Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Int J Cancer ; 151(11): 2043-2054, 2022 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-35932450

RESUMEN

Immune checkpoint blockade (ICB) has led to durable clinical responses in multiple cancer types. However, biomarkers that identify which patients are most likely to respond to ICB are not well defined. Many putative biomarkers developed from a small number of samples often fail to maintain their predictive status in larger validation cohorts. We show across multiple human malignancies and syngeneic murine tumor models that neither pretreatment T cell receptor (TCR) clonality nor changes in clonality after ICB correlate with response. Dissection of tumor infiltrating lymphocytes pre- and post-ICB by paired single-cell RNA sequencing and single-cell TCR sequencing reveals conserved and distinct transcriptomic features in expanded TCR clonotypes between anti-PD1 responder and nonresponder murine tumor models. Overall, our results indicate a productive anti-tumor response is agnostic of TCR clonal expansion. Further, we used single-cell transcriptomics to develop a CD8+ T cell specific gene signature for a productive anti-tumor response and show the response signature to be associated with overall survival (OS) on nivolumab monotherapy in CheckMate-067, a phase 3 clinical trial in metastatic melanoma. These results highlight the value of leveraging single-cell assays to dissect heterogeneous tumor and immune subsets and define cell-type specific transcriptomic biomarkers of ICB response.


Asunto(s)
Melanoma , Receptor de Muerte Celular Programada 1 , Animales , Linfocitos T CD8-positivos , Humanos , Inhibidores de Puntos de Control Inmunológico , Melanoma/tratamiento farmacológico , Melanoma/genética , Ratones , Nivolumab/farmacología , Nivolumab/uso terapéutico , Receptores de Antígenos de Linfocitos T/genética
2.
Sci Rep ; 12(1): 12298, 2022 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-35853984

RESUMEN

In an effort to identify rare alleles associated with adolescent idiopathic scoliosis (AIS) whole-exome sequencing was performed on a discovery cohort of 73 unrelated patients and 70 age-and sex matched controls, all of French-Canadian ancestry. A collapsing gene burden test was performed to analyze rare protein-altering variants using case-control statistics. Since no single gene achieved statistical significance, targeted exon sequencing was performed for 24 genes with the smallest p values, in an independent replication cohort of unrelated severely affected females with AIS and sex-matched controls (N = 96 each). An excess of rare, potentially protein-altering variants was noted in one particular gene, FAT3, although it did not achieve statistical significance. Independently, we sequenced the exomes of all members of a rare multiplex family of three affected sisters and unaffected parents. All three sisters were compound heterozygous for two rare protein-altering variants in FAT3. The parents were single heterozygotes for each variant. The two variants in the family were also present in our discovery cohort. A second validation step was done, using another independent replication cohort of 258 unrelated AIS patients having reach their skeletal maturity and 143 healthy controls to genotype nine FAT3 gene variants, including the two variants previously identified in the multiplex family: p.L517S (rs139595720) and p.L4544F (rs187159256). Interestingly, two FAT3 variants, rs139595720 (genotype A/G) and rs80293525 (genotype C/T), were enriched in severe scoliosis cases (4.5% and 2.7% respectively) compared to milder cases (1.4% and 0.7%) and healthy controls (1.6% and 0.8%). Our results implicate FAT3 as a new candidate gene in the etiology of AIS.


Asunto(s)
Cadherinas , Factor de Crecimiento Epidérmico , Cifosis , Escoliosis , Adolescente , Alelos , Cadherinas/genética , Factor de Crecimiento Epidérmico/genética , Exoma , Femenino , Predisposición Genética a la Enfermedad , Humanos , Cifosis/genética , Polimorfismo de Nucleótido Simple , Escoliosis/genética
3.
J Immunother Cancer ; 10(3)2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35304405

RESUMEN

BACKGROUND: The phase 3 CheckMate 214 trial demonstrated higher response rates and improved overall survival with nivolumab plus ipilimumab versus sunitinib in first-line therapy for advanced clear-cell renal cell carcinoma (RCC). An unmet need exists to identify patients with RCC who are most likely to benefit from treatment with nivolumab plus ipilimumab. METHODS: In exploratory analyses, pretreatment levels of programmed death ligand 1 were assessed by immunohistochemistry. Genomic and transcriptomic biomarkers (including tumor mutational burden and gene expression signatures) were also investigated. RESULTS: Biomarkers previously associated with benefit from immune checkpoint inhibitor-containing regimens in RCC were not predictive for survival in patients with RCC treated with nivolumab plus ipilimumab. Analysis of gene expression identified an association between an inflammatory response and progression-free survival with nivolumab plus ipilimumab. CONCLUSIONS: The exploratory analyses reveal relationships between molecular biomarkers and provide supportive data on how the inflammation status of the tumor microenvironment may be important for identifying predictive biomarkers of response and survival with combination immunotherapy in patients with RCC. Further validation may help to provide biomarker-driven precision treatment for patients with RCC.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Células Renales/patología , Ensayos Clínicos Fase III como Asunto , Humanos , Ipilimumab/farmacología , Ipilimumab/uso terapéutico , Neoplasias Renales/patología , Nivolumab/farmacología , Nivolumab/uso terapéutico , Sunitinib/uso terapéutico , Microambiente Tumoral
4.
J Immunother Cancer ; 9(3)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33658305

RESUMEN

BACKGROUND: Nivolumab is an immune checkpoint inhibitor targeting the programmed death-1 receptor that improves survival in a subset of patients with clear cell renal cell carcinoma (ccRCC). In contrast to other tumor types that respond to immunotherapy, factors such as programmed death ligand-1 (PD-L1) status and tumor mutational burden show limited predictive utility in ccRCC. To address this gap, we report here the first molecular characterization of nivolumab response using paired index lesions, before and during treatment of metastatic ccRCC. METHODS: We analyzed gene expression and T-cell receptor (TCR) clonality using lesion-paired biopsies provided in the CheckMate 009 trial and integrated the results with their PD-L1/CD4/CD8 status, genomic mutation status and serum cytokine assays. Statistical tests included linear mixed models, logistic regression models, Fisher's exact test, and Kruskal-Wallis rank-sum test. RESULTS: We identified transcripts related to response, both at baseline and on therapy, including several that are amenable to peripheral bioassays or to therapeutic intervention. At both timepoints, response was positively associated with T-cell infiltration but not associated with TCR clonality, and some non-Responders were highly infiltrated. Lower baseline T-cell infiltration correlated with elevated transcription of Wnt/ß-catenin signaling components and hypoxia-regulated genes, including the Treg chemoattractant CCL28. On treatment, analysis of the non-responding patients whose tumors were highly T-cell infiltrated suggests association of the RIG-I-MDA5 pathway in their nivolumab resistance. We also analyzed our data using previous transcriptional classifications of ccRCC and found they concordantly identified a molecular subtype that has enhanced nivolumab response but is sunitinib-resistant. CONCLUSION: Our study describes molecular characteristics of response and resistance to nivolumab in patients with metastatic ccRCC, potentially impacting patient selection and first-line treatment decisions. TRIAL REGISTRATION NUMBER: NCT01358721.


Asunto(s)
Biomarcadores de Tumor , Carcinoma de Células Renales/tratamiento farmacológico , Resistencia a Antineoplásicos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias Renales/tratamiento farmacológico , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Nivolumab/uso terapéutico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Linfocitos T/efectos de los fármacos , Antígeno B7-H1/genética , Biomarcadores de Tumor/sangre , Biomarcadores de Tumor/genética , Antígenos CD4/genética , Antígenos CD8/genética , Carcinoma de Células Renales/sangre , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/inmunología , Citocinas/sangre , Resistencia a Antineoplásicos/genética , Humanos , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Neoplasias Renales/sangre , Neoplasias Renales/genética , Neoplasias Renales/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Mutación , Nivolumab/efectos adversos , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T/inmunología , Linfocitos T/metabolismo , Factores de Tiempo , Resultado del Tratamiento
5.
Commun Med (Lond) ; 1: 56, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35602225

RESUMEN

Background: Tumor mutational burden (TMB) has been proposed as a predictive biomarker of response to immunotherapy. Efforts to standardize TMB scores for use in the clinic and to identify the factors that could impact TMB scores are of high importance. However, the biopsy collection site has not been assessed as a factor that may influence TMB scores. Methods: We examine a real-world cohort comprising 137,771 specimens across 47 tissues in 12 indications profiled by the FoundationOne assay (Foundation Medicine, Cambridge, MA) to assess the prevalence of biopsy sites for each indication and their TMB scores distribution. Results: We observe a wide variety of biopsy sites from which specimens are sent for genomic testing and show that TMB scores differ in a cancer- and tissue-specific manner. For example, brain or adrenal gland specimens from NSCLC patients show higher TMB scores than local lung specimens (mean difference 3.31 mut/Mb; p < 0.01, 3.90 mut/Mb; p < 0.01, respectively), whereas bone specimens show no difference. Conclusions: Our data shed light on the biopsied tissue as a driver of TMB measurement variability in clinical practice.

6.
ESMO Open ; 5(2)2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32312757

RESUMEN

INTRODUCTION: Somatic mutations in STK11 and KEAP1, frequently comutated in non-squamous non-small cell lung cancer (NSQ NSCLC), have been associated with poor response to immune checkpoint blockade (ICB). However, previous reports lack non-ICB controls needed to properly ascertain the predictive nature of those biomarkers. The objective of this study was to evaluate the predictive versus prognostic effect of STK11 or KEAP1 mutations in NSQ NSCLC. METHODS: Patients diagnosed with stage IIIB, IIIC, IVA or IVB NSQ NSCLC from a real-world data cohort from the Flatiron Health Network linked with genetic testing from Foundation Medicine were retrospectively assessed. Real-world, progression-free survival (rwPFS) and overall survival (OS) were calculated from time of initiation of first-line treatment. RESULTS: We analysed clinical and mutational data for 2276 patients including patients treated with anti-programmed death-1 (PD-1)/anti-programmed death ligand 1 (PD-L1) inhibitors at first line (n=574). Mutations in STK11 or KEAP1 were associated with poor outcomes across multiple therapeutic classes and were not specifically associated with poor outcomes in ICB cohorts. There was no observable interaction between STK11 mutations and anti-PD-1/anti-PD-L1 treatment on rwPFS (HR, 1.05; 95% CI 0.76 to 1.44; p=0.785) or OS (HR, 1.13; 95% CI 0.76 to 1.67; p=0.540). Similarly, there was no observable interaction between KEAP1 mutations and treatment on rwPFS (HR, 0.93; 95% CI 0.67 to 1.28; p=0.653) or OS (HR, 0.98; 95% CI 0.66 to 1.45; p=0.913). CONCLUSION: Our results show that STK11-KEAP1 mutations are prognostic, not predictive, biomarkers for anti-PD-1/anti-PD-L1 therapy.


Asunto(s)
Adenocarcinoma/genética , Proteína 1 Asociada A ECH Tipo Kelch/genética , Neoplasias Pulmonares/genética , Proteínas Serina-Treonina Quinasas/genética , Quinasas de la Proteína-Quinasa Activada por el AMP , Adenocarcinoma/mortalidad , Adenocarcinoma/patología , Biomarcadores de Tumor , Estudios de Cohortes , Femenino , Humanos , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Masculino , Mutación , Pronóstico , Análisis de Supervivencia
7.
Nature ; 576(7786): 274-280, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31802000

RESUMEN

Embryonal tumours with multilayered rosettes (ETMRs) are aggressive paediatric embryonal brain tumours with a universally poor prognosis1. Here we collected 193 primary ETMRs and 23 matched relapse samples to investigate the genomic landscape of this distinct tumour type. We found that patients with tumours in which the proposed driver C19MC2-4 was not amplified frequently had germline mutations in DICER1 or other microRNA-related aberrations such as somatic amplification of miR-17-92 (also known as MIR17HG). Whole-genome sequencing revealed that tumours had an overall low recurrence of single-nucleotide variants (SNVs), but showed prevalent genomic instability caused by widespread occurrence of R-loop structures. We show that R-loop-associated chromosomal instability can be induced by the loss of DICER1 function. Comparison of primary tumours and matched relapse samples showed a strong conservation of structural variants, but low conservation of SNVs. Moreover, many newly acquired SNVs are associated with a mutational signature related to cisplatin treatment. Finally, we show that targeting R-loops with topoisomerase and PARP inhibitors might be an effective treatment strategy for this deadly disease.


Asunto(s)
MicroARNs/genética , Neoplasias de Células Germinales y Embrionarias/genética , ARN Helicasas DEAD-box/genética , ADN-Topoisomerasas de Tipo I/genética , Humanos , Mutación , Neoplasias de Células Germinales y Embrionarias/diagnóstico , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Poli(ADP-Ribosa) Polimerasas/genética , Polimorfismo de Nucleótido Simple , ARN Largo no Codificante , Recurrencia , Ribonucleasa III/genética
8.
Nature ; 573(7773): 281-286, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31485078

RESUMEN

Enzymes that catalyse CpG methylation in DNA, including the DNA methyltransferases 1 (DNMT1), 3A (DNMT3A) and 3B (DNMT3B), are indispensable for mammalian tissue development and homeostasis1-4. They are also implicated in human developmental disorders and cancers5-8, supporting the critical role of DNA methylation in the specification and maintenance of cell fate. Previous studies have suggested that post-translational modifications of histones are involved in specifying patterns of DNA methyltransferase localization and DNA methylation at promoters and actively transcribed gene bodies9-11. However, the mechanisms that control the establishment and maintenance of intergenic DNA methylation remain poorly understood. Tatton-Brown-Rahman syndrome (TBRS) is a childhood overgrowth disorder that is defined by germline mutations in DNMT3A. TBRS shares clinical features with Sotos syndrome (which is caused by haploinsufficiency of NSD1, a histone methyltransferase that catalyses the dimethylation of histone H3 at K36 (H3K36me2)8,12,13), which suggests that there is a mechanistic link between these two diseases. Here we report that NSD1-mediated H3K36me2 is required for the recruitment of DNMT3A and maintenance of DNA methylation at intergenic regions. Genome-wide analysis shows that the binding and activity of DNMT3A colocalize with H3K36me2 at non-coding regions of euchromatin. Genetic ablation of Nsd1 and its paralogue Nsd2 in mouse cells results in a redistribution of DNMT3A to H3K36me3-modified gene bodies and a reduction in the methylation of intergenic DNA. Blood samples from patients with Sotos syndrome and NSD1-mutant tumours also exhibit hypomethylation of intergenic DNA. The PWWP domain of DNMT3A shows dual recognition of H3K36me2 and H3K36me3 in vitro, with a higher binding affinity towards H3K36me2 that is abrogated by TBRS-derived missense mutations. Together, our study reveals a trans-chromatin regulatory pathway that connects aberrant intergenic CpG methylation to human neoplastic and developmental overgrowth.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , ADN Intergénico/metabolismo , Histonas/metabolismo , Animales , Línea Celular , ADN Metiltransferasa 3A , Estudio de Asociación del Genoma Completo , Trastornos del Crecimiento/genética , Trastornos del Crecimiento/fisiopatología , Humanos , Ratones , Unión Proteica , Dominios Proteicos , Transporte de Proteínas , Síndrome de Sotos/genética , Síndrome de Sotos/fisiopatología
9.
Nat Commun ; 10(1): 1262, 2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30890717

RESUMEN

Lys-27-Met mutations in histone 3 genes (H3K27M) characterize a subgroup of deadly gliomas and decrease genome-wide H3K27 trimethylation. Here we use primary H3K27M tumor lines and isogenic CRISPR-edited controls to assess H3K27M effects in vitro and in vivo. We find that whereas H3K27me3 and H3K27me2 are normally deposited by PRC2 across broad regions, their deposition is severely reduced in H3.3K27M cells. H3K27me3 is unable to spread from large unmethylated CpG islands, while H3K27me2 can be deposited outside these PRC2 high-affinity sites but to levels corresponding to H3K27me3 deposition in wild-type cells. Our findings indicate that PRC2 recruitment and propagation on chromatin are seemingly unaffected by K27M, which mostly impairs spread of the repressive marks it catalyzes, especially H3K27me3. Genome-wide loss of H3K27me3 and me2 deposition has limited transcriptomic consequences, preferentially affecting lowly-expressed genes regulating neurogenesis. Removal of H3K27M restores H3K27me2/me3 spread, impairs cell proliferation, and completely abolishes their capacity to form tumors in mice.


Asunto(s)
Neoplasias Encefálicas/genética , Cromatina/metabolismo , Glioblastoma/genética , Histonas/genética , Complejo Represivo Polycomb 2/metabolismo , Adolescente , Anciano , Animales , Neoplasias Encefálicas/patología , Sistemas CRISPR-Cas , Carcinogénesis/genética , Línea Celular Tumoral , Proliferación Celular/genética , Niño , Islas de CpG/genética , Metilación de ADN/genética , Epigénesis Genética , Femenino , Edición Génica/métodos , Regulación Neoplásica de la Expresión Génica , Glioblastoma/patología , Células HEK293 , Código de Histonas/genética , Histonas/metabolismo , Humanos , Lisina/genética , Masculino , Metionina/genética , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mutación , Neurogénesis/genética , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Nat Genet ; 49(5): 780-788, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28394352

RESUMEN

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.


Asunto(s)
Neoplasias Cerebelosas/genética , Regulación Neoplásica de la Expresión Génica , Meduloblastoma/genética , Transcriptoma , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias Cerebelosas/patología , Niño , Preescolar , Análisis por Conglomerados , Variaciones en el Número de Copia de ADN , Femenino , Perfilación de la Expresión Génica/métodos , Heterogeneidad Genética , Estudio de Asociación del Genoma Completo , Humanos , Mutación INDEL , Masculino , Meduloblastoma/patología , Persona de Mediana Edad , Mutación , Polimorfismo de Nucleótido Simple , Análisis de Componente Principal , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
Nat Genet ; 49(2): 180-185, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28067913

RESUMEN

Human papillomavirus (HPV)-negative head and neck squamous cell carcinomas (HNSCCs) are deadly and common cancers. Recent genomic studies implicate multiple genetic pathways, including cell signaling, cell cycle and immune evasion, in their development. Here we analyze public data sets and uncover a previously unappreciated role of epigenome deregulation in the genesis of 13% of HPV-negative HNSCCs. Specifically, we identify novel recurrent mutations encoding p.Lys36Met (K36M) alterations in multiple H3 histone genes. histones. We further validate the presence of these alterations in multiple independent HNSCC data sets and show that, along with previously described NSD1 mutations, they correspond to a specific DNA methylation cluster. The K36M substitution and NSD1 defects converge on altering methylation of histone H3 at K36 (H3K36), subsequently blocking cellular differentiation and promoting oncogenesis. Our data further indicate limited redundancy for NSD family members in HPV-negative HNSCCs and suggest a potential role for impaired H3K36 methylation in their development. Further investigation of drugs targeting chromatin regulators is warranted in HPV-negative HNSCCs driven by aberrant H3K36 methylation.


Asunto(s)
Carcinoma de Células Escamosas/genética , Metilación de ADN/genética , Neoplasias de Cabeza y Cuello/genética , Histonas/genética , Carcinogénesis/genética , Diferenciación Celular/genética , Epigénesis Genética/genética , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Mutación/genética , Proteínas Nucleares/genética , Papillomaviridae/patogenicidad , Infecciones por Papillomavirus/genética , Carcinoma de Células Escamosas de Cabeza y Cuello
12.
Science ; 352(6287): 844-9, 2016 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-27174990

RESUMEN

Several types of pediatric cancers reportedly contain high-frequency missense mutations in histone H3, yet the underlying oncogenic mechanism remains poorly characterized. Here we report that the H3 lysine 36-to-methionine (H3K36M) mutation impairs the differentiation of mesenchymal progenitor cells and generates undifferentiated sarcoma in vivo. H3K36M mutant nucleosomes inhibit the enzymatic activities of several H3K36 methyltransferases. Depleting H3K36 methyltransferases, or expressing an H3K36I mutant that similarly inhibits H3K36 methylation, is sufficient to phenocopy the H3K36M mutation. After the loss of H3K36 methylation, a genome-wide gain in H3K27 methylation leads to a redistribution of polycomb repressive complex 1 and de-repression of its target genes known to block mesenchymal differentiation. Our findings are mirrored in human undifferentiated sarcomas in which novel K36M/I mutations in H3.1 are identified.


Asunto(s)
Neoplasias Óseas/genética , Carcinogénesis/genética , Condroblastoma/genética , Histonas/genética , Células Madre Mesenquimatosas/patología , Células Madre Neoplásicas/patología , Sarcoma/genética , Animales , Neoplasias Óseas/patología , Carcinogénesis/patología , Preescolar , Condroblastoma/patología , Regulación Neoplásica de la Expresión Génica , Histonas/metabolismo , Humanos , Lisina/genética , Células Madre Mesenquimatosas/metabolismo , Metionina/genética , Metilación , Metiltransferasas/genética , Metiltransferasas/metabolismo , Ratones , Mutación , Mutación Missense , Células Madre Neoplásicas/metabolismo , Nucleosomas/genética , Complejo Represivo Polycomb 1/metabolismo , Sarcoma/patología
13.
Nat Commun ; 7: 11185, 2016 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-27048880

RESUMEN

Diffuse Intrinsic Pontine Gliomas (DIPGs) are deadly paediatric brain tumours where needle biopsies help guide diagnosis and targeted therapies. To address spatial heterogeneity, here we analyse 134 specimens from various neuroanatomical structures of whole autopsy brains from nine DIPG patients. Evolutionary reconstruction indicates histone 3 (H3) K27M--including H3.2K27M--mutations potentially arise first and are invariably associated with specific, high-fidelity obligate partners throughout the tumour and its spread, from diagnosis to end-stage disease, suggesting mutual need for tumorigenesis. These H3K27M ubiquitously-associated mutations involve alterations in TP53 cell-cycle (TP53/PPM1D) or specific growth factor pathways (ACVR1/PIK3R1). Later oncogenic alterations arise in sub-clones and often affect the PI3K pathway. Our findings are consistent with early tumour spread outside the brainstem including the cerebrum. The spatial and temporal homogeneity of main driver mutations in DIPG implies they will be captured by limited biopsies and emphasizes the need to develop therapies specifically targeting obligate oncohistone partnerships.


Asunto(s)
Neoplasias del Tronco Encefálico/genética , Carcinogénesis/genética , Regulación Neoplásica de la Expresión Génica , Glioma/genética , Histonas/genética , Mutación , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo I/metabolismo , Autopsia , Mapeo Encefálico , Tronco Encefálico/metabolismo , Tronco Encefálico/patología , Neoplasias del Tronco Encefálico/metabolismo , Neoplasias del Tronco Encefálico/patología , Carcinogénesis/metabolismo , Carcinogénesis/patología , Cerebro/metabolismo , Cerebro/patología , Niño , Fosfatidilinositol 3-Quinasa Clase Ia , Evolución Clonal , Glioma/metabolismo , Glioma/patología , Histonas/metabolismo , Humanos , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/metabolismo , Proteína Fosfatasa 2C , Transducción de Señal , Técnicas Estereotáxicas , Factores de Tiempo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
14.
Acta Neuropathol ; 128(5): 615-27, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25077668

RESUMEN

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.


Asunto(s)
Neoplasias Encefálicas/genética , Carcinogénesis/genética , Epigénesis Genética/genética , Humanos , Pediatría
15.
Hum Mutat ; 35(11): 1285-9, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25130867

RESUMEN

Mutations in the nuclear-encoded mitochondrial aminoacyl-tRNA synthetases are associated with a range of clinical phenotypes. Here, we report a novel disorder in three adult patients with a phenotype including cataracts, short-stature secondary to growth hormone deficiency, sensorineural hearing deficit, peripheral sensory neuropathy, and skeletal dysplasia. Using SNP genotyping and whole-exome sequencing, we identified a single likely causal variant, a missense mutation in a conserved residue of the nuclear gene IARS2, encoding mitochondrial isoleucyl-tRNA synthetase. The mutation is homozygous in the affected patients, heterozygous in carriers, and absent in control chromosomes. IARS2 protein level was reduced in skin cells cultured from one of the patients, consistent with a pathogenic effect of the mutation. Compound heterozygous mutations in IARS2 were independently identified in a previously unreported patient with a more severe mitochondrial phenotype diagnosed as Leigh syndrome. This is the first report of clinical findings associated with IARS2 mutations.


Asunto(s)
Catarata/genética , Enanismo Hipofisario/genética , Pérdida Auditiva Sensorineural/genética , Isoleucina-ARNt Ligasa/genética , Enfermedad de Leigh/genética , Mutación , Enfermedades del Sistema Nervioso Periférico/genética , Adulto , Secuencia de Aminoácidos , Encéfalo/patología , Catarata/diagnóstico , Consanguinidad , Análisis Mutacional de ADN , Enanismo Hipofisario/diagnóstico , Femenino , Genes Recesivos , Pérdida Auditiva Sensorineural/diagnóstico , Humanos , Isoleucina-ARNt Ligasa/química , Enfermedad de Leigh/diagnóstico , Imagen por Resonancia Magnética , Masculino , Datos de Secuencia Molecular , Linaje , Enfermedades del Sistema Nervioso Periférico/diagnóstico , Fenotipo , Alineación de Secuencia , Síndrome
16.
Nat Genet ; 46(5): 462-6, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24705250

RESUMEN

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.


Asunto(s)
Receptores de Activinas Tipo I/genética , Astrocitoma/genética , Neoplasias Encefálicas/genética , Regulación Neoplásica de la Expresión Génica/genética , Genoma Humano/genética , Mutación/genética , Animales , Secuencia de Bases , Proteínas Morfogenéticas Óseas/metabolismo , Niño , Variaciones en el Número de Copia de ADN/genética , Metilación de ADN/genética , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Datos de Secuencia Molecular , Análisis de Secuencia de ADN , Proteínas Smad/metabolismo
17.
Nat Genet ; 46(1): 39-44, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24316981

RESUMEN

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.


Asunto(s)
Neoplasias Encefálicas/genética , Cromosomas Humanos Par 19 , ADN (Citosina-5-)-Metiltransferasas/genética , Proteínas de la Membrana/genética , MicroARNs/genética , Neoplasias de Células Germinales y Embrionarias/genética , Preescolar , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Fusión Génica , Humanos , Masculino , Isoformas de Proteínas , Proteína p130 Similar a la del Retinoblastoma/genética , Ensayos Antitumor por Modelo de Xenoinjerto , ADN Metiltransferasa 3B
18.
Bioinformatics ; 29(18): 2365-8, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23825369

RESUMEN

MOTIVATION: Feature selection is one of the main challenges in analyzing high-throughput genomic data. Minimum redundancy maximum relevance (mRMR) is a particularly fast feature selection method for finding a set of both relevant and complementary features. Here we describe the mRMRe R package, in which the mRMR technique is extended by using an ensemble approach to better explore the feature space and build more robust predictors. To deal with the computational complexity of the ensemble approach, the main functions of the package are implemented and parallelized in C using the openMP Application Programming Interface. RESULTS: Our ensemble mRMR implementations outperform the classical mRMR approach in terms of prediction accuracy. They identify genes more relevant to the biological context and may lead to richer biological interpretations. The parallelized functions included in the package show significant gains in terms of run-time speed when compared with previously released packages. AVAILABILITY: The R package mRMRe is available on Comprehensive R Archive Network and is provided open source under the Artistic-2.0 License. The code used to generate all the results reported in this application note is available from Supplementary File 1. CONTACT: bhaibeka@ircm.qc.ca SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Genómica/métodos , Programas Informáticos , Algoritmos , Antineoplásicos Fitogénicos/farmacología , Camptotecina/análogos & derivados , Camptotecina/farmacología , Resistencia a Antineoplásicos , Irinotecán
19.
J Am Med Inform Assoc ; 20(4): 597-602, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23355484

RESUMEN

BACKGROUND: An enduring challenge in personalized medicine lies in selecting the right drug for each individual patient. While testing of drugs on patients in large trials is the only way to assess their clinical efficacy and toxicity, we dramatically lack resources to test the hundreds of drugs currently under development. Therefore the use of preclinical model systems has been intensively investigated as this approach enables response to hundreds of drugs to be tested in multiple cell lines in parallel. METHODS: Two large-scale pharmacogenomic studies recently screened multiple anticancer drugs on over 1000 cell lines. We propose to combine these datasets to build and robustly validate genomic predictors of drug response. We compared five different approaches for building predictors of increasing complexity. We assessed their performance in cross-validation and in two large validation sets, one containing the same cell lines present in the training set and another dataset composed of cell lines that have never been used during the training phase. RESULTS: Sixteen drugs were found in common between the datasets. We were able to validate multivariate predictors for three out of the 16 tested drugs, namely irinotecan, PD-0325901, and PLX4720. Moreover, we observed that response to 17-AAG, an inhibitor of Hsp90, could be efficiently predicted by the expression level of a single gene, NQO1. CONCLUSION: These results suggest that genomic predictors could be robustly validated for specific drugs. If successfully validated in patients' tumor cells, and subsequently in clinical trials, they could act as companion tests for the corresponding drugs and play an important role in personalized medicine.


Asunto(s)
Antineoplásicos/uso terapéutico , Modelos Genéticos , Neoplasias/genética , Farmacogenética , Medicina de Precisión , Línea Celular Tumoral , Biología Computacional , Bases de Datos Genéticas , Humanos , Neoplasias/tratamiento farmacológico , Transcriptoma
20.
Am J Hum Genet ; 90(4): 693-700, 2012 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-22425360

RESUMEN

Joubert syndrome (JBTS) is an autosomal-recessive disorder characterized by a distinctive mid-hindbrain malformation, developmental delay with hypotonia, ocular-motor apraxia, and breathing abnormalities. Although JBTS was first described more than 40 years ago in French Canadian siblings, the causal mutations have not yet been identified in this family nor in most French Canadian individuals subsequently described. We ascertained a cluster of 16 JBTS-affected individuals from 11 families living in the Lower St. Lawrence region. SNP genotyping excluded the presence of a common homozygous mutation that would explain the clustering of these individuals. Exome sequencing performed on 15 subjects showed that nine affected individuals from seven families (including the original JBTS family) carried rare compound-heterozygous mutations in C5ORF42. Two missense variants (c.4006C>T [p.Arg1336Trp] and c.4690G>A [p.Ala1564Thr]) and a splicing mutation (c.7400+1G>A), which causes exon skipping, were found in multiple subjects that were not known to be related, whereas three other truncating mutations (c.6407del [p.Pro2136Hisfs*31], c.4804C>T [p.Arg1602*], and c.7477C>T [p.Arg2493*]) were identified in single individuals. None of the unaffected first-degree relatives were compound heterozygous for these mutations. Moreover, none of the six putative mutations were detected among 477 French Canadian controls. Our data suggest that mutations in C5ORF42 explain a large portion of French Canadian individuals with JBTS.


Asunto(s)
Enfermedades Cerebelosas/genética , Anomalías del Ojo/genética , Enfermedades Renales Quísticas/genética , Proteínas de la Membrana/genética , Mutación , Anomalías Múltiples , Adulto , Secuencia de Bases , Canadá , Cerebelo/anomalías , Niño , Preescolar , Exoma , Femenino , Heterocigoto , Homocigoto , Humanos , Masculino , Datos de Secuencia Molecular , Polimorfismo de Nucleótido Simple , Retina/anomalías
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...