1
|
Langenberg KPS, van Hooff SR, Koopmans B, Strijker JGM, Kholosy WM, Ober K, Zwijnenburg DA, van der Hoek JJF, Keller KM, Vernooij L, Schild LG, Looze EJ, Ebus ME, Essing AHW, Vree PD, Tas ML, Matser YAH, Wienke J, Volckmann R, Tops BBJ, Kester LA, Badloe S, Hehir-Kwa JY, Kemmeren P, Goemans BF, Zwaan CM, Oehme I, Jäger N, Witt O, van Eijkelenburg NKA, Dierselhuis MP, Tytgat GAM, Wijnen MHW, van Noesel MM, de Krijger RR, Eising S, Koster J, Dolman EM, Molenaar JJ. Exploring high-throughput drug sensitivity testing in neuroblastoma cell lines and patient-derived tumor organoids in the era of precision medicine. Eur J Cancer 2025; 218:115275. [PMID: 39954414 PMCID: PMC11884408 DOI: 10.1016/j.ejca.2025.115275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 01/17/2025] [Accepted: 01/29/2025] [Indexed: 02/17/2025]
Abstract
INTRODUCTION Despite druggable events to be present in 80 % of neuroblastomapatients within the Princess Máxima Center precision medicine program 'iTHER', clinical uptake of treatment recommendations has been low, and the clinical impact for individual patients remains hard to predict. This stresses the need for a method integrating genomics and transcriptomics with functional approaches into therapeutic decision making. METHODS We aimed to launch an online repository integrating genomics and transcriptomics with high-throughput drug screening (HTS) of nineteen commonly used neuroblastoma cell lines and fifteen neuroblastoma patient-derived organoids (NBL-PDOs). Cell lines, NBL-PDOs and their parental tumors were characterized utilizing (lc)WGS, WES and RNAseq. Cells were exposed to ∼200 compounds. Results were transferred to the R2 visualization platform. RESULTS A powerful reference set of cell lines is available, reflecting distinct known pharmacologic vulnerabilities. HTS identified additional therapeutic vulnerabilities, such as a striking correlation between a positive mesenchymal signature and sensitivity to BCL2-inhibitor venetoclax. Finally, we explored personalized drug sensitivities within iTHER, demonstrating HTS can support genomic and transcriptomic results, thereby strengthening the rationale for clinical uptake. CONCLUSION We established a dynamic publicly available dataset with detailed genomic, transcriptomic, and pharmacological annotation of classical neuroblastoma cell lines as well as novel sharable NBL-PDOs, representing the heterogeneous landscape of neuroblastoma. We anticipate that in vitro drug screening will be complementary to genomic-guided precision medicine by supporting clinical decision making, thereby improving prognosis for all neuroblastoma patients in the future.
Collapse
Affiliation(s)
- Karin P S Langenberg
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Sander R van Hooff
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Bianca Koopmans
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Josephine G M Strijker
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Waleed M Kholosy
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Kimberley Ober
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Danny A Zwijnenburg
- Department of Oncogenomics, Cancer Center Amsterdam, Amsterdam UMC, the Netherlands.
| | - Jessica J F van der Hoek
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands
| | - Kaylee M Keller
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Lindy Vernooij
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands
| | - Linda G Schild
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Eleonora J Looze
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Marli E Ebus
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands
| | - Anke H W Essing
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands
| | - Paula de Vree
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands
| | - Michelle L Tas
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands
| | - Yvette A H Matser
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Judith Wienke
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Richard Volckmann
- Department of Oncogenomics, Cancer Center Amsterdam, Amsterdam UMC, the Netherlands.
| | - Bastiaan B J Tops
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Lennart A Kester
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Shashi Badloe
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Jayne Y Hehir-Kwa
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Patrick Kemmeren
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands; Center for Molecular Medicine, University Medical Center Utrecht, Heidelberglaan 100, Utrecht 3584 CX, the Netherlands.
| | - Bianca F Goemans
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - C Michel Zwaan
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Ina Oehme
- Hopp Children's Cancer Center Heidelberg (KiTZ), German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, Heidelberg 69120 , the Netherlands; Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), and German Cancer Consortium (DKTK), Im Neuenheimer Feld 430, Heidelberg 69120, Germany.
| | - Nathalie Jäger
- Hopp Children's Cancer Center Heidelberg (KiTZ), German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, Heidelberg 69120 , the Netherlands.
| | - Olaf Witt
- Hopp Children's Cancer Center Heidelberg (KiTZ), German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, Heidelberg 69120 , the Netherlands; Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), and German Cancer Consortium (DKTK), Im Neuenheimer Feld 430, Heidelberg 69120, Germany; Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital,National Center for Tumor Diseases (NCT) Network, Heidelberg, Germany.
| | | | - Miranda P Dierselhuis
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Godelieve A M Tytgat
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Marc H W Wijnen
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Max M van Noesel
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands; Division Imaging & Cancer, University Medical Center Utrecht, Heidelberglaan 100, Utrecht 3584 CX, the Netherlands.
| | - Ronald R de Krijger
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands; Department of Pathology, University Medical Center Utrecht, Heidelberglaan 100, Utrecht 3584 CX, the Netherlands.
| | - Selma Eising
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Jan Koster
- Department of Oncogenomics, Cancer Center Amsterdam, Amsterdam UMC, the Netherlands.
| | - Emmy M Dolman
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Jan J Molenaar
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands; Department of Pharmaceutical Sciences, Utrecht University, Heidelberglaan 100, Utrecht 3584 CX, the Netherlands.
| |
Collapse
|
2
|
Berko ER, Naranjo A, Daniels AA, McNulty SN, Krytska K, Druley T, Zelley K, Koneru B, Chen L, Polkosnik G, Irwin MS, Bagatell R, Maris JM, Reynolds CP, DuBois SG, Park JR, Mossé YP. Frequency and Clinical Significance of Clonal and Subclonal Driver Mutations in High-Risk Neuroblastoma at Diagnosis: A Children's Oncology Group Study. J Clin Oncol 2025:JCO2402407. [PMID: 40036726 DOI: 10.1200/jco-24-02407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/20/2024] [Accepted: 02/03/2025] [Indexed: 03/06/2025] Open
Abstract
PURPOSE Relapsed high-risk neuroblastomas (NBLs) are enriched for targetable mutations in ALK and RAS-MAPK pathways, yet the prognostic effect of these aberrations and relevance of subclonal mutations at diagnosis remain undefined. We describe the spectrum and clinical significance of clonal and subclonal pathogenic alterations in high-risk NBL. METHODS We developed a focused high-risk NBL sequencing panel including ALK, NRAS, KRAS, HRAS, BRAF, PTPN11, TP53, and ATRX genes for ultra-deep sequencing and applied this assay to 242 pretherapy tumors from patients enrolled on the phase III trial Children's Oncology Group ANBL0532. We assessed the effect of clonal and subclonal mutations on event-free survival (EFS) and overall survival (OS). RESULTS ALK-activating mutations occurred in 21.5% of tumors (n = 52, 30 clonal, 22 subclonal), and 3.3% (n = 8) showed ALK amplification. EFS and OS for patients with any ALK-aberrant tumor were inferior to patients with wild-type (WT) ALK tumors (5-year OS 37.7% v 66.3%; hazard ratio [HR], 1.992; P = .0007). EFS and OS for patients with tumors harboring activating ALK mutations ≥5% variant allele frequency (VAF) were inferior to ALK WT (5-year OS 37.7% v 66.3%; HR, 1.966; P = .0041). The 5-year EFS and OS for patients with ALK-amplified tumors were 25.0%. RAS pathway mutations occurred in 7.9% of tumors (n = 19; four clonal, 15 subclonal), with EFS and OS for those with VAF ≥5% inferior to RAS-WT patients (5-year OS 19.1% v 60.0%; HR, 3.021; P = .0168). CONCLUSION Ultra-deep sequencing of high-risk NBLs demonstrates that oncogenic aberrations are more prevalent at diagnosis than previously recognized. ALK and RAS pathway aberrations confer inferior outcomes in patients treated with contemporary therapy, emphasizing the need for novel therapeutic approaches.
Collapse
Affiliation(s)
- Esther R Berko
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA
- Division of Hematology-Oncology, Schneider Children's Medical Center of Israel, Petach Tikvah, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Arlene Naranjo
- Department of Biostatistics, Children's Oncology Group Statistics and Data Center, University of Florida, Gainesville, FL
| | - Alexander A Daniels
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA
| | | | - Kateryna Krytska
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA
| | | | - Kirstin Zelley
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Balakrishna Koneru
- Department of Pediatrics and Cancer Center, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| | - Lulu Chen
- Department of Biostatistics, Children's Oncology Group Statistics and Data Center, University of Florida, Gainesville, FL
| | - Grace Polkosnik
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Meredith S Irwin
- Department of Pediatrics, Hospital for Sick Children and University of Toronto, Toronto, Canada
| | - Rochelle Bagatell
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA
| | - John M Maris
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - C Patrick Reynolds
- Department of Pediatrics and Cancer Center, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| | - Steven G DuBois
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Julie R Park
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN
| | - Yael P Mossé
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
3
|
Bruinsma RS, Lekkerkerker CWM, Fiocco M, Dierselhuis MP, Langenberg KPS, Tytgat GAM, van Noesel MM, Wijnen MHWA, van der Steeg AFW, de Krijger RR. Prognostic Value of Molecular Aberrations in Low- or Intermediate-Risk Neuroblastomas: A Systematic Review. Cancers (Basel) 2024; 17:13. [PMID: 39796644 PMCID: PMC11718975 DOI: 10.3390/cancers17010013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND The 5-year prognosis of non-high-risk neuroblastomas is generally good (>90%). However, a proportion of patients show progression and succumb to their disease. We aimed to identify molecular aberrations (not incorporated in the current risk stratification) associated with overall survival (OS) and/or event-free survival (EFS) in patients diagnosed with non-high-risk neuroblastoma. METHODS We conducted a systematic search in PubMed, Embase, Cochrane and Google Scholar. Two reviewers independently and blindly screened titles/abstracts, references of protocols/reviews and full texts. Risk of bias was assessed using a customized Quality in Prognostic Studies tool. Applicability was assessed using a tool designed by the researchers. GRADE criteria were used to determine quality of evidence. RESULTS Sixteen studies (4718 patients) were included. A segmental chromosomal aberration (SCA) profile was associated with lower survival. 1p loss of heterozygosity (LOH) and 17q gain were associated with lower OS and EFS. 1p deletion and 2p gain were associated with lower OS, but this was not the same for EFS. 3p deletion was not associated with worse outcome. Quality of evidence was downgraded because of imprecision and publication bias and upgraded because of moderate/large effect, resulting in a moderate quality of evidence. CONCLUSION The association of 1p LOH, 1p deletion, 2p gain and 17q gain with OS and EFS suggests that these SCAs may be added to the risk stratification to identify non-high-risk neuroblastomas with worse prognosis.
Collapse
Affiliation(s)
- Rixt S. Bruinsma
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
| | | | - Marta Fiocco
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Mathematical Institute, Leiden University, 2333 CC Leiden, The Netherlands
| | | | | | | | - Max M. van Noesel
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
- Division Imaging & Cancer, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | | | | | - Ronald R. de Krijger
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
- Department of Pathology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
4
|
Wei Z, Gong B, Li X, Chen C, Zhao Q. Event-free survival in neuroblastoma with MYCN amplification and deletion of 1p or 11q may be associated with altered immune status. BMC Cancer 2024; 24:1279. [PMID: 39407175 PMCID: PMC11481459 DOI: 10.1186/s12885-024-13044-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 10/08/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Neuroblastoma exhibits substantial heterogeneity, which is intricately linked to various genetic alterations. We aimed to explore immune status in the peripheral blood and prognosis of patients with neuroblastoma with different genetic characteristics. METHODS We enrolled 31 patients with neuroblastoma and collected samples to detect three genetic characteristics. Peripheral blood samples were tested for immune cells and cytokines by fluorescent microspheres conjugated with antibodies and flow cytometry. Event-free survival (EFS) was analyzed using the Kaplan‒Meier method. RESULTS Twenty-two patients had genetic aberrations, including MYCN amplification in 6 patients, chromosome 1p deletion in 9 patients, and chromosome 11q deletion in 14 patients. Two genetic alterations were present in seven patients. The EFS was worse in patients with MYCN amplification or 1p deletion than in the corresponding group, whereas 11q deletion was a prognostic factor only in patients with unamplified MYCN. Changes in immune status revealed a decrease in the proportion of T cells in blood, and an increase in regulatory T cells and immunosuppression-related cytokines such as interleukin (IL)-10. The EFS of the IL-10 high-level group was lower than that of the low-level group. Patients with concomitant genetic alterations and a high level of IL-10 had worse EFS than other patients. CONCLUSIONS Patients with neuroblastoma characterized by these genetic characteristics often have suppressed T cell response and an overabundance of immunosuppressive cells and cytokines in the peripheral blood. This imbalance is significantly associated with poor EFS. Moreover, if these patients show an elevated levels of immunosuppressive cytokines such as IL-10, the prognosis will be worse.
Collapse
Affiliation(s)
- Zixuan Wei
- Department of Pediatric Oncology, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, West Huan-Hu Rd, Ti Yuan Bei, Hexi District, 300060, Tianjin, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Baocheng Gong
- Department of Pediatric Oncology, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, West Huan-Hu Rd, Ti Yuan Bei, Hexi District, 300060, Tianjin, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Xin Li
- Tianjin Cancer Hospital Airport Hospital, Tianjin, China
| | - Chong Chen
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- National Human Genetic Resources Sharing Service Platform, Tianjin, China
| | - Qiang Zhao
- Department of Pediatric Oncology, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, West Huan-Hu Rd, Ti Yuan Bei, Hexi District, 300060, Tianjin, Tianjin, China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
- Tianjin's Clinical Research Center for Cancer, Tianjin, China.
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.
| |
Collapse
|
5
|
Bender HG, Irwin MS, Hogarty MD, Castleberry R, Maris JM, Kao PC, Zhang FF, Naranjo A, Cohn SL, London WB. Survival of Patients With Neuroblastoma After Assignment to Reduced Therapy Because of the 12- to 18-Month Change in Age Cutoff in Children's Oncology Group Risk Stratification. J Clin Oncol 2023; 41:3149-3159. [PMID: 37098238 PMCID: PMC10256433 DOI: 10.1200/jco.22.01946] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 12/01/2022] [Accepted: 02/23/2023] [Indexed: 04/27/2023] Open
Abstract
PURPOSE In 2006, Children's Oncology Group (COG) reclassified subgroups of toddlers diagnosed with neuroblastoma from high-risk to intermediate-risk, when the age cutoff for high-risk assignment was raised from 365 days (12 months) to 547 days (18 months). The primary aim of this retrospective study was to determine if excellent outcome was maintained after assigned reduction of therapy. PATIENTS AND METHODS Children <3 years old at diagnosis, enrolled on a COG biology study from 1990 to 2018, were eligible (n = 9,189). Assigned therapy was reduced for two cohorts of interest on the basis of the age cutoff change: 365-546 days old with International Neuroblastoma Staging System (INSS) stage 4, MYCN not amplified (MYCN-NA), favorable International Neuroblastoma Pathology Classification (INPC), hyperdiploid tumors (12-18mo/Stage4/FavBiology), and 365-546 days old with INSS stage 3, MYCN-NA, and unfavorable INPC tumors (12-18mo/Stage3/MYCN-NA/Unfav). Log-rank tests compared event-free survival (EFS) and overall survival (OS) curves. RESULTS For 12-18mo/Stage4/FavBiology, 5-year EFS/OS (± SE) before (≤2006; n = 40) versus after (>2006; n = 55) assigned reduction in therapy was similar: 89% ± 5.1%/89% ± 5.1% versus 87% ± 4.6%/94% ± 3.2% (P = .7; P = .4, respectively). For 12-18mo/Stage3/MYCN-NA/Unfav, the 5-year EFS and OS were both 100%, before (n = 6) and after (n = 4) 2006. The 12-18mo/Stage4/FavBiology plus 12-18mo/Stage3/MYCN-NA/Unfav classified as high-risk ≤2006 had an EFS/OS of 91% ± 4.4%/91% ± 4.5% versus 38% ± 1.3%/43% ± 1.3% for all other high-risk patients <3 years old (P < .0001; P < .0001, respectively). The 12-18mo/Stage4/FavBiology plus 12-18mo/Stage3/MYCN-NA/Unfav classified as intermediate-risk >2006 had an EFS/OS of 88% ± 4.3%/95% ± 2.9% versus 88% ± 0.9%/95% ± 0.6% for all other intermediate-risk patients <3 years old (P = .87; P = .85, respectively). CONCLUSION Excellent outcome was maintained among subsets of toddlers with neuroblastoma assigned to reduced treatment after reclassification of risk group from high to intermediate on the basis of new age cutoffs. Importantly, as documented in prior trials, intermediate-risk therapy is not associated with the degree of acute toxicity and late effects commonly observed with high-risk regimens.
Collapse
Affiliation(s)
- Hannah G. Bender
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Meredith S. Irwin
- Department of Pediatrics, The Hospital for Sick Children, University of Toronto, ON, Canada
| | - Michael D. Hogarty
- Department of Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | | | - John M. Maris
- Department of Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Pei-Chi Kao
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Fan F. Zhang
- Department of Biostatistics, Children's Oncology Group Statistics and Data Center, University of Florida, Gainesville, FL
| | - Arlene Naranjo
- Department of Biostatistics, Children's Oncology Group Statistics and Data Center, University of Florida, Gainesville, FL
| | - Susan L. Cohn
- Department of Pediatrics and Comer Children's Hospital, University of Chicago, Chicago, IL
| | - Wendy B. London
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| |
Collapse
|
6
|
Pinto N, Naranjo A, Ding X, Zhang FF, Hibbitts E, Kennedy R, Tibbetts R, Wong-Michalak S, Craig DW, Manojlovic Z, Hogarty MD, Kreissman S, Bagatell R, Irwin MS, Park JR, Asgharzadeh S. Impact of Genomic and Clinical Factors on Outcome of Children ≥18 Months of Age with Stage 3 Neuroblastoma with Unfavorable Histology and without MYCN Amplification: A Children's Oncology Group (COG) Report. Clin Cancer Res 2023; 29:1546-1556. [PMID: 36749880 PMCID: PMC10106446 DOI: 10.1158/1078-0432.ccr-22-3032] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/13/2022] [Accepted: 02/06/2023] [Indexed: 02/09/2023]
Abstract
PURPOSE Patients ≥18 months of age with International Neuroblastoma Staging System (INSS) stage 3 unfavorable histology (UH), MYCN-nonamplified (MYCN-NA) tumors have favorable survival rates compared with other high-risk neuroblastoma populations. The impact of select clinical and biological factors on overall survival (OS) and event-free survival (EFS) were evaluated. EXPERIMENTAL DESIGN Patients enrolled on Children's Oncology Group (COG) A3973 (n = 34), ANBL0532 (n = 27), and/or biology protocol ANBL00B1 (n = 72) were analyzed. Tumors with available DNA (n = 65) and RNA (n = 42) were subjected to whole-exome sequencing (WES) and RNA sequencing. WES analyses and gene expression profiling were evaluated for their impact on survival. Multivariate analyses of EFS/OS using significant factors from univariate analyses were performed. RESULTS 5-year EFS/OS for patients treated with high-risk therapy on A3973 and ANBL0532 were 73.0% ± 8.1%/87.9% ± 5.9% and 61.4% ± 10.2%/73.0% ± 9.2%, respectively (P = 0.1286 and P = 0.2180). In the A3973/ANBL0532 cohort, patients with less than partial response (PR; n = 5) at end-induction had poor outcomes (5-year EFS/OS: 0%/20.0% ± 17.9%. Univariate analyses of WES data revealed that subjects whose tumors had chromosome 1p or 11q loss/LOH and chromosome 5 or 9 segmental chromosomal aberrations had inferior EFS compared with those with tumors without these aberrations. Multivariate analysis revealed that 11q loss/LOH was an independent predictor of inferior OS [HR, 3.116 (95% confidence interval, 1.034-9.389), P = 0.0435]. CONCLUSIONS Patients ≥18 months of age at diagnosis who had tumors with UH and MYCN-NA INSS stage 3 neuroblastoma assigned to high-risk therapy had an 81.6% ± 5.3% 5-year OS. Less than PR to induction therapy and chromosome 11q loss/LOH are independent predictors of inferior outcome and identify patients who should be eligible for future high-risk clinical trials.
Collapse
Affiliation(s)
- Navin Pinto
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Arlene Naranjo
- Children’s Oncology Group Data and Statistics Center, Department of Biostatistics, University of Florida, Gainesville, Florida, USA
| | - Xiangming Ding
- Department of Pediatrics, University of Southern California, Los Angeles, California, USA
| | - Fan F. Zhang
- Children’s Oncology Group Data and Statistics Center, Monrovia, California, USA
| | - Emily Hibbitts
- Children’s Oncology Group Data and Statistics Center, Department of Biostatistics, University of Florida, Gainesville, Florida, USA
| | - Rebekah Kennedy
- Department of Pediatrics, University of Southern California, Los Angeles, California, USA
| | - Rachelle Tibbetts
- Department of Pediatrics, University of Southern California, Los Angeles, California, USA
| | - Shannon Wong-Michalak
- Department of Pediatrics, University of Southern California, Los Angeles, California, USA
| | - David W Craig
- Department of Pediatrics, University of Southern California, Los Angeles, California, USA
| | - Zarko Manojlovic
- Department of Pediatrics, University of Southern California, Los Angeles, California, USA
| | - Michael D. Hogarty
- Department of Pediatrics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Susan Kreissman
- Department of Pediatrics, Duke University, Durham, North Carolina, USA
| | - Rochelle Bagatell
- Department of Pediatrics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Meredith S. Irwin
- Department of Pediatrics, University of Toronto, Toronto, Ontario, CANADA
| | - Julie R. Park
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Shahab Asgharzadeh
- Department of Pediatrics, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
7
|
Wieczorek A, Szewczyk K, Klekawka T, Stefanowicz J, Ussowicz M, Drabik G, Pawinska-Wasikowska K, Balwierz W. Segmental chromosomal aberrations as the poor prognostic factor in children over 18 months with stage 3 neuroblastoma without MYCN amplification. Front Oncol 2023; 13:1134772. [PMID: 36865795 PMCID: PMC9972431 DOI: 10.3389/fonc.2023.1134772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 01/30/2023] [Indexed: 02/16/2023] Open
Abstract
Introduction Patients with stage 3 neuroblastoma (NBL) according to International Neuroblastoma Staging System (INSS) without MYCN amplification represent a heterogenous group with respect to disease presentation and prognosis. Methods Retrospective analysis of 40 stage 3 patients with NBL without MYCN amplification was performed. The prognostic value of age at diagnosis (under 18 vs over 18 months), International Neuroblastoma Pathology Classification (INPC) diagnostic category and presence of segmental or numerical chromosomes aberrations were evaluated, as well as biochemical markers. Array comparative genomic hybridization (aCGH) for analyzing copy number variations and Sanger sequencing for ALK point mutations were done. Results In 12 patients (two patients under 18 months), segmental chromosomal aberrations (SCA) were found and numerical chromosomal aberrations (NCA) were found in 16 patients (14 patients under 18 months). In children over 18 months SCA were more common (p=0.0001). Unfavorable pathology was significantly correlated with SCA genomic profile (p=0.04) and age over 18 months (p=0.008). No therapy failures occurred in children with NCA profile over or under 18 months or in children under 18 months, irrespective of pathology and CGH results. Three treatment failures occurred in the SCA group, in one patient CGH profile was not available. For the whole group at 3, 5 and 10-year OS and DFS were 0.95 (95% CI 0.81-0.99), 0.91 (95% CI 0.77-0.97) and 0.91 (95% CI 0.77-0.97), and 0.95 (95% CI 0.90-0.99), 0.92 (95% CI 0.85-0.98) and 0.86 (95% CI 0.78-0.97), respectively. DFS was significantly lower in the SCA group than in the NCA group (3-years, 5-years, and 10-years DFS 0.92 (95% CI 0.53-0.95), 0.80 (95% CI 0.40-0.95) and 0.60 (95% CI 0.16-0.87) vs 1.0, 1.0 and 1.0, respectively, p=0.005). Conclusions The risk of treatment failure was higher in patients with SCA profile, but only in patients over 18 months. All relapses occurred in children having obtained the complete remission, with no previous radiotherapy. In patients over 18 months, SCA profile should be taken into consideration for therapy stratification as it increases the risk of relapse and this group may require more intensive treatment.
Collapse
Affiliation(s)
- Aleksandra Wieczorek
- Department of Pediatric Oncology and Hematology, Medical College, Jagiellonian University, Krakow, Poland,Department of Pediatric Oncology and Hematology, University Children’s Hospital of Krakow, Krakow, Poland,*Correspondence: Aleksandra Wieczorek, ; Katarzyna Pawinska-Wasikowska,
| | - Katarzyna Szewczyk
- Department of Medical Genetics, Institute of Pediatrics, Medical College, Jagiellonian University, Krakow, Poland
| | - Tomasz Klekawka
- Department of Pediatric Oncology and Hematology, University Children’s Hospital of Krakow, Krakow, Poland
| | - Joanna Stefanowicz
- Department of Pediatrics, Pediatric Hematology and Oncology, Medical University of Gdansk, Gdansk, Poland
| | - Marek Ussowicz
- Department of Pediatric Bone Marrow Transplantation, Oncology, and Hematology, Wroclaw Medical University, Wroclaw, Poland
| | - Grazyna Drabik
- Department of Pathology, University Children’s Hospital of Krakow, Krakow, Poland
| | - Katarzyna Pawinska-Wasikowska
- Department of Pediatric Oncology and Hematology, Medical College, Jagiellonian University, Krakow, Poland,Department of Pediatric Oncology and Hematology, University Children’s Hospital of Krakow, Krakow, Poland,*Correspondence: Aleksandra Wieczorek, ; Katarzyna Pawinska-Wasikowska,
| | - Walentyna Balwierz
- Department of Pediatric Oncology and Hematology, Medical College, Jagiellonian University, Krakow, Poland,Department of Pediatric Oncology and Hematology, University Children’s Hospital of Krakow, Krakow, Poland
| |
Collapse
|
8
|
Du H, Cai W. Opsoclonus-myoclonus syndrome associated with neuroblastoma: Insights into antitumor immunity. Pediatr Blood Cancer 2022; 69:e29949. [PMID: 36094353 DOI: 10.1002/pbc.29949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 07/08/2022] [Accepted: 08/04/2022] [Indexed: 11/08/2022]
Abstract
Opsoclonus-myoclonus syndrome (OMS) is a rare neurological disorder. Half of these cases occur in children with neuroblastoma. Neuroblastoma patients with OMS usually have better oncological outcomes than those without OMS even after stratification by tumor stage and age, indicating that factors mediating OMS may also inhibit tumor cell proliferation. Although the mechanisms underlying OMS remain undefined, the cytokines and lymphocytes alterations in the cerebrospinal fluid support the concept that it is a pattern of neuroinflammation due to an autoimmune effect. The presence of lymphoid follicles consisting of follicular dendritic cells, CD20+ B lymphocytes, CD3+ T lymphocytes, and CD68+ macrophages in the tumor microenvironment in OMS-associated neuroblastoma support the autoimmune nature of this disorder. This review focuses on the clinical and genetic features of OMS-associated neuroblastoma, and we update readers on immune features of neuroblastoma with or without OMS to gain insights into antitumor immunity as it relates to tumor biology and prognosis.
Collapse
Affiliation(s)
- Hongmei Du
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Weisong Cai
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
9
|
Ognibene M, Cangelosi D, Sorrentino S, Zanardi S, Zara F, Pezzolo A, Parodi S. E2F3 gene expression is a potential negative prognostic marker for localised and MYCN not-amplified neuroblastoma: Results of in silico analysis of 786 samples. Pediatr Blood Cancer 2022; 69:e29800. [PMID: 35652628 DOI: 10.1002/pbc.29800] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 04/13/2022] [Accepted: 05/09/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Neuroblastoma (NB) is an enigmatic childhood malignancy characterised by a wide range of clinical behaviour. Many potential oncogenes for NB have recently been identified. Among them, E2 transcription factor 3 (E2F3) expression was associated with a poor survival in 134 stage 4S patients, but evidence for other stage groups remains poorly investigated. METHODS We have analysed the expression of E2F3 gene from a database of 786 NB samples. Overall and event-free survivals (EFS) were assessed by the Kaplan-Meier method, splitting the data on the median and tertile expression values. The Cox model was applied to control for the confounding by stage, age and MYCN amplification. Validation was performed by an in silico analysis of an independent cohort of 283 NB patients. Furthermore, an immunofluorescence analysis on 48 formalin-fixed, paraffin-embedded NB specimens was also performed. RESULTS E2F3 overexpression was associated with a poor survival (EFS = 84%, 95% CI: 79%-95%, for low expression levels; EFS = 62%, 95% CI: 56%-68% for middle levels; EFS = 30%, 95% CI: 24%-36%, for high levels, p < .001). This association was confirmed in multivariable analysis and was more evident in patients with MYCN not-amplified and localised stages. Immunofluorescence results and the validation on an independent cohort of NB primary samples confirmed these findings. CONCLUSIONS E2F3 is a new potential prognostic marker in NB with favourable characteristics at diagnosis. Further studies are needed to elucidate the potential role of E2F3 in NB oncogenesis and progression, in order to identify new targets for therapeutic interventions.
