Abstract

Integrative analysis of multi-omic datasets has proven to be extremely valuable in cancer research and precision medicine. However, obtaining multimodal data from the same samples is often difficult. Integrating multiple datasets of different omics remains a challenge, with only a few available algorithms developed to solve it. Here, we present INTEND (IntegratioN of Transcriptomic and EpigeNomic Data), a novel algorithm for integrating gene expression and DNA methylation datasets covering disjoint sets of samples. To enable integration, INTEND learns a predictive model between the two omics by training on multi-omic data measured on the same set of samples. In comprehensive testing on 11 TCGA (The Cancer Genome Atlas) cancer datasets spanning 4329 patients, INTEND achieves significantly superior results compared with four state-of-the-art integration algorithms. We also demonstrate INTEND’s ability to uncover connections between DNA methylation and the regulation of gene expression in the joint analysis of two lung adenocarcinoma single-omic datasets from different sources. INTEND’s data-driven approach makes it a valuable multi-omic data integration tool. The code for INTEND is available at https://github.com/Shamir-Lab/INTEND.

INTRODUCTION

Emerging technological advances in recent years have made high-throughput genome-wide sequencing a central tool for biological research. It allows the collective analysis of various types of biological data (commonly termed ‘omics’), in a single tissue or even at the level of a single cell. These include genomics—covering the DNA sequence itself; transcriptomics—the expression levels of genes in the form of mRNAs; epigenomics—reversible modifications on the genetic data, e.g. DNA methylation and chromatin accessibility; proteomics—the levels of translated proteins; and more. Although the analysis of a single omic may generate meaningful insights, it may be necessary to conduct a multi-omic integrative analysis to comprehensively understand a biological system and its complexities. For brevity, we will use throughout the term ‘integration’ for integrative analysis. Hence, integrating different omic datasets is one of the most interesting challenges in computational biology today, with the potential for opening up new avenues in cancer research and precision medicine (1–3).

Multi-omic integration: diverse problems, diverse approaches

One way to obtain multi-omics data for analysis is to simultaneously measure more than one omic from the same tissue. For example, TCGA (The Cancer Genome Atlas) (4) contains multimodal data for numerous tissues spanning dozens of cancer types. The main data types covered by TCGA are genotype, copy number variations, genome methylation, mRNA expression and microRNA (miRNA) expression, along with clinical data. Multimodal data can also be obtained at the cell level by simultaneously measuring multiple types of molecules within the cell (5–7). Such technologies are relatively new and expensive, and thus so far there are far fewer data of multiple omics from the same cells.

Schematically, we can categorize the integration problems into three scenarios (Figure 1A). (i) Single omic–multiple datasets (SO/MD). Here only one omic type is used but multiple datasets (typically experiments from different labs or studies) need to be analyzed together. (ii) Multiple omic–single dataset (MO/SD). Here there is one set of samples on which several omics were measured, and the feature sets of the different omics are disjoint. (iii) Multiple omics–multiple datasets (MO/MD). This problem generalizes both (i) and (ii).

Figure 1.

(A) Three scenarios of integration problems: green, single omic–multiple datasets (SO/MD); red, multiple omics–single dataset (MO/SD); blue, multiple omics–multiple datasets (MO/MD). (B) An overview of the two phases of INTEND: the training phase and the embedding phase.

Many algorithms were developed to handle the integration in the MO/SD setting. These include DIABLO (8), iCluster (9) and MOFA/MOFA+ (10,11), which use the latent variable analysis approach; iNMF (12), which uses non-negative matrix factorization; similarity-based methods such as SNF (13), NEMO (14,15) and MONET (16); and scAI (17), which specializes in single-cell data. Other algorithms were developed to tackle the integration in the SO/MD setting. These algorithms should balance the trade-off between the removal of batch effects and the conservation of biological variance (18). Relevant examples are MNN (19), Seurat v3 (20), scVI (21), Scanorama (22), LIGER (23), Conos (24) and Harmony (25).

The challenge we address herein is the composition of the two problems discussed above: MO/MD integration. Only a few algorithms have been developed to tackle this challenge. Both LIGER and Seurat v3 were used to integrate different omic datasets of disjoint sets of cells, specifically transcriptome and epigenome datasets. LIGER was shown to integrate single-cell RNA sequencing (scRNA-seq) with genome-wide DNA methylation, and Seurat to integrate scRNA-seq with scATAC-seq (measuring chromatin accessibility).

The motivation behind integrating datasets across different experiments arises from the difficulties in obtaining multimodal data from the same samples. These difficulties may be technical inabilities, as mentioned in the context of single-cell data, and economic, a significant factor also in the case of bulk sequencing data. An algorithm that can integrate two different omic datasets measured from disjoint sets of samples could assist researchers in utilizing data that have already been collected in the past, allowing a multi-omic systemic view on the investigated subject. This could increase efficiency, both in time and in cost. Consider the situation where the methylation patterns inside tumors of a specific cancer subtype are being investigated. The multi-omics approach could suggest further inquiry of the epigenome–transcriptome connections, i.e. obtaining mRNA sequencing from every tumor and conducting an integrative analysis of the methylation and gene expression patterns together. As RNA-seq data are widely available for many cancer subtypes, it may be the case that such RNA-seq data are already available for other samples of that cancer subtype. With an algorithm that can integrate RNA-seq and DNA methylation datasets measured on disjoint samples, the researcher could conduct an integrative multi-omic analysis while measuring only the methylation patterns, thus requiring fewer resources.

The algorithms for MO/MD integration can be classified according to the correspondence information they require as input. Some methods require partial correspondence between the samples (either tissues or cells). One example is the semi-supervised correspondence approach of the MAGAN algorithm (26). This approach uses matching pairs of samples from both datasets to learn the correct alignment of the datasets. Other methods, such as LIGER and Seurat, require correspondence information between the features of the different omics. Finally, some methods do not require any correspondence information and assume a common underlying structure that is maintained across technologies and omics. Such methods usually belong to the class of machine learning algorithms that solve the unsupervised manifold alignment problem. One algorithm that uses such techniques to integrate single-cell multi-omics data is the maximum mean discrepancy-manifold alignment (MMD-MA) algorithm (27). Another algorithm that can jointly embed two datasets, without any correspondence information between their features or samples, is the joint Laplacian manifold alignment algorithm (JLMA) (28). Using a method that does not require any correspondence information may sound appealing, but may not perform adequately when the assumed common underlying structure is weak.

In our study, we developed a method for the integration of transcriptomic and epigenomic data across different experiments. We focused on the integration of gene expression and DNA methylation. Specializing in two particular omics creates a less general method, but allows us to develop a stronger model: we can incorporate the known biological connections between gene expression and DNA methylation.

Associations between DNA methylation and gene expression

The regulation of gene expression allows cells to increase or decrease the production of proteins or RNA. Such adjustments enable a response to external changes in the environment and to internal signals within cells. In complex multicellular organisms, the regulation of genes in particular cellular contexts enables the differentiation and proliferation of cells. Epigenetic modifications mainly include DNA methylation and histone protein modifications, which alter the chromatin structure. These modifications are known to be key factors in the regulation of gene expression. In the last two decades, a strong connection has been established between epigenetic modifications and the development of cancer. Hence, the integration of transcriptomic and epigenomic data has the potential to broaden our understanding of the molecular mechanisms orchestrating the regulation of genes, in both normal and malignant tissues.

DNA methylation in mammals occurs almost exclusively in the 5′ position of a cytosine followed by a guanine, commonly termed a CpG site. CpG dinucleotides tend to cluster in CpG islands (CGIs), regions with a high frequency of CpG sites. The majority of CpG dinucleotides (75%) throughout the mammalian genomes are methylated (29), except for CGIs, which are mostly unmethylated. About 70% of the proximal promoters of human genes contain a CGI and, reciprocally, ∼50% of the CGIs are located near a gene's transcription start site (TSS). In fact, CGIs are strongly linked to the regulation of transcription (30). Although CGIs are mostly hypomethylated, there are known examples of their methylation, resulting in stable silencing of the associated promoter. However, it is believed that CGI methylation does not initiate the silencing of genes, but assists in making the silenced state permanent (30). For example, in X chromosome inactivation, the methylation process of CGIs in the X chromosome has been shown to start only after gene silencing. However, when DNA methylation is inhibited, genes in the X chromosome can be reactivated.

The connection between CGI hypermethylation and silencing of genes is not the only relationship observed between methylation and gene expression. There is evidence of both strong positive and strong negative correlations between gene body methylation and gene expression (31). Other studies have shown that hypermethylation of CGIs in cancer tissues is not always accompanied by a decrease in gene expression (32). These findings suggest that DNA methylation can play diverse roles in gene regulation, depending on the genomic context (33). This should be considered when using multi-omic integration algorithms such as LIGER and Seurat, which require correspondence information between the features of the different omics. The methods that are currently used to link the feature spaces of DNA methylation and gene expression assume a simplistic connection between the two (see LIGER description in the Supplementary data). The complex and not fully understood relationship between DNA methylation and gene expression stresses the necessity for a more sophisticated approach.

Our approach

Herein, we present a novel algorithm for the MO/MD problem. The algorithm is called INTEND (IntegratioN of Transcriptomic and EpigeNomic Data). Specifically, INTEND aims to integrate gene expression (GE) and DNA methylation (DM) datasets covering disjoint sets of samples. INTEND does not use any correspondence information between the samples in the two datasets (e.g. knowing which GE and DM profiles originated from the same individual). To handle the complex connections between DM and GE, INTEND learns a predictive model between the two, by training on multi-omic data measured on the same set of samples. To the best of our knowledge, this is the first use of a predictive model in the context of the studied problem.

As a preliminary step, for each gene, INTEND learns a function that predicts its expression based on the methylation levels in sites located proximal to it. To integrate the target methylation and GE datasets, INTEND first predicts for each methylation profile its expression profile. Then, it identifies a set of genes that will be used for the joint embedding of the expression and predicted expression datasets. At this stage, both datasets share the same feature space. INTEND then employs canonical-correlation analysis (CCA) to jointly reduce their dimension.

We evaluated the performance of INTEND by comparing it with four state-of-the-art MO/MD integration methods: LIGER, Seurat v3, JLMA and MMD-MA. The first two require correspondence information between the different omic features, in order to create a common feature space before the integration, whereas the last two do not require such information. We used 11 TCGA cancer datasets spanning 4329 patients for testing the algorithms in multiple integration tasks. We also showed the utility of the method in identifying SKCM (skin cutaneous melanoma) cancer subtypes and in joint analysis of LUAD (lung adenocarcinoma) using two single-omic datasets obtained from different individuals.

MATERIALS AND METHODS

INTEND algorithm

INTEND works in two phases (Figure 1B). The training phase receives as input training data consisting of GE and DM profiles measured on the same set of samples. The algorithm uses these data to learn the connections between the omics. This will allow it later to make accurate predictions of expression levels of specified genes based on a given methylation profile. The training process can be executed once for any number of future integration tasks. Intuitively, the multimodal data used in the training process should be ‘biologically similar’ to the datasets that INTEND will integrate subsequently. However, as we shall show, even when we used INTEND to integrate datasets covering tumor types that were different from those covered by the multimodal training data, it performed well.

For the embedding phase, INTEND’s inputs are from two disjoint cohorts, denoted T1 and T2. They include a DM matrix for T1 and a GE matrix for T2. It proceeds in three steps: (i) creation of a predicted GE matrix for T1 based on the DM data; (ii) selection of a subset of the genes based on the predicted GE for T1, the GE for T2 and the trained model from the preliminary step; and (iii) reducing jointly the dimension of the two GE datasets on the selected gene set.

The training phase

The preliminary training phase aims to learn connections between GE and DM using training data. Its inputs are expression and methylation profiles for the same set of samples. Its goal is to determine, for each gene, a function that predicts its expression level based on methylation levels of potentially relevant sites. We hypothesized that accurately predicting the expression levels of even a small number of genes, from an input methylation matrix, will enable successful integration. To achieve this goal, we developed a prediction model considering the known connections between methylation in promoter CGIs and gene expression (30), as well as gene body methylation (31). Furthermore, to capture the variation in the correlation between methylation and expression across the CGI, its shores and shelves, and also outside CGIs (32), the model uses the methylation levels in each probe separately.

For each gene, we considered all the probed methylation sites contained in the gene region or within 10 kb from its ends. While in certain cases more distal methylation sites were reported to affect gene expression (34), the main effect is usually due to proximal sites (30). We limited the range in order to have modest size gene models. As we will show, such models provide a good basis for the integration task.

INTEND uses the Lasso regression model (35,36) to learn the prediction function and select model features. Lasso was run using the glmnet R package, and the optimal value of the penalty constant was chosen using 10-fold cross-validation on the training set. Using Lasso allows the preliminary step to handle genes with a large number of methylation sites, by ignoring sites that have little relevance for the gene expression prediction. For example, in a TCGA training set that we used, spanning 10 cancer subtypes listed in Table 1 (excluding LUAD) and spanning 3852 tumor samples, for each gene 25 methylation sites were considered on average, with a maximum of 1055 sites per gene (Supplementary Figure S1). However, the maximal number of probes for which the regression resulted with non-zero coefficients for a specifed gene was only 424, with an average of 21 sites per gene (Supplementary Figure S2).

Table 1.

Summary information of TCGA cancer datasets used

Number of patient samples
Cancer typeAbbreviationGene expressionDNA methylationBoth
Acute myeloid leukemiaAML173194170
Bladder urothelial carcinomaBLCA427440425
Colon adenocarcinomaCOAD328353298
Brain lower-grade gliomaLGG534534530
Liver hepatocellular carcinomaLIHC424430414
Lung adenocarcinomaLUAD576507477
Pancreatic adenocarcinomaPAAD183195183
Prostate adenocarcinomaPRAD550553533
SarcomaSARC265269263
Skin cutaneous melanomaSKCM473475473
Thyroid carcinomaTHCA572571563
Number of patient samples
Cancer typeAbbreviationGene expressionDNA methylationBoth
Acute myeloid leukemiaAML173194170
Bladder urothelial carcinomaBLCA427440425
Colon adenocarcinomaCOAD328353298
Brain lower-grade gliomaLGG534534530
Liver hepatocellular carcinomaLIHC424430414
Lung adenocarcinomaLUAD576507477
Pancreatic adenocarcinomaPAAD183195183
Prostate adenocarcinomaPRAD550553533
SarcomaSARC265269263
Skin cutaneous melanomaSKCM473475473
Thyroid carcinomaTHCA572571563
Table 1.

Summary information of TCGA cancer datasets used

Number of patient samples
Cancer typeAbbreviationGene expressionDNA methylationBoth
Acute myeloid leukemiaAML173194170
Bladder urothelial carcinomaBLCA427440425
Colon adenocarcinomaCOAD328353298
Brain lower-grade gliomaLGG534534530
Liver hepatocellular carcinomaLIHC424430414
Lung adenocarcinomaLUAD576507477
Pancreatic adenocarcinomaPAAD183195183
Prostate adenocarcinomaPRAD550553533
SarcomaSARC265269263
Skin cutaneous melanomaSKCM473475473
Thyroid carcinomaTHCA572571563
Number of patient samples
Cancer typeAbbreviationGene expressionDNA methylationBoth
Acute myeloid leukemiaAML173194170
Bladder urothelial carcinomaBLCA427440425
Colon adenocarcinomaCOAD328353298
Brain lower-grade gliomaLGG534534530
Liver hepatocellular carcinomaLIHC424430414
Lung adenocarcinomaLUAD576507477
Pancreatic adenocarcinomaPAAD183195183
Prostate adenocarcinomaPRAD550553533
SarcomaSARC265269263
Skin cutaneous melanomaSKCM473475473
Thyroid carcinomaTHCA572571563

After calculating the prediction function for every gene, the 2000 genes with the highest R2 between predicted and observed gene expression are identified for use in the next stages of INTEND. For example, using the above training set, the average R2 of all 19 143 genes considered was 0.30, and the average R2 of the top 2000 genes was 0.68 (Supplementary Figure S3).

Note that when applying the preliminary step to certain cancer subtypes, the subsequent algorithmic steps use only data from other subtypes, in order to avoid overfitting.

The embedding phase

The inputs for the main phase of the algorithm are DM profiles for one target set of samples, and GE profiles for a second, disjoint target set of samples.

First, we use the learned prediction functions from the preliminary step to compute the predicted expression profiles based on the DM input profiles. For each DM profile, we predict its gene expression levels for each gene in the selected set of 2000 genes from the preliminary step. The expression of these genes has the highest likelihood to be predicted accurately by the methylation profile, at least in the tissue types and states included in the training set. However, the target datasets may originate from a different tissue type or state. Hence, an additional heuristic for feature selection is employed.

Genes may be regulated by mechanisms other than DNA methylation. Thus we assumed that the genes that are most likely to be regulated by the methylation profile are those with high variance in both methylation and expression levels. We consider the 2000 genes with the highest expression variability in the set of input GE profiles to be the set of genes with high variability in expression levels. We consider the 2000 genes with the highest variance in the predicted expression profiles to be the set of genes with high variance in methylation levels. From the selected set of 2000 genes from the preliminary step, we select only the genes that are contained in both sets of genes with high variability in expression and methylation levels. The size of the selected gene set varies depending on the training and target datasets. For the next step, we consider only these genes for the input expression profiles matrix and predicted expression profiles matrix. Each feature (gene) of these matrices is centered and scaled separately so that it has zero mean expression level and unit variance.

The last step applies CCA to the resulting matrices, and produces the integrated output matrix. CCA is a dimension reduction method that finds linear combinations of features across datasets such that these combinations have maximum correlation (37). It was used in computational genomics to project datasets that share the same samples but have different features (the MO/SD setting) from a common low-dimensional feature space. CCA has been used in this way, for example, in multi-omic clustering (15,38). In contrast, here we apply CCA to matrices that cover samples from different datasets but share the same set of selected genes (similar to the SO/MD setting). This approach for utilizing CCA was introduced in Seurat v2 (39).

A complete mathematical description of the training and embedding phases can be found in the Supplementary data. The code for INTEND is available at https://github.com/Shamir-Lab/INTEND.

Data

TCGA data

To assess performance, we used RNA-seq and DM data from TCGA (4) covering 11 different cancer types. See Table 1 for cancer types, their abbreviations and statistics. The data were downloaded using the TCGA-Assembler software (40,41). We used only 4329 samples for which both omics were measured.

The DM data we used were gathered with Illumina's Infinium HumanMethylation450 BeadChip assay. The levels of > 450 000 methylation sites were reported as β-values. The RNA-seq data were gathered with Illumina HiSeq assay, and quantified using RSEM (42). In each GE and DM sample, the zero counts were removed, then the raw count values were divided by the 75th percentile of the counts, and then multiplied by 1000. In both omics, we downloaded the data after these transformations from the TCGA website.

An additional LUAD gene expression dataset

In addition to the TCGA LUAD data, we used RNA-seq profiles from 172 tumors of LUAD patients from Singapore (43). GE was quantified with RSEM and normalized as done for the TCGA data.

Data pre-processing

To handle missing values, for each dataset, features with >5% missing values were removed, and then samples with >5% missing values were removed. Subsequently, the missing values per each feature were imputed to the mean of this feature across all samples. The number of features and samples in each dataset we used, before and after the handling of missing values, are described in Supplementary Table S1. Finally, for GE data from all sources and for all purposes, we added one pseudo-count to each value and log-transformed the result.

Running other algorithms

We evaluated the performance of INTEND by comparing it with four state-of-the-art MO/MD integration methods: LIGER, Seurat v3, JLMA and MMD-MA. The methods are briefly described in the Supplementary data. To use LIGER and Seurat, we supplied the algorithms with an aggregated gene-level methylation matrix as input, as they require correspondence information between features across omics. The aggregated matrix computation process is described in Supplementary Figure S4. JLMA and MMD-MA algorithms do not require correspondence information between the features. However, empirical results from (27) showed that JLMA failed to integrate GE and DM using the local geometry metric as a measure for cross-omic similarity. Hence, we computed the cross-omic similarity matrix for JLMA based on the aggregated gene-level methylation matrix. For MMD-MA, we used both the original methylation data and gene-level methylation matrix as inputs. We denoted the runs of JLMA and MMD-MA with the gene-level methylation matrix as JLMA WFCI (with features correspondence information) and MMD-MA WFCI. We ran all the algorithms with their default recommended hyper-parameters and, whenever applicable, we used the algorithm's pipeline for feature selection and normalization. Since MMD-MA and JLMA do not include a method for feature selection, when running them in the WFCI mode, we selected the n genes with the highest variance in expression, for n = 500 and 2000. Further details regarding how each of the algorithms was applied, including hyper-parameters and additional necessary pre-processing steps, are described in the Supplementary data.

Evaluating the quality of the results

For the TCGA data, we have the true pairing of samples that represent different omic measurements of the same patient. This pairing is not given as input to the integration algorithms and can therefore be used to evaluate their results. We use the metric defined in Liu et al. (27) to evaluate the algorithms. For GE and DM input datasets covering nE and nM samples, respectively, each algorithm produces a d-long vector of the projected expression ei for each sample i and a d-long vector of the projected estimated expression mj based on the methylation for each sample j. For patient i, let fi be the fraction of samples j with projections mj closer to ei than mi. We call it the ‘fraction of samples closer than the true match’ (FOSCTTM). FOSCTTM ranges from 0 to 1, where 0 means that the true match of a sample i is the closest to i in the projected space. We calculate the FOSCTTM for every sample in the GE and DM datasets, and average these values. A perfect integration will have a score of 0. For a random projection, the expected FOSCTTM is 0.5.

Clustering

For clustering , we used the k-means algorithm of Hartigan and Wong (44), with a maximum number of 100 iterations and 100 different starting solutions. We selected the desired number of clusters using the ‘elbow method’ as described in Rappoport and Shamir (14). Let v(i) be the total within-cluster sum of squares for a solution with i clusters, then we chose i for which the point v(i) had the maximum curvature. Specifically, we chose the i that maximized the following approximation of the second derivative of v:

$$\begin{equation*}v\left[ {i + 1} \right] + v\left[ {i - 1} \right] - 2v\left[ i \right]\end{equation*}$$
(1)

RESULTS

We applied INTEND in several settings. In the first part, we applied INTEND and four other algorithms in several integration tasks of GE and DM data, using 11 cancer datasets from TCGA. We also demonstrated the utility of the method in identifying SKCM cancer subtypes. In the second part, we used INTEND for the integration of datasets from two different sources, covering two populations of LUAD patients.

Our first set of analyses compared five algorithms: INTEND, LIGER, Seurat v3 (hereafter: Seurat), MMD-MA and JLMA. We used 11 datasets of different cancer types from TCGA. First, we integrated GE and DM data of the same cancer type, for each of the 11 types. Next, we integrated data of four cancer types simultaneously.

Single cancer dataset integration task

We first ran the algorithms with input datasets of a single cancer subtype. We used the 11 datasets listed in Table 1. For each dataset, we considered only the subset of samples measured in both omics. The total number of samples used in these integration tasks was 4329, where dataset sizes ranged from 170 to 563. For each cancer dataset, we trained a new regression model in INTEND’s preliminary phase, using the samples of the remaining 10 cancer datasets as the training set. To evaluate the results, we used the pairing information between samples from the two omics measured on the same tissue to calculate the FOSCTTM score.

We ran the algorithms using projected space dimension d ranging from 2 to 40, and recorded the best integration scores (average FOSCTTM). The results are summarized in Table 2 and Supplementary Figure S5. INTEND performed best across all datasets and all d values, and substantially better than the rest, with MMD-MA the second best performer. In fact, INTEND results were often 1–2 orders of magnitude better than those of all the other methods.

Table 2.

Average FOSCTTM of algorithms for integrating GE and DM data

Cancer/AlgINTENDLIGERSeurat v3MMD-MAMMD-MA WFCI (500)MMD-MA WFCI (2000)JLMA WFCI (500)JLMA WFCI (2000)
AML2.42 (25)29.83 (7)17.05 (36)23.63 (40)19.08 (40)22.35 (40)24.01 (8)28.38 (7)
BLCA0.04 (39)39.62 (9)13.86 (40)11.20 (40)16.34 (40)14.58 (40)34.80 (40)37.11 (40)
COAD0.02 (37)26.84 (19)19.14 (40)12.59 (40)12.19 (40)12.92 (40)32.98 (5)34.73 (4)
LGG6.82 (22)41.97 (8)32.06 (26)8.88 (40)15.50 (40)12.08 (40)37.41 (14)32.38 (12)
LIHC0.14 (36)42.34 (3)19.23 (38)16.04 (30)11.02 (30)12.94 (30)32.68 (21)36.03 (12)
LUAD0.06 (32)36.72 (4)16.36 (39)8.71 (40)14.11 (40)13.89 (40)29.60 (9)32.16 (8)
PAAD0.55 (30)36.68 (15)24.18 (35)11.07 (40)23.42 (40)16.27 (40)29.83 (3)27.44 (2)
PRAD0.37 (38)35.96 (8)16.32 (17)10.88 (40)11.15 (40)10.99 (40)27.14 (2)29.53 (2)
SARC0.05 (35)42.06 (15)12.86 (36)8.86 (40)20.97 (40)17.42 (40)34.47 (7)34.73 (5)
SKCM0.03 (39)42.20 (17)18.97 (37)16.02 (40)20.53 (40)16.62 (40)32.11 (15)34.71 (3)
THCA3.07 (11)32.58 (7)15.96 (36)6.71 (40)7.78 (40)6.65 (40)30.95 (2)27.52 (5)
Average (all datasets)1.23 (31)36.98 (10)18.73 (34)12.24 (39)15.64 (39)14.25 (39)31.45 (11)32.25 (9)
Cancer/AlgINTENDLIGERSeurat v3MMD-MAMMD-MA WFCI (500)MMD-MA WFCI (2000)JLMA WFCI (500)JLMA WFCI (2000)
AML2.42 (25)29.83 (7)17.05 (36)23.63 (40)19.08 (40)22.35 (40)24.01 (8)28.38 (7)
BLCA0.04 (39)39.62 (9)13.86 (40)11.20 (40)16.34 (40)14.58 (40)34.80 (40)37.11 (40)
COAD0.02 (37)26.84 (19)19.14 (40)12.59 (40)12.19 (40)12.92 (40)32.98 (5)34.73 (4)
LGG6.82 (22)41.97 (8)32.06 (26)8.88 (40)15.50 (40)12.08 (40)37.41 (14)32.38 (12)
LIHC0.14 (36)42.34 (3)19.23 (38)16.04 (30)11.02 (30)12.94 (30)32.68 (21)36.03 (12)
LUAD0.06 (32)36.72 (4)16.36 (39)8.71 (40)14.11 (40)13.89 (40)29.60 (9)32.16 (8)
PAAD0.55 (30)36.68 (15)24.18 (35)11.07 (40)23.42 (40)16.27 (40)29.83 (3)27.44 (2)
PRAD0.37 (38)35.96 (8)16.32 (17)10.88 (40)11.15 (40)10.99 (40)27.14 (2)29.53 (2)
SARC0.05 (35)42.06 (15)12.86 (36)8.86 (40)20.97 (40)17.42 (40)34.47 (7)34.73 (5)
SKCM0.03 (39)42.20 (17)18.97 (37)16.02 (40)20.53 (40)16.62 (40)32.11 (15)34.71 (3)
THCA3.07 (11)32.58 (7)15.96 (36)6.71 (40)7.78 (40)6.65 (40)30.95 (2)27.52 (5)
Average (all datasets)1.23 (31)36.98 (10)18.73 (34)12.24 (39)15.64 (39)14.25 (39)31.45 (11)32.25 (9)

Average FOSCTTM score (percentge) for each algorithm on each of the 11 cancer datasets. The optimal score is 0%, and the expected score for a random projection is 50%. The requested shared space dimension d ranges from 2 to 40 for each algorithm. The score shown is the best across all values of d, and the optimal d is given in parentheses. The numbers 500 and 2000 for MMD-MA and JLMA denote the number of selected genes in the WFCI runs.

Table 2.

Average FOSCTTM of algorithms for integrating GE and DM data

Cancer/AlgINTENDLIGERSeurat v3MMD-MAMMD-MA WFCI (500)MMD-MA WFCI (2000)JLMA WFCI (500)JLMA WFCI (2000)
AML2.42 (25)29.83 (7)17.05 (36)23.63 (40)19.08 (40)22.35 (40)24.01 (8)28.38 (7)
BLCA0.04 (39)39.62 (9)13.86 (40)11.20 (40)16.34 (40)14.58 (40)34.80 (40)37.11 (40)
COAD0.02 (37)26.84 (19)19.14 (40)12.59 (40)12.19 (40)12.92 (40)32.98 (5)34.73 (4)
LGG6.82 (22)41.97 (8)32.06 (26)8.88 (40)15.50 (40)12.08 (40)37.41 (14)32.38 (12)
LIHC0.14 (36)42.34 (3)19.23 (38)16.04 (30)11.02 (30)12.94 (30)32.68 (21)36.03 (12)
LUAD0.06 (32)36.72 (4)16.36 (39)8.71 (40)14.11 (40)13.89 (40)29.60 (9)32.16 (8)
PAAD0.55 (30)36.68 (15)24.18 (35)11.07 (40)23.42 (40)16.27 (40)29.83 (3)27.44 (2)
PRAD0.37 (38)35.96 (8)16.32 (17)10.88 (40)11.15 (40)10.99 (40)27.14 (2)29.53 (2)
SARC0.05 (35)42.06 (15)12.86 (36)8.86 (40)20.97 (40)17.42 (40)34.47 (7)34.73 (5)
SKCM0.03 (39)42.20 (17)18.97 (37)16.02 (40)20.53 (40)16.62 (40)32.11 (15)34.71 (3)
THCA3.07 (11)32.58 (7)15.96 (36)6.71 (40)7.78 (40)6.65 (40)30.95 (2)27.52 (5)
Average (all datasets)1.23 (31)36.98 (10)18.73 (34)12.24 (39)15.64 (39)14.25 (39)31.45 (11)32.25 (9)
Cancer/AlgINTENDLIGERSeurat v3MMD-MAMMD-MA WFCI (500)MMD-MA WFCI (2000)JLMA WFCI (500)JLMA WFCI (2000)
AML2.42 (25)29.83 (7)17.05 (36)23.63 (40)19.08 (40)22.35 (40)24.01 (8)28.38 (7)
BLCA0.04 (39)39.62 (9)13.86 (40)11.20 (40)16.34 (40)14.58 (40)34.80 (40)37.11 (40)
COAD0.02 (37)26.84 (19)19.14 (40)12.59 (40)12.19 (40)12.92 (40)32.98 (5)34.73 (4)
LGG6.82 (22)41.97 (8)32.06 (26)8.88 (40)15.50 (40)12.08 (40)37.41 (14)32.38 (12)
LIHC0.14 (36)42.34 (3)19.23 (38)16.04 (30)11.02 (30)12.94 (30)32.68 (21)36.03 (12)
LUAD0.06 (32)36.72 (4)16.36 (39)8.71 (40)14.11 (40)13.89 (40)29.60 (9)32.16 (8)
PAAD0.55 (30)36.68 (15)24.18 (35)11.07 (40)23.42 (40)16.27 (40)29.83 (3)27.44 (2)
PRAD0.37 (38)35.96 (8)16.32 (17)10.88 (40)11.15 (40)10.99 (40)27.14 (2)29.53 (2)
SARC0.05 (35)42.06 (15)12.86 (36)8.86 (40)20.97 (40)17.42 (40)34.47 (7)34.73 (5)
SKCM0.03 (39)42.20 (17)18.97 (37)16.02 (40)20.53 (40)16.62 (40)32.11 (15)34.71 (3)
THCA3.07 (11)32.58 (7)15.96 (36)6.71 (40)7.78 (40)6.65 (40)30.95 (2)27.52 (5)
Average (all datasets)1.23 (31)36.98 (10)18.73 (34)12.24 (39)15.64 (39)14.25 (39)31.45 (11)32.25 (9)

Average FOSCTTM score (percentge) for each algorithm on each of the 11 cancer datasets. The optimal score is 0%, and the expected score for a random projection is 50%. The requested shared space dimension d ranges from 2 to 40 for each algorithm. The score shown is the best across all values of d, and the optimal d is given in parentheses. The numbers 500 and 2000 for MMD-MA and JLMA denote the number of selected genes in the WFCI runs.

We also analyzed the contribution of the last step in INTEND, i.e. applying CCA for dimension reduction, to its performance. We measured the average FOSCTTM when using the original GE data and the imputed GE computed by INTEND, for the selected gene set (see the Materials and Methods). Excluding the CCA step resulted in poorer FOSCTTM scores. Notably, these scores were better than all other tested algorithms in all datasets, with only one exception (Supplementary Table S2).

In later analyses, we preferred to use the same space dimension d for all algorithms. MMD-MA and JLMA do not recommend a method for determining d. For Seurat, the authors originally suggested approaches to select d (39) but later noted that the identification of this value remains a challenge (20). After running all methods for |$d \in [ {2,40} ]$| for all datasets, we observed that most algorithms reach a plateau in the FOSCTTM score at around d =40 (Supplementary Figure S5). Hence, in subsequent runs we set d = 40 for all algorithms, with one exception: LIGER failed to run on the AML dataset with d = 40 or d = 39, so in that case we used d = 38.

Next, we analyzed the FOSCTTM per sample across all methods and datasets. Figure 2 shows boxplots of the FOSCTTM per sample for each algorithm and cancer dataset using d = 40. INTEND’s advantage was prominent, with the entire FOSCTTM interquartile range (IQR) at zero for 8 of the 11 datasets tested. In 6 of the 11 datasets, the FOSCTTM was perfect (zero) for >90% of the samples.

Figure 2.

Distribution of FOSCTTM (%) scores in INTEND results on each cancer type.

We analyzed in more detail the results for the colon adenocarcinoma (COAD) dataset. We used UMAP (45) for the 2D projection of the samples from the original omic feature spaces and from the integration shared space. Figure 3 shows the results for INTEND, LIGER, Seurat and MMD-MA algorithms. The results for JLMA WFCI and MMD-MA WFCI versions are presented in Supplementary Figure S6.

Figure 3.

Results of integration of GE and DM samples from the COAD dataset by different algorithms. (A) UMAP plots of the original data. (B) The same plots as in (A). To appreciate concordance between omics, 10 samples were randomly selected, and their matching points in both omics were labeled. (C–F) UMAP plots of the samples after they were projected to a shared space by each algorithm. (G–J) The same plots as in (C–F) with the selected points labeled. In all plots, colors correspond to omics.

Figure 3A and B shows the projections from the original feature spaces. One can appreciate that pairwise distances are not preserved between the omics. Figure 3CF show for each algorithm the projections from the shared feature space. It is evident that the level of mixing between the two omics is highest for INTEND, intermediate for MMD-MA and lower for Seurat and LIGER. Figure 3G–J show the same projections as in Figure 3CF with the 10 samples of Figure 3B marked. Evidently, INTEND does a much better job in projecting omics from the same sample to close positions. For example, the two points labeled 3 belong to distinct clusters of samples in both the DM and the GE spaces. INTEND was the only method to succeed in projecting the points from both omics into the same cluster in the shared space. A similar advantage of INTEND was obtained for all other cancer types, even when the average FOSCTTM was higher (Supplementary Figures S7–16).

Joint integration of multiple cancer types

In a second test, we applied the algorithms on four cancer datasets simultaneously. We used the datasets of COAD, liver hepatocellular carcinoma (LIHC), sarcoma (SARC) and SKCM, covering 1448 GE and DM profiles. We did not supply the cancer type of each sample to the algorithms. We used the remaining seven TCGA datasets as the training set in INTEND’s training phase. INTEND performed this task with the best FOSCTTM integration score (Supplementary Figure S17), with perfect FOSCTTM for >65% of the samples, and 1–2 orders of magnitude better than the other methods: The mean scores were 0.37% for INTEND, 41.59% for LIGER, 9.33% for Seurat and 4.01% for MMD-MA.

Figure 4 shows 2D projections of the mapping by each of the algorithms. INTEND, Seurat and MMD-MA projected the samples from the different cancer datasets into separate clusters in the shared space (Figure 4GJ). In contrast, LIGER failed to preserve the biological variance among the tissue types, mapping samples of different types to the same clusters (Figure 4I). While INTEND mixed the samples from both omics in each cancer type cluster, Seurat and MMD-MA created clusters with substantial separation between the samples from each omic (Figure 4CF).

Figure 4.

Results of joint integration of GE and DM samples of four cancer datasets: COAD, LIHC, SARC and SKCM. (A and B) UMAP plots of the original data colored by omic (A) and by cancer type (B). (C–J) UMAP plots of the sample projections into the shared space by INTEND, LIGER, Seurat v3 and MMD-MA, colored by omic (C–F) and by cancer type (G–J). (K–N) Confusion matrices for the classification of the DM sample projections into cancer types based on the majority vote among the five nearest GE samples in the shared space.

To further evaluate the results, we tested the quality of classifying the DM samples to specific cancer types based on the types of their neighboring GE samples in the shared space, as follows. Each DM sample was assigned by majority voting to the cancer type most represented among its five closest GE samples in the shared space. The confusion matrices between the inferred and true assignments are shown in Figure 4KN. INTEND performed best, with > 97% of DM samples in each cancer type correctly classified. MMD-MA performed slightly worse: three cancer types had high accuracy classification, but the SARC cancer type had > 9% of the samples misclassified as SKCM. For Seurat, three cancer types had high accuracy classification, but the SKCM cancer type had > 26% of the samples misclassified as SARC. The LIGER projections led to the lowest accuracy classification.

Using INTEND to identify subtypes in skin cutaneous melanoma

Clustering of single-omic cancer data is commonly used to identify subtypes. The quality of the clustering solution can be evaluated by the significance of separation in survival among subtypes. It has been observed that for certain cancer types, one omic may produce much better clustering than another. For example, Rappoport and Shamir (14) benchmarked eight clustering algorithms on the TCGA SKCM data, and observed that GE profile clustering produced clusters with a significant difference in survival in all algorithms, while in DM profile clustering only one algorithm showed such a result. We hypothesized that in such cases, we could use INTEND to obtain GE predictions from the DM data, then jointly embed in the shared space the predictions and a set of GE profiles from the same cancer subtype, and achieve higher significance of separation in survival between clusters of the embedded predictions.

We used a dataset of 473 SKCM samples from TCGA that had both GE and DM profiles. We created 30 random partitions of this set into two equal disjoint groups and, for each partition, we used the first group's DM profiles and the second's GE profiles. We used INTEND to obtain a predicted GE matrix (P) from the DM samples and then embed P jointly with the GE profiles. Call the embedded P data EP. For the training phase of the INTEND model, we used samples from all TCGA datasets listed in Table 1 but excluded the SKCM dataset.

We first clustered separately the original partitioned DM and GE data. We performed each clustering task using k-means (see the Materials and Methods) after selecting the 2000 features with the highest variance and normalizing the features to have zero mean and a standard deviation of one. We ran the algorithm for k between 2 and 15, and selected the desired number of clusters using the ‘elbow method’ (see the Materials and Methods). We measured differential survival between clusters by computing the P-value for the log-rank test. We estimated the P-values using permutation tests (46). As we hypothesized, in most cases, the clustering of the GE data obtained more significant differential survival between clusters than the clustering of the DM data, with the log-rank P-value of the first being lower in 27 of the 30 partitions.

Next, for each of the 30 partitions, we used INTEND’s joint embedding of the DM and GE samples to classify the DM samples based on the k-means clustering of the GE samples. Each DM sample was assigned by majority voting (with ties broken at random) to the cluster most represented among the five GE embeddings closest to its matching EP representation in the shared space. In 23 of the 30 splits, clustering the DM samples using this method obtained more significant differential survival than using the k-means clustering of the DM samples. The average log-rank P-values for the clusterings for all 30 random splits were: 0.07 for the GE k-means clustering, 0.56 for the DM k-means clustering and 0.21 for the integration-based DM clustering, as described above.

We further investigated one of the 23 partitions for which the integration-based DM clustering achieved more significant differential survival than the DM clustering. For that partition, the DM clustering resulted in two clusters with insignificant differential survival (P-value = 0.978, Figure 5A), whereas the GE clustering resulted in two clusters with significant differential survival (P-value = 0.018, Figure 5B). The integration-based DM clustering also obtained significant differential survival between clusters (P-value = 0.036, Figure 5C). See Supplementary Figure S18A–E for the UMAP plots and the clusters.

Figure 5.

Kaplan–Meier plots of clusters of SKCM patients obtained using DM profiles, GE profiles and their INTEND embeddings. (A) Plot for clusters of the original DM profiles. (B) Plot for clusters of the original GE profiles. (C) Plot for clusters of the DM profiles obtained by the integration-based clustering. See Supplementary Figure S18A–E for the UMAP plots and the clusters.

Next, we tested whether the subtypes obtained by the integration-based DM clustering were biologically or clinically more similar to those obtained by the GE k-means clustering. We found that primary tumor and metastases samples were represented in each of the DM k-means clusters exactly in their portion of all DM samples (18.26% of primary tumor samples in both clusters). In contrast, when looking at the GE clusters, the primary tumor samples were over-represented in one cluster and under-represented in the other (28.21% of primary tumor samples in the first cluster, 5.94% in the second, 17.89% in all GE samples). We observed a similar pattern in the integration-based DM clustering: 23.77% of primary tumor samples in one cluster and 11.34% in the other (and 18.26% in all DM samples). This example shows the potential of transferring biological information between GE and DM samples measured on different populations, using INTEND’s integration.

We also compared our results with iCluster, a widely used algorithm for multi-omic subtype identification (9). Since iCluster requires multi-omic measurements from each sample, in order to make a fair comparison, we used the entire multi-omic SKCM TCGA dataset, which comprises GE and DM profiles from 473 samples. We used the same feature selection and normalization as we used for the k-means clustering. To determine the lower dimension of the data in iCluster, we used the dimension with the maximal deviance ratio as defined by the authors. We ran iCluster for dimensions between 1 and 14, corresponding to the number of clusters between 2 and 15. We also ran that same procedure with k-means and INTEND, on the full set of 473 samples. Specifically, we clustered the 473 GE profiles using k-means and then obtained a clustering of the DM profiles based on the GE clustering, by assigning each DM profile to the cluster most represented amongst the five GE embeddings closest to its DM embedding. It is important to note that INTEND did not use the correspondence information between the GE and DM profiles, but only predicted the GE profiles from the DM profiles. Surprisingly, using INTEND’s joint embedding of the DM and GE samples to classify the DM samples based on the k-means clustering of the GE samples, we achieved a significantly better separation of survival between clusters compared with the multi-omic clustering provided by iCluster. The log-rank P-values for the clusterings were 0.39 for the DM k-means clustering, 0.0014 for the GE k-means clustering, 0.0062 for the integration-based DM clustering and 0.14 for the iCluster multi-omic clustering. Therefore, our results suggest that our method outperforms iCluster in multi-omic subtype identification.

Joint analysis of lung adenocarcinoma datasets from different sources

Our next goal was to test the utility of INTEND in joint analysis of two datasets, one of DM profiles and one of GE profiles, coming from different sources. We used data from two studies of LUAD: GE of 172 tumor samples from Chen et al. (43), and DM profiles of 477 samples from TCGA. The datasets were collected in different studies covering disjoint groups of LUAD patients.

Integration

For the training phase of the model, we used samples from all TCGA datasets listed in Table 1 but excluded the LUAD dataset. The integration results are summarized in Figure 6A and B. As the two target datasets here are disjoint, we cannot use FOSCTTM to evaluate their mixing in the embedding phase. As a sanity check, we considered for each sample its closest 32 neighbors (5% of the samples) in the shared space. We expected that if the local neighborhood of a sample is well mixed, the number of samples from each omic in the neighborhood would reflect the relative sizes of the target datasets. For each sample, we measured the ratio between the observed and expected number of samples from the other omic in its neighborhood. If the omics are fully separated, we would expect this ratio to be near zero, whereas for perfectly mixed samples we would expect it to be close to 1. The mean computed ratio for all samples in the shared space was 1.0003 (SD = ± 0.258), and the IQR was 0.82–1.15, indicating well-mixed samples across omics.

Figure 6.

INTEND results on LUAD GE profiles from Chen et al. (43) and DM profiles from TCGA. (A and B) UMAP plots of the original data (A) and of the projections into the shared space (B), colored by omic. (C) Scatterplot of the estimated correlations based on the matching of INTEND projections versus the observed correlations from the multi-omic TCGA dataset, for each of the considered 10.14 million gene–site pairs. The pairs for which the site is within 10 kb from the gene are colored in orange. These gene–site pairs were considered in the INTEND training phase on the TCGA datasets (excluding LUAD). (D) Histogram of the correlation between the estimated and TCGA-observed gene–site correlations, per gene. (E) Correlation coefficients between TK1 expression and methylation levels, at 964 sites located ± 1 Mb from TK1. y-axis: correlations when TK1 expression is based on INTEND projections; x-axis: correlations when both the GE and the paired DM data were taken from TCGA. Correlations with P-value <1e-5 based on both methods are colored in dark blue. (F) Estimated correlation coefficients based on INTEND projections in sites located ± 100 kb from TK1. The x-axis shows their genomic location (build GRCh37/hg19). Correlations with P-value <1e-5 are colored in dark blue; TK1 location is marked by the green arrow. The highlighted yellow regions indicate enhancer regions supported by at least four GeneHancer sources.

Correlations between methylation at specific sites and expression

Next, we wished to test if INTEND application on the two datasets can be used to reveal connections between specific distal DM sites and the regulation of GE in LUAD tumors, even though the GE profiles and DM profiles used here were collected from disjoint sets of patients. For this task, we extracted the estimated correlations between methylation levels at specific CpG sites and the expression levels of specified genes as follows.

We considered for every gene g, the methylation sites located within ± 1 Mb of g (including sites in g). There was a total of ∼10.14 million such gene–site pairs, for which the expression and methylation levels were measured, covering 18 553 different genes. Recall that the INTEND model was trained using proximal sites located only within ± 10 kb from each gene, while here we explore mostly distal methylation sites. To estimate the correlation between the methylation level at site s and the expression level of gene g, we used INTEND projections to get matchings between GE and DM profiles from different patients. First, to match GE and DM profiles, we found the mutual nearest neighbors between the projections of all DM and GE samples in the shared space, using the batchelor R package (19). A pair of a GE profile e and a DM profile m was considered a match if the projection of m was among the k-nearest neighbors of the projection of e, and vice versa (i.e. the projections of e and m are mutual k-nearest neighbors). For k = 5 we obtained 270 matches between GE and DM profiles (out of 172 × 477 = 82 044 possible matches). The matches provided an expression vector of length 270 for each gene g, and a corresponding vector of length 270 for each methylation site s, allowing the examination of the relationship between any gene and methylation site. Next, using the 270 matches, we computed the Pearson‘s correlation coefficient and tested the statistical significance of the association between the expression level and the methylation level of each considered gene–site pair.

We wished to assess the validity of the estimated correlations, based on the created 270 matchings of GE and DM samples from the two LUAD datasets (from here on: ‘estimated correlations’). We compared the estimations with the correlations obtained from 477 pairs of GE and DM profiles measured from the same tissue, from the multi-omic LUAD TCGA dataset. For each of the ∼10 million gene–site pairs previously described, we also computed the correlation between the expression of the gene and the methylation level of the relevant site, based on the multi-omic TCGA dataset (from here on: ‘TCGA-observed correlations’). Figure 6C shows for each gene–site pair the estimated correlation versus the TCGA-observed correlation. Approximately 5.08% of the considered gene–site pairs were detected with significant correlation (P-value <0.01), either positive or negative, according to both methods. For 95.63% of these significant pairs, the estimated correlation coefficient had the same sign as the TCGA-observed correlation. We also tested, for each of the considered genes, the correlation between the estimated correlation and the TCGA-observed correlation, for all sites relevant for that gene. Out of the 18 553 considered genes, there was a significant positive correlation between the estimated and TCGA-observed correlations (P-value <0.05) for 14 693 of the genes. The correlation between the estimated and TCGA-observed correlations was >0.8 for 1041 of the genes, and >0.9 for 180 of them (Figure 6D). This demonstrates the potential of the INTEND integration method to uncover connections between DNA methylation and the regulation of gene expression, for both proximal and distal methylation sites. Repeating the same procedure with the integration results of LIGER, Seurat and MMD-MA for the target LUAD datasets gave inferior results (Supplementary Table S3).

An in-depth look at the regulation of Thymidine Kinase 1

We chose to look in detail at the distal methylation sites of the gene Thymidine Kinase 1 (TK1). High expression of TK1 was recorded in many solid tumors, and was associated specifically with poor prognosis of patients with LUAD (47–49). We computed the correlation between the methylation levels in 964 sites within ± 1 Mb from TK1, and its expression level. The estimated correlations based on the matching of GE and DM profiles from INTEND projections were highly concordant with the correlations computed using the multi-omic TCGA dataset (R2 = 0.824, Figure 6E).

Methylation patterns in enhancer regions are known to be altered in cancer and are closely linked to changes in expression of cancer-related genes (34). Therefore, we checked if strong expression–methylation correlations extracted from INTEND projections can indicate potential distal enhancer regions. We used the GeneHancer database of enhancers and their inferred target genes (50) for information on TK1 enhancers. There were eight enhancer regions supported by at least four GeneHancer sources, seven of them within a 100 kb range from TK1. Figure 6F shows the enhancer regions located ± 100 kb from TK1, and the correlations between methylation and TK1 expression, for sites located in this range. A total of 14 out of the 15 sites in this range with strong negative correlation (P-value <1e-5), are located in one of the documented enhancer regions. Note that all but two of them fall outside the ± 10 kb used for the training phase.

Out of the 964 sites in the 1 Mb range from TK1, we investigated the 10 sites with the strongest negative estimated correlations (full details in Supplementary Table S4). Eight of them are located in two of the enhancer regions shown in Figure 6F (seven of them in a short interval of <500 bases). The other two sites, cg11868461 and cg05110391, are located ∼350 kb downstream and 400 kb upstream of the TSS, respectively. They were not in one of the regions marked by GeneHancer as TK1 enhancers. Nevertheless, both cg11868461 and cg05110391 were identified as ‘enhancer probes’ (not specifically related to TK1) by Mullen et al. (51), using H3K27ac ChIP-seq data from normal and tumor lung tissue samples to identify lung-relevant enhancer regions.

DISCUSSION

We presented the INTEND algorithm for integrating gene expression and DNA methylation from different datasets. We tested it on multiple multi-omic cancer datasets and compared it with extant multi-omic integration algorithms. INTEND showed significantly superior results on all tested datasets when integrating data from single and multiple cancer types, in terms of both FOSCTTM score and classification to cancer types according to the integration results. We demonstrated the potential of INTEND to transfer biological information between GE and DM samples measured on non-overlapping populations of SKCM patients. Clustering DM samples achieved higher significance of separation in survival between clusters when using the integration results of the DM and GE data than using the original DM data only. In another typical use case, we tested INTEND in joint analysis of two lung adenocarcinoma datasets from different sources. Here INTEND demonstrated its potential to uncover connections between DNA methylation and the regulation of gene expression.

INTEND’s novelty mainly resides in the incorporation of the prediction of a GE profile from a DM profile of a sample, into the MO/MD integration problem. Unlike algorithms such as LIGER and Seurat, which were developed mainly to solve the SO/MD problem and then were extended to solve the MO/MD problem, INTEND suggests another method to generate the correspondence information between features—a paramount part of the integration. INTEND presents a data-driven approach to generate a predicted GE matrix, thus effectively reducing the MO/MD problem of integrating GE and DM profiles to the simpler SO/MD problem of integrating multiple GE datasets. Importantly, the data necessary for the training phase of INTEND can represent different populations from the data used for the embedding phase. In all cases presented herein, the used training data originated from samples from cancer types other than those represented in the target datasets for integration. It is important to note that the goal of INTEND is not to predict expression from methylation for individual genes, but rather to enable integrated analysis. In the embedding phase, INTEND uses prediction data for a selected set of genes. Although only a small portion of the genes is selected, the integrated analysis allows the examination of the relationship between any gene and methylation site, as we demonstrated in the lung cancer analysis.

Several questions regarding the performance of INTEND merit additional study. How does the performance of the method depend on the size of the training dataset? How does it depend on the heterogeneity (in our case, the number of different cancer types) in the training set? The analyses above provide anecdotal evidence that both size and heterogeneity make a marked difference (see Supplementary Table S5), but a systematic study is called for. As all our development was done on TCGA, and methylation patterns of tumors may differ from those of normal tissues, how would INTEND work on normal tissues?

INTEND has several limitations. First, the training phase requires multi-omic data measured on the same set of samples, which is not required for the other algorithms we tested. While the training data are not required to be from a similar population to the target data, it is necessary that the omics will be measured in the same method on the training and target datasets. Obtaining multi-omic measurements may be harder in several scenarios, e.g. single-cell multi-omic data. Due to the lack of appropriate single-cell training data, we applied INTEND only on bulk data, which may bias the comparison against single-cell integration methods. Further testing would be needed as such data emerge. Second, the final step in the embedding, applying CCA, may be less effective when the target datasets contain non-overlapping sample populations (e.g. when one of the target datasets contains a group of samples from a cancer type which is not present in the second). Stuart et al. (20) addressed this limitation of using CCA as a final step and introduced a method to overcome it, using the concept of mutual nearest neighbors to identify anchors between the target datasets.

Lastly, we note two possible directions of extending this work. The first is the integration of other pairs of omics, in addition to GE and DM, in a similar method. Here we used an established, simple biological observation, namely the relationship between the state of proximal methylation sites and the gene's expression to build a model and uncover the connections between GE and DM based on available multi-omic data. This concept may be extended to other pairs of omics with available data measuring both on the same set of samples. Another future research direction is the incorporation of methods from algorithms tackling the SO/MD integration problem, after the first step in INTEND’s embedding phase, which results in the predicted GE matrix.

Data Availability

The code for INTEND is available at https://github.com/Shamir-Lab/INTEND or at https://zenodo.org/badge/latestdoi/539148741.

SUPPLEMENTARY DATA

Supplementary Data are available at NAR Online.

ACKNOWLEDGEMENTS

The results published here are in whole or part based upon data generated by the TCGA Research Network: https://www.cancer.gov/tcga. Graphical figures were created using BioRender.com.

FUNDING

Israel Science Foundation (ISF) – Israel Precision Medicine Partnership program [3165 / 19]; Raymond and Beverly Sackler Chair in Bioinformatics, Tel Aviv University; German Research Foundation (DFG) [RE 4193 / 1-1]; ISF [1339 / 18] (to R. S.); Herczeg Institute on Aging Planning and Budgeting Committee (PBC) [Fellowshipfor excellent PhD students in Data Science]; Tel Aviv University Healthy Longevity Research Center; Edmond J. Safra Center for Bioinformatics PhD fellowship (to N. R.); Funding for open access charge: ISF - Israel Precision Medicine Partnership program [3165 / 19], ISF [1339 / 18] and DFG [RE 4193 / 1-1].

Conflict of interest statement. None declared.

REFERENCES

1.

Chakraborty
S.
,
Hosen
M.I.
,
Ahmed
M.
,
Shekhar
H.U.
Onco-multi-OMICS approach: a new frontier in cancer research
.
Biomed Res. Int.
2018
;
2018
:
9836256
.

2.

Efremova
M.
,
Teichmann
S.A.
Computational methods for single-cell omics across modalities
.
Nat. Methods
.
2020
;
17
:
14
17
.

3.

Method of the Year 2019. Single-cell multimodal omics
.
Nat. Methods
.
2020
;
17
:
1
.

4.

Cancer Genome Atlas Research Network
Comprehensive genomic characterization defines human glioblastoma genes and core pathways
.
Nature
.
2008
;
455
:
1061
1068
.

5.

Angermueller
C.
,
Clark
S.J.
,
Lee
H.J.
,
Macaulay
I.C.
,
Teng
M.J.
,
Hu
T.X.
,
Krueger
F.
,
Smallwood
S.A.
,
Ponting
C.P.
,
Voet
T.
et al. .
Parallel single-cell sequencing links transcriptional and epigenetic heterogeneity
.
Nat. Methods
.
2016
;
13
:
229
232
.

6.

Clark
S.J.
,
Argelaguet
R.
,
Kapourani
C.A.
,
Stubbs
T.M.
,
Lee
H.J.
,
Alda-Catalinas
C.
,
Krueger
F.
,
Sanguinetti
G.
,
Kelsey
G.
,
Marioni
J.C.
et al. .
ScNMT-seq enables joint profiling of chromatin accessibility DNA methylation and transcription in single cells
.
Nat. Commun.
2018
;
9
:
781
.

7.

Argelaguet
R.
,
Clark
S.J.
,
Mohammed
H.
,
Stapel
L.C.
,
Krueger
C.
,
Kapourani
C.A.
,
Imaz-Rosshandler
I.
,
Lohoff
T.
,
Xiang
Y.
,
Hanna
C.W.
et al. .
Multi-omics profiling of mouse gastrulation at single-cell resolution
.
Nature
.
2019
;
576
:
487
491
.

8.

Singh
A.
,
Shannon
C.P.
,
Gautier
B.
,
Rohart
F.
,
Vacher
M.
,
Tebbutt
S.J.
,
Cao
K.A.L.
DIABLO: an integrative approach for identifying key molecular drivers from multi-omics assays
.
Bioinformatics
.
2019
;
35
:
3055
3062
.

9.

Shen
R.
,
Olshen
A.B.
,
Ladanyi
M.
Integrative clustering of multiple genomic data types using a joint latent variable model with application to breast and lung cancer subtype analysis
.
Bioinformatics
.
2009
;
25
:
2906
2912
.

10.

Argelaguet
R.
,
Velten
B.
,
Arnol
D.
,
Dietrich
S.
,
Zenz
T.
,
Marioni
J.C.
,
Buettner
F.
,
Huber
W.
,
Stegle
O.
Multi-omics factor analysis—a framework for unsupervised integration of multi-omics data sets
.
Mol. Syst. Biol.
2018
;
14
:
e8124
.

11.

Argelaguet
R.
,
Arnol
D.
,
Bredikhin
D.
,
Deloro
Y.
,
Velten
B.
,
Marioni
J.C.
,
Stegle
O.
MOFA+: a statistical framework for comprehensive integration of multi-modal single-cell data
.
Genome Biol.
2020
;
21
:
111
.

12.

Yang
Z.
,
Michailidis
G.
A non-negative matrix factorization method for detecting modules in heterogeneous omics multi-modal data
.
Bioinformatics
.
2016
;
32
:
1
8
.

13.

Wang
B.
,
Mezlini
A.M.
,
Demir
F.
,
Fiume
M.
,
Tu
Z.
,
Brudno
M.
,
Haibe-Kains
B.
,
Goldenberg
A.
Similarity network fusion for aggregating data types on a genomic scale
.
Nat. Methods
.
2014
;
11
:
333
337
.

14.

Rappoport
N.
,
Shamir
R.
NEMO: cancer subtyping by integration of partial multi-omic data
.
Bioinformatics
.
2019
;
35
:
3348
3356
.

15.

Rappoport
N.
,
Shamir
R.
Multi-omic and multi-view clustering algorithms: review and cancer benchmark
.
Nucleic Acids Res.
2018
;
46
:
10546
10562
.

16.

Rappoport
N.
,
Safra
R.
,
Shamir
R.
MONET: multi-omic module discovery by omic selection
.
PLoS Comput. Biol.
2020
;
16
:
e1008182
.

17.

Jin
S.
,
Zhang
L.
,
Nie
Q.
scAI: an unsupervised approach for the integrative analysis of parallel single-cell transcriptomic and epigenomic profiles
.
Genome Biol.
2020
;
21
:
25
.

18.

Luecken
M.D.
,
Büttner
M.
,
Chaichoompu
K.
,
Danese
A.
,
Interlandi
M.
,
Mueller
M.F.
,
Strobl
D.C.
,
Zappia
L.
,
Dugas
M.
,
Colomé-Tatché
M.
et al. .
Benchmarking atlas-level data integration in single-cell genomics
.
Nat. Methods
.
2022
;
19
:
41
50
.

19.

Haghverdi
L.
,
Lun
A.T.L.
,
Morgan
M.D.
,
Marioni
J.C.
Batch effects in single-cell RNA-sequencing data are corrected by matching mutual nearest neighbors
.
Nat. Biotechnol.
2018
;
36
:
421
427
.

20.

Stuart
T.
,
Butler
A.
,
Hoffman
P.
,
Hafemeister
C.
,
Papalexi
E.
,
Mauck
W.M.
,
Hao
Y.
,
Stoeckius
M.
,
Smibert
P.
,
Satija
R.
Comprehensive integration of single-cell data
.
Cell
.
2019
;
177
:
1888
1902
.

21.

Lopez
R.
,
Regier
J.
,
Cole
M.B.
,
Jordan
M.I.
,
Yosef
N.
Deep generative modeling for single-cell transcriptomics
.
Nat. Methods
.
2018
;
15
:
1053
1058
.

22.

Hie
B.
,
Bryson
B.
,
Berger
B.
Efficient integration of heterogeneous single-cell transcriptomes using Scanorama
.
Nat. Biotechnol.
2019
;
37
:
685
691
.

23.

Welch
J.D.
,
Kozareva
V.
,
Ferreira
A.
,
Vanderburg
C.
,
Martin
C.
,
Macosko
E.Z.
Single-cell multi-omic integration compares and contrasts features of brain cell identity
.
Cell
.
2019
;
177
:
1873
1887
.

24.

Barkas
N.
,
Petukhov
V.
,
Nikolaeva
D.
,
Lozinsky
Y.
,
Demharter
S.
,
Khodosevich
K.
,
Kharchenko
P.V.
Joint analysis of heterogeneous single-cell RNA-seq dataset collections
.
Nat. Methods
.
2019
;
16
:
695
698
.

25.

Korsunsky
I.
,
Millard
N.
,
Fan
J.
,
Slowikowski
K.
,
Zhang
F.
,
Wei
K.
,
Baglaenko
Y.
,
Brenner
M.
,
Loh
P.R.
,
Raychaudhuri
S.
Fast, sensitive and accurate integration of single-cell data with Harmony
.
Nat. Methods
.
2019
;
16
:
1289
1296
.

26.

Amodio
M.
,
Krishnaswamy
S.
MAGAN: aligning biological manifolds
.
35th International Conference on Machine Learning, ICML 2018
.
2018
;

27.

Liu
J.
,
Huang
Y.
,
Singh
R.
,
Vert
J.P.
,
Noble
W.S.
Jointly embedding multiple single-cell omics measurements
.
Leibniz Int. Proc. Informatics
.
2019
;
143
:
10
.

28.

Wang
C.
,
Mahadevan
S.
Manifold alignment without correspondence
.
IJCAI International Joint Conference on Artificial Intelligence
.
2009
;
1273
1278
.

29.

Tost
J.
DNA methylation: an introduction to the biology and the disease-associated changes of a promising biomarker
.
Mol. Biotechnol.
2010
;
44
:
71
81
.

30.

Deaton
A.M.
,
Bird
A.
CpG islands and the regulation of transcription
.
Genes Dev.
2011
;
25
:
1010
1022
.

31.

Jjingo
D.
,
Conley
A.B.
,
Yi
S.V.
,
Lunyak
V.V.
,
King Jordan
I.
On the presence and role of human gene-body DNA methylation
.
Oncotarget
.
2012
;
3
:
462
474
.

32.

Moarii
M.
,
Boeva
V.
,
Vert
J.P.
,
Reyal
F.
Changes in correlation between promoter methylation and gene expression in cancer
.
BMC Genomics
.
2015
;
16
:
873
.

33.

Bhasin
J.M.M.
,
Lee
B.H.H.
,
Matkin
L.
,
Taylor
M.G.G.
,
Hu
B.
,
Xu
Y.
,
Magi-Galluzzi
C.
,
Klein
E.A.A.
,
Ting
A.H.H.
Methylome-wide sequencing detects DNA hypermethylation distinguishing indolent from aggressive prostate cancer
.
Cell Rep.
2015
;
13
:
2135
2146
.

34.

Aran
D.
,
Sabato
S.
,
Hellman
A.
DNA methylation of distal regulatory sites characterizes dysregulation of cancer genes
.
Genome Biol.
2013
;
14
:
R21
.

35.

Tibshirani
R.
Regression shrinkage and selection via the lasso
.
J. R. Stat. Soc. Ser. B
.
1996
;
58
:
267
288
.

36.

Friedman
J.
,
Hastie
T.
,
Tibshirani
R.
Regularization paths for generalized linear models via coordinate descent
.
J. Stat. Softw.
2010
;
33
:
1
22
.

37.

Hotelling
H.
Relations between two sets of variates
.
Biometrika
.
1936
;
28
:
321
377
.

38.

Witten
D.M.
,
Tibshirani
R.J.
Extensions of sparse canonical correlation analysis with applications to genomic data
.
Stat. Appl. Genet. Mol. Biol.
2009
;
8
:
28
.

39.

Butler
A.
,
Hoffman
P.
,
Smibert
P.
,
Papalexi
E.
,
Satija
R.
Integrating single-cell transcriptomic data across different conditions, technologies, and species
.
Nat. Biotechnol.
2018
;
36
:
411
420
.

40.

Wei
L.
,
Jin
Z.
,
Yang
S.
,
Xu
Y.
,
Zhu
Y.
,
Ji
Y.
TCGA-assembler 2: software pipeline for retrieval and processing of TCGA/CPTAC data
.
Bioinformatics
.
2018
;
34
:
1615
1617
.

41.

Zhu
Y.
,
Qiu
P.
,
Ji
Y.
TCGA-assembler: open-source software for retrieving and processing TCGA data
.
Nat. Methods
.
2014
;
11
:
599
600
.

42.

Li
B.
,
Dewey
C.N.
RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome
.
BMC Bioinf.
2011
;
12
:
323
.

43.

Chen
J.
,
Yang
H.
,
Teo
A.S.M.
,
Amer
L.B.
,
Sherbaf
F.G.
,
Tan
C.Q.
,
Alvarez
J.J.S.
,
Lu
B.
,
Lim
J.Q.
,
Takano
A.
et al. .
Genomic landscape of lung adenocarcinoma in East Asians
.
Nat. Genet.
2020
;
52
:
177
186
.

44.

Hartigan
J.A.
,
Wong
M.A.
Algorithm AS 136: a k-means clustering algorithm
.
Appl. Stat.
1979
;
28
:
100
108
.

45.

McInnes
L.
,
Healy
J.
,
Melville
J.
UMAP: uniform manifold approximation and projection for dimension reduction
.
JOSS
.
2018
;
3
:
861
.

46.

Rappoport
N.
,
Shamir
R.
Inaccuracy of the log-rank approximation in cancer data analysis
.
Mol. Syst. Biol.
2019
;
15
:
2017
2019
.

47.

Malvi
P.
,
Janostiak
R.
,
Nagarajan
A.
,
Cai
G.
,
Wajapeyee
N.
Loss of thymidine kinase 1 inhibits lung cancer growth and metastatic attributes by reducing GDF15 expression
.
PLoS Genet.
2019
;
15
:
e1008439
.

48.

Jagarlamudi
K.K.
,
Shaw
M.
Thymidine kinase 1 as a tumor biomarker: technical advances offer new potential to an old biomarker
.
Biomark. Med.
2018
;
12
:
1038
1045
.

49.

He
E.
,
Xu
X.H.
,
Guan
H.
,
Chen
Y.
,
Chen
Z.H.
,
Pan
Z.L.
,
Tang
L.L.
,
Hu
G.Z.
,
Li
Y.
,
Zhang
M.
et al. .
Thymidine kinase 1 is a potential marker for prognosis and monitoring the response to treatment of patients with breast, lung, and esophageal cancer and non-Hodgkin's lymphoma
.
Nucleosides Nucleotides Nucleic Acids
.
2010
;
29
:
352
358
.

50.

Fishilevich
S.
,
Nudel
R.
,
Rappaport
N.
,
Hadar
R.
,
Plaschkes
I.
,
Iny Stein
T.
,
Rosen
N.
,
Kohn
A.
,
Twik
M.
,
Safran
M.
et al. .
GeneHancer: genome-wide integration of enhancers and target genes in GeneCards
.
Database (Oxford)
.
2017
;
2017
:
bax028
.

51.

Mullen
D.J.
,
Yan
C.
,
Kang
D.S.
,
Zhou
B.
,
Borok
Z.
,
Marconett
C.N.
,
Farnham
P.J.
,
Offringa
I.A.
,
Rhie
S.K.
TENET 2.0: identification of key transcriptional regulators and enhancers in lung adenocarcinoma
.
PLoS Genet.
2020
;
16
:
e1009023
.

This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial License (https://creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact journals.permissions@oup.com

Supplementary data

Comments

0 Comments
Submit a comment
You have entered an invalid code
Thank you for submitting a comment on this article. Your comment will be reviewed and published at the journal's discretion. Please check for further notifications by email.