119 results on '"Shago M"'
Search Results
2. Clinical phenotypes and prognostic features of embryonal tumours with multi-layered rosettes: a Rare Brain Tumor Registry study.
- Author
-
Khan, S, Solano-Paez, P, Suwal, T, Lu, M, Al-Karmi, S, Ho, B, Mumal, I, Shago, M, Hoffman, LM, Dodgshun, A, Nobusawa, S, Tabori, U, Bartels, U, Ziegler, DS, Hansford, JR, Ramaswamy, V, Hawkins, C, Dufour, C, André, N, Bouffet, E, Huang, A, Rare Brain Tumor Registry, Khan, S, Solano-Paez, P, Suwal, T, Lu, M, Al-Karmi, S, Ho, B, Mumal, I, Shago, M, Hoffman, LM, Dodgshun, A, Nobusawa, S, Tabori, U, Bartels, U, Ziegler, DS, Hansford, JR, Ramaswamy, V, Hawkins, C, Dufour, C, André, N, Bouffet, E, Huang, A, and Rare Brain Tumor Registry
- Abstract
BACKGROUND: Embryonal tumours with multi-layered rosettes (ETMRs) are a newly recognised, rare paediatric brain tumour with alterations of the C19MC microRNA locus. Due to varied diagnostic practices and scarce clinical data, disease features and determinants of outcomes for these tumours are poorly defined. We did an integrated clinicopathological and molecular analysis of primary ETMRs to define clinical phenotypes, and to identify prognostic factors of survival and key treatment modalities for this orphan disease. METHODS: Paediatric patients with primary ETMRs and tissue available for analyses were identified from the Rare Brain Tumor Consortium global registry. The institutional histopathological diagnoses were centrally re-reviewed as per the current WHO CNS tumour guidelines, using histopathological and molecular assays. Only patients with complete clinical, treatment, and survival data on Nov 30, 2019, were included in clinicopathological analyses. Among patients who received primary multi-modal curative regimens, event-free survival and overall survival were determined using Cox proportional hazard and log-rank analyses. Univariate and multivariable Cox proportional hazard regression was used to estimate hazard ratios (HRs) with 95% CIs for clinical, molecular, or treatment-related prognostic factors. FINDINGS: 159 patients had a confirmed molecular diagnosis of primary ETMRs (median age at diagnosis 26 months, IQR 18-36) and were included in our clinicopathological analysis. ETMRs were predominantly non-metastatic (94 [73%] of 128 patients), arising from multiple sites; 84 (55%) of 154 were cerebral tumours and 70 (45%) of 154 arose at sites characteristic of other brain tumours. Hallmark C19MC alterations were seen in 144 (91%) of 159 patients; 15 (9%) were ETMR not otherwise specified. In patients treated with curative intent, event-free survival was 57% (95% CI 47-68) at 6 months and 31% (21-42) at 2 years; overall survival was 29% (20-38) at 2 years an
- Published
- 2021
3. Functional differences between myeloid leukemia-initiating and transient leukemia cells in Down's syndrome
- Author
-
Chen, J, Li, Y, Doedens, M, Wang, P, Shago, M, Dick, J, and Hitzler, J K
- Published
- 2010
- Full Text
- View/download PDF
4. Antimicrobial Efficacy of Methanolic and Aqueous Extracts of Partially Purified Protein from Young and Matured Root of Guiera senegalensis (Moshi Medicine)
- Author
-
Jiyil, M. K., primary, Shago, M. I., primary, Mafuyai, C. E., primary, Silas, M., primary, and Olorunyomi, O. A., primary
- Published
- 2020
- Full Text
- View/download PDF
5. High incidence of CALM-AF10 fusion and the identification of a novel fusion transcript in acute megakaryoblastic leukemia in children without Down's syndrome
- Author
-
Abdelhaleem, M, Beimnet, K, Kirby-Allen, M, Naqvi, A, Hitzler, J, and Shago, M
- Published
- 2007
6. Alterations in ALK/ROS1/NTRK/MET drive a group of infantile hemispheric gliomas.
- Author
-
Guerreiro Stucklin, AS, Ryall, S, Fukuoka, K, Zapotocky, M, Lassaletta, A, Li, C, Bridge, T, Kim, B, Arnoldo, A, Kowalski, PE, Zhong, Y, Johnson, M, Ramani, AK, Siddaway, R, Nobre, LF, de Antonellis, P, Dunham, C, Cheng, S, Boué, DR, Finlay, JL, Coven, SL, de Prada, I, Perez-Somarriba, M, Faria, CC, Grotzer, MA, Rushing, E, Sumerauer, D, Zamecnik, J, Krskova, L, Garcia Ariza, M, Cruz, O, Morales La Madrid, A, Solano, P, Terashima, K, Nakano, Y, Ichimura, K, Nagane, M, Sakamoto, H, Gil-da-Costa, MJ, Silva, R, Johnston, DL, Michaud, J, Wilson, B, van Landeghem, FKH, Oviedo, A, McNeely, PD, Crooks, B, Fried, I, Zhukova, N, Hansford, JR, Nageswararao, A, Garzia, L, Shago, M, Brudno, M, Irwin, MS, Bartels, U, Ramaswamy, V, Bouffet, E, Taylor, MD, Tabori, U, Hawkins, C, Guerreiro Stucklin, AS, Ryall, S, Fukuoka, K, Zapotocky, M, Lassaletta, A, Li, C, Bridge, T, Kim, B, Arnoldo, A, Kowalski, PE, Zhong, Y, Johnson, M, Ramani, AK, Siddaway, R, Nobre, LF, de Antonellis, P, Dunham, C, Cheng, S, Boué, DR, Finlay, JL, Coven, SL, de Prada, I, Perez-Somarriba, M, Faria, CC, Grotzer, MA, Rushing, E, Sumerauer, D, Zamecnik, J, Krskova, L, Garcia Ariza, M, Cruz, O, Morales La Madrid, A, Solano, P, Terashima, K, Nakano, Y, Ichimura, K, Nagane, M, Sakamoto, H, Gil-da-Costa, MJ, Silva, R, Johnston, DL, Michaud, J, Wilson, B, van Landeghem, FKH, Oviedo, A, McNeely, PD, Crooks, B, Fried, I, Zhukova, N, Hansford, JR, Nageswararao, A, Garzia, L, Shago, M, Brudno, M, Irwin, MS, Bartels, U, Ramaswamy, V, Bouffet, E, Taylor, MD, Tabori, U, and Hawkins, C
- Abstract
Infant gliomas have paradoxical clinical behavior compared to those in children and adults: low-grade tumors have a higher mortality rate, while high-grade tumors have a better outcome. However, we have little understanding of their biology and therefore cannot explain this behavior nor what constitutes optimal clinical management. Here we report a comprehensive genetic analysis of an international cohort of clinically annotated infant gliomas, revealing 3 clinical subgroups. Group 1 tumors arise in the cerebral hemispheres and harbor alterations in the receptor tyrosine kinases ALK, ROS1, NTRK and MET. These are typically single-events and confer an intermediate outcome. Groups 2 and 3 gliomas harbor RAS/MAPK pathway mutations and arise in the hemispheres and midline, respectively. Group 2 tumors have excellent long-term survival, while group 3 tumors progress rapidly and do not respond well to chemoradiation. We conclude that infant gliomas comprise 3 subgroups, justifying the need for specialized therapeutic strategies.
- Published
- 2019
7. Antimicrobial Activity of Methanolic, Aqueous and Partially Purified Protein of Young and Matured Leaves of Guiera senegalensis (Moshi Medicine)
- Author
-
Jiyil, M. K., primary, Mafuyai, C. E., primary, Shago, M. I., primary, and Inuwa, H. M., primary
- Published
- 2019
- Full Text
- View/download PDF
8. Erratum: Functional differences between myeloid leukemia-initiating and transient leukemia cells in Down's syndrome
- Author
-
Chen, J, Li, Y, Doedens, M, Wang, P, Shago, M, Dick, J E, and Hitzler, J K
- Published
- 2010
- Full Text
- View/download PDF
9. Unusual X inactivation: an active ring(X)
- Author
-
Shago, M., Antinucci, D., Chakraborty, P., Sgro, M., Barozzino, T., Chitayat, D., and Teshima, I.
- Subjects
Human genetics -- Research ,Human chromosome abnormalities -- Research ,Genetic disorders -- Research ,Syndromes -- Genetic aspects ,Birth defects -- Genetic aspects ,Skeleton -- Abnormalities ,Biological sciences - Published
- 2001
10. Use of Molecular Diagnostic Techniques to Determinate the Epidemiology of Malaria Parasites in North Eastern Nigeria.
- Author
-
Lawan, M. M. and Shago, M. I.
