51
|
Jiang P, Zhang T, Wu B, Li X, Fu M, Xu B. Musashi-2 (MSI2) promotes neuroblastoma tumorigenesis through targeting MYC-mediated glucose-6-phosphate dehydrogenase (G6PD) transcriptional activation. Med Oncol 2023; 40:332. [PMID: 37843625 DOI: 10.1007/s12032-023-02199-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 09/19/2023] [Indexed: 10/17/2023]
Abstract
Neuroblastoma (NB) is the deadliest pediatric solid tumor due to its rapid proliferation. Aberrant expression of MYCN is deemed as the most remarkable feature for the predictive hallmark of NB progression and recurrence. However, the phenomenon that only detection of MYCN in the nearly 20% of NB patients hints that there should be other vital oncogenes in the progression of NB. Here, we firstly show that MSI2 mRNA is augmented by analyzing public GEO datasets in the malignant stage according to International Neuroblastoma Staging System (INSS) stages. Although accumulating evidences uncover the emerging roles of MSI2 in several cancers, the regulatory functions and underlying mechanisms of MSI2 in NB remain under-investigated. Herein, we identified that high-expressed MSI2 and low-expressed n-Myc group account for 43.1% of total NB clinical samples (n = 65). Meanwhile, MSI2 expression is profoundly upregulated along with NB malignancy and negatively associated with the survival outcome of NB patients in the NB tissue microarray (NB: n = 65; Ganglioneuroblastoma: n = 31; Ganglioneuroma: n = 27). In vitro, our results revealed that MSI2 promoted migration, invasion, and proliferation of NB cells via enhancing pentose phosphate pathway. Mechanistically, MSI2 upregulated the key enzyme glucose-6-phosphate dehydrogenase (G6PD) via directly binding to 3'-untranslated regions of c-Myc mRNA to facilitate its stability, resulting in enhancing pentose phosphate pathway. Our findings reveal that MSI2 promotes pentose phosphate pathway via activating c-Myc-G6PD signaling, suggesting that MSI2 exhibits a novel and powerful target for the diagnosis and treatment of NB.
Collapse
Affiliation(s)
- Ping Jiang
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Ting Zhang
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Bin Wu
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Xiaoqing Li
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Mingpeng Fu
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Banglao Xu
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China.
| |
Collapse
|
52
|
Qin X, Chen B. Comprehensive analysis and validation reveal potential MYCN regulatory biomarkers associated with neuroblastoma prognosis. J Biomol Struct Dyn 2023; 41:8902-8917. [PMID: 36300516 DOI: 10.1080/07391102.2022.2138977] [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: 05/30/2022] [Accepted: 10/16/2022] [Indexed: 10/31/2022]
Abstract
Neuroblastoma (NB) is an embryonic malignant tumor that occurs in the sympathetic nervous system. The treatment results of patients in the high-risk group are poor, and relapse and treatment failure can occur even with multiple combination treatments. The proto-oncogene MYCN is a BHLH Transcription Factor used as an independent prognostic factor for NB. The proportion of MYCN amplification in tumor tissues of high-risk patients reaches 40-50%. Hence, exploring new MYCN target genes is a meaningful approach in developing treatment for high-risk NB patients. The microarray datasets were obtained from Gene Expression Omnibus (GEO), and differentially expressed genes (DEGs) were identified. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and miRPathDB were used for enrichment analysis. STRING and Cytoscape were used to construct a protein-protein interaction (PPI) network and for modular analysis. The miRNet and NetworkAnalyst databases were used to predict and construct gene-miRNA and gene-TFs networks. The R2 database was used for expression, correlation, and prognostic analyses. The diagnostic value of the biomarkers was predicted by ROC analysis, and RT-qPCR was used to validate the identified hub genes. Finally, using specific MYCN siRNA and overexpressing plasmids, the correlation between the identified hub genes and MYCN was investigated. Our results showed that FBXO9, HECW2, MIB2, RNF19B, RNF213, TRIM36, and ZBTB16 are novel biomarkers that affect the prognosis of the NB patients. In addition, FBXO9, RNF19B, and TRIM36 were preliminarily confirmed as potential target genes of MYCN. Overall, FBXO9, HECW2, MIB2, RNF19B, RNF213, TRIM36, and ZBTB16 are expected to become novel biomarkers for the treatment of high-risk NB patients.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Xiuni Qin
- Guangzhou Concord Cancer Center, Guangzhou, China
| | - Bo Chen
- Medical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
53
|
刘 雪, 杨 玉, 蔡 慧, 张 耀, 范 方, 李 娴, 李 姗. [Aumolertinib inhibits proliferation, invasion and migration and promotes apoptosis of neuroblastoma cells by downregulating MMP2 and MMP9 expression]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2023; 43:1493-1499. [PMID: 37814863 PMCID: PMC10563090 DOI: 10.12122/j.issn.1673-4254.2023.09.06] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Indexed: 10/11/2023]
Abstract
OBJECTIVE To investigate the effects of aumolertinib, an epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI), on biological behaviors of neuroblastoma SH-SY5Y cells. METHODS CCK-8 assay, colony-forming assay, Transwell assay and flow cytometry were used to assess the effects of 2, 4 and 8 μmol/L aumolertinib on proliferation, survival, migration, invasion and apoptosis of SH-SY5Y cells, and the changes in ultrastructure of the cells were observed using transmission electron microscopy. The protein expressions of Bax, Bcl-2, E-cadherin, vimentin, MMP2, and MMP9 in the treated cells were detected using Western blotting. A nude mouse model bearing subcutaneous SH-SY5Y cell xenograft were treated with aumolertinib (15 mg/kg) or cyclophosphamide (20 mg/kg), and the tumor volume and body mass changes was measured. HE staining was used to observe adverse effects of the treatment on the heart, liver, spleen, lungs and kidneys. RESULTS Aumolertinib significantly inhibited the proliferation and viability of SH-SY5Y cells (P<0.05) with IC50 of 5.004, 3.728 and 3.228 µmol/L at 24, 48 and 72 h, respectively. Aumolertinib treatment induced obvious apoptosis of the cells, which showed characteristic morphological changes of apoptosis under transmission electron microscope. The treatment also inhibited the invasion and migration abilities of SH-SY5Y cells (P<0.01), up-regulated the expression levels of E-cadherin and Bax and lowered the expression levels of Bcl-2, vimentin, MMP2 and MMP9 (P<0.05). In the nude mouse models, treatment with aumolertinib effectively inhibited the growth of neuroblastoma without causing significant toxicity to the vital organs. CONCLUSION Aumolertinib inhibits proliferation, survival, invasion and migration and induces apoptosis in SH-SY5Y cells by downregulating MMP2 and MMP9 expression.
Collapse
Affiliation(s)
- 雪柔 刘
- />蚌埠医学院药学院, 安徽省生化药物工程技术研究中心, 安徽 蚌埠 233030School of Pharmacy, Bengbu Medical College, Anhui Provincial Engineering Technology Research Center of Biochemical Pharmaceuticals, Bengbu 233030, China
| | - 玉梅 杨
- />蚌埠医学院药学院, 安徽省生化药物工程技术研究中心, 安徽 蚌埠 233030School of Pharmacy, Bengbu Medical College, Anhui Provincial Engineering Technology Research Center of Biochemical Pharmaceuticals, Bengbu 233030, China
| | - 慧 蔡
- />蚌埠医学院药学院, 安徽省生化药物工程技术研究中心, 安徽 蚌埠 233030School of Pharmacy, Bengbu Medical College, Anhui Provincial Engineering Technology Research Center of Biochemical Pharmaceuticals, Bengbu 233030, China
| | - 耀帅 张
- />蚌埠医学院药学院, 安徽省生化药物工程技术研究中心, 安徽 蚌埠 233030School of Pharmacy, Bengbu Medical College, Anhui Provincial Engineering Technology Research Center of Biochemical Pharmaceuticals, Bengbu 233030, China
| | - 方田 范
- />蚌埠医学院药学院, 安徽省生化药物工程技术研究中心, 安徽 蚌埠 233030School of Pharmacy, Bengbu Medical College, Anhui Provincial Engineering Technology Research Center of Biochemical Pharmaceuticals, Bengbu 233030, China
| | - 娴 李
- />蚌埠医学院药学院, 安徽省生化药物工程技术研究中心, 安徽 蚌埠 233030School of Pharmacy, Bengbu Medical College, Anhui Provincial Engineering Technology Research Center of Biochemical Pharmaceuticals, Bengbu 233030, China
| | - 姗姗 李
- />蚌埠医学院药学院, 安徽省生化药物工程技术研究中心, 安徽 蚌埠 233030School of Pharmacy, Bengbu Medical College, Anhui Provincial Engineering Technology Research Center of Biochemical Pharmaceuticals, Bengbu 233030, China
| |
Collapse
|
54
|
Zhang A, Zhang S. Clinicopathological significance of vasculogenic mimicry and fetal hemoglobin expression in peripheral neuroblastic tumors in children. Am J Transl Res 2023; 15:4687-4698. [PMID: 37560203 PMCID: PMC10408510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 06/29/2023] [Indexed: 08/11/2023]
Abstract
PURPOSE Vasculogenic mimicry (VM) is present in a variety of malignant tumors, and is related to the degree of malignancy. Neuroblastoma (NB) can induce the expression of fetal hemoglobin (HB-F). The purpose of this study was to investigate the clinicopathological significance of the number of VMs and tumor cell expression of HB-F in children with peripheral neuroblastic tumors (pNTs). MATERIALS AND METHODS We collected tissue samples and clinical data from 101 children with pNTs; prepared serial sections of tissue wax blocks for hematoxylin and eosin staining, CD31/periodic acid-Schiff double staining, and HB-F immunohistochemical staining; and analyzed the experimental results. RESULTS There were significant differences in the number of VMs and HB-F expression in tumor cells according to the pathological classification of pNTs (P<0.001, collectively). Poorly differentiated NB had a median of 137 VMs and 25.5% HB-F expression. Differentiating NB had a median of 90.5 VMs and 8.5% HB-F expression. Ganglioneuroblastoma intermixed had a median of 6.0 VMs and 1.0% HB-F expression. Ganglioneuromas had no VM and a median of 0% HB-F expression. The number of VMs and the expression of HB-F were significantly higher in the poor prognosis group than the good prognosis group (P<0.001, collectively). There was a strong positive correlation between the number of VMs and HB-F expression in pNTs (r=0.891, P<0.001). CONCLUSION We confirmed VM and HB-F expression in pNTs. The number of VMs and HB-F expression were higher in poorly differentiated tumors. The number of VMs and level of HB-F expression in pNTs might be related to the prognosis.
Collapse
Affiliation(s)
- Aihua Zhang
- Graduate School, Tianjin Medical UniversityTianjin, China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical CenterTianjin, China
| |
Collapse
|
55
|
Schloo C, Kutscher LM. Modeling brain and neural crest neoplasms with human pluripotent stem cells. Neuro Oncol 2023; 25:1225-1235. [PMID: 36757217 PMCID: PMC10326493 DOI: 10.1093/neuonc/noad034] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Indexed: 02/10/2023] Open
Abstract
Pluripotent stem cells offer unique avenues to study human-specific aspects of disease and are a highly versatile tool in cancer research. Oncogenic processes and developmental programs often share overlapping transcriptomic and epigenetic signatures, which can be reactivated in induced pluripotent stem cells. With the emergence of brain organoids, the ability to recapitulate brain development and structure has vastly improved, making in vitro models more realistic and hence more suitable for biomedical modeling. This review highlights recent research and current challenges in human pluripotent stem cell modeling of brain and neural crest neoplasms, and concludes with a call for more rigorous quality control and for the development of models for rare tumor subtypes.
Collapse
Affiliation(s)
- Cedar Schloo
- Hopp Children’s Cancer Center (KiTZ), Heidelberg, Germany
- Division of Neuroblastoma Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lena M Kutscher
- Hopp Children’s Cancer Center (KiTZ), Heidelberg, Germany
- Developmental Origins of Pediatric Cancer Junior Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
56
|
Salamone TA, Rutigliano L, Pennacchi B, Cerra S, Matassa R, Nottola S, Sciubba F, Battocchio C, Marsotto M, Del Giudice A, Chumakov A, Davydok A, Grigorian S, Canettieri G, Agostinelli E, Fratoddi I. Thiol functionalised gold nanoparticles loaded with methotrexate for cancer treatment: From synthesis to in vitro studies on neuroblastoma cell lines. J Colloid Interface Sci 2023; 649:264-278. [PMID: 37348346 DOI: 10.1016/j.jcis.2023.06.078] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/05/2023] [Accepted: 06/13/2023] [Indexed: 06/24/2023]
Abstract
HYPOTHESIS Colloidal gold nanoparticles (AuNPs) functionalised with hydrophilic thiols can be used as drug delivery probes, thanks to their small size and hydrophilic character. AuNPs possess unique properties for their use in nanomedicine, especially in cancer treatment, as diagnostics and therapeutic tools. EXPERIMENTS Thiol functionalised AuNPs were synthesised and loaded with methotrexate (MTX). Spectroscopic and morphostructural characterisations evidenced the stability of the colloids upon interaction with MTX. Solid state (GISAXS, GIWAXS, FESEM, TEM, FTIR-ATR, XPS) and dispersed phase (UV-Vis, DLS, ζ-potential, NMR, SAXS) experiments allowed to understand structure-properties correlations. The nanoconjugate was tested in vitro (MTT assays) against two neuroblastoma cell lines: SNJKP and IMR5 with overexpressed n-Myc. FINDINGS Molar drug encapsulation efficiency was optimised to be >70%. A non-covalent interaction between the π system and the carboxylate moiety belonging to MTX and the charged aminic group of one of the thiols was found. The MTX loading slightly decreased the structural order of the system and increased the distance between the AuNPs. Free AuNPs showed no cytotoxicity whereas the AuNPs-MTX nanoconjugate had a more potent effect when compared to free MTX. The active role of AuNPs was evidenced by permeation studies: an improvement on penetration of the drug inside cells was evidenced.
Collapse
Affiliation(s)
- Tommaso A Salamone
- Department of Chemistry, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy.
| | - Lavinia Rutigliano
- Department of Molecular Medicine, Laboratory Affiliated to Istituto Pasteur Italia, Fondazione Cenci Bolognetti, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| | - Beatrice Pennacchi
- Department of Chemistry, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Sara Cerra
- Department of Chemistry, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Roberto Matassa
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Section of Human Anatomy, Sapienza University of Rome, Via A. Borelli 50, 00161 Rome, Italy
| | - Stefania Nottola
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Section of Human Anatomy, Sapienza University of Rome, Via A. Borelli 50, 00161 Rome, Italy
| | - Fabio Sciubba
- Department of Environmental Biology, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy; NMR-based Metabolomics Laboratory (NMLab), Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Chiara Battocchio
- Department of Sciences, Roma Tre University, Via della Vasca Navale 79, 00146 Rome, Italy
| | - Martina Marsotto
- Department of Sciences, Roma Tre University, Via della Vasca Navale 79, 00146 Rome, Italy
| | | | - Andrei Chumakov
- Deutsches Elektronen-Synchrotron DESY, Notkestr. 85, 22607 Hamburg, Germany
| | - Anton Davydok
- Institute of Material Physics, Helmholtz Zentrum Hereon, Notkestr. 85, 22607 Hamburg, Germany
| | - Souren Grigorian
- Department of Chemistry, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy; Institute of Physics, University of Siegen, Walter-Flex-Strasse 3, D-57068 Siegen, Germany
| | - Gianluca Canettieri
- Department of Molecular Medicine, Laboratory Affiliated to Istituto Pasteur Italia, Fondazione Cenci Bolognetti, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| | - Enzo Agostinelli
- Department of Sense Organs, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; International Polyamines Foundation "ETS-ONLUS", Via del Forte Tiburtino 98, 00159 Rome, Italy
| | - Ilaria Fratoddi
- Department of Chemistry, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy.
| |
Collapse
|
57
|
Julson JR, Quinn CH, Bownes LV, Hutchins SC, Stewart JE, Aye J, Yoon KJ, Beierle EA. Inhibition of PIM Kinases Promotes Neuroblastoma Cell Differentiation to a Neuronal Phenotype. J Pediatr Surg 2023; 58:1155-1163. [PMID: 36907773 PMCID: PMC10198809 DOI: 10.1016/j.jpedsurg.2023.02.018] [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: 01/24/2023] [Accepted: 02/10/2023] [Indexed: 02/19/2023]
Abstract
BACKGROUND Neuroblastoma arises from aberrancies in neural stem cell differentiation. PIM kinases contribute to cancer formation, but their precise role in neuroblastoma tumorigenesis is poorly understood. In the current study, we evaluated the effects of PIM kinase inhibition on neuroblastoma differentiation. METHODS Versteeg database query assessed the correlation between PIM gene expression and the expression of neuronal stemness markers and relapse free survival. PIM kinases were inhibited with AZD1208. Viability, proliferation, motility were measured in established neuroblastoma cells lines and high-risk neuroblastoma patient-derived xenografts (PDXs). qPCR and flow cytometry detected changes in neuronal stemness marker expression after AZD1208 treatment. RESULTS Database query showed increased levels of PIM1, PIM2, or PIM3 gene expression were associated with higher risk of recurrent or progressive neuroblastoma. Increased levels of PIM1 were associated with lower relapse free survival rates. Higher levels of PIM1 correlated with lower levels of neuronal stemness markers OCT4, NANOG, and SOX2. Treatment with AZD1208 resulted in increased expression of neuronal stemness markers. CONCLUSIONS Inhibition of PIM kinases differentiated neuroblastoma cancer cells toward a neuronal phenotype. Differentiation is a key component of preventing neuroblastoma relapse or recurrence and PIM kinase inhibition provides a potential new therapeutic strategy for this disease.
Collapse
Affiliation(s)
- Janet R Julson
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Colin H Quinn
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Laura V Bownes
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Sara C Hutchins
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Jerry E Stewart
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Jamie Aye
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Karina J Yoon
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Elizabeth A Beierle
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
| |
Collapse
|
58
|
Zhu K, Gao T, Wang Z, Zhang L, Tan K, Lv Z. RNA N6-methyladenosine reader IGF2BP3 interacts with MYCN and facilitates neuroblastoma cell proliferation. Cell Death Discov 2023; 9:151. [PMID: 37156775 PMCID: PMC10167253 DOI: 10.1038/s41420-023-01449-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/17/2023] [Accepted: 04/24/2023] [Indexed: 05/10/2023] Open
Abstract
Neuroblastoma (NB) is a kind of typical life-threatening extracranial tumor in children. N6-methyladenosine (m6A) modification is closely related to multiple cancer pathological processes. Insulin-like growth factor 2 mRNA binding protein 3 (IGF2BP3) is a top-ranked prognostic risk gene in NB; however, its function is uncertain. The expression of m6A-associated enzymes in patients with NB was analyzed using the Gene Expression Omnibus (GEO) and Therapeutically Applicable Research to Generate Effective Treatments (TARGET) database. The IGF2BP3 level in NB cell lines and primary samples was tested using quantitative real-time polymerase chain reaction (qRT-PCR), western blot method, and immunohistochemical analysis. The IGF2BP3 function in cell proliferation was clarified based on many functional in vitro and in vivo experiments. The interaction between IGF2BP3 and N-myc was researched via RNA immunoprecipitation (RIP), m6A RNA immunoprecipitation (MeRIP), and chromatin immunoprecipitation (ChIP) assays. The 16 m6A-regulated enzymes in NB were researched, and the result indicated that IGF2BP3 overexpression was related to cancer progression, COG risk, and survival based on the GEO and TARGET databases. Besides, the IGF2BP3 and MYCN levels were positively correlated. IGF2BP3 expression levels increased in MYCN-amplified NB clinical samples and cells. Knockdown of IGF2BP3 inhibited N-myc expression and NB cell proliferation in vitro and in vivo. IGF2BP3 regulates MYCN RNA stability by modifying m6A. In addition, we demonstrated that N-myc is a transcription factor that directly promotes IGF2BP3 expression in NB cells. IGF2BP3 regulates the proliferation of NB cells via m6A modification of MYCN. N-myc also acts as a transcription factor that regulates IGF2BP3 expression. A positive feedback loop between IGF2BP3 and N-myc facilitates NB cell proliferation.
Collapse
Affiliation(s)
- Kai Zhu
- Department of General Surgery, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, PR China
| | - Tingting Gao
- Department of General Surgery, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, PR China
| | - Zhiru Wang
- Department of General Surgery, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, PR China
| | - Liaoran Zhang
- Department of General Surgery, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, PR China
| | - Kezhe Tan
- Department of General Surgery, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, PR China
| | - Zhibao Lv
- Department of General Surgery, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, PR China.
| |
Collapse
|
59
|
Wang H, Xie M, Chen X, Zhu J, Ding H, Zhang L, Pan Z, He L. Development and validation of a CT-based radiomics signature for identifying high-risk neuroblastomas under the revised Children's Oncology Group classification system. Pediatr Blood Cancer 2023; 70:e30280. [PMID: 36881504 DOI: 10.1002/pbc.30280] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 02/09/2023] [Accepted: 02/13/2023] [Indexed: 03/08/2023]
Abstract
BACKGROUND To develop and validate a radiomics signature based on computed tomography (CT) for identifying high-risk neuroblastomas. PROCEDURE This retrospective study included 339 patients with neuroblastomas, who were classified into high-risk and non-high-risk groups according to the revised Children's Oncology Group classification system. These patients were then randomly divided into a training set (n = 237) and a testing set (n = 102). Pretherapy CT images of the arterial phase were segmented by two radiologists. Pyradiomics package and FeAture Explorer software were used to extract and process radiomics features. Radiomics models based on linear discriminant analysis (LDA), logistic regression (LR), and support vector machine (SVM) were constructed, and the area under the curve (AUC), 95% confidence interval (CI), and accuracy were calculated. RESULTS The optimal LDA, LR, and SVM models had 11, 12, and 14 radiomics features, respectively. The AUC of the LDA model in the training and testing sets were 0.877 (95% CI: 0.833-0.921) and 0.867 (95% CI: 0.797-0.937), with an accuracy of 0.823 and 0.804, respectively. The AUC of the LR model in the training and testing sets were 0.881 (95% CI: 0.839-0.924) and 0.855 (95% CI: 0.781-0.930), with an accuracy of 0.823 and 0.804, respectively. The AUC of the SVM model in the training and testing sets were 0.879 (95% CI: 0.836-0.923) and 0.862 (95% CI: 0.791-0.934), with an accuracy of 0.827 and 0.804, respectively. CONCLUSIONS CT-based radiomics is able to identify high-risk neuroblastomas and may provide additional image biomarkers for the identification of high-risk neuroblastomas.
Collapse
Affiliation(s)
- Haoru Wang
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Mingye Xie
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xin Chen
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Jin Zhu
- Department of Pathology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Hao Ding
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Li Zhang
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Zhengxia Pan
- Department of Cardiothoracic Surgery, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Ling He
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
60
|
Chai W, Wang X, Lu Z, Zhang S, Wang W, Wang H, Chen C, Yang W, Cheng H, Wang H, Feng J, Yang S, Li Q, Song W, Jin F, Zhang H, Su Y, Gui J. Elevated exosome-transferrable lncRNA EPB41L4A-AS1 in CD56 bright NK cells is responsible for the impaired NK function in neuroblastoma patients by suppressing cell glycolysis. Clin Immunol 2023; 250:109322. [PMID: 37024023 DOI: 10.1016/j.clim.2023.109322] [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: 12/26/2022] [Revised: 02/07/2023] [Accepted: 03/31/2023] [Indexed: 04/08/2023]
Abstract
NK cells are one of key immune components in neuroblastoma (NB) surveillance and eradication. Glucose metabolism as a major source of fuel for NK activation is exquisitely regulated. Our data revealed a diminished NK activation and a disproportionally augmented CD56bright subset in NB. Further study showed that NK cells in NB presented with an arrested glycolysis accompanied by an elevated expression of the long noncoding RNA (lncRNA) EPB41L4A-AS1, a known crucial participant in glycolysis regulation, in the CD56bright NK subset. The inhibitory function of lncRNA EPB41L4A-AS1 was recapitulated. Interestingly, our study demonstrated that exosomal lncRNA EPB41L4A-AS1 was transferrable from CD56bright NK to CD56dim NK and was able to quench the glycolysis of target NK. Our data demonstrated that an arrested glycolysis in patient NK cells was associated with an elevated lncRNA in CD56bright NK subset and a cross-talk between heterogeneous NK subsets was achieved by transferring metabolic inhibitory lncRNA through exosomes.
Collapse
Affiliation(s)
- Wenjia Chai
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, China; Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, China
| | - Xiaolin Wang
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, China; Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, China
| | - Zhengjing Lu
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, China; Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, China
| | - Shihan Zhang
- Medical Oncology Department, Pediatric Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Clinical Discipline of Pediatric Oncology, Key Laboratory of Major Diseases in Children, Ministry of Education, China
| | - Wei Wang
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, China; Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, China
| | - Hui Wang
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, China; Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, China
| | - Chenghao Chen
- Department of Thoracic Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Wei Yang
- Department of Surgical Oncology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, China
| | - Haiyan Cheng
- Department of Surgical Oncology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, China
| | - Huanmin Wang
- Department of Surgical Oncology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, China
| | - Jun Feng
- Department of Surgical Oncology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, China
| | - Shen Yang
- Department of Surgical Oncology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, China
| | - Qiliang Li
- Department of Clinical Laboratory Center, Beijing Children's Hospital, Capital Medical University, National Center for Children Health, Beijing 100045, China
| | - Wenqi Song
- Department of Clinical Laboratory Center, Beijing Children's Hospital, Capital Medical University, National Center for Children Health, Beijing 100045, China
| | - Fang Jin
- Department of Clinical Laboratory Center, Beijing Children's Hospital, Capital Medical University, National Center for Children Health, Beijing 100045, China
| | - Hui Zhang
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, China; Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, China
| | - Yan Su
- Medical Oncology Department, Pediatric Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Clinical Discipline of Pediatric Oncology, Key Laboratory of Major Diseases in Children, Ministry of Education, China
| | - Jingang Gui
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, China; Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, China.
| |
Collapse
|
61
|
TRAF4 Silencing Induces Cell Apoptosis and Improves Retinoic Acid Sensitivity in Human Neuroblastoma. Neurochem Res 2023; 48:2116-2128. [PMID: 36795185 DOI: 10.1007/s11064-023-03882-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/26/2023] [Accepted: 01/31/2023] [Indexed: 02/17/2023]
Abstract
Neuroblastoma (NB) is a pediatric malignancy that arises in the peripheral nervous system, and the prognosis in the high-risk group remains dismal, despite the breakthroughs in multidisciplinary treatments. The oral treatment with 13-cis-retinoic acid (RA) after high-dose chemotherapy and stem cell transplant has been proven to reduce the incidence of tumor relapse in children with high-risk neuroblastoma. However, many patients still have tumors relapsed following retinoid therapy, highlighting the need for the identification of resistant factors and the development of more effective treatments. Herein, we sought to investigate the potential oncogenic roles of the tumor necrosis factor (TNF) receptor-associated factor (TRAF) family in neuroblastoma and explore the correlation between TRAFs and retinoic acid sensitivity. We discovered that all TRAFs were efficiently expressed in neuroblastoma, but TRAF4, in particular, was found to be strongly expressed. The high expression of TRAF4 was associated with a poor prognosis in human neuroblastoma. The inhibition of TRAF4, rather than other TRAFs, improved retinoic acid sensitivity in two human neuroblastoma cell lines, SH-SY5Y and SK-N-AS cells. Further in vitro studies indicated that TRAF4 suppression induced retinoic acid-induced cell apoptosis in neuroblastoma cells, probably by upregulating the expression of Caspase 9 and AP1 while downregulating Bcl-2, Survivin, and IRF-1. Notably, the improved anti-tumor effects from the combination of TRAF4 knockdown and retinoic acid were confirmed in vivo using the SK-N-AS human neuroblastoma xenograft model. In conclusion, the highly expressed TRAF4 might be implicated in developing resistance to retinoic acid treatment in neuroblastoma, and the combination therapy with retinoic acid and TRAF4 inhibition may offer significant therapeutic advantages in the treatment of relapsed neuroblastoma.
Collapse
|
62
|
The RUNX Family Defines Trk Phenotype and Aggressiveness of Human Neuroblastoma through Regulation of p53 and MYCN. Cells 2023; 12:cells12040544. [PMID: 36831211 PMCID: PMC9954111 DOI: 10.3390/cells12040544] [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: 01/13/2023] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
The Runt-related transcription factor (RUNX) family, which is essential for the differentiation of cells of neural crest origin, also plays a potential role in neuroblastoma tumorigenesis. Consecutive studies in various tumor types have demonstrated that the RUNX family can play either pro-tumorigenic or anti-tumorigenic roles in a context-dependent manner, including in response to chemotherapeutic agents. However, in primary neuroblastomas, RUNX3 acts as a tumor-suppressor, whereas RUNX1 bifunctionally regulates cell proliferation according to the characterized genetic and epigenetic backgrounds, including MYCN oncogenesis. In this review, we first highlight the current knowledge regarding the mechanism through which the RUNX family regulates the neurotrophin receptors known as the tropomyosin-related kinase (Trk) family, which are significantly associated with neuroblastoma aggressiveness. We then focus on the possible involvement of the RUNX family in functional alterations of the p53 family members that execute either tumor-suppressive or dominant-negative functions in neuroblastoma tumorigenesis. By examining the tripartite relationship between the RUNX, Trk, and p53 families, in addition to the oncogene MYCN, we endeavor to elucidate the possible contribution of the RUNX family to neuroblastoma tumorigenesis for a better understanding of potential future molecular-based therapies.
Collapse
|
63
|
Wieczorek A, Zaniewska-Tekieli A, Ehlert K, Pawinska-Wasikowska K, Balwierz W, Lode H. Dinutuximab beta combined with chemotherapy in patients with relapsed or refractory neuroblastoma. Front Oncol 2023; 13:1082771. [PMID: 36816982 PMCID: PMC9936065 DOI: 10.3389/fonc.2023.1082771] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/23/2023] [Indexed: 02/05/2023] Open
Abstract
Prognosis in children with refractory and relapsed high-risk neuroblastoma is poor. Only a minority of patients obtain remission when treated with second-line chemotherapy regimens. Chemotherapy combined with anti-GD2 antibodies has previously been shown to increase response and survival rates. We retrospectively analyzed a cohort of 25 patients with relapsed or refractory high-risk neuroblastoma who were treated with irinotecan/temozolomide chemotherapy in combination with the anti-GD2 antibody dinutuximab beta. The therapy resulted in an objective response rate of 64%, with 32% of patients achieving a complete response. Response to treatment was observed in patients with refractory disease (n=5) and those with first (n=12) or consecutive (n=8) relapses, including patients with progressing disease. In four patients, best response was achieved after more than 5 cycles, suggesting that some patients may benefit from prolonged chemotherapy and dinutuximab beta treatment. Fourteen of our 25 patients had previously received dinutuximab beta, four of whom achieved complete response and six partial response (objective response rate 71%). The therapy was well tolerated, even in heavily pre-treated patients and those who had previously received dinutuximab beta treatment. Toxicities were comparable to those previously reported for the individual therapies, and no discontinuations due to toxicities occurred. Combination of chemotherapy with dinutuximab beta is a promising treatment option for patients with relapsed or refractory high-risk neuroblastoma and should be further explored in clinical studies.
Collapse
Affiliation(s)
- Aleksandra Wieczorek
- Pediatric Oncology and Hematology, Jagiellonian University Medical College, Krakow, Poland
- Pediatric Oncology and Hematology, University Children's Hospital of Krakow, Krakow, Poland
| | - Anna Zaniewska-Tekieli
- Pediatric Oncology and Hematology, University Children's Hospital of Krakow, Krakow, Poland
| | - Karoline Ehlert
- Pediatric Hematology and Oncology, University Medicine Greifswald, Greifswald, Germany
| | - Katarzyna Pawinska-Wasikowska
- Pediatric Oncology and Hematology, Jagiellonian University Medical College, Krakow, Poland
- Pediatric Oncology and Hematology, University Children's Hospital of Krakow, Krakow, Poland
| | - Walentyna Balwierz
- Pediatric Oncology and Hematology, Jagiellonian University Medical College, Krakow, Poland
- Pediatric Oncology and Hematology, University Children's Hospital of Krakow, Krakow, Poland
| | - Holger Lode
- Pediatric Hematology and Oncology, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
64
|
Breedy S, Ratnayake W, Lajmi L, Hill R, Acevedo-Duncan M. 14-3-3 and Smad2/3 are crucial mediators of atypical-PKCs: Implications for neuroblastoma progression. Front Oncol 2023; 13:1051516. [PMID: 36776326 PMCID: PMC9910080 DOI: 10.3389/fonc.2023.1051516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 01/06/2023] [Indexed: 01/28/2023] Open
Abstract
Neuroblastoma (NB) is a cancer that develops in the neuroblasts. It is the most common cancer in children under the age of 1 year, accounting for approximately 6% of all cancers. The prognosis of NB is linked to both age and degree of cell differentiation. This results in a range of survival rates for patients, with outcomes ranging from recurrence and mortality to high survival rates and tumor regression. Our previous work indicated that PKC-ι promotes cell proliferation in NB cells through the PKC-ι/Cdk7/Cdk2 cascade. We report on two atypical protein kinase inhibitors as potential therapeutic candidates against BE(2)-C and BE(2)-M17 cells: a PKC-ι-specific 5-amino-1-2,3-dihydroxy-4-(methylcyclopentyl)-1H-imidazole-4-carboxamide and a PKC-ζ specific 8-hydroxy-1,3,6-naphthalenetrisulfonic acid. Both compounds induced apoptosis and retarded the epithelial-mesenchymal transition (EMT) of NB cells. Proteins 14-3-3 and Smad2/3 acted as central regulators of aPKC-driven progression in BE(2)-C and BE(2)-M17 cells in relation to the Akt1/NF-κB and TGF-β pathways. Data indicates that aPKCs upregulate Akt1/NF-κB and TGF-β pathways in NB cells through an association with 14-3-3 and Smad2/3 that can be diminished by aPKC inhibitors. In summary, both inhibitors appear to be promising potential neuroblastoma therapeutics and merit further research.
Collapse
Affiliation(s)
- S. Breedy
- Department of Chemistry, University of South Florida, Tampa, FL, United States
| | - W.S. Ratnayake
- Department of Chemistry, University of South Florida, Tampa, FL, United States
| | - L. Lajmi
- Department of Chemistry, University of South Florida, Tampa, FL, United States
| | - R. Hill
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL, United States
| | - M. Acevedo-Duncan
- Department of Chemistry, University of South Florida, Tampa, FL, United States
| |
Collapse
|
65
|
Pulido R, Nunes-Xavier CE. Editorial: Cell and Developmental Signalling in Neuroblastoma. Front Cell Dev Biol 2023; 10:1126352. [PMID: 36684434 PMCID: PMC9846788 DOI: 10.3389/fcell.2022.1126352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 12/19/2022] [Indexed: 01/06/2023] Open
Affiliation(s)
- Rafael Pulido
- Biomarkers in Cancer, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain,Ikerbasque, Basque Foundation for Science, Bilbao, Spain,*Correspondence: Rafael Pulido, ; Caroline E. Nunes-Xavier,
| | - Caroline E. Nunes-Xavier
- Biomarkers in Cancer, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain,Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway,*Correspondence: Rafael Pulido, ; Caroline E. Nunes-Xavier,
| |
Collapse
|
66
|
Liu X, Wang Z, Xiong X, Li C, Wu Y, Su M, Yang S, Zeng M, Weng W, Huang K, Zhou D, Fang J, Xu L, Li P, Zhu Y, Qiu K, Ma Y, Lei J, Li Y. Arsenic Trioxide inhibits Activation of Hedgehog Pathway in Human Neuroblastoma Cell Line SK-N-BE(2) Independent of Itraconazole. Anticancer Agents Med Chem 2023; 23:2217-2224. [PMID: 37888819 DOI: 10.2174/0118715206259952230919173611] [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: 04/28/2023] [Revised: 07/21/2023] [Accepted: 08/18/2023] [Indexed: 10/28/2023]
Abstract
BACKGROUND Neuroblastoma (NB) remains associated with a low overall survival rate over the long term. Abnormal activation of the Hedgehog (HH) signaling pathway can activate the transcription of various downstream target genes that promote NB. Both arsenic trioxide (ATO) and itraconazole (ITRA) can inhibit tumor growth. OBJECTIVE To determine whether ATO combined with ITRA can be used to treat NB with HH pathway activation, we examined the effects of ATO and ITRA monotherapy or combined inhibition of the HH pathway in NB. METHODS Analysis of CCK8 and flow cytometry showed cell inhibition and cell cycle, respectively. Real-time PCR analysis was conducted to assess the mRNA expression of HH pathway. RESULTS We revealed that as concentrations of ATO and ITRA increased, the killing effects of both agents on SK-N-BE(2) cells became more apparent. During G2/M, the cell cycle was largely arrested by ATO alone and combined with ITRA, and in the G0/G1 phase by ITRA alone. In the HH pathway, ATO inhibited the transcription of the SHH, PTCH1, SMO and GLI2 genes, however, ITRA did not. Instead of showing synergistic effects in a combined mode, ITRA decreased ATO inhibitory effects. CONCLUSION We showed that ATO is an important inhibitor of HH pathway but ITRA can weaken the inhibitory effect of ATO. This study provides an experimental evidence for the clinical use of ATO and ITRA in the treatment of NB with HH pathway activation in cytology.
Collapse
Affiliation(s)
- Xiaoshan Liu
- Pediatric Hematology/Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhixuan Wang
- Pediatric Hematology/Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xilin Xiong
- Pediatric Hematology/Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Chunmou Li
- Department of Pediatrics, the Seventh Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
| | - Yu Wu
- Pediatric Hematology/Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Mingwei Su
- Pediatric Hematology/Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Shu Yang
- Pediatric Hematology/Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Meilin Zeng
- Pediatric Hematology/Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Wenjun Weng
- Pediatric Hematology/Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ke Huang
- Pediatric Hematology/Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Dunhua Zhou
- Pediatric Hematology/Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jianpei Fang
- Pediatric Hematology/Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Lvhong Xu
- Pediatric Hematology/Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Peng Li
- South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yafeng Zhu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Guangzhou, China
| | - Kunyin Qiu
- Pediatric Hematology/Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yuhan Ma
- Pediatric Hematology/Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jiaying Lei
- Pediatric Hematology/Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yang Li
- Pediatric Hematology/Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
67
|
Chen W, Lin P, Bai J, Fang Y, Zhang B. Establishment and validation of a nomogram to predict cancer-specific survival in pediatric neuroblastoma patients. Front Pediatr 2023; 11:1105922. [PMID: 36937951 PMCID: PMC10020339 DOI: 10.3389/fped.2023.1105922] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 02/16/2023] [Indexed: 03/06/2023] Open
Abstract
Background The term "neuroblastoma (NB)" refers to a type of solid pediatric tumor that develops from undivided neuronal cells. According to the American Cancer Society report, between 700 and 800 children under the age of 14 are diagnosed with NB every year in the United States (U.S.). About 6% of all cases of pediatric cancer in the U.S. are caused by NB. NB is the most frequent malignancy in children younger than 1 year; however, it is rarely found in those over the age of 10 and above. Objective To accurately predict cancer-specific survival (CSS) in children with NB, this research developed and validated an all-encompassing prediction model. Methods The present retrospective study used the Surveillance, Epidemiology, and End Results (SEER) database to collect information on 1,448 individuals diagnosed with NB between 1998 and 2019. The pool of potentially eligible patients was randomly split into two groups, a training cohort (N = 1,013) and a validation cohort (N = 435). Using multivariate Cox stepwise regression, we were able to identify the components that independently predicted outcomes. The accuracy of this nomogram was measured employing the consistency index (C-index), area under the time-dependent receiver operating characteristic curve (AUC), calibration curve, and decision-curve analysis (DCA). Results In this study, we found that age, primary location, tumor size, summary stage, chemotherapy, and surgery were all significant predictors of CSS outcomes and integrated them into our model accordingly. The C-index for the validation cohort was 0.812 (95% CI: 0.773-0.851), while for the training cohort it was 0.795 (95% CI: 0.767-0.823). The C-indexes and AUC values show that the nomogram is able to discriminate well enough. The calibration curves suggest that the nomogram is quite accurate. Also, the DCA curves demonstrated the prediction model's value. Conclusion A novel nomogram was developed and validated in this work to assess personalized CSS in NB patients, and it has been indicated that this model could be a useful tool for calculating NB patients' survival on an individual basis and enhancing therapeutic decision-making.
Collapse
Affiliation(s)
- Weiming Chen
- Department of Pediatric Surgery, Fujian Children’s Hospital (Fujian Branch of Shanghai Children’s Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Ping Lin
- Department of Hematology and Oncology, Fujian Children’s Hospital (Fujian Branch of Shanghai Children’s Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Jianxi Bai
- Department of Pediatric Surgery, Fujian Children’s Hospital (Fujian Branch of Shanghai Children’s Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Yifan Fang
- Department of Pediatric Surgery, Fujian Children’s Hospital (Fujian Branch of Shanghai Children’s Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- Correspondence: Bing Zhang Yifan Fang
| | - Bing Zhang
- Department of Pediatric Surgery, Fujian Children’s Hospital (Fujian Branch of Shanghai Children’s Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- Correspondence: Bing Zhang Yifan Fang
| |
Collapse
|
68
|
Remarkable Synergy When Combining EZH2 Inhibitors with YM155 Is H3K27me3-Independent. Cancers (Basel) 2022; 15:cancers15010208. [PMID: 36612203 PMCID: PMC9818370 DOI: 10.3390/cancers15010208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 12/20/2022] [Accepted: 12/28/2022] [Indexed: 01/01/2023] Open
Abstract
Targeting multiple molecules in the same biological network may maximize therapeutic efficacy. In this study, we identified a 27-gene module that is highly expressed in solid tumors, encoding actionable targets including EZH2 and BIRC5. The combination of EZH2 inhibitors and a BIRC5 inhibitor, YM155, results in a remarkable synergistic effect. The action of EZH2 inhibitors in this process is independent of the histone methyltransferase activity of polycomb repressive complex 2. Our study reveals a potential therapeutic approach for treating solid tumors by simultaneously targeting EZH2 and BIRC5.
Collapse
|
69
|
Singh A, Rawat S, Yadav G, Kushwaha R, Verma SP, Singh US. Study of Hemato-morphological Features in Neuroblastoma Infiltrating Marrow. J Lab Physicians 2022. [DOI: 10.1055/s-0042-1758667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Abstract
Objective Neuroblastoma typically affects children within the first 5 years of life and accounts for 10% of all pediatric malignancies. Neuroblastoma at onset may manifest as a localized or metastatic illness. The aim of this study was to identify hematomorphological features in neuroblastoma infiltrating marrow as well as to ascertain the prevalence of bone marrow infiltration in neuroblastoma.
Materials and Methods This retrospective study included newly diagnosed 79 cases of neuroblastoma, which were referred for bone marrow examination for the staging of the disease. Medical records were retrieved to acquire hematomorphological findings of peripheral blood and bone marrow smears. Statistical Package for Social Sciences, IBM Inc., USA, version 21.0 was used to analyze the data.
Results The interquartile age range of neuroblastoma cases was 24.0 to 72.0 months (median = 48 months) with a male to female ratio of 2.7:1. Also, 55.6% (44/79) of cases in the study population showed evidence of marrow infiltration. The bone marrow infiltration was significantly linked to thrombocytopenia (p = 0.043) and nucleated red blood cells (p = 0.003) in peripheral blood. The bone marrow smears of cases with infiltration showed a significant shift to the left in the myeloid series (p = 0.001) and an increased number of erythroid cells (p = 0.001).
Conclusion For neuroblastoma patients, a diligent, exhaustive search for infiltrating cells in bone marrow is advised if thrombocytopenia or nucleated red blood cells are identified on a peripheral blood smear and bone marrow smears showed myeloid left shift with an increased number of erythroid cells.
Collapse
Affiliation(s)
- Anurag Singh
- Department of Pathology, King George Medical University, Lucknow, Uttar Pradesh, India
| | - Shalini Rawat
- Department of Pathology, King George Medical University, Lucknow, Uttar Pradesh, India
| | - Geeta Yadav
- Department of Pathology, King George Medical University, Lucknow, Uttar Pradesh, India
| | - Rashmi Kushwaha
- Department of Pathology, King George Medical University, Lucknow, Uttar Pradesh, India
| | - Shailendra Prasad Verma
- Department of Clinical Hematology, King George Medical University, Lucknow, Uttar Pradesh, India
| | - Uma S. Singh
- Department of Pathology, King George Medical University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
70
|
Julson JR, Beierle EA. Embryology and surgical anatomy of pediatric solid tumors. Semin Pediatr Surg 2022; 31:151233. [PMID: 36423516 PMCID: PMC10084944 DOI: 10.1016/j.sempedsurg.2022.151233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- J R Julson
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - E A Beierle
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA.
| |
Collapse
|
71
|
SOX4 Mediates ATRA-Induced Differentiation in Neuroblastoma Cells. Cancers (Basel) 2022; 14:cancers14225642. [PMID: 36428735 PMCID: PMC9688885 DOI: 10.3390/cancers14225642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/10/2022] [Accepted: 11/12/2022] [Indexed: 11/19/2022] Open
Abstract
Neuroblastoma (NB), which is considered to be caused by the differentiation failure of neural crest cells, is the most common extracranial malignant solid tumor in children. The degree of tumor differentiation in patients with NB is closely correlated with the survival rate. To explore the potential targets that mediate NB cell differentiation, we analyzed four microarray datasets from GEO, and the overlapping down- or upregulated DEGs were displayed using Venn diagrams. SOX4 was one of the overlapping upregulated DEGs and was confirmed by RT-qPCR and Western blot in ATRA-treated NGP, SY5Y, and BE2 cells. To clarify whether SOX4 was the target gene regulating NB cell differentiation, the correlation between the expression of SOX4 and the survival of clinical patients was analyzed via the R2 database, SOX4 overexpression plasmids and siRNAs were generated to change the expression of SOX4, RT-qPCR and Western blot were performed to detect SOX4 expression, cell confluence or cell survival was detected by IncuCyte Zoom or CCK8 assay, immunocytochemistry staining was performed to detect cells' neurites, and a cell cycle analysis was implemented using Flow cytometry after PI staining. The results showed that the survival probabilities were positively correlated with SOX4 expression, in which overexpressing SOX4 inhibited NB cell proliferation, elongated the cells' neurite, and blocked the cell cycle in G1 phase, and that knockdown of the expression of SOX4 partially reversed the ATRA-induced inhibition of NB cell proliferation, the elongation of the cells' neurites, and the blocking of the cell cycle in the G1 phase. These indicate that SOX4 may be a target to induce NB cell differentiation.
Collapse
|
72
|
Nino N, Ishida T, Nakatani N, Lin KS, Win KHN, Mon CY, Nishimura A, Inoue S, Tamura A, Yamamoto N, Uemura S, Saito A, Mori T, Hasegawa D, Kosaka Y, Nozu K, Nishimura N. Minimal residual disease detected by droplet digital PCR in peripheral blood stem cell grafts has a prognostic impact on high-risk neuroblastoma patients. Heliyon 2022; 8:e10978. [PMID: 36276741 PMCID: PMC9578974 DOI: 10.1016/j.heliyon.2022.e10978] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 09/30/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022] Open
Abstract
More than half of high-risk neuroblastoma (NB) patients have experienced relapse due to the activation of chemoresistant minimal residual disease (MRD) even though they are treated by high-dose chemotherapy with autologous peripheral blood stem cell (PBSC) transplantation. Although MRD in high-risk NB patients can be evaluated by quantitative PCR with several sets of neuroblastoma-associated mRNAs (NB-mRNAs), the prognostic significance of MRD in PBSC grafts (PBSC-MRD) is unclear. In the present study, we collected 20 PBSC grafts from 20 high-risk NB patients and evaluated PBSC-MRD detected by droplet digital PCR (ddPCR) with 7NB-mRNAs (CRMP1, DBH, DDC, GAP43, ISL1, PHOX2B, and TH mRNA). PBSC-MRD in 11 relapsed patients was significantly higher than that in 9 non-relapsed patients. Patients with a higher PBSC-MRD had a lower 3-year event-free survival (P = 0.0148). The present study suggests that PBSC-MRD detected by ddPCR with 7NB-mRNAs has a prognostic impact on high-risk NB patients.
Collapse
Affiliation(s)
- Nanako Nino
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Toshiaki Ishida
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe, Japan
| | - Naoko Nakatani
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kyaw San Lin
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kaung Htet Nay Win
- Department of Public Health, Kobe University Graduate School of Health Science, Kobe, Japan
| | - Cho Yee Mon
- Department of Public Health, Kobe University Graduate School of Health Science, Kobe, Japan
| | - Akihiro Nishimura
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shotaro Inoue
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Akihiro Tamura
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Nobuyuki Yamamoto
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Suguru Uemura
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe, Japan
| | - Atsuro Saito
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe, Japan
| | - Takeshi Mori
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe, Japan
| | - Daiichiro Hasegawa
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe, Japan
| | - Yoshiyuki Kosaka
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe, Japan
| | - Kandai Nozu
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Noriyuki Nishimura
- Department of Public Health, Kobe University Graduate School of Health Science, Kobe, Japan,Corresponding author.
| |
Collapse
|
73
|
Parkinson LM, Gillen SL, Woods LM, Chaytor L, Marcos D, Ali FR, Carroll JS, Philpott A. The proneural transcription factor ASCL1 regulates cell proliferation and primes for differentiation in neuroblastoma. Front Cell Dev Biol 2022; 10:942579. [PMID: 36263020 PMCID: PMC9574099 DOI: 10.3389/fcell.2022.942579] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/26/2022] [Indexed: 11/27/2022] Open
Abstract
Neuroblastoma is believed to arise from sympathetic neuroblast precursors that fail to engage the neuronal differentiation programme, but instead become locked in a pro-proliferative developmental state. Achaete-scute homolog 1 (ASCL1) is a proneural master regulator of transcription which modulates both proliferation and differentiation of sympathetic neuroblast precursor cells during development, while its expression has been implicated in the maintenance of an oncogenic programme in MYCN-amplified neuroblastoma. However, the role of ASCL1 expression in neuroblastoma is not clear, especially as its levels vary considerably in different neuroblastoma cell lines. Here, we have investigated the role of ASCL1 in maintaining proliferation and controlling differentiation in both MYCN amplified and Anaplastic Lymphoma Kinase (ALK)-driven neuroblastoma cells. Using CRISPR deletion, we generated neuroblastoma cell lines lacking ASCL1 expression, and these grew more slowly than parental cells, indicating that ASCL1 contributes to rapid proliferation of MYCN amplified and non-amplified neuroblastoma cells. Genome-wide analysis after ASCL1 deletion revealed reduced expression of genes associated with neuronal differentiation, while chromatin accessibility at regulatory regions associated with differentiation genes was also attenuated by ASCL1 knock-out. In neuroblastoma, ASCL1 has been described as part of a core regulatory circuit of developmental regulators whose high expression is maintained by mutual cross-activation of a network of super enhancers and is further augmented by the activity of MYC/MYCN. Surprisingly, ASCL1 deletion had little effect on the transcription of CRC gene transcripts in these neuroblastoma cell lines, but the ability of MYC/MYCN and CRC component proteins, PHOX2B and GATA3, to bind to chromatin was compromised. Taken together, our results demonstrate several roles for endogenous ASCL1 in neuroblastoma cells: maintaining a highly proliferative phenotype, regulating DNA binding of the core regulatory circuit genes to chromatin, while also controlling accessibility and transcription of differentiation targets. Thus, we propose a model where ASCL1, a key developmental regulator of sympathetic neurogenesis, plays a pivotal role in maintaining proliferation while simultaneously priming cells for differentiation in neuroblastoma.
Collapse
Affiliation(s)
- Lydia M. Parkinson
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Sarah L. Gillen
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Laura M. Woods
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Lewis Chaytor
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Daniel Marcos
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Fahad R. Ali
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, United Kingdom
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Jason S. Carroll
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Anna Philpott
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, United Kingdom
| |
Collapse
|
74
|
Single-cell profiling of peripheral neuroblastic tumors identifies an aggressive transitional state that bridges an adrenergic-mesenchymal trajectory. Cell Rep 2022; 41:111455. [DOI: 10.1016/j.celrep.2022.111455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 04/16/2022] [Accepted: 09/14/2022] [Indexed: 11/21/2022] Open
|
75
|
Pastorino F, Capasso M, Brignole C, Giglio S, Bensa V, Cantalupo S, Lasorsa VA, Tondo A, Mura R, Sementa AR, Garaventa A, Ponzoni M, Amoroso L. Italian Precision Medicine in Pediatric Oncology: Moving beyond Actionable Alterations. Int J Mol Sci 2022; 23:ijms231911236. [PMID: 36232538 PMCID: PMC9570321 DOI: 10.3390/ijms231911236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Neuroblastoma (NB) is the most common extracranial solid tumor encountered in childhood. Although there has been significant improvement in the outcomes of patients with high-risk disease, the prognosis for patients with metastatic relapse or refractory disease is poor. Hence, the clinical integration of genome sequencing into standard clinical practice is necessary in order to develop personalized therapy for children with relapsed or refractory disease. The PeRsonalizEdMEdicine (PREME) project focuses on the design of innovative therapeutic strategies for patients suffering from relapsed NB. We performed whole exome sequencing (WES) of patient-matched tumor-normal samples to identify genetic variants amenable to precision medicine. Specifically, two patients were studied (First case: a three-year-old male with early relapsed NB; Second case: a 20-year-old male who relapsed 10 years after the first diagnosis of NB). Results were reviewed by a multi-disciplinary molecular tumor board (MTB) and clinical reports were issued to the ordering physician. WES revealed the mutation c.G320C in the CUL4A gene in case 1 and the mutation c.A484G in the PSMC2 gene in case 2. Both patients were treated according to these actionable alterations, with promising results. The effective treatment of NB is one of the main challenges in pediatric oncology. In the era of precision medicine, the need to design new therapeutic strategies for NB is fundamental. Our results demonstrate the feasibility of incorporating clinical WES into pediatric oncology practice.
Collapse
Affiliation(s)
- Fabio Pastorino
- Laboratorio di Terapie Sperimentali in Oncologia, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
- These authors contributed equally to this work
| | - Mario Capasso
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80131 Napoli, Italy
- CEINGE Biotecnologie Avanzate, 80131 Napoli, Italy
- These authors contributed equally to this work
| | - Chiara Brignole
- Laboratorio di Terapie Sperimentali in Oncologia, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Serena Giglio
- UOC Oncologia, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Veronica Bensa
- Laboratorio di Terapie Sperimentali in Oncologia, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Sueva Cantalupo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80131 Napoli, Italy
- CEINGE Biotecnologie Avanzate, 80131 Napoli, Italy
| | | | - Annalisa Tondo
- Dipartimento di Oncoematologia, Ospedale Meyer, 50139 Firenze, Italy
| | - Rossella Mura
- Oncoematologia Pediatrica, Ospedale Pediatrico Microcitemico “Antonio Cao” Azienda Ospedaliera Brotzu, 09121 Cagliari, Italy
| | - Angela Rita Sementa
- Dipartimento di Patologia, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | | | - Mirco Ponzoni
- Laboratorio di Terapie Sperimentali in Oncologia, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
- Correspondence: ; Tel.: +39-0105-636-3539; Fax: +39-0103-779-820
| | - Loredana Amoroso
- UOC Oncologia, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
- These authors contributed equally to this work
| |
Collapse
|
76
|
Arman T, Nelson PS. Endocrine and paracrine characteristics of neuroendocrine prostate cancer. Front Endocrinol (Lausanne) 2022; 13:1012005. [PMID: 36440195 PMCID: PMC9691667 DOI: 10.3389/fendo.2022.1012005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/24/2022] [Indexed: 11/12/2022] Open
Abstract
Prostate cancer is a common malignancy affecting men worldwide. While the vast majority of newly diagnosed prostate cancers are categorized as adenocarcinomas, a spectrum of uncommon tumor types occur including those with small cell and neuroendocrine cell features. Benign neuroendocrine cells exist in the normal prostate microenvironment, and these cells may give rise to primary neuroendocrine carcinomas. However, the more common development of neuroendocrine prostate cancer is observed after therapeutics designed to repress the signaling program regulated by the androgen receptor which is active in the majority of localized and metastatic adenocarcinomas. Neuroendocrine tumors are identified through immunohistochemical staining for common markers including chromogranin A/B, synaptophysin and neuron specific enolase (NSE). These markers are also common to neuroendocrine tumors that arise in other tissues and organs such as the gastrointestinal tract, pancreas, lung and skin. Notably, neuroendocrine prostate cancer shares biochemical features with nerve cells, particularly functions involving the secretion of a variety of peptides and proteins. These secreted factors have the potential to exert local paracrine effects, and distant endocrine effects that may modulate tumor progression, invasion, and resistance to therapy. This review discusses the spectrum of factors derived from neuroendocrine prostate cancers and their potential to influence the pathophysiology of localized and metastatic prostate cancer.
Collapse
Affiliation(s)
- Tarana Arman
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Peter S. Nelson
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, United States
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA, United States
- *Correspondence: Peter S. Nelson,
| |
Collapse
|
77
|
Uttinger KL, Riedmeier M, Reibetanz J, Meyer T, Germer CT, Fassnacht M, Wiegering A, Wiegering V. Adrenalectomies in children and adolescents in Germany - a diagnose related groups based analysis from 2009-2017. Front Endocrinol (Lausanne) 2022; 13:914449. [PMID: 35966067 PMCID: PMC9363694 DOI: 10.3389/fendo.2022.914449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/05/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Adrenalectomies are rare procedures especially in childhood. So far, no large cohort study on this topic has been published with data on to age distribution, operative procedures, hospital volume and operative outcome. METHODS This is a retrospective analysis of anonymized nationwide hospital billing data (DRG data, 2009-2017). All adrenal surgeries (defined by OPS codes) of patients between the age 0 and 21 years in Germany were included. RESULTS A total of 523 patient records were identified. The mean age was 8.6 ± 7.7 years and 262 patients were female (50.1%). The majority of patients were between 0 and 5 years old (52% overall), while 11.1% were between 6 and 11 and 38.8% older than 12 years. The most common diagnoses were malignant neoplasms of the adrenal gland (56%, mostly neuroblastoma) with the majority being younger than 5 years. Benign neoplasms in the adrenal gland (D350) account for 29% of all cases with the majority of affected patients being 12 years or older. 15% were not defined regarding tumor behavior. Overall complication rate was 27% with a clear higher complication rate in resection for malignant neoplasia of the adrenal gland. Bleeding occurrence and transfusions are the main complications, followed by the necessary of relaparotomy. There was an uneven patient distribution between hospital tertiles (low volume, medium and high volume tertile). While 164 patients received surgery in 85 different "low volume" hospitals (0.2 cases per hospital per year), 205 patients received surgery in 8 different "high volume" hospitals (2.8 cases per hospital per year; p<0.001). Patients in high volume centers were significant younger, had more extended resections and more often malignant neoplasia. In multivariable analysis younger age, extended resections and open procedures were independent predictors for occurrence of postoperative complications. CONCLUSION Overall complication rate of adrenalectomies in the pediatric population in Germany is low, demonstrating good therapeutic quality. Our analysis revealed a very uneven distribution of patient volume among hospitals.
Collapse
Affiliation(s)
- Konstantin L. Uttinger
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, Würzburg University Hospital, Würzburg, Germany
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, Leipzig University Hospital, Leipzig, Germany
| | - Maria Riedmeier
- Department of Pediatrics, Ped. Hematology, Oncology and Stem Cell Transplantation, Würzburg University Hospital, Würzburg, Germany
| | - Joachim Reibetanz
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, Würzburg University Hospital, Würzburg, Germany
| | - Thomas Meyer
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, Würzburg University Hospital, Würzburg, Germany
| | - Christoph Thomas Germer
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, Würzburg University Hospital, Würzburg, Germany
- Comprehensive Cancer Centre Mainfranken, University of Würzburg Medical Centre, Würzburg, Germany
| | - Martin Fassnacht
- Department of Endocrine Medicine, Würzburg University Hospital, Würzburg, Germany
| | - Armin Wiegering
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, Würzburg University Hospital, Würzburg, Germany
- Comprehensive Cancer Centre Mainfranken, University of Würzburg Medical Centre, Würzburg, Germany
- Department of Biochemistry and Molecular Biology , University of Würzburg, Würzburg, Germany
| | - Verena Wiegering
- Department of Pediatrics, Ped. Hematology, Oncology and Stem Cell Transplantation, Würzburg University Hospital, Würzburg, Germany
- *Correspondence: Verena Wiegering,
| |
Collapse
|
78
|
Liang J, Li X, Dong Y, Zhao B. Modeling Human Organ Development and Diseases With Fetal Tissue-Derived Organoids. Cell Transplant 2022; 31:9636897221124481. [PMID: 36121224 PMCID: PMC9490458 DOI: 10.1177/09636897221124481] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Recent advances in human organoid technology have greatly facilitated the study of organ development and pathology. In most cases, these organoids are derived from either pluripotent stem cells or adult stem cells for the modeling of developmental events and tissue homeostasis. However, due to the lack of human fetal tissue references and research model, it is still challenging to capture early developmental changes and underlying mechanisms in human embryonic development. The establishment of fetal tissue–derived organoids in rigorous time points is necessary. Here we provide an overview of the strategies and applications of fetal tissue–derived organoids, mainly focusing on fetal organ development research, developmental defect disease modeling, and organ–organ interaction study. Discussion of the importance of fetal tissue research also highlights the prospects and challenges in this field.
Collapse
Affiliation(s)
- Jianqing Liang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xinyang Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yateng Dong
- bioGenous Biotechnology, Inc., Hangzhou, China
| | - Bing Zhao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|