1
|
Yang L, Huang K, Cao L, Ma Y, Li S, Zhou J, Zhao Z, Wang S. Molecular profiling of core immune-escape genes highlights TNFAIP3 as an immune-related prognostic biomarker in neuroblastoma. Inflamm Res 2024; 73:1529-1545. [PMID: 39028490 DOI: 10.1007/s00011-024-01914-4] [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: 04/06/2024] [Revised: 06/28/2024] [Accepted: 07/03/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND Neuroblastoma (NB) is the most prevalent and deadliest pediatric solid tumor. With of over 50% of high-risk neuroblastoma cases relapse, the imperative for novel drug targets and therapeutic strategies is accentuated. In neuroblastoma, the existence of tumor-associated macrophages (TAMs) correlates with an unfavorable patient prognosis. However, the clinical relevance and prognostic implications of regulatory genes linked to TAMs infiltration in neuroblastoma remain unclear, and further study is required. METHODS We conducted a comprehensive analysis utilizing transcriptome expression profiles from three primary datasets associated with neuroblastoma (GSE45547, GSE49710, TARGET) to identify hub genes implicated in immune evasion within neuroblastoma. Subsequently, we utilized single-cell RNA sequencing analysis on 17 clinical neuroblastoma samples to investigate the expression and distribution of these hub genes, leading to the identification of TNFAIP3. The above three public databases were merged to allowed for the validation of TNFAIP3's molecular functions through GO and KEGG analysis. Furthermore, we assessed TNFAIP3's correlation with immune infiltration and its potential immunotherapeutic impact by multiple algorithms. Our single-cell transcriptome data revealed the role of TNFAIP3 in macrophage polarization. Finally, preliminary experimental verifications to confirm the biological functions of TNFAIP3-mediated TAMs in NB. RESULTS A total of 6 genes related to immune evasion were screened and we found that TNFAIP3 exhibited notably higher expression in macrophages than other immune cell types, based on the scRNA-sequencing data. GO and KEGG analysis showed that low expression of TNFAIP3 significantly correlated with the activation of multiple oncogenic pathways as well as immune-related pathways. Then validation affirmed that individuals within the TNFAIP3 high-expression cohort could potentially derive greater advantages from immunotherapeutic interventions, alongside exhibiting heightened immune responsiveness. Deciphering the pseudotime trajectory of macrophages, we revealed the potential of TNFAIP3 in inducing the polarization of macrophages towards the M1 phenotype. Finally, we confirmed that patients in the TNFAIP3 high expression group might benefit more from immunotherapy or chemotherapy as substantiated by RT-qPCR and immunofluorescence examinations. Moreover, the role of TNFAIP3 in macrophage polarization was validated. Preliminary experiment showed that TNFAIP3-mediated TAMs inhibit the proliferation, migration and invasion capabilities of NB cells. CONCLUSIONS Our results suggest that TNFAIP3 was first identified as a promising biomarker for immunotherapy and potential molecular target in NB. Besides, the presence of TNFAIP3 within TAMs may offer a novel therapeutic strategy for NB.
Collapse
Affiliation(s)
- Linyu Yang
- Department of Pediatric Surgical Oncology Children's Hospitial of Chongqinng Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Kai Huang
- Department of Pediatric Surgical Oncology Children's Hospitial of Chongqinng Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Lijian Cao
- Department of Pediatric Surgical Oncology Children's Hospitial of Chongqinng Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yue Ma
- Department of Pediatric Surgical Oncology Children's Hospitial of Chongqinng Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Suwen Li
- Department of Pediatric Surgical Oncology Children's Hospitial of Chongqinng Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Jianwu Zhou
- Department of Pediatric Surgical Oncology Children's Hospitial of Chongqinng Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Zhenzhen Zhao
- Department of Pediatric Surgical Oncology Children's Hospitial of Chongqinng Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Shan Wang
- Department of Pediatric Surgical Oncology Children's Hospitial of Chongqinng Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children's Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
2
|
Bhardwaj N, Rohilla M, Trehan A, Bansal D, Kakkar N, Srinivasan R. MYCN amplification and International Neuroblastoma Risk Group stratification on fine-needle aspiration biopsy and their correlation to survival in neuroblastoma. J Clin Pathol 2023; 76:599-605. [PMID: 35414524 DOI: 10.1136/jclinpath-2022-208177] [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: 01/20/2022] [Accepted: 03/25/2022] [Indexed: 11/03/2022]
Abstract
AIMS Risk stratification as per the International Neuroblastoma Risk Group (INRG) stratification is important for management of neuroblastoma. INRG incorporates various parameters including histological category as per the International Neuroblastoma Pathology Classification (INPC) and MYCN amplification, which were evaluated in fine needle aspiration biopsy (FNAB) samples of neuroblastoma patients to ascertain their impact in our population. METHODS This was a retrospective study including 60 neuroblastoma cases diagnosed on FNAB, staged and stratified by INRG. Mitosis Karyorrhexis Index (MKI), INPC morphological category and MYCN status by fluorescence in situ hybridisation (n=46) were evaluated and correlated to outcome. RESULTS The mean age was 29 months (21 days to 9 years) with 27 and 33 children ≥18 months; male: female ratio of 1.6: 1; INRG stage-30(M), 20(L2), 2(L1) and 2(MS); INRG-36 high-risk, 13 intermediate-risk and 11 low-risk categories, respectively. MKI was high, intermediate and low in 39, 4 and 7 cases, respectively. INPC morphological type included 2 ganglioneuroblastomas and 58 neuroblastomas, graded further as 25 undifferentiated and 33 poorly differentiated tumours. MYCN was amplified in 48% (22/46) cases and correlated with undifferentiated morphology (p=0.01). At a mean follow-up of 469 (7-835) days, 22/50 were disease free and 28/50 had relapsed/died. The overall survival correlated with age (p=0.03), stage (p=0.01), INRG group (p=0.0001) and tumour grade (p=0.036). MYCN status independently did not correlate with age (p=0.5), INRG stage (p=0.2) and overall survival (p=0.4). CONCLUSION FNAB is a complete modality for diagnosing neuroblastoma and providing all information required for risk stratification as per INRG including MKI, MYCN amplification, INPC category. Our cohort with predominant high-risk neuroblastoma cases highlights regional variation.
Collapse
Affiliation(s)
- Neha Bhardwaj
- Pathology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Manish Rohilla
- Cytology & Gynecological Pathology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Amita Trehan
- Department of Pediatrics (Hematology-Oncology Division), Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Deepak Bansal
- Department of Pediatrics (Hematology-Oncology Division), Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Nandita Kakkar
- Histopathology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Radhika Srinivasan
- Cytology & Gynecological Pathology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
3
|
Gao J, Fosbrook C, Gibson J, Underwood TJ, Gray JC, Walters ZS. Review: Targeting EZH2 in neuroblastoma. Cancer Treat Rev 2023; 119:102600. [PMID: 37467626 DOI: 10.1016/j.ctrv.2023.102600] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/10/2023] [Accepted: 07/12/2023] [Indexed: 07/21/2023]
Abstract
Neuroblastoma is one of the commonest extra-cranial pediatric tumors, and accounts for over 15% of all childhood cancer mortality. Risk stratification for children with neuroblastoma is based on age, stage, histology, and tumor cytogenetics. The majority of patients are considered to have high-risk neuroblastoma, for which the long-term survival is less than 50%. Current treatments combine surgical resection, chemotherapy, stem cell transplantation, radiotherapy, anti-GD2 based immunotherapy as well as the differentiating agent isotretinoin. Despite the intensive multimodal therapies applied, there are high relapse rates, and recurrent disease is often resistant to further therapy. Enhancer of Zeste Homolog 2 (EZH2), a catalytic subunit of Polycomb Repressive Complex 2 (PRC2), is a histone methyltransferase that represses transcription through trimethylation of lysine residue K27 on histone H3 (H3K27me3). It is responsible for epigenetic repression of transcription, making EZH2 an essential regulator for cell differentiation. Overexpression of EZH2 has been shown to promote tumorigenesis, cancer cell proliferation and prevent tumor cells from differentiating in a number of cancers. Therefore, research has been ongoing for the past decade, developing treatments that target EZH2 in neuroblastoma. This review summarises the role of EZH2 in neuroblastoma and evaluates the latest research findings on the therapeutic potential of targeting EZH2 in the treatment of neuroblastoma.
