251
|
Decock A, Ongenaert M, De Wilde B, Brichard B, Noguera R, Speleman F, Vandesompele J. Stage 4S neuroblastoma tumors show a characteristic DNA methylation portrait. Epigenetics 2016; 11:761-771. [PMID: 27599161 DOI: 10.1080/15592294.2016.1226739] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Stage 4S neuroblastoma (NB) is a special type of NB found in infants with metastases at diagnosis and is associated with an excellent outcome due to its remarkable capacity to undergo spontaneous regression. As genomics have not been able to explain this intriguing clinical presentation, we here aimed at profiling the DNA methylome of stage 4S NB to better understand this phenomenon. To this purpose, differential methylation analyses between International Neuroblastoma Staging System (INSS) stage 4S, stage 4 and stage 1/2 were performed, using methyl-CpG-binding domain (MBD) sequencing data of 14 stage 4S, 14 stage 4, and 13 stage 1/2 primary NB tumors (all MYCN non-amplified in order not to confound results). Stage 4S-specific hyper- and hypomethylated promoters were determined and further characterized for genomic localization and function by cytogenetic band enrichment, gene set enrichment, transcription factor target enrichment and differential RNA expression analyses. We show that specific chromosomal locations are enriched for stage 4S differentially methylated promoters and that stage 4S tumors show characteristic hypermethylation of specific subtelomeric promoters. Furthermore, genes involved in important oncogenic pathways, in neural crest development and differentiation, and in epigenetic processes are differentially methylated and expressed in stage 4S tumors. Based on these findings, we describe new biological mechanisms possibly contributing to the stage 4S-specific tumor biology and spontaneous regression. In conclusion, this study is the first to describe the highly characteristic stage 4S DNA methylome. These findings will open new avenues to further unravel the NB pathology in general and stage 4S disease specifically.
Collapse
Affiliation(s)
- Anneleen Decock
- a Center for Medical Genetics, Ghent University , Ghent , Belgium.,b Cancer Research Institute Ghent (CRIG) , Ghent , Belgium
| | - Maté Ongenaert
- a Center for Medical Genetics, Ghent University , Ghent , Belgium
| | - Bram De Wilde
- a Center for Medical Genetics, Ghent University , Ghent , Belgium.,b Cancer Research Institute Ghent (CRIG) , Ghent , Belgium.,c Department of Pediatric Hematology and Oncology , Ghent University Hospital , Ghent , Belgium
| | - Bénédicte Brichard
- d Cliniques Universitaires Saint-Luc, Université Catholique de Louvain , Brussels , Belgium
| | - Rosa Noguera
- e Department of Pathology , Medical School, University of Valencia, and Health Research Institute INCLIVA , Valencia , Spain
| | - Frank Speleman
- a Center for Medical Genetics, Ghent University , Ghent , Belgium.,b Cancer Research Institute Ghent (CRIG) , Ghent , Belgium
| | - Jo Vandesompele
- a Center for Medical Genetics, Ghent University , Ghent , Belgium.,b Cancer Research Institute Ghent (CRIG) , Ghent , Belgium.,f Bioinformatics Institute Ghent - From Nucleotides to Networks (BIG N2N) , Ghent , Belgium
| |
Collapse
|
252
|
Friedman DL. High-risk neuroblastoma: challenges in management in low- and middle-income countries. Pediatr Transplant 2016; 20:742-3. [PMID: 27501322 DOI: 10.1111/petr.12775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Debra L Friedman
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
253
|
Marrano P, Irwin MS, Thorner PS. Heterogeneity of MYCN amplification in neuroblastoma at diagnosis, treatment, relapse, and metastasis. Genes Chromosomes Cancer 2016; 56:28-41. [PMID: 27465929 DOI: 10.1002/gcc.22398] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 07/24/2016] [Accepted: 07/25/2016] [Indexed: 12/14/2022] Open
Abstract
Amplification of the MYCN gene in neuroblastoma is associated with a poor prognosis and is considered to remain unchanged in post-treatment specimens and metastases. While heterogeneity of MYCN copy number in tumor cells has been reported, serial samples have only been studied in a limited way, and the biologic relevance of this finding is not well understood. We used in situ hybridization on paraffin sections of 102 specimens from 30 patients with MYCN-amplified neuroblastoma to determine MYCN copy number in the primary tumor, pre- and post-treatment, and in metastatic samples. Nineteen cases (63%) showed diffuse MYCN amplification in all samples tested. Nine cases (30%) showed a reduction in MYCN copy number: five cases with diffuse amplification subsequently showed focal amplification, one case with diffuse MYCN amplification showed MYCN gain after treatment, and three focally amplified cases were non-amplified in later specimens. In two cases (7%), focal amplification became diffuse in subsequent samples. Histology was not predictive of the temporal or spatial pattern of MYCN amplification for a particular tumor. If extent of amplification (focal vs. diffuse) is not considered, 26/30 (87%) of cases were consistently MYCN-amplified. However, our data suggest that MYCN status can be heterogeneous between tumor sites, during tumor progression or following treatment, challenging the notion that MYCN copy number does not change for a particular neuroblastoma. Assessing the biologic significance of MYCN heterogeneity will require larger studies of clinically annotated tumor samples, and will depend on interpreting heterogeneity in MYCN status in combination with other genetic changes. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Paula Marrano
- Department of Pediatric Laboratory Medicine, the Hospital for Sick Children, Toronto, Canada
| | - Meredith S Irwin
- Department of Pediatrics, the Hospital for Sick Children, Toronto, Canada.,Department of Pediatrics, University of Toronto, Toronto, Canada
| | - Paul S Thorner
- Department of Pediatric Laboratory Medicine, the Hospital for Sick Children, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| |
Collapse
|
254
|
Serum-Based Quantification of MYCN Gene Amplification in Young Patients with Neuroblastoma: Potential Utility as a Surrogate Biomarker for Neuroblastoma. PLoS One 2016; 11:e0161039. [PMID: 27513929 PMCID: PMC4981470 DOI: 10.1371/journal.pone.0161039] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 07/28/2016] [Indexed: 11/19/2022] Open
Abstract
We previously developed a method for determining MYCN gene amplification status using cell-free DNA fragments released from cancer cells into the blood of patients with neuroblastoma (NB). Here, we analyzed the relationship between MYCN amplification (MNA) status and neuroblastoma prognosis. We screened serum samples from 151 patients with NB for MNA, using real-time quantitative PCR, and compared the results with MYCN status determined using paired tumor samples. We additionally investigated whether MNA status correlates with patient survival. When a cut-off value of 5 was used, serum-based MNA analysis was found to show good sensitivity (86%) and very high specificity (95%). The sensitivities for stage 1 and 2 might be acceptable, even though it is not as good as for stage 3 and 4 (67% for stage 1 and 2, 92% for stage 3, and 87% for stage 4). MNA status correlated with overall survival in our cohort of 82 patients, with survival data available (p < 0.01). The hazard ratio of MNA status was 4.98 in patients diagnosed at less than 18 months of age (95% confidence interval, 1.00–24.78), and 1.41 (95% confidence interval, 0.63–3.14) for those diagnosed at 18 months of age or older. Serum-based MNA analysis is rapid and non-invasive compared with tumor-based MNA analysis, and has potential to predict tumor MNA status. There is still a room to improve the sensitivity of the test for tumors of stages 1 and 2, nonetheless this assay might help to determine therapeutic strategies prior to tumor biopsy, especially for patients with a life-threatening condition, as well as for patients of less than 18 months of age whose risk-grouping and treatment allocation depends on their MNA status.
Collapse
|
255
|
He J, Wang F, Zhu J, Zhang R, Yang T, Zou Y, Xia H. Association of potentially functional variants in the XPG gene with neuroblastoma risk in a Chinese population. J Cell Mol Med 2016; 20:1481-1490. [PMID: 27019310 PMCID: PMC4956948 DOI: 10.1111/jcmm.12836] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 02/14/2016] [Indexed: 02/05/2023] Open
Abstract
XPG gene plays a critical role in the nucleotide excision repair pathway. However, the association between XPG gene polymorphisms and neuroblastoma risk has not been investigated. In this study with 256 neuroblastoma cases and 531 cancer-free controls, we investigated the effects of five potentially functional polymorphisms (rs2094258 C>T, rs751402 C>T, rs2296147 T>C, rs1047768 T>C and rs873601G>A) on neuroblastoma risk. We calculated odds ratio (OR) and 95% confidence interval (CI) to evaluate the association between the five selected polymorphisms and neuroblastoma risk. False-positive report probability (FPRP) was utilized to determine whether significant findings were noteworthy or because of a chance. We also performed genotype-phenotype association analysis to explore the biological plausibility of our findings. We found that the rs2094258 T allele was significantly associated with decreased neuroblastoma risk (CT versus CC: adjusted OR = 0.65, 95% CI = 0.47-0.90, P = 0.010; and CT/TT versus CC: adjusted OR = 0.71, 95% CI = 0.53-0.97, P = 0.030) after adjusting for age and gender. The association was more prominent for subjects with retroperitoneal tumour or early-stage tumour. We also found that carriers of the 2-3 risk genotypes had a significantly increased neuroblastoma risk when compared to carriers of the 0-1 risk genotypes. The association with risk genotypes was more predominant in older children, females and subjects with retroperitoneal tumour or early stage. Our results were further supported by FPRP analysis and genotype-phenotype association analysis. In conclusion, our study verified that the XPG gene rs2094258 C>T polymorphism may contribute to neuroblastoma susceptibility. Our findings require further validation by studies with larger sample size and concerning different ethnicities.
Collapse
Affiliation(s)
- Jing He
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Department of Experimental Research, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Fenghua Wang
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jinhong Zhu
- Molecular Epidemiology Laboratory and Department of Laboratory Medicine, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Ruizhong Zhang
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Tianyou Yang
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yan Zou
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Huimin Xia
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
256
|
Rached J, Nasr Z, Abdallah J, Abou-Antoun T. L1-CAM knock-down radiosensitizes neuroblastoma IMR-32 cells by simultaneously decreasing MycN, but increasing PTEN protein expression. Int J Oncol 2016; 49:1722-30. [PMID: 27432152 DOI: 10.3892/ijo.2016.3625] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 06/28/2016] [Indexed: 11/06/2022] Open
Abstract
Childhood neuroblastoma is one of the most malignant types of cancers leading to a high mortality rate. These cancerous cells can be highly metastatic and malignant giving rise to disease recurrence and poor prognosis. The proto-oncogene myelocytomatosis neuroblastoma (MycN) is known to be amplified in this type of cancer, thus, promoting high malignancy and resistance. The L1 cell adhesion molecule (L1-CAM) cleavage has been found upregulated in many types of malignant cancers. In the present study, we explored the interplay between L1-CAM, MycN and PTEN as well as the role played by PDGFR and VEGFR on tumorigenicity in neuroblastoma cells. We investigated the effect of L1-CAM knock-down (KD) and PDGFR/VEGFR inhibition with sunitinib malate (Sutent®) treatment on subsequent tumorsphere formation and cellular proliferation and migration in the MycN-amplified IMR-32 neuroblastoma cells. We further examined the effect of combined L1-CAM KD with Sutent treatment or radiotherapy on these cellular functions in our cells. Tumorsphere formation is one of the indicators of aggressiveness in malignant cancers, which was significantly inhibited in IMR-32 cells after L1-CAM KD or Sutent treatment, however, no synergistic effect was observed with dual treatments, rather L1-CAM KD alone showed a greater inhibition on tumorsphere formation compared to Sutent treatment alone. In addition, cellular proliferation and migration were significantly inhibited after L1-CAM KD in the IMR-32 cells with no synergistic effect observed on the rate of cell proliferation when combined with Sutent treatment. Again, L1-CAM KD alone exhibited greater inhibitory effect than Sutent treatment on cell proliferation. L1-CAM KD led to the simultaneous downregulation of MycN, but the upregulation of PTEN protein expression. Notably, radiotherapy (2 Gy) of the IMR-32 cells led to significant upregulation of both L1-CAM and MycN, which was abrogated with L1-CAM KD in our cells. In addition, L1-CAM KD radiosensitized the cells as exhibited by the synergistic effect on the reduction in cell proliferation compared to radiotherapy alone. Taken together, our data show the importance of L1-CAM interplay with MycN and PTEN on the MycN amplified neuroblastoma cell radioresistance, proliferation and motility.
Collapse
Affiliation(s)
- Johnny Rached
- Faculty of Sciences, University of Balamand, Koura, Lebanon
| | - Zeina Nasr
- Faculty of Sciences, University of Balamand, Koura, Lebanon
| | - Jad Abdallah
- School of Pharmacy, Pharmaceutical Sciences Department, Lebanese American University, Byblos, Lebanon
| | - Tamara Abou-Antoun
- School of Pharmacy, Pharmaceutical Sciences Department, Lebanese American University, Byblos, Lebanon
| |
Collapse
|
257
|
Maas SM, Vansenne F, Kadouch DJM, Ibrahim A, Bliek J, Hopman S, Mannens MM, Merks JHM, Maher ER, Hennekam RC. Phenotype, cancer risk, and surveillance in Beckwith-Wiedemann syndrome depending on molecular genetic subgroups. Am J Med Genet A 2016; 170:2248-60. [PMID: 27419809 DOI: 10.1002/ajmg.a.37801] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 05/29/2016] [Indexed: 12/22/2022]
Abstract
Patients with Beckwith-Wiedemann syndrome (BWS) have an increased risk to develop cancer in childhood, especially Wilms tumor and hepatoblastoma. The risk varies depending on the cause of BWS. We obtained clinical and molecular data in our cohort of children with BWS, including tumor occurrences, and correlated phenotype and genotype. We obtained similar data from larger cohorts reported in the literature. Phenotype, genotype and tumor occurrence were available in 229 of our own patients. Minor differences in phenotype existed depending on genotype/epigenotype, similar to earlier studies. By adding patients from the literature, we obtained data on genotype and tumor occurrence of in total 1,971 BWS patients. Tumor risks were highest in the IC1 (H19/IGF2:IG-DMR) hypermethylation subgroup (28%) and pUPD subgroup (16%) and were lower in the KCNQ1OT1:TSS-DMR (IC2) subgroup (2.6%), CDKN1C (6.9%) subgroup, and the group in whom no molecular defect was detectable (6.7%). Wilms tumors (median age 24 months) were frequent in the IC1 (24%) and pUPD (7.9%) subgroups. Hepatoblastoma occurred mostly in the pUPD (3.5%) and IC2 (0.7%) subgroups, never in the IC1 and CDKN1C subgroups, and always before 30 months of age. In the CDKN1C subgroup 2.8% of patients developed neuroblastoma. We conclude tumor risks in BWS differ markedly depending on molecular background. We propose a differentiated surveillance protocol, based on tumor risks in the various molecular subgroups causing BWS. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Saskia M Maas
- Department of Pediatrics, Academic Medical Center, Amsterdam, The Netherlands
- Department of Clinical Genetics, Academic Medical Center, Amsterdam, The Netherlands
| | - Fleur Vansenne
- Department of Clinical Genetics, University Medical Center Groningen, Groningen, The Netherlands
| | - Daniel J M Kadouch
- Department of Plastic Surgery, Academic Medical Center, Amsterdam, The Netherlands
| | - Abdulla Ibrahim
- Department of Medical Genetics, University of Cambridge and NHR Cambridge Biomedical Research Centre, Cambridge, United Kingdom
- Department of Clinical Genetics, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Jet Bliek
- Department of Clinical Genetics, DNA-Diagnostics Laboratory, Academic Medical Center, Amsterdam, The Netherlands
| | - Saskia Hopman
- Department of Genetics, University Medical Center, Utrecht, The Netherlands
| | - Marcel M Mannens
- Department of Clinical Genetics, DNA-Diagnostics Laboratory, Academic Medical Center, Amsterdam, The Netherlands
| | - Johannes H M Merks
- Department of Pediatrics, Academic Medical Center, Amsterdam, The Netherlands
| | - Eamonn R Maher
- Department of Medical Genetics, University of Cambridge and NHR Cambridge Biomedical Research Centre, Cambridge, United Kingdom
| | - Raoul C Hennekam
- Department of Pediatrics, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
258
|
Rios P, Bailey HD, Orsi L, Lacour B, Valteau-Couanet D, Levy D, Corradini N, Leverger G, Defachelles AS, Gambart M, Sirvent N, Thebaud E, Ducassou S, Clavel J. Risk of neuroblastoma, birth-related characteristics, congenital malformations and perinatal exposures: A pooled analysis of the ESCALE and ESTELLE French studies (SFCE). Int J Cancer 2016; 139:1936-48. [DOI: 10.1002/ijc.30239] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 06/07/2016] [Indexed: 01/11/2023]
Affiliation(s)
- Paula Rios
- Inserm U1153, Epidemiology and Biostatistics Sorbonne Paris Cité Centre (CRESS), Epidemiology of Childhood and Adolescent Cancers Team (EPICEA); Villejuif France
- Paris-Descartes University, UMRS-1153, Epidemiology and Biostatistics Sorbonne Paris Cité Centre (CRESS); Paris France
| | - Helen D Bailey
- Inserm U1153, Epidemiology and Biostatistics Sorbonne Paris Cité Centre (CRESS), Epidemiology of Childhood and Adolescent Cancers Team (EPICEA); Villejuif France
- Paris-Descartes University, UMRS-1153, Epidemiology and Biostatistics Sorbonne Paris Cité Centre (CRESS); Paris France
| | - Laurent Orsi
- Inserm U1153, Epidemiology and Biostatistics Sorbonne Paris Cité Centre (CRESS), Epidemiology of Childhood and Adolescent Cancers Team (EPICEA); Villejuif France
- Paris-Descartes University, UMRS-1153, Epidemiology and Biostatistics Sorbonne Paris Cité Centre (CRESS); Paris France
| | - Brigitte Lacour
- Inserm U1153, Epidemiology and Biostatistics Sorbonne Paris Cité Centre (CRESS), Epidemiology of Childhood and Adolescent Cancers Team (EPICEA); Villejuif France
- Paris-Descartes University, UMRS-1153, Epidemiology and Biostatistics Sorbonne Paris Cité Centre (CRESS); Paris France
- CHU Nancy, Registre National Des Tumeurs Solides De L'Enfant; Vandœuvre-lès-Nancy France
| | | | - Dominique Levy
- Institut Curie, Département D'Oncologie Pédiatrique, Adolescents Et Jeunes Adultes; Paris France
| | - Nadège Corradini
- Centre Léon Bérard; Institut D'Hématologie Et D'Oncologie Pédiatrique; Lyon France
| | - Guy Leverger
- Hôpital Armand-Trousseau; Assistance Publique - Hôpitaux De Paris, Service D'Oncohématologie Pédiatrique; Paris France
| | | | - Marion Gambart
- Hôpital Des Enfants, Sce D'Hématologie Oncologie Pédiatrique; Toulouse France
| | - Nicolas Sirvent
- Service d'Hémato-Oncologie Pédiatrique; Hôpital Arnaud De Villeneuve Montpellier France
| | - Estelle Thebaud
- Hôpital De La Mère Et L'enfant, Service D'Oncologie Pédiatrique; Nantes France
| | - Stéphane Ducassou
- Service D'Oncohématologie Pédiatrique, Hôpital Pellegrin Tripode; Bordeaux France
| | - Jacqueline Clavel
- Inserm U1153, Epidemiology and Biostatistics Sorbonne Paris Cité Centre (CRESS), Epidemiology of Childhood and Adolescent Cancers Team (EPICEA); Villejuif France
- Paris-Descartes University, UMRS-1153, Epidemiology and Biostatistics Sorbonne Paris Cité Centre (CRESS); Paris France
| |
Collapse
|
259
|
Parisi MT, Eslamy H, Park JR, Shulkin BL, Yanik GA. 131I-Metaiodobenzylguanidine Theranostics in Neuroblastoma: Historical Perspectives; Practical Applications. Semin Nucl Med 2016; 46:184-202. [DOI: 10.1053/j.semnuclmed.2016.02.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
260
|
Proust-Houdemont S, Pasqualini C, Blanchard P, Dufour C, Benhamou E, Goma G, Semeraro M, Raquin MA, Hartmann O, Valteau-Couanet D. Busulfan-melphalan in high-risk neuroblastoma: the 30-year experience of a single institution. Bone Marrow Transplant 2016; 51:1076-81. [PMID: 27042850 DOI: 10.1038/bmt.2016.75] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 01/10/2016] [Accepted: 02/19/2016] [Indexed: 11/09/2022]
Abstract
High-dose chemotherapy (HDC) was investigated in high-risk neuroblastoma (HR-NBL) to reduce the risk of relapse. We report the results of the 30-year experience of a cohort of patients with HR-NBL treated with high-dose (HD) busulfan (Bu)-containing regimens. From 1980 to 2009, 215 patients aged >1 year with stage 4 NBL were treated with HD Bu-containing regimens at Gustave Roussy. These data were prospectively recorded in the Pediatric Transplantation Database. The median age at diagnosis was 40 months (12-218 months). All patients had a stage 4 neuroblastoma. NMYC amplification was displayed in 24% of the tumors. The hematopoietic support consisted of bone marrow or PBSCs in 46% and 49% of patients, respectively. The 5-year event-free survival and overall survival rates of the whole cohort were 35.1% and 40%, respectively. Age at diagnosis, bone marrow involvement and tumor response after induction chemotherapy were significant prognostic factors. Toxicity was manageable and decreased over time, owing to both PBSC administration and better supportive care. Based on this experience, HD Bu-melphalan (Mel) has been implemented in Europe and compared with Carboplatin-Etoposide-Mel in the European SIOP Neuroblastoma (SIOPEN)/HR-NBL randomized protocol. It has now become the standard HDC in the SIOPEN HR strategy.
Collapse
Affiliation(s)
- S Proust-Houdemont
- Department of Pediatric Oncology, Centre Hospitalier Universitaire, Angers, France
| | - C Pasqualini
- Department of Pediatric and Adolescent Oncology, Gustave Roussy, Villejuif, France
| | - P Blanchard
- Biostatistics and Epidemiology Unit, Gustave Roussy, Villejuif, France
| | - C Dufour
- Department of Pediatric and Adolescent Oncology, Gustave Roussy, Villejuif, France
| | - E Benhamou
- Biostatistics and Epidemiology Unit, Gustave Roussy, Villejuif, France
| | - G Goma
- Biostatistics and Epidemiology Unit, Gustave Roussy, Villejuif, France
| | - M Semeraro
- Department of Pediatric and Adolescent Oncology, Gustave Roussy, Villejuif, France
| | - M-A Raquin
- Department of Pediatric and Adolescent Oncology, Gustave Roussy, Villejuif, France
| | - O Hartmann
- Department of Pediatric and Adolescent Oncology, Gustave Roussy, Villejuif, France
| | - D Valteau-Couanet
- Department of Pediatric and Adolescent Oncology, Gustave Roussy, Villejuif, France
| |
Collapse
|
261
|
Yáñez Y, Hervás D, Grau E, Oltra S, Pérez G, Palanca S, Bermúdez M, Márquez C, Cañete A, Castel V. TH and DCX mRNAs in peripheral blood and bone marrow predict outcome in metastatic neuroblastoma patients. J Cancer Res Clin Oncol 2016; 142:573-80. [PMID: 26498952 DOI: 10.1007/s00432-015-2054-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 10/09/2015] [Indexed: 12/26/2022]
Abstract
PURPOSE In metastatic neuroblastoma (NB) patients, accurate risk stratification and disease monitoring would reduce relapse probabilities. This study aims to evaluate the independent prognostic significance of detecting tyrosine hydroxylase (TH) and doublecortin (DCX) mRNAs by reverse transcriptase quantitative polymerase chain reaction (RT-qPCR) in peripheral blood (PB) and bone marrow (BM) samples from metastatic NB patients. PROCEDURES RT-qPCR was performed on PB and BM samples from metastatic NB patients at diagnosis, post-induction therapy and at the end of treatment for TH and DCX mRNAs detection. RESULTS High levels of TH and DCX mRNAs when detected in PB and BM at diagnosis independently predicted worse outcome in a cohort of 162 metastatic NB. In the subgroup of high-risk metastatic NB, TH mRNA detected in PB remained as independent predictor of EFS and OS at diagnosis. After the induction therapy, high levels of TH mRNA in PB and DCX mRNA in BM independently predicted poor EFS and OS. Furthermore TH mRNA when detected in BM predicted worse EFS. TH mRNA in PB samples at the end of treatment is an independent predictor of worse outcome. CONCLUSION TH and DCX mRNAs levels in PB and BM assessed by RT-qPCR should be considered in new pre-treatment risk stratification strategies to reliable estimate outcome differences in metastatic NB patients. In those high-risk metastatic NB, TH and DCX mRNA quantification could be used for the assessment of response to treatment and for early detection of progressive disease or relapses.
Collapse
Affiliation(s)
- Yania Yáñez
- Unidad de Oncología Pediátrica, Hospital La Fe, Avda. Fernando Abril Martorell, 106, 46026, Valencia, Spain.
| | - David Hervás
- Unidad de Bioestadística, Hospital La Fe, Valencia, Spain
| | - Elena Grau
- Unidad de Oncología Pediátrica, Hospital La Fe, Avda. Fernando Abril Martorell, 106, 46026, Valencia, Spain
| | - Silvestre Oltra
- Unidad de Genética y Diagnóstico Prenatal, Hospital La Fe, Valencia, Spain
| | - Gema Pérez
- Laboratorio de Biología Molecular, Hospital La Fe, Valencia, Spain
| | - Sarai Palanca
- Laboratorio de Biología Molecular, Hospital La Fe, Valencia, Spain
| | - Mar Bermúdez
- Servicio de Oncología Pediátrica, Hospital Virgen de la Arrixaca, Murcia, Spain
| | - Catalina Márquez
- Servicio de Oncología Pediátrica, Hospital Virgen del Rocio, Seville, Spain
| | - Adela Cañete
- Unidad de Oncología Pediátrica, Hospital La Fe, Avda. Fernando Abril Martorell, 106, 46026, Valencia, Spain
| | - Victoria Castel
- Unidad de Oncología Pediátrica, Hospital La Fe, Avda. Fernando Abril Martorell, 106, 46026, Valencia, Spain
| |
Collapse
|
262
|
Ploessl C, Pan A, Maples KT, Lowe DK. Dinutuximab: An Anti-GD2 Monoclonal Antibody for High-Risk Neuroblastoma. Ann Pharmacother 2016; 50:416-22. [PMID: 26917818 DOI: 10.1177/1060028016632013] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE To review the pharmacology, pharmacokinetics, efficacy, safety, dosage and administration, and formulary considerations for dinutuximab. DATA SOURCES MEDLINE was searched (1964 to January 2016) using the terms ch14.18, dinutuximab, immunotherapy, and neuroblastoma. Other information was identified from package insert, Biologics License Application, abstracts, news releases, and ClinicalTrials.gov. STUDY SELECTION AND DATA EXTRACTION Identified English-language articles were reviewed. Selected studies included phase I through III. DATA SYNTHESIS High-risk neuroblastoma is primarily a childhood cancer with 5-year survival rates of 40% to 50%. Treatment for high-risk neuroblastoma includes induction chemotherapy, surgery, myeloablative chemotherapy with autologous hematopoietic stem cell transplant, and radiation therapy. For patients achieving clinical remission, limited treatments exist for preventing relapse. Dinutuximab is a chimeric, human-murine, anti-GD2 monoclonal antibody approved in combination with granulocyte-macrophage colony-stimulating factor (GM-CSF), aldesleukin (interleukin-2 [IL-2]), and isotretinoin (13-cis-retinoic acid [RA]) for maintenance treatment of pediatric patients with high-risk neuroblastoma who achieve at least a partial response to first-line multiagent, multimodality therapy. In phase III trials, dinutuximab increased 2-year event-free survival and overall survival when compared to standard treatment. Severe adverse effects of dinutuximab include pain, hypersensitivity reactions, capillary leak syndrome, and hypotension. CONCLUSIONS Dinutuximab is the first anti-GD2 monoclonal antibody approved in combination with GM-CSF, IL-2, and RA for maintenance treatment of pediatric patients with high-risk neuroblastoma who achieve at least a partial response to first-line multiagent, multimodality therapy. Ongoing research will determine if dinutuximab could be used earlier in treatment, in nonresponders to initial therapies, in combination with chemotherapy, or in other cancers.
Collapse
Affiliation(s)
- Cady Ploessl
- Virginia Commonwealth University Health System/Medical College of Virginia Hospitals, Richmond, VA, USA
| | - Alice Pan
- Virginia Commonwealth University Health System/Medical College of Virginia Hospitals, Richmond, VA, USA
| | - Kathryn T Maples
- Virginia Commonwealth University Health System/Medical College of Virginia Hospitals, Richmond, VA, USA
| | - Denise K Lowe
- Virginia Commonwealth University Health System/Medical College of Virginia Hospitals, Richmond, VA, USA
| |
Collapse
|
263
|
Zhang R, Zou Y, Zhu J, Zeng X, Yang T, Wang F, He J, Xia H. The Association between GWAS-identified BARD1 Gene SNPs and Neuroblastoma Susceptibility in a Southern Chinese Population. Int J Med Sci 2016; 13:133-138. [PMID: 26941572 PMCID: PMC4764780 DOI: 10.7150/ijms.13426] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 01/05/2016] [Indexed: 02/07/2023] Open
Abstract
A previous genome-wide association study (GWAS) has found that some common variations in the BARD1 gene were associated with neuroblastoma susceptibility especially for high-risk subjects, and the associations have been validated in Caucasians and African-Americans. However, the associations between BARD1 gene polymorphisms and neuroblastoma susceptibility have not been studied among Asians, not to mention Chinese subjects. In the present study, we investigated the association of three BARD1 polymorphisms (rs7585356 G>A, rs6435862 T>G and rs3768716 A>G) with neuroblastoma susceptibility in 201 neuroblastoma patients and 531 controls using TaqMan methodology. Overall, none of these polymorphisms was significantly associated with neuroblastoma susceptibility. However, stratified analysis showed a more profound association between neuroblastoma risk and rs6435862 TG/GG variant genotypes among older children (adjusted OR=1.55, 95% CI=1.04-2.31), and children with adrenal gland-originated disease (adjusted OR=2.94, 95% CI=1.40-6.18), or with ISSN clinical stages III+IV disease (adjusted OR=1.75, 95% CI=1.09-2.84). Similar results were observed for the variant genotypes of rs3768716 A>G polymorphism among these three subgroups. Our results suggest that the BARD1 rs6435862 T>G and rs3768716 A>G polymorphisms may contribute to increased susceptibility to neuroblastoma, especially for the subjects at age ≥12 months, with adrenal gland-originated or with late clinical stage neuroblastoma. These findings need further validation by prospective studies with larger sample size with subjects enrolled from multicenter, involving different ethnicities.
Collapse
Affiliation(s)
- Ruizhong Zhang
- 1. Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Yan Zou
- 1. Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Jinhong Zhu
- 2. Molecular Epidemiology Laboratory and Department of Laboratory Medicine, Harbin Medical University Cancer Hospital, Harbin 150040, Heilongjiang, China
| | - Xinhao Zeng
- 1. Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Tianyou Yang
- 1. Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Fenghua Wang
- 1. Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Jing He
- 1. Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Huimin Xia
- 1. Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| |
Collapse
|
264
|
Loizidou MA, Hadjisavvas A, Tanteles GA, Spanou-Aristidou E, Kyriacou K, Christophidou-Anastasiadou V. Fanconi anemia-D1 due to homozygosity for the BRCA2 gene Cypriot founder mutation: A case report. Oncol Lett 2016; 11:471-473. [PMID: 26834852 DOI: 10.3892/ol.2015.3852] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 09/04/2015] [Indexed: 11/05/2022] Open
Abstract
Fanconi anemia (FA) is a rare disorder characterized by multiple congenital malformations, progressive bone marrow failure and susceptibility to malignancies. Biallelic mutations in the breast cancer 2, early onset (BRCA2) gene are responsible for the FA-D1 subgroup, which accounts for ~3% of all the FA cases. Patients with biallelic BRCA2 mutations generally display a more severe phenotype, with earlier onset and increased incidence of leukaemia and other solid tumors, than other patients with FA. In the present report, the first Cypriot patient with FA-D1 is described, which is the fifth case of a homozygote for the same null allele reported thus far, and the third known case of neuroblastoma in association with FA-D1.
Collapse
Affiliation(s)
- Maria A Loizidou
- Department of Electron Microscopy/Molecular Pathology, Cyprus Institute of Neurology and Genetics, Nicosia 1683, Cyprus
| | - Andreas Hadjisavvas
- Department of Electron Microscopy/Molecular Pathology, Cyprus Institute of Neurology and Genetics, Nicosia 1683, Cyprus; Cyprus School of Molecular Medicine, Cyprus Institute of Neurology and Genetics, Nicosia 1683, Cyprus
| | - George A Tanteles
- Department of Clinical Genetics, Makarios Medical Centre and Cyprus Institute of Neurology and Genetics, Nicosia 1683, Cyprus
| | - Elena Spanou-Aristidou
- Department of Clinical Genetics, Makarios Medical Centre and Cyprus Institute of Neurology and Genetics, Nicosia 1683, Cyprus
| | - Kyriacos Kyriacou
- Department of Electron Microscopy/Molecular Pathology, Cyprus Institute of Neurology and Genetics, Nicosia 1683, Cyprus; Cyprus School of Molecular Medicine, Cyprus Institute of Neurology and Genetics, Nicosia 1683, Cyprus
| | - Violetta Christophidou-Anastasiadou
- Cyprus School of Molecular Medicine, Cyprus Institute of Neurology and Genetics, Nicosia 1683, Cyprus; Department of Clinical Genetics, Makarios Medical Centre and Cyprus Institute of Neurology and Genetics, Nicosia 1683, Cyprus
| |
Collapse
|
265
|
Nakamichi N, Ishimoto T, Yamauchi Y, Masuo Y, Kato Y. Screening to Identify Multidrug Resistance-Associated Protein Inhibitors with Neuroblastoma-Selective Cytotoxicity. Biol Pharm Bull 2016; 39:1638-1645. [DOI: 10.1248/bpb.b16-00319] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Noritaka Nakamichi
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University
| | - Takahiro Ishimoto
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University
| | - Yoshihide Yamauchi
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University
| | - Yusuke Masuo
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University
| | - Yukio Kato
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University
| |
Collapse
|
266
|
Klimovich GV, Ramos-Irizarry CT, Falk GA, Loux T. Development of abdominal compartment syndrome secondary to tumor lysis in an infant with disseminated stage 4 neuroblastoma despite decompressive laparotomy. JOURNAL OF PEDIATRIC SURGERY CASE REPORTS 2016. [DOI: 10.1016/j.epsc.2015.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
267
|
Thompson D, Vo KT, London WB, Fischer M, Ambros PF, Nakagawara A, Brodeur GM, Matthay KK, DuBois SG. Identification of patient subgroups with markedly disparate rates of MYCN amplification in neuroblastoma: A report from the International Neuroblastoma Risk Group project. Cancer 2015; 122:935-45. [PMID: 26709890 DOI: 10.1002/cncr.29848] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 10/17/2015] [Accepted: 11/17/2015] [Indexed: 11/07/2022]
Abstract
BACKGROUND MYCN gene amplification (MNA) is a hallmark of aggressive neuroblastoma. This study was performed to determine univariate and multivariate predictors of tumor MNA. METHODS Data from the International Neuroblastoma Risk Group were analyzed for a subset of 7102 patients with known MYCN status. Chi-square testing and logistic regression were used to identify univariate and multivariate predictors of MYCN status. Recursive partitioning was used to identify groups of patients with maximal differences in rates of MNA. RESULTS All clinical features (age ≥ 18 months, high ferritin levels, high lactate dehydrogenase [LDH] levels, International Neuroblastoma Staging System stage 4, and adrenal sites) and pathological/biological features (DNA index ≤ 1, high mitosis-karyorrhexis index [MKI], undifferentiated/poorly differentiated grade, unfavorable histology according to the International Neuroblastoma Pathology Classification, and segmental chromosomal aberrations [SCAs]) were significantly associated with MNA. LDH (odds ratio [OR], 8.4; P < .001) and chromosomal 1p loss of heterozygosity (OR, 19.8; P < .001) were the clinical and biological variables, respectively, most strongly associated with MNA. In logistic regression, all variables except chromosome 17q aberration and pooled SCAs were independently predictive of MNA. Recursive partitioning identified subgroups with disparate rates of MNA, including subgroups with 85.7% MNA (patients with high LDH levels who had poorly differentiated adrenal tumors with chromosome 1p deletion) and 0.6% MNA (localized tumors having hyperdiploidy and low MKIs and lacking chromosome 1p aberrations). CONCLUSIONS MNA is strongly associated with other clinical and biological variables in neuroblastoma. Recursive partitioning has identified subgroups of neuroblastoma patients with highly disparate rates of MNA. These findings can be used to inform investigations of molecular mechanisms of MNA.
Collapse
Affiliation(s)
- Daria Thompson
- Department of Pediatrics, Benioff Children's Hospital, University of California San Francisco School of Medicine, San Francisco, California
| | - Kieuhoa T Vo
- Department of Pediatrics, Benioff Children's Hospital, University of California San Francisco School of Medicine, San Francisco, California
| | - Wendy B London
- Dana-Farber Children's Hospital Cancer and Blood Disorders Center, Harvard Medical School, Boston, Massachusetts
| | - Matthias Fischer
- Department of Pediatric Oncology, Children's Hospital and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Peter F Ambros
- Children's Cancer Research Institute, St. Anne Kinderkrebsforschung, Vienna, Austria
| | - Akira Nakagawara
- Department of Biochemistry, Chiba Cancer Center Research Institute and Chiba University, Chiba, Japan
| | - Garrett M Brodeur
- Department of Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Katherine K Matthay
- Department of Pediatrics, Benioff Children's Hospital, University of California San Francisco School of Medicine, San Francisco, California
| | - Steven G DuBois
- Department of Pediatrics, Benioff Children's Hospital, University of California San Francisco School of Medicine, San Francisco, California
| |
Collapse
|
268
|
Friedman GK, Beierle EA, Gillespie GY, Markert JM, Waters AM, Chen CY, Denton NL, Haworth KB, Hutzen B, Leddon JL, Streby KA, Wang PY, Cripe TP. Pediatric cancer gone viral. Part II: potential clinical application of oncolytic herpes simplex virus-1 in children. MOLECULAR THERAPY-ONCOLYTICS 2015; 2:S2372-7705(16)30018-3. [PMID: 26436134 PMCID: PMC4589754 DOI: 10.1038/mto.2015.16] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Oncolytic engineered herpes simplex viruses (HSVs) possess many biologic and functional attributes that support their use in clinical trials in children with solid tumors. Tumor cells, in an effort to escape regulatory mechanisms that would impair their growth and progression, have removed many mechanisms that would have protected them from virus infection and eventual virus-mediated destruction. Viruses engineered to exploit this weakness, like mutant HSV, can be safely employed as tumor cell killers, since normal cells retain these antiviral strategies. Many preclinical studies and early phase trials in adults demonstrated that oncolytic HSV can be safely used and are highly effective in killing tumor cells that comprise pediatric malignancies, without generating the toxic side effects of nondiscriminatory chemotherapy or radiation therapy. A variety of engineered viruses have been developed and tested in numerous preclinical models of pediatric cancers and initial trials in patients are underway. In Part II of this review series, we examine the preclinical evidence to support the further advancement of oncolytic HSV in the pediatric population. We discuss clinical advances made to date in this emerging era of oncolytic virotherapy.
Collapse
Affiliation(s)
- Gregory K Friedman
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Elizabeth A Beierle
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | - James M Markert
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Alicia M Waters
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Chun-Yu Chen
- Division of Hematology/Oncology/Blood and Marrow Transplantation, Cancer and Blood Diseases, Nationwide Children's Hospital, The Ohio State University, Columbus, Ohio, USA ; Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, The Ohio State University, Columbus, Ohio, USA
| | - Nicholas L Denton
- Division of Hematology/Oncology/Blood and Marrow Transplantation, Cancer and Blood Diseases, Nationwide Children's Hospital, The Ohio State University, Columbus, Ohio, USA
| | - Kellie B Haworth
- Division of Hematology/Oncology/Blood and Marrow Transplantation, Cancer and Blood Diseases, Nationwide Children's Hospital, The Ohio State University, Columbus, Ohio, USA ; Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, The Ohio State University, Columbus, Ohio, USA
| | - Brian Hutzen
- Division of Hematology/Oncology/Blood and Marrow Transplantation, Cancer and Blood Diseases, Nationwide Children's Hospital, The Ohio State University, Columbus, Ohio, USA
| | - Jennifer L Leddon
- Division of Hematology/Oncology/Blood and Marrow Transplantation, Cancer and Blood Diseases, Nationwide Children's Hospital, The Ohio State University, Columbus, Ohio, USA
| | - Keri A Streby
- Division of Hematology/Oncology/Blood and Marrow Transplantation, Cancer and Blood Diseases, Nationwide Children's Hospital, The Ohio State University, Columbus, Ohio, USA ; Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, The Ohio State University, Columbus, Ohio, USA
| | - Pin-Yi Wang
- Division of Hematology/Oncology/Blood and Marrow Transplantation, Cancer and Blood Diseases, Nationwide Children's Hospital, The Ohio State University, Columbus, Ohio, USA
| | - Timothy P Cripe
- Division of Hematology/Oncology/Blood and Marrow Transplantation, Cancer and Blood Diseases, Nationwide Children's Hospital, The Ohio State University, Columbus, Ohio, USA ; Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
269
|
Tulla M, Berthold F, Graf N, Rutkowski S, von Schweinitz D, Spix C, Kaatsch P. Incidence, Trends, and Survival of Children With Embryonal Tumors. Pediatrics 2015; 136:e623-32. [PMID: 26304823 DOI: 10.1542/peds.2015-0224] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Central nervous system (CNS) and non-CNS embryonal tumors occur principally in children and are rarely seen in adults. The incidence rates for rare entities such as atypical teratoid/rhabdoid tumors (AT/RT) or primitive neuroectodermal tumors in the CNS are rarely published. Incidence rates for certain subgroups, such as hepatoblastomas, have been increasing in some countries. METHODS Data of 8337 embryonal tumors, registered in children (0-14 years) between 1991 and 2012 (for AT/RT 2000-2012) in the population-based German Childhood Cancer Registry with complete national coverage were analyzed for incidence rates, time trends, and survival. RESULTS For most entities, the incidence rates were the highest for children <1 year. An important exception was medulloblastomas, which occurred mainly in 1- to 9-year-olds. Neuroblastomas and ganglioneuroblastomas as well as Wilms tumors (nephroblastomas) had the highest age standardized incidence rates (13.7 and 9.4 per million, respectively). A statistically significant increasing trend for hepatoblastomas (annual average percent change 4.6%) was detected. The survival probabilities varied between the diagnostic groups: primitive neuroectodermal tumors and AT/RT had the lowest and retinoblastomas the highest. The survival was dependent on the age at diagnosis, the most extreme examples being neuroblastomas, for which the survival probability declined steeply for children ≥1 year and medulloblastomas, for which the highest survival was seen for 10- to 14-year-olds. CONCLUSIONS This study presents a comprehensive overview of pediatric embryonal tumors from a well-established, complete nationwide cancer registry. Significant increasing trend for hepatoblastomas was detected for the first time in Europe.
Collapse
Affiliation(s)
- M Tulla
- German Childhood Cancer Registry, Institute for Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany;
| | - F Berthold
- Children's Hospital, Department of Pediatric Oncology and Hematology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - N Graf
- Department of Pediatric Hematology and Oncology, University of Saarland, Homburg/Saar, Germany
| | - S Rutkowski
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; and
| | - D von Schweinitz
- Department of Pediatric Surgery, University of Munich, Munich, Germany
| | - C Spix
- German Childhood Cancer Registry, Institute for Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - P Kaatsch
- German Childhood Cancer Registry, Institute for Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
270
|
Revealed: The spy who regulates neuroblastoma stem cells. Oncotarget 2015; 5:11014-6. [PMID: 25483101 PMCID: PMC4294329 DOI: 10.18632/oncotarget.2839] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Accepted: 12/01/2014] [Indexed: 01/24/2023] Open
|
271
|
Shao JB, Lu ZH, Huang WY, Lv ZB, Jiang H. A single center clinical analysis of children with neuroblastoma. Oncol Lett 2015; 10:2311-2318. [PMID: 26622841 DOI: 10.3892/ol.2015.3588] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Accepted: 06/16/2015] [Indexed: 12/14/2022] Open
Abstract
In the present study, the cases of 59 children diagnosed with neuroblastoma (NB) were retrospectively analyzed to assess the association between the short-term efficacy of treatment and prognostic factors. In total, 59 patients with NB that were diagnosed between July 1, 2008 and June 30, 2013 at Shanghai Children's Hospital were enrolled in the present study. The follow-up was performed until December 31, 2013, and the data revealed that 43 patients (72.9%) achieved complete remission (CR) or partial remission (PR). The 3-year overall survival (OS) rate of patients with stage I, II, III, IV and IVs disease was 100, 100, 65.6, 34.8 and 85.7%, respectively (P=0.02). The 3-year OS and event-free survival rates were evidently increased in patients with favorable histology compared with the rates in the patients with unfavorable histology (P=0.046 and 0.030, respectively). Univariate statistical analysis revealed that the factors significantly associated with prognosis were patient age, tumor stage and risk group (P=0.004, 0.02 and 0.001, respectively). The present study identified that tumor stage, risk group and patient age are important prognostic factors for NB. An age of 18 months was also hypothesized to be the cut-off for the prognosis of patients.
Collapse
Affiliation(s)
- Jing-Bo Shao
- Department of Hematology/Oncology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai 200040, P.R. China
| | - Zheng-Hua Lu
- Department of Hematology/Oncology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai 200040, P.R. China
| | - Wen-Yan Huang
- Department of Nephrology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai 200040, P.R. China
| | - Zhi-Bao Lv
- Department of General Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai 200040, P.R. China
| | - Hui Jiang
- Department of Hematology/Oncology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai 200040, P.R. China
| |
Collapse
|
272
|
|
273
|
Stewart E, Federico S, Karlstrom A, Shelat A, Sablauer A, Pappo A, Dyer MA. The Childhood Solid Tumor Network: A new resource for the developmental biology and oncology research communities. Dev Biol 2015; 411:287-293. [PMID: 26068307 DOI: 10.1016/j.ydbio.2015.03.001] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Significant advances have been made over the past 25 years in our understanding of the most common adult solid tumors such as breast, colon, lung and prostate cancer. Much less is known about childhood solid tumors because they are rare and because they originate in developing organs during fetal development, childhood and adolescence. It can be very difficult to study the cellular origins of pediatric solid tumors in developing organs characterized by rapid proliferative expansion, growth factor signaling, developmental angiogenesis, programmed cell death, tissue reorganization and cell migration. Not only has the etiology of pediatric cancer remained elusive because of their developmental origins, but it also makes it more difficult to treat. Molecular targeted therapeutics that alter developmental pathway signaling may have devastating effects on normal organ development. Therefore, basic research focused on the mechanisms of development provides an essential foundation for pediatric solid tumor translational research. In this article, we describe new resources available for the developmental biology and oncology research communities. In a companion paper, we present the detailed characterization of an orthotopic xenograft of a pediatric solid tumor derived from sympathoadrenal lineage during development.
Collapse
Affiliation(s)
- Elizabeth Stewart
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Sara Federico
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Asa Karlstrom
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Anang Shelat
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Andras Sablauer
- Department of Radiological Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Alberto Pappo
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Michael A Dyer
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
274
|
Al-Tonbary Y, Badr M, Mansour A, El Safy U, Saeed S, Hassan T, Elashery R, Nofal R, Darwish A. Clinico-epidemiology of neuroblastoma in north east Egypt: A 5-year multicenter study. Oncol Lett 2015; 10:1054-1062. [PMID: 26622625 DOI: 10.3892/ol.2015.3335] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 02/22/2015] [Indexed: 12/12/2022] Open
Abstract
Neuroblastoma, an embryonal malignancy of the sympathetic nervous system, is the most frequent extracranial solid tumor The clinico-epidemiological features of neuroblastoma in infants and children were investigated between January 2005 and January 2010 at the Pediatric Oncology units of Mansoura, Zagazig, and Tanta University Children's Hospitals (Egypt). Of 142 cases of neuroblastoma, 10 were omitted from the study due to defective data. The median age of the patients was 30 months, with 75.8% aged ≥1 year and 24.2% aged <1 year at time of diagnosis. The male-to-female ratio was 1.06. Suprarenal glands were the most common primary tumor site (72.7%). The majority of the patients (76.7%) had stage IV disease. Favorable pathology was observed in 43.8% of patients, while 56.2% exhibited unfavorable pathology. The estimated survival rate of patients was 30.7±10.0%, and mean survival time was 24.2±5.2 months. The rate of mortality was 28.6% for patients aged <1 year, and 81.8% for those aged ≥1 year (P=0.005). For patients with favorable pathology, the rate of mortality was significantly lower (28.6%) compared with that of patients with unfavorable pathology (77.8%; P=0.049). Although the association between outcome and each of the primary tumor sites, children's oncology group risk and gender was statistically insignificant, a large effect size was identified between outcome and primary tumor site, as well as children's oncology group risk and a medium effect size was identified between outcome and gender. Additionally, an age of ≥1 year was associated with unfavorable pathology (P=0.024), stage IV disease (P=0.026) and a suprarenal primary tumor site (P=0.001).
Collapse
Affiliation(s)
- Youssef Al-Tonbary
- Department of Pediatric Hematology and Oncology, Mansoura University Children's Hospital, Mansoura, Dakahlia 35516, Egypt
| | - Mohamed Badr
- Department of Pediatric Hematology and Oncology, Zagazig University Children's Hospital, Zagazig, Sharqia 44111, Egypt
| | - Ahmed Mansour
- Department of Pediatric Hematology and Oncology, Mansoura University Children's Hospital, Mansoura, Dakahlia 35516, Egypt
| | - Usama El Safy
- Department of Pediatric Hematology and Oncology, Zagazig University Children's Hospital, Zagazig, Sharqia 44111, Egypt
| | - Shebl Saeed
- Department of Pediatric Hematology and Oncology, Tanta University Children's Hospital, Tanta, Gharbia 31111, Egypt
| | - Tamer Hassan
- Department of Pediatric Hematology and Oncology, Zagazig University Children's Hospital, Zagazig, Sharqia 44111, Egypt
| | - Rasha Elashery
- Department of Pediatric Hematology and Oncology, Mansoura University Children's Hospital, Mansoura, Dakahlia 35516, Egypt
| | - Rofida Nofal
- Department of Pediatric Hematology and Oncology, Zagazig University Children's Hospital, Zagazig, Sharqia 44111, Egypt
| | - Ahmad Darwish
- Department of Pediatric Hematology and Oncology, Mansoura University Children's Hospital, Mansoura, Dakahlia 35516, Egypt
| |
Collapse
|
275
|
Kushner BH, Ostrovnaya I, Cheung IY, Kuk D, Kramer K, Modak S, Yataghene K, Cheung NK. Prolonged progression-free survival after consolidating second or later remissions of neuroblastoma with Anti-G D2 immunotherapy and isotretinoin: a prospective Phase II study. Oncoimmunology 2015; 4:e1016704. [PMID: 26140243 DOI: 10.1080/2162402x.2015.1016704] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 01/30/2015] [Accepted: 01/30/2015] [Indexed: 10/23/2022] Open
Abstract
Relapse of high-risk neuroblastoma (HR-NB) is deemed invariably fatal yet increasing numbers of HR-NB patients achieve a second complete/very good partial remission (CR/VGPR), hence the urgency to find a successful consolidative therapy. Identifying efficacy in patients without assessable disease, however, is problematic. We report the first study providing outcome data for this group of patients with poor prognosis. To prevent another relapse, HR-NB patients in second or later CR/VGPR received the anti-GD2 murine antibody 3F8 plus granulocyte-macrophage colony-stimulating factor plus isotretinoin in a Phase II trial. Upon meeting the target aim for progression-free survival (PFS) in the initial cohort of 33 patients, the trial was amended to allow patients who developed human anti-mouse antibody (HAMA) to receive rituximab to ablate HAMA with or without low-dose maintenance chemotherapy until immunotherapy could resume. For the total of 101 study patients, 5-year PFS and overall survival (OS) rates were 33% ± 5% and 48% ± 5%, respectively. Among the 33 long-term progression-free survivors, 19 had MYCN amplification, 19 had previously received anti-GD2 immunotherapy plus isotretinoin (as first-line therapy), and 15 never received maintenance chemotherapy. In a multivariate analysis of prognostic factors, only absence of minimal residual disease in bone marrow after 2 cycles of immunotherapy and before initiation of isotretinoin or anti-HAMA therapy was significantly favorable for both PFS and OS. Therefore, long-term PFS is possible for HR-NB patients who achieve at least a second CR/VGPR and receive consolidation that includes anti-GD2 immunotherapy plus isotretinoin, even if the patients received these biological treatments before relapse. Results from this prospective study will aid in the development of future Phase II studies for this growing ultra high-risk patient population.
Collapse
Key Words
- ASCT, autologous stem-cell transplantation
- BM, bone marrow
- CNS, central nervous system
- CR, complete remission
- GM-CSF, granulocyte-macrophage colony-stimulating factor
- HAMA, human anti-mouse antibody
- HR-NB: high-risk neuroblastoma
- INRC, International Neuroblastoma Response Criteria
- INRG, International Neuroblastoma Risk Group
- MIBG, metaiodobenzylguanidine
- MRD, minimal residual disease
- OS, overall survival
- PD, progressive disease
- PFS, progression-free survival
- VGPR, very good partial remission
- anti-GD2 antibody
- immunotherapy
- mAb, monoclonal antibody
- minimal residual disease
- salvage
- second remission
Collapse
Affiliation(s)
- Brian H Kushner
- Department of Pediatrics; Memorial Sloan-Kettering Cancer Center ; New York, NY USA
| | - Irina Ostrovnaya
- Department of Epidemiology and Biostatistics; Memorial Sloan Kettering Cancer Center ; New York, NY USA
| | - Irene Y Cheung
- Department of Pediatrics; Memorial Sloan-Kettering Cancer Center ; New York, NY USA
| | - Deborah Kuk
- Department of Epidemiology and Biostatistics; Memorial Sloan Kettering Cancer Center ; New York, NY USA
| | - Kim Kramer
- Department of Pediatrics; Memorial Sloan-Kettering Cancer Center ; New York, NY USA
| | - Shakeel Modak
- Department of Pediatrics; Memorial Sloan-Kettering Cancer Center ; New York, NY USA
| | - Karima Yataghene
- Department of Pediatrics; Memorial Sloan-Kettering Cancer Center ; New York, NY USA
| | - N K Cheung
- Department of Pediatrics; Memorial Sloan-Kettering Cancer Center ; New York, NY USA
| |
Collapse
|
276
|
Lorenz V, Hessenkemper W, Rödiger J, Kyrylenko S, Kraft F, Baniahmad A. Sodium butyrate induces cellular senescence in neuroblastoma and prostate cancer cells. Horm Mol Biol Clin Investig 2015; 7:265-72. [PMID: 25961265 DOI: 10.1515/hmbci.2011.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Accepted: 05/30/2011] [Indexed: 12/31/2022]
Abstract
Cellular senescence leads to an irreversible block of cellular division capacity both in cell culture and in vivo. The induction of an irreversible cell cycle arrest is very useful for treatment of cancer. Histone deacetylases (HDACs) are considered as therapeutic targets to treat cancer patients. HDAC inhibitors repress cancer growth and are used in various clinical trials. Here, we analyzed whether sodium butyrate (NaBu), an inhibitor of class I and II HDACs, induces cellular senescence in neuroblastoma and prostate cancer (PCa) including an androgen-dependent as well as an androgen-independent human PCa cell line. We found that the HDAC inhibitors NaBu and valproic acid (VPA) induce cellular senescence in tumor cells. Interestingly, also an inhibitor of SIRT1, a class HDAC III, induces cellular senescence. Both neuroblastoma and human prostate cancer cell lines express senescence markers, such as the Senescence Associated-β-galactosidase (SA-β-Gal) and Senescence Associated Heterochromatin Foci (SAHF). Furthermore, NaBu down-regulates the proto-oncogenes c-Myc, Cyclin D1 and E2F1 mRNA levels. The mRNA level of the cell cycle inhibitor p16 remains unchanged whereas that of the tumor suppressor p21 is strongly up-regulated. Interestingly, NaBu treatment robustly increases reactive oxygen species (ROS) levels. These results indicate an epigenetic regulation and an association of HDAC inhibition and ROS production with cellular senescence. The data underline that tumor cells can be driven towards cellular senescence by HDAC inhibitors, which may further arise as a potent possibility for tumor suppression.
Collapse
|
277
|
Abstract
OBJECTIVE To determine outcome of neuroblastoma (NBL) in children under 18 mo of age who had been treated with national protocols. METHODS The characteristics and treatment outcomes of 27 children were evaluated retrospectively. RESULTS The event-free survival (EFS) at 60 and 108 mo were 84.7 % ± 7.7 and 72.6 % ± 7.7, respectively. The overall survival (OS) was 91.7 % ± 8 at 108 mo. The only significant risk factor for OS in children with neuroblastoma was the treatment response at the end of therapy (p = 0.001). "Wait and see" policy was applied to two infants with low risk NBL and one infant with stage 4S neuroblastoma and all 3 of these infants have been in remission at last followup. Four of the five patients with MYCN-amplified neuroblastoma were alive at a median follow-up time of 54 mo (range: 5-108 mo). CONCLUSIONS The EFS and OS of the present group were similar to that of the previous series which included children under 18 mo of age with neuroblastoma. Well known prognostic factors did not affect EFS and OS significantly; this may be related to the retrospective design of the present study and the small number of patients reviewed. High survival rate in infants with MYCN-amplified tumors suggests the difference in the biology of infant neuroblastoma.
Collapse
|
278
|
Iodine-131 metaiodobenzylguanidine therapy for neuroblastoma: reports so far and future perspective. ScientificWorldJournal 2015; 2015:189135. [PMID: 25874239 PMCID: PMC4385691 DOI: 10.1155/2015/189135] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 08/01/2014] [Indexed: 12/13/2022] Open
Abstract
Neuroblastoma, which derives from neural crest, is the most common extracranial solid cancer in childhood. The tumors express the norepinephrine (NE) transporters on their cell membrane and take in metaiodobenzylguanidine (MIBG) via a NE transporter. Since iodine-131 (I-131) MIBG therapy was firstly reported, many trails of MIBG therapy in patients with neuroblastoma were performed. Though monotherapy with a low dose of I-131 MIBG could achieve high-probability pain reduction, the objective response was poor. In contrast, more than 12 mCi/kg I-131 MIBG administrations with or without hematopoietic cell transplantation (HCT) obtain relatively good responses in patients with refractory or relapsed neuroblastoma. The combination therapy with I-131 MIBG and other modalities such as nonmyeloablative chemotherapy and myeloablative chemotherapy with HCT improved the therapeutic response in patients with refractory or relapsed neuroblastoma. In addition, I-131 MIBG therapy incorporated in the induction therapy was proved to be feasible in patients with newly diagnosed neuroblastoma. To expand more the use of MIBG therapy for neuroblastoma, further studies will be needed especially in the use at an earlier stage from diagnosis, in the use with other radionuclide formations of MIBG, and in combined use with other therapeutic agents.
Collapse
|
279
|
Yáñez Y, Grau E, Rodríguez-Cortez VC, Hervás D, Vidal E, Noguera R, Hernández M, Segura V, Cañete A, Conesa A, Font de Mora J, Castel V. Two independent epigenetic biomarkers predict survival in neuroblastoma. Clin Epigenetics 2015; 7:16. [PMID: 25767620 PMCID: PMC4357365 DOI: 10.1186/s13148-015-0054-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 02/09/2015] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Neuroblastoma (NB) is the most common extracranial pediatric solid tumor with a highly variable clinical course, ranging from spontaneous regression to life-threatening disease. Survival rates for high-risk NB patients remain disappointingly low despite multimodal treatment. Thus, there is an urgent clinical need for additional biomarkers to improve risk stratification, treatment management, and survival rates in children with aggressive NB. RESULTS Using gene promoter methylation analysis in 48 neuroblastoma tumors with microarray technology, we found a strong association between survival and gene promoter hypermethylation (P = 0.036). Hypermethylation of 70 genes significantly differentiated high-risk survivor patients from those who died during follow-up time. Sixteen genes with relevant roles in cancer biology were further validated in an additional cohort of 83 neuroblastoma tumors by bisulfite pyrosequencing. High promoter methylation rates of these genes were found in patients with metastatic tumors (either stage metastatic (M) or metastatic special (MS)), 18 months or older at first diagnosis, MYCN amplification, relapsed, and dead. Notably, the degree of methylation of retinoblastoma 1 (RB1) and teratocarcinoma-derived growth factor 1 (TDGF1) predicts event-free and overall survival independently of the established risk factors. In addition, low RB1 mRNA expression levels associate with poor prognosis suggesting that promoter methylation could contribute to the transcriptional silencing of this gene in NB. CONCLUSIONS We found a new epigenetic signature predictive for NB patients' outcome: the methylation status of RB1 and TDGF1 associate with poorer survival. This information is useful to assess prognosis and improve treatment selection.
Collapse
Affiliation(s)
- Yania Yáñez
- Pediatric Oncology Unit, Hospital Universitari i Politècnic La Fe, Avda Fernando Abril Martorell, Valencia, 46026 Spain
| | - Elena Grau
- Pediatric Oncology Unit, Hospital Universitari i Politècnic La Fe, Avda Fernando Abril Martorell, Valencia, 46026 Spain
| | - Virginia C Rodríguez-Cortez
- Chromatin and Disease Group, Cancer Epigenetics and Biology Programme (PEBC) Bellvitge Biomedical Research Institute (IDIBELL), Gran Via de L'Hospitalet, Barcelona, 08908 Spain
| | - David Hervás
- Biostatistics Unit, Instituto de Investigación Sanitaria La Fe, Avda Fernando Abril Martorell, Valencia, 46026 Spain
| | - Enrique Vidal
- Genomics of Gene Expression Lab, Centro de Investigaciones Príncipe Felipe, Carrer d'Eduardo Primo Yúfera, Valencia, 46012 Spain
| | - Rosa Noguera
- Department of Pathology, Medical School, University of Valencia, Avda Blasco Ibáñez, Valencia, 46010 Spain
| | - Miguel Hernández
- Department of Pathology, Hospital Universitari i Politècnic La Fe, Avda Fernando Abril Martorell, Valencia, 46026 Spain
| | - Vanessa Segura
- Pediatric Oncology Unit, Hospital Universitari i Politècnic La Fe, Avda Fernando Abril Martorell, Valencia, 46026 Spain
| | - Adela Cañete
- Pediatric Oncology Unit, Hospital Universitari i Politècnic La Fe, Avda Fernando Abril Martorell, Valencia, 46026 Spain
| | - Ana Conesa
- Genomics of Gene Expression Lab, Centro de Investigaciones Príncipe Felipe, Carrer d'Eduardo Primo Yúfera, Valencia, 46012 Spain
| | - Jaime Font de Mora
- Laboratory of Cellular and Molecular Biology, Instituto de Investigación Sanitaria La Fe, Avda Fernando Abril Martorell, Valencia, 46026 Spain
| | - Victoria Castel
- Pediatric Oncology Unit, Hospital Universitari i Politècnic La Fe, Avda Fernando Abril Martorell, Valencia, 46026 Spain
| |
Collapse
|
280
|
Angelini P, Baruchel S, Marrano P, Irwin MS, Thorner PS. The neuroblastoma and ganglion components of nodular ganglioneuroblastoma are genetically similar: evidence against separate clonal origins. Mod Pathol 2015; 28:166-76. [PMID: 25081755 DOI: 10.1038/modpathol.2014.90] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 04/18/2014] [Accepted: 04/19/2014] [Indexed: 12/12/2022]
Abstract
Nodular ganglioneuroblastoma is characterized by a macroscopic nodule of neuroblastoma within a ganglioneuromatous component. These two components have been considered to originate from separate clones, with the neuroblastoma clone accounting for the clinical behavior of nodular ganglioneuroblastoma. In order to investigate the clonal origin of the cellular components (neuroblasts, ganglion cells, and Schwann cells) of nodular ganglioneuroblastoma, paraffin-embedded tumor samples from eight cases were analyzed by single nucleotide polymorphism array and in situ hybridization. DNA was extracted separately from neuroblastomatous and ganglioneuromatous areas. By in situ hybridization, MYCN gain (4-10 gene copies/nucleus) was detected in 7/8 neuroblastoma samples. In ganglioneuromatous regions, gains were also detected in ganglion cells but not in Schwann cells. Single-nucleotide polymorphism array studies identified chromosome losses (11q and 14q) and gains (12, 13q, 17q and 18q) in the neuroblastoma component, whereas the ganglioneuromatous component showed fewer or no genetic alterations. There were no unique copy number changes distinguishing nodular ganglioneuroblastoma from other subtypes of neuroblastoma. By in situ hybridization, ganglion cells but not Schwann cells showed the same alterations detected in neuroblasts. Thus, neuroblasts and ganglion cells in nodular ganglioneuroblastoma are genetically related and may arise from the same clone. In contrast, the Schwann cells have a different origin and may be derived from a non-neoplastic neural crest precursor. Our results suggest that the clinical behavior of nodular ganglioneuroblastoma cannot be explained by the presence of separate clones with distinct genetic signatures.
Collapse
Affiliation(s)
- Paola Angelini
- Division of Haematology/Oncology, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Sylvain Baruchel
- 1] Division of Haematology/Oncology, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada [2] Department of Pediatrics, University of Toronto, University of Toronto, Toronto, ON, Canada
| | - Paula Marrano
- Division of Pathology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Meredith S Irwin
- 1] Division of Haematology/Oncology, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada [2] Department of Pediatrics, University of Toronto, University of Toronto, Toronto, ON, Canada
| | - Paul S Thorner
- 1] Division of Pathology, The Hospital for Sick Children, Toronto, ON, Canada [2] Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
281
|
Abstract
Neuroblastoma (NB) is the third most common pediatric cancer. Although NB accounts for 7% of pediatric malignancies, it is responsible for more than 10% of childhood cancer-related mortality. Prognosis and treatment are determined by clinical and biological risk factors. Estimated 5-year survival rates for patients with non-high-risk and high-risk NB are more than 90% and less than 50%, respectively. Recent clinical trials have continued to reduce therapy for patients with non-high-risk NB, including the most favorable subsets who are often followed with observation approaches. In contrast, high-risk patients are treated aggressively with chemotherapy, radiation, surgery, and myeloablative and immunotherapies.
Collapse
|
282
|
Uslu L, Donig J, Link M, Rosenberg J, Quon A, Daldrup-Link HE. Value of 18F-FDG PET and PET/CT for evaluation of pediatric malignancies. J Nucl Med 2015; 56:274-86. [PMID: 25572088 DOI: 10.2967/jnumed.114.146290] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Successful management of solid tumors in children requires imaging tests for accurate disease detection, characterization, and treatment monitoring. Technologic developments aim toward the creation of integrated imaging approaches that provide a comprehensive diagnosis with a single visit. These integrated diagnostic tests not only are convenient for young patients but also save direct and indirect health-care costs by streamlining procedures, minimizing hospitalizations, and minimizing lost school or work time for children and their parents. (18)F-FDG PET/CT is a highly sensitive and specific imaging modality for whole-body evaluation of pediatric malignancies. However, recent concerns about ionizing radiation exposure have led to a search for alternative imaging methods, such as whole-body MR imaging and PET/MR. As we develop new approaches for tumor staging, it is important to understand current benchmarks. This review article will synthesize the current literature on (18)F-FDG PET/CT for tumor staging in children, summarizing questions that have been solved and providing an outlook on unsolved avenues.
