Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 May 1.
Published in final edited form as: Wiley Interdiscip Rev Comput Stat. 2021 Feb 7;14(3):e1553. doi: 10.1002/wics.1553

Integrative clustering methods for multi-omics data

Xiaoyu Zhang 1,#, Zhenwei Zhou 1,#, Hanfei Xu 1, Ching-Ti Liu 1
PMCID: PMC9097984  NIHMSID: NIHMS1674749  PMID: 35573155

Abstract

Integrative analysis of multi-omics data has drawn much attention from the scientific community due to the technological advancements which have generated various omics data. Leveraging these multi-omics data potentially provides a more comprehensive view of the disease mechanism or biological processes. Integrative multi-omics clustering is an unsupervised integrative method specifically used to find coherent groups of samples or features by utilizing information across multi-omics data. It aims to better stratify diseases and to suggest biological mechanisms and potential targeted therapies for the diseases. However, applying integrative multi-omics clustering is both statistically and computationally challenging due to various reasons such as high dimensionality and heterogeneity. In this review, we summarized integrative multi-omics clustering methods into three general categories: concatenated clustering, clustering of clusters, and interactive clustering based on when and how the multi-omics data are processed for clustering. We further classified the methods into different approaches under each category based on the main statistical strategy used during clustering. In addition, we have provided recommended practices tailored to four real-life scenarios to help researchers to strategize their selection in integrative multi-omics clustering methods for their future studies.

Keywords: clustering, integration, multi-view, omics

1 |. INTRODUCTION

Integrative analysis of multi-omics data has drawn the scientific community’s attention. Recent technological advances have generated various omics data with the hope of gaining greater insight into the underlying mechanism of diseases. For example, single-cell RNA sequencing has provided new biological mechanisms in some rare cell populations (B. Hwang et al., 2018; Stark et al., 2019); also, the newly developed Illumina EPIC array has revealed methylation differences associated with complex phenotypes (Mansell et al., 2019); and mass spectrometry-based proteomics has played an essential part in studying proteins that could have a significant impact on biology and medicine (Aebersold & Mann, 2003). Leveraging these multi-omics data potentially provides a more comprehensive view of the disease mechanism or of the biological processes. Based on the research question(s), different integrative methods may respond to different demands. On the other hand, dimensions of different types of omics data vary tremendously and each of them has its own biological properties. For example, each sample may have around 20,000 gene expression profiles, over 480,000 methylation sites, and millions of single-nucleotide polymorphism (SNP). Thus, the data types and the biological questions of interest should be both taken into consideration for multi-omics data analysis. For instance, when integrating SNP data and “other” types of omics data (e.g., transcriptomic, epigenomic, proteomic, and metabolomics), the idea that SNPs affect other omic markers may lead to genome-wide QTL analysis naturally (Sun & Hu, 2016). However, with various interaction networks between those “other” omics data types, the modeling strategy could be different from the one proposed for the integration of SNP data and a single type of “other” omics data (Dimitrakopoulos et al., 2018). Hasin et al. (2017) grouped multi-omics approaches into three categories, “genome first,” “phenotype first,” and “environment first,” depending on the focus of the investigation. This reflects the importance of determining the biological question before conducting the multi-omics analysis.

Disease subtyping is one of the major biological questions of interest which can be addressed by clustering methods. Integrative multi-omics clustering is an unsupervised integrative method specifically used to find coherent groups of samples or features by utilizing information across multi-omics data. It has wide applications, especially in cancer studies. For example, Curtis et al. (2012) integrated copy number and gene expression data and revealed novel subgroups of breast tumors with distinct clinical outcomes. The Cancer Genome Atlas Networks (TCGA) group further demonstrated the existence of four subtypes of breast cancer by combining data from different platforms, including genomic DNA copy number arrays, DNA methylation, exome sequencing, messenger RNA arrays, and microRNA sequencing (Cancer Genome Atlas Network, 2012). Mo et al. (2013) showed distinct tumor subtypes of colorectal cancer through the integrative clustering approach. These multi-omics clustering results provided a novel molecular stratification of cancers, suggesting both the biological mechanisms and potential targeted therapies for the diseases.

However, applying integrative multi-omics clustering is both statistically and computationally challenging due to various reasons, for example, high dimensionality and heterogeneity. Many proposed integrative multi-omics clustering methods have intended to address these challenges. Several recent reviews have discussed those methods (Chauvel et al., 2019; Pierre-Jean et al., 2019; Rappoport & Shamir, 2018; Tini et al., 2019; D. Wang & Gu, 2016). Chauvel et al. evaluated and grouped six methods into two categories based on the mathematical techniques implemented in the integrative clustering. Tini et al. compared five methods and discussed their performance under different conditions of noise and signal strength across data types. Pierre-Jean et al. extended the scope of their survey and included 13 methods. However, it lacked a systematic way to group methods, and its inclusion criteria mainly depended on the availability of R packages. Rappoport et al. gave a more comprehensive review, including multi-omics clustering methods developed either by the bioinformatic or machine learning community with a benchmark. But it lacked mathematical detail for each method. Wang et al. restricted their review to the application of cancer classifications. In general, these reviews did not provide a practical guideline for real-life applications. In this review, we summarized integrative multi-omics clustering methods based on not only the integrating strategy of the multi-omics data but also the statistical approach used during clustering. Specifically, we classified these methods into three categories: concatenated clustering, clustering of clusters, and interactive clustering. We further grouped the methods into different approaches under each category. In addition, we have provided recommended practices under different scenarios for real-life applications for future studies.

We organized the remainder of this review as follows: we first present the methods including their mathematical basis under three categories in Section 2. We then provide recommendations for researchers under four scenarios in Section 3. Finally, we conclude this review in Section 4 with the future challenges of integrative multi-omics clustering methods.

2 |. METHODS

Throughout this review, we denote A as a matrix, a as a vector, a as a scalar, and AT as the transpose of the matrix A. This notation is consistent when using other letters. Xk denotes the n × pk data matrix from the kth type of omics data, where n is the number of samples, pk is the number of features in the kth type of omics data, and K=1Kpk=p (i.e., the total number of features is p). Therefore, Xijk is the value of the jth feature for the ith sample in the k th data type.

We classified integrative multi-omics clustering methods into three major categories based on when and how the multiple omics data are processed for clustering (Figure 1). (1) Concatenated clustering: first construct one data matrix based on all omics data and then perform the clustering analysis. (2) Clustering of clusters: either perform clustering on each omics dataset, followed by integrating the primary clustering results; or transform each omics dataset into a particular form to summarize the samples’ relationship, followed by combining the processed data thus yielding the final clustering results. (3) Interactive clustering: simultaneously integrate data and perform clustering through assigning parameters or component allocation variables to connect multi-omics data. Under each category, we further classified each method into different groups based on its implementation strategy. We summarized all the methods mentioned in this review with their corresponding category and approach in Table 1.

FIGURE 1.

FIGURE 1

Three categories of integrative multi-omics clustering methods. Multi-omics data (e.g., DNA methylation, copy number variation, gene expression) are collected for each sample. Integrative multi-omics clustering methods can be used to analyze such data and produce sample clusters. We summarized those methods into three categories. Concatenated clustering: combine the multi-omics data into one matrix or search for the shared structure, followed by the final clustering; clustering of clusters: Obtain the clustering information from each omics dataset first and follow by the final clustering; interactive clustering: simultaneously integrate multi-omics data and perform clustering

TABLE 1.

Summary of integrative multi-omics clustering methods under three categories and different approaches

Category Approach Method Description Strength Weakness Implementation
Concatenated clustering Joint latent model iCluster (iClusterPlus, iClusterBayes) Assume all omics data originate from a low dimensional latent matrix which can be used for the final clustering with probabilistic model Feature selection Computationally intensive R
moCluster Use the sparse consensus principal component to define a set of latent variables to get the final clustering Efficient with convergence to a deterministic solution Delicate normalization procedure required R
Low-rank approximation LRAcluster Assume different omics data are independent conditional on the stacked parameter matrix with low-rank constraints Convex objective function leading to a global solution No feature selection R
JIVE Decompose each data into three parts: low-rank approximation for joint variation, low-rank individual variation, and residual noise Account for individual data variation; feature selection Only applicable to continuous data; not robust to outliers Matlab, R
Non-negative matrix factorization jNMF (iNMF, intNMF) Approximate each omics data by a product of two non-negative matrices and minimize the approximation error Feature selection Local optimal solution only Matlab, Python, R
K-means related IS-K means Extend sparse K-means for multi-omics data through normalization and incorporate prior knowledge to select biologically meaningful features Can incorporate prior knowledge Only applicable to continuous data; delicate normalization procedure required R
Graph-based PARADIGM Develop a probabilistic graphical model and construct an integrated pathway activity matrix for features which can be used for clustering Can incorporate prior knowledge Pathway knowledge required; need submit data into the designated website to run the analysis Web/API
Clustering of clusters Perturbation-aided COCA Implement consensus clustering approach (generate perturbed datasets through resampling) Direct apply on different omics data without the need of normalization No feature selection NA
PINS (PINSPlus) Generate perturbed datasets by adding Gaussian noise to the original data and choose the optimal number of clusters through perturbation Robust to data with noise No feature selection R
Similarity-based Spectrum Construct sample-wise similarity matrix for each omics data using its proposed kernel first and then combine them to construct a Laplacian matrix followed by spectral clustering to get the final clustering Robust to data with noise; computational efficient No feature selection R
SNF (ab-SNF, NEMO) Construct sample-wise similarity matrix for each omics data first and then fuse them together followed by the final clustering Computational efficient; can deal with mixed type of data No feature selection R, Matlab
CIMLR Multiple kernel learning method that learns the similarity matrix that best fits the data through an optimization procedure constructed by a set of Gaussian kernels Feature selection Gaussian kernels only R, Matlab
rMKL-LPP Multiple kernel learning method that simultaneously optimizes kernel weight and projects data into a lower dimensional space Flexibility of incorporating multiple different kernels No feature selection Upon request
Interactive clustering Dirichlet mixture model-based MDI Use Dirichlet-multinomial mixture model with data dependence captured by parameters at the allocation level Can deal with mixed type of data; no requirement for a consistent clustering structure Computational intense with many parameters to specify Matlab
BCC Use Dirichlet mixture model to simultaneously identify the dependence and heterogeneity across multi-omics data Allow heterogeneity of multi-omics data when identify the overall clustering No feature selection; a consistent clustering structure required R
PSDF Use two-level hierarchy of Dirichlet process mixture model to separate concordant samples with feature selection Feature selection; No requirement for a consistent clustering structure Only integrate two omics data; Discretization of input data required Matlab

Notes: Methods in the parenthesis are extended methods based on the original method in front of the parentheses. NA, not available.

2.1 |. Concatenated clustering

One intuitive strategy for integrative multi-omics clustering is to combine different omics data first and then apply a direct clustering method. When combining omics data, one can either put all the omics data into a big composed matrix or one can search for the shared structure among the omics data. Some normalized procedures may be applied to combine omics data to make them comparable. Performing dimension reduction by searching for the shared structure in the combined omics data can assist the following clustering. In the clustering analysis for multi-omics data, finding shared information from different omics is an inevitable part. The methods in this category find shared information first, and then perform clustering, which is different from the methods in another two categories discuss later. We further summarized methods into the following five groups based on their primary statistical approach.

2.1.1 |. Joint latent model

This group of methods assumes that all omics data share a set of low dimensional latent variables, which generate the observed high-dimensional data. Mathematically, we can write the general model for the kth omics data Xk, as:

XkT=WkZ+Ek,
Z~Nq(0,I),

where Z, a q × n matrix, is the joint latent structure shared by all K types of omics data, Wk is the omics-specific loading matrix, and Ek is the uncorrelated error matrix with zero mean and diagonal covariance matrix ψk=diag(σ1k2,,σpkk2). The core of this approach is to find the joint latent variables, followed by any type of standard clustering algorithm, such as K-means, hierarchical clustering, on the joint latent variables to obtain the final clustering results. In general, this type of approach is easy to implement; however, it has two major challenges: the difficulty in the biological interpretations of latent variables as well as the data heterogeneity among different omics data. Due to different scales, it often requires proper data normalization prior to the analysis. Below, we have illustrated a few representative methods.

iCluster (Shen et al., 2009) assumes a Gaussian joint latent model for which a low dimensional cluster membership matrix is shared by all omics data on the same samples. We can derive the joint likelihood on the assumption of conditional independence of observed omics data given the shared latent variable Z which collectively captures the correlative structure between omics data. iCluster incorporates a lasso type (L1-norm) penalty on the loading matrix W to identify essential features and uses the Expectation-Maximization (EM) algorithm to get the mean of the cluster membership matrix on which a final cluster assignment can be achieved through a standard K-means. iCluster is only applicable to continuous data. iClusterPlus (Mo et al., 2013), the extended iCluster, accommodates continuous, binary, count, and multicategory data through generalized linear models. However, both iCluster and iClusterPlus suffer from an intense computational burden due to evaluating an excessive number of tuning parameters and different numbers of clusters for the analysis. iClusterBayes (Mo et al., 2018) was thus developed to solve this issue. Instead of using regularization in the likelihood to select features, iClusterBayes introduced an additional indicator variable with value 0 or 1 for Bayesian variable selection. With a prior distribution setting for parameters, the Metropolis-Hasting algorithm can be applied to jointly sample the latent and indicator variables from their posterior distribution for statistical inference. As there is no longer a need to fine-tune parameters like in iCluster or iClusterPlus, iClusterBayes is more computationally efficient.

Instead of using the EM algorithm as suggested, above, for the iCluster series, moCluster (Meng et al., 2016) utilizes the sparse consensus principal component analysis and the nonlinear iterative partial least squares algorithm to estimate the latent variables. This method was reported to be 100× to 1000× faster than iCluster/iClusterPlus since it can converge to a deterministic solution. However, it requires a delicate normalization procedure. In addition to the centering and scaling features in each dataset, the authors also weight each data matrix by the inverse of its first eigenvalue to allow different matrices to contribute comparable variance to the first few joint latent variables.

2.1.2 |. Low-rank approximation

This type of approach aims to find the low-dimensional subspace of high-dimensional data and cluster on the reduced subspace. It shares a similar intuition with the joint latent model approach, that is, it assumes a low-dimensional matrix, which well represents the clustering structure of samples across different omics data. The main difference is that the low-rank approximation approach enforces certain constraints on data to introduce low-rank or low-dimensional space instead of assuming joint latent variables although there could be overlapping between these two approaches for some methods.

Low-rank-approximation-based multi-omics data clustering (LRAcluster) (Wu et al., 2015) develops its low-rank approximation based on an integrative probabilistic model. It assumes different datasets are independent conditioning on the parameter matrix. It sets low-rank constraints on the stacked parameter matrix Θ leading to a penalty of model complexity in the joint likelihood of data. The objective function becomes minΘK=1KL(Θk;Xk)+μ|Θ|*, where L(.) is the negative log likelihood function of data based on the probabilistic model (continuous data using Gaussian distribution, binary data using a Bernoulli distribution, and count data using a Poisson distribution), μ is a tuning parameter and |.|* denotes the nuclear norm of the matrix. Singular value thresholding-like method is used to optimize the objective function, and K-means is applied to find the final clustering assignment on the low-dimensional subspace. One appealing part of this method is that its objective function is convex, which leads to a global solution.

Joint and individual variation explained (JIVE) (Lock et al., 2013; O’Connell & Lock, 2016) decomposes each omics dataset into three parts: a low-rank approximation for joint variation, a low-rank individual variation, and residual noise. It can be expressed as XkT=Jk+Ak+Ek,J=(J1T,,JKT)T, where Jk is the submatrix of joint structure J associated with Xk, Ak is the individual structure of Xk, and Ek is the error matrix with zero expectation. Meanwhile, the joint and individual variations are assumed to be uncorrelated. Low-rank constraints are set for both joint variation and individual variation (i.e., rank(J) = q, rank(Ak) = qk). By minimizing the sum of squared error, the joint and individual structure can be estimated iteratively. A cluster analysis on either the joint structure or the individual structure can help cluster samples either based on all the omics data or concerning each omics dataset. Besides, JIVE can be viewed as an extension of the joint latent model by adding an omics-specific term: XkT=WkZ+UkSk+Ek, where the q × n matrix Z is the common score, summarizing the sample variability across K omics data, and Wk is the pk × q loading matrix for the first q components. UkSk is the additional omics-specific term where Uk is the loading matrix, and Sk is the score matrix for the kth omics data. Thus, the joint and individual structures, J and Ak, can be estimated through singular value decomposition-type methods. However, this method is not robust to outliers and only applicable to continuous data.

2.1.3 |. Non-negative matrix factorization

The basic idea of the non-negative matrix factorization (NMF) approach is to approximately decompose the kth omics data matrix Xk into a product of two non-negative matrices, that is, one common basis matrix Z and one omics-specific coefficient matrix Wk. The n × q matrix Z can be used to identify the sample clustering membership and the q × pk matrix Wk can be used to identify features that contribute to clusters. The objective function can be formulated as:

minZ,W1,,WKk=1KXkZWkF,s.t.Z0andWk0,

where ‖.‖F denotes the Frobenius norm. It can be solved through a multiplicative algorithm (Lee & Seung, 1999). This approach is closely related to the joint latent model approach when assuming Z is the common latent structure even though a non-negativity constraint is used here. However, the objective function of NMF is not convex, potentially leading to a local solution only.

Joint NMF (jNMF) (S. Zhang et al., 2012) applies the NMF framework to identify correlative modules (i.e., groups of correlated features for all or a subset of samples), also called multi-dimensional modules (md-modules), in multi-dimensional omics data, which can be further used to cluster samples. jNMF searches for the shared basis matrix Z across multi-omics datasets through minimizing the squared Euclidean error function: minZ,W1,,WKk=1KXkZWkF2.. The coefficient matrices Wk can be used to identify md-modules based on the user-defined criteria such as the largest entity of each column of Wk. Samples can be further grouped into either md-module-specific or not md-module-specific groups based on the columns of shared basis matrix Z for each md-module. jNMF uses the generalized multiplicative update rules to arrive at the shared basis matrix and the coefficient matrices iteratively. Because it is a local optimization procedure, multiple attempts using different initial values are required to reach the final solution. Two proposed integrative NMF methods (iNMF and intNMF) were developed based on the aforementioned jNMF method. iNMF (Yang & Michailidis, 2016) extends jNMF to accommodate the heterogeneous effects from different omics data via an additional term VkWk. Its objective function then becomes minZ,W1,,WK,V1,,VKK=1KXk(Z+Vk)WkF2+λK=1KVkWkF2,Vk0. In other words, iNMF puts a penalty on the Frobenius norm of the heterogeneous term to retain identifiability. It is more robust to heterogeneous noise across different omics data. Moreover, iNMF can additionally incorporate an L1-norm penalty on the elements of the coefficient matrix Wk to induce sparsity in feature selection. intNMF (Chalise & Fridley, 2017), in contrast, focuses on clustering samples using different omics data. It does not assume any distribution of data and uses weights θk to combine different data with objective function as minZ,W1,,WKK=1KθkXkZWk2. Defined by the users, the weights θk for example, can be the maximum of the mean sums of squares among all omics data divided by the mean sum of squares of each omics data (i.e.,θk=max(mean(Xk2),k=1,,K)mean{Xk2}). Instead of a multiplicative update algorithm, intNMF uses alternating least square algorithms to arrive at the estimation since the objective function becomes convex in Z given Wk and vice versa. Only basis matrix Z has to be initialized. The final sample clustering membership is determined by the largest entry in each row of basis matrix Z.

2.1.4 |. K-Means related

The K-means method originally discussed partitioning observations into k sets through minimizing the within-cluster sum of squares (WCSS) (MacQueen, 1967). Assuming the total number of clusters is D, we denote G = (G1, …, GD) the clustering results, with Gd be the collection of samples in cluster d. We can write the objective function of the K-means as:

minGd=1D1ndWCSS(Gd)=minGd=1D1ndi,i'Gdj(Xi,jXi',j)2,

where nd is the number of samples in cluster d, j stands for the jth feature. Minimizing the WCSS is equivalent to maximizing the between-cluster sum of squares (BCSS) since the total sum of squares (TSS) is a constant (i.e., TSS = WCSS + BCSS). Some extensions of K-means methods were proposed to handle omics data (Friedman & Meulman, 2004; Witten & Tibshirani, 2010). Sparse K-means, for example, can effectively select features and perform sample clustering simultaneously (Witten & Tibshirani, 2010). It targets at maximizing the weighted BCSS, subject to constraints on the weights to enforce a sparse solution. To apply a K-means-related approach to multi-omics data, a specific normalization procedure is required to make features from different omics data comparable. This method requires a preselected number of clusters, and it can only apply to continuous data.

Integrative sparse K-means (IS-K means) (Huo & Tseng, 2017) extends sparse K-means to deal with multi-omics data through normalizing the BCSS by the TSS, Rj(G)=BCSSj(G)TSSj(G). Also, it incorporates overlapping group structure of features through adding a group lasso penalty term in the objective function to select biologically meaningful features in the clustering procedure. The feature group information can come from prior knowledge like biological databases.

2.1.5 |. Graph-based

A graph-based approach considers the network structures between features such as pathways among genes, proteins, and so on when integrating multiple omics data. The probabilistic graphical model (PGM) is often used to represent the regulatory structure with parameter inferences, and prior knowledge on the pathways is usually required for this type of approach.

PAthway Recognition Algorithm using Data Integration on Genomic Models (PARADIGM) (Vaske et al., 2010) develops a PGM based on factor graphs that can integrate different genomic and functional genomic datasets to infer the molecular pathways altered in a patient sample. Each node in the factor graph represents different measurement levels of genes, proteins, and so on and can be either activated, nominal, or deactivated relative to a control level. It generates an integrated pathway activity (IPA) matrix whose rows represent different entities (e.g., a protein-coding gene, a small molecule, a complex, a gene family, or an abstract process) and columns represent samples based on multi-omics data. Entities belonging to the same pathway will be grouped in rows. Each element of the IPA matrix is an IPA score, a signed analog of the log-likelihood ratio, representing how likely the entity is activated/null/deactivated in the corresponding sample, for each pathway separately. The IPA matrix can then be used to cluster samples via hierarchical clustering. This method requires prior knowledge of pathways and requires users to upload the data to the designated website to run the analysis.

2.2 |. Clustering of clusters

As the name of this category suggests, clustering of clusters shares the idea of obtaining clustering information from each omics first and then constructing an overall grouping that represents the relationship between samples followed by a final clustering. The primary clustering information can come from many places such as: directly clustering on each omics dataset, clustering on perturbed data, or constructing a sample-wise similarity matrix on each omics dataset. These approaches extract the shared information from different omics’ data after performing clustering for each omics. The methods in this category exhibit a different logic compared to the concatenated clustering, and avoid the possible information loss from the dimension reduction step. We summarized methods under this category into two groups, a perturbation-aided approach and a similarity-based approach, according to their main strategy.

2.2.1 |. Perturbation-aided

The main goal of the perturbation-aided approach is to reach a more reliable clustering assignment with perturbed datasets by either using a resampling technique or adding noise to the original data. A standard clustering algorithm can then be applied to each of the perturbed omics datasets and the agreement of clustering results can be assessed among multiple runs. Assuming that we have H perturbed datasets, an n × n connectivity matrix M(h) represents samples relationship for the hth perturbed data, where n is the number of samples:

Mij(h)={1, if sample i and j belong to the same cluster 0, otherwise ,h=1,,H.

Then an overall n × n consensus matrix C can be calculated based on all connectivity matrices (i.e., M(h), h = 1, …, H) to help determine the final clustering. The construction of the consensus matric C varies among methods. In general, methods within this approach are robust to noisy data because of the perturbation procedure. More details about each method are described below.

Cluster-of-cluster assignments (COCA) (Hoadley et al., 2014) implemented the consensus clustering approach in the problem of integrative multi-omics clustering. Consensus clustering originates from the idea that the clustering results should be robust to the sampling variability (Monti et al., 2003). It uses a resampling technique to generate perturbed datasets followed by multiple runs of a standard clustering algorithm to create connectivity matrices. Then the consensus matrix C can be calculated as a normalized sum of connectivity matrices of all the perturbed datasets: Cij=hMij(h)hIij(h), where the indicator function Iij(h)=1 if both samples i and j are present in the hth perturbed dataset and 0 otherwise. Thus, each entry of C represents the proportion of multiple runs that two samples are clustered together. Hierarchical clustering can then be applied to the distance matrix, 1 − C, to determine the final clustering memberships. COCA implemented the consensus clustering method to integrate multi-omics data, where connectivity matrices were built based on each omics dataset and then put them together to construct the consensus matrix C to represent samples’ relationship. The method was applied on six platforms of omics data including DNA copy number, DNA methylation, mRNA expression, microRNA expression, protein expression, and somatic point mutation, from 12 types of cancer and clustered samples into 11 major subtypes which have shown clinical importance from the Kaplan-Meier survival analysis.

Perturbation clustering for data INtegration and disease Subtyping (PINS) (T. Nguyen et al., 2017) generates perturbed datasets by adding noise and aims to choose the optimal number of clusters through perturbation before conducting the final clustering. PINS first applies K-means on the original data and builds (D − 1) connectivity matrices M2, …, MD for all possible numbers of clusters, where D is the largest number of clusters considered. It then generates H perturbed datasets by adding Gaussian noise to the original data and repeating the same process to build connectivity matrices on each perturbed dataset. Then by averaging those connectivity matrices for each possible number of clusters, we can get perturbed connective matrices M2, …, MD. The difference matrix M˜d, with M˜ijd=|MijdM'ijd|, between the original and the perturbed connectivity matrix reflects the stability of clustering. The smaller M˜ijd, the more robust the connectivity between sample i and j, which can be used to determine the optimal number of clusters based on the most robust clustering results against perturbation. For K types of omics data, the optimal number of clusters is chosen based on each omics data as above and K connectivity matrices then can be built using the optimal number of clusters as M1, …, MK. Then the average pair-wise connectivity matrix (i.e., the consensus matrix) C can be calculated as Cij=K=1KMkK. A similarity-based clustering algorithm such as hierarchical clustering can be applied on the distance matrix 1 − C to determine the final clustering memberships. PINS is robust against noise by building the resilience of sample connectivity under perturbations. However, its running time is relatively long and there is no weight adjustment for different omics data.

PINSPlus (H. Nguyen et al., 2019) extended PINS by utilizing ensemble strategy (i.e., applying more than one type of clustering algorithm) on the consensus matrix to identify the clustering that agrees the most among multi-omics data. Thus, it can ensure the identified clusters are consistent and robust against the choice of clustering algorithms. Also, an early stopping criterion is implemented for the process of building perturbed connectivity matrices. Together with parallel computing, the computational efficiency of PINSPlus is largely improved compared to PINS.

2.2.2 |. Similarity-based

In general, this type of approach constructs a sample similarity matrix for each omics dataset first and then integrates similarity matrices across all omics data into one followed by the final clustering. Assuming each omics dataset, we have sample data (x1, …, xn) where xi, i = 1, …, n, is a vector for sample i with all features in that omics dataset. The key component of the similarity-based approach is to construct an n × n sample similarity matrix W based on different kernels defined by each method to represent the samples’ relationship. This approach becomes more popular these days since it can address the common challenges in the integrative multi-omics clustering methods. By constructing a sample-wise similarity matrix, it overcomes not only the data heterogeneity problem, but also the problem of small n large p. However, this approach generally cannot perform feature selection. Related methods are further described below.

Spectrum (John et al., 2020) extended the spectral clustering to be able to handle multi-omics data. Spectral clustering was originally designed for single-view data as a similarity- and graph-based method. It mainly uses eigenvectors of the Laplacian matrix derived from the data to perform further clustering (Ng et al., 2001). Given a dataset with n samples and a similarity matrix W, it first constructs the similarity graph to model the local neighborhood relationships between samples and then computes the Laplacian matrix L. It uses first d eigenvectors of L to represent data and follows with the standard K-means (von Luxburg, 2007). Equivalently, the objective function is maxtraceU(UTLU), s.t. UTU=I,s:t:UTU = I , where U is an n × d matrix. Apply the K-means algorithm on U to obtain the cluster memberships (Ng et al., 2001). Kumar et al. (2011) proposed a spectral clustering method under multi-view setting through a co-regularization framework with an objective function of maxU1,,UKk=1Ktrace(UkTLkUk)+λ1k,lK,kltrace(UkUkTUlUlT), s.t. UkTUk=I. However, it was not designed for omics data. Spectrum, on the other hand, was developed for multi-omics data. The authors of Spectrum proposed a self-tuning density-aware kernel as Wij=exp(ρ2(xi,xj)ϵiϵj(CNN(xi,xj)+1)), where, ρ(xi, xj) denotes the Euclidean distance between sample i and j, ϵi is local scaling parameter for sample i, CNN(xi, xj) is the number of samples in the inter-section between the two sets of nearest neighbors of sample i and j, based on Zelnik-Manor self-tuning kernel (Zelnik-Manor & Perona, 2005) and Zhang density-aware kernel (X. Zhang et al., 2011). Spectrum uses a tensor product graph integration and diffusion technique to combine similarity matrices across multi-omics data and reduce noise (Shu & Latecki, 2016). Finally, Laplacian matrix L is constructed based on the combined similarity matrix and Gaussian mixture model clustering is used to get the final clustering memberships on the eigenvector matrix of L.

Similarity network fusion (SNF) (B. Wang et al., 2014) is a well-represented method belonging to the similarity-based approach of integrative multi-omics clustering. SNF uses a scaled exponential similarity kernel to construct the sample similarity matrix W with Wij=exp(ρ2(xi,xj)μϵi,j), where, ρ(xi, xj) denotes the Euclidean distance for continuous variables between sample i and j, μ is a hyperparameter that can be empirically set and ϵi,j=mean(ρ(xi,Ni))+mean(ρ(xj,Nj))+ρ(xi,xj)3 which is used to eliminate the scaling problem, mean(ρ(xi, Ni)) is the average value of the distances between sample i and each of its neighbors. The authors proposed to use chi-squared distance for discrete variables and agreement-based measure for binary variables. After similarity matrices being constructed for samples from available datasets, they will be fused into one similarity matrix to represent the full spectrum of underlying data. An n × n normalized similarity matrix P and an n × n affinity matrix S are defined as below for the integration or fusion process for multiple similarity matrices based on different omics data:

Pij={Wij2liWil,ji12,j=iandSij={WijlNiWil,jNi0, otherwise ,

where Ni is the set of neighbors for sample i. Then the similarity fusion process is conducted iteratively by:

P(t+1)k=Sk×(mkP(t)mK1)×(Sk)T,k=1,,K,

for the tth iterations and the kth omics data. The fused normalized similarity matrix is Pfused =K=1KP(t+1)kK after t iterations. Spectral clustering (Ng et al., 2001) then can be applied on the Pfused to generate the final clustering.

Association-signal-annotation boosted SNF (ab-SNF) (Ruan et al., 2019) extended SNF by adding weights to features when constructing sample similarity matrix. The weights can be determined by the feature-level association strengths with the outcome, such as association p-value, as well as annotation signals, such as relationship indicators between a gene and a disease. More specifically, assuming we have a continuous feature m, we can calculate the p-value of feature m (i.e., pm), from the model comparing disease samples with normal samples at feature m. Then the feature-level weight can be defined as Wtm=log10(pm)m=1M(log10(pm)), where M is the total number of features of that data type. The weighted distance ρ(xi,xj)=m=1Mwtm(XimXjm)2. For a binary feature m, the weight can be an indicator function to represent whether a feature is important based on prior knowledge, such as if we know a gene m is a mutation gene for a certain disease, we can set wtm = 1 and wtm = 0 otherwise. Then the weighted distance becomes ρ(xi,xj)=m=1Mwtm|XimXjm|. By incorporating the weights, ab-SNF was reported to outperform the original SNF in disease subtyping. However, SNF and ab-SNF both need a separate procedure to handle missing values if not all omics data available for each sample. This could be an issue in real-world practice. NEighborhood based Multi-Omics clustering (NEMO) (Rappoport & Shamir, 2019) was, therefore, developed to address this issue by allowing some samples to have only partial omics data available. It uses a similarity measure based on the radial basis function kernel (Buhmann, 2009) to construct a similarity matrix for the kth omics dataset, Wijk=12πϵijkexp(xikxjk2ϵijk2) where is a normalizing factor which controls for the density of samples by averaging the squared distance of the ith and jth samples to their nearest neighbors and the squared distance between these two samples. Then NEMO defines the relative similarity matrix RS for each omics dataset to measure the similarity between two samples relative to their κ nearest neighbors, RSijk=WijkrrikWirk×I(jηik)+WijkrηjkWjrk×I(iηjk), where I(.) is the indicator function. We can obtain the average relative similarity matrix ARS by averaging the relative similarity matrix across omics data with observed values, ARSij=1|JMij|kJMijRSijk, where JMij denotes the omic types available for both samples. Again, spectral clustering can be applied to the ARS to get the final clustering.

Cancer Integration via Multi-kernel Learning (CIMLR) (Ramazzotti et al., 2018) aims to integrate multi-omics data to reveal molecular subtypes of cancer. It is an extension of Single-cell Interpretation via Multi-kernel LeaRning (SIMLR) (B. Wang et al., 2017). Unlike SNF that uses one kernel to construct a sample similarity matrix, SIMLR is a multikernel learning method that learns the similarity matrix that best fits the data by combing multiple kernels. SIMLR defines each kernel as: Kernel(xi,xj)=1ϵij2πexp(xixj222ϵij2), where ϵij can be calculated with different scales: ϵij=σ(μi+μj)2,μi=lKNN(xi)xixj2κ, where KNN(xi) represents samples that are top κ neighbors of sample i. Thus, each kernel is decided by a pair of parameters (σ, κ). CIMLR extended SIMLR by constructing the same number of Gaussian kernels for K omics. All constructed kernels will be used to find the n × n sample similarity matrix W through the following optimization procedure:

minW,L,vi,j,mvmKernelm(xi,xj)Wij+βWF2+γtr(LT(InW)L)  +ρmvmlogvm s.t. LTL=ID,mvm=1,vm0,jWij=1, and Wij0,

where D is the number of clusters, m is the kernel index over all kernels across K omics data, νm is the weight for the mth kernel, β and γ are non-negative tuning parameters, ‖. ‖F denotes the Frobenius norm, and L is an auxiliary low-dimensional matrix enforcing the low rank constraint on W. It then applies K-means based on the similarity matrix W for the final clustering. Additionally, CIMLR can also perform feature selection through a hypergeometric test.

Regularized multiple kernels learning with locality preserving projections (rMKL-LPP) (Speicher & Pfeifer, 2015) is also a multikernel learning method that can integrate multi-omics data and perform cancer subtype identification. It adopts the multiple kernels learning for dimensionality reduction (MKL-DR) framework (Lin et al., 2011) which integrates multiple kernel learning (i.e., optimizes the weights that linearly combines a set of kernel matrices to generate a unified kernel matrix) into the graph embedding (S. Yan et al., 2007) for dimensionality reduction. MKL-DR defines different types of kernels based on the data type, the mth dissimilarity-based kernel matrice Kernelm(xi,xj)=exp(dm2(xi,xj)ϵm2), where ϵm is a positive constant. rMKL-LPP extended MKL-DR by adding a regularization term to avoid overfitting and it applied locality preserving projections algorithm (LPP) (He & Niyogi, 2003) for dimensionality reduction with the optimization problem as:

minA,βi,j=1nATKiβAKjβ2Wij s.t. i,j=1nATKiβ2Rij=const.,β1=1 ,βm0,m=1,2,,MKi=( Kernel 1(x1,xi) Kernel M(x1,xi) Kernel 1(xn,xi) Kernel M(xn,xi)),

where A is an n × q projection matrix and q is user-defined (q is usually smaller than the number of features p to achieve dimension reduction), β is an M × 1 coefficient vector, Ki is an n × M kernel matrix for sample i which can be constructed by different kernel functions, M is the total number of kernels and each omics dataset may have multiple kernels. The (i, j)-entry of the similarity matrix W is equal to 1 if iNκ(j) or jNκ(i) and 0 otherwise, Nl(i) is the set of the κ nearest neighbors for sample i, and Rij=l=1nWil, if i = j and 0 otherwise. A and β are iteratively optimized until convergence and K-means can be performed on the projected samples ATKiβ,i=(1,,n) to obtain the final clustering. This method has the flexibility of incorporating multiple kernels per data type. This not only improves the performance but also removes the need of preselecting the optimal kernel. The authors applied this method on five cancer datasets and identified biologically meaningful subgroups for cancers with significantly different survival.

2.3 |. Interactive clustering

Instead of performing two-step procedures like those above-mentioned, methods covered in this category conduct the integration and clustering simultaneously. These methods assign parameters or allocation variables to link the dependence across different omics data. Under such settings, a consistent clustering structure across multi-omics data is not necessary, which provides a more flexible setup. Methods in this category usually incorporate ideas of the Dirichlet mixture model and Bayesian statistics.

2.3.1 |. Dirichlet mixture model-based

This type of approach usually assumes that data originate from a Dirichlet mixture model with a general form: p(x)=m=1Mπmf(xθm),m=1,,M, where p(x) denotes the probability density for data with M components, πm’s are mixture proportions, f is a parametric density with associated parameters θm and different types of data can be modeled using different densities. Let (u1, …, un) be the component allocation variables for n samples where ui ∈ {1, …, M}, i = 1, …, n and πm = P(ui = m). Under a Bayesian framework, Dirichlet priors are usually put on Π = (π1, …, πM). Gibbs sampling can be used to approximate the posterior distributions of parameters of interest and the posterior of component allocation variables are directly related to the final clustering of samples. When M → ∞, the model becomes a Dirichlet process (DP). On one hand, this approach provides flexible probabilistic models for different omics data. On the other hand, it naturally captures the clustering structure through decomposing data into M components. However, this approach usually requires one to specify many parameters in advance.

Multiple dataset integration (MDI) (Kirk et al., 2012) can integrate multiple omics data of different types (e.g., continuous, categorical, time series). It was originally designed for clustering genes but it can be easily applied for clustering samples. For n samples/genes from K omics data, MDI models each omics data using a Dirichlet-multinomial allocation (DMA) mixture model (Green & Richardson, 2001) with component allocation variables uik ∈{1, …M}, i = 1, …, n; k = 1, …, K; M is set by users and was set as n/2 here. MDI then links all DMA models at the level of the component allocation variables via the conditional prior p(ui1,,uiKϕ)k=1Kπuikkk=1K1l=k+1K(1+ϕklI(uik=uil)), where ϕ is a collection of K(K − 1)/2 parameters that controls the strength of association between pair of omics data, πuikk is the mixture proportion associated with component uik in the kth DMA model. Gibbs sampling is used to approximate the parameters’ posterior probabilities. MDI first identifies the samples/genes that tend to be allocated to the same component across a subset of or all omics data followed by the final clustering using the sampled component allocations. One main advantage of MDI is that it does not require the common clustering structure across all omics data. As there are many parameters to specify, the scalability of MDI needs improvement.

Bayesian consensus clustering (BCC) (Lock & Dunson, 2013) aims to simultaneously identify the dependence (i.e., overall clustering) and the heterogeneity (i.e., omics-specific clustering) across multi-omics data. Similar to MDI, BCC models each omics dataset with a Dirichlet mixture model. But BCC assumes there is an overall clustering across multi-omics data and a separate omics-specific clustering adhere to the overall clustering. Let Let lik(1,,M) represent the kth omics-specific component allocation variable and ui ∈ (1, …, M) represent the overall component allocation variable for sample i. The dependence between omics-specific clustering lk=(l1k,,lnk) and the overall clustering u = (u1, …, un) is modeled through the following:

P(lik=mui)=v(lik=m,ui,αk)={αk, if ui=m1αkM1, otherwise ,

where αk[1M,1] controls the adherence of the kth omics data to the overall clustering. M is selected to maximize the mean of omics-specific adherence to the overall clustering. Then the probability for a sample belongs to an omics-specific cluster is P(lik=mΠ)=πmαk+(1πm)1αkM1. And based on Bayes’ rule, the conditional distribution of ui can be written as P(ui=mΠ,{Lk,αk}k=1K)πmk=1Kv(lik,ui=m,αk). Again, through the Gibbs sampling procedure, posterior distributions of omics-specific and overall allocation variables can be estimated to determine the final clustering.

Patient-specific data fusion (PSDF) (Yuan et al., 2011) is based on Bayesian nonparametric modeling. Unlike MDI and BCC, it utilizes a two-level hierarchy of the DP mixture model (Antoniak, 1974; Ferguson, 1973) and mainly integrates two omics datasets (e.g., copy number and gene expression data). DP mixture model may be derived from the above general form of Dirichlet mixture model when M → ∞ (Rasmussen, 2000). The sample i is indicated as fused (ri = 1), if the clustering structure for this sample between two omics data is concordant, or as unfused (ri = 0) if it is contradictory. The prior probability of fusion is defined by P(ri = 1) = w, where w can be set by users, w = 0.5 in the PSDF paper. The hyperparameters of the hierarchical DP consist of the baseline probability measure H, and the concentration parameters γ and α0. This method allows the possibility of taking the product of likelihoods over the two omics data, so if sample i is fused, θi = (θ1i θ2i)~F3; if unfused, θ1i~F1 and θ2i~F2, where θ represents the likelihood parameters under each case. And then we have F1~DP(α0,F0(1)), F2~DP(α0,F0(2)), F3~DP(α0, F0), and F0(θ1, θ2)~DP(γ, H), where F0(k) represents the marginal distribution of θk under F0, k = 1, 2. Additional indicator parameters for features are introduced for the feature selection purpose. Gibbs sampling is used to estimate the posterior probability of parameters of interest. By incorporating the sample fusion information in the DP mixture model, it no longer requires the assumption of a consistent clustering structure between two omics datasets, which shares the intention with MDI. In principle, this method can be extended to integrate more than two omics datasets. However, the authors reported that it would become unwieldy if they did so. Another limitation of PSDF is that it can only handle discretized input data, so data preprocessing is required before modeling.

3 |. IMPLEMENT RECOMMENDATION

We have surveyed a number of integrative multi-omics clustering methods based on their theoretical properties. When it comes to real-life applications, it is necessary to pinpoint the appropriate methods for the various circumstances. Here we list four different situations that researchers commonly face with recommended methods in Table 2. We also point out the difference between methods under the same scenario. Overall, we hope our recommendations can help researchers to strategize their analyses using integrative multi-omics clustering for their future research.

TABLE 2.

Four scenarios with recommended methods

Scenarios Required characteristics for method Recommended methods
I. (Feature selection): The need to identify clinically relevant disease subtypes and driving molecular signatures which can be targeted for treatment Performing both sample clustering and feature selection iCluster; iClusterPlus; iClusterBayes; intNMF; IS-K means; CIMLR; PSDF
II. (Mixed-type data): Large scale genomic data of mixed-type in large consortia Integrating mixed type of data iClusterPlus; iClusterBayes; moCluster; LRAcluster; MDI; SNF; CIMLR; rMKL-LPP; PINS; PINSPlus
III. (Computational efficiency): Concern on the computational resources and consumption of time Computationally efficient Spectrum; SNF; ab-SNF; NEMO; CIMLR; rMKL-LPP
IV. (Knowledge integration): Leveraging the prior knowledge Incorporating prior information IS-K means; PARADIGM

Scenario I (feature selection):

Many large-scale cancer genomics studies have demonstrated the benefit of using proper integrative multi-omics clustering methods to generate biologically meaningful cancer subtypes and to identify potential therapeutic targets, for example, studies related to breast cancer, lung cancer, and stomach cancer (Cancer Genome Atlas Network, 2012; Cancer Genome Atlas Research Network, 2012, 2014). Therefore, the capabilities of clustering samples and feature selection are both desired, especially in translational research for precision medicine. The following methods would meet the needs: iCluster and its extensions (iClusterPlus and iClusterBayes), intNMF, IS-K means, CIMLR, and PSDF. iClusterPlus extended iCluster by allowing the integration of multi-type omics data through generalized linear models. iClusterBayes is more computationally efficient than iCluster and iClusterPlus based on a Bayesian model with no tuning parameters. intNMF is an NMF approach without assumption of model distribution. IS-K means can additionally incorporate prior knowledge to identify biologically meaningful driving features. CIMLR’s feature selection procedure cannot be achieved simultaneously with clustering. PSDF utilizes a more flexible nonparametric Bayesian method to cluster samples and select informative features but becomes unwieldy when applying to more than two omics datasets.

Scenario II (mixed-type data):

An unprecedented amount of mixed-type genomic data, binary (somatic mutation), categorical (copy number gain, normal, loss), and continuous (gene expression), for various cancers have been provided by large consortia or in public depositories such as TCGA http://cancergenome.nih/gov, Gene Expression Omnibus http://www.ncbi.nlm.nih/gov/geo/, and Sequence Read Archive http://www.ncbi.nlm.nih.gov/sra. The integrative clustering methods that can deal with mixed-type data are demanded for this scenario. Therefore, we recommended the following methods: iClusterPlus, iClusterBayes, moCluster, LRAcluster, MDI, SNF, CIMLR, rMKL-LPP, PINS, and PINSPlus. A generalized linear model is usually applied to incorporate mixed-type of data for likelihood-based methods such as iClusterPlus, iClusterBayes, and LRAclusters. iClusterBayes, moCluster, and LRAclusters have less computational cost than iClusterPlus. moCluster requires different normalization steps for different types of data. LRAcluster can result in a stable global solution as its objective function is convex. MDI does not require a consistent clustering structure across multi-omics data. SNF, CIMLR, and rMKL-LPP all belong to the similarity-based approach which is generally more computationally efficient. SNF cannot select features. CIMLR can prioritize features through a hypergeometric test. rMKL-LPP uses multiple kernels learning method and adds a regularization term to avoid overfitting during the optimization procedure. PINS and PINPlus both are robust to noisy data, but can be computationally intensive.

Scenario III (computational efficiency):

Although computational capacity has exponentially increased over time, not every researcher has access to a cost-efficient computing infrastructure. Even with enough computational resources, the scalability and consumption of time could be a concern when analyzing multi-omics data. Computationally efficient methods become much more appealing under this setting. Therefore, methods belonging to similarity-based approach are recommended since they are quite efficient by integrating omics data in the space of samples, including SNF and its extensions ab-SNF and NEMO, CIMLR, and rMKL-LPP. In general, the similarity-based approach cannot perform feature selection. Spectrum uses its own proposed kernel to construct sample-wise similarity matrix. SNF uses Euclidean distance between samples and exponential similarity kernel to construct the similarity matrix. ab-SNF is a weighted version of SNF which incorporates phenotype information. NEMO extended SNF to include partially available data for samples. CIMLR and rMKL-LPP are both multiple kernel learning methods with optimization procedure. CIMLR only considers Gaussian kernels, but it can select features. rMKL-LPP has the flexibility of incorporating different kernels per omics dataset.

Scenario IV (knowledge integration):

There is growing biological knowledge, for example, interactions between genes, the potential cis-acting regulatory mechanism between copy number variation, and methylation and gene expression. Researchers may want to utilize this accumulated knowledge to guide the clustering procedure for more biologically meaningful results. Also, there are many publicly available databases providing the related information, such as pathway databases KEGG (Ogata et al., 1999), Reactome (Joshi-Tope et al., 2005), Gene Ontology (Ashburner et al., 2000), and so on. However, only limited integrative multi-omics clustering methods can borrow prior biological information, including IS-K means and PARADIGM. IS-K means groups features based on prior knowledge (e.g., pathway information) and incorporates them in the objective function through a group lasso penalty. Thus, it can select critical groups (e.g., pathways) during the clustering procedure although it can only deal with continuous data. PARADIGM can incorporate pathway information, but it requires users to upload data to the designated website to perform the analysis.

4 |. CONCLUSION

We have summarized integrative multi-omics clustering methods into three general categories, concatenated clustering, clustering of clusters, and interactive clustering based on when and how multi-omics data are processed for clustering. Depending on the main strategy used during clustering, we further classified methods into different approaches under each category. We discussed the mathematical basis for each method and its strengths and weaknesses. One uniqueness of this work is that we also outlined four general scenarios with preferred methods for researchers to strategize their selection of integrative multi-omics clustering methods for their future studies.

Concatenated clustering is a straightforward strategy for integrative multi-omics clustering, following a natural logic that finds shared information first and performs clustering base on the shared information later. This is an obvious difference comparing to another two categories of methods. Joint latent model, low-rank approximation, and NMF intuit that a low-dimensional matrix can represent the sample clustering membership across multi-omics data. Notably, some methods can be grouped into more than one of these three approaches based on how the data matrix is decomposed. For example, JIVE can be viewed as a low-rank approximation approach if the data matrix is composed of the summation of low-rank constrained joint structure and individual structure, and an error matrix. JIVE also can be viewed as a joint latent model approach if the joint structure is further constructed by a product of a joint latent matrix and data-specific loading matrix. In addition, if all elements in the joint latent matrix and data specific loading matrix are restricted to be non-negative, JIVE even can be viewed as an NMF approach. Noisy data is commonly seen in the integrative analysis of multi-omics data, and concatenated clustering methods are generally sensitive to data with noise. Methods from clustering of clusters, on the contrary, perform well for noisy data. The perturbation-aided approach itself is based on perturbation generated by either resampling or adding noise to the original data. Therefore, methods from the perturbation-aided approach generally can provide a more reliable clustering result. Similarity-based approach such as Spectrum and SNF were also reported robust to noise (John et al., 2020; Tini et al., 2019). One major concern for clustering of clusters is whether the primary clustering information across different omics data is consistent before performing further integrative clustering. If not, the final clustering results may not be meaningful. Thus, it is worth applying some measures to evaluate the consistency of primary clustering results, such as the Rand index (Rand, 1971), adjusted Rand index (Hubert & Arabie, 1985), Jaccard similarity (Levandowsky & Winter, 1971), variation of information (Meilă, 2003), and so on.

As mentioned in the very beginning, determining the biological question before conducting the multi-omics analysis is very important. We focus on the disease subtyping using multi-omics data which can be addressed by the integrative multi-omics clustering methods included in this review. In addition, people may also be interested in identifying potential therapeutic targets for diseases which is related to clustering methods that can select features, as discussed in the above Scenario I of implement recommendation. Feature selection can be achieved by incorporating a penalty term in the objective function or by adding Bayesian indicator variables/parameters in many methods from concatenated clustering and interactive clustering. However, most methods from clustering of clusters cannot perform feature selection because either the perturbation procedure or the similarity construction procedure relies on information from all features, except for CIMLR which includes a second step to select features through a hypergeometric test. Feature selection also has been studied in the model-based clustering methods which incorporate variable selection procedure although not under multi-omics settings (Fop & Murphy, 2018; Guo et al., 2010; Zhou et al., 2009). Besides, some of our included methods can address some specific questions other than disease subtyping. For example, jNMF can identify correlated profiles across different type of measures (e.g., gene expression, DNA methylation, microRNA expression); PARADIGM can infer sample-specific pathway activities from multi-omics data.

Some limitations of this review must be acknowledged. First, we focus more on integrative multi-omics clustering methods that can lead to direct sample assignments. Some other integrative clustering methods were not included here, for example, correlation and covariance-based methods and their extensions (Hotelling, 1936; D. Hwang et al., 2004; Witten & Tibshirani, 2009; Wold et al., 2001). They can also be interesting if researchers want to disentangle the correlations between omics data. Second, because we assume that the same group of features contributes to the clusters instead of different groups of features contributing to different clusters, only one-dimensional clustering methods were discussed here. For the two-dimensional clustering methods, for example, bi-clustering, people may consult with a recent systematic review by Padilha and Campello (2017). Third, we only listed four common scenarios with recommendations in this work for a general guidance or as a starting point. In real applications, situations can be much more complicated and require more effort to choose the appropriate method.

Even though many integrative multi-omics clustering methods have been developed as described above, many potential areas still require further investigation. For example, most methods require a common clustering structure shared by all omics data. This may not be the case in real-world data, especially when the omics data are generated from very different aspects of the same samples. How to properly assess this inconsistent clustering structure among multi-omics data is difficult. Even though we have methods like MDI and PSDF which can address part of this issue, they have limitations such as too many parameters to specify and computational cost. Therefore, there is still much work to do in this area. Current clustering methods generally assume that samples can all be grouped into some clusters. However, some samples may just be outliers due to many reasons, for example, mistakenly measured. Therefore, these samples should not be considered for any clusters. Currently, there is a paucity of methods that can deal with this situation. This could be especially important in biological studies. As the quick increase of our domain knowledge, methods that can incorporate prior information is worthy of further study in the near future. Currently there are several methods available for performing clustering while taking into account for prior knowledge (Dotan-Cohen et al., 2007; Huang & Pan, 2006; Tari et al., 2009; Verbanck et al., 2013), as well as methods for integrative analysis for multi-omics data assisted by prior knowledge (de Tayrac et al., 2009; Tong et al., 2020; J. Yan et al., 2018). It is an interesting and important research topic to develop methods for clustering of multi-omics data that take advantage of prior information.

Funding information

National Heart, Lung, and Blood Institute, Grant/Award Number: R01HL151855; National INSTITUTE OF ARTHRITIS AND MUSCULOSKELETAL AND SKIN DISEASES, Grant/Award Number: R01AR072199; National Institute of Diabetes and Digestive and Kidney Diseases, Grant/Award Number: R01DK122503

Footnotes

CONFLICT OF INTEREST

The authors have declared no conflicts of interest for this article.

RELATED WIREs ARTICLE

Model-based cluster analysis

REFERENCES

  1. Aebersold R, & Mann M (2003). Mass spectrometry-based proteomics. Nature, 422(6928), 198–207. 10.1038/nature01511 [DOI] [PubMed] [Google Scholar]
  2. Antoniak CE (1974). Mixtures of Dirichlet processes with applications to Bayesian nonparametric problems. The Annals of Statistics, 2(6), 1152–1174. 10.1214/aos/1176342871 [DOI] [Google Scholar]
  3. Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, … Sherlock G (2000). Gene ontology: Tool for the unification of biology. The Gene Ontology Consortium. Nature Genetics, 25(1), 25–29. 10.1038/75556 [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Buhmann MD (2009). Radial basis functions. Cambridge: Cambridge University Press. [Google Scholar]
  5. Cancer Genome Atlas Network. (2012). Comprehensive molecular portraits of human breast tumours. Nature, 490(7418), 61–70. 10.1038/nature11412 [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Cancer Genome Atlas Research Network. (2012). Comprehensive genomic characterization of squamous cell lung cancers. Nature, 489 (7417), 519–525. 10.1038/nature11404 [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Cancer Genome Atlas Research Network. (2014). Comprehensive molecular characterization of gastric adenocarcinoma. Nature, 513(7517), 202–209. 10.1038/nature13480 [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Chalise P, & Fridley BL (2017). Integrative clustering of multi-level ‘omic data based on non-negative matrix factorization algorithm. PLoS One, 12(5), e0176278. 10.1371/journal.pone.0176278 [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Chauvel C, Novoloaca A, Veyre P, Reynier F, & Becker J (2019). Evaluation of integrative clustering methods for the analysis of multi-omics data. Briefings in Bioinformatics, 21, 541–552. 10.1093/bib/bbz015 [DOI] [PubMed] [Google Scholar]
  10. Curtis C, Shah SP, Chin SF, Turashvili G, Rueda OM, Dunning MJ, … Aparicio S (2012). The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature, 486(7403), 346–352. 10.1038/nature10983 [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. de Tayrac M, Le S, Aubry M, Mosser J, & Husson F (2009). Simultaneous analysis of distinct omics data sets with integration of biological knowledge: Multiple factor analysis approach. BMC Genomics, 10(1), 32. 10.1186/1471-2164-10-32 [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Dimitrakopoulos C, Hindupur SK, Hafliger L, Behr J, Montazeri H, Hall MN, & Beerenwinkel N (2018). Network-based integration of multi-omics data for prioritizing cancer genes. Bioinformatics, 34(14), 2441–2448. 10.1093/bioinformatics/bty148 [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Dotan-Cohen D, Melkman AA, & Kasif S (2007). Hierarchical tree snipping: Clustering guided by prior knowledge. Bioinformatics, 23 (24), 3335–3342. 10.1093/bioinformatics/btm526 [DOI] [PubMed] [Google Scholar]
  14. Ferguson TS (1973). A Bayesian analysis of some nonparametric problems. The Annals of Statistics, 1(2), 209–230. 10.1214/aos/1176342360 [DOI] [Google Scholar]
  15. Fop M, & Murphy TB (2018). Variable selection methods for model-based clustering. Statistics Surveys, 12(0), 18–65. 10.1214/18-ss119 [DOI] [Google Scholar]
  16. Friedman JH, & Meulman JJ (2004). Clustering objects on subsets of attributes (with discussion). Journal of the Royal Statistical Society Series B, 66(4), 815–849. [Google Scholar]
  17. Green PJ, & Richardson S (2001). Modelling heterogeneity with and without the Dirichlet process. Scandinavian Journal of Statistics, 28 (2), 355–375. 10.1111/1467-9469.00242 [DOI] [Google Scholar]
  18. Guo J, Levina E, Michailidis G, & Zhu J (2010). Pairwise variable selection for high-dimensional model-based clustering. Biometrics, 66 (3), 793–804. 10.1111/j.1541-0420.2009.01341.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Hasin Y, Seldin M, & Lusis A (2017). Multi-omics approaches to disease. Genome Biology, 18(1), 83. 10.1186/s13059-017-1215-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. He X, & Niyogi P (2004). Locality preserving projections. Advances in neural information processing systems, 16(16), 153–160. [Google Scholar]
  21. Hoadley KA, Yau C, Wolf DM, Cherniack AD, Tamborero D, Ng S, … Stuart JM (2014). Multiplatform analysis of 12 cancer types reveals molecular classification within and across tissues of origin. Cell, 158(4), 929–944. 10.1016/j.cell.2014.06.049 [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Hotelling H (1936). Relations between two sets of variates. Biometrika, 28(3/4), 321. 10.2307/2333955 [DOI] [Google Scholar]
  23. Huang D, & Pan W (2006). Incorporating biological knowledge into distance-based clustering analysis of microarray gene expression data. Bioinformatics, 22(10), 1259–1268. 10.1093/bioinformatics/btl065 [DOI] [PubMed] [Google Scholar]
  24. Hubert L, & Arabie P (1985). Comparing partitions. Journal of Classification, 2(1), 193–218. 10.1007/bf01908075 [DOI] [Google Scholar]
  25. Huo Z, & Tseng G (2017). Integrative sparse K-means with overlapping group lasso in genomic applications for disease subtype discovery. The Annals of Applied Statistics, 11(2), 1011–1039. 10.1214/17-AOAS1033 [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Hwang B, Lee JH, & Bang D (2018). Single-cell RNA sequencing technologies and bioinformatics pipelines. Experimental & Molecular Medicine, 50(8), 96. 10.1038/s12276-018-0071-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Hwang D, Stephanopoulos G, & Chan C (2004). Inverse modeling using multi-block PLS to determine the environmental conditions that provide optimal cellular function. Bioinformatics, 20(4), 487–499. 10.1093/bioinformatics/btg433 [DOI] [PubMed] [Google Scholar]
  28. John CR, Watson D, Barnes MR, Pitzalis C, Lewis MJ, & Cowen L (2020). Spectrum: Fast density-aware spectral clustering for single and multi-omic data. Bioinformatics, 36(4), 1159–1166. 10.1093/bioinformatics/btz704 [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Joshi-Tope G, Gillespie M, Vastrik I, D’Eustachio P, Schmidt E, de Bono B, … Stein L (2005). Reactome: A knowledgebase of biological pathways. Nucleic Acids Research, 33(Database issue), D428–D432. 10.1093/nar/gki072 [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Kirk P, Griffin JE, Savage RS, Ghahramani Z, & Wild DL (2012). Bayesian correlated clustering to integrate multiple datasets. Bioinformatics, 28(24), 3290–3297. 10.1093/bioinformatics/bts595 [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Kumar A, Rai P, & Daume H (2011). Co-regularized multi-view spectral clustering. Advances in neural information processing systems, 24, 1413–1421. [Google Scholar]
  32. Lee DD, & Seung HS (1999). Learning the parts of objects by non-negative matrix factorization. Nature, 401(6755), 788–791. 10.1038/44565 [DOI] [PubMed] [Google Scholar]
  33. Levandowsky M, & Winter D (1971). Distance between sets. Nature, 234(5323), 34–35. 10.1038/234034a0 [DOI] [Google Scholar]
  34. Lin YY, Liu TL, & Fuh CS (2011). Multiple kernel learning for dimensionality reduction. IEEE Transactions on Pattern Analysis and Machine Intelligence, 33(6), 1147–1160. 10.1109/TPAMI.2010.183 [DOI] [PubMed] [Google Scholar]
  35. Lock EF, & Dunson DB (2013). Bayesian consensus clustering. Bioinformatics, 29(20), 2610–2616. 10.1093/bioinformatics/btt425 [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Lock EF, Hoadley KA, Marron JS, & Nobel AB (2013). Joint and individual variation explained (Jive) for integrated analysis of multiple data types. The Annals of Applied Statistics, 7(1), 523–542. 10.1214/12-AOAS597 [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. MacQueen J (1967, June). Some methods for classification and analysis of multivariate observations. Paper presented at Proceedings of the fifth Berkeley symposium on mathematical statistics and probability (Vol. 1, No. 14, pp. 281–297). [Google Scholar]
  38. Mansell G, Gorrie-Stone TJ, Bao Y, Kumari M, Schalkwyk LS, Mill J, & Hannon E (2019). Guidance for DNA methylation studies: Statistical insights from the Illumina EPIC array. BMC Genomics, 20(1), 366. 10.1186/s12864-019-5761-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Meilă M (2003). Comparing clusterings by the variation of information. In Learning theory and kernel machines (pp. 173–187). Berlin, Heidelberg: Springer. [Google Scholar]
  40. Meng C, Helm D, Frejno M, & Kuster B (2016). moCluster: Identifying joint patterns across multiple Omics data sets. Journal of Proteome Research, 15(3), 755–765. 10.1021/acs.jproteome.5b00824 [DOI] [PubMed] [Google Scholar]
  41. Mo Q, Shen R, Guo C, Vannucci M, Chan KS, & Hilsenbeck SG (2018). A fully Bayesian latent variable model for integrative clustering analysis of multi-type omics data. Biostatistics, 19(1), 71–86. 10.1093/biostatistics/kxx017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Mo Q, Wang S, Seshan VE, Olshen AB, Schultz N, Sander C, … Shen R (2013). Pattern discovery and cancer gene identification in integrated cancer genomic data. Proceedings of the National Academy of Sciences of the United States of America, 110(11), 4245–4250. 10.1073/pnas.1208949110 [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Monti S, Tamayo P, Mesirov J, & Golub O (2003). Consensus clustering: A resampling-based method for class discovery and visualization of gene expression microarray data. Machine Learning, 52(1/2), 91–118. 10.1023/a:1023949509487 [DOI] [Google Scholar]
  44. Ng AY, Jordan MI, & Weiss Y (2002). On spectral clustering: Analysis and an algorithm. Advances in neural information processing systems, 2, 849–856. [Google Scholar]
  45. Nguyen H, Shrestha S, Draghici S, & Nguyen T (2019). PINSPlus: A tool for tumor subtype discovery in integrated genomic data. Bioinformatics, 35(16), 2843–2846. 10.1093/bioinformatics/bty1049 [DOI] [PubMed] [Google Scholar]
  46. Nguyen T, Tagett R, Diaz D, & Draghici S (2017). A novel approach for data integration and disease subtyping. Genome Research, 27 (12), 2025–2039. 10.1101/gr.215129.116 [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. O’Connell MJ, & Lock EF (2016). R.JIVE for exploration of multi-source molecular data. Bioinformatics, 32(18), 2877–2879. 10.1093/bioinformatics/btw324 [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Ogata H, Goto S, Sato K, Fujibuchi W, Bono H, & Kanehisa M (1999). KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Research, 27(1), 29–34. 10.1093/nar/27.1.29 [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Padilha VA, & Campello RJ (2017). A systematic comparative evaluation of biclustering techniques. BMC Bioinformatics, 18(1), 55. 10.1186/s12859-017-1487-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Pierre-Jean M, Deleuze JF, Le Floch E, & Mauger F (2019). Clustering and variable selection evaluation of 13 unsupervised methods for multi-omics data integration. Briefings in Bioinformatics, 21, 2011–2030. 10.1093/bib/bbz138 [DOI] [PubMed] [Google Scholar]
  51. Ramazzotti D, Lal A, Wang B, Batzoglou S, & Sidow A (2018). Multi-omic tumor data reveal diversity of molecular mechanisms that correlate with survival. Nature Communications, 9(1), 4453. 10.1038/s41467-018-06921-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Rand WM (1971). Objective criteria for the evaluation of clustering methods. Journal of the American Statistical Association, 66(336), 846–850. 10.1080/01621459.1971.10482356 [DOI] [Google Scholar]
  53. Rappoport N, & Shamir R (2018). Multi-omic and multi-view clustering algorithms: Review and cancer benchmark. Nucleic Acids Research, 46(20), 10546–10562. 10.1093/nar/gky889 [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Rappoport N, & Shamir R (2019). NEMO: Cancer subtyping by integration of partial multi-omic data. Bioinformatics, 35(18), 3348–3356. 10.1093/bioinformatics/btz058 [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Rasmussen CE (1999, December). The infinite Gaussian mixture model. Paper presented at NIPS (Vol. 12, pp. 554–560). [Google Scholar]
  56. Ruan P, Wang Y, Shen R, & Wang S (2019). Using association signal annotations to boost similarity network fusion. Bioinformatics, 35 (19), 3718–3726. 10.1093/bioinformatics/btz124 [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Shen R, Olshen AB, & Ladanyi M (2009). Integrative clustering of multiple genomic data types using a joint latent variable model with application to breast and lung cancer subtype analysis. Bioinformatics, 25(22), 2906–2912. 10.1093/bioinformatics/btp543 [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Shu L, & Latecki LJ (2016, February). Integration of single-view graphs with diffusion of tensor product graphs for multi-view spectral clustering. Paper presented at Asian Conference on Machine Learning (pp. 362–377). PMLR [Google Scholar]
  59. Speicher NK, & Pfeifer N (2015). Integrating different data types by regularized unsupervised multiple kernel learning with application to cancer subtype discovery. Bioinformatics, 31(12), i268–i275. 10.1093/bioinformatics/btv244 [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Stark R, Grzelak M, & Hadfield J (2019). RNA sequencing: The teenage years. Nature Reviews Genetics, 20(11), 631–656. 10.1038/s41576-019-0150-2 [DOI] [PubMed] [Google Scholar]
  61. Sun YV, & Hu YJ (2016). Integrative analysis of multi-omics data for discovery and functional studies of complex human diseases. Advances in Genetics, 93, 147–190. 10.1016/bs.adgen.2015.11.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Tari L, Baral C, & Kim S (2009). Fuzzy c-means clustering with prior biological knowledge. Journal of Biomedical Informatics, 42(1), 74–81. 10.1016/j.jbi.2008.05.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Tini G, Marchetti L, Priami C, & Scott-Boyer MP (2019). Multi-omics integration - A comparison of unsupervised clustering methodologies. Briefings in Bioinformatics, 20(4), 1269–1279. 10.1093/bib/bbx167 [DOI] [PubMed] [Google Scholar]
  64. Tong D, Tian Y, Zhou T, Ye Q, Li J, Ding K, & Li J (2020). Improving prediction performance of colon cancer prognosis based on the integration of clinical and multi-omics data. BMC Medical Informatics and Decision Making, 20(1), 22. 10.1186/s12911-020-1043-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Vaske CJ, Benz SC, Sanborn JZ, Earl D, Szeto C, Zhu J, … Stuart JM (2010). Inference of patient-specific pathway activities from multi-dimensional cancer genomics data using PARADIGM. Bioinformatics, 26(12), i237–i245. 10.1093/bioinformatics/btq182 [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Verbanck M, Lê S, & Pagès J (2013). A new unsupervised gene clustering algorithm based on the integration of biological knowledge into expression data. BMC Bioinformatics, 14(1), 1–11. 10.1186/1471-2105-14-42 [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. von Luxburg U (2007). A tutorial on spectral clustering. Statistics and Computing, 17(4), 395–416. 10.1007/s11222-007-9033-z [DOI] [Google Scholar]
  68. Wang B, Mezlini AM, Demir F, Fiume M, Tu Z, Brudno M, … Goldenberg A (2014). Similarity network fusion for aggregating data types on a genomic scale. Nature Methods, 11(3), 333–337. 10.1038/nmeth.2810 [DOI] [PubMed] [Google Scholar]
  69. Wang B, Zhu J, Pierson E, Ramazzotti D, & Batzoglou S (2017). Visualization and analysis of single-cell RNA-seq data by kernel-based similarity learning. Nature Methods, 14(4), 414–416. 10.1038/nmeth.4207 [DOI] [PubMed] [Google Scholar]
  70. Wang D, & Gu J (2016). Integrative clustering methods of multi-omics data for molecule-based cancer classifications. Quantitative Biology, 4(1), 58–67. 10.1007/s40484-016-0063-4 [DOI] [Google Scholar]
  71. Witten DM, & Tibshirani R (2010). A framework for feature selection in clustering. Journal of the American Statistical Association, 105 (490), 713–726. 10.1198/jasa.2010.tm09415 [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Witten DM, & Tibshirani RJ (2009). Extensions of sparse canonical correlation analysis with applications to genomic data. Statistical Applications in Genetics and Molecular Biology, 8(1), 1–27. 10.2202/1544-6115.1470 [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Wold S, Sjöström M, & Eriksson L (2001). PLS-regression: A basic tool of chemometrics. Chemometrics and Intelligent Laboratory Systems, 58(2), 109–130. 10.1016/s0169-7439(01)00155-1 [DOI] [Google Scholar]
  74. Wu D, Wang D, Zhang MQ, & Gu J (2015). Fast dimension reduction and integrative clustering of multi-omics data using low-rank approximation: Application to cancer molecular classification. BMC Genomics, 16, 1022. 10.1186/s12864-015-2223-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Yan J, Risacher SL, Shen L, & Saykin AJ (2018). Network approaches to systems biology analysis of complex disease: Integrative methods for multi-omics data. Briefings in Bioinformatics, 19(6), 1370–1381. 10.1093/bib/bbx066 [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Yan S, Xu D, Zhang B, Zhang H. j., Yang Q, & Lin S (2007). Graph embedding and extensions: A general framework for dimensionality reduction. IEEE Transactions on Pattern Analysis and Machine Intelligence, 29(1), 40–51. 10.1109/tpami.2007.250598 [DOI] [PubMed] [Google Scholar]
  77. Yang Z, & Michailidis G (2016). A non-negative matrix factorization method for detecting modules in heterogeneous omics multi-modal data. Bioinformatics, 32(1), 1–8. 10.1093/bioinformatics/btv544 [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Yuan Y, Savage RS, & Markowetz F (2011). Patient-specific data fusion defines prognostic cancer subtypes. PLoS Computational Biology, 7(10), e1002227. 10.1371/journal.pcbi.1002227 [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Zelnik-Manor L, & Perona P (2005). Self-tuning spectral clustering. Paper presented at the Advances in Neural Information Processing Systems. https://resolver.caltech.edu/CaltechAUTHORS:20160314-152424746 [Google Scholar]
  80. Zhang S, Liu C-C, Li W, Shen H, Laird PW, & Zhou XJ (2012). Discovery of multi-dimensional modules by integrative analysis of cancer genomic data. Nucleic Acids Research, 40(19), 9379–9391. 10.1093/nar/gks725 [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Zhang X, Li J, & Yu H (2011). Local density adaptive similarity measurement for spectral clustering. Pattern Recognition Letters, 32(2), 352–358. 10.1016/j.patrec.2010.09.014 [DOI] [Google Scholar]
  82. Zhou H, Pan W, & Shen X (2009). Penalized model-based clustering with unconstrained covariance matrices. Electronic Journal of Statistics, 3, 1473–1496. 10.1214/09-ejs487 [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES