Sequencing of breast cancer stem cell populations indicates a dynamic conversion between differentiation states in vivo

IntroductionThe cancer stem cell model implies a hierarchical organization within breast tumors maintained by cancer stem-like cells (CSCs). Accordingly, CSCs are a subpopulation of cancer cells with capacity for self-renewal, differentiation and tumor initiation. These cells can be isolated through the phenotypic markers CD44+/CD24-, expression of ALDH1 and an ability to form nonadherent, multicellular spheres in vitro. However, controversies to describe the stem cell model exist; it is unclear whether the tumorigenicity of CSCs in vivo is solely a proxy for a certain genotype. Moreover, in vivo evidence is lacking to fully define the reversibility of CSC differentiation.MethodsIn order to answer these questions, we undertook exome sequencing of CSCs from 12 breast cancer patients, along with paired primary tumor samples. As suggested by stem classical cell biology, we assumed that the number of mutations in the CSC subpopulation should be lower and distinct compared to the differentiated tumor cells with higher proliferation.ResultsOur analysis revealed that the majority of somatic mutations are shared between CSCs and bulk primary tumor, with similar frequencies in the two.ConclusionsThe data presented here exclude the possibility that CSCs are only a phenotypic consequence of certain somatic mutations, that is a distinct and non-reversible population of cells. In addition, our results imply that CSCs must be a population of cells that can dynamically switch from differentiated tumor cells, and vice versa. This finding increases our understanding of CSC function in tumor heterogeneity and the importance of identifying drugs to counter de-differentiation rather than targeting CSCs.

[1]  T. Taguchi,et al.  Stem cell marker aldehyde dehydrogenase 1‐positive breast cancers are characterized by negative estrogen receptor, positive human epidermal growth factor receptor type 2, and high Ki67 expression , 2009, Cancer science.

[2]  I. Weissman,et al.  Clonal Evolution of Preleukemic Hematopoietic Stem Cells Precedes Human Acute Myeloid Leukemia , 2012, Science Translational Medicine.

[3]  Corbin E. Meacham,et al.  Tumour heterogeneity and cancer cell plasticity , 2013, Nature.

[4]  Guy S. Salvesen,et al.  SnapShot: Caspases , 2011, Cell.

[5]  L. Dodd,et al.  Intratumoral heterogeneity in primary breast carcinoma: Study of concurrent parameters , 1997, Journal of surgical oncology.

[6]  Mohamed H. Sayegh,et al.  Identification of cells initiating human melanomas , 2008, Nature.

[7]  C. Blanpain,et al.  Unravelling cancer stem cell potential , 2013, Nature Reviews Cancer.

[8]  Kornelia Polyak,et al.  Heterogeneity for Stem Cell–Related Markers According to Tumor Subtype and Histologic Stage in Breast Cancer , 2010, Clinical Cancer Research.

[9]  I. Weissman,et al.  Stem cells, cancer, and cancer stem cells , 2001, Nature.

[10]  Aleix Prat Aparicio Comprehensive molecular portraits of human breast tumours , 2012 .

[11]  Irmtraud M. Meyer,et al.  The clonal and mutational evolution spectrum of primary triple-negative breast cancers , 2012, Nature.

[12]  S. Morrison,et al.  Prospective identification of tumorigenic breast cancer cells , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[13]  G. Dontu,et al.  In vitro propagation and transcriptional profiling of human mammary stem/progenitor cells. , 2003, Genes & development.

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

[15]  Steven J. M. Jones,et al.  Comprehensive molecular portraits of human breast tumours , 2013 .

[16]  M. Gönen,et al.  Cellular and genetic diversity in the progression of in situ human breast carcinomas to an invasive phenotype. , 2010, The Journal of clinical investigation.

[17]  R. Datar,et al.  Genetic and epigenetic analysis of putative breast cancer stem cell models , 2013, BMC Cancer.

[18]  J. Dick,et al.  A human colon cancer cell capable of initiating tumour growth in immunodeficient mice , 2007, Nature.

[19]  Markus Thomas,et al.  Established breast cancer stem cell markers do not correlate with in vivo tumorigenicity of tumor-initiating cells , 2012, International journal of oncology.

[20]  Joshua F. McMichael,et al.  The Origin and Evolution of Mutations in Acute Myeloid Leukemia , 2012, Cell.

[21]  R. Beroukhim,et al.  Molecular definition of breast tumor heterogeneity. , 2007, Cancer cell.

[22]  Paul A. Wiggins,et al.  Normal and neoplastic nonstem cells can spontaneously convert to a stem-like state , 2011, Proceedings of the National Academy of Sciences.

[23]  J. Dick,et al.  Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell , 1997, Nature Medicine.

[24]  J. Troge,et al.  Tumour evolution inferred by single-cell sequencing , 2011, Nature.

[25]  Susan G Hilsenbeck,et al.  Intrinsic resistance of tumorigenic breast cancer cells to chemotherapy. , 2008, Journal of the National Cancer Institute.

[26]  J. Salk Clonal evolution in cancer , 2010 .