Elsevier

Steroids

Volume 72, Issue 2, February 2007, Pages 135-143
Steroids

Estrogen receptor signaling pathways in human non-small cell lung cancer

https://doi.org/10.1016/j.steroids.2006.11.019Get rights and content

Abstract

Lung cancer is the most common cause of cancer mortality in male and female patients in the US. The etiology of non-small cell lung cancer (NSCLC) is not fully defined, but new data suggest that estrogens and growth factors promote tumor progression. In this work, we confirm that estrogen receptors (ER), both ERα and ERβ, occur in significant proportions of archival NSCLC specimens from the clinic, with receptor expression in tumor cell nuclei and in extranuclear sites. Further, ERα in tumor nuclei was present in activated forms as assessed by detection of ER phosphorylation at serines-118 and -167, residues commonly modulated by growth factor receptor as well as steroid signaling. In experiments using small interfering RNA (siRNA) constructs, we find that suppressing expression of either ERα or ERβ elicits a significant reduction in NSCLC cell proliferation in vitro. Estrogen signaling in NSCLC cells may also include steroid receptor coactivators (SRC), as SRC-3 and MNAR/PELP1 are both expressed in several lung cell lines, and both EGF and estradiol elicit serine phosphorylation of SRC-3 in vitro. EGFR and ER also cooperate in promoting early activation of p42/p44 MAP kinase in NSCLC cells. To assess new strategies to block NSCLC growth, we used Faslodex alone and with erlotinib, an EGFR kinase inhibitor. The drug tandem elicited enhanced blockade of the growth of NSCLC xenografts in vivo, and antitumor activity exceeded that of either agent given alone. The potential for use of antiestrogens alone and with growth factor receptor antagonists is now being pursued further in clinical trials.

Introduction

Lung cancer is the most common cause of cancer mortality in both male and female patients in the US. It is estimated that more than 180,000 new cases of non-small cell lung cancer (NSCLC) will be diagnosed this year in the US, and about 165,000 patients will succumb to the disease. Survival rates from NSCLC are unacceptably low [1], [2], and new therapeutic options are urgently needed. The etiology of most NSCLC is not fully defined, but several studies suggest a role of estrogens in progression [2], [3], [4], [5]. Clearly, cigarette smoking remains the primary risk factor for lung cancer with 85–90% of all lung cancer patients having smoked cigarettes at some time. Remaining lung cancer cases occur in nonsmokers, mainly women. In the past, men had higher lung cancer incidence due to higher smoking rates. However, rates of smoking in women have increased, with a 600% increase in the death rate from lung cancer leading to a “full blown epidemic” as noted by the US Surgeon General [2]. Mortality from lung cancer in women now exceeds that from breast cancer.

Estrogen status appears to be a significant factor in lung cancer in women, with evidence that exogenous and endogenous estrogen may play a role in development of lung cancer, especially adenocarcinoma [2], [5], [6]. Women have naturally higher circulating estrogen levels than men that may increase their susceptibility to lung cancer. In addition, estrogen biosynthesis due to activity of aromatase is reported in lung, suggesting that estrogens are produced locally in women and men and could affect tumor development [4], [7]. Despite earlier conflicting reports on the presence of ER in lung [8], new work clearly shows that both ERα and ERβ mRNA and protein are expressed in malignant lung epithelial cells [4], [9], [10], [11]. Moreover, these receptors may play important biologic roles in lung and respond to antiestrogens. In lung tumor cells in the laboratory, estrogens stimulate cell proliferation and enhance tumor progression in vivo. Blockade of this pathway by competition for estrogen binding to ER is the basis of the therapeutic tamoxifen, a partial agonist that limits proliferative effects of estrogen in breast cancer. However, in the uterine endometrium, tamoxifen has more prominent agonist effects and promotes growth, and it appears to have similar effects in lung [9], [11]. In contrast, we and others expect that new agents, such as Faslodex, an antiestrogen that downregulates ER [12], and aromatase inhibitors [13], downregulators of local estrogen production in tissues, may have previously unsuspected use to suppress lung cancer [4], [13].

Extranuclear and nuclear ER were postulated in early concepts of steroid interaction with target cells, such as breast and ovary [14], with the transcriptional activity of estrogen mediated by high-affinity ER in cell nuclei [15]. On estrogen binding in target cells, ER is phosphorylated and undergoes a conformational change that allows receptor dimerization and association of estrogen–ER complexes with specific estrogen response elements (ERE) in DNA, leading to transcription. Nuclear actions of estrogen are dependent, in part, on the subtype (ERα, ERβ) of ER, the gene promoter, and the steroid receptor coactivator and co-repressor proteins that modulate transcription [15], [16]. In addition, ER also regulates gene expression without direct binding to DNA. This occurs by protein–protein interaction with other transcription factors, such as AP-1, and with extranuclear signaling complexes that, in turn, modulate downstream gene transcription. Extranuclear signaling, such as MAPK activation, has a rapid onset and is mediated by ER in or tethered to membrane. In lung, as in breast, extranuclear ER appear to derive from the same transcript as nuclear ER [4], [17], [18], [19]. Nuclear and extranuclear ER act in concert with growth factor signaling pathways, such as EGFR [20], [21], to promote growth and survival [4], [11], [22]. The EGFR family of receptors, including EGFR (HER-1) and HER-2, are also implicated in lung cancer pathogenesis [23], [24]. This receptor axis is associated with progression of malignancy, inhibition of apoptosis and angiogenesis. EGF receptor antibodies or EGFR tyrosine kinase inhibitors elicit growth inhibition of lung tumors expressing these receptors. Moreover, EGFR/HER receptors regulate ligand-independent ER activation. Molecular details of interaction between ER and EGFR/HER are emerging, and ER is an important locus for signal convergence [4], [21], [22], [25]. In gene knockout mice lacking ERα, both estrogen- and EGF-stimulated growth in target tissue is blocked, while, in knockout mice lacking ERβ, disruption of normal lung development occurs [26]. Thus, ER may mediate transcription by integrating signals from growth factor pathways as well as from estrogen binding [27], [28], [29], [30]. Although some reports suggest that extranuclear ER is an alternative protein [31], [32], most studies indicate that extranuclear and nuclear ER derive from the same transcript [17], [33], [34], [35]. Post-translational ER modification can elicit membrane targeting [36], [37], [38], and association of ER with other adaptor or signaling proteins (e.g. shc, c-src or MNAR) may occur [39], [40], [41].

Section snippets

Cell culture

Human non-small cell lung cancer cells (NCI-H23 [H23], A549) and breast cancer cells (MCF-7 and SKBR3) were from ATCC. MCF-7 tumor cells with HER-2 overexpression (MCF-7/HER-2) were prepared as before [28]. Cell lines were routinely maintained in RPMI 1640 medium with 10% fetal bovine serum (FBS, Invitrogen/Life Technologies, Carlsbad, CA), 2 mM l-glutamine and 1% Antibiotic-Antimycotic solution 100× (Mediatech, Herndon, VA). For estrogen-free conditions, media were changed 48 h before

ERα and ERβ are both expressed in archival human NSCLC specimens from the clinic

The prevalence of ERα and ERβ in human NSCLC specimens was assessed by standard IHC methods using archival formalin-fixed, paraffin-embedded human tumor specimens. Fig. 1 presents representative examples of IHC staining patterns for ERα and ERβ. Appropriate tissue and reagent controls were done to confirm specificity [43]. For example, in the absence of antibodies to ERα or ERβ, no specific staining was observed (see Fig. 1A and D). Staining in tumor cell nuclei was observed for both ERα (Fig. 1

Discussion

Data from the present study confirm earlier work showing that estrogen as well as growth factors promote the progression of human NSCLC [4], [9], [10], [11]. Although previously not recognized, it is now known that estradiol promotes the growth of NSCLC [4], [11]. NSCLC cells harbor a complete estrogen signaling system, including estrogen receptors, estrogen-responsive elements in DNA and steroid hormone receptor coactivators. Using controlled homogenization and quantitative subcellular

Acknowledgments

We thank Dr. Hermes J. Garbán for his support in the preparation of this manuscript. Research supported by funding from NCI Lung Cancer SPORE Program at UCLA (P50 CA90388), Hamburger Fund of the Jonsson Cancer Center, Stiles Program in Integrative Oncology (in vitro work) and National Lung Cancer Partnership.

References (64)

  • R.J. Pietras et al.

    Estrogen receptors in uterine plasma membrane

    J Steroid Biochem

    (1979)
  • S. Dubey et al.

    Non-small-cell lung cancer and breast carcinoma: chemotherapy and beyond

    Lancet Oncol

    (2006)
  • J.C. Reese et al.

    Mutagenesis of cysteines in the hormone binding domain of the human estrogen receptor. Alterations in binding and transcriptional activation by covalently and reversibly attaching ligands

    J Biol Chem

    (1991)
  • J.D. Patel et al.

    Lung cancer in US women: a contemporary epidemic

    JAMA

    (2004)
  • L.P. Stabile et al.

    Estrogen receptor pathways in lung cancer

    Curr Oncol Rep

    (2004)
  • E. Taioli et al.

    Re: endocrine factors and adenocarcinoma of the lung in women

    J Natl Cancer Inst

    (1994)
  • C.W. Beattie et al.

    Steroid receptors in human lung cancer

    Cancer Res

    (1985)
  • P.A. Hershberger

    Regulation of endogenous gene expression in human non-small cell lung cancer cells by estrogen receptor ligands

    Cancer Res

    (2005)
  • E.A. Kirsch

    Estrogen acutely stimulates endothelial nitric oxide synthase in H441 human airway epithelial cells

    Am J Respir Cell Mol Biol

    (1999)
  • L.P. Stabile

    Human non-small cell lung tumors and cells derived from normal lung express both estrogen receptor alpha and beta and show biological responses to estrogen

    Cancer Res

    (2002)
  • A. Howell

    ICI 182,780 (Faslodex): development of a novel, “pure” antiestrogen

    Cancer

    (2000)
  • E.V. Jensen et al.

    Estrogen-receptor interaction

    Science

    (1973)
  • R.M. Evans

    The steroid and thyroid hormone receptor superfamily

    Science

    (1988)
  • M. Razandi

    Cell membrane and nuclear estrogen receptors (ERs) originate from a single transcript: studies of ERalpha and ERbeta expressed in Chinese hamster ovary cells

    Mol Endocrinol

    (1999)
  • A.M. Norfleet

    Antibodies to the estrogen receptor-alpha modulate rapid prolactin release from rat pituitary tumor cells through plasma membrane estrogen receptors

    FASEB J

    (2000)
  • D.C. Marquez et al.

    Membrane-associated binding sites for estrogen contribute to growth regulation of human breast cancer cells

    Oncogene

    (2001)
  • S.W. Curtis

    Physiological coupling of growth factor and steroid receptor signaling pathways: estrogen receptor knockout mice lack estrogen-like response to epidermal growth factor

    Proc Natl Acad Sci USA

    (1996)
  • D.C. Marquez

    Epidermal growth factor receptor and tyrosine phosphorylation of estrogen receptor

    Endocrine

    (2001)
  • C.J. Gruber

    Production and actions of estrogens

    N Engl J Med

    (2002)
  • T.J. Lynch

    Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib

    N Engl J Med

    (2004)
  • J.G. Paez

    EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy

    Science

    (2004)
  • L.P. Stabile

    Combined targeting of the estrogen receptor and the epidermal growth factor receptor in non-small cell lung cancer shows enhanced antiproliferative effects

    Cancer Res

    (2005)
  • Cited by (0)

    View full text