Fact-checked by Grok 2 weeks ago

Pathway analysis

Pathway analysis is a computational approach in bioinformatics and that interprets high-throughput molecular data—such as profiles from , transcriptomics, or —by mapping differentially expressed genes or proteins onto known biological pathways to identify those significantly enriched or perturbed under specific conditions, like disease versus control states. This method provides biological context to large lists of molecular changes, revealing coordinated functional alterations in processes such as , signaling, or , and aids in hypothesis generation for mechanistic studies. By leveraging curated databases like , Reactome, or WikiPathways, pathway analysis transforms raw data into interpretable insights about cellular networks and disease mechanisms. Originating from early genetic mapping efforts in the mid-20th century, pathway analysis gained prominence with the advent of high-throughput technologies following the in 2001, evolving into a cornerstone of research by the mid-2000s. Key methods include over-representation analysis (ORA), which tests for disproportionate gene enrichment in pathways using hypergeometric or Fisher's exact tests; functional class scoring (FCS), such as (GSEA), which evaluates cumulative pathway activity across all genes without arbitrary thresholds; and topology-based approaches (e.g., Signaling Pathway Impact Analysis or SPIA), which account for gene interactions and pathway structures to better capture regulatory dynamics. These techniques have been benchmarked extensively, with topology-based methods often outperforming others in accuracy for identifying truly impacted pathways, though challenges like database incompleteness (covering only about 45% of human genes) and statistical biases persist. In the multi-omics era, pathway analysis has expanded to integrate diverse data types, including and , enabling more holistic views of biological systems and supporting applications in , , and identification. Recent advances emphasize network-based and machine learning-enhanced methods to address limitations in traditional enrichment analyses, improving and across cohorts. As of , over 700 pathway databases exist, underscoring the field's maturity, yet ongoing efforts focus on standardizing annotations and handling complex, interconnected pathway topologies for robust interpretation.

Overview

Definition and purpose

Pathway analysis is a computational method in bioinformatics that integrates high-throughput data, such as profiles, protein abundances, or metabolite levels, with predefined biological pathways to identify those that are statistically enriched or perturbed under specific experimental conditions. This approach maps individual molecular changes onto structured networks of interacting genes, proteins, and metabolites, revealing coordinated alterations that might otherwise be obscured in . The primary purpose of pathway analysis is to provide biological context and mechanistic insights from lists of differentially expressed or regulated molecules, facilitating the interpretation of complex datasets in areas like disease pathology, response, or environmental perturbations. By focusing on pathways rather than isolated entities, it enables researchers to infer affected cellular processes, such as signaling cascades or metabolic routes, and prioritize hypotheses for further validation. Pathway analysis emerged in the early alongside the proliferation of technologies, which generated vast datasets requiring systematic interpretation beyond individual assessments. A pivotal advancement occurred in 2005 with the introduction of (GSEA) by Subramanian et al., which formalized a knowledge-based framework for detecting subtle, coordinated pathway-level changes without relying on arbitrary significance thresholds for single genes. This method addresses key limitations of traditional single-gene analysis, particularly the multiple testing problem, where stringent corrections for thousands of hypotheses often yield few significant findings despite evident biological effects. By evaluating sets collectively, pathway analysis enhances to modest changes across multiple components, reducing false negatives and providing a more holistic view of system-wide perturbations.

Types of biological pathways

Biological pathways are primarily categorized into three main types: metabolic, signaling, and regulatory, each representing distinct mechanisms of cellular function and interaction. These classifications provide foundational models for understanding how biological systems process information and maintain , serving as targets for pathway analysis in data. Metabolic pathways comprise sequences of enzymatic reactions that transform substrates into products, facilitating energy production and . Examples include , which converts glucose to pyruvate, and the Krebs cycle (tricarboxylic acid cycle), which generates reducing equivalents for . These pathways are typically modeled as directed graphs, with nodes representing metabolites or enzymes and edges denoting chemical reactions or conversions, often incorporating and directionality to reflect flux through the system. Signaling pathways consist of cascades that propagate extracellular signals to elicit cellular responses, involving sequential activation of molecular components. A prominent example is the (MAPK) pathway, which transmits signals from growth factors to regulate processes like through protein interactions and events. In graph representations, nodes correspond to proteins, receptors, or second messengers, while directed edges illustrate activations, inhibitions, or bindings, emphasizing the flow of information rather than material transformation. Regulatory pathways, often termed gene regulatory networks, govern the control of through interactions among , microRNAs (miRNAs), and other regulators. The p53 signaling network exemplifies this, where p53 acts as a responding to DNA damage to activate genes involved in arrest or . These are depicted as directed graphs with nodes as genes, proteins, or regulatory elements and edges indicating transcriptional activation, repression, or post-transcriptional modifications, capturing dynamic feedback loops. Across these types, biological pathways are commonly formalized as graphs where nodes denote biological entities such as genes, proteins, or metabolites, and edges capture interactions, reactions, or regulatory relationships, enabling computational analysis of structure and dynamics. Standards like BioPAX (Biological Pathway Exchange) facilitate the interchange of pathway data, supporting representations of metabolic, signaling, and regulatory processes at molecular and genetic levels, while SBML ( Markup Language) provides an XML-based format for encoding quantitative models of these pathways, including regulatory networks. In contrast to broader biological , which integrate diverse interactions across an entire system and often display scale-free topologies with high connectivity, pathways are curated, context-specific models emphasizing linear or branching sequences of functionally linked events rather than exhaustive connectivity.

Applications

In and transcriptomics

In and transcriptomics, pathway analysis typically begins with differential expression (DE) analysis of high-throughput sequencing data, such as or outputs, to identify genes with significant changes in expression levels between conditions. For data, tools like DESeq2 are commonly employed to normalize read counts, model variance, and compute s and p-values for DE genes, generating ranked lists based on statistics like log2 or adjusted p-values. These ranked gene lists or values are then fed into pathway enrichment methods, such as over-representation analysis or , to assess whether predefined biological pathways are dysregulated. This approach offers key advantages over single-gene analysis by accommodating genome-wide data scale, where thousands of genes are tested simultaneously, reducing false positives through multiple testing corrections while highlighting coordinated pathway-level perturbations that individual gene effects might obscure. For instance, it integrates subtle changes across multiple genes within a pathway, providing biological context and revealing mechanisms like signaling cascades that drive disease phenotypes, rather than isolated markers. In cancer transcriptomics, pathway analysis has been instrumental in identifying dysregulated pathways; for example, RNA-seq studies of tumor samples have shown enrichment in and immune activation pathways, such as those involving signaling, which correlate with tumor progression and response. Similarly, in genome-wide association studies (GWAS), single nucleotide polymorphisms (SNPs) are mapped to pathway genes using annotation resources, enabling enrichment analysis to link genetic variants to broader biological processes like regulation, thereby prioritizing candidate pathways for functional validation. Quantitative metrics from these analyses often include enrichment p-values, which indicate the of pathway dysregulation; in tumor datasets, the , implicated in oncogenesis, has shown significant enrichment with p-values as low as 2.90 × 10^{-52} in renal cancer samples treated with , highlighting its role in modulating β-catenin activity. As of 2025, recent advances integrate pathway analysis with single-cell (scRNA-seq) to detect cell-type-specific perturbations, such as heterogeneous immune pathway activations within tumor microenvironments, using graph-based models like GSDensity for pathway-centric dissection of transcriptomic heterogeneity.

In proteomics and metabolomics

In , pathway analysis leverages data to assess enrichment in biological pathways affected by post-translational modifications (), particularly in dynamic processes like signaling that influence drug responses. For instance, workflows identify sites on kinases, revealing pathway activations or inhibitions in response to therapeutic agents, such as targeted inhibitors in cancer cells. This approach has been instrumental in characterizing PTM landscapes in , where altered signaling pathways correlate with efficacy and resistance profiles. Interactive tools further enable of PTM dysregulation across signaling cascades, facilitating the pinpointing of regulatory hubs in progression. Pan-cancer studies using have demonstrated widespread PTM alterations in oncogenic pathways, underscoring their role in therapeutic targeting. In , pathway analysis maps profiles from liquid chromatography- (LC-MS) and gas chromatography- (GC-MS) to reconstruct metabolic networks, detecting flux imbalances such as those in the tricarboxylic acid () during . Untargeted has identified intermediates like citrate and α-ketoglutarate as dysregulated in , linking mitochondrial dysfunction to and . These analyses reveal broader network perturbations, including and , providing biomarkers for disease monitoring and intervention. Advanced strategies enhance resolution for low-abundance metabolites, enabling precise pathway mapping in diabetic complications like nephropathy. Multi-omics integration extends pathway analysis by combining and with to trace causal perturbations from genetic variants through protein modifications to metabolite outputs. For example, tools like Pathview overlay genomic, proteomic, and metabolomic data onto pathway maps, visualizing how gene mutations propagate to alter activity and downstream flux in metabolic disorders. This layered approach has elucidated regulatory mechanisms in complex diseases, such as integrating phosphoproteomics with to model signaling-metabolism . Comprehensive frameworks for transcriptomics-proteomics-metabolomics integration emphasize pathway-centric methods to uncover coordinated changes, improving predictive accuracy over single-omics analyses. Distinct challenges in these domains arise from the inherent variability and in protein and measurements, which exceed that in genomic data and demand advanced and imputation techniques for reliable enrichment. data often exhibits batch effects and low reproducibility due to PTM lability, complicating pathway inference. pathway modeling further requires stoichiometric constraints in to account for stoichiometries, yet incomplete coverage and issues hinder accurate reconstructions. In microbiome contexts, pathway analysis of host-pathogen interactions highlights these issues, as seen in recent genome-scale models revealing metabolic exchanges like nutrient competition in gut infections, where noisy multi-omics is essential for dissecting reciprocal influences.

Pathway Databases and Resources

Curated pathway databases

Curated pathway databases compile structured representations of biological pathways, including metabolic, signaling, and regulatory processes, derived from experimental evidence and literature. These databases provide graph-based models that capture interactions, reactions, and relationships among genes, proteins, metabolites, and other biomolecules, enabling detailed analysis of cellular functions. Major databases emphasize manual curation by domain experts to ensure accuracy and integration of supporting evidence, such as IDs (PMIDs) for referenced publications. The Kyoto Encyclopedia of Genes and Genomes (KEGG) is a foundational resource featuring manually drawn pathway maps that represent molecular interaction, reaction, and relation networks across organisms. As of November 2025, KEGG contains 581 pathway maps, including metabolic pathways annotated with Enzyme Commission (EC) numbers for catalytic reactions and signaling pathways detailing regulatory cascades. Curation involves expert annotation of pathways based on genomic and biochemical data, with regular updates incorporating new evidence from literature and experimental studies. Reactome offers a human-centric, peer-reviewed database of detailed reaction steps organized in a hierarchical format, covering 16,002 molecular events (reactions) in processes like and . Pathways are manually curated by biologists using a and linked to evidence from publications via PMIDs, ensuring traceability to primary sources. The database supports through export in formats like BioPAX (Biological Pathway ), a community standard for representing pathways at the molecular and cellular level in OWL XML. As of September 2025 (version 94), Reactome includes 2,825 human pathways with expanded annotations for disease-related processes. WikiPathways is an open, community-driven platform where researchers collaboratively curate and update pathway diagrams using the PathVisio editor, fostering rapid incorporation of emerging knowledge. It hosts 1,913 human pathways across 27 species as of 2023, with ongoing contributions from over 600 individuals, and emphasizes for visualization and analysis. Curation follows a process with literature-backed annotations, including PMIDs, and pathways are available in BioPAX and GPML formats for integration with other tools. To enhance coverage, integrative resources like PathDIP version 5 aggregate data from 12 curated databases, resulting in 6,535 pathways spanning 195,148 genes and 5,783 diseases in humans and 16 other organisms. This integration propagates pathway associations to indirectly connected proteins, addressing gaps in primary databases through propagation algorithms and manual validation. Similarly, the database version 12.5 (released in 2025, building on 2024 updates) incorporates pathway information into its protein association networks, adding regulatory directionality based on curated evidence for over 12,535 organisms. These databases serve as reference sets for pathway enrichment analysis, where gene sets derived from them inform functional interpretation of data.

Gene set collections

Gene set collections consist of predefined lists of genes or gene products grouped based on shared biological attributes, such as functional roles, regulatory mechanisms, or experimental perturbations, serving as resources for in . These collections differ from structured pathway databases by providing flat, unordered groupings that facilitate broad functional interpretation without requiring . The Molecular Signatures Database (MSigDB), maintained by the Broad Institute, is a leading resource with version 2025.1 containing 35,134 human gene sets divided into nine major collections. Hallmark gene sets (H, 50 sets) represent refined, high-confidence signatures of well-defined biological states, such as or , derived from multiple sources. The (GO) collection within MSigDB (C5, 16,228 sets, including 10,480 GO terms) organizes genes into categories for biological processes, molecular functions, and cellular components, enabling analysis of diverse functional annotations beyond pathways. Oncogenic gene sets (C6, 189 sets) capture cancer-related neighborhoods and modules curated from tumor profiles. Gene set types include canonical pathways, compiled from databases like and Reactome as part of MSigDB's collection (7,561 curated sets overall). Computational sets, such as motif-based regulatory targets (), infer gene groupings from sequence motifs or binding sites. Immunologic signatures (C7, 5,219 sets) encompass cell type-specific, state-specific, and perturbation-induced profiles from the ImmuneSigDB compendium. Curation processes emphasize manual annotation from peer-reviewed literature and validated databases, with MSigDB distinguishing categories like (literature- and database-derived) from computationally generated ones like . These collections offer advantages for enrichment analyses by simplifying computations with unordered lists, enabling rapid assessment of functional themes, and accommodating non-pathway groupings like cellular compartments or immunologic states that lack defined interactions. However, they inherently lack the topological details of pathway databases, such as gene regulatory directions or interaction strengths, potentially overlooking nuanced biological relationships.

Methods of Pathway Analysis

Over-representation analysis (ORA)

Over-representation analysis (ORA) is a threshold-based statistical method employed in pathway analysis to evaluate whether genes associated with a specific biological pathway are more prevalent in a list of significant features—such as differentially expressed genes from genomic or transcriptomic data—than would be expected by random chance. This approach is particularly useful for interpreting high-throughput experimental results by identifying enriched pathways that may underlie observed biological phenomena. ORA operates on a binarized gene list, where features are classified as significant or non-significant based on a predefined cutoff, typically an adjusted p-value threshold like 0.05. The core of ORA involves computing the enrichment of pathway genes within the significant list using the or, equivalently, , which models the selection process as sampling without replacement from a finite population. To calculate the one-sided for over-representation, the formula is: p = \sum_{i=k}^{\min(n, N_i)} \frac{\binom{N_i}{i} \binom{N - N_i}{n - i}}{\binom{N}{n}} Here, N represents the total number of genes in the background set (e.g., all genes assayed in the experiment), n is the size of the selected significant gene list, N_i is the number of genes annotated to the pathway of interest, and k is the observed number of overlaps between the significant list and the pathway genes. A low indicates significant over-representation, suggesting the pathway is biologically relevant to the experimental condition. This method was popularized in tools like GO::TermFinder for enrichment. The standard for ORA begins with defining the gene set to provide context for the , ensuring it reflects the experimental scope (e.g., genes detectable by the or sequencing platform). Next, the significant list is generated by applying a statistical to differential analysis results. Enrichment p-values are then computed for each pathway or gene set from curated databases, followed by correction for multiple testing to control the or ; common corrections include the Bonferroni method for stringent control or the Benjamini-Hochberg procedure for FDR. Pathways with corrected p-values below a chosen (e.g., 0.05) are deemed significantly enriched. Key assumptions underlying ORA include the of genes within pathways, meaning correlations or interactions among genes are not accounted for, and the of significance, which discards quantitative information on effect sizes or ranks. These assumptions simplify computation but can introduce biases if violated. For example, the tool has been used in analyses to identify over-representation in and apoptosis-related clusters from differentially expressed genes in treated versus control samples, aiding in functional interpretation. ORA's strengths lie in its straightforward and computational efficiency, enabling rapid of large lists without requiring complex . However, it is limited by its dependence on arbitrary thresholds, which can alter results and discard valuable information from non-significant but directionally consistent genes, and by a tendency to favor larger pathways due to higher baseline overlap probabilities.

Functional class scoring (FCS)

Functional class scoring (FCS) methods represent a category of pathway enrichment analyses that utilize the full spectrum of gene expression data by ranking all genes according to their differential expression statistics, thereby avoiding the need for arbitrary significance thresholds inherent in over-representation analysis (ORA). In FCS, individual gene scores—such as moderated t-statistics derived from linear models for microarray or RNA-seq data using tools like limma—are computed to rank the entire gene list from most up-regulated to most down-regulated. This ranked list is then interrogated for each pathway or gene set to quantify the degree of coordinated perturbation, emphasizing subtle but consistent shifts across multiple genes rather than extreme changes in a few. The core computation in FCS involves deriving an enrichment score (ES) for a given pathway, which aggregates the positions of pathway genes within the ranked list. A prominent variant, as implemented in (GSEA), employs a running-sum statistic resembling a Kolmogorov-Smirnov test to measure deviation from random expectation. The enrichment score is the maximum value of the running sum, calculated by walking down the ranked list L (from most up-regulated to most down-regulated): increase the running sum when encountering a gene in the set S (positive increment) and decrease it for genes out of S (negative increment). In the unweighted version, the increments are +1 for genes in S and -1 for genes not in S, normalized appropriately; significance is assessed via permutation tests. Key FCS methods include GSEA, which relies on this permutation-based framework to detect enriched pathways, and PADOG (Pathway Analysis with Down-weighting of Overlapping Genes), a robust aggregation approach that combines moderated t-test statistics for pathway genes while down-weighting those shared across multiple sets to mitigate overlap biases. In PADOG, the pathway score is the of the values of weighted moderated t-statistics for all s in the pathway, where weights are higher for s appearing in fewer sets, enabling sensitive detection in small sample sizes. Both methods process the ranked or scored list through these aggregation steps, followed by or resampling to derive p-values. An illustrative application of GSEA is in analyzing data to identify enrichment of EMT-related sets in chemoresistant samples, revealing coordinated changes in genes like VIM and CDH2 associated with aggressive behavior. FCS approaches offer advantages in sensitivity to modest, distributed expression changes that might be overlooked by threshold-dependent methods, as they leverage all data points without discarding information. Additionally, by normalizing contributions based on set size and total genes, FCS reduces biases toward large pathways, promoting equitable evaluation across biological processes of varying complexity.

Pathway topology analysis (PTA)

Pathway topology analysis () integrates the structural information of biological pathways, modeled as where nodes represent or proteins and edges denote interactions or regulatory relationships, to assess the impact of experimental data such as changes. Unlike over-representation (ORA) or functional class scoring (FCS), which treat genes within a pathway as equally contributing members, PTA propagates signals along the graph edges to weight the influence of each gene based on its and , thereby capturing how perturbations in upstream components may amplify or attenuate downstream effects. This approach enhances the detection of pathway dysregulation by accounting for the hierarchical and interconnected nature of signaling or metabolic processes. A seminal in PTA is Signaling Pathway Impact Analysis (SPIA), which combines evidence from ORA with a perturbation accumulation score to rank pathways. The perturbation factor P for a pathway is computed as P = \sum_i d_i \cdot |t_i|, where t_i is the or for i, and d_i is a reflecting the gene's position, such as its number of direct targets or a measure like betweenness in directed graphs. SPIA has been benchmarked on various datasets, demonstrating its ability to identify impacted pathways by accounting for , outperforming unweighted in detecting regulatory . Other key PTA methods include DEGraph, which employs Gaussian graphical models and multivariate tests like the Hotelling T^2-statistic to evaluate differential expression across the entire pathway graph, and , which uses factor graphs to infer patient-specific pathway activities by integrating multi-omics data and propagating probabilities through nodes and edges. DEGraph assesses whether the joint distribution of gene expressions differs between conditions while incorporating graph structure to boost statistical power, particularly for small sample sizes. models pathways as probabilistic graphical models, enabling the detection of shifts in pathway states, such as activation or inhibition, in cancer datasets. Topology metrics in PTA commonly include node degree (number of connections), centrality measures (e.g., betweenness for bottlenecks or closeness for influence), and distinctions between directed (e.g., signaling cascades) and undirected (e.g., metabolic interactions) graphs to reflect flow directionality. These metrics allow PTA to prioritize genes with high centrality, such as hubs or gatekeepers, whose dysregulation disproportionately affects pathway function. For instance, in directed pathways, incoming and outgoing edge weights propagate signals asymmetrically, unlike undirected models that assume symmetric interactions. The primary benefits of PTA lie in its ability to account for pathway bottlenecks and interaction dependencies, leading to improved over flat enrichment methods; studies show PTA detects more biologically relevant pathways in benchmark datasets by weighting topological positions. This structured weighting mitigates biases from gene length or expression levels, providing deeper insights into mechanistic dysregulation, such as in contexts where upstream mutations propagate broadly.

Network enrichment analysis (NEA)

Network enrichment analysis (NEA) integrates or other data with molecular interaction to detect enriched subnetworks, extending beyond predefined gene sets by leveraging connectivity in global like protein-protein interaction (PPI) maps. Unlike overlap-based methods, NEA quantifies enrichment through , such as the density of links between query and functional modules, enabling the identification of emergent biological associations not captured by isolated gene lists. This approach typically begins by constructing a comprehensive from resources like the database, which compiles interactions from experimental, computational, and literature sources across thousands of organisms. Key NEA methods employ diffusion or clustering to propagate scores across the network and score connected components. For instance, EnrichNet uses with restart (RWR) on a PPI network, where seed s from the query set initiate with a restart probability of 0.9; the steady-state probability for a v updates iteratively as S_v^{(t+1)} = (1 - \gamma) \sum_u A_{uv} S_u^{(t)} / \deg(u) + \gamma f_v, with \gamma = 0.9 as the restart parameter, A the , and f_v the seed indicator, allowing scores to diffuse and reveal pathway associations via subnetwork visualization. Similarly, PRINCE applies label on PPI networks to prioritize -associated genes and complexes, computing a smooth scoring function by iteratively diffusing priors from known genes across the , achieving high accuracy in gene prioritization for diseases like Alzheimer's. Module detection often incorporates algorithms like Markov Clustering (MCL), which simulates flow in the network to partition it into dense clusters, scoring these for enrichment against query sets to uncover functional modules. In contrast to pathway topology analysis (PTA), which relies on fixed, curated pathway structures with predefined topologies, NEA operates on larger, uncurated networks to discover de novo modules without assuming rigid pathway boundaries, thus capturing indirect or emergent interactions across broader biological contexts. For example, network-based analyses in using PPI networks have identified modules enriched for amyloid processing genes like APP. Recent advancements incorporate directional edges in signed networks to model activation/inhibition, enhancing module specificity in neurodegenerative analyses. Advantages include the ability to reveal novel interactions missed by pathway-centric methods, but NEA is computationally intensive due to network scale and propagation iterations, often requiring efficient randomization for significance testing. Recent advances as of 2025 include machine learning-enhanced NEA methods, such as graph neural networks for enrichment analysis (e.g., GNNenrich), and topology-aware approaches for data, improving detection in complex contexts.

Software and Tools

Open-source tools

Open-source tools for pathway analysis provide accessible platforms for researchers to perform enrichment analyses, visualize results, and integrate multi-omics data without licensing costs, often leveraging community-driven development within ecosystems like . These tools typically support methods such as over-representation analysis (ORA), functional class scoring (FCS), and network enrichment analysis (NEA), enabling users to input gene lists in formats like Ensembl IDs or IDs and generate outputs including p-value-adjusted enrichments and visualizations. The clusterProfiler is a widely used tool for comprehensive enrichment analysis, supporting ORA, FCS, and (GSEA) across thousands of species with up-to-date annotations from databases like MSigDB and . It features the compareCluster function for multi-set comparisons, allowing simultaneous analysis of multiple gene lists to identify overlapping enriched pathways, and produces visualizations such as dot plots and bubble plots for intuitive interpretation. As part of the project, clusterProfiler integrates seamlessly with other packages for downstream analyses, including support for non-coding data. For pathway visualization, the R package pathview enables mapping and rendering of user data onto pathway graphs, facilitating the integration of datasets like or onto predefined diagrams. It automatically handles data parsing and graph rendering, supporting inputs in various formats and producing publication-ready images that highlight node-level changes, such as fold-expression values. Pathview is also integrated into , allowing combination with tools like clusterProfiler for end-to-end workflows. Web-based options like Enrichr offer a user-friendly for gene set enrichment, aggregating libraries from MSigDB, , and over 100 other sources to perform ORA and visualization without installation. Users can upload gene lists via a web form, receiving interactive results including bar graphs, tables of top terms, and combined scores for pathway overlaps. Enrichr supports programmatic access through its , making it suitable for in pipelines. The GSEA desktop application, developed by the Broad Institute, is a standalone tool implementing FCS via the GSEA method, which assesses coordinated changes across ranked lists using metrics like normalized enrichment scores (NES). It processes microarray or RNA-seq data, supports custom sets, and generates detailed reports with heatmaps and leading-edge analyses to identify core enriched . The application is freely downloadable and runs on multiple operating systems, with updates maintaining compatibility with recent MSigDB releases. For multi-omics integration, the mixOmics provides multivariate methods like sparse partial least squares (sPLS) to combine datasets from , , and transcriptomics prior to pathway analysis, reducing dimensionality while preserving biological relevance. It supports pathway-level projections through integration with enrichment tools, enabling identification of correlated features across layers for downstream NEA or . Recent advancements include the STRING API update in 2025, which enhances NEA by incorporating directionality in protein-protein interaction networks and a new geneset_description function for automated enrichment on input sets, drawing from over 20 billion interactions across 12,535 . Similarly, WebGestalt 2024 introduces support for via integration with RaMP-DB and multi-omics analysis through multi-list functionality, accelerating ORA with over 600,000 functional categories and interactive visualizations like pathway maps, while improving computational speed for large datasets. In practice, users should standardize inputs to common identifiers (e.g., HGNC symbols for genes) to avoid mapping errors, and leverage Bioconductor's vignette tutorials for combining tools like clusterProfiler with pathview to generate plots overlaid on pathway diagrams. These open-source resources foster through version-controlled code and community forums.

Commercial solutions

Commercial solutions for pathway analysis primarily consist of platforms designed for pharmaceutical, , and clinical applications, offering integrated, user-friendly interfaces with extensive curated databases and advanced predictive capabilities. These tools emphasize , such as adherence to FDA guidelines for variant interpretation and reporting, and provide dedicated support for high-throughput data integration. Ingenuity Pathway Analysis (IPA), developed by , stands as a leading commercial platform, leveraging a comprehensive, human-curated encompassing over 200,000 biological molecules and millions of relationships derived from . It enables users to overlay experimental —such as or —onto pathways, facilitating the identification of upstream regulators, downstream effects, and potential drug targets through causal network modeling. Key features include predictive algorithms for biomarker discovery and toxicity assessment, with built-in tools like Analysis Match that align user datasets against more than 100,000 publicly available analyses for contextual insights. IPA's supports intuitive of pathway perturbations, making it particularly advantageous for pharmaceutical target validation and preclinical , where it accelerates generation while ensuring for regulatory submissions. In pharmaceutical applications, has been instrumental in dissecting COVID-19-related pathway perturbations, such as analyzing serum and to reveal dysregulated and pathways in infected patients, aiding in drug repurposing efforts. For instance, integrative analyses using identified key molecular signatures in host responses, supporting the validation of antiviral targets in early 2020s studies. MetaCore, offered by Clarivate as part of the Cortellis Drug Discovery Intelligence suite, provides an integrated web-based platform for multi-omics pathway analysis, allowing researchers to upload and interpret data from sources like next-generation sequencing and microarrays within curated disease maps and signaling pathways. It features advanced network-building tools for exploring changes in the context of biological processes, including and mechanism-of-action predictions, with a focus on accelerating innovation in by linking omics results to therapeutic opportunities. MetaCore's advantages lie in its seamless integration with Clarivate's broader intelligence resources, such as compound libraries and data, offering a polished for collaborative workflows and compliance with industry standards for reproducible analyses. Similar to , has supported research in pharma, where meta-transcriptomic analyses extracted drug targets from pathway databases, identifying 46 repurposable compounds by mapping to host pathways. As of 2025, commercial platforms continue to evolve with enhancements; notably, launched Interpret in late 2024, an extension that automates the interpretation of complex by generating narrative summaries of pathway activations and regulatory effects, building on the platform's and Summer 2025 releases that expanded content for enhanced analyses. The Fall 2025 release of further refines these -driven features for faster downstream predictions. While benefits from Clarivate's overarching infrastructure for in research intelligence, specific pathway prediction updates remain integrated within its multi-omics toolkit without standalone modules announced in 2025.

Limitations and Challenges

Annotation and coverage issues

One major challenge in pathway analysis stems from the incomplete annotation of genes and proteins within pathway databases, which limits the reliability of enrichment results. For instance, the database annotates approximately 36% of the roughly 20,000 human protein-coding genes, covering about 7,200 genes across its pathways, leaving a substantial portion unrepresented in comprehensive maps. This gap is even more pronounced for non-model organisms, where functional annotations are often insufficient due to limited experimental data and underrepresentation in major databases, hindering cross-species analyses. Annotation biases further exacerbate coverage issues, primarily arising from literature-driven priorities that favor well-studied biological processes. Pathways related to common diseases like cancer receive disproportionate attention, leading to overrepresentation of genes involved in oncogenesis while rare diseases and niche functions remain underexplored. Additionally, species-specific gaps persist, as many databases are human-centric or focused on model organisms such as Mus musculus or Saccharomyces cerevisiae, resulting in sparse coverage for microbial or plant pathways that may interact with human systems. These shortcomings directly impact pathway analysis outcomes, often producing false negatives in enrichment detection where relevant pathways are overlooked due to missing gene assignments. Consequently, analyses tend to overemphasize , well-annotated pathways, skewing interpretations toward familiar biological mechanisms and potentially missing novel or context-specific insights. For example, human-centric databases like frequently lack detailed microbial pathways from the , despite their critical role in host metabolism and immunity, leading to incomplete models of microbe-host interactions. Efforts to mitigate these issues include community-driven initiatives such as WikiPathways, which encourage collaborative curation to expand and update annotations beyond traditional databases. By 2025, advances in have further supported automated annotation, with integrations of tools like enabling predictions of protein structures and interactions to infer pathway components in under-annotated regions.

Computational and methodological limitations

Pathway analysis methods, particularly over-representation analysis (ORA), often rely on the assumption of gene independence, which is frequently violated due to biological interactions and pathway , leading to biased enrichment scores and inflated false positives. This assumption treats genes as modular entities without accounting for their interconnected roles in networks, resulting in siloed analyses that overlook how perturbations in one pathway propagate to others. Functional class scoring (FCS) methods exacerbate this by equally weighting all genes within a pathway, ignoring varying contributions to overall activity. Statistical challenges further compound these issues, including the need for rigorous multiple testing corrections when analyzing large sets, which can drastically reduce statistical power and increase type II errors. In small sample sizes, common in experimental studies, methods like ORA and FCS lack sufficient power to detect subtle pathway enrichments, often requiring larger cohorts for reliable results. Pathway analysis () and network enrichment analysis (NEA) face similar hurdles, as their reliance on aggregation neglects effect sizes, potentially overlooking biologically meaningful but statistically modest changes. Computationally, PTA and NEA can be intensive for network-based inferences involving a large number of genes or interactions, making them challenging for large-scale datasets without optimization. In the context of from single-cell , processing millions of cells demands GPU acceleration and to handle sparsity, zero-inflation, and batch effects, as traditional CPU-based implementations become infeasible. Annotation gaps can indirectly worsen these computational burdens by necessitating additional preprocessing to align incomplete pathway maps. Additional limitations include the over-reliance on static pathway representations, which fail to capture dynamic biological processes such as temporal regulation or context-specific interactions, limiting applicability to evolving systems like disease progression. Reproducibility is undermined by variability in permutation-based null distributions, where gene permutations assume independence and can yield inconsistent results across studies due to dataset-specific correlations. Looking ahead, hybrid approaches integrating with statistical methods, such as embedding-based models for pathway clustering, promise to address assumption violations and enhance interpretability in multi-omics data. Cloud-based platforms for scalable processing will facilitate handling of single-cell and longitudinal datasets, while efforts to incorporate dynamic modeling could better align analyses with biological reality.

References

  1. [1]
    Pathway Analysis: State of the Art - PMC - PubMed Central
    Abstract. Pathway analysis is a set of widely used tools for research in life sciences intended to give meaning to high-throughput biological data.
  2. [2]
    Identifying significantly impacted pathways: a comprehensive review ...
    Oct 9, 2019 · This article presents the most comprehensive comparative study on pathway analysis methods available to date.
  3. [3]
    Pathway Analysis Interpretation in the Multi-Omic Era - MDPI
    In bioinformatics, pathway analyses are used to interpret biological data by mapping measured molecules with known pathways to discover their functional ...<|control11|><|separator|>
  4. [4]
    comprehensive survey of the approaches for pathway analysis using ...
    In this article, we review 32 approaches that have been designed for the purpose of integrative pathway analysis in the multi-omics and/or multi-cohort setup.Introduction · Pre-analysis · Integrative pathway analysis · Summary and discussion
  5. [5]
    Gene set enrichment analysis: A knowledge-based approach for ...
    In this paper, we provide a full mathematical description of the GSEA methodology and illustrate its utility by applying it to several diverse biological ...
  6. [6]
    Ten Years of Pathway Analysis: Current Approaches and ... - NIH
    Feb 23, 2012 · We discuss the evolution of knowledge base–driven pathway analysis over its first decade, distinctly divided into three generations. We also ...
  7. [7]
    Pathway and Network Analysis Workflow - GitHub Pages
    Jul 1, 2019 · The main purpose of pathway and network analysis is to understand what a list of genes is telling us, ie gain mechanistic insights and interpret lists of ...
  8. [8]
    Gene Set Analysis: Challenges, Opportunities, and Future Research
    The presence of such a large number of multi-functional genes means single-gene analysis may lead to false or ambiguous conclusions. Single-gene approach may ...
  9. [9]
    Using graph theory to analyze biological networks - BioData Mining
    Apr 28, 2011 · SBML can represent metabolic networks, cell signaling pathways, regulatory networks, and many other kinds of systems [50]. Other file ...
  10. [10]
    Tutorial on biological networks - Wiley Interdisciplinary Reviews
    Jun 22, 2012 · In this section, we will present an overview of five types of biological networks: metabolic networks, gene regulatory networks, PPI networks, ...
  11. [11]
  12. [12]
    Systematic comparison and assessment of RNA-seq procedures for ...
    Nov 12, 2020 · RNA-seq data analysis typically involves several steps: trimming, alignment, counting and normalization of the sequenced reads, and, very often, ...
  13. [13]
    RNA-seq: questions and answers - STAR Protocols - Cell Press
    May 4, 2023 · You can use tools like DESeq2, edgeR, or limma for bulk RNA-seq data. Functional analysis: Conduct functional analysis to identify enriched ...
  14. [14]
    Pathway Analysis for Genome-wide Genetic Variation Data
    In this review, we focus on pathway analysis, also known as gene-set enrichment analysis, which stands at the forefront of the latest GWAS discoveries.Missing: paper | Show results with:paper
  15. [15]
    Transcriptome analysis reveals upregulation of immune response ...
    Jan 12, 2022 · Comprehensive pathway analysis showed enrichment of genes related to tumour functions such as inflammation, angiogenesis and metabolism at the ...
  16. [16]
    SNP-based pathway enrichment analysis for genome-wide ...
    SNP-based pathway enrichment analysis for GWAS involves selecting representative SNPs for each gene, then ranking and testing if pathway SNPs are significantly ...
  17. [17]
    A comprehensive analysis of Wnt/β-catenin signaling pathway ... - NIH
    We aimed to investigate the effect of As2O3 treatment on Wnt/β-catenin signaling pathway-related genes and pathways in renal cancer. Illumina-based RNA-seq ...
  18. [18]
    Pathway centric analysis for single-cell RNA-seq and spatial ...
    Dec 18, 2023 · We present GSDensity, a graph-modeling approach that allows users to obtain pathway-centric interpretation and dissection of single-cell and spatial ...
  19. [19]
    Proteomic characterization of post-translational modifications in drug ...
    Mass spectrometry-based proteomics technologies serve as a powerful approach for system-wide characterization of PTMs, which facilitates the identification of ...
  20. [20]
    PTMNavigator: interactive visualization of differentially regulated ...
    Jan 8, 2025 · Post-translational modifications (PTMs) play pivotal roles in regulating cellular signaling, fine-tuning protein function, and orchestrating ...
  21. [21]
    Pan-cancer analysis of post-translational modifications reveals ...
    Aug 14, 2023 · Post-translational modifications (PTMs) play key roles in regulating cell signaling and physiology in both normal and cancer cells.
  22. [22]
    Metabolomics uncovers the diabetes metabolic network
    Sep 4, 2025 · The coupling of MS with liquid chromatography (LC) or gas chromatography (GC) significantly improves metabolite separation and identification.
  23. [23]
    Advanced Mass Spectrometry-Based Biomarker Identification for ...
    May 27, 2024 · This paper summarizes advanced mass spectrometry for the application of metabolomics in diabetes mellitus, gestational diabetes mellitus, diabetic peripheral ...
  24. [24]
    PaintOmics 4: new tools for the integrative analysis of multi-omics ...
    May 24, 2022 · PaintOmics is a web server for the integrative analysis and visualisation of multi-omics datasets using biological pathway maps.Missing: Pathview | Show results with:Pathview
  25. [25]
    Strategies for Comprehensive Multi-Omics Integrative Data Analysis ...
    Oct 22, 2024 · In this article, we review the methods used for integrating transcriptomics, proteomics, and metabolomics data and summarize them in three approaches.
  26. [26]
    Metabolic Modeling of Host-Microbe Interactions - ScienceDirect
    Oct 3, 2025 · In this review, we examine recent applications of GEMs to host-microbe studies, with a focus on how they reveal reciprocal metabolic influences.
  27. [27]
    Current State, Challenges, and Opportunities in Genome-Scale ...
    Jun 28, 2024 · In the past decade, several frameworks have been introduced to incorporate proteome-related limitations using a genome-scale stoichiometric ...
  28. [28]
    KEGG: Kyoto Encyclopedia of Genes and Genomes
    KEGG is a database resource for understanding high-level functions and utilities of the biological system, such as the cell, the organism and the ecosystem.KEGG PATHWAY Database · KEGG Database · KEGG Software · KEGG Overview
  29. [29]
    Reactome Pathway Database: Home
    Reactome is pathway database which provides intuitive bioinformatics tools for the visualisation, interpretation and analysis of pathway knowledge.Manually · Pathway Browser · Download · Analysis Tools
  30. [30]
    WikiPathways: Home
    WikiPathways is an open science platform for biological pathways contributed, updated, and used by the research community. Read more Video tour.About · WikiPathways Data · Search · Analyze
  31. [31]
    KEGG PATHWAY Database
    KEGG PATHWAY is a collection of manually drawn pathway maps representing our knowledge of the molecular interaction, reaction and relation networks.Kegg network · KEGG Compound · KEGG Disease · KEGG Objects
  32. [32]
    KEGG - Current Statistics - GenomeNet
    Current Statistics. KEGG Database as of 2025/11/6. Systems information. KEGG PATHWAY, KEGG pathway maps, 581. KEGG BRITE, BRITE hierarchies and tables, 203.
  33. [33]
    KEGG: biological systems database as a model of the real world - NIH
    Oct 17, 2024 · KEGG (https://www.kegg.jp/) is a database resource for representation and analysis of biological systems. Pathway maps are the primary dataset in KEGG.
  34. [34]
    Reactome: a database of reactions, pathways and biological ... - NIH
    Reactome is an open source, open access, manually curated, peer-reviewed pathway database of human pathways and processes (1). Pathway annotations are created ...
  35. [35]
    The Reactome Pathway Knowledgebase 2024 - Oxford Academic
    Nov 6, 2023 · The Reactome Knowledgebase systematically links human proteins to their molecular functions, providing a resource that is both a textbook of ...
  36. [36]
    BioPAX – A community standard for pathway data sharing - PMC
    BioPAX (Biological Pathway Exchange) is a standard language to represent biological pathways at the molecular and cellular level.
  37. [37]
    WikiPathways 2024: next generation pathway database - PMC
    Nov 6, 2023 · WikiPathways contains a total of 1913 human-curated and reviewed pathways for 27 species. As a result of our unique approach to community ...
  38. [38]
    WikiPathways: connecting communities | Nucleic Acids Research
    Nov 19, 2020 · WikiPathways is a database of biological pathway models collected and curated by the research community. Anyone at any time can contribute ...
  39. [39]
    PathDIP 5: improving coverage and making enrichment analysis ...
    Nov 22, 2023 · Pathway Data Integration Portal (PathDIP) is an integrated pathway database that was developed to increase functional gene annotation coverage ...
  40. [40]
    pathDIP 4: an extended pathway annotations and enrichment ... - NIH
    Nov 16, 2019 · PathDIP 4 now integrates 24 major databases. To further reduce the number of proteins with no curated pathway annotation, pathDIP integrates ...
  41. [41]
    The STRING database in 2025: protein networks with directionality ...
    Jan 6, 2025 · The latest version, STRING 12.5, introduces a new 'regulatory network', for which it gathers evidence on the type and directionality of ...Missing: v12 | Show results with:v12
  42. [42]
    STRING: functional protein association networks
    ... 12,535 organisms. Includes 59.3 million proteins with over 20 billion interactions. A Core Data Resource as designated by. SEARCH. ↑. © STRING Consortium 2024.
  43. [43]
    MSigDB - GSEA
    MSigDB is a resource of annotated gene sets for GSEA software, allowing users to examine, browse, and search gene sets.Mouse MSigDB Collections · Human MSigDB Collections · Miscellaneous gene sets
  44. [44]
    Human MSigDB Collections - GSEA
    The 35134 gene sets in the Human Molecular Signatures Database (MSigDB) are divided into 9 major collections, and several subcollections.Mouse MSigDB Collections · Browse Human Gene Sets · Browse 16228 gene sets
  45. [45]
    MSigDB | Browse Human Gene Sets - GSEA
    MSigDB offers human gene sets by name, first letter, or collection, including hallmark, positional, curated, pathways, regulatory, and ontology sets.
  46. [46]
    tool for the unification of biology. The Gene Ontology Consortium
    Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat Genet. 2000 May;25(1):25-9. doi: 10.1038/75556. Authors. M Ashburner ...
  47. [47]
    Human MSigDB Collections: Details and Acknowledgments - GSEA
    ImmuneSigDB is composed of gene sets that represent a broad curation ... Copyright (c) 2004-2025 Broad Institute, Inc., Massachusetts Institute of ...
  48. [48]
    Pathway Analysis vs Gene Set Analysis | Advaita Bioinformatics
    in essence, pathways are models describing the interactions of genes, proteins, or metabolites within cells, tissues, or organisms, not simple lists of genes.
  49. [49]
    Prior biological knowledge-based approaches for the analysis of ...
    We will compare these methods and discuss their assumptions and pros and cons in Section 4. 3.1 Over-representation analysis. Over-representation analysis ...Missing: paper | Show results with:paper
  50. [50]
    The DAVID Gene Functional Classification Tool: a novel biological ...
    The DAVID gene functional classification tool uses a novel fuzzy clustering algorithm to condense a list of genes or associated biological terms into organized ...
  51. [51]
    A comprehensive survey of the approaches for pathway analysis ...
    This article points out pros and cons of integrative pathway analysis methods, as well as assesses each method's practicality, and discusses outstanding ...
  52. [52]
    A general modular framework for gene set enrichment analysis
    Feb 3, 2009 · Ackermann, M., Strimmer, K. A general modular framework for gene set enrichment analysis. BMC Bioinformatics 10, 47 (2009). https://doi.org ...
  53. [53]
    Ten Years of Pathway Analysis: Current Approaches and ...
    Feb 23, 2012 · This paper discusses the evolution of pathway analysis methods of high-throughput molecular measurements in the last decade, distinctly ...<|control11|><|separator|>
  54. [54]
    Identification of hub genes associated with EMT-induced ... - PubMed
    Jan 30, 2022 · GSEA revealed that EMT-related genes sets were enriched in the CR samples. Further, we found that EMT-induced breast cancer cells showed ...
  55. [55]
    Methods and approaches in the topology-based analysis ... - Frontiers
    Oct 9, 2013 · This review covers 22 such topology-based pathway analysis methods published in the last decade. We compare these methods based on: type of pathways analyzed.
  56. [56]
    A novel signaling pathway impact analysis - PMC - NIH
    We describe a novel signaling pathway impact analysis (SPIA) that combines the evidence obtained from the classical enrichment analysis with a novel type of ...
  57. [57]
    A novel signaling pathway impact analysis - PubMed - NIH
    Jan 1, 2009 · We describe a novel signaling pathway impact analysis (SPIA) that combines the evidence obtained from the classical enrichment analysis with a novel type of ...
  58. [58]
    Inference of patient-specific pathway activities from multi ...
    We describe a PGM framework based on factor graphs (Kschischang et al., 2001) that can integrate any number of genomic and functional genomic datasets to infer ...
  59. [59]
    A comparative study of topology-based pathway enrichment ...
    Nov 4, 2019 · This comparative study of nine network-based methods for pathway enrichment analysis aims to provide a systematic evaluation of their performance based on ...
  60. [60]
    A critical comparison of topology-based pathway analysis methods
    Here, we assessed the performance of seven representative methods identifying differentially expressed pathways between two groups of interest based on gene ...
  61. [61]
    Network enrichment analysis: extension of gene-set enrichment ...
    Sep 11, 2012 · We developed a method of network enrichment analysis (NEA) that extends the overlap statistic in GEA to network links between genes in the experimental set.Methods · Gene Network · Functional Gene Sets
  62. [62]
    EnrichNet: network-based gene set enrichment analysis
    We introduce an integrative analysis approach and web-application called EnrichNet. It combines a novel graph-based statistic with an interactive sub-network ...Abstract · MOTIVATION · SYSTEM AND METHODS · RESULTS AND DISCUSSION
  63. [63]
    Associating Genes and Protein Complexes with Disease via ... - PMC
    Jan 15, 2010 · Here, we provide a global, network-based method for prioritizing disease genes and inferring protein complex associations, which we call PRINCE.<|control11|><|separator|>
  64. [64]
    MCL - a cluster algorithm for graphs - Micans
    MCL, or Markov Cluster Algorithm, is a fast, scalable, unsupervised cluster algorithm for graphs based on simulation of flow in graphs.Missing: enrichment pathway
  65. [65]
    Benchmarking enrichment analysis methods with the disease ...
    This approach identifies Network Enrichment Analysis methods as the overall top performers compared with overlap-based methods.
  66. [66]
    Unveiling Gene Interactions in Alzheimer's Disease by Integrating ...
    Apr 1, 2024 · An over-representation analysis of the genes in the top 10% of dmGWAS modules revealed the enrichment of GO BP terms related to amyloid-beta ...2. Results · 2.2. Ad-Associated Top... · 3. Discussion<|control11|><|separator|>
  67. [67]
    NEAT: an efficient network enrichment analysis test
    Sep 5, 2016 · NEAT is a flexible and efficient test for network enrichment analysis that aims to overcome some limitations of existing resampling-based tests.
  68. [68]
    clusterProfiler 4.0: A universal enrichment tool for interpreting omics ...
    Aug 28, 2021 · clusterProfiler supports exploring functional characteristics of both coding and non-coding genomics data for thousands of species with up-to-date gene ...
  69. [69]
    Pathview: an R/Bioconductor package for pathway-based data ... - NIH
    Pathview is a novel tool set for pathway-based data integration and visualization. It maps and renders user data on relevant pathway graphs.
  70. [70]
    Pathview: An R package for pathway based data integration and ...
    The pathview R package is a tool set for pathway based data integration and visualization. It maps and renders user data on relevant pathway graphs.Overview · Examples · Installation
  71. [71]
    pathview package - Bioconductor
    No information is available for this page. · Learn why
  72. [72]
    Enrichr - Ma'ayan Laboratory, Computational Systems Biology
    Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinformatics. 2013; 128(14).Enrichr-KG · API Documentation · Login · WormEnrichrMissing: clusterProfiler | Show results with:clusterProfiler
  73. [73]
    GSEA-P: a desktop application for Gene Set Enrichment Analysis
    Abstract. Gene Set Enrichment Analysis (GSEA) is a computational method that assesses whether an a priori defined set of genes shows statistically signific.
  74. [74]
    mixOmics – From Single to Multi-Omics Data Integration
    mixOmics is an R package for integrating omics data, using multivariate methods to reduce data dimensionality and integrate single or multiple datasets.Network() · The mixOmics package · mixOmics Publications · The mixOmics teamMissing: pathway | Show results with:pathway
  75. [75]
    mixOmics: An R package for 'omics feature selection and multiple ...
    We introduce mixOmics, an R package dedicated to the multivariate analysis of biological data sets with a specific focus on data exploration, dimension ...
  76. [76]
    The STRING database in 2025: protein networks with directionality ...
    Nov 18, 2024 · The new API function, named geneset_description, requires only a set of genes as input and automatically performs enrichment analysis. It ...
  77. [77]
    WebGestalt 2024: faster gene set analysis and new support for ...
    May 29, 2024 · For the pathway analysis of metabolomics data, RaMP-DB is used as our primary source of pathway data. This database aggregates human metabolic ...Data update and support for... · Multi-list analysis · Performance improvements...
  78. [78]
    QIAGEN Ingenuity Pathway Analysis
    Use Ingenuity pathway analysis to predict downstream effects, identify new targets or candidate biomarkers and more. Click here to read more!RNA-seq Analysis Portal · Analyze with IPA · IPA Training · Product Login
  79. [79]
    New QCI Interpret 2025 Release enhances variant filtering ...
    Feb 14, 2025 · The QCI Interpret 2025 release delivers updates to enhance efficiency, flexibility and accuracy in your variant interpretation workflow.
  80. [80]
    [PDF] Unlock the power in your 'omics datasets - QIAGEN Digital Insights
    QIAGEN Ingenuity Pathway Analysis. (IPA) with Analysis Match automatically aligns analyses against over 100,000 curated publicly available datasets. It allows ...Missing: advantages | Show results with:advantages
  81. [81]
    The secrets to pathway analysis - Bioinformatics Software
    Oct 8, 2020 · Pathway analysis identifies interconnected genes and functional changes. QIAGEN IPA uses a large, updated database and predicts pathway ...Missing: definition | Show results with:definition
  82. [82]
    Integrative multi-omics approach for identifying molecular signatures ...
    Jun 12, 2023 · IPA on proteins selected from SIDA revealed that the gluconeogenesis I and glycolysis I pathways are determined to be associated with COVID-19.
  83. [83]
    Metacore - Integrated pathway analysis for multi-OMICs data
    MetaCore is a web-based bioinformatics suite that allows researchers to upload data analysis results from experiments such as microarray, next generation ...
  84. [84]
    MetaBase & MetaCore Bioinformatics Data Solutions - Clarivate
    MetaCore has given us the opportunity to confidently explore new biological pathways and has massively increased the value of our RNA-seq datasets.Analyze Your Biological Data... · Access The Critical Data You... · Want To Learn More?
  85. [85]
    Drug Discovery & Development Tools | Clarivate
    Molecular pathway analysis & bioinformatics. Understand the meaning of your omics data in the context of healthy and diseased molecular pathways, and ...Actionable Insights To... · Our Solutions · Drug Discovery And...
  86. [86]
    Drug repurposing for COVID-19 based on an integrative meta ...
    Drug database, extracted from the Metacore and IPA, identified 15 drug targets (with information on COVID-19 pathogenesis) with 46 existing drugs as potential- ...Missing: pharma | Show results with:pharma
  87. [87]
    QIAGEN Launches AI-Extension of Ingenuity Pathway Analysis for ...
    Dec 12, 2024 · Ingenuity Pathway Analysis Interpret extends analysis capabilities of human-curated knowledge base with AI technology.Missing: advantages | Show results with:advantages
  88. [88]
    Now available: QIAGEN IPA Summer Release
    Jul 11, 2025 · The 2025 Summer Release of QIAGEN Ingenuity Pathway Analysis (IPA) is here, delivering enhanced features and expanded content to improve ...Missing: integration | Show results with:integration
  89. [89]
    The QIAGEN IPA Fall Release is here - Bioinformatics Software
    Oct 16, 2025 · The Fall Release of QIAGEN Ingenuity Pathway Analysis is now available, with new features and improvements for enhanced 'omics analyses.Missing: integration | Show results with:integration<|separator|>
  90. [90]
    Systematic assessment of pathway databases, based on a diverse ...
    Sep 10, 2022 · Functional enrichment analysis, also termed 'gene set enrichment analysis', is a widely used approach to accomplish this. It is typically ...
  91. [91]
    Establishing genome sequencing and assembly for non-model and ...
    Apr 17, 2025 · For non-model organisms, structural and functional annotation can be difficult due to insufficient evidence and underrepresentation in databases ...
  92. [92]
    Functionally Enigmatic Genes in Cancer: Using TCGA Data to Map ...
    Mar 5, 2020 · Moreover, these genes are not missing at random but reflect that our information about genes is gathered in a biased manner: poorly studied ...
  93. [93]
    Impact of outdated gene annotations on pathway enrichment analysis
    Aug 7, 2025 · Enrichment tools often rely on humancentric or model organisms databases, creating challenges for applications in non-model systems including ...
  94. [94]
    New insights from uncultivated genomes of the global human gut ...
    Mar 13, 2019 · The genome sequences of many species of the human gut microbiome remain unknown, largely owing to challenges in cultivating microorganisms ...
  95. [95]
    Integrating artificial intelligence in drug discovery and early ... - NIH
    Mar 14, 2025 · AI models, such as AlphaFold, predict protein structures with high accuracy, aiding druggability assessments and structure-based drug design. AI ...Ai In Target Identification · Drug Discovery With... · Limitations Of Ai Applied To...<|separator|>
  96. [96]
    On the influence of several factors on pathway enrichment analysis
    Choice of gene set collection or pathway database. The selection of one gene set collection over another can lead to different results. Some collections or ...
  97. [97]
    [PDF] Sample Size and Reproducibility of Gene Set Analysis
    Abstract—Gene set analysis is widely used to gain insight from gene expression data. Achieving reproducible results is a fundamental part of any expression ...
  98. [98]
    Network modeling of single-cell omics data - Portland Press
    Here, we discuss the existing network modeling approaches developed for bulk tissue omics data, the unique challenges imposed by single-cell omics data for use ...