Cancer Letters

Cancer Letters

Volume 500, 1 March 2021, Pages 64-74
Cancer Letters

Cancer stem cells: Culprits in endocrine resistance and racial disparities in breast cancer outcomes

https://doi.org/10.1016/j.canlet.2020.12.014Get rights and content

Highlights

  • Endocrine therapy is highly efficacious though some patients experience relapse.

  • Breast cancer stem cells (BCSCs) contribute to endocrine resistance and relapse.

  • BCSCs likely play a causal role in racial disparities observed in breast cancer.

  • Targeting BCSCs is expected to improve luminal breast cancer patient outcomes.

Abstract

Breast cancer stem cells (BCSCs) promote endocrine therapy (ET) resistance, also known as endocrine resistance in hormone receptor (HR) positive breast cancer. Endocrine resistance occurs via mechanisms that are not yet fully understood. In vitro, in vivo and clinical data suggest that signaling cascades such as Notch, hypoxia inducible factor (HIF), and integrin/Akt promote BCSC-mediated endocrine resistance. Once HR positive breast cancer patients relapse on ET, targeted therapy agents such as cyclin dependent kinase inhibitors are frequently implemented, though secondary resistance remains a threat. Here, we discuss Notch, HIF, and integrin/Akt pathway regulation of BCSC activity and potential strategies to target these pathways to counteract endocrine resistance. We also discuss a plausible link between elevated BCSC-regulatory gene levels and reduced survival observed among African American women with basal-like breast cancer which lacks HR expression. Should future studies reveal a similar link for patients with luminal breast cancer, then the use of agents that impede BCSC activity could prove highly effective in improving clinical outcomes among African American breast cancer patients.

Introduction

More than 500,000 women die from breast cancer each year worldwide. Most women are diagnosed with hormone receptor (HR) positive or luminal breast cancer since their tumors express the estrogen receptor (ER), the progesterone receptor (PR), and in rarer instances, human epidermal growth factor receptor 2 (Her2). Luminal breast cancer is further divided into two subtypes: luminal A and luminal B, with the luminal A subtype exhibiting the best prognosis [1]. Endocrine therapy (ET) entails blocking the binding of estrogen to the ER often designated as estrogen receptor alpha (ERα), or inhibiting aromatase which converts androgens into estrogens.

Selective estrogen receptor modulator tamoxifen, acts as an estrogen antagonist in breast tissues yet behaves as an estrogen agonist in cardiac tissues and bone [2]. Aromatase inhibitors (AIs) have largely replaced tamoxifen as first line therapy for luminal breast cancer due to their enhanced efficacy [3]. Thus, tamoxifen is primarily used in women who cannot tolerate AIs. In pre-menopausal patients, AIs alone are insufficient to decrease estrogen levels due to the production of estrogen from the ovaries. It is therefore necessary to suppress ovarian function, which is commonly and reversibly achieved using luteinizing hormone-releasing hormone analogs. Patients who develop AI resistance are often treated with the selective estrogen receptor down-regulator fulvestrant in combination with agents such as cyclin dependent kinase (CDK) inhibitors [4].

Though ET often shows tremendous efficacy, relapse occurs in more than 40% of patients [5]. When patients develop resistance to one form of ET, they are often treated with an alternate form of ET. However, resistance to alternate forms of ET frequently occurs. Emerging evidence suggests that ET often enriches the breast cancer stem cell (BCSC) population to ultimately promote relapse [6] (Fig. 1A). Recently, AI-resistant cells were shown to exhibit high levels of stemness markers [7]. To eradicate BCSCs, it is necessary to identify cellular signaling pathways that promote BCSC development and function [8].

African American women experience higher rates of breast cancer mortality than European American women despite lower overall incidence [9]. Differences in tumor biology are believed to contribute to this survival disparity. This minireview will focus on molecular aberrations within breast cancer cells that promote stem cell survival and increase the risk of endocrine resistance onset. We will discuss the potential contribution of BCSCs to the breast cancer survival disparity observed among African American women. We will also discuss potential therapeutic strategies to overcome endocrine resistance based on preclinical and clinical studies.

Section snippets

Pathways that regulate BCSC activity in endocrine therapy resistance

Stem cells govern tissue development and homeostasis. BCSCs represent key drivers of metastasis and endocrine resistance as previously reviewed [10]. Signaling pathways that promote endocrine resistance frequently drive BCSC growth and function. For instance, an ERα splice variant promotes resistance by increasing ET-mediated enrichment of the BCSC population via activation of AKT/GSK3β signaling [11]. Notch and integrin/Akt signaling pathways are important players in stem cell function and

Race associated differences in breast cancer survival

While race is often defined as a social construct, disparities in breast cancer outcomes have been observed among certain racial and ethnic groups. Both genetic and epigenetic factors drive underlying tumor biology to suggest that designating ‘ancestry’ groups proves more accurate in a biological context than self-reported ‘race’. In most studies, the race/ethnicity designation given reflect the predominance toward the use of African ancestry in African Americans/Blacks as well as the use of

Clinical evaluation of agents designed to tackle endocrine resistance

Cyclin dependent kinase 4 (CDK4) promotes self-renewal within BCSCs in TNBC [112]. Whether CDK4 also promotes cancer stemness in luminal breast cancer is worth investigating since this may suggest why at least initially, CDK4/6 inhibitors enhance ET efficacy once patients relapse on ET. Insufficient elimination of BCSCs represents a key reason many combination therapies only marginally outperform established monotherapies.

A number of clinical trials have commenced to treat breast cancer

Perspectives and conclusions

ET benefits many patients with luminal breast cancer although the emergence of resistance leading to relapse is a constant threat to sustained efficacy. Targeting signaling pathways that promote stemness is important to improving breast cancer survival. Though BCSCs contribute substantially to aggressive breast cancer subtypes (e.g., TNBC) and appear to promote the survival disparity seen among African American breast cancer patients, we hypothesize that these cells may play an even greater

Funding

This work was supported in part by funds from the Department of Basic Sciences Loma Linda University Health (LLUH) School of Medicine, Department of Pharmaceutical and Administrative Sciences LLUH School of Pharmacy, the Grants for Research and School Partnerships (LLUH intramural grant), NIH/National Institute of General Medical Sciences grant (award number 2R25GM060507), PIP 0812 CONICET 2014–2016 (Argentina), the LLUH Center for Health Disparities and Molecular Medicine and the LLUH School

Author contributions

Nicole Mavingire: investigation, writing (original draft, review & editing) Petreena Campbell: investigation, writing (original draft, review & editing) Jonathan Wooten: investigation, writing (original draft, review & editing) Joyce Aja: investigation, writing (original draft, review & editing) Melissa Davis: conceptualization, investigation, writing (original draft, review & editing) Andrea Loaiza-Perez: conceptualization, investigation, writing (original draft, review & editing), funding

Declaration of competing interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgements

The authors thank Drs. Gayathri Nagaraj, In Jae Yoo, Willie Davis, Jr. and Ubaldo Soto for helpful discussions. The authors apologize for the publications that could not be cited due to space constraints. Fig. 1 was created with BioRender.com.

References (120)

  • M. Wormke

    Crosstalk between estrogen receptor alpha and the aryl hydrocarbon receptor in breast cancer cells involves unidirectional activation of proteasomes

    FEBS Lett.

    (2000)
  • S. Zhao

    Activation of the aryl hydrocarbon receptor represses mammosphere formation in MCF-7 cells

    Canc. Lett.

    (2012)
  • C. Massard

    First-in-human study of LY3039478, an oral Notch signaling inhibitor in advanced or metastatic cancer

    Ann. Oncol.

    (2018)
  • P. Alluri et al.

    Basal-like and triple negative breast cancers: searching for positives among many negatives

    (2014)
  • O. Najim

    The prevalence of estrogen receptor-1 mutation in advanced breast cancer: the estrogen receptor one study (EROS1)

    Canc. Treat. Res. Commun.

    (2019)
  • M.C. Cheang

    Ki67 index, HER2 status, and prognosis of patients with luminal B breast cancer

    J. Natl. Canc. Inst.

    (2009)
  • H. Michael

    Differential effects of selective oestrogen receptor modulators (SERMs) tamoxifen, ospemifene and raloxifene on human osteoclasts in vitro

    Br. J. Pharmacol.

    (2007)
  • S. Sammons et al.

    Fulvestrant-based combination therapy for second-line treatment of hormone receptor-positive advanced breast cancer

    Targeted Oncol.

    (2019)
  • H. Pan

    20-Year risks of breast-cancer recurrence after stopping endocrine therapy at 5 years

    N. Engl. J. Med.

    (2017)
  • K. Uchiumi

    Cancer stem-like properties of hormonal therapy-resistant breast cancer cells

    Breast Cancer

    (2019)
  • A. Acar

    A role for Notch signalling in breast cancer and endocrine resistance

    Stem Cell. Int.

    (2016)
  • G.H. Rauscher

    Racial disparity in survival from estrogen and progesterone receptor-positive breast cancer: implications for reducing breast cancer mortality disparities

    Breast Canc. Res. Treat.

    (2017)
  • D. Ojo

    Factors promoting tamoxifen resistance in breast cancer via stimulating breast cancer stem cell expansion

    Curr. Med. Chem.

    (2015)
  • V. Venkatesh

    Targeting Notch signalling pathway of cancer stem cells

    Stem Cell Invest.

    (2018)
  • D.L. Abravanel

    Notch promotes recurrence of dormant tumor cells following HER2/neu-targeted therapy

    J. Clin. Invest.

    (2015)
  • S. Mittal

    Cooperation of Notch and Ras/MAPK signaling pathways in human breast carcinogenesis

    Mol. Canc.

    (2009)
  • B. Cohen

    Cyclin D1 is a direct target of JAG1-mediated Notch signaling in breast cancer

    Breast Canc. Res. Treat.

    (2010)
  • N.E. Buckley

    BRCA1 is a key regulator of breast differentiation through activation of Notch signalling with implications for anti-endocrine treatment of breast cancers

    Nucleic Acids Res.

    (2013)
  • H. Harrison

    Regulation of breast cancer stem cell activity by signaling through the Notch4 receptor

    Canc. Res.

    (2010)
  • I.N. Colaluca

    NUMB controls p53 tumour suppressor activity

    Nature

    (2008)
  • S. Pece

    Loss of negative regulation by Numb over Notch is relevant to human breast carcinogenesis

    J. Cell Biol.

    (2004)
  • H.Y. Choi

    p53 destabilizing protein skews asymmetric division and enhances NOTCH activation to direct self-renewal of TICs

    Nat. Commun.

    (2020)
  • D.S. Peiffer

    DAXX suppresses tumor-initiating cells in estrogen receptor-positive breast cancer following endocrine therapy

    Canc. Res.

    (2019)
  • J. Yun

    Crosstalk between PKCalpha and Notch-4 in endocrine-resistant breast cancer cells

    Oncogenesis

    (2013)
  • M.P. Shea

    Antiestrogen therapy increases plasticity and cancer stemness of prolactin-induced ERalpha(+) mammary carcinomas

    Canc. Res.

    (2018)
  • L. Liu

    Identification of domains for efficient Notch signaling activity in immobilized Notch ligand proteins

    J. Cell. Biochem.

    (2017)
  • J.W. Bai

    Notch signaling pathway and endocrine resistance in breast cancer

    Front. Pharmacol.

    (2020)
  • P. Sansone

    Self-renewal of CD133(hi) cells by IL6/Notch3 signalling regulates endocrine resistance in metastatic breast cancer

    Nat. Commun.

    (2016)
  • J.C. Jun

    Hypoxia-inducible factors and cancer

    Curr. Sleep Med. Rep.

    (2017)
  • P.D. Tiburcio et al.

    Complex role of HIF in cancer: the known, the unknown, and the unexpected

    Hypoxia (Auckl)

    (2014)
  • J. Ajdukovic

    HIF-1--a big chapter in the cancer tale

    Exp. Oncol.

    (2016)
  • D. Samanta

    PHGDH expression is required for mitochondrial redox homeostasis, breast cancer stem cell maintenance, and lung metastasis

    Canc. Res.

    (2016)
  • G.L. Semenza

    Targeting HIF-1 for cancer therapy

    Nat. Rev. Canc.

    (2003)
  • M. Majumder

    COX-2 induces breast cancer stem cells via EP4/PI3K/AKT/NOTCH/WNT Axis

    Stem Cell.

    (2016)
  • K. Tominaga

    Addiction to the IGF2-ID1-IGF2 circuit for maintenance of the breast cancer stem-like cells

    Oncogene

    (2017)
  • A. Jogi

    Hypoxia alters gene expression in human neuroblastoma cells toward an immature and neural crest-like phenotype

    Proc. Natl. Acad. Sci. U. S. A.

    (2002)
  • A. Soeda

    Hypoxia promotes expansion of the CD133-positive glioma stem cells through activation of HIF-1alpha

    Oncogene

    (2009)
  • K. Balamurugan

    C/EBPdelta links IL-6 and HIF-1 signaling to promote breast cancer stem cell-associated phenotypes

    Oncogene

    (2019)
  • Y. Chen

    ZMYND8 acetylation mediates HIF-dependent breast cancer progression and metastasis

    J. Clin. Invest.

    (2018)
  • M.M. Ching et al.

    Eicosanoids in cancer: prostaglandin E2 receptor 4 in cancer therapeutics and immunotherapy

    Front. Pharmacol.

    (2020)
  • Cited by (6)

    • UCHL1 promotes cancer stemness in triple-negative breast cancer

      2022, Pathology Research and Practice
      Citation Excerpt :

      Cancer stem cells (CSCs) are tumor cells with the ability of self-renewal and infinite proliferation, and can produce heterogeneous tumor cells. It is characterized by unlimited self-renewal capacity, differentiation potential, high tumorigenicity and drug resistance [18,20]. Studies have shown that cells with CD44+/CD24- phenotype in breast cancer have stem cell features in the aspects of self-renewal, proliferation and differentiation, which has become a hot spot in breast cancer research [21,22].

    • JARID2 promotes stemness and cisplatin resistance in non-small cell lung cancer via upregulation of Notch1

      2021, International Journal of Biochemistry and Cell Biology
      Citation Excerpt :

      These data provide first evidence for the role of JARID2 in regulating CDDP sensitivity. Increased cancer stem cell-like properties is linked to development of drug resistance (Hao et al., 2021; Mavingire et al., 2021). Therefore, targeting cancer stem cells may be an important strategy to overcome drug resistance.

    1

    Present address: Frederick National Laboratory for Cancer Research, PO Box B, Bldg. 432, Room 232 Frederick, MD 21702-1201.

    View full text