101
|
Nakamura N, Kishimoto K, Ishida T, Nakamura S, Tamura A, Kozaki A, Saito A, Hasegawa D, Kosaka Y. Muscle mass change during chemotherapy in children with high-risk neuroblastoma: a retrospective case series of 24 patients. Eur J Pediatr 2021; 180:3265-3271. [PMID: 33970314 DOI: 10.1007/s00431-021-04106-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 12/17/2022]
Abstract
The clinical characteristics, cause, and risk factors of sarcopenia are unclear in children. The aim of this study was to describe the course of and identify the factors related to muscle mass change during chemotherapy in children with neuroblastoma. A total of 24 consecutive patients aged below 18 years with newly diagnosed high-risk neuroblastoma between 2010 and 2018 in our hospital were enrolled in a case-series study. The psoas muscle index (PMI) was calculated as a parameter of muscle mass based on computer tomography (CT) images of the psoas muscle. PMIs were evaluated at 4 time points (TPs): TP1, at the diagnosis of neuroblastoma; TP2, after the first cycle of chemotherapy; TP3, after the third cycle of chemotherapy; and TP4, at the end of the induction chemotherapy. PMI recovery was defined as an increase in PMI between TP2 and TP4. The mean PMI decreased by 15% between TP1 and TP2 (TP1 7.09 ± 0.99 vs. TP2 6.01 ± 0.98, P < 0.001) and by 10% between TP1 and TP4 (TP1 7.09 vs. TP4 6.35, P = 0.004). PMI recovery between TP1 and TP2 was observed in 7 (29%) patients. The median age of patients with PMI recovery was significantly lower (2 vs. 4 years, P = 0.028), and the proportion of boys was significantly higher in patients with PMI recovery (100% vs. 41%, P = 0.017).Conclusion: This study demonstrated that prominent PMI reduction occurs during the early time of chemotherapy, and a younger age and male sex may be predictive factors for PMI recovery. What is Known: • Sarcopenia is a common disorder in elderly people. • Several causes and risk factors have been reported in adults. • Children with previous hematological malignancies have decreased physical activity. What is New: • Prominent muscle mass loss was observed early in children with high-risk neuroblastoma during chemotherapy. • Age and sex were found to be potentially associated with muscle mass recovery.
Collapse
Affiliation(s)
- Natsumi Nakamura
- Pediatric Emergency Department, Hyogo Prefectural Amagasaki General Medical Center, Higashi-Naniwacho 2-17-77, Amagasaki, 660-8550, Japan
| | - Kenji Kishimoto
- Department of Hematology & Oncology, Children's Cancer Center, Kobe Children's Hospital, Minatojima-Minamimachi 1-6-7, Chuo-ku, Kobe, 650-0047, Japan.
| | - Toshiaki Ishida
- Department of Hematology & Oncology, Children's Cancer Center, Kobe Children's Hospital, Minatojima-Minamimachi 1-6-7, Chuo-ku, Kobe, 650-0047, Japan
| | - Sayaka Nakamura
- Department of Hematology & Oncology, Children's Cancer Center, Kobe Children's Hospital, Minatojima-Minamimachi 1-6-7, Chuo-ku, Kobe, 650-0047, Japan
| | - Akihiro Tamura
- Department of Hematology & Oncology, Children's Cancer Center, Kobe Children's Hospital, Minatojima-Minamimachi 1-6-7, Chuo-ku, Kobe, 650-0047, Japan
| | - Aiko Kozaki
- Department of Hematology & Oncology, Children's Cancer Center, Kobe Children's Hospital, Minatojima-Minamimachi 1-6-7, Chuo-ku, Kobe, 650-0047, Japan
| | - Atsuro Saito
- Department of Hematology & Oncology, Children's Cancer Center, Kobe Children's Hospital, Minatojima-Minamimachi 1-6-7, Chuo-ku, Kobe, 650-0047, Japan
| | - Daiichiro Hasegawa
- Department of Hematology & Oncology, Children's Cancer Center, Kobe Children's Hospital, Minatojima-Minamimachi 1-6-7, Chuo-ku, Kobe, 650-0047, Japan
| | - Yoshiyuki Kosaka
- Department of Hematology & Oncology, Children's Cancer Center, Kobe Children's Hospital, Minatojima-Minamimachi 1-6-7, Chuo-ku, Kobe, 650-0047, Japan
| |
Collapse
|
102
|
|
103
|
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: 259] [Impact Index Per Article: 64.8] [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
|
104
|
Ali Q, Bilal MI, Ansari FA, Gondal MUR, Arif A. Neuroblastoma: A Case of Massive Hepatomegaly. Cureus 2021; 13:e16731. [PMID: 34513362 PMCID: PMC8412194 DOI: 10.7759/cureus.16731] [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] [Accepted: 07/29/2021] [Indexed: 11/30/2022] Open
Abstract
Neuroblastoma is the most common embryonal tumor of childhood and has a variable presentation. Stage 4S neuroblastoma, described as a localized primary tumor in an infant with metastasis to skin, liver, or bone marrow, is an exception to the poor prognosis seen in widespread metastasis of neuroblastoma. Survival in infants with this stage of the disease is over 90%. Stage 4S with massive liver involvement, however, confers a poor prognosis. We need more research on the optimum treatment modality for patients with Stage 4S disease and massive hepatomegaly to improve patient outcomes.
Collapse
Affiliation(s)
- Qamar Ali
- Paediatrics, Shifa International Hospital Islamabad, Islamabad, PAK
| | | | - Fawwad A Ansari
- Internal Medicine, Shifa International Hospital Islamabad, Islamabad, PAK
| | | | - Adnan Arif
- Diagnostic Radiology, Shifa International Hospital Islamabad, Islamabad, PAK
| |
Collapse
|
105
|
Dijkstra S, Kraal KCJM, Tytgat GAM, van Noesel MM, Wijnen MHWA, Hoogerbrugge PM. Use of quality indicators in neuroblastoma treatment: A feasibility assessment. Pediatr Blood Cancer 2021; 68:e28301. [PMID: 32735384 DOI: 10.1002/pbc.28301] [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: 12/15/2019] [Revised: 03/15/2020] [Accepted: 03/16/2020] [Indexed: 11/08/2022]
Abstract
BACKGROUND Quality indicators (QIs) may be used to monitor the quality of neuroblastoma (NBL) care during treatment, in addition to survival and treatment toxicity, which can only be evaluated in the years after treatment. The present study aimed to assess the feasibility of a new set of indicators for the quality of NBL therapy. PROCEDURE Seven QIs have been proposed based on literature and consensus of experts: (a) duration of complete diagnostic work-up, (b) prescription of thyroid prophylaxis before metaiodobenzylguanidine imaging, (c) treatment intensity, (d) use of tumor board meetings, (e) number of outpatient visits and sedation procedures during follow-up, (f) protocolled follow-up, and (g) required apheresis sessions. A retrospective data analysis from October 2014 to November 2017 including all patients with NBL in the centralized Princess Máxima Center in the Netherlands was performed to assess these parameters and determine practicality of measurement. RESULTS A total number of 72 patients (aged between 2 weeks and 15 years) were analyzed. Adherence to all QIs could be determined for all eligible patients using their electronic medical records. Three indicators were compared over time, and an increase in adherence was observed. CONCLUSIONS Assessment of QIs in neuroblastoma treatment is feasible. Seven new QIs were found to be feasible to measure and showed improvement over time for three indicators. Monitoring of these QIs during treatment may provide tools for quality improvement activities and comparisons of treatment quality over time or between centers. Further study is required to investigate their association with long-term outcomes.
Collapse
Affiliation(s)
- Suzan Dijkstra
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Kathelijne C J M Kraal
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Godelieve A M Tytgat
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Max M van Noesel
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Marc H W A Wijnen
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Peter M Hoogerbrugge
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| |
Collapse
|
106
|
Imaging for Staging of Pediatric Abdominal Tumors: An Update, From the AJR Special Series on Cancer Staging. AJR Am J Roentgenol 2021; 217:786-799. [PMID: 33825502 DOI: 10.2214/ajr.20.25310] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The three most common pediatric solid tumors of the abdomen are neuroblastoma, Wilms tumor, and hepatoblastoma. These embryonal tumors most commonly present in the first decade of life. Each tumor has unique imaging findings, including locoregional presentation and patterns of distant spread. Neuroblastoma, Wilms tumor, and hepatoblastoma have unique staging systems that rely heavily on imaging and influence surgical and oncologic management. The staging systems include image-defined risk factors for neuroblastoma, the Children's Oncology Group staging system for Wilms tumor, and the pretreatment extent of tumor system (PRETEXT) for hepatoblastoma. It is important for radiologists to be aware of these staging systems to optimize image acquisition and interpretation. This article provides a practical and clinically oriented approach to the role of imaging in the staging of these common embryonal tumors of childhood. The selection among imaging modalities, key findings for determining tumor stage, and the role of imaging in posttreatment response evaluation and surveil-lance are discussed. Recent updates to the relevant staging systems are highlighted with attention to imaging findings of particular prognostic importance. The information presented will help radiologists tailor the imaging approach to the individual patient and guide optimal oncologic management.
Collapse
|
107
|
Garaventa A, Poetschger U, Valteau-Couanet D, Luksch R, Castel V, Elliott M, Ash S, Chan GCF, Laureys G, Beck-Popovic M, Vettenranta K, Balwierz W, Schroeder H, Owens C, Cesen M, Papadakis V, Trahair T, Schleiermacher G, Ambros P, Sorrentino S, Pearson ADJ, Ladenstein RL. Randomized Trial of Two Induction Therapy Regimens for High-Risk Neuroblastoma: HR-NBL1.5 International Society of Pediatric Oncology European Neuroblastoma Group Study. J Clin Oncol 2021; 39:2552-2563. [PMID: 34152804 DOI: 10.1200/jco.20.03144] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 04/07/2021] [Accepted: 04/22/2021] [Indexed: 02/06/2023] Open
Abstract
PURPOSE Induction therapy is a critical component of the therapy of high-risk neuroblastoma. We aimed to assess if the Memorial Sloan Kettering Cancer Center (MSKCC) N5 induction regimen (MSKCC-N5) would improve metastatic complete response (mCR) rate and 3-year event-free survival (EFS) compared with rapid COJEC (rCOJEC; cisplatin [C], vincristine [O], carboplatin [J], etoposide [E], and cyclophosphamide [C]). PATIENTS AND METHODS Patients (age 1-20 years) with stage 4 neuroblastoma or stage 4/4s aged < 1 year with MYCN amplification were eligible for random assignment to rCOJEC or MSKCC-N5. Random assignment was stratified according to national group and metastatic sites. Following induction, therapy comprised primary tumor resection, high-dose busulfan and melphalan, radiotherapy to the primary tumor site, and isotretinoin with ch14.18/CHO (dinutuximab beta) antibody with or without interleukin-2 immunotherapy. The primary end points were mCR rate and 3-year EFS. RESULTS A total of six hundred thirty patients were randomly assigned to receive rCOJEC (n = 313) or MSKCC-N5 (n = 317). Median age at diagnosis was 3.2 years (range, 1 month to 20 years), and 16 were younger than 1 year of age with MYCN amplification. mCR rate following rCOJEC induction (32%, 86/272 evaluable patients) was not significantly different from 35% (99/281) with MSKCC-N5 (P = .368), and 3-year EFS was 44% ± 3% for rCOJEC compared with 47% ± 3% for MSKCC-N5 (P = .527). Three-year overall survival was 60% ± 3% for rCOJEC compared with 65% ± 3% for MSKCC-N5 (P = .379). Toxic death rates with both regimens were 1%. However, nonhematologic CTC grade 3 and 4 toxicities were higher with MSKCC-N5: 68% (193/283) versus 48% (129/268) (P < .001); infection 35% versus 25% (P = .011); stomatitis 25% versus 3% (P < .001); nausea and vomiting 17% versus 7% (P < .001); and diarrhea 7% versus 3% (P = .011). CONCLUSION No difference in outcome was observed between rCOJEC and MSKCC-N5; however, acute toxicity was less with rCOJEC, and therefore rCOJEC is the preferred induction regimen for International Society of Pediatric Oncology European Neuroblastoma Group.
Collapse
Affiliation(s)
| | | | | | - Roberto Luksch
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Victoria Castel
- Pediatric Oncology Unit, Hospital Universitari I Politecnic La Fe, Valencia, Spain
| | - Martin Elliott
- Leeds Teaching Hospitals, NHS Trust, Leeds, United Kingdom
| | - Shifra Ash
- Ruth Rappaport Children's Hospital, Rambam Health Care Campus, Haifa, Israel
| | - Godfrey C F Chan
- University of Hong Kong and Hong Kong Children's Hospital, Hong Kong SAR, China
| | | | | | - Kim Vettenranta
- Children's Hospital, University of Helsinki, Helsinki, Finland
| | | | - Henrik Schroeder
- Department of Paediatrics, University Hospital of Aarhus, Denmark
| | | | | | | | | | | | - Peter Ambros
- Children's Cancer Research Institute, Vienna, Austria
| | | | - Andrew D J Pearson
- Retired. Institute of Cancer Research and Royal Marsden Hospital, Sutton, United Kingdom
| | - Ruth Lydia Ladenstein
- Department of Paediatrics, St Anna Children's Hospital and Children's Cancer Research Institute, Medical University, Vienna, Austria
| |
Collapse
|
108
|
Berthold F, Rosswog C, Christiansen H, Frühwald M, Hemstedt N, Klingebiel T, Fröhlich B, Schilling FH, Schmid I, Simon T, Hero B, Fischer M, Ernst A. Clinical and molecular characterization of patients with stage 4(M) neuroblastoma aged less than 18 months without MYCN amplification. Pediatr Blood Cancer 2021; 68:e29038. [PMID: 33826231 DOI: 10.1002/pbc.29038] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/23/2021] [Accepted: 03/13/2021] [Indexed: 11/06/2022]
Abstract
INTRODUCTION The survival of children with stage 4(M) neuroblastoma without MYCN amplification and below the age of 18 months is considered better than the still dismal outcome of older high-risk neuroblastoma patients. This study analyzes the impact of clinical and molecular characteristics on the long-term outcome. PATIENTS AND METHODS Clinical presentation, survival, and recurrence patterns of patients enrolled onto trials NB90, NB97, and NB2004 were retrospectively analyzed. Gene expression signatures based on RNA microarrays (TH10) were investigated if tumor material was available. RESULTS Between 1990 and 2015, 177 patients with stage 4(M) MYCN nonamplified neuroblastoma aged less than 18 months at diagnosis were eligible. After a median follow-up of 9.7 years (IQR 5.0, 13.4), the proportions of 10-year event-free survival (EFS) and overall survival (OS) were 73% (95% confidence interval [CI] 67-79%) and 86% (95% CI 80-92%), respectively. Of the 27 neuroblastoma recurrences, 44% occurred in more than one site. Four additional patients presented histologically mature ganglioneuroma at recurrence. Six patients developed a secondary malignancy. The secondary 5-year EFS and OS of the 27 patients with neuroblastoma recurrence were 44% and 59%, respectively. TH10 gene expression signature was not prognostically predictive in the investigated subcohort. CONCLUSION The outcome of patients with stage 4(M) neuroblastoma aged less than 18 months is favorable when treated with high-risk or otherwise intensive therapy. The development of secondary malignancies and the potential of maturation to ganglioneuroma call for a controlled stepwise reduction of treatment intensity.
Collapse
Affiliation(s)
- Frank Berthold
- Department of Pediatric Oncology and Hematology, University of Cologne, Cologne, Germany
| | - Carolina Rosswog
- Department of Experimental Pediatric Oncology, University of Cologne, Cologne, Germany.,Center for Molecular Medicine, Medical Faculty, University of Cologne, Cologne, Germany
| | - Holger Christiansen
- Department of Pediatric Oncology and Hematology, University of Leipzig, Leipzig, Germany
| | - Michael Frühwald
- Swabian Children's Cancer Center, Children's Hospital, University Hospital Augsburg, Augsburg, Germany
| | - Nadine Hemstedt
- Department of Experimental Pediatric Oncology, University of Cologne, Cologne, Germany.,Center for Molecular Medicine, Medical Faculty, University of Cologne, Cologne, Germany
| | - Thomas Klingebiel
- Department of Children and Adolescents, University Hospital, Goethe University Frankfurt (Main), Frankfurt, Germany
| | - Birgit Fröhlich
- Department of Pediatric Oncology and Hematology, University of Munster, Munster, Germany
| | - Freimut H Schilling
- Department of Pediatric Oncology and Hematology, Olgahospital Stuttgart, Stuttgart, Germany
| | - Irene Schmid
- Department of Pediatric Hematology and Oncology and Hematology, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Thorsten Simon
- Department of Pediatric Oncology and Hematology, University of Cologne, Cologne, Germany
| | - Barbara Hero
- Department of Pediatric Oncology and Hematology, University of Cologne, Cologne, Germany
| | - Matthias Fischer
- Department of Experimental Pediatric Oncology, University of Cologne, Cologne, Germany.,Center for Molecular Medicine, Medical Faculty, University of Cologne, Cologne, Germany
| | - Angela Ernst
- Institute of Medical Statistics and Computational Biology, University of Cologne, Cologne, Germany
| |
Collapse
|
109
|
Biko DM, Lichtenberger JP, Rapp JB, Khwaja A, Huppmann AR, Chung EM. Mediastinal Masses in Children: Radiologic-Pathologic Correlation. Radiographics 2021; 41:1186-1207. [PMID: 34086496 DOI: 10.1148/rg.2021200180] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Most pediatric masses in the chest are located in the mediastinum. These masses are often initially detected incidentally on chest radiographs in asymptomatic children, although some patients may present with respiratory symptoms. At chest radiography, the mediastinum has been anatomically divided into anterior, middle, and posterior compartments. However, with the International Thymic Malignancy Interest Group classification scheme, which is based on cross-sectional imaging findings, the mediastinum is divided into prevascular, visceral, and paravertebral compartments. In the prevascular compartment, tumors of thymic origin, lymphomas, germ cell tumors, and vascular tumors are encountered. In the visceral compartment, lymphadenopathy and masses related to the foregut are seen. In the paravertebral compartment, neurogenic tumors are most common. Using the anatomic location in combination with knowledge of the imaging and pathologic features of pediatric mediastinal masses aids in accurate diagnosis of these masses to guide treatment and management decisions. An invited commentary by Lee and Winant is available online. ©RSNA, 2021.
Collapse
Affiliation(s)
- David M Biko
- From the Pediatric Radiology Section (D.M.B., E.M.C.) and Thoracic Radiology Section (J.P.L.), American Institute for Radiologic Pathology, Silver Spring, Md; Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104 (D.M.B., J.B.R., A.K.); Department of Radiology, George Washington University, Washington, DC (J.P.L.); Department of Biomedical Sciences, University of South Carolina School of Medicine, Greenville, SC (A.R.H.); and Department of Radiology, Nationwide Children's Hospital, Columbus, Ohio (E.M.C)
| | - John P Lichtenberger
- From the Pediatric Radiology Section (D.M.B., E.M.C.) and Thoracic Radiology Section (J.P.L.), American Institute for Radiologic Pathology, Silver Spring, Md; Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104 (D.M.B., J.B.R., A.K.); Department of Radiology, George Washington University, Washington, DC (J.P.L.); Department of Biomedical Sciences, University of South Carolina School of Medicine, Greenville, SC (A.R.H.); and Department of Radiology, Nationwide Children's Hospital, Columbus, Ohio (E.M.C)
| | - Jordan B Rapp
- From the Pediatric Radiology Section (D.M.B., E.M.C.) and Thoracic Radiology Section (J.P.L.), American Institute for Radiologic Pathology, Silver Spring, Md; Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104 (D.M.B., J.B.R., A.K.); Department of Radiology, George Washington University, Washington, DC (J.P.L.); Department of Biomedical Sciences, University of South Carolina School of Medicine, Greenville, SC (A.R.H.); and Department of Radiology, Nationwide Children's Hospital, Columbus, Ohio (E.M.C)
| | - Asef Khwaja
- From the Pediatric Radiology Section (D.M.B., E.M.C.) and Thoracic Radiology Section (J.P.L.), American Institute for Radiologic Pathology, Silver Spring, Md; Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104 (D.M.B., J.B.R., A.K.); Department of Radiology, George Washington University, Washington, DC (J.P.L.); Department of Biomedical Sciences, University of South Carolina School of Medicine, Greenville, SC (A.R.H.); and Department of Radiology, Nationwide Children's Hospital, Columbus, Ohio (E.M.C)
| | - Alison R Huppmann
- From the Pediatric Radiology Section (D.M.B., E.M.C.) and Thoracic Radiology Section (J.P.L.), American Institute for Radiologic Pathology, Silver Spring, Md; Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104 (D.M.B., J.B.R., A.K.); Department of Radiology, George Washington University, Washington, DC (J.P.L.); Department of Biomedical Sciences, University of South Carolina School of Medicine, Greenville, SC (A.R.H.); and Department of Radiology, Nationwide Children's Hospital, Columbus, Ohio (E.M.C)
| | - Ellen M Chung
- From the Pediatric Radiology Section (D.M.B., E.M.C.) and Thoracic Radiology Section (J.P.L.), American Institute for Radiologic Pathology, Silver Spring, Md; Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104 (D.M.B., J.B.R., A.K.); Department of Radiology, George Washington University, Washington, DC (J.P.L.); Department of Biomedical Sciences, University of South Carolina School of Medicine, Greenville, SC (A.R.H.); and Department of Radiology, Nationwide Children's Hospital, Columbus, Ohio (E.M.C)
| |
Collapse
|
110
|
Zhang S, Zhang W, Jin M, Sun Q, Zhang Z, Qin H, Su Y, Zhi T, Xie Y, Gao YX, Zhang X, Li L, Liu R, Zhao W, Wang H, Huang D, Ma X. Biological features and clinical outcome in infant neuroblastoma: a multicenter experience in Beijing. Eur J Pediatr 2021; 180:2055-2063. [PMID: 33580827 DOI: 10.1007/s00431-021-03989-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/09/2021] [Accepted: 02/05/2021] [Indexed: 10/22/2022]
Abstract
Neuroblastoma (NB) is the most common extracranial solid tumor in childhood, with 37% of patients diagnosed during infancy. This study is aimed at evaluating the survival outcome in infants diagnosed with neuroblastoma. This was a retrospective cohort study including patients under the age of 12 months with neuroblastoma from four tertiary referral centers in Beijing, China (Beijing Children's Hospital, Beijing Tongren Hospital, Peking University First Hospital, and Capital Institute of Pediatrics). Two hundred and forty-seven infants with neuroblastoma were included (male = 132 and female = 115). 91.1% (n = 225) patients were classified as having low-risk or intermediate-risk disease and 8.9% (n = 22) as having high-risk disease. The most common metastatic site is distant lymph node (n=89, 36.0%), followed by liver (n=57, 23.1%), bone (n=42, 17.0%), bone marrow (n=37, 15.0%), soft tissue (n=25, 10%), and central nervous system (n=4, 1.6%). MYCN amplification was present in 9.9% of tumor samples, chromosome 1p or 11q aberration in 14%. Treatment involved surgery alone in 9.7% of patients (n=24, all with low-risk disease), surgery followed by adjuvant chemotherapy in 50.2% (n=124), neoadjuvant chemotherapy followed by surgery in 40.1% (n=97), and chemotherapy alone in 0.8% (n=2). 4.9% (n=12) patients died, and the major cause of death is disease progression. Three-year event-free and overall survival were 91.6%±2.1% and 97.4%±1.1%, respectively, in patients with low- or intermediate-risk disease, and 58.7%±11.5% and 63.6%±11.2%, respectively, in those with high-risk disease.Conclusions: Infants with neuroblastoma achieve a reasonable clinical outcome when treated with surgery with or without chemotherapy using a risk-stratified approach in China. Such information will facilitate counseling, therapeutic decision-making, and development of adapted standard-of-care guidelines for future patients in the country. What is Known: • NB is a disease of infancy; 37% of patients are diagnosed as infants. • Most children younger than 12 months of age have a good prognosis even in the presence of metastatic disease. What is New: • Infants with neuroblastoma achieve reasonable clinical outcome when treated with surgery with or without chemotherapy using a risk-stratified approach in China. • CNS metastasis in infants with neuroblastoma is very rare at diagnosis and had a worse prognosis than those without metastasis.
Collapse
Affiliation(s)
- Shihan Zhang
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics, Ministry of Education, Key Laboratory of Major Diseases in Children, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Weiling Zhang
- Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Mei Jin
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics, Ministry of Education, Key Laboratory of Major Diseases in Children, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Qing Sun
- Peking University First Hospital, Beijing, 100034, China
| | - Zhaoxia Zhang
- Capital Institute of Pediatrics, Beijing, 100020, China
| | - Hong Qin
- Department of Surgical Oncology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Yan Su
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics, Ministry of Education, Key Laboratory of Major Diseases in Children, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Tian Zhi
- Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Yao Xie
- Peking University First Hospital, Beijing, 100034, China
| | - Yang Xu Gao
- Peking University First Hospital, Beijing, 100034, China
| | - Xiaolun Zhang
- Capital Institute of Pediatrics, Beijing, 100020, China
| | - Long Li
- Capital Institute of Pediatrics, Beijing, 100020, China
| | - Rong Liu
- Capital Institute of Pediatrics, Beijing, 100020, China.
| | - Weihong Zhao
- Peking University First Hospital, Beijing, 100034, China.
| | - Huanmin Wang
- Department of Surgical Oncology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China.
| | - Dongsheng Huang
- Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| | - Xiaoli Ma
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics, Ministry of Education, Key Laboratory of Major Diseases in Children, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China.
| |
Collapse
|
111
|
Dhali A, Ray S, Dhali GK, Ghosh R, Sarkar A. Duodenal Ganglioneuroma: A Rare Tumor Causing Upper Gastrointestinal Bleed. Surg J (N Y) 2021; 7:e255-e258. [PMID: 34541318 PMCID: PMC8440055 DOI: 10.1055/s-0041-1735644] [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: 04/25/2021] [Accepted: 07/19/2021] [Indexed: 12/03/2022] Open
Abstract
Neuroblastic tumors (NTs) include neuroblastoma, ganglioneuroblastoma, and ganglioneuroma (GN). They are very rare in adults. The Surveillance, Epidemiology, and End Results identified 144 patients ≥20 years old at diagnosis (6.1%) from 1973 to 2002. GNs account for 14% of all localized NT. Since 1957, a total of four cases of GN of the duodenum have been reported. We report a novel case of GN of the periampullary region in the duodenum in a 41-year-old man presenting with chronic upper gastrointestinal bleed. Given the rarity of GNs in this age group and the nonspecificity of radiological features, this diagnosis is often missed until histopathology is done. This may negatively affect the prognosis of an otherwise well-prognosticated disease.
Collapse
Affiliation(s)
- Arkadeep Dhali
- Department of GI Surgery, School of Digestive and Liver Diseases, Institute of Postgraduate Medical Education and Research, Kolkata, West Bengal, India
| | - Sukanta Ray
- Department of GI Surgery, School of Digestive and Liver Diseases, Institute of Postgraduate Medical Education and Research, Kolkata, West Bengal, India
| | - Gopal Krishna Dhali
- Department of Gastroenterology, School of Digestive and Liver Diseases, Institute of Postgraduate Medical Education and Research, Kolkata, West Bengal, India
| | - Ranajoy Ghosh
- Department of GI Pathology, School of Digestive and Liver Diseases, Institute of Postgraduate Medical Education and Research, Kolkata, West Bengal, India
| | - Avik Sarkar
- Department of GI Radiology, School of Digestive and Liver Diseases, Institute of Postgraduate Medical Education and Research, Kolkata, West Bengal, India
| |
Collapse
|
112
|
Guo YF, Duan JJ, Wang J, Li L, Wang D, Liu XZ, Yang J, Zhang HR, Lv J, Yang YJ, Yang ZY, Cai J, Liao XM, Tang T, Huang TT, Wu F, Yang XY, Wen Q, Bian XW, Yu SC. Inhibition of the ALDH18A1-MYCN positive feedback loop attenuates MYCN-amplified neuroblastoma growth. Sci Transl Med 2021; 12:12/531/eaax8694. [PMID: 32075946 DOI: 10.1126/scitranslmed.aax8694] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 01/28/2020] [Indexed: 12/14/2022]
Abstract
MYCN-amplified neuroblastoma (NB) is characterized by poor prognosis, and directly targeting MYCN has proven challenging. Here, we showed that aldehyde dehydrogenase family 18 member A1 (ALDH18A1) exerts profound impacts on the proliferation, self-renewal, and tumorigenicity of NB cells and is a potential risk factor in patients with NB, especially those with MYCN amplification. Mechanistic studies revealed that ALDH18A1 could both transcriptionally and posttranscriptionally regulate MYCN expression, with MYCN reciprocally transactivating ALDH18A1 and thus forming a positive feedback loop. Using molecular docking and screening, we identified an ALDH18A1-specific inhibitor, YG1702, and demonstrated that pharmacological inhibition of ALDH18A1 was sufficient to induce a less proliferative phenotype and confer tumor regression and prolonged survival in NB xenograft models, providing therapeutic insights into the disruption of this reciprocal regulatory loop in MYCN-amplified NB.
Collapse
Affiliation(s)
- Yu-Feng Guo
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jiang-Jie Duan
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jun Wang
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Lin Li
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Di Wang
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xun-Zhou Liu
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jing Yang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Hua-Rong Zhang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jing Lv
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yong-Jun Yang
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Ze-Yu Yang
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jiao Cai
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xue-Mei Liao
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Tao Tang
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Ting-Ting Huang
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Feng Wu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xian-Yan Yang
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Qian Wen
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xiu-Wu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China. .,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Shi-Cang Yu
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China. .,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Key Laboratory of Tumor Immunopathology of the Ministry of Education, Third Military Medical University (Army Medical University), Chongqing 400038, China
| |
Collapse
|
113
|
Choi JH, Ro JY. Mediastinal neuroblastoma, ganglioneuroblastoma, and ganglioneuroma: Pathology review and diagnostic approach. Semin Diagn Pathol 2021; 39:120-130. [PMID: 34167847 DOI: 10.1053/j.semdp.2021.06.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 06/10/2021] [Indexed: 12/13/2022]
Abstract
Neuroblastic tumors are a group of tumors of the sympathetic ganglia and adrenal medulla that derive from primordial neural crest cells. These tumors include neuroblastoma, intermixed ganglioneuroblastoma, nodular ganglioneuroblastoma, and ganglioneuroma. Neuroblastomas are the most common extracranial solid tumor arising in childhood and may occur in different anatomic sites. Neuroblastic tumors are common mesenchymal tumors of the mediastinum. Herein, we describe advances in our understanding of neuroblastic tumor biology. Pathologists should be aware of diagnostic challenges associated with these tumors to ensure correct histologic diagnosis and appropriate clinical management. We describe updated mediastinal neuroblastic tumor pathology, focusing on morphological, immunohistochemical, and molecular features and differential diagnoses.
Collapse
Affiliation(s)
- Joon Hyuk Choi
- Department of Pathology, Yeungnam University College of Medicine, 170 Hyeonchung-ro, Namgu, Daegu, 42415, South Korea.
| | - Jae Y Ro
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Weill Medical College of Cornell University, Houston, TX, 77030, USA
| |
Collapse
|
114
|
Gautier M, Thirant C, Delattre O, Janoueix-Lerosey I. Plasticity in Neuroblastoma Cell Identity Defines a Noradrenergic-to-Mesenchymal Transition (NMT). Cancers (Basel) 2021; 13:2904. [PMID: 34200747 PMCID: PMC8230375 DOI: 10.3390/cancers13122904] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/05/2021] [Accepted: 06/07/2021] [Indexed: 12/14/2022] Open
Abstract
Neuroblastoma, a pediatric cancer of the peripheral sympathetic nervous system, is characterized by an important clinical heterogeneity, and high-risk tumors are associated with a poor overall survival. Neuroblastoma cells may present with diverse morphological and biochemical properties in vitro, and seminal observations suggested that interconversion between two phenotypes called N-type and S-type may occur. In 2017, two main studies provided novel insights into these subtypes through the characterization of the transcriptomic and epigenetic landscapes of a panel of neuroblastoma cell lines. In this review, we focus on the available data that define neuroblastoma cell identity and propose to use the term noradrenergic (NOR) and mesenchymal (MES) to refer to these identities. We also address the question of transdifferentiation between both states and suggest that the plasticity between the NOR identity and the MES identity defines a noradrenergic-to-mesenchymal transition, reminiscent of but different from the well-established epithelial-to-mesenchymal transition.
Collapse
Affiliation(s)
- Margot Gautier
- Institut Curie, PSL Research University, Inserm U830, Equipe Labellisée Ligue Contre le Cancer, 75005 Paris, France; (M.G.); (C.T.); (O.D.)
- SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, 75005 Paris, France
| | - Cécile Thirant
- Institut Curie, PSL Research University, Inserm U830, Equipe Labellisée Ligue Contre le Cancer, 75005 Paris, France; (M.G.); (C.T.); (O.D.)
- SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, 75005 Paris, France
| | - Olivier Delattre
- Institut Curie, PSL Research University, Inserm U830, Equipe Labellisée Ligue Contre le Cancer, 75005 Paris, France; (M.G.); (C.T.); (O.D.)
- SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, 75005 Paris, France
| | - Isabelle Janoueix-Lerosey
- Institut Curie, PSL Research University, Inserm U830, Equipe Labellisée Ligue Contre le Cancer, 75005 Paris, France; (M.G.); (C.T.); (O.D.)
- SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, 75005 Paris, France
| |
Collapse
|
115
|
Wang Y, Guan E, Li D, Sun L. miRNA-34a-5p regulates progression of neuroblastoma via modulating the Wnt/β-catenin signaling pathway by targeting SOX4. Medicine (Baltimore) 2021; 100:e25827. [PMID: 34011046 PMCID: PMC8137035 DOI: 10.1097/md.0000000000025827] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 04/06/2021] [Indexed: 01/05/2023] Open
Abstract
Neuroblastoma is an embryonal tumor of the autonomic nervous system with poor prognosis in children. In present study, we demonstrated the relationship of miRNA-34a-5p in the regulating of the Wnt/β-catenin signaling pathway by targeting SRY-related HMG-box (SOX4)Reverse transcription-quantitative PCR was used to detect the expression levels of miRNA-34a-5p and SoX4. Western blotting was performed to assess the protein expression levels of SoX4, Wnt, MMP9, Bax, and Bcl-2. The proliferation, apoptosis, migration and invasion of neuroblastoma cells were determined using MTT, flow cytometry and Transwell assays.In this study, we sought to investigate the role of miRNA-34a-5p on neuroblastoma and the possible molecular mechanism. We had performed in-vitro and in-vivo experiments to evaluate the effects of miRNA-34a-5p on neuroblastoma cell proliferation and invasion by altering its expression level via cell transfection. On the basis of our study, miRNA-34a-5p showed decreased expression levels in neuroblastoma. Subsequently, we manipulated miRNA-34a-5p expression through cell transfection and observed abnormal expression of β-catenin as well as the downstream targets of the Wnt/β-catenin pathway in neuroblastoma cells. With all these evidences, we determined that miRNA-34a-5p regulated Wnt/β-catenin pathway by targeting SOX4.In conclusion, our study demonstrates that miRNA-34a-5p can inhibit the over-activation of the Wnt/β-catenin signaling pathway via targeting SOX4 and further regulate proliferation, invasion of neuroblastoma cells.
Collapse
Affiliation(s)
- Yue Wang
- Qingdao Municipal Hospital Affiliated to Qingdao University
| | - Enqing Guan
- Qingdao Municipal Hospital Affiliated to Qingdao University
| | - Dehua Li
- Qingdao Women and Children's Hospital
| | - Lirong Sun
- The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, P.R. China
| |
Collapse
|
116
|
Dirks WG, Capes-Davis A, Eberth S, Fähnrich S, Wilting J, Nagel S, Steenpass L, Becker J. Cross contamination meets misclassification: Awakening of CHP-100 from sleeping beauty sleep-A reviewed model for Ewing's sarcoma. Int J Cancer 2021; 148:2608-2613. [PMID: 33460449 DOI: 10.1002/ijc.33474] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/09/2020] [Accepted: 01/04/2021] [Indexed: 11/11/2022]
Abstract
A human cell line of neuroblastic tissue, which was believed to have been lost to science due to its unavailability in public repositories, is revived and reclassified. In the 1970s, a triple set of neuroblastoma (NB) cell lines became available for research as MYCN-amplified vs nonamplified models (CHP-126/-134 and CHP-100, respectively). Confusingly, CHP-100 was used in subsequent years as a model for NB and, since the 1990s, as a model for neuroepithelioma and later as a model for Ewing's sarcoma (ES), which inevitably led to non-reproducible results. A deposit at a bioresource center revealed that globally available stocks of CHP-100 were identical to the prominent NB cell line IMR-32 and CHP-100 was included into the list of misidentified cell lines. Now we report on the rediscovery of an authentic CHP-100 cell line and provide evidence of incorrect classification during establishment. We show that CHP-100 cells carry a t(11;22)(q24;q12) type II EWSR1-FLI1 fusion and identify it as a classic ES. Although the question of whether CHP-100 was a virtual and never existing cell line from the beginning is now clarified, the results of all relevant publications should be considered questionable. Neither the time of the cross-contamination event with IMR-32 is known nor was the final classification as a model for Ewing family of tumors available with an associated short tandem repeat profile. After a long road of errors and confusion, authentic CHP-100 is now characterized as a type II EWSR1-FLI1 fusion model 44 years after its establishment.
Collapse
Affiliation(s)
- Wilhelm Gerhard Dirks
- Leibniz-Institute DSMZ - Deutsche Sammlung von Mikroorganismen und Zellkulturen, Braunschweig, Germany
| | - Amanda Capes-Davis
- Cell Bank Australia, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
| | - Sonja Eberth
- Leibniz-Institute DSMZ - Deutsche Sammlung von Mikroorganismen und Zellkulturen, Braunschweig, Germany
| | - Silke Fähnrich
- Leibniz-Institute DSMZ - Deutsche Sammlung von Mikroorganismen und Zellkulturen, Braunschweig, Germany
| | - Jörg Wilting
- Department of Anatomy and Cell Biology, University Medical School Göttingen, Göttingen, Germany
| | - Stefan Nagel
- Leibniz-Institute DSMZ - Deutsche Sammlung von Mikroorganismen und Zellkulturen, Braunschweig, Germany
| | - Laura Steenpass
- Leibniz-Institute DSMZ - Deutsche Sammlung von Mikroorganismen und Zellkulturen, Braunschweig, Germany
| | - Jürgen Becker
- Department of Anatomy and Cell Biology, University Medical School Göttingen, Göttingen, Germany
| |
Collapse
|
117
|
Karkoska K, Ricci K, VandenHeuvel K, Trout AT, Smith EA, Kotagal M, Weiss B. Metastatic neuroblastoma masquerading as infantile hemangioma in a 4-month-old child. Pediatr Blood Cancer 2021; 68:e28920. [PMID: 33644927 DOI: 10.1002/pbc.28920] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/18/2020] [Accepted: 01/10/2021] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Diffuse liver lesions in an infant have a differential diagnosis including infantile hemangioma (IH), which is common in the first year of life, and neuroblastoma (NBL) which presents at a median age of 18 months. RESULTS We describe the case of a 4-month-old girl with a known superficial/deep IH who presented with new axillary nodules and hepatosplenomegaly, initially suspected to reflect IH but later determined to be widely metastatic NBL. CONCLUSION Hepatic IH and metastatic NBL can present similarly. Clinicians must maintain a broad differential when evaluating new findings in a patient with previously diagnosed IH.
Collapse
Affiliation(s)
- Kristine Karkoska
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Kiersten Ricci
- Division of Hematology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Katherine VandenHeuvel
- Department of Pathology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Andrew T Trout
- Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Ethan A Smith
- Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Meera Kotagal
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Brian Weiss
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
118
|
Reichel CA. Rare Diseases of the Oral Cavity, Neck, and Pharynx. Laryngorhinootologie 2021; 100:S1-S24. [PMID: 34352905 PMCID: PMC8432966 DOI: 10.1055/a-1331-2851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Diseases occurring with an incidence of less than 1-10 cases per 10 000 individuals are considered as rare. Currently, between 5 000 and 8 000 rare or orphan diseases are known, every year about 250 rare diseases are newly described. Many of those pathologies concern the head and neck area. In many cases, a long time is required to diagnose an orphan disease. The lives of patients who are affected by those diseases are often determined by medical consultations and inpatient stays. Most orphan diseases are of genetic origin and cannot be cured despite medical progress. However, during the last years, the perception of and the knowledge about rare diseases has increased also due to the fact that publicly available databases have been created and self-help groups have been established which foster the autonomy of affected people. Only recently, innovative technical progress in the field of biogenetics allows individually characterizing the genetic origin of rare diseases in single patients. Based on this, it should be possible in the near future to elaborate tailored treatment concepts for patients suffering from rare diseases in the sense of translational and personalized medicine. This article deals with orphan diseases of the lip, oral cavity, pharynx, and cervical soft tissues depicting these developments. The readers will be provided with a compact overview about selected diseases of these anatomical regions. References to further information for medical staff and affected patients support deeper knowledge and lead to the current state of knowledge in this highly dynamic field.
Collapse
Affiliation(s)
- Christoph A Reichel
- Klinik und Poliklinik für Hals-Nasen-Ohrenheilkunde, KUM-Klinikum, Ludwig-Maximilians-Universität München, München
| |
Collapse
|
119
|
van Heerden J, Esterhuizen TM, Hendricks M, Poole J, Büchner A, Naidu G, du Plessis J, van Emmenes B, Uys R, Hadley GP, Kruger M. Age at diagnosis as a prognostic factor in South African children with neuroblastoma. Pediatr Blood Cancer 2021; 68:e28878. [PMID: 33484106 DOI: 10.1002/pbc.28878] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 12/06/2020] [Accepted: 12/13/2020] [Indexed: 11/11/2022]
Abstract
PURPOSE Low- and middle-income countries (LMICs) reported a higher median age at diagnosis of neuroblastoma (NB) compared to high-income countries. The aim was to determine if the optimal age at diagnosis, which maximizes the difference in overall survival between younger versus older patients in the South African population was similar to the internationally validated 18 months age cut-point. METHODS Four hundred sixty NB patients diagnosed between 2000 and 2016 were included. Receiver operating characteristic (ROC) curves were used to predict potential age cut-point values for overall survival in all risk group classifications. Risk ratios, sensitivity, specificity, and positive and negative predictive values at the specific cut-points were estimated with 95% confidence intervals, and time to mortality by age at the specific cut-points was shown with Kaplan-Meier curves and compared using log-rank tests. RESULTS The median age at diagnosis for the total cohort was 31.9 months (range 0.2-204.7). For high-risk (HR), intermediate-risk, low-risk, and very low-risk patients, the median age at diagnosis was, respectively, 36 months (range 0.4-204.7), 16.8 months (range 0.7-145.1), 14.2 months (range 2.0-143.5), and 8.7 months (range 0.2-75.6). The ROC curves for the total NB cohort (area under the curve [AUC] 0.696; P < .001) and HR (AUC 0.682; P < .001) were analyzed further. The optimal cut-point value for the total cohort was at 19.1 months (sensitivity 59%; specificity 78%). The HR cohort had potential cut-point values identified at 18.4 months age at diagnosis (sensitivity 45%; specificity 87%) and 31.1 months (sensitivity 67%; specificity 62%). The 19.1 months cut-point value in the total cohort and the 18.4 months cut-point value in HR were as useful in predicting overall survival as 18 months age at diagnosis. CONCLUSION The 18 months cut-point value appears to be the appropriate age for prognostic determination, despite the higher median age at diagnosis in South Africa.
Collapse
Affiliation(s)
- Jaques van Heerden
- Department of Paediatric Haematology and Oncology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg Hospital, Cape Town, South Africa.,Paediatric Haematology and Oncology, Department of Paediatrics, Antwerp University Hospital, Edegem, Belgium
| | - Tonya M Esterhuizen
- Division of Epidemiology and Biostatistics, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Marc Hendricks
- Department of Paediatrics and Child Health, Faculty of Health Sciences, Paediatric Haematology and Oncology Service, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa
| | - Janet Poole
- Faculty of Health Sciences, Division of Paediatric Haematology and Oncology, Department of Paediatrics and Child Health, University of the Witwatersrand, Charlotte Maxeke Johannesburg Academic Hospital, Cape Town, South Africa
| | - Ané Büchner
- Paediatric Haematology and Oncology, Department of Paediatrics, University of Pretoria, Steve Biko Academic Hospital, Pretoria, South Africa
| | - Gita Naidu
- Faculty of Health Sciences, Division of Paediatric Haematology and Oncology, Department of Paediatrics and Child Health, University of the Witwatersrand, Chris Hani Baragwanath Academic Hospital, Johannesburg, South Africa
| | - Jan du Plessis
- Department of Paediatrics, Faculty of Health Sciences, University of the Free State, Division of Paediatric Haematology and Oncology, Universitas Hospital, Bloemfontein, South Africa
| | - Barry van Emmenes
- Division of Paediatric Haematology and Oncology Hospital, Department of Paediatrics, Frere Hospital, East London, Eastern Cape, South Africa
| | - Ronelle Uys
- Department of Paediatric Haematology and Oncology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg Hospital, Cape Town, South Africa
| | - G P Hadley
- Department of Paediatric Surgery, Faculty of Health Sciences, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Berea, South Africa
| | - Mariana Kruger
- Department of Paediatric Haematology and Oncology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg Hospital, Cape Town, South Africa
| |
Collapse
|
120
|
Abstract
Surgeons caring for patients with neuroblastoma must be familiar with recent developments in assessing risk. In particular, the Children's Oncology Group, along with major international groups, uses the International Neuroblastoma Risk Group Staging System as a risk assessment tool. Accurate risk determination is essential for optimal surgical therapy. Some tumors like neonatal adrenal neuroblastomas and those in the metastatic category can be observed. Very-low-risk and low-risk neuroblastomas can be treated with surgery alone. Intermediate-risk tumors also often require systemic chemotherapy.
Collapse
Affiliation(s)
- Nikke Croteau
- Department of Surgery, Pediatric Service, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Jed Nuchtern
- Department of Surgery, Baylor College of Medicine, 6701 Fannin Street, Houston, TX 77030, USA
| | - Michael P LaQuaglia
- Department of Surgery, Pediatric Service, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
121
|
Zhou C, Wang Y, He L, Zhu J, Li J, Tang Y, Zhou H, He J, Wu H. Association between NER pathway gene polymorphisms and neuroblastoma risk in an eastern Chinese population. Mol Ther Oncolytics 2021; 20:3-11. [PMID: 33575466 PMCID: PMC7851491 DOI: 10.1016/j.omto.2020.12.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 12/10/2020] [Indexed: 02/07/2023] Open
Abstract
Neuroblastoma is a common childhood malignancy. Nucleotide excision repair (NER) polymorphisms have been shown to influence cancer susceptibility by modifying DNA repair efficiency. To investigate the association of NER gene polymorphisms with neuroblastoma risk, we constructed a three-center case-control study. A total of 19 candidate single-nucleotide polymorphisms (SNPs) in NER genes were analyzed. Odds ratios (ORs) and 95% confidential intervals (CIs) were calculated to evaluate the associations. We identified five independent SNPs that were significantly associated with neuroblastoma risk, including XPA rs1800975 (dominant model: adjusted OR = 0.73, 95% CI = 0.55-0.98, p = 0.033), XPA rs3176752 (recessive model: adjusted OR = 2.78, 95% CI = 1.12-6.91, p = 0.028), XPD rs3810366 (dominant: adjusted OR = 1.44, 95% CI = 1.05-1.97, p = 0.022; recessive: adjusted OR = 1.58, 95% CI = 1.18-2.11, p = 0.002), XPD rs238406 (dominant: adjusted OR = 0.64, 95% CI = 0.48-0.84, p = 0.002; recessive: adjusted OR = 0.67, 95% CI = 0.48-0.94, p = 0.021), and XPG rs2094258 (recessive: adjusted OR = 1.44, 95% CI = 1.03-2.04, p = 0.036). Stratified analysis was carried out. Furthermore, these findings were strengthened by false-positive report probability (FPRP) analysis and expression quantitative trait loci (eQTL) analysis. In conclusion, our study indicates that five SNPs in NER genes are correlated with neuroblastoma susceptibility in the eastern Chinese population, providing novel insight into the genetic underpinnings of neuroblastoma. However, further large-scale studies are required to verify these findings.
Collapse
Affiliation(s)
- Chunlei Zhou
- Department of Pathology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, Jiangsu, China
| | - Yizhen Wang
- Department of Pathology, Anhui Provincial Children’s Hospital, Hefei 230051, Anhui, China
| | - Lili He
- Department of Pathology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, Jiangsu, China
| | - Jinhong Zhu
- Department of Clinical Laboratory, Biobank, Harbin Medical University Cancer Hospital, Harbin 150040, Heilongjiang, China
| | - Jinghang Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Yingzi Tang
- Department of Pathology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, Jiangsu, China
| | - Haixia Zhou
- Department of Hematology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Haiyan Wu
- Department of Pathology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, Jiangsu, China
| |
Collapse
|
122
|
Biegel JM, Dhamdhere M, Gao S, Gowda CP, Kawasawa YI, Spiegelman VS. Inhibition of the mRNA-Binding Protein IGF2BP1 Suppresses Proliferation and Sensitizes Neuroblastoma Cells to Chemotherapeutic Agents. Front Oncol 2021; 11:608816. [PMID: 33796454 PMCID: PMC8008117 DOI: 10.3389/fonc.2021.608816] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 02/02/2021] [Indexed: 12/13/2022] Open
Abstract
Gain at chromosome 17q21 in neuroblastoma is associated with a poor prognosis, independent of MYCN amplification status. Several potential proto-oncogenes have been identified in this region, one of them-insulin-like growth-factor-2 mRNA binding protein (IGF2BP1)-is expressed at high levels in stage 4 tumors, and associated with overall lower patient survival. Here, we demonstrate that down-regulation of IGF2BP1 activity, either by transcript silencing or chemical inhibition, suppresses neuroblastoma cell growth. Furthermore, the combination of IGF2BP1 inhibition along with commonly used chemotherapeutics that broadly affect DNA synthesis, or cyclin-dependent kinase (CDK) inhibitors that disrupt signal transduction, have a synergistic effect on the suppression of neuroblastoma cell proliferation.
Collapse
Affiliation(s)
- Jason M. Biegel
- Division of Hematology and Oncology, Pediatric Department, Penn State College of Medicine, Hershey, PA, United States
| | - Mayura Dhamdhere
- Division of Hematology and Oncology, Pediatric Department, Penn State College of Medicine, Hershey, PA, United States
| | - Shuang Gao
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA, United States
| | - Chethana P. Gowda
- Division of Hematology and Oncology, Pediatric Department, Penn State College of Medicine, Hershey, PA, United States
| | - Yuka Imamura Kawasawa
- Departments of Pharmacology and Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, PA, United States
| | - Vladimir S. Spiegelman
- Division of Hematology and Oncology, Pediatric Department, Penn State College of Medicine, Hershey, PA, United States
| |
Collapse
|
123
|
Abstract
Neuroblastoma accounts for approximately 8% of all pediatric cancers, with 5% diagnosed during the neonatal period. Despite the disproportionate contribution of neuroblastoma to childhood cancer deaths, neonatal neuroblastoma has a favorable prognosis, often with little or no therapy required. Therefore, minimizing therapy and mitigating complications/toxicities are emphasized, including using a watch-and-wait approach for patients at low risk for disease progression/relapse. However, stage MS neuroblastoma exhibits a unique pattern of disseminated disease, can be challenging to manage, and may require early intervention with systemic chemotherapy. In this review, the epidemiology, treatment options, and anticipated outcomes for neonatal neuroblastoma are discussed.
Collapse
Affiliation(s)
- Andrew M Davidoff
- Department of Surgery, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105-3678, USA.
| |
Collapse
|
124
|
Ornell KJ, Mistretta KS, Ralston CQ, Coburn JM. Development of a stacked, porous silk scaffold neuroblastoma model for investigating spatial differences in cell and drug responsiveness. Biomater Sci 2021; 9:1272-1290. [PMID: 33336667 DOI: 10.1039/d0bm01153c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Development of in vitro, preclinical cancer models that contain cell-driven microenvironments remains a challenge. Engineering of millimeter-scale, in vitro tumor models with spatially distinct regions that can be independently assessed to study tumor microenvironments has been limited. Here, we report the use of porous silk scaffolds to generate a high cell density neuroblastoma (NB) model that can spatially recapitulate changes resulting from cell and diffusion driven changes. Using COMSOL modeling, a scaffold holder design that facilitates stacking of thin, 200 μm silk scaffolds into a thick, bulk millimeter-scale tumor model (2, 4, 6, and 8 stacked scaffolds) and supports cell-driven oxygen gradients was developed. Cell-driven oxygen gradients were confirmed through pimonidazole staining. Post-culture, the stacked scaffolds were separated for analysis on a layer-by-layer basis. The analysis of each scaffold layer demonstrated decreasing DNA and increasing expression of hypoxia related genes (VEGF, CAIX, and GLUT1) from the exterior scaffolds to the interior scaffolds. Furthermore, the expression of hypoxia related genes at the interior of the stacks was comparable to that of a single scaffold cultured under 1% O2 and at the exterior of the stacks was comparable to that of a single scaffold cultured under 21% O2. The four-stack scaffold model underwent further evaluation to determine if a hypoxia activated drug, tirapazamine, induced reduced cell viability within the internal stacks (region of reduced oxygen) as compared with the external stacks. Decreased DNA content was observed in the internal stacks as compared to the external stacks when treated with tirapazamine, which suggests the internal scaffold stacks had higher levels of hypoxia than the external scaffolds. This stacked silk scaffold system presents a method for creating a single culture model capable of generating controllable cell-driven microenvironments through different stacks that can be individually assessed and used for drug screening.
Collapse
Affiliation(s)
- Kimberly J Ornell
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, USA.
| | - Katelyn S Mistretta
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, USA.
| | - Coulter Q Ralston
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, USA.
| | - Jeannine M Coburn
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, USA.
| |
Collapse
|
125
|
Nawata A, Izumi R, Harada K, Kurisu H, Shimajiri S, Matsuki Y, Nakayama T. An elderly-onset neuroblastoma concomitant with minimal change nephrotic syndrome: the first autopsy case report. CEN Case Rep 2021; 10:414-421. [PMID: 33595829 DOI: 10.1007/s13730-021-00580-3] [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: 11/08/2020] [Accepted: 02/06/2021] [Indexed: 10/22/2022] Open
Abstract
BACKGROUND Neuroblastoma is a well-known embryonal cancer; however, adult-onset neuroblastomas are rare. The systemic symptoms are related to catecholamine excretion or intraabdominal mass effects. Only two cases of neuroblastoma with nephrotic syndrome have previously been reported. We herein present the first autopsy case of neuroblastoma in an older individual associated with minimal change nephrotic syndrome. CASE PRESENTATION A 63-year-old man was admitted to our hospital for investigation of general fatigue. His renal function was normal and his urine was negative for protein. A computed tomography scan showed a renal tumor and intraabdominal lymph node swelling. Approximately 4 months after admission, he suddenly developed acute renal failure and severe proteinuria, and hemodialysis was instituted. A computed tomography scan revealed an increase in the size of the renal tumor and lymph nodes. He died 1 month later and an autopsy was performed. The tumor exhibited diffuse proliferation of tumor cells with scant cytoplasm, namely small blue cell tumor with rosette formation. As a result of immunohistochemical study, a neuroblastoma was diagnosed. Despite the patient's severe renal failure, most glomeruli showed no remarkable changes. The tubular epithelium exhibited detachment and vacuolation. Electron microscopic study of the glomeruli showed diffuse effacement of the foot processes. These features indicate a diagnosis of minimal change nephrotic syndrome with acute tubular injury. CONCLUSIONS Minimal change nephrotic syndrome is the most common renal manifestation associated with lymphoproliferative malignancies. We here present an extremely rare case of adult-onset neuroblastoma with minimal change nephrotic syndrome.
Collapse
Affiliation(s)
- Aya Nawata
- The Department of Pathology and Oncology, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, Fukuoka, Japan. .,Department of Pathology, Kenwakai Otemachi Hospital, Kitakyushu, Japan.
| | - Ryo Izumi
- Department of Cardiovascular Medicine, Kitakyushu Municipal Medical Center, Kitakyushu, Japan
| | - Kohsuke Harada
- Department of Internal Medicine, Kenwakai Otemachi Hospital, Kitakyushu, Japan
| | - Hiroaki Kurisu
- Department of Urology, Kenwakai Otemachi Hospital, Kitakyushu, Japan
| | - Shohei Shimajiri
- Department of Pathology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yasumasa Matsuki
- Department of Pathology, Kenwakai Otemachi Hospital, Kitakyushu, Japan
| | - Toshiyuki Nakayama
- Department of Pathology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|
126
|
Benzekry S, Sentis C, Coze C, Tessonnier L, André N. Development and Validation of a Prediction Model of Overall Survival in High-Risk Neuroblastoma Using Mechanistic Modeling of Metastasis. JCO Clin Cancer Inform 2021; 5:81-90. [PMID: 33439729 DOI: 10.1200/cci.20.00092] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Prognosis of high-risk neuroblastoma (HRNB) remains poor despite multimodal therapies. Better prediction of survival could help to refine patient stratification and better tailor treatments. We established a mechanistic model of metastasis in HRNB relying on two processes: growth and dissemination relying on two patient-specific parameters: the dissemination rate μ and the minimal visible lesion size Svis. This model was calibrated using diagnosis values of primary tumor size, lactate dehydrogenase circulating levels, and the meta-iodobenzylguanidine International Society for Paediatric Oncology European (SIOPEN) score from nuclear imaging, using data from 49 metastatic patients. It was able to describe the data of total tumor mass (lactate dehydrogenase, R2 > 0.99) and number of visible metastases (SIOPEN, R2 = 0.96). A prediction model of overall survival (OS) was then developed using Cox regression. Clinical variables alone were not able to generate a model with sufficient OS prognosis ability (P = .507). The parameter μ was found to be independent of the clinical variables and positively associated with OS (P = .0739 in multivariable analysis). Critically, addition of this computational biomarker significantly improved prediction of OS with a concordance index increasing from 0.675 (95% CI, 0.663 to 0.688) to 0.733 (95% CI, 0.722 to 0.744, P < .0001), resulting in significant OS prognosis ability (P = .0422).
Collapse
Affiliation(s)
- Sébastien Benzekry
- MONC Team, Inria Bordeaux Sud-Ouest and Institut de Mathématiques de Bordeaux, CNRS, University of Bordeaux, Bordeaux, France
| | - Coline Sentis
- Paediatric Hematology and Oncology Department, Hôpital pour enfant de La Timone, AP-HM, Marseille, France
| | - Carole Coze
- Paediatric Hematology and Oncology Department, Hôpital pour enfant de La Timone, AP-HM, Marseille, France.,Aix Marseille University, Marseille, France
| | - Laëtitia Tessonnier
- Department of Nuclear Medicine, Hôpital de La Timone, AP-HM, Marseille, France
| | - Nicolas André
- Paediatric Hematology and Oncology Department, Hôpital pour enfant de La Timone, AP-HM, Marseille, France.,SMARTc Unit, Centre de Recherche en Cancérologie de Marseille, Inserm U1068, Aix Marseille University, Marseille, France
| |
Collapse
|
127
|
Morandi F, Sabatini F, Podestà M, Airoldi I. Immunotherapeutic Strategies for Neuroblastoma: Present, Past and Future. Vaccines (Basel) 2021; 9:43. [PMID: 33450862 PMCID: PMC7828327 DOI: 10.3390/vaccines9010043] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/07/2021] [Accepted: 01/10/2021] [Indexed: 12/22/2022] Open
Abstract
Neuroblastoma is the most common extracranial pediatric solid tumor with a heterogeneous clinical course, ranging from spontaneous regression to metastatic disease and death, irrespective of intensive chemotherapeutic regimen. On the basis of several parameters, children affected by neuroblastoma are stratified into low, intermediate and high risk. At present, more than 50% of high-risk patients with metastatic spread display an overall poor long-term outcome also complicated by devastating long-term morbidities. Thus, novel and more effective therapies are desperately needed to improve lifespan of high-risk patients. In this regard, adoptive cell therapy holds great promise and several clinical trials are ongoing, demonstrating safety and tolerability, with no toxicities. Starting from the immunological and clinical features of neuroblastoma, we here discuss the immunotherapeutic approaches currently adopted for high-risk patients and different innovative therapeutic strategies currently under investigation. The latter are based on the infusion of natural killer (NK) cells, as support of consolidation therapy in addition to standard treatments, or chimeric antigen receptor (CAR) T cells directed against neuroblastoma associated antigens (e.g., disialoganglioside GD2). Finally, future perspectives of adoptive cell therapies represented by γδ T lymphocyes and CAR NK cells are envisaged.
Collapse
Affiliation(s)
| | | | | | - Irma Airoldi
- Laboratorio Cellule Staminali Post-Natali e Terapie Cellulari, Istituto Giannina Gaslini (Istituto di Ricerca e Cura a Carattere Scientifico—IRCCS), Via G. Gaslini 5, 16147 Genova, Italy; (F.M.); (F.S.); (M.P.)
| |
Collapse
|
128
|
Kumar A, Rocke JPJ, Kumar BN. Evolving treatments in high-risk neuroblastoma. Expert Opin Orphan Drugs 2020. [DOI: 10.1080/21678707.2020.1865918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Abhinav Kumar
- Division of Medicine, University College London Medical School, London, UK
| | - John P J Rocke
- ENT Department, Royal Albert Edward Infirmary, Wigan, UK
| | - B Nirmal Kumar
- ENT Department, Wrightington, Wigan & Leigh Teaching NHS, Wigan, UK
| |
Collapse
|
129
|
Pérez-Torres-Lobato MR, De Las Morenas-Iglesias J, Llempén-López M, Gómez-Millán-Ruiz P, Márquez-Vega C, Espiñeira-Periñán MÁ, Coronel-Rodríguez C, Franco-Ruedas C, Balboa-Huguet B, Sánchez-Vicente JL. Paediatric Horner syndrome. A case series of 14 patients in a tertiary hospital. ARCHIVOS DE LA SOCIEDAD ESPAÑOLA DE OFTALMOLOGÍA 2020; 96:356-365. [PMID: 34217473 DOI: 10.1016/j.oftale.2020.06.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 06/22/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND Horner syndrome (HS) is characterised by the triad of upper eyelid ptosis, miosis, and facial anhidrosis. Due to its wide variety of causes, it can occur at any age, and is uncommon in paediatrics. The aetiology and diagnostic approach of paediatric HS (PHS) is controversial. OBJECTIVE The purpose of this study is to describe the clinical characteristics of a 14 case series, focusing on the aetiology of HS and the clinical evolution the patients presented. METHODS A retrospective observational study was conducted on patients under 14 years-old (enrolled between 1st January 2009 and 30th April 2020). Depending on the age at diagnosis (before or after the first 5 months of life), the study cases were divided into two groups: congenital or acquired. RESULTS Fourteen patients, with a mean age of 8.5 months, were enrolled. The most frequent cause of PHS were tumours (6/14), with the most representative neoplasm being neuroblastoma (4/14). Of the acquired cases (8/14), the most frequent cause was iatrogenic (5/8), mainly secondary to cervical or thoracic surgery. The main origin of congenital HS (6/14) was neuroblastoma (4/6), being the first manifestation of the disease in 50% of patients (2/4). CONCLUSION HS may be the first sign of a major underlying disease, such as neuroblastoma. For this reason, children presenting with HS of unknown origin require imaging studies to exclude a life threatening disease. A thorough examination is essential for early diagnosis of these patients.
Collapse
Affiliation(s)
| | | | - M Llempén-López
- Hospital Universitario Virgen del Rocío, Pediatría, servicio de Oncología Pediátrica, Sevilla, Spain
| | - P Gómez-Millán-Ruiz
- Hospital Universitario Virgen del Rocío, Radiología, sección General, Sevilla, Spain
| | - C Márquez-Vega
- Hospital Universitario Virgen del Rocío, Pediatría, servicio de Oncología Pediátrica, Sevilla, Spain
| | - M Á Espiñeira-Periñán
- Hospital Universitario Virgen del Rocío, Oftalmología, sección General, Sevilla, Spain
| | | | - C Franco-Ruedas
- Hospital Universitario Virgen del Rocío, Oftalmología, sección General, Sevilla, Spain
| | - B Balboa-Huguet
- Hospital Universitario Virgen del Rocío, Oftalmología, sección Oftalmología Infantil, Sevilla, Spain
| | - J L Sánchez-Vicente
- Hospital Universitario Virgen del Rocío, Oftalmología, sección de Retina Quirúrgica, Sevilla, Spain
| |
Collapse
|
130
|
The glycosphingolipid GD2 as an effective but enigmatic target of passive immunotherapy in children with aggressive neuroblastoma (HR-NBL). Cancer Lett 2020; 503:220-230. [PMID: 33271265 DOI: 10.1016/j.canlet.2020.11.044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 11/02/2020] [Accepted: 11/26/2020] [Indexed: 11/20/2022]
Abstract
Neuroblastoma (NBL), the most frequent and lethal pediatric cancer of children in pre-school age, is considered enigmatic in view of its extreme heterogeneity, from spontaneous regression in the IV-S form to incurable disease in approx. 40% of cases (High Risk, HR-NBL). It has an embryonal origin and a very heterogeneous genomic landscape, hampering the success of targeted strategies. The glycosphingolipid GD2 was shown to be expressed on NBL cells and utilized as target for passive immunotherapy with anti-GD2 antibodies (GD2-IMT). An international protocol was established with GD2-IMT, which increases remission length and survival in HR-NBL. By reviewing the different biological and molecular aspects of NBL and GD2-IMT, this mini-review questions the present lack of association between GD2-IMT and the underlying molecular landscape. The alternative model of Micro-Foci inducing virus (MFV) is presented, since MFV infection can induce extensive genomic aberrations (100X NMYC DNA-amplification). Since this family of viruses uses molecules for cell penetration similar to GD2 (i.e., GM2), it is hypothesized that GD2 is the port-of-entry for MFV and that success of anti-GD2 therapies is also associated to inhibition of this clastogenic virus in HR-NBL.
Collapse
|
131
|
Yan P, Qi F, Bian L, Xu Y, Zhou J, Hu J, Ren L, Li M, Tang W. Comparison of Incidence and Outcomes of Neuroblastoma in Children, Adolescents, and Adults in the United States: A Surveillance, Epidemiology, and End Results (SEER) Program Population Study. Med Sci Monit 2020; 26:e927218. [PMID: 33249420 PMCID: PMC7711874 DOI: 10.12659/msm.927218] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background This United States (U.S.) population study aimed to compare the incidence of neuroblastoma and outcomes in children, adolescents, and adults using the Surveillance, Epidemiology, and End Results (SEER) program database. Material/Methods Patients with neuroblastoma were identified in the SEER database from 1975 to 2013. According to the age at diagnosis, patients were divided into “Children” (≤14 years old) and “Adolescents/Adults” group (>14 years old). Then, comparisons in basic characteristics, incidence rates (IRs) and long-term survival outcomes between patients in 2 groups were made. Results A total of 4280 patients were identified, including 3998 children and 282 adolescent/adult patients. Adolescent/adult patients were more likely to have localized diseases than children and to be diagnosed with ganglioneuroblastoma (all P<0.05). The IR of neuroblastoma presented with upward and downward trends in children and adolescent/adult populations, respectively. Adolescents/adults had worse overall survival (OS) than children despite the earlier tumor stage. Lastly, multivariate Cox proportional hazards analyses showed that tumor stage, histology, sequence of primary malignancy, primary site, the administration of surgery, and treatment era were prognostic factors for children, and sequence of primary malignancy, primary site, undergoing surgery, and treatment era were tightly related to OS in adolescent/adult patients. Conclusions Analysis of the SEER program database between 1975 to 2013 showed that in the U.S., the incidence of neuroblastoma in children increased, but the incidence decreased in adolescents and adults. There was a trend for improved overall survival in all age groups despite the increased stage at presentation in children.
Collapse
Affiliation(s)
- Ping Yan
- Department of Neonatal Surgery, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Feng Qi
- Department of Urologic Surgery, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Lanzheng Bian
- Department of Nursing, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Yajuan Xu
- Department of Neonatal Surgery, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Jing Zhou
- Department of Nursing, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Jiajie Hu
- Department of Nursing, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Lei Ren
- Department of Nursing, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Mei Li
- Department of Nursing, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Weibin Tang
- Department of Neonatal Surgery, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| |
Collapse
|
132
|
Nalci OB, Nadaroglu H, Genc S, Hacimuftuoglu A, Alayli A. The effects of MgS nanoparticles-Cisplatin-bio-conjugate on SH-SY5Y neuroblastoma cell line. Mol Biol Rep 2020; 47:9715-9723. [PMID: 33191478 DOI: 10.1007/s11033-020-05987-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 11/06/2020] [Indexed: 10/23/2022]
Abstract
Magnesium sulfide nanoparticles (MgS NPs) is a nanomaterial that has an important place in diagnosis, treatment, diagnosis, and drug delivery systems. Neuroblastoma, a type of brain cancer, is an extremely difficult cancer to treat with today's treatment options. This study was carried out to determine the cytotoxic, oxidant, and antioxidant effects on the neuroblastoma cancer line (SH-SY5Y cell line) along with the green synthesis and characterization of MgS NPs structures. MgS NPs were synthesized by green synthesis using Na2S and Punica granatum, a cleaner method for toxic effects, and characterized using Scanning Electron Microscopy, Fourier Transform Infrared spectroscopy, X-Ray diffraction methods. In cell culture, SH-SY5Y cells were grown in a suitable nutrient medium under favorable conditions. Five different doses of MgS NPs (10, 25, 50, 75, and 100 µg/mL) were applied to the cell line for 24 h. The analysis of the MgS NPs applications was performed with MTT cytotoxicity test and total oxidant and total antioxidant tests. According to the data obtained, 75 μg/mL MgS NPs application decreased cancer cell viability up to 48.54%. MgS NPs exhibited a dose-dependent effect on the SH-SY5Y cell line. Also, it was determined that MgS NPs increased oxidant activity in neuroblastoma cells, which was compatible with the cytotoxicity test. As a result, MgS NPs exhibited an effective activity on the neuroblastoma cell line. It was clearly seen that NPs obtained by green synthesis prevented the related cancer line from proliferating.
Collapse
Affiliation(s)
- Ozge Balpinar Nalci
- Department of Medical Pharmacology, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - Hayrunnisa Nadaroglu
- Department of Food Technology, Erzurum Vocational College, Ataturk University, 25240, Erzurum, Turkey. .,Department of Nano-Science and Nano-Engineering, Institute of Science and Technology, Ataturk University, 25240, Erzurum, Turkey.
| | - Sidika Genc
- Department of Medical Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Ahmet Hacimuftuoglu
- Department of Medical Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Turkey.
| | - Azize Alayli
- Department of Nursing, Faculty of Health Sciences, Sakarya University of Applied Sciences, 54187, Sakarya, Turkey
| |
Collapse
|
133
|
Song J, Zhao Q, Xu Y, Zhu L. A signature of 29 immune-related genes pairs to predict prognosis in patients with neuroblastoma. Int Immunopharmacol 2020; 88:106994. [PMID: 33182060 DOI: 10.1016/j.intimp.2020.106994] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND Neuroblastoma (NB) is one of the most common childhood tumors that is associated with a poor prognosis. Recently, immunotherapy has been recognized as an effective strategy in the treatment of NB patients. In this study, an immune-related gene pair (IRGP) signature was established for predicting the prognosis of NB patients. METHODS The Treehouse Childhood Cancer Initiative dataset and the Gene Expression Omnibus (GEO) were included in this study, as the training set and testing set, respectively. Immune-related genes retrieved from the ImmPort database were used to construct the prognostic model. Least absolute shrinkage and selection operator (LASSO) regression was used to develop the prognostic signature. Kaplan-Meier survival curves and the log-rank test were used to compare the differences between high and low immune risk scores groups. Immune infiltration analysis was estimated using TIMER, quanTIseq, and MCP-counter algorithms. The Tumor ImmunoPhenotype (TIP) pipeline was used for cancer-immunity cycle studies. RESULTS We identified an IRGP model that was significantly correlated with survival rates and the immune risk score was calculated for each sample. The low immune risk group had significantly better prognostic outcome compared with the high immune risk group. Besides, age, MYCN status, histology, Children's Oncology Group (COG) risk, mitosis-karyorrhexis index (MKI), and immune risk scores were found to be independent prognostic factors. Moreover, the low immune risk group showed a negative correlation with the immune cell infiltration, which may activate the anti-cancer immune response. CONCLUSIONS This prognostic immune signature based on IRGP reflects the link between the NB patient outcome and immune infiltration, and provides new insights into the prediction of prognosis in NB.
Collapse
Affiliation(s)
- Jingjing Song
- Department of Pediatric Surgery, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang Province, China; Department of Children's Health Care, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang Province, China
| | - Qianlei Zhao
- Department of Pediatric Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang Province, China
| | - Yuexia Xu
- Department of Pediatric Surgery, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang Province, China; Department of Children's Health Care, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang Province, China
| | - Libin Zhu
- Department of Pediatric Surgery, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang Province, China.
| |
Collapse
|
134
|
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
|
135
|
Shimada H, Sano H, Hazard FK. Pathology of Peripheral Neuroblastic Tumors. CLINICAL PEDIATRIC HEMATOLOGY-ONCOLOGY 2020. [DOI: 10.15264/cpho.2020.27.2.73] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Hiroyuki Shimada
- Department of Pathology and Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Hideki Sano
- Department of Pathology Oncology, Fukushima Medical University Hospital, Fukushima, Japan
| | - Florette K. Hazard
- Department of Pathology and Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
136
|
Jarzembowski JA. New Prognostic Indicators in Pediatric Adrenal Tumors: Neuroblastoma and Adrenal Cortical Tumors, Can We Predict When These Will Behave Badly? Surg Pathol Clin 2020; 13:625-641. [PMID: 33183724 DOI: 10.1016/j.path.2020.08.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Pediatric adrenal tumors are unique entities with specific diagnostic, prognostic, and therapeutic challenges. The adrenal medulla gives rise to peripheral neuroblastic tumors (pNTs), pathologically defined by their architecture, stromal content, degree of differentiation, and mitotic-karyorrhectic index. Successful risk stratification of pNTs uses patient age, stage, tumor histology, and molecular/genetic aberrations. The adrenal cortex gives rise to adrenocortical tumors (ACTs), which present diagnostic and prognostic challenges. Histologic features that signify poor prognosis in adults can be meaningless in children, who have superior outcomes. The key clinical, pathologic, and molecular findings of pediatric ACTs have yet to be completely identified.
Collapse
Affiliation(s)
- Jason A Jarzembowski
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA; Pathology and Laboratory Medicine, Children's Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
137
|
Bahmad HF, Chalhoub RM, Harati H, Bou-Gharios J, Assi S, Ballout F, Monzer A, Msheik H, Araji T, Elajami MK, Ghanem P, Chamaa F, Kadara H, Abou-Antoun T, Daoud G, Fares Y, Abou-Kheir W. Tideglusib attenuates growth of neuroblastoma cancer stem/progenitor cells in vitro and in vivo by specifically targeting GSK-3β. Pharmacol Rep 2020; 73:211-226. [PMID: 33030673 DOI: 10.1007/s43440-020-00162-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 09/01/2020] [Accepted: 09/19/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Neuroblastoma (NB) is the most frequently diagnosed extracranial solid tumor among the pediatric population. It is an embryonic tumor with high relapse rates pertaining to the presence of dormant slowly dividing cancer stem cells (CSC) within the tumor bulk that are responsible for therapy resistance. Therefore, there is a dire need to develop new therapeutic approaches that specifically target NB CSCs. Glycogen synthase kinase (GSK)-3β is a serine/threonine kinase that represents a common signaling node at the intersection of many pathways implicated in NB CSCs. GSK-3β sustains the survival and maintenance of CSCs and renders them insensitive to chemotherapeutic agents and radiation. METHODS In our study, we aimed at evaluating the potential anti-tumor effect of Tideglusib (TDG), an irreversible GSK-3β inhibitor drug, on three human NB cell lines, SK-N-SH, SH-SY5Y, and IMR-32. RESULTS Our results showed that TDG significantly reduced cell proliferation, viability, and migration of the NB cells, in a dose- and time-dependent manner, and also significantly hindered the neurospheres formation eradicating the self-renewal ability of highly resistant CSCs. Besides, TDG potently reduced CD133 cancer stem cell marker expression in both SH-SY5Y cells and G1 spheres. Lastly, TDG inhibited NB tumor growth and progression in vivo. CONCLUSION Collectively, we concluded that TDG could serve as an effective treatment capable of targeting the NB CSCs and hence overcoming therapy resistance. Yet, future studies are warranted to further investigate its potential role in NB and decipher the subcellular and molecular mechanisms underlying this role.
Collapse
Affiliation(s)
- Hisham F Bahmad
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,Neuroscience Research Center, Faculty of Medicine, Lebanese University, Beirut, Lebanon.,Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL, USA
| | - Reda M Chalhoub
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,Medical Scientist Training Program, College of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Hayat Harati
- Neuroscience Research Center, Faculty of Medicine, Lebanese University, Beirut, Lebanon
| | - Jolie Bou-Gharios
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,Neuroscience Research Center, Faculty of Medicine, Lebanese University, Beirut, Lebanon
| | - Sahar Assi
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Farah Ballout
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Alissar Monzer
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Hiba Msheik
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Tarek Araji
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Mohamad K Elajami
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,Department of Internal Medicine, Mount Sinai Medical Center, Miami Beach, FL, USA
| | - Paola Ghanem
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Farah Chamaa
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Humam Kadara
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tamara Abou-Antoun
- School of Pharmacy, Department of Pharmaceutical Sciences, Lebanese American University, Byblos, Lebanon
| | - Georges Daoud
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.
| | - Youssef Fares
- Neuroscience Research Center, Faculty of Medicine, Lebanese University, Beirut, Lebanon.
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
138
|
LimFat GJ, Kukreti V. Visual Diagnosis: Rapid Deterioration of Respiratory Status and Lower Limb Hypotonia in a 6-month-old Infant. Pediatr Rev 2020; 41:e37-e41. [PMID: 33004671 DOI: 10.1542/pir.2017-0199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Guillaume J LimFat
- Undergraduate Medicine, Queen's University School of Medicine, Kingston, ON, Canada
| | - Vinay Kukreti
- Department of Pediatrics, Queen's University School of Medicine, Kingston General Hospital, Kingston, ON, Canada.,Department of Pediatrics, Lakeridge Health Oshawa, Oshawa, ON, Canada
| |
Collapse
|
139
|
Wei M, Ye M, Dong K, Dong R. Circulating tumor DNA in neuroblastoma. Pediatr Blood Cancer 2020; 67:e28311. [PMID: 32729220 DOI: 10.1002/pbc.28311] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/10/2020] [Accepted: 03/19/2020] [Indexed: 12/20/2022]
Abstract
As a sympathetic nervous system-derived tumor, aggressive neuroblastoma (NB) is currently attracting interest from researchers seeking diagnostic and prognostic markers via less invasive procedures. The analysis of circulating tumor DNA (ctDNA) in peripheral blood can provide genetic information from multiple tumor lesions and is not dependent on a surgical procedure. The identification of genetic alterations, chromosomal variations, and hypermethylation contained within plasma DNA yields clinical value in the diagnosis, risk stratification, monitoring of treatment effects, and survival prediction for patients. With the widespread application of genome sequencing, droplet digital polymerase chain reaction, and other advanced technologies, the detection of ctDNA may guide therapeutic schedules, enhance the quality of life, and improve the prognosis for patients with NB.
Collapse
Affiliation(s)
- Meng Wei
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai, China
| | - Mujie Ye
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai, China
| | - Kuiran Dong
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai, China
| | - Rui Dong
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai, China
| |
Collapse
|
140
|
Olsen HE, Campbell K, Bagatell R, DuBois SG. Trends in conditional survival and predictors of late death in neuroblastoma. Pediatr Blood Cancer 2020; 67:e28329. [PMID: 32735385 DOI: 10.1002/pbc.28329] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/29/2020] [Accepted: 03/30/2020] [Indexed: 11/11/2022]
Abstract
PURPOSE Significant advances in the treatment of neuroblastoma have been made in the past several decades. There are scant data examining how these improvements have changed over time and differentially affected conditional survival among high-risk and non-high-risk patient groups. METHODS We conducted a retrospective cohort study using the Surveillance, Epidemiology, and End Results database. We analyzed clinical characteristics and survival outcomes for 4717 neuroblastoma patients. Kaplan-Meier methods were used to estimate overall survival (OS) and conditional overall survival (COS) with estimates compared between groups using log-rank tests. RESULTS Five-year OS was 41.46% (95% CI 38.77-44.13) for the high-risk group and 91.13% (95% CI 89.49-92.53) for the non-high-risk group. Both groups saw significant improvements in OS by decade (P < .001). Five-year COS among 1-year survivors was 52.69% (CI 49.54-55.73) for the high-risk group and 96.75% (95% CI 95.57-97.62) for the non-high-risk group. One-year survivors in the high-risk group showed a statistically significant improvement in COS over time. No difference in COS was observed among 5-year high-risk survivors. In the high-risk and non-high-risk groups, 82% and 32% of late deaths were attributable to cancer, respectively. Statistically significant adverse prognostic factors for late death were age ≥ 1 year at diagnosis, metastatic disease, and nonthoracic primary site (P = .001). CONCLUSIONS Improvements in COS over time have largely benefited high-risk patients, though they are still at higher risk for late death due to cancer when compared to non-high-risk patients. Age, stage, and primary site, but not treatment decade, influence outcomes among 5-year survivors.
Collapse
Affiliation(s)
| | - Kevin Campbell
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, Massachusetts
| | - Rochelle Bagatell
- Department of Pediatrics, Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Steven G DuBois
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
141
|
Factors Impacting Time to Engraftment in Patients With High-risk Neuroblastoma Following Autologous Stem Cell Transplant. J Pediatr Hematol Oncol 2020; 42:e569-e574. [PMID: 32032244 DOI: 10.1097/mph.0000000000001731] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Despite advances in supportive measures, myeloablative chemotherapy with stem cell rescue remains limited by toxicity and treatment-related mortality. The purpose of this study was to identify factors influencing the rate of hematopoietic recovery following autologous stem cell transplant in high-risk neuroblastoma. PROCEDURE We retrospectively studied 54 patients with high-risk neuroblastoma who received a single autologous stem cell transplant between 2006 and 2016. Race, sex, conditioning regimen, chemotherapy delays and bone marrow involvement were analyzed using Kaplan-Meier Log-Rank test while the amount of cells infused, age, and length of hospital stay were analyzed using univariate Cox Proportional Hazards Regression. RESULTS The conditioning regimen administered was significant (P=0.016) for time to engraftment of neutrophils, with busulfan/melphalan (Bu/Mel) at 16.6 days, and carboplatin/etoposide/melphalan at 12.1 days. A delay of chemotherapy during induction (n=24) was significant (P<0.001) for time to platelet engraftment of >75,000/µL. Female patients had a longer time to engraftment (P=0.029). CONCLUSION Patients receiving Bu/Mel as a conditioning regimen, patients who had a delay in induction chemotherapy and patients of female sex were found to be significant for delayed engraftment of neutrophils, platelets, and hemoglobin, respectively, in patients with high-risk neuroblastoma undergoing autologous stem cell transplant. Knowing these factors may lead to new expectations and possible interventions to decrease the morbidity and mortality of treatment and recovery.
Collapse
|
142
|
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: 48] [Impact Index Per Article: 9.6] [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
|
143
|
TRIM proteins in neuroblastoma. Biosci Rep 2020; 39:221458. [PMID: 31820796 PMCID: PMC6928532 DOI: 10.1042/bsr20192050] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 01/01/2023] Open
Abstract
Neuroblastoma (NB) is the most common extracranial solid tumor in childhood. Outcome for children with high-risk NB remains unsatisfactory. Accumulating evidence suggests that tripartite motif (TRIM) family proteins express diversely in various human cancers and act as regulators of oncoproteins or tumor suppressor proteins. This review summarizes the TRIM proteins involving in NB and the underlying molecular mechanisms. We expect these new insights will provide important implications for the treatment of NB by targeting TRIM proteins.
Collapse
|
144
|
Pan J, Lin H, Yang T, Yang J, Hu C, Zhu J, Tan T, Li J, Xia H, He J, Zou Y. lncRNA-uc003opf.1 rs11752942 A>G polymorphism decreases neuroblastoma risk in Chinese children. Cell Cycle 2020; 19:2367-2372. [PMID: 32809919 PMCID: PMC7513837 DOI: 10.1080/15384101.2020.1808382] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 07/08/2020] [Accepted: 08/06/2020] [Indexed: 02/07/2023] Open
Abstract
Recent studies have revealed that long non-coding RNAs (lncRNAs) play critical roles in the tumorigenesis and proliferation of human cancer. Several polymorphisms of lncRNAs have been found to be involved in the risk of neuroblastoma (NB). However, studies on the relationship between polymorphisms in lncRNA exons and NB are infrequent. We evaluated the association between rs11752942 A > G polymorphism in lnc-RNA-uc003opf.1 exon and neuroblastoma susceptibility by performing a hospital-based study with 275 patients and 531 controls. Odds ratios (ORs) and 95% confidence intervals (CIs) assessed by using logistic regression models were used to determine the strength of the association. We found that the rs11752942 G allele is significantly associated with decreased neuroblastoma risk (AG vs. AA: adjusted OR = 0.72, 95% CI = 0.53-0.98, P = 0.038; and AG/GG vs. AA: adjusted OR = 0.74, 95% CI = 0.55-0.99, P = 0.045) after adjusting for age and gender. This association was more prominent in females, subjects with tumor in the mediastinum or early-stage. Furthermore, the expression quantitative trait locus analysis indicated that rs11752942 G was associated with decreased expression of its neighboring gene LRFN2 mRNA. These results indicate that lncRNA-uc003opf.1 may be a novel potentially functional lncRNA that may be used as a predictive marker, for it might contribute to decreased neuroblastoma risk.
Collapse
Affiliation(s)
- Jing Pan
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Huiran Lin
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Tianyou Yang
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jiliang Yang
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Chao Hu
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jinhong Zhu
- Department of Clinical Laboratory, Biobank, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Tianbao Tan
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jiahao Li
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Huimin Xia
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yan Zou
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
145
|
Meng X, Feng C, Fang E, Feng J, Zhao X. Combined analysis of RNA-sequence and microarray data reveals effective metabolism-based prognostic signature for neuroblastoma. J Cell Mol Med 2020; 24:10367-10381. [PMID: 32683778 PMCID: PMC7521294 DOI: 10.1111/jcmm.15650] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 06/21/2020] [Accepted: 06/25/2020] [Indexed: 12/17/2022] Open
Abstract
The relationship between metabolism reprogramming and neuroblastoma (NB) is largely unknown. In this study, one RNA-sequence data set (n = 153) was used as discovery cohort and two microarray data sets (n = 498 and n = 223) were used as validation cohorts. Differentially expressed metabolic genes were identified by comparing stage 4s and stage 4 NBs. Twelve metabolic genes were selected by LASSO regression analysis and integrated into the prognostic signature. The metabolic gene signature successfully stratifies NB patients into two risk groups and performs well in predicting survival of NB patients. The prognostic value of the metabolic gene signature is also independent with other clinical risk factors. Nine metabolism-related long non-coding RNAs (lncRNAs) were also identified and integrated into the metabolism-related lncRNA signature. The lncRNA signature also performs well in predicting survival of NB patients. These results suggest that the metabolic signatures have the potential to be used for risk stratification of NB. Gene set enrichment analysis (GSEA) reveals that multiple metabolic processes (including oxidative phosphorylation and tricarboxylic acid cycle, both of which are emerging targets for cancer therapy) are enriched in the high-risk NB group, and no metabolic process is enriched in the low-risk NB group. This result indicates that metabolism reprogramming is associated with the progression of NB and targeting certain metabolic pathways might be a promising therapy for NB.
Collapse
Affiliation(s)
- Xinyao Meng
- Department of Pediatric SurgeryTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanP. R. China
| | - Chenzhao Feng
- School of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhanP. R. China
| | - Erhu Fang
- Department of Pediatric SurgeryTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanP. R. China
| | - Jiexiong Feng
- Department of Pediatric SurgeryTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanP. R. China
| | - Xiang Zhao
- Department of Pediatric SurgeryTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanP. R. China
| |
Collapse
|
146
|
Aravindan N, Herman T, Aravindan S. Emerging therapeutic targets for neuroblastoma. Expert Opin Ther Targets 2020; 24:899-914. [PMID: 33021426 PMCID: PMC7554151 DOI: 10.1080/14728222.2020.1790528] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 06/29/2020] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Neuroblastoma (NB) is the prime cancer of infancy, and accounts for 9% of pediatric cancer deaths. While children diagnosed with clinically stable NB experience a complete cure, those with high-risk disease (HR-NB) do not recover, despite intensive therapeutic strategies. Development of novel and effective targeted therapies is needed to counter disease progression, and to benefit long-term survival of children with HR-NB. AREAS COVERED Recent studies (2017-2020) pertinent to NB evolution are selectively reviewed to recognize novel and effective therapeutic targets. The prospective and promising therapeutic targets/strategies for HR-NB are categorized into (a) targeting oncogene-like and/or reinforcing tumor suppressor (TS)-like lncRNAs; (b) targeting oncogene-like microRNAs (miRs) and/or mimicking TS-miRs; (c) targets for immunotherapy; (d) targeting epithelial-to-mesenchymal transition and cancer stem cells; (e) novel and beneficial combination approaches; and (f) repurposing drugs and other strategies in development. EXPERT OPINION It is highly unlikely that agents targeting a single candidate or signaling will be beneficial for an HR-NB cure. We must develop efficient drug deliverables for functional targets, which could be integrated and advance clinical therapy. Fittingly, the looming evidence indicated an aggressive evolution of promising novel and integrative targets, development of efficient drugs, and improvised strategies for HR-NB treatment.
Collapse
Affiliation(s)
| | - Terence Herman
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
- Stephenson Cancer Center, Oklahoma City, USA
| | | |
Collapse
|
147
|
Meng X, Li H, Fang E, Feng J, Zhao X. Comparison of Stage 4 and Stage 4s Neuroblastoma Identifies Autophagy-Related Gene and LncRNA Signatures Associated With Prognosis. Front Oncol 2020; 10:1411. [PMID: 32974147 PMCID: PMC7466450 DOI: 10.3389/fonc.2020.01411] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/03/2020] [Indexed: 01/01/2023] Open
Abstract
Background: The spontaneous regression of neuroblastoma (NB) is most prevalent and well-documented in stage 4s NB patients. However, whether autophagy plays roles in the spontaneous regression of NB is unknown. Objective: This study aimed to identify autophagy-related genes (ARGs) and autophagy-related long non-coding RNAs (lncRNAs) differentially expressed in stage 4 and stage 4s NB and to build prognostic risk signatures on the basis of the ARGs and autophagy-related lncRNAs. Methods: One RNA-sequence (RNA-Seq) dataset (TARGET NBL, n = 153) was utilized as discovery cohort, and two microarray datasets (n = 498 and n = 223) were used as validation cohorts. Differentially expressed ARGs were identified by comparing stage 4s and stage 4 NB samples. An ARG signature risk score and an autophagy-related lncRNA signature risk score were constructed. The receiver operating characteristic (ROC) curve analyses were used to evaluate the survival prediction ability of the two signatures. Gene function annotation and Gene Set Enrichment Analysis (GSEA) were performed to clarify the autophagic biological processes enriched in different risk groups. Results: Nine ARGs were integrated into the ARG signature. Patients in the high-risk group of the ARG signature had significantly poorer overall survival (OS) than patients in the low-risk group. The ROC curves analyses revealed that the ARG signature performed very well in predicting OS [5-year area under the curve (AUC) = 0.81]. Seven autophagy-related lncRNAs were integrated into the autophagy-related lncRNA signature. Patients in the high-risk group of the lncRNA signature had significantly poorer OS than patients in the low-risk group. The ROC curve analyses also revealed that the lncRNA signature performed well in predicting OS (5-year AUC = 0.77). Both the ARG signature and lncRNA signature are independent with other clinical risk factors in the multivariate Cox regression survival analyses. GSEAs revealed that autophagy-related biological processes are enriched in low-risk groups. Conclusions: Autophagy-related genes and lncRNAs are differentially expressed between stage 4 and stage 4s NB. The ARG signature and autophagy-related lncRNA signature successfully stratified NB patients into two risk groups. Autophagy-related biological processes are highly enriched in low-risk NB groups.
Collapse
Affiliation(s)
| | | | | | | | - Xiang Zhao
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
148
|
Hishiki T, Fujino A, Watanabe T, Tahara K, Ohno M, Yamada Y, Tomonaga K, Kutsukake M, Fujita T, Kawakubo N, Matsumoto K, Kiyotani C, Shioda Y, Miyazaki O, Fuji H, Yoshioka T, Kanamori Y. Definitive Tumor Resection after Myeloablative High Dose Chemotherapy Is a Feasible and Effective Option in the Multimodal Treatment of High-Risk Neuroblastoma: A Single Institution Experience. J Pediatr Surg 2020; 55:1655-1659. [PMID: 31575417 DOI: 10.1016/j.jpedsurg.2019.08.050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 08/23/2019] [Accepted: 08/29/2019] [Indexed: 01/06/2023]
Abstract
BACKGROUND/PURPOSE The delayed local treatment approach (DL) in high-risk neuroblastoma (HR-NB) refers to the process in which tumor resection is performed after the completion of all the courses of chemotherapy, including myeloablative high-dose chemotherapy (HDC). Alternatively, in the conventional local treatment approach (CL), tumor resection is performed during induction chemotherapy. In this study, we compared the surgical outcomes in HR-NB patients treated by CL and DL. METHOD Forty-seven patients with abdominal HR-NB underwent primary tumor resection from 2002 to 2018. The timing of surgery was generally determined by following the trials and guidelines available at the time. The outcomes and surgical complications between the two strategies were compared. RESULT Operation time, blood loss, and postoperative WBC counts were lower in the DL group (n = 25) when compared to the CL group (n = 22), statistical significance notwithstanding. Major vascular structures were less frequently encased in the DL group tumors, while immediate surgical complications were significantly more frequent in the CL group (P < 0.05). Furthermore, the 3-year EFSs were 50.0% and 53.9% in the DL and CL groups, respectively. CONCLUSION DL appears to be a feasible and effective treatment option for HR-NB. Nonetheless, further verifications using larger cohorts are warranted. LEVEL OF EVIDENCE Treatment study, Level III.
Collapse
Affiliation(s)
- Tomoro Hishiki
- Division of Surgical Oncology, Children's Cancer Center, National Center for Child Health and Development; Division of Surgery, Department of Surgical Specialties, National Center for Child Health and Development; Department of Pediatric Surgical Oncology, National Cancer Center Hospital.
| | - Akihiro Fujino
- Division of Surgery, Department of Surgical Specialties, National Center for Child Health and Development
| | - Toshihiko Watanabe
- Division of Surgery, Department of Surgical Specialties, National Center for Child Health and Development
| | - Kazunori Tahara
- Division of Surgery, Department of Surgical Specialties, National Center for Child Health and Development
| | - Michinobu Ohno
- Division of Surgery, Department of Surgical Specialties, National Center for Child Health and Development
| | - Yohei Yamada
- Division of Surgery, Department of Surgical Specialties, National Center for Child Health and Development
| | - Kotaro Tomonaga
- Division of Surgery, Department of Surgical Specialties, National Center for Child Health and Development
| | - Mai Kutsukake
- Division of Surgery, Department of Surgical Specialties, National Center for Child Health and Development
| | - Takuro Fujita
- Division of Surgery, Department of Surgical Specialties, National Center for Child Health and Development
| | - Naonori Kawakubo
- Division of Surgical Oncology, Children's Cancer Center, National Center for Child Health and Development; Department of Pediatric Surgical Oncology, National Cancer Center Hospital
| | - Kimikazu Matsumoto
- Children's Cancer Center, National Center for Child Health and Development
| | - Chikako Kiyotani
- Children's Cancer Center, National Center for Child Health and Development
| | - Yoko Shioda
- Children's Cancer Center, National Center for Child Health and Development
| | - Osamu Miyazaki
- Department of Radiology, National Center for Child Health and Development
| | - Hiroshi Fuji
- Department of Radiology, National Center for Child Health and Development
| | - Takako Yoshioka
- Department of Pathology, National Center for Child Health and Development
| | - Yutaka Kanamori
- Division of Surgery, Department of Surgical Specialties, National Center for Child Health and Development
| |
Collapse
|
149
|
Moroz V, Machin D, Hero B, Ladenstein R, Berthold F, Kao P, Obeng Y, Pearson ADJ, Cohn SL, London WB. The prognostic strength of serum LDH and serum ferritin in children with neuroblastoma: A report from the International Neuroblastoma Risk Group (INRG) project. Pediatr Blood Cancer 2020; 67:e28359. [PMID: 32472746 DOI: 10.1002/pbc.28359] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 03/12/2020] [Accepted: 04/02/2020] [Indexed: 12/29/2022]
Abstract
PURPOSE Age, MYCN status, stage, and histology have been used as neuroblastoma (NB) risk factors for decades. Serum lactate dehydrogenase (LDH) and serum ferritin are reproducible, easily obtained, and prognostic, though never used in risk stratification, except one German trial. We analyzed the prognostic strength of LDH and ferritin, overall, within high-risk NB, and by era, using the International Neuroblastoma Risk Group Data Commons. PATIENTS AND METHODS Children with NB (1990-2016) were categorized into LDH (n = 8867) and ferritin (n = 8575) risk groups using EFS. Cox models compared the prognostic strength of LDH and ferritin to age, MYCN status, and INSS stage. RESULTS Higher LDH conferred worse EFS, overall (5-year EFS) (100-899 IU/L: 76 ± 0.6%; 0-99 or 900-1399 IU/L: 60 ± 1.2%; ≥1400 IU/L: 36 ± 1.2%; P < .0001), and in high-risk NB post-2009 (3-year EFS) (117-381 IU/L: 67 ± 8.9%; 382-1334 IU/L: 58 ± 4.4%; 0-116 or ≥1335 IU/L: 46 ± 3.9%; P = .003). Higher ferritin conferred worse EFS, overall (5-year EFS) (1-29 ng/mL: 87 ± 0.9%; 0 or 30-89 ng/mL: 74 ± 0.8%; ≥90 ng/mL: 48 ± 0.9%; P < .0001), and in high-risk NB post-2009 (3-year EFS) (1-53 ng/mL: 71 ± 9.3%; 0 or 54-354 ng/mL: 55 ± 4.7%; ≥355 ng/mL: 34 ± 6.1%; P = .0008). In multivariable analyses adjusting for age, MYCN, and stage, LDH and ferritin maintained independent prognostic ability (P < .0001; adjusted HRs (95% CI): 1.7 (1.5-1.9), 2.3 (2.0-2.7), respectively). CONCLUSIONS LDH and ferritin are strongly prognostic in NB, overall and within high-risk NB patients treated post-2009 with modern therapy. LDH and ferritin show promise for (a) identifying ultra-high-risk; (b) refining risk stratification; and (c) clinical utility in low-/middle-income countries. Routine collection of LDH and ferritin should be reinitiated for evolving NB risk stratification.
Collapse
Affiliation(s)
- Veronica Moroz
- Cancer Research UK Trials Unit, University of Birmingham, Birmingham, UK
| | - David Machin
- Leicester Cancer Research Centre, University of Leicester, Leicester, UK
| | - Barbara Hero
- Department of Pediatric Oncology and Hematology, Children's Hospital, University of Cologne, Cologne, Germany
| | | | - Frank Berthold
- Department of Pediatric Oncology and Hematology, Children's Hospital, University of Cologne, Cologne, Germany
| | - Paige Kao
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, Massachusetts
| | - Yaa Obeng
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, Massachusetts
| | - Andrew D J Pearson
- Section of Paediatrics, Institute of Cancer Research and Royal Marsden Hospital, Surrey, UK
| | - Susan L Cohn
- Department of Pediatrics, The University of Chicago, Chicago, Illinois
| | - Wendy B London
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
150
|
Pan J, Zhu J, Wang M, Yang T, Hu C, Yang J, Zhang J, Cheng J, Zhou H, Xia H, He J, Zou Y. Association of MYC gene polymorphisms with neuroblastoma risk in Chinese children: A four-center case-control study. J Gene Med 2020; 22:e3190. [PMID: 32222109 DOI: 10.1002/jgm.3190] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 02/04/2020] [Accepted: 03/15/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Neuroblastoma is one of the most common malignant tumors in childhood. Polymorphisms in proto-oncogene MYC are implicated in many cancers, although their role in neuroblastoma remains unclear. In the present study, we attempted to investigate the association between MYC gene polymorphisms and neuroblastoma susceptibility in Chinese children. METHODS We included two MYC polymorphisms (rs4645943 and rs2070583) and assessed their effects on neuroblastoma risk in 505 cases and 1070 controls via the Taqman method. RESULTS In single and combined locus analysis, no significant association was found between the two selected polymorphisms and neuroblastoma susceptibility. In stratification analysis, the rs4645943 CT/TT genotypes were significantly associated with a decreased neuroblastoma risk in subjects with tumors originating from other sites [adjusted odds ratio (OR) = 0.42, 95% confidence interval (CI) = 0.21-0.84, p = 0.013]. Meanwhile, the presence of one or two protective genotypes was significantly associated with a decreased neuroblastoma risk in subjects with tumors arising from other sites (adjusted OR = 0.50, 95% CI = 0.26-0.96, p = 0.036). CONCLUSIONS The present study indicates that MYC gene polymorphisms may have a weak effect on the neuroblastoma risk, which neeeds to be verified further.
Collapse
Affiliation(s)
- Jing Pan
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jinhong Zhu
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Clinical Laboratory, Biobank, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Mi Wang
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Tianyou Yang
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Chao Hu
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jiliang Yang
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jiao Zhang
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jiwen Cheng
- Department of Pediatric Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Haixia Zhou
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Huimin Xia
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yan Zou
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|