Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-m9kch Total loading time: 0 Render date: 2024-04-30T13:34:09.283Z Has data issue: false hasContentIssue false

Chap 31 - SMALL ROUND CELL TUMORS

Published online by Cambridge University Press:  01 March 2011

Markku Miettinen
Affiliation:
Armed Forces Institute of Pathology, Washington DC
Get access

Summary

Small round blue cell tumor is the name given to a group of highly malignant neoplasms that occur mostly in the pediatric age group. Although this term could also apply to some adult neoplasms, notably small cell carcinoma of the lung, the term has become widely associated with childhood cancer. The name is derived from the primitive, highly cellular nature of these lesions, which typically present a vast sea of dark-blue nuclei on hematoxylin-based stains. Cytoplasmic abundance roughly correlates with cellular differentiation, which is often modest. Although cytoplasmic landmarks allow for cell type identification in some cases, ancillary techniques, such as immunohistochemistry, electron microscopy, and genetics, have become important in the diagnosis of these tumors.

Although childhood cancer constitutes a relatively small percentage of all malignancies, it causes a large percentage of pediatric mortality, being second only to accidents as a cause of death in industrialized nations. Pediatric cancer also differs from adult cancer in the great percentage of cases treated on multi-institutional protocols. Despite success in treatment, more than 100,000 person-years are lost each year to this group of cancers.

TYPES OF ROUND CELL TUMORS AND THEIR DIFFERENTIAL DIAGNOSES

Leukemias and brain tumors constitute most childhood cancers. Although the former can present as solid masses, these lesions do not typically enter into the differential diagnosis of small round cell tumors. Table 31.1 lists the most common lesions in the latter group. Of these tumors, neuroblastoma, rhabdomyosarcoma, lymphoma, and Wilms' tumor are most common.

Type
Chapter
Information
Modern Soft Tissue Pathology
Tumors and Non-Neoplastic Conditions
, pp. 896 - 929
Publisher: Cambridge University Press
Print publication year: 2010

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Triche, TJ, Askin, FB. Neuroblastoma and the differential diagnosis of small-, round-, blue-cell tumors. Hum Pathol 1983;14:569.Google ScholarPubMed
Donaldson, SS. Lessons from our children. Int J Radiat Oncol Biol Phys 1993;26:739.CrossRefGoogle ScholarPubMed
Ross, JA, Severson, RK, Pollock, BH, Robison, LL. Childhood cancer in the United States – A geographical analysis of cases from the Pediatric Cooperative Clinical Trials Groups. Cancer 1996;77:201.3.0.CO;2-7>CrossRefGoogle ScholarPubMed
Olshan, AF, Breslow, NE, Daling, JR. Wilms' tumor and paternal occupation. Cancer Res 1990;50:3212.Google ScholarPubMed
Grundy, P, Wilson, B, Telzerow, P. Uniparental disomy occurs infrequently in Wilms tumor patients. Am J Hum Genet 1994;54:282.Google ScholarPubMed
Koufos, A, Hansen, MF, Copeland, NG. Loss of heterozygosity in three embryonal tumours suggests a common pathogenetic mechanism. Nature 1985;316: 330.CrossRefGoogle ScholarPubMed
Heyn, R, Haeberlen, V, Newton, WA. Second malignant neoplasms in children treated for rhabdomyosarcoma. J Clin Oncol 1993;11:262.CrossRefGoogle ScholarPubMed
May, WA, Lessnick, SL, Braun, BS. The Ewing's sarcoma EWS/FLI-1 fusion gene encodes a more potent transcriptional activator and is a more powerful transforming gene than FLI-1. Mol Cell Biol 1993;13:7393.CrossRefGoogle ScholarPubMed
Whang-Peng, J, Triche, TJ, Knutsen, T. Cytogenetic characterization of selected small round cell tumors of childhood. Cancer Genet Cytogenet 1986;21:185.CrossRefGoogle ScholarPubMed
Oda, H, Imai, Y, Nakatsuru, Y. Somatic mutations of the APC gene in sporadic hepatoblastomas. Cancer Res 1996;56:3320.Google ScholarPubMed
Raffel, C, Jenkins, RB, Frederick, L. Sporadic medulloblastomas contain PTCH mutations. Cancer Res 1997;57:842.Google ScholarPubMed
Felix, CA, Chavez Kappel, C, Mitsudomi, T. Frequency and diversity of p53 mutations in childhood rhabdomyosarcoma. Cancer Res 1992;52:2243.Google ScholarPubMed
Knudson, AG. Antioncogenes and human cancer. Proc Natl Acad Sci U S A 1993;90:10914.CrossRefGoogle ScholarPubMed
Scrable, H, Witte, D, Shimada, H. Molecular differential pathology in rhabdomyosarcoma. Genes Chromosomes Cancer 1989;1:23.CrossRefGoogle ScholarPubMed
Hoovers, JM, Kalikin, LM, Johnson, . Multiple genetic loci within 11p15 defined by Beckwith-Wiedemann syndrome rearrangement breakpoints and subchromosomal transferable fragments. Proc Natl Acad Sci USA 1995;92:12456.Google ScholarPubMed
Jones, PA, Laird, PW. Cancer epigenetics comes of age. Nature Genet 1999;21:163.CrossRefGoogle ScholarPubMed
Baylin, SB. Tying it all together: epigenetics, genetics, cell cycle, and cancer [see comments]. Science 1997;277:1948.CrossRefGoogle Scholar
Chen, B, Dias, P, Jenkins, JJ. Methylation alterations of the MyoD1 upstream region are predictive of subclassification of human rhabdomyosarcomas. Am J Pathol 1998;152:1071.Google ScholarPubMed
Dehner, LP. Primitive neuroectodermal tumor and Ewing's sarcoma. Am J Surg Pathol 1993;17:1.CrossRefGoogle ScholarPubMed
Yunis, EJ. Ewing's sarcoma and related small round cell neoplasms in children. Am J Surg Pathol 1986;10(Supp.1):54.Google ScholarPubMed
Angervall, L, Enzinger, FM. Extraskeletal neoplasm resembling Ewing's sarcoma. Cancer 1975;36:240.3.0.CO;2-H>CrossRefGoogle ScholarPubMed
Tefft, M, Vawter, GF, Mitus, A. Paravertebral “round cell” tumors in children. Radiology 1969;92:1501.CrossRefGoogle ScholarPubMed
Schmidt, D, Mackay, B, Ayala, AG. Ewing's sarcoma with neuroblastoma-like features. Ultrastruct Pathol 1982;3:143.CrossRefGoogle ScholarPubMed
Mierau, GW. Extraskeletal Ewing's sarcoma (peripheral neuroepithelioma). Ultrastruct Pathol 1985;9:91.CrossRefGoogle Scholar
Seemayer, TA, Thelmo, WL, Bolande, RP. Peripheral neuroectodermal tumors. Perspect Pediatr Pathol 1975;2:151.Google ScholarPubMed
Askin, FB, Rosai, J, Sibley, RK. Malignant small cell tumor of the thoracopulmonary region in childhood: a distinctive clinicopathologic entity of uncertain histogenesis. Cancer 1979;43:2438.3.0.CO;2-9>CrossRefGoogle ScholarPubMed
Jaffe, R, Santamaria, M, Yunis, EJ. The neuroectodermal tumor of bone. Am J Surg Pathol 1984;8:885.CrossRefGoogle ScholarPubMed
Turc-Carel, C, Philip, I, Berger, MP. Chromosome study of Ewing's sarcoma (ES) cell lines. Consistency of a reciprocal translocation t(11;22)(q24;q12). Cancer Genet Cytogenet 1984;12:1.CrossRefGoogle Scholar
Whang-Peng, J, Triche, TJ, Knutsen, T. Chromosome translocation in peripheral neuroepithelioma. N Engl J Med 1984;311:584.CrossRefGoogle ScholarPubMed
Ladanyi, M, Lewis, R, Garin-Chesa, P. EWS rearrangement in Ewing's sarcoma and peripheral neuroectodermal tumor. Molecular detection and correlation with cytogenetic analysis and MIC2 expression. Diagn Mol Pathol 1993;2:141.Google ScholarPubMed
Pellin, A, Boix, J, Blesa, JR. EWS/FLI-1 rearrangement in small round cell sarcomas of bone and soft tissue detected by reverse transcriptase polymerase chain reaction amplification. Eur J Cancer [A] 1994;30A:827.CrossRefGoogle ScholarPubMed
Cavazzana, AO, Miser, JS, Jefferson, J. Experimental evidence for a neural origin of Ewing's sarcoma of bone. Am J Pathol 1987;127:507.Google ScholarPubMed
Stout, AP, Murray, MR. Neuroepithelioma of radial nerve with study of its behavior in vitro. Rev Can Biol 1942;1:651.Google Scholar
Delattre, O, Zucman, J, Melot, T. The Ewing family of tumors – A subgroup of small-round-cell tumors defined by specific chimeric transcripts. N Engl J Med 1994;331:294.CrossRefGoogle ScholarPubMed
Grier, HE. The Ewing family of tumors. Ewing's sarcoma and primitive neuroectodermal tumors. Pediatr Clin North Am 1997;44:991.CrossRefGoogle ScholarPubMed
Kissane, JM, Askin, FB, Foulkes, M. Ewing's sarcoma of bone: clinicopathologic aspects of 303 cases from the Intergroup Ewing's Sarcoma Study. Hum Pathol 1983;14:773.CrossRefGoogle ScholarPubMed
Soule, EH, Newton, W., Moon TE, et al. Extraskeletal Ewing's sarcoma: a preliminary review of 26 cases encountered in the Intergroup Rhabdomyosarcoma Study. Cancer 1978;42:259.3.0.CO;2-R>CrossRefGoogle ScholarPubMed
Harms, D. Soft tissue sarcomas in the Kiel Pediatric Tumor Registry. Current Topics in Pathology 1995;89:31.CrossRefGoogle ScholarPubMed
Holman, CDJ, Reynolds, PM, Byrne, MJJ. Possible infectious etiology of six cases of Ewing's sarcoma in Western Australia. Cancer 1983;52:1974.3.0.CO;2-9>CrossRefGoogle ScholarPubMed
Hutter, RVP, Francis, KD, Foote, FW. Ewing's sarcoma in siblings: report of the second known occurence. Am J Surg 1964;4:598.CrossRefGoogle Scholar
Gariepy, G, Drouin, R, Lemieux, N. Ultrastructural, immunohistochemical, and cytogenetic study of a malignant peripheral neuroectodermal tumor in a patient seropositive for human immunodeficiency virus. Am J Clin Pathol 1990;93:818.CrossRefGoogle Scholar
Ogawa, K. Embryonal neuroepithelial tumors induced by human adenovirus type 12 in rodents. 1. Tumor induction in the peripheral nervous system. Acta Neuropathol (Berl) 1989;77:244.CrossRefGoogle ScholarPubMed
Mirra, JM. Bone Tumors: Clinical, Radiologic, and Pathologic Correlations. Philadelphia, Lea & Febiger, 1989.Google Scholar
Marina, NM, Etcubanas, E, Parham, DM. Peripheral primitive neuroectodermal tumor (peripheral neuroepithelioma) in children: A review of the St. Jude experience and controversies in diagnosis and management. Cancer 1989;64:1952.3.0.CO;2-W>CrossRefGoogle ScholarPubMed
Shimada, H, Newton, WA., Soule EH, et al. Pathologic features of extraosseous Ewing's sarcoma: A report from the Intergroup Rhabdomyosarcoma Study. Hum Pathol 1988;19:442.CrossRefGoogle ScholarPubMed
Rodriguez-Galindo, C, Marina, NM, Fletcher, BD. Is primitive neuroectodermal tumor of the kidney a distinct entity?Cancer 1997;79:2243.3.0.CO;2-V>CrossRefGoogle ScholarPubMed
Sexton, CW, White, WL. Primary cutaneous Ewing's family sarcoma. Report of a case with immunostaining for glycoprotein p30/32 mic2. Am J Dermatopathol 1996;18:601.CrossRefGoogle ScholarPubMed
Smith, LM, Adams, RH, Brothman, AR. Peripheral primitive neuroectodermal tumor presenting with diffuse cutaneous involvement and 7;22 translocation. Med Pediatr Oncol 1998;30:357.3.0.CO;2-F>CrossRefGoogle ScholarPubMed
Ehrig, T, Billings, SC, Fanburg-Smith, JC. Superficial primitive neuroectodermal tumor/Ewing sarcoma (PN/ES): same tumor as deep PN/ES or new entity?Ann Diagn Pathol 2007;11:153–159.CrossRefGoogle ScholarPubMed
Kaspers, G-JJL, Kamphorst, W, Graaff, M. Primary spinal epidural extraosseous Ewing's sarcoma. Cancer 1991;68:648.3.0.CO;2-Q>CrossRefGoogle ScholarPubMed
Mobley, BC, Roulston, D, Shah, GV. Peripheral primitive neuroectodermal tumor/Ewing's sarcoma of the craniospinal vault: care reports and review. Hum Pathol 2006;37:845–853.CrossRefGoogle Scholar
Meis, JM, Enzinger, FM, Martz, KL. Malignant peripheral nerve sheath tumors (malignant schwannomas) in children. Am J Surg Pathol 1992;16:694.CrossRefGoogle ScholarPubMed
Hanna, SL, Fletcher, BD, Kaste, SC. Increased confidence of diagnosis of Ewing sarcoma using T2- weighted MR images. Magn Reson Imaging 1994;12:559.CrossRefGoogle ScholarPubMed
Lemmi, MA, Fletcher, BD, Marina, NM. Use of MR imaging to assess results of chemotherapy for Ewing sarcoma. AJR Am J Roentgenol 1990;155:343.CrossRefGoogle ScholarPubMed
Tsokos, M. Peripheral primitive neuroectodermal tumors. Diagnosis, classification, and prognosis. Perspect Pediatr Pathol 1992;16:27.Google ScholarPubMed
Carr, TF, Lockwood, L, Stevens, RF. Childhood B cell lymphomas arising in the mediastinum. J Clin Pathol 1993;46:513.CrossRefGoogle ScholarPubMed
Riopel, M, Dickman, PS, Link, MP. MIC2 analysis in pediatric lymphomas and leukemias. Hum Pathol 1994;25:396.CrossRefGoogle ScholarPubMed
Nascimento, AG, Unni, KK, Pritchard, DJ. A clinicopathologic study of 20 cases of large-cell (atypical) Ewing's sarcoma of bone. Am J Surg Pathol 1980;4:29.CrossRefGoogle ScholarPubMed
Cavazzana, AO, Ninfo, V, Roberts, J. Peripheral neuroepithelioma: a light microscopic, immunocytochemistry, and ultrastructural study. Mod Pathol 1992;5:71.Google ScholarPubMed
Williams, S, Parham, DM, Jenkins, JJ. Peripheral neuroepithelioma with ganglion cells: report of two cases and review of the literature. Pediatr Dev Pathol 1999;2:42.CrossRefGoogle ScholarPubMed
Kawamoto, EH, Weidner, N, Agostini, RM. Malignant ectomesenchymoma of soft tissue: report of two cases and review of the literature. Cancer 1987; 59: 1791.3.0.CO;2-#>CrossRefGoogle ScholarPubMed
Dehner, LP: Whence the primitive neuroectodermal tumor. Arch Pathol Lab Med 1990;114:16.Google ScholarPubMed
Llombart-Bosch, A, Peydro-Olaya, A. Scanning and transmission electron microscopy of Ewing's sarcoma of bone (typical and atypical variants). Virchows Arch [A] 1983;398: 329.CrossRefGoogle Scholar
Triche, TJ, Ross, WE. Glycogen-containing neuroblastoma with clinical and histopathologic features of Ewing's sarcoma. Cancer 1978;41:1425.3.0.CO;2-9>CrossRefGoogle ScholarPubMed
Tsokos, M. Peripheral primitive neuroectodermal tumors: diagnosis, classification, and prognosis. In Garvin, AJ, O'Leary, TJ, Bernstein, J, Rosenberg, HS. (eds): Pediatric Molecular Pathology: Quantitation and Applications. Perspectives in Pediatric Pathology, Vol. 16, p 27. Basel, Karger, 1992.Google Scholar
Damjanov, I, Tuma, B, Dominis, M. Electron microscopy of large cell undifferentiated and giant cell tumors. Pathol Res Pract 1989;184:137.CrossRefGoogle ScholarPubMed
Miettinen, M, Lehto, VP, Virtanen, I. Histogenesis of Ewing's sarcoma. An evaluation of intermediate filaments and endothelial cell markers. Virchows Arch [B] 1982;41:277.CrossRefGoogle ScholarPubMed
Llombart-Bosch, A, Lacombe, MJ, Peydro-Olaya, A. Malignant peripheral neuroectodermal tumours of bone other than Askin's neoplasm: characterization of 14 new cases with immunohistochemistry and electron microscopy. Virchows Arch [A] 1988;412:421.CrossRefGoogle ScholarPubMed
Pinto, A, Grant, LH, Hayes, FA. Immunohistochemical expression of neuron-specific enolase and Leu7 in Ewing's sarcoma of bone. Cancer 1989;64:1266.3.0.CO;2-0>CrossRefGoogle Scholar
Greco, MA, Steiner, GC, Fazzini, E. Ewing's sarcoma with epithelial differentiation: fine structural and immunocytochemical study. Ultrastruct Pathol 1988;12:317.CrossRefGoogle ScholarPubMed
Moll, R, Lee, I, Gould, VE. Immunocytochemical analysis of Ewing's tumors: patterns of expression of intermediate filaments and desmosomal proteins indicate cell type heterogeneity and pluripotential differentiation. Am J Pathol 1987;127:288.Google ScholarPubMed
Bridge, JA, Fidler, ME, Neff, JR. Adamantinoma-like Ewing's sarcoma: genomic confirmation, phenotypic drift. Am J Surg Pathol 1999;23:159.CrossRefGoogle ScholarPubMed
Ambros, IM, Ambros, PF, Strehl, S. 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;67:1886.3.0.CO;2-U>CrossRefGoogle ScholarPubMed
Hamilton, G, Mallinger, R, Hofbauer, S. The monoclonal HBA-71 antibody modulates proliferation of thymocytes and Ewing's sarcoma cells by interfering with the action of insulin-like growth factor I. Thymus 1991;18:33.Google ScholarPubMed
Ramani, P, Rampling, D, Link, M. Immunocytochemical study of 12E7 in small round-cell tumours of childhood: An assessment of its sensitivity and specificity. Histopathology 1993; 23: 557.CrossRefGoogle ScholarPubMed
Stevenson, AJ, Chatten, J, Bertoni, F. CD99 (p30/32MIC2) neuroectodermal/Ewing's sarcoma antigen as an immunohistochemical marker: review of more than 600 tumors and the literature experience. App Immunohistochem 1994;2:231.Google Scholar
Fellinger, EJ, Garin-Chesa, P, Glasser, DB. Comparison of cell surface antigen HBA71 (p30/32MIC2), neuron-specific enolase, and vimentin in the immunohistochemical analysis of Ewing's sarcoma of bone. Am J Surg Pathol 1992;16:746.CrossRefGoogle Scholar
Weiss, LM, Arber, DA, Chang, KL. CD45: a review. Appl Immunohistochem Mol Morphol 1993;1:166.Google Scholar
Ozdemirli, M, Fanburg-Smith, JC, Hartmann, DP. 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.CrossRefGoogle ScholarPubMed
Pappo, AS, Douglass, EC, Meyer, WH. Use of HBA71 and anti-b2-microglobulin to distinquish peripheral neuroepithelioma from neuroblastoma. Hum Pathol 1993;24:880.CrossRefGoogle Scholar
Molenaar, WM, Muntinghe, FL. Expression of neural cell adhesion molecules and neurofilament protein isoforms in skeletal muscle tumors. Hum Pathol 1998;29:1290.CrossRefGoogle ScholarPubMed
Strother, DR, Parham, DM, Houghton, PJ. Expression of the 5.1 H11 antigen, a fetal muscle surface antigen, in normal and neoplastic tissue. Arch Pathol Lab Med 1990;114:593.Google ScholarPubMed
Garin-Chesa, P, Fellinger, EJ, Huvos, AG. Immunohistochemical analysis of neural cell adhesion molecules: Differential expression in small round cell tumors of childhood and adolescence. Am J Pathol 1991;139:275.Google ScholarPubMed
Mhawech-Fauceglia, P, Herrmann, F, Penetrante, P. Diagnostic utility of FLI-1 monoclonal antibody and dual-color, break-apart probe fluorescence in situ (FISH) analysis in Ewing's sarcoma/primitive neuroectodermal tumour (EWS/PNET). A comparative study with CD99 and FLI-1 polyclonal antibodies. Histopathology 2006;49:569–575.CrossRefGoogle Scholar
Parham, DM, Dias, P, Kelly, DR. Desmin positivity in primitive neuroectodermal tumors of childhood. Am J Surg Pathol 1992;16:483.CrossRefGoogle ScholarPubMed
Douarin, NM, Ziller, C. Plasticity in neural crest cell differentiation. Curr Opin Cell Biol 1993;5:1036.CrossRefGoogle ScholarPubMed
Murre, C, McCaw, PS, Vaessin, H. Interactions between heterologous helix-loop-helix proteins generate complexes that bind specifically to a common DNA sequence. Cell 1989;58:537.CrossRefGoogle ScholarPubMed
Boue, DR, Parham, DM, Webber, B, Crist, WM, Qualman, SJ. Clinicopathologic study of ectomesenchymomas from Intergroup Rhabdomyosarcoma Study Groups III and IV. Pediatr Dev Pathol 2000;3:290–300.Google ScholarPubMed
Navarro, S, Cavazzana, AO, Llombart-Bosch, A. Comparison of Ewing's sarcoma of bone and peripheral neuroepithelioma: an immunocytochemical and ultrastructural analysis of two primitive neuroectodermal neoplasms. Arch Pathol Lab Med 1994;118:608.Google ScholarPubMed
Douglass, EC, Rowe, ST, Valentine, M. A second nonrandom translocation, der(16)t(1;16)(q21;q13), in Ewing sarcoma and peripheral neuroectodermal tumor. Cytogenet Cell Genet 1990;53:87.CrossRefGoogle Scholar
Maurici, D, Perez-Atayde, A, Grier, HE. Frequency and implications of chromosome 8 and 12 gains in Ewing sarcoma. Cancer Genet Cytogenet 1998;100:106.CrossRefGoogle ScholarPubMed
Mugneret, F, Lizard, S, Aurias, A. Chromosomes in Ewing's sarcoma. II. Nonrandom additional changes, trisomy 8 and der(16)t(1;16). Cancer Genet Cytogenet 1988;32:239.CrossRefGoogle Scholar
Hattinger, CM, Rumpler, S, Strehl, S. Prognostic impact of deletions at 1p36 and numerical aberrations in Ewing tumors. Genes Chromosomes Cancer 1999;24:243.3.0.CO;2-A>CrossRefGoogle ScholarPubMed
Armengol, G, Tarkkanen, M, Virolainen, M. Recurrent gains of 1q, 8 and 12 in the Ewing family of tumours by comparative genomic hybridization. Br J Cancer 1997;75:1403.CrossRefGoogle ScholarPubMed
Delattre, O, Zucman, J, Plougastel, B. Gene fusion with an ETS DNA-binding domain caused by chromosome translocation in human tumours. Nature 1992;359:162.CrossRefGoogle ScholarPubMed
Ladanyi, M. The emerging molecular genetics of sarcoma translocations. Diagn Mol Pathol 1995;4:162.CrossRefGoogle ScholarPubMed
Ouchida, M, Ohno, T, Fujimura, Y. Loss of tumorigenicity of Ewing's sarcoma cells expressing antisense RNA to EWS-fusion transcripts. Oncogene 1995;11:1049.Google ScholarPubMed
Ida, K, Kobayashi, S, Taki, T. EWS-FLI-1 and EWS-ERG chimeric mRNAs in Ewing's sarcoma and primitive neuroectodermal tumor. Int J Cancer 1995;63:500.CrossRefGoogle ScholarPubMed
Jeon, IS, Davis, JN, Braun, BS. A variant Ewing's sarcoma translocation (7;22) fuses the EWS gene to the ETS gene ETV1. Oncogene 1995;10:1229.Google ScholarPubMed
Peter, M, Couturier, J, Pacquement, H. A new member of the ETS family fused to EWS in Ewing tumors. Oncogene 1997;14:1159.CrossRefGoogle ScholarPubMed
Ishida, S, Yoshida, K, Kaneko, Y. The genomic breakpoint and chimeric transcripts in the EWSR1-ETV4/E1AF gene fusion in Ewing sarcoma. Cytogenet Cell Genet 1998;82:278.CrossRefGoogle ScholarPubMed
Downing, JR, Head, DR, Parham, DM. Detection of the (11;22)(q24;q12) translocation of Ewing's sarcoma and peripheral neuroectodermal tumor by reverse transcription polymerase chain reaction. Am J Pathol 1993;143:1294.Google ScholarPubMed
Desmaze, C, Zucman, J, Delattre, O. Interphase molecular cytogenetics of Ewing's sarcoma and peripheral neuroepithelioma t(11;22) with flanking and overlapping cosmid probes. Cancer Genet Cytogenet 1994;74:13.CrossRefGoogle Scholar
Kumar, S, Pack, S, Kumar, D. Detection of EWS-FLI-1 fusion in Ewing's sarcoma/peripheral primitive neuroectodermal tumor by fluorescence in situ hybridization using formalin-fixed paraffin-embedded tissue. Hum Pathol 1999;30:324.CrossRefGoogle ScholarPubMed
Bridge, RS, Rajamam, V, Dehner, LP. Molecular diagnosis of Ewing sarcoma/primitive neuroectodermal tumor in routinely processed tissue: a comparison of two FISH strategies and RT-PCR in malignant round cell tumors. Mod Pathol 2006;12:1–18.CrossRefGoogle Scholar
Zucman, J, Melot, T, Desmaze, C. Combinatorial generation of variable fusion proteins in the Ewing family of tumours. EMBO J 1993;12:4481.Google ScholarPubMed
Meier, VS, Kuhne, T, Jundt, G. Molecular diagnosis of Ewing tumors: improved detection of EWS-FLI-1 and EWS-ERG chimeric transcripts and rapid determination of exon combinations. Diagn Mol Pathol 1998;7:29.CrossRefGoogle ScholarPubMed
West, DC, Grier, HE, Swallow, MM. Detection of circulating tumor cells in patients with Ewing's sarcoma and peripheral primitive neuroectodermal tumor. J Clin Oncol 1997;15:583.CrossRefGoogle ScholarPubMed
Vermeulen, J, Ballet, S, Oberlin, O. Incidence and prognostic value of tumor cells detected by RT-PCR in peripheral blood stem cell collections from patients with Ewing's sarcoma. Br J Cancer 2006;95:1326–1333.CrossRefGoogle Scholar
Alava, E, Kawai, A, Healey, JH. EWS-FLI1 fusion transcript structure is an independent determinant of prognosis in Ewing's sarcoma. J Clin Oncol 1998;16:1248.CrossRefGoogle ScholarPubMed
Kovar, H, Jug, G, Aryee, DN. Among genes involved in the RB dependent cell cycle regulatory cascade, the p16 tumor suppressor gene is frequently lost in the Ewing family of tumors. Oncogene 1997;15:2225.CrossRefGoogle ScholarPubMed
Ladanyi, M, Lewis, R, Jhanwar, SC. MDM2 and CDK4 gene amplification in Ewing's sarcoma. J Pathol 1995;175:211.CrossRefGoogle ScholarPubMed
Lawlor, ER, Lim, JF, Tao, W. The Ewing tumor family of peripheral primitive neuroectodermal tumors expresses human gastrin-releasing peptide. Cancer Res 1998;58:2469.Google ScholarPubMed
Rodriguez-Galindo, C, Navid, F, Khoury, J, Krasin, M. Ewing sarcoma family of tumors. In Pappo, AS, ed, Pediatric Bone and Soft Tissue Tumors. Berlin, Springer, 2006, pp. 181–218.CrossRefGoogle Scholar
Schmidt, D, Herrmann, C, Jurgens, H. Malignant peripheral neuroectodermal tumor and its necessary distinction from Ewing's sarcoma. A report from the Kiel Pediatric Tumor Registry. Cancer 1991;68:2251.3.0.CO;2-X>CrossRefGoogle ScholarPubMed
Parham, DM, Hijazi, Y, Steinberg, SM. Neuroectodermal differentiation in Ewing's sarcoma family of tumors does not predict tumor behavior. Hum Pathol 1999;30:911.CrossRefGoogle Scholar
Oberlin, O, Patte, C, Demeocq, R. The response to initial chemotherapy as a prognostic factor in localized Ewing's sarcoma. Eur J Cancer Clin Oncol 1985;21:463.CrossRefGoogle ScholarPubMed
Picci, P, Bohling, T, Bacci, G. Chemotherapy-induced tumor necrosis as a prognostic factor in localized Ewing's sarcoma of the extremities. J Clin Oncol 1997;15: 1553.CrossRefGoogle ScholarPubMed
Bacci, G, Longhi, A, Ferrari, S. Prognostic factors in non-metastatic Ewing's sarcoma of bone: an analysis of 579 patients treated at a single institution with adjuvant or neoadjuvant chemotherapy between 1972 and 1998. Acta Oncol 2006;45:469–475.CrossRefGoogle ScholarPubMed
Aparicio, J, Munarriz, B, Pastor, M. Long-term follow-up and prognostic factors in Ewing's sarcoma. A multivariate analysis of 116 patients from a single institution. Oncology 1998; 55:20.CrossRefGoogle ScholarPubMed
Wexler, LH, Delaney, TF, Tsokos, M. Ifosfamide and etoposide plus vincristine, doxorubicin, and cyclophosphamide for newly diagnosed Ewing's sarcoma family of tumors. Cancer 1996;78:901.3.0.CO;2-X>CrossRefGoogle ScholarPubMed
Jürgens, H, Exner, U, Gadner, H. Multidisciplinary treatment of primary Ewing's sarcoma of bone: a 6-year experience of a European cooperative trial. Cancer 1988;61:23.3.0.CO;2-M>CrossRefGoogle ScholarPubMed
Ozaki, T, Hillmann, A, Hoffmann, C. Significance of surgical margin on the prognosis of patients with Ewing's sarcoma – A report from the cooperative Ewing's sarcoma study. Cancer 1996;78:892.3.0.CO;2-P>CrossRefGoogle ScholarPubMed
Evans, RG. The four S's of Ewing's sarcoma. Int J Radiat Oncol Biol Phys 1991;21:1671.CrossRefGoogle ScholarPubMed
Thorner, P, Squire, J, Chilton-MacNeill, S. Is the EWS/FLI-1 fusion transcript specific for Ewing sarcoma and peripheral primitive neuroectodermal tumor? A report of four cases showing this transcript in a wider range of tumor types. Am J Pathol 1996;148:1125.Google Scholar
Jambhekar, NA, Bagwan, IN, Ghule, P. Comparative analysis of routine histology, immunohistochemistry, reverse transcriptase polymerase chain reaction, and fluorescence in situ hybridization in Ewing family of tumors. Arch Pathol Lab Med 2006;130:1813–1818.Google ScholarPubMed
Ahmed, AA, Nava, VE, Pham, T. Ewing sarcoma family of tumors in unusual sites: confirmation by rt-PCR. Pediatr Devel Pathol 2006;9:488–495.CrossRefGoogle ScholarPubMed
Dehner, LP. The evolution of the diagnosis and understanding of primitive and embryonic neoplasms in children: living through an epoch [review] . Mod Pathol 1998;11:669–685.Google Scholar
Wright, JH. Neurocytoma or neuroblastoma, a kind of tumor not generally recognized. J Exp Med 1910;12:556–561.CrossRefGoogle Scholar
Willis, RA. The Borderland of Embryology and Pathology, 2 ed. London, Butterworths, 1962.Google Scholar
Beckwith, JB, Martin, RF. Observations on the histopathology of neuroblastomas. J Pediatr Surg 1968;3:106–110.CrossRefGoogle ScholarPubMed
Joshi, VV, Cantor, AB, Altshuler, G, Larkin, EW, Neill, JSA, Shuster, JJ. Age-linked prognostic categorization based on a new histologic grading system of neuroblastomas: A clinicopathologic study of 211 cases from the Pediatric Oncology Group. Cancer 1992;69:2197–2211.3.0.CO;2-X>CrossRefGoogle ScholarPubMed
Beckwith, JB, Perrin, EV. In situ neuroblastoma: a contribution to the natural history of the neural crest tumors. Am J Pathol 1963;43:1089–1104.Google ScholarPubMed
Acharya, S, Jayabose, S, Kogan, SJ, Tugal, O, Beneck, D, Leslie, D. Prenatally diagnosed neuroblastoma [review]. Cancer 1997;80:304–310.3.0.CO;2-Y>CrossRefGoogle Scholar
Brodeur, GM, Moley, JF. Biology of tumors of the peripheral nervous system. Cancer Metastasis Rev 1991;10:321–333.CrossRefGoogle ScholarPubMed
Sano, H, Bonadio, J, Gerbing, RB, London, WB, Matthay, KK, Lukens, JN. International neuroblastoma pathology classification adds independent prognostic information beyond the prognostic contribution of age. Eur J Cancer 2006;42:1113–1119.CrossRefGoogle ScholarPubMed
Navarro, S, Amann, G, Beiske, K, Cullinane, CJ, d'Amore, ES, Gambini, C. Prognostic value of International Neuroblastoma Pathology Classification in localized resectable peripheral neuroblastic tumors: a histopathologic study of localized neuroblastoma European Study Group 94.01 Trial and Protocol. J Clin Oncol 2006;24:695–699.CrossRefGoogle Scholar
Dehner, LP. Peripheral and central primitive neuroectodermal tumors. A nosologic concept seeking a consensus. Arch Pathol Lab Med 1986;110:997.Google ScholarPubMed
Matthay, KK. Neuroblastoma: a clinical challenge and biologic puzzle. Ca: A Cancer Journal for Clinicians 1995;45:179.Google ScholarPubMed
Evans, AE, D'Angio, GJ, Propert, K. Prognostic factors in neuroblastoma. Cancer 1987;59:1853.3.0.CO;2-F>CrossRefGoogle ScholarPubMed
Kelly, DR, Joshi, VV. Neuroblastoma and related tumors. In Parham, DM(eds): Pediatric Neoplasia: Morphology and Biology, p. 105. Philadelphia, Lippincott-Raven, 1996.Google Scholar
Bolande, RP. A natural immune system in pregnancy serum lethal to human neuroblastoma cells: A possible mechanism of spontaneous regression. Perspect Pediatr Pathol 1992;16:120.Google ScholarPubMed
Graeve, JLA, Alarcon, PA, Sato, Y. Miliary pulmonary neuroblastoma: A risk of autologous bone marrow transplantation. Cancer 1988;62:2125.3.0.CO;2-1>CrossRefGoogle ScholarPubMed
Telles, NC, Rabson, AS, Pomeroy, TC. Ewing's sarcoma: an autopsy study. Cancer 1978;41:2321.3.0.CO;2-Y>CrossRefGoogle Scholar
Asami, T, Otabe, N, Wakabayashi, M, Kakihara, T, Uchiyama, M, Asami, K. Screening for neuroblastoma: A 9-year birth cohort-based study in Niigata, Japan. Acta Paediatr 1995;84:1173–1176.CrossRefGoogle ScholarPubMed
Brodeur, GM, Pritchard, J, Berthold, F. Revisions of the international criteria for neuroblastoma diagnosis, staging, and response to treatment. J Clin Oncol 1993;11:1466.CrossRefGoogle ScholarPubMed
Katzenstein, HM, Kent, PM, London, WB, Cohn, SL. Treatment and outcome of 83 children with intraspinal neuroblastoma: the Pediatric Oncology Group experience. J Clin Oncol 2001;19:1047–1055.CrossRefGoogle ScholarPubMed
Escobar, MA, Grosfeld, JL, Powell, RL, West, KW, Scherer, LR, Fallon, RJ. Long-term outcomes in patients with stage IV neuroblastoma. J Pediatr Surg 2006;41:377–381.CrossRefGoogle ScholarPubMed
Acharya, S, Jayabose, S, Kogan, SJ. Prenatally diagnosed neuroblastoma. Cancer 1997;80:304.3.0.CO;2-Y>CrossRefGoogle ScholarPubMed
Joshi, VV, Silverman, JF, Altshuler, G. Systematization of primary histopathologic and fine-needle aspiration cytologic features and description of unusual histopathologic features of neuroblastic tumors: A report from the Pediatric Oncology Group. Hum Pathol 1993;24:493.CrossRefGoogle ScholarPubMed
Umehara, S, Nakagawa, A, Matthay, KK, Lukens, JN, Seeger, RC, Stram, . Histopathology defines prognostic subsets of ganglioneuroblastoma, nodular. Cancer 2000;89:1150–1161.3.0.CO;2-7>CrossRefGoogle ScholarPubMed
Shimada, H, Ambros, IM, Dehner, LP. Terminology and morphologic criteria of neuroblastic tumors: recommendations by the International Neuroblastoma Pathology Committee. Cancer 1999;86:349.3.0.CO;2-Y>CrossRefGoogle ScholarPubMed
Shimada, H, Chatten, J, Newton, WA., et al. Histopathologic prognostic factors in neuroblastic tumors: definition of subtypes of ganglioneuroblastoma and an age-linked classification of neuroblastomas. J Natl Cancer Inst 1984;73:405.CrossRefGoogle ScholarPubMed
Shimada, H, Ambros, IM, Dehner, LP. The International Neuroblastoma Pathology Classification (the Shimada system). Cancer 1999;86:364.3.0.CO;2-7>CrossRefGoogle Scholar
Cozzutto, C, Carbone, A. Pleomorphic (anaplastic) neuroblastoma. Arch Pathol 1988;112:621.Google ScholarPubMed
Gonzalez-Crussi, F, Hsueh, W. Bilateral adrenal ganglioneuroblastoma with neuromelanin. Clinical and pathologic observations. Cancer 1988;61:1159.3.0.CO;2-S>CrossRefGoogle ScholarPubMed
Tsokos, M, Scarpa, S, Ross, RA. Differentiation of human neuroblastoma recapitulates neural crest development. Study of morphology, neurotransmitter enzymes, and extracellular matrix proteins. Am J Pathol 1987;128:484.Google ScholarPubMed
Balázs, M. Mixed pheochromocytoma and ganglioneuroma of the adrenal medulla: A case report with electron microscopic examination. Hum Pathol 1988;19:1352.CrossRefGoogle ScholarPubMed
Blatt, J, Gula, MJ, Orlando, SJ. Indolent course of advanced neuroblastoma in children older than 6 years at diagnosis. Cancer 1995;76:890.3.0.CO;2-X>CrossRefGoogle ScholarPubMed
Mierau, GW, Berry, PJ, Malott, RL. Appraisal of the comparative utility of immunohistochemistry and electron microscopy in the diagnosis of childhood round cell tumors. Ultrastruct Pathol 1996;20:507.CrossRefGoogle Scholar
Thiele, CJ. Pediatric peripheral neuroectodermal tumors, oncogenes, and differentiation. Cancer Invest 1990;8:629.CrossRefGoogle Scholar
Dias, P, Kumar, P, Marsden, HB. N-myc gene is amplified in alveolar rhabdomyosarcomas (RMS) but not in embryonal RMS. Int J Cancer 1990;45:593.CrossRefGoogle ScholarPubMed
Driman, D, Thorner, PS, Greenberg, ML. MYCN gene amplification in rhabdomyosarcoma. Cancer 1994;73:2231.3.0.CO;2-E>CrossRefGoogle ScholarPubMed
Leader, M, Collins, M, Patel, J. Antineuron specific enolase staining reactions in sarcomas and carcinomas: its lack of neuroendocrine specificity. J Clin Pathol 1986;39:1186.CrossRefGoogle ScholarPubMed
Brodeur, GM, Green, AA, Hayes, FA. Cytogenetic features of human neuroblastomas and cell lines. Cancer Res 1981;41:4678.Google ScholarPubMed
Maris, JM, Matthay, KK. Molecular biology of neuroblastoma. J Clin Oncol 1999;17:2264.CrossRefGoogle ScholarPubMed
Caren, H, Ejeskar, K, Fransson, S, Hesson, L, Latif, F, Sjoberg, RM. A cluster of genes located in 1p36 are down-regulated in neuroblastomas with poor prognosis, but not due to CpG island methylation. Mol Cancer 2005;4:10.CrossRefGoogle Scholar
Fransson, S, Martinsson, T, Ejeskar, K. Neuroblastoma tumors with favorable and unfavorable outcomes: Significant differences in mRNA expression of genes mapped at 1p36.2. Genes Chromosomes Cancer 2007;46:45–52.CrossRefGoogle ScholarPubMed
Iehara, T, Hosoi, H, Akazawa, K, Matsumoto, Y, Yamamoto, K, Suita, S. MYCN gene amplification is a powerful prognostic factor even in infantile neuroblastoma detected by mass screening. Br J Cancer 2006;94:1510–1515.CrossRefGoogle ScholarPubMed
Shapiro, DN, Valentine, MB, Rowe, ST. Detection of N-myc gene amplification by fluorescence in situ hybridization. Diagnostic utility for neuroblastoma. Am J Pathol 1993;142:1339.Google ScholarPubMed
Bhargava, R, Oppenheimer, O, Gerald, W, Jhanwar, SC, Chen, B. Identification of MYCN gene amplification in neuroblastoma using chromogenic in situ hybridization (CISH): an alternative and practical method. Diagn Mol Pathol 2005;14:72–76.CrossRefGoogle ScholarPubMed
Look, AT, Hayes, FA, Nitschke, R. Cellular DNA content as a predictor of response to chemotherapy in infants with unresectable neuroblastoma. N Engl J Med 1984;311:231.CrossRefGoogle ScholarPubMed
Look, AT, Hayes, FA, Shuster, JJ. Clinical relevance of tumor cell ploidy and N-myc gene amplification in childhood neuroblastoma: A Pediatric Oncology Group study. J Clin Oncol 1991;9:581.CrossRefGoogle ScholarPubMed
Tanaka, T, Sugimoto, T, Sawada, T. Prognostic discrimination among neuroblastomas according to Ha-ras/trk A gene expression: a comparison of the profiles of neuroblastomas detected clinically and those detected through mass screening. Cancer 1998;83:1626.3.0.CO;2-Y>CrossRefGoogle ScholarPubMed
Maitra, A, Yashima, K, Rathi, A. The RNA component of telomerase as a marker of biologic potential and clinical outcome in childhood neuroblastic tumors. Cancer 1999;85:741.3.0.CO;2-6>CrossRefGoogle ScholarPubMed
Ohali, A, Avigad, S, Ash, S, Goshen, Y, Luria, D, Feinmesser, M. Telomere length is a prognostic factor in neuroblastoma. Cancer 2006;107:1391–1399.CrossRefGoogle ScholarPubMed
Brodeur, GM, Azar, C, Brother, M. Neuroblastoma: Effect of genetic factors on prognosis and treatment. Cancer 1992;70 Suppl.:1685.3.0.CO;2-H>CrossRefGoogle ScholarPubMed
Elimam, NA, Atra, AA, Fayea, NY, Al-Asaad, TG, Khattab, TM, Al-Sulami, GA. Stage 4S neuroblastoma, a disseminated tumor with excellent outcome. Saudi Med J 2006;27:1734–1736.Google ScholarPubMed
Navarro, S, Amann, G, Beiske, K, Cullinane, CJ, d'Amore, ES, Gambini, C. Prognostic value of International Neuroblastoma Pathology Classification in localized resectable peripheral neuroblastic tumors: a histopathologic study of localized neuroblastoma European Study Group 94.01 Trial and Protocol. J Clin Oncol 2006;24:695–699.CrossRefGoogle Scholar
Burgues, O, Navarro, S, Noguera, R, Pellin, A, Ruiz, A, Castel, V. Prognostic value of the International Neuroblastoma Pathology Classification in Neuroblastoma (Schwannian stroma-poor) and comparison with other prognostic factors: a study of 182 cases from the Spanish Neuroblastoma Registry. Virchows Arch 2006;449:410–420.CrossRefGoogle ScholarPubMed
Ordonez, NG. Desmoplastic small round cell tumor: I: a histopathologic study of 39 cases with emphasis on unusual histological patterns. Am J Surg Pathol 1998;22:1303.CrossRefGoogle ScholarPubMed
Gerald, WL, Miller, HK, Battifora, H. Intra-abdominal desmoplastic small round-cell tumor: report of 19 cases of a distinctive type of high-grade polyphenotypic malignancy affecting young individuals. Am J Surg Pathol 1991;15:499.CrossRefGoogle Scholar
Sawyer, JR, Tryka, AF, Lewis, JM. A novel reciprocal chromosome translocation t(11;22)(p13; q12) in an intraabdominal desmoplastic small round-cell tumor. Am J Surg Pathol 1992;16:411–416.CrossRefGoogle Scholar
Kretschmar, CS, Colbach, C, Bhan, I. Desmoplastic small cell tumor: a report of three cases and a review of the literature. J Pediatr Hematol Oncol 1996;18:293.CrossRefGoogle Scholar
Ordonez, NG, El-Naggar, AK, Ro, JY. Intra-abdominal desmoplastic small cell tumor: a light microscopic, immunocytochemical, ultrastructural, and flow cytometric study. Hum Pathol 1993;24:850.CrossRefGoogle ScholarPubMed
Kawano, N, Inayama, Y, Nagashima, Y, Miyagi, Y, Uemura, H, Saitoh, K. Desmoplastic small round-cell tumor of the paratesticular region: report of an adult case with demonstration of EWS and WT1 gene fusion using paraffin-embedded tissue. Mod Pathol 1999;12:729–734.Google ScholarPubMed
Adsay, V, Cheng, J, Athanasian, E. Primary desmoplastic small cell tumor of soft tissues and bone of the hand. Am J Surg Pathol 1999;23:1408.CrossRefGoogle ScholarPubMed
Bian, Y, Jordan, AG, Rupp, M. Effusion cytology of desmoplastic small round cell tumor of the pleura: A case report. Acta Cytol 1993;37:77.Google ScholarPubMed
Tison, V, Cerasoli, S, Morigi, F. Intracranial desmoplastic small-cell tumor – Report of a case. Am J Surg Pathol 1996;20:112.CrossRefGoogle ScholarPubMed
Karavitakis, EM, Moschovi, M, Stefanaki, K, Karamolegou, K, Dimitriadis, E, Pandis, N. Desmoplastic small round cell tumor of the pleura. Pediatr Blood Cancer 2007;49:335–338.CrossRefGoogle ScholarPubMed
Wang, LL, Perlman, EJ, Vujanic, GM, Zuppan, C, Brundler, MA, Cheung, CR. Desmoplastic small round cell tumor of the kidney in childhood. Am J Surg Pathol 2007;31:576–584.CrossRefGoogle ScholarPubMed
Finke, NM, Lae, ME, Lloyd, RV, Gehani, SK, Nascimento, AG. Sinonasal desmoplastic small round cell tumor: a case report. Am J Surg Pathol 2002;26:799–803.CrossRefGoogle ScholarPubMed
Schmidt, D, Koster, E, Harms, D. Intraabdominal desmoplastic small-cell tumor with divergent differentiation: clinicopathological findings and DNA ploidy. Med Pediatr Oncol 1994;22:97.CrossRefGoogle ScholarPubMed
Zhang, J, Dalton, J, Fuller, C. Epithelial marker-negative desmoplastic small round cell tumor with atypical morphology: definitive classification by fluorescence in situ hybridization. Arch Pathol Lab Med 2007;131:646–649.Google ScholarPubMed
Ordonez, NG. Desmoplastic small round cell tumor: II: an ultrastructural and immunohistochemical study with emphasis on new immunohistochemical markers. Am J Surg Pathol 1998;22:1314.CrossRefGoogle ScholarPubMed
Trupiano, JK, Machen, SK, Barr, FG. Cytokeratin-negative desmoplastic small round cell tumor: a report of two cases emphasizing the utility of reverse transcriptase-polymerase chain reaction. Mod Pathol 1999;12:849.Google ScholarPubMed
Sawyer, JR, Tryka, AF, Lewis, JM. A novel reciprocal chromosome translocation t(11;22)(p13; q12) in an intraabdominal desmoplastic small round-cell tumor. Am J Surg Pathol 1992;16:411.CrossRefGoogle Scholar
Ladanyi, M, Gerald, W. Fusion of the EWS and WT1 genes in the desmoplastic small round cell tumor. Cancer Res 1994;54:2837.Google ScholarPubMed
Park, S, Schalling, M, Bernard, A. The Wilms tumour gene WT1 is expressed in murine mesoderm-derived tissues and mutated in a human mesothelioma. Nature Genet 1993;4:415.CrossRefGoogle Scholar
Walker, C, Rutten, F, Yuan, X. Wilms' tumor suppressor gene expression in rat and human mesothelioma. Cancer Res 1994;54:3101.Google ScholarPubMed
Roberts, DJ, Haber, D, Sklar, J. Extrarenal Wilms' tumors. A study of their relationship with classical renal Wilms' tumor using expression of WT1 as a molecular marker. Lab Invest 1993;68:528.Google ScholarPubMed
Lee, SB, Kolquist, KA, Nichols, K. The EWS-WT1 translocation product induces PDGFA in desmoplastic small round-cell tumour. Nature Genet 1997;17:309.CrossRefGoogle ScholarPubMed
Alava, E, Ladanyi, M, Rosai, J. Detection of chimeric transcripts in desmoplasmic small round cell tumor and related developmental tumors by reverse transcriptase polymerase chain reaction – A specific diagnostic assay. Am J Pathol 1995;147:1584.Google Scholar
Argatoff, LH, O'Connell, JX, Mathers, JA. Detection of the EWS/WT1 gene fusion by reverse transcriptase-polymerase chain reaction in the diagnosis of intra-abdominal desmoplastic small round cell tumor. Am J Surg Pathol 1996;20:406.CrossRefGoogle Scholar
Katz, RL, Quezado, M, Senderowicz, AM. An intra-abdominal small round cell neoplasm with features of primitive neuroectodermal and desmoplastic round cell tumor and a EWS/FLI-1 fusion transcript. Hum Pathol 1997;28:502.CrossRefGoogle Scholar
Gardner, LJ, Ayala, AG, Monforte, HL, Dunphy, CH. Ewing sarcoma/peripheral primitive neuroectodermal tumor: adult abdominal tumors with an Ewing sarcoma gene rearrangement demonstrated by fluorescence in situ hybridization in paraffin sections. Appl Immunohistochem Mol Morphol 2004;12:160–165.CrossRefGoogle ScholarPubMed
Ordi, J, AE, , Torne, A, Mellado, B, Pardo-Mindan, J, Iglesias, X. Intraabdominal desmoplastic small round cell tumor with EWS/ERG fusion transcript. Am J Surg Pathol 1998;22:1026–1032.CrossRefGoogle ScholarPubMed
Alaggio, R, Rosolen, A, Sartori, F, Leszl, A, d'Amore, ES, Bisogno, G. Spindle cell tumor with EWS-WT1 transcript and a favorable clinical course: a variant of DSCT, a variant of leiomyosarcoma, or a new entity? Report of 2 pediatric cases. Am J Surg Pathol 2007;31:454–459.CrossRefGoogle ScholarPubMed
Amato, RJ, Ellerhorst, JA, Ayala, AG. Intraabdominal desmoplastic small cell tumor. Report and discussion of five cases. Cancer 1996;78:845.3.0.CO;2-U>CrossRefGoogle ScholarPubMed
Saab, R, Khoury, JD, Krasin, M, Davidoff, AM, Navid, F. Desmoplastic small round cell tumor in childhood: the St. Jude Children's Research Hospital experience. Pediatr Blood Cancer 2007;49:274–279.CrossRefGoogle ScholarPubMed
Coppes, MJ, Wilson, PCG, Weitzman, S. Extrarenal Wilms' tumor: Staging, treatment, and prognosis. J Clin Oncol 1991;9:167.CrossRefGoogle Scholar
Parham, DM, Weeks, DA, Beckwith, JB. The clinicopathologic spectrum of putative extrarenal rhabdoid tumors: an analysis of 42 cases studies with immunohistochemistry and/or electron microscopy. Am J Surg Pathol 1994;18: 1010.CrossRefGoogle ScholarPubMed
Leong, FJ, Leong, AS. Malignant rhabdoid tumor in adults – heterogenous tumors with a unique morphological phenotype. Pathol Res Pract 1996;192:796.CrossRefGoogle ScholarPubMed
White, FV, Dehner, LP, Belchis, DA. Congenital disseminated malignant rhabdoid tumor: distinct clinicopathologic entity demonstrating abnormalities of chromosome 22q11. Am J Surg Pathol 1999;23:249.CrossRefGoogle ScholarPubMed
Simons, J, Teshima, I, Zielenska, M. Analysis of chromosome 22q as an aid to the diagnosis of rhabdoid tumor: a case report. Am J Surg Pathol 1999;23:982.CrossRefGoogle Scholar
Beckwith, JB, Palmer, NF. Histopathology and prognosis of Wilms tumors: results from the First National Wilms' Tumor Study. Cancer 1978;41:1937.3.0.CO;2-U>CrossRefGoogle ScholarPubMed
Balaton, AJ, Vaury, P, Videgrain, M. Paravertebral malignant rhabdoid tumor in an adult. A case report with immunocytochemical study. Pathol Res Pract 1987;182:713.Google Scholar
Dabbs, DJ, Park, HK. Malignant rhabdoid skin tumor: an uncommon primary skin neoplasm. Ultrastructural and immunohistochemical analysis. J Cutan Pathol 1988;15:109.CrossRefGoogle ScholarPubMed
Tsuneyoshi, M, Daimaru, Y, Hashimoto, H. Malignant soft tissue neoplasms with the histologic features of renal rhabdoid tumors: an ultrastructural and immunohistochemical study. Hum Pathol 1985;16:1235.CrossRefGoogle ScholarPubMed
Gonzalez-Crussi, F, Goldschmidt, RA, Hsueh, W. Infantile sarcoma with intracytoplasmic filamentous inclusions: distinctive tumor of possible histiocytic origin. Cancer 1982;49:2365.3.0.CO;2-I>CrossRefGoogle ScholarPubMed
Lemos, LB, Hamoudi, AB. Malignant thymic tumor in an infant (malignant histiocytoma). Arch Pathol Lab Med 1978;102:84.Google Scholar
Hajdu, SI. Pathology of Soft Tissue Tumors. Philadelphia, Lea and Febiger, 1979.Google Scholar
Kodet, R, Newton, WA., Sachs, N. Rhabdoid tumors of soft tissues: A clinicopathologic study of 26 cases enrolled in the Intergroup Rhabdomyosarcoma Study. Hum Pathol 1991;22:674.CrossRefGoogle ScholarPubMed
Tsuneyoshi, M, Daimaru, Y, Hashimoto, H. The existence of rhabdoid cells in specified soft tissue sarcomas: histopathological, ultrastructural and immunohistochemical evidence. Virchows Arch [A] 1987;411:509.CrossRefGoogle ScholarPubMed
Kodet, R, Newton, WA., Hamoudi, AB. Rhabdomyosarcomas with intermediate-filament inclusions and features of rhabdoid tumors: Light microscopic and immunohistochemical study. Am J Surg Pathol 1991;15:257.CrossRefGoogle ScholarPubMed
Tsokos, M, Kouraklis, G, Chandra, RS. Malignant rhabdoid tumor of the kidney and soft tissues: evidence for a diverse morphological and immunocytochemical phenotype. Arch Pathol Lab Med 1989;113:115.Google ScholarPubMed
Fanburg-Smith, JC, Hengge, M, Hengge, UR, Smith, JS, Miettinen, M. Extrarenal rhabdoid tumors of soft tissue: a clinicopathologic and immunohistochemical study of 18 cases. Ann Diagn Pathol 1998;2:351–362.CrossRefGoogle ScholarPubMed
Rorke, LB, Packer, RJ, Biegel, JA. Central nervous system atypical teratoid/rhabdoid tumors of infancy and childhood: Definition of an entity. J Neurosurg 1996;85:56.CrossRefGoogle ScholarPubMed
Wick, MR, Ritter, JH, Dehner, LP. Malignant rhabdoid tumors: a clinicopathologic review and conceptual discussion [review]. Semin Diagn Pathol 1995;12:233–248.Google Scholar
Haas, JE, Palmer, NF, Weinberg, AG, Beckwith, JB. Ultrastructure of malignant rhabdoid tumor of the kidney. A distinctive renal tumor of children. Hum Pathol 1981;12:646–657.CrossRefGoogle ScholarPubMed
Chung, CJ, Cammoun, D, Munden, M. Rhabdoid tumor of the kidney presenting as an abdominal mass in a newborn. Pediatr Radiol 1990;20:562.CrossRefGoogle Scholar
Kent, AL, Mahoney, DH, Gresik, MV. Malignant rhabdoid tumor of the extremity. Cancer 1987;60:1056.3.0.CO;2-Q>CrossRefGoogle ScholarPubMed
Dominey, A, Paller, AS, Gonzalez-Crussi, F. Congenital rhabdoid sarcoma with cutaneous metastases. J Am Acad Dermatol 1990;22:969.CrossRefGoogle ScholarPubMed
Perez-Atayde, AR, Newbury, R, Fletcher, JA. Congenital “neurovascular hamartoma” of the skin. A possible marker of malignant rhabdoid tumor. Am J Surg Pathol 1994;18:1030.CrossRefGoogle ScholarPubMed
Jayabose, S, Iqbal, K, Newman, L. Hypercalcemia in childhood renal tumors. Cancer 1988;61:788.3.0.CO;2-H>CrossRefGoogle ScholarPubMed
Mayes, LC, Kasselberg, AG, Roloff, JS. Hypercalcemia associated with immunoreactive parathyroid hormone in a malignant rhabdoid tumor of the kidney (rhabdoid Wilms' tumor). Cancer 1984; 54: 882.3.0.CO;2-N>CrossRefGoogle Scholar
Rousseau-Merck, MF, Nogues, C, Roth, A. Hypercalcemic infantile renal tumors: morphological, clinical, and biological heterogeneity. Pediatr Pathol 1985;3:155.CrossRefGoogle ScholarPubMed
Bonnin, JM, Rubinstein, LJ, Palmer, NF. The association of embryonal tumours originating in the kidney and in the brain. Cancer 1984;54:2137.3.0.CO;2-D>CrossRefGoogle Scholar
Chang, CH, Ramirez, N, Sakr, WA. Primitive neuroectodermal tumor of the brain associated with malignant rhabdoid tumor of the liver: a histologic, immunohistochemical, and electron microscopic study. Pediatr Pathol 1989;9:307.CrossRefGoogle ScholarPubMed
Weeks, DA, Beckwith, JB, Mierau, GW. Rhabdoid tumor of kidney: A report of 111 cases from the National Wilms' Tumor Study Pathology Center. Am J Surg Pathol 1989;13:439.CrossRefGoogle ScholarPubMed
Suzuki, A, Ohta, S, Shimada, M. Gene expression of malignant rhabdoid tumor cell lines by reverse transcriptase-polymerase chain reaction. Diagn Mol Pathol 1997;6:326.CrossRefGoogle ScholarPubMed
Hoot, AC, Russo, P, Judkins, AR, Perlman, EJ, Biegel, JA. Immunohistochemical analysis of hSNF5/INI1 distinguishes renal and extra-renal malignant rhabdoid tumors from other pediatric soft tissue tumors. Am J Surg Pathol 2004;28:1485–1491.CrossRefGoogle ScholarPubMed
Fuller, CE, Pfeifer, J, Humphrey, P, Bruch, , Dehner, LP, Perry, A. Chromosome 22q dosage in composite extrarenal rhabdoid tumors: clonal evolution or a phenotypic mimic?Hum Pathol 2001;32:1102–1108.CrossRefGoogle ScholarPubMed
Wyatt-Ashmead, J, Kleinschmidt-DeMasters, B, Mierau, GW, Malkin, D, Orsini, E, McGavran, L. Choroid plexus carcinomas and rhabdoid tumors: phenotypic and genotypic overlap. Pediatr Dev Pathol 2001;4:545–549.CrossRefGoogle ScholarPubMed
Oda, Y, Tsuneyoshi, M. Extrarenal rhabdoid tumors of soft tissue: clinicopathological and molecular genetic review and distinction from other soft-tissue sarcomas with rhabdoid features. Pathol Int 2006;56:287–295.CrossRefGoogle ScholarPubMed
Modena, P, Lualdi, E, Facchinetti, F, Galli, L, Teixeira, MR, Pilotti, S. SMARCB1/INI1 tumor suppressor gene is frequently inactivated in epithelioid sarcomas. Cancer Res 2005;65:4012–4019.CrossRefGoogle ScholarPubMed
Donner, LR, Wainwright, LM, Zhang, F, Biegel, JA. Mutation of the INI1 gene in composite rhabdoid tumor of the endometrium. Hum Pathol 2007;38:935CrossRefGoogle ScholarPubMed
Cho, YM, Choi, J, Lee, OJ, Lee, HI, Han, DJ, Ro, JY. SMARCB1/INI1 missense mutation in mucinous carcinoma with rhabdoid features. Pathol Int 2006;56:702–706.CrossRefGoogle ScholarPubMed
Douglass, EC, Valentine, M, Rowe, ST. Malignant rhabdoid tumor: a highly malignant childhood tumor with minimal karyotypic changes. Genes Chromosomes Cancer 1990;2:210.CrossRefGoogle ScholarPubMed
Biegel, JA, Rorke, LB, Emanuel, BS. Monosomy 22 in rhabdoid or atypical teratoid tumors of the brain. N Engl J Med 1989;321:906.Google ScholarPubMed
Bhattacharjee, MB, Armstrong, DD, Vogel, H. Cytogenetic analysis of 120 primary pediatric brain tumors and literature review. Cancer Genet Cytogenet 1997;97:39.CrossRefGoogle ScholarPubMed
Schofield, , Beckwith, JB, Sklar, J. Loss of heterozygosity at chromosome regions 22q11–12 and 11p15.5 in renal rhabdoid tumors. Genes Chromosomes Cancer 1996;15:10.3.0.CO;2-7>CrossRefGoogle ScholarPubMed
Biegel, JA, Allen, CS, Kawasaki, K. Narrowing the critical region for a rhabdoid tumor locus in 22q11. Genes Chromosomes Cancer 1996;16:94.3.0.CO;2-Y>CrossRefGoogle ScholarPubMed
Versteege, I, Sevenet, N, Lange, J. Truncating mutations of hSNF5/INI1 in aggressive paediatric cancer. Nature 1998;394:203.CrossRefGoogle ScholarPubMed
Biegel, JA. Molecular genetics of atypical teratoid/rhabdoid tumor. Neurosurg Focus 2006;20:E11.CrossRefGoogle ScholarPubMed
Janson, K, Nedzi, , David, O, Schorin, M, Walsh, JW, Bhattacharjee, M. Predisposition to atypical teratoid/rhabdoid tumor due to an inherited INI1 mutation. Pediatr Blood Cancer 2006;47:279–284.CrossRefGoogle Scholar
White, FV, Dehner, LP, Belchis, DA, Conard, K, Davis, MM, Stocker, JT. Congenital disseminated malignant rhabdoid tumor: a distinct clinicopathologic entity demonstrating abnormalities of chromosome 22q11 [review]. Am J Surg Pathol 1999;23:249–256.CrossRefGoogle Scholar
Garvin, AJ, Re, GG, Tarnowski, BI. The G401 cell line, utilized for studies of chromosomal changes in Wilms' tumor, is derived from a rhabdoid tumor of the kidney. Am J Pathol 1993;142:375.Google ScholarPubMed
Reid, LH, Davies, C, Cooper, PR. A 1-Mb physical map and PAC contig of the imprinted domain in 11p15.5 that contains TAPA1 and the BWSCR1/WT2 region. Genomics 1997;43:366.CrossRefGoogle ScholarPubMed
Sabbioni, S, Barbanti-Brodano, G, Croce, CM. GOK: a gene at 11p15 involved in rhabdomyosarcoma and rhabdoid tumor development. Cancer Res 1997;57:4493.Google ScholarPubMed
Fruhwald, MC, Hasselblatt, M, Wirth, S, Kohler, G, Schneppenheim, R, Subero, JI. Non-linkage of familial rhabdoid tumors to SMARCB1 implies a second locus for the rhabdoid tumor predisposition syndrome. Pediatr Blood Cancer 2006;47:273–278.CrossRefGoogle ScholarPubMed
Bourdeaut, F, Freneaux, P, Thuille, B, Lellouch-Tubiana, A, Nicolas, A, Couturier, J. hSNF5/INI1-deficient tumours and rhabdoid tumours are convergent but not fully overlapping entities. J Pathol 2007;211:323–330.CrossRefGoogle Scholar
Bittesini, L, Dei Tos, AP, Fletcher, CDM. Metastatic malignant melanoma showing a rhabdoid phenotype: Further evidence of a non-specific histological pattern. Histopathology 1992;20:167.CrossRefGoogle ScholarPubMed
Guillou, L, Wadden, C, Coindre, JM. “Proximal-type” epithelioid sarcoma, a distinctive aggressive neoplasm showing rhabdoid features. Clinicopathologic, immunohistochemical, and ultrastructural study of a series. Am J Surg Pathol 1997;21:130.CrossRefGoogle ScholarPubMed
Raney, RB, Asmar, L, Newton, WA. Ewing's sarcoma of soft tissues in childhood: a report from the Intergroup Rhabdomyosarcoma Study, 1972–1991. J Clin Oncol 1997;15:574–582.CrossRefGoogle Scholar
Rud, RP, Reiman, HM, Pritchard, DJ. Extraosseous Ewing's sarcoma, a study of 42 cases. Cancer 1989;64:1548–1553.3.0.CO;2-W>CrossRefGoogle ScholarPubMed
Parkin, DM, Kramároᾓ, E, Draper, GJ. [eds] International Incidence of Childhood Cancer, Vol. II. IARC Scientific Publications, Lyon, 1998, Table 5.27.Google Scholar
London, WB, Look, AT, Brodeur, GM, Altmiller, DH, Thorner, PS. Natural history and biology of stage A neuroblastoma: a Pediatric Oncology Group Study. J Pediatr Hematol Oncol 2000;22:197–205.Google Scholar
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.CrossRefGoogle ScholarPubMed
Triche, TJ, Askin, FB. Neuroblastoma and the differential diagnosis of small-, round-, blue-cell tumors. Hum Pathol 1983;14:569.Google ScholarPubMed
Donaldson, SS. Lessons from our children. Int J Radiat Oncol Biol Phys 1993;26:739.CrossRefGoogle ScholarPubMed
Ross, JA, Severson, RK, Pollock, BH, Robison, LL. Childhood cancer in the United States – A geographical analysis of cases from the Pediatric Cooperative Clinical Trials Groups. Cancer 1996;77:201.3.0.CO;2-7>CrossRefGoogle ScholarPubMed
Olshan, AF, Breslow, NE, Daling, JR. Wilms' tumor and paternal occupation. Cancer Res 1990;50:3212.Google ScholarPubMed
Grundy, P, Wilson, B, Telzerow, P. Uniparental disomy occurs infrequently in Wilms tumor patients. Am J Hum Genet 1994;54:282.Google ScholarPubMed
Koufos, A, Hansen, MF, Copeland, NG. Loss of heterozygosity in three embryonal tumours suggests a common pathogenetic mechanism. Nature 1985;316: 330.CrossRefGoogle ScholarPubMed
Heyn, R, Haeberlen, V, Newton, WA. Second malignant neoplasms in children treated for rhabdomyosarcoma. J Clin Oncol 1993;11:262.CrossRefGoogle ScholarPubMed
May, WA, Lessnick, SL, Braun, BS. The Ewing's sarcoma EWS/FLI-1 fusion gene encodes a more potent transcriptional activator and is a more powerful transforming gene than FLI-1. Mol Cell Biol 1993;13:7393.CrossRefGoogle ScholarPubMed
Whang-Peng, J, Triche, TJ, Knutsen, T. Cytogenetic characterization of selected small round cell tumors of childhood. Cancer Genet Cytogenet 1986;21:185.CrossRefGoogle ScholarPubMed
Oda, H, Imai, Y, Nakatsuru, Y. Somatic mutations of the APC gene in sporadic hepatoblastomas. Cancer Res 1996;56:3320.Google ScholarPubMed
Raffel, C, Jenkins, RB, Frederick, L. Sporadic medulloblastomas contain PTCH mutations. Cancer Res 1997;57:842.Google ScholarPubMed
Felix, CA, Chavez Kappel, C, Mitsudomi, T. Frequency and diversity of p53 mutations in childhood rhabdomyosarcoma. Cancer Res 1992;52:2243.Google ScholarPubMed
Knudson, AG. Antioncogenes and human cancer. Proc Natl Acad Sci U S A 1993;90:10914.CrossRefGoogle ScholarPubMed
Scrable, H, Witte, D, Shimada, H. Molecular differential pathology in rhabdomyosarcoma. Genes Chromosomes Cancer 1989;1:23.CrossRefGoogle ScholarPubMed
Hoovers, JM, Kalikin, LM, Johnson, . Multiple genetic loci within 11p15 defined by Beckwith-Wiedemann syndrome rearrangement breakpoints and subchromosomal transferable fragments. Proc Natl Acad Sci USA 1995;92:12456.Google ScholarPubMed
Jones, PA, Laird, PW. Cancer epigenetics comes of age. Nature Genet 1999;21:163.CrossRefGoogle ScholarPubMed
Baylin, SB. Tying it all together: epigenetics, genetics, cell cycle, and cancer [see comments]. Science 1997;277:1948.CrossRefGoogle Scholar
Chen, B, Dias, P, Jenkins, JJ. Methylation alterations of the MyoD1 upstream region are predictive of subclassification of human rhabdomyosarcomas. Am J Pathol 1998;152:1071.Google ScholarPubMed
Dehner, LP. Primitive neuroectodermal tumor and Ewing's sarcoma. Am J Surg Pathol 1993;17:1.CrossRefGoogle ScholarPubMed
Yunis, EJ. Ewing's sarcoma and related small round cell neoplasms in children. Am J Surg Pathol 1986;10(Supp.1):54.Google ScholarPubMed
Angervall, L, Enzinger, FM. Extraskeletal neoplasm resembling Ewing's sarcoma. Cancer 1975;36:240.3.0.CO;2-H>CrossRefGoogle ScholarPubMed
Tefft, M, Vawter, GF, Mitus, A. Paravertebral “round cell” tumors in children. Radiology 1969;92:1501.CrossRefGoogle ScholarPubMed
Schmidt, D, Mackay, B, Ayala, AG. Ewing's sarcoma with neuroblastoma-like features. Ultrastruct Pathol 1982;3:143.CrossRefGoogle ScholarPubMed
Mierau, GW. Extraskeletal Ewing's sarcoma (peripheral neuroepithelioma). Ultrastruct Pathol 1985;9:91.CrossRefGoogle Scholar
Seemayer, TA, Thelmo, WL, Bolande, RP. Peripheral neuroectodermal tumors. Perspect Pediatr Pathol 1975;2:151.Google ScholarPubMed
Askin, FB, Rosai, J, Sibley, RK. Malignant small cell tumor of the thoracopulmonary region in childhood: a distinctive clinicopathologic entity of uncertain histogenesis. Cancer 1979;43:2438.3.0.CO;2-9>CrossRefGoogle ScholarPubMed
Jaffe, R, Santamaria, M, Yunis, EJ. The neuroectodermal tumor of bone. Am J Surg Pathol 1984;8:885.CrossRefGoogle ScholarPubMed
Turc-Carel, C, Philip, I, Berger, MP. Chromosome study of Ewing's sarcoma (ES) cell lines. Consistency of a reciprocal translocation t(11;22)(q24;q12). Cancer Genet Cytogenet 1984;12:1.CrossRefGoogle Scholar
Whang-Peng, J, Triche, TJ, Knutsen, T. Chromosome translocation in peripheral neuroepithelioma. N Engl J Med 1984;311:584.CrossRefGoogle ScholarPubMed
Ladanyi, M, Lewis, R, Garin-Chesa, P. EWS rearrangement in Ewing's sarcoma and peripheral neuroectodermal tumor. Molecular detection and correlation with cytogenetic analysis and MIC2 expression. Diagn Mol Pathol 1993;2:141.Google ScholarPubMed
Pellin, A, Boix, J, Blesa, JR. EWS/FLI-1 rearrangement in small round cell sarcomas of bone and soft tissue detected by reverse transcriptase polymerase chain reaction amplification. Eur J Cancer [A] 1994;30A:827.CrossRefGoogle ScholarPubMed
Cavazzana, AO, Miser, JS, Jefferson, J. Experimental evidence for a neural origin of Ewing's sarcoma of bone. Am J Pathol 1987;127:507.Google ScholarPubMed
Stout, AP, Murray, MR. Neuroepithelioma of radial nerve with study of its behavior in vitro. Rev Can Biol 1942;1:651.Google Scholar
Delattre, O, Zucman, J, Melot, T. The Ewing family of tumors – A subgroup of small-round-cell tumors defined by specific chimeric transcripts. N Engl J Med 1994;331:294.CrossRefGoogle ScholarPubMed
Grier, HE. The Ewing family of tumors. Ewing's sarcoma and primitive neuroectodermal tumors. Pediatr Clin North Am 1997;44:991.CrossRefGoogle ScholarPubMed
Kissane, JM, Askin, FB, Foulkes, M. Ewing's sarcoma of bone: clinicopathologic aspects of 303 cases from the Intergroup Ewing's Sarcoma Study. Hum Pathol 1983;14:773.CrossRefGoogle ScholarPubMed
Soule, EH, Newton, W., Moon TE, et al. Extraskeletal Ewing's sarcoma: a preliminary review of 26 cases encountered in the Intergroup Rhabdomyosarcoma Study. Cancer 1978;42:259.3.0.CO;2-R>CrossRefGoogle ScholarPubMed
Harms, D. Soft tissue sarcomas in the Kiel Pediatric Tumor Registry. Current Topics in Pathology 1995;89:31.CrossRefGoogle ScholarPubMed
Holman, CDJ, Reynolds, PM, Byrne, MJJ. Possible infectious etiology of six cases of Ewing's sarcoma in Western Australia. Cancer 1983;52:1974.3.0.CO;2-9>CrossRefGoogle ScholarPubMed
Hutter, RVP, Francis, KD, Foote, FW. Ewing's sarcoma in siblings: report of the second known occurence. Am J Surg 1964;4:598.CrossRefGoogle Scholar
Gariepy, G, Drouin, R, Lemieux, N. Ultrastructural, immunohistochemical, and cytogenetic study of a malignant peripheral neuroectodermal tumor in a patient seropositive for human immunodeficiency virus. Am J Clin Pathol 1990;93:818.CrossRefGoogle Scholar
Ogawa, K. Embryonal neuroepithelial tumors induced by human adenovirus type 12 in rodents. 1. Tumor induction in the peripheral nervous system. Acta Neuropathol (Berl) 1989;77:244.CrossRefGoogle ScholarPubMed
Mirra, JM. Bone Tumors: Clinical, Radiologic, and Pathologic Correlations. Philadelphia, Lea & Febiger, 1989.Google Scholar
Marina, NM, Etcubanas, E, Parham, DM. Peripheral primitive neuroectodermal tumor (peripheral neuroepithelioma) in children: A review of the St. Jude experience and controversies in diagnosis and management. Cancer 1989;64:1952.3.0.CO;2-W>CrossRefGoogle ScholarPubMed
Shimada, H, Newton, WA., Soule EH, et al. Pathologic features of extraosseous Ewing's sarcoma: A report from the Intergroup Rhabdomyosarcoma Study. Hum Pathol 1988;19:442.CrossRefGoogle ScholarPubMed
Rodriguez-Galindo, C, Marina, NM, Fletcher, BD. Is primitive neuroectodermal tumor of the kidney a distinct entity?Cancer 1997;79:2243.3.0.CO;2-V>CrossRefGoogle ScholarPubMed
Sexton, CW, White, WL. Primary cutaneous Ewing's family sarcoma. Report of a case with immunostaining for glycoprotein p30/32 mic2. Am J Dermatopathol 1996;18:601.CrossRefGoogle ScholarPubMed
Smith, LM, Adams, RH, Brothman, AR. Peripheral primitive neuroectodermal tumor presenting with diffuse cutaneous involvement and 7;22 translocation. Med Pediatr Oncol 1998;30:357.3.0.CO;2-F>CrossRefGoogle ScholarPubMed
Ehrig, T, Billings, SC, Fanburg-Smith, JC. Superficial primitive neuroectodermal tumor/Ewing sarcoma (PN/ES): same tumor as deep PN/ES or new entity?Ann Diagn Pathol 2007;11:153–159.CrossRefGoogle ScholarPubMed
Kaspers, G-JJL, Kamphorst, W, Graaff, M. Primary spinal epidural extraosseous Ewing's sarcoma. Cancer 1991;68:648.3.0.CO;2-Q>CrossRefGoogle ScholarPubMed
Mobley, BC, Roulston, D, Shah, GV. Peripheral primitive neuroectodermal tumor/Ewing's sarcoma of the craniospinal vault: care reports and review. Hum Pathol 2006;37:845–853.CrossRefGoogle Scholar
Meis, JM, Enzinger, FM, Martz, KL. Malignant peripheral nerve sheath tumors (malignant schwannomas) in children. Am J Surg Pathol 1992;16:694.CrossRefGoogle ScholarPubMed
Hanna, SL, Fletcher, BD, Kaste, SC. Increased confidence of diagnosis of Ewing sarcoma using T2- weighted MR images. Magn Reson Imaging 1994;12:559.CrossRefGoogle ScholarPubMed
Lemmi, MA, Fletcher, BD, Marina, NM. Use of MR imaging to assess results of chemotherapy for Ewing sarcoma. AJR Am J Roentgenol 1990;155:343.CrossRefGoogle ScholarPubMed
Tsokos, M. Peripheral primitive neuroectodermal tumors. Diagnosis, classification, and prognosis. Perspect Pediatr Pathol 1992;16:27.Google ScholarPubMed
Carr, TF, Lockwood, L, Stevens, RF. Childhood B cell lymphomas arising in the mediastinum. J Clin Pathol 1993;46:513.CrossRefGoogle ScholarPubMed
Riopel, M, Dickman, PS, Link, MP. MIC2 analysis in pediatric lymphomas and leukemias. Hum Pathol 1994;25:396.CrossRefGoogle ScholarPubMed
Nascimento, AG, Unni, KK, Pritchard, DJ. A clinicopathologic study of 20 cases of large-cell (atypical) Ewing's sarcoma of bone. Am J Surg Pathol 1980;4:29.CrossRefGoogle ScholarPubMed
Cavazzana, AO, Ninfo, V, Roberts, J. Peripheral neuroepithelioma: a light microscopic, immunocytochemistry, and ultrastructural study. Mod Pathol 1992;5:71.Google ScholarPubMed
Williams, S, Parham, DM, Jenkins, JJ. Peripheral neuroepithelioma with ganglion cells: report of two cases and review of the literature. Pediatr Dev Pathol 1999;2:42.CrossRefGoogle ScholarPubMed
Kawamoto, EH, Weidner, N, Agostini, RM. Malignant ectomesenchymoma of soft tissue: report of two cases and review of the literature. Cancer 1987; 59: 1791.3.0.CO;2-#>CrossRefGoogle ScholarPubMed
Dehner, LP: Whence the primitive neuroectodermal tumor. Arch Pathol Lab Med 1990;114:16.Google ScholarPubMed
Llombart-Bosch, A, Peydro-Olaya, A. Scanning and transmission electron microscopy of Ewing's sarcoma of bone (typical and atypical variants). Virchows Arch [A] 1983;398: 329.CrossRefGoogle Scholar
Triche, TJ, Ross, WE. Glycogen-containing neuroblastoma with clinical and histopathologic features of Ewing's sarcoma. Cancer 1978;41:1425.3.0.CO;2-9>CrossRefGoogle ScholarPubMed
Tsokos, M. Peripheral primitive neuroectodermal tumors: diagnosis, classification, and prognosis. In Garvin, AJ, O'Leary, TJ, Bernstein, J, Rosenberg, HS. (eds): Pediatric Molecular Pathology: Quantitation and Applications. Perspectives in Pediatric Pathology, Vol. 16, p 27. Basel, Karger, 1992.Google Scholar
Damjanov, I, Tuma, B, Dominis, M. Electron microscopy of large cell undifferentiated and giant cell tumors. Pathol Res Pract 1989;184:137.CrossRefGoogle ScholarPubMed
Miettinen, M, Lehto, VP, Virtanen, I. Histogenesis of Ewing's sarcoma. An evaluation of intermediate filaments and endothelial cell markers. Virchows Arch [B] 1982;41:277.CrossRefGoogle ScholarPubMed
Llombart-Bosch, A, Lacombe, MJ, Peydro-Olaya, A. Malignant peripheral neuroectodermal tumours of bone other than Askin's neoplasm: characterization of 14 new cases with immunohistochemistry and electron microscopy. Virchows Arch [A] 1988;412:421.CrossRefGoogle ScholarPubMed
Pinto, A, Grant, LH, Hayes, FA. Immunohistochemical expression of neuron-specific enolase and Leu7 in Ewing's sarcoma of bone. Cancer 1989;64:1266.3.0.CO;2-0>CrossRefGoogle Scholar
Greco, MA, Steiner, GC, Fazzini, E. Ewing's sarcoma with epithelial differentiation: fine structural and immunocytochemical study. Ultrastruct Pathol 1988;12:317.CrossRefGoogle ScholarPubMed
