1
|
Dos Santos MN, Soares FDS, Felix RCM, Pujatti PB. Retrospective analysis of neuroblastoma and pheochromocytoma therapy with I-131 metaiodobenzylguanidine at a reference oncology hospital in Brazil. Nucl Med Commun 2025:00006231-990000000-00429. [PMID: 40420793 DOI: 10.1097/mnm.0000000000001993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
BACKGROUND Metaiodobenzylguanidine (MIBG) is a norepinephrine analogue with high affinity and specificity for the norepinephrine transporter. I-131-labeled MIBG (I-131 MIBG) is a therapeutic radiopharmaceutical used in selected cases of refractory or metastatic neuroblastoma and pheochromocytoma, tumors that overexpress the norepinephrine transporter. While the use of I-131 MIBG in neuroblastoma and pheochromocytoma therapy is well-established, the literature shows significant variability in treatment response, regarding the dose of the radiopharmaceutical, previous therapies administered, and the clinical condition of the patients involved in the studies. To contribute to the current literature, this study analyzed the use of I-131 MIBG at a cancer treatment institution in Brazil. METHODS Retrospective, observational, single-center study was conducted, with a descriptive and exploratory character, involving patients diagnosed with neuroblastoma and pheochromocytoma treated with I-131 MIBG from 2010 to 2025. Demographic, clinical, and laboratory parameters were collected before and after I-131 MIBG therapy. The outcome was determined through survival analysis. RESULTS Thirty-two patients were treated with I-131 MIBG, including 24 patients with neuroblastoma and six patients with pheochromocytoma. Leukocytes and platelets showed a reduction, and aspartate aminotransferase (AST) levels exhibited a significant increase posttherapy in patients with neuroblastoma. Survival rate was 84% in patients with pheochromocytoma and 55% in patients with neuroblastoma in the first year following I-131 MIBG therapy; however, both groups showed a gradual reduction in the cumulative survival rates, reaching 20% after 5 years. CONCLUSION I-131 MIBG was primarily used for the treatment of neuroblastoma in pediatric patients, and pheochromocytomas in adults. Anemia, leukopenia, thrombocytopenia, and increased serum AST were the main adverse events, and cumulative survival rates were 20% after 5 years.
Collapse
|
2
|
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
|
3
|
Huang K, Yang L, Ma Y, Cao L, Li S, Zhao Z, Zhou J, Wang S. Integrated multi-omics characterization of neuroblastoma with bone or bone marrow metastasis. Genes Dis 2025; 12:101511. [PMID: 40070366 PMCID: PMC11894310 DOI: 10.1016/j.gendis.2024.101511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 11/17/2024] [Accepted: 12/18/2024] [Indexed: 03/14/2025] Open
Abstract
The pathogenesis of neuroblastoma with bone or bone marrow metastasis (NB-BBM) and its complex immune microenvironment remain poorly elucidated, hampering the advancement of effective risk prediction for BBM and limiting therapeutic strategies. Feature recognition of 142 paraffin-embedded hematoxylin-eosin-stained tumor section images was conducted using a Swin-Transformer for pathological histology to predict NB-BBM occurrence. Single-cell transcriptomics identified a tumor cell subpopulation (NB3) and two tumor-associated macrophage (TAM) subpopulations (SPP1+ TAMs and IGHM+ TAMs) closely associated with BBM and highlighted transketolase (TKT) as a key molecular marker for metastatic progression in NB. This extensive multi-omics investigation into NB-BBM enhances our understanding of single-cell transcriptional dynamics in NB beyond existing research, outlining the evolution from in situ carcinoma through tumorigenesis to bone marrow metastases. Furthermore, exploration of the immune microenvironment identified specific subpopulations of TAMs crucial in promoting NB-BBM, presenting new avenues for immunotherapy. These insights enhance our understanding of the metastatic process from NB to BBM and facilitate the development of more effective diagnostic and therapeutic strategies for this aggressive pediatric cancer.
Collapse
Affiliation(s)
| | | | - Yue Ma
- Department of Pediatric Surgical Oncology, The 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 Pediatric Metabolism and Inflammatory Diseases, Chongqing 400014, China
| | - Lijian Cao
- Department of Pediatric Surgical Oncology, The 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 Pediatric Metabolism and Inflammatory Diseases, Chongqing 400014, China
| | - Suwen Li
- Department of Pediatric Surgical Oncology, The 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 Pediatric Metabolism and Inflammatory Diseases, Chongqing 400014, China
| | - Zhenzhen Zhao
- Department of Pediatric Surgical Oncology, The 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 Pediatric Metabolism and Inflammatory Diseases, Chongqing 400014, China
| | - Jianwu Zhou
- Department of Pediatric Surgical Oncology, The 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 Pediatric Metabolism and Inflammatory Diseases, Chongqing 400014, China
| | - Shan Wang
- Department of Pediatric Surgical Oncology, The 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 Pediatric Metabolism and Inflammatory Diseases, Chongqing 400014, China
| |
Collapse
|
4
|
Chen Y, Zhu H, Luo Y, Xie T, Hu Y, Yan Z, Ji W, Wang Y, Yin Q, Xian H. ALDOC promotes neuroblastoma progression and modulates sensitivity to chemotherapy drugs by enhancing aerobic glycolysis. Front Immunol 2025; 16:1573815. [PMID: 40313939 PMCID: PMC12043483 DOI: 10.3389/fimmu.2025.1573815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 03/17/2025] [Indexed: 05/03/2025] Open
Abstract
Introduction Neuroblastoma (NB), a malignant extracranial solid tumor originating from the sympathetic nervous system, exhibits poor prognosis in high-risk cases, with a 5-year overall survival rate below 50%. Glycolysis has been implicated in NB pathogenesis, and targeting glycolysis-related pathways shows therapeutic potential. This study investigates the role of the glycolysis-associated gene ALDOC in NB pathogenesis and its impact on chemotherapy sensitivity. Methods Transcriptomic data from NB patients were analyzed to identify ALDOC as an independent risk factor for high-risk NB. Protein expression levels of ALDOC were assessed in NB cells versus normal cells using immunoblotting. Functional experiments, including proliferation and migration assays, were conducted in ALDOC-interfered NB cell lines. Glycolytic activity was evaluated by measuring glucose uptake, lactate production, and ATP generation. Additionally, the sensitivity of ALDOC-downregulated NB cells to cisplatin and cyclophosphamide was tested to explore its role in chemotherapy response. Results ALDOC was identified as a high-risk prognostic marker in NB, with elevated protein expression in NB cells compared to normal controls. Silencing ALDOC significantly inhibited NB cell proliferation and migration. Glycolytic activity was markedly reduced in ALDOC-downregulated cells, evidenced by decreased glucose uptake, lactate production, and ATP levels. Furthermore, ALDOC suppression enhanced NB cell sensitivity to cisplatin and cyclophosphamide, suggesting a glycolysis-dependent mechanism underlying chemotherapy resistance. Discussion Our findings highlight ALDOC as a critical driver of NB progression through glycolysis acceleration, with implications for therapeutic targeting. The observed increase in chemotherapy sensitivity upon ALDOC inhibition underscores its potential as a biomarker for treatment optimization. However, the complexity of glycolysis regulation, involving multiple genes and pathways, necessitates further mechanistic studies to clarify ALDOC's specific role. Despite this limitation, our work emphasizes the importance of aerobic glycolysis in NB pathogenesis and provides a foundation for developing novel therapeutic strategies targeting ALDOC or associated pathways. Future research should explore interactions between ALDOC and other glycolytic regulators to refine combinatorial treatment approaches.
Collapse
Affiliation(s)
- Yunpeng Chen
- School of Medicine, Nantong University, Nantong, China
| | - Haixia Zhu
- Cancer Research Center Nantong, Nantong Tumor Hospital, Nantong, Jiangsu, China
| | - Yishu Luo
- School of Medicine, Nantong University, Nantong, China
| | - Tianyue Xie
- Department of Endocrinology, Affiliated Hospital of Nantong University, Nantong, China
| | - Youyang Hu
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Zhiwei Yan
- School of Medicine, Nantong University, Nantong, China
| | - Weichao Ji
- School of Medicine, Nantong University, Nantong, China
| | - YaXuan Wang
- Department of Urology, Nantong Tumor Hospital, Nantong, China
| | - Qiyou Yin
- Department of Paediatric Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Hua Xian
- Department of Paediatric Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
5
|
Zhou Z, Yang X, Wang G, Wang X, Liu J, Xu Y, Ying K, Wang W, Yang J. The prognostic significance of semi-quantitative metabolic parameters and tumoral metabolic activity based on 123I-MIBG SPECT/CT in pretreatment neuroblastoma patients. Cancer Imaging 2025; 25:45. [PMID: 40165328 PMCID: PMC11956495 DOI: 10.1186/s40644-025-00858-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 03/10/2025] [Indexed: 04/02/2025] Open
Abstract
PURPOSE To assess the prognosis predictive value of semi-quantitative metabolic parameters and tumoral metabolic activity based on 123I-meta-iodobenzylguanidine (MIBG) SPECT/CT in pretreatment neuroblastoma (NB) patients. METHODS A total of 50 children (25 girls, 25 boys, median age 37 months, range 1-102 months) with newly diagnosed NB, consecutively examined with pretherapeutic 123I-MIBG SPECT/CT between 2018 and 2024, were included in this retrospective study. The semi-quantitative metabolic parameters and activity of primary tumor were measured, including Tmax/Lmax, Tmean/Lmean, Tmax/Lmean, Tmax/Mmax, Tmean/Mmean and asphericity (ASP). The ratio was maximum or mean count of primary tumor, liver and muscle. Clinical data and image-related factors was recorded as well. The outcome endpoint was event-free survival (EFS). Independent predictors were identified through univariate and multivariate logistic regression analyses. Receiver operating characteristic (ROC) and Kaplan Meier analysis with log-rank test for EFS were performed. RESULTS Median follow-up was 42 months (range 2.5-74 months; 4 patients showed disease progression/relapse, 7 patients died). The univariate and multivariate Cox regression analysis demonstrated that bone/bone marrow metastasis [95% confidence interval (CI): 1.051, 18.570, p = 0.043], Tmax/Lmax (95% CI: 1.074, 1.459, p = 0.004) and ASP (95% CI: 2.618, 273.477, p = 0.006) were independent predictors of EFS. The Kaplan Meier survival analyses demonstrated that Tmax/Lmax undefined[Formula: see text]]]>6 and ASP [Formula: see text]undefined]]>34% and with bone/bone marrow metastasis had worse outcomes. CONCLUSION In this exploratory study, pretherapeutic 123I-MIBG image-derived semi-quantitative metabolic parameters and tumor asphericity provided prognostic value for EFS in NB patients. Tmax/Lmax [Formula: see text]undefined]]>6 and ASP [Formula: see text]undefined]]>34%, along with the presence of bone/bone marrow metastasis, could be considered as supplementary factors alongside existing ones.
Collapse
Affiliation(s)
- Ziang Zhou
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, 100050, China
| | - Xu Yang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, 100050, China
| | - Guanyun Wang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, 100050, China
| | - Xiaoya Wang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, 100050, China
| | - Jun Liu
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, 100050, China
| | - Yanfeng Xu
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, 100050, China
| | - Kan Ying
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, 100050, China
| | - Wei Wang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, 100050, China
| | - Jigang Yang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, 100050, China.
| |
Collapse
|
6
|
Wang X, Mao AW, Pan S, Wang D, He L, Vogel H, Mao JH, Weiss W, Li T, Chang H. Cellular morphometric biomarkers and large language model predict prognosis and treatment response in neuroblastoma patients: A retrospective and double-blind prospective single arm clinical study. Eur J Cancer 2025; 218:115273. [PMID: 39908653 DOI: 10.1016/j.ejca.2025.115273] [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: 05/25/2024] [Revised: 01/07/2025] [Accepted: 01/29/2025] [Indexed: 02/07/2025]
Abstract
BACKGROUND The heterogeneity of Neuroblastoma (NB) leads to variation in response to treatment and outcomes. The aim of the current study is to discover AI-empowered cellular morphometric biomarkers (CMBs), to establish the corresponding CMB risk score (CMBRS), CMB risk group (CMBRG), large language model driven CMB risk score (CMB-LLM-RS), and large language model driven CMB risk group (CMB-LLM-RG), and to investigate and validate their prognostic and predictive power in NB. METHODS In this study, the retrospective cohort enrolled 84 primary NBs between 1/2020 and 12/2021, followed up through 11/22/2024; the prospective cohort enrolled 67 primary NBs between 1/2022 and 7/2023, followed up through 11/22/2024. RESULTS We identified 9 CMBs from a retrospective NB cohort, enabling the CMBRS, CMBRG, CMB-LLM-RS, and CMB-LLM-RG. Both CMBRG and CMB-LLM-RG are significantly associated with prognosis (p < 0.0001) and treatment response (p < 0.0001). Furthermore, we double-blindly validated the predictive power of CMBRG and CMB-LLM-RG in a prospective NB cohort, which confirms their potential value in real clinical settings. Importantly, CMBRG provides clinical value independent of the International Neuroblastoma Risk Group (INRG) classification system in both retrospective and prospective NB cohorts (p < 0.05); and the combination of CMBRG and INRG significantly increases prognostic and predictive performance for NB patients. CONCLUSIONS These findings suggest that CMBRG and CMB-LLM-RG have prognostic and predictive value for NB and warrants evaluation in larger multicenter cohorts.
Collapse
Affiliation(s)
- Xu Wang
- Department of Endocrinology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - April W Mao
- Department of Mathematics, University of California, Los Angeles, Los Angeles, United States
| | - Sirui Pan
- Department of Oncology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Dawei Wang
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States; Berkeley Biomedical Data Science Center, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Lili He
- Department of Pathology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Hannes Vogel
- Department of Pathology, Stanford University Medical Center, Stanford, CA, United States
| | - Jian-Hua Mao
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States; Berkeley Biomedical Data Science Center, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - William Weiss
- Department of Neurology, Neurological Surgery, and Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| | - Tao Li
- Department of Oncology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Hang Chang
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States; Berkeley Biomedical Data Science Center, Lawrence Berkeley National Laboratory, Berkeley, CA, United States.
