1
|
Bordbar F, Rigi A, Mastanabad MV, Rohani F, Ghaedi E, Dhiaa SM, Asadi F, Maragheh SM. Investigating miR-9 and miR-222 in CSF and Plasma of Neuroblastoma Patients as Metastatic and Apoptotic-Related Markers. Cell Biochem Biophys 2025; 83:1605-1615. [PMID: 39663279 DOI: 10.1007/s12013-024-01570-9] [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] [Accepted: 09/18/2024] [Indexed: 12/13/2024]
Abstract
Neuroblastoma is a cancer that occurs due to abnormal development of the sympathetic nervous system. The dysregulation of miR-9 and miR-222 plays a crucial role in neuroblastoma development. These microRNAs have a significant relationship with PTEN, caspase-9, and MMP14, which can potentially form the basis for the specific diagnosis and treatment of this disease. In our study, two neuroblastoma cell lines were divided into three groups based on whether they had been treated with miR-9, anti-miR-9, miR-222, or both. We evaluated various parameters in these groups, including migration (through a wound healing assay), apoptosis (using flow cytometry), and gene expression (through qRT-PCR). Additionally, we measured the expression levels of MMP14, miR-9, and miR-222 in plasma and CSF samples from neuroblastoma patients using ELISA and qRT-PCR. We found that patients with neuroblastoma had higher levels of MMP14 and miR-222 mRNA expression but lower levels of miR-9 mRNA expression. Furthermore, after treating the cell lines with anti-miR-9 and anti-miR-222, we observed increased levels of MMP14 expression, as well as PTEN and caspase-9. Additionally, the treatment with anti-miR-222 and anti-miR-9 led to an increase in the frequency of apoptosis and migration of cancer cells. Our research shows that the dysregulation of miR-9, miR-222, and MMP14 could be key indicators in the pathogenesis of neuroblastoma. We also found that up-regulation of miR-9 was associated with decreased disease severity, whereas up-regulation of miR-222 and MMP14 was linked to increased disease severity.
Collapse
Affiliation(s)
- Farhad Bordbar
- Key Laboratory of Chicken Genetics, Breeding And Reproduction, Ministry of Agriculture And Rural Affair, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Amir Rigi
- Department of Nursing, Young Researchers and Elite Club, Zahedan Branch, Islamic Azad University, Zahedan, Iran
| | - Mahsa Vafaei Mastanabad
- Neurosurgery Department, Faculty of Medicine, Qazvin University of Medical Science, Qazvin, Iran
| | - Fattah Rohani
- Faculty of Veterinary Medicine of Shahrekord, Shahrekord, Iran
| | - Elham Ghaedi
- Department of Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | | | - Fatemeh Asadi
- Department of Genetics, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| | - Salar Momen Maragheh
- Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran.
- Department of Biotechnology, Islamic Azad University, Central Tehran Branch, Tehran, Iran.
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Magalhães Gimenez T, Peralta VP, Giorgi RR, Morikawa K, Vince CC, Halley N, Siqueira SA, Bendit I, Cristofani LM, Filho VO, Novak EM. Novel variants of the ATRX gene identified in MYCN non-amplified Neuroblastoma in Brazilian patients. Clinics (Sao Paulo) 2025; 80:100652. [PMID: 40286729 PMCID: PMC12060459 DOI: 10.1016/j.clinsp.2025.100652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 03/14/2025] [Accepted: 04/02/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Neuroblastoma is one of the most common extracranial solid tumors in children and it frequently displays high heterogeneity throughout the course of the disease. It has previously been described those changes in the ATRX gene (Alpha Thalassemia/Mental Retardation, X-linked) are the most common recurring events in the indolent clinical subtype (∼30 %) of MYCN amplified neuroblastoma. There is no effective treatment for this type of neuroblastoma, which is associated with overall poor survival. On the other hand, few studies have detected an association between high-risk (stage IV) non-amplified MYCN neuroblastoma patients and mutant ATRX. METHODS In this study, 37 tumor samples from Brazilian patients with stages I to IV MYCN non-amplified neuroblastoma, according to the International Neuroblastoma Staging System (INSS), were analyzed using the panel Oncomine™ Childhood Cancer Research Assay. RESULTS The authors found two older children (NB1 and NB2) with advanced MYCN non-amplified neuroblastoma carried each one of the two following novel nonsense ATRX variants (p.Gln1670* or p.Glu1984*). These variants created a stop codon in the helicase domain of the ATRX gene, leading to ATRX loss-of-function. These mutations were confirmed by Sanger sequencing and the protein loss-of-function was confirmed by immunohistochemistry. The finding of these heterozygous mutations in two patients with MYCN non-amplified neuroblastoma deserves further investigation. Thus, the authors analyzed each of these cases to better understand how these mutations may be related to disease severity and prognosis. CONCLUSION ATRX loss-of-function from p.Gln1670* or p.Glu1984* mutations turn MYCN non-amplified neuroblastoma more aggressive and similar to what is seen in MYCN amplified neuroblastoma. This information may help clinical decision-making and facilitate establishing an accurate prognosis for patients with MYCN non-amplified neuroblastoma.
Collapse
Affiliation(s)
| | - Vanessa Pretes Peralta
- Laboratório de Investigação Médica em Pediatria Clínica -Lim-36.Instituto da Criança. Hospital de Clínicas, Faculdade de Medicina, Universidade de São Paulo (HC/FMUSP), São Paulo, Brazil
| | - Ricardo Rodrigues Giorgi
- Laboratório de Investigação Médica em Patogênese e Terapia dirigida em Onco-Imuno-Hematologia (Lim 31). Departamento de Hematologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP). São Paulo, São Paulo, Brazil
| | - Karina Morikawa
- Instituto do Cancer do Estado de Sao Paulo (ICESP/ITACI), São Paulo, Brazil
| | | | - Nathalia Halley
- Hospital Israelita Albert Einstein (HIAE). São Paulo, Brazil
| | - Sheila Aparecida Siqueira
- Divisão de Patologia, Hospital das Clínicas (HCFMUSP), Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Israel Bendit
- Laboratório de Investigação Médica em Patogênese e Terapia dirigida em Onco-Imuno-Hematologia (Lim 31). Departamento de Hematologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP). São Paulo, São Paulo, Brazil
| | | | - Vicente Odone Filho
- Departamento de Pediatria, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Estela Maria Novak
- Fundação Pró-Sangue Hemocentro de São Paulo, Departamento de Genética Molecular e Biotecnologia. São Paulo, Brazil.
| |
Collapse
|
5
|
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
|
6
|
Mossé YP, Polkosnik G, Pogoriler J, Mattei P, States LJ, Maris JM. Anaplastic Lymphoma Kinase Inhibition Therapy for Hereditary Neuroblastoma. JCO Precis Oncol 2025; 9:e2400886. [PMID: 40294355 PMCID: PMC12039971 DOI: 10.1200/po-24-00886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 03/10/2025] [Accepted: 04/01/2025] [Indexed: 04/30/2025] Open
Affiliation(s)
- Yaël P. Mossé
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Grace Polkosnik
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Jenny Pogoriler
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Peter Mattei
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Lisa J. States
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA
| | - John M. Maris
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
7
|
Li S, Wang J, Zhang Z, Ren C, He D. Individual risk and prognostic value prediction by interpretable machine learning for distant metastasis in neuroblastoma: A population-based study and an external validation. Int J Med Inform 2025; 196:105813. [PMID: 39904180 DOI: 10.1016/j.ijmedinf.2025.105813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 12/27/2024] [Accepted: 01/23/2025] [Indexed: 02/06/2025]
Abstract
PURPOSE Neuroblastoma (NB) is a childhood malignancy with a poor prognosis and a propensity for distant metastasis (DM). We aimed to establish machine learning (ML) based model to accurately predict risk of DM and prognosis of NB patients with DM. METHODS We analyzed NB patients from the Surveillance, Epidemiology, and End Results (SEER) database between 2000 and 2020. Univariate and multivariate logistic analysis were employed to select meaning variables. Recursive Feature Elimination (RFE) method based on 6 ML algorithms was utilized in feature selection. To construct predictive model, 13 ML algorithms were evaluated by area under the operating characteristic curve (AUC), accuracy, sensitivity, specificity, precision, cross-entropy, Brier scores, Balanced Accuracy and F-beta score. An optimal ML model was constructed to predict DM, and the predictive results were explained by SHapley Additive exPlanations (SHAP) framework. Meanwhile, 101 ML algorithm combinations were developed to select the best model with highest C-index to predict prognosis of NB patients with DM. RESULTS A total of 1,668 NB patients from SEER database was consecutively enrolled. We identified that tumor primary site, grade, surgery type, regional lymph nodes, radiotherapy and chemotherapy are significant risk factors for DM. CatBoost model was selected as the best prediction model, and AUC was 0.846 (95 %CI: [0.804,0.899]), 0.834 (95 %CI: [0.796,0.873]) and 0.813 (95 %CI: [0.776,0.852]) in training, internal test and external test sets, with 0.777 accuracy, 0.839 sensitivity, 0.72 specificity and 0.731 precision in training set. Grade, chemotherapy and radiotherapy had the greatest effects on DM according to SHAP results. For prognosis prediction, "RSF + GBM" algorithm was the best prognostic model with C-index of 0.656, 0.611 and 0.629 in training, internal test and external test sets. CONCLUSIONS Our ML models demonstrate excellent accuracy and reliability, offering more precise personalized metastasis diagnosis and prognostic prediction to NB patients.
Collapse
Affiliation(s)
- Shan Li
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, China; China International Science and Technology Cooperation base of Child Development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Jinkui Wang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, China; China International Science and Technology Cooperation base of Child Development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Zhaoxia Zhang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, China; China International Science and Technology Cooperation base of Child Development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Chunnian Ren
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, China; China International Science and Technology Cooperation base of Child Development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Dawei He
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, China; China International Science and Technology Cooperation base of Child Development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing 400014, China.
| |
Collapse
|
8
|
Williams KM, Shah NR, Chukkapalli S, King S, Grant CN, Brown EG, Avanzini S, Lal DR, Sarnacki S, Newman EA. Modern surgical strategies in pediatric neuroblastoma: Evolving approaches and treatment principles. Pediatr Blood Cancer 2025; 72 Suppl 2:e31317. [PMID: 39313754 DOI: 10.1002/pbc.31317] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/25/2024]
Abstract
Neuroblastoma, the most common extracranial solid tumor in children under the age of 5, has been described as early as the 19th century, and its complexity has continued to intrigue researchers, as well as medical and surgical specialists. At one end of the phenotypic spectrum, neuroblastoma is self-limiting with minimal to no intervention required, while on the opposite end exists the challenge of refractory disease despite aggressive management and toxic systemic treatments. The goal of this review is to describe a comprehensive surgical perspective and contemporary approach to neuroblastoma.
Collapse
Affiliation(s)
- Keyonna M Williams
- Section of Pediatric Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Nikhil R Shah
- Section of Pediatric Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Sahiti Chukkapalli
- Section of Pediatric Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Sarah King
- Section of Pediatric Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Christa N Grant
- Department of Pediatric Surgery, Maria Fareri Children's Hospital, Valhalla, New York, USA
| | - Erin G Brown
- Division of Pediatric Surgery, University of California Davis Medical Center, Sacramento, California, USA
| | - Stefano Avanzini
- Department of Pediatric Surgery, IRCCS Istituto G. Gaslini, Genoa, Italy
| | - Dave R Lal
- Department of Pediatric Surgery, Children's Wisconsin, Milwaukee, Wisconsin, USA
| | - Sabine Sarnacki
- Department of Pediatric Surgery, Hôpital Universitaire Necker, Paris, France
| | - Erika A Newman
- Section of Pediatric Surgery, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
9
|
Yu X, Kang S, Ge J, Wang J. A clinical observational study of dinutuximab beta as first-line maintenance treatment for patients with high-risk neuroblastoma in China. BMC Pediatr 2025; 25:203. [PMID: 40091019 PMCID: PMC11912632 DOI: 10.1186/s12887-025-05568-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND High-risk neuroblastoma (HR-NB) is associated with high metastatic and relapse rates that require intensive multimodal treatment. We evaluated the efficacy and safety of dinutuximab beta as first-line maintenance immunotherapy in pediatric patients with HR-NB in real-world clinical settings in China. METHODS We retrospectively reviewed the clinical records of pediatric patients with newly diagnosed HR-NB in the hospital from October 2021 to November 2023. Patients treated with dinutuximab beta in combination with granulocyte-macrophage colony-stimulating factor (GM-CSF) and isotretinoin as the first-line maintenance therapy were included in this study. Among patients with residual disease after completing induction and consolidation treatment, those with partial response (PR) or very good partial response (VGPR) except for bone marrow (BM) residue were also administrated vincristine/irinotecan/temozolomide (VIT) chemotherapy. RESULTS Fifty-one patients with newly diagnosed HR-NB who achieved at least PR before immunotherapy were evaluated. At the end of immunotherapy, the objective response rate (ORR) in 33 patients with evidence of disease was 60.6% (95% confidence interval (CI), 42.1-77.1%) and the complete response rate (CRR; n = 18) was 54.5% (95% CI, 36.4-71.9%). The 2-year event-free survival (EFS) rate and overall survival (OS) rate were 80.1% (95% CI, 66.2-88.8%) and 97.6% (95% CI, 84.3-99.7%), respectively. The 2-year EFS rate was higher in patients with CR (94.4%; 95% CI, 66.6-99.2%) than in non-CR patients (72.6%; 95% CI, 53.9-84.7%). Dinutuximab beta was well tolerated in patients and had fewer side effects, which decreased over time. Co-treatment of dinutuximab beta with VIT chemotherapy did not require discontinuation in patients undergoing immunochemotherapy. CONCLUSION The study showed promising efficacy and safety of dinutuximab beta as the first-line maintenance immunotherapy for pediatric patients with HR-NB. Notably, the combination of dinutuximab beta with GM-CSF and VIT chemotherapy could be used for treating patients who did not achieve CR after previous multimodal therapy.
Collapse
Affiliation(s)
- Xuedi Yu
- Department of Pediatric Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250000, China
| | - Suyi Kang
- Department of Pediatric Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250000, China
| | - Junjie Ge
- Department of Pediatric Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250000, China
| | - Jingfu Wang
- Department of Pediatric Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250000, China.
| |
Collapse
|
10
|
Yin H, Liu T, Wu D, Li X, Li G, Song W, Wang X, Xin S, Liu Y, Pan J. Exploring FAM13A-N-Myc interactions to uncover potential targets in MYCN-amplified neuroblastoma: a study of protein interactions and molecular dynamics simulations. BMC Cancer 2025; 25:470. [PMID: 40087586 PMCID: PMC11907995 DOI: 10.1186/s12885-025-13903-9] [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: 11/14/2024] [Accepted: 03/10/2025] [Indexed: 03/17/2025] Open
Abstract
Neuroblastoma (NB), a common infantile neuroendocrine tumor, presents a substantial therapeutic challenge when MYCN is amplified. Given that the protein structure of N-Myc is disordered, we utilized Alphafold for prediction and GROMACS for optimization of the N-Myc structure, thereby improving the reliability of the predicted structure. The publicly available datasets GSE49710 and GSE73517 were adopted, which contain the transcriptome data of clinical samples from 598 NB patients. Through various machine learning algorithms, FAM13A was identified as a characteristic gene of MYCN. Cell functional experiments, including those on cell proliferation, apoptosis, and cell cycle, also indicate that FAM13A is a potential risk factor. Additionally, Alphafold and GROMACS were employed to predict and optimize the structure of FAM13A. Protein-protein docking and molecular dynamic modeling techniques were then used to validate the enhanced protein stability resulting from the interaction between N-Myc and FAM13A. Consequently, targeting FAM13A holds the potential to reduce the stability of N-Myc, hinder the proliferation of NB cells, and increase the infiltration of immune cells. This multi-faceted approach effectively combats tumor cells, making FAM13A a prospective therapeutic target for MYCN-amplified NB.
Collapse
Affiliation(s)
- Hongli Yin
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215003, China.
| | - Tianyi Liu
- Department of Pharmaceutics, Dalian Women and Children's Medical Group, Dalian, Liaoning, 116012, China
| | - Di Wu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Xiaolu Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Gen Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Weiwei Song
- Department of Pharmacy, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Xiaodong Wang
- Department of Orthopaedics, Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Shan Xin
- Genetics and Cellular Engineering Group, Research Unit Signaling and Translation, Helmholtz Zentrum Munich, Ingolstaedter Landstr. 1, Neuherberg, 85764, Germany.
| | - Yisu Liu
- China College of Life Sciences and Chemistry, Hunan University of Technology, Zhuzhou, 412007, China.
| | - Jian Pan
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215003, China.
| |
Collapse
|
11
|
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
|
12
|
Westerveld ASR, Tytgat GAM, van Santen HM, van Noesel MM, Loonen J, de Vries ACH, Louwerens M, Koopman MMW, van der Heiden-van der Loo M, Janssens GO, de Krijger RR, Ronckers CM, van der Pal HJH, Kremer LCM, Teepen JC. Long-Term Risk of Subsequent Neoplasms in 5-Year Survivors of Childhood Neuroblastoma: A Dutch Childhood Cancer Survivor Study-LATER 3 Study. J Clin Oncol 2025; 43:154-166. [PMID: 39356982 DOI: 10.1200/jco.23.01430] [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: 07/06/2023] [Revised: 05/16/2024] [Accepted: 08/15/2024] [Indexed: 10/04/2024] Open
Abstract
PURPOSE Neuroblastoma survivors have an increased risk of developing subsequent malignant neoplasms (SMNs), but the risk of subsequent nonmalignant neoplasms (SNMNs) and risk factors are largely unknown. We analyzed the long-term risks and associated risk factors for developing SMNs and SNMNs in a well-characterized cohort of 5-year neuroblastoma survivors. METHODS We included 563 5-year neuroblastoma survivors from the Dutch Childhood Cancer Survivor Study (DCCSS)-LATER cohort, diagnosed during 1963-2014. Subsequent neoplasms were ascertained by linkages with the Netherlands Cancer Registry and the Dutch Nationwide Pathology Databank (Palga) and medical chart review. We calculated standardized incidence ratios (SIRs), absolute excess risk (AER), and cumulative incidences. Multivariable competing risk regression analysis was used to evaluate risk factors. RESULTS In total, 23 survivors developed an SMN and 60 an SNMN. After a median follow-up of 23.7 (range, 5.0-56.3) years, the risk of SMN was elevated compared with the general population (SIR, 4.0; 95% CI, 2.5 to 5.9; AER per 10,000 person-years, 15.1). The 30-year cumulative incidence was 3.4% (95% CI, 1.9 to 6.0) for SMNs and 10.4% (95% CI, 7.3 to 14.8) for SNMNs. Six survivors developed an SMN after iodine-metaiodobenzylguanidine (131IMIBG) treatment. Survivors treated with 131IMIBG had a higher risk of developing SMNs (subdistribution hazard ratio [SHR], 5.7; 95% CI, 1.8 to 17.8) and SNMNs (SHR, 2.6; 95% CI, 1.2 to 5.6) compared with survivors treated without 131IMIBG; results for SMNs were attenuated in high-risk patients only (SMNs SHR, 3.6; 95% CI, 0.9 to 15.3; SNMNs SHR, 1.5; 95% CI, 0.7 to 3.6). CONCLUSION Our results demonstrate that neuroblastoma survivors have an elevated risk of developing SMNs and a high risk of SNMNs. 131IMIBG may be a treatment-related risk factor for the development of SMN and SNMN, which needs further validation. Our results emphasize the need for awareness of subsequent neoplasms and the importance of follow-up care.
Collapse
Affiliation(s)
| | | | - Hanneke M van Santen
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Department of Pediatric Endocrinology, University Medical Center Utrecht, Wilhelmina Children's Hospital, Utrecht, the Netherlands
| | - Max M van Noesel
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Department of Imaging & Cancer, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jacqueline Loonen
- Department of Hematology, Radboudumc Center of Expertise for Cancer Survivorship, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Andrica C H de Vries
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Netherlands Department of Pediatric Oncology/Hematology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Marloes Louwerens
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Maria M W Koopman
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | | | - Geert O Janssens
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Ronald R de Krijger
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Cecile M Ronckers
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Division of Childhood Cancer Epidemiology, Institute of Medical Biostatistics Informatics and Epidemiology, University Medical Center of the JGU, Mainz, Germany
| | | | - Leontien C M Kremer
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- University Medical Center Utrecht, Wilhelmina Children's Hospital, Utrecht, the Netherlands
- Department of Pediatrics, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Jop C Teepen
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| |
Collapse
|
13
|
Bilger A, Lambert PF. Rapid-onset cancer. Tumour Virus Res 2025; 19:200312. [PMID: 39755235 PMCID: PMC11764593 DOI: 10.1016/j.tvr.2024.200312] [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: 11/13/2024] [Revised: 12/23/2024] [Accepted: 12/23/2024] [Indexed: 01/06/2025] Open
Abstract
Human cancers are generally thought to develop over the course of decades. Such slow progression is well documented for a variety of cancers that we designate "slow-onset" cancers. "Rapid-onset" cancers, in contrast, can develop in a matter of months in humans or in as little as 9 days in mice. These cancers often develop under conditions that might be expected to accelerate cancer development: early development, immune deficiency, or viral infection. We will discuss rapid-onset cancers in the context of the "hallmarks of cancer" - properties cells must acquire in order to become malignant - focusing on how viruses are particularly well suited to causing rapid-onset cancer.
Collapse
Affiliation(s)
- Andrea Bilger
- McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public Health, 1111 Highland Avenue, Madison, WI, 53705, USA.
| | - Paul F Lambert
- McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public Health, 1111 Highland Avenue, Madison, WI, 53705, USA.
| |
Collapse
|
14
|
Wertheim KY, Chisholm R, Richmond P, Walker D. Multicellular model of neuroblastoma proposes unconventional therapy based on multiple roles of p53. PLoS Comput Biol 2024; 20:e1012648. [PMID: 39715281 PMCID: PMC11723635 DOI: 10.1371/journal.pcbi.1012648] [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: 02/22/2024] [Revised: 01/10/2025] [Accepted: 11/18/2024] [Indexed: 12/25/2024] Open
Abstract
Neuroblastoma is the most common extra-cranial solid tumour in children. Over half of all high-risk cases are expected to succumb to the disease even after chemotherapy, surgery, and immunotherapy. Although the importance of MYCN amplification in this disease is indisputable, the mechanistic details remain enigmatic. Here, we present a multicellular model of neuroblastoma comprising a continuous automaton, discrete cell agents, and a centre-based mechanical model, as well as the simulation results we obtained with it. The continuous automaton represents the tumour microenvironment as a grid-like structure, where each voxel is associated with continuous variables such as the oxygen level therein. Each discrete cell agent is defined by several attributes, including its cell cycle position, mutations, gene expression pattern, and more with behaviours such as cell cycling and cell death being stochastically dependent on these attributes. The centre-based mechanical model represents the properties of these agents as physical objects, describing how they repel each other as soft spheres. By implementing a stochastic simulation algorithm on modern GPUs, we simulated the dynamics of over one million neuroblastoma cells over a period of months. Specifically, we set up 1200 heterogeneous tumours and tracked the MYCN-amplified clone's dynamics in each, revealed the conditions that favour its growth, and tested its responses to 5000 drug combinations. Our results are in agreement with those reported in the literature and add new insights into how the MYCN-amplified clone's reproductive advantage in a tumour, its gene expression profile, the tumour's other clones (with different mutations), and the tumour's microenvironment are inter-related. Based on the results, we formulated a hypothesis, which argues that there are two distinct populations of neuroblastoma cells in the tumour; the p53 protein is pro-survival in one and pro-apoptosis in the other. It follows that alternating between inhibiting MDM2 to restore p53 activity and inhibiting ARF to attenuate p53 activity is a promising, if unorthodox, therapeutic strategy. The multicellular model has the advantages of modularity, high resolution, and scalability, making it a potential foundation for creating digital twins of neuroblastoma patients.
Collapse
Affiliation(s)
- Kenneth Y. Wertheim
- Insigneo Institute for in Silico Medicine, University of Sheffield, Sheffield, United Kingdom
- School of Computer Science, University of Sheffield, Sheffield, United Kingdom
- Centre of Excellence for Data Science, Artificial Intelligence, and Modelling, University of Hull, Kingston upon Hull, United Kingdom
- School of Computer Science, University of Hull, Kingston upon Hull, United Kingdom
| | - Robert Chisholm
- School of Computer Science, University of Sheffield, Sheffield, United Kingdom
| | - Paul Richmond
- School of Computer Science, University of Sheffield, Sheffield, United Kingdom
| | - Dawn Walker
- Insigneo Institute for in Silico Medicine, University of Sheffield, Sheffield, United Kingdom
- School of Computer Science, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
15
|
He J, Wang Z, Yu X, Su Y, Hong M, Zhu K. Promoting application of enhanced recovery after surgery protocols during perioperative localized abdominal and thoracic neuroblastomas. Pediatr Surg Int 2024; 40:286. [PMID: 39487870 DOI: 10.1007/s00383-024-05884-w] [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: 10/26/2024] [Indexed: 11/04/2024]
Abstract
AIM To investigate the safety and efficacy of the application of enhanced recovery after surgery (ERAS) protocols in the perioperative period of abdominal and thoracic localized neuroblastomas (NBs). METHODS In this retrospective study, 68 children with NBs who underwent surgical resection of the tumor were enrolled. The ERAS protocols for NB excision were implemented in the ERAS group (n = 39) and the consequences were compared with children treated with traditional care (n = 29, TRAD group). The main outcomes of our interest included the incidence of surgery-related complications, the postoperative length of stay (LOS), and the Face/Legs/Activity/Cry/Consolability (FLACC) quantitative table from postoperative days (POD) 1-5. We also evaluated the median intraoperative fluid volume and anesthesia recovery time; blood glucose levels at the beginning of anesthesia, POD1, and 3; WBC counts, CRP values, and the concentration of plasma nutritional indicators on POD1 and 3; time of early ambulation, first anal exhaust, total enteral nutrition (TEN), and discontinue intravenous infusion postoperatively; usage proportion and duration of abdominal and thoracic drainages, nasogastric decompression tubes and urinary catheters; cost of hospitalization, parental satisfaction rate, and readmission rate of surgery ward within 30 days. RESULTS Compared to the TRAD group, the ERAS group had lower surgery-related complications, albeit not significantly (P > 0.05); the median postoperative LOS decreased from 11.0 to 8.0 days (P < 0.001), the LOS of abdominal NB was significantly shortened (P < 0.001) compared to thoracic NB (P = 0.07) between the two groups; the FLACC scores decreased significantly from POD1-5 (all P < 0.01). The ERAS group had an improved median intraoperative infusion speed (5.0 mL/kg/h vs 8.0 mL/kg/h), time of early ambulation (1.0 days vs 3.0 days), first anal exhaust (2.0 days vs 2.0 days), TEN (5.0 vs 7.0 days), discontinuation of intravenous infusion (5.0 days vs 8.0 days), and total cost of hospitalization (33,897.2 Yuan vs 38,876.3 Yuan); (all P < 0.01). The usage proportion and duration of surgical drainages and tubes were apparently reduced. The mean blood glucose level was higher at the beginning of anesthesia but lower on POD1 and 3 in the ERAS group (P < 0.01). No statistically significant difference was detected in WBC counts and concentrations of hemoglobin and albumin between the two groups of patients (P > 0.05), while the concentrations of prealbumin on POD3 were higher and the CRP level on POD1 was lower in the ERAS group than the TRAD group (P < 0.01). The satisfaction rate of parents was only slightly higher, but the difference was not statistically significant (P = 0.730). No obvious differences were observed in the aspects of NB resection (P = 0.462) and 30-day readmissions of surgery ward (P = 1.000). CONCLUSION The application of ERAS protocols has a significant potential to accelerate perioperative rehabilitation in children undergoing abdominal and thoracic NBs' surgical resection.
Collapse
Affiliation(s)
- Jingjing He
- Reproductive Medicine Center, Hefei Maternal and Child Health Hospital, Hefei, 230001, Anhui, China
| | - Zhiru Wang
- Department of General Surgery, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Xiyang Yu
- Department of Pediatric Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Yilin Su
- Department of Pediatric Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Mingyun Hong
- Reproductive Medicine Center, Hefei Maternal and Child Health Hospital, Hefei, 230001, Anhui, China.
| | - Kai Zhu
- Department of Pediatric Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| |
Collapse
|
16
|
Wei Z, Gong B, Li X, Chen C, Zhao Q. Event-free survival in neuroblastoma with MYCN amplification and deletion of 1p or 11q may be associated with altered immune status. BMC Cancer 2024; 24:1279. [PMID: 39407175 PMCID: PMC11481459 DOI: 10.1186/s12885-024-13044-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 10/08/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Neuroblastoma exhibits substantial heterogeneity, which is intricately linked to various genetic alterations. We aimed to explore immune status in the peripheral blood and prognosis of patients with neuroblastoma with different genetic characteristics. METHODS We enrolled 31 patients with neuroblastoma and collected samples to detect three genetic characteristics. Peripheral blood samples were tested for immune cells and cytokines by fluorescent microspheres conjugated with antibodies and flow cytometry. Event-free survival (EFS) was analyzed using the Kaplan‒Meier method. RESULTS Twenty-two patients had genetic aberrations, including MYCN amplification in 6 patients, chromosome 1p deletion in 9 patients, and chromosome 11q deletion in 14 patients. Two genetic alterations were present in seven patients. The EFS was worse in patients with MYCN amplification or 1p deletion than in the corresponding group, whereas 11q deletion was a prognostic factor only in patients with unamplified MYCN. Changes in immune status revealed a decrease in the proportion of T cells in blood, and an increase in regulatory T cells and immunosuppression-related cytokines such as interleukin (IL)-10. The EFS of the IL-10 high-level group was lower than that of the low-level group. Patients with concomitant genetic alterations and a high level of IL-10 had worse EFS than other patients. CONCLUSIONS Patients with neuroblastoma characterized by these genetic characteristics often have suppressed T cell response and an overabundance of immunosuppressive cells and cytokines in the peripheral blood. This imbalance is significantly associated with poor EFS. Moreover, if these patients show an elevated levels of immunosuppressive cytokines such as IL-10, the prognosis will be worse.
Collapse
Affiliation(s)
- Zixuan Wei
- Department of Pediatric Oncology, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, West Huan-Hu Rd, Ti Yuan Bei, Hexi District, 300060, Tianjin, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Baocheng Gong
- Department of Pediatric Oncology, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, West Huan-Hu Rd, Ti Yuan Bei, Hexi District, 300060, Tianjin, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Xin Li
- Tianjin Cancer Hospital Airport Hospital, Tianjin, China
| | - Chong Chen
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- National Human Genetic Resources Sharing Service Platform, Tianjin, China
| | - Qiang Zhao
- Department of Pediatric Oncology, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, West Huan-Hu Rd, Ti Yuan Bei, Hexi District, 300060, Tianjin, Tianjin, China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
- Tianjin's Clinical Research Center for Cancer, Tianjin, China.
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.
| |
Collapse
|
17
|
Han M, Niu H, Duan F, Wang Z, Zhang Z, Ren H. Research status and development trends of omics in neuroblastoma a bibliometric and visualization analysis. Front Oncol 2024; 14:1383805. [PMID: 39450262 PMCID: PMC11499224 DOI: 10.3389/fonc.2024.1383805] [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: 03/21/2024] [Accepted: 09/16/2024] [Indexed: 10/26/2024] Open
Abstract
Background Neuroblastoma (NB), a prevalent extracranial solid tumor in children, stems from the neural crest. Omics technologies are extensively employed in NB, and We analyzed published articles on NB omics to understand the research trends and hot topics in NB omics. Method We collected all articles related to NB omics published from 2005 to 2023 from the Web of Science Core Collection database. Subsequently, we conducted analyses using VOSviewer, CiteSpace, Bibliometrix, and the Bibliometric online analysis platform (https://bibliometric.com/ ). Results We included a total of 514 articles in our analysis. The increasing number of publications in this field since 2020 indicates growing attention to NB omics, gradually entering a mature development stage. These articles span 50 countries and 1,000 institutions, involving 3,669 authors and 292 journals. The United States has the highest publication output and collaboration with other countries, with Germany being the most frequent collaborator. Capital Medical University and the German Cancer Research Center are the institutions with the highest publication count. The Journal of Proteome Research and the Journal of Biological Chemistry are the most prolific journal and most co-cited journal, respectively. Wang, W, and Maris, JM are the scholars with the highest publication count and co-citations in this field. "Neuroblastoma" and "Expression" are the most frequent keywords, while "classification," "Metabolism," "Cancer," and "Diagnosis" are recent key terms. The article titled "Neuroblastoma" by John M. Maris is the most cited reference in this analysis. Conclusion The continuous growth in NB omics research underscores its increasing significance in the scientific community. Omics technologies have facilitated the identification of potential biomarkers, advancements in personalized medicine, and the development of novel therapeutic strategies. Despite these advancements, the field faces significant challenges, including tumor heterogeneity, data standardization issues, and the translation of research findings into clinical practice.
Collapse
Affiliation(s)
| | - Huizhong Niu
- First Department of General Surgery, Hebei Children’s Hospital,
Shijiazhuang, Hebei, China
| | | | | | | | | |
Collapse
|
18
|
Nian Z, Wang D, Wang H, Liu W, Ma Z, Yan J, Cao Y, Li J, Zhao Q, Liu Z. Single-cell RNA-seq reveals the transcriptional program underlying tumor progression and metastasis in neuroblastoma. Front Med 2024; 18:690-707. [PMID: 39014137 DOI: 10.1007/s11684-024-1081-7] [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: 01/08/2024] [Accepted: 04/18/2024] [Indexed: 07/18/2024]
Abstract
Neuroblastoma (NB) is one of the most common childhood malignancies. Sixty percent of patients present with widely disseminated clinical signs at diagnosis and exhibit poor outcomes. However, the molecular mechanisms triggering NB metastasis remain largely uncharacterized. In this study, we generated a transcriptomic atlas of 15 447 NB cells from eight NB samples, including paired samples of primary tumors and bone marrow metastases. We used time-resolved analysis to chart the evolutionary trajectory of NB cells from the primary tumor to the metastases in the same patient and identified a common 'starter' subpopulation that initiates tumor development and metastasis. The 'starter' population exhibited high expression levels of multiple cell cycle-related genes, indicating the important role of cell cycle upregulation in NB tumor progression. In addition, our evolutionary trajectory analysis demonstrated the involvement of partial epithelial-to-mesenchymal transition (p-EMT) along the metastatic route from the primary site to the bone marrow. Our study provides insights into the program driving NB metastasis and presents a signature of metastasis-initiating cells as an independent prognostic indicator and potential therapeutic target to inhibit the initiation of NB metastasis.
Collapse
Affiliation(s)
- Zhe Nian
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Dan Wang
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Hao Wang
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Wenxu Liu
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Zhenyi Ma
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Cell Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, 311121, China
| | - Jie Yan
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Yanna Cao
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Jie Li
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Qiang Zhao
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
| | - Zhe Liu
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Cell Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, 311121, China.
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
19
|
Sui X, Liu T, Zou Z, Zhang B, Zhang G. Effects and mechanisms of puerarin against neuroblastoma: insights from bioinformatics and in vitro experiments. BMC Complement Med Ther 2024; 24:257. [PMID: 38982456 PMCID: PMC11234716 DOI: 10.1186/s12906-024-04569-0] [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: 06/22/2023] [Accepted: 06/26/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND Neuroblastoma, a prevalent solid tumor in children, often manifests with hidden onset sites, rapid growth, and high metastatic potential. The prognosis for children with high-risk neuroblastoma remains poor, highlighting the urgent need for novel prognostic models and therapeutic avenues. In recent years, puerarin, as a kind of small molecule drug extracted from Chinese medicine Pueraria lobata, has demonstrated significant anticancer effects on various cancer cell types. In this study, through bioinformatics analysis and in vitro experiments, the potential and mechanism of puerarin in the treatment of neuroblastoma were investigated, and a prognostic model was established. METHODS A total of 9 drug-disease related targets were observed by constructing a database of drug targets and disease genes. Besides, GO and KEGG enrichment analysis was performed to explore the potential mechanism of its therapeutic effect. To construct the prognostic model, risk regression analysis and LASSO analysis were carried out for validation. Finally, the prognostic genes were identified. Parachute test and immunofluorescence staining were performed to verify the potential mechanism of puerarin in neuroblastoma treatment. RESULTS Three prognostic genes, i.e., BIRC5, TIMP2 and CASP9, were identified. In vitro studies verified puerarin's impact on BIRC5, TIMP2, and CASP9 expression, inhibiting proliferation in neuroblastoma SH-SY5Y cells. Puerarin disrupts the cytoskeleton, boosts gap junctional communication, curtailing invasion and migration, and induces mitochondrial damage in SH-SY5Y cells. CONCLUSIONS Based on network pharmacology and bioinformatics analysis, combined with in vitro experimental verification, puerarin was hereby observed to enhance GJIC in neuroblastoma, destroy cytoskeleton and thus inhibit cell invasion and migration, cause mitochondrial damage of tumor cells, and inhibit cell proliferation. Overall, puerarin, as a natural medicinal compound, does hold potential as a novel therapy for neuroblastoma.
Collapse
Affiliation(s)
- Xiaohui Sui
- Shandong University of Traditional Chinese Medicine, Jinan Shandong, 250014, China
| | - Tingting Liu
- Shandong University of Traditional Chinese Medicine, Jinan Shandong, 250014, China
| | - Zhiyun Zou
- Shandong University of Traditional Chinese Medicine, Jinan Shandong, 250014, China
| | - Baoqing Zhang
- Affiliated Hospital of Shandong, University of Traditional Chinese Medicine, Jinan Shandong, 250011, China
| | - Guiju Zhang
- Affiliated Hospital of Shandong, University of Traditional Chinese Medicine, Jinan Shandong, 250011, China.
| |
Collapse
|
20
|
Linga BG, Mohammed SGAA, Farrell T, Rifai HA, Al-Dewik N, Qoronfleh MW. Genomic Newborn Screening for Pediatric Cancer Predisposition Syndromes: A Holistic Approach. Cancers (Basel) 2024; 16:2017. [PMID: 38893137 PMCID: PMC11171256 DOI: 10.3390/cancers16112017] [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: 04/19/2024] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
As next-generation sequencing (NGS) has become more widely used, germline and rare genetic variations responsible for inherited illnesses, including cancer predisposition syndromes (CPSs) that account for up to 10% of childhood malignancies, have been found. The CPSs are a group of germline genetic disorders that have been identified as risk factors for pediatric cancer development. Excluding a few "classic" CPSs, there is no agreement regarding when and how to conduct germline genetic diagnostic studies in children with cancer due to the constant evolution of knowledge in NGS technologies. Various clinical screening tools have been suggested to aid in the identification of individuals who are at greater risk, using diverse strategies and with varied outcomes. We present here an overview of the primary clinical and molecular characteristics of various CPSs and summarize the existing clinical genomics data on the prevalence of CPSs in pediatric cancer patients. Additionally, we discuss several ethical issues, challenges, limitations, cost-effectiveness, and integration of genomic newborn screening for CPSs into a healthcare system. Furthermore, we assess the effectiveness of commonly utilized decision-support tools in identifying patients who may benefit from genetic counseling and/or direct genetic testing. This investigation highlights a tailored and systematic approach utilizing medical newborn screening tools such as the genome sequencing of high-risk newborns for CPSs, which could be a practical and cost-effective strategy in pediatric cancer care.
Collapse
Affiliation(s)
- BalaSubramani Gattu Linga
- Department of Research, Women’s Wellness and Research Center, Hamad Medical Corporation (HMC), P.O. Box 3050, Doha 0974, Qatar
- Translational and Precision Medicine Research, Women’s Wellness and Research Center (WWRC), Hamad Medical Corporation (HMC), Doha 0974, Qatar
| | | | - Thomas Farrell
- Department of Research, Women’s Wellness and Research Center, Hamad Medical Corporation (HMC), P.O. Box 3050, Doha 0974, Qatar
| | - Hilal Al Rifai
- Neonatal Intensive Care Unit (NICU), Newborn Screening Unit, Department of Pediatrics and Neonatology, Women’s Wellness and Research Center (WWRC), Hamad Medical Corporation (HMC), Doha 0974, Qatar
| | - Nader Al-Dewik
- Department of Research, Women’s Wellness and Research Center, Hamad Medical Corporation (HMC), P.O. Box 3050, Doha 0974, Qatar
- Translational and Precision Medicine Research, Women’s Wellness and Research Center (WWRC), Hamad Medical Corporation (HMC), Doha 0974, Qatar
- Neonatal Intensive Care Unit (NICU), Newborn Screening Unit, Department of Pediatrics and Neonatology, Women’s Wellness and Research Center (WWRC), Hamad Medical Corporation (HMC), Doha 0974, Qatar
- Genomics and Precision Medicine (GPM), College of Health & Life Science (CHLS), Hamad Bin Khalifa University (HBKU), Doha 0974, Qatar
- Faculty of Health and Social Care Sciences, Kingston University and St George’s University of London, Kingston upon Thames, Surrey, London KT1 2EE, UK
| | - M. Walid Qoronfleh
- Healthcare Research & Policy Division, Q3 Research Institute (QRI), Ann Arbor, MI 48197, USA
| |
Collapse
|
21
|
Hu X, Zhou Y, Hill C, Chen K, Cheng C, Liu X, Duan P, Gu Y, Wu Y, Ewing RM, Li Z, Wu Z, Wang Y. Identification of MYCN non-amplified neuroblastoma subgroups points towards molecular signatures for precision prognosis and therapy stratification. Br J Cancer 2024; 130:1841-1854. [PMID: 38553589 PMCID: PMC7616008 DOI: 10.1038/s41416-024-02666-y] [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: 12/07/2023] [Revised: 03/13/2024] [Accepted: 03/19/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUND Despite the extensive study of MYCN-amplified neuroblastomas, there is a significant unmet clinical need in MYCN non-amplified cases. In particular, the extent of heterogeneity within the MYCN non-amplified population is unknown. METHODS A total of 1566 samples from 16 datasets were identified in Gene Expression Omnibus (GEO) and ArrayExpress. Characterisation of the subtypes was analysed by ConsensusClusterPlus. Independent predictors for subgrouping were constructed from the single sample predictor based on the multiclassPairs package. Findings were verified using immunohistochemistry and CIBERSORTx analysis. RESULTS We demonstrate that MYCN non-amplified neuroblastomas are heterogeneous and can be classified into 3 subgroups based on their transcriptional signatures. Within these groups, subgroup_2 has the worst prognosis and this group shows a 'MYCN' signature that is potentially induced by the overexpression of Aurora Kinase A (AURKA); whilst subgroup_3 is characterised by an 'inflamed' gene signature. The clinical implications of this subtype classification are significant, as each subtype demonstrates a unique prognosis and vulnerability to investigational therapies. A total of 420 genes were identified as independent subgroup predictors with average balanced accuracy of 0.93 and 0.84 for train and test datasets, respectively. CONCLUSION We propose that transcriptional subtyping may enhance precision prognosis and therapy stratification for patients with MYCN non-amplified neuroblastomas.
Collapse
Affiliation(s)
- Xiaoxiao Hu
- Department of Paediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200092, China
- Department of Paediatric Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Division of Paediatric Oncology, Shanghai Institute of Paediatric Research, Shanghai, 200092, China
| | - Yilu Zhou
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Charlotte Hill
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Kai Chen
- Department of Paediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200092, China
- Division of Paediatric Oncology, Shanghai Institute of Paediatric Research, Shanghai, 200092, China
| | - Cheng Cheng
- Department of Paediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200092, China
- Division of Paediatric Oncology, Shanghai Institute of Paediatric Research, Shanghai, 200092, China
| | - Xiaowei Liu
- Department of Paediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200092, China
- Division of Paediatric Oncology, Shanghai Institute of Paediatric Research, Shanghai, 200092, China
| | - Peiwen Duan
- Department of Paediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200092, China
- Division of Paediatric Oncology, Shanghai Institute of Paediatric Research, Shanghai, 200092, China
| | - Yaoyao Gu
- Department of Paediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200092, China
- Division of Paediatric Oncology, Shanghai Institute of Paediatric Research, Shanghai, 200092, China
| | - Yeming Wu
- Department of Paediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200092, China
- Division of Paediatric Oncology, Shanghai Institute of Paediatric Research, Shanghai, 200092, China
- Department of Paediatric Surgery, Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Rob M Ewing
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Zhongrong Li
- Department of Paediatric Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Zhixiang Wu
- Department of Paediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200092, China.
- Division of Paediatric Oncology, Shanghai Institute of Paediatric Research, Shanghai, 200092, China.
- Department of Paediatric Surgery, Children's Hospital of Soochow University, Suzhou, 215003, China.
| | - Yihua Wang
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK.
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK.
| |
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
|
Tan J, McLoone JK, Wakefield CE, Nassar N, Cohn RJ, Signorelli C. Neuroblastoma survivors' self-reported late effects, quality of life, health-care use, and risk perceptions. Palliat Support Care 2024; 22:296-305. [PMID: 37311662 DOI: 10.1017/s1478951523000615] [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: 06/15/2023]
Abstract
BACKGROUND Survivors of childhood neuroblastoma are at risk of multiple treatment-related health problems (late effects), impacting their quality of life. While late effects and quality of life among Australia and New Zealand (ANZ) childhood cancer survivors have been reported, the outcomes of neuroblastoma survivors specifically have not been reported, limiting critical information to inform treatment and care. METHODS Young neuroblastoma survivors or their parents (as proxy for survivors <16 years) were invited to complete a survey and optional telephone interview. Survivors' late effects, risk perceptions, health-care use, and health-related quality of life were surveyed and analyzed using descriptive statistics and linear regression analyses. In-depth interviews explored participants' experiences, knowledge, and perception of late effects and information needs. Thematic content analysis was used to summarize the data. RESULTS Thirty-nine neuroblastoma survivors or parents completed questionnaires (median age = 16 years, 39% male), with 13 also completing interviews. Thirty-two participants (82%) reported experiencing at least 1 late effect, most commonly dental problems (56%), vision/hearing problems (47%), and fatigue (44%). Participants reported high overall quality of life (index = 0.9, range = 0.2-1.0); however, more participants experienced anxiety/depression compared to the population norm (50% met criteria versus 25%, χ2 = 13, p < 0.001). Approximately half of participants (53%) believed they were at risk of developing further late effects. Qualitatively, participants reported knowledge gaps in understanding their risk of developing late effects. CONCLUSION Many neuroblastoma survivors appear to experience late effects, anxiety/depression and have unmet cancer-related information needs. This study highlights important areas for intervention to reduce the impact of neuroblastoma and its treatment in childhood and young adulthood.
Collapse
Affiliation(s)
- Jessica Tan
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
- Discipline of Paediatrics & Child Health, School of Clinical Medicine, UNSW Medicine & Health, Randwick Clinical Campus, UNSW Sydney, Sydney, NSW, Australia
| | - Jordana K McLoone
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
- Discipline of Paediatrics & Child Health, School of Clinical Medicine, UNSW Medicine & Health, Randwick Clinical Campus, UNSW Sydney, Sydney, NSW, Australia
| | - Claire E Wakefield
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
- Discipline of Paediatrics & Child Health, School of Clinical Medicine, UNSW Medicine & Health, Randwick Clinical Campus, UNSW Sydney, Sydney, NSW, Australia
| | - Natasha Nassar
- Sydney School of Public Health, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Children's Hospital at Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Richard J Cohn
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
- Discipline of Paediatrics & Child Health, School of Clinical Medicine, UNSW Medicine & Health, Randwick Clinical Campus, UNSW Sydney, Sydney, NSW, Australia
| | - Christina Signorelli
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
- Discipline of Paediatrics & Child Health, School of Clinical Medicine, UNSW Medicine & Health, Randwick Clinical Campus, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
24
|
Primus PS, Wu CHY, Kao CL, Choo YM. Glabraquinone A and B, new bisanthraquinones from Prismatomeris glabra (Korth.) Valeton. Nat Prod Res 2024; 38:1406-1413. [PMID: 36416441 DOI: 10.1080/14786419.2022.2147932] [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: 02/09/2022] [Revised: 11/01/2022] [Accepted: 11/07/2022] [Indexed: 11/24/2022]
Abstract
Two new bisanthraquinones, glabraquinone A and B (1-2) were isolated from the root of Prismatomeris glabra (Korth.) Valeton. In addition to the new glabraquinones, six known anthraquinones, that is, 1-hydroxy-2-methoxy-6-methylanthraquinone (3), 1,2-dimethoxy-7-methylanthraquinone (4), lucidin (5), nordamnacanthal (6), damnacanthal (7) and 2-carboxaldehyde-3-hydroxyanthraquinone (8)) and an aromatic compound, that is, catechol diethyl ether (9) were isolated and characterized in this study. Compounds 1, 4 and 9 showed mild activity, reducing N2A cell viability to 77%, 82% and 77%, respectively, in anti-neuroblastoma assay.
Collapse
Affiliation(s)
- Phoebe Sussana Primus
- Department of Chemistry, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| | - Carol Hsin-Yi Wu
- Division of Cellular and Immune Therapy, Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Taiwan
| | - Chai-Lin Kao
- Division of Cellular and Immune Therapy, Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Taiwan
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Taiwan
| | - Yeun-Mun Choo
- Department of Chemistry, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
25
|
Feng L, Yao X, Lu X, Wang C, Wang W, Yang J. Differentiation of early relapse and late relapse in intermediate- and high-risk neuroblastoma with an 18F-FDG PET/CT-based radiomics nomogram. Abdom Radiol (NY) 2024; 49:888-899. [PMID: 38315193 DOI: 10.1007/s00261-023-04181-9] [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: 11/18/2023] [Revised: 12/25/2023] [Accepted: 12/27/2023] [Indexed: 02/07/2024]
Abstract
OBJECTIVES To develop and validate an 18F-FDG PET/CT-based radiomics nomogram for differentiating early relapse and late relapse of intermediate- and high-risk neuroblastoma (NB). METHODS A total of eighty-five patients with relapsed NB who underwent 18F-FDG PET/CT were retrospectively evaluated. All selected patients were randomly assigned to the training set and the validation set in a 7:3 ratio. Tumors were segmented using the 3D slicer, followed by radiomics features extraction. Features selection was performed using random forest, and the radiomics score was constructed by logistic regression analysis. Clinical risk factors were identified, and the clinical model was constructed using logistic regression analysis. A radiomics nomogram was constructed by combining the radiomics score and clinical risk factors, and its performance was evaluated by receiver operating characteristic (ROC) curves, calibration curves, and decision curve analysis (DCA). RESULTS Finally, the 12 most important radiomics features were used for modeling, with an area under the curve (AUC) of 0.835 and 0.824 in the training and validation sets, respectively. Age at diagnosis and International Neuroblastoma Pathology Classification were determined as clinical risk factors to construct the clinical model. In addition, the nomogram achieved an AUC of 0.902 and 0.889 for identifying early relapse in the training and validation sets, respectively, which is higher than the clinical model (AUC of 0.712 and 0.588, respectively). The predicted early relapse and actual early relapse in the calibration curves were in good agreement. The DCA showed that the radiomics nomogram was clinically useful. CONCLUSION Our 18F-FDG PET/CT-based radiomics nomogram can well predict early relapse and late relapse of intermediate- and high-risk NB, which contributes to follow-up and management in clinical practice.
Collapse
Affiliation(s)
- Lijuan Feng
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, China
| | - Xilan Yao
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, China
| | - Xia Lu
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, China
| | - Chao Wang
- SinoUnion Healthcare Inc., Beijing, China
| | - Wei Wang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, China
| | - Jigang Yang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, China.
| |
Collapse
|
26
|
Berthenet K, Aïmontché E, El Mrini S, Brière J, Pion N, Iacono I, Brejon S, Monier K, Catez F, Ichim G, Combaret V, Mertani HC, Diaz JJ, Albaret MA. Spatial sequestration of activated-caspase 3 in aggresomes mediates resistance of neuroblastoma cell to bortezomib treatment. Sci Rep 2024; 14:3768. [PMID: 38355966 PMCID: PMC10866921 DOI: 10.1038/s41598-024-54140-7] [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: 01/05/2023] [Accepted: 02/08/2024] [Indexed: 02/16/2024] Open
Abstract
Neuroblastoma (NB) is the most common pediatric tumor and is currently treated by several types of therapies including chemotherapies, such as bortezomib treatment. However, resistance to bortezomib is frequently observed by mechanisms that remain to be deciphered. Bortezomib treatment leads to caspase activation and aggresome formation. Using models of patients-derived NB cell lines with different levels of sensitivity to bortezomib, we show that the activated form of caspase 3 accumulates within aggresomes of NB resistant cells leading to an impairment of bortezomib-induced apoptosis and increased cell survival. Our findings unveil a new mechanism of resistance to chemotherapy based on an altered subcellular distribution of the executioner caspase 3. This mechanism could explain the resistance developed in NB patients treated with bortezomib, emphasizing the potential of drugs targeting aggresomes.
Collapse
Affiliation(s)
- Kévin Berthenet
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard, Cancer Research Center of Lyon, 69008, Lyon, France
| | - Eliézer Aïmontché
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard, Cancer Research Center of Lyon, 69008, Lyon, France
| | - Sara El Mrini
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard, Cancer Research Center of Lyon, 69008, Lyon, France
| | - Johan Brière
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard, Cancer Research Center of Lyon, 69008, Lyon, France
| | - Nathalie Pion
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard, Cancer Research Center of Lyon, 69008, Lyon, France
| | - Isabelle Iacono
- Department of Translational Research and Innovation, Centre Léon Bérard, 69373, Lyon, France
| | - Stéphanie Brejon
- Department of Translational Research and Innovation, Centre Léon Bérard, 69373, Lyon, France
| | - Karine Monier
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard, Cancer Research Center of Lyon, 69008, Lyon, France
| | - Frédéric Catez
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard, Cancer Research Center of Lyon, 69008, Lyon, France
| | - Gabriel Ichim
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard, Cancer Research Center of Lyon, 69008, Lyon, France
- Institut Convergence PLAsCAN, 69373, Lyon Cedex 08, France
| | - Valérie Combaret
- Department of Translational Research and Innovation, Centre Léon Bérard, 69373, Lyon, France
| | - Hichem C Mertani
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard, Cancer Research Center of Lyon, 69008, Lyon, France
| | - Jean-Jacques Diaz
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard, Cancer Research Center of Lyon, 69008, Lyon, France
- Institut Convergence PLAsCAN, 69373, Lyon Cedex 08, France
- DevWeCan Labex Laboratory, 69373, Lyon Cedex 08, France
| | - Marie Alexandra Albaret
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard, Cancer Research Center of Lyon, 69008, Lyon, France.
| |
Collapse
|
27
|
Sheng J, Li T, Xu H, Xu R, Cai X, Zhang H, Ji Q, Duan X, Xia W, Yang X. Evaluation of clinical and imaging features for differentiating rhabdomyosarcoma from neuroblastoma in pediatric soft tissue. Front Oncol 2024; 14:1289532. [PMID: 38406807 PMCID: PMC10884217 DOI: 10.3389/fonc.2024.1289532] [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: 09/06/2023] [Accepted: 01/16/2024] [Indexed: 02/27/2024] Open
Abstract
Background In this study, we developed a nomogram predictive model based on clinical, CT, and MRI parameters to differentiate soft tissue rhabdomyosarcoma (RMS) from neuroblastoma (NB) in children preoperatively. Materials and methods A total of 103 children with RMS (n=37) and NB (n=66) were enrolled in the study from December 2012 to July 2023. The clinical and imaging data (assessed by two experienced radiologists) were analyzed using univariate analysis, and significant factors were further analyzed by multivariable logistic regression using the forward LR method to develop the clinical model, radiological model, and integrated nomogram model, respectively. The diagnostic performances, goodness of fit, and clinical utility of the integrated nomogram model were assessed using the area under the curve (AUC) of the receiver operator characteristics curve (ROC) with a 95% confidence interval (95% CI), calibration curve, and decision curve analysis (DCA) curves, respectively. Diagnostic efficacy between the model and radiologists' interpretations was examined. Results The median age at diagnosis in the RMS group was significantly older than the NB group (36.0 months vs. 14.5 months; P=0.003); the fever rates in RMS patients were significantly lower than in patients with NB (0.0% vs.16.7%; P=0.022), and the incidence of palpable mass was higher in patients with RMS compared with the NB patients (89.2% vs. 34.8%; P<0.001). Compare NB on image features: RMS occurred more frequently in the head and neck and displayed homogeneous density on non-enhanced CT than NB (48.6% vs. 9.1%; 35.3% vs. 13.8%, respectively; all P<0.05), and the occurrence of characteristics such as calcification, encasing vessels, and intraspinal tumor extension was significantly less frequent in RMS children compared to children with NB (18.9% vs. 84.8%; 13.5% vs. 34.8%; 2.7% vs. 50.0%, respectively; all P <0.05). Two, three, and four features were identified as independent parameters by multivariate logistic regression analysis to develop the clinical, radiological, and integrated nomogram models, respectively. The AUC value (0.962), calibration curve, and DCA showed that the integrated nomogram model may provide better diagnostic performance, good agreement, and greater clinical net benefits than the clinical model, radiological model, and radiologists' subjective diagnosis. Conclusion The clinical and imaging features-based nomogram has potential for helping radiologists distinguish between pediatric soft tissue RMS and NB patients preoperatively, and reduce unnecessary interventions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Xiujun Yang
- Department of Radiology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
28
|
Muehling J, Fröba-Pohl A, Muensterer OJ, von Schweinitz D, Kappler R. Impact of BCL-2 Expression on Course of Disease in Neuroblastoma. Eur J Pediatr Surg 2024; 34:69-77. [PMID: 37774735 DOI: 10.1055/s-0043-1774798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/01/2023]
Abstract
OBJECTIVE The antiapoptotic BCL-2 protein has implications for maturation and differentiation of neural tissue and acts as a strong modulator of carcinogenesis in different tumors. Recent research focuses not only on its benefit as a prognostic factor, but also as a potential therapeutic target. The role of BCL-2 in neuroblastoma, the most common extracranial solid tumor in childhood, remains controversial. The aim of our study was to determine the gene expression level of BCL-2 in a large cohort of neuroblastoma patients and its correlation with clinical parameters. METHODS Tumor samples and clinical data were collected from 100 neuroblastoma patients treated according to the NB2004 protocol of the German Society of Pediatric Oncology and Hematology. BCL-2 gene expression levels were measured by quantitative reverse transcription polymerase chain reaction and correlated with clinical parameters. RESULTS BCL-2 expression was detected in all tumor samples. Relative BCL-2 expression levels were higher in females versus males (1.839 vs. 1.342; p = 0.0143), in patients with low versus high International Neuroblastoma Staging System stage (2.051 vs. 1.463; p = 0.0206), in nonmetastatic versus metastatic disease (1.801 vs. 1.342; p = 0.0242), as well as in patients without presurgical chemotherapy (2.145 vs. 1.402; p = 0.0016), but was not associated with overall survival and MYCN amplification. CONCLUSION Our study demonstrates the ubiquitous expression of BCL-2 in neuroblastoma and suggests the possibility for targeted therapy with BCL-2 inhibitors, even in lower-stage neuroblastoma. It also underlines the need for further research on concomitant genetic alterations for a better understanding of the impact of BCL-2 on this pediatric tumor type.
Collapse
Affiliation(s)
- Jakob Muehling
- Department of Pediatric Surgery, Munich University Hospital, Dr. von Hauner Children's Hospital, München, Germany
| | - Alexandra Fröba-Pohl
- Department of Pediatric Surgery, Munich University Hospital, Dr. von Hauner Children's Hospital, München, Germany
| | - Oliver J Muensterer
- Department of Pediatric Surgery, Munich University Hospital, Dr. von Hauner Children's Hospital, München, Germany
| | - Dietrich von Schweinitz
- Department of Pediatric Surgery, Munich University Hospital, Dr. von Hauner Children's Hospital, München, Germany
| | - Roland Kappler
- Department of Pediatric Surgery, Munich University Hospital, Dr. von Hauner Children's Hospital, München, Germany
| |
Collapse
|
29
|
Jiang H, Tiche SJ, He CJ, Jedoui M, Forgo B, Zhao M, He B, Li Y, Li AM, Truong AT, Ho J, Simmermaker C, Yang Y, Zhou MN, Hu Z, Cuthbertson DJ, Svensson KJ, Hazard FK, Shimada H, Chiu B, Ye J. Mitochondrial uncoupler and retinoic acid synergistically induce differentiation and inhibit proliferation in neuroblastoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.22.576741. [PMID: 38328117 PMCID: PMC10849550 DOI: 10.1101/2024.01.22.576741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Neuroblastoma is a leading cause of death in childhood cancer cases. Unlike adult malignancies, which typically develop from aged cells through accumulated damage and mutagenesis, neuroblastoma originates from neural crest cells with disrupted differentiation. This distinct feature provides novel therapeutic opportunities beyond conventional cytotoxic methods. Previously, we reported that the mitochondrial uncoupler NEN (niclosamide ethanolamine) activated mitochondria respiration to reprogram the epigenome, promoting neuronal differentiation. In the current study, we further combine NEN with retinoic acid (RA) to promote neural differentiation both in vitro and in vivo. The treatment increased the expression of RA signaling and neuron differentiation-related genes, resulting in a global shift in the transcriptome towards a more favorable prognosis. Overall, these results suggest that the combination of a mitochondrial uncoupler and the differentiation agent RA is a promising therapeutic strategy for neuroblastoma.
Collapse
Affiliation(s)
- Haowen Jiang
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | | | - Clifford JiaJun He
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Mohamed Jedoui
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Balint Forgo
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Meng Zhao
- Department of Pathology, Stanford University, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Bo He
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Yang Li
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Albert M. Li
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | | | - Jestine Ho
- Agilent Technologies, Inc., Santa Clara, CA, USA
| | | | - Yanan Yang
- Agilent Technologies, Inc., Santa Clara, CA, USA
| | - Meng-Ning Zhou
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Zhen Hu
- Olivia Consulting Service, Redwood City, CA, USA
| | | | - Katrin J. Svensson
- Department of Pathology, Stanford University, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | | | | | - Bill Chiu
- Department of Surgery, Stanford University, Stanford, CA, USA
| | - Jiangbin Ye
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| |
Collapse
|
30
|
Patel AG, Ashenberg O, Collins NB, Segerstolpe Å, Jiang S, Slyper M, Huang X, Caraccio C, Jin H, Sheppard H, Xu K, Chang TC, Orr BA, Shirinifard A, Chapple RH, Shen A, Clay MR, Tatevossian RG, Reilly C, Patel J, Lupo M, Cline C, Dionne D, Porter CBM, Waldman J, Bai Y, Zhu B, Barrera I, Murray E, Vigneau S, Napolitano S, Wakiro I, Wu J, Grimaldi G, Dellostritto L, Helvie K, Rotem A, Lako A, Cullen N, Pfaff KL, Karlström Å, Jané-Valbuena J, Todres E, Thorner A, Geeleher P, Rodig SJ, Zhou X, Stewart E, Johnson BE, Wu G, Chen F, Yu J, Goltsev Y, Nolan GP, Rozenblatt-Rosen O, Regev A, Dyer MA. A spatial cell atlas of neuroblastoma reveals developmental, epigenetic and spatial axis of tumor heterogeneity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.07.574538. [PMID: 38260392 PMCID: PMC10802404 DOI: 10.1101/2024.01.07.574538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Neuroblastoma is a pediatric cancer arising from the developing sympathoadrenal lineage with complex inter- and intra-tumoral heterogeneity. To chart this complexity, we generated a comprehensive cell atlas of 55 neuroblastoma patient tumors, collected from two pediatric cancer institutions, spanning a range of clinical, genetic, and histologic features. Our atlas combines single-cell/nucleus RNA-seq (sc/scRNA-seq), bulk RNA-seq, whole exome sequencing, DNA methylation profiling, spatial transcriptomics, and two spatial proteomic methods. Sc/snRNA-seq revealed three malignant cell states with features of sympathoadrenal lineage development. All of the neuroblastomas had malignant cells that resembled sympathoblasts and the more differentiated adrenergic cells. A subset of tumors had malignant cells in a mesenchymal cell state with molecular features of Schwann cell precursors. DNA methylation profiles defined four groupings of patients, which differ in the degree of malignant cell heterogeneity and clinical outcomes. Using spatial proteomics, we found that neuroblastomas are spatially compartmentalized, with malignant tumor cells sequestered away from immune cells. Finally, we identify spatially restricted signaling patterns in immune cells from spatial transcriptomics. To facilitate the visualization and analysis of our atlas as a resource for further research in neuroblastoma, single cell, and spatial-omics, all data are shared through the Human Tumor Atlas Network Data Commons at www.humantumoratlas.org.
Collapse
Affiliation(s)
- Anand G Patel
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
- These authors contributed equally
| | - Orr Ashenberg
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- These authors contributed equally
| | - Natalie B Collins
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
- These authors contributed equally
| | - Åsa Segerstolpe
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sizun Jiang
- Department of Pathology, Stanford University, Stanford, CA, USA
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Michal Slyper
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Xin Huang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Chiara Caraccio
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Hongjian Jin
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Heather Sheppard
- Comparative Pathology Core, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ke Xu
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ti-Cheng Chang
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Brent A Orr
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Abbas Shirinifard
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Richard H Chapple
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Amber Shen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michael R Clay
- Department of Pathology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Ruth G Tatevossian
- Cancer Biomarkers Laboratory, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Colleen Reilly
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jaimin Patel
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Marybeth Lupo
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Cynthia Cline
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Danielle Dionne
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Caroline B M Porter
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Julia Waldman
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yunhao Bai
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Bokai Zhu
- Department of Pathology, Stanford University, Stanford, CA, USA
| | | | - Evan Murray
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sébastien Vigneau
- Center for Cancer Genomics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sara Napolitano
- Center for Cancer Genomics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Isaac Wakiro
- Center for Cancer Genomics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jingyi Wu
- Center for Cancer Genomics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Grace Grimaldi
- Center for Cancer Genomics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Laura Dellostritto
- Center for Cancer Genomics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Karla Helvie
- Center for Cancer Genomics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Asaf Rotem
- Center for Cancer Genomics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ana Lako
- Center for Immuno-Oncology (CIO), Dana-Farber Cancer Institute, Boston, MA, USA
| | - Nicole Cullen
- Center for Immuno-Oncology (CIO), Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kathleen L Pfaff
- Center for Immuno-Oncology (CIO), Dana-Farber Cancer Institute, Boston, MA, USA
| | - Åsa Karlström
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Judit Jané-Valbuena
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ellen Todres
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Aaron Thorner
- Center for Cancer Genomics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Paul Geeleher
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Scott J Rodig
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Xin Zhou
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Elizabeth Stewart
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Bruce E Johnson
- Center for Cancer Genomics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Gang Wu
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Fei Chen
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Jiyang Yu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yury Goltsev
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Garry P Nolan
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Orit Rozenblatt-Rosen
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Current address: Research and Early Development, Genentech Inc., South San Francisco, CA, 94080, USA
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
- Koch Institute of Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Current address: Research and Early Development, Genentech Inc., South San Francisco, CA, 94080, USA
- Lead contacts
| | - Michael A Dyer
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Lead contacts
| |
Collapse
|
31
|
Cermakova K, Tao L, Dejmek M, Sala M, Montierth MD, Chan YS, Patel I, Chambers C, Loeza Cabrera M, Hoffman D, Parchem RJ, Wang W, Nencka R, Barbieri E, Hodges HC. Reactivation of the G1 enhancer landscape underlies core circuitry addiction to SWI/SNF. Nucleic Acids Res 2024; 52:4-21. [PMID: 37993417 PMCID: PMC10783513 DOI: 10.1093/nar/gkad1081] [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: 08/23/2023] [Revised: 09/29/2023] [Accepted: 10/27/2023] [Indexed: 11/24/2023] Open
Abstract
Several cancer core regulatory circuitries (CRCs) depend on the sustained generation of DNA accessibility by SWI/SNF chromatin remodelers. However, the window when SWI/SNF is acutely essential in these settings has not been identified. Here we used neuroblastoma (NB) cells to model and dissect the relationship between cell-cycle progression and SWI/SNF ATPase activity. We find that SWI/SNF inactivation impairs coordinated occupancy of non-pioneer CRC members at enhancers within 1 hour, rapidly breaking their autoregulation. By precisely timing inhibitor treatment following synchronization, we show that SWI/SNF is dispensable for survival in S and G2/M, but becomes acutely essential only during G1 phase. We furthermore developed a new approach to analyze the oscillating patterns of genome-wide DNA accessibility across the cell cycle, which revealed that SWI/SNF-dependent CRC binding sites are enriched at enhancers with peak accessibility during G1 phase, where they activate genes involved in cell-cycle progression. SWI/SNF inhibition strongly impairs G1-S transition and potentiates the ability of retinoids used clinically to induce cell-cycle exit. Similar cell-cycle effects in diverse SWI/SNF-addicted settings highlight G1-S transition as a common cause of SWI/SNF dependency. Our results illustrate that deeper knowledge of the temporal patterns of enhancer-related dependencies may aid the rational targeting of addicted cancers.
Collapse
Affiliation(s)
- Katerina Cermakova
- Department of Molecular and Cellular Biology, and Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
| | - Ling Tao
- Department of Pediatrics, Section of Hematology-Oncology, Texas Children's Cancer and Hematology Center, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Milan Dejmek
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Michal Sala
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Matthew D Montierth
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Graduate Program in Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX, USA
| | - Yuen San Chan
- Department of Molecular and Cellular Biology, and Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
| | - Ivanshi Patel
- Stem Cells and Regenerative Medicine Center, Center for Cell and Gene Therapy, and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA
| | - Courtney Chambers
- Department of Molecular and Cellular Biology, and Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
- Translational Biology and Molecular Medicine Graduate Program, Houston, TX, USA
| | - Mario Loeza Cabrera
- Department of Molecular and Cellular Biology, and Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
- Development, Disease Models and Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, USA
| | - Dane Hoffman
- Department of Molecular and Cellular Biology, and Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
- Cancer and Cell Biology Graduate Program, Baylor College of Medicine, Houston, TX, USA
| | - Ronald J Parchem
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Center for Cell and Gene Therapy, and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Wenyi Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Radim Nencka
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Eveline Barbieri
- Department of Pediatrics, Section of Hematology-Oncology, Texas Children's Cancer and Hematology Center, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - H Courtney Hodges
- Department of Molecular and Cellular Biology, and Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Bioengineering, Rice University, Houston, TX, USA
| |
Collapse
|
32
|
Mazar J, Brooks JK, Peloquin M, Rosario R, Sutton E, Longo M, Drehner D, Westmoreland TJ. The Oncolytic Activity of Zika Viral Therapy in Human Neuroblastoma In Vivo Models Confers a Major Survival Advantage in a CD24-dependent Manner. CANCER RESEARCH COMMUNICATIONS 2024; 4:65-80. [PMID: 38214542 PMCID: PMC10775766 DOI: 10.1158/2767-9764.crc-23-0221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 09/14/2023] [Accepted: 12/06/2023] [Indexed: 01/13/2024]
Abstract
Neuroblastoma is the most common extracranial tumor, accounting for 15% of all childhood cancer-related deaths. The long-term survival of patients with high-risk tumors is less than 40%, and MYCN amplification is one of the most common indicators of poor outcomes. Zika virus (ZIKV) is a mosquito-borne flavivirus associated with mild constitutional symptoms outside the fetal period. Our published data showed that high-risk and recurrent neuroblastoma cells are permissive to ZIKV infection, resulting in cell type-specific lysis. In this study, we assessed the efficacy of ZIKV as an oncolytic treatment for high-risk neuroblastoma using in vivo tumor models. Utilizing both MYCN-amplified and non-amplified models, we demonstrated that the application of ZIKV had a rapid tumoricidal effect. This led to a nearly total loss of the tumor mass without evidence of recurrence, offering a robust survival advantage to the host. Detection of the viral NS1 protein within the tumors confirmed that a permissive infection preceded tissue necrosis. Despite robust titers within the tumor, viral shedding to the host was poor and diminished rapidly, correlating with no detectable side effects to the murine host. Assessments from both primary pretreatment and recurrent posttreatment isolates confirmed that permissive sensitivity to ZIKV killing was dependent on the expression of CD24, which was highly expressed in neuroblastomas and conferred a proliferative advantage to tumor growth. Exploiting this viral sensitivity to CD24 offers the possibility of its use as a prognostic target for a broad population of expressing cancers, many of which have shown resistance to current clinical therapies. SIGNIFICANCE Sensitivity to the tumoricidal effect of ZIKV on high-risk neuroblastoma tumors is dependent on CD24 expression, offering a prognostic marker for this oncolytic therapy in an extensive array of CD24-expressing cancers.
Collapse
Affiliation(s)
- Joseph Mazar
- Nemours Children's Hospital, Nemours Parkway, Orlando, Florida
- Burnett School of Biological Sciences, The University of Central Florida College of Medicine, Orlando, Florida
| | | | | | - Rosa Rosario
- Nemours Children's Hospital, Nemours Parkway, Orlando, Florida
| | - Emma Sutton
- Nemours Children's Hospital, Nemours Parkway, Orlando, Florida
| | - Matthew Longo
- Nemours Children's Hospital, Nemours Parkway, Orlando, Florida
- Burnett School of Biological Sciences, The University of Central Florida College of Medicine, Orlando, Florida
| | - Dennis Drehner
- Nemours Children's Hospital, Nemours Parkway, Orlando, Florida
| | - Tamarah J. Westmoreland
- Nemours Children's Hospital, Nemours Parkway, Orlando, Florida
- Burnett School of Biological Sciences, The University of Central Florida College of Medicine, Orlando, Florida
| |
Collapse
|
33
|
Morin CE, Hasweh R, Anton C, Dillman JR, Orscheln E, Smith EA, Kotagal M, Weiss BD, Ouyang J, Zhang B, Trout AT, Towbin AJ. Gadolinium-based contrast media does not improve the staging of neuroblastoma image-defined risk factors at diagnosis. Pediatr Blood Cancer 2024; 71:e30724. [PMID: 37845799 DOI: 10.1002/pbc.30724] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 08/23/2023] [Accepted: 10/06/2023] [Indexed: 10/18/2023]
Abstract
BACKGROUND Neuroblastoma risk stratification relies on prognostic risk factors and image-defined risk factors (IDRFs). Evaluating neuroblastoma typically involves magnetic resonance imaging (MRI) with gadolinium-based contrast media (GBCM, "contrast"). However, there are concerns regarding adverse effects and cost of GBCM. We aimed to assess the impact of intravenous GBCM on interobserver agreement for neuroblastoma staging on baseline MRI. PROCEDURE We reviewed baseline MRI scans of 50 children with abdominopelvic neuroblastomas confirmed by histopathology. Duplicate sets of images were created, with post-contrast T1-weighted sequences removed from one set. Four pediatric radiologists independently analyzed the scans in a randomized manner. They recorded primary tumor size, presence of IDRFs, and metastatic lesions. Agreement among the reviewers was measured using kappa and Fleiss kappa statistics. RESULTS Mean age of included children was 3.3 years (range: 0.01-14.9 years), and 20 [40%] were females. Mean tumor size was 5.7 cm in greatest axial diameter. Pre-contrast versus post-contrast MRI showed excellent agreement for tumor measurement. Overlapping confidence intervals (CIs) were seen in nearly all categories of interobserver agreement on the presence or absence of individual IDRFs, with agreement ranging from poor to substantial, regardless of the presence of contrast. The overall interobserver agreement on the presence of at least one IDRF was substantial with contrast (kappa = .63; 95% CI: .52-.75) and moderate without contrast (kappa = .5; 95% CI: .39-.61); although the overlapping CIs suggest a lack of meaningful difference. Similarly, interobserver agreement on the presence or absence of individual sites of metastatic disease ranged from poor to substantial. The interobserver agreement on the overall determination of presence of metastatic disease was fair with contrast (kappa = .49; 95% CI: .38-.61) and moderate without contrast (kappa = .71; 95% CI: .59-.826). CONCLUSIONS Contrast does not improve tumor size measurement or radiologist agreement on the presence or absence of IDRFs or metastatic disease in children with newly diagnosed neuroblastoma.
Collapse
Affiliation(s)
- Cara E Morin
- Department of Radiology, Cincinnati Children's Hospital and University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Reem Hasweh
- Division of Radiology, Al-Balqa Applied University, Al-Salt, Jordan
| | - Chris Anton
- Department of Radiology, Cincinnati Children's Hospital and University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Jonathan R Dillman
- Department of Radiology, Cincinnati Children's Hospital and University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Emily Orscheln
- Department of Radiology, Children's Mercy Kansas City, Kansas City, Missouri, USA
| | - Ethan A Smith
- Department of Surgery, University of Cincinnati College of Medicine, Division of Pediatric and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Meera Kotagal
- Cancer and Blood Disease Institute, Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Brian D Weiss
- Department of Mathematical Sciences, University of Cincinnati, Cincinnati, Ohio, USA
| | - Jiarong Ouyang
- Division of Biostatistics and Epidemiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Bin Zhang
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Andrew T Trout
- Department of Radiology, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Alexander J Towbin
- Department of Radiology, Cincinnati Children's Hospital and University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
34
|
Abstract
Neuroblastoma is a malignant tumor of neuroblasts, immature nerve cells found in several areas of the body. It usually affects children under age of 5. As usual, the tumor has ability to grow rapidly and to expand vastly which ultimately leads to death. Mostly, management decisions can be drawn by the prediction of the stage of the disease as well as age at the time of its diagnosis. There are four main stages of neuroblastoma, and treatment is according to the low and high risk of the disease. Several cytotoxic agents along with other therapies (antibody therapy, gene therapy, and even immunological therapies, antiangiogenic therapy, etc.) are used. Immunotherapy also has an important treatment option used nowadays for neuroblastoma. The discovery of major neuroblastoma-predisposition gene anaplastic lymphoma kinase cause somatic transformation or gene strengthening in diagnosed neuroblastoma. Promising new antiangiogenic strategies have also been introduced for the treatment of neuroblastoma with multiple mylomas. To manage numerous myelomas and cancers, including neuroblastoma, bone marrow transplantation and peripheral blood stem cell transplantation may be used.
Collapse
Affiliation(s)
- Muhammad Imran Qadir
- Institute of Molecular Biology & Biotechnology, Bahauddin Zakariya University, Multan, Pakistan
| | - Bilal Ahmed
- University of Science And Technology of Fujairah, UAE; School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Sumaira Noreen
- Faculty of Pharmaceutical Sciences, Governemnet College University, Faisalabad, Pakistan
| |
Collapse
|
35
|
Zhang Y, Zhang WL, Huang DS, Wang YZ, Hu HM, Zhi T, Mei YY. Prognostic factors for intermediate- or high-risk neuroblastomas in children in China. BMC Pediatr 2023; 23:617. [PMID: 38053080 PMCID: PMC10699076 DOI: 10.1186/s12887-023-04258-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 08/21/2023] [Indexed: 12/07/2023] Open
Abstract
BACKGROUND Evidence regarding the characteristics and prognosis of neuroblastoma (NBL) in China is limited. We aimed to investigate the characteristics and prognosis of intermediate- or high-risk NBL in children in China. METHODS We included 147 patients with intermediate- or high-risk NBL evaluated from January 2006 to March 2015. The patients were aged 1 month to 15.5 years, 66% of them were boys, and 117 (79.6%) were diagnosed with high-risk NBL. RESULTS After a median follow-up of 32.5 months, 80 (45.6%) patients survived, with a median survival time of 48 months (95% confidence interval [CI]: 36.41-59.59). High-risk patients (hazard ratio [HR]: 12.467; 95% CI: 11.029-12.951), partial response (PR) (HR: 1.200; 95% CI: 1.475-2.509) or progression disease (PD) (HR: 1.924; 95% CI: 1.623-3.012) after induction chemotherapy, and intracranial metastasis (HR: 3.057; 95% CI: 0.941-4.892) were independent risk factors for survival (p < 0.05) and postrelapse survival (p < 0.05). NBL relapse, male sex, and PR or PD after induction chemotherapy were risk factors for event-free survival (p < 0.05). CONCLUSIONS In addition to previously established independent risk factors, such as age, risk group, and relapse, efficacy of induction chemotherapy and intracranial metastasis play significant roles in the prognosis of NBL.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Pediatrics, Beijing Tongren Hospital, Capital Medical University, No 2, Xihuan South Road, Daxing Zone, Beijing, China
| | - Wei-Ling Zhang
- Department of Pediatrics, Beijing Tongren Hospital, Capital Medical University, No 2, Xihuan South Road, Daxing Zone, Beijing, China
| | - Dong-Sheng Huang
- Department of Pediatrics, Beijing Tongren Hospital, Capital Medical University, No 2, Xihuan South Road, Daxing Zone, Beijing, China.
| | - Yi-Zhuo Wang
- Department of Pediatrics, Beijing Tongren Hospital, Capital Medical University, No 2, Xihuan South Road, Daxing Zone, Beijing, China
| | - Hui-Min Hu
- Department of Pediatrics, Beijing Tongren Hospital, Capital Medical University, No 2, Xihuan South Road, Daxing Zone, Beijing, China
| | - Tian Zhi
- Department of Pediatrics, Beijing Tongren Hospital, Capital Medical University, No 2, Xihuan South Road, Daxing Zone, Beijing, China
| | - Yan-Yan Mei
- Department of Pediatrics, Beijing Tongren Hospital, Capital Medical University, No 2, Xihuan South Road, Daxing Zone, Beijing, China
| |
Collapse
|
36
|
Feng L, Kan Y, Wang W, Wang C, Zhang H, Xie P, Yang J. Development and validation of a nomogram for predicting survival in intermediate- and high-risk neuroblastoma of the Children's Oncology Group risk stratification. J Cancer Res Clin Oncol 2023; 149:16377-16390. [PMID: 37702807 DOI: 10.1007/s00432-023-05398-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 09/02/2023] [Indexed: 09/14/2023]
Abstract
PURPOSE To develop and validate a nomogram for predicting survival in intermediate- and high-risk neuroblastoma patients and to compare the accuracy of the nomogram in predicting survival with Children's Oncology Group (COG) risk stratification. METHODS A total of 885 intermediate- and high-risk neuroblastoma patients were enrolled in this study, including 243 patients from our hospital (the training set) and 642 patients from the TARGET database (the validation set). The factors related to event-free survival (EFS) and overall survival (OS) in neuroblastoma were determined to construct the nomogram by Cox regression analysis. The C-index, calibration curves, and area under the time-dependent receiver operating characteristic curves (AUCs) were used to assess the predictive performance of the nomogram. RESULTS International Neuroblastoma Staging System stage and Mitosis-karyorrhexis index (MKI) were significant unfavorable factors for EFS, while MKI and MYCN status were significant unfavorable factors for OS. The C-index of the nomogram was 0.621 and 0.586 for predicting EFS, 0.650 and 0.570 for predicting OS in the training and validation sets, respectively. The calibration curves revealed good agreement in the EFS and OS predicted by the nomogram. The AUCs of the nomogram for 1-, 2-, 3-year EFS and OS were 0.633, 0.669, 0.604 and 0.672, 0.670, 0.702 in the training set, respectively. Moreover, the nomogram was able to classify patients into two groups according to risk scores, with the "high-risk" group having a lower survival rate than the "intermediate-risk" group. And the nomogram performed better than the COG risk stratification, which had a C-index of 0.537, 0.502 and 0.565, 0.572 for predicting EFS, OS in the training and validation sets, respectively. CONCLUSION We developed and validated a prognostic nomogram for intermediate- and high-risk neuroblastoma patients that clinicians can use to make more informed decisions for individual patients.
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
| | - Ying Kan
- 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
| | - Chao Wang
- SinoUnion Healthcare Inc., Beijing, 100192, China
| | - Hui Zhang
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Peng Xie
- Department of Nuclear Medicine, The Third Hospital, Hebei Medical University, Shijiazhuang, 050051, China.
| | - Jigang Yang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, 100050, China.
| |
Collapse
|
37
|
Kennedy PT, Zannoupa D, Son MH, Dahal LN, Woolley JF. Neuroblastoma: an ongoing cold front for cancer immunotherapy. J Immunother Cancer 2023; 11:e007798. [PMID: 37993280 PMCID: PMC10668262 DOI: 10.1136/jitc-2023-007798] [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] [Accepted: 10/28/2023] [Indexed: 11/24/2023] Open
Abstract
Neuroblastoma is the most frequent extracranial childhood tumour but effective treatment with current immunotherapies is challenging due to its immunosuppressive microenvironment. Efforts to date have focused on using immunotherapy to increase tumour immunogenicity and enhance anticancer immune responses, including anti-GD2 antibodies; immune checkpoint inhibitors; drugs which enhance macrophage and natural killer T (NKT) cell function; modulation of the cyclic GMP-AMP synthase-stimulator of interferon genes pathway; and engineering neuroblastoma-targeting chimeric-antigen receptor-T cells. Some of these strategies have strong preclinical foundation and are being tested clinically, although none have demonstrated notable success in treating paediatric neuroblastoma to date. Recently, approaches to overcome heterogeneity of neuroblastoma tumours and treatment resistance are being explored. These include rational combination strategies with the aim of achieving synergy, such as dual targeting of GD2 and tumour-associated macrophages or natural killer cells; GD2 and the B7-H3 immune checkpoint; GD2 and enhancer of zeste-2 methyltransferase inhibitors. Such combination strategies provide opportunities to overcome primary resistance to and maximize the benefits of immunotherapy in neuroblastoma.
Collapse
Affiliation(s)
- Paul T Kennedy
- Department of Pharmacology & Therapeutics, University of Liverpool, Liverpool, UK
| | - Demetra Zannoupa
- Department of Pharmacology & Therapeutics, University of Liverpool, Liverpool, UK
| | - Meong Hi Son
- Department of Pediatrics, Samsung Medical Center, Gangnam-gu, Seoul, Korea (the Republic of)
| | - Lekh N Dahal
- Department of Pharmacology & Therapeutics, University of Liverpool, Liverpool, UK
| | - John F Woolley
- Department of Pharmacology & Therapeutics, University of Liverpool, Liverpool, UK
| |
Collapse
|
38
|
Feng L, Zhang S, Wang C, Li S, Kan Y, Wang C, Zhang H, Wang W, Yang J. Axial Skeleton Radiomics of 18F-FDG PET/CT: Impact on Event-Free Survival Prediction in High-Risk Pediatric Neuroblastoma. Acad Radiol 2023; 30:2487-2496. [PMID: 36828720 DOI: 10.1016/j.acra.2023.01.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/24/2023] [Accepted: 01/24/2023] [Indexed: 02/25/2023]
Abstract
OBJECTIVES To construct and validate a combined model based on axial skeleton radiomics of 18F-FDG PET/CT for predicting event-free survival in high-risk pediatric neuroblastoma patients. MATERIALS AND METHODS Eighty-seven high-risk neuroblastoma patients were retrospectively enrolled in this study and randomized in a 7:3 ratio to the training and validation cohorts. The radiomics model was constructed using radiomics features that were extracted from the axial skeleton. A univariate Cox regression analysis was then performed to screen clinical risk factors associated with event-free survival for building clinical model. Radiomics features and clinical risk factors were incorporated to construct the combined model for predicting the event-free survival in high-risk neuroblastoma patients. The performance of the models was evaluated by the C-index. RESULTS Eighteen radiomics features were selected to build the radiomics model. The radiomics model achieved better event-free survival prediction than the clinical model in the training cohort (C-index: 0.846 vs. 0.612) and validation cohort (C-index: 0.754 vs. 0.579). The combined model achieved the best prognostic prediction performance with a C-index of 0.863 and 0.799 in the training and validation cohorts, respectively. CONCLUSION The combined model integrating radiomics features and clinical risk factors showed more accurate predictive performance for event-free survival in high-risk pediatric neuroblastoma patients, which helps to design individualized treatment strategies and regular follow-ups.
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
| | - Shuxin Zhang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China
| | - Chaoran Wang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China
| | - Siqi Li
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China
| | - Ying Kan
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China
| | - Chao Wang
- SinoUnion Healthcare Inc., Beijing, China
| | - Hui Zhang
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 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
|
39
|
Cheng H, Zhang L, Yang S, Ren Q, Chang S, Jin Y, Mou W, Qin H, Yang W, Zhang X, Zhang W, Wang H. Integration of clinical characteristics and molecular signatures of the tumor microenvironment to predict the prognosis of neuroblastoma. J Mol Med (Berl) 2023; 101:1421-1436. [PMID: 37712965 DOI: 10.1007/s00109-023-02372-x] [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: 04/10/2023] [Revised: 08/26/2023] [Accepted: 09/04/2023] [Indexed: 09/16/2023]
Abstract
This study aimed to analyze the clinical characteristics, cell types, and molecular characteristics of the tumor microenvironment to better predict the prognosis of neuroblastoma (NB). The gene expression data and corresponding clinical information of 498 NB patients were obtained from the Gene Expression Omnibus (GEO: GSE62564) and ArrayExpress (accession: E-MTAB-8248). The relative cell abundances were estimated using single-sample gene set enrichment analysis (ssGSEA) with the R gene set variation analysis (GSVA) package. We performed Cox regression analyses to identify marker genes indicating cell subsets and combined these with prognostically relevant clinical factors to develop a new prognostic model. Data from the E-MTAB-8248 cohort verified the predictive accuracy of the prognostic model. Single-cell RNA-seq data were analyzed by using the R Seurat package. Multivariate survival analysis for each gene, using clinical characteristics as cofactors, identified 34 prognostic genes that showed a significant correlation with both event-free survival (EFS) and overall survival (OS) (log-rank test, P value < 0.05). The pathway enrichment analysis revealed that these prognostic genes were highly enriched in the marker genes of NB cells with mesenchymal features and protein translation. Ultimately, USP39, RPL8, IL1RAPL1, MAST4, CSRP2, ATP5E, International Neuroblastoma Staging System (INSS) stage, age, and MYCN status were selected to build an optimized Cox model for NB risk stratification. These samples were divided into two groups using the median of the risk score as a cutoff. The prognosis of samples in the poor prognosis group (PP) was significantly worse than that of samples in the good prognosis group (GP) (log-rank test, P value < 0.0001, median EFS: 640.5 vs. 2247 days, median OS: 1279.5 vs. 2519 days). The risk model was also regarded as a prognostic indicator independent of MYCN status, age, and stage. Finally, through scRNA-seq data, we found that as an important prognostic marker, USP39 might participate in the regulation of RNA splicing in NB. Our study established a multivariate Cox model based on gene signatures and clinical characteristics to better predict the prognosis of NB and revealed that mesenchymal signature genes of NB cells, especially USP39, were more abundant in patients with a poor prognosis than in those with a good prognosis. KEY MESSAGES: Our study established a multivariate Cox model based on gene signatures and clinical characteristics to better predict the prognosis of NB and revealed that mesenchymal signature genes of NB cells, especially USP39, were more abundant in patients with a poor prognosis than in those with a good prognosis. USP39, RPL8, IL1RAPL1, MAST4, CSRP2, ATP5E, International Neuroblastoma Staging System (INSS) stage, age, and MYCN status were selected to build an optimized Cox model for NB risk stratification. These samples were divided into two groups using the median of the risk score as a cutoff. The prognosis of samples in the poor prognosis group (PP) was significantly worse than that of samples in the good prognosis group (GP). Finally, through scRNA-seq data, we found that as an important prognostic marker, USP39 might participate in the regulation of RNA splicing in NB.
Collapse
Affiliation(s)
- Haiyan Cheng
- Department of Surgical Oncology, MOE Key Laboratory of Major Diseases in Children, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 56 Nanlishi Road, Beijing, 100045, China
| | - Li Zhang
- Shanghai Institute of Precision Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Shen Yang
- Department of Surgical Oncology, MOE Key Laboratory of Major Diseases in Children, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 56 Nanlishi Road, Beijing, 100045, China
| | - Qinghua Ren
- Department of Surgical Oncology, MOE Key Laboratory of Major Diseases in Children, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 56 Nanlishi Road, Beijing, 100045, China
| | - Saishuo Chang
- Department of Surgical Oncology, MOE Key Laboratory of Major Diseases in Children, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 56 Nanlishi Road, 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
| | - Wenjun Mou
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Hong Qin
- Department of Surgical Oncology, MOE Key Laboratory of Major Diseases in Children, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 56 Nanlishi Road, Beijing, 100045, China
| | - Wei Yang
- Department of Surgical Oncology, MOE Key Laboratory of Major Diseases in Children, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 56 Nanlishi Road, Beijing, 100045, China
| | - Xianwei Zhang
- Zhengzhou Key Laboratory of Precise Diagnosis and Treatment of Children's Malignant Tumors, Department of Pediatric Oncology Surgery, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Wancun Zhang
- Zhengzhou Key Laboratory of Precise Diagnosis and Treatment of Children's Malignant Tumors, Department of Pediatric Oncology Surgery, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Huanmin Wang
- Department of Surgical Oncology, MOE Key Laboratory of Major Diseases in Children, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 56 Nanlishi Road, Beijing, 100045, China.
| |
Collapse
|
40
|
Valenti GE, Marengo B, Milanese M, Zuccari G, Brullo C, Domenicotti C, Alfei S. Imidazo-Pyrazole-Loaded Palmitic Acid and Polystyrene-Based Nanoparticles: Synthesis, Characterization and Antiproliferative Activity on Chemo-Resistant Human Neuroblastoma Cells. Int J Mol Sci 2023; 24:15027. [PMID: 37834475 PMCID: PMC10573130 DOI: 10.3390/ijms241915027] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Neuroblastoma (NB) is a childhood cancer, commonly treated with drugs, such as etoposide (ETO), whose efficacy is limited by the onset of resistance. Here, aiming at identifying new treatments for chemo-resistant NB, the effects of two synthesized imidazo-pyrazoles (IMPs) (4G and 4I) were investigated on ETO-sensitive (HTLA-230) and ETO-resistant (HTLA-ER) NB cells, detecting 4I as the more promising compound, that demonstrated IC50 values lower than those of ETO on HTLA ER. Therefore, to further improve the activity of 4I, we developed 4I-loaded palmitic acid (PA) and polystyrene-based (P5) cationic nanoparticles (P5PA-4I NPs) with high drug loading (21%) and encapsulation efficiency (97%), by a single oil-in-water emulsification technique. Biocompatible PA was adopted as an emulsion stabilizer, while synthesized P5 acted as an encapsulating agent, solubilizer and hydrophilic-lipophilic balance (HLB) improver. Optic microscopy and cytofluorimetric analyses were performed to investigate the micromorphology, size and complexity distributions of P5PA-4I NPs, which were also structurally characterized by chemometric-assisted Fourier transform infrared spectroscopy (FTIR). Potentiometric titrations allowed us to estimate the milliequivalents of PA and basic nitrogen atoms present in NPs. P5PA-4I NPs afforded dispersions in water with excellent buffer capacity, essential to escape lysosomal degradation and promote long residence time inside cells. They were chemically stable in an aqueous medium for at least 40 days, while in dynamic light scattering (DLS) analyses, P5PA-4I showed a mean hydrodynamic diameter of 541 nm, small polydispersity (0.194), and low positive zeta potentials (+8.39 mV), assuring low haemolytic toxicity. Biological experiments on NB cells, demonstrated that P5PA-4I NPs induced ROS-dependent cytotoxic effects significantly higher than those of pristine 4I, showing a major efficacy compared to ETO in reducing cell viability in HTLA-ER cells. Collectively, this 4I-based nano-formulation could represent a new promising macromolecular platform to develop a new delivery system able to increase the cytotoxicity of the anticancer drugs.
Collapse
Affiliation(s)
- Giulia Elda Valenti
- Department of Experimental Medicine (DIMES), University of Genova, Via Alberti L.B., 16132 Genoa, Italy; (G.E.V.); (B.M.)
| | - Barbara Marengo
- Department of Experimental Medicine (DIMES), University of Genova, Via Alberti L.B., 16132 Genoa, Italy; (G.E.V.); (B.M.)
| | - Marco Milanese
- Department of Pharmacy, Section of Chemistry and Pharmaceutical and Food Technologies, University of Genoa, Viale Cembrano, 4, 16148 Genoa, Italy;
| | - Guendalina Zuccari
- Department of Pharmacy (DIFAR), Section of Medicinal Chemistry and Cosmetic Product, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (G.Z.); (C.B.)
| | - Chiara Brullo
- Department of Pharmacy (DIFAR), Section of Medicinal Chemistry and Cosmetic Product, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (G.Z.); (C.B.)
| | - Cinzia Domenicotti
- Department of Experimental Medicine (DIMES), University of Genova, Via Alberti L.B., 16132 Genoa, Italy; (G.E.V.); (B.M.)
| | - Silvana Alfei
- Department of Pharmacy, Section of Chemistry and Pharmaceutical and Food Technologies, University of Genoa, Viale Cembrano, 4, 16148 Genoa, Italy;
| |
Collapse
|
41
|
Ghosh A, Yekeler E, Teixeira SR, Dalal D, States L. Role of MRI radiomics for the prediction of MYCN amplification in neuroblastomas. Eur Radiol 2023; 33:6726-6735. [PMID: 37178203 DOI: 10.1007/s00330-023-09628-7] [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: 08/31/2022] [Revised: 02/18/2023] [Accepted: 02/26/2023] [Indexed: 05/15/2023]
Abstract
OBJECTIVES We evaluate MR radiomics and develop machine learning-based classifiers to predict MYCN amplification in neuroblastomas. METHODS A total of 120 patients with neuroblastomas and baseline MR imaging examination available were identified of whom 74 (mean age ± standard deviation [SD] of 6 years and 2 months ± 4 years and 9 months; 43 females and 31 males, 14 MYCN amplified) underwent imaging at our institution. This was therefore used to develop radiomics models. The model was tested in a cohort of children with the same diagnosis but imaged elsewhere (n = 46, mean age ± SD: 5 years 11 months ± 3 years 9 months, 26 females and 14 MYCN amplified). Whole tumour volumes of interest were adopted to extract first-order histogram and second-order radiomics features. Interclass correlation coefficient and maximum relevance and minimum redundancy algorithm were applied for feature selection. Logistic regression, support vector machine, and random forest were employed as the classifiers. Receiver operating characteristic (ROC) analysis was performed to evaluate the diagnostic accuracy of the classifiers on the external test set. RESULTS The logistic regression model and the random forest both showed an AUC of 0.75. The support vector machine classifier obtained an AUC of 0.78 on the test set with a sensitivity of 64% and a specificity of 72%. CONCLUSION The study provides preliminary retrospective evidence demonstrating the feasibility of MRI radiomics in predicting MYCN amplification in neuroblastomas. Future studies are needed to explore the correlation between other imaging features and genetic markers and to develop multiclass predictive models. KEY POINTS • MYCN amplification in neuroblastomas is an important determinant of disease prognosis. • Radiomics analysis of pre-treatment MR examinations can be used to predict MYCN amplification in neuroblastomas. • Radiomics machine learning models showed good generalisability to external test set, demonstrating reproducibility of the computational models.
Collapse
Affiliation(s)
- Adarsh Ghosh
- Department of Radiology, Cincinnati Children's Hospital and Medical Centre, Cincinnati, OH, USA.
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| | - Ensar Yekeler
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sara Reis Teixeira
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Deepa Dalal
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lisa States
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
42
|
Kerkar AN, Chinnam D, Verma A, Peters NJ, Kakkar N, Trehan A, Singh M, Gupta K. MYCN amplification, TERT rearrangements and ATRX mutations in neuroblastoma: clinicopathological correlates- an Indian perspective. Virchows Arch 2023; 483:477-486. [PMID: 37460674 DOI: 10.1007/s00428-023-03604-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 06/14/2023] [Accepted: 07/11/2023] [Indexed: 10/28/2023]
Abstract
BACKGROUND Neuroblastoma (NB) is the most common extracranial solid tumour in childhood with a diverse clinical presentation and course. The early age of onset, high frequency of metastatic disease at diagnosis and tendency for spontaneous regression in infancy sets it apart from other childhood tumors. This heterogeneity is largely attributed to underlying genetic aberrations which are distinct in low-risk and high-risk NB. To this end, we sought to analyse our NB cases for the molecular alterations and find its correlation with clinical behaviour. METHODS NB cases (n = 50) diagnosed over last 7 years were retrospectively analysed for MYCN amplification (fluorescent-in-situ hybridization), TERT rearrangements (qRT-PCR), ATRX mutations (immunohistochemistry). These findings were correlated with demographic profiles, histologic features and clinical outcome. RESULTS Age ranged from 1 month to 30 years (mean 2.8 years) with male preponderance. Poorly differentiated subtype constituted the majority (64%), followed by differentiating (28%) and undifferentiated subtype (8%) which were equally distributed across all age groups. MYCN amplification, TERT-mRNA upregulation and ATRX mutations was observed in 30%, 42% and 24%, respectively. Cases with TERT-mRNA upregulation were distributed equally across all histological subtypes while those with ATRX mutations and MYCN amplification were frequent in poorly differentiated NB. ATRX mutation was mutually exclusive of TERT-mRNA upregulation and MYCN amplification. Kaplan-Meier analysis revealed significantly shorter overall and progression-free survival for tumors harboring MYCN amplification and TERT-mRNA upregulation, while that for ATRX mutant tumors was not significant. CONCLUSIONS Our results provide data indicating poor clinical outcome in NB carrying MYCN amplification and TERT-mRNA upregulation.
Collapse
Affiliation(s)
- Aadya N Kerkar
- Department of Pathology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | | | - Aanchal Verma
- Department of Histopathology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Nitin J Peters
- Department of Pediatric Surgery, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Nandita Kakkar
- Department of Histopathology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Amita Trehan
- Department of Pediatrics, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Minu Singh
- Department of Pediatrics, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Kirti Gupta
- Department of Histopathology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India.
| |
Collapse
|
43
|
Li M, Duan X, Li C, You D, Liu L. A novel clinical tool and risk stratification system for predicting the event-free survival of neuroblastoma patients: A TARGET-based study. Medicine (Baltimore) 2023; 102:e34925. [PMID: 37746942 PMCID: PMC10519501 DOI: 10.1097/md.0000000000034925] [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: 04/16/2023] [Revised: 07/11/2023] [Accepted: 08/03/2023] [Indexed: 09/26/2023] Open
Abstract
Neuroblastoma (NB), considered the most common non-intracranial solid tumor in children, accounts for nearly 8% of pediatric malignancies. This study aimed to develop a simple and practical nomogram to predict event-free survival (EFS) in NB patients and establish a new risk stratification system. In this study, 763 patients primarily diagnosed with NB in the Therapeutically Applicable Research to Generate Effective Treatments (TARGET) database were included and randomly assigned to a training set (70%) and a validation set (30%) in a 7:3 ratio. First, the independent prognostic factors of EFS for NB patients were identified through univariate and multivariate Cox regression analyses. Second, a nomogram was created based on these factors and was validated for calibration capability, discriminative, and clinical significance by C-curves, receiver operating characteristic (ROC) curves, and decision curve analysis. Finally, a new risk stratification system was established for NB patients based on the nomogram. The univariate Cox analysis demonstrated that NB patients with age at diagnosis >318 days, International Neuroblastoma Staging System (INSS) stage 4, DNA diploidy, MYCN amplification status, and children oncology group (COG) high-risk group had a relatively poor prognosis. However, according to the multivariate Cox regression analysis, only age, INSS stage, and DNA ploidy were independent predictive factors in NB patients regarding EFS, and a nomogram was created based on these factors. The area under the curve (AUC) values of the ROC curves for the 3-, 5-, and 10-year EFS of this nomogram were 0.681, 0.706, and 0.720, respectively. Additionally, the AUC values of individual independent prognostic factors of EFS were lower than those of the nomogram, suggesting that the developed nomogram had a higher predictive reliability for prognosis. In addition, a new risk stratification system was developed to better stratify NB patients and provide clinical practitioners with a better reference for clinical decision-making. NB patients' EFS could be predicted more accurately and easily through the constructed nomogram and event-occurrence risk stratification system, allowing clinicians to better differentiate NB patients and establish individualized treatment plans to maximize patient benefits.
Collapse
Affiliation(s)
- Mingzhen Li
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Nanguan District, Changchun, Jilin, People’s Republic of China
| | - Xiaoying Duan
- Department of Acupuncture and moxibustion, Second Hospital of Jilin University, Nanguan District, Changchun, Jilin, People’s Republic of China
| | - Chunyan Li
- Department of Endocrinology, The Affiliated Hospital of Beihua University, Chuanying District, Jilin, People’s Republic of China
| | - Di You
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Nanguan District, Changchun, Jilin, People’s Republic of China
| | - Linlin Liu
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Nanguan District, Changchun, Jilin, People’s Republic of China
| |
Collapse
|
44
|
Deng Z, Richardson DR. The Myc Family and the Metastasis Suppressor NDRG1: Targeting Key Molecular Interactions with Innovative Therapeutics. Pharmacol Rev 2023; 75:1007-1035. [PMID: 37280098 DOI: 10.1124/pharmrev.122.000795] [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: 12/06/2022] [Revised: 03/07/2023] [Accepted: 05/01/2023] [Indexed: 06/08/2023] Open
Abstract
Cancer is a leading cause of death worldwide, resulting in ∼10 million deaths in 2020. Major oncogenic effectors are the Myc proto-oncogene family, which consists of three members including c-Myc, N-Myc, and L-Myc. As a pertinent example of the role of the Myc family in tumorigenesis, amplification of MYCN in childhood neuroblastoma strongly correlates with poor patient prognosis. Complexes between Myc oncoproteins and their partners such as hypoxia-inducible factor-1α and Myc-associated protein X (MAX) result in proliferation arrest and pro-proliferative effects, respectively. Interactions with other proteins are also important for N-Myc activity. For instance, the enhancer of zest homolog 2 (EZH2) binds directly to N-Myc to stabilize it by acting as a competitor against the ubiquitin ligase, SCFFBXW7, which prevents proteasomal degradation. Heat shock protein 90 may also be involved in N-Myc stabilization since it binds to EZH2 and prevents its degradation. N-Myc downstream-regulated gene 1 (NDRG1) is downregulated by N-Myc and participates in the regulation of cellular proliferation via associating with other proteins, such as glycogen synthase kinase-3β and low-density lipoprotein receptor-related protein 6. These molecular interactions provide a better understanding of the biologic roles of N-Myc and NDRG1, which can be potentially used as therapeutic targets. In addition to directly targeting these proteins, disrupting their key interactions may also be a promising strategy for anti-cancer drug development. This review examines the interactions between the Myc proteins and other molecules, with a special focus on the relationship between N-Myc and NDRG1 and possible therapeutic interventions. SIGNIFICANCE STATEMENT: Neuroblastoma is one of the most common childhood solid tumors, with a dismal five-year survival rate. This problem makes it imperative to discover new and more effective therapeutics. The molecular interactions between major oncogenic drivers of the Myc family and other key proteins; for example, the metastasis suppressor, NDRG1, may potentially be used as targets for anti-neuroblastoma drug development. In addition to directly targeting these proteins, disrupting their key molecular interactions may also be promising for drug discovery.
Collapse
Affiliation(s)
- Zhao Deng
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Australia (Z.D., D.R.R.), and Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan (D.R.R.)
| | - Des R Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Australia (Z.D., D.R.R.), and Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan (D.R.R.)
| |
Collapse
|
45
|
Wu R, Li X, Chen Z, Shao Q, Zhang X, Tang W, Hu B. Development and validation of a nomogram based on common biochemical indicators for survival prediction of children with high-risk neuroblastoma: A valuable tool for resource-limited hospitals. BMC Pediatr 2023; 23:426. [PMID: 37633889 PMCID: PMC10463855 DOI: 10.1186/s12887-023-04228-2] [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: 04/13/2023] [Accepted: 08/04/2023] [Indexed: 08/28/2023] Open
Abstract
BACKGROUND Despite multiple attempts have been made to develop risk stratification within high-risk neuroblastoma (NB) patients (age of diagnosis ≥ 18 month-old with metastatic NB), the definition of "ultra high-risk NB" is still lack of consensus, and indicators for identifying this subgroup are still unclear. This study aimed to develop a nomogram based on easy-to-obtain blood-derived biofactors for identifying ultra high-risk NB patients with highest risk of death within 3 or 5 years. METHODS One hundred sixty-seven NB patients who treated at Sun Yat-sen University Cancer Center between 2015 and 2023 were recruited and clustered randomly into training and validation cohorts (116 and 51 cases, respectively). Univariate and multivariate Cox analysis were performed in training set to screen independent prognostic indicators for constructing nomogram model of predicting 1-, 3- and 5-year overall survival (OS). The discrimination power of the nomogram in training and validation sets were assessed by concordance index (C-index) and calibration plot. Based on the risk score obtained from nomogram model, the prognostic accuracy of 1-, 3- and 5-year OS rates in training and validation cohorts were further evaluated using the area under receiver operating characteristic (ROC) curves (AUC). RESULTS Through univariate and multivariate Cox analysis, independent prognostic indicators, including serum lactate dehydrogenase (LDH) and albumin (ALB), were identified in training set, and used to establish a nomogram model. The model showed good discrimination power with C-index in training cohort being 0.706 (95%CI: 0.633-0.788). According to the cut-point calculated based on the established nomogram, patients with a nomogram score > 34 points could be stratified to ultra high-risk NB subgroup, and this subgroup had poorer OS than those in non-ultra one (p < 0.001). AUC values of ROC curves for 3- and 5-year OS rates in the training set were 0.758 and 0.756, respectively. Moreover, based on the cut-point score (34 points) developed in training set, The model also showed good discrimination power with C-index of 0.773 (95%CI: 0.664-0.897) and powerful prognostic accuracy of AUC for 3- and 5-year OS rates being 0.825 and 0.826, respectively, in validation cohort. CONCLUSIONS We developed a simple-to-use nomogram based on common laboratory indicators to identify the subgroup of ultra high-risk NB before treatment, providing these children even from developing countries or regions access to intensified multimodal treatments earlier and thus improving their long-term outcome.
Collapse
Affiliation(s)
- Ruohao Wu
- Department of Pediatrics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, China
| | - Xiaohui Li
- Department of Laboratory Medicine, Sun Yat-Sen University Cancer Center, Sun Yat-Sen University, Guangzhou, 510060, Guangdong, China
| | - Zhishan Chen
- Department of Pathology, Panyu District Central Hospital, Guangzhou, 511400, Guangdong, China
| | - Qiong Shao
- Department of Research and Molecular Diagnostics, Sun Yat-Sen University Cancer Center, Sun Yat-Sen University, Guangzhou, 510060, Guangdong, China
| | - Xiao Zhang
- Department of Research and Molecular Diagnostics, Sun Yat-Sen University Cancer Center, Sun Yat-Sen University, Guangzhou, 510060, Guangdong, China
| | - Wenting Tang
- Department of Research and Molecular Diagnostics, Sun Yat-Sen University Cancer Center, Sun Yat-Sen University, Guangzhou, 510060, Guangdong, China.
| | - Bo Hu
- Department of Laboratory Medicine, Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, Guangdong, China.
| |
Collapse
|
46
|
Hu J, Song F, Kang W, Xia F, Song Z, Wang Y, Li J, Zhao Q. Integrative analysis of multi-omics data for discovery of ferroptosis-related gene signature predicting immune activity in neuroblastoma. Front Pharmacol 2023; 14:1162563. [PMID: 37521469 PMCID: PMC10373597 DOI: 10.3389/fphar.2023.1162563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 06/02/2023] [Indexed: 08/01/2023] Open
Abstract
Immunotherapy for neuroblastoma remains unsatisfactory due to heterogeneity and weak immunogenicity. Exploring powerful signatures for the evaluation of immunotherapy outcomes remain the primary purpose. We constructed a ferroptosis-related gene (FRG) signature by least absolute shrinkage and selection operator and Cox regression, identified 10 independent prognostic FRGs in a training cohort (GSE62564), and then verified them in an external validation cohort (TCGA). Associated with clinical factors, the signature accurately predicts overall survival of 3, 5, and 10 years. An independent prognostic nomogram, which included FRG risk, age, stage of the International Neuroblastoma Staging System, and an MYCN status, was constructed. The area under the curves showed satisfactory prognostic predicting performance. Through bulk RNA-seq and proteomics data, we revealed the relationship between hub genes and the key onco-promoter MYCN gene and then validated the results in MYCN-amplified and MYCN-non-amplified cell lines with qRT-PCR. The FRG signature significantly divided patients into high- and low-risk groups, and the differentially expressed genes between the two groups were enriched in immune actions, autophagy, and carcinogenesis behaviors. The low-risk group embodied higher positive immune component infiltration and a higher expression of immune checkpoints with a more favorable immune cytolytic activity (CYT). We verified the predictive power of this signature with data from melanoma patients undergoing immunotherapy, and the predictive power was satisfactory. Gene mutations were closely related to the signature and prognosis. AURKA and PRKAA2 were revealed to be nodal hub FRGs in the signature, and both were shown to have significantly different expressions between the INSS stage IV and other stages after immunohistochemical validation. With single-cell RNA-seq analysis, we found that genes related to T cells were enriched in TNFA signaling and interferon-γ hallmark. In conclusion, we constructed a ferroptosis-related gene signature that can predict the outcomes and work in evaluating the effects of immunotherapy.
Collapse
Affiliation(s)
- Jiajian Hu
- Tianjin Key Laboratory of Cancer Prevention and Therapy, Department of Pediatric Oncology, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Fengju Song
- Key Laboratory of Molecular Cancer Epidemiology, Department of Epidemiology and Biostatistics, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Wenjuan Kang
- Key Laboratory of Molecular Cancer Epidemiology, Department of Epidemiology and Biostatistics, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Fantong Xia
- Tianjin Key Laboratory of Cancer Prevention and Therapy, Department of Pediatric Oncology, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Zi’an Song
- Tianjin Key Laboratory of Cancer Prevention and Therapy, Department of Pediatric Oncology, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yangyang Wang
- Tianjin Key Laboratory of Cancer Prevention and Therapy, Department of Pediatric Oncology, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jie Li
- Tianjin Key Laboratory of Cancer Prevention and Therapy, Department of Pediatric Oncology, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Qiang Zhao
- Tianjin Key Laboratory of Cancer Prevention and Therapy, Department of Pediatric Oncology, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
47
|
Kato S. Lactoferrin inhibits the proliferation of IMR‑32 neuroblastoma cells even under X‑rays. MEDICINE INTERNATIONAL 2023; 3:33. [PMID: 37448769 PMCID: PMC10336960 DOI: 10.3892/mi.2023.93] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023]
Abstract
Neuroblastoma is a typical solid tumor common in childhood. The present study investigated the inhibitory effects of lactoferrin on the proliferation of IMR-32 neuroblastoma cells, including under X-ray irradiation. In controlled in vitro assays, it was found that lactoferrin inhibited cell proliferation, accompanied by cell membrane disruption. Furthermore, intracellular reactive oxygen species generation increased in IMR-32 cells treated with lactoferrin, causing membrane lipid peroxidation and the leakage of lactate dehydrogenase. The IC50 values for cell proliferation were ~2.0 nM for doxorubicin, 2.7 mM for dibutyryl-cAMP and 45.9 µM for lactoferrin. X-ray irradiation at 1 Gy decreased cell proliferation to ~30%, which was not restored by lactoferrin. In the Fenton reaction system with iron chloride, lactoferrin increased hydroxyl radical (OH·) formation via H2O2, as confirmed by electron spin resonance spectra. On the whole, the findings of the present study indicate that lactoferrin, found abundantly in milk, may help prevent or treat neuroblastoma in infants with modest efficacy, and does not exert a protective effect against X-rays.
Collapse
Affiliation(s)
- Shinya Kato
- Radioisotope Experimental Facility, Advanced Science Research Promotion Center, Mie University, Tsu, Mie 514-8507, Japan
| |
Collapse
|
48
|
Bender HG, Irwin MS, Hogarty MD, Castleberry R, Maris JM, Kao PC, Zhang FF, Naranjo A, Cohn SL, London WB. Survival of Patients With Neuroblastoma After Assignment to Reduced Therapy Because of the 12- to 18-Month Change in Age Cutoff in Children's Oncology Group Risk Stratification. J Clin Oncol 2023; 41:3149-3159. [PMID: 37098238 PMCID: PMC10256433 DOI: 10.1200/jco.22.01946] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 12/01/2022] [Accepted: 02/23/2023] [Indexed: 04/27/2023] Open
Abstract
PURPOSE In 2006, Children's Oncology Group (COG) reclassified subgroups of toddlers diagnosed with neuroblastoma from high-risk to intermediate-risk, when the age cutoff for high-risk assignment was raised from 365 days (12 months) to 547 days (18 months). The primary aim of this retrospective study was to determine if excellent outcome was maintained after assigned reduction of therapy. PATIENTS AND METHODS Children <3 years old at diagnosis, enrolled on a COG biology study from 1990 to 2018, were eligible (n = 9,189). Assigned therapy was reduced for two cohorts of interest on the basis of the age cutoff change: 365-546 days old with International Neuroblastoma Staging System (INSS) stage 4, MYCN not amplified (MYCN-NA), favorable International Neuroblastoma Pathology Classification (INPC), hyperdiploid tumors (12-18mo/Stage4/FavBiology), and 365-546 days old with INSS stage 3, MYCN-NA, and unfavorable INPC tumors (12-18mo/Stage3/MYCN-NA/Unfav). Log-rank tests compared event-free survival (EFS) and overall survival (OS) curves. RESULTS For 12-18mo/Stage4/FavBiology, 5-year EFS/OS (± SE) before (≤2006; n = 40) versus after (>2006; n = 55) assigned reduction in therapy was similar: 89% ± 5.1%/89% ± 5.1% versus 87% ± 4.6%/94% ± 3.2% (P = .7; P = .4, respectively). For 12-18mo/Stage3/MYCN-NA/Unfav, the 5-year EFS and OS were both 100%, before (n = 6) and after (n = 4) 2006. The 12-18mo/Stage4/FavBiology plus 12-18mo/Stage3/MYCN-NA/Unfav classified as high-risk ≤2006 had an EFS/OS of 91% ± 4.4%/91% ± 4.5% versus 38% ± 1.3%/43% ± 1.3% for all other high-risk patients <3 years old (P < .0001; P < .0001, respectively). The 12-18mo/Stage4/FavBiology plus 12-18mo/Stage3/MYCN-NA/Unfav classified as intermediate-risk >2006 had an EFS/OS of 88% ± 4.3%/95% ± 2.9% versus 88% ± 0.9%/95% ± 0.6% for all other intermediate-risk patients <3 years old (P = .87; P = .85, respectively). CONCLUSION Excellent outcome was maintained among subsets of toddlers with neuroblastoma assigned to reduced treatment after reclassification of risk group from high to intermediate on the basis of new age cutoffs. Importantly, as documented in prior trials, intermediate-risk therapy is not associated with the degree of acute toxicity and late effects commonly observed with high-risk regimens.
Collapse
Affiliation(s)
- Hannah G. Bender
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Meredith S. Irwin
- Department of Pediatrics, The Hospital for Sick Children, University of Toronto, ON, Canada
| | - Michael D. Hogarty
- Department of Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | | | - John M. Maris
- Department of Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Pei-Chi Kao
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Fan F. Zhang
- Department of Biostatistics, Children's Oncology Group Statistics and Data Center, University of Florida, Gainesville, FL
| | - Arlene Naranjo
- Department of Biostatistics, Children's Oncology Group Statistics and Data Center, University of Florida, Gainesville, FL
| | - Susan L. Cohn
- Department of Pediatrics and Comer Children's Hospital, University of Chicago, Chicago, IL
| | - Wendy B. London
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| |
Collapse
|
49
|
Slattery SM, Perez IA, Ceccherini I, Chen ML, Kurek KC, Yap KL, Keens TG, Khaytin I, Ballard HA, Sokol EA, Mittal A, Rand CM, Weese-Mayer DE. Transitional care and clinical management of adolescents, young adults, and suspected new adult patients with congenital central hypoventilation syndrome. Clin Auton Res 2023; 33:231-249. [PMID: 36403185 DOI: 10.1007/s10286-022-00908-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 10/31/2022] [Indexed: 11/21/2022]
Abstract
PURPOSE With contemporaneous advances in congenital central hypoventilation syndrome (CCHS), recognition, confirmatory diagnostics with PHOX2B genetic testing, and conservative management to reduce the risk of early morbidity and mortality, the prevalence of identified adolescents and young adults with CCHS and later-onset (LO-) CCHS has increased. Accordingly, there is heightened awareness and need for transitional care of these patients from pediatric medicine into a multidisciplinary adult medical team. Hence, this review summarizes key clinical and management considerations for patients with CCHS and LO-CCHS and emphasizes topics of particular importance for this demographic. METHODS We performed a systematic review of literature on diagnostics, pathophysiology, and clinical management in CCHS and LO-CCHS, and supplemented the review with anecdotal but extensive experiences from large academic pediatric centers with expertise in CCHS. RESULTS We summarized our findings topically for an overview of the medical care in CCHS and LO-CCHS specifically applicable to adolescents and adults. Care topics include genetic and embryologic basis of the disease, clinical presentation, management, variability in autonomic nervous system dysfunction, and clarity regarding transitional care with unique considerations such as living independently, family planning, exposure to anesthesia, and alcohol and drug use. CONCLUSIONS While a lack of experience and evidence exists in the care of adults with CCHS and LO-CCHS, a review of the relevant literature and expert consensus provides guidance for transitional care areas.
Collapse
Affiliation(s)
- Susan M Slattery
- Center for Autonomic Medicine in Pediatrics (CAMP), Division of Autonomic Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago and Stanley Manne Children's Research Center, 225 E. Chicago Ave, Box #165, Chicago, IL, 60611, USA.
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | - Iris A Perez
- Division of Pediatric Pulmonology and Sleep Medicine, Children's Hospital Los Angeles, Los Angeles, CA, USA
- Department of Pediatrics, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - Isabella Ceccherini
- Laboratory of Genetics and Genomics of Rare Diseases, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Maida L Chen
- Division of Pulmonary and Sleep Medicine, Seattle Children's Hospital, Seattle, WA, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Kyle C Kurek
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada
| | - Kai Lee Yap
- Molecular Diagnostics Laboratory, Department of Pathology & Laboratory Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Thomas G Keens
- Division of Pediatric Pulmonology and Sleep Medicine, Children's Hospital Los Angeles, Los Angeles, CA, USA
- Department of Pediatrics, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - Ilya Khaytin
- Center for Autonomic Medicine in Pediatrics (CAMP), Division of Autonomic Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago and Stanley Manne Children's Research Center, 225 E. Chicago Ave, Box #165, Chicago, IL, 60611, USA
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Heather A Ballard
- Department of Pediatric Anesthesiology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Department of Anesthesia, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Elizabeth A Sokol
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Division of Hematology/Oncology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Angeli Mittal
- Center for Autonomic Medicine in Pediatrics (CAMP), Division of Autonomic Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago and Stanley Manne Children's Research Center, 225 E. Chicago Ave, Box #165, Chicago, IL, 60611, USA
| | - Casey M Rand
- Center for Autonomic Medicine in Pediatrics (CAMP), Division of Autonomic Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago and Stanley Manne Children's Research Center, 225 E. Chicago Ave, Box #165, Chicago, IL, 60611, USA
| | - Debra E Weese-Mayer
- Center for Autonomic Medicine in Pediatrics (CAMP), Division of Autonomic Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago and Stanley Manne Children's Research Center, 225 E. Chicago Ave, Box #165, Chicago, IL, 60611, USA
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
50
|
Aschero R, Castillo-Ecija H, Baulenas-Farres M, Resa-Pares C, Jimenez-Cabaco A, Rodriguez E, Monterrubio C, Perez-Jaume S, Suñol M, Chantada GL, Lavarino C, Mora J, Carcaboso AM. Prognostic value of xenograft engraftment in patients with metastatic high-risk neuroblastoma. Pediatr Blood Cancer 2023; 70:e30318. [PMID: 36973999 DOI: 10.1002/pbc.30318] [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: 10/20/2022] [Revised: 02/28/2023] [Accepted: 03/03/2023] [Indexed: 03/29/2023]
Abstract
BACKGROUND Successful engraftment of human cancer biopsies in immunodeficient mice correlates with the poor prognosis of patients. This was reported 30 years ago for children with neuroblastoma, but the standard of care treatment evolved significantly during the last 15 years, leading to improved survival of these patients. Here, we evaluated the association of patient-derived xenograft (PDX) engraftment and prognosis in patients receiving up-to-date treatments for cancers classified as metastatic (stage M) high-risk neuroblastoma (HR-NB) by the International Neuroblastoma Risk Group Staging System (INRGSS). METHODS We obtained biopsies from patients with stage M HR-NB. We inoculated biopsy fragments subcutaneously in mice. We studied the association of PDX engraftment with event-free survival (EFS) and overall survival (OS) of patients. RESULTS Since 2009, we established 17 PDX from 97 samples of 66 patients with stage M HR-NB, with a follow-up of at least two years. Factors associated with higher probability of engraftment were the death as outcome (p = .0006) and the amplification of the gene MYCN in tumors (p = .0271). Patients whose biopsies established a PDX had significantly shorter EFS and OS (p = .0039 and .0002, respectively) than patients whose samples did not engraft. The association of PDX engraftment and OS was significant in patients without MYCN amplification (p = .0041), but not in patients with MYCN amplification (p = .2707). CONCLUSION Positive PDX engraftment is a factor related to poor prognosis and fatal outcome in patients with stage M HR-NB treated with up-to-date therapies.
Collapse
Affiliation(s)
- Rosario Aschero
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Barcelona, Spain
- Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - Helena Castillo-Ecija
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Barcelona, Spain
- Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - Merce Baulenas-Farres
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Barcelona, Spain
- Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - Claudia Resa-Pares
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Barcelona, Spain
- Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - Ana Jimenez-Cabaco
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Barcelona, Spain
- Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - Eva Rodriguez
- Department of Pathology, Hospital Sant Joan de Deu, Barcelona, Spain
| | - Carles Monterrubio
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Barcelona, Spain
| | - Sara Perez-Jaume
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Barcelona, Spain
- Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - Mariona Suñol
- Department of Pathology, Hospital Sant Joan de Deu, Barcelona, Spain
| | - Guillermo L Chantada
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Barcelona, Spain
- Institut de Recerca Sant Joan de Deu, Barcelona, Spain
- CONICET, Buenos Aires, Argentina
| | - Cinzia Lavarino
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Barcelona, Spain
- Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - Jaume Mora
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Barcelona, Spain
- Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - Angel M Carcaboso
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Barcelona, Spain
- Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| |
Collapse
|