You are on page 1of 15

Histopathology 2021, 78, 644–657. DOI: 10.1111/his.

14265

REVIEW

Updates from the 2020 World Health Organization


Classification of Soft Tissue and Bone Tumours
William J Anderson & Leona A Doyle
Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA

Date of submission 8 July 2020


Accepted for publication 24 September 2020
Published online Article Accepted 12 January 2021

Anderson W J & Doyle L A.


(2021) Histopathology 78, 644–657. https://doi.org/10.1111/his.14265
Updates from the 2020 World Health Organization classification of soft tissue and bone
tumours

The fifth edition of the World Health Organization refinements in classification and the development of
(WHO) classification of soft tissue and bone tumours was ancillary diagnostic tests, and have improved our under-
published in May 2020. This ‘Blue Book’, which is also standing of disease pathogenesis. Several new entities
available digitally for the first time, incorporates an array are also included. This review summarises the main
of new information on these tumours, amassed in the changes introduced in the 2020 WHO classification for
7 years since the previous edition. Major advances in each subcategory of soft tissue and bone tumours.
molecular characterisation have driven further
Keywords: bone, sarcoma, soft tissue, tumour, WHO

Introduction changes in each diagnostic category of the 2020


WHO classification.3
In the 7 years since the 2013 World Health Organi-
zation (WHO) classification of tumours of soft tissue
and bone, there have been considerable advances in Soft tissue
our understanding of this large and diverse group of ADIPOCYTIC TUMOURS
neoplasms.1 Many of these advances have been
underpinned by massively parallel sequencing, which The principal changes in this category are the addition
has become increasingly available during this period of two new entities: atypical spindle cell/pleomorphic
in both clinical and research settings. The identifica- lipomatous tumour and myxoid pleomorphic liposar-
tion of recurrent genetic events across the spectrum coma. Atypical spindle cell lipomatous tumour (ASLT)
of mesenchymal neoplasms has continued to drive and atypical pleomorphic lipomatous tumour (APLT)
improvements in diagnoses using molecular methods are low-grade adipocytic neoplasms that are thought to
and novel immunohistochemical markers.2 There constitute a single entity, on the basis of their shared
have also been important refinements in classification clinicopathological and molecular features.4,5 They are
and the recognition of new entities. In this review, therefore classified together as ‘atypical spindle cell/pleo-
we summarise the principal developments and morphic lipomatous tumour’. These tumours are often
larger, have less well-defined margins and have a wider
anatomical distribution than spindle cell/pleomorphic
lipoma. Their variable proportions of adipocytes, spindle
Address for correspondence: Leona A Doyle, MD, Department of
cells (often with mild to moderate atypia), lipoblasts, flo-
Pathology, Brigham and Women’s Hospital, 75 Francis Street, Bos-
ton, MA 02115, USA. e-mail: ladoyle@bwh.harvard.edu ret-like giant cells and extracellular matrix (myxoid or

© 2020 John Wiley & Sons Ltd.


WHO Classification 2020 645

and low-grade dedifferentiated liposarcoma, which are


A characterised by MDM2 amplification.
Myxoid pleomorphic liposarcoma (pleomorphic
myxoid liposarcoma), which is also included for the
first time, is an extremely rare and aggressive tumour
of children, adolescents and young adults that has a
tendency to arise in the mediastinum.6 Histologically,
it contains areas resembling both myxoid liposarcoma
and conventional pleomorphic liposarcoma, but it
lacks the characteristic molecular alterations of other
genetically defined liposarcomas (e.g. FUS/EWSR1
rearrangement and MDM2 amplification), and is now
considered to be a distinct entity.7
Finally, angiolipomas are now known to have
recurrent low-level mutations in the protein kinase
D2 gene (PRKD2), which is thought to drive tumori-
B genesis.8

FIBROBLASTIC AND MYOFIBROBLASTIC TUMOURS

This is the largest category of soft tissue tumours,


and it has grown even larger in the 2020 WHO
classification with the inclusion of several new enti-
ties. Among these is superficial CD34-positive fibrob-
lastic tumour, a low-grade tumour first described by
Carter et al. in 2014.9 It arises in the subcutis and
is typically well circumscribed. Histologically, it is
composed of spindled fibroblastic cells with ‘glassy’
cytoplasm admixed with scattered inflammatory cells
and foamy histiocytes (Figure 2).9,10 A key feature is
the presence of striking nuclear pleomorphism that
Figure 1. A, Atypical spindle cell lipomatous tumour is a benign occurs in the absence of significant mitotic activity
adipocytic neoplasm characterised by a prominent spindle cell com- or necrosis. In addition to diffuse positivity for CD34,
ponent in a collagenous or myxoid stroma. The spindle cells are focal positivity for keratins (mostly AE1/AE3) or des-
generally more numerous than in atypical lipomatous tumour/
min can be observed. Outcomes have so far been
well-differentiated liposarcoma, and show mild nuclear atypia. B,
Atypical pleomorphic lipomatous tumour has similar features, as very favourable, with only one reported metastasis
well as pleomorphic cells, floret-like multinucleated giant cells, and (to a regional lymph node) in the original series,
lipoblasts, which are often numerous. which included 13 patients with clinical follow-up
data - this tumor is therefore classified as intermedi-
ate with regards to biologic potential.9 Very recently,
collagenous) result in a broad morphological spectrum a subset has been shown to harbour PRDM10 rear-
(Figure 1). Immunohistochemically, ASLT may be posi- rangement.11
tive for CD34 (60%), S100 (~40%) and desmin (~20%).4 Angiofibroma of soft tissue is another new addition
Retinoblastoma protein expression is often lost in both to this category.12 This benign tumour arises most
ASLT and APLT, as these tumours commonly have dele- commonly in the lower extremities of adults, close to
tions of RB1 and its adjacent genes, i.e. RCBTB2, (and occasionally involving) large joints. It is well cir-
DLEU1, and ITM2B. They may recur locally, but, in con- cumscribed and composed of bland spindled fibroblas-
trast to atypical lipomatous tumours, they have no tic cells in a fibromyxoid stroma with prominent
known potential to dedifferentiate or metastasise and are thin-walled branching blood vessels (Figure 3). The
classified as benign. Both ASLT and APLT lack MDM2 latter have been likened to those of myxoid liposar-
amplification, and molecular methods can therefore be coma.12 Immunohistochemically, almost half of cases
helpful in resolving the differential diagnosis with atypi- show some expression of epithelial membrane anti-
cal lipomatous tumour/well-differentiated liposarcoma gen, and smaller subsets (~15%) show some degree of
© 2020 John Wiley & Sons Ltd, Histopathology, 78, 644–657.
646 W J Anderson and L A Doyle

Figure 2. Superficial CD34-positive fibroblastic tumour is a recently


described low-grade tumour comprising spindled and epithelioid Figure 4. EWSR1–SMAD3 fibroblastic tumour typically arises
cells with variable amounts of eosinophilic ‘glassy’ cytoplasm. superficially in the distal extremities. It is composed of intersecting
Despite considerable nuclear atypia reminiscent of a pleomorphic fascicles of bland fibroblastic spindle cells that infiltrate the sur-
sarcoma, mitoses are very scarce and necrosis is absent. rounding adipose tissue. Areas of stromal hyalinisation and calcifi-
cations are often present, either centrally, giving a zonation
architecture, or interspersed with the spindle cells, as in this
example.

be defined by EWSR1–SMAD3 fusion.14,15 Micro-


scopically, it often shows zonation by having a hya-
linised centre surrounded by cellular fascicles of
bland spindle cells (Figure 4). Immunohistochemi-
cally, ERG is consistently positive. Although only
small numbers of cases have been reported to date,
local recurrence after incomplete excision appears to
be common.
One of the chief molecular advances in this cate-
gory came in 2013, when NAB2–STAT6 fusion was
discovered as the defining molecular event in solitary
fibrous tumour (SFT).16 Immunohistochemistry for
signal transducer and activator of transcription 6
Figure 3. Soft tissue angiofibroma is characterised by haphazardly (STAT6) has since become an extremely reliable and
distributed bland spindle cells in a collagenous-to-myxoid stroma widely adopted means of establishing the diagnosis,
with prominent thin-walled branching blood vessels. The majority especially as the paracentric inversion of 12q that
harbour AHRR–NCOA2 fusion gene. produces the gene fusion is beyond the resolution of
fluorescence in-situ hybridisation (FISH).17 In more
recent years, TERT promoter mutations have been
characterised as secondary events associated with
CD34 or smooth muscle actin positivity. In the initial aggressive behaviour and metastasis.18,19
series there was a recurrent t(5;8) rearrangement, Importantly, improved risk stratification criteria for
which has subsequently been shown to target SFT are now included in the 2020 WHO classifica-
NCOA2, often resulting in AHRR–NCOA2 fusions.13 tion. In one widely adopted set of criteria—estab-
Also included for the first time is an emerging lished and later modified by Demicco et al.20—
entity provisionally termed EWSR1–SMAD3 fibrob- tumours are assigned low-risk, intermediate-risk or
lastic tumour. Two studies within the last year have high-risk status (specifically for metastasis) based on
described this as a benign tumour that arises most a combined assessment of clinical (patient age) and
commonly in females at acral sites and appears to pathological (tumour size, mitoses, and necrosis)
© 2020 John Wiley & Sons Ltd, Histopathology, 78, 644–657.
WHO Classification 2020 647

criteria. A separate risk calculator to predict overall In addition to those driven by WWTR1–CAMTA1
survival, local recurrence and metastasis has also fusion (>90% of cases), a small subset with a some-
been published by the French Sarcoma Group.21 what different morphology has been found to harbour
Finally, for several fibroblastic/myofibroblastic an alternative YAP1–TFE3 fusion.24 In this variant,
tumours there have been noteworthy advances in the tumour cells have more voluminous eosinophilic
our understanding of their genetics. Recurrent gene cytoplasm, and typically show a vasoformative
fusions have been identified in calcifying aponeurotic growth pattern as well as areas of solid growth (Fig-
fibroma (FN1–EGF) and lipofibromatosis (involving ure 5A).24 Immunohistochemistry for TFE3 shows
several genes encoding receptor tyrosine kinases),22 strong and diffuse nuclear staining (Figure 5B). Given
and insertion/duplication of EGFR exon 20 has been that all EHEs are classified as malignant, the use of
identified as a consistent event in fibrous hamartoma the term ‘malignant EHE’, which has been used to
of infancy.23 denote cases with higher-grade morphological fea-
tures, is no longer recommended. Pseudomyogenic
haemangioendothelioma, a vascular tumour of inter-
VASCULAR TUMOURS
mediate biological potential, was included for the first
Epithelioid haemangioendothelioma (EHE) is now time in the 2013 classification; it is now understood
recognised to comprise two distinct tumour subtypes. to be molecularly characterised by FOSB gene
rearrangements, as either SERPINE1–FOSB or
ACTB–FOSB fusions.25,26 Interestingly, FOSB activa-
A
tion—often via different fusions involving FOSB or
FOS—has also been identified in a subset of epithe-
lioid haemangiomas. FOSB immunohistochemistry
has therefore become a useful diagnostic marker for
both entities.25,27
Anastomosing haemangioma is included for the
first time, although it has been widely recognised
since 2009, when it was first described in the geni-
tourinary tract.28 This benign tumour most com-
monly occurs in the kidney and retroperitoneum.
Microscopically, it consists of delicately arborising
thin-walled vessels, whose architecture may resemble

Figure 5. A, Epithelioid haemangioendothelioma (EHE) with YAP1–


TFE3 accounts for a small subset of EHEs. It is composed of epithe- Figure 6. Anastomosing haemangioma is composed of numerous
lioid cells with abundant eosinophilic cytoplasm, and is typically thin-walled branching vascular channels that are lined by a single
vasoformative, in contrast to conventional EHE (driven by layer of endothelial cells with mildly protuberant nuclei. A charac-
WWTR1–CAMTA1). B, Diffuse nuclear positivity for TFE3 is charac- teristic feature is the presence of extramedullary haematopoiesis, as
teristic. evidenced by a megakaryocyte in this case.

© 2020 John Wiley & Sons Ltd, Histopathology, 78, 644–657.


648 W J Anderson and L A Doyle

angiosarcoma at low power (Figure 6). In contrast, strong and diffuse desmin expression, and can be mis-
however, the vessels are lined by a monolayer of uni- taken for a myogenic tumour such as leiomyosar-
form endothelial cells with only mildly protuberant coma. Interestingly, cellular myofibroma/
nuclei. When present, intracytoplasmic eosinophilic myopericytoma has a distinct SRF–RELA fusion (Fig-
globules, extramedullary haematopoiesis and minute ure 7).34 Glomus tumours that arise sporadically are
thrombi are helpful clues to the diagnosis. Most cases now known to commonly have MIR143–NOTCH1–3
have been shown to harbour activating mutations in fusions,36 and a small subset of those with intermedi-
GNAQ or GNA14.29 ate or malignant behaviour have been shown to have
Finally, tufted angioma and kaposiform haeman- KRAS and BRAF (p.V600E) mutations.37,38
gioendothelioma are now classified together more
clearly, reflecting the fact that they are most likely
SMOOTH MUSCLE TUMOURS
superficial and deep variants, respectively, of a single
entity. In addition to their shared clinical and mor- In this section, Epstein–Barr virus (EBV)-associated
phological features, recent work has also suggested smooth muscle tumours (EBV-SMTs) are now classi-
molecular overlap in a small number of cases.30 fied separately from leiomyosarcoma. EBV-SMTs

PERICYTIC (PERIVASCULAR) TUMOURS A

In the 2013 WHO classification, myopericytoma and


myofibroma were classified together, as it had become
apparent that they represent a morphological contin-
uum. Since then, PDGFRB mutation has been identi-
fied as a key driver event in myopericytoma and
myofibroma, being present in both familial and spo-
radic cases,31–33 and providing additional support for
their classification as a single entity. More recently, a
distinct subtype of myofibroma/myopericytoma has
been recognised that is characterised by increased cel-
lularity and is thus termed ‘cellular myofibroma/my-
opericytoma’.34,35 This subtype frequently shows

Figure 8. A, Epstein–Barr virus (EBV)-associated smooth muscle


Figure 7. Cellular myofibroma/myopericytoma with SRF–RELA is a tumour occurs in the setting of immunosuppression; histologically,
recently recognised variant of myofibroma/myopericytoma. Micro- fascicles of spindle cells with eosinophilic cytoplasm are present,
scopically, there is a hypercellular population of uniform ovoid-to- often with areas that have a more rounded cytomorphology and
spindled cells arranged in a fascicular or slightly nested pattern, as interspersed T lymphocytes. B, The tumour cells are positive for
well as the typical vascular component of myopericytoma EBV-encoded RNA by in-situ hybridisation.

© 2020 John Wiley & Sons Ltd, Histopathology, 78, 644–657.


WHO Classification 2020 649

occur in the setting of immunosuppression, being first tumours with hotspot mutations in MYOD1
described in patients with human immunodeficiency (p.L122R) that occur most often in adolescents and
virus/acquired immunodeficiency syndrome and young adults.45,46 Second, there are tumours har-
organ transplants.39,40 Their behaviour varies from bouring gene fusions involving VGLL2, NCOA2 or
benign to malignant, and is related to the degree of CITED2 that typically arise in paediatric patients and
immunosuppression. A proportion may be multifocal, that have a better prognosis.47,48 Third, there are
often because of separate localised EBV infections tumours that lack either of these alterations. Finally,
rather than metastasis. Histologically, EBV-SMTs tend it is noteworthy that there are very recently charac-
to have low-grade cytological features (mild to mod- terised variants of spindle cell/sclerosing rhab-
erate atypia and pleomorphism; low mitotic activity) domyosarcoma with TFCP2 rearrangement or
and lymphocytic infiltrates (Figure 8A).41 Detection MEIS–NCOA2 fusion that appear to have a predilec-
of EBV-encoded RNA by in-situ hybridisation is useful tion for bone.49–51 As a minor change, ectomes-
to confirm the diagnosis (Figure 8B). Inflammatory enchymoma is now assigned to this group, largely on
leiomyosarcoma, first described in 1995, is included the basis of its genetic overlap with embryonal rhab-
for the first time as a distinct entity.42 In contrast to domyosarcoma, having been previously classified as a
conventional leiomyosarcoma, it has a prominent nerve sheath tumour.
infiltrate of lymphocytes and/or histiocytes (Figure 9).
Recent work has confirmed earlier reports of its
CHONDRO-OSSEOUS TUMOURS
unique molecular profile, consisting of an unusual
haploid genome with few somatic mutations, thereby Soft tissue chondroma, which is a rare tumour that
supporting its separate classification.43,44 arises mostly in the hands and feet, has been shown
to have recurrent FN1–FGFR2 and FN1–FGFR1
fusions in 50% of cases.52 Interestingly, those with
SKELETAL MUSCLE TUMOURS
FN1 fusions all showed areas of so-called ‘grungy’
Spindle cell/sclerosing rhabdomyosarcoma is now calcification (also referred to as the ‘chondroblas-
known to comprise at least three subgroups of toma-like’ variant). This tumour therefore shows
tumours with quite distinct clinicopathological and morphological and molecular overlap with phospha-
molecular features. This represents a substantial turic mesenchymal tumour (PMT) (see below). Unlike
advance over the previous classification, when its PMT, however, soft tissue chondroma has no associa-
molecular basis was largely unknown. First, there are tion with fibroblast growth factor 23 expression or
osteomalacia, possibly reflecting its distinct ‘cell of ori-
gin’ or epigenetic profile.52

PERIPHERAL NERVE SHEATH TUMOURS.

An important molecular advance in this category


came with the discovery that a large proportion of
malignant peripheral nerve sheath tumours
(MPNSTs), ~80%, show inactivation of SUZ12 or
EED, which are genes that encode members of poly-
comb repressive complex 2 (PRC2).53,54 PRC2 is an
epigenetic ‘writer’ that normally directs the trimethy-
lation of histone 3 on lysine 27. Loss of PRC2 func-
tion in MPNST therefore erases this methylation
mark. Immunohistochemistry for H3K27me3, (his-
tone H3 trimethylated on lysine 27) to demonstrate
loss of expression, has become a useful diagnostic
marker, particularly in tumours toward the higher-
grade end of the spectrum, with a few limitations,
Figure 9. Inflammatory leiomyosarcoma comprises fascicles of spin-
such as patchy staining in synovial sarcoma and loss
dle cells with eosinophilic cytoplasm admixed with a prominent
infiltrate of lymphocytes and histiocytes. Its frequently haploid of expression in a subset of radiation-associated sarco-
genomic profile and favourable outcome support classification sepa- mas other than MPNST.55–59 Melanotic schwannoma
rate from conventional leiomyosarcoma. is now known as malignant melanotic nerve sheath
© 2020 John Wiley & Sons Ltd, Histopathology, 78, 644–657.
650 W J Anderson and L A Doyle

tumour, because large case series have better demon-


TUMOURS OF UNCERTAIN DIFFERENTIATION
strated its aggressive behaviour.60 Inactivating muta-
tions in PRKAR1A have also been recognised in the The principal change in this section is the inclusion
majority of cases, and loss of PRKAR1A immunohis- of NTRK-rearranged soft tissue neoplasms as an
tochemical expression can be used to support the ‘emerging’ entity. These tumours occur most com-
diagnosis.61 As a further advance, granular cell monly in children and adolescents, but the age range
tumour has been found to harbour mutations in is wide, and they are defined molecularly by rear-
ATP6AP1/ATP6AP2 in ~70% of cases; these are not rangement of NTRK1, NTRK2, or NTRK3, which are
only the probable inciting tumorigenic events, but genes that encode tropomyosin receptor kinases
also result in the abnormal vesicle accumulation that (although infantile fibrosarcoma, defined by ETV6–
imparts the characteristic granular cytoplasm of this NTRK3 fusion, remains in a separate category).63,64
tumour type.62 They include the recently described lipofibromatosis-
like neural tumour.65 Microscopically, they are typi-
cally composed of bland spindle cells in a variably
hyalinised stroma, and they may have solid and infil-
trative growth patterns (Figure 10A). Although most
A
tumours, particularly those in children, are clinically
benign or intermediate in biological potential (be-
cause of local recurrence), some pursue a more
aggressive clinical course, and this is often associated
with the presence of hypercellularity and a high mito-
tic rate. Immunohistochemically, many are positive
for both S100 and CD34, and reasonably sensitive
(but not entirely specific) pan-TRK antibodies have
also become available (Figure 10B).66,67
PMT was included for the first time in the 2013
WHO classification, and since then two large studies
have characterised FN1–FGFR1 and FN1–FGF1
fusions in almost 50% of cases.68,69

UNDIFFERENTIATED SMALL ROUND CELL


B SARCOMAS OF BONE AND SOFT TISSUE

Undifferentiated round cell sarcomas with CIC rear-


rangement, BCOR alterations or gene fusions involv-
ing non-ETS partner genes were each discussed only
briefly in the previous WHO classification. Our under-
standing of these tumours has grown considerably,
such that they are now classified individually in this
section separately from Ewing sarcoma, the tumour
for which many were mistaken in the past. CIC-rear-
ranged sarcomas are high-grade round cell sarcomas
that constitute the largest entity extracted from the
‘Ewing-like’ family of tumours. They are defined by
rearrangement of CIC (capicua), most commonly as a
gene fusion with DUX4, although rarer fusions with
non-DUX4 partner genes, such as FOXO4 and
Figure 10. A, NTRK-rearranged spindle cell neoplasm is an emerg- NUTM1, have also been described.70,71 While this
ing entity with heterogeneous morphology. A characteristic feature tumour was originally identified in paediatric
is the presence of stromal hyalinisation. There is often infiltrative
growth, as seen around a bronchiole in this pulmonary example
patients, it is now recognised to occur over a wide
which harboured an NTRK3 rearrangement. B, Immunohistochem- age range, and peaks in young adults.72,73 Histologi-
istry with a pan-TRK antibody usually shows cytoplasmic and/or cally, it comprises mildly pleomorphic round cells
nuclear positivity. with vesicular nuclei, prominent nucleoli, and
© 2020 John Wiley & Sons Ltd, Histopathology, 78, 644–657.
WHO Classification 2020 651

A
amphophilic cytoplasm, usually growing in a sheet-
like or lobular pattern (Figure 11A). In contrast to
Ewing sarcoma, it generally shows patchy CD99
expression. ETV4 and Wilms’ tumour 1 have emerged
as useful diagnostic markers, but are non-specific and
are best used as part of a panel of stains.74,75 These
tumours have a markedly worse prognosis than
Ewing sarcoma.73
It has also become apparent that a subgroup of
undifferentiated sarcomas with overlapping clinico-
pathological features harbour BCOR genetic alter-
ations. Such alterations, as either gene fusions (most
commonly BCOR–CCNB3) or internal tandem dupli-
cations (ITDs), result in BCOR activation or overex-
B pression.76 Sarcomas with BCOR–CCNB3 occur most
commonly in males in the first two decades of life,
and have a predilection for bone.77 However, sarco-
mas with BCOR ITD, in the form of infantile undiffer-
entiated round cell sarcoma or primitive myxoid
mesenchymal tumour of infancy, are more frequent
in soft tissues of the trunk and retroperitoneum. Mor-
phologically, sarcomas with BCOR alterations are
composed of primitive round to ovoid or occasionally
spindled cells (Figure 11B). Immunohistochemistry
for BCOR is sensitive but not specific, and SATB2 and
TLE1 are also often positive.78,79
Finally, rare sarcomas with EWSR1 fusions involv-
ing non-ETS partner genes have now also been
shown, by gene expression and methylation profiling,
C to be distinct from Ewing sarcoma. Sarcomas with
EWSR1–NFATC2 (or FUS–NFATC2) arise mostly in
the metaphysis or diaphysis of long bones.80,81 They
show a slight male predominance and occur over a
wide age range, but are most frequent in middle-aged
adults.80,81 Microscopically, they consist of round or
spindled cells often arranged in nests and cords,82
and they may show foci of osseous or cartilaginous
differentiation.
In contrast, sarcomas with EWSR1–PATZ1 are
more common in the deep soft tissues of the trunk
and have a more equal sex distribution.83 The mor-
phological appearances are variable (Figure 11C),
and there is immunohistochemical coexpression of
muscle and neurogenic (S100 and SOX10) mark-
ers.84
Figure 11. Round cell sarcomas. A, CIC-rearranged sarcoma typi-
cally has lobulated and sheet-like growth of round cells with mild
nuclear pleomorphism and prominent nucleoli. B, The tumour cells
of BCOR–CCNB3 sarcomas are usually small and round-to-ovoid Bone
with inconspicuous nucleoli, and a delicate vascular network may
be seen. C, EWSR1–PATZ1 sarcomas have a broad range of mor- CHONDROGENIC
phologies. This tumour shows small round cells with clear or
slightly amphophilic cytoplasm. The cells are in nests and trabecu- In the 2013 WHO classification, the term ‘atypical
lae separated by thin fibrous septae. cartilaginous tumour’ (ACT) was introduced as a sub-
stitute for grade 1 chondrosarcoma, as it was felt that
© 2020 John Wiley & Sons Ltd, Histopathology, 78, 644–657.
652 W J Anderson and L A Doyle

A B

Figure 12. A,
Chondroblastoma is composed
of relatively uniform ovoid cells
with grooved nuclei and
distinct eosinophilic cytoplasm
C D within an eosinophilic
chondroid matrix. B, Histone
H3 with K36M mutation
(H3K36M) shows strong
nuclear positivity in tumour
cells. C, Giant cell tumour of
bone in a small biopsy
specimen. D, Histone H3 with
G34W mutation (H3G34W) is
diffusely positive in the
mononuclear cells, but not in
the osteoclast-like giant cells.

such tumours, although locally aggressive, lacked fibronectin gene (FN1) rearrangements outlined ear-
overtly malignant behaviour. In the 2020 classifica- lier that show chondroid matrix production.52,87 It is
tion, it is proposed that ACT should be restricted to important to note that FN1/ACVR2A rearrangement
tumours arising in the appendicular skeleton, which may also be detected in chondrosarcomas that can
are more amenable to complete surgical resection rarely develop from synovial chondromatosis.88
than tumours located in the axial skeleton. Con- Chondromyxoid fibroma is now classified as a
versely, ‘chondrosarcoma, grade 1’ should be used for benign entity (with a local recurrence rate of approxi-
axial tumours. The use of these two different terms mately 10–15%).89 In addition, the majority of cases
based on tumour resectability is similar to atypical are now known to be driven by fusions of the gluta-
lipomatous tumour/well-differentiated liposarcoma. mate receptor gene (GRM1). These fusions are formed
An important molecular advance in this category by complex genomic rearrangements, i.e. chromo-
came in 2013, when Behjati et al. characterised recur- plexy and chromothripsis, and result in GRM1 up-
rent p.K36M mutations of H3-3B (H3F3B), or rarely regulation.90
H3-3A (H3F3A), in ~95% of chondroblastomas.85
Remarkably, the same study identified similar but dis-
OSTEOGENIC
tinct histone mutations in giant cell tumour of bone,
another giant cell-rich tumour of the epiphysis (see The most common benign bone-forming neoplasms,
below). This has led to the development of a sensitive i.e. osteoid osteoma and osteoblastoma, have quite
and specific immunohistochemical marker directed recently been demonstrated to share FOS (and, less
against the mutant histone, K36M, facilitating the commonly, FOSB) rearrangements.91 These molec-
diagnosis of chondroblastoma (Figure 12A,B).86 Given ular findings, together with their identical histolog-
its very low rate of metastasis (<1%), chondroblastoma ical features, further support a very close
is also now classified as benign. relationship between these tumours. Currently,
Synovial chondromatosis was previously considered however, they are still classified separately, on the
to be benign, but it is now classed as having interme- basis of their distinct clinical and radiological pre-
diate (locally aggressive) biological potential. Further- sentations and an arbitrary size cut-off (20 mm).
more, recurrent rearrangements of FN1 and/or FISH for FOS rearrangement and FOS immunohis-
ACVR2A have been characterised in 57% of tochemistry are emerging as valuable ancillary
cases.52,87,88 Synovial chondromatosis therefore joins tests with which to distinguish challenging cases
a growing list of mesenchymal tumours with from osteosarcoma.91

© 2020 John Wiley & Sons Ltd, Histopathology, 78, 644–657.


WHO Classification 2020 653

A B

C D

Figure 13. A, Poorly differentiated chordoma composed of sheets of epithelioid cells with moderate nuclear pleomorphism, vesicular chro-
matin, and occasional cells with a slightly rhabdoid appearance. B, Immunohistochemically, there is loss of expression of INI-1/SMARCB1.
C, Dedifferentiated chordoma is composed of highly pleomorphic atypical cells. D, There is loss of nuclear brachyury expression in the dedif-
ferentiated component.

It is of note that small-cell and telangiectatic Aneurysmal bone cyst, the third tumour in this
osteosarcoma are now described as variants of section, was previously in the intermediate (locally
osteosarcoma rather than as separate entities. aggressive) category but is now classified as benign.
The designation ‘benign fibrous histiocytoma of
bone’ is no longer recommended. Tumours with such
OSTEOCLASTIC GIANT CELL-RICH
morphology are now generally considered to be
Giant cell tumour of bone has been shown to harbour examples of non-ossifying fibroma when arising in
H3-3A (H3F3A) mutations in ~95% of cases, the the metaphysis, or giant cell tumour of bone (with
most common being p.G34W (~90%).85 Immunohis- regressive changes) when located in the epiphysis.
tochemically, G34W is positive in the mononuclear ‘Giant cell lesion of the small bones’ is also no longer
cells, and it has become a very helpful diagnostic regarded as a distinct entity.
marker (Figure 12C,D). However, G34W positivity is
not synonymous with benignity, as malignant giant
NOTOCHORDAL TUMOURS
cell tumour of bone can harbour the same mutation.
Non-ossifying fibroma is another benign osteoclast- Poorly differentiated chordoma and dedifferentiated
rich lesion whose molecular basis has now been chordoma are discussed in more detail in the 2020
determined. In a recent series of 59 cases, mutually WHO classification. Poorly differentiated chordoma
exclusive mutations of mitogen-activated protein was first described in 2010 as an aggressive chor-
kinase pathway genes were identified, including doma variant that typically occurs in the base of the
mutations of KRAS (64%), FGFR1 (14%), and NF1 skull or the upper cervical spine of children.94 Mor-
(two cases).92 These intriguing results highlight non- phologically, it is composed of epithelioid cells with
ossifying fibroma as another example of a ‘self-limit- eosinophilic cytoplasm and prominent nucleoli,
ing’ neoplasm, analogous to nodular fasciitis of soft arranged in a solid or nested growth pattern with
tissues, as it has long been recognised to undergo areas of necrosis (Figure 13A).95 Although it is histo-
spontaneous regression.92,93 logically distinct from conventional chordoma, it is

© 2020 John Wiley & Sons Ltd, Histopathology, 78, 644–657.


654 W J Anderson and L A Doyle

similarly positive for keratins and brachyury. In addi- Diseases for Oncology (ICD-O) coding (in the tables of
tion, this variant is characterised by homozygous tumour classifications), and for which there is no
deletions of SMARCB1, and, immunohistochemically, clear or corresponding diagnostic category, should be
shows loss of INI-1 (SMARCB1) expression (Fig- used with caution or avoided, e.g. ‘epithelioid liposar-
ure 13B).96 Dedifferentiated chordoma, in contrast, is coma’. Epithelioid liposarcoma refers only to a mor-
a high-grade sarcoma that derives from conventional phological variant of pleomorphic liposarcoma with
chordoma and was first described in 1987.97 Progres- predominantly epithelioid cytomorphology rather
sion to dedifferentiated chordoma is often histologi- than a distinct subtype of liposarcoma. Similarly,
cally abrupt, imparting a biphasic appearance, and is ‘clear cell sarcoma NOS’, ‘rhabdoid tumour NOS’ and
associated with loss of brachyury expression (Fig- ‘mesenchymoma NOS’ refer only to clear cell sar-
ure 13C,D).98 coma, malignant rhabdoid tumour and fibrocartilagi-
nous mesenchymoma, as there are no other defined
types of these tumours in soft tissue or bone.
OTHER MESENCHYMAL TUMOURS OF BONE
In summary, the 2020 WHO classification of soft
Fibrocartilaginous mesenchymoma is included as a tissue and bone tumours incorporates an array of
new entity in this category, having been originally new information that has been amassed in the
described by Dahlin et al. in 1984.99 This very rare 7 years since the previous edition, including major
tumour of adolescents and young adults tends to advances in the molecular characterisation of soft tis-
arise in the metaphysis of long bones and is locally sue and bone tumours, refinements in tumour classi-
aggressive. Histologically, it is composed of mildly fication, and novel ancillary diagnostic tests, all of
atypical spindle cells admixed with nodules of hyaline which are helping to improve our understanding of
cartilage that have a ‘growth plate-like’ appearance disease pathogenesis.
and undergo endochondral ossification. In a recent
small series, GNAS and IDH1/2 mutations were
absent, as was MDM2 amplification, providing molec- Conflicts of interest
ular support for fibrocartilaginous mesenchymoma
The authors have no conflicts of interest to disclose.
being distinct from several other entities that show
overlapping morphology (fibrocartilaginous dysplasia,
chondrosarcoma, and low-grade osteosarcoma, DATA AVAILABILITY STATEMENT
respectively).100
Data sharing is not applicable to this article as no
Adamantinoma, a biphasic fibro-osseous tumour
new data were created or analyzed in this study.
that almost always arises in the tibia and/or fibula,
has a variety of histological patterns that, in the past,
were all regarded as malignant. However, the osteofi- References
brous dysplasia (OFD)-like variant has now been clas- 1. Fletcher CDM, Bridge JA, Hogendoorn PCW et al. eds. World
sified as an intermediate-category (locally aggressive) Health Organization classification of tumours of soft tissue and
neoplasm. In contrast to the classic variant, which bone. 4th ed. Lyon: IARC Press, 2013.
recurs very frequently and can metastasise in approx- 2. Anderson WJ, Hornick JL. Immunohistochemical correlates of
imately 10–30% of cases, the OFD-like variant has a recurrent genetic alterations in sarcomas. Genes Chromosomes
Cancer 2019; 58; 111–123.
low local recurrence rate (20%), and metastases are 3. WHO Classification of Tumours Editorial Board eds. World
rare.101–103 Histologically, apart from resembling Health Organization classification of soft tissue and bone tumours.
OFD, this variant also differs from classic adamanti- 5th ed. Lyon: IARC Press, 2020.
noma by having less prominent epithelial nests. 4. Marino-Enriquez A, Nascimento AF, Ligon AH et al. Atypical
Finally, very rare examples of dedifferentiated spindle cell lipomatous tumor: clinicopathologic characteriza-
tion of 232 cases demonstrating a morphologic spectrum. Am.
adamantinoma are also now recognised; these show J. Surg. Pathol. 2017; 41; 234–244.
transition to a high-grade malignancy with pleomor- 5. Creytens D, Mentzel T, Ferdinande L et al. ‘Atypical’ pleomor-
phism and loss of the typical epithelial elements. phic lipomatous tumor: a clinicopathologic, immunohisto-
chemical and molecular study of 21 cases, emphasizing its
relationship to atypical spindle cell lipomatous tumor and sug-
gesting a morphologic spectrum (atypical spindle cell/pleomor-
Final comments and conclusions phic lipomatous tumor). Am. J. Surg. Pathol. 2017; 41; 1443–
1455.
Some terms introduced in the 2020 WHO classifica- 6. Alaggio R, Coffin CM, Weiss SW et al. Liposarcomas in young
tion for the purpose of International Classification of patients: a study of 82 cases occurring in patients younger

© 2020 John Wiley & Sons Ltd, Histopathology, 78, 644–657.


