Spatial transformation of multi-omics data unlocks novel insights into cancer biology

The application of next-generation sequencing (NGS) has transformed cancer research. As costs have decreased, NGS has increasingly been applied to generate multiple layers of molecular data from the same samples, covering genomics, transcriptomics, and methylomics. Integrating these types of multi-o...

Full description

Bibliographic Details
Main Authors: Mateo Sokač, Asbjørn Kjær, Lars Dyrskjøt, Benjamin Haibe-Kains, Hugo JWL Aerts, Nicolai J Birkbak
Format: Article
Language:English
Published: eLife Sciences Publications Ltd 2023-09-01
Series:eLife
Subjects:
Online Access:https://elifesciences.org/articles/87133
_version_ 1797692256756432896
author Mateo Sokač
Asbjørn Kjær
Lars Dyrskjøt
Benjamin Haibe-Kains
Hugo JWL Aerts
Nicolai J Birkbak
author_facet Mateo Sokač
Asbjørn Kjær
Lars Dyrskjøt
Benjamin Haibe-Kains
Hugo JWL Aerts
Nicolai J Birkbak
author_sort Mateo Sokač
collection DOAJ
description The application of next-generation sequencing (NGS) has transformed cancer research. As costs have decreased, NGS has increasingly been applied to generate multiple layers of molecular data from the same samples, covering genomics, transcriptomics, and methylomics. Integrating these types of multi-omics data in a combined analysis is now becoming a common issue with no obvious solution, often handled on an ad hoc basis, with multi-omics data arriving in a tabular format and analyzed using computationally intensive statistical methods. These methods particularly ignore the spatial orientation of the genome and often apply stringent p-value corrections that likely result in the loss of true positive associations. Here, we present GENIUS (GEnome traNsformatIon and spatial representation of mUltiomicS data), a framework for integrating multi-omics data using deep learning models developed for advanced image analysis. The GENIUS framework is able to transform multi-omics data into images with genes displayed as spatially connected pixels and successfully extract relevant information with respect to the desired output. We demonstrate the utility of GENIUS by applying the framework to multi-omics datasets from the Cancer Genome Atlas. Our results are focused on predicting the development of metastatic cancer from primary tumors, and demonstrate how through model inference, we are able to extract the genes which are driving the model prediction and are likely associated with metastatic disease progression. We anticipate our framework to be a starting point and strong proof of concept for multi-omics data transformation and analysis without the need for statistical correction.
first_indexed 2024-03-12T02:25:02Z
format Article
id doaj.art-5eb6910b14814b319ff2f70d309e251b
institution Directory Open Access Journal
issn 2050-084X
language English
last_indexed 2024-03-12T02:25:02Z
publishDate 2023-09-01
publisher eLife Sciences Publications Ltd
record_format Article
series eLife
spelling doaj.art-5eb6910b14814b319ff2f70d309e251b2023-09-05T16:47:40ZengeLife Sciences Publications LtdeLife2050-084X2023-09-011210.7554/eLife.87133Spatial transformation of multi-omics data unlocks novel insights into cancer biologyMateo Sokač0https://orcid.org/0000-0001-9896-1544Asbjørn Kjær1https://orcid.org/0009-0006-3307-0031Lars Dyrskjøt2Benjamin Haibe-Kains3Hugo JWL Aerts4Nicolai J Birkbak5https://orcid.org/0000-0003-1613-9587Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Bioinformatics Research Center, Aarhus University, Aarhus, DenmarkDepartment of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Bioinformatics Research Center, Aarhus University, Aarhus, DenmarkDepartment of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, DenmarkPrincess Margaret Cancer Centre, University Health Network, Temerty Faculty of Medicine, University of Toronto, Toronto, CanadaArtificial Intelligence in Medicine (AIM) Program, Mass General Brigham, Harvard Medical School, Boston, United States; Departments of Radiation Oncology and Radiology, Brigham and Women’s Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, United States; Radiology and Nuclear Medicine, CARIM & GROW, Maastricht University, Maastricht, NetherlandsDepartment of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Bioinformatics Research Center, Aarhus University, Aarhus, DenmarkThe application of next-generation sequencing (NGS) has transformed cancer research. As costs have decreased, NGS has increasingly been applied to generate multiple layers of molecular data from the same samples, covering genomics, transcriptomics, and methylomics. Integrating these types of multi-omics data in a combined analysis is now becoming a common issue with no obvious solution, often handled on an ad hoc basis, with multi-omics data arriving in a tabular format and analyzed using computationally intensive statistical methods. These methods particularly ignore the spatial orientation of the genome and often apply stringent p-value corrections that likely result in the loss of true positive associations. Here, we present GENIUS (GEnome traNsformatIon and spatial representation of mUltiomicS data), a framework for integrating multi-omics data using deep learning models developed for advanced image analysis. The GENIUS framework is able to transform multi-omics data into images with genes displayed as spatially connected pixels and successfully extract relevant information with respect to the desired output. We demonstrate the utility of GENIUS by applying the framework to multi-omics datasets from the Cancer Genome Atlas. Our results are focused on predicting the development of metastatic cancer from primary tumors, and demonstrate how through model inference, we are able to extract the genes which are driving the model prediction and are likely associated with metastatic disease progression. We anticipate our framework to be a starting point and strong proof of concept for multi-omics data transformation and analysis without the need for statistical correction.https://elifesciences.org/articles/87133deep learningmulti-omicscancerintegrated gradientsimmunotherapybladder cancer
spellingShingle Mateo Sokač
Asbjørn Kjær
Lars Dyrskjøt
Benjamin Haibe-Kains
Hugo JWL Aerts
Nicolai J Birkbak
Spatial transformation of multi-omics data unlocks novel insights into cancer biology
eLife
deep learning
multi-omics
cancer
integrated gradients
immunotherapy
bladder cancer
title Spatial transformation of multi-omics data unlocks novel insights into cancer biology
title_full Spatial transformation of multi-omics data unlocks novel insights into cancer biology
title_fullStr Spatial transformation of multi-omics data unlocks novel insights into cancer biology
title_full_unstemmed Spatial transformation of multi-omics data unlocks novel insights into cancer biology
title_short Spatial transformation of multi-omics data unlocks novel insights into cancer biology
title_sort spatial transformation of multi omics data unlocks novel insights into cancer biology
topic deep learning
multi-omics
cancer
integrated gradients
immunotherapy
bladder cancer
url https://elifesciences.org/articles/87133
work_keys_str_mv AT mateosokac spatialtransformationofmultiomicsdataunlocksnovelinsightsintocancerbiology
AT asbjørnkjær spatialtransformationofmultiomicsdataunlocksnovelinsightsintocancerbiology
AT larsdyrskjøt spatialtransformationofmultiomicsdataunlocksnovelinsightsintocancerbiology
AT benjaminhaibekains spatialtransformationofmultiomicsdataunlocksnovelinsightsintocancerbiology
AT hugojwlaerts spatialtransformationofmultiomicsdataunlocksnovelinsightsintocancerbiology
AT nicolaijbirkbak spatialtransformationofmultiomicsdataunlocksnovelinsightsintocancerbiology