1. Introduction
The omics high-throughput pipeline is transforming biological research by enabling comprehensive, large-scale analysis of diverse biomolecular data. These advanced technologies generate extensive data at multiple omics levels, including genomics, transcriptomics, proteomics and metabolomics. To handle the complexity and volume of this data, sophisticated bioinformatics pipelines are required that integrate various software tools and databases to pre-process, analyze and interpret the data, forming intricate workflows (
Figure 1). The integration of high-throughput omics is primarily based on two fundamental approaches: similarity-based methods and difference-based methods. Similarity-based methods aim to identify common patterns, correlations and common paths in different omics datasets. These methods are crucial for understanding overarching biological processes and identifying universal biomarkers. For example, correlation analysis evaluates the correlation between different omics levels (e.g., genomics, transcriptomics and proteomics) to find common trends and relationships and identify co-expressed genes or proteins in different datasets. Clustering algorithms, such as hierarchical clustering and k-means clustering, group similar data points from different omics datasets and uncover modules or networks of genes and proteins that work together. Network-based approaches, such as Similarity Network Fusion (SNF), construct similarity networks for each omics type and then integrate them into a single network, merging information from all omics levels to highlight commonalities and identify important biological pathways. On the other hand, difference-based methods focus on detecting unique features and variations between different omics levels, which is essential for understanding disease-specific mechanisms and for personalized medicine. Differential expression analysis compares the expression levels of genes or proteins between different states (e.g., healthy vs. diseased) to identify significant changes and recognize unique molecular signatures associated with specific conditions [
1]. Variance decomposition decomposes the total variance observed in the data into components attributable to different omics levels. This helps us to understand the contribution of each omics type to the overall variability and to identify omics-specific variation. Feature selection methods, such as LASSO (Least Absolute Shrinkage and Selection Operator) and Random Forests, select the most relevant features from each omics dataset and integrates these features into a comprehensive model that captures the unique aspects of each layer [
2]. Popular integration algorithms include Multi-Omics Factor Analysis (MOFA) and Canonical Correlation Analysis (CCA). MOFA is an unsupervised approach that uses Bayesian factor analysis to identify latent factors responsible for variation in multiple omics datasets and integrates the data to identify underlying biological signals. CCA identifies linear relationships between two or more omics datasets, facilitating the discovery of correlated traits and common pathways [
3].
In genomic analysis (
Table 1), tools such as Ensembl (
https://www.ensembl.org/, accessed on 1 June 2024) and Galaxy [
4] are essential. Ensembl provides comprehensive genomic data, while Galaxy [
4] offers a user-friendly platform for bioinformatics workflows, including genome assembly, variant calling, transcriptomics and epigenomic analysis. For multi-omics data integration and visualization, OmicsNet and NetworkAnalyst are invaluable. OmicsNet [
5] supports the integration of genomics, transcriptomics, proteomics and metabolomics data to create comprehensive biological networks. NetworkAnalyst [
6] provides data filtering, normalization, statistical analysis and network visualization capabilities. These tools enable researchers to uncover new pathways and molecular mechanisms, driving advancements in precision medicine and other fields.
The integration of high-throughput omics combines data from different omics technologies to gain a comprehensive understanding of biological systems (
Figure 1,
Table 1). This integration is essential to unravel the complexity of cellular processes and disease mechanisms. This review explores advanced technologies and computational methods that facilitate omics integration, covering platforms such as next-generation sequencing (NGS) for genomics, RNA sequencing (RNA-Seq) for transcriptomics, mass spectrometry for proteomics and nuclear magnetic resonance (NMR) spectroscopy for metabolomics. It addresses challenges such as heterogeneity, scale and standardization in data and proposes solutions such as advanced bioinformatics tools and machine learning techniques. Key applications include automated text mining techniques such as natural language processing (NLP) to extract meaningful information from scientific literature and genomic analyses to identify biomarkers for diseases to improve diagnostic tools and personalized medicine. Integrating data from resources such as the GWAS catalog helps identify genetic variants associated with different traits, supporting biomarker discovery and therapeutic targets. Proteomics, facilitated by mass spectrometry, provides insights into protein functions and interactions, and the integration of proteomics data with other omics datasets improves our understanding of disease mechanisms. Effective integration strategies, such as early, mixed, middle, late and hierarchical integration, are essential for comprehensive insights into complex biological systems.
2. Comprehensive Frameworks for High-Throughput Pipeline Omics Integration
High-throughput omics integration pipelines focus on the development of comprehensive frameworks for efficiently processing, analyzing and interpreting large amounts of biological data generated by genomics, transcriptomics, proteomics and metabolomics technologies [
7]. These integration pipelines address the challenges posed by the heterogeneity and complexity of multi-omics data, ensuring effective combination and meaningful insights. Advanced computational methods and bioinformatics tools are essential for this integration. Platforms such as OmicsNet and NetworkAnalyst [
6] are critical for managing and analyzing multi-omics data. OmicsNet facilitates the visual analysis of biological networks by integrating genomics, transcriptomics, proteomics and metabolomics data and provides an intuitive user interface and extensive visualization options. NetworkAnalyst [
6] provides robust tools for network-based visual analysis that support transcriptomics, proteomics and metabolomics data and include features for data filtering, normalization, statistical analysis and network visualization, all accessible without programming knowledge.
These integrated pipelines streamline workflows from data acquisition to analysis, promoting the discovery of complex biological relationships and pathways. By building detailed molecular networks, researchers gain deeper insights into cellular functions and disease mechanisms, facilitating the identification of novel biomarkers and therapeutic targets and ultimately advancing precision medicine [
8]. In addition, integrated multi-omics pipelines translate high-dimensional biological data into actionable knowledge by enabling the simultaneous investigation of different molecular layers and providing a holistic view of biological systems. This integrative approach is particularly valuable in the study of diseases such as cancer, where understanding the interplay between genetic mutations, changes in gene expression, protein modifications and metabolic shifts is critical to the development of effective treatments. These advanced pipelines also improve the reproducibility and accessibility of next-generation sequencing analyses, benefiting a wide range of research applications (
Table 2) [
9,
10].
2.1. Key Components and Technologies
High-throughput omics technologies have revolutionized biological research by enabling comprehensive analysis of the molecular components in cells. These technologies rely on several advanced methods and key components, each of which critical to capturing the complexity of biological systems. At the center of these technologies are data generation platforms, such as next-generation sequencing (NGS) for genomics, RNA sequencing (RNA-Seq) for transcriptomics, mass spectrometry for proteomics and nuclear magnetic resonance (NMR) spectroscopy for metabolomics. These platforms enable the rapid and large-scale collection of data on different molecular entities, providing a detailed view of the biological landscape [
9,
10].
NGS has revolutionized genomics by enabling the rapid sequencing of entire genomes at high speed and low cost. This technology is critical for identifying genetic variation, understanding the contribution of genes to disease and exploring evolutionary relationships. RNA-Seq provides comprehensive gene expression data by sequencing the RNA present in a sample and is therefore essential for transcriptomic studies. It enables researchers to quantify the level of gene expression, identify splice variants and investigate gene regulatory networks. In proteomics, mass spectrometry is the cornerstone technology for identifying and quantifying proteins in complex mixtures. It reveals changes in protein expression, post-translational modifications and protein–protein interactions, providing insights into cellular functions and signaling pathways. NMR spectroscopy, which is essential in metabolomics, provides detailed insights into the mechanisms of small molecule metabolites present in biological samples. It enables the investigation of metabolic pathways, interactions and changes associated with various physiological and pathological conditions [
11].
In addition to data generation, high-throughput omics requires a robust computational infrastructure for data analysis and integration. The extensive and heterogeneous nature of omics data requires advanced computational resources and bioinformatics frameworks capable of processing, integrating and visualizing this data. Platforms such as G-language Genome Analysis Environment [
12] and anvi’o [
13] provide comprehensive tools for gene prediction, pathway mapping and interactive data visualization. These platforms are essential for managing large-scale omics data and enable researchers to extract meaningful insights from complex datasets. The development of new computational techniques further enhances the ability to integrate and interpret complex omics datasets. Techniques such as blockwise sparse principal component analysis and network-based methods overcome challenges such as variable redundancy and computational instability. These methods ensure accurate and reliable data integration, enabling researchers to construct detailed molecular networks and gain new biological insights [
14,
15]. For example, the use of advanced clustering algorithms and network-based approaches, such as Similarity Network Fusion (SNF), enables the integration of multi-omics data into a unified framework that highlights common pathways and interactions across different molecular layers [
16,
17].
Effective data integration strategies are critical to advancing our understanding of complex biological systems. Integrative tools such as OmicsNet [
18] and NetworkAnalyst [
6] play a central role in managing and analyzing multi-omics data. OmicsNet facilitates the visual analysis of biological networks and supports the integration of genomics, transcriptomics, proteomics and metabolomics data to create comprehensive biological networks. Its intuitive user interface and extensive visualization options make it accessible to researchers, enabling them to explore complex interactions within biological systems. NetworkAnalyst, on the other hand, offers powerful features for network-based visual analysis of multi-omics data. This platform supports the integration of data from different omics layers and provides robust tools for data filtering, normalization, statistical analysis and network visualization. Thanks to its user-friendly design and extensive online tutorials, NetworkAnalyst is accessible to researchers with varying levels of computer literacy.
These integrative tools not only streamline the workflow from data acquisition to analysis, but also greatly enhance the discovery of complex biological relationships and pathways. By constructing detailed molecular networks, researchers can gain a deeper understanding of cell function and disease mechanisms. This comprehensive approach facilitates the identification of novel biomarkers and therapeutic targets, ultimately advancing precision medicine and other fields.
Table 2.
Concept And Need of High-Throughput Omics.
Table 2.
Concept And Need of High-Throughput Omics.
Aspect | Description | Examples | References |
---|
Concept | High-throughput omics technologies encompass genomics, transcriptomics, proteomics and metabolomics, enabling comprehensive analysis of molecular components. | Genomics: DNA sequencing, Transcriptomics: RNA sequencing, Proteomics: Mass spectrometry, Metabolomics: NMR spectroscopy. | [19] |
Need | The complexity and heterogeneity of biological systems necessitate advanced methods to capture molecular interactions and dynamics. | Multifactorial diseases like cancer require comprehensive data to understand gene–protein–metabolite interactions. | [20] |
Benefits | Provides detailed views of biological systems, identifies novel biomarkers and facilitates personalized medicine. | Improves disease understanding, targeted therapeutic strategies and customized treatment plans. | [21] |
Challenges | Managing and integrating vast, heterogeneous datasets and developing accurate computational models. | Data heterogeneity, computational resource requirements and the need for advanced bioinformatics tools. | [21] |
2.2. Challenges and Opportunities
High-throughput omics technologies offer unprecedented opportunities to advance biological research, but they also pose significant challenges. One of the biggest challenges is integrating and interpreting the vast and heterogeneous datasets that these technologies generate. The complexity and scale of omics data require sophisticated computational methods and bioinformatics infrastructures to effectively process and understand the information. Issues such as data heterogeneity, noise and the lack of standardized data formats further complicate the integration process. Advanced computational solutions such as Omics Integrator software and network-based approaches have been developed to overcome these challenges by integrating different datasets and uncovering molecular pathways. However, a critical bottleneck for many researchers remains the need for improved training in data analysis and bioinformatics. Despite these challenges, high-throughput omics technologies offer numerous opportunities for transformative advances in biological research and personalized medicine [
22]. The integration of multi-omics data provides a more comprehensive understanding of biological systems and enables the identification of new biomarkers and therapeutic targets. This integrative approach is particularly valuable in the study of complex diseases such as cancer, where understanding the interactions between different molecular components is crucial. In addition, the development of cloud computing and big data analytics offers promising solutions for storing, analyzing and sharing omics data on a large scale [
23]. These technological advances facilitate the creation of detailed molecular atlases and the development of predictive models that can improve clinical outcomes and advance the field of precision medicine (
Table 3).
Over the past decade, omics technology has undergone significant advancements, evolving from its initial focus on cataloguing genes, proteins and SNPs to performing disease-specific, in-depth analyses of various aspects of genomics, including meta-genetics, protein–protein interactions, modifications and pathway mapping. Large-scale genome-wide association studies and high-throughput techniques have become more efficient and productive in exploring previously uncharted biological systems [
24]. Discovery approaches now utilize multiplex technologies, such as rapid sequencing and advanced mass spectrometry instruments, with better resolution to detect complex protein mixtures. Targeted approaches that use mass spectrometry technology to monitor multiple reactions enable rapid quantification of multiple peptides without the need for antibodies. Despite its complexity, omics technology has brought us closer to understanding the final clinical phenotype in complex diseases where multiple genes, organs and environmental influences are likely to be involved [
11].
Translational research in disease greatly benefits from omics approaches, as these technologies can map entire cellular–molecular pathways to guide in vivo studies and validate bedside research. The interrelated issues of reproducibility, noise and the perception of omics as “pure fishing” are important to address. The problem of reproducibility stems from noise, and the existence of noisy datasets that cannot be reproduced, fuels this perception. Careful experiment design, targeted biological questions and appropriate interpretation and validation of results are needed to address this criticism. The core problem with omics methodology is reproducibility. A healthy skepticism about reproducibility has driven genomics research and led to the derivation of training sets from well-characterized cohorts and validation in separate cohorts. However, proteomics presents unique challenges due to its inherent diversity and instability compared to the genome. Proteomics studies are sensitive to changes in sample preparation protocols and instrumentation, which can introduce external noise into the experimental data. Well-designed and well-controlled experiments can mitigate these challenges by improving reproducibility through standardized workflows [
25].
Given the dynamic nature of proteins, reproducibility in proteomics may require a new perspective. Unlike the relatively stable genome, the proteome changes over time and under different conditions. This variability can be seen as an opportunity to gain more comprehensive biological insights. For example, studying a single organism with a good phenotype over time may provide more reliable targets for generating hypotheses than pooled heterogeneous samples. In proteomics, the concept of “reproducibility” may need to be reconsidered as “continuous convergence”, where the focus is on consistent detection of biological signatures over time rather than exact replication of results. Innovative bioinformatics and mathematical tools can aid in the storing, visualization and interpretation of these dynamic datasets and ensure that meaningful conclusions can be drawn despite the biological noise [
25,
26]. All biological systems generate noise. Early targeted approaches focusing on a single or a few biomarkers have produced successful clinical markers. However, the challenge in the omics era is to filter out a signal from the vast amount of data generated. Better instruments and omics technologies have not always translated into more clinically relevant biomarker discoveries, possibly due to the overfitting of data and the variable quality of samples and instruments. To counteract noise, omics studies should leverage the sensitivity of latest technologies to investigate specific pathways and interactions in smaller cohorts, with each individual serving as its own control. This approach can help discover new signals and reduce confounding variables. The study of noise itself can also provide valuable insights into biological variability and homeostasis and help to redefine the criteria for reproducibility in the context of biological noise (
Table 3) [
27].
The integration of genome and proteome analyses is a crucial aspect of high-throughput omics that enables a comprehensive understanding of biological systems. Genome analysis provides insights into the DNA sequence and its variations, while proteome analysis provides information on the expression, modification and interaction of proteins. The integration of these two datasets can reveal how genetic variations affect protein expression and function, and thus elucidate the molecular mechanisms underlying various diseases. For example, the integration of genomic and proteomic data has been shown to improve the understanding of cancer biology, allowing researchers to identify potential biomarkers and therapeutic targets with greater accuracy. Tools such as the Omics Integrator and methods such as network-based integration help to combine these datasets and facilitate the construction of detailed molecular interaction networks [
28,
29].
In addition, the integration of genomic and proteomic data has significant implications for personalized medicine. By correlating genetic variants with protein expression patterns, researchers can develop more precise diagnostic tools and treatment strategies tailored to individual patients. In colorectal cancer research, for example, integrated proteogenomic analyses have identified specific protein signatures that are associated with different clinical outcomes, improving patient stratification and facilitating treatment decisions. This approach not only increases the predictive power of genomic data, but also provides a functional context that helps prioritize candidate genes and proteins for further investigation. Bioinformatics platforms, like iProClass [
30] and iProXpress [
31], facilitate the integration and functional annotation of datasets, enabling comprehensive analyses and meaningful biological insights [
29,
32]. For instance, iProClass [
30] offers detailed data on protein sequences, structures, functions and pathways, facilitating the integration and analysis of complex proteomics datasets.
4. Integration and Interoperability of Omics Data
The integration and interoperability of omics data are critical to the advancement of biological research and enable a holistic understanding of complex biological systems. Effective integration combines different omics datasets, such as genomics, transcriptomics, proteomics and metabolomics, to provide comprehensive insights that individual data types alone cannot achieve. Strategies for data integration include early, mixed, intermediate, late and hierarchical integration, each of which offers unique advantages depending on the research context [
59]. Successful integration methods must overcome challenges such as heterogeneity of data, management of large datasets and standardization of biological identities [
105,
106]. In addition, novel approaches, such as regularized canonical correlation analysis and graphical lasso, have shown promise in identifying important biomarkers and deciphering complex biological interactions [
107]. As omics technologies continue to evolve, robust integration methods are essential to maximize the utility of multi-omics datasets and advance precision medicine and systems biology (
Table 7) [
12].
Effective data integration strategies are critical for promoting the interoperability of omics data and enabling comprehensive insights into complex biological systems. One prominent approach is early integration, in which all omics datasets are merged into a single matrix for subsequent analysis using machine learning models. This method utilizes the full spectrum of the data, but can be computationally intensive due to the high dimensionality [
59]. Another approach is mixed integration, where each omics dataset is independently transformed into a new representation before being combined for downstream analysis, striking a balance between computational efficiency and data dimensionality [
59].
Intermediate and late integration strategies provide additional flexibility by converting datasets into general and omics-specific representations simultaneously, or by analyzing each omics dataset separately before combining their predictions. Hierarchical integration, where datasets are organized based on previous regulatory relationships, provides a structured approach to data combination and improves the interpretability and relevance of results [
59]. Omics data integration using regularized canonical correlation analysis and graphical lasso has shown promise in identifying important biomarker candidates and deciphering complex interactions within biological systems [
107]. As high-throughput technologies continue to evolve, these integration strategies are crucial to maximizing the utility of multi-omics datasets and advancing precision medicine and systems biology.
Omics data integration and interoperability requires advanced tools and techniques to manage the complexity and heterogeneity of high-throughput datasets. One of the most important tools is the Omics Integrator software (
https://fraenkel-nsf.csbi.mit.edu/omicsintegrator/, accessed on 1 June 2024), which uses network-based approaches to integrate different omics data and identify the underlying molecular pathways. This software uses advanced network optimization algorithms to find subnetworks with high confidence, facilitating the interpretation of multi-omics data in a biologically meaningful way [
10]. Another notable tool is the OmicsPLS package, which implements Two-way Orthogonal Partial Least Squares (O2PLS) for efficient handling and integration of low and high dimensional datasets. This package enables inspection and visualization of integrated omics data, improving the ability to gain meaningful biological insights [
108].
Table 7.
Integration and Interoperability of Omics Data: Approaches, Tools, and Benefits.
Table 7.
Integration and Interoperability of Omics Data: Approaches, Tools, and Benefits.
Omics Type | Integration Approach | Interoperability Tools and Standards | Benefits | Limitations |
---|
Genomics | Cross-referencing genetic variants | VCF (Variant Call Format), dbSNP [109], Ensembl [110] | Identifies genetic variations and their effects | High data volume; interpretation of variants; privacy issues |
Transcriptomics | Aligning RNA-seq data with genome | FASTQ, SAM/BAM, GTF/GFF, GEO (https://genome.ucsc.edu/ENCODE/, accessed on 1 June 2024) | Reveals gene expression patterns and alternative splicing | Data complexity; variability between samples |
Proteomics | Correlating protein levels with gene expression | mzML [111], PRIDE [57], UniProt [112] | Understands protein abundance and functional roles | Sensitivity to sample preparation; high technical variability |
Metabolomics | Linking metabolites to metabolic pathways | mzTab, HMDB [113], KEGG [114] | Provides insights into cellular metabolism and pathway activities | Complex data integration; diverse chemical properties |
Epigenomics | Mapping epigenetic modifications | BED, WIG, GEO, ENCODE (https://genome.ucsc.edu/ENCODE/, accessed on 1 June 2024) | Studies DNA methylation, histone modifications and chromatin accessibility | High data complexity; dynamic nature of epigenetic changes |
Lipidomics | Profiling lipid species | LIPID MAPS [115], mzXML [111] | Explores lipid composition and its role in cell biology | Heterogeneity of lipids; difficulty in quantifying low-abundance lipids |
Glycomics | Characterizing glycan structures | GlyTouCan [116], UniCarb-DB [117] | Analyzes glycan functions and interactions | Structural complexity of glycans; limited analytical standards |
Microbiomics | Integrating microbiome with host data | QIIME [118], MG-RAST [119] | Examines microbial communities and their impact on host health | Variability in sample preparation; complex data interpretation |
Phenomics | Associating phenotypic traits with molecular data | PhenX [120], PheWAS [121], dbGaP [122] | Identifies molecular markers linked to phenotypes | High variability in phenotype data; integration with genomic data |
Pharmacogenomics | Linking drug response to genetic profiles | PharmGKB [123], CPIC | Personalizes medicine based on genetic profiles | Ethical concerns; variability in drug response among individuals |
In addition to these tools, various techniques have been developed to improve the integration of multi-omics data. XML is often used to facilitate cross-platform data integration as it is easy to learn, store and transfer. Techniques such as bio-warehousing, database federation and controlled vocabularies also play a crucial role in managing heterogeneous data sources [
124]. In addition, integration strategies such as early, mixed, medium, late and hierarchical integration meet different research needs and increase the flexibility and applicability of data integration processes [
59]. As this field progresses, the development of more robust, scalable and user-friendly tools and techniques will be crucial to fully exploit the potential of multi-omics data in biological research and precision medicine.
The integration and interoperability of omics data is a major challenge due to the heterogeneity and complexity of the datasets involved. One of the biggest challenges is the heterogeneity of the data, which includes different formats, scales and types of omics data such as genomics, proteomics and metabolomics. This diversity makes it difficult to bring these datasets together in a coherent framework for analysis. Furthermore, the amount of data generated by high-throughput technologies exacerbates these problems and makes data storage, management and retrieval complex tasks [
125]. Another major challenge is the lack of standardization of different data types and sources, which hinders the effective sharing and reuse of data. Standardization issues include differences in terminology, units of measurement and data formats, which can lead to difficulties in integrating and interpreting data [
12].
To overcome these challenges, various solutions have been proposed and developed. One effective approach is to use advanced bioinformatics tools and algorithms that can handle the complexity and scale of multi-omics data. For example, tools such as Omics Integrator and OmicsPLS facilitate the integration of different omics datasets by using sophisticated algorithms to identify meaningful biological pathways and interactions [
10]. In addition, the use of standardized data formats and controlled vocabularies can significantly improve data interoperability. For example, XML-based data integration techniques provide a way to standardize the storage, migration and validation of omics data so that it can be more easily integrated and analyzed across different platforms. In addition, the use of machine learning and deep learning techniques has shown promise for managing and interpreting large-scale multi-omics data. They offer new insights into complex biological systems and improve the possibilities of predictive modeling [
126].
An exemplary initiative in this area is NeDRexDB [
127], which integrates data from a variety of biomedical databases such as OMIM [
128], DisGeNET [
129], UniProt [
112], NCBI gene info, IID [
130], Reactome [
131], MONDO [
132], DrugBank [
133] and DrugCentral [
134]. By aggregating data from these different sources, NeDRexDB constructs heterogeneous networks that represent different biomedical entities (e.g., diseases, genes, drugs) and their interconnections [
127]. Researchers can access and explore these networks via NeDRexApp, NeDRexAPI [
127] and the Neo4j endpoint [
135] to NeDRexDB, creating a versatile platform for data analysis [
127]. NeDRexApp, a Cytoscape [
136] application, extends this functionality by providing implementations of state-of-the-art network algorithms, such as Multi-Steiner Trees (MuST) [
137], TrustRank [
138], Biclustering Constrained by Networks (BiCoN) [
139] and Disease Module Detection (DIAMOnD) [
140]. These advanced algorithms are accessible to users via the RESTful API and the user-friendly NeDRexApp interface. They require a list of user-selected genes (called seeds) as a starting point, with BiCoN being an exception as it uses gene expression data. These seeds can contain all or a subset of genes associated with a particular disease (disease genes) or genes within disease modules. In addition, expert knowledge can be used to select the seeds and the results can be statistically validated by calculating empirical
p-values. In this way, NeDRex enables state-of-the-art network medicine methods that allow researchers in pharmacology and biomedicine to use their expertise to discover candidates for drug repurposing and gain deeper insights into disease mechanisms. By integrating diverse datasets and applying sophisticated computational tools, NeDRex exemplifies how advanced bioinformatics solutions can overcome the challenges posed by the heterogeneity and complexity of multi-omics data and ultimately advance precision medicine and biomedical research. A recent study has illustrated the capabilities of NeDRex by identifying critical modules involved in glioblastoma multiforme (GBM) disease, such as MYC, EGFR, PIK3CA, SUZ12 and SPRK2. In addition, hub genes were identified in a comprehensive interaction network containing 7560 proteins associated with GBM disease and 3860 proteins associated with signaling pathways involved in GBM. By integrating these results and performing a centrality analysis, 11 key genes involved in GBM disease were identified [
141].
The integration of high-throughput omics data into precision medicine has significant implications for matching treatments to individual patients. By combining different types of data—such as genomics, proteomics, metabolomics and transcriptomics—researchers can create comprehensive molecular profiles that improve the understanding of disease mechanisms. This multi-omics approach enables the identification of specific biomarkers and therapeutic targets and facilitates personalized treatment plans that are more effective and have fewer side effects. In oncology, for example, the integration of multi-omics data has led to the discovery of novel cancer subtypes and the development of targeted therapies that improve patient outcomes [
142]. In addition, advanced computational techniques, such as machine learning and deep learning, are being used to analyze these complex datasets and further improve the precision of therapeutic interventions [
143]. Furthermore, the application of multi-omics data integration in precision medicine extends beyond cancer to other complex diseases. This holistic approach enables the stratification of patients based on their molecular profiles, leading to more precise diagnoses and tailored treatments. In the field of metabolic diseases, for example, the integration of multi-omics data has enabled the identification of key metabolic pathways and regulatory networks that can be targeted therapeutically [
80]. In addition, the integration of electronic health records (EHR) with multi-omics data supported by artificial intelligence has the potential to revolutionize patient care by providing real-time, data-driven insights into disease progression and treatment efficacy [
144]. These advances underscore the transformative impact of integrating high-throughput omics on the practice of precision medicine, which will ultimately lead to more personalized and effective healthcare solutions.