1
|
Sen A, Huo Y, Elster J, Zage PE, McVicker G. Allele-specific expression reveals genes with recurrent cis-regulatory alterations in high-risk neuroblastoma. Genome Biol 2022; 23:71. [PMID: 35246212 PMCID: PMC8896304 DOI: 10.1186/s13059-022-02640-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 02/23/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Neuroblastoma is a pediatric malignancy with a high frequency of metastatic disease at initial diagnosis. Neuroblastoma tumors have few recurrent protein-coding mutations but contain extensive somatic copy number alterations (SCNAs) suggesting that mutations that alter gene dosage are important drivers of tumorigenesis. Here, we analyze allele-specific expression in 96 high-risk neuroblastoma tumors to discover genes impacted by cis-acting mutations that alter dosage. RESULTS We identify 1043 genes with recurrent, neuroblastoma-specific allele-specific expression. While most of these genes lie within common SCNA regions, many of them exhibit allele-specific expression in copy neutral samples and these samples are enriched for mutations that are predicted to cause nonsense-mediated decay. Thus, both SCNA and non-SCNA mutations frequently alter gene expression in neuroblastoma. We focus on genes with neuroblastoma-specific allele-specific expression in the absence of SCNAs and find 26 such genes that have reduced expression in stage 4 disease. At least two of these genes have evidence for tumor suppressor activity including the transcription factor TFAP2B and the protein tyrosine phosphatase PTPRH. CONCLUSIONS In summary, our allele-specific expression analysis discovers genes that are recurrently dysregulated by both large SCNAs and other cis-acting mutations in high-risk neuroblastoma.
Collapse
Affiliation(s)
- Arko Sen
- Integrative Biology Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Yuchen Huo
- Department of Pediatrics, Division of Hematology-Oncology, University of California San Diego, La Jolla, California, USA
| | - Jennifer Elster
- Department of Pediatrics, Division of Hematology-Oncology, University of California San Diego, La Jolla, California, USA.,Peckham Center for Cancer and Blood Disorders, Rady Children's Hospital-San Diego, San Diego, California, USA
| | - Peter E Zage
- Department of Pediatrics, Division of Hematology-Oncology, University of California San Diego, La Jolla, California, USA.,Peckham Center for Cancer and Blood Disorders, Rady Children's Hospital-San Diego, San Diego, California, USA
| | - Graham McVicker
- Integrative Biology Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA.
| |
Collapse
|
2
|
MYCN in Neuroblastoma: "Old Wine into New Wineskins". Diseases 2021; 9:diseases9040078. [PMID: 34842635 PMCID: PMC8628738 DOI: 10.3390/diseases9040078] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/20/2021] [Accepted: 10/27/2021] [Indexed: 12/12/2022] Open
Abstract
MYCN Proto-Oncogene, BHLH Transcription Factor (MYCN) has been one of the most studied genes in neuroblastoma. It is known for its oncogenetic mechanisms, as well as its role in the prognosis of the disease and it is considered one of the prominent targets for neuroblastoma therapy. In the present work, we attempted to review the literature, on the relation between MYCN and neuroblastoma from all possible mechanistic sites. We have searched the literature for the role of MYCN in neuroblastoma based on the following topics: the references of MYCN in the literature, the gene's anatomy, along with its transcripts, the protein's anatomy, the epigenetic mechanisms regulating MYCN expression and function, as well as MYCN amplification. MYCN plays a significant role in neuroblastoma biology. Its functions and properties range from the forming of G-quadraplexes, to the interaction with miRNAs, as well as the regulation of gene methylation and histone acetylation and deacetylation. Although MYCN is one of the most primary genes studied in neuroblastoma, there is still a lot to be learned. Our knowledge on the exact mechanisms of MYCN amplification, etiology and potential interventions is still limited. The knowledge on the molecular mechanisms of MYCN in neuroblastoma, could have potential prognostic and therapeutic advantages.
Collapse
|
3
|
Takita J. Molecular Basis and Clinical Features of Neuroblastoma. JMA J 2021; 4:321-331. [PMID: 34796286 PMCID: PMC8580727 DOI: 10.31662/jmaj.2021-0077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 06/02/2021] [Indexed: 12/05/2022] Open
Abstract
Neuroblastoma, a neoplasm of the sympathetic nervous system, originates from neuroblastoma stem cells during embryogenesis. It exhibits unique clinical features including a tendency for spontaneous regression of tumors in infants and a high frequency of metastatic disease at diagnosis in patients aged over 18 months. Genetic risk factors and epigenetic dysregulation also play a significant role in the development of neuroblastoma. Over the past decade, our understanding of this disease has advanced considerably. This has included the identification of chromosomal copy number aberrations specific to neuroblastoma development, risk groups, and disease stage. However, high-risk neuroblastoma remains a therapeutic challenge for pediatric oncologists. New therapeutic approaches have been developed, either as alternatives to conventional chemotherapy or in combination, to overcome the dismal prognosis. Particularly promising strategies are targeted therapies that directly affect cancer cells or cancer stem cells while exhibiting minimal effect on healthy cells. This review summarizes our understanding of neuroblastoma biology and prognostic features and focuses on novel therapeutic strategies for this intractable disease.
Collapse
Affiliation(s)
- Junko Takita
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
4
|
Shawraba F, Hammoud H, Mrad Y, Saker Z, Fares Y, Harati H, Bahmad HF, Nabha S. Biomarkers in Neuroblastoma: An Insight into Their Potential Diagnostic and Prognostic Utilities. Curr Treat Options Oncol 2021; 22:102. [PMID: 34580780 DOI: 10.1007/s11864-021-00898-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2021] [Indexed: 12/23/2022]
Abstract
OPINION STATEMENT Neuroblastoma (NB) is a heterogeneous solid tumor of the pediatric population that originates from neural crest cells and affects the developing sympathetic nervous system. It is the most common neuroblastic tumor accounting for approximately 10% of all childhood cancers and 10-15% of pediatric tumor mortalities. The outcomes range from spontaneous tumor regression in low-risk groups to metastasis and death even after multimodal therapy in high-risk groups. Hence, the detection of NB at an early stage improves outcomes and provides a better prognosis for patients. Early detection and prognosis of NB depend on specific molecules termed biomarkers which can be tissue-specific or circulating. Certain biomarkers are employed in the classification of NB into different groups to improve the treatment and prognosis, and others can be used as therapeutic targets. Therefore, novel biomarker discovery is essential for the early detection of NB, predicting the course of the disease, and developing new targeted treatment strategies. In this review, we aim to summarize the literature pertinent to some important biomarkers of NB and discuss the prognostic role of these biomarkers as well as their potential role in targeted therapy.
Collapse
Affiliation(s)
- Fatima Shawraba
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Hadath, Beirut, Lebanon
| | - Hussein Hammoud
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Hadath, Beirut, Lebanon
| | - Yara Mrad
- Université Clermont Auvergne, Inserm, Neuro-Dol, Clermont-Ferrand, France
| | - Zahraa Saker
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Hadath, Beirut, Lebanon
| | - Youssef Fares
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Hadath, Beirut, Lebanon.,Department of Neurosurgery, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Hayat Harati
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Hadath, Beirut, Lebanon
| | - Hisham F Bahmad
- Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, 4300 Alton Rd, Miami Beach, FL, 33140, USA.
| | - Sanaa Nabha
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Hadath, Beirut, Lebanon.
| |
Collapse
|
5
|
Eleveld TF, Bakali C, Eijk PP, Stathi P, Vriend LE, Poddighe PJ, Ylstra B. Engineering large-scale chromosomal deletions by CRISPR-Cas9. Nucleic Acids Res 2021; 49:12007-12016. [PMID: 34230973 PMCID: PMC8643637 DOI: 10.1093/nar/gkab557] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/07/2021] [Accepted: 06/14/2021] [Indexed: 01/06/2023] Open
Abstract
Large-scale chromosomal deletions are a prevalent and defining feature of cancer. A high degree of tumor-type and subtype specific recurrencies suggest a selective oncogenic advantage. However, due to their large size it has been difficult to pinpoint the oncogenic drivers that confer this advantage. Suitable functional genomics approaches to study the oncogenic driving capacity of large-scale deletions are limited. Here, we present an effective technique to engineer large-scale deletions by CRISPR-Cas9 and create isogenic cell line models. We simultaneously induce double-strand breaks (DSBs) at two ends of a chromosomal arm and select the cells that have lost the intermittent region. Using this technique, we induced large-scale deletions on chromosome 11q (65 Mb) and chromosome 6q (53 Mb) in neuroblastoma cell lines. A high frequency of successful deletions (up to 30% of selected clones) and increased colony forming capacity in the 11q deleted lines suggest an oncogenic advantage of these deletions. Such isogenic models enable further research on the role of large-scale deletions in tumor development and growth, and their possible therapeutic potential.
Collapse
Affiliation(s)
- Thomas F Eleveld
- Department of Pathology, Cancer CenterAmsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Chaimaa Bakali
- Department of Pathology, Cancer CenterAmsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Paul P Eijk
- Department of Pathology, Cancer CenterAmsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Phylicia Stathi
- Department of Pathology, Cancer CenterAmsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Lianne E Vriend
- Department of Clinical Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Pino J Poddighe
- Department of Clinical Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Bauke Ylstra
- Department of Pathology, Cancer CenterAmsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| |
Collapse
|
6
|
Campos Cogo S, Gradowski Farias da Costa do Nascimento T, de Almeida Brehm Pinhatti F, de França Junior N, Santos Rodrigues B, Regina Cavalli L, Elifio-Esposito S. An overview of neuroblastoma cell lineage phenotypes and in vitro models. Exp Biol Med (Maywood) 2020; 245:1637-1647. [PMID: 32787463 PMCID: PMC7802384 DOI: 10.1177/1535370220949237] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
This review was conducted to present the main neuroblastoma (NB) clinical characteristics and the most common genetic alterations present in these pediatric tumors, highlighting their impact in tumor cell aggressiveness behavior, including metastatic development and treatment resistance, and patients' prognosis. The distinct three NB cell lineage phenotypes, S-type, N-type, and I-type, which are characterized by unique cell surface markers and gene expression patterns, are also reviewed. Finally, an overview of the most used NB cell lines currently available for in vitro studies and their unique cellular and molecular characteristics, which should be taken into account for the selection of the most appropriate model for NB pre-clinical studies, is presented. These valuable models can be complemented by the generation of NB reprogrammed tumor cells or organoids, derived directly from patients' tumor specimens, in the direction toward personalized medicine.
Collapse
Affiliation(s)
- Sheron Campos Cogo
- Graduate Program in Health Sciences, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil
| | | | | | - Nilton de França Junior
- Graduate Program in Health Sciences, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil
| | - Bruna Santos Rodrigues
- Graduate Program in Health Sciences, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil
| | - Luciane Regina Cavalli
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba 80250-060, Brazil
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20007, USA
| | - Selene Elifio-Esposito
- Graduate Program in Health Sciences, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil
| |
Collapse
|
7
|
Niu X, Nong S, Gong J, Zhang X, Tang H, Zhou T, Li W. MiR-194 promotes hepatocellular carcinoma through negative regulation of CADM1. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2020; 13:1518-1528. [PMID: 32782670 PMCID: PMC7414468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 04/29/2020] [Indexed: 06/11/2023]
Abstract
Aberrant expression of microRNAs may contribute to the initiation and progression of various types of human cancer and they may also constitute biomarkers for cancer diagnosis and treatment. However, the specific function of miR-194 in hepatocellular carcinoma (HCC), and the potential mechanism of its involvement in HCC were unclear. In the present study, we found that miR-194 inhibited CADM1 protein level expression by inhibiting mRNA translation of CADM1; the expression of CADM1 was low in liver cancer cells and tumor tissues, and the high expression of miR-194 was closely related to HCC. MiR-194 promoted proliferation, invasion, migration, and cell cycle progression of HCC cells, and such promotion effect was inhibited by CADM1. In addition, miR-194 may play a tumor-promoting action in a HCC xenograft tumor model. These results suggested that miR-194 may promote the occurrence and development of HCC by inhibiting CADM1. Therefore, miR-194 may be a promising novel therapy for diagnosis of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Xianli Niu
- Key Laboratory of Viral Biology Guangzhou, Department of Biology, Jinan UniversityGuangzhou, Guangdong, China
- Department of Biochemistry and Molecular Biology, Zhuhai Campus of Zunyi Medical UniversityZhuhai, Guangdong, China
| | - Shirong Nong
- Key Laboratory of Viral Biology Guangzhou, Department of Biology, Jinan UniversityGuangzhou, Guangdong, China
| | - Junyuan Gong
- Key Laboratory of Viral Biology Guangzhou, Department of Biology, Jinan UniversityGuangzhou, Guangdong, China
| | - Xin Zhang
- Key Laboratory of Viral Biology Guangzhou, Department of Biology, Jinan UniversityGuangzhou, Guangdong, China
| | - Hui Tang
- Key Laboratory of Viral Biology Guangzhou, Department of Biology, Jinan UniversityGuangzhou, Guangdong, China
| | - Tianhong Zhou
- Key Laboratory of Viral Biology Guangzhou, Department of Biology, Jinan UniversityGuangzhou, Guangdong, China
| | - Wei Li
- Key Laboratory of Viral Biology Guangzhou, Department of Biology, Jinan UniversityGuangzhou, Guangdong, China
| |
Collapse
|
8
|
Yang L, Li Y, Wei Z, Chang X. Coexpression network analysis identifies transcriptional modules associated with genomic alterations in neuroblastoma. Biochim Biophys Acta Mol Basis Dis 2017; 1864:2341-2348. [PMID: 29247836 DOI: 10.1016/j.bbadis.2017.12.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 12/02/2017] [Accepted: 12/11/2017] [Indexed: 01/28/2023]
Abstract
Neuroblastoma is a highly complex and heterogeneous cancer in children. Acquired genomic alterations including MYCN amplification, 1p deletion and 11q deletion are important risk factors and biomarkers in neuroblastoma. Here, we performed a co-expression-based gene network analysis to study the intrinsic association between specific genomic changes and transcriptome organization. We identified multiple gene coexpression modules which are recurrent in two independent datasets and associated with functional pathways including nervous system development, cell cycle, immune system process and extracellular matrix/space. Our results also indicated that modules involved in nervous system development and cell cycle are highly associated with MYCN amplification and 1p deletion, while modules responding to immune system process are associated with MYCN amplification only. In summary, this integrated analysis provides novel insights into molecular heterogeneity and pathogenesis of neuroblastoma. This article is part of a Special Issue entitled: Accelerating Precision Medicine through Genetic and Genomic Big Data Analysis edited by Yudong Cai & Tao Huang.
Collapse
Affiliation(s)
- Liulin Yang
- College of Electrical Engineering, Guangxi University, Nanning, Guangxi 530004, China; Department of Computer Science, New Jersey Institute of Technology, Newark, NJ 07102, USA
| | - Yun Li
- Department of Biostatistics and Epidemiology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zhi Wei
- Department of Computer Science, New Jersey Institute of Technology, Newark, NJ 07102, USA.
| | - Xiao Chang
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.
| |
Collapse
|
9
|
Takagi M, Yoshida M, Nemoto Y, Tamaichi H, Tsuchida R, Seki M, Uryu K, Nishii R, Miyamoto S, Saito M, Hanada R, Kaneko H, Miyano S, Kataoka K, Yoshida K, Ohira M, Hayashi Y, Nakagawara A, Ogawa S, Mizutani S, Takita J. Loss of DNA Damage Response in Neuroblastoma and Utility of a PARP Inhibitor. J Natl Cancer Inst 2017; 109:4096548. [PMID: 29059438 DOI: 10.1093/jnci/djx062] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 03/13/2017] [Indexed: 11/14/2022] Open
Abstract
Background Neuroblastoma (NB) is the most common solid tumor found in children, and deletions within the 11q region are observed in 11% to 48% of these tumors. Notably, such tumors are associated with poor prognosis; however, little is known regarding the molecular targets located in 11q. Methods Genomic alterations of ATM , DNA damage response (DDR)-associated genes located in 11q ( MRE11A, H2AFX , and CHEK1 ), and BRCA1, BARD1, CHEK2, MDM2 , and TP53 were investigated in 45 NB-derived cell lines and 237 fresh tumor samples. PARP (poly [ADP-ribose] polymerase) inhibitor sensitivity of NB was investigated in in vitro and invivo xenograft models. All statistical tests were two-sided. Results Among 237 fresh tumor samples, ATM, MRE11A, H2AFX , and/or CHEK1 loss or imbalance in 11q was detected in 20.7% of NBs, 89.8% of which were stage III or IV. An additional 7.2% contained ATM rare single nucleotide variants (SNVs). Rare SNVs in DDR-associated genes other than ATM were detected in 26.4% and were mutually exclusive. Overall, samples with SNVs and/or copy number alterations in these genes accounted for 48.4%. ATM-defective cells are known to exhibit dysfunctions in homologous recombination repair, suggesting a potential for synthetic lethality by PARP inhibition. Indeed, 83.3% NB-derived cell lines exhibited sensitivity to PARP inhibition. In addition, NB growth was markedly attenuated in the xenograft group receiving PARP inhibitors (sham-treated vs olaprib-treated group; mean [SD] tumor volume of sham-treated vs olaprib-treated groups = 7377 [1451] m 3 vs 298 [312] m 3 , P = .001, n = 4). Conclusions Genomic alterations of DDR-associated genes including ATM, which regulates homologous recombination repair, were observed in almost half of NBs, suggesting that synthetic lethality could be induced by treatment with a PARP inhibitor. Indeed, DDR-defective NB cell lines were sensitive to PARP inhibitors. Thus, PARP inhibitors represent candidate NB therapeutics.
Collapse
Affiliation(s)
- Masatoshi Takagi
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo, Japan; Department of Pediatrics, Graduate School of Medicine, Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, Laboratory of Sequence Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Pediatrics and Adolescent Medicine, School of Medicine, Juntendo University, Tokyo, Japan; Department of Pediatric Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan, Department of Pediatrics, Nagara Medical Center, Gifu, Japan; Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan; Division of Cancer Genomics, Saitama Cancer Center Research Institute, Saitama, Japan; Division of Cancer Genomics, Chiba Cancer Center, Chiba, Japan; Gunma Children's Medical Center, Gunma, Japan; Saga Medical Center, Saga, Japan
| | - Misa Yoshida
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo, Japan; Department of Pediatrics, Graduate School of Medicine, Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, Laboratory of Sequence Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Pediatrics and Adolescent Medicine, School of Medicine, Juntendo University, Tokyo, Japan; Department of Pediatric Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan, Department of Pediatrics, Nagara Medical Center, Gifu, Japan; Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan; Division of Cancer Genomics, Saitama Cancer Center Research Institute, Saitama, Japan; Division of Cancer Genomics, Chiba Cancer Center, Chiba, Japan; Gunma Children's Medical Center, Gunma, Japan; Saga Medical Center, Saga, Japan
| | - Yoshino Nemoto
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo, Japan; Department of Pediatrics, Graduate School of Medicine, Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, Laboratory of Sequence Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Pediatrics and Adolescent Medicine, School of Medicine, Juntendo University, Tokyo, Japan; Department of Pediatric Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan, Department of Pediatrics, Nagara Medical Center, Gifu, Japan; Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan; Division of Cancer Genomics, Saitama Cancer Center Research Institute, Saitama, Japan; Division of Cancer Genomics, Chiba Cancer Center, Chiba, Japan; Gunma Children's Medical Center, Gunma, Japan; Saga Medical Center, Saga, Japan
| | - Hiroyuki Tamaichi
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo, Japan; Department of Pediatrics, Graduate School of Medicine, Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, Laboratory of Sequence Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Pediatrics and Adolescent Medicine, School of Medicine, Juntendo University, Tokyo, Japan; Department of Pediatric Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan, Department of Pediatrics, Nagara Medical Center, Gifu, Japan; Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan; Division of Cancer Genomics, Saitama Cancer Center Research Institute, Saitama, Japan; Division of Cancer Genomics, Chiba Cancer Center, Chiba, Japan; Gunma Children's Medical Center, Gunma, Japan; Saga Medical Center, Saga, Japan
| | - Rika Tsuchida
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo, Japan; Department of Pediatrics, Graduate School of Medicine, Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, Laboratory of Sequence Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Pediatrics and Adolescent Medicine, School of Medicine, Juntendo University, Tokyo, Japan; Department of Pediatric Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan, Department of Pediatrics, Nagara Medical Center, Gifu, Japan; Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan; Division of Cancer Genomics, Saitama Cancer Center Research Institute, Saitama, Japan; Division of Cancer Genomics, Chiba Cancer Center, Chiba, Japan; Gunma Children's Medical Center, Gunma, Japan; Saga Medical Center, Saga, Japan
| | - Masafumi Seki
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo, Japan; Department of Pediatrics, Graduate School of Medicine, Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, Laboratory of Sequence Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Pediatrics and Adolescent Medicine, School of Medicine, Juntendo University, Tokyo, Japan; Department of Pediatric Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan, Department of Pediatrics, Nagara Medical Center, Gifu, Japan; Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan; Division of Cancer Genomics, Saitama Cancer Center Research Institute, Saitama, Japan; Division of Cancer Genomics, Chiba Cancer Center, Chiba, Japan; Gunma Children's Medical Center, Gunma, Japan; Saga Medical Center, Saga, Japan
| | - Kumiko Uryu
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo, Japan; Department of Pediatrics, Graduate School of Medicine, Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, Laboratory of Sequence Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Pediatrics and Adolescent Medicine, School of Medicine, Juntendo University, Tokyo, Japan; Department of Pediatric Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan, Department of Pediatrics, Nagara Medical Center, Gifu, Japan; Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan; Division of Cancer Genomics, Saitama Cancer Center Research Institute, Saitama, Japan; Division of Cancer Genomics, Chiba Cancer Center, Chiba, Japan; Gunma Children's Medical Center, Gunma, Japan; Saga Medical Center, Saga, Japan
| | - Rina Nishii
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo, Japan; Department of Pediatrics, Graduate School of Medicine, Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, Laboratory of Sequence Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Pediatrics and Adolescent Medicine, School of Medicine, Juntendo University, Tokyo, Japan; Department of Pediatric Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan, Department of Pediatrics, Nagara Medical Center, Gifu, Japan; Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan; Division of Cancer Genomics, Saitama Cancer Center Research Institute, Saitama, Japan; Division of Cancer Genomics, Chiba Cancer Center, Chiba, Japan; Gunma Children's Medical Center, Gunma, Japan; Saga Medical Center, Saga, Japan
| | - Satoshi Miyamoto
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo, Japan; Department of Pediatrics, Graduate School of Medicine, Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, Laboratory of Sequence Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Pediatrics and Adolescent Medicine, School of Medicine, Juntendo University, Tokyo, Japan; Department of Pediatric Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan, Department of Pediatrics, Nagara Medical Center, Gifu, Japan; Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan; Division of Cancer Genomics, Saitama Cancer Center Research Institute, Saitama, Japan; Division of Cancer Genomics, Chiba Cancer Center, Chiba, Japan; Gunma Children's Medical Center, Gunma, Japan; Saga Medical Center, Saga, Japan
| | - Masahiro Saito
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo, Japan; Department of Pediatrics, Graduate School of Medicine, Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, Laboratory of Sequence Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Pediatrics and Adolescent Medicine, School of Medicine, Juntendo University, Tokyo, Japan; Department of Pediatric Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan, Department of Pediatrics, Nagara Medical Center, Gifu, Japan; Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan; Division of Cancer Genomics, Saitama Cancer Center Research Institute, Saitama, Japan; Division of Cancer Genomics, Chiba Cancer Center, Chiba, Japan; Gunma Children's Medical Center, Gunma, Japan; Saga Medical Center, Saga, Japan
| | - Ryoji Hanada
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo, Japan; Department of Pediatrics, Graduate School of Medicine, Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, Laboratory of Sequence Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Pediatrics and Adolescent Medicine, School of Medicine, Juntendo University, Tokyo, Japan; Department of Pediatric Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan, Department of Pediatrics, Nagara Medical Center, Gifu, Japan; Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan; Division of Cancer Genomics, Saitama Cancer Center Research Institute, Saitama, Japan; Division of Cancer Genomics, Chiba Cancer Center, Chiba, Japan; Gunma Children's Medical Center, Gunma, Japan; Saga Medical Center, Saga, Japan
| | - Hideo Kaneko
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo, Japan; Department of Pediatrics, Graduate School of Medicine, Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, Laboratory of Sequence Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Pediatrics and Adolescent Medicine, School of Medicine, Juntendo University, Tokyo, Japan; Department of Pediatric Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan, Department of Pediatrics, Nagara Medical Center, Gifu, Japan; Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan; Division of Cancer Genomics, Saitama Cancer Center Research Institute, Saitama, Japan; Division of Cancer Genomics, Chiba Cancer Center, Chiba, Japan; Gunma Children's Medical Center, Gunma, Japan; Saga Medical Center, Saga, Japan
| | - Satoru Miyano
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo, Japan; Department of Pediatrics, Graduate School of Medicine, Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, Laboratory of Sequence Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Pediatrics and Adolescent Medicine, School of Medicine, Juntendo University, Tokyo, Japan; Department of Pediatric Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan, Department of Pediatrics, Nagara Medical Center, Gifu, Japan; Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan; Division of Cancer Genomics, Saitama Cancer Center Research Institute, Saitama, Japan; Division of Cancer Genomics, Chiba Cancer Center, Chiba, Japan; Gunma Children's Medical Center, Gunma, Japan; Saga Medical Center, Saga, Japan
| | - Keisuke Kataoka
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo, Japan; Department of Pediatrics, Graduate School of Medicine, Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, Laboratory of Sequence Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Pediatrics and Adolescent Medicine, School of Medicine, Juntendo University, Tokyo, Japan; Department of Pediatric Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan, Department of Pediatrics, Nagara Medical Center, Gifu, Japan; Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan; Division of Cancer Genomics, Saitama Cancer Center Research Institute, Saitama, Japan; Division of Cancer Genomics, Chiba Cancer Center, Chiba, Japan; Gunma Children's Medical Center, Gunma, Japan; Saga Medical Center, Saga, Japan
| | - Kenichi Yoshida
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo, Japan; Department of Pediatrics, Graduate School of Medicine, Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, Laboratory of Sequence Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Pediatrics and Adolescent Medicine, School of Medicine, Juntendo University, Tokyo, Japan; Department of Pediatric Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan, Department of Pediatrics, Nagara Medical Center, Gifu, Japan; Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan; Division of Cancer Genomics, Saitama Cancer Center Research Institute, Saitama, Japan; Division of Cancer Genomics, Chiba Cancer Center, Chiba, Japan; Gunma Children's Medical Center, Gunma, Japan; Saga Medical Center, Saga, Japan
| | - Miki Ohira
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo, Japan; Department of Pediatrics, Graduate School of Medicine, Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, Laboratory of Sequence Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Pediatrics and Adolescent Medicine, School of Medicine, Juntendo University, Tokyo, Japan; Department of Pediatric Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan, Department of Pediatrics, Nagara Medical Center, Gifu, Japan; Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan; Division of Cancer Genomics, Saitama Cancer Center Research Institute, Saitama, Japan; Division of Cancer Genomics, Chiba Cancer Center, Chiba, Japan; Gunma Children's Medical Center, Gunma, Japan; Saga Medical Center, Saga, Japan
| | - Yasuhide Hayashi
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo, Japan; Department of Pediatrics, Graduate School of Medicine, Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, Laboratory of Sequence Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Pediatrics and Adolescent Medicine, School of Medicine, Juntendo University, Tokyo, Japan; Department of Pediatric Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan, Department of Pediatrics, Nagara Medical Center, Gifu, Japan; Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan; Division of Cancer Genomics, Saitama Cancer Center Research Institute, Saitama, Japan; Division of Cancer Genomics, Chiba Cancer Center, Chiba, Japan; Gunma Children's Medical Center, Gunma, Japan; Saga Medical Center, Saga, Japan
| | - Akira Nakagawara
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo, Japan; Department of Pediatrics, Graduate School of Medicine, Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, Laboratory of Sequence Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Pediatrics and Adolescent Medicine, School of Medicine, Juntendo University, Tokyo, Japan; Department of Pediatric Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan, Department of Pediatrics, Nagara Medical Center, Gifu, Japan; Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan; Division of Cancer Genomics, Saitama Cancer Center Research Institute, Saitama, Japan; Division of Cancer Genomics, Chiba Cancer Center, Chiba, Japan; Gunma Children's Medical Center, Gunma, Japan; Saga Medical Center, Saga, Japan
| | - Seishi Ogawa
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo, Japan; Department of Pediatrics, Graduate School of Medicine, Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, Laboratory of Sequence Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Pediatrics and Adolescent Medicine, School of Medicine, Juntendo University, Tokyo, Japan; Department of Pediatric Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan, Department of Pediatrics, Nagara Medical Center, Gifu, Japan; Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan; Division of Cancer Genomics, Saitama Cancer Center Research Institute, Saitama, Japan; Division of Cancer Genomics, Chiba Cancer Center, Chiba, Japan; Gunma Children's Medical Center, Gunma, Japan; Saga Medical Center, Saga, Japan
| | - Shuki Mizutani
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo, Japan; Department of Pediatrics, Graduate School of Medicine, Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, Laboratory of Sequence Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Pediatrics and Adolescent Medicine, School of Medicine, Juntendo University, Tokyo, Japan; Department of Pediatric Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan, Department of Pediatrics, Nagara Medical Center, Gifu, Japan; Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan; Division of Cancer Genomics, Saitama Cancer Center Research Institute, Saitama, Japan; Division of Cancer Genomics, Chiba Cancer Center, Chiba, Japan; Gunma Children's Medical Center, Gunma, Japan; Saga Medical Center, Saga, Japan
| | - Junko Takita
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo, Japan; Department of Pediatrics, Graduate School of Medicine, Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, Laboratory of Sequence Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Pediatrics and Adolescent Medicine, School of Medicine, Juntendo University, Tokyo, Japan; Department of Pediatric Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan, Department of Pediatrics, Nagara Medical Center, Gifu, Japan; Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan; Division of Cancer Genomics, Saitama Cancer Center Research Institute, Saitama, Japan; Division of Cancer Genomics, Chiba Cancer Center, Chiba, Japan; Gunma Children's Medical Center, Gunma, Japan; Saga Medical Center, Saga, Japan
| |
Collapse
|
10
|
Cao Y, Jin Y, Yu J, Wang J, Yan J, Zhao Q. Research progress of neuroblastoma related gene variations. Oncotarget 2017; 8:18444-18455. [PMID: 28055978 PMCID: PMC5392342 DOI: 10.18632/oncotarget.14408] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 12/27/2016] [Indexed: 01/08/2023] Open
Abstract
Neuroblastoma, the most common extracranial solid tumor among children, is an embryonal tumor originating from undifferentiated neural crest cell. Neuroblastomas are highly heterogeneous, represented by the wide range of clinical presentations and likelihood of cure, ranging from spontaneous regression to relentless progression despite rigorous multimodal treatments. Approximately, 50% of cases are high-risk with overall survival rates less than 40%. With the efforts to collect large numbers of clinically annotated specimens and the advancements in technologies, researchers have revealed numerous genetic alterations that may drive tumor growth. However, the most lack mutations in genes that are recurrently mutated, which inspires researchers to identify disrupted pathways instead of single mutated genes to unearth biological systems perturbed in neuroblastoma. Stratification of patients and target therapy based on their molecular signatures have been the center of focus. This review provides a comprehensive summary of the recent advances in identification of candidate genes variations, targeted approaches to high-risk neuroblastoma and evaluates the methods utilized for detection, which will provide new avenues to develop therapies and further genetic researches.
Collapse
Affiliation(s)
- Yanna Cao
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, P.R. China
| | - Yan Jin
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, P.R. China
| | - Jinpu Yu
- Department of Cancer Molecular Diagnostic Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, P.R. China
| | - Jingfu Wang
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, P.R. China
| | - Jie Yan
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, P.R. China
| | - Qiang Zhao
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, P.R. China
| |
Collapse
|
11
|
Mlakar V, Jurkovic Mlakar S, Lopez G, Maris JM, Ansari M, Gumy-Pause F. 11q deletion in neuroblastoma: a review of biological and clinical implications. Mol Cancer 2017; 16:114. [PMID: 28662712 PMCID: PMC5492892 DOI: 10.1186/s12943-017-0686-8] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 06/25/2017] [Indexed: 12/12/2022] Open
Abstract
Deletion of the long arm of chromosome 11 (11q deletion) is one of the most frequent events that occur during the development of aggressive neuroblastoma. Clinically, 11q deletion is associated with higher disease stage and decreased survival probability. During the last 25 years, extensive efforts have been invested to identify the precise frequency of 11q aberrations in neuroblastoma, the recurrently involved genes, and to understand the molecular mechanisms of 11q deletion, but definitive answers are still unclear. In this review, it is our intent to compile and review the evidence acquired to date on 11q deletion in neuroblastoma.
Collapse
Affiliation(s)
- Vid Mlakar
- CANSEARCH Research Laboratory, Geneva University Medical School, Avenue de la Roseraie 64, 1205, Geneva, Switzerland
| | - Simona Jurkovic Mlakar
- CANSEARCH Research Laboratory, Geneva University Medical School, Avenue de la Roseraie 64, 1205, Geneva, Switzerland
| | - Gonzalo Lopez
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - John M Maris
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Marc Ansari
- CANSEARCH Research Laboratory, Geneva University Medical School, Avenue de la Roseraie 64, 1205, Geneva, Switzerland.,Department of Pediatrics, Onco-Hematology Unit, Geneva University Hospitals, Rue Willy-Donzé 6, 1205, Geneva, Switzerland
| | - Fabienne Gumy-Pause
- CANSEARCH Research Laboratory, Geneva University Medical School, Avenue de la Roseraie 64, 1205, Geneva, Switzerland. .,Department of Pediatrics, Onco-Hematology Unit, Geneva University Hospitals, Rue Willy-Donzé 6, 1205, Geneva, Switzerland.
| |
Collapse
|
12
|
Masecchia S, Coco S, Barla A, Verri A, Tonini GP. Genome instability model of metastatic neuroblastoma tumorigenesis by a dictionary learning algorithm. BMC Med Genomics 2015; 8:57. [PMID: 26358114 PMCID: PMC4566396 DOI: 10.1186/s12920-015-0132-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 08/28/2015] [Indexed: 12/21/2022] Open
Abstract
Background Metastatic neuroblastoma (NB) occurs in pediatric patients as stage 4S or stage 4 and it is characterized by heterogeneous clinical behavior associated with diverse genotypes. Tumors of stage 4 contain several structural copy number aberrations (CNAs) rarely found in stage 4S. To date, the NB tumorigenesis is not still elucidated, although it is evident that genomic instability plays a critical role in the genesis of the tumor. Here we propose a mathematical approach to decipher genomic data and we provide a new model of NB metastatic tumorigenesis. Method We elucidate NB tumorigenesis using Enhanced Fused Lasso Latent Feature Model (E-FLLat) modeling the array comparative chromosome hybridization (aCGH) data of 190 metastatic NBs (63 stage 4S and 127 stage 4). This model for aCGH segmentation, based on the minimization of functional dictionary learning (DL), combines several penalties tailored to the specificities of aCGH data. In DL, the original signal is approximated by a linear weighted combination of atoms: the elements of the learned dictionary. Results The hierarchical structures for stage 4S shows at the first level of the oncogenetic tree several whole chromosome gains except to the unbalanced gains of 17q, 2p and 2q. Conversely, the high CNA complexity found in stage 4 tumors, requires two different trees. Both stage 4 oncogenetic trees are marked diverged, up to five sublevels and the 17q gain is the most common event at the first level (2/3 nodes). Moreover the 11q deletion, one of the major unfavorable marker of disease progression, occurs before 3p loss indicating that critical chromosome aberrations appear at early stages of tumorigenesis. Finally, we also observed a significant (p = 0.025) association between patient age and chromosome loss in stage 4 cases. Conclusion These results led us to propose a genome instability progressive model in which NB cells initiate with a DNA synthesis uncoupled from cell division, that leads to stage 4S tumors, primarily characterized by numerical aberrations, or stage 4 tumors with high levels of genome instability resulting in complex chromosome rearrangements associated with high tumor aggressiveness and rapid disease progression. Electronic supplementary material The online version of this article (doi:10.1186/s12920-015-0132-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Simona Coco
- Lung Cancer Unit; IRCCS A.O.U. San Martino - IST, Genova, Italy.
| | - Annalisa Barla
- DIBRIS, Università degli Studi di Genova, Genova, Italy.
| | | | - Gian Paolo Tonini
- Neuroblastoma Laboratory, Onco/Hematology Laboratory, Department of Woman and Child Health, University of Padua, Pediatric Research Institute, Fondazione Città della Speranza, Padua, Corso Stati Uniti, 4, 35127, Padua, Italy.
| |
Collapse
|
13
|
Mandriota SJ, Valentijn LJ, Lesne L, Betts DR, Marino D, Boudal-Khoshbeen M, London WB, Rougemont AL, Attiyeh EF, Maris JM, Hogarty MD, Koster J, Molenaar JJ, Versteeg R, Ansari M, Gumy-Pause F. Ataxia-telangiectasia mutated (ATM) silencing promotes neuroblastoma progression through a MYCN independent mechanism. Oncotarget 2015; 6:18558-76. [PMID: 26053094 PMCID: PMC4621910 DOI: 10.18632/oncotarget.4061] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 05/14/2015] [Indexed: 12/13/2022] Open
Abstract
Neuroblastoma, a childhood cancer with highly heterogeneous biology and clinical behavior, is characterized by genomic aberrations including amplification of MYCN. Hemizygous deletion of chromosome 11q is a well-established, independent marker of poor prognosis. While 11q22-q23 is the most frequently deleted region, the neuroblastoma tumor suppressor in this region remains to be identified. Chromosome bands 11q22-q23 contain ATM, a cell cycle checkpoint kinase and tumor suppressor playing a pivotal role in the DNA damage response. Here, we report that haploinsufficiency of ATM in neuroblastoma correlates with lower ATM expression, event-free survival, and overall survival. ATM loss occurs in high stage neuroblastoma without MYCN amplification. In SK-N-SH, CLB-Ga and GI-ME-N human neuroblastoma cells, stable ATM silencing promotes neuroblastoma progression in soft agar assays, and in subcutaneous xenografts in nude mice. This effect is dependent on the extent of ATM silencing and does not appear to involve MYCN. Our findings identify ATM as a potential haploinsufficient neuroblastoma tumor suppressor, whose inactivation mirrors the increased aggressiveness associated with 11q deletion in neuroblastoma.
Collapse
Affiliation(s)
- Stefano J. Mandriota
- Department of Pediatrics, CANSEARCH Research Laboratory, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Linda J. Valentijn
- Department of Oncogenomics, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Laurence Lesne
- Department of Pediatrics, CANSEARCH Research Laboratory, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - David R. Betts
- Department of Clinical Genetics, Our Lady's Children's Hospital, Dublin, Ireland
| | - Denis Marino
- Department of Pediatrics, CANSEARCH Research Laboratory, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Mary Boudal-Khoshbeen
- Department of Pediatrics, CANSEARCH Research Laboratory, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Wendy B. London
- Division of Pediatric Hematology/Oncology, Harvard Medical School, Dana-Farber/Children's Hospital Cancer and Blood Disorders Center, Boston, MA, USA
| | | | - Edward F. Attiyeh
- Department of Pediatrics, Children's Hospital of Philadelphia and the University of Pennsylvania, Philadelphia, PA, USA
| | - John M. Maris
- Department of Pediatrics, Children's Hospital of Philadelphia and the University of Pennsylvania, Philadelphia, PA, USA
| | - Michael D. Hogarty
- Department of Pediatrics, Children's Hospital of Philadelphia and the University of Pennsylvania, Philadelphia, PA, USA
| | - Jan Koster
- Department of Oncogenomics, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Jan J. Molenaar
- Department of Oncogenomics, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Rogier Versteeg
- Department of Oncogenomics, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Marc Ansari
- Department of Pediatrics, CANSEARCH Research Laboratory, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Pediatrics, Onco-hematology Unit, University Hospital of Geneva, Geneva, Switzerland
| | - Fabienne Gumy-Pause
- Department of Pediatrics, CANSEARCH Research Laboratory, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Pediatrics, Onco-hematology Unit, University Hospital of Geneva, Geneva, Switzerland
| |
Collapse
|
14
|
Brodeur GM, Iyer R, Croucher JL, Zhuang T, Higashi M, Kolla V. Therapeutic targets for neuroblastomas. Expert Opin Ther Targets 2014; 18:277-92. [PMID: 24387342 DOI: 10.1517/14728222.2014.867946] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Neuroblastoma (NB) is the most common and deadly solid tumor in children. Despite recent improvements, the long-term outlook for high-risk NB is still < 50%. Further, there is considerable short- and long-term toxicity. More effective, less toxic therapy is needed, and the development of targeted therapies offers great promise. AREAS COVERED Relevant literature was reviewed to identify current and future therapeutic targets that are critical to malignant transformation and progression of NB. The potential or actual NB therapeutic targets are classified into four categories: i) genes activated by amplification, mutation, translocation or autocrine overexpression; ii) genes inactivated by deletion, mutation or epigenetic silencing; iii) membrane-associated genes expressed on most NBs but few other tissues; or iv) common target genes relevant to NB as well as other tumors. EXPERT OPINION Therapeutic approaches have been developed to some of these targets, but many remain untargeted at the present time. It is unlikely that single targeted agents will be sufficient for long-term cure, at least for high-risk NBs. The challenge will be how to integrate targeted agents with each other and with conventional therapy to enhance their efficacy, while simultaneously reducing systemic toxicity.
Collapse
Affiliation(s)
- Garrett M Brodeur
- Children's Hospital of Philadelphia, Division of Oncology , CTRB Rm. 3018, 3501 Civic Center Blvd., Philadelphia, PA 19104-4302 , USA +1 215 590 2817 ; +1 215 590 3770 ;
| | | | | | | | | | | |
Collapse
|
15
|
Domingo-Fernandez R, Watters K, Piskareva O, Stallings RL, Bray I. The role of genetic and epigenetic alterations in neuroblastoma disease pathogenesis. Pediatr Surg Int 2013; 29:101-19. [PMID: 23274701 PMCID: PMC3557462 DOI: 10.1007/s00383-012-3239-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/12/2012] [Indexed: 12/11/2022]
Abstract
Neuroblastoma is a highly heterogeneous tumor accounting for 15 % of all pediatric cancer deaths. Clinical behavior ranges from the spontaneous regression of localized, asymptomatic tumors, as well as metastasized tumors in infants, to rapid progression and resistance to therapy. Genomic amplification of the MYCN oncogene has been used to predict outcome in neuroblastoma for over 30 years, however, recent methodological advances including miRNA and mRNA profiling, comparative genomic hybridization (array-CGH), and whole-genome sequencing have enabled the detailed analysis of the neuroblastoma genome, leading to the identification of new prognostic markers and better patient stratification. In this review, we will describe the main genetic factors responsible for these diverse clinical phenotypes in neuroblastoma, the chronology of their discovery, and the impact on patient prognosis.
Collapse
Affiliation(s)
- Raquel Domingo-Fernandez
- Department of Cancer Genetics, Royal College of Surgeons in Ireland, Dublin, Ireland,Children’s Research Centre, Our Lady’s Children’s Hospital, Crumlin, Dublin, Ireland
| | - Karen Watters
- Department of Cancer Genetics, Royal College of Surgeons in Ireland, Dublin, Ireland,Children’s Research Centre, Our Lady’s Children’s Hospital, Crumlin, Dublin, Ireland
| | - Olga Piskareva
- Department of Cancer Genetics, Royal College of Surgeons in Ireland, Dublin, Ireland,Children’s Research Centre, Our Lady’s Children’s Hospital, Crumlin, Dublin, Ireland
| | - Raymond L. Stallings
- Department of Cancer Genetics, Royal College of Surgeons in Ireland, Dublin, Ireland,Children’s Research Centre, Our Lady’s Children’s Hospital, Crumlin, Dublin, Ireland
| | - Isabella Bray
- Department of Cancer Genetics, Royal College of Surgeons in Ireland, Dublin, Ireland,Children’s Research Centre, Our Lady’s Children’s Hospital, Crumlin, Dublin, Ireland
| |
Collapse
|
16
|
Villamón E, Berbegall AP, Piqueras M, Tadeo I, Castel V, Djos A, Martinsson T, Navarro S, Noguera R. Genetic instability and intratumoral heterogeneity in neuroblastoma with MYCN amplification plus 11q deletion. PLoS One 2013; 8:e53740. [PMID: 23341988 PMCID: PMC3544899 DOI: 10.1371/journal.pone.0053740] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 12/03/2012] [Indexed: 12/14/2022] Open
Abstract
Background/Aim Genetic analysis in neuroblastoma has identified the profound influence of MYCN amplification and 11q deletion in patients’ prognosis. These two features of high-risk neuroblastoma usually occur as mutually exclusive genetic markers, although in rare cases both are present in the same tumor. The purpose of this study was to characterize the genetic profile of these uncommon neuroblastomas harboring both these high-risk features. Methods We selected 18 neuroblastomas with MNA plus 11q loss detected by FISH. Chromosomal aberrations were analyzed using Multiplex Ligation-dependent Probe Amplification and Single Nucleotide Polymorphism array techniques. Results and Conclusion This group of tumors has approximately the same high frequency of aberrations as found earlier for 11q deleted tumors. In some cases, DNA instability generates genetic heterogeneity, and must be taken into account in routine genetic diagnosis.
Collapse
Affiliation(s)
- Eva Villamón
- Department of Pathology, Medical School, University of Valencia, Valencia, Spain
| | - Ana P. Berbegall
- Department of Pathology, Medical School, University of Valencia, Valencia, Spain
| | - Marta Piqueras
- Department of Pathology, Medical School, University of Valencia, Valencia, Spain
| | - Irene Tadeo
- Research Foundation of Hospital Clínico Universitario of Valencia, Valencia, Spain
| | - Victoria Castel
- Pediatric Oncology Unit, Hospital Universitario La Fe, Valencia, Spain
| | - Anna Djos
- Department of Clinical Genetics, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Tommy Martinsson
- Department of Clinical Genetics, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Samuel Navarro
- Department of Pathology, Medical School, University of Valencia, Valencia, Spain
| | - Rosa Noguera
- Department of Pathology, Medical School, University of Valencia, Valencia, Spain
- * E-mail:
| |
Collapse
|
17
|
Jeison M, Yaniv I, Ash S. Genetic stratification of neuroblastoma for treatment tailoring. Future Oncol 2012; 7:1087-99. [PMID: 21919696 DOI: 10.2217/fon.11.87] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Neuroblastoma is the most common extracranial tumor of childhood. The clinical behavior is variable, ranging from spontaneous regression to fatal progression despite aggressive therapy. The most highly statistically significant and clinically relevant factors that are currently used for classification include stage, age, histopathologic category, MYCN oncogene status, chromosome 11q status and DNA ploidy. These genetic markers were analyzed separately by classical methods until recently: mainly fluorescence in situ hybridization or loss of heterozygosity. The development of genome-wide techniques such as comparative genomic hybridization, array comparative genomic hybridization and single nucleotide polymorphism allows the analysis of copy number variations through the whole genome in one step. This enabled the investigators to refine different genetic subtypes for the better comprehension of neuroblastoma tumor behavior and reach the conclusion that these data together with a genomic profile based on gene expression should be included in future treatment stratification.
Collapse
Affiliation(s)
- Marta Jeison
- Department of Pediatric Hematology-Oncology, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | | | | |
Collapse
|
18
|
Neuroblastoma genetics and phenotype: a tale of heterogeneity. Semin Cancer Biol 2011; 21:238-44. [PMID: 21839839 DOI: 10.1016/j.semcancer.2011.07.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Accepted: 07/13/2011] [Indexed: 12/31/2022]
Abstract
Cancer is a complex disease driven by multiple genetic and epigenetic alterations. Understanding the (epi-)genetic changes and consequent deregulation of regulatory networks controlling the various normal critical cellular phenotypes that are perturbed in cancer cells can provide clues to new therapeutic opportunities. Moreover, such insights into the molecular pathology of a given cancer type can offer clinical relevant genetic markers or molecular signatures for assessment of prognosis and response to therapy, and prediction of risk for relapse. Therefore, as for many other tumour entities, neuroblastoma (NB) has been the subject of intensive ongoing genomic research. Here we will summarize the current state-of-the-art of these studies with focus on genome wide DNA copy number and gene expression analyses in relation to the relevance for present and future clinical management of NB patients.
Collapse
|
19
|
Ito T, Williams-Nate Y, Iwai M, Tsuboi Y, Hagiyama M, Ito A, Sakurai-Yageta M, Murakami Y. Transcriptional regulation of the CADM1 gene by retinoic acid during the neural differentiation of murine embryonal carcinoma P19 cells. Genes Cells 2011; 16:791-802. [DOI: 10.1111/j.1365-2443.2011.01525.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
20
|
Parodi F, Passoni L, Massimo L, Luksch R, Gambini C, Rossi E, Zuffardi O, Pistoia V, Pezzolo A. Identification of novel prognostic markers in relapsing localized resectable neuroblastoma. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2011; 15:113-21. [PMID: 21319993 DOI: 10.1089/omi.2010.0085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Patients with localized resectable neuroblastoma (NB) generally have an excellent prognosis and can be treated by surgery alone, but approximately 10% of them develop local recurrences or metastatic progression. The known predictive risk factors are important for the identification of localized resectable NB patients at risk of relapse and/or progression, who may benefit from early and aggressive treatment. These factors, however, identify only a subset of patients at risk, and the search for novel prognostic markers is warranted. This review focuses on the recent advances in the identification of new prognostic markers. Recently we addressed the search of novel genetic prognostic markers in a selected cohort of patients with stroma-poor localized resectable NB who underwent disease relapse or progression (group 1) or complete remission (group 2). High-resolution array-comparative genomic hybridization (CGH) DNA copy-number analysis technology was used. Chromosome 1p36.22p36.32 loss and 1q22qter gain, detected almost exclusively in group 1 patients, were significantly associated with poor event-free survival (EFS). Increasing evidence points to anaplastic lymphoma kinase (ALK) as a fundamental oncogene associated with NB. The immunohistochemical analysis of sporadic NB localized resectable primary tumors (stage 1-2) showed a correlation between aberrant ALK level of expression and tumor progression and clinical outcome. Moreover, other factors that might influence the clinical behavior of these tumors will be reviewed.
Collapse
Affiliation(s)
- Federica Parodi
- Laboratory of Oncology, IRCCS G.Gaslini Hospital, Genova, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
|
22
|
Chang G, Xu S, Dhir R, Chandran U, O'Keefe DS, Greenberg NM, Gingrich JR. Hypoexpression and epigenetic regulation of candidate tumor suppressor gene CADM-2 in human prostate cancer. Clin Cancer Res 2010; 16:5390-401. [PMID: 21062931 DOI: 10.1158/1078-0432.ccr-10-1461] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Cell adhesion molecules (CADM) comprise a newly identified protein family whose functions include cell polarity maintenance and tumor suppression. CADM-1, CADM-3, and CADM-4 have been shown to act as tumor suppressor genes in multiple cancers including prostate cancer. However, CADM-2 expression has not been determined in prostate cancer. EXPERIMENTAL DESIGN The CADM-2 gene was cloned and characterized and its expression in human prostatic cell lines and cancer specimens was analyzed by reverse transcription-PCR and an immunohistochemical tissue array, respectively. The effects of adenovirus-mediated CADM-2 expression on prostate cancer cells were also investigated. CADM-2 promoter methylation was evaluated by bisulfite sequencing and methylation-specific PCR. RESULTS We report the initial characterization of CADM-2 isoforms: CADM-2a and CADM-2b, each with separate promoters, in human chromosome 3p12.1. Prostate cancer cell lines, LNCaP and DU145, expressed negligible CADM-2a relative to primary prostate tissue and cell lines, RWPE-1 and PPC-1, whereas expression of CADM-2b was maintained. Using immunohistochemistry, tissue array results from clinical specimens showed statistically significant decreased expression in prostate carcinoma compared with normal donor prostate, benign prostatic hyperplasia, prostatic intraepithelial neoplasia, and normal tissue adjacent to tumor (P < 0.001). Adenovirus-mediated CADM-2a expression suppressed DU145 cell proliferation in vitro and colony formation in soft agar. The decrease in CADM-2a mRNA in cancer cell lines correlated with promoter region hypermethylation as determined by bisulfite sequencing and methylation-specific PCR. Accordingly, treatment of cells with the demethylating agent 5-aza-2'-deoxycytidine alone or in combination with the histone deacetylase inhibitor trichostatin A resulted in the reactivation of CADM-2a expression. CONCLUSIONS CADM-2a protein expression is significantly reduced in prostate cancer. Its expression is regulated in part by promoter methylation and implicates CADM-2 as a previously unrecognized tumor suppressor gene in a proportion of human prostate cancers.
Collapse
Affiliation(s)
- Guimin Chang
- Department of Urology, University of Pittsburgh, 5200 Centre Avenue, Pittsburgh, PA 15232, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Pediatric mixed-lineage leukemia (MLL)-rearranged acute monoblastic leukemia with t(9;11)(p22;q23) has a favorable outcome compared with other MLL-rearranged AML. The biologic background for this difference remains unknown. Therefore, we compared gene expression profiles (GEPs; Affymetrix HGU133 + 2.0) of 26 t(9;11)(p22;q23) patients with 42 other MLL-rearranged AML patients to identify differentially expressed genes. IGSF4, a cell-cell adhesion molecule, was found to be highly expressed in t(9;11)(p22;q23) patients, which was confirmed by real-time quantitative polymerase chain reaction and Western blot. IGSF4 expression within t(9;11)(p22;q23) patients was 4.9 times greater in French-American-British morphology classification (FAB)-M5 versus other FAB-types (P = .001). Methylation status investigation showed that high IGSF4-expressing t(9;11)(p22;q23) patients with FAB-M5 have no promoter hypermethylation, whereas all other cases do. Cell-line incubation with demethylating agent decitabine resulted in promoter demethylation and increased expression of IGSF4. Down-regulation of IGSF4 by siRNA did not affect proliferation or drug sensitivity. In a cohort of 79 MLL-rearranged AML cases, we show significant better overall survival for cases with high IGSF4 expression (5-year overall survival 0.70 vs 0.37, P = .03) In conclusion, we identified IGSF4 overexpression to be discriminative for t(9;11)(p22;q23) patients with FAB-M5, regulated partially by promoter methylation and resulting in survival benefit.
Collapse
|
24
|
Ming S, Gao J, Sun T. [The relationship between the TSLC1 silencing and DNA methylation in human lung cancer cells]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2010; 13:464-9. [PMID: 20677643 PMCID: PMC6000693 DOI: 10.3779/j.issn.1009-3419.2010.05.16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
背景与目的 TSLC1在多种肿瘤中表达下调或失活,其表达下调与DNA高甲基化有很大关系。本研究旨在探索TSLC1在肺癌细胞中的表达缺失与其启动子区DNA甲基化的关系。 方法 采用RT-PCR和Real-time PCR方法检测TSLC1在正常肺组织和3种肺癌细胞系(A549、NCI-H446和Calu-3)中的表达谱;运用亚硫酸氢盐修饰后测序(bisulfte sequencing)方法检测上述正常肺组织和肺癌细胞中TSLC1启动子区的甲基化状态;应用甲基化转移酶抑制剂5-氮-2-脱氧胞苷(5-Aza-dC)处理上述细胞后,采用Real-time PCR方法检测处理前后TSLC1的表达变化。 结果 TSLC1在正常肺组织和A549细胞中表达,其启动子区DNA未发生甲基化;而在NCI-H446和Calu-3细胞中表达缺失,其启动子区DNA发生高甲基化,并且5-Aza-dC处理NCI-H446和Calu-3细胞后可促进TSLC1的表达。 结论 TSLC1在肺癌细胞中的表达缺失是由其启动子区的DNA高甲基化引起。
Collapse
Affiliation(s)
- Shuhong Ming
- Department of Respiratory Medicine, Beijing Hospital Ministry of Health, Beijing 100730, China.
| | | | | |
Collapse
|
25
|
Chen QR, Song YK, Yu LR, Wei JS, Chung JY, Hewitt SM, Veenstra TD, Khan J. Global genomic and proteomic analysis identifies biological pathways related to high-risk neuroblastoma. J Proteome Res 2010; 9:373-82. [PMID: 19921788 PMCID: PMC2801773 DOI: 10.1021/pr900701v] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Neuroblastoma (NB) is a heterogeneous pediatric tumor. To better understand the biological pathways involved in the development of high-risk neuroblastoma, we performed parallel global protein and mRNA expression profiling on NB tumors of stage 4 MYCN-amplified (4+) and stage 1 MYCN-not-amplified (1-) using isotope-coded affinity tags (ICAT) and Affymetrix U133plus2 microarray, respectively. A total of 1461 proteins represented by 2 or more peptides were identified from the quantitative ICAT analysis, of which 433 and 130 proteins are up- or down-regulated, respectively, in 4+ tumor compared to the 1- tumor. Pathway analysis of the differentially expressed proteins showed the enrichment of glycolysis, DNA replication and cell cycle processes in the up-regulated proteins and cell adhesion, nervous system development and cell differentiation processes in the down-regulated proteins in 4+ tumor; suggesting a less mature neural and a more invasive phenotype of 4+ tumor. Myc targets and ribosomal proteins are overrepresented in the 4+ tumors as expected; functional gene sets reported to be enriched in neural and embryonic stem cells are significantly enriched in the 4+ tumor, indicating the existence of a stemness signature in MYCN-amplified stage 4 tumor. In addition, protein and mRNA expression are moderately correlated (r = 0.51, p < 0.0001), as approximately half of the up-regulated proteins in 4+ tumor have elevated mRNA level (n = 208), and one-third of down-regulated proteins have lower mRNA expression (n = 47). Further biological network analysis revealed that the differentially expressed proteins closely interact with other proteins of known networks; the important role of MYCN is confirmed and other transcription factors identified in the network may have potential roles in the biology of NB tumor. We used global genomic and proteomic analysis to identify biologically relevant proteins and pathways important to NB progression and development that may provide new insights into the biology of advanced neuroblastoma.
Collapse
Affiliation(s)
- Qing-Rong Chen
- Oncogenomics Section, Pediatric Oncology Branch, Advanced Technology Center, National Cancer Institute, 8717 Grovemont Circle, Gaithersburg, MD 20877
- Bioinformatics Support Group, Advanced Biomedical Computing Center, SAIC-Frederick, Inc., NCI-Frederick, Frederick, Maryland 21702
| | - Young K Song
- Oncogenomics Section, Pediatric Oncology Branch, Advanced Technology Center, National Cancer Institute, 8717 Grovemont Circle, Gaithersburg, MD 20877
| | - Li-Rong Yu
- Laboratory of Proteomics and Analytical Technologies, Advanced Technology Program, SAIC-Frederick, Inc., NCI-Frederick, Frederick, Maryland 21702
| | - Jun S. Wei
- Oncogenomics Section, Pediatric Oncology Branch, Advanced Technology Center, National Cancer Institute, 8717 Grovemont Circle, Gaithersburg, MD 20877
| | - Joon-Yong Chung
- Tissue Array Research Program, Laboratory of Pathology, National Cancer Institute, 8717 Grovemont Circle, Gaithersburg, MD 20877
| | - Stephen M. Hewitt
- Tissue Array Research Program, Laboratory of Pathology, National Cancer Institute, 8717 Grovemont Circle, Gaithersburg, MD 20877
| | - Timothy D. Veenstra
- Laboratory of Proteomics and Analytical Technologies, Advanced Technology Program, SAIC-Frederick, Inc., NCI-Frederick, Frederick, Maryland 21702
| | - Javed Khan
- Oncogenomics Section, Pediatric Oncology Branch, Advanced Technology Center, National Cancer Institute, 8717 Grovemont Circle, Gaithersburg, MD 20877
| |
Collapse
|
26
|
|
27
|
Van Roy N, De Preter K, Hoebeeck J, Van Maerken T, Pattyn F, Mestdagh P, Vermeulen J, Vandesompele J, Speleman F. The emerging molecular pathogenesis of neuroblastoma: implications for improved risk assessment and targeted therapy. Genome Med 2009; 1:74. [PMID: 19638189 PMCID: PMC2717400 DOI: 10.1186/gm74] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Neuroblastoma is one of the most common solid tumors of childhood, arising from immature sympathetic nervous system cells. The clinical course of patients with neuroblastoma is highly variable, ranging from spontaneous regression to widespread metastatic disease. Although the outcome for children with cancer has improved considerably during the past decades, the prognosis of children with aggressive neuroblastoma remains dismal. The clinical heterogeneity of neuroblastoma mirrors the biological and genetic heterogeneity of these tumors. Ploidy and MYCN amplification have been used as genetic markers for risk stratification and therapeutic decision making, and, more recently, gene expression profiling and genome-wide DNA copy number analysis have come into the picture as sensitive and specific tools for assessing prognosis. The applica tion of new genetic tools also led to the discovery of an important familial neuroblastoma cancer gene, ALK, which is mutated in approximately 8% of sporadic tumors, and genome-wide association studies have unveiled loci with risk alleles for neuroblastoma development. For some of the genomic regions that are deleted in some neuroblastomas, on 1p, 3p and 11q, candidate tumor suppressor genes have been identified. In addition, evidence has emerged for the contribution of epigenetic disturbances in neuroblastoma oncogenesis. As in other cancer entities, altered microRNA expression is also being recognized as an important player in neuroblastoma. The recent successes in unraveling the genetic basis of neuroblastoma are now opening opportunities for development of targeted therapies.
Collapse
Affiliation(s)
- Nadine Van Roy
- Center for Medical Genetics, Ghent University Hospital, De Pintelaan 185, B-9000 Ghent, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Vermeulen J, De Preter K, Naranjo A, Vercruysse L, Van Roy N, Hellemans J, Swerts K, Bravo S, Scaruffi P, Tonini GP, De Bernardi B, Noguera R, Piqueras M, Cañete A, Castel V, Janoueix-Lerosey I, Delattre O, Schleiermacher G, Michon J, Combaret V, Fischer M, Oberthuer A, Ambros PF, Beiske K, Bénard J, Marques B, Rubie H, Kohler J, Pötschger U, Ladenstein R, Hogarty MD, McGrady P, London WB, Laureys G, Speleman F, Vandesompele J. Predicting outcomes for children with neuroblastoma using a multigene-expression signature: a retrospective SIOPEN/COG/GPOH study. Lancet Oncol 2009; 10:663-71. [PMID: 19515614 PMCID: PMC3045079 DOI: 10.1016/s1470-2045(09)70154-8] [Citation(s) in RCA: 144] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND More accurate prognostic assessment of patients with neuroblastoma is required to better inform the choice of risk-related therapy. The aim of this study is to develop and validate a gene-expression signature to improve outcome prediction. METHODS 59 genes were selected using an innovative data-mining strategy, and were profiled in the largest neuroblastoma patient series (n=579) to date using real-time quantitative PCR starting from only 20 ng of RNA. A multigene-expression signature was built using 30 training samples, tested on 313 test samples, and subsequently validated in a blind study on an independent set of 236 tumours. FINDINGS The signature has a performance, sensitivity, and specificity of 85.4% (95% CI 77.7-93.2), 84.4% (66.5-94.1), and 86.5% (81.1-90.6), respectively, to predict patient outcome. Multivariate analysis indicates that the signature is a significant independent predictor of overall survival and progression-free survival after controlling for currently used risk factors: patients with high molecular risk have a higher risk of death from disease and higher risk of relapse or progression than patients with low molecular risk (odds ratio 19.32 [95% CI 6.50-57.43] and 3.96 [1.97-7.97] for overall survival and progression-free survival, respectively, both p<0.0001). Patients at an increased risk of an adverse outcome can also be identified in the current treatment groups, showing the potential of this signature for improved clinical management. These results were confirmed in the validation study, in which the signature was also independently statistically significant in a model adjusted for MYCN status, age, International Neuroblastoma Staging System stage, ploidy, International Neuroblastoma Pathology Classification grade of differentiation, and mitosis karyorrhexis index (odds ratios between 4.81 and 10.53 depending on the model for overall survival and 3.68 [95% CI 2.01-6.71] for progression-free survival). INTERPRETATION The 59-gene expression signature is an accurate predictor of outcome in patients with neuroblastoma. The signature is an independent risk predictor, identifying patients with an increased risk of poor outcome in the current clinical-risk groups. The method and signature is suitable for routine laboratory testing, and should be evaluated in prospective studies. FUNDING The Belgian Foundation Against Cancer, the Children Cancer Fund Ghent, the Belgian Society of Paediatric Haematology and Oncology, the Belgian Kid's Fund and the Fondation Nuovo-Soldati (JV), the Fund for Scientific Research Flanders (KDP, JH), the Fund for Scientific Research Flanders, the Institute for the Promotion of Innovation by Science and Technology in Flanders, Strategisch basisonderzoek, the Fondation Fournier Majoie pour l'Innovation, the Instituto Carlos III, the Italian Neuroblastoma Foundation, the European Community under the FP6, and the Belgian programme of Interuniversity Poles of Attraction.
Collapse
Affiliation(s)
- Joëlle Vermeulen
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|