Content area
Opinion Statement
Breast cancer metastasizing to the central nervous system (CNS) encompasses two distinct entities: brain metastases involving the cerebral parenchyma and infiltration of the leptomeningeal space, i.e., leptomeningeal disease. CNS metastases affect 10–15% of patients with hormone receptor-positive-status and nearly one-half of those with HER2-positive and triple-negative breast cancer with distant metastatic disease. Significant clinical morbidity and heterogeneous penetration of the blood–brain barrier by systemic therapies contribute to the poor prognosis associated with brain metastases. Recent advances in radiotherapy, including stereotactic approaches and morbidity-reducing strategies such as the use of memantine and hippocampal avoidance in whole brain radiation, coupled with the development of more effective CNS-penetrant systemic therapies, including small molecules and antibody–drug conjugates, have significantly improved patient outcomes. Consequently, patients with breast cancer CNS metastases have improved survival compared to prior decades, and longitudinal care has become increasingly complex, necessitating a multidisciplinary approach to achieve optimal outcomes for patients.
Introduction
Although often underestimated, 5–10% of breast cancer patients and 40% of patients with metastatic disease develop central nervous system (CNS) metastases, which include two distinct entities: cerebral parenchymal involvement (brain metastases, BM) and infiltration of the cerebrospinal fluid-filled leptomeningeal space (leptomeningeal disease, LMD) [1, 2, 3–4]. CNS involvement remains a major clinical challenge, in part because of the protective roles of the blood–brain and blood-CSF barriers, and because CNS disease frequently occurs in the setting of heavily pretreated, treatment-refractory cancer. Additionally, a unique microenvironment consisting of neurons, astrocytes, microglia, and oligodendrocytes, coupled with metabolic constraints that are exclusive to this area, imposes selective pressure on tumor cells [3, 4]. Consequently, metastases to the CNS represent a distinct entity with a unique evolutionary trajectory compared to extracranial metastases [5]. The combination of a distinct phenotype and heterogeneous penetration of the blood–brain barrier by systemic therapies could explain the frequent observation of progressive BM, even in patients with controlled extracranial disease. Consequently, cerebral involvement leads to a poor prognosis, resulting in neurological death in 20–50% of patients [6, 7]. Given that the risk of developing CNS metastases increases with the duration of metastatic disease and by line of therapy [8, 9] and that patients live longer with better control of extracranial disease through new systemic therapies, the incidence of CNS metastases is expected to rise. Furthermore, novel therapies that have improved survival and reduced extracranial relapse in early-stage breast cancer, such as pembrolizumab in early-stage triple-negative breast cancer (TNBC) and trastuzumab-emtansine in early-stage HER2-positive breast cancer with residual disease, have not reduced rates of CNS relapse [10, 11]. This trend highlights an urgent unmet need for patients with CNS metastases.
Metastatic breast cancer (MBC) is the second most common cause of BM and LMD, indicating a pronounced tropism for these sanctuary spaces, more pronounced for HER2-positive and triple-negative breast cancers (TNBC) than luminal subtypes [3]. Overall survival has improved for some BM subtypes, particularly those with HER2-positive BM [12]. This improvement is mainly attributed to the development of systemic therapies that are effective in the CNS and extracranially. The movement in the last decade to include patients with BM in registrational clinical trials has accelerated progress. Local therapy advances such as stereotactic radiosurgery have improved morbidity but not overall survival in most populations. The management of patients with CNS metastases has thus become complex, requiring a multidisciplinary approach to achieve optimal outcomes.
Epidemiology
Unfortunately, the exact incidence of BM in breast cancer remains unknown, as current registries do not reliably capture brain metastases after initial diagnosis of malignancy and because mandated central reporting of brain metastases is not currently standard in the United States, along with the fact that screening for BM is not routinely performed for many primary cancers, underreporting of new metastatic sites post-diagnosis, and potential underdiagnosis in patients with minimal symptoms. Ongoing screening programs are expected to provide more precise data on the incidence of BM soon (NCT04030507).
The main factors associated with the risk of CNS metastases are initial stage, metastatic disease-free interval, overall burden of metastatic disease, HER2-positive subtype, triple-negative subtype, young age, and BRCA2 germinal status [9, 13, 14–15]. A lobular histological type is a specific risk factor of LMD [16].
Population-based data from the SEER (Surveillance, Epidemiology, and End Results) registry in the United States indicates that 0.4% of patients, including early-stage patients, had BM at the initial breast cancer diagnosis. However, 7.6% of patients with de novo MBC had BM at diagnosis, including 11.5% of patients with Hormone Receptor-negative and HER2-positive (HR-/HER2 +) de novo MBC, 11.4% with de novo TNBC, and 5.56% with HR-positive/HER2-negative (HR + /HER2-) MBC [13]. In the metastatic setting within a large French national cohort (ESME), the cumulative incidence rates of CNS metastases at 24 months were 14.4%, 29.2%, 49.0%, and 44.8% for patients with HER2-/HR + , HER2 + /HR + , HER2 + /HR-, and TNBC, respectively. The incidence continued rising across all tumor subtypes with no evidence of plateauing. In this cohort, among patients with CNS metastases at MBC diagnosis (de novo), 43.1% had only CNS involvement (isolated CNS) [9]. An analysis based on the metastatic treatment line from the Flatiron cohort revealed that at metastatic diagnosis, the prevalence of BM was 6.3%, 11.2%; 2.7% and 11.1% HER2-/HR + , HER2 + /HR + , HER2 + /HR-, and TNBC, respectively. Then in the second/fourth line, the prevalence of BM was 4.4%/7.2%, 17.6%/26.1%, 31.2%/37.1%, and 17.6%/24.7% among patients with HER2-/HR + , HER2 + /HR + , HER2 + /HR-, and TNBC, respectively [8]. To note, the HER2-low expression did not impact BM prevalence within historical subtypes.
It is important to highlight the biomarker (i.e. Her2 and estrogen expression) discordance is often observed between primary tumors and their corresponding brain metastases. This discordance is characterized by a loss of HR alongside a gain of HER2 expression, resulting in nearly three-quarters of all CNS metastases expressing HER2 (positive or low) [17, 18–19].
LMD is less common and may affect 5–20% of MBC patients [20, 21–22]. LMD typically emerges as a late complication in MBC, often coinciding with systemic disease progression. The subtype distribution mirrors that of BM, with an overrepresentation of HER2-positive and TNBC subtypes: 18–27% for HER2-positive, 44–51% for HR-positive/HER2-negative, and 15–37% for TNBC, respectively. Risk factors for developing LMD in breast cancer patients include lobular histology, HER2-positive, TNBC subtype, existing BM (particularly those near the meningeal space or infratentorial), and BM resection [23, 24, 25, 26–27].
Prognostic factors among MBC Patients with CNS Metastases
CNS involvement is associated with inferior prognosis across all subtypes of MBC compared to those without CNS dissemination. Among patients with CNS metastases, the key factors independently associated with survival include performance status and tumor subtype, with HR + /HER2 + having a better prognosis than HR-/HER2 + , followed by HR + /HER2- and then TNBC [9, 28, 29]. Other significant prognostic factors include the patient's age, the number of BM, and the presence of extracranial metastases. These prognostic factors have been integrated into a Graded Prognostic Assessment (GPA) score [29, 30]. The median overall survival (OS) from the diagnosis of BM ranged from 6 months for those in the GPA 0 group to 36 months for patients in the GPA 3.5–4.0 group. The status of extracranial disease (stable or progressive) was also a major prognostic factor, with a doubling of OS observed in patients with stable extracranial disease (17.9 versus 8.0 months), as demonstrated in a meta-analysis encompassing all cancer types [31]. Finally, the Brain Metastasis Velocity (BMV) is a prognostic measure recently defined for patients experiencing cerebral recurrence after initial stereotactic radiosurgery (SRS) for BM, calculated by dividing the number of new BM that develop post-SRS by the time interval (in years) since the initial SRS. BMV has been identified as an independent prognostic factor in a large, multi-institutional, pan-tumor cohort. Median OS from the initial SRS for BMV categories as low-risk (< 4 brain metastases), intermediate-risk (4–13 metastases), and high-risk (> 13 new metastases per year) were 12.5 months, 7.0 months, and 4.6 months, respectively [32]. This finding was also corroborated in a separate cohort of patients with MBC, with a hazard ratio (HR) of 1.03 for each increment in BMV, regardless of the subtype [33].
Brain metastases prognosis can vary widely, and there is often a challenge with the competing risk of extracranial visceral metastases. With many systemic therapy options across breast cancer subtypes, understanding which patients are at highest risk of death from their brain metastases is of interest. One would prioritize treatment regimens with the highest intracranial penetration and CNS progression-free survival (CNS-PFS) in those with a presumed high risk of dying from their BM. Patients with HER2 + BM have a higher risk of neurological death compared to TNBC or HR + subtypes. Number of brain metastases, upfront whole brain radiotherapy, and progressive extracranial disease were also multivariate risk factors of neurologic death [7].
Patients with LMD have the direst prognosis, with OS ranging from 3 to 10.5 months. A similar trend to that seen in patients with brain metastases is observed according to subtype, with patients with HER2-positive disease having longer survival (4.4–20.0 months) compared to those with HR-positive (3.7–6.0 months) and TNBC (2.0–3.0 months) [16].
Treatment of Brain Metastases among MBC Patients
Clinical Categories of Brain Metastases
BM treatment aims to prevent or delay neurological deterioration and prolong survival with an acceptable quality of life [2, 34, 35]. However, BM presentations are heterogeneous and have distinct clinical scenarios with specific therapeutic implications. BM are usually described by their clinical stability and treatment history as either 1) treated/stable or 2) active. This has risen in relevancy due to the inclusion of stable BM patients in most trials but the exclusion of active BM patients. Treated/stable BM describes lesions that have previously received CNS-specific local treatment with subsequent validation of stable or non-progressive disease on imaging. Active BM describes either new BM that have not been treated with CNS-directed therapy or progressive BM despite local CNS-directed therapy [36].
The tumor burden, defined by the number and size of lesions, varies widely. Concurrent extracranial disease status, with isolated cerebral progression or multisite progressions, also adds to the complexity of treating BM patients. Finally, treatment may differ depending on the presence or absence of neurological symptoms [2, 34, 35]. Local CNS-directed therapies should be prioritized urgently in patients with symptoms. Patients with symptomatic BM historically have inferior OS compared to those with asymptomatic disease [37]. Treatment objectives and systemic therapy choice will differ based on stable/active status, symptomatic versus asymptomatic status, extracranial disease status, and number of brain metastases. For patients with stable BM without high-risk features of neurologic death, standard systemic therapy algorithms for extracranial disease apply after local therapy. For patients with active BM with progressive disease despite prior radiation, prioritizing regimens with the highest intracranial response rates is necessary. Algorithm treatment for BM breast cancer is shown in Fig. 1.
[See PDF for image]
Fig. 1
Algorithm treatment brain metastases breast cancer. BM, Brain metastases, CNS Central Nervous System.
Local Treatments
Limited Number of Brain Metastases
Surgery followed by cavity radiation is generally considered for a single metastasis and an option for patients with a limited number of BM, particularly in those with bulky, symptomatic lesions or diagnostic uncertainty when observation is less viable. Two prospective trials demonstrated a survival benefit of surgery followed by whole brain radiotherapy (WBRT) compared with WBRT alone in patients with CNS metastases from solid tumors, with a median OS of 10–9 months versus 3.5–6 months. Notably, no OS benefit was observed in the subgroup with active extracranial disease, and one prospective study showed no significant difference in patients with impaired performance status [38, 39–40]. In the randomized NCCTG-N107C trial, stereotactic radiosurgery (SRS) was compared with WBRT for treatment of the surgical cavity following resection of a single brain metastasis. Although no significant difference in overall survival was observed (with a median of approximately 12 months), there was a reduction in the recurrence rate of the cavity at one year in the WBRT group (40% vs. 20%) and a reduction in the rate of cognitive function decline at six months (52% vs. 85%) with SRS [41]. To date, no prospective data exist on surgical treatment of more than one brain metastasis. Therefore, combining surgery and radiotherapy is recommended to treat patients with a limited number of bulky lesions in the context of modest extracranial disease. This approach is also beneficial for patients without a primary cancer diagnosis, for evaluating pathologic diagnosis or re-evaluating biomarkers (like HER2 status), and for large metastases with mass effect. Stereotactic radiation administered to the postoperative cavity is often preferred to WBRT because of the better preservation of cognitive function. Note that SRS is associated with an increased risk of pachymeningeal seeding beyond the radiation field (approximately 8% although higher in some series) [42].
Extensive Brain Metastases
Radiation therapy, particularly SRS, is fundamental in treating patients with a limited number of metastases. The choice of cut-off between stereotactic radiosurgery (SRS) versus WBRT is still debated, but it likely involves both the number and volume of brain metastases [34, 43] among other factors such as patient age, performance status, brain metastasis velocity, primary tumor type, extracranial disease status, and available systemic options. SRS improves disease control and potentially enhances survival in patients who have not undergone resection compared to those who receive no radiation. Importantly, SRS was less associated with cognitive deterioration compared to WBRT. In contrast, WBRT provides greater intracranial control but is also associated with more cognitive decline. Combining SRS and WBRT leads to further cognitive deterioration without improving survival outcomes and has little role in the contemporary management of patients [44, 45]. Nonetheless, WBRT remains a crucial treatment for multiple BM, especially in cases of a large intracranial disease burden. In such cases, memantine and hippocampal avoidance, when viable, should be used to reduce neurocognitive toxicity [46, 47]. SRS can lead to adverse radiation effects, most commonly radiation necrosis, which is defined as an inflammatory and necrotic process occurring in the brain. The incidence is about 8%, although the incidence varies widely, and can appear months or even years after exposure (median ∼7–8 months post-radiation). Diagnosing this condition and differentiating it from a recurrent tumor is a significant diagnostic challenge, and it causes neurological symptoms in half of the cases [48]. Its incidence appears to be increased when co-administered with an antibody–drug conjugate (ADC) [49]. Treatment options for radiation necrosis include corticosteroids, and for refractory cases, options extend to bevacizumab, laser interstitial thermal therapy, surgical resection, or hyperbaric oxygen therapy [48].
Systemic Treatment Options per Subtype for Brain Metastases
It has long been believed that systemic therapies' ineffectiveness on BM was due to their inability to penetrate the BBB. However, the BBB is often significantly disrupted by CNS metastases, making it permeable to agents that typically do not cross it, such as monoclonal antibodies [50]. In retrospective studies, systemic therapy following local, regional therapy to BM improves overall survival in luminal and HER2 + breast cancer, even in the setting of isolated brain progression [51]. The choice of the optimal systemic treatment after BM is highly variable and patient-specific, dependent upon performance status, breast cancer subtype, extracranial disease status, stable or active BM, and prior treatments. Data from prospective trials assessing the intracerebral activity of systemic treatments based on subtype are summarized in Table 1.
Table 1. Prospective clinical trial with outcomes on MBC patients with Brain metastasis
Trial name and References | Treatment regimen | Type of BM | Number of BM patients | Rate of Prior BM local therapy | CNS-ORR, % (95% CI) | ORR % (95% CI) | CNS-PFS Median month (mo), (95% CI) | PFS, Median mo, (95% CI) | OS Median mo, (95% CI) |
|---|---|---|---|---|---|---|---|---|---|
HER2 positive | |||||||||
Antibody Drug Conjugates and monoclonal antibodies anti HER2 | |||||||||
DEBBRAH [52] | T-Dxd | Cohort 1: non-progressing BM after local therapy | 8 | 87.5% | - | 80.0% (28.4–99.5) | - | 16-week PFS rate 87.5% (47.3–99.7) | - |
Cohort 2: asymptomatic untreated BM | 4 | 2:0% | 50.0% (6.7–93.2) | 50.0% (6.7–93.2) | - | - | - | ||
Cohort 3: progressing BM after local therapy | 9 | 100% | 44.4% (13.7–78.8) | 66.7% (29.9–92.5) | - | - | - | ||
Cohort 5: HER2 + or HER2-Low with LMD | 7 | 0% | 20% | 0% | - | 8.9 mo (2.1 – NR) | 13.3 mo (2.5 – NR) | ||
TUXEDO [53] | T-Dxd | Active | 15 | - | 73.3% (48.1–89.1%) | - | - | 21 (13.3-NR) | NR (22.2-NR) |
DESTINY-Breast01 [54] | T-Dxd | Stable | 24 | 75% | 47.1% | - | - | 18.1 mo (6.7–18.1) | - |
DESTINY-Breast03 [55] | T-Dxd | Stable | 43 | 53.5% | 65.7% | - | - | 15.0 mo (12.5–22.2) | - |
TDM1 | 39 | 51.3% | 34.3% | - | - | 3.0 mo (0.8–5.8) | - | ||
Pooled-Analysis DESTINY 01 02 03 [56] | T-Dxd | Stable and Active BM | 148 | 70.3% | Stable = 45.2% Active = 45.5% | - | Stable = 12.3 mo (11.1–13.8) Active = 18.5 mo (13.6–23.3) | - | - |
Trastuzumab/Lapatinib + Capecitabine or TDM1 | 83 | 69.9% | Stable = 27.6% Active = 12.0% | - | Stable = 8.7 mo (6.3–11.8) Active = 4.0 mo(2.7–5.7) | - | - | ||
DESTINY-Breast12 [57] | T-Dxd | Stable and Active BM | 263 | 60.1% | Overall 71.7% (64.2–79.3) Stable 79.2% (70.2–88.3) Active 62.3% (50.1–74.5) | Overall 51.7% (45.7–57.8) Stable 49.7 (41.9–57.5) Active 54.7 (45.2–64.2) | Overall 17.3 (13.7–22.1) | ||
KAMILIA [58] | TDM1 | Stable and Active BM | 398 | 47% | 42.9% (34.1–52.0) | 21.4% (14.6–29.6) | - | 5.5 mo (5.3–5.6) | 18.9 mo (0.1–21.3) |
PATRICIA [59] | Pertuzumab + high dose trastuzumab (6 mg/Kg weekly) | Active pretreated BM with stable extracranial disease | 40 | 100% | 11% (3.0–25.4) | - | 4.6 mo (4.0–8.9) | 4.6 mo (4.0–8.9) | 27.2 mo (16.1–NR) |
Tyrosine kinase inhibitors anti HER2 | |||||||||
EGF105084 [60] | Lapatinib | Active | 242 | 100% | 6% | 15% | - | 2.73 mo (1.87–3.45) | 9.56 (6.18-n/e) |
LANDSCAPE [61] | Lapatinib + Capecitabine | Active not previously treated with WBRT, capecitabine, or lapatinib | 45 | 0% | 65.9% (50.1–79.5) | 44·1% (27.2–62.0) | 5·5 mo (4.5–6.1) | 5·5 mo (4·3–6·0) | 17·0 mo (13·7–24·9) |
LAPTEM [62] | lapatinib–temozolomide | Recurrent or progressive BM | 18 | 83% | 0% | - | - | 2.60 mo (1.82–3.37) | 10.94 mo (1.09–20.79) |
Lin et al. 2013 [63] | Lapatinib + WBRT | Active BM | 35 | 0% | 79% (59–92) | - | - | 4.8 mo (range 0–58.3) | 19 mo (range 1–62) |
NRG Oncology − KROG/ RTOG1119 [64] | WBRT or SRS + Lapatinib | unirradiated BM | 71 | 0% | 53% (39–67) | 51% (37,65) | - | - | - |
WBRT or SRS Alone | 65 | 66% (51–81) | 58% (42–74) | - | - | - | |||
TBCRC 022 [65] | Neratinib | Active BM | 40 | 100% | 8% (2 −22) | - | - | 1.9 months | 8.7 months |
TBCRC 022 [66] | Neratinib + Capecitabine | Active BM | 37 lapatinib naïve | 92% | 49% (32—66) | - | - | 5.5 mo (range, 0.8 to 18.8) | 13.3 mo (range, 2.2 to 27.6 |
TBCRC 022 [67] | Neratinib + TDM1 | Active BM | 6 previously untreated BM | 0% | 50.0% (18.8- 81.2) | - | - | - | - |
17 progressing after prior local therapy without prior exposure to T-DM1 | 100% | 25.0% (8.3–52.6) | - | - | - | - | |||
21 progressing after prior local therapy with prior exposure to T-DM1 | 100% | 38.1% (19.0–61.3%) | - | - | - | - | |||
NALA [68] | Capecitabine + Neratinib | Stable BMs | 51 | 86.3% | 26.3% | - | 12.4 mo (5.6–17.9) | 5.6 mo (3.7–7.1) | 13.9 (8.9–17.5) |
Capecitabine + Lapatinib | 50 | 74% | 15.4% | - | 8.3 mo (4.3–NE) | 4.3 mo (2.8–5.6) | 12.4 (9.7–16.9) | ||
HER2CLIMB [12, 69] | Capecitabine + Trastuzumab + Tucatinib | Active or Stable BM | 198 Active = 118 Stable = 80 | 70% | All: 47.3% (33.7% – 61.2%) Active: 47.3% (33.7–61.2) | - | 9.9 (8.4–11.7) Active 9.6 mo Stabl: 13.9mo | - | 21.6 mo (18.1–28.5) Active 21.4 Stable 21.6 |
Capecitabine + Trastuzumab + Placebo | 93 Active = 56 Stable = 37 | 70% | All 20,0% (5,7%- 43,7%) Active: 20.0% (5.7%−43.7%) | - | 4.2 mo (3.6–5.7) Active 4.0 mo Stable 5.6 mo | - | 12.5 mo (11.2–16.9) Active 11.8 mo Stable 16.4 mo | ||
HER2CLIM2 | Tucatinib + TDM1 | Active or Stable BM | 70 | - | - | - | - | 7.8 mo (6.7 – 10) | - |
TDM1 | 85 | - | - | - | - | 5.7 mo (4.6 – 7.5) | - | ||
BROPTIMA [70] | Radiotherapy with pyrotinib and capecitabine | New Active BM | 40 | 12.5% | 85% | - | 18.0 mo (15.5 – NR) | 17.6 mo (12.8–34.) | - |
CDK4/6 inhibitors | |||||||||
I3Y-MC-JPBO [71] | Abemaciclib + Trastuzumab + endocrine therapy | Active | 27 | 96% | 0% | 0% | 2.7 mo (1.4–4.0) | 7.3 mo (3.3–13.3) | 10.1 mo (4.2–14.3) |
HER2 negative | |||||||||
Targeted Therapy | |||||||||
I3Y-MC-JPBO [71] | Abemaciclib + endocrine therapy | HR + Active BM | 58 | 81% | 5.2% (0.0%–10.9%) | 3.4% (0.0–8.1) | 4.9 mo (2.9–5.6) | 6.6 mo (4.3–12.4) | 12.5 mo (9.3–16.4) |
Leone et al [72] | Bevacizumab + Carboplatin (+ Trastuzumab if HER2 +) | Active BM | 38 29 HER2 + 9 HER2- | 87% | 63% (46–78) | - | - | 5.62 mo (.03–6.51) | 14.1 mo (11.7- 20.7) |
Lu et al [73] | Bevacizumab followed by BEEP | Refractory to WBRT | 35 23 HER2 + 12 HER2- | 100% | 77.1% (59.9–89.6) | 7.3 mo (6.5–8.1) | 6.1 mo (5.0–7.2) | 10.5 mo (7.8–13.2) | |
Antibody Drug Conjugates | |||||||||
DESTINY-Breast04 [74] | T-Dxd | HER2- low Stable BM | 24 (11 HR +) | 100% | 25% | 9.7 mo (4.4–15.1) | 16.7 mon (6.7–24.5) | ||
capecitabine, eribulin, gemcitabine, paclitaxel, or nab-paclitaxel | 11 (8 HR +) | 100% | 0% | ||||||
ASCENT [75] | Sacituzumab Govitecan | TNBC Stable BM | 32 | 100% | - | 3% | - | 2.8 mo (1.5–3.9) | 7 mo (4.7–14.7) |
capecitabine, eribulin, vinorelbine, or gemcitabine | 29 | - | 0% | - | 1.6 mo (1.3–2.9) | 7.5 mo (4.7–11.1) | |||
TROPION-Breast01 | Datopotamab Deruxtecan | HR + Stable BM | 35 | 100% | - | - | - | 5.6 mo (3.0–8.1) | - |
capecitabine, eribulin, vinorelbine, or gemcitabine | 23 | - | - | - | 4.4 (1.4–5.7) | - | |||
Chemotherapy | |||||||||
A-PLUS [76] | 3 cycles of BEEP followed by WBRT | All subtype Active BM | 77 | 0% | 59.5% | - | 8.1 mo (range, 0.3–29.5) | 7.9 mo | 15.9 mo (range, 0.8–58.1) |
WRBT alone | 41 | 0% | 65.8% | - | 6.5 mo (range, 0.9–25.5) | 5.0 months | 16.4 (range, 1.1–58.2) | ||
Rviviera et al [77] | Capecitabine + Temozolomide | All subtype Active BM | 24 | 16% | – | 12 weeks (range, 3–70) | 8 weeks (range, 6–64) | - | |
Franciosi et al [78] | Cisplatin + Etoposide | All subtype Newly diagnosed BM | 56 | 0% | 38% | - | 4 mo | - | |
Christodoulou et al [79] | Cisplatin + Temozolomide | Progressive brain metastases, | 32 (15 breast) | 53% | 31% | - | - | 2.9 mo (2.2–3.7; | 5.5mo (3.4–7.7) |
MBC Metastatatic Breast Cacner, BM Brain Metastases, CNS Central Nervous System, 95% CI 95% Confidence interval, ORR objective Response Rate, PFS Progression Free Survival, OS Overall Survival, Mo: Months, LMD Leptomeningeal Disease, T-Dxd Tratuzumab-Deruxtecan, Dato-Dxd Daptopotamab-Deruxtecan, TDM1 Trastuzumab Emtansine, WBRT Whole Brain Radiation Therapy, SRS Stereotactic Radiation Surgery, BEEP bevacizumab, etoposide, and cisplatin, TNBC triple negative breast cancer
HER2-Positive
The combination of pertuzumab, trastuzumab (HP), and taxanes is the first-line treatment for HER2-positive MBC following the results of the CLEOPATRA trial, which demonstrated a survival benefit with this regimen compared to taxanes plus trastuzumab. However, patients with CNS metastases were excluded from this trial [80]. Evidence supports the benefit of dual HER2 blockade with HP with respect to CNS outcomes. First, in the CLEOPATRA trial, the median time to the appearance of CNS metastases as the first site of disease progression was significantly longer in the pertuzumab group compared to the placebo group (15.0 months vs. 11.9 months; HR = 0.58, p = 0.0049). However, the incidence of CNS metastases as the first site of disease progression was similar between the arms (around 13%) [81]. Second, real-world data from a cohort of 252 patients with de novo BM showed significantly longer OS with first-line HP compared to treatments with other or without HER2-targeting therapies (44 vs 17 vs 3 months, p < 0.001) [82].
For second-line treatment, two regimens are highly active for patients with HER2-positive BM: trastuzumab-deruxtecan (T-DXd) and the combination of tucatinib, trastuzumab and capecitabine (HER2CLIMB regimen). In the sub-analysis of the DESTINY-Breast 03 trial among patients with BM, the median progression-free survival (PFS) was 15 months [95% confidence interval (CI) 12.5–22.2 months] for T-DXd compared to 3 months (95%CI 2.8–5.8 months) for Trastuzumab-emtansine (TDM1) (HR = 0.25, 95%CI 0.31–0.45). The CNS objective response rate (CNS-ORR) observed in DESTINY-Breast03 was 65.7% with T-DXd versus 34.3% with T-DM1 [55]. These data are consistent with the results from the non-comparative trials TUXEDO-1 and DEBBRAH, which reported CNS-ORRs between 44.4% and 73.3% in small numbers of patients with active BM [52, 83]. Moreover, in a pooled analysis of the DESTINY-Breast 01, 02, and 03 trials, patients with active and stable BM were treated with T-DXd compared to the reference treatment (Trastuzumab/Lapatinib + Capecitabine or TDM1). The CNS-ORR was 45% for active and stable BM with T-DXd compared to 12.0% and 27.6% with the reference treatment. The median CNS-PFS in the T-DXd group vs. standard treatment was 18.5 months (95%CI 13.6–23.3) vs. 4.0 months (95%CI 2.7–5.7) and 12.3 months (95%CI 11.1–13.8) vs. 8.7 months (95% CI 6.3–11.8) (HR = 0.19 and 0.59), respectively [56]. However, in these studies, an active BM refers only to asymptomatic cerebral lesions that have not been locally treated, as per the inclusion criteria. Patients with brain metastases that had progressed after local therapy were excluded, and therefore the patient population was not representative of all patients with active BM [84]. The DESTINY-Breast study12 is a real-world phase 3b/4 trial with the largest enrollment of BM patients (n = 263), including stable (n = 157) and active (n = 106) patients, with a distinction between untreated active patients (n = 39) and BM patients in progression (treated/progressed; n = 67) [57]. In contrast to previous studies, patients had received a median of 1(0–4). prior therapy. The median PFS for all BM patients was 17.3 months (CI95% 13.7–22.1). The 12-month PFS-CNS rates were 57.8% (95% CI: 48.2–66.1) and 60.1% (95% CI: 49.2–69.4) in patients with stable and active BM, respectively. The ORR-CNS rates were 79.2% (95% CI: 70.2–88.3), 82.6% (95% CI: 67.1–98.1) and 50.0% (95% CI: 34.1–65.9) in patients with stable, untreated and previously treatment/progressing BM, respectively. Toxicity was characterized by interstitial lung disease and pneumonitis observed in 16% of cases (grade 5: 2%), reminding us of the importance of considering Pneumocystis jirovecii pneumonia prophylaxis in patients taking chronic corticosteroids for symptoms of BM [57].
In the HER2CLIMB clinical trial, which compared tucatinib, trastuzumab, and capecitabine versus trastuzumab plus capecitabine in patients previously treated with taxanes, HP, and T-DM1 who had stable or active/progressive HER2-positive BM, the tucatinib group demonstrated significant benefits. These included improved CNS-PFS and CNS-ORR compared to the control arm of 9.9 vs. 4.2 months (HR = 0.39; p < 0.001) and 47.3% vs. 20.0% and, respectively. In addition, there was a reduction in the risk of developing new BM as the first site of progression or death (HR = 0.55, 95%CI 0.36–0.85). Finally, there was a significant increase in OS of 21.6 vs. 12.5 months (95%CI, 18.1–28.5 vs. 11.2–16.9, HR = 0.60; p < 0.001). The results were consistent in patients with both active and stable BM [12]. Preliminary data from the HER2CLIMB-02 trial, which compared TDM1 plus tucatinib to placebo in patients with HER2-positive MBC, have been reported. Forty percent of the participants had BM, half of which were active. The median PFS for patients with BM was 7.8 months compared to 5.7 months for the placebo group (95%CI, 6.7–10.0; 4.6–7.5; HR = 0.64). We await data on intracranial endpoints from this clinical trial.
No direct comparison has been made between these two treatments in patients with HER2-positive BM. Given the differences in the populations selected in these two trials, particularly in the lines and types of prior therapies and the inclusion criteria around active BM. In addition, patients enrolled in DESTINY12 could not have received prior tucatinib treatment, making it impossible to assess cross-resistance. The DESTINY12 results confirm the intracranial activity of T-DXd, which should be used as second-line treatment in patients with and without central nervous system (CNS) metastases after progression on trastuzumab and doublet pertuzumab. The HER2CLIMB-04 trial (NCT04539938) will evaluate whether the combination of tucatinib and TDXd is beneficial in patients with HER2 + MBC with or without BM.
For subsequent lines of treatment, neratinib and lapatinib in combination with capecitabine have shown intracranial response rates in patients with active brain metastases in the TBCRC 022 and LANDSCAPE trials (non-irradiated). The CNS-ORRs were 49% (95%CI 32–66) and 65.9% (95%CI 50.1–79.5), respectively, with a median PFS of around 5 months [61, 66]. The activity of either regimen after prior tucatinib are unknown, although the TBCRC 022 study did demonstrate activity of neratinib after prior lapatinib exposure. The combination of neratinib with T-DM1 has also demonstrated intracerebral activity in patients with untreated brain metastases (n = 6), those progressing after prior CNS-directed local therapy without prior exposure to T-DM1 (n = 16), and those with prior exposure to T-DM1 (n = 21), with CNS-ORR of 50.0% (95% CI 18.8–81.2%), 25.0% (95% CI 8.3–52.6%), and 38.1% (95% CI 19.0–61.3%), respectively [67].
For patients with isolated CNS progression despite previous radiotherapy, high-dose intravenous trastuzumab (6 mg/kg per week) in combination with pertuzumab and the continuation of systemic therapy is an option. In the PATRICIA trial, among 40 heavily pretreated HER2-positive MBC patients with brain metastases (70% had received WBRT at the initial diagnosis of CNS metastases, and 70% had undergone SRS at the time of CNS progression), the CNS-ORR was 11% (95% CI: 3.0–25.4). The median CNS-PFS was 4.6 months (95%CI: 4.0–8.9), and the clinical benefit rate (CBR) in the CNS at six months was 51% (95%CI: 34.4–68.1) [59, 85].
HR-Positive
BM tend to occur after a longer interval of metastatic disease in patients with HR + /HER2- MBC. Pivotal trials for cyclin-dependent kinase 4 and 6 inhibitors (CDK4/6i) generally excluded patients with BM. The phase II JPBO trial assessed abemaciclib plus endocrine therapy in patients with active BM who had not previously received CDK4/6i. These patients had received a median of three prior treatments. The CNS-ORR was 5.2% (95% CI, 0.0–10.9). The median PFS and OS were 4.4 months (95% CI, 2.6–5.5) and 12.5 months (95% CI, 9.3–16.4) [71]. The post-marketing phase IIIb CompLEEment-1 study evaluated ribociclib plus letrozole in patients with HR + /HER2- MBC and demonstrated a safety profile, time to progression, and extracranial response rates similar between patients with CNS metastases (n = 51) and the overall population (n = 3,246). These results suggest a comparable benefit of Ribociclib in patients with CNS metastases from HR + /HER2- MBC as observed in the overall study population, but without providing information on intracranial responses [86].
Antibody–drug conjugates (ADC) are promising in treating BM across MBC subtypes. T-DXd was evaluated in a cohort of patients with HER2-low BM in the DESTINY-04 clinical trial. Among the 35 patients with BM, most of whom had HR-positive disease (approximately 75%), 24 received T-DXd, and 11 received chemotherapy. The median PFS for T-DXd was 9.7 months (95% CI, 4.4–15.1 months) compared to not evaluable for chemotherapy due to the small number of patients. The CNS objective response rate (ORR-CNS) was 6/24 (25%) (95% CI, 9.8%−46.7%) for T-DXd and 0/11 (95% CI, 0%−28.5%) for chemotherapy. In subsequent lines, preliminary data from the TROPION-Breast01 trial, which compared datopotamab-deruxtecan (Dato-DXd) with standard chemotherapy in patients with HR-positive MBC previously treated with chemotherapy, showed a significant improvement in PFS (HR 0.63; p < 0,0001). Among patients with stable and asymptomatic BM (35 in the Dato-DXd group and 23 in the chemotherapy group), there was no significant difference in median PFS, which was 5.6 months for the Dato-DXd group versus 4.4 months for the chemotherapy group (HR = 0.73, 95%CI 0.39–1.42) [87]. Data from the TROPICS trial of Sacituzumab-Govitecan (SG) in HR + patients with BM have not been released. However, preliminary data from a window-of-opportunity study planned one dose of SG to be administered prior to BM resection and continued until progression in 13 patients with MBC (all subtype). In this highly selected population, median PFS was 8 months (range 2–26.5 months) with a CNS ORS of 50%. Morover, SN-38 levels in bone marrow tissue reached therapeutic levels after a single injection [88]. These data suggest intracranial activity of SG, which will be validated in SWOG S2007 (NCT04647916), a phase 2 trial of SG in patients with HER2-negative MBC and BM. Finally, several cases have been reported in which endocrine therapies and a PI3K inhibitor, alpelisib, have led to durable responses in BM in patients with HR-positive tumors [89].
TNBC
In the ASCENT trial, 61 out of 529 (12%) enrolled patients had stable BM at the time of selection and received either SG (n = 32) or standard chemotherapy (n = 29). In this subgroup, the median PFS was 2.8 months (95%CI, 1.5–3.9) for SG compared to 1.6 months (95%CI, 1.3–2.9) for chemotherapy. The median OS was 6.8 months (95%CI, 4.7–14.1) for SG versus 7.5 months (95%CI, 4.7–11.1) for the chemotherapy group. The ORR for SG vs chemotherapy was 3% (1/32) versus 0%, respectively [75]. The efficacy of Sacituzumab-Govitecan in patients with active brain metastases is unknown and is currently under investigation (NCT04647916). We lack data on the combination of pembrolizumab with chemotherapy as a first-line treatment in TNBC due to the exclusion of patients with active BM and the small number of patients included with stable BM (3%). However, the presence of BM should not preclude the addition of pembrolizumab, considering the clinical benefit rate of 36.4% (4/7) observed in monotherapy among pretreated TNBC patients in a multi-histology BM phase II basket trial with pembrolizumab [90].
HER2-Low
In patients with HER2-low cancer, T-DXd demonstrated a significant median PFS gain of 4.8 months compared to standard treatment. In an exploratory study, among patients with stable asymptomatic BM (35 patients [24 T-DXd; 11 controls]), the CNS-ORR was 25% vs 0%. In the T-DXd group, the PFS and OS were 9.7 months (95%CI, 4.4–15.1) and 16.7 months (95%CI, 6.7–24.5), respectively [74]. These data align with findings from the DAISY trial, which evaluated T-DXd in patient cohorts with HER2-low and HER2-negative. Among patients with stable BM, the best objective responses (BOR) were observed at 30% (3/10; (95%CI 6.7–65.2) for the HER2-low group and 50% (1/2; (95% CI 1.3–98.7) for the HER2-negative group, respectively [91].
All Subtypes
Bevacizumab has shown benefits in PFS but not in OS in MBC; however, all phase 3 trials of bevacizumab in MBC strictly excluded patients with any history of brain metastases due to concern for intracranial hemorrhage. Since then, two phase 2 studies have demonstrated significant response rates of 63–77% in patients with BM when combined with chemotherapy (and trastuzumab if HER2-positive). No clinically significant intracranial hemorrhages were reported in either study [72, 73]. Chemotherapies have demonstrated activity in the CNS, largely based on case series capecitabine, anthracyclines, and platinum agents, with response rates ranging from 18–54%, 17–41%, and 28–38%, respectively [77, 78–79, 92, 93]. Retrospective data are also available for eribulin and irinotecan [94, 95]. Despite its known ability to cross the BBB, temozolomide has not shown efficacy in BM in randomized trials [96]. Finally, PARP inhibitors have also been the subject of case reports in patients with BRCA germline alteration [97].
Treatment of LMD among MBC Patients
Treatments for LMD are based on studies with low levels of evidence, including small prospective studies and retrospective cohorts. Treatment decisions should be made through a multidisciplinary approach tailored to each case.
Radiation Therapy
Radiation therapy (RT) remains an effective treatment for improving neurological symptoms in LMD. WBRT should be reserved for patients with extensive LMD and coexisting BM [98]. In contrast, local involvement could receive involved-field RT (IFRT) or hypo-fractionated stereotactic RT [99]. Craniospinal irradiation (CSI) with photons has been described but was rarely used due to its significant systemic side effects. However, proton craniospinal irradiation (pCSI) limits the radiation beyond the neurological axis, thereby reducing systemic side effects. A phase II study with solid tumors, including breast cancer, showed a significant advantage in terms of CNS-PFS and OS over pCSI compared to photon-based IFRT, median 7.5 months (CI95%, 6.6 months to not reached (NR)) vs. 2.3 months; (CI95%, 1.2- 5.8; p < 0.001) and 9.9 months (CI95%, 7.5-NR) vs. 6.0 months (CI95% 3.9-NR; p = 0.029) without new or additive toxicity [100]. Unfortunately, the utility of pCSI for the treatment of LMD is limited by cost and the relative scarcity of centers capable of delivering proton therapy. Further studies evaluating pCSI in the management of LMD are being developed.
Intrathecal Therapy
The rationale for choosing intrathecal administration is to achieve higher cerebrospinal drug concentrations while minimizing systemic toxicity. Initially developed with chemotherapy, several trials and cohorts have evaluated methotrexate (MTX), liposomal cytarabine, and irinotecan [16]. However, no trials have demonstrated an improvement in OS. The DEPOSEIN clinical trial, which compared liposomal-cytarabine combined with systemic treatment against systemic treatment alone, reported a median LM-PFS (leptomeningeal progression-free survival) of 2.2 months (95%CI 1.3–3.1) in the control group versus 3.8 months (95%CI 2.3–6.8) in the experimental group (HR = 0.61, p = 0.04). Despite these findings, the quality of life up to progression did not differ between groups [101]. Retrospective data have shown that intrathecal topotecan resulted in clearing of CSF malignancy in 10/21 (48%) patients with a median duration of 15.9 months (range, 1.4–34.5) [102]. A second intrathecal treatment option involves the administration of trastuzumab, which has demonstrated a tolerable safety profile and extended survival in two phase 2 trials, with OS ranging from 8 to 10 months [103, 104].
Systemic Therapy
Systemic therapy data for LMD come from retrospective cohorts or subgroups within prospective trials. However, promising efficacy data from new anti-HER2 therapies, notably T-Dxd, have been reported. Two cohorts from the United States and Japan involving heavily pretreated HER2-positive MBC patients with progressive LMD showed response rates of 50–78% and median OS exceeding 12 months [105, 106]. Additionally, the tucatinib regimen has demonstrated intracranial response, improved symptoms, improved quality of life, and extended survival in patients with LMD in the TBCRC049 study. Median OS was 10 months (95% CI: 4.1, not reached), and most patients with target deficits at baseline experienced improvement with the tucatinib regimen [107, 108]. Standard chemotherapies have not demonstrated long-term benefits for LMD patients. However, some agents like capecitabine, high-dose IV methotrexate and platinum-based therapies have shown some activity and may be considered [16]. Endocrine therapies have also shown some activity based on case. Abemaciclib was evaluated in the prospective JPBO clinical trial, which included an LMD cohort and observed one response in seven patients with a median PFS of 5.9 months (95% CI 0.7–8.6) and median OS of 8.4 months (95% CI 3.3–23.5) [71].
Surveillance and Follow-up of CNS Metastases among MBC Patients
Patients with CNS metastases should undergo regular clinical and radiologic follow-up to detect any treatment complications or intracranial progression [34, 109]. Clinical evaluation should include periodic neurologic and cognitive assessments, possibly using the Neurologic Assessment in Neuro-Oncology (NANO) scale [110]. Imaging should include MRI with intravenous gadolinium-based contrast every 2–3 months or earlier based on symptoms. Perfusion and spectroscopic MRI and amino acid PET scans can help distinguish between radionecrosis and progression of brain metastases in cases of doubt [111, 112–113]. Although the RANO-BM criteria were developed for clinical trials, they are also useful in routine practice. Unlike the RECIST criteria, RANO-BM considers changes in both target and non-target lesions on conventional contrast-enhanced MRI, neurological status, and steroid use [114]. The RANO-LM criteria provide support for the follow-up of patients with LMD. In addition to clinical examination and neuroaxis MRI, they also include CSF analysis [115].
Prevention of CNS Recurrence: Limited Success to Date
Unfortunately, few adjuvant therapies have been shown to reduce the risk of CNS recurrence. A meta-analysis of trials evaluating adjuvant trastuzumab showed that it was associated with a significant increase in the risk of CNS metastases as the site of first recurrence 2.56% versus 1.94% (RR = 1.35, p = 0.038), suggesting a greater reduction in extracranial than intracranial relapse risk with trastuzumab [116]. In the APHINITY trial, no difference was observed in the incidence of CNS metastases (2.0% in both groups) with the addition of pertuzumab to adjuvant trastuzumab [117]. Similarly, patients with residual disease after neoadjuvant treatment who received T-DM1 versus adjuvant trastuzumab in the KATHERINE trial had a higher risk of first CNS relapse (7.0% vs. 5.1%) [10]. However, in a post-hoc analysis, HR-positive/HER2-positive patients treated with adjuvant neratinib in the ExteNET trial had a lower cumulative incidence of CNS relapse at 5 years compared to placebo (0.7% vs. 2.1%), suggesting a potential signal in favor of the HER2 TKI [118]. The ongoing phase 3 clinical trial COMPASS-HER2 RD is comparing the combination of TDM1 with or without tucatinib for HER2 + patients with residual disease after neoadjuvant treatment. Brain metastases-free survival (BMFS) is a secondary objective to evaluate whether tucatinib prevents CNS metastases for these at-risk patients (NCT04457596).
In patients with TNBC, adding pembrolizumab to neoadjuvant and adjuvant chemotherapy in the KEYNOTE 522 trial reduced the overall risk of CNS relapse (3.6% vs. 2.1%). However, in the RCB-0 and RCB-1 categories, more than half of the recurrences occurred in the CNS, with 13/22 (59.1%) in the pembrolizumab group and 8/16 (50.0%) in the placebo group [11]. These results are consistent with retrospective data from patients with HER2 + MBC [6]. Brain micrometastases are "protected" within the CNS and require additional strategies for patients with complete pathologic responses.
Promising Novel Therapeutics
Numerous new treatments and combinations capable of crossing the BBB or blood-tumor barrier are under development and showing promising results in patients with CNS metastases. All active trials are summarized in Table 2. The results from T-Dxd have spurred significant development of new ADCs for patients with CNS metastases, including HER2-targeting ADCs like ARX788 and SHR-A1811, TROP2-targeting ADC SHR-A1921, and HER3-targeting ADC patritumab-deruxtecan. Innovative treatments such as CAR-T cell therapy targeting HER2 are currently being studied in a phase 1 trial (NCT03696030) in patients with BM and LMD.
Table 2. Recruiting or Not Yet recruiting clinical trials on systemic treatment in Breast Cancer Patients with CNS Metastases
NCT Number ACRONYM | Trial Name | Interventions | Primary Outcome Measures | Phases | Completion Date |
|---|---|---|---|---|---|
MBC patients with Brain Metastases | |||||
HER2-POSITIVE | |||||
NCT04334330 | Palbociclib, Trastuzumab, Pyrotinib and Fulvestrant Treatment in Patients with BM From ER/PR Positive, HER-2 Positive BC: A Multi-center, Prospective Study in China | Palbociclib, Trastuzumab, Pyrotinib and Fulvestrant | CNS-ORR (RECIST 1.1) | 2 | 2024–12 |
NCT06152822 | Pyrotinib Combined with Capecitabine and Bevacizumab for Patients with HER2 Positive BC and BM | Pyrotinib Combined with Capecitabine and Bevacizumab | CNS-ORR (RECIST 1.1) | 2 | 2025–11 |
NCT05553522 | Tucatinib, Trastuzumab, and Capecitabine with SRS for BM From HER-2 Positive BC | Combined use of SRS with Tucatinib, Trastuzumab, and Capecitabine | dose-limiting toxicities | 1 | 2025–11 |
NCT04582968 | Pyrotinib Combined with Brain Radiotherapy in BC Patients With BM | Pyrotinib Plus Capecitabine combined with brain radiotherapy | Assess safety and tolerability | 1/2 | 2023–08 |
NCT03417544 | Atezolizumab + Pertuzumab + Trastuzumab in CNS Mets In BC | Atezolizumab + Pertuzumab + Trastuzuma | CNS ORR (RANO-BM) | 2 | 2025–12 |
NCT04760431 HER2BRAI | TKIs vs. Pertuzumab in HER2 + BC Patients with Active BM (HER2BRAIN) | Anti-HER2 TKI versus Pertuzumab in Combination with Dose-dense Trastuzumab and Taxane | ORR | 2 | 2025–09 |
NCT05042791 | A Study of Pyrotinib Plus Capecitabine Combined with SRT in HER2 + MBC With BM | Pyrotinib Plus Capecitabine Combined With SRT | CNS-ORR, | 2 | 2025–09 |
NCT05018702 | ARX788 in HER2-positive BC Patients With BM | ARX788 ((HER2-targeting ADC) | CNS-CBR (RANO-BM) | 2 | 2023–06 |
NCT06361979 | SHR-A1811 Combined with Bevacizumab in HER2-positive BC With BM | SHR-A1811 (HER2-targeting ADC) Combined with Bevacizumab | CNS-ORR (RANO-BM) | 2 | 2026–05 |
NCT05769010 | Study of SHR-A1811 in HER2-expression MBC er with BM | SHR-A1811 (HER2-targeting ADC) plus Pyrotinib and Bevacizumab | CNS-ORR (RANO-BM) | 2 | 2026–04 |
NCT05323955 BRIDGET | Secondary BM Prevention After Isolated Intracranial Progression on Trastuzumab/Pertuzumab or T-DM1 in Patients With aDvanced Human Epidermal Growth Factor Receptor 2 + BC With the Addition of Tucatinib | Tucatinib plus Trastuzumab/Pertuzumab or T-DM1 | Bicompartmental PFS (RANO-BM) | 2 | 2025–04 |
NCT06015113 | Efficacy and Safety of Disitamab Vedotin Plus Pyrotinib or Naratinib in HER2-positive BC Patients With BM | Disitamab Vedotin Plus Pyrotinib or Naratinib | PFS (RECIST1.1) | 2 | 2027–04 |
NCT05041842 InTTercePT | Treatment With Tucatinib in Patients with an Isolated Brain Progression of a MBC | Tucatinib with Pertuzumab/Trastuzumab (and endocrine therpy) | PFS (RECIST1.1) | 2 | 2026–03 |
NCT03765983 | GDC-0084 in Combination with Trastuzumab for Patients with HER2-Positive BC BM | GDC-0084 (inhibitor of PI3K and mTOR) in Combination with Trastuzumab | ORR (RANO-BM) | 2 | 2025–11 |
NCT06088056 | A Phase II Study of T-DXd Plus SRT in HER2-positive BC BM | T-DXd Plus stereotactic radiotherapy | CNS-ORR (RANO-BM) | 2 | 2026–07 |
NCT03691051 | A Study of Pyrotinib Plus Capecitabine in Patients with BM From HER2-positive MBC | Pyrotinib Plus Capecitabine | CNS-ORR (RECIST1.1) | 2 | 2024–12 |
TNBCpé | |||||
NCT03483012 | Atezolizumab + Stereotactic Radiation in Triple-negative BC and BM | Atezolizumatrob + Stereotactic Radiation | bi-compartmental-PFS (RANO-BM) | 2 | 2025–09 |
NCT06210438 | SHR-A1921 Combined with Bevacizumab in Triple-negative BC With BM | SHR-A1921 (TROP2-targeting ADC)) Combined with Bevacizumab | CNS ORR (RANO-BM) | 2 | 2026–06 |
NCT06238921 | Sacituzumab Govitecan and Zimberelimab w/SRS in the Management of Metastatic TNBC With BM | Sacituzumab Govitecan and Zimberelimab (anti-PD1) w/ SRS | Phase I: Neurologic Toxicity, Phase II: PFS | 1/2 | 2027–02 |
NCT05866432 TUXEDO-2 | Phase II Study of Dato-DXd in Triple-negative BC Patients with Newly Diagnosed or Progressing BM | Dato-DXd | CNS-ORR (RANO-BM) | 2 | 2026–05 |
HR-positive / HER2-negative | |||||
NCT04791384 | Phase Ib/II Trial of Abemaciclib and Elacestrant in Patients with BM Due to HR + /Her2- BC | Abemaciclib and Elacestrant | Phase I: Safety Phase II: CNS-ORR (RANO-BM) | 1/2 | 2025–01 |
NCT05386108 ELECTRA | Study of Abemaciclib and Elacestrant in Patients with BM Due to HR + /HER2- BC | Abemaciclib and Elacestrant | Safety | 1/2 | 2025–12 |
NCT04923542 | Stereotactic Radiation & Abemaciclib in the Management of HR + /HER2- BC BM | Stereotactic Radiation & Abemaciclib | CNS-PFS (RANO-BM) | 1/2 | 2024–12 |
NCT05872347 | Efficacy and Safety of SPH4336 in Combination with Endocrine Therapy in BC Patients With BM | SPH4336 (CDK 4/6 inhibitor) in Combination with Endocrine Therapy | CNS-ORR (RANO-BM) | 2 | 2025–12 |
All Subtype | |||||
NCT05305365 | Study Assessing QBS72S For Treating BM | QBS72S | ORR (RANO-BM) | 2 | 2026–08 |
NCT03807765 | Stereotactic Radiation and Nivolumab in the Management of MBC BM | Nivolumab Stereotactic Radiosurgery | Safety and Neurologic dose limiting toxicities | 1 | 2024–04 |
NCT05357417 | Utidelone Plus Bevacizumab for MBC With BM | Utidelone Plus Bevacizumab | CNS-ORR (RECIST1.1) | 2 | 2024–05 |
NCT03328884 Phenomenal | Evaluation of the Efficacy and Safety of Nal-IRI for Progressing BM in BC Patients | Irinotecan Hydrochloride | CNS-ORR (RANO-BM) | 2 | 2025–06 |
NCT03449238 | Pembrolizumab And Stereotactic Radiosurgery (Srs) Of Selected BM In BC Patients | Pembrolizumab And Stereotactic Radiosurgery | Tumor response for non-irradiated brain (RECIST1.1) | 1/2 | 2027–12 |
NCT05535413 | UTD1 Combined with Capecitabine in Metastatic HER2-negative BC Patients With BM | UTD1 Combined with Capecitabine | CNS-ORR (RANO-BM) | 1/2 | 2025–07 |
NCT06048718 TUXEDO-4 | T-DXd Therapy for HER2-low Breast Cancer Patients With BM | T-DXd | CNS-ORR (RANO-BM) | 2 | 2026–07 |
NCT04711824 SOLARA | Study of Stereotactic Radiosurgery with Olaparib Followed by Durvalumab and Physician's Choice Systemic Therapy in Subjects with BC BM | Stereotactic Radiosurgery with Olaparib Followed by Durvalumab | Phase I: Safety Phase II: CNS-ORR (RANO-BM) | 1/2 | 2026–09 |
NCT05781633 | The Efficacy and Safety of Eutideron, Etoposide, and Bevacizumab in Patients with BM From BC | Eutideron, Etoposide, and Bevacizuma | CNS-ORR (RANO-BM) | 2 | 2025–07 |
NCT04348747 | Dendritic Cell Vaccines Against Her2/Her3 and Pembrolizumab for the Treatment of BM From TNBC or HER2 + BC | Dendritic Cell Vaccines Against Her2/Her3 and Pembrolizumab | CNS-ORR (RANO-BM) | 2 | 2026–12 |
NCT04647916 | Testing Sacituzumab Govitecan Therapy in Patients with HER2-Negative BC and BM | Sacituzumab Govitecan | ORR | 2 | 2026–12 |
NCT02595905 | Cisplatin With or Without Veliparib in Treating Patients with Recurrent or Metastatic TNBC and/or BRCA Mutation-Associated BC With or Without BM | Cisplatin With or Without Veliparib | PFS | 2 | 2024–10 |
MBC patients with Brain Metastases and or LMD | |||||
NCT06176261 DATO-BASE | DATO-BASE: DATOpotamab-deruxtecan for BC BM | Datopotamab Deruxtecan | ORR (RANO-BM) | 2 | 2029–01 |
NCT03696030 | HER2-CAR T Cells in Treating Patients with Recurrent Brain or LMD | HER2-CAR T Cells | Safety | 1 | 2025–02 |
MBC patients with LMD | |||||
NCT05746325 | Tumor Treating Fields for the Treatment of LMD of the Spine in Patients With BC | tumor treating fields (TTF) | Safety | NA | 2027–03 |
NCT03501979 | Tucatinib, Trastuzumab, and Capecitabine for the Treatment of HER2 + LMD | Tucatinib, Trastuzumab, and Capecitabine | OS | 2 | 2024–07 |
NCT03613181 ANGLeD | ANG1005 in Leptomeningeal Disease From BC | ANG1005 (taxane linked to Angiopep-2) | OS | 3 | 2024–12 |
NCT05800275 ETIC-LM | Capecitabine, Tucatinib, and Intrathecal Trastuzumab for Breast Cancer Patients With LMD | Capecitabine, Tucatinib, and Intrathecal Trastuzumab | OS | 2 | 2027–06 |
NCT04588545 | Radiation Therapy Followed by Intrathecal Trastuzumab/Pertuzumab in HER2 + Breast LMD | Radiation Therapy Followed by Intrathecal Trastuzumab/Pertuzumab | Phase 1: Maximum Tolerated Dose Phase 2: OS | 1/2 | 2024–11 |
NCT06016387 | Tucatinib With Brain and/or Spinal XRT in Patients with HER2 + MBC and LMD | Tucatinib With Brain and/or Spinal XRT | OS | 2 | 2028–10 |
BC Breast Cancer, MBC Metastatatic Breast Cacner, BM Brain Metastases, CNS Central Nervous System, TNBC triple negative breast Cancer
Utidelone, an epothilone analog engineered through genetic engineering, has recently shown promise in a phase 2 trial in combination with etoposide and bevacizumab in heavily pretreated breast cancer patients with CNS metastases, achieving a CNS-ORR of 73% (8/11 patients) [119]. The primary adverse event (AE) was peripheral neuropathies in 9% (1/11, grade 3) of patients. ANG1005, a novel taxane derivative consisting of paclitaxel covalently linked to Angiopep-2, designed to penetrate the CNS via the LRP1 transport system, demonstrated a CNS-ORR of 15% in a phase II study among adult patients with MBC CNS metastases (BM n = 72 and LDM n = 28). In the LMD subgroup, 79% of patients achieved intracranial disease control with an estimated median OS of 8.0 months (95%CI, 5.4–9.4) [120].
Innovative trials, such as the phase 2 Alliance A071701 trial (NCT03994796), are ongoing and evaluate genomic-guided treatment choices for BM. This trial assesses abemaciclib, GDC-0084 (a PI3K and mTOR inhibitor), and entrectinib in patients with alterations in NTRK, ROS1, CDK, or PI3K genes in brain metastasis. Also, trials like BRIDGET (NCT05323955) and InTTercePT (NCT05041842) are evaluating the addition of tucatinib and the continuation of ongoing treatment (HP or TDM1) in cases of isolated brain progression, thus potentially delaying radiotherapy.
Summary
CNS metastases are a common occurrence and an unmet need associated with significant morbidity and poor prognosis in MBC patients. However, advances in local and systemic treatments have significantly improved the OS of these patients. Multimodal treatment strategies require a multidisciplinary approach. Local treatment remains the cornerstone of active, especially newly diagnosed, BM. However, the emergence of highly effective systemic therapies may soon lead to a paradigm shift in the multidisciplinary therapeutic approach to BM.
Key References
Brastianos PK, Carter SL, Santagata S, Cahill DP, Taylor-Weiner A, Jones RT, et al. Genomic Characterization of Brain Metastases Reveals Branched Evolution and Potential Therapeutic Targets. Cancer Discovery. 2015;5:1164–77.
○ Genetic profiling of 86 matched brain metastases, primary tumors, and normal tissue demonstrated that in 53% of cases, we found potentially clinically informative alterations in the brain metastases that were not detected in the matched primary tumor sample.
Reese RA, Lamba N, Catalano PJ, Cagney DN, Wen PY, Aizer AA. Incidence and Predictors of Neurologic Death in Patients with Brain Metastases. World Neurosurgery. 2022;162:e401–15.
○ Study of incidence in CNS metastases patients across type of cancer
Lin NU, Murthy RK, Abramson V, Anders C, Bachelot T, Bedard PL, et al. Tucatinib vs Placebo, Both in Combination With Trastuzumab and Capecitabine, for Previously Treated ERBB2 (HER2)-Positive Metastatic Breast Cancer in Patients With Brain Metastases: Updated Exploratory Analysis of the HER2CLIMB Randomized Clinical Trial. JAMA Oncology. 2023;9:197–205.
○ The first phase 3 trial with significant improvement in CNS-PFS and OS that included patients with active brain metastases.
Sperduto PW, Mesko S, Li J, Cagney D, Aizer A, Lin NU, 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. International Journal of Radiation Oncology*Biology*Physics. 2020;107:334–43.
○ Study demonstrating the association with overall survival of the Graded Prognostic Assessment (Breast GPA).
McTyre ER, Soike MH, Farris M, Ayala-Peacock DN, Hepel JT, Page BR, et al. Multi-institutional validation of brain metastasis velocity, a recently defined predictor of outcomes following stereotactic radiosurgery. Radiotherapy and Oncology. 2020;142:168–74.
○ Study demonstrating the association with overall survival of brain metastasis velocity
Vecht CJ, Haaxma-Reiche H, Noordijk EM, Padberg GW, Voormolen JHC, Hoekstra FH, et al. Treatment of single brain metastasis: Radiotherapy alone or combined with neurosurgery. Annals of Neurology. 1993;33:583–90.
○ Phase 3 study demonstrated a significant OS gain for surgery followed by radiation therapy for a brain metastasis.
Patchell Roy A., Tibbs Phillip A., Walsh John W., Dempsey Robert J., Maruyama Yosh, Kryscio Richard J., et al. A Randomized Trial of Surgery in the Treatment of Single Metastases to the Brain. New England Journal of Medicine. 1990;322:494–500.
○ Phase 3 study demonstrated a significant OS gain for surgery followed by radiation therapy for a brain metastasis.
Brown PD, Pugh S, Laack NN, Wefel JS, Khuntia D, Meyers C, et al. Memantine for the prevention of cognitive dysfunction in patients receiving whole-brain radiotherapy: a randomized, double-blind, placebo-controlled trial. Neuro-Oncology. 2013;15:1429–37.
○ Study of memantine for prevention of cognitive dysfunction in patients receiving whole-brain radiotherapy
Brown PD, Gondi V, Pugh S, Tome WA, Wefel JS, Armstrong TS, et al. Hippocampal Avoidance During Whole-Brain Radiotherapy Plus Memantine for Patients With Brain Metastases: Phase III Trial NRG Oncology CC001. JCO. 2020;38:1019–29.
○ Study of hippocampal avoidance for prevention of cognitive dysfunction in patients receiving whole-brain radiotherapy
Hurvitz SA, Modi S, Li W, Park YH, Chung W, Kim S-B, et al. 377O A pooled analysis of trastuzumab deruxtecan (T-DXd) in patients (pts) with HER2-positive (HER2 +) metastatic breast cancer (mBC) with brain metastases (BMs) from DESTINY-Breast (DB) −01, −02, and −03. Annals of Oncology. 2023;34:S335–6.
○ Pooled analysis of DESTINY-Breast (DB) −01, −02, and −03 on patients with brain metastases (BMs)
Harbeck N, Ciruelos E, Jerusalem G, Müller V, Niikura N, Viale G, et al. Trastuzumab deruxtecan in HER2-positive advanced breast cancer with or without brain metastases: a phase 3b/4 trial. Nat Med. 2024;1–10.
○ Largest trial evaluating T-DXd in HER2-positive patients with stable, active BM
Yang JT, Wijetunga NA, Pentsova E, Wolden S, Young RJ, Correa D, et al. Randomized Phase II Trial of Proton Craniospinal Irradiation Versus Photon Involved-Field Radiotherapy for Patients With Solid Tumor Leptomeningeal Metastasis. Journal of Clinical Oncology [Internet]. 2022
○ Phase II study of Proton Craniospinal Irradiation for LMD patients
Acknowledgements
The authors used generative artificial intelligence tools such as ChatGPT and DeepL to improve the syntax and language. The first author would like to thank Lucile Cabanel for her unfailing help.
Author Contribution
All authors contributed to the reveiw conception . T.G wrote the first draft of the manuscript. S.S supervised the course of the article. All authors critically reviewed the manuscript and approved the final manuscript.
Data Availability
No datasets were generated or analysed during the current study.
Declarations
Conflict of Interests
TG discloses Travel fees from AstraZeneca, Gilead, and Pfizer. Consulting/Advisor role for AstraZeneca and a personal Grant from the Philippe Foundation.
AA notes research funding from Varian and NH TherAguix as well as consulting for Novartis and Seagen.
NUL declares consulting honorarium from Seagen, Daichii-Sankyo, AstraZeneca, Olema Pharmaceuticals, Janssen, Blueprint Medicines, Stemline/Menarini, Artera Inc., Eisai; travel support from Olema Pharmaceuticals; research support (to institution) from Genentech, Zion Pharmaceuticals (as part of GNE), Pfizer, Seagen, Merck, Olema Pharmaceuticals, AstraZeneca; royalties from Up To Date.
SS declares research funding to their institution from Astra Zeneca, Eli Lilly, Relay, SEAGEN and Sermonix; and consulting fees from Foundation Medicine, Astra Zeneca, Daichii Sankyo, Eli Lilly, Pfizer, Incyclix, Relay, Gilead, Sermonix and Novartis.
Human and Animal Rights and Informed Consent
This article does not contain any studies with human or animal subjects performed by any authors.
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
References and Recommended Reading
1. Lamba, N; Wen, PY; Aizer, AA. Epidemiology of brain metastases and leptomeningeal disease. Neuro Oncol; 2021; 23, pp. 1447-1456. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/33908612][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8408881][DOI: https://dx.doi.org/10.1093/neuonc/noab101]
2. Aizer, AA; Lamba, N; Ahluwalia, MS; Aldape, K; Boire, A; Brastianos, PK et al. Brain metastases: a society for neuro-oncology (SNO) consensus review on current management and future directions. Neuro Oncol; 2022; 24, pp. 1613-1646. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/35762249][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9527527][DOI: https://dx.doi.org/10.1093/neuonc/noac118] 1:CAS:528:DC%2BB3sXpsF2qsrc%3D
3. Achrol, AS; Rennert, RC; Anders, C; Soffietti, R; Ahluwalia, MS; Nayak, L et al. Brain metastases. Nat Rev Dis Primers; 2019; 5, pp. 1-26. [DOI: https://dx.doi.org/10.1038/s41572-018-0055-y]
4. Kim, AE; Nieblas-Bedolla, E; de Sauvage, MA; Brastianos, PK. Leveraging translational insights toward precision medicine approaches for brain metastases. Nat Cancer; 2023; 4, pp. 955-967. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/37491527][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10644911][DOI: https://dx.doi.org/10.1038/s43018-023-00585-0]
5. Brastianos, PK; Carter, SL; Santagata, S; Cahill, DP; Taylor-Weiner, A; Jones, RT et al. Genomic characterization of brain metastases reveals branched evolution and potential therapeutic targets. Cancer Discov; 2015; 5, pp. 1164-1177. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/26410082][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4916970][DOI: https://dx.doi.org/10.1158/2159-8290.CD-15-0369] 1:CAS:528:DC%2BC2MXhvVWqsr3O
6. Ferraro, E; Singh, J; Patil, S; Razavi, P; Modi, S; Chandarlapaty, S et al. Incidence of brain metastases in patients with early HER2-positive breast cancer receiving neoadjuvant chemotherapy with trastuzumab and pertuzumab. Npj Breast Cancer; 2022; 8, pp. 1-8. [DOI: https://dx.doi.org/10.1038/s41523-022-00380-7]
7. Reese, RA; Lamba, N; Catalano, PJ; Cagney, DN; Wen, PY; Aizer, AA. Incidence and predictors of neurologic death in patients with brain metastases. World Neurosurgery; 2022; 162, pp. e401-e415. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/35283357][DOI: https://dx.doi.org/10.1016/j.wneu.2022.03.028]
8. Sammons, S; Leone, J; Sanglier, T; Lambert, P; Montemurro, F; Poppe, R et al. Abstract PS11–01: Brain metastases in metastatic breast cancer: prevalence per line of treatment and cumulative incidence in a cohort of 18075 real-world patients. Cancer Res; 2024; 84, pp. PS11-01. [DOI: https://dx.doi.org/10.1158/1538-7445.SABCS23-PS11-01]
9. Darlix, A; Louvel, G; Fraisse, J; Jacot, W; Brain, E; Debled, M et al. Impact of breast cancer molecular subtypes on the incidence, kinetics and prognosis of central nervous system metastases in a large multicentre real-life cohort. Br J Cancer; 2019; 121, pp. 991-1000. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/31719684][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6964671][DOI: https://dx.doi.org/10.1038/s41416-019-0619-y] 1:CAS:528:DC%2BC1MXit12ks7zO
10. Loibl, S; Mano, M; Untch, M; Huang, C-S; Mamounas, E; Wolmark, N et al. Abstract GS03–12: Phase III study of adjuvant ado-trastuzumab emtansine vs trastuzumab for residual invasive HER2-positive early breast cancer after neoadjuvant chemotherapy and HER2-targeted therapy: KATHERINE final IDFS and updated OS analysis. Cancer Research; 2024; 84, pp. GS03-12. [DOI: https://dx.doi.org/10.1158/1538-7445.SABCS23-GS03-12]
11. Pusztai L, Denkert C, O’Shaughnessy J, Cortes J, Dent R, McArthur H, et al. Event-free survival by residual cancer burden with pembrolizumab in early-stage TNBC: exploratory analysis from KEYNOTE-522 ☆. Annals of Oncology [Internet]. 2024 [cited 2024 Apr 30];0. Available from: https://www.annalsofoncology.org/article/S0923-7534(24)00046-2/fulltext
12. Lin, NU; Murthy, RK; Abramson, V; Anders, C; Bachelot, T; Bedard, PL et al. Tucatinib vs placebo, both in combination with trastuzumab and capecitabine, for previously treated erbb2 (HER2)-positive metastatic breast cancer in patients with brain metastases: updated exploratory analysis of the HER2CLIMB randomized clinical trial. JAMA Oncol; 2023; 9, pp. 197-205. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/36454580][DOI: https://dx.doi.org/10.1001/jamaoncol.2022.5610]
13. Martin, AM; Cagney, DN; Catalano, PJ; Warren, LE; Bellon, JR; Punglia, RS et al. Brain metastases in newly diagnosed breast cancer: a population-based study. JAMA Oncol; 2017; 3, pp. 1069-1077. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/28301662][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5824221][DOI: https://dx.doi.org/10.1001/jamaoncol.2017.0001]
14. Garber, HR; Raghavendra, AS; Lehner, M; Qiao, W; Gutierrez-Barrera, AM; Tripathy, D et al. Incidence and impact of brain metastasis in patients with hereditary BRCA1 or BRCA2 mutated invasive breast cancer. NPJ Breast Cancer; 2022; 8, 46. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/35393462][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8990006][DOI: https://dx.doi.org/10.1038/s41523-022-00407-z] 1:CAS:528:DC%2BB38XhtlOisrbI
15. Song, Y; Barry, WT; Seah, DS; Tung, NM; Garber, JE; Lin, NU. Patterns of recurrence and metastasis in BRCA1/BRCA2-associated breast cancers. Cancer; 2020; 126, pp. 271-280. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/31581314][DOI: https://dx.doi.org/10.1002/cncr.32540] 1:CAS:528:DC%2BB3cXhtl2jtro%3D
16. Bartsch R, Jerzak KJ, Larrouquere L, Müller V, Rhun EL. Pharmacotherapy for leptomeningeal disease in breast cancer. Cancer Treatment Reviews [Internet]. 2024 [cited 2024 Apr 17];122. Available from: https://www.cancertreatmentreviews.com/article/S0305-7372(23)00146-9/fulltext
17. Grinda, T; Joyon, N; Lusque, A; Lefèvre, S; Arnould, L; Penault-Llorca, F et al. Phenotypic discordance between primary and metastatic breast cancer in the large-scale real-life multicenter French ESME cohort. Npj Breast Cancer; 2021; 7, pp. 1-9. [DOI: https://dx.doi.org/10.1038/s41523-021-00252-6]
18. Duchnowska, R; Dziadziuszko, R; Trojanowski, T; Mandat, T; Och, W; Czartoryska-Arłukowicz, B et al. Conversion of epidermal growth factor receptor 2 and hormone receptor expression in breast cancer metastases to the brain. Breast Cancer Res; 2012; 14, R119. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/22898337][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3680944][DOI: https://dx.doi.org/10.1186/bcr3244] 1:CAS:528:DC%2BC3sXjsFSjtLg%3D
19. Pereslete, A; Hughes, M; Patterson, A; Files, J; Nguyen, K; Buckley, L et al. Abstract PS11–06: Analysis of HER2 expression changes from breast primary to brain metastases including HER2 Low and impact on overall survival. Cancer Research; 2024; 84, pp. PS11-06. [DOI: https://dx.doi.org/10.1158/1538-7445.SABCS23-PS11-06]
20. Hyun, J-W; Jeong, IH; Joung, A; Cho, HJ; Kim, S-H; Kim, HJ. Leptomeningeal metastasis: Clinical experience of 519 cases. Eur J Cancer; 2016; 56, pp. 107-114. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/26841095][DOI: https://dx.doi.org/10.1016/j.ejca.2015.12.021]
21. Sinclair, H; Le Calvez, K; Chen, J; Pakzad-Shahabi, L; Dixon, L; Mohammed, W et al. Estimating population-based incidence of brain metastases – a comprehensive incident cohort study. Neuro-Oncology; 2021; 23, iv21. [DOI: https://dx.doi.org/10.1093/neuonc/noab195.053]
22. Clarke, JL; Perez, HR; Jacks, LM; Panageas, KS; DeAngelis, LM. Leptomeningeal metastases in the MRI era. Neurology; 2010; 74, pp. 1449-1454. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/20439847][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2871005][DOI: https://dx.doi.org/10.1212/WNL.0b013e3181dc1a69] 1:STN:280:DC%2BC3czgvVyktg%3D%3D
23. Tewarie, IA; Jessurun, CAC; Hulsbergen, AFC; Smith, TR; Mekary, RA; Broekman, MLD. Leptomeningeal disease in neurosurgical brain metastases patients: A systematic review and meta-analysis. Neuro-Oncol Adv; 2021; 3, vdad62.
24. Griguolo, G; Pouderoux, S; Dieci, MV; Jacot, W; Bourgier, C; Miglietta, F et al. Clinicopathological and treatment-associated prognostic factors in patients with breast cancer leptomeningeal metastases in relation to tumor biology. Oncologist; 2018; 23, pp. 1289-1299. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/30120164][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6291333][DOI: https://dx.doi.org/10.1634/theoncologist.2018-0200] 1:CAS:528:DC%2BC1MXitlKnur0%3D
25. Morikawa, A; Jordan, L; Rozner, R; Patil, S; Boire, A; Pentsova, E et al. Characteristics and outcomes of patients with breast cancer with leptomeningeal metastasis. Clin Breast Cancer; 2017; 17, pp. 23-28. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/27569275][DOI: https://dx.doi.org/10.1016/j.clbc.2016.07.002]
26. Niwińska, A; Pogoda, K; Michalski, W; Kunkiel, M; Jagiełło-Gruszfeld, A. Determinants of prolonged survival for breast cancer patient groups with leptomeningeal metastasis (LM). J Neurooncol; 2018; 138, pp. 191-198. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/29435818][DOI: https://dx.doi.org/10.1007/s11060-018-2790-z]
27. Le Rhun, E; Taillibert, S; Zairi, F; Kotecki, N; Devos, P; Mailliez, A et al. A retrospective case series of 103 consecutive patients with leptomeningeal metastasis and breast cancer. J Neurooncol; 2013; 113, pp. 83-92. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/23456656][DOI: https://dx.doi.org/10.1007/s11060-013-1092-8]
28. Riecke K, Müller V, Neunhöffer T, Park-Simon T-W, Weide R, Polasik A, et al. Long-term survival of breast cancer patients with brain metastases: subanalysis of the BMBC registry. ESMO Open [Internet]. 2023 [cited 2024 Oct 30];8.
29. Sperduto, PW; Mesko, S; Li, J; Cagney, D; Aizer, A; Lin, NU 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 Rad Oncol Biol Phys; 2020; 107, pp. 334-43. [DOI: https://dx.doi.org/10.1016/j.ijrobp.2020.01.051]
30. Sperduto, PW; Berkey, B; Gaspar, LE; 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 Rad Oncol Biol Phys; 2008; 70, pp. 510-4. [DOI: https://dx.doi.org/10.1016/j.ijrobp.2007.06.074]
31. Li, AY; Gaebe, K; Zulfiqar, A; Lee, G; Jerzak, KJ; Sahgal, A et al. Association of brain metastases with survival in patients with limited or stable extracranial disease: a systematic review and meta-analysis. JAMA Netw Open; 2023; 6, e230475. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/36821113][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9951042][DOI: https://dx.doi.org/10.1001/jamanetworkopen.2023.0475]
32. McTyre, ER; Soike, MH; Farris, M; Ayala-Peacock, DN; Hepel, JT; Page, BR et al. Multi-institutional validation of brain metastasis velocity, a recently defined predictor of outcomes following stereotactic radiosurgery. Radiother Oncol; 2020; 142, pp. 168-174. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/31526671][DOI: https://dx.doi.org/10.1016/j.radonc.2019.08.011]
33. Soike, M; Hughes, RT; Everett, A; Marcrom, S; Farris, M; Bredel, M et al. Multi-institutional analysis of brain metastasis velocity within breast cancer receptor subtypes. Int J Radiat Oncol Biol Phys; 2020; 108, [DOI: https://dx.doi.org/10.1016/j.ijrobp.2020.07.082] e712.
34. Vogelbaum, MA; Brown, PD; Messersmith, H; Brastianos, PK; Burri, S; Cahill, D et al. Treatment for brain metastases: ASCO-SNO-ASTRO guideline. JCO; 2022; 40, pp. 492-516. [DOI: https://dx.doi.org/10.1200/JCO.21.02314] 1:CAS:528:DC%2BB38XjtVOku7o%3D
35. Le Rhun, E; Rudà, R; Devos, P; Hoang-Xuan, K; Brandsma, D; Pérez Segura, P et al. Diagnosis and treatment patterns for patients with leptomeningeal metastasis from solid tumors across Europe. J Neurooncol; 2017; 133, pp. 419-427. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/28455788][DOI: https://dx.doi.org/10.1007/s11060-017-2452-6]
36. FDA C for DE and. Cancer Clinical Trial Eligibility Criteria: Brain Metastases [Internet]. FDA; 2020 [cited 2024 Apr 19]. Available from: https://www.fda.gov/regulatory-information/search-fda-guidance-documents/cancer-clinical-trial-eligibility-criteria-brain-metastases
37. Steindl, A; Yadavalli, S; Gruber, K-A; Seiwald, M; Gatterbauer, B; Dieckmann, K et al. Neurological symptom burden impacts survival prognosis in patients with newly diagnosed non–small cell lung cancer brain metastases. Cancer; 2020; 126, pp. 4341-4352. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/32678971][DOI: https://dx.doi.org/10.1002/cncr.33085]
38. Vecht, CJ; Haaxma-Reiche, H; Noordijk, EM; Padberg, GW; Voormolen, JHC; Hoekstra, FH et al. Treatment of single brain metastasis: Radiotherapy alone or combined with neurosurgery. Ann Neurol; 1993; 33, pp. 583-590. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/8498838][DOI: https://dx.doi.org/10.1002/ana.410330605] 1:STN:280:DyaK3s3ntVClsQ%3D%3D
39. Patchell, RA; Tibbs, PA; Walsh, JW; Dempsey, RJ; Yosh, M; Kryscio, RJ et al. A Randomized Trial of Surgery in the Treatment of Single Metastases to the Brain. New England J Med; 1990; 322, pp. 494-500. [DOI: https://dx.doi.org/10.1056/NEJM199002223220802] 1:STN:280:DyaK3c7js1OlsQ%3D%3D
40. Noordijk, EM; Vecht, CJ; Haaxma-Reiche, H; Padberg, GW; Voormolen, JHC; Hoekstra, FH et al. The choice of treatment of single brain metastasis should be based on extracranial tumor activity and age. Int J Radiat Oncol Biol Phys; 1994; 29, pp. 711-717. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/8040016][DOI: https://dx.doi.org/10.1016/0360-3016(94)90558-4] 1:STN:280:DyaK2czhsVGrug%3D%3D
41. Brown, PD; Ballman, KV; Cerhan, JH; Anderson, SK; Carrero, XW; Whitton, AC 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, pp. 1049-1060. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/28687377][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5568757][DOI: https://dx.doi.org/10.1016/S1470-2045(17)30441-2]
42. Cagney, DN; Lamba, N; Sinha, S; Catalano, PJ; Bi, WL; Alexander, BM et al. Association of neurosurgical resection with development of pachymeningeal seeding in patients with brain metastases. JAMA Oncol; 2019; 5, pp. 703-709. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/30844036][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6512273][DOI: https://dx.doi.org/10.1001/jamaoncol.2018.7204]
43. Schiff, D; Messersmith, H; Brastianos, PK; Brown, PD; Burri, S; Dunn, IF et al. Radiation therapy for brain metastases: ASCO guideline endorsement of ASTRO guideline. JCO; 2022; 40, pp. 2271-2276. [DOI: https://dx.doi.org/10.1200/JCO.22.00333] 1:CAS:528:DC%2BB38XhslyrurfK
44. Kocher, M; Soffietti, R; Abacioglu, U; Villà, S; Fauchon, F; Baumert, BG et al. Adjuvant whole-brain radiotherapy versus observation after radiosurgery or surgical resection of one to three cerebral metastases: Results of the EORTC 22952–26001 Study. JCO; 2011; 29, pp. 134-141. [DOI: https://dx.doi.org/10.1200/JCO.2010.30.1655]
45. Brown, PD; Jaeckle, K; Ballman, KV; Farace, E; Cerhan, JH; Anderson, SK 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, pp. 401-409. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/27458945][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5313044][DOI: https://dx.doi.org/10.1001/jama.2016.9839]
46. Brown, PD; Pugh, S; Laack, NN; Wefel, JS; Khuntia, D; Meyers, C et al. Memantine for the prevention of cognitive dysfunction in patients receiving whole-brain radiotherapy: a randomized, double-blind, placebo-controlled trial. Neuro Oncol; 2013; 15, pp. 1429-1437. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/23956241][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3779047][DOI: https://dx.doi.org/10.1093/neuonc/not114] 1:CAS:528:DC%2BC3sXhsFegsLzE
47. Brown, PD; Gondi, V; Pugh, S; Tome, WA; Wefel, JS; Armstrong, TS et al. Hippocampal avoidance during whole-brain radiotherapy plus memantine for patients with brain metastases: Phase III Trial NRG oncology CC001. JCO; 2020; 38, pp. 1019-1029. [DOI: https://dx.doi.org/10.1200/JCO.19.02767]
48. Vellayappan, B; Lim-Fat, MJ; Kotecha, R; Salles, AD; Fariselli, L; Levivier, M et al. A systematic review informing the management of symptomatic brain radiation necrosis after stereotactic radiosurgery and international stereotactic radiosurgery society recommendations. Int J Radiat Oncol Biol Phys; 2024; 118, pp. 14-28. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/37482137][DOI: https://dx.doi.org/10.1016/j.ijrobp.2023.07.015]
49. Lebow, ES; Pike, LRG; Seidman, AD; Moss, N; Beal, K; Yu, Y. Symptomatic necrosis with antibody-drug conjugates and concurrent stereotactic radiotherapy for brain metastases. JAMA Oncol; 2023; 9, pp. 1729-1733. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/37883079][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10603573][DOI: https://dx.doi.org/10.1001/jamaoncol.2023.4492]
50. Steeg, PS. The blood–tumour barrier in cancer biology and therapy. Nat Rev Clin Oncol; 2021; 18, pp. 696-714. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/34253912][DOI: https://dx.doi.org/10.1038/s41571-021-00529-6]
51. Niwińska, A. Brain metastases as site of first and isolated recurrence of breast cancer: the role of systemic therapy after local treatment. Clin Exp Metastasis; 2016; 33, pp. 677-685. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/27225754][DOI: https://dx.doi.org/10.1007/s10585-016-9802-1]
52. Pérez-García, JM; Vaz Batista, M; Cortez, P; Ruiz-Borrego, M; Cejalvo, JM; de la Haba-Rodriguez, J et al. Trastuzumab deruxtecan in patients with central nervous system involvement from HER2-positive breast cancer: The DEBBRAH trial. Neuro Oncol; 2023; 25, pp. 157-166. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/35639825][DOI: https://dx.doi.org/10.1093/neuonc/noac144]
53. Bartsch, R; Berghoff, AS; Furtner, J; Marhold, M; Bergen, ES; Roider-Schur, S et al. FINAL outcome analysis from the phase ii tuxedo-1 trial of trastuzumab-deruxtecan in HER2-positive breast cancer patients with active brain metastases. Neuro-Oncology; 2024; 26, noae23.
54. Jerusalem, G; Park, YH; Yamashita, T; Hurvitz, SA; Modi, S; Andre, F et al. Trastuzumab Deruxtecan in HER2-positive metastatic breast cancer patients with brain metastases: A DESTINY-Breast01 subgroup analysis. Cancer Discov; 2022; 12, pp. 2754-2762. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/36255231][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9716244][DOI: https://dx.doi.org/10.1158/2159-8290.CD-22-0837] 1:CAS:528:DC%2BB3sXivVarsbg%3D
55. Hurvitz SA, Kim S-B, Chung W-P, Im S-A, Park YH, Hegg R, et al. Trastuzumab deruxtecan versus trastuzumab emtansine in HER2-positive metastatic breast cancer patients with brain metastases from the randomized DESTINY-Breast03 trial. ESMO Open [Internet]. 2024 [cited 2024 Apr 24];0. Available from: https://www.esmoopen.com/article/S2059-7029(24)00692-6/fulltext
56. Hurvitz, SA; Modi, S; Li, W; Park, YH; Chung, W; Kim, S-B et al. 377O A pooled analysis of trastuzumab deruxtecan (T-DXd) in patients (pts) with HER2-positive (HER2+) metastatic breast cancer (mBC) with brain metastases (BMs) from DESTINY-Breast (DB) -01, -02, and -03. Ann Oncol; 2023; 34, pp. S335-S336. [DOI: https://dx.doi.org/10.1016/j.annonc.2023.09.554]
57. Harbeck N, Ciruelos E, Jerusalem G, Müller V, Niikura N, Viale G, et al. Trastuzumab deruxtecan in HER2-positive advanced breast cancer with or without brain metastases: a phase 3b/4 trial. Nat Med. 2024;1–10.
58. Montemurro, F; Delaloge, S; Barrios, CH; Wuerstlein, R; Anton, A; Brain, E 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, pp. 1350-1358. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/32634611][DOI: https://dx.doi.org/10.1016/j.annonc.2020.06.020] 1:STN:280:DC%2BB38npsV2isQ%3D%3D
59. Lin, NU; Kumthekar, P; Sahebjam, S; Ibrahim, N; Fung, A; Cheng, A et al. Pertuzumab plus high-dose trastuzumab for HER2-positive breast cancer with brain metastases: PATRICIA final efficacy data. Npj Breast Cancer; 2023; 9, pp. 1-10. [DOI: https://dx.doi.org/10.1038/s41523-023-00587-2] 1:CAS:528:DC%2BB3sXhslOmt7zN
60. Lin, NU; Diéras, V; Paul, D; Lossignol, D; Christodoulou, C; Stemmler, H-J et al. Multicenter phase II study of lapatinib in patients with brain metastases from HER2-positive breast cancer. Clin Cancer Res; 2009; 15, pp. 1452-1459. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/19228746][DOI: https://dx.doi.org/10.1158/1078-0432.CCR-08-1080] 1:CAS:528:DC%2BD1MXitVOktLo%3D
61. Bachelot, T; Romieu, G; Campone, M; Diéras, V; Cropet, C; Dalenc, F 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, pp. 64-71. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/23122784][DOI: https://dx.doi.org/10.1016/S1470-2045(12)70432-1] 1:CAS:528:DC%2BC3sXhsV2msQ%3D%3D
62. de Azambuja, E; Zardavas, D; Lemort, M; Rossari, J; Moulin, C; Buttice, A et al. Phase I trial combining temozolomide plus lapatinib for the treatment of brain metastases in patients with HER2-positive metastatic breast cancer: the LAPTEM trial. Ann Oncol; 2013; 24, pp. 2985-2989. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/24013582][DOI: https://dx.doi.org/10.1093/annonc/mdt359]
63. Lin, NU; Freedman, RA; Ramakrishna, N; Younger, J; Storniolo, AM; Bellon, JR 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, pp. 405-414. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/24197661][DOI: https://dx.doi.org/10.1007/s10549-013-2754-0] 1:CAS:528:DC%2BC3sXhslGlsbjN
64. Kim, IA; Winter, KA; Sperduto, PW; De Los Santos, JF; Peereboom, DM; Ogunleye, T et al. Concurrent Lapatinib with brain radiation therapy in patients with HER2+ breast cancer with brain metastases: NRG Oncology–KROG/RTOG 1119 Phase 2 Randomized Trial. Int J Rad Oncol Biol Phys; 2024; 118, pp. 1391-401. [DOI: https://dx.doi.org/10.1016/j.ijrobp.2023.07.019]
65. Freedman, RA; Gelman, RS; Wefel, JS; Melisko, ME; Hess, KR; Connolly, RM et al. Translational breast cancer research consortium (TBCRC) 022: A Phase II trial of neratinib for patients with human epidermal growth factor receptor 2–positive breast cancer and brain metastases. JCO; 2016; 34, pp. 945-952. [DOI: https://dx.doi.org/10.1200/JCO.2015.63.0343] 1:CAS:528:DC%2BC2sXnvFOhsg%3D%3D
66. Freedman, RA; Gelman, RS; Anders, CK; Melisko, ME; Parsons, HA; Cropp, AM 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. JCO; 2019; 37, pp. 1081-1089. [DOI: https://dx.doi.org/10.1200/JCO.18.01511] 1:CAS:528:DC%2BC1MXhs1WntLbP
67. Freedman, R; Ren, S; Tayob, N; Gelman, R; Smith, KL; Davis, R et al. Abstract PD7–03: translational breast cancer research consortium trial 022: Neratinib and Trastuzumab-Emtansine for HER2+ Breast Cancer Brain Metastases (BCBM). Cancer Research; 2023; 83, pp. PD7-03. [DOI: https://dx.doi.org/10.1158/1538-7445.SABCS22-PD7-03]
68. Hurvitz, SA; Saura, C; Oliveira, M; Trudeau, ME; Moy, B; Delaloge, S et al. Efficacy of neratinib plus capecitabine in the subgroup of patients with central nervous system involvement from the NALA Trial. Oncologist; 2021; 26, pp. e1327-e1338. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/34028126][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8342591][DOI: https://dx.doi.org/10.1002/onco.13830] 1:CAS:528:DC%2BB3MXhs1anurbJ
69. Lin, NU; Borges, V; Anders, C; Murthy, RK; Paplomata, E; Hamilton, E 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. JCO; 2020; 38, pp. 2610-2619. [DOI: https://dx.doi.org/10.1200/JCO.20.00775] 1:CAS:528:DC%2BB3cXitVChu77L
70. Yang, Z; Meng, J; Mei, X; Mo, M; Xiao, Q; Han, X et al. Brain Radiotherapy With Pyrotinib and Capecitabine in Patients With ERBB2-Positive Advanced Breast Cancer and Brain Metastases: A Nonrandomized Phase 2 Trial. JAMA Oncol; 2024; 10, pp. 335-341. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/38175627][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10767641][DOI: https://dx.doi.org/10.1001/jamaoncol.2023.5791]
71. Tolaney, SM; Sahebjam, S; Le Rhun, E; Bachelot, T; Kabos, P; Awada, A 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, pp. 5310-5319. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/32694159][DOI: https://dx.doi.org/10.1158/1078-0432.CCR-20-1764] 1:CAS:528:DC%2BB3cXisFCku73E
72. Leone, JP; Emblem, KE; Weitz, M; Gelman, RS; Schneider, BP; Freedman, RA et al. Phase II trial of carboplatin and bevacizumab in patients with breast cancer brain metastases. Breast Cancer Res; 2020; 22, 131. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/33256829][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7706261][DOI: https://dx.doi.org/10.1186/s13058-020-01372-w] 1:CAS:528:DC%2BB3cXis12isbfL
73. Lu, Y-S; Chen, TW-W; Lin, C-H; Yeh, D-C; Tseng, L-M; Wu, P-F 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, pp. 1851-8. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/25700303][DOI: https://dx.doi.org/10.1158/1078-0432.CCR-14-2075] 1:CAS:528:DC%2BC2MXmsl2gt7c%3D
74. Tsurutani, J; Jacot, W; Yamashita, T; Riaz, F; Yerushalmi, R; Im, S-A et al. 388P Subgroup analysis of patients (pts) with HER2-low metastatic breast cancer (mBC) with brain metastases (BMs) at baseline from DESTINY-Breast04, a randomized phase III study of trastuzumab deruxtecan (T-DXd) vs treatment of physician’s choice (TPC). Ann Oncol; 2023; 34, pp. S342-S343. [DOI: https://dx.doi.org/10.1016/j.annonc.2023.09.565]
75. Hurvitz, SA; Bardia, A; Punie, K; Kalinsky, K; Carey, LA; Rugo, HS et al. Subgroup analyses from the phase 3 ASCENT study of sacituzumab govitecan in metastatic triple-negative breast cancer. Npj Breast Cancer; 2024; 10, pp. 1-11.
76. Chen, TW-W; Dai, M-S; Tseng, L-M; Chen, S-C; Chao, T-Y; Chao, T-C et al. Whole-brain radiotherapy alone vs preceded by bevacizumab, etoposide, and cisplatin for untreated brain metastases from breast cancer: a randomized clinical trial. JAMA Oncology; 2024; 10, pp. 325-34. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/38127335][DOI: https://dx.doi.org/10.1001/jamaoncol.2023.5456]
77. Rivera, E; Meyers, C; Groves, M; Valero, V; Francis, D; Arun, B et al. Phase I study of capecitabine in combination with temozolomide in the treatment of patients with brain metastases from breast carcinoma. Cancer; 2006; 107, pp. 1348-1354. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/16909414][DOI: https://dx.doi.org/10.1002/cncr.22127] 1:CAS:528:DC%2BD28XhtVOns73K
78. Franciosi, V; Cocconi, G; Michiara, M; Di Costanzo, F; Fosser, V; Tonato, M et al. Front-line chemotherapy with cisplatin and etoposide for patients with brain metastases from breast carcinoma, nonsmall cell lung carcinoma, or malignant melanoma: a prospective study. Cancer; 1999; 85, pp. 1599-1605. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/10193952][DOI: https://dx.doi.org/10.1002/(SICI)1097-0142(19990401)85:7<1599::AID-CNCR23>3.0.CO;2-#] 1:CAS:528:DyaK1MXitlaqs7g%3D
79. Christodoulou, C; Bafaloukos, D; Linardou, H; Aravantinos, G; Bamias, A; Carina, M et al. Temozolomide (TMZ) combined with cisplatin (CDDP) in patients with brain metastases from solid tumors: a Hellenic Cooperative Oncology Group (HeCOG) Phase II study. J Neurooncol; 2005; 71, pp. 61-65. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/15719277][DOI: https://dx.doi.org/10.1007/s11060-004-9176-0] 1:CAS:528:DC%2BD2MXhtlCjsLfK
80. Swain, SM; Miles, D; Kim, S-B; Im, Y-H; Im, S-A; Semiglazov, V 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, pp. 519-530. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/32171426][DOI: https://dx.doi.org/10.1016/S1470-2045(19)30863-0] 1:CAS:528:DC%2BB3cXkvFCmu7o%3D
81. Swain, SM; Baselga, J; Miles, D; Im, Y-H; Quah, C; Lee, LF et al. 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, pp. 1116-1121. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/24685829][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4037862][DOI: https://dx.doi.org/10.1093/annonc/mdu133] 1:STN:280:DC%2BC2crovF2kug%3D%3D
82. Bergen, ES; Binter, A; Starzer, AM; Heller, G; Kiesel, B; Tendl-Schulz, K et al. Favourable outcome of patients with breast cancer brain metastases treated with dual HER2 blockade of trastuzumab and pertuzumab. Ther Adv Med Oncol; 2021; 13, 17588359211009002. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/33995593][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8072867][DOI: https://dx.doi.org/10.1177/17588359211009002] 1:CAS:528:DC%2BB3MXhsFCnsrzL
83. Bartsch, R; Berghoff, AS; Furtner, J; Marhold, M; Bergen, ES; Roider-Schur, S et al. Trastuzumab deruxtecan in HER2-positive breast cancer with brain metastases: a single-arm, phase 2 trial. Nat Med; 2022; 28, pp. 1840-1847. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/35941372][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9499862][DOI: https://dx.doi.org/10.1038/s41591-022-01935-8] 1:CAS:528:DC%2BB38XitV2lsLbN
84. Sammons S, Lin NU. Antibody–drug conjugates are active in patients with HER2-positive breast cancer brain metastases: where do we go from here? ESMO Open [Internet]. 2024 [cited 2024 Apr 25];0. Available from: https://www.esmoopen.com/article/S2059-7029(24)00758-0/fulltext
85. Lin, NU; Pegram, M; Sahebjam, S; Ibrahim, N; Fung, A; Cheng, A et al. Pertuzumab plus high-dose trastuzumab in patients with progressive brain metastases and HER2-positive metastatic breast cancer: primary analysis of a phase II study. JCO; 2021; 39, pp. 2667-2675. [DOI: https://dx.doi.org/10.1200/JCO.20.02822] 1:CAS:528:DC%2BB38XjtVOrtL8%3D
86. Cottu, P; Ring, A; Abdel-Razeq, H; Marchetti, P; Cardoso, F; Salvador Bofill, J et al. Ribociclib plus letrozole in subgroups of special clinical interest with hormone receptor–positive, human epidermal growth factor receptor 2–negative advanced breast cancer: Subgroup analysis of the phase IIIb CompLEEment-1 trial. Breast; 2022; 62, pp. 75-83. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/35131646][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9073296][DOI: https://dx.doi.org/10.1016/j.breast.2022.01.016]
87. Bardia, A; Jhaveri, K; Im, S-A; De Laurentiis, M; Xu, B; Pernas, S et al. Abstract GS02–01: Randomized phase 3 study of datopotamab deruxtecan vs chemotherapy for patients with previously-treated inoperable or metastatic hormone receptor-positive, HER2-negative breast cancer: Results from TROPION-Breast01. Cancer Research; 2024; 84, pp. GS02-01. [DOI: https://dx.doi.org/10.1158/1538-7445.SABCS23-GS02-01]
88. Balinda, HU; Kelly, WJ; Kaklamani, VG; Lathrop, KI; Canola, MM; Ghamasaee, P et al. Sacituzumab Govitecan in patients with breast cancer brain metastases and recurrent glioblastoma: a phase 0 window-of-opportunity trial. Nat Commun; 2024; 15, 6707. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/39112464][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11306739][DOI: https://dx.doi.org/10.1038/s41467-024-50558-9] 1:CAS:528:DC%2BB2cXhslKnsLzE
89. Jusino, S; Fadul, CE; Dillon, P. Systematic review of the management of brain metastases from hormone receptor positive breast cancer. J Neurooncol; 2023; 162, pp. 45-57. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/36884200][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10049940][DOI: https://dx.doi.org/10.1007/s11060-023-04276-9]
90. Brastianos, PK; Kim, AE; Giobbie-Hurder, A; Lee, EQ; Lin, NU; Overmoyer, B et al. Pembrolizumab in brain metastases of diverse histologies: phase 2 trial results. Nat Med; 2023; 29, pp. 1728-1737. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/37268724][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10644912][DOI: https://dx.doi.org/10.1038/s41591-023-02392-7] 1:CAS:528:DC%2BB3sXhtFGnu73O
91. Epaillard, N; Lusque, A; Pistilli, B; André, F; Bachelot, T; Pierga, J-Y et al. 260P Antitumor activity of trastuzumab deruxtecan (T-DXd) in patients with metastatic breast cancer (mBC) and brain metastases (BMs) from DAISY trial. Ann Oncol; 2022; 33, S656. [DOI: https://dx.doi.org/10.1016/j.annonc.2022.07.299]
92. Luitse, MJA; Boles, G; Sonke, GS; Brandsma, D. P11.61.B Capecitabine treatment of CNS metastases from breast cancer: intracranial response and survival. Neuro-Oncology; 2022; 24, 72. [DOI: https://dx.doi.org/10.1093/neuonc/noac174.250]
93. Linot, B; Campone, M; Augereau, P; Delva, R; Abadie-Lacourtoisie, S; Nebout-Mesgouez, N et al. Use of liposomal doxorubicin–cyclophosphamide combination in breast cancer patients with brain metastases: a monocentric retrospective study. J Neurooncol; 2014; 117, pp. 253-259. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/24481998][DOI: https://dx.doi.org/10.1007/s11060-014-1378-5] 1:CAS:528:DC%2BC2cXhsl2it7k%3D
94. Sachdev, JC; Munster, P; Northfelt, DW; Han, HS; Ma, C; Maxwell, F et al. Phase I study of liposomal irinotecan in patients with metastatic breast cancer: findings from the expansion phase. Breast Cancer Res Treat; 2021; 185, pp. 759-771. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/33201358][DOI: https://dx.doi.org/10.1007/s10549-020-05995-7] 1:CAS:528:DC%2BB3cXisV2gsbfJ
95. Sabatier, R; Martin, J; Vicier, C; Guérin, M; Monneur, A; Provansal, M et al. Eribulin efficacy on brain metastases in heavily pretreated patients with metastatic breast cancer. J Clin Med; 2021; 10, 1272. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/33803894][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8003126][DOI: https://dx.doi.org/10.3390/jcm10061272] 1:CAS:528:DC%2BB3MXhtFyitbvJ
96. Siena, S; Crinò, L; Danova, M; Prete, SD; Cascinu, S; Salvagni, S et al. Dose-dense temozolomide regimen for the treatment of brain metastases from melanoma, breast cancer, or lung cancer not amenable to surgery or radiosurgery: a multicenter phase II study. Ann Oncol; 2010; 21, pp. 655-661. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/19767314][DOI: https://dx.doi.org/10.1093/annonc/mdp343] 1:STN:280:DC%2BC3c7isVGmsA%3D%3D
97. Exman, P; Mallery, RM; Lin, NU; Parsons, HA. Response to Olaparib in a patient with germline BRCA2 mutation and breast cancer leptomeningeal carcinomatosis. Npj Breast Cancer; 2019; 5, pp. 1-5. [DOI: https://dx.doi.org/10.1038/s41523-019-0139-1]
98. Rhun EL, Weller M, Bent M van den, Brandsma D, Furtner J, Rudà R, et al. Leptomeningeal metastasis from solid tumours: EANO–ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up☆. ESMO Open [Internet]. 2023 [cited 2023 Oct 4];0. Available from: https://www.esmoopen.com/article/S2059-7029(23)00859-1/fulltext#%20
99. Wolf, A; Donahue, B; Silverman, JS; Chachoua, A; Lee, JK; Kondziolka, D. Stereotactic radiosurgery for focal leptomeningeal disease in patients with brain metastases. J Neurooncol; 2017; 134, pp. 139-143. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/28536993][DOI: https://dx.doi.org/10.1007/s11060-017-2497-6]
100. Yang JT, Wijetunga NA, Pentsova E, Wolden S, Young RJ, Correa D, et al. Randomized Phase II Trial of Proton Craniospinal Irradiation Versus Photon Involved-Field Radiotherapy for Patients With Solid Tumor Leptomeningeal Metastasis. J Clin Oncol [Internet]. 2022
101. Le Rhun, E; Wallet, J; Mailliez, A; Le Deley, MC; Rodrigues, I; Boulanger, T et al. Intrathecal liposomal cytarabine plus systemic therapy versus systemic chemotherapy alone for newly diagnosed leptomeningeal metastasis from breast cancer. Neuro Oncol; 2020; 22, pp. 524-538. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/31637444][DOI: https://dx.doi.org/10.1093/neuonc/noz201]
102. Jaeckle, KA; Dixon, JG; Anderson, SK; Moreno-Aspitia, A; Colon-Otero, G; Hebenstreit, K et al. Intra-CSF topotecan in treatment of breast cancer patients with leptomeningeal metastases. Cancer Med; 2020; 9, pp. 7935-7942. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/32885617][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7643683][DOI: https://dx.doi.org/10.1002/cam4.3422] 1:CAS:528:DC%2BB3cXitlWhtL%2FF
103. Kumthekar, PU; Avram, MJ; Lassman, AB; Lin, NU; Lee, E; Grimm, SA et al. A phase I/II study of intrathecal trastuzumab in human epidermal growth factor receptor 2-positive (HER2-positive) cancer with leptomeningeal metastases: Safety, efficacy, and cerebrospinal fluid pharmacokinetics. Neuro Oncol; 2023; 25, pp. 557-565. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/35948282][DOI: https://dx.doi.org/10.1093/neuonc/noac195] 1:CAS:528:DC%2BB3sXhs1Wmur3O
104. Bonneau, C; Paintaud, G; Trédan, O; Dubot, C; Desvignes, C; Dieras, V et al. Phase I feasibility study for intrathecal administration of trastuzumab in patients with HER2 positive breast carcinomatous meningitis. Eur J Cancer; 2018; 95, pp. 75-84. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/29635147][DOI: https://dx.doi.org/10.1016/j.ejca.2018.02.032] 1:CAS:528:DC%2BC1cXntF2ltrk%3D
105. Alder, L; Trapani, D; Bradbury, C; Van Swearingen, AED; Tolaney, SM; Khasraw, M et al. Durable responses in patients with HER2+ breast cancer and leptomeningeal metastases treated with trastuzumab deruxtecan. Npj Breast Cancer; 2023; 9, pp. 1-5. [DOI: https://dx.doi.org/10.1038/s41523-023-00519-0]
106. Niikura, N; Yamanaka, T; Nomura, H; Shiraishi, K; Kusama, H; Yamamoto, M et al. Treatment with trastuzumab deruxtecan in patients with HER2-positive breast cancer and brain metastases and/or leptomeningeal disease (ROSET-BM). Npj Breast Cancer; 2023; 9, pp. 1-8. [DOI: https://dx.doi.org/10.1038/s41523-023-00584-5]
107. Murthy, RK; O’Brien, B; Berry, DA; Singareeka-Raghavendra, A; Monroe, MG; Johnson, J et al. Abstract PD4–02: Safety and efficacy of a tucatinib-trastuzumab-capecitabine regimen for treatment of leptomeningeal metastasis (LM) in HER2-positive breast cancer: Results from TBCRC049, a phase 2 non-randomized study. Cancer Research; 2022; 82, pp. PD4-02. [DOI: https://dx.doi.org/10.1158/1538-7445.SABCS21-PD4-02]
108. O’Brien, BJ; Murthy, RK; Berry, DA; Singareeka Raghavendra, A; Gule-Monroe, M; Johnson, JM et al. Tucatinib-trastuzumab-capecitabine for treatment of leptomeningeal metastasis in HER2+ breast cancer: TBCRC049 phase 2 study results. JCO; 2024; 42, pp. 2018-2018. [DOI: https://dx.doi.org/10.1200/JCO.2024.42.16_suppl.2018]
109. Rhun, EL; Guckenberger, M; Smits, M; Dummer, R; Bachelot, T; Sahm, F et al. EANO–ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up of patients with brain metastasis from solid tumours☆. Ann Oncol; 2021; 32, pp. 1332-1347. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/34364998][DOI: https://dx.doi.org/10.1016/j.annonc.2021.07.016]
110. Nayak, L; DeAngelis, LM; Brandes, AA; Peereboom, DM; Galanis, E; Lin, NU et al. The neurologic assessment in neuro-oncology (NANO) scale: a tool to assess neurologic function for integration into the Response Assessment in Neuro-Oncology (RANO) criteria. Neuro Oncol; 2017; 19, pp. 625-635. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/28453751][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5464449][DOI: https://dx.doi.org/10.1093/neuonc/nox029]
111. Galldiks, N; Langen, K-J; Albert, NL; Chamberlain, M; Soffietti, R; Kim, MM et al. PET imaging in patients with brain metastasis—report of the RANO/PET group. Neuro Oncol; 2019; 21, pp. 585-595. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/30615138][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6502495][DOI: https://dx.doi.org/10.1093/neuonc/noz003]
112. Rock, JP; Scarpace, L; Hearshen, D; Gutierrez, J; Fisher, JL; Rosenblum, M et al. Associations among magnetic resonance spectroscopy, apparent diffusion coefficients, and image-guided histopathology with special attention to radiation necrosis. Neurosurgery; 2004; 54, 1111. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/15113465][DOI: https://dx.doi.org/10.1227/01.NEU.0000119328.56431.A7]
113. Horky, LL; Hsiao, EM; Weiss, SE; Drappatz, J; Gerbaudo, VH. Dual phase FDG-PET imaging of brain metastases provides superior assessment of recurrence versus post-treatment necrosis. J Neurooncol; 2011; 103, pp. 137-146. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/20838854][DOI: https://dx.doi.org/10.1007/s11060-010-0365-8]
114. Lin, NU; Lee, EQ; Aoyama, H; Barani, IJ; Barboriak, DP; Baumert, BG et al. Response assessment criteria for brain metastases: proposal from the RANO group. Lancet Oncol; 2015; 16, pp. e270-e278. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/26065612][DOI: https://dx.doi.org/10.1016/S1470-2045(15)70057-4]
115. Chamberlain, M; Junck, L; Brandsma, D; Soffietti, R; Rudà, R; Raizer, J et al. Leptomeningeal metastases: a RANO proposal for response criteria. Neuro Oncol; 2017; 19, pp. 484-492. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/28039364]
116. Olson, EM; Abdel-Rasoul, M; Maly, J; Wu, CS; Lin, NU; Shapiro, CL. Incidence and risk of central nervous system metastases as site of first recurrence in patients with HER2-positive breast cancer treated with adjuvant trastuzumab. Ann Oncol; 2013; 24, pp. 1526-1533. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/23463626][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3660080][DOI: https://dx.doi.org/10.1093/annonc/mdt036] 1:STN:280:DC%2BC3svjsFamuw%3D%3D
117. Piccart, M; Procter, M; Fumagalli, D; de Azambuja, E; Clark, E; Ewer, MS et al. Adjuvant Pertuzumab and Trastuzumab in Early HER2-Positive Breast Cancer in the APHINITY Trial: 6 Years’ Follow-Up. JCO; 2021; 39, pp. 1448-1457. [DOI: https://dx.doi.org/10.1200/JCO.20.01204] 1:CAS:528:DC%2BB38XhslalsLs%3D
118. Chan, A; Moy, B; Mansi, J; Ejlertsen, B; Holmes, FA; Chia, S et al. Final Efficacy Results of Neratinib in HER2-positive Hormone Receptor-positive Early-stage Breast Cancer From the Phase III ExteNET Trial. Clin Breast Cancer; 2021; 21, pp. 80-91.e7. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/33183970][DOI: https://dx.doi.org/10.1016/j.clbc.2020.09.014] 1:CAS:528:DC%2BB3cXitFCltrbE
119. Shi, Y; Wang, P; Zhu, Y; Wang, X; Li, S; Zhang, B et al. 401P Utidelone in combination with etoposide and bevacizumab in HER2-negative breast cancer with brain metastasis (UTOBIA-BM): A prospective, single-arm, phase II trial. Ann Oncol; 2023; 34, pp. S349-S350. [DOI: https://dx.doi.org/10.1016/j.annonc.2023.09.578]
120. Kumthekar, P; Tang, S-C; Brenner, AJ; Kesari, S; Piccioni, DE; Anders, C 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, pp. 2789-2799. [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/31969331][DOI: https://dx.doi.org/10.1158/1078-0432.CCR-19-3258] 1:CAS:528:DC%2BB3cXitVyktLjP
© The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature 2025.