Skip to main content
  • Research Note
  • Open access
  • Published:

A comparative study of clustering methods on gene expression data for lung cancer prognosis

Abstract

Lung cancer subtyping based on gene expression data is important for identifying patient subgroups with differing survival prognosis to facilitate customized treatment strategies for each subtype of patients. Unsupervised clustering methods are the traditional approach for clustering patients into subtypes. However, since those methods cluster patients based only on gene expression data, the resulting clusters may not always be relevant to the survival outcome of interest. In recent years, semi-supervised and supervised methods have been proposed, which leverage the survival outcome data to identify clusters more relevant to survival prognosis. This paper aims to compare the performance of different clustering methods for identifying clinically prognostic lung cancer subtypes based on two lung adenocarcinoma datasets. For each method, we clustered patients into two clusters and assessed the difference in patient survival time between clusters. Unsupervised methods were found to have large logrank p-values and no significant results in most cases. Semi-supervised and supervised methods had improved performance over unsupervised methods and very significant p-values. These results indicate that unsupervised methods are not capable of identifying clusters with significant differences in survival prognosis in most cases, while supervised and semi-supervised methods can better cluster patients into clinically useful subtypes.

Peer Review reports

Introduction

The leading cause of cancer death for both men and women is lung cancer [1]. However, risk levels vary between patients, so it is important to identify clinically prognostic subtypes of lung cancer with different risk levels and target higher risk subtypes for more aggressive treatment [2, 3]. Gene expression data may be used for this screening, as expression levels of certain genes are associated with cancer prognosis [4]. In addition to lung cancer, molecular subtypes have been widely investigated in many other cancer types, such as breast cancer [5] and colorectal cancer [6]. We refer the readers to [7,8,9,10] for comprehensive reviews of cancer subtyping.

Traditionally, there have been various unsupervised clustering algorithms used for this purpose. Unsupervised clustering methods treat all features equally, without considering their significance to the clinical outcome. As data typically contain multiple underlying structures, e.g. a set of documents may be clustered based on authorship, topic, or writing style [11], the results of unsupervised clustering can be driven by one dominant structure, or a mixture of several structures. There is no guarantee that the structures identified by unsupervised clustering is relevant to the outcome of interest [12]. In cancer subtyping studies, it has been shown that the subtypes identified by unsupervised clustering may sometimes be related to cell-of-origin [13] or histology [14] rather than patient’s survival time. Therefore, although subtypes from unsupervised clustering may be useful for investigating the biology of cancer, they are not always associated with clinical prognosis [12]. To combat this, semi-supervised methods, which attempt to consider both expression and clinical data, have been proposed [15]. These methods select genes that are more closely related to clinical outcome for clustering input [15]. Recently, supervised clustering methods have also been proposed, such as survClust [12], ogClust [16], and [17]. These methods directly incorporate clinical outcome information into the clustering process to ensure that the resulting clusters are clinically relevant.

In this paper, we compare the performance of these three types of clustering algorithms for identifying prognostic subtypes in lung cancer, providing a guidance on future applications of these approaches. The two datasets used are from patients with lung adenocarcinoma, one of the most common types of lung cancer [18]. We will apply each clustering method to identify subtypes and evaluate the performance based on the prognostic difference between subtypes.

Methods

Data

Two datasets were considered. The first dataset used was the lung adenocarcinoma dataset from The Cancer Genome Atlas Research Network (TCGA-LUAD) [18], which included patient gene expression data (transcript per million (TPM) values, 56,716 features), as well as clinical data, such as smoking history, cancer stage, patient survival time, patient censoring event (patient status at the time of leaving the study), patient age, and patient sex. The second dataset was from a study conducted by Shedden et al. [19], which included similar data variables, with fewer gene expression features (normalized microarray data with 22,283 features). Patients without both clinical and gene expression data were excluded. Furthermore, duplicate entries for patients were excluded, along with patients with survival time of 0. After applying a 2-component principal component analysis (PCA), outliers were found and removed. All gene expression data were scaled from 0 to 1 using min-max scaling. In the TCGA-LUAD dataset, 483 patients remained after data preparation, while in the Shedden dataset, 346 patients remained. A summary of patient characteristics is provided in Supplementary Table 1.

