Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-5nwft Total loading time: 0 Render date: 2024-05-01T10:00:06.975Z Has data issue: false hasContentIssue false

59 - Molecular oncology of neuroblastoma

from Part 3.2 - Molecular pathology: cancers of the nervous system

Published online by Cambridge University Press:  05 February 2015

Vandana Batra
Affiliation:
Center for Childhood Cancer Research, Department of Pediatrics, Childrens Hospital of Philadelphia, Philadelphia, PA, USA
Rebecca J. Deyell
Affiliation:
Center for Childhood Cancer Research, Department of Pediatrics, Childrens Hospital of Philadelphia, Philadelphia, PA, USA
John M. Maris
Affiliation:
Center for Childhood Cancer Research, Department of Pediatrics, Childrens Hospital of Philadelphia, Philadelphia, PA, 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

Introduction

Neuroblastoma, an embryonal tumor of the sympathetic nervous system, is the most common extra-cranial solid tumor of childhood and the third leading cause of childhood cancer-related mortality (1,2). With a median age at diagnosis of 17 months, neuroblastoma has an annual age-adjusted incidence rate of 10.0 per million children younger than 15 years (2,3). It arises from postganglionic sympathetic neuroblasts, typically involving the adrenal medulla or paraspinal ganglia, and clinical presentations range from the incidental detection of an isolated, asymptomatic mass to disseminated disease in which children often present with rapid clinical deterioration.

The hallmark of neuroblastoma is its distinctive clinical heterogeneity. Most infants have tumors which undergo spontaneous regression or behave in a benign manner, with survival rates exceeding 95%, despite disease that may involve the liver, skin, and bone marrow (4–6). In contrast, older children with high-risk neuroblastoma have aggressive disease that is often resistant to intensive multi-modality therapy (7). There is abundant evidence that these divergent neuroblastoma phenotypes are predicted by distinct molecular features that are prognostic of survival outcomes and currently guide therapeutic decisions in the broad sense of defining tumor aggressiveness (8). The unmet need in the field, as discussed herein, is to precisely define molecular genetic subsets of high-risk neuroblastoma that reliably predict unique oncogenic vulnerabilities that are exploitable therapeutically.

Type
Chapter
Information
Molecular Oncology
Causes of Cancer and Targets for Treatment
, pp. 669 - 678
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

Smith, MA, Seibel, NL, Altekruse, SF, et al. Outcomes for children and adolescents with cancer:challenges for the twenty-first century. Journal of Clinical Oncology 2010;28:2625–34.CrossRefGoogle ScholarPubMed
Surveillance, Epidemiology and End Results (SEER) database. National Cancer Institute 2010;Accessed July 2011;.
London, WB, Castleberry, RP, Matthay, KK, et al. Evidence for an age cutoff greater than 365 days for neuroblastoma risk group stratification in the Children's Oncology Group. Journal of Clinical Oncology 2005;23:6459–65.CrossRefGoogle ScholarPubMed
D’Angio, GJ, Evans, AE, Koop, CE. Special pattern of widespread neuroblastoma with a favourable prognosis. Lancet 1971;1:1046–9.CrossRef
De Bernardi, B, Gerrard, M, Boni, L, et al. Excellent outcome with reduced treatment for infants with disseminated neuroblastoma without MYCN gene amplification. Journal of Clinical Oncology 2009;27:1034–40.CrossRefGoogle ScholarPubMed
De Bernardi, B, Pianca, C, Boni, L, et al. Disseminated neuroblastoma (stage IV and IV-S) in the first year of life: outcome related to age and stage. Italian Cooperative Group on Neuroblastoma. Cancer 1992;70:1625–33.
Maris, JM, Maris, JM. Recent advances in neuroblastoma. New England Journal of Medicine 2010;362:2202–11.CrossRefGoogle ScholarPubMed
Ambros, PF, Ambros, IM, Brodeur, GM, et al. International consensus for neuroblastoma molecular diagnostics: report from the International Neuroblastoma Risk Group (INRG) Biology Committee. British Journal of Cancer 2009;100:1471–82.CrossRefGoogle ScholarPubMed
Matthay, KK, Reynolds, CP, Seeger, RC, et al. Long-term results for children with high-risk neuroblastoma treated on a randomized trial of myeloablative therapy followed by 13-cis-retinoic acid: a children's oncology group study. Journal of Clinical Oncology 2009;27:1007–13.CrossRefGoogle ScholarPubMed
Matthay, KK, Villablanca, JG, Seeger, RC, et al. Treatment of high-risk neuroblastoma with intensive chemotherapy, radiotherapy, autologous bone marrow transplantation, and 13-cis-retinoic acid. Children's Cancer Group. New England Journal of Medicine 1999;341:1165–73.CrossRefGoogle ScholarPubMed
Yu, AL, Gilman, AL, Ozkaynak, MF, et al. Anti-GD2 antibody with GM-CSF, interleukin-2, and isotretinoin for neuroblastoma. New England Journal of Medicine 2010;363:1324–34.CrossRefGoogle ScholarPubMed
Maris, JM, Hogarty, MD, Bagatell, R, et al. Neuroblastoma. Lancet 2007;369:2106–20.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
Griffin, CA, Hawkins, AL, Dvorak, C, et al. Recurrent involvement of 2p23 in inflammatory myofibroblastic tumors. Cancer Research 1999;59:2776–80.
Jazii, FR, Najafi, Z, Malekzadeh, R, et al. Identification of squamous cell carcinoma associated proteins by proteomics and loss of beta tropomyosin expression in esophageal cancer. World Journal of Gastroenterology 2006;12:7104–12.CrossRefGoogle ScholarPubMed
Morris, SW, Kirstein, MN, Valentine, MB, et al. Fusion of a kinase gene, ALK, to a nucleolar protein gene, NPM, in non-Hodgkin's lymphoma. [Erratum Science. 1995;267:316–7]. Science 1994;263:1281–4.
Rikova, K, Guo, A, Zeng, Q, et al. Global survey of phosphotyrosine signaling identifies oncogenic kinases in lung cancer. Cell 2007;131:1190–203.CrossRef
Soda, M, Choi, YL, Enomoto, M, et al. Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature 2007;448:561–6.CrossRef
Inamura, K, Takeuchi, K, Togashi, Y, et al. EML4-ALK fusion is linked to histological characteristics in a subset of lung cancers. Journal of Thoracic Oncology 2008;3:13–7.CrossRefGoogle Scholar
Caren, H, Abel, F, Kogner, P, et al. High incidence of DNA mutations and gene amplifications of the ALK gene in advanced sporadic neuroblastoma tumours. Biochemical Journal 2008;416:153–9.CrossRef
Chen, Y, Takita, J, Choi, YL, et al. Oncogenic mutations of ALK kinase in neuroblastoma. Nature 2008;455:971–4.CrossRef
George, RE, Sanda, T, Hanna, M, et al. Activating mutations in ALK provide a therapeutic target in neuroblastoma. Nature 2008;455:975–8.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
De Brouwer, S, De Preter, K, Kumps, C, et al. Meta-analysis of neuroblastomas reveals a skewed ALK mutation spectrum in tumors with MYCN amplification. Clinical Cancer Research 2010;16:4353–62.CrossRef
Weiser, D, Laudenslager, M, Rappaport, E, et al. Stratification of patients with neuroblastoma for targeted ALK inhibitor therapy. Journal of Clinical Oncology 2011;29 (suppl.; abstr. 9514).CrossRefGoogle Scholar
Schulte, JH, Bachmann, HS, Brockmeyer, B, et al. High ALK receptor tyrosine kinase expression supersedes ALK mutation as a determining factor of an unfavorable phenotype in primary neuroblastoma. Clinical Cancer Research 2011;17:5082–92.CrossRef
Mosse, YP, Wood, A, Maris, JM, Inhibition of ALK signaling for cancer therapy. Clinical Cancer Research 2009;15:5609–14.
Bresler, SC, Wood, AC, Haglund, EA, et al. Differential inhibitor sensitivity of anaplastic lymphoma kinase variants found in neuroblastoma. Science Translational Medicine 2011;3:108–14.
Bower, RJ, Adkins, JC. Ondine's curse and neurocristopathy. Clinical Pediatrics (Philadelphia) 1980;19:665–8.CrossRef
Knudson, AG, Jr., Amromin, GD. Neuroblastoma and ganglioneuroma in a child with multiple neurofibromatosis. Implications for the mutational origin of neuroblastoma. Cancer 1966;19:1032–7.3.0.CO;2-7>CrossRef
Knudson, AG, Jr., Meadows, AT. Developmental genetics of neuroblastoma. Journal of the National Cancer Institute 1976;57:675–82.CrossRefGoogle ScholarPubMed
Kushner, BH, Hajdu, SI, Helson, L. Synchronous neuroblastoma and von Recklinghausen's disease: a review of the literature. Journal of Clinical Oncology 1985;3:117–20.CrossRefGoogle ScholarPubMed
Maris, JM, Chatten, J, Meadows, AT, Biegel, JA, Brodeur, GM. Familial neuroblastoma: a three-generation pedigree and a further association with Hirschsprung disease. Medical and Pediatric Oncology 1997;28:1–5.3.0.CO;2-P>CrossRef
Amiel, J, Laudier, B, Attie-Bitach, T, et al. Polyalanine expansion and frameshift mutations of the paired-like homeobox gene PHOX2B in congenital central hypoventilation syndrome. Nature Genetics 2003;33:459–61.CrossRef
Mosse, YP, Laudenslager, M, Khazi, D, et al. Germline PHOX2B mutation in hereditary neuroblastoma. American Journal of Human Genetics 2004;75:727–30.CrossRefGoogle ScholarPubMed
Trochet, D, Bourdeaut, F, Janoueix-Lerosey, I, et al. Germline mutations of the paired-like homeobox 2B (PHOX2B) gene in neuroblastoma. American Journal of Human Genetics 2004;74:761–4.CrossRefGoogle ScholarPubMed
Raabe, EH, Laudenslager, M, Winter, C, et al. Prevalence and functional consequence of PHOX2B mutations in neuroblastoma. Oncogene 2008;27:469–76.CrossRef
Viprey, VF, Lastowska, MA, Corrias, MV, et al. Minimal disease monitoring by QRT-PCR:guidelines for identification and systematic validation of molecular markers prior to evaluation in prospective clinical trials. Journal of Pathology 2008;216:245–52.CrossRefGoogle ScholarPubMed
Tartaglia, M, Mehler, EL, Goldberg, R, et al. Mutations in PTPN11, encoding the protein tyrosine phosphatase SHP-2, cause Noonan syndrome. [Erratum appears in Nature Genetics 2001;29:491]. [Erratum appears in Nature Genetics 2002;30:123]. Nature Genetics 2001;29:465–8.
Bentires-Alj, M, Paez, JG, David, FS, et al. Activating mutations of the noonan syndrome-associated SHP2/PTPN11gene in human solid tumors and adult acute myelogenous leukemia. Cancer Research 2004;64:8816–20.CrossRef
Cotton, JL, Williams, RG. Noonan syndrome and neuroblastoma. Archives of Pediatric and Adolescent Medicine 1995;149:1280–1.CrossRef
Tartaglia, M, Niemeyer, CM, Fragale, A, et al. Somatic mutations in PTPN11 in juvenile myelomonocytic leukemia, myelodysplastic syndromes and acute myeloid leukemia. Nature Genetics 2003;34:148–50.CrossRef
Birch, JM, Alston, RD, McNally, RJ, et al. Relative frequency and morphology of cancers in carriers of germline TP53 mutations. Oncogene 2001;20:4621–8.CrossRef
Armstrong, R, Greenhalgh, KL, Rattenberry, E, et al. Succinate dehydrogenase subunit B (SDHB) gene deletion associated with a composite paraganglioma/neuroblastoma. Journal of Medical Genetics 2009;46:215–16.CrossRefGoogle ScholarPubMed
Cascon, A, Landa, I, Lopez-Jimenez, E, et al. Molecular characterisation of a common SDHB deletion in paraganglioma patients. Journal of Medical Genetics 2008;45:233–8.CrossRefGoogle ScholarPubMed
Schimke, RN, Collins, DL, Stolle, CA, et al. Paraganglioma, neuroblastoma, and a SDHB mutation:Resolution of a 30-year-old mystery. American Journal of Medical Genetics A 2010;152A:1531–5.Google Scholar
Capasso, M, Devoto, M, Hou, C, et al. Common variations in BARD1 influence susceptibility to high-risk neuroblastoma. Nature Genetics 2009;41:718–23.CrossRef
Diskin, SJ, Hou, C, Glessner, JT, et al. Copy number variation at 1q21.1 associated with neuroblastoma. Nature 2009;459:987–91.CrossRef
Maris, JM, Mosse, YP, Bradfield, JP, et al. Chromosome 6p22 locus associated with clinically aggressive neuroblastoma. New England Journal of Medicine 2008;358:2585–93.CrossRefGoogle ScholarPubMed
Nguyen, L, Diskin, S, Capasso, M, et al. Phenotype restricted genome-wide association study identifies three low-risk neuroblastoma susceptibility loci. PLoS Genetics 2011;7:e1002026.
Wang, K, Diskin, S, Zhang, H, et al. Integrative genomics identifies LMO1 as a neuroblastoma oncogene. Nature 2010;469:216–20.CrossRef
Brodeur, GM, Seeger, RC, Schwab, M, Varmus, HE, Bishop, JM. Amplification of N-myc in untreated human neuroblastomas correlates with advanced disease stage. Science 1984;224:1121–4.CrossRef
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
Seeger, RC, Brodeur, GM, Sather, H, et al. Association of multiple copies of the N-myc oncogene with rapid progression of neuroblastomas. New England Journal of Medicine 1985;313:1111–16.CrossRefGoogle ScholarPubMed
Janoueix-Lerosey, I, Schleiermacher, G, Michels, E, et al. Overall genomic pattern is a predictor of outcome in neuroblastoma. Journal of Clinical Oncology 2009;27:1026–33.CrossRefGoogle ScholarPubMed
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
White, PS, Thompson, PM, Gotoh, T, et al. Definition and characterization of a region of 1p36.3 consistently deleted in neuroblastoma. Oncogene 2005;24:2684–94.CrossRef
Maris, JM, Weiss, MJ, Guo, C, et al. Loss of heterozygosity at 1p36 independently predicts for disease progression but not decreased overall survival probability in neuroblastoma patients:a Children's Cancer Group study. Journal of Clinical Oncology 2000;18:1888–99.CrossRefGoogle Scholar
Caron, H, van Sluis, P, de Kraker, J, et al. Allelic loss of chromosome 1p as a predictor of unfavorable outcome in patients with neuroblastoma. New England Journal of Medicine 1996;334:225–30.CrossRefGoogle ScholarPubMed
Guo, C, White, PS, Weiss, MJ, et al. Allelic deletion at 11q23 is common in MYCN single copy neuroblastomas. Oncogene 1999;18:4948–57.CrossRef
Plantaz, D, Vandesompele, J, Van Roy, N, et al. Comparative genomic hybridization (CGH) analysis of stage 4 neuroblastoma reveals high frequency of 11q deletion in tumors lacking MYCN amplification. International Journal of Cancer 2001;91:680–6.3.0.CO;2-R>CrossRefGoogle ScholarPubMed
Spitz, R, Hero, B, Ernestus, K, et al. Deletions in chromosome arms 3p and 11q are new prognostic markers in localized and 4s neuroblastoma. Clinical Cancer Research 2003;9:52–8.
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
Bown, N, Cotterill, S, Lastowska, M, et al. Gain of chromosome arm 17q and adverse outcome in patients with neuroblastoma. New England Journal of Medicine 1999;340:1954–61.CrossRefGoogle ScholarPubMed
Lastowska, M, Cotterill, S, Pearson, AD, et al. Gain of chromosome arm 17q predicts unfavourable outcome in neuroblastoma patients. UK Children's Cancer Study Group and the UK Cancer Cytogenetics Group. European Journal of Cancer 1997;33:1627–33.CrossRefGoogle Scholar
Lastowska, M, Viprey, V, Santibanez-Koref, M, et al. Identification of candidate genes involved in neuroblastoma progression by combining genomic and expression microarrays with survival data. Oncogene 2007;26:7432–44.CrossRef
Wang, Q, Diskin, S, Rappaport, E, et al. Integrative genomics identifies distinct molecular classes of neuroblastoma and shows that multiple genes are targeted by regional alterations in DNA copy number. Cancer Research 2006;66:6050–62.CrossRef
Asgharzadeh, S, Pique-Regi, R, Sposto, R, et al. Prognostic significance of gene expression profiles of metastatic neuroblastomas lacking MYCN gene amplification. Journal of the National Cancer Institute 2006;98:1193–203.CrossRefGoogle ScholarPubMed
De Preter, K, Vermeulen, J, Brors, B, et al. Accurate outcome prediction in neuroblastoma across independent data sets using a multigene signature. Clinical Cancer Research 2010;16:1532–41.CrossRef
Oberthuer, A, Berthold, F, Warnat, P, et al. Customized oligonucleotide microarray gene expression-based classification of neuroblastoma patients outperforms current clinical risk stratification. Journal of Clinical Oncology 2006;24:5070–8.CrossRefGoogle ScholarPubMed
Oberthuer, A, Hero, B, Berthold, F, et al. Prognostic impact of gene expression-based classification for neuroblastoma. Journal of Clinical Oncology 2010;28:3506–15.CrossRefGoogle ScholarPubMed
Vermeulen, J, De Preter, K, Naranjo, A, et al. Predicting outcomes for children with neuroblastoma using a multigene-expression signature: a retrospective SIOPEN/COG/GPOH study. Lancet Oncology 2009;10:663–71.CrossRef
Wei, JS, Greer, BT, Westermann, F, et al. Prediction of clinical outcome using gene expression profiling and artificial neural networks for patients with neuroblastoma. [Erratum Cancer Research. 2005;65:374]. Cancer Research 2004;64:6883–91.
Morozova, O, Birol, I, Corbett, R, et al. Whole genome and transcriptome sequencing defines the spectrum of somatic changes in high-risk neuroblastoma. American Association for Cancer Research Meeting Abstracts April 2011.
Pugh, T, Lawrence, M, Sougnez, C, et al. Exome sequencing of 81 neuroblastomas identifies a wide diversity of somatic mutation. American Association for Cancer Research Meeting Abstracts April 2011.
Westermark, UK, Wilhelm, M, Frenzel, A, Henriksson, MA. The MYCN oncogene and differentiation in neuroblastoma. Seminars in Cancer Biology 2011;21:256–66.CrossRef
Brodeur, GM, Maris, JM, Yamashiro, DJ, Hogarty, MD, White, PS. Biology and genetics of human neuroblastomas. Journal of Pediatric Hematology/Oncology 1997;19:93–101.CrossRefGoogle ScholarPubMed
Adamson, PC, Blaney, SM. New approaches to drug development in pediatric oncology. Cancer Journal 2005;11:324–30.CrossRef
Caren, H, Djos, A, Nethander, M, et al. Identification of epigenetically regulated genes that predict patient outcome in neuroblastoma. BMC Cancer;11:66.
Wellstein, A, Toretsky, JA. Hunting ALK to feed targeted cancer therapy. Nature Medicine;17:290–1.
Mosse, Y. ADVL0912 A Phase 1/2 Study of PF-02341066, an oral small molecule inhibitor of anaplastic lymphoma kinase (ALK) and C-Met, in children with relapsed/refractory solid tumors and anaplastic large cell lymphoma. Children's Oncology Group Phase I Consortium 2009.
Di Paolo, D, Ambrogio, C, Pastorino, F, et al. Selective therapeutic targeting of the anaplastic lymphoma kinase with liposomal siRNA induces apoptosis and inhibits angiogenesis in neuroblastoma. Molecular Therapeutics 2011;19:2201–12.
Gambacorti-Passerini, C, Messa, C, Pogliani, EM. Crizotinib in anaplastic large-cell lymphoma. New England Journal of Medicine 2011;364:775–6.CrossRefGoogle ScholarPubMed
Schonherr, C, Ruuth, K, Yamazaki, Y, et al. Activating ALK mutations found in neuroblastoma are inhibited by Crizotinib and NVP-TAE684. Biochemical Journal 2011;440:405–13.CrossRef
Kwak, EL, Bang, YJ, Camidge, DR, et al. Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer. New England Journal of Medicine 2010;363:1693–703.CrossRefGoogle ScholarPubMed
Sasaki, T, Okuda, K, Zheng, W, et al. The neuroblastoma-associated F1174L ALK mutation causes resistance to an ALK kinase inhibitor in ALK-translocated cancers. Cancer Research 70:10 038–43.
Cheung, NK, Lazarus, H, Miraldi, FD, et al. Ganglioside GD2 specific monoclonal antibody 3F8: a Phase I study in patients with neuroblastoma and malignant melanoma. Journal of Clinical Oncology 1987;5:1430–40.CrossRefGoogle ScholarPubMed
Schulz, G, Cheresh, DA, Varki, NM, et al. Detection of ganglioside GD2 in tumor tissues and sera of neuroblastoma patients. Cancer Research 1984;44:5914–20.
Sebolt-Leopold, JS, Herrera, R, Ohren, JF. The mitogen-activated protein kinase pathway for molecular-targeted cancer treatment. Recent Results Cancer Research 2007;172:155–67.CrossRef
Vigil, D, Cherfils, J, Rossman, KL, Der, CJ. Ras superfamily GEFs and GAPs:validated and tractable targets for cancer therapy? Nature Reviews Cancer 2010;10:842–57.
Bollag, G, Freeman, S, Lyons, JF, Post, LE. Raf pathway inhibitors in oncology. Current Opinion in Investigative Drugs 2003;4:1436–41.
Bunney, TD, Katan, M. Phosphoinositide signalling in cancer:beyond PI3K and PTEN. Nature Reviews Cancer 2010;10:342–52.CrossRef
Sebolt-Leopold, JS, Herrera, R. Targeting the mitogen-activated protein kinase cascade to treat cancer. Nature Reviews Cancer 2004;4:937–47.CrossRef
Holzel, M, Huang, S, Koster, J, et al. NF1 is a tumor suppressor in neuroblastoma that determines retinoic acid response and disease outcome. Cell 2010;142:218–29.CrossRef
Opel, D, Poremba, C, Simon, T, Debatin, KM, Fulda, S. Activation of Akt predicts poor outcome in neuroblastoma. Cancer Research 2007;67:735–45.CrossRef
Dam, V, Morgan, BT, Mazanek, P, et al. Mutations in PIK3CA are infrequent in neuroblastoma. BMC Cancer 2006;6:177.CrossRef
Chesler, L, Schlieve, C, Goldenberg, DD, et al. Inhibition of phosphatidylinositol 3-kinase destabilizes Mycn protein and blocks malignant progression in neuroblastoma. Cancer Research 2006;66:8139–46.CrossRef
Matthay, KK, Quach, A, Huberty, J, et al. Iodine-131–metaiodobenzylguanidine double infusion with autologous stem-cell rescue for neuroblastoma: a new approache to neuroblastoma therapy Phase I study. Journal of Clinical Oncology 2009;27:1020–5.CrossRefGoogle Scholar
Matthay, KK, Quach, A, Huberty, J, et al. Iodine-131–metaiodobenzylguanidine double infusion with autologous stem-cell rescue for neuroblastoma: a new approache to neuroblastoma therapy Phase I study. Journal of Clinical Oncology 2009;27:1020–5.CrossRefGoogle Scholar
Matthay, KK, DeSantes, K, Hasegawa, B, et al. Phase I dose escalation of 131I-metaiodobenzylguanidine with autologous bone marrow support in refractory neuroblastoma. Journal of Clinical Oncology 1998;16:229–36.CrossRefGoogle ScholarPubMed
Matthay, KK, Yanik, G, Messina, J, et al. Phase II study on the effect of disease sites, age, and prior therapy on response to iodine-131-metaiodobenzylguanidine therapy in refractory neuroblastoma. Journal of Clinical Oncology 2007;25:1054–60.CrossRefGoogle ScholarPubMed
Vaidyanathan, G, Affleck, DJ, Alston, KL, et al. A kit method for the high level synthesis of [211At]MABG. Bioorganic and Medicinal Chemistry 2007;15:3430–6.CrossRef
Mairs, RJ, Boyd, M. Preclinical assessment of strategies for enhancement of metaiodobenzylgu-anidine therapy of neuroendocrine tumors. Seminars in Nuclear Medicine 2011;41:334–44.CrossRef
Maris, JM, Morton, CL, Gorlick, R, et al. Initial testing of the aurora kinase A inhibitor MLN8237 by the Pediatric Preclinical Testing Program (PPTP). Pediatric Blood Cancer 2010;55:26–34.
Maris, JM. Unholy matrimony: Aurora A and N-Myc as malignant partners in neuroblastoma. Cancer Cell 2009;15:5–6.CrossRef
Carol, H, Boehm, I, Reynolds, CP, et al. Efficacy and pharmacokinetic/pharmacodynamic evaluation of the Aurora kinase A inhibitor MLN8237 against preclinical models of pediatric cancer. Cancer Chemotherapy and Pharmacology 2011;68:1291–304.CrossRef
Shang, X, Burlingame, SM, Okcu, MF, et al. Aurora A is a negative prognostic factor and a new therapeutic target in human neuroblastoma. Molecular Cancer Therapeutics 2009;8:2461–9.CrossRef
Otto, T, Horn, S, Brockmann, M, et al. Stabilization of N-Myc is a critical function of Aurora A in human neuroblastoma. Cancer Cell 2009;15:67–78.CrossRef
Cole, KA, Huggins, J, Laquaglia, M, et al. RNAi screen of the protein kinome identifies checkpoint kinase 1 (CHK1) as a therapeutic target in neuroblastoma. Proceedings of the National Academy of Sciences USA 2011;108:3336–41.CrossRef
Xu, H, Cheung, IY, Wei, XX, et al. Checkpoint kinase inhibitor synergizes with DNA-damaging agents in G(1) checkpoint-defective neuroblastoma. International Journal of Cancer 2011;129:1953–62.CrossRefGoogle Scholar
Fredlund, E, Ringner, M, Maris, JM, Pahlman, S. High Myc pathway activity and low stage of neuronal differentiation associate with poor outcome in neuroblastoma. Proceedings of the National Academy of Sciences USA 2008;105:14 094–9.

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.

  • Molecular oncology of neuroblastoma
    • By Vandana Batra, Center for Childhood Cancer Research, Department of Pediatrics, Childrens Hospital of Philadelphia, Philadelphia, PA, USA, Rebecca J. Deyell, Center for Childhood Cancer Research, Department of Pediatrics, Childrens Hospital of Philadelphia, Philadelphia, PA, USA, John M. Maris, Center for Childhood Cancer Research, Department of Pediatrics, Childrens Hospital of Philadelphia, Philadelphia, PA, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.060
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.

  • Molecular oncology of neuroblastoma
    • By Vandana Batra, Center for Childhood Cancer Research, Department of Pediatrics, Childrens Hospital of Philadelphia, Philadelphia, PA, USA, Rebecca J. Deyell, Center for Childhood Cancer Research, Department of Pediatrics, Childrens Hospital of Philadelphia, Philadelphia, PA, USA, John M. Maris, Center for Childhood Cancer Research, Department of Pediatrics, Childrens Hospital of Philadelphia, Philadelphia, PA, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.060
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.

  • Molecular oncology of neuroblastoma
    • By Vandana Batra, Center for Childhood Cancer Research, Department of Pediatrics, Childrens Hospital of Philadelphia, Philadelphia, PA, USA, Rebecca J. Deyell, Center for Childhood Cancer Research, Department of Pediatrics, Childrens Hospital of Philadelphia, Philadelphia, PA, USA, John M. Maris, Center for Childhood Cancer Research, Department of Pediatrics, Childrens Hospital of Philadelphia, Philadelphia, PA, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.060
Available formats
×