The immune system and inflammation in breast cancer

https://doi.org/10.1016/j.mce.2013.06.003Get rights and content

Abstract

During different stages of tumor development the immune system can either identify and destroy tumors, or promote their growth. Therapies targeting the immune system have emerged as a promising treatment modality for breast cancer, and immunotherapeutic strategies are being examined in preclinical and clinical models. However, our understanding of the complex interplay between cells of the immune system and breast cancer cells is incomplete. In this article, we review recent findings showing how the immune system plays dual host-protective and tumor-promoting roles in breast cancer initiation and progression. We then discuss estrogen receptor α (ERα)-dependent and ERα-independent mechanisms that shield breast cancers from immunosurveillance and enable breast cancer cells to evade immune cell induced apoptosis and produce an immunosuppressive tumor microenvironment. Finally, we discuss protumorigenic inflammation that is induced during tumor progression and therapy, and how inflammation promotes more aggressive phenotypes in ERα positive breast cancers.

Introduction

Despite significant therapeutic achievements in recent years, in industrialized countries, breast cancer remains the most common cancer in women. It causes about 40,000 deaths in the United States each year (Basu et al., 2012). Estrogen receptorα (ERα) positive breast cancers represent more than 70% of breast tumors and endocrine therapies such as selective estrogen receptor modulators (SERMs) and aromatase inhibitors are still the standard adjuvant treatment for these tumors. However, the majority of patients will develop resistance to hormonal therapy and will need alternative therapies (Clarke et al., 2001, Clarke et al., 2003, Osborne and Schiff, 2011).

For over a century, the idea that the immune system can control cancer has been a subject of debate. Only very recently has it become generally accepted that the immune system has the ability not only to prevent tumor growth but also to promote it through a process called immunoediting. This process is comprised of three phases: elimination, equilibrium and escape (Schreiber et al., 2011, Vesely et al., 2011). Elimination is achieved through identification and destruction of nascent transformed cells by acute tumor-inhibiting inflammation, characterized by infiltration of effector cells of the innate and adaptive immune system as well as production of tumor-inhibiting cytokines. The escape phase is sustained by chronic tumor-promoting inflammation, which mainly involves immunosuppressive cells and soluble factors (Vesely et al., 2011). Evading immune destruction has recently been recognized as a hallmark of cancer (Hanahan and Weinberg, 2011). In general, use of immunosuppressants following organ transplantation or HIV infection increases the risk of tumors such as skin cancer, non-Hodgkin’s lymphoma or lung cancers, but not cancers of organs such as breast, brain, prostate and ovary (Kirk et al., 2007, Jiang et al., 2010). These studies suggest that breast cancer cells may be less immunogenic or simply take longer to develop (Vesely et al., 2011). Historically pre-existing inflammation or infection was not considered to be an underlying risk factor for the development of breast cancer. However, it is now clear that the infiltration of leukocytes, in the correct context, can either eliminate or promote the development of breast cancers (DeNardo and Coussens, 2007, Coussens and Pollard, 2011). Several studies have shown that immunity and inflammation-associated gene expression signatures are able to predict or classify tamoxifen-resistant breast cancers (Jansen et al., 2005, Chanrion et al., 2008, Vendrell et al., 2008). This supports the notion that endocrine resistance is associated with a dysregulated immune response and/or excessive inflammation in the tumor microenvironment (Osborne and Schiff, 2011). A recent study suggests that the immune response profile and inflammatory signature in breast cancer may provide useful information on patient prognosis and treatment (Kristensen et al., 2012). These studies suggest that research associated with inflammation and the immune system may enhance therapeutic possibilities for breast cancers, especially for those resistant to endocrine therapies. To better understand the battle and interplay between breast cancer cells and cells of the immune system, in this review we discuss following topics: (1) anti-breast cancer effector cells of the immune system, (2) mechanisms of breast cancer resistance to antitumor immunity, (3) protumorigenic inflammation in breast cancer and (4) inflammation promotion of aggressive phenotypes of ERα positive breast cancer.

Section snippets

Anti-breast cancer effector cells of the immune system

Breast cancer is often initiated by genetic and epigenetic changes in genes that regulate the function of the mammary epithelial cells (Coussens and Pollard, 2011). To prevent the development of breast cancer, diverse intrinsic tumor-suppressor mechanisms induce senescence or apoptosis of neoplastic cells (Lacroix et al., 2006, Xu et al., 2011, Nicholls et al., 2012). In parallel, the immune system is recognized as an extrinsic tumor-suppressor that can eliminate epithelial cells that have

Tumor cell-autonomous modifications that enable breast tumors to evade immune detection and destruction

Breast cancer cell have developed several ways to avoid immune cell-mediated killing thereby allowing the development of overt tumors.

Immunosuppressive microenvironment of breast cancer

It is now widely accepted that tumor cells are able to induce a suppressive microenvironment that supports tumor growth, and that the suppressive microenvironment is comprised of immunosuppressive cells and soluble factors (DeNardo and Coussens, 2007, Coussens and Pollard, 2011, Vesely et al., 2011). Major immunosuppressive cells that are often found in breast tumors include regulatory T (Treg) cells and myeloid-derived suppressor cells (MDSCs).

Protumorigenic inflammation in breast cancer

The main types of inflammation in tumorigenesis and cancer include: Chronic inflammation that precedes tumor development, tumor-associated inflammation and therapy-induced inflammation. Inflammation induces an angiogenic switch (Grivennikov et al., 2010). The roles of proinflammatory cytokines and chemokines, such as IL-1, IL-6, IL-8, TNF-α, MCP-1, CCL5 and CXCL12 in breast cancer have been reviewed previously (Ben-Baruch, 2003, Goldberg and Schwertfeger, 2010, Baumgarten and Frasor, 2012).

Inflammation as a promoter for more aggressive ERα+ breast cancer

Epidemiologic studies showed that regular use of nonsteroidal anti-inflammatory drugs (NSAIDS), such as aspirin, reduce the risk of ERα+ but not ERα breast cancers (Terry et al., 2004, Wang and Dubois, 2010). Recent research strongly suggests that a proinflammatory tumor microenvironment is an important factor contributing to resistance to endocrine therapy (Osborne and Schiff, 2011). Numerous studies have shown that ERα+ breast cancers are more responsive to proinflammatory cytokines. For

Concluding remarks

Breast cancer progression is not an entirely cell-autonomous process. Development and metastasis of breast tumors are influenced and even driven by cells of the immune system and associated inflammatory mediators in the tumor microenvironment. The balance between antitumor immunity and tumor-promoting inflammation determines whether the tumor will progress or be controlled or eliminated. Inflammation induced during the natural tumor progression is probably one of the main reasons that the

Acknowledgements

Our research and preparation of this review is supported by NIH Grant DK 071909 (to DJS). The authors wish to gratefully acknowledge Dr. Yon Sung (Stanford University) for critically reading this manuscript.

References (150)

  • P. Kannan-Thulasiraman et al.

    Modulators of inflammation use nuclear factor-kappa B and activator protein-1 sites to induce the caspase-1 and granzyme B inhibitor, proteinase inhibitor 9

    J. Biol. Chem.

    (2002)
  • H. Korkaya et al.

    Activation of an IL6 inflammatory loop mediates trastuzumab resistance in HER2+ breast cancer by expanding the cancer stem cell population

    Mol. Cell

    (2012)
  • A.J. Krieg et al.

    Interplay between estrogen response element sequence and ligands controls in vivo binding of estrogen receptor to regulated genes

    J. Biol. Chem.

    (2004)
  • D. Padua et al.

    TGFbeta primes breast tumors for lung metastasis seeding through angiopoietin-like 4

    Cell

    (2008)
  • S. Araki et al.

    TGF-beta1-induced expression of human Mdm2 correlates with late-stage metastatic breast cancer

    J. Clin. Invest.

    (2010)
  • R.C. Bargou et al.

    Overexpression of the death-promoting gene bax-alpha which is downregulated in breast cancer restores sensitivity to different apoptotic stimuli and reduces tumor growth in SCID mice

    J. Clin. Invest.

    (1996)
  • S. Basu et al.

    Eicosanoids and adipokines in breast cancer: from molecular mechanisms to clinical considerations

    Antioxid. Redox Signal.

    (2012)
  • G.J. Bates et al.

    Quantification of regulatory T cells enables the identification of high-risk breast cancer patients and those at risk of late relapse

    J. Clin. Oncol.

    (2006)
  • S.C. Baumgarten et al.

    Minireview: inflammation: an instigator of more aggressive estrogen receptor (ER) positive breast cancers

    Mol. Endocrinol.

    (2012)
  • A. Ben-Baruch

    Host microenvironment in breast cancer development: inflammatory cells, cytokines and chemokines in breast cancer progression: reciprocal tumor-microenvironment interactions

    Breast Cancer Res.

    (2003)
  • Y. Ben-Neriah et al.

    Inflammation meets cancer, with NF-kappaB as the matchmaker

    Nat. Immunol.

    (2011)
  • H. Bernhard et al.

    Adoptive transfer of autologous, HER2-specific, cytotoxic T lymphocytes for the treatment of HER2-overexpressing breast cancer

    Cancer Immunol. Immunother.

    (2008)
  • B.N. Bidwell et al.

    Silencing of Irf7 pathways in breast cancer cells promotes bone metastasis through immune escape

    Nat. Med.

    (2012)
  • P.V. Bodine et al.

    Suppression of ligand-dependent estrogen receptor activity by bone-resorbing cytokines in human osteoblasts

    Endocrinology

    (1999)
  • M. Chanrion et al.

    A gene expression signature that can predict the recurrence of tamoxifen-treated primary breast cancer

    Clin. Cancer Res.

    (2008)
  • R. Clarke et al.

    Cellular and molecular pharmacology of antiestrogen action and resistance

    Pharmacol. Rev.

    (2001)
  • R. Clarke et al.

    Antiestrogen resistance in breast cancer and the role of estrogen receptor signaling

    Oncogene

    (2003)
  • K.S. Clive et al.

    The GP2 peptide: a HER2/neu-based breast cancer vaccine

    J. Surg. Oncol.

    (2012)
  • L.M. Coussens et al.

    Leukocytes in mammary development and cancer

    Cold Spring Harb. Perspect. Biol.

    (2011)
  • E.M. de Kruijf et al.

    HLA-E and HLA-G expression in classical HLA class I-negative tumors is of prognostic value for clinical outcome of early breast cancer patients

    J. Immunol.

    (2010)
  • A. de la Torre et al.

    NGlycolylGM3/VSSP vaccine in metastatic breast cancer patients: results of phase I/IIa clinical trial

    Breast Cancer (Auckl)

    (2012)
  • M. De Palma et al.

    Cancer: macrophages limit chemotherapy

    Nature

    (2011)
  • D.G. DeNardo et al.

    Inflammation and breast cancer. Balancing immune response: crosstalk between adaptive and innate immune cells during breast cancer progression

    Breast Cancer Res.

    (2007)
  • D.G. DeNardo et al.

    Leukocyte complexity predicts breast cancer survival and functionally regulates response to chemotherapy

    Cancer Discov.

    (2011)
  • M.Z. Dewan et al.

    Natural killer cells in breast cancer cell growth and metastasis in SCID mice

    Biomed. Pharmacother.

    (2005)
  • M.L. Disis et al.

    Existent T-cell and antibody immunity to HER-2/neu protein in patients with breast cancer

    Cancer Res.

    (1994)
  • Y. Drabsch et al.

    TGF-beta signaling in breast cancer cell invasion and bone metastasis

    J. Mammary Gland Biol. Neoplasia.

    (2011)
  • O. Fainaru et al.

    Tumor growth and angiogenesis are dependent on the presence of immature dendritic cells

    FASEB J.

    (2010)
  • I. Feldman et al.

    Identification of proteins within the nuclear factor-kappa B transcriptional complex including estrogen receptor-alpha

    Am. J. Obstet. Gynecol.

    (2007)
  • G. Finak et al.

    Stromal gene expression predicts clinical outcome in breast cancer

    Nat. Med.

    (2008)
  • J. Frasor et al.

    Synergistic up-regulation of prostaglandin E synthase expression in breast cancer cells by 17beta-estradiol and proinflammatory cytokines

    Endocrinology

    (2008)
  • D.I. Gabrilovich et al.

    Myeloid-derived suppressor cells as regulators of the immune system

    Nat. Rev. Immunol.

    (2009)
  • C. Ginestier et al.

    CXCR1 blockade selectively targets human breast cancer stem cells in vitro and in xenografts

    J. Clin. Invest.

    (2010)
  • N. Gionet et al.

    NF-kappaB and estrogen receptor alpha interactions: Differential function in estrogen receptor-negative and -positive hormone-independent breast cancer cells

    J. Cell. Biochem.

    (2009)
  • M. Gobert et al.

    Regulatory T cells recruited through CCL22/CCR4 are selectively activated in lymphoid infiltrates surrounding primary breast tumors and lead to an adverse clinical outcome

    Cancer Res.

    (2009)
  • J.E. Goldberg et al.

    Proinflammatory cytokines in breast cancer: mechanisms of action and potential targets for therapeutics

    Curr. Drug Targets

    (2010)
  • A.D. Gregory et al.

    Tumor-associated neutrophils: new targets for cancer therapy

    Cancer Res.

    (2011)
  • A.D. Gritzapis et al.

    Quantitative fluorescence cytometric measurement of estrogen and progesterone receptors: correlation with the hormone binding assay

    Breast Cancer Res. Treat.

    (2003)
  • V. Groh et al.

    Tumour-derived soluble MIC ligands impair expression of NKG2D and T-cell activation

    Nature

    (2002)
  • L.S. Gutierrez et al.

    The Fas/Fas-ligand system: a mechanism for immune evasion in human breast carcinomas

    Breast Cancer Res. Treat.

    (1999)
  • Cited by (180)

    • Effect of metabolism on the immune microenvironment of breast cancer

      2023, Biochimica et Biophysica Acta - Reviews on Cancer
    View all citing articles on Scopus
    View full text