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
Doyle K, Hassan AE, Sutter M, Rodriguez M, Kumar P, Brown E. A Comparison of in vivo Tumor-Homing Abilities of Placental-Derived and Bone Marrow-Derived Mesenchymal Stromal Cells in High-Risk Neuroblastoma. J Pediatr Surg 2025; 60:161954. [PMID: 39379183 PMCID: PMC12068278 DOI: 10.1016/j.jpedsurg.2024.161954] [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: 09/09/2024] [Accepted: 09/17/2024] [Indexed: 10/10/2024]
Abstract
BACKGROUND Neuroblastoma is a highly lethal malignancy of young children. Mesenchymal stromal cells (MSCs) may represent a novel cellular delivery vehicle due to their innate tumor-homing properties. We compared in vivo homing abilities of placental-derived MSCs (PMSCs) and bone marrow-derived MSCs (BM-MSCs) in an orthotopic neuroblastoma xenograft. METHODS 28 mice underwent direct implantation of neuroblastoma cells (cell line NB1643) into the adrenal gland followed by intraperitoneal injection of 5 × 106 MSCs (PMSC n = 13, BM-MSC n = 13, PBS controls n = 2). MSC migration was monitored with in vivo imaging system (IVIS) radiance measurements at multiple timepoints post-MSC injection. Necropsy timepoints were 72 h (n = 10) and 7 days (n = 16). Ex vivo imaging was performed on all adrenal masses and select organ tissues. Immunohistochemistry (IHC) assessed the presence of MSCs in tumors. RESULTS IVIS demonstrated initial diffuse signal that migrated to the left abdomen. Radiance decreased over time, but MSC signal persisted at day 7 in all animals. Ex vivo IVIS demonstrated signal in the adrenal tumor but not other organs. There was no significant difference in average ex vivo adrenal mass radiance between MSC groups (p = 0.74). IHC confirmed presence of both MSC types within the tumor. CONCLUSION PMSCs and BM-MSCs successfully migrated to neuroblastoma tumor tissues in vivo without evidence of migration to other organs. MSCs migrate within 72 h and persisted within the tumor up to 7 days. There was no significant difference in homing capabilities of PMSCs compared to BM-MSCs, indicating that either cell type has potential as a drug delivery vehicle. TYPE OF STUDY Original Research. LEVEL OF EVIDENCE n/a.
Collapse
Affiliation(s)
- Kathleen Doyle
- Department of Surgery, University of California-Davis, Sacramento, CA, USA.
| | | | - Maria Sutter
- Surgical Bioengineering Laboratory, Department of Surgery, University of California-Davis, Sacramento, CA, USA
| | - Monica Rodriguez
- Surgical Bioengineering Laboratory, Department of Surgery, University of California-Davis, Sacramento, CA, USA
| | - Priyadarsini Kumar
- Surgical Bioengineering Laboratory, Department of Surgery, University of California-Davis, Sacramento, CA, USA
| | - Erin Brown
- Department of Surgery, Division of Pediatric Surgery, University of California-Davis, Sacramento, CA, USA
| |
Collapse
|
10
|
Purkayastha S, Shalu H, Gutman D, Holodny A, Modak S, Basu E, Kushner B, Kramer K, Haque S, Stember JN. Evolutionary Strategies AI Addresses Multiple Technical Challenges in Deep Learning Deployment: Proof-of-Principle Demonstration for Neuroblastoma Brain Metastasis Detection. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024; 37:2920-2930. [PMID: 38886289 PMCID: PMC11612045 DOI: 10.1007/s10278-024-01165-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/09/2024] [Accepted: 06/03/2024] [Indexed: 06/20/2024]
Abstract
Two significant obstacles hinder the advancement of Radiology AI. The first is the challenge of overfitting, where small training data sets can result in unreliable outcomes. The second challenge is the need for more generalizability, the lack of which creates difficulties in implementing the technology across various institutions and practices. A recent innovation, deep neuroevolution (DNE), has been introduced to tackle the overfitting issue by training on small data sets and producing accurate predictions. However, the generalizability of DNE has yet to be proven. This paper strives to overcome this barrier by demonstrating that DNE can achieve satisfactory results in diverse external validation sets. The main innovation of the work is thus showing that DNE can generalize to varied outside data. Our example use case is predicting brain metastasis from neuroblastoma, emphasizing the importance of AI with limited data sets. Despite image collection and labeling advancements, rare diseases will always constrain data availability. We optimized a convolutional neural network (CNN) with DNE to demonstrate generalizability. We trained the CNN with 60 MRI images and tested it on a separate diverse collection of images from over 50 institutions. For comparison, we also trained with the more traditional stochastic gradient descent (SGD) method, with the two variants of (1) training from scratch and (2) transfer learning. Our results show that DNE demonstrates excellent generalizability with 97% accuracy on the heterogeneous testing set, while neither form of SGD could reach 60% accuracy. DNE's ability to generalize from small training sets to external and diverse testing sets suggests that it or similar approaches may play an integral role in improving the clinical performance of AI.
Collapse
Affiliation(s)
- Subhanik Purkayastha
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Hrithwik Shalu
- Department of Aerospace Engineering, Indian Institute of Technology Madras, Chennai, India, 600036
| | - David Gutman
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Andrei Holodny
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Shakeel Modak
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Ellen Basu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Brian Kushner
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Kim Kramer
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Sofia Haque
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Joseph N Stember
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| |
Collapse
|
11
|
Finkelstein SR, Patel R, Deland K, Mercer J, Starr B, Zhu D, Min H, Reinsvold M, Campos LDS, Williams NT, Luo L, Ma Y, Neff J, Hoenerhoff MJ, Moding EJ, Kirsch DG. 56Fe-ion Exposure Increases the Incidence of Lung and Brain Tumors at a Similar Rate in Male and Female Mice. Radiat Res 2024; 202:734-744. [PMID: 39307527 DOI: 10.1667/rade-24-00004.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 08/23/2024] [Indexed: 09/25/2024]
Abstract
The main deterrent to long-term space travel is the risk of Radiation Exposure Induced Death (REID). The National Aeronautics and Space Administration (NASA) has adopted Permissible Exposure Levels (PELs) to limit the probability of REID to 3% for the risk of death due to radiation-induced carcinogenesis. The most significant contributor to current REID estimates for astronauts is the risk of lung cancer. Recently updated lung cancer estimates from Japan's atomic bomb survivors showed that the excess relative risk of lung cancer by age 70 is roughly fourfold higher in females compared to males. However, whether sex differences may impact the risk of lung cancer due to exposure to high charge and energy (HZE) radiation is not well studied. Thus, to evaluate the impact of sex differences on the risk of solid cancer development after HZE radiation exposure, we irradiated Rbfl/fl, Trp53fl/+ male and female mice infected with Adeno-Cre with various doses of 320 kVp X rays or 600 MeV/n 56Fe ions and monitored them for any radiation-induced malignancies. We conducted complete necropsy and histopathology of major organs on 183 male and 157 female mice after following them for 350 days postirradiation. We observed that lung adenomas/carcinomas and esthesioneuroblastomas (ENBs) were the most common primary malignancies in mice exposed to X rays and 56Fe ions, respectively. In addition, 1 Gy 56Fe-ion exposure compared to X-ray exposure led to a significantly increased incidence of lung adenomas/carcinomas (P = 0.02) and ENBs (P < 0.0001) in mice. However, we did not find a significantly higher incidence of any solid malignancies in female mice as compared to male mice, regardless of radiation quality. Furthermore, gene expression analysis of ENBs suggested a distinct gene expression pattern with similar hallmark pathways altered, such as MYC targets and MTORC1 signaling, in ENBs induced by X rays and 56Fe ions. Thus, our data revealed that 56Fe-ion exposure significantly accelerated the development of lung adenomas/carcinomas and ENBs compared to X rays, but the rate of solid malignancies was similar between male and female mice, regardless of radiation quality.
Collapse
Affiliation(s)
- Sophie R Finkelstein
- Department of Radiation Oncology, Duke University School of Medicine, Durham, North Carolina
| | - Rutulkumar Patel
- Department of Radiation Oncology, Baylor College of Medicine, Houston, Texas
| | - Katherine Deland
- Department of Radiation Oncology, Duke University School of Medicine, Durham, North Carolina
| | - Joshua Mercer
- Department of Radiation Oncology, Duke University School of Medicine, Durham, North Carolina
| | - Bryce Starr
- Department of Radiation Oncology, Duke University School of Medicine, Durham, North Carolina
| | - Daniel Zhu
- Department of Radiation Oncology, Duke University School of Medicine, Durham, North Carolina
| | - Hooney Min
- Department of Radiation Oncology, Duke University School of Medicine, Durham, North Carolina
| | - Michael Reinsvold
- Department of Radiation Oncology, Duke University School of Medicine, Durham, North Carolina
| | | | - Nerissa T Williams
- Department of Radiation Oncology, Duke University School of Medicine, Durham, North Carolina
| | - Lixia Luo
- Department of Radiation Oncology, Duke University School of Medicine, Durham, North Carolina
| | - Yan Ma
- Department of Radiation Oncology, Duke University School of Medicine, Durham, North Carolina
| | - Jadee Neff
- Department of Pathology, Duke University, Durham, North Carolina
| | - Mark J Hoenerhoff
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Everett J Moding
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - David G Kirsch
- Department of Radiation Oncology, Duke University School of Medicine, Durham, North Carolina
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, North Carolina
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario
- Department of Radiation Oncology and Department of Medical Biophysics, University of Toronto, Toronto, Ontario
| |
Collapse
|
12
|
Zhao Z, Yang C. Predictive value of 18 F-FDG PET/CT versus bone marrow biopsy and aspiration in pediatric neuroblastoma. Clin Exp Metastasis 2024; 41:627-638. [PMID: 38609536 DOI: 10.1007/s10585-024-10286-2] [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: 02/05/2024] [Accepted: 04/04/2024] [Indexed: 04/14/2024]
Abstract
BACKGROUND Neuroblastoma (NB) is the most prevalent solid extracranial malignancy in children, often with bone marrow metastases (BMM) are present. The conventional approach for detecting BMM is bone marrow biopsy and aspiration (BMBA). 18 F-fluorodeoxyglucose-positron emission tomography/computed tomography (18 F-FDG PET/CT) has become a staple for staging and is also capable of evaluating marrow infiltration. The consensus on the utility of 18 F-FDG PET/CT for assessing BMM in NB patients is still under deliberation. METHODS This retrospective study enrolled 266 pediatric patients with pathologically proven NB. All patients had pretherapy FDG PET/CT. BMBA, clinical, radiological, and follow-up data were also collected. The diagnostic accuracy of BMBA and 18 F-FDG PET/CT was assessed. RESULTS BMBAs identified BMM in 96 cases (36.1%), while 18 F-FDG PET/CT detected BMI in 106 cases (39.8%) within the cohort. The initial sensitivity, positive predictive value (PPV), specificity, and negative predictive value (NPV) of 18 F-FDG PET/CT were 93.8%, 84.9%, 90.6%, and 96.3%, respectively. After treatment, these values were 92.3%, 70.6%, 97.3%, and 99.4%, respectively. The kappa statistic, which measures agreement between BMBA and 18 F-FDG PET/CT, was 0.825 before treatment and 0.784 after treatment, with both values indicating a substantial agreement (P = 0.000). Additionally, the amplification of MYCN and a positive initial PET/CT scan were identified as independent prognostic factors for overall survival (OS). CONCLUSION 18 F-FDG-PET/CT is a valuable method for evaluating BMM in NB. The routine practice of performing a BMBA without discrimination may need to be reassessed. Negative result from 18 F-FDG-PET/CT could potentially spare children with invasive bone marrow biopsies.
Collapse
Affiliation(s)
- Zhenzhen Zhao
- Department of Surgical oncology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Chao Yang
- Department of Surgical oncology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
- , 136 Zhongshan 2nd Road, Yuzhong District, Chongqing, 400014, China.
| |
Collapse
|
13
|
Chang S, Ren D, Zhang L, Liu S, Yang W, Cheng H, Zhang X, Hong E, Geng D, Wang Y, Chen C, Zhang J, Shi T, Guo Y, Ni X, Wang H, Jin Y. Therapeutic SHPRH-146aa encoded by circ-SHPRH dynamically upregulates P21 to inhibit CDKs in neuroblastoma. Cancer Lett 2024; 598:217120. [PMID: 39002691 DOI: 10.1016/j.canlet.2024.217120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/03/2024] [Accepted: 07/10/2024] [Indexed: 07/15/2024]
Abstract
Recent research has underscored the significance of circular RNAs (circRNAs) in various cancers, including neuroblastoma (NB). Specifically, circ-SHPRH, a unique circRNA, has been revealed to inhibit tumor growth by sequestering miRNAs or producing the SHPRH-146aa protein. To explore circ-SHPRH's involvement in NB and its potential application in gene therapy, this study examined circ-SHPRH expression in 94 NB tissues and cell lines (SK-N-BE(2), SH-SY5Y) using real-time PCR and fluorescence in situ hybridization (FISH). Functional assays encompassing both overexpression and knockdown experiments in NB cell lines, as well as in vivo investigations, were conducted. RNA-seq analysis revealed a correlation between circ-SHPRH and the pathway of P21 (CDKN1A), a pivotal cell cycle regulator. Validation through PCR and other techniques confirmed that circ-SHPRH upregulated P21 expression. Furthermore, the regulatory role of circ-SHPRH in the P21-CDK pathway was corroborated through SHPRH-146aa expression analysis. Notably, adenovirus-mediated circ-SHPRH overexpression effectively curbed NB tumor growth in NSG mice, while combining circ-SHPRH with everolimus exhibited potential for NB treatment. This study elucidates the remarkable significance of circ-SHPRH in NB and its prospective utility in gene therapy, thereby paving the way for innovative therapeutic approaches.
Collapse
Affiliation(s)
- Saishuo Chang
- Department of Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Dong Ren
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Li Zhang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shan Liu
- Department of Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Wei Yang
- Department of Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Haiyan Cheng
- Department of Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Xuexi Zhang
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Enyu Hong
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Di Geng
- Biobank for Clinical Data and Samples in Pediatrics, Beijing Children's Hospital, National Center for Children's Health, Beijing Pediatric Research Institute, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Yadi Wang
- Biobank for Clinical Data and Samples in Pediatrics, Beijing Children's Hospital, National Center for Children's Health, Beijing Pediatric Research Institute, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Chenghao Chen
- Department of Thoracic Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Jie Zhang
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Tieliu Shi
- Center for Bioinformatics and Computational Biology and the Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yongli Guo
- Department of Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Xin Ni
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China; Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China.
| | - Huanmin Wang
- Department of Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China.
| | - Yaqiong Jin
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China.
| |
Collapse
|
14
|
Mao C, Poimenidou M, Craig BT. Current Knowledge and Perspectives of Immunotherapies for Neuroblastoma. Cancers (Basel) 2024; 16:2865. [PMID: 39199637 PMCID: PMC11353182 DOI: 10.3390/cancers16162865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/02/2024] [Accepted: 08/12/2024] [Indexed: 09/01/2024] Open
Abstract
Neuroblastoma (NBL) cells highly express disialoganglioside GD2, which is restricted and weakly expressed in selected healthy cells, making it a desirable target of immunotherapy. Over the past two decades, application of dinutuximab, an anti-GD2 monoclonal antibody (mAb), has been one of the few new therapies to substantially improve outcomes to current levels. Given the persistent challenge of relapse and therapeutic resistance, there is an urgent need for new effective and tolerable treatment options for high-risk NBL. Recent breakthroughs in immune checkpoint inhibitor (ICI) therapeutics have not translated into high-risk NBL, like many other major pediatric solid tumors. Given the suppressed tumor microenvironment (TME), single ICIs like anti-CTLA4 and anti-PD1 have not demonstrated significant antitumor response rates. Meanwhile, emerging studies are reporting novel advancements in GD2-based therapies, targeted therapies, nanomedicines, and other immunotherapies such as adoptive transfer of natural killer (NK) cells and chimeric antigen receptors (CARs), and these hold interesting promise for the future of high-risk NBL patient care. Herein, we summarize the current state of the art in NBL therapeutic options and highlight the unique challenges posed by NBL that have limited the successful adoption of immune-modifying therapies. Through this review, we aim to direct the field's attention to opportunities that may benefit from a combination immunotherapy strategy.
Collapse
Affiliation(s)
- Chenkai Mao
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
- Center for Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
| | - Maria Poimenidou
- Center for Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
| | - Brian T. Craig
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
- Center for Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
| |
Collapse
|
15
|
Wu TN, Hung JT, Hung TH, Wang YH, Wu JC, Yu AL. Effective suppression of tumor growth and hepatic metastasis of neuroblastoma by NKT-stimulatory phenyl glycolipid. Biomed Pharmacother 2024; 177:117040. [PMID: 38959605 DOI: 10.1016/j.biopha.2024.117040] [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: 04/05/2024] [Revised: 06/15/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024] Open
Abstract
Invariant natural killer T cell (iNKT) cells produce large amounts of cytokines in response to α-Galactosylceramide (α-GalCer) stimulation. An analog containing two phenyl rings on the acyl chain, C34, was previously found to be more Th1-biased than α-GalCer and triggered greater anticancer activities against breast cancer, melanoma and lung cancer in mice. Since liver is enriched in iNKT cells, we investigated anticancer efficacy of C34 on neuroblastoma with hepatic metastasis. C34 induced Th1-biased cytokine secretions in the liver, significantly suppressed neuroblastoma growth/metastasis and prolonged mouse survival. The anti-tumor efficacy might be attributed to greater expansions of hepatic NKT, NK, CD4+ T, and CD8+ T cells as well as reduction of the number of SSCloGr1intCD11b+ subset of myeloid-derived suppressor cells (MDSCs) in the liver of tumor-bearing mice, as compared to DMSO control group. C34 also upregulated expression of CD1d and CD11c, especially in the SSCloGr1intCD11b+ subset of MDSCs, which might be killed by C34-activated NKT cells, attributing to their reduced number. In addition, C34 also induced expansion of CD4+ T, CD8+ T, and NK cells, which might eliminate neuroblastoma cells. These immune-modulating effects of C34 might act in concert in the local milieu of liver to suppress the tumor growth. Further analysis of database of neuroblastoma revealed that patients with high CD11c expression in the monocytic MDSCs in the tumor had longer survival, suggesting the potential clinical application of C34 for treatment of neuroblastoma.
Collapse
Affiliation(s)
- Tai-Na Wu
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 115, Taiwan; Institute of Biotechnology, National Taiwan University, Taipei 115, Taiwan.
| | - Jung-Tung Hung
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan.
| | - Tsai-Hsien Hung
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan
| | - Ya-Hui Wang
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan
| | - Jen-Chine Wu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan
| | - Alice L Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan; Department of Pediatrics, University of California in San Diego, La Jolla, CA, USA
| |
Collapse
|
16
|
Mimura K, Fumino S, Yamashi K, Iguchi M, Inoue M, Takayama S, Kim K, Aoi S, Tajiri T, Ono S. Systemic inflammation enhances metastatic growth in a syngeneic neuroblastoma mouse model. Pediatr Surg Int 2024; 40:195. [PMID: 39017743 DOI: 10.1007/s00383-024-05788-9] [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] [Accepted: 07/12/2024] [Indexed: 07/18/2024]
Abstract
BACKGROUND We previously showed that total tumor resection enhances metastatic growth in a syngeneic metastatic mouse model of neuroblastoma. In this study, we further investigated which surgical factors contributed most to metastatic growth. METHODS Tumor cells derived from MYCN transgenic mice were subcutaneously injected into wild-type mice. Mice were randomly assigned to receive partial resection (PR group), subcutaneous implantation of a sponge (Sp group), or observation (Obs group). The lymph node metastasis volume and the frequency of lung metastasis were compared 14 days after assignment by measuring C-reactive protein (CRP) and interleukin-6 (IL-6) levels. RESULTS The lymph node metastasis volume in the Sp group was larger than in the Obs group (148.4 [standard deviation {SD}: 209.5] vs. 10.2 [SD 12.8] mm3). The frequency of lung metastasis was greater in the Sp group than in the PR group (11.9 [SD 12.2] vs. 6.6 [SD 4.0] counts/slide). The CRP level in the Sp group was higher than in the PR group (2.3 [SD 0.5] vs. 1.5 [SD 0.4] μg/mL), and the IL-6 level in the Sp group was higher than in the PR or Obs groups (28.4 [SD 34.5] vs. 12.4 [SD 19.0] vs. 5.4 [SD 8.1] pg/mL). CONCLUSION Metastatic growth may be enhanced by systemic inflammation.
Collapse
Affiliation(s)
- Kazuya Mimura
- Department of Pediatric Surgery, Kyoto Prefectural University of Medicine, 465 Kajii Cho, Kawaramachi Hirokoji, Kamigyo Ku, Kyoto, 602-8566, Japan.
| | - Shigehisa Fumino
- Department of Pediatric Surgery, Kyoto Prefectural University of Medicine, 465 Kajii Cho, Kawaramachi Hirokoji, Kamigyo Ku, Kyoto, 602-8566, Japan
| | - Kodai Yamashi
- Department of Pediatric Surgery, Kyoto Prefectural University of Medicine, 465 Kajii Cho, Kawaramachi Hirokoji, Kamigyo Ku, Kyoto, 602-8566, Japan
| | - Masafumi Iguchi
- Department of Pediatric Surgery, Kyoto Prefectural University of Medicine, 465 Kajii Cho, Kawaramachi Hirokoji, Kamigyo Ku, Kyoto, 602-8566, Japan
| | - Maho Inoue
- Department of Pediatric Surgery, Kyoto Prefectural University of Medicine, 465 Kajii Cho, Kawaramachi Hirokoji, Kamigyo Ku, Kyoto, 602-8566, Japan
- Department of Pediatric Surgery, SSM Health Cardinal Glennon Children's Hospital, St. Louis, USA
| | - Shohei Takayama
- Department of Pediatric Surgery, Kyoto Prefectural University of Medicine, 465 Kajii Cho, Kawaramachi Hirokoji, Kamigyo Ku, Kyoto, 602-8566, Japan
| | - Kiyokazu Kim
- Department of Pediatric Surgery, Kyoto Prefectural University of Medicine, 465 Kajii Cho, Kawaramachi Hirokoji, Kamigyo Ku, Kyoto, 602-8566, Japan
| | - Shigeyoshi Aoi
- Department of Pediatric Surgery, Kyoto Prefectural University of Medicine, 465 Kajii Cho, Kawaramachi Hirokoji, Kamigyo Ku, Kyoto, 602-8566, Japan
| | - Tatsuro Tajiri
- Department of Pediatric Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shigeru Ono
- Department of Pediatric Surgery, Kyoto Prefectural University of Medicine, 465 Kajii Cho, Kawaramachi Hirokoji, Kamigyo Ku, Kyoto, 602-8566, Japan
| |
Collapse
|
17
|
Djos A, Svensson J, Gaarder J, Umapathy G, Nilsson S, Ek T, Vogt H, Georgantzi K, Öra I, Träger C, Kogner P, Martinsson T, Fransson S. Loss of Chromosome Y in Neuroblastoma Is Associated With High-Risk Disease, 11q-Deletion, and Telomere Maintenance. Genes Chromosomes Cancer 2024; 63:e23260. [PMID: 39031441 DOI: 10.1002/gcc.23260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 07/22/2024] Open
Abstract
Neuroblastoma (NB) is a heterogeneous childhood cancer with a slightly higher incidence in boys than girls, with the reason for this gender disparity unknown. Given the growing evidence for the involvement of loss of the Y chromosome (LoY) in male diseases including cancer, we investigated Y chromosome status in NB. Male NB tumor samples from a Swedish cohort, analyzed using Cytoscan HD SNP-microarray, were selected. Seventy NB tumors were analyzed for aneuploidy of the Y chromosome, and these data were correlated with other genetic, biological, and clinical parameters. LoY was found in 21% of the male NB tumors and it was almost exclusively found in those with high-risk genomic profiles. Furthermore, LoY was associated with increased age at diagnosis and enriched in tumors with 11q-deletion and activated telomere maintenance mechanisms. In contrast, tumors with an MYCN-amplified genomic profile retained their Y chromosome. The understanding of LoY in cancer is limited, making it difficult to conclude whether LoY is a driving event in NB or function of increased genomic instability. Gene expression analysis of Y chromosome genes in male NB tumors showed low expression of certain genes correlating with worse overall survival. KDM5D, encoding a histone demethylase stands out as an interesting candidate for further studies. LoY has been shown to impact the epigenomic layer of autosomal loci in nonreproductive tissues, and KDM5D has been reported as downregulated and/or associated with poor survival in different malignancies. Further studies are needed to explore the mechanisms and functional consequences of LoY in NB.
Collapse
Affiliation(s)
- Anna Djos
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Johanna Svensson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jennie Gaarder
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ganesh Umapathy
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Staffan Nilsson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Torben Ek
- Children's Cancer Centre, Queen Silvia Children's Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Hartmut Vogt
- Crown Princess Victoria Children's Hospital, Division of Children's and Women's Health, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Kleopatra Georgantzi
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, and Pediatric Oncology, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Ingrid Öra
- Department of Pediatric Oncology, Skåne University Hospital, Lund, Sweden
| | - Catarina Träger
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Per Kogner
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, and Pediatric Oncology, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Tommy Martinsson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Susanne Fransson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
18
|
de Faria LL, Ponich Clementino C, Véras FASE, Khalil DDC, Otto DY, Oranges Filho M, Suzuki L, Bedoya MA. Staging and Restaging Pediatric Abdominal and Pelvic Tumors: A Practical Guide. Radiographics 2024; 44:e230175. [PMID: 38722785 DOI: 10.1148/rg.230175] [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: 06/06/2024]
Abstract
The most common abdominal malignancies diagnosed in the pediatric population include neuroblastoma, Wilms tumor, hepatoblastoma, lymphoma, germ cell tumor, and rhabdomyosarcoma. There are distinctive imaging findings and patterns of spread for each of these tumors that radiologists must know for diagnosis and staging and for monitoring the patient's response to treatment. The multidisciplinary treatment group that includes oncologists, surgeons, and radiation oncologists relies heavily on imaging evaluation to identify the best treatment course and prognostication of imaging findings, such as the image-defined risk factors for neuroblastomas, the PRETreatment EXtent of Disease staging system for hepatoblastoma, and the Ann Arbor staging system for lymphomas. It is imperative for radiologists to be able to correctly indicate the best imaging methods for diagnosis, staging, and restaging of each of these most prevalent tumors to avoid inconclusive or unnecessary examinations. The authors review in a practical manner the most updated key points in diagnosing and staging disease and assessing response to treatment of the most common pediatric abdominal tumors. ©RSNA, 2024 Supplemental material is available for this article.
Collapse
Affiliation(s)
- Luisa Leitão de Faria
- From the Radiology Department, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Rua Dr Ovídio Pires de Campos, 225 Cerqueira César, São Paulo, SP 36070-460, Brazil (L.L.d.F., C.P.C., F.A.S.e.V., D.d.C.K., D.Y.O., M.O.F., L.S.); and Department of Radiology, Boston Childrens Hospital, Harvard Medical School, Boston, Mass (M.A.B.)
| | - Carolina Ponich Clementino
- From the Radiology Department, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Rua Dr Ovídio Pires de Campos, 225 Cerqueira César, São Paulo, SP 36070-460, Brazil (L.L.d.F., C.P.C., F.A.S.e.V., D.d.C.K., D.Y.O., M.O.F., L.S.); and Department of Radiology, Boston Childrens Hospital, Harvard Medical School, Boston, Mass (M.A.B.)
| | - Felippe Augusto Silvestre E Véras
- From the Radiology Department, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Rua Dr Ovídio Pires de Campos, 225 Cerqueira César, São Paulo, SP 36070-460, Brazil (L.L.d.F., C.P.C., F.A.S.e.V., D.d.C.K., D.Y.O., M.O.F., L.S.); and Department of Radiology, Boston Childrens Hospital, Harvard Medical School, Boston, Mass (M.A.B.)
| | - Douglas da Cunha Khalil
- From the Radiology Department, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Rua Dr Ovídio Pires de Campos, 225 Cerqueira César, São Paulo, SP 36070-460, Brazil (L.L.d.F., C.P.C., F.A.S.e.V., D.d.C.K., D.Y.O., M.O.F., L.S.); and Department of Radiology, Boston Childrens Hospital, Harvard Medical School, Boston, Mass (M.A.B.)
| | - Deborah Yukiko Otto
- From the Radiology Department, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Rua Dr Ovídio Pires de Campos, 225 Cerqueira César, São Paulo, SP 36070-460, Brazil (L.L.d.F., C.P.C., F.A.S.e.V., D.d.C.K., D.Y.O., M.O.F., L.S.); and Department of Radiology, Boston Childrens Hospital, Harvard Medical School, Boston, Mass (M.A.B.)
| | - Marcelo Oranges Filho
- From the Radiology Department, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Rua Dr Ovídio Pires de Campos, 225 Cerqueira César, São Paulo, SP 36070-460, Brazil (L.L.d.F., C.P.C., F.A.S.e.V., D.d.C.K., D.Y.O., M.O.F., L.S.); and Department of Radiology, Boston Childrens Hospital, Harvard Medical School, Boston, Mass (M.A.B.)
| | - Lisa Suzuki
- From the Radiology Department, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Rua Dr Ovídio Pires de Campos, 225 Cerqueira César, São Paulo, SP 36070-460, Brazil (L.L.d.F., C.P.C., F.A.S.e.V., D.d.C.K., D.Y.O., M.O.F., L.S.); and Department of Radiology, Boston Childrens Hospital, Harvard Medical School, Boston, Mass (M.A.B.)
| | - M Alejandra Bedoya
- From the Radiology Department, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Rua Dr Ovídio Pires de Campos, 225 Cerqueira César, São Paulo, SP 36070-460, Brazil (L.L.d.F., C.P.C., F.A.S.e.V., D.d.C.K., D.Y.O., M.O.F., L.S.); and Department of Radiology, Boston Childrens Hospital, Harvard Medical School, Boston, Mass (M.A.B.)
| |
Collapse
|
19
|
Murphy C, Devis-Jauregui L, Struck R, Boloix A, Gallagher C, Gavin C, Cottone F, Fernandez AS, Madden S, Roma J, Segura MF, Piskareva O. In vivo cisplatin-resistant neuroblastoma metastatic model reveals tumour necrosis factor receptor superfamily member 4 (TNFRSF4) as an independent prognostic factor of survival in neuroblastoma. PLoS One 2024; 19:e0303643. [PMID: 38809883 PMCID: PMC11135766 DOI: 10.1371/journal.pone.0303643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/29/2024] [Indexed: 05/31/2024] Open
Abstract
Neuroblastoma is the most common solid extracranial tumour in children. Despite major advances in available therapies, children with drug-resistant and/or recurrent neuroblastoma have a dismal outlook with 5-year survival rates of less than 20%. Therefore, tackling relapsed tumour biology by developing and characterising clinically relevant models is a priority in finding targetable vulnerability in neuroblastoma. Using matched cisplatin-sensitive KellyLuc and resistant KellyCis83Luc cell lines, we developed a cisplatin-resistant metastatic MYCN-amplified neuroblastoma model. The average number of metastases per mouse was significantly higher in the KellyCis83Luc group than in the KellyLuc group. The vast majority of sites were confirmed as having lymph node metastasis. Their stiffness characteristics of lymph node metastasis values were within the range reported for the patient samples. Targeted transcriptomic profiling of immuno-oncology genes identified tumour necrosis factor receptor superfamily member 4 (TNFRSF4) as a significantly dysregulated MYCN-independent gene. Importantly, differential TNFRSF4 expression was identified in tumour cells rather than lymphocytes. Low TNFRSF4 expression correlated with poor prognostic indicators in neuroblastoma, such as age at diagnosis, stage, and risk stratification and significantly associated with reduced probability of both event-free and overall survival in neuroblastoma. Therefore, TNFRSF4 Low expression is an independent prognostic factor of survival in neuroblastoma.
Collapse
Affiliation(s)
- Catherine Murphy
- Department of Anatomy and Regenerative Medicine, Cancer Bioengineering Group, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Department of Anatomy and Regenerative Medicine, Tissue Engineering Research Group (TERG), RCSI University of Medicine and Health Sciences, Dublin, Ireland
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Laura Devis-Jauregui
- Faculty of Medicine, Cell Biology Unit, Department of Pathology and Experimental Therapeutics, University of Barcelona, Campus Bellvitge, Feixa Llarga s/n, L’Hospitalet de Llobregat, Spain
| | - Ronja Struck
- Department of Anatomy and Regenerative Medicine, Cancer Bioengineering Group, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Department of Anatomy and Regenerative Medicine, Tissue Engineering Research Group (TERG), RCSI University of Medicine and Health Sciences, Dublin, Ireland
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Ariadna Boloix
- Vall d’Hebron Research Institute, Group of Childhood Cancer & Blood Disorders, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ciara Gallagher
- Department of Anatomy and Regenerative Medicine, Cancer Bioengineering Group, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Department of Anatomy and Regenerative Medicine, Tissue Engineering Research Group (TERG), RCSI University of Medicine and Health Sciences, Dublin, Ireland
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Cian Gavin
- Department of Anatomy and Regenerative Medicine, Cancer Bioengineering Group, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Federica Cottone
- Department of Anatomy and Regenerative Medicine, Cancer Bioengineering Group, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Department of Anatomy and Regenerative Medicine, Tissue Engineering Research Group (TERG), RCSI University of Medicine and Health Sciences, Dublin, Ireland
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Aroa Soriano Fernandez
- Vall d’Hebron Research Institute, Group of Childhood Cancer & Blood Disorders, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Stephen Madden
- Data Science Centre, School of Population Health, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Josep Roma
- Vall d’Hebron Research Institute, Group of Childhood Cancer & Blood Disorders, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Miguel F. Segura
- Vall d’Hebron Research Institute, Group of Childhood Cancer & Blood Disorders, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Olga Piskareva
- Department of Anatomy and Regenerative Medicine, Cancer Bioengineering Group, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Department of Anatomy and Regenerative Medicine, Tissue Engineering Research Group (TERG), RCSI University of Medicine and Health Sciences, Dublin, Ireland
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| |
Collapse
|
20
|
Yeom A, Yoo SY, Jeon TY, Park J, Kim JH. [MR Imaging Features of Pediatric Bone Marrow]. JOURNAL OF THE KOREAN SOCIETY OF RADIOLOGY 2024; 85:505-519. [PMID: 38873387 PMCID: PMC11166593 DOI: 10.3348/jksr.2024.0039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 05/01/2024] [Accepted: 05/16/2024] [Indexed: 06/15/2024]
Abstract
MRI plays a crucial role in bone marrow (BM) assessment, and has very high sensitivity in diagnosing marrow disorders. However, for radiologists who may not frequently encounter pediatric imaging, distinguishing pathologic BM lesion from normal BM can be challenging. Conditions involving the BM in pediatric patients, such as leukemia and metastatic neuroblastoma, often manifest with diverse musculoskeletal symptoms and may be diagnosed using musculoskeletal MRI examinations. Accurate interpretation of pediatric MRI requires not only an understanding of the normal composition of BM but also an awareness of agerelated developmental changes in the marrow and familiarity with conditions that commonly involve pediatric BM. We aim to describe the composition of normal BM and outline the normal and abnormal MRI findings in pediatric BM. Additionally, we aim to present clinical cases of malignant BM disorders including leukemia, neuroblastoma metastasis, and other malignant BM disorders.
Collapse
|
21
|
Khan D, Sagar S, Shamim SA, Kaushik P, Kumar R. An Unusual Occurrence of Metastases to Multiple Muscles in Neuroblastoma. Indian J Nucl Med 2024; 39:222-223. [PMID: 39291061 PMCID: PMC11404728 DOI: 10.4103/ijnm.ijnm_37_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/18/2023] [Accepted: 05/06/2023] [Indexed: 09/19/2024] Open
Abstract
Neuroblastoma presenting with multiple muscles and subcutaneous tissue metastases is rarely reported in the literature. We would like to highlight such infrequent occurrences for increasing the clinical acumen of the medical fraternity with an aim to deliver proper therapy to patients.
Collapse
Affiliation(s)
- Dikhra Khan
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Sambit Sagar
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Shamim Ahmed Shamim
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Prateek Kaushik
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Rakesh Kumar
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
22
|
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
|
23
|
Dhamdhere MR, Spiegelman VS. Extracellular vesicles in neuroblastoma: role in progression, resistance to therapy and diagnostics. Front Immunol 2024; 15:1385875. [PMID: 38660306 PMCID: PMC11041043 DOI: 10.3389/fimmu.2024.1385875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 03/22/2024] [Indexed: 04/26/2024] Open
Abstract
Neuroblastoma (NB) is the most common extracranial solid pediatric cancer, and is one of the leading causes of cancer-related deaths in children. Despite the current multi-modal treatment regimens, majority of patients with advanced-stage NBs develop therapeutic resistance and relapse, leading to poor disease outcomes. There is a large body of knowledge on pathophysiological role of small extracellular vesicles (EVs) in progression and metastasis of multiple cancer types, however, the importance of EVs in NB was until recently not well understood. Studies emerging in the last few years have demonstrated the involvement of EVs in various aspects of NB pathogenesis. In this review we summarize these recent findings and advances on the role EVs play in NB progression, such as tumor growth, metastasis and therapeutic resistance, that could be helpful for future investigations in NB EV research. We also discuss different strategies for therapeutic targeting of NB-EVs as well as utilization of NB-EVs as potential biomarkers.
Collapse
Affiliation(s)
| | - Vladimir S. Spiegelman
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| |
Collapse
|
24
|
Newman J, Leelatian N, Liang J. Characterization of pediatric non-hematopoietic tumor metastases to the central nervous system: A single institution review. J Neuropathol Exp Neurol 2024; 83:268-275. [PMID: 38350468 DOI: 10.1093/jnen/nlae012] [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] [Indexed: 02/15/2024] Open
Abstract
Central nervous system (CNS) metastases represent a small portion of pediatric CNS neoplasms and data surrounding this condition with high morbidity is scarce. Single institutional archival institutional pathology records between 1999 and 2022 were searched for patients over 21 years old and younger with CNS, dura, cranial nerve, CSF, or leptomeningeal metastases; 41 cases were identified. We documented primary tumor types and locations, metastasis locations, types of invasion (direct extension vs distant metastasis), times from imaging or pathologic diagnosis to CNS involvement, and outcomes. Distant metastasis was the most common mechanism of metastasis (n = 32, 78%). Interval times to CNS metastasis varied by both tumor type and primary tumor location. In this cohort, osteosarcoma portended the shortest survival following CNS metastasis. This study highlights the diverse mechanisms and locations of CNS involvement in pediatric CNS metastases and illuminates a need for varied monitoring strategies when considering primary tumor type and anatomic location.
Collapse
Affiliation(s)
- John Newman
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nalin Leelatian
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jiancong Liang
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
25
|
Zhao X, Xu Z, Feng X. Clinical characteristics and prognoses in pediatric neuroblastoma with bone or liver metastasis: data from the SEER 2010-2019. BMC Pediatr 2024; 24:162. [PMID: 38454422 PMCID: PMC10921780 DOI: 10.1186/s12887-024-04570-z] [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: 07/03/2023] [Accepted: 01/16/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND To investigate clinical characteristics, prognoses, and impacts of treatments on prognoses of neuroblastoma patients with bone or liver metastasis. METHODS This retrospective cohort study extracted data from the Surveillance, Epidemiology, and End Results (SEER) database 2010-2019. The outcomes were 3-year cancer-specific survival (CSS) and 5-year CSS. Multivariable COX risk proportional models were established to assess the association between metastasis types and CSS. Hazard ratios (HRs) and 95% confidence intervals (CIs) were estimated. RESULTS Totally 425 patients with metastatic neuroblastoma were eligible for 3-year CSS analysis and 320 for 5-year CSS analysis. For 3-year follow-up, 62 (14.59%) patients had liver metastasis alone, 289 (0.68%) had bone metastasis alone, and 74 (17.41%) had both liver and bone metastasis. For 5-year follow-up, 44 (13.75%) patients had liver metastasis alone, 223 (69.69%) had bone metastasis alone, and 53 (16.56%) had both liver and bone metastasis. Significant differences were observed in age, tumor size, surgery for the primary site, chemotherapy, radiation, brain metastasis, lung metastasis, and vital status between patients with liver metastasis alone, bone metastasis alone, and both liver and bone metastasis (all P < 0.05). Compared with patients with liver metastasis alone, patients with bone metastasis alone (HR = 2.30, 95%CI: 1.10-4.82, P = 0.028) or both (HR = 2.35, 95%CI: 1.06-5.20, P = 0.035) had significantly poorer 3-year CSS; patients with bone metastasis alone (HR = 2.32, 95%CI: 1.14-4.70, P = 0.020) or both liver and bone metastasis (HR = 2.33, 95%CI: 1.07-5.07, P = 0.032) exhibited significantly worse 5-year CSS than those with liver metastasis alone. In patients with bone metastasis, those with chemotherapy had significantly better 3-year CSS than those without (HR = 0.24, 95%CI: 0.07-0.75, P = 0.014). Among patients with liver metastasis, receiving radiation was associated with significantly worse 3-year CSS (HR = 2.00, 95%CI: 1.05-3.81, P = 0.035). CONCLUSION Compared with patients with liver metastasis alone, those with bone metastasis alone or both had poorer 3- and 5-year CSS. For patients with bone metastasis, undergoing chemotherapy was associated with better 3-year CSS. For patients with liver metastasis, receiving radiation was associated with worse 3-year CSS.
Collapse
Affiliation(s)
- Xudong Zhao
- Department of Pediatric Surgery, Zhongshan City People's Hospital, No. 2 Sunwen East Road, Guangdong, Zhongshan, 528400, People's Republic of China
| | - Zhuofan Xu
- Department of Pediatric Surgery, Zhongshan City People's Hospital, No. 2 Sunwen East Road, Guangdong, Zhongshan, 528400, People's Republic of China
| | - Xiaochuan Feng
- Department of Pediatric Surgery, Zhongshan City People's Hospital, No. 2 Sunwen East Road, Guangdong, Zhongshan, 528400, People's Republic of China.
| |
Collapse
|
26
|
Feng L, Zhou Z, Liu J, Yao S, Wang C, Zhang H, Xiong P, Wang W, Yang J. 18F-FDG PET/CT-Based Radiomics Nomogram for Prediction of Bone Marrow Involvement in Pediatric Neuroblastoma: A Two-Center Study. Acad Radiol 2024; 31:1111-1121. [PMID: 37643929 DOI: 10.1016/j.acra.2023.07.018] [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: 06/13/2023] [Revised: 07/13/2023] [Accepted: 07/19/2023] [Indexed: 08/31/2023]
Abstract
RATIONALE AND OBJECTIVES To assess the predictive ability of an 18F-FDG PET/CT-based radiomics nomogram for bone marrow involvement in pediatric neuroblastoma. MATERIALS AND METHODS A total of 241 neuroblastoma patients who underwent 18F-FDG PET/CT at two medical centers were retrospectively evaluated. Data from center A (n = 200) were randomized into a training cohort (n = 140) and an internal validation cohort (n = 60), while data from center B (n = 41) constituted the external validation cohort. For each patient, two regions of interest were defined using the tumor and axial skeleton. The clinical factors and radiomics features were derived to construct the clinical and radiomics models. The radiomics nomogram was built by combining clinical factors and radiomics features. The area under the receiver operating characteristic curves (AUCs) were used to assess the performance of the models. RESULTS Radiomics models created from tumor and axial skeleton achieved AUCs of 0.773 and 0.900, and the clinical model had an AUC of 0.858 in the training cohort. By incorporating clinical risk factors and axial skeleton-based radiomics features, the AUC of the radiomics nomogram in the training cohort, internal validation cohort, and external validation cohort was 0.932, 0.887, and 0.733, respectively. CONCLUSION The axial skeleton-based radiomics model performed better than the tumor-based radiomics model in predicting bone marrow involvement. Moreover, the radiomics nomogram showed that combining axial skeleton-based radiomics features with clinical risk factors improved their performance.
Collapse
Affiliation(s)
- Lijuan Feng
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China (L.F., Z.Z., J.L., W.W., J.Y.)
| | - Ziang Zhou
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China (L.F., Z.Z., J.L., W.W., J.Y.)
| | - Jun Liu
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China (L.F., Z.Z., J.L., W.W., J.Y.)
| | - Shuang Yao
- Department of Nuclear Medicine, Beijing Fengtai YouAnMen Hospital, Beijing, China (S.Y.)
| | - Chao Wang
- Department of Clinical Research, SinoUnion Healthcare Inc., Beijing, China (C.W.)
| | - Hui Zhang
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China (H.Z.)
| | - Pingxiang Xiong
- Nanchang Rimag Medical Diagnosis Center, Nanchang, China (P.X.)
| | - Wei Wang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China (L.F., Z.Z., J.L., W.W., J.Y.)
| | - Jigang Yang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China (L.F., Z.Z., J.L., W.W., J.Y.).
| |
Collapse
|
27
|
Ouvrard E, Kaseb A, Poterszman N, Porot C, Somme F, Imperiale A. Nuclear medicine imaging for bone metastases assessment: what else besides bone scintigraphy in the era of personalized medicine? Front Med (Lausanne) 2024; 10:1320574. [PMID: 38288299 PMCID: PMC10823373 DOI: 10.3389/fmed.2023.1320574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 12/28/2023] [Indexed: 01/31/2024] Open
Abstract
Accurate detection and reliable assessment of therapeutic responses in bone metastases are imperative for guiding treatment decisions, preserving quality of life, and ultimately enhancing overall survival. Nuclear imaging has historically played a pivotal role in this realm, offering a diverse range of radiotracers and imaging modalities. While the conventional bone scan using 99mTc marked bisphosphonates has remained widely utilized, its diagnostic performance is hindered by certain limitations. Positron emission tomography, particularly when coupled with computed tomography, provides improved spatial resolution and diagnostic performance with various pathology-specific radiotracers. This review aims to evaluate the performance of different nuclear imaging modalities in clinical practice for detecting and monitoring the therapeutic responses in bone metastases of diverse origins, addressing their limitations and implications for image interpretation.
Collapse
Affiliation(s)
- Eric Ouvrard
- Nuclear Medicine and Molecular Imaging, Institut de Cancérologie Strasbourg Europe (ICANS), University Hospitals of Strasbourg, University of Strasbourg, Strasbourg, France
| | - Ashjan Kaseb
- Nuclear Medicine and Molecular Imaging, Institut de Cancérologie Strasbourg Europe (ICANS), University Hospitals of Strasbourg, University of Strasbourg, Strasbourg, France
- Radiology, College of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Nathan Poterszman
- Nuclear Medicine and Molecular Imaging, Institut de Cancérologie Strasbourg Europe (ICANS), University Hospitals of Strasbourg, University of Strasbourg, Strasbourg, France
| | - Clémence Porot
- Radiopharmacy, Institut de Cancérologie Strasbourg Europe (ICANS), Strasbourg, France
| | - Francois Somme
- Nuclear Medicine and Molecular Imaging, Institut de Cancérologie Strasbourg Europe (ICANS), University Hospitals of Strasbourg, University of Strasbourg, Strasbourg, France
| | - Alessio Imperiale
- Nuclear Medicine and Molecular Imaging, Institut de Cancérologie Strasbourg Europe (ICANS), University Hospitals of Strasbourg, University of Strasbourg, Strasbourg, France
- IPHC, UMR 7178, CNRS/Unistra, Strasbourg, France
| |
Collapse
|
28
|
Kushner BH, LaQuaglia MP, Cardenas FI, Basu EM, Gerstle JT, Kramer K, Roberts SS, Wolden SL, Cheung NKV, Modak S. Stage 4N neuroblastoma before and during the era of anti-G D2 immunotherapy. Int J Cancer 2023; 153:2019-2031. [PMID: 37602920 PMCID: PMC11925214 DOI: 10.1002/ijc.34693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/28/2023] [Accepted: 08/02/2023] [Indexed: 08/22/2023]
Abstract
Patients with stage 4N neuroblastoma (distant metastases limited to lymph nodes) stand out as virtually the only survivors of high-risk neuroblastoma (HR-NB) before myeloablative therapy (MAT) and immunotherapy with anti-GD2 monoclonal antibodies (mAbs) became standard. Because no report presents more recent results with 4N, we analyzed our large 4N experience. All 51 pediatric 4N patients (<18 years old) diagnosed 1985 to 2021 were reviewed. HR-NB included MYCN-nonamplified 4N diagnosed at age ≥18 months and MYCN-amplified 4N. Among 34 MYCN-nonamplified high-risk patients, 20 are relapse-free 1.5+ to 37.5+ (median 12.5+) years post-diagnosis, including 13 without prior MAT and 5 treated with little (1 cycle; n = 2) or no mAb (n = 3), while 14 patients (7 post-MAT, 8 post-mAbs) relapsed (all soft tissue). Of 15 MYCN-amplified 4N patients, 7 are relapse-free 2.1+ to 26.4+ (median 11.6+) years from the start of chemotherapy (all received mAbs; 3 underwent MAT) and 4 are in second remission 4.2+ to 21.8+ years postrelapse (all soft tissue). Statistical analyses showed no significant association of survival with either MAT or mAbs for MYCN-nonamplified HR-NB; small numbers prevented these analyses for MYCN-amplified patients. The two patients with intermediate-risk 4N (14-months-old) are relapse-free 7+ years postresection of primary tumors; distant disease spontaneously regressed. The natural history of 4N is marked by NB confined to soft tissue without early relapse in bones or bone marrow, where mAbs have proven efficacy. These findings plus curability without MAT, as seen elsewhere and at our center, support consideration of treatment reduction for MYCN-nonamplified 4N.
Collapse
Affiliation(s)
- Brian H Kushner
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Michael P LaQuaglia
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | - Ellen M Basu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Justin T Gerstle
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Kim Kramer
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Stephen S Roberts
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Suzanne L Wolden
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Shakeel Modak
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
29
|
Scherer S, Mayer BFB, Dietzel M, Esser M, Warmann SW, Lang P, Schuhmann MU, Schmidt A, Fuchs J. The role of surgery in the treatment of neuroblastoma metastases at rare sites. J Cancer Res Clin Oncol 2023; 149:12913-12921. [PMID: 37466795 PMCID: PMC10587098 DOI: 10.1007/s00432-023-05147-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 07/08/2023] [Indexed: 07/20/2023]
Abstract
PURPOSE Treatment of neuroblastoma metastases usually consists of chemotherapy and irradiation. However, in selected cases, surgical treatment is also indicated. In this study, we present three cases of patients with neuroblastoma metastases at rare sites that underwent surgery. MATERIALS AND METHODS We retrospectively analyzed data of patients who underwent surgery for neuroblastoma at our department of Pediatric Surgery and Pediatric Urology at the University Children's Hospital in Tuebingen and selected those patients who had surgery explicitly for a metastasis. RESULTS Between 2002 and 2020, 277 children underwent surgical treatment for neuroblastoma. Three cases with metastases at exceptional sites are presented here after therapy according to protocols. One patient had a penile metastasis and received surgery including a plastic reconstruction. The patient showed no signs of erectile or urinary dysfunction at follow-up. Another patient had a metastasis in the proximal ulna, which remained vital even after exhausted treatment after two relapses. Afterward there was no restriction of movement of the extremity. The third patient had, amongst others, metastases to the pancreatic body and to the liver. Both were surgically removed during primary tumor resection. This patient died after local tumor relapse. The other two patients showed no evidence of tumor relapse after a follow-up of 18 and 17 months, respectively. CONCLUSION Although children with neuroblastoma often present with metastases, there is no recommendation for surgical treatment other than diagnostic biopsies. In case of persistence of metastasis or after exhaustion of high-risk therapy, surgical resection must be considered.
Collapse
Affiliation(s)
- Simon Scherer
- Department of Pediatric Surgery and Pediatric Urology, University Children's Hospital Tuebingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany.
| | - Benjamin F B Mayer
- Department of Pediatric Surgery and Pediatric Urology, University Children's Hospital Tuebingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany
| | - Markus Dietzel
- Department of Pediatric Surgery and Pediatric Urology, University Children's Hospital Tuebingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany
| | - Michael Esser
- Department of Diagnostic and Interventional Radiology, University Hospital Tuebingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany
| | - Steven W Warmann
- Department of Pediatric Surgery and Pediatric Urology, University Children's Hospital Tuebingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany
| | - Peter Lang
- Department of General Pediatrics, Hematology and Oncology, University Children's Hospital Tuebingen, Hoppe-Seyler-Str. 1, 72076, Tübingen, Germany
| | - Martin U Schuhmann
- Department of Neurosurgery, University Hospital Tuebingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany
| | - Andreas Schmidt
- Department of Pediatric Surgery and Pediatric Urology, University Children's Hospital Tuebingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany
| | - Jörg Fuchs
- Department of Pediatric Surgery and Pediatric Urology, University Children's Hospital Tuebingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany
| |
Collapse
|
30
|
Jiang P, Zhang T, Wu B, Li X, Fu M, Xu B. Musashi-2 (MSI2) promotes neuroblastoma tumorigenesis through targeting MYC-mediated glucose-6-phosphate dehydrogenase (G6PD) transcriptional activation. Med Oncol 2023; 40:332. [PMID: 37843625 DOI: 10.1007/s12032-023-02199-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 09/19/2023] [Indexed: 10/17/2023]
Abstract
Neuroblastoma (NB) is the deadliest pediatric solid tumor due to its rapid proliferation. Aberrant expression of MYCN is deemed as the most remarkable feature for the predictive hallmark of NB progression and recurrence. However, the phenomenon that only detection of MYCN in the nearly 20% of NB patients hints that there should be other vital oncogenes in the progression of NB. Here, we firstly show that MSI2 mRNA is augmented by analyzing public GEO datasets in the malignant stage according to International Neuroblastoma Staging System (INSS) stages. Although accumulating evidences uncover the emerging roles of MSI2 in several cancers, the regulatory functions and underlying mechanisms of MSI2 in NB remain under-investigated. Herein, we identified that high-expressed MSI2 and low-expressed n-Myc group account for 43.1% of total NB clinical samples (n = 65). Meanwhile, MSI2 expression is profoundly upregulated along with NB malignancy and negatively associated with the survival outcome of NB patients in the NB tissue microarray (NB: n = 65; Ganglioneuroblastoma: n = 31; Ganglioneuroma: n = 27). In vitro, our results revealed that MSI2 promoted migration, invasion, and proliferation of NB cells via enhancing pentose phosphate pathway. Mechanistically, MSI2 upregulated the key enzyme glucose-6-phosphate dehydrogenase (G6PD) via directly binding to 3'-untranslated regions of c-Myc mRNA to facilitate its stability, resulting in enhancing pentose phosphate pathway. Our findings reveal that MSI2 promotes pentose phosphate pathway via activating c-Myc-G6PD signaling, suggesting that MSI2 exhibits a novel and powerful target for the diagnosis and treatment of NB.
Collapse
Affiliation(s)
- Ping Jiang
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Ting Zhang
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Bin Wu
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Xiaoqing Li
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Mingpeng Fu
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Banglao Xu
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China.
| |
Collapse
|
31
|
Feng L, Zhang S, Lu X, Yang X, Kan Y, Wang C, Zhang H, Wang W, Yang J. An Optimal Radiomics Nomogram Based on 18F-FDG PET/CT for Identifying Event-Free Survival in Pediatric Neuroblastoma. Acad Radiol 2023; 30:2309-2320. [PMID: 37393177 DOI: 10.1016/j.acra.2023.06.004] [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: 04/18/2023] [Revised: 05/13/2023] [Accepted: 06/02/2023] [Indexed: 07/03/2023]
Abstract
RATIONALE AND OBJECTIVES To investigate whether the 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography/computed tomography (PET/CT) radiomics features that combine tumor and bone marrow can more accurately identify event-free survival (EFS) in pediatric neuroblastoma. MATERIALS AND METHODS A total of 126 patients with neuroblastoma were retrospectively included and randomly divided into the training and validation cohorts (7:3 ratio). Radiomics features were extracted to develop a tumor- and bone marrow-based radiomics risk score (RRS). The Kaplan-Meier method was used to evaluate the effectiveness of RRS in EFS risk stratification. Univariate and multivariate Cox regression analyses were used to determine independent clinical risk factors and construct the clinical models. The conventional PET model was constructed based on conventional PET parameters, and the noninvasive combined model integrated the RRS and the noninvasive independent clinical risk factors. The performance of the models was evaluated using C-index, calibration curves, and decision curve analysis (DCA). RESULTS A total of 15 radiomics features were selected to build the RRS. According to Kaplan-Meier analysis, there was a significant difference in EFS between the low-risk and high-risk groups as defined by the value of RRS (P < .05). The noninvasive combined model combining RRS and the International Neuroblastoma Risk Group stage achieved the best prognostic prediction of EFS, with a C-index of 0.810 and 0.783 in the training and validation cohorts, respectively. The calibration curves and DCA indicated that the noninvasive combined model had good consistency and clinical utility. CONCLUSION The 18F-FDG PET/CT-based radiomics of neuroblastoma allows a reliable evaluation of EFS. The performance of the noninvasive combined model was superior to the clinical and conventional PET models.
Collapse
Affiliation(s)
- Lijuan Feng
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China (L.F., S.Z., X.L., X.Y., Y.K., W.W., J.Y.)
| | - Shuxin Zhang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China (L.F., S.Z., X.L., X.Y., Y.K., W.W., J.Y.)
| | - Xia Lu
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China (L.F., S.Z., X.L., X.Y., Y.K., W.W., J.Y.)
| | - Xu Yang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China (L.F., S.Z., X.L., X.Y., Y.K., W.W., J.Y.)
| | - Ying Kan
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China (L.F., S.Z., X.L., X.Y., Y.K., W.W., J.Y.)
| | - Chao Wang
- SinoUnion Healthcare Inc., Beijing, China (C.W.)
| | - Hui Zhang
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China (H,Z,)
| | - Wei Wang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China (L.F., S.Z., X.L., X.Y., Y.K., W.W., J.Y.)
| | - Jigang Yang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China (L.F., S.Z., X.L., X.Y., Y.K., W.W., J.Y.).
| |
Collapse
|
32
|
Dhamdhere MR, Spiegelman DV, Schneper L, Erbe AK, Sondel PM, Spiegelman VS. Generation of Novel Immunocompetent Mouse Cell Lines to Model Experimental Metastasis of High-Risk Neuroblastoma. Cancers (Basel) 2023; 15:4693. [PMID: 37835389 PMCID: PMC10571844 DOI: 10.3390/cancers15194693] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/18/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
NB, being a highly metastatic cancer, is one of the leading causes of cancer-related deaths in children. Increased disease recurrence and clinical resistance in patients with metastatic high-risk NBs (HR-NBs) result in poor outcomes and lower overall survival. However, the paucity of appropriate in vivo models for HR-NB metastasis has limited investigations into the underlying biology of HR-NB metastasis. This study was designed to address this limitation and develop suitable immunocompetent models for HR-NB metastasis. Here, we developed several highly metastatic immunocompetent murine HR-NB cell lines. Our newly developed cell lines show 100% efficiency in modeling experimental metastasis in C57BL6 mice and feature metastasis to the sites frequently observed in humans with HR-NB (liver and bone). In vivo validation demonstrated their specifically gained metastatic phenotype. The in vitro characterization of the cell lines showed increased cell invasion, acquired anchorage-independent growth ability, and resistance to MHC-I induction upon IFN-γ treatment. Furthermore, RNA-seq analysis of the newly developed cells identified a differentially regulated gene signature and an enrichment of processes consistent with their acquired metastatic phenotype, including extracellular matrix remodeling, angiogenesis, cell migration, and chemotaxis. The presented newly developed cell lines are, thus, suitable and promising tools for HR-NB metastasis and microenvironment studies in an immunocompetent system.
Collapse
Affiliation(s)
- Mayura R. Dhamdhere
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Dan V. Spiegelman
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53792, USA; (D.V.S.); (P.M.S.)
| | - Lisa Schneper
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Amy K. Erbe
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53792, USA; (D.V.S.); (P.M.S.)
| | - Paul M. Sondel
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53792, USA; (D.V.S.); (P.M.S.)
| | - Vladimir S. Spiegelman
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
33
|
Proestler E, Donzelli J, Nevermann S, Breitwieser K, Koch LF, Best T, Fauth M, Wickström M, Harter PN, Kogner P, Lavieu G, Larsson K, Saul MJ. The multiple functions of miR-574-5p in the neuroblastoma tumor microenvironment. Front Pharmacol 2023; 14:1183720. [PMID: 37731742 PMCID: PMC10507178 DOI: 10.3389/fphar.2023.1183720] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 08/07/2023] [Indexed: 09/22/2023] Open
Abstract
Neuroblastoma is the most common extracranial solid tumor in childhood and arises from neural crest cells of the developing sympathetic nervous system. Prostaglandin E2 (PGE2) has been identified as a key pro-inflammatory mediator of the tumor microenvironment (TME) that promotes neuroblastoma progression. We report that the interaction between the microRNA miR-574-5p and CUG-binding protein 1 (CUGBP1) induces the expression of microsomal prostaglandin E2 synthase 1 (mPGES-1) in neuroblastoma cells, which contributes to PGE2 biosynthesis. PGE2 in turn specifically induces the sorting of miR-574-5p into small extracellular vesicles (sEV) in neuroblastoma cell lines. sEV are one of the major players in intercellular communication in the TME. We found that sEV-derived miR-574-5p has a paracrine function in neuroblastoma. It acts as a direct Toll-like receptor 7/8 (TLR7/8) ligand and induces α-smooth muscle actin (α-SMA) expression in fibroblasts, contributing to fibroblast differentiation. This is particularly noteworthy as it has an opposite function to that in the TME of lung carcinoma, another PGE2 dependent tumor type. Here, sEV-derived miR-574-5p has an autokrine function that inhibits PGE2 biosynthesis in lung cancer cells. We report that the tetraspanin composition on the surface of sEV is associated with the function of sEV-derived miR-574-5p. This suggests that the vesicles do not only transport miRs, but also appear to influence their mode of action.
Collapse
Affiliation(s)
- Eva Proestler
- Fachbereich Biologie, Technische Universität Darmstadt, Darmstadt, Germany
| | - Julia Donzelli
- Fachbereich Biologie, Technische Universität Darmstadt, Darmstadt, Germany
| | - Sheila Nevermann
- Fachbereich Biologie, Technische Universität Darmstadt, Darmstadt, Germany
| | - Kai Breitwieser
- Fachbereich Biologie, Technische Universität Darmstadt, Darmstadt, Germany
| | - Leon F. Koch
- Fachbereich Biologie, Technische Universität Darmstadt, Darmstadt, Germany
| | - Tatjana Best
- Fachbereich Biologie, Technische Universität Darmstadt, Darmstadt, Germany
- Merck KGaA, Darmstadt, Germany
| | - Maria Fauth
- Fachbereich Biologie, Technische Universität Darmstadt, Darmstadt, Germany
- Prolytic GmbH, a Kymos Company, Frankfurt, Germany
| | - Malin Wickström
- Childhood Cancer Research Unit, Department of Children’s and Women’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Patrick N. Harter
- Institute of Neurology (Edinger-Institute), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Frankfurt, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt am Main, Frankfurt, Germany
| | - Per Kogner
- Childhood Cancer Research Unit, Department of Children’s and Women’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Grégory Lavieu
- INSERM U1316, UMR7057, Centre National de la Recherche Scientifique (CNRS), Université Paris Cité, Paris, France
| | - Karin Larsson
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Meike J. Saul
- Fachbereich Biologie, Technische Universität Darmstadt, Darmstadt, Germany
| |
Collapse
|
34
|
Smiles WJ, Catalano L, Stefan VE, Weber DD, Kofler B. Metabolic protein kinase signalling in neuroblastoma. Mol Metab 2023; 75:101771. [PMID: 37414143 PMCID: PMC10362370 DOI: 10.1016/j.molmet.2023.101771] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/20/2023] [Accepted: 06/30/2023] [Indexed: 07/08/2023] Open
Abstract
BACKGROUND Neuroblastoma is a paediatric malignancy of incredibly complex aetiology. Oncogenic protein kinase signalling in neuroblastoma has conventionally focussed on transduction through the well-characterised PI3K/Akt and MAPK pathways, in which the latter has been implicated in treatment resistance. The discovery of the receptor tyrosine kinase ALK as a target of genetic alterations in cases of familial and sporadic neuroblastoma, was a breakthrough in the understanding of the complex genetic heterogeneity of neuroblastoma. However, despite progress in the development of small-molecule inhibitors of ALK, treatment resistance frequently arises and appears to be a feature of the disease. Moreover, since the identification of ALK, several additional protein kinases, including the PIM and Aurora kinases, have emerged not only as drivers of the disease phenotype, but also as promising druggable targets. This is particularly the case for Aurora-A, given its intimate engagement with MYCN, a driver oncogene of aggressive neuroblastoma previously considered 'undruggable.' SCOPE OF REVIEW Aided by significant advances in structural biology and a broader understanding of the mechanisms of protein kinase function and regulation, we comprehensively outline the role of protein kinase signalling, emphasising ALK, PIM and Aurora in neuroblastoma, their respective metabolic outputs, and broader implications for targeted therapies. MAJOR CONCLUSIONS Despite massively divergent regulatory mechanisms, ALK, PIM and Aurora kinases all obtain significant roles in cellular glycolytic and mitochondrial metabolism and neuroblastoma progression, and in several instances are implicated in treatment resistance. While metabolism of neuroblastoma tends to display hallmarks of the glycolytic "Warburg effect," aggressive, in particular MYCN-amplified tumours, retain functional mitochondrial metabolism, allowing for survival and proliferation under nutrient stress. Future strategies employing specific kinase inhibitors as part of the treatment regimen should consider combinatorial attempts at interfering with tumour metabolism, either through metabolic pathway inhibitors, or by dietary means, with a view to abolish metabolic flexibility that endows cancerous cells with a survival advantage.
Collapse
Affiliation(s)
- William J Smiles
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Müllner Hauptstraße 48, 5020, Salzburg, Austria.
| | - Luca Catalano
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Müllner Hauptstraße 48, 5020, Salzburg, Austria
| | - Victoria E Stefan
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Müllner Hauptstraße 48, 5020, Salzburg, Austria
| | - Daniela D Weber
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Müllner Hauptstraße 48, 5020, Salzburg, Austria
| | - Barbara Kofler
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Müllner Hauptstraße 48, 5020, Salzburg, Austria
| |
Collapse
|
35
|
Serter Kocoglu S, Oy C, Secme M, Sunay FB. Investigation of the anticancer mechanism of monensin via apoptosis-related factors in SH-SY5Y neuroblastoma cells. Clin Transl Sci 2023; 16:1725-1735. [PMID: 37477356 PMCID: PMC10499413 DOI: 10.1111/cts.13593] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/22/2023] Open
Abstract
Monensin is an ionophore antibiotic that inhibits the growth of cancer cells. The aim of this study was to investigate the apoptosis-mediated anticarcinogenic effects of monensin in SH-SY5Y neuroblastoma cells. The effects of monensin on cell viability, invasion, migration, and colony formation were determined by XTT, matrigel-chamber, wound healing, and colony formation tests, respectively. The effects of monensin on apoptosis were determined by real-time polymerase chain reaction, TUNEL, Western blot, and Annexin V assay. We have shown that monensin suppresses neuroblastoma cell viability, invasion, migration, and colony formation. Moreover, we reported that monensin inhibits cell viability by triggering apoptosis of neuroblastoma cells. Monensin caused apoptosis by increasing caspase-3, 7, 8, and 9 expressions and decreasing Bax and Bcl-2 expressions in neuroblastoma cells. In Annexin V results, the rates of apoptotic cells were found to be 9.66 ± 0.01% (p < 0.001), 29.28 ± 0.88% (p < 0.01), and 62.55 ± 2.36% (p < 0.01) in the 8, 16, and 32 μM monensin groups, respectively. In TUNEL results, these values were, respectively; 35 ± 2% (p < 0.001), 34 ± 0.57% (p < 0.001), and 75 ± 2.51% (p < 0.001). Our results suggest that monensin may be a safe and effective therapeutic candidate for treating pediatric neuroblastoma.
Collapse
Affiliation(s)
- Sema Serter Kocoglu
- Department of Histology and EmbryologySchool of Medicine, Balikesir UniversityBalikesirTurkey
| | - Ceren Oy
- Department of Histology and EmbryologySchool of Medicine, Bursa Uludag UniversityBursaTurkey
| | - Mücahit Secme
- Department of Medical BiologySchool of Medicine, Ordu UniversityDenizliTurkey
| | - F. Bahar Sunay
- Department of Histology and EmbryologySchool of Medicine, Balikesir UniversityBalikesirTurkey
| |
Collapse
|
36
|
Rahavi SM, Aletaha M, Farrokhi A, Lorentzian A, Lange PF, Maxwell CA, Lim CJ, Reid GSD. Adaptation of the Th-MYCN Mouse Model of Neuroblastoma for Evaluation of Disseminated Disease. Int J Mol Sci 2023; 24:12071. [PMID: 37569447 PMCID: PMC10419036 DOI: 10.3390/ijms241512071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/23/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
High-risk neuroblastoma remains a profound clinical challenge that requires eradication of neuroblastoma cells from a variety of organ sites, including bone marrow, liver, and CNS, to achieve a cure. While preclinical modeling is a powerful tool for the development of novel cancer therapies, the lack of widely available models of metastatic neuroblastoma represents a significant barrier to the development of effective treatment strategies. To address this need, we report a novel luciferase-expressing derivative of the widely used Th-MYCN mouse. While our model recapitulates the non-metastatic neuroblastoma development seen in the parental transgenic strain, transplantation of primary tumor cells from disease-bearing mice enables longitudinal monitoring of neuroblastoma growth at distinct sites in immune-deficient or immune-competent recipients. The transplanted tumors retain GD2 expression through many rounds of serial transplantation and are sensitive to GD2-targeted immune therapy. With more diverse tissue localization than is seen with human cell line-derived xenografts, this novel model for high-risk neuroblastoma could contribute to the optimization of immune-based treatments for this deadly disease.
Collapse
Affiliation(s)
- Seyed M. Rahavi
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Maryam Aletaha
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Ali Farrokhi
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
- Department of Pediatrics, University of British Columbia, 950 W28th Avenue, Vancouver, BC V5Z 4H4, Canada
| | - Amanda Lorentzian
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Philipp F. Lange
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
- Department of Pathology, University of British Columbia, 950 W28th Avenue, Vancouver, BC V5Z 4H4, Canada
| | - Christopher A. Maxwell
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
- Department of Pediatrics, University of British Columbia, 950 W28th Avenue, Vancouver, BC V5Z 4H4, Canada
| | - Chinten James Lim
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
- Department of Pediatrics, University of British Columbia, 950 W28th Avenue, Vancouver, BC V5Z 4H4, Canada
| | - Gregor S. D. Reid
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
- Department of Pediatrics, University of British Columbia, 950 W28th Avenue, Vancouver, BC V5Z 4H4, Canada
| |
Collapse
|
37
|
Fanlo L, Gómez-González S, Rozalén C, Pérez-Núñez I, Sangrador I, Tomás-Daza L, Gautier EL, Usieto S, Rebollo E, Vila-Ubach M, Carcaboso AM, Javierre BM, Celià-Terrassa T, Lavarino C, Martí E, Le Dréau G. Neural crest-related NXPH1/α-NRXN signaling opposes neuroblastoma malignancy by inhibiting organotropic metastasis. Oncogene 2023:10.1038/s41388-023-02742-2. [PMID: 37301928 DOI: 10.1038/s41388-023-02742-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/16/2023] [Accepted: 06/01/2023] [Indexed: 06/12/2023]
Abstract
Neuroblastoma is a pediatric cancer that can present as low- or high-risk tumors (LR-NBs and HR-NBs), the latter group showing poor prognosis due to metastasis and strong resistance to current therapy. Whether LR-NBs and HR-NBs differ in the way they exploit the transcriptional program underlying their neural crest, sympatho-adrenal origin remains unclear. Here, we identified the transcriptional signature distinguishing LR-NBs from HR-NBs, which consists mainly of genes that belong to the core sympatho-adrenal developmental program and are associated with favorable patient prognosis and with diminished disease progression. Gain- and loss-of-function experiments revealed that the top candidate gene of this signature, Neurexophilin-1 (NXPH1), has a dual impact on NB cell behavior in vivo: whereas NXPH1 and its receptor α-NRXN1 promote NB tumor growth by stimulating cell proliferation, they conversely inhibit organotropic colonization and metastasis. As suggested by RNA-seq analyses, these effects might result from the ability of NXPH1/α-NRXN signalling to restrain the conversion of NB cells from an adrenergic state to a mesenchymal one. Our findings thus uncover a transcriptional module of the sympatho-adrenal program that opposes neuroblastoma malignancy by impeding metastasis, and pinpoint NXPH1/α-NRXN signaling as a promising target to treat HR-NBs.
Collapse
Affiliation(s)
- Lucía Fanlo
- Department of Cells and Tissues, Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, C/ Baldiri Reixac 10-15, 08028, Barcelona, Spain
| | - Soledad Gómez-González
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Déu, Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Catalina Rozalén
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), 08003, Barcelona, Spain
| | - Iván Pérez-Núñez
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), 08003, Barcelona, Spain
| | - Irene Sangrador
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), 08003, Barcelona, Spain
| | | | - Emmanuel L Gautier
- Institut National de la Santé et de la Recherche Médicale (Inserm, UMR_S 1166), Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Susana Usieto
- Department of Cells and Tissues, Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, C/ Baldiri Reixac 10-15, 08028, Barcelona, Spain
| | - Elena Rebollo
- Molecular Imaging Platform, Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, C/ Baldiri Reixac 10-15, 08028, Barcelona, Spain
| | - Mònica Vila-Ubach
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Déu, Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Angel M Carcaboso
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Déu, Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Biola M Javierre
- Josep Carreras Leukaemia Research Institute, Badalona, Barcelona, Spain
| | - Toni Celià-Terrassa
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), 08003, Barcelona, Spain
| | - Cinzia Lavarino
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Déu, Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Elisa Martí
- Department of Cells and Tissues, Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, C/ Baldiri Reixac 10-15, 08028, Barcelona, Spain
| | - Gwenvael Le Dréau
- Department of Cells and Tissues, Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, C/ Baldiri Reixac 10-15, 08028, Barcelona, Spain.
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France.
| |
Collapse
|
38
|
Finkelstein SR, Patel R, Deland K, Mercer J, Starr B, Zhu D, Min H, Reinsvold M, Campos LDS, Williams N, Luo L, Ma Y, Neff J, Hoenerhoff M, Moding EJ, Kirsch DG. 56 Fe ion exposure increases the incidence of lung and brain tumors at a similar rate in male and female mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.06.543754. [PMID: 37333373 PMCID: PMC10274718 DOI: 10.1101/2023.06.06.543754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
The main deterrent to long-term space travel is the risk of Radiation Exposure Induced Death (REID). The National Aeronautics and Space Administration (NASA) has adopted Permissible Exposure Levels (PELs) to limit the probability of REID to 3% for the risk of death due to radiation-induced carcinogenesis. The most significant contributor to current REID estimates for astronauts is the risk of lung cancer. Recently updated lung cancer estimates from Japan's atomic bomb survivors showed that the excess relative risk of lung cancer by age 70 is roughly four-fold higher in females compared to males. However, whether sex differences may impact the risk of lung cancer due to exposure to high charge and energy (HZE) radiation is not well studied. Thus, to evaluate the impact of sex differences on the risk of solid cancer development post-HZE radiation exposure, we irradiated Rb fl/fl ; Trp53 fl/+ male and female mice infected with Adeno-Cre with various doses of 320 kVp X-rays or 600 MeV/n 56 Fe ions and monitored them for any radiation-induced malignancies. We observed that lung adenomas/carcinomas and esthesioneuroblastomas (ENBs) were the most common primary malignancies in X-ray and 56 Fe ion-exposed mice, respectively. In addition, 1 Gy 56 Fe ion exposure compared to X-rays led to a significantly higher incidence of lung adenomas/carcinomas (p=0.02) and ENBs (p<0.0001). However, we did not find a significantly higher incidence of any solid malignancies in female mice as compared to male mice, regardless of radiation quality. Furthermore, gene expression analysis of ENBs suggested a distinct gene expression pattern with similar hallmark pathways altered, such as MYC targets and MTORC1 signaling, in X-ray and 56 Fe ion-induced ENBs. Thus, our data revealed that 56 Fe ion exposure significantly accelerated the development of lung adenomas/carcinomas and ENBs compared to X-rays, but the rate of solid malignancies was similar between male and female mice, regardless of radiation quality.
Collapse
|
39
|
Gundem G, Levine MF, Roberts SS, Cheung IY, Medina-Martínez JS, Feng Y, Arango-Ossa JE, Chadoutaud L, Rita M, Asimomitis G, Zhou J, You D, Bouvier N, Spitzer B, Solit DB, Dela Cruz F, LaQuaglia MP, Kushner BH, Modak S, Shukla N, Iacobuzio-Donahue CA, Kung AL, Cheung NKV, Papaemmanuil E. Clonal evolution during metastatic spread in high-risk neuroblastoma. Nat Genet 2023; 55:1022-1033. [PMID: 37169874 PMCID: PMC11481711 DOI: 10.1038/s41588-023-01395-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 04/12/2023] [Indexed: 05/13/2023]
Abstract
Patients with high-risk neuroblastoma generally present with widely metastatic disease and often relapse despite intensive therapy. As most studies to date focused on diagnosis-relapse pairs, our understanding of the genetic and clonal dynamics of metastatic spread and disease progression remain limited. Here, using genomic profiling of 470 sequential and spatially separated samples from 283 patients, we characterize subtype-specific genetic evolutionary trajectories from diagnosis through progression and end-stage metastatic disease. Clonal tracing timed disease initiation to embryogenesis. Continuous acquisition of structural variants at disease-defining loci (MYCN, TERT, MDM2-CDK4) followed by convergent evolution of mutations targeting shared pathways emerged as the predominant feature of progression. At diagnosis metastatic clones were already established at distant sites where they could stay dormant, only to cause relapses years later and spread via metastasis-to-metastasis and polyclonal seeding after therapy.
Collapse
Affiliation(s)
- Gunes Gundem
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Computational Oncology Service, Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Max F Levine
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Computational Oncology Service, Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Stephen S Roberts
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Irene Y Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Juan S Medina-Martínez
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Computational Oncology Service, Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yi Feng
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Juan E Arango-Ossa
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Computational Oncology Service, Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Loic Chadoutaud
- Computational Oncology Service, Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mathieu Rita
- Computational Oncology Service, Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Georgios Asimomitis
- Computational Oncology Service, Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joe Zhou
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Computational Oncology Service, Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daoqi You
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nancy Bouvier
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Barbara Spitzer
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David B Solit
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, New York, NY, USA
| | - Filemon Dela Cruz
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael P LaQuaglia
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Brian H Kushner
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Shakeel Modak
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Neerav Shukla
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Christine A Iacobuzio-Donahue
- The David M. Rubenstein Center for Pancreatic Cancer Research, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrew L Kung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elli Papaemmanuil
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Computational Oncology Service, Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
40
|
Bhavsar SP. Metastasis in neuroblastoma: the MYCN question. Front Oncol 2023; 13:1196861. [PMID: 37274289 PMCID: PMC10233040 DOI: 10.3389/fonc.2023.1196861] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 05/08/2023] [Indexed: 06/06/2023] Open
Abstract
Oncogenic drivers like MYCN in neuroblastoma subsets continues to present a significant challenge owing to its strong correlation with high-risk metastatic disease and poor prognosis. However, only a limited number of MYCN-regulatory proteins associated with tumor initiation and progression have been elucidated. In this minireview, I summarize the recent progress in understanding the functional role of MYCN and its regulatory partners in neuroblastoma metastasis.
Collapse
|
41
|
Rivera Z, Escutia C, Madonna MB, Gupta KH. Biological Insight and Recent Advancement in the Treatment of Neuroblastoma. Int J Mol Sci 2023; 24:ijms24108470. [PMID: 37239815 DOI: 10.3390/ijms24108470] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/26/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
One of the most frequent solid tumors in children is neuroblastoma, which has a variety of clinical behaviors that are mostly influenced by the biology of the tumor. Unique characteristics of neuroblastoma includes its early age of onset, its propensity for spontaneous tumor regression in newborns, and its high prevalence of metastatic disease at diagnosis in individuals older than 1 year of age. Immunotherapeutic techniques have been added to the previously enlisted chemotherapeutic treatments as therapeutic choices. A groundbreaking new treatment for hematological malignancies is adoptive cell therapy, specifically chimeric antigen receptor (CAR) T cell therapy. However, due to the immunosuppressive nature of the tumor microenvironment (TME) of neuroblastoma tumor, this treatment approach faces difficulties. Numerous tumor-associated genes and antigens, including the MYCN proto-oncogene (MYCN) and disialoganglioside (GD2) surface antigen, have been found by the molecular analysis of neuroblastoma cells. The MYCN gene and GD2 are two of the most useful immunotherapy findings for neuroblastoma. The tumor cells devise numerous methods to evade immune identification or modify the activity of immune cells. In addition to addressing the difficulties and potential advancements of immunotherapies for neuroblastoma, this review attempts to identify important immunological actors and biological pathways involved in the dynamic interaction between the TME and immune system.
Collapse
Affiliation(s)
- Zoriamin Rivera
- Division of Pediatric Surgery, Department of Surgery, Rush University Medical Center, Chicago, IL 60612, USA
| | - Carlos Escutia
- Division of Pediatric Surgery, Department of Surgery, Rush University Medical Center, Chicago, IL 60612, USA
| | - Mary Beth Madonna
- Division of Pediatric Surgery, Department of Surgery, Rush University Medical Center, Chicago, IL 60612, USA
| | - Kajal H Gupta
- Division of Pediatric Surgery, Department of Surgery, Rush University Medical Center, Chicago, IL 60612, USA
- Division of Surgical Oncology, Department of Surgery, Rush University Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
42
|
Hartley MJ, Gounder P, Oliphant H. Spontaneous periocular ecchymosis: a major review. Orbit 2023; 42:124-129. [PMID: 36374198 DOI: 10.1080/01676830.2022.2142944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Periocular ecchymosis, or periocular bruising, is a common clinical finding. Periocular skin is thin with an extensive vascular network, making this anatomical region prone to bruising. The most common etiology is trauma, but rarely, patients can present with spontaneous periocular ecchymosis (SPE). The pathophysiology of SPE is complex and varied. In this literature review of 121 articles, we assessed the frequency and variety of causation of this infrequent entity. The main finding was that by far the most common diagnosis causing SPE is amyloidosis and neoplasm, most notably neuroblastoma. Amyloidosis accounted for 23% articles (28/121) and neuroblastoma for 17% articles (21/121). Overall, neoplastic processes accounted for 30% of the articles (36/121), raised intracranial pressure and vascular malformations for 19% of the articles (23/121), migraine and atypical headache for 7% of the articles (8/121), while iatrogenic accounted for 5% of the articles (6/121). Through exploration and appreciation of the pathophysiology, we hope to foster a greater understanding in the clinician to establish underlying etiology, from benign to life-threatening, when presented with SPE.
Collapse
Affiliation(s)
- Matthew J Hartley
- Newcastle Eye Centre, Royal Victoria Infirmary, Newcastle-Upon-Tyne, UK
| | - Pav Gounder
- Royal Sussex County Hospital, Sussex Eye Hospital, Brighton, UK
| | - Huw Oliphant
- Royal Sussex County Hospital, Sussex Eye Hospital, Brighton, UK
| |
Collapse
|
43
|
Lynch JD, Tomboc PJ. Neuroblastoma Masquerading as a Septic Hip Infection in a Three-Year-Old. Cureus 2023; 15:e36350. [PMID: 37082496 PMCID: PMC10111876 DOI: 10.7759/cureus.36350] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2023] [Indexed: 03/21/2023] Open
Abstract
Metastatic neuroblastoma to the bone and septic joint shares the same incidence in age and clinical symptomology. Here we discuss a three-year-old male who presented with anemia, persistent hip pain, and a refusal to bear weight. A thorough evaluation based on a broad differential diagnosis allowed for an expedient diagnosis of metastatic neuroblastoma. The timely diagnosis allowed for rapid enrolment in a children's oncology group (COG) clinical trial for advanced neuroblastoma. The patient tolerated the therapy without adverse events and remains in remission.
Collapse
|
44
|
Jahangiri L. Metastasis in Neuroblastoma and Its Link to Autophagy. Life (Basel) 2023; 13:life13030818. [PMID: 36983973 PMCID: PMC10056181 DOI: 10.3390/life13030818] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Neuroblastoma is a paediatric malignancy originating from the neural crest that commonly occurs in the abdomen and adrenal gland, leading to cancer-related deaths in children. Distant metastasis can be encountered at diagnosis in greater than half of these neuroblastoma patients. Autophagy, a self-degradative process, plays a key role in stress-related responses and the survival of cells and has been studied in neuroblastoma. Accordingly, in the early stages of metastasis, autophagy may suppress cancer cell invasion and migration, while its role may be reversed in later stages, and it may facilitate metastasis by enhancing cancer cell survival. To that end, a body of literature has revealed the mechanistic link between autophagy and metastasis in neuroblastoma in multiple steps of the metastatic cascade, including cancer cell invasion and migration, anoikis resistance, cancer cell dormancy, micrometastasis, and metastatic outbreak. This review aims to take a step forward and discuss the significance of multiple molecular players and compounds that may link autophagy to metastasis and map their function to various metastatic steps in neuroblastoma.
Collapse
Affiliation(s)
- Leila Jahangiri
- School of Science and Technology, Nottingham Trent University, Clifton Site, Nottingham NG11 8NS, UK
- Division of Cellular and Molecular Pathology, Department of Pathology, Addenbrooke's Hospital, University of Cambridge, Cambridge CB2 0QQ, UK
| |
Collapse
|
45
|
Kuchalska K, Derwich K, Gotz-Więckowska A. Ophthalmologic Manifestations of Neuroblastoma: A Systemic Review. J Pediatr Hematol Oncol 2023; 45:47-56. [PMID: 36161977 DOI: 10.1097/mph.0000000000002543] [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] [Received: 05/19/2022] [Accepted: 07/21/2022] [Indexed: 11/26/2022]
Abstract
Neuroblastoma (NBL) is the most common extracranial solid tumor found in pediatric patients. It develops from the sympathetic tract tissue. Although the symptoms are associated with tumor localization, sometimes NBL is manifested as ophthalmologic disorders. In this paper, we describe their incidence and the correlation with the prognosis. We searched 2 databases (PubMed and Web of Science) for papers published before April 2022, and concerned pediatric patients with NBL, which caused ophthalmologic changes. We collected 7 papers assessing the occurrence of ophthalmologic findings in the NBL patients, as well as 68 case reports presenting children with orbital changes and NBL, or with other tumors stemming from the sympathetic ganglia. The statistical analysis was performed to synthetize the data. The ophthalmologic signs occurred in 17.89% of the NBL cases; however, they were observed on presentation in 10.68%. The isolated ocular presentation was found in 2.56% of patients, whereas Horner syndrome was most frequent. The ophthalmologic symptoms correlated with patients' age, primary tumor site, and survival rate. NBL may be challenging to diagnose in cases with isolated ophthalmologic manifestations. Numerous possible ocular signs can be observed, which emphasize the need for multidisciplinary care with regard to the NBL patients.
Collapse
|
46
|
Tringale KR, Wolden SL, Casey DL, Kushner BH, Luo L, Pandit-Taskar N, Souweidane M, Cheung NKV, Modak S, Basu EM, Kramer K. Clinical outcomes of pediatric patients receiving multimodality treatment of second central nervous system relapse of neuroblastoma. Pediatr Blood Cancer 2023; 70:e30075. [PMID: 36349892 DOI: 10.1002/pbc.30075] [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: 03/09/2022] [Revised: 09/09/2022] [Accepted: 10/09/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND In high-risk neuroblastoma, multimodality therapy including craniospinal irradiation (CSI) is effective for central nervous system (CNS) relapse. Management of post-CSI CNS relapse is not clearly defined. PROCEDURE Pediatric patients with neuroblastoma treated with CSI between 2000 and 2019 were identified. Treatment of initial CNS disease (e.g., CSI, intraventricular compartmental radioimmunotherapy [cRIT] with 131 I-monoclonal antibodies targeting GD2 or B7H3) and management of post-CSI CNS relapse ("second CNS relapse") were characterized. Cox proportional hazards models to evaluate factors associated with third CNS relapse and overall survival (OS) were used. RESULTS Of 128 patients (65% male, median age 4 years), 19 (15%) received CSI with protons and 115 (90%) had a boost. Most (103, 81%) received cRIT, associated with improved OS (hazard ratio [HR] 0.3, 95% confidence interval [CI]: 0.1-0.5, p < .001). Forty (31%) developed a second CNS relapse, associated with worse OS (1-year OS 32.5%, 95% CI: 19-47; HR 3.8; 95% CI: 2.4-6.0, p < .001), and more likely if the leptomeninges were initially involved (HR 2.5, 95% CI: 1.3-4.9, p = .006). Median time to second CNS relapse was 6.8 months and 51% occurred outside the CSI boost field. Twenty-five (63%) patients underwent reirradiation, most peri-operatively (18, 45%) with focal hypofractionation. Eight (20%) patients with second CNS relapse received cRIT, associated with improved OS (HR 0.1; 95% CI: 0.1-0.4, p < .001). CONCLUSIONS CNS relapse after CSI for neuroblastoma portends a poor prognosis. Surgery with hypofractionated radiotherapy was the most common treatment. Acknowledging the potential for selection bias, receipt of cRIT both at first and second CNS relapse was associated with improved survival. This finding necessitates further investigation.
Collapse
Affiliation(s)
- Kathryn R Tringale
- Department of Radiation Oncology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Suzanne L Wolden
- Department of Radiation Oncology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Dana L Casey
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Brian H Kushner
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Leo Luo
- Department of Radiation Oncology, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee, USA
| | - Neeta Pandit-Taskar
- Department of Nuclear Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Mark Souweidane
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Shakeel Modak
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ellen M Basu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Kim Kramer
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
47
|
Central Nervous System Metastasis in Neuroblastoma: From Three Decades Clinical Experience to New Considerations in the Immunotherapy Era. Cancers (Basel) 2022; 14:cancers14246249. [PMID: 36551734 PMCID: PMC9777007 DOI: 10.3390/cancers14246249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/15/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Central nervous system (CNS) metastatic spread in neuroblastoma (NB) is rare and occurs more often at relapse/progression. We report on CNS involvement in high risk (HR) NB over 25 years. For this retrospective study, we reviewed the CNS imaging of all the patients treated at Bambino Gesù Children Hospital from 1 July 1996 to 30 June 2022. A total of 128 patients with HR NB were diagnosed over 26 years. Out of 128 patients, CNS metastatic spread occurred in 6 patients: 3 patients presented a metastatic spread at diagnosis, while in 3 patients, CNS was involved at relapse. Overall, the rate of occurrence of CNS spread is 4.7% with the same distribution at diagnosis and at relapse, namely 2.3%. Interestingly, CNS spread at diagnosis was observed only before 2012, whereas CNS was observed at relapse only after 2012, in the immunotherapy era. CNS metastases presented similar imaging features at diagnosis and at relapse, with a peculiar hemorrhagic aspect and mainly hemispheric localization in patients with bone skull involvement at the time of diagnosis. The outcome is dismal, and 3 out of 6 patients died for progressive disease.
Collapse
|
48
|
Pastorino F, Capasso M, Brignole C, Giglio S, Bensa V, Cantalupo S, Lasorsa VA, Tondo A, Mura R, Sementa AR, Garaventa A, Ponzoni M, Amoroso L. Italian Precision Medicine in Pediatric Oncology: Moving beyond Actionable Alterations. Int J Mol Sci 2022; 23:ijms231911236. [PMID: 36232538 PMCID: PMC9570321 DOI: 10.3390/ijms231911236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Neuroblastoma (NB) is the most common extracranial solid tumor encountered in childhood. Although there has been significant improvement in the outcomes of patients with high-risk disease, the prognosis for patients with metastatic relapse or refractory disease is poor. Hence, the clinical integration of genome sequencing into standard clinical practice is necessary in order to develop personalized therapy for children with relapsed or refractory disease. The PeRsonalizEdMEdicine (PREME) project focuses on the design of innovative therapeutic strategies for patients suffering from relapsed NB. We performed whole exome sequencing (WES) of patient-matched tumor-normal samples to identify genetic variants amenable to precision medicine. Specifically, two patients were studied (First case: a three-year-old male with early relapsed NB; Second case: a 20-year-old male who relapsed 10 years after the first diagnosis of NB). Results were reviewed by a multi-disciplinary molecular tumor board (MTB) and clinical reports were issued to the ordering physician. WES revealed the mutation c.G320C in the CUL4A gene in case 1 and the mutation c.A484G in the PSMC2 gene in case 2. Both patients were treated according to these actionable alterations, with promising results. The effective treatment of NB is one of the main challenges in pediatric oncology. In the era of precision medicine, the need to design new therapeutic strategies for NB is fundamental. Our results demonstrate the feasibility of incorporating clinical WES into pediatric oncology practice.
Collapse
Affiliation(s)
- Fabio Pastorino
- Laboratorio di Terapie Sperimentali in Oncologia, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
- These authors contributed equally to this work
| | - Mario Capasso
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80131 Napoli, Italy
- CEINGE Biotecnologie Avanzate, 80131 Napoli, Italy
- These authors contributed equally to this work
| | - Chiara Brignole
- Laboratorio di Terapie Sperimentali in Oncologia, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Serena Giglio
- UOC Oncologia, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Veronica Bensa
- Laboratorio di Terapie Sperimentali in Oncologia, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Sueva Cantalupo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80131 Napoli, Italy
- CEINGE Biotecnologie Avanzate, 80131 Napoli, Italy
| | | | - Annalisa Tondo
- Dipartimento di Oncoematologia, Ospedale Meyer, 50139 Firenze, Italy
| | - Rossella Mura
- Oncoematologia Pediatrica, Ospedale Pediatrico Microcitemico “Antonio Cao” Azienda Ospedaliera Brotzu, 09121 Cagliari, Italy
| | - Angela Rita Sementa
- Dipartimento di Patologia, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | | | - Mirco Ponzoni
- Laboratorio di Terapie Sperimentali in Oncologia, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
- Correspondence: ; Tel.: +39-0105-636-3539; Fax: +39-0103-779-820
| | - Loredana Amoroso
- UOC Oncologia, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
- These authors contributed equally to this work
| |
Collapse
|
49
|
Abstract
Neuroblastomas are tumours of sympathetic origin, with a heterogeneous clinical course ranging from localized or spontaneously regressing to widely metastatic disease. Neuroblastomas recapitulate many of the features of sympathoadrenal development, which have been directly targeted to improve the survival outcomes in patients with high-risk disease. Over the past few decades, improvements in the 5-year survival of patients with metastatic neuroblastomas, from <20% to >50%, have resulted from clinical trials incorporating high-dose chemotherapy with autologous stem cell transplantation, differentiating agents and immunotherapy with anti-GD2 monoclonal antibodies. The next generation of trials are designed to improve the initial response rates in patients with high-risk neuroblastomas via the addition of immunotherapies, targeted therapies (such as ALK inhibitors) and radiopharmaceuticals to standard induction regimens. Other trials are focused on testing precision medicine strategies for patients with relapsed and/or refractory disease, enhancing the antitumour immune response and improving the effectiveness of maintenance regimens, in order to prolong disease remission. In this Review, we describe advances in delineating the pathogenesis of neuroblastoma and in identifying the drivers of high-risk disease. We then discuss how this knowledge has informed improvements in risk stratification, risk-adapted therapy and the development of novel therapies.
Collapse
Affiliation(s)
- Bo Qiu
- Department of Paediatrics, Division of Paediatric Hematology and Oncology, University of California San Francisco, San Francisco, CA, USA.
| | - Katherine K Matthay
- Department of Paediatrics, Division of Paediatric Hematology and Oncology, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
50
|
Amplification of CDK4 and MDM2: a detailed study of a high-risk neuroblastoma subgroup. Sci Rep 2022; 12:12420. [PMID: 35859155 PMCID: PMC9300649 DOI: 10.1038/s41598-022-16455-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 07/11/2022] [Indexed: 11/08/2022] Open
Abstract
In neuroblastoma, MYCN amplification and 11q-deletion are important, although incomplete, markers of high-risk disease. It is therefore relevant to characterize additional alterations that can function as prognostic and/or predictive markers. Using SNP-microarrays, a group of neuroblastoma patients showing amplification of one or multiple 12q loci was identified. Two loci containing CDK4 and MDM2 were commonly co-amplified, although amplification of either locus in the absence of the other was observed. Pharmacological inhibition of CDK4/6 with ribociclib or abemaciclib decreased proliferation in a broad set of neuroblastoma cell lines, including CDK4/MDM2-amplified, whereas MDM2 inhibition by Nutlin-3a was only effective in p53wild-type cells. Combined CDK4/MDM2 targeting had an additive effect in p53wild-type cell lines, while no or negative additive effect was observed in p53mutated cells. Most 12q-amplified primary tumors were of abdominal origin, including those of intrarenal origin initially suspected of being Wilms' tumor. An atypical metastatic pattern was also observed with low degree of bone marrow involvement, favoring other sites such as the lungs. Here we present detailed biological data of an aggressive neuroblastoma subgroup hallmarked by 12q amplification and atypical clinical presentation for which our in vitro studies indicate that CDK4 and/or MDM2 inhibition also could be beneficial.
Collapse
|