1
|
Claeyssens C, Witters P, Segers H, De Koster J, Levtchenko E, Vermeersch P. An unusual case of neuroblastoma presenting with prolonged watery diarrhea in a pediatric patient. Biochem Med (Zagreb) 2025; 35:020901. [PMID: 40520658 PMCID: PMC12161515 DOI: 10.11613/bm.2025.020901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 03/31/2025] [Indexed: 06/18/2025] Open
Abstract
Neuroblastomas represent a diverse group of neuroblastic tumors characterized by variability in their clinical progression and degree of differentiation. In rare cases, patients with neuroblastoma may present with paraneoplastic syndromes, such as watery diarrhea, hypokalemia, and achlorhydria (WDHA syndrome), linked to the secretion of vasoactive intestinal peptide (VIP). We report a case of a 14-month-old girl presented with a three-week history of watery diarrhea and signs of dehydration with no other symptoms. The patient's medical history was unremarkable, and no medication use was reported. Venous blood gas analysis revealed a normal anion gap metabolic acidosis with severe hypokalemia. The patient was referred to our hospital 48 hours post-admission due to persistent hypokalemic metabolic acidosis, unresponsive to intravenous fluid therapy. The primary causes of normal anion gap metabolic acidosis in young children are gastrointestinal bicarbonate loss due to diarrhea and renal bicarbonate loss. Semi-quantitative urine organic acid analysis, reported 48 hours after admission, revealed increased vanillylmandelic acid (VMA) (89 mmol/mol creatinine) and homovanillic acid (HVA) (21 mmol/mol creatinine), raising the suspicion of a neuroblastoma. Subsequent analysis of an acidified urine sample confirmed a more than threefold increase in VMA, HVA, normetanephrine, norepinephrine, and 3-methoxytyramine concentrations. In addition, VIP was markedly elevated (1994 ng/L) in a blood sample. The diagnosis of neuroblastoma was confirmed through imaging and histological examination. This case illustrates that chronic diarrhea with metabolic dysregulation (e.g. hypokalemia) can be the first and only symptom in patients with VIP-secreting neuroblastoma which can result in delayed diagnosis of neuroblastoma.
Collapse
Affiliation(s)
- Claire Claeyssens
- Clinical Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Peter Witters
- Clinical Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Heidi Segers
- Clinical Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| | - Jan De Koster
- Clinical Department of Pediatrics, Ziekenhuis Oost-Limburg, Genk, Belgium
| | - Elena Levtchenko
- Clinical Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
- Department of Development & Regeneration, KU Leuven, Belgium
| | - Pieter Vermeersch
- Clinical Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
- Department of Cardiovascular Sciences, KU Leuven, Belgium
| |
Collapse
|
2
|
Shih JC, Chen PY, Kuo CH, Hsieh CH, Chang HY, Lee HC, Huang CH, Hsu CH, Hsu WM, Huang HC, Juan HF. DHODH blockade induces ferroptosis in neuroblastoma by modulating the mevalonate pathway. Mol Cell Proteomics 2025:101014. [PMID: 40513779 DOI: 10.1016/j.mcpro.2025.101014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 06/06/2025] [Accepted: 06/10/2025] [Indexed: 06/16/2025] Open
Abstract
Neuroblastoma is the most common heterogeneous solid tumor in children, and current treatment options remain limited, especially for high-risk patients. Previous studies have identified dihydroorotate dehydrogenase (DHODH), a key enzyme in pyrimidine synthesis, as a potential therapeutic target in cancer. However, none of the existing FDA-approved DHODH inhibitors have shown effective inhibition of neuroblastoma cell growth. To address this challenge, we employed virtual screening to discover potential DHODH-targeting drugs, identifying Regorafenib as a promising candidate. Regorafenib significantly inhibited neuroblastoma growth in both neuroblastoma cells and patient-derived organoids. To unravel the underlying molecular mechanisms, we conducted Tandem Mass Tag (TMT)-based quantitative proteomics using LC-MS/MS. Our proteomic profiling revealed substantial regulation of lipid metabolism proteins, specifically those in the mevalonate pathway, correlating with ferroptosis induction. Further analysis showed that DHODH inhibition led to a reduction in total cholesterol, cholesterol esters, disrupted lipid droplet formation, and significantly decreased the expression of Squalene Epoxidase (SQLE), a key enzyme in lipid metabolism. Notably, we also observed an increase in nuclear SQLE expression following DHODH inhibition. In summary, our study highlights DHODH blockade as a novel approach to induce ferroptosis through lipid metabolism reprogramming, underscoring DHODH as a viable therapeutic target for neuroblastoma treatment. These insights open new avenues for metabolic-based interventions in aggressive pediatric cancers.
Collapse
Affiliation(s)
- Jui-Chia Shih
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei, Taiwan
| | - Pin-Yu Chen
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei, Taiwan
| | - Chuan-Hao Kuo
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei, Taiwan
| | - Chiao-Hui Hsieh
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Hsin-Yi Chang
- Graduate Institute of Medical Science, National Defense Medical Center, Taipei, Taiwan
| | - Hong-Chih Lee
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Chen-Hao Huang
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - Chun-Hua Hsu
- Department of Agricultural Chemistry, National Taiwan University, Taipei, Taiwan
| | - Wen-Ming Hsu
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Hsuan-Cheng Huang
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Hsueh-Fen Juan
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei, Taiwan; Department of Life Science, National Taiwan University, Taipei, Taiwan; Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan; enter for Computational and Systems Biology, National Taiwan University, Taipei, Taiwan; Center for Advanced Computing and Imaging in Biomedicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
3
|
Liu M, Lv D, Yan W, Wu Y, Wang S, Wang L, Lei J, Zeng D, Wang Z, Liu F, Deng B, Liu Q, He B, Yan M. SLIT3-mediated intratumoral crosstalk induces neuroblastoma differentiation via a spontaneous regression-like program. J Transl Med 2025; 23:598. [PMID: 40448172 DOI: 10.1186/s12967-025-06621-0] [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: 03/18/2025] [Accepted: 05/16/2025] [Indexed: 06/02/2025] Open
Abstract
BACKGROUND Neuroblastoma, the most common pediatric extracranial solid tumor, has heterogeneous clinical outcomes ranging from malignant progression to spontaneous regression. With the highest frequency of the elusive spontaneous regression, low-risk INSS Stage 4S neuroblastoma represents an ideal model for mechanistic investigation. Spontaneous regression is often accompanied by tumor differentiation, but the mechanisms underlying this process remain largely unclear. METHODS Single-nucleus transcriptomics (snRNA-seq) data of neuroblastoma samples were obtained from the Synapse repository to investigate the composition of heterogeneous tumor cell clusters. The feature of the Stage 4S-specific tumor cell subpopulation was revealed through differential expression analysis, pathway enrichment analysis and pseudotime analysis, followed by clinical significance validation on public cohort datasets. The biological function of secreted SLIT3 was validated using multiple in vitro models, including recombinant protein treatment, conditioned medium treatment, and cell lines coculture, to confirm the intratumoral crosstalk effect. Orthotopic and subcutaneous xenograft models were established to verify SLIT3's in vivo function. Cellular bulk RNA-seq analysis was performed with or without SLIT3 recombinant protein treatment to discover the downstream pathways activated by SLIT3, followed by validation with specific pathway inhibitors. RESULTS Analysis of snRNA-seq revealed a distinct subpopulation of tumor cells within INSS Stage 4S neuroblastoma, characterized by a spontaneous regression-like program progressing toward differentiation. Activated SLIT-ROBO signaling was found in the Stage 4S-specific tumor cell subpopulation, which strongly correlated with favorable prognosis. Further investigation into the secreted ligands in SLIT-ROBO related pathways revealed that SLIT3 displayed the most potent enrichment in Stage 4S tumors and the strongest differentiation-inducing effect. In vitro experiments using recombinant SLIT3 protein, conditioned medium, and cell lines coculture consistently demonstrated the capacity of SLIT3 to induce neuroblastoma cell differentiation via intratumoral crosstalk, as evidenced by increased neurite outgrowth and elevated expression of neuronal differentiation markers. Both orthotopic xenograft and subcutaneous xenograft models demonstrated that SLIT3 expression suppressed tumor growth, leading to in vivo tumor differentiation. Mechanistically, PLCβ/PKC signaling mediates the SLIT3-induced neuroblastoma cell differentiation. CONCLUSIONS Stage 4S-specific tumor cell subpopulation exhibits a spontaneous regression-like program, from which SLIT3 mediates intratumoral crosstalk and promotes neuroblastoma differentiation via PLCβ/PKC signaling. These findings provide new insights into the mechanism of spontaneous regression in neuroblastoma and offer novel therapeutic targets for differentiation-based treatment strategies.
Collapse
Affiliation(s)
- Meiling Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Psychobehavioral Cancer Research Center, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Dekang Lv
- Institute of Cancer Stem Cell, Dalian Medical University, 9 West Section, Lvshun South Road, Dalian, 116044, Liaoning, China
| | - Wenjing Yan
- Institute of Cancer Stem Cell, Dalian Medical University, 9 West Section, Lvshun South Road, Dalian, 116044, Liaoning, China
| | - Yi Wu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Psychobehavioral Cancer Research Center, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Shulan Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Psychobehavioral Cancer Research Center, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Luoxuan Wang
- Institute of Cancer Stem Cell, Dalian Medical University, 9 West Section, Lvshun South Road, Dalian, 116044, Liaoning, China
| | - Jie Lei
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Psychobehavioral Cancer Research Center, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Deshun Zeng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Psychobehavioral Cancer Research Center, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Zifeng Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Psychobehavioral Cancer Research Center, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Fang Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Psychobehavioral Cancer Research Center, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Bing Deng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Psychobehavioral Cancer Research Center, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Quentin Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Psychobehavioral Cancer Research Center, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
- Institute of Cancer Stem Cell, Dalian Medical University, 9 West Section, Lvshun South Road, Dalian, 116044, Liaoning, China.
| | - Bin He
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Psychobehavioral Cancer Research Center, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
| | - Min Yan
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Psychobehavioral Cancer Research Center, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
| |
Collapse
|
4
|
Liu J, Zhang M, Ouyang Y, Chang J, Zhang W, Zhou C, He J, Zhang X. Association between MYCN gene polymorphisms and neuroblastoma susceptibility: a case-control study in Chinese children from Jiangsu Province. BMC Cancer 2025; 25:892. [PMID: 40389877 PMCID: PMC12087095 DOI: 10.1186/s12885-025-14310-w] [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: 02/06/2025] [Accepted: 05/12/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Neuroblastoma, developed from the sympathetic nervous system, is a deadly childhood malignancy. There is an urgent need to elucidate its intricated etiology. MYCN amplification leads to aggressive neuroblastoma and represents a powerful marker of poor prognosis. However, the correlation between MYCN gene polymorphisms and neuroblastoma susceptibility remains largely unknown in Chinese Han children. METHODS We conducted a case-control study to evaluate the associations between MYCN gene polymorphisms and neuroblastoma susceptibility, involving 402 cases and 473 controls from Jiangsu Province, China. The association strength between the studied polymorphisms and neuroblastoma susceptibility was quantified using odds ratios and 95% confidence intervals. RESULTS Four studied polymorphisms (rs57961569 G > A, rs9653226 T > C, rs13034994 A > G, and rs60226897 G > A) were significantly associated with neuroblastoma susceptibility. Stratified analysis of two polymorphisms (rs13034994 A > G and rs60226897 G > A) demonstrated stronger associations with neuroblastoma susceptibility in specific subgroups. Moreover, survival analysis demonstrated elevated MYCN expression in high-risk patients, with reduced expression correlating to improved survival outcomes. CONCLUSION Our study indicated that MYCN gene polymorphisms are significantly associated with neuroblastoma susceptibility in the eastern Chinese population and that high expression of the MYCN gene may suggest a poor prognosis. Nevertheless, further verification should be conducted with large-scale and well-designed studies to confirm our findings.
Collapse
Affiliation(s)
- Jiabin Liu
- Department of Pediatric Surgery, Liuzhou Key Laboratory of Birth Defect Prevention and Control, Guangzhou Women and Children's Medical Center Liuzhou Hospital, Liuzhou, 545616, Guangxi, China
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Guangzhou, 510623, Guangdong, China
| | - Mengzhen Zhang
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Guangzhou, 510623, Guangdong, China
| | - Yu Ouyang
- Department of Pediatric Surgery, Liuzhou Key Laboratory of Birth Defect Prevention and Control, Guangzhou Women and Children's Medical Center Liuzhou Hospital, Liuzhou, 545616, Guangxi, China
| | - Jiaming Chang
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Guangzhou, 510623, Guangdong, China
| | - Wenli Zhang
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Guangzhou, 510623, Guangdong, China
| | - Chunlei Zhou
- Department of Pathology, Children's Hospital of Nanjing Medical University, Nanjing, 210008, Jiangsu, China
| | - Jing He
- Department of Pediatric Surgery, Liuzhou Key Laboratory of Birth Defect Prevention and Control, Guangzhou Women and Children's Medical Center Liuzhou Hospital, Liuzhou, 545616, Guangxi, China.
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Guangzhou, 510623, Guangdong, China.
| | - Xinxin Zhang
- Department of Pediatric Surgery, Liuzhou Key Laboratory of Birth Defect Prevention and Control, Guangzhou Women and Children's Medical Center Liuzhou Hospital, Liuzhou, 545616, Guangxi, China.
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Guangzhou, 510623, Guangdong, China.
| |
Collapse
|
5
|
Xie L, Luo D, Chen X, Zhang M, Zhang W, Chang J, Zhou H, Zhang X, He J, Chen L, Zhou C. RAS gene polymorphisms confer the risk of neuroblastoma in Chinese children from Jiangsu province. Pediatr Surg Int 2025; 41:130. [PMID: 40323475 DOI: 10.1007/s00383-025-06025-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/11/2025] [Indexed: 05/07/2025]
Abstract
INTRODUCTION To evaluate the potential association between single nucleotide polymorphisms in the RAS gene and neuroblastoma risk, we examined four candidate SNPs within this gene. METHODS Our hospital-based case-control study included 402 cases and 473 controls. Four SNPs (rs12587 G > T, rs7973450 A > G, and rs7312175 G > A in KRAS and rs2273267 A > T in NRAS) were genotyped using the TaqMan assay. The association between RAS gene polymorphisms and neuroblastoma susceptibility was assessed through odds ratios and 95% confidence intervals. RESULTS None of the four candidate SNPs exhibited a significant attribution to neuroblastoma risk. However, the concurrent presence of 2-3 KRAS risk genotypes significantly conferred an increased susceptibility to neuroblastoma (adjusted odds ratio [AOR] = 2.55, 95% confidence interval [CI] 1.33-4.89; P = 0.005). Further stratified analyses indicated that carriers of the KRAS rs12587 TT genotype tended to be more predisposed to neuroblastoma in males and in the subgroup with tumors originating from other sites. Additionally, the co-occurrence of 2-3 KRAS risk genotypes was found to be linked to an increased neuroblastoma risk in subgroups of individuals older than 18 months, males, tumors originating from retroperitoneum, mediastinum, or other sites, and those with tumors at clinical stage III + IV, respectively. CONCLUSIONS In summary, a single KRAS gene polymorphism may be weakly associated with an increased risk of childhood neuroblastoma in Jiangsu province, China, while the presence of more KRAS risk genotypes may increase the contribution to the risk of neuroblastoma.
