1
|
Xie W, Zhang Y, Xu J, Sun F, Zhu J, Que Y, Huang J, Zhen Z, Lu S, Wang J, Zhang Y. Characteristics, treatments, and outcomes of adolescents and adults with neuroblastoma: a retrospective study in China. Ther Adv Med Oncol 2025; 17:17588359251337494. [PMID: 40351327 PMCID: PMC12064894 DOI: 10.1177/17588359251337494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 04/08/2025] [Indexed: 05/14/2025] Open
Abstract
Background Neuroblastoma (NB) is rare in adolescents and adults, resulting in limited availability of data. Objectives We comprehensively investigated the characteristics, treatments, and outcomes of adolescent and adult patients with NB, aiming to provide a more in-depth insight into this disease. Design A retrospective, single-center study. Methods We retrieved and analyzed the medical data of patients with NB aged 10 years or older at diagnosis who were treated at Sun Yat-sen University Cancer Center between June 2005 and January 2024. Results Sixty-five patients (30 males and 35 females) were enrolled, with a median age of 20 years (interquartile range, 14-26 years), including 27 patients aged 10-18 years and 38 patients aged >18 years. Most patients were classified as M-stage disease (n = 40, 61.5%), high-risk (n = 42, 64.6%), and poorly differentiated NB (n = 27, 41.5%). Additionally, 3 (6.7%) patients had MYCN amplification, and 5 (25%) had ALK mutations. The genomic landscape revealed that mutations in the cell cycle and DNA repair pathways are related to chemotherapy sensitivity. After induction therapy, 34 (52.3%) patients achieved complete response (CR). The 5-year progression-free survival (PFS) and overall survival (OS) rates were 33.1% ± 6.9% and 55.1% ± 7.6%, respectively. Patients who achieved CR after induction therapy had superior PFS (p = 0.009), with 5-year PFS rates of 44.0% ± 10.6% compared to 18.5% ± 8.5% in non-CR patients. Conclusion Adolescent and adult patients with NB exhibit distinct characteristics, less chemotherapy sensitivity, and poorer outcomes compared to pediatric patients. Achieving CR after induction therapy is associated with better outcomes. Further investigation for new therapies is required.
Collapse
Affiliation(s)
- Weiji Xie
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Yu Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Jiaqian Xu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Feifei Sun
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Jia Zhu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Yi Que
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Junting Huang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Zijun Zhen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Suying Lu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Yuexiu District, Guangzhou City, Guangdong 510060, P.R. China
| | - Juan Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Yuexiu District, Guangzhou City, Guangdong 510060, P.R. China
| | - Yizhuo Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Yuexiu District, Guangzhou City, Guangdong 510060, P.R. China
| |
Collapse
|
2
|
Wang H, Ye W, Li T, Liao Q, Zhang Y, Liu L, He L, Chen X. Integrating neuron-specific enolase and venous-phase enhanced computed tomography radiomics for improved survival prediction in high-risk neuroblastoma: a two-center study. Pediatr Radiol 2025; 55:1178-1190. [PMID: 40327094 DOI: 10.1007/s00247-025-06247-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND High-risk neuroblastoma is associated with a poor prognosis, making it crucial to identify patients within this group who face an even higher risk of adverse outcomes. OBJECTIVE To determine if integrating clinical indicators and venous-phase enhanced computed tomography radiomics features could improve the prediction of overall survival in high-risk neuroblastoma. MATERIALS AND METHODS We retrospectively included high-risk neuroblastoma patients treated at a primary institution, randomly stratifying them into a training set (70%) and a test set (30%). Univariate and multivariate Cox regression analyses were used to identify independent clinical risk factors. We then extracted radiomics features from venous-phase enhanced computed tomography images. Clinical risk factors, radiomics score, and combined model were evaluated in the training, test, and external validation sets. RESULTS The training, test, and validation sets included 70, 30, and 40 patients, respectively. Neuron-specific enolase was identified as the independent clinical risk factor, with concordance indices of 0.616, 0.627, and 0.595 in the training, test, and validation sets, respectively. The radiomics score achieved concordance indices of 0.699, 0.690, and 0.684 in the training, test, and validation sets, respectively. The combined model showed concordance indices of 0.730, 0.707, and 0.690 in the training, test, and validation sets, respectively. The combined model predicted 5-year overall survival with an area under the receiver operating characteristic curve of 0.780 in the training set, 0.742 in the test set, and 0.710 in the validation set. CONCLUSION Combining neuron-specific enolase and venous-phase enhanced computed tomography radiomics improves survival prediction in high-risk neuroblastoma.
Collapse
Affiliation(s)
- Haoru Wang
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing, 400014, China
| | - Wenhong Ye
- Department of Radiology, Shenzhen Children's Hospital, Shenzhen, 518038, China
| | - Ting Li
- Department of Radiology, Medical Science Research Center, University-Town Hospital of Chongqing Medical University, Chongqing, 401331, China
| | - Qianyi Liao
- Department of Radiology, Shenzhen Children's Hospital, Shenzhen, 518038, China
| | - Ya Zhang
- Department of Radiology, Shenzhen Children's Hospital, Shenzhen, 518038, China
| | - Longping Liu
- Department of Radiology, Shenzhen Children's Hospital, Shenzhen, 518038, China
| | - Ling He
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing, 400014, China
| | - Xin Chen
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing, 400014, China.
| |
Collapse
|
3
|
Li S, Wang J, Zhang Z, Ren C, He D. Individual risk and prognostic value prediction by interpretable machine learning for distant metastasis in neuroblastoma: A population-based study and an external validation. Int J Med Inform 2025; 196:105813. [PMID: 39904180 DOI: 10.1016/j.ijmedinf.2025.105813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 12/27/2024] [Accepted: 01/23/2025] [Indexed: 02/06/2025]
Abstract
PURPOSE Neuroblastoma (NB) is a childhood malignancy with a poor prognosis and a propensity for distant metastasis (DM). We aimed to establish machine learning (ML) based model to accurately predict risk of DM and prognosis of NB patients with DM. METHODS We analyzed NB patients from the Surveillance, Epidemiology, and End Results (SEER) database between 2000 and 2020. Univariate and multivariate logistic analysis were employed to select meaning variables. Recursive Feature Elimination (RFE) method based on 6 ML algorithms was utilized in feature selection. To construct predictive model, 13 ML algorithms were evaluated by area under the operating characteristic curve (AUC), accuracy, sensitivity, specificity, precision, cross-entropy, Brier scores, Balanced Accuracy and F-beta score. An optimal ML model was constructed to predict DM, and the predictive results were explained by SHapley Additive exPlanations (SHAP) framework. Meanwhile, 101 ML algorithm combinations were developed to select the best model with highest C-index to predict prognosis of NB patients with DM. RESULTS A total of 1,668 NB patients from SEER database was consecutively enrolled. We identified that tumor primary site, grade, surgery type, regional lymph nodes, radiotherapy and chemotherapy are significant risk factors for DM. CatBoost model was selected as the best prediction model, and AUC was 0.846 (95 %CI: [0.804,0.899]), 0.834 (95 %CI: [0.796,0.873]) and 0.813 (95 %CI: [0.776,0.852]) in training, internal test and external test sets, with 0.777 accuracy, 0.839 sensitivity, 0.72 specificity and 0.731 precision in training set. Grade, chemotherapy and radiotherapy had the greatest effects on DM according to SHAP results. For prognosis prediction, "RSF + GBM" algorithm was the best prognostic model with C-index of 0.656, 0.611 and 0.629 in training, internal test and external test sets. CONCLUSIONS Our ML models demonstrate excellent accuracy and reliability, offering more precise personalized metastasis diagnosis and prognostic prediction to NB patients.
Collapse
Affiliation(s)
- Shan Li
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, China; China International Science and Technology Cooperation base of Child Development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Jinkui Wang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, China; China International Science and Technology Cooperation base of Child Development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Zhaoxia Zhang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, China; China International Science and Technology Cooperation base of Child Development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Chunnian Ren
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, China; China International Science and Technology Cooperation base of Child Development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Dawei He
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, China; China International Science and Technology Cooperation base of Child Development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing 400014, China.
| |
Collapse
|
4
|
Mora J, Chan GCF, Morgenstern DA, Amoroso L, Nysom K, Faber J, Wingerter A, Bear MK, Rubio-San-Simon A, de Las Heras BM, Tornøe K, Düring M, Kushner BH. The anti-GD2 monoclonal antibody naxitamab plus GM-CSF for relapsed or refractory high-risk neuroblastoma: a phase 2 clinical trial. Nat Commun 2025; 16:1636. [PMID: 39952926 PMCID: PMC11828896 DOI: 10.1038/s41467-025-56619-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 01/20/2025] [Indexed: 02/17/2025] Open
Abstract
In this single-arm, non-randomized, phase 2 trial (NCT03363373), 74 patients with relapsed/refractory high-risk neuroblastoma and residual disease in bone/bone marrow (BM) received naxitamab on Days 1, 3, and 5 (3 mg/kg/day) with granulocyte-macrophage colony-stimulating factor (Days -4 to 5) every 4 weeks, until complete response (CR) or partial response (PR) followed by 5 additional cycles every 4 weeks. Primary endpoint in the prespecified interim analysis was overall response (2017 International Neuroblastoma Response Criteria). Among 26 responders (CR + PR) in the efficacy population (N = 52), 58% had refractory disease, and 42% had relapsed disease. Overall response rate (ORR) was 50% (95% CI: 36-64%), and CR and PR were observed in 38% and 12%, respectively. With the 95% CI lower limit for ORR exceeding 20%, the primary endpoint of overall response was met. Patients with evaluable bone disease had a 58% (29/50) bone compartment response (CR, 40%; PR, 18%). BM compartment response was 74% (17/23; CR, 74%). One-year overall survival and progression-free survival (secondary endpoints) were 93% (95% CI: 80-98%) and 35% (95% CI: 16-54%), respectively. Naxitamab-related Grade 3 adverse events included hypotension (58%) and pain (54%). Overall, naxitamab demonstrated clinically meaningful efficacy with manageable safety in patients with residual neuroblastoma in bone/BM.
Collapse
Affiliation(s)
- Jaume Mora
- Pediatric Cancer Center Barcelona, Hospital Sant Joan de Déu, Barcelona, Spain.
| | - Godfrey C F Chan
- Queen Mary Hospital & Hong Kong Children's Hospital, Pok Fu Lam, Hong Kong
- The University of Hong Kong, Pok Fu Lam, Hong Kong
| | | | - Loredana Amoroso
- IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Department of Maternal Infantile and Urological Sciences, Pediatric Onco-Hematology Unit, Policlinico Umberto I, Sapienza, University of Rome, Rome, Italy
| | - Karsten Nysom
- Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Jörg Faber
- Department of Pediatric Hematology/Oncology/Hemostaseology, Center for Pediatric and Adolescent Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Arthur Wingerter
- Department of Pediatric Hematology/Oncology/Hemostaseology, Center for Pediatric and Adolescent Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | | | | | | | | | | | | |
Collapse
|
5
|
Grunewald L, Andersch L, Helmsauer K, Schwiebert S, Klaus A, Henssen AG, Straka T, Lodrini M, Wicha SG, Fuchs S, Hertwig F, Westermann F, Vitali A, Caramel C, Büchel G, Eilers M, Astrahantseff K, Eggert A, Höpken UE, Schulte JH, Blankenstein T, Anders K, Künkele A. Targeting MYCN upregulates L1CAM tumor antigen in MYCN-dysregulated neuroblastoma to increase CAR T cell efficacy. Pharmacol Res 2025; 212:107608. [PMID: 39828101 DOI: 10.1016/j.phrs.2025.107608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 12/18/2024] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
Current treatment protocols have limited success against MYCN-amplified neuroblastoma. Adoptive T cell therapy presents an innovative strategy to improve cure rates. However, L1CAM-targeting CAR T cells achieved only limited response against refractory/relapsed neuroblastoma so far. We investigated how oncogenic MYCN levels influence tumor cell response to CAR T cells, as one possible factor limiting clinical success. A MYCN-inducible neuroblastoma cell model was created. L1CAM-CAR T cell effector function was assessed (activation markers, cytokine release, tumor cytotoxicity) after coculture with the model or MYCN-amplified neuroblastoma cell lines. RNA sequencing datasets characterizing the model were compared to publicly available RNA/proteomic datasets. MYCN-directed L1CAM regulation was explored using public ChIP-sequencing datasets. Synergism between CAR T cells and the indirect MYCN inhibitor, MLN8237, was assessed in vitro using the Bliss model and in vivo in an immunocompromised mouse model. Inducing high MYCN levels in the neuroblastoma cell model reduced L1CAM expression and, consequently, L1CAM-CAR T cell effector function in vitro. Primary neuroblastomas possessing high MYCN levels expressed lower levels of both the L1CAM transcript and L1CAM tumor antigen. MLN8237 treatment restored L1CAM tumor expression and L1CAM-CAR T cell effector function. Combining MLN8237 and L1CAM-CAR T cell treatment synergistically enhanced MYCN-overexpressing tumor cytotoxicity in vitro and in vivo concomitant with severe in vivo toxicity. We identify target antigen downregulation as source of resistance against L1CAM-CAR T cells in MYCN-driven neuroblastoma cells. These data suggest that L1CAM-CAR T cell therapy combined with pharmacological MYCN inhibition may benefit patients with MYCN-amplified neuroblastoma.
Collapse
Affiliation(s)
- Laura Grunewald
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany; German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Lena Andersch
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany; German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany; Freie Universität Berlin, Kaiserswerther Str. 16-18, Berlin 14195, Germany
| | - Konstantin Helmsauer
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany; Neuroblastoma Research Group, Experimental and Clinical Research Center (ECRC) of the Charité and the Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Lindenberger Weg 80, Berlin 13125, Germany
| | - Silke Schwiebert
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany
| | - Anika Klaus
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany
| | - Anton G Henssen
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany; Neuroblastoma Research Group, Experimental and Clinical Research Center (ECRC) of the Charité and the Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Lindenberger Weg 80, Berlin 13125, Germany
| | - Teresa Straka
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany
| | - Marco Lodrini
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany
| | - Sebastian G Wicha
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Bundesstrasse 45, Hamburg 20146, Germany
| | - Steffen Fuchs
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany; German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany; German Cancer Consortium (DKTK), Partner Site Berlin, Virchowweg 23, Berlin 10117, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Anna-Louisa-Karsch-Strasse 2, Berlin 10178, Germany
| | - Falk Hertwig
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany
| | - Frank Westermann
- German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Alice Vitali
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany
| | - Carlotta Caramel
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany
| | - Gabriele Büchel
- Department of Biochemistry and Molecular Biology, Theodor Boveri Institute, Biocenter, University of Würzburg, Am Hubland, Würzburg 97074, Germany; Mildred Scheel Early Career Center, University Hospital Würzburg, Josef-Schneider-Str. 6, Würzburg 97080, Germany
| | - Martin Eilers
- Department of Biochemistry and Molecular Biology, Theodor Boveri Institute, Biocenter, University of Würzburg, Am Hubland, Würzburg 97074, Germany
| | - Kathy Astrahantseff
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany
| | - Angelika Eggert
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany; German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany; German Cancer Consortium (DKTK), Partner Site Berlin, Virchowweg 23, Berlin 10117, Germany
| | - Uta E Höpken
- Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Robert-Rössle Str. 10, Berlin 13125, Germany
| | - Johannes H Schulte
- Universitätsklinik für Kinder, und Jugendmedizin, Department of Pediatric Hematology and Oncology, Hoppe-Seyler-Straße 1, Tübingen 72076, Germany
| | - Thomas Blankenstein
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany; Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Robert-Rössle Str. 10, Berlin 13125, Germany
| | - Kathleen Anders
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany
| | - Annette Künkele
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Oncology and Hematology, Augustenburger Platz 1, Berlin 13353, Germany; German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany; German Cancer Consortium (DKTK), Partner Site Berlin, Virchowweg 23, Berlin 10117, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Anna-Louisa-Karsch-Strasse 2, Berlin 10178, Germany.
| |
Collapse
|
6
|
Casey RT, Hendriks E, Deal C, Waguespack SG, Wiegering V, Redlich A, Akker S, Prasad R, Fassnacht M, Clifton-Bligh R, Amar L, Bornstein S, Canu L, Charmandari E, Chrisoulidou A, Freixes MC, de Krijger R, de Sanctis L, Fojo A, Ghia AJ, Huebner A, Kosmoliaptsis V, Kuhlen M, Raffaelli M, Lussey-Lepoutre C, Marks SD, Nilubol N, Parasiliti-Caprino M, Timmers HHJLM, Zietlow AL, Robledo M, Gimenez-Roqueplo AP, Grossman AB, Taïeb D, Maher ER, Lenders JWM, Eisenhofer G, Jimenez C, Pacak K, Pamporaki C. International consensus statement on the diagnosis and management of phaeochromocytoma and paraganglioma in children and adolescents. Nat Rev Endocrinol 2024; 20:729-748. [PMID: 39147856 DOI: 10.1038/s41574-024-01024-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/17/2024] [Indexed: 08/17/2024]
Abstract
Phaeochromocytomas and paragangliomas (PPGL) are rare neuroendocrine tumours that arise not only in adulthood but also in childhood and adolescence. Up to 70-80% of childhood PPGL are hereditary, accounting for a higher incidence of metastatic and/or multifocal PPGL in paediatric patients than in adult patients. Key differences in the tumour biology and management, together with rare disease incidence and therapeutic challenges in paediatric compared with adult patients, mandate close expert cross-disciplinary teamwork. Teams should ideally include adult and paediatric endocrinologists, oncologists, cardiologists, surgeons, geneticists, pathologists, radiologists, clinical psychologists and nuclear medicine physicians. Provision of an international Consensus Statement should improve care and outcomes for children and adolescents with these tumours.
Collapse
Affiliation(s)
- Ruth T Casey
- Department of Medical Genetics, University of Cambridge and NIHR Cambridge Biomedical Research Centre, Cambridge, UK.
- Department of Endocrinology, Cambridge Cancer Centre and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| | - Emile Hendriks
- Department of Paediatric Diabetes and Endocrinology, Cambridge Cancer Centre and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Cheri Deal
- Endocrine and Diabetes Service, CHU Sainte-Justine and University of Montreal, Montreal, Québec, Canada
| | - Steven G Waguespack
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Verena Wiegering
- University Children's Hospital, Department of Paediatric Hematology, Oncology and Stem Cell Transplantation, University of Würzburg, Würzburg, Germany
| | - Antje Redlich
- Paediatric Oncology Department, Otto von Guericke University Children's Hospital, Magdeburg, Germany
| | - Scott Akker
- St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Rathi Prasad
- Centre for Endocrinology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Martin Fassnacht
- Department of Medicine, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, Würzburg, Germany
| | - Roderick Clifton-Bligh
- Department of Diabetes and Endocrinology, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Laurence Amar
- Université de Paris, Paris, France
- Hypertension Unit, Hôpital Européen Georges Pompidou, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Stefan Bornstein
- Department of Medicine III, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Letizia Canu
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, Azienda Ospedaliera Universitaria (AOU) Careggi, Florence, Italy
| | - Evangelia Charmandari
- Division of Endocrinology, Metabolism and Diabetes, First Department of Paediatrics, National and Kapodistrian University of Athens Medical School, 'Aghia Sophia' Children's Hospital, Athens, Greece
| | | | - Maria Currás Freixes
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Institute of Health Carlos III (ISCIII), Madrid, Spain
| | - Ronald de Krijger
- Princess Maxima Center for Paediatric Oncology, Utrecht, Netherlands
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Luisa de Sanctis
- Department of Public Health and Paediatric Sciences, University of Turin, Turin, Italy
| | - Antonio Fojo
- Division of Hematology/Oncology, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Amol J Ghia
- Department of Radiation Oncology, University Hospital of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Angela Huebner
- Department of Paediatrics, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Vasilis Kosmoliaptsis
- Department of Surgery, University of Cambridge and National Institute for Health Research Cambridge Biomedical Research Centre, Addenbrooke's Hospital, Cambridge, UK
- Blood and Transplant Research Unit in Organ Donation and Transplantation, National Institute for Health Research, University of Cambridge, Cambridge, UK
| | - Michaela Kuhlen
- Paediatrics and Adolescent Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Marco Raffaelli
- U.O.C. Chirurgia Endocrina e Metabolica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Istituto di Semeiotica Chirurgica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Charlotte Lussey-Lepoutre
- Service de médecine nucléaire, Inserm U970, Sorbonne université, Groupe hospitalier Pitié-Salpétrière, Paris, France
| | - Stephen D Marks
- Department of Paediatric Nephrology, Great Ormond Street Hospital for Children NHS Foundation Trust and NIHR GOSH Biomedical Research Centre, University College London Great Ormond Street Institute of Child Health, London, UK
| | - Naris Nilubol
- Surgical Oncology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mirko Parasiliti-Caprino
- Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Corso Dogliotti, Turin, Italy
| | - Henri H J L M Timmers
- Department of Internal Medicine, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Anna Lena Zietlow
- Clinical Child and Adolescent Psychology, Institute of Clinical Psychology and Psychotherapy, Department of Psychology, TU Dresden, Dresden, Germany
| | - Mercedes Robledo
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Institute of Health Carlos III (ISCIII), Madrid, Spain
| | - Anne-Paule Gimenez-Roqueplo
- Université Paris Cité, PARCC, INSERM, Paris, France
- Service de Génétique, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France
| | - Ashley B Grossman
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- Centre for Endocrinology, Barts and the London School of Medicine, London, UK
- ENETS Centre of Excellence, Royal Free Hospital, London, UK
| | - David Taïeb
- Department of Nuclear Medicine, La Timone University Hospital, CERIMED, Aix-Marseille University, Marseille, France
| | - Eamonn R Maher
- Department of Medical Genetics, University of Cambridge and NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Jacques W M Lenders
- Department of Internal Medicine, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Graeme Eisenhofer
- Department of Medicine III, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Camilo Jimenez
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Rockville, MD, USA
| | - Christina Pamporaki
- Department of Medicine III, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
7
|
Liu T, Sheng Q, Xu W, Lu L, Zhu L, Xiong J, Jiang S, Yang X, Liu J, Lv Z. Tracking changes in image-defined risk factors during neoadjuvant chemotherapy and their predictive value for surgical outcomes based on the International Neuroblastoma Surgical Report Form. Pediatr Blood Cancer 2024; 71:e31161. [PMID: 38987989 DOI: 10.1002/pbc.31161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/10/2024] [Accepted: 06/09/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND The capacity of presurgical image-defined risk factors (IDRFs) to predict secondary surgical outcomes in patients with neuroblastoma is controversial. METHODS The International Neuroblastoma Surgical Report Form (INSRF) was employed to retrospectively collect the clinical data of 53 patients diagnosed with neuroblastoma at our hospital from April 2014 to April 2020. IDRFs were identified at the time of diagnosis and reassessed during the course of neoadjuvant chemotherapy. Various statistical tests were used to evaluate the correlation between IDRFs and secondary surgical outcomes. RESULTS A total of 195 IDRFs were identified. Notably, by two courses of neoadjuvant chemotherapy, the number of "two body compartments," "intraspinal tumor extension," and "trachea-compressing" IDRFs decreased significantly (p = .001). The primary tumor volumes and the number of IDRFs decreased significantly by four courses of neoadjuvant chemotherapy, especially in "intraspinal tumor extension" IDRFs (p = .034). The median number of IDRF per patient was four (interquartile range [IQR]: 1-5) at diagnosis, which diminished to one (IQR: 1-3) subsequent to neoadjuvant chemotherapy. The presence of preoperative IDRFs was not associated with surgical complications (p = .286) or the extent of surgery (p = .188). However, the number of preoperative IDRFs linked to the extent of surgery (p = .002), not to operative complications (p = .669). Specifically, presurgery "renal vessel contact" IDRFs were predictive of surgical complications, while presurgery "infiltration of vital structures" IDRFs were associated with the extent of surgery. CONCLUSION The number of IDRFs decreased significantly by four courses of neoadjuvant chemotherapy. The number and type of presurgery IDRFs may predict secondary surgical outcomes, surpassing the mere consideration of their presence or absence.
Collapse
Affiliation(s)
- Tao Liu
- Department of General Surgery, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qingfeng Sheng
- Department of General Surgery, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Weijue Xu
- Department of General Surgery, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li Lu
- Department of General Surgery, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Linlin Zhu
- Department of General Surgery, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Xiong
- Department of General Surgery, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shayi Jiang
- Department of Hematology/Oncology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiujun Yang
- Department of Radiology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiangbin Liu
- Department of General Surgery, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhibao Lv
- Department of General Surgery, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
8
|
Flaadt T, Jaki C, Maier CP, Amorelli G, Klingebiel T, Schlegel PG, Eyrich M, Greil J, Schulte JH, Bader P, Handgretinger R, Lang P. Immune reconstitution after transplantation of autologous peripheral stem cells in children: a comparison between CD34+ selected and nonmanipulated grafts. Cytotherapy 2024; 26:1227-1235. [PMID: 38904583 DOI: 10.1016/j.jcyt.2024.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 05/02/2024] [Accepted: 05/10/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND AND AIMS High-dose chemotherapy (HDC) followed by autologous stem cell transplantation (ASCT) improves the prognosis in pediatric patients with several solid tumors and lymphomas. Little is known about the reconstitution of the immune system after ASCT and the influence of CD34+ cell selection on the reconstitution in pediatric patients. METHODS Between 1990 and 2001, 94 pediatric patients with solid tumors and lymphomas received autologous CD34+ selected or unmanipulated peripheral stem cells after HDC. CD34+ selection was carried out with magnetic microbeads. The absolute numbers of T cells, B cells and natural killer (NK) cells were measured and compared in both groups at various time points post-transplant. RESULTS Recovery of T cells was significantly faster in the unmanipulated group at day 30, with no significant difference later on. Reconstitution of B and NK cells was similar in both groups without significant differences at any time. The CD34+-selected group was divided into patients receiving less or more than 5.385 × 106/kg CD34+ cells. Patients in the CD34+ high-dose group displayed significantly faster reconstitutions of neutrophiles and lymphocyte subsets than the CD34+ low-dose group. CONCLUSIONS Engraftment and reconstitution of leukocytes, B cells and NK cells after transplantation of CD34+ selected stem cells were comparable to that in patients receiving unmanipulated grafts. T-cell recovery was faster in the unmanipulated group only within the first month. However, this delay could be compensated by transplantation of >5.385 × 106 CD34+ cells/kg. Especially for patients receiving immunotherapy after HDC large numbers of immune effector cells such as NK and T cells are necessary to mediate antibody-dependent cellular cytotoxicity. Therefore, in patients receiving autologous CD34+-selected grafts, our data emphasize the need to administer high stem cell counts.
Collapse
Affiliation(s)
- Tim Flaadt
- Department of Hematology and Oncology, University Children's Hospital, Eberhard Karls University Tuebingen, Tuebingen, Germany.
| | - Christina Jaki
- Department of Hematology and Oncology, University Children's Hospital, Eberhard Karls University Tuebingen, Tuebingen, Germany; Simulation Center STUPS, Klinikum Stuttgart, Stuttgart, Germany
| | - Claus-Philipp Maier
- Department of Hematology and Oncology, University Children's Hospital, Eberhard Karls University Tuebingen, Tuebingen, Germany; Department of Hematology, Oncology, Clinical Immunology and Rheumatology, Center for Internal Medicine, University Hospital Tuebingen, Tuebingen, Germany
| | - Germano Amorelli
- Department of Hematology and Oncology, University Children's Hospital, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Thomas Klingebiel
- Goethe University, University Hospital, Department of Pediatrics, Division for Stem Cell Transplantation and Immunology, Frankfurt, Germany
| | - Paul Gerhardt Schlegel
- Department of Pediatric Hematology and Oncology, University Children's Hospital, University Medical Center, Wuerzburg, Germany
| | - Matthias Eyrich
- Department of Pediatric Hematology and Oncology, University Children's Hospital, University Medical Center, Wuerzburg, Germany
| | - Johann Greil
- Department of Pediatric Oncology, Hematology and Immunology, University Hospital Heidelberg, Heidelberg, Germany
| | - Johannes H Schulte
- Department of Hematology and Oncology, University Children's Hospital, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Peter Bader
- Goethe University, University Hospital, Department of Pediatrics, Division for Stem Cell Transplantation and Immunology, Frankfurt, Germany
| | - Rupert Handgretinger
- Department of Hematology and Oncology, University Children's Hospital, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Peter Lang
- Department of Hematology and Oncology, University Children's Hospital, Eberhard Karls University Tuebingen, Tuebingen, Germany
| |
Collapse
|
9
|
Gorostegui M, Muñoz JP, Perez-Jaume S, Simao-Rafael M, Larrosa C, Garraus M, Salvador N, Lavarino C, Krauel L, Mañe S, Castañeda A, Mora J. Management of High-Risk Neuroblastoma with Soft-Tissue-Only Disease in the Era of Anti-GD2 Immunotherapy. Cancers (Basel) 2024; 16:1735. [PMID: 38730688 PMCID: PMC11083939 DOI: 10.3390/cancers16091735] [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: 04/09/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Neuroblastoma presents with two patterns of disease: locoregional or systemic. The poor prognostic risk factors of locoregional neuroblastoma (LR-NB) include age, MYCN or MDM2-CDK4 amplification, 11q, histology, diploidy with ALK or TERT mutations, and ATRX aberrations. Anti-GD2 immunotherapy has significantly improved the outcome of high-risk (HR) NB and is mostly effective against osteomedullary minimal residual disease (MRD), but less so against soft tissue disease. The question is whether adding anti-GD2 monoclonal antibodies (mAbs) benefits patients with HR-NB compounded by only soft tissue. We reviewed 31 patients treated at SJD for HR-NB with no osteomedullary involvement at diagnosis. All tumors had molecular genetic features of HR-NB. The outcome after first-line treatment showed 25 (80.6%) patients achieving CR. Thirteen patients remain in continued CR, median follow-up 3.9 years. We analyzed whether adding anti-GD2 immunotherapy to first-line treatment had any prognostic significance. The EFS analysis using Cox models showed a HR of 0.20, p = 0.0054, and an 80% decrease in the risk of relapse in patients treated with anti-GD2 immunotherapy in the first line. Neither EFS nor OS were significantly different by CR status after first-line treatment. In conclusion, adding treatment with anti-GD2 mAbs at the stage of MRD helps prevent relapse that unequivocally portends poor survival.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Jaume Mora
- Pediatric Cancer Center Barcelona, Hospital Sant Joan de Déu, 08950 Barcelona, Spain; (M.G.); (J.P.M.); (M.S.-R.); (C.L.); (M.G.); (N.S.); (C.L.); (L.K.); (S.M.); (A.C.)
| |
Collapse
|
10
|
Peggion S, Najem S, Kolman JP, Reinshagen K, Pagerols Raluy L. Revisiting Neuroblastoma: Nrf2, NF-κB and Phox2B as a Promising Network in Neuroblastoma. Curr Issues Mol Biol 2024; 46:3193-3208. [PMID: 38666930 PMCID: PMC11048850 DOI: 10.3390/cimb46040200] [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: 02/28/2024] [Revised: 03/29/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
Neuroblastoma is the most common solid extracranial tumor during childhood; it displays extraordinary heterogeneous clinical courses, from spontaneous regression to poor outcome in high-risk patients due to aggressive growth, metastasizing, and treatment resistance. Therefore, the identification and detailed analysis of promising tumorigenic molecular mechanisms are inevitable. This review highlights the abnormal regulation of NF-κB, Nrf2, and Phox2B as well as their interactions among each other in neuroblastoma. NF-κB and Nrf2 play a key role in antioxidant responses, anti-inflammatory regulation and tumor chemoresistance. Recent studies revealed a regulation of NF-κB by means of the Nrf2/antioxidant response element (ARE) system. On the other hand, Phox2B contributes to the differentiation of immature sympathetic nervous system stem cells: this transcription factor regulates the expression of RET, thereby facilitating cell survival and proliferation. As observed in other tumors, we presume striking interactions between NF-κB, Nrf2, and Phox2B, which might constitute an important crosstalk triangle, whose decompensation may trigger a more aggressive phenotype. Consequently, these transcription factors could be a promising target for novel therapeutic approaches and hence, further investigation on their regulation in neuroblastoma shall be reinforced.
Collapse
Affiliation(s)
| | | | | | | | - Laia Pagerols Raluy
- Department of Pediatric Surgery, University Medical Centre Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| |
Collapse
|
11
|
Fiz F, Cirone A, Righi S, Massollo M, Amoroso L, Bottoni G, Conte M, Gambaro M, Massone F, Orengo S, Bruzzone GS, Sorrentino S, Garaventa A, Piccardo A. Reaching the target dose with one single 131 I-mIBG administration in high-risk neuroblastoma: The determinant impact of the primary tumour. Pediatr Blood Cancer 2024; 71:e30806. [PMID: 38082548 DOI: 10.1002/pbc.30806] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 11/13/2023] [Accepted: 11/19/2023] [Indexed: 01/24/2024]
Abstract
BACKGROUND 131 I-metaiodobenzylguanidine (131 I-mIBG) effectiveness in children with metastasised neuroblastoma (NB) is linked to the effective dose absorbed by the target; a target of 4 Gy whole-body dose threshold has been proposed. Achieving this dose often requires administering 131 I-mIBG twice back-to-back, which may cause haematological toxicity. In this study, we tried identifying the factors predicting the achievement of 4 Gy whole-body dose with a single radiopharmaceutical administration. MATERIALS AND METHODS Children affected by metastatic NB and treated with a high 131 I-mIBG activity (>450 MBq (megabecquerel)/kg) were evaluated retrospectively. Kinetics measurements were carried out at multiple time points to estimate the whole-body dose, which was compared with clinical and activity-related parameters. RESULTS Seventeen children (12 females, median age 3 years, age range: 1.5-6.9 years) were included. Eleven of them still bore the primary tumour. The median whole-body dose was 2.88 Gy (range: 1.63-4.22 Gy). Children with a 'bulky' primary (>30 mL) received a higher whole-body dose than those with smaller or surgically removed primaries (3.42 ± 0.74 vs. 2.48 ± 0.65 Gy, respectively, p = .016). Conversely, the correlation between activity/kg and the whole-body dose was moderate (R: 0.42, p = .093). In the multivariate analysis, the volume of the primary tumour was the most relevant predictor of the whole-body dose (p = .002). CONCLUSIONS These data suggest that the presence of a bulky primary tumour can significantly prolong the 131 I-mIBG biological half-life, effectively increasing the absorbed whole-body dose. This information could be used to model the administered activity, allowing to attain the target dose without needing a two-step radiopharmaceutical administration.
Collapse
Affiliation(s)
- Francesco Fiz
- Department of Nuclear Medicine, Galliera Hospital, Genoa, Italy
- Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital, Tübingen, Germany
| | - Alessio Cirone
- Department of Medical Physics, Galliera Hospital, Genoa, Italy
| | - Sergio Righi
- Department of Medical Physics, Galliera Hospital, Genoa, Italy
| | | | | | | | - Massimo Conte
- Oncology Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Monica Gambaro
- Department of Medical Physics, Galliera Hospital, Genoa, Italy
| | | | - Stefano Orengo
- Department of Medical Physics, Galliera Hospital, Genoa, Italy
| | | | | | | | | |
Collapse
|
12
|
Karami Fath M, Bagherzadeh Torbati SM, Saqagandomabadi V, Yousefi Afshar O, Khalilzad M, Abedi S, Moliani A, Daneshdoust D, Barati G. The therapeutic effect of MSCs and their extracellular vesicles on neuroblastoma. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2024; 187:51-60. [PMID: 38373516 DOI: 10.1016/j.pbiomolbio.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 10/04/2023] [Accepted: 02/16/2024] [Indexed: 02/21/2024]
Abstract
Neuroblastoma is a common inflammatory-related cancer during infancy. Standard treatment modalities including surgical interventions, high-dose chemotherapy, radiotherapy, and immunotherapy are not able to increase survival rate and reduce tumor relapse in high-risk patients. Mesenchymal stem cells (MSCs) are known for their tumor-targeting and immunomodulating properties. MSCs could be engineered to express anticancer agents (i.e., growth factors, cytokines, pro-apoptotic agents) or deliver oncolytic viruses in the tumor microenvironment. As many functions of MSCs are mediated through their secretome, researchers have tried to use extracellular vesicles (EVs) from MSCs for targeted therapy of neuroblastoma. Here, we reviewed the studies to figure out whether the use of MSCs could be worthwhile in neuroblastoma therapy or not. Native MSCs have shown a promoting or inhibiting role in cancers including neuroblastoma. Therefore, MSCs are proposed as a vehicle to deliver anticancer agents such as oncolytic viruses to the neuroblastoma tumor microenvironment. Although modified MSCs or their EVs have been shown to suppress the tumorigenesis of neuroblastoma, further pre-clinical and clinical studies are required to come to a conclusion.
Collapse
Affiliation(s)
- Mohsen Karami Fath
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | | | - Vahid Saqagandomabadi
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | | | - Mohammad Khalilzad
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sara Abedi
- Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Afshin Moliani
- Isfahan Medical Students Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Danyal Daneshdoust
- Faculty of Medicine, Babol University of Medical Sciences, Mazandaran, Iran
| | | |
Collapse
|
13
|
Muehling J, Fröba-Pohl A, Muensterer OJ, von Schweinitz D, Kappler R. Impact of BCL-2 Expression on Course of Disease in Neuroblastoma. Eur J Pediatr Surg 2024; 34:69-77. [PMID: 37774735 DOI: 10.1055/s-0043-1774798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/01/2023]
Abstract
OBJECTIVE The antiapoptotic BCL-2 protein has implications for maturation and differentiation of neural tissue and acts as a strong modulator of carcinogenesis in different tumors. Recent research focuses not only on its benefit as a prognostic factor, but also as a potential therapeutic target. The role of BCL-2 in neuroblastoma, the most common extracranial solid tumor in childhood, remains controversial. The aim of our study was to determine the gene expression level of BCL-2 in a large cohort of neuroblastoma patients and its correlation with clinical parameters. METHODS Tumor samples and clinical data were collected from 100 neuroblastoma patients treated according to the NB2004 protocol of the German Society of Pediatric Oncology and Hematology. BCL-2 gene expression levels were measured by quantitative reverse transcription polymerase chain reaction and correlated with clinical parameters. RESULTS BCL-2 expression was detected in all tumor samples. Relative BCL-2 expression levels were higher in females versus males (1.839 vs. 1.342; p = 0.0143), in patients with low versus high International Neuroblastoma Staging System stage (2.051 vs. 1.463; p = 0.0206), in nonmetastatic versus metastatic disease (1.801 vs. 1.342; p = 0.0242), as well as in patients without presurgical chemotherapy (2.145 vs. 1.402; p = 0.0016), but was not associated with overall survival and MYCN amplification. CONCLUSION Our study demonstrates the ubiquitous expression of BCL-2 in neuroblastoma and suggests the possibility for targeted therapy with BCL-2 inhibitors, even in lower-stage neuroblastoma. It also underlines the need for further research on concomitant genetic alterations for a better understanding of the impact of BCL-2 on this pediatric tumor type.
Collapse
Affiliation(s)
- Jakob Muehling
- Department of Pediatric Surgery, Munich University Hospital, Dr. von Hauner Children's Hospital, München, Germany
| | - Alexandra Fröba-Pohl
- Department of Pediatric Surgery, Munich University Hospital, Dr. von Hauner Children's Hospital, München, Germany
| | - Oliver J Muensterer
- Department of Pediatric Surgery, Munich University Hospital, Dr. von Hauner Children's Hospital, München, Germany
| | - Dietrich von Schweinitz
- Department of Pediatric Surgery, Munich University Hospital, Dr. von Hauner Children's Hospital, München, Germany
| | - Roland Kappler
- Department of Pediatric Surgery, Munich University Hospital, Dr. von Hauner Children's Hospital, München, Germany
| |
Collapse
|
14
|
Bechmann N, Moskopp ML, Constantinescu G, Stell A, Ernst A, Berthold F, Westermann F, Jiang J, Lui L, Nowak E, Zopp S, Pacak K, Peitzsch M, Schedl A, Reincke M, Beuschlein F, Bornstein SR, Fassnacht M, Eisenhofer G. Asymmetric Adrenals: Sexual Dimorphism of Adrenal Tumors. J Clin Endocrinol Metab 2024; 109:471-482. [PMID: 37647861 PMCID: PMC11032253 DOI: 10.1210/clinem/dgad515] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/03/2023] [Accepted: 08/28/2023] [Indexed: 09/01/2023]
Abstract
CONTEXT Sexual dimorphism has direct consequences on the incidence and survival of cancer. Early and accurate diagnosis is crucial to improve prognosis. OBJECTIVE This work aimed to characterized the influence of sex and adrenal asymmetry on the emergence of adrenal tumors. METHODS We conducted a multicenter, observational study involving 8037 patients with adrenal tumors, including adrenocortical carcinoma (ACC), aldosterone-producing adenoma (APA), cortisol-secreting adrenocortical adenomas (CSAs), non-aldosterone-producing adrenal cortical adenoma (NAPACA), pheochromocytoma (PCC), and neuroblastoma (NB), and investigated tumor lateralization according to sex. Human adrenal tissues (n = 20) were analyzed with a multiomics approach that allows determination of gene expression, catecholamine, and steroid contents in a single sample. In addition, we performed a literature review of computed tomography and magnetic resonance imaging-based studies examining adrenal gland size. RESULTS ACC (n = 1858); CSA (n = 68), NAPACA (n = 2174), and PCC (n = 1824) were more common in females than in males (female-to-male ratio: 1.1:1-3.8:1), whereas NBs (n = 2320) and APAs (n = 228) were less prevalent in females (0.8:1). ACC, APA, CSA, NAPACA, and NB occurred more frequently in the left than in the right adrenal (left-to-right ratio: 1.1:1-1.8:1), whereas PCC arose more often in the right than in the left adrenal (0.8:1). In both sexes, the left adrenal was larger than the right adrenal; females have smaller adrenals than males. CONCLUSION Adrenal asymmetry in both sexes may be related to the pathogenesis of adrenal tumors and should be considered during the diagnosis of these tumors.
Collapse
Affiliation(s)
- Nicole Bechmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Mats Leif Moskopp
- Department of Neurosurgery, Vivantes Friedrichshain Hospital, Charité Academic Teaching Hospital, 10249 Berlin, Germany
| | - Georgiana Constantinescu
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Anthony Stell
- School of Computing and Information Systems, University of Melbourne, 3052 Melbourne, Australia
| | - Angela Ernst
- Institute of Medical Statistics and Computational Biology, Faculty of Medicine, University of Cologne, 50931 Cologne, Germany
| | - Frank Berthold
- Children's Hospital, University of Cologne, 50735 Cologne, Germany
| | - Frank Westermann
- Hopp Children's Cancer Center Heidelberg (KiTZ), 69120 Heidelberg, Germany
- Division of Neuroblastoma Genomics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Jingjing Jiang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, 200031 Shanghai, China
| | - Longfei Lui
- Department of Urology, Xiangya Hospital, Central South University, 410017 Changsha, China
| | - Elisabeth Nowak
- Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität Munich, 80539 Munich, Germany
| | - Stephanie Zopp
- Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität Munich, 80539 Munich, Germany
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Rockville, MD 20892, USA
| | - Mirko Peitzsch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Andreas Schedl
- Université Côte d’Azur, Inserm, CNRS, Institut de Biologie Valrose, 06108 Nice, France
| | - Martin Reincke
- Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität Munich, 80539 Munich, Germany
| | - Felix Beuschlein
- Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität Munich, 80539 Munich, Germany
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), 8091 Zurich, Switzerland
- Institute of Neuropathology, University of Zurich, 8091 Zurich, Switzerland
| | - Stefan R Bornstein
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Martin Fassnacht
- Division of Endocrinology and Diabetes, Department of Internal Medicine I, University Hospital of Würzburg, University of Würzburg, 97080 Würzburg, Germany
| | - Graeme Eisenhofer
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| |
Collapse
|
15
|
Campbell K, Siegel DA, Umaretiya PJ, Dai S, Heczey A, Lupo PJ, Schraw JM, Thompson TD, Scheurer ME, Foster JH. A comprehensive analysis of neuroblastoma incidence, survival, and racial and ethnic disparities from 2001 to 2019. Pediatr Blood Cancer 2024; 71:e30732. [PMID: 37867409 PMCID: PMC11018254 DOI: 10.1002/pbc.30732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/05/2023] [Accepted: 10/07/2023] [Indexed: 10/24/2023]
Abstract
BACKGROUND We characterize the incidence and 5-year survival of children and adolescents with neuroblastoma stratified by demographic and clinical factors based on the comprehensive data from United States Cancer Statistics (USCS) and the National Program of Cancer Registries (NPCR). METHODS We analyzed the incidence of neuroblastoma from USCS (2003-2019) and survival data from NPCR (2001-2018) for patients less than 20 years old. Incidence trends were calculated by average annual percent change (AAPC) using joinpoint regression. Differences in relative survival were estimated comparing non-overlapping confidence intervals (CI). RESULTS We identified 11,543 primary neuroblastoma cases in USCS. Age-adjusted incidence was 8.3 per million persons [95% CI: 8.2, 8.5], with an AAPC of 0.4% [95% CI: -0.1, 0.9]. Five-year relative survival from the NPCR dataset (n = 10,676) was 79.7% [95% CI: 78.9, 80.5]. Patients aged less than 1 year had the highest 5-year relative survival (92.5%). Five-year relative survival was higher for non-Hispanic White patients (80.7%) or Hispanic patients (80.8%) compared to non-Hispanic Black patients (72.6%). CONCLUSION Neuroblastoma incidence was stable during 2003-2019. Differences in relative survival exist by sex, age, race/ethnicity, and stage; patients who were male, older, non-Hispanic Black, or with distant disease had worse survival. Future studies could seek to assess the upstream factors driving disparities in survival, and evaluate interventions to address inequities and improve survival across all groups.
Collapse
Affiliation(s)
- Kevin Campbell
- Division of Hematology-Oncology and Bone Marrow Transplantation, Children’s Mercy Hospitals and Clinics, Kansas City, Missouri, USA
| | - David A. Siegel
- Division of Cancer Prevention and Control, National Center for Chronic Disease Prevention and Health Promotion, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Puja J. Umaretiya
- Division of Hematology-Oncology and Bone Marrow Transplantation, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Shifan Dai
- Cyberdata Technologies, Inc., Herndon, Virginia, USA
| | - Andras Heczey
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Cancer and Hematology Centers, Texas Children’s Hospital, Houston, Texas, USA
| | - Philip J. Lupo
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Cancer and Hematology Centers, Texas Children’s Hospital, Houston, Texas, USA
- Center for Epidemiology and Population Health, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Jeremy M. Schraw
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Cancer and Hematology Centers, Texas Children’s Hospital, Houston, Texas, USA
- Center for Epidemiology and Population Health, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Trevor D. Thompson
- Division of Cancer Prevention and Control, National Center for Chronic Disease Prevention and Health Promotion, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Michael E. Scheurer
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Cancer and Hematology Centers, Texas Children’s Hospital, Houston, Texas, USA
- Center for Epidemiology and Population Health, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Jennifer H. Foster
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Cancer and Hematology Centers, Texas Children’s Hospital, Houston, Texas, USA
| |
Collapse
|
16
|
Youlden DR, Baade PD, Frazier AL, Gupta S, Gottardo NG, Moore AS, Aitken JF. Temporal changes in childhood cancer incidence and survival by stage at diagnosis in Australia, 2000-2017. Acta Oncol 2023; 62:1256-1264. [PMID: 37647245 DOI: 10.1080/0284186x.2023.2251668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 08/20/2023] [Indexed: 09/01/2023]
Abstract
BACKGROUND The Toronto Paediatric Cancer Stage Guidelines are a compendium of staging systems developed to facilitate collection of consistent and comparable data on stage at diagnosis for childhood cancers by cancer registries. MATERIAL AND METHODS This retrospective, observational cohort study investigated changes in stage-specific incidence and survival for children diagnosed between 2000-2008 compared to 2009-2017 using the population-based Australian Childhood Cancer Registry. Information on mortality for each patient was available to 31st December 2020. Shifts in incidence by stage were evaluated using chi-square tests, and differences in stage-specific five-year observed survival for all causes of death over time were assessed using flexible parametric models. RESULTS Stage was assigned according to the Toronto Guidelines for 96% (n = 7944) of the total study cohort (n = 8292). Changes in the distribution of incidence by stage between the two diagnosis periods were observed for retinoblastoma, with stage 0 increasing from 26% to 37% of cases (p = 0.02), and hepatoblastoma, with metastatic disease increasing from 22% to 39% of cases (p = 0.04). There were large gains in stage-specific survival over time for stage IV rhabdomyosarcoma (five-year adjusted mortality hazard ratio for 2009-2017 compared to 2000-2008 of 0.38, 95% CI 0.19-0.77; p = 0.01), stage M3 for medulloblastoma (HR = 0.41, 95% CI 0.21-0.79; p = 0.01) and metastatic neuroblastoma excluding stage MS (HR = 0.61, 95% CI 0.44-0.84; p < 0.01). CONCLUSION These results indicate that improvements in childhood cancer survival in Australia are most likely due to refined management rather than changes in stage at diagnosis, particularly for metastatic solid tumours. Wide international uptake of the Toronto Guidelines will allow comprehensive evaluation of differences in survival between countries.
Collapse
Affiliation(s)
- Danny R Youlden
- Viertel Cancer Research Centre, Cancer Council Queensland, Brisbane, Australia
- Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia
| | - Peter D Baade
- Viertel Cancer Research Centre, Cancer Council Queensland, Brisbane, Australia
- Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia
- School of Mathematical Sciences, Queensland University of Technology, Brisbane, Australia
| | - A Lindsay Frazier
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, USA
| | - Sumit Gupta
- Division of Haematology/Oncology, Hospital for Sick Children, Toronto, Canada
- Department of Medicine, University of Toronto, Toronto, Canada
| | - Nicolas G Gottardo
- Department of Paediatric and Adolescent Oncology/Haematology, Perth Children's Hospital, Perth, Australia
- Brain Tumour Research Program, Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - Andrew S Moore
- Oncology Service, Queensland Children's Hospital, Children's Health Queensland Hospital and Health Service, Brisbane, Australia
- Child Health Research Centre, The University of Queensland, Brisbane, Australia
| | - Joanne F Aitken
- Viertel Cancer Research Centre, Cancer Council Queensland, Brisbane, Australia
- School of Public Health and Social Work, Queensland University of Technology, Brisbane, Australia
- School of Public Health, The University of Queensland, Brisbane, Australia
| |
Collapse
|
17
|
Wang P, Li T, Liu Z, Jin M, Su Y, Zhang J, Jing H, Zhuang H, Li F. [ 18F]MFBG PET/CT outperforming [ 123I]MIBG SPECT/CT in the evaluation of neuroblastoma. Eur J Nucl Med Mol Imaging 2023; 50:3097-3106. [PMID: 37160439 DOI: 10.1007/s00259-023-06221-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/02/2023] [Indexed: 05/11/2023]
Abstract
PURPOSE Iodine 123 labeled meta-iodobenzylguanidine ([123I]MIBG) scan with SPECT/CT imaging is one of the most commonly used imaging modalities in the evaluation of neuroblastoma. [18F]-meta-fluorobenzylguanidine ([18F]MFBG) is a novel positron emission tomography (PET) tracer which was reported to have a similar biodistribution to [123I]MIBG. However, the experience of using [18F]MFBG PET/CT in the evaluation of patients with neuroblastoma is limited. This preliminary investigation aims to assess the efficacy of [18F]MFBG PET/CT in the evaluation of neuroblastomas in comparison to [123I]MIBG scans with SPECT/CT. MATERIALS AND METHODS In this prospective, single-center study, 40 participants (mean age 6.0 ± 3.7 years) with history of neuroblastoma were enrolled. All children underwent both [123I]MIBG SPECT/CT and [18F]MFBG PET/CT studies. The number of lesions and the Curie scores revealed by each imaging method were recorded. RESULTS Six patients had negative findings on both [123I]MIBG and [18F]MFBG studies. Four of the 34 patients (11.8%) were negative on [123I]MIBG but positive on [18F]MFBG, while 30 patients were positive on both [123I]MIBG and [18F]MFBG studies. In these 34 patients, [18F]MFBG PET/CT identified 784 lesions while [123I]MIBG SPECT/CT detected 532 lesions (p < 0.001). The Curie scores obtained from [18F]MFBG PET/CT (11.32 ± 8.18, range 1-27) were statistically higher (p < 0.001) than those from [123I]MIBG SPECT/CT (7.74 ± 7.52, range 0-26). 30 of 34 patients (88.2%) with active disease on imaging had higher Curie scores based on the [18F]MFBG study than on the [123I]MIBG imaging. CONCLUSION [18F]MFBG PET/CT shows higher lesion detection rate than [123I]MIBG SPECT/CT in the evaluation of pediatric patients with neuroblastoma. CLINICAL TRIAL REGISTRATION Clinicaltrials.gov : NCT05069220 (Registered: 25 September 2021, retrospectively registered); Institute Review Board of Peking Union Medical College Hospital: ZS-2514.
Collapse
Affiliation(s)
- Peipei Wang
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, People's Republic of China
| | - Tuo Li
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, People's Republic of China
| | - Zhikai Liu
- Department of Radiation Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College, Beijing, People's Republic of China
| | - Mei Jin
- Department of Medical Oncology, Pediatric Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, People's Republic of China
| | - Yan Su
- Department of Medical Oncology, Pediatric Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, People's Republic of China.
| | - Jingjing Zhang
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hongli Jing
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, People's Republic of China.
| | - Hongming Zhuang
- Department of Radiology, Children's Hospital of Philadelphia University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Fang Li
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, People's Republic of China.
| |
Collapse
|
18
|
Karalexi MA, Servitzoglou M, Papadakis V, Kachanov D, Česen Mazič M, Baka M, Moschovi M, Kourti M, Polychronopoulou S, Stiakaki E, Hatzipantelis E, Dana H, Stefanaki K, Malama A, Themistocleous MS, Strantzia K, Shamanskaya T, Bouka P, Panagopoulou P, Kantzanou M, Ntzani E, Dessypris N, Petridou ET. Survival patterns of childhood neuroblastoma: an analysis of clinical data from Southern-Eastern European countries. Eur J Cancer Prev 2023; 32:254-263. [PMID: 32925511 DOI: 10.1097/cej.0000000000000614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The prognosis of children with neuroblastoma (NBL) can be dismal with significant variations depending on the stage and biology of the tumor. We assessed the event-free (EFS) and overall (OS) survival using harmonized data from three Southern-Eastern European (SEE) countries. Data for 520 incident NBL cases (2009-2018) were collected from Greece, Slovenia and Russia. Kaplan-Meier curves were fitted, and EFS/OS were derived from Cox proportional models by study variables including the protocol-based risk-group (low/observation, intermediate, high). Over one-third of cases were coded in the high-risk group, of which 23 children (4.4%) received treatment with anti-ganglioside 2 (GD2) mAb. Survival rates were inferior in older (OS 5-year; 1.5-4.9 years: 61%; EFS 5-year; 1.5-4.9 years: 48%) compared to children younger than 1.5 years (OS 5-year; <1.5 years: 91%; EFS 5-year; <1.5 years: 78%). Predictors of poor OS included stage 4 (hazard ratio, HR OS : 18.12, 95% confidence intervals, CI: 3.47-94.54), N-myc amplification (HR OS : 2.16, 95% CI: 1.40-3.34), no surgical excision (HR OS : 3.27, 95% CI: 1.91-5.61) and relapse/progression (HR OS : 5.46, 95% CI: 3.23-9.24). Similar unfavorable EFS was found for the same subsets of patients. By contrast, treatment with anti-GD2 antibody in high-risk patients was associated with decreased risk of death or unfavorable events (HR OS : 0.11, 95% CI: 0.02-0.79; HR EFS : 0.19, 95% CI: 0.07-0.52). Our results confirm the outstanding prognosis of the early NBL stages, especially in children <1.5 years, and the improved outcomes of the anti-GD2 treatment in high-risk patients. Ongoing high-quality clinical cancer registration is needed to ensure comparability of survival across Europe and refine our understanding of the NBL biology.
Collapse
Affiliation(s)
- Maria A Karalexi
- Department of Hygiene, Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens
| | - Marina Servitzoglou
- Department of Pediatric Hematology-Oncology, 'Pan. & Agl. Kyriakou' Children's Hospital
| | - Vassilios Papadakis
- Department of Pediatric Hematology-Oncology, 'Agia Sofia' Children's Hospital, Athens, Greece
| | - Denis Kachanov
- Department of Clinical Oncology, Federal Scientific and Clinical Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, Moscow, Russian Federation
| | - Maja Česen Mazič
- Department for Pediatric Hematology and Oncology, University Children Hospital Ljubljana, Ljubljana, Slovenia
| | - Margaret Baka
- Department of Pediatric Hematology-Oncology, 'Pan. & Agl. Kyriakou' Children's Hospital
| | - Maria Moschovi
- Pediatric Hematology/Oncology Unit, First Department of Pediatrics, University of Athens, 'Agia Sofia' Children's Hospital, Athens
| | - Maria Kourti
- Department of Pediatric Hematology and Oncology, Hippokration Hospital, Thessaloniki
| | - Sofia Polychronopoulou
- Department of Pediatric Hematology-Oncology, 'Agia Sofia' Children's Hospital, Athens, Greece
| | - Eftichia Stiakaki
- Department of Pediatric Hematology-Oncology, University of Crete, Heraklion
| | - Emmanuel Hatzipantelis
- Hematology-Oncology Unit, 2nd Pediatric Department, Aristotle University of Thessaloniki, AHEPA Hospital, Thessaloniki
| | - Helen Dana
- Pediatric Hematology-Oncology Department, 'Mitera' Children's Hospital
| | | | - Astero Malama
- Department of Imaging, National and Kapodistrian University of Athens
| | | | | | - Tatyana Shamanskaya
- Department of Clinical Oncology, Federal Scientific and Clinical Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, Moscow, Russian Federation
| | - Panagiota Bouka
- Department of Hygiene, Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens
| | - Paraskevi Panagopoulou
- Department of Hygiene, Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens
| | - Maria Kantzanou
- Department of Hygiene, Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens
| | - Evangelia Ntzani
- Department of Hygiene and Epidemiology, School of Medicine, University of Ioannina, Ioannina, Greece
- Center for Evidence Synthesis in Health, Brown University School of Public Health, Providence, RI, USA
| | - Nick Dessypris
- Department of Hygiene, Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens
| | - Eleni Th Petridou
- Department of Hygiene, Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens
- Clinical Epidemiology Unit, Department of Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
19
|
Abele N, Langner S, Felbor U, Lode H, Hosten N. Quantitative Diffusion-Weighted MRI of Neuroblastoma. Cancers (Basel) 2023; 15:cancers15071940. [PMID: 37046600 PMCID: PMC10092990 DOI: 10.3390/cancers15071940] [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: 01/26/2023] [Revised: 03/06/2023] [Accepted: 03/08/2023] [Indexed: 04/14/2023] Open
Abstract
Neuroblastoma is the most common extracranial, malignant, solid tumor found in children. In more than one-third of cases, the tumor is in an advanced stage, with limited resectability. The treatment options include resection, with or without (neo-/) adjuvant therapy, and conservative therapy, the latter even with curative intent. Contrast-enhanced MRI is used for staging and therapy monitoring. Diffusion-weighted imaging (DWI) is often included. DWI allows for a calculation of the apparent diffusion coefficient (ADC) for quantitative assessment. Histological tumor characteristics can be derived from ADC maps. Monitoring the response to treatment is possible using ADC maps, with an increase in ADC values in cases of a response to therapy. Changes in the ADC value precede volume reduction. The usual criteria for determining the response to therapy can therefore be supplemented by ADC values. While these changes have been observed in neuroblastoma, early changes in the ADC value in response to therapy are less well described. In this study, we evaluated whether there is an early change in the ADC values in neuroblastoma under therapy; if this change depends on the form of therapy; and whether this change may serve as a prognostic marker. We retrospectively evaluated neuroblastoma cases treated in our institution between June 2007 and August 2014. The examinations were grouped as 'prestaging'; 'intermediate staging'; 'final staging'; and 'follow-up'. A classification of "progress", "stable disease", or "regress" was made. For the determination of ADC values, regions of interest were drawn along the borders of all tumor manifestations. To calculate ADC changes (∆ADC), the respective MRI of the prestaging was used as a reference point or, in the case of therapies that took place directly after previous therapies, the associated previous staging. In the follow-up examinations, the previous examination was used as a reference point. The ∆ADC were grouped into ∆ADCregress for regressive disease, ∆ADCstable for stable disease, and ∆ADC for progressive disease. In addition, examinations at 60 to 120 days from the baseline were grouped as er∆ADCregress, er∆ADCstable, and er∆ADCprogress. Any differences were tested for significance using the Mann-Whitney test (level of significance: p < 0.05). In total, 34 patients with 40 evaluable tumor manifestations and 121 diffusion-weighted MRI examinations were finally included. Twenty-seven patients had INSS stage IV neuroblastoma, and seven had INSS stage III neuroblastoma. A positive N-Myc expression was found in 11 tumor diseases, and 17 patients tested negative for N-Myc (with six cases having no information). 26 patients were assigned to the high-risk group according to INRG and eight patients to the intermediate-risk group. There was a significant difference in mean ADC values from the high-risk group compared to those from the intermediate-risk group, according to INRG. The differences between the mean ∆ADC values (absolute and percentage) according to the course of the disease were significant: between ∆ADCregress and ∆ADCstable, between ∆ADCprogress and ∆ADCstable, as well as between ∆ADCregress and ∆ADCprogress. The differences between the mean er∆ADC values (absolute and percentage) according to the course of the disease were significant: between er∆ADCregress and er∆ADCstable, as well as between er∆ADCregress and er∆ADCprogress. Forms of therapy, N-Myc status, and risk groups showed no further significant differences in mean ADC values and ∆ADC/er∆ADC. A clear connection between the ADC changes and the response to therapy could be demonstrated. This held true even within the first 120 days after the start of therapy: an increase in the ADC value corresponds to a probable response to therapy, while a decrease predicts progression. Minimal or no changes were seen in cases of stable disease.
Collapse
Affiliation(s)
- Niklas Abele
- Department of Radiology, Germany University of Greifswald, 17475 Greifswald, Germany
- Institute of Pathology, University of Erlangen, 91054 Erlangen, Germany
| | - Soenke Langner
- Department of Radiology, Germany University of Greifswald, 17475 Greifswald, Germany
- Department of Radiology, University of Rostock, 18057 Rostock, Germany
| | - Ute Felbor
- Department of Human Genetics, University of Greifswald, 17475 Greifswald, Germany
- Interfaculty Institute of Genetics and Functional Genetics, University of Greifswald, 17475 Greifswald, Germany
| | - Holger Lode
- Department of Pediatric Hematology and Oncology, University of Greifswald, 17475 Greifswald, Germany
| | - Norbert Hosten
- Department of Radiology, Germany University of Greifswald, 17475 Greifswald, Germany
| |
Collapse
|
20
|
Awounou D, Lacour B, Desandes E, Guissou S, Cassoux N, Doz F, Dufour C, Minard-Colin V, Schleiermacher G, Taque S, Verschuur A, Clavel J, Goujon S. Seasonality of main childhood embryonal tumours and rhabdomyosarcoma, France, 2000-2015. Cancer Med 2023; 12:8789-8803. [PMID: 36726302 PMCID: PMC10134357 DOI: 10.1002/cam4.5624] [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: 07/22/2022] [Revised: 12/13/2022] [Accepted: 01/03/2023] [Indexed: 02/03/2023] Open
Abstract
Few studies have investigated the seasonal patterns of embryonal tumours. Based on data from the French National Registry of Childhood Cancers, the present study aimed to investigate seasonal variations in embryonal tumour incidence rates by month of birth and by month of diagnosis. The study included 6635 primary embryonal tumour cases diagnosed before the age of 15 years over the period 2000-2015 in mainland France. Assuming monthly variations in incidence rates were homogeneous over 2000-2015, we used a Poisson regression model to test for overall heterogeneity in standardised incidence ratios (SIRs) by month of birth or diagnosis. The seasonal scan statistic method was used to detect monthly excesses or deficits of embryonal tumour cases over the whole study period. The annual reproducibility of the observed monthly variations was formally tested. An overall heterogeneity in incidence rates by month of birth was observed for rhabdomyosarcoma in boys only. Based on the month of diagnosis, a seasonality was evidenced for unilateral retinoblastoma, with a lower incidence rate in the summer (SIRJul-Aug = 0.68, 95% CI = 0.52-0.87), whilst the incidence rate of rhabdomyosarcoma tended to be lower in August (SIRAug = 0.68, 95% CI = 0.52-0.89). No seasonality was detected for the other embryonal tumour groups by month of birth or month of diagnosis. This study is one of the largest to have investigated the seasonality of childhood embryonal tumours. The study showed a seasonal variation in the incidence rates by month of diagnosis for unilateral retinoblastoma and rhabdomyosarcoma. Our findings are likely to reflect a delay in consultation during the summer months. However, the role of seasonally varying environmental exposures cannot be ruled out.
Collapse
Affiliation(s)
- Danielle Awounou
- Inserm, UMR 1153 Centre of Research in Epidemiology and StatisticS (CRESS), Epidemiology of childhood and adolescent cancers team (EPICEA), Villejuif, France.,Université Paris Cité, Paris, France
| | - Brigitte Lacour
- Inserm, UMR 1153 Centre of Research in Epidemiology and StatisticS (CRESS), Epidemiology of childhood and adolescent cancers team (EPICEA), Villejuif, France.,Université Paris Cité, Paris, France.,French National Registry of Childhood Solid Tumours (RNTSE), CHU Nancy, Nancy, France
| | - Emmanuel Desandes
- Inserm, UMR 1153 Centre of Research in Epidemiology and StatisticS (CRESS), Epidemiology of childhood and adolescent cancers team (EPICEA), Villejuif, France.,Université Paris Cité, Paris, France.,French National Registry of Childhood Solid Tumours (RNTSE), CHU Nancy, Nancy, France
| | - Sandra Guissou
- Inserm, UMR 1153 Centre of Research in Epidemiology and StatisticS (CRESS), Epidemiology of childhood and adolescent cancers team (EPICEA), Villejuif, France.,Université Paris Cité, Paris, France.,French National Registry of Childhood Solid Tumours (RNTSE), CHU Nancy, Nancy, France
| | - Nathalie Cassoux
- Université Paris Cité, Paris, France.,Department of Surgical Oncology, Institut Curie, Paris, France
| | - François Doz
- Université Paris Cité, Paris, France.,SIREDO Centre (Care, Innovation, Research In Pediatric, Adolescent and Young Adult Oncology), Institut Curie, Paris, France
| | - Christelle Dufour
- Department of Paediatric and Adolescent Oncology, Institut Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Véronique Minard-Colin
- Department of Paediatric and Adolescent Oncology, Institut Gustave Roussy, Université Paris Saclay, Villejuif, France.,Inserm, UMR 1015, Université Paris Saclay, Villejuif, France
| | - Gudrun Schleiermacher
- SIREDO Centre (Care, Innovation, Research In Pediatric, Adolescent and Young Adult Oncology), Institut Curie, Paris, France
| | - Sophie Taque
- Department of Paediatrics, CHU Rennes, Rennes, France
| | - Arnauld Verschuur
- Department of Paediatric Haematology, Immunology and Oncology, Children Hospital of La Timone, APHM, Marseille, France
| | - Jacqueline Clavel
- Inserm, UMR 1153 Centre of Research in Epidemiology and StatisticS (CRESS), Epidemiology of childhood and adolescent cancers team (EPICEA), Villejuif, France.,Université Paris Cité, Paris, France.,French National Registry of Childhood Haematological Malignancies (RNHE), Villejuif, France
| | - Stéphanie Goujon
- Inserm, UMR 1153 Centre of Research in Epidemiology and StatisticS (CRESS), Epidemiology of childhood and adolescent cancers team (EPICEA), Villejuif, France.,Université Paris Cité, Paris, France.,French National Registry of Childhood Haematological Malignancies (RNHE), Villejuif, France
| |
Collapse
|
21
|
Li Q, Wang J, Cheng Y, Hu A, Li D, Wang X, Guo Y, Zhou Y, Chen G, Bao B, Gao H, Song J, Du X, Zheng L, Tong Q. Long-Term Survival of Neuroblastoma Patients Receiving Surgery, Chemotherapy, and Radiotherapy: A Propensity Score Matching Study. J Clin Med 2023; 12:jcm12030754. [PMID: 36769402 PMCID: PMC9918249 DOI: 10.3390/jcm12030754] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/01/2022] [Accepted: 12/08/2022] [Indexed: 01/20/2023] Open
Abstract
Neuroblastoma is the most common extracranial solid malignancy in children. This study was undertaken to determine the long-term survival of neuroblastoma patients receiving conventional therapeutics (surgery, chemotherapy, and radiotherapy). The neuroblastoma patients examined were registered in the Surveillance, Epidemiology and End Results (SEER) database (1975-2016). Using propensity score matching analysis, the patients were paired by record depending on whether they received surgery, chemotherapy, or radiotherapy. Univariate and multivariate analyses of the disease-specific survival of the paired patients were performed by the log-rank test and Cox regression assay. A total of 4568 neuroblastoma patients were included in this study. During 1975-2016, the proportion of histopathological grade III/IV cases receiving surgery gradually increased, while the number of patients with tumors of grade I to IV undergoing chemotherapy or radiotherapy was stable or even decreased. After propensity score analysis, for Grade I + II and Grade III tumors, surgery obviously improved the disease-specific survival of patients, while chemotherapy was unfavorable for patient prognosis, and radiotherapy exerted no obvious effect on the patients. However, no matter what treatment was chosen, the patients with advanced-histopathological-grade tumors had a poor prognosis. Meanwhile, for all histopathological grades, the patients receiving surgery and subsequent chemotherapy or radiotherapy suffered from worsen disease-specific survival than those simply undergoing surgery. Fortunately, the negative effects of surgery, chemotherapy, or radiotherapy improved gradually over time. Surgery improved the long-term survival of the neuroblastoma patients, while chemotherapy and radiotherapy exerted an unfavorable impact on patient outcome. These results provide an important reference for the clinical treatment of neuroblastoma.
Collapse
Affiliation(s)
- Qilan Li
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Jianqun Wang
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Yang Cheng
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Anpei Hu
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Dan Li
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Xiaojing Wang
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Yanhua Guo
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Yi Zhou
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Guo Chen
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Banghe Bao
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Haiyang Gao
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Jiyu Song
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Xinyi Du
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Liduan Zheng
- Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Correspondence: (L.Z.); (Q.T.); Tel.: +86-27-8572-6129 (L.Z.); +86-27-8535-0762 (Q.T.); Fax: +86-27-8572-6821 (L.Z. & Q.T.)
| | - Qiangsong Tong
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Correspondence: (L.Z.); (Q.T.); Tel.: +86-27-8572-6129 (L.Z.); +86-27-8535-0762 (Q.T.); Fax: +86-27-8572-6821 (L.Z. & Q.T.)
| |
Collapse
|
22
|
Liu Q, Wang S, Chu P, Sun J, Jin Y, Guo Y, Ma X, He L, Su Y, Zhang J, Peng Y, Li Y, Zhang X, Sun N, Liu Z, Ni X. Clinical and surgical outcome differences on the basis of pathology category in cervical neuroblastic tumors. J Pediatr Surg 2022; 57:926-933. [PMID: 35961818 DOI: 10.1016/j.jpedsurg.2022.07.008] [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: 02/22/2022] [Revised: 06/20/2022] [Accepted: 07/01/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND Cervical neuroblastic tumors (NTs) are rare but less aggressive cancer with an above-average survival rate. Little has been published regarding the management and surgical outcomes of patients with cervical NTs based on pathology category. This study compared and identified the preoperative characteristics of cervical NTs in different pathology categories and evaluated the outcomes of patients undergoing surgical resection. MATERIALS AND METHODS Upon the institutional review board's approval, a retrospective chart review was performed at Beijing Children's Hospital from April 2013 to August 2020. Demographics of patients, imaging data, lab test results, operation details and outcomes were recorded and analyzed. RESULTS Of 32 cervical NTs, 24(80%) were classified as neuroblastoma (NB) /ganglioneuroblastoma-nodular (GNBn) and 8(20%) as ganglioneuroblastoma-intermixed (GNBi)/ ganglioneuroma (GN). Patients with GNBi/GN were older than those with NB/GNBn (44.5 months (IQR 16-81) vs 9 months (IQR 1-47); P = 0.001). GNBi/GN patients presented more frequently with stage 1 disease compared with NB/GNBn patients (100% vs. 29.2%, P = 0.001), less frequently with tumor-related symptoms (0% vs. 70.8%, P = 0.001), artery encased tumor (0% vs. 41.7%, P = 0.035), and surgical complications (25% vs. 70.8%, P = 0.038). GNBi/GN patients were also less likely to show elevated neuron specific enolase (NSE) (12.5% vs. 79.2%, P = 0.002). CONCLUSIONS Cervical NB/GNBn and GNBi/GN patients had distinct characteristic clinical presentations and surgical outcomes. For children with features suggestive of benign disease (older age, asymptomatic, normal serum tumor markers) and no artery image-defined risk factors (IDRFs), upfront resection can be considered.
Collapse
Affiliation(s)
- Qiaoyin Liu
- Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Shengcai Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Ping Chu
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology Head and Neck Surgery, Beijing Pediatric Research Institute, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Jihang Sun
- Imaging center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yaqiong Jin
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology Head and Neck Surgery, Beijing Pediatric Research Institute, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yongli Guo
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology Head and Neck Surgery, Beijing Pediatric Research Institute, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Xiaoli Ma
- Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Lejian He
- Department of Pathology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yan Su
- Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Jie Zhang
- Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yun Peng
- Imaging center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yanzhen Li
- Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Xuexi Zhang
- Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Nian Sun
- Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Zhiyong Liu
- Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Xin Ni
- Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China.
| |
Collapse
|
23
|
Ghasemi A, Ghaffari K, Gohari A, Eghbali A, Yousefichaijan P, Falahati V. Kidney Failure in Children with Wilms Tumor: A Study Based on Urine Analysis and Ultrasound. Adv Biomed Res 2022; 11:89. [PMID: 36518858 PMCID: PMC9744077 DOI: 10.4103/abr.abr_367_21] [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: 11/20/2021] [Revised: 12/26/2021] [Accepted: 12/27/2021] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Renal insufficiency is one of the inevitable complications in patients with Wilms tumor (WT). The purpose of this study was to assess the renal function in children with WT at baseline and every 3 months to 2 years. MATERIALS AND METHODS In a descriptive-analytical study from 2018 to 2020, 48 children with WT were included in the study. Urine creatinine (UCr), serum calcium (SCr), blood pressure (BP), estimated glomerular filtration rate (eGFR), and urinary protein (UPro) were evaluated at baseline and every 3 months during the study. Spot UCa/UCr and spot UPro/UCr ratio were calculated. Kidney ultrasonography was used in all patients. Independent Sample t-test and Chi-square tests were utilized to compare age and sex, respectively. RESULTS The mean age of patients at follow-up was 7.3 years. There was no significant difference in mean UCr, SCr, eGFR, 24-h UPro, UCa/UCr ratio, and spot UPro/UCr ratio at baseline and end of study (P baseline> 0.05, P end of study> 0.05). Analysis of kidney size showed a statistical association with tumor stage (P < 0.05). Comparison of the kidney size in patients showed that there is a statistically significant difference (P < 0.0001) at baseline and end of the study. CONCLUSION This study showed that as WT progressed, the size of the kidneys increases without any renal insufficiency. Therefore, it seems that urinalysis of patients with WT along with sonography is necessary to determine renal insufficiency and the use of ultrasound alone to determine kidney insufficiency is not recommended.
Collapse
Affiliation(s)
- Ali Ghasemi
- Department of Biochemistry and Hematology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Kazem Ghaffari
- Department of Basic and Laboratory Sciences, Khomein University of Medical Sciences, Khomein, Iran
| | - Alireza Gohari
- Clinical Research Development Center of Amir Kabir Hospital, Arak University of Medical Sciences, Arak, Iran
| | - Aziz Eghbali
- Department of Pediatric Nephrology, Arak University of Medical Sciences, Arak, Iran
| | - Parsa Yousefichaijan
- Clinical Research Development Center of Aliasghar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Vahid Falahati
- Clinical Research Development Center of Amir Kabir Hospital, Arak University of Medical Sciences, Arak, Iran,Address for correspondence: Dr. Vahid Falahati, Clinical Research Development Center of Amir Kabir Hospital, Arak University of Medical Sciences, Arak, Iran. E-mail:
| |
Collapse
|
24
|
van Heerden J, Kruger M, Esterhuizen TM, van Zyl A, Hendricks M, Cox S, Mangray H, Poole J, Naidu G, Büchner A, de Villiers M, du Plessis J, van Emmenes B, Matthews E, Manickchund Y, Harrison DS. The pre-surgical factors that determine the decision to proceed to resection in children diagnosed with high-risk neuroblastoma in a resource limited setting. Pediatr Hematol Oncol 2022; 40:242-257. [PMID: 36271813 DOI: 10.1080/08880018.2022.2137610] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Surgical control has prognostic value in neuroblastoma (NB). Advanced NB is common at diagnosis in South Africa. We investigated the pre-surgery factors that influenced decisions to perform surgical resections. We included 204 patients with high-risk NB from a national retrospective study, who completed induction chemotherapy between 2000 and 2016.The median age was 32.4 months (IQR 15.1 - 53.5 months). Primary tumor resection was achieved in 76.9% of patients between 0-18 months of age, 51.8% between 18-60 months and 51.7% older than 60 months (p < 0.001). Only 43.2% of patients with distant metastatic disease had surgery done (p < 0.001). LDH was >750 U/L in 46.8% and ferritin >120 g/dL in 53.1% of those who had surgery (p = 0.005). The majority (80.4%), who had achieved post-induction metastatic complete remission (mCR), were operated, while 28.7% without mCR had surgery (p < 0.001). The long-term overall survival in patients with mCR and primary tumor resection was 36.5% compared to those with mCR without primary tumor resection (25.4%) and without mCR (≤3.0%)(p < 0.001). Age (p < 0.001), stage (p < 0.001), mCR (p < 0.001) and treatment setting (p < 0.001) were of prognostic significance. The tumor site and MYCN-amplification did not significantly predict resection rates. Post-induction mCR and stage were associated with surgical resection and five-year OS (p < 0.001) on multivariate analysis.Patients with high-risk NB who achieved mCR and had primary tumor resections are curable in limited resourced settings. Stage and post-induction mCR were significant variables that led to surgery. These variables should be included as indications in the management of metastatic NB in resource limited settings.
Collapse
Affiliation(s)
- Jaques van Heerden
- Paediatric Haematology and Oncology, Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg Hospital, Cape Town, South Africa.,Paediatric Haematology and Oncology, Department of Paediatrics, Antwerp University Hospital, Antwerp, Belgium.,Department of Medicine, Molecular imaging, Pathology, Radiotherapy and Oncology (MIPRO), University of Antwerp, Antwerp, Belgium
| | - Mariana Kruger
- Paediatric Haematology and Oncology, Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg Hospital, Cape Town, South Africa
| | - Tonya Marianne Esterhuizen
- Division of Epidemiology and Biostatistics, Faculty of Medicine and Health Sciences, Stellenbosch University, South Africa
| | - Anel van Zyl
- Paediatric Haematology and Oncology, Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg Hospital, Cape Town, South Africa
| | - Marc Hendricks
- Department of Paediatrics and Child Health, Faculty of Health Sciences, University of Cape Town.,Paediatric Haematology and Oncology Service, Red Cross War Memorial Children's Hospital, South Africa
| | - Sharon Cox
- Division of Paediatric Surgery, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa
| | - Hansraj Mangray
- Department of Paediatric Surgery, University of Kwa-Zulu Natal, Greys Hospital, Pietermaritzburg, 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, Johannesburg, 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
| | - Ané Büchner
- Paediatric Haematology and Oncology, Department of Paediatrics, University of Pretoria, Steve Biko Academic Hospital, Pretoria, South Africa
| | - Mariza de Villiers
- Department of Paediatric Surgery, University of Pretoria, Steve Biko Academic Hospital, Pretoria, 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, South Africa
| | - Barry van Emmenes
- Division of Paediatric Haematology and Oncology Hospital, Department of Paediatrics, Walter Sisulu University, South Africa
| | - Elmarie Matthews
- Paediatric Haematology Oncology, Department of Paediatrics and Child Health, Port Elizabeth Provincial Hospital, Walter Sisulu University, Port Elizabeth, South Africa
| | - Yashoda Manickchund
- Department of Paediatric Surgery, Walter Sisulu University, East London, South Africa
| | - Derek Stanley Harrison
- Department of Paediatric Surgery, Faculty of Health Sciences, University of the Witwatersrand, Chris Hani Baragwanath Academic Hospital, Johannesburg, South Africa
| |
Collapse
|
25
|
Pedersen C, Aboian M, McConathy JE, Daldrup-Link H, Franceschi AM. PET/MRI in Pediatric Neuroimaging: Primer for Clinical Practice. AJNR Am J Neuroradiol 2022; 43:938-943. [PMID: 35512826 DOI: 10.3174/ajnr.a7464] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/13/2021] [Indexed: 11/07/2022]
Abstract
Modern pediatric imaging seeks to provide not only exceptional anatomic detail but also physiologic and metabolic information of the pathology in question with as little radiation penalty as possible. Hybrid PET/MR imaging combines exquisite soft-tissue information obtained by MR imaging with functional information provided by PET, including metabolic markers, receptor binding, perfusion, and neurotransmitter release data. In pediatric neuro-oncology, PET/MR imaging is, in many ways, ideal for follow-up compared with PET/CT, given the superiority of MR imaging in neuroimaging compared with CT and the lower radiation dose, which is relevant in serial imaging and long-term follow-up of pediatric patients. In addition, although MR imaging is the main imaging technique for the evaluation of spinal pathology, PET/MR imaging may provide useful information in several clinical scenarios, including tumor staging and follow-up, treatment response assessment of spinal malignancies, and vertebral osteomyelitis. This review article covers neuropediatric applications of PET/MR imaging in addition to considerations regarding radiopharmaceuticals, imaging protocols, and current challenges to clinical implementation.
Collapse
Affiliation(s)
- C Pedersen
- From the Department of Radiology (C.P., M.A.), Yale School of Medicine, New Haven, Connecticut
| | - M Aboian
- From the Department of Radiology (C.P., M.A.), Yale School of Medicine, New Haven, Connecticut
| | - J E McConathy
- Division of Molecular Imaging and Therapeutics (J.E.M.), Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - H Daldrup-Link
- Department of Radiology and Pediatrics (H.D.-L.), Stanford University School of Medicine, Palo Alto, California
| | - A M Franceschi
- Neuroradiology Division (A.M.F.), Department of Radiology, Northwell Health/Donald and Barbara Zucker School of Medicine, Lenox Hill Hospital, New York, New York
| |
Collapse
|
26
|
Circulating Cell-Free DNA Assessment in Biofluids from Children with Neuroblastoma Demonstrates Feasibility and Potential for Minimally Invasive Molecular Diagnostics. Cancers (Basel) 2022; 14:cancers14092080. [PMID: 35565208 PMCID: PMC9099910 DOI: 10.3390/cancers14092080] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 04/15/2022] [Accepted: 04/19/2022] [Indexed: 12/20/2022] Open
Abstract
Simple Summary The invasive nature of surgical biopsies prevents their sequential application to monitor disease. Single biopsies fail to reflect cancer dynamics, intratumor heterogeneity, and drug sensitivities that change over time. Detection and characterization of cell-free circulating tumor DNA in biofluids from patients with solid tumors may better support disease monitoring and provide advanced molecular information for clinical decision-making toward personalized medicine. Here, we investigated the cell-free DNA characteristics in blood, bone marrow, cerebrospinal fluid, and urine provided from 84 infants and children with low-, intermediate-, or high-risk neuroblastoma. We report characteristic size distribution and concentration patterns for each biofluid to provide information to support the development of successful liquid biopsy biobanking strategies. We investigate potential correlations between disease activity and cfDNA concentration and provide strong evidence that markers specific for neuroblastoma can be detected in very small blood volumes from infants. Abstract Liquid biopsy strategies in pediatric patients are challenging due to low body weight. This study investigated cfDNA size distribution and concentration in blood, bone marrow, cerebrospinal fluid, and urine from 84 patients with neuroblastoma classified as low (n = 28), intermediate (n = 6), or high risk (n = 50) to provide key data for liquid biopsy biobanking strategies. The average volume of blood and bone marrow plasma provided ranged between 1 and 2 mL. Analysis of 637 DNA electropherograms obtained by Agilent TapeStation measurement revealed five different major profiles and characteristic DNA size distribution patterns for each of the biofluids. The proportion of samples containing primarily cfDNA was, at 85.5%, the highest for blood plasma. The median cfDNA concentration amounted to 6.28 ng/mL (blood plasma), 58.2 ng/mL (bone marrow plasma), 0.08 ng/mL (cerebrospinal fluid), and 0.49 ng/mL (urine) in samples. Meta-analysis of the dataset demonstrated that multiple cfDNA-based assays employing the same biofluid sample optimally require sampling volumes of 1 mL for blood and bone marrow plasma, 2 mL for cerebrospinal fluid, and as large as possible for urine samples. A favorable response to treatment was associated with a rapid decrease in blood-based cfDNA concentration in patients with high-risk neuroblastoma. Blood-based cfDNA concentration was not sufficient as a single parameter to indicate high-risk disease recurrence. We provide proof of concept that monitoring neuroblastoma-specific markers in very small blood volumes from infants is feasible.
Collapse
|
27
|
Castañeda A, Gorostegui M, Miralles SL, Chamizo A, Patiño SC, Flores MA, Garraus M, Lazaro JJ, Santa-Maria V, Varo A, Muñoz JP, Mora J. How we approach the treatment of patients with high-risk neuroblastoma with naxitamab: experience from the Hospital Sant Joan de Déu in Barcelona, Spain. ESMO Open 2022; 7:100462. [PMID: 35397431 PMCID: PMC9006652 DOI: 10.1016/j.esmoop.2022.100462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 03/08/2022] [Indexed: 12/01/2022] Open
Abstract
Naxitamab [humanized 3f8 (hu3F8)] is a humanized monoclonal antibody (mAb) targeting the disialoganglioside GD2. It was approved in 2020 by the United States Food and Drug Administration (FDA) in combination with granulocyte–macrophage colony-stimulating factor (GM-CSF) for treatment of pediatric and adult patients with relapsed/refractory high-risk neuroblastoma, limited to the bone or bone marrow (BM). The team at Sant Joan de Déu Children’s Hospital in Barcelona, Spain, have been using naxitamab to treat neuroblastoma under clinical trial protocols [e.g. Trial 201, and hu3F8, irinotecan, temozolomide, and sargramostim (GM-CSF) (HITS) study] and compassionate use since 2017. The team has experience with two primary regimens: naxitamab with GM-CSF only, or naxitamab in combination with irinotecan, temozolomide, and GM-CSF (chemoimmunotherapy). This article aims to provide a practical overview of the team’s experience with naxitamab to date, including preparing the treatment room and selecting the team. Adverse event management, including the use of ketamine to manage pain during anti-GD2 mAb infusions, is also discussed. We hope this will provide practical information for other health care providers considering offering this treatment. Immunotherapy with anti-GD2 antibodies has revolutionized the treatment of patients with high-risk neuroblastoma. In 2020, FDA approved naxitamab + GM-CSF for treatment of patients with R/R neuroblastoma in the bone and/or BM. Outpatient treatment with naxitamab-based immunotherapy may improve health-related quality of life. Naxitamab infusions require specific training and teamwork to prevent and efficiently manage most frequent adverse events.
Collapse
Affiliation(s)
- A Castañeda
- Sant Joan de Déu Barcelona Children's Hospital, Barcelona, Spain
| | - M Gorostegui
- Sant Joan de Déu Barcelona Children's Hospital, Barcelona, Spain
| | - S L Miralles
- Sant Joan de Déu Barcelona Children's Hospital, Barcelona, Spain
| | - A Chamizo
- Sant Joan de Déu Barcelona Children's Hospital, Barcelona, Spain
| | - S C Patiño
- Sant Joan de Déu Barcelona Children's Hospital, Barcelona, Spain
| | - M A Flores
- Sant Joan de Déu Barcelona Children's Hospital, Barcelona, Spain
| | - M Garraus
- Sant Joan de Déu Barcelona Children's Hospital, Barcelona, Spain
| | - J J Lazaro
- Sant Joan de Déu Barcelona Children's Hospital, Barcelona, Spain
| | - V Santa-Maria
- Sant Joan de Déu Barcelona Children's Hospital, Barcelona, Spain
| | - A Varo
- Sant Joan de Déu Barcelona Children's Hospital, Barcelona, Spain
| | - J P Muñoz
- Sant Joan de Déu Barcelona Children's Hospital, Barcelona, Spain
| | - J Mora
- Sant Joan de Déu Barcelona Children's Hospital, Barcelona, Spain.
| |
Collapse
|
28
|
Miao J, Wei H, Cui J, Zhang Q, Liu F, Mao Z, Zhang D. The prognosis of different distant metastases pattern in malignant tumors of the adrenal glands: A population-based retrospective study. PLoS One 2022; 17:e0264431. [PMID: 35290387 PMCID: PMC8923449 DOI: 10.1371/journal.pone.0264431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 02/09/2022] [Indexed: 11/18/2022] Open
Abstract
Introduction
The present existing data on the association of metastatic sites and prognosis of patients with metastatic adrenal malignancy are limited. This study aims to investigate the impact of different distant metastases pattern on the survival of patients with adrenal malignancy.
Methods
A dataset from the National Cancer Institute’s Surveillance, Epidemiology, and End Results (SEER) 18 Registries (2000–2017) was selected for a retrospective metastatic adrenal malignancy cohort study. There was information on distribution of metastatic lesions in bone, brain, liver, and lung in the SEER database. Kaplan-Meier analysis and nomogram analyses were applied to compare the survival distribution of cases. Univariate and multivariate cox regression models were used to analyze survival outcomes.
Results
From the SEER database, a total of 980 patients with primary metastatic adrenal malignancy from 2010 to 2017 were enrolled in this cohort study. Based on the initial metastatic sites, 42.3%, 38.4%, 30.5%, and 4.9% of patients were found bone, liver, lung, and brain metastasis, respectively. Patients who had a single site of distant metastases accounted for 52.6% (515/980) and had a better overall survival (OS) and cancer-specific survival (CSS) (both P < 0.001). In contrast with the tumor arising from the cortex, the tumor from the medulla showed better survival outcomes in both OS and CSS (P < 0.001).
Conclusion
Different histological types possess various metastatic features and prognostic values. Understanding these differences may contribute to designing targeted pre-treatment assessment of primary metastatic adrenal malignancy and creating a personalized curative intervention.
Collapse
Affiliation(s)
- Jia Miao
- Department of Urology, Taizhou First People’s Hospital, Taizhou, Zhejiang, China
| | - Haibin Wei
- Department of Urology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
- * E-mail: (HW); (DZ)
| | - Jianxin Cui
- Department of Urology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qi Zhang
- Department of Urology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Feng Liu
- Department of Urology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zujie Mao
- Department of Urology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Dahong Zhang
- Department of Urology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
- * E-mail: (HW); (DZ)
| |
Collapse
|
29
|
Abbas AA, Samkari AMN. High-Risk Neuroblastoma: Poor Outcomes Despite Aggressive Multimodal
Therapy. CURRENT CANCER THERAPY REVIEWS 2022. [DOI: 10.2174/1573394717666210805114226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
:
Neuroblastoma (NBL) is a highly malignant embryonal tumor that originates from the
primordial neural crest cells. NBL is the most common tumor in infants and the most common extracranial
solid tumor in children. The tumor is more commonly diagnosed in children of 1-4 years
of age. NBL is characterized by enigmatic clinical behavior that ranges from spontaneous regression
to an aggressive clinical course leading to frequent relapses and death. Based on the likelihood
of progression and relapse, the International Neuroblastoma Risk Group classification system categorized
NBL into very low risk, low risk, intermediate risk, and high risk (HR) groups. HR NBL is
defined based on the patient's age (> 18 months), disease metastasis, tumor histology, and MYCN
gene amplification. HR NBL is diagnosed in nearly 40% of patients, mainly those > 18 months of
age, and is associated with aggressive clinical behavior. Treatment strategies involve the use of intensive
chemotherapy (CTR), surgical resection, high dose CTR with hematopoietic stem cell support,
radiotherapy, biotherapy, and immunotherapy with Anti-ganglioside 2 monoclonal antibodies.
Although HR NBL is now better characterized and aggressive multimodal therapy is applied, the
outcomes of treatment are still poor, with overall survival and event-free survival of approximately
40% and 30% at 3-years, respectively. The short and long-term side effects of therapy are tremendous.
HR NBL carries a high mortality rate accounting for nearly 15% of pediatric cancer deaths.
However, most mortalities are attributed to the high frequency of disease relapse (50%) and disease
reactiveness to therapy (20%). Newer treatment strategies are therefore urgently needed. Recent
discoveries in the field of biology and molecular genetics of NBL have led to the identification
of several targets that can improve the treatment results. In this review, we discuss the different
aspects of the epidemiology, biology, clinical presentations, diagnosis, and treatment of HR
NBL, in addition to the recent developments in the management of the disease.
Collapse
Affiliation(s)
- Adil Abdelhamed Abbas
- College of Medicine King Saud bin Abdulaziz, University for Health Sciences Consultant Pediatric Hematology / Oncology
& BMT The Pediatric Hematology/Oncology Section Princess Nourah Oncology Centre King Abdulaziz Medical
City, Jeddah, Saudi Arabia
| | - Alaa Mohammed Noor Samkari
- College of Medicine King Saud bin Abdulaziz, University for Health Sciences Consultant
Anatomical Pathologist Department of Laboratory Medicine King Abdulaziz Medical City, Jeddah, Saudi Arabia
| |
Collapse
|
30
|
Jiang Z, Zhou X, Han L, Li F, Hao X, Dong Q, Chen X. miR-21 Targets Long Noncoding RNA PCAT29 to Promote Cell Proliferation in Neuroblastoma. Crit Rev Eukaryot Gene Expr 2022; 32:1-8. [PMID: 36017911 DOI: 10.1615/critreveukaryotgeneexpr.2022042471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Long noncoding RNA (lncRNA) PCAT29 has been characterized as a tumor suppressor in several types of cancer, although its involvement in neuroblastoma (NB) is unknown. In this study, we analyzed the role of PCAT29 in NB. In paired NB and nontumor tissues from 56 patients with NB, microRNA (miR)-21 and PCAT29 expression was determined with reverse transcription quantitative PCR. Correlation between miR-21 and PCAT29 was evaluated with linear regression. The interaction between miR-21 and PCAT29 was predicted by the IntaRNA 2.0 program. In NB cells, miR-21 and PCAT29 were overexpressed to explore their relationship. In NB cell proliferation, the roles of miR-21 and PCAT29 were analyzed with propidium iodide staining and Ki67 staining assays. The results showed that PCAT29 was downregulated and miR-21 was upregulated in NB. MiR-21 was inversely correlated with PCAT29. RNA-RNA interaction prediction revealed that miR-21 might target PCAT29. MiR-21 overexpression reduced PCAT29 expression and increased NB cell proliferation, whereas PCAT29 overexpression inhibited NB cell proliferation. PCAT29 overexpression promoted NB cell apoptosis, while miR-21 overexpression inhibited NB cell apoptosis and attenuated PCAT29 overexpression-mediated NB cell apoptosis. In conclusion, MiR-21 may target PCAT29 to promote cell apoptosis in NB.
Collapse
Affiliation(s)
- Zhong Jiang
- Department of Pediatric Surgery, the Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province 266000, People's Republic of China
| | - Xianjun Zhou
- Department of Pediatric Surgery, the Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province 266000, People's Republic of China
| | - Lulu Han
- Department of Operation Room, the Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province 266000, People's Republic of China
| | - Fujiang Li
- Department of Pediatric Surgery, the Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province 266000, People's Republic of China
| | - Xiwei Hao
- Department of Pediatric Surgery, the Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province 266000, People's Republic of China
| | - Qian Dong
- Department of Pediatric Surgery, the Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province 266000, People's Republic of China
| | - Xin Chen
- Department of Pediatric Surgery, the Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province 266000, People's Republic of China
| |
Collapse
|
31
|
Schmelz K, Toedling J, Huska M, Cwikla MC, Kruetzfeldt LM, Proba J, Ambros PF, Ambros IM, Boral S, Lodrini M, Chen CY, Burkert M, Guergen D, Szymansky A, Astrahantseff K, Kuenkele A, Haase K, Fischer M, Deubzer HE, Hertwig F, Hundsdoerfer P, Henssen AG, Schwarz RF, Schulte JH, Eggert A. Spatial and temporal intratumour heterogeneity has potential consequences for single biopsy-based neuroblastoma treatment decisions. Nat Commun 2021; 12:6804. [PMID: 34815394 PMCID: PMC8611017 DOI: 10.1038/s41467-021-26870-z] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 10/18/2021] [Indexed: 01/12/2023] Open
Abstract
Intratumour heterogeneity is a major cause of treatment failure in cancer. We present in-depth analyses combining transcriptomic and genomic profiling with ultra-deep targeted sequencing of multiregional biopsies in 10 patients with neuroblastoma, a devastating childhood tumour. We observe high spatial and temporal heterogeneity in somatic mutations and somatic copy-number alterations which are reflected on the transcriptomic level. Mutations in some druggable target genes including ALK and FGFR1 are heterogeneous at diagnosis and/or relapse, raising the issue whether current target prioritization and molecular risk stratification procedures in single biopsies are sufficiently reliable for therapy decisions. The genetic heterogeneity in gene mutations and chromosome aberrations observed in deep analyses from patient courses suggest clonal evolution before treatment and under treatment pressure, and support early emergence of metastatic clones and ongoing chromosomal instability during disease evolution. We report continuous clonal evolution on mutational and copy number levels in neuroblastoma, and detail its implications for therapy selection, risk stratification and therapy resistance.
Collapse
Affiliation(s)
- Karin Schmelz
- Charité-Universitätsmedizin Berlin, Berlin, Germany
- The German Cancer Consortium (DKTK), Partner Site Berlin, Berlin, Germany
- The German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Joern Toedling
- Charité-Universitätsmedizin Berlin, Berlin, Germany
- The German Cancer Consortium (DKTK), Partner Site Berlin, Berlin, Germany
- The German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Matt Huska
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Maja C Cwikla
- Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | | | - Jutta Proba
- Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Peter F Ambros
- Children's Cancer Research Institute, St. Anna Kinderkrebsforschung, 1090, Vienna, Austria
| | - Inge M Ambros
- Children's Cancer Research Institute, St. Anna Kinderkrebsforschung, 1090, Vienna, Austria
| | - Sengül Boral
- Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Marco Lodrini
- Charité-Universitätsmedizin Berlin, Berlin, Germany
- The German Cancer Consortium (DKTK), Partner Site Berlin, Berlin, Germany
- The German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Celine Y Chen
- Charité-Universitätsmedizin Berlin, Berlin, Germany
- Experimental and Clinical Research Center (ECRC) of the Charité and Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Martin Burkert
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Dennis Guergen
- Experimental Pharmacology and Oncology Berlin-Buch GmbH (EPO), Berlin, Germany
| | | | | | - Annette Kuenkele
- Charité-Universitätsmedizin Berlin, Berlin, Germany
- The German Cancer Consortium (DKTK), Partner Site Berlin, Berlin, Germany
- The German Cancer Research Center (DKFZ), Heidelberg, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Kerstin Haase
- Charité-Universitätsmedizin Berlin, Berlin, Germany
- The German Cancer Consortium (DKTK), Partner Site Berlin, Berlin, Germany
- The German Cancer Research Center (DKFZ), Heidelberg, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Matthias Fischer
- Department of Experimental Pediatric Oncology, Medical Faculty, University Children's Hospital of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Hedwig E Deubzer
- Charité-Universitätsmedizin Berlin, Berlin, Germany
- The German Cancer Consortium (DKTK), Partner Site Berlin, Berlin, Germany
- The German Cancer Research Center (DKFZ), Heidelberg, Germany
- Experimental and Clinical Research Center (ECRC) of the Charité and Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Falk Hertwig
- Charité-Universitätsmedizin Berlin, Berlin, Germany
- The German Cancer Consortium (DKTK), Partner Site Berlin, Berlin, Germany
- The German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Patrick Hundsdoerfer
- Charité-Universitätsmedizin Berlin, Berlin, Germany
- Helios Klinikum Berlin-Buch, Berlin, Germany
| | - Anton G Henssen
- Charité-Universitätsmedizin Berlin, Berlin, Germany.
- The German Cancer Consortium (DKTK), Partner Site Berlin, Berlin, Germany.
- The German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.
- Experimental and Clinical Research Center (ECRC) of the Charité and Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.
- Berlin Institute of Health (BIH), Berlin, Germany.
| | - Roland F Schwarz
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.
- BIFOLD-Berlin Institute for the Foundations of Learning and Data, Berlin, Germany.
| | - Johannes H Schulte
- Charité-Universitätsmedizin Berlin, Berlin, Germany.
- The German Cancer Consortium (DKTK), Partner Site Berlin, Berlin, Germany.
- The German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Berlin Institute of Health (BIH), Berlin, Germany.
| | - Angelika Eggert
- Charité-Universitätsmedizin Berlin, Berlin, Germany.
- The German Cancer Consortium (DKTK), Partner Site Berlin, Berlin, Germany.
- The German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Berlin Institute of Health (BIH), Berlin, Germany.
| |
Collapse
|
32
|
Stainczyk SA, Westermann F. Neuroblastoma-Telomere maintenance, deregulated signaling transduction and beyond. Int J Cancer 2021; 150:903-915. [PMID: 34636058 DOI: 10.1002/ijc.33839] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 09/06/2021] [Accepted: 09/27/2021] [Indexed: 11/11/2022]
Abstract
The childhood malignancy neuroblastoma belongs to the group of embryonal tumors and originates from progenitor cells of the sympathoadrenal lineage. Treatment options for children with high-risk and relapsed disease are still very limited. In recent years, an ever-growing molecular diversity was identified using (epi)-genetic profiling of neuroblastoma tumors, indicating that molecularly targeted therapies could be a promising therapeutic option. In this review article, we summarize the various molecular subtypes and genetic events associated with neuroblastoma and describe recent advances in targeted therapies. We lay a strong emphasis on the importance of telomere maintenance mechanisms for understanding tumor progression and risk classification of neuroblastoma.
Collapse
Affiliation(s)
- Sabine A Stainczyk
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany.,Neuroblastoma Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Frank Westermann
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany.,Neuroblastoma Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
33
|
YILMAZ E, SAMUR MB, ÖZCAN A, ÜNAL E, KARAKÜKÇÜ M. Transplantation for ultra high-risk neuroblastoma patients: effect of tandem autologous stem cell transplantation. JOURNAL OF HEALTH SCIENCES AND MEDICINE 2021. [DOI: 10.32322/jhsm.985592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
34
|
Genetic Alterations and Resectability Predict Outcome in Patients with Neuroblastoma Assigned to High-Risk Solely by MYCN Amplification. Cancers (Basel) 2021; 13:cancers13174360. [PMID: 34503173 PMCID: PMC8430929 DOI: 10.3390/cancers13174360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 08/19/2021] [Accepted: 08/24/2021] [Indexed: 11/26/2022] Open
Abstract
Simple Summary Currently, patients with high-risk neuroblastoma are uniformly treated with maximum therapy. This study investigated a high-risk subgroup characterized by the presence of the amplified MYCN oncogene in the tumor regardless of the stage. In contrast to the corresponding high-risk subgroup consisting of patients with metastases and age at diagnosis over 18 months, the investigated subgroup had generally a superior survival chance. However, the detection of mutations of specific genes in the tumor tissue (RAS and p53 pathway including ALK) had a strong, negative impact. These genes should be therefore also investigated in the future. Complete surgical removal of the primary tumor proved to be beneficial for high-risk neuroblastoma patients assigned to the high-risk category solely by MYCN amplification. Abstract Background: To identify variables predicting outcome in neuroblastoma patients assigned to the high-risk group solely by the presence of MYCN oncogene amplification (MNA). Methods: Clinical characteristics, genomic information, and outcome of 190 patients solely assigned to high-risk neuroblastoma by MNA were analyzed and compared to 205 patients with stage 4 neuroblastoma aged ≥18 months with MNA (control group). Results: Event-free survival (EFS) and overall survival (OS) at 10 years were 47% (95%-CI 39–54%) and 56% (95%-CI 49–63%), respectively, which was significantly better than EFS and OS of the control group (EFS 25%, 95%-CI 18–31%, p < 0.001; OS 32% 95%-CI 25–39%, p < 0.001). The presence of RAS-/p53-pathway gene alterations was associated with impaired 10-year EFS and OS (19% vs. 55%, and 19% vs. 67%, respectively; both p < 0.001). In time-dependent multivariable analyses, alterations of RAS-/p53-pathway genes and the extent of the best primary tumor resection were the only independent prognostic variables for OS (p < 0.001 and p = 0.011, respectively). Conclusions: Neuroblastoma patients attributed to high risk solely by MYCN amplification have generally a more favorable outcome. Mutations of genes of the RAS and/or p53 pathways and incomplete resection are the main risk factors predicting poor outcome.
Collapse
|
35
|
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
|
36
|
Zhang F, Tian Z, Li T, Hu X, Zhu J. Prognostic Value of c-MYC Expression in Patients with Peripheral Neuroblastic Tumors. Int J Gen Med 2021; 14:2901-2907. [PMID: 34234520 PMCID: PMC8254143 DOI: 10.2147/ijgm.s315982] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/14/2021] [Indexed: 01/08/2023] Open
Abstract
Objective Neuroblastic tumors are the most common solid tumors in children. The aim of this study was to explore the prognostic value of immunostaining for cellular-myelocytomatosis viral oncogene (c-MYC) expression in patients with peripheral neuroblastic tumors (NTs). Methods A retrospective study was conducted to compare the expression of c-MYC detected by immunohistochemistry and v-myc avian myelocytomatosis viral oncogene neuroblastoma derived homolog (MYCN) by fluorescence in situ hybridization among 177 cases of NTs and determine the associations of c-MYC and MYCN with the clinical stages, morphological types, and survival rates of NTs. Results The cases positive for c-MYC were mainly the favorable histology type in stage 3 or 4 with a poor NT prognosis, but no morphological changes related to the poor prognosis were observed in their samples under a microscope. The cases with positive c-MYC expression did not overlap those with MYCN amplification. Conclusion Positive c-MYC expression portends a poor prognosis in patients with NTs.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Otolaryngology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, People's Republic of China.,National Clinical Research Center for Child Health and Disorders, Chongqing, 400014, People's Republic of China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, People's Republic of China
| | - Zhuo Tian
- Department of Geratology, Chongqing General Hospital, Chongqing, 400014, People's Republic of China
| | - Tinghong Li
- Department of Pathology, Chongqing Medical University, Chongqing, 400016, People's Republic of China.,Institute of Neuroscience, Chongqing Medical University, Chongqing, 400016, People's Republic of China.,Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Xianjun Hu
- Department of Geratology, Chongqing General Hospital, Chongqing, 400014, People's Republic of China
| | - Jin Zhu
- Department of Pathology, Chongqing Medical University, Chongqing, 400016, People's Republic of China.,Institute of Neuroscience, Chongqing Medical University, Chongqing, 400016, People's Republic of China.,Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| |
Collapse
|
37
|
Jiang CY, Xu X, Jian BL, Zhang X, Yue ZX, Guo W, Ma XL. Chromosome 10 abnormality predicts prognosis of neuroblastoma patients with bone marrow metastasis. Ital J Pediatr 2021; 47:134. [PMID: 34108028 PMCID: PMC8190999 DOI: 10.1186/s13052-021-01085-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 05/26/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Neuroblastoma (NB) is the most common extracranial solid tumor in children. It is known for high heterogeneity and concealed onset. In recent years, the mechanism of its occurrence and development has been gradually revealed. The purpose of this study is to summarize the clinical characteristics of children with NB and abnormal chromosome 10, and to investigate the relationship between the number and structure of chromosome 10 abnormalities and NB prognosis. METHODS Chromosome G-banding was used at the time of diagnosis to evaluate the genetics of chromosomes in patients with NB and track their clinical characteristics and prognosis. All participants were diagnosed with NB in the Medical Oncology Department of the Beijing Children's Hospital from May 2015 to December 2018 and were followed up with for at least 1 year. RESULTS Of all 150 patients with bone marrow metastases, 42 were clearly diagnosed with chromosomal abnormalities. Thirteen patients showed abnormalities in chromosome 10, and chromosome 10 was the most commonly missing chromosome. These 13 patients had higher LDH and lower OS and EFS than children with chromosomal abnormalities who did not have an abnormality in chromosome 10. Eight patients had both MYCN amplification and 1p36 deletion. Two patients had optic nerve damage and no vision, and one patient had left supraorbital metastases 5 months after treatment. CONCLUSIONS The results indicated that chromosome 10 might be a new prognostic marker for NB. MYCN amplification and 1p36 deletion may be related to chromosome 10 abnormalities in NB. Additionally, NB patients with abnormal chromosome 10 were prone to orbital metastases.
Collapse
Affiliation(s)
- Chi-Yi Jiang
- Medical Oncology Department, Pediatric Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing Key Laboratory of Pediatric Hematology Oncology, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, 100045, China
| | - Xiao Xu
- Medical Oncology Department, Pediatric Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing Key Laboratory of Pediatric Hematology Oncology, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, 100045, China
| | - Bing-Lin Jian
- Medical Oncology Department, Pediatric Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing Key Laboratory of Pediatric Hematology Oncology, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, 100045, China
| | - Xue Zhang
- Medical Oncology Department, Pediatric Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing Key Laboratory of Pediatric Hematology Oncology, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, 100045, China
| | - Zhi-Xia Yue
- Medical Oncology Department, Pediatric Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing Key Laboratory of Pediatric Hematology Oncology, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, 100045, China
| | - Wei Guo
- MILS (Beijing) Medical Labortory, Beijing, China
| | - Xiao-Li Ma
- Medical Oncology Department, Pediatric Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing Key Laboratory of Pediatric Hematology Oncology, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, 100045, China.
| |
Collapse
|
38
|
Nile DL, Rae C, Walker DJ, Waddington JC, Vincent I, Burgess K, Gaze MN, Mairs RJ, Chalmers AJ. Inhibition of glycolysis and mitochondrial respiration promotes radiosensitisation of neuroblastoma and glioma cells. Cancer Metab 2021; 9:24. [PMID: 34011385 PMCID: PMC8136224 DOI: 10.1186/s40170-021-00258-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 04/13/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Neuroblastoma accounts for 7% of paediatric malignancies but is responsible for 15% of all childhood cancer deaths. Despite rigorous treatment involving chemotherapy, surgery, radiotherapy and immunotherapy, the 5-year overall survival rate of high-risk disease remains < 40%, highlighting the need for improved therapy. Since neuroblastoma cells exhibit aberrant metabolism, we determined whether their sensitivity to radiotherapy could be enhanced by drugs affecting cancer cell metabolism. METHODS Using a panel of neuroblastoma and glioma cells, we determined the radiosensitising effects of inhibitors of glycolysis (2-DG) and mitochondrial function (metformin). Mechanisms underlying radiosensitisation were determined by metabolomic and bioenergetic profiling, flow cytometry and live cell imaging and by evaluating different treatment schedules. RESULTS The radiosensitising effects of 2-DG were greatly enhanced by combination with the antidiabetic biguanide, metformin. Metabolomic analysis and cellular bioenergetic profiling revealed this combination to elicit severe disruption of key glycolytic and mitochondrial metabolites, causing significant reductions in ATP generation and enhancing radiosensitivity. Combination treatment induced G2/M arrest that persisted for at least 24 h post-irradiation, promoting apoptotic cell death in a large proportion of cells. CONCLUSION Our findings demonstrate that the radiosensitising effect of 2-DG was significantly enhanced by its combination with metformin. This clearly demonstrates that dual metabolic targeting has potential to improve clinical outcomes in children with high-risk neuroblastoma by overcoming radioresistance.
Collapse
Affiliation(s)
- Donna L Nile
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK.
- Present Address: Integrated Covid Hub North East (ICHNE) Innovation Laboratory, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, NE4 5BX, UK.
| | - Colin Rae
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - David J Walker
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK
- Present Address: School of Medicine, University of Dundee, Dundee, DD1 4HN, UK
| | | | - Isabel Vincent
- Glasgow Polyomics Facility, University of Glasgow, Glasgow, G61 1QH, UK
- Present Address: Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE, UK
| | - Karl Burgess
- Glasgow Polyomics Facility, University of Glasgow, Glasgow, G61 1QH, UK
- Present Address: School of Biological Sciences, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Mark N Gaze
- Department of Oncology, University College London Hospitals NHS Foundation Trust, London, NW1 2BU, UK
| | - Robert J Mairs
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Anthony J Chalmers
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| |
Collapse
|
39
|
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
|
40
|
Schumacher-Kuckelkorn R, Atra A, Belli ML, den Engelsman G, Fréneaux P, Gauthier A, Heijlaerts-Klever A, Scuderi F, Senent Peris L, Tewari S, Zapletal O, Ernst A, Berthold F. The reliability of bone marrow cytology as response criterion in metastatic neuroblastoma. Pediatr Blood Cancer 2021; 68:e28819. [PMID: 33245195 DOI: 10.1002/pbc.28819] [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: 09/10/2020] [Revised: 10/20/2020] [Accepted: 11/03/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND The quantitative assessment of neuroblastoma cell content in bone marrow aspirates for response evaluation has been introduced recently. Data on the concordance of interobserver reports are lacking so far. METHODS Investigators of seven European countries representing national reference or large oncological centers convened in 2016. They agreed to quantitatively assess routine bone marrow smears of the participating institutions and to discuss the discrepant results in joint meetings. RESULTS From 2017 through 2019, three cytology rounds with 24, 28, and 28 bone marrow samples were run evaluating the representativity of the smears (yes/[restricted]/no) and the presence of tumor cells (yes/no and %). The comparison of the reports using κ (Fleiss) and α (Krippendorff) statistics demonstrated no robust reliabilities. The agreement on the representativity was moderate to poor, on the presence of tumor cells moderate to good, and on the percentage of tumor cells slight to moderate. Though the value of cytology is unquestioned to detect even tiny metastatic cells in bone marrow, the investigators unanimously agreed that a reliable quantification of the tumor cell content in bone marrow smears is unrealistic. For the key issue of representativity, a new practical definition was developed. CONCLUSION For any work with bone marrow aspirates, the representativity of the material is of paramount importance. A practical definition is proposed. A reliable quantitative cytological assessment of tumor cell content in bone marrow aspirates is not feasible in metastatic neuroblastoma. Therefore, its use as response criterion should be reconsidered.
Collapse
Affiliation(s)
- Roswitha Schumacher-Kuckelkorn
- Department of Pediatric Oncology and Hematology, National Neuroblastoma Reference Cytology Lab, University of Cologne, Cologne, Germany
| | - Ayad Atra
- Department of Paediatric Oncology/Haematology, The Royal Marsden NHS Foundation Trust, Sutton, Surrey, UK
| | - Maria Luisa Belli
- Department of Haematology and Oncology, Cytomorphology Laboratory, IRCCS, G. Gaslini Institute, Genoa, Italy
| | | | - Paul Fréneaux
- Service de Pathologie, Pôle de médicine diagnosique et théranostic, Institut Curie, Paris, France
| | - Arnaud Gauthier
- Service de Pathologie, Pôle de médicine diagnosique et théranostic, Institut Curie, Paris, France
| | | | - Francesca Scuderi
- Department of Haematology and Oncology, Cytomorphology Laboratory, IRCCS, G. Gaslini Institute, Genoa, Italy
| | - Leonor Senent Peris
- Department of Hematology, Cytomorphology Laboratory, Diagnosis Unit, La Fe University and Polytechnic Hospital, Valencia, Spain.,La Fe Health Research Institute, Hematology Research Group, Valencia, Spain.,Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Sanjay Tewari
- Department of Paediatric Oncology/Haematology, The Royal Marsden NHS Foundation Trust, Sutton, Surrey, UK
| | - Ondrej Zapletal
- Department of Paediatric Haematology, University Hospital Brno, Brno, Czech Republic
| | - Angela Ernst
- Faculty of Medicine and University Hospital Cologne, Institute of Medical Statistics and Computational Biology (IMSB), University of Cologne, Cologne, Germany
| | - Frank Berthold
- Department of Pediatric Oncology and Hematology, National Neuroblastoma Reference Cytology Lab, University of Cologne, Cologne, Germany
| |
Collapse
|
41
|
Pulmonary Metastasis of Low-risk Perinatal Neuroblastoma After Resection: Implications for Surveillance. J Pediatr Hematol Oncol 2021; 43:e184-e186. [PMID: 31815890 DOI: 10.1097/mph.0000000000001693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 11/08/2019] [Indexed: 11/25/2022]
Abstract
In the wake of the Children's Oncology Group (COG) ANBL00P2 trial and the ongoing ANBL1232 trial, an increasing number of children with neonatal neuroblastoma are being managed nonoperatively. We report the case of a patient with low-risk, non-MYCN amplified, neuroblastoma that was diagnosed and resected in the neonatal period but subsequently developed pulmonary metastases by the age of 7 months. Though rare, the possibility of low-risk disease metastasizing during surveillance should be recognized and may not be identified by current protocols.
Collapse
|
42
|
The Potential of Mesenchymal Stromal Cells in Neuroblastoma Therapy for Delivery of Anti-Cancer Agents and Hematopoietic Recovery. J Pers Med 2021; 11:jpm11030161. [PMID: 33668854 PMCID: PMC7996318 DOI: 10.3390/jpm11030161] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/19/2021] [Accepted: 02/21/2021] [Indexed: 02/07/2023] Open
Abstract
Neuroblastoma is one of the most common pediatric cancers and a major cause of cancer-related death in infancy. Conventional therapies including high-dose chemotherapy, stem cell transplantation, and immunotherapy approach a limit in the treatment of high-risk neuroblastoma and prevention of relapse. In the last two decades, research unraveled a potential use of mesenchymal stromal cells in tumor therapy, as tumor-selective delivery vehicles for therapeutic compounds and oncolytic viruses and by means of supporting hematopoietic stem cell transplantation. Based on pre-clinical and clinical advances in neuroblastoma and other malignancies, we assess both the strong potential and the associated risks of using mesenchymal stromal cells in the therapy for neuroblastoma. Furthermore, we examine feasibility and safety aspects and discuss future directions for harnessing the advantageous properties of mesenchymal stromal cells for the advancement of therapy success.
Collapse
|
43
|
Menke K, Schwermer M, Schramm A, Zuzak TJ. [Präklinische Untersuchungen von Wechselwirkungen zwischen Mistel und Radio- oder Chemotherapie auf pädiatrische Tumorzellen]. Complement Med Res 2021; 28:308-316. [PMID: 33621978 DOI: 10.1159/000512670] [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: 04/15/2020] [Accepted: 10/23/2020] [Indexed: 11/19/2022]
Abstract
Hintergrund: Mistelanwendungen werden als komplementäre Therapien häufig in der pädiatrischen Onkologie zusammen mit einer Radio- oder Chemotherapie verabreicht. Wechselwirkungen bei simultaner Applikation sind gerade in der Pädiatrie von großer Bedeutung, sie sind allerdings nach wie vor unzureichend untersucht. Material und Methoden: Zytotoxische Effekte des Mistelextraktes abnobaVISCUM Fraxini (aVF) auf LAN-1 Neuroblastomzellen und deren Etoposid- bzw. Cisplatin-resistente Subzelllinien wurden mittels Viabilitätstest untersucht, sowie mögliche Synergieeffekte zwischen aVF und den Chemotherapeutika durch die Softwareprodukte Combenefit und CompuSyn analysiert. Effekte einer Kombinationstherapie aus aVF und Bestrahlung auf SH-SY5Y Zellen wurden mittels Koloniebildungstest untersucht und Auswirkungen auf die Reparatureffizienz strahleninduzierter Doppelstrangbrüche mit Hilfe durchflusszytometrischer Quantifizierungen von γ-H2AX-Foci nach PI/FITC Doppelfärbung analysiert. Ergebnisse: Die Chemotherapie-resistenten LAN-1 Subzelllinien erwiesen sich als resistenter gegenüber der Mistelbehandlung als die Ursprungszelllinie. Auf Basis vier verschiedener Referenz-modelle konnten vor allem synergistisch/additive Effekte zwischen aVF und den Zytostatika Etoposid und Cisplatin berechnet werden. Die Kombination aus Mistelbehandlung und Bestrahlung führte zu einer Verringerung der Koloniebildung und zu einer Verzögerung der Reparaturgeschwindigkeit von strahleninduzierten Doppelstrangbrüchen. Schlussfolgerung: Die präklinischen Daten könnten darauf hinweisen, dass die Verwendung des Mistelextraktes, aVF, eine unterstützende Wirkung auf Radio- und Chemotherapien hat. BACKGROUND Mistletoe therapies belong to the field of complementary medicines and are often administered simultaneously or successive to conventional radio- or chemotherapy. Drug-herb interactions are of great significance, especially in pediatrics, but are still insufficiently investigated. MATERIAL AND METHODS Cytotoxic effects of the mistletoe extract, abnobaVISCUM Fraxini (aVF), on LAN-1 neuroblastoma cell line and their chemotherapy-resistant (cisplatin; etoposide) subclones were investigated by cell viability assays. Potential synergistic or antagonistic effects of the co-treatment of aVF and cisplatin or etoposide, respectively, were analyzed by Combenefit and CompuSyn software. Combinational effects of mistletoe and irradiation were assessed by colony formation assays and repair efficiency of irradiation-induced double strand breaks was investigated by flow cytometric analyses of γ-H2AX foci after PI/FITC double staining. RESULTS Chemotherapy-resistant subclones were more resistant to mistletoe therapy than the parental cells. Based on four different reference models, primarily synergistic/additive effects between aVF and the cytostatic drugs could be calculated. Simultaneous application of mistletoe extract and irradiation led to a delay of irradiation-induced double strand break repair in neuroblastoma cells and a decreased colony formation compared to irradiation monotherapy. CONCLUSION The preclinical data may indicate that the use of aVF has a supportive effect on radio- and chemotherapies.
Collapse
Affiliation(s)
- Katrin Menke
- Abteilung für pädiatrische Onkologie und Hämatologie Universitäts Kinderklinik Essen, Essen, Deutschland
| | - Melanie Schwermer
- Abteilung für pädiatrische Onkologie und Hämatologie Universitäts Kinderklinik Essen, Essen, Deutschland.,Abteilung für Kinder- und Jugendmedizin, Gemeinschaftskrankenhaus Herdecke, Herdecke, Deutschland
| | - Alexander Schramm
- Medizinische Onkologie, Westdeutsches Tumorzentrum, Universitätskrankenhaus Essen, Universität Duisburg-Essen, Essen, Deutschland
| | - Tycho Jan Zuzak
- Abteilung für Kinder- und Jugendmedizin, Gemeinschaftskrankenhaus Herdecke, Herdecke, Deutschland,
| |
Collapse
|
44
|
Hochheuser C, Windt LJ, Kunze NY, de Vos DL, Tytgat GA, Voermans C, Timmerman I. Mesenchymal Stromal Cells in Neuroblastoma: Exploring Crosstalk and Therapeutic Implications. Stem Cells Dev 2021; 30:59-78. [PMID: 33287630 PMCID: PMC7826431 DOI: 10.1089/scd.2020.0142] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023] Open
Abstract
Neuroblastoma (NB) is the second most common solid cancer in childhood, accounting for 15% of cancer-related deaths in children. In high-risk NB patients, the majority suffers from metastasis. Despite intensive multimodal treatment, long-term survival remains <40%. The bone marrow (BM) is among the most common sites of distant metastasis in patients with high-risk NB. In this environment, small populations of tumor cells can persist after treatment (minimal residual disease) and induce relapse. Therapy resistance of these residual tumor cells in BM remains a major obstacle for the cure of NB. A detailed understanding of the microenvironment and its role in tumor progression is of utmost importance for improving the treatment efficiency of NB. In BM, mesenchymal stromal cells (MSCs) constitute an important part of the microenvironment, where they support hematopoiesis and modulate immune responses. Their role in tumor progression is not completely understood, especially for NB. Although MSCs have been found to promote epithelial-mesenchymal transition, tumor growth, and metastasis and to induce chemoresistance, some reports point toward a tumor-suppressive effect of MSCs. In this review, we aim to compile current knowledge about the role of MSCs in NB development and progression. We evaluate arguments that depict tumor-supportive versus -suppressive properties of MSCs in the context of NB and give an overview of factors involved in MSC-NB crosstalk. A focus lies on the BM as a metastatic niche, since that is the predominant site for NB metastasis and relapse. Finally, we will present opportunities and challenges for therapeutic targeting of MSCs in the BM microenvironment.
Collapse
Affiliation(s)
- Caroline Hochheuser
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Laurens J. Windt
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Nina Y. Kunze
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Dieuwke L. de Vos
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | | | - Carlijn Voermans
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Ilse Timmerman
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| |
Collapse
|
45
|
Bertholf RL. Disorders of the adrenal gland. HANDBOOK OF DIAGNOSTIC ENDOCRINOLOGY 2021:103-156. [DOI: 10.1016/b978-0-12-818277-2.00004-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
46
|
Arlt B, Zasada C, Baum K, Wuenschel J, Mastrobuoni G, Lodrini M, Astrahantseff K, Winkler A, Schulte JH, Finkler S, Forbes M, Hundsdoerfer P, Guergen D, Hoffmann J, Wolf J, Eggert A, Kempa S, Deubzer HE. Inhibiting phosphoglycerate dehydrogenase counteracts chemotherapeutic efficacy against MYCN-amplified neuroblastoma. Int J Cancer 2020; 148:1219-1232. [PMID: 33284994 DOI: 10.1002/ijc.33423] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 11/03/2020] [Accepted: 11/11/2020] [Indexed: 01/12/2023]
Abstract
Here we sought metabolic alterations specifically associated with MYCN amplification as nodes to indirectly target the MYCN oncogene. Liquid chromatography-mass spectrometry-based proteomics identified seven proteins consistently correlated with MYCN in proteomes from 49 neuroblastoma biopsies and 13 cell lines. Among these was phosphoglycerate dehydrogenase (PHGDH), the rate-limiting enzyme in de novo serine synthesis. MYCN associated with two regions in the PHGDH promoter, supporting transcriptional PHGDH regulation by MYCN. Pulsed stable isotope-resolved metabolomics utilizing 13 C-glucose labeling demonstrated higher de novo serine synthesis in MYCN-amplified cells compared to cells with diploid MYCN. An independence of MYCN-amplified cells from exogenous serine and glycine was demonstrated by serine and glycine starvation, which attenuated nucleotide pools and proliferation only in cells with diploid MYCN but did not diminish these endpoints in MYCN-amplified cells. Proliferation was attenuated in MYCN-amplified cells by CRISPR/Cas9-mediated PHGDH knockout or treatment with PHGDH small molecule inhibitors without affecting cell viability. PHGDH inhibitors administered as single-agent therapy to NOG mice harboring patient-derived MYCN-amplified neuroblastoma xenografts slowed tumor growth. However, combining a PHGDH inhibitor with the standard-of-care chemotherapy drug, cisplatin, revealed antagonism of chemotherapy efficacy in vivo. Emergence of chemotherapy resistance was confirmed in the genetic PHGDH knockout model in vitro. Altogether, PHGDH knockout or inhibition by small molecules consistently slows proliferation, but stops short of killing the cells, which then establish resistance to classical chemotherapy. Although PHGDH inhibition with small molecules has produced encouraging results in other preclinical cancer models, this approach has limited attractiveness for patients with neuroblastoma.
Collapse
Affiliation(s)
- Birte Arlt
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.,Neuroblastoma Research Group, Experimental and Clinical Research Center (ECRC) of the Charité and the Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Lindenberger Weg 80, 13125, Berlin, Germany.,Berliner Institut für Gesundheitsforschung (BIH), Anna-Louisa-Karsch-Straβe 2, 10178, Berlin, Germany.,Integrative Proteomics and Metabolomics, Berlin Institute for Medical Systems Biology at the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Hannoversche Str. 28, 10115, Berlin, Germany
| | - Christin Zasada
- Integrative Proteomics and Metabolomics, Berlin Institute for Medical Systems Biology at the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Hannoversche Str. 28, 10115, Berlin, Germany
| | - Katharina Baum
- Mathematical Modelling of Cellular Processes, Max-Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Robert-Rössle-Straβe 10, 13125, Berlin, Germany
| | - Jasmin Wuenschel
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.,Neuroblastoma Research Group, Experimental and Clinical Research Center (ECRC) of the Charité and the Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Lindenberger Weg 80, 13125, Berlin, Germany
| | - Guido Mastrobuoni
- Integrative Proteomics and Metabolomics, Berlin Institute for Medical Systems Biology at the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Hannoversche Str. 28, 10115, Berlin, Germany
| | - Marco Lodrini
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.,Neuroblastoma Research Group, Experimental and Clinical Research Center (ECRC) of the Charité and the Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Lindenberger Weg 80, 13125, Berlin, Germany
| | - Kathy Astrahantseff
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Annika Winkler
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Johannes H Schulte
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.,Berliner Institut für Gesundheitsforschung (BIH), Anna-Louisa-Karsch-Straβe 2, 10178, Berlin, Germany.,German Cancer Consortium (DKTK), Partner Site Berlin, Berlin, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sabine Finkler
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.,Neuroblastoma Research Group, Experimental and Clinical Research Center (ECRC) of the Charité and the Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Lindenberger Weg 80, 13125, Berlin, Germany
| | - Martin Forbes
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.,Neuroblastoma Research Group, Experimental and Clinical Research Center (ECRC) of the Charité and the Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Lindenberger Weg 80, 13125, Berlin, Germany.,Integrative Proteomics and Metabolomics, Berlin Institute for Medical Systems Biology at the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Hannoversche Str. 28, 10115, Berlin, Germany
| | - Patrick Hundsdoerfer
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.,Department of Pediatric Oncology, Helios Klinikum Berlin Buch, Schwanebecker Chaussee 50, 13125, Berlin, Germany
| | - Dennis Guergen
- Experimental Pharmacology and Oncology Berlin-Buch GmbH (EPO), Robert-Rössle-Straβe 10, 13125, Berlin, Germany
| | - Jens Hoffmann
- Experimental Pharmacology and Oncology Berlin-Buch GmbH (EPO), Robert-Rössle-Straβe 10, 13125, Berlin, Germany
| | - Jana Wolf
- Mathematical Modelling of Cellular Processes, Max-Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Robert-Rössle-Straβe 10, 13125, Berlin, Germany
| | - Angelika Eggert
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.,Berliner Institut für Gesundheitsforschung (BIH), Anna-Louisa-Karsch-Straβe 2, 10178, Berlin, Germany.,German Cancer Consortium (DKTK), Partner Site Berlin, Berlin, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan Kempa
- Integrative Proteomics and Metabolomics, Berlin Institute for Medical Systems Biology at the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Hannoversche Str. 28, 10115, Berlin, Germany
| | - Hedwig E Deubzer
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.,Neuroblastoma Research Group, Experimental and Clinical Research Center (ECRC) of the Charité and the Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Lindenberger Weg 80, 13125, Berlin, Germany.,Berliner Institut für Gesundheitsforschung (BIH), Anna-Louisa-Karsch-Straβe 2, 10178, Berlin, Germany.,German Cancer Consortium (DKTK), Partner Site Berlin, Berlin, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
47
|
Turnock S, Turton DR, Martins CD, Chesler L, Wilson TC, Gouverneur V, Smith G, Kramer-Marek G. 18F-meta-fluorobenzylguanidine ( 18F-mFBG) to monitor changes in norepinephrine transporter expression in response to therapeutic intervention in neuroblastoma models. Sci Rep 2020; 10:20918. [PMID: 33262374 PMCID: PMC7708446 DOI: 10.1038/s41598-020-77788-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 11/13/2020] [Indexed: 02/07/2023] Open
Abstract
Targeted radiotherapy with 131I-mIBG, a substrate of the human norepinephrine transporter (NET-1), shows promising responses in heavily pre-treated neuroblastoma (NB) patients. Combinatorial approaches that enhance 131I-mIBG tumour uptake are of substantial clinical interest but biomarkers of response are needed. Here, we investigate the potential of 18F-mFBG, a positron emission tomography (PET) analogue of the 123I-mIBG radiotracer, to quantify NET-1 expression levels in mouse models of NB following treatment with AZD2014, a dual mTOR inhibitor. The response to AZD2014 treatment was evaluated in MYCN amplified NB cell lines (Kelly and SK-N-BE(2)C) by Western blot (WB) and immunohistochemistry. PET quantification of 18F-mFBG uptake post-treatment in vivo was performed, and data correlated with NET-1 protein levels measured ex vivo. Following 72 h AZD2014 treatment, in vitro WB analysis indicated decreased mTOR signalling and enhanced NET-1 expression in both cell lines, and 18F-mFBG revealed a concentration-dependent increase in NET-1 function. AZD2014 treatment failed however to inhibit mTOR signalling in vivo and did not significantly modulate intratumoural NET-1 activity. Image analysis of 18F-mFBG PET data showed correlation to tumour NET-1 protein expression, while further studies are needed to elucidate whether NET-1 upregulation induced by blocking mTOR might be a useful adjunct to 131I-mIBG therapy.
Collapse
Affiliation(s)
- Stephen Turnock
- Preclinical Molecular Imaging, Division of Radiotherapy and Imaging, The Institute of Cancer Research, 123 Old Brompton Road, London, SW7 3RP, UK
| | - David R Turton
- PET Radiochemistry, Division of Radiotherapy and Imaging, The Institute of Cancer Research, 123 Old Brompton Road, London, SW7 3RP, UK
| | - Carlos Daniel Martins
- Preclinical Molecular Imaging, Division of Radiotherapy and Imaging, The Institute of Cancer Research, 123 Old Brompton Road, London, SW7 3RP, UK
| | - Louis Chesler
- Division of Clinical Studies, The Institute of Cancer Research, 123 Old Brompton Road, London, SW7 3RP, UK
| | - Thomas C Wilson
- Department of Chemistry, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, UK
| | - Véronique Gouverneur
- Department of Chemistry, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, UK
| | - Graham Smith
- PET Radiochemistry, Division of Radiotherapy and Imaging, The Institute of Cancer Research, 123 Old Brompton Road, London, SW7 3RP, UK
| | - Gabriela Kramer-Marek
- Preclinical Molecular Imaging, Division of Radiotherapy and Imaging, The Institute of Cancer Research, 123 Old Brompton Road, London, SW7 3RP, UK.
| |
Collapse
|
48
|
Zhai K, Brockmüller A, Kubatka P, Shakibaei M, Büsselberg D. Curcumin's Beneficial Effects on Neuroblastoma: Mechanisms, Challenges, and Potential Solutions. Biomolecules 2020; 10:biom10111469. [PMID: 33105719 PMCID: PMC7690450 DOI: 10.3390/biom10111469] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/17/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023] Open
Abstract
Curcumin, a natural polyphenolic compound derived from the South Asian turmeric plant (Curcuma longa), has well-characterized antioxidant, anti-inflammatory, anti-protein-aggregate, and anticancer properties. Neuroblastoma (NB) is a cancer of the nervous system that arises primarily in pediatric patients. In order to reduce the multiple disadvantages and side effects of conventional oncologic modalities and to potentially overcome cancer drug resistance, natural substances such as curcumin are examined as complementary and supportive therapies against NB. In NB cell lines, curcumin by itself promotes apoptosis and cell cycle arrest through the suppression of serine–threonine kinase Akt and nuclear factor kappa of activated B-cells (NF-κB) signaling, induction of mitochondrial dysfunction, and upregulation of p53 and caspase signaling. While curcumin demonstrates anti-NB efficacy in vitro, cross-validation between NB cell types is currently lacking for many of its specific mechanistic activities. Furthermore, curcumin’s low bioavailability by oral administration, poor absorption, and relative insolubility in water pose challenges to its clinical introduction. Numerous curcumin formulations, including nanoparticles, nanocarriers, and microemulsions, have been developed, with these having some success in the treatment of NB. In the future, standardization and further basic and preclinical trials will be required to ensure the safety of curcumin formulations. While the administration of curcumin is clinically safe even at high doses, clinical trials are necessary to substantiate the practical efficacy of curcumin in the prevention and treatment of NB.
Collapse
Affiliation(s)
- Kevin Zhai
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| | - Aranka Brockmüller
- Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, 80336 Munich, Germany; (A.B.); (M.S.)
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Mehdi Shakibaei
- Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, 80336 Munich, Germany; (A.B.); (M.S.)
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
- Correspondence: ; Tel.: +974-4492-8334
| |
Collapse
|
49
|
Gunda V, Pathania AS, Chava S, Prathipati P, Chaturvedi NK, Coulter DW, Pandey MK, Durden DL, Challagundla KB. Amino Acids Regulate Cisplatin Insensitivity in Neuroblastoma. Cancers (Basel) 2020; 12:cancers12092576. [PMID: 32927667 PMCID: PMC7563727 DOI: 10.3390/cancers12092576] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/08/2020] [Accepted: 09/08/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Neuroblastomas mostly show poor response to the Cisplatin therapy. Amino acids serve as building blocks for proteins, which are acquired either through diet or protein breakdown. Our study reveals high amino acid pools and dependence of Cisplatin-tolerant neuroblastomas cells on amino acids for their survival, especially, in drug treated conditions. Our study also demonstrates that response of neuroblastomas to Cisplatin can be improved by decreasing cellular amino acid levels either by reducing amino acid supplements or by applying autophagy inhibitor, Hydroxychloroquine. Thus, our findings establish that neuroblastomas can be sensitized to Cisplatin by targeting amino acid metabolism. Abstract Neuroblastoma are pediatric, extracranial malignancies showing alarming survival prognosis outcomes due to their resilience to current aggressive treatment regimens, including chemotherapies with cisplatin (CDDP) provided in the first line of therapy regimens. Metabolic deregulation supports tumor cell survival in drug-treated conditions. However, metabolic pathways underlying cisplatin-resistance are least studied in neuroblastoma. Our metabolomics analysis revealed that cisplatin-insensitive cells alter their metabolism; especially, the metabolism of amino acids was upregulated in cisplatin-insensitive cells compared to the cisplatin-sensitive neuroblastoma cell line. A significant increase in amino acid levels in cisplatin-insensitive cells led us to hypothesize that the mechanisms upregulating intracellular amino acid pools facilitate insensitivity in neuroblastoma. We hereby report that amino acid depletion reduces cell survival and cisplatin-insensitivity in neuroblastoma cells. Since cells regulate their amino acids levels through processes, such as autophagy, we evaluated the effects of hydroxychloroquine (HCQ), a terminal autophagy inhibitor, on the survival and amino acid metabolism of cisplatin-insensitive neuroblastoma cells. Our results demonstrate that combining HCQ with CDDP abrogated the amino acid metabolism in cisplatin-insensitive cells and sensitized neuroblastoma cells to sub-lethal doses of cisplatin. Our results suggest that targeting of amino acid replenishing mechanisms could be considered as a potential approach in developing combination therapies for treating neuroblastomas.
Collapse
Affiliation(s)
- Venugopal Gunda
- Department of Biochemistry and Molecular Biology & The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (V.G.); (A.S.P.); (S.C.)
| | - Anup S. Pathania
- Department of Biochemistry and Molecular Biology & The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (V.G.); (A.S.P.); (S.C.)
| | - Srinivas Chava
- Department of Biochemistry and Molecular Biology & The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (V.G.); (A.S.P.); (S.C.)
| | - Philip Prathipati
- Laboratory of Bioinformatics, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki City, Osaka 567-0085, Japan;
| | - Nagendra K. Chaturvedi
- Department of Pediatrics, Division of Hematology/Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (N.K.C.); (D.W.C.)
| | - Don W. Coulter
- Department of Pediatrics, Division of Hematology/Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (N.K.C.); (D.W.C.)
| | - Manoj K. Pandey
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, 401 South Broadway, Camden, NJ 08103, USA;
| | - Donald L. Durden
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Moores Cancer Center, University of California, San Diego, 3855 Health Science Drive, MC-0815, La Jolla, CA 92093, USA;
- SignalRx Pharmaceuticals, Inc. 8330, Loveland Drive, Omaha, NE 68124, USA
| | - Kishore B. Challagundla
- Department of Biochemistry and Molecular Biology & The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (V.G.); (A.S.P.); (S.C.)
- The Children’s Health Research Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Correspondence: ; Tel.: +1-402-559-9032
| |
Collapse
|
50
|
Su Y, Wang L, Zhao Q, Yue Z, Zhao W, Wang X, Duan C, Jin M, Zhang D, Chen S, Yin J, Qiu L, Cheng X, Xu Z, Ma X. Implementation of the plasma MYCN/NAGK ratio to detect MYCN amplification in patients with neuroblastoma. Mol Oncol 2020; 14:2884-2893. [PMID: 32896084 PMCID: PMC7607162 DOI: 10.1002/1878-0261.12794] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 07/28/2020] [Accepted: 09/01/2020] [Indexed: 01/26/2023] Open
Abstract
Detection of amplification of the MYCN gene is essential for determining optimal treatment and estimating prognosis of patients with neuroblastoma (NB). DNA FISH with neuroblastoma tissues or patient‐derived bone marrow cells is the standard clinical practice for the detection of MYCN amplification. As tumor cells may often be unavailable, we developed a method to detect MYCN amplification in the plasma of patients with neuroblastoma. Taking single‐copy NAGK DNA as reference, we used real‐time quantitative PCR (qPCR) to determine the MYCN/NAGK ratio in the plasma of 115 patients diagnosed with NB. An increased MYCN/NAGK ratio in the plasma was consistent with MYCN amplification as assessed by DNA FISH. The AUC for a MYCN/NAGK ratio equal to 6.965 was 0.943, with 86% sensitivity and 100% specificity. Beyond the threshold of 6.965, the MYCN/NAGK ratio correlated with a heavier tumor burden. Event‐free and overall survival of two years were significantly shortened in stage 4 patients with a MYCN/NAGK ratio higher than 6.965. Plasma MYCN/NAGK ratios increased in patients with progressive disease and relapse. Thus, we conclude that the determination of the plasma MYCN/NAGK ratio by qPCR is a noninvasive and reproducible method to measure MYCN amplification in patients with NB.
Collapse
Affiliation(s)
- Yan Su
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Discipline of Pediatrics, Ministry of Education, MOE Key Laboratory of Major Diseases in Children, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Lijun Wang
- Beijing Keyin Technology Company Limited, Beijing Keyin Evergreen Institutes for Medical Research Company Limited, China
| | - Qian Zhao
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Discipline of Pediatrics, Ministry of Education, MOE Key Laboratory of Major Diseases in Children, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Zhixia Yue
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Discipline of Pediatrics, Ministry of Education, MOE Key Laboratory of Major Diseases in Children, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Wen Zhao
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Discipline of Pediatrics, Ministry of Education, MOE Key Laboratory of Major Diseases in Children, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Xisi Wang
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Discipline of Pediatrics, Ministry of Education, MOE Key Laboratory of Major Diseases in Children, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Chao Duan
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Discipline of Pediatrics, Ministry of Education, MOE Key Laboratory of Major Diseases in Children, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Mei Jin
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Discipline of Pediatrics, Ministry of Education, MOE Key Laboratory of Major Diseases in Children, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Dawei Zhang
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Discipline of Pediatrics, Ministry of Education, MOE Key Laboratory of Major Diseases in Children, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Shenglan Chen
- Taizhou Genewill Medical Laboratory Company Limited, China
| | - Jianfeng Yin
- Taizhou Genewill Medical Laboratory Company Limited, China
| | - Lihua Qiu
- Beijing Keyin Technology Company Limited, Beijing Keyin Evergreen Institutes for Medical Research Company Limited, China
| | - Xianfeng Cheng
- Beijing Keyin Technology Company Limited, Beijing Keyin Evergreen Institutes for Medical Research Company Limited, China
| | - Zhong Xu
- Beijing Keyin Technology Company Limited, Beijing Keyin Evergreen Institutes for Medical Research Company Limited, China
| | - Xiaoli Ma
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Discipline of Pediatrics, Ministry of Education, MOE Key Laboratory of Major Diseases in Children, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| |
Collapse
|