Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Biotechnol ; 40(4): 585-597, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35361996

RESUMO

Profiling of circulating tumor DNA (ctDNA) in the bloodstream shows promise for noninvasive cancer detection. Chromatin fragmentation features have previously been explored to infer gene expression profiles from cell-free DNA (cfDNA), but current fragmentomic methods require high concentrations of tumor-derived DNA and provide limited resolution. Here we describe promoter fragmentation entropy as an epigenomic cfDNA feature that predicts RNA expression levels at individual genes. We developed 'epigenetic expression inference from cell-free DNA-sequencing' (EPIC-seq), a method that uses targeted sequencing of promoters of genes of interest. Profiling 329 blood samples from 201 patients with cancer and 87 healthy adults, we demonstrate classification of subtypes of lung carcinoma and diffuse large B cell lymphoma. Applying EPIC-seq to serial blood samples from patients treated with PD-(L)1 immune-checkpoint inhibitors, we show that gene expression profiles inferred by EPIC-seq are correlated with clinical response. Our results indicate that EPIC-seq could enable noninvasive, high-throughput tissue-of-origin characterization with diagnostic, prognostic and therapeutic potential.


Assuntos
Ácidos Nucleicos Livres , Neoplasias , Adulto , Biomarcadores Tumorais/genética , Ácidos Nucleicos Livres/genética , Fragmentação do DNA , Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Mutação
2.
Artigo em Inglês | MEDLINE | ID: mdl-29990066

RESUMO

Gene-expression-based classification and regression are major concerns in translational genomics. If the feature-label distribution is known, then an optimal classifier can be derived. If the predictor-target distribution is known, then an optimal regression function can be derived. In practice, neither is known, data must be employed, and, for small samples, prior knowledge concerning the feature-label or predictor-target distribution can be used in the learning process. Optimal Bayesian classification and optimal Bayesian regression provide optimality under uncertainty. With optimal Bayesian classification (or regression), uncertainty is treated directly on the feature-label (or predictor-target) distribution. The fundamental engineering problem is prior construction. The Regularized Expected Mean Log-Likelihood Prior (REMLP) utilizes pathway information and provides viable priors for the feature-label distribution, assuming that the training data contain labels. In practice, the labels may not be observed. This paper extends the REMLP methodology to a Gaussian mixture model (GMM) when the labels are unknown. Prior construction bundled with prior update via Bayesian sampling results in Monte Carlo approximations to the optimal Bayesian regression function and optimal Bayesian classifier. Simulations demonstrate that the GMM REMLP prior yields better performance than the EM algorithm for small data sets. We apply it to phenotype classification when the prior knowledge consists of colon cancer pathways.


Assuntos
Perfilação da Expressão Gênica/métodos , Genômica/métodos , Modelos Estatísticos , Algoritmos , Teorema de Bayes , Neoplasias do Colo/genética , Bases de Dados Genéticas , Humanos , Distribuição Normal
3.
J Clin Oncol ; 36(28): 2845-2853, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30125215

RESUMO

PURPOSE: Outcomes for patients with diffuse large B-cell lymphoma remain heterogeneous, with existing methods failing to consistently predict treatment failure. We examined the additional prognostic value of circulating tumor DNA (ctDNA) before and during therapy for predicting patient outcomes. PATIENTS AND METHODS: We studied the dynamics of ctDNA from 217 patients treated at six centers, using a training and validation framework. We densely characterized early ctDNA dynamics during therapy using cancer personalized profiling by deep sequencing to define response-associated thresholds within a discovery set. These thresholds were assessed in two independent validation sets. Finally, we assessed the prognostic value of ctDNA in the context of established risk factors, including the International Prognostic Index and interim positron emission tomography/computed tomography scans. RESULTS: Before therapy, ctDNA was detectable in 98% of patients; pretreatment levels were prognostic in both front-line and salvage settings. In the discovery set, ctDNA levels changed rapidly, with a 2-log decrease after one cycle (early molecular response [EMR]) and a 2.5-log decrease after two cycles (major molecular response [MMR]) stratifying outcomes. In the first validation set, patients receiving front-line therapy achieving EMR or MMR had superior outcomes at 24 months (EMR: EFS, 83% v 50%; P = .0015; MMR: EFS, 82% v 46%; P < .001). EMR also predicted superior 24-month outcomes in patients receiving salvage therapy in the first validation set (EFS, 100% v 13%; P = .011). The prognostic value of EMR and MMR was further confirmed in the second validation set. In multivariable analyses including International Prognostic Index and interim positron emission tomography/computed tomography scans across both cohorts, molecular response was independently prognostic of outcomes, including event-free and overall survival. CONCLUSION: Pretreatment ctDNA levels and molecular responses are independently prognostic of outcomes in aggressive lymphomas. These risk factors could potentially guide future personalized risk-directed approaches.


Assuntos
Biomarcadores Tumorais/sangue , DNA Tumoral Circulante/sangue , Linfoma Difuso de Grandes Células B/sangue , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Adulto , Idoso , Biomarcadores Tumorais/genética , Feminino , Humanos , Linfoma Difuso de Grandes Células B/patologia , Masculino , Pessoa de Meia-Idade , Prognóstico , Intervalo Livre de Progressão , Resultado do Tratamento
4.
Cancer Discov ; 7(12): 1394-1403, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28899864

RESUMO

Identifying molecular residual disease (MRD) after treatment of localized lung cancer could facilitate early intervention and personalization of adjuvant therapies. Here, we apply cancer personalized profiling by deep sequencing (CAPP-seq) circulating tumor DNA (ctDNA) analysis to 255 samples from 40 patients treated with curative intent for stage I-III lung cancer and 54 healthy adults. In 94% of evaluable patients experiencing recurrence, ctDNA was detectable in the first posttreatment blood sample, indicating reliable identification of MRD. Posttreatment ctDNA detection preceded radiographic progression in 72% of patients by a median of 5.2 months, and 53% of patients harbored ctDNA mutation profiles associated with favorable responses to tyrosine kinase inhibitors or immune checkpoint blockade. Collectively, these results indicate that ctDNA MRD in patients with lung cancer can be accurately detected using CAPP-seq and may allow personalized adjuvant treatment while disease burden is lowest.Significance: This study shows that ctDNA analysis can robustly identify posttreatment MRD in patients with localized lung cancer, identifying residual/recurrent disease earlier than standard-of-care radiologic imaging, and thus could facilitate personalized adjuvant treatment at early time points when disease burden is lowest. Cancer Discov; 7(12); 1394-403. ©2017 AACR.See related commentary by Comino-Mendez and Turner, p. 1368This article is highlighted in the In This Issue feature, p. 1355.


Assuntos
DNA Tumoral Circulante/genética , Neoplasias Pulmonares/genética , Neoplasia Residual/diagnóstico , Feminino , Humanos , Masculino , Neoplasia Residual/patologia
5.
BMC Bioinformatics ; 18(Suppl 14): 552, 2017 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-29297278

RESUMO

BACKGROUND: Phenotypic classification is problematic because small samples are ubiquitous; and, for these, use of prior knowledge is critical. If knowledge concerning the feature-label distribution - for instance, genetic pathways - is available, then it can be used in learning. Optimal Bayesian classification provides optimal classification under model uncertainty. It differs from classical Bayesian methods in which a classification model is assumed and prior distributions are placed on model parameters. With optimal Bayesian classification, uncertainty is treated directly on the feature-label distribution, which assures full utilization of prior knowledge and is guaranteed to outperform classical methods. RESULTS: The salient problem confronting optimal Bayesian classification is prior construction. In this paper, we propose a new prior construction methodology based on a general framework of constraints in the form of conditional probability statements. We call this prior the maximal knowledge-driven information prior (MKDIP). The new constraint framework is more flexible than our previous methods as it naturally handles the potential inconsistency in archived regulatory relationships and conditioning can be augmented by other knowledge, such as population statistics. We also extend the application of prior construction to a multinomial mixture model when labels are unknown, which often occurs in practice. The performance of the proposed methods is examined on two important pathway families, the mammalian cell-cycle and a set of p53-related pathways, and also on a publicly available gene expression dataset of non-small cell lung cancer when combined with the existing prior knowledge on relevant signaling pathways. CONCLUSION: The new proposed general prior construction framework extends the prior construction methodology to a more flexible framework that results in better inference when proper prior knowledge exists. Moreover, the extension of optimal Bayesian classification to multinomial mixtures where data sets are both small and unlabeled, enables superior classifier design using small, unstructured data sets. We have demonstrated the effectiveness of our approach using pathway information and available knowledge of gene regulating functions; however, the underlying theory can be applied to a wide variety of knowledge types, and other applications when there are small samples.


Assuntos
Algoritmos , Animais , Teorema de Bayes , Carcinoma Pulmonar de Células não Pequenas/genética , Ciclo Celular , Entropia , Humanos , Teoria da Informação , Neoplasias Pulmonares/genética , Mamíferos/metabolismo , Probabilidade , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo
6.
Sci Transl Med ; 8(364): 364ra155, 2016 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-27831904

RESUMO

Patients with diffuse large B cell lymphoma (DLBCL) exhibit marked diversity in tumor behavior and outcomes, yet the identification of poor-risk groups remains challenging. In addition, the biology underlying these differences is incompletely understood. We hypothesized that characterization of mutational heterogeneity and genomic evolution using circulating tumor DNA (ctDNA) profiling could reveal molecular determinants of adverse outcomes. To address this hypothesis, we applied cancer personalized profiling by deep sequencing (CAPP-Seq) analysis to tumor biopsies and cell-free DNA samples from 92 lymphoma patients and 24 healthy subjects. At diagnosis, the amount of ctDNA was found to strongly correlate with clinical indices and was independently predictive of patient outcomes. We demonstrate that ctDNA genotyping can classify transcriptionally defined tumor subtypes, including DLBCL cell of origin, directly from plasma. By simultaneously tracking multiple somatic mutations in ctDNA, our approach outperformed immunoglobulin sequencing and radiographic imaging for the detection of minimal residual disease and facilitated noninvasive identification of emergent resistance mutations to targeted therapies. In addition, we identified distinct patterns of clonal evolution distinguishing indolent follicular lymphomas from those that transformed into DLBCL, allowing for potential noninvasive prediction of histological transformation. Collectively, our results demonstrate that ctDNA analysis reveals biological factors that underlie lymphoma clinical outcomes and could facilitate individualized therapy.


Assuntos
DNA Tumoral Circulante/genética , Linfoma de Células B/genética , Linfoma Difuso de Grandes Células B/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Algoritmos , Biomarcadores Tumorais/sangue , Biópsia , Sistema Livre de Células , Feminino , Genótipo , Humanos , Imunoglobulinas/química , Linfoma de Células B/sangue , Linfoma Difuso de Grandes Células B/sangue , Masculino , Pessoa de Meia-Idade , Mutação , Prognóstico , Recidiva , Resultado do Tratamento
7.
BMC Bioinformatics ; 12 Suppl 10: S9, 2011 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-22166046

RESUMO

BACKGROUND: Accumulation of gene mutations in cells is known to be responsible for tumor progression, driving it from benign states to malignant states. However, previous studies have shown that the detailed sequence of gene mutations, or the steps in tumor progression, may vary from tumor to tumor, making it difficult to infer the exact path that a given type of tumor may have taken. RESULTS: In this paper, we propose an effective probabilistic algorithm for reconstructing the tumor progression process based on partial knowledge of the underlying gene regulatory network and the steady state distribution of the gene expression values in a given tumor. We take the BNp (Boolean networks with pertubation) framework to model the gene regulatory networks. We assume that the true network is not exactly known but we are given an uncertainty class of networks that contains the true network. This network uncertainty class arises from our partial knowledge of the true network, typically represented as a set of local pathways that are embedded in the global network. Given the SSD of the cancerous network, we aim to simultaneously identify the true normal (healthy) network and the set of gene mutations that drove the network into the cancerous state. This is achieved by analyzing the effect of gene mutation on the SSD of a gene regulatory network. At each step, the proposed algorithm reduces the uncertainty class by keeping only those networks whose SSDs get close enough to the cancerous SSD as a result of additional gene mutation. These steps are repeated until we can find the best candidate for the true network and the most probable path of tumor progression. CONCLUSIONS: Simulation results based on both synthetic networks and networks constructed from actual pathway knowledge show that the proposed algorithm can identify the normal network and the actual path of tumor progression with high probability. The algorithm is also robust to model mismatch and allows us to control the trade-off between efficiency and accuracy.


Assuntos
Algoritmos , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Modelos Estatísticos , Neoplasias/genética , Perfilação da Expressão Gênica , Humanos , Processamento de Imagem Assistida por Computador , Mutação , Proteínas Proto-Oncogênicas c-mdm2/genética , Transdução de Sinais , Proteína Supressora de Tumor p53/genética , Incerteza
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA