Advertisement
Original article| Volume 22, ISSUE 6, P521-537, August 2022

Identification of Breast Cancer Subtypes Based on Gene Expression Profiles in Breast Cancer Stroma

  • Md. Nazim Uddin
    Affiliations
    Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China

    Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China

    Big Data Research Institute, China Pharmaceutical University, Nanjing, China

    Institute of Food Science and Technology, Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka, Bangladesh
    Search for articles by this author
  • Xiaosheng Wang
    Correspondence
    Address for correspondence: Xiaosheng Wang, MD, PhD, Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
    Affiliations
    Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China

    Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China

    Big Data Research Institute, China Pharmaceutical University, Nanjing, China
    Search for articles by this author
Published:April 04, 2022DOI:https://doi.org/10.1016/j.clbc.2022.04.001

      Abstract

      Background

      Tumor stroma is a heterogeneous cellular component in the tumor microenvironment of breast cancer. However, very few studies have explored the identification of breast cancer subtypes based on highly heterogeneous tumor stromal signatures.

      Materials and Methods

      Using a combined dataset composed of 8 gene expression profiling datasets for breast tumor stroma, we clustered breast cancers based on the expression levels of 100 genes whose expression values were most variable across all samples. Furthermore, we investigated the molecular features of the breast cancer subtypes identified.

      Results

      We identified 2 breast cancer subtypes, termed SBCS-1 and SBCS-2. We found that the contents of stroma and immune cells were lower in SBCS-1 than in SBCS-2, while the proportion of tumor cells was higher in SBCS-1. SBCS-1 was enriched in cancer-associated pathways, including ribosomes, cell cycle, RNA degradation, RNA polymerase, DNA replication, oxidative phosphorylation, proteasome, spliceosome, and glycolysis/gluconeogenesis. SBCS-2 was enriched in pathways of graft versus host disease, type 1 diabetes mellitus, intestinal immune network for IgA production, allograft rejection, and steroid hormone biosynthesis. Moreover, many oncogenic biological processes were highly activated in SBCS-1, including proliferation, stemness, epithelial-to-mesenchymal transition (EMT), and angiogenesis. Gene co-expression network analysis identified prognostic hub genes, transcription factor encoding genes (PFDN5 and EZH2), and protein kinase encoding gene (AURKA) in a gene module highly enriched in SBCS-1.

      Conclusion

      Based on the gene expression profiles in breast cancer stroma, breast cancer can be divided into 2 subtypes, which have significantly different molecular, and clinical characteristics. The identification of new subtypes of breast cancer has clinical implications for the management of this disease.

      Keywords

      Abbreviations:

      SBCS-1 (stromal breast cancer subtype 1), SBCS-2 (stromal breast cancer subtype 2), TME (tumor microenvironment), NCBI (National Center for Biotechnology Information), TCGA (The Cancer Genome Atlas), GDC (Genomic Data Commons), RSEM (RNA-Seq by expectation-maximization), ssGSEA (single-sample gene-set enrichment analysis), TILs (tumor-infiltrating lymphocytes), GSEA (gene set enrichment analysis), EMT (epithelial-mesenchymal transition), WGCNA (weighted correlation network analysis), KEGG (Kyoto Encyclopedia of Genes and Genomes), TF (transcription factor), RFS (recurrence-free survival)
      To read this article in full you will need to make a payment

      Purchase one-time access:

      Academic & Personal: 24 hour online accessCorporate R&D Professionals: 24 hour online access
      One-time access price info
      • For academic or personal research use, select 'Academic and Personal'
      • For corporate R&D use, select 'Corporate R&D Professionals'

      Subscribe:

      Subscribe to Clinical Breast Cancer
      Already a print subscriber? Claim online access
      Already an online subscriber? Sign in
      Institutional Access: Sign in to ScienceDirect

      References

        • Valkenburg KC
        • de Groot AE
        • Pienta KJ.
        Targeting the tumour stroma to improve cancer therapy.
        Nat Rev Clin Oncol. 2018; 15: 366-381https://doi.org/10.1038/s41571-018-0007-1
        • Novin A
        • Suhail Y
        • Ajeti V
        • et al.
        Diversity in cancer invasion phenotypes indicates specific stroma regulated programs.
        Human Cell. 2021; 34: 111-121https://doi.org/10.1007/s13577-020-00427-6
        • Uddin MN
        • Wang X.
        The landscape of long non-coding RNAs in tumor stroma.
        Life Sci. 2021; 264118725https://doi.org/10.1016/j.lfs.2020.118725
        • Tarin D.
        Role of the host stroma in cancer and its therapeutic significance.
        Cancer Metastasis Rev. 2013; 32: 553-566https://doi.org/10.1007/s10555-013-9438-4
        • Merlino G
        • Miodini P
        • Callari M
        • D'Aiuto F
        • Cappelletti V
        • Daidone MG
        Prognostic and functional role of subtype-specific tumor–stroma interaction in breast cancer.
        Mol Oncol. 2017; 11: 1399-1412https://doi.org/10.1002/1878-0261.12107
        • Conklin MW
        • Keely PJ.
        Why the stroma matters in breast cancer.
        Cell Adh Migr. 2012; 6: 249-260https://doi.org/10.4161/cam.20567
        • van der Spek YM
        • Kroep JR
        • Tollenaar RAEM
        • Mesker WE.
        Chemotherapy resistance and stromal targets in breast cancer treatment: a review.
        Mol Biol Rep. 2020; 47: 8169-8177https://doi.org/10.1007/s11033-020-05853-1
        • Plava J
        • Cihova M
        • Burikova M
        • Matuskova M
        • Kucerova L
        • Miklikova S.
        Recent advances in understanding tumor stroma-mediated chemoresistance in breast cancer.
        Mol Cancer. 2019; 18: 67https://doi.org/10.1186/s12943-019-0960-z
        • Hofmeister V
        • Schrama D
        • Becker JC.
        Anti-cancer therapies targeting the tumor stroma.
        Cancer Immunol Immunother. 2008; 57: 1-17https://doi.org/10.1007/s00262-007-0365-5
        • Hofmeister V
        • Vetter C
        • Schrama D
        • Bröcker EB
        • Becker JC.
        Tumor stroma-associated antigens for anti-cancer immunotherapy.
        Cancer Immunol Immunother. 2006; 55: 481-494https://doi.org/10.1007/s00262-005-0070-1
        • Kraman M
        • Bambrough PJ
        • Arnold JN
        • et al.
        Suppression of antitumor immunity by stromal cells expressing fibroblast activation protein-alpha.
        Science. 2010; 330: 827-830https://doi.org/10.1126/science.1195300
        • Ren Q
        • Zhu P
        • Zhang H
        • et al.
        Identification and validation of stromal-tumor microenvironment-based subtypes tightly associated with PD-1/PD-L1 immunotherapy and outcomes in patients with gastric cancer.
        Cancer Cell Int. 2020; 20: 92https://doi.org/10.1186/s12935-020-01173-3
        • Puré E
        • Lo A
        Can targeting stroma pave the way to enhanced antitumor immunity and immunotherapy of solid tumors?.
        Cancer Immunol Res. 2016; 4: 269-278https://doi.org/10.1158/2326-6066.CIR-16-0011
        • Yuan J
        • Liu M
        • Yang L
        • et al.
        Acquisition of epithelial-mesenchymal transition phenotype in the tamoxifen-resistant breast cancer cell: a new role for G protein-coupled estrogen receptor in mediating tamoxifen resistance through cancer-associated fibroblast-derived fibronectin and β1-integrin signaling pathway in tumor cells.
        Breast Cancer Res. 2015; 17: 69https://doi.org/10.1186/s13058-015-0579-y
        • Shiga K
        • Hara M
        • Nagasaki T
        • Sato T
        • Takahashi H
        • Takeyama H.
        Cancer-associated fibroblasts: their characteristics and their roles in tumor growth.
        Cancers. 2015; 7: 2443-2458https://doi.org/10.3390/cancers7040902
      1. IL-6 secreted by cancer-associated fibroblasts induces tamoxifen resistance in luminal breast cancer.
        Oncogene. 2014; 33: 4450https://doi.org/10.1038/onc.2014.224
        • Junttila MR
        • de Sauvage FJ.
        Influence of tumour micro-environment heterogeneity on therapeutic response.
        Nature. 2013; 501: 346-354https://doi.org/10.1038/nature12626
        • Yang SX
        • Polley EC.
        Systemic treatment and radiotherapy, breast cancer subtypes, and survival after long-term clinical follow-up.
        Breast Cancer Res Treat. 2019; 175: 287-295https://doi.org/10.1007/s10549-019-05142-x
        • Bonsang-Kitzis H
        • Sadacca B
        • Hamy-Petit AS
        • et al.
        Biological network-driven gene selection identifies a stromal immune module as a key determinant of triple-negative breast carcinoma prognosis.
        Oncoimmunology. 2016; 5e1061176https://doi.org/10.1080/2162402X.2015.1061176
        • Hamy AS
        • Bonsang-Kitzis H
        • Lae M
        • et al.
        A stromal immune module correlated with the response to neoadjuvant chemotherapy, prognosis and lymphocyte infiltration in her2-positive breast carcinoma is inversely correlated with hormonal pathways.
        PLoS One. 2016; 11e0167397https://doi.org/10.1371/journal.pone.0167397
        • Finak G
        • Bertos N
        • Pepin F
        • et al.
        Stromal gene expression predicts clinical outcome in breast cancer.
        Nat Med. 2008; 14: 518-527https://doi.org/10.1038/nm1764
        • Winslow S
        • Leandersson K
        • Edsjö A
        • Larsson C.
        Prognostic stromal gene signatures in breast cancer.
        Breast Cancer Res. 2015; 17https://doi.org/10.1186/s13058-015-0530-2
        • Sims AH
        • Howell A
        • Howell SJ
        • Clarke RB.
        Origins of breast cancer subtypes and therapeutic implications.
        Nat Clin Pract Oncol. 2007; 4: 516-525https://doi.org/10.1038/ncponc0908
        • Liu H
        • Dowdle JA
        • Khurshid S
        • et al.
        Discovery of stromal regulatory networks that suppress ras-sensitized epithelial cell proliferation.
        Dev Cell. 2017; 41: 392-407.e6https://doi.org/10.1016/j.devcel.2017.04.024
        • Harvell DME
        • Kim J
        • O'Brien J
        • et al.
        Genomic signatures of pregnancy-associated breast cancer epithelia and stroma and their regulation by estrogens and progesterone.
        Horm Cancer. 2013; 4: 140-153https://doi.org/10.1007/s12672-013-0136-z
        • Planche A
        • Bacac M
        • Provero P
        • et al.
        Identification of prognostic molecular features in the reactive stroma of human breast and prostate cancer.
        PLoS One. 2011; 6: e18640https://doi.org/10.1371/journal.pone.0018640
        • Casey T
        • Bond J
        • Tighe S
        • et al.
        Molecular signatures suggest a major role for stromal cells in development of invasive breast cancer.
        Breast Cancer Res Treat. 2009; 114: 47-62https://doi.org/10.1007/s10549-008-9982-8
        • Karnoub AE
        • Dash AB
        • Vo AP
        • et al.
        Mesenchymal stem cells within tumour stroma promote breast cancer metastasis.
        Nature. 2007; 449: 557-563https://doi.org/10.1038/nature06188
        • Knudsen ES
        • Ertel A
        • Davicioni E
        • Kline J
        • Schwartz GF
        • Witkiewicz AK.
        Progression of ductal carcinoma in situ to invasive breast cancer is associated with gene expression programs of EMT and myoepithelia.
        Breast Cancer Res Treat. 2012; 133: 1009-1024https://doi.org/10.1007/s10549-011-1894-3
        • Ma XJ
        • Dahiya S
        • Richardson E
        • Erlander M
        • Sgroi DC.
        Gene expression profiling of the tumor microenvironment during breast cancer progression.
        Breast Cancer Res. 2009; 11: R7https://doi.org/10.1186/bcr2222
        • Xia J
        • Gill EE
        • Hancock REW.
        NetworkAnalyst for statistical, visual and network-based meta-analysis of gene expression data.
        Nat Protoc. 2015; 10: 823-844https://doi.org/10.1038/nprot.2015.052
        • Johnson WE
        • Li C
        • Rabinovic A.
        Adjusting batch effects in microarray expression data using empirical Bayes methods.
        Biostatistics. 2007; 8: 118-127https://doi.org/10.1093/biostatistics/kxj037
        • Koboldt DC
        • Fulton RS
        • McLellan MD
        • et al.
        Comprehensive molecular portraits of human breast tumours.
        Nature. 2012; 490: 61-70https://doi.org/10.1038/nature11412
        • Loi S
        • Haibe-Kains B
        • Desmedt C
        • et al.
        Predicting prognosis using molecular profiling in estrogen receptor-positive breast cancer treated with tamoxifen.
        BMC Genomics. 2008; 9: 239https://doi.org/10.1186/1471-2164-9-239
        • Loi S
        • Haibe-Kains B
        • Majjaj S
        • et al.
        PIK3CA mutations associated with gene signature of low mTORC1 signaling and better outcomes in estrogen receptor-positive breast cancer.
        Proc Natl Acad Sci U S A. 2010; 107: 10208-10213https://doi.org/10.1073/pnas.0907011107
        • Yoshihara K
        • Shahmoradgoli M
        • Martínez E
        • et al.
        Inferring tumour purity and stromal and immune cell admixture from expression data.
        Nat Commun. 2013; 4: 2612https://doi.org/10.1038/ncomms3612
        • Aran D
        • Hu Z
        • Butte AJ.
        xCell: digitally portraying the tissue cellular heterogeneity landscape.
        Genome Biol. 2017; 18https://doi.org/10.1186/s13059-017-1349-1
        • Hänzelmann S
        • Castelo R
        • Guinney J.
        GSVA: gene set variation analysis for microarray and RNA-seq data.
        BMC Bioinformatics. 2013; 14: 7https://doi.org/10.1186/1471-2105-14-7
        • Subramanian A
        • Tamayo P
        • Mootha VK
        • et al.
        Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles.
        Proc Natl Acad Sci U S A. 2005; 102: 15545-15550https://doi.org/10.1073/pnas.0506580102
        • Mootha VK
        • Lindgren CM
        • Eriksson KF
        • et al.
        PGC-1alpha-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes.
        Nat Genet. 2003; 34: 267-273https://doi.org/10.1038/ng1180
        • Yu G
        • Wang LG
        • Han Y
        • He QY.
        clusterProfiler: an R package for comparing biological themes among gene clusters.
        OMICS. 2012; 16: 284-287https://doi.org/10.1089/omi.2011.0118
        • Kanehisa M
        • Furumichi M
        • Tanabe M
        • Sato Y
        • Morishima K.
        KEGG: new perspectives on genomes, pathways, diseases and drugs.
        Nucleic Acids Res. 2017; 45: D353-D361https://doi.org/10.1093/nar/gkw1092
        • He Y
        • Jiang Z
        • Chen C
        • Wang X.
        Classification of triple-negative breast cancers based on Immunogenomic profiling.
        J Exp Clin Cancer Res. 2018; 37https://doi.org/10.1186/s13046-018-1002-1
        • Miranda A
        • Hamilton PT
        • Zhang AW
        • et al.
        Cancer stemness, intratumoral heterogeneity, and immune response across cancers.
        Proc Natl Acad Sci U S A. 2019; 116: 9020-9029https://doi.org/10.1073/pnas.1818210116
        • Langfelder P
        • Horvath S.
        WGCNA: an R package for weighted correlation network analysis.
        BMC Bioinformatics. 2008; 9: 559https://doi.org/10.1186/1471-2105-9-559
        • Szklarczyk D
        • Gable AL
        • Lyon D
        • et al.
        STRING v11: protein–protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets.
        Nucleic Acids Res. 2019; 47: D607-D613https://doi.org/10.1093/nar/gky1131
        • Chin CH
        • Chen SH
        • Wu HH
        • Ho CW
        • Ko MT
        • Lin CY.
        cytoHubba: identifying hub objects and sub-networks from complex interactome.
        BMC Syst Biol. 2014; 8: S11https://doi.org/10.1186/1752-0509-8-S4-S11
        • Shannon P
        • Markiel A
        • Ozier O
        • et al.
        Cytoscape: a software environment for integrated models of biomolecular interaction networks.
        Genome Res. 2003; 13: 2498-2504https://doi.org/10.1101/gr.1239303
      2. Therneau T. A package for survival analysis in R. :95.

        • Mizuno H
        • Kitada K
        • Nakai K
        • Sarai A.
        PrognoScan: a new database for meta-analysis of the prognostic value of genes.
        BMC Med Genomics. 2009; 2: 18https://doi.org/10.1186/1755-8794-2-18
        • Ritchie ME
        • Phipson B
        • Wu D
        • et al.
        limma powers differential expression analyses for RNA-sequencing and microarray studies.
        Nucleic Acids Res. 2015; 43: e47https://doi.org/10.1093/nar/gkv007
        • Bindea G
        • Mlecnik B
        • Tosolini M
        • et al.
        Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer.
        Immunity. 2013; 39: 782-795https://doi.org/10.1016/j.immuni.2013.10.003
        • Yearley JH
        • Gibson C
        • Yu N
        • et al.
        PD-L2 expression in human tumors: relevance to anti-PD-1 therapy in cancer.
        Clin Cancer Res. 2017; 23: 3158-3167https://doi.org/10.1158/1078-0432.CCR-16-1761
        • Rowshanravan B
        • Halliday N
        • Sansom DM.
        CTLA-4: a moving target in immunotherapy.
        Blood. 2018; 131: 58-67https://doi.org/10.1182/blood-2017-06-741033
        • Choi J
        • Kim DH
        • Jung WH
        • Koo JS.
        Metabolic interaction between cancer cells and stromal cells according to breast cancer molecular subtype.
        Breast Cancer Res. 2013; 15: R78https://doi.org/10.1186/bcr3472
        • Singh S
        • Chakrabarti R.
        Consequences of EMT-driven changes in the immune microenvironment of breast cancer and therapeutic response of cancer cells.
        J Clin Med. 2019; 8https://doi.org/10.3390/jcm8050642
        • Pinto MP
        • Dye WW
        • Jacobsen BM
        • Horwitz KB.
        Malignant stroma increases luminal breast cancer cell proliferation and angiogenesis through platelet-derived growth factor signaling.
        BMC Cancer. 2014; 14https://doi.org/10.1186/1471-2407-14-735
        • Dolezal JM
        • Dash AP
        • Prochownik EV.
        Diagnostic and prognostic implications of ribosomal protein transcript expression patterns in human cancers.
        BMC Cancer. 2018; 18: 275https://doi.org/10.1186/s12885-018-4178-z
        • Yoshida T
        • Kitaura H
        • Hagio Y
        • Sato T
        • Iguchi-Ariga SMM
        • Ariga H.
        Negative regulation of the Wnt signal by MM-1 through inhibiting expression of the wnt4 gene.
        Exp Cell Res. 2008; 314: 1217-1228https://doi.org/10.1016/j.yexcr.2008.01.002
        • Hirukawa A
        • Smith HW
        • Zuo D
        • et al.
        Targeting EZH2 reactivates a breast cancer subtype-specific anti-metastatic transcriptional program.
        Nat Commun. 2018; 9https://doi.org/10.1038/s41467-018-04864-8
        • Sahai E
        • Astsaturov I
        • Cukierman E
        • et al.
        A framework for advancing our understanding of cancer-associated fibroblasts.
        Nat Rev Cancer. 2020; 20: 174-186https://doi.org/10.1038/s41568-019-0238-1
        • Parker JS
        • Mullins M
        • Cheang MCU
        • et al.
        Supervised risk predictor of breast cancer based on intrinsic subtypes.
        J Clin Oncol. 2009; 27: 1160-1167https://doi.org/10.1200/JCO.2008.18.1370
        • Yao J
        • Li S
        • Wang X.
        Identification of breast cancer immune subtypes by analyzing bulk tumor and single cell transcriptomes.
        Front Cell Dev Biol. 2022; 9 (Accessed at: March 11, 2022Accessed from:)