You are on page 1of 13

Small Round Cell Tumors of Bone

Meera Hameed, MD

● Context.—Primary small round cell tumors of the bone Data Sources.—A literature search using PubMed and
are a heterogeneous group of malignant neoplasms pre- Ovid MEDLINE was performed, and data were obtained
senting predominantly in children and adolescents. They from various articles pertaining to clinicopathologic, bio-
include Ewing sarcoma/peripheral neuroectodermal tumor logical, and genetic findings in these tumors. Additionally,
or Ewing family tumors, lymphoma, mesenchymal chon- findings from rare cases have been included from author’s
drosarcoma, and small cell osteosarcoma. Even though subspecialty experience.
they share many morphological similarities, their unique Conclusion.—The diagnosis of small round cell tumors
biological and genetic characteristics have provided sub- can be made accurately by applying clinicopathologic cri-
stantial insights into the pathology of these diverse neo- teria, as well as a panel of immunohistochemical and ge-
plasms. netic studies in appropriate cases. Molecular genetic stud-
Objective.—To provide an overview of the clinical, ra- ies may provide further insight into the biology, histogen-
diologic, pathologic, and genetic characteristics of these esis, and prognosis of these tumors.
tumors along with a pertinent review of the literature. (Arch Pathol Lab Med. 2007;131:192–204)

EWING SARCOMA FAMILY OF TUMORS bones, the femur is most commonly affected, followed by
Since the original descriptions of tumors of the Ewing tibia, fibula, and humerus. However, virtually any bone
sarcoma family in 1918 by Arthur Purdy Stout1 and in can be affected, including the acral skeleton, craniofacial
1921 by James Ewing, who called the tumor diffuse endo- bones, and clavicle.13 Neoplasms in the long bones are of-
thelioma of bone,2 several nomenclatures have been assigned ten diaphyseal.14,15
to these tumors, albeit with much skepticism in the liter- Clinical Presentation
ature3 A possible relationship between Ewing sarcoma Local tenderness or mass is the most common clinical
(ES) and peripheral neuroectodermal tumor (PNET) was presentation. In about 10% to 15% of the cases, there is a
established after the description of extraosseous ES by An- pathological fracture when the femur is involved.16 It is
gerwall and Enzinger4 and PNET of the bone by Jaffe.5 At not uncommon to encounter children with systemic symp-
the present time, ES and PNET, otherwise called Ewing toms such as fever, fatigue, leukocytosis, and raised eryth-
family tumors (EFTs), form a single neoplastic entity rocyte sedimentation rate mimicking signs and symptoms
sharing common histologic and molecular features, and of acute osteomyelitis.15 This is an important clinical and
differing only in their extent of cellular differentiation. radiologic differential diagnosis.
Askin’s tumor of the thoracopulmonary region is also
currently grouped under EFT. Nonrandom translocation Radiographic Features
involving the EWS gene with one of the ETS family of Radiographic correlation is a prerequisite to all ortho-
transcription factors is the hallmark of their oncogene- pedic pathology and more so in tumors such as EFT,
sis.6–8 where clinical findings may point to benign processes
Incidence and Location such as osteomyelitis. The radiographic features are that
of a diaphyseal or metadiaphyseal lesion in the long
Ewing family tumor commonly occurs in the adolescent bones. Typically, the lesion is intramedullary (symmetric
age group between 10 and 20 years, but is extremely rare or eccentric) and is associated with cortical thickening and
in African American children.9 The annual incidence is cyclical periosteal reaction, giving rise to the characteristic
about 3 per million, and there is a slight male predomi- onion-skin appearance (Figure 1). The rapid growth of the
nance.10 These neoplasms constitute the second most com- tumor produces a continuous lifting of the periosteum
mon pediatric malignancy occurring in the skeletal sys- leading to perpendicular striations. Within the marrow,
tem.10 Adult patients (older than 40 years) with EFT are the lesion is poorly marginated, with a permeative pattern
rare, but isolated cases in patients up to 81 years old have of osteolysis (without a beginning and an end) (Figure 1).
been reported.11,12 Magnetic resonance imaging reveals an accompanying
Ewing family tumor involves both the axial and appen- soft tissue mass in about 90% of cases (Figures 2 and 3).
dicular skeleton, including the pelvis. Among long tubular Computed tomographic scan will demonstrate that the le-
sion itself does not produce any matrix (bone or cartilage).
Accepted for publication April 19, 2006. Occasionally there is prominent diffuse thickening of the
From the University of Medicine and Dentistry of New Jersey–New bone and cloudy opacities mimicking osteosarcoma (OS).
Jersey Medical School, Newark. Radiologic differential diagnoses include osteomyelitis,
The author has no relevant financial interest in the products or com-
panies described in this article.
Langerhans histiocytosis, lymphoma, and OS.17 Imaging
Reprints: Meera Hameed, MD, Surgical Pathology, UMDNJ–New Jer- workup of these patients involves skeletal scintigraphy
sey Medical School, Newark, NJ 07103 (e-mail: hameedmr@ (bone scan) and computed tomography of the chest to rule
umdnj.edu). out metastatic disease at the time of presentation.
192 Arch Pathol Lab Med—Vol 131, February 2007 Small Round Cell Tumors of Bone—Hameed
Figure 1. Plain x-ray. Diaphyseal intramedullary lesion of radius showing cyclical periosteal reaction with periosteal elevation and onion skinning.
Figure 2. Coronal magnetic resonance imaging without contrast showing the lesion occupies the entire marrow cavity encircling the entire cortex
with soft tissue extension.

Pathology abundant glycogen, which can be confirmed with periodic


An open biopsy is usually performed to obtain adequate acid–Schiff stain. Necrosis and apoptosis are common. The
material for pathological examination and ancillary stud- presence of apoptotic cells sometimes gives the biphasic
ies. Multiple core biopsies may be performed in patients appearance of dark and light cells (Figure 4). Homer-
with large, accessible soft tissue component of the tumors, Wright rosettes and pseudorosettes can be present (Figure
keeping in mind that additional studies such as cytoge- 5). Stromal elements are minimal, and the tumor generally
netics will be needed. Fine-needle aspiration can be used fills the medullary cavity and replaces bone marrow ele-
in an appropriate setting where adequate material can be ments. Interspersed collagen bundles are usually seen in
obtained. Grossly, the biopsied material is often hemor- the soft tissue component. A vascular component is not
rhagic and necrotic and usually has a tan-gray appear- prominent and is usually composed of slitlike capillaries.
ance. Touch imprints of the submitted pieces allow one Thick-walled vessels are seen in areas where there are
not only to examine morphology, but also to select viable prominent stroma and collagen bundles. A minimal de-
material for cytogenetic analysis. Molecular studies (re- gree of spindle cell change can be present. The tumor cells
verse transcriptase polymerase chain reaction [RT-PCR] permeate the cortex and elicit periosteal new bone for-
and fluorescence in situ hybridization [FISH]) can be done mation with reactive osteoblasts, osteoclasts, and some-
using frozen and paraffin-embedded tissue. If tissue is times cartilaginous metaplasia. The tumor generally does
snap-frozen for molecular studies, care should be taken to not induce an inflammatory response.
freeze tumor material in a timely manner to avoid RNA
degradation. Whenever possible, bony spicules should be Morphological Variants
separated from soft tissue to avoid unnecessary decalci-
fication of the soft tissue component. A number of variants have been described in EFT. These
Microscopic examination of classic EFT shows a highly include atypical ES (large cell EFT),18 adamantinoma-like
cellular neoplasm consisting of a monotonous population ES,19,20 sclerosing ES, and spindle cell sarcoma–like EFT.21
of uniform round blue cells (diameter, about 14 ␮m) ar- The tumor cells of the atypical or large cell EFT are het-
ranged in a nested, sheetlike, or solid pattern. The nuclei erogeneous compared with classic EFT. Their size ranges
are round with fine chromatin, scant clear or eosinophilic between 20 and 24 ␮m, and they have irregular nuclear
cytoplasm, indistinct cytoplasmic membranes, and 1 to 3 membranes and prominent nucleoli (Figure 6). The cyto-
small to medium-sized nucleoli. The cytoplasm contains plasm is vacuolated, and cytoplasmic glycogen is less
Arch Pathol Lab Med—Vol 131, February 2007 Small Round Cell Tumors of Bone—Hameed 193
Figure 3. Axial T2-weighted magnetic resonance imaging without contrast showing the lesion occupies the entire marrow cavity encircling the
entire cortex with soft tissue extension.
Figure 4. Photomicrograph of Ewing sarcoma showing diffuse monotonous round cells with dark and light pattern (hematoxylin-eosin, original
magnification ⫻400)
Figure 5. Rosettelike pattern in Ewing sarcoma (hematoxylin-eosin, original magnification ⫻400).
Figure 6. Photomicrograph of large cell Ewing family tumor with irregular nuclear contours and prominent nucleoli (hematoxylin-eosin, original
magnification ⫻400).
Figure 7. Pelvic atypical Ewing sarcoma showing nests of tumor cells in a desmoplastic stroma (hematoxylin-eosin, original magnification ⫻200).
Figure 8. Interphase fluorescence in situ hybridization section showing split signal with EWS probes (yellow signal normal and green and red
signal representing split EWS).

abundant than in conventional EFT. In some cases, the (Figures 7 and 8). RT-PCR for EWS-FLI-1, ERG, and WT1
large cells can be accompanied by spindled tumor cells. fusion transcripts were negative in this tumor. Sclerosing
Adamantinoma-like EFT has been described in the tibia EFT resembles sclerosing epithelioid fibrosarcoma or scle-
and fibula and shares histological and immunohistochem- rosing rhabdomyosarcoma because of the presence of hy-
ical features of adamantinoma, such as distinct nests of alinized eosinophilic matrix. However, there are always at
moderately pleomorphic tumor cells with peripheral pal- least focal areas showing typical features of classic EFT.21
isading, a prominent desmoplastic host response, and pos- Spindle cell sarcoma–like EFT shows a greater degree of
itive immunohistochemistry for cytokeratins. These tu- spindling than classic EFT and thus can resemble synovial
mors are also positive for CD99 and FLI-1 and show EWS- sarcoma.21
FLI-1 fusion transcript.19 We have seen a tumor of the pel-
vis with similar histology which showed positive staining Ultrastructure
for pan-cytokeratin with a variant EWS translocation, as Transmission electron microscopy of EFTs demonstrates
evidenced by positive break-apart FISH with EWS probe densely packed undifferentiated tumor cells with cyto-

Table 1. Immunohistochemical Profile*


Tumor CD99 FLI-1 LCA B Cell T Cell TdT CK Chr S100
EFT ⫹ ⫹ ⫺ ⫺ ⫺ ⫺ ⫹/⫺ ⫺ ⫹/⫺
LBL/ALL ⫹ ⫹ ⫹/⫺ ⫹/⫺ ⫹/⫺ ⫹ ⫹/⫺ ⫺ ⫺
NHL–other ⫹/⫺ ⫹ ⫹ ⫹ ⫹ ⫺ ⫺ ⫺ ⫺
Mesenchymal chondrosarcoma ⫹ ⫺ ⫺ ⫺ ⫺ ⫺ ⫺ ⫺ ⫹
Small cell OS ⫹/⫺ ? ⫺ ⫺ ⫺ ⫺ ⫹/⫺ ⫺ ⫹/⫺
Rhabdomyosarcoma ⫹/⫺ ⫺ ⫺ ⫺ ⫺ ⫺ ⫺ ⫺ ⫹/⫺
DSCRT ⫹ ⫹/⫺ ⫺ ⫺ ⫺ ⫺ ⫹ ⫹/⫺ ⫹/⫺
Neuroblastoma ⫺ ⫺ ⫺ ⫺ ⫺ ⫺ ⫺ ⫹ ⫹/⫺
* LCA indicates leukocyte common antigen; TdT, terminal deoxynucleotidyltransferase; CK, pan-cytokeratin; Chr, chromogranin; EFT, Ewing
family tumors; LBL/ALL, lymphoblastic lymphoma/acute lymphoblastic leukemia; NHL, non-Hodgkin lymphoma; OS, osteosarcoma; DSCRT, des-
moplastic small round cell tumor; and ?, not known.

194 Arch Pathol Lab Med—Vol 131, February 2007 Small Round Cell Tumors of Bone—Hameed
plasmic glycogen and scarce organelles. Occasionally, des-
mosome-like junctions are seen. Evidence of neural differ-
entiation such as dense core neurosecretory granules, den-
dritic processes, and microtubules can be present.22,23
Immunohistochemistry
It is advisable to use a panel of immunohistochemical
markers in small round cell tumors of the bone which in-
clude vimentin, CD99, FLI-1, leucocyte common antigen,
terminal deoxynucleotidyl transferase (TdT), B-cell, T-cell
markers, pan-cytokeratin, desmin, MYOD1/myogenin,
chromogranin, and S100 protein. These markers assist in
differentiating EFT from other tumors such as lympho-
blastic lymphoma/acute lymphoblastic leukemia, rhab-
domyosarcoma, desmoplastic small round cell tumor, and
neuroblastoma. Other differential diagnoses to consider
would be mesenchymal chondrosarcoma, non-Hodgkin
lymphoma (NHL) (other), and small cell OS. The most
useful, though nonspecific, marker in the diagnosis of EFT
is CD99, which often produces a diffuse membranous
staining pattern in these tumors (Figure 9). This product
is a 32-kd transmembrane protein whose function is not
well established; it is encoded by the MIC2 gene located
in the pseudoautosomal region of X and Y chromo-
somes.24–26 Apart from EFT, this protein is also highly ex-
pressed in T and B lymphocytes, hence also expressed by
lymphoblastic lymphoma/acute lymphoblastic leukemia.
In addition, positive staining has been observed in some
rhabdomyosarcomas, desmoplastic small round cell tu-
mors, synovial sarcomas, and mesenchymal chondrosar-
comas.27–30 Another useful immunomarker is the FLI-1
protein, which is expressed in about 84% of EFTs (Figure
10).31 This transcription factor is also expressed by lym-
phoblastic lymphomas and other NHLs.31 Other variably
expressed markers in EFT include vimentin, cytokeratin
(up to 25%), desmin (very rare), and neural markers such Figure 9. Immunohistochemical stain for CD99 showing diffuse pos-
as synaptophysin, CD57, S100, and chromogranin.21,23,32–34 itivity in typical Ewing sarcoma (original magnification ⫻400).
The presence of neural markers in an EFT indicates neu-
Figure 10. Immunohistochemical staining for FLI-1 showing intense
roectodermal differentiation (PNET). High molecular cy- nuclear staining in tumor cells.
tokeratin (34␤E12) is positive in adamantinoma-like ES
and not in classic and other variants of EFT.21 Table 1 sum-
marizes the immunohistochemical profile of these tumors. some 22q12 to the FLI-1 gene at chromosome 11q24. The
EWS-FLI-1 chimeric protein on the (der)22, contains the 5’
Cytogenetics and Molecular Genetics end of the EWS gene and the 3’ end of the FLI-1 gene.38
The pathogenetic relationship of ES and PNET was de- The second most common translocation in EFT is t(12;
fined by the discovery of the unique and specific nonran- 22)(q22;q12), seen in about 5% to 10% of the cases, re-
dom chromosomal translocation, seen in about 85% to 95% sulting in the fusion of EWS to ERG at chromosome
of the tumors, involving chromosome 11q24 and chro- 12q22.39 Other variant translocations reported in bone EFT
mosome 22q12, t(11;22)(q24;q12) (Figure 11).35–37 This accounting for less than 1% of cases include t(7;22)(p22;
leads to an in-frame fusion of the EWS gene at chromo- q12), t(17;22)(q12;q12), and t(2;22)(q23;q12), involving fu-
sion of EWS to ETV1, E1AF, and FEV genes, respectively
(Table 2).40–43
The EWS gene has 17 exons, of which the first 7 exons
Table 1. Extended
encode the N-terminal region responsible for regulating
Desmin Myogenin/MYOD1 WT1 its RNA binding capacity. It belongs to the TET family of
⫺ ⫺ ⫺ RNA binding proteins (EWS, TLS, TAFII68) whose amino
⫺ ⫺ ⫹/⫺ terminal (NTD) has the capacity to bind to DNA-binding
⫺ ⫺ ⫺ domains of various transcription factors, thus providing a
⫹/⫺ ⫹/⫺ ⫺ strong transcriptional activating domain to the resulting
⫹/⫺ ? ?
⫹ ⫹ ⫹/⫺(cyto)
chimeric protein.44,45 EWS itself is ubiquitously expressed
⫹ ⫺ ⫹ in all tissues.46 The partner genes of EWS in the EFT of
⫺ ⫺ ⫺ bone, such as FLI-1, ERG, ETV1, EIAF, and FEV, belong to
the ETS family of transcription factors possessing a con-
served ETS domain recognizing a core DNA motif of
GGAA/T.47 FLI-1 expression is seen in high levels in he-
Arch Pathol Lab Med—Vol 131, February 2007 Small Round Cell Tumors of Bone—Hameed 195
Figure 11. Karyotyping of Ewing sarcoma with t(11;22)(q24;q12).
Figure 12. EWS-FLI-1 gene fusion diagram showing common fusion products with long arrows indicating the most common break points. All
fusions include exons 1 through 7 of the EWS gene (NTD) and FLI-1 exon 9 (DNA binding domain). The involved exons are shown as numbers
for each transcript of EWS-FLI-1.
Figure 13. Reverse transcriptase polymerase chain reaction showing 329 bp type I fusion product of EWS-FLI-1.
Figure 14. Metaphase fluorescence in situ hybridization with EWS dual-color break-apart probe showing translocation of green signal from
chromosome 22 to chromosome 11.

matopoietic cells, endothelial cells, and mesenchyme de- the EWS-ETS fusion are FISH and RT-PCR. Multiple chi-
rived from neural crest cells.48,49 The product of EWS-ETS meric transcripts are possible, all of which involve exons
translocation results in fusion proteins containing the C- 1 through 7 of EWS and exon 9 of the ETS gene. The
terminal region of the FLI-1 and N-terminal region of genomic break points are intronic, and occur at 1 of 4
EWS, thus bringing the ETS component under the pow- EWS introns and, in the case of EWS-FLI-1, 1 of 6 FLI-1
erful transactivating domain of EWS.50 These fusion prod- introns (Figure 12).51 Type 1 (exon 7 of EWS to exon 6 of
ucts modulate multiple target genes, resulting in malig- FLI-1) and type II (exon 7 of EWS to exon 5 of FLI-1) are
nant transformation. the most common (⬎85%) fusion transcripts.52 Commonly
The most commonly used molecular assays to identify used RT-PCR assays use EWS exon 7 forward primer and
FLI-1 exon 9 reverse primer, which amplifies all fusion
products, or EWS exon 7 forward primer and FLI-1 exon
Table 2. Translocation and Fusion Genes in Ewing 6 to detect type 1 or type II fusion products, which would
Family Tumors of Bone identify 85% of the cases (Figure 13). In addition, RT-PCR
Translocation Fusion Genes Frequency for EWS-ERG fusion product can be performed, which
t(11;22)(q24;q12) EWS-FLI-1 85%–95% would identify the 10% of cases with this fusion. A mul-
t(21;22)(q22;q12) EWS-ERG 5%–10% tiplex real-time PCR has also been used to identify EWS-
t(7;22)(p22;q12) EWS-ETV1 ⬍1% ETS fusion products.53 RT-PCR has also been applied to
t(17;22)(q12;q12) EWS-E1AF ⬍1% formalin-fixed, paraffin-embedded tissues, and a combi-
t(2;22)(q23;q12) EWS-FEV ⬍1% nation of FISH and RT-PCR may yield increased sensitiv-
196 Arch Pathol Lab Med—Vol 131, February 2007 Small Round Cell Tumors of Bone—Hameed
Figure 15. Gross photograph (A) and spec-
imen x-ray (B) of resected Ewing sarcoma of
radius after chemotherapy. The grid lines
serve as a map for histology sections.
Figure 16. Plain x-ray of proximal femur
showing a destructive moth-eaten lesion with
mixed lytic and sclerotic areas involving me-
taphysis and diaphysis. Periostitis is seen ad-
jacent to lateral cortex.

ity and accuracy due to limitations of formalin-fixed, par- ponse in resected specimens, which has some prognostic
affin-embedded tissue material with suboptimal preser- significance and has a role in selection of agents for con-
vation of nucleic acids.54 Fluorescence in situ hybridization tinuation chemotherapy. A sagittal section of the resected
assay with the commercially available (such as Vysis) bone, with mapping and examining the entire central face
dual-color break-apart DNA probes flanking the EWS of the tumor and adjacent tissue after careful decalcifica-
break point region on chromosome 22 detects the trans- tion, allows one to assess the chemotherapeutic response
location of EWS and can be used in fresh and formalin- (Figure 15).58 The grading system used to assess chemo-
fixed, paraffin-embedded tissues. In a metaphase spread, response is similar to that used in OS (Table 3).58,59
the split signal enables one to identify the chromosome
involved in the translocation; however, the partner gene is Prognosis and Outcome
not identified by this methodology (Figure 14). Up to one third of patients with EFT have metastatic
Additional reported secondary karyotypic changes in disease at the time of presentation, and their outcome re-
EFT include gains of chromosomes 8, 12, and 18, deletion mains poor in spite of aggressive chemotherapy.60 The
of the short arm of chromosome 1, unbalanced translo- most common sites of metastases are lung, bone, and bone
cation t(1;16)(q12;q11), and other rare structural abnor- marrow. Patients who present only with lung metastases
malities.55,56 have better survival compared with those who present
Treatment and Treatment Response with bone metastases.61 One third of the patients with lo-
calized disease are also prone to distant relapse, especially
The management of EFTs includes systemic chemother- if there are residual viable tumor cells at definitive resec-
apy followed by surgery, with or without radiation and tion.61,62 Multiple biological factors, such as type of fusion
continuation chemotherapy.57 Induction chemotherapy be- transcript, mutations in INK4a gene, mutations in p53, de-
fore definitive surgery allows us to assess the chemores- letion of p16, telomerase, IGF-1, and aneuploidy, have been
implicated to play a role in the prognosis of EFT.63,64 In
patients with localized disease, EWS-FLI-1 type 1 fusion
Table 3. Chemotherapy Response—Pediatric transcript has been reported as being associated with im-
Oncology Group Study
proved outcome compared to other fusion transcript
Grade Tumor Response 3-y Survival, % types, and it appears that type 1 fusion transcript encodes
I No chemotherapy effect 30 a less active chimeric protein.65,66 Reports on the detection
IIA 1%–10% necrosis 30 of minimal residual disease by RT-PCR for the chimeric
IIB 11%–90% necrosis 49 proteins in peripheral blood and bone marrow remain
III 91%–99% necrosis 73 controversial with respect to disease progression.67–70 Cur-
IV 100% necrosis 100
rent chemotherapeutic regimens with selective surgery
Arch Pathol Lab Med—Vol 131, February 2007 Small Round Cell Tumors of Bone—Hameed 197
have significantly improved the overall survival up to 60% tumor may evoke a sclerotic response and present as a
to 80% in nonmetastatic EFT.71 Radiation-induced sarco- blastic lesion, which can sometimes be mistaken for Paget
mas and chemotherapy-related acute leukemias are the disease. Computed tomography and magnetic resonance
most frequent secondary malignancies.13 imaging are useful in assessing extent of disease, and pos-
itron emission tomography and bone scans are used for
Conclusion staging and assessment of remission after therapy. Of im-
Even though the cell of origin is still unknown in EFT, portance is the fact that occasionally plain radiographs can
considerable strides have been made since the original de- be normal, and persistence of clinical symptoms would
scription by Drs Stout and Ewing. The combination of warrant further investigation such as magnetic resonance
morphology, immunohistochemistry, and molecular ge- imaging, which will demonstrate the signal abnormalities
netics has opened many doors, including some novel ap- of the marrow. Pathologic fracture can be present in 22%
proaches to therapeutics, such as the use of CD99 as a of cases.89 Due to the nonspecificity of the radiographic
target,72 inhibition of IGF-1 receptor with antisense oligo- findings, the differential diagnoses include OS, eosino-
nucleotides,73 and small interfering RNA (siRNA)74 all of philic granuloma, metastases of solid tumors, round cell
which have shown promising results in cell lines and an- tumors (ES, neuroblastoma, rhabdomyosrcaoma), and
imal models. chronic osteomyelitis.
NON-HODGKIN LYMPHOMA Pathology
Primary NHL of bone was described as a series, first by
Grossly, bone lymphomas tend to be fleshy gray-white
Jackson and Parker in 1939.75 Before this, Oberling in 1928
or hemorrhagic, and the gross appearance is also related
had reported a case of reticuloendothelial sarcoma histo-
to the response of the host bone, leading to sometimes
logically indistinguishable from ES.76 The description of
very sclerotic and bony biopsy specimens requiring de-
this rare entity is restricted to lymphoma involving a sin-
calcification. Histologically, the most common finding is a
gle skeletal site with or without regional lymph node in-
diffuse pattern of growth with tumor cells permeating be-
volvement or multiple skeletal sites without visceral or
tween bony trabeculae and invading Haversian channels
lymph node involvement.77
of the cortex. There is often a sclerotic or collagenized
Incidence and Location stroma, and the tumor cells can invade in cords, nests, or
solid sheets. As is often evidenced in the x-ray, the host
Approximately 3% to 7% of extranodal lymphomas pre-
bony trabeculae can be thickened and irregular with ex-
sent as primary bone neoplasms,78–81 and lymphomas con-
tensive sclerosis. The most frequent type (92%)82 of pri-
stitute about 7% of all bone malignancies.77 The disease
mary NHL of bone is diffuse large B-cell lymphoma
tends to involve older adults (older than 40 years), and
(DLBCL). Unlike nodal lymphomas, DLBCL of bone char-
there is a male predominance, with male-female ratio
acteristically shows a polymorphous appearance (Figures
ranging from 1.2:1 to 1.6:1.82 The femur, pelvis, vertebra,
18 and 19). The tumor cells themselves show marked var-
and humerus are the most common sites. In the long
iation in size and shape, often with multilobation, and are
bones, the preferred site is the metadiaphyseal region.83,84
accompanied by a mixture of small and medium-sized
Involvement of the small bones of the hand and feet is
lymphocytes that are reactive B and T cells. The large tu-
extremely unusual. It is not uncommon to encounter mul-
mor cells can have prominent nucleoli, and the cytoplasm
tifocality, and soft tissue extension may be present.77,83
is not usually abundant. Occasionally, the fibrotic stromal
Clinical Presentation reaction elicited by the tumor cells can result in tumor cell
spindling with storiform features, mimicking a sarcoma.
Most patients present with pain, and depending on lo-
The polymorphous nature of the infiltrate can lead to a
cation, such as spine, neurological symptoms may be pres-
mistaken diagnosis of osteomyelitis, which is also a radio-
ent. Systemic symptoms such as fever (as seen in ES) and
logic differential diagnosis. Crush artifact is fairly com-
B symptoms of nodal lymphomas are unusual.83 Occa-
mon due to the delicate nature of the tumor cells and can
sionally patients can present with hypercalcemia, lethargy,
be a vexing problem in pathologic diagnosis. Apart from
constipation, etc.85 For a neoplasm to be classified as a
DLBCL, other NHLs, which can occasionally present as
primary bone lymphoma, an interval of 4 to 6 months
primary neoplasms of bone, include anaplastic large cell
between the skeletal manifestation and extraskeletal dis-
lymphoma, lymphoblastic lymphoma, Burkitt lymphoma,
ease is required.86 Thus appropriate staging procedures,
and T-cell lymphomas.82,90,91 Of these, lymphoblastic lym-
such as skeletal imaging, computed tomographic scans,
phoma can be a diagnostic challenge due to morphological
bone scan/positron emission tomography, and bone mar-
and immunohistochemical overlap with ES.90 Follicular
row biopsy, are an integral part of the patient workup.87
lymphomas and small lymphocytic lymphomas such as
Radiographic Features chronic lymphocytic lymphoma generally do not present
as primary destructive bone lesions.77
On conventional x-rays, primary lymphomas are lytic
radiolucent lesions with a moth-eaten or permeative de-
Immunohistochemistry and Genetics
structive pattern. This may be associated with sclerotic ar-
eas (Figure 16).84,88 These changes are nonspecific and In a suspected case of NHL, a panel of markers is used
overlap with other round cell tumors. Cortical erosion and to delineate the lineage and type of the neoplasm. They
destruction with soft tissue extension is frequently seen include leucocyte common antigen, B-cell markers (CD20),
(Figure 17). Periosteal reaction (interrupted or solid single- and T-cell markers (CD2, CD3, CD5, CD7), and additional
layer) can be present, but less so than in ES. Generally the markers such as CD30, epithelial membrane antigen, and
disease tends to involve the long bones extensively, some- Alk-1 to rule out anaplastic large cell lymphoma. In a case
times occupying the entire length. In some instances, the with a monomorphic morphology such as lymphoblastic
198 Arch Pathol Lab Med—Vol 131, February 2007 Small Round Cell Tumors of Bone—Hameed
Figure 17. Computed tomographic scan showing sclerosis in medullary canal with anterior cortical destruction and soft tissue extension.
Figure 18. Photomicrograph of diffuse large B-cell lymphoma showing a diffuse polymorphous infiltrate composed of small, medium, and large
cells (hematoxylin-eosin, original magnification ⫻200).
Figure 19. Immunohistochemical stain showing diffuse positivity for CD20, B-cell marker (original magnification ⫻400).
Figure 20. Photomicrograph of atypical Burkitt lymphoma with large lymphoid cells with prominent nucleoli and mitotic figures (hematoxylin-
eosin, original magnification ⫻400).

lymphoma, the panel should include all of the lymphoma such as flow cytometry can be used in appropriate cases.
and other round cell tumor markers (CD45, CD20, CD2, B-cell and T-cell rearrangement studies can be performed
CD3, CD5, CD7, TdT, vimentin, CD99, FLI-1, keratin, des- in fresh and paraffin-embedded tissues to confirm clon-
min, chromogranin). In addition, other lymphoid antigens ality in doubtful cases. Specific cytogenetic studies of pri-
such as CD79a and CD43 may be required, as lympho- mary bone lymphomas are sparse. There is a single report
blastic lymphomas are often negative for CD20 and posi- of a pediatric DLBCL of the spine with t(3;22)(q27;q11).92
tive for CD99 and FLI-1. They can also be negative for Recently we encountered a case of an atypical Burkitt lym-
leucocyte common antigen, and in some instances show phoma arising in the calcaneus of an elderly male patient
focal cytoplasmic keratin positivity, a major pitfall that can histologically mimicking DLBCL (Figure 20); however, cy-
lead to an erroneous diagnosis of ES.90 In pediatric cases togenetic analysis showed t(8;14)(q24.1;q32), characteristic
of small round cell tumors, TdT appears to be a sensitive of a Burkitt lymphoma.
and specific marker for precursor B or T lymphoblastic
lymphomas and should be included in the immunopanel Treatment and Prognosis
(Table 1). Other special stains such as ␬ and ␭ can be used The primary modality of therapy for primary bone lym-
to ensure monoclonality, but the results are often variable phomas is chemotherapy and radiotherapy, depending on
due to technical limitations. Primary T-cell lymphomas of the stage of disease and the grade and type of the lym-
the bone are extremely rare, and the diagnosis is estab- phoma.93 The prognosis is dependent upon the stage of
lished based on morphology of the polymorphous popu- the disease. Patients with stages I and II have a very good
lation with large tumor cells and loss of pan–T-cell anti- prognosis compared with patients with stages III and IV.94
gens such as CD7. Two cases of primary adult T-cell leu- The 5-year overall survival ranges between 56% and
kemia of the bone have been reported.91 Additional studies 61%.82,95
Arch Pathol Lab Med—Vol 131, February 2007 Small Round Cell Tumors of Bone—Hameed 199
flicts young adults and teenagers, with a peak incidence
during the third decade.100 Males and females are equally
affected.98 Craniofacial bones, especially the jaw, are most
frequently affected.101 Other commonly involved sites in-
clude vertebrae, ribs, pelvis, and humerus. About one
third of these tumors are extraskeletal and arise in soft
tissues, and meninges are a favored extraskeletal location
for these tumors.98,99 Skeletal neoplasms can be multifo-
cal.99
Clinical Presentation
Pain and swelling are the most common clinical fea-
tures, usually several days to months in duration,102 and
oncogenic osteomalacia (tumor-induced osteomalacia) has
been reported.103
Radiographic Features
Plain x-ray reveals a radiolucent lesion, usually eccen-
tric with stippled calcifications denoting its chondroid na-
ture (Figure 21). The lesion is often destructive, with ex-
pansion of bone, cortical destruction, and extraosseous ex-
tension (Figure 22). Sometimes the calcifications can be
large and present as discrete opacities or heavily calcified
masses. Rarely, a sharply demarcated sclerotic margin is
present. Magnetic resonance imaging appearance can be
variable and can include a heterogeneous, low attenuation
on T1-weighted images (Figure 23) and high signal inten-
sity on T2-weighted images.104,105
Pathology
Biopsy of the lesion generally shows a grossly grey to
pink tissue with foci of mineralization. Microscopically,
the tumor shows a biphasic pattern made up of solid areas
of round or spindle mesenchymal cells interspersed with
islands of well-differentiated cartilage (Figure 24). The
mesenchymal component often exhibits a hemangioperi-
Figure 21. Plain x-ray of proximal femur showing a permeative pat-
cytic pattern with multiple vascular spaces. The cartilage
tern in the intertrochanteric and subtrochanteric region with endosteal component is variable and can show endochondral ossi-
scalloping across the distalmost aspect of the neoplasm. There are fication with bone formation, coarse calcifications, or
questionable foci of matrix calcification proximally. (Courtesy of Dr loosely arranged mature hyaline cartilage. There can be
Joseph Benevenia, Department of Orthopedics, University of Medicine distinct demarcation or gradual blending between the
and Dentistry of New Jersey, Newark.) mesenchymal component and the chondroid component
(Figure 25).98,99 The mesenchymal cells when they are
round can simulate ES or small cell OS in biopsy speci-
Conclusion mens. On the other hand, the cartilage component alone
Primary NHL of bone is a rare but well-defined entity in a biopsy can lead to the erroneous diagnosis of a benign
with radiologic and morphologic heterogeneity. The his- cartilaginous neoplasm.
tologic and immunohistochemical similarities, ranging
from benign conditions such as osteomyelitis to monoto- Ultrastructure
nous round cell sarcomas, should be borne in mind when The primitive mesenchymal cells show round to oval
assessing these neoplasms to avoid misdiagnosis and in- cells with little intercellular matrix, and the cartilaginous
appropriate treatment. foci reveal mature cells containing glycogen, rough en-
doplasmic reticulum, and numerous mitochondria.106
MESENCHYMAL CHONDROSARCOMA
This rare neoplasm was first described by Lichtenstein Immunohistochemistry
and Bernstein96 in 1959 as an unusual variant of chondro- The chondroid foci stain for S100 protein, and the prim-
sarcoma affecting bone and soft tissues. This was followed itive mesenchymal cells are positive for CD99 (Figure
by recognition of 9 more cases from the files of the Mayo 26).107 Both components are vimentin positive. It has been
Clinic by Dahlin and Henderson.97 Subsequently it has proposed that immunohistochemical positivity for colla-
been established as a separate entity distinct from conven- gen II and IIA, which are considered to be markers of
tional and dedifferentiated chondrosarcoma. chondroprogenitor cells, could be used to differentiate
mesenchymal chondrosarcoma from other small round
Incidence and Location cell malignancies, such as small cell OS and ES.108 Another
Mesenchymal chondrosarcomas constitute less than 2% study109 has shown that the transcription factor Sox9, a
of all chondrosarcomas.98,99 The tumor most commonly af- master regulator of chondrogenesis,110,111 differentiates
200 Arch Pathol Lab Med—Vol 131, February 2007 Small Round Cell Tumors of Bone—Hameed
Figure 22. Computed tomographic scan. Mesenchymal chondrosarcoma of the rib with cortical destruction and soft tissue mass with calcification.
(Courtesy of Dr John Reith, Department of Pathology and Orthopedics, University of Florida, Gainesville.)
Figure 23. T1-weighted magnetic resonance imaging of mesenchymal chondrosarcoma showing lobulated heterogeneous soft tissue mass of low
signal intensity. (Courtesy of Dr John Reith, Department of Pathology and Orthopedics, University of Florida, Gainesville.)

mesenchymal chondrosarcoma from various small cell SMALL CELL OSTEOSARCOMA


malignancies, including small cell OS, lymphoma, and ES, This tumor is discussed here in the context of the dif-
by immunohistochemical analysis using a rabbit polyclon- ferential diagnosis of the above-mentioned small round
al antibody. cell tumors of the bone. This is a distinctive microscopic
variant of OS, originally described in 1979, as a tumor
Cytogenetics and Molecular Genetics simulating ES.121 This rare tumor accounts for about 1%
There are a few reported cytogenetic alterations in mes- to 1.5% of OSs and has similar age, sex, and skeletal dis-
enchymal chondrosarcoma, and so far no specific or re- tribution as conventional OS.122 Radiologically the tumor
current translocations have been identified. Case reports can resemble ES or conventional OS. The majority of the
of near-tetraploid and other diverse structural abnormal- lesions show a mixed lytic and blastic pattern, but they
ities,112 translocation der(13;21)(q10;q10) with loss of chro- can be purely lytic or permeative; they usually involve
mosomes 8 and 20 material and gain of chromosome 12 metaphysis and may extend into epiphysis. A soft tissue
material in 2 cases,113 and 1 case with t(11;22)(q24;q12) component is present in most of the cases.123–126 Three his-
similar to ES have been reported.114 Molecular genetic tologic patterns have been described: ES-like, lymphoma-
analyses for mutational alteration of p53 and p16 tumor like, and small spindle cell.123 The cells can be round and
suppressor genes have shown a low incidence (⬍25%) in uniform, similar to ES, or show variation in size. Nucleoli
these tumors.115,116 Biologic pathways involving PKC-␣, can be inconspicuous to prominent. The chromatin can be
PDGFR-␣, and Bcl-2 expression seem to play a role in fine to hyperchromatic. The nuclei can be very small to
pathogenesis.117 medium (6.7–15 ␮m), thus showing a spectrum of sizes
between ES and large cell lymphoma.125 Hemangioperi-
Treatment and Prognosis cytic pattern, myxoid change, cordlike arrangement, and
epithelioid features can be seen. The presence of malig-
Mesenchymal chondrosarcomas are primarily treated nant cartilage in some tumors can simulate mesenchymal
with surgery. They are aggressive lesions with a high pro- chondrosarcoma.125 The presence of osteiod is a prereq-
pensity for metastases; however, some patients can have a uisite for diagnosis. The new osteoid matrix is usually
protracted clinical course, so the behavior can be unpre- lacelike and can be difficult to find in small biopsies (Fig-
dictable.118 The overall 10-year survival rate is approxi- ure 27). The presence of mineralized matrix in imaging
mately 25%.99 Tumors of the jawbones tend to have a more studies supports the diagnosis of OS and is helpful in
indolent course.119 Negative correlation between survival differentiating other small blue cell tumors of bone. ES
times and proliferation rates using Ki-67 in 10 patients has may show a blastic component in the medullary cavity,
been recently reported.120 Distant metastases which can but production of matrix outside the bone is seen only in
occur after many years can involve other bones, lung, OS.123,127,128 Cytoplasmic glycogen can be seen in both neo-
etc.99,118 Hence long-term follow-up of these patients is ad- plasms.123 Immunohistochemical features do not distin-
vocated. guish small cell OS from sarcomas, as CD99 can be pos-
Arch Pathol Lab Med—Vol 131, February 2007 Small Round Cell Tumors of Bone—Hameed 201
Figure 24. Microscopic photograph of mesenchymal chondrosarcoma showing a biphasic pattern of chondroid and round and spindle cells
(hematoxylin-eosin, original magnification ⫻200).
Figure 25. High-power view showing malignant cartilage and primitive round cell components (hematoxylin-eosin, original magnification ⫻400).
Figure 26. Immunohistochemical stain for CD99 showing positivity in tumor cells (original magnification ⫻400).
Figure 27. Photomicrograph of small cell osteosarcoma showing small spindle cells with a diffuse growth pattern (hematoxylin-eosin, original
magnification ⫻400).

itive in small cell OS.129 Ewing family tumor–specific SUMMARY


translocations such as t(11;22) have not been reported in Small round cell tumors of bone are a heterogeneous
small cell OS. Lymphomas can be distinguished using group of neoplasms with overlapping clinical, radiologic,
lymphoma-specific markers.130 The treatment of small cell morphologic, and immunohistochemical features. Even
OS is similar to conventional OS, which includes neoad- though questions remain regarding cell of origin and his-
juvant chemotherapy followed by surgery, usually a limb togenesis, cytogenetic and molecular genetic studies have
salvage procedure. According to some authors, when there enhanced our understanding in bringing together some of
is questionable matrix formation in a small round cell tu- these tumors into single entities. In addition, cDNA mi-
mor, it is best to err on the diagnosis of ES rather than croarray analysis and the availability of techniques to un-
small cell OS.125 ravel the various signaling pathways will open many
OTHER TUMORS doors toward targeted chemotherapy in these aggressive,
highly lethal tumors.
Rhabdomyosarcoma and desmoplastic round cell tu-
The author thanks Seena Aisner, MD, Department of Pathology,
mors are primarily soft tissue neoplasms; rare primary UMDNJ-Newark, for critical review of the manuscript.
bone tumors are cited in the literature as case reports.131,132
References
Neuroblastomas can present as metastases in the bone and 1. Stout AP. A tumor of the ulnar nerve. Proc N Y Pathol Soc. 1918;12:2–12.
can be distinguished from other primary round cell tu- 2. Ewing J. Diffuse endothelioma of bone. Proc N Y Pathol Soc. 1921;21:17–
mors of bone with immunohistochemical positivity for 24.
neuron-specific enolase, synaptophysin, and chromogran- 3. Cohn I. Epithelial neoplasms of peripheral and cranial nerves. Report of
three cases: review of the literature. Arch Surg. 1928;17:117–160.
in and absent staining for CD99,133 lymphoid markers, 4. Angerwall L, Enzinger FM. Extraskeletal neoplasm resembling Ewing’s sar-
desmin, myogenin, and MyoD1. coma. Cancer. 1975;36:240–251.

202 Arch Pathol Lab Med—Vol 131, February 2007 Small Round Cell Tumors of Bone—Hameed
5. Jaffe R, Santamaria M, Yunis EJ, et al. The neuroectodermal tumor of bone. 37. Whang-Peng J, Triche TJ, Knutsen T, Miser J, Douglass EC, Israel MA. Chro-
Am J Surg Pathol. 1990;8:885–898. mosome translocation in peripheral neuroepithelioma. N Engl J Med. 1984;311:
6. Ushigome U, Machinami R, Sorensen PH. Ewing sarcoma/primitive neu- 584–585.
roectodermal tumor (PNET). In: Fletcher CDM, Unni KK, Mertens F, eds. Pathol- 38. May WA, Gishizky ML, Lessnik SL, et al. Ewing sarcoma 11;22 translo-
ogy and Genetics of Tumours of Soft Tissue and Bone. Lyon, France: IARC Press; cation produces a chimeric transcription factor that requires the DNA-binding
2002:298–300. World Health Organization Classification of Tumours. domain encoded by FLI1 for transformation. Proc Natl Acad Sci U S A. 1993;90:
7. Turc-Carel C, Philip I, Berger MP, Phili T, Lenoir GM. Chromosomal trans- 5752–5756.
locations in Ewing’s sarcoma. N Engl J Med. 1983;309:497–498. 39. Sorensen PH, Lessnick SL, Lopez-Terrada D, et al. A second Ewing’s sar-
8. Whang-Peng J, Triche TJ, Knutsen T, Miser J, Douglass EC, Israel MA. Chro- coma translocation, t(21;22), fuses the EWS gene to another ETS-family transcrip-
mosome translocation in peripheral neuroepithelioma. N Engl J Med. 1984;311: tion factor, ERG. Nat Genet. 1994;6:146–151.
584–585. 40. Jeon IS, Davis JN, Braun BS, et al. A variant Ewing’s sarcoma translocation
9. Gurney JG, Severson RK, Davis S, Robison LL. Incidence of cancer in chil- (7;22) fuses the EWS gene to the ETS gene ETV1. Oncogene. 1995;10:1229–
dren in the United States: sex-, race-, and 1-year age-specific rates by histologic 1234.
type. Cancer. 1995;75:2186–2195. 41. Kaneko Y, Yoshida K, Handa M, et al. Fusion of an ETS-family gene, E1AF,
10. Carvajal R, Meyers P. Ewing’s sarcoma and primitive neuroectodermal fam- to EWS by t(7;22)(q12;q12) chromosome translocation in an undifferentiated sar-
ily of tumors. Hematol Oncol Clin North Am. 2005;19:501–525. coma of infancy. Genes Chromosomes Cancer. 1996;15:115–121.
11. Fizazi K, Dohollou N, Blay JY, et al. Ewing’s family of tumors in adults: 42. Urano F, Umezawa A, Hong W, et al. A novel chimera gene between EWS
multivariate analysis of survival and long-term results of multimodality therapy in and E1A-F, encoding the adenovirus E1A enhancer-binding protein, in extraos-
182 patients. J Clin Oncol. 1998;16:3736–3743. seous Ewing’s sarcoma. Biochem Biophys Res Commun. 1996;219:608–612.
12. Levine RG, Bono CM, Hameed M, et al. Ewing sarcoma in an octogenar- 43. Peter M, Couturier J, Pacquement H, et al. A new member of the ETS family
ian: a case report. J Bone J Surg Am. 2002;84:445–448. fused to EWS in Ewing tumors. Oncogene. 1997;14:1159–1164.
13. Dorfman H, Czerniak B. Bone cancers. Cancer. 1995;75(suppl):203–210. 44. Ohno T, Ouchida M, Lee L, et al. The EWS gene, involved in Ewing family
14. Himelstein BP, Dormans JP. Malignant bone tumors of childhood. Pediatr of tumors, malignant melanoma of soft tumors and desmoplastic small round cell
Clin North Am. 1996;43:967–983. tumors, codes for an RNA binding protein with novel regulatory domains. On-
15. Widhe B, Widhe T. Initial symptoms and clinical features in osteosarcoma cogene. 1994;9:3087–3097.
and Ewing sarcoma. J Bone Joint Surg Am. 2000;82:667–674. 45. Rossow KL, Janknecht R. The Ewing’s sarcoma gene product functions as
16. Wagner LM, Neel MD, Pappo AS, et al. Fractures in pediatric Ewing sar- a transcriptional activator. Cancer Res. 2001;61:2690–2695.
coma. J Pediatric Hematol Oncol. 2001;23:568–571. 46. Aman P, Panagopoulos I, Lassen C, et al. Expression patterns of the human
17. Hoffer FA. Primary skeletal neoplasms: osteosarcoma and Ewing sarcoma. sarcoma-associated genes FUS and EWS and the genomic structure of FUS. Gen-
Top Magn Reson Imaging. 2002;13:231–240. omics. 1996;37:1–8.
18. Nascimento AG, Unni K, Pritchard D, Cooper KL, Dahlin DC. A clinico- 47. Sementchenko VI, Watson DK. Ets target genes: past, present and future.
pathological study of 20 cases of large-cell (atypical) Ewing’s sarcoma of bone. Oncogene. 2000;19:6533–6548.
Am J Surg Pathol. 1980;4:29–36. 48. Folpe A, Chand EM, Goldblum JR, et al. Expression of Fli-1, a nuclear
19. Julia BA, Fidler ME, James NR, et al. Adamantinoma-like Ewing’s sarcoma: transcription factor, distinguishes vascular neoplasms from potential mimics. Am
genomic confirmation, phenotypic drift. Am J Surg Pathol. 1999;23:159–165. J Surg Pathol. 2001;25:1061–1066.
20. Hauben E, van den Broek LC, Van Marck E, Hogendoorn PC. Adamanti- 49. Kovar H. Context matters: the hen or egg problem in Ewing’s sarcoma.
noma-like Ewing’s sarcoma and Ewing’s-like adamantinoma: the t(11;22),t(12;22) Semin Cancer Biol. 2005;15:189–196.
status. J Pathol. 2001;195:218–221. 50. Amandine HLT, Yaacov BD. The role of Fli-1 in normal cell function and
21. Folpe AL, Goldblum JR, Rubin BP, et al. Morphologic and immunophe- malignant transformation. Oncogene. 2000;19:6482–6489.
notypic diversity in Ewing family tumors. Am J Surg Pathol. 2005;29:1025–1033. 51. Zucman J, Melot T, Desmaze C, et al. Combinatorial generation of variable
22. Suh CH, Ordonez NG, Hicks J, Mackay B. Ultrastructure of the Ewing’s fusion proteins in the Ewing family of tumours. EMBO J. 1993;12:4481–4487.
sarcoma family of tumors. Ultrastruct Pathol. 2002;26:67–76. 52. Kovar H. Ewing tumor biology: perspectives for innovative treatment ap-
23. Franchi A, Pasquinelli G, Cenacchi G, et al. Immunohistochemical and proaches. Adv Exp Med Biol. 2003;27–37.
ultrastructural investigation of neural differentiation in Ewing sarcoma/PNET of 53. Peter M, Gilbert E, Delattre O. A multiplex real-time PCR assay for the
bone and soft tissues. Ultrastruct Pathol. 2001;25:219–225. detection of gene fusions observed in solid tumors. Lab Invest. 2001;81:905–912.
24. Ambros IM, Ambros PF, Strehl S, Kovar H, Gadner H, Salzer-Kuntschik M. 54. Xiang Q, Long J, Brandon S, Rhett PK, Syed J, Ricardo L. Molecular di-
MIC2 is a specific marker for Ewing’s sarcoma and peripheral primitive neuro- agnosis of Ewing’s sarcoma/primitive neuroectodermal tumor in formalin-fixed
ectodermal tumors: evidence for a common histogenesis of Ewing’s sarcoma and paraffin-embedded tissues by RT-PCR and fluorescence in situ hybridization.
peripheral primitive neuroectodermal tumors from MIC2 expression and specific Diagn Mol Pathol. 14:23–28.
chromosome aberration. Cancer. 1991;67:1886–1893. 55. Udayakumar AM, Sundareshan TS, Mallana GT, et al. Cytogenetic char-
25. Bantig GS, Pym B, Darling SM, Goodfellow PN. The MIC2 gene product: acterization of Ewing tumors using fine needle aspiration samples: a 10 year
epitope mapping and structural prediction analysis define an integral membrane experience and review of literature. Cancer Genet Cytogenet. 2001;127:42–48.
protein. Mol Immunol. 1989;26:181–188. 56. Udayakumar AM, Sundareshan TS. Cytogenetic characterization of Ewing
26. Darling SM, Goodfello PJ, Pym B, Banting GS, Pritchard C, Goodfellow tumors: further update on 20 cases [letter]. Cancer Genet Cytogenet. 2002;133:
PN. Molecular genetics of MIC2: a gene shared by the human X and Y chro- 102–103.
mosomes. Cold Spring Harb Symp Quant Biol. 1986;51:205–212. 57. Rodiguez-Galindo C. Pharmacological management of Ewing sarcoma
27. Folpe AL, McKenney JK, Bridge JA, Weiss SW. Sclerosing rhabdomyosar- family of tumours. Expert Opin Pharmacother. 2004;17:329a.
coma in adults: report of four cases of a hyalinizing, matrix-rich variant of rhab- 58. Carpentieri DF, Qualman SJ, Bowen J, Krausz T, Marchevsky A, Dickman
domyosarcoma that may be confused with osteosarcoma, chondrosarcoma, or PS. Protocol for the examination of the specimens from pediatric and adult pa-
angiosarcoma. Am J Surg Pathol. 2002;26:1175–1183. tients with osseous and extraosseous Ewing sarcoma family of tumors, including
28. Lae ME, Roche PC, Jin L, Lloyd RV, Nascimento AG. Desmoplastic small peripheral primitive neuroectodermal tumor and Ewing sarcoma. Arch Pathol Lab
round cell tumor: a clinicopathologic, immunohistochemical and molecular Med. 2005;129:866–873.
study of 32 tumors. Am J Surg Pathol. 2002;26:823–835. 59. Schoedel K, Dickman PS, Krailo M, Perlman EJ, Miser J, Grier H. Histologic
29. Ozdemirli M, Fanburg-Smith JC, Hartmann DP, Azumi N, Miettinen M.
response to chemotherapy and prognosis in osseous Ewing’s sarcoma/primitive
Differentiating lymphoblastic lymphoma and Ewing’s sarcoma:lymphocyte mark-
neuroectodermal tumor (ES/PNET) [abstract]. Int Surg Pathol. 1995;2(suppl):443.
ers and gene arrangement. Mod Pathol. 2001;14:1175–1182.
60. Cotterill SJ, Ahrens S, Paulussen M, et al. Prognostic factors in Ewing’s
30. Granter SR, Renshaw AA, Fletcher CD, Bhan AK, Rosenberg AE. CD99
tumor of bone: analysis of 975 patients from the European intergroup cooperative
reactivity in mesenchymal chondrosarcoma. Hum Pathol. 1996;27:1273–1276.
31. Llombart-Bosch A, Navarro S. Immunohistochemical detection of EWS and Ewing’s sarcoma study group. J Clin Oncol. 2000;18:3108–3114.
FLI-1 proteins in Ewing sarcoma and primitive neuroectodermal tumors:compar- 61. Delepine N, Delepine G, Cornille H, Voisin MC, Brun B, Desbois JC. Prog-
ative analysis with CD99 (MIC-2) expression. Appl Immmunohistochem Mol Mor- nostic factors in patients with localized Ewing’s sarcoma: the effect on survival
phol. 2001;9:255–260. of actual received drug dose intensity and of histologic response to induction
32. Fellinger EJ, Garin-Chesa P, Glasser DB, Huvos AG, Rettig WJ. Comparison therapy. J Chemother. 1997;9:352–363.
of cell surface antigen HBA71 (p30/32MIC2), neuron-specific enolase and vi- 62. Picci P, Rougraff BT, Bacci G, et al. Prognostic significance of histopath-
mentin in the immunohistochemical study of Ewing’s sarcoma of bone. Am J Surg ologic response to chemotherapy in nonmetastatic Ewing’s sarcoma of extremi-
Pathol. 1992;16:746–755. ties. J Clin Oncol. 1993;11:1763–1769.
33. Amann G, Zoubek A, Salzer-Kuntschik M, Windhager R, Kovar H. Relation 63. Burchill SA. Ewing’s sarcoma: diagnostic, prognostic and therapeutic im-
of neurological marker expression and EWS gene fusion types in MIC2/CD99- plications of molecular abnormalities. J Clin Pathol. 2003;56:96–102.
positive tumors of the Ewing Family. Hum Pathol. 1999;30:1058–1064. 64. Amiel A, Ohali A, Fejgin M, et al. Molecular cytogenetic parameters in
34. Gu M, Antonesu CR, Guiter G, Huvos AG, Ladanyi M, Zakowski MF. Cy- Ewing sarcoma. Cancer Genet Cytogenet. 2003;140:107–112.
tokeratin immunoreactivity in Ewing’s sarcoma: prevalence in 50 cases confirmed 65. Zoubek A, Dockhorn-Dwarniczak B, Delatire O, et al. Does expression of
by molecular diagnostic studies. Am J Surg Pathol. 2000;24:410–416. different EWS transcripts define clinically distinct risk groups of Ewing tumor
35. Aurias A, Rimbaut C, Buffe C, Dubousset J, Mazabraud A. Chromosomal patients? J Clin Oncol. 1996;14:245–251.
translocations in Ewing’s sarcoma. N Engl J Med. 1983;309:496–497. 66. Lin PP, Brody RJ, Hamelin AC, et al. Differential transactivation by alter-
36. Miozzo M, Sozzi G, Calderone C, et al. t(11;22) in three cases of periph- native EWS-FLI1 fusion proteins correlates with clinical heterogeneity in Ewing’s
eral neuroepithelioma. Genes Chromosomes Cancer. 1990;2:163–165. sarcoma. Cancer Res. 1999;59:1428–1455.

Arch Pathol Lab Med—Vol 131, February 2007 Small Round Cell Tumors of Bone—Hameed 203
67. Fagnou C, Michon J, Peter M, et al. Presence of tumor cells in bone mar- 101. Salvador AH, Beabout JW, Dahin DC. Mesenchymal chondrosarcoma:
row but not in blood is associated with adverse prognosis in patients with Ewing’s observation on 30 new cases. Cancer. 1971;28:605–616.
tumor: Societe Francaise d’Oncologie Pediatrique. J Clin Oncol. 1998;16:1707– 102. Harwood AR, Krajbich JI, Fornasier VL. Mesenchymal chondrosarcoma:
1711. a report of 17 cases. Clin Orthop Relat Res. 1981;158:144–148.
68. Zoubek A, Ladenstein R, Windhager R, et al. Predictive potential of testing 103. Zura RD, Minasi JS, Kahler DM. Tumor-induced osteomalacia and symp-
for bone marrow involvement in Ewing’s tumor patients by RT-PCR: a preliminary tomatic looser zones secondary to mesenchymal chondrosarcoma. J Surg Oncol.
evaluation. Int J Cancer. 1998;79:56–60. 1999;71:58–62.
69. West DC, Grier HE, Swallow MM, et al. Detection of circulating tumor 104. Shinaver CN, Mafee MF, Choi KH. MRI of mesenchymal chondrosarcoma
cells in patients with Ewing’s sarcoma and primitive peripheral neuroectodermal of the orbit: case report and review of literature. Neuroradiology. 1997;39:296–
tumor. J Clin Oncol. 1997;15:583–588. 301.
70. Schleiermacher G, Peter M, Oberlin O, et al. Increased risk of systemic 105. Ariyoshi Y, Shimahara D. Mesenchymal chondrosarcoma of the maxilla:
relapses associated with bone marrow micrometastases and circulating tumor report of a case. J Oral Maxillofac Surg. 1999;57:733–737.
cells in localized Ewing’s tumor. J Clin Oncol. 2003;21:85–91. 106. Steiner GC, Mirra JM, Bullough PG. Mesenchymal chondrosarcoma: a
71. Kennedy JG, Frelinghuysen P, Hoang BH. Ewing sarcoma: current concepts study of the ultrastucture. Cancer. 1973;32:926–939.
in diagnosis and treatment. Curr Opin Pediatr. 2003;15:53–57. 107. Granter SR, Renshaw AA, Fletcher CD, Bhan AK, Rosenberg AE. CD99
72. Kovar J, Aryee D, Zoubek A. The Ewing family of tumors and the search reactivity in mesenchymal chondrosarcoma. Hum Pathol. 1996;27:1273–1276.
for the Achilles’ heel. Curr Opin Oncol. 1999;11:275–292. 108. Aigner T, Loos S, Muller S, Sandell LJ, Unni KK, Kirshner T. Cell differ-
73. Prieur A, Tirode F, Cohen P, et al. EWS/FLI-1 silencing and gene profiling entiation and matrix gene expression in mesenchymal chondrosarcoma. Am J
of Ewing cells reveal downstream oncogenic pathways and a crucial role for Pathol. 2000;156:1327–1335.
repression of insulin-like growth factor binding protein 3. Mol Cell Biol. 2004; 109. Wehrli BM, Huang W, De Crombrugghe B, Ayala AG, Czerniak B. Sox9,
24:7275–7283. a master regulator of chondrogenesis, distinguishes mesenchymal chondrosar-
74. Ouchida M, Ohno T, Fujimara Y, et al. Loss of tumorigenecity of Ewing’s coma from other small round cell tumors. Hum Pathol. 2003;34:263–269.
sarcoma cells expressing antisense RNA to EWS-fusion transcripts. Oncogene. 110. Ng LJ, Wheatley S, Muscat GEO, et al. Sox9 binds DNA, activates tran-
1995;11:1049–1054. scription and co express with type II collagen during chondrogenesis in mouse.
75. Parker F, Jackson H Jr. Primary reticulum cell sarcoma of bone. Surg Gy- Dev Biol. 1997;183:108–121.
necol Obstet. 1939;68:45–53. 111. Bi W, Deng JM, Zhang Z, et al. Sox9 is required for cartilage formation.
76. Oberling C. Les reticulosarcomes et les reticuloendotheliosarcomes de la Nat Genet. 1999;22:85–89.
moelle osseuse (sarcoma d’Ewing). Bull Assoc Fr Etude Cancer (Paris). 1928;17: 112. Dobin SM, Donner LR, Speights VO Jr. Mesenhymal chondrosarcoma: a
259–296. cytogeentic, immunohistochemical and ultrastructural study. Cancer Genet Cy-
77. Unni KK, Hogendoorn PCW. Malignant lymphoma. In: Fletcher CDM, togenet. 1005;83:56–60.
Unni KK, Mertens F, eds. Pathology and Genetics of Tumours of Soft Tissue and 113. Naumann S, Krallman PA, Unni KK, Fidler ME, Neff JR, Bridge JA. Trans-
Bone. Lyon, France: IARC Press; 2002. World Health Organization Classification location der(13;21)(q10;q10) in skeletal and extraskeletal mesenchymal chondro-
of Tumours. sarcoma. Mod Pathol. 2002;15:572–576.
78. Freeman C, Berg JW, Cutler SJ. Occurrence and prognosis of extranodal 114. Sainati L, Scapinello A, Montaldi A, et al. Mesenchymal chondrosarcoma
lymphomas. Cancer. 1972;28:252–260. of a child with reciprocal translocation (11;22)(q24;q12), Cancer Genet Cytoge-
79. Rodman D, Raymond AK, Phillips WC. Case report 201: primary lympho- net. 1993;71:144–147.
ma of bone (PLB)—left fibula. Skeletal Radiol. 1982;8:235–237. 115. Park YK, Park HR, Chi SG, et al. Overexpression of p53 and rare genetic
80. Rosenberg SA, Diamond HD, Jasowitz B, Craver LF. Lymphosarcoma: a mutation in mesenchymal chondrosarcoma. Oncol Rep. 2000;7:1041–1047.
review of 1269 cases. Medicine. 1961;40:31–81. 116. Bae DK, Park YK, Chi SG, Lee CW, Unni KK. Mutational alterations of
81. Rudder RA, Ross ME, De Lellis RA. Primary extranodal lymphoma: re- the p16CDKN2A tumor suppressor gene have low incidence in mesenchymal
sponse to treatment and factors influencing prognosis. Cancer. 1978;42:406–416. chondrosarcoma. Oncol Res. 2000;12:5–10.
82. Heyning FH, Hogendoorn PC, Kramer MN, et al. Primary non-Hodgkin’s 117. Brown RE, Boyle JL. Mesenchymal chondrosarcoma: molecular charac-
lymphoma of bone: a clinicopathological investigation of 60 cases. Leukemia. terization by a proteomic approach with morphogenic and therapeutic implica-
1999;13:2094–2098. tions. Ann Clin Lab Sci. 2003;33:131–141.
83. Barr J, Burkes RL, Bell R, Blackstein M, Fernandes B, Langer F. Primary 118. Dowling EA. Mesenchymal chondrosarcoma. J Bone Joint Surg. 1964;46-
non-Hodgkin’s lymphoma of bone. Cancer. 1994;73:1194–1199. A:747–754.
84. Dubey P, Chul SH, Besa P, et al. Localized primary malignant lymphoma 119. Vencio EF, Reeve CM, Unni KK, Nascimento AG. Mesenchymal chon-
of bone. Int J Radiat Oncol Biol Phys. 1997;37:1087–1093. drosarcoma of the jaw bones: clinicopathologic study of 19 cases. Cancer. 1998;
85. Moses AM, Spencer H. Hypercalcemia in patients with malignant lym- 82:2350–2355.
phoma. Ann Int Med. 1963;59:531–536. 120. Nussbeck W, Neureiter D, Soder S, Inwards C, Aigner T. Mesenchymal
86. Dorfman H, Cherniak B. Bone Tumors. St Louis, Mo: Mosby; 1998. chondrosarcoma: an immunohistochemical study of 10 cases examining prog-
87. Braunstein EM, White SJ. Non-Hodgkin’s lymphoma of bone. Radiology. nostic significance of proliferative activity and cellular differentiation. Pathology.
1980;135:59–63. 2004;36:230–233.
88. Clayton F, Butler JJ, Ayala AG, Ro JY, Zornoza J. Non-Hodgkin’s lymphoma 121. Sim FH, Unni KK, Beabout JW, Dahlin DC. Osteosarcoma with small
in bone. Cancer. 1987;60:2494–2501. cells simulating Ewing’s tumor. J Bone Joint Surg Am. 1979;61:207–215.
89. Mulligan ME, McRae GA, Murphy MD. Imaging features of primary lym- 122. Dahlin DC, Unni KK. General Aspects and Data on 11087 Cases. 5th
phoma of bone. Amer J Radiol. 1999;173:1691–1697. ed. Philadelphia, Pa: Lippincott-Raven, 1996.
90. Ozdemirli M, Fanburg-Smith J, Hartmann DP, et al. Precursor B-lympho- 123. Ayala AG, Ro JY, Raymond K, et al. Small cell osteosarcoma: a clinico-
blastic lymphoma presenting as a solitary bone tumor and mimicking Ewing’s pathologic study of 27 cases. Cancer. 1989;64:2162–2173.
sarcoma: a report of four cases and review of the literature. Am J Surg Pathol. 124. Ayala AG, Ro JY, Papadopoulos NK, Raymond KA, Edeiken J. Osteosar-
1998;22:795–804. coma in Adolescents and Young Adults. Boston, Mass: Kluwer Academic Publish-
91. Takemoto S, Matsuoka M, Sakata K, et al. Primary adult T cell leukemia ers; 1993.
of bone: two patients with primary bone lesion showing monoclonal integration 125. Nakajima H, Sim FH, Bond JR, Unni KK. Small cell osteosarcoma of
of HTLV-1 proviral DNA. Leukemia. 1996;10:333–337. bone: review of 72 cases. Cancer. 1997;79:2095–2106.
92. Antillon F, Behm FG, Raimondi SC, Kaste SC, Sandland JT, Pappo AS. 126. Park SH, Kim I. Small cell osteogenic sarcoma of the ribs: cytological,
Pediatric primary diffuse large cell lymphoma of bone with t(3;22)(q27;q11). J immunohistochemical and ulttrastructural study with literature review. Ultra
Pediatr Hematol Oncol. 1998;20:552–555. Pathol. 1999;23:133–140.
93. Misgeld E, Wehmeier A, Kromeke O, Gattermann N. Primary non-Hodg- 127. Bertoni F, Present D, Bacchini P, Pignatti G, Picci P, Campanacci M. The
kin’s lymphoma of bone: three cases and a short review of the literature. Ann Instituto Rizzoli experience with small cell osteosarcoma. Cancer. 1989;64:2591–
Hematol. 2003;82:440–443. 2599.
94. Ostowski M, Unni KK, Banks PM, et al. Malignant lymphoma of bone. 128. Martin SE, Dwyer A, Kissane TJ. Small cell osteosarcoma. Cancer. 1982;
Cancer. 1986;58:2646–2655. 50:990–996.
95. Mendenhall NP, Jones JJ, Kramer BS, et al. The management of primary 129. Devoe K, Weidner N. Immunohistochemistry of small round cell tumors.
lymphoma of bone. Radiother Oncol. 1987;8:137–145. Semin Diagn Pathol. 2000;17:216–224.
96. Lichtenstein L, Bernstein D. Unusual benign and malignant chondroid tu- 130. Devaney K, Vinh TN, Sweet DE. Small cell osteosarcoma: an immuno-
mors of bone. Cancer. 1959;12:1142–1157. histochemical study with differential diagnostic considerations. Hum Pathol.
97. Dahlin DC, Henderson ED. Mesenchymal chondrosarcoma: further obser- 1993;24:1211–1225.
vations on a new entity. Cancer. 1962;15:410–417. 131. Thomas F, Lipton J, Barbera C, Vigorita V, Bryk E. Primary rhabdomyo-
98. Bertoni F, Picci P, Bacchini P. Mesenchymal chondrosarcoma of bone and sarcoma of the humerus: a case report and review of the literature. J Bone Joint
soft tissues. Cancer. 1983;52:533–541. Surg Am. 2002;84-A:813–817.
99. Nakashima Y, Unni KK, Shives TC, Sweet RG, Dahlin DC. Mesenchymal 132. Murphy A, Stallings RL, Howard J, et al. Primary desmoplastic small
chondrosarcoma of bone and soft tissue: a review of 111 cases. Cancer. 1986; round cell tumor of bone: report of a case with cytogenetic confirmation. Cancer
57:2444–2453. Genet Cytogenet. 2005;156:167–171.
100. Dabska M, Huvos AG. Mesenchymal chondrosarcoma in the young: a 133. Pappo AS, Douglass EC, Meyer WH, et al. Use of HBA-71 and anti-beta
clinicopathological study of 19 patients with explanation of histogenesis. Vir- microglobulin to distinguish peripheral neuroepithelioma from neuroblastoma.
chows Arch Pathol Histopathol. 1983;399:89–104. Hum Pathol. 1993;24:880–885.

204 Arch Pathol Lab Med—Vol 131, February 2007 Small Round Cell Tumors of Bone—Hameed

You might also like