CSCO expert consensus on the diagnosis and treatment of breast cancer brain metastasis
Consensus

CSCO expert consensus on the diagnosis and treatment of breast cancer brain metastasis

Tao Wang1#, Jiayi Chen2#, Jin Yang3, Minjie Fu4, Wei Hua4, Wang Jia5, Yueping Liu6, Biyun Wang7, Min Yan8, Juan Zhou1, Chunfang Hao9, Jiaxin Chen1, Dan Ou2, Tao Jiang5, Ying Mao4, Zefei Jiang1, Yuee Teng, Qingyuan Zhang, Man Li, Feng Jin, Zefei Jiang, Tao Wang, Juyi Wen, Chunfang Hao, Cuizhi Geng, Yunjiang Liu, Yueping Liu, Jun Zhang, Min Yan, Haibo Wan, Zhigang Yu, Yongmei Yin, Yueyin Pan, Jiayi Chen, Biyun Wang, Xiaojia Wang, Quchang Ouyang, Jia Liu, Shu Liu, Qiang Liu, Kun Wang, Shusen Wang, Jianyun Nie, Hongyuan Li, Xinlan Liu, Gang Sun, Yan Xue, Jin Yang; the CSCO expert panel of breast cancer*

1Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China; 2Department of Radiotherapy, Ruijin Affiliated Hospital of Shanghai Jiaotong University School of Medicine, Shanghai, China; 3Department of Oncology, The First Affiliated Hospital of Xi‘an Jiaotong University, Xi’an, China; 4Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China; 5Department of Neurosurgery, Tiantan Hospital, Beijing, China; 6Department of Pathology, Fourth Hospital Affiliated of Hebei Medical University, Shijiazhuang, China; 7Department of Oncology, Cancer Hospital Affiliated to Fudan University, ShanghaiChina; 8Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China; 9Department of Oncology, Tumor Hospital of Tianjin, Tianjin, China

#These authors contributed equally to this work and should be considered as co-first authors.

*Members of the expert panel: Yuee Teng (The First Affiliated Hospital of China Medical University), Qingyuan Zhang (Heilongjiang Province Cancer Hospital), Man Li (The Second Affiliated Hospital of Dalian Medical University), Feng Jin (The First Affiliated Hospital of China Medical University), Zefei Jiang (the fifth Medical Center of PLA General Hospital),Tao Wang (the fifth Medical Center of PLA General Hospital), Juyi Wen (The Sixth Medical Center of PLA General Hospital), Chunfang Hao (Tumor Hospital of Tianjin), Cuizhi Geng (Fourth Hospital Affiliated of Hebei Medical University), Yunjiang Liu (Fourth Hospital Affiliated of Hebei Medical University), Yueping Liu (Fourth Hospital Affiliated of Hebei Medical University), Jun Zhang (Fourth Hospital Affiliated of Hebei Medical University), Min Yan (The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital), Haibo Wan (Affiliated Hospital of Qingdao University), Zhigang Yu (Second Hospital of Shandong University), Yongmei Yin (Jiangsu Provincial People’s Hospital), Yueyin Pan (The First Affiliated Hospital of University of Science and Technology of China), Jiayi Chen (Ruijin Affiliated Hospital of Shanghai Jiaotong University School of Medicine), Biyun Wang (Cancer Hospital Affiliated to Fudan University), Xiaojia Wang (Zhejiang Province Cancer Hospital), Quchang Ouyang (Hunan Province Cancer Hospital), Jia Liu (Fujian Province Cancer Hospital), Shu Liu (Affiliated Hospital of Guizhou Medical University), Qiang Liu (Sun Yat-sen Memorial Hospital, Sun Yat-sen University), Kun Wang (Guangdong Provincial People’s Hospital), Shusen Wang (Sun Yat-sen Memorial Hospital, Sun Yat-sen University), Jianyun Nie (Yunnan Province Cancer Hospital), Hongyuan Li (The First Affiliated Hospital of Chongqing Medical University), Xinlan Liu (Cancer Hospital of Ningxia Medical University), Gang Sun (Cancer Hospital Affiliated to Xinjiang Medical University), Yan Xue (Cancer Hospital of Xi’an International Medical Center), Jin Yang (The First Affiliated Hospital of Xi’an Jiaotong University).

Correspondence to: Tao Jiang. Department of Neurosurgery, Tiantan Hospital, Beijing, China. Email: taojiang1964@163.com; Ying Mao. Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China. Email: maoying@fudan.edu.cn; Zefei Jiang. Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China. Email: jiangzefei@csco.org.cn.

Abstract: Breast cancer is one of the most common malignancies among women worldwide. According to the International Agency for Research on Cancer, breast cancer affected more Chinese women than any other cancer in 2020. The brain is an increasingly common metastatic sites of breast cancer. Although the risk of developing brain metastases (BMs) is lower in breast cancer than in lung cancer and melanoma, due to its high prevalence, it is the second most common cause of BM among solid tumors, being second only to lung cancer. The incidence of breast cancer brain metastasis (BCBM) differs by molecular subtype. Half of patients with advanced human epidermal growth factor receptor-2 (HER2)-positive and one-third of patients with triple-negative breast cancer (TNBC) develop BM. The clinical manifestations of leptomeningeal metastasis (LM) are often non-specific and may manifest as a variety of signs and symptoms, mainly including brain parenchyma involvement and meningeal irritation syndromes cranial nerve involvement, increased intracranial pressure, and progressive brain dysfunction. Therefore, the Chinese Society of Clinical Oncology (CSCO) Breast Cancer Committee has developed this expert consensus on BM, in an effort to improve the overall prognosis of BCBM and promote the standardized diagnosis and treatment of this disease. During the development of this expert consensus, we carried out a comprehensive literature review and referred to some of the most authoritative guidelines in China and abroad. In this consensus, we will discuss clinical manifestations, imaging examinations, pathological diagnosis, treatments, prognosis, follow-up and monitoring. We hope this consensus will be of help to all the clinicians majored in breast cancer and other similar professions.

Keywords: Breast cancer (BC); brain metastases (BMs); expert consensus


Received: 29 June 2022; Accepted: 28 July 2022; Published: 30 July 2022.

doi: 10.21037/tbcr-22-30


Introduction

Breast cancer metastasis to the brain, including breast cancer brain metastasis (BCBM) and leptomeningeal metastasis (LM), occurs in 10% to 20% of breast cancer (BC) cases. About 80% of breast cancer brain metastases (BMs) occur in the cerebral hemispheres, 15% in the cerebellum, and 5% in the brain stem. In a study by Cacho-Díaz et al., 47.6% of the patients had multiple lesions and 26.4% had solitary lesions, most of which were located at the cerebral cortex-medulla junction, where the vascular branches are relatively narrow. Only a small proportion of cases had LM (about 6.9%), and 3.6% had both BMs and LM, which was associated with a poorer prognosis (1,2).

The incidence of BCBM differs by molecular subtype. About 15% of patients are diagnosed with advanced hormone receptor (HR)-positive BC, 50% of patients with advanced human epidermal growth factor receptor-2 (HER2)-positive BC, and one-third of patients with triple-negative breast cancer (TNBC) develop BM (3,4). The occurrence of BCBM is also associated with BRCA1 (BReast CAncer gene 1) and BRCA2 (BReast CAncer gene 2) mutations. BRCA1/2 mutation carriers have been found to have a significantly higher rate of BM (5).


Methods

Professor Zefei Jiang, Vice President and Secretary General of the Chinese Society of Clinical Oncology (CSCO) and Chairman of the CSCO Breast cancer Expert Committee, took the lead in formulating an expert consensus on BCBM. On the 17th March 2022, the BM consensus expert group held an online meeting to define the consensus on the diagnosis and treatment of BCBM from the epidemiological characteristics, clinical manifestations, diagnostic methods, treatment, prognosis, and monitoring follow-up five aspects, and written by the experts’ division of labor. Finally, the CSCO expert consensus on the diagnosis and treatment of BCBM was formed through discussion and summary of several online meetings.


Results and discussion

Clinical manifestations

Patients with BCBM experience some common clinical manifestations, which may vary among individual patients due to differences in the histobiological features, location, size, and the number of metastatic lesions. The most common symptoms in patients with BCBM include headache (35%), vomiting (26%), nausea (23%), hemiplegia (22%), visual changes (13%), and seizures (12%) (6). Common clinical manifestations include headache, symptoms related to increased intracranial pressure, focal neurological dysfunction, seizures, and psychiatric disorder.

The clinical manifestations of LM are often non-specific and may manifest as a variety of signs and symptoms, mainly including brain parenchyma involvement and meningeal irritation syndromes (e.g., headache, vomiting, nuchal rigidity, cognitive disorders, clouding of the consciousness, and the onset of symptomatic epilepsy), cranial nerve involvement, increased intracranial pressure, and progressive brain dysfunction. Sometimes it is difficult to differentiate LM from BM or treatment-associated toxicities. In some cases, patients with LM present only with progressive neck and shoulder pain. If the tumor also spreads along the spinal membrane, spinal cord, and spinal nerve root, stimulation can occur, manifesting as radicular pain, segmental sensory disturbance, limb numbness, sensory ataxia, and reduced or absent deep tendon reflexes (7-10).

Imaging examinations

Magnetic resonance imaging (MRI)

Cranial MRI plays an important role in the diagnosis, response evaluation, and post-treatment monitoring and follow-up of patients with metastases (11). It is the preferred imaging modality for BM and LM due to being radiation-free and offering high-resolution soft-tissue imaging, multi-parameter imaging, and high sensitivity; however, it is not feasible for patients with magnetically sensitive metals in their bodies or those with claustrophobia. Meningeal metastases of breast cancer can be divided into LM, dural metastases, and mixed meningeal metastases. On MRI, dural metastases are shown as a continuous uneven linear meningeal thickening on the convex surface of the brain or tentorium, which typically does not extend into the sulcus or the brain fissure. The involvement can be extensive, and in severe cases, local nodules or irregular masses may form, which are markedly enhanced after contrast application. Leptomeningeal metastases on MRI are mostly arc-like enhancements along the surface of the gyrus, sulci, fissures, cistern, and subependymal zone. They are irregular in shape, and nodules can also form. Invasion of adjacent brain tissues can cause local parenchymal edema. In some cases, meningeal metastases of breast cancer may only manifest as hydrocephalus without meningeal enhancement, which is difficult to diagnose with MRI alone, and cerebrospinal fluid (CSF) examination may be helpful.

Computed tomography (CT)

On CT, BCBM typically manifests as iso- or low-density shadows, although the density may be slightly elevated in the presence of hemorrhage. The typical manifestations of BCBM on contrast-enhanced CT scans are small tumors and large edema with an obvious enhancement of solid components, which are manifested as nodular, annular, or irregular enhancements. In cases where patients have contraindications to MRI, CT remains a valuable adjunct. Notably, small isodense metastases are difficult to detect, and metastases in the posterior fossa are often overlooked due to the presence of surrounding bony structures. Calcification of BM from breast cancer is extremely rare.

Positron emission tomography/computed tomography (PET-CT)

PET-CT can provide both anatomical and metabolic information and is valuable for assessing systemic tumor load. However, due to the predominance of glucose metabolism in cerebral gray matter, the background radioactivity of normal brain tissue is high, which makes the differentiation between brain tumors and the surrounding normal brain tissues difficult. As a result, imaging of local structures is often required. In patients with clinical symptoms of BM, a timely head MRI or CT scan should be performed to determine the systemic tumor load.

Pathological diagnosis

For patients who have a high clinical suspicion of BCBM, a biopsy of the metastatic lesions is recommended, if clinically feasible, to confirm the diagnosis. Histological morphology examination and immunohistochemical staining of the primary tumor and metastases should also be performed to determine whether it is BCBM. Panels of immunohistochemical markers are recommended; these may include cytokeratin 7 (CK7), GATA binding protein 3 (GATA3), gross cystic disease fluid protein 15 (GCDFP-15), mammaglobin, trichorhinophalangeal syndrome 1 (TRPS1), and SRY-related HMG-box (SOX10). For patients from whom biopsy tissue cannot be obtained, CSF detection, along with cytological and immunohistochemical staining, may be performed to identify BCBM.

The high degree of temporal and spatial heterogeneity among advanced breast cancer may result in inconsistent molecular typing results between metastases and primary lesions. Re-assessment of the molecular subtype of a metastatic lesion based on estrogen receptor (ER), progesterone receptor (PR), HER2, and KI67 status is recommended. In particular, the HER2 status of metastases should be determined wherever possible. Immunohistochemistry plus in situ hybridization (ISH) is recommended to detect HER2 status (12): (I) HER2 positivity: IHC 3+ or IHC 2+ AND ISH positive; (II) low HER2 expression: IHC 1+ or 2+ AND ISH negative; and (III) HER2 negativity: IHC 0.

Treatment

Both systemic therapy and treatment of BM are important for the treatment of BCBM, and multidisciplinary treatment is always preferred. Localized treatments including surgery, whole-brain radiation therapy (WBRT), and stereotactic radiosurgery (SRS) remain the mainstay of treatment for BCBM. Medical therapies have also shown efficacy in treating certain types of breast cancer. The aim of treatment for BCBM is same as that for advanced breast cancer: to improve quality of life and prolong survival.

Surgical treatment

Surgical removal of BM can lower intracranial pressure, relieve symptoms, prevent focal neurological dysfunction and epilepsy, and reduce steroid use. Surgical specimens are useful in making a definite pathological diagnosis, and they can also be used in molecular pathology and targeted therapy. Surgery is an effective treatment for patients with solitary BM, especially those with giant lesions and compressive symptoms, for whom the benefit is higher than it is for patients with multiple BM or systemic symptoms. Patchell et al. randomized 48 patients with BM (including 3 BCBM cases) into surgery, whole brain radiation therapy (WBRT), and biopsy with WBRT groups, and found that the recurrence rate in the surgery group was significantly lower than that in the WBRT group (20% vs. 52%) and the median survival was significantly longer (40 vs. 15 weeks) (13). Patients with two or three BMs whose general condition is satisfactory can also benefit from surgery, with comparable outcomes to those of patients with solitary BMs (level IIIb) (14).

Margin status is an important prognostic factor, with the postoperative residual tumor being significantly associated with tumor recurrence and progression (level IIIb) (15). A meta-analysis (16) found that in patients with posterior fossa metastases, radical resection (R0) reduced the recurrence rate; the rate of leptomeningeal dissemination was only 5 to 6%, which was significantly lower than that after partial resection (level IIIb). In another study, residual tumor after metastasis extirpation was observed on early postoperative MRI in nearly 20% of patients and was significantly correlated with local recurrence (level IIIb). Multimodal imaging and navigation techniques such as preoperative functional MRI, intraoperative neuronavigation, and pyramidal tract reconstruction (level IV) can protect brain function and reduce complications while completely resecting BM.

Magnetic resonance-guided laser-induced thermotherapy (LITT) is an emerging treatment technique. It represents a new treatment opportunity for patients with deep brain lesions, older or frail patients who cannot tolerate a long surgical operation, and those with radiation-induced necrosis. A case-control study reported that laser-induced thermotherapy was as effective as a surgical resection for local control of BM. The 6-month local control rates of recurrent BM and radiation-induced necrosis ranged from 54% to 81.9% and 56.5% to 100%, respectively (17,18). In a meta-analysis and a retrospective clinical trial, the performance of laser-induced thermotherapy was equal or even superior to that of bevacizumab for the control of radiation-induced necrosis in patients with BMs (19,20). Moreover, laser-induced thermotherapy treatment did not reduce the Karnofsky Performance Status (KPS) score or the quality of life of patients (17), and to a certain extent, reduced the use of steroids (17,21). Another multicenter prospective clinical trial showed that laser-induced thermotherapy effectively controlled BMs in areas difficult to reach by surgery (22). Brain edema caused by radiation-induced necrosis can also be effectively controlled by laser-induced thermotherapy therapy. Some self-developed LITT treatment system in China has shown that it is effective in controlling both newly diagnosed and recurrent brain metastases, as well as radiation-induced necrosis. According to the 2021 guidelines on the treatment of BMs jointly released by the European Association of Neuro-Oncology (EANO) and the European Society for Medical Oncology (ESMO), laser-induced thermotherapy is a new technology for the treatment of recurrent BMs and radiation-induced necrosis, but its value needs to be further investigated.

Radiotherapy

Goals

The overall goals of radiotherapy for BCBM include intracranial lesion control, neurological symptom improvement, cognitive function and quality of life preservation, and maximization of survival benefits. In clinical practice, treatment strategies based on different treatment goals are often developed based on individual patients’ expected survival. Some non-prospective studies have suggested that the Diagnosis-Specific Graded Prognostic Assessment (DS-GPA) score may improve the treatment decision-making for patients with BM through prognostic stratification (23,24). In a survey on local therapies for multiple BM, one-third of physicians preferred to use the Recursive Partitioning Analysis (RPA) or GPA score as the basis for selecting SRS (25). Therefore, an updated Graded Prognostic Assessment (Breast GPA) is recommended to determine treatment goals and guide the treatment of BM in a stratified, reasonable, and orderly manner. This scoring system will be continuously optimized with the innovation of medical therapies and local treatment methods.

Currently, radiotherapy for BCBM includes stereotactic radiotherapy (SRT) and WBRT with or without hippocampal avoidance.

SRT

Owing to its advantages including more precise positioning, higher dose, a shorter course of treatment, and lower toxicity, SRT can effectively protect cognitive function while controlling the progression of intracranial lesions and relieving neurological symptoms. Consequently, it has gradually replaced WBRT as a mainstay of local treatment for BM. With regard to dose fractionation, SRT is divided into SRS and fractionated stereotactic radiotherapy (FSRT).

(I) Postoperative SRT
Half of the patients with BMs who undergo surgical resection alone suffer from intracranial local recurrence within the first 6 months after surgery (26). Postoperative WBRT can decrease both the risk of local recurrence and the risk of intra-cranial distant recurrence by 50%, as well as prolong survival (27-29). Several observational studies have explored the efficacy of postoperative SRT (30-33). After single- or multiple-fraction postoperative SRT, the 1-year local control rates of the intracranial surgical bed reached 73% to 90%, which were comparable to the local control rates of lesions after postoperative WBRT. Mahajan et al. (34) analyzed 132 patients with one to three BMs who were randomized to receive postoperative SRS or undergo observation and confirmed that postoperative SRS significantly improved the local control of the surgical cavity. The NCCTG N107C/CEC3 study (35) enrolled 194 patients who had undergone surgery for BM (at least one resected BM and no more than three unresected BM). While there was no difference in overall survival between the treatment groups, the median cognitive-deterioration-free survival rate and the cognitive deterioration rate at 6 months were superior in the SRS group. The surgical site control rates were lower in the SRT group than in the WBRT group (80% vs. 87% at 6 months, and 61% vs. 81% at 12 months), which was likely due to 40% of the patients had an operative cavity width greater than 3 cm.

Therefore, we recommend that for patients with a limited number of BCBM, postoperative radiotherapy is necessary to improve intracranial local control. Compared with postoperative WBRT, postoperative tumor bed SRT can achieve equally effective local control of the surgical cavity while being associated with fewer cognitive impairments without compromising survival. Therefore, if postoperative SRT is technically accessible, it should be the therapeutic choice of priority, followed by WBRT; however, the risk of intracranial recurrence should not be neglected, especially for patients with a surgical cavity larger than 3 cm. Close follow-up is thus required.

(II) SRT alone
The RTOG 9508 study (36) confirmed the efficacy and safety of WBRT + SRS in patients with one to three BMs. Subsequent clinical studies compared the efficacy and toxicity of SRS with WBRT versus SRS alone in the same population (26,37-39). Compared with SRS alone, SRS with WBRT was found to reduce the risk of intracranial disease progression by about 50%; however, it not only failed to prolong overall survival but also increased the risk of toxic effects, such as cognitive decline. Several randomized trials have supported the use of SRS as initial therapy for patients with a small number of BMs who are suitable candidates (Table 1). Meanwhile, due to the toxicities associated with WBRT, it is generally preferable to defer the use of WBRT for most patients who have a limited number of BMs and can be initially treated with SRS. Most of these previous randomized clinical trials included patients with no more than 4 BMs of no larger than 3 cm in diameter (Table 1).

Table 1

Prospective randomized controlled trials on radiotherapy for brain metastases

Study n Inclusion criteria Groups Radiotherapy dosage Intracranial local
control (RR or PFS)
OS
Patchell et al. (13) 48 >18 y; solitary; KPS ≥70 Surgery + WBRT (n=25); WBRT (n=23) 36 Gy/12 F 5/25 vs. 12/23; P<0.02 40 vs. 15 w; P<0.01
Vecht et al. (28) 63 >18 y; solitary; PS ≤1 Surgery + WBRT (n=32); WBRT (n=31) 40 Gy/20 F NA 10 vs. 6 m; P=0.04
Mintz et al. (29) 84 <80 y; solitary; KPS ≥50 Surgery + WBRT (n=41); WBRT (n=43) 30 Gy/10 F NA 5.6 vs. 6.3 m; P=0.24
Patchell et al. (27) 95 >18 y; solitary; KPS ≥70 Surgery + WBRT (n=49); Surgery (n=46) 50.4 Gy/28 F 18% vs. 70%; P<0.001 48 vs. 43 w; P=0.39
NCCTG N107C/CEC3 study; Brown et al. (35) 194 ≥18 y; PS ≤2; including 1 resected BM with a surgical cavity <5 cm; 0–3 unresectable BMs with max. diameter <3 cm (77% were solitary) Surgery + SRS (n=98); Surgery + WBRT (n=96) WBRT: 30 Gy/10 F and 37.5 Gy/15 F; SRS: 12–24 Gy* 6.4 vs. 27.5 m; P<0.0001 12.2 vs. 11.6 m; P=0.70
Kayama et al. (40) 271 PS ≤2 or PS =3 only because of neurological symptoms; 1–4 BMs
were surgically removed, and 1 lesion sized >3 cm
Surgery + SRS for residual tumor (n=134); Surgery + WBRT (n=137) WBRT: 37.5 Gy/
15 F
4.0 vs. 10.4 m 15.6 vs. 15.6 m
EORTC22952-26001 (26) 359 PS ≤2; 1–3 BMs;
surgical resection
(n=199) or SRS (n=160)
WBRT (n=180); Wait-
and-see (n=179)
30 Gy/10 F 4.6 vs. 3.6 m; P=0.02 10.9 vs. 10.7 m; P=0.89
Mahajan et al. (34) 132 KPS ≥70; surgical resection of 1–3 BMs SRS (n=64); Wait-and-see (n=68) SRS: 12–18 Gy 72% vs. 43%; P=0.015 17 vs. 18 m; P=0.24
RTOG 9508 (36) 333 ≥18 y; KPS ≥70;
1–3 BMs ≤4 cm
WBRT + SRS (n=167); WBRT (n=164) WBRT: 37.5 Gy/
15 F; SRS: 15–
24 Gy
1 y: 82% vs. 71%; P=0.01 6.5 vs. 5.7 m; P=0.14
Aoyama et al. (38) 132 ≥18 y; KPS ≥70;
1–4 BMs ≤3 cm
SRS + WBRT (n=65);
SRS (n=67)
WBRT: 30 Gy/10 F; SRS: 18–25 Gy 1 y tumor bed recurrence: 46.8% vs. 76.4%; P<0.001 7.5 vs. 8.0 m; P=0.42
Chang et al. (39) 58 ≥18 y; KPS ≥70; RPA grade 1–2; 1–2 BMs SRS + WBRT (n=28);
SRS (n=30)
WBRT: 30 Gy/12 F; SRS: 15–20 Gy 1 y: 73% vs. 27% NA
Brown et al. (37) 213 ≥18 y; PS ≥2; 1–3 BMs ≤3 cm SRS + WBRT (n=102); SRS (n=111) WBRT: 30 Gy/12 F; SRS: 18–24 Gy 1 y: 84.6% vs. 50.5%; P<0.001 7.4 vs. 10.4 m; P=0.92
NCT02353000 (41) 29 ≥18 y; KPS ≥70;
4–10 BMs ≤30 cm3
SRS (n=15);
WBRT (n=14)
SRS: 15–24 Gy/1 F and 24 Gy/3 F; WBRT: 20 Gy/5 F 1 y: 50% vs. 78% (P=0.22) 1 y: 57% vs. 31% (P=0.52)
NRG ONCOLOGY CC001 (42) 518 ≥18 y; KPS ≥70 HA-WBRT + memantine hydrochloride (n=261); WBRT + memantine hydrochloride (n=257) 30 Gy/10 F 5.0 vs. 5.3 m; P=0.21 6.3 vs. 7.6 m; P=0.31

*, surgical cavity SRS (volume/dose): <4.2 cm3/20–24 Gy; 4.2–7.9 cm3/18 Gy; 8.0–14.3 cm3/17 Gy; 14.4–19.9 cm3/15 Gy; 20–29.9 cm3/14 Gy; ≥30 cm3 and not exceeding 5 cm/12 Gy. KPS, Karnofsky Performance Status; PS, performance status; BM, brain metastasis; SRS, stereotactic radiosurgery; RPA, Recursive Partitioning Analysis; WBRT, whole-brain radiation therapy; HA, hippocampal avoidance; RR, regional recurrence; PFS, progression-free survival; NA, not available; m, month; y, year; OS, overall survival; w, week.

The strongest evidence currently supporting the use of SRS in patients with more than 4 BMs comes from a Japanese prospective single-arm multicenter study [JLGK0901 (43)] which included 1,194 patients with between 1 and 10 BMs. Compared with patients with 2 to 4 BMs, those with 5 to 10 BMs had similar overall survival, toxicities, and subsequent central nervous system failure rates after SRS. Another randomized controlled study [NCT02353000 (41)] compared the toxicity and efficacy of WBRT versus SRS in patients with 4 to 10 BMs. Although the study was prematurely ended due to poor accrual, analysis of the 29 patients enrolled showed that in the SRS group, the actuarial 1-year brain salvage-free survival rate was 50% and the 1-year survival rate was 57% with good quality of life maintained.

Therefore, we recommend that effective SRT can safely postpone WBRT for patients with a limited number of BMs (i.e., <4) while ensuring survival, thereby allowing them to avoid WBRT-related neurotoxicity. For patients with multiple BMs (>4), SRT has shown good efficacy and may be recommended when technically feasible.

(III) Fractionation
No prospective randomized controlled trials have investigated the clinical benefits of different dose/fractionation regimens of SRT. The dose of postoperative single-fraction SRS can refer to the NCCTG N107C/CEC3 study (35) (Table 1). Only some non-randomized studies (33,44-46) have explored postoperative multiple-fraction SRT, and a study to evaluate the fractionation regimens of postoperative SRT (NCT04114981) is ongoing.

In a dose-escalation study of SRT alone (RTOG 9005), the maximum tolerated doses of single-fraction SRS were 24, 18, and 15 Gy for BMs with a maximum diameter of equal or less than 20, 21 to 30, and 31 to 40 mm, respectively (47). Meanwhile, in the above-mentioned multiple prospective studies, the single-fraction dose was 20 to 24 Gy for BMs with a maximum diameter of equal or less than 2cm or a volume of less than 4 cm3 (26,37,38,43). In a large-scale retrospective study, the local control of tumors no larger than 2 cm was satisfactory using a 24-Gy/single-fraction scheme; however, tumors larger than 2 cm were less effectively controlled by a 15 to 18 Gy/single-fraction scheme (48). Multi-fractional SRT has shown a higher local control rate and lower risk of brain necrosis in this population, especially for solitary BMs with a maximum diameter of larger than 3 cm (49). Acceptable fractions of SRT include 27 Gy/3 Fx or 30 Gy/5 Fx and 3,500 cGy/5 Fx (50). In addition to the maximum diameter, the intracranial substructure of the lesion and its tolerable dose also affect the recommended dose of SRS/SRT. Among the different fractionation schemes of SRT, a biologically effective dose (BED10) of equal or more than 50 Gy was associated with better local tumor control (51).

Therefore, we recommend single-fraction SRS of 20 to 24 Gy for BMs with a maximum diameter of equal to or small than 2 cm, single-fraction SRS of 18 Gy, or multi-fraction SRT for lesions with maximum diameter of 2.1 to 2.9 cm, and multi-fraction SRT for lesions with a maximum diameter of 3.1–4.0 cm. Since previous prospective studies using single-fraction SRS for the treatment of BMs have not included lesions larger than 4 cm, we recommend multi-fraction SRT for these lesions if technically feasible. Nevertheless, SRT is currently discouraged for tumors larger than 6 cm due to lack of evidence (52).

WBRT with or without hippocampal avoidance

Although the indications for WBRT are continually challenged by SRT (53). WBRT remains an appropriate and optional treatment for patients with diffuse BMs with or without leptomeningeal involvement. Research has shown that patients with diffuse BMs (arbitrarily defined as 20 or more lesions) have a chance to survive long enough after WBRT (54). The cognitive decline caused by WBRT has been widely recognized, and less than 10% of BMs are within 5 mm of the hippocampus. In the RTOG 0933 study (55), cognitive function deterioration was less significant when the dose of hippocampal irradiation was reduced. The NRG ONCOLOGY CC001 study (42) further confirmed that hippocampal avoidance WBRT (HA-WBRT) combined with memantine therapy could improve cognitive function without affecting local control or overall survival.

In general, for patients with diffused BMs who have a good prognosis and whose closest distance between the lesions and the hippocampus is not less than 1 cm, we recommend the use of HA-WBRT combined with memantine, as this protocol has high efficacy and low toxicity. Also, HA-WBRT combined with SRT for some metastases can be considered to obtain a better local control rate. The dose of WBRT can refer to the NCCTG N107C/CEC3 study (35), with the recommended dose being 30 Gy/10 Fx (Table 1).

The QUARTZ study (56) confirmed that WBRT did not show an advantage over supportive care in patients with poor prognosis. For these patients, more reasonable options include palliative or end-of-life care; or, for patients with symptomatic BMs, short-course WBRT (i.e., 20 Gy/5 Fx) may be applied.

Radiotherapeutic strategy for BMs may be particularly important for patients with a limited number of BMs (i.e., patients with 1 to 4 anatomically independent metastases). More treatment options are available for these patients. Stratified and classified treatment may be arranged based on their general conditions, BM prognosis, and treatment willingness. Figure 1 summarizes the recommendations for local therapy in patients with a limited number of BMs. During SRS, FSRT, or WBRT with or without hippocampal avoidance, the total BED to the metastases always affects local control.

Figure 1 Recommendations on local therapy for patients with a limited number of brain metastases. PS, performance status; HA-WBRT, hippocampal avoidance whole-brain radiotherapy; SRS, stereotactic radiosurgery; SRT, stereotactic radiotherapy.
Timing of radiotherapy and systemic therapy

Since the blood-brain barrier (BBB) blocks the entry of therapeutic substances into the brain, medical therapy was previously believed to have a very limited effect on intracranial lesions from breast cancer. However, preclinical studies have shown that exposure to radiation can damage the BBB and enhance drug permeability (57,58), which provides a theoretical basis for the application of drugs combined with brain radiotherapy in the treatment of BMs (59,60). Many clinical studies have demonstrated that small molecule tyrosine kinase inhibitors (TKIs) such as lapatinib, tucatinib, epertinib, and pyrotinib, as well as macromolecular monoclonal antibodies, have certain therapeutic effects on intracranial lesions from HER2-positive breast cancer (61-67).

In general, targeted therapies with small molecule TKIs, macromolecular monoclonal antibodies, and/or antibody-drug conjugates (ADCs) are effective for HER2-positive BCBM, which also justifies the view that systemic therapy can be used upfront and intracranial radiotherapy may be deferred for HER2-positive patients. Therefore, we recommend for patients with newly diagnosed BMs from HER2-positive breast cancer, anti-HER2 therapy should be the treatment of choice if local symptoms are minor and under control; for patients with other molecular subtypes of BCBM, intracranial radiotherapy should not be postponed due to the lack of effective systemic treatments.

Medical treatment

Chemotherapy

Chemotherapy drugs have poor BBB permeability because they have large molecular weights, carry charges, and can easily bind to albumin. Therefore, chemotherapy alone typically has poor effect in treating BMs. There is no evidence that anthracyclines and taxanes can penetrate the BBB (68). Furthermore, since anthracyclines and taxanes are widely used in adjuvant or salvage therapy, few studies have investigated their roles in BM treatment. Other chemotherapeutic drugs such as capecitabine, platinum, topotecan, methotrexate, and temozolomide, when used alone or in combination, have achieved an objective response rate (ORR) of 4% to 55% in patients with BCBM, and the reported progression-free survival (PFS) is less than 4 months (69-75). However, such evidence has been obtained from clinical exploratory studies with small sample sizes, most of which were single-arm trials that used chemotherapeutic drugs in combination with radiotherapy. Therefore, there is no sufficient evidence to support the use of a single conventional chemotherapeutic agent as the mainstay of treatment for BMs.

HER2-targeted therapy

For patients with BMs from HER2-positive breast cancer, HER2-targeted therapies have shown definite efficacy. Drugs with HER2 inhibitory activity fall into three categories: TKIs, monoclonal antibodies, and ADCs.

(I) Epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs)
The four currently approved EGFR-TKIs have achieved good efficacy in patients with BCBM.

Lapatinib is a reversible, dual TKI of human epidermal growth factor receptor type 1 (HER1) and type 2 (HER2). A pooled analysis of 799 patients showed that lapatinib was effective in patients with BMs who have received different lines of different therapies (76). In a prospective single-arm phase 2 study (LANDSCAPE) (77), the combination of lapatinib plus capecitabine in patients who had HER2-positive metastatic breast cancer with BMs who had not previously been treated with WBRT achieved a central nervous system objective response rate (CNS-ORR) of 57.1% and postponed radiotherapy by 8.3 months. The LANDSCAPE study, for the first time, demonstrated the therapeutic effect of a small-molecule TKI on BMs.

Pyrotinib is an irreversible potent TKI targeting HER1, HER2, and HER4. The prospective, randomized, controlled, phase III PHOEBE study (78) confirmed that pyrotinib was significantly more effective than lapatinib. The phase III PHENIX study showed that pyrotinib could delay the progression of intracranial lesions in patients with asymptomatic BMs (79). In a multicenter, single-arm, two-cohort, phase 2 trial (PERMEATE), pyrotinib combined with capecitabine achieved a CNS-ORR of 42.1% and a PFS of 5.6 months in patients with BMs that progressed after radiotherapy; in patients with radiotherapy-naive HER2-positive BMs, the CNS-ORR was 74.6% and the PFS was 11.3 months, which were comparable to those in patients with extracranial lesions (80). Thus, pyrotinib plus capecitabine can serve as the preferred systemic treatment for EGFR TKI-naïve active BMs with controllable local symptoms.

Neratinib is an irreversible TKI also targeting HER1, HER2, and HER4. The phase III NALA study showed fewer interventions for central nervous system disease occurred in the neratinib group (81). A single-arm study of neratinib in the treatment of BMs that progressed after radiotherapy showed that the CNS-ORR was 33% to 49% and the median PFS was 3.1 to 5.5 months (82).

Tucatinib is a highly selective TKI targeting HER2, which has shown good control of BMs in patients with advanced breast cancer in the second-line and later setting (64). In the phase III HER2CLIMB study that enrolled 291 (47%) patients with BMs, the addition of tucatinib to trastuzumab plus capecitabine significantly prolonged the time to intracranial progression; the CNS-PFS increased from 4.2 to 9.9 months and the overall survival prolonged from 12.0 to 18.1 months (83). Accordingly, tucatinib has been approved for the treatment of BCBM by the US Food and Drug Administration.

(II) Monoclonal antibodies
Macromolecular monoclonal antibodies provide no distinct advantage over small-molecule TKIs in penetrating the BBB. No study has shown that macromolecular monoclonal antibodies can significantly improve brain lesions. Trastuzumab and pertuzumab are monoclonal antibodies targeting the HER2 extracellular domains (ECDs) IV and II, respectively. In the phase III trial CLEOPATRA, the combination with pertuzumab delayed the median time to development of CNS metastases at the first site of disease progression (84); in the PHEREXA study, the addition of pertuzumab to trastuzumab and capecitabine showed a trend in PFS benefit (85); in a single-arm, prospective, phase II study, however, patients with HER2-positive metastatic breast cancer with BMs and CNS progression despite prior radiotherapy received pertuzumab plus high-dose trastuzumab (6 mg/kg weekly), and the CNS-ORR was only 11%, showing a non-significant result (67).

(III) ADCs
ADCs containing trastuzumab and a cytotoxic payload have shown promising therapeutic effects in patients with stable BMs. Trastuzumab emtansine (T-DM1) is an ADC composed of trastuzumab and microtubule polymerization inhibitor DM1. In two phase III studies, a total of 443 patients with asymptomatic BMs were treated with T-DM1, and the median PFS was 5.5 to 5.9 months; in 126 patients with measurable BMs, the best overall response rate was 21.4% (86,87).

Trastuzumab deruxtecan (T-Dxd) is an ADC composed of trastuzumab and the topoisomerase I inhibitor deruxtecan. It has shown excellent efficacy in patients with stable BMs after treatment. In the DESTINY-Breast01 and DESTINY-Breast03 studies, 67 patients with stable, asymptomatic BMs after local treatment were enrolled. The PFS reached 15 to 18.1 months, and in patients with intracranial measurable lesions, the CNS-ORR reached 46.7% to 67.4%, which showed a significant advantage of T-Dxd over T-DM1 (88,89). In existing studies, T-Dxd has shown good efficacy for treating BMs, and efficacy trials in patients with newly-diagnosed BMs or progressive disease after local therapy are still ongoing.

Other targeted therapies

None of the currently approved targeted therapies for HER2-negative advanced breast cancer have shown definite effectiveness in treating BMs. The incidence of BMs is relatively low in patients with HR-positive breast cancer, and BMs usually appear late in the process of tumor recurrence and metastasis. CDK4/6 inhibitors have become a standard of care for patients with advanced HR-positive breast cancer. The CDK4/6 inhibitor abemaciclib was found to be able to penetrate the BBB, resulting in a CNS-ORR of 5.2% in patients with ER+/HER2− BC (90). However, its efficiency is not satisfactory and needs further investigation.

In a substudy from the phase 3 IMpassion130 trial, there was no clinical benefit from the PD-L1 inhibitor atezolizumab in the BM subgroup (91). Two phase III studies of poly (ADP-ribose) polymerase (PARP) inhibitors enrolled patients with HER2-negative metastatic breast cancer and a germline BRCA mutation (92,93). Compared with standard therapy, olaparib and talazoparib prolonged PFS in patients with BMs (94). Also, the anti-angiogenic drug bevacizumab can relieve brain edema caused by radiotherapy (95). When used in combination with chemotherapy, it yielded a CNS-ORR of 47% to 77% and a PFS of 5.6 to 6.1 months in patients with BCBM who progressed after WBRT (96,97). For patients with HER2-negative BMs, targeted therapies have limited value and there is a paucity of evidence. Local treatment of intracranial lesions is preferred, and medical treatment options may be comprehensively considered based on the systemic conditions.

Intrathecal medications

Intrathecal injection refers to the direct injection of drug molecules into the subarachnoid space, to thereby increase the drug concentration in the CSF to kill tumor cells. Intrathecal therapy is widely used for the treatment of LM. Intrathecal trastuzumab can be considered for patients with LM from HER2-positive breast cancer. In a meta-analysis of 58 patients who received intrathecal trastuzumab, 55% showed clinical remission, suggesting that intrathecal trastuzumab was safe and effective (98). Intrathecal administration of chemotherapy drugs (e.g., methotrexate and cytarabine) can be considered in patients with HER2-negative breast cancer (99,100). However, intrathecal therapy can cause a wide spectrum of adverse effects, such as neurotoxicity; therefore, co-administration with glucocorticoids during chemotherapy may be helpful.

Symptomatic and supportive treatment

During disease diagnosis and treatment, patients with BCBM often suffer from a variety of symptoms, which undermine their quality of life and can even become life-threatening. Therefore, symptomatic and supportive treatment is an important component of the whole-course management of BCBM.

Brain edema caused by BMs increases intracranial pressure, which can lead to symptoms such as headache, nausea, and vomiting, and raise the risk of seizures. Initially, aggressive dehydration and diuretic therapies (e.g., mannitol, glycerol fructose, and furosemide) should be applied to lower the intracranial pressure. For instance, 125 to 250 mL of 20% mannitol may be intravenously administered every 6 to 8 hours according to the patient’s symptoms, and plasma electrolyte concentrations and urine output should be closely monitored. Glucocorticoids, especially dexamethasone, can alleviate cerebral edema, improve quality of life, and reduce meningeal irritation, although they do not improve prognosis (101,102). Dexamethasone is widely used, often in combination with mannitol. Glucocorticoids should only be used when specifically indicated, and at the lowest dose possible, for the shortest possible time. Insufficient evidence exists to make a treatment recommendation for patients with BMs who are asymptomatic without mass effect. Glucocorticoid use before surgical resection of BMs can alleviate preoperative and postoperative cerebral edema, and glucocorticoid administration during radiotherapy can mitigate early radiotherapy reactions. Intrathecal chemotherapy is an important treatment for meningeal metastases. Administration of glucocorticoids during intrathecal injection of chemotherapy drugs can reduce chemotherapy-induced neurotoxicities and relieve symptoms. Nevertheless, it is necessary to be alert to possible adverse reactions to glucocorticoids, including peptic ulcers and elevated blood sugar. Furthermore, glucocorticoids must be used with caution in patients with diabetes. Furosemide is administered routinely as a rapid intravenous bolus in doses of 20 to 40 mg, and its dosage is adjusted according to the increased intracranial pressure, clinical symptoms, and 24-h urine output; however, changes in plasma electrolytes, especially hyponatremia and hypokalemia, must be closely monitored. Bevacizumab has also been shown to reduce cerebral edema and improve radiation-induced necrosis (103). A ventriculoperitoneal shunt can provide durable relief for symptomatic hydrocephalus. For headaches, nausea, and vomiting that are not promptly relieved after the above treatments, symptomatic treatments such as antiemetics and analgesics can be administered.

Managing epilepsy is integral to the diagnosis and treatment of BM. Since antiepileptic drugs cannot lower the risk of seizures in patients with BM without epilepsy symptoms, they are generally only used in patients who exhibit symptoms of epilepsy and are not recommended as primary prevention (101-104). Seizures should be treated with anticonvulsants that do not interact with systemic therapy (e.g., levetiracetam, lamotrigine, and lacosamide, which are superior to phenytoin, carbamazepine, and valproic acid). Secondary prophylaxis may be considered for patients who experience seizures. Medical staff must be alert to the potential side effects of antiepileptic therapy, such as abnormal liver function, cognitive impairment, and ataxia.

Antitumor therapy can also cause symptoms. For example, patients may experience dizziness, headache, nausea, loss of appetite, and fatigue after radiotherapy, and the risk of infection can be high in bedridden patients. Symptomatic management including nutritional support, moderate exercise, monitoring of electrolyte balance, and infection prevention can be offered. The risk of Pneumocystis jiroveci pneumonia (PJP) may be increased in patients who have received glucocorticoid therapy for a few weeks. Trimethoprim-sulfamethoxazole (TMP-SMX) prophylaxis should be considered if additional systemic immunosuppressive therapy is administered. Thromboprophylaxis should be considered for patients who are hospitalized or bedridden with acute illness. Low-molecular-weight heparin or unfractionated heparin is recommended for the primary prevention and treatment of venous thromboembolic events. Risk factors for thromboembolic events in patients with BM include specific primary tumors, glucocorticoid use, chemotherapy, high body mass index, and prolonged bed rest or immobilization. The risk of intracranial hemorrhage may not be increased in patients treated with low-molecular-weight heparin, and other risk factors for bleeding should be considered. Data on direct oral anticoagulants in patients with BM are lacking (101-104).

Prognosis

The prognosis of BCBM is closely related to the molecular subtype of the primary cancer. In a study of 1,147 patients with invasive breast cancer, OS was significantly shorter in patients with triple-negative (TN) breast cancer than in those with HER2-enriched tumors (P<0.001). The median duration of survival following brain metastasis (SFBM) was 386, 310, and 147 days in patients with luminal, HER2-enriched and TNBC, respectively (P=0.029). Patients with luminal breast cancer had a lower risk of developing BMs and the longest BMFS, whereas those with HER2-positive or TNBC had a significantly higher risk of developing BMs. Compared with that in the TNBC group, the duration of SFBM was doubled in the HER2-enriched group (105). Another study that included 206,913 breast cancer patients in the Surveillance, Epidemiology, and End Results (SEER) 18 registry showed that the median survival of patients with luminal A, luminal B, HER2, and TNBC and BM was 12, 23, 10, and 6 months, respectively (P<0.001), and in patients with BM without visceral metastasis it was 14, 34, 17, and 8 months, respectively (P<0.001). On multivariate analysis, among all patients with BM, the subtype order by favorable prognosis was luminal B, luminal A, HER2, and TNBC, while for those with BM without visceral metastasis, the order was luminal B, HER2, luminal A, and TNBC (106).

Previous studies have shown that the prognostic factors for patients with parenchymal BM from breast cancer include systemic organ and nervous system functional status (KPS score), age, primary tumor (location and extent, pathological type, and control), the number and location of BMs, surgical resection, extracranial metastases, recurrence, and the time from primary diagnosis to development of BM (107). Several prognostic indices have been developed accordingly. The value of the Breast GPA, which was proposed by Sperduto et al. on top of the GPA, has been well documented (108). In the original Breast GPA, the prognostic grade of BCBM was divided into three grades, and the prognostic factors included the KPS score, molecular subtype of breast cancer, and age (only for patients with a KPS score of 60–80). In 2015, researchers from the University of Texas MD Anderson Cancer Center modified the Breast GPA to develop a new index, the MDACC-GPA, in which a four-tiered grading system refined the prognostic grade of brain BCBM to grade 4. After the number of BMs was added as a prognostic factor, the concordance index was increased from 0.78 (95% CI, 0.77 to 0.80) to 0.84 (95% CI, 0.83 to 0.85) (Table 2) (109). In 2020, Sperduto et al. updated the Breast GPA, into which the extracranial metastasis and the time interval from primary diagnosis to development of BM were incorporated as prognostic factors, and the four-tier grading system was still used (Table 3) (110). Sperduto et al. have published their scoring tools on the following website: https://brainmetgpa.com/.

Table 2

MD Anderson Cancer Center Graded Prognostic Assessment scoring system

Prognostic factors 0 point 0.5 points 1.0 points 1.5 points
KPS score ≤50 60 70–80 90–100
Molecular subtype TNBC HR+ HER2+/HR– HER2+/HR–
Age >50 ≤50
No. of lesions >3 1–3

KPS, Karnofsky Performance Status; TNBC, triple-negative breast cancer; HR, hormone receptor; HER2, human epidermal growth factor receptor-2.

Table 3

Updated Breast-GPA scoring system

Prognostic factors 0 point 0.5 points 1.0 points 1.5 points
KPS score ≤60 70–80 90–100
Molecular subtype TNBC Luminal A HER2+ and Luminal B
Age, years >60 ≤60
No. of lesions >1 1
Extracranial metastasis Yes No

GPA, Graded Prognostic Assessment; KPS, Karnofsky Performance Status; TNBC, triple-negative breast cancer; HER2, human epidermal growth factor receptor-2.

LM is an uncommon complication of breast cancer; however, it is highly fatal. It indicates that the disease is already in its advanced stage and the prognosis is extremely poor. Without proper treatment, the median survival is only 6 weeks to 2 months. After therapeutic interventions, the median survival may reach 3 to 6 months; only 15% of patients survive for more than 1 year. Death is often caused by progressive neurological disorders. The prognosis is related to disease grade, CSF protein level, molecular subtype of the primary cancer, age, tumor size, metastasis to other sites, and KPS score at the time of diagnosis (111).

Follow-up and monitoring

Although the incidence of BMs is rising annually, guidelines on breast cancer do not recommend routine screening for patients with BMs due to the lack of evidence of survival benefits. Therefore, most BMs are detected based on neurological symptoms, and further active measures are required. HER2 positivity is a widely recognized risk factor for BM. Nearly half of all patients with HER2-positive breast cancer eventually acquire BMs. BMs from HER2-positive breast cancer seem to be an ongoing event that can occur even years after diagnosis. Furthermore, about half of patients with HER2-positive BCBM die due to central nervous system progression even after treatment. Therefore, given the high incidence of BMs in HER2-positive breast cancer and the complexity of BM treatment, brain MRI should be performed timely in patients with this subtype who present with neurological symptoms, to achieve early diagnosis and early treatment, although routine screening with brain MRI for these patients is not recommended by the current guidelines.

It is generally believed that BCBM patients should be regularly followed up after diagnosis and treatment and receive a series of examinations including history-taking, physical examinations, testing of serum tumor markers, and medical imaging. Follow-up visits should be arranged at 1- to 2-month intervals after treatment, and the patient should seek medical treatment if an abnormality is detected. For stable craniocerebral metastases, the intervals between visits can be extended to 3 to 6 months (Table 4).

Table 4

Recommendations on the treatment of breast cancer brain metastasis in the CSCO BC Guidelines 2022

Stratification Level I recommendations Level II recommendations
For patients presenting with 1–3 BMs (I) Well-controlled extracranial disease and KPS ≥60 points: (i)
Surgical resection (1A); postoperative SRS to the resection cavity; (ii) Due to the lack of survival benefit data and the risk of neurocognitive impairment, WBRT is not routinely recommended after surgery or SRT. (II) Poorly controlled extracranial disease, with a low KPS score: consider WBRT or supportive care
(I) SRT may be considered for lesions sized ≤3–3.5 cm (1B); (II) SRT may be considered for inoperable lesions (1B); (III) Medical therapy may be considered for patients with HER2+ BC (2B)
For patients presenting with >3 BMs WBRT or SRT Medical therapy may be considered for patients with HER2+ BC (2B)
Meningeal metastasis Radiotherapy (1A) Intrathecal injection (2B)

CSCO BC, Chinese Society of Clinical Oncology Breast Cancer; BM, brain metastasis; KPS, Karnofsky Performance Status; WBRT, whole-brain radiotherapy; SRT, stereotactic radiotherapy; BC, breast cancer; HER2, human epidermal growth factor receptor-2.


Acknowledgments

Funding: None.


Footnote

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tbcr.amegroups.com/article/view/10.21037/tbcr-22-30/coif). ZJ serves as the Editor-in-Chief of Translational Breast Cancer Research from November 2019 to October 2024. JC, YL, CH serve as unpaid Editorial Board Members of Translational Breast Cancer Research from May 2021 to April 2023. The other authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Xia C, Dong X, Li H, et al. Cancer statistics in China and United States, 2022: profiles, trends, and determinants. Chin Med J (Engl) 2022;135:584-90. [Crossref] [PubMed]
  2. Cacho-Díaz B, Lorenzana-Mendoza NA, Chávez-Hernandez JD, et al. Clinical manifestations and location of brain metastases as prognostic markers. Curr Probl Cancer 2019;43:312-23. [Crossref] [PubMed]
  3. Gabos Z, Sinha R, Hanson J, et al. Prognostic significance of human epidermal growth factor receptor positivity for the development of brain metastasis after newly diagnosed breast cancer. J Clin Oncol 2006;24:5658-63. [Crossref] [PubMed]
  4. Martin AM, Cagney DN, Catalano PJ, et al. Brain Metastases in Newly Diagnosed Breast Cancer: A Population-Based Study. JAMA Oncol 2017;3:1069-77. [Crossref] [PubMed]
  5. Song Y, Barry WT, Seah DS, et al. Patterns of recurrence and metastasis in BRCA1/BRCA2-associated breast cancers. Cancer 2020;126:271-80. [Crossref] [PubMed]
  6. Achrol AS, Rennert RC, Anders C, et al. Brain metastases. Nat Rev Dis Primers 2019;5:5. [Crossref] [PubMed]
  7. Rostami R, Mittal S, Rostami P, et al. Brain metastasis in breast cancer: a comprehensive literature review. J Neurooncol 2016;127:407-14. [Crossref] [PubMed]
  8. Madhusoodanan S, Ting MB, Farah T, et al. Psychiatric aspects of brain tumors: A review. World J Psychiatry 2015;5:273-85. [Crossref] [PubMed]
  9. Madhusoodanan S, Danan D, Moise D. Psychiatric manifestations of brain tumors: diagnostic implications. Expert Rev Neurother 2007;7:343-9. [Crossref] [PubMed]
  10. Brain metastases and cognitive dysfunction: a review. China Medical Innovation 2021;18:5.
  11. Derks SHAE, van der Veldt AAM, Smits M. Brain metastases: the role of clinical imaging. Br J Radiol 2022;95:20210944. [Crossref] [PubMed]
  12. Franchet C, Djerroudi L, Maran-Gonzalez A, et al. 2021 update of the GEFPICS' recommendations for HER2 status assessment in invasive breast cancer in France. Ann Pathol 2021;41:507-20. [Crossref] [PubMed]
  13. Patchell RA, Tibbs PA, Walsh JW, et al. A randomized trial of surgery in the treatment of single metastases to the brain. N Engl J Med 1990;322:494-500. [Crossref] [PubMed]
  14. Pollock BE, Brown PD, Foote RL, et al. Properly selected patients with multiple brain metastases may benefit from aggressive treatment of their intracranial disease. J Neurooncol 2003;61:73-80. [Crossref] [PubMed]
  15. Kamp MA, Rapp M, Bühner J, et al. Early postoperative magnet resonance tomography after resection of cerebral metastases. Acta Neurochir (Wien) 2015;157:1573-80. [Crossref] [PubMed]
  16. Patel AJ, Suki D, Hatiboglu MA, et al. Impact of surgical methodology on the complication rate and functional outcome of patients with a single brain metastasis. J Neurosurg 2015;122:1132-43. [Crossref] [PubMed]
  17. Ahluwalia M, Barnett GH, Deng D, et al. Laser ablation after stereotactic radiosurgery: a multicenter prospective study in patients with metastatic brain tumors and radiation necrosis. J Neurosurg 2018;130:804-11. [Crossref] [PubMed]
  18. Srinivasan ES, Grabowski MM, Nahed BV, et al. Laser interstitial thermal therapy for brain metastases. Neurooncol Adv 2021;3:v16-25. [Crossref] [PubMed]
  19. Sujijantarat N, Hong CS, Owusu KA, et al. Laser interstitial thermal therapy (LITT) vs. bevacizumab for radiation necrosis in previously irradiated brain metastases. J Neurooncol 2020;148:641-9. [Crossref] [PubMed]
  20. Palmisciano P, Haider AS, Nwagwu CD, et al. Bevacizumab vs laser interstitial thermal therapy in cerebral radiation necrosis from brain metastases: a systematic review and meta-analysis. J Neurooncol 2021;154:13-23. [Crossref] [PubMed]
  21. Sankey EW, Grabowski MM, Srinivasan ES, et al. Time to Steroid Independence After Laser Interstitial Thermal Therapy vs Medical Management for Treatment of Biopsy-Proven Radiation Necrosis Secondary to Stereotactic Radiosurgery for Brain Metastasis. Neurosurgery 2022;90:684-90. [Crossref] [PubMed]
  22. Rennert RC, Khan U, Tatter SB, et al. Patterns of Clinical Use of Stereotactic Laser Ablation: Analysis of a Multicenter Prospective Registry. World Neurosurg 2018;116:e566-70. [Crossref] [PubMed]
  23. Aoyama H, Tago M, Shirato H, et al. Stereotactic Radiosurgery With or Without Whole-Brain Radiotherapy for Brain Metastases: Secondary Analysis of the JROSG 99-1 Randomized Clinical Trial. JAMA Oncol 2015;1:457-64. [Crossref] [PubMed]
  24. Ou D, Cao L, Xu C, et al. Upfront brain radiotherapy may improve survival for unfavorable prognostic breast cancer brain metastasis patients with Breast-GPA 0-2.0. Breast J 2019;25:1134-42. [Crossref] [PubMed]
  25. Bergen ES, Binter A, Starzer AM, 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. [Crossref] [PubMed]
  26. Kocher M, Soffietti R, Abacioglu U, 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. J Clin Oncol 2011;29:134-41. [Crossref] [PubMed]
  27. Patchell RA, Tibbs PA, Regine WF, et al. Postoperative radiotherapy in the treatment of single metastases to the brain: a randomized trial. JAMA 1998;280:1485-9. [Crossref] [PubMed]
  28. Vecht CJ, Haaxma-Reiche H, Noordijk EM, et al. Treatment of single brain metastasis: radiotherapy alone or combined with neurosurgery? Ann Neurol 1993;33:583-90. [Crossref] [PubMed]
  29. Mintz AH, Kestle J, Rathbone MP, et al. A randomized trial to assess the efficacy of surgery in addition to radiotherapy in patients with a single cerebral metastasis. Cancer 1996;78:1470-6. [Crossref] [PubMed]
  30. Brennan C, Yang TJ, Hilden P, et al. A phase 2 trial of stereotactic radiosurgery boost after surgical resection for brain metastases. Int J Radiat Oncol Biol Phys 2014;88:130-6. [Crossref] [PubMed]
  31. Karlovits BJ, Quigley MR, Karlovits SM, et al. Stereotactic radiosurgery boost to the resection bed for oligometastatic brain disease: challenging the tradition of adjuvant whole-brain radiotherapy. Neurosurg Focus 2009;27:E7. [Crossref] [PubMed]
  32. Hartford AC, Paravati AJ, Spire WJ, et al. Postoperative stereotactic radiosurgery without whole-brain radiation therapy for brain metastases: potential role of preoperative tumor size. Int J Radiat Oncol Biol Phys 2013;85:650-5. [Crossref] [PubMed]
  33. Minniti G, Esposito V, Clarke E, et al. Multidose stereotactic radiosurgery (9 Gy × 3) of the postoperative resection cavity for treatment of large brain metastases. Int J Radiat Oncol Biol Phys 2013;86:623-9. [Crossref] [PubMed]
  34. Mahajan A, Ahmed S, McAleer MF, et al. Post-operative stereotactic radiosurgery versus observation for completely resected brain metastases: a single-centre, randomised, controlled, phase 3 trial. Lancet Oncol 2017;18:1040-8. [Crossref] [PubMed]
  35. Brown PD, Ballman KV, Cerhan JH, et al. Postoperative stereotactic radiosurgery compared with whole brain radiotherapy for resected metastatic brain disease (NCCTG N107C/CEC·3): a multicentre, randomised, controlled, phase 3 trial. Lancet Oncol 2017;18:1049-60. [Crossref] [PubMed]
  36. Andrews DW, Scott CB, Sperduto PW, et al. Whole brain radiation therapy with or without stereotactic radiosurgery boost for patients with one to three brain metastases: phase III results of the RTOG 9508 randomised trial. Lancet 2004;363:1665-72. [Crossref] [PubMed]
  37. Brown PD, Jaeckle K, Ballman KV, et al. Effect of Radiosurgery Alone vs Radiosurgery With Whole Brain Radiation Therapy on Cognitive Function in Patients With 1 to 3 Brain Metastases: A Randomized Clinical Trial. JAMA 2016;316:401-9. [Crossref] [PubMed]
  38. Aoyama H, Shirato H, Tago M, et al. Stereotactic radiosurgery plus whole-brain radiation therapy vs stereotactic radiosurgery alone for treatment of brain metastases: a randomized controlled trial. JAMA 2006;295:2483-91. [Crossref] [PubMed]
  39. Chang EL, Wefel JS, Hess KR, et al. Neurocognition in patients with brain metastases treated with radiosurgery or radiosurgery plus whole-brain irradiation: a randomised controlled trial. Lancet Oncol 2009;10:1037-44. [Crossref] [PubMed]
  40. Kayama T, Sato S, Sakurada K, et al. Effects of Surgery With Salvage Stereotactic Radiosurgery Versus Surgery With Whole-Brain Radiation Therapy in Patients With One to Four Brain Metastases (JCOG0504): A Phase III, Noninferiority, Randomized Controlled Trial. J Clin Oncol 2018; Epub ahead of print. [Crossref] [PubMed]
  41. Hartgerink D, Bruynzeel A, Eekers D, et al. A Dutch phase III randomized multicenter trial: whole brain radiotherapy versus stereotactic radiotherapy for 4-10 brain metastases. Neurooncol Adv 2021;3:vdab021. [Crossref] [PubMed]
  42. Brown PD, Gondi V, Pugh S, et al. Hippocampal Avoidance During Whole-Brain Radiotherapy Plus Memantine for Patients With Brain Metastases: Phase III Trial NRG Oncology CC001. J Clin Oncol 2020;38:1019-29. [Crossref] [PubMed]
  43. Serizawa T, Yamamoto M, Higuchi Y, et al. Local tumor progression treated with Gamma Knife radiosurgery: differences between patients with 2-4 versus 5-10 brain metastases based on an update of a multi-institutional prospective observational study (JLGK0901). J Neurosurg 2019;132:1480-9. [Crossref] [PubMed]
  44. Keller A, Doré M, Cebula H, et al. Hypofractionated Stereotactic Radiation Therapy to the Resection Bed for Intracranial Metastases. Int J Radiat Oncol Biol Phys 2017;99:1179-89. [Crossref] [PubMed]
  45. Rogers S, Stauffer A, Lomax N, et al. Five fraction stereotactic radiotherapy after brain metastasectomy: a single-institution experience and literature review. J Neurooncol 2021;155:35-43. [Crossref] [PubMed]
  46. Cleary RK, Meshman J, Dewan M, et al. Postoperative Fractionated Stereotactic Radiosurgery to the Tumor Bed for Surgically Resected Brain Metastases. Cureus 2017;9:e1279. [Crossref] [PubMed]
  47. Shaw E, Scott C, Souhami L, et al. Single dose radiosurgical treatment of recurrent previously irradiated primary brain tumors and brain metastases: final report of RTOG protocol 90-05. Int J Radiat Oncol Biol Phys 2000;47:291-8. [Crossref] [PubMed]
  48. Vogelbaum MA, Angelov L, Lee SY, et al. Local control of brain metastases by stereotactic radiosurgery in relation to dose to the tumor margin. J Neurosurg 2006;104:907-12. [Crossref] [PubMed]
  49. Minniti G, Scaringi C, Paolini S, et al. Single-Fraction Versus Multifraction (3 × 9 Gy) Stereotactic Radiosurgery for Large (>2 cm) Brain Metastases: A Comparative Analysis of Local Control and Risk of Radiation-Induced Brain Necrosis. Int J Radiat Oncol Biol Phys 2016;95:1142-8. [Crossref] [PubMed]
  50. Ernst-Stecken A, Ganslandt O, Lambrecht U, et al. Phase II trial of hypofractionated stereotactic radiotherapy for brain metastases: results and toxicity. Radiother Oncol 2006;81:18-24. [Crossref] [PubMed]
  51. Remick JS, Kowalski E, Khairnar R, et al. A multi-center analysis of single-fraction versus hypofractionated stereotactic radiosurgery for the treatment of brain metastasis. Radiat Oncol 2020;15:128. [Crossref] [PubMed]
  52. Gattozzi DA, Alvarado A, Kitzerow C, et al. Very Large Metastases to the Brain: Retrospective Study on Outcomes of Surgical Management. World Neurosurg 2018;116:e874-81. [Crossref] [PubMed]
  53. Gullhaug A, Hjermstad MJ, Yri O, et al. Use of radiotherapy in breast cancer patients with brain metastases: a retrospective 11-year single center study. J Med Imaging Radiat Sci 2021;52:214-22. [Crossref] [PubMed]
  54. Nieder C, Yobuta R, Mannsåker B. Patterns of Treatment and Outcome in Patients With 20 or More Brain Metastases. In Vivo 2019;33:173-6. [Crossref] [PubMed]
  55. Gondi V, Pugh SL, Tome WA, et al. Preservation of memory with conformal avoidance of the hippocampal neural stem-cell compartment during whole-brain radiotherapy for brain metastases (RTOG 0933): a phase II multi-institutional trial. J Clin Oncol 2014;32:3810-6. [Crossref] [PubMed]
  56. Mulvenna P, Nankivell M, Barton R, et al. Dexamethasone and supportive care with or without whole brain radiotherapy in treating patients with non-small cell lung cancer with brain metastases unsuitable for resection or stereotactic radiotherapy (QUARTZ): results from a phase 3, non-inferiority, randomised trial. Lancet 2016;388:2004-14. [Crossref] [PubMed]
  57. Diserbo M, Agin A, Lamproglou I, et al. Blood-brain barrier permeability after gamma whole-body irradiation: an in vivo microdialysis study. Can J Physiol Pharmacol 2002;80:670-8. [Crossref] [PubMed]
  58. Stemmler HJ, Schmitt M, Willems A, et al. 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;18:23-8. [Crossref] [PubMed]
  59. Fauquette W, Amourette C, Dehouck MP, et al. Radiation-induced blood-brain barrier damages: an in vitro study. Brain Res 2012;1433:114-26. [Crossref] [PubMed]
  60. Yonemori K, Tsuta K, Ono M, et al. Disruption of the blood brain barrier by brain metastases of triple-negative and basal-type breast cancer but not HER2/neu-positive breast cancer. Cancer 2010;116:302-8. [Crossref] [PubMed]
  61. Lin NU, Carey LA, Liu MC, et al. Phase II trial of lapatinib for brain metastases in patients with human epidermal growth factor receptor 2-positive breast cancer. J Clin Oncol 2008;26:1993-9. [Crossref] [PubMed]
  62. Lin NU, Diéras V, Paul D, et al. Multicenter phase II study of lapatinib in patients with brain metastases from HER2-positive breast cancer. Clin Cancer Res 2009;15:1452-9. [Crossref] [PubMed]
  63. Murthy RK, Loi S, Okines A, et al. Tucatinib, Trastuzumab, and Capecitabine for HER2-Positive Metastatic Breast Cancer. N Engl J Med 2020;382:597-609. [Crossref] [PubMed]
  64. Lin NU, Borges V, Anders C, et al. Intracranial Efficacy and Survival With Tucatinib Plus Trastuzumab and Capecitabine for Previously Treated HER2-Positive Breast Cancer With Brain Metastases in the HER2CLIMB Trial. J Clin Oncol 2020;38:2610-9. [Crossref] [PubMed]
  65. Krop IE, Kim SB, Martin AG, et al. Trastuzumab emtansine versus treatment of physician's choice in patients with previously treated HER2-positive metastatic breast cancer (TH3RESA): final overall survival results from a randomised open-label phase 3 trial. Lancet Oncol 2017;18:743-54. [Crossref] [PubMed]
  66. Mills MN, Walker C, Thawani C, et al. Trastuzumab Emtansine (T-DM1) and stereotactic radiation in the management of HER2+ breast cancer brain metastases. BMC Cancer 2021;21:223. [Crossref] [PubMed]
  67. Lin NU, Pegram M, Sahebjam S, 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. J Clin Oncol 2021;39:2667-75. [Crossref] [PubMed]
  68. Brufsky AM, Mayer M, Rugo HS, et al. Central nervous system metastases in patients with HER2-positive metastatic breast cancer: incidence, treatment, and survival in patients from registHER. Clin Cancer Res 2011;17:4834-43. [Crossref] [PubMed]
  69. Bazan F, Dobi E, Royer B, et al. Systemic high-dose intravenous methotrexate in patients with central nervous system metastatic breast cancer. BMC Cancer 2019;19:1029. [Crossref] [PubMed]
  70. Siena S, Crinò L, Danova M, 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;21:655-61. [Crossref] [PubMed]
  71. Oberhoff C, Kieback DG, Würstlein R, et al. Topotecan chemotherapy in patients with breast cancer and brain metastases: results of a pilot study. Onkologie 2001;24:256-60. [PubMed]
  72. Viñolas N, Graus F, Mellado B, et al. Phase II trial of cisplatinum and etoposide in brain metastases of solid tumors. J Neurooncol 1997;35:145-8. [Crossref] [PubMed]
  73. Cocconi G, Lottici R, Bisagni G, et al. Combination therapy with platinum and etoposide of brain metastases from breast carcinoma. Cancer Invest 1990;8:327-34. [Crossref] [PubMed]
  74. Franciosi V, Cocconi G, Michiara 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;85:1599-605. [Crossref] [PubMed]
  75. Christodoulou C, Bafaloukos D, Linardou H, 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;71:61-5. [Crossref] [PubMed]
  76. Petrelli F, Ghidini M, Lonati V, et al. The efficacy of lapatinib and capecitabine in HER-2 positive breast cancer with brain metastases: A systematic review and pooled analysis. Eur J Cancer 2017;84:141-8. [Crossref] [PubMed]
  77. Bachelot T, Romieu G, Campone M, et al. Lapatinib plus capecitabine in patients with previously untreated brain metastases from HER2-positive metastatic breast cancer (LANDSCAPE): a single-group phase 2 study. Lancet Oncol 2013;14:64-71. [Crossref] [PubMed]
  78. Xu B, Yan M, Ma F, et al. Pyrotinib plus capecitabine versus lapatinib plus capecitabine for the treatment of HER2-positive metastatic breast cancer (PHOEBE): a multicentre, open-label, randomised, controlled, phase 3 trial. Lancet Oncol 2021;22:351-60. [Crossref] [PubMed]
  79. Yan M, Bian L, Hu X, et al. Pyrotinib plus capecitabine for human epidermal growth factor receptor 2-positive metastatic breast cancer after trastuzumab and taxanes (PHENIX): a randomized, double-blind, placebo-controlled phase 3 study. Transl Breast Cancer Res 2020;1:13. [Crossref]
  80. Yan M, Ouyang Q, Sun T, et al. Pyrotinib plus capecitabine for patients with human epidermal growth factor receptor 2-positive breast cancer and brain metastases (PERMEATE): a multicentre, single-arm, two-cohort, phase 2 trial. Lancet Oncol 2022;23:353-61. [Crossref] [PubMed]
  81. Saura C, Oliveira M, Feng YH, et al. Neratinib Plus Capecitabine Versus Lapatinib Plus Capecitabine in HER2-Positive Metastatic Breast Cancer Previously Treated With ≥ 2 HER2-Directed Regimens: Phase III NALA Trial. J Clin Oncol 2020;38:3138-49. [Crossref] [PubMed]
  82. Freedman RA, Gelman RS, Anders CK, et al. TBCRC 022: A Phase II Trial of Neratinib and Capecitabine for Patients With Human Epidermal Growth Factor Receptor 2-Positive Breast Cancer and Brain Metastases. J Clin Oncol 2019;37:1081-9. [Crossref] [PubMed]
  83. Moulder SL, Borges VF, Baetz T, et al. Phase I Study of ONT-380, a HER2 Inhibitor, in Patients with HER2+-Advanced Solid Tumors, with an Expansion Cohort in HER2+ Metastatic Breast Cancer (MBC). Clin Cancer Res 2017;23:3529-36. [Crossref] [PubMed]
  84. Swain SM, Baselga J, Miles D, 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;25:1116-21. [Crossref] [PubMed]
  85. Urruticoechea A, Rizwanullah M, Im SA, et al. Randomized Phase III Trial of Trastuzumab Plus Capecitabine With or Without Pertuzumab in Patients With Human Epidermal Growth Factor Receptor 2-Positive Metastatic Breast Cancer Who Experienced Disease Progression During or After Trastuzumab-Based Therapy. J Clin Oncol 2017;35:3030-8. [Crossref] [PubMed]
  86. Krop IE, Lin NU, Blackwell K, et al. Trastuzumab emtansine (T-DM1) versus lapatinib plus capecitabine in patients with HER2-positive metastatic breast cancer and central nervous system metastases: a retrospective, exploratory analysis in EMILIA. Ann Oncol 2015;26:113-9. [Crossref] [PubMed]
  87. Montemurro F, Delaloge S, Barrios CH, et al. Trastuzumab emtansine (T-DM1) in patients with HER2-positive metastatic breast cancer and brain metastases: exploratory final analysis of cohort 1 from KAMILLA, a single-arm phase IIIb clinical trial☆. Ann Oncol 2020;31:1350-8. [Crossref] [PubMed]
  88. Modi S, Saura C, Yamashita T, et al. Trastuzumab Deruxtecan in Previously Treated HER2-Positive Breast Cancer. N Engl J Med 2020;382:610-21. [Crossref] [PubMed]
  89. Cortés J, Kim SB, Chung WP, et al. Trastuzumab Deruxtecan versus Trastuzumab Emtansine for Breast Cancer. N Engl J Med 2022;386:1143-54. [Crossref] [PubMed]
  90. Tolaney SM, Sahebjam S, Le Rhun E, et al. A Phase II Study of Abemaciclib in Patients with Brain Metastases Secondary to Hormone Receptor-Positive Breast Cancer. Clin Cancer Res 2020;26:5310-9. [Crossref] [PubMed]
  91. Schmid P, Adams S, Rugo HS, et al. Atezolizumab and Nab-Paclitaxel in Advanced Triple-Negative Breast Cancer. N Engl J Med 2018;379:2108-21. [Crossref] [PubMed]
  92. Robson M, Im SA, Senkus E, et al. Olaparib for Metastatic Breast Cancer in Patients with a Germline BRCA Mutation. N Engl J Med 2017;377:523-33. [Crossref] [PubMed]
  93. Litton JK, Rugo HS, