<<

in Diagnosis of Soft Tumours Cyril Fisher

To cite this version:

Cyril Fisher. Immunohistochemistry in Diagnosis of Tumours. Histopathology, Wiley, 2010, 58 (7), pp.1001. ￿10.1111/j.1365-2559.2010.03707.x￿. ￿hal-00613811￿

HAL Id: hal-00613811 https://hal.archives-ouvertes.fr/hal-00613811 Submitted on 6 Aug 2011

HAL is a multi-disciplinary open access L’archive ouverte pluridisciplinaire HAL, est archive for the deposit and dissemination of sci- destinée au dépôt et à la diffusion de documents entific research documents, whether they are pub- scientifiques de niveau recherche, publiés ou non, lished or not. The documents may come from émanant des établissements d’enseignement et de teaching and research institutions in France or recherche français ou étrangers, des laboratoires abroad, or from public or private research centers. publics ou privés. Histopathology

Immunohistochemistry in Diagnosis of Soft Tissue Tumours

ForJournal: Histopathology Peer Review Manuscript ID: HISTOP-08-10-0420

Wiley - Manuscript type: Review

Date Submitted by the 01-Aug-2010 Author:

Complete List of Authors: fisher, cyril; royal marsden hospital, histopathology

Keywords: soft tissue tumours, immunohistochemistry, , diagnosis

Published on behalf of the British Division of the International Academy of Page 1 of 39 Histopathology

Immunohistochemistry in Diagnosis of Soft Tissue Tumours

For PeerCyril FisherReview Royal Marsden Hospital, London UK

Correspondence to:

Prof Cyril Fisher MD DSc FRCPath

Dept of Histopathology The Royal Marsden Hospital 203 Fulham Road London SW3 6JJ UK

Email: [email protected] Tel: +44 207 808 2631 Fax +44 207 808 2578

Running Title : Soft Tissue Tumour Immunohistochemistry

Key Words Immunohistochemistry, sarcoma, diagnosis, soft tissue tumour

1 Published on behalf of the British Division of the International Academy of Pathology Histopathology Page 2 of 39

Abstract

Immunohistochemistry in soft tissue tumours, and especially , is used to identify differentiation in the neoplastic cells. In some cases, specific antigens are expressed; however, an initial panel of is often required in order to establish the broad lineage, with a subsequent, more focussed panel to allow classification.

Immunohistochemical evaluation must be employed with the clinical picture, the morphology, and, Forwhen necessary, Peer other ancillary Review techniques such as molecular genetics and cytogenetics.

While some diagnoses are evident on morphology, many soft tissue are seen microscopically as spindle , epithelioid cell, small round cell or pleomorphic tumours which need further to be characterized. This article reviews selected applications of immunohistochemistry in diagnosis of each of the principal morphological groups, concentrating on areas of most use in daily practice.

2 Published on behalf of the British Division of the International Academy of Pathology Page 3 of 39 Histopathology

Introduction

Soft tissue tumours comprise numerous different tumour types that are classified

according to the type of mesenchymal tissue which they resemble. The main object of

immunohistochemistry in soft tissue neoplasms, and especially sarcomas, is to identify

differentiation in the neoplastic cells. This is straightforward for those tumours that

correspond to normal tissue types, but less so for those sarcomas characterized by specific chromosomalFor translocations Peer since they Review represent novel lineages. In some cases, specific antigens are expressed; however, an initial panel of antibodies is often required

in order to establish the broad lineage, with a subsequent, more focused panel to allow

precise classification.

Tumours with adipocytic or osteochondroid differentiation or vascular space

formation can often be recognized from the microscopic appearances. Many soft

tissue neoplasms, however, show spindle cell, epithelioid cell, small round cell or

pleomorphic morphology on initial examination and require further characterisation by

immunohistochemistry, electron microscopy (although this now has a limited role 1), and

genetic analysis. This article reviews selected applications of immunohistochemistry in

diagnosis in each of the principal morphological groups, concentrating on areas of most

use in daily practice. It is important to remember that immunohistochemical evaluation

must be employed with the clinical picture, the morphology, and, when necessary, other

ancillary techniques such as molecular genetics and cytogenetics.

Spindle Cell Tumours

BENIGN SPINDLE CELL NEOPLASMS

3 Published on behalf of the British Division of the International Academy of Pathology Histopathology Page 4 of 39

The differential diagnosis of the principal benign soft tissue tumours is straightforward when the lineage is obvious. Difficulty is sometimes encountered, especially in core , in assessing , the morphological criteria for which vary according to the tumour type. In specific situations, immunohistochemistry is occasionally of assistance in distinguishing benign from malignant lesions. For example, diffuse S100 positivity in a spindled nerve sheath tumour is more suggestive of schwannoma than malignant peripheralFor nerve Peer sheath tumour Review (MPNST), especially if there are portions of capsule in the . Recently, it has been suggested that a rare CD34-positive cellular fibrous can be distinguished from the commonly CD34-positive dermatofibrosarcoma by the expression of podoplanin (D2-40) in the former but not the latter. 2 Combinations of CD34 with other antibodies can be useful in selected circumstances (Table 1). -related and proliferation markers are of little use in individual cases. The proliferation index with Ki67 can be indicative of malignancy when high, but it can also be high in reactive lesions such as nodular .

INTERMEDIATE SPINDLE CELL NEOPLASMS

Fibromatosis is a non-metastasizing lesion which is a consideration in the differential diagnostic of many types of spindle cell sarcoma. The lesional cells are which are focally positive for (SMA) and occasionally for in scattered cells. can be focally positive but CD34 is negative. Nuclear immunoreactivity for beta- is found in variable numbers of nuclei in about 80% of deep fibromatoses, but only 5% of superficial ones. 3, 4 Non-specific paranuclear staining is common in other myofibroblastic lesions and must not be misinterpreted. Nuclear

4 Published on behalf of the British Division of the International Academy of Pathology Page 5 of 39 Histopathology

beta-catenin positivity can also be found in about 40% of solitary fibrous tumours,

synovial sarcomas and endometrial stromal sarcomas, as well as in hypertrophic scars. It

is absent from gastrointestinal stromal tumour, , inflammatory

myofibroblastic tumour and MPNST, which is of use in the differential diagnosis of intra-

abdominal spindle cell neoplasms. 5 Conversely, is negative for h-

caldesmon, ALK, CD34, CD117 and DOG1. Solitary fibrousFor tumour Peer is diffusely positiveReview for CD34 in 95% of cases, as well as for bcl-2, CD99 and beta-catenin.3, 6-8 Occasional cells expressing (CK),9

epithelial membrane antigen (EMA), or S100 protein can be seen, which does not alter

the diagnosis. Malignant solitary fibrous tumour, manifesting as spindle or polygonal

cell sarcoma contiguous with (or recurrent at the site of a previous) typical solitary

fibrous tumour, usually retains CD34 expression diffusely or focally.10, 11,12

Angiomatoid fibrous histiocytoma expresses desmin in about 50% of cases, but

myogenin is negative. Other antigens that are sometimes positive are h-caldesmon, SMA

(but the morphology is unlike that of smooth muscle tumours), CD99 and EMA.13 This

tumour has three translocations resulting in the fusion EWSR1-CREB1 (the

commonest), EWSR1-ATF1 , or FUS-ATF1 .14 The first two are shared with clear cell

sarcoma (see below).

MALIGNANT SPINDLE CELL NEOPLASMS

These include , malignant peripheral nerve sheath tumour,

leiomyosarcoma, myofibrosarcoma and , and spindle cell variants of

and angiosarcoma. Non-mesenchymal tumours such as sarcomatoid

5 Published on behalf of the British Division of the International Academy of Pathology Histopathology Page 6 of 39

, and follicular dendritic cell sarcoma can present in soft tissue locations and thereby enter the differential diagnosis.

Synovial sarcoma is diagnostically obvious when biphasic. The monophasic form is a spindle cell sarcoma composed of closely packed uniform cells with overlapping nuclei and scanty , while poorly differentiated synovial sarcoma has polygonal or small round cells with high proliferation index. 15 All types of synovial sarcoma exhibit epithelial differentiationFor Peer with focal positivity Review for (including CK7 and CK19) and for EMA in most cases. Diffuse expression of bcl-2 and focal positivity for

CD99, S100 protein, CD56 and calponin are also found in some, and other non-specific markers of occasional diagnostic value include beta-catenin (nuclear) 3 and calretinin

(nuclear). 16 CD34 is almost always negative in synovial sarcoma 17 which is useful in excluding solitary fibrous tumour, since the immunophenotype of these two tumours can otherwise overlap. TLE1, an derived from expression profiling studies, is emerging as a highly sensitive (nuclear) marker for synovial sarcoma of all morphologic types. 18, 19 It is also not wholly specific, since occasional examples of nerve sheath tumours can display positivity. However, its sensitivity means that it is useful in excluding the diagnosis of synovial sarcoma when the result is negative. Reduced expression of INI1 (see below) has been described in some synovial sarcomas. 20

Synovial sarcoma is characterized by a specific chromosomal translocation t(X;18)(p11.2;q11.2), which results in fusion of the SS18 (SYT ) gene from

18 with one of several variants of the SSX gene on the .21 Recently, an antibody to the SS18 has been shown to be sensitive for identification of synovial sarcoma but it is not specific and has not found general application. 22

6 Published on behalf of the British Division of the International Academy of Pathology Page 7 of 39 Histopathology

Malignant peripheral nerve sheath tumours (MPNST) have focal nuclear

positivity for S100 protein in up to 67% of cases, which correlates with ultrastructural

evidence of differentiation. 23 There are no other ‘specific’ indicators of

nerve sheath lineage (although electron microscopy can be contributory in S100 protein-

negative examples 23 ) but many MPNST are focally positive for CD34, and a variable

proportion of cases express GFAP, basic protein, CD57 and . Cytokeratins are occasionally positiveFor but thePeer presence of CK7Review or CK19 is more indicative of synovial sarcoma than of MPNST. 24 Immunoreactivity for NSE or PGP9.5 is sometimes found in

MPNST but these markers are non-specific and of no value in diagnosis. 25 EMA

positivity is occasionally seen in MPNST with Schwannian differentiation, but is most

useful in diagnosis of benign perineurioma 26 and its rare malignant counterpart,27 which

usually lack S100 protein. Another useful marker of perineurial cell differentiation is

claudin-1 which, as a -associated protein,28 has a granular distribution in

normal perineurial cells and in perineurioma.29 Claudin-1 has also been reported as

positive in low-grade fibromyxoid sarcoma 30 which can resemble perineurioma,

especially in cutaneous lesions, so that genetic analysis (see below) can be required for

diagnosis.

Leiomyosarcoma is most often morphologically distinctive, with its non-tapered

cells with cytoplasm and squared-off nuclei, arranged in cellular fascicles in

a distinctive rectilinear pattern. Immunohistochemically, desmin, SMA, muscle specific

actin (MSA), h-caldesmon, calponin and smooth muscle (SMM) are expressed in

the majority of . Some examples co-express cytokeratins (frequently

with dot pattern), EMA and S100 protein but usually only when the muscle-specific

7 Published on behalf of the British Division of the International Academy of Pathology Histopathology Page 8 of 39

antigens are present. CD99 can also be seen in a paranuclear dot pattern. CD34 expression is variable but generally negative in leiomyosarcoma, and CD117 is absent from smooth muscle tumours. A spindle cell expressing SMA but not desmin or h-caldesmon is unlikely to be a smooth muscle tumour and is more likely to be myofibroblastic.31

Myofibrosarcomas are spindle cell or pleomorphic sarcomas which display myofibroblastic differentiation.For Peer32 Low grade Reviewmyofibrosarcomas are composed of tapered cells containing ovoid nuclei and punctate nucleoli, arranged in sheets and fascicles. 33

They are positive for SMA (with a ‘tram-track’ subplasmalemmal accentuation), and usually express calponin and less commonly desmin. H-caldesmon and SMM are negative, distinguishing myofibrosarcoma from leiomyosarcoma. 34 CD34 and S100 protein are also negative; the combination of CD34 and desmin immunoreactivity is found in mammary-type myofibroblastomas, 35 but their ultrastructure is more like that of smooth muscle cells than of true myofibroblasts. 36 Inflammatory myofibroblastic tumour

(IMT) is a low-grade myofibroblastic malignancy with fasciitis-like, fascicular and sclerosing histological patterns. 37 In addition to the usual myofibroblastic markers, about half of IMT, especially those in viscera and in children, express ALK, and recent evidence suggests that lack of ALK is prognostically adverse.38 In IMT, the ALK gene is rearranged with one of six partner genes, most of which result in cytoplasmic staining on immunohistochemistry, except for ALK-RANBP2 . The latter gives a distinctive nuclear membranous distribution as RANBP2 binding are localised at the complex.39

8 Published on behalf of the British Division of the International Academy of Pathology Page 9 of 39 Histopathology

Adult fibrosarcoma is now a rare tumour which is by definition negative for all

lineage markers ( express but this is a wholly non-specific marker

which need not usually be included in diagnostic panels). However, specific subtypes of

fibrosarcoma are positive for CD34, including those arising in dermatofibrosarcoma,

about half of myxofibrosarcomas, and a smaller proportion of myxoinflammatory

fibroblastic sarcomas. 40 Some superficial fibrosarcomas that are strongly CD34 positive but lack an apparentFor dermatofibrosarcomatous Peer Review component have been shown to have the same COL1A1-PDGFRB fusion gene transcripts as dermatofibrosarcoma. 41 In a CD34-

negative fibrosarcoma, an occasional SMA-positive cell can sometimes be seen which is

attributable to focal myofibroblastic differentiation, but diffuse or multifocal SMA

staining indicates categorization as myofibrosarcoma. 32

Low grade fibromyxoid sarcoma occasionally has SMA or CD34 positivity (and,

as previously noted, can express EMA and claudin-130 ), but the diagnosis is best

confirmed by genetic demonstration of the t(7;16)(q33;p11) or FUS-CREB3L2 (or rarely

FUS-CREB3L1 ) fusion gene, 42-44 which has also been reported in some examples of

sclerosing epithelioid fibrosarcoma. 45

Spindle cell rhabdomyosarcoma of juvenile (adolescent) 46, 47 or adult type 48-50 is

diffusely positive for desmin, and at least focally so in nuclei for myogenin and MyoD1.

The latter is less sensitive, but in the rare sclerosing pseudovascular rhabdomyosarcoma,

MyoD1 is more widely expressed than myogenin. 51 It should be noted that these are

nuclear antigens and that cytoplasmic staining for either is non-specific and does not

denote rhabdomyoblastic differentiation. Myoglobin, a previously widely used

9 Published on behalf of the British Division of the International Academy of Pathology Histopathology Page 10 of 39

cytoplasmic marker for differentiation, is of low sensitivity and specificity and is no longer recommended for routine use.

Spindle cell angiosarcoma is an uncommon soft tissue sarcoma of skin or soft tissue in which diagnostic vasoformative areas can be present only focally and therefore absent from a core biopsy. The tumour can be identified by its expression of CD31, and less commonly of CD34 and other endothelial markers (see discussion of epithelioid endothelial neoplasmsFor below). Peer CD117 Review positivity has also been reported in angiosarcoma 52 and in Kaposi sarcoma (KS). The latter, however, demonstrates highly specific immunoreactivity for HHV8 (KSHV) in the nuclei of its spindled and endothelial cells, which is not found in other endothelial neoplasms. 53 Podoplanin (D2-40) is a lymphatic endothelial marker that has a membranous staining pattern in Kaposi sarcoma, but also in some angiosarcomas and gastrointestinal stromal tumours (which also display

CD34 and CD117, as in angiosarcoma).54 Antibodies to vascular endothelial growth factor and its (such as VEGF-C and VEGFR-3) are also immunoreactive in

KS. 55, 56

Follicular dendritic cell sarcoma (FDCS), which sometimes arises in soft tissue sites,57-59 is characterised by fascicles and whorls of cells in which the nuclei typically display prominent membranes and a speckled chromatin pattern. Immunoreactivity for

CD21, CD35 (the two are more sensitive when used together as a cocktail 60 ) and CD23 is diagnostic, and other markers reportedly helpful in identifying FDCS include , clusterin 61 and D2-40.62 Many examples are also positive for S100 protein and EMA. 63

Sarcomatoid carcinoma can express cytokeratins focally, especially those of high molecular weight, but can also be negative. 64 The diagnosis can be facilitated by finding

10 Published on behalf of the British Division of the International Academy of Pathology Page 11 of 39 Histopathology

areas of epithelial differentiation or surface dysplasia, or a nested reticulin pattern.

Spindle cell carcinoma can also express SMA, but desmin, h-caldesmon, CD34 and S100

protein are absent. SMA-positive/CK-negative , which is usually

a pleomorphic neoplasm, can be difficult to distinguish from pleomorphic

myofibrosarcoma without knowledge of the history and the location of the tumour.

Gastrointestinal stromal tumours (GIST) can arise not only in the but also in For extra-gastrointestinal Peer locations, Review especially omentum, mesentery and retroperitoneum. The two predominant patterns are spindled or epithelioid, which have a

similar immunophenotype. GIST typically express CD117, the product of the KIT gene,

which encodes a receptor tyrosine kinase, and which in GIST shows a variety of

prognosis-related . 65-67 A small number of GIST are CD117-negative, and

some of these have mutations in the gene which encodes -derived growth factor

receptor A (PDGFRA). 67 These are often epithelioid gastric GIST. The antibody DOG1,

identified by profiling, has high sensitivity and specificity for GIST and

can identify most KIT-negative tumours. 68, 69 Other antigens found in GIST include

CD34 in 50-100% of tumours (varying with site 70 ), SMA (varying inversely with CD34

expression), h-caldesmon, bcl-2 and, less commonly, desmin or S100 protein. Beta-

catenin is negative in nuclei, in contrast to fibromatosis. 5

Epithelioid and clear cell tumours

Epithelioid soft tissue neoplasms include , and epithelioid variants of

MPNST, leiomyosarcoma, and fibrosarcoma, as well as clear sarcoma, alveolar soft part

sarcoma, PEComa (Table 2), and melanoma and carcinoma when presenting in soft

tissue locations.

11 Published on behalf of the British Division of the International Academy of Pathology Histopathology Page 12 of 39

Epithelioid sarcoma (EpS), of both classical and proximal types, is positive for the epithelial markers cytokeratins and EMA and, in about 50% of cases for CD34. 71

This is particularly useful in making the distinction from primary or metastatic , which are CD34 negative. In addition, CK5/6, a useful marker for carcinomas especially those with squamous differentiation, is detectable in only a small proportion of epithelioid sarcomas.72 EpS generally lacks desmin, S100 protein and FLI- 1, 73 which help For in diagnosis Peer from rhabdomyosarcoma, Review melanoma, and epithelioid angiosarcoma respectively. Antibodies to INI1 are negative in about 90% of EpS, in malignant rhabdoid tumours (MRT), in about half the cases of epithelioid MPNST, and in some extraskeletal myxoid with rhabdoid cytomorphology.74-76 This relates to the location of the SMARC/INI gene on chromosome 22q, which is mutated or deleted in MRT and EpS.74 Absence of INI1 is extremely useful for diagnosis of these tumours since all other epithelioid or polygonal cell tumours in the differential diagnosis display nuclear positivity for this marker. MRT can be distinguished from EpS by absence of CD34, although clinical data are required to distinguish MRT from CD34- negative EpS.

Epithelioid MPNST differs from its spindle cell counterpart in displaying diffuse

S100 protein expression in most cases.77 It differs from melanoma in having more uniform cytological features, frequent association with a typical spindle cell nerve sheath component, and absence of melanoma-specific antigens. INI-negative cases should not be misdiagnosed as epithelioid sarcoma, or INI-positive cases as epithelioid angiosarcoma, since both of these tumour types are S100 protein negative.

12 Published on behalf of the British Division of the International Academy of Pathology Page 13 of 39 Histopathology

Epithelioid endothelial neoplasms manifest varying combinations of vascular

endothelial cell markers. These include CD34, CD31, FLI-1 (in nuclei), FVIIIRAg,

and CD117. In epithelioid angiosarcoma and malignant variants of

epithelioid haemangioendothelioma these antigens are selectively expressed so that not

all are detectable by immunohistochemistry. CD31 and FVIIIRAg are the most specific,

although the latter is not very sensitive. These tumours show diffuse nuclear immunoreactivity For for INI1, 76 Peer and D2-40 isReview positive in some cases.54 In addition, epithelioid angiosarcoma is frequently cytokeratin-positive, while usually lacking

EMA. 78, 79 Epithelioid sarcoma, especially the proximal variant, can be morphologically

similar to epithelioid angiosarcoma and can also co-express CD34 and cytokeratins, but

lacks CD31, FVIIIRAg, FLI-1, and INI1. Epithelioid sarcoma-like

haemangioendothelioma is a very rare low-grade spindle cell neoplasm that displays

positivity for cytokeratins, CD31, and FL1-1, but lacks CD34 (Table 1).80, 81

PEComa is the term used for a group of clinically disparate tumours composed of

supposed perivascular epithelioid cells – spindle or polygonal cells with clear or granular

cytoplasm and uniform nuclei, which are arranged in nests around sinusoidal vessels.82, 83

Members of the PEComa ‘family’ include , ,

clear cell ‘sugar’ tumour of and other sites, and clear cell myomelanocytic tumour.

PEComas co-express SMA and the melanocytic antigens HMB-45, melan-A, and micro-

ophthalmia factor (MITF). It has become recognized that PEComas can

also focally express S100 protein, desmin and h-caldesmon as well as TFE3 in some

cases. 84 Most PEComas are benign but malignant examples are increasingly reported.84, 85

The principal differential diagnosis (Table 2) is with clear sarcoma of and

13 Published on behalf of the British Division of the International Academy of Pathology Histopathology Page 14 of 39

aponeuroses which also expresses melanoma-specific antigens, but which has diffuse

S100 protein positivity, lacks SMA and h-caldesmon, and is characterized by a specific translocation t(12;22)(q13;q12) with fusion of EWS and ATF-1 genes. 86 The morphologically similar of the gastrointestinal tract with osteoclast- like giant cells is also S100 protein positive but lacks melan-A and HMB45 expression, and usually has t(2;22)(q33;q12) with EWS-CREB1 fusion. 87 However, examples of both fusions have beenFor described Peer at both sites. Review88 The same fusions are also found in angiomatoid fibrous histiocytoma, 89 representing one of several examples of promiscuity among gene fusions. 90

Alveolar soft part sarcoma has long been an enigmatic tumour in which the occasional report of immunoreactivity for desmin and, for a time, MyoD1, led to the assumption that the tumour showed skeletal muscle differentiation. This tumour, however, is now known to be a translocation-associated sarcoma characterized by t(X;17)(p11;q25) with fusion of TFE3 and ASPL genes.91 Antibodies to TFE3 are available which demonstrate nuclear positivity in alveolar soft part sarcoma; similar staining is also found in some translocation-associated carcinomas of , in some

PEComas and in examples of granular cell tumour.92

Pleomorphic tumours

Pleomorphic sarcomas include (pleomorphic and dedifferentiated), rhabdomyosarcoma, MPNST, leiomyosarcoma, myofibrosarcoma, and undifferentiated pleomorphic sarcoma (also known as pleomorphic malignant fibrous histiocytoma or

14 Published on behalf of the British Division of the International Academy of Pathology Page 15 of 39 Histopathology

MFH). Undifferentiated or sarcomatoid carcinoma, melanoma and rarely lymphoma can

also have similar microscopic appearances.

Pleomorphic neoplasms are composed predominantly of pleomorphic spindle,

polygonal and giant cells with mitoses, necrosis and . In addition the

specific subtypes have foci of differentiation perceived either morphologically

(lipoblasts) or immunohistochemically (skeletal, smooth muscle, melanocytic or epithelial antigens).For In many Peercases without apparentReview lineage, the pleomorphic cells have ultrastructural features of with a variable proportion of cells showing

myofibroblastic differentiation. 93 Thus, MFH and undifferentiated areas of other

pleomorphic sarcomas can display focal SMA positivity, with a characteristic

subplasmalemmal linear distribution imparting a ‘tram-track’ appearance. 94 Desmin

positivity is occasionally seen but h-caldesmon is absent. It is important to identify

pleomorphic sarcomas with any type of myogenic differentiation at

immunohistochemical level since, counter-intuitively, they have a worse prognosis than

undifferentiated pleomorphic sarcomas. 95, 96 Pleomorphic rhabdomyosarcoma usually

has diffuse desmin positivity and focal nuclear staining for myogenin. MyoD1 is a less

sensitive marker except in sclerosing pseudovascular rhabdomyosarcoma.51, 97 In

pleomorphic leiomyosarcoma , typical fascicular areas occupying <25% of the

pleomorphic tumour are usually required for the diagnosis.98 However,

immunohistochemical evidence of smooth muscle differentiation (desmin, SMA and h-

caldesmon) without corresponding morphological features is associated with a worse

prognosis.95, 96 MFH-like undifferentiated sarcomas with widespread SMA positivity can

be termed pleomorphic myofibrosarcomas; the distinction is important since

15 Published on behalf of the British Division of the International Academy of Pathology Histopathology Page 16 of 39

myofibroblastic differentiation, although less well-studied than myoid differentiation, also appears prognostically unfavorable. 94

Dedifferentiated liposarcoma , like well differentiated liposarcoma, has MDM2 and CDK4 amplification, detectable by fluorescence in situ hybridization (FISH) and also by immunohistochemistry.99 Most retroperitoneal (and some extremity 100 ) undifferentiated pleomorphic sarcomas (MFH) and myxofibrosarcomas express these markers (in nuclei)For and are nowPeer considered Review to represent dedifferentiated even when no well-differentiated component remains. 100-103 In addition, dedifferentiated liposarcoma can express actin, desmin and CD34 focally in the pleomorphic areas,101 which should not be misinterpreted as leiomyosarcoma.

Sarcomatoid carcinoma can appear as a pleomorphic neoplasm. This usually expresses cytokeratins in a multifocal or diffuse distribution that is best demonstrated with a broad spectrum antibody or ‘cytokeratin cocktail’. Even very focal cytokeratin expression should prompt consideration of a primary carcinoma, and it can be accepted as evidence of epithelial differentiation in an organ-based (visceral) pleomorphic neoplasm.

It should be remembered, however, that some undifferentiated pleomorphic sarcomas in soft tissue have (aberrant) expression of cytokeratin, usually in a few scattered cells. 104,

105 Also, as previously noted, spindled carcinomas can acquire myofibroblastic differentiation and thereby be focally positive for SMA (but not desmin or h-caldesmon).

As always, the final diagnosis must summate all data including clinical and radiological findings.

Small round cell tumours

16 Published on behalf of the British Division of the International Academy of Pathology Page 17 of 39 Histopathology

This group of soft tissue neoplasms includes Ewing sarcoma/primitive

neuroectodermal tumour (ES), desmoplastic small round cell tumour (DSRCT), alveolar

rhabdomyosarcoma (ARMS), (NB), and poorly differentiated synovial

sarcoma (PDSS). Most of these occur predominantly in childhood and adolescence, but

are also seen sporadically in adults. 106 Several of these sarcoma subtypes have specific,

translocations and consequent fusion genes that allow precise diagnosis, and in some instances provide For prognostic Peer information, usingReview FISH or PCR-based methods. 90, 107 A molecular pathology service is not always readily available, but the immunophenotypes

of these small round cell sarcomas differ sufficiently to enable diagnosis in many cases.

Lymphomas, especially of lymphoblastic type, must also be considered in the differential

diagnosis, and TdT should be part of the diagnostic panel in the appropriate clinical

setting. Similarly leukaemic deposits, and small cell carcinomas might need exclusion by

immunostaining for myeloid markers or cytokeratins, neuroendocrine antigens and TTF1

respectively.

Ewing sarcoma is a translocation-associated tumour, which can arise in or

extraskeletal sites; examples with evidence of neural differentiation are termed PNET. 90

The large majority of ES display diffuse membranous staining for CD99, and some

express proteins and NSE. About 70% show nuclear immunoreactivity for

FLI-1. 108 A small number are also positive for S100 protein or display dot positivity with

broad spectrum anticytokeratin antibodies and membranous staining for EMA. CD56 is

usually negative, unlike in small cell neuroendocrine carcinoma and PDSS.109 The latter

also expresses TLE1, but some cases have a similar immunophenotype to ES and

although there are subtle morphological differences (ES tends to be more uniform and

17 Published on behalf of the British Division of the International Academy of Pathology Histopathology Page 18 of 39

lacks intercellular reticulin), genetic analysis may be required to make the diagnosis. The morphologic spectrum of ES is expanding due to increased recognition of atypical variants aided, in part, by molecular studies.110, 111 Although there is a wide range of available antibodies at least three of a panel composed of CD99, FLI-1, HNK1, and -1 reportedly show immunoreactivity in all cases with caveolin-1 being positive in CD99-negative cases. 111 DesmoplasticFor small roundPeer cell tumour Review was originally described in the abdomen and pelvis in adolescent males, and is now recognized in other locations. It shows presumed primitive mesothelial differentiation, and has a characteristic t(11;22)(p13;q12) rearrangement with formation of an EWS-WT1 fusion gene. Immunohistochemically,

DSRCT shows nuclear staining with antibodies to the C-terminal end of WT1 as well as multilineage differentiation with dot positivity for desmin, cytokeratins (CK20 and

CK5/6 are negative), and neural markers including , , , and CD56. 112, 113 However, not all lineage markers are expressed in every case.114 DSRCT is usually negative for CD99 but 10-20% (and especially those with variant translocations) can be positive which does not therefore exclude the diagnosis. 115, 116

Alveolar rhabdomyosarcoma , as with other rhabdomyosarcoma subtypes, has diffuse cytoplasmic staining for desmin. Myogenin is expressed in a majority of tumour cell nuclei and is more sensitive than MyoD1. 117 CD56 is also positive as in other types of rhabdomyosarcoma. 70-90% of ARMS have either t(2;13)(q35;q14) creating a PAX3-

FOXO1 fusion gene or t(1;13)(p36;q14) with resulting PAX7-FOXO1 fusion. 118

18 Published on behalf of the British Division of the International Academy of Pathology Page 19 of 39 Histopathology

Immunohistochemistry with an antibody to PAX5 can detect all translocation-positive

cases of either type.119

Neuroblastoma can be recognized not only by its clinical context (the vast

majority are in patients <4 years old and arise in adrenal or other sympathetic nervous

system locations), but also by its expression of NB84, NSE, CD56 and neurofilament

proteins. 120 Antibodies to these markers, which can be used as a panel, are particularly useful in detectionFor of residual Peer Review in bone marrow specimens taken after . Neuroblastoma is negative for cytokeratins, desmin and CD99, which can

be particularly helpful in diagnosis of rare examples in adults. 121

Adipose tumours

The distinction between and atypical lipomatous tumour (ALT)/well

differentiated liposarcoma can be difficult since it depends on focal morphologic

subtleties which are not always represented in the samples taken. The diagnosis is

facilitated by the demonstration of immunohistochemical positivity for products of the

murine double minute type 2 ( MDM2) and cyclin-dependent kinase 4 ( CDK4 ) genes

which are overexpressed in ALT as a result of amplification in the 12q14-15

chromosomal region. All ALT are immunoreactive in nuclei for MDM2 (often focally)

and 91% for CDK4 (more diffusely).99 These have application not only in the differential

diagnosis of lipoma and ALT, but also in determining whether the fatty tissue at the

margin of excision of ALT is neoplastic or not. As mentioned above, in dedifferentiated

liposarcoma (DDL), which is genetically related to ALT, CDK4 and MDM2 are similarly

overexpressed.102, 122 Their specificity in DDL is reduced since a small proportion of

19 Published on behalf of the British Division of the International Academy of Pathology Histopathology Page 20 of 39

MPNST, myxofibrosarcomas and embryonal are immunoreactive with both 99 but, apart from morphological considerations, the diagnosis of DDL is supported by positivity for these two markers in the differentiated adipose tissue adjacent to the pleomorphic neoplasm.

Conclusion Existing antibody Forpanels, when Peer correctly selected, Review have a major role in diagnosis of soft tissue sarcomas. Diagnostically useful new antibodies, such as DOG1 and TLE1, are emerging from gene profiling studies, and in addition to the usual reagents, antibodies to fusion gene products might theoretically be of value in detecting translocation sarcomas.

For example, antibodies raised against TLS/EWS-CHOP chimeric oncoproteins have shown high specificity and sensitivity for myxoid and round cell liposarcoma. 123 These approaches can be expected to lead to improved diagnosis of soft tissue tumours and perhaps to more efficient use of immunohistochemistry with limited and therefore less expensive panels.

Acknowledgment

I acknowledge NHS funding to the NIHR Biomedical Research Centre .

20 Published on behalf of the British Division of the International Academy of Pathology Page 21 of 39 Histopathology

References

1. Fisher C. The comparative roles of electron microscopy and

immunohistochemistry in the diagnosis of soft tissue tumours. Histopathology

2006; 48 ;32-41.

2. Bandarchi B, Ma L, Marginean C, Hafezi S, Zubovits J, Rasty G. D2-40, a novel immunohistochemicalFor marker Peerin differentiating Review from dermatofibrosarcoma protuberans. Mod Pathol 2010; 23 ;434-438.

3. Ng TL, Gown AM, Barry TS et al. Nuclear beta-catenin in mesenchymal tumors.

Mod Pathol 2005; 18 ;68-74.

4. Bhattacharya B, Dilworth HP, Iacobuzio-Donahue C et al. Nuclear beta-catenin

expression distinguishes deep fibromatosis from other benign and malignant fibroblastic

and myofibroblastic lesions. Am J Surg Pathol 2005; 29 ;653-659.

5. Montgomery E, Torbenson MS, Kaushal M, Fisher C, Abraham SC. Beta-catenin

immunohistochemistry separates mesenteric fibromatosis from gastrointestinal stromal

tumor and sclerosing mesenteritis. Am J Surg Pathol 2002; 26 ;1296-1301.

6. Renshaw AA PG, Corson JM. CD34 and AE1/AE3. Diagnostic discriminants in

the distinction of of the pleura from sarcomatoid .

Appl Immunohistochem 1994; 2;94-102.

7. Chan JK. Solitary fibrous tumour--everywhere, and a diagnosis in vogue.

Histopathology 1997; 31 ;568-576.

8. Chilosi M, Facchetti F, Tos APD et al. bcl-2 expression in pleural and

extrapleural solitary fibrous tumours. J Pathol 1997; 181 ;362-367.

21 Published on behalf of the British Division of the International Academy of Pathology Histopathology Page 22 of 39

9. McGuire LJ, Chan HS, Pang J. Solitary fibrous tumor of pleura: expression of cytokeratins. Pathology 1990; 22 ;232-234.

10. Nielsen GP, O'Connell JX, Dickersin GR, Rosenberg AE. Solitary fibrous tumor of soft tissue: a report of 15 cases, including 5 malignant examples with light microscopic, immunohistochemical, and ultrastructural data. Mod Pathol 1997; 10 ;1028-

1037. 11. Vallat-DecouvelaereFor AV,Peer Dry SM, Fletcher Review CD. Atypical and malignant solitary fibrous tumors in extrathoracic locations: evidence of their comparability to intra-thoracic tumors. Am J Surg Pathol 1998; 22 ;1501-1511.

12. Fisher C. Low-grade sarcomas with CD34-positive fibroblasts and low-grade myofibroblastic sarcomas. Ultrastruct Pathol 2004; 28 ;291-305.

13. Thway K. Angiomatoid fibrous histiocytoma: a review with recent genetic findings. Arch Pathol Lab Med 2008; 132 ;273-277.

14. Antonescu CR, Dal Cin P, Nafa K et al. EWSR1-CREB1 is the predominant gene fusion in angiomatoid fibrous histiocytoma. Genes 2007; 46 ;1051-

1060.

15. Fisher C. Synovial sarcoma. Ann Diagn Pathol 1998; 2;401-421.

16. Miettinen M, Limon J, Niezabitowski A, Lasota J. Calretinin and other mesothelioma markers in synovial sarcoma: analysis of antigenic similarities and differences with malignant mesothelioma. Am J Surg Pathol 2001; 25 ;610-617.

17. Pelmus M, Guillou L, Hostein I, Sierankowski G, Lussan C, Coindre JM.

Monophasic fibrous and poorly differentiated synovial sarcoma: immunohistochemical

22 Published on behalf of the British Division of the International Academy of Pathology Page 23 of 39 Histopathology

reassessment of 60 t(X;18)(SYT-SSX)-positive cases. Am J Surg Pathol 2002; 26 ;1434-

1440.

18. Kosemehmetoglu K, Vrana JA, Folpe AL. TLE1 expression is not specific for

synovial sarcoma: a whole section study of 163 soft tissue and bone neoplasms. Mod

Pathol 2009; 22 ;872-878.

19. Jagdis A, Rubin BP, Tubbs RR, Pacheco M, Nielsen TO. Prospective evaluation of TLE1 as a diagnosticFor immunohistochemical Peer Review marker in synovial sarcoma. Am J Surg Pathol 2009; 33 ;1743-1751.

20. Kohashi K, Oda Y, Yamamoto H et al. Reduced expression of SMARCB1/INI1

protein in synovial sarcoma. Mod Pathol 2010; 23 ;981-990.

21. Ladanyi M, Antonescu CR, Leung DH et al. Impact of SYT-SSX fusion type on

the clinical behavior of synovial sarcoma: a multi-institutional retrospective study of 243

patients. Cancer Res 2002; 62 ;135-140.

22. He R, Patel RM, Alkan S et al. Immunostaining for SYT protein discriminates

synovial sarcoma from other soft tissue tumors: analysis of 146 cases. Mod Pathol

2007; 20 ;522-528.

23. Fisher C, Carter RL, Ramachandra S, Thomas DM. Peripheral nerve sheath

differentiation in malignant soft tissue tumours: an ultrastructural and

immunohistochemical study. Histopathology 1992; 20 ;115-125.

24. Smith TA, Machen SK, Fisher C, Goldblum JR. Usefulness of cytokeratin subsets

for distinguishing monophasic synovial sarcoma from malignant peripheral nerve sheath

tumor. Am J Clin Pathol 1999; 112 ;641-648.

23 Published on behalf of the British Division of the International Academy of Pathology Histopathology Page 24 of 39

25. Campbell LK, Thomas JR, Lamps LW, Smoller BR, Folpe AL. Protein gene product 9.5 (PGP 9.5) is not a specific marker of neural and nerve sheath tumors: an immunohistochemical study of 95 mesenchymal neoplasms. Mod Pathol 2003; 16 ;963-

969.

26. Hornick JL, Fletcher CD. Soft tissue perineurioma: clinicopathologic analysis of

81 cases including those with atypical histologic features. Am J Surg Pathol 2005; 29 ;845- 858. For Peer Review 27. Hirose T, Scheithauer BW, Sano T. Perineurial malignant peripheral (MPNST). A clinicopathologic, immunohistochemical, and ultrastructural study of seven cases. Am J Surg Pathol 1998; 22 ;1368-1378.

28. Billings SD, Walsh SV, Fisher C, Nusrat A, Weiss SW, Folpe AL. Aberrant expression of tight junction-related proteins ZO-1, claudin-1 and occludin in synovial sarcoma: an immunohistochemical study with ultrastructural correlation. Mod Pathol

2004; 17 ;141-149.

29. Folpe AL, Billings SD, McKenney JK, Walsh SV, Nusrat A, Weiss SW.

Expression of claudin-1, a recently described tight junction-associated protein, distinguishes soft tissue perineurioma from potential mimics. Am J Surg Pathol

2002; 26 ;1620-1626.

30. Thway K, Fisher C, Debiec-Rychter M, Calonje E. Claudin-1 is expressed in perineurioma-like low-grade fibromyxoid sarcoma. Hum Pathol 2009; 40 ;1586-1590.

31. Ceballos KM, Nielsen GP, Selig MK, O'Connell JX. Is anti-h-caldesmon useful for distinguishing smooth muscle and myofibroblastic tumors? An immunohistochemical study. Am J Clin Pathol 2000; 114 ;746-753.

24 Published on behalf of the British Division of the International Academy of Pathology Page 25 of 39 Histopathology

32. Fisher C. Myofibrosarcoma. Virchows Arch 2004; 445 ;215-223.

33. Eyden B, Banerjee SS, Shenjere P, Fisher C. The and its tumours. J

Clin Pathol 2009; 62 ;236-249.

34. Fisher C. Myofibroblastic . Adv Anat Pathol 2004; 11 ;190-201.

35. McMenamin ME, Fletcher CD. Mammary-type myofibroblastoma of soft tissue: a

tumor closely related to . Am J Surg Pathol 2001; 25 ;1022-1029. 36. Eyden BP, ForShanks JH, PeerIoachim E, Ali ReviewHH, Christensen L, Howat AJ. Myofibroblastoma of breast: evidence favoring smooth-muscle rather than

myofibroblastic differentiation. Ultrastruct Pathol 1999; 23 ;249-257.

37. Gleason BC, Hornick JL. Inflammatory myofibroblastic tumours: where are we

now? J Clin Pathol 2008; 61 ;428-437.

38. Coffin CM, Hornick JL, Fletcher CD. Inflammatory myofibroblastic tumor:

comparison of clinicopathologic, histologic, and immunohistochemical features including

ALK expression in atypical and aggressive cases. Am J Surg Pathol 2007; 31 ;509-520.

39. Wilken N, Senecal JL, Scheer U, Dabauvalle MC. Localization of the Ran-GTP

binding protein RanBP2 at the cytoplasmic side of the nuclear pore complex. Eur J Cell

Biol 1995; 68 ;211-219.

40. Meis-Kindblom JM, Kindblom LG. Acral myxoinflammatory fibroblastic

sarcoma: a low-grade tumor of the hands and feet. Am J Surg Pathol 1998; 22 ;911-924.

41. Sheng WQ, Hashimoto H, Okamoto S et al. Expression of COL1A1-PDGFB

fusion transcripts in superficial adult fibrosarcoma suggests a close relationship to

dermatofibrosarcoma protuberans. J Pathol 2001; 194 ;88-94.

25 Published on behalf of the British Division of the International Academy of Pathology Histopathology Page 26 of 39

42. Reid R, de Silva MV, Paterson L, Ryan E, Fisher C. Low-grade fibromyxoid sarcoma and hyalinizing spindle cell tumor with giant rosettes share a common t(7;16)(q34;p11) translocation. Am J Surg Pathol 2003; 27 ;1229-1236.

43. Mertens F, Fletcher CD, Antonescu CR et al. Clinicopathologic and molecular genetic characterization of low-grade fibromyxoid sarcoma, and cloning of a novel

FUS/CREB3L1 fusion gene. Lab Invest 2005; 85 ;408-415. 44. MatsuyamaFor A, Hisaoka Peer M, Shimajiri S Review et al. Molecular detection of FUS- CREB3L2 fusion transcripts in low-grade fibromyxoid sarcoma using formalin-fixed, paraffin-embedded tissue specimens. Am J Surg Pathol 2006; 30 ;1077-1084.

45. Guillou L, Benhattar J, Gengler C et al. Translocation-positive low-grade fibromyxoid sarcoma: clinicopathologic and molecular analysis of a series expanding the morphologic spectrum and suggesting potential relationship to sclerosing epithelioid fibrosarcoma: a study from the French Sarcoma Group. Am J Surg Pathol 2007; 31 ;1387-

1402.

46. Cavazzana AO, Schmidt D, Ninfo V et al. Spindle cell rhabdomyosarcoma. A prognostically favorable variant of rhabdomyosarcoma. Am J Surg Pathol 1992; 16 ;229-

235.

47. Leuschner I. Spindle cell rhabdomyosarcoma: histologic variant of embryonal rhabdomyosarcoma with association to favorable prognosis. Curr Top Pathol

1995; 89 ;261-272.

48. Nascimento AF, Fletcher CD. Spindle cell rhabdomyosarcoma in adults. Am J

Surg Pathol 2005; 29 ;1106-1113.

26 Published on behalf of the British Division of the International Academy of Pathology Page 27 of 39 Histopathology

49. Mentzel T, Kuhnen C. Spindle cell rhabdomyosarcoma in adults:

clinicopathological and immunohistochemical analysis of seven new cases. Virchows

Arch 2006; 449 ;554-560.

50. Stock N, Chibon F, Binh MB et al. Adult-type rhabdomyosarcoma: analysis of 57

cases with clinicopathologic description, identification of 3 morphologic patterns and

prognosis. Am J Surg Pathol 2009; 33 ;1850-1859. 51. Folpe AL, McKenneyFor JK,Peer Bridge JA, ReviewWeiss SW. Sclerosing rhabdomyosarcoma in adults: report of four cases of a hyalinizing, matrix-rich variant of rhabdomyosarcoma

that may be confused with , , or angiosarcoma. Am J Surg

Pathol 2002; 26 ;1175-1183.

52. Miettinen M, Lasota J. KIT (CD117): a review on expression in normal and

neoplastic tissues, and mutations and their clinicopathologic correlation. Appl

Immunohistochem Mol Morphol 2005; 13 ;205-220.

53. Boshoff C, Schulz TF, Kennedy MM et al. Kaposi's sarcoma-associated

herpesvirus infects endothelial and spindle cells. Nat Med 1995; 1;1274-1278.

54. Ordonez NG. Podoplanin: a novel diagnostic immunohistochemical marker. Adv

Anat Pathol 2006; 13 ;83-88.

55. Dupin N, Fisher C, Kellam P et al. Distribution of herpesvirus-8 latently

infected cells in Kaposi's sarcoma, multicentric Castleman's disease, and primary effusion

lymphoma. Proc Natl Acad Sci U S A 1999; 96 ;4546-4551.

56. Folpe AL, Veikkola T, Valtola R, Weiss SW. Vascular endothelial growth factor

receptor-3 (VEGFR-3): a marker of vascular tumors with presumed lymphatic

27 Published on behalf of the British Division of the International Academy of Pathology Histopathology Page 28 of 39

differentiation, including Kaposi's sarcoma, kaposiform and Dabska-type hemangioendotheliomas, and a subset of angiosarcomas. Mod Pathol 2000; 13 ;180-185.

57. Chan JK, Fletcher CD, Nayler SJ, Cooper K. Follicular dendritic cell sarcoma.

Clinicopathologic analysis of 17 cases suggesting a malignant potential higher than currently recognized. Cancer 1997; 79 ;294-313.

58. Fisher C, Magnusson B, Hardarson S, Smith ME. Myxoid variant of follicular dendritic cell sarcomaFor arising inPeer the breast. Ann Review Diagn Pathol 1999; 3;92-98. 59. Biddle DA, Ro JY, Yoon GS et al. Extranodal follicular dendritic cell sarcoma of the head and neck region: three new cases, with a review of the literature. Mod Pathol

2002; 15 ;50-58.

60. Chan JKC, Fletcher CDM, Nayler SJ, Cooper K. Follicular dendritic cell sarcoma.

Clinicopathologic analysis of 17 cases suggesting a malignant potential higher than currently recognized. Cancer 1997; 79 ;294-313.

61. Grogg KL, Macon WR, Kurtin PJ, Nascimento AG. A survey of clusterin and fascin expression in sarcomas and spindle cell neoplasms: strong clusterin immunostaining is highly specific for follicular dendritic cell tumor. Mod Pathol

2005; 18 ;260-266.

62. Yu H, Gibson JA, Pinkus GS, Hornick JL. Podoplanin (D2-40) is a novel marker for follicular dendritic cell tumors. Am J Clin Pathol 2007; 128 ;776-782.

63. Chan JKC. Proliferative lesions of follicular dendritic cells: an overview, including a detailed account of follicular dendritic cell sarcoma, a neoplasm with many faces and uncommon etiologic associations. Adv Anat Pathol 1997; 6;387-411.

28 Published on behalf of the British Division of the International Academy of Pathology Page 29 of 39 Histopathology

64. Balercia G, Bhan AK, Dickersin GR. Sarcomatoid carcinoma: an ultrastructural

study with light microscopic and immunohistochemical correlation of 10 cases from

various anatomic sites. Ultrastructural Pathol 1995; 19 ;249-263.

65. Miettinen M, Lasota J. Gastrointestinal stromal tumors: review on morphology,

molecular pathology, prognosis, and differential diagnosis. Arch Pathol Lab Med

2006; 130 ;1466-1478. 66. Andersson ForJ, Bumming Peer P, Meis-Kindblom Review JM et al. Gastrointestinal stromal tumors with KIT 11 deletions are associated with poor prognosis. Gastroenterology

2006; 130 ;1573-1581.

67. Lasota J, Miettinen M. KIT and PDGFRA mutations in gastrointestinal stromal

tumors (GISTs). Semin Diagn Pathol 2006; 23 ;91-102.

68. Espinosa I, Lee CH, Kim MK et al. A novel monoclonal antibody against DOG1

is a sensitive and specific marker for gastrointestinal stromal tumors. Am J Surg Pathol

2008; 32 ;210-218.

69. West RB, Corless CL, Chen X et al. The novel marker, DOG1, is expressed

ubiquitously in gastrointestinal stromal tumors irrespective of KIT or PDGFRA

status. Am J Pathol 2004; 165 ;107-113.

70. Miettinen M, Sobin LH, Sarlomo-Rikala M. Immunohistochemical spectrum of

GISTs at different sites and their differential diagnosis with a reference to CD117 (KIT).

Mod Pathol 2000; 13 ;1134-1142.

71. Fisher C. Epithelioid sarcoma of Enzinger. Adv Anat Pathol 2006; 13 ;114-121.

29 Published on behalf of the British Division of the International Academy of Pathology Histopathology Page 30 of 39

72. Lin L, Skacel M, Sigel JE et al. Epithelioid sarcoma: an immunohistochemical analysis evaluating the utility of cytokeratin 5/6 in distinguishing superficial epithelioid sarcoma from spindled . J Cutan Pathol 2003; 30 ;114-117.

73. Folpe AL, Chand EM, Goldblum JR, Weiss SW. Expression of Fli-1, a nuclear transcription factor, distinguishes vascular neoplasms from potential mimics. Am J Surg

Pathol 2001; 25 ;1061-1066. 74. Modena P, ForLualdi E, FacchinettiPeer F et al.Review SMARCB1/INI1 tumor suppressor gene is frequently inactivated in epithelioid sarcomas. Cancer Res 2005; 65 ;4012-4019.

75. Orrock JM, Abbott JJ, Gibson LE, Folpe AL. INI1 and GLUT-1 expression in epithelioid sarcoma and its cutaneous neoplastic and nonneoplastic mimics. Am J

Dermatopathol 2009; 31 ;152-156.

76. Hornick JL, Dal Cin P, Fletcher CD. Loss of INI1 expression is characteristic of both conventional and proximal-type epithelioid sarcoma. Am J Surg Pathol

2009; 33 ;542-550.

77. Laskin WB, Weiss SW, Bratthauer GL. Epithelioid variant of malignant peripheral nerve sheath tumor (malignant epithelioid schwannoma). Am J Surg Pathol

1991; 15 ;1136-1145.

78. Gray MH, Rosenberg AE, Dickersin GR, Bhan AK. Cytokeratin expression in epithelioid vascular neoplasms. Hum Pathol 1990; 21 ;212-217.

79. Meis-Kindblom JM, Kindblom LG. Angiosarcoma of soft tissue: a study of 80 cases. Am J Surg Pathol 1998; 22 ;683-697.

80. Billings SD, Folpe AL, Weiss SW. Epithelioid sarcoma-like hemangioendothelioma. Am J Surg Pathol 2003; 27 ;48-57.

30 Published on behalf of the British Division of the International Academy of Pathology Page 31 of 39 Histopathology

81. Watabe A, Okuyama R, Hashimoto A et al. Epithelioid sarcoma-like

haemangioendothelioma: a case report. Acta Derm Venereol 2009; 89 ;208-209.

82. Pea M, Martignoni G, Zamboni G, Bonetti F. Perivascular epithelioid cell. Am J

Surg Pathol 1996; 20 ;1149-1153.

83. Folpe AL, Kwiatkowski DJ. Perivascular epithelioid cell neoplasms: pathology

and . Hum Pathol 2010; 41 ;1-15. 84. Folpe AL, MentzelFor T, LehrPeer HA, Fisher Review C, Balzer BL, Weiss SW. Perivascular epithelioid cell neoplasms of soft tissue and gynecologic origin: a clinicopathologic study

of 26 cases and review of the literature. Am J Surg Pathol 2005; 29 ;1558-1575.

85. Harris GC, McCulloch TA, Perks G, Fisher C. Malignant perivascular epithelioid

cell tumour ("PEComa") of soft tissue: a unique case. Am J Surg Pathol 2004; 28 ;1655-

1658.

86. Meis-Kindblom JM. Clear cell sarcoma of tendons and aponeuroses: a historical

perspective and tribute to the man behind the entity. Adv Anat Pathol 2006; 13 ;286-292.

87. Kosemehmetoglu K FA. Clear cell sarcoma of tendons and aponeuroses, and

osteoclast-rich tumour of the gastrointestinal tract with features resembling clear cell

sarcoma of soft parts: a review and update. J Clin Pathol 2010; 63 ;416-423.

88. Wang WL, Mayordomo E, Zhang W et al. Detection and characterization of

EWSR1/ATF1 and EWSR1/CREB1 chimeric transcripts in clear cell sarcoma (melanoma

of soft parts). Mod Pathol 2009; 22 ;1201-1209.

89. Rossi S, Szuhai K, Ijszenga M et al. EWSR1-CREB1 and EWSR1-ATF1 fusion

genes in angiomatoid fibrous histiocytoma. Clin Cancer Res 2007; 13 ;7322-7328.

31 Published on behalf of the British Division of the International Academy of Pathology Histopathology Page 32 of 39

90. Ordonez JL, Osuna D, Herrero D, de Alava E, Madoz-Gurpide J. Advances in

Ewing's sarcoma research: where are we now and what lies ahead? Cancer Res

2009; 69 ;7140-7150.

91. Ladanyi M, Lui MY, Antonescu CR et al. The der(17)t(X;17)(p11;q25) of human alveolar soft part sarcoma fuses the TFE3 transcription factor gene to ASPL, a novel gene at 17q25. 2001; 20 ;48-57. 92. Argani P, LalFor P, Hutchinson Peer B, Lui MY, Review Reuter VE, Ladanyi M. Aberrant nuclear immunoreactivity for TFE3 in neoplasms with TFE3 gene fusions: a sensitive and specific immunohistochemical assay. Am J Surg Pathol 2003; 27 ;750-761.

93. Fisher C. The value of electron microscopy and immunohistochemistry in the diagnosis of soft tissue sarcomas: a study of 200 cases. Histopathology 1990; 16 ;441-445.

94. Montgomery E, Fisher C. Myofibroblastic differentiation in malignant fibrous histiocytoma (pleomorphic myofibrosarcoma): a clinicopathological study.

Histopathology 2001; 38 ;499-509.

95. Deyrup AT, Haydon RC, Huo D et al. Myoid differentiation and prognosis in adult pleomorphic sarcomas of the extremity: an analysis of 92 cases. Cancer

2003; 98 ;805-813.

96. Massi D, Beltrami G, Capanna R, Franchi A. Histopathological re-classification of extremity pleomorphic soft tissue sarcoma has clinical relevance. Eur J Surg Oncol

2004; 30 ;1131-1136.

97. Kuhnen C, Herter P, Leuschner I et al. Sclerosing pseudovascular rhabdomyosarcoma-immunohistochemical, ultrastructural, and genetic findings indicating a distinct subtype of rhabdomyosarcoma. Virchows Arch 2006; 449 ;572-578.

32 Published on behalf of the British Division of the International Academy of Pathology Page 33 of 39 Histopathology

98. Oda Y, Miyajima K, Kawaguchi K et al. Pleomorphic leiomyosarcoma:

clinicopathologic and immunohistochemical study with special emphasis on its

distinction from ordinary leiomyosarcoma and malignant fibrous histiocytoma. Am J Surg

Pathol 2001; 25 ;1030-1038.

99. Binh MB, Sastre-Garau X, Guillou L et al. MDM2 and CDK4 immunostainings

are useful adjuncts in diagnosing well-differentiated and dedifferentiated liposarcoma subtypes: a comparativeFor analysis Peer of 559 soft tissueReview neoplasms with genetic data. Am J Surg Pathol 2005; 29 ;1340-1347.

100. Chung L, Lau SK, Jiang Z et al. Overlapping features between dedifferentiated

liposarcoma and undifferentiated high-grade pleomorphic sarcoma. Am J Surg Pathol

2009; 33 ;1594-1600.

101. Hasegawa T, Seki K, Hasegawa F et al. Dedifferentiated liposarcoma of

retroperitoneum and mesentery: varied growth patterns and histological grades - a

clinicopathologic study of 32 cases. Hum Pathol 2000; 31 ;717-727.

102. Coindre JM, Mariani O, Chibon F et al. Most malignant fibrous histiocytomas

developed in the retroperitoneum are dedifferentiated liposarcomas: a review of 25 cases

initially diagnosed as malignant fibrous histiocytoma. Mod Pathol 2003; 16 ;256-262.

103. Coindre JM, Hostein I, Maire G et al. Inflammatory malignant fibrous

histiocytomas and dedifferentiated liposarcomas: histological review, genomic profile,

and MDM2 and CDK4 status favour a single entity. J Pathol 2004; 203 ;822-830.

104. Lawson CW, Fisher C, Gatter KC. An immunohistochemical study of

differentiation in malignant fibrous histiocytoma. Histopathology 1987; 11 ;375-383.

33 Published on behalf of the British Division of the International Academy of Pathology Histopathology Page 34 of 39

105. Weiss SW BG, Morris PA. Postirradiation malignant fibrous histiocytoma expressing cytokeratin. Implications for the immunodiagnosis of sarcomas. Am J Surg

Pathol 1988; 12 ;554-558.

106. Verrill MW, Judson IR, Harmer CL, Fisher C, Thomas JM, Wiltshaw E. Ewing's sarcoma and primitive neuroectodermal tumor in adults: are they different from Ewing's sarcoma and primitive neuroectodermal tumor in children? J Clin Oncol 1997; 15 ;2611- 2621. For Peer Review 107. Lessnick SL, Dei Tos AP, Sorensen PH et al. Small round cell sarcomas. Semin

Oncol 2009; 36 ;338-346.

108. Folpe AL, Hill CE, Parham DM, O'Shea PA, Weiss SW. Immunohistochemical detection of FLI-1 protein expression: a study of 132 round cell tumors with emphasis on

CD99-positive mimics of Ewing's sarcoma/primitive neuroectodermal tumor. Am J Surg

Pathol 2000; 24 ;1657-1662.

109. Olsen SH, Thomas DG, Lucas DR. Cluster analysis of immunohistochemical profiles in synovial sarcoma, malignant peripheral nerve sheath tumor, and Ewing sarcoma. Mod Pathol 2006; 19 ;659-668.

110. Folpe AL, Goldblum JR, Rubin BP et al. Morphologic and immunophenotypic diversity in Ewing family tumors: a study of 66 genetically confirmed cases. Am J Surg

Pathol 2005; 29 ;1025-1033.

111. Llombart-Bosch A, Machado I, Navarro S et al. Histological heterogeneity of

Ewing's sarcoma/PNET: an immunohistochemical analysis of 415 genetically confirmed cases with clinical support. Virchows Arch 2009; 455 ;397-411.

34 Published on behalf of the British Division of the International Academy of Pathology Page 35 of 39 Histopathology

112. Ordonez NG. Desmoplastic small round cell tumor: II: an ultrastructural and

immunohistochemical study with emphasis on new immunohistochemical markers [In

Process Citation]. Am J Surg Pathol 1998; 22 ;1314-1327.

113. Lae ME, Roche PC, Jin L, Lloyd RV, Nascimento AG. Desmoplastic small round

cell tumor: a clinicopathologic, immunohistochemical, and molecular study of 32 tumors.

Am J Surg Pathol 2002; 26 ;823-835. 114. Trupiano JK,For Machen SK,Peer Barr FG, GoldblumReview JR. Cytokeratin-negative desmoplastic small round cell tumor: a report of two cases emphasizing the utility of

reverse transcriptase-polymerase chain reaction. Mod Pathol 1999; 12 ;849-853.

115. Ordi J, de Alava E, Torne A et al. Intraabdominal desmoplastic small round cell

tumor with EWS/ERG fusion transcript. Am J Surg Pathol 1998; 22 ;1026-1032.

116. Sebire NJ, Gibson S, Rampling D, Williams S, Malone M, Ramsay AD.

Immunohistochemical findings in embryonal small round cell tumors with molecular

diagnostic confirmation. Appl Immunohistochem Mol Morphol 2005; 13 ;1-5.

117. Folpe AL. MyoD1 and myogenin expression in human neoplasia: a review and

update. Adv Anat Pathol 2002; 9;198-203.

118. Fisher C. Soft tissue sarcomas with non-EWS translocations: molecular genetic

features and pathologic and clinical correlations. Virchows Arch 2010; 456 ;153-166.

119. Sullivan LM, Atkins KA, LeGallo RD. PAX immunoreactivity identifies alveolar

rhabdomyosarcoma. Am J Surg Pathol 2009; 33 ;775-780.

120. Wachowiak R, Rawnaq T, Metzger R et al. Universal expression of

NCAM in neuroblastoma in contrast to : implications for different roles in

tumor biology of neuroblastoma? Pediatr Surg Int 2008; 24 ;1361-1364.

35 Published on behalf of the British Division of the International Academy of Pathology Histopathology Page 36 of 39

121. Hasegawa T, Hirose T, Ayala AG et al. Adult neuroblastoma of the retroperitoneum and abdomen: clinicopathologic distinction from primitive neuroectodermal tumor. Am J Surg Pathol 2001; 25 ;918-924.

122. Chibon F, Mariani O, Derre J et al. A subgroup of malignant fibrous histiocytomas is associated with genetic changes similar to those of well-differentiated liposarcomas. Cancer Genet Cytogenet 2002; 139 ;24-29. 123. Oikawa K, ForIshida T, Imamura Peer T et al. ReviewGeneration of the novel monoclonal antibody against TLS/EWS-CHOP chimeric oncoproteins that is applicable to one of the most sensitive assays for myxoid and round cell liposarcomas. Am J Surg Pathol

2006; 30 ;351-356.

36 Published on behalf of the British Division of the International Academy of Pathology Page 37 of 39 Histopathology

Table 1. CD34 and other markers in selected spindle and epithelioid soft tissue tumours

CD34 CK SMA S100pr Desmin Other diagnostic markers

Dermatofibrosarcoma + - - + (Bednar) − For Peer Review Ectopic hamartomatous thymoma + + − − −

Epithelioid angiosarcoma + + − − − CD31, INI1

Epithelioid sarcoma + + ± − − INI1 negative

Gastrointestinal stromal tumour + − + ± CD117, DOG1, h-caldesmon

Mammary type myofibroblastoma + − ± − +

Neurofibroma + − − + −

Perineurioma + − − − − EMA, claudin-1

Peripheral nerve sheath tumour, malignant + ± − + focal + (Triton)

Schwannoma + ± − + diffuse − GFAP

Solitary fibrous tumour + − − − − Bcl-2, CD99, CD56

Epithelioid malignant peripheral nerve sheath tumor − − − + − INI1 (50% negative)

Epithelioid sarcoma-like haemangioendothelioma − + − − − CD31+, FLI1+

Fibromatosis − − + ± ± Beta-catenin

Published on behalf of the British Division of the International Academy of Pathology Histopathology Page 38 of 39

Inflammatory myofibroblastic tumour − ± + − − ALK (50%)

Myofibrosarcoma − − + − ± Calponin, h-caldesmon negative

Retroperitoneal fibrosis − − + − − IgG4/IgG ratio

Smooth muscle tumours − ± + + h-caldesmon, SMM For Peer Review Spindle cell carcinoma − + + − − EMA, CK5/6, CK34betaE12

Synovial sarcoma − + − ± − TLE1, CD99, CD56

± = in some cases, CK = cytokeratin. EMA = epithelial membrane antigen, GFAP = glial fibrillary acidic protein, SMM = smooth muscle myosin

Published on behalf of the British Division of the International Academy of Pathology Page 39 of 39 Histopathology

Table 2. Differential diagnosis of soft tissue tumours with clear cells

S100 protein HMB45 Melan-A SMA Desmin TFE3 Other diagnostic findings

Clear cell sarcoma (soft tissue) + (diffuse) + + - - - t(12;22)(q13;q12), EWSR1-ATF1

Clear cell sarcoma (GIT) +For (diffuse) Peer - Review - - - - t (2;22)(q33;q12), EWSR1-CREB1

PEComa ± (focal) + + + ± ±

Paraganglioma - (except sustentacular - - - - - CD56+, NF+, CG+ cells)

Alveolar soft part sarcoma - - - - ± + t(X;17)(p11;q25) , ASPL-TFE3

± = in some cases , GIT = gastrointestinal tract, SMA = smooth muscle actin, NF = neurofilament, CG = chromogranin,

Published on behalf of the British Division of the International Academy of Pathology