Elsevier

Current Problems in Cancer

Volume 40, Issues 2–4, March–August 2016, Pages 95-105
Current Problems in Cancer

Endocrine therapy and strategies to overcome therapeutic resistance in breast cancer

https://doi.org/10.1016/j.currproblcancer.2016.09.001Get rights and content

Abstract

Despite the remarkable success of endocrine therapy in the treatment of patients with estrogen receptor (ER)- positive breast cancer, not all patients derive benefit from such therapy, or may benefit only temporarily before disease progression or relapse occurs. The value of endocrine therapy, which blocks ER signaling by a variety of strategies, lies in its simplicity, lower toxicity, and better alignment with preserved quality of life, particularly when compared to chemotherapy, which is more toxic and has only modest benefits for many patients with ER-positive breast cancer. It is therefore critical that we discover ways to extend endocrine therapy benefit in patients and prevent therapeutic resistance whenever possible.

The tremendous evolution in our understanding of endocrine resistance mechanisms, coupled with the increasing availability of novel agents that target resistance pathways, has led to enhanced treatment approaches for patients with ER-positive breast cancer, primarily through combinations of endocrine agents with a variety of pathway inhibitors. Despite these treatment advances and our changing view of ER-positive breast cancer, there is much work that needs to be done. It remains a problem that we cannot reliably predict which subsets of patients will experience disease relapse or progression on endocrine therapy, and as such, combination strategies with targeted agents have largely been used in unselected patients with ER-positive breast cancer, including those who continue to have endocrine-sensitive disease. Patient selection is a significant issue since most of the targeted therapeutics that we use with endocrine therapy are expensive and can be toxic, and we may be inadvertently overtreating patients whose disease can still be controlled with endocrine therapy alone. In this article, we will review current and future strategies in the treatment of ER-positive breast cancer, as well as the evolving role of targeted therapy in the management of endocrine-resistance.

Introduction

Breast cancer is a major health problem worldwide with more than 1 million new cases diagnosed each year and approximately 400,000 deaths annually.1 Approximately 70%-80% of all breast cancers express the estrogen receptors (ER) and progesterone receptors (PgR),2 at times referred to collectively as hormone receptor-positive breast cancer. The presence of hormone receptors is associated with certain predictable tumor characteristics and a biologic behavior that is more indolent when compared with hormone receptor-negative tumors. Importantly, ER and its ligand estrogen play a key role in the development and progression of breast cancer, and disruption of the ER signaling pathway using a variety of endocrine strategies has been a mainstay in the treatment of this disease (Fig).

More than 120 years ago, and long before we knew about the existence of ER in breast cancer, Beatson3 described the benefit of estrogen deprivation by oophorectomy in treatment of premenopausal women with advanced breast cancer. Although this estrogen deprivation strategy was the original endocrine therapy described, and the first targeted therapy approach, endocrine strategies evolved to include agents that target ER itself, with the selective estrogen receptor modulator tamoxifen as the prototype endocrine therapy modality for decades.4, 5, 6, 7 Although effective, tamoxifen was limited by the problem of therapeutic resistance2 and additional treatment modalities were needed. With time, therapeutic options for treatment of ER-positive breast cancer evolved further with the addition of several new and more powerful anti-ER strategies, including third-generation aromatase inhibitors (AIs), which cause estrogen deprivation in postmenopausal women,8 and fulvestrant, which leads to the degradation of ER.9

Despite the addition of these expanded endocrine therapy options with more effective inhibition of ER signaling,10 therapeutic resistance remained a major problem limiting the benefit of all forms of endocrine therapy, but also created more opportunity to study mechanisms of resistance and to target new resistance pathways to improve endocrine therapy action and modulate resistance. This has become possible with significant advances in our understanding of ER biology and the unraveling of an intricate interplay between ER and other survival and growth factor signaling pathways in a breast cancer cell (Fig).2, 11 Consequently, we have witnessed a progressive shift away from viewing ER as an isolated and static pathway in breast cancer, and increasingly view it as an interactive partner with other signaling pathways, as well as a dynamic target in response to the effect of treatment.

In this article, we highlight 2 key strategies that can be used to modulate endocrine response and resistance in patients. The first strategy involves inhibition of the PI3K/AKT/mTOR pathway, which is a key driver of endocrine resistance in patients with progressive disease on endocrine therapy. The second strategy involves cell cycle inhibition using agents that target cyclin dependent kinases (CDK) 4 and 6, and this strategy is increasingly taking center stage as a reliable approach to enhance endocrine effect, for both second and first-line metastatic disease. We will also highlight the evolving role of fulvestrant and ER degradation as a unique endocrine therapy approach to use in combination with resistance pathway inhibitors and future strategies. Finally, we will discuss the subset of ER-positive breast cancer that expresses the human epidermal growth factor receptor-2 (HER2) and potential implications for endocrine resistance. There are of course many facets and dimensions involved in endocrine resistance, and through continued understanding of ER biology and dynamicity we will continue to refine our therapeutic options in individual patients with ER-positive breast cancer and the timing of when to most effectively use the different available strategies to modulate endocrine therapy.

Section snippets

Role of PI3K/AKT/mTOR pathway in endocrine resistance

One of the most critical signaling pathways that emerge after prolonged estrogen deprivation in ER-positive breast cancer is the PI3K/AKT/mTOR survival pathway12, 13 and has, therefore, been an important feature of progressive disease biology after AI failure.14 Preclinical models of ER-positive breast cancer have consistently shown that inhibition of PI3K/AKT/mTOR signaling can restore endocrine sensitivity and halt tumor regrowth.15, 16, 17 Clinical data also support the role of this pathway

Role of CDK4/6 inhibition to modulate endocrine response

CDK 4/6 inhibition has recently emerged as an effective and feasible strategy to prevent as well as to overcome endocrine resistance in metastatic ER-positive breast cancer. Palbociclib, an oral inhibitor of CDK 4 and CDK 6, has shown activity in both the first-line and second-line metastatic disease settings when combined with endocrine therapy. PALOMA-1 was a phase II randomized clinical trial in patients with newly metastatic ER-positive breast cancer, which compared the combination of

Is ER degradation a more effective endocrine strategy to use with targeted therapy?

Theoretically, a strategy of completely or nearly completely destroying the ER, may lead to a more effective inhibition of highly ER-dependent tumors. Fulvestrant, which degrades ER, has emerged as a valuable endocrine modality for patients with metastatic ER-positive breast cancer, but there were questions on how best to use it in patients, with earlier issues related to timing of its use, proper dosing, and whether it should be combined with other agents for optimal effect.33, 34, 35 Despite

The interplay between ER and HER2

HER2 is amplified or overexpressed in 20%-30% of all invasive breast cancers, with approximately 10% of breast tumors expressing both ER and HER2.57, 58 HER2 is a critical driver of cancer cell survival that is associated with decreased benefit from endocrine therapy when present.59, 60 Preclinical studies show that functional “crosstalk” between HER2 and ER can be a mechanism of resistance to endocrine therapy and inhibition of HER2 improves endocrine sensitivity in these studies.61, 62

There

Future directions

Endocrine therapy has come a long way since it was initially described and continues to play a central role in the management of patients with ER-positive breast cancer. Our improved understanding of endocrine-resistance mechanisms over the past 15 years has produced tangible benefits for patients and we should continue to carefully study mechanisms of resistance to further refine therapy. In particular, we need to better identify which patients are most likely to derive the most benefit from

Funding source

This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

References (80)

  • J.F. Robertson et al.

    Ganitumab with either exemestane or fulvestrant for postmenopausal women with advanced, hormone-receptor-positive breast cancer: a randomised, controlled, double-blind, phase 2 trial

    Lancet Oncol

    (2013)
  • C. Thomas et al.

    Estrogen receptor mutations and functional consequences for breast cancer

    Trends Endocrinol Metab

    (2015)
  • J. Baselga et al.

    Lapatinib with trastuzumab for HER2-positive early breast cancer (NeoALTTO): a randomised, open-label, multicentre, phase 3 trial

    Lancet

    (2012)
  • L. Gianni et al.

    Efficacy and safety of neoadjuvant pertuzumab and trastuzumab in women with locally advanced, inflammatory, or early HER2-positive breast cancer (NeoSphere): a randomised multicentre, open-label, phase 2 trial

    Lancet Oncol

    (2012)
  • L. Gianni et al.

    Neoadjuvant and adjuvant trastuzumab in patients with HER2-positive locally advanced breast cancer (NOAH): follow-up of a randomised controlled superiority trial with a parallel HER2-negative cohort

    Lancet Oncol

    (2014)
  • I. Vaz-Luis et al.

    Human epidermal growth factor receptor-2-positive breast cancer: does estrogen receptor status define two distinct subtypes?

    Ann Oncol

    (2013)
  • F. Montemurro et al.

    Human epidermal growth factor receptor 2 (HER2)-positive and hormone receptor-positive breast cancer: new insights into molecular interactions and clinical implications

    Ann Oncol

    (2013)
  • I. Helen

    Global health inequalities and breast cancer: an impending public health problem for developing countries

    Breast J

    (2008)
  • S. Massarweh et al.

    Unraveling the mechanisms of endocrine resistance in breast cancer: new therapeutic opportunities

    Clin Cancer Res

    (2007)
  • G.T. Beatson

    On the treatment of inoperable cases of carcinogen of the mamma: suggestions for a new method of treatment with illustrative cases

    Lancet

    (1896)
  • I.A. Jaiyesimi et al.

    Use of tamoxifen for breast cancer: twenty-eight years later

    J Clin Oncol

    (1995)
  • EBCTCG

    Tamoxifen for early breast cancer: an overview of the randomised trials

    The Lancet

    (1998)
  • B. Fisher et al.

    Tamoxifen for prevention of breast cancer: report of the National Surgical Adjuvant Breast and Bowel Project P-1 Study

    J Natl Cancer Inst

    (1998)
  • C.K. Osborne

    Aromatase inhibitors in relation to other forms of endocrine therapy for breast cancer

    Endocr Relat Cancer

    (1999)
  • A. Howell et al.

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

    Cancer

    (2000)
  • A.U. Buzdar

    Advances in endocrine treatments for postmenopausal women with metastatic and early breast cancer

    Oncologist

    (2003)
  • R. Schiff et al.

    Breast cancer endocrine resistance: how growth factor signaling and estrogen receptor coregulators modulate response

    Clin Cancer Res

    (2003)
  • G. Sabnis et al.

    Inhibition of the phosphatidylinositol 3-kinase/Akt pathway improves response of long-term estrogen-deprived breast cancer xenografts to antiestrogens

    Clin Cancer Res

    (2007)
  • R.J. Santen et al.

    Long-term estradiol deprivation in breast cancer cells up-regulates growth factor signaling and enhances estrogen sensitivity

    Endocr Relat Cancer

    (2005)
  • M. Sun et al.

    Phosphatidylinositol-3-OH Kinase (PI3K)/AKT2, activated in breast cancer, regulates and is induced by estrogen receptor alpha (ERalpha) via interaction between ERalpha and PI3K

    Cancer Res

    (2001)
  • T.W. Miller et al.

    Phosphatidylinositol 3-kinase and antiestrogen resistance in breast cancer

    J Clin Oncol

    (2011)
  • Comprehensive molecular portraits of human breast tumours

    Nature

    (2012)
  • K. Stemke-Hale et al.

    An integrative genomic and proteomic analysis of PIK3CA, PTEN, and AKT mutations in breast cancer

    Cancer Res

    (2008)
  • J. Baselga et al.

    Everolimus in postmenopausal hormone-receptor-positive advanced breast cancer

    N Engl J Med

    (2012)
  • T. Bachelot

    Randomized phase II trial of everolimus in combination with tamoxifen in patients with hormone receptor-positive, human epidermal growth factor receptor 2-negative metastatic breast cancer with prior exposure to aromatase inhibitors: a GINECO Study

    J Clin Oncol

    (2012)
  • S. Massarweh et al.

    A phase II study of combined fulvestrant and everolimus in patients with metastatic estrogen receptor (ER)-positive breast cancer after aromatase inhibitor (AI) failure

    Breast Cancer Res Treat

    (2014)
  • G.N. Hortobagyi et al.

    Correlative analysis of genetic alterations and everolimus benefit in hormone receptor-positive, human epidermal growth factor receptor 2-negative advanced breast cancer: results from BOLERO-2

    J Clin Oncol

    (2016)
  • S. Massarweh et al.

    Effect of mutations in distinct components of the PI3K/AKT/mTOR pathway on sensitivity to endocrine therapy in estrogen receptor (ER)-positive breast cancer

    J Clin Oncol

    (2015)
  • N.L. Busaidy et al.

    Management of metabolic effects associated with anticancer agents targeting the PI3K-Akt-mTOR pathway

    J Clin Oncol

    (2012)
  • RS Finn et al.

    PALOMA-2: primary results from a phase III trial of palbociclib (P) with letrozole (L) compared with letrozole alone in postmenopausal women with ER+/HER2-advanced breast cancer (ABC)

    J Clin Oncol

    (2016)
  • Cited by (0)

    View full text