Collapse
Affiliation(s)
- Marzia Ognibene
- U.O.C. Genetica Medica, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Davide Cangelosi
- Unità di Bioinformatica Clinica, Direzione Scientifica, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Stefania Sorrentino
- U.O.C. Divisione di Oncologia, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Sabrina Zanardi
- U.O.S.I.D. Epidemiologia e Biostatistica, Direzione Scientifica, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Federico Zara
- U.O.C. Genetica Medica, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | | | - Stefano Parodi
- Direzione Scientifica, IRCCS Istituto Giannina Gaslini, Genova, Italy
| |
Collapse
|
10
|
Kuick CH, Tan JY, Jasmine D, Sumanty T, Ng AYJ, Venkatesh B, Chen H, Loh E, Jain S, Seow WY, Ng EHQ, Lian DWQ, Soh SY, Chang KTE, Chen ZX, Loh AHP. Mutations of 1p genes do not consistently abrogate tumor suppressor functions in 1p-intact neuroblastoma. BMC Cancer 2022; 22:717. [PMID: 35768791 PMCID: PMC9245282 DOI: 10.1186/s12885-022-09800-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 06/21/2022] [Indexed: 11/26/2022] Open
Abstract
Background Deletion of 1p is associated with poor prognosis in neuroblastoma, however selected 1p-intact patients still experience poor outcomes. Since mutations of 1p genes may mimic the deleterious effects of chromosomal loss, we studied the incidence, spectrum and effects of mutational variants in 1p-intact neuroblastoma. Methods We characterized the 1p status of 325 neuroblastoma patients, and correlated the mutational status of 1p tumor suppressors and neuroblastoma candidate genes with survival outcomes among 100 1p-intact cases, then performed functional validation of selected novel variants of 1p36 genes identified from our patient cohort. Results Among patients with adverse disease characteristics, those who additionally had 1p deletion had significantly worse overall survival. Among 100 tumor-normal pairs sequenced, somatic mutations of 1p tumor suppressors KIF1Bβ and CHD5 were most frequent (2%) after ALK and ATRX (8%), and BARD1 (3%). Mutations of neuroblastoma candidate genes were associated with other synchronous mutations and concurrent 11q deletion (P = 0.045). In total, 24 of 38 variants identified were novel and predicted to be deleterious or pathogenic. Functional validation identified novel KIF1Bβ I1355M variant as a gain-of-function mutation with increased expression and tumor suppressive activity, correlating with indolent clinical behavior; another novel variant CHD5 E43Q was a loss-of-function mutation with decreased expression and increased long-term cell viability, corresponding with aggressive disease characteristics. Conclusions Our study showed that chromosome 1 gene mutations occurred frequently in 1p-intact neuroblastoma, but may not consistently abrogate the function of bonafide 1p tumor suppressors. These findings may augment the evolving model of compounding contributions of 1p gene aberrations toward tumor suppressor inactivation in neuroblastoma. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09800-0.
Collapse
Affiliation(s)
- Chik Hong Kuick
- Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore, 229899, Singapore
| | - Jia Ying Tan
- Neurodevelopment and Cancer Laboratory, NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Deborah Jasmine
- Neurodevelopment and Cancer Laboratory, NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Tohari Sumanty
- Comparative and Medical Genomics Laboratory, Institute of Molecular and Cell Biology, A*STAR, Singapore, 138673, Singapore
| | - Alvin Y J Ng
- Comparative and Medical Genomics Laboratory, Institute of Molecular and Cell Biology, A*STAR, Singapore, 138673, Singapore
| | - Byrrappa Venkatesh
- Comparative and Medical Genomics Laboratory, Institute of Molecular and Cell Biology, A*STAR, Singapore, 138673, Singapore
| | - Huiyi Chen
- Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore, 229899, Singapore
| | - Eva Loh
- Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore, 229899, Singapore
| | - Sudhanshi Jain
- Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore, 229899, Singapore
| | - Wan Yi Seow
- Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore, 229899, Singapore
| | - Eileen H Q Ng
- Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore, 229899, Singapore
| | - Derrick W Q Lian
- Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore, 229899, Singapore
| | - Shui Yen Soh
- VIVA-KKH Paediatric Brain and Solid Tumour Programme, Children's Blood and Cancer Centre, KK Women's and Children's Hospital, Singapore, 229899, Singapore.,Department of Paediatric Subspecialties Haematology Oncology Service, KK Women's and Children's Hospital, Singapore, 229899, Singapore.,Duke NUS Medical School, Singapore, 169857, Singapore
| | - Kenneth T E Chang
- Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore, 229899, Singapore.,VIVA-KKH Paediatric Brain and Solid Tumour Programme, Children's Blood and Cancer Centre, KK Women's and Children's Hospital, Singapore, 229899, Singapore.,Duke NUS Medical School, Singapore, 169857, Singapore
| | - Zhi Xiong Chen
- Neurodevelopment and Cancer Laboratory, NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore. .,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore. .,VIVA-KKH Paediatric Brain and Solid Tumour Programme, Children's Blood and Cancer Centre, KK Women's and Children's Hospital, Singapore, 229899, Singapore. .,National University Cancer Institute, Singapore, 119074, Singapore.
| | - Amos H P Loh
- VIVA-KKH Paediatric Brain and Solid Tumour Programme, Children's Blood and Cancer Centre, KK Women's and Children's Hospital, Singapore, 229899, Singapore. .,Duke NUS Medical School, Singapore, 169857, Singapore. .,Department of Paediatric Surgery, KK Women's and Children's Hospital, Singapore, 229899, Singapore.
| |
Collapse
|
11
|
Irwin MS, Naranjo A, Zhang FF, Cohn SL, London WB, Gastier-Foster JM, Ramirez NC, Pfau R, Reshmi S, Wagner E, Nuchtern J, Asgharzadeh S, Shimada H, Maris JM, Bagatell R, Park JR, Hogarty MD. Revised Neuroblastoma Risk Classification System: A Report From the Children's Oncology Group. J Clin Oncol 2021; 39:3229-3241. [PMID: 34319759 PMCID: PMC8500606 DOI: 10.1200/jco.21.00278] [Citation(s) in RCA: 241] [Impact Index Per Article: 60.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 06/10/2021] [Accepted: 06/30/2021] [Indexed: 01/25/2023] Open
Abstract
PURPOSE Treatment planning for children with neuroblastoma requires accurate assessment of prognosis. The most recent Children's Oncology Group (COG) risk classification system used tumor stage as defined by the International Neuroblastoma Staging System. Here, we validate a revised classifier using the International Neuroblastoma Risk Group Staging System (INRGSS) and incorporate segmental chromosome aberrations (SCA) as an additional genomic biomarker. METHODS Newly diagnosed patients enrolled on the COG neuroblastoma biology study ANBL00B1 between 2007 and 2017 with known age, International Neuroblastoma Staging System, and INRGSS stage were identified (N = 4,832). Tumor MYCN status, ploidy, SCA status (1p and 11q), and International Neuroblastoma Pathology Classification histology were determined centrally. Survival analyses were performed for combinations of prognostic factors used in COG risk classification according to the prior version 1, and to validate a revised algorithm (version 2). RESULTS Most patients with locoregional tumors had excellent outcomes except for those with image-defined risk factors (INRGSS L2) with MYCN amplification (5-year event-free survival and overall survival: 76.3% ± 5.8% and 79.9% ± 5.5%, respectively) or patients age ≥ 18 months with L2 MYCN nonamplified tumors with unfavorable International Neuroblastoma Pathology Classification histology (72.7% ± 5.4% and 82.4% ± 4.6%), which includes the majority of L2 patients with SCA. For patients with stage M (metastatic) and MS (metastatic, special) disease, genomic biomarkers affected risk group assignment for those < 12 months (MYCN) or 12-18 months (MYCN, histology, ploidy, and SCA) of age. In a retrospective analysis of patient outcome, the 5-year event-free survival and overall survival using COG version 1 were low-risk: 89.4% ± 1.1% and 97.9% ± 0.5%; intermediate-risk: 86.1% ± 1.3% and 94.9% ± 0.8%; high-risk: 50.8% ± 1.4% and 61.9% ± 1.3%; and using COG version 2 were low-risk: 90.7% ± 1.1% and 97.9% ± 0.5%; intermediate-risk: 85.1% ± 1.4% and 95.8% ± 0.8%; high-risk: 51.2% ± 1.4% and 62.5% ± 1.3%, respectively. CONCLUSION A revised 2021 COG neuroblastoma risk classifier (version 2) that uses the INRGSS and incorporates SCAs has been adopted to prospectively define COG clinical trial eligibility and treatment assignment.
Collapse
Affiliation(s)
- Meredith S. Irwin
- Department of Pediatrics, Hospital for Sick Children, Toronto, ON, Canada
| | - Arlene Naranjo
- Children's Oncology Group Statistics and Data Center, Department of Biostatistics, University of Florida, Gainesville, FL
| | - Fan F. Zhang
- Children's Oncology Group Statistics and Data Center, Monrovia, CA
| | - Susan L. Cohn
- Department of Pediatrics, The University of Chicago, Chicago, IL
| | - Wendy B. London
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Julie M. Gastier-Foster
- Institute for Genomic Medicine and Biopathology Center, Nationwide Children's Hospital, Columbus, OH
- Departments of Pathology and Pediatrics, Ohio State University, Columbus, OH
| | - Nilsa C. Ramirez
- Institute for Genomic Medicine and Biopathology Center, Nationwide Children's Hospital, Columbus, OH
- Departments of Pathology and Pediatrics, Ohio State University, Columbus, OH
| | - Ruthann Pfau
- Institute for Genomic Medicine and Biopathology Center, Nationwide Children's Hospital, Columbus, OH
- Departments of Pathology and Pediatrics, Ohio State University, Columbus, OH
| | - Shalini Reshmi
- Institute for Genomic Medicine and Biopathology Center, Nationwide Children's Hospital, Columbus, OH
- Departments of Pathology and Pediatrics, Ohio State University, Columbus, OH
| | - Elizabeth Wagner
- Institute for Genomic Medicine and Biopathology Center, Nationwide Children's Hospital, Columbus, OH
| | - Jed Nuchtern
- Division of Pediatric Surgery, Department of Surgery, Texas Children's Hospital, Baylor College of Medicine, Houston, TX
| | - Shahab Asgharzadeh
- Division of Hematology/Oncology, Children's Hospital of Los Angeles, Los Angeles, CA
| | - Hiroyuki Shimada
- Departments of Pathology and Pediatrics, Stanford University, Stanford, CA
| | - John M. Maris
- Department of Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Rochelle Bagatell
- Department of Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Julie R. Park
- Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA
| | - Michael D. Hogarty
- Department of Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
12
|
Why Is Aneuploidy Associated with Favorable Outcome in Neuroblastoma? Biomolecules 2021; 11:biom11081116. [PMID: 34439783 PMCID: PMC8391846 DOI: 10.3390/biom11081116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 11/17/2022] Open
Abstract
Neuroblastoma is a pediatric cancer, onset with localized as well as metastatic disease. Localized tumors usually show a high content of aneuploid cells. It is suggested that aneuploid cells with numerical copy number variation (CNV) are generated by chromosome instability (CIN). Patients with a localized tumor respond well to the therapy and show a good outcome. On the contrary, patients with a metastatic tumor have worse outcomes and the cells with structural CNV show high levels of CIN. It is proposed that a favorable outcome in patients with localized disease is associated to the grade of CIN.
Collapse
|
13
|
Long-Term Outcome and Role of Biology within Risk-Adapted Treatment Strategies: The Austrian Neuroblastoma Trial A-NB94. Cancers (Basel) 2021; 13:cancers13030572. [PMID: 33540616 PMCID: PMC7867286 DOI: 10.3390/cancers13030572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 01/08/2021] [Accepted: 01/28/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Neuroblastoma, the most common extracranial malignancy of childhood, shows a highly variable course of disease ranging from spontaneous regression or maturation into a benign tumor to an aggressive and intractable cancer in up to 60% of patients. To adapt treatment intensity, risk staging at diagnosis is of utmost importance. The A-NB94 trial was the first in Austria to stratify therapy intensity according to tumor staging, patient’s age, and MYCN amplification status, the latter being a biologic marker turning otherwise low-risk tumors into high-risk disease. Recent publications showed a prognostic impact of various genomic features including segmental chromosomal aberrations (SCAs). We retrospectively investigated the relevance of SCAs within this risk-adapted treatment strategy. The A-NB94 approach resulted in an excellent long-term survival for the majority of patients with acceptable long-term morbidity. An age- and stage-dependent frequency of SCAs was confirmed and SCAs should always be considered in future treatment decision making processes. Abstract We evaluated long-term outcome and genomic profiles in the Austrian Neuroblastoma Trial A-NB94 which applied a risk-adapted strategy of treatment (RAST) using stage, age and MYCN amplification (MNA) status for stratification. RAST ranged from surgery only to intensity-adjusted chemotherapy, single or multiple courses of high-dose chemotherapy (HDT) followed by autologous stem cell rescue depending on response to induction chemotherapy, and irradiation to the primary tumor site. Segmental chromosomal alterations (SCAs) were investigated retrospectively using multi- and pan-genomic techniques. The A-NB94 trial enrolled 163 patients. Patients with localized disease had an excellent ten-year (10y) event free survival (EFS) and overall survival (OS) of 99 ± 1% and 93 ± 2% whilst it was 80 ± 13% and 90 ± 9% for infants with stage 4S and for infants with stage 4 non-MNA disease both 83 ± 15%. Stage 4 patients either >12 months or ≤12 months but with MNA had a 10y-EFS and OS of 45 ± 8% and 47 ± 8%, respectively. SCAs were present in increasing frequencies according to stage and age: in 29% of localized tumors but in 92% of stage 4 tumors (p < 0.001), and in 39% of patients ≤ 12 months but in 63% of patients > 12 months (p < 0.001). RAST successfully reduced chemotherapy exposure in low- and intermediate-risk patients with excellent long-term results while the outcome of high-risk disease met contemporary trials.
Collapse
|
14
|
Ambros IM, Tonini GP, Pötschger U, Gross N, Mosseri V, Beiske K, Berbegall AP, Bénard J, Bown N, Caron H, Combaret V, Couturier J, Defferrari R, Delattre O, Jeison M, Kogner P, Lunec J, Marques B, Martinsson T, Mazzocco K, Noguera R, Schleiermacher G, Valent A, Van Roy N, Villamon E, Janousek D, Pribill I, Glogova E, Attiyeh EF, Hogarty MD, Monclair TF, Holmes K, Valteau-Couanet D, Castel V, Tweddle DA, Park JR, Cohn S, Ladenstein R, Beck-Popovic M, De Bernardi B, Michon J, Pearson ADJ, Ambros PF. Age Dependency of the Prognostic Impact of Tumor Genomics in Localized Resectable MYCN-Nonamplified Neuroblastomas. Report From the SIOPEN Biology Group on the LNESG Trials and a COG Validation Group. J Clin Oncol 2020; 38:3685-3697. [PMID: 32903140 PMCID: PMC7605396 DOI: 10.1200/jco.18.02132] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/07/2020] [Indexed: 12/15/2022] Open
Abstract
PURPOSE For localized, resectable neuroblastoma without MYCN amplification, surgery only is recommended even if incomplete. However, it is not known whether the genomic background of these tumors may influence outcome. PATIENTS AND METHODS Diagnostic samples were obtained from 317 tumors, International Neuroblastoma Staging System stages 1/2A/2B, from 3 cohorts: Localized Neuroblastoma European Study Group I/II and Children's Oncology Group. Genomic data were analyzed using multi- and pangenomic techniques and fluorescence in-situ hybridization in 2 age groups (cutoff age, 18 months) and were quality controlled by the International Society of Pediatric Oncology European Neuroblastoma (SIOPEN) Biology Group. RESULTS Patients with stage 1 tumors had an excellent outcome (5-year event-free survival [EFS] ± standard deviation [SD], 95% ± 2%; 5-year overall survival [OS], 99% ± 1%). In contrast, patients with stage 2 tumors had a reduced EFS in both age groups (5-year EFS ± SD, 84% ± 3% in patients < 18 months of age and 75% ± 7% in patients ≥ 18 months of age). However, OS was significantly decreased only in the latter group (5-year OS ± SD in < 18months and ≥ 18months, 96% ± 2% and 81% ± 7%, respectively; P = .001). In < 18months, relapses occurred independent of segmental chromosome aberrations (SCAs); only 1p loss decreased EFS (5-year EFS ± SD in patients 1p loss and no 1p loss, 62% ± 13% and 87% ± 3%, respectively; P = .019) but not OS (5-year OS ± SD, 92% ± 8% and 97% ± 2%, respectively). In patients ≥ 18 months, only SCAs led to relapse and death, with 11q loss as the strongest marker (11q loss and no 11q loss: 5-year EFS ± SD, 48% ± 16% and 85% ± 7%, P = .033; 5-year OS ± SD, 46% ± 22% and 92% ± 6%, P = .038). CONCLUSION Genomic aberrations of resectable non-MYCN-amplified stage 2 neuroblastomas have a distinct age-dependent prognostic impact. Chromosome 1p loss is a risk factor for relapse but not for diminished OS in patients < 18 months, SCAs (especially 11q loss) are risk factors for reduced EFS and OS in those > 18months. In older patients with SCA, a randomized trial of postoperative chemotherapy compared with observation alone may be indicated.
Collapse
Affiliation(s)
- Inge M. Ambros
- Children’s Cancer Research Institute, St Anna Kinderkrebsforschung, Vienna, Austria
| | - Gian-Paolo Tonini
- Paediatric Research Institute, Fondazione Città della Speranza, Neuroblastoma Laboratory, Padua, Italy
| | - Ulrike Pötschger
- Children’s Cancer Research Institute, St Anna Kinderkrebsforschung, Vienna, Austria
| | - Nicole Gross
- Pediatric Oncology Research, Department of Pediatrics, University Hospital, Lausanne, Switzerland
| | | | - Klaus Beiske
- Department of Pathology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Ana P. Berbegall
- Department of Pathology, Medical School, University of Valencia–Fundación de Investigación del Hospital Clínico Universitario de Valencia, Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
| | - Jean Bénard
- Département de Biologie et de Pathologie Médicales, Service de Pathologie Moléculaire, Institut Gustave Roussy, Villejuif, France
| | - Nick Bown
- Northern Genetics Service, Newcastle upon Tyne, United Kingdom
| | - Huib Caron
- Department of Pediatric Oncology, Emma Children's Hospital, Academic Medical Center, Amsterdam, the Netherlands
| | - Valérie Combaret
- Centre Léon Bérard, Laboratoire de Recherche Translationnelle, Lyon, France
| | - Jerome Couturier
- Unité de Génétique Somatique et Cytogénétique, Institut Curie, Paris, France
| | | | - Olivier Delattre
- INSERM U830, Laboratoire de Génétique et Biologie des Cancers, Paris, France
| | - Marta Jeison
- Ca-Cytogenetic Laboratory, Pediatric Hematology Oncology Department, Schneider Children's Medical Center of Israel, Petah Tikvah, Israel
| | - Per Kogner
- Childhood Cancer Research Unit, Karolinska Institutet, Astrid Lindgren Children's Hospital, Stockholm, Sweden
| | - John Lunec
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Barbara Marques
- Centro de Genética Humana, Instituto Nacional de Saude doutor Ricardo Jorge, Lisbon, Portugal
| | - Tommy Martinsson
- Department of Clinical Genetics, Institute of Biomedicine, University of Gothenburg, Sahlgrenska University Hospital, Göteborg, Sweden
| | - Katia Mazzocco
- Department of Pathology, Istituto G. Gaslini, Genoa, Italy
| | - Rosa Noguera
- Department of Pathology, Medical School, University of Valencia–Fundación de Investigación del Hospital Clínico Universitario de Valencia, Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
| | - Gudrun Schleiermacher
- INSERM U830, Laboratoire de Génétique et Biologie des Cancers, Paris, France
- Département de Pédiatrie, Institut Curie, Paris, France
| | - Alexander Valent
- Département de Biologie et de Pathologie Médicales, Service de Pathologie Moléculaire, Institut Gustave Roussy, Villejuif, France
| | - Nadine Van Roy
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Eva Villamon
- Department of Pathology, Medical School, University of Valencia–Fundación de Investigación del Hospital Clínico Universitario de Valencia, Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
| | - Dasa Janousek
- Children’s Cancer Research Institute, St Anna Kinderkrebsforschung, Vienna, Austria
| | - Ingrid Pribill
- Children’s Cancer Research Institute, St Anna Kinderkrebsforschung, Vienna, Austria
| | - Evgenia Glogova
- Children’s Cancer Research Institute, St Anna Kinderkrebsforschung, Vienna, Austria
| | - Edward F. Attiyeh
- Division of Oncology, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Michael D. Hogarty
- Division of Oncology, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Tom F. Monclair
- Section for Paediatric Surgery, Division of Surgery, Rikshospitalet University Hospital, Oslo, Norway
| | - Keith Holmes
- Department of Paediatric Surgery, St George's Hospital, London, UK
| | | | - Victoria Castel
- Unidad de Oncologia Pediatrica Hospital Universitario La Fe, Valencia, Spain
| | - Deborah A. Tweddle
- Wolfson Childhood Cancer Research Centre, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Julie R. Park
- Seattle Children’s Hospital and University of Washington School of Medicine, Seattle, WA
| | - Sue Cohn
- Department of Pediatrics, The University of Chicago, Chicago, IL
| | - Ruth Ladenstein
- Children’s Cancer Research Institute, St Anna Kinderkrebsforschung, Vienna, Austria
- Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Maja Beck-Popovic
- Pediatric Hematology Oncology Unit, University Hospital of Lausanne, Lausanne, Switzerland
| | - Bruno De Bernardi
- Department of Paediatric Haematology and Oncology, Giannina Gaslini Children's Hospital, Genova, Italy
| | - Jean Michon
- Département de Pédiatrie, Institut Curie, Paris, France
| | - Andrew D. J. Pearson
- Institute of Cancer Research, Royal Marsden Hospital, Sutton, Surrey, United Kingdom
| | - Peter F. Ambros
- Children’s Cancer Research Institute, St Anna Kinderkrebsforschung, Vienna, Austria
- Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
15
|
Sokol E, Desai AV, Applebaum MA, Valteau-Couanet D, Park JR, Pearson ADJ, Schleiermacher G, Irwin MS, Hogarty M, Naranjo A, Volchenboum S, Cohn SL, London WB. Reply to K. Beiske et al. J Clin Oncol 2020; 38:3720-3721. [PMID: 32931402 DOI: 10.1200/jco.20.02147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Elizabeth Sokol
- Elizabeth Sokol, MD, Department of Pediatrics and Lurie Children's Hospital, Northwestern University, Chicago, IL; Ami V. Desai, MD, MSCE, and Mark A. Applebaum, MD, Department of Pediatrics and Comer Children's Hospital, University of Chicago, Chicago, IL; Dominique Valteau-Couanet, MD, PhD, Institute Gustave Roussy, Villejuif, France; Julie R. Park, MD, Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA; Andrew D.J. Pearson, MD, Paediatric Drug Development, Children and Young People's Unit, Royal Marsden Hospital, London, UK; Gudrun Schleiermacher, MD, PhD, Department of Pediatric, Adolescents, and Young Adults Oncology and INSERM U830, Institut Curie, Paris, France; Meredith S. Irwin, MD, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada; Michael Hogarty, MD, Department of Pediatrics, University of Pennsylvania, Philadelphia, PA; Arlene Naranjo, PhD, Department of Biostatistics, Children's Oncology Group Statistics and Data Center, University of Florida, Gainesville, FL; Samuel Volchenboum, MD, PhD, and Susan L. Cohn, MD, Department of Pediatrics and Comer Children's Hospital, University of Chicago, Chicago, IL; and Wendy B. London, PhD, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Ami V Desai
- Elizabeth Sokol, MD, Department of Pediatrics and Lurie Children's Hospital, Northwestern University, Chicago, IL; Ami V. Desai, MD, MSCE, and Mark A. Applebaum, MD, Department of Pediatrics and Comer Children's Hospital, University of Chicago, Chicago, IL; Dominique Valteau-Couanet, MD, PhD, Institute Gustave Roussy, Villejuif, France; Julie R. Park, MD, Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA; Andrew D.J. Pearson, MD, Paediatric Drug Development, Children and Young People's Unit, Royal Marsden Hospital, London, UK; Gudrun Schleiermacher, MD, PhD, Department of Pediatric, Adolescents, and Young Adults Oncology and INSERM U830, Institut Curie, Paris, France; Meredith S. Irwin, MD, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada; Michael Hogarty, MD, Department of Pediatrics, University of Pennsylvania, Philadelphia, PA; Arlene Naranjo, PhD, Department of Biostatistics, Children's Oncology Group Statistics and Data Center, University of Florida, Gainesville, FL; Samuel Volchenboum, MD, PhD, and Susan L. Cohn, MD, Department of Pediatrics and Comer Children's Hospital, University of Chicago, Chicago, IL; and Wendy B. London, PhD, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Mark A Applebaum
- Elizabeth Sokol, MD, Department of Pediatrics and Lurie Children's Hospital, Northwestern University, Chicago, IL; Ami V. Desai, MD, MSCE, and Mark A. Applebaum, MD, Department of Pediatrics and Comer Children's Hospital, University of Chicago, Chicago, IL; Dominique Valteau-Couanet, MD, PhD, Institute Gustave Roussy, Villejuif, France; Julie R. Park, MD, Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA; Andrew D.J. Pearson, MD, Paediatric Drug Development, Children and Young People's Unit, Royal Marsden Hospital, London, UK; Gudrun Schleiermacher, MD, PhD, Department of Pediatric, Adolescents, and Young Adults Oncology and INSERM U830, Institut Curie, Paris, France; Meredith S. Irwin, MD, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada; Michael Hogarty, MD, Department of Pediatrics, University of Pennsylvania, Philadelphia, PA; Arlene Naranjo, PhD, Department of Biostatistics, Children's Oncology Group Statistics and Data Center, University of Florida, Gainesville, FL; Samuel Volchenboum, MD, PhD, and Susan L. Cohn, MD, Department of Pediatrics and Comer Children's Hospital, University of Chicago, Chicago, IL; and Wendy B. London, PhD, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Dominique Valteau-Couanet
- Elizabeth Sokol, MD, Department of Pediatrics and Lurie Children's Hospital, Northwestern University, Chicago, IL; Ami V. Desai, MD, MSCE, and Mark A. Applebaum, MD, Department of Pediatrics and Comer Children's Hospital, University of Chicago, Chicago, IL; Dominique Valteau-Couanet, MD, PhD, Institute Gustave Roussy, Villejuif, France; Julie R. Park, MD, Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA; Andrew D.J. Pearson, MD, Paediatric Drug Development, Children and Young People's Unit, Royal Marsden Hospital, London, UK; Gudrun Schleiermacher, MD, PhD, Department of Pediatric, Adolescents, and Young Adults Oncology and INSERM U830, Institut Curie, Paris, France; Meredith S. Irwin, MD, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada; Michael Hogarty, MD, Department of Pediatrics, University of Pennsylvania, Philadelphia, PA; Arlene Naranjo, PhD, Department of Biostatistics, Children's Oncology Group Statistics and Data Center, University of Florida, Gainesville, FL; Samuel Volchenboum, MD, PhD, and Susan L. Cohn, MD, Department of Pediatrics and Comer Children's Hospital, University of Chicago, Chicago, IL; and Wendy B. London, PhD, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Julie R Park
- Elizabeth Sokol, MD, Department of Pediatrics and Lurie Children's Hospital, Northwestern University, Chicago, IL; Ami V. Desai, MD, MSCE, and Mark A. Applebaum, MD, Department of Pediatrics and Comer Children's Hospital, University of Chicago, Chicago, IL; Dominique Valteau-Couanet, MD, PhD, Institute Gustave Roussy, Villejuif, France; Julie R. Park, MD, Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA; Andrew D.J. Pearson, MD, Paediatric Drug Development, Children and Young People's Unit, Royal Marsden Hospital, London, UK; Gudrun Schleiermacher, MD, PhD, Department of Pediatric, Adolescents, and Young Adults Oncology and INSERM U830, Institut Curie, Paris, France; Meredith S. Irwin, MD, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada; Michael Hogarty, MD, Department of Pediatrics, University of Pennsylvania, Philadelphia, PA; Arlene Naranjo, PhD, Department of Biostatistics, Children's Oncology Group Statistics and Data Center, University of Florida, Gainesville, FL; Samuel Volchenboum, MD, PhD, and Susan L. Cohn, MD, Department of Pediatrics and Comer Children's Hospital, University of Chicago, Chicago, IL; and Wendy B. London, PhD, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Andrew D J Pearson
- Elizabeth Sokol, MD, Department of Pediatrics and Lurie Children's Hospital, Northwestern University, Chicago, IL; Ami V. Desai, MD, MSCE, and Mark A. Applebaum, MD, Department of Pediatrics and Comer Children's Hospital, University of Chicago, Chicago, IL; Dominique Valteau-Couanet, MD, PhD, Institute Gustave Roussy, Villejuif, France; Julie R. Park, MD, Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA; Andrew D.J. Pearson, MD, Paediatric Drug Development, Children and Young People's Unit, Royal Marsden Hospital, London, UK; Gudrun Schleiermacher, MD, PhD, Department of Pediatric, Adolescents, and Young Adults Oncology and INSERM U830, Institut Curie, Paris, France; Meredith S. Irwin, MD, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada; Michael Hogarty, MD, Department of Pediatrics, University of Pennsylvania, Philadelphia, PA; Arlene Naranjo, PhD, Department of Biostatistics, Children's Oncology Group Statistics and Data Center, University of Florida, Gainesville, FL; Samuel Volchenboum, MD, PhD, and Susan L. Cohn, MD, Department of Pediatrics and Comer Children's Hospital, University of Chicago, Chicago, IL; and Wendy B. London, PhD, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Gudrun Schleiermacher
- Elizabeth Sokol, MD, Department of Pediatrics and Lurie Children's Hospital, Northwestern University, Chicago, IL; Ami V. Desai, MD, MSCE, and Mark A. Applebaum, MD, Department of Pediatrics and Comer Children's Hospital, University of Chicago, Chicago, IL; Dominique Valteau-Couanet, MD, PhD, Institute Gustave Roussy, Villejuif, France; Julie R. Park, MD, Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA; Andrew D.J. Pearson, MD, Paediatric Drug Development, Children and Young People's Unit, Royal Marsden Hospital, London, UK; Gudrun Schleiermacher, MD, PhD, Department of Pediatric, Adolescents, and Young Adults Oncology and INSERM U830, Institut Curie, Paris, France; Meredith S. Irwin, MD, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada; Michael Hogarty, MD, Department of Pediatrics, University of Pennsylvania, Philadelphia, PA; Arlene Naranjo, PhD, Department of Biostatistics, Children's Oncology Group Statistics and Data Center, University of Florida, Gainesville, FL; Samuel Volchenboum, MD, PhD, and Susan L. Cohn, MD, Department of Pediatrics and Comer Children's Hospital, University of Chicago, Chicago, IL; and Wendy B. London, PhD, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Meredith S Irwin
- Elizabeth Sokol, MD, Department of Pediatrics and Lurie Children's Hospital, Northwestern University, Chicago, IL; Ami V. Desai, MD, MSCE, and Mark A. Applebaum, MD, Department of Pediatrics and Comer Children's Hospital, University of Chicago, Chicago, IL; Dominique Valteau-Couanet, MD, PhD, Institute Gustave Roussy, Villejuif, France; Julie R. Park, MD, Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA; Andrew D.J. Pearson, MD, Paediatric Drug Development, Children and Young People's Unit, Royal Marsden Hospital, London, UK; Gudrun Schleiermacher, MD, PhD, Department of Pediatric, Adolescents, and Young Adults Oncology and INSERM U830, Institut Curie, Paris, France; Meredith S. Irwin, MD, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada; Michael Hogarty, MD, Department of Pediatrics, University of Pennsylvania, Philadelphia, PA; Arlene Naranjo, PhD, Department of Biostatistics, Children's Oncology Group Statistics and Data Center, University of Florida, Gainesville, FL; Samuel Volchenboum, MD, PhD, and Susan L. Cohn, MD, Department of Pediatrics and Comer Children's Hospital, University of Chicago, Chicago, IL; and Wendy B. London, PhD, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Michael Hogarty
- Elizabeth Sokol, MD, Department of Pediatrics and Lurie Children's Hospital, Northwestern University, Chicago, IL; Ami V. Desai, MD, MSCE, and Mark A. Applebaum, MD, Department of Pediatrics and Comer Children's Hospital, University of Chicago, Chicago, IL; Dominique Valteau-Couanet, MD, PhD, Institute Gustave Roussy, Villejuif, France; Julie R. Park, MD, Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA; Andrew D.J. Pearson, MD, Paediatric Drug Development, Children and Young People's Unit, Royal Marsden Hospital, London, UK; Gudrun Schleiermacher, MD, PhD, Department of Pediatric, Adolescents, and Young Adults Oncology and INSERM U830, Institut Curie, Paris, France; Meredith S. Irwin, MD, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada; Michael Hogarty, MD, Department of Pediatrics, University of Pennsylvania, Philadelphia, PA; Arlene Naranjo, PhD, Department of Biostatistics, Children's Oncology Group Statistics and Data Center, University of Florida, Gainesville, FL; Samuel Volchenboum, MD, PhD, and Susan L. Cohn, MD, Department of Pediatrics and Comer Children's Hospital, University of Chicago, Chicago, IL; and Wendy B. London, PhD, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Arlene Naranjo
- Elizabeth Sokol, MD, Department of Pediatrics and Lurie Children's Hospital, Northwestern University, Chicago, IL; Ami V. Desai, MD, MSCE, and Mark A. Applebaum, MD, Department of Pediatrics and Comer Children's Hospital, University of Chicago, Chicago, IL; Dominique Valteau-Couanet, MD, PhD, Institute Gustave Roussy, Villejuif, France; Julie R. Park, MD, Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA; Andrew D.J. Pearson, MD, Paediatric Drug Development, Children and Young People's Unit, Royal Marsden Hospital, London, UK; Gudrun Schleiermacher, MD, PhD, Department of Pediatric, Adolescents, and Young Adults Oncology and INSERM U830, Institut Curie, Paris, France; Meredith S. Irwin, MD, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada; Michael Hogarty, MD, Department of Pediatrics, University of Pennsylvania, Philadelphia, PA; Arlene Naranjo, PhD, Department of Biostatistics, Children's Oncology Group Statistics and Data Center, University of Florida, Gainesville, FL; Samuel Volchenboum, MD, PhD, and Susan L. Cohn, MD, Department of Pediatrics and Comer Children's Hospital, University of Chicago, Chicago, IL; and Wendy B. London, PhD, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Samuel Volchenboum
- Elizabeth Sokol, MD, Department of Pediatrics and Lurie Children's Hospital, Northwestern University, Chicago, IL; Ami V. Desai, MD, MSCE, and Mark A. Applebaum, MD, Department of Pediatrics and Comer Children's Hospital, University of Chicago, Chicago, IL; Dominique Valteau-Couanet, MD, PhD, Institute Gustave Roussy, Villejuif, France; Julie R. Park, MD, Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA; Andrew D.J. Pearson, MD, Paediatric Drug Development, Children and Young People's Unit, Royal Marsden Hospital, London, UK; Gudrun Schleiermacher, MD, PhD, Department of Pediatric, Adolescents, and Young Adults Oncology and INSERM U830, Institut Curie, Paris, France; Meredith S. Irwin, MD, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada; Michael Hogarty, MD, Department of Pediatrics, University of Pennsylvania, Philadelphia, PA; Arlene Naranjo, PhD, Department of Biostatistics, Children's Oncology Group Statistics and Data Center, University of Florida, Gainesville, FL; Samuel Volchenboum, MD, PhD, and Susan L. Cohn, MD, Department of Pediatrics and Comer Children's Hospital, University of Chicago, Chicago, IL; and Wendy B. London, PhD, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Susan L Cohn
- Elizabeth Sokol, MD, Department of Pediatrics and Lurie Children's Hospital, Northwestern University, Chicago, IL; Ami V. Desai, MD, MSCE, and Mark A. Applebaum, MD, Department of Pediatrics and Comer Children's Hospital, University of Chicago, Chicago, IL; Dominique Valteau-Couanet, MD, PhD, Institute Gustave Roussy, Villejuif, France; Julie R. Park, MD, Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA; Andrew D.J. Pearson, MD, Paediatric Drug Development, Children and Young People's Unit, Royal Marsden Hospital, London, UK; Gudrun Schleiermacher, MD, PhD, Department of Pediatric, Adolescents, and Young Adults Oncology and INSERM U830, Institut Curie, Paris, France; Meredith S. Irwin, MD, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada; Michael Hogarty, MD, Department of Pediatrics, University of Pennsylvania, Philadelphia, PA; Arlene Naranjo, PhD, Department of Biostatistics, Children's Oncology Group Statistics and Data Center, University of Florida, Gainesville, FL; Samuel Volchenboum, MD, PhD, and Susan L. Cohn, MD, Department of Pediatrics and Comer Children's Hospital, University of Chicago, Chicago, IL; and Wendy B. London, PhD, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Wendy B London
- Elizabeth Sokol, MD, Department of Pediatrics and Lurie Children's Hospital, Northwestern University, Chicago, IL; Ami V. Desai, MD, MSCE, and Mark A. Applebaum, MD, Department of Pediatrics and Comer Children's Hospital, University of Chicago, Chicago, IL; Dominique Valteau-Couanet, MD, PhD, Institute Gustave Roussy, Villejuif, France; Julie R. Park, MD, Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA; Andrew D.J. Pearson, MD, Paediatric Drug Development, Children and Young People's Unit, Royal Marsden Hospital, London, UK; Gudrun Schleiermacher, MD, PhD, Department of Pediatric, Adolescents, and Young Adults Oncology and INSERM U830, Institut Curie, Paris, France; Meredith S. Irwin, MD, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada; Michael Hogarty, MD, Department of Pediatrics, University of Pennsylvania, Philadelphia, PA; Arlene Naranjo, PhD, Department of Biostatistics, Children's Oncology Group Statistics and Data Center, University of Florida, Gainesville, FL; Samuel Volchenboum, MD, PhD, and Susan L. Cohn, MD, Department of Pediatrics and Comer Children's Hospital, University of Chicago, Chicago, IL; and Wendy B. London, PhD, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| |
Collapse
|
16
|
Liang WH, Federico SM, London WB, Naranjo A, Irwin MS, Volchenboum SL, Cohn SL. Tailoring Therapy for Children With Neuroblastoma on the Basis of Risk Group Classification: Past, Present, and Future. JCO Clin Cancer Inform 2020; 4:895-905. [PMID: 33058692 PMCID: PMC7608590 DOI: 10.1200/cci.20.00074] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2020] [Indexed: 12/12/2022] Open
Abstract
For children with neuroblastoma, the likelihood of cure varies widely according to age at diagnosis, disease stage, and tumor biology. Treatments are tailored for children with this clinically heterogeneous malignancy on the basis of a combination of markers that are predictive of risk of relapse and death. Sequential risk-based, cooperative-group clinical trials conducted during the past 4 decades have led to improved outcome for children with neuroblastoma. Increasingly accurate risk classification and refinements in treatment stratification strategies have been achieved with the more recent discovery of robust genomic and molecular biomarkers. In this review, we discuss the history of neuroblastoma risk classification in North America and Europe and highlight efforts by the International Neuroblastoma Risk Group (INRG) Task Force to develop a consensus approach for pretreatment stratification using seven risk criteria including an image-based staging system-the INRG Staging System. We also update readers on the current Children's Oncology Group risk classifier and outline plans for the development of a revised 2021 Children's Oncology Group classifier that will incorporate INRG Staging System criteria to facilitate harmonization of risk-based frontline treatment strategies conducted around the globe. In addition, we discuss new approaches to establish increasingly robust, future risk classification algorithms that will further refine treatment stratification using machine learning tools and expanded data from electronic health records and the INRG Data Commons.
Collapse
Affiliation(s)
- Wayne H. Liang
- Department of Pediatrics and Informatics Institute, University of Alabama at Birmingham, Birmingham, AL
| | - Sara M. Federico
- Department of Oncology, St Jude Children’s Research Hospital, Memphis, TN
| | - Wendy B. London
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Arlene Naranjo
- Department of Biostatistics, Children’s Oncology Group Statistics and Data Center, University of Florida, Gainesville, FL
| | - Meredith S. Irwin
- Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Samuel L. Volchenboum
- Department of Pediatrics and Comer Children’s Hospital, University of Chicago, Chicago, IL
| | - Susan L. Cohn
- Department of Pediatrics and Comer Children’s Hospital, University of Chicago, Chicago, IL
| |
Collapse
|
17
|
Juan Ribelles A, Gargallo P, Ferriol C, Segura V, Yáñez Y, Juan B, Cañada AJ, Font de Mora J, Cañete A, Castel V. Distribution of segmental chromosomal alterations in neuroblastoma. Clin Transl Oncol 2020; 23:1096-1104. [PMID: 32948984 DOI: 10.1007/s12094-020-02497-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/05/2020] [Indexed: 11/26/2022]
Abstract
BACKGROUND Neuroblastoma (NB) is a heterogeneous tumor with extremely diverse prognosis according to clinical and genetic factors such as specific combinations of chromosomal imbalances. METHODS Molecular karyotyping data from a national neuroblastic tumor database of 155 NB samples were analyzed and related to clinical data. RESULTS Segmental chromosomal alterations (SCA) were detected in 102 NB, whereas 45 only displayed numerical alterations. Incidence of SCA was higher in stage M (92%) and MYCN amplified (MNA) NB (96%). Presence of SCA was associated with older age, especially 1q gain and 3p deletion. 96% of the deaths were observed in the SCA group and 85% of the relapsed NB contained SCA. The alteration most commonly associated with a higher number of other segmental rearrangements was 11q deletion, followed by 4p deletion. Whole-chromosome 19 gain was associated with lower stages, absence of SCA and better outcome. CONCLUSIONS SCA are not randomly distributed and are concentrated on recurrent chromosomes. The most frequently affected chromosomes identify prognostic factors in specific risk groups. SCA are associated with older age and MNA. We have identified a small subset of patients with better outcome that share whole-chromosome 19 numeric gain, suggesting its use as a prognostic biomarker in NB.
Collapse
Affiliation(s)
- A Juan Ribelles
- Pediatric Oncology and Hematology Unit, Hospital U i P La Fe, Av. Fernando Abril Martorell, 106, Valencia, Spain.
| | - P Gargallo
- Clinical and Translational Oncology Research Group, Instituto de Investigación La Fe, Valencia, Spain
| | - C Ferriol
- Universitat de València, Valencia, Spain
| | - V Segura
- Clinical and Translational Oncology Research Group, Instituto de Investigación La Fe, Valencia, Spain
| | - Y Yáñez
- Clinical and Translational Oncology Research Group, Instituto de Investigación La Fe, Valencia, Spain
| | - B Juan
- Universitat de València, Valencia, Spain
| | - A J Cañada
- Biostatistics Department, Instituto de Investigación La Fe, Valencia, Spain
| | - J Font de Mora
- Clinical and Translational Oncology Research Group, Instituto de Investigación La Fe, Valencia, Spain
| | - A Cañete
- Pediatric Oncology and Hematology Unit, Hospital U i P La Fe, Av. Fernando Abril Martorell, 106, Valencia, Spain
| | - V Castel
- Clinical and Translational Oncology Research Group, Instituto de Investigación La Fe, Valencia, Spain
| |
Collapse
|
18
|
Southgate HED, Chen L, Curtin NJ, Tweddle DA. Targeting the DNA Damage Response for the Treatment of High Risk Neuroblastoma. Front Oncol 2020; 10:371. [PMID: 32309213 PMCID: PMC7145987 DOI: 10.3389/fonc.2020.00371] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 03/03/2020] [Indexed: 12/14/2022] Open
Abstract
Despite intensive multimodal therapy, the survival rate for high risk neuroblastoma (HR-NB) remains <50%. Most cases initially respond to treatment but almost half will subsequently relapse with aggressive treatment resistant disease. Novel treatments exploiting the molecular pathology of NB and/or overcoming resistance to current genotoxic therapies are needed before survival rates can significantly improve. DNA damage response (DDR) defects are frequently observed in HR-NB including allelic deletion and loss of function mutations in key DDR genes, oncogene induced replication stress and cell cycle checkpoint dysfunction. Exploiting defects in the DDR has been a successful treatment strategy in some adult cancers. Here we review the genetic features of HR-NB which lead to DDR defects and the emerging molecular targeting agents to exploit them.
Collapse
Affiliation(s)
- Harriet E D Southgate
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Lindi Chen
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Nicola J Curtin
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Deborah A Tweddle
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
19
|
Qin C, He X, Zhao Y, Tong CY, Zhu KY, Sun Y, Cheng C. Systematic computational identification of prognostic cytogenetic markers in neuroblastoma. BMC Med Genomics 2019; 12:192. [PMID: 31831008 PMCID: PMC6909636 DOI: 10.1186/s12920-019-0620-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Accepted: 11/12/2019] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Neuroblastoma (NB) is the most common extracranial solid tumor found in children. The frequent gain/loss of many chromosome bands in tumor cells and absence of mutations found at diagnosis suggests that NB is a copy number-driven cancer. Despite the previous work, a systematic analysis that investigates the relationship between such frequent gain/loss of chromosome bands and patient prognosis has yet to be implemented. METHODS First, we analyzed two NB CNV datasets to select chromosomal bands with a high frequency of gain or loss. Second, we applied a computational approach to infer sample-specific CNVs for each chromosomal band selected in step 1 based on gene expression data. Third, we applied univariate Cox proportional hazards models to examine the association between the resulting inferred copy number values (iCNVs) and patient survival. Finally, we applied multivariate Cox proportional hazards models to select chromosomal bands that remained significantly associated with prognosis after adjusting for critical clinical variables, including age, stage, gender, and MYCN amplification status. RESULTS Here, we used a computational method to infer the copy number variations (CNVs) of sample-specific chromosome bands from NB patient gene expression profiles. The resulting inferred CNVs (iCNVs) were highly correlated with the experimentally determined CNVs, demonstrating CNVs can be accurately inferred from gene expression profiles. Using this iCNV metric, we identified 58 frequent gain/loss chromosome bands that were significantly associated with patient survival. Furthermore, we found that 7 chromosome bands were still significantly associated with patient survival even when clinical factors, such as MYCN status, were considered. Particularly, we found that the chromosome band chr11p14 has high potential as a novel candidate cytogenetic biomarker for clinical use. CONCLUSION Our analysis resulted in a comprehensive list of prognostic chromosome bands supported by strong statistical evidence. In particular, the chr11p14 gain event provided additional prognostic value in addition to well-established clinical factors, including MYCN status, and thereby represents a novel candidate cytogenetic biomarker with high clinical potential. Additionally, this computational framework could be readily extended to other cancer types, such as leukemia.
Collapse
Affiliation(s)
- Chao Qin
- Beijing Key Lab of Traffic Data Analysis and Mining, School of Computer and Information Technology, Beijing Jiaotong University, No.3 Shangyuancun, Beijing, 100044 Haidian District China
- Department of Medicine, Baylor College of Medicine, BCM451, Suite 100D, Houston, TX 77030 USA
| | - Xiaoyan He
- Center for Clinical Molecular Medicine, Children’s Hospital, Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, 400014 China
| | - Yanding Zhao
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Lebanon, NH 03766 USA
| | - Chun-Yip Tong
- Department of Medicine, Baylor College of Medicine, BCM451, Suite 100D, Houston, TX 77030 USA
| | - Kenneth Y. Zhu
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755 USA
| | - Yongqi Sun
- Beijing Key Lab of Traffic Data Analysis and Mining, School of Computer and Information Technology, Beijing Jiaotong University, No.3 Shangyuancun, Beijing, 100044 Haidian District China
| | - Chao Cheng
- Department of Medicine, Baylor College of Medicine, BCM451, Suite 100D, Houston, TX 77030 USA
| |
Collapse
|
20
|
Depuydt P, Boeva V, Hocking TD, Cannoodt R, Ambros IM, Ambros PF, Asgharzadeh S, Attiyeh EF, Combaret V, Defferrari R, Fischer M, Hero B, Hogarty MD, Irwin MS, Koster J, Kreissman S, Ladenstein R, Lapouble E, Laureys G, London WB, Mazzocco K, Nakagawara A, Noguera R, Ohira M, Park JR, Pötschger U, Theissen J, Tonini GP, Valteau-Couanet D, Varesio L, Versteeg R, Speleman F, Maris JM, Schleiermacher G, De Preter K. Genomic Amplifications and Distal 6q Loss: Novel Markers for Poor Survival in High-risk Neuroblastoma Patients. J Natl Cancer Inst 2019. [PMID: 29514301 PMCID: PMC6186524 DOI: 10.1093/jnci/djy022] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Neuroblastoma is characterized by substantial clinical heterogeneity. Despite intensive treatment, the survival rates of high-risk neuroblastoma patients are still disappointingly low. Somatic chromosomal copy number aberrations have been shown to be associated with patient outcome, particularly in low- and intermediate-risk neuroblastoma patients. To improve outcome prediction in high-risk neuroblastoma, we aimed to design a prognostic classification method based on copy number aberrations. Methods In an international collaboration, normalized high-resolution DNA copy number data (arrayCGH and SNP arrays) from 556 high-risk neuroblastomas obtained at diagnosis were collected from nine collaborative groups and segmented using the same method. We applied logistic and Cox proportional hazard regression to identify genomic aberrations associated with poor outcome. Results In this study, we identified two types of copy number aberrations that are associated with extremely poor outcome. Distal 6q losses were detected in 5.9% of patients and were associated with a 10-year survival probability of only 3.4% (95% confidence interval [CI] = 0.5% to 23.3%, two-sided P = .002). Amplifications of regions not encompassing the MYCN locus were detected in 18.1% of patients and were associated with a 10-year survival probability of only 5.8% (95% CI = 1.5% to 22.2%, two-sided P < .001). Conclusions Using a unique large copy number data set of high-risk neuroblastoma cases, we identified a small subset of high-risk neuroblastoma patients with extremely low survival probability that might be eligible for inclusion in clinical trials of new therapeutics. The amplicons may also nominate alternative treatments that target the amplified genes.
Collapse
Affiliation(s)
- Pauline Depuydt
- Center for Medical Genetics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| | - Valentina Boeva
- Institut Cochin, Inserm U1016, CNRS UMR 8104, Université Paris Descartes UMR-S1016, Paris, France.,Institut Curie, Inserm U900, Mines ParisTech, PSL Research University, Paris, France
| | - Toby D Hocking
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Robrecht Cannoodt
- Center for Medical Genetics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium.,Data Mining and Modelling for Biomedicine Group, VIB Center for Inflammation Research, Ghent, Belgium
| | - Inge M Ambros
- Children's Cancer Research Institute, Austria.,Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Peter F Ambros
- Children's Cancer Research Institute, Austria.,Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Shahab Asgharzadeh
- Division of Hematology/Oncology, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA
| | - Edward F Attiyeh
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA.,Center for Childhood Cancer Research, University of Pennsylvania, Philadelphia, PA.,Department of Pediatrics, University of Pennsylvania, Philadelphia, PA
| | - Valérie Combaret
- Centre Léon-Bérard, Laboratoire de Recherche Translationnelle, Lyon, France
| | | | - Matthias Fischer
- Department of Experimental Pediatric Oncology, University of Cologne, Cologne, Germany.,University Children's Hospital Cologne, Medical Faculty, and Center for Molecular Medicine Cologne
| | - Barbara Hero
- Department of Pediatric Oncology and Hematology, University of Cologne, Cologne, Germany
| | - Michael D Hogarty
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA.,Perelman School of Medicine (MDH), University of Pennsylvania, Philadelphia, PA
| | - Meredith S Irwin
- Division of Hematology-Oncology, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Jan Koster
- Department of Oncogenomics, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Susan Kreissman
- Department of Pediatrics, Duke University School of Medicine, Durham, NC
| | - Ruth Ladenstein
- Children's Cancer Research Institute, Austria.,Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Eve Lapouble
- Genetic Somatic Unit.,Institut Curie, Paris, France
| | - Geneviève Laureys
- Department of Pediatric Hematology and Oncology, Ghent University Hospital, De Pintelaan, Ghent, Belgium
| | - Wendy B London
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Katia Mazzocco
- Department of Pathology, Istituto Giannina Gaslini, Genova, Italy
| | | | - Rosa Noguera
- Pathology Department, Medical School, University of Valencia, Valencia, Spain.,Medical Research Foundation INCLIVA, Valencia, Spain.,CIBERONC, Madrid, Spain
| | - Miki Ohira
- Research Institute for Clinical Oncology Saitama Cancer Center, Saitama, Japan
| | - Julie R Park
- Seattle Children's Hospital and University of Washington, Seattle, WA
| | | | - Jessica Theissen
- Department of Experimental Pediatric Oncology, University of Cologne, Cologne, Germany
| | - Gian Paolo Tonini
- Laboratory of Neuroblastoma, Onco/Haematology Laboratory, University of Padua, Pediatric Research Institute (IRP)-Città della Speranza, Padova, Italy
| | | | - Luigi Varesio
- Laboratory of Molecular Biology (LV), Istituto Giannina Gaslini, Genova, Italy
| | - Rogier Versteeg
- Department of Oncogenomics, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Frank Speleman
- Center for Medical Genetics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| | - John M Maris
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA.,Center for Childhood Cancer Research, University of Pennsylvania, Philadelphia, PA.,Department of Pediatrics, University of Pennsylvania, Philadelphia, PA.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA.,Abramson Family Cancer Research Institute, Philadelphia, PA
| | - Gudrun Schleiermacher
- U830 INSERM, Recherche Translationelle en Oncologie Pédiatrique (RTOP) and Department of Pediatric Oncology
| | - Katleen De Preter
- Center for Medical Genetics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| |
Collapse
|
21
|
Trigg RM, Shaw JA, Turner SD. Opportunities and challenges of circulating biomarkers in neuroblastoma. Open Biol 2019; 9:190056. [PMID: 31088252 PMCID: PMC6544987 DOI: 10.1098/rsob.190056] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 04/23/2019] [Indexed: 12/11/2022] Open
Abstract
Molecular analysis of nucleic acid and protein biomarkers is becoming increasingly common in paediatric oncology for diagnosis, risk stratification and molecularly targeted therapeutics. However, many current and emerging biomarkers are based on analysis of tumour tissue, which is obtained through invasive surgical procedures and in some cases may not be accessible. Over the past decade, there has been growing interest in the utility of circulating biomarkers such as cell-free nucleic acids, circulating tumour cells and extracellular vesicles as a so-called liquid biopsy of cancer. Here, we review the potential of emerging circulating biomarkers in the management of neuroblastoma and highlight challenges to their implementation in the clinic.
Collapse
Affiliation(s)
- Ricky M. Trigg
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Jacqui A. Shaw
- Leicester Cancer Research Centre, College of Life Sciences, University of Leicester, Leicester LE2 7LX, UK
| | - Suzanne D. Turner
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge CB2 0QQ, UK
| |
Collapse
|
22
|
Abstract
INTRODUCTION Neuroblastoma ranks third among pediatric malignancies. CASE REPORT The case of a 3-year-old child is presented, who suddenly had frequent, unproductive, emetic cough; fever; and weight loss. Lung X-ray showed an opacity situated in the posterior superior mediastinum. Thoracic ultrasound revealed a slightly inhomogeneous, hypoechoic mass located in the posterior superior mediastinum. Computed tomography evidenced a tumor mass with homogeneous appearance in the costo-vertebral groove. Histological examination confirmed the diagnosis of ganglioneuroblastoma. CONCLUSION Although history and clinical examination provided few elements, diagnosis was made based on imaging and histopathological examination.
Collapse
|
23
|
Meany HJ. Non-High-Risk Neuroblastoma: Classification and Achievements in Therapy. CHILDREN (BASEL, SWITZERLAND) 2019; 6:E5. [PMID: 30626019 PMCID: PMC6352142 DOI: 10.3390/children6010005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 12/23/2018] [Accepted: 12/28/2018] [Indexed: 12/12/2022]
Abstract
Neuroblastoma, a tumor of the sympathetic nervous system, is the most common extra-cranial neoplasm of childhood. Variables with prognostic significance in patients with neuroblastoma, including age at diagnosis, disease stage, tumor histology, MYCN gene amplification, tumor cell ploidy, and the presence of segmental chromosomal aberrations are utilized to classify patients based on risk of disease recurrence. Patients with non-high-risk neuroblastoma, low- and intermediate-risk categories, represent nearly half of all newly diagnosed cases. This group has an excellent event-free and overall survival with current therapy. Over time, the objective in treatment of non-high-risk neuroblastoma has been reduction of therapy intensity to minimize short- and long-term adverse events all the while maintaining excellent outcomes.
Collapse
Affiliation(s)
- Holly J Meany
- Center for Cancer and Blood Disorders, Children's National Health System, The George Washington University School of Medicine and Health Sciences, Washington, DC 20010, USA.
| |
Collapse
|
24
|
Fusco P, Esposito MR, Tonini GP. Chromosome instability in neuroblastoma. Oncol Lett 2018; 16:6887-6894. [PMID: 30546420 PMCID: PMC6256707 DOI: 10.3892/ol.2018.9545] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 08/30/2018] [Indexed: 12/28/2022] Open
Abstract
Neuroblastoma is a neural crest-derived tumor that accounts for 7-10% of all malignancies in children and ~15% of all childhood cancer-associated mortalities. Approximately 50% of patients are characterized as high-risk (HR) and have an overall survival of <40% at 5 years from diagnosis. HR patients with unfavorable prognosis exhibit several structural copy number variations (CNVs), whereas localized tumors belonging to patients in the low- and intermediate-risk classes, have favorable outcomes and display several numerical CNVs. Taken together these results are indicative of chromosome instability (CIN) in neuroblastoma tumor cells. The present review discusses multiple aspects of CIN including methods of measuring CIN, CIN targeting as a therapeutic strategy in cancer and the effects of CIN in neuroblastoma development and aggressiveness with particular emphasis on the CIN gene signature associated with HR neuroblastoma patients.
Collapse
Affiliation(s)
- Pina Fusco
- Neuroblastoma Laboratory, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, I-35127 Padua, Italy
| | - Maria Rosaria Esposito
- Neuroblastoma Laboratory, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, I-35127 Padua, Italy
| | - Gian Paolo Tonini
- Neuroblastoma Laboratory, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, I-35127 Padua, Italy
| |
Collapse
|
25
|
Esposito MR, Binatti A, Pantile M, Coppe A, Mazzocco K, Longo L, Capasso M, Lasorsa VA, Luksch R, Bortoluzzi S, Tonini GP. Somatic mutations in specific and connected subpathways are associated with short neuroblastoma patients' survival and indicate proteins targetable at onset of disease. Int J Cancer 2018; 143:2525-2536. [PMID: 29992558 DOI: 10.1002/ijc.31748] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 05/24/2018] [Accepted: 06/21/2018] [Indexed: 12/29/2022]
Abstract
Neuroblastoma (NB) is an embryonic malignancy of the sympathetic nervous system with heterogeneous biological, morphological, genetic and clinical characteristics. Although genomic studies revealed the specific biological features of NB pathogenesis useful for new therapeutic approaches, the improvement of high-risk (HR)-NB patients overall survival remains unsatisfactory. To further clarify the biological basis of disease aggressiveness, we used whole-exome sequencing to examine the genomic landscape of HR-NB patients at stage M with short survival (SS) and long survival (LS). Only a few genes, including SMARCA4, SMO, ZNF44 and CHD2, were recurrently and specifically mutated in the SS group, confirming the low recurrence of common mutations in this tumor. A systems biology approach revealed that in the two patient groups, mutations occurred in different pathways. Mutated genes (ARHGEF11, CACNA1G, FGF4, PTPRA, PTK2, ANK3, SMO, NTNG2, VCL and NID2) regulate the MAPK pathway associated with the organization of the extracellular matrix, cell motility through PTK2 signaling and matrix metalloproteinase activity. Moreover, we detected mutations in LAMA2, PTK2, LAMA4, and MMP14 genes, impairing MET signaling, in SFI1 and CHD2 involved in centrosome maturation and chromosome remodeling, in AK7 and SPTLC2, which regulate the metabolism of nucleotides and lipoproteins, and in NALCN, SLC12A1, SLC9A9, which are involved in the transport of small molecules. Notably, connected networks of somatically mutated genes specific for SS patients were identified. The detection of mutated genes present at the onset of disease may help to address an early treatment of HR-NB patients using FDA-approved compounds targeting the deregulated pathways.
Collapse
Affiliation(s)
- Maria Rosaria Esposito
- Neuroblastoma Laboratory, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
| | - Andrea Binatti
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Marcella Pantile
- Neuroblastoma Laboratory, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
| | - Alessandro Coppe
- Department of Women's and Children's Health, University of Padova, Padua, Italy
| | - Katia Mazzocco
- Translational Research Department, Laboratory Medicine, Diagnostics and Services U.O.C. Pathological Anatomy, IRCCS Giannina Gaslini Institute, Genoa, Italy
| | - Luca Longo
- U.O.C. Bioterapie, Ospedale Policlinico San Martino, Genoa, Italy
| | - Mario Capasso
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy.,CEINGE Biotecnologie Avanzate, Naples, Italy.,IRCCS SDN, Istituto di Ricerca Diagnostica e Nucleare, Naples, Italy
| | | | - Roberto Luksch
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Gian Paolo Tonini
- Neuroblastoma Laboratory, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
| |
Collapse
|
26
|
Berbegall AP, Bogen D, Pötschger U, Beiske K, Bown N, Combaret V, Defferrari R, Jeison M, Mazzocco K, Varesio L, Vicha A, Ash S, Castel V, Coze C, Ladenstein R, Owens C, Papadakis V, Ruud E, Amann G, Sementa AR, Navarro S, Ambros PF, Noguera R, Ambros IM. Heterogeneous MYCN amplification in neuroblastoma: a SIOP Europe Neuroblastoma Study. Br J Cancer 2018; 118:1502-1512. [PMID: 29755120 PMCID: PMC5988829 DOI: 10.1038/s41416-018-0098-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 03/01/2018] [Accepted: 04/06/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND In neuroblastoma (NB), the most powerful prognostic marker, the MYCN amplification (MNA), occasionally shows intratumoural heterogeneity (ITH), i.e. coexistence of MYCN-amplified and non-MYCN-amplified tumour cell clones, called heterogeneous MNA (hetMNA). Prognostication and therapy allocation are still unsolved issues. METHODS The SIOPEN Biology group analysed 99 hetMNA NBs focussing on the prognostic significance of MYCN ITH. RESULTS Patients <18 months (18 m) showed a better outcome in all stages as compared to older patients (5-year OS in localised stages: <18 m: 0.95 ± 0.04, >18 m: 0.67 ± 0.14, p = 0.011; metastatic: <18 m: 0.76 ± 0.15, >18 m: 0.28 ± 0.09, p = 0.084). The genomic 'background', but not MNA clone sizes, correlated significantly with relapse frequency and OS. No relapses occurred in cases of only numerical chromosomal aberrations. Infiltrated bone marrows and relapse tumour cells mostly displayed no MNA. However, one stage 4s tumour with segmental chromosomal aberrations showed a homogeneous MNA in the relapse. CONCLUSIONS This study provides a rationale for the necessary distinction between heterogeneous and homogeneous MNA. HetMNA tumours have to be evaluated individually, taking age, stage and, most importantly, genomic background into account to avoid unnecessary upgrading of risk/overtreatment, especially in infants, as well as in order to identify tumours prone to developing homogeneous MNA.
Collapse
Affiliation(s)
- Ana P Berbegall
- Department of Pathology, Medical School, University of Valencia/INCLIVA Biomedical Research Institute, 46010, Valencia, Spain
- Ciberonc, Madrid, Spain
| | - Dominik Bogen
- Department of Tumour Biology CCRI, Children's Cancer Research Institute, St. Anna Kinderkrebsforschung, 1090, Vienna, Austria
| | - Ulrike Pötschger
- S2IRP: Studies and Statistics for Integrated Research and Projects CCRI, Children's Cancer Research Institute, St. Anna Kinderkrebsforschung, 1090, Vienna, Austria
| | - Klaus Beiske
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo and Department of Pathology, Norwegian Radium Hospital, Oslo University Hospital, 0372, Oslo, Norway
| | - Nick Bown
- Northern Genetics Service, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Institute of Genetic Medicine, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| | - Valérie Combaret
- Centre Léon Bérard, Laboratoire de Recherche Translationnelle, 28 rue Laennec, Lyon, 69008, France
| | - Raffaella Defferrari
- Department of Pathology, Gaslini Institute, Largo G. Gaslini 5, 16147, Genoa, Italy
| | - Marta Jeison
- Cancer Cytogenetic and Molecular Cytogenetic Laboratory, Schneider Children's Medical Center of Israel, 49202, Petach Tikva, Israel
| | - Katia Mazzocco
- Department of Pathology, Gaslini Institute, Largo G. Gaslini 5, 16147, Genoa, Italy
| | - Luigi Varesio
- Laboratory of Molecular Biology, Gaslini Institute, Largo G. Gaslini 5, 16147, Genoa, Italy
| | - Ales Vicha
- Department of Pediatric Hematology and Oncology, Charles University in Prague, Second Faculty of Medicine and University Hospital Motol, 15006, Prague, Czech Republic
| | - Shifra Ash
- Department of Paediatric Haematology-Oncology, Schneider Children's Medical Center of Israel, 49202, Petach Tikva, Israel
| | - Victoria Castel
- Pediatric Oncology Unit, Hospital Universitari i Politècnic La Fe, 46026, Valencia, Spain
| | - Carole Coze
- Department of Paediatric Haematology-Oncology, Aix-Marseille University and APHM, Hôpital d' Enfants de La Timone, 13385, Marseille, France
| | - Ruth Ladenstein
- S2IRP: Studies and Statistics for Integrated Research and Projects CCRI, Children's Cancer Research Institute, St. Anna Kinderkrebsforschung, 1090, Vienna, Austria
- St Anna Children's Hospital and Department of Paediatrics of the Medical University, 1090, Vienna, Austria
| | - Cormac Owens
- Our Lady's Children's Hospital, Crumlin, Dublin, D12 N512, Ireland
| | - Vassilios Papadakis
- Department of Paediatric Haematology-Oncology, Agia Sofia Children's Hospital Athens, 11528, Athens, Greece
| | - Ellen Ruud
- Department of Paediatric Medicine, Rikshospitalet, Oslo University Hospital, 0372, Oslo, Norway
| | - Gabriele Amann
- Institute of Clinical Pathology, Medical University Vienna, Vienna, Austria
| | - Angela R Sementa
- Department of Pathology, Gaslini Institute, Largo G. Gaslini 5, 16147, Genoa, Italy
| | - Samuel Navarro
- Department of Pathology, Medical School, University of Valencia/INCLIVA Biomedical Research Institute, 46010, Valencia, Spain
- Ciberonc, Madrid, Spain
| | - Peter F Ambros
- Department of Tumour Biology CCRI, Children's Cancer Research Institute, St. Anna Kinderkrebsforschung, 1090, Vienna, Austria
- Department of Paediatrics, Medical University Vienna, Vienna, Austria
| | - Rosa Noguera
- Department of Pathology, Medical School, University of Valencia/INCLIVA Biomedical Research Institute, 46010, Valencia, Spain.
- Ciberonc, Madrid, Spain.
| | - Inge M Ambros
- Department of Tumour Biology CCRI, Children's Cancer Research Institute, St. Anna Kinderkrebsforschung, 1090, Vienna, Austria.
| |
Collapse
|
27
|
Neuroblastoma: clinical and biological approach to risk stratification and treatment. Cell Tissue Res 2018; 372:195-209. [PMID: 29572647 DOI: 10.1007/s00441-018-2821-2] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 02/28/2018] [Indexed: 01/15/2023]
Abstract
Neuroblastoma is the most common extra-cranial solid tumor of childhood and the most common in the first year of life. It is a unique malignancy in that infants often present with either localized or metastatic disease that can spontaneously regress without intervention while older children can succumb to the disease after months to years of arduous therapy. Given this wide range of outcomes, the International Neuroblastoma Risk Group was created to stratify patients based on presenting characteristics and tumor biology in order to guide intensity of treatment strategies. The goal has been to decrease therapy for low-risk patients to avoid long-term complications while augmenting and targeting therapies for high-risk patients to improve overall survival. The international risk stratification depends on age, stage, histology, MYCN gene amplification status, tumor cell ploidy and segmental chromosomal abnormalities. Treatment for asymptomatic low-risk patients with an estimated survival of > 98% is often observation or surgical resection alone, whereas intermediate-risk patients with an estimated survival of > 90% require moderate doses of response-adjusted chemotherapy along with resection. High-risk patients undergo multiple cycles of combination chemotherapy before surgery, followed by consolidation with myeloablative autologous hematopoietic stem cell transplantation and local radiation and finally immunotherapy with differentiation therapy as maintenance phase. With this approach, outcome for patients with neuroblastoma has improved, as the field continues to expand efforts in more targeted therapies for high-risk patients.
Collapse
|
28
|
Abstract
Opsoclonus myoclonus syndrome (OMS), often called "dancing eyed syndrome," is a rare neurological condition associated with neuroblastoma in the majority of all childhood cases. Genomic copy number profiles have shown to be of prognostic significance for neuroblastoma patients. The aim of this retrospective multicenter study was to analyze the genomic copy number profiles of tumors from children with neuroblastoma presenting with OMS at diagnosis. In 44 cases of neuroblastoma associated with OMS, overall genomic profiling by either array-comparative genomic hybridization or single nucleotide polymorphism array proved successful in 91% of the cases, distinguishing tumors harboring segmental chromosome alterations from those with numerical chromosome alterations only. A total of 23/44 (52%) tumors showed an segmental chromosome alterations genomic profile, 16/44 (36%) an numerical chromosome alterations genomic profile, and 1 case displayed an atypical profile (12q amplicon). No recurrently small interstitial copy number alterations were identified. With no tumor relapse nor disease-related deaths, the overall genomic profile was not of prognostic impact with regard to the oncological outcome in this series of patients. Thus, the observation of an excellent oncological outcome, even for those with an unfavorable genomic profile of neuroblastoma, supports the hypothesis that an immune response might be involved in tumor control in these patients with OMS.
Collapse
|
29
|
Zhong ZY, Shi BJ, Zhou H, Wang WB. CD133 expression and MYCN amplification induce chemoresistance and reduce average survival time in pediatric neuroblastoma. J Int Med Res 2018; 46:1209-1220. [PMID: 29322842 PMCID: PMC5972240 DOI: 10.1177/0300060517732256] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Objectives Neuroblastoma (NB) is the most common pediatric solid tumor derived from the sympathetic nervous system. MYCN is amplified in nearly half of patients with NB, and its association with rapid disease progression and poor outcome is controversial. Characterization of cancer stem cells (CSCs) in NBs has been rarely studied. This study was performed to determine whether MYCN and CD133+ CSCs are associated with chemotherapy resistance and the survival time of patients with NB. Methods Fifty patients with an unequivocal pathological diagnosis of NB were recruited. MYCN expression levels were measured before therapy. CSCs were derived and their multipotency tested by directed differentiation. The patients’ responses to chemotherapy and average survival time were compared among the groups as follows: CD133+, CD133−, MYCN amplification ≥5 times (i.e. MYCN≥5), MYCN<5, CD133+ plus MYCN≥5, and CD133− plus MYCN<5. Results CD133+ CSCs differentiated into neuron-like cells. CD133+ patients had a significantly poorer response to chemotherapy than did CD133− patients. CD133+ plus MYCN≥5 patients had a significantly shorter average survival time than did CD133− plus MYCN<5 patients. Conclusions CD133+ CSCs are chemoresistance. CD133 expression and MYCN amplification can be used together as a prognostic indicator of disease outcome.
Collapse
Affiliation(s)
- Zhi-Yong Zhong
- Pediatric Surgery Department, The Second of Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, P.R. China
| | - Bao-Jun Shi
- Pediatric Surgery Department, The Second of Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, P.R. China
| | - Hui Zhou
- Pediatric Surgery Department, The Second of Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, P.R. China
| | - Wen-Bo Wang
- Pediatric Surgery Department, The Second of Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, P.R. China
| |
Collapse
|
30
|
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: 11.5] [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
|
31
|
Tonini GP. Growth, progression and chromosome instability of Neuroblastoma: a new scenario of tumorigenesis? BMC Cancer 2017; 17:20. [PMID: 28056863 PMCID: PMC5217541 DOI: 10.1186/s12885-016-2986-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 12/08/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Neuroblastoma is a pediatric cancer with a low survival rate of patients with metastatic stage 4 disease. Tumor aggressiveness and progression have been associated with structural copy number variations (CNVs) that are observed in malignant cells. In contrast, localized Neuroblastomas, which are associated with a low number of structural CNVs but frequent numerical CNVs, are less aggressive, and patients have good outcomes. Finally, whole-genome and whole-exome sequencing of Neuroblastoma tissues have shown few damaging mutations in these tumors. CONCLUSIONS In the present report it is proposed that chromosome instability (CIN) plays a major role in Neuroblastoma tumorigenesis and that CIN is already present in the early phases of tumor development. High CIN can promote several types of chromosomal damage including chromothripsis, gene deletion, amplification and rearrangements, which deregulate gene expression. Indeed, gene rearrangements have been reported as a new scenario in the development of Neuroblastoma, which supports the hypothesis that CIN is an early step preliminary to the late catastrophic events leading to tumor development.
Collapse
Affiliation(s)
- Gian Paolo Tonini
- Neuroblastoma Laboratory, Italian Neuroblastoma Foundation, Pediatric Research Institute, Fondazione Città della Speranza, Corso Stati Uniti, 4, 35127, Padua, Italy.
| |
Collapse
|
32
|
Abstract
Neuroblastoma is the most common extracranial solid tumour occurring in childhood and has a diverse clinical presentation and course depending on the tumour biology. Unique features of these neuroendocrine tumours are the early age of onset, the high frequency of metastatic disease at diagnosis and the tendency for spontaneous regression of tumours in infancy. The most malignant tumours have amplification of the MYCN oncogene (encoding a transcription factor), which is usually associated with poor survival, even in localized disease. Although transgenic mouse models have shown that MYCN overexpression can be a tumour-initiating factor, many other cooperating genes and tumour suppressor genes are still under investigation and might also have a role in tumour development. Segmental chromosome alterations are frequent in neuroblastoma and are associated with worse outcome. The rare familial neuroblastomas are usually associated with germline mutations in ALK, which is mutated in 10-15% of primary tumours, and provides a potential therapeutic target. Risk-stratified therapy has facilitated the reduction of therapy for children with low-risk and intermediate-risk disease. Advances in therapy for patients with high-risk disease include intensive induction chemotherapy and myeloablative chemotherapy, followed by the treatment of minimal residual disease using differentiation therapy and immunotherapy; these have improved 5-year overall survival to 50%. Currently, new approaches targeting the noradrenaline transporter, genetic pathways and the tumour microenvironment hold promise for further improvements in survival and long-term quality of life.
Collapse
|
33
|
Factors associated with recurrence and survival length following relapse in patients with neuroblastoma. Br J Cancer 2016; 115:1048-1057. [PMID: 27701387 PMCID: PMC5117794 DOI: 10.1038/bjc.2016.302] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 07/11/2016] [Accepted: 08/29/2016] [Indexed: 12/29/2022] Open
Abstract
Background: Despite therapeutic advances, survival following relapse for neuroblastoma patients remains poor. We investigated clinical and biological factors associated with length of progression-free and overall survival following relapse in UK neuroblastoma patients. Methods: All cases of relapsed neuroblastoma, diagnosed during 1990–2010, were identified from four Paediatric Oncology principal treatment centres. Kaplan–Meier and Cox regression analyses were used to calculate post-relapse overall survival (PROS), post-relapse progression-free survival (PRPFS) between relapse and further progression, and to investigate influencing factors. Results: One hundred eighty-nine cases were identified from case notes, 159 (84.0%) high risk and 17 (9.0%), unresectable, MYCN non-amplified (non-MNA) intermediate risk (IR). For high-risk patients diagnosed >2000, median PROS was 8.4 months (interquartile range (IQR)=3.0–17.4) and median PRPFS was 4.7 months (IQR=2.1–7.1). For IR, unresectable non-MNA patients, median PROS was 11.8 months (IQR 9.0–51.6) and 5-year PROS was 24% (95% CI 7–45%). MYCN amplified (MNA) disease and bone marrow metastases at diagnosis were independently associated with worse PROS for high-risk cases. Eighty percent of high-risk relapses occurred within 2 years of diagnosis compared with 50% of unresectable non-MNA IR disease. Conclusions: Patients with relapsed HR neuroblastomas should be treatment stratified according to MYCN status and PRPFS should be the primary endpoint in early phase clinical trials. The failure to salvage the majority of IR neuroblastoma is concerning, supporting investigation of intensification of upfront treatment regimens in this group to determine whether their use would diminish likelihood of relapse.
Collapse
|
34
|
Wright DC, Adayapalam N, Bain N, Bain SM, Brown A, Buzzacott N, Carey L, Cross J, Dun K, Joy C, McCarthy C, Moore S, Murch AR, O'Malley F, Parker E, Watt J, Wilkin H, Fagan K, Pertile MD, Peters GB. Chromosome microarray proficiency testing and analysis of quality metric data trends through an external quality assessment program for Australasian laboratories. Pathology 2016; 48:586-96. [PMID: 27575971 DOI: 10.1016/j.pathol.2016.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 04/19/2016] [Accepted: 05/20/2016] [Indexed: 10/21/2022]
Abstract
Chromosome microarrays are an essential tool for investigation of copy number changes in children with congenital anomalies and intellectual deficit. Attempts to standardise microarray testing have focused on establishing technical and clinical quality criteria, however external quality assessment programs are still needed. We report on a microarray proficiency testing program for Australasian laboratories. Quality metrics evaluated included analytical accuracy, result interpretation, report completeness, and laboratory performance data: sample numbers, success and abnormality rate and reporting times. Between 2009 and 2014 nine samples were dispatched with variable results for analytical accuracy (30-100%), correct interpretation (32-96%), and report completeness (30-92%). Laboratory performance data (2007-2014) showed an overall mean success rate of 99.2% and abnormality rate of 23.6%. Reporting times decreased from >90 days to <30 days for normal results and from >102 days to <35 days for abnormal results. Data trends showed a positive correlation with improvement for all these quality metrics, however only 'report completeness' and reporting times reached statistical significance. Whether the overall improvement in laboratory performance was due to participation in this program, or from accumulated laboratory experience over time, is not clear. Either way, the outcome is likely to assist referring clinicians and improve patient care.
Collapse
Affiliation(s)
- D C Wright
- The Children's Hospital at Westmead, Westmead, NSW, Australia.
| | - N Adayapalam
- Royal Brisbane Hospital, Brisbane, Qld, Australia
| | - N Bain
- Hunter Area Pathology, Newcastle, NSW, Australia
| | - S M Bain
- SA Pathology, Adelaide, SA, Australia
| | - A Brown
- Wellington Hospital, Wellington, New Zealand
| | - N Buzzacott
- Western Genome Diagnostics, Perth, WA, Australia
| | - L Carey
- The Children's Hospital at Westmead, Westmead, NSW, Australia
| | - J Cross
- The Children's Hospital at Westmead, Westmead, NSW, Australia
| | - K Dun
- Royal Hobart Hospital, Hobart, Tas, Australia
| | - C Joy
- Mater Hospital, Brisbane, Qld, Australia
| | - C McCarthy
- Queensland Fertility Group, Brisbane, Qld, Australia
| | - S Moore
- SA Pathology, Adelaide, SA, Australia
| | - A R Murch
- Retired, formerly at Pathwest Laboratory Medicine WA, QEII Medical Centre, Nedlands, WA, Australia
| | - F O'Malley
- St Vincents Hospital, Melbourne, Vic, Australia
| | - E Parker
- Canterbury Health Laboratories, Christchurch, New Zealand
| | - J Watt
- Canterbury Health Laboratories, Christchurch, New Zealand
| | - H Wilkin
- Monash Medical Centre, Melbourne, Vic, Australia
| | - K Fagan
- Retired, formerly at Hunter Area Pathology Service, John Hunter Hospital, Newcastle, NSW, Australia
| | - M D Pertile
- Murdoch Children's Research Institute, Melbourne, Vic, Australia
| | - G B Peters
- The Children's Hospital at Westmead, Westmead, NSW, Australia
| |
Collapse
|
35
|
Pinto N, Mayfield JR, Raca G, Applebaum MA, Chlenski A, Sukhanova M, Bagatell R, Irwin MS, Little A, Rawwas J, Gosiengfiao Y, Delattre O, Janoueix-Lerosey I, Lapouble E, Schleiermacher G, Cohn SL. Segmental Chromosomal Aberrations in Localized Neuroblastoma Can be Detected in Formalin-Fixed Paraffin-Embedded Tissue Samples and Are Associated With Recurrence. Pediatr Blood Cancer 2016; 63:1019-23. [PMID: 26864375 PMCID: PMC5109976 DOI: 10.1002/pbc.25934] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 01/07/2016] [Accepted: 01/08/2016] [Indexed: 02/05/2023]
Abstract
BACKGROUND Array comparative genomic hybridization (CGH) analyses of frozen tumors have shown strong associations between the pattern of chromosomal aberrations and outcome in patients with advanced-stage neuroblastoma. New platforms for analyzing chromosomal aberrations using formalin-fixed paraffin-embedded (FFPE) tissue have recently been developed. We sought to determine whether chromosomal microarray analysis (CMA) using FFPE tumors is feasible and if segmental chromosomal aberrations were prognostic of recurrence in localized neuroblastoma. METHODS Patients with MYCN nonamplified International Neuroblastoma Staging System stage 1 and 2 disease who recurred were identified. CMA was performed with diagnostic FFPE samples using OncoScan™ FFPE Express 2.0. The prognostic significance of chromosomal pattern was validated in 105 patients with available CGH results. RESULTS In 26 evaluable patients, 11 recurred locally, nine had metastatic relapse, and six remained progression free >3 years from diagnosis. No chromosomal aberrations were identified in four tumors. Numerical chromosomal aberrations (NCAs) without segmental chromosomal aberration (SCA) were identified in 11 patients: six progressed locally, two had metastatic progression and 3 remained progression-free. Eleven patients had SCAs: four progressed locally, six developed metastatic progression and one remained progression-free. Five or more SCAs were only detected in tumors from patients who developed metastases (P = 0.0004). In the validation cohort, SCAs were associated with inferior event-free survival (EFS) compared to NCA (5-year EFS 68% ± 8.3% vs. 91% ± 3.6%, respectively; P = 0.0083). CONCLUSIONS It is feasible to evaluate chromosomal aberrations using FFPE neuroblastoma tissue. SCA is associated with inferior EFS in localized neuroblastoma patients, and multiple SCAs may be predictive of metastatic relapse.
Collapse
Affiliation(s)
- Navin Pinto
- Division of Pediatric Hematology/Oncology, Seattle Children’s Hospital, Seattle, Washington
| | - Jodi R. Mayfield
- Department of Pediatrics, University of New Mexico, Albuquerque, New Mexico
| | - Gordana Raca
- Department of Pediatrics, University of Chicago, Chicago, Illinois
| | - Mark A. Applebaum
- Department of Pediatrics, University of Southern California, Los Angeles, California
| | - Alexandre Chlenski
- Department of Pediatrics, University of Southern California, Los Angeles, California
| | - Madina Sukhanova
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Rochelle Bagatell
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Meredith S. Irwin
- Department of Pediatrics, Hospital for Sick Children (Toronto), Toronto, Ontario, Canada
| | - Anthony Little
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Jawhar Rawwas
- Children’s Hospitals and Clinics of Minnesota, Minneapolis, Minnesota
| | | | - Olivier Delattre
- Laboratoire de Génétique et Biologie des Cancers, Institut Curie, Paris, France
| | | | - Eve Lapouble
- Unité de Génétique Somatique, Institut Curie, Paris, France
| | - Gudrun Schleiermacher
- Laboratoire de Génétique et Biologie des Cancers, Institut Curie, Paris, France
- Unité de Génétique Somatique, Institut Curie, Paris, France
- Département de Pédiatrie, Institut Curie, Paris, France
| | - Susan L. Cohn
- Department of Pediatrics, University of Chicago, Chicago, Illinois
| |
Collapse
|