- Subjects
PLASMODIUM ,EPIDEMIOLOGY ,MICROSCOPY ,BLOOD testing ,DRUG abuse - Abstract
The emergence of resistance to all antimalarial drugs in clinical use is now making it necessary to discover the markers responsible for the resistance. The principal aim of this research is the use of molecular diagnostic techniques to Determine the epidemiology of malaria parasites. Thirty blood samples were analyzed by microscopy and molecular techniques to monitor the relative efficiency in malaria diagnosis. Molecular analysis revealed 28 out of 30 samples as positive for malaria while Microscopic analysis revealed 27out of 30 samples as positive malaria parasite. The molecular analysis was particularly useful to unveil parasites presence in infections not detectable by blood smear analysis. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
11. LG-01 * BRAF MUTATION AND CDKN2A DELETION DEFINE A CLINICALLY DISTINCT SUBGROUP OF CHILDHOOD SECONDARY HIGH-GRADE GLIOMA
- Author
-
Mistry, M., primary, Zhukova, N., additional, Merico, D., additional, Rakopoulos, P., additional, Krishnatry, R., additional, Shago, M., additional, Stavropoulos, J., additional, Pole, J., additional, Ray, P., additional, Remke, M., additional, Buczkowicz, P., additional, Ramaswamy, V., additional, Shlien, A., additional, Rutka, J., additional, Dirks, P., additional, Taylor, M., additional, Malkin, D., additional, Bouffet, E., additional, Hawkins, C., additional, and Tabori, U., additional
- Published
- 2015
- Full Text
- View/download PDF
12. The impact of category, cytopathology and cytogenetics on development and progression of clonal and malignant myeloid transformation in inherited bone marrow failure syndromes
- Author
-
Cada, M., primary, Segbefia, C. I., additional, Klaassen, R., additional, Fernandez, C. V., additional, Yanofsky, R. A., additional, Wu, J., additional, Pastore, Y., additional, Silva, M., additional, Lipton, J. H., additional, Brossard, J., additional, Michon, B., additional, Abish, S., additional, Steele, M., additional, Sinha, R., additional, Belletrutti, M., additional, Breakey, V., additional, Jardine, L., additional, Goodyear, L., additional, Sung, L., additional, Shago, M., additional, Beyene, J., additional, Sharma, P., additional, Zlateska, B., additional, and Dror, Y., additional
- Published
- 2015
- Full Text
- View/download PDF
13. BI-21 * BRAF MUTATION AND CDKN2A DELETIONS DEFINE A CLINICALLY DISTINCT SUBGROUP OF CHILDHOOD SECONDARY HIGH GRADE GLIOMA
- Author
-
Mistry, M., primary, Zhukova, N., additional, Merico, D., additional, Rakopoulos, P., additional, Krishnatry, R., additional, Shago, M., additional, Stavropoulos, J., additional, Ray, P., additional, Mangerel, J., additional, Remke, M., additional, Buczkowicz, P., additional, Ramaswamy, V., additional, Rutka, J., additional, Dirks, P., additional, Taylor, M., additional, Bouffet, E., additional, Malkin, D., additional, Huang, A., additional, Hawkins, C., additional, and Tabori, U., additional
- Published
- 2014
- Full Text
- View/download PDF
14. PEDIATRICS CLINICAL RESEARCH
- Author
-
Antony, R., primary, Zagardo, M., additional, Gujrati, M., additional, Lin, J., additional, Antony, R., additional, Al-Rahawan, M., additional, Broniscer, A., additional, Bhardwaj, R., additional, Hampton, C., additional, Ozols, V., additional, Chakravadhanula, M., additional, Bouffet, E., additional, Hawkins, C., additional, Scheinemann, K., additional, Zelcer, S., additional, Johnston, D., additional, Lafay-Cousin, L., additional, Larouche, V., additional, Jabado, N., additional, Carret, A. S., additional, Hukin, J., additional, Eisenstat, D., additional, Pond, G., additional, Poskitt, K., additional, Wilson, B., additional, Bartels, U., additional, Tabori, U., additional, Dhall, G., additional, Haley, K., additional, Finlay, J., additional, Rushing, T., additional, Sposto, R., additional, Seeger, R., additional, Garvin, J., additional, Rupani, K., additional, Stark, E., additional, Anderson, R., additional, Feldstein, N., additional, Grill, J., additional, Hargrave, D., additional, Massimino, M., additional, Jaspan, T., additional, Varlet, P., additional, Jones, C., additional, Morgan, P., additional, Le Deley, M. C., additional, Azizi, A., additional, Canete, A., additional, Saran, F., additional, Bachir, J., additional, Bubuteishvili-Pacaud, L., additional, Rousseau, R., additional, Vassal, G., additional, Gupta, S., additional, Robinson, N., additional, Dhir, N., additional, Wong, K., additional, Zhou, S., additional, Kumabe, T., additional, Kawaguchi, T., additional, Saito, R., additional, Kanamori, M., additional, Yamashita, Y., additional, Sonoda, Y., additional, Tominaga, T., additional, Miyagawa, T., additional, Nwachukwu, C., additional, Youland, R., additional, Laack, N., additional, Filipek, I., additional, Drogosiewicz, M., additional, Polnik, M. P.-, additional, Swieszkowska, E., additional, Dembowska-Baginska, B., additional, Jurkiewicz, E., additional, Perek, D., additional, Grajkowska, W., additional, Roszkowski, M., additional, Sobol, G., additional, Musiol, K., additional, Wachowiak, J., additional, Kazmierczak, B., additional, Pogorzelski, J. P. -, additional, Mlynarski, W., additional, Szewczyk, B. Z.-, additional, Wysocki, M., additional, Niedzielska, E., additional, Kowalczyk, J., additional, Slusarz, H. W. -, additional, Balwierz, W., additional, Czepko, E. Z. -, additional, Szolkiewicz, A., additional, Perek-Polnik, M., additional, Lastowska, M., additional, Chojnacka, M., additional, Tarasinska, M., additional, Perreault, S., additional, Chao, K., additional, Ramaswamy, V., additional, Shih, D., additional, Remke, M., additional, Luu, B., additional, Schubert, S., additional, Fisher, P., additional, Partap, S., additional, Vogel, H., additional, Taylor, M., additional, Goumnerova, L., additional, Cho, Y.-J., additional, Robison, N., additional, Brown, R., additional, Cloughesy, T., additional, Davidson, T. B., additional, Krieger, M., additional, Berger, M., additional, Perry, A., additional, Gilles, F., additional, Finlay, J. L., additional, Khemani, J., additional, Britt, B., additional, Grimm, J., additional, Ruge, M. I., additional, Blau, T., additional, Hafkemeyer, V., additional, Hamisch, C., additional, Klinger, K., additional, Simon, T., additional, Sadighi, Z., additional, Ellezam, B., additional, Guindani, M., additional, Ater, J., additional, Shimizu, Y., additional, Arai, H., additional, Miyajima, M., additional, Shimoji, K., additional, Kondo, A., additional, Shinohara, E., additional, Perkins, S., additional, DeWees, T., additional, Slavc, I., additional, Chocholous, M., additional, Leiss, U., additional, Haberler, C., additional, Peyrl, A., additional, Azizi, A. A., additional, Dieckmann, K., additional, Woehrer, A., additional, Dorfer, C., additional, Czech, T., additional, Spence, T., additional, Picard, D., additional, Barszczyk, M., additional, Kim, S.-K., additional, Ra, Y.-S., additional, Fangusaro, J., additional, Toledano, H., additional, Nakamura, H., additional, Fan, X., additional, Muraszko, K. M., additional, Ng, H.-K., additional, Halliday, W., additional, Shago, M., additional, Hawkins, C. E., additional, Huang, A., additional, Suzuki, M., additional, van Zanten, S. V., additional, Jansen, M., additional, van Vuurden, D., additional, Hulleman, E., additional, Idema, S., additional, Noske, D., additional, Wolf, N., additional, Hendrikse, H., additional, Vandertop, P., additional, Kaspers, G. J., additional, Muller, K., additional, Schlamann, A., additional, Warmuth-Metz, M., additional, Pietsch, T., additional, Pietschmann, S., additional, Kortmann, R.-D., additional, Kramm, C. M., additional, von Bueren, A. O., additional, Walston, S., additional, Williams, T., additional, Hamstra, D., additional, Oh, K., additional, Pelloski, C., additional, Zhukova, N., additional, Pole, J., additional, Mistry, M., additional, Fried, I., additional, Lapperiere, N., additional, Dirks, P., additional, An, J., additional, Alon, N., additional, Nathan, P., additional, Greenberg, M., additional, and Malkin, D., additional
- Published
- 2013
- Full Text
- View/download PDF
15. Telomere maintenance and dysfunction predict recurrence in paediatric ependymoma
- Author
-
Tabori, U, primary, Wong, V, additional, Ma, J, additional, Shago, M, additional, Alon, N, additional, Rutka, J, additional, Bouffet, E, additional, Bartels, U, additional, Malkin, D, additional, and Hawkins, C, additional
- Published
- 2008
- Full Text
- View/download PDF
16. Molecular and clinical characterization of de novo and familial cases with microduplication 3q29: guidelines for copy number variation case reporting
- Author
-
Goobie, S., primary, Knijnenburg, J., additional, FitzPatrick, D., additional, Sharkey, F.H., additional, Lionel, A.C., additional, Marshall, C.R., additional, Azam, T., additional, Shago, M., additional, Chong, K., additional, Mendoza-Londono, R., additional, den Hollander, N.S., additional, Ruivenkamp, C., additional, Maher, E., additional, Tanke, H.J., additional, Szuhai, K., additional, Wintle, R.F., additional, and Scherer, S.W., additional
- Published
- 2008
- Full Text
- View/download PDF
17. High incidence of CALM-AF10 fusion and the identification of a novel fusion transcript in acute megakaryoblastic leukemia in children without Down's syndrome
- Author
-
Abdelhaleem, M, primary, Beimnet, K, additional, Kirby-Allen, M, additional, Naqvi, A, additional, Hitzler, J, additional, and Shago, M, additional
- Published
- 2006
- Full Text
- View/download PDF
18. Expression of a retinoic acid response element-hsplacZ transgene defines specific domains of transcriptional activity during mouse embryogenesis.
- Author
-
Rossant, J, primary, Zirngibl, R, additional, Cado, D, additional, Shago, M, additional, and Giguère, V, additional
- Published
- 1991
- Full Text
- View/download PDF
19. Molecular and clinical characterization of de novo and familial cases with microduplication 3q29: guidelines for copy number variation case reporting.
- Author
-
Goobie, S., Knijnenburg, J., FitzPatrick, D., Sharkey, F. H., Lionel, A. C., Marshall, C. R., Azam, T., Shago, M., K. Chong, Mendoza-Londono, R., den Hollander, N. S., Ruivenkamp, C., Maher, E., Tanke, H. J., Szuhai, K., Wintle, R. F., and Scherer, S. W.
- Subjects
CYTOGENETICS ,GENETIC polymorphisms ,GENOMICS ,PHENOTYPES ,NANOTECHNOLOGY - Abstract
Microdeletions of 3q29 have previously been reported, but the postulated reciprocal microduplication has only recently been observed. Here, cases from four families, two ascertained in Toronto (Canada) and one each from Edinburgh (UK) and Leiden (Netherlands), carrying microduplications of 3q29 are presented. These families have been characterized by cytogenetic and molecular techniques, and all individuals have been further characterized with genome-wide, high density single nucleotide polymorphism (SNP) arrays run at a single centre (The Centre for Applied Genomics, Toronto). In addition to polymorphic copy-number variants (CNV), all carry duplications of 3q29 ranging in size from 1.9 to 2.4 Mb, encompassing multiple genes and defining a minimum region of overlap of about 1.6 Mb bounded by clusters of segmental duplications that is remarkably similar in location to previously reported 3q29 microdeletions. Consistent with other reports, the phenotype is variable, although developmental delay and significant ophthalmological findings were recurrent, suggesting that dosage sensitivity of genes located within 3q29 is important for eye and CNS development. We also consider CNVs found elsewhere in the genome for their contribution to the phenotype. We conclude by providing preliminary guidelines for management and anticipatory care of families with this microduplication, thereby establishing a standard for CNV reporting. Copyright © 2009 S. Karger AG, Basel [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
20. Identification of a novel isoform of the retinoic acid receptor gamma expressed in the mouse embryo
- Author
-
Giguère, V, primary, Shago, M, additional, Zirngibl, R, additional, Tate, P, additional, Rossant, J, additional, and Varmuza, S, additional
- Published
- 1990
- Full Text
- View/download PDF
21. Isolation of a novel retinoic acid-responsive gene by selection of genomic fragments derived from CpG-island-enriched DNA
- Author
-
Shago, M and Giguére, V
- Abstract
One of the primary goals in transcription factor research is the elucidation of the genetic networks controlled by a factor or by members of a family of closely related factors. The pleiotropic effects of retinoic acid (PA) in the developing and adult animal are mediated by ligand-inducible transcription factors (RA receptors [RARs] and retinoid X receptors [RXRs]) that belong to the superfamily of nuclear receptors. Regulatory regions of PA effector genes contain RAR and RXR binding sites (RAR elements [RAREs] and RXR elements [RXREs]) that generally consist of direct or everted repeats of the core half-site motif, (A/G)G(G/T)TCA. In order to identify novel genes regulated by RA, we devised a selection strategy based on the premise that regulatory regions of a large number of housekeeping and tissue-specific genes are embodied within CpG island DNA. In this method, referred to as CpG-selected and amplified binding, fragments derived from the CpG island fraction of the murine genome are selected by a gel mobility shift assay using in vitro-transcribed and -translated RXR-RAR. Multiple rounds of selection coupled with amplification of the fragments by PCR enabled us to clone a population of CG-rich fragments of which approximately one-fifth contained consensus RAREs or RXREs. Twelve genomic fragments containing novel response elements are described, and the transcription unit associated with one of them, NN-84AG, was characterized in detail. The mouse NN-84AG transcript is upregulated by RA in F9 embryonal carcinoma cells and is homologous to an expressed sequence tag (EST41159) derived from a human infant brain cDNA library. Cloning of the murine NN8-4AG genomic sequence places the RXRE in the proximity of the transcription initiation sites of the gene. Although sequence analysis indicates that the EST41159 gene product is novel, a region of amino acid identity with sequences of a yeast polypeptide of, as yet, unknown function and the Drosophila trithorax protein suggests the presence of an evolutionarily and functionally conserved domain. Our study demonstrates that transcription factor binding sites and corresponding regulated genes can be identified by selecting fragments derived from the CpG island fraction of the genome.
- Published
- 1996
- Full Text
- View/download PDF
22. BI-21BRAF MUTATION AND CDKN2A DELETIONS DEFINE A CLINICALLY DISTINCT SUBGROUP OF CHILDHOOD SECONDARY HIGH GRADE GLIOMA
- Author
-
Mistry M, Zhukova N, Merico D, Rakopoulos P, Krishnatry R, Shago M, Stavropoulos J, Ray P, Mangerel J, Marc Remke, Buczkowicz P, Ramaswamy V, Rutka J, Dirks P, Taylor M, Bouffet E, Malkin D, Huang A, Hawkins C, and Tabori U
- Subjects
Abstracts ,Cancer Research ,Oncology ,Neurology (clinical) - Abstract
PURPOSE: Pediatric secondary high grade glioma (sHGG), which result from malignant transformation of low grade glioma (PLGG), are a poorly understood group of tumors with devastating outcomes. PATIENTS AND METHODS: We performed a population-based study combined with long-term follow-up of PLGG that transformed to sHGG. Exonic-sequencing and copy number alterations were investigated on a discovery cohort, followed by detailed genetic analysis of all tumors. Clinical and outcome data analysis of a genetically distinct subgroup was performed. RESULTS: sHGG were observed in 28/888 (3.2%) patients treated in Southern Ontario for PLGG with a median latency of 2.74 years (range, 0.18-20.3 years). sHGG were characterized by a high somatic mutation load (23 per genome). Alterations in chromatin modifying genes and major telomere maintenance pathways were observed in 57% and 54% of sHGG respectively. However, specific mutations in IDH1, H3F3A G34 and ATRX were extremely rare. The most recurrent somatic alterations were the oncogenic BRAF V600E mutation and deletion of the tumor suppressor gene CDKN2A, observed in 39% and 57% of sHGG respectively. Importantly, all BRAF V600E and 80% of CDKN2A alterations could be traced to the patient-matched PLGG counterparts. These early alterations were rarely observed in non-transformed PLGG (p < 0.0001) and primary childhood high grade glioma (p = 0.0023). The BRAF mutant sHGG subgroup was characterized by longer latency periods to transformation than non-BRAF mutant sHGG (median 6.59 versus 1.62 years; p < 0.0001). Furthermore, 5-year overall survival of children with BRAF mutant and wild-type PLGG that transformed were 75% ± 15% and 29% ± 12% respectively (p = 0.024). CONCLUSION: BRAF V600E mutations and CDKN2A deletions constitute a clinically distinct subtype of sHGG. The prolonged course to transformation provides a window of opportunity for aggressive surgical interventions, targeted therapy against oncogenic BRAF, and extended surveillance to potentially mitigate the devastating transformation event.
23. Alterations in ALK/ROS1/NTRK/MET drive a group of infantile hemispheric gliomas
- Author
-
Elisabeth J. Rushing, Bruce Crooks, Scott L. Coven, Uri Tabori, Eric Bouffet, Claire Li, Christopher Li, Josef Zamecnik, Ute Bartels, Cynthia Hawkins, P. Daniel McNeely, Inmaculada de Prada, Michael Brudno, Michael D. Taylor, Bev Wilson, Claudia C. Faria, Livia Garzia, Vijay Ramaswamy, Lenka Krskova, Christopher Dunham, Roberto Silva, Andres Morales La Madrid, Sylvia Cheng, Ofelia Cruz, Arun K. Ramani, Michael A. Grotzer, Donna L. Johnston, Jonathan L. Finlay, David Sumerauer, Maria Joao Gil-da-Costa, Scott Ryall, Ana Guerreiro Stucklin, Yvonne Zhong, Pasqualino De Antonellis, Anthony Arnoldo, Daniel R. Boue, Koichi Ichimura, Miguel Garcia Ariza, Jean Michaud, Marta Perez-Somarriba, Motoo Nagane, Frank van Landeghem, Kohei Fukuoka, Hiroaki Sakamoto, Paul E. Kowalski, Meredith S. Irwin, Michal Zapotocky, Taylor Bridge, Iris Fried, Liana Nobre, Monique Johnson, Jordan R. Hansford, Robert Siddaway, Mary Shago, Nataliya Zhukova, Byungjin Kim, Palma Solano, Yoshiko Nakano, Keita Terashima, Alvaro Lassaletta, Angelica Oviedo, Amulya NageswaraRao, Repositório da Universidade de Lisboa, Guerreiro Stucklin, A. S., Ryall, S., Fukuoka, K., Zapotocky, M., Lassaletta, A., Li, C., Bridge, T., Kim, B., Arnoldo, A., Kowalski, P. E., Zhong, Y., Johnson, M., Ramani, A. K., Siddaway, R., Nobre, L. F., de Antonellis, P., Dunham, C., Cheng, S., Boue, D. R., Finlay, J. L., Coven, S. L., de Prada, I., Perez-Somarriba, M., Faria, C. C., Grotzer, M. A., Rushing, E., Sumerauer, D., Zamecnik, J., Krskova, L., Garcia Ariza, M., Cruz, O., Morales La Madrid, A., Solano, P., Terashima, K., Nakano, Y., Ichimura, K., Nagane, M., Sakamoto, H., Gil-da-Costa, M. J., Silva, R., Johnston, D. L., Michaud, J., Wilson, B., van Landeghem, F. K. H., Oviedo, A., Mcneely, P. D., Crooks, B., Fried, I., Zhukova, N., Hansford, J. R., Nageswararao, A., Garzia, L., Shago, M., Brudno, M., Irwin, M. S., Bartels, U., Ramaswamy, V., Bouffet, E., Taylor, M. D., Tabori, U., and Hawkins, C.
- Subjects
MAPK/ERK pathway ,Oncology ,Epigenomics ,Male ,General Physics and Astronomy ,Whole Exome Sequencing ,Receptor tyrosine kinase ,0302 clinical medicine ,Protein-Tyrosine Kinase ,Cancer genomics ,Anaplastic lymphoma kinase ,Anaplastic Lymphoma Kinase ,lcsh:Science ,Exome sequencing ,Proto-Oncogene Protein ,Multidisciplinary ,Molecular medicine ,biology ,Brain Neoplasms ,Glioma ,Protein-Tyrosine Kinases ,Proto-Oncogene Proteins c-met ,Receptor Protein-Tyrosine Kinase ,Gene Expression Regulation, Neoplastic ,030220 oncology & carcinogenesis ,Female ,Survival Analysi ,Human ,medicine.medical_specialty ,Epigenomic ,Science ,Article ,General Biochemistry, Genetics and Molecular Biology ,Brain Neoplasm ,03 medical and health sciences ,Internal medicine ,Proto-Oncogene Proteins ,Exome Sequencing ,medicine ,ROS1 ,Humans ,Receptor, trkA ,Survival analysis ,business.industry ,Infant, Newborn ,Infant ,Receptor Protein-Tyrosine Kinases ,General Chemistry ,DNA Methylation ,medicine.disease ,Survival Analysis ,biology.protein ,lcsh:Q ,business ,030217 neurology & neurosurgery - Abstract
© The Author(s) 2019. Open Access. This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/., Infant gliomas have paradoxical clinical behavior compared to those in children and adults: low-grade tumors have a higher mortality rate, while high-grade tumors have a better outcome. However, we have little understanding of their biology and therefore cannot explain this behavior nor what constitutes optimal clinical management. Here we report a comprehensive genetic analysis of an international cohort of clinically annotated infant gliomas, revealing 3 clinical subgroups. Group 1 tumors arise in the cerebral hemispheres and harbor alterations in the receptor tyrosine kinases ALK, ROS1, NTRK and MET. These are typically single-events and confer an intermediate outcome. Groups 2 and 3 gliomas harbor RAS/MAPK pathway mutations and arise in the hemispheres and midline, respectively. Group 2 tumors have excellent long-term survival, while group 3 tumors progress rapidly and do not respond well to chemoradiation. We conclude that infant gliomas comprise 3 subgroups, justifying the need for specialized therapeutic strategies.
- Published
- 2019
24. Determinants of survival after first relapse of acute lymphoblastic leukemia: a Children's Oncology Group study.
- Author
-
Rheingold SR, Bhojwani D, Ji L, Xu X, Devidas M, Kairalla JA, Shago M, Heerema NA, Carroll AJ, Breidenbach H, Borowitz M, Wood BL, Angiolillo AL, Asselin BL, Bowman WP, Brown P, Dreyer ZE, Dunsmore KP, Hilden JM, Larsen E, Maloney K, Matloub Y, Mattano LA, Winter SS, Gore L, Winick NJ, Carroll WL, Hunger SP, Raetz EA, and Loh ML
- Subjects
- Humans, Child, Female, Male, Infant, Child, Preschool, Prognosis, Adolescent, Recurrence, Survival Rate, Neoplasm Recurrence, Local pathology, Neoplasm Recurrence, Local mortality, Oncogene Proteins, Fusion genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma mortality, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
Limited prognostic factors have been associated with overall survival (OS) post-relapse in childhood Acute Lymphoblastic Leukemia (ALL). Patients enrolled on 12 Children's Oncology Group frontline ALL trials (1996-2014) were analyzed to assess for additional prognostic factors associated with OS post-relapse. Among 16,115 patients, 2053 (12.7%) relapsed. Relapse rates were similar for B-ALL (12.5%) and T-ALL (11.2%) while higher for infants (34.2%). Approximately 50% of B-ALL relapses occurred late (≥36 months) and 72.5% involved the marrow. Conversely, 64.8% of T-ALL relapses occurred early (<18 months) and 47.1% involved the central nervous system. The 5-year OS post-relapse for the entire cohort was 48.9 ± 1.2%; B-ALL:52.5 ± 1.3%, T-ALL:35.5 ± 3.3%, and infant ALL:21.5 ± 3.9%. OS varied by early, intermediate and late time-to-relapse; 25.8 ± 2.4%, 49.5 ± 2.2%, and 66.4 ± 1.8% respectively for B-ALL and 29.8 ± 3.9%, 33.3 ± 7.6%, 58 ± 9.8% for T-ALL. Patients with ETV6::RUNX1 or Trisomy 4 + 10 had median time-to-relapse of 43 months and higher OS post-relapse 74.4 ± 3.1% and 70.2 ± 3.6%, respectively. Patients with hypodiploidy, KMT2A-rearrangement, and TCF3::PBX1 had short median time-to-relapse (12.5-18 months) and poor OS post-relapse (14.2 ± 6.1%, 31.9 ± 7.7%, 36.8 ± 6.6%). Site-of-relapse varied by cytogenetic subtype. This large dataset provided the opportunity to identify risk factors for OS post-relapse to inform trial design and highlight populations with dismal outcomes post-relapse., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
25. Genomic Determinants of Outcome in Acute Lymphoblastic Leukemia.
- Author
-
Chang TC, Chen W, Qu C, Cheng Z, Hedges D, Elsayed A, Pounds SB, Shago M, Rabin KR, Raetz EA, Devidas M, Cheng C, Angiolillo A, Baviskar P, Borowitz M, Burke MJ, Carroll A, Carroll WL, Chen IM, Harvey R, Heerema N, Iacobucci I, Wang JR, Jeha S, Larsen E, Mattano L, Maloney K, Pui CH, Ramirez NC, Salzer W, Willman C, Winick N, Wood B, Hunger SP, Wu G, Mullighan CG, and Loh ML
- Subjects
- Humans, Child, Male, Female, Case-Control Studies, Child, Preschool, Adolescent, Genomics, Infant, PAX5 Transcription Factor genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma mortality, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy
- Abstract
Purpose: Although cure rates for childhood acute lymphoblastic leukemia (ALL) exceed 90%, ALL remains a leading cause of cancer death in children. Half of relapses arise in children initially classified with standard-risk (SR) disease., Materials and Methods: To identify genomic determinants of relapse in children with SR ALL, we performed genome and transcriptome sequencing of diagnostic and remission samples of children with SR (n = 1,381) or high-risk B-ALL with favorable cytogenetic features (n = 115) enrolled on Children's Oncology Group trials. We used a case-control study design analyzing 439 patients who relapsed and 1,057 who remained in complete remission for at least 5 years., Results: Genomic subtype was associated with relapse, which occurred in approximately 50% of cases of PAX5 -altered ALL (odds ratio [OR], 3.31 [95% CI, 2.17 to 5.03]; P = 3.18 × 10
-8 ). Within high-hyperdiploid ALL, gain of chromosome 10 with disomy of chromosome 7 was associated with favorable outcome (OR, 0.27 [95% CI, 0.17 to 0.42]; P = 8.02 × 10-10 ; St Jude Children's Research Hospital validation cohort: OR, 0.22 [95% CI, 0.05 to 0.80]; P = .009), and disomy of chromosomes 10 and 17 with gain of chromosome 6 was associated with relapse (OR, 7.16 [95% CI, 2.63 to 21.51]; P = 2.19 × 10-5 ; validation cohort: OR, 21.32 [95% CI, 3.62 to 119.30]; P = .0004). Genomic alterations were associated with relapse in a subtype-dependent manner, including alterations of INO80 in ETV6::RUNX1 ALL, IKZF1 , and CREBBP in high-hyperdiploid ALL and FHIT in BCR::ABL1 -like ALL. Genomic alterations were also associated with the presence of minimal residual disease, including NRAS and CREBBP in high-hyperdiploid ALL., Conclusion: Genetic subtype, patterns of aneuploidy, and secondary genomic alterations determine risk of relapse in childhood ALL. Comprehensive genomic analysis is required for optimal risk stratification.- Published
- 2024
- Full Text
- View/download PDF
26. 45,X/46,XY mosaicism: Clinical manifestations and long term follow-up.
- Author
-
Alkhunaizi E, Albrecht JP, Aarabi M, Witchel SF, Wherrett D, Babul-Hirji R, Dupuis A, Chiniara L, Chater-Diehl E, Shago M, Shuman C, Rajkovic A, Yatsenko SA, and Chitayat D
- Subjects
- Child, Humans, Male, Female, Mosaicism, Follow-Up Studies, Retrospective Studies, Phenotype, Turner Syndrome diagnosis, Turner Syndrome genetics, Gonadal Dysgenesis, Mixed genetics, Neoplasms
- Abstract
45,X/46,XY chromosomal mosaicism presents a range of clinical manifestations, including phenotypes from Turner syndrome through genital abnormalities to apparently unaffected phenotypic males; however, the full clinical spectrum has not yet been fully delineated since prior studies on the clinical phenotype and associated risk of gonadal tumors included small cohorts and limited follow-up. To better describe the clinical manifestations and long-term outcome of patients with 45,X/46,XY mosaicism. We conducted a retrospective chart review of patients with 45,X/46,XY from three health centers (Hospital for Sick Children and Mount Sinai Hospital in Canada, and University of Pittsburgh Medical Center in United States). Of 100 patients with 45,X/46,XY karyotype, 47 were raised as females and 53 as males. Females were significantly shorter than males (p = 0.04) and height Z-score was significantly decreased with age for both genders (p = 0.02). Growth hormone (GH) treatment did not result in a significant height increase compared to the untreated group (p = 0.5). All females required puberty induction in contrast to majority of males. Five females were diagnosed with gonadal tumors, while no males were affected. Around 58% of patients exhibited at least one Turner syndrome stigmata. This study expands the clinical spectrum, long-term outcomes, and associated tumor risk in a large cohort of patients with 45,X/46,XY mosaicism. Additionally, it highlights our experience with GH therapy and prophylactic gonadectomy., (© 2023 Wiley Periodicals LLC.)
- Published
- 2024
- Full Text
- View/download PDF
27. Genomic landscape of Down syndrome-associated acute lymphoblastic leukemia.
- Author
-
Li Z, Chang TC, Junco JJ, Devidas M, Li Y, Yang W, Huang X, Hedges DJ, Cheng Z, Shago M, Carroll AJ, Heerema NA, Gastier-Foster J, Wood BL, Borowitz MJ, Sanclemente L, Raetz EA, Hunger SP, Feingold E, Rosser TC, Sherman SL, Loh ML, Mullighan CG, Yu J, Wu G, Lupo PJ, Rabin KR, and Yang JJ
- Subjects
- Animals, Mice, Mutation, Risk Factors, Genomics, Chromosome Aberrations, Down Syndrome complications, Down Syndrome genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma complications, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
Trisomy 21, the genetic cause of Down syndrome (DS), is the most common congenital chromosomal anomaly. It is associated with a 20-fold increased risk of acute lymphoblastic leukemia (ALL) during childhood and results in distinctive leukemia biology. To comprehensively define the genomic landscape of DS-ALL, we performed whole-genome sequencing and whole-transcriptome sequencing (RNA-Seq) on 295 cases. Our integrated genomic analyses identified 15 molecular subtypes of DS-ALL, with marked enrichment of CRLF2-r, IGH::IGF2BP1, and C/EBP altered (C/EBPalt) subtypes compared with 2257 non-DS-ALL cases. We observed abnormal activation of the CEBPD, CEBPA, and CEBPE genes in 10.5% of DS-ALL cases via a variety of genomic mechanisms, including chromosomal rearrangements and noncoding mutations leading to enhancer hijacking. A total of 42.3% of C/EBP-activated DS-ALL also have concomitant FLT3 point mutations or insertions/deletions, compared with 4.1% in other subtypes. CEBPD overexpression enhanced the differentiation of mouse hematopoietic progenitor cells into pro-B cells in vitro, particularly in a DS genetic background. Notably, recombination-activating gene-mediated somatic genomic abnormalities were common in DS-ALL, accounting for a median of 27.5% of structural alterations, compared with 7.7% in non-DS-ALL. Unsupervised hierarchical clustering analyses of CRLF2-rearranged DS-ALL identified substantial heterogeneity within this group, with the BCR::ABL1-like subset linked to an inferior event-free survival, even after adjusting for known clinical risk factors. These results provide important insights into the biology of DS-ALL and point to opportunities for targeted therapy and treatment individualization., (© 2023 by The American Society of Hematology.)
- Published
- 2023
- Full Text
- View/download PDF
28. Myeloproliferative Neoplasm Driven by ETV6-ABL1 in an Adolescent with Recent History of Burkitt Leukemia.
- Author
-
Renzi S, Algawahmed F, Davidson S, Langenberg KPS, Fuligni F, Ali S, Anderson N, Brunga L, Bartram J, Abdelhaleem M, Naqvi A, Beimnet K, Schuh A, Tierens A, Malkin D, Shlien A, Shago M, and Villani A
- Subjects
- Male, Humans, Adolescent, Protein-Tyrosine Kinases genetics, In Situ Hybridization, Fluorescence, Imatinib Mesylate therapeutic use, Burkitt Lymphoma, Leukemia, Myeloid, Acute genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology
- Abstract
ETV6-ABL1 gene fusion is a rare genetic rearrangement in a variety of malignancies, including myeloproliferative neoplasms (MPN), acute lymphoblastic leukemia (ALL), and acute myeloid leukemia (AML). Here, we report the case of a 16-year-old male diagnosed with a MPN, 7 months post-completion of treatment for Burkitt leukaemia. RNA sequencing analysis confirmed the presence of an ETV6-ABL1 fusion transcript, with an intact, in-frame ABL tyrosine-kinase domain. Of note, secondary ETV6-ABL1 -rearranged neoplastic diseases have not been reported to date. The patient was started on a tyrosine kinase inhibitor (TKI; imatinib) and, subsequently, underwent a 10/10 matched unrelated haematopoietic stem cell transplant. He is disease-free five years post-transplant. Definitive evidence of the prognostic influence of the ETV6-ABL1 fusion in haematological neoplasms is lacking; however, overall data suggest that it is a poor prognostic factor, particularly in patients with ALL and AML. The presence of this ETV6-ABL1 fusion should be more routinely investigated, especially in patients with a CML-like picture. More routine use of whole-genome and RNA sequencing analyses in clinical diagnostic care, in conjunction with conventional cytogenetics, will facilitate these investigations.
- Published
- 2023
- Full Text
- View/download PDF
29. The clinical utility of integrative genomics in childhood cancer extends beyond targetable mutations.
- Author
-
Villani A, Davidson S, Kanwar N, Lo WW, Li Y, Cohen-Gogo S, Fuligni F, Edward LM, Light N, Layeghifard M, Harripaul R, Waldman L, Gallinger B, Comitani F, Brunga L, Hayes R, Anderson ND, Ramani AK, Yuki KE, Blay S, Johnstone B, Inglese C, Hammad R, Goudie C, Shuen A, Wasserman JD, Venier RE, Eliou M, Lorenti M, Ryan CA, Braga M, Gloven-Brown M, Han J, Montero M, Spatare F, Whitlock JA, Scherer SW, Chun K, Somerville MJ, Hawkins C, Abdelhaleem M, Ramaswamy V, Somers GR, Kyriakopoulou L, Hitzler J, Shago M, Morgenstern DA, Tabori U, Meyn S, Irwin MS, Malkin D, and Shlien A
- Subjects
- Young Adult, Adolescent, Humans, Child, Mutation, Genomics, Transcriptome genetics, Homologous Recombination, Neoplasms drug therapy, Neoplasms genetics
- Abstract
We conducted integrative somatic-germline analyses by deeply sequencing 864 cancer-associated genes, complete genomes and transcriptomes for 300 mostly previously treated children and adolescents/young adults with cancer of poor prognosis or with rare tumors enrolled in the SickKids Cancer Sequencing (KiCS) program. Clinically actionable variants were identified in 56% of patients. Improved diagnostic accuracy led to modified management in a subset. Therapeutically targetable variants (54% of patients) were of unanticipated timing and type, with over 20% derived from the germline. Corroborating mutational signatures (SBS3/BRCAness) in patients with germline homologous recombination defects demonstrates the potential utility of PARP inhibitors. Mutational burden was significantly elevated in 9% of patients. Sequential sampling identified changes in therapeutically targetable drivers in over one-third of patients, suggesting benefit from rebiopsy for genomic analysis at the time of relapse. Comprehensive cancer genomic profiling is useful at multiple points in the care trajectory for children and adolescents/young adults with cancer, supporting its integration into early clinical management., (© 2022. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
30. Characterization of unusual iAMP21 B-lymphoblastic leukemia (iAMP21-ALL) from the Mayo Clinic and Children's Oncology Group.
- Author
-
Koleilat A, Smadbeck JB, Zepeda-Mendoza CJ, Williamson CM, Pitel BA, Golden CL, Xu X, Greipp PT, Ketterling RP, Hoppman NL, Peterson JF, Harrison CJ, Akkari YMN, Tsuchiya KD, Shago M, and Baughn LB
- Subjects
- Chromosome Aberrations, Humans, In Situ Hybridization, Fluorescence, Retrospective Studies, Core Binding Factor Alpha 2 Subunit genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
Acute lymphoblastic leukemia (B-ALL) with intrachromosomal amplification of chromosome 21 (iAMP21-ALL) represents a recurrent high-risk cytogenetic abnormality and accurate identification is critical for appropriate clinical management. Identification of iAMP21-ALL has historically relied on fluorescence in situ hybridization (FISH) using a RUNX1 probe. Current classification requires ≥ five copies of RUNX1 per cell and ≥ three additional copies of RUNX1 on a single abnormal iAMP21-chromosome. We sought to evaluate the performance of the RUNX1 probe in the identification of iAMP21-ALL. This study was a retrospective evaluation of iAMP21-ALL in the Mayo Clinic and Children's Oncology Group cohorts. Of 207 cases of iAMP21-ALL, 188 (91%) were classified as "typical" iAMP21-ALL, while 19 (9%) cases were classified as "unusual" iAMP21-ALL. The "unusual" iAMP21 cases did not meet the current definition of iAMP21 by FISH but were confirmed to have iAMP21 by chromosomal microarray. Half of the "unusual" iAMP21-ALL cases had less than five RUNX1 signals, while the remainder had ≥ five RUNX1 signals with some located apart from the abnormal iAMP21-chromosome. Nine percent of iAMP21-ALL cases fail to meet the FISH definition of iAMP21-ALL demonstrating that laboratories are at risk of misidentification of iAMP21-ALL when relying only on the RUNX1 FISH probe. Incorporation of chromosomal microarray testing circumvents these risks., (© 2022 The Authors. Genes, Chromosomes and Cancer published by Wiley Periodicals LLC.)
- Published
- 2022
- Full Text
- View/download PDF
31. Hypomorphic GINS3 variants alter DNA replication and cause Meier-Gorlin syndrome.
- Author
-
McQuaid ME, Ahmed K, Tran S, Rousseau J, Shaheen R, Kernohan KD, Yuki KE, Grover P, Dreseris ES, Ahmed S, Dupuis L, Stimec J, Shago M, Al-Hassnan ZN, Tremblay R, Maass PG, Wilson MD, Grunebaum E, Boycott KM, Boisvert FM, Maddirevula S, Faqeih EA, Almanjomi F, Khan ZU, Alkuraya FS, Campeau PM, Kannu P, Campos EI, and Wurtele H
- Subjects
- Animals, Chromosomal Proteins, Non-Histone, Congenital Microtia, DNA Replication genetics, Growth Disorders, Humans, Mice, Minichromosome Maintenance Proteins genetics, Patella abnormalities, Micrognathism genetics, Saccharomyces cerevisiae
- Abstract
The eukaryotic CDC45/MCM2-7/GINS (CMG) helicase unwinds the DNA double helix during DNA replication. The GINS subcomplex is required for helicase activity and is, therefore, essential for DNA replication and cell viability. Here, we report the identification of 7 individuals from 5 unrelated families presenting with a Meier-Gorlin syndrome-like (MGS-like) phenotype associated with hypomorphic variants of GINS3, a gene not previously associated with this syndrome. We found that MGS-associated GINS3 variants affecting aspartic acid 24 (D24) compromised cell proliferation and caused accumulation of cells in S phase. These variants shortened the protein half-life, altered key protein interactions at the replisome, and negatively influenced DNA replication fork progression. Yeast expressing MGS-associated variants of PSF3 (the yeast GINS3 ortholog) also displayed impaired growth, S phase progression defects, and decreased Psf3 protein stability. We further showed that mouse embryos homozygous for a D24 variant presented intrauterine growth retardation and did not survive to birth, and that fibroblasts derived from these embryos displayed accelerated cellular senescence. Taken together, our findings implicate GINS3 in the pathogenesis of MGS and support the notion that hypomorphic variants identified in this gene impaired cell and organismal growth by compromising DNA replication.
- Published
- 2022
- Full Text
- View/download PDF
32. Clinical phenotypes and prognostic features of embryonal tumours with multi-layered rosettes: a Rare Brain Tumor Registry study.
- Author
-
Khan S, Solano-Paez P, Suwal T, Lu M, Al-Karmi S, Ho B, Mumal I, Shago M, Hoffman LM, Dodgshun A, Nobusawa S, Tabori U, Bartels U, Ziegler DS, Hansford JR, Ramaswamy V, Hawkins C, Dufour C, André N, Bouffet E, and Huang A
- Subjects
- Brain Neoplasms mortality, Chemotherapy, Adjuvant, Child, Child, Preschool, Female, Gene Expression Regulation, Neoplastic, Humans, Infant, Infant, Newborn, Male, Neoplasms, Germ Cell and Embryonal mortality, Neurosurgical Procedures, Prognosis, Progression-Free Survival, Proportional Hazards Models, RNA, Messenger, Radiotherapy, Adjuvant, Brain Neoplasms pathology, Brain Neoplasms therapy, Combined Modality Therapy, Neoplasms, Germ Cell and Embryonal pathology, Neoplasms, Germ Cell and Embryonal therapy
- Abstract
Background: Embryonal tumours with multi-layered rosettes (ETMRs) are a newly recognised, rare paediatric brain tumour with alterations of the C19MC microRNA locus. Due to varied diagnostic practices and scarce clinical data, disease features and determinants of outcomes for these tumours are poorly defined. We did an integrated clinicopathological and molecular analysis of primary ETMRs to define clinical phenotypes, and to identify prognostic factors of survival and key treatment modalities for this orphan disease., Methods: Paediatric patients with primary ETMRs and tissue available for analyses were identified from the Rare Brain Tumor Consortium global registry. The institutional histopathological diagnoses were centrally re-reviewed as per the current WHO CNS tumour guidelines, using histopathological and molecular assays. Only patients with complete clinical, treatment, and survival data on Nov 30, 2019, were included in clinicopathological analyses. Among patients who received primary multi-modal curative regimens, event-free survival and overall survival were determined using Cox proportional hazard and log-rank analyses. Univariate and multivariable Cox proportional hazard regression was used to estimate hazard ratios (HRs) with 95% CIs for clinical, molecular, or treatment-related prognostic factors., Findings: 159 patients had a confirmed molecular diagnosis of primary ETMRs (median age at diagnosis 26 months, IQR 18-36) and were included in our clinicopathological analysis. ETMRs were predominantly non-metastatic (94 [73%] of 128 patients), arising from multiple sites; 84 (55%) of 154 were cerebral tumours and 70 (45%) of 154 arose at sites characteristic of other brain tumours. Hallmark C19MC alterations were seen in 144 (91%) of 159 patients; 15 (9%) were ETMR not otherwise specified. In patients treated with curative intent, event-free survival was 57% (95% CI 47-68) at 6 months and 31% (21-42) at 2 years; overall survival was 29% (20-38) at 2 years and 27% (18-37) at 4 years. Overall survival was associated with non-metastatic disease (HR 0·48, 95% CI 0·28-0·80; p=0·0057) and non-brainstem location (0·42 [0·22-0·81]; p=0·013) on univariate analysis, as well as with gross total resection (0·30, 0·16-0·58; p=0·0014), high-dose chemotherapy (0·35, 0·19-0·67; p=0·0020), and radiotherapy (0·21, 0·10-0·41; p<0·0001) on multivariable analysis. 2-year event-free and overall survival was 0% at 2 years in patients treated with conventional chemotherapy without radiotherapy (regardless of surgery extent), and 21% (95% CI 1-41) and 30% (6-54), respectively, in patients treated with high-dose chemotherapy, and gross total resection without radiotherapy. 2-year event-free survival in patients treated with high-dose chemotherapy and radiotherapy was 66% (95% CI 39-93) for patients with gross total resection and 44% (7-81) for patients with sub-total resection. 2-5-year overall survival was 66% (95% CI 33-99, p=0·038) for patients with gross total resection and 67% (36-98, p=0·0020) for patients with sub-total resection., Interpretation: Prompt molecular diagnosis and post-surgical treatment with intensive multi-modal therapy tailored to patient-specific risk features could improve ETMR survival., Funding: Canadian Institute of Health Research, Canada Research Chair Awards, Australian Lions Childhood Cancer Research Foundation, Spanish Society of Pediatrics, Consejería de Salud y Familias de la Junta de Andalucía, Miracle Marnie, Phoebe Rose Rocks, Tali's Funds, Garron Cancer Centre, Grace's Walk, Meagan's Hug, Brainchild, Nelina's Hope, and Jean Martel Foundation., Competing Interests: Declaration of interests We declare no competing interests., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
33. Practice patterns of prenatal and perinatal testing in Canadian cytogenetics laboratories.
- Author
-
Ashfield T, McCready E, Shago M, Wang H, Sinclair-Bourque E, Cappa E, Piche Marolt A, and Chun K
- Subjects
- Adult, Canada, Cytogenetics instrumentation, Cytogenetics methods, Cytogenetics statistics & numerical data, Female, Humans, Noninvasive Prenatal Testing trends, Practice Patterns, Physicians' statistics & numerical data, Pregnancy, Surveys and Questionnaires, Noninvasive Prenatal Testing methods, Practice Patterns, Physicians' trends
- Abstract
Objective: To survey patterns of practice in Canadian cytogenetics laboratories and evaluate whether newer technologies have influenced testing algorithms for the detection of common aneuploidies and other genomic imbalances in the prenatal and perinatal settings., Methods: Cytogenetics laboratories across Canada were invited to participate in two patterns-of-practice surveys: one in 2016 and one in 2019. They were asked to identify the prenatal and perinatal specimen types tested at their facility and which testing methods were used for initial testing and for follow-up., Results: All clinical laboratories performing prenatal testing offer rapid aneuploidy detection (RAD). Most laboratories also offer microarray analysis. A positive result is either followed up by karyotyping or no further testing is performed. For prenatal samples, a negative result may be followed up by microarray or karyotyping and is dependent on the reason for referral. For perinatal samples, availability of microarray to follow up a negative result is increasing., Conclusions: Since 2016, the availability of RAD as a first-line test in Canadian cytogenetics laboratories remains consistent, while microarray has become the preferred follow-up testing method over traditional karyotyping following a normal RAD result. Despite a universal healthcare system, disparities in prenatal and perinatal cytogenetic testing algorithms are apparent., (© 2021 John Wiley & Sons Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
34. Pediatric fibromyxoid soft tissue tumor with PLAG1 fusion: A novel entity?
- Author
-
Chung CT, Antonescu CR, Dickson BC, Chami R, Marrano P, Fan R, Shago M, Hameed M, and Thorner PS
- Subjects
- 14-3-3 Proteins genetics, Child, Preschool, Female, Fibroma pathology, Foot Diseases genetics, Head and Neck Neoplasms pathology, Humans, Infant, Male, Peptide Elongation Factor 1 genetics, RNA-Seq, Scalp, Skin Neoplasms pathology, DNA-Binding Proteins genetics, Fibroma genetics, Head and Neck Neoplasms genetics, Oncogene Fusion, Skin Neoplasms genetics
- Abstract
The classification of undifferentiated soft tissue tumors continues to evolve with the expanded application of molecular analysis in clinical practice. We report three cases of a unique soft tissue tumor in young children (5 months to 2 years old) displaying a purely fibromyxoid histology, with positive staining for desmin and CD34. In two cases, RNA sequencing detected a YWHAZ-PLAG1 gene fusion, while in the third case, a previously unreported EEF1A1-PLAG1 fusion was identified. PLAG1 fusions have been reported in several pathologic entities including pleomorphic adenoma, myoepithelial tumors of skin and soft tissue, and lipoblastoma, the latter occurring preferentially in young children. In these tumors, expression of a full length PLAG1 protein comes under the control of the constitutively active promoter of the partner gene in the fusion, and the current cases conform to that model. Overexpression of PLAG1 was confirmed by diffusely positive immunostaining for PLAG1 in all three cases. Our findings raise the possibility of a novel fibromyxoid neoplasm in childhood associated with these rare PLAG1 fusion variants. The only other report of a PLAG1-YWHAZ fusion occurred in a pediatric tumor diagnosed as a "fibroblastic lipoblastoma." This finding raises the possibility of a relationship with our three cases, even though our cases lacked any fat component. Further studies with regard to a shared pathogenesis are required., (© 2020 Wiley Periodicals LLC.)
- Published
- 2021
- Full Text
- View/download PDF
35. TERT promotor variant associated with poor clinical outcome in a patient with novel RBM15-MKL1 fusion-positive pediatric acute megakaryoblastic leukemia.
- Author
-
Langenberg-Ververgaert K, Renzi S, Fuligni F, Davidson S, Abdelhaleem M, Lo W, Malkin D, Shlien A, Shago M, Villani A, and Abla O
- Subjects
- Antineoplastic Combined Chemotherapy Protocols therapeutic use, Fatal Outcome, Female, Humans, Infant, Leukemia, Megakaryoblastic, Acute drug therapy, Leukemia, Megakaryoblastic, Acute genetics, Leukemia, Megakaryoblastic, Acute pathology, Oncogene Proteins, Fusion genetics, Polymorphism, Single Nucleotide, Promoter Regions, Genetic, RNA-Binding Proteins genetics, Telomerase genetics, Trans-Activators genetics
- Published
- 2021
- Full Text
- View/download PDF
36. First report of t(5;11) KMT2A-MAML1 fusion in de novo infant acute lymphoblastic leukemia.
- Author
-
Tandon S, Shago M, Davidson S, Kanwar N, Fuligni F, Shlien A, Whitlock J, Villani A, and Abla O
- Subjects
- Humans, Infant, Male, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma genetics, Prognosis, Chromosomes, Human, Pair 11 genetics, Chromosomes, Human, Pair 5 genetics, DNA-Binding Proteins genetics, Histone-Lysine N-Methyltransferase genetics, Myeloid-Lymphoid Leukemia Protein genetics, Oncogene Proteins, Fusion genetics, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma pathology, Transcription Factors genetics, Translocation, Genetic
- Abstract
Infant acute lymphoblastic leukemia (ALL) comprises 2.5%-5% of pediatric ALL with inferior survival compared to older children. A majority of infants (80%) with ALL harbor KMT2A gene rearrangement, which portends a poor prognosis. Approximately 94 different partner genes have been identified to date. The common rearrangements include t(4;11)(q21;q23)KMT2A-AFF1,t(11;19) (q23;p13.3)KMT2A-MLLT1 and t(9;11)(p22;q23)KMT2A-MLLT3. We report a novel translocation t(5;11)(q35;q23)KMT2A-MAML1 in newly diagnosed infant precursor B-ALL. Long-term follow-up and a larger number of patients are needed to better understand its prognostic significance., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
37. Evidence-based review of genomic aberrations in B-lymphoblastic leukemia/lymphoma: Report from the cancer genomics consortium working group for lymphoblastic leukemia.
- Author
-
Akkari YMN, Bruyere H, Hagelstrom RT, Kanagal-Shamanna R, Liu J, Luo M, Mikhail FM, Pitel BA, Raca G, Shago M, Shao L, Smith LR, Smolarek TA, Yenamandra A, and Baughn LB
- Subjects
- Adult, Age Factors, Child, Clinical Decision-Making, Cytogenetic Analysis, Disease-Free Survival, High-Throughput Nucleotide Sequencing, Humans, In Situ Hybridization, Fluorescence, Oligonucleotide Array Sequence Analysis, Patient Selection, Practice Guidelines as Topic, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma mortality, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy, Prognosis, Risk Assessment methods, Risk Assessment standards, Chromosome Aberrations, Genomics standards, Medical Oncology standards, Precursor Cell Lymphoblastic Leukemia-Lymphoma diagnosis
- Abstract
Clinical management and risk stratification of B-lymphoblastic leukemia/ lymphoma (B-ALL/LBL) depend largely on identification of chromosomal abnormalities obtained using conventional cytogenetics and Fluorescence In Situ Hybridization (FISH) testing. In the last few decades, testing algorithms have been implemented to support an optimal risk-oriented therapy, leading to a large improvement in overall survival. In addition, large scale genomic studies have identified multiple aberrations of prognostic significance that are not routinely tested by existing modalities. However, as chromosomal microarray analysis (CMA) and next-generation sequencing (NGS) technologies are increasingly used in clinical management of hematologic malignancies, these abnormalities may be more readily detected. In this article, we have compiled a comprehensive, evidence-based review of the current B-ALL literature, focusing on known and published subtypes described to date. More specifically, we describe the role of various testing modalities in the diagnosis, prognosis, and therapeutic relevance. In addition, we propose a testing algorithm aimed at assisting laboratories in the most effective detection of the underlying genomic abnormalities., Competing Interests: Declaration of Competing Interest Tanner Hagelstrom is employed by Illumina Inc. All other authors declare no conflict of interest., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
38. Integrated Molecular and Clinical Analysis of 1,000 Pediatric Low-Grade Gliomas.
- Author
-
Ryall S, Zapotocky M, Fukuoka K, Nobre L, Guerreiro Stucklin A, Bennett J, Siddaway R, Li C, Pajovic S, Arnoldo A, Kowalski PE, Johnson M, Sheth J, Lassaletta A, Tatevossian RG, Orisme W, Qaddoumi I, Surrey LF, Li MM, Waanders AJ, Gilheeney S, Rosenblum M, Bale T, Tsang DS, Laperriere N, Kulkarni A, Ibrahim GM, Drake J, Dirks P, Taylor MD, Rutka JT, Laughlin S, Shroff M, Shago M, Hazrati LN, D'Arcy C, Ramaswamy V, Bartels U, Huang A, Bouffet E, Karajannis MA, Santi M, Ellison DW, Tabori U, and Hawkins C
- Subjects
- Adolescent, Brain Neoplasms classification, Brain Neoplasms pathology, Child, Child, Preschool, Cohort Studies, Female, Gene Expression Profiling, Glioma classification, Glioma pathology, Humans, Infant, Infant, Newborn, Male, Mitogen-Activated Protein Kinases genetics, Neurofibromin 1 genetics, Oncogene Proteins, Fusion genetics, Proto-Oncogene Proteins B-raf genetics, ras Proteins genetics, Biomarkers, Tumor genetics, Brain Neoplasms genetics, DNA Copy Number Variations, Gene Expression Regulation, Neoplastic, Gene Rearrangement, Glioma genetics, Mutation
- Abstract
Pediatric low-grade gliomas (pLGG) are frequently driven by genetic alterations in the RAS-mitogen-activated protein kinase (RAS/MAPK) pathway yet show unexplained variability in their clinical outcome. To address this, we characterized a cohort of >1,000 clinically annotated pLGG. Eighty-four percent of cases harbored a driver alteration, while those without an identified alteration also often exhibited upregulation of the RAS/MAPK pathway. pLGG could be broadly classified based on their alteration type. Rearrangement-driven tumors were diagnosed at a younger age, enriched for WHO grade I histology, infrequently progressed, and rarely resulted in death as compared with SNV-driven tumors. Further sub-classification of clinical-molecular correlates stratified pLGG into risk categories. These data highlight the biological and clinical differences between pLGG subtypes and opens avenues for future treatment refinement., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
39. Genetic diversity in alveolar soft part sarcoma: A subset contain variant fusion genes, highlighting broader molecular kinship with other MiT family tumors.
- Author
-
Dickson BC, Chung CT, Hurlbut DJ, Marrano P, Shago M, Sung YS, Swanson D, Zhang L, and Antonescu CR
- Abstract
Alveolar soft part sarcoma (ASPS) is a rare malignancy that, since its initial description, remains a neoplasm of uncertain histogenesis. The disease-defining molecular event characterizing the diagnosis of ASPS is the ASPSCR1-TFE3 fusion gene. Following identification of an index case of ASPS with a novel TFE3 fusion partner, we performed a retrospective review to determine whether this represents an isolated event. We identified two additional cases, for a total of three cases lacking ASPSCR1 partners. The average patient age was 46 years (range, 17-65); two patients were female. The sites of origin included the transverse colon, foot, and dura. Each case exhibited a histomorphology typical of ASPS, and immunohistochemistry was positive for TFE3 in all cases. Routine molecular testing of the index patient demonstrated a HNRNPH3-TFE3 gene fusion; the remaining cases were found to have DVL2-TFE3 or PRCC-TFE3 fusion products. The latter two fusions have previously been identified in renal cell carcinoma; to our knowledge, this is the first report of a HNRNPH3-TFE3 gene fusion. These findings highlight a heretofore underrecognized genetic diversity in ASPS, which appears to more broadly molecularly overlap with that of translocation-associated renal cell carcinoma and PEComa. These results have immediate implications in the diagnosis of ASPS since assays reliant upon ASPSCR1 may yield a false negative result. While these findings further understanding of the molecular pathogenesis of ASPS, issues related to the histogenesis of this unusual neoplasm remain unresolved., (© 2019 Wiley Periodicals, Inc.)
- Published
- 2020
- Full Text
- View/download PDF
40. Masked hypodiploidy: Hypodiploid acute lymphoblastic leukemia (ALL) mimicking hyperdiploid ALL in children: A report from the Children's Oncology Group.
- Author
-
Carroll AJ, Shago M, Mikhail FM, Raimondi SC, Hirsch BA, Loh ML, Raetz EA, Borowitz MJ, Wood BL, Maloney KW, Mattano LA Jr, Larsen EC, Gastier-Foster J, Stonerock E, Ell D, Kahwash S, Devidas M, Harvey RC, Chen IL, Willman CL, Hunger SP, Winick NJ, Carroll WL, Rao KW, and Heerema NA
- Subjects
- Child, Chromosomes, Human, Humans, Karyotyping, Mosaicism, Diploidy, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
Hyperdiploidy with greater than 50 chromosomes is usually associated with favorable prognosis in pediatric acute lymphoblastic leukemia (ALL), whereas hypodiploidy with ≤43 chromosomes is associated with extremely poor prognosis. Sometimes, hypodiploidy is "masked" and patients do not have a karyotypically visible clone with ≤43 chromosomes. Instead, their abnormal karyotypes contain 50-78 or more chromosomes from doubling of previously hypodiploid cells. When the hypodiploid and doubled hyperdiploid clones are both present, patients can be identified by traditional test methods [karyotype, DNA Index (DI), fluorescence in situ hybridization (FISH)], but the incidence of masked hypodiploid cases in which only the doubled clone is visible is unknown. We analyzed 7013 patients with B-ALL enrolled in COG AALL03B1 (2003-2011) for whom chromosome studies were available. Of 115 patients with hypodiploidy (25-39 chromosomes), karyotypes of 40 showed only the hypodiploid clone, 47 showed mosaicism with both hypodiploid and hyperdiploid (doubled) karyotypes, and 28 with masked hypodiploidy showed only a hyperdiploid (doubled) clone. Unique karyotypic signatures were identified, and widespread loss of heterozygosity (LOH) was seen in the microsatellite panel for all patients with masked hypodiploidy. An increased awareness of the unusual karyotypic profile associated with a doubled hypodiploid clone and coordinated use of DI, FISH, and LOH studies when indicated can identify patients with masked hypodiploidy and allow appropriate treatment selection., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
41. Alterations in ALK/ROS1/NTRK/MET drive a group of infantile hemispheric gliomas.
- Author
-
Guerreiro Stucklin AS, Ryall S, Fukuoka K, Zapotocky M, Lassaletta A, Li C, Bridge T, Kim B, Arnoldo A, Kowalski PE, Zhong Y, Johnson M, Li C, Ramani AK, Siddaway R, Nobre LF, de Antonellis P, Dunham C, Cheng S, Boué DR, Finlay JL, Coven SL, de Prada I, Perez-Somarriba M, Faria CC, Grotzer MA, Rushing E, Sumerauer D, Zamecnik J, Krskova L, Garcia Ariza M, Cruz O, Morales La Madrid A, Solano P, Terashima K, Nakano Y, Ichimura K, Nagane M, Sakamoto H, Gil-da-Costa MJ, Silva R, Johnston DL, Michaud J, Wilson B, van Landeghem FKH, Oviedo A, McNeely PD, Crooks B, Fried I, Zhukova N, Hansford JR, Nageswararao A, Garzia L, Shago M, Brudno M, Irwin MS, Bartels U, Ramaswamy V, Bouffet E, Taylor MD, Tabori U, and Hawkins C
- Subjects
- Anaplastic Lymphoma Kinase genetics, Anaplastic Lymphoma Kinase metabolism, Brain Neoplasms classification, Brain Neoplasms metabolism, Female, Glioma classification, Glioma metabolism, Humans, Infant, Infant, Newborn, Male, Protein-Tyrosine Kinases genetics, Protein-Tyrosine Kinases metabolism, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-met genetics, Proto-Oncogene Proteins c-met metabolism, Receptor Protein-Tyrosine Kinases metabolism, Receptor, trkA genetics, Receptor, trkA metabolism, Survival Analysis, Exome Sequencing methods, Brain Neoplasms genetics, DNA Methylation, Epigenomics methods, Gene Expression Regulation, Neoplastic, Glioma genetics, Receptor Protein-Tyrosine Kinases genetics
- Abstract
Infant gliomas have paradoxical clinical behavior compared to those in children and adults: low-grade tumors have a higher mortality rate, while high-grade tumors have a better outcome. However, we have little understanding of their biology and therefore cannot explain this behavior nor what constitutes optimal clinical management. Here we report a comprehensive genetic analysis of an international cohort of clinically annotated infant gliomas, revealing 3 clinical subgroups. Group 1 tumors arise in the cerebral hemispheres and harbor alterations in the receptor tyrosine kinases ALK, ROS1, NTRK and MET. These are typically single-events and confer an intermediate outcome. Groups 2 and 3 gliomas harbor RAS/MAPK pathway mutations and arise in the hemispheres and midline, respectively. Group 2 tumors have excellent long-term survival, while group 3 tumors progress rapidly and do not respond well to chemoradiation. We conclude that infant gliomas comprise 3 subgroups, justifying the need for specialized therapeutic strategies.
- Published
- 2019
- Full Text
- View/download PDF
42. Immunohistochemistry for ATRX Can Miss ATRX Mutations: Lessons From Neuroblastoma.
- Author
-
Chami R, Marrano P, Teerapakpinyo C, Arnoldo A, Shago M, Shuangshoti S, and Thorner PS
- Subjects
- Adolescent, Child, Child, Preschool, False Negative Reactions, Female, Humans, Infant, Male, Mutation, Reproducibility of Results, X-linked Nuclear Protein genetics, Young Adult, Immunohistochemistry methods, Neuroblastoma genetics, X-linked Nuclear Protein analysis
- Abstract
Neuroblastoma is the most common extracranial solid tumor of childhood with a median age of presentation of 17 months. A common theme in high-risk neuroblastoma is maintenance of telomeres, one mechanism for which involves alternate lengthening of telomeres (ALT) associated with ATRX gene mutations. Mutations are believed to result in loss of ATRX protein, and therefore immunohistochemistry is used to detect mutations. We screened 133 cases of neuroblastoma by ATRX immunohistochemistry, and found 9 cases with partial to total absence of ATRX. Sequencing for ATRX mutations detected a mutation in 1 of 9 cases, suggesting immunostaining was not reliable for detecting mutations. To correlate immunostaining with ALT, fluorescence in situ hybridization (FISH) for ALT was performed in 6 of these cases and 5 (from 4 patients) showed ALT, implying impaired ATRX protein function, despite the failure to identify a mutation. Two other cases with large deletions in the ATRX gene showed diffusely positive staining for ATRX protein but showed ALT by FISH. Four of the 6 patients with ALT-positive tumors were over 5 years old. Therefore, 29 additional patients 5 years old and above with ATRX-positive tumors were screened for ALT by FISH and 6 additional cases with ALT were detected, bringing the total to 29% (10/34) of children 5 years old and above, 70% of which showed positive ATRX immunohistochemistry. Patients with ATRX mutations in neuroblastoma tend to have a more chronic and progressive course of disease. Screening neuroblastoma tumors at diagnosis for ATRX mutations may help identify patients who might benefit from personalized therapy directed against ALT. However, relaying on negative immunohistochemistry for ATRX protein to identify ALT in neuroblastoma may miss a significant proportion of patients. The addition of FISH for ALT as part of the diagnostic workup, especially for older children (5 y old and above), would help ensure that patients are correctly identified for anti-ALT therapy.
- Published
- 2019
- Full Text
- View/download PDF
43. An aggressive central giant cell granuloma in a pediatric patient: case report and review of literature.
- Author
-
Wang Y, Le A, El Demellawy D, Shago M, Odell M, and Johnson-Obaseki S
- Subjects
- Child, Diagnosis, Differential, Disease Progression, Female, Granuloma, Giant Cell diagnostic imaging, Granuloma, Giant Cell surgery, Humans, Mandibular Diseases diagnostic imaging, Mandibular Diseases surgery, Granuloma, Giant Cell pathology, Mandibular Diseases pathology
- Abstract
Background: Central giant cell granulomas are benign tumours of the mandible, presenting in children and young adults. Divided into non- and aggressive subtypes, the aggressive subtype is relatively rare and can occasionally progress rapidly, resulting in significant morbidity., Case Presentation: We present a case of an aggressive central giant cell granuloma (CGCG) in a six year-old female. The lesion originated in the right mandibular ramus and progressed rapidly to involve the condyle. Diagnosis was made using a combination of imaging and pathology. A timely en bloc resection of the hemi-mandible was performed with placement of a reconstructive titanium plate and condylar prosthesis., Conclusion: Our case demonstrates the importance of considering CGCG in the differential diagnosis of rapidly progressive mandibular lesions in the pediatric population. Prompt diagnosis and management can greatly improve long-term outcomes.
- Published
- 2019
- Full Text
- View/download PDF
44. MYCN Amplified Relapse Following Resolution of MYCN Nonamplified 4S Neuroblastoma With Placental Involvement: A Case Report and Review of the Literature.
- Author
-
Langenberg-Ververgaert KPS, Renzi S, Chung CT, Shago M, Lo W, Davidson S, Villani A, Baruchel S, Irwin MS, and Morgenstern DA
- Subjects
- Adult, Chromosome Aberrations, Female, Gene Amplification, Humans, Infant, Neuroblastoma congenital, Neuroblastoma pathology, Placenta Diseases, Pregnancy, Recurrence, N-Myc Proto-Oncogene Protein genetics, Neuroblastoma genetics
- Abstract
Congenital neuroblastoma with placental involvement is exceptionally rare, but mortality is high. Detailed examination of placenta including MYCN amplification and segmental chromosomal aberrations should be performed in all suspected cases, as it is noninvasive and readily available. Maternal dissemination has not been reported. In this manuscript, we describe an infant with placental diagnosis of MYCN nonamplified congenital neuroblastoma. This is the first report of a recurrence of congenital 4S neuroblastoma following resolution in which MYCN amplification is only detected in the recurrence. Germline sequencing using a large comprehensive cancer panel did not reveal variants in candidate cancer predisposition genes.
- Published
- 2019
- Full Text
- View/download PDF
45. Clinicopathologic Features of a Series of Primary Renal CIC-rearranged Sarcomas With Comprehensive Molecular Analysis.
- Author
-
Mangray S, Kelly DR, LeGuellec S, Fridman E, Aggarwal S, Shago M, Matoso A, Madison R, Pramanik S, Zhong S, Li R, Lombardo KA, Cramer S, Pressey J, Ross JS, Corona RJ, Bratslavsky G, Argani P, Coindre JM, Somers GR, Ali SM, and Yakirevich E
- Subjects
- Adolescent, Adult, Aged, 80 and over, Biomarkers, Tumor analysis, Biopsy, Needle, Female, Genetic Predisposition to Disease, Homeobox Protein Nkx-2.2, Homeodomain Proteins analysis, Homeodomain Proteins genetics, Humans, Immunohistochemistry, In Situ Hybridization, Fluorescence, Kidney Neoplasms chemistry, Kidney Neoplasms pathology, Kidney Neoplasms surgery, Middle Aged, Neoplasm Proteins genetics, Nephrectomy, Nuclear Proteins genetics, Phenotype, Sarcoma chemistry, Sarcoma pathology, Sarcoma surgery, Transcription Factors, Biomarkers, Tumor genetics, Gene Fusion, Gene Rearrangement, Kidney Neoplasms genetics, Repressor Proteins genetics, Sarcoma genetics
- Abstract
CIC-rearranged sarcomas rarely occur in visceral organs including the kidney. The most common fusion partner with CIC is the DUX4 gene, but variant fusion partners have also been reported. Herein, we describe the clinicopathologic features and comprehensive molecular profiling of 4 cases of primary renal CIC-rearranged sarcomas. All cases occurred in females, age range 13 to 82 years and included 3 resections and 1 needle biopsy specimen. There was a tendency for development of metastatic disease predominantly to the lungs and poor disease outcome despite different treatment strategies. Histologically, variable round cell (20% to 100%), spindle cell (0% to 80%), and rhabdoid morphologies (0% to 20%) were seen. By immunohistochemistry diffuse WT1 nuclear (2 to 3+, ∼90%) labeling was present in 1 case, with cytoplasmic staining in the others (3+, 40% to 75%). CD99 was focally positive in all 4 cases (≤10%); 1 case each was diffusely positive for c-myc (2 to 3+, ∼90%) and ETV4 (3+, ∼90%); 1 case was focally positive for c-myc (2+, ∼5%) and calretinin (2+, ∼5%); and all cases were negative for cytokeratin and NKX2.2. CIC rearrangement by fluorescence in situ hybridization was present in the 3 cases tested. Comprehensive genomic profiling (CGP) of 3 cases revealed a CIC-DUX4 fusion in 2 cases, and 1 CIC-NUTM1 fusion. All 4 CIC-rearranged renal sarcomas had low mutation burden, and except HLA-A and MLL mutations lacked genomic alterations in other oncogenic drivers. Material from the needle biopsy was insufficient for CGP but that case was positive with the DUX4 immunohistochemical stain as were the 2 CIC-DUX4 tumors. In conclusion, CIC-rearranged sarcomas rarely occur in the kidney with a tendency for poor outcome and in this series we illustrate an example with CIC-NUTM1 fusion, an emerging variant, at a visceral site. Testing by fluorescence in situ hybridization or CGP is optimal to avoid missing cases that harbor variant fusion partners.
- Published
- 2018
- Full Text
- View/download PDF
46. Aggressive embryonal rhabdomyosarcoma in a 3-month-old boy: A clinical and molecular analysis.
- Author
-
Renzi S, Langenberg-Ververgaert K, Fuligni F, Ryan AL, Davidson S, Anderson N, Hayes R, Hopyan S, Gerstle JT, Shago M, Chami R, Malkin D, Shlien A, Villani A, and Gupta AA
- Subjects
- Forkhead Box Protein O1 genetics, Humans, Infant, Lipase genetics, Male, Oncogene Proteins, Fusion genetics, Paired Box Transcription Factors genetics, Rhabdomyosarcoma, Embryonal pathology, Ubiquitin-Protein Ligases genetics, Chromosomes, Human, Pair 15 genetics, Chromosomes, Human, Pair 2 genetics, Rhabdomyosarcoma, Embryonal genetics, Translocation, Genetic
- Abstract
Rhabdomyosarcoma (RMS) represents the most common soft tissue sarcoma in the pediatric age group. While RMS has been traditionally classified on the basis of its histological appearance (with embryonal and alveolar being most common), it is now clear that the PAX-FOXO1 fusion product drives prognosis. We report here a case of pelvic embryonal RMS in a 3-month-old male who was subsequently found to have developed brain metastases during the course of chemotherapy. Cytogenetic analysis of the brain metastases at the time of autopsy as well as next-generation sequencing analysis revealed a reciprocal translocation involving the SH3 domain containing ring finger 3 gene (SH3RF3, on chromosome 2q13) and the Lipase C gene (LIPC, on chromosome 15q21.3). Due to the poor quality of the pretreatment and postresection samples, cytogenetics and NGS analysis looking for the presence of this balanced translocation in these specimens could not be performed. To the authors' knowledge, this translocation has never been described in RMS. Further studies are needed to determine the biological and clinical implications of this novel translocation.
- Published
- 2018
- Full Text
- View/download PDF
47. Rearrangement bursts generate canonical gene fusions in bone and soft tissue tumors.
- Author
-
Anderson ND, de Borja R, Young MD, Fuligni F, Rosic A, Roberts ND, Hajjar S, Layeghifard M, Novokmet A, Kowalski PE, Anaka M, Davidson S, Zarrei M, Id Said B, Schreiner LC, Marchand R, Sitter J, Gokgoz N, Brunga L, Graham GT, Fullam A, Pillay N, Toretsky JA, Yoshida A, Shibata T, Metzler M, Somers GR, Scherer SW, Flanagan AM, Campbell PJ, Schiffman JD, Shago M, Alexandrov LB, Wunder JS, Andrulis IL, Malkin D, Behjati S, and Shlien A
- Subjects
- Adolescent, Bone Neoplasms pathology, Child, DNA Replication, Evolution, Molecular, Female, Genome, Human, Humans, Male, Mutation, Neoplasm Metastasis, Neoplasm Recurrence, Local genetics, Neoplasm Recurrence, Local pathology, Soft Tissue Neoplasms pathology, Bone Neoplasms genetics, Gene Rearrangement, Oncogene Proteins, Fusion genetics, Sarcoma, Ewing genetics, Soft Tissue Neoplasms genetics
- Abstract
Sarcomas are cancers of the bone and soft tissue often defined by gene fusions. Ewing sarcoma involves fusions between EWSR1 , a gene encoding an RNA binding protein, and E26 transformation-specific (ETS) transcription factors. We explored how and when EWSR1-ETS fusions arise by studying the whole genomes of Ewing sarcomas. In 52 of 124 (42%) of tumors, the fusion gene arises by a sudden burst of complex, loop-like rearrangements, a process called chromoplexy, rather than by simple reciprocal translocations. These loops always contained the disease-defining fusion at the center, but they disrupted multiple additional genes. The loops occurred preferentially in early replicating and transcriptionally active genomic regions. Similar loops forming canonical fusions were found in three other sarcoma types. Chromoplexy-generated fusions appear to be associated with an aggressive form of Ewing sarcoma. These loops arise early, giving rise to both primary and relapse Ewing sarcoma tumors, which can continue to evolve in parallel., (Copyright © 2018 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2018
- Full Text
- View/download PDF
48. Fluorescent In Situ Hybridization for TP53 in the Diagnosis of Pediatric Osteogenic Sarcoma.
- Author
-
Marrano P, Shago M, Somers GR, and Thorner PS
- Subjects
- Age of Onset, Biomarkers, Tumor analysis, Bone Neoplasms chemistry, Bone Neoplasms pathology, Humans, Immunohistochemistry, Introns, Osteosarcoma chemistry, Osteosarcoma pathology, Predictive Value of Tests, Tumor Suppressor Protein p53 analysis, Biomarkers, Tumor genetics, Bone Neoplasms genetics, Gene Rearrangement, In Situ Hybridization, Fluorescence, Osteosarcoma genetics, Tumor Suppressor Protein p53 genetics
- Abstract
Osteogenic sarcoma (OS) is the most common malignant bone tumor in children and adolescents. Despite advances in molecular genetic characterization of pediatric and adult tumors, the diagnosis of OS still depends almost entirely on light microscopy. The lack of consistent genetic changes in OS has greatly hindered the development of any diagnostic molecular test. Recently, whole-genome sequencing has shown that ~50% of cases of OS have a translocation involving the TP53 gene with breakpoints confined to the first intron. We developed a 2 color break-apart fluorescent in situ hybridization (FISH) probe for intron 1 of TP53 and applied it to an archived series to assess its diagnostic utility. The study group included 37 cases of OS (including osteoblastic, chondroblastic, and fibroblastic), as well as 53 cases of non-OS pediatric sarcomas (including Ewing sarcoma, rhabdomyosarcoma, undifferentiated small cell sarcoma, CCNB3-BCOR sarcoma, CIC-DUX sarcoma, synovial sarcoma, and malignant peripheral nerve sheath tumor) and 27 cases of benign bone lesions (including osteoblastoma, chondromyxoid fibroma, fibrous dysplasia, and fibro-osseous dysplasia). A rearranged signal was found in 20/37 cases (54%) of OS and in none of the other sarcomas or benign bone lesions, giving the FISH test 100% specificity for a diagnosis of OS. p53 immunostaining was generally not predictive of the results obtained by FISH and could not substitute for this test. This FISH probe offers a simple and specific genetic test to aid in the diagnosis of OS, despite the genetic complexity of this tumor.
- Published
- 2018
- Full Text
- View/download PDF
49. Multiplex Detection of Pediatric Low-Grade Glioma Signature Fusion Transcripts and Duplications Using the NanoString nCounter System.
- Author
-
Ryall S, Arnoldo A, Krishnatry R, Mistry M, Khor K, Sheth J, Ling C, Leung S, Zapotocky M, Guerreiro Stucklin A, Lassaletta A, Shago M, Tabori U, and Hawkins CE
- Subjects
- Biomarkers, Tumor genetics, Biopsy, Brain Neoplasms diagnosis, Cohort Studies, Comparative Genomic Hybridization, Female, Glioma diagnosis, Humans, In Situ Hybridization, Fluorescence, Male, Mutation genetics, Pediatrics, Receptor, Fibroblast Growth Factor, Type 1 genetics, Brain Neoplasms genetics, Glioma genetics, Oncogene Proteins, Fusion genetics, Proto-Oncogene Proteins B-raf genetics
- Abstract
Previous studies identified recurrent fusion and duplication events in pediatric low-grade glioma (pLGG). In addition to their role in diagnosis, the presence of these events aid in dictating therapy and predicting patient survival. Clinically, BRAF alterations are most commonly identified using fluorescent in situ hybridization (FISH). However, this method is costly, labor-intensive and does not identify nonBRAF events. Here, we evaluated the NanoString nCounter gene expression system for detecting 32 of the most commonly reported fusion/duplication events in pLGG. The assay was validated on 90 pLGG samples using FISH as the gold standard and showed sensitivity and specificity of 97% and 98%, respectively. We next profiled formalin-fixed paraffin-embedded preserved biopsy specimens from 429 pLGG cases. 171 (40%) of the cases within our cohort tested positive for a fusion or duplication event contained within our panel. These events, in order of prevalence, were KIAA1549-BRAF 16;9 (89/171, 52.0%), KIAA1549-BRAF 15;9 (42/171, 24.6%), KIAA1549-BRAF 16;11 (14/171, 8.2%), FGFR1-TACC1 17;7 (13/171, 7.6%), MYBL1 duplication (5/171, 2.9%), KIAA1549-BRAF 18;10 (4/171, 2.3%), KIAA1549-BRAF 15;11 (2/171, 1.2%), FAM131B-BRAF 2;9 (1/171, 0.6%), and RNF130-BRAF 3;9 (1/171, 0.6%). This work introduces NanoString as a viable clinical replacement for the detection of fusion and duplication events in pLGG., (© 2017 American Association of Neuropathologists, Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
50. The clinical impact of copy number variants in inherited bone marrow failure syndromes.
- Author
-
Waespe N, Dhanraj S, Wahala M, Tsangaris E, Enbar T, Zlateska B, Li H, Klaassen RJ, Fernandez CV, Cuvelier GDE, Wu JK, Pastore YD, Silva M, Lipton JH, Brossard J, Michon B, Abish S, Steele M, Sinha R, Belletrutti MJ, Breakey VR, Jardine L, Goodyear L, Kofler L, Cada M, Sung L, Shago M, Scherer SW, and Dror Y
- Abstract
Inherited bone marrow failure syndromes (IBMFSs) comprise a genetically heterogeneous group of diseases with hematopoietic failure and a wide array of physical malformations. Copy number variants (CNVs) were reported in some IBMFSs. It is unclear what impact CNVs play in patients evaluated for a suspected diagnosis of IBMFS. Clinical and genetic data of 323 patients from the Canadian Inherited Marrow Failure Registry from 2001 to 2014, who had a documented genetic work-up, were analyzed. Cases with pathogenic CNVs (at least 1 kilobasepairs) were compared to cases with other mutations. Genotype-phenotype correlations were performed to assess the impact of CNVs. Pathogenic nucleotide-level mutations were found in 157 of 303 tested patients (51.8%). Genome-wide CNV analysis by single nucleotide polymorphism arrays or comparative genomic hybridization arrays revealed pathogenic CNVs in 11 of 67 patients tested (16.4%). In four of these patients, identification of CNV was crucial for establishing the correct diagnosis as their clinical presentation was ambiguous. Eight additional patients were identified to harbor pathogenic CNVs by other methods. Of the 19 patients with pathogenic CNVs, four had compound-heterozygosity of a CNV with a nucleotide-level mutation. Pathogenic CNVs were associated with more extensive non-hematological organ system involvement ( p =0.0006), developmental delay ( p =0.006) and short stature ( p =0.04) compared to nucleotide-level mutations. In conclusion, a significant proportion of patients with IBMFSs harbor pathogenic CNVs which were associated with a more extensive non-hematological phenotype in this cohort. Patients with a phenotype suggestive of IBMFSs but without identification of pathogenic nucleotide-level mutations should undergo specific testing for CNVs., Competing Interests: CONFLICT OF INTEREST All authors declare no conflict of interest.
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.