351
|
Shang X, Vasudevan SA, Yu Y, Ge N, Ludwig AD, Wesson CL, Wang K, Burlingame SM, Zhao YJ, Rao PH, Lu X, Russell HV, Okcu MF, Hicks MJ, Shohet JM, Donehower LA, Nuchtern JG, Yang J. Dual-specificity phosphatase 26 is a novel p53 phosphatase and inhibits p53 tumor suppressor functions in human neuroblastoma. Oncogene 2010; 29:4938-46. [PMID: 20562916 DOI: 10.1038/onc.2010.244] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Chemoresistance is a major cause of treatment failure and poor outcome in neuroblastoma. In this study, we investigated the expression and function of dual-specificity phosphatase 26 (DUSP26), also known as mitogen-activated protein kinase phophatase-8, in human neuroblastoma. We found that DUSP26 was expressed in a majority of neuroblastoma cell lines and tissue specimens. Importantly, we found that DUSP26 promotes the resistance of human neuroblastoma to doxorubicin-induced apoptosis by acting as a p53 phosphatase to downregulate p53 tumor suppressor function in neuroblastoma cells. Inhibiting DUSP26 expression in the IMR-32 neuroblastoma cell line enhanced doxorubicin-induced p53 phosphorylation at Ser20 and Ser37, p21, Puma, Bax expression as well as apoptosis. In contrast, DUSP26 overexpression in the SK-N-SH cell line inhibited doxorubicin-induced p53 phosphorylation at Ser20 and Ser37, p21, Puma, Bax expression and apoptosis. Using in vitro and in vivo assays, we found that DUSP26 binds to p53 and dephosphorylates p53 at Ser20 and Ser37. In this report, we show that DUSP26 functions as a p53 phosphatase, which suppresses downstream p53 activity in response to genotoxic stress. This suggests that inhibition of this phosphatase may increase neuroblastoma chemosensitivity and DUSP26 is a novel therapeutic target for this aggressive pediatric malignancy.
Collapse
Affiliation(s)
- X Shang
- Texas Children's Cancer Center, Department of Pediatrics, Dan L Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
352
|
Wolter J, Angelini P, Irwin M. p53 family: Therapeutic targets in neuroblastoma. Future Oncol 2010; 6:429-44. [PMID: 20222799 DOI: 10.2217/fon.09.176] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Survival rates for metastatic neuroblastoma remain poor, despite significant increase in the intensity of therapy. Although it represents approximately 7% of pediatric cancer, neuroblastoma accounts for approximately 15% of childhood cancer deaths. Thus, novel approaches to enhance neuroblastoma chemotherapy sensitivity and prevent or bypass chemoresistance are required. Disruption of the p53 pathway is a common mechanism leading to defects in apoptosis in cancer cells. Increasing evidence suggests that the p53 pathway may be inactivated in neuroblastoma. Inactivation of the p53 pathway occurs most commonly at the time of relapse, and probably contributes to chemoresistance. The p53 family proteins, p73 and p63, can also induce apoptosis, and early studies suggest that p73 may be important in neuroblastoma pathogenesis and response to treatment. This article focuses on current therapies and novel drugs targeting p53 and p73 signaling pathways in neuroblastoma. Understanding the balance between the p53 family proteins in neuroblastoma and how their expression and activity are regulated will hopefully lead to the discovery of agents that target these pathways to induce neuroblastoma cell death, alone or in combination with chemotherapies.
Collapse
Affiliation(s)
- Jennifer Wolter
- Department of Medical Biophysics, University of Toronto, Hospital for Sick Children, ON, Canada
| | | | | |
Collapse
|
353
|
Affiliation(s)
- John M Maris
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, and Abramson Family Cancer Research Institute, Philadelphia, PA 19104-4318, USA.
| |
Collapse
|
354
|
Park JR, Eggert A, Caron H. Neuroblastoma: biology, prognosis, and treatment. Hematol Oncol Clin North Am 2010; 24:65-86. [PMID: 20113896 DOI: 10.1016/j.hoc.2009.11.011] [Citation(s) in RCA: 337] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Neuroblastoma, a neoplasm of the sympathetic nervous system, is the second most common extracranial malignant tumor of childhood and the most common solid tumor of infancy. Neuroblastoma is a heterogeneous malignancy with prognosis ranging from near uniform survival to high risk for fatal demise. Neuroblastoma serves as a paradigm for the prognostic utility of biologic and clinical data and the potential to tailor therapy for patient cohorts at low, intermediate, and high risk for recurrence. This article summarizes our understanding of neuroblastoma biology and prognostic features and discusses their impact on current and proposed risk stratification schemas, risk-based therapeutic approaches, and the development of novel therapies for patients at high risk for failure.
Collapse
Affiliation(s)
- Julie R Park
- Division of Hematology and Oncology, University of Washington School of Medicine and Seattle Children's Hospital, 4800 Sand Point Way NE, Seattle, WA 98105-0371, USA.
| | | | | |
Collapse
|
355
|
Jeison M, Ash S, Halevy-Berko G, Mardoukh J, Luria D, Avigad S, Feinberg-Gorenshtein G, Goshen Y, Hertzel G, Kapelushnik J, Ben Barak A, Attias D, Steinberg R, Stein J, Stark B, Yaniv I. 2p24 Gain region harboring MYCN gene compared with MYCN amplified and nonamplified neuroblastoma: biological and clinical characteristics. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 176:2616-25. [PMID: 20395439 DOI: 10.2353/ajpath.2010.090624] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Although the role of MYCN amplification in neuroblastoma is well established, the biological and clinical characteristics of the 2p gain region harboring the MYCN gene remain unclear. The aim of this study was to compare the biological and clinical characteristics of these tumors with MYCN amplified and nonamplified neuroblastoma and to determine their impact on disease outcome. Samples from 177 patients were analyzed by fluorescence in situ hybridization, including MYCN, 1p, 17q, and 11q regions; 2p gain was identified in 25 patients, MYCN amplification in 31, and no amplification in 121 patients. Patients with 2p gain had a significantly worse 5-year event-free survival rate than patients with no MYCN amplified (P < 0.001), and an intermediate 5-year overall survival rate difference existed between the MYCN amplified tumors (P = 0.025) and nonamplified (P = 0.003) groups. All of the 2p gain samples were associated with segmental and/or numerical alterations in the other tested regions. The presence of segmental alterations with or without MYCN amplification was recently found to be the strongest predictor of relapse in a multivariate analysis. The results of the present study suggest that the determination of MYCN gene copy number relative to chromosome 2, when evaluating MYCN status at diagnosis, may help to reveal the underlying genetic pattern of these tumors and better understand their clinical behavior.
Collapse
Affiliation(s)
- Marta Jeison
- Ca-Cytogenetic Lab, Schneider Children's Medical Center of Israel, Kaplan St. 14, 49202 Petah Tikva, Israel.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
356
|
Harder T, Plagemann A, Harder A. Birth weight and risk of neuroblastoma: a meta-analysis. Int J Epidemiol 2010; 39:746-56. [PMID: 20236985 DOI: 10.1093/ije/dyq040] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Neuroblastoma is the most common solid tumour in infancy but its aetiology is largely unknown. Prenatal factors might play a key role in its pathogenesis. Previous studies investigated whether birth weight is associated with risk of neuroblastoma, with conflictive results. We conducted a meta-analysis to quantitatively summarize the published evidence. METHODS Results from 10 case-control studies and one cohort study (1966 to December 2008) were included, involving a total of 3004 children with neuroblastoma. We constructed random-effects and fixed-effects models, performed 'pool-first' analyses, assessed heterogeneity and publication bias and performed sensitivity and influence analyses. RESULTS High birth weight (>4000 g) was associated with increased risk of neuroblastoma [odds ratio (OR) 1.19; 95% confidence interval (CI) 1.04-1.36]. Results for high birth weight were highly homogenous (I(2) = 0%). Low birth weight (<2500 g) was also related to increased risk of neuroblastoma (OR 1.24; 95% CI 1.0-1.55), but results were more heterogeneous (I(2 )= 30%). No evidence for particularly influential studies or for publication bias was found. However, sensitivity analysis indicated the presence of bias in studies on the association with low birth weight. Above 2500 g each 1000-g increase in birth weight was associated with a 13% (95% CI 3-25) increase in risk of neuroblastoma. CONCLUSIONS This meta-analysis shows that high birth weight is highly reproducibly associated with increased risk of neuroblastoma. The association with low birth weight was found to be less robust and deserves further studies.
Collapse
Affiliation(s)
- Thomas Harder
- Clinic of Obstetrics, Division of Experimental Obstetrics, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | | | | |
Collapse
|
357
|
Banelli B, Bonassi S, Casciano I, Mazzocco K, Di Vinci A, Scaruffi P, Brigati C, Allemanni G, Borzì L, Tonini GP, Romani M. Outcome prediction and risk assessment by quantitative pyrosequencing methylation analysis of the SFN gene in advanced stage, high-risk, neuroblastic tumor patients. Int J Cancer 2010; 126:656-68. [DOI: 10.1002/ijc.24768] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
358
|
|
359
|
A new familial cancer syndrome including predisposition to Wilms tumor and neuroblastoma. Fam Cancer 2010; 9:425-30. [DOI: 10.1007/s10689-009-9319-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
360
|
Scaruffi P, Stigliani S, Moretti S, Coco S, De Vecchi C, Valdora F, Garaventa A, Bonassi S, Tonini GP. Transcribed-Ultra Conserved Region expression is associated with outcome in high-risk neuroblastoma. BMC Cancer 2009; 9:441. [PMID: 20003513 PMCID: PMC2804711 DOI: 10.1186/1471-2407-9-441] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2009] [Accepted: 12/15/2009] [Indexed: 02/08/2023] Open
Abstract
Background Neuroblastoma is the most common, pediatric, extra-cranial, malignant solid tumor. Despite multimodal therapeutic protocols, outcome for children with a high-risk clinical phenotype remains poor, with long-term survival still less than 40%. Hereby, we evaluated the potential of non-coding RNA expression to predict outcome in high-risk, stage 4 neuroblastoma. Methods We analyzed expression of 481 Ultra Conserved Regions (UCRs) by reverse transcription-quantitative real-time PCR and of 723 microRNAs by microarrays in 34 high-risk, stage 4 neuroblastoma patients. Results First, the comparison of 8 short- versus 12 long-term survivors showed that 54 UCRs were significantly (P < 0.0491) over-expressed in the former group. For 48 Ultra Conserved Region (UCRs) the expression levels above the cut-off values defined by ROC curves were strongly associated with good-outcome (OS: 0.0001 <P < 0.0185, EFS: 0.0001 <P < 0.0491). Then we tested the Transcribed-UCR (T-UCR) threshold risk-prediction model on an independent cohort of 14 patients. The expression profile of 28 T-UCRs was significantly associated to prognosis and at least 15 up-regulated T-UCRs are needed to discriminate (P < 0.0001) short- from long-survivors at the highest sensitivity and specificity (94.12%). We also identified a signature of 13 microRNAs differently expressed between long- and short-surviving patients. The comparative analysis of the two classes of non-coding RNAs disclosed that 9 T-UCRs display their expression level that are inversely correlated with expression of 5 complementary microRNAs of the signature, indicating a negative regulation of T-UCRs by direct interaction with microRNAs. Moreover, 4 microRNAs down-regulated in tumors of long-survivors target 3 genes implicated in neuronal differentiation, that are known to be over-expressed in low-risk tumors. Conclusions Our pilot study suggests that a deregulation of the microRNA/T-UCR network may play an important role in the pathogenesis of neuroblastoma. After further validation on a larger independent set of samples, such findings may be applied as the first T-UCR prognostic signature for high-risk neuroblastoma patients.
Collapse
Affiliation(s)
- Paola Scaruffi
- Translational Paediatric Oncology, National Cancer Research Institute (IST), Genoa, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
361
|
|
362
|
|
363
|
|
364
|
Passoni L, Longo L, Collini P, Coluccia AML, Bozzi F, Podda M, Gregorio A, Gambini C, Garaventa A, Pistoia V, Del Grosso F, Tonini GP, Cheng M, Gambacorti-Passerini C, Anichini A, Fossati-Bellani F, Di Nicola M, Luksch R. Mutation-independent anaplastic lymphoma kinase overexpression in poor prognosis neuroblastoma patients. Cancer Res 2009; 69:7338-46. [PMID: 19723661 DOI: 10.1158/0008-5472.can-08-4419] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase predominantly expressed in the developing nervous system. Recently, mutated ALK has been identified as a major oncogene associated with familial and sporadic neuroblastomas (NBL). Yet, a direct correlation between endogenous expression level of the ALK protein, oncogenic potential, and clinical outcome has not been established. We investigated ALK genetic mutations, protein expression/phosphorylation, and functional inhibition both in NBL-derived cell lines and in 34 localized and 48 advanced/metastatic NBL patients. ALK constitutive phosphorylation/activation was observed in high-ALK expressing cells, harboring either a mutated or a wild-type receptor. No activation was found in cell lines with low expression of wild-type ALK. After 72 hours of treatments, small molecule ALK inhibitor CEP-14083 (60 nmol/L) induced growth arrest and cell death in NBL cells overexpressing wild-type (viability: ALK(high) 12.8%, ALK(low) 73%, P = 0.0035; cell death: ALK(high) 56.4%, ALK(low) 16.2%, P = 0.0001) or mutated ALK. ALK protein expression was significantly up-regulated in advanced/metastatic compared with localized NBLs (ALK overexpressing patients: stage 1-2, 23.5%; stage 3-4, 77%; P < 0.0001). Interestingly, protein levels did not always correlate with ALK genetic alterations and/or mRNA abundance. Both mutated and wild-type ALK receptor can exert oncogenic activity in NBL cells. However, wild-type ALK receptor requires a critical threshold of expression to achieve oncogenic activation. Overexpression of either mutated or wild-type ALK defines poor prognosis patients. Alternative mechanisms other than direct mutations and/or gene amplification regulate the ALK level of expression in NBL cells. Wild-type ALK is a potential therapeutic target for advanced/metastatic NBLs.
Collapse
Affiliation(s)
- Lorena Passoni
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
365
|
Vasudevan SA, Shang X, Shang X, Chang S, Ge N, Diaz-Miron JL, Russell HV, Hicks MJ, Ludwig AD, Wesson CL, Burlingame SM, Kim ES, Khan J, Yang J, Nuchtern JG. Neuroblastoma-derived secretory protein is a novel secreted factor overexpressed in neuroblastoma. Mol Cancer Ther 2009; 8:2478-89. [PMID: 19671756 DOI: 10.1158/1535-7163.mct-08-1132] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Secreted proteins such as growth factors, cytokines, and chemokines play important roles in tumor development. Through expression microarray and bioinformatic analysis, we discovered a novel secreted protein, neuroblastoma-derived secretory protein (NDSP). The NDSP gene is found on chromosome 1q25.2 and encodes a 167 amino acid protein with a putative signal peptide. Using real-time PCR and immunoblotting, we find that NDSP is specifically overexpressed in neuroblastoma at much higher levels than other adult and pediatric malignancies and normal tissues. NDSP is an 18-kDa protein that can be secreted by NDSP-transfected HEK-293T cells, as well as, neuroblastoma cell lines endogenously expressing NDSP. Inhibiting NDSP expression in neuroblastoma cell lines with retrovirally transduced NDSP small hairpin interfering RNA, sh-NDSP, results in decreased cellular proliferation and colony formation. We also find inhibited extracellular signal-regulated kinase (ERK)1/2 phosphorylation in the sh-NDSP cell line. Treating the parental cell line with MAP/ERK kinase 1/2 inhibitors, which diminish ERK1/2 phosphorylation, results in decreased cell proliferation. Culturing these transduced cells with recombinant NDSP, reintroducing NDSP overexpression in the knockdown cell line, or inducing Ras oncogene overexpression for constitutive ERK1/2 activation results in a reversal of the growth-inhibited phenotype and proliferation rates similar to the control cells. In addition, reintroduction of NDSP overexpression in the sh-NDSP cell line results in ERK1/2 phosphorylation similar to control. We conclude that NDSP is specifically overexpressed in neuroblastoma and actively secreted from tumor cells. Furthermore, NDSP serves as a growth factor for neuroblastoma tumor cells through activation of the ERK-mediated proliferation pathway.
Collapse
Affiliation(s)
- Sanjeev A Vasudevan
- Michael E DeBakey Department of Surgery, Dan L Duncan Cancer Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
366
|
Abstract
OBJECTIVE AND METHOD The survival of the patients with neuroblastoma has improved in last few decades. But still it depends on various clinical and biological factors. To assess the clinical features and trends in survival, the data for 500 newly diagnosed patients between January 1972 and December 2004 from a single center were retrospectively analyzed. RESULTS Histopathologic subtypes were neuroblastoma (NBL) in 462 patients (92.4%) and ganglioneuroblastoma in 38 patients (7.6%). The median age was 2.9 years and Male/Female ratio was 1.3/1. Primary tumor sites were abdomen, thorax, pelvis, neck, and others with the frequency of 72.2%, 14.9%, 3.8%, 3.2%, and 5.9%, respectively. There were 30, 49, 133, 257, 31 patients with stage 1, 2, 3, 4, 4S disease and their 10-year survival rates were 100%, 75.8%, 34.1%, 6.5%, and 59.4%, respectively. The outcome has significantly improved according to 10-year periods. The 5-year overall survival rates were 14%, 26.1%, 39.2%, and 52.4% for the years of 1970s, 1980s, 1990s, and after 2000. Surgical procedure involving total or near total tumor removal improved the survival (P=0.002). Both 5-year overall survival and event free survival rates were higher when partial resection was performed, especially in stage 3 disease (P=0.002 and P=0.02). In multivariate analysis, age above 18 months at diagnosis (P=0.01), stage 4 disease (P<0.001), abdominal primary tumor site (P<0.001), NBL subtype in histopathology (P=0.001), responsiveness to chemotherapy (P<0.001) positive or high Vanillyl mandelic acid levels (P=0.02) and male sex (P=0.008) were the determinants of poor prognosis. CONCLUSIONS The survival rates in children with local disease are comparable with the results of developed countries; however, the results in children with advanced disease are still not satisfactory. To improve the outcome, especially in children with advanced disease, more effective chemotherapy regimens and molecular therapies should be investigated. Sharing the knowledge and capacity building to improve the treatment results in NBL are also critical for developing countries.
Collapse
|
367
|
Oberthuer A, Theissen J, Westermann F, Hero B, Fischer M. Molecular characterization and classification of neuroblastoma. Future Oncol 2009; 5:625-39. [DOI: 10.2217/fon.09.41] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
For many decades, neuroblastoma has remained a challenging disease for both clinicians and researchers. Now, techniques that efficiently specify both comprehensive genetic and gene-expression alterations of neuroblastoma tumors have provided molecular markers that indicate tumor behavior and patient outcome with very high accuracy. Once the anticipated value of these markers has been confirmed in ongoing studies, patients may profit from more accurate risk assessment by integrating these markers into clinical routine. Moreover, disclosing further tumor-initiating events, such as the recently revealed oncogenic mutations of ALK, will further promote the elucidation of the genetic etiology of the disease. Together with recent information on altered signaling pathways in aggressively growing tumors, this knowledge will help to establish therapeutic strategies specifically targeting molecular key factors of neuroblastoma tumor progression.
Collapse
Affiliation(s)
- André Oberthuer
- University Children’s Hospital, Department of Pediatric Oncology, Kerpener Strasse 62, 50924 Cologne, Germany
| | - Jessica Theissen
- University of Cologne, Children’s Hospital, Department of Pediatric Oncology, Kerpener Strasse 62, 50924 Cologne, Germany
| | - Frank Westermann
- Department of Tumor Genetics German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Barbara Hero
- University of Cologne, Children’s Hospital, Department of Pediatric Oncology, Kerpener Strasse 62, 50924 Cologne, Germany
| | - Matthias Fischer
- University of Cologne, Children’s Hospital, Department of Pediatric Oncology, Kerpener Strasse 62, 50924 Cologne, Germany
| |
Collapse
|
368
|
Abstract
Neuroblastoma is the third most common pediatric cancer in the United States and is responsible for 15% of pediatric cancer-related deaths. Despite major advances in multimodal therapy, the clinical outcome for several patients remains poor. Due to the desperate need for innovativation and improved success in the treatment and management of neuroblastoma, research interests in immunotherapy have been on the rise in recent years. Current immunotherapeutic approaches under investigation include antibodies targeting the neuroblastoma antigen GD2, cytokine stimulation of immune cells, use of immunocytokine conjugates, radioimmunotherapy, and tumor-primed dendritic cells. Immunotherapy could serve as a safe alternative or adjunct to current therapeutic protocols and would presumptively have fewer deleterious effects making it more favorable to patients.
Collapse
Affiliation(s)
- Latania Y Booker
- Department of Surgery The University of Texas Medical Branch Galveston, TX 77555
| | | | | | | |
Collapse
|
369
|
Hunsberger S, Albert PS, London WB. A finite mixture survival model to characterize risk groups of neuroblastoma. Stat Med 2009; 28:1301-14. [PMID: 19184977 PMCID: PMC4559264 DOI: 10.1002/sim.3543] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Neuroblastoma is a childhood cancer with patients experiencing heterogeneous survival outcomes despite aggressive treatment. Disease outcomes range from early death to spontaneous regression of the tumor followed by cure. Owing to this heterogeneity, it is of interest to identify patients with similar types of neuroblastoma so that specific types of treatment can be developed. Oncologists are especially interested in identifying patients who will be cured so that the minimum amount of a potentially toxic treatment can be given to this group of patients. We analyze a large cohort of neuroblastoma patients and develop a finite mixture model that uses covariates to predict the probability of being in a cure group or other (one or more) risk groups. A prediction method is developed that uses the estimated probabilities to assign a patient to different risk groups. The robustness of the model and the prediction method is examined via simulation by looking at misclassification rates under misspecified models.
Collapse
Affiliation(s)
- Sally Hunsberger
- Biometric Research Branch, National Cancer Institute, 6130 Executive Boulevard, Rm 8120, Rockville MD, 20852
| | - Paul S. Albert
- Biometric Research Branch, National Cancer Institute, 6130 Executive Boulevard, Rm 8120, Rockville MD, 20852
| | - Wendy B. London
- Research Associate Professor & Assoc Program Director, Children’s Oncology Group (COG), University of Florida, 104 N. Main St, #600, Gainesville, FL 32601
| |
Collapse
|
370
|
Pezzolo A, Rossi E, Gimelli S, Parodi F, Negri F, Conte M, Pistorio A, Sementa A, Pistoia V, Zuffardi O, Gambini C. Presence of 1q gain and absence of 7p gain are new predictors of local or metastatic relapse in localized resectable neuroblastoma. Neuro Oncol 2009; 11:192-200. [PMID: 18923191 PMCID: PMC2718991 DOI: 10.1215/15228517-2008-086] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2008] [Accepted: 04/29/2008] [Indexed: 11/19/2022] Open
Abstract
We have addressed the search of novel genetic prognostic markers in a selected cohort of patients with stroma-poor localized resectable neuroblastoma (NB) who underwent relapse or progression (group 1) or complete remission (group 2) over a minimum follow-up of 32 months from diagnosis. Twenty-three Italian patients with localized resectable NB (stages 1 and 2) diagnosed from 1994 through 2005 were studied. All patients received surgical treatment. Chemotherapy was administered only to the three stage 2 patients who had MYCN-amplified tumors. High-resolution array-comparative genomic hybridization (CGH) DNA copy-number analysis technology was used to identify novel prognostic markers. Chromosome 1p36.22p36.32 loss and 1q22qter gain, detected almost exclusively in group 1 patients, were significantly associated with poor event-free survival (EFS) (p = 0.0024 and p = 0.024, respectively). In contrast, patients with 7p11.2p22 gain, who belonged predominantly to group 2, had a significantly better EFS (p = 0.015). The frequency of 17q gain or 3p and 11q losses did not differ significantly in group 1 versus group 2 NBs. The sensitive technique allowed us to define the smallest region of 1p deletion. In conclusion, 1q22qter gain and 7p11.2p22 gain might represent new prognostic markers in localized resectable NB, but the small study size and the retrospective nature of the findings warrant further validation of the results in larger studies.
Collapse
Affiliation(s)
- Annalisa Pezzolo
- Department of Oncology, IRCCS G. Gaslini Hospital, Genova, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
371
|
Volchenboum SL, Cohn SL. Progress in defining and treating high-risk neuroblastoma: lessons from the bench and bedside. J Clin Oncol 2009; 27:1003-4. [PMID: 19171702 DOI: 10.1200/jco.2008.20.2739] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
372
|
Matthay KK, Reynolds CP, Seeger RC, Shimada H, Adkins ES, Haas-Kogan D, Gerbing RB, London WB, Villablanca JG. Long-term results for children with high-risk neuroblastoma treated on a randomized trial of myeloablative therapy followed by 13-cis-retinoic acid: a children's oncology group study. J Clin Oncol 2009; 27:1007-13. [PMID: 19171716 DOI: 10.1200/jco.2007.13.8925] [Citation(s) in RCA: 654] [Impact Index Per Article: 40.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED PURPOSE We assessed the long-term outcome of patients enrolled on CCG-3891, a high-risk neuroblastoma study in which patients were randomly assigned to undergo autologous purged bone marrow transplantation (ABMT) or to receive chemotherapy, and subsequent treatment with 13-cis-retinoic acid (cis-RA). PATIENTS AND METHODS Patients received the same induction chemotherapy, with random assignment (N = 379) to consolidation with myeloablative chemotherapy, total-body irradiation, and ABMT versus three cycles of intensive chemotherapy. Patients who completed consolidation without disease progression were randomly assigned to receive no further therapy or cis-RA for 6 months. Results The event-free survival (EFS) for patients randomly assigned to ABMT was significantly higher than those randomly assigned to chemotherapy; the 5-year EFS (mean +/- SE) was 30% +/- 4% versus 19% +/- 3%, respectively (P = .04). The 5-year EFS (42% +/- 5% v 31% +/- 5%) from the time of second random assignment was higher for cis-RA than for no further therapy, though it was not significant. Overall survival (OS) was significantly higher for each random assignment by a test of the log(-log(.)) transformation of the survival estimates at 5 years (P < .01). The 5-year OS from the second random assignment of patients who underwent both random assignments and who were assigned to ABMT/cis-RA was 59% +/- 8%; for ABMT/no cis-RA, it was 41% +/- 8% [corrected]; for continuing chemotherapy/cis-RA, it was 38% +/- 7%; and for chemotherapy/no cis-RA, it was 36% +/- 7%. CONCLUSION Myeloablative therapy and autologous hematopoietic cell rescue result in significantly better 5-year EFS than nonmyeloablative chemo therapy; neither myeloablative therapy with [corrected] autologous hematopoietic cell rescue nor cis-RA given after consolidation therapy significantly improved OS.
Collapse
Affiliation(s)
- Katherine K Matthay
- University of California School of Medicine, 505 Parnassus Ave, Room M647, San Francisco, CA, 94143-0106, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
373
|
Suh JM, Lee SG, Yoo KH, Sung KW, Koo HH, Kim JY, Cho EJ, Lee SK, Kim J, Lim DH. Outcome of patients with neuroblastoma aged less than 1 year at diagnosis. KOREAN JOURNAL OF PEDIATRICS 2009. [DOI: 10.3345/kjp.2009.52.1.93] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Jung Min Suh
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sang Goo Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Keon Hee Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ki Woong Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hong Hoe Koo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ju Youn Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Eun Joo Cho
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Suk Koo Lee
- Department of Pediatric Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jhingook Kim
- Department of Thoracic Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Do Hoon Lim
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
374
|
Hogarty MD, Norris MD, Davis K, Liu X, Evageliou NF, Hayes CS, Pawel B, Guo R, Zhao H, Sekyere E, Keating J, Thomas W, Cheng NC, Murray J, Smith J, Sutton R, Venn N, London WB, Buxton A, Gilmour SK, Marshall GM, Haber M. ODC1 is a critical determinant of MYCN oncogenesis and a therapeutic target in neuroblastoma. Cancer Res 2008; 68:9735-45. [PMID: 19047152 PMCID: PMC2596661 DOI: 10.1158/0008-5472.can-07-6866] [Citation(s) in RCA: 204] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Neuroblastoma is a frequently lethal childhood tumor in which MYC gene deregulation, commonly as MYCN amplification, portends poor outcome. Identifying the requisite biopathways downstream of MYC may provide therapeutic opportunities. We used transcriptome analyses to show that MYCN-amplified neuroblastomas have coordinately deregulated myriad polyamine enzymes (including ODC1, SRM, SMS, AMD1, OAZ2, and SMOX) to enhance polyamine biosynthesis. High-risk tumors without MYCN amplification also overexpress ODC1, the rate-limiting enzyme in polyamine biosynthesis, when compared with lower-risk tumors, suggesting that this pathway may be pivotal. Indeed, elevated ODC1 (independent of MYCN amplification) was associated with reduced survival in a large independent neuroblastoma cohort. As polyamines are essential for cell survival and linked to cancer progression, we studied polyamine antagonism to test for metabolic dependence on this pathway in neuroblastoma. The Odc inhibitor alpha-difluoromethylornithine (DFMO) inhibited neuroblast proliferation in vitro and suppressed oncogenesis in vivo. DFMO treatment of neuroblastoma-prone genetically engineered mice (TH-MYCN) extended tumor latency and survival in homozygous mice and prevented oncogenesis in hemizygous mice. In the latter, transient Odc ablation permanently prevented tumor onset consistent with a time-limited window for embryonal tumor initiation. Importantly, we show that DFMO augments antitumor efficacy of conventional cytotoxics in vivo. This work implicates polyamine biosynthesis as an arbiter of MYCN oncogenesis and shows initial efficacy for polyamine depletion strategies in neuroblastoma, a strategy that may have utility for this and other MYC-driven embryonal tumors.
Collapse
Affiliation(s)
- Michael D Hogarty
- Division of Oncology, The Children's Hospital of Philadelphia, Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104-4318, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
375
|
Cohn SL, Pearson ADJ, London WB, Monclair T, Ambros PF, Brodeur GM, Faldum A, Hero B, Iehara T, Machin D, Mosseri V, Simon T, Garaventa A, Castel V, Matthay KK. The International Neuroblastoma Risk Group (INRG) classification system: an INRG Task Force report. J Clin Oncol 2008; 27:289-97. [PMID: 19047291 DOI: 10.1200/jco.2008.16.6785] [Citation(s) in RCA: 1296] [Impact Index Per Article: 76.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
PURPOSE Because current approaches to risk classification and treatment stratification for children with neuroblastoma (NB) vary greatly throughout the world, it is difficult to directly compare risk-based clinical trials. The International Neuroblastoma Risk Group (INRG) classification system was developed to establish a consensus approach for pretreatment risk stratification. PATIENTS AND METHODS The statistical and clinical significance of 13 potential prognostic factors were analyzed in a cohort of 8,800 children diagnosed with NB between 1990 and 2002 from North America and Australia (Children's Oncology Group), Europe (International Society of Pediatric Oncology Europe Neuroblastoma Group and German Pediatric Oncology and Hematology Group), and Japan. Survival tree regression analyses using event-free survival (EFS) as the primary end point were performed to test the prognostic significance of the 13 factors. RESULTS Stage, age, histologic category, grade of tumor differentiation, the status of the MYCN oncogene, chromosome 11q status, and DNA ploidy were the most highly statistically significant and clinically relevant factors. A new staging system (INRG Staging System) based on clinical criteria and tumor imaging was developed for the INRG Classification System. The optimal age cutoff was determined to be between 15 and 19 months, and 18 months was selected for the classification system. Sixteen pretreatment groups were defined on the basis of clinical criteria and statistically significantly different EFS of the cohort stratified by the INRG criteria. Patients with 5-year EFS more than 85%, more than 75% to < or = 85%, > or = 50% to < or = 75%, or less than 50% were classified as very low risk, low risk, intermediate risk, or high risk, respectively. CONCLUSION By defining homogenous pretreatment patient cohorts, the INRG classification system will greatly facilitate the comparison of risk-based clinical trials conducted in different regions of the world and the development of international collaborative studies.
Collapse
Affiliation(s)
- Susan L Cohn
- Department of Pediatrics, The University of Chicago, Chicago, IL 60637, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
376
|
Dubois SG, London WB, Zhang Y, Matthay KK, Monclair T, Ambros PF, Cohn SL, Pearson A, Diller L. Lung metastases in neuroblastoma at initial diagnosis: A report from the International Neuroblastoma Risk Group (INRG) project. Pediatr Blood Cancer 2008; 51:589-92. [PMID: 18649370 PMCID: PMC2746936 DOI: 10.1002/pbc.21684] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Neuroblastoma is the most common extracranial pediatric solid cancer. Lung metastasis is rarely detected in children with newly diagnosed neuroblastoma. We aimed to describe the incidence, clinical characteristics, and outcome of patients with lung metastasis at initial diagnosis using a large international database. PROCEDURE The subset of patients from the International Neuroblastoma Risk Group database with INSS stage 4 neuroblastoma and known data regarding lung metastasis at diagnosis was selected for analysis. Clinical and biological characteristics were compared between patients with and without lung metastasis. Survival for patients with and without lung metastasis was estimated by Kaplan-Meier methods. Cox proportional hazards methods were used to determine the independent prognostic value of lung metastasis at diagnosis. RESULTS Of the 2,808 patients with INSS stage 4 neuroblastoma diagnosed between 1990 and 2002, 100 patients (3.6%) were reported to have lung metastasis at diagnosis. Lung metastasis was more common among patients with MYCN amplified tumors, adrenal primary tumors, or elevated lactate dehydrogenase (LDH) levels (P < 0.02 in each case). Five-year overall survival +/- standard error for patients with lung metastasis was 34.5% +/- 6.8% compared to 44.7% +/- 1.3% for patients without lung metastasis (P = 0.0002). However, in multivariable analysis, the presence of lung metastasis was not independently predictive of outcome. CONCLUSIONS Lung metastasis at initial diagnosis of neuroblastoma is associated with MYCN amplification and elevated LDH levels. Although lung metastasis at diagnosis was not independently predictive of outcome in this analysis, it remains a useful prognostic marker of unfavorable outcome.
Collapse
Affiliation(s)
- Steven G Dubois
- Department of Pediatrics, University of California, San Francisco, California, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
377
|
Yanagisawa T, Bartels U, Bouffet E. Role of prognostic factors in the management of pediatric solid tumors. Ann N Y Acad Sci 2008; 1138:32-42. [PMID: 18837880 DOI: 10.1196/annals.1414.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The importance of prognostic factors in predicting outcome in pediatric oncology is largely recognized, and most current protocols tailor treatment based on risk stratification. Further refinements of classical staging systems are ongoing, and the future of pediatric oncology is in the development of strategies based on individual tumor characteristics. This review details significant advances in our understanding of prognostic factors in the most common pediatric solid tumors and potential applications for clinical management.
Collapse
Affiliation(s)
- Takaaki Yanagisawa
- Division of Paediatric Neuro-Oncology, Department of Neuro-Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | | | | |
Collapse
|
378
|
McLaughlin CC, Baptiste MS, Schymura MJ, Zdeb MS, Nasca PC. Perinatal risk factors for neuroblastoma. Cancer Causes Control 2008; 20:289-301. [PMID: 18941915 DOI: 10.1007/s10552-008-9243-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2008] [Accepted: 09/26/2008] [Indexed: 10/21/2022]
Abstract
Neuroblastoma is the most common cancer among infants, suggesting an etiologic role for prenatal factors. In this case-cohort study, neuroblastoma cases (n = 529) diagnosed between 1985 and 2001 were identified from the New York State Cancer Registry and were matched to the electronic birth records for 1983-2001 from New York State and New York City. Controls (n = 12,010) were selected from the same birth cohorts. Analysis was stratified by age at diagnosis, with one to six months (younger infants), seven to 18 months (older infants), and older than 18 months (older children) analyzed separately. Perinatal exposure data was obtained from the birth certificates. No risk factors were identified to be consistently associated with risk across all three age groups. Generally, more risk factors were identified as associated with neuroblastoma among younger infants relative to older ages, including high birth weight, heavier maternal gestational weight gain, maternal hypertension, older maternal age, ultrasound, and respiratory distress. Among older infants, low birth weight was associated with increased risk while heavier maternal gestational weight gain was protective. In the oldest age group, first born status, primary cesarean delivery, prolonged labor and premature rupture of the membranes were associated with increased risk.
Collapse
Affiliation(s)
- Colleen C McLaughlin
- New York State Department of Health, New York State Cancer Registry, Empire State Plaza, Albany, NY 12237, USA.
| | | | | | | | | |
Collapse
|
379
|
Bilke S, Chen QR, Wei JS, Khan J. Whole chromosome alterations predict survival in high-risk neuroblastoma without MYCN amplification. Clin Cancer Res 2008; 14:5540-7. [PMID: 18765546 PMCID: PMC2535913 DOI: 10.1158/1078-0432.ccr-07-4461] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Patients with stage IV neuroblastoma over the age of 500 days without MYCN amplification have a survival rate of <30% and there are currently no reliable means of predicting which of these patients will survive or succumb to the disease. The goal of this study is to develop a DNA copy number-based prognostic profile for these patients. EXPERIMENTAL DESIGN We have used comparative genomic hybridization to identify genome copy number changes that can predict outcome in patients with stage IV neuroblastoma without MYCN amplification. RESULTS A strong correlation of patient survival with the presence of whole chromosome changes (WCC >or=2) was observed, even in the group of patients older than 500 days at time of diagnosis. This novel prognostic marker showed a significant dependence on the date of diagnosis; patients with WCC >or=2 diagnosed after 1998 had a significantly higher probability of survival compared with those diagnosed earlier. At the same time, no such time dependence was found among the samples with WCC <2, suggesting that medical progress patients in recent years has particularly benefited those patients with a stage IV non-MYCN-amplified disease if WCC >or=2 were present. CONCLUSIONS In this pilot study, we present a novel prognostic marker for survival of high-risk neuroblastoma patients over the age of 500 days without MYCN amplification and diagnosed after 1998. Further validation study is required to establish this risk stratification for these patients.
Collapse
Affiliation(s)
- Sven Bilke
- Oncogenomics Section, Pediatric Oncology Branch, Advanced Technology Center, National Cancer Institute, 8717 Grovemont Circle, Gaithersburg, MD 20877
- Cancer Genetics Branch, Center for Cancer Researcher, National Cancer Institute, 37 Convent Drive, Bethesda, MD 20892
| | - Qing-Rong Chen
- Oncogenomics Section, Pediatric Oncology Branch, Advanced Technology Center, National Cancer Institute, 8717 Grovemont Circle, Gaithersburg, MD 20877
- Advanced Biomedical Computing Center, SAIC-Frederick, Inc., NCI-Frederick, Frederick, MD 21702
| | - Jun S. Wei
- Oncogenomics Section, Pediatric Oncology Branch, Advanced Technology Center, National Cancer Institute, 8717 Grovemont Circle, Gaithersburg, MD 20877
| | - Javed Khan
- Oncogenomics Section, Pediatric Oncology Branch, Advanced Technology Center, National Cancer Institute, 8717 Grovemont Circle, Gaithersburg, MD 20877
| |
Collapse
|
380
|
28 years of high-dose therapy and SCT for neuroblastoma in Europe: lessons from more than 4000 procedures. Bone Marrow Transplant 2008; 41 Suppl 2:S118-27. [PMID: 18545256 DOI: 10.1038/bmt.2008.69] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Between 1978 and 2006, the European Group for Blood and Marrow Transplantation registered 4098 high-dose therapy (HDT) procedures followed by stem cell rescue (SCR) (3974 autologous/124 allogeneic) in patients with neuroblastoma. The 5-year rates for overall (OS) and event-free survival are 37 and 32%, respectively. The median age at diagnosis is 3.9 years (0.3-62 years) with 76 patients older than 18 years. Patients above 10 years carry a 2.5-fold higher risk. Younger patients cure significantly (<0.001) better with OS rates of 40 and 30% for age groups 2-4 years and 4-10 years, respectively. Their risks are about twofold higher than that of patients below 2 years with OS rates of 60%. The better the quality of remission status before HDT/SCT the better are the observed OS rates: 43% in CR1 (1199 patients) and 42% for CR2 (140 patients), and 36% for those in very good partial or partial remission (1413 patients) and 21% for those with sensitive relapse (134 patients). Patients reported with stable disease in first remission still had an OS rate of 30%. Multivariate analysis shows significantly better OS in the age group of less than 2 years (<0.0001), as well as a better quality of remission status before HDT/SCT (P<0.0001), with the use of peripheral stem cells (P=0.014), autologous SCT (P=0.031) and busulphan/melphalan HDT (P<0.001). Busulphan/melphalan HDT/SCT in first remission achieves an OS of 48%, while it is only 35% with other regimens (P<0.001), including melphalan alone, other melphalan-containing regimens, a variety of other drugs given as a single HDT as well as the addition of TBI or sequential HDT/SCT procedures. Further progress in the field may only be expected from large-scale international randomized trials.
Collapse
|
381
|
DuBois SG, Matthay KK. Radiolabeled metaiodobenzylguanidine for the treatment of neuroblastoma. Nucl Med Biol 2008; 35 Suppl 1:S35-48. [PMID: 18707633 PMCID: PMC2633223 DOI: 10.1016/j.nucmedbio.2008.05.002] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2008] [Revised: 05/01/2008] [Accepted: 05/06/2008] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Neuroblastoma is the most common pediatric extracranial solid cancer. This tumor is characterized by metaiodobenzylguanidine (MIBG) avidity in 90% of cases, prompting the use of radiolabeled MIBG for targeted radiotherapy in these tumors. METHODS The available English language literature was reviewed for original research investigating in vitro, in vivo and clinical applications of radiolabeled MIBG for neuroblastoma. RESULTS MIBG is actively transported into neuroblastoma cells by the norepinephrine transporter. Preclinical studies demonstrate substantial activity of radiolabeled MIBG in neuroblastoma models, with (131)I-MIBG showing enhanced activity in larger tumors compared to (125)I-MIBG. Clinical studies of (131)I-MIBG in patients with relapsed or refractory neuroblastoma have identified myelosuppression as the main dose-limiting toxicity, necessitating stem cell reinfusion at higher doses. Most studies report a response rate of 30-40% with (131)I-MIBG in this population. More recent studies have focused on the use of (131)I-MIBG in combination with chemotherapy or myeloablative regimens. CONCLUSIONS (131)I-MIBG is an active agent for the treatment of patients with neuroblastoma. Future studies will need to define the optimal role of this targeted radiopharmaceutical in the therapy of this disease.
Collapse
Affiliation(s)
- Steven G DuBois
- Department of Pediatrics, UCSF School of Medicine, Box 0106, San Francisco, CA 94143-0106, USA
| | | |
Collapse
|
382
|
Bénard J, Raguénez G, Kauffmann A, Valent A, Ripoche H, Joulin V, Job B, Danglot G, Cantais S, Robert T, Terrier-Lacombe MJ, Chassevent A, Koscielny S, Fischer M, Berthold F, Lipinski M, Tursz T, Dessen P, Lazar V, Valteau-Couanet D. MYCN-non-amplified metastatic neuroblastoma with good prognosis and spontaneous regression: a molecular portrait of stage 4S. Mol Oncol 2008; 2:261-71. [PMID: 19383347 DOI: 10.1016/j.molonc.2008.07.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2008] [Accepted: 07/16/2008] [Indexed: 10/21/2022] Open
Abstract
Stage 4 neuroblastoma (NB) are heterogeneous regarding their clinical presentations and behavior. Indeed infants (stage 4S and non-stage 4S of age <365days at diagnosis) show regression contrasting with progression in children (>365days). Our study aimed at: (i) identifying age-based genomic and gene expression profiles of stage 4 NB supporting this clinical stratification; and (ii) finding a stage 4S NB signature. Differential genome and transcriptome analyses of a learning set of MYCN-non amplified stage 4 NB tumors at diagnosis (n=29 tumors including 12 stage 4S) were performed using 1Mb BAC microarrays and Agilent 22K probes oligo-microarrays. mRNA chips data following filtering yielded informative genes before supervised hierarchical clustering to identify relationship among tumor samples. After confirmation by quantitative RT-PCR, a stage 4S NB's gene cluster was obtained and submitted to a validation set (n=22 tumors). Genomic abnormalities of infant's tumors (whole chromosomes gains or loss) differ radically from that of children (intra-chromosomal rearrangements) but could not discriminate infants with 4S from those without this presentation. In contrast, differential gene expression by looking at both individual genes and whole biological pathways leads to a molecular stage 4S NB portrait which provides new biological clues about this fascinating entity.
Collapse
Affiliation(s)
- Jean Bénard
- Molecular Interactions in Cancer CNRS-UMR 8126, IFR54, Institut Gustave Roussy, 39, rue C. Desmoulins, Villejuif 94805 Cedex, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
383
|
Abstract
The medical records of 241 children with stage II osteosarcoma were reviewed to assess whether growth characteristics at the time of diagnosis are related to treatment outcome. We defined the maximal growth rate period as an age between 12 and 14 for girls and between 13 and 15 for boys. Accordingly, we subdivided patients into 3 groups and analyzed for clinical characteristics and metastasis-free survival (MFS). Most of the clinical characteristics showed no difference among the 3 groups. After a median follow-up of 54 months (range: 6 to 153 mo), the 5-year MFS of the entire cohort was 67.5%+/-3.1%. Patients in the period of maximal growth rate did worst (57.0%+/-5.6%, P=0.02), whereas patients before and after the period of maximal growth rate fared better (77.0%+/-5.1% and 69.4%+/-4.9%, respectively). On multivariate analysis, disease occurrence in the period of maximal growth rate [relative risk (RR) 2.44; 95% confidence interval (CI) 1.37-4.38; P=0.003], American Joint Committee on Cancer stage IIB disease (RR 2.49; 95% CI 1.49-4.17; P=0.001), and a poor histologic response (RR 2.91; 95% CI 1.78-4.77; P<0.001) independently shortened the MFS. The growth characteristics of patients that are available at the time of diagnosis carry prognostic significance and these could be used as a base to design risk-adapted therapy for osteosarcoma.
Collapse
|
384
|
Voth H, Oberthuer A, Simon T, Kahlert Y, Berthold F, Fischer M. Identification of DEIN, a novel gene with high expression levels in stage IVS neuroblastoma. Mol Cancer Res 2008; 5:1276-84. [PMID: 18171985 DOI: 10.1158/1541-7786.mcr-06-0258] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Neuroblastoma at stage IVS, defined by dissemination to specific tissues and age <1 year at diagnosis, regularly follows spontaneous regression without cytotoxic treatment. To uncover the molecular characteristics of this subtype, Serial Analysis of Gene Expression (SAGE) profiles from stage IVS and fatal stage IV tumors were compared. A SAGE tag (GCAACTTAAC) was detected that was overrepresented in stage IVS disease and that had no reliable match in current National Center for Biotechnology Information databases of SAGE profiles, thus pointing to a novel gene. The corresponding gene, which maps to chromosome 4q33-34, was identified using a modified 3'- and 5'-rapid amplification of cDNA ends PCR and was designated as DEIN (differentially expressed in neuroblastoma). The gene comprises five transcript variants and its sequence overlaps with expressed sequences of the yet uncharacterized UniGene cluster Hs.61435. DEIN exhibits nucleotide sequence conservation over a broad range of species with an overall homology of 65% between human and mouse. As none of the predicted amino acid sequences is homologous to known proteins, it remains to be determined whether DEIN represents a coding or noncoding RNA. Northern blot analysis and semiquantitative reverse transcription-PCR showed high DEIN expression in neuroblastoma, whereas expression was absent or weak in most normal adult tissues. Analysis of 121 primary neuroblastomas by real-time reverse transcription-PCR revealed a strong association with age at diagnosis <1 year and particularly with stage IVS disease (both P < 0.001). The characteristic expression pattern of DEIN suggests a specific role of this gene in the unique biology of stage IVS tumors and may help to molecularly define this special subtype of neuroblastoma.
Collapse
Affiliation(s)
- Harald Voth
- Department of Pediatric Oncology and Hematology, University Children's Hospital of Cologne, Kerpener Strasse 62, 50924 Cologne, Germany
| | | | | | | | | | | |
Collapse
|
385
|
Abstract
Neuroblastoma, a neoplasm of the sympathetic nervous system, is the second most common extracranial malignant tumor of childhood and the most common solid tumor of infancy. Neuroblastoma is a heterogeneous malignancy with prognosis ranging from near uniform survival to high risk for fatal demise. Neuroblastoma serves as a paradigm for the prognostic utility of biologic and clinical data and the potential to tailor therapy for patient cohorts at low, intermediate, and high risk for recurrence. This article summarizes our understanding of neuroblastoma biology and prognostic features and discusses their impact on current and proposed risk stratification schemas, risk-based therapeutic approaches, and the development of novel therapies for patients at high risk for failure.
Collapse
Affiliation(s)
- Julie R Park
- Division of Hematology and Oncology, University of Washington School of Medicine and Children's Hospital and Regional Medical Center, Seattle, WA 98105-0371, USA.
| | | | | |
Collapse
|
386
|
Lee JA, Kim DH, Lim JS, Park KD, Song WS, Lee SY, Jeon DG. The survival of osteosarcoma patients 10 years old or younger is not worse than the survival of older patients: a retrospective analysis. Cancer Res Treat 2007; 39:160-4. [PMID: 19746238 DOI: 10.4143/crt.2007.39.4.160] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2007] [Accepted: 12/18/2007] [Indexed: 11/21/2022] Open
Abstract
PURPOSE This study aimed to assess whether a young age at the time of diagnosis with osteosarcoma has value to predict the prognosis. MATERIALS AND METHODS Sixty-seven children with stage II osteosarcoma were stratified according to the age of 10. There were 32 preadolescents (<or=10 years) and 35 adolescents (10<age<or=15 years). The patients were analyzed for their clinical characteristics, the histologic response to preoperative chemotherapy, event-free survival (EFS) and the patterns of relapse. RESULTS After a median follow-up of 54 months (range: 6 approximately 153 months), the 5-year EFS of the preadolescent and adolescent groups was 64.5+/-9.3% and 58.2+/-9.1%, respectively, and age did not have any statistical significance for survival (p=0.55). Cox regression analysis revealed that both the serum level of alkaline phosphatase and the histologic response to preoperative chemotherapy were significantly related to survival of the 67 patients. Those patients aged less than 7 years responded poorly to preoperative chemotherapy and their rate of amputation was 43%. However, their 5-year EFS was not statistically different from the older patients (57.1+/-18.7 vs 67.7+/-6.3%, respectively, p=0.58). CONCLUSIONS We could not find any statistical difference in the clinical characteristics and survival from osteosarcoma for the preadolescents and adolescents, so the current approach of having the same protocol for both groups of patients seems to be reasonable.
Collapse
Affiliation(s)
- Jun Ah Lee
- Department of Pediatrics, Korea Cancer Center Hospital, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
387
|
|
388
|
Friedman GK, Castleberry RP. Changing trends of research and treatment in infant neuroblastoma. Pediatr Blood Cancer 2007; 49:1060-5. [PMID: 17943963 DOI: 10.1002/pbc.21354] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Neuroblastoma is the most common malignancy in infants and 40% of neuroblastomas are diagnosed in the first year of life. While generally neuroblastoma behaves less aggressively in this age group, tumors that have adverse biologic characteristics do not differ in their behavior from counterparts in older children. Clinical and biologic behavior of neuroblastoma in children up to 460 days of age is similar to that in children less than 1 year of age. Thus the categorization of children up to 18 months of age into risk category is critically dependent on biologic characterization and assignment to appropriate treatment intensity categories.
Collapse
Affiliation(s)
- Gregory K Friedman
- Department of Pediatrics, Division of Pediatric Hematology Oncology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
| | | |
Collapse
|
389
|
Hiyama E, Yamaoka H, Kondo S, Yoneda A, Tajiri T, Fukuzawa M, Sugiyama M, Hayashi Y, Sasaki F, Ohtaki M. Heterogeneous subgroups in human neuroblastoma for clinically relevant risk stratification. Pediatr Surg Int 2007; 23:1051-8. [PMID: 17701415 DOI: 10.1007/s00383-007-1998-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/30/2007] [Indexed: 11/30/2022]
Abstract
Neuroblastoma is a heterogeneous tumor and that may have a favorable or unfavorable prognosis. In Japan, a nation-wide neuroblastoma mass-screening (MS) project assessed 6-month-old infants between 1985 and 2003, and almost all neuroblastomas, including regressing or maturing tumors were thought to be detected in this period. To evaluate the heterogeneity of neuroblastoma subgroups, we analyzed patients with neuroblastoma who had been diagnosed during this period. The clinical courses of 4,209 patients with neuroblastoma, including 1,560 MS detected patients, whose tumors had been diagnosed between 1971 and 1995 were registered. The 2,520 cases registered between 1985 and 1995 were compared to 1,050 cases registered between 1971 and 1980 and analyzed by a multi-gene target model to determine the age distribution of neuroblastoma incidence. We hypothesized that three target genes were responsible for the progression of neuroblastoma: one pair of tumor suppressor gene alleles, one oncogene, and one gene controlling regression/differentiation. This simulation study revealed that the age distribution at initial diagnosis of neuroblastoma was divided into four groups based on post-fertilization age: 20-40, 40-50, 60-90, and 160-200 weeks. Since neuroblatoma in the first group occurred prenatal, post-natal clinical neuroblastoma can be classified into three age groups: 0-6 months, 1-2 years, and 3-4 years. The 0- to 6-month group consisted of mostly benign tumors, and the two older groups had predominantly malignant phenotypes. Our proposed model could explain qualitatively the distribution of neuroblastoma consisting of one subgroup with a favorable prognosis and two subgroups with unfavorable prognosis. For clinically relevant risk stratification, an age cutoff should be considered by the age distribution of these heterogeneous subgroups.
Collapse
Affiliation(s)
- Eiso Hiyama
- Natural Science Center for Basic Research and Development, Hiroshima University, 1-2-3, Kasumi, Hiroshima 734-8551, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
390
|
Abstract
Neuroblastoma is the most common extracranial solid tumor of childhood. It commonly presents in children younger than 2 years of age, with 90% being younger than 5 years of age. There is marked variability in clinical behavior ranging from spontaneous regression or differentiation into benign tumors to rapid and progressive fatal disease. Approximately 50% of patients will have metastases at presentation. The management is dependent on age, stage of disease, and biological and biochemical markers. Nuclear medicine plays an important role in the initial staging, as a prognostic indicator, for assessment of response to treatment, and also in therapy. The most common nuclear medicine diagnostic studies are (99m)Tc-disphosphonate bone scintigraphy and (123)I-MIBG (metaiodobenzylguanidine) scintigraphy. Bone scintigraphy has been the main investigational modality to diagnose skeletal metastases. Whole body imaging with (123)I-MIBG has become the preferred diagnostic test because this agent accumulates in neuroblastoma in 90% to 95% of cases and will accumulate in the primary tumor and metastases particularly in bone, bone marrow, lymph nodes, and soft tissues. MIBG can be used to assess therapy response and is a significant prognostic indicator. Other diagnostic techniques include positron emission tomography (PET)/computed tomography, mainly using (18)F-fluorodeoxyglucose. Other more experimental PET agents, as well as radiolabeled antibodies and octreotide, also are being investigated. Therapy has mainly focused on palliation and has been used alone or in combination with chemotherapy in high-risk refractory or relapsed patients. Major attention is being placed on stratification of patients to try and reduce the side effects associated with intensive megatherapy in the low to intermediate risk patients. Neuroendocrine tumors (NETs) are rare in childhood, but nuclear medicine techniques, mainly using MIBG and somatostatin receptor agents, have a role in diagnosis, staging, and a limited role in therapy. Newer radiopharmaceuticals, including PET agents, are being evaluated for the assessment of NET. Nuclear medicine techniques play a major role in the management of neuroblastoma and NET.
Collapse
Affiliation(s)
- Robert Howman-Giles
- Department of Nuclear Medicine, Children's Hospital at Westmead, Sydney, NSW, Australia.
| | | | | | | |
Collapse
|
391
|
Rabady DZ, Simon JW, Lopasic N. Pediatric horner syndrome: etiologies and roles of imaging and urine studies to detect neuroblastoma and other responsible mass lesions. Am J Ophthalmol 2007; 144:481-2; author reply 482. [PMID: 17765444 DOI: 10.1016/j.ajo.2007.06.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2007] [Revised: 05/16/2007] [Accepted: 06/08/2007] [Indexed: 11/27/2022]
|
392
|
Gutierrez JC, Fischer AC, Sola JE, Perez EA, Koniaris LG. Markedly improving survival of neuroblastoma: a 30-year analysis of 1,646 patients. Pediatr Surg Int 2007; 23:637-46. [PMID: 17476512 DOI: 10.1007/s00383-007-1933-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/28/2007] [Indexed: 11/30/2022]
Abstract
We sought to define current incidence trends and outcomes for pediatric patients with neuroblastoma. The SEER registry was queried from 1973 to 2002. Overall, 1,646 patients with neuroblastoma were identified. The annual incidence has remained unchanged at 0.9 per 100,000. The median age of the population was 1 year, with 42% of patients presenting at <1 year of age. The majority of tumors arose in the retroperitoneum (75.6%) with the remainder located in the mediastinum (15.3%), cervical region (6.6%) and pelvis (2.2%). Markedly improved survival has been noted in each decade (P < 0.002). Sixteen percent of lesions were over 10 cm in greatest dimension, while 84% were high-grade. Disease-specific survival at 1, 2, 5 and 20 years for the entire cohort was 81, 70, 61 and 59%, respectively. Superior survival was observed for infants <1 year of age (P < 0.001). Neuroblastomas in the mediastinum and pelvis had a better prognosis (P < 0.05) while high-grade and lesions over 10 cm carried a worse prognosis (P < 0.022). Surgery but not radiotherapy was associated with improvement in survival (P < 0.001). Multivariate analysis identified age, tumor location, stage, decade of diagnosis and surgical treatment as independent prognostic factors. Neuroblastoma remains a common malignancy with markedly improving patient outcomes. Early diagnosis and surgical therapy continue to provide the best chance for cure. More effective therapies for patients presenting over 1 year of age or those with advanced disease are still needed.
Collapse
Affiliation(s)
- Juan C Gutierrez
- Division of Surgical Oncology, DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | | | | | | | | |
Collapse
|
393
|
Abstract
The clinical hallmark of neuroblastoma is heterogeneity, with the likelihood of cure varying widely according to age at diagnosis, extent of disease, and tumour biology. A subset of tumours will undergo spontaneous regression while others show relentless progression. Around half of all cases are currently classified as high-risk for disease relapse, with overall survival rates less than 40% despite intensive multimodal therapy. This Seminar focuses on recent advances in our understanding of the biology of this complex paediatric solid tumour. We outline plans for the development of a uniform International Neuroblastoma Risk Group (INRG) classification system, and summarise strategies for risk-based therapies. We also update readers on new discoveries related to the underlying molecular pathogenesis of this tumour, with special emphasis on advances that are translatable to the clinic. Finally, we discuss new approaches to treatment, including recently discovered molecular targets that might provide more effective treatment strategies with the potential for less toxicity.
Collapse
Affiliation(s)
- John M Maris
- Children's Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, PA 19104-4318, USA.
| | | | | | | |
Collapse
|
394
|
Schleiermacher G, Michon J, Huon I, d'Enghien CD, Klijanienko J, Brisse H, Ribeiro A, Mosseri V, Rubie H, Munzer C, Thomas C, Valteau-Couanet D, Auvrignon A, Plantaz D, Delattre O, Couturier J. Chromosomal CGH identifies patients with a higher risk of relapse in neuroblastoma without MYCN amplification. Br J Cancer 2007; 97:238-46. [PMID: 17579628 PMCID: PMC2360301 DOI: 10.1038/sj.bjc.6603820] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Whereas neuroblastoma (NB) with MYCN amplification presents a poor prognosis, no single marker allows to reliably predict outcome in tumours without MYCN amplification. We report here an extensive analysis of 147 NB samples at diagnosis, without MYCN amplification, by chromosomal comparative genomic hybridisation (CGH), providing a comprehensive overview of their genomic imbalances. Comparative genomic hybridisation profiles showed gains or losses of entire chromosomes (type 1) in 71 cases, whereas partial chromosome gains or losses (type 2), including gain involving 17q were observed in 68 cases. Atypical profiles were present in eight cases. A type 1 profile was observed more frequently in localised disease (P<0.0001), and in patients of less than 12 months at diagnosis (P<0.0001). A type 2 genomic profile was associated with a higher risk of relapse in the overall population (log-rank test; P<0.0001), but also in the subgroup of patients with localised disease (log-rank test, P=0.007). In multivariate analysis, the genomic profile was the strongest independent prognostic factor. In conclusion, the genomic profile is of prognostic impact in patients without MYCN amplification, making it a help in the management of low-stage NB. Further studies using higher-resolution CGH are needed to better characterise atypical genomic alterations.
Collapse
|
395
|
Sposto R, London WB, Alonzo TA. Criteria for Optimizing Prognostic Risk Groups in Pediatric Cancer: Analysis of Data From the Children's Oncology Group. J Clin Oncol 2007; 25:2070-7. [PMID: 17513812 DOI: 10.1200/jco.2006.09.1983] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PurposePhysicians who treat cancer often attempt to identify patient subgroups that are homogeneous in their chance of recurrence or death as a way to target the more toxic and presumably more effective treatments to patients with the worst prognosis. However, to date, prognosis-based treatment assignment in pediatric cancer has not been based on a quantitative assessment of the risks and benefits of different treatment strategies or on morbidity and efficacy outcome measures that are relevant to children.MethodsWe performed a quantitative analysis of the risks and benefits of prognosis-based treatment assignment in two examples from the Children's Oncology Group using a mathematical model of cancer cure and permanent treatment morbidity and defined an optimality criterion for assigning treatments to specific risk groups.ResultsIn stage 4 MYCN-unamplified neuroblastoma, age-based risk grouping distinguishes clearly between patients with high and low risk of recurrence. However, our analysis suggests that the optimal age cut point depends profoundly on the morbidity of the treatments being considered and agrees with current published recommendations only for treatments that add significant morbidity. In Hodgkin's lymphoma, under our model, no clearly optimal risk groupings exist, and a compelling quantitative rationale for defining risks group at all may not exist.ConclusionOur analysis illustrates the inadequacy of naïve application of statistical criteria for defining prognostic risk groups in pediatric cancer and highlights the importance of quantifying treatment morbidity when defining risk groups or when deciding whether risk grouping is warranted.
Collapse
Affiliation(s)
- Richard Sposto
- Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, Los Angeles, CA 90027-6016, USA.
| | | | | |
Collapse
|
396
|
Martin DT, Gendron RL, Jarzembowski JA, Perry A, Collins MH, Pushpanathan C, Miskiewicz E, Castle VP, Paradis H. Tubedown Expression Correlates with the Differentiation Status and Aggressiveness of Neuroblastic Tumors. Clin Cancer Res 2007; 13:1480-7. [PMID: 17332292 DOI: 10.1158/1078-0432.ccr-06-1716] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The discovery and validation of new prognostic factors and further refinement of risk group stratification are needed to improve clinical interpretation of neuroblastoma. Our laboratory isolated and characterized a developmentally regulated gene named TUBEDOWN against which we have raised a monoclonal antibody (OE5). Tubedown becomes down-regulated postnatally yet remains strongly expressed in some neuroblastomas. The purpose of this study is to define the utility of Tubedown expression as a new measure of the differentiation status and aggressiveness of neuroblastic tumors. EXPERIMENTAL DESIGN Tubedown protein expression was quantitatively assessed in neuroblastic tumors (neuroblastomas, ganglioneuroblastomas, and ganglioneuromas) and normal adrenal tissues using Western blot and OE5 immunohistochemistry. Regulation of Tubedown expression during retinoic acid-induced neuronal differentiation in neuroblastoma cell lines was assessed by Western blotting. RESULTS High levels of Tubedown expression are observed in tumors with significant neuroblastic component, unfavorable histopathology, advanced stage, high-risk group, and poor outcome. In contrast, more differentiated subsets of neuroblastic tumors, ganglioneuroblastomas with favorable histopathology and ganglioneuromas, express low levels of Tubedown. In vitro, marked retinoic acid-induced neuronal differentiation responses of neuroblastoma cells are associated with a significant decrease in Tubedown expression, whereas limited neuronal differentiation responses to retinoic acid were associated with little or no decrease in Tubedown expression. CONCLUSIONS Our results indicate that the levels of Tubedown expression are linked to the differentiation status and aggressiveness of neuroblastic tumors and represent an independent prognostic factor for neuroblastoma. Tubedown expression may be useful to more accurately define different neuroblastic tumor subsets and ultimately provide more adequate assessment and treatment for neuroblastoma patients.
Collapse
Affiliation(s)
- Darryl T Martin
- Department of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
397
|
Current World Literature. Curr Opin Oncol 2007; 19:65-9. [PMID: 17133115 DOI: 10.1097/cco.0b013e328012d5fa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
398
|
Weber A, Starke S, Bergmann E, Christiansen H. The Coamplification Pattern of the MYCN Amplicon Is an Invariable Attribute of Most MYCN-Amplified Human Neuroblastomas. Clin Cancer Res 2006; 12:7316-21. [PMID: 17189403 DOI: 10.1158/1078-0432.ccr-06-0837] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Fifteen percent to 20% of human neuroblastomas show amplification of the MYCN oncogene physiologically located at chromosome 2p24-25, indicating an aggressive subtype of human neuroblastoma with a poor clinical outcome. Recent findings revealed that the structure of the amplicon differs interindividually and that coamplification of genes in telomeric proximity to MYCN might play a relevant role in neuroblastoma development and response to treatment, respectively. We now asked if the amplicon structure is an invariable attribute of an individual tumor or if the coamplification pattern could change during progress or in case of recurrent disease. EXPERIMENTAL DESIGN We used a previously described multiplex PCR approach to analyze the coamplification status of MYCN-amplified human neuroblastomas (n = 33) in tumor tissue at the time of initial diagnosis and in consecutive tissue specimens at later time points after initial treatment or from relapsing disease. The MYCN copy number per haploid genome (Mcn/hg) in these specimens was determined in a separate duplex PCR. RESULTS In 32 of the 33 investigated tumors, the amplicon structure showed no changes after initial chemotherapy and in recurrent disease. Mcn/hg showed a decrease after initial treatment (n = 23), whereas we found a significant increase in recurrent disease (n = 10). CONCLUSION Our data indicate that the initial determined structure of the 2p24-25 amplicon is a consistent attribute in the great majority of the individual MYCN-amplified neuroblastomas and shows no plasticity during or after chemotherapy. Observed changes in the Mcn/hg over the course of disease are in line with preexisting cell culture findings.
Collapse
Affiliation(s)
- Axel Weber
- Children's Hospital, Pediatric Hematology and Oncology, University of Marburg, Marburg, Germany
| | | | | | | |
Collapse
|
399
|
Nanni C, Rubello D, Castellucci P, Farsad M, Franchi R, Rampin L, Gross MD, Al-Nahhas A, Fanti S. 18F-FDG PET/CT fusion imaging in paediatric solid extracranial tumours. Biomed Pharmacother 2006; 60:593-606. [PMID: 16978824 DOI: 10.1016/j.biopha.2006.07.091] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2006] [Accepted: 07/28/2006] [Indexed: 12/21/2022] Open
Abstract
This paper aims at discussing the utility of 18F-FDG PET/CT in the evaluation of paediatric solid extracranial tumours. Following a brief discussion of the basic principles and methodology of PET/CT system, it reviews the main characteristics of the tumours that can be visualised with 18F-FDG PET and presents examples of cases where the combined use of 18F-FDG PET/CT fusion imaging helped in the management of patients. It will also discuss the physiologic biodistribution of 18F-FDG, outlining the normal variants in the paediatric patients that may lead to misinterpretation.
Collapse
Affiliation(s)
- C Nanni
- Nuclear Medicine Department, PET/CT Unit, Policlinico S. Orsola-Malpighi, Bologna, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
400
|
Kushner BH, Cheung NKV. What factors predict a favorable outcome in young children with disseminated neuroblastoma? ACTA ACUST UNITED AC 2006; 3:242-3. [PMID: 16683000 DOI: 10.1038/ncponc0493] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2006] [Accepted: 03/20/2006] [Indexed: 11/09/2022]
Affiliation(s)
- Brian H Kushner
- Neuroblastoma Program, Department of Pediatrics, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA.
| | | |
Collapse
|