Skip to main content

EDITORIAL article

Front. Oncol., 03 March 2023
Sec. Breast Cancer
This article is part of the Research Topic Cancer Stem Cells as Attractive Targets for Breast Cancer Therapy View all 5 articles

Editorial: Cancer stem cells as attractive targets for breast cancer therapy

  • 1Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt
  • 2Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
  • 3Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany

Breast cancer is the most prevalent cancer that affects females worldwide. It accounts for approximately 31% of cancers in women (1). Breast cancer can be classified into various intrinsic subtypes based on the expression of estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2), and the proliferative index Ki-67: luminal A (ER-positive, PR-positive, HER2-negative, Ki-67 with less than 20% positivity); luminal B-HER2- negative (ER-positive, HER2- negative, and either PR-low or Ki-67 with more than 20% positivity); luminal B HER2-positive (ER-positive or PR-positive, HER2-positive or amplified, and regardless of Ki-67 status); HER2-enriched (ER-negative, PR-negative, HER-2-positive or amplified, and regardless of Ki-67 status); and triple-negative (ER-negative, PR-negative, and HER2-negative) breast cancer (24). Despite recent advances in breast carcinoma treatment, including chemotherapy, radiotherapy, and hormone (endocrine) therapy, as well as innovations in targeted therapeutics and immunotherapy (5), development of treatment resistance and tumor recurrence are still major challenges. In contrast to differentiated cells, cancer stem cells (CSCs) or tumor initiating cells (TICs), which constitute a subpopulation of cells within the tumor bulk, have been linked to tumor relapse and resistance to conventional therapy (69). Therefore, targeting CSCs has emerged as a promising therapeutic strategy against cancer, including breast cancer. CSCs or TICs possess unique features, including self-renewal, expression of multidrug resistance proteins, high proliferative capacity, efficient DNA repair capability, resistance to apoptosis, and overexpression of therapeutic efflux proteins (1015). Breast CSCs are characterized by expression of cell surface makers CD44+/CD24- and/or increased detoxifying aldehyde dehydrogenase (ALDH) activity (6, 16). Breast CSCs are influenced not only by cancer-cell autonomous factors but also by diverse cues derived from the tumor microenvironment (TME) (17).

This Research Topic collection comprises four papers (two original research, one perspective, and one review article) coauthored by 28 researchers. We aimed to provide original research and opinion pieces on breast CSCs, focusing on new molecular markers and therapeutic targets, crosstalk between tumor microenvironmental cells and breast CSCs, and signaling pathways involved in breast CSC regulation.

Discovering new molecular markers expressed by breast CSCs is of particular interest for breast cancer management. Yang et al. showed that two splicing variants of very-low-density lipoprotein receptor (VLDLR-I, II), a member of the low-density lipoprotein receptor (LDLR) superfamily involved in lipid metabolism, are overexpressed in breast CSCs relative to non-breast CSCs. Using gain-and loss-function experiments, the authors further verified the biological role of VLDR-I and -II expression in regulating breast CSC functions in vitro and in vivo, namely xenograft tumor growth, tumor cell proliferation, and spheroid and colony formation. Suppression of breast tumor growth is mediated by VLDR knockdown-induced cellular quiescence in a ligand-independent manner. Increased expression of VLDR is correlated with elevated energy production and ribosome biogenesis, as well as associated with poor clinical outcome in breast cancer patients.

Breast CSCs express many immunosuppressive molecules (e.g., the immune checkpoint ligands-programmed death-ligand (PD-L)1 and PD-L2), which impede the functions of natural killer and T- immune cells and result in promoting immune evasion and stemness. Ruiu et al. addressed in their perspective article the immunological properties of breast CSCs and questioned the suitability of their exploitation as targets for immunotherapeutics (e.g., vaccines, adoptive cell therapy and chimeric antigen receptor (CAR)-T cells, monoclonal antibodies, and small molecules) based on preclinical studies and clinical trials. The authors reported encouraging results of some clinical trials. However, the applicability of immunotherapies to target breast CSCs is still under study and has not been proven yet.

Inflammatory breast cancer (IBC), the deadliest and highly metastatic form of breast cancer, is enriched with CSCs in comparison with non-inflammatory types of breast cancer. Tumor-associated macrophages (TAMs), an important component of IBC TME, play an essential role in IBC pathogenesis and regulate breast CSCs. Macrophages infected with the human cytomegalovirus (HCMV) act as “mobile vectors” for virus dissemination into breast tissues. Mohamed et al. demonstrated that the secretome (containing IL-6, IL-8, and MCP-1) of TAMs infected with HCMV induced IBC SUM149 cells to undergo proliferation, invasion, colony formation and breast CSC-related gene expression via activation of STAT3, AMPKα, PRAS40, and SAPK/JNK compared to untreated SUM149 cells.

It is well known that breast CSCs are regulated by development-associated signaling pathways, including Wnt, Notch, and Hedgehog pathways. Walker et al. discussed and provided evidence for the role of prostaglandin E2 pathway in breast CSCs using published transcriptome data. The authors also addressed the relevance of targeting the prostaglandin E2 pathway for breast cancer treatment, especially for CSCs-enriched tumor subtypes.

Overall, the publications within this Research Topic collection provide further support for the breast CSC concept and provide novel avenues of targeting a key tumor cell population associated with therapeutic resistance and recurrence.

Author contributions

All authors made substantial contribution to the work and approved it for publication.

Acknowledgments

Guest editors would like to thank all researchers for their valuable contributions to this Research Topic collection.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA. Cancer J Clin (2023) 73:17–48. doi: 10.3322/caac.21763

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Perou CM, Sørlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, et al. Molecular portraits of human breast tumours. Nature (2000) 406:747–52. doi: 10.1038/35021093

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Sørlie T, Tibshirani R, Parker J, Hastie T, Marron JS, Nobel A, et al. Repeated observation of breast tumor subtypes in independent gene expression data sets. Proc Natl Acad Sci (2003) 100:8418–23. doi: 10.1073/pnas.0932692100

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Li X, Zhou J, Xiao M, Zhao L, Zhao Y, Wang S, et al. Uncovering the subtype-specific molecular characteristics of breast cancer by multiomics analysis of prognosis-associated genes, driver genes, signaling pathways, and immune activity. Front Cell Dev Biol (2021) 9:689028. doi: 10.3389/fcell.2021.689028

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Sun X, Liu K, Lu S, He W, Du Z. Targeted therapy and immunotherapy for heterogeneous breast cancer. Cancers (Basel). (2022) 14:5456. doi: 10.3390/cancers14215456

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Ibrahim SA, Gadalla R, El-Ghonaimy EA, Samir O, Mohamed HT, Hassan H, et al. Syndecan-1 is a novel molecular marker for triple negative inflammatory breast cancer and modulates the cancer stem cell phenotype via the IL-6/STAT3, notch and EGFR signaling pathways. Mol Cancer (2017) 16:57. doi: 10.1186/s12943-017-0621-z

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Ibrahim SA, Hassan H, Vilardo L, Kumar SK, Kumar AV, Kelsch R, et al. Syndecan-1 (CD138) modulates triple-negative breast cancer stem cell properties via regulation of LRP-6 and IL-6-Mediated STAT3 signaling. PloS One (2013) 8:e85737. doi: 10.1371/journal.pone.0085737

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Kumar Katakam S, Tria V, Sim W, Yip GW, Molgora S, Karnavas T, et al. The heparan sulfate proteoglycan syndecan-1 regulates colon cancer stem cell function via a focal adhesion kinase–wnt signaling axis. FEBS J (2021) 288:486–506. doi: 10.1111/febs.15356

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Troschel FM, Minte A, Ismail YM, Kamal A, Abdullah MS, Ahmed SH, et al. Knockdown of musashi RNA binding proteins decreases radioresistance but enhances cell motility and invasion in triple-negative breast cancer. Int J Mol Sci (2020) 21:2169. doi: 10.3390/ijms21062169

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Vitale D, Kumar Katakam S, Greve B, Jang B, Oh E, Alaniz L, et al. Proteoglycans and glycosaminoglycans as regulators of cancer stem cell function and therapeutic resistance. FEBS J (2019) 286:2870–82. doi: 10.1111/febs.14967

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Greve B, Kelsch R, Spaniol K, Eich HT, Götte M. Flow cytometry in cancer stem cell analysis and separation. Cytom. Part A (2012) 81(4):284–93. doi: 10.1002/cyto.a.22022

CrossRef Full Text | Google Scholar

12. Saha T, Lukong KE. Breast cancer stem-like cells in drug resistance: A review of mechanisms and novel therapeutic strategies to overcome drug resistance. Front Oncol (2022) 12:856974. doi: 10.3389/fonc.2022.856974

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci (2003) 100:3983–8. doi: 10.1073/pnas.0530291100

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Zheng Q, Zhang M, Zhou F, Zhang L, Meng X. The breast cancer stem cells traits and drug resistance. Front Pharmacol (2021) 11:599965. doi: 10.3389/fphar.2020.599965

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Teixeira FCOB, Vijaya Kumar A, Kumar Katakam S, Cocola C, Pelucchi P, Graf M, et al. The heparan sulfate sulfotransferases HS2ST1 and HS3ST2 are novel regulators of breast cancer stem-cell properties. Front Cell Dev Biol (2020) 8:559554. doi: 10.3389/fcell.2020.559554

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Ginestier C, Hur MH, Charafe-Jauffret E, Monville F, Dutcher J, Brown M, et al. ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell (2007) 1:555–67. doi: 10.1016/j.stem.2007.08.014

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Ciummo SL, D’Antonio L, Sorrentino C, Fieni C, Lanuti P, Stassi G, et al. The c-X-C motif chemokine ligand 1 sustains breast cancer stem cell self-renewal and promotes tumor progression and immune escape programs. Front Cell Dev Biol (2021) 9:689286. doi: 10.3389/fcell.2021.689286

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: cancer stem cells, cancer initiating cells, breast cancer, metastasis, epithelial-mesenchymal transition, tumor microenvironment, immunotherapy

Citation: Ibrahim SA, Yip GW and Götte M (2023) Editorial: Cancer stem cells as attractive targets for breast cancer therapy. Front. Oncol. 13:1151742. doi: 10.3389/fonc.2023.1151742

Received: 26 January 2023; Accepted: 27 February 2023;
Published: 03 March 2023.

Edited and Reviewed by:

Kara Britt, Peter MacCallum Cancer Centre, Australia

Copyright © 2023 Ibrahim, Yip and Götte. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Sherif Abdelaziz Ibrahim, isherif@cu.edu.eg; isherif@sci.cu.edu.eg; Martin Götte, mgotte@uni-muenster.de

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.