Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
NPJ Precis Oncol ; 5(1): 71, 2021 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-34302041

RESUMO

The FDA recently approved eight targeted therapies for acute myeloid leukemia (AML), including the BCL-2 inhibitor venetoclax. Maximizing efficacy of these treatments requires refining patient selection. To this end, we analyzed two recent AML studies profiling the gene expression and ex vivo drug response of primary patient samples. We find that ex vivo samples often exhibit a general sensitivity to (any) drug exposure, independent of drug target. We observe that this "general response across drugs" (GRD) is associated with FLT3-ITD mutations, clinical response to standard induction chemotherapy, and overall survival. Further, incorporating GRD into expression-based regression models trained on one of the studies improved their performance in predicting ex vivo response in the second study, thus signifying its relevance to precision oncology efforts. We find that venetoclax response is independent of GRD but instead show that it is linked to expression of monocyte-associated genes by developing and applying a multi-source Bayesian regression approach. The method shares information across studies to robustly identify biomarkers of drug response and is broadly applicable in integrative analyses.

2.
Nicotine Tob Res ; 23(11): 1869-1879, 2021 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-33991191

RESUMO

INTRODUCTION: The availability of a variety of e-cigarettes flavors is one of the frequently cited reasons for their adoption. An active stream of discussion about flavoring can be observed online. Analyzing these real-time conversations offers nuanced insights into key factors related to the adoption of flavors, subsequently supporting public health interventions. METHODS: Google's BERT, a state-of-the-art deep learning method was employed to model the first sentiment corpus on JUUL flavors. BERT, which is pre-trained with the complete English Wikipedia was fine-tuned by integrating a classification model, with human labeled Tweets, as training data. A collection of 30 075 Tweets about JUUL flavors was classified into positive and negative sentiments. Finally, using topic models, we identify and grouped thematic areas into positive and negative Tweets. RESULTS: With an average of 89% cross-validation precision for classifying Tweets, the fine-tuned BERT model classified 24 114 Tweets as positive and 5961 Tweets as negative. Through the topic modeling approach 10 thematic topics were identified from the predicted positive and negative sentiments expressed in the Tweets. CONCLUSIONS: JUUL flavors, notably mango, mint, and cucumber, provoke overwhelmingly positive sentiments indicating a strong likeness due to favorable taste and odor. Negative discourse about JUUL flavors revolve around addictiveness, high nicotine content, and youth targeted marketing. IMPLICATIONS: Limiting the content related to flavors and positive perceptions on social media is necessary to minimize exposure to youth. The novel methodology used in this study may be adopted to monitor e-cigarette discourse periodically, as well as other critical public health phenomena online.


Assuntos
Sistemas Eletrônicos de Liberação de Nicotina , Mídias Sociais , Adolescente , Aromatizantes , Humanos , Aprendizado de Máquina , Paladar
3.
Bioinformatics ; 33(14): i359-i368, 2017 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-28881998

RESUMO

MOTIVATION: A prime challenge in precision cancer medicine is to identify genomic and molecular features that are predictive of drug treatment responses in cancer cells. Although there are several computational models for accurate drug response prediction, these often lack the ability to infer which feature combinations are the most predictive, particularly for high-dimensional molecular datasets. As increasing amounts of diverse genome-wide data sources are becoming available, there is a need to build new computational models that can effectively combine these data sources and identify maximally predictive feature combinations. RESULTS: We present a novel approach that leverages on systematic integration of data sources to identify response predictive features of multiple drugs. To solve the modeling task we implement a Bayesian linear regression method. To further improve the usefulness of the proposed model, we exploit the known human cancer kinome for identifying biologically relevant feature combinations. In case studies with a synthetic dataset and two publicly available cancer cell line datasets, we demonstrate the improved accuracy of our method compared to the widely used approaches in drug response analysis. As key examples, our model identifies meaningful combinations of features for the well known EGFR, ALK, PLK and PDGFR inhibitors. AVAILABILITY AND IMPLEMENTATION: The source code of the method is available at https://github.com/suleimank/mvlr . CONTACT: muhammad.ammad-ud-din@helsinki.fi or suleiman.khan@helsinki.fi. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Antineoplásicos/uso terapêutico , Biologia Computacional/métodos , Modelos Biológicos , Neoplasias/tratamento farmacológico , Medicina de Precisão/métodos , Software , Algoritmos , Antineoplásicos/farmacologia , Teorema de Bayes , Humanos , Modelos Lineares , Neoplasias/genética , Neoplasias/metabolismo , Transdução de Sinais/efeitos dos fármacos
4.
Bioinformatics ; 32(17): i455-i463, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27587662

RESUMO

MOTIVATION: A key goal of computational personalized medicine is to systematically utilize genomic and other molecular features of samples to predict drug responses for a previously unseen sample. Such predictions are valuable for developing hypotheses for selecting therapies tailored for individual patients. This is especially valuable in oncology, where molecular and genetic heterogeneity of the cells has a major impact on the response. However, the prediction task is extremely challenging, raising the need for methods that can effectively model and predict drug responses. RESULTS: In this study, we propose a novel formulation of multi-task matrix factorization that allows selective data integration for predicting drug responses. To solve the modeling task, we extend the state-of-the-art kernelized Bayesian matrix factorization (KBMF) method with component-wise multiple kernel learning. In addition, our approach exploits the known pathway information in a novel and biologically meaningful fashion to learn the drug response associations. Our method quantitatively outperforms the state of the art on predicting drug responses in two publicly available cancer datasets as well as on a synthetic dataset. In addition, we validated our model predictions with lab experiments using an in-house cancer cell line panel. We finally show the practical applicability of the proposed method by utilizing prior knowledge to infer pathway-drug response associations, opening up the opportunity for elucidating drug action mechanisms. We demonstrate that pathway-response associations can be learned by the proposed model for the well-known EGFR and MEK inhibitors. AVAILABILITY AND IMPLEMENTATION: The source code implementing the method is available at http://research.cs.aalto.fi/pml/software/cwkbmf/ CONTACTS: muhammad.ammad-ud-din@aalto.fi or samuel.kaski@aalto.fi SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Genômica , Neoplasias , Algoritmos , Teorema de Bayes , Sistemas de Liberação de Medicamentos , Descoberta de Drogas , Humanos , Redes e Vias Metabólicas , Software
5.
Nat Commun ; 7: 12460, 2016 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-27549343

RESUMO

Rheumatoid arthritis (RA) affects millions world-wide. While anti-TNF treatment is widely used to reduce disease progression, treatment fails in ∼one-third of patients. No biomarker currently exists that identifies non-responders before treatment. A rigorous community-based assessment of the utility of SNP data for predicting anti-TNF treatment efficacy in RA patients was performed in the context of a DREAM Challenge (http://www.synapse.org/RA_Challenge). An open challenge framework enabled the comparative evaluation of predictions developed by 73 research groups using the most comprehensive available data and covering a wide range of state-of-the-art modelling methodologies. Despite a significant genetic heritability estimate of treatment non-response trait (h(2)=0.18, P value=0.02), no significant genetic contribution to prediction accuracy is observed. Results formally confirm the expectations of the rheumatology community that SNP information does not significantly improve predictive performance relative to standard clinical traits, thereby justifying a refocusing of future efforts on collection of other data.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Artrite Reumatoide/tratamento farmacológico , Predisposição Genética para Doença/genética , Polimorfismo de Nucleotídeo Único , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Adulto , Idoso , Anticorpos Monoclonais/uso terapêutico , Antirreumáticos/uso terapêutico , Artrite Reumatoide/genética , Artrite Reumatoide/patologia , Certolizumab Pegol/uso terapêutico , Estudos de Coortes , Crowdsourcing , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Resultado do Tratamento , Fator de Necrose Tumoral alfa/imunologia
6.
J Chem Inf Model ; 54(8): 2347-59, 2014 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-25046554

RESUMO

With data from recent large-scale drug sensitivity measurement campaigns, it is now possible to build and test models predicting responses for more than one hundred anticancer drugs against several hundreds of human cancer cell lines. Traditional quantitative structure-activity relationship (QSAR) approaches focus on small molecules in searching for their structural properties predictive of the biological activity in a single cell line or a single tissue type. We extend this line of research in two directions: (1) an integrative QSAR approach predicting the responses to new drugs for a panel of multiple known cancer cell lines simultaneously and (2) a personalized QSAR approach predicting the responses to new drugs for new cancer cell lines. To solve the modeling task, we apply a novel kernelized Bayesian matrix factorization method. For maximum applicability and predictive performance, the method optionally utilizes genomic features of cell lines and target information on drugs in addition to chemical drug descriptors. In a case study with 116 anticancer drugs and 650 cell lines, we demonstrate the usefulness of the method in several relevant prediction scenarios, differing in the amount of available information, and analyze the importance of various types of drug features for the response prediction. Furthermore, after predicting the missing values of the data set, a complete global map of drug response is explored to assess treatment potential and treatment range of therapeutically interesting anticancer drugs.


Assuntos
Antineoplásicos/farmacologia , Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/genética , Relação Quantitativa Estrutura-Atividade , Bibliotecas de Moléculas Pequenas/farmacologia , Antineoplásicos/química , Teorema de Bayes , Biomarcadores Farmacológicos , Linhagem Celular Tumoral , Análise Fatorial , Humanos , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/metabolismo , Bibliotecas de Moléculas Pequenas/química
7.
Nat Biotechnol ; 32(12): 1202-12, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24880487

RESUMO

Predicting the best treatment strategy from genomic information is a core goal of precision medicine. Here we focus on predicting drug response based on a cohort of genomic, epigenomic and proteomic profiling data sets measured in human breast cancer cell lines. Through a collaborative effort between the National Cancer Institute (NCI) and the Dialogue on Reverse Engineering Assessment and Methods (DREAM) project, we analyzed a total of 44 drug sensitivity prediction algorithms. The top-performing approaches modeled nonlinear relationships and incorporated biological pathway information. We found that gene expression microarrays consistently provided the best predictive power of the individual profiling data sets; however, performance was increased by including multiple, independent data sets. We discuss the innovations underlying the top-performing methodology, Bayesian multitask MKL, and we provide detailed descriptions of all methods. This study establishes benchmarks for drug sensitivity prediction and identifies approaches that can be leveraged for the development of new methods.


Assuntos
Antineoplásicos/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/genética , Perfilação da Expressão Gênica , Neoplasias/tratamento farmacológico , Algoritmos , Antineoplásicos/efeitos adversos , Epigenômica/métodos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genômica/métodos , Humanos , Neoplasias/genética , Proteômica/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA