Skip to main content

CTpathway: a CrossTalk-based pathway enrichment analysis method for cancer research

Abstract

Background

Pathway enrichment analysis (PEA) is a common method for exploring functions of hundreds of genes and identifying disease-risk pathways. Moreover, different pathways exert their functions through crosstalk. However, existing PEA methods do not sufficiently integrate essential pathway features, including pathway crosstalk, molecular interactions, and network topologies, resulting in many risk pathways that remain uninvestigated.

Methods

To overcome these limitations, we develop a new crosstalk-based PEA method, CTpathway, based on a global pathway crosstalk map (GPCM) with >440,000 edges by combing pathways from eight resources, transcription factor-gene regulations, and large-scale protein-protein interactions. Integrating gene differential expression and crosstalk effects in GPCM, we assign a risk score to genes in the GPCM and identify risk pathways enriched with the risk genes.

Results

Analysis of >8300 expression profiles covering ten cancer tissues and blood samples indicates that CTpathway outperforms the current state-of-the-art methods in identifying risk pathways with higher accuracy, reproducibility, and speed. CTpathway recapitulates known risk pathways and exclusively identifies several previously unreported critical pathways for individual cancer types. CTpathway also outperforms other methods in identifying risk pathways across all cancer stages, including early-stage cancer with a small number of differentially expressed genes. Moreover, the robust design of CTpathway enables researchers to analyze both bulk and single-cell RNA-seq profiles to predict both cancer tissue and cell type-specific risk pathways with higher accuracy.

Conclusions

Collectively, CTpathway is a fast, accurate, and stable pathway enrichment analysis method for cancer research that can be used to identify cancer risk pathways. The CTpathway interactive web server can be accessed here http://www.jianglab.cn/CTpathway/. The stand-alone program can be accessed here https://github.com/Bioccjw/CTpathway.

Background

Over 15 years, significant efforts have been made to annotate the functions of individual genes and construct higher order functional knowledgebases, such as the Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) [1, 2]. However, it still remains quite challenging to systematically interpret biological meaning from the expression changes of thousands of genes in a specific model system, such as disease versus control. To determine this, a routinely used method is the screening of differentially expressed genes (DEGs) followed by pathway enrichment analysis (PEA).

PEA methods could be categorized into four generations [3]. First-generation methods (e.g., DAVID [4], WebGestalt [5], and several others [6,7,8]) usually conduct over-representation analysis (ORA) using a hypergeometric or Fisher’s exact test to assess whether the number of input DEGs is significantly higher than that of the genes expected by chance. However, these ORA methods have several limitations. Based on an arbitrary threshold, these methods only select the DEGs that have large expression fold changes (FC) or significant P-values and treat each selected gene equally. Consequently, these methods achieve highly inconsistent results with small changes (e.g., 1.5 FC versus 2.0 FC) in thresholds.

To address these limitations, the second-generation methods called functional class scoring (FCS) were developed [9]. FCS methods hypothesize that even though changes of individual genes are small in magnitude, their coordinated expression changes may have a greater impact in modulating a pathway/gene set [10, 11]. A well-known FCS method is the gene set enrichment analysis (GSEA) [11]. GSEA first ranks genes by differential expression FC. Enrichment scores (ESs) are then calculated for predefined gene sets (pathways or functional gene sets) by considering how well the gene sets are enriched at the top or bottom of the ranked gene lists, which indicate their activation or repression, respectively. Therefore, FCS methods address the limitations of ORA methods.

Previous studies hypothesized that genes with different topological properties have different weights for the linked pathways [12,13,14]. Because topology information of pathways was used, pathway topology-based (PT) approaches were demonstrated to perform better than the previous approaches and regarded as the third generation of PEA methods [15]. The method CePa calculated the weight of a pathway node based on the network centralities [13]. SPIA considered the influence of the neighboring nodes [14]. TPEA integrated the global upstream/downstream positions and the degrees of all nodes in pathways [12]. A significant drawback of PT methods is that they analyze pathways independently and neglect pathway crosstalk, a common and critical event in biology and disease development.

From the perspective of systems biology, genes may iteratively affect many other genes that exist in multiple pathways, causing pathway crosstalk that accounts for the phenotypes, such as crosstalk between ERK and WNT signaling in tumorigenesis [16]. The latest generation of the PEA method is network topology-based (NT) approaches, which consider pathway crosstalk systematically in a network, such as latent pathway identification analysis (LPIA) [17] and pathways based on network information (PathNet) [18]. LPIA regarded each pathway as one node to construct an edge-weighted pathway network based on shared GO functions and DEGs. LPIA identified pathways by random walk algorithm according to network topology. Although LPIA considered pathway crosstalk, it ignores internal topology property within the pathway. PathNet integrated direct evidence (gene differential expression) and indirect evidence (neighbor gene differential expression), which considered gene interactions in both inter- and intra-pathway, as combined evidence for genes to assess their impacts on pathways. However, the gene interactions only depended on directed neighbors in the pathway network, and it ignored the impact of other genes.

As more biological knowledge was gained, protein-protein interaction (PPI) network and gene expression data were used to detect crosstalk between pathways [19, 20]. Recently, by integrating pathway information, PPI network and gene expression data, Kelder et al. identified indirect associations between pathways in insulin-resistant mouse liver [21]. However, in addition to PPI, transcription factor (TF) regulations also provide additional valuable information about molecular interactions. Additionally, these methods performed enrichment analysis mostly based on KEGG or GO, while numerous high-quality pathways or functional gene sets were also publicly available, such as Reactome [22], PANTHER [23], HumanCyc [24], INOH [25], NetPath [26], PID [27], and WikiPathways [28]. Moreover, NT methods consume more time and more space because of their complexity. For example, LPIA may consume several hours for one test. Therefore, although commonly and widely used, current PEA methods have significant limitations, posing barriers to discovery.

In this study, we provided a new NT method for gene enrichment analysis called CTpathway: a crosstalk-based PEA method in a global pathway crosstalk map (GPCM) (Fig. 1). To obtain better speed, our method was optimized for running time to less than 1 min. Compared with existing methods, including DAVID, GSEA, TPEA, PathNet, and LPIA, CTpathway outperformed in terms of accuracy, robustness, and running time. In addition, CTpathway identified several important cancer pathways, which were not identified by other methods. Furthermore, CTpathway was useful even for data sets with fewer DEGs. By applying CTpathway for several cancer types of different stages (I, II, III, and IV), cancer target pathways were identified in early-stage tissues and blood samples. For breast cancer (BRCA) single-cell RNA-seq (scRNA-seq) data, CTpathway could identify the cell type-related pathways. We also developed an online web tool (http://www.jianglab.cn/CTpathway/) and the stand-alone program (https://github.com/Bioccjw/CTpathway) [29], which allows users to simply upload the gene symbols or entrez gene IDs with log2FC or P-values to identify risk pathways in a specific condition (e.g., disease) by performing the CTpathway method.

Fig. 1
figure 1

The workflow diagram for CTpathway. a GPCM and crosstalk effect matrix construction. GPCM was constructed by integrating pathway, TF regulation, and PPI information. Then, we evaluated the crosstalk effects in GPCM by applying the multi-RWR algorithm to calculate a crosstalk effect matrix C. b Gene differential expression (DE) score calculation. We integrated FC and P-value to calculate DE scores. Next, risk score (RS) for each gene and pathway score (PS) for each pathway were calculated to identify the significant risk pathways. c Visualization of significant pathways by bar graph, bubble plot, and enrichment map

Methods

Pathway data

We collected eight knowledgebases of human pathways including KEGG [1], PANTHER [23], Reactome [22], HumanCyc [24], INOH [25], NetPath [26], PID [27], and WikiPathways [28]. The interactions of gene and gene products in pathways of Reactome, HumanCyc, INOH, NetPath, PID, and WikiPathways were obtained from Pathway Commons version 10 [30]. Because the information of KEGG and PANTHER in Pathway Commons was not updated, we extracted the interactions in KEGG and PANTHER in March 2019. For KEGG pathways, we downloaded KGML files of 299 pathways and extracted interaction information by iSubpathwayMiner R package [31]. For PANTHER pathways, we downloaded BioPAX files of 138 pathways, and NetPathminer R package [32] was used to extract interaction information. For the other six sources of pathways, we used gene interactions in Pathway Commons. In total, we obtained 375,256 interactions, including 11,556 genes from 2563 pathways involved in eight pathway databases (details in Table 1).

Table 1 Summary of data source information in GPCM

TF-gene regulation data

We obtained experimentally validated TF-gene regulations from the TRANSFAC Professional database (release: February 2014) [33]. TF-gene regulations, of which at least one node belongs to a pathway, were retained, including 491 TFs, 1614 genes, and 4657 pairs of regulation (Table 1).

PPI data

We obtained PPIs from 12 sources (Additional file 1: Table S1) collected by previous researchers [34, 35]. To obtain more reliable information, PPIs included in ≥2 sources were retained. Furthermore, we used interactions of which at least one of the interacting nodes belongs to a pathway. Finally, 79,262 PPIs, including 11,054 genes, were used for the next analysis (Table 1).

Constructing a global pathway crosstalk map (GPCM)

To consider pathway enrichment more systematically, we integrated three kinds of interactions including pathways, PPIs, and TF regulation for constructing a GPCM to simulate natural pathway crosstalk and adding biological knowledge. We used the union of pathway, PPI, and TF-gene information described above. For simplicity here, it was regarded as an undirected network. In total, the network includes 15,292 nodes and 442,439 edges (details in Table 1).

Gold standard data sets

For comparing CTpathway accuracy with other PEA methods, we used gold standard data sets from the KEGGdzPathwaysGEO R package [36]. It contained 24 data sets involving 12 diseases and 12 target pathways (Additional file 1: Table S2). One disease is corresponding to one target pathway. This data set was widely used as the gold standard for benchmarking in other methods [15, 36, 37]. In addition, to test whether CTpathway could be applied in data sets with fewer DEGs, we analyzed 12 of 24 gold standard data sets with different numbers of DEGs (details in Additional file 1: Table S3).

Cancer data sets from Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) databases

To evaluate reproducibility, we used both microarray data from the GEO database [38] and RNA-seq data from the TCGA database [39] for each of four cancer types (COAD, LIHC, LUAD, and OV). We downloaded eight gene expression data of four cancer types from the GEO database (GSE100179 [40], GSE101685 [41], GSE116959 [42], GSE9891 [43]) and TCGA database (Additional file 1: Table S2). Each data set includes case (cancer) and control (normal) samples (details in Additional file 1: Table S2).

Furthermore, to test whether CTpathway could be applied in early-stage cancer samples, we analyzed ten data sets consisting of different cancer types available in TCGA or GEO (GSE20189; peripheral blood samples of LUAD patients [44]) database. Each data set includes cancer samples of different cancer stages (I, II, III, and IV) and normal samples (details in Additional file 1: Table S2).

To test whether CTpathway could be applied in scRNA-seq data, we downloaded breast cancer (BRCA) scRNA-seq data from the GEO database (GSE118389 [45]) (Additional file 1: Table S2). The BRCA scRNA-seq data contains 1112 cells from six triple-negative breast cancer patients. Here, we used cell type annotation results according to the previous study [45] including B cell, T cell, endothelial cell, epithelial cell, macrophage, and stromal cell. More details about the data sets are shown in Additional file 1: Table S2.

Differential expression

For the GEO microarray data set, we performed differential expression analysis by R package limma [46] to obtain FC and P-value. For the TCGA RNA-seq data set, we used R package DESeq2 [47] to obtain FC and P-value. OV data differential expression profile was from a previous study [48]. For BRCA scRNA-seq data, we performed differential expression analysis between one cell type and the others using function "FindAllMarkers" in R package Seurat V3.2.2 [49]. For some compared methods, which need a set of genes as input, such as DAVID, genes with FC > 2 or FC < 0.5 and P-value < 0.05 were used for functional enrichment analysis. For LPIA, |log2FC| value was used as differential expression score.

Gene differential expression score

The FC and P-value are both important indexes to reflect the differential expression level of genes. Previous studies demonstrated that incorporating FC and P-value could provide significant improvement to meet the practical needs [50, 51]. We calculated a gene differential expression score to represent the impact of its disrupted expression on pathways. P-value ranges from 0 to 1. In order to make the FC value between 0 and 1, and keep genes with an FC value with n and 1/n having the same contribution weight, the gene differential expression score was calculated by Eq. (1).

$$DE=\left\{\begin{array}{l}{\left(1-P\right)}^{\alpha}\times {\left[1-{(FC)}^{-\frac{1}{2}}\right]}^{\beta }, FC\geq 1\\ {}{\left(1-P\right)}^{\alpha}\times {\left[1-{(FC)}^{\frac{1}{2}}\right]}^{\beta },0\le FC< 1\end{array}\right.$$
(1)

When P-value (or FC) is available or not, α (or β) equals to 1 or 0. DE is the differential expression score, represented as a vector:

$$DE=\left[\begin{array}{c}{DE}_1\\ {}{DE}_2\\ {}\vdots \\ {}{DE}_i\\ {}\vdots \\ {}{DE}_L\end{array}\right]$$
(2)

where L is the intersection number of genes in the expression profile and genes in the GPCM.

Risk score (RS)

Here in GPCM, for one gene, we calculated a risk score integrating all the nodes (genes) impact on this node (gene). The GPCM was defined as a simple undirected graph G = (V, E), where a v V represents a gene and a e E represents an edge. First, one gene in the expression file was taken as a seed (i.e., i), and given an initial weight score of 1. Then, a random walk with restart (RWR) algorithm [52] was used to simulate the propagation process of crosstalk effect Ci from one to others.

$${C}_i^t =r\times W\times{C}_i^{t-1} +\left(1-r\right)\times {N}_i$$
(3)

where \({C}_i^1= {N}_i\), r is the restart coefficient, t is iteration times, W was a |N| × |N| column-normalized adjacent matrix of graph G, and Ni is a |N| × 1 vector with ith element equal to 1 and others all equal to 0. Next, with respect to all the genes in the expression file, we iterated over each gene as a seed. This process was called a multiple random walk with restart (multi-RWR) algorithm (Fig. 2). Here, we measured the magnitude of change between states t and t-1 as the sum of the absolute difference of the Ct and Ct-1. The threshold was set as 10−10 to control the iteration times. When it was less than 10−10, the iterative computation would stop. Finally, we obtained a crosstalk effect for all genes, named as the crosstalk effect matrix, represented as a matrix:

$$C={\left[\begin{array}{cccccc}{C}_{11}& {C}_{12}& \cdots & {C}_{1j}& \cdots & {C}_{1N}\\ {}{C}_{21}& {C}_{22}& \cdots & {C}_{2j}& \cdots & {C}_{2N}\\ {}\vdots & \vdots & \vdots \vdots \vdots & \vdots & \vdots \vdots \vdots & \vdots \\ {}{C}_{i1}& {C}_{i2}& \cdots & {C}_{ij}& \cdots & {C}_{iN}\\ {}\vdots & \vdots & \vdots \vdots \vdots & \vdots & \vdots \vdots \vdots & \vdots \\ {}{C}_{L1}& {C}_{L2}& \cdots & {C}_{Lj}& \cdots & {C}_{LN}\end{array}\right]}^T$$
(4)

where Cij represents the crosstalk effect of gene j impacted by gene i. Last, we integrated C matrix and DE vector to calculate the risk score (RS) as follows:

$$RS=C\times DE$$
(5)
Fig. 2
figure 2

CTpathway algorithm diagram including Multi-RWR. First, gene 1 in the expression profile was taken out as a seed and RWR was used to obtain the crosstalk effect C1 on all nodes in the network. Next, another gene i was chosen to repeat this progress and obtained Ci. Finally, we obtained the crosstalk effect matrix C after all genes in the profile were taken out as a seed. We also calculated the differential expression (DE) score by integrating FC and P-value. Using both crosstalk and differential expression, we obtained the risk score \({RS}_{j_i}\) of gene j impacted by gene i. We integrated the risk score of gene j impacted by all genes as gene j risk score RSj. Finally, we obtained a pathway risk score (PS) by averaging all gene risk scores in a pathway and calculated the significance level by permutation

For example, we calculated an RS of gene j impacted by gene i as follows:

$${RS}_{j_i}={C}_{ij}\times {DE}_i$$
(6)

where \({RS}_{j_i}\) represents the risk score of gene j impacted by gene i and DEi represents the differential expression score of gene i.

For gene j, we integrated scores impacted by all genes as the final gene risk score (RSj) as follows:

$${RS}_j =\sum\limits_{i=1}^L{RS}_{j_i}$$
(7)

where L is the intersection number of genes in the expression profile and genes in the GPCM.

The pathway enrichment score

We obtained RS of each gene in GPCM. For a pathway k, we calculated a pathway enrichment score PSk as the average of RS values for the genes in pathway k. The formula is as follows:

$${PS}_k =\frac{1}{n}\sum\limits_{j=1}^n{RS}_j$$
(8)

where n represents the number of genes in pathway k.

Identification of significant pathways

We performed the permutation analysis to estimate the significance of the pathway. First, we shuffled genes in the differential expression profile. Then, we calculated the pathway enrichment score for each pathway. The background distribution was generated after performing h permutations. For a pathway, the empirical P-value was defined as the proportion of random pathway enrichment scores (PSrandom) larger than the real pathway enrichment score (PS): P-value = \({}^{\left({N}_{PS_{random}> PS}\right)\!}\left/ \!{}_{h}\right.\), where \({N}_{PS_{random}> PS}\) was the number of random pathways that had larger scores than the real pathway. Here, h was 1000. However, because of the limited number of permutations, it often produces a P-value of 0. To solve this problem, we estimated the exact P-value by using the generalized Pareto distribution (GPD) [53]. Because many pathways were involved in this analysis, it was necessary to perform multiple hypothesis testing methods to control the proportion of false positives. We applied the false discovery rate (FDR) to account for false positives [54]. The pathways with FDR < 0.01 were considered as significant pathways. In addition, CTpathway automatically clusters significant pathways into non-redundant groups. Pairwise similarities between any two significant pathways are computed based on a Jaccard similarity coefficient. According to user’s input cutoff of Jaccard similarity coefficient, a pathway similarity network is constructed. A default coefficient of 0.3 was set up in this study, which could be customized by the users using our web server. The Markov Cluster (MCL) algorithm [55] was employed to perform clustering process. CTpathway chooses the most significant (lowest FDR) pathway within each cluster to represent the cluster. To obtain a better visualization, CTpathway shows the top 20 non-redundant pathways or clusters with low FDR, if there are more than 20 clusters or pathways. For each cluster, the top 10 pathways with lower FDR are shown in the enrichment map if there are more than 10 pathways which are within one cluster.

Rank difference (DR) and time difference (DT) values

DR value was calculated to represent the rank difference (before and after optimization) of the target pathway as follows:

$$DR=\sum\limits_{m=1}^M\frac{\left|{R}_{b_m}-{R}_{a_m}\right|}{K}/M$$
(9)

where \({R}_{b_m}\) represents the rank of the target pathway in data set m before optimization and \({R}_{a_m}\) represents the rank of the target pathway in data set m after optimization. M is the number of data sets. K is the number of total KEGG pathways. Here, M is 24 and K is 299.

DT value was calculated to represent the running time difference (before and after optimization) as follows:

$$DT=\sum\limits_{m=1}^M\frac{T_{b_m}-{T}_{a_m}}{T_{b_m}}/M$$
(10)

where \({T}_{b_m}\) represents the running time in data set m before the optimization, and \({T}_{a_m}\) represents the running time in data set m after the optimization.

Rank ratio (RR) value

RR value was used as the criteria to compare the accuracy of different tools. Each data set m had an RR value for its target pathway, represented as RRm, which was the rank ratio of the target pathway in data set m, and calculated as follows:

$${RR}_m=\frac{R_{a_m}}{M}$$
(11)

where \({R}_{a_m}\) and M were described as above. To make it comparable between different methods, we used KEGG pathways as candidate pathways.

Stability (S) value

S value was used as the criteria to compare the stability or reproducibility of different tools. First, for eight data sets of four cancer types, each of which has microarray data and RNA-seq data, we identified risk pathways by using different methods. For each cancer type, we compared shared significant pathways identified from microarray data and RNA-Seq data. Because compared methods just identified few pathways or no pathways when using routine FDR or adjusted P-value as a cutoff, pathways with P-value < 0.05 were identified as significant pathways for all methods here. S value was calculated as follows:

$$S=\sum\limits_{d=1}^D\frac{J_d}{D}$$
(12)

where Jd represents the Jaccard similarity coefficient of different data sets of cancer type d and D represents the number of all cancer types. In this study, D is 4.

Statistics analysis

Differential expression analysis was performed by R package limma for the GEO data set and R package DESeq2 for the TCGA data set. P-value < 0.05 was considered to be statistically significant for DEGs. For CTpathway, permutation analysis was performed to estimate the significance of the pathway; FDR < 0.01 was considered as significant. For GSEA, FDR < 0.01 was considered as significant. For other compared methods, P-value < 0.05 was considered as significant because few pathways or no pathways were identified when using routine FDR or adjusted P-value as a cutoff.

Benchmarking

In this study, CTpathway was compared with five widely used tools, including DAVID, GSEA, TPEA, LPIA, and PathNet, in terms of accuracy, reproducibility, and running time. For accuracy, we compared RR values for each method using 24 gold standard data sets (Additional file 1: Table S2). For reproducibility, we compared the S value calculated for four cancer types (COAD, LIHC, LUAD, and OV) based on different sources (TCGA RNA-seq data and GEO microarray data) of eight gene expression data (Additional file 1: Table S2). For the running time, we used simulated data sets of 500, 1000, 5000, 10,000, and 20,000 genes. Because most of these methods only focused on the pathways defined in KEGG, we used KEGG pathways for comparative analysis when benchmarking.

Hardware platform

All benchmarks were performed on a computer with 2*Intel Xeon E5-2609 V4 Processor, 2*64G DDR4 RDIMM, 8 DIMM slots, 1*128G SSD 2.5, 1*2TB SATA 3.5, and 2*1080Ti.

Code availability

CTpathway web server is available at http://www.jianglab.cn/CTpathway/. The CTpathway stand-alone program is available at https://github.com/Bioccjw/CTpathway [29]. Other custom codes used in this study are available from the corresponding authors upon reasonable request.

Results

Global pathway crosstalk map (GPCM) and its properties

By integrating three kinds of interactions including the regulation of TFs to genes from TRANSFAC [33], the PPIs from multiple sources in previous studies [34, 35], and the pathways from eight databases (KEGG [1], Reactome [22], PANTHER [23], HumanCyc [49], INOH [25], NetPath [26], PID [27], and WikiPathways [28]), we constructed a GPCM that included 15,292 nodes and 442,439 edges (Fig. 3a and Table 1). Next, we investigated the topological properties of the GPCM. The degree distribution approximately displayed a power law distribution (Fig. 3b), indicating the network satisfied scale-free topology, a general concept for biological networks. There are some well-known signaling and transcription factor genes with a high degree in the GPCM, such as EGFR, AKT, MYC, and p53 (Additional file 1: Table S4). The gene with the highest degree in the network is GNB1, a subunit of G proteins, which are modulators or transducers in various transmembrane signaling pathways and included in 234 pathways. In addition, we determined that ~75% (n > 8800) of the genes participate in more than one pathway (Fig. 3c). Density distribution of pathways showed a positively skewed distribution, which suggested that only a few pathways include a higher number of genes (Fig. 3d). Most of the genes participate in multiple pathways, which suggest that crosstalk exists. Pathway crosstalk was represented by integrating molecular interactions and pathways into a GPCM.

Fig. 3
figure 3

Overview and characteristics of the GPCM. a GPCM. Different colors represent different kinds of nodes. Gray dots represent genes from at least two kinds of interactions (pathway, PPI, or TF regulation). b The degree distribution of GPCM. c The cumulative distribution of the number of genes in pathways. The pie chart shows the proportion of genes in one pathway and in more than one pathway. d The density distribution of the number of pathways

Crosstalk effect evaluation and pathway identification

Pathways are usually affected by each other in the process of performing functions due to crosstalk [16]. We evaluated the crosstalk effects in GPCM by applying a multi-RWR algorithm to calculate a crosstalk effect matrix, C, which exploits the complete network topology (Fig. 2) (details in the “Methods” section). Then, we integrated FC and P-value as gene differential expression score (DE) to reflect the disturbed level of gene expression (details in the “Methods” section). Next, we integrated the C matrix and differential expression score (DE) to calculate a risk score (RS) as the impact of the gene on the pathways (Fig. 2, details in the “Methods” section). For gene i, RSi reflects the risk score of the node i in the context of GPCM. We further tested the relationship between RS and |log2FC| based on a lung adenocarcinoma (LUAD) data set (Additional file 1: Table S2) available in the GEO database (GSE116959 [42]). Despite a higher positive correlation (Pearson correlation coefficient R ≈ 0.84) (Additional file 2: Fig. S1), we determined several known lung cancer-associated genes with high RS and low |log2FC| (Table 2 and Additional file 2: Fig. S1), such as TRIM28, APP, ESR1, MYC, and EGFR [56,57,58,59]. However, these genes would be overlooked by most of the existing PEA methods because they only consider significant DEGs or high |log2FC| genes.

Table 2 The representative 10 genes with high RS and low |log2FC| value in LUAD (GSE116959)

Additionally, we questioned whether RS would reflect gene risk better than |log2FC|. First, we downloaded cancer causal genes (CCGs) from the Cancer Gene Census (CGC) [60]. We obtained CCGs for four cancer types (COAD, LIHC, LUAD, and OV) separately (Additional file 1: Tables S2 and S5). Then, we obtained two gene expression data sets for each of these cancer types from two independent sources (TCGA and GEO, Additional file 1: Table S2) and performed differential expression analysis. For each data set, we ranked genes according to their |log2FC| and RS from high to low, separately. Next, we evaluated if CCGs were located in the top of the rank list by the GSEA method [11]. The results showed that CCGs were significantly located in the top of the RS rank list for all 8 data sets at a significance level of FDR <0.1, whereas all |log2FC|-based FDRs were >0.1 (Additional file 2: Fig. S2 and Additional file 1: Table S6). Here, the CCGs with low |log2FC| achieved high RS through crosstalk with those high |log2FC| genes in GPCM. These results indicated that RS was a better index for identifying casual genes, and thus, pathways enriched with high RS genes are likely to have important roles. Moreover, the proportion of risk genes in the top 100 of the RS rank list with |log2FC| <1 varied from 17 to 60% for eight data sets. This set of high-risk genes would have been overlooked if only considering the DEG analysis (Additional file 2: Fig. S3).

Finally, we calculated a pathway enrichment score, PS, by integrating the RS of all nodes in the pathway. We took the average of the RS values in a pathway k as PSk. By permutation, we identified the significant dysregulated pathways (details in the “Methods” section).

Parameter optimization and improved performance compared to existing tools

We tested the performance of different r values based on 24 gold standard data sets involving 12 human diseases (Additional file 1: Table S2). In general, there was a slight variance on the performance with different r values. In this study, r was set as 0.7 because CTpathway had the best performance (Fig. 4a). In addition, we only kept ε digits and set values smaller than 10ε to 0 for the C matrix to improve running speed. Here, the threshold ε was set to 3, which consequently completes the job in less than 50 s (86.3% reduction of running time) without compromising the quality of the results (rank difference = 0.018) (Fig. 4b).

Fig. 4
figure 4

CTpathway outperforms other methods. a Box plot of target pathway RR values for different r values. b The impact of different ε values (x-axis) on DT (left y-axis) and DR (right y-axis) values. cf Comparative analysis of the performance of different methods in terms of accuracy (RR and ROC curve), reproducibility, and running time, respectively. “*” represents two-sided t-test P-value < 0.05; “**” represents P-value < 0.01; “***” represents P-value < 0.001; ns represents not significant

To illustrate the effectiveness of the proposed method in identifying dysregulated pathways, our results were compared with five widely used tools, including DAVID (first-generation method) [4], GSEA (second-generation method) [11], TPEA (third-generation method) [12], LPIA (fourth-generation method) [17], and PathNet (fourth-generation method) [18]. Because most of these methods only focused on the pathways defined in KEGG, we used KEGG pathways for this comparative analysis (details in the “Methods” section).

First, accuracy was compared by using 24 gold standard data sets (Additional file 1: Table S2) [36]. We compared the RR values of the target pathways obtained from different tools (Fig. 4c). CTpathway had the significantly lower RR values than other methods (no significant change compared to PathNet), indicating that our method was more accurate. Moreover, the comparisons of ROC curves and AUC values also indicated CTpathway had the best performance (Fig. 4d).

Reproducibility is also very important. Currently, most of the PEA methods are not sufficiently reproducible because of only using DEGs and insufficiently using pathway topology and molecular interaction information. To evaluate the stability of the methods, we calculated the S value (details in the “Methods” section) for four cancer types (COAD, LIHC, LUAD, and OV) based on different sources (TCGA RNA-seq data and GEO microarray data) of eight gene expression data sets (Additional file 1: Table S2). The results showed that CTpathway achieved the highest S value in all comparisons (Fig. 4e). Therefore, our data showed that CTpathway outperformed other tools in generating reproducible results.

Next, we compared the running time of CTpathway with other methods such as GSEA, TPEA, PathNet, and LPIA. Because DAVID was used on the web server, the running time of which might be interfered by the internet connection speed, it was excluded. We used simulated data sets of 500, 1000, 5000, 10,000, and 20,000 genes. Our results demonstrated that CTpathway outperformed other methods, particularly as gene number increased. As the number of genes rose, increased running time was observed in TPEA, PathNet, and GSEA, whereas no change in running time occurred in CTpathway and LPIA (Fig. 4f). However, LPIA running time was days compared to CTpathway, which took less than 50 s to analyze one set of data regardless of gene number, demonstrating that our method was independent of gene set size. Taken together, our data show that CTpathway has greater accuracy, higher reproducibility, and less running time compared to other methods.

CTpathway identifies risk pathways in cancers

To demonstrate the utility of CTpathway, we firstly applied it to eight gene expression data sets of four tumor types (COAD, LIHC, LUAD, and OV; Additional file 1: Table S2). In eight pathway databases, we identified significant pathways for the four tumor types at a significance level of FDR < 0.01 (Fig. 5a and b, and Additional file 1: Table S7-S14). The number of identified pathways in different pathway databases differed. In general, the number of identified pathways in the Reactome database was relatively higher because of more candidate pathways. The total number of significant pathways for the eight gene expression data varied from approximately 300 to 500, accounting for 11.7~19.5% of all candidate pathways (Fig. 5b). Some well-known cancer pathways were significant in more than one cancer type (Fig. 5a and Additional file 1: Table S7-S14). For example, the “AP-1 transcription factor network” was identified as a significant pathway across four cancer types over all eight data sets. The AP-1 transcription factor is involved in a wide range of biological processes, such as cell growth, proliferation, differentiation, apoptosis, migration, and invasion [61,62,63,64]. “FOXM1 transcription factor network,” “Degradation of the extracellular matrix,” and “Activation of matrix metalloproteinases” appeared in seven data sets. Many previous studies have demonstrated that these pathways are altered in multiple cancer types, indicating their pan-cancer regulation potential [65,66,67,68,69,70]. We also identified pathways unique to a single cancer. For example, transport-related pathways (“Transferrin endocytosis and recycling” and “Passive transport by Aquaporins”) were reproducibly identified in COAD in both the GSE100179 [40] and TCGA patient cohort and not in other cancers. Many metabolism-linked pathways (“Pyruvate metabolism,” “Glycerolipid metabolism,” “Glycogen degradation II,” “Acetate conversion to acetyl-CoA,” and “Caffeine metabolism”) were specifically identified in LIHC patient cohorts in TCGA and GEO (GSE101685 [41]). Previous reports demonstrated that a large number of metabolic processes are dysregulated in LIHC to fuel tumorigenesis [71], suggesting our method accurately identifies dysregulated pathways in cancer. Several transcription or signal transduction-related pathways (“RUNX1 regulates transcription of genes involved in differentiation of HSCs,” “NOTCH1 Intracellular Domain Regulates Transcription,” “Constitutive Signaling by NOTCH1 PEST Domain Mutants”) were shared by GEO (GSE116959 [42]) and TCGA LUAD patient cohorts, but not in other cancers. In OV, immune and EMT/migration/invasion-related pathways were observed, such as “TCR,” “IL3,” “E-cadherin signaling in the nascent adherens junction,” “RUNX2 regulates genes involved in cell migration,” “Adherens junction,” and “Stabilization and expansion of the E-cadherin adherens junction” pathways. Importantly, these pathways have been reported to impact cell and/or organ functions and/or tumorigenesis [72,73,74,75,76,77,78,79,80,81,82]. Collectively, our results show that CTpathway accurately identifies well-known cancer risk pathways.

Fig. 5
figure 5

CTpathway accurately identifies well-known cancer risk pathways. a The comparison of identified pathways in four cancers for eight data sets. Gray color represents identified pathways across multiple cancer types (AC). Other colors represent cancer type-specific identified pathways. GT represents both GEO and TCGA data sets; TS and GS represent TCGA specific and GEO specific, respectively. For cancer type-specific identified pathways, the proportion of pathways for different data sets is shown as pie charts. b Heatmap of pathways identified for eight cancer data sets by CTpathway in eight pathway databases. The number of candidate pathways for each database is indicated in parentheses. c The bar graph shows the top 10 KEGG pathways

Most of the pathways are previously verified known risk pathways for the individual cancer types, indicating CTpathway is a highly reliable tool for prioritizing the risk pathways. Taking GEO LUAD and OV data sets as examples, all the top 10 pathways for LUAD and nine of the top 10 pathways for OV have been reported (Fig. 5c, Table 3 and Additional file 1: Table S15-S19). We also compared risk pathways identified by different methods. We determined that all of the top 10 risk pathways for both LUAD and OV in CTpathway were also identified by other methods (Table 3 and Additional file 1: Table S15-S19). However, four of the top 10 pathways for LUAD or OV were identified by only one or two existing methods (Additional file 1: Table S15-S19), such as “Tuberculosis,” “Focal adhesion,” and “ECM-receptor interaction,” which play critical roles in cancer pathogenesis or progression [83,84,85]. Moreover, we determined the pathways only identified by CTpathway for OV (Table 4 and Additional file 1: Table S17-S19), and most of these pathways were cancer-related such as “MAPK signaling pathway” [86, 87], “Wnt signaling pathway” [88], and “Hippo signaling pathway” [89]. We also determined that these pathways had a lower proportion of DEG (Additional file 2: Fig. S4). For example, CTpathway identified “MAPK signaling pathway” (Table 4), which has important roles in the development and survival of many cancer types including ovarian cancer [86, 87]. Also, there is a crosstalk between “MAPK signaling pathway” and “ECM-receptor interaction” (Fig. 6a), which had been demonstrated to aid in EMT/migration/invasion process [90,91,92]. Five of six methods including CTpathway identified “ECM-receptor interaction” as a risk pathway (Table 3); however, all of the other compared methods were unable to determine “MAPK signaling pathway” as a risk pathway in EMT in OV. Furthermore, we determined that there was a lower proportion of DEGs in the “MAPK signaling pathway” (1.4% [4/295]) than that in the “ECM-receptor interaction” pathway (18.5% [15/81]); thus, most methods will identify “ECM-receptor interaction” instead of “MAPK signaling pathway.” Because the “MAPK signaling pathway” has crosstalk with “ECM-receptor interaction,” with most DEGs (14/15) in the “ECM-receptor interaction” having a direct connection with the “MAPK signaling pathway,” most “MAPK signaling pathway” genes have high RS (Fig. 6a, b). Therefore, only our method identified “MAPK signaling pathway” as a risk pathway. Moreover, we determined that, in the top 100 of the RS rank list, there are 36 EMT genes [93], of which seven have low |log2FC| (|log2FC| < 1) (Fig. 6c and Additional file 1: Table S20). These genes were easily overlooked by other methods that only considered DEGs as risk genes. Taken together, CTpathway could identify cancer risk pathways that were identified by existing methods, and importantly, also significant pathways and risk genes that were overlooked by other methods.

Table 3 Top 10 significant pathways identified by CTpathway based on the GSE116959 LUAD data set
Table 4 Top 10 significant pathways only identified by CTpathway based on the GSE9891 OV data set
Fig. 6
figure 6

“MAPK signaling pathway” aid in EMT in OV. a Crosstalk between “MAPK signaling pathway” and “ECM-receptor interaction.” The red nodes represent DEGs and the gray nodes are non-DEGs. Fourteen of 15 DEGs in the “ECM-receptor interaction” pathway have a direct connection with the “MAPK signaling pathway.” b The risks of genes in the “MAPK signaling pathway.” Risks were measured by FC, FDR, DEG, and RS. c The bar graph shows EMT genes in the top 100 of the RS rank list in OV

CTpathway identifies risk pathways in data sets with fewer DEGs

Because of its algorithmic properties, we postulated that CTpathway would be useful for data sets with a small number of DEGs. To test this, we screened DEGs for 24 gold standard data sets at a level of |log2FC| > 1 and FDR < 0.1 and selected 12 representative gold standard data sets with different numbers of DEGs ranging from 0 to 1702 (Fig. 7a and Additional file 1: Table S3). We compared KEGG pathways identified by CTpathway with those by other five methods (DAVID, GSEA, TPEA, PathNe, and LPIA) at a significance level of FDR-corrected P-value < 0.05 (Fig. 7a). The number of significant pathways identified by CTpathway was independent from the number of DEGs. For data sets with fewer DEGs, CTpathway could identify more pathways than all other methods. However, other methods, including DAVID, GSEA, and TPEA, showed a greater dependency on the number of DEGs. They could only identify a small number of significant pathways for data sets with fewer DEGs (e.g., GSE6956C [94] and GSE1297 [95]). Furthermore, CTpathway could identify target pathways for most (9/12) of the data sets, whereas other methods had a lower rate of identification and overlooked them, especially for data sets with fewer DEGs. We also compared significant pathways at a level of nominal P-value < 0.05 (Additional file 2: Fig. S5), and CTpathway could identify target pathways independent on the number of DEGs. These results demonstrated that CTpathway outperformed other methods for target pathway identification, particularly when there are a small number of DEGs.

Fig. 7
figure 7

CTpathway identifies significant pathways in data sets with few DEGs and early-stage cancer patients. a The comparison of identified pathways for data sets with a different number of DEGs in six methods. The bar graph shows the number of DEGs for 12 representative data sets. The heatmap shows the number of significant pathways at the significance level of FDR < 0.05, identified by different methods for data sets with a different number of DEGs, divided by the number of all candidate pathways. The target pathways are marked as green stars. b The heatmap of enrichment results (P-value) for the target pathways of 10 early-stage cancer types using different methods. c, d Enrichment results (P-value, y-axis) for the target pathways of TCGA COAD data sets of tissue samples (c) and LUAD blood samples (d) of different stages using different methods. The dashed line represents a cutoff (P-value = 0.05)

CTpathway identifies risk pathways in early-stage cancer

Cancer diagnosis relies on detecting symptoms followed by histology/pathology evaluation. Identification of altered pathways indicative of pre-malignancy or early-stage cancer is critical for disease prevention and earlier treatment, leading to improved outcomes for patients. Early-stage cancers usually show smaller changes at the molecular level than late-stage cancers. We tested whether CTpathway could identify risk pathways for early-stage disease in cancer patients. First, samples of 10 cancer types that included stages I, II, III, and IV were obtained from TCGA. We selected the KEGG annotated ten pathways specific for the ten cancer types (Additional file 1: Table S2). For each cancer type of each stage, pathway enrichment analysis was performed by CTpathway and other methods. The P-values of target pathways were compared by different methods. The results showed that CTpathway performed better than the other methods for tissue samples (Fig. 7b, c and Additional file 2: Fig. S6). In general, the CTpathway P-values of target pathways were smaller than those of other methods. Even in the early-stage (stage I) patients, 9/10 target pathways of cancer types could be identified by CTpathway at a significance level of P-value < 0.05, whereas all but one other method could either not identify any or only identified one target pathway in early-stage patients for one cancer type (Fig. 7b). In addition, we also evaluated blood samples, which are easier to obtain from patients as compared to tissue samples. CTpathway was applied to the data sets of the blood samples (GSE20189 [44]) from LUAD patients of different stages (I, II, III, and IV), and it identified the target pathway in the early stage as well as performed better than the other methods across all cancer stages (Fig. 7d). These results demonstrate that CTpathway may be useful for early disease diagnosis.

CTpathway identifies cell type-related pathways in scRNA-seq data

Due to characteristics of scRNA-seq data such as drop-out events and low library sizes, the number of DEGs for a subgroup or cell type is typically low. Because CTpathway is not limited by DEG number, we postulated it could be utilized in scRNA-seq data. To test this, we obtained BRCA scRNA-seq data (GSE118389 [45]). Cell types were annotated (B cell, T cell, macrophage, endothelial cell, epithelial cell, and stromal cell) according to the reported study [45]. Differential expression analysis was performed between one cell type and the others by Seurat V3.2.2 [49]. Then, CTpathway was applied to each cell type. The pathway enrichment results showed that CTpathway could identify known cell type-related pathways in each cell type (Fig. 8). For example, “B cell receptor signaling pathway” was significant in B cell (FDR = 1.48×10−6) [96]; “Neurophilin interactions with VEGF and VEGFR” was significant in endothelial cell (FDR = 5.06×10−6) [97]; “Toll-like receptor signaling pathway” was significant in macrophage (FDR = 6.47×10−6) [98]; “ECM-receptor interaction” was significant in stromal cell (FDR = 1.61×10−6) [99]; “TCR” was significant in T cell (FDR = 6.21×10−7) [100]. Compared to other methods, CTpathway showed the lowest RR value for the “B cell receptor signaling pathway” in B cell (Additional file 2: Fig. S7). These results demonstrated that CTpathway could effectively identify cell type-related functions or pathways in scRNA-seq data.

Fig. 8
figure 8

CTpathway identifies cell type-related pathways in BRCA scRNA-seq data. The heatmap of enrichment results (PS value) of pathways in each cell type determined by CTpathway

Reduction of pathway redundancy

Redundancy is a frequently neglected problem for most PEA methods. Pathways sharing genes lead to functional similarities. As a result, it is difficult to extract representative pathways from redundant information [7, 101, 102]. CTpathway automatically clusters enriched pathways into non-redundant groups. Briefly, we constructed a similarity network after obtaining significant pathways based on a particular cutoff of the Jaccard similarity coefficient for shared genes among all significant pathway pairs. MCL clustering algorithm [55] was employed to absorb most redundancies into representative clusters. Each cluster was renamed as the name of the most significant pathway in this cluster. Taking TCGA COAD stage I patient samples as an example (Additional file 2: Fig. S8), we determined some clusters with two or more pathways, and our method enables robust identification of the remaining single node clusters, indicating that these risk pathways reveal potentially targetable pathways, as they have the least amount of crosstalk with other pathways. Therefore, CTpathway is designed to obtain non-redundant pathway information to better interpret pathway enrichment results, and this is dictated according to the needs of the user who input a cutoff of the Jaccard similarity coefficient on the web server.

Web-based implementation of CTpathway

We provided an online web tool for users to perform pathway enrichment analysis with CTpathway (Additional file 2: Fig. S9). Users can input data including gene (gene symbol or entrez ID), both log2FC and P-value or either. By selecting several parameters, input Email address, and clicking the “run” button (Additional file 2: Fig. S9a and b, more details in the Web Manual page), CTpathway returns enrichment results shown in the table in the result page of the web server. The results are also visualized by a bar graph, a bubble plot, and an enrichment map (Additional file 2: Fig. S9c-e). Users can choose any or all results according to their needs. In the web server, results only take a few minutes.

Discussion

PEA is a useful method for exploring gene set function. However, most existing methods did not consider pathway crosstalk and priori knowledge. In this study, we designed and provided to the research community CTpathway, a crosstalk-based PEA method through a global pathway crosstalk map (GPCM) by using multiple sources of pathways and priori knowledge in human.

First, we collected TF-gene regulation, PPI, and gene-gene interaction and constructed a GPCM. The topological property analysis showed that the degree distribution approximately displayed a power law distribution, which was similar to most biological networks. Then, we integrated FC and P-value for each gene from differential expression analysis as gene differential expression score (DE). Next, we obtained a crosstalk effect matrix by the multi-RWR algorithm and calculated a final risk score (RS) by integrating the DE and crosstalk effects. By enrichment analysis of the CGC genes, we demonstrated that RS was a better index for identifying risk genes, and identified important genes with a high RS and low |log2FC| that were overlooked by other methods that relied on |log2FC|. Finally, we calculated a pathway enrichment score by averaging RS for genes in the pathway and identified significantly dysregulated pathways by permutation. Our optimization process reduced ~86.3% of the original running time. Furthermore, the performance of CTpathway is significantly better compared with existing methods in terms of accuracy (RR and AUC value), reproducibility, and running time. In addition, by applying CTpathway to cancer patient samples, we determined that CTpathway could identify critical pathways, which were not identified by other methods. For the data sets with a small number of DEGs, CTpathway was also useful and outperformed the other methods. Notably, CTpathway outperformed other methods in identifying target pathways in early-stage cancer tissues and blood samples. For scRNA-seq data, which can have small DEG numbers, CTpathway could effectively identify cell type-related pathways. Our results demonstrate that CTpathway could be applied in disease analysis, and especially for data sets with fewer DEGs, early cancer diagnosis, which may lead to starting treatment earlier, and scRNA-seq data. We also developed an online web tool to allow users to easily and freely perform PEA with CTpathway.

This study provides a new useful PEA method, CTpathway, for over 2500 pathways in eight pathway databases, and showed that CTpathway performed better than other widely used methods. We evaluated CTpathway performance using the commonly used standard data sets. However, these data sets are limiting because there are only 24 target pathways for 24 diseases, indicating a need in the field for more gold standard data sets for the evaluation of pathway enrichment analysis methods. If the data sets contained additional known risk pathways for diseases, the methods could be evaluated more precisely using the precision-recall curve and AUPRC. In addition, CTpathway still has limitations related to reproducibility, which is consistent with PEA methods overall. For example, when different data sets belonging to the same disease serve as input, the results may differ. While differences in samples and sample handling and processing between different labs contribute to reproducibility challenges, CTpathway was more reproducible than the other methods, showing ~35% overlap between different data sets tested.

Of note, the NT methods are highly dependent on the information of interactions, such as TF-gene regulations, PPIs, and gene-gene interactions, and thus, incomplete information will limit the development of these methods. In this study, TF-gene regulations come from the TRANSFAC database. Recently, several other resources of TF-gene regulation have been provided [103, 104]. Adding more TF-gene regulations might lead to a potential improvement of CTpathway. Notably, CTpathway could be extended to predict non-coding RNA (ncRNA) functions by adding ncRNA regulations or interactions into GPCM. Moreover, CTpathway only focuses on Homo sapiens in this version. Through constructing GPCM for other species, CTpathway could be used to identify risk pathways of other species. Although future studies will be needed to investigate these areas, CTpathway provides a new publicly available method that should result in new discoveries in multiple fields of biology and disease research.

Conclusions

This study presents a novel pathway crosstalk-based method, CTpathway, for performing pathway enrichment analysis. CTpathway outperformed existing methods on accuracy, reproducibility, and speed. CTpathway exclusively identified critical pathways in several cancer types. Furthermore, CTpathway was useful even for data sets with few differentially expressed genes and could identify target pathways in early-stage cancer patient samples, which could lead to earlier treatment, and identify cell type-related pathways for scRNA-seq data. Finally, we provide an interactive and easy-to-use web server so users can conveniently perform pathway enrichment analysis and discover disease-risk pathways.

Availability of data and materials

The CTpathway web server is available from http://www.jianglab.cn/CTpathway/. The CTpathway stand-alone program is available from https://github.com/Bioccjw/CTpathway [29]. All data used could be obtained from public sources (details in Additional file 1: Table S2). Twenty-four gold standard data sets were obtained from the KEGGdzPathwaysGEO R package [36]. Eight gene expression data of four cancer types (COAD, LIHC, LUAD, and OV) were downloaded from the GEO database [38] (https://www.ncbi.nlm.nih.gov/geo/) under accession numbers GSE100179 [40], GSE101685 [41], GSE116959 [42], and GSE9891 [43] and the TCGA database [39] (https://portal.gdc.cancer.gov/). Cancer data of different stages were downloaded from the TCGA (https://portal.gdc.cancer.gov/) and GEO (https://www.ncbi.nlm.nih.gov/geo/, under accession number GSE20189 [44]) databases. BRCA scRNA-seq data were obtained from the GEO database (https://www.ncbi.nlm.nih.gov/geo/) under accession number GSE118389 [45]. Cancer causal genes were obtained from the Cancer Gene Census (CGC) [60] (https://cancer.sanger.ac.uk/census/).

Abbreviations

BRCA:

Breast cancer

C:

Crosstalk effect matrix

COAD:

Colon adenocarcinoma

DE:

Differential expression score

DEGs:

Differentially expressed genes

DR:

Rank difference

DT:

Time difference

ESs:

Enrichment scores

FC:

Fold change

FCS:

Functional class scoring

FDR:

False discovery rate

GEO:

Gene Expression Omnibus

GO:

Gene Ontology

GPCM:

Global pathway crosstalk map

GPD:

Generalized Pareto distribution

GSEA:

Gene set enrichment analysis

KEGG:

Kyoto Encyclopedia of Genes and Genomes

LIHC:

LIVER hepatocellular carcinoma

LPIA:

Latent pathway identification analysis

LUAD:

Lung adenocarcinoma

MCL:

Markov Cluster

multi-RWR:

Multiple random walk with restart

NT:

Network topology-based

ORA:

Over-representation analysis

PathNet:

Pathways based on network information

PEA:

Pathway enrichment analysis

PPI:

Protein-protein interaction

PS:

Pathway enrichment score

PT:

Pathway topology-based

RR:

Rank ratio

RS:

Risk score

S:

Stability

scRNA-seq:

Single-cell RNA-seq

TCGA:

The Cancer Genome Atlas

TF:

Transcription factor

THCA:

Thyroid cancer

References

  1. Kanehisa M, Furumichi M, Tanabe M, Sato Y, Morishima K. KEGG: new perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Res. 2017;45:D353–61.

    Article  CAS  PubMed  Google Scholar 

  2. Gene Ontology C. Gene Ontology Consortium: going forward. Nucleic Acids Res. 2015;43:D1049–56.

    Article  Google Scholar 

  3. Wang X, Yin TS, Boyi LI, Jiang XL, Sun H, Dou YG, et al. Progress in gene functional enrichment analysis. Sci Sin Vitae. 2016;46:363-73.

  4. Dennis G Jr, Sherman BT, Hosack DA, Yang J, Gao W, Lane HC, et al. DAVID: database for annotation, visualization, and integrated discovery. Genome Biol. 2003;4:P3.

    Article  PubMed  Google Scholar 

  5. Liao Y, Wang J, Jaehnig EJ, Shi Z, Zhang B. WebGestalt 2019: gene set analysis toolkit with revamped UIs and APIs. Nucleic Acids Res. 2019;47:W199–205.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Bindea G, Mlecnik B, Hackl H, Charoentong P, Tosolini M, Kirilovsky A, et al. ClueGO: a Cytoscape plug-in to decipher functionally grouped gene ontology and pathway annotation networks. Bioinformatics. 2009;25:1091–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Zhou Y, Zhou B, Pache L, Chang M, Khodabakhshi AH, Tanaseichuk O, et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat Commun. 2019;10:1523.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Beissbarth T, Speed TP. GOstat: find statistically overrepresented Gene Ontologies within a group of genes. Bioinformatics. 2004;20:1464–5.

    Article  CAS  PubMed  Google Scholar 

  9. Yu G, Wang LG, Han Y, He QY. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS. 2012;16:284–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Hanzelmann S, Castelo R, Guinney J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics. 2013;14:7.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005;102:15545–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Yang Q, Wang S, Dai E, Zhou S, Liu D, Liu H, et al. Pathway enrichment analysis approach based on topological structure and updated annotation of pathway. Brief Bioinform. 2019;20:168–77.

    Article  CAS  PubMed  Google Scholar 

  13. Gu Z, Wang J. CePa: an R package for finding significant pathways weighted by multiple network centralities. Bioinformatics. 2013;29:658–60.

    Article  CAS  PubMed  Google Scholar 

  14. Tarca AL, Draghici S, Khatri P, Hassan SS, Mittal P, Kim JS, et al. A novel signaling pathway impact analysis. Bioinformatics. 2009;25:75–82.

    Article  CAS  PubMed  Google Scholar 

  15. Bayerlova M, Jung K, Kramer F, Klemm F, Bleckmann A, Beissbarth T. Comparative study on gene set and pathway topology-based enrichment methods. BMC Bioinformatics. 2015;16:334.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Jeong WJ, Ro EJ, Choi KY. Interaction between Wnt/beta-catenin and RAS-ERK pathways and an anti-cancer strategy via degradations of beta-catenin and RAS by targeting the Wnt/beta-catenin pathway. NPJ Precis Oncol. 2018;2:5.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Pham L, Christadore L, Schaus S, Kolaczyk ED. Network-based prediction for sources of transcriptional dysregulation using latent pathway identification analysis. Proc Natl Acad Sci U S A. 2011;108:13347–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Dutta B, Wallqvist A, Reifman J. PathNet: a tool for pathway analysis using topological information. Source Code Biol Med. 2012;7:10.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Li Y, Agarwal P, Rajagopalan D. A global pathway crosstalk network. Bioinformatics. 2008;24:1442–7.

    Article  CAS  PubMed  Google Scholar 

  20. Liu ZP, Wang Y, Zhang XS, Chen L. Identifying dysfunctional crosstalk of pathways in various regions of Alzheimer’s disease brains. BMC Syst Biol. 2010;4(Suppl 2):S11.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Kelder T, Eijssen L, Kleemann R, van Erk M, Kooistra T, Evelo C. Exploring pathway interactions in insulin resistant mouse liver. BMC Syst Biol. 2011;5:127.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Fabregat A, Jupe S, Matthews L, Sidiropoulos K, Gillespie M, Garapati P, et al. The reactome pathway knowledgebase. Nucleic Acids Res. 2018;46:D649–55.

    Article  CAS  PubMed  Google Scholar 

  23. Mi H, Thomas P. PANTHER pathway: an ontology-based pathway database coupled with data analysis tools. Methods Mol Biol. 2009;563:123–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Romero P, Wagg J, Green ML, Kaiser D, Krummenacker M, Karp PD. Computational prediction of human metabolic pathways from the complete human genome. Genome Biol. 2005;6:R2.

    Article  PubMed  Google Scholar 

  25. Yamamoto S, Sakai N, Nakamura H, Fukagawa H, Fukuda K, Takagi T. INOH: ontology-based highly structured database of signal transduction pathways. Database (Oxford). 2011;2011:bar052.

    Article  Google Scholar 

  26. Kandasamy K, Mohan SS, Raju R, Keerthikumar S, Kumar GS, Venugopal AK, et al. NetPath: a public resource of curated signal transduction pathways. Genome Biol. 2010;11:R3.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Schaefer CF, Anthony K, Krupa S, Buchoff J, Day M, Hannay T, et al. PID: the pathway interaction database. Nucleic Acids Res. 2009;37:D674–9.

    Article  CAS  PubMed  Google Scholar 

  28. Slenter DN, Kutmon M, Hanspers K, Riutta A, Windsor J, Nunes N, et al. WikiPathways: a multifaceted pathway database bridging metabolomics to other omics research. Nucleic Acids Res. 2018;46:D661–7.

    Article  CAS  PubMed  Google Scholar 

  29. Liu H, Yuan M, Mitra R, Zhou X, Long M, Lei W, et al. CTpathway: a crosstalk-based pathway enrichment analysis method: Github; 2022. https://doi.org/10.5281/zenodo.7089771. https://github.com/Bioccjw/CTpathway/

    Book  Google Scholar 

  30. Rodchenkov I, Babur O, Luna A, Aksoy BA, Wong JV, Fong D, et al. Pathway Commons 2019 Update: integration, analysis and exploration of pathway data. Nucleic Acids Res. 2020;48:D489–97.

    CAS  PubMed  Google Scholar 

  31. Li C, Li X, Miao Y, Wang Q, Jiang W, Xu C, et al. SubpathwayMiner: a software package for flexible identification of pathways. Nucleic Acids Res. 2009;37:e131.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Mohamed A, Hancock T, Nguyen CH, Mamitsuka H. NetPathMiner: R/Bioconductor package for network path mining through gene expression. Bioinformatics. 2014;30:3139–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Matys V, Fricke E, Geffers R, Gossling E, Haubrock M, Hehl R, et al. TRANSFAC: transcriptional regulation, from patterns to profiles. Nucleic Acids Res. 2003;31:374–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Cheng F, Desai RJ, Handy DE, Wang R, Schneeweiss S, Barabasi AL, et al. Network-based approach to prediction and population-based validation of in silico drug repurposing. Nat Commun. 2018;9:2691.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Cheng F, Kovacs IA, Barabasi AL. Network-based prediction of drug combinations. Nat Commun. 2019;10:1197.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Tarca AL, Draghici S, Bhatti G, Romero R. Down-weighting overlapping genes improves gene set analysis. BMC Bioinformatics. 2012;13:136.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Zyla J, Marczyk M, Domaszewska T, Kaufmann SHE, Polanska J, Weiner J. Gene set enrichment for reproducible science: comparison of CERNO and eight other algorithms. Bioinformatics. 2019;35:5146–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Barrett T, Wilhite SE, Ledoux P, Evangelista C, Kim IF, Tomashevsky M, et al. NCBI GEO: archive for functional genomics data sets--update. Nucleic Acids Res. 2013;41:D991–5.

    Article  CAS  PubMed  Google Scholar 

  39. Tomczak K, Czerwinska P, Wiznerowicz M. The Cancer Genome Atlas (TCGA): an immeasurable source of knowledge. Contemp Oncol (Pozn). 2015;19:A68–77.

    Google Scholar 

  40. Szigeti KA, Kalmar A, Galamb O, Valcz G, Bartak BK, Nagy ZB, et al. Global DNA hypomethylation of colorectal tumours detected in tissue and liquid biopsies may be related to decreased methyl-donor content. BMC Cancer. 2022;22:605.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Lee YS. Gene expression profile of hepatocellular carcinoma patients in Taiwan. 2019.

    Google Scholar 

  42. Moreno Leon L, Gautier M, Allan R, Ilie M, Nottet N, Pons N, et al. The nuclear hypoxia-regulated NLUCAT1 long non-coding RNA contributes to an aggressive phenotype in lung adenocarcinoma through regulation of oxidative stress. Oncogene. 2019;38:7146–65.

    Article  CAS  PubMed  Google Scholar 

  43. Tothill RW, Tinker AV, George J, Brown R, Fox SB, Lade S, et al. Novel molecular subtypes of serous and endometrioid ovarian cancer linked to clinical outcome. Clin Cancer Res. 2008;14:5198–208.

    Article  CAS  PubMed  Google Scholar 

  44. Rotunno M, Hu N, Su H, Wang C, Goldstein AM, Bergen AW, et al. A gene expression signature from peripheral whole blood for stage I lung adenocarcinoma. Cancer Prev Res (Phila). 2011;4:1599–608.

    Article  CAS  Google Scholar 

  45. Karaayvaz M, Cristea S, Gillespie SM, Patel AP, Mylvaganam R, Luo CC, et al. Unravelling subclonal heterogeneity and aggressive disease states in TNBC through single-cell RNA-seq. Nat Commun. 2018;9:3588.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43:e47.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15:550.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Mitra R, Chen X, Greenawalt EJ, Maulik U, Jiang W, Zhao Z, et al. Decoding critical long non-coding RNA in ovarian cancer epithelial-to-mesenchymal transition. Nat Commun. 2017;8:1604.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Stuart T, Butler A, Hoffman P, Hafemeister C, Papalexi E, Mauck WM 3rd, et al. Comprehensive integration of single-cell data. Cell. 2019;177:1888–1902.e1821.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. McCarthy DJ, Smyth GK. Testing significance relative to a fold-change threshold is a TREAT. Bioinformatics. 2009;25:765–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Xiao Y, Hsiao TH, Suresh U, Chen HI, Wu X, Wolf SE, et al. A novel significance score for gene selection and ranking. Bioinformatics. 2014;30:801–7.

    Article  CAS  PubMed  Google Scholar 

  52. Rosvall M, Bergstrom CT. Maps of random walks on complex networks reveal community structure. Proc Natl Acad Sci U S A. 2008;105:1118–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Knijnenburg TA, Wessels LF, Reinders MJ, Shmulevich I. Fewer permutations, more accurate P-values. Bioinformatics. 2009;25:i161–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Benjamini Y, Drai D, Elmer G, Kafkafi N, Golani I. Controlling the false discovery rate in behavior genetics research. Behav Brain Res. 2001;125:279–84.

    Article  CAS  PubMed  Google Scholar 

  55. Enright AJ, Van Dongen S, Ouzounis CA. An efficient algorithm for large-scale detection of protein families. Nucleic Acids Res. 2002;30:1575–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Chen L, Chen DT, Kurtyka C, Rawal B, Fulp WJ, Haura EB, et al. Tripartite motif containing 28 (Trim28) can regulate cell proliferation by bridging HDAC1/E2F interactions. J Biol Chem. 2012;287:40106–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Ito S, Miki Y, Saito R, Inoue C, Okada Y, Sasano H. Amyloid precursor protein and its phosphorylated form in non-small cell lung carcinoma. Pathol Res Pract. 2019;215:152463.

    Article  CAS  PubMed  Google Scholar 

  58. Siegfried JM, Hershberger PA, Stabile LP. Estrogen receptor signaling in lung cancer. Semin Oncol. 2009;36:524–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Cancer Genome Atlas Research N. Comprehensive molecular profiling of lung adenocarcinoma. Nature. 2014;511:543–50.

    Article  Google Scholar 

  60. Sondka Z, Bamford S, Cole CG, Ward SA, Dunham I, Forbes SA. The COSMIC Cancer Gene Census: describing genetic dysfunction across all human cancers. Nat Rev Cancer. 2018;18:696–705.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Karin M, Liu Z, Zandi E. AP-1 function and regulation. Curr Opin Cell Biol. 1997;9:240–6.

    Article  CAS  PubMed  Google Scholar 

  62. Angel P, Karin M. The role of Jun, Fos and the AP-1 complex in cell-proliferation and transformation. Biochim Biophys Acta. 1991;1072:129–57.

    CAS  PubMed  Google Scholar 

  63. Ameyar M, Wisniewska M, Weitzman JB. A role for AP-1 in apoptosis: the case for and against. Biochimie. 2003;85:747–52.

    Article  CAS  PubMed  Google Scholar 

  64. Ibrahim SAE, Abudu A, Johnson E, Aftab N, Conrad S, Fluck M. The role of AP-1 in self-sufficient proliferation and migration of cancer cells and its potential impact on an autocrine/paracrine loop. Oncotarget. 2018;9:34259–78.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Lopez-Bergami P, Lau E, Ronai Z. Emerging roles of ATF2 and the dynamic AP1 network in cancer. Nat Rev Cancer. 2010;10:65–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Szabo E, Riffe ME, Steinberg SM, Birrer MJ, Linnoila RI. Altered cJUN expression: an early event in human lung carcinogenesis. Cancer Res. 1996;56:305–15.

    CAS  PubMed  Google Scholar 

  67. Tessari G, Ferrara C, Poletti A, Dubrovich A, Corsini A, Del Favero G, et al. The expression of proto-oncogene c-jun in human pancreatic cancer. Anticancer Res. 1999;19:863–7.

    CAS  PubMed  Google Scholar 

  68. Gartel AL. FOXM1 in cancer: interactions and vulnerabilities. Cancer Res. 2017;77:3135–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Najafi M, Farhood B, Mortezaee K. Extracellular matrix (ECM) stiffness and degradation as cancer drivers. J Cell Biochem. 2019;120:2782–90.

    Article  CAS  PubMed  Google Scholar 

  70. Kessenbrock K, Plaks V, Werb Z. Matrix metalloproteinases: regulators of the tumor microenvironment. Cell. 2010;141:52–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Tenen DG, Chai L, Tan JL. Metabolic alterations and vulnerabilities in hepatocellular carcinoma. Gastroenterol Rep (Oxf). 2021;9:1–13.

    Article  Google Scholar 

  72. Nordgaard I, Mortensen PB. Digestive processes in the human colon. Nutrition. 1995;11:37–45.

    CAS  PubMed  Google Scholar 

  73. Prutki M, Poljak-Blazi M, Jakopovic M, Tomas D, Stipancic I, Zarkovic N. Altered iron metabolism, transferrin receptor 1 and ferritin in patients with colon cancer. Cancer Lett. 2006;238:188–96.

    Article  CAS  PubMed  Google Scholar 

  74. Moon C, Soria JC, Jang SJ, Lee J, Obaidul Hoque M, Sibony M, et al. Involvement of aquaporins in colorectal carcinogenesis. Oncogene. 2003;22:6699–703.

    Article  CAS  PubMed  Google Scholar 

  75. Ma X, Cui Y, Zhou H, Li Q. Function of mitochondrial pyruvate carriers in hepatocellular carcinoma patients. Oncol Lett. 2018;15:9110–6.

    PubMed  PubMed Central  Google Scholar 

  76. Prentki M, Madiraju SR. Glycerolipid metabolism and signaling in health and disease. Endocr Rev. 2008;29:647–76.

    Article  CAS  PubMed  Google Scholar 

  77. Schug ZT, Vande Voorde J, Gottlieb E. The metabolic fate of acetate in cancer. Nat Rev Cancer. 2016;16:708–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Saab S, Mallam D, Cox GA 2nd, Tong MJ. Impact of coffee on liver diseases: a systematic review. Liver Int. 2014;34:495–504.

    Article  PubMed  Google Scholar 

  79. Ramsey J, Butnor K, Peng Z, Leclair T, van der Velden J, Stein G, et al. Loss of RUNX1 is associated with aggressive lung adenocarcinomas. J Cell Physiol. 2018;233:3487–97.

    Article  CAS  PubMed  Google Scholar 

  80. Westhoff B, Colaluca IN, D'Ario G, Donzelli M, Tosoni D, Volorio S, et al. Alterations of the Notch pathway in lung cancer. Proc Natl Acad Sci U S A. 2009;106:22293–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Coukos G, Tanyi J, Kandalaft LE. Opportunities in immunotherapy of ovarian cancer. Ann Oncol. 2016;27(Suppl 1):i11–5.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Shanmughapriya S, Senthilkumar G, Vinodhini K, Das BC, Vasanthi N, Natarajaseenivasan K. Viral and bacterial aetiologies of epithelial ovarian cancer. Eur J Clin Microbiol Infect Dis. 2012;31:2311–7.

    Article  CAS  PubMed  Google Scholar 

  83. Keikha M, Esfahani BN. The relationship between tuberculosis and lung cancer. Adv Biomed Res. 2018;7:58.

    Article  PubMed  PubMed Central  Google Scholar 

  84. Dy GK. The role of focal adhesion kinase in lung cancer. Anticancer Agents Med Chem. 2013;13:581–3.

    Article  CAS  PubMed  Google Scholar 

  85. Stevens LE, Cheung WKC, Adua SJ, Arnal-Estape A, Zhao M, Liu Z, et al. Extracellular matrix receptor expression in subtypes of lung adenocarcinoma potentiates outgrowth of micrometastases. Cancer Res. 2017;77:1905–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Burotto M, Chiou VL, Lee JM, Kohn EC. The MAPK pathway across different malignancies: a new perspective. Cancer. 2014;120:3446–56.

    Article  CAS  PubMed  Google Scholar 

  87. Bast RC Jr, Hennessy B, Mills GB. The biology of ovarian cancer: new opportunities for translation. Nat Rev Cancer. 2009;9:415–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Arend RC, Londono-Joshi AI, Straughn JM Jr, Buchsbaum DJ. The Wnt/beta-catenin pathway in ovarian cancer: a review. Gynecol Oncol. 2013;131:772–9.

    Article  CAS  PubMed  Google Scholar 

  89. Hall CA, Wang R, Miao J, Oliva E, Shen X, Wheeler T, et al. Hippo pathway effector Yap is an ovarian cancer oncogene. Cancer Res. 2010;70:8517–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Olea-Flores M, Zuniga-Eulogio MD, Mendoza-Catalan MA, Rodriguez-Ruiz HA, Castaneda-Saucedo E, Ortuno-Pineda C, et al. Extracellular-signal regulated kinase: a central molecule driving epithelial-mesenchymal transition in cancer. Int J Mol Sci. 2019;20(12):2885.

    Article  CAS  PubMed Central  Google Scholar 

  91. Shin S, Buel GR, Nagiec MJ, Han MJ, Roux PP, Blenis J, et al. ERK2 regulates epithelial-to-mesenchymal plasticity through DOCK10-dependent Rac1/FoxO1 activation. Proc Natl Acad Sci U S A. 2019;116:2967–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Janiszewska M, Primi MC, Izard T. Cell adhesion in cancer: beyond the migration of single cells. J Biol Chem. 2020;295:2495–505.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Zhao M, Kong L, Liu Y, Qu H. dbEMT: an epithelial-mesenchymal transition associated gene resource. Sci Rep. 2015;5:11459.

    Article  PubMed  PubMed Central  Google Scholar 

  94. Wallace TA, Prueitt RL, Yi M, Howe TM, Gillespie JW, Yfantis HG, et al. Tumor immunobiological differences in prostate cancer between African-American and European-American men. Cancer Res. 2008;68:927–36.

    Article  CAS  PubMed  Google Scholar 

  95. Blalock EM, Geddes JW, Chen KC, Porter NM, Markesbery WR, Landfield PW. Incipient Alzheimer’s disease: microarray correlation analyses reveal major transcriptional and tumor suppressor responses. Proc Natl Acad Sci U S A. 2004;101:2173–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Dal Porto JM, Gauld SB, Merrell KT, Mills D, Pugh-Bernard AE, Cambier J. B cell antigen receptor signaling 101. Mol Immunol. 2004;41:599–613.

    Article  CAS  PubMed  Google Scholar 

  97. Shibuya M. Vascular endothelial growth factor (VEGF) and its receptor (VEGFR) signaling in angiogenesis: a crucial target for anti- and pro-angiogenic therapies. Genes Cancer. 2011;2:1097–105.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Sanjuan MA, Dillon CP, Tait SW, Moshiach S, Dorsey F, Connell S, et al. Toll-like receptor signalling in macrophages links the autophagy pathway to phagocytosis. Nature. 2007;450:1253–7.

    Article  CAS  PubMed  Google Scholar 

  99. Gehmert S, Lehoczky G, Loibl M, Jung F, Prantl L, Gehmert S. Interaction between extracellular cancer matrix and stromal breast cells. Clin Hemorheol Microcirc. 2020;74:45–52.

    Article  CAS  PubMed  Google Scholar 

  100. Pitcher LA, van Oers NS. T-cell receptor signal transmission: who gives an ITAM? Trends Immunol. 2003;24:554–60.

    Article  CAS  PubMed  Google Scholar 

  101. Huang da W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 2009;4:44–57.

    Article  PubMed  Google Scholar 

  102. Reimand J, Isserlin R, Voisin V, Kucera M, Tannus-Lopes C, Rostamianfar A, et al. Pathway enrichment analysis and visualization of omics data using g:Profiler, GSEA, Cytoscape and EnrichmentMap. Nat Protoc. 2019;14:482–517.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Liu A, Trairatphisan P, Gjerga E, Didangelos A, Barratt J, Saez-Rodriguez J. From expression footprints to causal pathways: contextualizing large signaling networks with CARNIVAL. NPJ Syst Biol Appl. 2019;5:40.

    Article  PubMed  PubMed Central  Google Scholar 

  104. Garcia-Alonso L, Holland CH, Ibrahim MM, Turei D, Saez-Rodriguez J. Benchmark and integration of resources for the estimation of human transcription factor activities. Genome Res. 2019;29:1363–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Funding

This work was supported by the National Natural Science Foundation of China [61872183, 62172213] to WJ. This work was also supported by the National Institute of Health/National Cancer Institute grants R01CA177786 (CME), R01CA236853 (CME), and P30CA056036 that supports the Sidney Kimmel Cancer Center at Thomas Jefferson University, and the Herbert A. Rosenthal Endowed Chair fund (CME) and the Steinfort Research Fund.

Author information

Authors and Affiliations

Authors

Contributions

WJ designed the study; HZL, MQY, and RM carried out the data acquisition and analysis and drafted the manuscript; XZ, ML, and WYL performed the statistical analysis; SHZ, YEH, and FH provided scientific advice and contributed to result interpretations; RM and CME provided scientific advice and contributed to result interpretations and the writing of the manuscript. The authors read and approved the final manuscript. HZL, MQY, and RM contributed equally to this work.

Corresponding authors

Correspondence to Christine M. Eischen or Wei Jiang.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

Dr. Eischen discloses research funding from AbbVie unrelated to this manuscript. The remaining authors declare they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1: Table S1.

Information of protein-protein (PPI) included in this study. Table S2. Summary of data sets analyzed in this study. Table S3. Summary of 12 gold standard data sets. Table S4. The degree of genes in the GPCM. Table S5. List of CGC genes of four cancer types. Table S6. GSEA enrichment results of RS or |log2FC| for CGC genes. Tables S7-S14. Significant (FDR < 0.01) pathways identified by CTpathway for GEO COAD, TCGA COAD, GEO LIHC, TCGA LIHC, GEO LUAD, TCGA LUAD, GEO OV, TCGA OV. Tables S15-S18. Comparative results show significant KEGG pathways identified by CTpathway with other methods from GEO LUAD, TCGA LUAD, GEO OV, TCGA OV data. Table S19. Significant (FDR < 0.01) KEGG pathways for GEO OV data only identified by CTpathway. Table S20. EMT genes in the top 100 of RS rank list.

Additional file 2: Figure S1.

Crosstalk effect consideration detects genes known to be associated with LUAD. Figure S2. CGC genes are in top of the RS rank list, but not top of the |log2FC| rank list. Figure S3. The proportion of risk genes, overlooked by other methods. Figure S4. Comparison of DEG proportions between pathways specifically identified by CTpathway and non-specific pathways. Figure S5. Significant pathways in data sets with a small number of DEGs, identified by CTpathway. Figure S6. Comparison of enrichment result (P-value) of target pathways for TCGA cancer stages (I, II, III and IV), obtained by different methods. Figure S7. Enrichment result (RR value) comparison of target pathways for B cell by different methods. Figure S8. An enrichment map constructed from early-stage COAD data set. Figure S9. The publicly available CTpathway web tool.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, H., Yuan, M., Mitra, R. et al. CTpathway: a CrossTalk-based pathway enrichment analysis method for cancer research. Genome Med 14, 118 (2022). https://doi.org/10.1186/s13073-022-01119-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13073-022-01119-6

Keywords