Collapse
Affiliation(s)
- Lili Xie
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Dongyuan Luo
- Department of Stomatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Xuemei Chen
- Department of Pharmacy, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, 650118, Yunnan, China
| | - Mengzhen Zhang
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Wenli Zhang
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Jiaming Chang
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Haixia Zhou
- Department of Hematology, The Key Laboratory of Pediatric Hematology and Oncology Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Xinxin Zhang
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Liping Chen
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China.
| | - Chunlei Zhou
- Department of Pathology, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
6
|
Wang Z, Liu Y, Zhang Y, Shi J, Xie S, Yi M, Zhang X, Tao D, Yang Y. TSPYL5-driven G3BP1 nuclear membrane translocation facilitates p53 cytoplasm sequestration via accelerating RanBP2-mediated p53 sumoylation and nuclear export in neuroblastoma. Cell Death Dis 2025; 16:358. [PMID: 40319028 PMCID: PMC12049415 DOI: 10.1038/s41419-025-07694-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 04/07/2025] [Accepted: 04/24/2025] [Indexed: 05/07/2025]
Abstract
Cytoplasmic sequestration of wild-type p53, representing a nonmutational event of p53 activity suppression, is a characteristic phenotype of undifferentiated neuroblastoma (NB); however, the underlying mechanism is yet to be defined. In the present study, we observed that TSPYL5 effectively tethers p53 in the cytoplasm and greatly inhibits its function as a transcription factor. Mechanistically, the binding of TSPYL5 with G3BP1 enhances G3BP1 Ser149 phosphorylation to drive G3BP1 nuclear membrane translocation, which recruits more p53 for nucleoporin RanBP2 by the formation of the RanBP2-G3BP1-p53 complex. Thus, the accelerating p53 sumoylation promotes its nuclear export. With this signal pathway, TSPYL5 augments the malignant characteristics of neuroblastoma cells. Our findings unravel a detailed TSPYL5-driven molecular axis that sheds light on the regulating system of the p53 sumoylation-based cytoplasmic sequestration in NB cells, paving the way for the novel therapeutic opportunities for NB cancers by antagonizing TSPYL5 function.
Collapse
Affiliation(s)
- Zhaokun Wang
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yunqiang Liu
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yangwei Zhang
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jiaying Shi
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Shengyu Xie
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ming Yi
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xinyue Zhang
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Dachang Tao
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yuan Yang
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
7
|
Zhu B, Pitts MG, Buoncristiani MD, Bryant LT, Lopez-Nunez O, Gurria JP, Shedlock C, Ribas R, Keohane S, Liu J, Wang C, Gentry MS, Shelman NR, Allison DB, Evers BM, Sun RC, Rellinger EJ. GDP-mannose 4,6-dehydratase is a key driver of MYCN-amplified neuroblastoma core fucosylation and tumorigenesis. Oncogene 2025; 44:1272-1283. [PMID: 39956863 PMCID: PMC12048354 DOI: 10.1038/s41388-025-03297-0] [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: 07/10/2024] [Revised: 01/08/2025] [Accepted: 02/03/2025] [Indexed: 02/18/2025]
Abstract
MYCN-amplification is a genetic hallmark of ~40% of high-risk neuroblastomas (NBs). Altered glycosylation is a common feature of adult cancer progression, but little is known about how genetic signatures such as MYCN-amplification alter glycosylation profiles. Herein, matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) revealed increased core fucosylated glycan abundance within neuroblast-rich regions of human MYCN-amplified NB tumors. GDP-mannose 4,6-dehydratase (GMDS) is responsible for the first-committed and rate-limiting step of de novo GDP-fucose synthesis. High GMDS expression was found to be associated with poor patient survival, advanced stage disease, and MYCN-amplification in human NB tumors. Chromatin immunoprecipitation and promoter reporter assays demonstrated that N-MYC directly binds and activates the GMDS promoter in NB cells. When GMDS was blocked through either genetic or pharmacological mechanisms, NBs were found to be dependent upon de novo GDP-fucose production to sustain cell surface and secreted core fucosylated glycan abundance, as well as adherence and motility. Moreover, genetic knockdown of GMDS inhibited tumor formation and progression in vivo. These critical findings identify de novo GDP-fucose production as a novel metabolic vulnerability that may be exploited in designing new treatment strategies for MYCN-amplified NBs.
Collapse
Affiliation(s)
- Beibei Zhu
- University of Kentucky, Department of Surgery, Lexington, KY, 40536, USA
| | - Michelle G Pitts
- University of Kentucky, Department of Surgery, Lexington, KY, 40536, USA
| | | | - Lindsay T Bryant
- University of Kentucky, Department of Surgery, Lexington, KY, 40536, USA
| | - Oscar Lopez-Nunez
- Cincinnati Children's Hospital Medical Center, Department of Pathology, Cincinnati, OH, 45229, USA
| | - Juan P Gurria
- Cincinnati Children's Hospital Medical Center, Department of Pediatric Surgery, Cincinnati, OH, 45229, USA
| | - Cameron Shedlock
- University of Florida, Department of Biochemistry & Molecular Biology, Gainesville, FL, 32610, USA
| | - Roberto Ribas
- University of Florida, Department of Biochemistry & Molecular Biology, Gainesville, FL, 32610, USA
| | - Shannon Keohane
- University of Florida, Department of Biochemistry & Molecular Biology, Gainesville, FL, 32610, USA
| | - Jinpeng Liu
- University of Kentucky, Department of Bioinformatics, Lexington, KY, 40536, USA
| | - Chi Wang
- University of Kentucky, Department of Bioinformatics, Lexington, KY, 40536, USA
| | - Matthew S Gentry
- University of Florida, Department of Biochemistry & Molecular Biology, Gainesville, FL, 32610, USA
| | - Nathan R Shelman
- University of Kentucky, Department of Pathology, Lexington, KY, 40536, USA
| | - Derek B Allison
- University of Kentucky, Department of Pathology, Lexington, KY, 40536, USA
- University of Kentucky, Markey Cancer Center, Lexington, KY, 40536, USA
| | - B Mark Evers
- University of Kentucky, Department of Surgery, Lexington, KY, 40536, USA
- University of Kentucky, Markey Cancer Center, Lexington, KY, 40536, USA
| | - Ramon C Sun
- University of Florida, Department of Biochemistry & Molecular Biology, Gainesville, FL, 32610, USA
| | - Eric J Rellinger
- University of Kentucky, Department of Surgery, Lexington, KY, 40536, USA.
- University of Kentucky, Markey Cancer Center, Lexington, KY, 40536, USA.
| |
Collapse
|
8
|
Liu Z, Hong JJ, Zhang X, Sayers CM, Fang W, Xu M, Loria S, Maskal S, Lei H, Wu H, Swenson R, Meier JL, Shern JF, Thiele CJ. MYCN and KAT2A form a feedforward loop to drive an oncogenic transcriptional program in neuroblastoma. Oncogenesis 2025; 14:13. [PMID: 40274766 PMCID: PMC12022051 DOI: 10.1038/s41389-025-00557-2] [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: 08/26/2024] [Revised: 04/03/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025] Open
Abstract
The oncoprotein MYCN drives malignancy in various cancer types, including neuroblastoma (NB). However, our understanding of the mechanisms underlying its transcriptional activity and oncogenic function, as well as effective strategies to target it, remains limited. We discovered that MYCN interacts with the transcriptional coactivator KAT2A, and this interaction significantly contributes to MYCN's activity in NB. Our genome-wide analyses indicate MYCN recruits KAT2A to bind to DNA, thereby transcriptionally regulating genes associated with ribosome biogenesis and RNA processing. Moreover, we identified that MYCN directly activates KAT2A transcription, while KAT2A acetylates MYCN, increasing MYCN protein stability. Consequently, MYCN and KAT2A establish a feedforward loop that effectively regulates global gene expression, governing the malignant NB phenotype. Treatment of NB cells with a KAT2A Proteolysis Targeting Chimera (PROTAC) degrader reduces MYCN protein levels, antagonizes MYCN-mediated gene transcription regulation and suppresses cell proliferation. This study highlights the potential of transcriptional cofactors as viable targets for developing anti-MYCN therapies.
Collapse
Affiliation(s)
- Zhihui Liu
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, MD, USA.
| | - Jason J Hong
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Xiyuan Zhang
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Carly M Sayers
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Wendy Fang
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Man Xu
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Sydney Loria
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Sakereh Maskal
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Haiyan Lei
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Haitao Wu
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Rolf Swenson
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Jordan L Meier
- Chemical Biology Laboratory, National Cancer Institute, Frederick, MD, USA
| | - Jack F Shern
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Carol J Thiele
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
9
|
Nguyen LD, Sengupta S, Cho KI, Floru A, George RE, Krichevsky AM. A drug that induces the microRNA miR-124 enables differentiation of retinoic acid-resistant neuroblastoma cells. Sci Signal 2025; 18:eads2641. [PMID: 40233178 DOI: 10.1126/scisignal.ads2641] [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: 08/05/2024] [Accepted: 03/27/2025] [Indexed: 04/17/2025]
Abstract
Tumor cell heterogeneity in neuroblastoma, a pediatric cancer arising from neural crest-derived progenitor cells, presents clinical challenges. Unlike adrenergic (ADRN) neuroblastoma cells, neuroblastoma cells with a mesenchymal (MES) identity are resistant to chemotherapy and retinoid therapy, which contributes to relapses and treatment failures. We explored whether up-regulation of the neurogenic, tumor suppressor microRNA miR-124 could promote the differentiation of retinoic acid-resistant MES neuroblastoma cells. Leveraging our screen for miRNA-modulatory small molecules, we identified and validated the tyrosine and phosphoinositide kinase inhibitor PP121 as a robust inducer of miR-124. Combining PP121 and BDNF-activating bufalin synergistically arrested proliferation and promoted the sustained differentiation of MES/heterogeneous SK-N-AS cells over several weeks. This protocol also resulted in the differentiation of multiple MES neuroblastoma and glioblastoma cell lines. RNA-seq analysis of differentiated MES/heterogeneous SK-N-AS cells revealed the replacement of the ADRN core regulatory circuitry with circuitries associated with chromaffin cells and Schwann cell precursors. Furthermore, differentiation was associated with inhibition of the CDK4/CDK6 pathway and activation of a transcriptional program that correlated with improved outcomes for patients with neuroblastoma. Our findings suggest an approach with translational potential to induce the differentiation of therapy-resistant cancers of the nervous system. Moreover, these long-lived, differentiated cells could be used to study mechanisms underlying cancer biology and therapies.
Collapse
Affiliation(s)
- Lien D Nguyen
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Satyaki Sengupta
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02115, USA
| | - Kevin I Cho
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Alexander Floru
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02115, USA
| | - Rani E George
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02115, USA
| | - Anna M Krichevsky
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
10
|
Elmenawi S, Fawzy M. 15 Years Old ALK Gene from Birth to Adolescence; Where to in NBL. Curr Oncol Rep 2025; 27:431-445. [PMID: 40064818 PMCID: PMC11976753 DOI: 10.1007/s11912-025-01650-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] [Subscribe] [Scholar Register] [Accepted: 02/03/2025] [Indexed: 04/09/2025]
Abstract
PURPOSE OF REVIEW This review provides a comprehensive understanding of the ALK gene, encompassing its prevalence, genetic alterations, and significance in neuroblastoma diagnosis, outcome prediction, and targeted therapy utilization. The insights presented aim to inform future research directions and clinical practices in this field. RECENT FINDINGS High risk neuroblastoma, comprising approximately 50% of all cases, presents a particularly poor prognosis. In 2008, the discovery of ALK aberrations in neuroblastoma marked a significant breakthrough, leading to the recognition of ALK as a target for tumors with activating ALK alterations. This discovery has paved the way for the development of various ALK inhibitors, which have shown promising clinical efficacy. ALK amplification, often observed alongside MYCN amplification, has been associated with unfavorable outcomes in patients. Activating mutations in the kinase domain of ALK, particularly at hotspot positions F1174, R1275, and F1245, have been identified. These mutations can occur at clonal or subclonal levels, posing challenges for early detection and potentially influencing disease progression and therapy resistance. The availability of ALK inhibitors, initially developed for adult cancers, has expedited the translation of this knowledge into targeted therapies for neuroblastoma. However, resistance to ALK inhibitors can emerge as a result of treatment or preexist as subclones within the tumor prior to therapy. Future trials should focus on identifying additional targets complementing ALK inhibition to enhance treatment efficacy and overcome acquired resistance. Furthermore, the utilization of circulating tumor DNA as a non-invasive approach for longitudinal monitoring of ALK-positive neuroblastoma patients, in combination with radiographic evaluation of treatment response, holds promise for understanding dynamic tumor changes over time.
Collapse
Affiliation(s)
- Salma Elmenawi
- Clinical Research Department, Children's Cancer Hospital Egypt, 57357, 1-Sekket Elemam-Sayeda Zeinab, Cairo, Egypt.
| | - Mohamed Fawzy
- Pediatric Oncology Department, Children's Cancer Hospital Egypt, 57357, 1-Sekket Elemam-Sayeda Zeinab, Cairo, Egypt
- Pediatric Oncology Department, National Cancer Institute, Cairo, Egypt
| |
Collapse
|
11
|
Corrias MV, Di Marco E, Bonaretti C, Squillario M, Amoroso L, Conte M, Ponzoni M, Biassoni R. In Infants with Neuroblastoma Standard Therapy Only Partially Reverts the Fecal Microbiome Dysbiosis Present at Diagnosis. Microorganisms 2025; 13:691. [PMID: 40142583 PMCID: PMC11946756 DOI: 10.3390/microorganisms13030691] [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: 02/21/2025] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
The fecal microbiomes of 15 infants with neuroblastoma (NB) at the onset of the disease and after standard-of-care therapy have been prospectively analyzed compared to those of age-matched healthy infants. By applying several algorithms to 16S sequencing, we found that the fecal microbiomes of infants with NB at onset were abundant in Pseudomonadota, including different descendants of Gammaproteobacteria. After completing therapy, their abundance decreased to a level like that observed in healthy infants. In contrast, the Bacillota that showed at the onset an abundance like that of healthy infants doubled their amount after treatment. In infants with NB, the beta diversity of the fecal microbiomes was significantly reduced compared to healthy infants and patients at the end of treatment. The Random Forest algorithm and the Reingold-Tilford heat tree showed that Enterobacteriaceae had a higher abundance at the onset, which declined after therapy. Picrust2 inferred pathway analysis indicated that the drug treatment was associated with a reduction in the polyamine pathway, highly represented in samples of NB at the onset. In conclusion, the dysbiosis observed in infants with NB at onset changed following standard-of-care treatment. Still, the composition at the end of treatment did not completely resemble that of healthy infants.
Collapse
Affiliation(s)
- Maria Valeria Corrias
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16147 Genova, Italy;
| | - Eddi Di Marco
- Central Laboratory, IRCCS Istituto Giannina Gaslini, 00161 Genova, Italy;
| | - Carola Bonaretti
- Molecular Diagnostics, IRCCS Istituto Giannina Gaslini, 00161 Genova, Italy; (C.B.); (R.B.)
| | | | - Loredana Amoroso
- Oncology Unit, IRCCS Istituto Giannina Gaslini, 00161 Genova, Italy; (L.A.); (M.C.)
| | - Massimo Conte
- Oncology Unit, IRCCS Istituto Giannina Gaslini, 00161 Genova, Italy; (L.A.); (M.C.)
| | - Mirco Ponzoni
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16147 Genova, Italy;
| | - Roberto Biassoni
- Molecular Diagnostics, IRCCS Istituto Giannina Gaslini, 00161 Genova, Italy; (C.B.); (R.B.)
| |
Collapse
|
12
|
AlKhazal A, Chohan S, Ross DJ, Kim J, Brown EG. Emerging clinical and research approaches in targeted therapies for high-risk neuroblastoma. Front Oncol 2025; 15:1553511. [PMID: 40104501 PMCID: PMC11913827 DOI: 10.3389/fonc.2025.1553511] [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: 12/30/2024] [Accepted: 02/06/2025] [Indexed: 03/20/2025] Open
Abstract
Neuroblastoma is a pediatric cancer that originates from neural crest cells and is the most common extracranial solid tumor in children under five years of age. While low-risk neuroblastoma often regresses spontaneously, high-risk neuroblastoma poses a significant clinical challenge. Recent advances in understanding neuroblastoma's molecular mechanisms have led to the development of targeted therapies that aim to selectively inhibit specific pathways involved in tumor growth and progression, improving patient outcomes while minimizing side effects. This review provides a comprehensive review of neuroblastoma biology and emerging therapeutic strategies. Key topics include (a) immunotherapies and immunotargets, (b) non-coding RNAs (long non-coding RNA, microRNA, and circular RNA), (c) molecular biomarkers and pathways, and (d) limitations and future directions.
Collapse
Affiliation(s)
- Albatool AlKhazal
- Department of Surgery, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Samiha Chohan
- Department of Surgery, School of Medicine, University of California, Davis, Davis, CA, United States
- Department of Biological Sciences, California State University, Sacramento, Sacramento, CA, United States
| | - Destani J Ross
- Department of Surgery, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Jinhwan Kim
- Department of Surgery, School of Medicine, University of California, Davis, Davis, CA, United States
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States
| | - Erin G Brown
- Department of Surgery, School of Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
13
|
Santo G, di Santo G, Cicone F, Virgolini I. Peptide receptor radionuclide therapy with somatostatin analogs beyond gastroenteropancreatic neuroendocrine tumors. J Neuroendocrinol 2025; 37:e70013. [PMID: 40064181 PMCID: PMC11919479 DOI: 10.1111/jne.70013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 02/07/2025] [Accepted: 02/18/2025] [Indexed: 03/20/2025]
Abstract
First isolated by Brazeau et al. in 1972, somatostatin (SST) is a neuropeptide known for regulating various signaling pathways through its specific cell surface receptors. Somatostatin receptors (SSTRs) comprise a family of five G protein-coupled receptors that are widely distributed across the human body and are expressed by various tumor types. The growing understanding of their clinical potential led to the introduction of both cold and radiolabeled somatostatin analogs (SSAs), which have revolutionized the management of several cancers, especially neuroendocrine tumors. As a direct consequence, advances in peptide receptor radionuclide therapy (PRRT) over the last 30 years led to the approval of 177Lu-DOTATATE for the treatment of gastroenteropancreatic neuroendocrine tumors (GEPNETs). Theoretically, any cancer patients whose tumors express SSTR, as demonstrated in vivo through SSTR-based molecular imaging, could be candidates for PRRT, especially those with limited treatment options. However, evidence on the efficacy of PRRT in non-GEPNET SSTR-expressing tumors is limited, and mainly derived from small retrospective studies. Given the limited therapeutic options for advanced/metastatic patients, there is a clear need for randomized trials to formally approve PRRT with SSAs for patients who may benefit from this treatment, particularly in certain types of neuroendocrine neoplasms such as lung carcinoids, paragangliomas, and meningiomas, where high rates of disease control (up to 80%) can be achieved. In addition, emerging evidence supports the potential of combination therapies, alpha emitters, and non-SSTR-based radionuclide therapy in tumors beyond GEPNET. This review aims to provide a comprehensive overview of PRRT's role in cancers beyond GEPNET, exploring new possibilities and future directions for most SSTR highly expressing tumors.
Collapse
Affiliation(s)
- Giulia Santo
- Department of Nuclear MedicineMedical University of InnsbruckInnsbruckAustria
- Department of Experimental and Clinical Medicine“Magna Graecia” University of CatanzaroCatanzaroItaly
| | - Gianpaolo di Santo
- Department of Nuclear MedicineMedical University of InnsbruckInnsbruckAustria
| | - Francesco Cicone
- Department of Experimental and Clinical Medicine“Magna Graecia” University of CatanzaroCatanzaroItaly
| | - Irene Virgolini
- Department of Nuclear MedicineMedical University of InnsbruckInnsbruckAustria
| |
Collapse
|
14
|
Antal P, Kuchár J, Rigamonti L, Kvasnicová M, Gonzalez G, Rárová L, Strnad M, Kopel P. Co(II), Cu(II), and Zn(II) thio-bis(benzimidazole) complexes induce apoptosis via mitochondrial pathway. J Inorg Biochem 2025; 264:112786. [PMID: 39644804 DOI: 10.1016/j.jinorgbio.2024.112786] [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: 08/09/2024] [Revised: 11/22/2024] [Accepted: 11/22/2024] [Indexed: 12/09/2024]
Abstract
The copper(II), cobalt(II), and zinc(II) complexes with 2-(1H-benzimidazol-2-ylmethylsulfanylmethyl)-1H-benzimidazole (tbb) and 2-[2-[2-(1H-benzimidazol-2-yl)ethylsulfanyl]ethyl]-1H-benzimidazole (tebb), [Cu(tbb)Cl2] (1), [Co(tbb)Cl2] (2), [Zn(tbb)Cl2] (3), [Cu(tebb)Cl(H2O)]Cl (4), [Co(tebb)Cl2]n·nCH3OH (5) and [Zn(tebb)Cl(H2O)]Cl (6), have been prepared and evaluated for antiproliferative activity. The structure of (4) was proved by X-ray diffraction crystallography. The coordination compounds were tested for their cytotoxic activities in cancer cell lines in vitro. The lower IC50 values were obtained for Co(II), Cu(II), and Zn(II) complexes with tebb in comparison with tbb complexes. Complex 2 showed strong antiproliferative selectivity for leukemia CEM cells and nontoxicity towards other tested cell lines and normal human cells (BJ and RPE-1). Proapoptotic activity of 2 and 5 were weaker than positive control cisplatin, but the big advantage of these complexes was their zero-cytotoxicity for normal healthy cells in contrast to the high cytotoxicity of cisplatin. The activation of apoptotic initiation phase was detected in neuroblastoma cancer cell line SH-SY5Y where 5 was cytotoxic without fragmentation of cells. Interestingly, complexes 5, 6, and tebb, together with cisplatin, dramatically impaired the mitochondrial membrane potential of SH-SY5Y after 72 h. Taken together, we demonstrated that our compounds trigger apoptosis via the mitochondrial pathway.
Collapse
Affiliation(s)
- Peter Antal
- Department of Inorganic Chemistry, Faculty of Science, Palacký University, 17. listopadu 12, CZ-779 00 Olomouc, Czech Republic
| | - Juraj Kuchár
- Department of Inorganic Chemistry, Institute of Chemistry, Faculty of Science, P. J. Šafárik University in Košice, Moyzesova 11, SK-041 54 Košice, Slovakia
| | - Luca Rigamonti
- Dipartimento di Scienze Chimiche e Geologiche, Università degli Studi di Modena e Reggio Emilia, via G. Campi 103, 41125 Modena, Italy
| | - Marie Kvasnicová
- Department of Experimental Biology, Faculty of Science, Palacký University, Šlechtitelů 27, CZ-779 00 Olomouc, Czech Republic; Laboratory of Growth Regulators, Faculty of Science, Palacký University, Institute of Experimental Botany of the Czech Academy of Sciences, Šlechtitelů 27, CZ-779 00 Olomouc, Czech Republic
| | - Gabriel Gonzalez
- Department of Experimental Biology, Faculty of Science, Palacký University, Šlechtitelů 27, CZ-779 00 Olomouc, Czech Republic; Department of Neurology, University Hospital in Olomouc, I. P. Pavlova 6, CZ-775 20 Olomouc, Czech Republic
| | - Lucie Rárová
- Department of Experimental Biology, Faculty of Science, Palacký University, Šlechtitelů 27, CZ-779 00 Olomouc, Czech Republic; Laboratory of Growth Regulators, Faculty of Science, Palacký University, Institute of Experimental Botany of the Czech Academy of Sciences, Šlechtitelů 27, CZ-779 00 Olomouc, Czech Republic.
| | - Miroslav Strnad
- Laboratory of Growth Regulators, Faculty of Science, Palacký University, Institute of Experimental Botany of the Czech Academy of Sciences, Šlechtitelů 27, CZ-779 00 Olomouc, Czech Republic
| | - Pavel Kopel
- Department of Inorganic Chemistry, Faculty of Science, Palacký University, 17. listopadu 12, CZ-779 00 Olomouc, Czech Republic.
| |
Collapse
|
15
|
Olgun N, Arayici ME, Kızmazoglu D, Cecen RE. Assessment of Chemo-Immunotherapy Regimens in Patients with Refractory or Relapsed Neuroblastoma: A Systematic Review with Meta-Analysis of Critical Oncological Outcomes. J Clin Med 2025; 14:934. [PMID: 39941606 PMCID: PMC11818460 DOI: 10.3390/jcm14030934] [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: 01/11/2025] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
Background: Neuroblastoma is a highly aggressive pediatric cancer, particularly in children with refractory or relapsed disease, where survival outcomes remain poor despite advancements in treatment. Combining anti-GD2 antibodies, such as dinutixumab beta, dinutixumab, and naxitanab, with conventional chemotherapy has emerged as a promising approach to improve clinical outcomes in this high-risk population. This chemo-immunotherapy regimen meta-analysis aimed to investigate the efficacy of these combination regimens by analyzing objective response rate (ORR), overall survival (OS), and event-free survival (EFS) outcomes across multiple studies. Methods: A systematic review and meta-analysis were conducted following PRISMA guidelines. PubMed, Web of Science, and Scopus databases were searched, yielding studies comprising the related reports. Both randomized controlled trials and non-randomized studies were included. The primary outcome of interest was ORR, and the secondary outcome of interest was EFS. A random-effects model using the DerSimonian-Laird method and Knapp-Hartung-Sidik-Jonkman adjustments was employed to pool effect sizes, and heterogeneity was assessed using I2 statistics. Results: A total of ten reports from eight studies were deemed eligible and included in the meta-analysis. The pooled ORR across the studies was 0.45 (95% CI: 0.35-0.54, p < 0.001), indicating that approximately 45% of patients showed a favorable treatment response, with moderate heterogeneity (I2 = 52.78%). The pooled analysis showed an OS of 75% (95% CI: 53-96, p < 0.001), and the pooled EFS effect size was 0.59 (95% CI: 0.45-0.73, p < 0.001), despite substantial heterogeneity (I2 = 60.54%). Conclusions: anti-GD2 antibodies combined with conventional chemotherapy may significantly improve response rates and event-free survival in children with refractory or relapsed neuroblastoma. Future research should focus on identifying predictive biomarkers to tailor therapies to individual patients, enhancing both efficacy and safety in this vulnerable population.
Collapse
Affiliation(s)
- Nur Olgun
- Department of Pediatric Oncology, Institute of Oncology, Dokuz Eylul University, İzmir 35340, Turkey; (D.K.); (R.E.C.)
- Acıbadem Kent Hospital, Karsiyaka, İzmir 35630, Turkey
| | - Mehmet Emin Arayici
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Dokuz Eylul University, İzmir 35340, Turkey;
- Department of Public Health, Faculty of Medicine, Dokuz Eylul University, İzmir 35340, Turkey
| | - Deniz Kızmazoglu
- Department of Pediatric Oncology, Institute of Oncology, Dokuz Eylul University, İzmir 35340, Turkey; (D.K.); (R.E.C.)
| | - Refik Emre Cecen
- Department of Pediatric Oncology, Institute of Oncology, Dokuz Eylul University, İzmir 35340, Turkey; (D.K.); (R.E.C.)
| |
Collapse
|
16
|
Bley IA, Behrens S, Spohn M, Müller I, Schattling B. Genetic Risk Profiling Reveals Altered Glycosyltransferase Expression as a Predictor for Patient Outcome in Neuroblastoma. J Clin Med 2025; 14:527. [PMID: 39860532 PMCID: PMC11766279 DOI: 10.3390/jcm14020527] [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: 11/20/2024] [Revised: 12/17/2024] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Neuroblastoma is a highly aggressive pediatric cancer that arises from immature nerve cells and exhibits a broad spectrum of clinical presentations. While low- and intermediate-risk neuroblastomas often have favorable outcomes, high-risk neuroblastomas are associated with poor prognosis and significant treatment challenges. The complex genetic networks driving these high-risk cases remain poorly understood. This study aims to investigate differences in gene expression patterns that may contribute to disease outcomes. Methods: We employed an in silico approach to analyze a cohort of 493 neuroblastoma tumor samples that underwent mRNA sequencing (GSE49711). This dataset was reanalyzed in depth with a non-hypothesis-driven approach to identify the expression patterns and regulatory mechanisms associated with a poor prognosis. Results: By exploring global gene expression and the integration of clinical parameters, we stratified the samples into two groups with highly distinct gene expression profiles. MYCN amplification emerged as a major driver not only of poor prognosis but also of specific gene regulatory patterns. Notably, tumors with MYCN amplification exhibited the strong regulation of immune response genes and less immune infiltration, suggesting potential immune evasion. However, while we observed only minor changes in immune checkpoint expression, there was a strong modulation of glycosyltransferase genes in MYCN-amplified tumors. Using this information, we were able to construct a risk profile based on 12 glycosylation-related genes, which correlates with the survival outcomes of neuroblastoma patients. Conclusions: This study highlights the role of MYCN amplification in driving a poor prognosis in neuroblastoma through the regulation of immune response and glycosylation-related genes. Based on this finding, we developed a genetic risk profile that correlates with survival outcomes in neuroblastoma patients.
Collapse
Affiliation(s)
- Isabelle Ariane Bley
- Research Institute Children’s Cancer Center Hamburg, 20251 Hamburg, Germany
- Division of Pediatric Stem Cell Transplantation and Immunology, Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Stefan Behrens
- Research Institute Children’s Cancer Center Hamburg, 20251 Hamburg, Germany
| | - Michael Spohn
- Research Institute Children’s Cancer Center Hamburg, 20251 Hamburg, Germany
- Bioinformatics Core, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Ingo Müller
- Research Institute Children’s Cancer Center Hamburg, 20251 Hamburg, Germany
- Division of Pediatric Stem Cell Transplantation and Immunology, Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Benjamin Schattling
- Research Institute Children’s Cancer Center Hamburg, 20251 Hamburg, Germany
- Division of Pediatric Stem Cell Transplantation and Immunology, Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
17
|
Huertas-Castaño C, Martínez-López L, Cabrera-Roldán P, Pastor N, Mateos JC, Mateos S, Pardal R, Domínguez I, Orta ML. Influence of stromal neural crest progenitor cells on neuroblastoma radioresistance. Int J Radiat Biol 2025; 101:153-163. [PMID: 39750107 DOI: 10.1080/09553002.2024.2440865] [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: 09/09/2024] [Revised: 11/12/2024] [Accepted: 11/25/2024] [Indexed: 01/04/2025]
Abstract
PURPOSE A substantial proportion of children with high risk Neuroblastoma die within the first 5 years post-diagnosis despite the complex treatment applied. In the recent years, tumor environment has been revealed as key factor for cancer treatment efficacy. In this sense, non-tumorigenic Neural Crest progenitor cells from high risk patients, have been described as part of Neuroblastoma stroma, promoting tumor growth and contributing to mesenchyme formation. In this paper we wanted to study the radiobiological behavior of these cells (NB14t) and how they influence the growth of tumorigenic neuroblasts after radiotherapy. MATERIALS AND METHODS To achieve our aim, we employed a wide list of methods either using NB14t cells as well as commercial NB cells. We have analyzed viability, survival, cell cyle profiles and differentiation. In addition, cocultured experiments were performed to monitor the influence of stroma cells to tumorigenic neuroblasts. RESULTS We found that stromal progenitor cells showed an extraordinary radio-resistance either cultured in attached or suspension conditions. In good agreement, we found an enhanced repair of irradiation-induced DNA lesions as compared with commercial cell lines. In addition, according to our data these cells differentiate into a Cancer Associated Fibroblasts (CAFs)-like phenotype, hence contributing to the formation of mesenchymal stroma enhancing the growth of tumor cells after irradiation. CONCLUSION Our data show that neural progenitor cells from high risk NB stroma are radio-resistant and promote cancer growth after irradiation. This paper can help to understand the complex cell relationships within a tumor that will determine patient prognosis after radiotherapy.
Collapse
Affiliation(s)
| | | | - Patricia Cabrera-Roldán
- Departamento de Oncología Radioterápica, Hospital Universitario Virgen del Rocío (HUVR), Seville, Spain
| | - Nuria Pastor
- Departamento de Biología Celular, Universidad de Sevilla, Seville, Spain
| | - Juan Carlos Mateos
- Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, Seville, Spain
| | - Santiago Mateos
- Departamento de Biología Celular, Universidad de Sevilla, Seville, Spain
| | - Ricardo Pardal
- Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, Seville, Spain
- Instituto de Biomedicina de Sevilla (IBiS) (Universidad de Sevilla, HUVR, Junta de Andalucía, CSIC), Seville, Spain
| | | | - Manuel Luis Orta
- Departamento de Biología Celular, Universidad de Sevilla, Seville, Spain
| |
Collapse
|
18
|
Murra N, Pommert NS, Schmidt B, Issa RS, Kaehler M, Bruckmueller H, Tim V, Cascorbi I, Waetzig V. Regulation and Function of CCL2 and N-Myc in Retinoic Acid-treated Neuroblastoma Cells. Cancer Genomics Proteomics 2025; 22:90-102. [PMID: 39730182 PMCID: PMC11696317 DOI: 10.21873/cgp.20490] [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: 09/29/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 12/29/2024] Open
Abstract
BACKGROUND/AIM Treatment with retinoic acid (RA) often promotes neuroblastoma differentiation and growth inhibition, including the suppression of the expression of the MYCN oncogene. However, RA also targets protumoral chemokines, such as CCL2, which may contribute to the development of resistance. The present study aimed to investigate the regulation and function of CCL2 and N-Myc in RA-treated neuroblastoma cells. MATERIALS AND METHODS In Kelly or SH-SY5Y cells, viability was quantified by cell fitness assays. Expression was analyzed using quantitative PCR and the regulation of proteins using enzyme-linked immunoabsorbent assays (ELISA) or western blots. RESULTS In MYCN-amplified Kelly cells, endogenous CCL2 levels were significantly lower compared to MYCN non-amplified SH-SY5Y cells. Treatment with 5 μM RA increased CCL2 release in both cell lines, but reduced N-Myc levels and cell numbers in Kelly cells. Over-expression of MYCN enhanced viability in SH-SY5Y cells, but did not affect RA-induced CCL2 release, while supplementation of CCL2 in Kelly cells did not prevent RA-mediated growth reduction. Impaired N-Myc or CCL2 signaling reduced the survival of all RA-treated cells and inhibition of N-Myc also decreased CCL2 levels. However, attenuated survival signaling was not generally associated with reduced levels of N-Myc or CCL2. Co-application of RA and the growth factor receptor inhibitors cediranib or crizotinib decreased N-Myc levels only in Kelly cells, while CCL2 release was dependent on the cell type and stimulus. CONCLUSION CCL2 and N-Myc promote the viability of RA-treated cells, although the levels of these mediators were not consistently correlated with cellular outcomes, especially during apoptotic signaling.
Collapse
Affiliation(s)
- Nanke Murra
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Nina Sophie Pommert
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Berit Schmidt
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Reema Sami Issa
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Meike Kaehler
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Henrike Bruckmueller
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Vera Tim
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Ingolf Cascorbi
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Vicki Waetzig
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| |
Collapse
|
19
|
Lin H, Liao F, Liu J, Yang Z, Zhang J, Cheng J, Zhou H, Li S, Li L, Li Y, Zhuo Z, He J. Neuroblastoma susceptibility and association of N7-methylguanosine modification gene polymorphisms: multi-center case-control study. Pediatr Res 2025; 97:153-159. [PMID: 38871802 DOI: 10.1038/s41390-024-03318-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 04/02/2024] [Accepted: 05/18/2024] [Indexed: 06/15/2024]
Abstract
BACKGROUND Neuroblastoma (NB) is a common extracranial solid malignancy in children. The N7-methylguanosine (m7G) modification gene METTL1/WDR4 polymorphisms may serve as promising molecular markers for identifying populations susceptible to NB. METHODS TaqMan probes was usded to genotype METTL1/WDR4 single nucleotide polymorphisms (SNPs) in 898 NB patients and 1734 healthy controls. A logistic regression model was utilized to calculate the odds ratio (OR) and 95% confidence interval (CI), evaluating the association between genotype polymorphisms and NB susceptibility. The analysis was also stratified by age, sex, tumor origin site, and clinical stage. RESULTS Individual polymorphism of the METTL1/WDR4 gene investigated in this study did not show significant associations with NB susceptibility. However, combined genotype analysis revealed that carrying all 5 WDR4 protective genotypes was associated with a significantly lower NB risk compared to having 0-4 protective genotypes (AOR = 0.82, 95% CI = 0.69-0.96, P = 0.014). Further stratified analyses revealed that carrying 1-3 METTL1 risk genotypes, the WDR4 rs2156316 CG/GG genotype, the WDR4 rs2248490 CG/GG genotype, and having all five WDR4 protective genotypes were all significantly correlated with NB susceptibility in distinct subpopulations. CONCLUSIONS In conclusion, our findings suggest significant associations between m7G modification gene METTL1/WDR4 SNPs and NB susceptibility in specific populations. IMPACT Genetic variation in m7G modification gene is associated with susceptibility to NB. Single nucleotide polymorphisms in METTL1/WDR4 are associated with susceptibility to NB. Single nucleotide polymorphisms of METTL1/WDR4 can be used as a biomarker for screening NB susceptible populations.
Collapse
Affiliation(s)
- Huiran Lin
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
- Faculty of Medicine, Macau University of Science and Technology, Macau, 999078, China
| | - Fan Liao
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Jiabin Liu
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Zhonghua Yang
- Department of Pediatric Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China
| | - Jiao Zhang
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Jiwen Cheng
- Department of Pediatric Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Haixia Zhou
- Department of Hematology, The Key Laboratory of Pediatric Hematology and Oncology Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Suhong Li
- Department of Pathology, Children Hospital and Women Health Center of Shanxi, Taiyuan, 030013, Shannxi, China
| | - Li Li
- Kunming Key Laboratory of Children Infection and Immunity, Yunnan Key Laboratory of Children's Major Disease Research, Yunnan Institute of Pediatrics Research, Yunnan Medical Center for Pediatric Diseases, Kunming Children's Hospital, Kunming, 650228, Yunnan, China
| | - Yong Li
- Department of Pediatric Surgery, Hunan Children's Hospital, Changsha, 410004, Hunan, China
| | - Zhenjian Zhuo
- Laboratory Animal Center, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China.
- Faculty of Medicine, Macau University of Science and Technology, Macau, 999078, China.
| |
Collapse
|
20
|
Wang Y, Cao N, Cui X, Liu Z, Yuan X, Chen S, Xu H, Yi M, Ti Y, Zheng F, Cai K. Detection of circulating tumor cells using a microfluidic chip for diagnostics and therapeutic prediction in mediastinal neuroblastoma. Eur J Pediatr 2024; 184:93. [PMID: 39702653 DOI: 10.1007/s00431-024-05896-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 11/15/2024] [Accepted: 11/20/2024] [Indexed: 12/21/2024]
Abstract
Circulating tumor cells (CTCs) have served as noninvasive tumor biomarkers in many types of cancer. Here, we detected CTCs in mediastinal neuroblastoma (mNB) patients for use as diagnostic and treatment response predictive biomarkers. We employed a cascaded filter deterministic lateral displacement microfluidic chip (CFD-Chip) to enrich CTCs in peripheral blood from 32 mNB patients and 7 healthy children. CTCs were identified by immunofluorescence staining and integrated neoplastic cell morphology. In total, 66.67% of newly diagnosed mNB patients were positive for CTCs while no CTCs were detected in healthy children. Moreover, CTC count differed significantly across different International Neuroblastoma Staging System, International Neuroblastoma Risk Group staging system, and risk stratifications. CTC count was also significantly higher in children with metastasis than those without metastasis. Additionally, CTC demonstrated a significant difference among patients with different clinical responses to therapy. CTC count decreased or fluctuated at low levels in patients with complete and partial response, compared to considerably increased in patients with stable and progressive diseases.Conclusion: CTCs may serve as non-invasive indicators for mNB diagnosis, staging, and metastasis prediction, and demonstrate promising potential as a liquid biopsy biomarker for the dynamic monitoring of therapeutic efficacy.
Collapse
Affiliation(s)
- Yuanxiang Wang
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Cardiothoracic Surgery, Shenzhen Children's Hospital, Shenzhen, 518038, China
| | - Nianhua Cao
- Shenzhen Zigzag Biotechnology Co., Ltd, Shenzhen, 518107, China
| | - Xiufang Cui
- Shenzhen Zigzag Biotechnology Co., Ltd, Shenzhen, 518107, China
| | - Zongbin Liu
- Shenzhen Zigzag Biotechnology Co., Ltd, Shenzhen, 518107, China
| | - Xiuli Yuan
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, 518038, China
| | - Senmin Chen
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, 518038, China
| | - Huanli Xu
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, 518038, China
| | - Meng Yi
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, 518038, China
| | - Yunxing Ti
- Department of Cardiothoracic Surgery, Shenzhen Children's Hospital, Shenzhen, 518038, China
| | - Fengnan Zheng
- Department of Cardiothoracic Surgery, Shenzhen Children's Hospital, Shenzhen, 518038, China
| | - Kaican Cai
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
21
|
Hergenhahn L, Padutsch N, Azawi S, Weiskirchen R, Liehr T, Rinčic M. Cytogenomic Characterization of Murine Neuroblastoma Cell Line Neuro-2a and Its Two Derivatives Neuro-2a TR-Alpha and Neuro-2a TR-Beta. Cells 2024; 13:1889. [PMID: 39594637 PMCID: PMC11593031 DOI: 10.3390/cells13221889] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/07/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND The Neuro-2a cell line, derived from a murine neuroblastoma (NB), was established as early as 1969 and originates from a transplantable tumor that arose spontaneously in an A/Jax male mouse in 1940. Since then, it has been applied in over 10,000 studies and is used by the World Organization for Animal Health for the routine diagnosis of rabies. Surprisingly, however, Neuro-2a has never been genetically characterized in detail; this study fills that gap. METHODS The Neuro-2a cell line and two of its derivatives, Neuro-2a TR-alpha and Neuro-2a TR-beta, were analyzed for their chromosomal constitution using molecular cytogenetic approaches. Array comparative genomic hybridization was performed to characterize copy number alterations. RESULTS Neuro-2A has a hyper-tetraploid karyotype with 70 to 97 chromosomes per cell, and the karyotypes of its two examined derivatives were quite similar. Neither of them had a Y-chromosome. The complex karyotype of Neuro-2a includes mitotically stable dicentres, neocentrics, and complex rearrangements resembling chromothripsis events. Although no amplification of euchromatin or oncogenes was detected, there are five derivative chromosomes with the amplification of centromere-near heterochromatic material and 1-5 additional derivatives consisting only of such material. CONCLUSIONS Since satellite DNA amplification has recently been found in advanced human tumors, this finding may be the corresponding equivalent in mice. An in silico translation of the obtained results into the human genome indicated that Neuro-2A is suitable as a model for advanced human NB.
Collapse
Affiliation(s)
- Lioba Hergenhahn
- Jena University Hospital, Friedrich Schiller University, Institute of Human Genetics, D-07747 Jena, Germany; (L.H.); (N.P.); (S.A.)
| | - Niklas Padutsch
- Jena University Hospital, Friedrich Schiller University, Institute of Human Genetics, D-07747 Jena, Germany; (L.H.); (N.P.); (S.A.)
| | - Shaymaa Azawi
- Jena University Hospital, Friedrich Schiller University, Institute of Human Genetics, D-07747 Jena, Germany; (L.H.); (N.P.); (S.A.)
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH, University Hospital Aachen, D-52074 Aachen, Germany;
| | - Thomas Liehr
- Jena University Hospital, Friedrich Schiller University, Institute of Human Genetics, D-07747 Jena, Germany; (L.H.); (N.P.); (S.A.)
| | - Martina Rinčic
- Croatian Institute for Brain Research, School of Medicine University of Zagreb, C-10000 Zagreb, Croatia;
| |
Collapse
|
22
|
Fan J, Tang S, Kong X, Cun Y. Integrating multi-omics data reveals neuroblastoma subtypes in the tumor microenvironment. Life Sci 2024; 359:123236. [PMID: 39532261 DOI: 10.1016/j.lfs.2024.123236] [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: 09/02/2024] [Revised: 11/04/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
Neuroblastoma (NB) is a severe pediatric tumor originating from the developing sympathetic nervous system, characterized by diverse clinical outcomes, including spontaneous regression and aggressive metastasis. This variability suggests the existence of different NB subtypes, necessitating accurate classification for effective targeted treatment. In this study, we employed the similarity network fusion (SNF) algorithm and identified three NB subtypes, including mesenchymal-like (MES), MYCN-like (MYCN), and neurogenic-like (Neurogenic). The MES subtype exhibited the highest activation of immune-related pathways. The MYCN subtype demonstrated the worst prognosis, with enrichment in cell growth and proliferation pathways. Conversely, the Neurogenic subtype showed the best prognosis, with enrichment in sympathetic nervous system development processes. Through single-cell RNA sequencing (scRNA-seq) analysis, we examined the tumor microenvironments of these distinct NB subtypes, revealing divergent differentiation trajectories for adrenergic cells within the MYCN and Neurogenic subtypes. We also identified a significant presence of naïve T cells in the MES subtype, as well as mesenchymal cell subtypes associated with the unique plasticity observed in both the MES and MYCN subtypes. Drug sensitivity prediction analysis suggested that the MES subtype may respond favorably to MEK inhibitors, while the MYCN subtype may be susceptible to Bcl-2 inhibitors. Our integrative multi-omics approach enabled precise stratification of NB into biologically distinct subtypes, potentially facilitating the development of subtype-specific therapeutic strategies for improved patient management and survival outcomes.
Collapse
Affiliation(s)
- Jinhua Fan
- Pediatric Research Institute, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Shuxin Tang
- Pediatric Research Institute, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Xiangru Kong
- Departments of Oncological Surgery, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Yupeng Cun
- Pediatric Research Institute, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China.
| |
Collapse
|
23
|
Zhang L, Li H, Sun F, Wu Q, Jin L, Xu A, Chen J, Yang R. Identification of novel markers for neuroblastoma immunoclustering using machine learning. Front Immunol 2024; 15:1446273. [PMID: 39559348 PMCID: PMC11570813 DOI: 10.3389/fimmu.2024.1446273] [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: 06/09/2024] [Accepted: 10/15/2024] [Indexed: 11/20/2024] Open
Abstract
Background Due to the unique heterogeneity of neuroblastoma, its treatment and prognosis are closely related to the biological behavior of the tumor. However, the effect of the tumor immune microenvironment on neuroblastoma needs to be investigated, and there is a lack of biomarkers to reflect the condition of the tumor immune microenvironment. Methods The GEO Database was used to download transcriptome data (both training dataset and test dataset) on neuroblastoma. Immunity scores were calculated for each sample using ssGSEA, and hierarchical clustering was used to categorize the samples into high and low immunity groups. Subsequently, the differences in clinicopathological characteristics and treatment between the different groups were examined. Three machine learning algorithms (LASSO, SVM-RFE, and Random Forest) were used to screen biomarkers and synthesize their function in neuroblastoma. Results In the training set, there were 362 samples in the immunity_L group and 136 samples in the immunity_H group, with differences in age, MYCN status, etc. Additionally, the tumor microenvironment can also affect the therapeutic response of neuroblastoma. Six characteristic genes (BATF, CXCR3, GIMAP5, GPR18, ISG20, and IGHM) were identified by machine learning, and these genes are associated with multiple immune-related pathways and immune cells in neuroblastoma. Conclusions BATF, CXCR3, GIMAP5, GPR18, ISG20, and IGHM may serve as biomarkers that reflect the conditions of the immune microenvironment of neuroblastoma and hold promise in guiding neuroblastoma treatment.
Collapse
Affiliation(s)
- Longguo Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Huixin Li
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Fangyan Sun
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Qiuping Wu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Leigang Jin
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Jiarui Chen
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Ranyao Yang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Clinical Pharmacy, Jining First People’s Hospital, Shandong First Medical University, Jining, China
| |
Collapse
|
24
|
Inuwa RF, Moss D, Quayle J, Swadi R. Epac activation reduces trans-endothelial migration of undifferentiated neuroblastoma cells and cellular differentiation with a CDK inhibitor further enhances Epac effect. PLoS One 2024; 19:e0304547. [PMID: 39495719 PMCID: PMC11534210 DOI: 10.1371/journal.pone.0304547] [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: 05/14/2024] [Accepted: 10/18/2024] [Indexed: 11/06/2024] Open
Abstract
Neuroblastoma (NB) is the most common solid extracranial neoplasm found in children and is derived from primitive sympathoadrenal neural precursor. The disease accounts for 15% of all cancer deaths in children. The mortality rate is high in patients presenting with a metastatic tumour even with extensive treatments. This signifies the need for further research towards the development of new additional therapies that can combat not only tumour growth but metastasis, especially amongst the high-risk groups. During metastasis, primary tumour cells become migratory and travel towards a capillary within the tumour. They then degrade the matrix surrounding the pericytes and endothelial cells traversing the endothelial barrier twice to establish a secondary. This led to the hypothesis that modulation of the endothelial cell junctional stability could have an influence on tumour metastasis. To test this hypothesis, agents that modulate endothelial permeability on NB cell line migration and invasion were assessed in vitro in a tissue culture model. The cAMP agonist and its antagonists were found to have no obvious effect on both SK-N-BE2C and SK-N-AS migration, invasion and proliferation. Next, NB cells were cocultured with HDMEC cells and live cell imaging was used to assess the effect of an Epac agonist on trans-endothelial cell migration of NB cells. Epac1 agonist remarkably reduced the trans-endothelial migration of both SK-N-BE2C and SK-N-AS cells. These results demonstrate that an Epac1 agonist may perhaps serve as an adjuvant to currently existing therapies for the high-risk NB patients.
Collapse
Affiliation(s)
- Rabiu Fage Inuwa
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
- Faculty of Basic Medical Sciences, Department of Anatomy, College of Health Sciences, Bayero University, Kano, Nigeria
| | - Diana Moss
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - John Quayle
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
- ALESS/ALESA Program, Center for Global Communication Strategies, Center for Global Education, University of Tokyo, Bunkyō, Japan
| | - Rasha Swadi
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, United Kingdom
- Institute for Health Research, Liverpool John Moores University, Liverpool, United Kingdom
| |
Collapse
|
25
|
Meng X, Tan Z, Qiu B, Zhang J, Wang R, Ni W, Fan J. METTL3-induced lncARSR aggravates neuroblastoma tumorigenic properties through stabilizing PHOX2B. Pathol Res Pract 2024; 263:155670. [PMID: 39461245 DOI: 10.1016/j.prp.2024.155670] [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: 05/20/2024] [Revised: 08/25/2024] [Accepted: 10/19/2024] [Indexed: 10/29/2024]
Abstract
Neuroblastoma (NB), the most common extracranial solid tumor in pediatric patients, manifests with considerable variability across multiple primary sites. Despite this, the extent of genetic heterogeneity within these tumor foci and the identification of consistent oncogenic drivers remains largely unexplored. Of particular interest, genetic mutations in PHOX2B have been linked to familial cases of NB, yet the underlying molecular mechanisms are not fully delineated. In our research, we focus on unraveling the role of a novel functional long non-coding RNA (lncRNA) associated with PHOX2B in the context of NB. Using NB cell models with overexpressed PHOX2B, combined with lncRNA microarray analysis, we discovered that lncARSR is significantly upregulated in response to PHOX2B overexpression. Subsequent biological assays demonstrated that lncARSR promotes both the proliferation and metastasis of NB cells. Further molecular investigations revealed that lncARSR plays a crucial role in stabilizing PHOX2B expression within NB cells. Moreover, we identified that the expression of lncARSR is regulated by methylation through methyltransferase-like 3 (METTL3), which itself is positively correlated with PHOX2B expression. Rescue experiments underscored the functional importance of METTL3, lncARSR, and PHOX2B in NB cells. In summary, our findings provide new insights into the molecular functions of PHOX2B in the progression of neuroblastoma and propose a novel therapeutic target for this aggressive malignancy.
Collapse
Affiliation(s)
- Xiangyi Meng
- Department of Pediatrics, Shenzhen University General Hospital, China
| | - Zhu Tan
- Department of Pediatrics, Shenzhen University General Hospital, China
| | - Bihua Qiu
- Department of Pediatrics, Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine, China
| | - Jie Zhang
- Department of Pediatrics, Shenzhen University General Hospital, China
| | - Ruobing Wang
- Department of Pediatrics, Shenzhen University General Hospital, China
| | - Wensi Ni
- Department of Pediatrics, Shenzhen University General Hospital, China.
| | - Jialing Fan
- Department of Pediatrics, Shenzhen University General Hospital, China.
| |
Collapse
|
26
|
Wu H, Zhang G, Liu Z, Liu W, Wang X, Zhao Y. Enhanced anti-tumor activity mediated by combination chimeric antigen receptor T cells targeting GD2 and GPC2 in high-risk neuroblastoma. Cytotherapy 2024; 26:1308-1319. [PMID: 38904586 DOI: 10.1016/j.jcyt.2024.05.023] [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: 01/26/2024] [Revised: 05/26/2024] [Accepted: 05/27/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND AIMS Chimeric antigen receptor T (CAR-T) cells targeting single antigens show limited activity against solid tumors due to poor T cell persistence, low efficiency infiltration, and exhaustion together with heterogeneous tumor-associated antigen (TAA) expression. This is also true in high-risk neuroblastoma (HRNB), a lethal pediatric extracranial malignancy. To overcome these obstacles, a combinational strategy using GD2-specific and GPC2-specific CAR-T cells was developed to improve immunotherapeutic efficacy. METHODS We individually developed GD2-specific and GPC2-specific CARs containing a selective domain (sCAR) which was a peptide of 10 amino acids derived from human nuclear autoantigen La/SS-B. These constructs allowed us to generate two different HRNB antigen-specific CAR-T cells with enhanced biological activity through stimulating sCAR-engrafted T cells via a selective domain-specific monoclonal antibody (SmAb). Binding affinity and stimulation of GD2- and GPC2-specific sCARs by SmAb were measured, and transient and persistent anti-tumor cytotoxicity of GD2sCAR-T and GPC2sCAR-T cells were quantified in neuroblastoma cell lines expressing different TAA levels. The anti-tumor pharmaceutical effects and cellular mechanisms mediated by single or combinational sCAR-T cells were evaluated in vitro and in vivo. RESULTS GD2- and GPC2-specific sCARs had antigen-specific binding affinity similar to their parental counterparts and were recognized by SmAb. SmAb-mediated stimulation selectively activated sCAR-T proliferation and increased central memory T cells in the final products. SmAb-stimulated sCAR-T cells had enhanced transient cytolytic activity, and combination therapy extended long-term anti-tumor activity in vitro through TNF-α and IL-15 release. Stimulated sCAR-T cells overcame heterogeneous antigen expression in HRNB, and the multi-TAA-targeting strategy was especially efficacious in vivo, inducing apoptosis through the caspase-3/PARP pathway and inhibiting the release of several tumor-promoting cytokines. CONCLUSIONS These data suggest that combined targeting of multiple TAAs is a promising strategy to overcome heterogenous antigen expression in solid tumors and extend CAR-T cell persistence for HRNB immunotherapy.
Collapse
Affiliation(s)
- Huantong Wu
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
| | - Guangji Zhang
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
| | - Zhongfeng Liu
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
| | - Weihua Liu
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
| | - Xuan Wang
- Department of Oncology, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong Province, China
| | - Yu Zhao
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China.
| |
Collapse
|
27
|
Huang S, Yang X, Gao Y, Huang H, Li T, Li M, Wu F, Yang H, Li C. Multifunctional nano co-delivery system for efficiently eliminating neuroblastoma by overcoming cancer heterogeneity. Biomed Mater 2024; 19:065033. [PMID: 39419089 DOI: 10.1088/1748-605x/ad8826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 10/17/2024] [Indexed: 10/19/2024]
Abstract
The high heterogeneity of neuroblastoma (NB) is currently the main challenge in clinical treatment, impeding the complete eradication of the tumor through monotherapy alone. In this study, we propose a combination strategy using a targeted nano co-delivery system (ADRF@Ag2Se) comprising phototheranostic agents, differentiation inducers and chemotherapy drugs for sequential therapy of NB. Upon intravenous injection, ADRF@Ag2Se demonstrates effective tumor targeting by the specific binding of AF7P to MMP14, which is overexpressed on the surface of NB cells. Subsequent implementation of local photothermal therapy (PTT) leverages the robust photothermal conversion capabilities of the amphiphilic photothermal reagent PF. This is followed by the temperature-triggered release of differentiation-inducing agent 13-cis-retinoic acid and chemo-drug doxorubicin to synergistically eliminate the residual lesions. This nanotherapeutic strategy facilitatesin vivotargeted delivery and PTT under the supervision of NIR-II fluorescence, and it also enhances the chemotherapeutic response through differentiation induction of poorly differentiated cancer cells. In the NB tumor model, this co-delivery strategy effectively inhibited tumor growth and significantly prolonged the survival of the mice.
Collapse
Affiliation(s)
- Shungen Huang
- Pediatric Surgery, Children's Hospital of Soochow University, Suzhou 215025, People's Republic of China
| | - Xian Yang
- Pediatric Surgery, Children's Hospital of Soochow University, Suzhou 215025, People's Republic of China
- Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Yajuan Gao
- Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Haoying Huang
- Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Tuanwei Li
- Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Meng Li
- Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Feng Wu
- Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Hongcao Yang
- Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Chunyan Li
- Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| |
Collapse
|
28
|
Marimpietri D, Corrias MV, Tripodi G, Gramignoli R, Airoldi I, Morandi F. Immunomodulatory properties of extracellular vesicles isolated from bone marrow of patients with neuroblastoma: role of PD-L1 and HLA-G. Front Immunol 2024; 15:1469771. [PMID: 39512342 PMCID: PMC11540764 DOI: 10.3389/fimmu.2024.1469771] [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: 07/24/2024] [Accepted: 10/03/2024] [Indexed: 11/15/2024] Open
Abstract
Introduction Extracellular vesicles (EVs) can be released by any cell and are crucial for cell-to-cell communications. EVs have been characterized in patients with solid and hematological tumors, where they play an important role in tumor progression and metastasis. EVs may express different surface proteins derived from the parental cells, including immunomodulatory molecules, such as HLA-G and PDL1. Methods We isolated EV from bone marrow (BM) samples of patients with Neuroblastoma (NB) and healthy controls and we analyzed the expression of CD56, GD2 and immune checkpoints on EV by flow cytometry. Next, we analyzed the function of T cells in vitro in the presence or absence of NB patients' BM-derived EV, in terms of proliferation and cytokine production. Finally, we analyzed the correlation between the expression of immune checkpoints on EV and the clinical outcome of patients. Results We found a higher expression of CD56 on EVs derived from BM of patients with NB than in those from healthy donors (HD). However, CD56 expression was not dependent on BM infiltration of NB cells. Moreover, the analysis of GD2 expression revealed that only a small fraction of EVs was released by infiltrating NB cells, whereas the majority may derive from BM-resident cells. BM-derived EVs from NB patients display a higher expression of HLA-G and PD-L1 than those derived from HD. Nonetheless, such EVs are able to modulate T cell immune responses. We measured a robust response, in vitro, towards a common bacterial antigen, including the release of GM-CSF and proinflammatory cytokines, like IFN-a and IL-6, from mononuclear cells. Some of these immunomodulatory features are dependent on the expression of HLA-G and PD-L1, whereas others may rely on other mechanism(s). Finally, a high expression of CD56, HLA-G and PD-L1 on BM-derived EVs may represent a good prognostic factor. Conclusions We described the presence of HLA-G and PDL1-bearing EVs in the BM of NB patients, which may represent a mechanism performed by resident BM cells to counteract the inflammation occurring in the BM microenvironment of NB patients.
Collapse
Affiliation(s)
- Danilo Marimpietri
- UOSD Laboratorio di Terapie Cellulari, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Maria Valeria Corrias
- UOSD Terapie Sperimentali in Oncologia, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Gino Tripodi
- UOC Servizio di Immunoematologia e Medicina Trasfusionale, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Roberto Gramignoli
- UOSD Laboratorio di Terapie Cellulari, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Irma Airoldi
- UOSD Laboratorio di Terapie Cellulari, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Fabio Morandi
- UOSD Laboratorio di Terapie Cellulari, IRCCS Istituto Giannina Gaslini, Genova, Italy
| |
Collapse
|
29
|
Yuan K, Tang Y, Ding Z, Peng L, Zeng J, Wu H, Yi Q. Mutant ATRX: pathogenesis of ATRX syndrome and cancer. Front Mol Biosci 2024; 11:1434398. [PMID: 39479502 PMCID: PMC11521912 DOI: 10.3389/fmolb.2024.1434398] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 10/04/2024] [Indexed: 11/02/2024] Open
Abstract
The transcriptional regulator ATRX, a genetic factor, is associated with a range of disabilities, including intellectual, hematopoietic, skeletal, facial, and urogenital disabilities. ATRX mutations substantially contribute to the pathogenesis of ATRX syndrome and are frequently detected in gliomas and many other cancers. These mutations disrupt the organization, subcellular localization, and transcriptional activity of ATRX, leading to chromosomal instability and affecting interactions with key regulatory proteins such as DAXX, EZH2, and TERRA. ATRX also functions as a transcriptional regulator involved in the pathogenesis of neuronal disorders and various diseases. In conclusion, ATRX is a central protein whose abnormalities lead to multiple diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Huaying Wu
- Key Laboratory of Model Animals and Stem Cell Biology, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Qi Yi
- Key Laboratory of Model Animals and Stem Cell Biology, Hunan Normal University School of Medicine, Changsha, Hunan, China
| |
Collapse
|
30
|
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
|
31
|
Xue L, Luo K, Hou K, Huo W, Ruan P, Xue Y, Yao X, Meng C, Xia D, Tang Y, Zhao W, Yuan H, Zhao L, Gao L, Yuan Q, Gao X, Cao K. Targeted Gold Nanoclusters for Synergistic High-Risk Neuroblastoma Therapy through Noncanonical Ferroptosis. ACS APPLIED MATERIALS & INTERFACES 2024; 16:53555-53566. [PMID: 39327976 DOI: 10.1021/acsami.4c11979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
Children with extracranial high-risk neuroblastoma (NB) have a poor prognosis due to resistance against apoptosis. Recently, ferroptosis, another form of programmed cell death, has been tested in clinical trials for high-risk NB; however, drug resistance and side effects have also been observed. Here, we find that the gold element in gold nanoclusters can significantly affect iron metabolism and sensitize high-risk NB cells to ferroptosis. Accordingly, we developed a gold nanocluster conjugated with a modified NB-targeting peptide. This gold nanocluster, namely, NANT, shows excellent NB targeting efficiency and dramatically promotes ferroptosis. Surprisingly, this effect is exerted by elevating the noncanonical ferroptosis pathway, which is dependent on heme oxygenase-1-regulated Fe(II) accumulation. Furthermore, NANT dramatically inhibits the growth of high-risk NB in both tumor spheroid and xenograft models by promoting noncanonical ferroptosis evidenced by enhanced intratumoral Fe(II) and heme oxygenase-1. Importantly, this strategy shows excellent cardiosafety, offering a promising strategy to overcome ferroptosis resistance for the efficient and safe treatment of children with high-risk neuroblastoma.
Collapse
Affiliation(s)
- Liyuan Xue
- Department of Chemistry, College of Chemistry and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Kaidi Luo
- Department of Chemistry, College of Chemistry and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Kaixiao Hou
- Department of Chemistry, College of Chemistry and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Wendi Huo
- Department of Chemistry, College of Chemistry and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Panpan Ruan
- Department of Chemistry, College of Chemistry and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Yilin Xue
- Department of Chemistry, College of Chemistry and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Xiuxiu Yao
- Department of Chemistry, College of Chemistry and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Cong Meng
- Department of Chemistry, College of Chemistry and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Dongfang Xia
- Department of Chemistry, College of Chemistry and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Yuhua Tang
- Department of Chemistry, College of Chemistry and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Wencong Zhao
- Department of Chemistry, College of Chemistry and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Hui Yuan
- Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Liang Zhao
- Department of Chemistry, College of Chemistry and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Liang Gao
- Department of Chemistry, College of Chemistry and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Qing Yuan
- Department of Chemistry, College of Chemistry and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Xueyun Gao
- Department of Chemistry, College of Chemistry and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Kai Cao
- Department of Chemistry, College of Chemistry and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| |
Collapse
|
32
|
Xu M, Hong JJ, Zhang X, Sun M, Liu X, Kang J, Stack H, Fang W, Lei H, Lacoste X, Okada R, Jung R, Nguyen R, Shern JF, Thiele CJ, Liu Z. Targeting SWI/SNF ATPases reduces neuroblastoma cell plasticity. EMBO J 2024; 43:4522-4541. [PMID: 39174852 PMCID: PMC11480351 DOI: 10.1038/s44318-024-00206-1] [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: 09/26/2023] [Revised: 07/01/2024] [Accepted: 07/17/2024] [Indexed: 08/24/2024] Open
Abstract
Tumor cell heterogeneity defines therapy responsiveness in neuroblastoma (NB), a cancer derived from neural crest cells. NB consists of two primary subtypes: adrenergic and mesenchymal. Adrenergic traits predominate in NB tumors, while mesenchymal features becomes enriched post-chemotherapy or after relapse. The interconversion between these subtypes contributes to NB lineage plasticity, but the underlying mechanisms driving this phenotypic switching remain unclear. Here, we demonstrate that SWI/SNF chromatin remodeling complex ATPases are essential in establishing an mesenchymal gene-permissive chromatin state in adrenergic-type NB, facilitating lineage plasticity. Targeting SWI/SNF ATPases with SMARCA2/4 dual degraders effectively inhibits NB cell proliferation, invasion, and notably, cellular plasticity, thereby preventing chemotherapy resistance. Mechanistically, depletion of SWI/SNF ATPases compacts cis-regulatory elements, diminishes enhancer activity, and displaces core transcription factors (MYCN, HAND2, PHOX2B, and GATA3) from DNA, thereby suppressing transcriptional programs associated with plasticity. These findings underscore the pivotal role of SWI/SNF ATPases in driving intrinsic plasticity and therapy resistance in neuroblastoma, highlighting an epigenetic target for combinational treatments in this cancer.
Collapse
Affiliation(s)
- Man Xu
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Jason J Hong
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Xiyuan Zhang
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Ming Sun
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Xingyu Liu
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Jeeyoun Kang
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Hannah Stack
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Wendy Fang
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Haiyan Lei
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Xavier Lacoste
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Reona Okada
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Raina Jung
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Rosa Nguyen
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Jack F Shern
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Carol J Thiele
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| | - Zhihui Liu
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
33
|
Zhao Z, Yang C. Predictive value of 18 F-FDG PET/CT versus bone marrow biopsy and aspiration in pediatric neuroblastoma. Clin Exp Metastasis 2024; 41:627-638. [PMID: 38609536 DOI: 10.1007/s10585-024-10286-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/04/2024] [Indexed: 04/14/2024]
Abstract
BACKGROUND Neuroblastoma (NB) is the most prevalent solid extracranial malignancy in children, often with bone marrow metastases (BMM) are present. The conventional approach for detecting BMM is bone marrow biopsy and aspiration (BMBA). 18 F-fluorodeoxyglucose-positron emission tomography/computed tomography (18 F-FDG PET/CT) has become a staple for staging and is also capable of evaluating marrow infiltration. The consensus on the utility of 18 F-FDG PET/CT for assessing BMM in NB patients is still under deliberation. METHODS This retrospective study enrolled 266 pediatric patients with pathologically proven NB. All patients had pretherapy FDG PET/CT. BMBA, clinical, radiological, and follow-up data were also collected. The diagnostic accuracy of BMBA and 18 F-FDG PET/CT was assessed. RESULTS BMBAs identified BMM in 96 cases (36.1%), while 18 F-FDG PET/CT detected BMI in 106 cases (39.8%) within the cohort. The initial sensitivity, positive predictive value (PPV), specificity, and negative predictive value (NPV) of 18 F-FDG PET/CT were 93.8%, 84.9%, 90.6%, and 96.3%, respectively. After treatment, these values were 92.3%, 70.6%, 97.3%, and 99.4%, respectively. The kappa statistic, which measures agreement between BMBA and 18 F-FDG PET/CT, was 0.825 before treatment and 0.784 after treatment, with both values indicating a substantial agreement (P = 0.000). Additionally, the amplification of MYCN and a positive initial PET/CT scan were identified as independent prognostic factors for overall survival (OS). CONCLUSION 18 F-FDG-PET/CT is a valuable method for evaluating BMM in NB. The routine practice of performing a BMBA without discrimination may need to be reassessed. Negative result from 18 F-FDG-PET/CT could potentially spare children with invasive bone marrow biopsies.
Collapse
Affiliation(s)
- Zhenzhen Zhao
- Department of Surgical oncology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Chao Yang
- Department of Surgical oncology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
- , 136 Zhongshan 2nd Road, Yuzhong District, Chongqing, 400014, China.
| |
Collapse
|
34
|
Zeng C, Li Z, Wei Z, Chen T, Wang J, Huang J, Sun F, Zhu J, Lu S, Zhen Z. Mechanism of Drug Resistance to First-Line Chemotherapeutics Mediated by TXNDC17 in Neuroblastomas. Cancer Rep (Hoboken) 2024; 7:e70033. [PMID: 39411839 PMCID: PMC11480999 DOI: 10.1002/cnr2.70033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 06/17/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND The prognosis of high-risk neuroblastomas (NB) that are resistant to first-line induction chemotherapy is relatively poor. This study explored the mechanism of resistance to first-line chemotherapeutics mediated by TXNDC17 and its potential solutions in NB. METHODS The genetic and clinical data of patients with NB were obtained from the Therapeutically Applicable Research to Generate Effective Treatments dataset. TXNDC17 and BECN1 expressions in NB cells were up- and downregulated by transfection with plasmids and shRNA, respectively. Autophagy-related proteins were detected by western blot. Cell viability was determined using cell proliferation and toxicity experiments. Apoptotic cells were detected using flow cytometry. RESULTS Overall, 1076 pediatric and adolescent patients with NB were enrolled in this study. The 10-year overall survival (OS) rates and event-free survival (EFS) rates for the patients with a mutation of BECN1 were 37.4 ± 9.1% and 34.5 ± 8.8%, respectively. For patients with a mutation of TXNDC17, the 10-year OS and EFS were 41.4 ± 5.9% and 24.3 ± 5.1%, respectively, which were significantly lower than those in the unaltered group. The overexpression of BECN1 and TXNDC17 reduced NB sensitivity to cisplatin (DDP), etoposide (VP16), and cyclophosphamide (CTX). Autophagy mediated by BECN1 was regulated by TXNDC17, and this process was involved in the resistance to DDP, VP16, and CTX in NB. Suberoylanilide hydroxamic acid (SAHA) can enhance the sensitivity and apoptosis of NB cells to chemotherapeutics by inhibiting TXNDC17, ultimately decreasing autophagy-mediated chemoresistance. CONCLUSIONS Acquired resistance to first-line chemotherapeutics was associated with autophagy mediated by BECN1 and regulated by TXNDC17, which can be reversed by SAHA.
Collapse
Affiliation(s)
- Chenggong Zeng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouPR China
- Collaborative Innovation Center of Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
- Department of Pediatric OncologySun Yat‐Sen University Cancer CenterGuangzhouPR China
| | - Zhuoran Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouPR China
- Collaborative Innovation Center of Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
- Department of Pediatric OncologySun Yat‐Sen University Cancer CenterGuangzhouPR China
| | - Zhiqing Wei
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouPR China
- Collaborative Innovation Center of Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
- Department of Pediatric OncologySun Yat‐Sen University Cancer CenterGuangzhouPR China
| | - Tingting Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouPR China
- Collaborative Innovation Center of Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
- Department of Pediatric OncologySun Yat‐Sen University Cancer CenterGuangzhouPR China
| | - Juan Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouPR China
- Collaborative Innovation Center of Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
- Department of Pediatric OncologySun Yat‐Sen University Cancer CenterGuangzhouPR China
| | - Junting Huang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouPR China
- Collaborative Innovation Center of Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
- Department of Pediatric OncologySun Yat‐Sen University Cancer CenterGuangzhouPR China
| | - Feifei Sun
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouPR China
- Collaborative Innovation Center of Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
- Department of Pediatric OncologySun Yat‐Sen University Cancer CenterGuangzhouPR China
| | - Jia Zhu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouPR China
- Collaborative Innovation Center of Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
- Department of Pediatric OncologySun Yat‐Sen University Cancer CenterGuangzhouPR China
| | - Suying Lu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouPR China
- Collaborative Innovation Center of Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
- Department of Pediatric OncologySun Yat‐Sen University Cancer CenterGuangzhouPR China
| | - Zijun Zhen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouPR China
- Collaborative Innovation Center of Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
- Department of Pediatric OncologySun Yat‐Sen University Cancer CenterGuangzhouPR China
| |
Collapse
|
35
|
Ibarra-García-Padilla R, Nambiar A, Hamre TA, Singleton EW, Uribe RA. Expansion of a neural crest gene signature following ectopic MYCN expression in sympathoadrenal lineage cells in vivo. PLoS One 2024; 19:e0310727. [PMID: 39292691 PMCID: PMC11410271 DOI: 10.1371/journal.pone.0310727] [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: 04/02/2024] [Accepted: 08/26/2024] [Indexed: 09/20/2024] Open
Abstract
Neural crest cells (NCC) are multipotent migratory stem cells that originate from the neural tube during early vertebrate embryogenesis. NCCs give rise to a variety of cell types within the developing organism, including neurons and glia of the sympathetic nervous system. It has been suggested that failure in correct NCC differentiation leads to several diseases, including neuroblastoma (NB). During normal NCC development, MYCN is transiently expressed to promote NCC migration, and its downregulation precedes neuronal differentiation. Overexpression of MYCN has been linked to high-risk and aggressive NB progression. For this reason, understanding the effect overexpression of this oncogene has on the development of NCC-derived sympathoadrenal progenitors (SAP), which later give rise to sympathetic nerves, will help elucidate the developmental mechanisms that may prime the onset of NB. Here, we found that overexpressing human EGFP-MYCN within SAP lineage cells in zebrafish led to the transient formation of an abnormal SAP population, which displayed expanded and elevated expression of NCC markers while paradoxically also co-expressing SAP and neuronal differentiation markers. The aberrant NCC signature was corroborated with in vivo time-lapse confocal imaging in zebrafish larvae, which revealed transient expansion of sox10 reporter expression in MYCN overexpressing SAPs during the early stages of SAP development. In these aberrant MYCN overexpressing SAP cells, we also found evidence of dampened BMP signaling activity, indicating that BMP signaling disruption occurs following elevated MYCN expression. Furthermore, we discovered that pharmacological inhibition of BMP signaling was sufficient to create an aberrant NCC gene signature in SAP cells, phenocopying MYCN overexpression. Together, our results suggest that MYCN overexpression in SAPs disrupts their differentiation by eliciting abnormal NCC gene expression programs, and dampening BMP signaling response, having developmental implications for the priming of NB in vivo.
Collapse
Affiliation(s)
- Rodrigo Ibarra-García-Padilla
- Department of Biosciences, Rice University, Houston, Texas, United States of America
- Biochemistry and Cell Biology Graduate Program, Rice University, Houston, Texas, United States of America
| | - Annika Nambiar
- Department of Biosciences, Rice University, Houston, Texas, United States of America
| | - Thomas A Hamre
- Department of Biosciences, Rice University, Houston, Texas, United States of America
| | - Eileen W Singleton
- Department of Biosciences, Rice University, Houston, Texas, United States of America
| | - Rosa A Uribe
- Department of Biosciences, Rice University, Houston, Texas, United States of America
| |
Collapse
|
36
|
Sun L, Shao W, Lin Z, Lin J, Zhao F, Yu J. Single-cell RNA sequencing explored potential therapeutic targets by revealing the tumor microenvironment of neuroblastoma and its expression in cell death. Discov Oncol 2024; 15:409. [PMID: 39235657 PMCID: PMC11377405 DOI: 10.1007/s12672-024-01286-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 08/28/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND Neuroblastoma (NB) is the most common extracranial solid tumor in childhood and is closely related to the early development and differentiation of neuroendocrine (NE) cells. The disease is mainly represented by high-risk NB, which has the characteristics of high mortality and difficult treatment. The survival rate of high-risk NB patients is not ideal. In this article, we not only conducted a comprehensive study of NB through single-cell RNA sequencing (scRNA-seq) but also further analyzed cuproptosis, a new cell death pathway, in order to find clinical treatment targets from a new perspective. MATERIALS AND METHODS The Seurat software was employed to process the scRNA-seq data. This was followed by the utilization of GO enrichment analysis and GSEA to unveil pertinent enriched pathways. The inferCNV software package was harnessed to investigate chromosomal copy number variations. pseudotime analyses involved the use of Monocle 2, CytoTRACE, and Slingshot software. CellChat was employed to analyze the intercellular communication network for NB. Furthermore, PySCENIC was deployed to review the profile of transcription factors. RESULT Using scRNA-seq, we studied cells from patients with NB. NE cells exhibited superior specificity in contrast to other cell types. Among NE cells, C1 PCLAF + NE cells showed a close correlation with the genesis and advancement of NB. The key marker genes, cognate receptor pairing, developmental trajectories, metabolic pathways, transcription factors, and enrichment pathways in C1 PCLAF + NE cells, as well as the expression of cuproptosis in C1 PCLAF + NE cells, provided new ideas for exploring new therapeutic targets for NB. CONCLUSION The results revealed the specificity of malignant NE cells in NB, especially the key subset of C1 PCLAF + NE cells, which enhanced our understanding of the key role of the tumor microenvironment in the complexity of cancer progression. Of course, cell death played an important role in the progression of NB, which also promoted our research on new targets. The scrutiny of these findings proved advantageous in uncovering innovative therapeutic targets, thereby bolstering clinical interventions.
Collapse
Affiliation(s)
- Lei Sun
- Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, China
| | - Wenwen Shao
- Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, China
| | - Zhiheng Lin
- Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, China
| | - Jingheng Lin
- Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, China
| | - Fu Zhao
- Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, China
| | - Juan Yu
- Pediatric Tuina Health Care Clinic, Shandong University of Traditional Chinese Medicine Affiliated Hospital, No. 16369, Jingshi Road, Jinan, 250014, Shandong, China.
| |
Collapse
|
37
|
Ognibene M, Parodi S, Amoroso L, Zara F, Pezzolo A. Overexpression of H2AFX gene in neuroblastoma is associated with worse prognosis. Pediatr Blood Cancer 2024; 71:e31146. [PMID: 38938078 DOI: 10.1002/pbc.31146] [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: 02/16/2024] [Revised: 05/22/2024] [Accepted: 05/31/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND Neuroblastoma (NB) is the most common solid tumour in childhood, and rises in the sympathetic nervous system. Here, we addressed the in silico analysis of the association between the expression of H2AFX gene involved in DNA damage response, and the survival of a cohort of 786 NB patients. METHODS In silico gene expression was retrieved from the publicly available dataset summarised by Cangelosi et al., including 13,696 gene expression profiles of 786 NB tumours at onset of disease. The prognostic value of H2AFX (H2A histone family member X) gene expression for event-free survival (EFS) and overall survival (OS) was evaluated by Kaplan-Meier and Cox regression analysis. The main results were validated on another openly accessible in silico database (NRC-283) containing 13,489 gene expressions in 283 NB patients. The expression of H2AFX protein was then tested by immunofluorescence on 48 primary NB samples of different tumour stages. H2AFX activity as an oncogene has been further validated in vitro by silencing the molecule in two NB cell lines, characterised by MYCN amplified or not, and performing cell growth and migration assays. RESULTS A strong inverse association between H2AFX expression and patients' survival was observed and confirmed by immunofluorescence results on primary NB tissue sections. Cox regression analysis also disclosed H2AFX as an independent predictor of EFS and OS. The gene-silencing experiments strongly suggested an oncogenic role for H2AFX on NB cells, regardless of MYCN amplification. CONCLUSIONS H2AFX is a prognostic marker for unfavourable NB and could be considered a target for therapeutic interventions.
Collapse
Affiliation(s)
- Marzia Ognibene
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Stefano Parodi
- Epidemiologia e Biostatistica, Direzione Scientifica, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Loredana Amoroso
- UOC Oncologia Pediatrica, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Federico Zara
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | | |
Collapse
|
38
|
Pascual-Pasto G, McIntyre B, Hines MG, Giudice AM, Garcia-Gerique L, Hoffmann J, Mishra P, Matlaga S, Lombardi S, Shraim R, Schürch PM, Yarmarkovich M, Hofmann TJ, Alikarami F, Martinez D, Tsang M, Gil-de-Gómez L, Spear TT, Bernt KM, Wolpaw AJ, Dimitrov DS, Li W, Bosse KR. CAR T-cell-mediated delivery of bispecific innate immune cell engagers for neuroblastoma. Nat Commun 2024; 15:7141. [PMID: 39164224 PMCID: PMC11336212 DOI: 10.1038/s41467-024-51337-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 08/06/2024] [Indexed: 08/22/2024] Open
Abstract
Novel chimeric antigen receptor (CAR) T-cell approaches are needed to improve therapeutic efficacy in solid tumors. High-risk neuroblastoma is an aggressive pediatric solid tumor that expresses cell-surface GPC2 and GD2 with a tumor microenvironment infiltrated by CD16a-expressing innate immune cells. Here we engineer T-cells to express a GPC2-directed CAR and simultaneously secrete a bispecific innate immune cell engager (BiCE) targeting both GD2 and CD16a. In vitro, GPC2.CAR-GD2.BiCE T-cells induce GPC2-dependent cytotoxicity and secrete GD2.BiCE that promotes GD2-dependent activation of antitumor innate immunity. In vivo, GPC2.CAR-GD2.BiCE T-cells locally deliver GD2.BiCE and increase intratumor retention of NK-cells. In mice bearing neuroblastoma patient-derived xenografts and reconstituted with human CD16a-expressing immune cells, GD2.BiCEs enhance GPC2.CAR antitumor efficacy. A CAR.BiCE strategy should be considered for tumor histologies where antigen escape limits CAR efficacy, especially for solid tumors like neuroblastoma that are infiltrated by innate immune cells.
Collapse
Affiliation(s)
- Guillem Pascual-Pasto
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Brendan McIntyre
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Margaret G Hines
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Anna M Giudice
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Laura Garcia-Gerique
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, 19104, USA
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Jennifer Hoffmann
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Pamela Mishra
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Stephanie Matlaga
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Simona Lombardi
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Rawan Shraim
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Patrick M Schürch
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Mark Yarmarkovich
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- New York University, Perlmutter Cancer Center, Grossman School of Medicine, New York, NY, 10016, USA
| | - Ted J Hofmann
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Fatemeh Alikarami
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Daniel Martinez
- Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Matthew Tsang
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Luis Gil-de-Gómez
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Molecular Biology, University of Cantabria School of Medicine, Santander, Cantabria, 39011, Spain
| | - Timothy T Spear
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Kathrin M Bernt
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Adam J Wolpaw
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Dimiter S Dimitrov
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Wei Li
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Kristopher R Bosse
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
39
|
López-Carrasco A, Vieco-Martí I, Granados-Aparici S, Acevedo-León D, Estañ-Capell N, Portugal R, Huerta-Aragonés J, Cañete A, Navarro S, Noguera R. Vitronectin Levels in the Plasma of Neuroblastoma Patients and Culture Media of 3D Models: A Prognostic Circulating Biomarker? Int J Mol Sci 2024; 25:8733. [PMID: 39201421 PMCID: PMC11354570 DOI: 10.3390/ijms25168733] [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: 07/09/2024] [Revised: 08/02/2024] [Accepted: 08/09/2024] [Indexed: 09/02/2024] Open
Abstract
Vitronectin is a glycoprotein present in plasma and the extracellular matrix that is implicated in cell migration. The high amount of vitronectin found in neuroblastoma biopsies has been associated with poor prognosis. Moreover, increased vitronectin levels have been described in the plasma of patients with different cancers. Our aim was to assess vitronectin as a potential circulating biomarker of neuroblastoma prognosis. Vitronectin concentration was quantified using ELISA in culture media of four neuroblastoma cell lines grown in a monolayer and in 3D models, and in the plasma of 114 neuroblastoma patients. Three of the neuroblastoma cell lines secreted vitronectin to culture media when cultured in a monolayer and 3D models. Vitronectin release was higher by neuroblastoma cells cultured in 3D models than in the monolayer and was still elevated when cells were grown in 3D scaffolds with cross-linked vitronectin. Vitronectin secretion occurred independently of cell numbers in cultures. Its concentration in the plasma of neuroblastoma patients ranged between 52.4 and 870 µg/mL (median, 218 µg/mL). A ROC curve was used to establish a cutoff of 361 µg/mL, above which patients over 18 months old had worse prognosis (p = 0.0018). Vitronectin could be considered a new plasma prognostic biomarker in neuroblastoma and warrants confirmation in collaborative studies. Drugs inhibiting vitronectin interactions with cells and/or the extracellular matrix could represent a significant improvement in survival for neuroblastoma patients.
Collapse
Affiliation(s)
- Amparo López-Carrasco
- Incliva Biomedical Health Research Institute, 46010 Valencia, Spain; (A.L.-C.); (S.G.-A.); (S.N.)
- CIBER of Cancer (CIBERONC), 28029 Madrid, Spain
| | - Isaac Vieco-Martí
- Incliva Biomedical Health Research Institute, 46010 Valencia, Spain; (A.L.-C.); (S.G.-A.); (S.N.)
- CIBER of Cancer (CIBERONC), 28029 Madrid, Spain
| | - Sofía Granados-Aparici
- Incliva Biomedical Health Research Institute, 46010 Valencia, Spain; (A.L.-C.); (S.G.-A.); (S.N.)
- CIBER of Cancer (CIBERONC), 28029 Madrid, Spain
| | | | | | | | | | - Adela Cañete
- Politechnic and University Hospital La Fe, 46026 Valencia, Spain
| | - Samuel Navarro
- Incliva Biomedical Health Research Institute, 46010 Valencia, Spain; (A.L.-C.); (S.G.-A.); (S.N.)
- CIBER of Cancer (CIBERONC), 28029 Madrid, Spain
- Pathology Department, Medical School, University of Valencia, 46010 Valencia, Spain
| | - Rosa Noguera
- Incliva Biomedical Health Research Institute, 46010 Valencia, Spain; (A.L.-C.); (S.G.-A.); (S.N.)
- CIBER of Cancer (CIBERONC), 28029 Madrid, Spain
- Pathology Department, Medical School, University of Valencia, 46010 Valencia, Spain
| |
Collapse
|
40
|
Gökçek-Saraç Ç, Çetin E, Ateş K, Özen Ş, Karakurt S. Different duration of exposure to a pulsed magnetic field can cause changes in mRNA expression of apoptotic genes in oleic acid-treated neuroblastoma cells. Int J Radiat Biol 2024; 100:1471-1480. [PMID: 39088733 DOI: 10.1080/09553002.2024.2386968] [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: 01/10/2024] [Revised: 06/20/2024] [Accepted: 07/22/2024] [Indexed: 08/03/2024]
Abstract
PURPOSE Neuroblastoma, a prevalent childhood tumor, poses significant challenges in therapeutic interventions, especially for high-risk cases. This study aims to fill a crucial gap in our understanding of neuroblastoma treatment by investigating the potential molecular impacts of short- and long-term pulsed magnetic field exposure on the neuronal apoptosis mechanism in an in vitro model of neuroblastoma treated with oleic acid (OA). MATERIALS AND METHODS Cells were cultured and divided into six following experimental groups: (I) Nontreated group (NT); (II) OA-treated group (OA); (III) Group treated with OA after being exposed to the pulsed magnetic field for 15-min (15 min PEMF + OA); (IV) Group treated with OA after being exposed to the pulsed magnetic field for 12 h (12 h PEMF + OA); (V) Group exposed to the pulsed magnetic field for 15 min (15 min PEMF); and (VI) Group exposed to the pulsed magnetic field for 12 h (12 h PEMF). Cell viability, rates of apoptosis, and mRNA levels of key apoptotic genes (TP53, Bcl2, Bax, and Caspase-3) were assessed. RESULTS Significant reductions in cell viability were observed, particularly in the group treated with OA following long-term pulsed magnetic field exposure. Flow cytometry revealed elevated apoptosis rates, notably in the early stages of apoptosis. qRT-PCR analysis demonstrated increased expression of cleaved Caspase-3, Bax/Bcl2 ratio, and TP53 in cells treated with OA following long-term pulsed magnetic field exposure, signifying enhanced apoptotic pathways. CONCLUSIONS The findings indicate that long-term pulsed magnetic field exposure and OA treatment exhibit potential synergistic effects leading to the induction of apoptosis in SH-SY5Y cells. We have concluded that stimulations of pulsed magnetic field have the potential to serve as an adjuvant therapy for oleic acid-based treatment of neuroblastoma.
Collapse
Affiliation(s)
- Çiğdem Gökçek-Saraç
- Department of Biomedical Engineering, Faculty of Engineering, Akdeniz University, Antalya, Turkey
| | - Ebru Çetin
- Department of Biomedical Engineering, Faculty of Engineering, Akdeniz University, Antalya, Turkey
| | - Kayhan Ateş
- Department of Electrical and Electronics Engineering, Faculty of Engineering, Akdeniz University, Antalya, Turkey
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy
| | - Şükrü Özen
- Department of Electrical and Electronics Engineering, Faculty of Engineering, Akdeniz University, Antalya, Turkey
| | - Serdar Karakurt
- Department of Biochemistry, Faculty of Science, Selçuk University, Konya, Turkey
| |
Collapse
|
41
|
Sundaramoorthy S, Colombo DF, Sanalkumar R, Broye L, Balmas Bourloud K, Boulay G, Cironi L, Stamenkovic I, Renella R, Kuttler F, Turcatti G, Rivera MN, Mühlethaler-Mottet A, Bardet AF, Riggi N. Preclinical spheroid models identify BMX as a therapeutic target for metastatic MYCN nonamplified neuroblastoma. JCI Insight 2024; 9:e169647. [PMID: 39133652 PMCID: PMC11383371 DOI: 10.1172/jci.insight.169647] [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: 02/13/2023] [Accepted: 06/10/2024] [Indexed: 09/11/2024] Open
Abstract
The development of targeted therapies offers new hope for patients affected by incurable cancer. However, multiple challenges persist, notably in controlling tumor cell plasticity in patients with refractory and metastatic illness. Neuroblastoma (NB) is an aggressive pediatric malignancy originating from defective differentiation of neural crest-derived progenitors with oncogenic activity due to genetic and epigenetic alterations and remains a clinical challenge for high-risk patients. To identify critical genes driving NB aggressiveness, we performed combined chromatin and transcriptome analyses on matched patient-derived xenografts (PDXs), spheroids, and differentiated adherent cultures derived from metastatic MYCN nonamplified tumors. Bone marrow kinase on chromosome X (BMX) was identified among the most differentially regulated genes in PDXs and spheroids versus adherent models. BMX expression correlated with high tumor stage and poor patient survival and was crucial to the maintenance of the self-renewal and tumorigenic potential of NB spheroids. Moreover, BMX expression positively correlated with the mesenchymal NB cell phenotype, previously associated with increased chemoresistance. Finally, BMX inhibitors readily reversed this cellular state, increased the sensitivity of NB spheroids toward chemotherapy, and partially reduced tumor growth in a preclinical NB model. Altogether, our study identifies BMX as a promising innovative therapeutic target for patients with high-risk MYCN nonamplified NB.
Collapse
Affiliation(s)
| | | | - Rajendran Sanalkumar
- Experimental Pathology Service, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Liliane Broye
- Experimental Pathology Service, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Katia Balmas Bourloud
- Department Woman-Mother-Child, Division of Pediatrics, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Gaylor Boulay
- Department of Pathology and Cancer Center, Massachusetts General Hospital and Harvard Medical School
| | - Luisa Cironi
- Experimental Pathology Service, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Ivan Stamenkovic
- Experimental Pathology Service, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Raffaele Renella
- Department Woman-Mother-Child, Division of Pediatrics, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Fabien Kuttler
- Biomolecular Screening Facility, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Gerardo Turcatti
- Biomolecular Screening Facility, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Miguel N Rivera
- Department of Pathology and Cancer Center, Massachusetts General Hospital and Harvard Medical School
| | - Annick Mühlethaler-Mottet
- Department Woman-Mother-Child, Division of Pediatrics, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Anaïs Flore Bardet
- Biotechnology and Cell Signaling (BSC), CNRS UMR7242, University of Strasbourg, Illkirch, France
- Institute of Genetics and Molecular and Cellular Biology (IGBMC), CNRS UMR7104, University of Strasbourg, INSERM U1258, Illkirch, France
| | - Nicolò Riggi
- Experimental Pathology Service, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
42
|
Tu J, Toh Y, Aldana AM, Wen JJ, Wu L, Jacob J, Li L, Pan S, Carmon KS, Liu QJ. Antitumor Activity of a Pyrrolobenzodiazepine Antibody-Drug Conjugate Targeting LGR5 in Preclinical Models of Neuroblastoma. Pharmaceutics 2024; 16:943. [PMID: 39065640 PMCID: PMC11279891 DOI: 10.3390/pharmaceutics16070943] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/05/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Neuroblastoma (NB) is a cancer of the peripheral nervous system found in children under 15 years of age. It is the most frequently diagnosed cancer during infancy, accounting for ~12% of all cancer-related deaths in children. Leucine-rich repeat-containing G-protein-coupled receptor 5 (LGR5) is a membrane receptor that is associated with the primary tumor formation and metastasis of cancers in the gastrointestinal system. Remarkably, high levels of LGR5 are found in NB tumor cells, and high LGR5 expression is strongly correlated with poor survival. Antibody-drug conjugates (ADCs) are monoclonal antibodies that are covalently linked to cell-killing cytotoxins to deliver the payloads into cancer cells. We generated an ADC with an anti-LGR5 antibody and pyrrolobenzodiazepine (PBD) dimer-based payload SG3199 using a chemoenzymatic conjugation method. The resulting anti-LGR5 ADC was able to inhibit the growth of NB cells expressing LGR5 with high potency and specificity. Importantly, the ADC was able to completely inhibit the growth of NB xenograft tumors in vivo at a clinically relevant dose for the PBD class of ADCs. The findings support the potential of targeting LGR5 using the PBD class of payload for the treatment of high-risk NBs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Qingyun J. Liu
- The Brown Foundation Institute of Molecular Medicine, Center for Translational Cancer Research, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
43
|
Fredlund E, Andersson S, Hilgert E, Monferrer E, Álvarez-Hernán G, Karakaya S, Loontiens S, Bek JW, Gregor T, Lecomte E, Magnusson E, Miltenyte E, Cabirol M, Kyknas M, Engström N, Henriksson MA, Hammarlund E, Rosenblum JS, Noguera R, Speleman F, van Nes J, Mohlin S. MOXD1 is a lineage-specific gene and a tumor suppressor in neuroblastoma. SCIENCE ADVANCES 2024; 10:eado1583. [PMID: 38905335 PMCID: PMC11192077 DOI: 10.1126/sciadv.ado1583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 05/14/2024] [Indexed: 06/23/2024]
Abstract
Neuroblastoma is a childhood developmental cancer; however, its embryonic origins remain poorly understood. Moreover, in-depth studies of early tumor-driving events are limited because of the lack of appropriate models. Herein, we analyzed RNA sequencing data obtained from human neuroblastoma samples and found that loss of expression of trunk neural crest-enriched gene MOXD1 associates with advanced disease and worse outcome. Further, by using single-cell RNA sequencing data of human neuroblastoma cells and fetal adrenal glands and creating in vivo models of zebrafish, chick, and mouse, we show that MOXD1 is a determinate of tumor development. In addition, we found that MOXD1 expression is highly conserved and restricted to mesenchymal neuroblastoma cells and Schwann cell precursors during healthy development. Our findings identify MOXD1 as a lineage-restricted tumor-suppressor gene in neuroblastoma, potentiating further stratification of these tumors and development of novel therapeutic interventions.
Collapse
Affiliation(s)
- Elina Fredlund
- Division of Pediatrics, Department of Clinical Sciences, Lund University, Lund, Sweden
- Lund University Cancer Center, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Stina Andersson
- Division of Pediatrics, Department of Clinical Sciences, Lund University, Lund, Sweden
- Lund University Cancer Center, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Elien Hilgert
- Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - Ezequiel Monferrer
- Department of Pathology, Medical School, University of Valencia-INCLIVA Biomedical Health Research Institute, Valencia, Spain
- Low Prevalence Tumors, Centro de Investigación Biomédica En Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Guadalupe Álvarez-Hernán
- Division of Pediatrics, Department of Clinical Sciences, Lund University, Lund, Sweden
- Lund University Cancer Center, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Sinan Karakaya
- Division of Pediatrics, Department of Clinical Sciences, Lund University, Lund, Sweden
- Lund University Cancer Center, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Siebe Loontiens
- Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - Jan Willem Bek
- Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - Tomas Gregor
- Division of Pediatrics, Department of Clinical Sciences, Lund University, Lund, Sweden
- Lund University Cancer Center, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Estelle Lecomte
- Division of Pediatrics, Department of Clinical Sciences, Lund University, Lund, Sweden
- Lund University Cancer Center, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Emma Magnusson
- Division of Pediatrics, Department of Clinical Sciences, Lund University, Lund, Sweden
- Lund University Cancer Center, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Enrika Miltenyte
- Division of Pediatrics, Department of Clinical Sciences, Lund University, Lund, Sweden
- Lund University Cancer Center, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Marie Cabirol
- Division of Pediatrics, Department of Clinical Sciences, Lund University, Lund, Sweden
- Lund University Cancer Center, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Michail Kyknas
- Division of Pediatrics, Department of Clinical Sciences, Lund University, Lund, Sweden
- Lund University Cancer Center, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Niklas Engström
- Division of Pediatrics, Department of Clinical Sciences, Lund University, Lund, Sweden
- Lund University Cancer Center, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Marie Arsenian Henriksson
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum B7, Karolinska Institute, Stockholm, Sweden
- Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Emma Hammarlund
- Lund University Cancer Center, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
- Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Jared S. Rosenblum
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rosa Noguera
- Department of Pathology, Medical School, University of Valencia-INCLIVA Biomedical Health Research Institute, Valencia, Spain
- Low Prevalence Tumors, Centro de Investigación Biomédica En Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Frank Speleman
- Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - Johan van Nes
- Department of Oncogenomics, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Sofie Mohlin
- Division of Pediatrics, Department of Clinical Sciences, Lund University, Lund, Sweden
- Lund University Cancer Center, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
| |
Collapse
|
44
|
Wang C, Tan J, Jin Y, Li Z, Yang J, Jia Y, Xia Y, Gong B, Dong Q, Zhao Q. A mitochondria-related genes associated neuroblastoma signature - based on bulk and single-cell transcriptome sequencing data analysis, and experimental validation. Front Immunol 2024; 15:1415736. [PMID: 38962012 PMCID: PMC11220120 DOI: 10.3389/fimmu.2024.1415736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 06/03/2024] [Indexed: 07/05/2024] Open
Abstract
Background Neuroblastoma (NB), characterized by its marked heterogeneity, is the most common extracranial solid tumor in children. The status and functionality of mitochondria are crucial in regulating NB cell behavior. While the significance of mitochondria-related genes (MRGs) in NB is still missing in key knowledge. Materials and methods This study leverages consensus clustering and machine learning algorithms to construct and validate an MRGs-related signature in NB. Single-cell data analysis and experimental validation were employed to characterize the pivotal role of FEN1 within NB cells. Results MRGs facilitated the classification of NB patients into 2 distinct clusters with considerable differences. The constructed MRGs-related signature and its quantitative indicators, mtScore and mtRisk, effectively characterize the MRGs-related patient clusters. Notably, the MRGs-related signature outperformed MYCN in predicting NB patient prognosis and was adept at representing the tumor microenvironment (TME), tumor cell stemness, and sensitivity to the chemotherapeutic agents Cisplatin, Topotecan, and Irinotecan. FEN1, identified as the most contributory gene within the MRGs-related signature, was found to play a crucial role in the communication between NB cells and the TME, and in the developmental trajectory of NB cells. Experimental validations confirmed FEN1's significant influence on NB cell proliferation, apoptosis, cell cycle, and invasiveness. Conclusion The MRGs-related signature developed in this study offers a novel predictive tool for assessing NB patient prognosis, immune infiltration, stemness, and chemotherapeutic sensitivity. Our findings unveil the critical function of FEN1 in NB, suggesting its potential as a therapeutic target.
Collapse
Affiliation(s)
- Chaoyu Wang
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Jiaxiong Tan
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yan Jin
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Zongyang Li
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Jiaxing Yang
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yubin Jia
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yuren Xia
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Baocheng Gong
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Qiuping Dong
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Qiang Zhao
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| |
Collapse
|
45
|
Zhao JD, Lu XY, Chen TP, Duan XL, Zuo W, Sai K, Zhu LR, Gao Q. Development and validation of a novel nomogram for predicting overall survival patients with neuroblastoma. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:108321. [PMID: 38598875 DOI: 10.1016/j.ejso.2024.108321] [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: 12/13/2023] [Revised: 03/26/2024] [Accepted: 04/03/2024] [Indexed: 04/12/2024]
Abstract
PURPOSE The aim of this study was to develop a nomogram specially for predicting overall survival (OS) for Chinese patients with neuroblastoma (NB). METHODS Patients with pathologically confirmed NB who were newly diagnosed and received treatments at our hospital from October 2013 to October 2021 were retrospectively reviewed. The nomogram for OS were built based on Cox regression analysis. The validation of the prognostic model was evaluated by concordance index (C-index), calibration curves, and decision curve analyses (DCAs). RESULTS A total of 254 patients with NB were included in this study. They were randomly divided into a training cohort (n = 178) and a validation cohort (n = 76) at a ratio of 7:3. Multivariate analyses revealed that prognostic variables significantly related to the OS were age at diagnosis, bone metastasis, hepatic metastasis, INSS stage, MYCN status and DNA ploidy. The nomogram was constructed based on above 6 factors. The C-index values of the nomogram for predicting 3-year and 5-year OS were 0.926 and 0.964, respectively. The calibration curves of the nomogram showed good consistency between nomogram prediction and actual survival. The DCAs showed great clinical usefulness of the nomograms. Furthermore, patients with low-risk identified by our nomogram had much higher OS than those with high-risk (p < 0.001). CONCLUSION The nomogram we constructed exhibited good predictive performance and could be used to assist clinicians in their decision-making process.
Collapse
Affiliation(s)
- Jin-du Zhao
- Department of Oncology Surgery, Anhui Medical University Children's Medical Center, Anhui Provincial Children's Hospital, Hefei, 230051, Anhui, China
| | - Xian-Ying Lu
- Department of Oncology Surgery, Anhui Medical University Children's Medical Center, Anhui Provincial Children's Hospital, Hefei, 230051, Anhui, China
| | - Tian-Ping Chen
- Department of Hematology and Oncology, Anhui Medical University Children's Medical Center, Anhui Provincial Children's Hospital, Hefei, 230051, Anhui, China
| | - Xian-Lun Duan
- Department of Thoracic Surgery, Anhui Medical University Children's Medical Center, Anhui Provincial Children's Hospital, Hefei, 230051, Anhui, China
| | - Wei Zuo
- Department of Neonatal Surgery, Anhui Medical University Children's Medical Center, Anhui Provincial Children's Hospital, Hefei, 230051, Anhui, China
| | - Kai Sai
- Department of Oncology Surgery, Anhui Medical University Children's Medical Center, Anhui Provincial Children's Hospital, Hefei, 230051, Anhui, China
| | - Li-Ran Zhu
- Anhui Institute of Pediatric Research, Anhui Medical University Children's Medical Center, Anhui Provincial Children's Hospital, Hefei, 230051, Anhui, China
| | - Qun Gao
- Department of Oncology Surgery, Anhui Medical University Children's Medical Center, Anhui Provincial Children's Hospital, Hefei, 230051, Anhui, China.
| |
Collapse
|
46
|
Chan C, Stip M, Nederend M, Jansen M, Passchier E, van den Ham F, Wienke J, van Tetering G, Leusen J. Enhancing IgA-mediated neutrophil cytotoxicity against neuroblastoma by CD47 blockade. J Immunother Cancer 2024; 12:e008478. [PMID: 38782540 PMCID: PMC11116899 DOI: 10.1136/jitc-2023-008478] [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: 05/06/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Approximately half of the neuroblastoma patients develop high-risk neuroblastoma. Current treatment involves a multimodal strategy, including immunotherapy with dinutuximab (IgG ch14.18) targeting GD2. Despite achieving promising results, the recurrence rate remains high and poor survival persists. The therapeutic efficacy of dinutuximab is compromised by suboptimal activation of neutrophils and severe neuropathic pain, partially induced by complement activation. METHODS To enhance neutrophil cytotoxicity, IgG ch14.18 was converted to the IgA isotype, resulting in potent neutrophil-mediated antibody-dependent cell-mediated cytotoxicity (ADCC), without complement activation. However, myeloid checkpoint molecules hamper neutrophil cytotoxicity, for example through CD47 that is overexpressed on neuroblastomas and orchestrates an immunosuppressive environment upon ligation to signal regulatory protein alpha (SIRPα) expressed on neutrophils. In this study, we combined IgA therapy with CD47 blockade. RESULTS In vitro killing assays showed enhanced IgA-mediated ADCC by neutrophils targeting neuroblastoma cell lines and organoids in comparison to IgG. Notably, when combined with CD47 blockade, both IgG and IgA therapy were enhanced, though the combination with IgA resulted in the greatest improvement of ADCC. Furthermore, in a neuroblastoma xenograft model, we systemically blocked CD47 with a SIRPα fusion protein containing an ablated IgG1 Fc, and compared IgA therapy to IgG therapy. Only IgA therapy combined with CD47 blockade increased neutrophil influx to the tumor microenvironment. Moreover, the IgA combination strategy hampered tumor outgrowth most effectively and prolonged tumor-specific survival. CONCLUSION These promising results highlight the potential to enhance immunotherapy efficacy against high-risk neuroblastoma through improved neutrophil cytotoxicity by combining IgA therapy with CD47 blockade.
Collapse
Affiliation(s)
- Chilam Chan
- Center for Translational Immunology, UMC Utrecht, Utrecht, The Netherlands
| | - Marjolein Stip
- Center for Translational Immunology, UMC Utrecht, Utrecht, The Netherlands
| | - Maaike Nederend
- Center for Translational Immunology, UMC Utrecht, Utrecht, The Netherlands
| | - Marco Jansen
- Center for Translational Immunology, UMC Utrecht, Utrecht, The Netherlands
| | | | - Femke van den Ham
- Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Judith Wienke
- Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Geert van Tetering
- Center for Translational Immunology, UMC Utrecht, Utrecht, The Netherlands
| | - Jeanette Leusen
- Center for Translational Immunology, UMC Utrecht, Utrecht, The Netherlands
| |
Collapse
|
47
|
Hussein R, Abou-Shanab AM, Badr E. A multi-omics approach for biomarker discovery in neuroblastoma: a network-based framework. NPJ Syst Biol Appl 2024; 10:52. [PMID: 38760476 PMCID: PMC11101461 DOI: 10.1038/s41540-024-00371-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 04/16/2024] [Indexed: 05/19/2024] Open
Abstract
Neuroblastoma (NB) is one of the leading causes of cancer-associated death in children. MYCN amplification is a prominent genetic marker for NB, and its targeting to halt NB progression is difficult to achieve. Therefore, an in-depth understanding of the molecular interactome of NB is needed to improve treatment outcomes. Analysis of NB multi-omics unravels valuable insight into the interplay between MYCN transcriptional and miRNA post-transcriptional modulation. Moreover, it aids in the identification of various miRNAs that participate in NB development and progression. This study proposes an integrated computational framework with three levels of high-throughput NB data (mRNA-seq, miRNA-seq, and methylation array). Similarity Network Fusion (SNF) and ranked SNF methods were utilized to identify essential genes and miRNAs. The specified genes included both miRNA-target genes and transcription factors (TFs). The interactions between TFs and miRNAs and between miRNAs and their target genes were retrieved where a regulatory network was developed. Finally, an interaction network-based analysis was performed to identify candidate biomarkers. The candidate biomarkers were further analyzed for their potential use in prognosis and diagnosis. The candidate biomarkers included three TFs and seven miRNAs. Four biomarkers have been previously studied and tested in NB, while the remaining identified biomarkers have known roles in other types of cancer. Although the specific molecular role is yet to be addressed, most identified biomarkers possess evidence of involvement in NB tumorigenesis. Analyzing cellular interactome to identify potential biomarkers is a promising approach that can contribute to optimizing efficient therapeutic regimens to target NB vulnerabilities.
Collapse
Affiliation(s)
- Rahma Hussein
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Ahmed M Abou-Shanab
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Eman Badr
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12578, Egypt.
- Faculty of Computers and Artificial Intelligence, Cairo University, Giza, 12613, Egypt.
| |
Collapse
|
48
|
Polychronopoulos PA, Bedoya-Reina OC, Johnsen JI. The Neuroblastoma Microenvironment, Heterogeneity and Immunotherapeutic Approaches. Cancers (Basel) 2024; 16:1863. [PMID: 38791942 PMCID: PMC11119056 DOI: 10.3390/cancers16101863] [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: 04/08/2024] [Revised: 05/02/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Neuroblastoma is a peripheral nervous system tumor that almost exclusively occurs in young children. Although intensified treatment modalities have led to increased patient survival, the prognosis for patients with high-risk disease is still around 50%, signifying neuroblastoma as a leading cause of cancer-related deaths in children. Neuroblastoma is an embryonal tumor and is shaped by its origin from cells within the neural crest. Hence, neuroblastoma usually presents with a low mutational burden and is, in the majority of cases, driven by epigenetically deregulated transcription networks. The recent development of Omic techniques has given us detailed knowledge of neuroblastoma evolution, heterogeneity, and plasticity, as well as intra- and intercellular molecular communication networks within the neuroblastoma microenvironment. Here, we discuss the potential of these recent discoveries with emphasis on new treatment modalities, including immunotherapies which hold promise for better future treatment regimens.
Collapse
Affiliation(s)
- Panagiotis Alkinoos Polychronopoulos
- Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, 11883 Stockholm, Sweden; (P.A.P.); (O.C.B.-R.)
| | - Oscar C. Bedoya-Reina
- Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, 11883 Stockholm, Sweden; (P.A.P.); (O.C.B.-R.)
- School of Medical Sciences, Örebro University, 70182 Örebro, Sweden
| | - John Inge Johnsen
- Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, 11883 Stockholm, Sweden; (P.A.P.); (O.C.B.-R.)
| |
Collapse
|
49
|
Gerges A, Canning U. Neuroblastoma and its Target Therapies: A Medicinal Chemistry Review. ChemMedChem 2024; 19:e202300535. [PMID: 38340043 DOI: 10.1002/cmdc.202300535] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/22/2024] [Accepted: 02/07/2024] [Indexed: 02/12/2024]
Abstract
Neuroblastoma (NB) is a childhood malignant tumour belonging to a group of embryonic tumours originating from progenitor cells of the sympathoadrenal lineage. The heterogeneity of NB is reflected in the survival rates of those with low and intermediate risk diseases who have survival rates ranging from 85 to 90 %. However, for those identified with high-risk Stage 4 NB, the treatment options are much more limited. For this group, current treatment consists of immunotherapy (monoclonal antibodies) in combination with anti-cancer drugs and has a 40 to 50 % survival rate. The purpose of this review is to summarise NB research from a medicinal chemistry perspective and to highlight advances in targeted drug therapy in the field. The review examines the medicinal chemistry of a number of drugs tested in research, some of which are currently under clinical trial. It concludes by proposing that future medicinal chemistry research into NB should consider other possible target therapies and adopt a multi-target drug approach rather than a one-drug-one-target approach for improved efficacy and less drug-drug interaction for the treatment of NB Stage 4 (NBS4) patients.
Collapse
Affiliation(s)
- A Gerges
- Bioscience Department, London Metropolitan University, 166-220 Holloway Road, London, N7 8DB, England, United Kingdom
| | - U Canning
- Bioscience Department, London Metropolitan University, 166-220 Holloway Road, London, N7 8DB, England, United Kingdom
| |
Collapse
|
50
|
Wang Z, Zhang Y, Li K. Nuclear miRNAs as transcriptional regulators in processes related to various cancers (Review). Int J Oncol 2024; 64:56. [PMID: 38606502 PMCID: PMC11015916 DOI: 10.3892/ijo.2024.5644] [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: 12/13/2023] [Accepted: 03/29/2024] [Indexed: 04/13/2024] Open
Abstract
MicroRNAs (miRNAs) are noncoding small nucleic acids that contain ~22 nucleotides and are considered to promote the degradation or inhibit the translation of mRNA by targeting its 3'‑untranslated region. However, growing evidence has revealed that nuclear miRNAs, combined with gene promoters or enhancers, are able to directly mediate gene transcription. These miRNAs exert a critical influence on cancer progression by affecting cell growth, migration and invasion. In this review, the direct regulation of gene expression by nuclear miRNAs at the transcriptional level was discussed and summarized, and their mechanisms of action in cancers were highlighted with reference to the various body systems.
Collapse
Affiliation(s)
- Ziqiang Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Yu Zhang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Kun Li
- Department of Nuclear Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
| |
Collapse
|