scholarly journals Constructing cancer patient-specific and group-specific gene networks with multi-omics data

2020 ◽  
Vol 13 (S6) ◽  
Author(s):  
Wook Lee ◽  
De-Shuang Huang ◽  
Kyungsook Han
2021 ◽  
Author(s):  
Mai Adachi Nakazawa ◽  
Yoshinori Tamada ◽  
Yoshihisa Tanaka ◽  
Marie Ikeguchi ◽  
Kako Higashihara ◽  
...  

The identification of cancer subtypes is important for the understanding of tumor heterogeneity. In recent years, numerous computational methods have been proposed for this problem based on the multi-omics data of patients. It is widely accepted that different cancer subtypes are induced by different molecular regulatory networks. However, only a few incorporate the differences between their molecular systems into the classification processes. In this study, we present a novel method to classify cancer subtypes based on patient-specific molecular systems. Our method quantifies patient-specific gene networks, which are estimated from their transcriptome data. By clustering their quantified networks, our method allows for cancer subtyping, taking into consideration the differences in the molecular systems of patients. Comprehensive analyses of The Cancer Genome Atlas (TCGA) datasets applied to our method confirmed that they were able to identify more clinically meaningful cancer subtypes than the existing subtypes and found that the identified subtypes comprised different molecular features. Our findings show that the proposed method, based on a simple classification using the patient-specific molecular systems, can identify cancer subtypes even with single omics data, which cannot otherwise be captured by existing methods using multi-omics data.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Mai Adachi Nakazawa ◽  
Yoshinori Tamada ◽  
Yoshihisa Tanaka ◽  
Marie Ikeguchi ◽  
Kako Higashihara ◽  
...  

AbstractThe identification of cancer subtypes is important for the understanding of tumor heterogeneity. In recent years, numerous computational methods have been proposed for this problem based on the multi-omics data of patients. It is widely accepted that different cancer subtypes are induced by different molecular regulatory networks. However, only a few incorporate the differences between their molecular systems into the identification processes. In this study, we present a novel method to identify cancer subtypes based on patient-specific molecular systems. Our method realizes this by quantifying patient-specific gene networks, which are estimated from their transcriptome data, and by clustering their quantified networks. Comprehensive analyses of The Cancer Genome Atlas (TCGA) datasets applied to our method confirmed that they were able to identify more clinically meaningful cancer subtypes than the existing subtypes and found that the identified subtypes comprised different molecular features. Our findings also show that the proposed method can identify the novel cancer subtypes even with single omics data, which cannot otherwise be captured by existing methods using multi-omics data.


Author(s):  
Minsik Oh ◽  
Sungjoon Park ◽  
Sun Kim ◽  
Heejoon Chae

Abstract Gene expressions are subtly regulated by quantifiable measures of genetic molecules such as interaction with other genes, methylation, mutations, transcription factor and histone modifications. Integrative analysis of multi-omics data can help scientists understand the condition or patient-specific gene regulation mechanisms. However, analysis of multi-omics data is challenging since it requires not only the analysis of multiple omics data sets but also mining complex relations among different genetic molecules by using state-of-the-art machine learning methods. In addition, analysis of multi-omics data needs quite large computing infrastructure. Moreover, interpretation of the analysis results requires collaboration among many scientists, often requiring reperforming analysis from different perspectives. Many of the aforementioned technical issues can be nicely handled when machine learning tools are deployed on the cloud. In this survey article, we first survey machine learning methods that can be used for gene regulation study, and we categorize them according to five different goals: gene regulatory subnetwork discovery, disease subtype analysis, survival analysis, clinical prediction and visualization. We also summarize the methods in terms of multi-omics input types. Then, we explain why the cloud is potentially a good solution for the analysis of multi-omics data, followed by a survey of two state-of-the-art cloud systems, Galaxy and BioVLAB. Finally, we discuss important issues when the cloud is used for the analysis of multi-omics data for the gene regulation study.


2019 ◽  
Vol 12 (S8) ◽  
Author(s):  
Byungkyu Park ◽  
Wook Lee ◽  
Inhee Park ◽  
Kyungsook Han

Abstract Background Molecular characterization of individual cancer patients is important because cancer is a complex and heterogeneous disease with many possible genetic and environmental causes. Many studies have been conducted to identify diagnostic or prognostic gene signatures for cancer from gene expression profiles. However, some gene signatures may fail to serve as diagnostic or prognostic biomarkers and gene signatures may not be found in gene expression profiles. Methods In this study, we developed a general method for constructing patient-specific gene correlation networks and for identifying prognostic gene pairs from the networks. A patient-specific gene correlation network was constructed by comparing a reference gene correlation network from normal samples to a network perturbed by a single patient sample. The main difference of our method from previous ones includes (1) it is focused on finding prognostic gene pairs rather than prognostic genes and (2) it can identify prognostic gene pairs from gene expression profiles even when no significant prognostic genes exist. Results Evaluation of our method with extensive data sets of three cancer types (breast invasive carcinoma, colon adenocarcinoma, and lung adenocarcinoma) showed that our approach is general and that gene pairs can serve as more reliable prognostic signatures for cancer than genes. Conclusions Our study revealed that prognosis of individual cancer patients is associated with the existence of prognostic gene pairs in the patient-specific network and the size of a subnetwork of the prognostic gene pairs in the patient-specific network. Although preliminary, our approach will be useful for finding gene pairs to predict survival time of patients and to tailor treatments to individual characteristics. The program for dynamically constructing patient-specific gene networks and for finding prognostic gene pairs is available at http://bclab.inha.ac.kr/pancancer.


2015 ◽  
Author(s):  
Vasileios Stathias ◽  
Chiara Pastori ◽  
Ricardo Komotar ◽  
Ming Zhang ◽  
Stephan Schürer ◽  
...  

2021 ◽  
Author(s):  
Adriaan-Alexander Ludl ◽  
Tom Michoel

Causal gene networks model the flow of information within a cell. Reconstructing causal networks from omics data is challenging because correlation does not imply causation. When genomics and transcriptomics data...


2021 ◽  
Vol 0 (0) ◽  
Author(s):  
Shinjo Yada

Abstract Cancer tissue samples obtained via biopsy or surgery were examined for specific gene mutations by genetic testing to inform treatment. Precision medicine, which considers not only the cancer type and location, but also the genetic information, environment, and lifestyle of each patient, can be applied for disease prevention and treatment in individual patients. The number of patient-specific characteristics, including biomarkers, has been increasing with time; these characteristics are highly correlated with outcomes. The number of patients at the beginning of early-phase clinical trials is often limited. Moreover, it is challenging to estimate parameters of models that include baseline characteristics as covariates such as biomarkers. To overcome these issues and promote personalized medicine, we propose a dose-finding method that considers patient background characteristics, including biomarkers, using a model for phase I/II oncology trials. We built a Bayesian neural network with input variables of dose, biomarkers, and interactions between dose and biomarkers and output variables of efficacy outcomes for each patient. We trained the neural network to select the optimal dose based on all background characteristics of a patient. Simulation analysis showed that the probability of selecting the desirable dose was higher using the proposed method than that using the naïve method.


Sign in / Sign up

Export Citation Format

Share Document