WHO Classification 2020 655

than 22 years of age. Am. J. Surg. Pathol. 2009; 33; 645– 24. Antonescu CR, Le Loarer F, Mosquera JM et al. Novel YAP1-
658. TFE3 fusion defines a distinct subset of epithelioid heman-
7. Hofvander J, Jo VY, Ghanei I et al. Comprehensive genetic gioendothelioma. Genes Chromosomes Cancer 2013; 52; 775–
analysis of a paediatric pleomorphic myxoid liposarcoma 784.
reveals near-haploidization and loss of the RB1 gene. 25. Walther C, Tayebwa J, Lilljebjorn H et al. A novel SERPINE1–
Histopathology 2016; 69; 141–147. FOSB fusion gene results in transcriptional up-regulation of
8. Hofvander J, Arbajian E, Stenkula KG et al. Frequent low-level FOSB in pseudomyogenic haemangioendothelioma. J. Pathol.
mutations of protein kinase D2 in angiolipoma. J. Pathol. 2014; 232; 534–540.
2017; 241; 578–582. 26. Agaram NP, Zhang L, Cotzia P, Antonescu CR. Expanding
9. Carter JM, Weiss SW, Linos K et al. Superficial CD34-positive the spectrum of genetic alterations in pseudomyogenic
fibroblastic tumor: report of 18 cases of a distinctive low-grade hemangioendothelioma with recurrent novel ACTB-FOSB
mesenchymal neoplasm of intermediate (borderline) malig- gene fusions. Am. J. Surg. Pathol. 2018; 42; 1653–1661.
nancy. Mod. Pathol. 2014; 27; 294–302. 27. Hung YP, Fletcher CD, Hornick JL. FOSB is a useful diagnostic
10. Lao IW, Yu L, Wang J. Superficial CD34-positive fibroblastic marker for pseudomyogenic hemangioendothelioma. Am. J.
tumour: a clinicopathological and immunohistochemical study Surg. Pathol. 2017; 41; 596–606.
of an additional series. Histopathology 2017; 70; 394–401. 28. Montgomery E, Epstein JI. Anastomosing hemangioma of the
11. Puls F, Pillay N, Fagman H et al. PRDM10-rearranged soft tis- genitourinary tract: a lesion mimicking angiosarcoma. Am. J.
sue tumor: a clinicopathologic study of 9 cases. Am. J. Surg. Surg. Pathol. 2009; 33; 1364–1369.
Pathol. 2019; 43; 504–513. 29. Bean GR, Joseph NM, Gill RM et al. Recurrent GNAQ muta-
12. Marino-Enriquez A, Fletcher CD. Angiofibroma of soft tissue: tions in anastomosing hemangiomas. Mod. Pathol. 2017; 30;
clinicopathologic characterization of a distinctive benign 722–727.
fibrovascular neoplasm in a series of 37 cases. Am. J. Surg. 30. Lim YH, Bacchiocchi A, Qiu J et al. GNA14 somatic mutation
Pathol. 2012; 36; 500–508. causes congenital and sporadic vascular tumors by MAPK
13. Jin Y, Moller E, Nord KH et al. Fusion of the AHRR and activation. Am. J. Hum. Genet. 2016; 99; 443–450.
NCOA2 genes through a recurrent translocation t(5;8)(p15; 31. Agaimy A, Bieg M, Michal M et al. Recurrent somatic
q13) in soft tissue angiofibroma results in upregulation of aryl PDGFRB mutations in sporadic infantile/solitary adult myofi-
hydrocarbon receptor target genes. Genes Chromosomes Cancer bromas but not in angioleiomyomas and myopericytomas.
2012; 51; 510–520. Am. J. Surg. Pathol. 2017; 41; 195–203.
14. Kao YC, Flucke U, Eijkelenboom A et al. Novel EWSR1- 32. Hung YP, Fletcher CDM. Myopericytomatosis: clinicopatho-
SMAD3 gene fusions in a group of acral fibroblastic spindle logic analysis of 11 cases with molecular identification of
cell neoplasms. Am. J. Surg. Pathol. 2018; 42; 522–528. recurrent PDGFRB alterations in myopericytomatosis and
15. Michal M, Kazakov DV, Michalova K, Michal M. Atypical myopericytoma. Am. J. Surg. Pathol. 2017; 41; 1034–1044.
multivacuolated lipoblasts and atypical mitoses are not com- 33. Martignetti JA, Tian L, Li D et al. Mutations in PDGFRB cause
patible with the diagnosis of spindle cell/pleomorphic lipoma autosomal-dominant infantile myofibromatosis. Am. J. Hum.
—reply. Hum. Pathol. 2018; 74; 189. Genet. 2013; 92; 1001–1007.
16. Robinson DR, Wu YM, Kalyana-Sundaram S et al. Identifica- 34. Antonescu CR, Sung YS, Zhang L et al. Recurrent SRF-RELA
tion of recurrent NAB2-STAT6 gene fusions in solitary fibrous fusions define a novel subset of cellular myofibroma/myopericy-
tumor by integrative sequencing. Nat. Genet. 2013; 45; 180– toma: a potential diagnostic pitfall with sarcomas with myo-
185. genic differentiation. Am. J. Surg. Pathol. 2017; 41; 677–684.
17. Doyle LA, Vivero M, Fletcher CD et al. Nuclear expression of 35. Linos K, Carter JM, Gardner JM et al. Myofibromas with atypi-
STAT6 distinguishes solitary fibrous tumor from histologic cal features: expanding the morphologic spectrum of a benign
mimics. Mod. Pathol. 2014; 27; 390–395. entity. Am. J. Surg. Pathol. 2014; 38; 1649–1654.
18. Bahrami A, Lee S, Schaefer IM et al. TERT promoter muta- 36. Mosquera JM, Sboner A, Zhang L et al. Novel MIR143-
tions and prognosis in solitary fibrous tumor. Mod. Pathol. NOTCH fusions in benign and malignant glomus tumors.
2016; 29; 1511–1522. Genes Chromosomes Cancer 2013; 52; 1075–1087.
19. Demicco EG, Wani K, Ingram D et al. TERT promoter muta- 37. Chakrapani A, Warrick A, Nelson D et al. BRAF and KRAS
tions in solitary fibrous tumour. Histopathology 2018; 73; mutations in sporadic glomus tumors. Am. J. Dermatopathol.
843–851. 2012; 34; 533–535.
20. Demicco EG, Wagner MJ, Maki RG et al. Risk assessment in 38. Karamzadeh Dashti N, Bahrami A, Lee SJ et al. BRAF V600E
solitary fibrous tumors: validation and refinement of a risk mutations occur in a subset of glomus tumors, and are asso-
stratification model. Mod. Pathol. 2017; 30; 1433–1442. ciated with malignant histologic characteristics. Am. J. Surg.
21. Salas S, Resseguier N, Blay JY et al. Prediction of local and Pathol. 2017; 41; 1532–1541.
metastatic recurrence in solitary fibrous tumor: construction 39. Lee ES, Locker J, Nalesnik M et al. The association of Epstein-
of a risk calculator in a multicenter cohort from the French Barr virus with smooth-muscle tumors occurring after organ
Sarcoma Group (FSG) database. Ann. Oncol. 2017; 28; 1979– transplantation. N. Engl. J. Med. 1995; 332; 19–25.
1987. 40. McClain KL, Leach CT, Jenson HB et al. Association of
22. Al-Ibraheemi A, Folpe AL, Perez-Atayde AR et al. Aberrant Epstein-Barr virus with leiomyosarcomas in young people
receptor tyrosine kinase signaling in lipofibromatosis: a clini- with AIDS. N. Engl. J. Med. 1995; 332; 12–18.
copathological and molecular genetic study of 20 cases. Mod. 41. Deyrup AT, Lee VK, Hill CE et al. Epstein-Barr virus-associated
Pathol. 2019; 32; 423–434. smooth muscle tumors are distinctive mesenchymal tumors
23. Park JY, Cohen C, Lopez D et al. EGFR exon 20 insertion/du- reflecting multiple infection events: a clinicopathologic and
plication mutations characterize fibrous hamartoma of molecular analysis of 29 tumors from 19 patients. Am. J.
infancy. Am. J. Surg. Pathol. 2016; 40; 1713–1718. Surg. Pathol. 2006; 30; 75–82.