Collapse
Affiliation(s)
- Lebriz Uslu
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, California; and
| | - Jessica Donig
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, California; and
| | - Michael Link
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Jarrett Rosenberg
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, California; and
| | - Andrew Quon
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, California; and
| | - Heike E Daldrup-Link
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, California; and
| |
Collapse
|
283
|
An Introspective Update on the Influence of miRNAs in Breast Carcinoma and Neuroblastoma Chemoresistance. GENETICS RESEARCH INTERNATIONAL 2014; 2014:743050. [PMID: 25548681 PMCID: PMC4273469 DOI: 10.1155/2014/743050] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 10/23/2014] [Accepted: 11/04/2014] [Indexed: 02/08/2023]
Abstract
Chemoresistance to conventional cytotoxic drugs may occur in any type of cancer and this can either be inherent or develop through time. Studies have linked this acquired resistance to the abnormal expression of microRNAs (miRNAs) that normally silence genes. At abnormal levels, miRNAs can either gain ability to silence tumour suppressor genes or else lose ability to silence oncogenes. miRNAs can also affect pathways that are involved in drug metabolism, such as drug efflux pumps, resulting in a resistant phenotype. The scope of this review is to provide an introspective analysis on the specific niches of breast carcinoma and neuroblastoma research.
Collapse
|
284
|
Defferrari R, Mazzocco K, Ambros IM, Ambros PF, Bedwell C, Beiske K, Bénard J, Berbegall AP, Bown N, Combaret V, Couturier J, Erminio G, Gambini C, Garaventa A, Gross N, Haupt R, Kohler J, Jeison M, Lunec J, Marques B, Martinsson T, Noguera R, Parodi S, Schleiermacher G, Tweddle DA, Valent A, Van Roy N, Vicha A, Villamon E, Tonini GP. Influence of segmental chromosome abnormalities on survival in children over the age of 12 months with unresectable localised peripheral neuroblastic tumours without MYCN amplification. Br J Cancer 2014; 112:290-5. [PMID: 25356804 PMCID: PMC4453444 DOI: 10.1038/bjc.2014.557] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 09/22/2014] [Accepted: 10/04/2014] [Indexed: 01/06/2023] Open
Abstract
Background: The prognostic impact of segmental chromosome alterations (SCAs) in children older than 1 year, diagnosed with localised unresectable neuroblastoma (NB) without MYCN amplification enrolled in the European Unresectable Neuroblastoma (EUNB) protocol is still to be clarified, while, for other group of patients, the presence of SCAs is associated with poor prognosis. Methods: To understand the role of SCAs we performed multilocus/pangenomic analysis of 98 tumour samples from patients enrolled in the EUNB protocol. Results: Age at diagnosis was categorised into two groups using 18 months as the age cutoff. Significant difference in the presence of SCAs was seen in tumours of patients between 12 and 18 months and over 18 months of age at diagnosis, respectively (P=0.04). A significant correlation (P=0.03) was observed between number of SCAs per tumour and age. Event-free (EFS) and overall survival (OS) were calculated in both age groups, according to both the presence and number of SCAs. In older patients, a poorer survival was associated with the presence of SCAs (EFS=46% vs 75%, P=0.023; OS=66.8% vs 100%, P=0.003). Moreover, OS of older patients inversely correlated with number of SCAs (P=0.002). Finally, SCAs provided additional prognostic information beyond histoprognosis, as their presence was associated with poorer OS in patients over 18 months with unfavourable International Neuroblastoma Pathology Classification (INPC) histopathology (P=0.018). Conclusions: The presence of SCAs is a negative prognostic marker that impairs outcome of patients over the age of 18 months with localised unresectable NB without MYCN amplification, especially when more than one SCA is present. Moreover, in older patients with unfavourable INPC tumour histoprognosis, the presence of SCAs significantly affects OS.
Collapse
Affiliation(s)
- R Defferrari
- Department of Pathology, Istituto Giannina Gaslini, Genova 16148, Italy
| | - K Mazzocco
- Department of Pathology, Istituto Giannina Gaslini, Genova 16148, Italy
| | - I M Ambros
- Children's Cancer Research Institute, St Anna Kinderkrebsforschung, Vienna 1090, Austria
| | - P F Ambros
- Children's Cancer Research Institute, St Anna Kinderkrebsforschung, Vienna 1090, Austria
| | - C Bedwell
- Northern Genetics Service, Newcastle upon Tyne NEI 3 BZ, UK
| | - K Beiske
- Department of Pathology, Oslo University Hospital Rikshopitalet, Oslo 0424, Norway
| | - J Bénard
- Département de Biologie et de Pathologie Médicales, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - A P Berbegall
- Department of Pathology, Medical School of Valencia, University of Valencia, Valencia 46010, Spain
| | - N Bown
- Northern Genetics Service, Newcastle upon Tyne NEI 3 BZ, UK
| | - V Combaret
- Laboratoire de Recherche Translationnelle, Centre Léon-Bérard, Lyon 69008, France
| | - J Couturier
- Unité de Génétique Somatique et Cytogénétique, Institut Curie, Paris Cedex 05 75248, France
| | - G Erminio
- Epidemiology, Biostatistics and Committees Unit, Istituto Giannina Gaslini, Genova 16148, Italy
| | - C Gambini
- Department of Pathology, Istituto Giannina Gaslini, Genova 16148, Italy
| | - A Garaventa
- Department of Haematology-Oncology, Istituto Giannina Gaslini, Genova 16148, Italy
| | - N Gross
- Pediatric Oncology Research Unit, Lausanne University Hospital (CHUV), Lausanne 1011, Switzerland
| | - R Haupt
- Epidemiology, Biostatistics and Committees Unit, Istituto Giannina Gaslini, Genova 16148, Italy
| | - J Kohler
- Department of Paediatric Oncology, Southampton General Hospital, Southampton S016 6YD, UK
| | - M Jeison
- Cancer Cytogenetique and Molecular Cytogenetique Laboratory, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
| | - J Lunec
- Northern Institute for Cancer Research, Newcastle University, Newcastle NE2 4HH, UK
| | - B Marques
- Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge, Lisbon 1649-016, Portugal
| | - T Martinsson
- Department of Clinical Genetics, Göteborg University, Sahlgrenska University Hospital, Göteborg 413 45, Sweden
| | - R Noguera
- Department of Pathology, Medical School of Valencia, University of Valencia, Valencia 46010, Spain
| | - S Parodi
- Institute of Electronics, Computer and Telecommunication Engineering, National Research Council, Genova 16149, Italy
| | - G Schleiermacher
- 1] INSERM U830, Laboratoire de Génétique et Biologie des Cancers, Paris Cedex 05 75248, France [2] Département d'Oncologie Pédiatrique, Institut Curie, Paris Cedex 05 75248, France
| | - D A Tweddle
- Northern Institute for Cancer Research, Newcastle University, Newcastle NE2 4HH, UK
| | - A Valent
- Département de Biologie et de Pathologie Médicales, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - N Van Roy
- Center for Medical Genetics, Ghent University Hospital, Ghent 9000, Belgium
| | - A Vicha
- Department of Paediatric Haematology and Oncology, Charles University and University Hospital Motol, Prague 15008, Czech Republic
| | - E Villamon
- Department of Hematology, Hospital Universitari i Politècnic La Fe, Valencia 46009, Spain
| | - G P Tonini
- Laboratory of Neuroblastoma, Onco/Haematology Laboratory, University of Padua, Pediatric Research Institute (IRP)-Città della Speranza, Corso Stati Uniti 4, Padova 35127, Italy
| |
Collapse
|
285
|
Meany HJ, London WB, Ambros PF, Matthay KK, Monclair T, Simon T, Garaventa A, Berthold F, Nakagawara A, Cohn SL, Pearson ADJ, Park JR. Significance of clinical and biologic features in Stage 3 neuroblastoma: a report from the International Neuroblastoma Risk Group project. Pediatr Blood Cancer 2014; 61:1932-9. [PMID: 25044743 DOI: 10.1002/pbc.25134] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 05/13/2014] [Indexed: 11/08/2022]
Abstract
BACKGROUND International Neuroblastoma Staging System (INSS) Stage 3 neuroblastoma is a heterogeneous disease. Data from the International Neuroblastoma Risk Group (INRG) database were analyzed to define patient and tumor characteristics predictive of outcome. PROCEDURE Of 8,800 patients in the INRG database, 1,483 with INSS Stage 3 neuroblastoma and complete follow-up data were analyzed. Secondary analysis was performed in 1,013 patients (68%) with MYCN-non-amplified (NA) tumors. Significant prognostic factors were identified via log-rank test comparisons of survival curves. Multivariable Cox proportional hazards regression model was used to identify factors independently predictive of event-free survival (EFS). RESULTS Age at diagnosis (P < 0.0001), tumor MYCN status (P < 0.0001), and poorly differentiating/undifferentiated histology (P = 0.03) were independent predictors of EFS. Compared to other Stage 3 subgroups, outcome was inferior for patients ≥ 547 days with MYCN-NA neuroblastoma (P < 0.0001), and within this cohort, serum ferritin ≥ 96 ng/ml was associated with inferior EFS (P = 0.02). For patients <547 days of age with MYCN-NA tumors, serum ferritin levels were prognostic of overall survival (OS) (P = 0.04) and chromosome 11q aberration was prognostic of EFS (P = 0.03). CONCLUSIONS Among patients with INSS Stage 3 neuroblastoma patients, age at diagnosis, MYCN status and histology predict outcome. Patients <547 days of age with MYCN-NA tumors that lack chromosome 11q aberrations or those with serum ferritin <96 ng/ml have excellent prognosis and should be considered for therapy reduction. Prospective clinical trials are needed to identify optimal therapy for those patients ≥ 547 days of age with undifferentiated histology or elevated serum ferritin.
Collapse
Affiliation(s)
- Holly J Meany
- Department of Hematology/Oncology, Children's National Medical Center, Washington, District of Columbia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
286
|
Darwish MH, Farah RA, Farhat GN, Torbey PHN, Ghandour FA, Bejjani-Doueihy NA, Dhaini HR. Association of CYP3A4/5 genotypes and expression with the survival of patients with neuroblastoma. Mol Med Rep 2014; 11:1462-8. [PMID: 25370902 DOI: 10.3892/mmr.2014.2835] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 07/30/2014] [Indexed: 11/06/2022] Open
Abstract
Neuroblastoma (NB) is a rare pediatric disease in Lebanon for which poor prognosis remains a major challenge. Genetic polymorphism of genes coding for drug‑metabolizing enzymes may influence the response of a patient to chemotherapy. This study investigates a possible association between CYP3A4/5 polymorphism and expression levels and survival in NB patients. All patients with stage III and IV NB diagnosed between 1993 and 2012 in three major hospitals in Beirut were included (n=27). Demographic information and survival time were obtained from medical records. CYP3A4 and CYP3A5 genotypes and expression levels were determined in archival tumors by polymerase chain reaction (PCR) and restriction fragment length polymorphism and quantitative PCR, respectively. Additionally, MYCN amplification was assessed. A Cox proportional hazards model was used to evaluate potential associations, adjusting for MYCN amplification. A statistically significant increase in the risk of mortality was observed in patients with MYCN amplification [hazard ratio (HR) 4.11, 95% confidence interval (CI) 1.14‑14.80]. Patients with CYP3A5 expression levels above the median had a lower risk of mortality (HR 0.61, 95% CI 0.21‑1.74) and patients with CYP3A4 expression levels above the median had a higher risk of mortality (HR 2.00, 95% CI 0.67‑5.90). CYP3A5*3/*3 homozygote mutants had a 4.3‑fold increase in the risk of mortality compared with that of homozygote wild‑type or heterozygote mutants (HR 4.30, 95% CI 0.56‑33.30). Carriers of the CYP3A4*1B mutant allele had a 52% lower risk of mortality compared with that of non‑carriers (HR 0.48, 95% CI 0.06‑3.76). Although the results of the present study did not achieve statistical significance, associations were observed, which indicates that CYP3A4 and CYP3A5 may modulate the clinical outcome of NB. Further studies with larger sample sizes are required to characterize the effects of the polymorphism and expression levels of CYP3A4/5 on the survival of patients with NB.
Collapse
Affiliation(s)
- Mohamad H Darwish
- Department of Medical Lab Sciences, Faculty of Health Sciences, University of Balamand, Beirut 1100‑2807, Lebanon
| | - Roula A Farah
- Department of Pediatric Oncology, Saint George Hospital University Medical Center, Beirut 1100‑2807, Lebanon
| | - Ghada N Farhat
- Department of Medical Lab Sciences, Faculty of Health Sciences, University of Balamand, Beirut 1100‑2807, Lebanon
| | - Paul-Henri N Torbey
- Department of Pediatrics, Hotel Dieu De France Hospital, Beirut 1100‑2190, Lebanon
| | - Fatima A Ghandour
- Department of Pathology, Saint George Hospital University Medical Center, Beirut 1100‑2807, Lebanon
| | - Noha A Bejjani-Doueihy
- Department of Pathology, University Medical Center ‑ Rizk Hospital, Beirut 11-3288, Lebanon
| | - Hassan R Dhaini
- Department of Medical Lab Sciences, Faculty of Health Sciences, University of Balamand, Beirut 1100‑2807, Lebanon
| |
Collapse
|
287
|
Abstract
Neuroblastoma is a developmental tumor of young children arising from the embryonic sympathoadrenal lineage of the neural crest. Neuroblastoma is the primary cause of death from pediatric cancer for children between the ages of one and five years and accounts for ∼13% of all pediatric cancer mortality. Its clinical impact and unique biology have made this aggressive malignancy the focus of a large concerted translational research effort. New insights into tumor biology are driving the development of new classification schemas. Novel targeted therapeutic approaches include small-molecule inhibitors as well as epigenetic, noncoding-RNA, and cell-based immunologic therapies. In this review, recent insights regarding the pathogenesis and biology of neuroblastoma are placed in context with the current understanding of tumor biology and tumor/host interactions. Systematic classification of patients coupled with therapeutic advances point to a future of improved clinical outcomes for this biologically distinct and highly aggressive pediatric malignancy.
Collapse
Affiliation(s)
- Chrystal U Louis
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas 77030; ,
| | | |
Collapse
|
288
|
Adult neuroblastoma complicated by increased intracranial pressure: a case report and review of the literature. Case Rep Oncol Med 2014; 2014:341980. [PMID: 25328733 PMCID: PMC4190830 DOI: 10.1155/2014/341980] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 08/30/2014] [Accepted: 09/03/2014] [Indexed: 11/29/2022] Open
Abstract
Neuroblastoma is the third most commonly occurring malignancy of the pediatric population, although it is extremely rare in the adult population. In adults, neuroblastoma is often metastatic and portends an extremely poor overall survival. Our case report documents metastatic neuroblastoma occurring in a healthy 29-year-old woman whose course was complicated by an unusual presentation of elevated intracranial pressures. The patient was treated with systemic chemotherapy, I131 metaiodobenzylguanidine (MIBG) radiotherapy, and autologous stem cell transplant (SCT). Unfortunately the patient's response to therapy was limited and she subsequently died. We aim to review neuroblastoma in the context of increased intracranial pressure and the limited data of neuroblastoma occurring in the adult population, along with proposed treatment options.
Collapse
|
289
|
Oberthuer A, Juraeva D, Hero B, Volland R, Sterz C, Schmidt R, Faldum A, Kahlert Y, Engesser A, Asgharzadeh S, Seeger R, Ohira M, Nakagawara A, Scaruffi P, Tonini GP, Janoueix-Lerosey I, Delattre O, Schleiermacher G, Vandesompele J, Speleman F, Noguera R, Piqueras M, Bénard J, Valent A, Avigad S, Yaniv I, Grundy RG, Ortmann M, Shao C, Schwab M, Eils R, Simon T, Theissen J, Berthold F, Westermann F, Brors B, Fischer M. Revised risk estimation and treatment stratification of low- and intermediate-risk neuroblastoma patients by integrating clinical and molecular prognostic markers. Clin Cancer Res 2014; 21:1904-15. [PMID: 25231397 DOI: 10.1158/1078-0432.ccr-14-0817] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 08/05/2014] [Indexed: 11/16/2022]
Abstract
PURPOSE To optimize neuroblastoma treatment stratification, we aimed at developing a novel risk estimation system by integrating gene expression-based classification and established prognostic markers. EXPERIMENTAL DESIGN Gene expression profiles were generated from 709 neuroblastoma specimens using customized 4 × 44 K microarrays. Classification models were built using 75 tumors with contrasting courses of disease. Validation was performed in an independent test set (n = 634) by Kaplan-Meier estimates and Cox regression analyses. RESULTS The best-performing classifier predicted patient outcome with an accuracy of 0.95 (sensitivity, 0.93; specificity, 0.97) in the validation cohort. The highest potential clinical value of this predictor was observed for current low-risk patients [5-year event-free survival (EFS), 0.84 ± 0.02 vs. 0.29 ± 0.10; 5-year overall survival (OS), 0.99 ± 0.01 vs. 0.76 ± 0.11; both P < 0.001] and intermediate-risk patients (5-year EFS, 0.88 ± 0.06 vs. 0.41 ± 0.10; 5-year OS, 1.0 vs. 0.70 ± 0.09; both P < 0.001). In multivariate Cox regression models for low-risk/intermediate-risk patients, the classifier outperformed risk assessment of the current German trial NB2004 [EFS: hazard ratio (HR), 5.07; 95% confidence interval (CI), 3.20-8.02; OS: HR, 25.54; 95% CI, 8.40-77.66; both P < 0.001]. On the basis of these findings, we propose to integrate the classifier into a revised risk stratification system for low-risk/intermediate-risk patients. According to this system, we identified novel subgroups with poor outcome (5-year EFS, 0.19 ± 0.08; 5-year OS, 0.59 ± 0.1), for whom we propose intensified treatment, and with beneficial outcome (5-year EFS, 0.87 ± 0.05; 5-year OS, 1.0), who may benefit from treatment de-escalation. CONCLUSIONS Combination of gene expression-based classification and established prognostic markers improves risk estimation of patients with low-risk/intermediate-risk neuroblastoma. We propose to implement our revised treatment stratification system in a prospective clinical trial.
Collapse
Affiliation(s)
- André Oberthuer
- Children's Hospital, Department of Pediatric Oncology and Hematology, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Dilafruz Juraeva
- Department of Theoretical Bioinformatics (B080), German Cancer Research Center, Heidelberg, Germany
| | - Barbara Hero
- Children's Hospital, Department of Pediatric Oncology and Hematology, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Ruth Volland
- Children's Hospital, Department of Pediatric Oncology and Hematology, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Carolina Sterz
- Children's Hospital, Department of Pediatric Oncology and Hematology, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Rene Schmidt
- Institute of Biostatistics and Clinical Research, University of Muenster, Muenster, Germany
| | - Andreas Faldum
- Institute of Biostatistics and Clinical Research, University of Muenster, Muenster, Germany
| | - Yvonne Kahlert
- Children's Hospital, Department of Pediatric Oncology and Hematology, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Anne Engesser
- Children's Hospital, Department of Pediatric Oncology and Hematology, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Shahab Asgharzadeh
- Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, Los Angeles, California
| | - Robert Seeger
- Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, Los Angeles, California
| | - Miki Ohira
- Laboratory of Cancer Genomics, Chiba Cancer Center Research Institute, Chuoh-ku, Chiba, Japan
| | - Akira Nakagawara
- Division of Biochemistry and Innovative Cancer Therapeutics, Chiba Cancer Center Research Institute, Chuoh-ku, Chiba, Japan
| | - Paola Scaruffi
- Center of Physiopathology of Human Reproduction, Department of Obstetrics and Gynecology, IRCCS San Martino Hospital, National Cancer Research Institute (IST), Genoa, Italy
| | - Gian Paolo Tonini
- Laboratory of Neuroblastoma, Onco/Hematology Laboratory Department SDB University of Padua, Pediatric Research Institute, Padua, Italy
| | | | | | | | - Jo Vandesompele
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Frank Speleman
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Rosa Noguera
- Department of Pathology, University of Valencia, Valencia, Spain
| | - Marta Piqueras
- Department of Pathology, University of Valencia, Valencia, Spain
| | - Jean Bénard
- Department of Tumor Genetics, Institut Gustave Roussy, Villejuif, France
| | - Alexander Valent
- Department of Tumor Genetics, Institut Gustave Roussy, Villejuif, France
| | - Smadar Avigad
- Schneider Children's Medical Center of Israel, Pediatric Hematology Oncology, Petah Tikva, Israel. Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Isaac Yaniv
- Schneider Children's Medical Center of Israel, Pediatric Hematology Oncology, Petah Tikva, Israel. Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Richard G Grundy
- Children's Cancer Leukaemia Group, University of Leicester, Leicester, United Kingdom
| | - Monika Ortmann
- Department of Pathology, University of Cologne, Cologne, Germany
| | - Chunxuan Shao
- Department of Neuroblastoma Genomics (B087), German Cancer Research Center, Heidelberg, Germany
| | - Manfred Schwab
- Department of Neuroblastoma Genomics (B087), German Cancer Research Center, Heidelberg, Germany
| | - Roland Eils
- Department of Theoretical Bioinformatics (B080), German Cancer Research Center, Heidelberg, Germany. Department for Bioinformatics and Functional Genomics, Institute for Pharmacy and Molecular Biotechnology (IPMB) and BioQuant, Heidelberg University, Heidelberg, Germany
| | - Thorsten Simon
- Children's Hospital, Department of Pediatric Oncology and Hematology, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Jessica Theissen
- Children's Hospital, Department of Pediatric Oncology and Hematology, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Frank Berthold
- Children's Hospital, Department of Pediatric Oncology and Hematology, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Frank Westermann
- Department of Neuroblastoma Genomics (B087), German Cancer Research Center, Heidelberg, Germany
| | - Benedikt Brors
- Department of Theoretical Bioinformatics (B080), German Cancer Research Center, Heidelberg, Germany
| | - Matthias Fischer
- Children's Hospital, Department of Pediatric Oncology and Hematology, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
| |
Collapse
|
290
|
Brickler MM, Basel DG, Gheorghe G, Margolis DM, Kelly ME, Ehrhardt MJ. Early therapy-related myeloid sarcoma and deletion of 9q22.32 to q31.1. Pediatr Blood Cancer 2014; 61:1701-3. [PMID: 24668947 DOI: 10.1002/pbc.25040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Accepted: 03/05/2014] [Indexed: 11/07/2022]
Abstract
Survival following childhood neuroblastoma is improving with low rates of secondary myeloid neoplasms. We describe a 13-month-old male with intermediate risk neuroblastoma who developed an isolated scalp therapy-related myeloid sarcoma (t-MS). Developmental delays and two distinct malignancies prompted constitutional evaluation. Chromosomal microarray identified a 7.3 Mb deletion of 9q22.32 to 9q31.1. He remains in remission 11 months following hematopoietic cell transplant. Unusual presentations of rare diseases necessitate a multidisciplinary approach and adaptation of standardized protocols to accommodate increased risks imposed by genetic variants.
Collapse
Affiliation(s)
- Molly M Brickler
- Pediatric Hematology, Oncology, and Bone Marrow Transplant, Medical College of Wisconsin, Milwaukee, Wisconsin; Midwest Center for Cancer and Blood Disorders, Milwaukee, Wisconsin
| | | | | | | | | | | |
Collapse
|
291
|
Abstract
Congenital anomalies of the upper urinary tract are common and frequently diagnosed on prenatal ultrasound. In the absence of infection, these anomalies are often asymptomatic. This article reviews key features and long-term implications to assist in discussions with families. In contrast, a perinatal renal tumor is rare but extremely alarming. This update on the most common tumors and their treatment is useful in reassuring parents that most infants, after primary surgical resection, are cured without adjuvant therapies. To understand renal agenesis and other congenital renal malformations and their associated anomalies, a brief review of normal renal development is presented.
Collapse
Affiliation(s)
- Ellen Shapiro
- Department of Urology, New York University School of Medicine, 150 32nd Street, 2nd Floor, New York, NY 10017, USA.
| |
Collapse
|
292
|
Schleiermacher G, Janoueix-Lerosey I, Delattre O. Recent insights into the biology of neuroblastoma. Int J Cancer 2014; 135:2249-61. [PMID: 25124476 DOI: 10.1002/ijc.29077] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 05/08/2014] [Indexed: 01/24/2023]
Abstract
Neuroblastoma (NB) is an embryonal tumor of the sympathetic nervous system which accounts for 8-10% of pediatric cancers. It is characterized by a broad spectrum of clinical behaviors from spontaneous regression to fatal outcome despite aggressive therapies. Considerable progress has been made recently in the germline and somatic genetic characterization of patients and tumors. Indeed, predisposition genes that account for a significant proportion of familial and syndromic cases have been identified and genome-wide association studies have retrieved a number of susceptibility loci. In addition, genome-wide sequencing, copy-number and expression studies have been conducted on tumors and have detected important gene modifications, profiles and signatures that have strong implications for the therapeutic stratification of patients. The identification of major players in NB oncogenesis, including MYCN, ALK, PHOX2B and LIN28B, has enabled the development of new animal models. Our review focuses on these recent advances, on the insights they provide on the mechanisms involved in NB development and their applications for the clinical management of patients.
Collapse
Affiliation(s)
- Gudrun Schleiermacher
- Equipe SIRIC Recherche Translationnelle en Oncologie Pédiatrique, Département de Recherche Translationnelle et Inserm U830, Centre de Recherche, Paris Cedex, 05, France; Département de pédiatrie, Institut Curie, Paris Cedex, 05, France; Unité Génétique et Biologie des Cancers, Inserm U830, Centre de Recherche, Institut Curie, Paris Cedex, 05, France
| | | | | |
Collapse
|
293
|
Theissen J, Oberthuer A, Hombach A, Volland R, Hertwig F, Fischer M, Spitz R, Zapatka M, Brors B, Ortmann M, Simon T, Hero B, Berthold F. Chromosome 17/17q gain and unaltered profiles in high resolution array-CGH are prognostically informative in neuroblastoma. Genes Chromosomes Cancer 2014; 53:639-49. [PMID: 24737690 DOI: 10.1002/gcc.22174] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 03/31/2014] [Indexed: 12/22/2022] Open
Abstract
The prognostic relevance of chromosome 17 gain in neuroblastoma is still discussed. This investigation specifies the frequency, type, size, and transcriptional relevance in a large patient cohort. Primary tumor material of 202 patients was analyzed using high-resolution oligonucleotide array-based comparative genomic hybridization (aCGH) and correlated with clinical and survival data. A subset (n = 145) was correlated for differentially expressed genes (DEG) by microarray analysis. Chromosome 17 aCGH analysis showed numerical gain in 94/202 patients (47%), partial gain in 93/202 patients (46%), and no gain in 15/202 patients (7%). The frequency of partial gain was higher in stage 4 neuroblastoma (stage 1 15%; stage 2 12%; stage 3 16%; stage 4S 7%; and stage 4 50%). Overall survival (OS) was superior in patients with numerical gain compared with patients with partial gain or no gain (5-y-OS: 0.95 ± 0.02 vs. 0.63 ± 0.05 vs. 0.60 ± 0.13; P < 0.001). Gene expression analysis demonstrated 95/130 DEGs between tumors with numerical or partial chromosome/no gain. Only one DEG (CCKBR) was detected comparing tumors with partial gain and those with no gain. In patients with partial gain, the distribution of breakpoints did not correlate with stage and 11q status, but with MYCN amplification and 1p status. The "best" breakpoints in cases with partial 17q gain were at 42.5 Mb for event-free and 26.6 Mb for OS. Numerical gain of chromosome 17 is associated with a better prognosis than partial and no gain. The group of tumors with partial gain was similar to the group without gain with respect to stage distribution, outcome, and gene expression profile.
Collapse
Affiliation(s)
- Jessica Theissen
- Department of Pediatric Oncology and Hematology, Children's Hospital, University of Cologne, Cologne, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
294
|
Mossé YP, Deyell RJ, Berthold F, Nagakawara A, Ambros PF, Monclair T, Cohn SL, Pearson AD, London WB, Matthay KK. Neuroblastoma in older children, adolescents and young adults: a report from the International Neuroblastoma Risk Group project. Pediatr Blood Cancer 2014; 61:627-35. [PMID: 24038992 DOI: 10.1002/pbc.24777] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 08/21/2013] [Indexed: 11/11/2022]
Abstract
BACKGROUND Neuroblastoma in older children and adolescents has a distinctive, indolent phenotype, but little is known about the clinical and biological characteristics that distinguish this rare subgroup. Our goal was to determine if an optimal age cut-off exists that defines indolent disease and if accepted prognostic factors and treatment approaches are applicable to older children. PROCEDURE Using data from the International Neuroblastoma Risk Group, among patients ≥18 months old (n = 4,027), monthly age cut-offs were tested to determine the effect of age on survival. The prognostic effect of baseline characteristics and autologous hematopoietic cell transplant (AHCT) for advanced disease was assessed within two age cohorts; ≥5 to <10 years (n = 730) and ≥10 years (n = 200). RESULTS Older age was prognostic of poor survival, with outcome gradually worsening with increasing age at diagnosis, without statistical evidence for an optimal age cut-off beyond 18 months. Among patients ≥5 years, factors significantly prognostic of lower event-free survival (EFS) and overall survival (OS) in multivariable analyses were INSS stage 4, MYCN amplification and unfavorable INPC histology classification. Among stage 4 patients, AHCT provided a significant EFS and OS benefit. Following relapse, patients in both older cohorts had prolonged OS compared to those ≥18 months to <5 years (P < 0.0001). CONCLUSIONS Despite indolent disease and infrequent MYCN amplification, older children with advanced disease have poor survival, without evidence for a specific age cut-off. Our data suggest that AHCT may provide a survival benefit in older children with advanced disease. Novel therapeutic approaches are required to more effectively treat these patients.
Collapse
Affiliation(s)
- Yaël P Mossé
- Division of Oncology, Children's Hospital of Philadelphia and Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | | | | | | | | | | | | | | |
Collapse
|
295
|
Viprey VF, Gregory WM, Corrias MV, Tchirkov A, Swerts K, Vicha A, Dallorso S, Brock P, Luksch R, Valteau-Couanet D, Papadakis V, Laureys G, Pearson AD, Ladenstein R, Burchill SA. Neuroblastoma mRNAs predict outcome in children with stage 4 neuroblastoma: a European HR-NBL1/SIOPEN study. J Clin Oncol 2014; 32:1074-83. [PMID: 24590653 DOI: 10.1200/jco.2013.53.3604] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2024] Open
Abstract
PURPOSE To evaluate the hypothesis that detection of neuroblastoma mRNAs by reverse transcriptase quantitative polymerase chain reaction (RTqPCR) in peripheral blood (PB) and bone marrow aspirates (BM) from children with stage 4 neuroblastoma are clinically useful biomarkers of risk. METHODS RTqPCR for paired-like homeobox 2b (PHOX2B), tyrosine hydroxylase (TH), and doublecortin (DCX) mRNA in PB and BM of children enrolled onto the High-Risk Neuroblastoma Trial-1 of the European Society of Pediatric Oncology Neuroblastoma Group (HR-NBL1/SIOPEN) was performed at diagnosis and after induction therapy. RESULTS High levels of TH, PHOX2B, or DCX mRNA in PB or BM at diagnosis strongly predicted for worse event-free survival (EFS) and overall survival (OS) in a cohort of 290 children. After induction therapy, high levels of these mRNAs predicted worse EFS and OS in BM but not in PB. Combinations of mRNAs in BM did not add to the predictive power of any single mRNA. However, in the original (n = 182) and validation (n = 137) PB cohorts, high TH (log10TH > 0.8) or high PHOX2B (log10PHOX2B > 0.28) identify 19% of children as ultrahigh risk, with 5-year EFS and OS rates of 0%; OS rate was 25% (95% CI, 16% to 36%) and EFS rate was 38% (95% CI, 28% to 49%) in the remaining children. The magnitude of reduction in mRNA level between diagnosis and postinduction therapy in BM or PB was not of additional predictive value. CONCLUSION High levels of TH and PHOX2B mRNA in PB at diagnosis objectively identify children with ultrahigh-risk disease who may benefit from novel treatment approaches. The level of TH, PHOX2B, and DCX mRNA in BM and/or PB at diagnosis might contribute to an algorithm to improve stratification of children for treatment.
Collapse
Affiliation(s)
- Virginie F Viprey
- Virginie F. Viprey and Susan A. Burchill, Leeds Institute of Cancer and Pathology; Walter M. Gregory, Clinical Trials Research Unit, University of Leeds, Leeds; Penelope Brock, Great Ormond Street Hospital, London; Andrew D. Pearson, Institute of Cancer Research/Royal Marsden National Health Service Foundation Trust, Sutton, United Kingdom; Maria V. Corrias and Sandro Dallorso, Gaslini Institute, Genoa; Roberto Luksch, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milano, Italy; Andrei Tchirkov, Centre Hospitalier Universitaire Clermont-Ferrand and Clermont Université, Université d'Auvergne, Clermont-Ferrand; Dominique Valteau-Couanet, Institut Gustave Roussy, Villejuif, France; Katrien Swerts and Genevieve Laureys, University Hospital Ghent, Ghent, Belgium; Ales Vicha, Charles University and University Hospital Motol, Prague, Czech Republic; Vassilios Papadakis, Agia Sofia Children's Hospital, Athens, Greece; and Ruth Ladenstein, Children's Cancer Research Institute/St Anna Children's Hospital, Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
296
|
Morgenstern DA, London WB, Stephens D, Volchenboum SL, Hero B, Di Cataldo A, Nakagawara A, Shimada H, Ambros PF, Matthay KK, Cohn SL, Pearson ADJ, Irwin MS. Metastatic neuroblastoma confined to distant lymph nodes (stage 4N) predicts outcome in patients with stage 4 disease: A study from the International Neuroblastoma Risk Group Database. J Clin Oncol 2014; 32:1228-35. [PMID: 24663047 DOI: 10.1200/jco.2013.53.6342] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
PURPOSE The presence of distant metastases is one of the most powerful predictors of outcome in patients with neuroblastoma. However, the pattern of metastatic spread is not incorporated into current risk stratification systems. Small case series have suggested that patients with neuroblastoma who have metastatic disease limited to distant lymph nodes (4N disease) may have improved outcomes. PATIENTS AND METHODS We analyzed retrospective data from the International Neuroblastoma Risk Group database for patients diagnosed from 1990 to 2002. 4N patients were compared with the remaining stage 4 patients (non-4N), excluding those with missing metastatic site data. RESULTS In all, 2,250 International Neuroblastoma Staging System stage 4 patients with complete data were identified, of whom 146 (6.5%) had 4N disease. For 4N patients, event-free survival (EFS; 5-year, 77% ± 4%) and overall survival (OS; 5-year, 85% ± 3%) were significantly better than EFS (5-year, 35% ± 1%) and OS (5-year, 42% ± 1%) for non-4N stage 4 patients (P < .001). 4N patients were more likely to be younger (P < .001) and have tumors with favorable characteristics, including absence of MYCN amplification (89% v 69%; P < .001). In a multivariable analysis, 4N disease remained a significant predictor of outcome (hazard ratio for non-4N v 4N: 3.40 for EFS and 3.69 for OS). Within subgroups defined by age at diagnosis and tumor MYCN status, 4N disease was significantly associated with improved outcomes. CONCLUSION 4N represents a subgroup with better outcome than that of other patients with metastatic disease. These findings suggest that the biology and treatment response of 4N tumors differ from other stage 4 tumors, and less intensive therapy should be considered for this cohort. Future exploration of biologic factors determining the pattern of metastatic spread is warranted.
Collapse
Affiliation(s)
- Daniel A Morgenstern
- Daniel A. Morgenstern and Meredith S. Irwin, Hospital for Sick Children and University of Toronto; Derek Stephens, Hospital for Sick Children, Toronto, ON, Canada; Daniel A. Morgenstern, Great Ormond Street Hospital for Children National Health Service (NHS) Foundation Trust, London; Andrew D.J. Pearson, The Royal Marsden NHS Foundation Trust, Sutton, Surrey, United Kingdom; Wendy B. London, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Samuel L. Volchenboum and Susan L. Cohn, University of Chicago, Chicago, IL; Barbara Hero, University Children's Hospital, Köln, Germany; Andrea Di Cataldo, University of Catania, Catania, Italy; Akira Nakagawara, Chiba University School of Medicine, Chiba, Japan; Hiroyuki Shimada, University of Southern California at Los Angeles, Los Angeles; Katherine K. Matthay, University of California at San Francisco, San Francisco, CA; and Peter F. Ambros, St. Anna Kinderkrebsforschung, Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
297
|
Jin C, Yu D, Čančer M, Nilsson B, Leja J, Essand M. Tat-PTD-modified oncolytic adenovirus driven by the SCG3 promoter and ASH1 enhancer for neuroblastoma therapy. Hum Gene Ther 2014; 24:766-75. [PMID: 23889332 DOI: 10.1089/hum.2012.132] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Secretogranin III (SGC3) belongs to the granin family and is highly expressed in endocrine and neural tissues. The human SCG3 promoter has not yet been characterized. We identified that a 0.5-kb DNA fragment upstream of the SCG3 gene can selectively drive transgene expression in neuroblastoma cell lines. The strength of transgene expression was further increased, with specificity maintained, by addition of the human achaete-scute complex homolog 1 (ASH1) enhancer. We developed an oncolytic serotype 5-based adenovirus, in which the SCG3 promoter and ASH1 enhancer drive E1A gene expression. The virus was further modified with a cell-penetrating peptide (Tat-PTD) in the viral capsid, which we have previously shown results in increased adenovirus transduction efficiency of many neuroblastoma cell lines. The virus, Ad5PTD(ASH1-SCG3-E1A), shows selective and efficient killing of neuroblastoma cell lines in vitro, including cisplatin-, etoposide-, and doxorubicin-insensitive neuroblastoma cells. Furthermore, it delays tumor growth and thereby prolonged survival for nude mice harboring subcutaneous human neuroblastoma xenograft. In conclusion, we report a novel oncolytic adenovirus with potential use for neuroblastoma therapy.
Collapse
Affiliation(s)
- Chuan Jin
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Uppsala University, Uppsala SE-75185, Sweden
| | | | | | | | | | | |
Collapse
|
298
|
Wnt inhibitory factor-1 functions as a tumor suppressor through modulating Wnt/β-catenin signaling in neuroblastoma. Cancer Lett 2014; 348:12-9. [PMID: 24561119 DOI: 10.1016/j.canlet.2014.02.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 01/22/2014] [Accepted: 02/10/2014] [Indexed: 02/05/2023]
Abstract
Neuroblastoma is the most common extracranial solid tumor in childhood and is associated with serious morbidity and mortality. The effective treatment of neuroblastoma remains one of the major challenges in pediatric oncology. The Wnt signaling pathway has been shown to play a significant role in the pathogenesis of adult and pediatric tumors. WIF-1 has been identified as an important Wnt antagonist which inhibits Wnt/β-catenin signaling by directly binding to Wnt proteins. However, the expression and function of WIF-1 in neuroblastoma remains unknown. The present study showed that WIF-1 was downregulated with high level promoter methylation in neuroblastoma cells, and was significantly upregulated after exposure to demethylating agent. This finding suggests that downregulation of WIF-1 was associated with its promoter methylation in neuroblastoma. To further study the potential function of WIF-1 in neuroblastoma, we constructed a plasmid that over-expressed WIF-1 and transfected the plasmid into one neuroblastoma cell line SK-N-SH. We found that restoration of WIF-1 inhibited the growth and proliferation of neuroblastoma cells in vitro. Moreover, Wnt/β-catenin signaling activity and target genes expression were reduced by WIF-1 restoration. These results provide support that WIF-1 is downregulated and functions as a tumor suppressor by antagonizing Wnt/β-catenin signaling in neuroblastoma, suggesting a potential role as a therapeutic target in neuroblastoma.
Collapse
|
299
|
High incidence of MYCN amplification in a Moroccan series of neuroblastic tumors: comparison to current biological data. ACTA ACUST UNITED AC 2014; 22:112-8. [PMID: 23628823 DOI: 10.1097/pdm.0b013e318277448e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
MYCN protooncogene status was assessed for the first time in Morocco in peripheral neuroblastic tumors, including neuroblastoma, ganglioneuroblastoma, and ganglioneuroma. Correlations with age at diagnosis, stage, mitosis-karyorrhexis index, differentiation, and Shimada histology were evaluated. Thirty-six formalin-fixed, paraffin-embedded peripheral neuroblastic tumor tissue specimens collected between 2007 and 2010 from the Pathology Department were assessed for MYCN amplification using fluorescence in situ hybridization. MYCN amplification was found in 27.8% of cases. An association of MYCN amplification with unfavorable Shimada grading, higher mitosis-karyorrhexis index, and undifferentiated morphologic phenotype was found. We found no correlation with older age, advanced stage, or the presence of metastasis. Our results suggested that the presence of MYCN amplification is a strong biological indicator of a poor outcome and aggressive disease in neuroblastoma and nodular ganglioneuroblastoma.
Collapse
|
300
|
Molecular oncology of neuroblastoma. Mol Oncol 2013. [DOI: 10.1017/cbo9781139046947.060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|