Clustering methods

The clustering methods tested were unsupervised methods, semi-supervised methods, and outcome-guided methods. For unsupervised methods, k-means (KM) [20], Gaussian mixture (GM), and agglomerative clustering (AC) were considered. Implementations of KM, GM, and AC models were provided by Scikit-learn [21]. In addition, we also included a Consensus clustering method based on a voting consensus of KM, GM, and AC clustering results, which follows the idea of [22]. Specifically, the method with the best agreement with the other two was selected as the reference, with the other two methods’ results adjusted to this reference and a best-of-3 voting deciding the final cluster of each patient. Note that our approach to select the reference method differed from that in [22] because we did not leverage any clinical information in order to make the method unsupervised. Strategies to include the clinical outcome variable in combination with Consensus clustering or other unsupervised methods to enable semi-supervised learning are described in the next paragraph. Prior to clustering analysis, a PCA was performed to reduce the dimension of features. The PCA considered gene expression data, as well as data on sex and age. The top principal components (PCs) sufficient to explain 95% of the variance in the data along with two important clinical variables, i.e. cancer stage and smoking status, were used as features in the clustering analysis.

For semi-supervised methods, feature selection methods were first used to select features, and these selected features were then used by clustering methods to cluster patients. Specifically, feature selection methods included Cox proportional-hazards regression [23] and random survival forests (RF) [24]. Implementation of Cox regression was provided in Python by lifelines [25], and the implementation of RF was provided in R [26]. Each feature selection model used patient survival time as the outcome variable and considered the same set of features as unsupervised methods. Based on each feature selection method, 20 features were selected for consideration in clustering. For the Cox regression, each feature was fitted univariately to the survival outcome, and the features with the lowest p-values were selected. For RF, the top 20 variables selected in a minimal-depth search were used. Based on the feature-selected data, unsupervised clustering methods, including KM, GM, AC, and Consensus, were applied. We considered the following eight combinations of feature selection and clustering methods for each dataset (TCGA or Shedden): Cox-KM, Cox-GM, Cox-AC, Cox-Consensus, RF-KM, RF-GM, RF-AC, and RF-Consensus.

For supervised methods, survClust (SC) [12] was considered. Patient survival time was used as the target value, and the same set of features as unsupervised methods were considered. survClust was trained using cross-validation, with final clustering results based on the consensus after 10 rounds of 3-fold cross-validation, as suggested by the authors [12].

All methods were set to cluster into 2 clusters. The number of clusters was selected to evaluate the ability of the methods to separate patients into good and bad prognostic subtypes. Method parameters that were changed from defaults are provided in Supplementary Table 2. Each method was run 200 times and averaged results were reported to reduce variation in performance due to random seeds, etc. Note that survClust had 200 trials of the 10-round 3-fold cross-validation run.

Evaluation

To evaluate the performance of the methods, Kaplan-Meier survival curves [27] were plotted to visualize the difference in survival time distribution between the two clusters predicted by a clustering method.

Utilizing the survival curves, p-values based on logrank tests were calculated to characterize the performance of the methods. A lower p-value indicates a better-performing method. A significance threshold of p = 0.05 was set to indicate the best performing methods. Since all methods were run 200 times, the average p-value is used to represent the performance of a method. Furthermore, Adjusted Rand Index (ARI) [28] was used to assess the similarity between the clusters of two methods. Specifically, each of the 200 replicates from one method was compared to each of the 200 replicates from the other method to obtain an ARI value. The averaged ARI of those 40,000 values was reported.

Results

We applied unsupervised, semi-supervised, and supervised clustering methods to the TCGA-LUAD [18] and Shedden [19] datasets to investigate which methods could identify clinically prognostic subtypes. Based on each method, we identified two clusters, and compared the survival time distributions of the two clusters of patients. Table 1; Fig. 1 summarize the overall results for every method, with mean, standard deviation, and 95% CI based on 200 trials. Supplementary Figs. 1 and 2 display the Kaplan-Meier survival curves of the two clusters based on a representative trial of each method for TCGA-LUAD and Shedden data, respectively.

Table 1 Overall results of different clustering methods
Fig. 1
figure 1

Comparison of p-values from different clustering methods. Note: These forest plots display -log10 p-value for comparing survival distribution between the two clusters identified by each clustering method based on (A) TCGA-LUAD or (B) Shedden data. The methods are grouped based on clustering algorithms, with unsupervised methods on top, semi-supervised methods in the center, and survClust (SC), the supervised method, on the bottom. Semi-supervised methods and SC present the best performance of all the methods. The solid square indicates -log10(mean p-value) and horizonal line indicates the corresponding 95% CI over 200 trials for each method. The significance threshold of p = 0.05 is marked with a vertical line, with significant values to the right of the line

Unsupervised methods presented non-significant differences (p-value > 0.05) in patient survival time between clusters in most cases, except for the AC and Consensus methods on TCGA-LUAD data and the KM method on Shedden data (Table 1; Fig. 1). The Kaplan-Meier survival curves for those non-significant cases also did not present strong separation between clusters (Supplementary Figs. 1 and 2). Therefore, the unsupervised clustering methods failed to identify clinically meaningful prognostic subtypes in most cases.

As for semi-supervised methods, both random forest- and Cox Regression-selected models yielded highly significant differences in survival time between clusters for both datasets (Table 1; Fig. 1). The corresponding Kaplan-Meier survival curves also showed strong separation between clusters (Supplementary Figs. 1 and 2).

The supervised clustering survClust method also presented a highly significant difference between clusters for both datasets (Table 1; Fig. 1). It had a strong separation between clusters, which are clearly distinguishable in the survival curves (Supplementary Figs. 1 and 2).

To assess the consistency in clustering results across different methods, we calculated the ARI for each pair of methods. As shown in Fig. 2, the clustering results from semi-supervised and supervised methods are highly consistent with each other, and are dissimilar to the results from unsupervised methods (except for AC with TCGA-LUAD data). Therefore, semi-supervised and supervised methods were able to obtain subtypes distinct from those obtained from unsupervised methods.

Fig. 2
figure 2

Evaluation of the consistency of clustering results from different clustering methods. Note: The heatmaps present the ARI for each pair of clustering methods based on (A) TCGA-LUAD and (B) Shedden data. The results present unsupervised methods first, followed by semi-supervised and supervised

The analyses above performed clustering using PCs, which is a frequently used approach to reduce the dimension of features but may introduce biases. To make a more unbiased evaluation, we performed a separate set of analyses using all the original features instead of PCs based on TCGA-LUAD data. Results are provided in Supplementary Tables 3 and Supplementary Figs. 35. Due to the large number of original features, survClust was unable to generate clustering results. Unsupervised methods and semi-supervised methods with random forest for feature selection also yielded less significant results. Specifically, all p-values from unsupervised methods were greater than 0.05, and only the p-value from RF-GM was smaller than 0.05. Semi-supervised methods with Cox regression for feature selection appeared to be less affected by the switch from PCs to original features, with all p-values remaining highly significant.

Discussion

Our study showed that unsupervised clustering methods, regardless of the specific clustering algorithm, were unable to identify lung cancer subtypes with different prognosis in many cases. In contrast, semi-supervised and supervised clustering methods were able to identify subtypes with significant difference in patient survival time. Therefore, when the purpose of clustering is to identify prognostic subtypes, supervised and semi-supervised methods would be preferred.

The supervised method, survClust, is unable to converge with large amounts of features, and some measure of feature reduction, such as PCA or a feature selection algorithm, may be required before clustering. It is also more computationally expensive than the other methods tested. Both supervised and semi-supervised methods require the outcome of patients to cluster, so unsupervised methods may be required in a study without outcomes already available, although the clinical interpretation of the clustering results can be ambiguous.

Another item of note is that PCA can improve clustering results. Based on our analysis of TCGA-LUAD data, the analyses based on PCs (Table 1; Fig. 1) yielded smaller p-values than the analyses based on all the original features (Supplementary Tables 3 and Supplementary Fig. 4) for all unsupervised and semi-supervised methods. This is likely due to the reduction in noise that PCA provides.

Limitations

Our study focuses on gene expression features for the identification of lung cancer prognostic subtypes. It has been well-documented that other molecular features, such as single nucleotide variants, copy number variations, and fusions, are also associated with lung cancer prognosis [10, 28]. Future studies that include those features will provide a more comprehensive assessment and comparison of the performance of different clustering methods. In addition, simulation studies, where there are known “true” clusters, may also need to be conducted to further elucidate the strength and weakness of different clustering methods.

Data Availability

The TCGA-LUAD dataset analyzed during the current study is available in the Genomic Data Commons of the National Cancer Institute, https://gdc.cancer.gov. The Shedden et al. dataset is available in the Gene Expression Omnibus of the National Cancer for Biotechnology Information, https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE68465.

Abbreviations

TCGA-LUAD:

The Cancer Genome Atlas Research Network lung adenocarcinoma dataset

TPM:

Transcript per million

PCA:

Principal component analysis

KM:

k-means

GM:

Gaussian mixture

AC:

Agglomerative clustering

PC:

Principal component

RF:

random survival forests

SC:

survClust

ARI:

Adjusted Rand Index

CI:

Confidence interval

References

  1. Spiro SG, Silvestri GA. One hundred years of Lung cancer. Am J Respir Crit Care Med. 2005;172(5):523–9.

    Article  PubMed  Google Scholar 

  2. Hayes DN, Monti S, Parmigiani G, Gilks CB, Naoki K, Bhattacharjee A, et al. Gene expression profiling reveals reproducible human lung adenocarcinoma subtypes in multiple Independent patient cohorts. J Clin Oncol. 2006;24(31):5079–90.

    Article  CAS  PubMed  Google Scholar 

  3. Bhattacharjee A, Richards WG, Staunton J, Li C, Monti S, Vasa P et al. Classification of human lung carcinomas by mRNA expression profiling reveals distinct adenocarcinoma subclasses. Proceedings of the National Academy of Sciences. 2001;98(24):13790-5.

  4. Lu Y, Lemon W, Liu P-Y, Yi Y, Morrison C, Yang P, et al. A gene expression signature predicts survival of patients with stage I non-small cell Lung cancer. PLoS Med. 2006;3(12):e467.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Perou CM, Sørlie T, Eisen MB, Van De Rijn M, Jeffrey SS, Rees CA, et al. Molecular portraits of human breast tumours. Nature. 2000;406(6797):747–52.

    Article  CAS  PubMed  Google Scholar 

  6. De Sousa E, Melo F, Wang X, Jansen M, Fessler E, Trinh A, De Rooij LP, et al. Poor-prognosis colon Cancer is defined by a molecularly distinct subtype and develops from serrated precursor lesions. Nat Med. 2013;19(5):614–8.

    Article  Google Scholar 

  7. Zhao L, Lee VH, Ng MK, Yan H, Bijlsma MF. Molecular subtyping of cancer: current status and moving toward clinical applications. Brief Bioinform. 2019;20(2):572–84.

    Article  CAS  PubMed  Google Scholar 

  8. Wang C, Zhang H, Liu Y, Wang Y, Hu H, Wang G. Molecular subtyping in Colorectal cancer: a bridge to personalized therapy. Oncol Lett. 2023;25(6):1–13.

    Article  Google Scholar 

  9. Chia N-Y, Tan P. Molecular classification of gastric cancer. Ann Oncol. 2016;27(5):763–9.

    Article  PubMed  Google Scholar 

  10. Inamura K. Lung cancer: understanding its molecular pathology and the 2015 WHO classification. Front Oncol. 2017;7:193.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Xing E, Jordan M, Russell SJ, Ng A. Distance metric learning with application to clustering with side-information. Adv Neural Inf Process Syst. 2002;15.

  12. Arora A, Olshen AB, Seshan VE, Shen R. Pan-cancer identification of clinically relevant genomic subtypes using outcome-weighted integrative clustering. Genome Med. 2020;12:1–13.

    Article  Google Scholar 

  13. Hoadley KA, Yau C, Hinoue T, Wolf DM, Lazar AJ, Drill E, et al. Cell-of-origin patterns dominate the molecular classification of 10,000 tumors from 33 types of cancer. Cell. 2018;173(2):291–304. e6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Network CGAR. Integrated genomic characterization of oesophageal carcinoma. Nature. 2017;541(7636):169.

    Article  Google Scholar 

  15. Bair E, Tibshirani R. Semi-supervised methods to predict patient survival from gene expression data. PLoS Biol. 2004;2(4):e108.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Liu P, Fang Y, Ren Z, Tang L, Tseng GC. Outcome-Guided Disease Subtyping for High-Dimensional Omics Data. arXiv preprint arXiv:200711123. 2020.

  17. Chang W, Wan C, Zang Y, Zhang C, Cao S. Supervised clustering of high-dimensional data using regularized mixture modeling. Brief Bioinform. 2021;22(4):bbaa291.

    Article  PubMed  Google Scholar 

  18. Network CGAR. Comprehensive molecular profiling of lung adenocarcinoma. Nature. 2014;511(7511):543.

    Article  Google Scholar 

  19. Gene expression–. Based survival prediction in lung adenocarcinoma: a multi-site, blinded validation study. Nat Med. 2008;14(8):822–7.

    Article  Google Scholar 

  20. MacQueen J. In:. Some methods for classification and analysis of multivariate observations. Oakland, CA, USA: Proceedings of the fifth Berkeley symposium on mathematical statistics and probability; 1967.

    Google Scholar 

  21. Pedregosa F, Varoquaux G, Gramfort A, Michel V, Thirion B, Grisel O, et al. Scikit-learn: machine learning in Python. J Mach Learn Res. 2011;12:2825–30.

    Google Scholar 

  22. Manganaro L, Bianco S, Bironzo P, Cipollini F, Colombi D, Corà D, et al. Consensus clustering methodology to improve molecular stratification of non-small cell Lung cancer. Sci Rep. 2023;13(1):7759.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Harrell J, Frank E, Harrell FE. Cox proportional hazards regression model. Regression modeling strategies: With applications to linear models, logistic and ordinal regression, and survival analysis. 2015:475–519.

  24. Ishwaran H, Kogalur U, Blackstone E, Lauer M. Random Survival Forests. The annals of applied statistics. 2008; 2 (3): 841–60.

  25. Davidson-Pilon C. Lifelines: survival analysis in Python. J Open Source Softw. 2019;4(40):1317.

    Article  Google Scholar 

  26. Ishwaran H, Kogalur U. Package ‘RandomForestSRC’. Fast Unified Random Forests for Survival, Regression, and Classification (RF-SRC). Version 3.1. 1. 2022.

  27. Goel MK, Khanna P, Kishore J. Understanding survival analysis: Kaplan-Meier estimate. Int J Ayurveda Res. 2010;1(4):274.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Pender A, Popat S. Understanding Lung cancer molecular subtypes. Clin Pract. 2014;11(4):441.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This research was supported by National Institutes of Health (R03CA259670) and the Biostatistics and Bioinformatics Shared Resource Facilities of the University of Kentucky Markey Cancer Center (P30CA177558).

Author information

Authors and Affiliations

Authors

Contributions

JZ collected data from clustering methods and analyzed the data. CW provided guidance and assistance and was a major contributor in writing and reviewing the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Chi Wang.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

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

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, J.Z., Wang, C. A comparative study of clustering methods on gene expression data for lung cancer prognosis. BMC Res Notes 16, 319 (2023). https://doi.org/10.1186/s13104-023-06604-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13104-023-06604-8

Keywords