© 2020 John Wiley & Sons Ltd, Histopathology, 78, 644–657.


656 W J Anderson and L A Doyle

42. Merchant W, Calonje E, Fletcher CD. Inflammatory 59. Panse G, Mito JK, Ingram DRet al.Radiation-associated sarco-
leiomyosarcoma: a morphological subgroup within the mas other than malignant peripheral nerve sheath tumors
heterogeneous family of so-called inflammatory malignant demonstrate loss of histone H3K27 trimethylation.
fibrous histiocytoma. Histopathology 1995; 27; 525–532. Histopathology 2020. E-pub ahead of print, 31 July.
43. Dal Cin P, Sciot R, Fletcher CD et al. Inflammatory 60. Khoo M, Pressney I, Hargunani R, Tirabosco R. Melanotic
leiomyosarcoma may be characterized by specific near-haploid schwannoma: an 11-year case series. Skeletal Radiol. 2016;
chromosome changes. J. Pathol. 1998; 185; 112–115. 45; 29–34.
44. Arbajian E, Koster J, Vult von Steyern F, Mertens F. Inflam- 61. Wang L, Zehir A, Sadowska J et al. Consistent copy number
matory leiomyosarcoma is a distinct tumor characterized by changes and recurrent PRKAR1A mutations distinguish
near-haploidization, few somatic mutations, and a primitive melanotic schwannomas from melanomas: SNP-array and
myogenic gene expression signature. Mod. Pathol. 2018; 31; next generation sequencing analysis. Genes Chromosomes Can-
93–100. cer 2015; 54; 463–471.
45. Kohsaka S, Shukla N, Ameur N et al. A recurrent neomorphic 62. Pareja F, Brandes AH, Basili T et al. Loss-of-function muta-
mutation in MYOD1 defines a clinically aggressive subset of tions in ATP6AP1 and ATP6AP2 in granular cell tumors.
embryonal rhabdomyosarcoma associated with PI3K-AKT Nat. Commun. 2018; 9; 3533.
pathway mutations. Nat. Genet. 2014; 46; 595–600. 63. Suurmeijer AJH, Dickson BC, Swanson D et al. A novel group
46. Szuhai K, de Jong D, Leung WY et al. Transactivating muta- of spindle cell tumors defined by S100 and CD34 co-expres-
tion of the MYOD1 gene is a frequent event in adult spindle sion shows recurrent fusions involving RAF1, BRAF, and
cell rhabdomyosarcoma. J. Pathol. 2014; 232; 300–307. NTRK1/2 genes. Genes Chromosomes Cancer 2018; 57; 611–
47. Alaggio R, Zhang L, Sung YS et al. A molecular study of pedi- 621.
atric spindle and sclerosing rhabdomyosarcoma: identification 64. Yamazaki F, Nakatani F, Asano N et al. Novel NTRK3 fusions
of novel and recurrent VGLL2-related fusions in infantile in fibrosarcomas of adults. Am. J. Surg. Pathol. 2019; 43;
cases. Am. J. Surg. Pathol. 2016; 40; 224–235. 523–530.
48. Mosquera JM, Sboner A, Zhang L et al. Recurrent NCOA2 65. Agaram NP, Zhang L, Sung YS et al. Recurrent NTRK1 gene
gene rearrangements in congenital/infantile spindle cell rhab- fusions define a novel subset of locally aggressive lipofibro-
domyosarcoma. Genes Chromosomes Cancer 2013; 52; 538– matosis-like neural tumors. Am. J. Surg. Pathol. 2016; 40;
550. 1407–1416.
49. Watson S, Perrin V, Guillemot D et al. Transcriptomic defini- 66. Hechtman JF, Benayed R, Hyman DM et al. Pan-Trk immuno-
tion of molecular subgroups of small round cell sarcomas. J. histochemistry is an efficient and reliable screen for the detec-
Pathol. 2018; 245; 29–40. tion of NTRK fusions. Am. J. Surg. Pathol. 2017; 41; 1547–
50. Dashti NK, Wehrs RN, Thomas BC et al. Spindle cell rhab- 1551.
domyosarcoma of bone with FUS–TFCP2 fusion: confirmation 67. Hung YP, Fletcher CDM, Hornick JL. Evaluation of pan-TRK
of a very recently described rhabdomyosarcoma subtype. immunohistochemistry in infantile fibrosarcoma, lipofibro-
Histopathology 2018; 73; 514–520. matosis-like neural tumour and histological mimics.
51. Agaram NP, Zhang L, Sung YS et al. Expanding the spectrum Histopathology 2018; 73; 634–644.
of intraosseous rhabdomyosarcoma: correlation between 2 68. Lee JC, Jeng YM, Su SY et al. Identification of a novel FN1–
distinct gene fusions and phenotype. Am. J. Surg. Pathol. FGFR1 genetic fusion as a frequent event in phosphaturic
2019; 43; 695–702. mesenchymal tumour. J. Pathol. 2015; 235; 539–545.
52. Amary F, Perez-Casanova L, Ye H et al. Synovial chondro- 69. Lee JC, Su SY, Changou CA et al. Characterization of FN1–
matosis and soft tissue chondroma: extraosseous cartilaginous FGFR1 and novel FN1–FGF1 fusion genes in a large series of
tumor defined by FN1 gene rearrangement. Mod. Pathol. phosphaturic mesenchymal tumors. Mod. Pathol. 2016; 29;
2019; 32; 1762–1771. 1335–1346.
53. Lee W, Teckie S, Wiesner T et al. PRC2 is recurrently inacti- 70. Le Loarer F, Pissaloux D, Watson S et al. Clinicopathologic
vated through EED or SUZ12 loss in malignant peripheral features of CIC-NUTM1 sarcomas, a new molecular variant of
nerve sheath tumors. Nat. Genet. 2014; 46; 1227–1232. the family of CIC-fused sarcomas. Am. J. Surg. Pathol. 2019;
54. Zhang M, Wang Y, Jones S et al. Somatic mutations of SUZ12 43; 268–276.
in malignant peripheral nerve sheath tumors. Nat. Genet. 71. Sugita S, Arai Y, Tonooka A et al. A novel CIC-FOXO4 gene
2014; 46; 1170–1172. fusion in undifferentiated small round cell sarcoma: a geneti-
55. Schaefer IM, Fletcher CD, Hornick JL. Loss of H3K27 cally distinct variant of Ewing-like sarcoma. Am. J. Surg.
trimethylation distinguishes malignant peripheral nerve Pathol. 2014; 38; 1571–1576.
sheath tumors from histologic mimics. Mod. Pathol. 2016; 29; 72. Richkind KE, Romansky SG, Finklestein JZ. t(4;19)(q35;
4–13. q13.1): a recurrent change in primitive mesenchymal
56. Prieto-Granada CN, Wiesner T, Messina JL et al. Loss of tumors? Cancer Genet. Cytogenet. 1996; 87; 71–74.
H3K27me3 expression is a highly sensitive marker for spo- 73. Antonescu CR, Owosho AA, Zhang L et al. Sarcomas
radic and radiation-induced MPNST. Am. J. Surg. Pathol. with CIC-rearrangements are a distinct pathologic entity
2016; 40; 479–489. with aggressive outcome: a clinicopathologic and molecu-
57. Cleven AH, Sannaa GA, Briaire-de Bruijn I et al. Loss of lar study of 115 cases. Am. J. Surg. Pathol. 2017; 41; 941–
H3K27 tri-methylation is a diagnostic marker for malignant 949.
peripheral nerve sheath tumors and an indicator for an infe- 74. Hung YP, Fletcher CD, Hornick JL. Evaluation of ETV4 and
rior survival. Mod. Pathol. 2016; 29; 582–590. WT1 expression in CIC-rearranged sarcomas and histologic
58. Mentzel T, Kiss K. Reduced H3K27me3 expression in radia- mimics. Mod. Pathol. 2016; 29; 1324–1334.
tion-associated angiosarcoma of the breast. Virchows Arch. 75. Le Guellec S, Velasco V, Perot G et al. ETV4 is a useful mar-
2018; 472; 361–368. ker for the diagnosis of CIC-rearranged undifferentiated

© 2020 John Wiley & Sons Ltd, Histopathology, 78, 644–657.


WHO Classification 2020 657

round-cell sarcomas: a study of 127 cases including mimick- 89. Bhamra JS, Al-Khateeb H, Dhinsa BS et al. Chondromyxoid
ing lesions. Mod. Pathol. 2016; 29; 1523–1531. fibroma management: a single institution experience of 22
76. Pierron G, Tirode F, Lucchesi C et al. A new subtype of bone cases. World J. Surg. Oncol. 2014; 12; 283.
sarcoma defined by BCOR-CCNB3 gene fusion. Nat. Genet. 90. Nord KH, Lilljebjorn H, Vezzi F et al. GRM1 is upregulated
2012; 44; 461–466. through gene fusion and promoter swapping in chondromyx-
77. Puls F, Niblett A, Marland G et al. BCOR-CCNB3 (Ewing-like) oid fibroma. Nat. Genet. 2014; 46; 474–477.
sarcoma: a clinicopathologic analysis of 10 cases, in compar- 91. Fittall MW, Mifsud W, Pillay N et al. Recurrent rearrange-
ison with conventional Ewing sarcoma. Am. J. Surg. Pathol. ments of FOS and FOSB define osteoblastoma. Nat. Commun.
2014; 38; 1307–1318. 2018; 9; 2150.
78. Li WS, Liao IC, Wen MC et al. BCOR–CCNB3-positive soft tissue 92. Baumhoer D, Kovac M, Sperveslage J et al. Activating muta-
sarcoma with round-cell and spindle-cell histology: a series of tions in the MAP-kinase pathway define non-ossifying
four cases highlighting the pitfall of mimicking poorly differen- fibroma of bone. J. Pathol. 2019; 248; 116–122.
tiated synovial sarcoma. Histopathology 2016; 69; 792–801. 93. Bovee JV, Hogendoorn PC. Non-ossifying fibroma: a RAS-
79. Kao YC, Owosho AA, Sung YS et al. BCOR-CCNB3 fusion pos- MAPK driven benign bone neoplasm. J. Pathol. 2019; 248;
itive sarcomas: a clinicopathologic and molecular analysis of 127–130.
36 cases with comparison to morphologic spectrum and clini- 94. Mobley BC, McKenney JK, Bangs CD et al. Loss of SMARCB1/
cal behavior of other round cell sarcomas. Am. J. Surg. Pathol. INI1 expression in poorly differentiated chordomas. Acta Neu-
2018; 42; 604–615. ropathol. 2010; 120; 745–753.
80. Diaz-Perez JA, Nielsen GP, Antonescu C et al. EWSR1/FUS- 95. Shih AR, Cote GM, Chebib I et al. Clinicopathologic character-
NFATc2 rearranged round cell sarcoma: clinicopathological istics of poorly differentiated chordoma. Mod. Pathol. 2018;
series of 4 cases and literature review. Hum. Pathol. 2019; 31; 1237–1245.
90; 45–53. 96. Hasselblatt M, Thomas C, Hovestadt V et al. Poorly differenti-
81. Perret R, Escuriol J, Velasco V et al. NFATc2-rearranged sar- ated chordoma with SMARCB1/INI1 loss: a distinct molecular
comas: clinicopathologic, molecular, and cytogenetic study of entity with dismal prognosis. Acta Neuropathol. 2016; 132;
7 cases with evidence of AGGRECAN as a novel diagnostic 149–151.
marker. Mod. Pathol. 2020; 33; 1930–1944. 97. Meis JM, Raymond AK, Evans HL et al. ‘Dedifferentiated’
82. Wang GY, Thomas DG, Davis JL et al. EWSR1-NFATC2 chordoma. A clinicopathologic and immunohistochemical
translocation-associated sarcoma clinicopathologic findings in study of three cases. Am. J. Surg. Pathol. 1987; 11; 516–
a rare aggressive primary bone or soft tissue tumor. Am. J. 525.
Surg. Pathol. 2019; 43; 1112–1122. 98. Vujovic S, Henderson S, Presneau N et al. Brachyury, a cru-
83. Bridge JA, Sumegi J, Druta M et al. Clinical, pathological, and cial regulator of notochordal development, is a novel biomar-
genomic features of EWSR1-PATZ1 fusion sarcoma. Mod. ker for chordomas. J. Pathol. 2006; 209; 157–165.
Pathol. 2019; 32; 1593–1604. 99. Dahlin DC, Bertoni F, Beabout JW, Campanacci M. Fibrocarti-
84. Chougule A, Taylor MS, Nardi V et al. Spindle and round cell laginous mesenchymoma with low-grade malignancy. Skeletal
sarcoma with EWSR1-PATZ1 gene fusion: a sarcoma with Radiol. 1984; 12; 263–269.
polyphenotypic differentiation. Am. J. Surg. Pathol. 2019; 43; 100. Gambarotti M, Righi A, Vanel D et al. Fibrocartilaginous mes-
220–228. enchymoma of bone: a single-institution experience with
85. Behjati S, Tarpey PS, Presneau N et al. Distinct H3F3A and molecular investigations and a review of the literature.
H3F3B driver mutations define chondroblastoma and giant Histopathology 2017; 71; 134–142.
cell tumor of bone. Nat. Genet. 2013; 45; 1479–1482. 101. Scholfield DW, Sadozai Z, Ghali C et al. Does osteofibrous dys-
86. Amary MF, Berisha F, Mozela R et al. The H3F3 K36M mutant plasia progress to adamantinoma and how should they be
antibody is a sensitive and specific marker for the diagnosis of treated? Bone Joint J. 2017; 99-b; 409–416.
chondroblastoma. Histopathology 2016; 69; 121–127. 102. Keeney GL, Unni KK, Beabout JW, Pritchard DJ. Adamanti-
87. Agaram NP, Zhang L, Dickson BC et al. A molecular study of noma of long bones. A clinicopathologic study of 85 cases.
synovial chondromatosis. Genes Chromosomes Cancer 2020; Cancer 1989; 64; 730–737.
59; 144–151. 103. Houdek MT, Sherman CE, Inwards CY et al. Adamantinoma
88. Totoki Y, Yoshida A, Hosoda F et al. Unique mutation por- of bone: long-term follow-up of 46 consecutive patients. J.
traits and frequent COL2A1 gene alteration in chondrosar- Surg. Oncol. 2018; 118; 1150–1154.
coma. Genome Res. 2014; 24; 1411–1420.

© 2020 John Wiley & Sons Ltd, Histopathology, 78, 644–657.

You might also like