Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nucleic Acids Res ; 51(D1): D1242-D1248, 2023 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-36259664

RESUMO

Extensive in vitro cancer drug screening datasets have enabled scientists to identify biomarkers and develop machine learning models for predicting drug sensitivity. While most advancements have focused on omics profiles, cancer drug sensitivity scores precalculated by the original sources are often used as-is, without consideration for variabilities between studies. It is well-known that significant inconsistencies exist between the drug sensitivity scores across datasets due to differences in experimental setups and preprocessing methods used to obtain the sensitivity scores. As a result, many studies opt to focus only on a single dataset, leading to underutilization of available data and a limited interpretation of cancer pharmacogenomics analysis. To overcome these caveats, we have developed CREAMMIST (https://creammist.mtms.dev), an integrative database that enables users to obtain an integrative dose-response curve, to capture uncertainty (or high certainty when multiple datasets well align) across five widely used cancer cell-line drug-response datasets. We utilized the Bayesian framework to systematically integrate all available dose-response values across datasets (>14 millions dose-response data points). CREAMMIST provides easy-to-use statistics derived from the integrative dose-response curves for various downstream analyses such as identifying biomarkers, selecting drug concentrations for experiments, and training robust machine learning models.


Assuntos
Antineoplásicos , Bases de Dados Factuais , Neoplasias , Humanos , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Teorema de Bayes , Biomarcadores , Aprendizado de Máquina , Neoplasias/tratamento farmacológico , Neoplasias/genética
2.
Genome Med ; 13(1): 189, 2021 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-34915921

RESUMO

While understanding molecular heterogeneity across patients underpins precision oncology, there is increasing appreciation for taking intra-tumor heterogeneity into account. Based on large-scale analysis of cancer omics datasets, we highlight the importance of intra-tumor transcriptomic heterogeneity (ITTH) for predicting clinical outcomes. Leveraging single-cell RNA-seq (scRNA-seq) with a recommender system (CaDRReS-Sc), we show that heterogeneous gene-expression signatures can predict drug response with high accuracy (80%). Using patient-proximal cell lines, we established the validity of CaDRReS-Sc's monotherapy (Pearson r>0.6) and combinatorial predictions targeting clone-specific vulnerabilities (>10% improvement). Applying CaDRReS-Sc to rapidly expanding scRNA-seq compendiums can serve as in silico screen to accelerate drug-repurposing studies. Availability: https://github.com/CSB5/CaDRReS-Sc .


Assuntos
Neoplasias , Transcriptoma , Células Clonais , Perfilação da Expressão Gênica , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Medicina de Precisão , Análise de Sequência de RNA , Análise de Célula Única , Software
3.
Bioinformatics ; 37(Supplement_1): i76-i83, 2021 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-34000002

RESUMO

MOTIVATION: Large-scale cancer omics studies have highlighted the diversity of patient molecular profiles and the importance of leveraging this information to deliver the right drug to the right patient at the right time. Key challenges in learning predictive models for this include the high-dimensionality of omics data and heterogeneity in biological and clinical factors affecting patient response. The use of multi-task learning techniques has been widely explored to address dataset limitations for in vitro drug response models, while domain adaptation (DA) has been employed to extend them to predict in vivo response. In both of these transfer learning settings, noisy data for some tasks (or domains) can substantially reduce the performance for others compared to single-task (domain) learners, i.e. lead to negative transfer (NT). RESULTS: We describe a novel multi-task unsupervised DA method (TUGDA) that addresses these limitations in a unified framework by quantifying uncertainty in predictors and weighting their influence on shared feature representations. TUGDA's ability to rely more on predictors with low-uncertainty allowed it to notably reduce cases of NT for in vitro models (94% overall) compared to state-of-the-art methods. For DA to in vivo settings, TUGDA improved over previous methods for patient-derived xenografts (9 out of 14 drugs) as well as patient datasets (significant associations in 9 out of 22 drugs). TUGDA's ability to avoid NT thus provides a key capability as we try to integrate diverse drug-response datasets to build consistent predictive models with in vivo utility. AVAILABILITYAND IMPLEMENTATION: https://github.com/CSB5/TUGDA. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.

5.
Bioinformatics ; 34(22): 3907-3914, 2018 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-29868820

RESUMO

Motivation: As we move toward an era of precision medicine, the ability to predict patient-specific drug responses in cancer based on molecular information such as gene expression data represents both an opportunity and a challenge. In particular, methods are needed that can accommodate the high-dimensionality of data to learn interpretable models capturing drug response mechanisms, as well as providing robust predictions across datasets. Results: We propose a method based on ideas from 'recommender systems' (CaDRReS) that predicts cancer drug responses for unseen cell-lines/patients based on learning projections for drugs and cell-lines into a latent 'pharmacogenomic' space. Comparisons with other proposed approaches for this problem based on large public datasets (CCLE and GDSC) show that CaDRReS provides consistently good models and robust predictions even across unseen patient-derived cell-line datasets. Analysis of the pharmacogenomic spaces inferred by CaDRReS also suggests that they can be used to understand drug mechanisms, identify cellular subtypes and further characterize drug-pathway associations. Availability and implementation: Source code and datasets are available at https://github.com/CSB5/CaDRReS. Supplementary information: Supplementary data are available at Bioinformatics online.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias , Humanos , Neoplasias/tratamento farmacológico , Farmacogenética , Medicina de Precisão , Software
6.
Cell ; 173(2): 305-320.e10, 2018 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-29625049

RESUMO

The Cancer Genome Atlas (TCGA) has catalyzed systematic characterization of diverse genomic alterations underlying human cancers. At this historic junction marking the completion of genomic characterization of over 11,000 tumors from 33 cancer types, we present our current understanding of the molecular processes governing oncogenesis. We illustrate our insights into cancer through synthesis of the findings of the TCGA PanCancer Atlas project on three facets of oncogenesis: (1) somatic driver mutations, germline pathogenic variants, and their interactions in the tumor; (2) the influence of the tumor genome and epigenome on transcriptome and proteome; and (3) the relationship between tumor and the microenvironment, including implications for drugs targeting driver events and immunotherapies. These results will anchor future characterization of rare and common tumor types, primary and relapsed tumors, and cancers across ancestry groups and will guide the deployment of clinical genomic sequencing.


Assuntos
Carcinogênese/genética , Genômica , Neoplasias/patologia , Reparo do DNA/genética , Bases de Dados Genéticas , Genes Neoplásicos , Humanos , Redes e Vias Metabólicas/genética , Instabilidade de Microssatélites , Mutação , Neoplasias/genética , Neoplasias/imunologia , Transcriptoma , Microambiente Tumoral/genética
7.
Cell ; 173(2): 371-385.e18, 2018 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-29625053

RESUMO

Identifying molecular cancer drivers is critical for precision oncology. Multiple advanced algorithms to identify drivers now exist, but systematic attempts to combine and optimize them on large datasets are few. We report a PanCancer and PanSoftware analysis spanning 9,423 tumor exomes (comprising all 33 of The Cancer Genome Atlas projects) and using 26 computational tools to catalog driver genes and mutations. We identify 299 driver genes with implications regarding their anatomical sites and cancer/cell types. Sequence- and structure-based analyses identified >3,400 putative missense driver mutations supported by multiple lines of evidence. Experimental validation confirmed 60%-85% of predicted mutations as likely drivers. We found that >300 MSI tumors are associated with high PD-1/PD-L1, and 57% of tumors analyzed harbor putative clinically actionable events. Our study represents the most comprehensive discovery of cancer genes and mutations to date and will serve as a blueprint for future biological and clinical endeavors.


Assuntos
Neoplasias/patologia , Algoritmos , Antígeno B7-H1/genética , Biologia Computacional , Bases de Dados Genéticas , Entropia , Humanos , Instabilidade de Microssatélites , Mutação , Neoplasias/genética , Neoplasias/imunologia , Análise de Componente Principal , Receptor de Morte Celular Programada 1/genética
8.
Cancer Res ; 78(1): 290-301, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29259006

RESUMO

Existing cancer driver prediction methods are based on very different assumptions and each of them can detect only a particular subset of driver genes. Here we perform a comprehensive assessment of 18 driver prediction methods on more than 3,400 tumor samples from 15 cancer types, all to determine their suitability in guiding precision medicine efforts. We categorized these methods into five groups: functional impact on proteins in general (FI) or specific to cancer (FIC), cohort-based analysis for recurrent mutations (CBA), mutations with expression correlation (MEC), and methods that use gene interaction network-based analysis (INA). The performance of driver prediction methods varied considerably, with concordance with a gold standard varying from 9% to 68%. FI methods showed relatively poor performance (concordance <22%), while CBA methods provided conservative results but required large sample sizes for high sensitivity. INA methods, through the integration of genomic and transcriptomic data, and FIC methods, by training cancer-specific models, provided the best trade-off between sensitivity and specificity. As the methods were found to predict different subsets of driver genes, we propose a novel consensus-based approach, ConsensusDriver, which significantly improves the quality of predictions (20% increase in sensitivity) in patient subgroups or even individual patients. Consensus-based methods like ConsensusDriver promise to harness the strengths of different driver prediction paradigms.Significance: These findings assess state-of-the-art cancer driver prediction methods and develop a new and improved consensus-based approach for use in precision oncology. Cancer Res; 78(1); 290-301. ©2017 AACR.


Assuntos
Algoritmos , Neoplasias/genética , Medicina de Precisão/métodos , Biologia Computacional/métodos , Genes , Humanos , Mutação , Transcriptoma
9.
Bioinformatics ; 31(12): i250-7, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-26072489

RESUMO

In this article, we described a new database framework to perform integrative "gene-set, network, and pathway analysis" (GNPA). In this framework, we integrated heterogeneous data on pathways, annotated list, and gene-sets (PAGs) into a PAG electronic repository (PAGER). PAGs in the PAGER database are organized into P-type, A-type and G-type PAGs with a three-letter-code standard naming convention. The PAGER database currently compiles 44 313 genes from 5 species including human, 38 663 PAGs, 324 830 gene-gene relationships and two types of 3 174 323 PAG-PAG regulatory relationships-co-membership based and regulatory relationship based. To help users assess each PAG's biological relevance, we developed a cohesion measure called Cohesion Coefficient (CoCo), which is capable of disambiguating between biologically significant PAGs and random PAGs with an area-under-curve performance of 0.98. PAGER database was set up to help users to search and retrieve PAGs from its online web interface. PAGER enable advanced users to build PAG-PAG regulatory networks that provide complementary biological insights not found in gene set analysis or individual gene network analysis. We provide a case study using cancer functional genomics data sets to demonstrate how integrative GNPA help improve network biology data coverage and therefore biological interpretability. The PAGER database can be accessible openly at http://discovery.informatics.iupui.edu/PAGER/.


Assuntos
Bases de Dados Genéticas , Redes Reguladoras de Genes , Humanos , Software
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA