1
|
Wang SM, Kong XY, Zhao DQ, Li M. Association analysis of genetic polymorphisms of METTL3 with clinical outcomes in a Chinese pediatric population with primary brain tumors. Gene 2025; 941:149232. [PMID: 39800197 DOI: 10.1016/j.gene.2025.149232] [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: 10/28/2024] [Revised: 01/04/2025] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
BACKGROUND Methyltransferase-like 3 (METTL3) regulates numerous biological processes and diverse cancers. OBJECTIVE To explore the frequency distribution of METTL3 rs1061026, rs1139130, and rs1263801 polymorphisms, and their potential impacts on clinical outcomes and chemotherapy-induced toxicities in a cohort of Chinese pediatric patients diagnosed with primary brain tumors (PBTs). METHODS Genotyping for three investigated SNPs was performed in 107 pediatric patients with PBTs using the Sequenom MassARRAY iPLEX platform. Serum METTL3 levels were determined by Enzyme-Linked Immunosorbent Assay. Serum methotrexate (MTX) concentrations were quantified utilizing fluorescence polarization immunoassay. RESULTS The three investigated SNPs were not significantly associated with the risks of relapse and metastasis after adjusting all confounders. Compared to individuals with the rs1139130 GG genotype, GA genotype carriers exhibited a significantly higher risk of oral mucositis (adjusted OR: 7.504; 95 % CI, 1.931-29.436; P = 0.004). The rs1139130 GA (adjusted OR: 5.091; 95 % CI, 1.351-19.176; P = 0.016) and AA (adjusted OR: 9.588; 95 % CI, 1.769-51.949; P = 0.009) genotype carriers exhibited a significantly lower risk of fever than GG genotype carriers. The median dose-normalized MTX concentrations at 42 h were lower with borderline significance in children with rs1061026 GT and GG genotypes (0.004 μmol/L per g/m2) than the TT genotype carriers (0.006 μmol/L per g/m2, P = 0.048). Patients with the rs1139130 GA genotype had significantly higher median serum METTL3 protein levels (59.91 ng/mL) than GG genotype carriers (44.57 ng/mL, P = 0.015). CONCLUSION This study demonstrated the association of the rs1139130 polymorphism with the development of oral mucositis and fever and the rs1061026 polymorphism with MTX exposure.
Collapse
Affiliation(s)
- Shu-Mei Wang
- Department of Pharmacy, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China.
| | - Xiao-Yan Kong
- Department of Pharmacy, Armed Police Beijing Corps Hospital, Beijing, China
| | - Dan-Qi Zhao
- Department of Pharmacy, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China; Department of Clinical Pharmacy, College of Pharmacy, Capital Medical University, Beijing 100069, China
| | - Miao Li
- Department of Pediatrics, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China.
| |
Collapse
|
2
|
Varga P, Obeidat M, Máté V, Kói T, Kiss-Dala S, Major GS, Tímár ÁE, Li X, Szilágyi Á, Csáki Z, Engh MA, Garami M, Hegyi P, Túri I, Tuboly E. From simple factors to artificial intelligence: evolution of prognosis prediction in childhood cancer: a systematic review and meta-analysis. EClinicalMedicine 2024; 78:102902. [PMID: 39640942 PMCID: PMC11617957 DOI: 10.1016/j.eclinm.2024.102902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 12/07/2024] Open
Abstract
Background Current paediatric cancer care requires innovative approaches to predict prognosis that facilitates personalised stratification, yet studies on the performance, composition and limitations of contemporary prognostic models are lacking. We aimed to compare the accuracy of traditional and advanced prognostic models. Methods A systematic search for this systematic review and meta-analysis (CRTN42022370251) was conducted in PubMed, Embase, Scopus, and the Cochrane Library databases on 28 June 2024. Studies on the accuracy of prognostic markers or models used in paediatric haematological malignancies, central nervous system (CNS), or non-CNS solid tumours (NCNSST) were included. Three model categories were defined using: 1-clinical parameters, 2-genomic-transcriptomic data, and 3-artificial intelligence (AI). Primary outcomes were area under the receiver operating characteristic curve with a 95% confidence interval (CI) for various overall survival intervals and event-free survival. Two independent groups performed selection and data extraction. We used data published by the authors and publicly available databases. Findings Of 12,982 studies, 358 were included in the meta-analysis and 27 in the systematic review, with limited data on AI-approaches. Most data were reported on NCNSST at 5-year OS, where a statistically significant difference was observed between Category-1 (0.75 CI: 0.72-0.79) and Category-2 (0.85 CI: 0.82-0.88) (p < 0.001), but not between Categories-2 and -3 (p = 0.2834) (0.82 CI: 0.77-0.88). Internal validation studies showed significantly better performance compared to those using external validation, highlighting the high risk of bias (ROB) inherent in internal validation. High ROB was most commonly experienced in the outcomes and statistical analysis domains, assessed using PROBAST and QUIPS. Interpretation It is advisable to introduce Category-2 and -3 models in a clinical setting, especially for NCNSST prognostic for aiding risk-stratification. Although AI-supported predictions in paediatric oncology are at an early stage of development, it is imperative to further explore their potential. This requires structured data collection and ethical sharing from paediatric oncology patients in sufficient quantity and quality. Funding None.
Collapse
Affiliation(s)
- Petra Varga
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Heim Pál National Pediatric Institute, Budapest, Hungary
| | - Mahmoud Obeidat
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Vanda Máté
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Pediatric Center, Semmelweis University, Budapest, Hungary
| | - Tamás Kói
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Department of Stochastics, Institute of Mathematics, Budapest University of Technology and Economics, Budapest, Hungary
| | - Szilvia Kiss-Dala
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Gréta Szilvia Major
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Heim Pál National Pediatric Institute, Budapest, Hungary
| | - Ágnes Eszter Tímár
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Heim Pál National Pediatric Institute, Budapest, Hungary
| | - Ximeng Li
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Ádám Szilágyi
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Zsófia Csáki
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Marie Anne Engh
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Miklós Garami
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Pediatric Center, Semmelweis University, Budapest, Hungary
| | - Péter Hegyi
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Institute of Pancreatic Diseases, Semmelweis University, Budapest, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Ibolya Túri
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Pető András Faculty, Semmelweis University, Budapest, Hungary
| | - Eszter Tuboly
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Hungarian Pediatric Oncology Network, Budapest, Hungary
| |
Collapse
|
3
|
Kumar S, Arwind DA, Kumar B H, Pandey S, Nayak R, Vithalkar MP, Kumar N, Pai KSR. Inhibition of STAT3: A promising approach to enhancing the efficacy of chemotherapy in medulloblastoma. Transl Oncol 2024; 46:102023. [PMID: 38852276 PMCID: PMC11220551 DOI: 10.1016/j.tranon.2024.102023] [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: 02/03/2024] [Revised: 05/27/2024] [Accepted: 06/01/2024] [Indexed: 06/11/2024] Open
Abstract
Medulloblastoma is a type of brain cancer that primarily affects children. While chemotherapy has been shown to be effective in treating medulloblastoma, the development of chemotherapy resistance remains a challenge. One potential therapeutic approach is to selectively inhibit the inducible transcription factor called STAT3, which is known to play a crucial role in the survival and growth of tumor cells. The activation of STAT3 has been linked to the growth and progression of various cancers, including medulloblastoma. Inhibition of STAT3 has been shown to sensitize medulloblastoma cells to chemotherapy, leading to improved treatment outcomes. Different approaches to STAT3 inhibition have been developed, including small-molecule inhibitors and RNA interference. Preclinical studies have shown the efficacy of STAT3 inhibitors in medulloblastoma, and clinical trials are currently ongoing to evaluate their safety and effectiveness in patients with various solid tumors, including medulloblastoma. In addition, researchers are also exploring ways to optimize the use of STAT3 inhibitors in combination with chemotherapy and identify biomarkers that can predict treatment that will help to develop personalized treatment strategies. This review highlights the potential of selective inhibition of STAT3 as a novel approach for the treatment of medulloblastoma and suggests that further research into the development of STAT3 inhibitors could lead to improved outcomes for patients with aggressive cancer.
Collapse
Affiliation(s)
- Sachindra Kumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, India
| | - Dube Aakash Arwind
- Department of Pharmacology and toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Vaishali-844102, Bihar, India
| | - Harish Kumar B
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, India
| | - Samyak Pandey
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, India
| | - Raksha Nayak
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, India
| | - Megh Pravin Vithalkar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, India
| | - Nitesh Kumar
- Department of Pharmacology and toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Vaishali-844102, Bihar, India
| | - K Sreedhara Ranganath Pai
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, India.
| |
Collapse
|
4
|
Minasi S, Gianno F, Bargiacchi L, Barresi V, Miele E, Antonelli M, Buttarelli FR. Case report of a pediatric medulloblastoma with concurrent MYC and MYCN subclonal amplification in distinct populations of neoplastic cells. Virchows Arch 2024; 485:153-158. [PMID: 37212894 DOI: 10.1007/s00428-023-03560-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 03/17/2023] [Accepted: 05/10/2023] [Indexed: 05/23/2023]
Abstract
Medulloblastomas (MDBs) are classified into molecular groups showing peculiar immunohistochemical and genetic features and distinct DNA methylation profile. Group 3 and group 4 MDBs have the worst prognosis; the former is treated with high-risk protocols and features MYC amplification, whereas the latter receives standard-risk protocols and harbors MYCN amplification. Herein, we report a unique case of MDB showing histological and immunohistochemical features consistent with non-SHH/non-WNT classic MDB, with both MYCN (30% of tumor cells) and MYC (5-10% tumor cells) amplification in distinct subclones of neoplastic cells at fluorescence in situ hybridization (FISH), characterized by specific patterns. In spite of MYC amplification in only a small percentage of tumor cells, this case had DNA methylation profile consistent with group 3, emphasizing the importance to test both MYC and MYCN amplifications at a single cell level using highly sensitive methods, such as FISH, for diagnostic and therapeutic purposes.
Collapse
Affiliation(s)
- Simone Minasi
- Department of Radiological, Oncological and Anatomo-Pathological Sciences, "Sapienza" University of Rome, Viale Regina Elena, 324-00161, Rome, Italy.
| | - Francesca Gianno
- Department of Radiological, Oncological and Anatomo-Pathological Sciences, "Sapienza" University of Rome, Viale Regina Elena, 324-00161, Rome, Italy
| | - Lavinia Bargiacchi
- Department of Radiological, Oncological and Anatomo-Pathological Sciences, "Sapienza" University of Rome, Viale Regina Elena, 324-00161, Rome, Italy
| | - Valeria Barresi
- Department of Diagnostics and Public Health, Section of Anatomic Pathology, University of Verona, Verona, Italy
| | - Evelina Miele
- Department of Oncology/Hematology, Gene and Cell Therapy and Hemopoietic Transplant, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Manila Antonelli
- Department of Radiological, Oncological and Anatomo-Pathological Sciences, "Sapienza" University of Rome, Viale Regina Elena, 324-00161, Rome, Italy
| | - Francesca Romana Buttarelli
- Department of Radiological, Oncological and Anatomo-Pathological Sciences, "Sapienza" University of Rome, Viale Regina Elena, 324-00161, Rome, Italy
| |
Collapse
|
5
|
Shahab SW, Roggeveen CM, Sun J, Kunhiraman H, McSwain LF, Juraschka K, Kumar SA, Saulnier O, Taylor MD, Schniederjan M, Schnepp RW, MacDonald TJ, Kenney AM. The LIN28B-let-7-PBK pathway is essential for group 3 medulloblastoma tumor growth and survival. Mol Oncol 2023; 17:1784-1802. [PMID: 37341142 PMCID: PMC10483609 DOI: 10.1002/1878-0261.13477] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/28/2023] [Accepted: 06/19/2023] [Indexed: 06/22/2023] Open
Abstract
Children with Group 3 medulloblastoma (G3 MB) have a very poor prognosis, and many do not survive beyond 5 years after diagnosis. A factor that may contribute to this is the lack of available targeted therapy. Expression of protein lin-28 homolog B (LIN28B), a regulator of developmental timing, is upregulated in several cancers, including G3 MB, and is associated with worse survival in this disease. Here, we investigate the role of the LIN28B pathway in G3 MB and demonstrate that the LIN28B-lethal-7 (let-7; a microRNA that is a tumor suppressor)-lymphokine-activated killer T-cell-originated protein kinase (PBK; also known as PDZ-binding kinase) axis promotes G3 MB proliferation. LIN28B knockdown in G3-MB-patient-derived cell lines leads to a significant reduction in cell viability and proliferation in vitro and in prolonged survival of mice with orthotopic tumors. The LIN28 inhibitor N-methyl-N-[3-(3-methyl-1,2,4-triazolo[4,3-b]pyridazin-6-yl)phenyl]acetamide (1632) significantly reduces G3 MB cell growth and demonstrates efficacy in reducing tumor growth in mouse xenograft models. Inhibiting PBK using HI-TOPK-032 also results in a significant reduction in G3 MB cell viability and proliferation. Together, these results highlight a critical role for the LIN28B-let-7-PBK pathway in G3 MB and provide preliminary preclinical results for drugs targeting this pathway.
Collapse
Affiliation(s)
- Shubin W. Shahab
- Aflac Cancer and Blood Disorders CenterChildren's Healthcare of AtlantaGAUSA
- Department of PediatricsEmory University School of MedicineAtlantaGAUSA
| | | | - Jiarong Sun
- Emory College of Arts and SciencesEmory UniversityAtlantaGAUSA
| | | | - Leon F. McSwain
- Department of PediatricsEmory University School of MedicineAtlantaGAUSA
| | - Kyle Juraschka
- Department of Neurosurgery, The Hospital for Sick ChildrenUniversity of TorontoONCanada
- Department of Laboratory Medicine and PathologyUniversity of TorontoONCanada
| | - Sachin A. Kumar
- Department of Laboratory Medicine and PathologyUniversity of TorontoONCanada
| | - Olivier Saulnier
- The Arthur and Sonia Labatt Brain Tumor Research Centre, The Hospital for Sick ChildrenUniversity of TorontoONCanada
- Developmental and Stem Cell Biology Program, The Hospital for Sick ChildrenUniversity of TorontoONCanada
| | - Michael D. Taylor
- Department of Neurosurgery, The Hospital for Sick ChildrenUniversity of TorontoONCanada
- Department of Laboratory Medicine and PathologyUniversity of TorontoONCanada
| | | | - Robert W. Schnepp
- Aflac Cancer and Blood Disorders CenterChildren's Healthcare of AtlantaGAUSA
- Department of PediatricsEmory University School of MedicineAtlantaGAUSA
- The Janssen PharmaceuticalAmblerPAUSA
| | - Tobey J MacDonald
- Aflac Cancer and Blood Disorders CenterChildren's Healthcare of AtlantaGAUSA
- Department of PediatricsEmory University School of MedicineAtlantaGAUSA
- Winship Cancer InstituteAtlantaGAUSA
| | - Anna Marie Kenney
- Department of PediatricsEmory University School of MedicineAtlantaGAUSA
- Winship Cancer InstituteAtlantaGAUSA
| |
Collapse
|
6
|
Ismail M, Craig S, Ahmed R, de Blank P, Tiwari P. Opportunities and Advances in Radiomics and Radiogenomics for Pediatric Medulloblastoma Tumors. Diagnostics (Basel) 2023; 13:2727. [PMID: 37685265 PMCID: PMC10487205 DOI: 10.3390/diagnostics13172727] [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: 07/24/2023] [Revised: 08/19/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
Recent advances in artificial intelligence have greatly impacted the field of medical imaging and vastly improved the development of computational algorithms for data analysis. In the field of pediatric neuro-oncology, radiomics, the process of obtaining high-dimensional data from radiographic images, has been recently utilized in applications including survival prognostication, molecular classification, and tumor type classification. Similarly, radiogenomics, or the integration of radiomic and genomic data, has allowed for building comprehensive computational models to better understand disease etiology. While there exist excellent review articles on radiomics and radiogenomic pipelines and their applications in adult solid tumors, in this review article, we specifically review these computational approaches in the context of pediatric medulloblastoma tumors. Based on our systematic literature research via PubMed and Google Scholar, we provide a detailed summary of a total of 15 articles that have utilized radiomic and radiogenomic analysis for survival prognostication, tumor segmentation, and molecular subgroup classification in the context of pediatric medulloblastoma. Lastly, we shed light on the current challenges with the existing approaches as well as future directions and opportunities with using these computational radiomic and radiogenomic approaches for pediatric medulloblastoma tumors.
Collapse
Affiliation(s)
- Marwa Ismail
- Department of Radiology, University of Wisconsin-Madison, Madison, WI 53706, USA; (S.C.); (P.T.)
| | - Stephen Craig
- Department of Radiology, University of Wisconsin-Madison, Madison, WI 53706, USA; (S.C.); (P.T.)
| | - Raheel Ahmed
- Department of Neurosurgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA;
| | - Peter de Blank
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA;
| | - Pallavi Tiwari
- Department of Radiology, University of Wisconsin-Madison, Madison, WI 53706, USA; (S.C.); (P.T.)
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
7
|
Stepien N, Senfter D, Furtner J, Haberler C, Dorfer C, Czech T, Lötsch-Gojo D, Mayr L, Hedrich C, Baumgartner A, Aliotti-Lippolis M, Schned H, Holler J, Bruckner K, Slavc I, Azizi AA, Peyrl A, Müllauer L, Madlener S, Gojo J. Proof-of-Concept for Liquid Biopsy Disease Monitoring of MYC-Amplified Group 3 Medulloblastoma by Droplet Digital PCR. Cancers (Basel) 2023; 15:2525. [PMID: 37173990 PMCID: PMC10177279 DOI: 10.3390/cancers15092525] [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: 03/04/2023] [Revised: 04/21/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
BACKGROUND Liquid biopsy diagnostic methods are an emerging complementary tool to imaging and pathology techniques across various cancer types. However, there is still no established method for the detection of molecular alterations and disease monitoring in MB, the most common malignant CNS tumor in the pediatric population. In the presented study, we investigated droplet digital polymerase chain reaction (ddPCR) as a highly sensitive method for the detection of MYC amplification in bodily fluids of group 3 MB patients. METHODS We identified a cohort of five MYC-amplified MBs by methylation array and FISH. Predesigned and wet-lab validated probes for ddPCR were used to establish the detection method and were validated in two MYC-amplified MB cell lines as well as tumor tissue of the MYC-amplified cohort. Finally, a total of 49 longitudinal CSF samples were analyzed at multiple timepoints during the course of the disease. RESULTS Detection of MYC amplification by ddPCR in CSF showed a sensitivity and specificity of 90% and 100%, respectively. We observed a steep increase in amplification rate (AR) at disease progression in 3/5 cases. ddPCR was proven to be more sensitive than cytology for the detection of residual disease. In contrast to CSF, MYC amplification was not detectable by ddPCR in blood samples. CONCLUSIONS ddPCR proves to be a sensitive and specific method for the detection of MYC amplification in the CSF of MB patients. These results warrant implementation of liquid biopsy in future prospective clinical trials to validate the potential for improved diagnosis, disease staging and monitoring.
Collapse
Affiliation(s)
- Natalia Stepien
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (N.S.); (S.M.)
| | - Daniel Senfter
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (N.S.); (S.M.)
| | - Julia Furtner
- Division of Neuroradiology and Musculoskeletal Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
- Research Center for Medical Image Analysis and Artificial Intelligence (MIAAI), Faculty of Medicine and Dentistry, Danube Private University, 3500 Krems-Stein, Austria
| | - Christine Haberler
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria
| | - Christian Dorfer
- Department of Neurosurgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Thomas Czech
- Department of Neurosurgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Daniela Lötsch-Gojo
- Department of Neurosurgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Lisa Mayr
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (N.S.); (S.M.)
| | - Cora Hedrich
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (N.S.); (S.M.)
| | - Alicia Baumgartner
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (N.S.); (S.M.)
| | - Maria Aliotti-Lippolis
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (N.S.); (S.M.)
| | - Hannah Schned
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (N.S.); (S.M.)
| | - Johannes Holler
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (N.S.); (S.M.)
| | - Katharina Bruckner
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (N.S.); (S.M.)
| | - Irene Slavc
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (N.S.); (S.M.)
| | - Amedeo A. Azizi
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (N.S.); (S.M.)
| | - Andreas Peyrl
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (N.S.); (S.M.)
| | - Leonhard Müllauer
- Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria
| | - Sibylle Madlener
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (N.S.); (S.M.)
| | - Johannes Gojo
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (N.S.); (S.M.)
| |
Collapse
|
8
|
Katsushima K, Pokhrel R, Mahmud I, Yuan M, Murad R, Baral P, Zhou R, Chapagain P, Garrett T, Stapleton S, Jallo G, Bettegowda C, Raabe E, Wechsler-Reya RJ, Eberhart CG, Perera RJ. The oncogenic circular RNA circ_63706 is a potential therapeutic target in sonic hedgehog-subtype childhood medulloblastomas. Acta Neuropathol Commun 2023; 11:38. [PMID: 36899402 PMCID: PMC10007801 DOI: 10.1186/s40478-023-01521-0] [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: 01/23/2023] [Accepted: 01/24/2023] [Indexed: 03/12/2023] Open
Abstract
Medulloblastoma (MB) develops through various genetic, epigenetic, and non-coding (nc) RNA-related mechanisms, but the roles played by ncRNAs, particularly circular RNAs (circRNAs), remain poorly defined. CircRNAs are increasingly recognized as stable non-coding RNA therapeutic targets in many cancers, but little is known about their function in MBs. To determine medulloblastoma subgroup-specific circRNAs, publicly available RNA sequencing (RNA-seq) data from 175 MB patients were interrogated to identify circRNAs that differentiate between MB subgroups. circ_63706 was identified as sonic hedgehog (SHH) group-specific, with its expression confirmed by RNA-FISH analysis in clinical tissue samples. The oncogenic function of circ_63706 was characterized in vitro and in vivo. Further, circ_63706-depleted cells were subjected to RNA-seq and lipid profiling to identify its molecular function. Finally, we mapped the circ_63706 secondary structure using an advanced random forest classification model and modeled a 3D structure to identify its interacting miRNA partner molecules. Circ_63706 regulates independently of the host coding gene pericentrin (PCNT), and its expression is specific to the SHH subgroup. circ_63706-deleted cells implanted into mice produced smaller tumors, and mice lived longer than parental cell implants. At the molecular level, circ_63706-deleted cells elevated total ceramide and oxidized lipids and reduced total triglyceride. Our study implicates a novel oncogenic circular RNA in the SHH medulloblastoma subgroup and establishes its molecular function and potential as a future therapeutic target.
Collapse
Affiliation(s)
- Keisuke Katsushima
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, 1650 Orleans St., Baltimore, MD, 21231, USA.,Johns Hopkins All Children's Hospital, St. Petersburg, USA
| | - Rudramani Pokhrel
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, 1650 Orleans St., Baltimore, MD, 21231, USA.,Johns Hopkins All Children's Hospital, St. Petersburg, USA
| | - Iqbal Mahmud
- Department Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, USA.,Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, USA
| | - Menglang Yuan
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, 1650 Orleans St., Baltimore, MD, 21231, USA.,Johns Hopkins All Children's Hospital, St. Petersburg, USA
| | - Rabi Murad
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, USA
| | - Prabin Baral
- Department of Physics, Florida International University, Miami, USA
| | - Rui Zhou
- Johns Hopkins All Children's Hospital, St. Petersburg, USA
| | - Prem Chapagain
- Department of Physics, Florida International University, Miami, USA.,Biomolecular Sciences Institute, Florida International University, Miami, USA
| | - Timothy Garrett
- Department Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, USA
| | | | - George Jallo
- Johns Hopkins All Children's Hospital, St. Petersburg, USA.,Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Chetan Bettegowda
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, 1650 Orleans St., Baltimore, MD, 21231, USA.,Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Eric Raabe
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, 1650 Orleans St., Baltimore, MD, 21231, USA.,Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, USA
| | | | - Charles G Eberhart
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, 1650 Orleans St., Baltimore, MD, 21231, USA.,Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Ranjan J Perera
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, 1650 Orleans St., Baltimore, MD, 21231, USA. .,Johns Hopkins All Children's Hospital, St. Petersburg, USA. .,Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, USA.
| |
Collapse
|
9
|
Zou H, Poore B, Brown EE, Qian J, Xie B, Asimakidou E, Razskazovskiy V, Ayrapetian D, Sharma V, Xia S, Liu F, Chen A, Guan Y, Li Z, Wanggou S, Saulnier O, Ly M, Fellows-Mayle W, Xi G, Tomita T, Resnick AC, Mack SC, Raabe EH, Eberhart CG, Sun D, Stronach BE, Agnihotri S, Kohanbash G, Lu S, Herrup K, Rich JN, Gittes GK, Broniscer A, Hu Z, Li X, Pollack IF, Friedlander RM, Hainer SJ, Taylor MD, Hu B. A neurodevelopmental epigenetic programme mediated by SMARCD3-DAB1-Reelin signalling is hijacked to promote medulloblastoma metastasis. Nat Cell Biol 2023; 25:493-507. [PMID: 36849558 PMCID: PMC10014585 DOI: 10.1038/s41556-023-01093-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 01/17/2023] [Indexed: 03/01/2023]
Abstract
How abnormal neurodevelopment relates to the tumour aggressiveness of medulloblastoma (MB), the most common type of embryonal tumour, remains elusive. Here we uncover a neurodevelopmental epigenomic programme that is hijacked to induce MB metastatic dissemination. Unsupervised analyses of integrated publicly available datasets with our newly generated data reveal that SMARCD3 (also known as BAF60C) regulates Disabled 1 (DAB1)-mediated Reelin signalling in Purkinje cell migration and MB metastasis by orchestrating cis-regulatory elements at the DAB1 locus. We further identify that a core set of transcription factors, enhancer of zeste homologue 2 (EZH2) and nuclear factor I X (NFIX), coordinates with the cis-regulatory elements at the SMARCD3 locus to form a chromatin hub to control SMARCD3 expression in the developing cerebellum and in metastatic MB. Increased SMARCD3 expression activates Reelin-DAB1-mediated Src kinase signalling, which results in a MB response to Src inhibition. These data deepen our understanding of how neurodevelopmental programming influences disease progression and provide a potential therapeutic option for patients with MB.
Collapse
Affiliation(s)
- Han Zou
- Xiangya School of Medicine, Central South University, Changsha, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Changsha, China
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- John G. Rangos Sr Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Bradley Poore
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- John G. Rangos Sr Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Emily E Brown
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jieqi Qian
- John G. Rangos Sr Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Bin Xie
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Evridiki Asimakidou
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- John G. Rangos Sr Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Vladislav Razskazovskiy
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- John G. Rangos Sr Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Deanna Ayrapetian
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- John G. Rangos Sr Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Vaibhav Sharma
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- John G. Rangos Sr Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Shunjin Xia
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Fei Liu
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
| | - Apeng Chen
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- John G. Rangos Sr Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Yongchang Guan
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- John G. Rangos Sr Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Zhengwei Li
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- John G. Rangos Sr Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Siyi Wanggou
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Olivier Saulnier
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Michelle Ly
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Wendy Fellows-Mayle
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Guifa Xi
- Division of Pediatric Neurosurgery, Ann and Robert H. Lurie Children's Hospital, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Tadanori Tomita
- Division of Pediatric Neurosurgery, Ann and Robert H. Lurie Children's Hospital, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Adam C Resnick
- Center for Data-Driven Discovery in Biomedicine, Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Stephen C Mack
- Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Eric H Raabe
- Division of Pediatric Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Charles G Eberhart
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dandan Sun
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Beth E Stronach
- Office of Research, University of Pittsburgh Health Sciences, Pittsburgh, PA, USA
| | - Sameer Agnihotri
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- John G. Rangos Sr Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Gary Kohanbash
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- John G. Rangos Sr Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Songjian Lu
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Karl Herrup
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jeremy N Rich
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - George K Gittes
- John G. Rangos Sr Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Alberto Broniscer
- John G. Rangos Sr Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Zhongliang Hu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Changsha, China
| | - Ian F Pollack
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- John G. Rangos Sr Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Robert M Friedlander
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sarah J Hainer
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA.
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
| | - Michael D Taylor
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada.
| | - Baoli Hu
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA.
- John G. Rangos Sr Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA.
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
10
|
Skitchenko R, Dinikina Y, Smirnov S, Krapivin M, Smirnova A, Morgacheva D, Artomov M. Case report: Somatic mutations in microtubule dynamics-associated genes in patients with WNT-medulloblastoma tumors. Front Oncol 2023; 12:1085947. [PMID: 36713498 PMCID: PMC9877404 DOI: 10.3389/fonc.2022.1085947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/07/2022] [Indexed: 01/14/2023] Open
Abstract
Medulloblastoma (MB) is the most common pediatric brain tumor which accounts for about 20% of all pediatric brain tumors and 63% of intracranial embryonal tumors. MB is considered to arise from precursor cell populations present during an early brain development. Most cases (~70%) of MB occur at the age of 1-4 and 5-9, but are also infrequently found in adults. Total annual frequency of pediatric tumors is about 5 cases per 1 million children. WNT-subtype of MB is characterized by a high probability of remission, with a long-term survival rate of about 90%. However, in some rare cases there may be increased metastatic activity, which dramatically reduces the likelihood of a favorable outcome. Here we report two cases of MB with a histological pattern consistent with desmoplastic/nodular (DP) and classic MB, and genetically classified as WNT-MB. Both cases showed putative causal somatic protein truncating mutations identified in microtubule-associated genes: ARID2, TUBB4A, and ANK3.
Collapse
Affiliation(s)
- Rostislav Skitchenko
- Almazov National Medical Research Centre, St. Petersburg, Russia,Computer Technologies Laboratory, ITMO University, St. Petersburg, Russia
| | - Yulia Dinikina
- Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Sergey Smirnov
- Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Mikhail Krapivin
- Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Anna Smirnova
- Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Daria Morgacheva
- Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Mykyta Artomov
- Almazov National Medical Research Centre, St. Petersburg, Russia,Computer Technologies Laboratory, ITMO University, St. Petersburg, Russia,The Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH, United States,Department of Pediatrics, Ohio State University, Columbus, OH, United States,*Correspondence: Mykyta Artomov,
| |
Collapse
|
11
|
Douyère M, Gong C, Richard M, Pellegrini-Moïse N, Daouk J, Pierson J, Chastagner P, Boura C. NRP1 inhibition modulates radiosensitivity of medulloblastoma by targeting cancer stem cells. Cancer Cell Int 2022; 22:377. [PMID: 36457009 PMCID: PMC9714111 DOI: 10.1186/s12935-022-02796-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 11/18/2022] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND Medulloblastoma (MB) is the most common pediatric malignant brain tumor. Despite current therapies, the morbidity and recurrent risk remains significant. Neuropilin-1 receptor (NRP1) has been implicated in the tumor progression of MB. Our recent study showed that NRP1 inhibition stimulated MB stem cells differentiation. Consequently, we hypothesized that targeting NRP1 in medulloblastoma could improve current treatments. METHODS NRP1 inhibition with a novel peptidomimetic agent, MR438, was evaluated with radiotherapy (RT) in MB models (DAOY, D283-Med and D341-Med) in vitro on cancer stem-like cells as well as in vivo on heterotopic and orthotopic xenografts. RESULTS We show that NRP1 inhibition by MR438 radiosensitizes MB stem-like cells in vitro. In heterotopic DAOY models, MR438 improves RT efficacy as measured by tumor growth and mouse survival. In addition, clonogenic assays after tumor dissociation showed a significant reduction in cancer stem cells with the combination treatment. In the same way, a benefit of the combined therapy was observed in the orthotopic model only for a low cumulative irradiation dose of 10 Gy but not for 20 Gy. CONCLUSIONS Finally, our results demonstrated that targeting NRP1 with MR438 could be a potential new strategy and could limit MB progression by decreasing the stem cell number while reducing the radiation dose.
Collapse
Affiliation(s)
- Manon Douyère
- grid.462787.80000 0001 2151 8763Université de Lorraine, CNRS, CRAN, UMR 7039, 54000 Nancy, France
| | - Caifeng Gong
- grid.462787.80000 0001 2151 8763Université de Lorraine, CNRS, CRAN, UMR 7039, 54000 Nancy, France ,grid.506261.60000 0001 0706 7839Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Can-Cer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Bei-Jing, 100021 China
| | - Mylène Richard
- Université de Lorraine, CNRS, L2CM, UMR 7053, Campus Science, 54500 Vandœuvre-Lès-Nancy, France
| | - Nadia Pellegrini-Moïse
- Université de Lorraine, CNRS, L2CM, UMR 7053, Campus Science, 54500 Vandœuvre-Lès-Nancy, France
| | - Joël Daouk
- grid.462787.80000 0001 2151 8763Université de Lorraine, CNRS, CRAN, UMR 7039, 54000 Nancy, France
| | - Julien Pierson
- grid.462787.80000 0001 2151 8763Université de Lorraine, CNRS, CRAN, UMR 7039, 54000 Nancy, France
| | - Pascal Chastagner
- grid.462787.80000 0001 2151 8763Université de Lorraine, CNRS, CRAN, UMR 7039, 54000 Nancy, France ,grid.410527.50000 0004 1765 1301Service d’Onco-Hématologie Pédiatrique, CHRU-Nancy, 54000 Nancy, France
| | - Cédric Boura
- grid.462787.80000 0001 2151 8763Université de Lorraine, CNRS, CRAN, UMR 7039, 54000 Nancy, France
| |
Collapse
|
12
|
Molecular subgrouping of medulloblastoma in pediatric population using the NanoString assay and comparison with immunohistochemistry methods. BMC Cancer 2022; 22:1221. [PMID: 36437460 PMCID: PMC9703680 DOI: 10.1186/s12885-022-10328-6] [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: 10/25/2021] [Accepted: 11/16/2022] [Indexed: 11/29/2022] Open
Abstract
PURPOSE Molecular subgrouping of medulloblastoma has become important due to its impact on risk group stratification. Immunohistochemistry (IHC) has been widely used but it has innate limitations. The NanoString assay has been proposed as an alternative method. This study aims to present the characteristics of medulloblastoma subgrouped by the NanoString assay and to compare the subgrouping results with the IHC method. METHODS Pediatric patients with histological diagnosis of medulloblastoma who underwent surgery from 2007 to 2021 were included. Clinical characteristics, pathological findings were reviewed. Molecular subgrouping was performed by IHC and by NanoString nCounter Elements TagSets assay. Test for concordance between two methods was made. RESULTS Among a total of 101 patients analyzed, subgrouping using the NanoString assay resulted in 14 (13.8%) WNT, 20 (19.8%) SHH, 18 (17.8%) Group 3, and 39 (38.6%) Group 4 subgroup cases. Survival analysis revealed the following from best to worse prognosis: WNT, Group 4, SHH, and Group 3. In SHH subgroup the large cell/anaplastic histology was present in 30% of cases. Seventy-one cases were analyzed for concordance between NanoString and IHC. Cohen's kappa value indicated moderate agreement but identification of Groups 3 and 4 with IHC using NPR3 and KCNA1 markers exhibited poor results. CONCLUSIONS The NanoString assay of Korean medulloblastoma patients revealed a more aggressive clinical course in the SHH subgroup which may be explained by a higher proportion of large cell/anaplastic histology being present in this subgroup. IHC did not distinguish Group 3 or 4 accurately. The NanoString assay may represent a good alternative method for practical use in the clinical field.
Collapse
|
13
|
Rao P, Furst L, Meyran D, Mayoh C, Neeson PJ, Terry R, Khuong-Quang DA, Mantamadiotis T, Ekert PG. Advances in CAR T cell immunotherapy for paediatric brain tumours. Front Oncol 2022; 12:873722. [PMID: 36505819 PMCID: PMC9727400 DOI: 10.3389/fonc.2022.873722] [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: 02/20/2022] [Accepted: 11/02/2022] [Indexed: 11/24/2022] Open
Abstract
Brain tumours are the most common solid tumour in children and the leading cause of cancer related death in children. Current treatments include surgery, chemotherapy and radiotherapy. The need for aggressive treatment means many survivors are left with permanent severe disability, physical, intellectual and social. Recent progress in immunotherapy, including genetically engineered T cells with chimeric antigen receptors (CARs) for treating cancer, may provide new avenues to improved outcomes for patients with paediatric brain cancer. In this review we discuss advances in CAR T cell immunotherapy, the major CAR T cell targets that are in clinical and pre-clinical development with a focus on paediatric brain tumours, the paediatric brain tumour microenvironment and strategies used to improve CAR T cell therapy for paediatric tumours.
Collapse
Affiliation(s)
- Padmashree Rao
- Translational Tumour Biology, Children’s Cancer Institute, Randwick, NSW, Australia
| | - Liam Furst
- Department of Microbiology & Immunology, The University of Melbourne, Victoria, VIC, Australia,Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, VIC, Australia
| | - Deborah Meyran
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia,Université de Paris, Inserm, U976 Human Immunology Pathophysiology Immunotherapy (HIPI) Unit, Institut de Recherche Saint-Louis, Paris, France,Children’s Cancer Centre, Royal Children’s Hospital, Parkville, VIC, Australia
| | - Chelsea Mayoh
- Translational Tumour Biology, Children’s Cancer Institute, Randwick, NSW, Australia,School of Women and Children’s Health, University of New South Wales, Randwick, NSW, Australia
| | - Paul J. Neeson
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Rachael Terry
- Translational Tumour Biology, Children’s Cancer Institute, Randwick, NSW, Australia,School of Women and Children’s Health, University of New South Wales, Randwick, NSW, Australia
| | - Dong-Anh Khuong-Quang
- Translational Tumour Biology, Children’s Cancer Institute, Randwick, NSW, Australia,Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, VIC, Australia,Children’s Cancer Centre, Royal Children’s Hospital, Parkville, VIC, Australia
| | - Theo Mantamadiotis
- Department of Microbiology & Immunology, The University of Melbourne, Victoria, VIC, Australia,Department of Surgery Royal Melbourne Hospital (RMH), The University of Melbourne, Parkville, VIC, Australia,*Correspondence: Theo Mantamadiotis, ; Paul G. Ekert,
| | - Paul G. Ekert
- Translational Tumour Biology, Children’s Cancer Institute, Randwick, NSW, Australia,Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, VIC, Australia,Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia,School of Women and Children’s Health, University of New South Wales, Randwick, NSW, Australia,Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia,*Correspondence: Theo Mantamadiotis, ; Paul G. Ekert,
| |
Collapse
|
14
|
microRNA-155-3p delivered by M2 macrophages-derived exosomes enhances the progression of medulloblastoma through regulation of WDR82. J Transl Med 2022; 20:13. [PMID: 34983581 PMCID: PMC8728908 DOI: 10.1186/s12967-021-03156-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 11/19/2021] [Indexed: 01/12/2023] Open
Abstract
Objective Exosomes, membranous nanovesicles, naturally bringing proteins, mRNAs, and microRNAs (miRNAs), play crucial roles in tumor pathogenesis. This study was to investigate the role of miR-155-3p from M2 macrophages-derived exosomes (M2-Exo) in promoting medulloblastoma (MB) progression by mediating WD repeat domain 82 (WDR82). Methods miR-155-3p expression was detected by RT-qPCR. The relationship of miR-155-3p with clinicopathological features of MB patients was analyzed. M2-Exo were isolated and identified by TEM, NTA and Western blot. CCK-8 assay, colony formation assay, flow cytometry, wound healing assay, and Transwell assay were performed to explore the role of miR-155-3p-enriched M2-Exo on the progression of MB cells. Luciferase assay were used to identify the relationship between miR-155-3p and WDR82. The effect of miR-155-3p-enriched M2-Exo on tumorigenesis of MB was confirmed by the xenograft nude mice model. Results miR-155-3p was up-regulated in MB tissues of patients and MB cell lines. High miR-155-3p expression was correlated with the pathological type and molecular subtype classification of MB patients. WDR82 was a direct target of miR-155-3p. miR-155-3p was packaged into M2-Exo. miR-155-3p-enriched M2-Exo promoted the progression of Daoy cells. miR-155-3p-enriched M2-Exo promoted in vivo tumorigenesis. Conclusion The study highlights that miR-155-3p-loaded M2-Exo enhances the growth of MB cells via down-regulating WDR82, which might provide a deep insight into MB mechanism. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-03156-y.
Collapse
|
15
|
Brown NJ, Wilson B, Shahrestani S, Choi EH, Lien BV, Paladugu A, Tran K, Ransom SC, Tafreshi AR, Ransom RC, Sahyouni R, Chan AY, Yang I. The 100 Most Influential Publications on Medulloblastoma: Areas of Past, Current, and Future Focus. World Neurosurg 2020; 146:119-139. [PMID: 33212273 DOI: 10.1016/j.wneu.2020.11.038] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/07/2020] [Accepted: 11/07/2020] [Indexed: 11/17/2022]
Abstract
BACKGROUND This article is the first to identify the most influential articles on medulloblastoma using the citation analysis methodology. OBJECTIVE To perform a bibliometric analysis of the 100 most-cited articles on medulloblastoma. METHODS Using the Web of Science database, search criteria included the title-specific keyword "medulloblastoma" OR "cerebellar primitive neuroectodermal tumor (PNET)" OR "cerebellar PNET." Publications from 1900 to 2020 labeled "article," "review," "data set," or "clinical trial" were chosen and ranked based on total number of citations in descending order. Each article was evaluated based on the following variables: total citations, average citations per year, first author, institution of first author, title, publication year, country of origin, SCImago Journal Rank, and Scopus SNIP (Source Normalized Impact per Paper). RESULTS Our search yielded 4928 articles on medulloblastoma. The 100 most-cited articles ranged from 192 to 2017 across 42 unique journals; Journal of Clinical Oncology accounted for the most publications (16%). Paul A. Northcott was first author of the most articles on the list (n = 7.7%), and the most widely cited article was "Altered neural cell fates and medulloblastoma in mouse patched mutants" by Goodrich et al., published in Science (1997). CONCLUSIONS Because medulloblastoma represents the most common form of pediatric cancerous brain tumor, it is important to identify works that have significantly contributed to the body of knowledge regarding this disease. The 100 most-cited medulloblastoma articles comprise a significant collection of data regarding the histopathologic and molecular classification of medulloblastoma as well as clinical outcomes of therapeutics used to treat this disease.
Collapse
Affiliation(s)
- Nolan J Brown
- Department of Neurological Surgery, University of California, Irvine, Orange, California, USA.
| | - Bayard Wilson
- Department of Neurological Surgery, University of California, Los Angeles, California, USA
| | - Shane Shahrestani
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA; Department of Medical Engineering, California Institute of Technology, Pasadena, California, USA
| | - Elliot H Choi
- School of Medicine, Case Western Reserve University, Cleveland, California, USA
| | - Brian V Lien
- Department of Neurological Surgery, University of California, Irvine, Orange, California, USA
| | - Anushka Paladugu
- Department of Neurological Surgery, University of California, Irvine, Orange, California, USA
| | - Katelynn Tran
- Department of Neuroscience, University of Southern California, Los Angeles, California, USA
| | - Seth C Ransom
- College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Ali R Tafreshi
- Department of Neurological Surgery, Geisinger Health System, Danville, Pennsylvania, USA
| | - Ryan Chase Ransom
- Department of Neurologic Surgery, Mayo Clinic Alix School of Medicine, Rochester, Minnesota, USA
| | - Ronald Sahyouni
- Department of Neurological Surgery, University of California, San Diego, La Jolla, California, USA
| | - Alvin Y Chan
- Department of Neurological Surgery, University of California, Irvine, Orange, California, USA
| | - Isaac Yang
- Department of Neurological Surgery, University of California, Los Angeles, California, USA
| |
Collapse
|
16
|
Dhanyamraju PK, Patel TN, Dovat S. Medulloblastoma: "Onset of the molecular era". Mol Biol Rep 2020; 47:9931-9937. [PMID: 33159234 DOI: 10.1007/s11033-020-05971-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 11/02/2020] [Indexed: 10/23/2022]
Abstract
Among brain tumors, Medulloblastoma (MB) is one of the most common, malignant, pediatric tumors of the cerebellum. It accounts for ~20% of all childhood central nervous system (CNS) tumors. Despite, tremendous advances in drug development processes, as well as novel drugs for MB the morbidity and mortality rates, remain high. Craniospinal radiation, high-dose chemotherapy, and surgical resection are the primary therapeutic strategies. Tremendous progress in the field of "genomics" with vast amounts of data has led to the identification of four distinct molecular subgroups in medulloblastoma: WNT group, SHH group, group-III, and group-IV. The identification of these subgroups has led to individualized treatment strategies for each subgroup. Here, we discuss the various molecular subgroups of medulloblastoma as well as the differences between them. We also highlight the latest treatment strategies available for medulloblastoma.
Collapse
Affiliation(s)
- Pavan Kumar Dhanyamraju
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
| | - Trupti N Patel
- Department of Integrative Biology, Vellore Institute of Technology, Vellore, India
| | - Sinisa Dovat
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| |
Collapse
|