1
|
Simon J, Reita D, Guerin E, Lhermitte B, Weingertner N, Lefebvre F, Karanian M, Masliah-Planchon J, Lindner V, Onea A, Jannier S, Salmon A, Bergthold G, Vincent F, Deschuyter M, Barbaza MO, Entz-Werlé N. Clinical impact of large genomic explorations at diagnosis in 198 pediatric solid tumors: a monocentric study aiming practical feasibility of precision oncology. BMC Cancer 2024; 24:1296. [PMID: 39433989 PMCID: PMC11492794 DOI: 10.1186/s12885-024-13034-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 10/07/2024] [Indexed: 10/23/2024] Open
Abstract
INTRODUCTION Faced to the growing development of collecting systematic molecular analyses in relapsed pediatric cancers to transform their targeted matched therapies, this study aimed to assess the clinical and therapeutic indications of systematic diagnostic genomic explorations performed in pediatric solid cancers to determine which type of screening and if it afford at relapse time an accurate targeted strategy. METHODS A total of 280 patients less than 22 years, referred at the University Hospitals of Strasbourg for a newly diagnosed solid tumor from January 2015 to December 2021, were prospectively genomically investigated since diagnosis. Using 7 different molecular tests going from single-gene methods (IHC, FISH, RT-PCR, Sanger sequencing, droplet digital PCR) to largescale analyses (Next-Generation sequencing, RNAsequencing and FoundationOne®CDx), we explored retrospectively the molecular findings in those pediatric solid tumors (except hematolymphoid cancers) to improve diagnosis, prognosis assessment and relapse therapeutics. RESULTS One hundred and ninety-eight patients (71%) underwent molecular biology (MB) at diagnosis. Thirty-eight different histologies were grouped into cerebral tumors (30%), sarcomas (26%, bone and soft tissues), various blastomas (27%), and other entities (17%). Over a median 40-month follow-up, the overall survival rate of patients was 85% and the relapse rate 28%. Of the 326 analyses carried out, 245 abnormalities (single nucleotide variations: 50%, fusions: 25%, copy number alteration: 20%) concerning 70 oncogenes were highlighted. The overall clinical impact rate was 84%. Broad-spectrum analyses had a higher therapeutic impact (57%) than the targeted analyses (28%). 75% of broad-spectrum tests found an actionable variant conducting 23% of patients to receive rapidly a matched targeted therapy since first relapse. CONCLUSION Our experience highlighted the clinical utility of molecular profiling of solid tumors as soon as at diagnosis in children to expect improving access to innovative agents at relapse.
Collapse
Affiliation(s)
- Juliette Simon
- Pediatric Onco-Hematology Department, University Hospitals of Strasbourg, 1 Avenue Molière, Strasbourg, France
| | - Damien Reita
- Department of Cancer Molecular Genetics, Laboratory of Biochemistry and Molecular Biology, University Hospitals of Strasbourg, Strasbourg, France
- Laboratory of Bioimaging and Pathologies - Team OnKO-3T - Translational, Transversal and Therapeutic Oncology - UMR CNRS 7021, University of Strasbourg, Strasbourg, France
| | - Eric Guerin
- Department of Cancer Molecular Genetics, Laboratory of Biochemistry and Molecular Biology, University Hospitals of Strasbourg, Strasbourg, France
- Laboratory of Bioimaging and Pathologies - Team OnKO-3T - Translational, Transversal and Therapeutic Oncology - UMR CNRS 7021, University of Strasbourg, Strasbourg, France
| | - Benoit Lhermitte
- Laboratory of Bioimaging and Pathologies - Team OnKO-3T - Translational, Transversal and Therapeutic Oncology - UMR CNRS 7021, University of Strasbourg, Strasbourg, France
- Department of Pathology, University Hospitals of Strasbourg, Strasbourg, France
- Centre de Ressources Biologiques (CRB), University Hospitals of Strasbourg, Strasbourg, France
| | - Noelle Weingertner
- Department of Pathology, University Hospitals of Strasbourg, Strasbourg, France
| | - François Lefebvre
- Public Health Unit, University Hospitals of Strasbourg, Strasbourg, France
| | - Marie Karanian
- Department of Biopathology, Léon Berard Center, Lyon, France
| | | | - Veronique Lindner
- Department of Pathology, University Hospitals of Strasbourg, Strasbourg, France
| | - Alina Onea
- Department of Pathology, University Hospitals of Strasbourg, Strasbourg, France
| | - Sarah Jannier
- Pediatric Onco-Hematology Department, University Hospitals of Strasbourg, 1 Avenue Molière, Strasbourg, France
| | - Alexandra Salmon
- Pediatric Onco-Hematology Department, University Hospitals of Strasbourg, 1 Avenue Molière, Strasbourg, France
| | - Guillaume Bergthold
- Pediatric Onco-Hematology Department, University Hospitals of Strasbourg, 1 Avenue Molière, Strasbourg, France
| | - Florence Vincent
- Pediatric Onco-Hematology Department, University Hospitals of Strasbourg, 1 Avenue Molière, Strasbourg, France
| | - Marlène Deschuyter
- Laboratory of Bioimaging and Pathologies - Team OnKO-3T - Translational, Transversal and Therapeutic Oncology - UMR CNRS 7021, University of Strasbourg, Strasbourg, France
| | | | - Natacha Entz-Werlé
- Pediatric Onco-Hematology Department, University Hospitals of Strasbourg, 1 Avenue Molière, Strasbourg, France.
- Laboratory of Bioimaging and Pathologies - Team OnKO-3T - Translational, Transversal and Therapeutic Oncology - UMR CNRS 7021, University of Strasbourg, Strasbourg, France.
| |
Collapse
|
2
|
Murakami K, Tago SI, Takishita S, Morikawa H, Kojima R, Yokoyama K, Ogawa M, Fukushima H, Takamori H, Nannya Y, Imoto S, Fuji M. Pathogenicity Prediction of Gene Fusion in Structural Variations: A Knowledge Graph-Infused Explainable Artificial Intelligence (XAI) Framework. Cancers (Basel) 2024; 16:1915. [PMID: 38791993 PMCID: PMC11120556 DOI: 10.3390/cancers16101915] [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: 04/04/2024] [Revised: 04/26/2024] [Accepted: 05/01/2024] [Indexed: 05/26/2024] Open
Abstract
When analyzing cancer sample genomes in clinical practice, many structural variants (SVs), other than single nucleotide variants (SNVs), have been identified. To identify driver variants, the leading candidates must be narrowed down. When fusion genes are involved, selection is particularly difficult, and highly accurate predictions from AI is important. Furthermore, we also wanted to determine how the prediction can make more reliable diagnoses. Here, we developed an explainable AI (XAI) suitable for SVs with gene fusions, based on the XAI technology we previously developed for the prediction of SNV pathogenicity. To cope with gene fusion variants, we added new data to the previous knowledge graph for SVs and we improved the algorithm. Its prediction accuracy was as high as that of existing tools. Moreover, our XAI could explain the reasons for these predictions. We used some variant examples to demonstrate that the reasons are plausible in terms of pathogenic basic mechanisms. These results can be seen as a hopeful step toward the future of genomic medicine, where efficient and correct decisions can be made with the support of AI.
Collapse
Affiliation(s)
- Katsuhiko Murakami
- Computing Laboratories, Fujitsu Research, Fujitsu Ltd., Kawasaki 211-8588, Kanagawa, Japan
| | - Shin-ichiro Tago
- Computing Laboratories, Fujitsu Research, Fujitsu Ltd., Kawasaki 211-8588, Kanagawa, Japan
| | - Sho Takishita
- Computing Laboratories, Fujitsu Research, Fujitsu Ltd., Kawasaki 211-8588, Kanagawa, Japan
| | - Hiroaki Morikawa
- Computing Laboratories, Fujitsu Research, Fujitsu Ltd., Kawasaki 211-8588, Kanagawa, Japan
| | - Rikuhiro Kojima
- Computing Laboratories, Fujitsu Research, Fujitsu Ltd., Kawasaki 211-8588, Kanagawa, Japan
| | - Kazuaki Yokoyama
- Division of Hematopoietic Disease Control, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Miho Ogawa
- Division of Hematopoietic Disease Control, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- The University of Tokyo Hospital, The University of Tokyo, Tokyo 113-8655, Japan
| | - Hidehito Fukushima
- Division of Hematopoietic Disease Control, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Hiroyuki Takamori
- Division of Hematopoietic Disease Control, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Yasuhito Nannya
- Division of Hematopoietic Disease Control, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Seiya Imoto
- Division of Health Medical Intelligence, Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Masaru Fuji
- Computing Laboratories, Fujitsu Research, Fujitsu Ltd., Kawasaki 211-8588, Kanagawa, Japan
| |
Collapse
|
3
|
Sahu S, Rao AR, Sahu TK, Pandey J, Varshney S, Kumar A, Gaikwad K. Predictive Role of Cluster Bean ( Cyamopsis tetragonoloba) Derived miRNAs in Human and Cattle Health. Genes (Basel) 2024; 15:448. [PMID: 38674383 PMCID: PMC11049822 DOI: 10.3390/genes15040448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/22/2023] [Accepted: 09/11/2023] [Indexed: 04/28/2024] Open
Abstract
MicroRNAs (miRNAs) are small non-coding conserved molecules with lengths varying between 18-25nt. Plants miRNAs are very stable, and probably they might have been transferred across kingdoms via food intake. Such miRNAs are also called exogenous miRNAs, which regulate the gene expression in host organisms. The miRNAs present in the cluster bean, a drought tolerant legume crop having high commercial value, might have also played a regulatory role for the genes involved in nutrients synthesis or disease pathways in animals including humans due to dietary intake of plant parts of cluster beans. However, the predictive role of miRNAs of cluster beans for gene-disease association across kingdoms such as cattle and humans are not yet fully explored. Thus, the aim of the present study is to (i) find out the cluster bean miRNAs (cb-miRs) functionally similar to miRNAs of cattle and humans and predict their target genes' involvement in the occurrence of complex diseases, and (ii) identify the role of cb-miRs that are functionally non-similar to the miRNAs of cattle and humans and predict their targeted genes' association with complex diseases in host systems. Here, we predicted a total of 33 and 15 functionally similar cb-miRs (fs-cb-miRs) to human and cattle miRNAs, respectively. Further, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis revealed the participation of targeted genes of fs-cb-miRs in 24 and 12 different pathways in humans and cattle, respectively. Few targeted genes in humans like LCP2, GABRA6, and MYH14 were predicted to be associated with disease pathways of Yesinia infection (hsa05135), neuroactive ligand-receptor interaction (hsa04080), and pathogenic Escherichia coli infection (hsa05130), respectively. However, targeted genes of fs-cb-miRs in humans like KLHL20, TNS1, and PAPD4 are associated with Alzheimer's, malignant tumor of the breast, and hepatitis C virus infection disease, respectively. Similarly, in cattle, targeted genes like ATG2B and DHRS11 of fs-cb-miRs participate in the pathways of Huntington disease and steroid biosynthesis, respectively. Additionally, the targeted genes like SURF4 and EDME2 of fs-cb-miRs are associated with mastitis and bovine osteoporosis, respectively. We also found a few cb-miRs that do not have functional similarity with human and cattle miRNAs but are found to target the genes in the host organisms and as well being associated with human and cattle diseases. Interestingly, a few genes such as NRM, PTPRE and SUZ12 were observed to be associated with Rheumatoid Arthritis, Asthma and Endometrial Stromal Sarcoma diseases, respectively, in humans and genes like SCNN1B associated with renal disease in cattle.
Collapse
Affiliation(s)
- Sarika Sahu
- Indian Agricultural Statistics Research Institute, ICAR, New Delhi 110012, India; (S.S.); (J.P.); (S.V.)
- Amity Institute of Biotechnology, Amity University, Noida 201303, India;
| | - Atmakuri Ramakrishna Rao
- Indian Agricultural Statistics Research Institute, ICAR, New Delhi 110012, India; (S.S.); (J.P.); (S.V.)
- Indian Council of Agricultural Research, New Delhi 110001, India
| | - Tanmaya Kumar Sahu
- Indian Grassland and Fodder Research Institute, ICAR, Jhansi 284003, India;
| | - Jaya Pandey
- Indian Agricultural Statistics Research Institute, ICAR, New Delhi 110012, India; (S.S.); (J.P.); (S.V.)
| | - Shivangi Varshney
- Indian Agricultural Statistics Research Institute, ICAR, New Delhi 110012, India; (S.S.); (J.P.); (S.V.)
| | - Archna Kumar
- Amity Institute of Biotechnology, Amity University, Noida 201303, India;
| | - Kishor Gaikwad
- National Institute for Plant Biotechnology, ICAR, New Delhi 110012, India;
| |
Collapse
|
4
|
Krynina O, de Ståhl TD, Jylhä C, Arthur C, Giraud G, Nyman P, Fritzberg A, Sandgren J, Tham E, Sandvik U. The potential of liquid biopsy for detection of the KIAA1549-BRAF fusion in circulating tumor DNA from children with pilocytic astrocytoma. Neurooncol Adv 2024; 6:vdae008. [PMID: 38371226 PMCID: PMC10874216 DOI: 10.1093/noajnl/vdae008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2024] Open
Abstract
Background Low-grade gliomas (LGGs) represent children's most prevalent central nervous system tumor, necessitating molecular profiling to diagnose and determine the most suitable treatment. Developing highly sensitive screening techniques for liquid biopsy samples is particularly beneficial, as it enables the early detection and molecular characterization of tumors with minimally invasive samples. Methods We examined CSF and plasma samples from patients with pilocytic astrocytoma (PA) using custom multiplexed droplet digital polymerase chain reaction (ddPCR) assays based on whole genome sequencing data. These assays included a screening test to analyze BRAF duplication and a targeted assay for the detection of patient-specific KIAA1549::BRAF fusion junction sequences or single nucleotide variants. Results Our findings revealed that 5 out of 13 individual cerebrospinal fluid (CSF) samples tested positive for circulating tumor DNA (ctDNA). Among these cases, 3 exhibited the KIAA1549::BRAF fusion, which was detected through copy number variation (CNV) analysis (n = 1) or a fusion-specific probe (n = 2), while 1 case each displayed the BRAF V600E mutation and the FGFR1 N577K mutation. Additionally, a quantitative analysis of cell-free DNA (cfDNA) concentrations in PA CSF samples showed that most cases had low cfDNA levels, below the limit of detection of our assay (<1.9 ng). Conclusions While CNV analysis of CSF samples from LGGs still has some limitations, it has the potential to serve as a valuable complementary tool. Furthermore, it can also be multiplexed with other aberrations, for example, to the BRAF V600 test, to provide important insights into the molecular characteristics of LGGs.
Collapse
Affiliation(s)
- Olha Krynina
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | | | - Cecilia Jylhä
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Clinical Genetics and Genomics, Karolinska University Hospital, Stockholm, Sweden
| | - Cecilia Arthur
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Clinical Genetics and Genomics, Karolinska University Hospital, Stockholm, Sweden
| | - Geraldine Giraud
- Department of Immunology, Genetic and Pathology, Neuro-oncology, and Neurodegeneration Program Rudbeck Laboratory, Uppsala, Sweden
- Department of Women and Children’s Health, Akademiska University Hospital, Uppsala, Sweden
| | - Per Nyman
- Department of Health, Crown Princess Victoria Children´s Hospital, Linköping University Hospital, Linköping, Sweden
- Department of Medicine and Caring Sciences, Linköping University, Linköping, Sweden
- Centre for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - Anders Fritzberg
- Daycare Unit of Oncology and Hematology, Clinic of Pediatrics Falun Hospital, Dalarna Region, Sweden
| | - Johanna Sandgren
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| | - Emma Tham
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Clinical Genetics and Genomics, Karolinska University Hospital, Stockholm, Sweden
| | - Ulrika Sandvik
- Department of Clinical Neuroscience, Division of Neurosurgery, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
5
|
Selt F, El Damaty A, Schuhmann MU, Sigaud R, Ecker J, Sievers P, Kocher D, Herold-Mende C, Oehme I, von Deimling A, Pfister SM, Sahm F, Jones DTW, Witt O, Milde T. Generation of patient-derived pediatric pilocytic astrocytoma in-vitro models using SV40 large T: evaluation of a modeling workflow. J Neurooncol 2023; 165:467-478. [PMID: 37999877 PMCID: PMC10752915 DOI: 10.1007/s11060-023-04500-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 11/02/2023] [Indexed: 11/25/2023]
Abstract
PURPOSE Although pediatric low-grade gliomas (pLGG) are the most common pediatric brain tumors, patient-derived cell lines reflecting pLGG biology in culture are scarce. This also applies to the most common pLGG subtype pilocytic astrocytoma (PA). Conventional cell culture approaches adapted from higher-grade tumors fail in PA due to oncogene-induced senescence (OIS) driving tumor cells into arrest. Here, we describe a PA modeling workflow using the Simian Virus large T antigen (SV40-TAg) to circumvent OIS. METHODS 18 pLGG tissue samples (17 (94%) histological and/or molecular diagnosis PA) were mechanically dissociated. Tumor cell positive-selection using A2B5 was perfomed in 8/18 (44%) cases. All primary cell suspensions were seeded in Neural Stem Cell Medium (NSM) and Astrocyte Basal Medium (ABM). Resulting short-term cultures were infected with SV40-TAg lentivirus. Detection of tumor specific alterations (BRAF-duplication and BRAF V600E-mutation) by digital droplet PCR (ddPCR) at defined time points allowed for determination of tumor cell fraction (TCF) and evaluation of the workflow. DNA-methylation profiling and gene-panel sequencing were used for molecular profiling of primary samples. RESULTS Primary cell suspensions had a mean TCF of 55% (+/- 23% (SD)). No sample in NSM (0/18) and ten samples in ABM (10/18) were successfully transduced. Three of these ten (30%) converted into long-term pLGG cell lines (TCF 100%), while TCF declined to 0% (outgrowth of microenvironmental cells) in 7/10 (70%) cultures. Young patient age was associated with successful model establishment. CONCLUSION A subset of primary PA cultures can be converted into long-term cell lines using SV40-TAg depending on sample intrinsic (patient age) and extrinsic workflow-related (e.g. type of medium, successful transduction) parameters. Careful monitoring of sample-intrinsic and extrinsic factors optimizes the process.
Collapse
Affiliation(s)
- Florian Selt
- Hopp Children's Cancer Center Heidelberg (KiTZ), Im Neuenheimer Feld 430, 69120, Heidelberg, Germany.
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany.
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany.
- National Center for Tumor Diseases (NCT), Heidelberg, Germany.
| | - Ahmed El Damaty
- Hopp Children's Cancer Center Heidelberg (KiTZ), Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
- Pediatric Neurosurgery Division, Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Martin U Schuhmann
- Section of Pediatric Neurosurgery, Department of Neurosurgery, University Hospital Tübingen, Tübingen, Germany
| | - Romain Sigaud
- Hopp Children's Cancer Center Heidelberg (KiTZ), Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Jonas Ecker
- Hopp Children's Cancer Center Heidelberg (KiTZ), Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Philipp Sievers
- Hopp Children's Cancer Center Heidelberg (KiTZ), Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniela Kocher
- Hopp Children's Cancer Center Heidelberg (KiTZ), Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | | | - Ina Oehme
- Hopp Children's Cancer Center Heidelberg (KiTZ), Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Andreas von Deimling
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan M Pfister
- Hopp Children's Cancer Center Heidelberg (KiTZ), Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Felix Sahm
- Hopp Children's Cancer Center Heidelberg (KiTZ), Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - David T W Jones
- Hopp Children's Cancer Center Heidelberg (KiTZ), Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Olaf Witt
- Hopp Children's Cancer Center Heidelberg (KiTZ), Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Till Milde
- Hopp Children's Cancer Center Heidelberg (KiTZ), Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
| |
Collapse
|
6
|
Sahm F, Brandner S, Bertero L, Capper D, French PJ, Figarella-Branger D, Giangaspero F, Haberler C, Hegi ME, Kristensen BW, Kurian KM, Preusser M, Tops BBJ, van den Bent M, Wick W, Reifenberger G, Wesseling P. Molecular diagnostic tools for the World Health Organization (WHO) 2021 classification of gliomas, glioneuronal and neuronal tumors; an EANO guideline. Neuro Oncol 2023; 25:1731-1749. [PMID: 37279174 PMCID: PMC10547522 DOI: 10.1093/neuonc/noad100] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Indexed: 06/08/2023] Open
Abstract
In the 5th edition of the WHO CNS tumor classification (CNS5, 2021), multiple molecular characteristics became essential diagnostic criteria for many additional CNS tumor types. For those tumors, an integrated, "histomolecular" diagnosis is required. A variety of approaches exists for determining the status of the underlying molecular markers. The present guideline focuses on the methods that can be used for assessment of the currently most informative diagnostic and prognostic molecular markers for the diagnosis of gliomas, glioneuronal and neuronal tumors. The main characteristics of the molecular methods are systematically discussed, followed by recommendations and information on available evidence levels for diagnostic measures. The recommendations cover DNA and RNA next-generation-sequencing, methylome profiling, and select assays for single/limited target analyses, including immunohistochemistry. Additionally, because of its importance as a predictive marker in IDH-wildtype glioblastomas, tools for the analysis of MGMT promoter methylation status are covered. A structured overview of the different assays with their characteristics, especially their advantages and limitations, is provided, and requirements for input material and reporting of results are clarified. General aspects of molecular diagnostic testing regarding clinical relevance, accessibility, cost, implementation, regulatory, and ethical aspects are discussed as well. Finally, we provide an outlook on new developments in the landscape of molecular testing technologies in neuro-oncology.
Collapse
Affiliation(s)
- Felix Sahm
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany
- CCU Neuropathology, German Concortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sebastian Brandner
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology and Division of Neuropathology, The National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - Luca Bertero
- Pathology Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| | - David Capper
- Department of Neuropathology, Charité, Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Pim J French
- Department of Neurology, Brain Tumor Center at Erasmus MC Cancer Center, 3015 GD Rotterdam, The Netherlands
| | - Dominique Figarella-Branger
- Aix-Marseille University, APHM, CNRS, INP, Institute Neurophysiopathol, CHU Timone, Service d’Anatomie Pathologique et de Neuropathologie, Marseille, France
| | - Felice Giangaspero
- Department of Radiological, Oncological and Anatomo-Pathological Sciences, University Sapienza of Rome, Rome, Italy
| | - Christine Haberler
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Austria
| | - Monika E Hegi
- Neuroscience Research Center and Neurosurgery, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Bjarne W Kristensen
- Department of Clinical Medicine and Biotech Research and Innovation Center (BRIC), University of Copenhagen, Denmark
- Department of Pathology, The Bartholin Institute, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | | | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Austria
| | - Bastiaan B J Tops
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Martin van den Bent
- The Brain Tumor Center at Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Wolfgang Wick
- Department of Neurology and Neurooncology Program, National Center for Tumor Diseases, Heidelberg University Hospital
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Guido Reifenberger
- Institute of Neuropathology, Heinrich Heine University, Medical Faculty, and University Hospital Düsseldorf, and German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Düsseldorf, Germany
| | - Pieter Wesseling
- Department of Pathology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands (P.W.)
| |
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
|
Candido MF, Medeiros M, Veronez LC, Bastos D, Oliveira KL, Pezuk JA, Valera ET, Brassesco MS. Drugging Hijacked Kinase Pathways in Pediatric Oncology: Opportunities and Current Scenario. Pharmaceutics 2023; 15:pharmaceutics15020664. [PMID: 36839989 PMCID: PMC9966033 DOI: 10.3390/pharmaceutics15020664] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023] Open
Abstract
Childhood cancer is considered rare, corresponding to ~3% of all malignant neoplasms in the human population. The World Health Organization (WHO) reports a universal occurrence of more than 15 cases per 100,000 inhabitants around the globe, and despite improvements in diagnosis, treatment and supportive care, one child dies of cancer every 3 min. Consequently, more efficient, selective and affordable therapeutics are still needed in order to improve outcomes and avoid long-term sequelae. Alterations in kinases' functionality is a trademark of cancer and the concept of exploiting them as drug targets has burgeoned in academia and in the pharmaceutical industry of the 21st century. Consequently, an increasing plethora of inhibitors has emerged. In the present study, the expression patterns of a selected group of kinases (including tyrosine receptors, members of the PI3K/AKT/mTOR and MAPK pathways, coordinators of cell cycle progression, and chromosome segregation) and their correlation with clinical outcomes in pediatric solid tumors were accessed through the R2: Genomics Analysis and Visualization Platform and by a thorough search of published literature. To further illustrate the importance of kinase dysregulation in the pathophysiology of pediatric cancer, we analyzed the vulnerability of different cancer cell lines against their inhibition through the Cancer Dependency Map portal, and performed a search for kinase-targeted compounds with approval and clinical applicability through the CanSAR knowledgebase. Finally, we provide a detailed literature review of a considerable set of small molecules that mitigate kinase activity under experimental testing and clinical trials for the treatment of pediatric tumors, while discuss critical challenges that must be overcome before translation into clinical options, including the absence of compounds designed specifically for childhood tumors which often show differential mutational burdens, intrinsic and acquired resistance, lack of selectivity and adverse effects on a growing organism.
Collapse
Affiliation(s)
- Marina Ferreira Candido
- Department of Cell Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - Mariana Medeiros
- Regional Blood Center, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - Luciana Chain Veronez
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - David Bastos
- Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-901, SP, Brazil
| | - Karla Laissa Oliveira
- Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-901, SP, Brazil
| | - Julia Alejandra Pezuk
- Departament of Biotechnology and Innovation, Anhanguera University of São Paulo, UNIAN/SP, São Paulo 04119-001, SP, Brazil
| | - Elvis Terci Valera
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - María Sol Brassesco
- Departament of Biotechnology and Innovation, Anhanguera University of São Paulo, UNIAN/SP, São Paulo 04119-001, SP, Brazil
- Correspondence: ; Tel.: +55-16-3315-9144; Fax: +55-16-3315-4886
| |
Collapse
|
9
|
AlShail E, Alahmari AN, Dababo AAM, Alsagob M, Al-Hindi H, Khalil H, Al Masseri Z, AlSalamah R, Almohseny E, Alduhaish A, Colak D, Kaya N. A molecular study of pediatric pilomyxoid and pilocytic astrocytomas: Genome-wide copy number screening, retrospective analysis of clinicopathological features and long-term clinical outcome. Front Oncol 2023; 13:1034292. [PMID: 36860324 PMCID: PMC9968872 DOI: 10.3389/fonc.2023.1034292] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 01/13/2023] [Indexed: 02/17/2023] Open
Abstract
Background Pilocytic Astrocytoma (PA) is the most common pediatric brain tumors. PAs are slow-growing tumors with high survival rates. However, a distinct subgroup of tumors defined as pilomyxoid astrocytoma (PMA) presents unique histological characteristics and have more aggressive clinical course. The studies on genetics of PMA are scarce. Methods In this study, we report one of the largest cohort of pediatric patients with pilomyxoid (PMA) and pilocytic astrocytomas (PA) in Saudi population providing a comprehensive clinical picture, retrospective analysis with long-term follow-up, genome-wide copy number changes, and clinical outcome of these pediatric tumors. We examined and compared genome-wide copy number aberrations (CNAs) and the clinical outcome of the patients with PA and PMA. Results The median progression free survival for the whole cohort was 156 months and it was 111 months for the PMA, however, not statistically significantly different between the groups (log-rank test, P = 0.726). We have identified 41 CNAs (34 gains and 7 losses) in all tested patients. Our study yielded the previously reported KIAA1549-BRAF Fusion gene in over 88% of the tested patients (89% and 80% in PMA and PA, respectively). Besides the fusion gene, twelve patients had additional genomic CNAs. Furthermore, pathway and gene network analyses of genes in the fusion region revealed alterations in retinoic acid mediated apoptosis and MAPK signaling pathways and key hub genes that may potentially be involved in tumor growth and progression, including BRAF, LUC7L2, MKRN1, RICTOR, TP53, HIPK2, HNF4A, POU5F, and SOX4. Conclusion Our study is the first report of a large cohort of patients with PMA and PA in the Saudi population that provides detailed clinical features, genomic copy number changes, and outcome of these pediatric tumors and may help better diagnosis and characterization of PMA.
Collapse
Affiliation(s)
- Essam AlShail
- Department of Neurosciences, King Faisal Specialist Hospital and Research Centre (KFSHRC), Riyadh, Saudi Arabia
| | - Ahmed Nasser Alahmari
- Department of Neurosciences, King Faisal Specialist Hospital and Research Centre (KFSHRC), Riyadh, Saudi Arabia
| | - Anas A. M. Dababo
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Centre (KFSHRC), Riyadh, Saudi Arabia
| | - Maysoon Alsagob
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Centre (KFSHRC), Riyadh, Saudi Arabia,Applied Genomics Technologies Institute, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Hindi Al-Hindi
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Centre (KFSHRC), Riyadh, Saudi Arabia
| | - Hala Khalil
- Department of Biostatistics, Epidemiology and Scientific Computing, King Faisal Specialist Hospital and Research Centre (KFSHRC), Riyadh, Saudi Arabia
| | - Zainab Al Masseri
- Medical Genetics Department, King Faisal Specialist Hospital and Research Centre (KFSHRC), Riyadh, Saudi Arabia
| | - Razan AlSalamah
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Centre (KFSHRC), Riyadh, Saudi Arabia
| | - Ethar Almohseny
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Centre (KFSHRC), Riyadh, Saudi Arabia
| | - Amjad Alduhaish
- Neuroscience Department, King Abdullah Medical City, Mecca, Saudi Arabia
| | - Dilek Colak
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre (KFSHRC), Riyadh, Saudi Arabia,*Correspondence: Namik Kaya, ; ; Dilek Colak,
| | - Namik Kaya
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Centre (KFSHRC), Riyadh, Saudi Arabia,*Correspondence: Namik Kaya, ; ; Dilek Colak,
| |
Collapse
|
10
|
Tsuriel S, Hannes V, Hasona A, Raz M, Hershkovitz D. Digital PCR-Based Method for Detecting CDKN2A Loss in Brain Tumours. Mol Diagn Ther 2022; 26:689-698. [PMID: 36129665 DOI: 10.1007/s40291-022-00610-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2022] [Indexed: 12/30/2022]
Abstract
INTRODUCTION CDKN2A is a key tumour suppressor gene and loss of CDKN2A can be found in many tumours. In astrocytoma grade IV, CDKN2A is deleted in more than 50% of tumours. In many instances, low-grade gliomas with homozygous loss of CDKN2A behave like high grade tumours. The available techniques for CDKN2A loss are laborious, expensive, unreliable, or unavailable in most pathology institutes. Therefore, although it is essential for accurate brain tumour diagnosis, the routine diagnosis does not include testing for CDKN2A deletion. METHODS We developed a digital polymerase chain reaction (dPCR) assay for CDKN2A loss detection. The assay is based on counting the copy number of CDKN2A gene and of a reference gene on the same chromosome. It was tested for the detection limit with regard to tumour content and minimal DNA quantity. It was then tested on 24 clinical samples with known CDKN2A status. Additionally, we tested 44 gliomas with unknown CDKN2A status. RESULTS We found that the newly developed assay is reliable in tissue with more than 50% tumour content and more than 0.4 ng of DNA. The validation cohort showed complete concordance, and we were able to detect homozygous loss in 16 gliomas with unknown CDKN2A status. DISCUSSION The method presented can give a fast, cost-effective, clinically reliable evaluation of CDKN2A loss in tissue with more than 50% tumour content. Its ability to work with old samples and with low amounts of DNA makes it the favoured assay in cases where other techniques fail.
Collapse
Affiliation(s)
- Shlomo Tsuriel
- Institute of Pathology, Tel-Aviv Sourasky Medical Center, 62431, Tel-Aviv, Israel.
| | - Victoria Hannes
- Institute of Pathology, Tel-Aviv Sourasky Medical Center, 62431, Tel-Aviv, Israel
| | - Asala Hasona
- Institute of Pathology, Tel-Aviv Sourasky Medical Center, 62431, Tel-Aviv, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, 69978, Tel-Aviv, Israel
| | - Michal Raz
- Institute of Pathology, Tel-Aviv Sourasky Medical Center, 62431, Tel-Aviv, Israel
| | - Dov Hershkovitz
- Institute of Pathology, Tel-Aviv Sourasky Medical Center, 62431, Tel-Aviv, Israel.
