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.
Nat Cancer ; 2024 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-38637658

RESUMO

Tailoring optimal treatment for individual cancer patients remains a significant challenge. To address this issue, we developed PERCEPTION (PERsonalized Single-Cell Expression-Based Planning for Treatments In ONcology), a precision oncology computational pipeline. Our approach uses publicly available matched bulk and single-cell (sc) expression profiles from large-scale cell-line drug screens. These profiles help build treatment response models based on patients' sc-tumor transcriptomics. PERCEPTION demonstrates success in predicting responses to targeted therapies in cultured and patient-tumor-derived primary cells, as well as in two clinical trials for multiple myeloma and breast cancer. It also captures the resistance development in patients with lung cancer treated with tyrosine kinase inhibitors. PERCEPTION outperforms published state-of-the-art sc-based and bulk-based predictors in all clinical cohorts. PERCEPTION is accessible at https://github.com/ruppinlab/PERCEPTION . Our work, showcasing patient stratification using sc-expression profiles of their tumors, will encourage the adoption of sc-omics profiling in clinical settings, enhancing precision oncology tools based on sc-omics.

2.
bioRxiv ; 2023 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-37886558

RESUMO

Immune checkpoint blockade (ICB) is a promising cancer therapy; however, resistance often develops. To learn more about ICB resistance mechanisms, we developed IRIS (Immunotherapy Resistance cell-cell Interaction Scanner), a machine learning model aimed at identifying candidate ligand-receptor interactions (LRI) that are likely to mediate ICB resistance in the tumor microenvironment (TME). We developed and applied IRIS to identify resistance-mediating cell-type-specific ligand-receptor interactions by analyzing deconvolved transcriptomics data of the five largest melanoma ICB therapy cohorts. This analysis identifies a set of specific ligand-receptor pairs that are deactivated as tumors develop resistance, which we refer to as resistance deactivated interactions (RDI). Quite strikingly, the activity of these RDIs in pre-treatment samples offers a markedly stronger predictive signal for ICB therapy response compared to those that are activated as tumors develop resistance. Their predictive accuracy surpasses the state-of-the-art published transcriptomics biomarker signatures across an array of melanoma ICB datasets. Many of these RDIs are involved in chemokine signaling. Indeed, we further validate on an independent large melanoma patient cohort that their activity is associated with CD8+ T cell infiltration and enriched in hot/brisk tumors. Taken together, this study presents a new strongly predictive ICB response biomarker signature, showing that following ICB treatment resistant tumors turn inhibit lymphocyte infiltration by deactivating specific key ligand-receptor interactions.

3.
J Biol Chem ; 297(3): 101023, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34343564

RESUMO

Ammonia is a cytotoxic molecule generated during normal cellular functions. Dysregulated ammonia metabolism, which is evident in many chronic diseases such as liver cirrhosis, heart failure, and chronic obstructive pulmonary disease, initiates a hyperammonemic stress response in tissues including skeletal muscle and in myotubes. Perturbations in levels of specific regulatory molecules have been reported, but the global responses to hyperammonemia are unclear. In this study, we used a multiomics approach to vertically integrate unbiased data generated using an assay for transposase-accessible chromatin with high-throughput sequencing, RNA-Seq, and proteomics. We then horizontally integrated these data across different models of hyperammonemia, including myotubes and mouse and human muscle tissues. Changes in chromatin accessibility and/or expression of genes resulted in distinct clusters of temporal molecular changes including transient, persistent, and delayed responses during hyperammonemia in myotubes. Known responses to hyperammonemia, including mitochondrial and oxidative dysfunction, protein homeostasis disruption, and oxidative stress pathway activation, were enriched in our datasets. During hyperammonemia, pathways that impact skeletal muscle structure and function that were consistently enriched were those that contribute to mitochondrial dysfunction, oxidative stress, and senescence. We made several novel observations, including an enrichment in antiapoptotic B-cell leukemia/lymphoma 2 family protein expression, increased calcium flux, and increased protein glycosylation in myotubes and muscle tissue upon hyperammonemia. Critical molecules in these pathways were validated experimentally. Human skeletal muscle from patients with cirrhosis displayed similar responses, establishing translational relevance. These data demonstrate complex molecular interactions during adaptive and maladaptive responses during the cellular stress response to hyperammonemia.


Assuntos
Genômica , Hiperamonemia/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Proteômica , Transcriptoma , Animais , Citometria de Fluxo , Humanos , Hiperamonemia/genética , Immunoblotting/métodos , Camundongos , Reação em Cadeia da Polimerase em Tempo Real , Reprodutibilidade dos Testes
4.
Nat Commun ; 11(1): 4391, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32873806

RESUMO

Deep learning with Convolutional Neural Networks has shown great promise in image-based classification and enhancement but is often unsuitable for predictive modeling using features without spatial correlations. We present a feature representation approach termed REFINED (REpresentation of Features as Images with NEighborhood Dependencies) to arrange high-dimensional vectors in a compact image form conducible for CNN-based deep learning. We consider the similarities between features to generate a concise feature map in the form of a two-dimensional image by minimizing the pairwise distance values following a Bayesian Metric Multidimensional Scaling Approach. We hypothesize that this approach enables embedded feature extraction and, integrated with CNN-based deep learning, can boost the predictive accuracy. We illustrate the superior predictive capabilities of the proposed framework as compared to state-of-the-art methodologies in drug sensitivity prediction scenarios using synthetic datasets, drug chemical descriptors as predictors from NCI60, and both transcriptomic information and drug descriptors as predictors from GDSC.


Assuntos
Antineoplásicos/farmacologia , Aprendizado Profundo , Processamento de Imagem Assistida por Computador/métodos , Neoplasias/tratamento farmacológico , Antineoplásicos/uso terapêutico , Teorema de Bayes , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Conjuntos de Dados como Assunto , Resistencia a Medicamentos Antineoplásicos , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Perfilação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Neoplasias/patologia , Análise de Sequência com Séries de Oligonucleotídeos
5.
Brief Bioinform ; 20(5): 1734-1753, 2019 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-31846027

RESUMO

Recent years have seen an increase in the availability of pharmacogenomic databases such as Genomics of Drug Sensitivity in Cancer (GDSC) and Cancer Cell Line Encyclopedia (CCLE) that provide genomic and functional characterization information for multiple cell lines. Studies have alluded to the fact that specific characterizations may be inconsistent between different databases. Analysis of the potential discrepancies in the different databases is highly significant, as these sources are frequently used to analyze and validate methodologies for personalized cancer therapies. In this article, we review the recent developments in investigating the correspondence between different pharmacogenomics databases and discuss the potential factors that require attention when incorporating these sources in any modeling analysis. Furthermore, we explored the consistency among these databases using copulas that can capture nonlinear dependencies between two sets of data.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/genética , Farmacogenética , Linhagem Celular Tumoral , Bases de Dados Genéticas , Humanos , Neoplasias/patologia
6.
BMC Bioinformatics ; 20(Suppl 12): 317, 2019 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-31216980

RESUMO

BACKGROUND: Clinical studies often track dose-response curves of subjects over time. One can easily model the dose-response curve at each time point with Hill equation, but such a model fails to capture the temporal evolution of the curves. On the other hand, one can use Gompertz equation to model the temporal behaviors at each dose without capturing the evolution of time curves across dosage. RESULTS: In this article, we propose a parametric model for dose-time responses that follows Gompertz law in time and Hill equation across dose approximately. We derive a recursion relation for dose-response curves over time capturing the temporal evolution and then specify a regression model connecting the parameters controlling the dose-time responses with individual level proteomic data. The resultant joint model allows us to predict the dose-response curves over time for new individuals. CONCLUSION: We have compared the efficacy of our proposed Recursive Hybrid model with individual dose-response predictive models at desired time points. We note that our proposed model exhibits a superior performance compared to the individual ones for both synthetic data and actual pharmacological data. For the desired dose-time varying genetic characterization and drug response values, we have used the HMS-LINCS database and demonstrated the effectiveness of our model for all available anticancer compounds.


Assuntos
Modelos Teóricos , Farmacologia , Simulação por Computador , Bases de Dados como Assunto , Relação Dose-Resposta a Droga , Humanos , Fatores de Tempo
7.
Sci Rep ; 9(1): 1628, 2019 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-30733524

RESUMO

Drug sensitivity prediction for individual tumors is a significant challenge in personalized medicine. Current modeling approaches consider prediction of a single metric of the drug response curve such as AUC or IC50. However, the single summary metric of a dose-response curve fails to provide the entire drug sensitivity profile which can be used to design the optimal dose for a patient. In this article, we assess the problem of predicting the complete dose-response curve based on genetic characterizations. We propose an enhancement to the popular ensemble-based Random Forests approach that can directly predict the entire functional profile of a dose-response curve rather than a single summary metric. We design functional regression trees with node costs modified based on dose/response region dependence methodologies and response distribution based approaches. Our results relative to large pharmacological databases such as CCLE and GDSC show a higher accuracy in predicting dose-response curves of the proposed functional framework in contrast to univariate or multivariate Random Forest predicting sensitivities at different dose levels. Furthermore, we also considered the problem of predicting functional responses from functional predictors i.e., estimating the dose-response curves with a model built on dose-dependent expression data. The superior performance of Functional Random Forest using functional data as compared to existing approaches have been shown using the HMS-LINCS dataset. In summary, Functional Random Forest presents an enhanced predictive modeling framework to predict the entire functional response profile considering both static and functional predictors instead of predicting the summary metrics of the response curves.


Assuntos
Relação Dose-Resposta a Droga , Modelos Teóricos , Área Sob a Curva , Linhagem Celular , Bases de Dados de Produtos Farmacêuticos , Humanos , Análise Multivariada , Neoplasias/tratamento farmacológico , Neoplasias/genética , Análise de Regressão , Reprodutibilidade dos Testes
8.
BMC Bioinformatics ; 19(Suppl 17): 497, 2018 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-30591023

RESUMO

BACKGROUND: In precision medicine, scarcity of suitable biological data often hinders the design of an appropriate predictive model. In this regard, large scale pharmacogenomics studies, like CCLE and GDSC hold the promise to mitigate the issue. However, one cannot directly employ data from multiple sources together due to the existing distribution shift in data. One way to solve this problem is to utilize the transfer learning methodologies tailored to fit in this specific context. RESULTS: In this paper, we present two novel approaches for incorporating information from a secondary database for improving the prediction in a target database. The first approach is based on latent variable cost optimization and the second approach considers polynomial mapping between the two databases. Utilizing CCLE and GDSC databases, we illustrate that the proposed approaches accomplish a better prediction of drug sensitivities for different scenarios as compared to the existing approaches. CONCLUSION: We have compared the performance of the proposed predictive models with database-specific individual models as well as existing transfer learning approaches. We note that our proposed approaches exhibit superior performance compared to the abovementioned alternative techniques for predicting sensitivity for different anti-cancer compounds, particularly the nonlinear mapping model shows the best overall performance.


Assuntos
Algoritmos , Antineoplásicos/uso terapêutico , Neoplasias/tratamento farmacológico , Área Sob a Curva , Bases de Dados Factuais , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias/genética
9.
Annu Int Conf IEEE Eng Med Biol Soc ; 2018: 1246-1249, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30440616

RESUMO

Recent years have observed a number of Pharmacogenomics databases being published that enable testing of various predictive modeling techniques for personalized therapy applications. However, the consistencies between the databases are usually limited in spite of having significant number of common cell lines and drugs. In this article, we consider the problem of whether we can use the model learned from one secondary database to improve the prediction for the other target database. We illustrate using two pharmacogenomics databases that representing the databases using common basis vectors can improve prediction performance as compared to the naive application of a model trained on one database to another. We also elucidate the robustness of using PCA based basis vectors for scenarios with low correlated input features.


Assuntos
Farmacogenética , Bases de Dados Factuais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...