Moll, R, Lee, I, Gould, VE. Immunocytochemical analysis of Ewing's tumors: patterns of expression of intermediate filaments and desmosomal proteins indicate cell type heterogeneity and pluripotential differentiation. Am J Pathol 1987;127:288.Google ScholarPubMed
Bridge, JA, Fidler, ME, Neff, JR. Adamantinoma-like Ewing's sarcoma: genomic confirmation, phenotypic drift. Am J Surg Pathol 1999;23:159.CrossRefGoogle ScholarPubMed
Ambros, IM, Ambros, PF, Strehl, S. 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;67:1886.3.0.CO;2-U>CrossRefGoogle ScholarPubMed
Hamilton, G, Mallinger, R, Hofbauer, S. The monoclonal HBA-71 antibody modulates proliferation of thymocytes and Ewing's sarcoma cells by interfering with the action of insulin-like growth factor I. Thymus 1991;18:33.Google ScholarPubMed
Ramani, P, Rampling, D, Link, M. Immunocytochemical study of 12E7 in small round-cell tumours of childhood: An assessment of its sensitivity and specificity. Histopathology 1993; 23: 557.CrossRefGoogle ScholarPubMed
Stevenson, AJ, Chatten, J, Bertoni, F. CD99 (p30/32MIC2) neuroectodermal/Ewing's sarcoma antigen as an immunohistochemical marker: review of more than 600 tumors and the literature experience. App Immunohistochem 1994;2:231.Google Scholar
Fellinger, EJ, Garin-Chesa, P, Glasser, DB. Comparison of cell surface antigen HBA71 (p30/32MIC2), neuron-specific enolase, and vimentin in the immunohistochemical analysis of Ewing's sarcoma of bone. Am J Surg Pathol 1992;16:746.CrossRefGoogle Scholar
Weiss, LM, Arber, DA, Chang, KL. CD45: a review. Appl Immunohistochem Mol Morphol 1993;1:166.Google Scholar
Ozdemirli, M, Fanburg-Smith, JC, Hartmann, DP. 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.CrossRefGoogle ScholarPubMed
Pappo, AS, Douglass, EC, Meyer, WH. Use of HBA71 and anti-b2-microglobulin to distinquish peripheral neuroepithelioma from neuroblastoma. Hum Pathol 1993;24:880.CrossRefGoogle Scholar
Molenaar, WM, Muntinghe, FL. Expression of neural cell adhesion molecules and neurofilament protein isoforms in skeletal muscle tumors. Hum Pathol 1998;29:1290.CrossRefGoogle ScholarPubMed
Strother, DR, Parham, DM, Houghton, PJ. Expression of the 5.1 H11 antigen, a fetal muscle surface antigen, in normal and neoplastic tissue. Arch Pathol Lab Med 1990;114:593.Google ScholarPubMed
Garin-Chesa, P, Fellinger, EJ, Huvos, AG. Immunohistochemical analysis of neural cell adhesion molecules: Differential expression in small round cell tumors of childhood and adolescence. Am J Pathol 1991;139:275.Google ScholarPubMed
Mhawech-Fauceglia, P, Herrmann, F, Penetrante, P. Diagnostic utility of FLI-1 monoclonal antibody and dual-color, break-apart probe fluorescence in situ (FISH) analysis in Ewing's sarcoma/primitive neuroectodermal tumour (EWS/PNET). A comparative study with CD99 and FLI-1 polyclonal antibodies. Histopathology 2006;49:569–575.CrossRefGoogle Scholar
Parham, DM, Dias, P, Kelly, DR. Desmin positivity in primitive neuroectodermal tumors of childhood. Am J Surg Pathol 1992;16:483.CrossRefGoogle ScholarPubMed
Douarin, NM, Ziller, C. Plasticity in neural crest cell differentiation. Curr Opin Cell Biol 1993;5:1036.CrossRefGoogle ScholarPubMed
Murre, C, McCaw, PS, Vaessin, H. Interactions between heterologous helix-loop-helix proteins generate complexes that bind specifically to a common DNA sequence. Cell 1989;58:537.CrossRefGoogle ScholarPubMed
Boue, DR, Parham, DM, Webber, B, Crist, WM, Qualman, SJ. Clinicopathologic study of ectomesenchymomas from Intergroup Rhabdomyosarcoma Study Groups III and IV. Pediatr Dev Pathol 2000;3:290–300.Google ScholarPubMed
Navarro, S, Cavazzana, AO, Llombart-Bosch, A. Comparison of Ewing's sarcoma of bone and peripheral neuroepithelioma: an immunocytochemical and ultrastructural analysis of two primitive neuroectodermal neoplasms. Arch Pathol Lab Med 1994;118:608.Google ScholarPubMed
Douglass, EC, Rowe, ST, Valentine, M. A second nonrandom translocation, der(16)t(1;16)(q21;q13), in Ewing sarcoma and peripheral neuroectodermal tumor. Cytogenet Cell Genet 1990;53:87.CrossRefGoogle Scholar
Maurici, D, Perez-Atayde, A, Grier, HE. Frequency and implications of chromosome 8 and 12 gains in Ewing sarcoma. Cancer Genet Cytogenet 1998;100:106.CrossRefGoogle ScholarPubMed
Mugneret, F, Lizard, S, Aurias, A. Chromosomes in Ewing's sarcoma. II. Nonrandom additional changes, trisomy 8 and der(16)t(1;16). Cancer Genet Cytogenet 1988;32:239.CrossRefGoogle Scholar
Hattinger, CM, Rumpler, S, Strehl, S. Prognostic impact of deletions at 1p36 and numerical aberrations in Ewing tumors. Genes Chromosomes Cancer 1999;24:243.3.0.CO;2-A>CrossRefGoogle ScholarPubMed
Armengol, G, Tarkkanen, M, Virolainen, M. Recurrent gains of 1q, 8 and 12 in the Ewing family of tumours by comparative genomic hybridization. Br J Cancer 1997;75:1403.CrossRefGoogle ScholarPubMed
Delattre, O, Zucman, J, Plougastel, B. Gene fusion with an ETS DNA-binding domain caused by chromosome translocation in human tumours. Nature 1992;359:162.CrossRefGoogle ScholarPubMed
Ladanyi, M. The emerging molecular genetics of sarcoma translocations. Diagn Mol Pathol 1995;4:162.CrossRefGoogle ScholarPubMed
Ouchida, M, Ohno, T, Fujimura, Y. Loss of tumorigenicity of Ewing's sarcoma cells expressing antisense RNA to EWS-fusion transcripts. Oncogene 1995;11:1049.Google ScholarPubMed
Ida, K, Kobayashi, S, Taki, T. EWS-FLI-1 and EWS-ERG chimeric mRNAs in Ewing's sarcoma and primitive neuroectodermal tumor. Int J Cancer 1995;63:500.CrossRefGoogle ScholarPubMed
Jeon, IS, Davis, JN, Braun, BS. A variant Ewing's sarcoma translocation (7;22) fuses the EWS gene to the ETS gene ETV1. Oncogene 1995;10:1229.Google ScholarPubMed
Peter, M, Couturier, J, Pacquement, H. A new member of the ETS family fused to EWS in Ewing tumors. Oncogene 1997;14:1159.CrossRefGoogle ScholarPubMed
Ishida, S, Yoshida, K, Kaneko, Y. The genomic breakpoint and chimeric transcripts in the EWSR1-ETV4/E1AF gene fusion in Ewing sarcoma. Cytogenet Cell Genet 1998;82:278.CrossRefGoogle ScholarPubMed
Downing, JR, Head, DR, Parham, DM. Detection of the (11;22)(q24;q12) translocation of Ewing's sarcoma and peripheral neuroectodermal tumor by reverse transcription polymerase chain reaction. Am J Pathol 1993;143:1294.Google ScholarPubMed
Desmaze, C, Zucman, J, Delattre, O. Interphase molecular cytogenetics of Ewing's sarcoma and peripheral neuroepithelioma t(11;22) with flanking and overlapping cosmid probes. Cancer Genet Cytogenet 1994;74:13.CrossRefGoogle Scholar
Kumar, S, Pack, S, Kumar, D. Detection of EWS-FLI-1 fusion in Ewing's sarcoma/peripheral primitive neuroectodermal tumor by fluorescence in situ hybridization using formalin-fixed paraffin-embedded tissue. Hum Pathol 1999;30:324.CrossRefGoogle ScholarPubMed
Bridge, RS, Rajamam, V, Dehner, LP. Molecular diagnosis of Ewing sarcoma/primitive neuroectodermal tumor in routinely processed tissue: a comparison of two FISH strategies and RT-PCR in malignant round cell tumors. Mod Pathol 2006;12:1–18.CrossRefGoogle Scholar
Zucman, J, Melot, T, Desmaze, C. Combinatorial generation of variable fusion proteins in the Ewing family of tumours. EMBO J 1993;12:4481.Google ScholarPubMed
Meier, VS, Kuhne, T, Jundt, G. Molecular diagnosis of Ewing tumors: improved detection of EWS-FLI-1 and EWS-ERG chimeric transcripts and rapid determination of exon combinations. Diagn Mol Pathol 1998;7:29.CrossRefGoogle ScholarPubMed
West, DC, Grier, HE, Swallow, MM. Detection of circulating tumor cells in patients with Ewing's sarcoma and peripheral primitive neuroectodermal tumor. J Clin Oncol 1997;15:583.CrossRefGoogle ScholarPubMed
Vermeulen, J, Ballet, S, Oberlin, O. Incidence and prognostic value of tumor cells detected by RT-PCR in peripheral blood stem cell collections from patients with Ewing's sarcoma. Br J Cancer 2006;95:1326–1333.CrossRefGoogle Scholar
Alava, E, Kawai, A, Healey, JH. EWS-FLI1 fusion transcript structure is an independent determinant of prognosis in Ewing's sarcoma. J Clin Oncol 1998;16:1248.CrossRefGoogle ScholarPubMed
Kovar, H, Jug, G, Aryee, DN. Among genes involved in the RB dependent cell cycle regulatory cascade, the p16 tumor suppressor gene is frequently lost in the Ewing family of tumors. Oncogene 1997;15:2225.CrossRefGoogle ScholarPubMed
Ladanyi, M, Lewis, R, Jhanwar, SC. MDM2 and CDK4 gene amplification in Ewing's sarcoma. J Pathol 1995;175:211.CrossRefGoogle ScholarPubMed
Lawlor, ER, Lim, JF, Tao, W. The Ewing tumor family of peripheral primitive neuroectodermal tumors expresses human gastrin-releasing peptide. Cancer Res 1998;58:2469.Google ScholarPubMed
Rodriguez-Galindo, C, Navid, F, Khoury, J, Krasin, M. Ewing sarcoma family of tumors. In Pappo, AS, ed, Pediatric Bone and Soft Tissue Tumors. Berlin, Springer, 2006, pp. 181–218.CrossRefGoogle Scholar
Schmidt, D, Herrmann, C, Jurgens, H. Malignant peripheral neuroectodermal tumor and its necessary distinction from Ewing's sarcoma. A report from the Kiel Pediatric Tumor Registry. Cancer 1991;68:2251.3.0.CO;2-X>CrossRefGoogle ScholarPubMed
Parham, DM, Hijazi, Y, Steinberg, SM. Neuroectodermal differentiation in Ewing's sarcoma family of tumors does not predict tumor behavior. Hum Pathol 1999;30:911.CrossRefGoogle Scholar
Oberlin, O, Patte, C, Demeocq, R. The response to initial chemotherapy as a prognostic factor in localized Ewing's sarcoma. Eur J Cancer Clin Oncol 1985;21:463.CrossRefGoogle ScholarPubMed
Picci, P, Bohling, T, Bacci, G. Chemotherapy-induced tumor necrosis as a prognostic factor in localized Ewing's sarcoma of the extremities. J Clin Oncol 1997;15: 1553.CrossRefGoogle ScholarPubMed
Bacci, G, Longhi, A, Ferrari, S. Prognostic factors in non-metastatic Ewing's sarcoma of bone: an analysis of 579 patients treated at a single institution with adjuvant or neoadjuvant chemotherapy between 1972 and 1998. Acta Oncol 2006;45:469–475.CrossRefGoogle ScholarPubMed
Aparicio, J, Munarriz, B, Pastor, M. Long-term follow-up and prognostic factors in Ewing's sarcoma. A multivariate analysis of 116 patients from a single institution. Oncology 1998; 55:20.CrossRefGoogle ScholarPubMed
Wexler, LH, Delaney, TF, Tsokos, M. Ifosfamide and etoposide plus vincristine, doxorubicin, and cyclophosphamide for newly diagnosed Ewing's sarcoma family of tumors. Cancer 1996;78:901.3.0.CO;2-X>CrossRefGoogle ScholarPubMed
Jürgens, H, Exner, U, Gadner, H. Multidisciplinary treatment of primary Ewing's sarcoma of bone: a 6-year experience of a European cooperative trial. Cancer 1988;61:23.3.0.CO;2-M>CrossRefGoogle ScholarPubMed
Ozaki, T, Hillmann, A, Hoffmann, C. Significance of surgical margin on the prognosis of patients with Ewing's sarcoma – A report from the cooperative Ewing's sarcoma study. Cancer 1996;78:892.3.0.CO;2-P>CrossRefGoogle ScholarPubMed
Evans, RG. The four S's of Ewing's sarcoma. Int J Radiat Oncol Biol Phys 1991;21:1671.CrossRefGoogle ScholarPubMed
Thorner, P, Squire, J, Chilton-MacNeill, S. Is the EWS/FLI-1 fusion transcript specific for Ewing sarcoma and peripheral primitive neuroectodermal tumor? A report of four cases showing this transcript in a wider range of tumor types. Am J Pathol 1996;148:1125.Google Scholar
Jambhekar, NA, Bagwan, IN, Ghule, P. Comparative analysis of routine histology, immunohistochemistry, reverse transcriptase polymerase chain reaction, and fluorescence in situ hybridization in Ewing family of tumors. Arch Pathol Lab Med 2006;130:1813–1818.Google ScholarPubMed
Ahmed, AA, Nava, VE, Pham, T. Ewing sarcoma family of tumors in unusual sites: confirmation by rt-PCR. Pediatr Devel Pathol 2006;9:488–495.CrossRefGoogle ScholarPubMed
Dehner, LP. The evolution of the diagnosis and understanding of primitive and embryonic neoplasms in children: living through an epoch [review] . Mod Pathol 1998;11:669–685.Google Scholar
Wright, JH. Neurocytoma or neuroblastoma, a kind of tumor not generally recognized. J Exp Med 1910;12:556–561.CrossRefGoogle Scholar
Willis, RA. The Borderland of Embryology and Pathology, 2 ed. London, Butterworths, 1962.Google Scholar
Beckwith, JB, Martin, RF. Observations on the histopathology of neuroblastomas. J Pediatr Surg 1968;3:106–110.CrossRefGoogle ScholarPubMed
Joshi, VV, Cantor, AB, Altshuler, G, Larkin, EW, Neill, JSA, Shuster, JJ. Age-linked prognostic categorization based on a new histologic grading system of neuroblastomas: A clinicopathologic study of 211 cases from the Pediatric Oncology Group. Cancer 1992;69:2197–2211.3.0.CO;2-X>CrossRefGoogle ScholarPubMed
Beckwith, JB, Perrin, EV. In situ neuroblastoma: a contribution to the natural history of the neural crest tumors. Am J Pathol 1963;43:1089–1104.Google ScholarPubMed
Acharya, S, Jayabose, S, Kogan, SJ, Tugal, O, Beneck, D, Leslie, D. Prenatally diagnosed neuroblastoma [review]. Cancer 1997;80:304–310.3.0.CO;2-Y>CrossRefGoogle Scholar
Brodeur, GM, Moley, JF. Biology of tumors of the peripheral nervous system. Cancer Metastasis Rev 1991;10:321–333.CrossRefGoogle ScholarPubMed
Sano, H, Bonadio, J, Gerbing, RB, London, WB, Matthay, KK, Lukens, JN. International neuroblastoma pathology classification adds independent prognostic information beyond the prognostic contribution of age. Eur J Cancer 2006;42:1113–1119.CrossRefGoogle ScholarPubMed
Navarro, S, Amann, G, Beiske, K, Cullinane, CJ, d'Amore, ES, Gambini, C. Prognostic value of International Neuroblastoma Pathology Classification in localized resectable peripheral neuroblastic tumors: a histopathologic study of localized neuroblastoma European Study Group 94.01 Trial and Protocol. J Clin Oncol 2006;24:695–699.CrossRefGoogle Scholar
Dehner, LP. Peripheral and central primitive neuroectodermal tumors. A nosologic concept seeking a consensus. Arch Pathol Lab Med 1986;110:997.Google ScholarPubMed
Matthay, KK. Neuroblastoma: a clinical challenge and biologic puzzle. Ca: A Cancer Journal for Clinicians 1995;45:179.Google ScholarPubMed
Evans, AE, D'Angio, GJ, Propert, K. Prognostic factors in neuroblastoma. Cancer 1987;59:1853.3.0.CO;2-F>CrossRefGoogle ScholarPubMed
Kelly, DR, Joshi, VV. Neuroblastoma and related tumors. In Parham, DM(eds): Pediatric Neoplasia: Morphology and Biology, p. 105. Philadelphia, Lippincott-Raven, 1996.Google Scholar
Bolande, RP. A natural immune system in pregnancy serum lethal to human neuroblastoma cells: A possible mechanism of spontaneous regression. Perspect Pediatr Pathol 1992;16:120.Google ScholarPubMed
Graeve, JLA, Alarcon, PA, Sato, Y. Miliary pulmonary neuroblastoma: A risk of autologous bone marrow transplantation. Cancer 1988;62:2125.3.0.CO;2-1>CrossRefGoogle ScholarPubMed
Telles, NC, Rabson, AS, Pomeroy, TC. Ewing's sarcoma: an autopsy study. Cancer 1978;41:2321.3.0.CO;2-Y>CrossRefGoogle Scholar
Asami, T, Otabe, N, Wakabayashi, M, Kakihara, T, Uchiyama, M, Asami, K. Screening for neuroblastoma: A 9-year birth cohort-based study in Niigata, Japan. Acta Paediatr 1995;84:1173–1176.CrossRefGoogle ScholarPubMed
Brodeur, GM, Pritchard, J, Berthold, F. Revisions of the international criteria for neuroblastoma diagnosis, staging, and response to treatment. J Clin Oncol 1993;11:1466.CrossRefGoogle ScholarPubMed
Katzenstein, HM, Kent, PM, London, WB, Cohn, SL. Treatment and outcome of 83 children with intraspinal neuroblastoma: the Pediatric Oncology Group experience. J Clin Oncol 2001;19:1047–1055.CrossRefGoogle ScholarPubMed
Escobar, MA, Grosfeld, JL, Powell, RL, West, KW, Scherer, LR, Fallon, RJ. Long-term outcomes in patients with stage IV neuroblastoma. J Pediatr Surg 2006;41:377–381.CrossRefGoogle ScholarPubMed
Acharya, S, Jayabose, S, Kogan, SJ. Prenatally diagnosed neuroblastoma. Cancer 1997;80:304.3.0.CO;2-Y>CrossRefGoogle ScholarPubMed
Joshi, VV, Silverman, JF, Altshuler, G. Systematization of primary histopathologic and fine-needle aspiration cytologic features and description of unusual histopathologic features of neuroblastic tumors: A report from the Pediatric Oncology Group. Hum Pathol 1993;24:493.CrossRefGoogle ScholarPubMed
Umehara, S, Nakagawa, A, Matthay, KK, Lukens, JN, Seeger, RC, Stram, . Histopathology defines prognostic subsets of ganglioneuroblastoma, nodular. Cancer 2000;89:1150–1161.3.0.CO;2-7>CrossRefGoogle ScholarPubMed
Shimada, H, Ambros, IM, Dehner, LP. Terminology and morphologic criteria of neuroblastic tumors: recommendations by the International Neuroblastoma Pathology Committee. Cancer 1999;86:349.3.0.CO;2-Y>CrossRefGoogle ScholarPubMed
Shimada, H, Chatten, J, Newton, WA., et al. Histopathologic prognostic factors in neuroblastic tumors: definition of subtypes of ganglioneuroblastoma and an age-linked classification of neuroblastomas. J Natl Cancer Inst 1984;73:405.CrossRefGoogle ScholarPubMed
Shimada, H, Ambros, IM, Dehner, LP. The International Neuroblastoma Pathology Classification (the Shimada system). Cancer 1999;86:364.3.0.CO;2-7>CrossRefGoogle Scholar
Cozzutto, C, Carbone, A. Pleomorphic (anaplastic) neuroblastoma. Arch Pathol 1988;112:621.Google ScholarPubMed
Gonzalez-Crussi, F, Hsueh, W. Bilateral adrenal ganglioneuroblastoma with neuromelanin. Clinical and pathologic observations. Cancer 1988;61:1159.3.0.CO;2-S>CrossRefGoogle ScholarPubMed
Tsokos, M, Scarpa, S, Ross, RA. Differentiation of human neuroblastoma recapitulates neural crest development. Study of morphology, neurotransmitter enzymes, and extracellular matrix proteins. Am J Pathol 1987;128:484.Google ScholarPubMed
Balázs, M. Mixed pheochromocytoma and ganglioneuroma of the adrenal medulla: A case report with electron microscopic examination. Hum Pathol 1988;19:1352.CrossRefGoogle ScholarPubMed
Blatt, J, Gula, MJ, Orlando, SJ. Indolent course of advanced neuroblastoma in children older than 6 years at diagnosis. Cancer 1995;76:890.3.0.CO;2-X>CrossRefGoogle ScholarPubMed
Mierau, GW, Berry, PJ, Malott, RL. Appraisal of the comparative utility of immunohistochemistry and electron microscopy in the diagnosis of childhood round cell tumors. Ultrastruct Pathol 1996;20:507.CrossRefGoogle Scholar
Thiele, CJ. Pediatric peripheral neuroectodermal tumors, oncogenes, and differentiation. Cancer Invest 1990;8:629.CrossRefGoogle Scholar
Dias, P, Kumar, P, Marsden, HB. N-myc gene is amplified in alveolar rhabdomyosarcomas (RMS) but not in embryonal RMS. Int J Cancer 1990;45:593.CrossRefGoogle ScholarPubMed
Driman, D, Thorner, PS, Greenberg, ML. MYCN gene amplification in rhabdomyosarcoma. Cancer 1994;73:2231.3.0.CO;2-E>CrossRefGoogle ScholarPubMed
Leader, M, Collins, M, Patel, J. Antineuron specific enolase staining reactions in sarcomas and carcinomas: its lack of neuroendocrine specificity. J Clin Pathol 1986;39:1186.CrossRefGoogle ScholarPubMed
Brodeur, GM, Green, AA, Hayes, FA. Cytogenetic features of human neuroblastomas and cell lines. Cancer Res 1981;41:4678.Google ScholarPubMed
Maris, JM, Matthay, KK. Molecular biology of neuroblastoma. J Clin Oncol 1999;17:2264.CrossRefGoogle ScholarPubMed
Caren, H, Ejeskar, K, Fransson, S, Hesson, L, Latif, F, Sjoberg, RM. A cluster of genes located in 1p36 are down-regulated in neuroblastomas with poor prognosis, but not due to CpG island methylation. Mol Cancer 2005;4:10.CrossRefGoogle Scholar
Fransson, S, Martinsson, T, Ejeskar, K. Neuroblastoma tumors with favorable and unfavorable outcomes: Significant differences in mRNA expression of genes mapped at 1p36.2. Genes Chromosomes Cancer 2007;46:45–52.CrossRefGoogle ScholarPubMed
Iehara, T, Hosoi, H, Akazawa, K, Matsumoto, Y, Yamamoto, K, Suita, S. MYCN gene amplification is a powerful prognostic factor even in infantile neuroblastoma detected by mass screening. Br J Cancer 2006;94:1510–1515.CrossRefGoogle ScholarPubMed
Shapiro, DN, Valentine, MB, Rowe, ST. Detection of N-myc gene amplification by fluorescence in situ hybridization. Diagnostic utility for neuroblastoma. Am J Pathol 1993;142:1339.Google ScholarPubMed
Bhargava, R, Oppenheimer, O, Gerald, W, Jhanwar, SC, Chen, B. Identification of MYCN gene amplification in neuroblastoma using chromogenic in situ hybridization (CISH): an alternative and practical method. Diagn Mol Pathol 2005;14:72–76.CrossRefGoogle ScholarPubMed
Look, AT, Hayes, FA, Nitschke, R. Cellular DNA content as a predictor of response to chemotherapy in infants with unresectable neuroblastoma. N Engl J Med 1984;311:231.CrossRefGoogle ScholarPubMed
Look, AT, Hayes, FA, Shuster, JJ. Clinical relevance of tumor cell ploidy and N-myc gene amplification in childhood neuroblastoma: A Pediatric Oncology Group study. J Clin Oncol 1991;9:581.CrossRefGoogle ScholarPubMed
Tanaka, T, Sugimoto, T, Sawada, T. Prognostic discrimination among neuroblastomas according to Ha-ras/trk A gene expression: a comparison of the profiles of neuroblastomas detected clinically and those detected through mass screening. Cancer 1998;83:1626.3.0.CO;2-Y>CrossRefGoogle ScholarPubMed
Maitra, A, Yashima, K, Rathi, A. The RNA component of telomerase as a marker of biologic potential and clinical outcome in childhood neuroblastic tumors. Cancer 1999;85:741.3.0.CO;2-6>CrossRefGoogle ScholarPubMed
Ohali, A, Avigad, S, Ash, S, Goshen, Y, Luria, D, Feinmesser, M. Telomere length is a prognostic factor in neuroblastoma. Cancer 2006;107:1391–1399.CrossRefGoogle ScholarPubMed
Brodeur, GM, Azar, C, Brother, M. Neuroblastoma: Effect of genetic factors on prognosis and treatment. Cancer 1992;70 Suppl.:1685.3.0.CO;2-H>CrossRefGoogle ScholarPubMed
Elimam, NA, Atra, AA, Fayea, NY, Al-Asaad, TG, Khattab, TM, Al-Sulami, GA. Stage 4S neuroblastoma, a disseminated tumor with excellent outcome. Saudi Med J 2006;27:1734–1736.Google ScholarPubMed
Navarro, S, Amann, G, Beiske, K, Cullinane, CJ, d'Amore, ES, Gambini, C. Prognostic value of International Neuroblastoma Pathology Classification in localized resectable peripheral neuroblastic tumors: a histopathologic study of localized neuroblastoma European Study Group 94.01 Trial and Protocol. J Clin Oncol 2006;24:695–699.CrossRefGoogle Scholar
Burgues, O, Navarro, S, Noguera, R, Pellin, A, Ruiz, A, Castel, V. Prognostic value of the International Neuroblastoma Pathology Classification in Neuroblastoma (Schwannian stroma-poor) and comparison with other prognostic factors: a study of 182 cases from the Spanish Neuroblastoma Registry. Virchows Arch 2006;449:410–420.CrossRefGoogle ScholarPubMed
Ordonez, NG. Desmoplastic small round cell tumor: I: a histopathologic study of 39 cases with emphasis on unusual histological patterns. Am J Surg Pathol 1998;22:1303.CrossRefGoogle ScholarPubMed
Gerald, WL, Miller, HK, Battifora, H. Intra-abdominal desmoplastic small round-cell tumor: report of 19 cases of a distinctive type of high-grade polyphenotypic malignancy affecting young individuals. Am J Surg Pathol 1991;15:499.CrossRefGoogle Scholar
Sawyer, JR, Tryka, AF, Lewis, JM. A novel reciprocal chromosome translocation t(11;22)(p13; q12) in an intraabdominal desmoplastic small round-cell tumor. Am J Surg Pathol 1992;16:411–416.CrossRefGoogle Scholar
Kretschmar, CS, Colbach, C, Bhan, I. Desmoplastic small cell tumor: a report of three cases and a review of the literature. J Pediatr Hematol Oncol 1996;18:293.CrossRefGoogle Scholar
Ordonez, NG, El-Naggar, AK, Ro, JY. Intra-abdominal desmoplastic small cell tumor: a light microscopic, immunocytochemical, ultrastructural, and flow cytometric study. Hum Pathol 1993;24:850.CrossRefGoogle ScholarPubMed
Kawano, N, Inayama, Y, Nagashima, Y, Miyagi, Y, Uemura, H, Saitoh, K. Desmoplastic small round-cell tumor of the paratesticular region: report of an adult case with demonstration of EWS and WT1 gene fusion using paraffin-embedded tissue. Mod Pathol 1999;12:729–734.Google ScholarPubMed
Adsay, V, Cheng, J, Athanasian, E. Primary desmoplastic small cell tumor of soft tissues and bone of the hand. Am J Surg Pathol 1999;23:1408.CrossRefGoogle ScholarPubMed
Bian, Y, Jordan, AG, Rupp, M. Effusion cytology of desmoplastic small round cell tumor of the pleura: A case report. Acta Cytol 1993;37:77.Google ScholarPubMed
Tison, V, Cerasoli, S, Morigi, F. Intracranial desmoplastic small-cell tumor – Report of a case. Am J Surg Pathol 1996;20:112.CrossRefGoogle ScholarPubMed
Karavitakis, EM, Moschovi, M, Stefanaki, K, Karamolegou, K, Dimitriadis, E, Pandis, N. Desmoplastic small round cell tumor of the pleura. Pediatr Blood Cancer 2007;49:335–338.CrossRefGoogle ScholarPubMed
Wang, LL, Perlman, EJ, Vujanic, GM, Zuppan, C, Brundler, MA, Cheung, CR. Desmoplastic small round cell tumor of the kidney in childhood. Am J Surg Pathol 2007;31:576–584.CrossRefGoogle ScholarPubMed
Finke, NM, Lae, ME, Lloyd, RV, Gehani, SK, Nascimento, AG. Sinonasal desmoplastic small round cell tumor: a case report. Am J Surg Pathol 2002;26:799–803.CrossRefGoogle ScholarPubMed
Schmidt, D, Koster, E, Harms, D. Intraabdominal desmoplastic small-cell tumor with divergent differentiation: clinicopathological findings and DNA ploidy. Med Pediatr Oncol 1994;22:97.CrossRefGoogle ScholarPubMed
Zhang, J, Dalton, J, Fuller, C. Epithelial marker-negative desmoplastic small round cell tumor with atypical morphology: definitive classification by fluorescence in situ hybridization. Arch Pathol Lab Med 2007;131:646–649.Google ScholarPubMed
Ordonez, NG. Desmoplastic small round cell tumor: II: an ultrastructural and immunohistochemical study with emphasis on new immunohistochemical markers. Am J Surg Pathol 1998;22:1314.CrossRefGoogle ScholarPubMed
Trupiano, JK, Machen, SK, Barr, FG. Cytokeratin-negative desmoplastic small round cell tumor: a report of two cases emphasizing the utility of reverse transcriptase-polymerase chain reaction. Mod Pathol 1999;12:849.Google ScholarPubMed
Sawyer, JR, Tryka, AF, Lewis, JM. A novel reciprocal chromosome translocation t(11;22)(p13; q12) in an intraabdominal desmoplastic small round-cell tumor. Am J Surg Pathol 1992;16:411.CrossRefGoogle Scholar
Ladanyi, M, Gerald, W. Fusion of the EWS and WT1 genes in the desmoplastic small round cell tumor. Cancer Res 1994;54:2837.Google ScholarPubMed
Park, S, Schalling, M, Bernard, A. The Wilms tumour gene WT1 is expressed in murine mesoderm-derived tissues and mutated in a human mesothelioma. Nature Genet 1993;4:415.CrossRefGoogle Scholar
Walker, C, Rutten, F, Yuan, X. Wilms' tumor suppressor gene expression in rat and human mesothelioma. Cancer Res 1994;54:3101.Google ScholarPubMed
Roberts, DJ, Haber, D, Sklar, J. Extrarenal Wilms' tumors. A study of their relationship with classical renal Wilms' tumor using expression of WT1 as a molecular marker. Lab Invest 1993;68:528.Google ScholarPubMed
Lee, SB, Kolquist, KA, Nichols, K. The EWS-WT1 translocation product induces PDGFA in desmoplastic small round-cell tumour. Nature Genet 1997;17:309.CrossRefGoogle ScholarPubMed
Alava, E, Ladanyi, M, Rosai, J. Detection of chimeric transcripts in desmoplasmic small round cell tumor and related developmental tumors by reverse transcriptase polymerase chain reaction – A specific diagnostic assay. Am J Pathol 1995;147:1584.Google Scholar
Argatoff, LH, O'Connell, JX, Mathers, JA. Detection of the EWS/WT1 gene fusion by reverse transcriptase-polymerase chain reaction in the diagnosis of intra-abdominal desmoplastic small round cell tumor. Am J Surg Pathol 1996;20:406.CrossRefGoogle Scholar
Katz, RL, Quezado, M, Senderowicz, AM. An intra-abdominal small round cell neoplasm with features of primitive neuroectodermal and desmoplastic round cell tumor and a EWS/FLI-1 fusion transcript. Hum Pathol 1997;28:502.CrossRefGoogle Scholar
Gardner, LJ, Ayala, AG, Monforte, HL, Dunphy, CH. Ewing sarcoma/peripheral primitive neuroectodermal tumor: adult abdominal tumors with an Ewing sarcoma gene rearrangement demonstrated by fluorescence in situ hybridization in paraffin sections. Appl Immunohistochem Mol Morphol 2004;12:160–165.CrossRefGoogle ScholarPubMed
Ordi, J, AE, , Torne, A, Mellado, B, Pardo-Mindan, J, Iglesias, X. Intraabdominal desmoplastic small round cell tumor with EWS/ERG fusion transcript. Am J Surg Pathol 1998;22:1026–1032.CrossRefGoogle ScholarPubMed
Alaggio, R, Rosolen, A, Sartori, F, Leszl, A, d'Amore, ES, Bisogno, G. Spindle cell tumor with EWS-WT1 transcript and a favorable clinical course: a variant of DSCT, a variant of leiomyosarcoma, or a new entity? Report of 2 pediatric cases. Am J Surg Pathol 2007;31:454–459.CrossRefGoogle ScholarPubMed
Amato, RJ, Ellerhorst, JA, Ayala, AG. Intraabdominal desmoplastic small cell tumor. Report and discussion of five cases. Cancer 1996;78:845.3.0.CO;2-U>CrossRefGoogle ScholarPubMed
Saab, R, Khoury, JD, Krasin, M, Davidoff, AM, Navid, F. Desmoplastic small round cell tumor in childhood: the St. Jude Children's Research Hospital experience. Pediatr Blood Cancer 2007;49:274–279.CrossRefGoogle ScholarPubMed
Coppes, MJ, Wilson, PCG, Weitzman, S. Extrarenal Wilms' tumor: Staging, treatment, and prognosis. J Clin Oncol 1991;9:167.CrossRefGoogle Scholar
Parham, DM, Weeks, DA, Beckwith, JB. The clinicopathologic spectrum of putative extrarenal rhabdoid tumors: an analysis of 42 cases studies with immunohistochemistry and/or electron microscopy. Am J Surg Pathol 1994;18: 1010.CrossRefGoogle ScholarPubMed
Leong, FJ, Leong, AS. Malignant rhabdoid tumor in adults – heterogenous tumors with a unique morphological phenotype. Pathol Res Pract 1996;192:796.CrossRefGoogle ScholarPubMed
White, FV, Dehner, LP, Belchis, DA. Congenital disseminated malignant rhabdoid tumor: distinct clinicopathologic entity demonstrating abnormalities of chromosome 22q11. Am J Surg Pathol 1999;23:249.CrossRefGoogle ScholarPubMed
Simons, J, Teshima, I, Zielenska, M. Analysis of chromosome 22q as an aid to the diagnosis of rhabdoid tumor: a case report. Am J Surg Pathol 1999;23:982.CrossRefGoogle Scholar
Beckwith, JB, Palmer, NF. Histopathology and prognosis of Wilms tumors: results from the First National Wilms' Tumor Study. Cancer 1978;41:1937.3.0.CO;2-U>CrossRefGoogle ScholarPubMed
Balaton, AJ, Vaury, P, Videgrain, M. Paravertebral malignant rhabdoid tumor in an adult. A case report with immunocytochemical study. Pathol Res Pract 1987;182:713.Google Scholar
Dabbs, DJ, Park, HK. Malignant rhabdoid skin tumor: an uncommon primary skin neoplasm. Ultrastructural and immunohistochemical analysis. J Cutan Pathol 1988;15:109.CrossRefGoogle ScholarPubMed
Tsuneyoshi, M, Daimaru, Y, Hashimoto, H. Malignant soft tissue neoplasms with the histologic features of renal rhabdoid tumors: an ultrastructural and immunohistochemical study. Hum Pathol 1985;16:1235.CrossRefGoogle ScholarPubMed
Gonzalez-Crussi, F, Goldschmidt, RA, Hsueh, W. Infantile sarcoma with intracytoplasmic filamentous inclusions: distinctive tumor of possible histiocytic origin. Cancer 1982;49:2365.3.0.CO;2-I>CrossRefGoogle ScholarPubMed
Lemos, LB, Hamoudi, AB. Malignant thymic tumor in an infant (malignant histiocytoma). Arch Pathol Lab Med 1978;102:84.Google Scholar
Hajdu, SI. Pathology of Soft Tissue Tumors. Philadelphia, Lea and Febiger, 1979.Google Scholar
Kodet, R, Newton, WA., Sachs, N. Rhabdoid tumors of soft tissues: A clinicopathologic study of 26 cases enrolled in the Intergroup Rhabdomyosarcoma Study. Hum Pathol 1991;22:674.CrossRefGoogle ScholarPubMed
Tsuneyoshi, M, Daimaru, Y, Hashimoto, H. The existence of rhabdoid cells in specified soft tissue sarcomas: histopathological, ultrastructural and immunohistochemical evidence. Virchows Arch [A] 1987;411:509.CrossRefGoogle ScholarPubMed
Kodet, R, Newton, WA., Hamoudi, AB. Rhabdomyosarcomas with intermediate-filament inclusions and features of rhabdoid tumors: Light microscopic and immunohistochemical study. Am J Surg Pathol 1991;15:257.CrossRefGoogle ScholarPubMed
Tsokos, M, Kouraklis, G, Chandra, RS. Malignant rhabdoid tumor of the kidney and soft tissues: evidence for a diverse morphological and immunocytochemical phenotype. Arch Pathol Lab Med 1989;113:115.Google ScholarPubMed
Fanburg-Smith, JC, Hengge, M, Hengge, UR, Smith, JS, Miettinen, M. Extrarenal rhabdoid tumors of soft tissue: a clinicopathologic and immunohistochemical study of 18 cases. Ann Diagn Pathol 1998;2:351–362.CrossRefGoogle ScholarPubMed
Rorke, LB, Packer, RJ, Biegel, JA. Central nervous system atypical teratoid/rhabdoid tumors of infancy and childhood: Definition of an entity. J Neurosurg 1996;85:56.CrossRefGoogle ScholarPubMed
Wick, MR, Ritter, JH, Dehner, LP. Malignant rhabdoid tumors: a clinicopathologic review and conceptual discussion [review]. Semin Diagn Pathol 1995;12:233–248.Google Scholar
Haas, JE, Palmer, NF, Weinberg, AG, Beckwith, JB. Ultrastructure of malignant rhabdoid tumor of the kidney. A distinctive renal tumor of children. Hum Pathol 1981;12:646–657.CrossRefGoogle ScholarPubMed
Chung, CJ, Cammoun, D, Munden, M. Rhabdoid tumor of the kidney presenting as an abdominal mass in a newborn. Pediatr Radiol 1990;20:562.CrossRefGoogle Scholar
Kent, AL, Mahoney, DH, Gresik, MV. Malignant rhabdoid tumor of the extremity. Cancer 1987;60:1056.3.0.CO;2-Q>CrossRefGoogle ScholarPubMed
Dominey, A, Paller, AS, Gonzalez-Crussi, F. Congenital rhabdoid sarcoma with cutaneous metastases. J Am Acad Dermatol 1990;22:969.CrossRefGoogle ScholarPubMed
Perez-Atayde, AR, Newbury, R, Fletcher, JA. Congenital “neurovascular hamartoma” of the skin. A possible marker of malignant rhabdoid tumor. Am J Surg Pathol 1994;18:1030.CrossRefGoogle ScholarPubMed
Jayabose, S, Iqbal, K, Newman, L. Hypercalcemia in childhood renal tumors. Cancer 1988;61:788.3.0.CO;2-H>CrossRefGoogle ScholarPubMed
Mayes, LC, Kasselberg, AG, Roloff, JS. Hypercalcemia associated with immunoreactive parathyroid hormone in a malignant rhabdoid tumor of the kidney (rhabdoid Wilms' tumor). Cancer 1984; 54: 882.3.0.CO;2-N>CrossRefGoogle Scholar
Rousseau-Merck, MF, Nogues, C, Roth, A. Hypercalcemic infantile renal tumors: morphological, clinical, and biological heterogeneity. Pediatr Pathol 1985;3:155.CrossRefGoogle ScholarPubMed
Bonnin, JM, Rubinstein, LJ, Palmer, NF. The association of embryonal tumours originating in the kidney and in the brain. Cancer 1984;54:2137.3.0.CO;2-D>CrossRefGoogle Scholar
Chang, CH, Ramirez, N, Sakr, WA. Primitive neuroectodermal tumor of the brain associated with malignant rhabdoid tumor of the liver: a histologic, immunohistochemical, and electron microscopic study. Pediatr Pathol 1989;9:307.CrossRefGoogle ScholarPubMed
Weeks, DA, Beckwith, JB, Mierau, GW. Rhabdoid tumor of kidney: A report of 111 cases from the National Wilms' Tumor Study Pathology Center. Am J Surg Pathol 1989;13:439.CrossRefGoogle ScholarPubMed
Suzuki, A, Ohta, S, Shimada, M. Gene expression of malignant rhabdoid tumor cell lines by reverse transcriptase-polymerase chain reaction. Diagn Mol Pathol 1997;6:326.CrossRefGoogle ScholarPubMed
Hoot, AC, Russo, P, Judkins, AR, Perlman, EJ, Biegel, JA. Immunohistochemical analysis of hSNF5/INI1 distinguishes renal and extra-renal malignant rhabdoid tumors from other pediatric soft tissue tumors. Am J Surg Pathol 2004;28:1485–1491.CrossRefGoogle ScholarPubMed
Fuller, CE, Pfeifer, J, Humphrey, P, Bruch, , Dehner, LP, Perry, A. Chromosome 22q dosage in composite extrarenal rhabdoid tumors: clonal evolution or a phenotypic mimic?Hum Pathol 2001;32:1102–1108.CrossRefGoogle ScholarPubMed
Wyatt-Ashmead, J, Kleinschmidt-DeMasters, B, Mierau, GW, Malkin, D, Orsini, E, McGavran, L. Choroid plexus carcinomas and rhabdoid tumors: phenotypic and genotypic overlap. Pediatr Dev Pathol 2001;4:545–549.CrossRefGoogle ScholarPubMed
Oda, Y, Tsuneyoshi, M. Extrarenal rhabdoid tumors of soft tissue: clinicopathological and molecular genetic review and distinction from other soft-tissue sarcomas with rhabdoid features. Pathol Int 2006;56:287–295.CrossRefGoogle ScholarPubMed
Modena, P, Lualdi, E, Facchinetti, F, Galli, L, Teixeira, MR, Pilotti, S. SMARCB1/INI1 tumor suppressor gene is frequently inactivated in epithelioid sarcomas. Cancer Res 2005;65:4012–4019.CrossRefGoogle ScholarPubMed
Donner, LR, Wainwright, LM, Zhang, F, Biegel, JA. Mutation of the INI1 gene in composite rhabdoid tumor of the endometrium. Hum Pathol 2007;38:935CrossRefGoogle ScholarPubMed
Cho, YM, Choi, J, Lee, OJ, Lee, HI, Han, DJ, Ro, JY. SMARCB1/INI1 missense mutation in mucinous carcinoma with rhabdoid features. Pathol Int 2006;56:702–706.CrossRefGoogle ScholarPubMed
Douglass, EC, Valentine, M, Rowe, ST. Malignant rhabdoid tumor: a highly malignant childhood tumor with minimal karyotypic changes. Genes Chromosomes Cancer 1990;2:210.CrossRefGoogle ScholarPubMed
Biegel, JA, Rorke, LB, Emanuel, BS. Monosomy 22 in rhabdoid or atypical teratoid tumors of the brain. N Engl J Med 1989;321:906.Google ScholarPubMed
Bhattacharjee, MB, Armstrong, DD, Vogel, H. Cytogenetic analysis of 120 primary pediatric brain tumors and literature review. Cancer Genet Cytogenet 1997;97:39.CrossRefGoogle ScholarPubMed
Schofield, , Beckwith, JB, Sklar, J. Loss of heterozygosity at chromosome regions 22q11–12 and 11p15.5 in renal rhabdoid tumors. Genes Chromosomes Cancer 1996;15:10.3.0.CO;2-7>CrossRefGoogle ScholarPubMed
Biegel, JA, Allen, CS, Kawasaki, K. Narrowing the critical region for a rhabdoid tumor locus in 22q11. Genes Chromosomes Cancer 1996;16:94.3.0.CO;2-Y>CrossRefGoogle ScholarPubMed
Versteege, I, Sevenet, N, Lange, J. Truncating mutations of hSNF5/INI1 in aggressive paediatric cancer. Nature 1998;394:203.CrossRefGoogle ScholarPubMed
Biegel, JA. Molecular genetics of atypical teratoid/rhabdoid tumor. Neurosurg Focus 2006;20:E11.CrossRefGoogle ScholarPubMed
Janson, K, Nedzi, , David, O, Schorin, M, Walsh, JW, Bhattacharjee, M. Predisposition to atypical teratoid/rhabdoid tumor due to an inherited INI1 mutation. Pediatr Blood Cancer 2006;47:279–284.CrossRefGoogle Scholar
White, FV, Dehner, LP, Belchis, DA, Conard, K, Davis, MM, Stocker, JT. Congenital disseminated malignant rhabdoid tumor: a distinct clinicopathologic entity demonstrating abnormalities of chromosome 22q11 [review]. Am J Surg Pathol 1999;23:249–256.CrossRefGoogle Scholar
Garvin, AJ, Re, GG, Tarnowski, BI. The G401 cell line, utilized for studies of chromosomal changes in Wilms' tumor, is derived from a rhabdoid tumor of the kidney. Am J Pathol 1993;142:375.Google ScholarPubMed
Reid, LH, Davies, C, Cooper, PR. A 1-Mb physical map and PAC contig of the imprinted domain in 11p15.5 that contains TAPA1 and the BWSCR1/WT2 region. Genomics 1997;43:366.CrossRefGoogle ScholarPubMed
Sabbioni, S, Barbanti-Brodano, G, Croce, CM. GOK: a gene at 11p15 involved in rhabdomyosarcoma and rhabdoid tumor development. Cancer Res 1997;57:4493.Google ScholarPubMed
Fruhwald, MC, Hasselblatt, M, Wirth, S, Kohler, G, Schneppenheim, R, Subero, JI. Non-linkage of familial rhabdoid tumors to SMARCB1 implies a second locus for the rhabdoid tumor predisposition syndrome. Pediatr Blood Cancer 2006;47:273–278.CrossRefGoogle ScholarPubMed
Bourdeaut, F, Freneaux, P, Thuille, B, Lellouch-Tubiana, A, Nicolas, A, Couturier, J. hSNF5/INI1-deficient tumours and rhabdoid tumours are convergent but not fully overlapping entities. J Pathol 2007;211:323–330.CrossRefGoogle Scholar
Bittesini, L, Dei Tos, AP, Fletcher, CDM. Metastatic malignant melanoma showing a rhabdoid phenotype: Further evidence of a non-specific histological pattern. Histopathology 1992;20:167.CrossRefGoogle ScholarPubMed
Guillou, L, Wadden, C, Coindre, JM. “Proximal-type” epithelioid sarcoma, a distinctive aggressive neoplasm showing rhabdoid features. Clinicopathologic, immunohistochemical, and ultrastructural study of a series. Am J Surg Pathol 1997;21:130.CrossRefGoogle ScholarPubMed
Raney, RB, Asmar, L, Newton, WA. Ewing's sarcoma of soft tissues in childhood: a report from the Intergroup Rhabdomyosarcoma Study, 1972–1991. J Clin Oncol 1997;15:574–582.CrossRefGoogle Scholar
Rud, RP, Reiman, HM, Pritchard, DJ. Extraosseous Ewing's sarcoma, a study of 42 cases. Cancer 1989;64:1548–1553.3.0.CO;2-W>CrossRefGoogle ScholarPubMed
Parkin, DM, Kramároᾓ, E, Draper, GJ. [eds] International Incidence of Childhood Cancer, Vol. II. IARC Scientific Publications, Lyon, 1998, Table 5.27.Google Scholar
London, WB, Look, AT, Brodeur, GM, Altmiller, DH, Thorner, PS. Natural history and biology of stage A neuroblastoma: a Pediatric Oncology Group Study. J Pediatr Hematol Oncol 2000;22:197–205.Google Scholar
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.CrossRefGoogle ScholarPubMed

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×