Next Article in Journal
A Prospective Feasibility Trial to Challenge Patient–Derived Pancreatic Cancer Organoids in Predicting Treatment Response
Next Article in Special Issue
Hypnosis Sedation Reduces the Duration of Different Side Effects of Cancer Treatments in Breast Cancer Patients Receiving Neoadjuvant Chemotherapy
Previous Article in Journal
Socio-Economic Deprivation and Symptom Burden in UK Hospice Patients with Advanced Cancer—Findings from a Longitudinal Study
Previous Article in Special Issue
A Novel Three-Gene Score as a Predictive Biomarker for Pathologically Complete Response after Neoadjuvant Chemotherapy in Triple-Negative Breast Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Present and Future of Neoadjuvant Endocrine Therapy for Breast Cancer Treatment

by
Covadonga Martí
1,2,* and
José Ignacio Sánchez-Méndez
1,2,3,*
1
Breast Cancer Unit, Hospital Universitario La Paz, 28046 Madrid, Spain
2
Gynaecology Department, Hospital Universitario La Paz, 28046 Madrid, Spain
3
IdiPaz—Instituto de Investigación La Paz, 28046 Madrid, Spain
*
Authors to whom correspondence should be addressed.
Cancers 2021, 13(11), 2538; https://doi.org/10.3390/cancers13112538
Submission received: 8 April 2021 / Revised: 16 May 2021 / Accepted: 19 May 2021 / Published: 21 May 2021
(This article belongs to the Special Issue Neoadjuvant Therapy in Breast Cancer)

Abstract

:

Simple Summary

The treatment of breast cancer has evolved considerably over the last two decades, leading toward individualized disease management. Hormone-sensitive breast cancers constitute the vast majority of cases and endocrine therapy is the mainstay of their treatment. On the other hand, neoadjuvant or pre-surgical treatments provide a number of advantages for tumor management. In this review we will discuss the existing evidence on neoadjuvant endocrine therapy, as well as its possible future indications.

Abstract

Endocrine therapy (ET) has established itself as an efficacious treatment for estrogen receptor-positive (ER+) breast cancers, with a reduction in recurrence rates and increased survival rates. The pre-surgical approach with chemotherapy (NCT) has become a common form of management for large, locally advanced, or high-risk tumors. However, a good response to NCT is not usually expected in ER+ tumors. Good results with primary ET, mainly in elderly women, have encouraged studies in other stages of life, and nowadays neoadjuvant endocrine treatment (NET) has become a useful approach to many ER+ breast cancers. The aim of this review is to provide an update on the current state of art regarding the present and the future role of NET.

1. Introduction

ET is a key pillar in the treatment of ER+ tumors, since it has widely demonstrated its efficacy in improving survival and reducing recurrences. In recent years, the neoadjuvant or presurgical approach has established itself as a very useful strategy in breast cancer management, because it offers numerous advantages. In the first place, tumor downstaging may be achieved, thus increasing breast-conserving surgery (BCS) rates and, in some cases, reducing axillary dissection. On the other hand, assessing the in vivo response enables us to determine drug efficacy, as well as to study any biological or molecular changes that may lead us to explore new biomarkers. Finally, neoadjuvant treatment provides a unique opportunity for validating new treatments, alone or in combination, given that results can be obtained in short periods of time. These advantages have established neoadjuvant chemotherapy (NCT) as a widely accepted approach to estrogen receptor negative (ER−) tumors, but neoadjuvant endocrine therapy (NET) still remains an underutilized tool for ER+ breast cancers, and is frequently relegated to the treatment of elderly or frail patients who are not candidates for chemotherapy.
In this review we will describe the evidence that supports the current use of NET, as well as its potential for future expansion.

2. Patient Selection

As is the case with endocrine adjuvant therapy, NET should be considered for luminal-like tumors with ER expression. However, although ET is recommended for tumors even with low ER levels (i.e. <1%) [1,2], NET is normally used only with high ER expression tumors (All red score 7–8), since most trials have achieved better results in these cases [3,4]. Nevertheless, high-ER expression does not always predict a good response to NET. Other factors, such as progesterone receptor (PR) [5] or HER-2 expression, as well as histologic grade, may also influence results [6]. HER-2 overexpression generally discourages the use of NET [5,6], although a few studies (IMPACT, P024) included small subpopulations of HER-2 positive patients and reported over 50% of clinical responses, especially when aromatase inhibitors were employed [7,8].
Regarding histologic subtypes, most studies do not differentiate between ductal and invasive lobular breast cancers in their analysis of response, an exception being a 2011 report confined to lobular breast cancers and published by Dixon et al. [9]. It indicates very good responses in lobular breast cancers alone, with clinical response rates of almost 92% and a mean reduction in tumor volume after three months of 66%.

Menopausal Status

Most studies concerning NET are confined to postmenopausal women, and this leads most authors and guidelines to reject its use in premenopausal conditions [3,10,11]. Despite this fact, there is evidence of NET response in premenopausal women. The STAGE trial was a single phase III study that enrolled 197 premenopausal ER+/HER-2 negative breast cancer patients who were randomized to either anastrozole or tamoxifen, plus goserelin, for 24 weeks before surgery. The primary endpoint was tumor response assessed by caliper measurement, and this favored the anastrozole arm (70.4% vs. 50.5%), while also more patients in this group benefited from BCS. The authors concluded that anastrozole plus goserelin constitutes a safe choice for premenopausal women with early-stage ER+ breast cancer and can be considered an alternative neoadjuvant treatment [12].
The GEICAM/2006-03 study evaluated differences between NCT and NET in 95 patients with luminal breast cancer, 50% of whom were premenopausal (treated with goserelin plus exemestane). NET was found to be a useful strategy in this group, although responses were higher in the NCT group and this was not observed in postmenopausal women. The authors suggested that these differences might be due to the time it takes to achieve ovarian suppression (four weeks) [13].
Torrisi et al. published a small but very interesting study that included only premenopausal women with locally advanced operable luminal tumors. Patients were treated with letrozole plus triptorelin or goserelin for a median time of four months. Good clinical response (50%, 95% CI 32—68%) and a 50% rate of breast conserving surgery (BCS) were reported. A biological response, in terms of downregulation of the estrogenic signaling, was observed in all patients. The authors considered NET to be a safe alternative in premenopausal women when estradiol suppression is complete [14].
In conclusion, although evidence from clinical trials is scarce, results from other studies show that NET, combined with ovarian suppression, can be considered for premenopausal women, especially when treating luminal A-like tumors (strongly ER+, well differentiated, and with low proliferation rates).

3. Clinical Implications of NET

3.1. NET vs. NCT

Only three randomized clinical trials have compared NET with NCT. None of them involved a large number of patients, but they all reached similar conclusions.
Semiglazov et al. conducted a phase II study in which 239 postmenopausal patients with stage IIA–IIIB ER+ breast cancer were randomized for three months to either NET (exemestane/anastrozole) or NCT (doxorubicin + paclitaxel). Rates of pathological complete response (pCR) and disease progression did not differ significantly between the arms. BCS rates favored NET and toxicity was higher in the NCT group [15].
The Spanish GEICAM/2006-03 study randomized 95 patients with IHC-defined luminal disease to receive neoadjuvant exemestane for 24 weeks or chemotherapy (four cycles of epirubicin + cyclophosphamide followed by four cycles of docetaxel). Premenopausal women were included in this trial, and they were also treated with goserelin while receiving exemestane. No statistical difference was found between the two arms in terms of clinical response rate, but there was a trend toward a worse outcome in the exemestane arm for premenopausal patients and those with high tumor Ki67 expression [13].
Finally, the NEOCENT trial aimed to compare NCT (epirubicin, 5-fluorouracil, cyclophosphamide) vs. NET with letrozole in postmenopausal women with strongly ER+ breast cancers. Rates of clinical response and radiological response were similar in both arms. Unfortunately, only 44 patients were recruited for this trial, and this weakens the conclusions [16]. The characteristics of these studies are summarized in Table 1.
The meta-analysis published by Spring et al. [17] using data from the abovementioned trials indicated no significant difference in the clinical response rate (OR, 1.08; 95% CI, 0.50–2.35; p = 0.85; n = 378), radiological response rate (OR, 1.38; 95% CI, 0.92–2.07; p = 0.12; n = 378), pCR (OR, 1.99; 95% CI, 0.62–6.39; p= 0.25; n = 378), or BCS rate (OR, 0.65; 95% CI, 0.41–1.03; p = 0.07; n = 334). However, toxicity was significantly increased in the NCT arm in the three studies.
A recently published meta-analysis including these trials, together with other small studies that compared NCT and NET, found a higher pCR rate with NCT (pooled OR= 0.48; 95% CI, 0.26–0.90), whereas no significant difference in overall response rate (ORR) was observed between those who underwent these two treatment schemes (pooled OR = 1.05; 95% CI, 0.73–1.52) [18].
To sum up, NET does not offer any clear advantages in terms of clinical response or BCS rates when compared to NCT, but it definitely implies less toxicity and better tolerance, thus constituting a safe and effective option for ER+ breast cancers [17,18].

3.2. Effect of NET on Breast and Axilla Surgery

3.2.1. Increasing BCS Rates and Enhancing Surgery

NET was first employed in the early 1980s as a therapeutic alternative for elderly women who had inoperable tumors and were unfit for chemotherapy. The primary endpoint of these studies was to evaluate the efficacy of ET as an alternative to surgery, rather than as a neoadjuvant approach. Most of the authors reported response rates over 50%, despite higher locoregional relapse rates. These higher recurrence rates were probably due to the fact that most of these patients did not undergo subsequent surgery, although this did not impact overall survival [19,20,21].
More recent trials involving NET have also demonstrated its value in achieving higher rates of breast conserving surgery and, therefore, an improved cosmesis outcome. In those studies with a treatment duration of more than three months, BCS likelihood can range between 40–80% when using aromatase inhibitors [22,23]. According to Montagna et al. [24], up to 77% of noneligible BCS patients were candidates for breast preservation after NET, although this rate was lower in those with low PR expression.
In parallel with the promotion of BCS, NET has shown itself to be an appropriate alternative for locally advanced, sometimes inoperable, ER+ tumors, [25,26,27] especially with older patients or low grade, luminal A-like tumors. Hence, NET provides a very helpful and safe tool for treating cases with tumors over 2 cm, when there is discordance between tumor and breast size, or in inoperable cases of locally advanced ER+ breast cancers [4].

3.2.2. Axilla Management after NET

It is well established that, after NCT, ER+/HER-2 negative tumors do not achieve a good response in both breast and axilla, with pCR rates below 15% [28,29]. Unfortunately, although axillary-positive patients have been included in most NET trials, very little has been concluded regarding changes brought about by endocrine treatment at this level. Most authors find that NET is less likely to de-escalate surgery in the axilla than in the breast, even though pCR rates range from 1.3% to 11% [24,30]. A slight increase in these rates was observed by Rusz et al., reaching 13% node pCR in N+ patients after one year of NET with letrozole [31]. In a recent review, Stafford et al., after analyzing 4580 ER+ N+ breast cancers from the National Cancer Database, concluded that nodal pCR could be achieved in up to 14.5% of patients [32]. As reported by other authors, lobular breast cancer had a significantly lower percentage of nodal pCR when compared to ductal histologies (12.4% vs. 15.26%, p < 0.05) [32,33]. Longer treatment duration (>6 months) seems to increase the likelihood of nodal pCR [24,31].
Axillary management after NCT has been a controversial issue in the past few years, mainly because of the concern that disease may be left behind if an axillary dissection (AD) is not performed. Three trials have proven sentinel node biopsy (SLNB) to be a safe technique when cN1 axilla changed to cN0 after NCT [34,35,36], and this has brought about a widespread change in practice patterns, with a general avoidance of AD when a complete response is presumed. Nevertheless, data regarding surgical management of the axilla after NET is limited. As previously stated, good responses in the axilla after NET are rarely expected, so this might create more difficulties when carrying out a SLNB. Paradoxically, most surgeons seem to apply the same criteria to NET as they do to NCT [37]; however, the context of NCT and, above all, of the high-risk tumors treated in this way, does not seem applicable to NET, since any low-burden disease left in the axilla after NET may not significantly impact the prognosis, as pointed out by Kantor et al. [37,38] These authors concluded that axillary management strategies employed with upfront-surgery patients may also be used with NET patients, which would mean that Z0011 criteria for avoiding AD may be safely applied.
In conclusion, axillary response is rather uncommon after NET, although the prognostic significance of axillary burden after treatment is not different from that of patients managed with upfront surgery.

3.2.3. Surgery Avoidance in Frail Patients

As mentioned above, the initial studies of NET were developed among elderly and/or frail women cohorts. Tamoxifen then arose as a possible alternative to surgery, with clinical responses in over 50% of the patients. As most of the cases did not complete the treatment with surgery, a large number of recurrent events was reported, although this did not affect survival [17,18,19]. In 2007, the Cochrane Library published a review comparing surgery, with or without tamoxifen, with tamoxifen alone for older women with operable breast cancer. When surgery with adjuvant endocrine therapy was compared to endocrine therapy alone, there was no significant difference in overall survival (HR: 0.86), but there was a significant difference in progression-free survival (HR 0.65) for surgery plus endocrine therapy vs. endocrine therapy alone [39]. Later studies using aromatase inhibitors have yielded similar results [40,41], with no survival benefit in spite of an increase in risk progression. In the review performed by Morgan et al. [42], randomized and nonrandomized controlled trials were included that recruited women over 70 years of age with operable breast cancer and treated with NET. A long median time to progression (~49 months), and a quite long duration of clinical benefit (~30 months) were reported, which, together with its low toxicity, make primary endocrine therapy a useful treatment strategy in selected patients. The authors concluded that NET can be an option in cases of short life expectancy (<2 years) and, given their greater efficacy, aromatase inhibitors should be preferred over tamoxifen [42]. Tumor cryoablation or surgery under local anesthesia can be considered in some cases when a tumor reduction accomplished with NET makes a less extensive resection possible [43,44,45].
In other words, although surgery avoidance should not be considered the standard of care, it can be contemplated in the case of a specific group of elderly or frail patients with ER+ breast cancer who offer considerable possibilities of response or disease stabilization with NET.

4. Choosing the Best Endocrine Agent

4.1. Tamoxifen versus Aromatase Inhibitors

The third-generation aromatase inhibitors (AIs), anastrozole, letrozole, and exemestane, are nowadays considered the standard adjuvant treatment for women with ER+ breast cancer because of their advantages over tamoxifen, and so studies have been conducted in order to confirm their superiority in the neoadjuvant setting.
Four phase III randomized trials have studied the differences between tamoxifen and AIs in that setting (Table 2). Three of these studies were conducted in postmenopausal women and one in premenopausal women.
P024 was a four-month, double-blind randomized trial that compared letrozole 2.5 mg with tamoxifen 20 mg in the preoperative treatment of 337 postmenopausal women who had ER+ locally advanced breast cancer and were ineligible for BCS. Letrozole showed a higher objective response (55% vs. 36%, p = 0.0001), as well as a better BCS rate (45% vs. 35%, p = 0.02) [7].
Two trials compared anastrozole with tamoxifen. The IMPACT study randomized 330 postmenopausal women with operable or potentially operable ER+ breast cancer to receive either tamoxifen 20 mg, anastrozole 1 mg, or a combination of both [8]. No differences in objective response were found among the three arms, although BCS rates were double in the anastrozole arm in patients who were deemed eligible for BCS by their surgeons (46% vs. 22%, p = 0.03). No benefit was obtained from the combination of letrozole and tamoxifen. The three-month PROACT trial compared anastrozole vs. tamoxifen in 451 postmenopausal women with ER+ large, operable (T2-3/N0-2) tumors, with or without chemotherapy [46]. Improvement in feasible surgery at baseline to actual surgery at three months was found to be numerically higher in the anastrozole group compared with the tamoxifen group, although this difference did not reach significance. However, significant differences were observed favoring the anastrozole arm in BCS rates of patients who did not receive chemotherapy (43% vs. 30.8% p = 0.04).
Finally, exemestane efficacy was also reported by Semiglazov in a randomized trial with 151 postmenopausal women [47]. Tamoxifen or exemestane were given preoperatively for three months. Significant differences were found in terms of objective response (exemestane 76% vs. tamoxifen 40%, p = 0.05) and in BCS (37% vs. 20%, p = 0.05), with a shorter response observed in the exemestane arm (57 days vs. 70 days, p < 0.05).
A meta-analysis of these trials demonstrated that preoperative use of AIs was more effective than tamoxifen. Pooled results for clinical efficacy revealed a clinical response rate (OR, 1.69; 95% CI, 1.36–2.10; p < 0.001), radiological response rate (OR, 1.49; 95% CI, 1.18–1.89; p < 0.001), and BCS rate (OR, 1.62; 95% CI, 1.24–2.12; p < 0.001) favoring Ais [17].
The only study comparing AIs with tamoxifen in the premenopausal condition was the aforementioned STAGE trial. This showed anastrozole to be superior to tamoxifen when both were combined with goserelin in premenopausal women with ER+ breast cancer [12]. Therefore, AIs constitute a first-choice option in neoadjuvant therapy, just as they already do in adjuvant treatment.

4.2. Comparison of Aromatase Inhibitors

The Z1031 trial is the only one to have compared the three third-generation AIs. This was a phase II study that randomized 377 postmenopausal women with clinical stage II/III ER+ breast cancer to receive an AI (anastrozole, letrozole, or exemestane) for four months before surgery [48]. The authors concluded that neoadjuvant AI treatment markedly improved surgical outcomes with no significant differences in clinical, biological, or surgical outcomes among the three agents. It is worth highlighting that half of the patients that were candidates for mastectomy benefited from BCS after NET. Consequently, the three commercially available AIs are all equally effective in neoadjuvant therapy, as is the case when they are used in adjuvant and metastatic treatment.

4.3. Selective Estrogen Down -Regulators versus Aromatase Inhibitors

Fulvestrant has been compared to anastrozole in two trials. Quenel-Tueux et al. randomized 108 postmenopausal women who had ER+ breast cancer and were not initially eligible for BCS to receive 500 mg fulvestrant or 1 mg anastrozole for six months [50]. No significant differences were found between the drugs, with similar objective responses (58.9% anastrozole vs. 53.8% fulvestrant) and BCS rates (58.9% in the anastrozole arm vs. 50.0% for fulvestrant).
With very similar results, the phase II UNICANCER CARMINA trial compared fulvestrant with anastrozole in 116 postmenopausal women with ER+/Her-2 negative, operable breast cancer [49]. Clinical response rates at six months were 52.6% (95% CI, 41–64%) for anastrozole and 36.8 % (95% CI, 25–49%) for fulvestrant. BCS was performed in 57.6% in the anastrozole arm and 50% in the fulvestrant arm. Relapse-free survival rates at three years were comparable (94.9% for anastrozole and 91.2% for fulvestrant). Both agents demonstrated effectiveness and were well tolerated, although results were slightly better with anastrozole.

5. Treatment Duration

NET duration is usually established at 3–6 months, based on the regular duration of NCT, although the optimal period for this therapy is not yet clearly determined. Several trials have been conducted with the aim of defining an appropriate duration (Table 3). Most of these studies found that a prolongation of NET produced extra benefits [31,51,52,53,54,55,56].
The first study was published by a German group led by Krainick-Strobel et al., concerning a cohort of 32 postmenopausal patients who received letrozole for a period of four months (early responders) to eight months (slow responders) [51]. At diagnosis, all patients were unsuitable for BCS, but the majority achieved sufficient response for BCS after four months, although the objective response rate reached 72.4% after eight months. Similarly, Dixon et al. compared letrozole for periods of three or more than three months [52]. After three months almost 70% of the patients had responded and 60% of them received BCS. With a longer treatment (>3 months), tumor downstaging reached 83.5%, and BCS rates 72%. A Spanish phase II trial recruited 70 postmenopausal women treated with letrozole for 3–12 months [53]. 6.8% of the patients achieved an objective response, 51.8% being partial and 25.0% complete. The median time to objective response was 3.9 months (95% CI, 3.3–4.5) and the median time to maximum response was 4.2 months (95% CI, 4.0–4.5), although 37.1% patients achieved the maximal response within 6–12 months. Despite these results, BCS rates reached only 43%. Another study, one that continued NET up to 12 months, stated that the optimal duration for obtaining BCS was a median of 7.5 months [54]. One Italian trial randomized 120 postmenopausal ER+ breast cancer patients to receive letrozole for 4, 8 or 12 months [56]. Clinical response, as well as pCR and BCS rates, were analyzed. After four months, only 2.5% pCR was observed, although BCS was feasible in 80% of the patients. After 12 months, pCR rates reached 17.5% and almost 90% of the women benefited from BCS. Slightly lower pCR rates (14.3%) are published by Rusz et al. [31], after a one-year treatment with letrozole, although they report relatively modest BCS rates (45%). Consequently, low pCR rates attributed to NET may be due to treatments that are not sufficiently long.
Only two trials have assessed the ideal duration of NET employing exemestane. The study conducted by Fontein et al. (TEAM IIA) [55] reported a significant increase in objective response rate with longer treatments (58.7% <3 months vs. 68.3% >3 months, p = 0.031), as well as an improvement in the feasibility of BCS (61.8% vs. 70.6%, p = 0.012). On the other hand, the Japanese PTEX46 trial [57] did not find significant differences between treating with exemestane for four months or six months, either in objective response rates (42.3% four months vs. 48% six months, p = 0.89) or in BCS rates (50% four months vs. 48% six months).
Because of this variability, clinical guidelines and expert consensus do not specify a recommended duration for NET. It was only at the 2013 St. Gallen Conference that the majority of the panelists supported the use of NET until maximal response [58].
Overall, even though the findings of the studies are mixed, it can be concluded that good responses and BCS rates can be achieved with a NET duration of six months or less, but higher pCR rates and objective responses may be obtained with up to one year of treatment.
A major concern raised when NET is maintained for long periods is the possibility of disease progression due to endocrine resistance. Studies of elderly and/or frail women with ER+ breast cancers who had been treated exclusively with tamoxifen found no differences in overall survival when compared with women treated with surgery, although regional control was poorer in the nonsurgical cohort [59,60].
Interestingly, several of these trials found no difference at all between the surgical and nonsurgical cohorts for the first 18–24 months. Willsher et al. also observed that a partial or complete response at six months was a strong predictor of excellent control at three years [61], especially with highly ER+ tumors. Hence, with an exclusively NET approach, resistance is unlikely to appear during the first year. Beyond 12 months, NET can be maintained in selected cases of elderly women [26], in particular those with a documented early tumor response.

6. Monitoring Response to NET

For their primary analyses, adjuvant trials typically employ survival goals (disease-free or breast cancer mortality). However, in the neoadjuvant setting the response criteria are based on the dynamic changes produced during treatment. These may be changes in surgical outcome, tumor size, or histopathological characteristics after therapy.

6.1. Assessing Response by Imaging

As with NCT, clinical examination and radiological control are of great importance. The combination of both clinical exploration and imaging constitutes the usual method for assessing response. Unfortunately, the precision of clinical breast examination for determining pCR in patients with locally advanced breast cancer after NET or NCT is only 57%, which is inferior to mammography (74%) and ultrasound (79%) [62], mainly because of the clinical changes (post-therapy fibrosis, undefined tumor margins) produced by treatment. A combination of mammography plus ultrasound (US) appears to be a very reliable way of checking response (80% likelihood) [63], although little has been published concerning these two techniques and NET evaluation.
Based on experience with NCT, many authors recommend MRI as the best procedure for assessing NET response, since it is a dynamic technique [63,64,65,66] that enables changes within the tumor to be verified. Estimating final pathology size after treatment is crucial when employing neoadjuvant therapies, since it allows a surgical approach to be planned. Correlation between post-treatment size and final pathology size is better with MRI than with clinical exploration [67], but no studies comparing MRI accuracy with US or US-mammography are available in the NET setting, although in the NCT scenario some authors have found US to be as accurate as MRI [68]. On the other hand, MRI could provide not only data on responsiveness to therapy but could also predict which patients would benefit most from it. In this sense, minimal or mild background parenchymal enhancement was described as a predictive factor of good response to NET in a small sample published by Hilal et al. [64]. Changes in contralateral breast parenchymal enhancement on MRI during NET might also differentiate between patients with a good prognosis and those with an intermediate/poor prognosis at final pathology [69].
The introduction of novel quantitative functional imaging, such as advanced MR or radionuclide imaging, may be also useful for evaluating changes caused by therapy response [63]. Metabolic changes produced by NET can be assessed using FDG-PET/CT scans. These have been proven to be a helpful tool since there is a correlation between tumor SUVmax and cell-cycle response (Ki67 levels) [70,71] and they may even be considered as a surrogate marker for monitoring tumor response [71]. Nevertheless, there are some limitations to this technique, as low-proliferation tumors (luminal A-like, lobular), which otherwise are the best candidates for NET, have a lower glucose metabolism [72] and this may affect the interpretation of the response. An interesting alternative would be to employ 18F-fluoroestradiol (18F-FES) as radionuclide [73,74,75]. 18F-FES PET/CT can measure the in vivo binding of estrogens and thus can be used to assess the biological activity of ER. Earlier trials reported a high positive and negative percentage agreement of 18F-FES PET (87% and 91%, respectively), with ER determined by immunohistochemical testing [76]. Other studies have indicated that FES avidity can be a pharmacodynamic biomarker for ER-directed therapy and could therefore predict a better response to NET and no benefit from NCT [74]. Unfortunately, these novel radiotracers are still not available in most centers and remain at the research stage, although they look very promising as an option in the NET setting. The combination of techniques such as MR imaging and FDG PET-TC, which are available in only a few facilities, might offer advantages for primary breast cancer therapy response assessment [63,73], yet further investigation is required in this field.
Therefore, monitoring NET response may be performed reasonably well with traditional procedures like US or US-mammography, although dynamic or functional techniques can provide greater accuracy in this evaluation.

6.2. Pathological Complete Response

Pathological complete response is a validated measure of long-term outcomes in NCT studies, mainly in biologically aggressive phenotypes such as triple negative or HER-2 positive breast cancers [77]. Nevertheless, pCR rates in ER+ breast cancers are very low with both NCT and NET [17], although this does not impact survival as much as happens with non-luminal tumors [77]. Consequently, pCR is not an optimal indicator of outcomes and other biomarkers must be used when validating NET.

6.3. Ki67

Ki67 is a nuclear antigen expressed in proliferating tissues, in all cell-cycle phases. It is absent in quiescent cells, and correlates well with other markers of proliferation such as the S-phase fraction or mitotic index, [78] thus being considered a surrogate marker of cell proliferation. Neoadjuvant treatments can induce a reduction in Ki67 value, and such changes have been identified only a few days after the initiation of NET [8,79], indicating a cell-cycle arrest induced by therapy [10]. But in addition, the changes that appear in Ki67 with NET constitute a biomarker with prognostic and predictive power [8,12,44,46], much more precisely related to long-term results than baseline Ki67 [80]. A Ki67 > 10% after two or four weeks of endocrine therapy has been suggested as a cutoff for early identification of non-responders with increased risk of relapse [81], and it is actually considered by many authors in various trials as the necessary threshold for maintaining or withdrawing NET [79,81,82,83,84,85]. Early Ki67 changes produced by NET served as the basis for the trial of PeriOperative Endocrine Therapy: Individualizing Care (POETIC) [79]. This “window of opportunity” study, with over 4300 women recruited, randomized patients to receive either an AI for two weeks before and two weeks after surgery, or no treatment at all. Although this short-course treatment did not improve outcomes, it clearly identified three groups of patients: those with low Ki67 levels before and two weeks after treatment, with a five-year recurrence risk (RR) of 4.3% (95% CI, 2.9–6.3); those whose Ki67 was initially high but became low after two weeks, with a RR of 8.4% (95% CI, 2.9–6.3); and finally, those with permanently high Ki67 scores (RR: 21.5% 95% CI, 17.1–27.0). The authors concluded that patients with low Ki67 or low post-AI-induced Ki67 probably do well enough with standard endocrine therapy, whereas those with high Ki67 scores might benefit from further adjuvant treatments. Interestingly, tumors that do not lower Ki67 after 2–4 weeks also fail to respond well to chemotherapy, indicating an intrinsic endocrine resistance, which makes it essential to consider alternative therapies [81]. The PerELISA trial also found that a Ki67 reduction could also serve to identify subgroups of ER+/HER-2 + patients who had a better prognosis, and for whom ET plus anti-HER-2 treatments, without chemotherapy, could be sufficient [86].
Ki67 presents certain weaknesses as a biomarker, such as intratumor heterogeneous expression, perhaps underrepresented in the biopsy, as well as interobserver variability when quantifying Ki67 staining [87]. Regardless of this, the clinical validity of Ki67 is fairly well established, and it is a useful and accessible tool that may make it possible to distinguish between endocrine sensitive or resistant tumors [87] and, in addition, determine a prognosis.

6.4. PEPI Score

Data from the follow-up of the P024 trial enabled Ellis et al. to develop a prognostic model that incorporated standard pathologic staging variables and “on-treatment” biomarker values. In the multivariable analysis, the four factors that showed a significant impact on survival (either relapse-free survival or breast cancer-specific survival) were: pathological tumor size, pathological node status, ER status, and Ki67 natural log intervals—all derived from the surgical specimen analysis. They were later reanalyzed to derive a final hazard ratio associated with each factor. This scoring process generated three risk groups (risk score 0, 1–3, and ≥4) that were associated with relapse risks of 10%, 23%, and 48% (p < 0.001) and breast cancer deaths of 2%, 11%, and 17% (p < 0.001). This was named the Preoperative Endocrine Prognostic Index (PEPI score (Table 4) [88]. An independent validation of this model was later performed with data from the IMPACT trial, with more favorable outcomes being shown in patients with T1 tumors and a PEPI score of 0; i.e., with a Ki67 score of less than 2.7% and ER expression. After two weeks, the PEPI score may be considered a more robust marker of response than Ki67 in some tumors, especially those with low pretreatment Ki67 or with a Ki67 rebound after the first 2–4 weeks [87]. With specific regard to its clinical application, the PEPI approach could be analogous to pCR with an escalation of therapy for those patients with PEPI > 0 and a de-escalation (avoiding chemotherapy) for patients with PEPI-0 status [82]. A modified PEPI score, including primary PR expression (PEPI-P), was described by Kurozumi et al., with better correlation with breast cancer-specific survival and relapse-free survival than PEPI alone [89]. One inconvenience of the PEPI score is that nodal involvement is a qualitative measurement, so a positive axilla always implies a PEPI score ≥ 3, regardless of whether one or more than three nodes are involved, and so an equivalence between PEPI ≥ 3 and chemotherapy should not always be categorically assumed.

