Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 14(1): 6669, 2023 10 20.
Article in English | MEDLINE | ID: mdl-37863903

ABSTRACT

Atypical teratoid rhabdoid tumors (ATRT) are divided into MYC, TYR and SHH subgroups, suggesting diverse lineages of origin. Here, we investigate the imaging of human ATRT at diagnosis and the precise anatomic origin of brain tumors in the Rosa26-CreERT2::Smarcb1flox/flox model. This cross-species analysis points to an extra-cerebral origin for MYC tumors. Additionally, we clearly distinguish SHH ATRT emerging from the cerebellar anterior lobe (CAL) from those emerging from the basal ganglia (BG) and intra-ventricular (IV) regions. Molecular characteristics point to the midbrain-hindbrain boundary as the origin of CAL SHH ATRT, and to the ganglionic eminence as the origin of BG/IV SHH ATRT. Single-cell RNA sequencing on SHH ATRT supports these hypotheses. Trajectory analyses suggest that SMARCB1 loss induces a de-differentiation process mediated by repressors of the neuronal program such as REST, ID and the NOTCH pathway.


Subject(s)
Brain Neoplasms , Rhabdoid Tumor , Teratoma , Humans , Rhabdoid Tumor/genetics , Multiomics , SMARCB1 Protein/genetics , Transcription Factors/genetics , Brain Neoplasms/genetics , Diagnostic Imaging , Teratoma/pathology , Hedgehog Proteins/genetics
2.
J Neuropathol Exp Neurol ; 77(3): 207-215, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29361006

ABSTRACT

Pediatric chordomas are rare malignant neoplasms, and few data are available for optimizing therapeutic strategies and outcome. This study aimed at evaluating how best to manage them and to identify prognostic factors. This multicentric retrospective study included 40 children diagnosed with chordomas between 1966 and 2012. Clinical, radiological, and histopathological data, treatment modalities, and outcomes were reviewed. The median age was 12 years old. Most chordomas were histologically classical forms (45.5%) and were mostly located at the skull base (72.5%). The overall survival (OS) was 66.6% and 58.6%, and progression-free survival (PFS) was 55.7% and 52% at 5 and 10 years, respectively. Total resection was correlated with a better outcome (p = 0.04 for OS and PFS, log-rank). A histopathological/immunohistochemical grading system recently crafted for adults was applied. In a multivariate analysis, it significantly correlated with outcome (PFS and OS, p = 0.004), and the loss of BAF47 immunoexpression appeared to be a significant independent prognostic factor (PFS, p = 0.033). We also identified clinical and histopathological parameters that correlated with prognosis. A new grading system combined with the quality of surgical resection could help classify patients to postpone radiotherapy in case of low risk. Targeted therapy and reirradiation at recurrence may be considered as potential therapeutic strategies.


Subject(s)
Brain Neoplasms/diagnosis , Brain Neoplasms/therapy , Chordoma/diagnosis , Chordoma/therapy , Adolescent , Child , Child, Preschool , Female , Fetal Proteins/metabolism , Follow-Up Studies , Humans , Ki-67 Antigen/metabolism , Male , Prognosis , Radiotherapy , Retrospective Studies , SMARCB1 Protein/metabolism , STAT3 Transcription Factor/metabolism , T-Box Domain Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Vascular Endothelial Growth Factor A/metabolism
3.
Cell Rep ; 21(7): 1737-1745, 2017 Nov 14.
Article in English | MEDLINE | ID: mdl-29141209

ABSTRACT

Rhabdoid tumors (RTs) are aggressive tumors of early childhood characterized by SMARCB1 inactivation. Their poor prognosis highlights an urgent need to develop new therapies. Here, we performed a high-throughput screening of approved drugs and identified broad inhibitors of tyrosine kinase receptors (RTKs), including pazopanib, and the potassium channel inhibitor clofilium tosylate (CfT), as SMARCB1-dependent candidates. Pazopanib targets were identified as PDGFRα/ß and FGFR2, which were the most highly expressed RTKs in a set of primary tumors. Combined genetic inhibition of both these RTKs only partially recapitulated the effect of pazopanib, emphasizing the requirement for broad inhibition. CfT perturbed protein metabolism and endoplasmic reticulum stress and, in combination with pazopanib, induced apoptosis of RT cells in vitro. In vivo, reduction of tumor growth by pazopanib was enhanced in combination with CfT, matching the efficiency of conventional chemotherapy. These results strongly support testing pazopanib/CfT combination therapy in future clinical trials for RTs.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery/methods , High-Throughput Screening Assays/methods , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Quaternary Ammonium Compounds/pharmacology , Rhabdoid Tumor/metabolism , Sulfonamides/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Endoplasmic Reticulum Stress/drug effects , Humans , Indazoles , Receptor, Platelet-Derived Growth Factor alpha/antagonists & inhibitors , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , SMARCB1 Protein/metabolism
4.
Oncotarget ; 8(21): 34245-34257, 2017 May 23.
Article in English | MEDLINE | ID: mdl-28427232

ABSTRACT

Extra-cranial rhabdoid tumors (RT) are highly aggressive malignancies of infancy, characterized by undifferentiated histological features and loss of SMARCB1 expression. The diagnosis is all the more challenging that other poorly differentiated cancers lose SMARCB1 expression, such as epithelioid sarcomas (ES), renal medullary carcinomas (RMC) or undifferentiated chordomas (UC). Moreover, late cases occurring in adults are now increasingly reported, raising the question of differential diagnoses and emphasizing nosological issues. To address this issue, we have analyzed the expression profiles of a training set of 32 SMARCB1-deficient tumors (SDT), with ascertained diagnosis of RT (n = 16, all < 5 years of age), ES (n = 8, all > 10 years of age), UC (n = 3) and RMC (n = 5). As compared with other SDT, RT are characterized by an embryonic signature, and up-regulation of key-actors of de novo DNA methylation processes. Using this signature, we then analysed the expression profiling of 37 SDT to infer the appropriate diagnosis. Thirteen adult onset tumors showed strong similarity with pediatric RT, in spite of older age; by exome sequencing, these tumors also showed genomic features indistinguishable from pediatric RT. In contrary, 8 tumors were reclassified within carcinoma, ES or UC categories, while the remaining could not be related to any of those entities. Our results demonstrate that embryonic signature is shared by all RT, whatever the age at diagnosis; they also illustrate that many adult-onset SDT of ambiguous histological diagnosis are clearly different from RT. Finally, our study paves the way for the routine use of expression-based signatures to give accurate diagnosis of SDT.


Subject(s)
Exome Sequencing/methods , Gene Expression Profiling/methods , Rhabdoid Tumor/diagnosis , Rhabdoid Tumor/genetics , SMARCB1 Protein/deficiency , Adolescent , Adult , Age of Onset , Child , Child, Preschool , DNA Methylation , Diagnosis, Differential , Gene Regulatory Networks , Humans , Infant , Male , Young Adult
5.
Eur Urol ; 69(6): 1055-61, 2016 06.
Article in English | MEDLINE | ID: mdl-26433572

ABSTRACT

BACKGROUND: Renal medullary carcinoma (RMC) is a rare and highly aggressive neoplasm that most often occurs in the setting of sickle cell trait or sickle cell disease (SCD). Most patients present with metastatic disease resistant to conventional chemotherapy, and therefore there is an urgent need for molecular insight to propose new therapies. OBJECTIVE: To determine the molecular alterations and oncogenic pathways that drive RMC development. DESIGN, SETTING, AND PARTICIPANTS: A series of five frozen samples of patients with RMC was investigated by means of gene expression profiling, array comparative genomic hybridization, and RNA and whole exome sequencing (WES). OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: RNA and DNA sequencing read data were analyzed to detect gene fusions and somatic mutations. Gene fusions mutations were validated by real-time polymerase chain reaction and fluorescence in situ hybridization. Gene expression profiling was analyzed by unsupervised hierarchical clustering and Gene Set Enrichment Analysis (Broad Institute, Cambridge, MA, USA). RESULTS AND LIMITATIONS: We observed inactivation of the tumor suppressor gene SMARCB1 in all tumors. In all four cases developed in patients with SCD, we identified an original mechanism of interchromosomal balanced translocations that disrupt the SMARCB1 sequence and thus contribute to its inactivation. Gene expression profiling revealed that RMC shares common oncogenic pathways with pediatric malignant rhabdoid tumors, another tumor subtype characterized by SMARCB1 deficiency. CONCLUSIONS: RMCs are characterized by an original mechanism of interchromosomal balanced translocations that disrupt the SMARCB1 sequence. WES reveals that RMCs show no other recurrent genetic alteration and an overall stable genome, underscoring the oncogenic potency of SMARCB1 inactivation. PATIENT SUMMARY: Our comprehensive molecular study supports a pivotal role of the tumor suppressor gene SMARCB1 in the development of renal medullary carcinoma. The use of therapeutic strategies based on the biologic effects of its inactivation should now open new perspectives for this typically lethal malignancy.


Subject(s)
Carcinoma/genetics , Kidney Neoplasms/genetics , SMARCB1 Protein/genetics , Translocation, Genetic , Adolescent , Adult , Anemia, Sickle Cell/complications , Anemia, Sickle Cell/genetics , Calpain/genetics , Carcinogenesis/genetics , Carcinoma/complications , Child , Comparative Genomic Hybridization , DNA-Binding Proteins/genetics , Gene Expression Profiling , Gene Fusion , Humans , Kidney Neoplasms/complications , Nuclear Proteins/genetics , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , RNA, Long Noncoding/genetics , Sequence Analysis, RNA , Trans-Activators , Transcription Factors/genetics , Exome Sequencing
SELECTION OF CITATIONS
SEARCH DETAIL