Skip to main content
Chronic Diseases and Translational Medicine logoLink to Chronic Diseases and Translational Medicine
. 2017 Mar 8;3(1):21–32. doi: 10.1016/j.cdtm.2017.01.004

Management of breast cancer brain metastases: Focus on human epidermal growth factor receptor 2-positive breast cancer

Peng Yuan 1,, Song-Lin Gao 1
PMCID: PMC5627687  PMID: 29063053

Abstract

After the introduction of trastuzumab, a monoclonal antibody that binds to human epidermal growth factor receptor 2 (HER2), the overall survival (OS) among patients with HER2-positive breast cancer has been substantially improved. However, among these patients, the incidence of brain metastases (BM) has been increasing and an increased proportion of them have died of intracranial progression, which makes HER2-positive breast cancer brain metastases (BCBM) a critical issue of concern. For local control of limited BM, stereotactic radiosurgery (SRS) and surgical resection are available modalities with different clinical indications. Postoperative or preoperative radiation is usually delivered in conjunction with surgical resection to boost local control. Adjuvant whole-brain radiotherapy (WBRT) should be deferred for limited BM because of its impairment of neurocognitive function while having no benefit for OS. Although WBRT is still the standard treatment for local control of diffuse BM, SRS is a promising treatment for diffuse BM as the technique continues to improve. Although large molecules have difficulty crossing the blood brain barrier, trastuzumab-containing regimens are critical for treating HER2-positive BCBM patients because they significantly prolong OS. Tyrosine kinase inhibitors are more capable of crossing into the brain and they have been shown to be beneficial for treating BM in HER2-positive patients, especially lapatinib combined with capecitabine. The antiangiogenic agent, bevacizumab, can be applied in the HER2-positive BCBM scenario as well. In this review, we also discuss several strategies for delivering drugs into the central nervous system and several microRNAs that have the potential to become biomarkers of BCBM.

Keywords: Breast cancer brain metastases, Human epidermal growth factor receptor 2-positive breast cancer, Local control, Targeted therapy, MicroRNA

Introduction

Breast cancer is the second-leading cause of central nervous system (CNS) metastases among solid malignancies.1 The incidence of developing brain metastases (BM) has been reported to range from 10% to 16% among advanced breast cancer patients,2 and autopsy studies indicate that this figure may underestimate the true incidence since another 10% of BM are asymptomatic and not diagnosed before death.3 Patients with human epidermal growth factor receptor 2 (HER2)-positive cancer or triple negative breast cancer (TNBC) have a higher risk of developing BM than patients with luminal-like disease.4, 5, 6 Several studies have shown that HER2-positivity is associated with a biological propensity to metastasize to the brain.7 After the introduction of trastuzumab, which has significantly improved overall survival (OS) among patients with HER2-positive breast cancer, the incidence of BM among HER2-positive patients (ranging from 30% to 55%5, 8, 9, 10, 11, 12) has been increasing.10, 13, 14, 15 Unlike BM in TNBC, which often develops with concurrent extracranial disease progression,16 BM often occurs in a setting of stable extracranial control among HER2-positive patients.17

In the past, even after treatment by whole-brain radiotherapy (WBRT), the median survival of patients with breast cancer brain metastases (BCBM) was poor, ranging from 3 to 6 months.18 Before the trastuzumab era, the OS was shorter among patients with HER2-positive brain metastases compared with those with HER2-negative disease, which was mainly attributed to the progression of systemic disease.19 After the introduction of effective anti-HER2 therapy, survival after diagnosis of BM among HER2-positive patients has been significantly improved compared with that among patients with HER2-negative disease, mainly due to the improvement of extracranial disease control.20, 21 Several retrospective studies have reported that the median OS after diagnosis of BM is around 2 years for HER2-positive patients.20, 21, 22, 23 Meanwhile, with the OS significantly prolonged, the proportion of people dying of cerebral progression has been increasing. A retrospective study reported that up to 50% of HER2-positive patients died of cerebral progression,10 which makes BM among HER2-positive patients a critical issue. To improve management, the American Society of Clinical Oncology (ASCO) published a guideline focusing on this issue in 2014.24

In this review, we will discuss treatments for HER2-positive BCBM, including local treatment and targeted therapy. In addition, several cancer biomarkers for BCBM will also be discussed.

Local control

Management of limited BM (1–4 BM)

Surgery

In order to achieve long-lasting control, surgical resection is a standard treatment for patients with a favorable prognosis and a solitary lesion, especially a large lesion (over 3–4 cm). There were several randomized control trials conducted to define the role of surgical resection in solitary BM, and they demonstrated a significant survival benefit for patients receiving surgical resection.25, 26, 27, 28 Surgical resection is also used for immediate mass effect relief in patients with limited BM (2–4 lesions) who have a large lesion causing neurologic symptoms; however, the effect of surgery on survival of these patients with limited BM is still unknown. Since there is a high recurrence rate after surgical resection,29 postoperative radiation is usually recommended to improve local control, which will be discussed in the postoperative and preoperative radiation therapy section.

Stereotactic radiosurgery

Stereotactic radiosurgery (SRS) is a radiation therapy technique using intersected beams to deliver a highly conformal and high dose of radiation to a target volume in order to produce an ablative effect with minimal damage to surrounding normal tissues. SRS is usually delivered in a single fraction, but it can also be delivered in multiple fractions [fractionated stereotactic radiotherapy (FSRT)]. For local control of BM, SRS can be used as a therapy alone, a boost after WBRT, or an adjuvant treatment preoperatively or postoperatively.30

There are different techniques available for stereotactic radiosurgery including Gamma Knife® (GK) and CyberKnife (CK). GK used to be the standard device for SRS. It is based on an invasive head frame system coupled with cobalt-60 sources and is mainly used for intracranial indications,31, 32 while CK is based on a linear accelerator system without head fixation and can be used for both intra- and extracranial lesions.33 Because it is frameless and has a wider range of indications, CK has become increasingly popular over the last few decades and has been shown to not be inferior to GK in the accuracy of dose delivery.34, 35 In dosimetry, CK shows a more homogeneous dose distribution across the entire lesion,36, 37 while GK shows an inhomogeneous distribution with a higher dose in the center of the lesion36, 38 that may help to minimize local tumor recurrence.39 However, a matched-pair analysis demonstrated that the obvious differences in treatment-related parameters between GK and CK had no effect on clinical outcomes after radiosurgery.40

Although both SRS and surgery can treat patients with limited BM, there is no prospective randomized trial comparing these two modalities. Actually, they are not competitive modalities in most cases, but are prescribed for different clinical indications. The choice of modality usually depends on the size of the lesion, the surgical accessibility, symptoms, and the status of the patient. Surgery can alleviate the mass effect of large lesions immediately and has advantages in treating lesions adjacent to critical structures or lesions over 4 cm in size. SRS is an alternative treatment for inoperable patients with limited BM with lesions 4 cm or less. SRS has several advantages over surgery including being noninvasive, able to treat several lesions simultaneously, and it can treat surgically inaccessible lesions, such as lesions in a deep area.41

SRS combined with WBRT or SRS alone as primary treatment has been shown to have an excellent rate of local control for patients with limited BM.42, 43 Because of the decline of neurocognitive function and quality of life (QoL) associated with WBRT,44, 45 several trials were conducted to determine whether WBRT could be omitted after SRS for patients with limited BM. Most of them demonstrated that the addition of WBRT to SRS significantly increased local control, but had no beneficial effect on OS.42, 46 Based on these findings, the Choosing Wisely List published by the American Society for Radiation Oncology (ASTRO) in September 2014 recommended that adjuvant WBRT should not be routinely added to SRS for patients with limited BM.47 However, this recommendation provoked a lot of debate. Several researchers thought that there were not enough patients involved in these randomized trials to demonstrate WBRT's effect on OS, since several trials consistently showed that adjuvant WBRT significantly improved intracranial control and reduced the rate of neurologic causes of death. They argued that more data about adjuvant WBRT should be collected to reevaluate this scenario.48

Postoperative and preoperative radiation therapy

Because of the high recurrence rate after surgical resection,29, 46 postoperative radiation is usually recommended as a boost for intracranial control. Adjuvant WBRT is the standard treatment in the postoperative setting.49 Multiple studies have demonstrated that postoperative WBRT can significantly reduce the risk of local recurrence, distant brain recurrence, and neurologic causes of death, but has no benefit for OS.29, 46 Because of potential toxicity associated with WBRT, clinicians also use postoperative SRS to defer WBRT and to improve intracranial control. Because of the size limitation of SRS, FSRT has been used for large surgical cavities over 3 cm and has a similar control rate as SRS.50 Several trials demonstrated that SRS/FSRT as a postoperative treatment alone provided acceptable local control,50, 51, 52, 53, 54 and the addition of a 2 mm margin around the resection cavity showed an improved local control compared to the technique with no margin.55

A retrospective study demonstrated that compared with WBRT, SRS alone administered to the surgical cavity showed a similar local recurrence rate, a higher rate of leptomeningeal dissemination (LMD), and an inferior distant brain control.56 Interestingly, a study reported that patients with breast cancer histology were at a higher risk of developing LMD (at 1 year, 24% vs. 9%) after postoperative SRS as compared with patients with non-breast cancer histology, but whether the rate of LMD is inherently higher with breast histology or the inclusion of WBRT could decrease this risk of LMD is unknown.57 A prospective randomized trial (NCT01372774) conducted by the Alliance for Clinical Trials in Oncology (N107C) is ongoing to determine the role of WBRT versus SRS in the postoperative setting.

With a clear target volume definition, preoperative SRS may be a promising treatment to reduce LMD and radiation necrosis (RN) caused by postoperative SRS.58 Despite limited supporting data, preoperative SRS has been increasingly used in recent years.30 In a multi-institutional analysis with 180 patients receiving preoperative SRS or postoperative SRS, Patel et al59 demonstrated a significantly lower rate of symptomatic RN (16.4% vs. 4.9%; P < 0.010) and LMD (16.6% vs. 3.2%; P < 0.010) in the preoperative SRS arm, but showed similar rates of local recurrence, distant brain recurrence, and OS between both arms.

Management of diffuse BM

Whole-brain radiation therapy

WBRT has been used for more than 50 years for treatment of BM. Although the development of SRS and concerns about its toxicity have decreased the use of WBRT in patients with limited BM, WBRT remains the standard treatment for patients with multiple BM. The typical dose and fractionation schedule for WBRT is 30 Gray (Gy) in 10 fractions. If patients have a short life expectancy, WBRT could also be delivered in 5 fractions with 20 Gy in total.41 Compared with the typical schedule (30 Gy in 10 fractions daily), several studies demonstrated that altered dose-fractionation schedules of WBRT do not show any improvement in OS, neurologic function, or symptom control. Meanwhile, additive radiosensitizers failed to show a benefit for OS or brain response but may increase the toxicity to patients.60

Aside from a good local control, WBRT is also associated with acute toxicity and long-term side effects. Clinicians are concerned about the decline of neurocognitive function44 and QoL45 caused by WBRT, since patients with HER2-positive breast cancer brain metastases have a relatively long survival. In order to ameliorate the decline of neurocognitive function caused by WBRT, several new strategies have been developed, including using intensity-modulated radiotherapy (IMRT) and prescribing medication such as memantine or donepezil. Since the hippocampus is a reservoir of neurological stem cells and plays an important role in memory preservation, IMRT is used to deliver a conformal WBRT to reduce the dose to the bilateral hippocampi, which is called hippocampal-avoidance WBRT (HAWBRT). Gondi et al61 have demonstrated that compared with historical controls, HAWBRT for BM can significantly mitigate radiation-induced neurocognitive and QoL decline. Memantine, a noncompetitive N-methyl-D-aspartate (NMDA) receptor antagonist, is a medication used to treat Alzheimer's disease. In a phase III trial, Brown et al62 demonstrated that a delayed time to cognitive decline and better cognitive function were observed among patients in the WBRT plus memantine arm. In addition, a phase III randomized placebo-controlled clinical trial has demonstrated that a different dementia medication, donepezil, contributes to modest improvements in several cognitive functions, especially among patients with greater pretreatment impairment.63

Stereotactic radiosurgery

In order to avoid the toxicity associated with WBRT, SRS has also been used for treating multiple BM. In a prospective observational study, Yamamoto et al64 demonstrated that SRS alone in patients with 5–10 brain metastases was non-inferior to that in patients with 2–4 brain metastases. In addition, Grandhi et al65 reported a high rate of local control in patients with ≥10 BM receiving SRS alone, but the median OS was only 4 months. Since a standard GK system can only treat one lesion at a time, treating multiple BM with SRS takes several hours. However, using a single isocenter, several forms of SRS such as intensity-modulated stereotactic radiosurgery,66 volumetric-modulated arc radiosurgery,67, 68, 69 and tomotherapy can deliver doses to multiple brain metastases simultaneously, which as a consequence decreases the treatment time down to minutes. Several studies have demonstrated that these forms of SRS can produce comparable clinical outcomes with those of conventional SRS for treating multiple intracranial metastases but in a much shorter treatment time,66, 67, 68, 69 which makes SRS a promising treatment for patients with multiple BM.

Targeted therapy for HER2-positive BCBM

Unlike estrogen and progesterone status, expression of HER2 has been reported to be highly concordant between the primary and brain metastatic tumors,70 which makes targeted therapy possible for treating patients with HER2-positive BCBM. Although the OS of patients with HER2-positive breast cancer has improved substantially in the trastuzumab era,4, 5, 6 the incidence of BM among these patients has been increasing in recent years. One of main reasons for this is that the blood brain barrier (BBB) makes the CNS a perfect sanctuary for tumor cells. The BBB is a barrier that selectively chooses molecules to enter the CNS. It consists of endothelial cells, a basement membrane, and astrocyte foot processes. The permeability of the BBB decreases 100-fold as the molecular weight of the drug increases from 200 Da to 450 Da.71 As a large molecule (145,531 Da), trastuzumab cannot penetrate the intact BBB, but multiple factors can disrupt the BBB, including metastatic tumors, surgery, and radiotherapy, which then allows limited amounts of large molecular agents to penetrate the CNS. A study has shown a change in the permeability of the BBB induced by WBRT, with a ratio of median trastuzumab level in the serum to cerebrospinal fluid 420:1 and 76:1 before and after WBRT, respectively.72 Despite the limited permeability of BBB to large molecular agents, it is important for patients with HER2-positive BCBM to improve systemic control, which can significantly prolong OS. Small molecular tyrosine kinase inhibitors (TKIs) have an improved ability to cross the BBB and can block multiple receptors of the erb-b2 receptor tyrosine kinase 2 (ERBB2) family at the same time, and are promising treatments for HER2-positive BCBM. In this section, we are going to discuss targeted therapy for HER2-positive BCBM.

Trastuzumab-containing regimens

Trastuzumab alone

Despite the limited blood-brain permeability, several studies demonstrated that trastuzumab alone for the treatment of HER2-positive breast cancer results in a prolonged time to BM and longer survival time after the diagnosis of BM. For example, Park et al21 demonstrated that patients receiving trastuzumab had a significantly longer median time to BM (15 months vs. 10 months, P = 0.035) and median time to death (14.9 vs. 4.0 months, P = 0.0005) than patients who were not treated with trastuzumab. Similarly, Rostami et al73 also demonstrated that the mean survival of patients with HER2-positive BCBM was prolonged when treated with trastuzumab (17.5 vs. 11 months).

Trastuzumab, pertuzumab, and taxane

Based on the landmark Clinical Evaluation of Pertuzumab and Trastuzumab (CLEOPATRA) trial, which has shown both significantly prolonged progression-free survival (PFS) and OS after adding pertuzumab to treatment with trastuzumab and docetaxel,74 current guidelines recommend dual anti-HER2 blockade with pertuzumab and trastuzumab plus a taxane as the preferred frontline regimen for HER2-positive metastatic disease.75 Although patients with BM were excluded from this trial, an exploratory analysis of patients who developed BM during the trial was performed. In this analysis, while the rate of BM as the first site of disease progression was similar between the 2 arms, a significantly delayed onset of BM was observed in the pertuzumab arm compared with the control arm. The median OS in the subset of patients who developed BM as the first site of disease progression tended to be longer in the pertuzumab arm (34.4 months) compared with the control arm (26.3 months). The comparison of OS between these 2 arms showed no significance on a log-rank test (P = 0.1139), but was significant on a Wilcoxon test (P = 0.0449).76 Furthermore, in a case report, pertuzumab with trastuzumab plus docetaxel was effective in reducing the recurrence of BCBM in a patient who was heavily pretreated.77 Based on these findings, further trials are warranted to investigate the effect of this regimen among patients with HER2-positive BCBM.

Trastuzumab emtansine

Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate of trastuzumab and the anti-microtubule agent emtansine. Based on a phase III trial, which demonstrated that T-DM1 and T-DM1 with pertuzumab were non-inferior in PFS and possibly better tolerated for some patients compared with trastuzumab plus taxane, the NCCN guidelines (version 1.2016) included T-DM1 as one of the first-line options for the treatment of patients with HER2-positive metastatic breast cancer (MBC).75 Furthermore, although the exploratory analysis of the EMILIA trial demonstrated a similar rate of CNS progression between the T-DM1 and the capecitabine-lapatinib arm, treatment with T-DM1 significantly improved the median OS in patients with CNS metastases (26.8 vs. 12.9 months, P = 0.008).78

Tyrosine kinase inhibitors

Small molecular tyrosine kinase inhibitors (TKIs) are promising anticancer agents for HER2-positive BCBM. TKIs have an improved ability as compared with antibodies to penetrate the BBB and can block multiple receptors of the ERBB2 family at the same time. Lapatinib plus capecitabine has been shown to benefit patients with HER2-positive BCBM. In addition, several other TKIs are involved in ongoing clinical trials to determine their usefulness for patients with HER2-positive BCBM.

Lapatinib

Lapatinib, which was approved by American Food and Drug Administration (FDA) in 2007, is a dual tyrosine kinase inhibitor of human epidermal growth factor receptor 1 (HER1) and HER2. As a small molecular and lipophilic agent, it can in theory penetrate the BBB. However, Taskar et al79 demonstrated that the lapatinib concentration in the CNS was quite variable among patients with BM, and the average lapatinib concentration in brain lesions was only 10–20% of that in peripheral metastatic lesions. Only in 17% of brain lesions did the lapatinib concentrations approach those in peripheral metastatic lesions. A modest CNS objective response was observed in patients receiving lapatinib as a monotherapy in a multicenter phase II study. Although the rate of CNS objective response was low, an association was observed between volumetric reduction and improvement in PFS and neurologic signs and symptoms.80 However, when lapatinib was administrated with capecitabine, the response rate in the brain increased to 20% and the rate of patients with a ≥20% volumetric reduction increased to 40%. In the LANDSCAPE trial, Bachelot et al81 demonstrated that among 44 previously untreated patients, an objective CNS response was observed in 29 patients (65.9%). This study also showed that treatment with lapatinib plus capecitabine at the time of diagnosis of BM delayed WBRT. The median time to WBRT was 8.3 months in this study, clinically relevant for a population with a short OS.81 According to the ASCO practice guideline on management of patients with advanced HER2-positive breast cancer and BM, if patients have asymptomatic, low-volume BM and have not received radiation therapy, upfront therapy with lapatinib and capecitabine is an option, although radiation therapy in this setting is still the primary option.24

Afatinib

Afatinib is an orally available, covalent, and irreversible inhibitor of all ERBB family members.82 In a phase II study, clinical activity was observed among trastuzumab-refractory patients receiving afatinib. In addition, Hoffknecht et al83 demonstrated that afatinib appeared to penetrate into the CNS with concentrations high enough to have a clinical effect on non-small cell lung cancer patients with CNS metastases. However, Cortés et al84 showed that afatinib alone or afatinib plus vinorelbine did not result in better outcomes in patients with HER2-positive BCBM compared with the investigator's choice of treatment.

Neratinib

Neratinib is an orally available, covalent, and irreversible inhibitor of HER1, HER2, and human epidermal growth factor receptor 4 (HER4). As a monotherapy, neratinib has been demonstrated to be an effective agent for either trastuzumab-naïve or heavily pretreated patients with HER2-positive breast cancer. In addition, compared with standard therapy alone, adding neratinib to standard therapy increases the rate of a pathological complete response among patients with HER2-positive, hormone receptor-negative, breast cancer.85 Neratinib plus capecitabine has shown remarkable systemic activity in patients with metastatic HER2-positive breast cancer in a phase I/II trial.86 Surprisingly, compared with the trastuzumab-paclitaxel arm, a lower incidence of CNS recurrence and prolonged time to BM were observed in the neratinib-paclitaxel arm in the NEFERT-T trial,87 which showed neratinib could be a promising agent for treating patients with HER2-positive BCBM. However, recently, a phase II trial demonstrated that neratinib alone resulted in a low CNS objective response rate. Despite the promising effect as monotherapy, the effect of neratinib combined with other agents for patients with HER2-positive BCBM is still unknown.

Trastuzumab vs. lapatinib

There were several studies that compared trastuzumab with lapatinib in patients with HER2-positive BCBM. Yap et al88 demonstrated that the OS after BM in patients with HER2-positive BCBM treated with lapatinib alone and trastuzumab alone were 21.4 months and 10.5 months, respectively; the best survival benefit (25.9 months) was observed in patients treated with both trastuzumab and lapatinib. Similarly, Kaplan et al89 also demonstrated that median OS in patients treated with lapatinib plus capecitabine was significantly increased compared with that in patients treated with trastuzumab-based therapy (19.1 vs. 12 months; P = 0.039). Recently, the CEREBEL trial showed the rates of CNS metastases as first site of relapse were similar in the lapatinib–capecitabine arm and trastuzumab–capecitabine arm (3% vs. 5%, P = 0.360) but they were both far lower than the expected rates of 12% and 20%, respectively. In addition, the overall rates of CNS progression at any time in both arms (7% vs. 6%; P = 0.8646) were lower than anticipated as well. Despite the low rates, this trial was inconclusive as to the prophylactic effect of both regimens.90

Bevacizumab

Bevacizumab is a monoclonal antibody binding to the ligand of vascular endothelial growth factor (VEGF) and the FDA has approved bevacizumab plus paclitaxel as a first-line therapy in patients with MBC.91 Several studies have reported that there is a significant positive correlation between VEGF and HER2 expression, suggesting bevacizumab may be useful for treating HER2-positive breast cancer.92 Despite its large molecular weight, bevacizumab has shown benefits for patients with some primary brain tumors such as glioblastoma multiforme, which gives researchers insight into treating BM.93 In the past, patients with BM had been excluded from bevacizumab trials since a hepatocellular carcinoma patient with undiagnosed BM suffered a fatal cerebral hemorrhage during a bevacizumab trial in 1997.94 However, a retrospective exploratory analysis demonstrated that the risk of developing cerebral hemorrhage among patients with CNS metastases was independent of bevacizumab therapy.95 In a case series of 3 patients with progressive HER2-positive BCBM pretreated by WBRT, the administration of bevacizumab plus anti-HER2 increased the OS of these 3 patients.96 In a phase II study, bevacizumab followed by etoposide and cisplatin (BEEP regimen), appeared highly effective in BCBM patients (not HER2-positive specific) who were refractory to WBRT. Surprisingly, the clinical efficacy of the BEEP regimen for HER2-positive patients (23 of 35) in this study was higher than that observed in clinical trials in which patients received lapatinib and capecitabine after WBRT.97 However, the analysis of the AVEREL trial (a phase III trial for HER2-positive MBC) showed the reduction in the rate of BM was just a trend in the bevacizumab arm, failing to reach statistical significance.98

Other strategies to deliver drugs to the CNS

In order to penetrate the BBB, researchers have developed several strategies to deliver drugs with a higher concentration to the brain, such as delivering them mediated with radiation or ultrasound, or with the help of nanotechnology.

Focused ultrasound plus microbubbles

Previous studies have shown that focused ultrasound (FUS) plus microbubbles could temporarily disrupt the tight junctions of the BBB, allowing drugs to penetrate.99, 100 Park et al101 demonstrated for the first time that the combination of trastuzumab and FUS had an anticancer activity for HER2-positive breast tumor inoculated into rats' brains. During this experiment, after 6 weekly trastuzumab treatments mediated with focused ultrasound, the mean tumor volume was significantly reduced and the survival was significantly prolonged without sequelae. Furthermore, a recent study has also shown a delayed progression of brain metastases from HER2-positive breast cancer in some of the rats receiving treatment with trastuzumab and pertuzumab mediated with FUS.102 These findings show the therapeutic potential of this noninvasive technique for targeted drug delivery to the brain.

Nanotechnology

Nanoparticles can conjugate with many anticancer agents and have been successfully used as vehicles to deliver therapeutic agents across the BBB.103 Patil et al104 demonstrated that compared with mice receiving phosphate buffered saline, a significantly prolonged survival was observed in mice with HER2-positive BCBM that were treated with targeted nanodrugs. In addition, Hamilton et al105 also showed that nanoparticles coated with a tumor-penetrating peptide (iRGD) may be a promising treatment for prevention of BM. Several clinical trials are ongoing for nano-therapeutic drugs such as MM-302 (NCT02735798) in order to determine their effect on patients with BCBM.

Biomarker for BCBM

Although much progress has been made in recent years, BCBM still seems incurable. A cancer biomarker would represent a significant step towards preventing, delaying, and eliminating brain metastases. microRNAs (miRNAs) are a complex network of non-coding RNA molecules that have been demonstrated to play an important role in regulating tumor metastases.106 Since a miRNA test is a stable, noninvasive, and highly sensitive approach to reflect tumors inside the body, miRNAs are promising prognostic and diagnostic markers for BM.107 In this section, we will discuss some miRNAs associated with BCBM (Table 1).

Table 1.

Several miRNAs associated with brain metastases from breast cancer.

miRNA Mechanism of action Expression in brain metastases
miR-7108 Modulate KLF4 gene expression Down-regulated
miR-146a109 Up-regulate B-catenin and down-regulate hnRNPC Down-regulated
miR-509109 Modulate the RhoC-TNF-α network Down-regulated
miR-19a110 Down-regulate tissue factor expression by binding 3′-UTR of the tissue factor transcript111 Down-regulated
miR-29c110 Induce apoptosis by reducing MCL-1112 Down-regulated
miR-1258113 Inhibit the expression and activity of heparanase in BCBM cells Down-regulated
miR-122109 Suppress glucose uptake by niche cells Up-regulated
miR-200114 Prevent TGF beta-induced EMT by regulating E-cadherin transcriptional repressors ZEB1 and ZEB2115 Up-regulated
miR-21050, 110 Promote cancer proliferation by targeting PTP1b and HIF-1α116 Up-regulated

miRNAs: microRNAs; KLF4: Kruppel-like factor 4; hnRNPC: heterogeneous nuclear ribonucleoproteins C1/C2; RhoC: ras homolog gene family, member C; TNF-α: tumor necrosis factor-α; 3′-UTR: 3′-untranslated region; MCL-1: myeloid cell leukemia-1; BCBM: breast cancer brain metastases; TGF: transforming growth factor; EMT: epithelial to mesenchymal transition; ZEB1: zinc finger E-box binding homeobox 1; ZEB2: zinc finger E-box binding homeobox 2; PTP1b: protein tyrosine phosphatase-1b; HIF-1α: hypoxia-inducible factor-1α.

There are several miRNAs that are involved in the mechanism of BM from patients with breast cancer and have the potential to serve as signatures to predict it. miRNAs such as miR-181 and miR-122 are able to facilitate brain metastasis of breast cancer.117, 118 For example, Fong et al118 demonstrated that miR-122 was able to suppress glucose uptake in distant organs, including the brain and lungs, through downregulating the glycolytic enzyme pyruvate kinase, and thus increasing the incidence of metastasis. In contrast, several miRNAs including miR-7, miR-146a, and miR-509 can inhibit BM of breast cancer.108, 109, 119 For example, Okuda et al108 demonstrated that kruppel-like factor 4 (KLF4) and miR-7 were dysregulated in brain metastatic tumors. MiR-146a is absent from brain metastatic tumors and can suppress the migratory and invasive potential of breast cancer cells by upregulating β-catenin and down-regulating heterogeneous nuclear ribonucleoproteins C1/C2 (hnRNPC).109

Conclusions

After the introduction of effective anti-HER2 treatment, patients with HER2-positive BCBM have experienced a significantly prolonged survival mainly because of better extracranial control. However, the incidence of BM among HER2-positive breast cancer patients has been increasing and an increased proportion of patients with HER2-positive breast cancer have died of intracranial progression in recent years, which makes BM from HER2-positive breast cancer an important issue of concern. For local treatment, QoL and local control should both be taken into account. Adjuvant WBRT should be delayed in treating limited BM because of its impairment of neurocognitive function while having no benefit for OS. With advances in technology, SRS has been shown to not only be an increasingly better treatment for limited BM, but also a promising treatment for diffuse brain metastases. Despite the limited penetrability of large molecular agents, it is critical for patients with HER2-positive BCBM to receive targeted treatment, which can significantly prolong OS. With improved BBB penetrability, TKIs have been shown to be beneficial for patients with HER2-positive BCBM, especially lapatinib combined with capecitabine. While several new drugs have been recently approved and demonstrated better extracranial control, new strategies and more drugs with better BBB permeability need to be developed and confirmed in future studies for better intracranial control. Furthermore, several miRNAs have been found to be involved in the mechanism of BM developing from breast cancer, showing clinical potential as preventive, diagnostic, and prognostic biomarkers for BCBM.

Conflicts of interest

The authors declare that they have no conflicts of interest.

Acknowledgements

This study was supported by the National Natural Science Foundation of China (No. 81672634).

Edited by Pei-Fang Wei

Footnotes

Peer review under responsibility of Chinese Medical Association.

References

  • 1.Weil R.J., Palmieri D.C., Bronder J.L., Stark A.M., Steeg P.S. Breast cancer metastasis to the central nervous system. Am J Pathol. 2005;167:913–920. doi: 10.1016/S0002-9440(10)61180-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Lin N.U. Breast cancer brain metastases: new directions in systemic therapy. Ecancermedicalscience. 2013;7:307. doi: 10.3332/ecancer.2013.307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Arslan C., Dizdar O., Altundag K. Systemic treatment in breast-cancer patients with brain metastasis. Expert Opin Pharmacother. 2010;11:1089–1100. doi: 10.1517/14656561003702412. [DOI] [PubMed] [Google Scholar]
  • 4.Lin N.U., Winer E.P. Brain metastases: the HER2 paradigm. Clin Cancer Res. 2007;13:1648–1655. doi: 10.1158/1078-0432.CCR-06-2478. [DOI] [PubMed] [Google Scholar]
  • 5.Pestalozzi B.C., Holmes E., de Azambuja E. CNS relapses in patients with HER2-positive early breast cancer who have and have not received adjuvant trastuzumab: a retrospective substudy of the HERA trial (BIG 1-01) Lancet Oncol. 2013;14:244–248. doi: 10.1016/S1470-2045(13)70017-2. [DOI] [PubMed] [Google Scholar]
  • 6.Aversa C., Rossi V., Geuna E. Metastatic breast cancer subtypes and central nervous system metastases. Breast. 2014;23:623–628. doi: 10.1016/j.breast.2014.06.009. [DOI] [PubMed] [Google Scholar]
  • 7.Pestalozzi B.C., Zahrieh D., Price K.N. Identifying breast cancer patients at risk for Central Nervous System (CNS) metastases in trials of the International Breast Cancer Study Group (IBCSG) Ann Oncol. 2006;17:935–944. doi: 10.1093/annonc/mdl064. [DOI] [PubMed] [Google Scholar]
  • 8.Kennecke H., Yerushalmi R., Woods R. Metastatic behavior of breast cancer subtypes. J Clin Oncol. 2010;28:3271–3277. doi: 10.1200/JCO.2009.25.9820. [DOI] [PubMed] [Google Scholar]
  • 9.Brufsky A.M., Mayer M., Rugo H.S. Central nervous system metastases in patients with HER2-positive metastatic breast cancer: incidence, treatment, and survival in patients from registHER. Clin Cancer Res. 2011;17:4834–4843. doi: 10.1158/1078-0432.CCR-10-2962. [DOI] [PubMed] [Google Scholar]
  • 10.Bendell J.C., Domchek S.M., Burstein H.J. Central nervous system metastases in women who receive trastuzumab-based therapy for metastatic breast carcinoma. Cancer. 2003;97:2972–2977. doi: 10.1002/cncr.11436. [DOI] [PubMed] [Google Scholar]
  • 11.Olson E.M., Najita J.S., Sohl J. Clinical outcomes and treatment practice patterns of patients with HER2-positive metastatic breast cancer in the post-trastuzumab era. Breast. 2013;22:525–531. doi: 10.1016/j.breast.2012.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Olson E.M., Abdel-Rasoul M., Maly J., Wu C.S., Lin N.U., Shapiro C.L. 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;24:1526–1533. doi: 10.1093/annonc/mdt036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Jung S.Y., Rosenzweig M., Sereika S.M., Linkov F., Brufsky A., Weissfeld J.L. Factors associated with mortality after breast cancer metastasis. Cancer Causes Control. 2012;23:103–112. doi: 10.1007/s10552-011-9859-8. [DOI] [PubMed] [Google Scholar]
  • 14.Niikura N., Liu J., Hayashi N. Loss of human epidermal growth factor receptor 2 (HER2) expression in metastatic sites of HER2-overexpressing primary breast tumors. J Clin Oncol. 2012;30:593–599. doi: 10.1200/JCO.2010.33.8889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Clayton A.J., Danson S., Jolly S. Incidence of cerebral metastases in patients treated with trastuzumab for metastatic breast cancer. Br J Cancer. 2004;91:639–643. doi: 10.1038/sj.bjc.6601970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Lin N.U., Claus E., Sohl J., Razzak A.R., Arnaout A., Winer E.P. Sites of distant recurrence and clinical outcomes in patients with metastatic triple-negative breast cancer: high incidence of central nervous system metastases. Cancer. 2008;113:2638–2645. doi: 10.1002/cncr.23930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Dawood S., Broglio K., Esteva F.J. Defining prognosis for women with breast cancer and CNS metastases by HER2 status. Ann Oncol. 2008;19:1242–1248. doi: 10.1093/annonc/mdn036. [DOI] [PubMed] [Google Scholar]
  • 18.Mahmoud-Ahmed A.S., Suh J.H., Lee S.Y., Crownover R.L., Barnett G.H. Results of whole brain radiotherapy in patients with brain metastases from breast cancer: a retrospective study. Int J Radiat Oncol Biol Phys. 2002;54:810–817. doi: 10.1016/s0360-3016(02)02967-x. [DOI] [PubMed] [Google Scholar]
  • 19.Tham Y.L., Sexton K., Kramer R., Hilsenbeck S., Elledge R. Primary breast cancer phenotypes associated with propensity for central nervous system metastases. Cancer. 2006;107:696–704. doi: 10.1002/cncr.22041. [DOI] [PubMed] [Google Scholar]
  • 20.Kirsch D.G., Ledezma C.J., Mathews C.S. Survival after brain metastases from breast cancer in the trastuzumab era. J Clin Oncol. 2005;23:2114–2116. doi: 10.1200/JCO.2005.05.249. author reply 2116–2117. [DOI] [PubMed] [Google Scholar]
  • 21.Park Y.H., Park M.J., Ji S.H. Trastuzumab treatment improves brain metastasis outcomes through control and durable prolongation of systemic extracranial disease in HER2-overexpressing breast cancer patients. Br J Cancer. 2009;100:894–900. doi: 10.1038/sj.bjc.6604941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Gori S., Rimondini S., De Angelis V. Central nervous system metastases in HER-2 positive metastatic breast cancer patients treated with trastuzumab: incidence, survival, and risk factors. Oncologist. 2007;12:766–773. doi: 10.1634/theoncologist.12-7-766. [DOI] [PubMed] [Google Scholar]
  • 23.Sperduto P.W., Kased N., Roberge D. Summary report on the graded prognostic assessment: an accurate and facile diagnosis-specific tool to estimate survival for patients with brain metastases. J Clin Oncol. 2012;30:419–425. doi: 10.1200/JCO.2011.38.0527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Ramakrishna N., Temin S., Chandarlapaty S. Recommendations on disease management for patients with advanced human epidermal growth factor receptor 2-positive breast cancer and brain metastases: American Society of Clinical Oncology clinical practice guideline. J Clin Oncol. 2014;32:2100–2108. doi: 10.1200/JCO.2013.54.0955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Patchell R.A., Tibbs P.A., Walsh J.W. A randomized trial of surgery in the treatment of single metastases to the brain. N Engl J Med. 1990;322:494–500. doi: 10.1056/NEJM199002223220802. [DOI] [PubMed] [Google Scholar]
  • 26.Noordijk E.M., Vecht C.J., Haaxma-Reiche H. The choice of treatment of single brain metastasis should be based on extracranial tumor activity and age. Int J Radiat Oncol Biol Phys. 1994;29:711–717. doi: 10.1016/0360-3016(94)90558-4. [DOI] [PubMed] [Google Scholar]
  • 27.Mintz A.H., Kestle J., Rathbone M.P. A randomized trial to assess the efficacy of surgery in addition to radiotherapy in patients with a single cerebral metastasis. Cancer. 1996;78:1470–1476. doi: 10.1002/(sici)1097-0142(19961001)78:7<1470::aid-cncr14>3.0.co;2-x. [DOI] [PubMed] [Google Scholar]
  • 28.Tabouret E., Metellus P., Gonçalves A. Assessment of prognostic scores in brain metastases from breast cancer. Neuro Oncol. 2014;16:421–428. doi: 10.1093/neuonc/not200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Patchell R.A., Tibbs P.A., Regine W.F. Postoperative radiotherapy in the treatment of single metastases to the brain: a randomized trial. JAMA. 1998;280:1485–1489. doi: 10.1001/jama.280.17.1485. [DOI] [PubMed] [Google Scholar]
  • 30.Badiyan S.N., Regine W.F., Mehta M. Stereotactic radiosurgery for treatment of brain metastases. J Oncol Pract. 2016;12:703–712. doi: 10.1200/JOP.2016.012922. [DOI] [PubMed] [Google Scholar]
  • 31.Coffey R.J., Lunsford L.D. Stereotactic radiosurgery using the 201 cobalt-60 source gamma knife. Neurosurg Clin N Am. 1990;1:933–954. [PubMed] [Google Scholar]
  • 32.Mack A., Czempiel H., Kreiner H.J., Dürr G., Wowra B. Quality assurance in stereotactic space. A system test for verifying the accuracy of aim in radiosurgery. Med Phys. 2002;29:561–568. doi: 10.1118/1.1463062. [DOI] [PubMed] [Google Scholar]
  • 33.Adler J.R., Chang S.D., Murphy M.J., Doty J., Geis P., Hancock S.L. The Cyberknife: a frameless robotic system for radiosurgery. Stereotact Funct Neurosurg. 1997;69:124–128. doi: 10.1159/000099863. [DOI] [PubMed] [Google Scholar]
  • 34.Chang S.D., Main W., Martin D.P., Gibbs I.C., Heilbrun M.P. An analysis of the accuracy of the CyberKnife: a robotic frameless stereotactic radiosurgical system. Neurosurgery. 2003;52:140–146. doi: 10.1097/00006123-200301000-00018. discussion 146–147. [DOI] [PubMed] [Google Scholar]
  • 35.Yu C., Main W., Taylor D., Kuduvalli G., Apuzzo M.L., Adler J.R. An anthropomorphic phantom study of the accuracy of Cyberknife spinal radiosurgery. Neurosurgery. 2004;55:1138–1149. doi: 10.1227/01.neu.0000141080.54647.11. [DOI] [PubMed] [Google Scholar]
  • 36.Yu C., Jozsef G., Apuzzo M.L., Petrovich Z. Dosimetric comparison of CyberKnife with other radiosurgical modalities for an ellipsoidal target. Neurosurgery. 2003;53:1155–1162. doi: 10.1227/01.neu.0000088805.01793.5a. discussion 1162–1163. [DOI] [PubMed] [Google Scholar]
  • 37.Collins S.P., Coppa N.D., Zhang Y., Collins B.T., McRae D.A., Jean W.C. CyberKnife radiosurgery in the treatment of complex skull base tumors: analysis of treatment planning parameters. Radiat Oncol. 2006;1:46. doi: 10.1186/1748-717X-1-46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Flickinger J.C., Lunsford L.D., Wu A., Maitz A.H., Kalend A.M. Treatment planning for gamma knife radiosurgery with multiple isocenters. Int J Radiat Oncol Biol Phys. 1990;18:1495–1501. doi: 10.1016/0360-3016(90)90326-f. [DOI] [PubMed] [Google Scholar]
  • 39.Leith J.T., Cook S., Chougule P. Intrinsic and extrinsic characteristics of human tumors relevant to radiosurgery: comparative cellular radiosensitivity and hypoxic percentages. Acta Neurochir Suppl. 1994;62:18–27. doi: 10.1007/978-3-7091-9371-6_5. [DOI] [PubMed] [Google Scholar]
  • 40.Wowra B., Muacevic A., Tonn J.C. Quality of radiosurgery for single brain metastases with respect to treatment technology: a matched-pair analysis. J Neurooncol. 2009;94:69–77. doi: 10.1007/s11060-009-9802-y. [DOI] [PubMed] [Google Scholar]
  • 41.Fontanella C., De Carlo E., Cinausero M., Pelizzari G., Venuti I., Puglisi F. Central nervous system involvement in breast cancer patients: is the therapeutic landscape changing too slowly. Cancer Treat Rev. 2016;46:80–88. doi: 10.1016/j.ctrv.2016.03.014. [DOI] [PubMed] [Google Scholar]
  • 42.Aoyama H., Shirato H., Tago M. Stereotactic radiosurgery plus whole-brain radiation therapy vs stereotactic radiosurgery alone for treatment of brain metastases: a randomized controlled trial. JAMA. 2006;295:2483–2491. doi: 10.1001/jama.295.21.2483. [DOI] [PubMed] [Google Scholar]
  • 43.Kondziolka D., Patel A., Lunsford L.D., Kassam A., Flickinger J.C. Stereotactic radiosurgery plus whole brain radiotherapy versus radiotherapy alone for patients with multiple brain metastases. Int J Radiat Oncol Biol Phys. 1999;45:427–434. doi: 10.1016/s0360-3016(99)00198-4. [DOI] [PubMed] [Google Scholar]
  • 44.Sperduto P.W., Wang M., Robins H.I. A phase 3 trial of whole brain radiation therapy and stereotactic radiosurgery alone versus WBRT and SRS with temozolomide or erlotinib for non-small cell lung cancer and 1 to 3 brain metastases: Radiation Therapy Oncology Group 0320. Int J Radiat Oncol Biol Phys. 2013;85:1312–1318. doi: 10.1016/j.ijrobp.2012.11.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Soffietti R., Kocher M., Abacioglu U.M. A European Organisation for Research and Treatment of Cancer phase III trial of adjuvant whole-brain radiotherapy versus observation in patients with one to three brain metastases from solid tumors after surgical resection or radiosurgery: quality-of-life results. J Clin Oncol. 2013;31:65–72. doi: 10.1200/JCO.2011.41.0639. [DOI] [PubMed] [Google Scholar]
  • 46.Kocher M., Soffietti R., Abacioglu U. Adjuvant whole-brain radiotherapy versus observation after radiosurgery or surgical resection of one to three cerebral metastases: results of the EORTC 22952-26001 study. J Clin Oncol. 2011;29:134–141. doi: 10.1200/JCO.2010.30.1655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Hahn C., Kavanagh B., Bhatnagar A. Choosing wisely: the American Society for Radiation Oncology's top 5 list. Pract Radiat Oncol. 2014;4:349–355. doi: 10.1016/j.prro.2014.06.003. [DOI] [PubMed] [Google Scholar]
  • 48.Fogarty G.B., Hong A., Gondi V. Debate: adjuvant whole brain radiotherapy or not? More data is the wiser choice. BMC Cancer. 2016;16:372. doi: 10.1186/s12885-016-2433-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Tsao M.N., Rades D., Wirth A. Radiotherapeutic and surgical management for newly diagnosed brain metastasis(es): an American Society for Radiation Oncology evidence-based guideline. Pract Radiat Oncol. 2012;2:210–225. doi: 10.1016/j.prro.2011.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Minniti G., Esposito V., Clarke E. Multidose stereotactic radiosurgery (9 Gy × 3) of the postoperative resection cavity for treatment of large brain metastases. Int J Radiat Oncol Biol Phys. 2013;86:623–629. doi: 10.1016/j.ijrobp.2013.03.037. [DOI] [PubMed] [Google Scholar]
  • 51.Jensen C.A., Chan M.D., McCoy T.P. Cavity-directed radiosurgery as adjuvant therapy after resection of a brain metastasis. J Neurosurg. 2011;114:1585–1591. doi: 10.3171/2010.11.JNS10939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Brennan C., Yang T.J., Hilden P. A phase 2 trial of stereotactic radiosurgery boost after surgical resection for brain metastases. Int J Radiat Oncol Biol Phys. 2014;88:130–136. doi: 10.1016/j.ijrobp.2013.09.051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Robbins J.R., Ryu S., Kalkanis S. Radiosurgery to the surgical cavity as adjuvant therapy for resected brain metastasis. Neurosurgery. 2012;71:937–943. doi: 10.1227/NEU.0b013e31826909f2. [DOI] [PubMed] [Google Scholar]
  • 54.Rao G., Ahmed S., Hess K., Mahajan A. 215 postoperative stereotactic radiosurgery vs observation for completely resected brain metastases: results of a prospective randomized study. Neurosurgery. 2016;63 Suppl 1:184. [Google Scholar]
  • 55.Choi C.Y., Chang S.D., Gibbs I.C. Stereotactic radiosurgery of the postoperative resection cavity for brain metastases: prospective evaluation of target margin on tumor control. Int J Radiat Oncol Biol Phys. 2012;84:336–342. doi: 10.1016/j.ijrobp.2011.12.009. [DOI] [PubMed] [Google Scholar]
  • 56.Patel K.R., Prabhu R.S., Kandula S. Intracranial control and radiographic changes with adjuvant radiation therapy for resected brain metastases: whole brain radiotherapy versus stereotactic radiosurgery alone. J Neurooncol. 2014;120:657–663. doi: 10.1007/s11060-014-1601-4. [DOI] [PubMed] [Google Scholar]
  • 57.Atalar B., Modlin L.A., Choi C.Y. Risk of leptomeningeal disease in patients treated with stereotactic radiosurgery targeting the postoperative resection cavity for brain metastases. Int J Radiat Oncol Biol Phys. 2013;87:713–718. doi: 10.1016/j.ijrobp.2013.07.034. [DOI] [PubMed] [Google Scholar]
  • 58.Asher A.L., Burri S.H., Wiggins W.F. A new treatment paradigm: neoadjuvant radiosurgery before surgical resection of brain metastases with analysis of local tumor recurrence. Int J Radiat Oncol Biol Phys. 2014;88:899–906. doi: 10.1016/j.ijrobp.2013.12.013. [DOI] [PubMed] [Google Scholar]
  • 59.Patel K.R., Burri S.H., Asher A.L. Comparing preoperative with postoperative stereotactic radiosurgery for resectable brain metastases: a multi-institutional analysis. Neurosurgery. 2016;79:279–285. doi: 10.1227/NEU.0000000000001096. [DOI] [PubMed] [Google Scholar]
  • 60.Tsao M.N., Lloyd N., Wong R.K. Whole brain radiotherapy for the treatment of newly diagnosed multiple brain metastases. Cochrane Database Syst Rev. 2012;4:CD003869. doi: 10.1002/14651858.CD003869.pub3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Gondi V., Pugh S.L., Tome W.A. Preservation of memory with conformal avoidance of the hippocampal neural stem-cell compartment during whole-brain radiotherapy for brain metastases (RTOG 0933): a phase II multi-institutional trial. J Clin Oncol. 2014;32:3810–3816. doi: 10.1200/JCO.2014.57.2909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Brown P.D., Pugh S., Laack N.N. Memantine for the prevention of cognitive dysfunction in patients receiving whole-brain radiotherapy: a randomized, double-blind, placebo-controlled trial. Neuro Oncol. 2013;15:1429–1437. doi: 10.1093/neuonc/not114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Rapp S.R., Case L.D., Peiffer A. Donepezil for irradiated brain tumor survivors: a phase III randomized placebo-controlled clinical trial. J Clin Oncol. 2015;33:1653–1659. doi: 10.1200/JCO.2014.58.4508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Yamamoto M., Serizawa T., Shuto T. Stereotactic radiosurgery for patients with multiple brain metastases (JLGK0901): a multi-institutional prospective observational study. Lancet Oncol. 2014;15:387–395. doi: 10.1016/S1470-2045(14)70061-0. [DOI] [PubMed] [Google Scholar]
  • 65.Grandhi R., Kondziolka D., Panczykowski D. Stereotactic radiosurgery using the Leksell Gamma Knife Perfexion unit in the management of patients with 10 or more brain metastases. J Neurosurg. 2012;117:237–245. doi: 10.3171/2012.4.JNS11870. [DOI] [PubMed] [Google Scholar]
  • 66.Nath S.K., Lawson J.D., Simpson D.R. Single-isocenter frameless intensity-modulated stereotactic radiosurgery for simultaneous treatment of multiple brain metastases: clinical experience. Int J Radiat Oncol Biol Phys. 2010;78:91–97. doi: 10.1016/j.ijrobp.2009.07.1726. [DOI] [PubMed] [Google Scholar]
  • 67.Lau S.K., Zakeri K., Zhao X. Single-isocenter frameless volumetric modulated arc radiosurgery for multiple intracranial metastases. Neurosurgery. 2015;77:233–240. doi: 10.1227/NEU.0000000000000763. discussion 240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Thomas A., Niebanck M., Juang T., Wang Z., Oldham M. A comprehensive investigation of the accuracy and reproducibility of a multitarget single isocenter VMAT radiosurgery technique. Med Phys. 2013;40:121725. doi: 10.1118/1.4829518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Thomas E.M., Popple R.A., Wu X. Comparison of plan quality and delivery time between volumetric arc therapy (RapidArc) and Gamma Knife radiosurgery for multiple cranial metastases. Neurosurgery. 2014;75:409–417. doi: 10.1227/NEU.0000000000000448. discussion 417–418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Shen Q., Sahin A.A., Hess K.R. Breast cancer with brain metastases: clinicopathologic features, survival, and paired biomarker analysis. Oncologist. 2015;20:466–473. doi: 10.1634/theoncologist.2014-0107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Koo T., Kim I.A. Brain metastasis in human epidermal growth factor receptor 2-positive breast cancer: from biology to treatment. Radiat Oncol J. 2016;34:1–9. doi: 10.3857/roj.2016.34.1.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Stemmler H.J., Schmitt M., Willems A., Bernhard H., Harbeck N., Heinemann V. Ratio of trastuzumab levels in serum and cerebrospinal fluid is altered in HER2-positive breast cancer patients with brain metastases and impairment of blood-brain barrier. Anticancer Drugs. 2007;18:23–28. doi: 10.1097/01.cad.0000236313.50833.ee. [DOI] [PubMed] [Google Scholar]
  • 73.Rostami R., Mittal S., Rostami P., Tavassoli F., Jabbari B. Brain metastasis in breast cancer: a comprehensive literature review. J Neurooncol. 2016;127:407–414. doi: 10.1007/s11060-016-2075-3. [DOI] [PubMed] [Google Scholar]
  • 74.Swain S.M., Baselga J., Kim S.B. Pertuzumab, trastuzumab, and docetaxel in HER2-positive metastatic breast cancer. N Engl J Med. 2015;372:724–734. doi: 10.1056/NEJMoa1413513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Gradishar W.J., Anderson B.O., Balassanian R. Invasive breast cancer version 1.2016, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2016;14:324–354. doi: 10.6004/jnccn.2016.0037. [DOI] [PubMed] [Google Scholar]
  • 76.Swain S.M., Baselga J., Miles D. Incidence of central nervous system metastases in patients with HER2-positive metastatic breast cancer treated with pertuzumab, trastuzumab, and docetaxel: results from the randomized phase III study CLEOPATRA. Ann Oncol. 2014;25:1116–1121. doi: 10.1093/annonc/mdu133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Senda N., Yamaguchi A., Nishimura H., Shiozaki T., Tsuyuki S. Pertuzumab, trastuzumab and docetaxel reduced the recurrence of brain metastasis from breast cancer: a case report. Breast Cancer. 2016;23:323–328. doi: 10.1007/s12282-015-0623-x. [DOI] [PubMed] [Google Scholar]
  • 78.Verma S., Miles D., Gianni L. Trastuzumab emtansine for HER2-positive advanced breast cancer. N Engl J Med. 2012;367:1783–1791. doi: 10.1056/NEJMoa1209124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Taskar K.S., Rudraraju V., Mittapalli R.K. Lapatinib distribution in HER2 overexpressing experimental brain metastases of breast cancer. Pharm Res. 2012;29:770–781. doi: 10.1007/s11095-011-0601-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Lin N.U., Diéras V., Paul D. Multicenter phase II study of lapatinib in patients with brain metastases from HER2-positive breast cancer. Clin Cancer Res. 2009;15:1452–1459. doi: 10.1158/1078-0432.CCR-08-1080. [DOI] [PubMed] [Google Scholar]
  • 81.Bachelot T., Romieu G., Campone M. Lapatinib plus capecitabine in patients with previously untreated brain metastases from HER2-positive metastatic breast cancer (LANDSCAPE): a single-group phase 2 study. Lancet Oncol. 2013;14:64–71. doi: 10.1016/S1470-2045(12)70432-1. [DOI] [PubMed] [Google Scholar]
  • 82.Zhang X., Munster P.N. New protein kinase inhibitors in breast cancer: afatinib and neratinib. Expert Opin Pharmacother. 2014;15:1277–1288. doi: 10.1517/14656566.2014.913570. [DOI] [PubMed] [Google Scholar]
  • 83.Hoffknecht P., Tufman A., Wehler T. Efficacy of the irreversible ErbB family blocker afatinib in epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI)-pretreated non-small-cell lung cancer patients with brain metastases or leptomeningeal disease. J Thorac Oncol. 2015;10:156–163. doi: 10.1097/JTO.0000000000000380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Cortés J., Dieras V., Ro J. Afatinib alone or afatinib pl12us vinorelbine versus investigator's choice of treatment for HER2-positive breast cancer with progressive brain metastases after trastuzumab, lapatinib, or both (LUX-Breast 3): a randomised, open-label, multicentre, phase 2 trial. Lancet Oncol. 2015;16:1700–1710. doi: 10.1016/S1470-2045(15)00373-3. [DOI] [PubMed] [Google Scholar]
  • 85.Park J.W., Liu M.C., Yee D. Adaptive randomization of neratinib in early breast cancer. N Engl J Med. 2016;375:11–22. doi: 10.1056/NEJMoa1513750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Saura C., Garcia-Saenz J.A., Xu B. Safety and efficacy of neratinib in combination with capecitabine in patients with metastatic human epidermal growth factor receptor 2-positive breast cancer. J Clin Oncol. 2014;32:3626–3633. doi: 10.1200/JCO.2014.56.3809. [DOI] [PubMed] [Google Scholar]
  • 87.Awada A., Colomer R., Inoue K. Neratinib plus paclitaxel vs trastuzumab plus paclitaxel in previously untreated metastatic ERBB2-positive breast cancer: the NEfERT-T randomized clinical trial. JAMA Oncol. 2016;2:1557–1564. doi: 10.1001/jamaoncol.2016.0237. [DOI] [PubMed] [Google Scholar]
  • 88.Yap Y.S., Cornelio G.H., Devi B.C. Brain metastases in Asian HER2-positive breast cancer patients: anti-HER2 treatments and their impact on survival. Br J Cancer. 2012;107:1075–1082. doi: 10.1038/bjc.2012.346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Kaplan M.A., Isikdogan A., Koca D. Biological subtypes and survival outcomes in breast cancer patients with brain metastases (study of the Anatolian Society of Medical Oncology) Oncology. 2012;83:141–150. doi: 10.1159/000338782. [DOI] [PubMed] [Google Scholar]
  • 90.Pivot X., Manikhas A., Żurawski B. CEREBEL (EGF111438): a phase III, randomized, open-label study of lapatinib plus capecitabine versus trastuzumab plus capecitabine in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer. J Clin Oncol. 2015;33:1564–1573. doi: 10.1200/JCO.2014.57.1794. [DOI] [PubMed] [Google Scholar]
  • 91.Alvarez R.H., Valero V., Hortobagyi G.N. Emerging targeted therapies for breast cancer. J Clin Oncol. 2010;28:3366–3379. doi: 10.1200/JCO.2009.25.4011. [DOI] [PubMed] [Google Scholar]
  • 92.Konecny G.E., Meng Y.G., Untch M. Association between HER-2/neu and vascular endothelial growth factor expression predicts clinical outcome in primary breast cancer patients. Clin Cancer Res. 2004;10:1706–1716. doi: 10.1158/1078-0432.ccr-0951-3. [DOI] [PubMed] [Google Scholar]
  • 93.Stavrou D. Monoclonal antibodies in neuro-oncology. Neurosurg Rev. 1990;13:7–18. doi: 10.1007/BF00638887. [DOI] [PubMed] [Google Scholar]
  • 94.Gordon M.S., Margolin K., Talpaz M. Phase I safety and pharmacokinetic study of recombinant human anti-vascular endothelial growth factor in patients with advanced cancer. J Clin Oncol. 2001;19:843–850. doi: 10.1200/JCO.2001.19.3.843. [DOI] [PubMed] [Google Scholar]
  • 95.Besse B., Lasserre S.F., Compton P., Huang J., Augustus S., Rohr U.P. Bevacizumab safety in patients with central nervous system metastases. Clin Cancer Res. 2010;16:269–278. doi: 10.1158/1078-0432.CCR-09-2439. [DOI] [PubMed] [Google Scholar]
  • 96.Sajjad M., Pan E., Minton S., Ismail-Khan R. Control of brain metastases for HER2-positive breast cancer with bevacizumab: a report of three patients. J Solid Tumors. 2013;3:1–6. [Google Scholar]
  • 97.Lu Y.S., Chen T.W., Lin C.H. Bevacizumab preconditioning followed by Etoposide and Cisplatin is highly effective in treating brain metastases of breast cancer progressing from whole-brain radiotherapy. Clin Cancer Res. 2015;21:1851–1858. doi: 10.1158/1078-0432.CCR-14-2075. [DOI] [PubMed] [Google Scholar]
  • 98.Ilhan-Mutlu A., Osswald M., Liao Y. Bevacizumab prevents brain metastases formation in lung adenocarcinoma. Mol Cancer Ther. 2016;15:702–710. doi: 10.1158/1535-7163.MCT-15-0582. [DOI] [PubMed] [Google Scholar]
  • 99.Shang X., Wang P., Liu Y., Zhang Z., Xue Y. Mechanism of low-frequency ultrasound in opening blood-tumor barrier by tight junction. J Mol Neurosci. 2011;43:364–369. doi: 10.1007/s12031-010-9451-9. [DOI] [PubMed] [Google Scholar]
  • 100.Sheikov N., McDannold N., Sharma S., Hynynen K. Effect of focused ultrasound applied with an ultrasound contrast agent on the tight junctional integrity of the brain microvascular endothelium. Ultrasound Med Biol. 2008;34:1093–1104. doi: 10.1016/j.ultrasmedbio.2007.12.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Park E.J., Zhang Y.Z., Vykhodtseva N., McDannold N. Ultrasound-mediated blood-brain/blood-tumor barrier disruption improves outcomes with trastuzumab in a breast cancer brain metastasis model. J Control Release. 2012;163:277–284. doi: 10.1016/j.jconrel.2012.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Kobus T., Zervantonakis I.K., Zhang Y., McDannold N.J. Growth inhibition in a brain metastasis model by antibody delivery using focused ultrasound-mediated blood-brain barrier disruption. J Control Release. 2016;238:281–288. doi: 10.1016/j.jconrel.2016.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Auffinger B., Thaci B., Nigam P., Rincon E., Cheng Y., Lesniak M.S. New therapeutic approaches for malignant glioma: in search of the Rosetta stone. F1000 Med Rep. 2012;4:18. doi: 10.3410/M4-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Patil R., Ljubimov A.V., Gangalum P.R. MRI virtual biopsy and treatment of brain metastatic tumors with targeted nanobioconjugates: nanoclinic in the brain. ACS Nano. 2015;9:5594–5608. doi: 10.1021/acsnano.5b01872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Hamilton A.M., Aidoudi-Ahmed S., Sharma S. Nanoparticles coated with the tumor-penetrating peptide iRGD reduce experimental breast cancer metastasis in the brain. J Mol Med Berl. 2015;93:991–1001. doi: 10.1007/s00109-015-1279-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Alsidawi S., Malek E., Driscoll J.J. MicroRNAs in brain metastases: potential role as diagnostics and therapeutics. Int J Mol Sci. 2014;15:10508–10526. doi: 10.3390/ijms150610508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Xing F., Watabe K. miRNAs as biomarkers for brain metastasis of breast cancer. Biomark Med. 2013;7:387–390. doi: 10.2217/bmm.13.43. [DOI] [PubMed] [Google Scholar]
  • 108.Okuda H., Xing F., Pandey P.R. miR-7 suppresses brain metastasis of breast cancer stem-like cells by modulating KLF4. Cancer Res. 2013;73:1434–1444. doi: 10.1158/0008-5472.CAN-12-2037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Hwang S.J., Seol H.J., Park Y.M. MicroRNA-146a suppresses metastatic activity in brain metastasis. Mol Cells. 2012;34:329–334. doi: 10.1007/s10059-012-0171-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Camacho L., Guerrero P., Marchetti D. MicroRNA and protein profiling of brain metastasis competent cell-derived exosomes. PLoS One. 2013;8:e73790. doi: 10.1371/journal.pone.0073790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Zhang X., Yu H., Lou J.R. MicroRNA-19 (miR-19) regulates tissue factor expression in breast cancer cells. J Biol Chem. 2011;286:1429–1435. doi: 10.1074/jbc.M110.146530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Mott J.L., Kobayashi S., Bronk S.F., Gores G.J. mir-29 regulates Mcl-1 protein expression and apoptosis. Oncogene. 2007;26:6133–6140. doi: 10.1038/sj.onc.1210436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Zhang L., Sullivan P.S., Goodman J.C., Gunaratne P.H., Marchetti D. MicroRNA-1258 suppresses breast cancer brain metastasis by targeting heparanase. Cancer Res. 2011;71:645–654. doi: 10.1158/0008-5472.CAN-10-1910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Teplyuk N.M., Mollenhauer B., Gabriely G. MicroRNAs in cerebrospinal fluid identify glioblastoma and metastatic brain cancers and reflect disease activity. Neuro Oncol. 2012;14:689–700. doi: 10.1093/neuonc/nos074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Gregory P.A., Bert A.G., Paterson E.L. The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat Cell Biol. 2008;10:593–601. doi: 10.1038/ncb1722. [DOI] [PubMed] [Google Scholar]
  • 116.Li L., Huang K., You Y. Hypoxia-induced miR-210 in epithelial ovarian cancer enhances cancer cell viability via promoting proliferation and inhibiting apoptosis. Int J Oncol. 2014;44:2111–2120. doi: 10.3892/ijo.2014.2368. [DOI] [PubMed] [Google Scholar]
  • 117.Wood H.A., Bancroft J.B. Activation of a plant virus by related incomplete nucleoprotein particles. Virology. 1965;27:94–102. doi: 10.1016/0042-6822(65)90146-7. [DOI] [PubMed] [Google Scholar]
  • 118.Fong M.Y., Zhou W., Liu L. Breast-cancer-secreted miR-122 reprograms glucose metabolism in premetastatic niche to promote metastasis. Nat Cell Biol. 2015;17:183–194. doi: 10.1038/ncb3094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Xing F., Sharma S., Liu Y. miR-509 suppresses brain metastasis of breast cancer cells by modulating RhoC and TNF α. Oncogene. 2015;34:4890–4900. doi: 10.1038/onc.2014.412. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Chronic Diseases and Translational Medicine are provided here courtesy of Chinese Medical Association

RESOURCES