Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 148(1-2): 59-71, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22265402

ABSTRACT

Genomic rearrangements are thought to occur progressively during tumor development. Recent findings, however, suggest an alternative mechanism, involving massive chromosome rearrangements in a one-step catastrophic event termed chromothripsis. We report the whole-genome sequencing-based analysis of a Sonic-Hedgehog medulloblastoma (SHH-MB) brain tumor from a patient with a germline TP53 mutation (Li-Fraumeni syndrome), uncovering massive, complex chromosome rearrangements. Integrating TP53 status with microarray and deep sequencing-based DNA rearrangement data in additional patients reveals a striking association between TP53 mutation and chromothripsis in SHH-MBs. Analysis of additional tumor entities substantiates a link between TP53 mutation and chromothripsis, and indicates a context-specific role for p53 in catastrophic DNA rearrangements. Among these, we observed a strong association between somatic TP53 mutations and chromothripsis in acute myeloid leukemia. These findings connect p53 status and chromothripsis in specific tumor types, providing a genetic basis for understanding particularly aggressive subtypes of cancer.


Subject(s)
Brain Neoplasms/genetics , Gene Rearrangement , Medulloblastoma/genetics , Tumor Suppressor Protein p53/genetics , Animals , Child , Chromosome Aberrations , DNA Copy Number Variations , DNA Mutational Analysis , Disease Models, Animal , Humans , Leukemia, Myeloid, Acute/genetics , Li-Fraumeni Syndrome/physiopathology , Mice , Middle Aged
2.
Nature ; 482(7384): 226-31, 2012 Jan 29.
Article in English | MEDLINE | ID: mdl-22286061

ABSTRACT

Glioblastoma multiforme (GBM) is a lethal brain tumour in adults and children. However, DNA copy number and gene expression signatures indicate differences between adult and paediatric cases. To explore the genetic events underlying this distinction, we sequenced the exomes of 48 paediatric GBM samples. Somatic mutations in the H3.3-ATRX-DAXX chromatin remodelling pathway were identified in 44% of tumours (21/48). Recurrent mutations in H3F3A, which encodes the replication-independent histone 3 variant H3.3, were observed in 31% of tumours, and led to amino acid substitutions at two critical positions within the histone tail (K27M, G34R/G34V) involved in key regulatory post-translational modifications. Mutations in ATRX (α-thalassaemia/mental retardation syndrome X-linked) and DAXX (death-domain associated protein), encoding two subunits of a chromatin remodelling complex required for H3.3 incorporation at pericentric heterochromatin and telomeres, were identified in 31% of samples overall, and in 100% of tumours harbouring a G34R or G34V H3.3 mutation. Somatic TP53 mutations were identified in 54% of all cases, and in 86% of samples with H3F3A and/or ATRX mutations. Screening of a large cohort of gliomas of various grades and histologies (n = 784) showed H3F3A mutations to be specific to GBM and highly prevalent in children and young adults. Furthermore, the presence of H3F3A/ATRX-DAXX/TP53 mutations was strongly associated with alternative lengthening of telomeres and specific gene expression profiles. This is, to our knowledge, the first report to highlight recurrent mutations in a regulatory histone in humans, and our data suggest that defects of the chromatin architecture underlie paediatric and young adult GBM pathogenesis.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Chromatin/genetics , Glioblastoma/genetics , Histones/genetics , Mutation/genetics , Adaptor Proteins, Signal Transducing/genetics , Base Sequence , Child , Chromatin/metabolism , Co-Repressor Proteins , DNA Helicases/genetics , DNA Mutational Analysis , Exome/genetics , Gene Expression Profiling , Histones/metabolism , Humans , Molecular Chaperones , Molecular Sequence Data , Nuclear Proteins/genetics , Telomere/genetics , Tumor Suppressor Protein p53/genetics , X-linked Nuclear Protein
3.
Pediatr Blood Cancer ; 64(2): 275-278, 2017 02.
Article in English | MEDLINE | ID: mdl-27718322

ABSTRACT

A cerebellar pilocytic astrocytoma (PA) in a child recurred first with a PA histology and then with features of a ganglioglioma (GG). Molecular genetic analyses of the tumors confirmed a BRAF V600E mutation in all. They also all harbored a T202M mutation in ERK1, a kinase downstream of BRAF that is implicated in glial versus neuronal differentiation. The GG sample contained several variants that were not present in the PA samples; in particular, it had a truncating mutation in MAP2. These findings not only underscore the role of BRAF as oncogenic driver but also suggest that other genes may influence tumor morphology.


Subject(s)
Astrocytoma/genetics , Biomarkers, Tumor/genetics , Brain Neoplasms/etiology , Cerebellar Neoplasms/genetics , Ganglioglioma/etiology , Mutation/genetics , Proto-Oncogene Proteins B-raf/genetics , Astrocytoma/complications , Astrocytoma/pathology , Brain Neoplasms/pathology , Cerebellar Neoplasms/complications , Cerebellar Neoplasms/pathology , Child, Preschool , DNA Mutational Analysis , Female , Ganglioglioma/pathology , Humans , Neoplasm Recurrence, Local , Prognosis
4.
Acta Neuropathol ; 128(5): 615-27, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25077668

ABSTRACT

A remarkably large number of "epigenetic regulators" have been recently identified to be altered in cancers and a rapidly expanding body of literature points to "epigenetic addiction" (an aberrant epigenetic state to which a tumor is addicted) as a new previously unsuspected mechanism of oncogenesis. Although mutations are also found in canonical signaling pathway genes, we and others identified chromatin-associated proteins to be more commonly altered by somatic alterations than any other class of oncoprotein in several subgroups of childhood high-grade brain tumors. Furthermore, as these childhood malignancies carry fewer non-synonymous somatic mutations per case in contrast to most adult cancers, these mutations are likely drivers in these tumors. Herein, we will use as examples of this novel hallmark of oncogenesis high-grade astrocytomas, including glioblastoma, and a subgroup of embryonal tumors, embryonal tumor with multilayered rosettes (ETMR) to describe the novel molecular defects uncovered in these deadly tumors. We will further discuss evidence for their profound effects on the epigenome. The relative genetic simplicity of these tumors promises general insights into how mutations in the chromatin machinery modify downstream epigenetic signatures to drive transformation, and how to target this plastic genetic/epigenetic interface.


Subject(s)
Brain Neoplasms/genetics , Carcinogenesis/genetics , Epigenesis, Genetic/genetics , Humans , Pediatrics
5.
Lancet Oncol ; 14(12): 1200-7, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24140199

ABSTRACT

BACKGROUND: Recurrent medulloblastoma is a therapeutic challenge because it is almost always fatal. Studies have confirmed that medulloblastoma consists of at least four distinct subgroups. We sought to delineate subgroup-specific differences in medulloblastoma recurrence patterns. METHODS: We retrospectively identified a discovery cohort of all recurrent medulloblastomas at the Hospital for Sick Children (Toronto, ON, Canada) from 1994 to 2012 (cohort 1), and established molecular subgroups using a nanoString-based assay on formalin-fixed paraffin-embedded tissues or frozen tissue. The anatomical site of recurrence (local tumour bed or leptomeningeal metastasis), time to recurrence, and survival after recurrence were assessed in a subgroup-specific manner. Two independent, non-overlapping cohorts (cohort 2: samples from patients with recurrent medulloblastomas from 13 centres worldwide, obtained between 1991 and 2012; cohort 3: samples from patients with recurrent medulloblastoma obtained at the NN Burdenko Neurosurgical Institute [Moscow, Russia] between 1994 and 2011) were analysed to confirm and validate observations. When possible, molecular subgrouping was done on tissue obtained from both the initial surgery and at recurrence. RESULTS: Cohort 1 consisted of 30 patients with recurrent medulloblastomas; nine with local recurrences, and 21 with metastatic recurrences. Cohort 2 consisted of 77 patients and cohort 3 of 96 patients with recurrent medulloblastoma. Subgroup affiliation remained stable at recurrence in all 34 cases with available matched primary and recurrent pairs (five pairs from cohort 1 and 29 pairs from cohort 2 [15 SHH, five group 3, 14 group 4]). This finding was validated in 17 pairs from cohort 3. When analysed in a subgroup-specific manner, local recurrences in cohort 1 were more frequent in SHH tumours (eight of nine [89%]) and metastatic recurrences were more common in group 3 and group 4 tumours (17 of 20 [85%] with one WNT, p=0·0014, local vs metastatic recurrence, SHH vs group 3 vs group 4). The subgroup-specific location of recurrence was confirmed in cohort 2 (p=0·0013 for local vs metastatic recurrence, SHH vs group 3 vs group 4,), and cohort 3 (p<0·0001). Treatment with craniospinal irradiation at diagnosis was not significantly associated with the anatomical pattern of recurrence. Survival after recurrence was significantly longer in patients with group 4 tumours in cohort 1 (p=0·013) than with other subgroups, which was confirmed in cohort 2 (p=0·0075), but not cohort 3 (p=0·70). INTERPRETATION: Medulloblastoma does not change subgroup at the time of recurrence, reinforcing the stability of the four main medulloblastoma subgroups. Significant differences in the location and timing of recurrence across medulloblastoma subgroups have potential treatment ramifications. Specifically, intensified local (posterior fossa) therapy should be tested in the initial treatment of patients with SHH tumours. Refinement of therapy for patients with group 3 or group 4 tumours should focus on metastases.


Subject(s)
Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , Medulloblastoma/genetics , Medulloblastoma/secondary , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Adolescent , Canada , Cerebellar Neoplasms/classification , Cerebellar Neoplasms/mortality , Cerebellar Neoplasms/therapy , Child , Child, Preschool , Europe , Female , Genetic Predisposition to Disease , Humans , Kaplan-Meier Estimate , Male , Medulloblastoma/classification , Medulloblastoma/mortality , Medulloblastoma/therapy , Neoplasm Recurrence, Local/classification , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/therapy , Phenotype , Principal Component Analysis , Prognosis , Retrospective Studies , Risk Factors , Time Factors , United States
6.
Surg Oncol Clin N Am ; 33(2): 387-408, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38401916

ABSTRACT

Soft tissue sarcomas (STSs), including gastrointestinal stromal tumors (GISTs), are mesenchymal neoplasms with heterogeneous clinical behavior and represent broad categories comprising multiple distinct biologic entities. Multidisciplinary management of these rare tumors is critical. To date, multiple studies have outlined the importance of biological characterization of mesenchymal tumors and have identified key molecular alterations which drive tumor biology. GIST has represented a flagship for targeted therapy in solid tumors with the advent of imatinib which has revolutionized the way we treat this malignancy. Herein, the authors discuss the importance of biological and molecular diagnostics in managing STS and GIST patients.


Subject(s)
Gastrointestinal Stromal Tumors , Sarcoma , Soft Tissue Neoplasms , Humans , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/genetics , Precision Medicine , Sarcoma/drug therapy , Sarcoma/genetics , Sarcoma/pathology , Imatinib Mesylate/therapeutic use , Sodium Tetradecyl Sulfate/therapeutic use
7.
Curr Opin Oncol ; 25(6): 665-73, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24097106

ABSTRACT

PURPOSE OF REVIEW: Glial tumors of the central nervous system (CNS) are the leading cause of cancer-related death and morbidity in children. Their diagnosis/prognosis relies mainly on clinical and histopathological factors. However, pathological grading is particularly challenging as there is substantial molecular heterogeneity in pediatric CNS tumors, which results in variable biological behavior in tumors with potentially identical histological diagnoses or limited reliable measures of classification for given subgroups. Novel molecular markers/pathways identified by integrated genomic/transcriptomic/epigenomic studies of cohorts of pediatric gliomas are revolutionizing this field and are summarized herein. RECENT FINDINGS: Studies of pediatric gliomas have identified unexpected oncogenic pathways implicated in gliomagenesis. These range from a single pathway/molecule defect such as abnormalities of the mitogen-activated-protein-kinase pathway considered to be a hallmark of pilocytic astrocytomas, to alterations in epigenomic modulators in higher-grade tumors. Importantly, the type, timing, and spatial clustering of these molecular alterations provide a better understanding of the pathogenesis of gliomas and critical markers for therapy that will help refine pathological grading. SUMMARY: Reappraisal of glioma classification using these novel biomarkers will likely change practice toward molecular pathology and their integration into clinical trials will enable personalized therapies based on the molecular fingerprint of individual tumors.


Subject(s)
Astrocytoma/pathology , Biomarkers, Tumor , Central Nervous System Neoplasms/pathology , Genetic Therapy , Glioma/pathology , Adolescent , Adult , Astrocytoma/genetics , Astrocytoma/therapy , Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/therapy , Child , Child, Preschool , Epigenomics , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/therapy , Humans , Immunohistochemistry , Male , Mutation , Prognosis , Signal Transduction
8.
Acta Neuropathol ; 126(2): 291-301, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23660940

ABSTRACT

Pilocytic astrocytomas (PAs) are the most common brain tumors in pediatric patients and can cause significant morbidity, including chronic neurological deficiencies. They are characterized by activating alterations in the mitogen-activated protein kinase pathway, but little else is known about their development. To map the global DNA methylation profiles of these tumors, we analyzed 62 PAs and 7 normal cerebellum samples using Illumina 450K microarrays. These data revealed two subgroups of PA that separate according to tumor location (infratentorial versus supratentorial), and identified key neural developmental genes that are differentially methylated between the two groups, including NR2E1 and EN2. Integration with transcriptome microarray data highlighted significant expression differences, which were unexpectedly associated with a strong positive correlation between methylation and expression. Differentially methylated probes were often identified within the gene body and/or regions up- or downstream of the gene, rather than at the transcription start site. We also identified a large number of differentially methylated genes between cerebellar PAs and normal cerebellum, which were again enriched for developmental genes. In addition, we found a significant association between differentially methylated genes and SUZ12 binding sites, indicating potential disruption of the polycomb repressor complex 2 (PRC2). Taken together, these data suggest that PA from different locations in the brain may arise from region-specific cells of origin, and highlight the potential disruption of key developmental regulators during tumorigenesis. These findings have implications for future basic research and clinical trials, as therapeutic targets and drug sensitivity may differ according to tumor location.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , Cerebellar Neoplasms/genetics , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Astrocytoma/pathology , Binding Sites/genetics , Brain Neoplasms/pathology , Cerebellar Neoplasms/pathology , Child , DNA Methylation/genetics , Gene Expression Profiling , Genes, Developmental/genetics , Humans , Neoplasm Proteins , Oligonucleotide Array Sequence Analysis , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Transcription Factors
9.
Acta Neuropathol ; 125(5): 659-69, 2013 May.
Article in English | MEDLINE | ID: mdl-23417712

ABSTRACT

Recurrent mutations affecting the histone H3.3 residues Lys27 or indirectly Lys36 are frequent drivers of pediatric high-grade gliomas (over 30% of HGGs). To identify additional driver mutations in HGGs, we investigated a cohort of 60 pediatric HGGs using whole-exome sequencing (WES) and compared them to 543 exomes from non-cancer control samples. We identified mutations in SETD2, a H3K36 trimethyltransferase, in 15% of pediatric HGGs, a result that was genome-wide significant (FDR = 0.029). Most SETD2 alterations were truncating mutations. Sequencing the gene in this cohort and another validation cohort (123 gliomas from all ages and grades) showed SETD2 mutations to be specific to high-grade tumors affecting 15% of pediatric HGGs (11/73) and 8% of adult HGGs (5/65) while no SETD2 mutations were identified in low-grade diffuse gliomas (0/45). Furthermore, SETD2 mutations were mutually exclusive with H3F3A mutations in HGGs (P = 0.0492) while they partly overlapped with IDH1 mutations (4/14), and SETD2-mutant tumors were found exclusively in the cerebral hemispheres (P = 0.0055). SETD2 is the only H3K36 trimethyltransferase in humans, and SETD2-mutant tumors showed a substantial decrease in H3K36me3 levels (P < 0.001), indicating that the mutations are loss-of-function. These data suggest that loss-of-function SETD2 mutations occur in older children and young adults and are specific to HGG of the cerebral cortex, similar to the H3.3 G34R/V and IDH mutations. Taken together, our results suggest that mutations disrupting the histone code at H3K36, including H3.3 G34R/V, IDH1 and/or SETD2 mutations, are central to the genesis of hemispheric HGGs in older children and young adults.


Subject(s)
Brain Neoplasms/genetics , Glioma/genetics , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Mutation/genetics , Adolescent , Adult , Age Factors , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Case-Control Studies , Child , Cohort Studies , Exome , Glioma/metabolism , Glioma/pathology , Histone Methyltransferases , Humans , Infant , Methylation , Middle Aged , Neoplasm Grading , Young Adult
10.
Acta Neuropathol ; 124(5): 615-25, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22886134

ABSTRACT

Gliomas are the most common primary brain tumors in children and adults. We recently identified frequent alterations in chromatin remodelling pathways including recurrent mutations in H3F3A and mutations in ATRX (α-thalassemia/mental-retardation-syndrome-X-linked) in pediatric and young adult glioblastoma (GBM, WHO grade IV astrocytoma). H3F3A mutations were specific to pediatric high-grade gliomas and identified in only 3.4 % of adult GBM. Using sequencing and/or immunohistochemical analyses, we investigated ATRX alterations (mutation/loss of expression) and their association with TP53 and IDH1 or IDH2 mutations in 140 adult WHO grade II, III and IV gliomas, 17 pediatric WHO grade II and III astrocytomas and 34 pilocytic astrocytomas. In adults, ATRX aberrations were detected in 33 % of grade II and 46 % of grade III gliomas, as well as in 80 % of secondary and 7 % of primary GBMs. They were absent in the 17 grade II and III astrocytomas in children, and the 34 pilocytic astrocytomas. ATRX alterations closely overlapped with mutations in IDH1/2 (p < 0.0001) and TP53 (p < 0.0001) in samples across all WHO grades. They were prevalent in astrocytomas and oligoastrocytomas, but were absent in oligodendrogliomas (p < 0.0001). No significant association of ATRX mutation/loss of expression and alternative lengthening of telomeres was identified in our cohort. In summary, our data show that ATRX alterations are frequent in adult diffuse gliomas and are specific to astrocytic tumors carrying IDH1/2 and TP53 mutations. Combined alteration of these genes may contribute to drive the neoplastic growth in a major subset of diffuse astrocytomas in adults.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , DNA Helicases/genetics , Gene Expression Regulation, Neoplastic/genetics , Mutation/genetics , Nuclear Proteins/genetics , Tumor Suppressor Protein p53/genetics , Adult , Aged , Astrocytoma/classification , Astrocytoma/pathology , Brain Neoplasms/classification , Brain Neoplasms/pathology , Female , Humans , Isocitrate Dehydrogenase/genetics , Male , Middle Aged , Mutation Rate , RNA, Messenger/metabolism , X-linked Nuclear Protein , Young Adult
11.
Acta Neuropathol ; 124(3): 439-47, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22661320

ABSTRACT

Pediatric glioblastomas (GBM) including diffuse intrinsic pontine gliomas (DIPG) are devastating brain tumors with no effective therapy. Here, we investigated clinical and biological impacts of histone H3.3 mutations. Forty-two DIPGs were tested for H3.3 mutations. Wild-type versus mutated (K27M-H3.3) subgroups were compared for HIST1H3B, IDH, ATRX and TP53 mutations, copy number alterations and clinical outcome. K27M-H3.3 occurred in 71 %, TP53 mutations in 77 % and ATRX mutations in 9 % of DIPGs. ATRX mutations were more frequent in older children (p < 0.0001). No G34V/R-H3.3, IDH1/2 or H3.1 mutations were identified. K27M-H3.3 DIPGs showed specific copy number changes, including all gains/amplifications of PDGFRA and MYC/PVT1 loci. Notably, all long-term survivors were H3.3 wild type and this group of patients had better overall survival. K27M-H3.3 mutation defines clinically and biologically distinct subgroups and is prevalent in DIPG, which will impact future therapeutic trial design. K27M- and G34V-H3.3 have location-based incidence (brainstem/cortex) and potentially play distinct roles in pediatric GBM pathogenesis. K27M-H3.3 is universally associated with short survival in DIPG, while patients wild-type for H3.3 show improved survival. Based on prognostic and therapeutic implications, our findings argue for H3.3-mutation testing at diagnosis, which should be rapidly integrated into the clinical decision-making algorithm, particularly in atypical DIPG.


Subject(s)
Brain Stem Neoplasms/genetics , Glioma/genetics , Histones/genetics , Pons/pathology , Adolescent , Brain Stem Neoplasms/mortality , Brain Stem Neoplasms/pathology , Child , Child, Preschool , Female , Gene Expression Profiling , Glioma/mortality , Glioma/pathology , Humans , Infant , Male , Mutation , Prognosis , Survival Rate
12.
J Trauma Acute Care Surg ; 89(6): 1225-1232, 2020 12.
Article in English | MEDLINE | ID: mdl-32925577

ABSTRACT

BACKGROUND: Hemothorax is a common sequelae following thoracic trauma and is associated with significant morbidity and mortality. Current guidelines recommend all traumatic hemothoraces be considered for drainage with tube thoracostomy (TT), regardless of size. With increasing use of computed tomography, smaller hemothoraces not seen on x-ray (defined as an occult hemothorax) are frequently detected. METHODS: This systematic review was performed to gather data on patients with occult hemothorax managed with TT or without TT (termed expectant management [EM]). MEDLINE, EMBASE, and Cochrane databases from inception to October 2019 were searched for relevant articles. The primary outcome was rates of failure of expectant (conservative) management. Secondary outcomes of interest included predictors of TT insertion, predictors of failure of EM, and morbidity and mortality in patients with occult hemothorax. RESULTS: We screened 1,329 abstracts from which 6 articles reporting 1,405 patients with occult hemothorax were included. Of these patients, 601 (43.68%) were managed initially with TT, and 802 (56.32%) were managed expectantly. Of the 802 patients managed expectantly, 212 failed conservative management and underwent TT insertion (23.1% pooled failure rate estimate [95% confidence interval, 17.1-29.1%]). The presence of concomitant pneumothorax predicted upfront TT insertion. Of the patients who failed EM, the need for mechanical ventilation and the presence of a large hemothorax predicted failure. Mortality was similar in both groups. CONCLUSIONS: Conservative treatment of occult hemothorax fails in 23.1% of patients. The presence of hemothorax greater than 300 mL and the need for mechanical ventilation predicted failure of conservative treatment and the need for TT. There was no difference in mortality between EM and TT cohorts. These data suggest that it may be possible to safely observe patients with occult hemothoraces less than 300 mL (1.5 cm pleural stripe) secondary to blunt trauma without upfront TT insertion. LEVEL OF EVIDENCE: Systematic review and meta-analysis, level III.


Subject(s)
Hemothorax/therapy , Pneumothorax/therapy , Thoracic Injuries/complications , Thoracostomy/methods , Chest Tubes , Disease Management , Drainage , Hemothorax/diagnosis , Humans , Pneumothorax/diagnosis , Tomography, X-Ray Computed
13.
Trauma Surg Acute Care Open ; 5(1): e000568, 2020.
Article in English | MEDLINE | ID: mdl-33409372

ABSTRACT

BACKGROUND: Trauma is a cause of significant morbidity and mortality globally, and patients with major trauma require specialized settings for multidisciplinary care. We sought to enumerate the variability of costs of caring for patients at a Canadian level 1 trauma center. METHODS: A retrospective analysis of all adult patients admitted to The Ottawa Hospital trauma service between June 2013 and June 2018 was conducted. Hospital costs and clinical data were collected. Descriptive statistics and multivariable regression analysis using generalized linear model were performed to assess cost variation with patient characteristics. Quintile-based analyses were used to characterize patients in different cost categories. Hospital costs were reported in 2018 Canadian dollars. RESULTS: A total of 2381 admissions were identified in the 5-year cohort. The mean age of patients was 50.2 years, the mean Injury Severity Score (ISS) was 18.7, the mean Charlson Comorbidity Index (CCI) score was 0.35, and the median total cost was $10 048.54. ISS and CCI score were associated with higher costs (ISS >15; p<0.0001). The most expensive mechanisms of injury (MOIs) were those involving heavy machinery (median total cost $24 074.38), pedestrians involved in road traffic collisions ($20 965.45), patients in motor vehicle collisions ($17 621.01) and motorcycle collisions ($16 220.89), and acts of self-injury ($13 903.69). Patients who experienced in-hospital adverse events were associated with higher costs (p<0.0001). Our multivariable regression analysis showed variation in costs related to male gender, penetrating/violent MOI, ISS, adverse hospital events, CCI score, urgent admission status, hospital 1-year mortality risk score, and alternate level of care designation (p<0.05). Quintile-based analyses demonstrated clinically significant differences between the highest and lowest cost groups. DISCUSSION: Major trauma was associated with high hospital costs. Modifiable and non-modifiable patient factors were shown to correlate with differing total hospital costs. These findings can aid in the development of funding strategies and resource allocation for this complex patient population. LEVEL OF EVIDENCE: Level III evidence for economic and value-based evaluations.

14.
Nat Genet ; 49(5): 780-788, 2017 May.
Article in English | MEDLINE | ID: mdl-28394352

ABSTRACT

Spatial heterogeneity of transcriptional and genetic markers between physically isolated biopsies of a single tumor poses major barriers to the identification of biomarkers and the development of targeted therapies that will be effective against the entire tumor. We analyzed the spatial heterogeneity of multiregional biopsies from 35 patients, using a combination of transcriptomic and genomic profiles. Medulloblastomas (MBs), but not high-grade gliomas (HGGs), demonstrated spatially homogeneous transcriptomes, which allowed for accurate subgrouping of tumors from a single biopsy. Conversely, somatic mutations that affect genes suitable for targeted therapeutics demonstrated high levels of spatial heterogeneity in MB, malignant glioma, and renal cell carcinoma (RCC). Actionable targets found in a single MB biopsy were seldom clonal across the entire tumor, which brings the efficacy of monotherapies against a single target into question. Clinical trials of targeted therapies for MB should first ensure the spatially ubiquitous nature of the target mutation.


Subject(s)
Cerebellar Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Medulloblastoma/genetics , Transcriptome , Adult , Aged , Aged, 80 and over , Cerebellar Neoplasms/pathology , Child , Child, Preschool , Cluster Analysis , DNA Copy Number Variations , Female , Gene Expression Profiling/methods , Genetic Heterogeneity , Genome-Wide Association Study , Humans , INDEL Mutation , Male , Medulloblastoma/pathology , Middle Aged , Mutation , Polymorphism, Single Nucleotide , Principal Component Analysis , Reverse Transcriptase Polymerase Chain Reaction
15.
Crit Rev Oncog ; 20(3-4): 271-99, 2015.
Article in English | MEDLINE | ID: mdl-26349420

ABSTRACT

Primary malignant brain tumors remain a disproportionate cause of morbidity and mortality in humans. A number of studies exploring the cancer genome of brain tumors across ages using integrated genetics and epigenetics and next-generation sequencing technologies have recently emerged. This has led to considerable advances in the understanding of the basic biology and pathogenesis of brain tumors, including the most malignant and common variants in children: gliomas and medulloblastoma. Notably, studies of pediatric brain tumors have identified unexpected oncogenic pathways implicated in tumorigenesis. These range from a single pathway/molecule defect such as abnormalities of the mitogen-activated protein kinase pathway, considered to be a hallmark of pilocytic astrocytomas, to alterations in the epigenome as a critical component altered in many subgroups of high-grade brain tumors. Importantly, the type, timing, and spatial clustering of these molecular alterations provide a better understanding of the pathogenesis of the respective brain tumor they target and critical markers for therapy that will help refine pathological grading. We summarize these novel findings in pediatric brain tumors, which also are put in the context of the evolving notion of molecular pathology, now a mandated tool for proper classification and therapy assignment in the clinical setting.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/therapy , Epigenomics/trends , Genomics/trends , Animals , Brain Neoplasms/diagnosis , Child , Epigenomics/methods , Genomics/methods , Glioma/diagnosis , Glioma/genetics , Glioma/therapy , Humans , Neural Stem Cells/pathology
16.
Oncotarget ; 6(31): 31844-56, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26378811

ABSTRACT

Pilocytic astrocytoma (PA) is the most common brain tumor in children but is rare in adults, and hence poorly studied in this age group. We investigated 222 PA and report increased aneuploidy in older patients. Aneuploid genomes were identified in 45% of adult compared with 17% of pediatric PA. Gains were non-random, favoring chromosomes 5, 7, 6 and 11 in order of frequency, and preferentially affecting non-cerebellar PA and tumors with BRAF V600E mutations and not with KIAA1549-BRAF fusions or FGFR1 mutations. Aneuploid PA differentially expressed genes involved in CNS development, the unfolded protein response, and regulators of genomic stability and the cell cycle (MDM2, PLK2),whose correlated programs were overexpressed specifically in aneuploid PA compared to other glial tumors. Thus, convergence of pathways affecting the cell cycle and genomic stability may favor aneuploidy in PA, possibly representing an additional molecular driver in older patients with this brain tumor.


Subject(s)
Aneuploidy , Astrocytoma/classification , Biomarkers, Tumor/genetics , Brain Neoplasms/classification , Adult , Age Factors , Astrocytoma/genetics , Astrocytoma/pathology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Child , Cohort Studies , Female , Gene Expression Profiling , Humans , Male , Mutation/genetics , Neoplasm Staging , Prognosis , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-mdm2/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptor, Fibroblast Growth Factor, Type 1/genetics , Reverse Transcriptase Polymerase Chain Reaction , Young Adult
17.
Nat Genet ; 46(1): 39-44, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24316981

ABSTRACT

Embryonal tumors with multilayered rosettes (ETMRs) are rare, deadly pediatric brain tumors characterized by high-level amplification of the microRNA cluster C19MC. We performed integrated genetic and epigenetic analyses of 12 ETMR samples and identified, in all cases, C19MC fusions to TTYH1 driving expression of the microRNAs. ETMR tumors, cell lines and xenografts showed a specific DNA methylation pattern distinct from those of other tumors and normal tissues. We detected extreme overexpression of a previously uncharacterized isoform of DNMT3B originating at an alternative promoter that is active only in the first weeks of neural tube development. Transcriptional and immunohistochemical analyses suggest that C19MC-dependent DNMT3B deregulation is mediated by RBL2, a known repressor of DNMT3B. Transfection with individual C19MC microRNAs resulted in DNMT3B upregulation and RBL2 downregulation in cultured cells. Our data suggest a potential oncogenic re-engagement of an early developmental program in ETMR via epigenetic alteration mediated by an embryonic, brain-specific DNMT3B isoform.


Subject(s)
Brain Neoplasms/genetics , Chromosomes, Human, Pair 19 , DNA (Cytosine-5-)-Methyltransferases/genetics , Membrane Proteins/genetics , MicroRNAs/genetics , Neoplasms, Germ Cell and Embryonal/genetics , Child, Preschool , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Gene Expression Regulation, Neoplastic , Gene Fusion , Humans , Male , Protein Isoforms , Retinoblastoma-Like Protein p130/genetics , Xenograft Model Antitumor Assays , DNA Methyltransferase 3B
18.
J Clin Oncol ; 32(9): 886-96, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24493713

ABSTRACT

PURPOSE: Medulloblastoma comprises four distinct molecular subgroups: WNT, SHH, Group 3, and Group 4. Current medulloblastoma protocols stratify patients based on clinical features: patient age, metastatic stage, extent of resection, and histologic variant. Stark prognostic and genetic differences among the four subgroups suggest that subgroup-specific molecular biomarkers could improve patient prognostication. PATIENTS AND METHODS: Molecular biomarkers were identified from a discovery set of 673 medulloblastomas from 43 cities around the world. Combined risk stratification models were designed based on clinical and cytogenetic biomarkers identified by multivariable Cox proportional hazards analyses. Identified biomarkers were tested using fluorescent in situ hybridization (FISH) on a nonoverlapping medulloblastoma tissue microarray (n = 453), with subsequent validation of the risk stratification models. RESULTS: Subgroup information improves the predictive accuracy of a multivariable survival model compared with clinical biomarkers alone. Most previously published cytogenetic biomarkers are only prognostic within a single medulloblastoma subgroup. Profiling six FISH biomarkers (GLI2, MYC, chromosome 11 [chr11], chr14, 17p, and 17q) on formalin-fixed paraffin-embedded tissues, we can reliably and reproducibly identify very low-risk and very high-risk patients within SHH, Group 3, and Group 4 medulloblastomas. CONCLUSION: Combining subgroup and cytogenetic biomarkers with established clinical biomarkers substantially improves patient prognostication, even in the context of heterogeneous clinical therapies. The prognostic significance of most molecular biomarkers is restricted to a specific subgroup. We have identified a small panel of cytogenetic biomarkers that reliably identifies very high-risk and very low-risk groups of patients, making it an excellent tool for selecting patients for therapy intensification and therapy de-escalation in future clinical trials.


Subject(s)
Biomarkers, Tumor/genetics , Hedgehog Proteins , Medulloblastoma/genetics , Wnt Proteins , Adolescent , Child , Child, Preschool , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 14 , Cytogenetics , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/genetics , Humans , In Situ Hybridization, Fluorescence , Infant , Kruppel-Like Transcription Factors/genetics , Male , Medulloblastoma/mortality , Medulloblastoma/pathology , Medulloblastoma/therapy , Nuclear Proteins/genetics , Predictive Value of Tests , Prognosis , Proportional Hazards Models , Proto-Oncogene Proteins c-myc/genetics , Reproducibility of Results , Risk Assessment , Risk Factors , Tissue Array Analysis , Wnt Proteins/genetics , Young Adult , Zinc Finger Protein Gli2
19.
Nat Genet ; 46(5): 462-6, 2014 May.
Article in English | MEDLINE | ID: mdl-24705250

ABSTRACT

Pediatric midline high-grade astrocytomas (mHGAs) are incurable with few treatment targets identified. Most tumors harbor mutations encoding p.Lys27Met in histone H3 variants. In 40 treatment-naive mHGAs, 39 analyzed by whole-exome sequencing, we find additional somatic mutations specific to tumor location. Gain-of-function mutations in ACVR1 occur in tumors of the pons in conjunction with histone H3.1 p.Lys27Met substitution, whereas FGFR1 mutations or fusions occur in thalamic tumors associated with histone H3.3 p.Lys27Met substitution. Hyperactivation of the bone morphogenetic protein (BMP)-ACVR1 developmental pathway in mHGAs harboring ACVR1 mutations led to increased levels of phosphorylated SMAD1, SMAD5 and SMAD8 and upregulation of BMP downstream early-response genes in tumor cells. Global DNA methylation profiles were significantly associated with the p.Lys27Met alteration, regardless of the mutant histone H3 variant and irrespective of tumor location, supporting the role of this substitution in driving the epigenetic phenotype. This work considerably expands the number of potential treatment targets and further justifies pretreatment biopsy in pediatric mHGA as a means to orient therapeutic efforts in this disease.


Subject(s)
Activin Receptors, Type I/genetics , Astrocytoma/genetics , Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , Genome, Human/genetics , Mutation/genetics , Animals , Base Sequence , Bone Morphogenetic Proteins/metabolism , Child , DNA Copy Number Variations/genetics , DNA Methylation/genetics , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Molecular Sequence Data , Sequence Analysis, DNA , Smad Proteins/metabolism
20.
Brain Pathol ; 23(2): 210-6, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23432647

ABSTRACT

Primary brain tumors occur in 8 out of 100 000 people and are the leading cause of cancer-related death in children. Among brain tumors, high-grade astrocytomas (HGAs) including glioblastoma multiforme (GBM) are aggressive and are lethal human cancers. Despite decades of concerted therapeutic efforts, HGAs remain essentially incurable in adults and children. Recent discoveries have revolutionized our understanding of these tumors in children and young adults. Recurrent somatic driver mutations in the tail of histone 3 variant 3 (H3.3), leading to amino acid substitutions at key residues, namely lysine (K) 27 (K27M) and glycine 34 (G34R/G34V), were identified as a new molecular mechanism in pediatric GBM. These mutations represent the pediatric counterpart of the recurrent mutations in isocitrate dehydrogenases (IDH) identified in young adult gliomas and provide a much-needed new pathway that can be targeted for therapeutic development. This review will provide an overview of the potential role of these mutations in altering chromatin structure and affecting specific molecular pathways ultimately leading to gliomagenesis. The distinct changes in chromatin structure and the specific downstream events induced by each mutation need characterizing independently if progress is to be made in tackling this devastating cancer.


Subject(s)
Brain Neoplasms/genetics , Chromatin Assembly and Disassembly , Glioblastoma/genetics , Histones/genetics , Adolescent , Brain Neoplasms/pathology , Child , Glioblastoma/pathology , Humans , Mutation , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL