Single-cell RNA-seq enables comprehensive tumour and immune cell profiling in primary breast cancer

Single-cell transcriptome profiling of tumour tissue isolates allows the characterization of heterogeneous tumour cells along with neighbouring stromal and immune cells. Here we adopt this powerful approach to breast cancer and analyse 515 cells from 11 patients. Inferred copy number variations from the single-cell RNA-seq data separate carcinoma cells from non-cancer cells. At a single-cell resolution, carcinoma cells display common signatures within the tumour as well as intratumoral heterogeneity regarding breast cancer subtype and crucial cancer-related pathways. Most of the non-cancer cells are immune cells, with three distinct clusters of T lymphocytes, B lymphocytes and macrophages. T lymphocytes and macrophages both display immunosuppressive characteristics: T cells with a regulatory or an exhausted phenotype and macrophages with an M2 phenotype. These results illustrate that the breast cancer transcriptome has a wide range of intratumoral heterogeneity, which is shaped by the tumour cells and immune cells in the surrounding microenvironment.

[1]  R. Sutherland,et al.  c-Myc or Cyclin D1 Mimics Estrogen Effects on Cyclin E-Cdk2 Activation and Cell Cycle Reentry , 1998, Molecular and Cellular Biology.

[2]  Ash A. Alizadeh,et al.  Genome-wide analysis of DNA copy-number changes using cDNA microarrays , 1999, Nature Genetics.

[3]  C K Osborne,et al.  Estrogen receptor status by immunohistochemistry is superior to the ligand-binding assay for predicting response to adjuvant endocrine therapy in breast cancer. , 1999, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[4]  Carsten O. Peterson,et al.  Estrogen receptor status in breast cancer is associated with remarkably distinct gene expression patterns. , 2001, Cancer research.

[5]  Andrea Califano,et al.  Transcriptional analysis of the B cell germinal center reaction , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[6]  Pablo Tamayo,et al.  Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[7]  Alberto Mantovani,et al.  Transcriptional Profiling of the Human Monocyte-to-Macrophage Differentiation and Polarization: New Molecules and Patterns of Gene Expression1 , 2006, The Journal of Immunology.

[8]  C. Liu,et al.  Targeting tumor-associated macrophages as a novel strategy against breast cancer. , 2006, The Journal of clinical investigation.

[9]  Z. Trajanoski,et al.  Type, Density, and Location of Immune Cells Within Human Colorectal Tumors Predict Clinical Outcome , 2006, Science.

[10]  M. Probst-Kepper,et al.  Signatures of human regulatory T cells: an encounter with old friends and new players , 2006, Genome Biology.

[11]  Gianluca Bontempi,et al.  Biological Processes Associated with Breast Cancer Clinical Outcome Depend on the Molecular Subtypes , 2008, Clinical Cancer Research.

[12]  C. Sotiriou,et al.  Meta-analysis of gene expression profiles in breast cancer: toward a unified understanding of breast cancer subtyping and prognosis signatures , 2007, Breast Cancer Research.

[13]  L. V. van't Veer,et al.  Clinical application of the 70-gene profile: the MINDACT trial. , 2008, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[14]  S. Paik,et al.  Development of the 21-gene assay and its application in clinical practice and clinical trials. , 2008, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[15]  F. Pépin,et al.  Stromal gene expression predicts clinical outcome in breast cancer , 2008, Nature Medicine.

[16]  Thomas D. Wu,et al.  Genetic Alterations and Oncogenic Pathways Associated with Breast Cancer Subtypes , 2009, Molecular Cancer Research.

[17]  Richard Durbin,et al.  Sequence analysis Fast and accurate short read alignment with Burrows – Wheeler transform , 2009 .

[18]  Renaud Gaujoux,et al.  A flexible R package for nonnegative matrix factorization , 2010, BMC Bioinformatics.

[19]  I. Ellis,et al.  Triple-Negative Breast Cancer: Distinguishing between Basal and Nonbasal Subtypes , 2009, Clinical Cancer Research.

[20]  Peter Olson,et al.  Cancer-Associated Fibroblasts Are Activated in Incipient Neoplasia to Orchestrate Tumor-Promoting Inflammation in an NF-kappaB-Dependent Manner. , 2010, Cancer cell.

[21]  M. DePristo,et al.  A framework for variation discovery and genotyping using next-generation DNA sequencing data , 2011, Nature Genetics.

[22]  J. Baselga,et al.  Targeted therapies for breast cancer. , 2011, The Journal of clinical investigation.

[23]  Rachel Schiff,et al.  Mechanisms of endocrine resistance in breast cancer. , 2011, Annual review of medicine.

[24]  X. Chen,et al.  Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. , 2011, The Journal of clinical investigation.

[25]  I. Ellis,et al.  The prognostic significance of B lymphocytes in invasive carcinoma of the breast , 2012, Breast Cancer Research and Treatment.

[26]  Colin N. Dewey,et al.  RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome , 2011, BMC Bioinformatics.

[27]  Justin Guinney,et al.  GSVA: gene set variation analysis for microarray and RNA-Seq data , 2013, BMC Bioinformatics.

[28]  Xi Chen,et al.  TNBCtype: A Subtyping Tool for Triple-Negative Breast Cancer , 2012, Cancer informatics.

[29]  Tatiana Popova,et al.  Supplementary Methods , 2012, Acta Neuropsychiatrica.

[30]  F. Markowetz,et al.  The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups , 2012, Nature.

[31]  Chris Williams,et al.  RNA-SeQC: RNA-seq metrics for quality control and process optimization , 2012, Bioinform..

[32]  John Quackenbush,et al.  A three-gene model to robustly identify breast cancer molecular subtypes. , 2012, Journal of the National Cancer Institute.

[33]  A. Sivachenko,et al.  Sequence analysis of mutations and translocations across breast cancer subtypes , 2012, Nature.

[34]  Steven J. M. Jones,et al.  Comprehensive molecular portraits of human breast tumors , 2012, Nature.

[35]  G. Getz,et al.  Inferring tumour purity and stromal and immune cell admixture from expression data , 2013, Nature Communications.

[36]  R. Clarke,et al.  Recent advances reveal IL-8 signaling as a potential key to targeting breast cancer stem cells , 2013, Breast Cancer Research.

[37]  Z. Trajanoski,et al.  Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. , 2013, Immunity.

[38]  Thomas R. Gingeras,et al.  STAR: ultrafast universal RNA-seq aligner , 2013, Bioinform..

[39]  H. Schreiber,et al.  Innate and adaptive immune cells in the tumor microenvironment , 2013, Nature Immunology.

[40]  A. Sivachenko,et al.  Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples , 2013, Nature Biotechnology.

[41]  N. Navin,et al.  Clonal Evolution in Breast Cancer Revealed by Single Nucleus Genome Sequencing , 2014, Nature.

[42]  J. Hackney,et al.  The immunoreceptor TIGIT regulates antitumor and antiviral CD8(+) T cell effector function. , 2014, Cancer cell.

[43]  C. Sotiriou,et al.  Transfer of clinically relevant gene expression signatures in breast cancer: from Affymetrix microarray to Illumina RNA-Sequencing technology , 2014, BMC Genomics.

[44]  Jeffrey W Pollard,et al.  Tumor-associated macrophages: from mechanisms to therapy. , 2014, Immunity.

[45]  L. Nguyen,et al.  Clinical blockade of PD1 and LAG3 — potential mechanisms of action , 2014, Nature Reviews Immunology.

[46]  N. Neff,et al.  Quantitative assessment of single-cell RNA-sequencing methods , 2013, Nature Methods.

[47]  Shawn M. Gillespie,et al.  Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma , 2014, Science.

[48]  Do-Hyun Nam,et al.  Single-cell mRNA sequencing identifies subclonal heterogeneity in anti-cancer drug responses of lung adenocarcinoma cells , 2015, Genome Biology.

[49]  B. Zhu,et al.  T-cell exhaustion in the tumor microenvironment , 2015, Cell Death and Disease.

[50]  N. Navin,et al.  The first five years of single-cell cancer genomics and beyond , 2015, Genome research.

[51]  Evan Z. Macosko,et al.  Highly Parallel Genome-wide Expression Profiling of Individual Cells Using Nanoliter Droplets , 2015, Cell.

[52]  Anne Vincent-Salomon,et al.  Intra-tumor genetic heterogeneity and alternative driver genetic alterations in breast cancers with heterogeneous HER2 gene amplification , 2015, Genome Biology.

[53]  J. Wolchok,et al.  Immune Checkpoint Blockade in Cancer Therapy. , 2015, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[54]  Daniel E. Speiser,et al.  Inhibitory Receptors Beyond T Cell Exhaustion , 2015, Front. Immunol..

[55]  Chih-Yang Wang,et al.  Single-cell analysis reveals a stem-cell program in human metastatic breast cancer cells , 2015, Nature.

[56]  Kun Zhang,et al.  Fluorescent in situ sequencing (FISSEQ) of RNA for gene expression profiling in intact cells and tissues , 2015, Nature Protocols.

[57]  T. Nielsen,et al.  The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: recommendations by an International TILs Working Group 2014. , 2015, Annals of oncology : official journal of the European Society for Medical Oncology.

[58]  N. Navin Delineating cancer evolution with single-cell sequencing , 2015, Science Translational Medicine.

[59]  J. Rosenblatt,et al.  B cell regulation of the anti-tumor response and role in carcinogenesis , 2016, Journal of Immunotherapy for Cancer.

[60]  Charles H. Yoon,et al.  Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq , 2016, Science.