- Sackler Faculty of Medicine, Tel-Aviv University, 69978, Tel-Aviv, Israel.
| |
Collapse
|
11
|
Pagès M, Debily M, Fina F, Jones DTW, Saffroy R, Castel D, Blauwblomme T, Métais A, Bourgeois M, Lechapt‐Zalcman E, Tauziède‐Espariat A, Andreiuolo F, Chrétien F, Grill J, Boddaert N, Figarella‐Branger D, Beroukhim R, Varlet P. The genomic landscape of dysembryoplastic neuroepithelial tumours and a comprehensive analysis of recurrent cases. Neuropathol Appl Neurobiol 2022; 48:e12834. [PMID: 35836307 PMCID: PMC9542977 DOI: 10.1111/nan.12834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 05/24/2022] [Accepted: 07/07/2022] [Indexed: 11/28/2022]
Abstract
AIMS Dysembryoplastic neuroepithelial tumour (DNT) is a glioneuronal tumour that is challenging to diagnose, with a wide spectrum of histological features. Three histopathological patterns have been described: specific DNTs (both the simple form and the complex form) comprising the specific glioneuronal element, and also the non-specific/diffuse form which lacks it, and has unclear phenotype-genotype correlations with numerous differential diagnoses. METHODS We used targeted methods (immunohistochemistry, fluorescence in situ hybridisation and targeted sequencing) and large-scale genomic methodologies including DNA methylation profiling to perform an integrative analysis to better characterise a large retrospective cohort of 82 DNTs, enriched for tumours that showed progression on imaging. RESULTS We confirmed that specific DNTs are characterised by a single driver event with a high frequency of FGFR1 variants. However, a subset of DNA methylation-confirmed DNTs harbour alternative genomic alterations to FGFR1 duplication/mutation. We also demonstrated that a subset of DNTs sharing the same FGFR1 alterations can show in situ progression. In contrast to the specific forms, "non-specific/diffuse DNTs" corresponded to a heterogeneous molecular group encompassing diverse, newly-described, molecularly distinct entities. CONCLUSIONS Specific DNT is a homogeneous group of tumours sharing characteristics of paediatric low-grade gliomas: a quiet genome with a recurrent genomic alteration in the RAS-MAPK signalling pathway, a distinct DNA methylation profile and a good prognosis but showing progression in some cases. The "non-specific/diffuse DNTs" subgroup encompasses various recently described histomolecular entities, such as PLNTY and diffuse astrocytoma, MYB or MYBL1 altered.
Collapse
Affiliation(s)
- Mélanie Pagès
- GHU‐Paris – Sainte‐Anne Hospital, Department of NeuropathologyParis UniversityParisFrance
- Department of GeneticsInstitut CurieParisFrance
- SIREDO Paediatric Cancer CenterInstitut CurieParisFrance
- INSERM U830, Laboratory of Translational Research in Paediatric OncologyInstitut CurieParisFrance
- Paris Sciences Lettres Research UniversityParisFrance
| | - Marie‐Anne Debily
- Molecular Predictors and New Targets in Oncology, INSERM U981, Gustave RoussyUniversité Paris‐SaclayVillejuifFrance
- Département de Biologie, Univ. EvryUniversité Paris‐SaclayEvryFrance
| | - Frédéric Fina
- APHM, CHU TimoneService d'Anatomie Pathologique et de NeuropathologieMarseilleFrance
| | - David T. W. Jones
- Pediatric Glioma ResearchHopp Children's Cancer Center (KiTZ)HeidelbergGermany
- Pediatric Glioma Research GroupGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Raphael Saffroy
- Oncogenetics Department, Assistance Publique‐Hôpitaux de Paris, Paul Brousse HospitalUniversité Paris‐SaclayVillejuifFrance
| | - David Castel
- Molecular Predictors and New Targets in Oncology, INSERM U981, Gustave RoussyUniversité Paris‐SaclayVillejuifFrance
- Département de Biologie, Univ. EvryUniversité Paris‐SaclayEvryFrance
| | - Thomas Blauwblomme
- Pediatric Neurosurgery Department, AP‐HPHôpital Universitaire Necker‐Enfants MaladesParisFrance
- Université de Paris‐ CitéParisFrance
| | - Alice Métais
- GHU‐Paris – Sainte‐Anne Hospital, Department of NeuropathologyParis UniversityParisFrance
| | - Marie Bourgeois
- Pediatric Neurosurgery Department, AP‐HPHôpital Universitaire Necker‐Enfants MaladesParisFrance
| | | | | | - Felipe Andreiuolo
- Department of NeuropathologyInstituto Estadual do Cérebro Paulo NiemeyerRio de JaneiroBrazil
- Pathology Division, D'Or Research Institute (IDOR)D'Or Hospitals NetworkRio de JaneiroBrazil
| | - Fabrice Chrétien
- GHU‐Paris – Sainte‐Anne Hospital, Department of NeuropathologyParis UniversityParisFrance
- Université de Paris‐ CitéParisFrance
| | - Jacques Grill
- Molecular Predictors and New Targets in Oncology, INSERM U981, Gustave RoussyUniversité Paris‐SaclayVillejuifFrance
- Département de Biologie, Univ. EvryUniversité Paris‐SaclayEvryFrance
- Department of Pediatric and Adolescent OncologyInstitut Gustave RoussyVillejuifFrance
| | - Nathalie Boddaert
- Pediatric Radiology Department, AP‐HPHôpital Universitaire Necker‐Enfants MaladesParisFrance
- INSERM ERL UA10Université de ParisParisFrance
- Institut ImagineUniversité de Paris, UMR 1163ParisFrance
| | - Dominique Figarella‐Branger
- APHM, CHU TimoneService d'Anatomie Pathologique et de NeuropathologieMarseilleFrance
- Institute of NeuroPhysiopatholyAix‐Marseille Univ, CNRS, INPMarseilleFrance
| | - Rameen Beroukhim
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
- Cancer ProgramBroad InstituteCambridgeMassachusettsUSA
- Department of MedicineHarvard Medical SchoolBostonMassachusettsUSA
| | - Pascale Varlet
- GHU‐Paris – Sainte‐Anne Hospital, Department of NeuropathologyParis UniversityParisFrance
| |
Collapse
|
12
|
Yuan R, Bai X, Hu X, Zhang H, Hou C, Long Q, Luo Y. CATCH: high specific transcriptome-focused fusion gene variants discrimination. Chem Commun (Camb) 2022; 58:7618-7621. [PMID: 35730730 DOI: 10.1039/d2cc02660k] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
We develop a CRISPR-Cas13a triggered catalytic hairpin assembly (CATCH) approach for accurate and impartial identification of variants by integrating the fusion gene-selected recognition of CRISPR-Cas13a with collateral cleavage-assisted catalytic hairpin assembly amplification. This approach achieved an accuracy of 100% in a pilot experiment involving 34 clinical samples.
Collapse
Affiliation(s)
- Rui Yuan
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, P. R. China.,Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing 400044, P. R. China
| | - Xiaopeng Bai
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing 400016, P. R. China
| | - Xiaolin Hu
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing 400044, P. R. China
| | - Hong Zhang
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing 400044, P. R. China.,Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong 250033, P. R. China
| | - Changjun Hou
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, P. R. China
| | - Quanxin Long
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing 400016, P. R. China
| | - Yang Luo
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing 400044, P. R. China.,Department of Clinical Laboratory, Fuling Hospital, Chongqing University, Chongqing, 408099, P. R. China.,Department of Clinical Laboratory, Jiangjin Hospital, Chongqing University, Chongqing, 402260, P. R. China
| |
Collapse
|
13
|
Wolter M, Felsberg J, Malzkorn B, Kaulich K, Reifenberger G. Droplet digital PCR-based analyses for robust, rapid, and sensitive molecular diagnostics of gliomas. Acta Neuropathol Commun 2022; 10:42. [PMID: 35361262 PMCID: PMC8973808 DOI: 10.1186/s40478-022-01335-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 02/20/2022] [Indexed: 11/10/2022] Open
Abstract
Classification of gliomas involves the combination of histological features with molecular biomarkers to establish an integrated histomolecular diagnosis. Here, we report on the application and validation of a set of molecular assays for glioma diagnostics based on digital PCR technology using the QX200™ Droplet Digital™ PCR (ddPCR) system. The investigated ddPCR-based assays enable the detection of diagnostically relevant glioma-associated mutations in the IDH1, IDH2, H3-3A, BRAF, and PRKCA genes, as well as in the TERT promoter. In addition, ddPCR-based assays assessing diagnostically relevant copy number alterations were studied, including 1p/19q codeletion, gain of chromosome 7 and loss of chromosome 10 (+ 7/-10), EGFR amplification, duplication of the BRAF locus, and CDKN2A homozygous deletion. Results obtained by ddPCR were validated by other methods, including immunohistochemistry, Sanger sequencing, pyrosequencing, microsatellite analyses for loss of heterozygosity, as well as real-time PCR- or microarray-based copy number assays. Particular strengths of the ddPCR approach are (1) its high analytical sensitivity allowing for reliable detection of mutations even with low mutant allele frequencies, (2) its quantitative determination of mutant allele frequencies and copy number changes, and (3) its rapid generation of results within a single day. Thus, in line with other recent studies our findings support ddPCR analysis as a valuable approach for molecular glioma diagnostics in a fast, quantitative and highly sensitive manner.
Collapse
|
14
|
Dandapath I, Chakraborty R, Kaur K, Mahajan S, Singh J, Sharma MC, Sarkar C, Suri V. Molecular alterations of low-grade gliomas in young patients: Strategies and platforms for routine evaluation. Neurooncol Pract 2021; 8:652-661. [PMID: 34777834 PMCID: PMC8579091 DOI: 10.1093/nop/npab053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In recent years, it has been established that molecular biology of pediatric low-grade gliomas (PLGGs) is entirely distinct from adults. The majority of the circumscribed pediatric gliomas are driven by mitogen-activated protein kinase (MAPK) pathway, which has yielded important diagnostic, prognostic, and therapeutic biomarkers. Further, the Consortium to Inform Molecular and Practical Approaches to CNS Tumor Taxonomy (cIMPACT) Steering Committee in their fourth meeting, suggested including a panel of molecular markers for integrated diagnosis in "pediatric-type" diffuse gliomas. However, a designated set of platforms for the evaluation of these alterations has yet not been mentioned for easier implementation in routine molecular diagnostics. Herein, we have reviewed the relevance of analyzing these markers and discussed the strategies and platforms best apposite for clinical laboratories.
Collapse
Affiliation(s)
- Iman Dandapath
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | | | - Kavneet Kaur
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Swati Mahajan
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Jyotsna Singh
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Mehar C Sharma
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Chitra Sarkar
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Vaishali Suri
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
15
|
Gu J, Wang Y, Yu J. Cerebral astroblastoma with oligodendroglial-like cells: A case report. Medicine (Baltimore) 2021; 100:e27570. [PMID: 34713831 PMCID: PMC8556017 DOI: 10.1097/md.0000000000027570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 10/07/2021] [Indexed: 01/05/2023] Open
Abstract
RATIONALE Astroblastoma is a rare tumor of the central nervous system with uncertain biological behavior and origin. Its histopathological features have been well established, while, to our knowledge, astroblastoma with oligodendroglial-like cells have not been reported. PATIENT CONCERNS A 15-year-old girl presented with nausea, vomiting, headache, and visual disturbance. DIAGNOSIS Magnetic resonance imaging revealed a large neoplasm in the left temporal. Histologically, the tumor showed solid and pseudopapillary structure. Immunohistochemical staining showed that the tumor cells were positive for glial fibrillary acidic protein and vimentin. The oligodendroglial-like cells were positive for glial fibrillary acidic protein, vimentin, and oligodendrocyte transcription factor 2. The antigen KI67 labeling index was about 4%. Sequencing for isocitrate dehydrogenase (IDH) 1 codon 132 and IDH2 codon 172 gene mutations showed negative results. Furthermore, fluorescent analysis revealed neither 1p nor 19q deletion in the lesion. Based on these findings, the girl was finally diagnosed as astroblastoma. INTERVENTIONS A craniotomy with total excision of the tumor was performed. OUTCOMES The follow-up time was 1 year, no evidence of disease recurrence was found in magnetic resonance imaging. LESSONS Cerebral astroblastoma with oligodendroglial-like cells is a clinically rare tumor of central nervous system. Clear distinction and diagnosis are critical.
Collapse
Affiliation(s)
- Jian Gu
- Department of Pathology, the First Affiliated Hospital of China Medical University,Shenyang, China
- Department of Pathology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yihua Wang
- Department of Pathology, the First Affiliated Hospital of China Medical University,Shenyang, China
- Department of Pathology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Juanhan Yu
- Department of Pathology, the First Affiliated Hospital of China Medical University,Shenyang, China
- Department of Pathology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| |
Collapse
|
16
|
Métais A, Appay R, Pagès M, Gallardo C, Silva K, Siegfried A, Perbet R, Maurage CA, Scavarda D, Fina F, Uro-Coste E, Riffaud L, Colin C, Figarella-Branger D. Low-grade epilepsy-associated neuroepithelial tumours with a prominent oligodendroglioma-like component: The diagnostic challenges. Neuropathol Appl Neurobiol 2021; 48:e12769. [PMID: 34551121 DOI: 10.1111/nan.12769] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/23/2021] [Accepted: 09/12/2021] [Indexed: 12/24/2022]
Abstract
AIMS We searched for recurrent pathological features and molecular alterations in a retrospective series of 72 low-grade epilepsy-associated neuroepithelial tumours (LEATs) with a prominent oligodendroglioma-like component, in order to classify them according to the 2021 World Health Organization (WHO) classification of central nervous system (CNS) tumours. METHODS Centralised pathological examination was performed as well as targeted molecular analysis of v-Raf murine sarcoma viral oncogene homologue B (BRAF) and fibroblast growth factor receptor 1 (FGFR1) by multiplexed digital polymerase chain reaction (mdPCR). DNA methylation profiling was performed in cases with sufficient DNA. In cases with no genetic alteration by mdPCR and sufficient material, RNA sequencing was done. RESULTS We first reclassified our cohort into three groups: ganglioglioma (GG, n = 14), dysembryoplastic neuroepithelial tumours (DNTs, n = 19) and glioneuronal tumours/paediatric-type low-grade glioma (LGG) not otherwise specified (GNT/PLGG NOS, n = 39). mdPCR found an alteration in 38/72 cases. Subsequent RNA sequencing revealed a fusion transcript involving BRAF, FGFR1/2/3 or neurotrophic tyrosine kinase receptor type 2 [NTRK2] in 9/25 cases. DNA methylation profiling found 12/46 cases with a calibrated score ≥0.9. Unsupervised hierarchical clustering revealed two clusters: Cluster 1 was enriched with cases classified as DNT at histology, belonging to the LGG-DNT methylation class (MC), with haematopoietic progenitor cell antigen (CD34) negativity and FGRF1 alterations; Cluster 2 was enriched with cases classified at histology as GG, belonging to the LGG-GG MC MC, with BRAF V600E mutation and CD34 positivity. The tumours reclassified as GNT/PLGG NOS were equally distributed across both clusters. Interestingly, all polymorphous low-grade neuroepithelial tumour of the young belonged to Cluster 2, whereas diffuse LGG mitogen-activated protein kinase (MAPK) pathway-altered were equally distributed among the two clusters. This led us to build an algorithm to classify LEATs with a prominent oligodendroglioma-like component. CONCLUSIONS Integrated histomolecular diagnosis of LEATs with a prominent oligodendroglioma-like component remains challenging. Because these tumours can be split into two major clusters of biological significance, the clinicopathological relevance of the four types recognised by the WHO CNS5 within this spectrum of tumours is questionable.
Collapse
Affiliation(s)
- Alice Métais
- AP-HM, CHU Timone, Service d'Anatomie Pathologique et de Neuropathologie, Marseille, France.,Service d'Anatomie et Cytologie Pathologiques, CHU Pontchaillou, Rennes, France
| | - Romain Appay
- AP-HM, CHU Timone, Service d'Anatomie Pathologique et de Neuropathologie, Marseille, France.,Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| | - Mélanie Pagès
- GHU-Paris Sainte-Anne Hospital, Paris University, Paris, France.,Department of Genetics, Institut Curie, Paris, France.,SIREDO Paediatric Cancer Center, Institut Curie, Paris, France.,INSERM U830, Laboratory of Translational Research in Paediatric Oncology, Institut Curie, Paris, France
| | - Catherine Gallardo
- AP-HM, CHU Timone, Service d'Anatomie Pathologique et de Neuropathologie, Marseille, France
| | - Karen Silva
- Groupe Hospitalier Est, Département de Neuropathologie, Hospices Civils de Lyon, Bron, France
| | - Aurore Siegfried
- Department of Pathology, Toulouse University Hospital, Toulouse, France.,INSERM U1037, Cancer Research Center of Toulouse (CRCT), Toulouse, France.,Université Paul Sabatier, Toulouse III, Toulouse, France
| | - Romain Perbet
- Univ. Lille, Inserm, CHU Lille, Alzheimer and Tauopathies, Lille Neuroscience & Cognition, UMR-S1172, Lille, France
| | - Claude-Alain Maurage
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, Lille, France
| | - Didier Scavarda
- Aix-Marseille Univ, AP-HM, Institut de Neurosciences des Systèmes, CHU Timone, Service de Neurochirurgie infantile, Marseille, France
| | - Frédéric Fina
- AP-HM, CHU Timone, Service d'Anatomie Pathologique et de Neuropathologie, Marseille, France.,ID Solutions, Research and Development, Grabels, France
| | - Emmanuelle Uro-Coste
- Department of Pathology, Toulouse University Hospital, Toulouse, France.,INSERM U1037, Cancer Research Center of Toulouse (CRCT), Toulouse, France.,Université Paul Sabatier, Toulouse III, Toulouse, France
| | - Laurent Riffaud
- Department of Pediatric Neurosurgery, Rennes University Hospital, Rennes, France.,INSERM MediCIS, unit U1099 LTSI, Rennes 1 University, Rennes, France
| | - Carole Colin
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| | - Dominique Figarella-Branger
- AP-HM, CHU Timone, Service d'Anatomie Pathologique et de Neuropathologie, Marseille, France.,Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| | | |
Collapse
|
17
|
Forest F, Dal Col P, Laville D, Court A, Rillardon M, Ramirez C, Rivoirard R, Stephan JL, Vassal F, Péoc'h M. Cyclin D1 expression in ganglioglioma, pleomorphic xanthoastrocytoma and pilocytic astrocytoma. Exp Mol Pathol 2021; 121:104652. [PMID: 34022185 DOI: 10.1016/j.yexmp.2021.104652] [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: 03/12/2021] [Revised: 05/11/2021] [Accepted: 05/17/2021] [Indexed: 11/18/2022]
Abstract
Ganglioglioma, pleomorphic xanthoastrocytoma (PXA) and pilocytic astrocytoma are rare brain neoplasms with frequent activation of mitogen-activated protein (MAP) kinase pathway. A downstream marker of MAP-kinase pathway activation is cyclin D1. However, the expression of cyclin D1 has not been studied in the differential diagnosis between these brain tumors. The aim of this work is to compare the expression of cyclin D1 in ganglioglioma, PXA, pilocytic astrocytoma. We also compared cyclin D1 expression in giant cell glioblastoma and in IDH wild type glioblastoma. Our work shows that roughly half of gangliogliomas have ganglion cells stained by cyclin D1 while two third of PXA have pleormophic cells stained by cyclin D1 and 15% of giant cell glioblastoma have pleomorphic cells stained by cyclin D1 (p < 0.001). Cyclin D1 never stains normal neurons either in the adjacent cortex of circumscribed tumor, or in entrapped neurons in IDH wild type glioblastomas. The expression of cyclin D1 is correlated to the presence of BRAF V600E mutation in ganglioglioma and PXA (p = 0.002). To conclude, cyclin D1 positivity might be used to confirm the neoplastic nature of ganglion cells. Cyclin D1 is expressed in most cases of BRAF V600E mutated gangliogliomas but also in cases without BRAF mutations suggesting an activation of MAP-kinase pathway through another way. Cyclin D1 immunohistochemistry has currently no or little role in the differential diagnosis of pilocytic astrocytoma. Its role in the differential diagnosis between PXA and giant cell glioblastoma needs to be further investigated on external series.
Collapse
Affiliation(s)
- Fabien Forest
- University Hospital of Saint Etienne, North Hospital, Department of Pathology, Avenue Albert Raimond, 42055, Saint Etienne CEDEX 2, France; University Hospital of Saint Etienne, North Hospital, Department of Molecular Biology of Tumors, Avenue Albert Raimond, 42055, Saint Etienne CEDEX 2, France; Corneal Graft Biology, Engineering and Imaging Laboratory, BiiGC, EA2521, Federative Institute of Research in Sciences and Health Engineering, Faculty of Medicine, Jean Monnet University, Saint-Etienne, France.
| | - Pierre Dal Col
- University Hospital of Saint Etienne, North Hospital, Department of Pathology, Avenue Albert Raimond, 42055, Saint Etienne CEDEX 2, France
| | - David Laville
- University Hospital of Saint Etienne, North Hospital, Department of Pathology, Avenue Albert Raimond, 42055, Saint Etienne CEDEX 2, France
| | - Alice Court
- University Hospital of Saint Etienne, North Hospital, Department of Pathology, Avenue Albert Raimond, 42055, Saint Etienne CEDEX 2, France
| | - Maxime Rillardon
- University Hospital of Saint Etienne, North Hospital, Department of Pathology, Avenue Albert Raimond, 42055, Saint Etienne CEDEX 2, France
| | - Carole Ramirez
- University Hospital of Saint Etienne, North Hospital, Department of Neurology, Avenue Albert Raimond, 42055, Saint Etienne CEDEX 2, France; Lucien Neuwith Cancer Institute, Department of Medical Oncology, Avenue Albert Raimond, 108 bis Avenue Albert Raimond, 42270 Saint-Priest-en-Jarez, France
| | - Romain Rivoirard
- Lucien Neuwith Cancer Institute, Department of Medical Oncology, Avenue Albert Raimond, 108 bis Avenue Albert Raimond, 42270 Saint-Priest-en-Jarez, France
| | - Jean-Louis Stephan
- University Hospital of Saint Etienne, North Hospital, Department of Pediatric Oncology, Avenue Albert Raimond. 42055, Saint Etienne CEDEX 2, France
| | - François Vassal
- University Hospital of Saint Etienne, North Hospital, Department of Neurosurgery, Avenue Albert Raimond, 42055, Saint Etienne CEDEX 2, France
| | - Michel Péoc'h
- University Hospital of Saint Etienne, North Hospital, Department of Pathology, Avenue Albert Raimond, 42055, Saint Etienne CEDEX 2, France; Corneal Graft Biology, Engineering and Imaging Laboratory, BiiGC, EA2521, Federative Institute of Research in Sciences and Health Engineering, Faculty of Medicine, Jean Monnet University, Saint-Etienne, France
| |
Collapse
|
18
|
Esami citologici, istologici, immunoistochimici e genetici dei tumori del sistema nervoso centrale. Neurologia 2021. [DOI: 10.1016/s1634-7072(21)45000-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
19
|
Barets D, Appay R, Heinisch M, Battistella M, Bouvier C, Chotard G, Le Loarer F, Macagno N, Perbet R, Pissaloux D, Rousseau A, Tauziède-Espariat A, Varlet P, Vasiljevic A, Colin C, Fina F, Figarella-Branger D. Specific and Sensitive Diagnosis of BCOR-ITD in Various Cancers by Digital PCR. Front Oncol 2021; 11:645512. [PMID: 33718245 PMCID: PMC7948083 DOI: 10.3389/fonc.2021.645512] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 01/27/2021] [Indexed: 11/25/2022] Open
Abstract
BCOR is an epigenetic regulator altered by various mechanisms including BCOR-internal tandem duplication (BCOR-ITD) in a wide range of cancers. Six different BCOR-ITD in the 3’-part of the coding sequence of exon 15 have been reported ranging from 89 to 114 bp in length. BCOR-ITD is a common genetic alteration found in clear cell sarcoma of the kidney and primitive myxoid mesenchymal tumor of infancy (PMMTI) and it characterizes a new type of central nervous system tumor: “CNS tumor with BCOR-ITD”. It can also be detected in undifferentiated round cell sarcoma (URCS) and in high-grade endometrial stromal sarcoma (HGESS). Therefore, it is of utmost importance to search for this genetic alteration in these cancers with the most frequent technique being RNA-sequencing. Here, we developed a new droplet PCR assay (dPCR) to detect the six sequences characterizing BCOR-ITD. To achieve this goal, we used a single colored probe to detect both the duplicated region and the normal sequence that acts as a reference. We first generated seven synthetic DNA sequences: ITD0 (the normal sequence) and ITD1 to ITD6 (the duplicated sequences described in the literature) and then we set up the optima dPCR conditions. We validated our assay on 19 samples from a representative panel of human tumors (9 HGNET-BCOR, 5 URCS, 3 HGESS, and 2 PMMTI) in which BCOR-ITD status was known using at least one other method including RNA sequencing, RT-PCR or DNA-methylation profiling for CNS tumors. Our results showed that our technique was 100% sensitive and specific. DPCR detected BCOR-ITD in 13/19 of the cases; in the remaining 6 cases additional RNA-sequencing revealed BCOR gene fusions. To conclude, in the era of histomolecular classification of human tumors, our modified dPCR assay is of particular interest to detect BCOR-ITD since it is a robust and less expensive test that can be applied to a broad spectrum of cancers that share this alteration.
Collapse
Affiliation(s)
- Doriane Barets
- APHM, CHU Timone, Service d'Anatomie Pathologique et de Neuropathologie, Marseille, France
| | - Romain Appay
- APHM, CHU Timone, Service d'Anatomie Pathologique et de Neuropathologie, Marseille, France.,Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| | - Marie Heinisch
- APHM, CHU Timone, Service d'Anatomie Pathologique et de Neuropathologie, Marseille, France
| | - Maxime Battistella
- Department of Pathology, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Université de Paris, Inserm U976, Paris, France
| | - Corinne Bouvier
- APHM, CHU Timone, Service d'Anatomie Pathologique et de Neuropathologie, Marseille, France
| | - Guillaume Chotard
- Service de Pathologie, Groupe Hospitalier Pellegrin, CHU de Bordeaux, Bordeaux, France
| | | | - Nicolas Macagno
- APHM, CHU Timone, Service d'Anatomie Pathologique et de Neuropathologie, Marseille, France
| | - Romain Perbet
- Institute of Pathology, CHU Lille, Lille, France.,LilNCog, Lille Neuroscience and Cognition, Univ. Lille, Inserm, CHU Lille, U1172, Lille, France
| | - Daniel Pissaloux
- Department of Translational Research and Innovation, Léon Bérard Cancer Center, Lyon, France.,Claude Bernard University Lyon 1, INSERM 1052, CNRS 5286, Cancer Research Center of Lyon, Centre Léon Bérard, Lyon, France
| | - Audrey Rousseau
- Département de Pathologie Cellulaire et Tissulaire, CHU Angers, Angers, France
| | - Arnaud Tauziède-Espariat
- Department of Neuropathology, GHU Paris-Psychiatrie Et Neurosciences, Sainte-Anne Hospital, Paris, France
| | - Pascale Varlet
- Department of Neuropathology, GHU Paris-Psychiatrie Et Neurosciences, Sainte-Anne Hospital, Paris, France
| | - Alexandre Vasiljevic
- Centre de Pathologie Est, Groupement Hospitalier Est, Hospices Civils de Lyon, Bron, France
| | - Carole Colin
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| | - Frédéric Fina
- APHM, CHU Timone, Service d'Anatomie Pathologique et de Neuropathologie, Marseille, France.,ID Solutions, Research and Development, Grabels, France
| | - Dominique Figarella-Branger
- APHM, CHU Timone, Service d'Anatomie Pathologique et de Neuropathologie, Marseille, France.,Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| |
Collapse
|
20
|
Bobach IS, Stougaard M. SNP-based detection of allelic imbalance: A novel approach for identifying KIAA1549-BRAF fusion in pilocytic astrocytoma using DNA sequencing. Exp Mol Pathol 2021; 120:104621. [PMID: 33626378 DOI: 10.1016/j.yexmp.2021.104621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/09/2021] [Accepted: 02/16/2021] [Indexed: 11/19/2022]
Abstract
Pilocytic astrocytoma (PA) is the most common glioma subtype found in children, and it is a non-malignant tumor type. The majority of PAs is caused by an approximately 2 Mb tandem duplication within 7q34 which creates an in-frame KIAA1549-BRAF fusion gene. The kinase domain of BRAF is fused to the N-terminal of KIAA1549, whereby BRAF is constitutively activated. We here present a novel approach for identifying KIAA1549-BRAF fusion based on single nucleotide polymorphism (SNP) analysis and next generation sequencing (NGS). Highly polymorphic SNPs in the duplicated area and in adjacent areas were selected and a custom targeted amplicon based NGS panel was designed. The panel was tested on DNA extracted from formalin fixed and paraffin embedded tissue from a retrospective cohort, consisting of biopsies from patients with PA, anaplastic astrocytoma, oligodendroglioma and glioblastoma as well as two non-tumor biopsies. The panel could distinguish chromosome 7 gain from BRAF fusion and correctly identified 8/9 PA samples with KIAA1549-BRAF fusion confirmed by RNA sequencing. The one biopsy where no fusion was detected was fresh frozen and from the RNA sequencing expected to have very low tumor content. No allelic imbalance was detected in either oligodendroglioma or in the non-tumor biopsies.
Collapse
Affiliation(s)
- Ida Schwartz Bobach
- Department of Clinical Medicine, Aarhus University, Denmark; Department of Pathology, Aarhus University Hospital, Denmark
| | - Magnus Stougaard
- Department of Clinical Medicine, Aarhus University, Denmark; Department of Pathology, Aarhus University Hospital, Denmark.
| |
Collapse
|
21
|
Tan JY, Wijesinghe IVS, Alfarizal Kamarudin MN, Parhar I. Paediatric Gliomas: BRAF and Histone H3 as Biomarkers, Therapy and Perspective of Liquid Biopsies. Cancers (Basel) 2021; 13:cancers13040607. [PMID: 33557011 PMCID: PMC7913734 DOI: 10.3390/cancers13040607] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 01/10/2023] Open
Abstract
Simple Summary Gliomas are major causes of worldwide cancer-associated deaths in children. Generally, paediatric gliomas can be classified into low-grade and high-grade gliomas. They differ significantly from adult gliomas in terms of prevalence, molecular alterations, molecular mechanisms and predominant histological types. The aims of this review article are: (i) to discuss the current updates of biomarkers in paediatric low-grade and high-grade gliomas including their diagnostic and prognostic values, and (ii) to discuss potential targeted therapies in treating paediatric low-grade and high-grade gliomas. Our findings revealed that liquid biopsy is less invasive than tissue biopsy in obtaining the samples for biomarker detections in children. In addition, future clinical trials should consider blood-brain barrier (BBB) penetration of therapeutic drugs in paediatric population. Abstract Paediatric gliomas categorised as low- or high-grade vary markedly from their adult counterparts, and denoted as the second most prevalent childhood cancers after leukaemia. As compared to adult gliomas, the studies of diagnostic and prognostic biomarkers, as well as the development of therapy in paediatric gliomas, are still in their infancy. A body of evidence demonstrates that B-Raf Proto-Oncogene or V-Raf Murine Sarcoma Viral Oncogene Homolog B (BRAF) and histone H3 mutations are valuable biomarkers for paediatric low-grade gliomas (pLGGs) and high-grade gliomas (pHGGs). Various diagnostic methods involving fluorescence in situ hybridisation, whole-genomic sequencing, PCR, next-generation sequencing and NanoString are currently used for detecting BRAF and histone H3 mutations. Additionally, liquid biopsies are gaining popularity as an alternative to tumour materials in detecting these biomarkers, but still, they cannot fully replace solid biopsies due to several limitations. Although histone H3 mutations are reliable prognosis biomarkers in pHGGs, children with these mutations have a dismal prognosis. Conversely, the role of BRAF alterations as prognostic biomarkers in pLGGs is still in doubt due to contradictory findings. The BRAF V600E mutation is seen in the majority of pLGGs (as seen in pleomorphic xanthoastrocytoma and gangliomas). By contrast, the H3K27M mutation is found in the majority of paediatric diffuse intrinsic pontine glioma and other midline gliomas in pHGGs. pLGG patients with a BRAF V600E mutation often have a lower progression-free survival rate in comparison to wild-type pLGGs when treated with conventional therapies. BRAF inhibitors (Dabrafenib and Vemurafenib), however, show higher overall survival and tumour response in BRAF V600E mutated pLGGs than conventional therapies in some studies. To date, targeted therapy and precision medicine are promising avenues for paediatric gliomas with BRAF V600E and diffuse intrinsic pontine glioma with the H3K27M mutations. Given these shortcomings in the current treatments of paediatric gliomas, there is a dire need for novel therapies that yield a better therapeutic response. The present review discusses the diagnostic tools and the perspective of liquid biopsies in the detection of BRAF V600E and H3K27M mutations. An in-depth understanding of these biomarkers and the therapeutics associated with the respective challenges will bridge the gap between paediatric glioma patients and the development of effective therapies.
Collapse
Affiliation(s)
| | | | | | - Ishwar Parhar
- Correspondence: ; Tel.: +603-5514-6304; Fax: +603-5515-6341
| |
Collapse
|
22
|
Appay R, Fina F, Barets D, Gallardo C, Nanni-Metellus I, Scavarda D, Henaff D, Vincent J, Grewis L, Pourquier P, Colin C, Figarella-Branger D. Multiplexed Droplet Digital PCR Assays for the Simultaneous Screening of Major Genetic Alterations in Tumors of the Central Nervous System. Front Oncol 2020; 10:579762. [PMID: 33282733 PMCID: PMC7689380 DOI: 10.3389/fonc.2020.579762] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 10/15/2020] [Indexed: 01/18/2023] Open
Abstract
The increased integration of molecular alterations to define tumor type or grade in central nervous system (CNS) tumor classification brings new challenges for the pathologist to make the best use of a precious limited tissue specimen for molecular studies. Within the different methods available to identify gene alterations, the droplet digital PCR (dPCR) constitutes a rapid, cost-effective, and very sensitive tool. In this study, we describe the development and validation of five multiplexed dPCR assays to detect major CNS biomarkers by using only small amounts of DNA extracted from formalin-fixed paraffin-embedded specimens. When compared to HRM-sequencing, NGS-sequencing, RNA-sequencing, or simplex digital PCR assays used as “gold standard” methods, these multiplexed dPCR assays displayed 100% specificity and sensitivity for the simultaneous detection of: 1/BRAF V600E mutation and KIAA1549:BRAF fusion; 2/FGFR1 N546K and K656E mutations and FGFR1 duplication; 3/H3F3A K27M and G34R/V mutations; 4/IDH1 R132X and IDH2 R172X mutations; and 5/TERT promoter mutations C228T and C250T. In light of the increased integration of molecular alteration, we believe that such strategies might help laboratories to optimize their screening strategies for routine diagnosis of pediatric and adult CNS tumors.
Collapse
Affiliation(s)
- Romain Appay
- APHM, CHU Timone, Service d'Anatomie Pathologique et de Neuropathologie, Marseille, France.,Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| | - Frederic Fina
- APHM, CHU Timone, Service d'Anatomie Pathologique et de Neuropathologie, Marseille, France.,ID Solutions, Research and Development, Grabels, France
| | - Doriane Barets
- APHM, CHU Timone, Service d'Anatomie Pathologique et de Neuropathologie, Marseille, France
| | - Catherine Gallardo
- APHM, CHU Timone, Service d'Anatomie Pathologique et de Neuropathologie, Marseille, France
| | - Isabelle Nanni-Metellus
- APHM, CHU Nord, Service de Transfert d'Oncologie Biologique, Laboratoire de Biologie Médicale, Marseille, France
| | - Didier Scavarda
- APHM, CHU Timone, Service de Neurochirurgie pédiatrique, Marseille, France
| | - Daniel Henaff
- ID Solutions, Research and Development, Grabels, France
| | | | - Lise Grewis
- ID Solutions, Research and Development, Grabels, France
| | | | - Carole Colin
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| | - Dominique Figarella-Branger
- APHM, CHU Timone, Service d'Anatomie Pathologique et de Neuropathologie, Marseille, France.,Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| |
Collapse
|
23
|
Diffuse leptomeningeal glioneuronal tumor: a double misnomer? A report of two cases. Acta Neuropathol Commun 2020; 8:95. [PMID: 32605662 PMCID: PMC7325675 DOI: 10.1186/s40478-020-00978-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 06/22/2020] [Indexed: 11/25/2022] Open
Abstract
Diffuse leptomeningeal glioneuronal tumor (DLGNT) was introduced, for the first time, as a provisional entity in the 2016 WHO classification of central nervous system tumors. DLGNT mainly occur in children and characterized by a widespread leptomeningeal growth occasionally associated with intraspinal tumor nodules, an oligodendroglial-like cytology, glioneuronal differentiation and MAP-Kinase activation associated with either solitary 1p deletion or 1p/19q codeletion in the absence of IDH mutation. We report here two unexpected DLGNTs adult cases, characterized by a unique supratentorial circumscribed intraparenchymal tumor without leptomeningeal involvement in spite of long follow-up. In both cases, the diagnosis of DLGNT was made after DNA-methylation profiling which demonstrated that one case belonged to the DLGNT class whereas the other remained not classifiable but showed on CNV the characteristic genetic findings recorded in DLGNT. Both cases harbored 1p/19q codeletion associated with KIAA1549:BRAF fusion in one case and with BRAF V600E and PIK3CA E545A mutations, in the other. Our study enlarges the clinical and molecular spectrum of DLGNTs, and points out that the terminology of DLGNTs is not fully appropriate since some cases could have neither diffuse growth nor leptomeningeal dissemination. This suggests that DLGNTs encompass a wide spectrum of tumors that has yet to be fully clarified.
Collapse
|
24
|
Chai RC, Zhang YW, Liu YQ, Chang YZ, Pang B, Jiang T, Jia WQ, Wang YZ. The molecular characteristics of spinal cord gliomas with or without H3 K27M mutation. Acta Neuropathol Commun 2020; 8:40. [PMID: 32228694 PMCID: PMC7106747 DOI: 10.1186/s40478-020-00913-w] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 03/09/2020] [Indexed: 12/11/2022] Open
Abstract
Due to the rare incidence of spinal cord astrocytomas, their molecular features remain unclear. Here, we characterized the landscapes of mutations in H3 K27M, isocitrate dehydrogenase 1 (IDH1) R132H, BRAF V600E, and the TERT promoter in 83 diffuse spinal cord astrocytic tumors. Among these samples, thirty-five patients had the H3 K27M mutation; this mutant could be observed in histological grade II (40%), III (40%), and IV (20%) astrocytomas. IDH1 mutations were absent in 58 of 58 cases tested. The BRAF V600E mutation (7/57) was only observed in H3-wildtype astrocytomas, and was associated with a better prognosis in all histological grade II/III astrocytomas. TERT promoter mutations were observed in both H3 K27M-mutant (4/25) and -wildtype (9/33) astrocytomas, and were associated with a poor prognosis in H3-wildtype histological grade II/III astrocytomas. In the 2016 WHO classification of CNS tumors, H3 K27M-mutant diffuse midline gliomas, including spinal cord astrocytomas, are categorized as WHO grade IV. Here, we noticed that the median overall survival of histological grade II/III H3 K27M-mutant cases (n = 28) was significantly longer than that of either the total histological grade IV cases (n = 12) or the H3 K27M-mutant histological grade IV cases (n = 7). We also directly compared H3 K27M-mutant astrocytomas to H3-wildtype astrocytomas of the same histological grade. In histological grade II astrocytomas, compared to H3-wildtype cases (n = 37), H3 K27M-mutant patients (n = 14) had showed a significantly higher Ki-67-positive rate and poorer survival rate. However, no significant differences in these parameters were observed in histological grade III and IV astrocytoma patients. In conclusion, these findings indicate that spinal cord astrocytomas are considerably different from hemispheric and brainstem astrocytomas in terms of their molecular profiles, and that the histological grade cannot be ignored when assessing the prognosis of H3 K27M-mutant spinal cord astrocytomas.
Collapse
|
25
|
Ryall S, Tabori U, Hawkins C. Pediatric low-grade glioma in the era of molecular diagnostics. Acta Neuropathol Commun 2020; 8:30. [PMID: 32164789 PMCID: PMC7066826 DOI: 10.1186/s40478-020-00902-z] [Citation(s) in RCA: 222] [Impact Index Per Article: 44.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 02/21/2020] [Indexed: 12/17/2022] Open
Abstract
Low grade gliomas are the most frequent brain tumors in children and encompass a spectrum of histologic entities which are currently assigned World Health Organisation grades I and II. They differ substantially from their adult counterparts in both their underlying genetic alterations and in the infrequency with which they transform to higher grade tumors. Nonetheless, children with low grade glioma are a therapeutic challenge due to the heterogeneity in their clinical behavior – in particular, those with incomplete surgical resection often suffer repeat progressions with resultant morbidity and, in some cases, mortality. The identification of up-regulation of the RAS–mitogen-activated protein kinase (RAS/MAPK) pathway as a near universal feature of these tumors has led to the development of targeted therapeutics aimed at improving responses while mitigating patient morbidity. Here, we review how molecular information can help to further define the entities which fall under the umbrella of pediatric-type low-grade glioma. In doing so we discuss the specific molecular drivers of pediatric low grade glioma and how to effectively test for them, review the newest therapeutic agents and their utility in treating this disease, and propose a risk-based stratification system that considers both clinical and molecular parameters to aid clinicians in making treatment decisions.
Collapse
|
26
|
The histomolecular criteria established for adult anaplastic pilocytic astrocytoma are not applicable to the pediatric population. Acta Neuropathol 2020; 139:287-303. [PMID: 31677015 PMCID: PMC6989446 DOI: 10.1007/s00401-019-02088-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 12/18/2022]
Abstract
Pilocytic astrocytoma (PA) is the most common pediatric glioma, arising from a single driver MAPK pathway alteration. Classified as a grade I tumor according to the 2016 WHO classification, prognosis is excellent with a 10-year survival rate > 95% after surgery. However, rare cases present with anaplastic features, including an unexpected high mitotic/proliferative index, thus posing a diagnostic and therapeutic challenge. Based on small histomolecular series and case reports, such tumors arising at the time of diagnosis or recurrence have been designated by many names including pilocytic astrocytoma with anaplastic features (PAAF). Recent DNA methylation-profiling studies performed mainly on adult cases have revealed that PAAF exhibit a specific methylation signature, thus constituting a distinct methylation class from typical PA [methylation class anaplastic astrocytoma with piloid features-(MC-AAP)]. However, the diagnostic and prognostic significance of MC-AAP remains to be determined in children. We performed an integrative work on the largest pediatric cohort of PAAF, defined according to strict criteria: morphology compatible with the diagnosis of PA, with or without necrosis, ≥ 4 mitoses for 2.3 mm2, and MAPK pathway alteration. We subjected 31 tumors to clinical, imaging, morphological and molecular analyses, including DNA methylation profiling. We identified only one tumor belonging to the MC-AAP (3%), the others exhibiting a methylation profile typical for PA (77%), IDH-wild-type glioblastoma (7%), and diffuse leptomeningeal glioneuronal tumor (3%), while three cases (10%) did not match to a known DNA methylation class. No significant outcome differences were observed between PAAF with necrosis versus no necrosis (p = 0.07), or with 4-6 mitoses versus 7 or more mitoses (p = 0.857). Our findings argue that the diagnostic histomolecular criteria established for anaplasia in adult PA are not of diagnostic or prognostic value in a pediatric setting. Further extensive and comprehensive integrative studies are necessary to accurately define this exceptional entity in children.
Collapse
|
27
|
Kurani H, Gurav M, Shetty O, Chinnaswamy G, Moiyadi A, Gupta T, Jalali R, Epari S. Pilocytic astrocytomas: BRAFV600E and BRAF fusion expression patterns in pediatric and adult age groups. Childs Nerv Syst 2019; 35:1525-1536. [PMID: 31321520 DOI: 10.1007/s00381-019-04282-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 06/26/2019] [Indexed: 12/16/2022]
Abstract
PURPOSE Pilocytic astrocytomas (PCAs) are characterized by two dominant molecular alterations of the BRAF gene, i.e., BRAFV600E mutation and KIAA1549-BRAF fusions which show a differential pattern of frequency across different age-groups. METHODS Formalin-fixed paraffin-embedded tissues of 358 (pediatric 276 and adult 82) consecutive PCAs were evaluated for BRAFV600E mutation by Sanger sequencing and KIAA1549:BRAF fusion transcripts (KIAA1549:BRAF 16-9, KIAA1549:BRAF 15-9, and KIAA1549:BRAF 16-11) by reverse transcriptase polymerase chain reaction, which were correlated with different clinicopathological features. RESULTS BRAFV600E mutation was detected in 8.9% pediatric and 9.75% adult PCAs, whereas 41.1% and 25.7% of pediatric and adult cases showed KIAA1549-BRAF fusions respectively. BRAFV600E did not show any statistically significant correlation with any of the clinical parameters (age, location, and gender). KIAA1549:BRAF fusions showed a significant statistical association with the pediatric age group and cerebellar location. KIAA1549-BRAF 16-9 was the commonest variant and was predominantly associated with cerebellar location than non-cerebellar whereas fusion variant 15-9 negatively correlated with cerebellar locations. CONCLUSIONS The present study showed overall frequency of 53.5% and 37.3% BRAF alterations in pediatric and adult PCA cases respectively. BRAF fusion in PCA cases showed a different distribution pattern across age groups and locations; while no such differential pattern was observed for BRAFV600E.
Collapse
Affiliation(s)
- Hetakshi Kurani
- Division of Molecular Pathology, Department of Pathology, Tata Memorial Centre, Homi Baba National Institute, Mumbai, 400012, India
| | - Mamta Gurav
- Division of Molecular Pathology, Department of Pathology, Tata Memorial Centre, Homi Baba National Institute, Mumbai, 400012, India
| | - Omshree Shetty
- Division of Molecular Pathology, Department of Pathology, Tata Memorial Centre, Homi Baba National Institute, Mumbai, 400012, India
| | - Girish Chinnaswamy
- Department of Pediatric Oncology, Tata Memorial Centre, Homi Baba National Institute, Mumbai, 400012, India
| | - Aliasagar Moiyadi
- Division of Neurosurgery, Department of Surgical Oncology, Tata Memorial Hospital and ACTREC, Tata Memorial Centre, Homi Baba National Institute, Mumbai, 400012, India
| | - Tejpal Gupta
- Department of Radiation Oncology, Tata Memorial Hospital and ACTREC, Tata Memorial Centre, Homi Baba National Institute, Mumbai, 400012, India
| | - Rakesh Jalali
- Department of Radiation Oncology, Tata Memorial Hospital and ACTREC, Tata Memorial Centre, Homi Baba National Institute, Mumbai, 400012, India
| | - Sridhar Epari
- Division of Molecular Pathology, Department of Pathology, Tata Memorial Centre, Homi Baba National Institute, Mumbai, 400012, India.
- Department of Pathology (& Division of Molecular Pathology), Tata Memorial Hospital and ACTREC, Tata Memorial Centre, Homi Baba National Institute, Mumbai, 400012, India.
| |
Collapse
|
28
|
Bret D, Chappuis V, Poncet D, Ducray F, Silva K, Mion F, Vasiljevic A, Ferraro-Peyret C, Mottolese C, Leblond P, Gabut M, Frappaz D, Streichenberger N, Meyronet D, Bringuier PP, Barritault M. A Multiplex Quantitative Reverse Transcription Polymerase Chain Reaction Assay for the Detection of KIAA1549-BRAF Fusion Transcripts in Formalin-Fixed Paraffin-Embedded Pilocytic Astrocytomas. Mol Diagn Ther 2019; 23:537-545. [PMID: 31087282 DOI: 10.1007/s40291-019-00403-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
BACKGROUND AND OBJECTIVE Genomic duplications and fusion involving BRAF and KIAA1549 that create fusion proteins with constitutive B-RAF kinase activity are a hallmark of pilocytic astrocytomas (PAs). The detection of KIAA1549-BRAF fusion transcripts is of paramount importance to classify these tumors and to identify patients who could benefit from BRAF inhibitors. In a clinical setting, the available material for molecular analysis from these pediatric tumors is often limited to formalin-fixed paraffin-embedded (FFPE) tissue. The aim of the present study was to develop a new method to detect the three most frequent KIAA1549-BRAF fusion transcripts, 15-9, 16-11, and 16-9, where numbers refer to the exons fused together, using a FFPE-compatible multiplex quantitative reverse transcription polymerase chain reaction (qRT-PCR). METHODS We compared performance of the assay to a reference singleplex method on a collection of 46 FFPE PAs. RESULTS The results showed that both methods are comparable. The multiplex method had an overall 97% sensitivity and 100% specificity compared to the singleplex method, and agreement between the two techniques was almost perfect (Cohen's kappa: 0.97). There was no evidence of a significant difference between the qRT-PCR efficiencies of the multiplex technique and of the singleplex assay for all fusion transcripts and for GAPDH, the latter used as a reference gene. The multiplex method consumed four times less complementary DNA (cDNA), cost less, and required half the hands-on technical time. CONCLUSION The results show that it could be beneficial to implement the multiplex method in a clinical setting, where samples presenting low quantity of degraded RNA are not unusual.
Collapse
Affiliation(s)
- David Bret
- Service de Cytologie et d'Anatomie Pathologique, Département de Biopathologie Moléculaire et de Département de Neuropathologie, Hospices Civils de Lyon, Groupement Hospitalier Est, Lyon, France.,University of Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Valentin Chappuis
- Service de Cytologie et d'Anatomie Pathologique, Département de Biopathologie Moléculaire et de Département de Neuropathologie, Hospices Civils de Lyon, Groupement Hospitalier Est, Lyon, France.,University of Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Delphine Poncet
- Service de Cytologie et d'Anatomie Pathologique, Département de Biopathologie Moléculaire et de Département de Neuropathologie, Hospices Civils de Lyon, Groupement Hospitalier Est, Lyon, France.,University of Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - François Ducray
- University of Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Service de Neuro-oncologie, Hospices Civils de Lyon, Hôpital Neurologique, Lyon, France.,Transcriptome Diversity in Stem Cells Laboratory, Cancer Cell Plasticity Department, Cancer Research Centre of Lyon, INSERM U1052, CNRS, UMR 5286, Lyon, France
| | - Karen Silva
- Service de Cytologie et d'Anatomie Pathologique, Département de Biopathologie Moléculaire et de Département de Neuropathologie, Hospices Civils de Lyon, Groupement Hospitalier Est, Lyon, France
| | - Fabrice Mion
- Service de Cytologie et d'Anatomie Pathologique, Département de Biopathologie Moléculaire et de Département de Neuropathologie, Hospices Civils de Lyon, Groupement Hospitalier Est, Lyon, France
| | - Alexandre Vasiljevic
- Service de Cytologie et d'Anatomie Pathologique, Département de Biopathologie Moléculaire et de Département de Neuropathologie, Hospices Civils de Lyon, Groupement Hospitalier Est, Lyon, France.,University of Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Carole Ferraro-Peyret
- Service de Cytologie et d'Anatomie Pathologique, Département de Biopathologie Moléculaire et de Département de Neuropathologie, Hospices Civils de Lyon, Groupement Hospitalier Est, Lyon, France.,University of Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Carmine Mottolese
- Service de Neurochirurgie Lyon, Hospices Civils de Lyon, Groupement Hospitalier Est, Lyon, France
| | - Pierre Leblond
- Service d'Oncologie, Institut d'Hématologie et d'Oncologie Pédiatrique, Lyon, France
| | - Mathieu Gabut
- Transcriptome Diversity in Stem Cells Laboratory, Cancer Cell Plasticity Department, Cancer Research Centre of Lyon, INSERM U1052, CNRS, UMR 5286, Lyon, France
| | - Didier Frappaz
- Service d'Oncologie, Institut d'Hématologie et d'Oncologie Pédiatrique, Lyon, France
| | - Nathalie Streichenberger
- Service de Cytologie et d'Anatomie Pathologique, Département de Biopathologie Moléculaire et de Département de Neuropathologie, Hospices Civils de Lyon, Groupement Hospitalier Est, Lyon, France.,University of Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - David Meyronet
- Service de Cytologie et d'Anatomie Pathologique, Département de Biopathologie Moléculaire et de Département de Neuropathologie, Hospices Civils de Lyon, Groupement Hospitalier Est, Lyon, France.,University of Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Transcriptome Diversity in Stem Cells Laboratory, Cancer Cell Plasticity Department, Cancer Research Centre of Lyon, INSERM U1052, CNRS, UMR 5286, Lyon, France
| | - Pierre-Paul Bringuier
- Service de Cytologie et d'Anatomie Pathologique, Département de Biopathologie Moléculaire et de Département de Neuropathologie, Hospices Civils de Lyon, Groupement Hospitalier Est, Lyon, France.,University of Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Marc Barritault
- Service de Cytologie et d'Anatomie Pathologique, Département de Biopathologie Moléculaire et de Département de Neuropathologie, Hospices Civils de Lyon, Groupement Hospitalier Est, Lyon, France. .,University of Lyon, Université Claude Bernard Lyon 1, Lyon, France. .,Transcriptome Diversity in Stem Cells Laboratory, Cancer Cell Plasticity Department, Cancer Research Centre of Lyon, INSERM U1052, CNRS, UMR 5286, Lyon, France.
| |
Collapse
|
29
|
Multiplex ligation-dependent probe amplification analysis is useful for detecting a copy number gain of the FGFR1 tyrosine kinase domain in dysembryoplastic neuroepithelial tumors. J Neurooncol 2019; 143:27-33. [DOI: 10.1007/s11060-019-03138-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 02/26/2019] [Indexed: 01/01/2023]
|