6.5. Other Biomarkers

Although Ki67 and the PEPI score are useful and available in clinics, more accurate biomarkers are being investigated. In a small prospective French study, high levels of the ZNF217 gene were found to be significantly related to reduced responses to NET and independent of Ki67 expression. [90] In another study, a higher expression of the nonproliferative genes KRAS, CUL2, FAM13A, ADCK2, and LILRA2 was significantly associated with tumor shrinkage, and KRAS, MMS19, and IVD were related to a lower PEPI score (≤3) and, again, this gene expression had no correlation with Ki67 reduction [91]. This seems to indicate that gene expression may eventually provide a more accurate biomarker; however, more research is still needed.

7. NET and Targeted Therapies

Good results with targeted therapies combined with ET in locally advanced or metastatic ER+ disease have encouraged the development of trials in the neoadjuvant setting, although outcomes in this scenario have been mixed (Table 5).
The combination of phosphatidylinositol 3-kinase (PI3K) inhibitors with aromatase inhibitors has not yielded the expected results. The LORELEI trial recruited 334 patients with early-stage ER+ breast cancer, randomized to letrozole + placebo or to letrozole + taselisib. A slight significant difference was found in terms of objective response rates in the taselisib arm (50% vs. 39%, p = 0.049), but no significance was observed concerning pCR (1.8% in letrozole + taselisib vs. 0.9% in letrozole + placebo) [98]. Results in the NEO-ORB study, which randomized 257 patients to letrozole + placebo or letrozole + alpelisib, were even more disappointing. Addition of alpelisib to 24-week neoadjuvant letrozole treatment did not improve response in patients with HR+ early breast cancer with respect to pCR, objective response rates, and Ki67 reduction [93].
Good results in the HER-2+ population encouraged the study of the role of Tyrosine Kinase inhibitors in ER+/HER-2 negative. Guarneri et al. conducted a randomized trial that recruited 92 women to receive letrozole plus a placebo or letrozole plus lapatinib [92]. No differences were observed concerning clinical response (63% in the placebo arm and 70% in the lapatinib arm), but significant results were found in the presence of PIK3C mutation (objective response rate, 93% vs. 63% in PIK3C wild-type, p = 0.040).
The addition of CDK4/6 inhibitors (palbociclib, ribociclib, and abemaciclib) to NET has been studied in six different trials, with similar results. [94,95,96,99,100,101] The MONALEESA study was a very small (only 14 patients) phase II “window of opportunity” trial that compared Ki67 results after 14 days with letrozole or letrozole + ribociclib (400/600 mg) [101]. A greater Ki67 reduction was observed in the ribociclib arm (69% reduction with placebo vs. 96% and 92 % with ribociclib 400 and 600 mg), as well as a decreased expression of Cyclin D-cyclin-dependent kinase genes. In three trials that combined palbociclib or abemaciclib with NET (NeoPalAna, PALLET, and NeoMONARCH) [94,99,100], a higher complete cell-cycle arrest was observed in the CDK4/6 arm, although little or no benefit was found with regard to pCR. The two remaining trials compared the combination of letrozole with either palbociclib (NeoPAL) [95] or ribociclib (CORALLEEN) [96] with different chemotherapy regimens. The NeoPAL trial randomized 106 patients to either letrozole + palbociclib for 19 weeks or to NCT (5FU 500 mg/m2, epirubicin 100 mg/m2, cyclophosphamide 500 mg/m2 × 3 cycles followed by docetaxel × 3 cycles). Although pCR and Ki67 geometric mean reduction were slightly larger in the CT arm (5.9% vs. 3.8%), the PEPI 0 score was higher in the letrozole-palbociclib subgroup. No differences were observed in terms of BCS rates (69% in both arms). Because of the similarities between the arms, the authors concluded that NET + CDK4/6 inhibitors may represent a safer and less toxic alternative for high-risk ER+ tumors. In the Spanish CORALLEEN trial, 106 patients were randomized to letrozole-ribociclib for six months or to NCT (doxorubicin-cyclophosphamide x 4 followed by weekly docetaxel × 12). The primary endpoint of this study was to evaluate the proportion of patients with PAM50 low risk-of-relapse (ROR) disease at surgery. Most patients belonged to a high-ROR group at baseline (85% and 89%), and at surgery 46.9% (95% CI, 32.5–61.7) of patients in the ribociclib-plus-letrozole group and 46.1% (32.9–61.5) of patients in the chemotherapy group were low ROR; therefore, some patients with high-risk, early-stage ER+ breast cancer could achieve a molecular downstaging with CDK4/6 inhibitors and endocrine therapy.
Finally, the combination of NET with mTOR inhibitors such as everolimus was described by Baselga et al. [97] in a phase II trial in which 270 patients were randomly assigned to receive letrozole + placebo or letrozole + everolimus over four months. Response rate by clinical palpation in the everolimus arm was non-significantly higher than with letrozole alone (68.1% vs. 59.1% p = 0.06). Ki67 reduction was also better in the everolimus group (57% vs. 30% p < 0.01). No differences in pCR were observed and with evrolimus up to 22.6% of patients presented toxicity (grade 3–4 adverse events).
In conclusion, adding targeted therapies to NET frequently achieves a better cell-cycle arrest and Ki67 drop, although the clinical benefit seems meager. The addition of CDK4/6 inhibitors to NET in high-risk ER+ breast cancer may be comparable to NCT in terms of clinical value and molecular downstaging. In light of the risk-benefit balance, mTOR inhibitors do not appear to be an alternative, due to their toxicity. In any case, further studies are needed, mainly involving long-term survival outcomes.

8. The Role of Genomic Assays in NET

Genomic assays have become a very useful tool for guiding adjuvant treatments in early ER+ breast cancer. The possibility of performing these assays in the core biopsy increases their utility to the neoadjuvant setting, and most of them have already demonstrated their capacity for predicting NCT response [102,103,104,105,106,107,108,109]. We will now review those that have been investigated in the context of NET.

8.1. MammaPrint®/BluePrint®

MammaPrint®, a 70-gene signature, was the first validated genomic assay and it has been largely used to guide treatments in the adjuvant scenario [110]. BluePrint® is a genomic profile that studies the expression of 80 additional genes, and, combined with MammaPrint®, makes it possible to establish a molecular classification of the tumor into the luminal A, luminal B, Her-2, or basal type [111]. The NBRST trial, with over 420 ER+/Her-2 negative patients, demonstrated that the MammaPrint®/BluePrint® tandem was more accurate than immunohistochemistry in predicting response to either NCT (with a higher response rate in the basal and Her-2 molecular subtypes) [111] or NET (68% of clinical response to AI in the luminal subtypes) [112]. Hence, luminal breast cancer patients diagnosed by Mammaprint®/BluePrint® could be considered for NET, and those with luminal A subtypes have a favorable prognosis. At all events, further prospective research is required.

8.2. Oncotype DX

The Oncotype DX® Breast Recurrence Score (RS) assay is a 21-gene validated genomic profile. It was first developed to assess recurrence risk for patients with early ER+/HER-2 negative breast cancer treated with tamoxifen [113] and was initially applied only to node-negative patients, but more recently it has been used regardless of node status [114,115]. The RS stratifies patients into three risk categories: low (<18), intermediate (18–30), and high (>31). In postmenopausal women, a low or intermediate RS profile implies no benefit from additional chemotherapy, while high RS tumors have better outcomes with it [116]. In the neoadjuvant setting, performing Oncotype DX® in the core biopsy has proven to be a useful tool for choosing the best primary therapy so, in those patients with low-intermediate RS, NET might be an effective strategy [108,117]. In a Japanese study by Ueno et al., correlation between RS and NET with exemestane was investigated. Exemestane was given for 24 weeks to 102 patients and RS was analyzed in both the core biopsy and the surgical sample. The clinical response rate in patients with a low or intermediate RS was significantly higher than in those with a high RS (59.4% and 58.8% vs. 20.0% p = 0.015). Differences were also observed in BCS rates, which were higher in the low (90.6%) and intermediate (76.5%) RS cases and lower in the high RS group (46.8%, p = 0.028) [118]. The phase III TransNEOS trial evaluated data from 295 postmenopausal ER+, node-negative breast cancer patients treated primarily with letrozole for six months [117]. The primary endpoint was to establish differences in clinical response rates according to RS (low vs. high). A larger proportion of patients with low RS had clinical responses (54%), compared to those with high RS (22%, p < 0.001). In multivariable analyses, a continuous RS result was associated with clinical response (p < 0.001). An RS <18 was also associated with better BCS rates (p = 0.010). These results confirmed the capacity of this genomic profile to predict the clinical response to neoadjuvant letrozole. Therefore, Oncotype DX® RS has proved itself to be a very helpful tool for determining which neoadjuvant treatment is better with regard to RS, the lower scores being more suitable for NET and the higher ones (>31) better candidates for NCT.

8.3. EndoPredict®

EndoPredict® (EP) is a 12-gene genomic assay that provides prognostic information on the risk of a distant recurrence in breast cancer patients [119], and it is valid for both negative- and positive-node status. EP prognostic power has been improved by creating EPclin: a risk score that incorporates pathological data like tumor size or nodal status. However, in the neoadjuvant setting, only the molecular analysis (EP) is possible. Dubsky et al. analyzed 217 samples of diagnostic core biopsies from the ABCSG-34 trial [106], in which women had already been selected to receive NCT or NET according to clinical or immunohistochemical criteria. Tumors with a low molecular score (MS) were unlikely to benefit from NCT (NPV 100% (95% CI, 66.4%–100%)), whereas a high MS predicted resistance to NET (NPV 92.3% (95% CI, 79.1%–98.4%)). EP MS predicted a residual cancer burden after treatment with neoadjuvant therapies for patients with HR-positive, HER2-negative early-stage breast cancer. On the other hand, this genomic assay may also have a part to play in estimating prognosis after treatment with CDK4/6 inhibitors. In the N007 study, the aim of the authors was to evaluate PEPI score, pathologic response, and gene expression on the tumor samples of 20 patients who were treated for 24 weeks with letrozole + Palbociclib [120]. EPclin was found to be a better parameter than the PEPI score for estimating a prognosis, since some of the high PEPI-score risks had a low-risk EPclin. Furthermore, changes in gene expression after letrozole + palbociclib were reported, implying that not only proliferative activity was reduced but also immune signaling was modified. The biological information provided by EndoPredict® can therefore facilitate patient selection for neoadjuvant treatments, and EPclin might be a more accurate biomarker than PEPI score, especially in NET treatments combined with CDK4/6 inhibitors. In any case, additional studies will be necessary in order to confirm these results.

8.4. A Four-Gene Model

Turnbull et al. developed a four-gene model from pretreatment and on-treatment samples of 89 postmenopausal women who were receiving NET with letrozole [121].
With an accuracy of 96% for predicting response status, this model was created on the basis of the level of two genes before treatment (IL6ST, associated with immune signaling, and NGFRAP1, related to apoptosis) and on the levels of two proliferation genes (ASPM, MCM4) after two weeks of treatment. A further independent validation was performed in a similar cohort treated with anastrozole, with 91% accuracy. The predictive power was increased when the two-week on-treatment biopsies were added to the pretreatment biopsies. The four-gene signature also significantly predicted recurrence-free survival (p = 0.029) and breast cancer–specific survival (p = 0.009), and it is feasible that the results can be obtained by means of PCR and immunohistochemistry techniques. This approach offers the advantage of taking into account the dynamic changes produced by NET, suggesting that a full standard course of several months of neoadjuvant treatment may not be required, and a short two-week pre-surgical therapy might be sufficient to improve the accuracy of long-term predictions of outcomes [122]. Nevertheless, these results are based on retrospective analysis, so larger prospective studies should be conducted.

8.5. Other Gene Expression Profiles

Several research groups have described different forms of gene expression related to NET response. The Italian group led by Mello-Grand et al. defined an expression signature of 54 genes that predicted response in 17 patients treated with anastrozole for three months [123]. In responding tumors, down-regulated genes after treatment were mainly related to cell cycle, many of them transcriptionally activated by ESR1 (estrogen receptor1), whereas up-regulated genes were associated with immune response, histamine metabolism and inflammation, cell adhesion, and cytoskeleton. Nonresponding tumors showed an increased immune response as well as androgen receptor nuclear signaling, and this might explain endocrine resistance. Five of these genes were found to predict response on an independent dataset of 52 breast cancers treated with letrozole (p = 0.0056). Miller et al. developed a 99-gene signature that evaluated long-term estrogen-deprived (LTED) gene expression [124]. The genes up- or down-regulated in LTED cells were expressed at higher or lower levels, respectively, in tumors where cell proliferation was not suppressed by letrozole. These results were later validated in another dataset of patients treated with anastrozole. The LTED gene expression signature was found to be predictive of high tumor-cell proliferation following neoadjuvant therapy with anastrozole and letrozole, therefore making it possible to identify prospectively patients at risk of early recurrence during or following ET. Xu-Liang et al. performed gene expression profiling on 86 pre-NET and post-NET tumor samples from the CARMINA trial, which randomized patients to either letrozole or fulvestrant [125]. In NET responders, involved immune-associated genes enriched in activated Th1 pathway were found, but these remained unchanged in nonresponders. Gene expression also revealed that lipid metabolism was the main molecular function related to prognosis. Finally, the cell cycle-apoptosis pathway and the PIK3CA/AKT/mTOR pathway were altered significantly more frequently in nonresponders than in responders. Inda et al. have recently described an ER pathway activity score (ERPAS) involving the expression of 27 high-evidence target genes of the ER transcription factor in pre- and post-treatment tumors treated with letrozole, exemestane, or fulvestrant [126]. One third of the ER+ breast cancers presented an inactive ERPAS at baseline. Highly respondent tumors had a higher pretreatment ERPAS and a greater magnitude of decrease after two weeks of therapy. As a result, this score may serve to identify ER+ tumors that will not respond to ET.
To sum up, genomic profiling performed on diagnostic core biopsies as well as on early on-treatment biopsies may become a very useful tool for enabling NET response or endocrine resistance to be predicted. Larger prospective studies will be necessary in order to establish which group of genes can provide the maximum information, and therefore act as a reliable biomarker.

9. Ongoing Trials

There are currently numerous clinical trials focusing on NET (Table 6), the majority of which involve the combination of NET with targeted therapies.
At least seven of these uses any of the CDK4/6 inhibitors together with an AI or tamoxifen. Other biological molecules such as PI3K inhibitors, tyrosine kinase inhibitors [127], anti-VEGF, Ros inhibitors, or histone deacetylase inhibitors, combined with an AI, are also being studied. It is worth highlighting the great interest of the immune role in ER+ breast cancer, especially the antitumor immune response mediated by the programmed death 1/programmed death ligand 1 (PD1/PD-L1) and cytotoxic T lymphocyte antigen-4 inhibitors. Anti-PD-1/PD-L1 are already emerging as a promising treatment in non-luminal phenotypes and there are, to our knowledge, three clinical trials (NCT03573648, NCT02997995, NCT03874325) that are studying avelumab or durvalumab, combined with an AI, in the ER+ breast cancer neoadjuvant setting.
Genomic assays are being used for screening patients for various neoadjuvant alternatives. MammaPrint is the basis of the NCT03900637 trial, which randomizes ER+ breast cancer patients who are not suitable for BCS to either NCT or NET. Oncotype DX is being used in the DxCARTES trial to select candidates for NET with letrozole or letrozole + palbociclib.
Invasive lobular carcinoma accounts for only 15% of breast cancers, but it is known that these are usually especially responsive to NET, even though studies involving this histological subtype are scarce [128]. Only one “window of opportunity” trial focuses on the characteristics of NET in invasive lobular tumors (NCT02206984), comparing Ki67 changes and ER/PR expression after a 21-day cycle of tamoxifen or anastrozole or two doses of fulvestrant.
Finally, premenopausal patients continue to be a subject of concern, since as yet there is insufficient evidence of NET’s safety for this group. Several of the studies mentioned previously recruited premenopausal women, and they are also the exclusive focus of two Chinese trials (NCT0339753, NCT02535221) that compare NCT with NET in younger patients.

10. Conclusions

The evidence available to date shows that NET is an effective and safe alternative for the treatment of ER+/Her-2 negative postmenopausal patients, enabling higher BCS rates to be achieved, together with survival rates equivalent to those provided by NCT, even in cases of low axillary burden. NET has also proven to offer unquestionable opportunities for testing new therapeutic agents. In the future, the role of CDK4/6 inhibitors, as well as other target treatments, will probably extend NET indications to high-risk ER+ breast cancer patients. Nevertheless, a few questions remain unclear. In the first place, treatment duration, although usually recommended for 4–6 months, might sometimes be insufficient, and the optimal treatment length has yet to be determined, and probably needs to be individualized. Second, the innate endocrine resistance of some ER+ tumors raises concerns about the proper selection of patients. In this regard, genomic assays and other multigene tests performed on the diagnostic biopsy provide a very useful tool for determining endocrine sensitivity. Early on-treatment biopsies and Ki67 analysis may also offer valuable information about treatment response as well as prognosis; however, more reliable biomarkers should be investigated. Finally, offering NET to premenopausal women still remains controversial because there has been too little research on this group. Nevertheless, large luminal A-like tumors may be safely managed in this way in order to achieve BCS. The results of trials now in progress, as well as further research, will help broaden NET indications for these women.

Author Contributions

Conception and design: C.M.; drafting the manuscript and final editing: C.M., J.I.S.-M.; review of the literature: C.M., J.I.S.-M.; critical revision of the manuscript: C.M., J.I.S.-M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

The authors wish to thank Ralph Pemberton for his invaluable contribution to the translation and correction of this manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Krauss, K.; Stickeler, E. Endocrine Therapy in Early Breast Cancer. Breast Care 2020, 337–346. [Google Scholar] [CrossRef]
  2. Burstein, H.J.; Curigliano, G.; Loibl, S.; Dubsky, P.; Gnant, M.; Poortmans, P.; Colleoni, M.; Denkert, C.; Piccart-Gebhart, M.; Regan, M.; et al. Estimating the benefits of therapy for early-stage breast cancer: The St. Gallen International Consensus Guidelines for the primary therapy of early breast cancer 2019. Ann. Oncol. 2019, 30, 1541–1557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. NCCN. NCCN Clinical Practice Guidelines in Oncology. Breast Cancer. Version 1.2014. 2014. Available online: http://www.nccn.com (accessed on 8 April 2021).
  4. Arthur, L.M.; Turnbull, A.K.; Khan, L.R.; Dixon, J.M. Pre-operative Endocrine Therapy. Curr. Breast Cancer Rep. 2017, 9, 202–209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Rastelli, F.; Crispino, S. Factors predictive of response to hormone therapy in breast cancer. Tumori 2008, 94, 370–383. [Google Scholar] [CrossRef] [PubMed]
  6. Van Dam, P.A.; Van Dam, V.C.N.; Altintas, S.; Papadimitriou, K.; Rolfo, C.; Trinh, X.B. Neoadjuvant endocrine treatment in early breast cancer: An overlooked alternative? Eur. J. Surg. Oncol. 2016, 42, 333–342. [Google Scholar] [CrossRef] [PubMed]
  7. Eiermann, W.; Paepke, S.; Appfelstaedt, J.; Llombart-Cussac, A.; Eremin, J.; Vinholes, J.; Mauriac, L.; Ellis, M.; Lassus, M.; Chaudri-Ross, H.A.; et al. Preoperative treatment of postmenopausal breast cancer patients with letrozole: A randomized double-blind multicenter study. Ann. Oncol. 2001, 12, 1527–1532. [Google Scholar] [CrossRef] [PubMed]
  8. Smith, I.E.; Dowsett, M.; Ebbs, S.R.; Dixon, J.M.; Skene, A.; Blohmer, J.U.; Ashley, S.E.; Francis, S.; Boeddinghaus, I.; Walsh, G. Neoadjuvant treatment of postmenopausal breast cancer with anastrozole, tamoxifen, or both in combination: The Immediate Preoperative Anastrozole, Tamoxifen, or Combined With Tamoxifen (IMPACT) multicenter double-blind randomized trial. J. Clin. Oncol. 2005, 23, 5108–5116. [Google Scholar] [CrossRef] [PubMed]
  9. Dixon, J.M.; Renshaw, L.; Dixon, J.; Thomas, J. Invasive lobular carcinoma: Response to neoadjuvant letrozole therapy. Breast Cancer Res. Treat. 2011, 130, 871–877. [Google Scholar] [CrossRef]
  10. Barroso-Sousa, R.; Reis Silva, D.D.A.F.; Alessi, J.V.M.; Mano, M.S. Neoadjuvant endocrine therapy in breast cancer: Current role and future perspectives. Ecancermedicalscience 2016, 10, 1–15. [Google Scholar] [CrossRef] [Green Version]
  11. NICE. Overview|Early and Locally Advanced Breast Cancer: Diagnosis and Management|Guidance|NICE; NICE Guidelines. 2018. Available online: http://www.nice.org (accessed on 8 April 2021).
  12. Masuda, N.; Sagara, Y.; Kinoshita, T.; Iwata, H.; Nakamura, S.; Yanagita, Y.; Nishimura, R.; Iwase, H.; Kamigaki, S.; Takei, H.; et al. Neoadjuvant anastrozole versus tamoxifen in patients receiving goserelin for premenopausal breast cancer (STAGE): A double-blind, randomised phase 3 trial. Lancet Oncol. 2012, 13, 345–352. [Google Scholar] [CrossRef]
  13. Alba, E.; Calvo, L.; Albanell, J.; De la Haba, J.R.; Lanza, A.A.; Chacon, J.I.; Sanchez-Rovira, P.; Plazaola, A.; Lopez Garcia-Asenjo, J.A.; Bermejo, B.; et al. Chemotherapy (CT) and hormonotherapy (HT) as neoadjuvant treatment in luminal breast cancer patients: Results from the GEICAM/2006-03, a multicenter, randomized, phase-II study. Ann. Oncol. 2012, 23, 3069–3074. [Google Scholar] [CrossRef] [PubMed]
  14. Torrisi, R.; Bagnardi, V.; Pruneri, G.; Ghisini, R.; Bottiglieri, L.; Magni, E.; Veronesi, P.; D’Alessandro, C.; Luini, A.; Dellapasqua, S.; et al. Antitumour and biological effects of letrozole and GnRH analogue as primary therapy in premenopausal women with ER and PgR positive locally advanced operable breast cancer. Br. J. Cancer 2007, 97, 802–808. [Google Scholar] [CrossRef] [PubMed]
  15. Semiglazov, V.F.; Semiglazov, V.V.; Dashyan, G.A.; Ziltsova, E.K.; Ivanov, V.G.; Bozhok, A.A.; Melnikova, O.A.; Paltuev, R.M.; Kletzel, A.; Berstein, L.M. Phase 2 randomized trial of primary endocrine therapy versus chemotherapy in postmenopausal patients with estrogen receptor-positive breast cancer. Cancer 2007, 110, 244–254. [Google Scholar] [CrossRef]
  16. Palmieri, C.; Cleator, S.; Kilburn, L.S.; Kim, S.B.; Ahn, S.H.; Beresford, M.; Gong, G.; Mansi, J.; Mallon, E.; Reed, S.; et al. NEOCENT: A randomised feasibility and translational study comparing neoadjuvant endocrine therapy with chemotherapy in ER-rich postmenopausal primary breast cancer. Breast Cancer Res. Treat. 2014, 148, 581–590. [Google Scholar] [CrossRef]
  17. Spring, L.M.; Gupta, A.; Reynolds, K.L.; Gadd, M.A.; Ellisen, L.W.; Isakoff, S.J.; Moy, B.; Bardia, A. Neoadjuvant endocrine therapy for estrogen receptor-positive breast cancer a systematic review and meta-Analysis. JAMA Oncol. 2016, 2, 1477–1486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Wang, Y.; He, L.; Song, Y.; Wu, Q.; Wang, H.; Zhang, B.; Ma, X. The tumour response of postmenopausal hormone receptor-positive breast cancers undergoing different types of neoadjuvant therapy: A meta-analysis. BMC Women’s Health 2020, 20, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Preece, R.A.; Wood, C.R.; Mackie, A.C. A prospective comparative clinical trial Tamoxifen as initial sole treatment of localised breast cancer in elderly women: A pilot study. Br. Med. J. 1982, 284, 869–870. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Gazet, J.C.; Ford, H.T.; Bland, J.M.; Markopoulos Coombes, R.C.; Dixon, R.C. Prospective Randomised Trial Of Tamoxifen versus Surgery In Elderly Patients With Breast Cancer. Lancet 1988, 1, 679–681. [Google Scholar] [CrossRef]
  21. Gazet, J.C.; Ford, H.T.; Coombes, R.C.; Bland, J.M.; Sutcliffe, R.; Quilliam, J.; Lowndes, S. Prospective randomized trial of tamoxifen vs surgery in elderly patients with breast cancer. Eur. J. Surg. Oncol. 1994, 20, 207–214. [Google Scholar] [PubMed]
  22. Li, J.J.; Shao, Z.M. Endocrine therapy as adjuvant or neoadjuvant therapy for breast cancer: Selecting the best agents, the timing and duration of treatment. Chin. Clin. Oncol. 2016, 5, 1–12. [Google Scholar] [CrossRef]
  23. Takei, H.; Kurosumi, M.; Yoshida, T.; Hayashi, Y.; Higuchi, T.; Uchida, S.; Ninomiya, J.; Oba, H.; Inoue, K.; Nagai, S.; et al. Neoadjuvant endocrine therapy of breast cancer: Which patients would benefit and what are the advantages? Breast Cancer 2011, 18, 85–91. [Google Scholar] [CrossRef] [PubMed]
  24. Montagna, G.; Sevilimedu, V.; Fornier, M.; Jhaveri, K.; Morrow, M.; Pilewskie, M.L. How Effective is Neoadjuvant Endocrine Therapy (NET) in Downstaging the Axilla and Achieving Breast-Conserving Surgery? Ann. Surg. Oncol. 2020, 27, 4702–4710. [Google Scholar] [CrossRef] [PubMed]
  25. Cardoso, F.; Senkus, E.; Costa, A.; Papadopoulos, E.; Aapro, M.; André, F.; Harbeck, N.; Lopez, B.A.; Barrios, C.H.; Bergh, J.; et al. 4th ESO-ESMO international consensus guidelines for advanced breast cancer (ABC 4). Ann. Oncol. 2018, 29, 1634–1657. [Google Scholar] [CrossRef] [PubMed]
  26. Madigan, L.I.; Dinh, P.; Graham, J.D. Neoadjuvant endocrine therapy in locally advanced estrogen or progesterone receptor-positive breast cancer: Determining the optimal endocrine agent and treatment duration in postmenopausal women—A literature review and proposed guidelines. Breast Cancer Res. 2020, 22, 77. [Google Scholar] [CrossRef] [PubMed]
  27. Charehbili, A.; Fontein, D.B.Y.; Kroep, J.R.; Liefers, G.J.; Mieog, J.S.D.; Nortier, J.W.R.; van de Velde, C.J. Neoadjuvant hormonal therapy for endocrine sensitive breast cancer: A systematic review. Cancer Treat. Rev. 2014, 40, 86–92. [Google Scholar] [CrossRef] [PubMed]
  28. Samiei, S.; van Kaathoven, B.N.; Boersma, L.; Granzier, R.W.Y.; Siesling, S.; Engelen, S.M.E.; de Munck, L.; van Kuijk, S.M.J.; van der Hulst, R.R.J.W.; Lobbes, M.B.I.; et al. Risk of Positive Sentinel Lymph Node After Neoadjuvant Systemic Therapy in Clinically Node-Negative Breast Cancer: Implications for Postmastectomy Radiation Therapy and Immediate Breast Reconstruction. Ann. Surg. Oncol. 2019, 26, 3902–3909. [Google Scholar] [CrossRef] [Green Version]
  29. Laws, A.; Hughes, M.E.; Hu, J.; Barry, W.T.; Dominici, L.; Nakhlis, F.; Barbie, T.; Duggan, M.; Weiss, A.; Rhei, E.; et al. Impact of Residual Nodal Disease Burden on Technical Outcomes of Sentinel Lymph Node Biopsy for Node-Positive (cN1) Breast Cancer Patients Treated with Neoadjuvant Chemotherapy. Ann. Surg. Oncol. 2019, 26, 3846–3855. [Google Scholar] [CrossRef]
  30. Hammond, J.B.; Parnall, T.H.; Scott, D.W.; Kosiorek, H.E.; Pockaj, B.A.; Ernst, B.J.; Northfelt, D.W.; McCullough, A.E.; Ocal, I.T.; Cronin, P.A. Gauging the efficacy of neoadjuvant endocrine therapy in breast cancer patients with known axillary disease. J. Surg. Oncol. 2020, 122, 619–622. [Google Scholar] [CrossRef] [PubMed]
  31. Rusz, O.; Vörös, A.; Varga, Z.; Kelemen, G.; Uhercsák, G.; Nikolényi, A.; Ormándi, K.; Simonka, Z.; Kahán, Z. One-Year Neoadjuvant Endocrine Therapy in Breast Cancer. Pathol. Oncol. Res. 2015, 21, 977–984. [Google Scholar] [CrossRef] [PubMed]
  32. Stafford, A.; Williams, A.; Edmiston, K.; Cocilovo, C.; Cohen, R.; Bruce, S.; Yoon-Flannery, K.; De La Cruz, L. Axillary Response in Patients Undergoing Neoadjuvant Endocrine Treatment for Node-Positive Breast Cancer: Systematic Literature Review and NCDB Analysis. Ann. Surg. Oncol. 2020, 27, 4669–4677. [Google Scholar] [CrossRef]
  33. Weiss, A.; Wong, S.; Golshan, M.; Freedman, R.A.; Metzger, O.; Bellon, J.; Mittendorf, E.A.; King, T.A. Patterns of Axillary Management in Stages 2 and 3 Hormone Receptor-Positive Breast Cancer by Initial Treatment Approach. Ann. Surg. Oncol. 2019, 26, 4326–4336. [Google Scholar] [CrossRef]
  34. Boughey, J.C.; Suman, V.J.; Mittendorf, E.A.; Ahrendt, G.M.; Wilke, L.G.; Taback, B.; Leitch, A.M.; Kuerer, H.M.; Bowling, M.; Flippo-Morton, T.S.; et al. Sentinel lymph node surgery after neoadjuvant chemotherapy in patients with node-positive breast cancer: The ACOSOG Z1071 (alliance) clinical trial. JAMA J. Am. Med. Assoc. 2013, 310, 1455–1461. [Google Scholar] [CrossRef] [Green Version]
  35. Boileau, J.F.; Poirier, B.; Basik, M.; Holloway, C.M.B.; Gaboury, L.; Sideris, L.; Meterissian, S.; Arnaout, A.; Brackstone, M.; McCready, D.R.; et al. Sentinel node biopsy after neoadjuvant chemotherapy in biopsy-proven node-positive breast cancer: The SN FNAC study. J. Clin. Oncol. 2015, 33, 258–264. [Google Scholar] [CrossRef] [PubMed]
  36. Kuehn, T.; Bauerfeind, I.; Fehm, T.; Fleige, B.; Hausschild, M.; Helms, G.; Lebeau, A.; Liedtke, C.; von Minckwitz, G.; Nekljudova, V.; et al. Sentinel-lymph-node biopsy in patients with breast cancer before and after neoadjuvant chemotherapy (SENTINA): A prospective, multicentre cohort study. Lancet Oncol. 2013, 14, 609–618. [Google Scholar] [CrossRef]
  37. Kantor, O.; Wong, S.; Weiss, A.; Metzger, O.; Mittendorf, E.A.; King, T.A. Prognostic significance of residual nodal disease after neoadjuvant endocrine therapy for hormone receptor-positive breast cancer. Breast Cancer 2020, 6, 5–10. [Google Scholar] [CrossRef]
  38. Kantor, O.; Wakeman, M.; Weiss, A.; Wong, S.; Laws, A.; Grossmith, S.; Mittendorf, E.A.; King, T.A. Axillary Management After Neoadjuvant Endocrine Therapy for Hormone Receptor-Positive Breast Cancer. Ann. Surg. Oncol. 2020. [Google Scholar] [CrossRef] [PubMed]
  39. Hind, D.; Wyld, L.; Reed, M.W. Surgery, with or without tamoxifen, vs tamoxifen alone for older women with operable breast cancer: Cochrane review. Br. J. Cancer 2007, 96, 1025–1029. [Google Scholar] [CrossRef] [Green Version]
  40. Wink, C.J.; Woensdregt, K.; Nieuwenhuijzen, G.A.P.; Van Der Sangen, M.J.C.; Hutschemaekers, S.; Roukema, J.A.; Tjan-Heijnen, V.C.; Voogd, A.C. Hormone treatment without surgery for patients aged 75 years or older with operable breast cancer. Ann. Surg. Oncol. 2012, 19, 1185–1191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Osborn, G.; Jones, M.; Champ, C.; Gower-Thomas, K.; Vaughan-Williams, E. Is primary endocrine therapy effective in treating the elderly, unfit patient with breast cancer? Ann. R. Coll. Surg. Engl. 2011, 93, 286–289. [Google Scholar] [CrossRef] [PubMed]
  42. Morgan, J.L.; Reed, M.W.; Wyld, L. Primary endocrine therapy as a treatment for older women with operable breast cancer—A comparison of randomized controlled trial and cohort study findings. Eur. J. Surg. Oncol. 2014, 40, 676–684. [Google Scholar] [CrossRef] [PubMed]
  43. Carlson, G.W. Total mastectomy under local anesthesia: The tumescent technique. Breast J. 2005, 11, 100–102. [Google Scholar] [CrossRef] [PubMed]
  44. Sabel, M.S.; Kaufman, C.S.; Whitworth, P.; Chang, H.; Stocks, L.H.; Simmons, R.; Schultz, M. Cryoablation of early-stage breast cancer: Work-in-progress report of a multi-institutional trial. Ann. Surg. Oncol. 2004, 11, 542–549. [Google Scholar] [CrossRef] [PubMed]
  45. Simmons, R.M.; Ballman, K.V.; Cox, C.; Carp, N.; Sabol, J.; Hwang, R.F.; Attai, D.; Sabel, M.; Nathanson, D.; Kenler, A.; et al. A Phase II Trial Exploring the Success of Cryoablation Therapy in the Treatment of Invasive Breast Carcinoma: Results from ACOSOG (Alliance) Z1072. Ann. Surg. Oncol. 2016, 23, 2438–2445. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Cataliotti, L.; Buzdar, A.U.; Noguchi, S.; Bines, J.; Takatsuka, Y.; Petrakova, K.; Dube, P.; de Oliveira, C.T. Comparison of anastrozole versus tamoxifen as preoperative therapy in postmenopausal women with hormone receptor-positive breast cancer: The Pre-Operative “Arimidex” Compared to Tamoxifen (PROACT) trial. Cancer 2006, 106, 2095–2103. [Google Scholar] [CrossRef] [PubMed]
  47. Semiglazov, V.; Kletsel, A.; Semiglazov, V.; Zhiltzova, E.; Ivanov, V.; Dashyan, G.; Bozhok, A.; Melnikova, O.; Paltuev, R.; Berstein, L. Exemestane (E) vs tamoxifen (T) as neoadjuvant endocrine therapy for postmenopausal women with ER+ breast cancer (T2N1–2, T3N0–1, T4N0M0). J. Clin. Oncol. 2005, 23, 530. [Google Scholar] [CrossRef]
  48. Ellis, M.J.; Suman, V.J.; Hoog, J.; Lin, L.; Snider, J.; Prat, A.; Parker, J.S.; Luo, J.; DeSchryver, K.; Allred, D.C.; et al. Randomized phase II neoadjuvant comparison between letrozole, anastrozole, and exemestane for postmenopausal women with estrogen receptor-rich stage 2 to 3 breast cancer: Clinical and biomarker outcomes and predictive value of the baseline PAM50-based intrinsic subtype-ACOSOGZ1031. J. Clin. Oncol. 2011, 29, 2342–2349. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Lerebours, F.; Rivera, S.; Mouret-Reynier, M.A.; Alran, S.; Venat-Bouvet, L.; Kerbrat, P.; Salmon, R.; Becette, V.; Bourgier, C.; Cherel, P.; et al. Randomized phase 2 neoadjuvant trial evaluating anastrozole and fulvestrant efficacy for postmenopausal, estrogen receptor–positive, human epidermal growth factor receptor 2–negative breast cancer patients: Results of the UNICANCER CARMINA 02 french trial. Cancer 2016, 122, 3032–3040. [Google Scholar] [CrossRef]
  50. Quenel-Tueux, N.; Debled, M.; Rudewicz, J.; MacGrogan, G.; Pulido, M.; Mauriac, L.; Dalenc, F.; Bachelot, T.; Lortal, B.; Breton-Callu, C.; et al. Clinical and genomic analysis of a randomised phase II study evaluating anastrozole and fulvestrant in postmenopausal patients treated for large operable or locally advanced hormone-receptor-positive breast cancer. Br. J. Cancer 2015, 113, 585–594. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Krainick-Strobel, U.E.; Lichtenegger, W.; Wallwiener, D.; Tulusan, A.H.; Jänicke, F.; Bastert, G.; Kiesel, L.; Wackwitz, B.; Paepke, S. Neoadjuvant letrozole in postmenopausal estrogen and/or progesterone receptor positive breast cancer: A phase IIb/III trial to investigate optimal duration of preoperative endocrine therapy. BMC Cancer 2008, 8, 62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Dixon, J.M.; Renshaw, L.; MacAskill, E.J.; Young, O.; Murray, J.; Cameron, D.; Kerr, G.R.; Evans, D.B.; Miller, W.R. Increase in response rate by prolonged treatment with neoadjuvant letrozole. Breast Cancer Res. Treat. 2009, 113, 145–151. [Google Scholar] [CrossRef] [PubMed]
  53. Llombart-Cussac, A.; Guerrero, Á.; Galán, A.; Carañana, V.; Buch, E.; Rodríguez-Lescure, Á.; Ruiz, A.; Fuster, D.C.; Guillem Porta, V. Phase II trial with letrozole to maximum response as primary systemic therapy in postmenopausal patients with ER/PgR[+] operable breast cancer. Clin. Transl. Oncol. 2012, 14, 125–131. [Google Scholar] [CrossRef] [PubMed]
  54. Carpenter, R.; Doughty, J.C.; Cordiner, C.; Moss, N.; Gandhi, A.; Wilson, C.; Andrews, C.; Ellis, G.; Gui, G.; Skene, A.I. Optimum duration of neoadjuvant letrozole to permit breast conserving surgery. Breast Cancer Res. Treat. 2014, 144, 569–576. [Google Scholar] [CrossRef]
  55. Fontein, D.B.Y.; Charehbili, A.; Nortier, J.W.R.; Kranenbarg, E.M.-K.; Kroep, J.R.; Putter, H.; van Riet, Y.; Nieuwenhuijzen, G.A.; de Valk, B.; Terwogt, J.M.; et al. Efficacy of six month neoadjuvant endocrine therapy in postmenopausal, hormone receptor-positive breast cancer patients—A phase II trial. Eur. J. Cancer 2014, 50, 2190–2200. [Google Scholar] [CrossRef] [PubMed]
  56. Allevi, G.; Strina, C.; Andreis, D.; Zanoni, V.; Bazzola, L.; Bonardi, S.; Foroni, C.; Milani, M.; Cappelletti, M.R.; Gussago, F.; et al. Increased pathological complete response rate after a long-term neoadjuvant letrozole treatment in postmenopausal oestrogen and/or progesterone receptor-positive breast cancer. Br. J. Cancer 2013, 108, 1587–1592. [Google Scholar] [CrossRef] [Green Version]
  57. Hojo, T.; Kinoshita, T.; Imoto, S.; Shimizu, C.; Isaka, H.; Ito, H.; Imi, K.; Wada, N.; Ando, M.; Fujiwara, Y. Use of the neo-adjuvant exemestane in post-menopausal estrogen receptor-positive breast cancer: A randomized phase II trial (PTEX46) to investigate the optimal duration of preoperative endocrine therapy. Breast 2013, 22, 263–267. [Google Scholar] [CrossRef]
  58. Goldhirsch, A.; Winer, E.P.; Coates, A.S.; Gelber, R.D.; Piccart-Gebhart, M.; Thrlimann, B.; Senn, H.J. Personalizing the treatment of women with early breast cancer: Highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2013. Ann. Oncol. 2013, 24, 2206–2223. [Google Scholar] [CrossRef]
  59. Johnston, S.J.; Kenny, F.S.; Syed, B.M.; Robertson, J.F.R.; Pinder, S.E.; Winterbottom, L.; Ellis, I.O.; Blamey, R.W.; Cheung, K.L. A randomised trial of primary tamoxifen versus mastectomy plus adjuvant tamoxifen in fit elderly women with invasive breast carcinoma of high oestrogen receptor content: Long-term results at 20 years of follow-up. Ann. Oncol. 2012, 23, 2296–2300. [Google Scholar] [CrossRef]
  60. Pepping, R.M.C.; Portielje, J.E.A.; van de Water, W.; de Glas, N.A. Primary Endocrine Therapy in Older Women with Breast Cancer. Curr. Geriatr. Rep. 2017, 6, 239–246. [Google Scholar] [CrossRef] [Green Version]
  61. Willsher, P.C.; Robertson, J.F.R.; Jackson, L.; Al-Hilaly, M.; Blarney, R.W. Investigation of primary tamoxifen therapy for elderly patients with operable breast cancer. Breast 1997, 6, 150–154. [Google Scholar] [CrossRef]
  62. Croshaw, R.; Shapiro-Wright, H.; Svensson, E.; Erb, K.; Julian, T. Accuracy of clinical examination, digital mammogram, ultrasound, and MRI in determining postneoadjuvant pathologic tumor response in operable breast cancer patients. Ann. Surg. Oncol. 2011, 18, 3160–3163. [Google Scholar] [CrossRef] [PubMed]
  63. Fowler, A.M.; Mankoff, D.A.; Joe, B.N. Imaging neoadjuvant therapy response in breast cancer. Radiology 2017, 285, 358–375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Hilal, T.; Covington, M.; Kosiorek, H.E.; Zwart, C.; Ocal, I.T.; Pockaj, B.A.; Northfelt, D.W.; Patel, B.K. Breast MRI phenotype and background parenchymal enhancement may predict tumor response to neoadjuvant endocrine therapy. Breast J. 2018, 24, 1010–1014. [Google Scholar] [CrossRef] [PubMed]
  65. Zhang, K.; Li, J.; Zhu, Q.; Chang, C. Prediction of pathologic complete response by ultrasonography and magnetic resonance imaging after neoadjuvant chemotherapy in patients with breast cancer. Cancer Manag. Res. 2020, 12, 2603–2612. [Google Scholar] [CrossRef] [Green Version]
  66. Takeda, K.; Kanao, S.; Okada, T.; Ueno, T.; Toi, M.; Ishiguro, H.; Mikami, Y.; Tanaka, S.; Togashi, K. MRI evaluation of residual tumor size after neoadjuvant endocrine therapy vs. neoadjuvant chemotherapy. Eur. J. Radiol. 2012, 81, 2148–2153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Reis, J.; Lindstrøm, J.C.; Boavida, J.; Gjesdal, K.I.; Park, D.; Bahrami, N.; Seyedzadeh, M.; Melles, W.A.; Sauer, T.; Geisler, J.; et al. Accuracy of breast MRI in patients receiving neoadjuvant endocrine therapy: Comprehensive imaging analysis and correlation with clinical and pathological assessments. Breast Cancer Res. Treat. 2020, 184, 407–420. [Google Scholar] [CrossRef]
  68. Vriens, B.E.P.J.; De Vries, B.; Lobbes, M.B.I.; Van Gastel, S.M.; Van Den Berkmortel, F.W.P.J.; Smilde, T.J.; van Warmerdam, L.J.; de Boer, M.; van Spronsen, D.J.; Smidt, M.L.; et al. Ultrasound is at least as good as magnetic resonance imaging in predicting tumour size post-neoadjuvant chemotherapy in breast cancer. Eur. J. Cancer 2016, 52, 67–76. [Google Scholar] [CrossRef]
  69. Ragusi, M.A.A.; Loo, C.E.; van der Velden, B.H.M.; Wesseling, J.; Linn, S.C.; Beets-Tan, R.G.; Elias, S.G.; Gilhuijs, K.G.A. Contralateral parenchymal enhancement on breast MRI before and during neoadjuvant endocrine therapy in relation to the preoperative endocrine prognostic index. Eur. Radiol. 2020, 30, 6740–6748. [Google Scholar] [CrossRef]
  70. Ueda, S.; Tsuda, H.; Saeki, T.; Omata, J.; Osaki, A.; Shigekawa, T.; Ishida, J.; Tamura, K.; Abe, Y.; Moriya, T.; et al. Early metabolic response to neoadjuvant letrozole, measured by FDG PET/CT, is correlated with a decrease in the Ki67 labeling index in patients with hormone receptor-positive primary breast cancer: A pilot study. Breast Cancer 2011, 18, 299–308. [Google Scholar] [CrossRef]
  71. Boughdad, S.; Champion, L.; Becette, V.; Cherel, P.; Fourme, E.; Lemonnier, J.; Lerebours, F.; Alberini, J. L.Early metabolic response of breast cancer to neoadjuvant endocrine therapy: Comparison to morphological and pathological response. Cancer Imaging 2020, 20, 11. [Google Scholar] [CrossRef]
  72. Gil-Rendo, A.; Martínez-Regueira, F.; Zornoza, G.; García-Velloso, M.J.; Beorlegui, C.; Rodriguez-Spiteri, N. Association between [18F]fluorodeoxyglucose uptake and prognostic parameters in breast cancer. Br. J. Surg. 2009, 96, 166–170. [Google Scholar] [CrossRef]
  73. Ming, Y.; Wu, N.; Qian, T.; Li, X.; Wan, D.Q.; Li, C.; Li, Y.; Wu, Z.; Wang, X.; Liu, J.; et al. Progress and Future Trends in PET/CT and PET/MRI Molecular Imaging Approaches for Breast Cancer. Front. Oncol. 2020, 10, 1301. [Google Scholar] [CrossRef] [PubMed]
  74. Chae, S.Y.; Kim, S.B.; Ahn, S.H.; Kim, H.O.; Yoon, D.H.; Ahn, J.H.; Jung, K.H.; Han, S.; Oh, S.J.; Lee, S.J.; et al. A randomized feasibility study of 18F-fluoroestradiol PET to predict pathologic response to neoadjuvant therapy in estrogen receptor-rich postmenopausal breast cancer. J. Nucl. Med. 2017, 58, 563–568. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Groheux, D. 18F-fluoroestradiol PET to predict the response to neoadjuvant treatment of luminal breast cancer. J. Nucl. Med. 2017, 58, 683. [Google Scholar] [CrossRef] [Green Version]
  76. Mortimer, J.E.; Dehdashti, F.; Siegel, B.A.; Katzenellenbogen, J.A.; Fracasso, P.; Welch, M.J. Positron emission tomography with 2-[18F]fluoro-2-deoxy-D-glucose and 16α-[18F]fluoro-17β-estradiol in breast cancer: Correlation with estrogen receptor status and response to systemic therapy. Clin. Cancer Res. 1996, 2, 933–939. [Google Scholar]
  77. Cortazar, P.; Zhang, L.; Untch, M.; Mehta, K.; Costantino, J.P.; Wolmark, N.; Bonnefoi, H.; Cameron, D.; Gianni, L.; Valagussa, P.; et al. Pathological complete response and long-term clinical benefit in breast cancer: The CTNeoBC pooled analysis. Lancet 2014, 384, 164–172. [Google Scholar] [CrossRef] [Green Version]
  78. Urruticoechea, A.; Smith, I.E.; Dowsett, M. Proliferation marker Ki-67 in early breast cancer. J. Clin. Oncol. 2005, 23, 7212–7220. [Google Scholar] [CrossRef] [PubMed]
  79. Smith, I.; Robertson, J.; Kilburn, L.; Wilcox, M.; Evans, A.; Holcombe, C.; Horgan, K.; Kirwan, C.; Mallon, E.; Sibbering, M.; et al. Long-term outcome and prognostic value of Ki67 after perioperative endocrine therapy in postmenopausal women with hormone-sensitive early breast cancer (POETIC): An open-label, multicentre, parallel-group, randomised, phase 3 trial. Lancet Oncol. 2020, 21, 1443–1454. [Google Scholar] [CrossRef]
  80. Dowsett, M.; Smith, I.E.; Ebbs, S.R.; Dixon, J.M.; Skene, A.; Griffith, C.; Boeddinghaus, I.; Salter, J.; Detre, S.; Hills, M.; et al. Short-term changes in Ki-67 during neoadjuvant treatment of primary breast cancer with anastrozole or tamoxifen alone or combined correlate with recurrence-free survival. Clin. Cancer Res. 2005, 11, 951–959. [Google Scholar]
  81. Ellis, M.J.; Suman, V.J.; Hoog, J.; Goncalves, R.; Sanati, S.; Creighton, C.J.; DeSchryver, K.; Crouch, E.; Brink, A.; Watson, M.; et al. Ki67 proliferation index as a tool for chemotherapy decisions during and after neoadjuvant aromatase inhibitor treatment of breast cancer: Results from the American college of surgeons oncology group Z1031 trial (Alliance). J. Clin. Oncol. 2017, 35, 1061–1069. [Google Scholar] [CrossRef]
  82. Ellis, M.J. Lessons in precision oncology from neoadjuvant endocrine therapy trials in ER+ breast cancer. Breast 2017, 34, S104–S107. [Google Scholar] [CrossRef] [Green Version]
  83. Ma, C.X.; Suman, V.J.; Leitch, A.M.; Sanati, S.; Vij, K.R.; Unzeitig, G.W.; Unzeitig, G.W.; Hoog, J.; Watson, M.; Hahn, O.M.; et al. ALTERNATE: Neoadjuvant endocrine treatment (NET) approaches for clinical stage II or III estrogen receptor-positive HER2-negative breast cancer (ER+ HER2- BC) in postmenopausal (PM) women: Alliance A011106. J. Clin. Oncol. 2020, 38 (Suppl. S15), 504. [Google Scholar] [CrossRef]
  84. Hofmann, D.; Nitz, U.; Gluz, O.; Kates, R.E.; Schinkoethe, T.; Staib, P.; Harbeck, N. WSG ADAPT—Adjuvant dynamic marker-adjusted personalized therapy trial optimizing risk assessment and therapy response prediction in early breast cancer: Study protocol for a prospective, multi-center, controlled, non-blinded, randomized, investigator initiated phase II/III trials. Trials 2013, 14, 261. [Google Scholar] [CrossRef] [Green Version]
  85. Martí, C.; Sánchez-Méndez, J.I. Neoadjuvant endocrine therapy for luminal breast cancer treatment: A first-choice alternative in times of crisis such as the COVID-19 pandemic. Ecancermedicalscience 2020, 14, 1–6. [Google Scholar] [CrossRef] [PubMed]
  86. Guarneri, V.; Dieci, M.V.; Bisagni, G.; Frassoldati, A.; Bianchi, G.V.; De Salvo, G.L.; Orvieto, E.; Urso, L.; Pascual, T.; Paré, L.; et al. De-escalated therapy for HR+/HER2+ breast cancer patients with Ki67 response after 2-week letrozole: Results of the PerELISA neoadjuvant study. Ann. Oncol. 2019, 30, 921–926. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Guerrero-zotano, A.L.; Arteaga, C.L.; Biology, C.; Program, B.C.; Cancer, V. Neoadjuvant trials in ER+ breast cancer: A tool for acceleration of drug development and discovery. Cancer Discov. 2017, 7, 561–574. [Google Scholar] [CrossRef] [Green Version]
  88. Ellis, M.J.; Tao, Y.; Luo, J.; A’Hern, R.; Evans, D.B.; Bhatnagar, A.S.; Chaudri Ross, H.A.; von Kameke, A.; Miller, W.R.; Smith, I.; et al. Outcome prediction for estrogen receptor-positive breast cancer based on postneoadjuvant endocrine therapy tumor characteristics. J. Natl. Cancer Inst. 2008, 100, 1380–1388. [Google Scholar] [CrossRef]
  89. Kurozumi, S.; Matsumoto, H.; Inoue, K.; Tozuka, K.; Hayashi, Y.; Kurosumi, M.; Oyama, T.; Fujii, T.; Horiguchi, J.; Kuwano, H. Impact of combining the progesterone receptor and preoperative endocrine prognostic index (PEPI) as a prognostic factor after neoadjuvant endocrine therapy using aromatase inhibitors in postmenopausal ER positive and HER2 negative breast cancer. PLoS ONE 2018, 13, e0201846. [Google Scholar] [CrossRef]
  90. Vendrell, J.A.; Solassol, J.; Gyo, B.; Vilquin, P.; Jarlier, M.; Donini, C.F.; Gamba, L.; Maudelonde, T.; Rouanet, P.; Cohen, P.A. Evaluating ZNF217 mRNA expression levels as a predictor of response to endocrine therapy in ER+ breast cancer. Front. Pharmacol. 2019, 9, 1581. [Google Scholar] [CrossRef] [PubMed]
  91. Goto-Yamaguchi, L.; Yamamoto-Ibusuki, M.; Yamamoto, Y.; Fujiki, Y.; Tomiguchi, M.; Sueta, A.; Takeshita, T.; Iwase, H. Therapeutic predictors of neoadjuvant endocrine therapy response in estrogen receptor-positive breast cancer with reference to optimal gene expression profiling. Breast Cancer Res. Treat. 2018, 172, 353–362. [Google Scholar] [CrossRef]
  92. Guarneri, V.; Generali, D.G.; Frassoldati, A.; Artioli, F.; Boni, C.; Cavanna, L.; Tagliafico, E.; Maiorana, A.; Bottini, A.; Cagossi, K.; et al. Double-blind, placebo-controlled, multicenter, randomized, phase IIB neoadjuvant study of letrozole-lapatinib in postmenopausal hormone receptor-positive, human epidermal growth factor receptor 2-negative, operable breast cancer. J. Clin. Oncol. 2014, 32, 1050–1057. [Google Scholar] [CrossRef] [Green Version]
  93. Mayer, I.A.; Prat, A.; Egle, D.; Blau, S.; Fidalgo, J.A.P.; Fasching, P.A.; Fasching, P.A.; Colleoni, M.; Wolff, A.C.; Winer, E.P.; et al. Growth Factor Receptor 2-Negative Breast Cancer (NEO-ORB). Clin. Cancer Res. 2019, 25, 2975–2987. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Ma, C.X.; Gao, F.; Luo, J.; Northfelt, D.W.; Goetz, M.; Forero, A.; Hoog, J.; Naughton, M.; Ademuyiwa, F.; Suresh, R.; et al. NeoPalAna: Neoadjuvant palbociclib, a cyclin-dependent kinase 4/6 inhibitor, and anastrozole for clinical stage 2 or 3 estrogen receptor–positive breast cancer. Clin. Cancer Res. 2017, 23, 4055–4065. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Cottu, P.; D’Hondt, V.; Dureau, S.; Lerebours, F.; Desmoulins, I.; Heudel, P.E.; Duhoux, F.P.; Levy, C.; Mouret-Reynier, M.A.; Dalenc, F.; et al. Letrozole and palbociclib versus chemotherapy as neoadjuvant therapy of high-risk luminal breast cancer. Ann. Oncol. 2018, 29, 2334–2340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Prat, A.; Saura, C.; Pascual, T.; Hernando, C.; Muñoz, M.; Paré, L.; González Farré, B.; Fernández, P.L.; Galván, P.; Chic, N.; et al. Ribociclib plus letrozole versus chemotherapy for postmenopausal women with hormone receptor-positive, HER2-negative, luminal B breast cancer (CORALLEEN): An open-label, multicentre, randomised, phase 2 trial. Lancet Oncol. 2020, 21, 33–43. [Google Scholar] [CrossRef]
  97. Baselga, J.; Semiglazov, V.; Van Dam, P.; Manikhas, A.; Bellet, M.; Mayordomo, J.; Campone, M.; Kubista, E.; Greil, R.; Bianchi, G.; et al. Phase II randomized study of neoadjuvant everolimus plus letrozole compared with placebo plus letrozole in patients with estrogen receptor-positive breast cancer. J. Clin. Oncol. 2009, 27, 2630–2637. [Google Scholar] [CrossRef] [PubMed]
  98. Saura, C.; Hlauschek, D.; Oliveira, M.; Zardavas, D.; Jallitsch-Halper, A.; de la Peña, L.; Nuciforo, P.; Ballestrero, A.; Dubsky, P.; Lombard, J.M.; et al. Neoadjuvant letrozole plus taselisib versus letrozole plus placebo in postmenopausal women with oestrogen receptor-positive, HER2-negative, early-stage breast cancer (LORELEI): A multicentre, randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Oncol. 2019, 20, 1226–1238. [Google Scholar] [CrossRef]
  99. Johnston, S.; Puhalla, S.; Wheatley, D.; Ring, A.; Barry, P.; Holcombe, C.; Boileau, J.F.; Provencher, L.; Robidoux, A.; Rimawi, M.; et al. Randomized phase II study evaluating palbociclib in addition to letrozole as neoadjuvant therapy in estrogen receptor–positive early breast cancer: Pallet trial. J. Clin. Oncol. 2019, 37, 178–189. [Google Scholar] [CrossRef] [PubMed]
  100. Hurvitz, S.A.; Martin, M.; Press, M.F.; Chan, D.; Fernandez-abad, M.; Petru, E.; Rostorfer, R.; Guarneri, V.; Huang, C.S.; Barriga, S.; et al. Potent Cell-Cycle Inhibition and Upregulation of Immune Response with Abemaciclib and Anastrozole in neoMONARCH, Phase II Neoadjuvant Study in HR+/HER2− Breast Cancer. Clin. Cancer Res. 2020, 26, 566–580. [Google Scholar] [CrossRef] [Green Version]
  101. Curigliano, G.; Pardo, P.G.; Meric-Bernstam, F.; Conte, P.; Lolkema, M.P.; Beck, J.T.; Bardia, A.; García, M.M.; Penault-Llorca, F.; Dhuria, S.; et al. Ribociclib plus letrozole in early breast cancer: A presurgical, window-of-opportunity study. Breast 2016, 28, 191–198. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Straver, M.E.; Glas, A.M.; Hannemann, J.; Wesseling, J.; Van De Vijver, M.J.; Rutgers, E.J.T.; Vrancken Peeters, M.J.; van Tinteren, H.; Van’t Veer, L.J.; Rodenhuis, S. The 70-gene signature as a response predictor for neoadjuvant chemotherapy in breast cancer. Breast Cancer Res. Treat. 2010, 119, 551–558. [Google Scholar] [CrossRef] [Green Version]
  103. Parker, J.S.; Mullins, M.; Cheung, M.C.U.; Leung, S.; Voduc, D.; Vickery, T.; Fauron, C.; He, X.; Hu, Z.; Quackenbush, J.F.; et al. Supervised risk predictor of breast cancer based on intrinsic subtypes. J. Clin. Oncol. 2009, 27, 1160–1167. [Google Scholar] [CrossRef] [PubMed]
  104. Parker, J.; Prat, A.; Cheang, M.; Lenburg, S.; Paik, J.; Perou, C. Breast Cancer Molecular Subtypes Predict Response to Anthracycline/Taxane-Based Chemotherapy. Cancer Res. 2009, 69, 598s. [Google Scholar]
  105. Iwao-Koizumi, K.; Matoba, R.; Ueno, N.; Kim, S.J.; Ando, A.; Miyoshi, Y.; Miyoshi, Y.; Maeda, E.; Noguchi, S.; Kato, K. Prediction of docetaxel response in human breast cancer by gene expression profiling. J. Clin. Oncol. 2005, 23, 422–431. [Google Scholar] [CrossRef]
  106. Dubsky, P.C.; Singer, C.F.; Egle, D.; Wette, V.; Petru, E.; Balic, M.; Pichler, A.; Greil, R.; Petzer, A.L.; Bago-Horvath, Z.; et al. The EndoPredict score predicts response to neoadjuvant chemotherapy and neoendocrine therapy in hormone receptor-positive, human epidermal growth factor receptor 2-negative breast cancer patients from the ABCSG-34 trial. Eur. J. Cancer 2020, 134, 99–106. [Google Scholar] [CrossRef]
  107. Soliman, H.; Wagner, S.; Flake, D.D.; Robson, M.; Schwartzberg, L.; Sharma, P.; Magliocco, A.; Kronenwett, R.; Lancaster, J.M.; Lanchbury, J.S.; et al. Evaluation of the 12-Gene Molecular Score and the 21-Gene Recurrence Score as Predictors of Response to Neo-adjuvant Chemotherapy in Estrogen Receptor-Positive, HER2-Negative Breast Cancer. Ann. Surg. Oncol. 2020, 27, 765–771. [Google Scholar] [CrossRef]
  108. Bear, H.D.; Wan, W.; Robidoux, A.; Rubin, P.; Limentani, S.; White, R.L.; Granfortuna, J.; Hopkins, J.O.; Oldham, D.; Rodriguez, A.; et al. Using the 21-gene assay from core needle biopsies to choose neoadjuvant therapy for breast cancer: A multicenter trial. J. Surg. Oncol. 2017, 115, 917–923. [Google Scholar] [CrossRef]
  109. Glück, S.; De Snoo, F.; Peeters, J.; Stork-Sloots, L.; Somlo, G. Molecular subtyping of early-stage breast cancer identifies a group of patients who do not benefit from neoadjuvant chemotherapy. Breast Cancer Res. Treat. 2013, 139, 759–767. [Google Scholar] [CrossRef] [PubMed]
  110. Van de Vijver, M.J.; He, Y.D.; van ’t Veer, L.J.; Dai, H.; Hart, A.A.M.; Voskuil, D.W.; Schreiber, G.J.; Peterse, J.L.; Roberts, C.; Marton, M.J.; et al. A Gene-Expression Signature as a Predictor of Survival in Breast Cancer. N. Engl. J. Med. 2002, 347, 1999–2009. [Google Scholar] [CrossRef] [Green Version]
  111. Whitworth, P.; Stork-Sloots, L.; de Snoo, F.A.; Richards, P.; Rotkis, M.; Beatty, J.; Mislowsky, A.; Pellicane, J.V.; Nguyen, B.; Lee, L.; et al. Chemosensitivity Predicted by BluePrint 80-Gene Functional Subtype and MammaPrint in the Prospective Neoadjuvant Breast Registry Symphony Trial (NBRST). Ann. Surg. Oncol. 2014, 21, 3261–3267. [Google Scholar] [CrossRef] [Green Version]
  112. Whitworth, P.; Beitsch, P.; Mislowsky, A.; Pellicane, J.V.; Nash, C.; Murray, M.; Lee, L.A.; Dul, C.L.; Rotkis, M.; Baron, P.; et al. Chemosensitivity and Endocrine Sensitivity in Clinical Luminal Breast Cancer Patients in the Prospective Neoadjuvant Breast Registry Symphony Trial (NBRST) Predicted by Molecular Subtyping. Ann. Surg. Oncol. 2017, 24, 669–675. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Paik, S.; Shak, S.; Tang, G.; Kim, C.; Baker, J.; Cronin, M.; Baehner, F.L.; Walker, M.G.; Watson, D.; Park, T.; et al. A Multigene Assay to Predict Recurrence of Tamoxifen-Treated, Node-Negative Breast Cancer. N. Engl. J. Med. 2004, 351, 2817–2826. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Albain, K.S.; Barlow, W.E.; Shak, S.; Hortobagyi, G.N.; Livingston, R.B.; Yeh, I.T.; Ravdin, P.; Bugarini, R.; Baehner, F.L.; Davidson, N.E.; et al. Prognostic and predictive value of the 21-gene recurrence score assay in postmenopausal women with node-positive, oestrogen-receptor-positive breast cancer on chemotherapy: A retrospective analysis of a randomised trial. Lancet Oncol. 2010, 11, 55–65. [Google Scholar] [CrossRef] [Green Version]
  115. Kalinsky, K.; Barlow, W.E.; Meric-Bernstam, F.; Gralow, J.R.; Albain, K.S.; Lin, N.; Edith, N.L.; Perez, A.; Goldstein, L.J.; Chia, S.; et al. First results from a phase III randomized clinical trial of standard adjuvant endocrine therapy (ET) +/− chemotherapy (CT) in patients (pts) with 1-3 positive nodes, hormone receptor-positive (HR+) and HER2-negative (HER2-) breast cancer (BC) with recurrence score (RS) <25: SWOG S1007 (RxPonder). Cancer Res. 2021, 81 (Suppl. S4), GS3-00. [Google Scholar] [CrossRef]
  116. Sparano, J.A.; Gray, R.J.; Makower, D.F.; Pritchard, K.I.; Albain, K.S.; Hayes, D.F.; Geyer, C.E., Jr.; Dees, E.C.; Goetz, M.P.; Olson, J.A., Jr.; et al. Adjuvant Chemotherapy Guided by a 21-Gene Expression Assay in Breast Cancer. N. Engl. J. Med. 2018, 379, 111–121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Iwata, H.; Masuda, N.; Yamamoto, Y.; Fujisawa, T.; Toyama, T.; Kashiwaba, M.; Ohtani, S.; Taira, N.; Sakai, T.; Hasegawa, Y.; et al. Validation of the 21-gene test as a predictor of clinical response to neoadjuvant hormonal therapy for ER+, HER2-negative breast cancer: The TransNEOS study. Breast Cancer Res. Treat. 2019, 173, 123–133. [Google Scholar] [CrossRef] [Green Version]
  118. Ueno, T.; Masuda, N.; Yamanaka, T.; Saji, S.; Kuroi, K.; Sato, N.; Takei, H.; Yamamoto, Y.; Ohno, S.; Yamashita, H.; et al. Evaluating the 21-gene assay Recurrence Score® as a predictor of clinical response to 24 weeks of neoadjuvant exemestane in estrogen receptor-positive breast cancer. Int. J. Clin. Oncol. 2014, 19, 607–613. [Google Scholar] [CrossRef] [Green Version]
  119. Filipits, M.; Rudas, M.; Jakesz, R.; Dubsky, P.; Fitzal, F.; Singer, C.F.; Dietze, O.; Greil, R.; Jelen, A.; Sevelda, P.; et al. A new molecular predictor of distant recurrence in ER-positive, HER2-negative breast cancer adds independent information to conventional clinical risk factors. Clin. Cancer Res. 2011, 17, 6012–6020. [Google Scholar] [CrossRef] [Green Version]
  120. Chow, L.W.C.; Morita, S.; Chow, C.Y.C.; Ng, W.K.; Toi, M. Neoadjuvant palbociclib on ER+ breast cancer (N007): Clinical response and EndoPredict’s value. Endocr. Relat. Cancer 2018, 25, 123–130. [Google Scholar] [CrossRef] [Green Version]
  121. Turnbull, A.K.; Arthur, L.M.; Renshaw, L.; Larionov, A.A.; Kay, C.; Dunbier, A.K.; Thomas, J.S.; Dowsett, M.; Sims, A.H.; Dixon, J.M.; et al. Accurate prediction and validation of response to endocrine therapy in breast cancer. J. Clin. Oncol. 2015, 33, 2270–2278. [Google Scholar] [CrossRef]
  122. Selli, C.; Sims, A.H. Neoadjuvant Therapy for Breast Cancer as a Model for Translational Research. Breast Cancer Basic Clin. Res. 2019, 13, 117822341982907. [Google Scholar] [CrossRef]
  123. Mello-Grand, M.; Singh, V.; Ghimenti, C.; Scatolini, M.; Regolo, L.; Grosso, E.; Zambelli, A.; Da Prada, G.A.; Villani, L.; Fregoni, V.; et al. Gene expression profiling and prediction of response to hormonal neoadjuvant treatment with anastrozole in surgically resectable breast cancer. Breast Cancer Res. Treat. 2010, 121, 399–411. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Miller, T.W.; Balko, J.M.; Ghazoui, Z.; Dunbier, A.; Anderson, H.; Dowsett, M.; González-Angulo, A.M.; Mills, G.B.; Miller, W.R.; Wu, H.; et al. A gene expression signature from human breast cancer cells with acquired hormone independence identifies MYC as a mediator of antiestrogen resistance. Clin. Cancer Res. 2011, 17, 2024–2034. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Liang, X.; Briaux, A.; Becette, V.; Benoist, C.; Boulai, A.; Chemlali, W.; Schnitzler, A.; Baulande, S.; Rivera, S.; Mouret-Reynier, M.A.; et al. Molecular profiling of hormone receptor-positive, HER2-negative breast cancers from patients treated with neoadjuvant endocrine therapy in the CARMINA 02 trial (UCBG-0609). J. Hematol. Oncol. 2018, 11, 124. [Google Scholar] [CrossRef] [Green Version]
  126. Inda, M.A.; Blok, E.J.; Kuppen, P.J.K.; Charehbili, A.; den Biezen-Timmermans, E.C.; van Brussel, A.; Fruytier, S.E.; Meershoek-Klein Kranenbarg, E.; Kloet, S.; van der Burg, B.; et al. Estrogen receptor pathway activity score to predict clinical response or resistance to neoadjuvant endocrine therapy in primary breast cancer. Mol. Cancer Ther. 2020, 19, 680–689. [Google Scholar] [CrossRef] [Green Version]
  127. Lim, J.S.J.; Wong, A.L.A.; Ow, S.G.W.; Eng, L.S.; Sundar, R.; Chan, G.H.J.; Yadav, K.; Heong, V.; Peng Tan, D.S.; Soo, R.A.; et al. A phase Ib/II trial of lenvatinib (len) and letrozole (let) incorporating pharmacodynamics studies in postmenopausal women with hormone receptor positive (HR+) locally advanced/metastatic breast cancer (LABC/MBC). J. Clin. Oncol. 2019, 37 (Suppl. S15), 1045. [Google Scholar] [CrossRef]
  128. Da Silva, L.R.; de Andrade, C.A.; Brenelli, F.; Ramalho, S.; Reinert, T.; da Silva, A.E.R.; da Silva, A.E.R.; Kraft, M.B.; de Vasconcelos, V.C.A.; Frasson, A.L.; et al. Real-world data on neoadjuvant endocrine therapy in ER-positive/HER2-negative breast cancer. Breast Cancer Res. Treat. 2021, 186, 753–760. [Google Scholar] [CrossRef]
Table 1. Clinical trials comparing NET vs. NCT.
Table 1. Clinical trials comparing NET vs. NCT.
Clinical TrialNoCharacteristicsChemotherapyEndocrine TherapyPrimary
Endpoint
ResponseBCS Rate
Semiglazov et al., 2007 [15]239Postmenopausal ER+ and/or PR + stage IIA − IIIBDoxorubicin+ paclitaxel × 4 cyclesAnastrozole/
Exemestane
× 12 wk
CR64% CT vs. 64% ET24% CT vs. 33% ET
Alba et al., 2012 (GEICAM 2006-03) [13]95Pre and postmenopausal ER+/PR+/HER2−EC × 4, followed by docetaxel × 4Exemestane × 24 wk (+goserelin if premenopausal)OR
(RECIST, MRI)
66% CT vs. 48% ET47% CT vs. 56% ET
Palmieri et al., 2014 (NEOCENT) [16]44Postmenopausal ER+5-FU + EC × 6,
switched to docetaxel
after 3 cycles if stability/
progression
Letrozole × 18 wkOR
(US,
mammogram)
55% CT vs. 59% ET55% CT vs. 68% ET
ER = Estrogen receptor; PR = Progesteron receptor; CR = clinical response; CT = chemotherapy; EC = epirubicin + cyclophosphamide; ET = endocrine therapy; OR = objective response; US = ultrasound, wk = week.
Table 2. Studies comparing tamoxifen vs. aromatase inhibitors vs. fulvestrant.
Table 2. Studies comparing tamoxifen vs. aromatase inhibitors vs. fulvestrant.
Clinical TrialNo of
Patients
Duration (months)DrugsOutcomes
P024 (2001) [7]3374Letrozole
vs. TMX
CRR 55% vs. 36% (p < 0.001)
RRR 35% vs. 25% (p = 0.042)
BCS 45% vs. 35% (p = 0.022)
PROACT (2005) [46]4513Anastrozole
vs. TMX
CRR 50.0% vs. 46.2% (p = 0.37)
RRR 39.5% vs. 35.4% (p = 0.29)
BCS: 43% vs. 30.8% (p = 0.04)
IMPACT (2006) [8]3303Anastrozole vs.
TMX vs.
Anastrozole + TMX
CRR 38% vs. 36% vs. 39% (ns)
RRR 24% vs. 20% vs. 28%
BCS: 43% vs. 30.8% vs. 24% (NS)
SEMIGLAZOV [47]1513Exemestane vs. TMXOR 76% vs. 40% (p = 0.05)
BCS 37% vs. 20% (p = 0.05)
STAGE (2012) [12]1976Anastrozole + Goserelin vs.
TMX + Goserelin
CRR 70.4% vs. 50.5% (p = 0.004)
RRR 58.2% vs. 42.4%, (p = 0.027)
BCS 86% vs. 68%
ACOSOG Z1031 (2011) [48]3774Exemestane vs.
Letrozole vs.
Anastrozole
CRR 62.9% vs. 74.8% vs. 69.1% (NS)
CARMINA (2016) [49]1164 or 6Anastrozole vs.
Fulvestrant
CRR 52.6% vs. 36.8% (NS)
BCS 57.6% vs. 50% (NS)
QUENEL-TUEUX (2016) [50]1086Anastrozole vs.
Fulvestrant
CRR 59% vs. 54% (NS)
BCS 59% vs. 50%
TMX = tamoxifen; CRR = clinical response rate; OR = objective response; RRR = radiological response rate; BCS = breast conserving surgery, ns = non-significant.
Table 3. Duration of NET.
Table 3. Duration of NET.
StudyNo of PatientsType of NETDuration (months)AssessmentOutcomes
Krainick-Strobel et al. 2008 [50]32Letrozole4 to 8Monthly: palpation
3 monthly:
MRI, Mammogram, or US
4 mo 55% ORR 71% BCS
8 mo 72.4% ORR/>80% BCS
Dixon et al.
2009 [49]
182Letrozole3 vs. >3Clinical and US measurement (0,2,6,12 wk)
Mammogram (0.12 wk)
Review/3 mo
3 mo: 70% ORR 60% BCS
>3 mo: 83% ORR
72% BCS
Llombart-Cussac et al. 2012 [51]70Letrozole4–12Monthly: clinical examination
Mammogram and US/8wk
for first 4 mo
76.8% ORR (25% CR and 51.8% PR)
43% BCS
Allevi et al. 2013 [54]120Letrozole4,8 or 12Monthly: clinical palpation (caliper)
Mammogram and US at baseline and before surgery
4 mo: pCR 2.5%; ORR 45%; BCS 80%
8 mo: pCR 5%; ORR 86.8%; BCS 85%
12 mo: pCR 17.5%; ORR 95%; BCS 87.5%
Hojo et al. 2013 [56]52Exemestane4 vs. 6Monthly: caliper measurement and toxicity assessment
Ultrasound and Mammogram
if progression suspected
4 mo: pCR 0%; ORR 42.3%; BCS 50%
6 mo: pCR 4 %; ORR 48%; BCS 48%
Carpenter et al. 2014 [52]139Letrozoleup to 12Clinical examination and bimodal US/2 mo until BCSORR 85% (3.2% CR and 81.5% PR)
66% BCS
Median time to achieve tumor response to allow BCS: 7.5 mo
Fontein et al. 2014 [53]102Exemestane3 to 6Monthly: clinical palpation
3 monthly:
MRI, Mammogram, or US
pCR: 0.98%
3 mo: ORR 58.7% BCS 58.7%
>3 mo: ORR 68.3%; BCS 70.6%
Rusz et al. 2015 [55]42Letrozole12Clinical palpation every 3 months. Imaging as necessarypCR: 14.3% operated cases
ORR: 88%
BCS: 45%
Mo = month; wk = week; US = ultrasound; pCR = pathological Complete Response; ORR = Objective Response Rate; CR = Clinical Response; PR = Partial Response; BCS = breast-conserving surgery.
Table 4. PEPI score.
Table 4. PEPI score.
Pathological Characteristics of Surgical SpecimenRFSBCSS
HRScoreHRScore
Tumor size
T1/2
T3/4

-
2.8

0
3

-
4.4

0
3
Nodal status
Negative
Positive

-
3.2

0
3

-
3.9

0
3
Ki67 level
0%–2.7%
>2.7%–7.3%
>7.3%–19.7%
>19.7%–53.1%
>53.1%

-
1.3
1.7
2.2
2.9

0
1
1
2
3

-
1.4
2.0
2.7
3.8

0
1
2
3
3
ER status (Allred score)
0–2
3–8

2.8
-

3
0

7.0
-

3
0
With permission from Ellis M. et al. [85]. HR = hazard ratio; RFS = relapse free survival; BCSS = breast cancer specific survival.
Table 5. NET and Targeted Therapies. Phase II trials.
Table 5. NET and Targeted Therapies. Phase II trials.
Drug GroupStudyNo of PatientsAITargeted
Therapy
DesignOutcomespCR
PI3K
Inhibitors
LORELEI
[88]
334LTaselisib
(T)
L + T/L + PORR
50%/39%
p = 0.049

1.8%/0.6%
ns
NEO-ORB
[89]
257LAlpelisib
(Al)
L + P/L + AlORR
61.0%/63.4%
ns

1.7%/2.8%
ns
Tyr Kin
inhibitors
Guarneri et al.
[91]
92LLapatinib
(Lp)
L + P/L + LpORR
63%/70%
ns
93% ORR in PI3K mut
CDK4/6
inhibitors
MONALEESA-1 [90]14LRibociclib
400 mg (R400)
600 mg (R600)
L 2 wk/
L + R400 2 wk/
L + R600 2 wk
Higher Ki67 reduction
in the R arm
-
NeoPalAna
[92]
50APalbociclib
(Pal)

A → A + Pal
CCCA
higher after adding Pal
C1D15/C1D1
87%/26%,
p < 0.001
-
NeoPAL
[93]
106LPalbociclib
(Pal)
L + Pal 19 wk
vs.
FEC/21 d × 3 +
docetaxel/21 d × 3
RCB
higher in the CT arm
BCS equal in both arms
PEPI 0
17.6%/8.0%
3.8%/5.9%
NeoMONARCH [94]224AAbemaciclib
(Ab)
A/Ab/Ab + A 2 wk
and 2nd core biopsy
Ab + A 14 wk
CCCA
higher in Ab arms
14%/58%/68%
p < 0.001
4%
PALLET
[95]
307LPalbociclib
(Pal)
L 14 wk/
L 2 wk → L + Pal 14 wk
Pal 2 wk → L + Pal 14 wk
L + Pal 14 wk
Ki67 reduction and CCCA higher in the L + Pal arm 90%/59%.
p < 0.001
CR
49.5%/54.3% ns
ns
1.1% in L vs. 3.3% in L + Pal
CORALLEEN
[96]
106LRibociclib
(R)
L + R 28 d × 6/
DC/21 d × 4
→ Paclitaxel/7 d × 12
ROR
at surgery
46.9%/46.1%
2%/3%
mTOR
inhibitor
Baselga et al.
[97]
270LEverolimus
(E)
L/L + E
4 mo
CR
59.1%/68.1%
ns

0.8/1.4%
ns
AI = Aromatase Inhibitor; L = Letrozole; A = Anastrozole; ORR = objective response rate; ns = non-significant; wk = week; mo = month; CCCA = complete cell cycle arrest (central Ki67 <2.7%); C1D1 = cycle 1 day 1; C1D15 = cycle 1 day 15; FEC = 5FU 500 mg/m2 + epirubicin 100 mg/m2 + cyclophosphamide 500 mg/m2; RCB = Residual Cancer Burden; CT = chemotherapy; CR = Clinical response; DC = doxorubicin + cyclophosphamide; CR = clinical response; → = followed by.
Table 6. Ongoing Trials with targeted therapies.
Table 6. Ongoing Trials with targeted therapies.
Targeted TherapyStudyPopulationDesignArmsPrimary EndpointsSecondary EndpointsStatus
CDK4/6
inhibitor
NCT04293393
(CARABELA)
Stages II-III
Surgery feasible
Phase
II
Arm A:
AC/21 days × 4 cycles → Paclitaxel/7 d × 12 wk or
3-weekly docetaxel/21 d × 4 cycles (wk)
Arm B:
letrozole plus abemaciclib ± LHRH up to 12 mo
RCBKi67 changes
RCB 0+ I versus RCB-II versus RCB-III
PEPI score
IEFS
Molecular downstaging for high-risk genomic groups
Recruiting
NCT03969121T > 15 mm
Ki67 > 14%
Phase
III
Arm A:
Letrozole (+LHRH if premenopausal) + placebo 16 wk
Arm B:
Letrozole(+LHRH if premenopausal) + palbociclib 16 wk
PEPI score
EPclin score
CRR
Ki67 change
pCR
BCS
Adverse events
Recruiting
NCT03819010
(DxCARTES)
T > 2 cm
Ki67 ≥ 20%
Phase
II
Pretreatment RS 18.25:
letrozole (+LHRH if premenopausal) + palbociclib/6 cycles
Pretreatment RS 26-100:
letrozole (+LHRH if premenopausal) + palbociclib/6 cycles
Difference on RS
pre- and post-
treatment
(molecular results)
Molecular changes
concordance rate between the RCB score (0–I vs. II–III) and pCR and post-treatment RS
Completed
NCT03065621
(NeoRHEA)
T2–T3
N0–N1
Phase
II
Single arm:
palbociclib 125 mg × 4 cycles + letrozole/tamoxifen
Biomarkers
of resistance
Radiological responseCompleted
NCT02712723ER+ >66%/
Allred score 6–8
Stage II-III
Phase
II
Arm A: letrozole + placebo 22 wk
Arm B: letrozole + ribociclib 600 mg 22 wk
Arm C: letrozole + ribocilib 400 mg 22 wk
PEPI score 0CCCA
pCR
BCS
RFS
Active, not recruiting
NCT02603679
(PREDIXLumB)
Luminal B any N
Luminal A N+
Phase
II
Arm A: Weekly paclitaxel 12 wk → switch to NET + palbociclib
Arm B: tamoxifen + palbociclib 12 wkArm C: AI + palbociclib 12 wk switch to
Arm D: goserelin + AI + palbociclib 12 wk weekly paclitaxel
RRR after 12 wkpORR
BCS
RFS
BCSS
OS
Recruiting
NCT02592083
(PREDIXLumA)
Luminal A
IDC
>40 y
Phase
II
Arm A:
ET (tamoxifen or AI or AI + goserelin)
Arm B:
ET + palbociclib
CRR
RRR
pORR
BCS
RFS
BCSS
OS
Recruiting
ImmunotherapyNCT03573648Stage II-IIIPhase
II
Arm A:
ET
Arm B:
ET + palbociclib (PET) in a 1:2 ratio.
After 1 cycle (1 mo) both arms will receive avelumab (A) × 3 cycles
CCRAdverse eventsRecruiting
NCT02997995Postmenopausal
Luminal A
Phase
II
1st phase:
immune attractant + exemestane 6 wk.
After three wk (± 3 days), a tumor biopsy
2nd phase:
Patients > 10% CD8+ cells in the tumor after 3 wk
a durvalumab 1500 mg Q4W IV,
combined with exemestane (25 mg daily), for six months.
pCRNumber of CD8+ T cell
Clinical response
Assessment of Ki67
Toxicities
Predictive value of Mutational load for efficacy of Durvalumab
Predictive value of PDL1
Completed
NCT03874325T2–T4c,
any N
Phase
II
Single arm:
1500 mg durvalumab i.v/4 wk × 6 cycles + anastrozole 1 mg
PEPI score = 0CRR
CPD
CSD
Active, no recruiting
Anti-VEGFNCT00773695Her-2neg BC
T > 25 mm
Phase
II
CT arm:
FEC 12 wk → taxane × 12 wk
FEC 12 wk → taxane × 12 wk + bevacizumab
ET arm: AI 24 wk
AI + bevacizumab
pCRpORR
type of surgery
Active,
recruiting
PI3K inhNCT01275859Postmenopausal
T2N0-3
Phase
II
Single arm:
letrozole 2.5 mg + lapatinib 1500 mg po for 18–21 wk
pCRCRR
RRR
DFS
OS
Completed
Tyr Kin inhNCT02562118ER+
Postmenopausal
Phase
I–II
Single arm:
lenvatinib × 2 wk → letrozole 2.5 mg daily + lenvatinib × 12 wk
CRRpCR
PFS
Recruiting
Ros inh.NCT04551495
(ROSALINE)
LIC negPhase
II
Single arm:
428-day of letrozole 2.5 mg + entrectinib 600 mg daily.
(+goserelin if premenopausal)
RCBpCR
ORR (by MRI)
Adverse events
Not yet
recruiting
Histone
deacetylase inh.
NCT04465097Stage II/IIIPhase
II
Single arm:
exemestane from week 1 to week 26
and tucidinostat BIW from week 3 to week 26
(+leuprorelin/goserelin if premenopausal)
ORR
evaluated by MRI
ORR evaluated by US
pCR
Adverse events
RCB
Recruiting
Other Ongoing Trials
Lobular breast cancerNCT02206984T > 1 cmPhase
II
(WoT)
Arm A: tamoxifen 21 d
Arm B: anastrozole 21 d
Arm C: fulvestrant day 1 and 14
Change in Ki67ER expression
ER gene expression
PR expression
Recruiting
NCT03397537T > 1 cmPhase
II
letrozole 2.5 mg × 6 mo
(+aGnRH if premenopausal)
ORR-Completed
PremenopausalNCT02535221>35 < 55 y
T2N0M0
Phase
III
Arm A: goserelin + TAM + AI (1st 4 wk TAM → AI)
Arm B: FEC × 4–6 cycles
RRR by USpCR (Miller-Paine)Recruiting
MammaPrint®NCT03900637Stage I–IIIA
BCS not feasible
Phase
II
MammaPrint® high risk: NCT × 4 followed by docetaxel × 4
MammaPrint® low risk: letrozole 2.5 mg × 16–24 wk
(+leuprorelin if premenopausal)
BCS
conversion rate
pCR
CRR
Tumor size reduction rate
DFS
IBTRBluePrint®
Recruiting
BC = breast cancer; ER = estrogen receptor; HR = hormone receptor; AC = Adriamycin-Cyclophosphamide; Inh = inhibitor; wk = week; mo = month; IDC = infiltrating ductal carcinoma; ILC = invasive lobular carcinoma; BCS= breast conserving surgery; IEFS = invasive event free-survival; CCCA = complete cell cycle arrest; RFS = relapse free survival; PFS: progression free survival; NCT = neoadjuvant chemotherapy; ET = endocrine therapy; AC = Adriamycin/Cyclophosphamide; FEC = Fluorouracil + epirubicin + cyclophosphamide; CCR = clinical complete response; CRR = clinical response rate; RRR = radiological response rate; DFS = disease free survival; IBTR = ipsilateral breast tumor recurrence; CPD = clinical progressive disease; CSD = clinical stable disease; ORR = overall response rate; pORR = pathological objective response rate; pCR = pathological complete response; RCB = residual cancer burden; WoT = window of opportunity trial; IHC = immunohistochemistry; TAM = tamoxifen; AI = aromatase inhibitor; aGnRH = gonadotropin-releasing hormone agonist; → = followed by.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Martí, C.; Sánchez-Méndez, J.I. The Present and Future of Neoadjuvant Endocrine Therapy for Breast Cancer Treatment. Cancers 2021, 13, 2538. https://doi.org/10.3390/cancers13112538

AMA Style

Martí C, Sánchez-Méndez JI. The Present and Future of Neoadjuvant Endocrine Therapy for Breast Cancer Treatment. Cancers. 2021; 13(11):2538. https://doi.org/10.3390/cancers13112538

Chicago/Turabian Style

Martí, Covadonga, and José Ignacio Sánchez-Méndez. 2021. "The Present and Future of Neoadjuvant Endocrine Therapy for Breast Cancer Treatment" Cancers 13, no. 11: 2538. https://doi.org/10.3390/cancers13112538

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop