Elsevier

Seminars in Cancer Biology

Volume 31, April 2015, Pages 3-15
Seminars in Cancer Biology

Review
The impact of tumor stroma on drug response in breast cancer

https://doi.org/10.1016/j.semcancer.2014.05.006Get rights and content

Abstract

In the last two decades the breast cancer mortality rate has steadily declined, in part, due to the availability of better treatment options. However, drug resistance still remains a major challenge. Resistance can be an inherent feature of breast cancer cells, but can also arise from the tumor microenvironment. This review aims to focus on the modulatory effect of the tumor microenvironment on the differing response of breast cancer subtypes to targeted drugs and chemotherapy.

Introduction

Breast cancer is the most frequent cancer in women and accounts for 14% of cancer deaths in the United States annually [1]. Due to early detection by mammographic screening and also in part due to improved treatments we are beginning to see an impact on breast cancer outcome. Since 1990, mortality rates have decreased 2.2% each year, while incidence rates fluctuated up until 2003 and remained relatively stable since. Additionally, reductions in breast cancer mortality have also been observed in European studies [2].

Breast cancer is a heterogenous disease. The classical, and still routine way, to subtype breast cancer is by immunohistochemical analysis. Based on this analysis, breast cancer can be divided into three main groups: (1) estrogen receptor (ER)α-positive, (2) human epidermal growth factor receptor (Her)2 positive and (3) triple-negative (ERα-negative, progesterone receptor-negative, Her2-negative) subtypes. These subtypes are of clinical relevance in terms of prognosis and targeted treatments against ERα and Her2 are routinely used in two of the three subtypes [3], [4]. With the availability of molecular tools, breast cancer has also been subtyped with greater detail by gene expression profiling. In a seminal paper, Perou and co-workers showed that breast cancer subtypes can be distinguished by clusters of genes that are predominantly expressed in luminal breast epithelial cells, myoepithelial (basal) cells and cells whose expression is associated with elevated Her2 levels [5]. These subtypes are called luminal-like, basal-like and Her2-enriched, respectively. The molecular subtypes overlap with the immunohistochemical subtypes, such that most luminal-like tumors are ERα-positive, basal-like tumors triple-negative and Her2-enriched Her2-positive and vice versa [6]. However, the gene expression portraits allowed greater distinction in tumor subtypes within the three molecular subtypes leading to the identification of a luminal B as well as a luminal A subtype within the luminal-like tumors [7]. The gene expression subtypes are also of clinical importance, as patients with luminal B tumors have a worse prognosis than those with luminal A disease [8]. Later, an additional molecular subtype, the claudinlow subtype, was characterized by a mesenchymal/cancer stem cell-like gene expression profile [9]. Hence, the use of molecular tools allows an even more precise breast cancer subtyping [10], which puts us well on the path to personalized therapy by targeting the key factors to which individual cancer (cells) are addicted [11].

Chemotherapeutic drugs are commonly used to treat breast cancer. These drugs interfere with one of the key hallmarks of cancer, proliferation [12]. Chemotherapeutic drugs either damage DNA and thus prevent DNA synthesis or inhibit genes involved in proliferation [13]. Anthracyclines, such as epirubicin and doxorubicin, combined with fluorouracil and cyclophosphamide are effective treatment options for patients with early breast cancer [14]. Taxanes (paclitaxel or docetaxel) are also often used in Her2-positive breast cancers [15]. In triple-negative disease, treatments with taxane or anthracycline alone or in combination are common [16].

Targeted drugs have also been available for many years. For example, ERα-dependent breast cancers can be successfully treated by anti-estrogens, such as tamoxifen or fulvestrant, or aromatase inhibitors, such as letrozole, in a setting called endocrine therapy [3]. Interestingly, between 1992 and 2008, a shift in the incidence toward ERα-positive tumors has been observed in the United States [1]. This may additionally account for the decline in mortality rate since ERα-positive tumors are associated with better prognosis and targeted endocrine treatments are available. To specifically target Her2-positive breast cancer, very effective drugs are available, such as the anti-Her2 antibodies trastuzumab and pertuzumab, or Her2 kinase inhibitors, such as lapatinib [4]. These Her2 targeted drugs are often combined with taxanes for optimal treatment [15]. Triple-negative breast cancers are still only treated with chemotherapy, as no specific targeted drugs can currently be recommended. Drugs against potential targets, such as epidermal growth factor receptor (EGFR) or poly(adenosine diphosphate ribose) polymerase (PARP), have shown limited success in this subtype [17], [18]. Recently, further analysis of the triple negative subtype revealed additional heterogeneity indicating subsets of triple negative disease that may be more sensitive to specific drugs, such as the luminal androgen receptor (LAR) triple negative subtype which has been shown to respond to androgen receptor antagonists [19].

Despite successes in developing powerful anti-cancer drugs, drug resistance remains a major obstacle in cancer treatment. Drug resistance may be a pre-existing, inherent feature of cancer cells (intrinsic drug resistance) or may be acquired during the course of treatment leading to a relapse after months or years (acquired resistance). Intrinsic or acquired drug resistance is well established on a genetic and/or epigenetic level resulting in a blockage of pro-apoptotic pathways, in overexpression of anti-apoptotic proteins or in more efficient DNA damage repair [20]. Multidrug resistance can also be found through the promotion of drug efflux.

Even for specific drugs, such as anti-estrogens or anti-Her2 antibodies, many different mechanisms have been described that can lead to resistance [21], [22], [23]. For example, Her2-positive breast cancer cells can acquire resistance by mechanisms ranging as different as downregulation of phosphatase and tensin homologue (PTEN), an inhibitor of the phosphatidylinositol-3-kinase (PI3K) pathway [24] through to activation of integrin β1-dependent signaling [25]. Likewise, in the same breast cancer cell line (MCF-7), resistance against the anti-estrogen tamoxifen can be achieved by overexpression of the androgen receptor [26] or by fostering the crosstalk between the G-protein coupled receptor (GPR)30 with EGFR [27]. The complexity of these signaling pathways and their ability to crosstalk with other pathways may explain the wide range of mechanisms that can lead to resistance. For example, ERα shows ligand-dependent and ligand-independent activities and can act as a transcription factor in a genomic fashion or as component of signaling cascades in a non-genomic fashion. Endocrine resistance may be accompanied by loss of ERα and activation of an alternative survival pathway or, more often, by a switch from estrogen-dependent to estrogen-independent ERα-driven survival [22], [23]. Despite these different mechanisms, endocrine resistance is often accompanied by increased activity of the PI3K pathway [28], which can be targeted in variety of ways [29]. One possibility is to inhibit the mammalian target of rapamycin (mTOR), an important effector of the PI3K pathway, and shown to be involved in critical cellular functions, such as cell growth, proliferation and survival [30]. In a recent study it was shown that breast cancer patients that developed endocrine resistance responded positively to the rapamycin homologue RAD001 [31] suggesting that blocking the PI3K survival pathway is a potential therapy option to overcome endocrine resistance in patients [28].

Most cancers are heterogeneous and within a single tumor different subpopulations of cancer cells can be found. In theory, the higher the number of subpopulations present in a tumor, the higher the chance that one will be drug resistant (Fig. 1) leading to a pool of surviving cells called minimal residual disease. Tumor cell heterogeneity therefore confers tumor robustness allowing it to withstand drug treatment, even though only a few cells may survive.

There are two alternative hypotheses that could explain tumor heterogeneity. One hypothesis claims that heterogeneity is caused by clonal evolution, where different cancer cell subpopulations emerge from a single clone in the course of tumor progression [32]. Alternatively, it has also been proposed that cancer stem cells (CSCs) are responsible for tumor heterogeneity [33], [34]. CSCs are cells that give rise to heterogenous progenies and are present in small numbers in primary breast cancers and breast cancer cell lines [35]. Both concepts (clonal evolution and CSCs) may not be mutually exclusive as different subpopulations of CSCs may exist that generate different progenies (CSC heterogeneity) [36], [37]. Supporting the concept of CSC heterogeneity and its impact on the tumor cell phenotype a single mutation in the breast cancer-1 (BRCA-1) gene has been shown to alter breast cancer progenitor cell fate [38].

The concept of CSCs is an attractive model to explain intrinsic drug resistance [39]. Through overexpressing ATP binding cassette (ABC) drug transporter proteins, CSCs are able to export many drugs and are therefore considered to be multi-drug resistant cells [35]. In addition, there are many more features, such as higher expression of anti-apoptotic proteins or a more effective DNA damaging repair system, that render CSCs drug-resistant [40]. Also, as CSCs differ from non-CSC cancer cells in many ways, CSCs may be dependent upon other signaling pathways than their non-CSC neighbors. For example, in ERα-positive breast cancer, CSCs show much lower ERα-levels than bulk tumor and, at the same time, display an expression pattern indicative of a hyperactive PI3K pathway [41]. Hence, in theory, endocrine therapy would be unlikely to affect CSCs and could lead to drug resistance. Nevertheless, there is evidence that CSCs in ERα-positive breast cancers may need ERα-positive non-CSC neighbor cells for activity [42]. This is in line with the concept of stem cell niches which provide an environment that supports stem cells to maintain their stemness [43]. Irrespective of the potential dependency of CSCs on the bulk tumor, CSCs are considered to be a major risk for relapse. Hence, CSC-specific drugs in combination with commonly used drugs that target the bulk tumor may help to improve the outcome of cancer patients [44].

Section snippets

Environment-mediated drug resistance

The stroma is a critical compartment in normal tissue and in tumor tissue. Normal stroma and tumor stroma are quite distinct. There is evidence demonstrating that normal stroma has anti-cancer activity [45]. This anti-cancer effect may also contribute to longevity. It seems that the ability of the stroma to suppress carcinogenesis correlates with the life span of a species [46]. However, once converted to tumor stroma, the effect is different and in most cases promotes cancer progression. It

Conclusions

The tumor microenvironment affects drug response in all three major breast cancer subtypes, the ERα-positive, Her-positive and triple-negative breast cancers. The modulatory effect of the stroma depends on a combination of the drug, the subtype and the surrounding stromal components. The majority of drug resistance data are available in ERα-positive breast cancer cells that were treated with the anti-estrogen tamoxifen. In this subtype, the drug resistance mechanism often involves the

Conflict of interest

The authors declare that there are no conflicts of interest.

Acknowledgments

The authors thank Angela Dittmer and Jacqueline Hall for critically reading the manuscript. This work was supported by the Deutsche Krebshilfe, Grant No. 109271.

References (214)

  • Z. Castano et al.

    The bed and the bugs: interactions between the tumor microenvironment and cancer stem cells

    Semin Cancer Biol

    (2012)
  • E.S. Nakasone et al.

    Imaging tumor–stroma interactions during chemotherapy reveals contributions of the microenvironment to resistance

    Cancer Cell

    (2012)
  • C.A. Sherman-Baust et al.

    Remodeling of the extracellular matrix through overexpression of collagen VI contributes to cisplatin resistance in ovarian cancer cells

    Cancer Cell

    (2003)
  • S. Huang et al.

    MED12 controls the response to multiple cancer drugs through regulation of TGF-beta receptor signaling

    Cell

    (2012)
  • M.T. de la Fuente et al.

    Involvement of p53 in alpha4beta1 integrin-mediated resistance of B-CLL cells to fludarabine

    Biochem Biophys Res Commun

    (2003)
  • C. Margadant et al.

    Mechanisms of integrin activation and trafficking

    Curr Opin Cell Biol

    (2011)
  • H. Schafer et al.

    Combined treatment of L1CAM antibodies and cytostatic drugs improve the therapeutic response of pancreatic and ovarian carcinoma

    Cancer Lett

    (2012)
  • C. Rolny et al.

    HRG inhibits tumor growth and metastasis by inducing macrophage polarization and vessel normalization through downregulation of PlGF

    Cancer Cell

    (2011)
  • J. Cook et al.

    Tumour-associated macrophages and cancer

    Curr Opin Pharmacol

    (2013)
  • A.T. Toriola et al.

    Trends in breast cancer incidence and mortality in the United States: implications for prevention

    Breast Cancer Res Treat

    (2013)
  • S. Njor et al.

    Breast cancer mortality in mammographic screening in Europe: a review of incidence-based mortality studies

    J Med Screen

    (2012)
  • C. Davies et al.

    Relevance of breast cancer hormone receptors and other factors to the efficacy of adjuvant tamoxifen: patient-level meta-analysis of randomised trials

    Lancet

    (2011)
  • C.A. Hudis

    Trastuzumab – mechanism of action and use in clinical practice

    N Engl J Med

    (2007)
  • C.M. Perou et al.

    Molecular portraits of human breast tumours

    Nature

    (2000)
  • T. Sorlie et al.

    Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications

    Proc Natl Acad Sci USA

    (2001)
  • J.S. Parker et al.

    Supervised risk predictor of breast cancer based on intrinsic subtypes

    J Clin Oncol

    (2009)
  • A. Prat et al.

    Phenotypic and molecular characterization of the claudin-low intrinsic subtype of breast cancer

    Breast Cancer Res

    (2010)
  • C.G.A. Network

    Comprehensive molecular portraits of human breast tumours

    Nature

    (2012)
  • M.L. Whitfield et al.

    Common markers of proliferation

    Nat Rev Cancer

    (2006)
  • J. Greene et al.

    The role of anthracyclines in the treatment of early breast cancer

    J Oncol Pharm Pract

    (2014)
  • N. Wilcken et al.

    Systemic treatment of HER2-positive metastatic breast cancer: a systematic review

    Asia Pac J Clin Oncol

    (2014)
  • S.L. Davis et al.

    Triple-negative breast cancer: bridging the gap from cancer genomics to predictive biomarkers

    Ther Adv Med Oncol

    (2014)
  • H. Masuda et al.

    Role of epidermal growth factor receptor in breast cancer

    Breast Cancer Res Treat

    (2012)
  • C.L. Griffiths et al.

    Triple negative breast cancer: a brief review of its characteristics and treatment options

    J Pharm Pract

    (2012)
  • B.D. Lehmann et al.

    Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies

    J Clin Invest

    (2011)
  • E.L. de Oliveira Niero et al.

    The multiple facets of drug resistance: one history, different approaches

    J Exp Clin Cancer Res

    (2014)
  • T.A. Bailey et al.

    Mechanisms of Trastuzumab resistance in ErbB2-driven breast cancer and newer opportunities to overcome therapy resistance

    J Carcinog

    (2011)
  • E.A. Musgrove et al.

    Biological determinants of endocrine resistance in breast cancer

    Nat Rev Cancer

    (2009)
  • C.K. Osborne et al.

    Mechanisms of endocrine resistance in breast cancer

    Annu Rev Med

    (2011)
  • C. Huang et al.

    beta1 integrin mediates an alternative survival pathway in breast cancer cells resistant to lapatinib

    Breast Cancer Res

    (2011)
  • F. De Amicis et al.

    Androgen receptor overexpression induces tamoxifen resistance in human breast cancer cells

    Breast Cancer Res Treat

    (2010)
  • Z. Mo et al.

    GPR30 as an initiator of tamoxifen resistance in hormone-dependent breast cancer

    Breast Cancer Res

    (2013)
  • P. Liu et al.

    Targeting the phosphoinositide 3-kinase pathway in cancer

    Nat Rev Drug Discov

    (2009)
  • B. Magnuson et al.

    Regulation and function of ribosomal protein S6 kinase (S6K) within mTOR signalling networks

    Biochem J

    (2012)
  • J. Baselga et al.

    Everolimus in postmenopausal hormone-receptor-positive advanced breast cancer

    N Engl J Med

    (2012)
  • H. Clevers

    The cancer stem cell: premises, promises and challenges

    Nat Med

    (2011)
  • L.V. Nguyen et al.

    Cancer stem cells: an evolving concept

    Nat Rev Cancer

    (2012)
  • J.E. Visvader

    Cells of origin in cancer

    Nature

    (2011)
  • E. Charafe-Jauffret et al.

    Aldehyde dehydrogenase 1-positive cancer stem cells mediate metastasis and poor clinical outcome in inflammatory breast cancer

    Clin Cancer Res

    (2009)
  • J.E. Visvader et al.

    Cancer stem cells in solid tumours: accumulating evidence and unresolved questions

    Nat Rev Cancer

    (2008)
  • Cited by (82)

    • Nanoparticle-assisted targeting of the tumour microenvironment

      2022, OpenNano
      Citation Excerpt :

      The TME is thought to be refined, shaped and constantly co-evolving with cancer cells, orchestrated through various cellular and chemical signals which aid in the acquisition of cancer hallmarks and response to intrinsic and extrinsic stress, fundamentally promoting survival of malignant cells [41]. The reactive plasticity of the TME is best encapsulated by the stroma of solid tumours which consists of the ECM, CAFs and myofibroblasts, and provides a mechanical scaffold for proliferation of cancer cells, characterized by reprogrammed metabolism, activated transcription, altered synthesis of repair-associated proteins and a remodelled matrix [42,43]. The TME in turn, expresses tumour-promoting factors including immunosuppressive effector molecules and effector cells such as regulatory T cells (Treg), tumour-associated macrophages (TAMs), and myeloid-derived suppressor cells (MDSCs) [41].

    • In-vitro tumor microenvironment models containing physical and biological barriers for modelling multidrug resistance mechanisms and multidrug delivery strategies

      2021, Journal of Controlled Release
      Citation Excerpt :

      CAFs have been shown to activate tamoxifen resistance because that they activate Ras/Raf/MEK1/ERK1/2 and/or PI3K/AKT pathways [110–112]. TANs and TAMs are recruited to the drug-mediated tumor environment and contribute to drug resistance [113–115]. DeNardo and co-workers [116] showed that TAM blocking drugs, such as erlotinib or gefitinib, improved the response of breast cancer cells to chemotherapeutics [117,118].

    View all citing articles on Scopus
    View full text