Collapse
Affiliation(s)
- Jinhui Gao
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, SO16 6YD, UK.
| | - Claire Fosbrook
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, SO16 6YD, UK
| | - Jane Gibson
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, SO16 6YD, UK
| | - Timothy J Underwood
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, SO16 6YD, UK
| | - Juliet C Gray
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, SO16 6YD, UK
| | - Zoë S Walters
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, SO16 6YD, UK
| |
Collapse
|
4
|
Fetahu IS, Esser-Skala W, Dnyansagar R, Sindelar S, Rifatbegovic F, Bileck A, Skos L, Bozsaky E, Lazic D, Shaw L, Tötzl M, Tarlungeanu D, Bernkopf M, Rados M, Weninger W, Tomazou EM, Bock C, Gerner C, Ladenstein R, Farlik M, Fortelny N, Taschner-Mandl S. Single-cell transcriptomics and epigenomics unravel the role of monocytes in neuroblastoma bone marrow metastasis. Nat Commun 2023; 14:3620. [PMID: 37365178 DOI: 10.1038/s41467-023-39210-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 05/29/2023] [Indexed: 06/28/2023] Open
Abstract
Metastasis is the major cause of cancer-related deaths. Neuroblastoma (NB), a childhood tumor has been molecularly defined at the primary cancer site, however, the bone marrow (BM) as the metastatic niche of NB is poorly characterized. Here we perform single-cell transcriptomic and epigenomic profiling of BM aspirates from 11 subjects spanning three major NB subtypes and compare these to five age-matched and metastasis-free BM, followed by in-depth single cell analyses of tissue diversity and cell-cell interactions, as well as functional validation. We show that cellular plasticity of NB tumor cells is conserved upon metastasis and tumor cell type composition is NB subtype-dependent. NB cells signal to the BM microenvironment, rewiring via macrophage mgration inhibitory factor and midkine signaling specifically monocytes, which exhibit M1 and M2 features, are marked by activation of pro- and anti-inflammatory programs, and express tumor-promoting factors, reminiscent of tumor-associated macrophages. The interactions and pathways characterized in our study provide the basis for therapeutic approaches that target tumor-to-microenvironment interactions.
Collapse
Affiliation(s)
- Irfete S Fetahu
- St. Anna Children's Cancer Research Institute, Vienna, Austria.
| | - Wolfgang Esser-Skala
- Department of Biosciences and Medical Biology, University of Salzburg, Salzburg, Austria
| | - Rohit Dnyansagar
- Department of Biosciences and Medical Biology, University of Salzburg, Salzburg, Austria
| | - Samuel Sindelar
- Department of Biosciences and Medical Biology, University of Salzburg, Salzburg, Austria
| | | | - Andrea Bileck
- University of Vienna, Department of Analytical Chemistry, Faculty of Chemistry, Vienna, Austria
- Joint Metabolomics Facility, University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Lukas Skos
- University of Vienna, Department of Analytical Chemistry, Faculty of Chemistry, Vienna, Austria
| | - Eva Bozsaky
- St. Anna Children's Cancer Research Institute, Vienna, Austria
| | - Daria Lazic
- St. Anna Children's Cancer Research Institute, Vienna, Austria
| | - Lisa Shaw
- Medical University of Vienna, Department of Dermatology, Vienna, Austria
| | - Marcus Tötzl
- St. Anna Children's Cancer Research Institute, Vienna, Austria
| | | | - Marie Bernkopf
- St. Anna Children's Cancer Research Institute, Vienna, Austria
| | - Magdalena Rados
- St. Anna Children's Cancer Research Institute, Vienna, Austria
| | - Wolfgang Weninger
- Medical University of Vienna, Department of Dermatology, Vienna, Austria
| | - Eleni M Tomazou
- St. Anna Children's Cancer Research Institute, Vienna, Austria
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Medical University of Vienna, Institute of Artificial Intelligence, Center for Medical Data Science, Vienna, Austria
| | - Christopher Gerner
- University of Vienna, Department of Analytical Chemistry, Faculty of Chemistry, Vienna, Austria
- Joint Metabolomics Facility, University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Ruth Ladenstein
- St. Anna Children's Hospital and St. Anna Children's Cancer Research Institute, Department of Studies and Statistics for Integrated Research and Projects, Vienna, Austria
- Medical University of Vienna, Department of Pediatrics, Vienna, Austria
| | - Matthias Farlik
- Medical University of Vienna, Department of Dermatology, Vienna, Austria
| | - Nikolaus Fortelny
- Department of Biosciences and Medical Biology, University of Salzburg, Salzburg, Austria.
| | | |
Collapse
|
5
|
Liu Y, Dan W, Yan B. A light-operated dual-mode method for neuroblastoma diagnosis based on a Tb-MOF: from biometabolite detection to logic devices. Inorg Chem Front 2023. [DOI: 10.1039/d2qi02701a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Tb-DBA can not only serve as a light-operated dual-mechanism driven platform to detect VMA (an early pathological feature of neuroblastoma), but can also produce a different fluorescence response to epinephrine (EP, the metabolic precursor of VMA).
Collapse
Affiliation(s)
- Yanhong Liu
- Shanghai Key Lab of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Siping Road 1239, Shanghai 200092, China
| | - Wenyan Dan
- Shanghai Key Lab of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Siping Road 1239, Shanghai 200092, China
| | - Bing Yan
- Shanghai Key Lab of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Siping Road 1239, Shanghai 200092, China
| |
Collapse
|
6
|
Ciceri S, Carenzo A, Iannó MF, Bertolotti A, Morosi C, Luksch R, Spreafico F, Collini P, Radice P, Massimino M, De Cecco L, Perotti D. Gene expression-based dissection of inter-histotypes, intra-histotype and intra-tumor heterogeneity in pediatric tumors. Sci Rep 2022; 12:17837. [PMID: 36284197 PMCID: PMC9596396 DOI: 10.1038/s41598-022-20536-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 09/14/2022] [Indexed: 02/07/2023] Open
Abstract
Intra-tumor heterogeneity (ITH) fosters tumor evolution, resistance to therapy, and relapse. Recently, many evidence have been accumulated on the occurrence of genetic ITH in pediatric cancers. With this study we aimed to address the downstream effects that genetic and epigenetic ITH, and tumor-microenvironment interactions may produce within a tumor mass. To this aim, we investigated by high-throughput gene expression multiple samples of 5 hepatoblastomas, 5 neuroblastomas, 5 rhabdomyosarcomas, and 5 Wilms tumors. Principal component analysis, single sample hallmark gene sets analysis, and weighted gene co-expression network analysis were performed on gene expression data. We observed that the different tumors clustered by histotype, and then by case, and in addition, a variable degree of ITH was visible in all the investigated cases. The ITH highlighted in this study can represent a challenge in tumor treatment since we demonstrated that different druggable hallmarks and targets may be heterogeneously present within the same tumor mass, and this can potentially lead to therapeutic failure. Despite this heterogeneity, we could highlight some commonalities among the different histotypes investigated, supporting the feasibility to move in the clinic from a histotype-driven to a target-driven, sometimes agnostic, approach at least in some cases.
Collapse
Affiliation(s)
- Sara Ciceri
- grid.417893.00000 0001 0807 2568Molecular Bases of Genetic Risk and Genetic Testing Unit, Department of Research, Fondazione IRCCS Istituto Nazionale Dei Tumori, Via Venezian 1, 20133 Milano, Italy
| | - Andrea Carenzo
- grid.417893.00000 0001 0807 2568Molecular Mechanisms Unit, Department of Research, Fondazione IRCCS Istituto Nazionale Dei Tumori, Via Venezian 1, 20133 Milano, Italy
| | - Maria Federica Iannó
- grid.417893.00000 0001 0807 2568Integrated Biology Platform, Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Alessia Bertolotti
- grid.417893.00000 0001 0807 2568Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Carlo Morosi
- grid.417893.00000 0001 0807 2568Department of Radiology, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Roberto Luksch
- grid.417893.00000 0001 0807 2568Pediatric Oncology Unit, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Filippo Spreafico
- grid.417893.00000 0001 0807 2568Pediatric Oncology Unit, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Paola Collini
- grid.417893.00000 0001 0807 2568Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Paolo Radice
- grid.417893.00000 0001 0807 2568Molecular Bases of Genetic Risk and Genetic Testing Unit, Department of Research, Fondazione IRCCS Istituto Nazionale Dei Tumori, Via Venezian 1, 20133 Milano, Italy
| | - Maura Massimino
- grid.417893.00000 0001 0807 2568Pediatric Oncology Unit, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Loris De Cecco
- grid.417893.00000 0001 0807 2568Molecular Mechanisms Unit, Department of Research, Fondazione IRCCS Istituto Nazionale Dei Tumori, Via Venezian 1, 20133 Milano, Italy
| | - Daniela Perotti
- grid.417893.00000 0001 0807 2568Molecular Bases of Genetic Risk and Genetic Testing Unit, Department of Research, Fondazione IRCCS Istituto Nazionale Dei Tumori, Via Venezian 1, 20133 Milano, Italy
| |
Collapse
|
7
|
Gomez RL, Ibragimova S, Ramachandran R, Philpott A, Ali FR. Tumoral heterogeneity in neuroblastoma. Biochim Biophys Acta Rev Cancer 2022; 1877:188805. [PMID: 36162542 DOI: 10.1016/j.bbcan.2022.188805] [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: 04/06/2022] [Revised: 08/28/2022] [Accepted: 09/17/2022] [Indexed: 10/31/2022]
Abstract
Neuroblastoma is a solid, neuroendocrine tumor with divergent clinical behavior ranging from asymptomatic to fatal. The diverse clinical presentations of neuroblastoma are directly linked to the high intra- and inter-tumoral heterogeneity it presents. This heterogeneity is strongly associated with therapeutic resistance and continuous relapses, often leading to fatal outcomes. The development of successful risk assessment and tailored treatment strategies lies in evaluating the extent of heterogeneity via the accurate genetic and epigenetic profiling of distinct cell subpopulations present in the tumor. Recent studies have focused on understanding the molecular mechanisms that drive tumoral heterogeneity in pursuing better therapeutic and diagnostic approaches. This review describes the cellular, genetic, and epigenetic aspects of neuroblastoma heterogeneity. In addition, we summarize the recent findings on three crucial factors that can lead to heterogeneity in solid tumors: the inherent diversity of the progenitor cells, the presence of cancer stem cells, and the influence of the tumor microenvironment.
Collapse
Affiliation(s)
- Roshna Lawrence Gomez
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Healthcare City, Dubai, United Arab Emirates
| | - Shakhzada Ibragimova
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Healthcare City, Dubai, United Arab Emirates
| | - Revathy Ramachandran
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Healthcare City, Dubai, United Arab Emirates
| | - Anna Philpott
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom; Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Center, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Fahad R Ali
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Healthcare City, Dubai, United Arab Emirates.
| |
Collapse
|
8
|
Spence T, Dubuc AM. Copy Number Analysis in Cancer Diagnostic Testing. Clin Lab Med 2022; 42:451-468. [DOI: 10.1016/j.cll.2022.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
9
|
Molecular Basis of Beckwith–Wiedemann Syndrome Spectrum with Associated Tumors and Consequences for Clinical Practice. Cancers (Basel) 2022; 14:cancers14133083. [PMID: 35804856 PMCID: PMC9265096 DOI: 10.3390/cancers14133083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/19/2022] [Accepted: 06/20/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary Beckwith–Wiedemann syndrome (BWS, OMIM 130650) is an inborn overgrowth disorder caused by molecular alterations in chromosome 11p15.5. These molecular changes affect so-called imprinted genes, i.e., genes which underlie a complex regulation which is linked to the parental origin of the gene copy. Thus, either the maternal gene copy is expressed or the paternal, but this balanced regulation is prone to disturbances. In fact, different types of molecular variants have been identified in BWS, resulting in a variable phenotype; thus, it was consented that the syndromic entity was extended to the Beckwith–Wiedemann spectrum (BWSp). Some molecular subgroups of BWSp are associated with an increased embryonic tumor risk and have different likelihoods for specific tumors. Therefore, the precise determination of the molecular subgroup is needed for precise monitoring and treatment, but the molecular diagnostic procedure has several limitations and challenges which have to be considered. Abstract Beckwith–Wiedemann syndrome (BWS, OMIM 130650) is a congenital imprinting condition with a heterogenous clinical presentation of overgrowth and an increased childhood cancer risk (mainly nephroblastoma, hepatoblastoma or neuroblastoma). Due to the varying clinical presentation encompassing classical, clinical BWS without a molecular diagnosis and BWS-related phenotypes with an 11p15.5 molecular anomaly, the syndromic entity was extended to the Beckwith–Wiedemann spectrum (BWSp). The tumor risk of up to 30% depends on the molecular subtype of BWSp with causative genetic or epigenetic alterations in the chromosomal region 11p15.5. The molecular diagnosis of BWSp can be challenging for several reasons, including the range of causative molecular mechanisms which are frequently mosaic. The molecular basis of tumor formation appears to relate to stalled cellular differentiation in certain organs that predisposes persisting embryonic cells to accumulate additional molecular defects, which then results in a range of embryonal tumors. The molecular subtype of BWSp not only influences the overall risk of neoplasia, but also the likelihood of specific embryonal tumors.
Collapse
|
10
|
Lodrini M, Graef J, Thole-Kliesch TM, Astrahantseff K, Sprüssel A, Grimaldi M, Peitz C, Linke RB, Hollander JF, Lankes E, Künkele A, Oevermann L, Schwabe G, Fuchs J, Szymansky A, Schulte JH, Hundsdörfer P, Eckert C, Amthauer H, Eggert A, Deubzer HE. Targeted Analysis of Cell-free Circulating Tumor DNA is Suitable for Early Relapse and Actionable Target Detection in Patients with Neuroblastoma. Clin Cancer Res 2022; 28:1809-1820. [PMID: 35247920 DOI: 10.1158/1078-0432.ccr-21-3716] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/12/2021] [Accepted: 02/16/2022] [Indexed: 11/16/2022]
Abstract
PURPOSE Treating refractory or relapsed neuroblastoma remains challenging. Monitoring body fluids for tumor-derived molecular information indicating minimal residual disease supports more frequent diagnostic surveillance and may have the power to detect resistant subclones before they give rise to relapses. If actionable targets are identified from liquid biopsies, targeted treatment options can be considered earlier. EXPERIMENTAL DESIGN Droplet digital PCR assays assessing MYCN and ALK copy numbers and allelic frequencies of ALK p.F1174L and ALK p.R1275Q mutations were applied to longitudinally collected liquid biopsies and matched tumor tissue samples from 31 patients with high-risk neuroblastoma. Total cell-free DNA (cfDNA) levels and marker detection were compared with data from routine clinical diagnostics. RESULTS Total cfDNA concentrations in blood plasma from patients with high-risk neuroblastoma were higher than in healthy controls and consistently correlated with neuron-specific enolase levels and lactate dehydrogenase activity but not with 123I-meta-iodobenzylguanidine scores at relapse diagnosis. Targeted cfDNA diagnostics proved superior for early relapse detection to all current diagnostics in 2 patients. Marker analysis in cfDNA indicated intratumor heterogeneity for cell clones harboring MYCN amplifications and druggable ALK alterations that were not detectable in matched tumor tissue samples in 17 patients from our cohort. Proof of concept is provided for molecular target detection in cerebrospinal fluid from patients with isolated central nervous system relapses. CONCLUSIONS Tumor-specific alterations can be identified and monitored during disease course in liquid biopsies from pediatric patients with high-risk neuroblastoma. This approach to cfDNA surveillance warrants further prospective validation and exploitation for diagnostic purposes and to guide therapeutic decisions.
Collapse
Affiliation(s)
- Marco Lodrini
- Department of Pediatric Hematology and Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Experimental and Clinical Research Center (ECRC) of the Charité and the Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| | - Josefine Graef
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Theresa M Thole-Kliesch
- Department of Pediatric Hematology and Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Kathy Astrahantseff
- Department of Pediatric Hematology and Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Annika Sprüssel
- Department of Pediatric Hematology and Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Maddalena Grimaldi
- Department of Pediatric Hematology and Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Experimental and Clinical Research Center (ECRC) of the Charité and the Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| | - Constantin Peitz
- Department of Pediatric Hematology and Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Experimental and Clinical Research Center (ECRC) of the Charité and the Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| | - Rasmus B Linke
- Department of Pediatric Hematology and Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Experimental and Clinical Research Center (ECRC) of the Charité and the Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| | - Jan F Hollander
- Department of Pediatric Hematology and Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Experimental and Clinical Research Center (ECRC) of the Charité and the Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| | - Erwin Lankes
- Department of Pediatric Endocrinology and Diabetes, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Center for Chronically Sick Children, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Annette Künkele
- Department of Pediatric Hematology and Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berliner Institut für Gesundheitsforschung (BIH), Berlin, Germany
| | - Lena Oevermann
- Department of Pediatric Hematology and Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Georg Schwabe
- Children's Hospital, Carl-Thiem-Klinikum, Cottbus, Germany
| | - Jörg Fuchs
- Department of Pediatric Surgery and Pediatric Urology, University Children's Hospital, Eberhard-Karls-University Tuebingen, Tuebingen, Germany
| | - Annabell Szymansky
- Department of Pediatric Hematology and Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Johannes H Schulte
- Department of Pediatric Hematology and Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berliner Institut für Gesundheitsforschung (BIH), Berlin, Germany.,German Cancer Consortium (DKTK), partner site Berlin, Berlin, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Patrick Hundsdörfer
- Department of Pediatric Hematology and Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Pediatric Oncology, Helios Klinikum Berlin Buch, Berlin, Germany
| | - Cornelia Eckert
- Department of Pediatric Hematology and Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Holger Amthauer
- Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| | - Angelika Eggert
- Department of Pediatric Hematology and Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berliner Institut für Gesundheitsforschung (BIH), Berlin, Germany.,German Cancer Consortium (DKTK), partner site Berlin, Berlin, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hedwig E Deubzer
- Department of Pediatric Hematology and Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Experimental and Clinical Research Center (ECRC) of the Charité and the Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany.,Berliner Institut für Gesundheitsforschung (BIH), Berlin, Germany.,German Cancer Consortium (DKTK), partner site Berlin, Berlin, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
11
|
van Gerven MR, Bozsaky E, Matser YAH, Vosseberg J, Taschner-Mandl S, Koster J, Tytgat GAM, Molenaar JJ, van den Boogaard M. The mutational spectrum of ATRX aberrations in neuroblastoma and the associated patient and tumor characteristics. Cancer Sci 2022; 113:2167-2178. [PMID: 35384159 PMCID: PMC9207354 DOI: 10.1111/cas.15363] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 03/18/2022] [Accepted: 04/02/2022] [Indexed: 11/30/2022] Open
Abstract
Neuroblastoma is the most common extracranial solid tumor in children. The chromatin remodeler ATRX is frequently mutated in high‐risk patients with a poor prognosis. Although many studies have reported ATRX aberrations and the associated clinical characteristics in neuroblastoma, a comprehensive overview is currently lacking. In this study, we extensively characterize the mutational spectrum of ATRX aberrations in neuroblastoma tumors reported in previous studies and present an overview of patient and tumor characteristics. We collected the data of a total of 127 neuroblastoma patients and three cell lines with ATRX aberrations originating from 20 papers. We subdivide the ATRX aberrations into nonsense, missense, and multiexon deletions (MEDs) and show that 68% of them are MEDs. Of these MEDs, 75% are predicted to be in‐frame. Furthermore, we identify a missense mutational hotspot region in the helicase domain. We also confirm that all three ATRX mutation types are more often identified in patients diagnosed at an older age, but still approximately 40% of the patients are aged 5 years or younger at diagnosis. Surprisingly, we found that 11q deletions are enriched in neuroblastomas with ATRX deletions compared to a reference cohort, but not in neuroblastomas with ATRX point mutations. Taken together, our data emphasizes a distinct ATRX mutation spectrum in neuroblastoma, which should be considered when studying molecular phenotypes and therapeutic strategies.
Collapse
Affiliation(s)
| | - Eva Bozsaky
- Tumor biology group, St Anna Children's Cancer Research Institute, Vienna, Austria
| | - Yvette A H Matser
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Julian Vosseberg
- Theoretical Biology and Bioinformatics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | | | - Jan Koster
- Department of Oncogenomics, Amsterdam UMC, location AMC, Amsterdam, the Netherlands
| | | | - Jan J Molenaar
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.,Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | | |
Collapse
|
12
|
Chan T, Chen Y, Tan KT, Wu C, Wu W, Li W, Wang J, Shiue Y, Li C. Biological significance of MYC and CEBPD coamplification in urothelial carcinoma: Multilayered genomic, transcriptional and posttranscriptional positive feedback loops enhance oncogenic glycolysis. Clin Transl Med 2021; 11:e674. [PMID: 34954904 PMCID: PMC8710299 DOI: 10.1002/ctm2.674] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 11/18/2021] [Accepted: 11/25/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE The aim of this study is to decipher the underlying mechanisms of CCAAT/enhancer-binding protein delta (CEBPD)-enhanced glycolysis as well as the biological significance of CEBPD and MYC coamplification in urothelial carcinoma (UC). METHODS In vitro analyses were conducted to examine the effects of altered CEBPD or MYC expression on UC cells. The in vivo effects of CEBPD overexpression in a high-glucose environment on tumour growth were investigated in xenografted induced diabetic severe combined immunodeficiency/beige mice. Data mining was used to cross-validate the associations between CEBPD and MYC copy number and transcriptional expression, quantitative reverse transcription-polymerase chain reaction, immunohistochemistry, chromogenic in situ hybridization, and in situ hybridization targeting microRNA were performed on 635 UC patient samples and xenograft samples. UC patient survival in relation to diabetes was validated by using the National Health Insurance Research Database. RESULTS CEBPD and MYC coamplification (29.6%) occurred at a high frequency, MYC expression promoted chromosomal instability, facilitating CEBPD copy number gain and expression. CEBPD promoted glucose uptake and lactate production by upregulating SLC2A1 and HK2, leading to mitochondrial fission, increased extracellular acidification rate and decreased oxygen consumption rate to fuel cell growth. CEBPD upregulated HK2 expression through multiple regulation pathways including MYC stabilization, suppression of FBXW7 transactivation and MYC-independent transcriptional suppression of hsa-miR-429. Clinical and xenografted experiments confirmed the growth advantage of CEBPD in relation to glucose metabolic dysregulation and the significant correlations between the expression of these genes. CONCLUSIONS We confirmed that CEBPD has an oncogenic role in UC by activating AKT signalling and initiating metabolic reprogramming from mitochondrial oxidative phosphorylation to glycolysis to satisfy glucose addiction. These novel CEBPD- and MYC-centric multilayered positive feedback loops enhance cancer growth that could complement theranostic approaches.
Collapse
Affiliation(s)
- Ti‐Chun Chan
- Department of Medical ResearchChi Mei Medical CenterTainanTaiwan
- National Institute of Cancer ResearchNational Health Research InstitutesTainanTaiwan
| | - Yi‐Ting Chen
- Department of Biotechnology and Bioindustry SciencesCollege of Bioscience and BiotechnologyNational Cheng Kung UniversityTainanTaiwan
| | | | | | - Wen‐Jeng Wu
- Graduate Institute of Clinical MedicineCollege of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Department of UrologyKaohsiung Medical University HospitalKaohsiungTaiwan
- Department of UrologySchool of MedicineCollege of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Department of UrologyMinistry of Health and Welfare Pingtung HospitalPingtungTaiwan
| | - Wei‐Ming Li
- Graduate Institute of Clinical MedicineCollege of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Department of UrologyKaohsiung Medical University HospitalKaohsiungTaiwan
- Department of UrologySchool of MedicineCollege of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Department of UrologyMinistry of Health and Welfare Pingtung HospitalPingtungTaiwan
| | - Ju‐Ming Wang
- Department of Biotechnology and Bioindustry SciencesCollege of Bioscience and BiotechnologyNational Cheng Kung UniversityTainanTaiwan
| | - Yow‐Ling Shiue
- Institute of Precision MedicineNational Sun Yat‐Sen UniversityKaohsiungTaiwan
- Department of PathologySchool of MedicineCollege of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
| | - Chien‐Feng Li
- Department of Medical ResearchChi Mei Medical CenterTainanTaiwan
- National Institute of Cancer ResearchNational Health Research InstitutesTainanTaiwan
- Institute of Precision MedicineNational Sun Yat‐Sen UniversityKaohsiungTaiwan
- Department of PathologySchool of MedicineCollege of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
| |
Collapse
|
13
|
López-Carrasco A, Berbegall AP, Martín-Vañó S, Blanquer-Maceiras M, Castel V, Navarro S, Noguera R. Intra-Tumour Genetic Heterogeneity and Prognosis in High-Risk Neuroblastoma. Cancers (Basel) 2021; 13:5173. [PMID: 34680323 PMCID: PMC8534138 DOI: 10.3390/cancers13205173] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 12/15/2022] Open
Abstract
Spatial ITH is defined by genomic and biological variations within a tumour acquired by tumour cell evolution under diverse microenvironments, and its role in NB patient prognosis is understudied. In this work, we applied pangenomic techniques to detect chromosomal aberrations in at least two different areas of each tumour and/or in simultaneously obtained solid and liquid biopsies, detecting ITH in the genomic profile of almost 40% of HR-NB. ITH was better detected when comparing one or more tumour pieces and liquid biopsy (50%) than between different tumour pieces (21%). Interestingly, we found that patients with ITH analysed by pangenomic techniques had a significantly better survival rate that those with non-heterogeneous tumours, especially in cases without MYCN amplification. Moreover, all patients in the studied cohort with high ITH (defined as 50% or more genomic aberration differences between areas of a tumour or simultaneously obtained samples) survived after 48 months. These results clearly support analysing at least two solid tumour areas (separately or mixed) and liquid samples to provide more accurate genomic diagnosis, prognosis and therapy options in HR-NB.
Collapse
Affiliation(s)
- Amparo López-Carrasco
- Department of Pathology, Medical School, University of Valencia-INCLIVA, 46010 Valencia, Spain; (A.L.-C.); (A.P.B.); (S.M.-V.); (M.B.-M.); (S.N.)
- CIBER of Cancer (CIBERONC), 28029 Madrid, Spain
| | - Ana P. Berbegall
- Department of Pathology, Medical School, University of Valencia-INCLIVA, 46010 Valencia, Spain; (A.L.-C.); (A.P.B.); (S.M.-V.); (M.B.-M.); (S.N.)
- CIBER of Cancer (CIBERONC), 28029 Madrid, Spain
| | - Susana Martín-Vañó
- Department of Pathology, Medical School, University of Valencia-INCLIVA, 46010 Valencia, Spain; (A.L.-C.); (A.P.B.); (S.M.-V.); (M.B.-M.); (S.N.)
- CIBER of Cancer (CIBERONC), 28029 Madrid, Spain
| | - Maite Blanquer-Maceiras
- Department of Pathology, Medical School, University of Valencia-INCLIVA, 46010 Valencia, Spain; (A.L.-C.); (A.P.B.); (S.M.-V.); (M.B.-M.); (S.N.)
- CIBER of Cancer (CIBERONC), 28029 Madrid, Spain
| | - Victoria Castel
- Clinical and Translational Oncology Research Group, Investigation Institute La Fe, 46026 Valencia, Spain;
| | - Samuel Navarro
- Department of Pathology, Medical School, University of Valencia-INCLIVA, 46010 Valencia, Spain; (A.L.-C.); (A.P.B.); (S.M.-V.); (M.B.-M.); (S.N.)
- CIBER of Cancer (CIBERONC), 28029 Madrid, Spain
| | - Rosa Noguera
- Department of Pathology, Medical School, University of Valencia-INCLIVA, 46010 Valencia, Spain; (A.L.-C.); (A.P.B.); (S.M.-V.); (M.B.-M.); (S.N.)
- CIBER of Cancer (CIBERONC), 28029 Madrid, Spain
| |
Collapse
|
14
|
Di Giannatale A, Di Paolo PL, Curione D, Lenkowicz J, Napolitano A, Secinaro A, Tomà P, Locatelli F, Castellano A, Boldrini L. Radiogenomics prediction for MYCN amplification in neuroblastoma: A hypothesis generating study. Pediatr Blood Cancer 2021; 68:e29110. [PMID: 34003574 DOI: 10.1002/pbc.29110] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/13/2021] [Accepted: 04/23/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND MYCN amplification represents a powerful prognostic factor in neuroblastoma (NB) and may occasionally account for intratumoral heterogeneity. Radiomics is an emerging field of advanced image analysis that aims to extract a large number of quantitative features from standard radiological images, providing valuable clinical information. PROCEDURE In this retrospective study, we aimed to create a radiogenomics model by correlating computed tomography (CT) radiomics analysis with MYCN status. NB lesions were segmented on pretherapy CT scans and radiomics features subsequently extracted using a dedicated library. Dimensionality reduction/features selection approaches were then used for features procession and logistic regression models have been developed for the considered outcome. RESULTS Seventy-eight patients were included in this study, as training dataset, of which 24 presented MYCN amplification. In total, 232 radiomics features were extracted. Eight features were selected through Boruta algorithm and two features were lastly chosen through Pearson correlation analysis: mean of voxel intensity histogram (p = .0082) and zone size non-uniformity (p = .038). Five-times repeated three-fold cross-validation logistic regression models yielded an area under the curve (AUC) value of 0.879 on the training set. The model was then applied to an independent validation cohort of 21 patients, of which five presented MYCN amplification. The validation of the model yielded a 0.813 AUC value, with 0.85 accuracy on previously unseen data. CONCLUSIONS CT-based radiomics is able to predict MYCN amplification status in NB, paving the way to the in-depth analysis of imaging based biomarkers that could enhance outcomes prediction.
Collapse
Affiliation(s)
- Angela Di Giannatale
- Department of Pediatric Hematology/Oncology and Cell and Gene Therapy, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | | | - Davide Curione
- Department of Imaging, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Jacopo Lenkowicz
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Antonio Napolitano
- Medical Physics Department, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Aurelio Secinaro
- Department of Imaging, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Paolo Tomà
- Department of Imaging, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Franco Locatelli
- Department of Pediatric Hematology/Oncology and Cell and Gene Therapy, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy.,Department of Gynecology/Obstetrics and Pediatrics, Sapienza University of Rome, Rome, Italy
| | - Aurora Castellano
- Department of Pediatric Hematology/Oncology and Cell and Gene Therapy, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Luca Boldrini
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| |
Collapse
|
15
|
Bellini A, Pötschger U, Bernard V, Lapouble E, Baulande S, Ambros PF, Auger N, Beiske K, Bernkopf M, Betts DR, Bhalshankar J, Bown N, de Preter K, Clément N, Combaret V, Font de Mora J, George SL, Jiménez I, Jeison M, Marques B, Martinsson T, Mazzocco K, Morini M, Mühlethaler-Mottet A, Noguera R, Pierron G, Rossing M, Taschner-Mandl S, Van Roy N, Vicha A, Chesler L, Balwierz W, Castel V, Elliott M, Kogner P, Laureys G, Luksch R, Malis J, Popovic-Beck M, Ash S, Delattre O, Valteau-Couanet D, Tweddle DA, Ladenstein R, Schleiermacher G. Frequency and Prognostic Impact of ALK Amplifications and Mutations in the European Neuroblastoma Study Group (SIOPEN) High-Risk Neuroblastoma Trial (HR-NBL1). J Clin Oncol 2021; 39:3377-3390. [PMID: 34115544 PMCID: PMC8791815 DOI: 10.1200/jco.21.00086] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
PURPOSE In neuroblastoma (NB), the ALK receptor tyrosine kinase can be constitutively activated through activating point mutations or genomic amplification. We studied ALK genetic alterations in high-risk (HR) patients on the HR-NBL1/SIOPEN trial to determine their frequency, correlation with clinical parameters, and prognostic impact. MATERIALS AND METHODS Diagnostic tumor samples were available from 1,092 HR-NBL1/SIOPEN patients to determine ALK amplification status (n = 330), ALK mutational profile (n = 191), or both (n = 571). RESULTS Genomic ALK amplification (ALKa) was detected in 4.5% of cases (41 out of 901), all except one with MYCN amplification (MNA). ALKa was associated with a significantly poorer overall survival (OS) (5-year OS: ALKa [n = 41] 28% [95% CI, 15 to 42]; no-ALKa [n = 860] 51% [95% CI, 47 to 54], [P < .001]), particularly in cases with metastatic disease. ALK mutations (ALKm) were detected at a clonal level (> 20% mutated allele fraction) in 10% of cases (76 out of 762) and at a subclonal level (mutated allele fraction 0.1%-20%) in 3.9% of patients (30 out of 762), with a strong correlation between the presence of ALKm and MNA (P < .001). Among 571 cases with known ALKa and ALKm status, a statistically significant difference in OS was observed between cases with ALKa or clonal ALKm versus subclonal ALKm or no ALK alterations (5-year OS: ALKa [n = 19], 26% [95% CI, 10 to 47], clonal ALKm [n = 65] 33% [95% CI, 21 to 44], subclonal ALKm (n = 22) 48% [95% CI, 26 to 67], and no alteration [n = 465], 51% [95% CI, 46 to 55], respectively; P = .001). Importantly, in a multivariate model, involvement of more than one metastatic compartment (hazard ratio [HR], 2.87; P < .001), ALKa (HR, 2.38; P = .004), and clonal ALKm (HR, 1.77; P = .001) were independent predictors of poor outcome. CONCLUSION Genetic alterations of ALK (clonal mutations and amplifications) in HR-NB are independent predictors of poorer survival. These data provide a rationale for integration of ALK inhibitors in upfront treatment of HR-NB with ALK alterations.
Collapse
Affiliation(s)
- Angela Bellini
- Equipe SiRIC RTOP Recherche Translationelle en Oncologie Pédiatrique, Institut Curie, Paris, France.,INSERM U830, Laboratoire de Génétique et Biologie des Cancers, Institut Curie, Paris, France.,SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| | - Ulrike Pötschger
- Department for Studies and Statistics and Integrated Research, Vienna, Austria.,St Anna Children's Cancer Research Institute, Vienna, Austria
| | - Virginie Bernard
- Institut Curie Genomics of Excellence (ICGex) Platform, Research Center, Institut Curie, Paris, France
| | - Eve Lapouble
- Unité de Génétique Somatique, Service de Génétique, Hospital Group, Institut Curie, Paris, France
| | - Sylvain Baulande
- Institut Curie Genomics of Excellence (ICGex) Platform, Research Center, Institut Curie, Paris, France
| | - Peter F Ambros
- St Anna Children's Cancer Research Institute, Vienna, Austria
| | - Nathalie Auger
- Service de Génétique des tumeurs; Institut Gustave Roussy, Villejuif, France
| | - Klaus Beiske
- Department of Pathology, Oslo University Hospital, and Medical Faculty, University of Oslo, Oslo, Norway
| | - Marie Bernkopf
- St Anna Children's Cancer Research Institute, Vienna, Austria
| | - David R Betts
- Department of Clinical Genetics, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Jaydutt Bhalshankar
- Equipe SiRIC RTOP Recherche Translationelle en Oncologie Pédiatrique, Institut Curie, Paris, France.,INSERM U830, Laboratoire de Génétique et Biologie des Cancers, Institut Curie, Paris, France.,SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| | - Nick Bown
- Northern Genetics Service, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | | | - Nathalie Clément
- Equipe SiRIC RTOP Recherche Translationelle en Oncologie Pédiatrique, Institut Curie, Paris, France.,INSERM U830, Laboratoire de Génétique et Biologie des Cancers, Institut Curie, Paris, France.,SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| | - Valérie Combaret
- Translational Research Laboratory, Centre Léon Bérard, Lyon, France
| | | | - Sally L George
- Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom
| | - Irene Jiménez
- Equipe SiRIC RTOP Recherche Translationelle en Oncologie Pédiatrique, Institut Curie, Paris, France.,INSERM U830, Laboratoire de Génétique et Biologie des Cancers, Institut Curie, Paris, France.,SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| | - Marta Jeison
- Schneider Children's Medical Center of Israel, Tel Aviv University, Tel Aviv, Israel
| | - Barbara Marques
- Departamento de Genética Humana, Instituto Nacional de Saúde Doutor Ricardo Jorge, Lisbon, Portugal
| | | | - Katia Mazzocco
- Department of Pathology, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Martina Morini
- Laboratory of Molecular Biology, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Annick Mühlethaler-Mottet
- Pediatric Hematology-Oncology Research Laboratory, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Rosa Noguera
- Department of Pathology, Medical School, University of Valencia-Incliva Health Research Institute/CIBERONC, Madrid, Spain
| | - Gaelle Pierron
- Unité de Génétique Somatique, Service de Génétique, Hospital Group, Institut Curie, Paris, France
| | - Maria Rossing
- Center for Genomic Medicine, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | | | | | - Ales Vicha
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Louis Chesler
- Paediatric Tumour Biology, Division of Clinical Studies, The Institute of Cancer Research, Sutton, United Kingdom
| | - Walentyna Balwierz
- Department of Pediatric Oncology and Hematology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Victoria Castel
- Clinical and Translational Oncology Research Group, Health Research Institute La Fe, Valencia, Spain
| | - Martin Elliott
- Leeds Children's Hospital, Leeds General Infirmary, Leeds, United Kingdom
| | - Per Kogner
- Karolinska University Hospital, Stockholm, Sweden
| | - Geneviève Laureys
- Department of Paediatric Haematology and Oncology, Princess Elisabeth Children's Hospital, Ghent University Hospital, Ghent, Belgium
| | - Roberto Luksch
- Paediatric Oncology, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - Josef Malis
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Maja Popovic-Beck
- Pediatric Hematology-Oncology Unit, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Shifra Ash
- Ruth Rappaport Children's Hospital, Rambam Health Care Campus, Haifa, Israel
| | - Olivier Delattre
- INSERM U830, Laboratoire de Génétique et Biologie des Cancers, Institut Curie, Paris, France.,SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France.,Institut Curie Genomics of Excellence (ICGex) Platform, Research Center, Institut Curie, Paris, France
| | | | - Deborah A Tweddle
- Wolfson Childhood Cancer Research Centre, Newcastle Centre for Cancer, Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Ruth Ladenstein
- Department for Studies and Statistics and Integrated Research, St Anna Children's Hospital, St Anna Children's Cancer Research Institute, Vienna, Austria.,Department of Paediatrics, Medical University of Vienna, Vienna, Austria
| | - Gudrun Schleiermacher
- Equipe SiRIC RTOP Recherche Translationelle en Oncologie Pédiatrique, Institut Curie, Paris, France.,INSERM U830, Laboratoire de Génétique et Biologie des Cancers, Institut Curie, Paris, France.,SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| |
Collapse
|
16
|
Nakazawa A. Biological categories of neuroblastoma based on the international neuroblastoma pathology classification for treatment stratification. Pathol Int 2021; 71:232-244. [PMID: 33657257 DOI: 10.1111/pin.13085] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 01/27/2021] [Indexed: 11/28/2022]
Abstract
The International Neuroblastoma Pathology Classification (INPC), which distinguishes a favorable histology (FH) and an unfavorable histology (UH), is one of the most powerful prognostic factors in patients with neuroblastoma. FH that shows spontaneous regression or age-appropriate tumor differentiation/maturation, is common in infants and has mutual interaction with Schwann cells via the NGF/NTRK1 pathway and gain of whole chromosome 17. In contrast, UH is prevalent in older children and is molecularly heterogeneous. MYCN amplification is the most frequent genomic abnormality in tumors with UH. MYCN-amplified tumors demonstrate characteristic histology, the same as MYC-positive neuroblastoma. Chromosome 1pLOH is often associated with MYCN amplification, but on the other hand, chromosome 11qLOH rarely occurs in combination with MYCN amplification. 11qLOH has an inferior prognostic impact in UH without MYCN amplification. The high expression of ALK protein is a negative prognostic factor in both ALK mutated or amplified tumors and FH, but not in UH. Abnormal maintenance/elongation of telomeres; overexpression of telomerase reverse transcriptase (TERT) and the alternative lengthening of telomeres (ALT) phenotype due to ATRX mutation, are another molecular event in UH. The INPC, incorporating immunohistochemistry for MYCN, MYC, ALK, TERT and ATRX, represents a practical and implementable approach to create the biological category for the future management of patients with this unique disease.
Collapse
Affiliation(s)
- Atsuko Nakazawa
- Department of Clinical Research, Saitama Children's Medical Center, Saitama, Japan
| |
Collapse
|
17
|
Long-Term Outcome and Role of Biology within Risk-Adapted Treatment Strategies: The Austrian Neuroblastoma Trial A-NB94. Cancers (Basel) 2021; 13:cancers13030572. [PMID: 33540616 PMCID: PMC7867286 DOI: 10.3390/cancers13030572] [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: 11/23/2020] [Revised: 01/08/2021] [Accepted: 01/28/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Neuroblastoma, the most common extracranial malignancy of childhood, shows a highly variable course of disease ranging from spontaneous regression or maturation into a benign tumor to an aggressive and intractable cancer in up to 60% of patients. To adapt treatment intensity, risk staging at diagnosis is of utmost importance. The A-NB94 trial was the first in Austria to stratify therapy intensity according to tumor staging, patient’s age, and MYCN amplification status, the latter being a biologic marker turning otherwise low-risk tumors into high-risk disease. Recent publications showed a prognostic impact of various genomic features including segmental chromosomal aberrations (SCAs). We retrospectively investigated the relevance of SCAs within this risk-adapted treatment strategy. The A-NB94 approach resulted in an excellent long-term survival for the majority of patients with acceptable long-term morbidity. An age- and stage-dependent frequency of SCAs was confirmed and SCAs should always be considered in future treatment decision making processes. Abstract We evaluated long-term outcome and genomic profiles in the Austrian Neuroblastoma Trial A-NB94 which applied a risk-adapted strategy of treatment (RAST) using stage, age and MYCN amplification (MNA) status for stratification. RAST ranged from surgery only to intensity-adjusted chemotherapy, single or multiple courses of high-dose chemotherapy (HDT) followed by autologous stem cell rescue depending on response to induction chemotherapy, and irradiation to the primary tumor site. Segmental chromosomal alterations (SCAs) were investigated retrospectively using multi- and pan-genomic techniques. The A-NB94 trial enrolled 163 patients. Patients with localized disease had an excellent ten-year (10y) event free survival (EFS) and overall survival (OS) of 99 ± 1% and 93 ± 2% whilst it was 80 ± 13% and 90 ± 9% for infants with stage 4S and for infants with stage 4 non-MNA disease both 83 ± 15%. Stage 4 patients either >12 months or ≤12 months but with MNA had a 10y-EFS and OS of 45 ± 8% and 47 ± 8%, respectively. SCAs were present in increasing frequencies according to stage and age: in 29% of localized tumors but in 92% of stage 4 tumors (p < 0.001), and in 39% of patients ≤ 12 months but in 63% of patients > 12 months (p < 0.001). RAST successfully reduced chemotherapy exposure in low- and intermediate-risk patients with excellent long-term results while the outcome of high-risk disease met contemporary trials.
Collapse
|
18
|
Fernández-Blanco B, Berbegall AP, Martin-Vañó S, Castel V, Navarro S, Noguera R. Imbalance between genomic gain and loss identifies high-risk neuroblastoma patients with worse outcomes. Neoplasia 2020; 23:12-20. [PMID: 33190090 PMCID: PMC7674617 DOI: 10.1016/j.neo.2020.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/03/2020] [Accepted: 11/03/2020] [Indexed: 12/13/2022] Open
Abstract
Survival in high-risk neuroblastoma (HR-NB) patients remains poor despite multimodal treatment. We aimed to identify HR-NB patients with worse outcomes by analyzing the genomic instability derived from segmental chromosomal aberrations. We calculated 3 genomic instability indexes for primary tumor SNP array profiles from 127 HR-NB patients: (1) Copy number aberration burden (%gainslength+%losseslength), (2) copy number load (CNL) (%gainslength-%losseslength) and (3) net genomic load (NGL) (%gainsamount-%lossesamount). Tumors were classified according to positive or negative CNL and NGL genomic subtypes. The impact of the genomic instability indexes on overall survival (OS) was assessed with Cox regression. We identified 38% of HR-NB patients with poor 5-year OS. A negative CNL genomic background was related to poor prognosis in patients ≥18 months showing tumors with homogeneous MYCN amplification (9.5% survival probability, P < 0.05) and patients with non-MYCN amplified NB (18.8% survival probability related to >2.4% CNL, P < 0.01). A positive CNL genomic background was associated with worse outcome in patients with heterogeneous MYCN amplification (22.5% survival probability, P < 0.05). We conclude that characterizing a tumor genomic background according to predominance of genome gained or lost contributes toward improved outcome prediction and brings greater insight into the tumor biology of HR-NB patients.
Collapse
Affiliation(s)
| | - Ana Pilar Berbegall
- Pathology Department, Medical School, University of Valencia-INCLIVA, Valencia, Spain; CIBERONC, Madrid, Spain
| | - Susana Martin-Vañó
- Pathology Department, Medical School, University of Valencia-INCLIVA, Valencia, Spain; CIBERONC, Madrid, Spain
| | - Victoria Castel
- Clinical and Translational Oncology Research Group, Investigation Institute La Fe, Valencia, Spain
| | - Samuel Navarro
- Pathology Department, Medical School, University of Valencia-INCLIVA, Valencia, Spain; CIBERONC, Madrid, Spain
| | - Rosa Noguera
- Pathology Department, Medical School, University of Valencia-INCLIVA, Valencia, Spain; CIBERONC, Madrid, Spain.
| |
Collapse
|
19
|
Su Y, Wang L, Zhao Q, Yue Z, Zhao W, Wang X, Duan C, Jin M, Zhang D, Chen S, Yin J, Qiu L, Cheng X, Xu Z, Ma X. Implementation of the plasma MYCN/NAGK ratio to detect MYCN amplification in patients with neuroblastoma. Mol Oncol 2020; 14:2884-2893. [PMID: 32896084 PMCID: PMC7607162 DOI: 10.1002/1878-0261.12794] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 07/28/2020] [Accepted: 09/01/2020] [Indexed: 01/26/2023] Open
Abstract
Detection of amplification of the MYCN gene is essential for determining optimal treatment and estimating prognosis of patients with neuroblastoma (NB). DNA FISH with neuroblastoma tissues or patient‐derived bone marrow cells is the standard clinical practice for the detection of MYCN amplification. As tumor cells may often be unavailable, we developed a method to detect MYCN amplification in the plasma of patients with neuroblastoma. Taking single‐copy NAGK DNA as reference, we used real‐time quantitative PCR (qPCR) to determine the MYCN/NAGK ratio in the plasma of 115 patients diagnosed with NB. An increased MYCN/NAGK ratio in the plasma was consistent with MYCN amplification as assessed by DNA FISH. The AUC for a MYCN/NAGK ratio equal to 6.965 was 0.943, with 86% sensitivity and 100% specificity. Beyond the threshold of 6.965, the MYCN/NAGK ratio correlated with a heavier tumor burden. Event‐free and overall survival of two years were significantly shortened in stage 4 patients with a MYCN/NAGK ratio higher than 6.965. Plasma MYCN/NAGK ratios increased in patients with progressive disease and relapse. Thus, we conclude that the determination of the plasma MYCN/NAGK ratio by qPCR is a noninvasive and reproducible method to measure MYCN amplification in patients with NB.
Collapse
Affiliation(s)
- Yan Su
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Discipline of Pediatrics, Ministry of Education, MOE Key Laboratory of Major Diseases in Children, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Lijun Wang
- Beijing Keyin Technology Company Limited, Beijing Keyin Evergreen Institutes for Medical Research Company Limited, China
| | - Qian Zhao
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Discipline of Pediatrics, Ministry of Education, MOE Key Laboratory of Major Diseases in Children, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Zhixia Yue
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Discipline of Pediatrics, Ministry of Education, MOE Key Laboratory of Major Diseases in Children, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Wen Zhao
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Discipline of Pediatrics, Ministry of Education, MOE Key Laboratory of Major Diseases in Children, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Xisi Wang
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Discipline of Pediatrics, Ministry of Education, MOE Key Laboratory of Major Diseases in Children, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Chao Duan
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Discipline of Pediatrics, Ministry of Education, MOE Key Laboratory of Major Diseases in Children, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Mei Jin
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Discipline of Pediatrics, Ministry of Education, MOE Key Laboratory of Major Diseases in Children, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Dawei Zhang
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Discipline of Pediatrics, Ministry of Education, MOE Key Laboratory of Major Diseases in Children, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Shenglan Chen
- Taizhou Genewill Medical Laboratory Company Limited, China
| | - Jianfeng Yin
- Taizhou Genewill Medical Laboratory Company Limited, China
| | - Lihua Qiu
- Beijing Keyin Technology Company Limited, Beijing Keyin Evergreen Institutes for Medical Research Company Limited, China
| | - Xianfeng Cheng
- Beijing Keyin Technology Company Limited, Beijing Keyin Evergreen Institutes for Medical Research Company Limited, China
| | - Zhong Xu
- Beijing Keyin Technology Company Limited, Beijing Keyin Evergreen Institutes for Medical Research Company Limited, China
| | - Xiaoli Ma
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Discipline of Pediatrics, Ministry of Education, MOE Key Laboratory of Major Diseases in Children, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| |
Collapse
|
20
|
Peitz C, Sprüssel A, Linke RB, Astrahantseff K, Grimaldi M, Schmelz K, Toedling J, Schulte JH, Fischer M, Messerschmidt C, Beule D, Keilholz U, Eggert A, Deubzer HE, Lodrini M. Multiplexed Quantification of Four Neuroblastoma DNA Targets in a Single Droplet Digital PCR Reaction. J Mol Diagn 2020; 22:1309-1323. [PMID: 32858250 DOI: 10.1016/j.jmoldx.2020.07.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 07/16/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022] Open
Abstract
The detection and characterization of cell-free DNA (cfDNA) in peripheral blood from neuroblastoma patients may serve as a minimally invasive approach to liquid biopsy. Major challenges in the analysis of cfDNA purified from blood samples are small sample volumes and low cfDNA concentrations. Droplet digital PCR (ddPCR) is a technology suitable for analyzing low levels of cfDNA. Reported here are two quadruplexed ddPCR assay protocols that reliably quantify MYCN and ALK copy numbers in a single reaction together with the two reference genes, NAGK and AFF3, and accurately estimate ALKF1174L (exon 23 position 3522, C>A) and ALKR1275Q (exon 25 position 3824, G>A) mutant allele fractions using cfDNA as input. The separation of positive and negative droplets was optimized for detecting two targets in each ddPCR fluorescence channel by the adjustment of the probe and primer concentrations of each target molecule. The quadruplexed assays were validated using a panel of 10 neuroblastoma cell lines and paired blood plasma and primary neuroblastoma samples from nine patients. Accuracy and sensitivity thresholds in quadruplexed assays corresponded well with those from the respective duplexed assays. Presented are two robust quadruplexed ddPCR protocols applicable in the routine clinical setting and that require only minimal plasma volumes for the assessment of MYCN and ALK oncogene status.
Collapse
Affiliation(s)
- Constantin Peitz
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Berlin, Germany; Neuroblastoma Research Group, Experimental and Clinical Research Center, Berlin, Germany
| | - Annika Sprüssel
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Berlin, Germany; Neuroblastoma Research Group, Experimental and Clinical Research Center, Berlin, Germany
| | - Rasmus B Linke
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Berlin, Germany; Neuroblastoma Research Group, Experimental and Clinical Research Center, Berlin, Germany
| | - Kathy Astrahantseff
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Maddalena Grimaldi
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Berlin, Germany; Neuroblastoma Research Group, Experimental and Clinical Research Center, Berlin, Germany
| | - Karin Schmelz
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Berlin, Germany; German Cancer Consortium, partner site Berlin, Berlin, Germany; German Cancer Research Center, Heidelberg, Germany
| | - Joern Toedling
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Johannes H Schulte
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Berlin, Germany; German Cancer Consortium, partner site Berlin, Berlin, Germany; German Cancer Research Center, Heidelberg, Germany; Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin Institute of Health, Berlin, Germany
| | - Matthias Fischer
- Department of Experimental Pediatric Oncology, University Children's Hospital of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, Cologne, Germany
| | - Clemens Messerschmidt
- Core Unit Bioinformatics, Charité-Universitätsmedizin Berlin, Berlin, Germany; Department of Computer Science, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Dieter Beule
- Core Unit Bioinformatics, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Angelika Eggert
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Berlin, Germany; German Cancer Consortium, partner site Berlin, Berlin, Germany; German Cancer Research Center, Heidelberg, Germany; Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin Institute of Health, Berlin, Germany
| | - Hedwig E Deubzer
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Berlin, Germany; Neuroblastoma Research Group, Experimental and Clinical Research Center, Berlin, Germany; German Cancer Consortium, partner site Berlin, Berlin, Germany; German Cancer Research Center, Heidelberg, Germany; Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin Institute of Health, Berlin, Germany.
| | - Marco Lodrini
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Berlin, Germany; Neuroblastoma Research Group, Experimental and Clinical Research Center, Berlin, Germany
| |
Collapse
|
21
|
Campbell K, Naranjo A, Hibbitts E, Gastier-Foster JM, Bagatell R, Irwin MS, Shimada H, Hogarty M, Park JR, DuBois SG. Association of heterogeneous MYCN amplification with clinical features, biological characteristics and outcomes in neuroblastoma: A report from the Children's Oncology Group. Eur J Cancer 2020; 133:112-119. [PMID: 32492633 DOI: 10.1016/j.ejca.2020.04.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/02/2020] [Accepted: 04/14/2020] [Indexed: 12/17/2022]
Abstract
PURPOSE MYCN amplification (MNA) is associated with poor outcomes in neuroblastoma. Less is known about heterogeneous MNA within a tumour. We compared clinical characteristics, biologic features and clinical outcomes of patients with heterogeneous MNA to patients with either homogeneous MNA or MYCN wild-type tumours. PATIENTS AND METHODS In this retrospective cohort study, we categorized patients as having tumours with MYCN wild-type, homogeneous MNA (>20% amplified tumour cells) or heterogeneous MNA (≤20% amplified tumour cells). We used chi-squared or Fisher's exact tests to compare features between groups. We used log-rank tests and Cox models to compare event-free survival (EFS) and overall survival (OS) between groups. RESULTS MYCN status and heterogeneity status (if amplified) could be ascertained in diagnostic tumour samples from 5975 patients, including 57 (1%) with heterogeneous MNA, 981 (16.4%) with homogeneous MNA, and 4937 (82.6%) with MYCN wild-type tumours. Multiple clinical and biological features differed between patients with heterogeneous vs. homogeneous MNA, including enrichment for thoracic primary sites and paucity of 1p loss of heterozygosity with heterogeneous MNA (p < 0.0001). Importantly, EFS and OS were not significantly different between patients with heterogeneous vs. homogeneous MNA. Further, EFS and OS for patients with heterogeneous MNA were significantly inferior to patients with wild-type MYCN. CONCLUSION Although neuroblastomas with heterogeneous MNA demonstrate significantly different biological and clinical patterns compared with homogeneous MNA, prognosis is similar between the two groups. These results support current practice that treats patients with heterogeneous MNA similarly to patients with homogeneous MNA.
Collapse
Affiliation(s)
- Kevin Campbell
- Dana-Farber / Boston Children's Cancer and Blood Disorders Center and Harvard Medical School, Boston, MA, USA
| | - Arlene Naranjo
- Children's Oncology Group Statistics and Data Center, University of Florida, Gainesville, FL, USA
| | - Emily Hibbitts
- Children's Oncology Group Statistics and Data Center, University of Florida, Gainesville, FL, USA
| | - Julie M Gastier-Foster
- Institute for Genomic Medicine, Nationwide Children's Hospital, Departments of Pathology and Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Rochelle Bagatell
- Department of Pediatrics, Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Meredith S Irwin
- Department of Pediatrics, The Hospital for Sick Children, Toronto, Canada
| | | | - Michael Hogarty
- Department of Pediatrics, Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Julie R Park
- Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA, USA
| | - Steven G DuBois
- Dana-Farber / Boston Children's Cancer and Blood Disorders Center and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
22
|
Duan K, Dickson BC, Marrano P, Thorner PS, Chung CT. Adult‐onset neuroblastoma: Report of seven cases with molecular genetic characterization. Genes Chromosomes Cancer 2019; 59:240-248. [DOI: 10.1002/gcc.22826] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 11/13/2019] [Accepted: 11/19/2019] [Indexed: 01/10/2023] Open
Affiliation(s)
- Kai Duan
- Department of Laboratory Medicine and Pathobiology University of Toronto Toronto Ontario Canada
| | - Brendan C. Dickson
- Department of Laboratory Medicine and Pathobiology University of Toronto Toronto Ontario Canada
- Department of Pathology and Laboratory Medicine Mount Sinai Hospital Toronto Ontario Canada
| | - Paula Marrano
- Division of Pathology The Hospital for Sick Children Toronto Ontario Canada
| | - Paul S. Thorner
- Department of Laboratory Medicine and Pathobiology University of Toronto Toronto Ontario Canada
- Division of Pathology The Hospital for Sick Children Toronto Ontario Canada
| | - Catherine T. Chung
- Department of Laboratory Medicine and Pathobiology University of Toronto Toronto Ontario Canada
- Division of Pathology The Hospital for Sick Children Toronto Ontario Canada
| |
Collapse
|
23
|
Huang X, Zhao J, Zhu J, Chen S, Fu W, Tian X, Lou S, Ruan J, He J, Zhou H. MYCN gene polymorphisms and Wilms tumor susceptibility in Chinese children. J Clin Lab Anal 2019; 33:e22988. [PMID: 31343784 PMCID: PMC7938399 DOI: 10.1002/jcla.22988] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/03/2019] [Accepted: 07/05/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Wilms tumor, derived from embryonic cells, accounts for a large proportion of pediatric renal tumors. MYCN encoded by MYCN proto-oncogene, a member of the MYC family, is a BHLH transcription factor. It plays a critical role in tumorigenesis and predicts poor clinical outcomes in various types of cancer. However, the role of MYCN remained unclarified in Wilms tumor. In this study, we investigated the association between MYCN gene polymorphisms and Wilms tumor susceptibility. METHODS Four MYCN gene polymorphisms (rs57961569 G > A, rs9653226 T > C, rs13034994 A > G, and rs60226897 G > A) were genotyped in 183 cases and 603 controls. Adjusted odds ratios (AORs) and 95% confidence intervals (CIs) were calculated to evaluate the association between MYCN gene polymorphisms and Wilms tumor susceptibility. RESULTS Overall, no significant association was found for any of the four MYCN gene polymorphisms. Interestingly, in the stratification analysis, the rs57961569 was found to be associated with decreased Wilms tumor susceptibility in the children older than 18 months (AOR = 0.65, 95% CI = 0.42-1.00, P = .050). Moreover, older children carrying 2-4 risk genotypes were at increased risk of Wilms tumor (OR = 1.55, 95% CI = 1.001-2.40, P = .0497). Haplotype GCAA was shown to significantly increased Wilms tumor risk (AOR = 2.40, 95% CI = 1.12-5.14, P = .024). CONCLUSION Our study demonstrated that these MYCN gene polymorphisms might be low penetrant variants in Wilms tumor.
Collapse
Affiliation(s)
- Xiaokai Huang
- Department of HematologyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Jie Zhao
- Department of HematologyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Jinhong Zhu
- Department of Clinical LaboratoryBiobankHarbin Medical University Cancer HospitalHarbinChina
| | - Shanshan Chen
- Department of HematologyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Wen Fu
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhouChina
| | - Xiaoqian Tian
- Department of HematologyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Susu Lou
- Department of HematologyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Jichen Ruan
- Department of HematologyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Jing He
- Department of HematologyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhouChina
| | - Haixia Zhou
- Department of HematologyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| |
Collapse
|
24
|
MYCN Amplified Relapse Following Resolution of MYCN Nonamplified 4S Neuroblastoma With Placental Involvement: A Case Report and Review of the Literature. J Pediatr Hematol Oncol 2019; 41:388-391. [PMID: 31094905 DOI: 10.1097/mph.0000000000001515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Congenital neuroblastoma with placental involvement is exceptionally rare, but mortality is high. Detailed examination of placenta including MYCN amplification and segmental chromosomal aberrations should be performed in all suspected cases, as it is noninvasive and readily available. Maternal dissemination has not been reported. In this manuscript, we describe an infant with placental diagnosis of MYCN nonamplified congenital neuroblastoma. This is the first report of a recurrence of congenital 4S neuroblastoma following resolution in which MYCN amplification is only detected in the recurrence. Germline sequencing using a large comprehensive cancer panel did not reveal variants in candidate cancer predisposition genes.
Collapse
|