4,113
Views
228
CrossRef citations to date
0
Altmetric
Review

Overview and recent advances in the treatment of neuroblastoma

, , , , &
Pages 369-386 | Received 14 Jun 2016, Accepted 18 Jan 2017, Published online: 15 Mar 2017

References

  • Ries L, Smith M, Gurney J, et al. Cancer incidence and survival among children and adolescents: united States SEER Program 1975–1995, National Cancer Institute, SEER Program. Bethesda (MD): NIH Pub; 1999. p. 99–4649.
  • Stiller CA, Parkin DM. International variations in the incidence of neuroblastoma. Int J Cancer. 1992;52:538–543.
  • Heck JE, Ritz B, Hung RJ, et al. The epidemiology of neuroblastoma: a review. Paediatr Perinat Epidemiol. 2009;23:125–143.
  • Irwin MS, Park JR. Neuroblastoma: paradigm for precision medicine. Pediatr Clin NA. 2015;62:225–256.
  • Henderson TO, Bhatia S, Pinto N, et al. Racial and ethnic disparities in risk and survival in children with neuroblastoma: a Children’s Oncology Group study. J Clin Oncol. 2011;29:76–82.
  • Yamamoto K, Ohta S, Ito E, et al. Marginal decrease in mortality and marked increase in incidence as a result of neuroblastoma screening at 6 months of age: cohort study in seven prefectures in Japan. J Clin Oncol. 2002;20:1209–1214.
  • Hiyama E, Iehara T, Sugimoto T, et al. Effectiveness of screening for neuroblastoma at 6 months of age: a retrospective population-based cohort study. Lancet. 2008;371:1173–1180.
  • Schilling FH, Spix C, Berthold F, et al. Neuroblastoma screening at one year of age. N Engl J Med. 2002;346:1047–1053.
  • Woods WG, Gao RN, Shuster JJ, et al. Screening of infants and mortality due to neuroblastoma. N Engl J Med. 2002;346:1041–1046.
  • Maris JM, Woods WG. Screening for neuroblastoma: a resurrected idea? Lancet. 2008;371:1142–1143.
  • Cook MN, Olshan AF, Guess HA, et al. Maternal medication use and neuroblastoma in offspring. Am J Epidemiol. 2004;159:721–731.
  • Menegaux F, Olshan AF, Nelgia JP, et al. Day care, childhood infections, and risk of neuroblastoma. Am J Epidmiol. 2004;159:843–851.
  • McDermott S, Salzberg DC, Anderson AP, et al. Systematic review of chromium and nickel exposure during pregnancy and impact on child outcomes. J Toxicol Environ Health A. 2015;78:1348–1368.
  • Shojaei-Brosseau T, Chompret A, Abel A, et al. Genetic epidemiology of neuroblastoma: a study of 426 cases at the Institut Gustave-Roussy in France. Pediatr Blood Cancer. 2004;42:99–105.
  • Trochet D, Bourdeaut F, Janoueix-Lerosey I, et al. Germline mutations of the paired-like homeobox 2B (PHOX2B) gene in neuroblastoma. Am J Hum Genet. 2004;74:761–764.
  • Rohrer T, Trachsel D, Engelcke G, et al. Congenital central hypoventilation syndrome associated with Hirschsprung’s disease and neuroblastoma: case of multiple neurocristopathies. Pediatr Pulmonol. 2002;33:71–76.
  • Mosse YP, Laudenslager M, Khazi D, et al. Germline PHOX2B mutation in hereditary neuroblastoma. Am J Hum Genet. 2004;75:727–730.
  • Clausen N, Andersson P, Tommerup N. Familial occurrence of neuroblastoma, von Recklinghausen’s neurofibromatosis, Hirschsprung’s agangliosis and jaw-winking syndrome. Acta Paediatr Scand. 1989;78:736–741.
  • Mosse YP, Laudenslager M, Longo L, et al. Identification of ALK as a major familial neuroblastoma predisposition gene. Nature. 2008;455:930–935.
  • Janoueix-Lerosey I, Lequin D, Brugieres L, et al. Somatic and germline activating mutations of the ALK kinase receptor in neuroblastoma. Nature. 2008;455:967–970.
  • George RE, Sanda T, Hanna M, et al. Activating mutations in ALK provide a therapeutic target in neuroblastoma. Nature. 2008;455:975–978.
  • Chen Y, Takita J, Choi YL, et al. Oncogenic mutations of ALK kinase in neuroblastoma. Nature. 2008;455:971–974.
  • 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. Clin Cancer Res. 2011;17:5082–5092.
  • Schleiermacher G, Javanmardi N, Bernard V, et al. Emergence of new ALK mutations at relapse of neuroblastoma. J Clin Oncol. 2014;32:2727–2734.
  • Bellini A, Bernard V, Leroy Q, et al. Deep sequencing reveals occurrence of subclonal ALK mutations in neuroblastoma at diagnosis. Clin Cancer Res. 2015;21:4913–4921.
  • Diskin SJ, Capasso M, Schnepp RW, et al. Common variation at 6q16 within HACE1 and LIN28B influences susceptibility to neuroblastoma. Nat Genet. 2012;44:1126–1130.
  • Capasso M, Devoto M, Hou C, et al. Common variations in BARD1 influence susceptibility to high-risk neuroblastoma. Nat Genet. 2009;41:718–723.
  • Wang K, Diskin SJ, Zhang H, et al. Integrative genomics identifies LMO1 as a neuroblastoma oncogene. Nature. 2011;469:216–220.
  • Whittle SB, Reyes S, Du M, et al. A polymorphism in the FGFR4 gene is associated with risk of neuroblastoma and altered receptor degradation. J Pediatr Hematol Oncol. 2016;38:131–138.
  • Maris JM, Mosse YP, Bradfield JP, et al. Chromosome 6p22 locus associated with clinically aggressive neuroblastoma. N Engl J Med. 2008;358:2585–2593.
  • Diskin SJ, Hou C, Glessner JT, et al. Copy number variation at 1q21.1 associated with neuroblastoma. Nature. 2009;459:987–991.
  • De Bernardi B, Balwierz W, Bejent J, et al. Epidural compression in neuroblastoma: diagnostic and therapeutic aspects. Cancer Lett. 2005;228:283–299.
  • Scheibel E, Rechnitzer C, Fahrenkrug J, et al. Vasoactive intestinal polypeptide (VIP) in children with neural crest tumours. Acta Paediatr Scand. 1982;71:721–725.
  • Matthay KK, Blaes F, Hero B, et al. Opsoclonus myoclonus syndrome in neuroblastoma: a report from a workshop on the dancing eyes syndrome at the advances in neuroblastoma meeting in Genoa, Italy, 2004. Cancer Lett. 2005;228:275–282.
  • Gorman MP. Update on diagnosis, treatment, and prognosis in opsoclonus-myoclonus-ataxia syndrome. Curr Opin Pediatr. 2010;22:745–750.
  • De Grandis E, Parodi S, Conte M, et al. Long-term follow-up of neuroblastoma-associated opsoclonus-myoclonus-ataxia syndrome. Neuropediatrics. 2009;40:103–111.
  • Catsman-Berrevoets CE, Aarsen FK, Van Hemsbergen ML, et al. Improvement of neurological status and quality of life in children with opsoclonus myoclonus syndrome at long-term follow-up. Pediatr Blood Cancer. 2009;53:1048–1053.
  • Sharp SE, Shulkin BL, Gelfand MJ, et al. 123I-MIBG scintigraphy and 18F-FDG PET in neuroblastoma. J Nucl Med. 2009;50:1237–1243.
  • Taggart DR, Han MM, Quach A, et al. Comparison of iodine-123 metaiodobenzylguanidine (MIBG) scan and [18F]fluorodeoxyglucose positron emission tomography to evaluate response after iodine-131 MIBG therapy for relapsed neuroblastoma. J Clin Oncol. 2009;27:5343–5349.
  • Bleeker G, Tytgat GAM, Adam JA, et al. 123I-MIBG scintigraphy and 18F-FDG-PET imaging for diagnosing neuroblastoma. Cochrane Database Syst Rev. 2015;9:CD009263. DOI:10.1002/14651858.CD009263.pub2
  • Cheung NK, Heller G, Kushner BH, et al. Detection of neuroblastoma in bone marrow by immunocytology: is a single marrow aspirate adequate? Med Pediatr Oncol. 1999;32:84–87.
  • Zage PE, Louis CU, Cohn SL. New aspects of neuroblastoma treatment: ASPHO 2011 symposium review. Pediatr Blood Cancer. 2012;58:1099–1105.
  • Shimada H, Ambros IM, Dehner LP, et al. Terminology and morphologic criteria of neuroblastic tumors: recommendations by the international neuroblastoma pathology committee. Cancer. 1999;86:349–363.
  • Peuchmaur M, d’Amore ES, Joshi VV, et al. Revision of the international neuroblastoma pathology classification: confirmation of favorable and unfavorable prognostic subsets in ganglioneuroblastoma, nodular. Cancer. 2003;98:2274–2281.
  • Navarro S, Amann G, Beiske K, et al. 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.
  • Brodeur GM, Seeger RC, Barrett A, et al. International criteria for diagnosis, staging, and response to treatment in patients with neuroblastoma. J Clin Oncol. 1988;6:1874–1881.
  • Brodeur GM, Pritchard J, Berthold F, et al. Revisions of the international criteria for neuroblastoma diagnosis, staging, and response to treatment. J Clin Oncol. 1993;11:1466–1477.
  • Cohn SL, Pearson AD, London WB, et al. The International Neuroblastoma Risk Group (INRG) classification system: an INRG task force report. J Clin Oncol. 2009;27:289–297.
  • Monclair T, Brodeur GM, Ambros PF, et al. The International Neuroblastoma Risk Group (INRG) staging system: an INRG task force report. J Clin Oncol. 2009;27:298–303.
  • Monclair T, Mosseri V, Cecchetto G, et al. Influence of image-defined risk factors on the outcome of patients with localised neuroblastoma. a report from the LNESG1 study of the European International Society of Paediatric Oncology Neuroblastoma group. Pediatr Blood Cancer. 2015;62:1536–1542.
  • Baker DL, Schmidt ML, Cohn SL, et al. Outcome after reduced chemotherapy for intermediate-risk neuroblastoma. New Engl J Med. 2010;363:1313–1323.
  • Strother DR, London WB, Schmidt ML, et al. Outcome after surgery alone or with restricted use of chemotherapy for patients with low risk neuroblastoma. J Clin Oncol. 2012;30:1842–1848.
  • Alvarado CS, London WB, Look AT, et al. Natural history and biology of stage A neuroblastoma: a Pediatric Oncology Group study. J Pediatr Hematol Oncol. 2000;22:197–205.
  • Perez CA, Matthay KK, Atkinson JB, et al. Biologic variables in the outcome of stages I and II neuroblastoma treated with surgery as primary therapy: a Childrens Cancer Group study. J Clin Oncol. 2000;18:18–26.
  • De Bernardi B, Mosseri V, Rubie H, et al. Treatment of localized resectable neuroblastoma. Results of the LNESG1 study by the SIOP Europe Neuroblastoma Group. Br J Cancer. 2008;99:1027–1033.
  • Nuchtern JG, London WB, Barnewolt CE, et al. A prospective study of expectant observation as primary therapy for neuroblastoma in young infants: a Children’s Oncology Group study. Ann Surgery. 2012;256:573–580.
  • Lavarino C, Cheung N-K, Garcia I, et al. Specific gene expression profiles and chromosomal abnormalities are associated with infant disseminated neuroblastoma. BMC Cancer. 2009;9:44.
  • Twist C, London WB, Naranjo A, et al. Maintaining Outstanding outcomes using response and biology based therapy for intermediate risk neuroblastoma: A report from the Children’s Oncology Group study ANBL0531. Presented at the Advances in Neuroblastoma Research Conference, Cologne, Germany, 2014 June.
  • Kohler JA, Rubie H, Castel V, et al. Treatment of children over the age of one year with unresectable localised neuroblastoma without MYCN amplification; results of the SIOPEN study. Eur J Cancer. 2013;49:3671–3679.
  • Schleiermacher G, Michon J, Ribero A, et al. Segmental chromosomal alterations lead to a higher risk of relapse in infant with MYCN-nonamplified localised unresectable/disseminiated neuroblastoma (a SIOPEN collaborative study). Br J Cancer. 2011;105:1940–1948.
  • Berthold F, Boos J, Burdach S, et al. Myeloablative megatherapy with autologous stem cell rescue versus oral maintenance chemotherapy as consolidation treatment in patients with high-risk neuroblastoma: a randomised controlled trial. Lancet Oncol. 2005;6:649–658.
  • Pearson AD, Pinkerton CR, Lewis IJ, et al. High-dose rapid and standard induction chemotherapy for patients aged over 1 year with stage 4 neuroblastoma: a randomised trial. Lancet Oncol. 2008;9:247–256.
  • 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. J Clin Oncol. 2009;27:1007–1013.
  • Kreissman SG, Seeger RC, Matthay KK, et al. Purged vs. non-purged peripheral blood stem cell transplantation for high risk neuroblastoma (COG A3973): a randomized phase III trial. Lancet Oncol. 2013;14:999–1008.
  • Yanik GA, Parisi MT, Shulkin BL, et al. Semiquantitative MIBG scoring as a prognostic indicator in patients with stage 4 neuroblastoma: a report from the Children’s Oncology Group. J Nucl Med. 2013;54:541–548.
  • Sorrentino S, Gigliotti AR, Sementa AR, et al. Neuroblastoma in the adult: the Italian experience with 21 patients. J Pediatr Hematol Oncol. 2014;36:e499–505.
  • Mosse YP, Deyell RJ, Berthold F, et al. Neuroblastoma in older children, adolescents and young adults: a report from the International Neuroblastoma Risk Group project. Pediatr Blood Cancer. 2014;61:627–635.
  • Park JR, Kreissman SG, London WB, et al. A Phase 3 Randomized Clinical Trial (RCT) of Tandem Myeloablative Autologous Stem Cell Transplant (ASCT) Using Peripheral Blood Stem Cell (PBSC) as Consolidation Therapy for High-Risk Neuroblastoma (HR-NB): A Children’s Oncology Group Study. Presented at the Advances in Neuroblastoma Research Conference, Cairns, Australia, 2016 June.
  • Schrey D, Vaidya SJ, Levine D, et al. Additional Therapies to improve metastatic response to induction therapy in Children with High Risk Neuroblastoma. J Pediatr Hematol Oncol. 2015;37:e 150–153.
  • Simon T, Haberle B, Hero B, et al. Role of Surgery in the treatment of patients with stage 4 Neuroblastoma age 18 months or older at diagnosis. J Clin Oncol. 2013;31:752–758.
  • Du L, Liu L, Zhang C, et al. Role of Surgery in the treatment of patients with high risk neuroblastoma who have poor response to induction chemotherapy. J Pediatr Surg. 2014;49:528–533.
  • Castel V, Tovar JA, Costa E, et al. The Role of Surgery in Stage IV Neuroblastoma. J Pediatr Surg. 2002;37:1574–1578.
  • Mazloom A, Louis CU, Nuchtern J, et al. Radiation Therapy to the primary and post induction chemotherapy MIBG avid sites in high risk neuroblastoma. Int J Radiat Oncol Biol Phys. 2014;90:858–862.
  • Rombi B, MacDonald SM, Maurizio A, et al. Proton radiotherapy for childhood tumors: an overview of early clinical results. J Nucl Med Radiat Ther. 2013;4:4.
  • Doyen J, Falk AT, Floquet V, et al. Proton beams in cancer treatments: clinical outcomes and dosimetric comparisons with photon therapy. Cancer Treat Rev. 2016;43:104–112.
  • Hattangadi JA, Rombi B, Yock TI, et al. Proton Radiotherapy for High-Risk Pediatric Neuroblastoma: early Outcomes and Dose Comparison. Int J Radiat Oncol Biol Phys. 2012;83:1015–1022.
  • Oshiro Y, Mizumoto M, Okkumura T, et al. Clinical Results of Proton Beam Therapy for Advanced Neuroblastoma. Radiat Oncol. 2013;8:142.
  • Yalcin B, Kremer LCM, van Dalen EC. High dose chemotherapy and autologous hematopoietic stem cell rescue for children with high risk neuroblastoma (Review). Cochrane Database Syst Rev. 2015;10:CD006301. doi: 10.1002/14651858.CD006301.pub4.
  • Grupp SA, Cohn SL, Wall D, et al. Collection, storage, and infusion of stem cells in children with high-risk neuroblastoma: saving for a rainy day. Pedaitr Blood Cancer. 2006;46:719–722.
  • Fish JD, Grupp SA. Stem cell transplantation for neuroblastoma. Bone Marrow Transplant. 2008;41:159–165.
  • Ladenstein R, Poetschger U, Luksch R, et al. Busulfan-Melphalan as a myeloablative therapy (MAT) for high risk neuroblastoma: results from HR-NBL1/SIOPEN trial. J Clin Oncol. 2011;29:2.
  • Soni S, Pai V, Gross TG, et al. Busulfan and Melphalan as consolidation therapy with autologous peripheral blood stem cell transplantation following COG induction platform for high risk neuroblastoma; early results from a single institution. Pediatr Trans. 2014;18:217–220.
  • Seif AE, Naranjo A, Baker DL, et al. A pilot study of tandem high dose chemotherapy with stem cell rescue as consolidation for high risk neuroblastoma: Children’s Oncology Group study ANBL00P1. Bone Marrow Transplant. 2013;48:947–952.
  • Pasqualani C, Dufour C, Goma G, et al. Tandem high dose chemotherapy with thiotepa and busulfan-melphalan and autologous stem cell transplantation in very high risk neuroblastoma patients. Bone Marrow Transplant. 2016;51:227–231.
  • Kushner BH, Ostrovnaya I, Cheung IY, et al. Lack of survival advantage with autologous stem-cell transplantation in high-risk neuroblastoma consolidated by anti-GD2 immunotherapy and isotretinoin. Oncotarget. 2016;7:4155–4166.
  • 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. N Engl J Med. 1999;341:1165–1173.
  • Yu AL, Gilman AL, Ozkaynak F, et al. Anti-GD2 antibody with GM-CSF, interleukin-2, and isotretinoin for neuroblastoma. N Engl J Med. 2010;363:1324–1334.
  • Cheung NK, Kushner BH, Cheung IY, et al. Anti-GD2 antibody treatment of minimal residual stage 4 neuroblastoma diagnosed at more than 1 year of age. J Clin Oncol. 1998;16:3053–3060.
  • Cheung NK, Cheung IY, Kushner BH, et al. Murine anti-GD2 monoclonal antibody 3F8 combined with granulocyte-macrophage colony-stimulating factor and 13-cis-retinoic acid in high-risk patients with stage 4 neuroblastoma in first remission. J Clin Oncol. 2012;30:3264–3270.
  • Hogarty MD, Norris MD, Davis K, et al. ODC1 is a critical determinant of MYCN oncogenesis and a therapeutic target in neuroblastoma. Cancer Res. 2008;68:9735–9745.
  • Geerts D, Koster J, Albert D, et al. The polyamine metabolism genes ornithine decarboxylase and antizyme 2 predict aggressive behavior in neuroblastomas with and without MYCN amplification. Int J Cancer. 2010;126:2012–2024.
  • Sholler G, Ferguson W, Bergendahl G, et al. A phase II prevention trial of DFMO in patients with high risk neuroblastoma in remission prevents relapse and increases event free and overall survival. Presented at the American Society of Pediatric Hematology/Oncology annual meeting. Minneapolis. 2016 May.
  • Sholler G, Ferguson W, Bergandahl G, et al. DFMO maintains remission and increases overall survival in high-risk neuroblastoma: results of a phase ii prevention trial. Presented at the Advances in Neuroblastoma Research Conference, Cairns, Australia, 2016 June.
  • London WB, Castel V, Monclair T, et al. Clinical and biologic features predictive of survival after relapse of neuroblastoma: a report from the International Neuroblastoma Risk Group project. J Clin Oncol. 2011;29:3286–3292.
  • Lau L, Tai DWeitzman S, et al. Factors influencing survival in children with recurrent neuroblastoma. J Pediatr Hematol Oncol. 2004;26:227–232.
  • Garaventa A, Parodi S, De Bernardi B, et al. Outcome of children with neuroblastoma after progression or relapse. A retrospective study of the Italian neuroblastoma registry. Eur J Cancer. 2009;45:2835–2842.
  • Moreno L, Rubie H, Varo A, et al.Outcome of children with relapsed or refractory neuroblastoma: A meta-analysis of ITCC/SIOPEN European phase II clinical trials. Pediatr Blood Cancer. 2017; 64:25–31.
  • Modak S Generation and administration of autologous T cells transduced with a 3rd generation GD2 chimeric antigen receptor for patients with relapsed or refractory neuroblastoma. Presented at the Advances in Neuroblastoma Research Conference, Cologne, Germany,2014 June.
  • Houghton PJ, Cheshire PJ, Hallman JD, et al. Efficacy of topoisomerase I inhibitors, topotecan and irinotecan, administered at low dose levels in protracted schedules to mice bearing xenografts of human tumors. Cancer Chemother Pharmacol. 1995;36:393–403.
  • Carol H, Houghton PJ, Morton CL, et al. Initial testing of topotecan by the pediatric preclinical testing program. Pediatr Blood Cancer. 2010;54(7): 07–715.
  • Blaney S, Berg SL, Pratt C, et al. A phase I study of irinotecan in pediatric patients: a Pediatric Oncology Group study. Clin Cancer Res. 2001;7:32–37.
  • Blaney SM, Needle MN, Gillespie A, et al. Phase II trial of topotecan administered as 72-hour continuous infusion in children with refractory solid tumors: a Collaborative Pediatric Branch, National Cancer Institute, and Children’s Cancer Group study. Clin Cancer Res. 1998;4:357–360.
  • Hawkins DS, Bradfield S, Whitlock JA, et al. Topotecan by 21-day continuous infusion in children with relapsed or refractory solid tumors: a Children’s Oncology Group study. Pediatr Blood Cancer. 2006;47:790–794.
  • Bomgaars LR, Bernstein M, Krailo M, et al. Phase II trial of irinotecan in children with refractory solid tumors: a Children’s Oncology Group study. J Clin Oncol. 2007;25:4622–4627.
  • Saylors RL, Stine KC, Sullivan J, et al. Cyclophosphamide plus topotecan in children with recurrent or refractory solid tumors: a Pediatric Oncology Group phase II study. J Clin Oncol. 2001;19:3463–3469.
  • London WB, Frantz CN, Campbell LA, et al. Phase II randomized comparison of topotecan plus cyclophosphamide versus topotecan alone in children with recurrent or refractory neuroblastoma: a Children’s Oncology Group study. J Clin Oncol. 2010;28:3808–3815.
  • Kushner BH, Kramer K, Modak S, et al. Differential impact of high-dose cyclophosphamide, topotecan, and vincristine in clinical subsets of patients with chemoresistant neuroblastoma. Cancer. 2010;116:3054–3060.
  • Garaventa A, Luksch R, Biasotti S, et al. A phase II study of topotecan with vincristine and doxorubicin in children with recurrent/refractory neuroblastoma. Cancer. 2003;98:2488–2494.
  • Kushner BH, Kramer K, Modak S, et al. Irinotecan plus temozolomide for relapsed or refractory neuroblastoma. J Clin Oncol. 2006;24:5271–5276.
  • Bagatell R, London WB, Wagner LM, et al. Phase II study of irinotecan and temozolomide in children with relapsed or refractory neuroblastoma: a Children’s Oncology Group study. J Clin Oncol. 2011;29:208–213.
  • Kushner BH, Modak S, Kramer K, et al. Ifosfamide, carboplatin, and etoposide for neuroblastoma: a high-dose salvage regimen and review of the literature. Cancer. 2013;119:665–667.
  • Adbel Rahman H, Moussa EAH, Zekri WZK, et al. Did Salvage ICE chemotherapy improve the outcome in primary resistant/relapsing stage III/IV neuroblastoma? J Egypt Nat Cancer Inst. 2011;23:47–53.
  • Donfrancesco A, Jenkner A, Castellano A, et al. Ifosfamide/carboplatin/etoposide (ICE) as front-line, topotecan/cyclophosphamide as second-line and oral temozolomide as third-line treatment for advanced neuroblastoma over one year of age. Acta Pediatr Suppl. 2004;445:6–11.
  • Hutchinson RJ, Sisson JC, Shapiro B, et al. 131-I-metaiodobenzylguanidine treatment in patients with refractory advanced neuroblastoma. Am J Clin Oncol. 1992;15:226–232.
  • Klingebiel T, Berthold F, Treuner J, et al. Metaiodobenzylguanidine (MIBG) in treatment of 47 patients with neuroblastoma: results of the German neuroblastoma trial. Med Pediatr Oncol. 1991;19:84–88.
  • Lashford LS, Lewis IJ, Fielding SL, et al. Phase I/II study of iodine 131 metaiodobenzylguanidine in chemoresistant neuroblastoma: a United Kingdom Children’s Cancer Study Group investigation. J Clin Oncol. 1992;10:1889–1896.
  • 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. J Clin Oncol. 2007;25:1054–1060.
  • Caren H, Abel F, Kogner P, et al. High incidence of DNA mutations and gene amplifications of the ALK gene in advanced sporadic neuroblastoma tumors. Biochem J. 2008;416:153–159.
  • Passoni L, Longo L, Collini P, et al. Mutation-independent anaplastic lymphoma kinase overexpression in poor prognosis neuroblastoma patients. Cancer Res. 2009;69:7338–7346.
  • Mosse YP, Lim MS, Voss SD, et al. Safety and activity of crizotinib for paediatric patients with refractory solid tumours or anaplastic large-cell lymphoma: a Children’s Oncology Group phase 1 consortium study. Lancet Oncol. 2013;14:472–480.
  • Moore NF, Azarova AM, Bhatnagar N, et al. Molecular rationale for the use of PI3K/AKT/mTOR pathway inhibitors in combination with crizotinib in ALK-mutated neuroblastoma. Oncotarget. 2014;5:8737–8749.
  • 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 Res. 2010;70:10038–10043.
  • Berry T, Luther W, Bhatnagar N, et al. The ALK(F1174L) mutation potentiates the oncogenic activity of MYCN in neuroblastoma. Cancer Cell. 2012;22:117–130.
  • Zhou H, Kuang J, Zhong L, et al. Tumour amplified kinase STK15/BTAK induces centrosome amplification, aneuploidy, and transformation. Nat Genet. 1998;20:189–193.
  • Sen S, Katayama H, Sasai K. Functional significance of aurora kinase A in centrosome amplification and genomic instability. Adv Exp Med Biol. 2008;617:99–108.
  • Shang X, Burlingame SM, Okcu MF, et al. Aurora A is a negative prognostic factor and a new therapeutic target in human neuroblastoma. Mol Cancer Ther. 2009;8:2461–2469.
  • 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.
  • Mosse´ YP, Lipsitz E, Fox E, et al. Pediatric phase I trial and pharmacokinetic study of MLN8237, an investigational oral selective small-molecule inhibitor of Aurora kinase A: a Children’s Oncology Group phase I consortium study. Clin Cancer Res. 2012;18:6058–6064.
  • Dubois SG, Marachelian A, Fox E, et al. Phase I study of the aurora A kinase inhibitor alisertib in combination with irinotecan and temozolomide for patients with relapsed or refractory neuroblastoma: A NANT (New Approaches to Neuroblastoma Therapy) Trial. J Clin Oncol. 2016;34:1368–1375.
  • Saulnier Sholler GL, Gerner EW, Bergendahl G, et al. A phase I trial of DFMO targeting polyamine addiction in patients with relapsed/refractory neuroblastoma. Plos ONE. 2015;10:e0127246.
  • Peirce SK, Findley HW, Prince C, et al. The PI-3 kinase-Akt-MDM2-survivin signaling axis in high-risk neuroblastoma: a target for PI-3 kinase inhibitor intervention. Cancer Chemother Pharmacol. 2011;68:325–335.
  • Bern C. Antitrypanosomal therapy for chronic Chagas’ disease. New Engl J Med. 2011;364:2527–2534.
  • Rassi A, Rassi A, Marin-Neto JA. Chagas disease. Lancet. 2010;375:1388–1402.
  • Saulnier Sholler G, Brard L, Straub JA, et al. Nifurtimox induces apoptosis of neuroblastoma cells in vitro and in vivo. J Pediatr Hematol Oncol. 2009;31:187–193.
  • Du M, Zhang L, Scorsone KA, et al. Nifurtimox is effective against neural tumor cells and is synergistic with buthionine sulfoximine. Sci Rep. 2016;6:27458.
  • Saulnier Sholler GL, Bergendahl GM, Brard L, et al. A phase 1 study of nifurtimox in patients with relapsed/refractory neuroblastoma. J Pediatr Hematol Oncol. 2011;33:25–30.
  • Saulnier Sholler GL, Ferguson W, Bergendahl G, et al. A pilot trial testing the feasibility of using molecular-guided therapy in patients with recurrent neuroblastoma. J Cancer Ther. 2012;3:602–612.
  • Saulnier Sholler GL, Bond JP, Bergendahl G, et al. Feasibility of implementing molecular-guided therapy for the treatment of patients with relapsed or refractory neuroblastoma. Cancer Med. 2015;4:871–886.
  • Mody RJ, Wu Y-M, Lonigro RJ, et al. Integrative clinical sequencing in the management of refractory or relapsed cancer in youth. JAMA. 2015;314:913–925.
  • Cha A, Asgharzedeh S, Czarnecki S, et al. Dinutuximab combined with chemotherapy in patients with multiply relapsed/refractory high risk neuroblastoma (HR-NBL). Presented at the Advances in Neuroblastoma Research Conference, Cairns, Australia, 2016 June.
  • Shusterman S, London WB, Gillies SD, et al. Antitumor activity of hu14.18-IL2 in patients with relapsed/refractory neuroblastoma: a Children’s Oncology Group (COG) phase II study. J Clin Oncol. 2010;28:4969–4975.
  • Delgado DC, Hank JA, Kolesar J, et al. Genotypes of NK cell KIR receptors, their ligands, and Fcγ receptors in the response of neuroblastoma patients to Hu14.18-IL2 immunotherapy. Cancer Res. 2010;70:9554–9561.
  • Cheung NK, Sowers R, Vickers AJ, et al. FCGR2A polymorphism is correlated with clinical outcome after immunotherapy of neuroblastoma with anti-GD2 antibody and granulocyte macrophage colony stimulating factor. J Clin Oncol. 2006;24:2885–2890.
  • Merchant MS, Baird K, Wexler LH, et al. Ipilimumab: first results of a phase I trial in pediatric patients with advanced solid tumors. J Clin Oncol. 2012;30:9545.
  • Russell HV, Strother D, Mei Z, et al. Phase I trial of vaccination with autologous neuroblastoma tumor cells genetically modified to secrete IL-2 and lymphotactin. J Immunother. 2007;30:227–233.
  • Krishnadas DK, Shapiro T, Lucas K. Complete remission following decitabine/dendritic cell vaccine for relapsed neuroblastoma. Pediatrics. 2013;131:e336–e341.
  • Kanold J, Paillard C, Tchirkov A, et al. NK Cell immunotherapy for high-risk neuroblastoma relapse after haploidentical HSCT. Pediatr Blood Cancer. 2012;59:739–742.
  • Lee DW, Barrett DM, Mackall C, et al. The future is now: chimeric antigen receptors as new targeted therapies for childhood cancer. Clin Cancer Res. 2012;18:2780–2790.
  • Louis CU, Savoldo B, Dotti G, et al. Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. Blood. 2011;118:6050–6056.
  • Pule MA, Savoldo B, Myers GD, et al. Virus-specific T cells engineered to coexpress tumor-specific receptors: persistence and antitumor activity in individuals with neuroblastoma. Nat Med. 2008;14:1264–1270.
  • Jena B, Dotti G, Cooper LJN. Redirecting T-cell specificity by introducing a tumor-specific chimeric antigen receptor. Blood. 2010;116:1035–1044.
  • Pession A, Tonelli R. The MYCN oncogene as a specific and selective drug target for peripheral and central nervous system tumors. Curr Cancer Drug Targets. 2005;5:273–283.
  • Puissant A, Frumm SM, Alexe G, et al. Targeting MYCN in neuroblastoma by BET bromodomain inhibition. Cancer Discov. 2013;3:308–323.
  • Shukla N, Ameur N, Yilman I, et al. Oncogene mutation profiling of pediatric solid tumors reveals significant subsets of embryonal rhabdomyosarcoma and neuroblastoma with mutated genes in growth signaling pathways. Clin Cancer Res. 2011;18:748–757.
  • Eleveld TF, Oldridge DA, Bernard V, et al. Relapsed neuroblastomas show frequent RAS-MAPK pathway mutations. Nat Gen. 2015;47:864–871.
  • 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;142:218–229.
  • Woodfield SE, Zhang L, Scorsone K, et al. Binimetinib Inhibits MEK and is effective against neuroblastoma tumor cells with low NF1 expression. BMC Cancer. 2016;16:172.
  • Oppenheimer O, Cheung N-K, Gerald WL. The RET oncogene is a critical component of transcriptional programs associated with retinoic acid-induced differentiation in neuroblastoma. Mol Cancer Ther. 2007;6:1300–1309.
  • Zage PE, Zeng L, Palla S, et al. A novel therapeutic combination for neuroblastoma: the VEGFR/EGFR/RET inhibitor vandetanib with 13-cis-retinoic acid. Cancer. 2010;116:2465–2475.
  • Grinshtein N, Datti A, Fujitani N, et al. Small molecule kinase inhibitor screen identifies polo-like kinase 1 as a target for neuroblastoma tumor-initiating cells. Cancer Res. 2011;71:1385–1395.
  • 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. Proc Natl Acad Sci USA. 2011;108:3336–3341.
  • Balamuth NJ, Wood A, Wang Q, et al. Serial transcriptome analysis and cross-species integration identifies centromere-associated protein E as a novel neuroblastoma target. Cancer Res. 2010;70:2749–2758.
  • Cohen LE, Gordon JH, Popovsky EY, et al. Late effects in children treated with intensive multimodal therapy for high-risk neuroblastoma: high incidence of endocrine and growth problems. Bone Marrow Transplant. 2014;49:502–508.
  • Moreno L, Vaidya SJ, Pinkerton CR, et al. Long-term follow-up of children with high-risk neuroblastoma: the ENSG5 trial experience. Pediatr Blood Cancer. 2013;60:1135–1140.
  • Laverdiere C, Cheung NK, Kushner BH, et al. Long-term complications in survivors of advanced stage neuroblastoma. Pediatr Blood Cancer. 2005;45:324–332.
  • Trahair TN, Vowels MR, Johnston K, et al. Long-term outcomes in children with high-risk neuroblastoma treated with autologous stem cell transplantation. Bone Marrow Transplant. 2007;40:741–746.
  • Laverdière C, Liu Q, Yasui Y, et al. Long-term outcomes in survivors of neuroblastoma: a report from the Childhood Cancer Survivor Study. J Natl Cancer Inst. 2009;101:1131–1140.
  • Hobbie WL, Moshang T, Carlson CA, et al. Late effects in survivors of tandem peripheral blood stem cell transplant for high-risk neuroblastoma. Pediatr Blood Cancer. 2008;51:679–683.
  • Fawzy M, El-Beltagy M, Shafei ME, et al. Intraspinal neuroblastoma: treatment options and neurological outcome of spinal cord compression. Oncol Lett. 2015;9:907–911.
  • Applebaum MA, Henderson TO, Lee SM, et al. Second malignancies in patients with neuroblastoma: the effects of risk-based therapy. Pediatr Blood Cancer. 2015;62:128–133.
  • Federico SM, Allewelt HB, Spunt SL, et al. Subsequent malignant neoplasms in pediatric patients initially diagnosed with neuroblastoma. J Pediatr Hematol Oncol. 2015;37:e6–e12.
  • Martin A, Schneiderman J, Helenowski IB, et al. Secondary malignant neoplasms after high-dose chemotherapy and autologous stem cell rescue for high-risk neuroblastoma. Pediatr Blood Cancer. 2014;61:1350–1356.
  • Polishchuk AL, DuBois SG, Haas-Kogan D, et al. Response, survival, and toxicity after iodine-131-metaiodobenzylguanidine therapy for neuroblastoma in preadolescents, adolescents, and adults. Cancer. 2011;117:4286–4293.
  • Quach A, Ji L, Mishra V, et al. Thyroid and hepatic function after high-dose 131I metaiodobenzylguanidine (131I-MIBG) therapy for neuroblastoma. Pediatr Blood Cancer. 2011;56:191–201.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.