Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 320
Filtrer
1.
Childs Nerv Syst ; 40(6): 1919-1924, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38459146

RÉSUMÉ

Pediatric fibromyxoid soft tissue tumors may be associated with gene fusions such as YHWAZ::PLAG1, with only three reported cases in the literature. We present the fourth case, a 13-year-old male with a pediatric fibromyxoid brachial plexus tumor with YWHAZ::PLAG1 gene fusion. This is also the first case to be reported in an adolescent, in the brachial plexus, and in the Philippines. The patient presented with a 10-year history of a slowly growing left supraclavicular mass and a 1-year history of intermittent dysesthesia in the left upper extremity. Neurologic examination was unremarkable. Imaging revealed a large left supraclavicular lesion with intrathoracic extension. Surgical excision was performed, and histopathology revealed a fibromyxoid tumor with YWHAZ::PLAG1 gene fusion. Although previous examples of this gene fusion pointed toward lipoblastoma as their primary pathology, our tumor does not completely fulfill the current diagnostic criteria for a lipoblastoma and may represent an intermediate form of the disease. Our case is unique not only because it is the first reported adolescent patient harboring such a lesion but also because of the relatively lengthy natural history exhibited by the tumor prior to its resection. This provided us with valuable information about its behavior, which suggests a more indolent growth pattern. This case also highlights the clinical importance of molecular testing of tumors, where recognition of disease entities can assist clinicians in deciding and advocating for the proper management.


Sujet(s)
Plexus brachial , Humains , Mâle , Adolescent , Plexus brachial/chirurgie , Fusion de gènes/génétique , Protéines 14-3-3/génétique , Fibrome/génétique , Fibrome/chirurgie , Tumeurs du système nerveux périphérique/génétique , Tumeurs du système nerveux périphérique/chirurgie , Tumeurs du système nerveux périphérique/anatomopathologie , Protéines de liaison à l'ADN/génétique , Tumeurs des tissus mous/génétique , Tumeurs des tissus mous/chirurgie , Tumeurs des tissus mous/anatomopathologie
2.
Virchows Arch ; 484(5): 859-864, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38260988

RÉSUMÉ

Gastroblastoma is a rare gastric biphasic tumor composed of mesenchymal and epithelial elements in variable proportions. These tumors usually arise in the gastric antrum of children and young adults and are reported to harbor a recurrent MALAT1::GLI1 fusion. Herein we report a case of gastroblastoma in a 19-year-old male who presented with intermittent epigastric abdominal discomfort. Antrectomy revealed a 5.6-cm multi-lobulated, tan-pink mass with solid and focally cystic areas involving the submucosa, muscularis propria, and subserosa. All tumor cells demonstrated immunoreactivity for GLI-1, CD56, and vimentin; epithelial elements expressed pancytokeratins (AE1/AE3 and Oscar), and mesenchymal cells demonstrated focal positivity for CD10. Next generation sequencing revealed a novel ACTB::GLI1 fusion without evidence of the recurrent MALAT1::GLI1 fusion. Nine months after surgery, the patient is well without evidence of recurrence or metastases. To our knowledge, this is the first case of gastroblastoma harboring this novel ACTB::GLI1 fusion.


Sujet(s)
Tumeurs de l'estomac , Protéine à doigt de zinc GLI1 , Humains , Mâle , Protéine à doigt de zinc GLI1/génétique , Jeune adulte , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/chirurgie , Marqueurs biologiques tumoraux/génétique , Fusion de gènes/génétique , Protéines de fusion oncogènes/génétique , Résultat thérapeutique
3.
Pigment Cell Melanoma Res ; 37(1): 6-14, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-37475109

RÉSUMÉ

We describe the first cases of pediatric melanoma with ALK fusion gene arising within giant congenital melanocytic nevi. Two newborn boys presented with large pigmented nodular plaques and numerous smaller satellite nevi. Additional expansile nodules developed within both nevi and invasive melanomas were diagnosed before 10 months of age in both boys. Oncogenic driver mutations in NRAS and BRAF were absent in both cases. Instead, oncogenic ZEB2::ALK fusion genes were identified in both the nevus and melanoma developing within the nevus. In both cases, tumors were noted by ultrasound in utero, demonstrated significant nodularity at birth, and progressed to melanoma in the first year of life suggesting that congenital nevi with ALK fusion genes may behave more aggressively than those with other mutations. As ALK kinase inhibitors are effective against a range of tumors with similar ALK fusion kinases, identifying ALK fusion genes in congenital melanocytic nevi may provide an opportunity for targeted therapy.


Sujet(s)
Mélanome , Naevus à cellules épithelioïdes et fusiformes , Naevus pigmentaire , Tumeurs cutanées , Enfant , Humains , Nourrisson , Nouveau-né , Mâle , Kinase du lymphome anaplasique/génétique , Fusion de gènes/génétique , Mélanome/génétique , Mélanome/anatomopathologie , Naevus pigmentaire/anatomopathologie , Tumeurs cutanées/génétique , Tumeurs cutanées/anatomopathologie
4.
Ann Clin Lab Sci ; 53(5): 800-805, 2023 Sep.
Article de Anglais | MEDLINE | ID: mdl-37945013

RÉSUMÉ

Secretory carcinoma (SC), also known as mammary analogue secretory carcinoma (MASC), is a rare salivary gland neoplasm with distinctive morphology that harbors a diagnostic ETV6 gene rearrangement. MASC was first described as a type of salivary gland neoplasm in 2010 and resembles breast secretory carcinoma. It is often mistaken for other neoplasms. It usually acts as an indolent tumor but can occasionally behave in an aggressive manner. We present a rare case of a patient with an aggressive SC/MASC of maxillary gingivobuccal sulcus with microcystic, solid and papillary patterns that showed ETV6 gene rearrangement by fluorescence in situ hybridization. Next-generation sequencing revealed t(12;15)(p13;q25) ETV6-NTRK3 translocation. Because SC/MASCs harbor the ETV6-NTRK3 translocation, molecular studies and immunostains are crucial to confirm the diagnosis and direct therapy.


Sujet(s)
Carcinomes , Carcinome sécrétoire pseudomammaire , Tumeurs des glandes salivaires , Humains , Hybridation fluorescente in situ , Gencive/anatomopathologie , Métastase lymphatique , Protéines de fusion oncogènes/génétique , Marqueurs biologiques tumoraux/génétique , Carcinomes/composition chimique , Carcinome sécrétoire pseudomammaire/génétique , Translocation génétique/génétique , Tumeurs des glandes salivaires/génétique , Tumeurs des glandes salivaires/anatomopathologie , Fusion de gènes/génétique
5.
Cells ; 12(8)2023 04 11.
Article de Anglais | MEDLINE | ID: mdl-37190044

RÉSUMÉ

The ability to identify the broadest range of targetable gene fusions is crucial to facilitate personalized therapy selection for advanced lung adenocarcinoma (LuADs) patients harboring targetable receptor tyrosine kinase (RTK) genomic alterations. In order to evaluate the most effective testing approach for LuAD targetable gene fusion detection, we analyzed 210 NSCLC selected clinical samples, comparing in situ (Fluorescence In Situ Hybridization, FISH, and ImmunoHistoChemistry, IHC) and molecular (targeted RNA Next-Generation Sequencing, NGS, and RealTime-PCR, RT-PCR) approaches. The overall concordance among these methods was high (>90%), and targeted RNA NGS was confirmed to be the most efficient technique for gene fusion identification in clinical practice, allowing the simultaneous analysis of a large set of genomic rearrangements at the RNA level. However, we observed that FISH was useful to detect targetable fusions in those samples with inadequate tissue material for molecular testing as well as in those few cases whose fusions were not identified by the RNA NGS panel. We conclude that the targeted RNA NGS analysis of LuADs allows accurate RTK fusion detection; nevertheless, standard methods such as FISH should not be dismissed, as they can crucially contribute to the completion of the molecular characterization of LuADs and, most importantly, the identification of patients as candidates for targeted therapies.


Sujet(s)
Adénocarcinome pulmonaire , Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Humains , Tumeurs du poumon/anatomopathologie , Kinase du lymphome anaplasique/génétique , Hybridation fluorescente in situ/méthodes , Carcinome pulmonaire non à petites cellules/anatomopathologie , Récepteurs à activité tyrosine kinase/génétique , ARN/usage thérapeutique , Fusion de gènes/génétique
7.
Pathobiology ; 89(6): 393-406, 2022.
Article de Anglais | MEDLINE | ID: mdl-35350025

RÉSUMÉ

INTRODUCTION: The neurotrophic tropomyosin-related kinase (NTRK) genes encode the tropomyosin receptor kinases (TRKs). Patients with solid tumors harboring an oncogenic NTRK fusion are eligible for treatment with TRK inhibitors. NTRK fusion is often associated with TRK overexpression. Pan-TRK immunohistochemistry (IHC) is used to screen for NTRK fusions, but immunoreactivity patterns are poorly defined. METHODS: Data on pan-TRK immunoreactivity patterns in 2,669 solid tumors (comprising carcinomas, sarcomas, and melanocytic lesions) were retrospectively collected by nine laboratories and comprised tumor type, percentage of pan-TRK-positive tumor cells, staining intensity, cytoplasmic, membrane and/or nuclear staining pattern, and the presence or absence of NTRK fusion. RESULTS: Overall, 2,457 tumors (92%) were pan-TRK negative and 212 neoplasms (8%) were pan-TRK positive. Twenty-two pan-TRK-positive tumors (0.8%) harbored an NTRK fusion, representing 10% of all pan-TRK-positive tumors. Cytoplasmic immunoreactivity was most often observed, followed by membrane immunoreactivity. Nuclear pan-TRK positivity was least frequent, but was most often (33%) associated with NTRK fusion. CONCLUSION: Pan-TRK IHC can be used to screen for NTRK fusions, especially in commonly diagnosed solid tumors with low NTRK fusion prevalence. In case of pan-TRK immunoreactivity, regardless of its intensity and tumor cell percentage, subsequent molecular tests should be performed to formally confirm the presence or absence of NTRK fusions.


Sujet(s)
Tumeurs , Récepteurs à activité tyrosine kinase , Humains , Immunohistochimie , Tumeurs/diagnostic , Tumeurs/génétique , Récepteur trkA/génétique , Études rétrospectives , Sarcomes/génétique , Tropomyosine/génétique , Récepteurs à activité tyrosine kinase/génétique , Fusion de gènes/génétique , Dépistage précoce du cancer
8.
BMC Cancer ; 22(1): 165, 2022 Feb 12.
Article de Anglais | MEDLINE | ID: mdl-35151276

RÉSUMÉ

BACKGROUND: In breast cancer (BC), recurrent fusion genes of estrogen receptor alpha (ESR1) and AKAP12, ARMT1 and CCDC170 have been reported. In these gene fusions the ligand binding domain of ESR1 has been replaced by the transactivation domain of the fusion partner constitutively activating the receptor. As a result, these gene fusions can drive tumor growth hormone independently as been shown in preclinical models, but the clinical value of these fusions have not been reported. Here, we studied the prognostic and predictive value of different frequently reported ESR1 fusion transcripts in primary BC. METHODS: We evaluated 732 patients with primary BC (131 ESR1-negative and 601 ESR1-positive cases), including two ER-positive BC patient cohorts: one cohort of 322 patients with advanced disease who received first-line endocrine therapy (ET) (predictive cohort), and a second cohort of 279 patients with lymph node negative disease (LNN) who received no adjuvant systemic treatment (prognostic cohort). Fusion gene transcript levels were measured by reverse transcriptase quantitative PCR. The presence of the different fusion transcripts was associated, in uni- and multivariable Cox regression analysis taking along current clinico-pathological characteristics, to progression free survival (PFS) during first-line endocrine therapy in the predictive cohort, and disease- free survival (DFS) and overall survival (OS) in the prognostic cohort. RESULTS: The ESR1-CCDC170 fusion transcript was present in 27.6% of the ESR1-positive BC subjects and in 2.3% of the ESR1-negative cases. In the predictive cohort, none of the fusion transcripts were associated with response to first-line ET. In the prognostic cohort, the median DFS and OS were respectively 37 and 93 months for patients with an ESR1-CCDC170 exon 8 gene fusion transcript and respectively 91 and 212 months for patients without this fusion transcript. In a multivariable analysis, this ESR1-CCDC170 fusion transcript was an independent prognostic factor for DFS (HR) (95% confidence interval (CI): 1.8 (1.2-2.8), P = 0.005) and OS (HR (95% CI: 1.7 (1.1-2.7), P = 0.023). CONCLUSIONS: Our study shows that in primary BC only ESR1-CCDC170 exon 8 gene fusion transcript carries prognostic value. None of the ESR1 fusion transcripts, which are considered to have constitutive ER activity, was predictive for outcome in BC with advanced disease treated with endocrine treatment.


Sujet(s)
Tumeurs du sein/génétique , Tumeurs du sein/mortalité , Récepteur alpha des oestrogènes/génétique , Fusion de gènes/génétique , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Antinéoplasiques hormonaux/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Études cas-témoins , Survie sans rechute , Femelle , Humains , Adulte d'âge moyen , Valeur prédictive des tests , Pronostic , Modèles des risques proportionnels , Études rétrospectives
9.
Eur J Endocrinol ; 186(4): K5-K8, 2022 Feb 23.
Article de Anglais | MEDLINE | ID: mdl-35118995

RÉSUMÉ

NTRK: rearrangements represent a very rare genomic abnormality among all cancers but can be detected in thyroid cancer with a non-negligible frequency of 2%. Dramatic clinical responses to therapies targeting NTRK chimeric proteins are now well described in the literature. SQSTM1-NTRK1fusions have not yet been described in a full clinical case report. We report a patient with a papillary thyroid carcinoma harboring this unique rearrangement, with an impressive clinical and radiologic response to larotrectinib, a highly specific inhibitor.


Sujet(s)
Pyrazoles/usage thérapeutique , Pyrimidines/usage thérapeutique , Récepteur trkA/génétique , Séquestosome-1/génétique , Cancer papillaire de la thyroïde/imagerie diagnostique , Cancer papillaire de la thyroïde/génétique , Tumeurs de la thyroïde/génétique , Femelle , Fusion de gènes/génétique , Humains , Adulte d'âge moyen , Cancer papillaire de la thyroïde/traitement médicamenteux , Tumeurs de la thyroïde/imagerie diagnostique , Tumeurs de la thyroïde/traitement médicamenteux , Résultat thérapeutique
10.
Pathol Int ; 72(3): 187-192, 2022 Mar.
Article de Anglais | MEDLINE | ID: mdl-35102630

RÉSUMÉ

NTRK fusions represent a new biomarker-defined population that can be treated with TRK inhibitors. Although rare, NTRK fusions are detected across a wide range of solid tumors. Previous reports suggest that NTRK fusions are limited to the secretory subtype of breast cancer. Here we examined NTRK fusions in a large real world next-generation sequencing (NGS) dataset and confirmed secretory versus non-secretory status using H&E images. Of 23 NTRK fusion-positive cases, 11 were classified as secretory, 11 as non-secretory, and one as mixed status. The secretory subtype trended younger, was predominantly estrogen receptor (ER)-, had lower tumor mutational burden, and exhibited lower levels of genomic loss of heterozygosity. The non-secretory subtype was enriched for TP53 mutations. The secretory subtype was enriched for ETV6-NTRK3 fusions in 7 of 11 cases, and the non-secretory subtype had NTRK1 fusions in 7 of 11 cases, each with a different fusion partner. Our data suggests NTRK fusions are present in both secretory and non-secretory subtypes, and that comprehensive genomic profiling should be considered across all clinically advanced breast cancers to identify patients that could receive benefit from TRK inhibitors.


Sujet(s)
Tumeurs du sein/génétique , Carcinomes/diagnostic , Récepteur trkA/génétique , Sujet âgé , Tumeurs du sein/diagnostic , Carcinomes/génétique , Femelle , Fusion de gènes/effets des médicaments et des substances chimiques , Fusion de gènes/génétique , Séquençage nucléotidique à haut débit/méthodes , Séquençage nucléotidique à haut débit/statistiques et données numériques , Humains , Immunohistochimie/méthodes , Immunohistochimie/statistiques et données numériques , Adulte d'âge moyen , Récepteur trkA/effets indésirables , Récepteur trkC/génétique
11.
Cell Rep ; 38(1): 110190, 2022 01 04.
Article de Anglais | MEDLINE | ID: mdl-34986355

RÉSUMÉ

Translocation renal cell carcinoma (tRCC) is a poorly characterized subtype of kidney cancer driven by MiT/TFE gene fusions. Here, we define the landmarks of tRCC through an integrative analysis of 152 patients with tRCC identified across genomic, clinical trial, and retrospective cohorts. Most tRCCs harbor few somatic alterations apart from MiT/TFE fusions and homozygous deletions at chromosome 9p21.3 (19.2% of cases). Transcriptionally, tRCCs display a heightened NRF2-driven antioxidant response that is associated with resistance to targeted therapies. Consistently, we find that outcomes for patients with tRCC treated with vascular endothelial growth factor receptor inhibitors (VEGFR-TKIs) are worse than those treated with immune checkpoint inhibitors (ICI). Using multiparametric immunofluorescence, we find that the tumors are infiltrated with CD8+ T cells, though the T cells harbor an exhaustion immunophenotype distinct from that of clear cell RCC. Our findings comprehensively define the clinical and molecular features of tRCC and may inspire new therapeutic hypotheses.


Sujet(s)
Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines/génétique , Néphrocarcinome/génétique , Tumeurs du rein/génétique , Facteur de transcription associé à la microphtalmie/génétique , Protéines de fusion oncogènes/génétique , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Lymphocytes T CD8+/immunologie , Néphrocarcinome/traitement médicamenteux , Néphrocarcinome/anatomopathologie , Régulation de l'expression des gènes tumoraux , Fusion de gènes/génétique , Humains , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Tumeurs du rein/traitement médicamenteux , Tumeurs du rein/anatomopathologie , Protéines de fusion oncogènes/métabolisme , Inhibiteurs de protéines kinases/usage thérapeutique , Récepteur-1 au facteur croissance endothéliale vasculaire/antagonistes et inhibiteurs
12.
Nucleic Acids Res ; 50(D1): D1221-D1230, 2022 01 07.
Article de Anglais | MEDLINE | ID: mdl-34755868

RÉSUMÉ

A knowledgebase of the systematic functional annotation of fusion genes is critical for understanding genomic breakage context and developing therapeutic strategies. FusionGDB is a unique functional annotation database of human fusion genes and has been widely used for studies with diverse aims. In this study, we report fusion gene annotation updates aided by deep learning (FusionGDB 2.0) available at https://compbio.uth.edu/FusionGDB2/. FusionGDB 2.0 has substantial updates of contents such as up-to-date human fusion genes, fusion gene breakage tendency score with FusionAI deep learning model based on 20 kb DNA sequence around BP, investigation of overlapping between fusion breakpoints with 44 human genomic features across five cellular role's categories, transcribed chimeric sequence and following open reading frame analysis with coding potential based on deep learning approach with Ribo-seq read features, and rigorous investigation of the protein feature retention of individual fusion partner genes in the protein level. Among ∼102k fusion genes, about 15k kept their ORF as In-frames, which is two times compared to the previous version, FusionGDB. FusionGDB 2.0 will be used as the reference knowledgebase of fusion gene annotations. FusionGDB 2.0 provides eight categories of annotations and it will be helpful for diverse human genomic studies.


Sujet(s)
Bases de données génétiques , Fusion de gènes/génétique , Génome humain/génétique , Génomique , Séquence d'acides aminés/génétique , Apprentissage profond , Humains , Bases de connaissances , Annotation de séquence moléculaire
13.
Curr Opin Neurol ; 34(6): 840-847, 2021 12 01.
Article de Anglais | MEDLINE | ID: mdl-34766555

RÉSUMÉ

PURPOSE OF REVIEW: Glioma represents of variety of brain malignancies, the majority of which confer a poor prognosis despite treatment. With the widespread use of next-generation sequencing, gene fusions are being found in greater numbers. Gene fusions in glioma represent an opportunity to deliver targeted therapies to those with limited options for treatment. RECENT FINDINGS: Extensive studies on these gene fusions have shown that they can exhibit distinct phenotypes, such as PTPRZ1-MET fusions in secondary glioblastoma or FGFR3-TACC3 fusions in IDH wildtype gliomas. Responses have been observed with the use of targeted therapies but some have been short lived because of the development of treatment resistance. SUMMARY: Increasing detection of gene fusions in glioma along with basket trials have helped define different fusion phenotypes and paved the way for targeted kinase inhibitor-based therapies. Targeting NTRK fusions has been the most successful fusion-guided therapy to date and evaluating all patients for these fusions may be warranted.


Sujet(s)
Tumeurs du cerveau , Glioblastome , Gliome , Tumeurs du cerveau/génétique , Tumeurs du cerveau/thérapie , Fusion de gènes/génétique , Gliome/génétique , Gliome/thérapie , Séquençage nucléotidique à haut débit , Humains , Protéines associées aux microtubules/génétique , Receptor-Like Protein Tyrosine Phosphatases, Class 5/génétique
14.
Pharmacogenomics ; 22(13): 833-847, 2021 08.
Article de Anglais | MEDLINE | ID: mdl-34525844

RÉSUMÉ

Although gene fusions occur rarely in non-small-cell lung cancer (NSCLC) patients, they represent a relevant target in treatment decision algorithms. To date, immunohistochemistry and fluorescence in situ hybridization are the two principal methods used in clinical trials. However, using these methods in routine clinical practice is often impractical and time consuming because they can only analyze single genes and the quantity of tissue material is often insufficient. Thus, novel technologies, able to test multiple genes in a single run with minimal sample input, are being under investigation. Here, we discuss the utility of next-generation sequencing and nCounter technologies in detecting simultaneous gene fusions in NSCLC patients.


Sujet(s)
Carcinome pulmonaire non à petites cellules/génétique , Fusion de gènes/génétique , Tumeurs du poumon/génétique , Oncologie médicale/méthodes , Oncologie médicale/tendances , Séquençage nucléotidique à haut débit , Humains
15.
J Med Genet ; 58(12): 789-795, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34462289

RÉSUMÉ

Gene fusion, a genomic event that generates a novel gene from two independent genes, has long been known to be implicated in tumourigenesis and cancer progression. It has thus served as a diagnostic and prognostic biomarker in cancer, as well as an ideal therapeutic target in cancer therapy. Gene fusion can arise from chromosomal rearrangement and alternative splicing of transcripts, resulting in deregulation of proto-oncogenes or creation of an oncogenic novel gene. Largely facilitated by next generation sequencing technologies, a plethora of novel gene fusions have been identified in a variety of cancers, which leaves us the challenge of functionally characterising these candidate gene fusions. In this review, we summarise the molecular mechanisms, the oncogenic consequences and the therapeutic implications of verified gene fusions. We also discuss recent studies on gene fusions in both common and rare subtypes of ovarian tumours and how these findings can be translated to cancer therapies to benefit patients carrying these gene fusions.


Sujet(s)
Carcinogenèse/génétique , Fusion de gènes/génétique , Protéines de fusion oncogènes/génétique , Tumeurs de l'ovaire/génétique , Femelle , Analyse de profil d'expression de gènes/méthodes , Régulation de l'expression des gènes tumoraux , Génomique/méthodes , Séquençage nucléotidique à haut débit/méthodes , Humains , Modèles génétiques
16.
Oncogene ; 40(40): 5902-5912, 2021 10.
Article de Anglais | MEDLINE | ID: mdl-34354240

RÉSUMÉ

Leukemia patients bearing t(6;11)(q27;q23) translocations can be divided in two subgroups: those with breakpoints in the major breakpoint cluster region of MLL (introns 9-10; associated mainly with AML M1/4/5), and others with breakpoints in the minor breakpoint cluster region (introns 21-23), associated with T-ALL. We cloned all four of the resulting fusion genes (MLL-AF6, AF6-MLL, exMLL-AF6, AF6-shMLL) and subsequently transfected them to generate stable cell culture models. Their molecular function was tested by inducing gene expression for 48 h in a Doxycycline-dependent fashion. Here, we present our results upon differential gene expression (DGE) that were obtained by the "Massive Analyses of cDNA Ends" (MACE-Seq) technology, an established 3'-end based RNA-Seq method. Our results indicate that the PHD/BD domain, present in the AF6-MLL and the exMLL-AF6 fusion protein, is responsible for chromatin activation in a genome-wide fashion. This led to strong deregulation of transcriptional processes involving protein-coding genes, pseudogenes, non-annotated genes, and RNA genes, e.g., LincRNAs and microRNAs, respectively. While cooperation between the MLL-AF6 and AF6-MLL fusion proteins appears to be required for the above-mentioned effects, exMLL-AF6 is able to cause similar effects on its own. The exMLL-AF6/AF6-shMLL co-expressing cell line displayed the induction of a myeloid-specific and a T-cell specific gene signature, which may explain the T-ALL disease phenotype observed in patients with such breakpoints. This again demonstrated that MLL fusion proteins are instructive and allow to study their pathomolecular mechanisms.


Sujet(s)
Fusion de gènes/génétique , Leucémie aigüe myéloïde/génétique , Translocation génétique/génétique , Humains , Transfection
17.
Genes Chromosomes Cancer ; 60(12): 833-836, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34369017

RÉSUMÉ

Aneurysmal bone cyst is a benign bone neoplasm that most commonly arises from the metaphyses of long bones in the first and second decades of life. Here, we describe a case of an aneurysmal bone cyst that occurred in the distal tibial diaphysis of a 72-year-old female that was concerning for malignancy on imaging, demonstrating cortical breakthrough and soft tissue extension. Histologically, the tumor showed the characteristic morphologic features of aneurysmal bone cyst. Fluorescence in situ hybridization was positive for USP6 rearrangement, and RNA sequencing revealed a USP6 gene fusion with VDR, a novel partner that encodes the vitamin D receptor and that has not been implicated previously in human neoplasia. This case highlights the diagnostic challenges presented by aneurysmal bone cyst in elderly adults, and it expands the genetic spectrum of USP6 rearrangements.


Sujet(s)
Kystes osseux anévrismaux/génétique , Tumeurs osseuses/génétique , Récepteur calcitriol/génétique , Ubiquitin thiolesterase/génétique , Sujet âgé , Kystes osseux anévrismaux/diagnostic , Kystes osseux anévrismaux/imagerie diagnostique , Kystes osseux anévrismaux/anatomopathologie , Tumeurs osseuses/diagnostic , Tumeurs osseuses/imagerie diagnostique , Tumeurs osseuses/anatomopathologie , Femelle , Fusion de gènes/génétique , Réarrangement des gènes/génétique , Humains , Hybridation fluorescente in situ , Régions promotrices (génétique)/génétique , Protéines proto-oncogènes/génétique
18.
Acta Neuropathol Commun ; 9(1): 135, 2021 08 13.
Article de Anglais | MEDLINE | ID: mdl-34389065

RÉSUMÉ

The cIMPACT-NOW Update 7 has replaced the WHO nosology of "ependymoma, RELA fusion positive" by "Supratentorial-ependymoma, C11orf95-fusion positive". This modification reinforces the idea that supratentorial-ependymomas exhibiting fusion that implicates the C11orf95 (now called ZFTA) gene with or without the RELA gene, represent the same histomolecular entity. A hot off the press molecular study has identified distinct clusters of the DNA methylation class of ZFTA fusion-positive tumors. Interestingly, clusters 2 and 4 comprised tumors of different morphologies, with various ZFTA fusions without involvement of RELA. In this paper, we present a detailed series of thirteen cases of non-RELA ZFTA-fused supratentorial tumors with extensive clinical, radiological, histopathological, immunohistochemical, genetic and epigenetic (DNA methylation profiling) characterization. Contrary to the age of onset and MRI aspects similar to RELA fusion-positive EPN, we noted significant histopathological heterogeneity (pleomorphic xanthoastrocytoma-like, astroblastoma-like, ependymoma-like, and even sarcoma-like patterns) in this cohort. Immunophenotypically, these NFκB immunonegative tumors expressed GFAP variably, but EMA constantly and L1CAM frequently. Different gene partners were fused with ZFTA: NCOA1/2, MAML2 and for the first time MN1. These tumors had epigenetic homologies within the DNA methylation class of ependymomas-RELA and were classified as satellite clusters 2 and 4. Cluster 2 (n = 9) corresponded to tumors with classic ependymal histological features (n = 4) but also had astroblastic features (n = 5). Various types of ZFTA fusions were associated with cluster 2, but as in the original report, ZFTA:MAML2 fusion was frequent. Cluster 4 was enriched with sarcoma-like tumors. Moreover, we reported a novel anatomy of three ZFTA:NCOA1/2 fusions with only 1 ZFTA zinc finger domain in the putative fusion protein, whereas all previously reported non-RELA ZFTA fusions have 4 ZFTA zinc fingers. All three cases presented a sarcoma-like morphology. This genotype/phenotype association requires further studies for confirmation. Our series is the first to extensively characterize this new subset of supratentorial ZFTA-fused ependymomas and highlights the usefulness of ZFTA FISH analysis to confirm the existence of a rearrangement without RELA abnormality.


Sujet(s)
Épendymome/génétique , Protéines/génétique , Tumeurs sus-tentorielles/génétique , Adolescent , Adulte , Enfant , Enfant d'âge préscolaire , Méthylation de l'ADN/génétique , Épendymome/classification , Épendymome/métabolisme , Épendymome/anatomopathologie , Femelle , Fusion de gènes/génétique , Génotype , Protéine gliofibrillaire acide/métabolisme , Humains , Nourrisson , Mâle , Facteur de transcription NF-kappa B/métabolisme , Molécule d'adhérence cellulaire neurale L-1/métabolisme , Coactivateur-1 de récepteur nucléaire/génétique , Coactivateur-2 de récepteur nucléaire/génétique , Phénotype , Tumeurs sus-tentorielles/classification , Tumeurs sus-tentorielles/métabolisme , Tumeurs sus-tentorielles/anatomopathologie , Transactivateurs/génétique , Facteur de transcription RelA/génétique , Protéines suppresseurs de tumeurs/génétique , Jeune adulte
19.
Clin. transl. oncol. (Print) ; 23(8): 1529-1541, ago. 2021. ilus, tab
Article de Anglais | IBECS | ID: ibc-222151

RÉSUMÉ

The recent identification of rearrangements of neurotrophic tyrosine receptor kinase (NTRK) genes and the development of specific fusion protein inhibitors, such as larotrectinib and entrectinib, have revolutionised the diagnostic and clinical management of patients presenting with tumours with these alterations. Tumours that harbour NTRK fusions are found in both adults and children; and they are either rare tumours with common NTRK fusions that may be diagnostic, or more prevalent tumours with rare NTRK fusions. To assess currently available evidence on this matter, three key Spanish medical societies (the Spanish Society of Medical Oncology (SEOM), the Spanish Society of Pathological Anatomy (SEAP), and the Spanish Society of Paediatric Haematology and Oncology (SEHOP) have brought together a group of experts to develop a consensus document that includes guidelines on the diagnostic, clinical, and therapeutic aspects of NTRK-fusion tumours. This document also discusses the challenges related to the routine detection of these genetic alterations in a mostly public Health Care System (AU)


Sujet(s)
Humains , Enfant , Adulte , Tumeurs/thérapie , Glycoprotéines/génétique , Thérapie moléculaire ciblée , Tumeurs/génétique , Fusion de gènes/génétique , Fusion oncogène/génétique , Facteurs âges , Benzamides/usage thérapeutique , Séquençage nucléotidique à haut débit , Immunohistochimie , Hybridation fluorescente in situ , Fluorescence , Tumeurs/diagnostic , Sociétés médicales , Consensus , Espagne
20.
Cancer Res ; 81(15): 3971-3984, 2021 08 01.
Article de Anglais | MEDLINE | ID: mdl-34099491

RÉSUMÉ

Gene fusions frequently result from rearrangements in cancer genomes. In many instances, gene fusions play an important role in oncogenesis; in other instances, they are thought to be passenger events. Although regulatory element rearrangements and copy number alterations resulting from these structural variants are known to lead to transcriptional dysregulation across cancers, the extent to which these events result in functional dependencies with an impact on cancer cell survival is variable. Here we used CRISPR-Cas9 dependency screens to evaluate the fitness impact of 3,277 fusions across 645 cell lines from the Cancer Dependency Map. We found that 35% of cell lines harbored either a fusion partner dependency or a collateral dependency on a gene within the same topologically associating domain as a fusion partner. Fusion-associated dependencies revealed numerous novel oncogenic drivers and clinically translatable alterations. Broadly, fusions can result in partner and collateral dependencies that have biological and clinical relevance across cancer types. SIGNIFICANCE: This study provides insights into how fusions contribute to fitness in different cancer contexts beyond partner-gene activation events, identifying partner and collateral dependencies that may have direct implications for clinical care.


Sujet(s)
Survie cellulaire/génétique , Fusion de gènes/génétique , Tumeurs/génétique , Humains
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...