Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
PLoS Genet ; 12(2): e1005888, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26925970

RESUMEN

Retinoblastoma (Rb), the most common pediatric intraocular neoplasm, results from inactivation of both alleles of the RB1 tumor suppressor gene. The second allele is most commonly lost, as demonstrated by loss of heterozygosity studies. RB1 germline carriers usually develop bilateral tumors, but some Rb families display low penetrance and variable expressivity. In order to decipher the underlying mechanisms, 23 unrelated low penetrance pedigrees segregating the common c.1981C>T/p.Arg661Trp mutation and other low penetrance mutations were studied. In families segregating the c.1981C>T mutation, we demonstrated, for the first time, a correlation between the gender of the transmitting carrier and penetrance, as evidenced by Fisher's exact test: the probability of being unaffected is 90.3% and 32.5% when the mutation is inherited from the mother and the father, respectively (p-value = 7.10(-7). Interestingly, a similar correlation was observed in families segregating other low penetrance alleles. Consequently, we investigated the putative involvement of an imprinted, modifier gene in low penetrance Rb. We first ruled out a MED4-driven mechanism by MED4 methylation and expression analyses. We then focused on the differentially methylated CpG85 island located in intron 2 of RB1 and showing parent-of-origin-specific DNA methylation. This differential methylation promotes expression of the maternal c.1981C>T allele. We propose that the maternally inherited c.1981C>T/p.Arg661Trp allele retains sufficient tumor suppressor activity to prevent retinoblastoma development. In contrast, when the mutation is paternally transmitted, the low residual activity would mimic a null mutation and subsequently lead to retinoblastoma. This implies that the c.1981C>T mutation is not deleterious per se but needs to be destabilized in order to reach pRb haploinsufficiency and initiate tumorigenesis. We suggest that this phenomenon might be a general mechanism to explain phenotypic differences in low penetrance Rb families.


Asunto(s)
Mutación , Neoplasias de la Retina/genética , Proteína de Retinoblastoma/genética , Retinoblastoma/genética , Islas de CpG , Metilación de ADN , Femenino , Heterocigoto , Humanos , Masculino , Complejo Mediador/genética , Complejo Mediador/metabolismo , Linaje , Penetrancia , Fenotipo , Neoplasias de la Retina/patología , Retinoblastoma/patología , Proteína de Retinoblastoma/metabolismo
2.
Nat Commun ; 14(1): 6695, 2023 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-37932267

RESUMEN

Mismatch Repair Deficiency (dMMR)/Microsatellite Instability (MSI) is a key biomarker in colorectal cancer (CRC). Universal screening of CRC patients for MSI status is now recommended, but contributes to increased workload for pathologists and delayed therapeutic decisions. Deep learning has the potential to ease dMMR/MSI testing and accelerate oncologist decision making in clinical practice, yet no comprehensive validation of a clinically approved tool has been conducted. We developed MSIntuit, a clinically approved artificial intelligence (AI) based pre-screening tool for MSI detection from haematoxylin-eosin (H&E) stained slides. After training on samples from The Cancer Genome Atlas (TCGA), a blind validation is performed on an independent dataset of 600 consecutive CRC patients. Inter-scanner reliability is studied by digitising each slide using two different scanners. MSIntuit yields a sensitivity of 0.96-0.98, a specificity of 0.47-0.46, and an excellent inter-scanner agreement (Cohen's κ: 0.82). By reaching high sensitivity comparable to gold standard methods while ruling out almost half of the non-MSI population, we show that MSIntuit can effectively serve as a pre-screening tool to alleviate MSI testing burden in clinical practice.


Asunto(s)
Neoplasias Colorrectales , Inestabilidad de Microsatélites , Humanos , Inteligencia Artificial , Reproducibilidad de los Resultados , Detección Precoz del Cáncer , Neoplasias Colorrectales/genética , Reparación de la Incompatibilidad de ADN
3.
Eur J Cancer ; 174: 90-98, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35985252

RESUMEN

BACKGROUND: The need for developing new biomarkers is increasing with the emergence of many targeted therapies. Artificial Intelligence (AI) algorithms have shown great promise in the medical imaging field to build predictive models. We developed a prognostic model for solid tumour patients using AI on multimodal data. PATIENTS AND METHODS: Our retrospective study included examinations of patients with seven different cancer types performed between 2003 and 2017 in 17 different hospitals. Radiologists annotated all metastases on baseline computed tomography (CT) and ultrasound (US) images. Imaging features were extracted using AI models and used along with the patients' and treatments' metadata. A Cox regression was fitted to predict prognosis. Performance was assessed on a left-out test set with 1000 bootstraps. RESULTS: The model was built on 436 patients and tested on 196 patients (mean age 59, IQR: 51-6, 411 men out of 616 patients). On the whole, 1147 US images were annotated with lesions delineation, and 632 thorax-abdomen-pelvis CTs (total of 301,975 slices) were fully annotated with a total of 9516 lesions. The developed model reaches an average concordance index of 0.71 (0.67-0.76, 95% CI). Using the median predicted risk as a threshold value, the model is able to significantly (log-rank test P value < 0.001) isolate high-risk patients from low-risk patients (respective median OS of 11 and 31 months) with a hazard ratio of 3.5 (2.4-5.2, 95% CI). CONCLUSION: AI was able to extract prognostic features from imaging data, and along with clinical data, allows an accurate stratification of patients' prognoses.


Asunto(s)
Inteligencia Artificial , Neoplasias , Biomarcadores , Humanos , Masculino , Persona de Mediana Edad , Neoplasias/diagnóstico por imagen , Estudios Retrospectivos , Tomografía Computarizada por Rayos X/métodos
4.
Nat Commun ; 12(1): 5578, 2021 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-34552068

RESUMEN

Retinoblastoma is the most frequent intraocular malignancy in children, originating from a maturing cone precursor in the developing retina. Little is known on the molecular basis underlying the biological and clinical behavior of this cancer. Here, using multi-omics data, we demonstrate the existence of two retinoblastoma subtypes. Subtype 1, of earlier onset, includes most of the heritable forms. It harbors few genetic alterations other than the initiating RB1 inactivation and corresponds to differentiated tumors expressing mature cone markers. By contrast, subtype 2 tumors harbor frequent recurrent genetic alterations including MYCN-amplification. They express markers of less differentiated cone together with neuronal/ganglion cell markers with marked inter- and intra-tumor heterogeneity. The cone dedifferentiation in subtype 2 is associated with stemness features including low immune and interferon response, E2F and MYC/MYCN activation and a higher propensity for metastasis. The recognition of these two subtypes, one maintaining a cone-differentiated state, and the other, more aggressive, associated with cone dedifferentiation and expression of neuronal markers, opens up important biological and clinical perspectives for retinoblastomas.


Asunto(s)
Células Fotorreceptoras Retinianas Conos/patología , Células Ganglionares de la Retina/metabolismo , Neoplasias de la Retina/clasificación , Retinoblastoma/clasificación , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Desdiferenciación Celular/genética , Preescolar , Metilación de ADN , Femenino , Expresión Génica , Heterogeneidad Genética , Humanos , Lactante , Masculino , Mutación , Proteína Proto-Oncogénica N-Myc/genética , Metástasis de la Neoplasia , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Ganglionares de la Retina/patología , Neoplasias de la Retina/genética , Neoplasias de la Retina/metabolismo , Neoplasias de la Retina/patología , Retinoblastoma/genética , Retinoblastoma/metabolismo , Retinoblastoma/patología
5.
Nat Commun ; 12(1): 634, 2021 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-33504775

RESUMEN

The SARS-COV-2 pandemic has put pressure on intensive care units, so that identifying predictors of disease severity is a priority. We collect 58 clinical and biological variables, and chest CT scan data, from 1003 coronavirus-infected patients from two French hospitals. We train a deep learning model based on CT scans to predict severity. We then construct the multimodal AI-severity score that includes 5 clinical and biological variables (age, sex, oxygenation, urea, platelet) in addition to the deep learning model. We show that neural network analysis of CT-scans brings unique prognosis information, although it is correlated with other markers of severity (oxygenation, LDH, and CRP) explaining the measurable but limited 0.03 increase of AUC obtained when adding CT-scan information to clinical variables. Here, we show that when comparing AI-severity with 11 existing severity scores, we find significantly improved prognosis performance; AI-severity can therefore rapidly become a reference scoring approach.


Asunto(s)
COVID-19/diagnóstico , COVID-19/fisiopatología , Aprendizaje Profundo , Redes Neurales de la Computación , Tomografía Computarizada por Rayos X/métodos , Inteligencia Artificial , COVID-19/clasificación , Humanos , Modelos Biológicos , Análisis Multivariante , Pronóstico , Radiólogos , Índice de Severidad de la Enfermedad
6.
Nat Commun ; 11(1): 3877, 2020 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-32747659

RESUMEN

Deep learning methods for digital pathology analysis are an effective way to address multiple clinical questions, from diagnosis to prediction of treatment outcomes. These methods have also been used to predict gene mutations from pathology images, but no comprehensive evaluation of their potential for extracting molecular features from histology slides has yet been performed. We show that HE2RNA, a model based on the integration of multiple data modes, can be trained to systematically predict RNA-Seq profiles from whole-slide images alone, without expert annotation. Through its interpretable design, HE2RNA provides virtual spatialization of gene expression, as validated by CD3- and CD20-staining on an independent dataset. The transcriptomic representation learned by HE2RNA can also be transferred on other datasets, even of small size, to increase prediction performance for specific molecular phenotypes. We illustrate the use of this approach in clinical diagnosis purposes such as the identification of tumors with microsatellite instability.


Asunto(s)
Biología Computacional/métodos , Aprendizaje Profundo , Regulación Neoplásica de la Expresión Génica , Procesamiento de Imagen Asistido por Computador/métodos , Neoplasias/genética , RNA-Seq/métodos , Algoritmos , Perfilación de la Expresión Génica/métodos , Humanos , Inestabilidad de Microsatélites , Modelos Genéticos , Neoplasias/diagnóstico , Neoplasias/metabolismo
7.
Nat Med ; 25(10): 1519-1525, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31591589

RESUMEN

Malignant mesothelioma (MM) is an aggressive cancer primarily diagnosed on the basis of histological criteria1. The 2015 World Health Organization classification subdivides mesothelioma tumors into three histological types: epithelioid, biphasic and sarcomatoid MM. MM is a highly complex and heterogeneous disease, rendering its diagnosis and histological typing difficult and leading to suboptimal patient care and decisions regarding treatment modalities2. Here we have developed a new approach-based on deep convolutional neural networks-called MesoNet to accurately predict the overall survival of mesothelioma patients from whole-slide digitized images, without any pathologist-provided locally annotated regions. We validated MesoNet on both an internal validation cohort from the French MESOBANK and an independent cohort from The Cancer Genome Atlas (TCGA). We also demonstrated that the model was more accurate in predicting patient survival than using current pathology practices. Furthermore, unlike classical black-box deep learning methods, MesoNet identified regions contributing to patient outcome prediction. Strikingly, we found that these regions are mainly located in the stroma and are histological features associated with inflammation, cellular diversity and vacuolization. These findings suggest that deep learning models can identify new features predictive of patient survival and potentially lead to new biomarker discoveries.


Asunto(s)
Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/patología , Mesotelioma/diagnóstico , Mesotelioma/patología , Pronóstico , Aprendizaje Profundo , Femenino , Humanos , Neoplasias Pulmonares/clasificación , Masculino , Mesotelioma/clasificación , Mesotelioma Maligno , Clasificación del Tumor , Redes Neurales de la Computación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA