Fact-checked by Grok 2 weeks ago

Multiomics

Multi-omics is an integrative approach in that combines data from multiple high-throughput technologies, such as , transcriptomics, , , , microbiomics, and emerging modalities like single-cell and spatial omics, to generate a comprehensive, multidimensional view of biological systems and their dysregulation in . This method addresses the limitations of single-omics studies by capturing interactions across molecular layers—from DNA alterations to protein function and metabolite profiles—enabling deeper insights into genotype-phenotype relationships and complex pathways. The origins of multi-omics trace back to foundational advancements, including the completed in 2003, which established as a cornerstone for high-throughput analysis. The field gained momentum in the with innovations in next-generation sequencing (NGS), for , and computational tools for , leading to a surge in publications—particularly in cancer and human health studies—by the late . Key milestones include cost reductions in whole-genome sequencing to under $500 by 2021 and the rise of single-cell technologies around 2016, which expanded multi-omics to resolve heterogeneity at cellular resolution. In applications, multi-omics has transformed precision medicine by facilitating discovery, patient subtyping, prediction, and drug target identification across diseases like cancer, Alzheimer's, Parkinson's, , and aging-related conditions. For instance, in , it integrates genomic mutations with transcriptomic and proteomic data to match therapies, improving response rates in trials like WINTHER (from 23% to 35% treatment matching). Beyond human health, it supports agricultural and environmental research by elucidating molecular mechanisms in model organisms. Despite its promise, multi-omics faces challenges in data heterogeneity, integration complexity, missing values, high computational demands, and ensuring through standardized pipelines and (findable, accessible, interoperable, reusable) principles. Ongoing developments in and multimodal technologies aim to overcome these hurdles, positioning multi-omics as a for future biological discovery.

Definition and Fundamentals

Definition and Scope

Multiomics refers to the simultaneous and of multiple "omics" datasets, such as , transcriptomics, , and , to capture complex interactions across biological scales from DNA to . This approach, also known as panomics, emphasizes the combined study of diverse molecular layers to provide a more complete picture of biological systems than isolated analyses. By integrating data from at least three omics layers derived from different regulatory levels, multiomics enables the elucidation of dynamic processes that govern cellular function. The core objectives of multiomics include achieving a holistic view of cellular mechanisms, identifying regulatory networks that link molecular events, and predicting phenotypic outcomes from genotypic variations. These goals address the inherent limitations of single-omics studies, which often fail to account for post-transcriptional, post-translational, or environmental influences—for instance, genomic data alone cannot reveal how transcriptomic regulation modulates protein activity. Multiomics overcomes these constraints by layering complementary data types, thereby revealing causal relationships and reducing noise from individual modalities to support applications like biomarker discovery. Key concepts in multiomics distinguish between vertical integration, which aligns data across molecular layers (e.g., genome to metabolome) within the same samples to trace regulatory cascades, and horizontal integration, which combines data from the same omics layer across different samples or conditions to identify population-level patterns. For example, multiomics can uncover emergent properties such as pathway crosstalk, where interactions between signaling pathways like Hippo and immune responses in tumors become evident only through integrated analyses, highlighting regulatory dynamics invisible in single-omics views.

Historical Development

The historical development of multiomics originated from the sequential emergence of individual omics disciplines, beginning with foundational advances in . In 1977, and colleagues developed the chain-termination method for , enabling the analysis of genetic material at scale and setting the stage for . The , initiated in 1990 and culminating in a draft sequence in 2001 and a complete version in 2003, represented a landmark effort that sequenced the entire , fostering the birth of high-throughput genomic studies. Building on this, transcriptomics advanced in 1995 with the introduction of DNA microarrays by Patrick Brown and colleagues, which allowed simultaneous measurement of thousands of levels. , meanwhile, gained momentum in the early 2000s through refinements in —pioneered in 1975—and , with the Human Proteome Organization (HUPO) established in 2001 to coordinate global efforts. Key technological drivers in the mid-2000s transformed these silos into integrated possibilities. The advent of next-generation sequencing (NGS) technologies, such as 454 introduced by 454 Life Sciences in 2005, dramatically reduced costs and increased throughput, making large-scale data generation feasible. Concurrently, the (ENCODE) project, launched by the in 2003, began systematically mapping functional elements across the genome, providing layered datasets that highlighted the need for multi-level analyses. , a pioneer in , played a crucial role by founding the Institute for Systems Biology in 2000 and advocating for holistic biological profiling that integrated , , and beyond, influencing the shift toward systems-oriented . The concept of multiomics coalesced in the late and early as computational and experimental tools matured to handle . The term "multi-omics" first appeared in in 2002, reflecting efforts to combine multiple layers for a more comprehensive view of biological systems, as seen in early reviews on disease modeling. A pivotal milestone was the 2012 launch of the Integrative Human Microbiome Project (iHMP) by the , the first large-scale initiative to profile microbial communities using integrated genomic, transcriptomic, proteomic, and metabolomic data from human cohorts. By the mid-, single-cell multiomics emerged, with technologies like combined single-cell sequencing and profiling post-2015 enabling resolution of cellular heterogeneity, exemplified by methods such as those developed in 2016 for multiplexed assays. These advancements, culminating in AI-assisted integration by the 2020s, underscored multiomics' evolution from isolated to a unified framework for understanding complex biology.

Omics Data Types

Core Omics Layers

The core omics layers form the foundational components of multiomics research, capturing sequential levels of biological information from genetic material to downstream functional outputs. These layers—genomics, transcriptomics, proteomics, metabolomics, and epigenomics—enable a comprehensive view of molecular processes by profiling distinct biomolecular entities, their generation typically relies on high-throughput technologies, and their data exhibit unique characteristics suited to specific analytical approaches. Together, they reflect the central dogma of molecular biology, where genetic information flows from DNA to RNA, proteins, and ultimately metabolites, allowing researchers to trace regulatory cascades and phenotypic outcomes. Genomics focuses on the complete set of DNA within an organism, including its sequence variants, structural features, and functional elements. It primarily examines genetic variations such as single nucleotide polymorphisms (SNPs) and copy number variations (CNVs), which influence disease susceptibility and trait inheritance. Genomic data are generated through next-generation sequencing (NGS) technologies, such as Illumina platforms, which enable high-throughput reading of DNA fragments to produce raw sequence reads or processed variant calls. The resulting data consist of aligned sequences, variant frequency tables, or genome-wide maps, often quantified in terms of coverage depth (e.g., 30x for whole-genome sequencing) to ensure accuracy. Transcriptomics investigates the , defined as the full repertoire of RNA molecules transcribed from the , to assess dynamics and events. It reveals how environmental cues or genetic factors modulate RNA abundance, providing insights into cellular responses and regulatory networks. Transcriptomic profiles are commonly produced using RNA sequencing (), which involves reverse transcription of RNA to cDNA followed by NGS, yielding high-resolution data on transcript diversity. Key data outputs include read counts per , normalized metrics like fragments per kilobase of transcript per million mapped reads (FPKM), or differential expression matrices, capturing both coding and non-coding RNAs. Proteomics targets the , the entire collection of proteins expressed by a , emphasizing their abundance, post-translational modifications (e.g., ), and interactions that determine cellular function. Unlike nucleic acid-based layers, accounts for the dynamic and context-dependent nature of proteins, which cannot be fully predicted from genomic or transcriptomic data due to regulatory discrepancies. Data are primarily generated via (MS), including liquid chromatography-tandem MS (LC-MS/MS), where proteins are digested into peptides, ionized, and fragmented for identification and quantification. Outputs comprise spectral intensities, peptide-spectrum matches, or values, often revealing thousands of proteins per sample with variability in detection limits around 10-100 ng/mL. Metabolomics profiles the , encompassing all small-molecule metabolites (typically <1,500 Da) that reflect the end products of cellular and physiological states. It captures biochemical pathways influenced by , , and , offering a snapshot of organismal closer to clinical outcomes than upstream . Metabolites are detected using analytical platforms like liquid chromatography-mass spectrometry (LC-MS) or gas chromatography-MS (GC-MS), which separate and ionize compounds for identification against databases. Data characteristics include peak areas or relative concentrations for hundreds to thousands of features, normalized to internal standards, with challenges in absolute quantification due to matrix effects. Epigenomics examines epigenetic modifications across the genome, such as and marks, which regulate without altering the DNA sequence. These layers provide mechanistic insights into how environmental factors induce heritable changes in structure and accessibility. Common methods include for and followed by sequencing (ChIP-seq) for modifications, enabling genome-wide mapping. Data are represented as enrichment scores, beta values (0-1 for methylation levels), or peak calls at regulatory regions like promoters, highlighting dynamic patterns in development and disease. These core layers interconnect hierarchically: genomic variations underpin transcriptomic regulation, which translates to proteomic outputs, further influencing metabolomic profiles, thus forming a unified framework for dissecting complex biological systems.

Emerging Omics Modalities

Emerging omics modalities encompass specialized layers that extend the scope of multiomics by capturing niche molecular and environmental features, such as lipid dynamics, modifications, microbial ecosystems, and protein interaction networks, thereby enhancing the understanding of cellular function and host-environment interactions. These approaches, often enabled by post-2010 technological refinements in and sequencing, provide complementary data to core omics layers, revealing functional dimensions like signaling pathways and community-level influences that are critical for . Lipidomics profiles the diverse lipid species within cells and tissues, focusing on their roles in membrane structure, energy storage, and signaling cascades. Shotgun lipidomics, which analyzes intact lipids directly from crude extracts without prior separation, has seen significant advancements since 2010, including higher-resolution that enables quantitative detection of hundreds of lipid subclasses with improved sensitivity and throughput. For instance, multidimensional mass spectrometry strategies in shotgun lipidomics have facilitated the identification of lipid alterations in diseases like , where specific changes correlate with inflammatory responses. This adds depth to multiomics by linking lipid composition to dynamic processes, such as vesicle trafficking and receptor activation, which are not fully captured by genomic or proteomic data alone. Glycomics investigates the structure and function of glycans—complex carbohydrates attached to proteins and —essential for processes like , immune recognition, and interactions. (MALDI-MS) has emerged as a key technique for profiling, allowing high-throughput analysis of patterns in biological samples. In immunity and cancer, glycomics reveals aberrant structures, such as truncated sialylated glycans on tumor cells that promote immune evasion and . Recent MALDI-MS innovations, including variants, enable spatial mapping of glycans in tissues, contributing to multiomics by elucidating how modulates protein function and influences disease progression in contexts like . Microbiomics examines the composition, diversity, and functional potential of microbial communities, particularly in the human gut, , and other niches, highlighting their impact on host physiology through metabolite production and immune modulation. 16S rRNA gene sequencing targets conserved bacterial ribosomal genes to assess taxonomic diversity, while shotgun provides functional gene content via whole-community genome sequencing. These methods have uncovered host-microbe interactions, such as how gut microbiota-derived regulate epithelial barrier integrity and . In multiomics frameworks, microbiomics introduces an environmental dimension, integrating microbial data with host to model in conditions like . Interactomics maps protein-protein interactions (PPIs) to construct networks that reveal signaling complexes and regulatory hubs within cells. Techniques like yeast two-hybrid (Y2H) screening detect binary interactions , while affinity purification-mass spectrometry (AP-MS) captures native complexes for proteomic identification. Recent AP-MS advancements, including quantitative crosslinking and , have expanded interactome maps to over 10,000 human PPIs, visualized as dynamic networks that inform pathway perturbations. This modality enriches multiomics by providing structural insights into functional assemblies, such as kinase-substrate networks in cancer signaling. Technological enablers have further propelled these modalities since 2012, notably CRISPR-based epigenome editing, which uses deactivated fused to epigenetic modifiers to precisely alter or marks without sequence changes. This tool facilitates targeted epigenomic profiling in multiomics studies, linking regulatory landscapes to in and . Complementing this, long-read sequencing technologies, such as and Oxford Nanopore, resolve complex structural variants like insertions and inversions that short-read methods miss, enhancing genomic context for emerging layers. For example, long-read data has identified microbiome-associated structural variants influencing host susceptibility to infections, adding layers of genomic-environmental interplay.

Data Acquisition and Integration

Multiomic Data Collection Methods

Multiomic data collection can occur through sequential or simultaneous approaches, each tailored to capture multiple molecular layers from the same biological sample while minimizing technical artifacts such as batch effects. In sequential collection, analytes are extracted in a stepwise manner from bulk tissues, allowing for the isolation of DNA, RNA, and proteins from a single sample to ensure direct comparability across layers. For instance, commercial kits like the Qiagen AllPrep DNA/RNA/Protein Mini Kit, introduced in the late 2000s, enable the simultaneous purification of high-quality genomic DNA and total RNA from cells and tissues in a single spin-column format, reducing sample heterogeneity and preserving molecular integrity for downstream analyses. This method has been widely adopted in bulk multiomics studies to avoid splitting samples, which could introduce variability. Simultaneous collection, often via multimodal assays, integrates multiple omics layers in a single experimental workflow, particularly advantageous for single-cell resolutions where sample scarcity is a concern. These assays couple techniques like oligonucleotide-tagged antibodies or transposase-based indexing to profile alongside other modalities without physical separation. A prominent example is (cellular indexing of transcriptomes and epitopes by sequencing), which simultaneously measures single-cell mRNA expression and surface protein levels using DNA-barcoded antibodies, enabling the detection of up to 100 proteins alongside the full in thousands of cells. Similarly, SHARE-seq facilitates joint profiling of chromatin accessibility and by combining ATAC-seq-like tagmentation with RNA capture, allowing high-throughput analysis of regulatory elements and transcriptional states in hundreds of single nuclei from frozen tissues. Such methods enhance compatibility by processing all layers in parallel, reducing batch effects inherent in separate assays. Sample preparation poses significant challenges in multiomic collection, particularly in preserving the integrity of diverse biomolecules across scales from tissues to single cells. Effective preservation methods, such as snap-freezing samples in immediately after collection, are essential to halt degradation of labile components like and proteins while maintaining structure for epigenomic assays. This approach is critical for extractions, where tissues are homogenized prior to multi-layer isolation, and extends to single-cell workflows, where dissociation protocols must cell viability with multiomic yield—often favoring nuclei isolation over whole cells to improve recovery and reduce cytoplasmic . Scaling from (millions of cells) to single-cell levels requires optimized conditions to avoid cross-contamination between layers, with challenges amplified in heterogeneous tissues like tumors. Experimental designs in multiomics emphasize strategies that capture temporal or population-level dynamics while ensuring robust data across layers. Longitudinal sampling, involving repeated collections from the same subjects over time, is particularly valuable for studying disease progression or treatment responses, as it links multiomic changes to clinical trajectories without confounding inter-individual variability. Large-scale studies exemplify this, such as (TCGA), a multiomics initiative spanning 2006 to 2016 that profiled genomic, transcriptomic, epigenomic, and proteomic data from approximately 11,000 samples across 33 cancer types, providing a for minimizing batch effects through standardized protocols and sample matching. These designs typically incorporate power calculations for cohort sizes to detect subtle multi-layer associations, prioritizing paired samples to enhance signal-to-noise ratios. Quality control during multiomic data collection is vital for cross-layer comparability, focusing on normalization techniques to account for technical variations in yield and sequencing depth. Spike-in controls, such as External RNA Controls Consortium (ERCC) synthetic transcripts added at known concentrations prior to library preparation, serve as internal standards for calibrating abundance across samples and layers, enabling accurate normalization of transcriptomic data relative to genomic or proteomic profiles. In multimodal assays, additional QC metrics like library complexity checks and batch effect assessments via ensure that variations arise from rather than processing artifacts, with thresholds for read depth and coverage uniformity applied to low-quality samples before .

Data Integration Strategies

Data integration strategies in multiomics involve computational and statistical frameworks designed to combine heterogeneous datasets from multiple omics layers, such as , transcriptomics, and , to uncover correlated patterns, infer causal relationships, and enhance biological insights. These strategies are broadly classified into and supervised approaches, as well as integration paradigms, addressing challenges like high dimensionality, data heterogeneity, and missing values. Vertical integration fuses different types measured on the same samples to model joint distributions, while horizontal integration aligns similar data across independent studies or cohorts to mitigate batch effects and enable meta-analyses. Unsupervised methods focus on and latent factor discovery without prior labels, facilitating exploratory analysis of multiomics data. (PCA) and t-distributed stochastic neighbor embedding (t-SNE) are commonly applied for reducing high-dimensional multiomics datasets into lower-dimensional representations that preserve global and local structures, respectively, allowing visualization of cross-omics relationships. A seminal unsupervised framework is Multi-Omics Factor Analysis (MOFA), introduced in 2018, which decomposes variation across omics layers into shared and modality-specific factors using a probabilistic model, enabling the identification of principal sources of variation in datasets like those from cancer studies. Supervised approaches incorporate phenotypic or outcome information to guide , often through network-based or probabilistic models for targeted correlation and . Network-based methods, such as Weighted Analysis (WGCNA), construct co-expression modules across layers to reveal interconnected modules associated with traits, as demonstrated in transcriptomics-proteomics integrations. Bayesian models provide probabilistic fusion by modeling uncertainties and dependencies, for instance, through hierarchical frameworks that integrate multiomics data with missing values via sampling, supporting joint in disease association studies. Vertical integration employs layer-specific models for joint analysis of matched multiomics data on the same samples, exemplified by iCluster, a 2009 method that performs integrative clustering via a joint . iCluster maximizes a joint likelihood across to identify sample clusters and subtype-specific features, formulated as: L = \sum \log P(\mathbf{X}_g | \mathbf{Z}, \theta_g) + \log P(\mathbf{Z} | \theta_z) where \mathbf{X}_g denotes data from the g-th layer, \mathbf{Z} is the shared latent cluster assignment, and \theta parameters are estimated via expectation-maximization, applied successfully to and subtype analysis. Horizontal integration facilitates by combining similar data from multiple studies, addressing heterogeneity through batch correction techniques like , which adjusts for known or unknown batch effects using empirical Bayes estimation to remove non-biological variance while preserving signals. This approach enhances statistical power in large-scale consortia, such as those integrating transcriptomics across cohorts for . To ensure and , multiomics adheres to standards like the Minimum Information About BIobank data Sharing (MIABIS) ontology, which standardizes metadata descriptions for biobanks and samples, including attributes for data and experimental conditions. Workflows often leverage tools such as the R-based MixOmics package, which implements multivariate methods like sparse partial for and supervised , supporting correlation analysis across in diverse biological contexts.

Advanced Techniques

Single-Cell Multiomics

Single-cell multiomics extends multiomic profiling to the resolution of individual cells, allowing the simultaneous measurement of multiple molecular layers such as transcriptomes, epigenomes, and proteomes within heterogeneous populations. This approach overcomes the averaging effects of bulk multiomics by capturing cellular diversity, enabling the dissection of rare subpopulations and dynamic processes that are masked in population-level analyses. Key advancements include methods that integrate single-cell RNA sequencing () with assays for chromatin accessibility or surface proteins, facilitating a deeper understanding of regulatory mechanisms at the single-cell level. Core technologies in single-cell multiomics have evolved to jointly profile and other modalities. For instance, sci-CAR combines scRNA-seq with single-cell (scATAC-seq) through combinatorial indexing, enabling scalable profiling of both and accessibility in thousands of cells from complex tissues like the . Similarly, REAP-seq ( expression and ) uses oligonucleotide-tagged antibodies to simultaneously quantify transcripts and surface proteins, such as immune markers, in human T cells, providing insights into protein-RNA correlations without spatial information. These methods build on foundational scRNA-seq platforms, adapting them for multimodal capture. The typical workflow for single-cell multiomics begins with cell isolation, often using fluorescence-activated cell sorting (FACS) for enrichment of specific populations or droplet-based encapsulation for high-throughput processing, as in the Chromium system, which partitions s into nanoliter droplets for efficient handling of up to 10,000 cells per run. s or nuclei are then barcoded with unique molecular identifiers (UMIs) during reverse transcription or amplification, followed by pooled library preparation and joint sequencing on platforms like Illumina NovaSeq, where multimodal reads are demultiplexed based on barcodes to assign features to individual s. This process ensures traceability of layers to the same while minimizing batch effects. Single-cell multiomic data are characterized by high dimensionality and sparsity, typically represented as count matrices with 10^4 to 10^5 features (e.g., , peaks, or ) per , but with only a fraction detected due to technical dropout rates exceeding 80% for lowly expressed features. To address missing values arising from this sparsity, imputation methods like (Markov affinity-based imputation of ) apply diffusion-based smoothing across similar , recovering continuous patterns and reducing noise without introducing artifacts. These data structures demand specialized preprocessing to handle zero-inflation and variability. A primary advantage of single-cell multiomics lies in its ability to resolve heterogeneous states, such as distinguishing malignant subclones in tumors or tracking trajectories during embryonic , where bulk methods fail to capture transitions. For example, tools like order s along pseudotime based on multiomic features, revealing regulatory dynamics in processes like myoblast . This has illuminated fate decisions that drive phenotypic diversity. However, single-cell multiomics faces resolution limits due to inherent trade-offs between throughput and depth; while platforms like enable profiling up to 10^6 cells in extended runs, this often reduces per-cell molecular coverage to a few thousand reads, limiting detection of rare transcripts or low-abundance proteins. Increasing depth via deeper sequencing improves sensitivity but scales costs quadratically, constraining applications to targeted subsets rather than whole tissues. Ongoing optimizations aim to balance these constraints for broader utility. As of 2025, deep learning-based methods like scAI and DCCA have advanced integration of single-cell multi-omics data, preserving biological signals more effectively than traditional approaches.

Spatial Multiomics

Spatial multiomics refers to the integration of multiple omics layers—such as transcriptomics, proteomics, and genomics—while preserving the spatial context within tissues, enabling the mapping of molecular profiles to specific locations to uncover tissue organization, cellular interactions, and functional heterogeneity. Unlike dissociated single-cell approaches, spatial multiomics maintains the positional information of cells and molecules, revealing how omics data varies across microenvironments in intact tissues. This approach has become essential for studying complex biological systems where location influences function, such as in developmental biology and pathology. Key technologies in spatial multiomics include sequencing-based and imaging-based platforms that capture multiomic data at varying resolutions. The Visium platform from 10x Genomics, introduced in 2019, enables unbiased spatial transcriptomics by capturing mRNA on barcoded slides with approximately 55 µm spot resolution, allowing whole-transcriptome profiling across tissue sections. For protein and DNA analysis, NanoString's GeoMx Digital Spatial Profiler uses targeted probes to quantify up to hundreds of analytes in user-defined regions of interest, supporting multiomic overlays on formalin-fixed paraffin-embedded samples. High-resolution imaging methods like MERFISH (multiplexed error-robust fluorescence in situ hybridization), developed in 2015, achieve subcellular resolution for RNA detection, imaging thousands of transcripts per cell through combinatorial barcoding and error correction. These technologies generate data structured as voxel-based matrices, where each spatial coordinate (e.g., spot or pixel) is associated with multiomic features, facilitating the visualization of molecular distributions in 2D or 3D tissue maps. Integration of multiomic layers in spatial contexts involves overlaying datasets from complementary modalities to correlate, for instance, expression with protein localization. Platforms like NanoString's CosMx, launched in , enable simultaneous high-plex imaging of over 1,000 RNAs and 100 proteins at subcellular resolution (<1 µm), allowing direct multiomic profiling within the same tissue section to study co-localization patterns. Deconvolution algorithms such as enhance integration by inferring cell-type compositions in low-resolution spots using single-cell references, improving the granularity of multiomic maps without additional experiments. In applications like mapping tumor microenvironments, spatial multiomics has revealed heterogeneous interactions between cancer cells, immune infiltrates, and stromal components, such as altered signaling gradients in that drive therapeutic resistance. For example, integrating transcriptomic and proteomic data has identified spatially restricted immune checkpoints in solid tumors, informing targeted immunotherapies. Post-2020 advances have focused on multimodal platforms that combine sequencing and imaging for broader coverage and higher throughput. SeqFISH+, an extension of sequential introduced in 2019, profiles over 10,000 genes at near-single-molecule resolution by iterative hybridization rounds, enabling dense multiomic atlases of tissues like the with minimal optical crowding. These developments have expanded spatial multiomics to include epigenomic layers, such as spatial for accessibility, integrated with transcriptomics to link regulatory elements to spatial . As of 2025, methods like MultiGATE provide integrative analysis and regulatory inference in spatial multi-omics, jointly profiling and epigenome/protein markers. Despite these progresses, challenges persist, including resolution trade-offs—spot-level methods like Visium average signals across multiple cells, while subcellular imaging like MERFISH demands extensive computational resources for error correction and data alignment. High-dimensional multiomic datasets also require scalable algorithms to handle noise, batch effects, and the computational intensity of 3D reconstructions, limiting widespread adoption in resource-constrained settings.

Computational Tools

Software and Algorithms

Software and algorithms are essential for handling the complexity of multiomic data, encompassing preprocessing to ensure , to combine disparate layers, to reveal patterns, and comprehensive platforms for end-to-end workflows. These tools emphasize to manage terabyte-scale datasets and through standardized formats like those in , an R-based ecosystem with over 2,360 packages (as of release 3.22 in 2025) facilitating multiomic analyses. Benchmarks highlight their performance in high-throughput environments. In preprocessing, FastQC provides quality control for next-generation sequencing data by evaluating metrics like per-base sequence quality and , generating interactive reports for rapid assessment. STAR, an ultrafast aligner for reads, supports spliced alignments and handles up to billions of reads efficiently, achieving speeds significantly faster than many comparable tools, as demonstrated in early benchmarks. For multiomic-specific preprocessing, MultiQC aggregates outputs from tools like FastQC and STAR across multiple samples and types into unified reports. Integration platforms enable the fusion of multiomic datasets to uncover shared variations. MOFA2, an extension of Multi-Omics Factor Analysis, uses unsupervised to decompose multi-view data into latent factors, supporting diverse modalities like and while handling through variational for scalable training on large datasets. NEMO (Neighborhood-based Multi-Omics clustering) integrates partial multiomic profiles via kernel-based similarity measures, performing robust clustering even when some are unavailable for subsets of samples, as demonstrated in cancer subtyping benchmarks where it outperformed similarity-network fusion methods in accuracy. Visualization tools aid in interpreting integrated results through intuitive representations. Cytoscape facilitates network-based views of multiomic interactions, with apps like enabling layered displays of genomic and proteomic edges for exploring correlations. In , ggplot2 extensions such as support multi-layer heatmaps to depict hierarchical patterns across omics, allowing annotation tracks for metadata and scalable rendering for datasets with millions of features. Comprehensive suites streamline multiomic pipelines. , a web-based platform introduced in 2005, offers modular workflows integrating preprocessing tools like and integration methods like MOFA2, promoting reproducibility through shareable histories and scalability via cloud deployments handling petabyte-scale data. Partek Flow, a solution, provides a graphical interface for multiomic analysis, including automated alignment, variant calling, and . Selection criteria for these tools prioritize scalability and , exemplified by Bioconductor's S4 classes for seamless data exchange between packages like MOFA2 and tools.

Machine Learning Applications

has revolutionized multiomics analysis by enabling the handling of high-dimensional, heterogeneous data to uncover patterns that traditional statistical methods often miss. In paradigms, random forests have been particularly effective for discovery, as they aggregate predictions from multiple decision trees to identify key features across layers such as and . For instance, random forests integrate multiomics data to predict disease outcomes while ranking biomarkers based on their contribution to accuracy. Feature importance in random forests is derived from the mean decrease in Gini impurity across trees, where the Gini index for a node with class proportions p_i is given by Gini = 1 - \sum_{i=1}^{C} p_i^2, and importance is the weighted sum of impurity reductions from splits on that feature, scaled by the number of samples at each node. This approach has demonstrated superior performance in identifying robust biomarkers from integrated transcriptomic and metabolomic datasets. Deep learning techniques further enhance multiomics by capturing nonlinear relationships and reducing dimensionality. Autoencoders, particularly variational autoencoders (VAEs), compress multiomic profiles into low-dimensional latent spaces while preserving biological variance, facilitating downstream tasks like integration of single-cell RNA-seq with epigenomic data. A seminal example is scVI, a deep generative model that uses a VAE framework to model count-based omics data, accounting for technical noise and batch effects to yield biologically interpretable embeddings. Graph neural networks (GNNs) extend this by modeling interactomes, where nodes represent genes or proteins and edges capture interactions from multiomics sources; GNNs propagate information through these graphs to predict functional modules, improving accuracy in cancer subtyping by 10-20% compared to non-graph methods. Unsupervised learning with VAEs enables clustering of multiomic samples to reveal subtypes without labels, as seen in frameworks that jointly embed genomic, transcriptomic, and proteomic data into a shared for . These models leverage the (ELBO) objective to balance fidelity and regularization, yielding clusters that align with known heterogeneity. Anomaly detection, another unsupervised application, identifies outliers in cohorts by reconstructing multiomic profiles and flagging high errors, which has proven useful in detecting rare variants in cancer progression studies. For example, VAE-based detectors have isolated anomalous metabolic profiles in tumor cohorts with precision exceeding 85%. Recent advances incorporate large language models (LLMs) for interpreting multiomic outputs, where post-2023 models fine-tuned on sequences generate hypotheses about regulatory mechanisms, such as linking variants to pathway disruptions with contextual explanations. addresses privacy concerns in multi-site multiomics by training models across distributed datasets without centralizing sensitive information, using techniques like to aggregate gradients; this has enabled collaborative analysis of genomic cohorts from multiple hospitals while maintaining . A practical case is predicting response from multiomic profiles using the Genomics of Drug Sensitivity in Cancer (GDSC) database, where models integrate mutation, expression, and copy number data to forecast sensitivity, achieving Pearson correlations of 0.7-0.8 for hundreds of compounds. These predictions guide personalized therapies by prioritizing drugs targeting patient-specific vulnerabilities, as validated in cross-cohort evaluations.

Applications in Biology and Medicine

Health and Disease Studies

Multiomics approaches have revolutionized the of cancer by integrating genomic, transcriptomic, epigenomic, and proteomic to uncover driver mutations and molecular subtypes. (TCGA) Pan-Cancer Analysis Project, initiated in 2013, initially profiled tumors across 12 cancer types, revealing recurrent driver mutations such as those in TP53 and PIK3CA, and defining subtypes like the four intrinsic subtypes (luminal A, luminal B, HER2-enriched, and basal-like) through multiplatform analyses including , RNA expression, and profiling. These efforts extended into the Pan-Cancer Atlas, which analyzed over 11,000 tumors across 33 cancer types, further delineating pan-cancer themes in pathway alterations and immune landscapes, enabling the identification of actionable therapeutic targets across tumor types. In contexts, multiomic signatures have predicted responses to checkpoint inhibitors; for instance, profiling of patients treated with anti-PD-1 therapy identified immunosuppressive features in non-responding tumors, such as reduced T-cell infiltration and altered metabolic profiles, achieving predictive accuracy with an area under the curve () of 0.84. In neurodegenerative diseases, multiomics has elucidated complex interactions between pathology markers and metabolic pathways, particularly in Alzheimer's disease (AD). The Accelerating Medicines Partnership-Alzheimer's Disease (AMP-AD) consortium, launched in 2014 with key analyses from 2015 onward, integrated transcriptomic, proteomic, and metabolomic data from brain tissues and cohorts like the Religious Orders Study and Rush Memory and Aging Project, revealing links between amyloid-beta plaques, tau tangles, and dysregulated metabolites such as altered lipid and amino acid pathways that exacerbate neuronal damage. A comprehensive metabolomics study within AMP-AD identified 298 brain metabolites associated with AD traits, including 188 linked to tau tangles and 34 to amyloid-beta deposition, highlighting pathways like sphingolipid metabolism as potential therapeutic targets and correlating these changes with cognitive decline severity. Proteomic analyses from the consortium further mapped tau interactomes, showing synaptic and mitochondrial disruptions tied to amyloid-tau interactions, which inform disease progression models. For infectious diseases, multiomics profiling has mapped host responses to pathogens, exemplified by studies in 2020 that integrated single-cell sequencing, , and to characterize immune dysregulation. Analyses integrating data from over 1,000 patients revealed severity-specific signatures, such as hyperinflammation in severe cases driven by elevated cytokines (e.g., IL-6) and monocyte activation, alongside metabolic shifts toward supporting immune exhaustion. These profiles distinguished mild from critical disease with high specificity, identifying biomarkers like and IFN-stimulated genes that predict outcomes and guide antiviral therapies. Multiomics supports precision medicine by integrating pharmacogenomic data with other layers to enable personalized dosing and treatment selection. The National Cancer Institute's Molecular Analysis for Therapy Choice (NCI-MATCH) trial, launched in 2015, screened tumors for over 140 actionable mutations using genomic profiling, assigning patients to targeted therapies regardless of cancer type, which demonstrated responses in mutation-matched arms and informed dosing adjustments based on pharmacokinetic interactions. Broader pharmacogenomics applications incorporate multiomics to predict ; for example, combining CYP450 with transcriptomic and metabolomic data has optimized and dosing, reducing adverse events by identifying variability in drug clearance pathways. Overall, multiomics integration in and studies has enhanced survival predictions, with models combining genomic and clinical data yielding improvements in concordance indices for cancer outcomes compared to single-omics approaches; for instance, frameworks on TCGA breast cancer data increased c-index from approximately 0.62 (single-omics) to 0.64 (multiomics), enabling better risk stratification and therapeutic planning. Recent advances as of 2025 include the integration of multi-omics with for precision medicine in gastrointestinal diseases, such as using on and host data to predict (IBD) therapeutic responses and (CRC) prognosis through signatures.

Microbiome and Immunology

The Integrative Human Microbiome Project (iHMP), conducted from 2012 to 2022 as the second phase of the NIH Project, generated longitudinal multi-omic datasets from over 300 subjects across multiple clinical sites to investigate dynamic host-microbiome interactions in health and disease. This initiative focused on three cohorts—, (IBD), and / onset—employing , , , , host transcriptomics, and immune assays to capture microbiome stability and perturbations. In the IBD cohort, involving 132 participants, multi-omic profiling of 1,785 stool samples, 651 biopsies, and 529 blood samples revealed greater temporal instability in the gut of (CD) patients compared to (UC) or controls, with frequent shifts in microbial composition linked to disease flares. Key findings highlighted functional during active IBD, characterized by reduced short-chain production (e.g., butyrate) and increased primary bile acids, correlating with host immune markers such as anti-Saccharomyces cerevisiae antibodies (ASCA) and elevated fecal calprotectin. Systems immunology leverages multi-omic approaches to map immune cell states and regulatory networks, integrating transcriptomics, , and to uncover hidden drivers of immunity. For instance, single-cell RNA sequencing (scRNA-seq) and () have delineated T-cell heterogeneity and activation pathways, such as Runx3-mediated CD8+ T-cell residency in tissues. Integrations with repositories like the Immunology Database and Portal (ImmPort) facilitate mining of curated immune datasets, enabling cross-study analyses of and cellular phenotypes. Cytokine-proteome correlations, analyzed via tools like NetBID, reveal regulatory interactions, such as Hippo Mst1 influencing metabolism and networks in CD8α+ cells. Host-microbe multi-omics studies combine and host to elucidate mechanisms, particularly in metabolic disorders like . In a of 100 Italian women, multi-omic profiling (16S rRNA, , , ) identified low microbial diversity and transcriptional inactivity in pathways among obese individuals with uncontrolled eating behaviors, correlating with elevated primary bile acids and sterols. These microbial shifts influenced related to and neuroendocrine signaling, underscoring -driven alterations in physiology. Spatial multi-omics techniques, such as spatial host-microbiome sequencing (SHM-seq), further reveal localized interactions at the gut barrier, profiling bacterial distributions (e.g., in the , Pseudobutyrivibrio in mucosa) alongside transcripts like Muc2 in colon tissues. This approach demonstrates bacterial co-organization with goblet cells and colonocytes, modulating immune signaling and barrier integrity in specific morphological niches. Multi-omic analyses have yielded insights into microbial metabolites shaping host , with (SCFAs) like butyrate acting as inhibitors to alter structure and in intestinal cells. Metagenomic and epigenomic integrations in IBD cohorts show microbiota-derived and supplying methyl donors (e.g., S-adenosylmethionine) for , linking to disease-associated epigenetic patterns. In , baseline multi-omic signatures from B- and T-cell profiles predict vaccine responses; for example, integration of transcriptomics, , and in a hepatitis B study identified JAK-STAT and TLR4 pathway markers that forecast antibody titers with high accuracy using supervised models like DIABLO. The Project 2 (HMP2), launched in 2017 as an extension of iHMP, applied multi-omics to the cohort, collecting longitudinal data from 96 pregnancies to track vaginal and placental microbiomes. Findings indicated dynamic shifts in microbial communities preceding preterm delivery, with reduced dominance and increased diversity correlating with host inflammatory responses.

Challenges and Future Directions

Current Limitations

Multiomics research generates vast volumes of , often reaching petabyte-scale datasets, which pose significant challenges for and . The inherent heterogeneity across layers—such as differences in data formats, resolution, and noise levels from , transcriptomics, and —complicates integration and analysis. Storage costs further exacerbate these issues, with substantial expenses for archiving large repositories in comprehensive projects. Analysis of multiomics data is hindered by missing values, where imputation methods can introduce biases that skew downstream inferences, particularly when missingness is not completely at random. remains a critical barrier in single-cell multiomics, as processing datasets exceeding 10^6 cells demands (HPC) resources to handle the computational intensity without prohibitive delays. Ethical concerns in multiomics prominently include privacy risks associated with genomic data sharing, necessitating compliance with regulations like the EU's (GDPR) enacted in 2018 to safeguard identifiable information. Additionally, inequities in diversity persist, with underrepresented populations often excluded from studies, limiting the generalizability of findings and perpetuating health disparities. Reproducibility in multiomics is undermined by the absence of standardized pipelines, leading to variability in results across labs and hindering validation efforts. High-dimensional statistical analyses are particularly susceptible to p-hacking, where selective reporting of p-values inflates false positives and erodes trust in discoveries. Resource limitations further restrict multiomics adoption, with costs for processing multiomic samples, which can range from hundreds to thousands of dollars depending on the layers included and facility. is especially challenging in low-resource settings, where limited and funding prevent routine implementation, widening global research gaps. In the coming years, technological advances in multiomics are poised to accelerate through of diverse datasets, enabling dynamic and predictive modeling in biological systems. For instance, models trained on longitudinal multi-omics are expected to facilitate clinical by adjusting therapeutic interventions based on evolving profiles. quantum-classical platforms are being developed to handle the complexity of multiomics network , building on frameworks like the encode-search-build approach for in multiomics . As of , advancements in and sequencing have reduced costs, facilitating wider adoption, alongside tools like multimodal large language models for . Clinical applications of multiomics are shifting toward routine diagnostics, particularly through advanced liquid biopsies that combine (ctDNA) with metabolomic profiles to improve early detection and monitoring of diseases like cancer. This integration allows for comprehensive analysis in non-invasive samples, supporting personalized strategies and reducing reliance on invasive procedures. The global multiomics market is projected to reach approximately $11 billion by 2034, driven by increasing adoption in diagnostics and therapeutics. Interdisciplinary fusions are expanding multiomics into imaging and environmental domains, with multiomic MRI techniques correlating molecular profiles with spatial imaging data to reveal intratumor heterogeneity and disease progression. Similarly, exposomics—the study of cumulative environmental exposures—is integrating with multiomics to map gene-environment interactions, providing a holistic view of health influences through longitudinal multi-omics signatures. In precision medicine, multiomic avatars—digital representations of patient biology derived from integrated omics data—are enabling tailored therapies by simulating treatment responses at the individual level. For example, these avatars support predictive modeling in oncology, guiding drug selection and minimizing adverse effects. Extensions of the Human Cell Atlas initiative are advancing single-cell spatial multiomics atlases, creating comprehensive maps of cellular diversity across tissues in the 2020s to inform organ-specific interventions. Post-2025 trends indicate mainstream adoption of multiomics in pharmaceutical , with -enhanced pipelines accelerating and clinical trials through omics-based personalization. To address accompanying challenges, ethical frameworks for multiomics emphasize data privacy, algorithmic fairness, and , ensuring responsible integration into healthcare systems.

References

  1. [1]
    Applications of multi‐omics analysis in human diseases - PMC
    Jul 31, 2023 · Multi‐omics usually refers to the crossover application of multiple high‐throughput screening technologies represented by genomics, ...
  2. [2]
    State of the Field in Multi-Omics Research - PubMed Central - NIH
    We define multi-omics as three or more omic datasets coming from different layers of biological regulation – not necessarily within one level (exclusively ...
  3. [3]
    The Essentials of Multiomics - PMC - PubMed Central
    Feb 22, 2022 · Through multiomics, the combined use many available technologies, a more complete and dynamic vision of cancer can be obtained.
  4. [4]
    State of the Field in Multi-Omics Research: From Computational ...
    We define multi-omics as three or more omic datasets coming from different layers of biological regulation – not necessarily within one level (exclusively ...
  5. [5]
    Top Trends in Multiomics Research: Evaluation of 52 Published ...
    ... multiomics, integromics, and panomics. Articles were evaluated and compared ... However, in some studies, synonyms for multiomics, such as ...
  6. [6]
    Vertical and horizontal integration of multi-omics data with miodin
    Dec 10, 2019 · The package allows users to integrate omics data from different experiments on the same samples (vertical integration) or across studies on the ...
  7. [7]
    Pan‐cancer multi‐omics analyses reveal crosstalk between the ...
    Dec 2, 2021 · Pan-cancer multi-omics analyses reveal crosstalk between the Hippo and immune signaling pathways in the tumor microenvironment
  8. [8]
    Multi-Omics Profiling for Health - PMC - PubMed Central
    This review highlights current and emerging multi-omics modalities for precision health and discusses applications in the following areas.
  9. [9]
    Multi-omics data integration using ratio-based quantitative profiling ...
    Sep 7, 2023 · The principle of the central dogma was well reflected in the Quartet multi-omics data, as it could be seen that the abundance of RNAs was ...
  10. [10]
    Genomics in medicine: A new era in medicine - PMC
    In this review article, we have reviewed the evolution of genomic methodologies/tools, their limitations, and scope, for current and future clinical application ...
  11. [11]
    Transcriptomics technologies - PMC - NIH
    Transcriptomics technologies are the techniques used to study an organism's transcriptome, the sum of all of its RNA transcripts.
  12. [12]
    Proteomics: Concepts and applications in human medicine - PMC
    Proteomics is the complete evaluation of the function and structure of proteins to understand an organism's nature.
  13. [13]
    Metabolomics: an emerging but powerful tool for precision medicine
    Metabolomics is the comprehensive analysis of metabolites, measuring all low-molecular-weight molecules in a biological specimen.
  14. [14]
    Epigenomics—Technologies and Applications - PMC
    The goal is to create an ultimate picture of the epigenome integrating DNA methylation, chromatin dynamics and accessibility, and expression. Some of the more ...
  15. [15]
    Genome-resolved metagenomics: a game changer for microbiome ...
    Jul 1, 2024 · This review provides an overview of the capabilities and methods of genome-resolved metagenomics for studying the human microbiome.
  16. [16]
    Novel Advances in Shotgun Lipidomics for Biology and Medicine
    This review focuses on shotgun lipidomics. After briefly introducing its fundamentals, the major materials of this article cover its recent advances.Missing: post- seminal paper
  17. [17]
    Multi-dimensional mass spectrometry-based shotgun lipidomics and ...
    Aug 10, 2025 · These developments include new strategies and refinements for shotgun lipidomic approaches that use direct infusion, including novel ...
  18. [18]
    Recent Advances in the Mass Spectrometry Methods for Glycomics ...
    The review focuses on recent aspects (last three years) of glycosylation analyses that provide relevant information about cancer. It includes recent development ...Missing: paper | Show results with:paper
  19. [19]
    MALDI mass spectrometry imaging of N-linked glycans in cancer ...
    Abstract. Glycosylated proteins account for a majority of the post-translation modifications of cell surface, secreted and circulating proteins.Missing: paper | Show results with:paper
  20. [20]
  21. [21]
    Characterization of the Gut Microbiome Using 16S or Shotgun ...
    The two main approaches for analyzing the microbiome, 16S ribosomal RNA (rRNA) gene amplicons and shotgun metagenomics, are illustrated with analyses of ...Missing: microbiomics | Show results with:microbiomics
  22. [22]
    system biology perspective on environment–host–microbe interactions
    In this review we discuss the role of the gut microbiome in complex diseases and the possible causal scenarios behind its interactions with the host genome and ...A System Biology Perspective... · Gut Microbiome Is A Complex... · Environment--Genetics--Microbiome...Missing: microbiomics | Show results with:microbiomics
  23. [23]
    Mass spectrometry‐based protein–protein interaction networks for ...
    Jan 12, 2021 · Here, we review MS techniques that have been instrumental for the identification of protein–protein interactions at a system‐level.
  24. [24]
    Recent Advances in Mass Spectrometry-Based Protein Interactome ...
    Nov 26, 2024 · This review summarizes recent advances in MS-based interactomics, focusing on the development of techniques that capture protein-protein, protein-metabolite, ...
  25. [25]
    CRISPR/Cas9-based epigenome editing: An overview of ... - PubMed
    Jul 15, 2019 · Here, we review strategies for recruitment of effector domains, used in gene regulation and epigenome editing, to the dCas9 DNA-targeting protein.
  26. [26]
    Structural variation in 1,019 diverse humans based on long ... - Nature
    Jul 23, 2025 · Imputation of structural variants using a multi-ancestry long-read sequencing panel enables identification of disease associations. Preprint ...
  27. [27]
    Long-read genome sequencing and multi-omics in aging ... - medRxiv
    Oct 29, 2025 · Long-read sequencing revealed structural variants (SVs) (many not detected with short-read sequencing) that we interpreted using matched ...
  28. [28]
    AllPrep DNA/RNA Mini Kit (50) - QIAGEN
    In stock $40 delivery 30-day returnsAllPrep DNA/RNA Kits are designed for simultaneous purification of DNA and RNA from cells and tissues, in spin-column and 96-well formats.Allprep Dna/rna Mini Kit... · Performance · PublicationsMissing: history 2008
  29. [29]
    Simultaneous Extraction of High-Quality RNA and DNA from Small ...
    Oct 7, 2008 · The first was the Qiagen AllPrep DNA/RNA Micro kit, which we used according to the manufacturer's instructions. For homogenization, we used the ...
  30. [30]
    Sample Preparation for Multi‐Omics Analysis: Considerations and ...
    Jun 23, 2025 · While the term genomics was first used in 1986, DNA sequencing technologies began to develop in the late 1970s with two pioneering approaches ...Missing: timeline | Show results with:timeline
  31. [31]
    The Cancer Genome Atlas Program (TCGA) - NCI
    The Cancer Genome Atlas (TCGA) is a landmark cancer genomics program that sequenced and molecularly characterized over 11000 cases of primary cancer samples ...Using TCGA · TCGA Molecular... · TCGA by the Numbers · GDC Data PortalMissing: multiomics experimental designs longitudinal 2006-2016
  32. [32]
    A guide to multi-omics data collection and integration for ...
    Multi-omics involves collecting and integrating patient samples, aiming to detect disease patterns, subtype identification, diagnosis, drug response, and ...2. Methods · 6.1. Subtype Identification · 7.2. Multi-Omics Pathway...
  33. [33]
    A real-world multi-center RNA-seq benchmarking study using the ...
    Jul 22, 2024 · The MAQC Consortium utilized these samples with spike-ins of 92 synthetic RNA from the External RNA Control Consortium (ERCC) to assess RNA-seq ...
  34. [34]
    A technical review of multi-omics data integration methods
    Aug 1, 2025 · Here, we comprehensively review state-of-the-art multi-omics integration methods with a focus on deep generative models, particularly ...
  35. [35]
    Unsupervised Multi-Omics Data Integration Methods - Frontiers
    Mar 21, 2022 · This review aims to provide an overview of multi-Omics data integration methods with different statistical approaches, focusing on unsupervised learning tasks.
  36. [36]
    WGCNA: an R package for weighted correlation network analysis
    Dec 29, 2008 · Overview of WGCNA methodology. This flowchart presents a brief overview of the main steps of Weighted Gene Co-expression Network Analysis.
  37. [37]
    Bayesian integrative model for multi-omics data with missingness
    Such Bayesian hierarchical modelling has gained popularity in multi-omics integrative analysis due to its flexibility in model construction for complex ...<|separator|>
  38. [38]
    Integrative clustering of multiple genomic data types using a joint ...
    In this study, we applied iCluster to integrate copy number and gene expression data. The joint latent variable model is completely scalable to include ...
  39. [39]
    Update of the Minimum Information About BIobank Data Sharing ...
    We reviewed the descriptive structure and metadata of the MIABIS terminology. All the attribute descriptions were synchronized across the component, and the ...Missing: multiomics | Show results with:multiomics
  40. [40]
    Single-cell multiomics: technologies and data analysis methods
    Sep 15, 2020 · Here, we summarize the technologies for single-cell multiomics analyses (mRNA-genome, mRNA-DNA methylation, mRNA-chromatin accessibility, and mRNA-protein)
  41. [41]
    Joint profiling of chromatin accessibility and gene expression in ...
    Cao et al. present sci-CAR, a pooled barcode method that jointly analyzes both the RNA transcripts and chromatin profiles of single cells. By applying sci-CAR ...
  42. [42]
    Massively parallel digital transcriptional profiling of single cells
    Jan 16, 2017 · We developed a droplet-based system that enables 3′ messenger RNA (mRNA) digital counting of thousands of single cells.
  43. [43]
    Methods and applications for single-cell and spatial multi-omics
    Mar 2, 2023 · In this Review, we highlight advances in the fast-developing field of single-cell and spatial multi-omics technologies (also known as multimodal omics ...
  44. [44]
    Visium Spatial Platform | 10x Genomics
    The Visium platform delivers unbiased, whole transcriptome spatial gene expression analysis at single cell scale with unmatched spatial data quality.Best-In-Class Spatial Data... · Visium Hd Wt Panel · New! Visium Hd 3'Missing: seminal paper
  45. [45]
    Spatially resolved, highly multiplexed RNA profiling in single cells
    We report multiplexed error-robust FISH (MERFISH), a single-molecule imaging method that allows thousands of RNA species to be imaged in single cells.
  46. [46]
    Transcriptome-scale super-resolved imaging in tissues by RNA ...
    Mar 25, 2019 · The transcriptome-level profiling of seqFISH+ allows unbiased identification of cell classes and their spatial organization in tissues. In ...Missing: spatial paper
  47. [47]
    Benchmarking multi-omics integration algorithms across single-cell ...
    Mar 16, 2024 · In this paper, we benchmarked 12 multi-omics integration methods on three integration tasks via qualitative visualization and quantitative metrics.Missing: STAR | Show results with:STAR
  48. [48]
    MOFA+: a statistical framework for comprehensive integration of ...
    May 11, 2020 · We present Multi-Omics Factor Analysis v2 (MOFA+), a statistical framework for the comprehensive and scalable integration of single-cell multi-modal data.
  49. [49]
    NEMO: cancer subtyping by integration of partial multi-omic data
    We present NEMO (NEighborhood based Multi-Omics clustering), a novel algorithm for multi-omics clustering. Importantly, NEMO can be applied to partial datasets.
  50. [50]
    A random forest based biomarker discovery and power analysis ...
    Nov 23, 2020 · The random forest algorithm is a powerful prediction method that is known to be able to capture complex dependency patterns between the outcome ...Missing: seminal | Show results with:seminal
  51. [51]
    multiomics data integration using graph convolutional networks
    SUPREME is a node classification framework that integrates multiple data modalities using graph convolutional networks for multiomics data.Missing: interactomes | Show results with:interactomes
  52. [52]
    Integrated multi-omics analysis of ovarian cancer using variational ...
    Mar 18, 2021 · An unsupervised deep learning framework with variational autoencoders for genome-wide dna methylation analysis and biologic feature ...Methods · Clustering And... · Results
  53. [53]
    Omics-based large language models: A new engine for drug ...
    Oct 28, 2025 · This review provides a systematic overview of the applications of LLMs in omics technologies and their potential for drug discovery from three ...
  54. [54]
    PPML-Omics: A privacy-preserving federated machine learning ...
    Jan 31, 2024 · We proposed a secure and privacy-preserving machine learning method (PPML-Omics) by designing a decentralized differential private federated learning algorithm.
  55. [55]
    Deep learning and multi-omics approach to predict drug responses ...
    Nov 28, 2022 · We propose a novel deep neural network model that integrates multi-omics data available as gene expressions, copy number variations, gene mutations, reverse ...
  56. [56]
    The Cancer Genome Atlas Pan-Cancer analysis project - Nature
    Sep 26, 2013 · The Pan-Cancer initiative compares the first 12 tumor types profiled by TCGA. Analysis of the molecular aberrations and their functional roles ...Molecular Profiling Of... · Author Information · The Cancer Genome Atlas...
  57. [57]
    Multiomic profiling of checkpoint inhibitor-treated melanoma
    Jan 10, 2022 · Multiomic analysis predicts response ... Li, et al. Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response.
  58. [58]
    Accelerating Medicines Partnership® Program for Alzheimer's ...
    Oct 15, 2024 · The first iteration of the AMP Program for AD was focused on discovering new therapeutic targets and on evaluating the usefulness of tau imaging ...Missing: multiomics metabolite 2015
  59. [59]
    The landscape of metabolic brain alterations in Alzheimer's disease
    Jul 13, 2022 · The majority of the 298 metabolites was associated with one of three AD traits: cognitive decline (n = 201), tau tangles (n = 188), and global ...
  60. [60]
    Tau interactome maps synaptic and mitochondrial processes ...
    Feb 17, 2022 · A multiomics approach on multiple cohorts from the Accelerating Medicines Partnership—Alzheimer's Disease (AMP-AD) consortium identified six co- ...
  61. [61]
    Multi-omic profiling reveals widespread dysregulation of innate ...
    To investigate how immune responses vary between different severities of COVID-19, we profiled peripheral blood immune cells from 64 patients with COVID-19 and ...
  62. [62]
    NCI-MATCH trial will link targeted cancer drugs to gene abnormalities
    Jun 1, 2015 · The trial seeks to determine whether targeted therapies for people whose tumors have specific gene mutations will be effective regardless of their cancer type.Missing: pharmacogenomics personalized
  63. [63]
    Revolutionizing Personalized Medicine: Synergy with Multi-Omics ...
    Genomics plays a crucial role in identifying biomarkers for diseases such as cancer, enabling early diagnosis, monitoring, and personalized treatments. Cancer ...
  64. [64]
    Deep learning based feature-level integration of multi-omics data for ...
    Sep 15, 2020 · In this study, we aim to improve breast cancer overall survival prediction by integrating multi-omics data (e.g., gene expression, DNA ...Tcga-Brca Breast Cancer... · Single-Modality Network · Novel Multi-Modality...<|control11|><|separator|>
  65. [65]
    Human Microbiome Project (HMP) - NIH Common Fund
    iHMP studies and datasets in preterm birth, inflammatory bowel disease, and prediabetes that expand our undestanding of host/microbiome interactions. More ...Missing: 2012-2022 multiomic subjects findings stability
  66. [66]
    The Integrative Human Microbiome Project - PMC - PubMed Central
    May 29, 2019 · The HMP2 expanded the repertoire of biological properties analysed for both host and microbiome in three longitudinal cohort studies of ...Missing: extensions | Show results with:extensions
  67. [67]
    Multi-omics of the gut microbial ecosystem in inflammatory bowel ...
    May 29, 2019 · Here we present the results, which provide a comprehensive view of functional dysbiosis in the gut microbiome during inflammatory bowel disease activity.
  68. [68]
    Systems immunology: Integrating multi-omics data to infer regulatory ...
    Mar 12, 2019 · Transcriptome analysis has provided instrumental insights into the mechanisms of immune system development and homeostasis under steady state, ...Missing: multiomic | Show results with:multiomic
  69. [69]
    Multi-omics gut microbiome signatures in obese women: role of diet ...
    Dec 27, 2022 · In an attempt to advance our knowledge of the gut-microbiome-brain axis in the obese phenotype, we thoroughly characterized the gut microbiome ...
  70. [70]
    Spatial host–microbiome sequencing reveals niches in the mouse gut
    Nov 20, 2023 · We present spatial host–microbiome sequencing (SHM-seq), an all-sequencing-based approach that captures tissue histology, polyadenylated RNAs and bacterial 16S ...
  71. [71]
  72. [72]
    Multi-Omic Data Integration Allows Baseline Immune Signatures to ...
    Nov 29, 2020 · We used DIABLO to derive a multi-omics model capable of predicting vaccine response from baseline molecular profiles. Critically, this ...
  73. [73]
    2025 Trends: Multiomics
    Jan 6, 2025 · Multiomics is going mainstream. But as researchers revel in single cell resolution, challenges in storing and harnessing the data loom large.
  74. [74]
    Multiomics Research: Principles and Challenges in Integrated ...
    Multiomics research is a transformative approach in the biological sciences that integrates data from genomics, transcriptomics, proteomics, metabolomics, and ...
  75. [75]
    Integrating Machine Learning and Multi-Omics to Explore Neutrophil ...
    Sep 5, 2025 · In practice, the implementation of ML for multi-omics analysis incurs significant computational and data storage costs [133]. The high ...
  76. [76]
    Missing data in multi-omics integration: Recent advances through ...
    Feb 9, 2023 · Missing samples in multi-omics data are rarely MCAR, and thus inference based on a complete case analysis is likely to be biased. A separate ...
  77. [77]
    ScaleSC: a superfast and scalable single-cell RNA-seq data ...
    Jul 17, 2025 · ScaleSC delivers over a 20× speedup through GPU computing and significantly improves scalability, handling datasets of 10–20 million cells with ...
  78. [78]
    Policy Brief: can genomic data be anonymised? - GA4GH
    The GDPR does not regulate anonymised data at all, and insists on keeping data in an identifiable form for no longer than necessary for the purposes for which ...Missing: concerns multiomics
  79. [79]
    A Framework for Promoting Diversity, Equity, and Inclusion in ...
    Apr 15, 2022 · We propose a novel framework for promoting diversity, equity, and inclusion in genomics research. ... Newer approaches to cohort-based research ...
  80. [80]
    Integrated omics: tools, advances and future approaches in
    We discuss recent approaches, existing tools and potential caveats in the integration of omics datasets for development of standardized analytical pipelines.<|separator|>
  81. [81]
    The reproducibility of research and the misinterpretation of p-values
    Dec 6, 2017 · The weak evidence provided by p-values between 0.01 and 0.05 is explored by exact calculations of false positive risks.
  82. [82]
    Mass Spectrometry Proteomics Core Fees | BCM
    Pricing Information (effective Nov. 1, 2025) ; Multiomic Project Setup, Per Project. $300 ; Multiomic Sample Processing- Frozen, Per Project. $90.
  83. [83]
    Implementation of multi-omics in diagnosis of pediatric rare diseases
    Nov 19, 2024 · Testing is often unavailable in low-resource medical settings, necessitating referrals to specialized centers, which can lead to increased time ...