Next Article in Journal
The Deubiquitinating Enzyme USP48 Interacts with the Retinal Degeneration-Associated Proteins UNC119a and ARL3
Next Article in Special Issue
Genetic Heterogeneity, Tumor Microenvironment and Immunotherapy in Triple-Negative Breast Cancer
Previous Article in Journal
Reactive Centre Loop Mutagenesis of SerpinB3 to Target TMPRSS2 and Furin: Inhibition of SARS-CoV-2 Cell Entry and Replication
Previous Article in Special Issue
LC-PDA-MS and GC-MS Analysis of Scorzonera hispanica Seeds and Their Effects on Human Breast Cancer Cell Lines
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advances in the Management of Central Nervous System Metastases from Breast Cancer

1
Department of Internal Medicine, St Elizabeth’s Medical Center, 736 Cambridge St., Boston, MA 02135, USA
2
Department of Medicine, Tufts University School of Medicine, Boston, MA 02111, USA
3
Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave., Boston, MA 02215, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(20), 12525; https://doi.org/10.3390/ijms232012525
Submission received: 21 September 2022 / Revised: 17 October 2022 / Accepted: 17 October 2022 / Published: 19 October 2022
(This article belongs to the Special Issue Targeting Breast Cancer: Strategies and Hopes)

Abstract

:
Central nervous system (CNS) metastases are common in breast cancer (BC) patients and are particularly relevant as new treatments for BC are prolonging survival. Here, we review advances in the treatment of CNS metastases from BC, including radiotherapy, systemic therapies, and the evolving role of immunotherapy. The use of radiotherapy and chemotherapy is the cornerstone of treatment for CNS metastases. However, new targeted therapies have recently been developed, including anti-HER2 agents and antibody–drug conjugates that have presented promising results for the treatment of these patients.

1. Introduction

Breast cancer (BC) is the second most common cancer overall and the most common among women, representing one of the leading causes of mortality among women [1]. Around 30% of patients with BC will develop brain metastases (BM) during the course of their disease, which can have a devastating effect on prognosis, functional status, and quality of life [2]. The incidence of BM seems to have increased in recent years, most likely due to the prolonged survival of patients, the development of more efficient treatments, and the availability of better imaging techniques that lead to the increased detection of this complication [3].
The risk of CNS metastases is higher in BC patients that are hormone receptor (HR)-negative or human epidermal growth factor receptor 2 (HER2)-positive or have a high tumor grade [4]. According to this, BC subtypes have different tendencies to metastasize to the brain. Studies have shown that 25–46% of triple-negative breast cancer (TNBC), 11–48% of HER-2 positive, 8–15% of luminal A, and 11% of luminal B BC patients can develop CNS metastases [5,6].
The median overall survival (MOS) after the development of brain metastases has been described as 9.3 months for the luminal subtype, 16.5 months for the luminal HER-2 subtype, 11.5 months HER2 subtype, and 4.9 months for the TN subtype [7].
Different indexes have been created to assess the prognostic factors of patients with breast cancer brain metastases (BCBM). The recursive partitioning assessment (RPA) divides patients into three groups, class I, II, and III, with median survival (MS) times of 7.7 months, 4.5 months, and 2.3 months, respectively [8]. The graded prognostic assessment (GPA) divides patients into four groups based on a score obtained from clinical and biological characteristics: from 0 to 1, with an MS of 2.6 months; from 1.5 to 2.5 with an MS of 3.8 months; 3 with an MS of 6.9 months; and from 3.5 to 4 with an MS of 11 months [9]. More recently, a breast-specific GPA was developed, which provided a more accurate description of survival for these patients. The MS values for GPA 0–1, 1.5–2, 2.5–3, and 3.5–4 were 6, 13, 24, and 36 months, respectively [10].
It has been shown that patients with HER2-positive BC have a significantly higher incidence of central nervous system (CNS) metastases after treatment with trastuzumab, probably secondary to improvements in systemic disease control and longer MOS associated with this pharmacological therapy [11]. The registHER study, a prospective observational study of over 1000 HER2-positive metastatic BC patients, showed that the MOS after the diagnosis of CNS metastases was improved from 3.8 to 17.5 months with the administration of trastuzumab [12].
One of the first treatments for BM was whole-brain radiation therapy (WBRT). Other important local therapy options available today include stereotactic radiosurgery (SRS) and neurosurgical resection [13]. Surgical resection followed by radiotherapy (RT) could be curative for a small, solitary BM [14]. However, the current treatment options for patients with extracranial disease and/or multiple BM remain mainly palliative [6]. Most recently, targeted systemic therapies and immunotherapy appeared in the multidisciplinary management of BM, leading to an improvement in intracranial control, survival, and neurocognitive preservation among these patients [13].
The blood–brain barrier (BBB), a neurovascular unit composed of endothelial cells, astrocytes, and pericytes [15], makes the treatment of BM challenging, as it forms a selective barrier between the CNS and the systemic circulation [16]. The BBB is disrupted during tumor progression and forms the blood–tumor barrier (BTB), which is more permeable than the BBB, allowing multiple drugs into the CNS to target the tumor and healthy brain parenchyma [17]. The permeability of the BTB varies depending on the subtype of cancer [18]. For example, BM from TNBC or basal-type BC may often disrupt the BBB, whereas BM from HER2-positive BC tend to preserve the BBB [19].
Although there is no doubt the BTB is more permeable than the BBB, it still significantly restricts the delivery of anticancer drugs and obstructs the systemic chemotherapeutics of brain tumors [20]. RT, besides providing cytotoxicity, can cause disruption of the BBB, resulting in increased permeability into the surrounding brain parenchyma [21]. A better understanding of both the BBB and BTB is needed to develop treatments with higher penetration to the CNS or increased manipulation of the CNS barrier.
In this review, we summarize local and systemic treatments for BCBM. First, we discuss in detail the existing data supporting the different treatment options, including surgical resection, various types of RT, chemotherapy, and targeted therapies. Then, we put all the data into context in a clinical algorithm for the recommended management of these patients. This may provide a better understanding of this topic and encourage the development of new strategies for the management of BCBM.

2. Local Treatment for BM

Local treatment for BM includes surgery, WBRT, SRS, fractionated stereotactic radiotherapy (fSRT), or a combination of these [6,22]. The decision of choosing one over another may be based on the estimated prognosis and the goals of the treatment [23]. Surgical resection is the standard treatment for a single BM, especially when it is large in size and causes a mass effect or obstructive hydrocephalus [24]. In patients with limited BM (defined as one to four BM), surgical resection and SRS are considered acceptable treatments as well. WBRT remains beneficial in certain situations and is often used in patients with widely disseminated BM [25].
The use of surgery is most often reserved for patients with a good performance status and good extracranial disease control (or absence of extracranial disease) [14]. However, surgery alone is considered inadequate for local control when compared to surgery plus RT [26] Surgery followed by RT improves MOS and symptom control compared with RT alone [27]. Studies have shown that, in patients with a single BM, WBRT after surgical resection reduces the rate of recurrence at the initial metastatic site and other brain sites [26].
The use of WBRT alone is indicated only in patients with more than ten BM for whom local treatment is not appropriate and in patients with new lesions on which additional SRS cannot be performed [28]. Studies have shown improvements in symptoms in 64–83% of patients after treatment with WBRT alone [29,30,31] and have also shown an increase in MOS from 1 month with no treatment to 3.7 months after WBRT [32]. However, this treatment is associated with toxicities, including dermatitis, alopecia, nausea, cerebral edema, and even cognitive deterioration [25], making the use of other techniques preferable over WBRT.
Patients with limited intracranial disease can be offered SRS, which delivers large doses of radiation to a well-defined area [33]. Recent studies affirmed that, while WBRT reduces the risk of local and regional intracranial disease progression relative to SRS alone, these benefits come at the cost of increased neurotoxicity without improved survival [34]. Kayama et al. evaluated whether salvage SRS alone within 21 days of surgery is as effective as postoperative WBRT on the OS of patients with 1–4 BM. The authors concluded that salvage SRS can be a standard therapy for patients with this number of BM, as it was observed to be noninferior to WBRT, with an MOS of 15.6 months in both arms (hazard ratio: 1.05; p for noninferiority = 0.027) [35].
Andrews et al. compared survival between WBRT alone and WBRT followed by SRS in patients with 1–3 BM and discovered that MOS did not differ between these two groups, but WBRT plus SRS resulted in better survival for patients with a single unresectable BM when compared to WBRT alone (MOS: 6.5 vs. 4.9 months, respectively; p = 0.039) [36].
Other studies have investigated the difference between SRS alone and WBRT plus SRS and showed that patients with 1-4 BM treated with the latter option did not have a longer survival (8 vs. 7.5 months; p = 0.42) [37]. This was also reported in patients with 1–3 BM by Brown et al. (p = 0.92) [34], who showed that using SRS alone could be a better treatment strategy for patients with less than five BM.
A multi-institutional trial of WBRT vs. SRS for patients with 1–4 BM after the resection of 1 metastasis showed that WBRT was associated with improved local control (80.6% vs. 60.5%; p = <0.001) and intracranial control (78.6% vs. 54.7%, p < 0.001). However, it did not show any differences in OS, and cognitive deterioration was higher at 6 months in the WBRT group [38].
In patients with five or more BM, it is still unclear whether WBRT or SRS should be used. Li et al. compared SRS with WBRT for patients with 4–15 BM, and they described no difference in MOS between the two groups (10.4 vs. 8.4 months; p = 0.45); however, in the same study, SRS reduced the risk of cognitive deterioration; suggesting that there may be a benefit in avoiding WBRT in this setting [39].
As mentioned before, fSRT has also been evaluated for BCBM. fSRT is less costly and more comfortable for patients [40], and retrospective reviews comparing fSRT to SRS have shown no difference in the local control of BCBM [41]. Meanwhile, emerging studies suggest improved local control in fSRT when compared to SRS for BM [42,43]. However, there is limited information on fSRT when compared to WBRT. The use of fSRT in 1–10 BM is currently being evaluated in the NCT04061408 study [44].
Hippocampal-avoidance WBRT (HA-WBRT) has been shown to be effective in protecting against cognitive decline when compared to standard WBRT [45]. This technique is considered safe, as BCBM has a low risk of metastases in the hippocampal region [46].
Table 1 summarizes the major clinical trials for BCBM treated with RT.

3. Systemic Therapies for BCBM

Plasma proteins such as immunoglobulin G (IgG) molecules generally do not cross the brain capillary endothelial wall, which forms the BBB under normal conditions. However, certain monoclonal antibodies in the circulation cross the BBB by a process of receptor-mediated transcytosis [47]. In this case, the monoclonal antibody is directed against a specific receptor located on the luminal membrane of the brain capillary endothelium, and the monoclonal antibody binds to exofacial epitopes on the BBB receptors. The reverse transcytosis of IgG molecules across the BBB is most likely mediated by an Fc receptor situated on the abluminal membrane of the brain capillary endothelium, and rat models have demonstrated a rapid IgG efflux with a half-life of 48 min [48]. In the following paragraphs, we will mention different drugs that have been evaluated for BCBM. However, their passage through the BBB/BTB is still under study.

3.1. Human Epidermal Growth Factor Receptor 2 (HER2)

3.1.1. Trastuzumab

Trastuzumab is a monoclonal antibody directed against the extracellular domain of HER2. However, it has a large molecular weight that makes it difficult to cross the BBB, and regular intravenous (IV) administration may not be effective for BCBM [49].
Under impaired BBB conditions such as meningeal carcinomatosis or RT, trastuzumab levels in cerebrospinal fluid (CSF) increase; this evidence supports the concept of continuing trastuzumab therapy in patients with BM treated by RT. Monitoring trastuzumab in the serum and CSF may enable individualized therapies in BCBM [50].
Nonetheless, Park et al. demonstrated that patients receiving trastuzumab for BCBM had a significant longer time to death (14.5 vs. 4.0 months; p = 0.0005) [51], and other authors discovered that this treatment appeared to prolong the MOS by controlling extracranial disease [52,53]. The registHER study also revealed that treatment with trastuzumab after the diagnosis of BCBM significantly decreased the risk of death [12]. Dawood et al. showed that patients with HER2-positive disease treated with trastuzumab had a longer median time to CNS metastasis compared with similar patients who never received trastuzumab (13.1 vs. 2.1 months; p = 0.0008) [54].
Several case reports have described a benefit of intrathecal trastuzumab administration to treat carcinomatous meningitis in patients with HER2-overexpressing metastatic BC [55,56]. Bousquet et al. described the effect of intrathecal trastuzumab in a patient with HER-2 BCBM, and after maintaining an efficacious concentration of this drug in the CSF, they were able to achieve the stabilization of brain and epidural metastases previously resistant to radiation and chemotherapy [57]. More recently, Kumthekar et al. evaluated intrathecal trastuzumab in a multicenter phase I/II study on patients with HER2-positive leptomeningeal disease (LMD). Partial responses were observed in 19.2% of the patients, and stable disease was seen in 50% of the participants. This study suggests the potential for improved outcomes in HER2-positive LMD [58]. However, further analysis needs to be conducted for the evaluation of intrathecal trastuzumab.
Trastuzumab was the first established therapy targeted against HER2-positive BCBM; unfortunately, despite initial success, patients developed disease progression [18].

3.1.2. Trastuzumab Emtansine (T-DM1)

Trastuzumab emtansine (T-DM1) is an antibody–drug conjugate composed of trastuzumab linked to the cytotoxic agent DM1 (a maytansine derivative), and it has been evaluated in multiple studies of patients with HER-2 positive BCBM, with reports of benefits in the median progression-free survival (PFS) [59,60].
EMILIA, a phase III trial, compared the efficacy of T-DM1 alone vs. lapatinib + capecitabine in HER2-positive advanced BC previously treated with trastuzumab and a taxane, and it showed that the T-DM1 group had improvements in PFS and OS (p < 0.001). In patients with treated asymptomatic CNS metastases at baseline, OS was improved in the T-DM1 group when compared to the lapatinib + capecitabine group (MOS: 26.8 vs. 12.9 months; p = 0.008) [61].
In 2020, Montemurro et al. published data from a subgroup of 398 patients with BM from the KAMILLA trial, a single-arm phase IIIb study of T-DM1 in patients with HER2-positive locally advanced/metastatic BC. The study confirmed the efficacy and safety of this drug, with a BM response rate of 21.4%, a median PFS of 5.5 months, and an MOS of 18.9 months [62].

3.1.3. Trastuzumab Deruxtecan (T-DXd)

Trastuzumab Deruxtecan (T-DXd) is an antibody–drug conjugate of trastuzumab and an exatecan derivative (topoisomerase 1 inhibitor). The DESTINY-Breast01 trial investigated the efficacy of this drug in HER2-positive metastatic breast cancer (MBC) previously treated with T-DM1 and showed a response rate of 60.9% and a median PFS of 16.4 months [63].
DESTINY-Breast03, a phase III trial that compared T-DXd vs. T-DM1 in HER2 + MBC patients, showed that the estimated 12-month OS was 75.8% for the T-DXd group vs. 34.1% for the T-DM1 group (p = 7.8 × 10−22) [64]. Approximately 15% of the patients in each arm had the presence of brain metastases at baseline; among these patients, the median PFS was 15 months for T-DXd vs. 3 months for T-DM1. In addition, intracranial responses were also higher in the T-DXd arm. Complete responses were observed in 27.8% of T-DXd patients and 2.8% of T-DM1 patients. Partial responses were observed in 36.1% and 30.6% of T-DXd and T-DM1 patients, respectively [65].
DESTINY-Breast 04, a phase III trial that included patients with BM, compared T-DXd vs. the treatment of physician’s choice (TPC) in previously treated HER2-low advanced breast cancer and showed that patients treated with T-DXd presented significantly longer PFS and OS than the TPC group (23.9 vs. 17.5 months; p = 0.003) [66].

3.1.4. Pertuzumab

Pertuzumab is a humanized monoclonal antibody that inhibits the dimerization of HER2 with other HER receptors and can be considered for the treatment of CNS metastases [67]. The phase III trial CLEOPATRA compared pertuzumab, trastuzumab, and docetaxel with placebo, trastuzumab, and docetaxel; the results from this trial showed that the median time to progression in the CNS was 11.9 in the placebo group and 15.0 months in the pertuzumab group (p = 0.0049); however, the incidence of CNS metastases as the first site of disease progression was similar in both arms (13.7 % vs. 12.6%) [68]. The final results from this study stated that the MOS was improved in the group that received pertuzumab when compared to the placebo group (57.1 vs. 40.8 months, hazard ratio: 0.69) [69].
In the phase II PATRICIA study, patients with HER2-positive MBC with CNS metastases received pertuzumab plus high-dose trastuzumab, and although the overall CNS response rate was modest (11%), 68% of patients experienced a clinical benefit at 4 months [70].

3.1.5. Lapatinib

Lapatinib is a dual tyrosine kinase inhibitor of the EGFR and HER2 that is able to cross the BBB due to its low molecular weight [17]. Studies have shown that single-agent lapatinib is active in patients with HER2-positive BCBM, with a modest response between and 2.6% and 6% [71,72]. In order to increase the response rate, lapatinib has been combined with capecitabine. Studies have shown that this combination is superior to capecitabine alone in HER2-positive MBC [73] as well as in BCMB, with an objective response rate (ORR) of 20%; in addition, volumetric reduction was observed in the CNS lesions of the patients treated with this combined regimen [72].
LANDSCAPE, a single-arm phase II trial, investigated the efficacy of combining lapatinib and capecitabine in patients with an initial recurrence of BM not previously treated with WBRT. This study presented a CNS response rate of 65.9% [74].
CEREBEL, a phase III randomized study designed to investigate the incidence of the CNS being the first progression in patients with HER2-positive MBC, compared lapatinib + capecitabine vs. trastuzumab + capecitabine, and it showed that trastuzumab + capecitabine had a longer PFS (hazard ratio: 1.30; p = 0.021) [75].
A pooled analysis of 12 studies about the use of lapatinib + capecitabine or lapatinib alone in HER2-positive BCBM demonstrated an ORR of 21.4%, a PFS of 4.1 months, and an OS of 11.2 months [76].
The use of lapatinib combined with trastuzumab has also been studied. Patients with HER2-positive MBC or recurrent breast cancer (RBC) with BM treated with this combination therapy had a significantly longer survival than those treated with trastuzumab alone, lapatinib alone, or no HER2-targeted therapy (p < 0.001) [77]. Other studies have shown that patients treated with lapatinib + taxane had a significantly shorter PFS (hazard ratio: 1.48; p < 0.001) than those treated with trastuzumab + taxane [78].
Lin et al. have also studied the combination of lapatinib and WBRT, which has shown a higher ORR (79%) when compared to the historical results of WBRT alone [79].

3.1.6. Neratinib

Neratinib is an irreversible inhibitor of EGFR, HER1, HER2 and HER4 and has demonstrated activity both as a single agent and in combination with paclitaxel [80].
NEfERT-T, a phase II trial, compared the use of neratinib + paclitaxel vs. trastuzumab + paclitaxel in HER2-positive MBC and showed that the incidence of CNS metastases was lower (8.3% vs. 17.3%; p = 0.002), and the time to CNS metastases was delayed in the group receiving neratinib + paclitaxel; however, both groups had similar median PFS values [81].
The TBCRC 022 trial evaluated neratinib as a single agent in HER2-positive BCBM and presented a low CNS objective response of 8%. However, the trial was expanded and showed that neratinib + capecitabine resulted in a CNS response of 49% in the lapatinib-naïve group vs. 33% in the cohort of lapatinib pretreated patients, reaffirming the synergy observed between lapatinib and capecitabine [82].
The NALA trial compared neratinib + capecitabine vs. lapatinib + capecitabine in HER2-positive MBC and demonstrated a longer PFS in the neratinib group (8.8 vs. 6.6 months; p = 0.003). In this study, the cumulative incidences of intervention for CNS disease were 22.8% in the neratinib group and 29.2% in the lapatinib group (p = 0.043) [83].

3.1.7. Tucatinib

Tucatinib is a specific inhibitor of HER2 and a substrate of p glycoprotein (p-gp), which would be expected to limit distribution to CNS. However, a reduced number of efflux transporters, acid interstitial pH, and leaky junctions enhance tucatinib permeability into the tumor [84].
Different studies have shown an increase in CNS response (12%) and better PFS when combined with trastuzumab, and when combined with T-DM1, the results have presented a median PFS of 6.7 months and a 36% CNS response [85,86].
The combination of tucatinib + trastuzumab + capecitabine has been explored in various studies, including a phase I trial that resulted in a CNS response of 42% and a median PFS of 6.7 months [87]. HER2CLIMB, a phase II trial that studied the efficacy if this triple combination on the intracranial response and survival in HER2-positive BCBM, showed a significant improvement in PFS, with a 1-year rate of 24.9% in the tucatinib group vs. 0 in the placebo group (p < 0.001) and median PFS values of 7.6 vs. 5.4 months, respectively [88]. A subgroup of patients from this trial was analyzed to determine the intracranial response (47.3% vs. 20%), CNS PFS (9.9 vs. 4.2 months), MOS (18.1 vs. 12.0 months), and a reduced risk of death (42%) in the tucatinib arm [89].
Data from HER2CLIMB also demonstrated a 62% reduction in disease progression or death and a longer PFS (7.6 vs. 4.1 months) in the patients receiving tucatinib vs. placebo [90,91].
The final OS analysis from HER2CLIMB showed that the MOS was 24.7 months for the tucatinib combination vs. 19.2 months for the placebo group (p = 0.004) [92].
Currently, HER2CLIMB-02 is evaluating the use of tucatinib + T-DM1 in patients with HER2-positive MBC.

3.1.8. Pyrotinib

Pyrotinib, an irreversible pan-HER receptor inhibitor, has shown higher efficacy when compared to lapatinib in HER2-positive MBC [93]. However, its benefit in BCBM is still unknown. Nonetheless, there is an ongoing phase II trial of pyrotinib + vinorelbine in HER2-positive BCBM patients (NCT03933982).
PERMEATE, a phase II trial that studied pyrotinib + capecitabine for patients with HER2-positive BCBM, demonstrated a intracranial objective response rates of 74.6% in radiotherapy-naïve patients and 42.1% in those with progressive disease after RT [94].

3.2. Vascular Endothelial Growth Factor (VEGF)

Bevacizumab

Bevacizumab is a recombinant humanized monoclonal antibody against VEGF and has been studied in MBC. A phase III trial compared paclitaxel + bevacizumab vs. paclitaxel alone in MBC and observed a longer PFS in the combined treatment group; however, there was no improvement in OS, and unfortunately this study did not include CNS metastases [95].
A phase II trial evaluated the efficacy of carboplatin and bevacizumab in BCBM, and presented an ORR of 63%, a median PFS of 5.62 months, and an MOS of 14.10 months [96]. Another phase II study in patients with BCBM demonstrated a CNS ORR of 77% with bevacizumab followed by etoposide and cisplatin [97].

3.3. Phosphatidylinositol 3-kinase (PI3K)/Mammalian Target of Rapamycin (mTOR)

Approximately 40% of HR-positive, HER2-negative BCs display PIK3CA mutations.
Although certain studies have proven the efficacy of alpelisib in the treatment of MBC [98,99], these have excluded patients with active or untreated BCBM. Four cases of patients with BCBM have been described in the literature about reduced size or stable disease with the use of alpelisib in combination with hormone therapy [100].
Buparlisib, a pan-class I PI3K inhibitor, has been shown to penetrate the BBB [101] and presented a better PFS (hazard ratio: 0.67; p = 0.0003) in the phase III BELLE trial when comparing buparlisib + fulvestrant vs. placebo for MBC patients [102].
Everolimus was also analyzed by Hurvitz et al. in a phase Ib/II single-arm trial investigating a triple therapy of lapatinib + everolimus + capecitabine in patients with HER2-positive BCBM. The results showed a 27% CNS ORR at 12 weeks and 6.2 months of PFS [103]. Everolimus was also studied in combination with trastuzumab + vinorelbine in BCBM and resulted in an intracranial response rate of 4% and a CNS clinical benefit of 27% at 6 months [104]. Paxalisib, a dual PI3L/mTOR inhibitor, is being studied to evaluate its clinical efficacy on HER2-positive BCBM [18].

3.4. Cyclin-Dependent Kinase 4/6

CDK 4–6 inhibitors block the accelerated cell cycle transition from G1 to S phase and therefore suppress cell cycle dysregulation [67].
Abemaciclib, a CDK 4/6 inhibitor, has shown improvement in ORR and PFS in patients with HR-positive BC when used in combination with either fulvestrant or aromatase inhibitors [105]. A phase II trial of abemaciclib that included patients with leptomeningeal metastases as well as BM treated with surgical resection showed that abemaciclib achieved therapeutic concentrations in BM tissue, confirming that this drug crosses the BBB [106].
Another phase II trial of abemaciclib in HR-positive/HER2-negative BCBM revealed a 38% intracranial response and a intracranial benefit rate of 25% [107].
Ribociclib combined with letrozole, presented positive results in the CompLEEment-1 study, a phase IIIb trial including HR-positive/HER2-negative MBC patients, with an ORR of 20.5% [108], and patients from this study with CNS metastases presented favorable results, with an ORR of 41.2% [109].
The MONALEESA trials studied the use of ribociclib in combination with endocrine therapy (ET) in patients with MBC. MONALEESA-2 and MONALEESA-7 included patients with CNS metastases, and in both of them the addition of ribociclib with ET resulted in measurable tumor reductions conferring an advantage in ORR (54.5% vs. 38.8%; p < 0.001 and 51% vs. 36%; p < 0.001, respectively) [110].
Palbociclib is also being evaluated to demonstrate its efficacy in BCBM, and we await the results from ongoing trials.

3.5. Poly (ADP-Ribose) Polymerase Inhibitors

Veliparib + WBRT was studied in a phase I trial that included BCBM and an MOS of 7.7 months was reported, compared to 4.9 months based on historical data [111].
The phase III EMBRACA trial explored the efficacy of talazoparib treatment in patients with BRCA-mutated MBC. The median PFS was higher (8.6 vs. 5.6 months) and the ORR was also higher (62.6% vs. 27.2%; p < 0.001) in the talazoparib group, and a subanalysis of patients from this study with BM presented similar results [112].
OlympiAD, a phase III trial, evaluated olaparib for MBC, and while there was no statistically significant improvement in OS with olaparib, there was a possibly of meaningful OS benefit among patients who had not received chemotherapy for MBC [113].
In BROCADE3, a phase III trial that compared veliparib + carboplatin + paclitaxel vs. placebo + carboplatin + paclitaxel in BRCA-mutated advanced BC, the addition of veliparib to carboplatin + paclitaxel provided a longer PFS than that observed in the placebo group (14.5 vs. 12.6 months; p = 0.0016). However, patients with active brain metastases were excluded from this study, and only 5% of the participants had a history of BCBM [114].
The SWOG trial, a phase II study of cisplatin +/− veliparib in MBC, showed that the addition of veliparib significantly improved PFS (5.7 vs. 4.3 months; p = 0.023) [115].

3.6. Sacituzumab Govitecan

Sacituzumab govitecan (SG) is an antibody–drug conjugate composed of an antitrophoblast cell-surface antigen 2 (Trop-2) IgG1 kappa antibody coupled to SN-38 (the active metabolite of irinotecan) that has presented positive results when combined with single-agent TPC [116]. Currently the SWOG 2007 study (NCT04647916) is evaluating intracranial ORR in patients with HER2-negative BCBM.
The primary results from TROPiCS-02, a randomized phase III study of SG vs. TPC in patients with HR-positive/HER2-negative MBC, demonstrated that SG improved median PFS when compared to TPC (5.5 vs. 4 months, hazard ratio: 0.66; p = 0.0003) as well as PFS at 6 and 12 months (46% vs. 30% and 21% vs. 7%, respectively) [117].

3.7. Etirinotecan Pegol

Etirinotecan pegol (EP), a novel long-acting topoisomerase-1 inhibitor, was studied in patients with BCBM by Cortes et al. in the phase III BEACON trial that compared EP to TPC. EP was associated with a significant reduction in the risk of death when compared to TPC (p < 0.01). The MOS was also higher on the EP group (10 vs. 4.8 months) [118].

3.8. Immune Checkpoint Inhibitors for BCBM

The expression of the programmed cell death protein 1 receptor ligand (PD-L1) has been suggested to be a therapeutic target of immune checkpoint inhibitors in BCBM [119].
KEYNOTE-012, a phase Ib randomized trial of single-agent pembrolizumab that included patients with advanced PD-L1-positive TNBC (and other types of malignancies) with treated and stable BM, demonstrated an overall response rate of 18.5% [120].
KEYNOTE-355, a phase III trial that studied pembrolizumab + chemotherapy in advanced TNBC, including patients with previously treated BM, resulted in a significantly longer MOS in the pembrolizumab-chemotherapy group vs. the placebo-chemotherapy group (23 vs. 16.1 months, hazard ratio: 0.73; p = 0.0185) [121].
Impassion130, a phase III clinical trial investigating Atezolizumab + Nab-Paclitaxel vs. placebo + Nab-paclitaxel reported a prolonged MOS among patients with metastatic TNBC (including BCBM) in both the intention-to-treat population (21.3 vs. 17.6 months, hazard ratio: 0.84; p = 0.08) and the PD-L1 + subgroup (25 vs. 15.5 months, hazard ratio: 0.62; p < 0.001) [122].
New studies are being developed to investigate the efficacy of immune checkpoint inhibitors in BCBM, including a phase I/II trial evaluating pembrolizumab + SRS (NCT03449238), a phase I study analyzing SRS after nivolumab (NCT03807765), and a randomized phase III trial exploring the use of nivolumab and iplimumab in solid tumor BM (NCT04434560), among others [67].

3.9. Other Therapies for BCBM

There are currently studies recruiting patients to evaluate the efficacy of chimeric antigen receptor (CAR)-T cell therapies in HER2-positive BCBM (NCT02442297 and NCT03696030). Other trials investigating the effects of dendritic cell therapy for BCBM (NCT03638765) and proteome-based immunotherapy of BCBM (NCT01782274) are under development [123].
ANG1005, a novel taxane derivative consisting of three paclitaxel molecules covalently linked to Angiopep-2, designed to cross the BBB and BTB and to penetrate malignant cells via the lipoprotein-receptor-related protein 1 (LRP-1) transport system, has demonstrated intracranial response, symptom improvement, and prolonged overall survival compared to historical controls in different studies on BCBM [124,125]. Unlike native paclitaxel, ANG1005 was proven not to be a substrate for the multiple drug resistance (MDR) efflux pump in in vitro studies and has shown similar brain penetration in mice. This indicates that ANG1005 can be used to treat both peripheral metastatic BC and BCBM, even if the tumor develops resistance to conventional taxanes [126].
The efficacy of these therapies needs further evaluation.
Table 2 summarizes the majority of the systemic therapies for BCBM.

4. Clinical Approach

The treatment of BCBM will depend on the number of BM, the ER/PR and HER2 status of the tumor, and the control or progression of intra- and extracranial disease (Figure 1).
For patients with a single BM, surgical resection is suggested, as it can improve OS, especially in symptomatic patients when systemic disease is well-controlled. If patients present one to four BM, surgery + SRS with or without WBRT should be considered to improve local control [127]. Memantine and hippocampal avoidance should be offered to patients with no hippocampal lesions and 4 months or more of expected survival [128].
In patients with HER2-positive metastatic disease and limited asymptomatic intracranial disease, upfront systemic therapy can be the initial treatment instead of RT. For patients who are T-DM1-naïve, a deferral of RT and treatment with T-DM1 with the intent to treat BM may be appropriate [62].
For patients who have no evidence of extracranial disease and achieve an excellent clinical response rate after local treatment for BCBM, there are no prospective data to inform the benefits of systemic treatment.
Patients with progressive/new intracranial and progressive extracranial disease or no feasible local therapy option should undergo treatment with systemic therapy based on their HER2 status.
If patients have HER2-negative disease, single-agent chemotherapy with drugs such as fluorouracil, capecitabine, platinums, or doxorubicin has been described to have activity against CNS metastases [129,130]. Patients with hormone-receptor-positive disease can receive tamoxifen, aromatase inhibitors, or other similar agents; however, ET should not be used alone as a monotherapy, and local therapies are still necessary [131,132,133]. This is where CDK 4–6 inhibitors can be used as well.
If patients have HER2-positive disease, patients should receive a combination of Pertuzumab + Trastuzumab + Taxane-based chemotherapy. If patients progress after this, the use of a combination with Tucatinib + Trastuzumab + Capecitabine is recommended [86]. If patients progress after the previous treatment, the data recommend the use of T-DXd [134]. Meanwhile, some authors recommend the use of T-DM1 instead [61]. Additional lines may include the use of neratinib in combination with capecitabine, which has shown an intracranial response in BCBM [82].

5. Conclusions

The presence of CNS metastases in BC is associated with limited survival, and its incidence is increasing with the new development of BC therapies. However, although still limited, the treatment of BCBM has shown promising results for a better prognosis and prolonged PFS in multiple studies. Local treatments, including surgery, WBRT, and SRS, are becoming more conservative, limiting cognitive decline, and enhancing quality of life.
Targeted therapies for BCBM have been established, and multiple clinical trials with drugs directed against HER2, VEGF, mTOR, PI3K, EGFR, and CKD4/6; immune checkpoint inhibitors; and even CAR-T-cell therapy have presented positive outcomes in this disease.
The promising results from the trials summarized in this article should encourage further studies of the treatment of BCBM.

Author Contributions

J.A. prepared the manuscript; J.P.L. revised the manuscript; J.A. and J.P.L. designed the manuscript, performed the literature search, and wrote the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

Jorge Avila declares no conflict of interest. José Pablo Leone has received research funding from Kazia therapeutics and consulting fees from Minerva Biotechnologies.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ferlay, J.; Soerjomataram, I.; Dikshit, R.; Eser, S.; Mathers, C.; Rebelo, M.; Parkin, D.M.; Forman, D.; Bray, F. Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int. J. Cancer 2015, 136, E359–E386. [Google Scholar] [CrossRef]
  2. Chew, S.; Carroll, H.K.; Darwish, W.; Boychak, O.; Higgins, M.; McCaffrey, J.; Kelly, C.M. Characterization of Treatments and Disease Course for Women with Breast Cancer Brain Metastases: 5-Year Retrospective Single Institution Experience. Cancer Manag. Res. 2021, 13, 8191–8198. [Google Scholar] [CrossRef] [PubMed]
  3. Kim, H.-J.; Im, S.-A.; Keam, B.; Kim, Y.-J.; Han, S.-W.; Kim, T.M.; Oh, D.-Y.; Kim, J.H.; Lee, S.-H.; Chie, E.K.; et al. Clinical outcome of central nervous system metastases from breast cancer: Differences in survival depending on systemic treatment. J. Neuro-Oncol. 2011, 106, 303–313. [Google Scholar] [CrossRef]
  4. Minisini, A.M.; Moroso, S.; Gerratana, L.; Giangreco, M.; Iacono, D.; Poletto, E.; Guardascione, M.; Fontanella, C.; Fasola, G.; Puglisi, F. Risk factors and survival outcomes in patients with brain metastases from breast cancer. Clin. Exp. Metastasis 2013, 30, 951–956. [Google Scholar] [CrossRef] [PubMed]
  5. Lin, N.U.; Claus, E.; Sohl, J.; Razzak, A.R.; Arnaout, A.; Winer, E.P. Sites of distant recurrence and clinical outcomes in patients with metastatic triple-negative breast cancer: High incidence of central nervous system metastases. Cancer 2008, 113, 2638–2645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Kim, J.S.; Kim, I.A. Evolving treatment strategies of brain metastases from breast cancer: Current status and future direction. Ther. Adv. Med. Oncol. 2020, 12, 1758835920936117. [Google Scholar] [CrossRef] [PubMed]
  7. Niikura, N.; Hayashi, N.; Masuda, N.; Takashima, S.; Nakamura, R.; Watanabe, K.; Kanbayashi, C.; Ishida, M.; Hozumi, Y.; Tsuneizumi, M.; et al. Treatment outcomes and prognostic factors for patients with brain metastases from breast cancer of each subtype: A multicenter retrospective analysis. Breast Cancer Res. Treat. 2014, 147, 103–112. [Google Scholar] [CrossRef]
  8. Miyazawa, K.; Shikama, N.; Okazaki, S.; Koyama, T.; Takahashi, T.; Kato, S. Predicting prognosis of short survival time after palliative whole-brain radiotherapy. J. Radiat. Res. 2018, 59, 43–49. [Google Scholar] [CrossRef] [Green Version]
  9. Sperduto, P.W.; Berkey, B.; Gaspar, L.E.; Mehta, M.; Curran, W. A new prognostic index and comparison to three other indices for patients with brain metastases: An analysis of 1,960 patients in the RTOG database. Int. J. Radiat. Oncol. Biol. Phys. 2008, 70, 510–514. [Google Scholar] [CrossRef]
  10. Sperduto, P.W.; Mesko, S.; Li, J.; Cagney, D.; Aizer, A.; Lin, N.U.; Nesbit, E.; Kruser, T.J.; Chan, J.; Braunstein, S.; et al. Beyond an Updated Graded Prognostic Assessment (Breast GPA): A Prognostic Index and Trends in Treatment and Survival in Breast Cancer Brain Metastases From 1985 to Today. Int. J. Radiat. Oncol. Biol. Phys. 2020, 107, 334–343. [Google Scholar] [CrossRef]
  11. Musolino, A.; Ciccolallo, L.; Panebianco, M.; Fontana, E.; Zanoni, D.; Bozzetti, C.; Michiara, M.; Silini, E.M.; Ardizzoni, A. Multifactorial central nervous system recurrence susceptibility in patients with HER2-positive breast cancer: Epidemiological and clinical data from a population-based cancer registry study. Cancer 2011, 117, 1837–1846. [Google Scholar] [CrossRef] [PubMed]
  12. Brufsky, A.M.; Mayer, M.; Rugo, H.S.; Kaufman, P.A.; Tan-Chiu, E.; Tripathy, D.; Tudor, L.C.; Wang, L.I.; Brammer, M.G.; Shing, M.; et al. Central nervous system metastases in patients with HER2-positive metastatic breast cancer: Incidence, treatment, and survival in patients from registHER. Clin. Cancer Res. 2011, 17, 4834–4843. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Diao, K.; Sosa, A.J.; Zada, G.; Nagpal, S.; Chang, E.L. Management of complications from brain metastasis treatment: A narrative review. Chin. Clin. Oncol. 2021, 11, 11. [Google Scholar] [CrossRef]
  14. Patchell, R.A.; Tibbs, P.A.; Walsh, J.W.; Dempsey, R.J.; Maruyama, Y.; Kryscio, R.J.; Markesbery, W.R.; Macdonald, J.S.; Young, B. A randomized trial of surgery in the treatment of single metastases to the brain. N. Engl. J. Med. 1990, 322, 494–500. [Google Scholar] [CrossRef]
  15. Abbott, N.J. Blood-brain barrier structure and function and the challenges for CNS drug delivery. J. Inherit. Metab. Dis. 2013, 36, 437–449. [Google Scholar] [CrossRef] [PubMed]
  16. Rhea, E.M.; Banks, W.A. Role of the Blood-Brain Barrier in Central Nervous System Insulin Resistance. Front. Neurosci. 2019, 13, 521. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Lockman, P.R.; Mittapalli, R.K.; Taskar, K.S.; Rudraraju, V.; Gril, B.; Bohn, K.A.; Adkins, C.; Roberts, A.; Thorsheim, H.; Gaasch, J.A.; et al. Heterogeneous blood-tumor barrier permeability determines drug efficacy in experimental brain metastases of breast cancer. Clin. Cancer Res. 2010, 16, 5664–5678. [Google Scholar] [CrossRef] [Green Version]
  18. Cali Daylan, A.E.; Leone, J.P. Targeted Therapies for Breast Cancer Brain Metastases. Clin. Breast Cancer 2021, 21, 263–270. [Google Scholar] [CrossRef]
  19. Yonemori, K.; Tsuta, K.; Ono, M.; Shimizu, C.; Hirakawa, A.; Hasegawa, T.; Hatanaka, Y.; Narita, Y.; Shibui, S.; Fujiwara, Y. Disruption of the blood brain barrier by brain metastases of triple-negative and basal-type breast cancer but not HER2/neu-positive breast cancer. Cancer 2010, 116, 302–308. [Google Scholar] [CrossRef]
  20. Upadhyay, R.K. Drug delivery systems, CNS protection, and the blood brain barrier. Biomed. Res. Int. 2014, 2014, 869269. [Google Scholar] [CrossRef]
  21. Hart, E.; Odé, Z.; Derieppe, M.P.P.; Groenink, L.; Heymans, M.W.; Otten, R.; Lequin, M.H.; Janssens, G.O.R.; Hoving, E.W.; van Vuurden, D.G. Blood-brain barrier permeability following conventional photon radiotherapy–A systematic review and meta-analysis of clinical and preclinical studies. Clin. Transl. Radiat. Oncol. 2022, 35, 44–55. [Google Scholar] [CrossRef] [PubMed]
  22. Mampre, D.; Mehkri, Y.; Rajkumar, S.; Sriram, S.; Hernandez, J.; Lucke-Wold, B.; Chandra, V. Treatment of breast cancer brain metastases: Radiotherapy and emerging preclinical approaches. Diagn. Ther. 2022, 1, 25–38. [Google Scholar] [CrossRef] [PubMed]
  23. Tsao, M.N.; Rades, D.; Wirth, A.; Lo, S.S.; Danielson, B.L.; Gaspar, L.E.; Sperduto, P.W.; Vogelbaum, M.A.; Radawski, J.D.; Wang, J.Z.; et al. Radiotherapeutic and surgical management for newly diagnosed brain metastasis(es): An American Society for Radiation Oncology evidence-based guideline. Pract. Radiat. Oncol. 2012, 2, 210–225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Bhangoo, S.S.; Linskey, M.E.; Kalkanis, S.N. Evidence-Based Guidelines for the Management of Brain Metastases. Neurosurg. Clin. N. Am. 2011, 22, 97–104. [Google Scholar] [CrossRef] [PubMed]
  25. McTyre, E.; Scott, J.; Chinnaiyan, P. Whole brain radiotherapy for brain metastasis. Surg. Neurol. Int. 2013, 4 (Suppl. 4), S236–S244. [Google Scholar] [CrossRef] [PubMed]
  26. Patchell, R.A.; Tibbs, P.A.; Regine, W.F.; Dempsey, R.J.; Mohiuddin, M.; Kryscio, R.J.; Markesbery, W.R.; Foon, K.A.; Young, B. Postoperative radiotherapy in the treatment of single metastases to the brain: A randomized trial. JAMA 1998, 280, 1485–1489. [Google Scholar] [CrossRef]
  27. Kalkanis, S.N.; Kondziolka, D.; Gaspar, L.E.; Burri, S.H.; Asher, A.L.; Cobbs, C.S.; Ammirati, M.; Robinson, P.D.; Andrews, D.W.; Loeffler, J.S.; et al. The role of surgical resection in the management of newly diagnosed brain metastases: A systematic review and evidence-based clinical practice guideline. J. Neurooncol. 2010, 96, 33–43. [Google Scholar] [CrossRef] [Green Version]
  28. Le Rhun, É.; Dhermain, F.; Noël, G.; Reyns, N.; Carpentier, A.; Mandonnet, E.; Tailibert, S.; Metellus, P.; ANOCEF: L’Association des Neuro-Oncologues d’Expression Francaise. ANOCEF guidelines for the management of brain metastases. Cancer Radiother. 2015, 19, 66–71. [Google Scholar] [CrossRef]
  29. Borgelt, B.; Gelber, R.; Kramer, S.; Brady, L.W.; Chang, C.H.; Davis, L.W.; Perez, C.A.; Hendrickson, F.R. The palliation of brain metastases: Final results of the first two studies by the Radiation Therapy Oncology Group. Int. J. Radiat. Oncol. Biol. Phys. 1980, 6, 1–9. [Google Scholar] [CrossRef]
  30. Lokich, J.J. The management of cerebral metastasis. JAMA 1975, 234, 748–751. [Google Scholar] [CrossRef]
  31. Sneed, P.K.; Larson, D.A.; Wara, W.M. Radiotherapy for cerebral metastases. Neurosurg. Clin. N. Am. 1996, 7, 505–515. [Google Scholar] [CrossRef]
  32. Gaspar, L.; Scott, C.; Rotman, M.; Asbell, S.; Phillips, T.; Wasserman, T.; McKenna, W.G.; Byhardt, R. Recursive partitioning analysis (RPA) of prognostic factors in three Radiation Therapy Oncology Group (RTOG) brain metastases trials. Int. J. Radiat. Oncol. Biol. Phys. 1997, 37, 745–751. [Google Scholar] [CrossRef]
  33. Tanguturi, S.; Warren, L.E.G. The Current and Evolving Role of Radiation Therapy for Central Nervous System Metastases from Breast Cancer. Curr. Oncol. Rep. 2019, 21, 50. [Google Scholar] [CrossRef] [PubMed]
  34. Brown, P.D.; Jaeckle, K.; Ballman, K.V.; Farace, E.; Cerhan, J.H.; Anderson, S.K.; Carrero, X.W.; Barker, F.G.; Deming, R.; Burri, S.H.; et al. Effect of Radiosurgery Alone vs Radiosurgery With Whole Brain Radiation Therapy on Cognitive Function in Patients With 1 to 3 Brain Metastases: A Randomized Clinical Trial. JAMA 2016, 316, 401–409. [Google Scholar] [CrossRef]
  35. Kayama, T.; Sato, S.; Sakurada, K.; Mizusawa, J.; Nishikawa, R.; Narita, Y.; Sumi, M.; Miyakita, Y.; Kumabe, T.; Sonoda, Y.; et al. Effects of Surgery With Salvage Stereotactic Radiosurgery Versus Surgery With Whole-Brain Radiation Therapy in Patients With One to Four Brain Metastases (JCOG0504): A Phase III, Noninferiority, Randomized Controlled Trial. J. Clin. Oncol. 2018, 36, 3282–3289. [Google Scholar] [CrossRef]
  36. Andrews, D.W.; Scott, C.B.; Sperduto, P.W.; Flanders, A.E.; Gaspar, L.E.; Schell, M.C.; Werner-Wasik, M.; Demas, W.; Ryu, J.; Bahary, J.P.; et al. Whole brain radiation therapy with or without stereotactic radiosurgery boost for patients with one to three brain metastases: Phase III results of the RTOG 9508 randomised trial. Lancet 2004, 363, 1665–1672. [Google Scholar] [CrossRef]
  37. Aoyama, H.; Shirato, H.; Tago, M.; Nakagawa, K.; Toyoda, T.; Hatano, K.; Kenjyo, M.; Oya, N.; Hirota, S.; Shioura, H.; et al. Stereotactic radiosurgery plus whole-brain radiation therapy vs stereotactic radiosurgery alone for treatment of brain metastases: A randomized controlled trial. JAMA 2006, 295, 2483–2491. [Google Scholar] [CrossRef]
  38. Brown, P.D.; Ballman, K.V.; Cerhan, J.H.; Anderson, S.K.; Carrero, X.W.; Whitton, A.C.; Greenspoon, J.; Parney, I.F.; Laack, N.N.; Ashman, J.B.; et al. Postoperative stereotactic radiosurgery compared with whole brain radiotherapy for resected metastatic brain disease (NCCTG N107C/CEC·3): A multicentre, randomised, controlled, phase 3 trial. Lancet Oncol. 2017, 18, 1049–1060. [Google Scholar] [CrossRef]
  39. Li, J.; Ludmir, E.B.; Wang, Y.; Guha-Thakurta, N.; McAleer, M.F.; Settle, S.H.; Yeboa, D.N.; Ghia, A.J.; McGovern, S.L.; Chung, C.; et al. Stereotactic Radiosurgery versus Whole-brain Radiation Therapy for Patients with 4-15 Brain Metastases: A Phase III Randomized Controlled Trial. Int. J. Radiat. Oncol. Biol. Phys. 2020, 108, S21–S22. [Google Scholar] [CrossRef]
  40. Manning, M.A.; Cardinale, R.M.; Benedict, S.H.; Kavanagh, B.D.; Zwicker, R.D.; Amir, C.; Broaddus, W.C. Hypofractionated stereotactic radiotherapy as an alternative to radiosurgery for the treatment of patients with brain metastases. Int. J. Radiat. Oncol. Biol. Phys. 2000, 47, 603–608. [Google Scholar] [CrossRef]
  41. Kim, Y.J.; Cho, K.H.; Kim, J.Y.; Lim, Y.K.; Min, H.S.; Lee, S.H.; Kim, H.J.; Gwak, H.S.; Yoo, H.; Lee, S.H. Single-dose versus fractionated stereotactic radiotherapy for brain metastases. Int. J. Radiat. Oncol. Biol. Phys. 2011, 81, 483–489. [Google Scholar] [CrossRef] [PubMed]
  42. Redmond, K.J.; De Salles, A.A.F.; Fariselli, L.; Levivier, M.; Ma, L.; Paddick, I.; Pollock, B.; Regis, J.; Sheehan, J.; Suh, J.; et al. Stereotactic Radiosurgery for Postoperative Metastatic Surgical Cavities: A Critical Review and International Stereotactic Radiosurgery Society (ISRS) Practice Guidelines. Int. J. Radiat. Oncol. Biol. Phys. 2021, 111, 68–80. [Google Scholar] [CrossRef] [PubMed]
  43. Single Fraction Stereotactic Radiosurgery Compared with Fractionated Stereotactic Radiosurgery in Treating Patients with Resected Metastatic Brain Disease. Available online: https://ClinicalTrials.gov/show/NCT04114981 (accessed on 10 October 2022).
  44. Ippolito, E.; Silipigni, S.; Matteucci, P.; Greco, C.; Carrafiello, S.; Palumbo, V.; Tacconi, C.; Talocco, C.; Fiore, M.; D’Angelillo, R.; et al. Radiotherapy for HER 2 Positive Brain Metastases: Urgent Need for a Paradigm Shift. Cancers 2022, 14, 1514. [Google Scholar] [CrossRef] [PubMed]
  45. Gondi, V.; Pugh, S.L.; Tome, W.A.; Caine, C.; Corn, B.; Kanner, A.; Rowley, H.; Kundapur, V.; DeNittis, A.; Greenspoon, J.N.; et al. Preservation of memory with conformal avoidance of the hippocampal neural stem-cell compartment during whole-brain radiotherapy for brain metastases (RTOG 0933): A phase II multi-institutional trial. J. Clin. Oncol. 2014, 32, 3810–3816. [Google Scholar] [CrossRef]
  46. Sun, B.; Huang, Z.; Wu, S.; Shen, G.; Cha, L.; Meng, X.; Ding, L.; Wang, J.; Song, S. Incidence and relapse risk of intracranial metastases within the perihippocampal region in 314 patients with breast cancer. Radiother. Oncol. 2016, 118, 181–186. [Google Scholar] [CrossRef] [PubMed]
  47. Pardridge, W.M.; Buciak, J.L.; Friden, P.M. Selective transport of an anti-transferrin receptor antibody through the blood-brain barrier in vivo. J. Pharmacol. Exp. Ther. 1991, 259, 66–70. [Google Scholar]
  48. Zhang, Y.; Pardridge, W.M. Mediated efflux of IgG molecules from brain to blood across the blood-brain barrier. J. Neuroimmunol. 2001, 114, 168–172. [Google Scholar] [CrossRef]
  49. Hudis, C.A. Trastuzumab--mechanism of action and use in clinical practice. N. Engl. J. Med. 2007, 357, 39–51. [Google Scholar] [CrossRef] [Green Version]
  50. Stemmler, H.J.; Schmitt, M.; Willems, A.; Bernhard, H.; Harbeck, N.; Heinemann, V. Ratio of trastuzumab levels in serum and cerebrospinal fluid is altered in HER2-positive breast cancer patients with brain metastases and impairment of blood-brain barrier. Anticancer Drugs 2007, 18, 23–28. [Google Scholar] [CrossRef]
  51. Park, Y.H.; Park, M.J.; Ji, S.H.; Yi, S.Y.; Lim, D.H.; Nam, D.H.; Lee, J.I.; Park, W.; Choi, D.H.; Huh, S.J.; et al. Trastuzumab treatment improves brain metastasis outcomes through control and durable prolongation of systemic extracranial disease in HER2-overexpressing breast cancer patients. Br. J. Cancer 2009, 100, 894–900. [Google Scholar] [CrossRef]
  52. Bendell, J.C.; Domchek, S.M.; Burstein, H.J.; Harris, L.; Younger, J.; Kuter, I.; Bunnell, C.; Rue, M.; Gelman, R.; Winer, E. Central nervous system metastases in women who receive trastuzumab-based therapy for metastatic breast carcinoma. Cancer 2003, 97, 2972–2977. [Google Scholar] [CrossRef] [PubMed]
  53. Clayton, A.J.; Danson, S.; Jolly, S.; Ryder, W.D.; Burt, P.A.; Stewart, A.L.; Wilkinson, P.M.; Welch, R.S.; Magee, B.; Wilson, G.; et al. Incidence of cerebral metastases in patients treated with trastuzumab for metastatic breast cancer. Br. J. Cancer 2004, 91, 639–643. [Google Scholar] [CrossRef] [PubMed]
  54. Dawood, S.; Broglio, K.; Esteva, F.J.; Ibrahim, N.K.; Kau, S.W.; Islam, R.; Aldape, K.D.; Yu, T.K.; Hortobagyi, G.N.; Gonzalez-Angulo, A.M. Defining prognosis for women with breast cancer and CNS metastases by HER2 status. Ann. Oncol. 2008, 19, 1242–1248. [Google Scholar] [CrossRef] [PubMed]
  55. Stemmler, H.J.; Schmitt, M.; Harbeck, N.; Willems, A.; Bernhard, H.; Lässig, D.; Schoenber, S.; Heinemann, V. Application of intrathecal trastuzumab (Herceptintrade mark) for treatment of meningeal carcinomatosis in HER2-overexpressing metastatic breast cancer. Oncol. Rep. 2006, 15, 1373–1377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Mir, O.; Ropert, S.; Alexandre, J.; Lemare, F.; Goldwasser, F. High-dose intrathecal trastuzumab for leptomeningeal metastases secondary to HER-2 overexpressing breast cancer. Ann. Oncol. 2008, 19, 1978–1980. [Google Scholar] [CrossRef]
  57. Bousquet, G.; Darrouzain, F.; de Bazelaire, C.; Ternant, D.; Barranger, E.; Winterman, S.; Madelaine-Chambin, I.; Thiebaut, J.; Polivka, M.; Paintaud, G.; et al. Intrathecal Trastuzumab Halts Progression of CNS Metastases in Breast Cancer. J. Clin. Oncol. 2014, 34, e151–e155. [Google Scholar] [CrossRef]
  58. Kumthekar, P.U.; Avram, M.J.; Lassman, A.B.; Lin, N.U.; Lee, E.; Grimm, S.A.; Schwartz, M.; Burdett, K.L.; Lukas, R.; Dixit, K.; et al. A Phase I/II Study of Intrathecal Trastuzumab in HER-2 Positive Cancer with Leptomeningeal Metastases: Safety, Efficacy, and Cerebrospinal Fluid Pharmacokinetics. Neuro-Oncology 2022, in press. [Google Scholar] [CrossRef]
  59. Fabi, A.; Alesini, D.; Valle, E.; Moscetti, L.; Caputo, R.; Caruso, M.; Carbognin, L.; Ciccarese, M.; La Verde, N.; Arpino, G.; et al. T-DM1 and brain metastases: Clinical outcome in HER2-positive metastatic breast cancer. Breast 2018, 41, 137–143. [Google Scholar] [CrossRef]
  60. Jacot, W.; Pons, E.; Frenel, J.S.; Guiu, S.; Levy, C.; Heudel, P.E.; Bachelot, T.; D’Hondt, V.; Darlix, A.; Firmin, N.; et al. Efficacy and safety of trastuzumab emtansine (T-DM1) in patients with HER2-positive breast cancer with brain metastases. Breast Cancer Res. Treat. 2016, 157, 307–318. [Google Scholar] [CrossRef]
  61. Krop, I.E.; Lin, N.U.; Blackwell, K.; Guardino, E.; Huober, J.; Lu, M.; Miles, D.; Samant, M.; Welslau, M.; Dieras, V. Trastuzumab emtansine (T-DM1) versus lapatinib plus capecitabine in patients with HER2-positive metastatic breast cancer and central nervous system metastases: A retrospective, exploratory analysis in EMILIA. Ann. Oncol. 2015, 26, 113–119. [Google Scholar] [CrossRef]
  62. Montemurro, F.; Delaloge, S.; Barrios, C.H.; Wuerstlein, R.; Anton, A.; Brain, E.; Hatschek, T.; Kelly, C.M.; Pena-Murillo, C.; Yilmaz, M.; et al. Trastuzumab emtansine (T-DM1) in patients with HER2-positive metastatic breast cancer and brain metastases: Exploratory final analysis of cohort 1 from KAMILLA, a single-arm phase IIIb clinical trial(☆). Ann. Oncol. 2020, 31, 1350–1358. [Google Scholar] [CrossRef] [PubMed]
  63. Lee, J.; Park, Y.H. Trastuzumab deruxtecan for HER2+ advanced breast cancer. Future Oncol. 2022, 18, 7–19. [Google Scholar] [CrossRef] [PubMed]
  64. Cortés, J.; Kim, S.; Chung, W.; Im, S.; Park, Y.; Hegg, R.; Kim, M.; Tseng, L.; Perty, V.; Chung, C.; et al. LBA1 Trastuzumab deruxtecan (T-DXd) vs trastuzumab emtansine (T-DM1) in patients (Pts) with HER2+ metastatic breast cancer (mBC): Results of the randomized phase III DESTINY-Breast03 study. Ann. Oncol. 2021, 32, S1287–S1288. [Google Scholar] [CrossRef]
  65. Hurvitz, S.; Kim, S.B.; Chung, W.P.; Im, S.A.; Park, Y.H.; Hegg, R.; Kim, M.H.; Tseng, L.M.; Petry, V.; Chung, C.F.; et al. GS3-01. Trastuzumab Deruxtecan (T-DXd; DS-8201a) vs. Trastuzumab Emtansine (T-DM1) in Patients (pts) with HER2+ Metastatic Breast Cancer (mBC): Subgroup Analyses from the Randomized Phase 3 Study DESTINY-Breast03. 2021. Available online: https://www.abstractsonline.com/pp8/#!/10462/presentation/649 (accessed on 16 January 2022).
  66. Modi, S.; Jacot, W.; Yamashita, T.; Sohn, J.; Vidal, M.; Tokunaga, E.; Tsurutani, J.; Ueno, N.T.; Prat, A.; Chae, Y.S.; et al. Trastuzumab Deruxtecan in Previously Treated HER2-Low Advanced Breast Cancer. N. Engl. J. Med. 2022, 387, 9–20. [Google Scholar] [CrossRef]
  67. Watase, C.; Shiino, S.; Shimoi, T.; Noguchi, E.; Kaneda, T.; Yamamoto, Y.; Yonemori, K.; Takayama, S.; Suto, A. Breast Cancer Brain Metastasis-Overview of Disease State, Treatment Options and Future Perspectives. Cancers 2021, 13, 1078. [Google Scholar] [CrossRef]
  68. Swain, S.M.; Baselga, J.; Miles, D.; Im, Y.H.; Quah, C.; Lee, L.F.; Cortés, J. Incidence of central nervous system metastases in patients with HER2-positive metastatic breast cancer treated with pertuzumab, trastuzumab, and docetaxel: Results from the randomized phase III study CLEOPATRA. Ann. Oncol. 2014, 25, 1116–1121. [Google Scholar] [CrossRef]
  69. Swain, S.M.; Miles, D.; Kim, S.B.; Im, Y.H.; Im, S.A.; Semiglazov, V.; Ciruelos, E.; Schneeweiss, A.; Loi, S.; Monturus, E.; et al. Pertuzumab, trastuzumab, and docetaxel for HER2-positive metastatic breast cancer (CLEOPATRA): End-of-study results from a double-blind, randomised, placebo-controlled, phase 3 study. Lancet Oncol. 2020, 21, 519–530. [Google Scholar] [CrossRef]
  70. Lin, N.U.; Pegram, M.; Sahebjam, S.; Ibrahim, N.; Fung, A.; Cheng, A.; Nicholas, A.; Kirschbrown, W.; Kumthekar, P. Pertuzumab Plus High-Dose Trastuzumab in Patients With Progressive Brain Metastases and HER2-Positive Metastatic Breast Cancer: Primary Analysis of a Phase II Study. J. Clin. Oncol. 2021, 39, 2667–2675. [Google Scholar] [CrossRef]
  71. Lin, N.U.; Carey, L.A.; Liu, M.C.; Younger, J.; Come, S.E.; Ewend, M.; Harris, G.J.; Bullitt, E.; Van den Abbeele, A.; Henson, J.W.; et al. Phase II trial of lapatinib for brain metastases in patients with human epidermal growth factor receptor 2-positive breast cancer. J. Clin. Oncol. 2008, 26, 1993–1999. [Google Scholar] [CrossRef] [Green Version]
  72. Lin, N.U.; Diéras, V.; Paul, D.; Lossignol, D.; Christodoulou, C.; Stemmler, H.J.; Roche, H.; Liu, M.C.; Greli, R.; Ciruelos, E.; et al. Multicenter phase II study of lapatinib in patients with brain metastases from HER2-positive breast cancer. Clin. Cancer Res. 2009, 15, 1452–1459. [Google Scholar] [CrossRef] [Green Version]
  73. Metro, G.; Foglietta, J.; Russillo, M.; Stocchi, L.; Vidiri, A.; Giannarelli, D.; Crino, L.; Papaldo, P.; Mottolese, M.; Cognetti, F.; et al. Clinical outcome of patients with brain metastases from HER2-positive breast cancer treated with lapatinib and capecitabine. Ann. Oncol. 2011, 22, 625–630. [Google Scholar] [CrossRef] [PubMed]
  74. Bachelot, T.; Romieu, G.; Campone, M.; Diéras, V.; Cropet, C.; Dalenc, F.; Jimenez, M.; Le Rhun, E.; Pierga, J.Y.; Goncalvez, A.; et al. Lapatinib plus capecitabine in patients with previously untreated brain metastases from HER2-positive metastatic breast cancer (LANDSCAPE): A single-group phase 2 study. Lancet Oncol. 2013, 14, 64–71. [Google Scholar] [CrossRef]
  75. Pivot, X.; Manikhas, A.; Żurawski, B.; Chmielowska, E.; Karaszewska, B.; Allerton, R.; Chan, S.; Fabi, A.; Bidoli, P.; Gori, S.; et al. CEREBEL (EGF111438): A Phase III, Randomized, Open-Label Study of Lapatinib Plus Capecitabine Versus Trastuzumab Plus Capecitabine in Patients With Human Epidermal Growth Factor Receptor 2-Positive Metastatic Breast Cancer. J. Clin. Oncol. 2015, 33, 1564–1573. [Google Scholar] [CrossRef] [PubMed]
  76. Petrelli, F.; Ghidini, M.; Lonati, V.; Tomasello, G.; Borgonovo, K.; Ghilardi, M.; Cabiddu, M.; Barni, S. The efficacy of lapatinib and capecitabine in HER-2 positive breast cancer with brain metastases: A systematic review and pooled analysis. Eur. J. Cancer 2017, 84, 141–148. [Google Scholar] [CrossRef] [PubMed]
  77. Hayashi, N.; Niikura, N.; Masuda, N.; Takashima, S.; Nakamura, R.; Watanabe, K.; Kanbayashi, C.; Ishida, M.; Hozumi, Y.; Tsuneizumi, M.; et al. Prognostic factors of HER2-positive breast cancer patients who develop brain metastasis: A multicenter retrospective analysis. Breast Cancer Res. Treat. 2015, 149, 277–284. [Google Scholar] [CrossRef] [PubMed]
  78. Gelmon, K.A.; Boyle, F.M.; Kaufman, B.; Huntsman, D.G.; Manikhas, A.; Di Leo, A.; Martin, M.; Schwartzberg, L.S.; Lemieux, J.; Aparicio, S.; et al. Lapatinib or Trastuzumab Plus Taxane Therapy for Human Epidermal Growth Factor Receptor 2-Positive Advanced Breast Cancer: Final Results of NCIC CTG MA.31. J. Clin. Oncol. 2015, 33, 1574–1583. [Google Scholar] [CrossRef]
  79. Lin, N.U.; Freedman, R.A.; Ramakrishna, N.; Younger, J.; Storniolo, A.M.; Bellon, J.R.; Come, S.E.; Gelman, R.S.; Harris, G.J.; Henderson, M.A.; et al. A phase I study of lapatinib with whole brain radiotherapy in patients with Human Epidermal Growth Factor Receptor 2 (HER2)-positive breast cancer brain metastases. Breast Cancer Res. Treat. 2013, 142, 405–414. [Google Scholar] [CrossRef]
  80. Burstein, H.J.; Sun, Y.; Dirix, L.Y.; Jiang, Z.; Paridaens, R.; Tan, A.R.; Awada, A.; Ranade, A.; Jiao, S.; Schwartz, G.; et al. Neratinib, an irreversible ErbB receptor tyrosine kinase inhibitor, in patients with advanced ErbB2-positive breast cancer. J. Clin. Oncol. 2010, 28, 1301–1307. [Google Scholar] [CrossRef]
  81. Awada, A.; Colomer, R.; Inoue, K.; Bondarenko, I.; Badwe, R.A.; Demetriou, G.; Lee, S.C.; Mehta, A.O.; Kin, S.B.; Bachelot, T.; et al. Neratinib Plus Paclitaxel vs Trastuzumab Plus Paclitaxel in Previously Untreated Metastatic ERBB2-Positive Breast Cancer: The NEfERT-T Randomized Clinical Trial. JAMA Oncol. 2016, 2, 1557–1564. [Google Scholar] [CrossRef]
  82. Freedman, R.A.; Gelman, R.S.; Anders, C.K.; Melisko, M.E.; Parsons, H.A.; Cropp, A.M.; Silvestri, K.; Cotter, C.M.; Componeschi, K.P.; Marte, J.M.; et al. TBCRC 022: A Phase II Trial of Neratinib and Capecitabine for Patients With Human Epidermal Growth Factor Receptor 2-Positive Breast Cancer and Brain Metastases. J. Clin. Oncol. 2019, 37, 1081–1089. [Google Scholar] [CrossRef]
  83. Saura, C.; Oliveira, M.; Feng, Y.H.; Dai, M.S.; Chen, S.W.; Hurvitz, S.A.; Kim, S.B.; Moy, B.; Delaloge, S.; Gradishar, W.; et al. Neratinib Plus Capecitabine Versus Lapatinib Plus Capecitabine in HER2-Positive Metastatic Breast Cancer Previously Treated with ≥ 2 HER2-Directed Regimens: Phase III NALA Trial. J. Clin. Oncol. 2020, 38, 3138–3149. [Google Scholar] [CrossRef] [PubMed]
  84. Committee for Medicinal Products for Human Use (CHMP). Tukysa. International Non-Proprietary Name: Tucatinib; European Medicines Agency: Amsterdam, The Netherlands, 2021; pp. 1–148. [Google Scholar]
  85. Metzger Filho, O.; Leone, J.P.; Li, T.; Tan-Wasielewski, Z.; Trippa, L.; Barry, W.T.; Younger, J.; Lawler, E.; Walker, L.; Freedman, R.A.; et al. Phase I dose-escalation trial of tucatinib in combination with trastuzumab in patients with HER2-positive breast cancer brain metastases. Ann. Oncol. 2020, 31, 1231–1239. [Google Scholar] [CrossRef] [PubMed]
  86. Hurvitz, S.A.; Vahdat, L.T.; Harbeck, N.; Wolff, A.C.; Tolaney, S.M.; Loi, S.; Masuda, N.; O’Shaughnessy, J.; Dong, C.; Walker, L.; et al. 353TiP HER2CLIMB-02: A randomized, double-blind, phase III study of tucatinib or placebo with T-DM1 for unresectable locally advanced or metastatic HER2+ breast cancer. Ann. Oncol. 2020, 31, S390. [Google Scholar] [CrossRef]
  87. Murthy, R.; Borges, V.F.; Conlin, A.; Chaves, J.; Chamberlain, M.; Gray, T.; Vo, A.; Hamilton, E. Tucatinib with capecitabine and trastuzumab in advanced HER2-positive metastatic breast cancer with and without brain metastases: A non-randomised, open-label, phase 1b study. Lancet Oncol. 2018, 19, 880–888. [Google Scholar] [CrossRef]
  88. Murthy, R.K.; Loi, S.; Okines, A.; Paplomata, E.; Hamilton, E.; Hurvitz, S.A.; Lin, N.U.; Borges, V.; Abramson, V.; Anders, C.; et al. Tucatinib, Trastuzumab, and Capecitabine for HER2-Positive Metastatic Breast Cancer. N. Engl. J. Med. 2020, 382, 597–609. [Google Scholar] [CrossRef]
  89. Lin, N.U.; Borges, V.; Anders, C.; Murthy, R.K.; Paplomata, E.; Hamilton, E.; Hurvitz, S.; Loi, S.; Okines, A.; Abramson, V.; et al. Intracranial Efficacy and Survival With Tucatinib Plus Trastuzumab and Capecitabine for Previously Treated HER2-Positive Breast Cancer With Brain Metastases in the HER2CLIMB Trial. J. Clin. Oncol. 2020, 38, 2610–2619. [Google Scholar] [CrossRef]
  90. Bachelot, T.; Lin, N.U.; Murthy, R.K.; Hurvitz, S.A.; Borges, V.; Oliveira, M.; Oakman, C.; Khan, S.; Lynch, C.; Westbrook, K.; et al. 293P Impact of tucatinib on progression free survival in patients with HER2+ metastatic breast cancer and stable or active brain metastases. Ann. Oncol. 2020, 31, S359–S360. [Google Scholar] [CrossRef]
  91. Borges, V.F.; Ferrario, C.; Aucoin, N.; Falkson, C.; Khan, Q.; Krop, I.; Welch, S.; Conlin, A.; Chaves, J.; Bedard, P.L.; et al. Tucatinib Combined With Ado-Trastuzumab Emtansine in Advanced ERBB2/HER2-Positive Metastatic Breast Cancer: A Phase 1b Clinical Trial. JAMA Oncol. 2018, 4, 1214–1220. [Google Scholar] [CrossRef] [Green Version]
  92. Curigliano, G.; Mueller, V.; Borges, V.; Hamilton, E.; Hurvitz, S.; Loi, S.; Murthy, R.; Okines, A.; Paplomata, E.; Cameron, D.; et al. Tucatinib versus placebo added to trastuzumab and capecitabine for patients with pretreated HER2+ metastatic breast cancer with and without brain metastases (HER2CLIMB): Final overall survival analysis. Ann. Oncol. 2021, 33, 321–329. [Google Scholar] [CrossRef]
  93. Ma, F.; Ouyang, Q.; Li, W.; Jiang, Z.; Tong, Z.; Liu, Y.; Li, H.; Yu, S.; Feng, J.; Wang, S.; et al. Pyrotinib or Lapatinib Combined With Capecitabine in HER2-Positive Metastatic Breast Cancer With Prior Taxanes, Anthracyclines, and/or Trastuzumab: A Randomized, Phase II Study. J. Clin. Oncol. 2019, 37, 2610–2619. [Google Scholar] [CrossRef]
  94. Yan, M.; Ouyang, Q.; Sun, T.; Niu, L.; Yang, J.; Li, L.; Song, Y.; Hao, C.; Chen, Z.; Orlandi, A.; et al. Pyrotinib plus capecitabine for patients with human epidermal growth factor receptor 2-positive breast cancer and brain metastases (PERMEATE): A multicentre, single-arm, two-cohort, phase 2 trial. Lancet Oncol. 2022, 23, 353–361. [Google Scholar] [CrossRef]
  95. Miller, K.; Wang, M.; Gralow, J.; Dickler, M.; Cobleigh, M.; Perez, E.A.; Shenkier, T.; Cella, D.; Davidson, N.E. Paclitaxel plus bevacizumab versus paclitaxel alone for metastatic breast cancer. N. Engl. J. Med. 2007, 357, 2666–2676. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Leone, J.P.; Emblem, K.E.; Weitz, M.; Gelman, R.S.; Schneider, B.P.; Freedman, R.A.; Younger, J.; Pinho, M.C.; Sorensen, A.G.; Gerstner, E.R.; et al. Phase II trial of carboplatin and bevacizumab in patients with breast cancer brain metastases. Breast Cancer Res. 2020, 22, 131. [Google Scholar] [CrossRef] [PubMed]
  97. Lu, Y.S.; Chen, T.W.; Lin, C.H.; Yeh, D.C.; Tseng, L.M.; Wu, P.F.; Rau, K.M.; Chen, B.B.; Chao, T.C.; Huang, S.M.; et al. Bevacizumab preconditioning followed by Etoposide and Cisplatin is highly effective in treating brain metastases of breast cancer progressing from whole-brain radiotherapy. Clin. Cancer Res. 2015, 21, 1851–1858. [Google Scholar] [CrossRef] [Green Version]
  98. André, F.; Ciruelos, E.; Rubovszky, G.; Campone, M.; Loibl, S.; Rugo, H.S.; Iwata, H.; Conte, P.; Mayer, I.A.; Kaufman, B.; et al. Alpelisib for PIK3CA-Mutated, Hormone Receptor-Positive Advanced Breast Cancer. N. Engl. J. Med. 2019, 380, 1929–1940. [Google Scholar] [CrossRef]
  99. Dent, S.; Cortés, J.; Im, Y.H.; Diéras, V.; Harbeck, N.; Krop, I.E.; Wilson, T.R.; Cui, N.; Schimmoller, F.; Hsu, J.Y.; et al. Phase III randomized study of taselisib or placebo with fulvestrant in estrogen receptor-positive, PIK3CA-mutant, HER2-negative, advanced breast cancer: The SANDPIPER trial. Ann. Oncol. 2021, 32, 197–207. [Google Scholar] [CrossRef]
  100. Batalini, F.; Moulder, S.L.; Winer, E.P.; Rugo, H.S.; Lin, N.U.; Wulf, G.M. Response of Brain Metastases From PIK3CA-Mutant Breast Cancer to Alpelisib. JCO Precis. Oncol. 2020, 4, 572–578. [Google Scholar] [CrossRef]
  101. Maira, M.; Schnell, C.; Lollini, P.; Chouaid, C.; Schmid, P.; Nanni, P.; Lam, D.; Di Tomaso, E.; Massaces, C.; Rodon, J. Preclinical and Preliminary Clinical Activity of NVP-BKM120, an Oral Pan-Class I PI3K Inhibitor, in the Brain. Ann. Oncol. 2012, 23, ix537. [Google Scholar] [CrossRef]
  102. Di Leo, A.; Johnston, S.; Lee, K.S.; Ciruelos, E.; Lønning, P.E.; Janni, W.; O’Regan, R.; Mouret-Reynier, M.A.; Kalev, D.; Egle, D.; et al. Buparlisib plus fulvestrant in postmenopausal women with hormone-receptor-positive, HER2-negative, advanced breast cancer progressing on or after mTOR inhibition (BELLE-3): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2018, 19, 87–100. [Google Scholar] [CrossRef]
  103. Hurvitz, S.; Singh, R.; Adams, B.; Taguchi, J.A.; Chan, D.; Dichmann, R.A.; Castrellon, A.; Hu, E.; Berkowitz, J.; Mani, A.; et al. Phase Ib/II single-arm trial evaluating the combination of everolimus, lapatinib and capecitabine for the treatment of HER2-positive breast cancer with brain metastases (TRIO-US B-09). Ther. Adv. Med. Oncol. 2018, 10, 1758835918807339. [Google Scholar] [CrossRef] [Green Version]
  104. Van Swearingen, A.E.D.; Siegel, M.B.; Deal, A.M.; Sambade, M.J.; Hoyle, A.; Hayes, D.N.; Jo, H.; Little, P.; Dees, E.C.; Muss, H.; et al. LCCC 1025: A phase II study of everolimus, trastuzumab, and vinorelbine to treat progressive HER2-positive breast cancer brain metastases. Breast Cancer Res. Treat. 2018, 171, 637–648. [Google Scholar] [CrossRef] [PubMed]
  105. Sledge, G.W., Jr.; Toi, M.; Neven, P.; Sohn, J.; Inoue, K.; Pivot, X.; Burdaeva, O.; Okera, M.; Masuda, N.; Kaufman, P.; 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, 2875–2884. [Google Scholar] [CrossRef] [PubMed]
  106. Tolaney, S.M.; Sahebjam, S.; Le Rhun, E.; Bachelot, T.; Kabos, P.; Awada, A.; Yardley, D.; Chan, A.; Conte, P.; Diéras, V.; et al. A Phase II Study of Abemaciclib in Patients with Brain Metastases Secondary to Hormone Receptor-Positive Breast Cancer. Clin. Cancer Res. 2020, 26, 5310–5319. [Google Scholar] [CrossRef] [PubMed]
  107. Anders, C.K.; Rhun, E.L.; Bachelot, T.D.; Yardley, D.A.; Awada, A.; Conte, P.F.; Kabos, P.; Bear, M.; Yang, Z.; Chen, Y.; et al. A phase II study of abemaciclib in patients (pts) with brain metastases (BM) secondary to HR+, HER2- metastatic breast cancer (MBC). J. Clin. Oncol. 2019, 37 (Suppl. 15), 1017. [Google Scholar] [CrossRef]
  108. DeLaurentiis, M.; Borstnar, S.; Campone, M.; Warner, E.; Bofill, S.; Jacot, W.; Dent, S.F.; Martin, M.; Ring, A.E.; Cottu, P.; et al. Interim results from the full population of the phase 3b CompLEEment-1 study of ribociclib (RIBO) plus letrozole (LET) in the treatment of HR+/HER2– advanced breast cancer (ABC). J. Clin. Oncol. 2019, 37 (Suppl. 15), 1041. [Google Scholar] [CrossRef]
  109. Cottu, P.H.; De Laurentiis, M.; Marchetti, P.; Coltelli, L.; Califaretti, N.; Debled, M.; Patti, S.; Evron, E.; Duhoux, F.P.; Meno-Singh, L.; et al. Ribociclib (RIB) + letrozole (LET) in patients (pts) with hormone receptor-positive (HR+), human epidermal growth factor receptor-2–negative (HER2–;) advanced breast cancer (ABC) and central nervous system (CNS) metastases: Subgroup analysis from the phase IIIb CompLEEment-1 trial. Ann. Oncol. 2019, 30, v118. [Google Scholar] [CrossRef]
  110. Yardley, D.A. MONALEESA clinical program: A review of ribociclib use in different clinical settings. Future Oncol. 2019, 15, 2673–2686. [Google Scholar] [CrossRef] [Green Version]
  111. Mehta, M.P.; Wang, D.; Wang, F.; Kleinberg, L.; Brade, A.; Robins, H.I.; Turaka, A.; Leahy, T.; Medina, D.; Xiong, H.; et al. Veliparib in combination with whole brain radiation therapy in patients with brain metastases: Results of a phase 1 study. J. Neurooncol. 2015, 122, 409–417. [Google Scholar] [CrossRef]
  112. Litton, J.K.; Rugo, H.S.; Ettl, J.; Hurvitz, S.A.; Goncalves, A.; Lee, K.H.; Fehrenbacher, L.; Yerushalmi, R.; Mina, L.A.; Martin, M.; et al. Talazoparib in Patients with Advanced Breast Cancer and a Germline BRCA Mutation. N. Engl. J. Med. 2018, 379, 753–763. [Google Scholar] [CrossRef]
  113. Robson, M.E.; Tung, N.; Conte, P.; Im, S.A.; Senkus, E.; Xu, B.; Masuda, N.; Delaloge, S.; Li, W.; Armstong, A.; 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, 558–566. [Google Scholar] [CrossRef]
  114. Diéras, V.; Han, H.S.; Kaufman, B.; Wildiers, H.; Friedlander, M.; Ayoub, J.-P.; Puhalla, S.L.; Bondarenko, I.; Campone, M.; Jakobsen, E.H.; et al. Veliparib with carboplatin and paclitaxel in BRCA-mutated advanced breast cancer (BROCADE3): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2020, 21, 1269–1282. [Google Scholar] [CrossRef]
  115. Sharma, P.; Rodler, E.; Barlow, W.E.; Gralow, J.; Huggins-Puhalla, S.L.; Anders, C.K.; Goldstein, L.J.; Brown-Glaberman, U.A.; Huynh, T.T.; Szyarto, C.S.; et al. Results of a phase II randomized trial of cisplatin +/− veliparib in metastatic triple-negative breast cancer (TNBC) and/or germline BRCA-associated breast cancer (SWOG S1416). J. Clin. Oncol. 2020, 38 (Suppl. 15), 1001. [Google Scholar] [CrossRef]
  116. Bardia, A.; Hurvitz, S.A.; Tolaney, S.M.; Loirat, D.; Punie, K.; Oliveira, M.; Brufsky, A.; Sardesi, S.D.; Kalinsky, K.; Zelnak, A.B.; et al. Sacituzumab Govitecan in Metastatic Triple-Negative Breast Cancer. N. Engl. J. Med. 2021, 384, 1529–1541. [Google Scholar] [CrossRef] [PubMed]
  117. Rugo, H.S.; Bardia, A.; Marmé, F.; Cortes, J.; Schmid, P.; Loirat, D.; Trendan, O.; Ciruelos, E.; Dalenc, F.; Gomez-Pardo, P.; 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 (Suppl. 17), LBA1001. [Google Scholar] [CrossRef]
  118. Cortés, J.; Rugo, H.S.; Awada, A.; Twelves, C.; Perez, E.A.; Im, S.A.; Gomez-Pardo, P.; Schwartzberg, L.S.; Diéras, V.; Yardley, D.; et al. Prolonged survival in patients with breast cancer and a history of brain metastases: Results of a preplanned subgroup analysis from the randomized phase III BEACON trial. Breast Cancer Res. Treat. 2017, 165, 329–341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Duchnowska, R.; Pęksa, R.; Radecka, B.; Mandat, T.; Trojanowski, T.; Jarosz, B.; Czartoryska-Arłukowicz, B.; Olszewski, W.P.; Och, W.; Kalinka-Warzocha, E.; et al. Immune response in breast cancer brain metastases and their microenvironment: The role of the PD-1/PD-L axis. Breast Cancer Res. 2016, 18, 43. [Google Scholar] [CrossRef] [Green Version]
  120. Nanda, R.; Chow, L.Q.; Dees, E.C.; Berger, R.; Gupta, S.; Geva, R.; Pusztai, L.; Pathiraja, K.; Aktan, G.; Cheng, J.D.; et al. Pembrolizumab in Patients With Advanced Triple-Negative Breast Cancer: Phase Ib KEYNOTE-012 Study. J. Clin. Oncol. 2016, 34, 2460–2467. [Google Scholar] [CrossRef]
  121. Cortes, J.; Rugo, H.S.; Cescon, D.W.; Im, S.A.; Yusof, M.M.; Gallardo, C.; Lipatov, O.; Barrios, C.H.; Perez-Garcia, J.; Iwata, H.; et al. Pembrolizumab plus Chemotherapy in Advanced Triple-Negative Breast Cancer. N. Engl. J. Med. 2022, 387, 217–226. [Google Scholar] [CrossRef]
  122. Schmid, P.; Adams, S.; Rugo, H.S.; Schneeweiss, A.; Barrios, C.H.; Iwata, H.; Diéras, V.; Hegg, R.; Im, S.A.; Wright, G.S.; et al. Atezolizumab and Nab-Paclitaxel in Advanced Triple-Negative Breast Cancer. N. Engl. J. Med. 2018, 379, 2108–2121. [Google Scholar] [CrossRef]
  123. Nieblas-Bedolla, E.; Nayyar, N.; Singh, M.; Sullivan, R.J.; Brastianos, P.K. Emerging Immunotherapies in the Treatment of Brain Metastases. Oncologist 2021, 26, 231–241. [Google Scholar] [CrossRef]
  124. Kumthekar, P.; Tang, S.-C.; Brenner, A.J.; Kesari, S.; Piccioni, D.E.; Anders, C.K.; Carrillo, J.A.; Chalasani, P.; Kabos, P.; Puhalla, S.; et al. ANG1005, a novel brain-penetrant taxane derivative, for the treatment of recurrent brain metastases and leptomeningeal carcinomatosis from breast cancer. J. Clin. Oncol. 2016, 34 (Suppl. 15), 2004. [Google Scholar] [CrossRef]
  125. Kumthekar, P.; Tang, S.C.; Brenner, A.J.; Kesari, S.; Piccioni, D.E.; Anders, C.; Carrillo, J.; Chalasani, P.; Kabos, P.; Puhalla, S.; et al. ANG1005, a Brain-Penetrating Peptide-Drug Conjugate, Shows Activity in Patients with Breast Cancer with Leptomeningeal Carcinomatosis and Recurrent Brain Metastases. Clin. Cancer Res. 2020, 26, 2789–2799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Li, F.; Tang, S.C. Targeting metastatic breast cancer with ANG1005, a novel peptide-paclitaxel conjugate that crosses the blood-brain-barrier (BBB). Genes Dis. 2017, 4, 1–3. [Google Scholar] [CrossRef]
  127. Leone, J.P.; Leone, B.A. Breast cancer brain metastases: The last frontier. Exp. Hematol. Oncol. 2015, 4, 33. [Google Scholar] [CrossRef] [Green Version]
  128. Vogelbaum, M.A.; Brown, P.D.; Messersmith, H.; Brastianos, P.K.; Burri, S.; Cahill, D.; Dunn, I.F.; Gaspar, L.E.; Gaston, N.T.N.; Gondi, V.; et al. Treatment for Brain Metastases: ASCO-SNO-ASTRO Guideline. J. Clin. Oncol. 2022, 40, 492–516. [Google Scholar] [CrossRef] [PubMed]
  129. Lin, N.U.; Bellon, J.R.; Winer, E.P. CNS metastases in breast cancer. J. Clin. Oncol. 2004, 22, 3608–3617. [Google Scholar] [CrossRef] [PubMed]
  130. Rivera, E.; Meyers, C.; Groves, M.; Valero, V.; Francis, D.; Arun, B.; Broglio, K.; Yin, G.; Hortobagyi, G.N.; Buchholz, T. Phase I study of capecitabine in combination with temozolomide in the treatment of patients with brain metastases from breast carcinoma. Cancer 2006, 107, 1348–1354. [Google Scholar] [CrossRef] [PubMed]
  131. Stewart, D.J.; Dahrouge, S. Response of brain metastases from breast cancer to megestrol acetate: A case report. J. Neurooncol. 1995, 24, 299–301. [Google Scholar] [CrossRef]
  132. Salvati, M.; Cervoni, L.; Innocenzi, G.; Bardella, L. Prolonged stabilization of multiple and single brain metastases from breast cancer with tamoxifen. Report of three cases. Tumori 1993, 79, 359–362. [Google Scholar] [CrossRef]
  133. Madhup, R.; Kirti, S.; Bhatt, M.L.; Srivastava, P.K.; Srivastava, M.; Kumar, S. Letrozole for brain and scalp metastases from breast cancer--a case report. Breast 2006, 15, 440–442. [Google Scholar] [CrossRef]
  134. Bartsch, R.; Berghoff, A.S.; Furtner, J.; Bergen, E.S.; Roider-Schur, S.; Marhold, M.; Starzer, A.M.; Forstner, H.; Rottenmanner, B.; Dieckmann, K.S.; et al. 280P Intracranial activity of trastuzumab-deruxtecan (T-DXd) in HER2-positive breast cancer patients with active brain metastases: Results from the first stage of the phase II TUXEDO-1 trial. Ann. Oncol. 2021, 32, S486. [Google Scholar] [CrossRef]
Figure 1. Suggested treatment algorithm for patients with breast cancer brain metastases. Abbreviations: BCBM, breast cancer brain metastases; KPS, Karnofsky Performance Scale; RT, radiotherapy; SRS, stereotactic radiosurgery; WBRT, whole-brain radiation therapy; HER-2, human epidermal growth factor receptor 2; T-DXd, trastuzumab-deruxtecan; T-DM1, trastuzumab-emtansine; HR, hormone receptor.
Figure 1. Suggested treatment algorithm for patients with breast cancer brain metastases. Abbreviations: BCBM, breast cancer brain metastases; KPS, Karnofsky Performance Scale; RT, radiotherapy; SRS, stereotactic radiosurgery; WBRT, whole-brain radiation therapy; HER-2, human epidermal growth factor receptor 2; T-DXd, trastuzumab-deruxtecan; T-DM1, trastuzumab-emtansine; HR, hormone receptor.
Ijms 23 12525 g001
Table 1. Major clinical trials of local treatments for BCBM.
Table 1. Major clinical trials of local treatments for BCBM.
TreatmentAuthorClinical Trial NumberStudy PopulationPhasePrimary Outcome
Surgery plus WBRT vs. WBRT alonePatchell et al. Single BMIIIOS
Surgery plus WBRT vs. surgery alonePatchell et al. Single BMIIIRecurrence of tumor in the brain
Preoperative SRS vs. postoperative SRS(Ongoing)NCT03741673BM under 4 cm for single fraction and over 7 cm for multifractional therapyIIILMD-free rate
WBRT with memantine vs. WBRT without memantineBrown et al.NCT0056685Pathologically proven BMIIICognitive function
Preoperative SRS(Ongoing)NCT033686251–6 BM (2–4 cm)IIRadiation toxicity
Postoperative SRS vs. WBRTBrown et al.NCT01372774Single BMIIICognitive-deterioration-free survival and OS
Salvage SRS vs. postoperative WBRTKayama et al.JCOG05041–4 resected BM with only one lesion > 3 cmIIIOS
WBRT alone vs. WBRT followed by SRSAndrews et al.NCT000027081–3 BMIIIOS
SRS alone vs. SRS plus WBRTBrown et al.NCT003771561–3 BMIIICognitive deterioration at 3 months
HA-WBRT + memantine vs. WBRT + memantineBrown et al.NTG Oncology CC001 (NCT02360215)BM outside a 5 mm margin around either hippocampusIIITime to cognitive function failure
SRS vs. HA-WBRT + memantine(Ongoing)NCT035503915–15 BMIIIOS and neurocognitive PFS
SRS or surgery with/without WBRTKocher et al.NCT000028991–3 BMIIITime to PS deterioration more than 2
SRS plus WBRT vs. SRS aloneAoyama et al.C000004121–4 BM, each under 3 cmIIIOS
SRS for 2–4 BM vs. 5–10 BMYamamoto et al.UMIN000001812Patients with BM who received SRSIIIOS
SRS vs. WBRTLie et al.NCT015929684–15 untreated nonmelanoma BMIIILocal control rate and proportion of patients with neurocognitive decline at 4 months
SRS vs. HA-WBRT(Ongoing)NCT03075075–20 BMIIIQoL
HA-WBRTGondi et al.NCT01227954BM outside a 5 mm margin around either hippocampusIICognitive function
FSRT(Ongoing)NCT04061401–10 HER2-positive BCBMIIIntracranial local tumor control rate
Abbreviations: BC, breast cancer; MBC, metastatic breast cancer; BM, brain metastases; BCBM, breast cancer brain metastases; WBRT, whole-brain radiation therapy; SRS, stereotactic radiosurgery; HA-WBRT, hippocampal-avoidance whole-brain radiotherapy; OS, overall survival; PFS, progress-free survival; QoL, quality of life; LMD, leptomeningeal disease; FRST, fractionated stereotactic radiotherapy.
Table 2. Major clinical trials of systemic treatments for BCBM.
Table 2. Major clinical trials of systemic treatments for BCBM.
TreatmentAuthorClinical Trial Name (Number)Study PopulationPhasePrimary Outcome
Intra-arterial cerebral infusion of trastuzumab(Ongoing)NCT02571530HER2-positive BCBMIMTD, adverse events, and dose-limiting toxicities
LapatinibLin et al.EGF 105084 (NCT00263588)Progressive HER2-positive BCBM after prior trastuzumab and cranial RTIIORR in CNS
Lapatinib + capecitabine vs. lapatinib + topotecanLin et al.NCT00437073HER2-positive BC with progressive BCBM after trastuzumab and cranial RTIIICNS objective response
Lapatinib + capecitabine vs. trastuzumab + capecitabinePivot et al.CEREBEL (NCT00820222)HER2-positive MBC without BMIIIIncidence of CNS metastases as first site of relapse
Intermittent high-dose lapatinib + capecitabineMorikawa et al.(NCT02650752)HER2-positive BCBMIMTD
Lapatinib + capecitabineBachelot et al.LANDSCAPE (NCT00967031)HER2-positive BCBM not previously treated with WBRT, capecitabine, or lapatinibIIORR in CNS
Lapatinib + capecitabine vs. capecitabine aloneGeyer et al.EGF100151 (NCT00078572)ABC with progression on trastuzumabIIITime to progression
Lapatinib + everolimus + capecitabineHurvitz et al.TRIO-US B-09 (NCT01783756)HER2-positive BCBMI/IICNS ORR
Lapatinib + WBRTLin et al.NCT00470847HER2-positive BCBMIMaximum tolerated dose of concurrent lapatinib with WBRT
Lapatinib + WBRTChristodoulou et al.NCT01218529BM from HER2-positive BC or lung cancerIIResponse rate in brain as assessed by volumetric analysis in brain MRI
Lapatinib + WBRT/SRS vs. WBRT/SRSKim et al.NCT01622868HER2-positive BCBMIICR rate in measurable BM
Pertuzumab + high-dose trastuzumab(Ongoing)PATRICIA (NCT02536339)HER2-positive progressive BCBM after RTIIPercentage of participants with ORR in the CNS
Pertuzumab + trastuzumab and docetaxel vs. trastuzumab + docetaxelSwain et al.CLEOPATRA (NCT00597190)HER2-positive locally recurrent, unresectable, or MBC without prior chemotherapy or biologic therapy for their advanced diseaseIIIPFS
T-DM1Montemurro et al.KAMILLA (NCT01702571)HER2-positive locally advanced or MBC with prior HER2-targeted therapy and chemotherapyIIIBest overall response rate and clinical benefit rate
T-DM1 vs. lapatinibKrop et al.EMILIA (NCT00829166)HER2-positive ABC previously treated with prior HER2-targeted therapy and chemotherapyIIIPercentage of participants with progressive disease or death, PFS, and OS
T-DM1 alone vs. T-DM1 plus metronomic temozolomide(Ongoing)NCT03190967HER2-positive BCBM treated with SRSI/IIMTD of temozolomide when used with T-DM1
NeratinibChan et al.ExteNET (NCT00878709)Stage II and IIIC HER2-positive BC with node-positive diseaseIIIInvasive disease-free survival at year 2
Neratinib + paclitaxel vs. trastuzumab + paclitaxelAwada et al.NEfERT-T (NCT00915018)Previously untreated recurrent and/or metastatic HER2-positive BCIIIPFS
Neratinib + capecitabineFreedman et al.TBCRC 022 (NCT01494662)Measurable, progressive, HER2-positive BCBMIIORR
Neratinib + capecitabine vs. lapatinib + capecitabineSaura et al.NALA trial (NCT01808573)HER2-positive MBC with two or more previous HER2-directed MBC treatmentsIIIPFS and OS
Afatinib vs. Afatinib + 2Gy RT vs. afatinib + 4Gy RT(Ongoing)NCT02768337BM from BC or lung cancerI/IIRatio of afatinib concentration in resected BM/plasma
Afatinib alone vs. afatinib + vinorelbine vs. investigator’s choiceCortes et al.LUX-Breast 3 (NCT01441596)HER2-positive BCBM with recurrence or progression during or after trastuzumab and lapatinibIIPatient benefit at 12 weeks
TucatinibMurthy et al.HER2CLIMB (NCT02614794)HER2-positive MBC previously treated with trastuzumab, pertuzumab, and Trastuzumab emtansineIIPFS
Tucatinib + T-DM1Borges et al.NCT01983501HER2-positive MBCIIncidence of adverse effects
Tucatinib + T-DM1 vs. T-DM1Hurvitz et al.HER2CLIMB-02 (NCT03975647)HER2-positive MBC previously treated with a taxane and/or trastuzumabIIIPFS
Pyrotinib + vinorelbine(Ongoing)NCT03933982HER2-positive BCBMIIORR of CNS
Pyrotinib + capecitabineYan et al.PERMEATE (NCT03691051)HER2-positive BCBMIIORR of CNS
GRN1005 (paclitaxel trevatide) vs. HRN1005 + trastuzumabBates et al.GRABM-BHER2-positive BCBMIIORR in CNS
GDC-0084 + trastuzumab(Ongoing)NCT03765983HER2-positive BCBMIIORR in CNS
Trastuzumab DeruxtecanJerusalem et al.DESTINY-Breast01 (NCT03248492)HER2-positive MBC that had received previous treatment with T-DM1IIORR
Trastuzumab Deruxtecan vs. Trastuzumab emtansineCortés et al.DESTINY-Breast03HER2-positive unresectable and/or metastatic breast cancer previously treated with Trastuzumab and TaxaneIIIPFS
Trastuzumab Deruxtecan vs. physician’s choice of treatmentModi et al.DESTINY-Breast04Previously treated HER2-low advanced breast cancerIIIPFS
Etirinotecan pegol vs. physician’s treatment of choiceCortes et al.BEACON (NCT01492101)Primary brain tumor or BMIIIOS
Erinotecan pegol vs. phisician’s treatment of choiceTripathy et al.ATTAINStable BCBM previously treated with an anthracycline, a taxane, and capecitabineIIIOS
Everolimus + vinorelbine + trastuzumab Van Swearingeng et al.NCT01305941HER2-positive BCBMIIORR in CNS
Alpelisib + capecitabine vs. buparlisib + capecitabine vs. buparlisib + capecitabine + trastuzumab vs. buparlisib + capecitabine + lapatinibMcRee et al.NCT01300962MBC, including BCBMIMTD and DLT
Buparlisib + fulvestrant vs. fulvestrant aloneBasega et al.BELLE-2 (NCT01610284)HR-positive/HER2-negative locally advanced or MBS with postmenopausal statusIIIPFS
Buparlisib + fulvestrant vs. fulvestrant aloneDi Leo et al.BELLE-3 (NCT01633060)HER2-negative, locally advanced, or MBC that had relapsed on or after endocrine therapy and mTOR inhibitorsIIIPFS
Buparlisib + capecitabine (+ trastuzumab in HER2-positive group)CompletedNCT02000882BCBMIICBR
Palbociclib(Ongoing)NCT02896335Measurable progressive luminal-type BCBMIIClinical benefit rate at 8 weeks
AbemaciclibTolaney et al.NCT02308020BM from luminal-type BC, NSCLC, or melanomaIIORR in CNS
Abemaciclib or PI3K inhibitor GDC-0084 or entrectinib, selected by genetic test(Ongoing)NCT03994796New or progressive BMIIORR in CNS
Palbociclib + trastuzumab + lapatinib + fulvestrant(Ongoing)NCT04334330ER-positive/HER2-positive BCBMIIORR
Ribociclib + LetrozoleHortobagyi et al.MONALEESA-2 (NCT01958021)Postmenopausal women with HR+, HER2-negative ABCIIIPFS
Ribociclib + Endocrine therapy vs. placebo + endocrine therapyTripathy et al.MONALEESA-7 (NCT02278120)Advanced HR+, HER2-negative BCIIIOS
Ribociclib + buparlisib + fulvestrant vs. ribociclib + alpelisib + fulvestrant vs. ribociclib + fulvestrantTolaney et al.NCT02088684Postmenopausal women with HR-positive/HER2-negative locally recurrent or advanced MBSI/IIIncidence of DLTs and PFS
Ribociclib + letrozoleDe Laurentis et al.ComPLEEment-1Advanced HR+, HER2- BCIIIbSafety, tolerability, and efficacy
PembrolizumabNanda et al.KEYNOTE-012 (NCT01848834)Advanced TNBC, advanced head and neck cancer, advanced urothelial cancer, or advanced gastric cancerIAdverse events and overall response rate
Pembrolizumab + SRS(Ongoing)NCT03449238BCBMI/IITumor response at 8 weeks
Pembrolizumab vs. chemotherapyWiner et al.KEYNOTE-119Metastatic TNBCIIIOS
Pembrolizumab + paclitaxel vs. pembrolizumab + nab-paclitaxel vs. pembrolizumab +gemcitabine/carboplatin vs. pembrolizumab + chemotherapy vs. chemotherapyCortes et al.KEYNOTE-355Previously untreated locally recurrent inoperable BC or metastatic TNBCIIIAdverse events and PFS
Atezolizumab + nab-paclitaxel vs. nab-paclitaxelSchmid et al.IMpassion130 (NCT01004172)Unresectable locally advanced or metastatic TNBCIIIPFS and OS
Atezolizumab + paclitaxel vs. Paclitaxel aloneMiles et al.IMpassion131 (NCT03125902)Previously untreated locally advanced or metastatic TNBCIIIPFS
Atezolizumab + SRSActive, not recruitingNCT03483012TN-type BCBMIIPFS
Atezolizumab + chemotherapy vs. chemotherapyKyte et al.ALICE (NCT03164993)Locally advanced or metastatic TNBCIIToxicity and PFS
Carboplatin and bevacizumabLeone et al.NCT01004172New or progressive BCBMIIORR in CNS
Bevacizumab, etoposide, and cisplatinWu et al.NCT01281696BC brain and or leptomeningeal metastasesIIORR in CNS
Nivolumab + SRSAhmed et al.NCT03807762BCBMIINumber of participants who experience DLTs
Talazoparib vs. single-agent chemotherapy of investigator’s choiceLitton et al.EMBRACA (NCT01945775)Advanced and/or MBC patients with BRCA mutation who received no more than three prior chemotherapy-inclusive regimens for locally advanced and/or metastatic diseaseIIIPFS
Olaparib vs. single-agent chemotherapy of investigator’s choiceRobson et al.OlympiAD (NCT02000622)MBC patients who had received no more than two previous chemotherapy regimens for metastatic diseaseIIIPFS
Veliparib + carboplatin + paclitaxel vs. Carboplatin + paclitaxelDieras et al.BROCADE3 (NCT02163694)Advanced HER2-negative BC with BRCA1 or BRCA2 mutationIIIPFS
Cisplatin + veliparib vs. Cisplatin aloneSharma et al.SOWG S1416 (NCT02595905)Recurrent or metastatic TNBC, including BMIIPFS
RucaparibPatsouris et al.RUBY (NCT0250548)HER2-negative MBC with BRCAnessIICBR
Niraparib + pembrolizumabVinayak et al.TOPACIO/KEYNOTE-162 (NCT02657889)Advanced or metastatic TNBC or recurrent ovarian cancerI/IINumber of subjects reporting DLTs and ORR
Sorafenib + WBRTMorikawa et al.NCT01724606BCBMIMTD and toxicity by number of adverse events
HER2-CAR T cellsRecruitingNCT03696030HER2-positive recurrent BCBM or leptomeningeal metastasesIIncidence of DLTs and number of adverse events
HER2-CAR T cells(Ongoing)NCT02442297HER2-positive CNS tumorsIORR in CNS
Abbreviations: BC, breast cancer; MBC, metastatic breast cancer; BM, brain metastases; BCBM, breast cancer brain metastases; TN, triple-negative; TNB, triple-negative breast cancer; ABC, advanced breast cancer; HR, hormone receptor; RT, radiotherapy; WBRT, whole-brain radiotherapy; SRS, stereotactic radiosurgery; HER2, human epidermal growth factor receptor 2; OS, overall survival; CAR, chimeric antigen receptors; CBR, clinical benefit rate; CR, clinical response; PFS, progression-free survival; ORR, objective response rate; PS, performance status; CNS, central nervous system; T-DM1, trastuzumab emtansine; NSCLS, non-small-cell lung cancer; BRCA, breast-cancer gene; MRI, magnetic resonance imaging; MTD, maximum tolerated dose; DLTs, dose-limiting toxicities.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Avila, J.; Leone, J.P. Advances in the Management of Central Nervous System Metastases from Breast Cancer. Int. J. Mol. Sci. 2022, 23, 12525. https://doi.org/10.3390/ijms232012525

AMA Style

Avila J, Leone JP. Advances in the Management of Central Nervous System Metastases from Breast Cancer. International Journal of Molecular Sciences. 2022; 23(20):12525. https://doi.org/10.3390/ijms232012525

Chicago/Turabian Style

Avila, Jorge, and José Pablo Leone. 2022. "Advances in the Management of Central Nervous System Metastases from Breast Cancer" International Journal of Molecular Sciences 23, no. 20: 12525. https://doi.org/10.3390/ijms232012525

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