Recent Developments in the Therapeutic Landscape of Advanced or Metastatic Hormone Receptor–Positive Breast Cancer

Article information

Cancer Res Treat. 2023;55(4):1065-1076
Publication date (electronic) : 2023 October 5
doi : https://doi.org/10.4143/crt.2023.846
1Department of Medicine, University of California San Francisco, San Francisco, CA, USA
2Cancer Research Institute, Seoul National University, Seoul, Korea
3Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
4Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
5Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
Correspondence: Seock-Ah Im, Department of Internal Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea Tel: 82-2-2072-0850 Fax: 82-2-762-9662 E-mail: moisa@snu.ac.kr
*Eunice Yoojin Lee and Dae-Won Lee contributed equally to this work.
Received 2023 July 18; Accepted 2023 October 2.

Abstract

Hormone receptor–positive (HR+) disease is the most frequently diagnosed subtype of breast cancer. Among tumor subtypes, natural course of HR+ breast cancer is indolent with favorable prognosis compared to other subtypes such as human epidermal growth factor protein 2–positive disease and triple-negative disease. HR+ tumors are dependent on steroid hormone signaling and endocrine therapy is the main treatment option. Recently, the discovery of cyclin-dependent kinase 4/6 inhibitors and their synergistic effects with endocrine therapy has dramatically improved treatment outcome of advanced HR+ breast cancer. The demonstrated efficacy of additional nonhormonal agents, such as targeted therapy against mammalian target of rapamycin and phosphatidylinositol 3-kinase signaling, poly(ADP-ribose) polymerase inhibitors, antibody-drug conjugates, and immunotherapeutic agents have further expanded the available therapeutic options. This article reviews the latest advancements in the treatment of HR+ breast cancer, and in doing so discusses not only the development of currently available treatment regimens but also emerging therapies that invite future research opportunities in the field.

Introduction

Breast cancer is the most commonly diagnosed cancer worldwide. It is a leading cause of death among women worldwide, with an estimated 2.3 million new cases each year and accounting for over 680,000 deaths in 2020 [1].

Breast cancer is not a single disease entity. Microarray-based gene expression profiling has identified four intrinsic subtypes of breast cancer, which include luminal A, luminal B, human epidermal growth factor protein 2 (HER2)–overexpression, and triple-negative breast cancer (TNBC) [2]. Each subtype also corresponds to a distinct histopathological profile based on the expression of hallmark biomarkers, namely the hormone receptors (HR) for estrogen (ER) or progesterone, HER2, and the cell proliferation marker Ki67. Briefly, luminal A tumors are HR+, HER2–, and Ki67-low; luminal B tumors are further subdivided into HR+/HER2–/ Ki67-high and HR+/HER2+/Ki67-low subtypes; HER2- overexpression refers to HR–/HER2+ tumors; and TNBC is negative for both HR and HER2 [3]. The luminal A and B subtypes are collectively referred to as HR+ breast cancer, which represents over two-thirds of all breast cancer diagnoses and has a more favorable prognosis compared to TNBC [4]. This can be attributed to not only its natural history but also its dependence on estrogen signaling, which is targeted with endocrine therapy agents such as selective ER modulators (SERMs), aromatase inhibitors (AIs), and selective ER degraders (SERDs) [5].

While endocrine therapy continues to be a mainstay of treatment for HR+ breast cancer, the discovery of additional nonhormonal molecular targets—namely cyclin-dependent kinase 4/6 (CDK4/6), mammalian target of rapamycin (mTOR), protein kinase B (also known as Akt), and phosphatidylinositol 3-kinase (PI3K)—and their translation into novel therapeutic agents have further improved patient outcomes. Despite these milestones, advanced HR+ breast cancer remains an incurable condition, often with mechanisms independent of hormonal signaling that contribute to recurrence, therapeutic resistance, and/or metastasis, underscoring the need for further translational research in this realm. In this review, we will discuss the currently available and emerging therapeutic agents in advanced HR+ breast cancer, highlighting landmark studies that have paved their introduction into clinical practice.

Endocrine Therapy Plus CDK4/6 Inhibitors as First-Line Treatment for Advanced HR+ Breast Cancer

Endocrine therapy is the main treatment option for advanced HR+ breast cancer. Endocrine therapy can be broadly classified into SERMs, AIs, and SERDs. SERMs are competitive inhibitors of estrogen-ER binding and display either agonist or antagonist activity depending on the target tissue; for instance, tamoxifen, the most commonly used SERM, has anti-proliferative effects in breast tissue but agonistic effects in the endometrium, bone, and cardiovascular tissues [6]. AIs inhibit the conversion of androgens to estradiol in non-ovarian tissues and are used to reduce estrogen levels in the postmenopausal setting. Nonsteroidal AIs, which include anastrozole and letrozole, are noncovalent, reversible competitive inhibitors of androgen-aromatase binding, whereas the steroidal AI exemestane covalently and irreversibly binds to the aromatase substrate-binding site [7]. SERDs such as fulvestrant are a more recently developed class of endocrine agents that antagonize ER signaling and result in ER degradation. Endocrine therapy is effective in most patients with HR+ advanced breast cancer. However, cancer cells acquire new mutations or lose HR expression, developing endocrine therapy resistance over time [5]. Traditionally, primary endocrine resistance is defined as patients who relapse during the first 2 years of adjuvant endocrine therapy or disease progression within the first 6 months of first-line endocrine therapy for advanced or metastatic breast cancer. Secondary (acquired) resistance is defined as relapse during adjuvant endocrine therapy but after the first 2 years, relapse within 12 months of completing adjuvant endocrine therapy or disease progression 6 months after endocrine therapy for advanced or metastatic breast cancer [8].

In addition to endocrine therapy, the discovery of CDK4/6 inhibitors has further transformed the treatment landscape for advanced HR+ breast cancer. CDK4 and CDK6 are serinethreonine kinases that facilitate G1/S progression in the cell cycle by inhibiting the retinoblastoma pathway, and studies have shown that their activation contributes to an ER-independent growth signal in HR+ breast cancers [9]. In 2009, the CDK4/6 inhibitor, palbociclib was shown to exhibit growth inhibitory effects on ER+ breast cancer cell lines in vitro [10]. This included both synergy with tamoxifen as well as potency against cell lines with tamoxifen resistance, inviting a novel therapeutic opportunity for HR+ breast cancers refractory to endocrine therapy [10]. Subsequent drug discovery efforts also led to the development of ribociclib and abemaciclib; today, all three CDK4/6 inhibitors are approved by the U.S. Food and Drug Administration (FDA) and Ministry of Food and Drug Safety (MFDS) in Korea as first-line treatment for advanced HR+ breast cancer combined with endocrine therapy. Landmark studies of CDK4/6 inhibitors are summarized in Table 1 [11-26].

Landmark studies in dual CDK4/6 inhibitor and endocrine therapy for advanced HR+breast cancer

The phase II PALOMA-1 study demonstrated that firstline palbociclib and letrozole combination therapy significantly increased progression-free survival (PFS) compared to letrozole monotherapy in postmenopausal patients with advanced ER+/HER2– breast cancer (20.2 vs. 10.2 months; hazard ratio, 0.488; p=0.0004) [11]. These results, which were further confirmed by the phase III PALOMA-2 trial [12], led to accelerated U.S. FDA approval of palbociclib in combination with letrozole for ER+/HER2– breast cancer in 2015. The MONALEESA-2 trial showed increased PFS with ribociclib/letrozole compared to letrozole alone (25.3 vs. 16.0 months; hazard ratio, 0.568; p=9.63×10–8) [13,14], and MONALEESA-7 was the first study to demonstrate overall survival (OS) benefits for adding ribociclib to therapy with goserelin and either letrozole or tamoxifen (58.7 vs. 48.0 months; hazard ratio, 0.76; 95% confidence interval [CI], 0.61 to 0.96) [15,16] in premenopausal women. The PFS benefits of abemaciclib in combination with either anastrozole or letrozole relative to AI plus placebo was first demonstrated in the MONARCH-3 trial (28.2 vs. 14.8 months; hazard ratio, 0.540; p=0.000002) [17,27]. OS was also reported to be significantly prolonged in the abemaciclib arm (67.1 vs. 65.1 months; hazard ratio, 0.754; p=0.0301) [18].

The MONALEESA-3 trial investigated the efficacy of ribociclib plus fulvestrant as first-line or second-line treatment in advanced HR+ breast cancer patients. Around half of patients were treatment naïve for advanced disease and prior chemotherapy for advanced disease was not allowed. Overall, patients in the ribociclib plus fulvestrant arm showed prolonged PFS (20.5 vs. 12.8 months; hazard ratio, 0.593; p < 0.001) and OS (53.7 vs. 41.5 months; hazard ratio, 0.73; 95% CI, 0.59 to 0.90) relative to those in the control arm [19,28]. Subgroup analysis of those who had not received endocrine therapy in the advanced setting (treatment naïve patients) also showed prolonged PFS (32% vs. 51% event rate; hazard ratio, 0.577; 95% CI, 0.415 to 0.802) and OS (34.5% vs. 52.7% deaths; hazard ratio, 0.62; 95% CI, 0.41 to 0.95) in the ribociclib plus fulvestrant arm [19,28].

As a result, combined treatment with endocrine therapy and CDK4/6 inhibitors is recommended as first-line therapy for advanced HR+ breast cancer [8,29]. CDK4/6 inhibitors are effective in de novo or recurrent metastatic breast cancer, and in patients with primary or secondary endocrine resistance [8]. Although chemotherapy was previously regarded as first-line therapy in the setting of visceral crisis, studies are underway to evaluate the safety and efficacy of CDK4/6 inhibitors in this setting as well. Most recently, preliminary results from the phase II RIGHT choice trial reported a statistically significant PFS benefit among premenopausal advanced HR+/HER2– breast cancer patients with 10% or higher expression of ER by immunohistochemistry and aggressive features (rapidly progressing or highly symptomatic disease, including visceral crises) in the ribociclib+letrozole/anastrazole+goserelin arm relative to those receiving chemotherapy (24.0 vs. 13.0 months; hazard ratio, 0.54; p=0.0007) [30].

With regards to the preferred CDK4/6 inhibitor for treatment, the preferred agent is decided on a case-by-case basis based on patient risk factors for adverse events, as well as PFS and/or OS benefits seen in pivotal clinical trials [31]. Neutropenia is a common adverse effect of all three CDK4/6 inhibitors, though it is more frequently observed with palbociclib and ribociclib. For instance, across the three trials that investigated dual therapy with fulvestrant and a CDK4/6 inhibitor, the incidence of all grade neutropenia with palbociclib, ribociclib, and abemaciclib were 78.8%, 69.6%, and 46.0%, respectively [20,21,28]. This discrepancy has been attributed to the greater selectivity of abemaciclib for CDK4 over CDK6 [32,33]. Furthermore, unlike chemotherapy-induced neutropenia, neutropenia associated with CDK4/6 inhibition is rapidly reversible [32,33]. Diarrhea is more common with abemaciclib, which was observed in 86.4% of patients in the MONARCH-2 trial, whereas 19.1% and 29.0% of patients in the PALOMA-3 (palbociclib) and MONARCH-2 (ribociclib) trials reported diarrhea [20,21,28]. Overall, the consensus is that adverse events associated with CDK4/6 inhibitors are well managed with standard supportive therapy and when indicated, dose interruptions or reductions [32,33].

If the patients did not recur while on adjuvant nonsteroidal AI or within 12 months after completing therapy, AI plus CDK4/6 inhibitor is recommended. There was no clear benefit of fulvestrant plus CDK4/6 inhibitor over AI plus CDK4/6 inhibitor in a phase II study [34]. Likewise, AI plus CDK4/6 inhibitor is recommended in patients who had relapsed on adjuvant tamoxifen. Fulvestrant plus CDK4/6 inhibitors are recommended in patients who relapsed on adjuvant AI, or within 12 months after completing adjuvant AI therapy.

Endocrine Therapy Plus CDK4/6 Inhibitors as Second- or Subsequent-Line Treatment for Advanced HR+ Breast Cancer

Numerous studies have also shown the benefits of combined CDK4/6 inhibitor and endocrine therapy in the second-line or early relapse setting. In addition to demonstrating the benefits of combined ribociclib and endocrine therapy as first-line treatment for advanced HR+ breast cancer, the MONALEESA-7 trial also investigated the benefits of adding ribociclib in patients who received no more than one line of chemotherapy for metastasis. Around 14% of patients enrolled in the MONALEESA-7 trial received 1 line of chemotherapy for advanced disease. Compared to those who received goserelin and either letrozole or tamoxifen alone, those who received ribociclib, goserelin and endocrine therapy showed improved PFS (46.8% vs. 61.7%; hazard ratio, 0.55; 95% CI, 0.31 to 0.95) but not OS (53.2% vs. 65.9% deaths; hazard ratio, 0.747; 95% CI, 0.441 to 1.266) [16,22].

As for regimens with fulvestrant as the backbone, the PALOMA-3 trial was the first to investigate the efficacy of combined palbociclib and fulvestrant as second-line or later line of therapy for advanced HR+ breast cancer patients who progressed on prior endocrine therapy [20]. Patients who had received one prior line of chemotherapy in the advanced setting were also included. Specifically, patients on dual-palbociclib/fulvestrant therapy exhibited significantly longer PFS compared to the placebo/fulvestrant group (9.5 vs. 4.6 months; hazard ratio, 0.46; p < 0.0001) [23]. Additionally, patients who received one line of prior chemotherapy for metastasis exhibited prolonged PFS (7.7 vs. 3.5 months; HR, 0.43; 95% CI, 0.29 to 0.64). Subsequent analyses showed OS benefits when restricted to patients with sensitivity to previous endocrine therapy (39.7 vs. 29.7 months; hazard ratio, 0.72), although statistical significance was not reached in analyses of the entire trial group [35].

The MONARCH-2 trial also tested the efficacy of combined abemaciclib/fulvestrant therapy in patients who progressed while on or 12 months after neoadjuvant or adjuvant endocrine therapy, or while receiving endocrine therapy in the advanced setting. Since subgroup analyses were performed based on primary or secondary endocrine therapy resistance as defined by the European School of Oncology (ESO)/European Society of Medical Oncology (ESMO) guidelines [36], the specific PFS and OS values for patients who progressed on endocrine therapy for advanced breast cancer are unknown. Nonetheless, the overall cohort of individuals in the abemaciclib/fulvestrant arm exhibited prolonged PFS (16.4 vs. 9.3 months; hazard ratio, 0.553; p < 0.001) and OS (45.8 vs. 37.2 months; hazard ratio, 0.784; 95% CI, 0.644 to 0.955) compared to the placebo/fulvestrant arm [21,37]. For patients who received treatment as second-line therapy or who had early relapse MONALEESA-3 reported improved PFS (14.6 vs. 9.1 months; hazard ratio, 0.57; 95% CI, 0.44 to 0.74) and OS (40.2 vs. 32,5 months; hazard ratio, 0.73; 95% CI, 0.53 to 1.00) with dual ribociclib/fulvestrant therapy relative to placebo/fulvestrant [38].

Finally, abemaciclib is the only CDK4/6 inhibitor that is approved as monotherapy for treatment-refractory HR+/HER2– metastatic breast cancer. The phase II single-arm MONARCH-1 trial reported an overall response rate (ORR) of 19.7%, median PFS of 6.0 months, and a median OS of 17.7 months with abemaciclib monotherapy in a cohort where 90.2% of patients had visceral disease and 50.8% had at least three sites of metastasis [39]. More recently, the phase II nextMONARCH trial randomized patients with endocrinerefractory HR+/HER2– metastatic breast cancer who previously received chemotherapy to abemaciclib 150 mg+tamoxifen 20 mg (A+T), twice-daily abemaciclib 150 mg (A-150) monotherapy, or once-daily abemaciclib 200 mg+loperamide (A-200) for diarrhea prophylaxis. There were no significant PFS differences between A+T and A-150 (9.1 vs. 7.4 months; hazard ratio, 0.815; p=0.293) and between A-150 and A-200 (6.5 vs. 7.4 months; hazard ratio, 1.045; p=0.811) [40].

Endocrine Therapy Combined with Agents Targeting the PI3K/AKT/mTOR Pathway

There is growing evidence that aberrant activation of the PI3K/Akt/mTOR pathway in association with ER signaling underlies acquired resistance to endocrine therapy in HR+ breast cancer [41]. Studies have reported somatic gain-of-function mutations in PIK3CA, which encodes the tyrosine kinase PI3K, in approximately 40% of patients with HR+ breast cancer [42]. While CDK4/6 inhibitor combined with endocrine therapy is an initial treatment option for advanced HR+ breast cancer, there are additional agents for subsequent treatment targeting the PI3K/Akt/mTOR pathway. Current guidelines for this population recommend combination therapy with fulvestrant, a SERD, and the PI3K inhibitor, alpelisib as second-line therapy [8,31]. The SOLAR-1 trial is a phase III randomized controlled trial that compared fulvestrant with alpelisib to fulvestrant with placebo in HR+ breast cancer. Patients with PIK3CA mutations who received fulvestrant and alpelisib had prolonged PFS (11.0 vs. 5.7 months; hazard ratio, 0.65; p < 0.001) and ORR, (26.6% vs. 12.8%) compared to those who received fulvestrant and placebo [43]. The combination regimen did not significantly prolong PFS nor ORR in individuals without PIK3CA mutations. In the subsequent BYLieve trial, the efficacy of alpelisib plus fulvestrant was maintained in PIK3CA-mutated HR+ breast cancer, who had progression on a CDK4/6 inhibitor plus an aromatase inhibitor [44]. These results led to U.S. FDA and Korean MFDS approval of fulvestrant and alpelisib combination therapy in advanced HR+ breast cancer with PIK3CA mutations. However, alpelisib is not without its safety risks; hyperglycemia was one of the most common adverse effects in the SOLAR-1 trial with an incidence of 63.7% in the alpelisib/fulvestrant arm compared to 9.8% in the placebo/fulvestrant arm [43]. Of note, individuals with known type 1 diabetes or uncontrolled type 2 diabetes (hemoglobin A1c > 6.4% or fasting plasma glucose > 140 mg/dL) were excluded from the study. Other common side effects included gastrointestinal toxicities (i.e., diarrhea in 57.7% and 15.7%, respectively, nausea in 44.7% and 22.3%, decreased appetite in 35.6% vs. 10.5%) and rash (35.6% and 5.9%).

More recently, the phase II FAKTION trial reported that combination therapy with capivasertib, an AKT inhibitor, and fulvestrant prolonged PFS (10.3 vs. 4.8 months; hazard ratio, 0.56; p=0.0023) and OS (29.3 vs. 23.4 months; hazard ratio, 0.66; p=0.035) relative to placebo/fulvestrant in advanced HR+/HER2– breast cancer with relapse or progression on an AI [45,46]. A phase III CAPItello-291 trial investigated the safety and efficacy of this dual regimen in patients with AI-resistant advanced HR+/HER2– breast cancer, notably including those who have previously received CDK4/6 inhibitors [47]. PFS was significantly prolonged in both the overall cohort (7.2 vs. 3.6 months; hazard ratio, 0.60; p < 0.001) and in patients with genetic alterations in the AKT pathway (7.3 vs. 3.1 months; hazard ratio, 0.50; p < 0.001) [47].

Everolimus, an mTOR inhibitor, has also been investigated for advanced HR+ breast cancer previously treated with nonsteroidal AI. The phase III BOLERO-2 trial demonstrated improved PFS in advanced HR+ breast cancer patients on exemestane in conjunction with everolimus compared to exemestane alone (7.8 vs. 3.2 months; hazard ratio, 0.45; p < 0.0001) [48,49]. Although there was no significant improvement in OS [50], the combination therapy was approved for metastatic HR+ breast cancer that progressed on an AI in 2012. Subsequent analyses on the cell-free DNA for activating mutations in the ER-encoding ESR1 gene found that while patients with ESR1 mutations overall exhibited shorter PFS compared to those with wild-type ESR1, those with D538G mutations who received exemestane and everolimus demonstrated a similar PFS to wild-type individuals [51]. These results highlight the relationship between ER and mTOR signaling, as well as the utility of circulating biomarkers for monitoring therapeutic response and prognosis.

Oral SERDs as Backbone Endocrine Therapy Agents in Advanced HR+ Breast Cancer

Despite the efficacy of fulvestrant as a backbone endocrine therapy agent in combined treatment regimens for advanced HR+ breast cancer, it is available only via intramuscular injection due to its poor oral bioavailability, which imposes a practical barrier to its long-term use. As a result, there have been considerable efforts to develop orally available SERDs such as elacestrant, which demonstrated improved PFS over standard-of-care endocrine monotherapy in the phase III EMERALD trial (6-month PFS rates 34.3% vs. 20.4%; 12-month PFS rates 22.3% vs. 9.4%; HR, 0.70; p=0.0018) [52]. Importantly, the trial enrolled patients with advanced HR+/ HER2– breast cancer who progressed on previous dual therapy with a CDK4/6 inhibitor and fulvestrant or an AI, and PFS benefits were also observed among patients with ESR1 mutations (6-month PFS rates 40.8% vs. 19.1%; 12-month PFS rates 26.8% vs. 8.2%; hazard ratio, 0.55; p=0.0005). Moreover, improved PFS was reported when elacestrant was specifically compared against fulvestrant in both the overall cohort (6-month PFS rates 40.8% vs. 20.8%; 12-month PFS rates 26.8% vs. 8.4%; hazard ratio, 0.50; p=0.0005) and in patients with altered ESR1 (6-month PFS rates 34.3% vs. 22.9%; 12-month PFS rates 22.3% vs. 10.2%; p=0.0049). In 2023, elacestrant was FDA-approved for postmenopausal women or adult men with HR+/HER2–, ESR1-mutated advanced or metastatic breast cancer who progressed on at least one line of endocrine therapy.

In addition to elacestrant, many oral SERDs are under investigation [53]. In patients with HR+/HER2– breast cancer who progressed after at least one line of endocrine therapy, camizestrant improved PFS at both the 75 mg (7.2 vs. 3.7 months; hazard ratio, 0.58; 95% CI, 0.41 to 0.81; p=0.0124) and 150 mg (7.7 vs. 3.7 months; hazard ratio, 0.67; 95% CI, 0.48 to 0.92; p=0.0161) doses compared to fulvestrant in the SERENA-2 trial [54]. The phase III SERENA-4 and SERENA-6 trials are currently enrolling patients to investigate the relative efficacy of combination therapy with a CDK4/6 inhibitor and either camizestrant or an AI [55,56]. In the acelERA BC trial which included HR+/HER2– breast cancer who progressed after at least one line of endocrine therapy, although the study did not meet its primary endpoint, giredestrant showed numerical improvement of PFS compared to physician choice of endocrine monotherapy especially in those with ESR1 mutation (5.3 vs. 3.5 months; hazard ratio, 0.60; 95% CI, 0.35 to 1.03, p=0.061) [57]. Finally, newer classes of agents that target the ER, including proteolysis targeting chimeras (PROTACs), selective ER covalent antagonists (SERCAs), and complete ER antagonists (CERANs), are currently being developed and tested for use in both the early and advanced settings [53]. Briefly, PROTACs simultaneously bind to the ER and an E3 ubiquitin ligase for ubiquitinmediated proteasomal degradation of the ER; SERCAs result in ER inactivation via a covalent bond to a key cysteine residue of wildtype and mutant ER proteins; CERANs inhibit ER-mediated transcriptional activation [58,59].

Emerging Non-endocrine Therapies in Advanced HR+ Breast Cancer

1. Poly(ADP-ribose) polymerase inhibitors for BRCA1/2-mutant cancer

Perhaps the most well-established genetic factor for breast cancer are pathogenic mutations in BRCA1/2, which are tumor suppressor genes that encode DNA repair enzymes involved in homologous recombination. Briefly, the recognition of a double-strand break (DSB) leads to the phosphorylation of BRCA1 as well as other downstream targets, which together act as a scaffold for the complex machinery that mediates DSB repair [60]. The ends of damaged DNA are then resected to form single-strand templates for repair, after which BRCA2, in a complex with additional DNA repair proteins, loads the critical RAD51 protein onto the site of damage for strand invasion and homologous recombination [60]. Pathogenic BRCA1/2 mutations are highly penetrant and significantly increase lifetime risk of breast cancer in an autosomal dominant fashion, accounting for approximately 5%-10% of breast cancers [61,62]. Another key family of DNA repair enzymes are poly(ADP-ribose) polymerase (PARP) proteins, which facilitate single-strand break (SSB) repair as well as base excision repair. Suppression of PARP activity leads to the accumulation of unresolved SSBs that degrade into DSBs, which in BRCA1/2-mutant cells that are incapable of HR, results in genomic instability and cell death [61]. This phenomenon has been leveraged in the development of PARP inhibitors as therapeutic agents for breast cancer. The phase III OlympiAD trial demonstrated the efficacy of monotherapy with the PARP inhibitor olaparib compared to single-agent chemotherapy in HER2– metastatic breast cancer patients with a germline BRCA1/2 mutation [63]. Significantly prolonged PFS was reported in the olaparib arm (7.0 vs. 4.2 months; hazard ratio, 0.58; p < 0.01), and while there was no significant improvement in OS (19.3 vs. 17.1 months; hazard ratio, 0.90; p=0.513), subgroup analyses suggested potential benefit in individuals who had not received prior chemotherapy in the metastatic setting [63,64]. Similarly, talazoparib also exhibited significantly prolonged PFS (8.6 vs. 5.6 months; hazard ratio, 0.54; p < 0.001) and increased ORR (62.6% vs. 27.2%; odds ratio, 5.0; p < 0.001) in germline BRCA1/2 mutant advanced breast cancer relative to singleagent chemotherapy in the phase III EMBRACA trial [65]. Both medications have been approved for germline BRCA1/2 mutant advanced breast cancer as of 2014 and 2018, respectively. Consideration for monotherapy with PARP inhibitors is also recommended for patients with pathogenic variations in PALB2, which encodes a protein that co-localizes with and stabilizes BRCA2 to mediate RAD51 loading for homologous recombination [8].

2. HER2-directed therapy for HER2-low cancers

The development of HER2-directed therapy is considered one of the greatest advancements in breast cancer treatment, and trastuzumab, a monoclonal anti-HER2 antibody, is a well-established agent for the treatment of HER2+ breast cancer. Nonetheless, there is growing evidence for HER2- therapy in metastatic HR+ tumors with low expression of HER2 (HER2 1+ or 2+ on immunohistochemistry and negative on fluorescence in situ hybridization), which account for 45%-55% of breast cancers [66]. Trastuzumab-deruxtecan (T-Dxd) is an antibody-drug conjugate (ADC) in which trastuzumab is linked to the topoisomerase I inhibitor deruxtecan. Upon its release into the HER2-expressing target cell, the deruxtecan “payload” portion of T-DXd exerts a potent, cytotoxic “bystander effect” on surrounding cells, including HER2– cells [67]. A phase Ib study of T-DXd demonstrated antitumor effects in patients with advanced HER2-low breast cancer refractory to standard therapy (median duration of response 10.4 months; 95% CI, 8.8 months to not evaluable) [68]. Following this, the DESTINY-Breast04 phase III randomized clinical trial demonstrated that T-DXd significantly prolonged both PFS (10.1 vs. 5.4 months; hazard ratio, 0.51; p < 0.001) and OS (23.9 months vs. 17.5 months; hazard ratio, 0.64; p=0.003) compared to the physician’s choice of chemotherapy [69]. These results held true not just for the HR+ group, but also among all enrolled patients regardless of HR+ disease, and led to U.S. FDA approval of T-DXd for advanced HER2-low breast cancer.

3. Sacituzumab govitecan

Sacitizumab-govitecan is another ADC that is being actively investigated for use in HR+ tumors. Recently approved for metastatic TNBC, the ADC consists of an anti-Trop2 antibody and a topoisomerase I payload. Trop2, which is overexpressed in a variety of malignant tumors including invasive breast cancer, is a transmembrane glycoprotein that upregulates proliferative markers while downregulating pro-apoptotic proteins to ultimately promote tumor growth [70]. A phase I/II trial of 54 patients with metastatic HR+/HER2– who progressed despite endocrine therapy and received at least one round of chemotherapy in the metastatic setting demonstrated encouraging results with an ORR of 31.5% (95% CI, 19.5 to 45.6) and a PFS of 5.5 months (95% CI, 3.6 to 7.6) [71]. Recently, a phase III TROPiCS-02 trial demonstrate an improved PFS (5.5 vs. 4.0 months; HR, 0.66; p=0.0003) and OS (14.4 vs. 11.2 months; HR, 0.79; p=0.020) with sacituzumab-govitecan compared to conventional chemotherapy in patients with relapsed or refractory HR+/HER2– breast cancer [72,73].

4. Immunotherapy in advanced HR+ breast cancer

In line with recent breakthroughs in immunotherapy for solid tumor malignancies, there are increasing efforts to adopt immune checkpoint inhibitors into available therapeutic agents for breast cancer. Immune checkpoint inhibitors were initially introduced in the setting of TNBCs, since HR+ tumors have been traditionally described as “immunologically cold” with low numbers of tumor-infiltrating lymphocytes. The limited efficacy of the anti-PD1 antibody pembrolizumab in the phase Ib KEYNOTE-028 trial, which enrolled metastatic HR+/HER2– breast cancer patients with >1% programmed death-ligand 1 (PD-L1)–expressing stroma or tumor cells (ORR, 12%; 95% CI, 2.5 to 31.2; no complete responses) [74], has been attributed to this finding; however, there is active research into the mechanisms underlying the architecture of the tumor microenvironment in HR+ breast cancer and ways to increase its susceptibility to cancer immunotherapy [75]. Similarly, initial trials testing the combination of pembrolizumab with chemotherapeutic agents such as eribulin or capecitabine did not demonstrate significant PFS or ORR benefits [76,77]. However, a more recent single-arm phase II trial of pembrolizumab/ eribulin therapy in metastatic HR+ breast cancer reported promising results (median PFS of 6.0 months; 95% CI, 3.7 to 8.4; 1-year OS, 59.1%; 95% CI, 45.8 to 76.2), which authors attributed to the degree of pre-treatment with chemotherapy and differences in PD-L1 scoring methods [78]. In addition, nivolumab plus eribulin showed ORR of 53.3% and median PFS of 5.6 months (95% CI, 4.3 to 6.8) in a phase IB/II study of HR+HER2– patients [79]. The currently ongoing KEYNOTE-B49 phase III trial is also evaluating the comparative efficacy of pembrolizumab in combination with one of four chemotherapy regimens, paclitaxel, nab-paclitaxel, doxorubicin, or capecitabine [80]. Additionally, a phase I/II trial of triple therapy with palbociclib, pembrolizumab, and letrozole in metastatic HR+/HER2– breast cancer observed 31% and 25% complete and partial response rates, inviting further research into the potential synergistic effects of immune checkpoint therapy with CDK4/6 inhibitors [81]. The role for immune checkpoint inhibitor-based therapeutic strategies in advanced HR+ breast cancer therefore remains an open question that warrants continued investment to maximize their potential benefits.

In summary, both existing and emerging therapeutic agents for advanced HR+ breast cancer serve as a testament to the wealth of recent scientific and clinical developments that have transformed disease management, prognosis, and patient outcomes (Fig. 1). In turn, these discoveries have fueled further research into disease biology that ensure the continued evolution of available treatment regimens for advanced HR+ breast cancer. Key unanswered questions include the identification of biomarkers for disease monitoring, response to targeted therapy, and/or resistance, optimization of combined treatment regimens—particularly those that prolong OS—and the minimization of treatment-related toxicity.

Fig. 1.

Treatment of advanced/metastatic hormone receptor (HR)–positive, human epidermal growth factor protein 2 (HER2)–negative breast cancer. ESR1, estrogen receptor 1; ET, endocrine therapy; OFS, ovarian function suppression; PD, progressive disease; SERD, selective estrogen receptor degrader; T-Dxd, trastuzumab-deruxtecan; ULN, upper limit of normal.

Notes

Author Contributions

Wrote the paper: Lee EY, Lee DW, Lee KH, Im SA.

Revision: Lee EY, Lee DW, Lee KH, Im SA.

Conflicts of Interest

Conflict of interest relevant to this article was not reported.

References

1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2021;71:209–49.
2. Sorlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci U S A 2001;98:10869–74.
3. Goldhirsch A, Wood WC, Coates AS, Gelber RD, Thurlimann B, Senn HJ, et al. Strategies for subtypes: dealing with the diversity of breast cancer: highlights of the St. Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2011. Ann Oncol 2011;22:1736–47.
4. Zhang MH, Man HT, Zhao XD, Dong N, Ma SL. Estrogen receptor-positive breast cancer molecular signatures and therapeutic potentials (Review). Biomed Rep 2014;2:41–52.
5. Huppert LA, Gumusay O, Idossa D, Rugo HS. Systemic therapy for hormone receptor-positive/human epidermal growth factor receptor 2-negative early stage and metastatic breast cancer. CA Cancer J Clin 2023;73:480–515.
6. Patel HK, Bihani T. Selective estrogen receptor modulators (SERMs) and selective estrogen receptor degraders (SERDs) in cancer treatment. Pharmacol Ther 2018;186:1–24.
7. Miller WR, Bartlett J, Brodie AM, Brueggemeier RW, di Salle E, Lonning PE, et al. Aromatase inhibitors: are there differences between steroidal and nonsteroidal aromatase inhibitors and do they matter? Oncologist 2008;13:829–37.
8. Gennari A, Andre F, Barrios CH, Cortes J, de Azambuja E, DeMichele A, et al. ESMO Clinical Practice Guideline for the diagnosis, staging and treatment of patients with metastatic breast cancer. Ann Oncol 2021;32:1475–95.
9. Scott SC, Lee SS, Abraham J. Mechanisms of therapeutic CDK4/6 inhibition in breast cancer. Semin Oncol 2017;44:385–94.
10. Finn RS, Dering J, Conklin D, Kalous O, Cohen DJ, Desai AJ, et al. PD 0332991, a selective cyclin D kinase 4/6 inhibitor, preferentially inhibits proliferation of luminal estrogen receptorpositive human breast cancer cell lines in vitro. Breast Cancer Res 2009;11:R77.
11. Finn RS, Crown JP, Lang I, Boer K, Bondarenko IM, Kulyk SO, et al. The cyclin-dependent kinase 4/6 inhibitor palbociclib in combination with letrozole versus letrozole alone as firstline treatment of oestrogen receptor-positive, HER2-negative, advanced breast cancer (PALOMA-1/TRIO-18): a randomised phase 2 study. Lancet Oncol 2015;16:25–35.
12. Finn RS, Martin M, Rugo HS, Jones S, Im SA, Gelmon K, et al. Palbociclib and letrozole in advanced breast cancer. N Engl J Med 2016;375:1925–36.
13. Hortobagyi GN, Stemmer SM, Burris HA, Yap YS, Sonke GS, Paluch-Shimon S, et al. Ribociclib as first-line therapy for HR-positive, advanced breast cancer. N Engl J Med 2016;375:1738–48.
14. Hortobagyi GN, Stemmer SM, Burris HA, Yap YS, Sonke GS, Paluch-Shimon S, et al. Updated results from MONALEESA-2, a phase III trial of first-line ribociclib plus letrozole versus placebo plus letrozole in hormone receptor-positive, HER2- negative advanced breast cancer. Ann Oncol 2018;29:1541–7.
15. Im SA, Lu YS, Bardia A, Harbeck N, Colleoni M, Franke F, et al. Overall survival with ribociclib plus endocrine therapy in breast cancer. N Engl J Med 2019;381:307–16.
16. Lu YS, Im SA, Colleoni M, Franke F, Bardia A, Cardoso F, et al. Updated Overall survival of ribociclib plus endocrine therapy versus endocrine therapy alone in pre- and perimenopausal patients with HR+/HER2- advanced breast cancer in MONALEESA-7: a phase III randomized clinical trial. Clin Cancer Res 2022;28:851–9.
17. Johnston S, Martin M, Di Leo A, Im SA, Awada A, Forrester T, et al. MONARCH 3 final PFS: a randomized study of abemaciclib as initial therapy for advanced breast cancer. NPJ Breast Cancer 2019;5:5.
18. Goetz MP, Toi M, Huober J, Sohn J, Tredan O, Park IH, et al. LBA15 MONARCH 3: interim overall survival (OS) results of abemaciclib plus a nonsteroidal aromatase inhibitor (NSAI) in patients (pts) with HR+, HER2- advanced breast cancer (ABC). Ann Oncol 2022;33:S1384.
19. Slamon DJ, Neven P, Chia S, Jerusalem G, De Laurentiis M, Im S, et al. Ribociclib plus fulvestrant for postmenopausal women with hormone receptor-positive, human epidermal growth factor receptor 2-negative advanced breast cancer in the phase III randomized MONALEESA-3 trial: updated overall survival. Ann Oncol 2021;32:1015–24.
20. Turner NC, Ro J, Andre F, Loi S, Verma S, Iwata H, et al. Palbociclib in hormone-receptor-positive advanced breast cancer. N Engl J Med 2015;373:209–19.
21. Sledge GW Jr, Toi M, Neven P, Sohn J, Inoue K, Pivot X, et al. MONARCH 2: abemaciclib in combination with fulvestrant in women with HR+/HER2- advanced breast cancer who had progressed while receiving endocrine therapy. J Clin Oncol 2017;35:2875–84.
22. Tripathy D, Im SA, Colleoni M, Franke F, Bardia A, Harbeck N, et al. Ribociclib plus endocrine therapy for premenopausal women with hormone-receptor-positive, advanced breast cancer (MONALEESA-7): a randomised phase 3 trial. Lancet Oncol 2018;19:904–15.
23. Cristofanilli M, Turner NC, Bondarenko I, Ro J, Im SA, Masuda N, et al. Fulvestrant plus palbociclib versus fulvestrant plus placebo for treatment of hormone-receptor-positive, HER2-negative metastatic breast cancer that progressed on previous endocrine therapy (PALOMA-3): final analysis of the multicentre, double-blind, phase 3 randomised controlled trial. Lancet Oncol 2016;17:425–39.
24. Finn RS, Rugo HS, Dieras VC, Harbeck N, Im SA, Gelmon KA et al. Overall survival (OS) with first-line palbociclib plus letrozole (PAL+LET) versus placebo plus letrozole (PBO+LET) in women with estrogen receptor–positive/human epidermal growth factor receptor 2–negative advanced breast cancer (ER+/HER2− ABC): Analyses from PALOMA-2. J Clin Oncol 2022;40(17 Suppl):LBA1003.
25. Hortobagyi GN, Stemmer SM, Burris HA, Yap YS, Sonke GS, Hart L, et al. Overall survival with ribociclib plus letrozole in advanced breast cancer. N Engl J Med 2022;386:942–50.
26. Sledge GW Jr, Toi M, Neven P, Sohn J, Inoue K, Pivot X, et al. The effect of abemaciclib plus fulvestrant on overall survival in hormone receptor-positive, ERBB2-negative breast cancer that progressed on endocrine therapy-MONARCH 2: a randomized clinical trial. JAMA Oncol 2020;6:116–24.
27. Goetz MP, Toi M, Campone M, Sohn J, Paluch-Shimon S, Huober J, et al. MONARCH 3: abemaciclib as initial therapy for advanced breast cancer. J Clin Oncol 2017;35:3638–46.
28. Slamon DJ, Neven P, Chia S, Fasching PA, De Laurentiis M, Im SA, et al. Phase III randomized study of ribociclib and fulvestrant in hormone receptor-positive, human epidermal growth factor receptor 2-negative advanced breast cancer: MONALEESA-3. J Clin Oncol 2018;36:2465–72.
29. Gradishar WJ, Anderson BO, Abraham J, Aft R, Agnese D, Allison KH, et al. Breast cancer, version 3.2020, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw 2020;18:452–78.
30. Lu YS, Mahidin EI, Azim H, Eralp Y, Yap YS, Im SA, et al. Abstract GS1-10: primary results from the randomized phase II RIGHT choice trial of premenopausal patients with aggressive HR+/HER2− advanced breast cancer treated with ribociclib+endocrine therapy vs. physician’s choice combination chemotherapy. Cancer Res 2023;83(5 Suppl):GS1–10.
31. McAndrew NP, Finn RS. Clinical review on the management of hormone receptor-positive metastatic breast cancer. JCO Oncol Pract 2022;18:319–27.
32. Spring LM, Zangardi ML, Moy B, Bardia A. Clinical management of potential toxicities and drug interactions related to cyclin-dependent kinase 4/6 inhibitors in breast cancer: practical considerations and recommendations. Oncologist 2017;22:1039–48.
33. Thill M, Schmidt M. Management of adverse events during cyclin-dependent kinase 4/6 (CDK4/6) inhibitor-based treatment in breast cancer. Ther Adv Med Oncol 2018;10:1758835918793326.
34. Llombart-Cussac A, Perez-Garcia JM, Bellet M, Dalenc F, GilGil M, Ruiz-Borrego M, et al. Fulvestrant-palbociclib vs. letrozole-palbociclib as initial therapy for endocrine-sensitive, hormone receptor-positive, ERBB2-negative advanced breast cancer: a randomized clinical trial. JAMA Oncol 2021;7:1791–9.
35. Turner NC, Slamon DJ, Ro J, Bondarenko I, Im SA, Masuda N, et al. Overall survival with palbociclib and fulvestrant in advanced breast cancer. N Engl J Med 2018;379:1926–36.
36. Cardoso F, Costa A, Norton L, Senkus E, Aapro M, Andre F, et al. ESO-ESMO 2nd international consensus guidelines for advanced breast cancer (ABC2). Breast 2014;23:489–502.
37. Llombart-Cussac A, Sledge G, Toi M, Neven P, Sohn JH, Inoue K, et al. Abstract PD13-11: PD13-11 final overall survival analysis of Monarch 2 : a phase 3 trial of abemaciclib plus fulvestrant in patients with hormone receptor-positive, HER2-negative advanced breast cancer. Cancer Res 2023;83(5 Suppl):PD13–11.
38. Slamon DJ, Neven P, Chia S, Fasching PA, De Laurentiis M, Im SA, et al. Overall survival with ribociclib plus fulvestrant in advanced breast cancer. N Engl J Med 2020;382:514–24.
39. Dickler MN, Tolaney SM, Rugo HS, Cortes J, Dieras V, Patt D, et al. MONARCH 1, a phase II study of abemaciclib, a CDK4 and CDK6 inhibitor, as a single agent, in patients with refractory HR(+)/HER2(-) metastatic breast cancer. Clin Cancer Res 2017;23:5218–24.
40. Hamilton E, Cortes J, Ozyilkan O, Chen SC, Petrakova K, Manikhas A, et al. nextMONARCH: abemaciclib monotherapy or combined with tamoxifen for metastatic breast cancer. Clin Breast Cancer 2021;21:181–90.
41. Elliott MJ, Cescon DW. Development of novel agents for the treatment of early estrogen receptor positive breast cancer. Breast 2022;62(Suppl 1):S34–42.
42. Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumours. Nature 2012;490:61–70.
43. Andre F, Ciruelos E, Rubovszky G, Campone M, Loibl S, Rugo HS, et al. Alpelisib for PIK3CA-mutated, hormone receptorpositive advanced breast cancer. N Engl J Med 2019;380:1929–40.
44. Rugo HS, Lerebours F, Ciruelos E, Drullinsky P, Ruiz-Borrego M, Neven P, et al. Alpelisib plus fulvestrant in PIK3CAmutated, hormone receptor-positive advanced breast cancer after a CDK4/6 inhibitor (BYLieve): one cohort of a phase 2, multicentre, open-label, non-comparative study. Lancet Oncol 2021;22:489–98.
45. Jones RH, Casbard A, Carucci M, Cox C, Butler R, Alchami F, et al. Fulvestrant plus capivasertib versus placebo after relapse or progression on an aromatase inhibitor in metastatic, oestrogen receptor-positive breast cancer (FAKTION): a multicentre, randomised, controlled, phase 2 trial. Lancet Oncol 2020;21:345–57.
46. Howell SJ, Casbard A, Carucci M, Ingarfield K, Butler R, Morgan S, et al. Fulvestrant plus capivasertib versus placebo after relapse or progression on an aromatase inhibitor in metastatic, oestrogen receptor-positive, HER2-negative breast cancer (FAKTION): overall survival, updated progression-free survival, and expanded biomarker analysis from a randomised, phase 2 trial. Lancet Oncol 2022;23:851–64.
47. Turner NC, Oliveira M, Howell SJ, Dalenc F, Cortes J, Gomez Moreno HL, et al. Capivasertib in hormone receptor-positive advanced breast cancer. N Engl J Med 2023;388:2058–70.
48. Baselga J, Campone M, Piccart M, Burris HA 3rd, Rugo HS, Sahmoud T, et al. Everolimus in postmenopausal hormonereceptor-positive advanced breast cancer. N Engl J Med 2012;366:520–9.
49. Yardley DA, Noguchi S, Pritchard KI, Burris HA 3rd, Baselga J, Gnant M, et al. Everolimus plus exemestane in postmenopausal patients with HR(+) breast cancer: BOLERO-2 final progression-free survival analysis. Adv Ther 2013;30:870–84.
50. Piccart M, Hortobagyi GN, Campone M, Pritchard KI, Lebrun F, Ito Y, et al. Everolimus plus exemestane for hormonereceptor-positive, human epidermal growth factor receptor2-negative advanced breast cancer: overall survival results from BOLERO-2dagger. Ann Oncol 2014;25:2357–62.
51. Chandarlapaty S, Chen D, He W, Sung P, Samoila A, You D, et al. Prevalence of ESR1 mutations in cell-free DNA and outcomes in metastatic breast cancer: a secondary analysis of the BOLERO-2 clinical trial. JAMA Oncol 2016;2:1310–5.
52. Bidard FC, Kaklamani VG, Neven P, Streich G, Montero AJ, Forget F, et al. Elacestrant (oral selective estrogen receptor degrader) versus standard endocrine therapy for estrogen receptor-positive, human epidermal growth factor receptor 2-negative advanced breast cancer: results from the randomized phase III EMERALD trial. J Clin Oncol 2022;40:3246–56.
53. Corti C, De Angelis C, Bianchini G, Malorni L, Giuliano M, Hamilton E, et al. Novel endocrine therapies: what is next in estrogen receptor positive, HER2 negative breast cancer? Cancer Treat Rev 2023;117:102569.
54. Oliveira M, Pominchuck D, Nowecki Z, Hamilton E, Kulyaba Y, Andabekov T, et al. Abstract GS3-02: GS3-02 camizestrant, a next generation oral SERD vs. fulvestrant in post-menopausal women with advanced ER-positive HER2-negative breast cancer: results of the randomized, multi-dose phase 2 SERENA-2 trial. Cancer Res 2023;83(5 Suppl):GS3–02.
55. Im SA, Hamilton EP, Llombart Cussac A, Baird RD, Ettl J, Goetz MP, et al. SERENA-4: a phase 3 comparison of AZD9833 (camizestrant) plus palbociclib, versus anastrozole plus palbociclib, for patients with ER-positive, HER2-negative advanced breast cancer who have not previously received systemic treatment for advanced disease. J Clin Oncol 2021;39(15 Suppl):TPS1101.
56. Bidard FC, Kalinsky K, Cristofanilli M, Bianchini G, Chia SK, Janni W, et al. Abstract OT2-11-05: SERENA-6: a phase III study to assess the efficacy and safety of AZD9833 (camizestrant) compared with aromatase inhibitors when given in combination with palbociclib or abemaciclib in patients with HR+/HER2- metastatic breast cancer with detectable ESR1m who have not experienced disease progression on first-line therapy. Cancer Res 2022;82(4 Suppl):OT2–11.
57. Jimenez MM, Lim E, Gregor MC, Bardia A, Wu J, Zhang Q, et al. 211MO Giredestrant (GDC-9545) vs. physician choice of endocrine monotherapy (PCET) in patients (pts) with ER+, HER2– locally advanced/metastatic breast cancer (LA/mBC): primary analysis of the phase II, randomised, open-label acelERA BC study. Ann Oncol 2022;33(Suppl 7):S633–4.
58. Patel R, Klein P, Tiersten A, Sparano JA. An emerging generation of endocrine therapies in breast cancer: a clinical perspective. NPJ Breast Cancer 2023;9:20.
59. Ferraro E, Walsh EM, Tao JJ, Chandarlapaty S, Jhaveri K. Accelerating drug development in breast cancer: new frontiers for ER inhibition. Cancer Treat Rev 2022;109:102432.
60. Prakash R, Zhang Y, Feng W, Jasin M. Homologous recombination and human health: the roles of BRCA1, BRCA2, and associated proteins. Cold Spring Harb Perspect Biol 2015;7:a016600.
61. Turk AA, Wisinski KB. PARP inhibitors in breast cancer: bringing synthetic lethality to the bedside. Cancer 2018;124:2498–506.
62. Alsop K, Fereday S, Meldrum C, deFazio A, Emmanuel C, George J, et al. BRCA mutation frequency and patterns of treatment response in BRCA mutation-positive women with ovarian cancer: a report from the Australian Ovarian Cancer Study Group. J Clin Oncol 2012;30:2654–63.
63. Robson M, Im SA, Senkus E, Xu B, Domchek SM, Masuda N, et al. Olaparib for metastatic breast cancer in patients with a germline BRCA mutation. N Engl J Med 2017;377:523–33.
64. Robson ME, Tung N, Conte P, Im SA, Senkus E, Xu B, et al. OlympiAD final overall survival and tolerability results: olaparib versus chemotherapy treatment of physician’s choice in patients with a germline BRCA mutation and HER2-negative metastatic breast cancer. Ann Oncol 2019;30:558–66.
65. Litton JK, Rugo HS, Ettl J, Hurvitz SA, Goncalves A, Lee KH, et al. Talazoparib in patients with advanced breast cancer and a germline BRCA mutation. N Engl J Med 2018;379:753–63.
66. Zhang H, Katerji H, Turner BM, Hicks DG. HER2-low breast cancers. Am J Clin Pathol 2022;157:328–36.
67. Ogitani Y, Hagihara K, Oitate M, Naito H, Agatsuma T. Bystander killing effect of DS-8201a, a novel anti-human epidermal growth factor receptor 2 antibody-drug conjugate, in tumors with human epidermal growth factor receptor 2 heterogeneity. Cancer Sci 2016;107:1039–46.
68. Modi S, Park H, Murthy RK, Iwata H, Tamura K, Tsurutani J, et al. Antitumor activity and safety of trastuzumab deruxtecan in patients with HER2-low-expressing advanced breast cancer: results from a phase Ib study. J Clin Oncol 2020;38:1887–96.
69. Modi S, Jacot W, Yamashita T, Sohn J, Vidal M, Tokunaga E, et al. Trastuzumab deruxtecan in previously treated HER2-low advanced breast cancer. N Engl J Med 2022;387:9–20.
70. Liu X, Deng J, Yuan Y, Chen W, Sun W, Wang Y, et al. Advances in Trop2-targeted therapy: novel agents and opportunities beyond breast cancer. Pharmacol Ther 2022;239:108296.
71. Kalinsky K, Diamond JR, Vahdat LT, Tolaney SM, Juric D, O’Shaughnessy J, et al. Sacituzumab govitecan in previously treated hormone receptor-positive/HER2-negative metastatic breast cancer: final results from a phase I/II, single-arm, basket trial. Ann Oncol 2020;31:1709–18.
72. Rugo HS, Bardia A, Marme F, Cortes J, Schmid P, Loirat D, et al. Primary results from TROPiCS-02: a randomized phase 3 study of sacituzumab govitecan (SG) versus treatment of physician’s choice (TPC) in patients (Pts) with hormone receptor– positive/HER2-negative (HR+/HER2-) advanced breast cancer. J Clin Oncol 2022;40(17 Suppl):LBA1001.
73. Rugo HS, Bardia A, Marme F, Cortes J, Schmid P, Loirat D, et al. Overall survival with sacituzumab govitecan in hormone receptor-positive and human epidermal growth factor receptor 2-negative metastatic breast cancer (TROPiCS-02): a randomised, open-label, multicentre, phase 3 trial. Lancet 2023 Aug 23 [Epub]. https://doi.org/10.1016/S0140-6736(23)01245-X.
74. Rugo HS, Delord JP, Im SA, Ott PA, Piha-Paul SA, Bedard PL, et al. Safety and antitumor activity of pembrolizumab in patients with estrogen receptor-positive/human epidermal growth factor receptor 2-negative advanced breast cancer. Clin Cancer Res 2018;24:2804–11.
75. Chaudhuri S, Thomas S, Munster P. Immunotherapy in breast cancer: a clinician’s perspective. J Natl Cancer Cent 2021;1:47–57.
76. Tolaney SM, Barroso-Sousa R, Keenan T, Li T, Trippa L, VazLuis I, et al. Effect of eribulin with or without pembrolizumab on progression-free survival for patients with hormone receptor-positive, ERBB2-negative metastatic breast cancer: a randomized clinical trial. JAMA Oncol 2020;6:1598–605.
77. Shah AN, Flaum L, Helenowski I, Santa-Maria CA, Jain S, Rademaker A, et al. Phase II study of pembrolizumab and capecitabine for triple negative and hormone receptor-positive, HER2-negative endocrine-refractory metastatic breast cancer. J Immunother Cancer 2020;8e000173.
78. Perez-Garcia JM, Llombart-Cussac A, Cortes MG, Curigliano G, Lopez-Miranda E, Alonso JL, et al. Pembrolizumab plus eribulin in hormone-receptor-positive, HER2-negative, locally recurrent or metastatic breast cancer (KELLY): an openlabel, multicentre, single-arm, phase Ⅱ trial. Eur J Cancer 2021;148:382–94.
79. Kim SH, Suh KJ, Im SA, Lee KH, Kim MH, Sohn J, et al. A phase IB/II study of nivolumab in combination with eribulin in HER2-negative metastatic breast cancer (KCSG BR18-16). J Clin Oncol 2022;40(16 Suppl):1098.
80. Rugo HS, Sohn J, Jerez Gilarranz Y, Gonzalez-Cortijo L, Sonnenblick A, Sabanathan D, et al. KEYNOTE-B49: a phase 3, randomized, double-blind, placebo-controlled study of pembrolizumab plus chemotherapy in patients with HR+/HER2- locally recurrent inoperable or metastatic breast cancer. J Clin Oncol 2022;40(16 Suppl):TPS1118.
81. Yuan Y, Lee JS, Yost SE, Frankel PH, Ruel C, Egelston CA, et al. Phase I/II trial of palbociclib, pembrolizumab and letrozole in patients with hormone receptor-positive metastatic breast cancer. Eur J Cancer 2021;154:11–20.

Article information Continued

Fig. 1.

Treatment of advanced/metastatic hormone receptor (HR)–positive, human epidermal growth factor protein 2 (HER2)–negative breast cancer. ESR1, estrogen receptor 1; ET, endocrine therapy; OFS, ovarian function suppression; PD, progressive disease; SERD, selective estrogen receptor degrader; T-Dxd, trastuzumab-deruxtecan; ULN, upper limit of normal.

Table 1.

Landmark studies in dual CDK4/6 inhibitor and endocrine therapy for advanced HR+breast cancer

Trial Phase Treatment arms No. of patients Menopausal status First or second-line setting (if second-line, indications are included) Median PFS (95% CI, mo) Median OS (95% CI, mo)
Trials with AIs as the backbone endocrine therapy agent
PALOMA-1 II Palbociclib+letrozole vs. letrozole 165 Postmenopausal First-line 20.2 (13.8-27.5) vs. 10.2 (5.7-12.6) 37.5 (31.4-47.8) vs. 34.5 (27.4-42.6)
HR 0.488 (0.319-0.748, p=0.0004) [11] HR 0.897 (0.623-1.294, p=0.281) [11]
PALOMA-2 III Palbociclib+letrozole vs. placebo+letrozole 666 Postmenopausal First-line 24.8 (22.1-not estimable) vs. 14.5 (12.9-17.1) 53.9 (49.8-60.8) vs. 51.2 (43.7-58.9)
HR 0.58 (95% CI, 0.46-9.72, p < 0.001) [12] HR 0.956 (0.777-1.177, p=0.03378) [24]
MONALEESA-2 III Ribociclib+letrozole vs. placebo+letrozole 668 Postmenopausal First-line 25.3 (23.0-30.3) vs. 16.0 (13.4-18.2) 63.9 (52.4-71.0) vs. 51.4 (47.2-59.7)
HR 0.568 (0.457-0.704, p=9.63×10-8) [13,14] HR 0.76, (0.63-0.93, p=0.008) [25]
MONARCH-3 III Abemaciclib+NSAI vs. placebo+NSAI 493 Postmenopausal First-line 28.18 vs. 14.76; 95% CIs, not reported 67.1 vs. 65.1; 95% CIs, not reported
HR 0.540 (0.418-0.698, p=0.000002) [17] HR 0.754 (0.584-0.974, p=0.0301) [18]
MONALEESA-7 III Ribociclib+tamoxifen or NSAI+goserelin vs. placebo+tamoxifen or NSAI+goserelin 672 Premenopausal First and second-line (for patients who received up to one previous line of chemotherapy for advanced disease) Overall: 23.8 (19.2-not reached) vs. 13.0 (11.0-16.4) Overall: 58.7 vs. 48.0; 95% CIs, not reported
HR 0.55 (0.44-0.69, p < 0.0001) [22] HR 0.76 (0.61-0.96 p=not reported) [15,16]
Chemotherapy subgroup: 46.8% vs. 61.7% event rate; HR 0.55 (0.31-0.95) Chemotherapy subgroup: 27.7% vs. 40.4% event rate; HR 0.67 (0.33-1.35)
Trials with fulvestrant as the backbone endocrine therapy agent
PALOMA-3 III Palbociclib+fulvestrant vs. placebo+fulvestrant 521 Any Second-line (progression during or ≤ 1 month after for endocrine therapy metastasis, progression during or ≤ 12 months after completion or discontinuation of adjuvant therapy) 9.5 (9.2-11.0) vs. 4.6 (3.5-5.6) 34.9 (28.8-40.0) vs. 28.0 (23.6-34.6)
Note: pre or perimenopausal patients received goserelin HR 0.46 (0.36-0.59, p < 0.0001) [20,23] HR 0.81 (0.64-1.03, p=0.09) [20,23]
MONARCH-2 III Abemaciclib+fulvestrant vs. placebo+fulvestrant 669 Any Second-line (progression on neoadjuvant or adjuvant endocrine therapy, ≤ 12 months after adjuvant endocrine therapy, or during endocrine therapy for advanced disease) 16.4 vs. 9.3; 95% CIs, not reported 46.7 vs. 37.3; 95% CIs, not reported
HR 0.553 (0.449-0.681, p < 0.001) [21] HR 0.757 (0.606-0.945, p=0.01) [26]
MONALEESA-3 III Ribociclib+fulvestrant vs. placebo+fulvestrant 484 Postmenopausal First- and second-line (for patients who progressed after one line of endocrine therapy for advanced disease without prior (neo)adjuvant treatment for early disease) 20.5 (18.5-23.5) vs. 12.8 (10.9-16.3) Not reached vs. 51.8
HR 0.593 (0.480-0.732, p < 0.001) [19] HR 0.64 (0.46-0.88) [19]
Endocrine therapy subgroup: 56% vs. 77% event rate; HR 0.565 (0.428-0.744) Endocrine therapy subgroup: 61.6% vs. 70.3% event rate; HR 0.74 (0.56-0.98)

CDK4/6, cyclin-dependent kinase 4/6; CI, confidence interval; HR, hazard ratio; NSAI, nonsteroidal aromatase inhibitor; OS, overall survival; PFS, progression-free survival.