Endocrine therapy resistance in breast cancer

Last updated

Endocrine therapy is a common treatment for estrogen receptor positive breast cancer. However, resistance to this therapy can develop, leading to relapse and progression of disease. This highlights the need for new strategies to combat this resistance.

Contents

Breast cancer overview

Hormonal status in breast cancer

Globally, breast cancer is the second most common type of cancer, comprising 11.6% of total worldwide cancer cases in 2018. [1] In the United Kingdom, breast cancer is the most common cancer type, affecting around 1 in 8 women. [2] It can be loosely divided into non-invasive, where the cancer is localized to the ducts or lobules in which it originated (in-situ), or invasive, where the cancer cells have spread beyond the initial duct or lobule into the surrounding breast tissue or other part of the body. [3] Risk factors that may predispose to breast cancer include increasing age and a family history of breast cancer. [2] Despite the increase in incidence of breast cancer with age, there is a notable deceleration of this increase following menopause. [4] Moreover, breast cancer risk is heightened following use of the combined oral contraceptive pill and combined hormone replacement therapy. [4] Armed with this evidence that endogenous and exogenous changes in estrogen and progesterone levels modulate the risk of breast cancer, it is apparent that hormones can play a key role in breast cancer.

Indeed, breast tumors can express certain hormone receptors, and use these to grow and proliferate. These hormone receptors (HR) are the estrogen receptor (ER) and the progesterone receptor (PR). Immunohistochemical analysis of a tumor sample, taken via biopsy or after surgery, is utilized to determine the presence of these receptors. To classify a tumor as ER or PR negative or positive, the Allred score is utilized, which takes into account both the proportion of ERs and PRs present and the overall intensity score of the staining - this method is considered controversial, however. [5]

Another receptor that often plays a role in breast cancer, although it is not a hormone receptor, is the human epidermal growth factor receptor 2 (HER2). The overexpression of HER2 is determined by immunohistochemistry (IHC), or with fluorescent in situ hybridization in those equivocal cases where IHC does not provide a clear result.

Different molecular subtypes of breast cancer have also been described, which loosely align with receptor status:

Additionally, cancers can be ER-/PR+ or ER+/PR-, but these are unnamed and relatively rare. [7] The receptor status of a cancer is assessed for all breast cancers as it has important implications on prognosis of the patient. It also dictates the treatment given: cancers that do express ER are likely to respond to endocrine therapy, but this type of therapy will have no effect on triple-negative breast cancers.

Mechanism of estrogen receptor action

75% of breast cancers are ER+. [8] ER is a transcription factor containing a DNA-binding domain which allows binding DNA at specific sequences called estrogen response elements (EREs), defined as 5’-GGTCAnnnTGACC-3’, where "n" refers to any nucleotide. [9] The N-terminal of the ER holds the activation function 1 (AF1) domain; the AF2 domain of the ER is contained within the ligand binding domain. The AF2 domain contains binding sites for coactivators. [10]

Estrogen is a steroid hormone and can cross the cell membrane freely. It can then bind to an ER, which is located in the cytoplasm of the cell. Upon binding, the ligand-receptor complex dimerizes with another, and this homodimer moves into the nucleus. Binding of an estrogen ligand exposes a site on the AF2 domain for coactivators to attach to the ER. [9] By recruiting coactivators and transcription factors, transcription of downstream genes, referred to as estrogen responsive genes, is initiated. These genes are of diverse function, but may potentiate aberrant cell survival and proliferation, contributing to tumorigenesis and cancer progression. For example, the ER can promote transcription of cyclin D1, an important regulator of the cell cycle. [10] By interfering with the expression of this regulator, estrogen and estrogen receptor signaling can disrupt the cell cycle.

Endocrine therapies interrupt this pathway, blocking the binding of the receptor to the response element and subsequent transcription of these estrogen-responsive genes. ER+/PR+ cancers have been found to show better results with administration of SERMs than ER+/PR- cancers. This trend has been found with AI therapy also, albeit in fewer studies. The biological basis of this remains unknown. [11]

Endocrine therapy overview

Estrogen

Estrogens are responsible for the regulation of the female reproductive system and development of secondary sexual characteristics. [12]

17β-Estradiol is the biologically active form of the sex hormone known as estrogen. [13] It is secreted by granulose cells of the ovarian follicle and the corpus luteum, and is the primary form of circulating form of estrogen. [12]

Other forms of endogenous estrogen exist, such as estrone, estriol and estretrol. Estretrol and estriol are primarily found in the body during pregnancy, and estrone is present during the menopause. [12]

All of the forms of estrogen found in the human body are able to bind to estrogen receptors (ER) present on cells. This initiates transcription in these cells, resulting in control of gene expression. [12]

Treatment strategies that work by blocking the effect of estrogen on breast cancer are referred to as endocrine (or hormone) therapies. These can target hormone regulation in two distinct ways. [14]

Blocking estrogen synthesis

Firstly, some forms of endocrine therapy can be administered to block estrogen synthesis. In pre-menopausal women, this can be achieved through surgical ovarian ablation (by oophorectomy) or chemical ovarian suppression (using luteinizing hormone releasing hormone agonists, such as goserilin). In postmenopausal women, the ovaries cease to be the main source of estrogen production, with estrogen being instead synthesized from regulatory steroid hormones called androgens in tissues of the bone, fat, and breast through the activity of the enzyme aromatase. [15] This can be blocked through the administration of aromatase inhibitors (AIs), which can be reversible inhibitors (non-steroidal) such as anastrozole and letrozole, or irreversible (steroidal) inhibitors like exemestane. [15]

Influencing the effect of estrogen

Secondly, other endocrine therapies can directly influence the effect of estrogen in cancer cells. ER function can be chemically blocked in pre- and post-menopausal women using antiestrogens, including:

Endocrine resistance overview

Definition of endocrine resistance

The term endocrine resistance describes a resistance to estrogen signaling suppression. [17] However, in many scientific studies endocrine resistance refers to resistance to estrogen or estrogen receptor suppression. [17] This resistance occurs due to the development of escape pathways which provide new ways for cancer cells to survive in the presence of endocrine therapy. [18] If a patient with ER+ breast cancer develops endocrine resistance, the endocrine therapy used to treat the cancer will no longer be effective. Approximately 30-50% of ER+ breast cancer patients will relapse as a result of endocrine resistance, proving it to be a predominant challenge in the treatment of ER+ breast cancer patients. [19]

Types of endocrine resistance

There are two types of endocrine resistance: primary and secondary. Primary resistance can also be described as de novo resistance or intrinsic resistance, which means the resistance existed before any treatment was initiated. [18] Secondary resistance is acquired, meaning that it occurs after some initial response to treatment due to exposure to endocrine therapy. [18]

Characterization of endocrine resistance

The table below summarizes how the two types of resistance are characterized in clinical settings.

Type of resistanceDescriptionRelapseProgressive diseaseResponse to further treatment
Primary. [18] De novo. [18] During first 2 years of adjuvant endocrine therapy. [20] Within the first 6 months of first-line endocrine therapy for metastatic breast cancer. [20] No response to subsequent endocrine therapy. [18]
Secondary. [18] Acquired. [18] After the first 2 years of treatment, during adjuvant endocrine therapy. [20]

OR

Within 1 year of finishing adjuvant endocrine therapy. [20]

6 or more months after starting endocrine therapy. [20] Shorter stages of response to a series of endocrine therapies until there is then no further response to endocrine treatment. [18]

Implications of resistance

Endocrine resistance in breast cancer leads to the progression of the disease and relapse. Resistance to endocrine therapy, leading to metastasis (spread of cancer beyond its place of origin) of breast cancer, which is one of the main reasons for death in breast cancer patients. [19] This highlights the need for new strategies to overcome endocrine resistance and better treat ER+ breast cancer.

Endocrine resistance mechanisms

ESR1 Modifications

Mutations in the ESR1 gene have been associated with the development of resistance to endocrine therapy. The ESR1 gene codes for estrogen receptors (ERα and ERβ). Whilst it is still not fully understood how ERβ impacts breast cancer development, ERα  is known to have a role in tumor growth and cell survival. [19]

Missense mutations, which are small mutations within a gene that can change the resulting protein, can lead to estrogen independence. This means that endocrine therapies blocking estrogen synthesis, such as aromatase inhibitors, lose efficacy because in a state of estrogen independence, the ER is stimulated without the need for estrogen, thus leading to constant ER activity. These mutations are rare in primary tumors; however, the frequency can be up to 30x higher in metastatic lesions. [19]

Another mechanism by which modifications to the ESR1 gene can lead to endocrine therapy resistance is by the occurrence of translocations. During translocations, a fusion protein is formed between YAP1, a gene crucial for proliferation and apoptosis (programmed cell death) of cells, and ESR1. The creation of this fusion protein can allow the tumor to grow independent of estrogen, develop anti-estrogen resistance, have induced cell motility and constitutive expression of ER target genes. [19]

Finally, it has been shown that there can be ESR1 gene amplification of up to 37% in breast cancer patients. However as of now there is no research showing a clear correlation between this amplification and endocrine resistance. [19]

miRNA regulators

miRNAs are non-coding sections of RNA that are involved in gene expression. It has been shown that upregulation of some of these miRNAs may be associated with the development of anti-estrogen resistance via two potential pathways. First by activating alternative growth pathways and secondly by inhibition of ER expression. Some of examples of this are described below. [19]

miR-155 stimulates the STAT3 signaling pathway, and this is associated with cell survival and endocrine resistance, specifically resistance to tamoxifen. In terms of inhibition, certain miRNAs are known to inhibit key inhibitors of the p27 pathway, hence allowing it carry on unchecked. Inhibition of the p27 pathway is well known for causing cell proliferation in tumors, and the levels of p27 can be measured and used as a marker for tamoxifen resistance. [19]

In contrast to this, downregulation of other miRNAs is also associated with the development of endocrine resistance. Some miRNAs act as inhibitors to ERα-36 which acts as an ERβ antagonist and an agonist of estrogen-like SERM effects. Therefore, low levels of these mRNAs are associated with ERα-36 mediated resistance. [19]

Another mechanism by which down-regulation of miRNAs may be implicated in the development of endocrine therapy resistance is by leading to overexpression of CDK3 and enhanced transcriptional activity of ER. [19]

GPER

GPER is a transmembrane protein that acts as an estrogen receptor, and it is present in 50-60% of breast cancer cases. The cellular location of GPER seems to correlate somewhat with tumor prognosis, where cytoplasmic GPER is associated with lower grade tumors and nuclear GPER with higher rates of metastasis. [21]

The signaling pathways associated with the GPER protein involve tyrosine kinases (from the Src family). Additionally, there is crosstalk between the GPER and EGFR signaling pathways. [21]

Tamoxifen has been shown to increase expression of GPER by binding to its receptor. The signaling pathways initiated lead not only to tamoxifen resistance, but also a higher risk of metastasis and cell growth. The overall survival rate hence decreases for patients displaying high GPER expression. [21]

PR expression

PR (progesterone receptor) is a gene expressed in around 50% of ER+ breast cancer patients. Clinical studies have shown that ER+/PR+ cases are more sensitive to endocrine therapy than ER+/PR- cases, suggesting that the loss of PR expression is associated with the development of endocrine therapy resistance. PR levels have been shown to be downregulated when there is activation of growth factors via the PI3K/Akt/mTOR pathway. [11]

HER2 over-expression

Over-expression of HER2 is found to be present in around 30% of metastatic breast tumors and is associated with a poorer prognosis. Studies have demonstrated that there is crosstalk between HER2 and ER signaling pathways. Specifically, studies have shown there is crosstalk between HER2 and the ER coactivator A1B1 which could enhance the estrogen agonist activity of tamoxifen bound ER. Additionally, HER2 has physical associations with the cell membrane ER and interactions between them have been shown to block ER-initiated apoptosis. [11]

Tackling endocrine resistance

When tackling endocrine resistance in breast cancer, the current strategies are focused around combining hormonal agents with drugs targeting several escape pathways, as outlined in the mechanisms of endocrine resistance section. The aim is to block all the tumor survival escapes. [11]

The main approach in overcoming endocrine resistance in those breast cancers that are both ER+ and HER2+ is by using a combination of endocrine and HER2-targeting agents. [22]

In trials conducted with a combination of anti-HER2 agents and an aromatase inhibitor, significant clinical benefit and improved progression-free survival have been observed. [22]

Trastuzumab is an example of an anti-HER2 agent which mainly depresses the growth of cells which are over-expressing HER2. [23]

Many endocrine resistant breast cancers which don’t respond to aromatase inhibitors still depend on ER signaling. [23]

Selective Estrogen Receptor Degraders (SERDs) can be used in these cases as they destabilize the ER and act as pure antagonists at the ER receptor. An example SERD is fulvestrant which binds to the ER to block its dimerization and nuclear localization. The fulvestrant-ER complex is unstable, resulting in degradation. [23]

There has also been research into the combined inhibition of the ER and EGFR. An example of a pure EGFR inhibitor is gefitinib, which has been used in phase II trials to evaluate its addition to tamoxifen in patients with HR-positive advanced breast cancer. [22]

The PI3K-Akt-mTOR signaling pathway and crosstalk with the ER signaling pathway is thought to be involved in the development of resistance to endocrine therapy. Therefore, studies have been done to assess the efficacy of using mTOR inhibitors, such as everolimus or temsirolimus, in ER-positive breast cancers. [22]

Some preclinical and clinical studies have shown the possibility of mTOR inhibitors as a first step treatment to shrink the breast cancer tumor before the main treatment of an aromatase inhibitor is given. It is agreed that more clinical trials are needed to confirm this issue.   [11]

Additionally, cyclin-dependent kinase (CDK) 4/6 inhibition has shown to improve the efficacy of endocrine treatment. [23]

Related Research Articles

Autocrine signaling is a form of cell signaling in which a cell secretes a hormone or chemical messenger that binds to autocrine receptors on that same cell, leading to changes in the cell. This can be contrasted with paracrine signaling, intracrine signaling, or classical endocrine signaling.

<span class="mw-page-title-main">Tamoxifen</span> Medication

Tamoxifen, sold under the brand name Nolvadex among others, is a selective estrogen receptor modulator used to prevent breast cancer in women and men. It is also being studied for other types of cancer. It has been used for Albright syndrome. Tamoxifen is typically taken daily by mouth for five years for breast cancer.

<span class="mw-page-title-main">Aromatase inhibitor</span> Class of drugs

Aromatase inhibitors (AIs) are a class of drugs used in the treatment of breast cancer in postmenopausal women and in men, and gynecomastia in men. They may also be used off-label to reduce estrogen conversion when supplementing testosterone exogenously. They may also be used for chemoprevention in women at high risk for breast cancer.

<span class="mw-page-title-main">Estrogen receptor</span> Proteins activated by the hormone estrogen

Estrogen receptors (ERs) are a group of proteins found inside cells. They are receptors that are activated by the hormone estrogen (17β-estradiol). Two classes of ER exist: nuclear estrogen receptors, which are members of the nuclear receptor family of intracellular receptors, and membrane estrogen receptors (mERs), which are mostly G protein-coupled receptors. This article refers to the former (ER).

<span class="mw-page-title-main">PELP-1</span> Mammalian protein found in Homo sapiens

Proline-, glutamic acid- and leucine-rich protein 1 (PELP1) also known as modulator of non-genomic activity of estrogen receptor (MNAR) and transcription factor HMX3 is a protein that in humans is encoded by the PELP1 gene. is a transcriptional corepressor for nuclear receptors such as glucocorticoid receptors and a coactivator for estrogen receptors.

Hormonal therapy in oncology is hormone therapy for cancer and is one of the major modalities of medical oncology, others being cytotoxic chemotherapy and targeted therapy (biotherapeutics). It involves the manipulation of the endocrine system through exogenous or external administration of specific hormones, particularly steroid hormones, or drugs which inhibit the production or activity of such hormones. Because steroid hormones are powerful drivers of gene expression in certain cancer cells, changing the levels or activity of certain hormones can cause certain cancers to cease growing, or even undergo cell death. Surgical removal of endocrine organs, such as orchiectomy and oophorectomy can also be employed as a form of hormonal therapy.

<span class="mw-page-title-main">Nuclear receptor coactivator 3</span> Protein-coding gene in the species Homo sapiens

The nuclear receptor coactivator 3 also known as NCOA3 is a protein that, in humans, is encoded by the NCOA3 gene. NCOA3 is also frequently called 'amplified in breast 1' (AIB1), steroid receptor coactivator-3 (SRC-3), or thyroid hormone receptor activator molecule 1 (TRAM-1).

<span class="mw-page-title-main">GPER</span> Protein-coding gene in the species Homo sapiens

G protein-coupled estrogen receptor 1 (GPER), also known as G protein-coupled receptor 30 (GPR30), is a protein that in humans is encoded by the GPER gene. GPER binds to and is activated by the female sex hormone estradiol and is responsible for some of the rapid effects that estradiol has on cells.

Breast cancer management takes different approaches depending on physical and biological characteristics of the disease, as well as the age, over-all health and personal preferences of the patient. Treatment types can be classified into local therapy and systemic treatment. Local therapy is most efficacious in early stage breast cancer, while systemic therapy is generally justified in advanced and metastatic disease, or in diseases with specific phenotypes.

Antiestrogens, also known as estrogen antagonists or estrogen blockers, are a class of drugs which prevent estrogens like estradiol from mediating their biological effects in the body. They act by blocking the estrogen receptor (ER) and/or inhibiting or suppressing estrogen production. Antiestrogens are one of three types of sex hormone antagonists, the others being antiandrogens and antiprogestogens. Antiestrogens are commonly used to stop steroid hormones, estrogen, from binding to the estrogen receptors leading to the decrease of estrogen levels. Decreased levels of estrogen can lead to complications in sexual development. Antiandrogens are sex hormone antagonists which are able to lower the production and the effects that testosterone can have on female bodies.

The estrogen receptor test (ERT) is a laboratory test to determine whether cancer cells have estrogen receptors. This information can guide treatment of the cancer.

Antihormone therapy is a type of hormone therapy that suppresses selected hormones or their effects, in contrast with hormone replacement therapy, which encourages hormone activity.

A hormone-receptor-positive (HR+) tumor is a tumor which consists of cells that express receptors for certain hormones. The term most commonly refers to estrogen receptor positive tumors, but can also include progesterone receptor positive tumors. Estrogen-receptor-positive tumors depend on the presence of estrogen for ongoing proliferation.

Breast cancer classification divides breast cancer into categories according to different schemes criteria and serving a different purpose. The major categories are the histopathological type, the grade of the tumor, the stage of the tumor, and the expression of proteins and genes. As knowledge of cancer cell biology develops these classifications are updated.

<span class="mw-page-title-main">Male breast cancer</span> Medical condition

Male breast cancer (MBC) is a cancer in males that originates in their breasts. Males account for less than 1% of new breast cancers with about 20,000 new cases being diagnosed worldwide every year. Its incidence rates in males vs. females are, respectively, 0.4 and 66.7 per 100,000 person-years. The worldwide incidences of male as well as female breast cancers have been increasing over the last few decades. Currently, one of every 800 men are estimated to develop this cancer during their lifetimes.

<span class="mw-page-title-main">Palbociclib</span> Medication for HR+ HER2− breast cancer

Palbociclib, sold under the brand name Ibrance among others, is a medication developed by Pfizer for the treatment of HR-positive and HER2-negative breast cancer. It is a selective inhibitor of the cyclin-dependent kinases CDK4 and CDK6. Palbociclib was the first CDK4/6 inhibitor to be approved as a cancer therapy.

Steroidal aromatase inhibitors are a class of drugs that are mostly used for treating breast cancer in postmenopausal women. High levels of estrogen in breast tissue increases the risk of developing breast cancer and the enzyme aromatase is considered to be a good therapeutic target when treating breast cancer due to it being involved in the final step of estrogen biosynthetic pathway and also its inhibition will not affect production of other steroids. Aromatase Inhibitors are classified into two categories based on their structure, nonsteroidal and steroidal; the latter resemble the structure of androstenedione. Steroidal aromatase inhibitors irreversibly inhibit the enzyme by binding covalently to the binding site of aromatase so the substrate cannot access it.

<span class="mw-page-title-main">Elacestrant</span> Chemical compound

Elacestrant, sold under the brand name Orserdu, is an anticancer medication which is used in the treatment of breast cancer. It is taken by mouth.

Estrogen deprivation therapy, also known as endocrine therapy, is a form of hormone therapy that is used in the treatment of breast cancer. Modalities include antiestrogens or estrogen blockers such as selective estrogen receptor modulators (SERMs) like tamoxifen, selective estrogen receptor degraders like fulvestrant, and aromatase inhibitors like anastrozole and ovariectomy.

A hormone-sensitive cancer, or hormone-dependent cancer, is a type of cancer that is dependent on a hormone for growth and/or survival. Examples include breast cancer, which is dependent on estrogens like estradiol, and prostate cancer, which is dependent on androgens like testosterone.

References

  1. Bray, Freddie; Ferlay, Jacques; Soerjomataram, Isabelle; Siegel, Rebecca L.; Torre, Lindsey A.; Jemal, Ahmedin (2018). "Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries". CA: A Cancer Journal for Clinicians. 68 (6): 398. doi: 10.3322/caac.21492 . PMID   30207593.
  2. 1 2 "Breast cancer in women". nhs.uk. 2017-10-20. Retrieved 2021-10-06.
  3. Akram, Muhammad; Iqbal, Mehwish; Daniyal, Muhammad; Khan, Asmat Ullah (2017). "Awareness and current knowledge of breast cancer". Biological Research. 50 (1): 33. doi: 10.1186/s40659-017-0140-9 . ISSN   0717-6287. PMC   5625777 . PMID   28969709.
  4. 1 2 Libson, Shai; Lippman, Marc (2013). "A review of clinical aspects of breast cancer". International Review of Psychiatry. 26 (1): 4–15. doi:10.3109/09540261.2013.852971. ISSN   0954-0261. PMID   24716497. S2CID   5951469.
  5. Brouckaert, O.; Paridaens, R.; Floris, G.; Rakha, E.; Osborne, K.; Neven, P. (2013). "A critical review why assessment of steroid hormone receptors in breast cancer should be quantitative". Annals of Oncology. 24 (1): 47–53. doi: 10.1093/annonc/mds238 . ISSN   1569-8041. PMID   22847811.
  6. Fahad Ullah, Mohammad (2019), Ahmad, Aamir (ed.), "Breast Cancer: Current Perspectives on the Disease Status", Breast Cancer Metastasis and Drug Resistance, Advances in Experimental Medicine and Biology, Cham: Springer International Publishing, vol. 1152, pp. 51–64, doi:10.1007/978-3-030-20301-6_4, ISBN   978-3-030-20300-9, PMID   31456179 , retrieved 2021-10-06
  7. Yip, Cheng-Har; Rhodes, Anthony (2014). "Estrogen and progesterone receptors in breast cancer". Future Oncology (London, England). 10 (14): 2293–2301. doi:10.2217/fon.14.110. ISSN   1744-8301. PMID   25471040.
  8. Chan, Hei Jason; Petrossian, Karineh; Chen, Shiuan (2016). "Structural and functional characterization of aromatase, estrogen receptor, and their genes in endocrine-responsive and -resistant breast cancer cells". The Journal of Steroid Biochemistry and Molecular Biology. 161: 73–83. doi:10.1016/j.jsbmb.2015.07.018. ISSN   1879-1220. PMC   4752924 . PMID   26277097.
  9. 1 2 Gruber, Christian J; Gruber, Doris M; Gruber, Isabel M.L; Wieser, Fritz; Huber, Johannes C (2004). "Anatomy of the estrogen response element". Trends in Endocrinology & Metabolism. 15 (2): 73–78. doi:10.1016/j.tem.2004.01.008. PMID   15036253. S2CID   12593038.
  10. 1 2 Moghadam, Sonia Javan; Hanks, Amanda M.; Keyomarsi, Khandan (2011). "Breaking the cycle: An insight into the role of ERα in eukaryotic cell cycles". Journal of Carcinogenesis. 10: 25. doi: 10.4103/1477-3163.90440 . ISSN   1477-3163. PMC   3243079 . PMID   22190867.
  11. 1 2 3 4 5 Fan, Weimin; Chang, Jinjia; Fu, Peifeng (2015). "Endocrine therapy resistance in breast cancer: current status, possible mechanisms and overcoming strategies". Future Medicinal Chemistry. 7 (12): 1511–1519. doi:10.4155/fmc.15.93. ISSN   1756-8919. PMC   5558537 . PMID   26306654.
  12. 1 2 3 4 Fuentes, Nathalie; Silveyra, Patricia (2019-01-01), Donev, Rossen (ed.), "Chapter Three - Estrogen receptor signaling mechanisms", Advances in Protein Chemistry and Structural Biology, Intracellular Signalling Proteins, Academic Press, 116: 135–170, doi:10.1016/bs.apcsb.2019.01.001, PMC   6533072 , PMID   31036290
  13. Behl, Christian (June 2002). "Oestrogen as a neuroprotective hormone". Nature Reviews Neuroscience. 3 (6): 433–442. doi:10.1038/nrn846. ISSN   1471-0048. PMID   12042878. S2CID   6292171.
  14. Martínez-Pérez, Carlos; Turnbull, Arran K; Dixon, J Michael (February 2019). "The evolving role of receptors as predictive biomarkers for metastatic breast cancer". Expert Review of Anticancer Therapy. 19 (2): 121–138. doi:10.1080/14737140.2019.1552138. hdl: 20.500.11820/a19d24db-2713-4a0b-abfb-f409bed96c75 . ISSN   1473-7140. PMID   30501540. S2CID   54565167.
  15. 1 2 Clarke, Robert; Skaar, Todd C.; Bouker, Kerrie B.; Davis, Natalie; Lee, Y.Richard; Welch, James N.; Leonessa, Fabio (March 2001). "Molecular and pharmacological aspects of antiestrogen resistance". The Journal of Steroid Biochemistry and Molecular Biology. 76 (1–5): 71–84. doi:10.1016/s0960-0760(00)00193-x. ISSN   0960-0760. PMID   11384865. S2CID   38235536.
  16. Peng, Jing; Sengupta, Surojeet; Jordan, V Craig (June 2009). "Potential of Selective Estrogen Receptor Modulators as Treatments and Preventives of Breast Cancer". Anti-Cancer Agents in Medicinal Chemistry. 9 (5): 481–499. doi:10.2174/187152009788451833. ISSN   1871-5206. PMC   3767174 . PMID   19519291.
  17. 1 2 Hanker, Ariella B.; Sudhan, Dhivya R.; Arteaga, Carlos L. (April 2020). "Overcoming Endocrine Resistance in Breast Cancer". Cancer Cell. 37 (4): 496–513. doi:10.1016/j.ccell.2020.03.009. PMC   7169993 . PMID   32289273.
  18. 1 2 3 4 5 6 7 8 9 AlFakeeh, A.; Brezden-Masley, C. (2018-06-01). "Overcoming Endocrine Resistance in Hormone Receptor–Positive Breast Cancer". Current Oncology. 25 (11): 18–27. doi:10.3747/co.25.3752. ISSN   1718-7729. PMC   6001756 . PMID   29910644.
  19. 1 2 3 4 5 6 7 8 9 10 Szostakowska, Małgorzata; Trębińska-Stryjewska, Alicja; Grzybowska, Ewa Anna; Fabisiewicz, Anna (November 2018). "Resistance to endocrine therapy in breast cancer: molecular mechanisms and future goals". Breast Cancer Research and Treatment. 173 (3): 489–497. doi:10.1007/s10549-018-5023-4. ISSN   0167-6806. PMC   6394602 . PMID   30382472.
  20. 1 2 3 4 5 D’Souza, Anishka; Spicer, Darcy; Lu, Janice (2018-06-11). "Overcoming endocrine resistance in metastatic hormone receptor-positive breast cancer". Journal of Hematology & Oncology. 11 (1): 80. doi: 10.1186/s13045-018-0620-6 . ISSN   1756-8722. PMC   5996460 . PMID   29891002.
  21. 1 2 3 Rondón-Lagos, Milena; Villegas, Victoria E.; Rangel, Nelson; Sánchez, Magda Carolina; Zaphiropoulos, Peter G. (August 2016). "Tamoxifen Resistance: Emerging Molecular Targets". International Journal of Molecular Sciences. 17 (8): 1357. doi: 10.3390/ijms17081357 . PMC   5000752 . PMID   27548161.
  22. 1 2 3 4 Milani, Andrea; Geuna, Elena; Mittica, Gloria; Valabrega, Giorgio (2014-12-10). "Overcoming endocrine resistance in metastatic breast cancer: Current evidence and future directions". World Journal of Clinical Oncology. 5 (5): 990–1001. doi: 10.5306/wjco.v5.i5.990 . ISSN   2218-4333. PMC   4259959 . PMID   25493235.
  23. 1 2 3 4 Hartkopf, Andreas D.; Grischke, Eva-Maria; Brucker, Sara Y. (2020). "Endocrine-Resistant Breast Cancer: Mechanisms and Treatment". Breast Care. 15 (4): 347–354. doi:10.1159/000508675. ISSN   1661-3791. PMC   7490658 . PMID   32982644.