1
|
He J, Wang Z, Yu X, Su Y, Hong M, Zhu K. Promoting application of enhanced recovery after surgery protocols during perioperative localized abdominal and thoracic neuroblastomas. Pediatr Surg Int 2024; 40:286. [PMID: 39487870 DOI: 10.1007/s00383-024-05884-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/26/2024] [Indexed: 11/04/2024]
Abstract
AIM To investigate the safety and efficacy of the application of enhanced recovery after surgery (ERAS) protocols in the perioperative period of abdominal and thoracic localized neuroblastomas (NBs). METHODS In this retrospective study, 68 children with NBs who underwent surgical resection of the tumor were enrolled. The ERAS protocols for NB excision were implemented in the ERAS group (n = 39) and the consequences were compared with children treated with traditional care (n = 29, TRAD group). The main outcomes of our interest included the incidence of surgery-related complications, the postoperative length of stay (LOS), and the Face/Legs/Activity/Cry/Consolability (FLACC) quantitative table from postoperative days (POD) 1-5. We also evaluated the median intraoperative fluid volume and anesthesia recovery time; blood glucose levels at the beginning of anesthesia, POD1, and 3; WBC counts, CRP values, and the concentration of plasma nutritional indicators on POD1 and 3; time of early ambulation, first anal exhaust, total enteral nutrition (TEN), and discontinue intravenous infusion postoperatively; usage proportion and duration of abdominal and thoracic drainages, nasogastric decompression tubes and urinary catheters; cost of hospitalization, parental satisfaction rate, and readmission rate of surgery ward within 30 days. RESULTS Compared to the TRAD group, the ERAS group had lower surgery-related complications, albeit not significantly (P > 0.05); the median postoperative LOS decreased from 11.0 to 8.0 days (P < 0.001), the LOS of abdominal NB was significantly shortened (P < 0.001) compared to thoracic NB (P = 0.07) between the two groups; the FLACC scores decreased significantly from POD1-5 (all P < 0.01). The ERAS group had an improved median intraoperative infusion speed (5.0 mL/kg/h vs 8.0 mL/kg/h), time of early ambulation (1.0 days vs 3.0 days), first anal exhaust (2.0 days vs 2.0 days), TEN (5.0 vs 7.0 days), discontinuation of intravenous infusion (5.0 days vs 8.0 days), and total cost of hospitalization (33,897.2 Yuan vs 38,876.3 Yuan); (all P < 0.01). The usage proportion and duration of surgical drainages and tubes were apparently reduced. The mean blood glucose level was higher at the beginning of anesthesia but lower on POD1 and 3 in the ERAS group (P < 0.01). No statistically significant difference was detected in WBC counts and concentrations of hemoglobin and albumin between the two groups of patients (P > 0.05), while the concentrations of prealbumin on POD3 were higher and the CRP level on POD1 was lower in the ERAS group than the TRAD group (P < 0.01). The satisfaction rate of parents was only slightly higher, but the difference was not statistically significant (P = 0.730). No obvious differences were observed in the aspects of NB resection (P = 0.462) and 30-day readmissions of surgery ward (P = 1.000). CONCLUSION The application of ERAS protocols has a significant potential to accelerate perioperative rehabilitation in children undergoing abdominal and thoracic NBs' surgical resection.
Collapse
Affiliation(s)
- Jingjing He
- Reproductive Medicine Center, Hefei Maternal and Child Health Hospital, Hefei, 230001, Anhui, China
| | - Zhiru Wang
- Department of General Surgery, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Xiyang Yu
- Department of Pediatric Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Yilin Su
- Department of Pediatric Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Mingyun Hong
- Reproductive Medicine Center, Hefei Maternal and Child Health Hospital, Hefei, 230001, Anhui, China.
| | - Kai Zhu
- Department of Pediatric Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| |
Collapse
|
2
|
Tirelli M, Bonfiglio F, Cantalupo S, Montella A, Avitabile M, Maiorino T, Diskin SJ, Iolascon A, Capasso M. Integrative genomic analyses identify neuroblastoma risk genes involved in neuronal differentiation. Hum Genet 2024; 143:1293-1309. [PMID: 39192051 PMCID: PMC11522082 DOI: 10.1007/s00439-024-02700-2] [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: 06/12/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024]
Abstract
Genome-Wide Association Studies (GWAS) have been decisive in elucidating the genetic predisposition of neuroblastoma (NB). The majority of genetic variants identified in GWAS are found in non-coding regions, suggesting that they can be causative of pathogenic dysregulations of gene expression. Nonetheless, pinpointing the potential causal genes within implicated genetic loci remains a major challenge. In this study, we integrated NB GWAS and expression Quantitative Trait Loci (eQTL) data from adrenal gland to identify candidate genes impacting NB susceptibility. We found that ZMYM1, CBL, GSKIP and WDR81 expression was dysregulated by NB predisposing variants. We further investigated the functional role of the identified genes through computational analysis of RNA sequencing (RNA-seq) data from single-cell and whole-tissue samples of NB, neural crest, and adrenal gland tissues, as well as through in vitro differentiation assays in NB cell cultures. Our results indicate that dysregulation of ZMYM1, CBL, GSKIP, WDR81 may lead to malignant transformation by affecting early and late stages of normal program of neuronal differentiation. Our findings enhance the understanding of how specific genes contribute to NB pathogenesis by highlighting their influence on neuronal differentiation and emphasizing the impact of genetic risk variants on the regulation of genes involved in critical biological processes.
Collapse
Affiliation(s)
- Matilde Tirelli
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131, Naples, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80145, Naples, Italy
| | - Ferdinando Bonfiglio
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131, Naples, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80145, Naples, Italy
| | - Sueva Cantalupo
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80145, Naples, Italy
| | - Annalaura Montella
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131, Naples, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80145, Naples, Italy
| | | | - Teresa Maiorino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131, Naples, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80145, Naples, Italy
| | - Sharon J Diskin
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, 19104, Philadelphia, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, 19104, Philadelphia, USA
| | - Achille Iolascon
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131, Naples, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80145, Naples, Italy
| | - Mario Capasso
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131, Naples, Italy.
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80145, Naples, Italy.
| |
Collapse
|
3
|
An H, Hong Y, Goh YT, Koh CWQ, Kanwal S, Zhang Y, Lu Z, Yap PML, Neo SP, Wong CM, Wong AST, Yu Y, Ho JSY, Gunaratne J, Goh WSS. m 6Am sequesters PCF11 to suppress premature termination and drive neuroblastoma differentiation. Mol Cell 2024:S1097-2765(24)00827-X. [PMID: 39481383 DOI: 10.1016/j.molcel.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 08/08/2024] [Accepted: 10/01/2024] [Indexed: 11/02/2024]
Abstract
N6,2'-O-dimethyladenosine (m6Am) is an abundant mRNA modification that impacts multiple diseases, but its function remains controversial because the m6Am reader is unknown. Using quantitative proteomics, we identified transcriptional terminator premature cleavage factor II (PCF11) as a m6Am-specific reader in human cells. Direct quantification of mature versus nascent RNAs reveals that m6Am does not regulate mRNA stability but promotes nascent transcription. Mechanistically, m6Am functions by sequestering PCF11 away from proximal RNA polymerase II (RNA Pol II). This suppresses PCF11 from dissociating RNA Pol II near transcription start sites, thereby promoting full-length transcription of m6Am-modified RNAs. m6Am's unique relationship with PCF11 means m6Am function is enhanced when PCF11 is reduced, which occurs during all-trans-retinoic-acid (ATRA)-induced neuroblastoma-differentiation therapy. Here, m6Am promotes expression of ATF3, which represses neuroblastoma biomarker MYCN. Depleting m6Am suppresses MYCN repression in ATRA-treated neuroblastoma and maintains their tumor-stem-like properties. Collectively, we characterize m6Am as an anti-terminator RNA modification that suppresses premature termination and modulates neuroblastoma's therapeutic response.
Collapse
Affiliation(s)
- Huihui An
- Shenzhen Bay Laboratory, Shenzhen, China; School of Biological Sciences, University of Hong Kong, Hong Kong, China; Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Yifan Hong
- Shenzhen Bay Laboratory, Shenzhen, China
| | | | | | | | - Yi Zhang
- Shenzhen Bay Laboratory, Shenzhen, China
| | - Zhaoqi Lu
- Shenzhen Bay Laboratory, Shenzhen, China
| | | | - Suat Peng Neo
- Institute of Molecular and Cell Biology, Singapore, Singapore
| | - Chun-Ming Wong
- Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Alice S T Wong
- School of Biological Sciences, University of Hong Kong, Hong Kong, China
| | - Yang Yu
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Jessica Sook Yuin Ho
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | | | | |
Collapse
|
4
|
Tempora P, D'Amico S, Gragera P, Damiani V, Krol K, Scaldaferri V, Pandey K, Chung S, Lucarini V, Giorda E, Scarsella M, Volpe G, Pezzullo M, De Stefanis C, D'Oria V, De Angelis L, Giovannoni R, De Ioris MA, Melaiu O, Purcell AW, Locatelli F, Fruci D. Combining ERAP1 silencing and entinostat therapy to overcome resistance to cancer immunotherapy in neuroblastoma. J Exp Clin Cancer Res 2024; 43:292. [PMID: 39438988 PMCID: PMC11494811 DOI: 10.1186/s13046-024-03180-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 09/04/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Checkpoint immunotherapy unleashes tumor control by T cells, but it is undermined in non-immunogenic tumors, e.g. with low MHC class I expression and low neoantigen burden, such as neuroblastoma (NB). Endoplasmic reticulum aminopeptidase 1 (ERAP1) is an enzyme that trims peptides before loading on MHC class I molecules. Inhibition of ERAP1 results in the generation of new antigens able of inducing potent anti-tumor immune responses. Here, we identify a novel non-toxic combinatorial strategy based on genetic inhibition of ERAP1 and administration of the HDAC inhibitor (HDACi) entinostat that increase the immunogenicity of NB, making it responsive to PD-1 therapy. METHODS CRISPR/Cas9-mediated gene editing was used to knockout (KO) the ERAP1 gene in 9464D NB cells derived from spontaneous tumors of TH-MYCN transgenic mice. The expression of MHC class I and PD-L1 was evaluated by flow cytometry (FC). The immunopeptidome of these cells was studied by mass spectrometry. Cocultures of splenocytes derived from 9464D bearing mice and tumor cells allowed the assessment of the effect of ERAP1 inhibition on the secretion of inflammatory cytokines and activation and migration of immune cells towards ERAP1 KO cells by FC. Tumor cell killing was evaluated by Caspase 3/7 assay and flow cytometry analysis. The effect of ERAP1 inhibition on the immune content of tumors was analyzed by FC, immunohistochemistry and multiple immunofluorescence. RESULTS We found that inhibition of ERAP1 makes 9464D cells more susceptible to immune cell-mediated killing by increasing both the recall and activation of CD4+ and CD8+ T cells and NK cells. Treatment with entinostat induces the expression of MHC class I and PD-L1 molecules in 9464D both in vitro and in vivo. This results in pronounced changes in the immunopeptidome induced by ERAP1 inhibition, but also restrains the growth of ERAP1 KO tumors in vivo by remodelling the tumor-infiltrating T-cell compartment. Interestingly, the absence of ERAP1 in combination with entinostat and PD-1 blockade overcomes resistance to PD-1 immunotherapy and increases host survival. CONCLUSIONS These findings demonstrate that ERAP1 inhibition combined with HDACi entinostat treatment and PD-1 blockade remodels the immune landscape of a non-immunogenic tumor such as NB, making it responsive to checkpoint immunotherapy.
Collapse
Affiliation(s)
| | | | - Paula Gragera
- Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Kamila Krol
- Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Kirti Pandey
- Department of Biochemistry and Molecular Biology, Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
| | - Shanzou Chung
- Department of Biochemistry and Molecular Biology, Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
| | | | - Ezio Giorda
- Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | | | | | | | | | | | | | | | - Ombretta Melaiu
- Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Anthony W Purcell
- Department of Biochemistry and Molecular Biology, Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
| | - Franco Locatelli
- Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Department of Life Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Doriana Fruci
- Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
| |
Collapse
|
5
|
Bonine N, Zanzani V, Van Hemelryk A, Vanneste B, Zwicker C, Thoné T, Roelandt S, Bekaert SL, Koster J, Janoueix-Lerosey I, Thirant C, Van Haver S, Roberts SS, Mus LM, De Wilde B, Van Roy N, Everaert C, Speleman F, Vermeirssen V, Scott CL, De Preter K. NBAtlas: A harmonized single-cell transcriptomic reference atlas of human neuroblastoma tumors. Cell Rep 2024; 43:114804. [PMID: 39368085 DOI: 10.1016/j.celrep.2024.114804] [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: 02/01/2024] [Revised: 06/11/2024] [Accepted: 09/12/2024] [Indexed: 10/07/2024] Open
Abstract
Neuroblastoma, a rare embryonic tumor arising from neural crest development, is responsible for 15% of pediatric cancer-related deaths. Recently, several single-cell transcriptome studies were performed on neuroblastoma patient samples to investigate the cell of origin and tumor heterogeneity. However, these individual studies involved a small number of tumors and cells, limiting the conclusions that could be drawn. To overcome this limitation, we integrated seven single-cell or single-nucleus datasets into a harmonized cell atlas covering 362,991 cells across 61 patients. We use this atlas to decipher the transcriptional landscape of neuroblastoma at single-cell resolution, revealing associations between transcriptomic profiles and clinical outcomes within the tumor compartment. In addition, we characterize the complex immune-cell landscape and uncover considerable heterogeneity among tumor-associated macrophages. Finally, we showcase the utility of our atlas as a resource by expanding it with additional data and using it as a reference for data-driven cell-type annotation.
Collapse
Affiliation(s)
- Noah Bonine
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB-UGent Center for Medical Biotechnology, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Vittorio Zanzani
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB-UGent Center for Medical Biotechnology, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; Laboratory for Computational Biology, Integromics and Gene Regulation (CBIGR), Ghent University, Ghent, Belgium
| | - Annelies Van Hemelryk
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; Laboratory of Myeloid Cell Biology in Tissue Damage and Inflammation, VIB-UGent Center for Inflammation Research, Technologiepark-Zwijnaarde 71, 9052 Ghent, Belgium
| | - Bavo Vanneste
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; Laboratory of Myeloid Cell Biology in Tissue Damage and Inflammation, VIB-UGent Center for Inflammation Research, Technologiepark-Zwijnaarde 71, 9052 Ghent, Belgium
| | - Christian Zwicker
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; Laboratory of Myeloid Cell Biology in Tissue Damage and Inflammation, VIB-UGent Center for Inflammation Research, Technologiepark-Zwijnaarde 71, 9052 Ghent, Belgium
| | - Tinne Thoné
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; Laboratory of Myeloid Cell Biology in Tissue Damage and Inflammation, VIB-UGent Center for Inflammation Research, Technologiepark-Zwijnaarde 71, 9052 Ghent, Belgium
| | - Sofie Roelandt
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB-UGent Center for Medical Biotechnology, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Sarah-Lee Bekaert
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Jan Koster
- Amsterdam UMC Location University of Amsterdam, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Isabelle Janoueix-Lerosey
- Inserm U830, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, Institut Curie Research Center, Paris, France
| | - Cécile Thirant
- Inserm U830, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, Institut Curie Research Center, Paris, France
| | - Stéphane Van Haver
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium; Tow Center for Developmental Oncology, Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Stephen S Roberts
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| | - Liselot M Mus
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Bram De Wilde
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Nadine Van Roy
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Celine Everaert
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB-UGent Center for Medical Biotechnology, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Frank Speleman
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Vanessa Vermeirssen
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; Laboratory for Computational Biology, Integromics and Gene Regulation (CBIGR), Ghent University, Ghent, Belgium
| | - Charlotte L Scott
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; Laboratory of Myeloid Cell Biology in Tissue Damage and Inflammation, VIB-UGent Center for Inflammation Research, Technologiepark-Zwijnaarde 71, 9052 Ghent, Belgium.
| | - Katleen De Preter
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB-UGent Center for Medical Biotechnology, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| |
Collapse
|
6
|
Bento CA, Arnaud-Sampaio VF, Glaser T, Adinolfi E, Coutinho-Silva R, Ulrich H, Lameu C. P2X7 receptor in macrophage polarization and its implications in neuroblastoma tumor behavior. Purinergic Signal 2024:10.1007/s11302-024-10051-w. [PMID: 39425818 DOI: 10.1007/s11302-024-10051-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 09/15/2024] [Indexed: 10/21/2024] Open
Abstract
Tumor-associated macrophages (TAMs) exhibit antitumor or protumor responses related to inflammatory (or M1) and alternative (or M2) phenotypes, respectively. The P2X7 receptor plays a key role in macrophage polarization, influencing inflammation and immunosuppression. In this study, we investigated the role of the P2X7 receptor in TAMs. Using P2X7 receptor-deficient macrophages, we analyzed gene expression profiles and their implications for neuroblastoma invasion and chemoresistance. Our results showed that P2X7 receptor deficiency altered the expression of classical polarization markers, such as nitric oxide synthase 2 (Nos2) and tumor necrosis factor-α (Tnf), as well as alternative phenotype markers, including mannose receptor C-type 1 (Mrc1) and arginase 1 (Arg1). P2X7 deficiency also influenced the expression of the ectonucleotidases Entpd1 and Nt5e and other purinergic receptors, especially P2ry2, suggesting compensatory mechanisms involved in macrophage polarization. In particular, TAMs deficient in P2X7 showed a phenotype with characteristics intermideiate between resting macrophages (M0) and M1 polarization rather than the M2-type phenotype like and wild-type TAM macrophages. In addition, P2rx7-/- TAMs regulated the expression of P2X7 receptor isoforms in neuroblastoma cells, with downregulation of the P2X7 A and B isoforms leading to a decrease in chemotherapy-induced cell death. However, TAMs expressing P2X7 downregulated only the B isoform, suggesting that TAMs play a role in modulating tumor behavior through P2X7 receptor isoform regulation. Taken together, our data underscore the regulatory function of the P2X7 receptor in orchestrating alternative macrophage polarization and in the interplay between tumor cells and TAMs. These findings help to clarify the complex interplay of purinergic signaling in cancer progression and open up avenues for future research and therapeutic interventions.
Collapse
Affiliation(s)
- Carolina Adriane Bento
- Metastasis Molecular Mechanisms Laboratory and Neurosciences Laboratory, Institute of Chemistry, Biochemistry Department, University of Sao Paulo, Sao Paulo, Brazil
| | - Vanessa Fernandes Arnaud-Sampaio
- Metastasis Molecular Mechanisms Laboratory and Neurosciences Laboratory, Institute of Chemistry, Biochemistry Department, University of Sao Paulo, Sao Paulo, Brazil
| | - Talita Glaser
- Metastasis Molecular Mechanisms Laboratory and Neurosciences Laboratory, Institute of Chemistry, Biochemistry Department, University of Sao Paulo, Sao Paulo, Brazil
| | - Elena Adinolfi
- Section of General Pathology, Department of Experimental and Diagnostic Medicine, University of Ferrara, Ferrara, Italy
| | - Robson Coutinho-Silva
- Laboratory of Immunophysiology Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Henning Ulrich
- Metastasis Molecular Mechanisms Laboratory and Neurosciences Laboratory, Institute of Chemistry, Biochemistry Department, University of Sao Paulo, Sao Paulo, Brazil
| | - Claudiana Lameu
- Metastasis Molecular Mechanisms Laboratory and Neurosciences Laboratory, Institute of Chemistry, Biochemistry Department, University of Sao Paulo, Sao Paulo, Brazil.
| |
Collapse
|
7
|
Wei Z, Gong B, Li X, Chen C, Zhao Q. Event-free survival in neuroblastoma with MYCN amplification and deletion of 1p or 11q may be associated with altered immune status. BMC Cancer 2024; 24:1279. [PMID: 39407175 PMCID: PMC11481459 DOI: 10.1186/s12885-024-13044-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 10/08/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Neuroblastoma exhibits substantial heterogeneity, which is intricately linked to various genetic alterations. We aimed to explore immune status in the peripheral blood and prognosis of patients with neuroblastoma with different genetic characteristics. METHODS We enrolled 31 patients with neuroblastoma and collected samples to detect three genetic characteristics. Peripheral blood samples were tested for immune cells and cytokines by fluorescent microspheres conjugated with antibodies and flow cytometry. Event-free survival (EFS) was analyzed using the Kaplan‒Meier method. RESULTS Twenty-two patients had genetic aberrations, including MYCN amplification in 6 patients, chromosome 1p deletion in 9 patients, and chromosome 11q deletion in 14 patients. Two genetic alterations were present in seven patients. The EFS was worse in patients with MYCN amplification or 1p deletion than in the corresponding group, whereas 11q deletion was a prognostic factor only in patients with unamplified MYCN. Changes in immune status revealed a decrease in the proportion of T cells in blood, and an increase in regulatory T cells and immunosuppression-related cytokines such as interleukin (IL)-10. The EFS of the IL-10 high-level group was lower than that of the low-level group. Patients with concomitant genetic alterations and a high level of IL-10 had worse EFS than other patients. CONCLUSIONS Patients with neuroblastoma characterized by these genetic characteristics often have suppressed T cell response and an overabundance of immunosuppressive cells and cytokines in the peripheral blood. This imbalance is significantly associated with poor EFS. Moreover, if these patients show an elevated levels of immunosuppressive cytokines such as IL-10, the prognosis will be worse.
Collapse
Affiliation(s)
- Zixuan Wei
- Department of Pediatric Oncology, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, West Huan-Hu Rd, Ti Yuan Bei, Hexi District, 300060, Tianjin, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Baocheng Gong
- Department of Pediatric Oncology, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, West Huan-Hu Rd, Ti Yuan Bei, Hexi District, 300060, Tianjin, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Xin Li
- Tianjin Cancer Hospital Airport Hospital, Tianjin, China
| | - Chong Chen
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- National Human Genetic Resources Sharing Service Platform, Tianjin, China
| | - Qiang Zhao
- Department of Pediatric Oncology, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, West Huan-Hu Rd, Ti Yuan Bei, Hexi District, 300060, Tianjin, Tianjin, China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
- Tianjin's Clinical Research Center for Cancer, Tianjin, China.
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.
| |
Collapse
|
8
|
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
|
9
|
Maher S, Wynne K, Zhernovkov V, Halasz M. A temporal (phospho-)proteomic dataset of neurotrophic receptor tyrosine kinase signalling in neuroblastoma. Sci Data 2024; 11:1111. [PMID: 39389992 PMCID: PMC11467210 DOI: 10.1038/s41597-024-03965-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 10/02/2024] [Indexed: 10/12/2024] Open
Abstract
Neurotrophic receptor tyrosine kinases (TrkA, TrkB, TrkC), despite their homology, contribute to the clinical heterogeneity of the childhood cancer neuroblastoma. TrkA expression is associated with low-stage disease and is often seen with spontaneous tumour regression. Conversely, TrkB is present in unfavourable neuroblastomas that often harbour amplification of the MYCN oncogene. The role of TrkC is less clearly defined, although some studies suggest its association with a favourable outcome. Understanding the differences in activity of Trk receptors that drive divergent clinical phenotypes as well as the influence of MYCN amplification on downstream Trk receptor signalling remains poorly understood. Here, we present a comprehensive label-free mass spectrometry-based total proteomics and phosphoproteomics dataset (432 raw files with FragPipe search outputs; available on PRIDE with accession number PXD054441) where we identified and quantified 4,907 proteins, 16,744 phosphosites and 5,084 phosphoproteins, derived from NGF/BDNF/NT-3 treated TrkA/B/C-overexpressing neuroblastoma cells with differential MYCN status. Analysing our dataset offers valuable insights into TrkA/B/C receptor signalling in neuroblastoma and its modulation by MYCN status; and holds potential for advancing therapeutic strategies in this challenging childhood cancer.
Collapse
Affiliation(s)
- Stephanie Maher
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin, Ireland
| | - Kieran Wynne
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Vadim Zhernovkov
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin, Ireland
| | - Melinda Halasz
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin, Ireland.
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland.
| |
Collapse
|
10
|
Chilamakuri R, Agarwal S. Repurposing of c-MET Inhibitor Tivantinib Inhibits Pediatric Neuroblastoma Cellular Growth. Pharmaceuticals (Basel) 2024; 17:1350. [PMID: 39458991 PMCID: PMC11510580 DOI: 10.3390/ph17101350] [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: 08/28/2024] [Revised: 09/22/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Dysregulation of receptor tyrosine kinase c-MET is known to promote tumor development by stimulating oncogenic signaling pathways in different cancers, including pediatric neuroblastoma (NB). NB is an extracranial solid pediatric cancer that accounts for almost 15% of all pediatric cancer-related deaths, with less than a 50% long-term survival rate. Results: In this study, we analyzed a large cohort of primary NB patient data and revealed that high MET expression strongly correlates with poor overall survival, disease progression, relapse, and high MYCN levels in NB patients. To determine the effects of c-MET in NB, we repurposed a small molecule inhibitor, tivantinib, and found that c-MET inhibition significantly inhibits NB cellular growth. Tivantinib significantly blocks NB cell proliferation and 3D spheroid tumor formation and growth in different MYCN-amplified and MYCN-non-amplified NB cell lines. Furthermore, tivantinib blocks the cell cycle at the G2/M phase transition and induces apoptosis in different NB cell lines. As expected, c-MET inhibition by tivantinib inhibits the expression of multiple genes in PI3K, STAT, and Ras cell signaling pathways. Conclusions: Overall, our data indicate that c-MET directly regulates NB growth and 3D spheroid growth, and c-MET inhibition by tivantinib may be an effective therapeutic approach for high-risk NB. Further developing c-MET targeted therapeutic approaches and combining them with current therapies may pave the way for effectively translating novel therapies for NB and other c-MET-driven cancers.
Collapse
Affiliation(s)
| | - Saurabh Agarwal
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, New York, NY 11439, USA
| |
Collapse
|
11
|
Chen Y, Zhuo R, Sun L, Tao Y, Li G, Zhu F, Xu Y, Wang J, Li Z, Yu J, Yin H, Wu D, Li X, Fang F, Xie Y, Hu Y, Wang H, Yang C, Shi L, Wang X, Zhang Z, Pan J. Super-enhancer-driven IRF2BP2 enhances ALK activity and promotes neuroblastoma cell proliferation. Neuro Oncol 2024; 26:1878-1894. [PMID: 38864832 PMCID: PMC11449008 DOI: 10.1093/neuonc/noae109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND Super-enhancers (SEs) typically govern the expression of critical oncogenes and play a fundamental role in the initiation and progression of cancer. Focusing on genes that are abnormally regulated by SE in cancer may be a new strategy for understanding pathogenesis. In the context of this investigation, we have identified a previously unreported SE-driven gene IRF2BP2 in neuroblastoma (NB). METHODS The expression and prognostic value of IRF2BP2 were detected in public databases and clinical samples. The effect of IRF2BP2 on NB cell growth and apoptosis was evaluated through in vivo and in vitro functional loss experiments. The molecular mechanism of IRF2BP2 was investigated by the study of chromatin regulatory regions and transcriptome sequencing. RESULTS The sustained high expression of IRF2BP2 results from the activation of a novel SE established by NB master transcription factors MYCN, MEIS2, and HAND2, and they form a new complex that regulates the gene network associated with the proliferation of NB cell populations. We also observed a significant enrichment of the AP-1 family at the binding sites of IRF2BP2. Remarkably, within NB cells, AP-1 plays a pivotal role in shaping the chromatin accessibility landscape, thereby exposing the binding site for IRF2BP2. This orchestrated action enables AP-1 and IRF2BP2 to collaboratively stimulate the expression of the NB susceptibility gene ALK, thereby upholding the highly proliferative phenotype characteristic of NB. CONCLUSIONS Our findings indicate that SE-driven IRF2BP2 can bind to AP-1 to maintain the survival of tumor cells via regulating chromatin accessibility of the NB susceptibility gene ALK.
Collapse
Affiliation(s)
- Yanling Chen
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Ran Zhuo
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Lichao Sun
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China
| | - Yanfang Tao
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Gen Li
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Frank Zhu
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Yunyun Xu
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Jianwei Wang
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Zhiheng Li
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Juanjuan Yu
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Hongli Yin
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Di Wu
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Xiaolu Li
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Fang Fang
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Yi Xie
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Yizhou Hu
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Hairong Wang
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Chun Yang
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Lei Shi
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaodong Wang
- Department of Orthopedics, Children’s Hospital of Soochow University, Suzhou, China
| | - Zimu Zhang
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Jian Pan
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| |
Collapse
|
12
|
Westerveld ASR, Tytgat GAM, van Santen HM, van Noesel MM, Loonen J, de Vries ACH, Louwerens M, Koopman MMW, van der Heiden-van der Loo M, Janssens GO, de Krijger RR, Ronckers CM, van der Pal HJH, Kremer LCM, Teepen JC. Long-Term Risk of Subsequent Neoplasms in 5-Year Survivors of Childhood Neuroblastoma: A Dutch Childhood Cancer Survivor Study-LATER 3 Study. J Clin Oncol 2024:JCO2301430. [PMID: 39356982 DOI: 10.1200/jco.23.01430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 05/16/2024] [Accepted: 08/15/2024] [Indexed: 10/04/2024] Open
Abstract
PURPOSE Neuroblastoma survivors have an increased risk of developing subsequent malignant neoplasms (SMNs), but the risk of subsequent nonmalignant neoplasms (SNMNs) and risk factors are largely unknown. We analyzed the long-term risks and associated risk factors for developing SMNs and SNMNs in a well-characterized cohort of 5-year neuroblastoma survivors. METHODS We included 563 5-year neuroblastoma survivors from the Dutch Childhood Cancer Survivor Study (DCCSS)-LATER cohort, diagnosed during 1963-2014. Subsequent neoplasms were ascertained by linkages with the Netherlands Cancer Registry and the Dutch Nationwide Pathology Databank (Palga) and medical chart review. We calculated standardized incidence ratios (SIRs), absolute excess risk (AER), and cumulative incidences. Multivariable competing risk regression analysis was used to evaluate risk factors. RESULTS In total, 23 survivors developed an SMN and 60 an SNMN. After a median follow-up of 23.7 (range, 5.0-56.3) years, the risk of SMN was elevated compared with the general population (SIR, 4.0; 95% CI, 2.5 to 5.9; AER per 10,000 person-years, 15.1). The 30-year cumulative incidence was 3.4% (95% CI, 1.9 to 6.0) for SMNs and 10.4% (95% CI, 7.3 to 14.8) for SNMNs. Six survivors developed an SMN after iodine-metaiodobenzylguanidine (131IMIBG) treatment. Survivors treated with 131IMIBG had a higher risk of developing SMNs (subdistribution hazard ratio [SHR], 5.7; 95% CI, 1.8 to 17.8) and SNMNs (SHR, 2.6; 95% CI, 1.2 to 5.6) compared with survivors treated without 131IMIBG; results for SMNs were attenuated in high-risk patients only (SMNs SHR, 3.6; 95% CI, 0.9 to 15.3; SNMNs SHR, 1.5; 95% CI, 0.7 to 3.6). CONCLUSION Our results demonstrate that neuroblastoma survivors have an elevated risk of developing SMNs and a high risk of SNMNs. 131IMIBG may be a treatment-related risk factor for the development of SMN and SNMN, which needs further validation. Our results emphasize the need for awareness of subsequent neoplasms and the importance of follow-up care.
Collapse
Affiliation(s)
| | | | - Hanneke M van Santen
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Department of Pediatric Endocrinology, University Medical Center Utrecht, Wilhelmina Children's Hospital, Utrecht, the Netherlands
| | - Max M van Noesel
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Department of Imaging & Cancer, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jacqueline Loonen
- Department of Hematology, Radboudumc Center of Expertise for Cancer Survivorship, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Andrica C H de Vries
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Netherlands Department of Pediatric Oncology/Hematology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Marloes Louwerens
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Maria M W Koopman
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | | | - Geert O Janssens
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Ronald R de Krijger
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Cecile M Ronckers
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Division of Childhood Cancer Epidemiology, Institute of Medical Biostatistics Informatics and Epidemiology, University Medical Center of the JGU, Mainz, Germany
| | | | - Leontien C M Kremer
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- University Medical Center Utrecht, Wilhelmina Children's Hospital, Utrecht, the Netherlands
- Department of Pediatrics, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Jop C Teepen
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| |
Collapse
|
13
|
Chen X, Chen Q, Liu Y, Qiu Y, Lv L, Zhang Z, Yin X, Shu F. Radiomics models to predict bone marrow metastasis of neuroblastoma using CT. CANCER INNOVATION 2024; 3:e135. [PMID: 38948899 PMCID: PMC11212276 DOI: 10.1002/cai2.135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 02/01/2024] [Accepted: 02/02/2024] [Indexed: 07/02/2024]
Abstract
Background Bone marrow is the leading site for metastasis from neuroblastoma and affects the prognosis of patients with neuroblastoma. However, the accurate diagnosis of bone marrow metastasis is limited by the high spatial and temporal heterogeneity of neuroblastoma. Radiomics analysis has been applied in various cancers to build accurate diagnostic models but has not yet been applied to bone marrow metastasis of neuroblastoma. Methods We retrospectively collected information from 187 patients pathologically diagnosed with neuroblastoma and divided them into training and validation sets in a ratio of 7:3. A total of 2632 radiomics features were retrieved from venous and arterial phases of contrast-enhanced computed tomography (CT), and nine machine learning approaches were used to build radiomics models, including multilayer perceptron (MLP), extreme gradient boosting, and random forest. We also constructed radiomics-clinical models that combined radiomics features with clinical predictors such as age, gender, ascites, and lymph gland metastasis. The performance of the models was evaluated with receiver operating characteristics (ROC) curves, calibration curves, and risk decile plots. Results The MLP radiomics model yielded an area under the ROC curve (AUC) of 0.97 (95% confidence interval [CI]: 0.95-0.99) on the training set and 0.90 (95% CI: 0.82-0.95) on the validation set. The radiomics-clinical model using an MLP yielded an AUC of 0.93 (95% CI: 0.89-0.96) on the training set and 0.91 (95% CI: 0.85-0.97) on the validation set. Conclusions MLP-based radiomics and radiomics-clinical models can precisely predict bone marrow metastasis in patients with neuroblastoma.
Collapse
Affiliation(s)
- Xiong Chen
- Department of Paediatric Urology, Guangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhouChina
- Department of Paediatric Surgery, Guangzhou Institute of PaediatricsGuangzhou Medical UniversityGuangzhouChina
| | - Qinchang Chen
- Department of Pediatric Cardiology, Guangdong Provincial People's HospitalGuangdong Academy of Medical Sciences, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Structural Heart DiseaseGuangzhouChina
| | - Yuanfang Liu
- Department of Radiology, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Ya Qiu
- Department of Radiologythe First People's Hospital of Kashi PrefectureKashiChina
| | - Lin Lv
- Medical SchoolSun Yat‐sen UniversityGuangzhouChina
| | - Zhengtao Zhang
- Department of Paediatric Urology, Guangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhouChina
- Department of Paediatric Surgery, Guangzhou Institute of PaediatricsGuangzhou Medical UniversityGuangzhouChina
| | - Xuntao Yin
- Department of RadiologyGuangzhou Women and Children's Medical CenterGuangzhouChina
| | - Fangpeng Shu
- Department of Paediatric Urology, Guangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhouChina
- Department of Paediatric Surgery, Guangzhou Institute of PaediatricsGuangzhou Medical UniversityGuangzhouChina
| |
Collapse
|
14
|
Altun Z, Ceyhan M, Yuan H, Kızmazoğlu D, Aktaş S, Olgun N. Low Expression of CASP8 Could be a Prognostic Biomarker in Neuroblastoma Patients. J Child Neurol 2024; 39:386-394. [PMID: 39234689 DOI: 10.1177/08830738241273431] [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] [Indexed: 09/06/2024]
Abstract
The aim of study was to investigate whether CASP8 (CASPASE8) could be a biomarker for prognosis in neuroblastoma. The prognostic value of CASP8 was determined by analyzing CASP8 methylation status and gene expressions in the tumor tissues of 37 neuroblastoma patients. Bisulfite and quantitative multiplex-methylation-specific polymerase chain reaction (PCR) were used to identify the methylation status. CASP8 messenger ribonucleic acid (RNA) expression levels were determined using reverse transcriptase-quantitative PCR. CASP8 expression levels associated with prognostic value were also analyzed using the TARGET NBL (141 cases) database through PDX for Childhood Cancer Therapeutics (PCAT) and SEQC (498 cases) via the R2 platform. CASP8 methylation status was associated with risk groups, MYCN amplification, and 17q gain status. CASP8 expression was found to be statistically different between high- and low-risk neuroblastoma groups. Low expression of CASP8 was associated with MYCN amplification status. Low expression of CASP8 has shown statistically significant prognostic value through TARGET NBL and SEQC-498 data sets. CASP8 messenger RNA expressions and methylation status were associated with the MYCN amplified high-risk group in neuroblastoma. CASP8 messenger RNA expressions may be considered as a clinical prognostic marker in neuroblastoma.
Collapse
Affiliation(s)
- Zekiye Altun
- Department of Basic Oncology, Dokuz Eylül University Institute of Oncology, Izmir, Turkey
| | - Metin Ceyhan
- Department of Basic Oncology, Dokuz Eylül University Institute of Oncology, Izmir, Turkey
| | - Hongling Yuan
- Department of Basic Oncology, Dokuz Eylül University Institute of Oncology, Izmir, Turkey
| | - Deniz Kızmazoğlu
- Department of Pediatric Oncology, Dokuz Eylül University Institute of Oncology, Izmir, Turkey
| | - Safiye Aktaş
- Department of Basic Oncology, Dokuz Eylül University Institute of Oncology, Izmir, Turkey
| | - Nur Olgun
- Department of Pediatric Oncology, Dokuz Eylül University Institute of Oncology, Izmir, Turkey
| |
Collapse
|
15
|
Espinoza AF, Bagatell R, McHugh K, Naranjo AH, Van Ryn C, Rojas Y, Lyons K, Paul Guillerman R, Kirby C, Brock P, Volchenboum S, Simon T, States L, Miller A, Krug B, Sarnacki S, Irtan S, Brisse HJ, Valteau-Couanet D, von Schweinitz D, Kammer B, Granata C, Pio L, Park JR, Nuchtern JG. A subset of image-defined risk factors predict completeness of resection in children with high-risk neuroblastoma: An international multicenter study. Pediatr Blood Cancer 2024; 71:e31218. [PMID: 39072986 PMCID: PMC11500268 DOI: 10.1002/pbc.31218] [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: 06/16/2024] [Accepted: 07/10/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND Image-defined risk factors (IDRFs) were promulgated for predicting the feasibility and safety of complete primary tumor resection in children with neuroblastoma (NB). There is limited understanding of the impact of individual IDRFs on resectability of the primary tumor or patient outcomes. A multicenter database of patients with high-risk NB was interrogated to answer this question. DESIGN/METHODS Patients with high-risk NB (age <20 years) were eligible if cross-sectional imaging was performed at least twice prior to resection. IDRFs and primary tumor measurements were recorded for each imaging study. Extent of resection was determined from operative reports. RESULTS There were 211 of 229 patients with IDRFs at diagnosis, and 171 patients with IDRFs present pre-surgery. A ≥90% resection was significantly more likely in the absence of tumor invading or encasing the porta hepatis, hepatoduodenal ligament, superior mesenteric artery (SMA), renal pedicles, abdominal aorta/inferior vena cava (IVC), iliac vessels, and/or diaphragm at diagnosis or an overlapping subset of IDRFs (except diaphragm) at pre-surgery. There were no significant differences in event-free survival (EFS) and overall survival (OS) when patients were stratified by the presence versus absence of any IDRF either at diagnosis or pre-surgery. CONCLUSION Two distinct but overlapping subsets of IDRFs present either at diagnosis or after induction chemotherapy significantly influence the probability of a complete resection in children with high-risk NB. The presence of IDRFs was not associated with significant differences in OS or EFS in this cohort.
Collapse
Affiliation(s)
| | | | - Kieran McHugh
- Great Ormond Street Hospital for Children, London, United Kingdom
| | - Arlene H Naranjo
- University of Florida Colleges of Medicine and Public Health & Health Professions, Children’s Oncology Group Statistics & Data Center, Gainesville, USA
| | - Collin Van Ryn
- University of Florida Colleges of Medicine and Public Health & Health Professions, Children’s Oncology Group Statistics & Data Center, Gainesville, USA
| | - Yesenia Rojas
- Texas Children’s Hospital/Baylor College of Medicine, Houston, USA
| | - Karen Lyons
- Texas Children’s Hospital/Baylor College of Medicine, Houston, USA
| | | | | | - Penelope Brock
- Great Ormond Street Hospital for Children, London, United Kingdom
| | | | - Thorsten Simon
- Department of Radiology, University of Cologne, Cologne, Germany
| | - Lisa States
- Children’s Hospital of Philadelphia, Philadelphia, USA
| | | | - Barbara Krug
- Department of Radiology, University of Cologne, Cologne, Germany
| | - Sabine Sarnacki
- Necker-Enfants Malades Hospital – APHP and Université de Paris Cité, Paris, France
| | - Sabine Irtan
- Necker-Enfants Malades Hospital – APHP and Université de Paris Cité, Paris, France
| | | | | | - Dietrich von Schweinitz
- Department of Radiology, LMU University Hospital, LMU, University of Munich, Munich, Germany
| | - Birgit Kammer
- Department of Radiology, LMU University Hospital, LMU, University of Munich, Munich, Germany
| | | | - Luca Pio
- Giannina Gaslini Children’s Hospital, Genoa, Italy
- St. Jude Children’s Research Hospital, Memphis, USA
| | | | - Jed G. Nuchtern
- Texas Children’s Hospital/Baylor College of Medicine, Houston, USA
| |
Collapse
|
16
|
Werr L, Bartenhagen C, Rosswog C, Cartolano M, Voegele C, Sexton-Oates A, Di Genova A, Ernst A, Kahlert Y, Hemstedt N, Höppner S, Mansuet Lupo A, Pelosi G, Brcic L, Papotti M, George J, Bosco G, Quaas A, Tang LH, Robzyk K, Kadota K, Roh MS, Fanaroff RE, Falcon CJ, Büttner R, Lantuejoul S, Rekhtman N, Rudin CM, Travis WD, Alcala N, Fernandez-Cuesta L, Foll M, Peifer M, Thomas RK, Fischer M. TERT Expression and Clinical Outcome in Pulmonary Carcinoids. J Clin Oncol 2024:JCO2302708. [PMID: 39348606 DOI: 10.1200/jco.23.02708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/30/2024] [Accepted: 07/16/2024] [Indexed: 10/02/2024] Open
Abstract
PURPOSE The clinical course of pulmonary carcinoids ranges from indolent to fatal disease, suggesting that specific molecular alterations drive progression toward the fully malignant state. A similar spectrum of clinical phenotypes occurs in pediatric neuroblastoma, in which activation of telomerase reverse transcriptase (TERT) is decisive in determining the course of disease. We therefore investigated whether TERT expression defines the clinical fate of patients with pulmonary carcinoid. METHODS TERT expression was examined by RNA sequencing in a test cohort and a validation cohort of pulmonary carcinoids (n = 88 and n = 105, respectively). A natural TERT expression cutoff was determined in the test cohort on the basis of the distribution of TERT expression, and its prognostic value was assessed by Kaplan-Meier survival estimates and multivariable analyses. Telomerase activity was validated by telomere repeat amplification protocol assay. RESULTS Similar to neuroblastoma, TERT expression exhibited a bimodal distribution in pulmonary carcinoids, separating tumors into TERT-high and TERT-low subgroups. A natural TERT cutoff discriminated unfavorable from favorable clinical courses with high accuracy both in the test cohort (5-year overall survival [OS], 0.547 ± 0.132 v 1.0; P < .001) and the validation cohort (5-year OS, 0.788 ± 0.063 v 0.913 ± 0.048; P < .001). In line with these findings, telomerase activity was largely absent in TERT-low tumors, whereas it was readily detectable in TERT-high carcinoids. In multivariable analysis considering TERT expression, histology (typical v atypical carcinoid), and stage (≤IIA v ≥IIB), high TERT expression was an independent prognostic marker for poor survival, with a hazard ratio of 5.243 (95% CI, 1.943 to 14.148; P = .001). CONCLUSION Our data demonstrate that high TERT expression defines clinically aggressive pulmonary carcinoids with fatal outcome, similar to neuroblastoma, indicating that activation of TERT may be a defining feature of lethal cancers.
Collapse
Affiliation(s)
- Lisa Werr
- Department of Experimental Pediatric Oncology, University Children's Hospital of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Medical Faculty, University of Cologne, Cologne, Germany
| | - Christoph Bartenhagen
- Department of Experimental Pediatric Oncology, University Children's Hospital of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Medical Faculty, University of Cologne, Cologne, Germany
| | - Carolina Rosswog
- Department of Experimental Pediatric Oncology, University Children's Hospital of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Medical Faculty, University of Cologne, Cologne, Germany
- Else Kröner Forschungskolleg Clonal Evolution in Cancer, University Hospital Cologne, Cologne, Germany
| | - Maria Cartolano
- Center for Molecular Medicine Cologne (CMMC), Medical Faculty, University of Cologne, Cologne, Germany
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Catherine Voegele
- Rare Cancers Genomics Team (RCG), Genomic Epidemiology Branch (GEM), International Agency for Research on Cancer/World Health Organisation (IARC/WHO), Lyon, France
| | - Alexandra Sexton-Oates
- Rare Cancers Genomics Team (RCG), Genomic Epidemiology Branch (GEM), International Agency for Research on Cancer/World Health Organisation (IARC/WHO), Lyon, France
| | - Alex Di Genova
- Rare Cancers Genomics Team (RCG), Genomic Epidemiology Branch (GEM), International Agency for Research on Cancer/World Health Organisation (IARC/WHO), Lyon, France
| | - Angela Ernst
- Institute of Medical Statistics and Computational Biology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Yvonne Kahlert
- Department of Experimental Pediatric Oncology, University Children's Hospital of Cologne, Cologne, Germany
| | - Nadine Hemstedt
- Department of Experimental Pediatric Oncology, University Children's Hospital of Cologne, Cologne, Germany
| | - Stefanie Höppner
- Department of Experimental Pediatric Oncology, University Children's Hospital of Cologne, Cologne, Germany
| | - Audrey Mansuet Lupo
- Department of Pathology, Assistance Publique-Hôpitaux de Paris, Cochin Hospital, Paris Cité University, Paris, France
| | - Giuseppe Pelosi
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Luka Brcic
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Mauro Papotti
- Department of Oncology, University of Turin, Torino, Italy
| | - Julie George
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Cologne, Cologne, Germany
| | - Graziella Bosco
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Alexander Quaas
- Institute of Pathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Laura H Tang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kenneth Robzyk
- Sloan Kettering Institue, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kyuichi Kadota
- Molecular Oncologic Pathology, Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Takamatsu, Japan
| | - Mee Sook Roh
- Department of Pathology, Dong-A University College of Medicine, Busan, South Korea
| | | | - Christina J Falcon
- Sloan Kettering Institue, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Reinhard Büttner
- Institute of Pathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Sylvie Lantuejoul
- Department of Biopathology, Centre de Lutte Contre le Cancer UNICANCER Léon Bérard, Grenoble Alpes University, Lyon, France
| | - Natasha Rekhtman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Charles M Rudin
- Thoracic Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - William D Travis
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Nicolas Alcala
- Rare Cancers Genomics Team (RCG), Genomic Epidemiology Branch (GEM), International Agency for Research on Cancer/World Health Organisation (IARC/WHO), Lyon, France
| | - Lynnette Fernandez-Cuesta
- Rare Cancers Genomics Team (RCG), Genomic Epidemiology Branch (GEM), International Agency for Research on Cancer/World Health Organisation (IARC/WHO), Lyon, France
| | - Matthieu Foll
- Rare Cancers Genomics Team (RCG), Genomic Epidemiology Branch (GEM), International Agency for Research on Cancer/World Health Organisation (IARC/WHO), Lyon, France
| | - Martin Peifer
- Center for Molecular Medicine Cologne (CMMC), Medical Faculty, University of Cologne, Cologne, Germany
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Roman K Thomas
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute of Pathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Matthias Fischer
- Department of Experimental Pediatric Oncology, University Children's Hospital of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Medical Faculty, University of Cologne, Cologne, Germany
| |
Collapse
|
17
|
Liu Y, Fleishman JS, Wang H, Huo L. Pharmacologically Targeting Ferroptosis and Cuproptosis in Neuroblastoma. Mol Neurobiol 2024:10.1007/s12035-024-04501-0. [PMID: 39331355 DOI: 10.1007/s12035-024-04501-0] [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: 02/07/2024] [Accepted: 09/12/2024] [Indexed: 09/28/2024]
Abstract
Neuroblastoma is a deadly pediatric cancer that originates from the neural crest and frequently develops in the abdomen or adrenal gland. Although multiple approaches, including chemotherapy, radiotherapy, targeted therapy, and immunotherapy, are recommended for treating neuroblastoma, the tumor will eventually develop resistance, leading to treatment failure and cancer relapse. Therefore, a firm understanding of the molecular mechanisms underlying therapeutic resistance is vital for the development of new effective therapies. Recent research suggests that cancer-specific modifications to multiple subtypes of nonapoptotic regulated cell death (RCD), such as ferroptosis and cuproptosis, contribute to therapeutic resistance in neuroblastoma. Targeting these specific types of RCD may be viable novel targets for future drug discovery in the treatment of neuroblastoma. In this review, we summarize the core mechanisms by which the inability to properly execute ferroptosis and cuproptosis can enhance the pathogenesis of neuroblastoma. Therefore, we focus on emerging therapeutic compounds that can induce ferroptosis or cuproptosis, delineating their beneficial pharmacodynamic effects in neuroblastoma treatment. Cumulatively, we suggest that the pharmacological stimulation of ferroptosis and ferroptosis may be a novel and therapeutically viable strategy to target neuroblastoma.
Collapse
Affiliation(s)
- Ying Liu
- Department of Pediatrics, The Fourth Affiliated Hospital of China Medical University, Shenyang, 100012, China.
| | - Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Hongquan Wang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China
| | - Liang Huo
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 11004, China.
| |
Collapse
|
18
|
Williams KM, Shah NR, Chukkapalli S, King S, Grant CN, Brown EG, Avanzini S, Lal DR, Sarnacki S, Newman EA. Modern surgical strategies in pediatric neuroblastoma: Evolving approaches and treatment principles. Pediatr Blood Cancer 2024:e31317. [PMID: 39313754 DOI: 10.1002/pbc.31317] [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: 07/05/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/25/2024]
Abstract
Neuroblastoma, the most common extracranial solid tumor in children under the age of 5, has been described as early as the 19th century, and its complexity has continued to intrigue researchers, as well as medical and surgical specialists. At one end of the phenotypic spectrum, neuroblastoma is self-limiting with minimal to no intervention required, while on the opposite end exists the challenge of refractory disease despite aggressive management and toxic systemic treatments. The goal of this review is to describe a comprehensive surgical perspective and contemporary approach to neuroblastoma.
Collapse
Affiliation(s)
- Keyonna M Williams
- Section of Pediatric Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Nikhil R Shah
- Section of Pediatric Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Sahiti Chukkapalli
- Section of Pediatric Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Sarah King
- Section of Pediatric Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Christa N Grant
- Department of Pediatric Surgery, Maria Fareri Children's Hospital, Valhalla, New York, USA
| | - Erin G Brown
- Division of Pediatric Surgery, University of California Davis Medical Center, Sacramento, California, USA
| | - Stefano Avanzini
- Department of Pediatric Surgery, IRCCS Istituto G. Gaslini, Genoa, Italy
| | - Dave R Lal
- Department of Pediatric Surgery, Children's Wisconsin, Milwaukee, Wisconsin, USA
| | - Sabine Sarnacki
- Department of Pediatric Surgery, Hôpital Universitaire Necker, Paris, France
| | - Erika A Newman
- Section of Pediatric Surgery, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
19
|
Li M, Sun F, Wang J, Lu S, Que Y, Song M, Chen H, Xiong X, Xie W, Zhu J, Huang J, Zhang Y, Zhang Y. SUV39H1 epigenetically modulates the MCPIP1-AURKA signaling axis to enhance neuroblastoma tumorigenesis. Oncogene 2024:10.1038/s41388-024-03164-4. [PMID: 39300256 DOI: 10.1038/s41388-024-03164-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 09/04/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024]
Abstract
Epigenetic regulation is a pivotal factor during neuroblastoma (NB) pathogenesis and investigations into cancer epigenetics are actively underway to identify novel therapeutic strategies for NB patients. SUV39H1, a member of the H3K9 methyltransferase family, contributing to tumorigenesis across multiple malignancies. However, its specific role in NB remains unexplored. In this study, we conducted a high-throughput screen utilizing a compound library containing 288 epigenetic drugs, leading to the identification of chaetocin as the most potent NB inhibitor by targeting SUV39H1. Genetic manipulation and therapeutic inhibition of SUV39H1 significantly impacted proliferation, migration, cell cycle phases, and apoptosis in NB cells. Concurrently, chaetocin demonstrated robust anti-tumor efficacy in vivo with tolerable toxicity. RNA-seq unveiled that SUV39H1 knockdown and inhibition down-regulated cell cycle pathways, impacting vital genes such as AURKA. Besides, MCPIP1 emerged as a novel tumor suppressor following SUV39H1 inhibition, which decreased AURKA expression in NB. In detail, SUV39H1 mediated the enrichment of H3K9me3 at the promoter region of MCPIP1, repressing the MCPIP1-mediated degradation of AURKA and facilitating the subsequent accumulation of AURKA, which revealed the oncogenic role of SUV39H1 via the SUV39H1-MCPIP1-AURKA signaling axis in NB. Therapeutic inhibition of SUV39H1 using chaetocin emerges as an effective and safe strategy for NB patients. Illustration of the oncogenic pathway regulated by SUV39H1 in NB.
Collapse
Affiliation(s)
- Mengzhen Li
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Feifei Sun
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Juan Wang
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Suying Lu
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yi Que
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Mengjia Song
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Huimou Chen
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- Department of Oncology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, No. 107 Yanjiang Road, Guangzhou, 510120, China
| | - Xiangyu Xiong
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Weiji Xie
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Jia Zhu
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Junting Huang
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yu Zhang
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| | - Yizhuo Zhang
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
20
|
Rados M, Landegger A, Schmutzler L, Rabidou K, Taschner-Mandl S, Fetahu IS. Natural killer cells in neuroblastoma: immunological insights and therapeutic perspectives. Cancer Metastasis Rev 2024:10.1007/s10555-024-10212-8. [PMID: 39294470 DOI: 10.1007/s10555-024-10212-8] [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: 04/15/2024] [Accepted: 09/10/2024] [Indexed: 09/20/2024]
Abstract
Natural killer (NK) cells have multifaceted roles within the complex tumor milieu. They are pivotal components of innate immunity and shape the dynamic landscape of tumor-immune cell interactions, and thus can be leveraged for use in therapeutic interventions. NK-based immunotherapies have had remarkable success in hematological malignancies, but these therapies are met with many challenges in solid tumors, including neuroblastoma (NB), a childhood tumor arising from the sympathetic nervous system. With a focus on NB, this review outlines the mechanisms employed by NK cells to recognize and eliminate malignant cells, delving into the dynamic relationship between ligand-receptor interactions, cytokines, and other molecules that facilitate the cross talk between NK and NB cells. We discuss the immunomodulatory functions of NK cells and the mechanisms that contribute to loss of this immunosurveillance in NB, with a focus on how this dynamic has been utilized in recent immunotherapy advancements for NB.
Collapse
Affiliation(s)
- Magdalena Rados
- St. Anna Children's Cancer Research Institute, Vienna, Austria
| | | | - Lukas Schmutzler
- Department of Otorhinolaryngology - Head and Neck Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Kimberlie Rabidou
- Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, USA
| | | | - Irfete S Fetahu
- Department of Neurology, Division of Neuropathology and Neurochemistry, Medical University of Vienna, Vienna, Austria.
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
21
|
Wang JX, Zhang HY, Yan ZJ, Cao ZY, Shao JB, Zou L. Identification and validation of a novel five-gene signature in high-risk MYCN-not-amplified neuroblastoma. Discov Oncol 2024; 15:456. [PMID: 39292372 PMCID: PMC11410741 DOI: 10.1007/s12672-024-01318-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 09/05/2024] [Indexed: 09/19/2024] Open
Abstract
OBJECTIVE High-risk neuroblastoma patients often have poor outcomes despite multi-treatment options. The risk stratification of high-risk MYCN-not-amplified (HR-MYCN-NA) patients remains difficult. This study aims to identify a gene set signature that can help further stratify HR-MYCN-NA patients for a potential personalized therapeutic strategy. METHODS Three microarrays and one single-cell RNA sequence dataset were acquired and analyzed. Firstly, the prognostic-related genes (PRGs) in HR-MYCN-NA tumor cells were identified using TARGET-NB and GSE137804 datasets. Then, the prognostic model was established by LASSO-Cox regression, and verified in external cohort (GSE49710, GSE45547). Moreover, a time-dependent receiver operating characteristic curve (ROC) and area under the ROC (AUC) was used to assess survival prediction. A nomogram was established to predict the 1-, 3- and 5-year overall survival (OS) of HR-MYCN-NA patients. RESULTS In the training set, a five-PRGs signature, which include GAL, GFRA3, MARCKS, PSMD13, and ZNHIT3 genes, was identified and successfully stratified HR-MYCN-NA patients into ultra-high risk (UHR) and high-risk (HR) subtypes (HR = 4.29, P < 0.001). ROC curve analysis confirmed its predictive power (AUC = 0.74-0.82), suggesting a good predictive efficacy. Consistently, high-risk scores also predicted worse OS (HR = 2, P = 0.033) in the external validation dataset (AUC = 0.67-0.71). Moreover, the overall C-index of the nomogram was 0.75 (P < 0.001), which indicated good agreement between the observed and predicted survival rates. Further integrating the five PRGs signature with clinical factors, these 5 gene signature (HR = 4.45, P < 0.001) and tumor grade (HR = 4.15, P = 0.02) were found to be independent prognostic factors for HR-MYCN-NA patients. CONCLUSION The novel five PRGs signature could well predict the survival of HR-MYCN-NA patients, which may provide constructive information for these subsets.
Collapse
Affiliation(s)
- Jin-Xia Wang
- Clinical Research Unit, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, China
| | - Hong-Yang Zhang
- Clinical Research Unit, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, China
| | - Zi-Jun Yan
- Clinical Research Unit, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, China
| | - Zi-Yang Cao
- Clinical Research Unit, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, China
| | - Jing-Bo Shao
- Department of Hematology and Cancer, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, China.
| | - Lin Zou
- Clinical Research Unit, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, China.
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, China.
| |
Collapse
|
22
|
Li C, Lu X, Zhang F, Huang S, Ding L, Wang H, Chen S. Neuroblastoma with high ASPM reveals pronounced heterogeneity and poor prognosis. BMC Cancer 2024; 24:1151. [PMID: 39289658 PMCID: PMC11406734 DOI: 10.1186/s12885-024-12912-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 09/06/2024] [Indexed: 09/19/2024] Open
Abstract
OBJECTIVE We explored the preliminary value of abnormal spindle-like microcephaly- associated (ASPM) protein in aiding precise risk sub-stratification, prediction of metabolic heterogeneity, and prognosis of neuroblastoma (NB). METHODS This retrospective study enrolled newly diagnosed patients with NB who underwent positron emission tomography/computed tomography (PET/CT) before therapy, and tumor tissue was collected after surgery. Regression analysis was used to evaluate ASPM expression and risk stratification in patients with NB. The expression levels of ASPM, clinical information, and PET/CT text features were analyzed using univariate and multivariate survival analyses. Finally, a correlation analysis was used to explore the relationship between ASPM and tumor metabolic heterogeneity. RESULTS There were 48 patients with NB in this study (35 boys and 13 girls); 22 patients progressed and 16 died. We found that the level of ASPM was highly associated with risk stratification (OR = 5.295, 95%IC: 1.348-41.722, p = 0.021). Patients with NB and high-risk stratification with high ASPM level had a lower 3-year progression-free survival (PFS) rate (14.28%) and 1-year PFS rate (57.14%) than those with low ASPM level (57.14% and 93.75%, respectively). Using univariate and multivariate survival analyses, this study revealed that ASPM and LDH were independent risk factors for both PFS and overall survival (OS), whales GLZLM_ZLNU was only a risk factor for PFS. CONCLUSION ASPM holds promise as a novel biomarker for refining current risk stratification and predicting prognosis in neuroblastoma. Elevated levels of ASPM, LDH, and GLZLM_ZLNU may be associated with poorer survival outcomes in neuroblastoma patients.
Collapse
Affiliation(s)
- Chao Li
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Xueyuan Lu
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Fengxian Zhang
- Department of Nuclear Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, 507 Zhengmin Road, Shanghai, 200433, China
| | - Shuo Huang
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Lin Ding
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Hui Wang
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China.
| | - Suyun Chen
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China.
| |
Collapse
|
23
|
Alfei S, Giannoni P, Signorello MG, Torazza C, Zuccari G, Athanassopoulos CM, Domenicotti C, Marengo B. The Remarkable and Selective In Vitro Cytotoxicity of Synthesized Bola-Amphiphilic Nanovesicles on Etoposide-Sensitive and -Resistant Neuroblastoma Cells. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1505. [PMID: 39330662 PMCID: PMC11434613 DOI: 10.3390/nano14181505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/04/2024] [Accepted: 09/11/2024] [Indexed: 09/28/2024]
Abstract
Neuroblastoma (NB) is a solid tumor occurring in infancy and childhood. Its high-risk form has currently a survival rate <50%, despite aggressive treatments. This worrying scenario is worsened by drug-induced secondary tumorigenesis and the emergency of drug resistance, calling for the urgent development of new extra-genomic treatments. Triphenyl phosphonium salts (TPPs) are mitochondria-targeting compounds that exert anticancer effects, impair mitochondria functions, and damage DNA at the same time. Despite several biochemical applications, TPP-based bola-amphiphiles self-assembling nanoparticles (NPs) in water have never been tested as antitumor agents. Here, with the aim of developing new antitumor devices to also counteract resistant forms of HR-NB, the anticancer effects of a TPP-based bola-amphiphile molecule have been investigated in vitro for the first time. To this end, we considered the previously synthesized and characterized sterically hindered quaternary phosphonium salt (BPPB). It embodies both the characteristics of mitochondria-targeting compounds and those of bola-amphiphiles. The anticancer effects of BPPB were assessed against HTLA-230 human stage-IV NB cells and their counterpart, which is resistant to etoposide (ETO), doxorubicin (DOX), and many other therapeutics (HTLA-ER). Very low IC50 values of 0.2 µM on HTLA-230 and 1.1 µM on HTLA-ER (538-fold lower than that of ETO) were already determined after 24 h of treatment. The very low cell viability observed after 24 h did not significantly differ from that observed for the longest exposure timing. The putative future inclusion of BPPB in a chemotherapeutic cocktail for HR-NB was assessed by investigating in vitro its cytotoxic effects against mammalian cell lines. These included monkey kidney cells (Cos-7, IC50 = 4.9 µM), human hepatic cells (HepG2, IC50 = 9.6 µM), a lung-derived fibroblast cell line (MRC-5, IC50 = 2.8 µM), and red blood cells (RBCs, IC50 = 14.9 µM). Appreciable to very high selectivity indexes (SIs) have been determined after 24 h treatments (SIs = 2.5-74.6), which provided evidence that both NB cell populations were already fully exterminated. These in vitro results pave the way for future investigations of BPPB on animal models and upon confirmation for the possible development of BPPB as a novel therapeutic to treat MDR HR-NB cells.
Collapse
Affiliation(s)
- Silvana Alfei
- Department of Pharmacy, University of Genoa, Viale Cembrano, 16148 Genoa, Italy; (C.T.); (G.Z.)
| | - Paolo Giannoni
- Department of Experimental Medicine (DIMES), University of Genova, Via Alberti L.B., 16132 Genoa, Italy; (P.G.); (C.D.)
| | - Maria Grazia Signorello
- Biochemistry Laboratory, Department of Pharmacy, University of Genoa, Viale Benedetto XV 3, 16132 Genova, Italy;
| | - Carola Torazza
- Department of Pharmacy, University of Genoa, Viale Cembrano, 16148 Genoa, Italy; (C.T.); (G.Z.)
| | - Guendalina Zuccari
- Department of Pharmacy, University of Genoa, Viale Cembrano, 16148 Genoa, Italy; (C.T.); (G.Z.)
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, Via G. Gaslini 5, 16147 Genoa, Italy
| | | | - Cinzia Domenicotti
- Department of Experimental Medicine (DIMES), University of Genova, Via Alberti L.B., 16132 Genoa, Italy; (P.G.); (C.D.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Barbara Marengo
- Department of Experimental Medicine (DIMES), University of Genova, Via Alberti L.B., 16132 Genoa, Italy; (P.G.); (C.D.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| |
Collapse
|
24
|
Liu X, Peng X, Yang S, Liu H, Zhang S, Wang J, Ma Y, Wu Y, Wang Z, Weng W, Li Y. Salvage chemotherapy regimens with arsenic trioxide for relapsed or refractory neuroblastoma: a promising approach. BMC Cancer 2024; 24:1140. [PMID: 39266997 PMCID: PMC11395222 DOI: 10.1186/s12885-024-12884-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 09/02/2024] [Indexed: 09/14/2024] Open
Abstract
In patients with relapsed or refractory neuroblastoma (NB), the limited efficacy of conventional chemotherapies necessitates the exploration of new treatment options. Previous studies have highlighted the anti-tumor properties of arsenic trioxide (ATO) in high-risk NB (HR-NB). This study aims to assess the effectiveness and safety of ATO combined with salvage chemotherapy regimens, featuring cyclophosphamide and topotecan, as a foundational treatment for children with relapsed or refractory NB. Eleven patients (four relapsed, seven refractory NB) were retrospectively analyzed for efficacy and treatment relevance. Salvage treatments, incorporating ATO (0.18 mg/kg daily for 8 h intravenously on days 1 to 10), were administered upon disease progression or relapse, with assessments conducted every two cycles. Treatments had 63.6% efficacy, with six cases of partial response, one case of stable disease, and four cases of disease progression. The overall response rate was 54.5%, and the disease control rate was 63.6%. Importantly, the systemic toxicity experienced by patients following salvage chemotherapy with ATO was mild. Salvage chemotherapy regimens featuring ATO demonstrated potential for prolonging disease stabilization for relapsed or refractory HR-NB patients, exhibiting both favorable efficacy and safety profiles. This suggests further clinical exploration and promotion of this therapeutic approach in the treatment of NB.
Collapse
Affiliation(s)
- Xiaoshan Liu
- Pediatric Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No.107, Yanjiang West Road, Yuexiu District, Guangzhou, 510120, Guangdong, China
| | - Xiaomin Peng
- Pediatric Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No.107, Yanjiang West Road, Yuexiu District, Guangzhou, 510120, Guangdong, China
| | - Shu Yang
- Pediatric Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No.107, Yanjiang West Road, Yuexiu District, Guangzhou, 510120, Guangdong, China
| | - Haijin Liu
- Department of Pediatric Surgery, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, Ganzhou, 341000, Jiangxi, China
| | - Shouhua Zhang
- Department of General Surgery, Jiangxi Provincial Children's Hospital, Nanchang, 330006, Jiangxi, China
| | - Jinhu Wang
- Department of Surgical Oncology, Children's Hospital Zhejiang University School of Medicine, Hangzhou, 310052, Zhejiang, China
| | - Yuhan Ma
- Pediatric Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No.107, Yanjiang West Road, Yuexiu District, Guangzhou, 510120, Guangdong, China
| | - Yu Wu
- Pediatric Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No.107, Yanjiang West Road, Yuexiu District, Guangzhou, 510120, Guangdong, China
| | - Zhixuan Wang
- Pediatric Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No.107, Yanjiang West Road, Yuexiu District, Guangzhou, 510120, Guangdong, China
| | - Wenjun Weng
- Pediatric Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No.107, Yanjiang West Road, Yuexiu District, Guangzhou, 510120, Guangdong, China
| | - Yang Li
- Pediatric Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No.107, Yanjiang West Road, Yuexiu District, Guangzhou, 510120, Guangdong, China.
| |
Collapse
|
25
|
Miera-Maluenda M, Pérez-Torres M, Mañas A, Rubio-San-Simón A, Butjosa-Espín M, Ruiz-Duran P, Seoane JA, Moreno L, Segura MF. Advances in the approaches used to repurpose drugs for neuroblastoma. Expert Opin Drug Discov 2024:1-11. [PMID: 39258785 DOI: 10.1080/17460441.2024.2402413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 09/05/2024] [Indexed: 09/12/2024]
Abstract
INTRODUCTION Neuroblastoma (NB) remains a challenging pediatric malignancy with limited treatment options, particularly for high-risk cases. Drug repurposing offers a convenient and cost-effective strategy for treating rare diseases like NB. Using existing drugs with known safety profiles accelerates the availability of new treatments, reduces development costs, and mitigates risks, offering hope for improved patient outcomes in challenging conditions. AREAS COVERED This review provides an overview of the advances in approaches used to repurpose drugs for NB therapy. The authors discuss strategies employed in drug repurposing, including computational and experimental methods, and rational drug design, highlighting key examples of repurposed drugs with promising clinical results. Additionally, the authors examine the challenges and opportunities associated with drug repurposing in NB and discuss future directions and potential areas for further research. EXPERT OPINION The fact that only one new drug has been approved in the last 30 years for the treatment of neuroblastoma plus a significant proportion of high-risk NB patients that remain uncurable, evidences the need for new fast and cost-effective alternatives. Drug repurposing may accelerate the treatment development process while reducing expenses and risks. This approach can swiftly bring effective NB therapies to market, enhancing survival rates and patient quality of life.
Collapse
Affiliation(s)
- Marta Miera-Maluenda
- Childhood Cancer and Blood Disorders Group, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - María Pérez-Torres
- Department of Pediatric Oncology and Hematology, Vall D'Hebron University Hospital, Barcelona, Spain
| | - Adriana Mañas
- Translational Research in Pediatric Oncology, Hematopoietic Transplantation and Cell Therapy, IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
- IdiPAZ-CNIO Pediatric Onco-Hematology Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Alba Rubio-San-Simón
- Pediatric Oncology and Hematology Department, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Maria Butjosa-Espín
- Cancer Computational Biology Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Paula Ruiz-Duran
- Childhood Cancer and Blood Disorders Group, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jose A Seoane
- Cancer Computational Biology Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Lucas Moreno
- Childhood Cancer and Blood Disorders Group, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Pediatric Oncology and Hematology, Vall D'Hebron University Hospital, Barcelona, Spain
| | - Miguel F Segura
- Childhood Cancer and Blood Disorders Group, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
26
|
Park SWS, Fransson S, Sundquist F, Nilsson JN, Grybäck P, Wessman S, Strömgren J, Djos A, Fagman H, Sjögren H, Georgantzi K, Herold N, Kogner P, Granberg D, Gaze MN, Martinsson T, Karlsson K, Stenman JJE. Heterogeneous SSTR2 target expression and a novel KIAA1549:: BRAF fusion clone in a progressive metastatic lesion following 177Lutetium-DOTATATE molecular radiotherapy in neuroblastoma: a case report. Front Oncol 2024; 14:1408729. [PMID: 39324010 PMCID: PMC11422106 DOI: 10.3389/fonc.2024.1408729] [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: 03/28/2024] [Accepted: 06/20/2024] [Indexed: 09/27/2024] Open
Abstract
In this case report, we present the treatment outcomes of the first patient enrolled in the LuDO-N trial. The patient is a 21-month-old girl diagnosed with high-risk neuroblastoma (NB) and widespread skeletal metastasis. The patient initially underwent first-line therapy according to SIOPEN HRNBL-1 but was switched to second-line treatments due to disease progression, and she was finally screened for enrollment in the LuDO-N trial due to refractory disease. Upon enrollment, the patient received two rounds of the radiolabeled somatostatin analogue lutetium-177 octreotate (177Lu-DOTATATE), which was well tolerated. A dosimetry analysis revealed a heterogeneous uptake across tumor lesions, resulting in a significant absorbed dose of 54 Gy in the primary tumor, but only 2 Gy at one of the metastatic sites in the distal femur. While the initial treatment response showed disease stabilization, the distal femoral metastasis continued to progress, leading to the eventual death of the patient. A tissue analysis of the biopsies collected throughout the course of the disease revealed heterogeneous drug target expression of somatostatin receptor 2 (SSTR2) across and within tumor lesions. Furthermore, genomic profiling revealed a novel KIAA1549::BRAF fusion oncogene amplification in the distal femoral metastasis at recurrence that might be related with resistance to radiation, possibly through the downregulation of SSTR2. This case report demonstrates a mixed response to molecular radiotherapy (MRT) with 177Lu-DOTATATE. The observed variation in SSTR2 expression between tumor lesions suggests that heterogeneous target expression may have been the reason for treatment failure in this patient's case. Further investigation within the LuDO-N trial will give a more comprehensive understanding of the correlation between SSTR2 expression levels and treatment outcomes, which will be important to advance treatment strategies based on MRT for children with high-risk NB.
Collapse
Affiliation(s)
- Se Whee Sammy Park
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Susanne Fransson
- Department of Laboratory Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Fredrik Sundquist
- Department of Urology, Karolinska University Hospital, Stockholm, Sweden
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Joachim N Nilsson
- Department of Medical Radiation Physics and Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Per Grybäck
- Department of Medical Radiation Physics and Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Sandra Wessman
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| | - Jacob Strömgren
- Department of Pediatric Radiology, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Djos
- Department of Laboratory Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Fagman
- Department of Laboratory Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Helene Sjögren
- Clinical Genetics and Genomics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Kleopatra Georgantzi
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Department of Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Nikolas Herold
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Department of Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Per Kogner
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Department of Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Dan Granberg
- Department of Breast, Endocrine Tumors and Sarcomas, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Mark N Gaze
- Department of Oncology, University College London Hospitals National Health Service (NHS) Foundation Trust, London, United Kingdom
| | - Tommy Martinsson
- Department of Laboratory Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kasper Karlsson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Jakob J E Stenman
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Department of Pediatric Surgery, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
27
|
Zhu X, Si Y, Gai C, Li Z. Investigating the molecular mechanisms of Fuzheng Yiliu Shenji prescription in SH-SY5Y neuroblastoma cells. Front Oncol 2024; 14:1447666. [PMID: 39319058 PMCID: PMC11420165 DOI: 10.3389/fonc.2024.1447666] [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: 06/12/2024] [Accepted: 08/15/2024] [Indexed: 09/26/2024] Open
Abstract
Background Neuroblastoma is the most common extracranial solid tumor in childhood. Fuzheng Yiliu Shenji Prescription (FYSP) has shown potential in treating malignant pediatric tumors in clinical settings. This study aims to explore the molecular mechanisms behind its effects, specifically in the context of neuroblastoma cell lines. Objective To elucidate the active compounds in FYSP and their mechanisms of action in inhibiting neuroblastoma cell viability, inducing apoptosis, and affecting the cell cycle in SH-SY5Y cells through network pharmacology and empirical validation. Materials and methods We identified the major compounds in FYSP and their predicted targets, constructing a protein-protein interaction (PPI) network and performing GO and KEGG pathway analyses. The effects of FYSP were empirically validated through assays on cell viability, cell cycle, apoptosis, and protein expression in SH-SY5Y cells. Results The study identified 172 active chemical components in FYSP, with 188 common targets related to neuroblastoma. Network analysis highlighted the PI3K-Akt pathway as a significant target. Experimental validation in SH-SY5Y cells confirmed that FYSP could inhibit cell viability, induce G2/M cell cycle arrest, and promote apoptosis through modulation of the PI3K-Akt pathway, specifically upregulating caspase-3 and downregulating Bcl-2/Bax expression. Conclusion The study elucidates the molecular basis of FYSP's effects on neuroblastoma cells in vitro, demonstrating its ability to modulate key pathways involved in cell cycle and apoptosis. While these findings suggest a potential therapeutic role for FYSP, they are limited to in vitro observations, and further research, including in vivo studies, is necessary to explore its clinical applicability.
Collapse
Affiliation(s)
- Xueying Zhu
- Department of Anatomy, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yinchu Si
- Department of Anatomy, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Cong Gai
- Department of Anatomy, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zhong Li
- Department of Oncology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
28
|
Nokchan N, Suthapot P, Choochuen P, Khongcharoen N, Hongeng S, Anurathapan U, Surachat K, Sangkhathat S, Thai Pediatric Cancer Atlas Tpca Consortium. Whole-Exome Sequencing Reveals Novel Candidate Driver Mutations and Potential Druggable Mutations in Patients with High-Risk Neuroblastoma. J Pers Med 2024; 14:950. [PMID: 39338204 PMCID: PMC11433071 DOI: 10.3390/jpm14090950] [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: 05/01/2024] [Revised: 08/26/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Neuroblastoma is the most prevalent solid tumor in early childhood, with a 5-year overall survival rate of 40-60% in high-risk cases. Therefore, the identification of novel biomarkers for the diagnosis, prognosis, and therapy of neuroblastoma is crucial for improving the clinical outcomes of these patients. In this study, we conducted the whole-exome sequencing of 48 freshly frozen tumor samples obtained from the Biobank. Somatic variants were identified and selected using a bioinformatics analysis pipeline. The mutational signatures were determined using the Mutalisk online tool. Cancer driver genes and druggable mutations were predicted using the Cancer Genome Interpreter. The most common mutational signature was single base substitution 5. MUC4, MUC16, and FLG were identified as the most frequently mutated genes. Using the Cancer Genome Interpreter, we identified five recurrent cancer driver mutations spanning MUC16, MUC4, ALK, and CTNND1, with the latter being novel and containing a missense mutation, R439C. We also identified 11 putative actionable mutations including NF1 Q1798*, Q2616*, and S636X, ALK F1174L and R1275Q, SETD2 P10L and Q1829E, BRCA1 R612S, NOTCH1 D1670V, ATR S1372L, and FGFR1 N577K. Our findings provide a comprehensive overview of the novel information relevant to the underlying molecular pathogenesis and therapeutic targets of neuroblastoma.
Collapse
Affiliation(s)
- Natakorn Nokchan
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
- Translational Medicine Research Center, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Praewa Suthapot
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
- Division of Hematology and Oncology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Pongsakorn Choochuen
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
- Translational Medicine Research Center, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Natthapon Khongcharoen
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
- Translational Medicine Research Center, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Suradej Hongeng
- Division of Hematology and Oncology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Usanarat Anurathapan
- Division of Hematology and Oncology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Komwit Surachat
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
- Translational Medicine Research Center, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Surasak Sangkhathat
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
- Translational Medicine Research Center, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
- Department of Surgery, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | | |
Collapse
|
29
|
Lin M, Hua Z, Li Z. FTO diversely influences sensitivity of neuroblastoma cells to various chemotherapeutic drugs. Front Pharmacol 2024; 15:1384141. [PMID: 39295930 PMCID: PMC11409730 DOI: 10.3389/fphar.2024.1384141] [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: 02/08/2024] [Accepted: 08/16/2024] [Indexed: 09/21/2024] Open
Abstract
Chemotherapy resistance is a significant factor in treatment failure in patients with neuroblastoma (NB), and it directly affects patient prognosis. Therefore, identifying novel therapeutic targets to enhance chemosensitivity is essential to improve the cure rate and prognosis of patients with NB. In this study, we investigated the role of FTO in chemosensitivity of NB cells to various chemotherapeutic drugs. Our results showed that high FTO expression was positively correlated with increased survival probability and favorable prognostic factors in patients with NB. FTO overexpression inhibited cell proliferation, whereas FTO knockdown promoted cell proliferation in NB cells. FTO expression alteration had contrasting effects on NB cells' sensitivity to etoposide but had no significant impact on sensitivity to cisplatin. Downregulation of FTO reduced the sensitivity of NB cells to paclitaxel, whereas upregulation of FTO enhanced its sensitivity. Additionally, the sensitivities between patients with lower and higher FTO expression to various chemotherapeutic drugs or small-molecule inhibitors were different. Thus, FTO affects the sensitivities of NB cells differently depending on the different chemotherapeutic drugs and small-molecule inhibitors. This finding may guide physicians and patients choose the appropriate chemotherapeutic drugs or small-molecule inhibitors for treatment.
Collapse
Affiliation(s)
- Meizhen Lin
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
- Liaoning Key Laboratory of Research and Application of Animal Models for Environmental and Metabolic Diseases, Medical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhongyan Hua
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
- Liaoning Key Laboratory of Research and Application of Animal Models for Environmental and Metabolic Diseases, Medical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhijie Li
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
- Liaoning Key Laboratory of Research and Application of Animal Models for Environmental and Metabolic Diseases, Medical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
30
|
Ekstrom TL, Hussain S, Bedekovics T, Ali A, Paolini L, Mahmood H, Rosok RM, Koster J, Johnsen SA, Galardy PJ. USP44 Overexpression Drives a MYC-Like Gene Expression Program in Neuroblastoma through Epigenetic Reprogramming. Mol Cancer Res 2024; 22:812-825. [PMID: 38775808 PMCID: PMC11372370 DOI: 10.1158/1541-7786.mcr-23-0454] [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: 06/05/2023] [Revised: 04/05/2024] [Accepted: 05/20/2024] [Indexed: 09/05/2024]
Abstract
Neuroblastoma is an embryonic cancer that contributes disproportionately to death in young children. Sequencing data have uncovered few recurrently mutated genes in this cancer, although epigenetic pathways have been implicated in disease pathogenesis. We used an expression-based computational screen that examined the impact of deubiquitinating enzymes on patient survival to identify potential new targets. We identified the histone H2B deubiquitinating enzyme USP44 as the enzyme with the greatest impact on survival in patients with neuroblastoma. High levels of USP44 significantly correlate with metastatic disease, unfavorable histology, advanced patient age, and MYCN amplification. The subset of patients with tumors expressing high levels of USP44 had significantly worse survival, including those with tumors lacking MYCN amplification. We showed experimentally that USP44 regulates neuroblastoma cell proliferation, migration, invasion, and neuronal development. Depletion of the histone H2B ubiquitin ligase subunit RNF20 resulted in similar findings, strongly implicating this histone mark as the target of USP44 activity in this disease. Integration of transcriptome and epigenome in analyses demonstrates a distinct set of genes that are regulated by USP44, including those in Hallmark MYC target genes in both murine embryonic fibroblasts and the SH-SY5Y neuroblastoma cell line. We conclude that USP44 is a novel epigenetic regulator that promotes aggressive features and may be a novel target in neuroblastoma. Implications: This study identifies a new genetic marker of aggressive neuroblastoma and identifies the mechanisms by which its overactivity contributes to the pathophysiology of this disease.
Collapse
Affiliation(s)
- Thomas L. Ekstrom
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, Minnesota.
- Robert Bosch Center for Tumor Diseases, Stuttgart, Germany.
| | - Sajjad Hussain
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota.
- Department of Family Medicine, Mayo Clinic, Rochester, Minnesota.
| | - Tibor Bedekovics
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota.
| | - Asma Ali
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota.
| | - Lucia Paolini
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota.
- Department of Pediatrics, University of Milano-Bicocca, San Gerardo Hospital, Monza, Italy.
| | - Hina Mahmood
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota.
| | - Raya M. Rosok
- Robert Bosch Center for Tumor Diseases, Stuttgart, Germany.
| | - Jan Koster
- Department of CEMM, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.
| | | | - Paul J. Galardy
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota.
- Division of Pediatric Hematology-Oncology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
31
|
Ek T, Ibrahim RR, Vogt H, Georgantzi K, Träger C, Gaarder J, Djos A, Rahmqvist I, Mellström E, Pujol-Calderón F, Vannas C, Hansson L, Fagman H, Treis D, Fransson S, Österlund T, Chuang TP, Verhoeven BM, Ståhlberg A, Palmer RH, Hallberg B, Martinsson T, Kogner P, Dalin M. Long-Lasting Response to Lorlatinib in Patients with ALK-Driven Relapsed or Refractory Neuroblastoma Monitored with Circulating Tumor DNA Analysis. CANCER RESEARCH COMMUNICATIONS 2024; 4:2553-2564. [PMID: 39177282 PMCID: PMC11440348 DOI: 10.1158/2767-9764.crc-24-0338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/21/2024] [Accepted: 08/21/2024] [Indexed: 08/24/2024]
Abstract
Patients with anaplastic lymphoma kinase (ALK)-driven neuroblastoma may respond to tyrosine kinase inhibitors, but resistance to treatment occurs and methods currently used for detection of residual disease have limited sensitivity. Here, we present a national unselected cohort of five patients with relapsed or refractory ALK-driven neuroblastoma treated with lorlatinib as monotherapy and test the potential of targeted circulating tumor DNA (ctDNA) analysis as a guide for treatment decisions in these patients. We developed a sequencing panel for ultrasensitive detection of ALK mutations associated with neuroblastoma or resistance to tyrosine kinase inhibitors and used it for ctDNA analysis in 83 plasma samples collected longitudinally from the four patients who harbored somatic ALK mutations. All four patients with ALK p.R1275Q experienced major responses and were alive 35 to 61 months after starting lorlatinib. A fifth patient with ALK p.F1174L initially had a partial response but relapsed after 10 months of treatment. In all cases, ctDNA was detected at the start of lorlatinib single-agent treatment and declined gradually, correlating with clinical responses. In the two patients exhibiting relapse, ctDNA increased 9 and 3 months, respectively, before clinical detection of disease progression. In one patient harboring HRAS p.Q61L in the relapsed tumor, retrospective ctDNA analysis showed that the mutation appeared de novo after 8 months of lorlatinib treatment. We conclude that some patients with relapsed or refractory high-risk neuroblastoma show durable responses to lorlatinib as monotherapy, and targeted ctDNA analysis is effective for evaluation of treatment and early detection of relapse in ALK-driven neuroblastoma. SIGNIFICANCE We present five patients with ALK-driven relapsed or refractory neuroblastoma treated with lorlatinib as monotherapy. All patients responded to treatment, and four of them were alive after 3 to 5 years of follow-up. We performed longitudinal ctDNA analysis with ultra-deep sequencing of the ALK tyrosine kinase domain. We conclude that ctDNA analysis may guide treatment decisions in ALK-driven neuroblastoma, also when the disease is undetectable using standard clinical methods.
Collapse
Affiliation(s)
- Torben Ek
- Children’s Cancer Centre, Queen Silvia Children’s Hospital, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden.
- Department of Pediatrics, Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
| | - Raghda R. Ibrahim
- Department of Pediatrics, Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - Hartmut Vogt
- Department of Biomedical and Clinical Sciences, Crown Princess Victoria Children’s Hospital, and Division of Children’s and Women’s Health, Linköping University, Linköping, Sweden.
| | - Kleopatra Georgantzi
- Department of Pediatric Hematology and Oncology, Karolinska University Hospital, Stockholm, Sweden.
- Childhood Cancer Research Unit, Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden.
| | - Catarina Träger
- Childhood Cancer Research Unit, Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden.
- Department of Pediatric Hematology and Oncology, Academic Children’s Hospital, Uppsala, Sweden.
- Department of Women’s and Children’s Health, Uppsala University, Uppsala, Sweden.
| | - Jennie Gaarder
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
- Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden.
| | - Anna Djos
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
| | - Ida Rahmqvist
- Department of Pediatrics, Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - Elisabeth Mellström
- Children’s Cancer Centre, Queen Silvia Children’s Hospital, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden.
- Department of Pediatrics, Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - Fani Pujol-Calderón
- Department of Pediatrics, Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - Christoffer Vannas
- Department of Laboratory Medicine, Sahlgrenska Center for Cancer Research, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
- Department of Oncology, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden.
| | - Lina Hansson
- Department of Oncology, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden.
| | - Henrik Fagman
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
- Department of Clinical Pathology, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden.
| | - Diana Treis
- Department of Pediatric Hematology and Oncology, Karolinska University Hospital, Stockholm, Sweden.
- Childhood Cancer Research Unit, Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden.
| | - Susanne Fransson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
- Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden.
| | - Tobias Österlund
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.
- Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden.
- Department of Laboratory Medicine, Sahlgrenska Center for Cancer Research, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
| | - Tzu-Po Chuang
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
| | - Bronte Manouk Verhoeven
- Childhood Cancer Research Unit, Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden.
| | - Anders Ståhlberg
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.
- Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden.
- Department of Laboratory Medicine, Sahlgrenska Center for Cancer Research, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
| | - Ruth H. Palmer
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
| | - Bengt Hallberg
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
| | - Tommy Martinsson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
| | - Per Kogner
- Department of Pediatric Hematology and Oncology, Karolinska University Hospital, Stockholm, Sweden.
- Childhood Cancer Research Unit, Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden.
| | - Martin Dalin
- Children’s Cancer Centre, Queen Silvia Children’s Hospital, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden.
- Department of Pediatrics, Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
32
|
Qiao S, Jia Y, Xie L, Jing W, Xia Y, Song Y, Zhang J, Cao T, Song H, Meng L, Shi L, Zhang X. KLF7 promotes neuroblastoma differentiation through the GTPase signaling pathway by upregulating neuroblast differentiation-associated protein AHNAKs and glycerophosphodiesterase GDPD5. FEBS J 2024; 291:3870-3888. [PMID: 38924469 DOI: 10.1111/febs.17208] [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: 09/29/2023] [Revised: 03/10/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024]
Abstract
The arrest of neural crest-derived sympathoadrenal neuroblast differentiation contributes to neuroblastoma formation, and overriding this blocked differentiation is a clear strategy for treating high-risk neuroblastoma. A better understanding of neuroblast or neuroblastoma differentiation is essential for developing new therapeutic approaches. It has been proposed that Krueppel-like factor 7 (KLF7) is a neuroblastoma super-enhancer-associated transcription factor gene. Moreover, KLF7 was found to be intensely active in postmitotic neuroblasts of the developing nervous system during embryogenesis. However, the role of KLF7 in the differentiation of neuroblast or neuroblastoma is unknown. Here, we find a strong association between high KLF7 expression and favorable clinical outcomes in neuroblastoma. KLF7 induces differentiation of neuroblastoma cells independently of the retinoic acid (RA) pathway and acts cooperatively with RA to induce neuroblastoma differentiation. KLF7 alters the GTPase activity and multiple differentiation-related genes by binding directly to the promoters of neuroblast differentiation-associated protein (AHNAK and AHNAK2) and glycerophosphodiester phosphodiesterase domain-containing protein 5 (GDPD5) and regulating their expression. Furthermore, we also observe that silencing KLF7 in neuroblastoma cells promotes the adrenergic-to-mesenchymal transition accompanied by changes in enhancer-mediated gene expression. Our results reveal that KLF7 is an inducer of neuroblast or neuroblastoma differentiation with prognostic significance and potential therapeutic value.
Collapse
Affiliation(s)
- Shupei Qiao
- Heilongjiang Province Key Laboratory of Child Development and Genetic Research, Harbin Medical University, China
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, China
| | - Ying Jia
- Heilongjiang Province Key Laboratory of Child Development and Genetic Research, Harbin Medical University, China
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, China
| | - Li Xie
- Heilongjiang Province Key Laboratory of Child Development and Genetic Research, Harbin Medical University, China
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, China
| | - Wenwen Jing
- Heilongjiang Province Key Laboratory of Child Development and Genetic Research, Harbin Medical University, China
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, China
| | - Yang Xia
- Heilongjiang Province Key Laboratory of Child Development and Genetic Research, Harbin Medical University, China
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, China
| | - Yue Song
- Heilongjiang Province Key Laboratory of Child Development and Genetic Research, Harbin Medical University, China
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, China
| | - Jiahui Zhang
- Heilongjiang Province Key Laboratory of Child Development and Genetic Research, Harbin Medical University, China
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, China
| | - Tianhua Cao
- Heilongjiang Province Key Laboratory of Child Development and Genetic Research, Harbin Medical University, China
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, China
| | - Huilin Song
- Heilongjiang Province Key Laboratory of Child Development and Genetic Research, Harbin Medical University, China
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, China
| | - Lingdi Meng
- Heilongjiang Province Key Laboratory of Child Development and Genetic Research, Harbin Medical University, China
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, China
| | - Lei Shi
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, China
| | - Xue Zhang
- Heilongjiang Province Key Laboratory of Child Development and Genetic Research, Harbin Medical University, China
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, China
| |
Collapse
|
33
|
Chang YH, Yu CH, Lu MY, Jou ST, Lin CY, Lin KH, Chang HH, Ni YL, Chou SW, Ko KY, Lin DT, Hsu WM, Chen HY, Yang YL. Higher tumor mutational burden is associated with inferior outcomes among pediatric patients with neuroblastoma. Pediatr Blood Cancer 2024; 71:e31176. [PMID: 38967585 DOI: 10.1002/pbc.31176] [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: 11/06/2023] [Revised: 05/18/2024] [Accepted: 06/17/2024] [Indexed: 07/06/2024]
Abstract
INTRODUCTION Neuroblastoma is a pediatric malignancy with heterogeneous clinical outcomes. Our aim was to identify prognostic genetic markers for patients with neuroblastoma, who were treated with the Taiwan Pediatric Oncology Group (TPOG) neuroblastoma N2002 protocol, to improve risk stratification and inform treatment. METHODS Our analysis was based on 53 primary neuroblastoma specimens, diagnosed pre-chemotherapy, and 11 paired tumor relapse specimens. Deep sequencing of 113 target genes was performed using a custom panel. Multiplex ligation-dependent probe amplification was performed to identify clinical outcomes related to copy-number variations. RESULTS We identified 128 variations associated with survival, with the number of variations being higher in the relapse than that in the diagnostic specimen (p = .03). The risk of event and mortality was higher among patients with a tumor mutational burden ≥10 than that in patients with a lower burden (p < .0001). Multivariate analysis identified tumor mutational burden, MYCN amplification, and chromosome 3p deletion as significant prognostic factors, independent of age at diagnosis, sex, and tumor stage. The 5-year event-free survival and overall survival rate was lower among patients with high tumor burden than in patients with low tumor burden. Furthermore, there was no survival of patients with an ALK F1147L variation at 5 years after diagnosis. CONCLUSIONS Genome sequencing to determine the tumor mutational burden and ALK variations can improve the risk classification of neuroblastoma and inform treatment.
Collapse
Affiliation(s)
- Ya-Hsuan Chang
- Institute of Molecular and Genomic Medicine, National Health Research Institute, Miaoli, Taiwan
| | - Chih-Hsiang Yu
- Institute of Statistical Science Academia Sinica, Taipei, Taiwan
- Departments of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Meng-Yao Lu
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
- Department of Pediatrics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shiann-Tarng Jou
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
- Department of Pediatrics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chien-Yu Lin
- Institute of Statistical Science Academia Sinica, Taipei, Taiwan
| | - Kai-Hsin Lin
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Hsiu-Hao Chang
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
- Department of Pediatrics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Ling Ni
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Shu-Wei Chou
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Kuan-Yin Ko
- Department of Nuclear Medicine, National Taiwan University Cancer Center, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Dong-Tsamn Lin
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Wen-Ming Hsu
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
- Department of Surgery, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsuan-Yu Chen
- Institute of Statistical Science Academia Sinica, Taipei, Taiwan
| | - Yung-Li Yang
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Department of Laboratory Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Laboratory Medicine and Medical Service, National Taiwan University Cancer Center, Taipei, Taiwan
| |
Collapse
|
34
|
Oyemolade TA, Adeolu AA, Oyewo OL, Oko-Azu VC. Neuroblastoma of the thoracic spine in an adult: a case report. J Surg Case Rep 2024; 2024:rjae602. [PMID: 39324103 PMCID: PMC11421993 DOI: 10.1093/jscr/rjae602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 09/09/2024] [Indexed: 09/27/2024] Open
Abstract
Neuroblastoma (NB) in the adults is extremely rare. Even less encountered is NB involving the adult spine with only a few reported cases. Because of its rarity, there are as yet no well-established treatment guidelines for NB in the adults. Treatment strategies, therefore, are often extrapolated from paediatric data. We report a case of a 51-year-old man with NB involving the thoracic spine who had surgical excision with a good postoperative outcome in the short term.
Collapse
Affiliation(s)
- Toyin A Oyemolade
- Division of Neurosurgery, Department of Surgery, Federal Medical Center Owo, PMB 1053, Owo, Ondo, Nigeria
| | - Augustine A Adeolu
- Division of Neurosurgery, Department of Surgery, College of Medicine, University of Ibadan, Queen Elizabeth Road, PMB 5116, Ibadan, Oyo State, Nigeria
- Department of Neurological Surgery, University College Hospital Ibadan, Queen Elizabeth Road, PMB 5116, Ibadan, Oyo, Nigeria
| | - Oluwafunmito L Oyewo
- Division of Neurosurgery, Department of Surgery, Federal Medical Center Owo, PMB 1053, Owo, Ondo, Nigeria
| | - Vivian C Oko-Azu
- Division of Neurosurgery, Department of Surgery, Federal Medical Center Owo, PMB 1053, Owo, Ondo, Nigeria
| |
Collapse
|
35
|
Dushnitzky S, Ishtayeh H, Ashkenazi A. The new kids on the block: RNA-binding proteins regulate autophagy in disease. FEBS J 2024; 291:3811-3819. [PMID: 38825737 DOI: 10.1111/febs.17195] [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: 02/28/2024] [Revised: 04/30/2024] [Accepted: 05/24/2024] [Indexed: 06/04/2024]
Abstract
Mammalian autophagy is a highly regulated and conserved cellular homeostatic process. Its existence allows the degradation of self-components to mediate cell survival in different stress conditions. Autophagy is involved in the regulation of cellular metabolic needs, protecting the cell or tissue from starvation through the degradation and recycling of cytoplasmic materials and organelles to basic molecular building blocks. It also plays a critical role in eliminating damaged or harmful proteins, organelles, and intracellular pathogens. Thus, a deterioration of the process may result in pathological conditions, such as aging-associated disorders and cancer. Understanding the crucial role of autophagy in maintaining the normal physiological function of cells, tissue, or organs has led to copious and expansive research regarding the regulation of this process. So far, most of the research has revolved around transcriptional and post-translational regulation. Here, we discuss the regulation of autophagy-related (ATG) mRNA transcripts by RNA-binding proteins (RBPs). This analysis focuses on how RBPs modulate autophagy in disease. A deeper understanding of the involvement of RBPs in autophagy can facilitate further research and treatment of a variety of human diseases.
Collapse
Affiliation(s)
- Shai Dushnitzky
- The Department of Cell and Developmental Biology, Faculty of Medical & Health Sciences, Tel Aviv University, Israel
| | - Hasan Ishtayeh
- The Department of Cell and Developmental Biology, Faculty of Medical & Health Sciences, Tel Aviv University, Israel
| | - Avraham Ashkenazi
- The Department of Cell and Developmental Biology, Faculty of Medical & Health Sciences, Tel Aviv University, Israel
- Sagol School of Neuroscience, Tel Aviv University, Israel
| |
Collapse
|
36
|
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
|
37
|
Liu KX, Shaaban SG, Chen JJ, Bagatell R, Lerman BJ, Catalano PJ, DuBois SG, Shusterman S, Ioakeim-Ioannidou M, Yock TI, Shamberger RC, Mattei P, Vu L, Elhalawani H, Dusenbery KE, Vo KT, Huang MS, Friedmann AM, Diller LR, Marcus KJ, MacDonald SM, Terezakis SA, Braunstein SE, Hill-Kayser CE, Haas-Kogan DA. Patterns of recurrence after radiotherapy for high-risk neuroblastoma: Implications for radiation dose and field. Radiother Oncol 2024; 198:110384. [PMID: 38880415 DOI: 10.1016/j.radonc.2024.110384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 05/29/2024] [Accepted: 06/12/2024] [Indexed: 06/18/2024]
Abstract
BACKGROUND Prognosis for patients with high-risk neuroblastoma (HR-NBL) is guarded despite aggressive therapy, and few studies have characterized outcomes after radiotherapy in relation to radiation treatment fields. METHODS Multi-institutional retrospective cohort of 293 patients with HR-NBL who received autologous stem cell transplant (ASCT) and EBRT between 1997-2021. LRR was defined as recurrence at the primary site or within one nodal echelon beyond disease present at diagnosis. Follow-up was defined from the end of EBRT. Event-free survival (EFS) and OS were analyzed by Kaplan-Meier method. Cumulative incidence of locoregional progression (CILP) was analyzed using competing risks of distant-only relapse and death with Gray's test. RESULTS Median follow-up was 7.0 years (range: 0.01-22.4). Five-year CILP, EFS, and OS were 11.9 %, 65.2 %, and 77.5 %, respectively. Of the 31 patients with LRR and imaging review, 15 (48.4 %) had in-field recurrences (>12 Gy), 6 (19.4 %) had marginal failures (≤12 Gy), and 10 (32.3 %) had both in-field and marginal recurrences. No patients receiving total body irradiation (12 Gy) experienced marginal-only failures (p = 0.069). On multivariable analyses, MYCN amplification had higher risk of LRR (HR: 2.42, 95 % CI: 1.06-5.50, p = 0.035) and post-consolidation isotretinoin and anti-GD2 antibody therapy (HR: 0.42, 95 % CI: 0.19-0.94, p = 0.035) had lower risk of LRR. CONCLUSIONS Despite EBRT, LRR remains a contributor to treatment failure in HR-NBL with approximately half of LRRs including a component of marginal failure. Future prospective studies are needed to explore whether radiation fields and doses should be defined based on molecular features such as MYCN amplification, and/or response to chemotherapy.
Collapse
Affiliation(s)
- Kevin X Liu
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sherif G Shaaban
- Department of Radiation Oncology & Molecular Radiation Sciences, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Jie Jane Chen
- Department of Radiation Oncology, University of California at San Francisco, UCSF Benioff Children's Hospital, San Francisco, CA, USA; Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rochelle Bagatell
- Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Benjamin J Lerman
- Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Pediatrics, UCSF Benioff Children's Hospital and UCSF School of Medicine, San Francisco, CA, USA
| | - Paul J Catalano
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, and Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Steven G DuBois
- Department of Pediatrics, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | - Suzanne Shusterman
- Department of Pediatrics, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | - Myrsini Ioakeim-Ioannidou
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Torunn I Yock
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Robert C Shamberger
- Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Peter Mattei
- Department of Surgery, University of Pennsylvania and the Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lan Vu
- Department of Surgery, UCSF Benioff Children's Hospital and UCSF School of Medicine, San Francisco, CA, USA
| | - Hesham Elhalawani
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kathryn E Dusenbery
- Department of Radiation Oncology, University of Minnesota, Minneapolis, MN, USA
| | - Kieuhoa T Vo
- Department of Pediatrics, UCSF Benioff Children's Hospital and UCSF School of Medicine, San Francisco, CA, USA
| | - Mary S Huang
- Department of Pediatric Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Alison M Friedmann
- Department of Pediatric Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Lisa R Diller
- Department of Pediatrics, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | - Karen J Marcus
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Shannon M MacDonald
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Steve E Braunstein
- Department of Radiation Oncology, University of California at San Francisco, UCSF Benioff Children's Hospital, San Francisco, CA, USA
| | - Christine E Hill-Kayser
- Department of Radiation Oncology, University of Pennsylvania and the Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Daphne A Haas-Kogan
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
38
|
Olsen TK, Otte J, Mei S, Embaie BT, Kameneva P, Cheng H, Gao T, Zachariadis V, Tsea I, Björklund Å, Kryukov E, Hou Z, Johansson A, Sundström E, Martinsson T, Fransson S, Stenman J, Fard SS, Johnsen JI, Kogner P, Adameyko I, Enge M, Kharchenko PV, Baryawno N. Joint single-cell genetic and transcriptomic analysis reveal pre-malignant SCP-like subclones in human neuroblastoma. Mol Cancer 2024; 23:180. [PMID: 39217332 PMCID: PMC11365129 DOI: 10.1186/s12943-024-02091-y] [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: 05/18/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Neuroblastoma (NB) is a heterogeneous embryonal malignancy and the deadliest tumor of infancy. It is a complex disease that can result in diverse clinical outcomes. In some children, tumors regress spontaneously. Others respond well to existing treatments. But for the high-risk group, which constitutes approximately 40% of all patients, the prognosis remains dire despite collaborative efforts in basic and clinical research. While its exact cellular origin is still under debate, NB is assumed to arise from the neural crest cell lineage including multipotent Schwann cell precursors (SCPs), which differentiate into sympatho-adrenal cell states eventually producing chromaffin cells and sympathoblasts. METHODS To investigate clonal development of neuroblastoma cell states, we performed haplotype-specific analysis of human tumor samples using single-cell multi-omics, including joint DNA/RNA sequencing of sorted single cells (DNTR-seq). Samples were also assessed using immunofluorescence stainings and fluorescence in-situ hybridization (FISH). RESULTS Beyond adrenergic tumor cells, we identify subpopulations of aneuploid SCP-like cells, characterized by clonal expansion, whole-chromosome 17 gains, as well as expression programs of proliferation, apoptosis, and a non-immunomodulatory phenotype. CONCLUSION Aneuploid pre-malignant SCP-like cells represent a novel feature of NB. Genetic evidence and tumor phylogeny suggest that these clones and malignant adrenergic populations originate from aneuploidy-prone cells of migrating neural crest or SCP origin, before lineage commitment to sympatho-adrenal cell states. Our findings expand the phenotypic spectrum of NB cell states. Considering the multipotency of SCPs in development, we suggest that the transformation of fetal SCPs may represent one possible mechanism of tumor initiation in NB with chromosome 17 aberrations as a characteristic element.
Collapse
Affiliation(s)
- Thale K Olsen
- Department of Women's and Children's Health, Childhood Cancer Research Unit, Karolinska Institutet, Stockholm, 171 77, Sweden
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, 752 36, Sweden
| | - Jörg Otte
- Department of Women's and Children's Health, Childhood Cancer Research Unit, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Shenglin Mei
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02115, USA
| | - Bethel Tesfai Embaie
- Department of Women's and Children's Health, Childhood Cancer Research Unit, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Polina Kameneva
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Huaitao Cheng
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Teng Gao
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02115, USA
| | - Vasilios Zachariadis
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Ioanna Tsea
- Department of Women's and Children's Health, Childhood Cancer Research Unit, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Åsa Björklund
- National Bioinformatics Infrastructure Sweden, SciLifeLab, Uppsala University, Uppsala, 752 36, Sweden
| | - Emil Kryukov
- Department of Ophthalmology, Schepens Eye Research Institute of Massachusetts, Eye and Ear, Boston, MA, 02115, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, 02115, USA
| | - Ziyi Hou
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02115, USA
| | - Anna Johansson
- National Bioinformatics Infrastructure Sweden, SciLifeLab, Uppsala University, Uppsala, 752 36, Sweden
| | - Erik Sundström
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Tommy Martinsson
- Department of Laboratory Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, 413 45, Sweden
| | - Susanne Fransson
- Department of Laboratory Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, 413 45, Sweden
| | - Jakob Stenman
- Department of Women's and Children's Health, Childhood Cancer Research Unit, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Shahrzad Shirazi Fard
- Department of Women's and Children's Health, Childhood Cancer Research Unit, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - John Inge Johnsen
- Department of Women's and Children's Health, Childhood Cancer Research Unit, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Per Kogner
- Department of Women's and Children's Health, Childhood Cancer Research Unit, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Igor Adameyko
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, 171 77, Sweden
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, 1090, Austria
| | - Martin Enge
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, 171 77, Sweden.
| | - Peter V Kharchenko
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02115, USA.
- San Diego Institute of Science, Altos Labs, San Diego, CA, 94022, USA.
| | - Ninib Baryawno
- Department of Women's and Children's Health, Childhood Cancer Research Unit, Karolinska Institutet, Stockholm, 171 77, Sweden.
| |
Collapse
|
39
|
Liu X, Zhou C, Cheng B, Xiong Y, Zhou Q, Wan E, He Y. Genipin promotes the apoptosis and autophagy of neuroblastoma cells by suppressing the PI3K/AKT/mTOR pathway. Sci Rep 2024; 14:20231. [PMID: 39215133 PMCID: PMC11364629 DOI: 10.1038/s41598-024-71123-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: 12/11/2023] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
This study investigated the underlying function and mechanism of genipin in neuroblastoma (NB). Using flow cytometry analysis and cytotoxicity tests, in vitro studies were conducted to assess the effects of genipin on the SK-N-SH cell line. The mechanism of action of genipin was explored through immunofluorescence staining, Western blotting, and caspase-3 activity assays. In addition, we also created a xenograft tumour model to investigate the effects of genipin in vivo. This research confirmed that genipin suppressed cell viability, induced apoptosis, and promoted autophagy, processes that are likely linked to the inhibition of the PI3K/AKT/mTOR signalling pathway. Autophagy inhibition increases the sensitivity of SK-N-SH cells to genipin. Furthermore, combination treatment with a PI3K inhibitor enhanced the therapeutic efficacy of genipin. These results highlight the potential of genipin as a candidate drug for the treatment of NB.
Collapse
Affiliation(s)
- Xinying Liu
- Department of Paediatrics, Affiliated Hospital of North Sichuan Medical College, No. 1 Maoyuan South Road, Shunqing District, Nanchong, 637000, Sichuan, China
- Science and Technology Innovation Centre, North Sichuan Medical College, Shunqing District, Nanchong, 637000, Sichuan, China
- Institute of Hepatobiliary Research, North Sichuan Medical College, Shunqing District, Nanchong, 637000, Sichuan, China
| | - Can Zhou
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Shunqing District, Nanchong, 637000, Sichuan, China
| | - Boli Cheng
- Department of Paediatrics, Affiliated Hospital of North Sichuan Medical College, No. 1 Maoyuan South Road, Shunqing District, Nanchong, 637000, Sichuan, China
| | - Yan Xiong
- Department of Paediatrics, Affiliated Hospital of North Sichuan Medical College, No. 1 Maoyuan South Road, Shunqing District, Nanchong, 637000, Sichuan, China
| | - Qin Zhou
- Department of Paediatrics, Affiliated Hospital of North Sichuan Medical College, No. 1 Maoyuan South Road, Shunqing District, Nanchong, 637000, Sichuan, China
| | - Enyu Wan
- Department of Paediatrics, Affiliated Hospital of North Sichuan Medical College, No. 1 Maoyuan South Road, Shunqing District, Nanchong, 637000, Sichuan, China
| | - Yun He
- Department of Paediatrics, Affiliated Hospital of North Sichuan Medical College, No. 1 Maoyuan South Road, Shunqing District, Nanchong, 637000, Sichuan, China.
| |
Collapse
|
40
|
Chang S, Ren D, Zhang L, Liu S, Yang W, Cheng H, Zhang X, Hong E, Geng D, Wang Y, Chen C, Zhang J, Shi T, Guo Y, Ni X, Wang H, Jin Y. Therapeutic SHPRH-146aa encoded by circ-SHPRH dynamically upregulates P21 to inhibit CDKs in neuroblastoma. Cancer Lett 2024; 598:217120. [PMID: 39002691 DOI: 10.1016/j.canlet.2024.217120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/03/2024] [Accepted: 07/10/2024] [Indexed: 07/15/2024]
Abstract
Recent research has underscored the significance of circular RNAs (circRNAs) in various cancers, including neuroblastoma (NB). Specifically, circ-SHPRH, a unique circRNA, has been revealed to inhibit tumor growth by sequestering miRNAs or producing the SHPRH-146aa protein. To explore circ-SHPRH's involvement in NB and its potential application in gene therapy, this study examined circ-SHPRH expression in 94 NB tissues and cell lines (SK-N-BE(2), SH-SY5Y) using real-time PCR and fluorescence in situ hybridization (FISH). Functional assays encompassing both overexpression and knockdown experiments in NB cell lines, as well as in vivo investigations, were conducted. RNA-seq analysis revealed a correlation between circ-SHPRH and the pathway of P21 (CDKN1A), a pivotal cell cycle regulator. Validation through PCR and other techniques confirmed that circ-SHPRH upregulated P21 expression. Furthermore, the regulatory role of circ-SHPRH in the P21-CDK pathway was corroborated through SHPRH-146aa expression analysis. Notably, adenovirus-mediated circ-SHPRH overexpression effectively curbed NB tumor growth in NSG mice, while combining circ-SHPRH with everolimus exhibited potential for NB treatment. This study elucidates the remarkable significance of circ-SHPRH in NB and its prospective utility in gene therapy, thereby paving the way for innovative therapeutic approaches.
Collapse
Affiliation(s)
- Saishuo Chang
- Department of Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Dong Ren
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Li Zhang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shan Liu
- Department of Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Wei Yang
- Department of Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Haiyan Cheng
- Department of Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Xuexi Zhang
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Enyu Hong
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Di Geng
- Biobank for Clinical Data and Samples in Pediatrics, Beijing Children's Hospital, National Center for Children's Health, Beijing Pediatric Research Institute, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Yadi Wang
- Biobank for Clinical Data and Samples in Pediatrics, Beijing Children's Hospital, National Center for Children's Health, Beijing Pediatric Research Institute, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Chenghao Chen
- Department of Thoracic Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Jie Zhang
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Tieliu Shi
- Center for Bioinformatics and Computational Biology and the Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yongli Guo
- Department of Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Xin Ni
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China; Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China.
| | - Huanmin Wang
- Department of Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China.
| | - Yaqiong Jin
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China.
| |
Collapse
|
41
|
Zhu D, Ren X, Xie W, Chen J, Liang S, Jiang M, Wang J, Zheng Z. Potential of gamma/delta T cells for solid tumor immunotherapy. Front Immunol 2024; 15:1466266. [PMID: 39253082 PMCID: PMC11381238 DOI: 10.3389/fimmu.2024.1466266] [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: 07/17/2024] [Accepted: 08/06/2024] [Indexed: 09/11/2024] Open
Abstract
Gamma/delta T (γδ T)cells possess a unique mechanism for killing tumors, making them highly promising and distinguished among various cell therapies for tumor treatment. This review focuses on the major histocompatibility complex (MHC)-independent recognition of antigens and the interaction between γδ T cells and solid tumor cells. A comprehensive review is provided regarding the classification of human gamma-delta T cell subtypes, the characteristics and mechanisms underlying their functions, as well as their r545egulatory effects on tumor cells. The involvement of γδ T cells in tumorigenesis and migration was also investigated, encompassing potential therapeutic targets such as apoptosis-related molecules, the TNF receptor superfamily member 6(FAS)/FAS Ligand (FASL) pathways, butyrophilin 3A-butyrophilin 2A1 (BTN3A-BTN2A1) complexes, and interactions with CD4, CD8, and natural killer (NK) cells. Additionally, immune checkpoint inhibitors such as programmed cell death protein 1/Programmed cell death 1 ligand 1 (PD-1/PD-L1) have the potential to augment the cytotoxicity of γδ T cells. Moreover, a review on gamma-delta T cell therapy products and their corresponding clinical trials reveals that chimeric antigen receptor (CAR) gamma-delta T therapy holds promise as an approach with encouraging preclinical outcomes. However, practical issues pertaining to manufacturing and clinical aspects need resolution, and further research is required to investigate the long-term clinical side effects of CAR T cells. In conclusion, more comprehensive studies are necessary to establish standardized treatment protocols aimed at enhancing the quality of life and survival rates among tumor patients utilizing γδ T cell immunotherapy.
Collapse
Affiliation(s)
- Dantong Zhu
- Oncology Department, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Xijing Ren
- Oncology Department, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
- School of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Wanting Xie
- Nursing Department, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Jianjun Chen
- Oncology Department, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Shiying Liang
- Oncology Department, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
- School of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Mingzhe Jiang
- Oncology Department, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Junyi Wang
- Oncology Department, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Zhendong Zheng
- Oncology Department, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| |
Collapse
|
42
|
Ma Y, He C, Lin W, Wang J, Xu C, Pan D, Wang Z, Yao W, Dong R, Jia D, Li K. CAMK2G Promotes Neuronal Differentiation and Inhibits Migration in Neuroblastoma. J Pediatr Surg 2024:161679. [PMID: 39266386 DOI: 10.1016/j.jpedsurg.2024.08.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 08/03/2024] [Accepted: 08/05/2024] [Indexed: 09/14/2024]
Abstract
PURPOSE Neuroblastoma (NB) originates from differentiation arrest of sympathoadrenal progenitors in the neural crest. It is necessary to reveal the differentiation mechanism of NB. Previously, we reported that Purkinje cell protein 4 (PCP4) is a well-differentiated marker of NB tissues. Herein, we explored the underlying mechanism of PCP4 induced differentiation in order to find better treatment options for patients. METHODS We screened the interacting proteins of PCP4 by co-immunoprecipitation (Co-IP) and liquid chromatography-mass spectrometry (LC-MS/MS). Then we investigated the relevance between expression of calmodulin-dependent protein kinase II gamma (CAMK2G) and clinical features using R2 platform. We also explored the function of CAMK2G in NB cells by knockdown and RNA sequencing. RESULTS Here, we verified the binding of PCP4 and calmodulin (CaM) by Co-IP and identified a target kinase of CaM, CAMK2G, by LC-MS/MS. PCP4 overexpression activates the autophosphorylation of CAMK2G. Patients with high CAMK2G expression had better survival while low CAMK2G was associated with unfavorable clinical features including MYCN-amplification, unfavorable histology, progression and high INSS stage. CAMK2G knockdown inhibited neurite outgrowth and down-regulated neuronal differentiation markers (NF-H, MAP2), yet promoted migration, invasion and proliferation. Gene Ontology (GO) analysis showed that knockdown of CAMK2G downregulated the expression of neuronal differentiation-related genes. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis showed that knockdown of CAMK2G upregulated the expression of migration-related genes. CONCLUSION These findings indicate that CAMK2G activated by PCP4/CaM complex promotes differentiation and inhibits migration in NB cells. LEVEL OF EVIDENCE Not applicable.
Collapse
Affiliation(s)
- Yujie Ma
- Department of Pediatric Surgery, Children's Hospital of Fudan University, National Children's Medical Center, 399 Wanyuan Road, Shanghai 201102, China
| | - Cong He
- Laboratory of Cancer Genomics and Biology, Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai 201321, China
| | - Weihong Lin
- Department of Pediatric Surgery, Children's Hospital of Fudan University, National Children's Medical Center, 399 Wanyuan Road, Shanghai 201102, China
| | - Jing Wang
- Department of Pediatric Surgery, Children's Hospital of Fudan University, National Children's Medical Center, 399 Wanyuan Road, Shanghai 201102, China
| | - Chaoliang Xu
- Laboratory of Cancer Genomics and Biology, Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Deshen Pan
- Laboratory of Cancer Genomics and Biology, Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Zuopeng Wang
- Department of Pediatric Surgery, Children's Hospital of Fudan University, National Children's Medical Center, 399 Wanyuan Road, Shanghai 201102, China
| | - Wei Yao
- Department of Pediatric Surgery, Children's Hospital of Fudan University, National Children's Medical Center, 399 Wanyuan Road, Shanghai 201102, China
| | - Rui Dong
- Department of Pediatric Surgery, Children's Hospital of Fudan University, National Children's Medical Center, 399 Wanyuan Road, Shanghai 201102, China
| | - Deshui Jia
- Laboratory of Cancer Genomics and Biology, Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Kai Li
- Department of Pediatric Surgery, Children's Hospital of Fudan University, National Children's Medical Center, 399 Wanyuan Road, Shanghai 201102, China.
| |
Collapse
|
43
|
Cheng J, Ji D, Ma J, Zhang Q, Zhang W, Yang L. Proteomic analysis of serum small extracellular vesicles identifies diagnostic biomarkers for neuroblastoma. Front Oncol 2024; 14:1367159. [PMID: 39228987 PMCID: PMC11368728 DOI: 10.3389/fonc.2024.1367159] [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: 01/08/2024] [Accepted: 07/30/2024] [Indexed: 09/05/2024] Open
Abstract
Background Neuroblastoma (NB) primarily arises in children who are <10 years of age, and originates from developing sympathetic nervous system, which results in tumors in adrenal glands and/or sympathetic ganglia. The diagnosis of NB involves a combination of laboratory and imaging tests, and biopsies. Small extracellular vesicles (sEVs) have gained attention as potential biomarkers for various types of tumors. Here, we performed proteomic analysis of serum sEVs and identified potential biomarkers for NB. Methods Label-free proteomics of serum sEVs were performed in the discovery phase. A bulk RNA-seq dataset of NB tissues was used to analyze the association between genes encoding sEVs proteins and prognosis. Potential biomarkers were validated via multiple reaction monitoring (MRM) or western blot analysis in the validation phase. A public single-cell RNA-seq (scRNA-seq) dataset was integrated to analyze the tissue origin of sEVs harboring biomarkers. Results A total of 104 differentially expressed proteins were identified in NB patients with label-free proteomics, and 26 potential biomarkers were validated with MRM analysis. Seven proteins BSG, HSP90AB1, SLC44A1, CHGA, ATP6V0A1, ITGAL and SELL showed the strong ability to distinguish NB patients from healthy controls and non-NB patients as well. Integrated analysis of scRNA-seq and sEVs proteomics revealed that these sEVs-derived biomarkers originated from different cell populations in tumor tissues. Conclusion sEVs-based biomarkers may aid the molecular diagnosis of NB, representing an innovative strategy to improve NB detection and management.
Collapse
Affiliation(s)
- Juan Cheng
- Department of Clinical Laboratory, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Dongrui Ji
- Wayen Biotechnologies (Shanghai), Inc., Shanghai, China
| | - Jing Ma
- Department of Pathology, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qinghua Zhang
- Wayen Biotechnologies (Shanghai), Inc., Shanghai, China
| | - Wanglin Zhang
- Department of Orthopaedics, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lin Yang
- Department of Clinical Laboratory, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
44
|
Mao C, Poimenidou M, Craig BT. Current Knowledge and Perspectives of Immunotherapies for Neuroblastoma. Cancers (Basel) 2024; 16:2865. [PMID: 39199637 PMCID: PMC11353182 DOI: 10.3390/cancers16162865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/02/2024] [Accepted: 08/12/2024] [Indexed: 09/01/2024] Open
Abstract
Neuroblastoma (NBL) cells highly express disialoganglioside GD2, which is restricted and weakly expressed in selected healthy cells, making it a desirable target of immunotherapy. Over the past two decades, application of dinutuximab, an anti-GD2 monoclonal antibody (mAb), has been one of the few new therapies to substantially improve outcomes to current levels. Given the persistent challenge of relapse and therapeutic resistance, there is an urgent need for new effective and tolerable treatment options for high-risk NBL. Recent breakthroughs in immune checkpoint inhibitor (ICI) therapeutics have not translated into high-risk NBL, like many other major pediatric solid tumors. Given the suppressed tumor microenvironment (TME), single ICIs like anti-CTLA4 and anti-PD1 have not demonstrated significant antitumor response rates. Meanwhile, emerging studies are reporting novel advancements in GD2-based therapies, targeted therapies, nanomedicines, and other immunotherapies such as adoptive transfer of natural killer (NK) cells and chimeric antigen receptors (CARs), and these hold interesting promise for the future of high-risk NBL patient care. Herein, we summarize the current state of the art in NBL therapeutic options and highlight the unique challenges posed by NBL that have limited the successful adoption of immune-modifying therapies. Through this review, we aim to direct the field's attention to opportunities that may benefit from a combination immunotherapy strategy.
Collapse
Affiliation(s)
- Chenkai Mao
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
- Center for Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
| | - Maria Poimenidou
- Center for Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
| | - Brian T. Craig
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
- Center for Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
| |
Collapse
|
45
|
Persaud NV, Park JA, Cheung NKV. High-Risk Neuroblastoma Challenges and Opportunities for Antibody-Based Cellular Immunotherapy. J Clin Med 2024; 13:4765. [PMID: 39200906 PMCID: PMC11355836 DOI: 10.3390/jcm13164765] [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: 05/08/2024] [Revised: 07/12/2024] [Accepted: 07/20/2024] [Indexed: 09/02/2024] Open
Abstract
Immunotherapy has emerged as an attractive option for patients with relapsed or refractory high-risk neuroblastoma (HRNB). Neuroblastoma (NB), a sympathetic nervous system cancer arising from an embryonic neural crest cell, is heterogeneous clinically, with outcomes ranging from an isolated abdominal mass that spontaneously regresses to a widely metastatic disease with cure rates of about 50% despite intensive multimodal treatment. Risk group stratification and stage-adapted therapy to achieve cure with minimal toxicities have accomplished major milestones. Targeted immunotherapeutic approaches including monoclonal antibodies, vaccines, adoptive cellular therapies, their combinations, and their integration into standard of care are attractive therapeutic options, although curative challenges and toxicity concerns remain. In this review, we provide an overview of immune approaches to NB and the tumor microenvironment (TME) within the clinical translational framework. We propose a novel T cell-based therapeutic approach that leverages the unique properties of tumor surface antigens such as ganglioside GD2, incorporating specific monoclonal antibodies and recent advancements in adoptive cell therapy.
Collapse
Affiliation(s)
- Natasha V. Persaud
- Department of Pediatrics Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Jeong A. Park
- Pediatrics Inha University Hospital, Icheon 22332, Republic of Korea;
| | - Nai Kong V. Cheung
- Department of Pediatrics Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| |
Collapse
|
46
|
Zhou J, Li Q, Deng X, Peng L, Sun J, Zhang Y, Du Y. Comprehensive analysis identifies ubiquitin ligase FBXO42 as a tumor-promoting factor in neuroblastoma. Sci Rep 2024; 14:18697. [PMID: 39134694 PMCID: PMC11319589 DOI: 10.1038/s41598-024-69760-2] [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: 01/14/2024] [Accepted: 08/08/2024] [Indexed: 08/15/2024] Open
Abstract
Neuroblastoma, the deadliest solid tumor in children, exhibits alarming mortality rates, particularly among high-risk cases. To enhance survival rates, a more precise risk stratification for patients is imperative. Utilizing proteomic data from 34 cases with or without N-Myc amplification, we identified 28 differentially expressed ubiquitination-related proteins (URGs). From these, a prognostic signature comprising 6 URGs was constructed. A nomogram incorporating clinical-pathological parameters yielded impressive AUC values of 0.88, 0.93, and 0.95 at 1, 3, and 5 years, respectively. Functional experiments targeting the E3 ubiquitin ligase FBXO42, a component of the prognostic signature, revealed its TP53-dependent promotion of neuroblastoma cell proliferation. In conclusion, our ubiquitination-related prognostic model robustly predicts patient outcomes, guiding clinical decisions. Additionally, the newfound pro-proliferative role of FBXO42 offers a novel foundation for understanding the molecular mechanisms of neuroblastoma.
Collapse
Affiliation(s)
- Jianwu Zhou
- Department of Pediatric Surgical Oncology, Children's Hospital of Chongqing Medical University; and the 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, 400014, People's Republic of China
| | - Qijun Li
- Laboratory Animal Center, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Xiaobin Deng
- Department of Pediatric Surgical Oncology, Children's Hospital of Chongqing Medical University; and the 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, 400014, People's Republic of China
| | - Liang Peng
- Department of Pediatric Surgical Oncology, Children's Hospital of Chongqing Medical University; and the 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, 400014, People's Republic of China
| | - Jian Sun
- Department of Pediatric Surgical Oncology, Children's Hospital of Chongqing Medical University; and the 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, 400014, People's Republic of China
| | - Yao Zhang
- Department of Pediatric Surgical Oncology, Children's Hospital of Chongqing Medical University; and the 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, 400014, People's Republic of China
| | - Yifei Du
- Department of Pediatric Surgical Oncology, Children's Hospital of Chongqing Medical University; and the 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, 400014, People's Republic of China.
| |
Collapse
|
47
|
Zhang Y. LncRNA-encoded peptides in cancer. J Hematol Oncol 2024; 17:66. [PMID: 39135098 PMCID: PMC11320871 DOI: 10.1186/s13045-024-01591-0] [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: 06/28/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024] Open
Abstract
Long non-coding RNAs (lncRNAs), once considered transcriptional noise, have emerged as critical regulators of gene expression and key players in cancer biology. Recent breakthroughs have revealed that certain lncRNAs can encode small open reading frame (sORF)-derived peptides, which are now understood to contribute to the pathogenesis of various cancers. This review synthesizes current knowledge on the detection, functional roles, and clinical implications of lncRNA-encoded peptides in cancer. We discuss technological advancements in the detection and validation of sORFs, including ribosome profiling and mass spectrometry, which have facilitated the discovery of these peptides. The functional roles of lncRNA-encoded peptides in cancer processes such as gene transcription, translation regulation, signal transduction, and metabolic reprogramming are explored in various types of cancer. The clinical potential of these peptides is highlighted, with a focus on their utility as diagnostic biomarkers, prognostic indicators, and therapeutic targets. The challenges and future directions in translating these findings into clinical practice are also discussed, including the need for large-scale validation, development of sensitive detection methods, and optimization of peptide stability and delivery.
Collapse
Affiliation(s)
- Yaguang Zhang
- Laboratory of Gastrointestinal Tumor Epigenetics and Genomics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
48
|
Zhang L, Mo J, Shi H, Xiong J, Aierken Y, Chen F, Tang Y, Zhao K, Lv Z, Tan K. CRISPR-Cas9 screening develops an epigenetic and transcriptional gene signature for risk stratification and target prediction in neuroblastoma. Front Cell Dev Biol 2024; 12:1433008. [PMID: 39175876 PMCID: PMC11338898 DOI: 10.3389/fcell.2024.1433008] [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: 05/15/2024] [Accepted: 07/22/2024] [Indexed: 08/24/2024] Open
Abstract
Objectives: Neuroblastoma (NB), a pediatric malignancy of the peripheral nervous system, is characterized by epigenetic and transcriptional (EP-TF) anomalies. This study aimed to develop an EP-TF clinical prognostic model for NB using CRISPR-Cas9 knockout screening. Results: An integrative analysis was conducted using CRISPR-Cas9 screening in vitro and in vivo with public NB datasets to identify 35 EP-TF genes that exhibited the highest expression in NB and were highly dependent on cancer viability. After univariate analysis, 27 of these 35 genes were included in the least absolute shrinkage and selection operator screen. We established and biologically validated a prognostic EP-TF model encompassing RUVBL1, LARP7, GTF3C4, THAP10, SUPT16H, TIGD1, SUV39H2, TAF1A, SMAD9, and FEM1B across diverse NB cohorts. MYCN serves a potential upstream regulator of EP-TF genes. The high-risk subtype exhibited traits associated with the malignant cell cycle, MYCN-linked signaling and chromatin remodeling, all of which are correlated with poor prognosis and immunosuppression. MEK inhibitors have emerged as promising therapeutic agents for targeting most EP-TF risk genes in NB. Conclusion: Our novel prognostic model shows significant potential for predicting and evaluating the overall survival of NB patients, offering insights into therapeutic targets.
Collapse
Affiliation(s)
- Liaoran Zhang
- Department of General Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jialin Mo
- Shanghai Key Laboratory of Reproductive Medicine, Department of Histoembryology, Genetics and Developmental Biology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hao Shi
- Department of General Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Xiong
- Department of General Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yeerfan Aierken
- Department of General Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Feng Chen
- Department of General Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yujie Tang
- Shanghai Key Laboratory of Reproductive Medicine, Department of Histoembryology, Genetics and Developmental Biology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kewen Zhao
- State Key Laboratory of Oncogenes and Related Genes, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Department of Pathophysiology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhibao Lv
- Department of General Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kezhe Tan
- Department of General Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
49
|
Janssen FW, Lak NSM, Janda CY, Kester LA, Meister MT, Merks JHM, van den Heuvel-Eibrink MM, van Noesel MM, Zsiros J, Tytgat GAM, Looijenga LHJ. A comprehensive overview of liquid biopsy applications in pediatric solid tumors. NPJ Precis Oncol 2024; 8:172. [PMID: 39097671 PMCID: PMC11297996 DOI: 10.1038/s41698-024-00657-z] [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: 02/20/2024] [Accepted: 07/15/2024] [Indexed: 08/05/2024] Open
Abstract
Liquid biopsies are emerging as an alternative source for pediatric cancer biomarkers with potential applications during all stages of patient care, from diagnosis to long-term follow-up. While developments within this field are reported, these mainly focus on dedicated items such as a specific liquid biopsy matrix, analyte, and/or single tumor type. To the best of our knowledge, a comprehensive overview is lacking. Here, we review the current state of liquid biopsy research for the most common non-central nervous system pediatric solid tumors. These include neuroblastoma, renal tumors, germ cell tumors, osteosarcoma, Ewing sarcoma, rhabdomyosarcoma and other soft tissue sarcomas, and liver tumors. Within this selection, we discuss the most important or recent studies involving liquid biopsy-based biomarkers, anticipated clinical applications, and the current challenges for success. Furthermore, we provide an overview of liquid biopsy-based biomarker publication output for each tumor type based on a comprehensive literature search between 1989 and 2023. Per study identified, we list the relevant liquid biopsy-based biomarkers, matrices (e.g., peripheral blood, bone marrow, or cerebrospinal fluid), analytes (e.g., circulating cell-free and tumor DNA, microRNAs, and circulating tumor cells), methods (e.g., digital droplet PCR and next-generation sequencing), the involved pediatric patient cohort, and proposed applications. As such, we identified 344 unique publications. Taken together, while the liquid biopsy field in pediatric oncology is still behind adult oncology, potentially relevant publications have increased over the last decade. Importantly, steps towards clinical implementation are rapidly gaining ground, notably through validation of liquid biopsy-based biomarkers in pediatric clinical trials.
Collapse
Affiliation(s)
| | | | | | | | - Michael T Meister
- Princess Máxima Center, Utrecht, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Johannes H M Merks
- Princess Máxima Center, Utrecht, the Netherlands
- Division of Imaging and Oncology, University Medical Center Utrecht, University of Utrecht, Utrecht, the Netherlands
| | - Marry M van den Heuvel-Eibrink
- Princess Máxima Center, Utrecht, the Netherlands
- Wilhelmina Children's Hospital-Division of CHILDHEALTH, University Medical Center Utrech, University of Utrecht, Utrecht, the Netherlands
| | - Max M van Noesel
- Princess Máxima Center, Utrecht, the Netherlands
- Division of Imaging and Oncology, University Medical Center Utrecht, University of Utrecht, Utrecht, the Netherlands
| | | | - Godelieve A M Tytgat
- Princess Máxima Center, Utrecht, the Netherlands
- Department of Genetics, University Medical Center Utrecht, University of Utrecht, Utrecht, the Netherlands
| | - Leendert H J Looijenga
- Princess Máxima Center, Utrecht, the Netherlands.
- Department of Pathology, University Medical Center Utrecht, University of Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
50
|
Nri-Ezedi CA, Ulasi TO, Menkiti FE, Ndukwe CO, Igwenagu CM, Akpuaka FC. Epidemiological trends and treatment abandonment of paediatric solid tumours in a nigerian tertiary hospital: a seven-year review (2016-2022). BMC Cancer 2024; 24:943. [PMID: 39095784 PMCID: PMC11295635 DOI: 10.1186/s12885-024-12723-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Paediatric solid tumours, both benign and malignant, present significant health challenges, particularly in Sub-Saharan Africa where comprehensive data is limited. This study aims to elucidate the prevalence, distribution, and treatment outcomes of paediatric solid neoplasms in a tertiary hospital in South-East Nigeria over a seven-year period. METHODS A retrospective cohort study was conducted at Nnamdi Azikiwe University Teaching Hospital (NAUTH), Nnewi, Nigeria. Clinical details and histological slides of confirmed cases from January 2016 to December 2022 were reviewed. Data extraction focused on socio-demographic variables and treatment outcomes, analysed using statistical methods. RESULTS The study included 293 children diagnosed with solid tumours (58.1% malignant, 41.9% benign), with a female predominance (61.8%). The median age at diagnosis was 12 years. Fibroadenoma was the most common benign tumour (61.8% of benign cases), while non-Hodgkin lymphoma was the predominant malignant tumour (18.2% of malignant cases). Treatment abandonment rates differed significantly between benign (13.8%) and malignant (51.2%) tumours. Significant associations were found between treatment outcomes and factors such as gender (p = 0.0001 for benign tumours), age category (p = 0.0001 for benign tumours), and specific diagnoses (p = 0.0001 for both benign and malignant tumours). CONCLUSION This study underscores the substantial burden of paediatric solid tumours in South-East Nigeria and highlights the critical need for improved treatment adherence strategies, particularly for malignant cases. The findings emphasize the importance of tailored interventions based on tumour type, age, and gender. These insights can inform future research, policy formulation, and healthcare strategies aimed at enhancing the management and outcomes of paediatric solid neoplasms in resource-limited settings.
Collapse
Affiliation(s)
| | | | - Felix Emeka Menkiti
- Department of Anatomic Pathology and Forensic Medicine, Nnamdi Azikiwe University, Awka, Anambra, Nigeria.
| | - Chinedu Onwuka Ndukwe
- Department of Anatomic Pathology and Forensic Medicine, Nnamdi Azikiwe University, Awka, Anambra, Nigeria
| | - Chinelo M Igwenagu
- Department of Industrial Mathematics and Applied Statistics, Enugu State University of Science and Technology, Enugu, Nigeria
| | - Frank Chinedu Akpuaka
- Department of Surgery, Chukwuemeka Odumegwu Ojukwu University, Igbariam, Anambra, Nigeria
| |
Collapse
|