Skip to main content Accessibility help
×
Hostname: page-component-7479d7b7d-jwnkl Total loading time: 0 Render date: 2024-07-10T09:35:29.666Z Has data issue: false hasContentIssue false

77 - Pediatric solid tumors: embryonal cell oncogenesis

from Part 3.7 - Molecular pathology: pediatric solid tumors

Published online by Cambridge University Press:  05 February 2015

Jeffrey A. Toretsky
Affiliation:
Departments of Oncology and Pediatrics, Georgetown University, Washington,DC, USA
Aerang Kim
Affiliation:
Department of Pediatrics, Children’s National Medical Center, George Washington University, Washington, DC, USA
Edward P. Gelmann
Affiliation:
Columbia University, New York
Charles L. Sawyers
Affiliation:
Memorial Sloan-Kettering Cancer Center, New York
Frank J. Rauscher, III
Affiliation:
The Wistar Institute Cancer Centre, Philadelphia
Get access

Summary

Solid tumors that occur in children and adolescents are rare, but have significantly contributed to the understanding of cancer biology. A famous example is Knudson's epidemiologic study of retinoblastoma patients that predicted the role of tumor-suppressor genes in human cancers (1–3). Pediatric solid-tumor investigations were critical to the discovery of pathognomonic chromosomal translocations, which classified difficult-to-diagnose, small round blue-cell tumors, and led to potential targets for novel therapeutics (4). This chapter will provide synopses of pediatric solid tumors where mechanistic insights have provided an understanding of pathobiology and targets for therapy.

Ewing sarcoma

Ewing sarcoma (ES) are comprised of highly malignant neoplasms of bone and soft tissue occurring in children, adolescents, and young adults. ES are defined by the characteristic chromosomal translocation t(11:22) and its fusion-protein product EWS-FLI1. The translocation, or a related variant, occurs in 95% of tumors (5) between the central exons of the EWSR1 gene (EWing Sarcoma breakpoint region 1; chromosome 22) to the central exons of an ets family gene combining it with either FLI1 (Friend Leukemia Integration 1; chromosome 11) or ERG (v-ets erythroblastosis virus E26 oncogene homolog; chromosome 21) t(11;22) and t(21;22), respectively. Additional chromosomal abnormalities associated with disease progression have also been identified (6,7). While rare, deletions of p53 or p16 portend a poor outcome and thus may be linked to the pathophysiology, potentially through tumor resistance to therapy (8).

Type
Chapter
Information
Molecular Oncology
Causes of Cancer and Targets for Treatment
, pp. 826 - 835
Publisher: Cambridge University Press
Print publication year: 2013

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

Friend, SH, Bernards, R, Rogelj, S, et al. A human DNA segment with properties of the gene that predisposes to retinoblastoma and osteosarcoma. Nature 1986;323:643–6.CrossRef
Lee, WH, Bookstein, R, Hong, F, et al. Human retinoblastoma susceptibility gene: cloning, identification, and sequence. Science 1987;235:1394–9.CrossRef
Knudson, AG Mutation and cancer: statistical study of retinoblastoma. Proceedings of the National Academy of Sciences USA 1971;68:820–3.CrossRef
Helman, LJ, Meltzer, P. Mechanisms of sarcoma development. Nature Reviews Cancer 2003;3:685–94.CrossRef
Delattre, O, Zucman, J, Melot, T, et al. The Ewing family of tumors-a subgroup of small-round-cell tumors defined by specific chimeric transcripts. New England Journal of Medicine 1994;331:294–9.CrossRefGoogle ScholarPubMed
Maurici, D, Perez-Atayde, A, Grier, HE, et al. Frequency and implications of chromosome 8 and 12 gains in Ewing sarcoma. Cancer Genetics and Cytogenetics 1998;100:106–10.CrossRef
Ozaki, T, Paulussen, M, Poremba, C, et al. Genetic imbalances revealed by comparative genomic hybridization in Ewing tumors. Genes, Chromosomes and Cancer 2001;32:164–71.CrossRef
Huang, HY, Illei, PB, Zhao, Z, et al. Ewing sarcomas with p53 mutation or p16/p14ARF homozygous deletion: a highly lethal subset associated with poor chemoresponse. Journal of Clinical Oncology 2005;23:548–58.CrossRefGoogle ScholarPubMed
Riggi, N, Suva, ML, Stamenkovic, I. Ewing's sarcoma origin: from duel to duality. Expert Review of Anticancer Therapy 2009;9:1025–30.CrossRef
Kovar, H. Context matters: the hen or egg problem in Ewing's sarcoma. Seminars in Cancer Biology 2005;15:189–96.CrossRef
Tanaka, K, Iwakuma, T, Harimaya, K, Sato, H, Iwamoto, Y. EWS-Fli1 antisense oligodeoxynucleotide inhibits proliferation of human Ewing's sarcoma and primitive neuroectodermal tumor cells. Journal of Clinical Investigation 1997;99:239–47.CrossRefGoogle ScholarPubMed
Toretsky, JA, Connell, Y, Neckers, L, Bhat, NK. Inhibition of EWS-FLI-1 fusion protein with antisense oligodeoxynucleotides. Journal of Neurooncology 1997;31:9–16.CrossRefGoogle ScholarPubMed
Ouchida, M, Ohno, T, Fujimura, Y, Rao, VN, Reddy, ES. Loss of tumorigenicity of Ewing's sarcoma cells expressing antisense RNA to EWS-fusion transcripts. Oncogene 1995;11:1049–54.
Kovar, H, Aryee, DN, Jug, G, et al. EWS/FLI-1 antagonists induce growth inhibition of Ewing tumor cells in vitro. Cell Growth and Differentiation 1996;7:429–37.
Maksimenko, A, Lambert, G, Bertrand, JR, et al. Therapeutic potentialities of EWS-Fli-1 mRNA-targeted vectorized antisense oligonucleotides. Annals of the New York Academy of Science 2003;1002:72–7.CrossRef
Hu-Lieskovan, S, Heidel, JD, Bartlett, DW, Davis, ME, Triche, TJ. Sequence-specific knockdown of EWS-FLI1 by targeted, nonviral delivery of small interfering RNA inhibits tumor growth in a murine model of metastatic Ewing's sarcoma. Cancer Research. 2005;65:8984–92.CrossRef
Toretsky, JA, Kalebic, T, Blakesley, V, LeRoith, D, Helman, LJ. The insulin-like growth factor-I receptor is required for EWS/FLI-1 transformation of fibroblasts. Journal of Biological Chemistry 1997;272:30 822–7.CrossRefGoogle ScholarPubMed
Erkizan, HV, Kong, Y, Merchant, M, et al. A small molecule blocking oncogenic protein EWS-FLI1 interaction with RNA helicase A inhibits growth of Ewing's sarcoma. Nature Medicine 2009;15:750–6.CrossRef
Uren, A, Toretsky, JA. Ewing's sarcoma oncoprotein EWS-FLI1: the perfect target without a therapeutic agent. Future Oncology 2005;1:521–8.CrossRef
Maris JM, MC, Gorlick, R, Kolb, EA, et al. Initial testing of the aurora kinase a inhibitor MLN8237 by the Pediatric Preclinical Testing Program (PPTP). Pediatric Blood amd Cancer 2010;55:26–34.
Houghton, PJ, Morton, CL, Gorlick, R, et al. Initial testing of a monoclonal antibody (IMC-A12) against IGF-1R by the pediatric preclinical testing program. Pediatric Blood and Cancer 2010;54:921–6.
Manara, MC, Landuzzi, L, Nanni, P, et al. Preclinical in vivo study of new insulin-like growth factor-I receptor–specific inhibitor in Ewing's sarcoma. Clinical Cancer Research 2007;13:1322–30.CrossRef
Pappo, AS, Patel, SR, Crowley, J, et al. R1507, a monoclonal antibody to the insulin-like growth factor 1 receptor, in patients with recurrent or refractory Ewing sarcoma family of tumors: results of a Phase II Sarcoma Alliance for Research through Collaboration study. Journal of Clinical Oncology 2011;29:4541–7.CrossRefGoogle ScholarPubMed
Juergens, H, Daw, NC, Geoerger, B, et al. Preliminary efficacy of the anti-insulin-like growth factor type 1 receptor antibody figitumumab in patients with refractory Ewing sarcoma. Journal of Clinical Oncology 2011;29:4534–40.CrossRefGoogle ScholarPubMed
Tap, WD, Demetri, G, Barnette, P, et al. Phase II study of ganitumab, a fully human anti-type-1 insulin-like growth factor receptor antibody, in patients with metastatic Ewing family tumors or desmoplastic small round cell tumors. Journal of Clinical Oncology 2012;30:1849–56.CrossRefGoogle ScholarPubMed
O’Neill, A, Shah, N, Zitomersky, N, et al. Insulin-like growth factor 1 receptor (IGF-1R) as a therapeutic target in Ewing sarcoma: lack of consistent up-regulation or recurrent mutation and a review of clinical trial literature. Sarcoma 2013:450578.
Tomlinson, GE, Douglass, EC, Pollock, BH, Finegold, MJ, Schneider, NR. Cytogenetic evaluation of a large series of hepatoblastomas: numerical abnormalities with recurring aberrations involving 1q12-q21. Genes, Chromosomes and Cancer 2005;44:177–84.CrossRef
DeBaun, MR, Tucker, MA. Risk of cancer during the first four years of life in children from The Beckwith-Wiedemann Syndrome Registry. Journal of Pediatrics 1998;132:398–400.CrossRefGoogle ScholarPubMed
Giardiello, FM, Offerhaus, GJ, Krush, AJ, et al. Risk of hepatoblastoma in familial adenomatous polyposis. Journal of Pediatrics 1991;119:766–8.CrossRefGoogle ScholarPubMed
Hughes, LJ, Michels, VV. Risk of hepatoblastoma in familial adenomatous polyposis. American Journal of Medical Genetics 1992;43:1023–5.CrossRefGoogle ScholarPubMed
Groden, J, Thliveris, A, Samowitz, W, et al. Identification and characterization of the familial adenomatous polyposis coli gene. Cell 1991;66:589–600.CrossRef
Nishisho, I, Nakamura, Y, Miyoshi, Y, et al. Mutations of chromosome 5q21 genes in FAP and colorectal cancer patients. Science 1991;253:665–9.CrossRef
Koufos, A, Grundy, P, Morgan, K, et al. Familial Wiedemann-Beckwith syndrome and a second Wilms tumor locus both map to 11p15.5. American Journal of Human Genetics 1989;44:711–19.Google Scholar
Li, FP, Thurber, WA, Seddon, J, Holmes, GE. Hepatoblastoma in families with polyposis coli. Journal of the American Medical Association 1987;257:2475–7.CrossRefGoogle ScholarPubMed
Giardiello, FM, Petersen, GM, Brensinger, JD, et al. Hepatoblastoma and APC gene mutation in familial adenomatous polyposis. Gut 1996;39:867–9.CrossRef
Koufos, A, Hansen, MF, Copeland, NG, et al. Loss of heterozygosity in three embryonal tumours suggests a common pathogenetic mechanism. Nature 1985;316:330–4.CrossRef
Byrne, JA, Simms, LA, Little, MH, Algar, EM, Smith, PJ. Three non-overlapping regions of chromosome arm 11p allele loss identified in infantile tumors of adrenal and liver. Genes, Chromosomes and Cancer 1993;8:104–11.CrossRef
Albrecht, S, von Schweinitz, D, Waha, A, et al. Loss of maternal alleles on chromosome arm 11p in hepatoblastoma. Cancer Research 1994;54:5041–4.
Wei, Y, Fabre, M, Branchereau, S, et al. Activation of beta-catenin in epithelial and mesenchymal hepatoblastomas. Oncogene 2000;19:498–504.CrossRef
Takayasu, H, Horie, H, Hiyama, E, et al. Frequent deletions and mutations of the beta-catenin gene are associated with overexpression of cyclin D1 and fibronectin and poorly differentiated histology in childhood hepatoblastoma. Clinical Cancer Research 2001;7:901–8.
Curia, MC, Zuckermann, M, De Lellis, L, et al. Sporadic childhood hepatoblastomas show activation of beta-catenin, mismatch repair defects and p53 mutations. Modern Pathology 2008;21:7–14.CrossRef
Koch, A, Denkhaus, D, Albrecht, S, et al. Childhood hepatoblastomas frequently carry a mutated degradation targeting box of the beta-catenin gene. Cancer Research 1999;59:269–73.
Rubinfeld, B, Albert, I, Porfiri, E, et al. Binding of GSK3beta to the APC-beta-catenin complex and regulation of complex assembly. Science 1996;272:1023–6.CrossRef
Schneider, NR, Cooley, LD, Finegold, MJ, Douglass, EC, Tomlinson, GE. The first recurring chromosome translocation in hepatoblastoma: der(4)t(1;4)(q12;q34). Genes, Chromosomes and Cancer 1997;19:291–4.3.0.CO;2-J>CrossRef
Sainati, L, Leszl, A, Stella, M, et al. Cytogenetic analysis of hepatoblastoma: hypothesis of cytogenetic evolution in such tumors and results of a multicentric study. Cancer Genetics and Cytogenetics 1998;104:39–44.CrossRef
Parada, LA, Limon, J, Iliszko, M, et al. Cytogenetics of hepatoblastoma: further characterization of 1q rearrangements by fluorescence in situ hybridization: an international collaborative study. Medical and Pediatric Oncology 2000;34:165–70.3.0.CO;2-T>CrossRef
Surace, C, Leszl, A, Perilongo, G, et al. Fluorescent in situ hybridization (FISH) reveals frequent and recurrent numerical and structural abnormalities in hepatoblastoma with no informative karyotype. Medical and Pediatric Oncology 2002;39:536–9.CrossRef
Weber, RG, Pietsch, T, von Schweinitz, D, Lichter, P. Characterization of genomic alterations in hepatoblastomas: a role for gains on chromosomes 8q and 20 as predictors of poor outcome. American Journal of Pathology 2000;157:571–8.CrossRefGoogle ScholarPubMed
Li, X, Adam, G, Cui, H, et al. Expression, promoter usage and parental imprinting status of insulin-like growth factor II (IGF2) in human hepatoblastoma: uncoupling of IGF2 and H19 imprinting. Oncogene 1995;11:221–9.
Hartmann, W, Waha, A, Koch, A, et al. p57(KIP2) is not mutated in hepatoblastoma but shows increased transcriptional activity in a comparative analysis of the three imprinted genes p57(KIP2), IGF2, and H19. American Journal of Pathology 2000;157:1393–403.CrossRefGoogle Scholar
Davies, SM. Maintenance of genomic imprinting at the IGF2 locus in hepatoblastoma. Cancer Research 1993;53:4781–3.
Rainier, S, Dobry, CJ, Feinberg, AP. Loss of imprinting in hepatoblastoma. Cancer Research 1995;55:1836–8.
Huse, JT, Holland, EC. Targeting brain cancer: advances in the molecular pathology of malignant glioma and medulloblastoma. Nature Reviews Cancer 2010;10:319–31.CrossRef
Schwartzbaum, JA, Fisher, JL, Aldape, KD, Wrensch, M. Epidemiology and molecular pathology of glioma. Nature Clinical Practice Neurology 2006;2:494–503.CrossRef
Yan, H, Parsons, DW, Jin, G, et al. IDH1 and IDH2 mutations in gliomas. New England Journal of Medicine 2009;360:765–73.CrossRefGoogle ScholarPubMed
Ries, L, Smith, M, Gurney, J, et al. Cancer incidence and survival among children and adolescents: United States SEER Program 1975–1995. Bethesda, MD: NIH Pub. No. 99–4649;1999.
Schuller, U, Heine, VM, Mao, J, et al. Acquisition of granule neuron precursor identity is a critical determinant of progenitor cell competence to form Shh-induced medulloblastoma. Cancer Cell 2008;14:123–34.CrossRef
Northcott, PA, Korshunov, A, Witt, H, et al. Medulloblastoma comprises four distinct molecular variants. Journal of Clinical Oncology 2011;29:1408–14.CrossRefGoogle ScholarPubMed
Maris, JM. Recent advances in neuroblastoma. New England Journal of Medicine 2010;362:2202–11.CrossRefGoogle ScholarPubMed
Schwab, M, Alitalo, K, Klempnauer, KH, et al. Amplified DNA with limited homology to myc cellular oncogene is shared by human neuroblastoma cell lines and a neuroblastoma tumour. Nature 1983;305:245–8.CrossRef
Attiyeh, EF, London, WB, Mosse, YP, et al. Chromosome 1p and 11q deletions and outcome in neuroblastoma. New England Journal of Medicine 2005;353:2243–53.CrossRefGoogle ScholarPubMed
Monclair, T, Brodeur, GM, Ambros, PF, et al. The International Neuroblastoma Risk Group (INRG) staging system: an INRG Task Force report. Journal of Clinical Oncology 2009;27:298–303.CrossRefGoogle ScholarPubMed
Cohn, SL, Pearson, AD, London, WB, et al. The International Neuroblastoma Risk Group (INRG) classification system: an INRG Task Force report. Journal of Clinical Oncology 2009;27:289–97.CrossRefGoogle ScholarPubMed
Bachetti, T, Di Paolo, D, Di Lascio, S, et al. PHOX2B-mediated regulation of ALK expression: in vitro identification of a functional relationship between two genes involved in neuroblastoma. PLoS One 2010;5:e13108.
George, RE, Sanda, T, Hanna, M, et al. Activating mutations in ALK provide a therapeutic target in neuroblastoma. Nature 2008;455:975–8.CrossRef
Chen, Y, Takita, J, Choi, YL, et al. Oncogenic mutations of ALK kinase in neuroblastoma. Nature 2008;455:971–4.CrossRef
Janoueix-Lerosey, I, Lequin, D, Brugieres, L, et al. Somatic and germline activating mutations of the ALK kinase receptor in neuroblastoma. Nature 2008;455:967–70.CrossRef
Mosse, YP, Laudenslager, M, Longo, L, et al. Identification of ALK as a major familial neuroblastoma predisposition gene. Nature 2008;455:930–5.CrossRef
Buckley, JD, Pendergrass, TW, Buckley, CM, et al. Epidemiology of osteosarcoma and Ewing's sarcoma in childhood: a study of 305 cases by the Children's Cancer Group. Cancer 1998;83:1440–8.3.0.CO;2-3>CrossRef
Lau, CC, Harris, CP, Lu, XY, et al. Frequent amplification and rearrangement of chromosomal bands 6p12-p21 and 17p11.2 in osteosarcoma. Genes, Chromosomes and Cancer 2004;39:11–21.
Kleinerman, RA, Tucker, MA, Tarone, RE, et al. Risk of new cancers after radiotherapy in long-term survivors of retinoblastoma: an extended follow-up. Journal of Clinical Oncology 2005;23:2272–9.CrossRefGoogle Scholar
Wang, LL, Levy, ML, Lewis, RA, et al. Clinical manifestations in a cohort of 41 Rothmund-Thomson syndrome patients. American Journal of Medical Genetics 2001;102:11–17.3.0.CO;2-A>CrossRefGoogle Scholar
Park, M, Dean, M, Cooper, CS, et al. Mechanism of met oncogene activation. Cell 1986;45:895–904.CrossRef
Patane, S, Avnet, S, Coltella, N, et al. MET overexpression turns human primary osteoblasts into osteosarcomas. Cancer Research 2006;66:4750–7.CrossRef
Ferracini, R, Di Renzo, MF, Scotlandi, K, et al. The Met/HGF receptor is over-expressed in human osteosarcomas and is activated by either a paracrine or an autocrine circuit. Oncogene 1995;10:739–49.
Sampson, ER, Martin, BA, Morris, AE, et al. The orally bioavailable met inhibitor PF-2341066 inhibits osteosarcoma growth and osteolysis/matrix production in a xenograft model. Journal of Bone and Mineral Research 2011;26:1283–94.CrossRefGoogle Scholar
Khanna, C, Wan, X, Bose, S, et al. The membrane-cytoskeleton linker ezrin is necessary for osteosarcoma metastasis. Nature Medicine 2004;10:182–6.CrossRef
Modder, UI, Oursler, MJ, Khosla, S, Monroe, DG. Wnt10b activates the Wnt, Notch and NFkappaB pathways in U2OS osteosarcoma cells. Journal of Cellular Biochemistry 2011;112:1392–402.CrossRefGoogle ScholarPubMed
Hoang, BH, Kubo, T, Healey, JH, et al. Expression of LDL receptor-related protein 5 (LRP5) as a novel marker for disease progression in high-grade osteosarcoma. International Journal of Cancer 2004;109:106–11.CrossRefGoogle ScholarPubMed
Rubin, EM, Guo, Y, Tu, K, et al. Wnt inhibitory factor 1 decreases tumorigenesis and metastasis in osteosarcoma. Molecular Cancer Therapeutics 2010;9:731–41.CrossRef
Dieudonne, FX, Marion, A, Hay, E, Marie, PJ, Modrowski, D. High Wnt signaling represses the proapoptotic proteoglycan syndecan-2 in osteosarcoma cells. Cancer Research 2010;70:5399–408.CrossRef
Chen, K, Fallen, S, Abaan, HO, et al. Wnt10b induces chemotaxis of osteosarcoma and correlates with reduced survival. Pediatric Blood and Cancer 2008;51:349–55.CrossRef
Abdeen, A, Chou, AJ, Healey, JH, et al. Correlation between clinical outcome and growth factor pathway expression in osteogenic sarcoma. Cancer 2009;115:5243–50.CrossRef
Scotlandi, K, Serra, M, Nicoletti, G, et al. Multidrug resistance and malignancy in human osteosarcoma. Cancer Research 1996;56:2434–9.
Gorlick, R, Huvos, AG, Heller, G, et al. Expression of HER2/erbB-2 correlates with survival in osteosarcoma. Journal of Clinical Oncology 1999;17:2781–8.CrossRefGoogle ScholarPubMed
Chan, HS, Grogan, TM, Haddad, G, DeBoer, G, Ling, V. P-glycoprotein expression: critical determinant in the response to osteosarcoma chemotherapy. Journal of the National Cancer Institute 1997;89:1706–15.CrossRefGoogle ScholarPubMed
Anninga, JK, van de Vijver, MJ, Cleton-Jansen, AM, et al. Overexpression of the HER-2 oncogene does not play a role in high-grade osteosarcomas. European Journal of Cancer 2004;40:963–70.CrossRefGoogle Scholar
Chou, AJ, Kleinerman, ES, Krailo, MD, et al. Addition of muramyl tripeptide to chemotherapy for patients with newly diagnosed metastatic osteosarcoma: a report from the Children's Oncology Group. Cancer 2009;115:5339–48.CrossRef
Hurwitz, RL, Shields, CL, Shields, JA, et al. Retinoblastoma. In: Pizzo PA, Poplock DG, editors, Principles and Practice of Pediatric Oncology, fourth edn. Philadelphia: Lippincott Williams & Wilkins;2002:825–46.
Schefler, AC, Abramson, DH. Retinoblastoma: what is new in 2007–2008. Current Opinion in Ophthalmology 2008;19:526–34.CrossRef
Toguchida, J, McGee, TL, Paterson, JC, et al. Complete genomic sequence of the human retinoblastoma susceptibility gene. Genomics 1993;17:535–43.CrossRef
Buchkovich, K, Duffy, LA, Harlow, E. The retinoblastoma protein is phosphorylated during specific phases of the cell cycle. Cell 1989;58:1097–105.CrossRef
Dyson, N. The regulation of E2F by pRB-family proteins. Genes and Development 1998;12:2245–62.CrossRef
Nielsen, SJ, Schneider, R, Bauer, UM, et al. Rb targets histone H3 methylation and HP1 to promoters. Nature 2001;412:561–5.CrossRef
Lohmann, DR. RB1 gene mutations in retinoblastoma. Human Mutation 1999;14:283–8.3.0.CO;2-J>CrossRef
Lohmann, DR, Gallie, BL. Retinoblastoma: revisiting the model prototype of inherited cancer. American Journal of Medical Genetics C, Seminars in Medical Genetics 2004;129C:23–8.CrossRefGoogle Scholar
Lohmann, DR, Gerick, M, Brandt, B, et al. Constitutional RB1-gene mutations in patients with isolated unilateral retinoblastoma. American Journal of Human Genetics 1997;61:282–94.CrossRefGoogle ScholarPubMed
Eng, C, Li, FP, Abramson, DH, et al. Mortality from second tumors among long-term survivors of retinoblastoma. Journal of the National Cancer Institute 1993;85:1121–8.CrossRefGoogle ScholarPubMed
DiCiommo, D, Gallie, BL, Bremner, R. Retinoblastoma: the disease, gene and protein provide critical leads to understand cancer. Seminars in Cancer Biology 2000;10:255–69.CrossRef
Madhavan, J, Ganesh, A, Kumaramanickavel, G. Retinoblastoma: from disease to discovery. Ophthalmic Research 2008;40:221–6.CrossRef
Corson, TW, Gallie, BL. One hit, two hits, three hits, more? Genomic changes in the development of retinoblastoma. Genes, Chromosomes and Cancer 2007;46:617–34.CrossRef
Zielinski, B, Gratias, S, Toedt, G, et al. Detection of chromosomal imbalances in retinoblastoma by matrix-based comparative genomic hybridization. Genes, Chromosomes and Cancer 2005;43:294–301.CrossRef
Lillington, DM, Kingston, JE, Coen, PG, et al. Comparative genomic hybridization of 49 primary retinoblastoma tumors identifies chromosomal regions associated with histopathology, progression, and patient outcome. Genes, Chromosomes and Cancer 2003;36:121–8.CrossRef
Mairal, A, Pinglier, E, Gilbert, E, et al. Detection of chromosome imbalances in retinoblastoma by parallel karyotype and CGH analyses. Genes, Chromosomes and Cancer 2000;28:370–9.3.0.CO;2-8>CrossRef
Herzog, S, Lohmann, DR, Buiting, K, et al. Marked differences in unilateral isolated retinoblastomas from young and older children studied by comparative genomic hybridization. Human Genetics 2001;108:98–104.CrossRef
Chen, D, Gallie, BL, Squire, JA. Minimal regions of chromosomal imbalance in retinoblastoma detected by comparative genomic hybridization. Cancer Genetics and Cytogenetics 2001;129:57–63.CrossRef
Bowles, E, Corson, TW, Bayani, J, et al. Profiling genomic copy number changes in retinoblastoma beyond loss of RB1. Genes, Chromosomes and Cancer 2007;46:118–29.CrossRef
Corson, TW, Huang, A, Tsao, MS, Gallie, BL. KIF14 is a candidate oncogene in the 1q minimal region of genomic gain in multiple cancers. Oncogene 2005;24:4741–53.CrossRef
Wexler, LH, Crist, WM, Helman, LJ. Rhabdomyosarcomas and the undifferentiated sarcomas. In: Pizzo PA, Poplock DG, editors, Principles and Practice for Pediatric Oncology. Philadelphia: Lippincott Williams & Wilkins; 2002:939–71.
Li, FP, Fraumeni, JFRhabdomyosarcoma in children: epidemiologic study and identification of a familial cancer syndrome. Journal of the National Cancer Institute 1969;43:1365–73.Google ScholarPubMed
Ferrari, A, Bisogno, G, Macaluso, A, et al. Soft-tissue sarcomas in children and adolescents with neurofibromatosis type 1. Cancer 2007;109:1406–12.CrossRef
Mercado, GE, Barr, FG. Fusions involving PAX and FOX genes in the molecular pathogenesis of alveolar rhabdomyosarcoma: recent advances. Current Molecular Medicine 2007;7:47–61.CrossRef
El-Badry, OM, Minniti, C, Kohn, EC, et al. Insulin-like growth factor II acts as an autocrine growth and motility factor in human rhabdomyosarcoma tumors. Cell Growth and Differentiation 1990;1:325–31.
Kalebic, T, Blakesley, V, Slade, C, et al. Expression of a kinase-deficient IGF-I-R suppresses tumorigenicity of rhabdomyosarcoma cells constitutively expressing a wild type IGF-I-R. International Journal of Cancer 1998;76:223–7.3.0.CO;2-Z>CrossRefGoogle ScholarPubMed
Zhan, S, Shapiro, DN, Helman, LJ. Activation of an imprinted allele of the insulin-like growth factor II gene implicated in rhabdomyosarcoma. Journal of Clinical Investigation 1994;94:445–8.CrossRefGoogle ScholarPubMed
Barr, FG. Gene fusions involving PAX and FOX family members in alveolar rhabdomyosarcoma. Oncogene 2001;20:5736–46.CrossRef
Galili, N, Davis, RJ, Fredericks, WJ, et al. Fusion of a fork head domain gene to PAX3 in the solid tumour alveolar rhabdomyosarcoma. Nature Genetics 1993;5:230–5.CrossRef
Barr, FG, Galili, N, Holick, J, et al. Rearrangement of the PAX3 paired box gene in the paediatric solid tumour alveolar rhabdomyosarcoma. Nature Genetics 1993;3:113–17.CrossRef
Davis, RJ, D’Cruz, CM, Lovell, MA, Biegel, JA, Barr, FG. Fusion of PAX7 to FKHR by the variant t(1;13)(p36;q14) translocation in alveolar rhabdomyosarcoma. Cancer Research 1994;54:2869–72.
Accili, D, Arden, KC. FoxOs at the crossroads of cellular metabolism, differentiation, and transformation. Cell 2004;117:421–6.CrossRef
Williamson, D, Missiaglia, E, de Reynies, A, et al. Fusion gene-negative alveolar rhabdomyosarcoma is clinically and molecularly indistinguishable from embryonal rhabdomyosarcoma. Journal of Clinical Oncology 2010;28:2151–8.CrossRefGoogle ScholarPubMed
Missiaglia, E, Williamson, D, Chisholm, J, et al. PAX3/FOXO1 fusion gene status is the key prognostic molecular marker in rhabdomyosarcoma and significantly improves current risk stratification. Journal of Clinical Oncology 2012;30:1670–7.CrossRefGoogle ScholarPubMed
Ginsberg, JP, Davis, RJ, Bennicelli, JL, Nauta, LE, Barr, FG. Up-regulation of MET but not neural cell adhesion molecule expression by the PAX3-FKHR fusion protein in alveolar rhabdomyosarcoma. Cancer Research 1998;58:3542–6.
Jeffers, M, Rong, S, Woude, GF. Hepatocyte growth factor/scatter factor-Met signaling in tumorigenicity and invasion/metastasis. Journal of Molecular Medicine 1996;74:505–13.CrossRefGoogle ScholarPubMed
Matsumoto, K, Nakamura, T. Emerging multipotent aspects of hepatocyte growth factor. Journal of Biochemistry 1996;119:591–600.CrossRefGoogle ScholarPubMed
Epstein, JA, Shapiro, DN, Cheng, J, Lam, PY, Maas, RL. Pax3 modulates expression of the c-Met receptor during limb muscle development. Proceedings of the National Academy of Sciences USA 1996;93:4213–18.CrossRef
Libura, J, Drukala, J, Majka, M, et al. CXCR4-SDF-1 signaling is active in rhabdomyosarcoma cells and regulates locomotion, chemotaxis, and adhesion. Blood 2002;100:2597–606.CrossRef
Merlino, G, Helman, LJ. Rhabdomyosarcoma-working out the pathways. Oncogene 1999;18:5340–8.CrossRef
Felix, CA, Kappel, CC, Mitsudomi, T, et al. Frequency and diversity of p53 mutations in childhood rhabdomyosarcoma. Cancer Research 1992;52:2243–7.
Stratton, MR, Fisher, C, Gusterson, BA, Cooper, CS. Detection of point mutations in N-ras and K-ras genes of human embryonal rhabdomyosar-comas using oligonucleotide probes and the polymerase chain reaction. Cancer Research 1989;49:6324–7.
Wexler, LH, Helman, LJ. Pediatric soft tissue sarcomas. CA, A Cancer Journal for Clinicians 1994;44:211–47.CrossRef
Konieczny, SF, Drobes, BL, Menke, SL, Taparowsky, EJ. Inhibition of myogenic differentiation by the H-ras oncogene is associated with the down regulation of the MyoD1 gene. Oncogene 1989;4:473–81.
Lassar, AB, Buskin, JN, Lockshon, D, et al. MyoD is a sequence-specific DNA binding protein requiring a region of myc homology to bind to the muscle creatine kinase enhancer. Cell 1989;58:823–31.CrossRef
Dias, P, Kumar, P, Marsden, HB, et al. N-myc gene is amplified in alveolar rhabdomyosarcomas (RMS) but not in embryonal RMS. International Journal of Cancer 1990;45:593–6.CrossRefGoogle ScholarPubMed
Taylor, JGt, Cheuk, AT, Tsang, PS, et al. Identification of FGFR4-activating mutations in human rhabdomyosar-comas that promote metastasis in xenotransplanted models. Journal of Clinical Investigation 2009;119:3395–407.Google ScholarPubMed
Grundy, PE, Green, DM, Coppes, MJ, et al. Renal tumors. In: Pizzo PA, Poplock DG, editors, Principles and Practice for Pediatric Oncology. Philadelphia: Lippincott Williams & Wilkins; 2002:865–93.
Hastie, ND. The genetics of Wilms’ tumor–a case of disrupted development. Annual Review of Genetics 1994;28:523–58.CrossRef
Mierau, GW, Beckwith, JB, Weeks, DA. Ultrastructure and histogenesis of the renal tumors of childhood: an overview. Ultrastructural Pathology 1987;11:313–33.CrossRef
Brown, KW, Malik, KT. The molecular biology of Wilms tumour. Expert Review of Molecular Medicine 2001:1–16.
Blackmore, C, Coppes, MJ, Narendran, A. Candidate genes and potential targets for therapeutics in Wilms’ tumour. Clinical and Translational Oncology 2010;12:597–605.CrossRef
Francke, U, Holmes, LB, Atkins, L, Riccardi, VM. Aniridia-Wilms’ tumor association: evidence for specific deletion of 11p13. Cytogenetics and Cell Genetics 1979;24:185–92.CrossRef
Gronskov, K, Olsen, JH, Sand, A, et al. Population-based risk estimates of Wilms tumor in sporadic aniridia: a comprehensive mutation screening procedure of PAX6 identifies 80% of mutations in aniridia. Human Genetics 2001;109:11–18.CrossRef
Muto, R, Yamamori, S, Ohashi, H, Osawa, M. Prediction by FISH analysis of the occurrence of Wilms tumor in aniridia patients. American Journal of Medical Genetics 2002;108:285–9.CrossRefGoogle ScholarPubMed
Pelletier, J, Bruening, W, Kashtan, CE, et al. Germline mutations in the Wilms’ tumor suppressor gene are associated with abnormal urogenital development in Denys-Drash syndrome. Cell 1991;67:437–47.CrossRef
Heathcott, RW, Morison, IM, Gubler, MC, Corbett, R, Reeve, AE. A review of the phenotypic variation due to the Denys-Drash syndrome-associated germline WT1 mutation R362X. Human Mutation. 2002;19:462.CrossRef
Bruening, W, Bardeesy, N, Silverman, BL, et al. Germline intronic and exonic mutations in the Wilms’ tumour gene (WT1) affecting urogenital development. Nature Genetics 1992;1:144–8.CrossRef
Pelletier, J, Bruening, W, Li, FP, et al. WT1 mutations contribute to abnormal genital system development and hereditary Wilms’ tumour. Nature 1991;353:431–4.CrossRef
Kreidberg, JA, Sariola, H, Loring, JM, et al. WT-1 is required for early kidney development. Cell 1993;74:679–91.CrossRef
Drummond, IA, Madden, SL, Rohwer-Nutter, P, et al. Repression of the insulin-like growth factor II gene by the Wilms tumor suppressor WT1. Science 1992;257:674–8.CrossRef
Mayo, MW, Wang, CY, Drouin, SS, et al. WT1 modulates apoptosis by transcriptionally upregulating the bcl-2 proto-oncogene. EMBO Journal 1999;18:3990–4003.CrossRef
Ruteshouser, EC, Robinson, SM, Huff, V. Wilms tumor genetics: mutations in WT1, WTX, and CTNNB1 account for only about one-third of tumors. Genes, Chromosomes and Cancer. 2008;47:461–70.CrossRef
Koesters, R, Ridder, R, Kopp-Schneider, A, et al. Mutational activation of the beta-catenin proto-oncogene is a common event in the development of Wilms’ tumors. Cancer Research 1999;59:3880–2.
Maiti, S, Alam, R, Amos, CI, Huff, V. Frequent association of beta-catenin and WT1 mutations in Wilms tumors. Cancer Research 2000;60:6288–92.
Li, CM, Kim, CE, Margolin, AA, et al. CTNNB1 mutations and overexpression of Wnt/beta-catenin target genes in WT1-mutant Wilms’ tumors. American Journal of Pathology 2004;165:1943–53.CrossRefGoogle ScholarPubMed
Major, MB, Camp, ND, Berndt, JD, et al. Wilms tumor suppressor WTX negatively regulates WNT/beta-catenin signaling. Science 2007;316:1043–6.CrossRef
Rivera, MN, Kim, WJ, Wells, J, et al. An X chromosome gene, WTX, is commonly inactivated in Wilms tumor. Science 2007;315:642–5.CrossRef
Ping, AJ, Reeve, AE, Law, DJ, et al. Genetic linkage of Beckwith-Wiedemann syndrome to 11p15. American Journal of Human Genetics 1989;44:720–3.Google ScholarPubMed
Ogawa, O, Eccles, MR, Szeto, J, et al. Relaxation of insulin-like growth factor II gene imprinting implicated in Wilms’ tumour. Nature 1993;362:749–51.CrossRef
Moulton, T, Chung, WY, Yuan, L, et al. Genomic imprinting and Wilms’ tumor. Medical and Pediatric Oncology 1996;27:476–83.3.0.CO;2-8>CrossRef
Feinberg, AP. Imprinting of a genomic domain of 11p15 and loss of imprinting in cancer: an introduction. Cancer Research 1999;59:1743s–6s.
Steenman, MJ, Rainier, S, Dobry, CJ, et al. Loss of imprinting of IGF2 is linked to reduced expression and abnormal methylation of H19 in Wilms’ tumour. Nature Genetics 1994;7:433–9.CrossRef
Moulton, T, Crenshaw, T, Hao, Y, et al. Epigenetic lesions at the H19 locus in Wilms’ tumour patients. Nature Genetics 1994;7:440–7.CrossRef
Grundy, RG, Pritchard, J, Scambler, P, Cowell, JK. Loss of heterozygosity for the short arm of chromosome 7 in sporadic Wilms tumour. Oncogene 1998;17:395–400.CrossRef
Powlesland, RM, Charles, AK, Malik, KT, et al. Loss of heterozygosity at 7p in Wilms’ tumour development. British Journal of Cancer 2000;82:323–9.CrossRefGoogle ScholarPubMed
Hing, S, Lu, YJ, Summersgill, B, et al. Gain of 1q is associated with adverse outcome in favorable histology Wilms’ tumors. American Journal of Pathology 2001;158:393–8.CrossRefGoogle ScholarPubMed
Maw, MA, Grundy, PE, Millow, LJ, et al. A third Wilms’ tumor locus on chromosome 16q. Cancer Research 1992;52:3094–8.
Grundy, PE, Breslow, NE, Li, S, et al. Loss of heterozygosity for chromosomes 1p and 16q is an adverse prognostic factor in favorable-histology Wilms tumor: a report from the National Wilms Tumor Study Group. Journal of Clinical Oncology 2005;23:7312–21.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
×