Small Round Cell Tumors of Bone

Small Round Cell Tumors of Bone Meera Hameed, MD ● Context.—Primary small round cell tumors of the bone are a heterogeneous group of malignant neopla...
Author: Randolf Summers
29 downloads 1 Views 2MB Size
Small Round Cell Tumors of Bone Meera Hameed, MD

● Context.—Primary small round cell tumors of the bone are a heterogeneous group of malignant neoplasms presenting predominantly in children and adolescents. They include Ewing sarcoma/peripheral neuroectodermal tumor or Ewing family tumors, lymphoma, mesenchymal chondrosarcoma, and small cell osteosarcoma. Even though they share many morphological similarities, their unique biological and genetic characteristics have provided substantial insights into the pathology of these diverse neoplasms. Objective.—To provide an overview of the clinical, radiologic, pathologic, and genetic characteristics of these tumors along with a pertinent review of the literature.

Data Sources.—A literature search using PubMed and Ovid MEDLINE was performed, and data were obtained from various articles pertaining to clinicopathologic, biological, and genetic findings in these tumors. Additionally, findings from rare cases have been included from author’s subspecialty experience. Conclusion.—The diagnosis of small round cell tumors can be made accurately by applying clinicopathologic criteria, as well as a panel of immunohistochemical and genetic studies in appropriate cases. Molecular genetic studies may provide further insight into the biology, histogenesis, and prognosis of these tumors. (Arch Pathol Lab Med. 2007;131:192–204)

EWING SARCOMA FAMILY OF TUMORS Since the original descriptions of tumors of the Ewing sarcoma family in 1918 by Arthur Purdy Stout1 and in 1921 by James Ewing, who called the tumor diffuse endothelioma of bone,2 several nomenclatures have been assigned to these tumors, albeit with much skepticism in the literature3 A possible relationship between Ewing sarcoma (ES) and peripheral neuroectodermal tumor (PNET) was established after the description of extraosseous ES by Angerwall and Enzinger4 and PNET of the bone by Jaffe.5 At the present time, ES and PNET, otherwise called Ewing family tumors (EFTs), form a single neoplastic entity sharing common histologic and molecular features, and differing only in their extent of cellular differentiation. Askin’s tumor of the thoracopulmonary region is also currently grouped under EFT. Nonrandom translocation involving the EWS gene with one of the ETS family of transcription factors is the hallmark of their oncogenesis.6–8

bones, the femur is most commonly affected, followed by tibia, fibula, and humerus. However, virtually any bone can be affected, including the acral skeleton, craniofacial bones, and clavicle.13 Neoplasms in the long bones are often diaphyseal.14,15

Incidence and Location Ewing family tumor commonly occurs in the adolescent age group between 10 and 20 years, but is extremely rare in African American children.9 The annual incidence is about 3 per million, and there is a slight male predominance.10 These neoplasms constitute the second most common pediatric malignancy occurring in the skeletal system.10 Adult patients (older than 40 years) with EFT are rare, but isolated cases in patients up to 81 years old have been reported.11,12 Ewing family tumor involves both the axial and appendicular skeleton, including the pelvis. Among long tubular Accepted for publication April 19, 2006. From the University of Medicine and Dentistry of New Jersey–New Jersey Medical School, Newark. The author has no relevant financial interest in the products or companies described in this article. Reprints: Meera Hameed, MD, Surgical Pathology, UMDNJ–New Jersey Medical School, Newark, NJ 07103 (e-mail: hameedmr@ umdnj.edu). 192 Arch Pathol Lab Med—Vol 131, February 2007

Clinical Presentation Local tenderness or mass is the most common clinical presentation. In about 10% to 15% of the cases, there is a pathological fracture when the femur is involved.16 It is not uncommon to encounter children with systemic symptoms such as fever, fatigue, leukocytosis, and raised erythrocyte sedimentation rate mimicking signs and symptoms of acute osteomyelitis.15 This is an important clinical and radiologic differential diagnosis. Radiographic Features Radiographic correlation is a prerequisite to all orthopedic pathology and more so in tumors such as EFT, where clinical findings may point to benign processes such as osteomyelitis. The radiographic features are that of a diaphyseal or metadiaphyseal lesion in the long bones. Typically, the lesion is intramedullary (symmetric or eccentric) and is associated with cortical thickening and cyclical periosteal reaction, giving rise to the characteristic onion-skin appearance (Figure 1). The rapid growth of the tumor produces a continuous lifting of the periosteum leading to perpendicular striations. Within the marrow, the lesion is poorly marginated, with a permeative pattern of osteolysis (without a beginning and an end) (Figure 1). Magnetic resonance imaging reveals an accompanying soft tissue mass in about 90% of cases (Figures 2 and 3). Computed tomographic scan will demonstrate that the lesion itself does not produce any matrix (bone or cartilage). Occasionally there is prominent diffuse thickening of the bone and cloudy opacities mimicking osteosarcoma (OS). Radiologic differential diagnoses include osteomyelitis, Langerhans histiocytosis, lymphoma, and OS.17 Imaging workup of these patients involves skeletal scintigraphy (bone scan) and computed tomography of the chest to rule out metastatic disease at the time of presentation. 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 An open biopsy is usually performed to obtain adequate material for pathological examination and ancillary studies. Multiple core biopsies may be performed in patients with large, accessible soft tissue component of the tumors, keeping in mind that additional studies such as cytogenetics will be needed. Fine-needle aspiration can be used in an appropriate setting where adequate material can be obtained. Grossly, the biopsied material is often hemorrhagic and necrotic and usually has a tan-gray appearance. Touch imprints of the submitted pieces allow one not only to examine morphology, but also to select viable material for cytogenetic analysis. Molecular studies (reverse transcriptase polymerase chain reaction [RT-PCR] and fluorescence in situ hybridization [FISH]) can be done using frozen and paraffin-embedded tissue. If tissue is snap-frozen for molecular studies, care should be taken to freeze tumor material in a timely manner to avoid RNA degradation. Whenever possible, bony spicules should be separated from soft tissue to avoid unnecessary decalcification of the soft tissue component. Microscopic examination of classic EFT shows a highly cellular neoplasm consisting of a monotonous population of uniform round blue cells (diameter, about 14 ␮m) arranged in a nested, sheetlike, or solid pattern. The nuclei are round with fine chromatin, scant clear or eosinophilic cytoplasm, indistinct cytoplasmic membranes, and 1 to 3 small to medium-sized nucleoli. The cytoplasm contains Arch Pathol Lab Med—Vol 131, February 2007

abundant glycogen, which can be confirmed with periodic acid–Schiff stain. Necrosis and apoptosis are common. The presence of apoptotic cells sometimes gives the biphasic appearance of dark and light cells (Figure 4). HomerWright rosettes and pseudorosettes can be present (Figure 5). Stromal elements are minimal, and the tumor generally fills the medullary cavity and replaces bone marrow elements. Interspersed collagen bundles are usually seen in the soft tissue component. A vascular component is not prominent and is usually composed of slitlike capillaries. Thick-walled vessels are seen in areas where there are prominent stroma and collagen bundles. A minimal degree of spindle cell change can be present. The tumor cells permeate the cortex and elicit periosteal new bone formation with reactive osteoblasts, osteoclasts, and sometimes cartilaginous metaplasia. The tumor generally does not induce an inflammatory response. Morphological Variants A number of variants have been described in EFT. These include atypical ES (large cell EFT),18 adamantinoma-like ES,19,20 sclerosing ES, and spindle cell sarcoma–like EFT.21 The tumor cells of the atypical or large cell EFT are heterogeneous compared with classic EFT. Their size ranges between 20 and 24 ␮m, and they have irregular nuclear membranes and prominent nucleoli (Figure 6). The cytoplasm is vacuolated, and cytoplasmic glycogen is less 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 large cells can be accompanied by spindled tumor cells. Adamantinoma-like EFT has been described in the tibia and fibula and shares histological and immunohistochemical features of adamantinoma, such as distinct nests of moderately pleomorphic tumor cells with peripheral palisading, a prominent desmoplastic host response, and positive immunohistochemistry for cytokeratins. These tumors are also positive for CD99 and FLI-1 and show EWSFLI-1 fusion transcript.19 We have seen a tumor of the pelvis with similar histology which showed positive staining for pan-cytokeratin with a variant EWS translocation, as evidenced by positive break-apart FISH with EWS probe

Table 1. Tumor

CD99

FLI-1

(Figures 7 and 8). RT-PCR for EWS-FLI-1, ERG, and WT1 fusion transcripts were negative in this tumor. Sclerosing EFT resembles sclerosing epithelioid fibrosarcoma or sclerosing rhabdomyosarcoma because of the presence of hyalinized eosinophilic matrix. However, there are always at least focal areas showing typical features of classic EFT.21 Spindle cell sarcoma–like EFT shows a greater degree of spindling than classic EFT and thus can resemble synovial sarcoma.21 Ultrastructure Transmission electron microscopy of EFTs demonstrates densely packed undifferentiated tumor cells with cyto-

Immunohistochemical Profile* 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, desmoplastic 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, desmosome-like junctions are seen. Evidence of neural differentiation such as dense core neurosecretory granules, dendritic 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 include 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 lymphoblastic lymphoma/acute lymphoblastic leukemia, rhabdomyosarcoma, 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 chromosomes.24–26 Apart from EFT, this protein is also highly expressed 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 tumors, synovial sarcomas, and mesenchymal chondrosarcomas.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 lymphoblastic lymphomas and other NHLs.31 Other variably expressed markers in EFT include vimentin, cytokeratin (up to 25%), desmin (very rare), and neural markers such as synaptophysin, CD57, S100, and chromogranin.21,23,32–34 The presence of neural markers in an EFT indicates neuroectodermal differentiation (PNET). High molecular cytokeratin (34␤E12) is positive in adamantinoma-like ES and not in classic and other variants of EFT.21 Table 1 summarizes the immunohistochemical profile of these tumors. Cytogenetics and Molecular Genetics The pathogenetic relationship of ES and PNET was defined by the discovery of the unique and specific nonrandom chromosomal translocation, seen in about 85% to 95% of the tumors, involving chromosome 11q24 and chromosome 22q12, t(11;22)(q24;q12) (Figure 11).35–37 This leads to an in-frame fusion of the EWS gene at chromo-

Table 1.

Extended

Desmin

Myogenin/MYOD1

WT1

⫺ ⫺ ⫺ ⫹/⫺ ⫹/⫺ ⫹ ⫹ ⫺

⫺ ⫺ ⫺ ⫹/⫺ ? ⫹ ⫺ ⫺

⫺ ⫹/⫺ ⫺ ⫺ ? ⫹/⫺(cyto) ⫹ ⫺

Arch Pathol Lab Med—Vol 131, February 2007

Figure 9. Immunohistochemical stain for CD99 showing diffuse positivity in typical Ewing sarcoma (original magnification ⫻400). Figure 10. Immunohistochemical staining for FLI-1 showing intense nuclear staining in tumor cells.

some 22q12 to the FLI-1 gene at chromosome 11q24. The EWS-FLI-1 chimeric protein on the (der)22, contains the 5’ end of the EWS gene and the 3’ end of the FLI-1 gene.38 The second most common translocation in EFT is t(12; 22)(q22;q12), seen in about 5% to 10% of the cases, resulting in the fusion of EWS to ERG at chromosome 12q22.39 Other variant translocations reported in bone EFT accounting for less than 1% of cases include t(7;22)(p22; q12), t(17;22)(q12;q12), and t(2;22)(q23;q12), involving fusion 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 encode the N-terminal region responsible for regulating 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 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 conserved ETS domain recognizing a core DNA motif of GGAA/T.47 FLI-1 expression is seen in high levels in heSmall 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 derived from neural crest cells.48,49 The product of EWS-ETS translocation results in fusion proteins containing the Cterminal region of the FLI-1 and N-terminal region of EWS, thus bringing the ETS component under the powerful transactivating domain of EWS.50 These fusion products modulate multiple target genes, resulting in malignant transformation. The most commonly used molecular assays to identify Table 2.

Translocation and Fusion Genes in Ewing Family Tumors of Bone

Translocation

t(11;22)(q24;q12) t(21;22)(q22;q12) t(7;22)(p22;q12) t(17;22)(q12;q12) t(2;22)(q23;q12)

Fusion Genes

Frequency

EWS-FLI-1 EWS-ERG EWS-ETV1 EWS-E1AF EWS-FEV

85%–95% 5%–10% ⬍1% ⬍1% ⬍1%

196 Arch Pathol Lab Med—Vol 131, February 2007

the EWS-ETS fusion are FISH and RT-PCR. Multiple chimeric transcripts are possible, all of which involve exons 1 through 7 of EWS and exon 9 of the ETS gene. The genomic break points are intronic, and occur at 1 of 4 EWS introns and, in the case of EWS-FLI-1, 1 of 6 FLI-1 introns (Figure 12).51 Type 1 (exon 7 of EWS to exon 6 of FLI-1) and type II (exon 7 of EWS to exon 5 of FLI-1) are the most common (⬎85%) fusion transcripts.52 Commonly 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 6 to detect type 1 or type II fusion products, which would identify 85% of the cases (Figure 13). In addition, RT-PCR for EWS-ERG fusion product can be performed, which would identify the 10% of cases with this fusion. A multiplex real-time PCR has also been used to identify EWSETS fusion products.53 RT-PCR has also been applied to formalin-fixed, paraffin-embedded tissues, and a combination of FISH and RT-PCR may yield increased sensitivSmall Round Cell Tumors of Bone—Hameed

Figure 15. Gross photograph (A) and specimen 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 metaphysis and diaphysis. Periostitis is seen adjacent to lateral cortex.

ity and accuracy due to limitations of formalin-fixed, paraffin-embedded tissue material with suboptimal preservation of nucleic acids.54 Fluorescence in situ hybridization assay with the commercially available (such as Vysis) dual-color break-apart DNA probes flanking the EWS break point region on chromosome 22 detects the translocation of EWS and can be used in fresh and formalinfixed, paraffin-embedded tissues. In a metaphase spread, the split signal enables one to identify the chromosome involved in the translocation; however, the partner gene is not identified by this methodology (Figure 14). Additional reported secondary karyotypic changes in EFT include gains of chromosomes 8, 12, and 18, deletion of the short arm of chromosome 1, unbalanced translocation t(1;16)(q12;q11), and other rare structural abnormalities.55,56 Treatment and Treatment Response The management of EFTs includes systemic chemotherapy followed by surgery, with or without radiation and continuation chemotherapy.57 Induction chemotherapy before definitive surgery allows us to assess the chemoresTable 3. Grade

I IIA IIB III IV

Chemotherapy Response—Pediatric Oncology Group Study Tumor Response

3-y Survival, %

No chemotherapy effect 1%–10% necrosis 11%–90% necrosis 91%–99% necrosis 100% necrosis

30 30 49 73 100

Arch Pathol Lab Med—Vol 131, February 2007

ponse in resected specimens, which has some prognostic significance and has a role in selection of agents for continuation chemotherapy. A sagittal section of the resected bone, with mapping and examining the entire central face of the tumor and adjacent tissue after careful decalcification, allows one to assess the chemotherapeutic response (Figure 15).58 The grading system used to assess chemoresponse is similar to that used in OS (Table 3).58,59 Prognosis and Outcome Up to one third of patients with EFT have metastatic disease at the time of presentation, and their outcome remains poor in spite of aggressive chemotherapy.60 The most common sites of metastases are lung, bone, and bone marrow. Patients who present only with lung metastases have better survival compared with those who present with bone metastases.61 One third of the patients with localized disease are also prone to distant relapse, especially if there are residual viable tumor cells at definitive resection.61,62 Multiple biological factors, such as type of fusion transcript, mutations in INK4a gene, mutations in p53, deletion 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 transcript has been reported as being associated with improved outcome compared to other fusion transcript types, and it appears that type 1 fusion transcript encodes a less active chimeric protein.65,66 Reports on the detection of minimal residual disease by RT-PCR for the chimeric proteins in peripheral blood and bone marrow remain controversial with respect to disease progression.67–70 Current chemotherapeutic regimens with selective surgery Small Round Cell Tumors of Bone—Hameed

197

have significantly improved the overall survival up to 60% to 80% in nonmetastatic EFT.71 Radiation-induced sarcomas and chemotherapy-related acute leukemias are the most frequent secondary malignancies.13 Conclusion Even though the cell of origin is still unknown in EFT, considerable strides have been made since the original description by Drs Stout and Ewing. The combination of morphology, immunohistochemistry, and molecular genetics has opened many doors, including some novel approaches to therapeutics, such as the use of CD99 as a target,72 inhibition of IGF-1 receptor with antisense oligonucleotides,73 and small interfering RNA (siRNA)74 all of which have shown promising results in cell lines and animal models. NON-HODGKIN LYMPHOMA Primary NHL of bone was described as a series, first by Jackson and Parker in 1939.75 Before this, Oberling in 1928 had reported a case of reticuloendothelial sarcoma histologically indistinguishable from ES.76 The description of this rare entity is restricted to lymphoma involving a single skeletal site with or without regional lymph node involvement or multiple skeletal sites without visceral or lymph node involvement.77 Incidence and Location Approximately 3% to 7% of extranodal lymphomas present as primary bone neoplasms,78–81 and lymphomas constitute about 7% of all bone malignancies.77 The disease tends to involve older adults (older than 40 years), and there is a male predominance, with male-female ratio ranging from 1.2:1 to 1.6:1.82 The femur, pelvis, vertebra, and humerus are the most common sites. In the long bones, the preferred site is the metadiaphyseal region.83,84 Involvement of the small bones of the hand and feet is extremely unusual. It is not uncommon to encounter multifocality, and soft tissue extension may be present.77,83 Clinical Presentation Most patients present with pain, and depending on location, such as spine, neurological symptoms may be present. Systemic symptoms such as fever (as seen in ES) and B symptoms of nodal lymphomas are unusual.83 Occasionally patients can present with hypercalcemia, lethargy, constipation, etc.85 For a neoplasm to be classified as a primary bone lymphoma, an interval of 4 to 6 months between the skeletal manifestation and extraskeletal disease is required.86 Thus appropriate staging procedures, such as skeletal imaging, computed tomographic scans, bone scan/positron emission tomography, and bone marrow biopsy, are an integral part of the patient workup.87 Radiographic Features On conventional x-rays, primary lymphomas are lytic radiolucent lesions with a moth-eaten or permeative destructive pattern. This may be associated with sclerotic areas (Figure 16).84,88 These changes are nonspecific and overlap with other round cell tumors. Cortical erosion and destruction with soft tissue extension is frequently seen (Figure 17). Periosteal reaction (interrupted or solid singlelayer) can be present, but less so than in ES. Generally the disease tends to involve the long bones extensively, sometimes occupying the entire length. In some instances, the 198 Arch Pathol Lab Med—Vol 131, February 2007

tumor may evoke a sclerotic response and present as a blastic lesion, which can sometimes be mistaken for Paget disease. Computed tomography and magnetic resonance imaging are useful in assessing extent of disease, and positron emission tomography and bone scans are used for staging and assessment of remission after therapy. Of importance is the fact that occasionally plain radiographs can be normal, and persistence of clinical symptoms would warrant further investigation such as magnetic resonance imaging, which will demonstrate the signal abnormalities of the marrow. Pathologic fracture can be present in 22% of cases.89 Due to the nonspecificity of the radiographic findings, the differential diagnoses include OS, eosinophilic granuloma, metastases of solid tumors, round cell tumors (ES, neuroblastoma, rhabdomyosrcaoma), and chronic osteomyelitis. Pathology Grossly, bone lymphomas tend to be fleshy gray-white or hemorrhagic, and the gross appearance is also related to the response of the host bone, leading to sometimes very sclerotic and bony biopsy specimens requiring decalcification. Histologically, the most common finding is a diffuse pattern of growth with tumor cells permeating between bony trabeculae and invading Haversian channels of the cortex. There is often a sclerotic or collagenized stroma, and the tumor cells can invade in cords, nests, or solid sheets. As is often evidenced in the x-ray, the host bony trabeculae can be thickened and irregular with extensive sclerosis. The most frequent type (92%)82 of primary NHL of bone is diffuse large B-cell lymphoma (DLBCL). Unlike nodal lymphomas, DLBCL of bone characteristically shows a polymorphous appearance (Figures 18 and 19). The tumor cells themselves show marked variation in size and shape, often with multilobation, and are accompanied by a mixture of small and medium-sized lymphocytes that are reactive B and T cells. The large tumor cells can have prominent nucleoli, and the cytoplasm is not usually abundant. Occasionally, the fibrotic stromal reaction elicited by the tumor cells can result in tumor cell spindling with storiform features, mimicking a sarcoma. The polymorphous nature of the infiltrate can lead to a mistaken diagnosis of osteomyelitis, which is also a radiologic differential diagnosis. Crush artifact is fairly common due to the delicate nature of the tumor cells and can be a vexing problem in pathologic diagnosis. Apart from DLBCL, other NHLs, which can occasionally present as primary neoplasms of bone, include anaplastic large cell lymphoma, lymphoblastic lymphoma, Burkitt lymphoma, and T-cell lymphomas.82,90,91 Of these, lymphoblastic lymphoma can be a diagnostic challenge due to morphological and immunohistochemical overlap with ES.90 Follicular lymphomas and small lymphocytic lymphomas such as chronic lymphocytic lymphoma generally do not present as primary destructive bone lesions.77 Immunohistochemistry and Genetics In a suspected case of NHL, a panel of markers is used to delineate the lineage and type of the neoplasm. They include leucocyte common antigen, B-cell markers (CD20), and T-cell markers (CD2, CD3, CD5, CD7), and additional markers such as CD30, epithelial membrane antigen, and Alk-1 to rule out anaplastic large cell lymphoma. In a case with a monomorphic morphology such as lymphoblastic 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 (hematoxylineosin, original magnification ⫻400).

lymphoma, the panel should include all of the lymphoma and other round cell tumor markers (CD45, CD20, CD2, CD3, CD5, CD7, TdT, vimentin, CD99, FLI-1, keratin, desmin, chromogranin). In addition, other lymphoid antigens such as CD79a and CD43 may be required, as lymphoblastic lymphomas are often negative for CD20 and positive for CD99 and FLI-1. They can also be negative for leucocyte common antigen, and in some instances show focal cytoplasmic keratin positivity, a major pitfall that can lead to an erroneous diagnosis of ES.90 In pediatric cases of small round cell tumors, TdT appears to be a sensitive and specific marker for precursor B or T lymphoblastic lymphomas and should be included in the immunopanel (Table 1). Other special stains such as ␬ and ␭ can be used to ensure monoclonality, but the results are often variable due to technical limitations. Primary T-cell lymphomas of the bone are extremely rare, and the diagnosis is established based on morphology of the polymorphous population with large tumor cells and loss of pan–T-cell antigens such as CD7. Two cases of primary adult T-cell leukemia of the bone have been reported.91 Additional studies Arch Pathol Lab Med—Vol 131, February 2007

such as flow cytometry can be used in appropriate cases. B-cell and T-cell rearrangement studies can be performed in fresh and paraffin-embedded tissues to confirm clonality in doubtful cases. Specific cytogenetic studies of primary bone lymphomas are sparse. There is a single report of a pediatric DLBCL of the spine with t(3;22)(q27;q11).92 Recently we encountered a case of an atypical Burkitt lymphoma arising in the calcaneus of an elderly male patient histologically mimicking DLBCL (Figure 20); however, cytogenetic analysis showed t(8;14)(q24.1;q32), characteristic of a Burkitt lymphoma. Treatment and Prognosis The primary modality of therapy for primary bone lymphomas is chemotherapy and radiotherapy, depending on the stage of disease and the grade and type of the lymphoma.93 The prognosis is dependent upon the stage of the disease. Patients with stages I and II have a very good prognosis compared with patients with stages III and IV.94 The 5-year overall survival ranges between 56% and 61%.82,95 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 include 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 multifocal.99 Clinical Presentation Pain and swelling are the most common clinical features, 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 eccentric with stippled calcifications denoting its chondroid nature (Figure 21). The lesion is often destructive, with expansion of bone, cortical destruction, and extraosseous extension (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 intensity on T2-weighted images.104,105

Figure 21. Plain x-ray of proximal femur showing a permeative pattern in the intertrochanteric and subtrochanteric region with endosteal scalloping across the distalmost aspect of the neoplasm. There are questionable foci of matrix calcification proximally. (Courtesy of Dr Joseph Benevenia, Department of Orthopedics, University of Medicine and Dentistry of New Jersey, Newark.)

Conclusion Primary NHL of bone is a rare but well-defined entity with radiologic and morphologic heterogeneity. The histologic and immunohistochemical similarities, ranging from benign conditions such as osteomyelitis to monotonous round cell sarcomas, should be borne in mind when assessing these neoplasms to avoid misdiagnosis and inappropriate treatment. MESENCHYMAL CHONDROSARCOMA This rare neoplasm was first described by Lichtenstein and Bernstein96 in 1959 as an unusual variant of chondrosarcoma affecting bone and soft tissues. This was followed by recognition of 9 more cases from the files of the Mayo Clinic by Dahlin and Henderson.97 Subsequently it has been established as a separate entity distinct from conventional and dedifferentiated chondrosarcoma. Incidence and Location Mesenchymal chondrosarcomas constitute less than 2% of all chondrosarcomas.98,99 The tumor most commonly af200 Arch Pathol Lab Med—Vol 131, February 2007

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 hemangiopericytic pattern with multiple vascular spaces. The cartilage component is variable and can show endochondral ossification with bone formation, coarse calcifications, or loosely arranged mature hyaline cartilage. There can be distinct demarcation or gradual blending between the 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 specimens. On the other hand, the cartilage component alone in a biopsy can lead to the erroneous diagnosis of a benign cartilaginous neoplasm. Ultrastructure The primitive mesenchymal cells show round to oval cells with little intercellular matrix, and the cartilaginous foci reveal mature cells containing glycogen, rough endoplasmic reticulum, and numerous mitochondria.106 Immunohistochemistry The chondroid foci stain for S100 protein, and the primitive mesenchymal cells are positive for CD99 (Figure 26).107 Both components are vimentin positive. It has been proposed that immunohistochemical positivity for collagen II and IIA, which are considered to be markers of chondroprogenitor cells, could be used to differentiate mesenchymal chondrosarcoma from other small round cell malignancies, such as small cell OS and ES.108 Another study109 has shown that the transcription factor Sox9, a master regulator of chondrogenesis,110,111 differentiates 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 malignancies, including small cell OS, lymphoma, and ES, by immunohistochemical analysis using a rabbit polyclonal antibody. Cytogenetics and Molecular Genetics There are a few reported cytogenetic alterations in mesenchymal chondrosarcoma, and so far no specific or recurrent translocations have been identified. Case reports of near-tetraploid and other diverse structural abnormalities,112 translocation der(13;21)(q10;q10) with loss of chromosomes 8 and 20 material and gain of chromosome 12 material in 2 cases,113 and 1 case with t(11;22)(q24;q12) similar to ES have been reported.114 Molecular genetic analyses for mutational alteration of p53 and p16 tumor suppressor genes have shown a low incidence (⬍25%) in these tumors.115,116 Biologic pathways involving PKC-␣, PDGFR-␣, and Bcl-2 expression seem to play a role in pathogenesis.117 Treatment and Prognosis Mesenchymal chondrosarcomas are primarily treated with surgery. They are aggressive lesions with a high propensity for metastases; however, some patients can have a protracted clinical course, so the behavior can be unpredictable.118 The overall 10-year survival rate is approximately 25%.99 Tumors of the jawbones tend to have a more indolent course.119 Negative correlation between survival times and proliferation rates using Ki-67 in 10 patients has been recently reported.120 Distant metastases which can occur after many years can involve other bones, lung, etc.99,118 Hence long-term follow-up of these patients is advocated. Arch Pathol Lab Med—Vol 131, February 2007

SMALL CELL OSTEOSARCOMA This tumor is discussed here in the context of the differential diagnosis of the above-mentioned small round cell tumors of the bone. This is a distinctive microscopic variant of OS, originally described in 1979, as a tumor simulating ES.121 This rare tumor accounts for about 1% to 1.5% of OSs and has similar age, sex, and skeletal distribution as conventional OS.122 Radiologically the tumor can resemble ES or conventional OS. The majority of the lesions show a mixed lytic and blastic pattern, but they can be purely lytic or permeative; they usually involve metaphysis and may extend into epiphysis. A soft tissue component is present in most of the cases.123–126 Three histologic patterns have been described: ES-like, lymphomalike, and small spindle cell.123 The cells can be round and uniform, similar to ES, or show variation in size. Nucleoli can be inconspicuous to prominent. The chromatin can be fine to hyperchromatic. The nuclei can be very small to medium (6.7–15 ␮m), thus showing a spectrum of sizes between ES and large cell lymphoma.125 Hemangiopericytic pattern, myxoid change, cordlike arrangement, and epithelioid features can be seen. The presence of malignant cartilage in some tumors can simulate mesenchymal chondrosarcoma.125 The presence of osteiod is a prerequisite for diagnosis. The new osteoid matrix is usually lacelike and can be difficult to find in small biopsies (Figure 27). The presence of mineralized matrix in imaging studies supports the diagnosis of OS and is helpful in differentiating other small blue cell tumors of bone. ES may show a blastic component in the medullary cavity, but production of matrix outside the bone is seen only in OS.123,127,128 Cytoplasmic glycogen can be seen in both neoplasms.123 Immunohistochemical features do not distinguish small cell OS from sarcomas, as CD99 can be posSmall 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 translocations such as t(11;22) have not been reported in small cell OS. Lymphomas can be distinguished using lymphoma-specific markers.130 The treatment of small cell OS is similar to conventional OS, which includes neoadjuvant chemotherapy followed by surgery, usually a limb salvage procedure. According to some authors, when there is questionable matrix formation in a small round cell tumor, it is best to err on the diagnosis of ES rather than small cell OS.125 OTHER TUMORS Rhabdomyosarcoma and desmoplastic round cell tumors are primarily soft tissue neoplasms; rare primary bone tumors are cited in the literature as case reports.131,132 Neuroblastomas can present as metastases in the bone and can be distinguished from other primary round cell tumors of bone with immunohistochemical positivity for neuron-specific enolase, synaptophysin, and chromogranin and absent staining for CD99,133 lymphoid markers, desmin, myogenin, and MyoD1. 202 Arch Pathol Lab Med—Vol 131, February 2007

SUMMARY Small round cell tumors of bone are a heterogeneous group of neoplasms with overlapping clinical, radiologic, morphologic, and immunohistochemical features. Even though questions remain regarding cell of origin and histogenesis, cytogenetic and molecular genetic studies have enhanced our understanding in bringing together some of these tumors into single entities. In addition, cDNA microarray analysis and the availability of techniques to unravel the various signaling pathways will open many doors toward targeted chemotherapy in these aggressive, highly lethal tumors. The author thanks Seena Aisner, MD, Department of Pathology, UMDNJ-Newark, for critical review of the manuscript. References 1. Stout AP. A tumor of the ulnar nerve. Proc N Y Pathol Soc. 1918;12:2–12. 2. Ewing J. Diffuse endothelioma of bone. Proc N Y Pathol Soc. 1921;21:17– 24. 3. Cohn I. Epithelial neoplasms of peripheral and cranial nerves. Report of three cases: review of the literature. Arch Surg. 1928;17:117–160. 4. Angerwall L, Enzinger FM. Extraskeletal neoplasm resembling Ewing’s sarcoma. Cancer. 1975;36:240–251.

Small Round Cell Tumors of Bone—Hameed

5. Jaffe R, Santamaria M, Yunis EJ, et al. The neuroectodermal tumor of bone. Am J Surg Pathol. 1990;8:885–898. 6. Ushigome U, Machinami R, Sorensen PH. Ewing sarcoma/primitive neuroectodermal tumor (PNET). In: Fletcher CDM, Unni KK, Mertens F, eds. Pathology and Genetics of Tumours of Soft Tissue and Bone. Lyon, France: IARC Press; 2002:298–300. World Health Organization Classification of Tumours. 7. Turc-Carel C, Philip I, Berger MP, Phili T, Lenoir GM. Chromosomal translocations in Ewing’s sarcoma. N Engl J Med. 1983;309:497–498. 8. Whang-Peng J, Triche TJ, Knutsen T, Miser J, Douglass EC, Israel MA. Chromosome translocation in peripheral neuroepithelioma. N Engl J Med. 1984;311: 584–585. 9. Gurney JG, Severson RK, Davis S, Robison LL. Incidence of cancer in children in the United States: sex-, race-, and 1-year age-specific rates by histologic type. Cancer. 1995;75:2186–2195. 10. Carvajal R, Meyers P. Ewing’s sarcoma and primitive neuroectodermal family of tumors. Hematol Oncol Clin North Am. 2005;19:501–525. 11. Fizazi K, Dohollou N, Blay JY, et al. Ewing’s family of tumors in adults: multivariate analysis of survival and long-term results of multimodality therapy in 182 patients. J Clin Oncol. 1998;16:3736–3743. 12. Levine RG, Bono CM, Hameed M, et al. Ewing sarcoma in an octogenarian: a case report. J Bone J Surg Am. 2002;84:445–448. 13. Dorfman H, Czerniak B. Bone cancers. Cancer. 1995;75(suppl):203–210. 14. Himelstein BP, Dormans JP. Malignant bone tumors of childhood. Pediatr Clin North Am. 1996;43:967–983. 15. Widhe B, Widhe T. Initial symptoms and clinical features in osteosarcoma and Ewing sarcoma. J Bone Joint Surg Am. 2000;82:667–674. 16. Wagner LM, Neel MD, Pappo AS, et al. Fractures in pediatric Ewing sarcoma. J Pediatric Hematol Oncol. 2001;23:568–571. 17. Hoffer FA. Primary skeletal neoplasms: osteosarcoma and Ewing sarcoma. Top Magn Reson Imaging. 2002;13:231–240. 18. Nascimento AG, Unni K, Pritchard D, Cooper KL, Dahlin DC. A clinicopathological study of 20 cases of large-cell (atypical) Ewing’s sarcoma of bone. Am J Surg Pathol. 1980;4:29–36. 19. Julia BA, Fidler ME, James NR, et al. Adamantinoma-like Ewing’s sarcoma: genomic confirmation, phenotypic drift. Am J Surg Pathol. 1999;23:159–165. 20. Hauben E, van den Broek LC, Van Marck E, Hogendoorn PC. Adamantinoma-like Ewing’s sarcoma and Ewing’s-like adamantinoma: the t(11;22),t(12;22) status. J Pathol. 2001;195:218–221. 21. Folpe AL, Goldblum JR, Rubin BP, et al. Morphologic and immunophenotypic diversity in Ewing family tumors. Am J Surg Pathol. 2005;29:1025–1033. 22. Suh CH, Ordonez NG, Hicks J, Mackay B. Ultrastructure of the Ewing’s sarcoma family of tumors. Ultrastruct Pathol. 2002;26:67–76. 23. Franchi A, Pasquinelli G, Cenacchi G, et al. Immunohistochemical and ultrastructural investigation of neural differentiation in Ewing sarcoma/PNET of bone and soft tissues. Ultrastruct Pathol. 2001;25:219–225. 24. Ambros IM, Ambros PF, Strehl S, Kovar H, Gadner H, Salzer-Kuntschik M. MIC2 is a specific marker for Ewing’s sarcoma and peripheral primitive neuroectodermal tumors: evidence for a common histogenesis of Ewing’s sarcoma and peripheral primitive neuroectodermal tumors from MIC2 expression and specific chromosome aberration. Cancer. 1991;67:1886–1893. 25. Bantig GS, Pym B, Darling SM, Goodfellow PN. The MIC2 gene product: epitope mapping and structural prediction analysis define an integral membrane protein. Mol Immunol. 1989;26:181–188. 26. Darling SM, Goodfello PJ, Pym B, Banting GS, Pritchard C, Goodfellow PN. Molecular genetics of MIC2: a gene shared by the human X and Y chromosomes. Cold Spring Harb Symp Quant Biol. 1986;51:205–212. 27. Folpe AL, McKenney JK, Bridge JA, Weiss SW. Sclerosing rhabdomyosarcoma in adults: report of four cases of a hyalinizing, matrix-rich variant of rhabdomyosarcoma that may be confused with osteosarcoma, chondrosarcoma, or angiosarcoma. Am J Surg Pathol. 2002;26:1175–1183. 28. 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. 29. Ozdemirli M, Fanburg-Smith JC, Hartmann DP, Azumi N, Miettinen M. Differentiating lymphoblastic lymphoma and Ewing’s sarcoma:lymphocyte markers and gene arrangement. Mod Pathol. 2001;14:1175–1182. 30. Granter SR, Renshaw AA, Fletcher CD, Bhan AK, Rosenberg AE. CD99 reactivity in mesenchymal chondrosarcoma. Hum Pathol. 1996;27:1273–1276. 31. Llombart-Bosch A, Navarro S. Immunohistochemical detection of EWS and FLI-1 proteins in Ewing sarcoma and primitive neuroectodermal tumors:comparative analysis with CD99 (MIC-2) expression. Appl Immmunohistochem Mol Morphol. 2001;9:255–260. 32. Fellinger EJ, Garin-Chesa P, Glasser DB, Huvos AG, Rettig WJ. Comparison of cell surface antigen HBA71 (p30/32MIC2), neuron-specific enolase and vimentin in the immunohistochemical study of Ewing’s sarcoma of bone. Am J Surg Pathol. 1992;16:746–755. 33. Amann G, Zoubek A, Salzer-Kuntschik M, Windhager R, Kovar H. Relation of neurological marker expression and EWS gene fusion types in MIC2/CD99positive tumors of the Ewing Family. Hum Pathol. 1999;30:1058–1064. 34. Gu M, Antonesu CR, Guiter G, Huvos AG, Ladanyi M, Zakowski MF. Cytokeratin immunoreactivity in Ewing’s sarcoma: prevalence in 50 cases confirmed by molecular diagnostic studies. Am J Surg Pathol. 2000;24:410–416. 35. Aurias A, Rimbaut C, Buffe C, Dubousset J, Mazabraud A. Chromosomal translocations in Ewing’s sarcoma. N Engl J Med. 1983;309:496–497. 36. Miozzo M, Sozzi G, Calderone C, et al. t(11;22) in three cases of peripheral neuroepithelioma. Genes Chromosomes Cancer. 1990;2:163–165.

Arch Pathol Lab Med—Vol 131, February 2007

37. Whang-Peng J, Triche TJ, Knutsen T, Miser J, Douglass EC, Israel MA. Chromosome translocation in peripheral neuroepithelioma. N Engl J Med. 1984;311: 584–585. 38. May WA, Gishizky ML, Lessnik SL, et al. Ewing sarcoma 11;22 translocation produces a chimeric transcription factor that requires the DNA-binding domain encoded by FLI1 for transformation. Proc Natl Acad Sci U S A. 1993;90: 5752–5756. 39. Sorensen PH, Lessnick SL, Lopez-Terrada D, et al. A second Ewing’s sarcoma translocation, t(21;22), fuses the EWS gene to another ETS-family transcription factor, ERG. Nat Genet. 1994;6:146–151. 40. Jeon IS, Davis JN, Braun BS, et al. A variant Ewing’s sarcoma translocation (7;22) fuses the EWS gene to the ETS gene ETV1. Oncogene. 1995;10:1229– 1234. 41. Kaneko Y, Yoshida K, Handa M, et al. Fusion of an ETS-family gene, E1AF, to EWS by t(7;22)(q12;q12) chromosome translocation in an undifferentiated sarcoma of infancy. Genes Chromosomes Cancer. 1996;15:115–121. 42. Urano F, Umezawa A, Hong W, et al. A novel chimera gene between EWS and E1A-F, encoding the adenovirus E1A enhancer-binding protein, in extraosseous Ewing’s sarcoma. Biochem Biophys Res Commun. 1996;219:608–612. 43. Peter M, Couturier J, Pacquement H, et al. A new member of the ETS family fused to EWS in Ewing tumors. Oncogene. 1997;14:1159–1164. 44. Ohno T, Ouchida M, Lee L, et al. The EWS gene, involved in Ewing family of tumors, malignant melanoma of soft tumors and desmoplastic small round cell tumors, codes for an RNA binding protein with novel regulatory domains. Oncogene. 1994;9:3087–3097. 45. Rossow KL, Janknecht R. The Ewing’s sarcoma gene product functions as a transcriptional activator. Cancer Res. 2001;61:2690–2695. 46. Aman P, Panagopoulos I, Lassen C, et al. Expression patterns of the human sarcoma-associated genes FUS and EWS and the genomic structure of FUS. Genomics. 1996;37:1–8. 47. Sementchenko VI, Watson DK. Ets target genes: past, present and future. Oncogene. 2000;19:6533–6548. 48. Folpe A, Chand EM, Goldblum JR, et al. Expression of Fli-1, a nuclear transcription factor, distinguishes vascular neoplasms from potential mimics. Am J Surg Pathol. 2001;25:1061–1066. 49. Kovar H. Context matters: the hen or egg problem in Ewing’s sarcoma. Semin Cancer Biol. 2005;15:189–196. 50. Amandine HLT, Yaacov BD. The role of Fli-1 in normal cell function and malignant transformation. Oncogene. 2000;19:6482–6489. 51. Zucman J, Melot T, Desmaze C, et al. Combinatorial generation of variable fusion proteins in the Ewing family of tumours. EMBO J. 1993;12:4481–4487. 52. Kovar H. Ewing tumor biology: perspectives for innovative treatment approaches. Adv Exp Med Biol. 2003;27–37. 53. Peter M, Gilbert E, Delattre O. A multiplex real-time PCR assay for the detection of gene fusions observed in solid tumors. Lab Invest. 2001;81:905–912. 54. Xiang Q, Long J, Brandon S, Rhett PK, Syed J, Ricardo L. Molecular diagnosis of Ewing’s sarcoma/primitive neuroectodermal tumor in formalin-fixed paraffin-embedded tissues by RT-PCR and fluorescence in situ hybridization. Diagn Mol Pathol. 14:23–28. 55. Udayakumar AM, Sundareshan TS, Mallana GT, et al. Cytogenetic characterization of Ewing tumors using fine needle aspiration samples: a 10 year experience and review of literature. Cancer Genet Cytogenet. 2001;127:42–48. 56. Udayakumar AM, Sundareshan TS. Cytogenetic characterization of Ewing tumors: further update on 20 cases [letter]. Cancer Genet Cytogenet. 2002;133: 102–103. 57. Rodiguez-Galindo C. Pharmacological management of Ewing sarcoma family of tumours. Expert Opin Pharmacother. 2004;17:329a. 58. Carpentieri DF, Qualman SJ, Bowen J, Krausz T, Marchevsky A, Dickman PS. Protocol for the examination of the specimens from pediatric and adult patients with osseous and extraosseous Ewing sarcoma family of tumors, including peripheral primitive neuroectodermal tumor and Ewing sarcoma. Arch Pathol Lab Med. 2005;129:866–873. 59. Schoedel K, Dickman PS, Krailo M, Perlman EJ, Miser J, Grier H. Histologic response to chemotherapy and prognosis in osseous Ewing’s sarcoma/primitive neuroectodermal tumor (ES/PNET) [abstract]. Int Surg Pathol. 1995;2(suppl):443. 60. Cotterill SJ, Ahrens S, Paulussen M, et al. Prognostic factors in Ewing’s tumor of bone: analysis of 975 patients from the European intergroup cooperative Ewing’s sarcoma study group. J Clin Oncol. 2000;18:3108–3114. 61. Delepine N, Delepine G, Cornille H, Voisin MC, Brun B, Desbois JC. Prognostic factors in patients with localized Ewing’s sarcoma: the effect on survival of actual received drug dose intensity and of histologic response to induction therapy. J Chemother. 1997;9:352–363. 62. Picci P, Rougraff BT, Bacci G, et al. Prognostic significance of histopathologic response to chemotherapy in nonmetastatic Ewing’s sarcoma of extremities. J Clin Oncol. 1993;11:1763–1769. 63. Burchill SA. Ewing’s sarcoma: diagnostic, prognostic and therapeutic implications of molecular abnormalities. J Clin Pathol. 2003;56:96–102. 64. Amiel A, Ohali A, Fejgin M, et al. Molecular cytogenetic parameters in Ewing sarcoma. Cancer Genet Cytogenet. 2003;140:107–112. 65. Zoubek A, Dockhorn-Dwarniczak B, Delatire O, et al. Does expression of different EWS transcripts define clinically distinct risk groups of Ewing tumor patients? J Clin Oncol. 1996;14:245–251. 66. Lin PP, Brody RJ, Hamelin AC, et al. Differential transactivation by alternative EWS-FLI1 fusion proteins correlates with clinical heterogeneity in Ewing’s sarcoma. Cancer Res. 1999;59:1428–1455.

Small Round Cell Tumors of Bone—Hameed

203

67. Fagnou C, Michon J, Peter M, et al. Presence of tumor cells in bone marrow but not in blood is associated with adverse prognosis in patients with Ewing’s tumor: Societe Francaise d’Oncologie Pediatrique. J Clin Oncol. 1998;16:1707– 1711. 68. Zoubek A, Ladenstein R, Windhager R, et al. Predictive potential of testing for bone marrow involvement in Ewing’s tumor patients by RT-PCR: a preliminary evaluation. Int J Cancer. 1998;79:56–60. 69. West DC, Grier HE, Swallow MM, et al. Detection of circulating tumor cells in patients with Ewing’s sarcoma and primitive peripheral neuroectodermal tumor. J Clin Oncol. 1997;15:583–588. 70. Schleiermacher G, Peter M, Oberlin O, et al. Increased risk of systemic relapses associated with bone marrow micrometastases and circulating tumor cells in localized Ewing’s tumor. J Clin Oncol. 2003;21:85–91. 71. Kennedy JG, Frelinghuysen P, Hoang BH. Ewing sarcoma: current concepts in diagnosis and treatment. Curr Opin Pediatr. 2003;15:53–57. 72. Kovar J, Aryee D, Zoubek A. The Ewing family of tumors and the search for the Achilles’ heel. Curr Opin Oncol. 1999;11:275–292. 73. Prieur A, Tirode F, Cohen P, et al. EWS/FLI-1 silencing and gene profiling of Ewing cells reveal downstream oncogenic pathways and a crucial role for repression of insulin-like growth factor binding protein 3. Mol Cell Biol. 2004; 24:7275–7283. 74. Ouchida M, Ohno T, Fujimara Y, et al. Loss of tumorigenecity of Ewing’s sarcoma cells expressing antisense RNA to EWS-fusion transcripts. Oncogene. 1995;11:1049–1054. 75. Parker F, Jackson H Jr. Primary reticulum cell sarcoma of bone. Surg Gynecol Obstet. 1939;68:45–53. 76. Oberling C. Les reticulosarcomes et les reticuloendotheliosarcomes de la moelle osseuse (sarcoma d’Ewing). Bull Assoc Fr Etude Cancer (Paris). 1928;17: 259–296. 77. Unni KK, Hogendoorn PCW. Malignant lymphoma. In: Fletcher CDM, Unni KK, Mertens F, eds. Pathology and Genetics of Tumours of Soft Tissue and Bone. Lyon, France: IARC Press; 2002. World Health Organization Classification of Tumours. 78. Freeman C, Berg JW, Cutler SJ. Occurrence and prognosis of extranodal lymphomas. Cancer. 1972;28:252–260. 79. Rodman D, Raymond AK, Phillips WC. Case report 201: primary lymphoma of bone (PLB)—left fibula. Skeletal Radiol. 1982;8:235–237. 80. Rosenberg SA, Diamond HD, Jasowitz B, Craver LF. Lymphosarcoma: a review of 1269 cases. Medicine. 1961;40:31–81. 81. Rudder RA, Ross ME, De Lellis RA. Primary extranodal lymphoma: response to treatment and factors influencing prognosis. Cancer. 1978;42:406–416. 82. Heyning FH, Hogendoorn PC, Kramer MN, et al. Primary non-Hodgkin’s lymphoma of bone: a clinicopathological investigation of 60 cases. Leukemia. 1999;13:2094–2098. 83. Barr J, Burkes RL, Bell R, Blackstein M, Fernandes B, Langer F. Primary non-Hodgkin’s lymphoma of bone. Cancer. 1994;73:1194–1199. 84. Dubey P, Chul SH, Besa P, et al. Localized primary malignant lymphoma of bone. Int J Radiat Oncol Biol Phys. 1997;37:1087–1093. 85. Moses AM, Spencer H. Hypercalcemia in patients with malignant lymphoma. Ann Int Med. 1963;59:531–536. 86. Dorfman H, Cherniak B. Bone Tumors. St Louis, Mo: Mosby; 1998. 87. Braunstein EM, White SJ. Non-Hodgkin’s lymphoma of bone. Radiology. 1980;135:59–63. 88. Clayton F, Butler JJ, Ayala AG, Ro JY, Zornoza J. Non-Hodgkin’s lymphoma in bone. Cancer. 1987;60:2494–2501. 89. Mulligan ME, McRae GA, Murphy MD. Imaging features of primary lymphoma of bone. Amer J Radiol. 1999;173:1691–1697. 90. Ozdemirli M, Fanburg-Smith J, Hartmann DP, et al. Precursor B-lymphoblastic lymphoma presenting as a solitary bone tumor and mimicking Ewing’s sarcoma: a report of four cases and review of the literature. Am J Surg Pathol. 1998;22:795–804. 91. Takemoto S, Matsuoka M, Sakata K, et al. Primary adult T cell leukemia of bone: two patients with primary bone lesion showing monoclonal integration of HTLV-1 proviral DNA. Leukemia. 1996;10:333–337. 92. Antillon F, Behm FG, Raimondi SC, Kaste SC, Sandland JT, Pappo AS. Pediatric primary diffuse large cell lymphoma of bone with t(3;22)(q27;q11). J Pediatr Hematol Oncol. 1998;20:552–555. 93. Misgeld E, Wehmeier A, Kromeke O, Gattermann N. Primary non-Hodgkin’s lymphoma of bone: three cases and a short review of the literature. Ann Hematol. 2003;82:440–443. 94. Ostowski M, Unni KK, Banks PM, et al. Malignant lymphoma of bone. Cancer. 1986;58:2646–2655. 95. Mendenhall NP, Jones JJ, Kramer BS, et al. The management of primary lymphoma of bone. Radiother Oncol. 1987;8:137–145. 96. Lichtenstein L, Bernstein D. Unusual benign and malignant chondroid tumors of bone. Cancer. 1959;12:1142–1157. 97. Dahlin DC, Henderson ED. Mesenchymal chondrosarcoma: further observations on a new entity. Cancer. 1962;15:410–417. 98. Bertoni F, Picci P, Bacchini P. Mesenchymal chondrosarcoma of bone and soft tissues. Cancer. 1983;52:533–541. 99. Nakashima Y, Unni KK, Shives TC, Sweet RG, Dahlin DC. Mesenchymal chondrosarcoma of bone and soft tissue: a review of 111 cases. Cancer. 1986; 57:2444–2453. 100. Dabska M, Huvos AG. Mesenchymal chondrosarcoma in the young: a clinicopathological study of 19 patients with explanation of histogenesis. Virchows Arch Pathol Histopathol. 1983;399:89–104.

204 Arch Pathol Lab Med—Vol 131, February 2007

101. Salvador AH, Beabout JW, Dahin DC. Mesenchymal chondrosarcoma: observation on 30 new cases. Cancer. 1971;28:605–616. 102. Harwood AR, Krajbich JI, Fornasier VL. Mesenchymal chondrosarcoma: a report of 17 cases. Clin Orthop Relat Res. 1981;158:144–148. 103. Zura RD, Minasi JS, Kahler DM. Tumor-induced osteomalacia and symptomatic looser zones secondary to mesenchymal chondrosarcoma. J Surg Oncol. 1999;71:58–62. 104. Shinaver CN, Mafee MF, Choi KH. MRI of mesenchymal chondrosarcoma of the orbit: case report and review of literature. Neuroradiology. 1997;39:296– 301. 105. Ariyoshi Y, Shimahara D. Mesenchymal chondrosarcoma of the maxilla: report of a case. J Oral Maxillofac Surg. 1999;57:733–737. 106. Steiner GC, Mirra JM, Bullough PG. Mesenchymal chondrosarcoma: a study of the ultrastucture. Cancer. 1973;32:926–939. 107. Granter SR, Renshaw AA, Fletcher CD, Bhan AK, Rosenberg AE. CD99 reactivity in mesenchymal chondrosarcoma. Hum Pathol. 1996;27:1273–1276. 108. Aigner T, Loos S, Muller S, Sandell LJ, Unni KK, Kirshner T. Cell differentiation and matrix gene expression in mesenchymal chondrosarcoma. Am J Pathol. 2000;156:1327–1335. 109. Wehrli BM, Huang W, De Crombrugghe B, Ayala AG, Czerniak B. Sox9, a master regulator of chondrogenesis, distinguishes mesenchymal chondrosarcoma from other small round cell tumors. Hum Pathol. 2003;34:263–269. 110. Ng LJ, Wheatley S, Muscat GEO, et al. Sox9 binds DNA, activates transcription and co express with type II collagen during chondrogenesis in mouse. Dev Biol. 1997;183:108–121. 111. Bi W, Deng JM, Zhang Z, et al. Sox9 is required for cartilage formation. Nat Genet. 1999;22:85–89. 112. Dobin SM, Donner LR, Speights VO Jr. Mesenhymal chondrosarcoma: a cytogeentic, immunohistochemical and ultrastructural study. Cancer Genet Cytogenet. 1005;83:56–60. 113. Naumann S, Krallman PA, Unni KK, Fidler ME, Neff JR, Bridge JA. Translocation der(13;21)(q10;q10) in skeletal and extraskeletal mesenchymal chondrosarcoma. Mod Pathol. 2002;15:572–576. 114. Sainati L, Scapinello A, Montaldi A, et al. Mesenchymal chondrosarcoma of a child with reciprocal translocation (11;22)(q24;q12), Cancer Genet Cytogenet. 1993;71:144–147. 115. Park YK, Park HR, Chi SG, et al. Overexpression of p53 and rare genetic mutation in mesenchymal chondrosarcoma. Oncol Rep. 2000;7:1041–1047. 116. Bae DK, Park YK, Chi SG, Lee CW, Unni KK. Mutational alterations of the p16CDKN2A tumor suppressor gene have low incidence in mesenchymal chondrosarcoma. Oncol Res. 2000;12:5–10. 117. Brown RE, Boyle JL. Mesenchymal chondrosarcoma: molecular characterization by a proteomic approach with morphogenic and therapeutic implications. Ann Clin Lab Sci. 2003;33:131–141. 118. Dowling EA. Mesenchymal chondrosarcoma. J Bone Joint Surg. 1964;46A:747–754. 119. Vencio EF, Reeve CM, Unni KK, Nascimento AG. Mesenchymal chondrosarcoma of the jaw bones: clinicopathologic study of 19 cases. Cancer. 1998; 82:2350–2355. 120. Nussbeck W, Neureiter D, Soder S, Inwards C, Aigner T. Mesenchymal chondrosarcoma: an immunohistochemical study of 10 cases examining prognostic significance of proliferative activity and cellular differentiation. Pathology. 2004;36:230–233. 121. Sim FH, Unni KK, Beabout JW, Dahlin DC. Osteosarcoma with small cells simulating Ewing’s tumor. J Bone Joint Surg Am. 1979;61:207–215. 122. Dahlin DC, Unni KK. General Aspects and Data on 11087 Cases. 5th ed. Philadelphia, Pa: Lippincott-Raven, 1996. 123. Ayala AG, Ro JY, Raymond K, et al. Small cell osteosarcoma: a clinicopathologic study of 27 cases. Cancer. 1989;64:2162–2173. 124. Ayala AG, Ro JY, Papadopoulos NK, Raymond KA, Edeiken J. Osteosarcoma in Adolescents and Young Adults. Boston, Mass: Kluwer Academic Publishers; 1993. 125. Nakajima H, Sim FH, Bond JR, Unni KK. Small cell osteosarcoma of bone: review of 72 cases. Cancer. 1997;79:2095–2106. 126. Park SH, Kim I. Small cell osteogenic sarcoma of the ribs: cytological, immunohistochemical and ulttrastructural study with literature review. Ultra Pathol. 1999;23:133–140. 127. Bertoni F, Present D, Bacchini P, Pignatti G, Picci P, Campanacci M. The Instituto Rizzoli experience with small cell osteosarcoma. Cancer. 1989;64:2591– 2599. 128. Martin SE, Dwyer A, Kissane TJ. Small cell osteosarcoma. Cancer. 1982; 50:990–996. 129. Devoe K, Weidner N. Immunohistochemistry of small round cell tumors. Semin Diagn Pathol. 2000;17:216–224. 130. Devaney K, Vinh TN, Sweet DE. Small cell osteosarcoma: an immunohistochemical study with differential diagnostic considerations. Hum Pathol. 1993;24:1211–1225. 131. Thomas F, Lipton J, Barbera C, Vigorita V, Bryk E. Primary rhabdomyosarcoma of the humerus: a case report and review of the literature. J Bone Joint Surg Am. 2002;84-A:813–817. 132. Murphy A, Stallings RL, Howard J, et al. Primary desmoplastic small round cell tumor of bone: report of a case with cytogenetic confirmation. Cancer Genet Cytogenet. 2005;156:167–171. 133. Pappo AS, Douglass EC, Meyer WH, et al. Use of HBA-71 and anti-beta microglobulin to distinguish peripheral neuroepithelioma from neuroblastoma. Hum Pathol. 1993;24:880–885.

Small Round Cell Tumors of Bone—Hameed

Suggest Documents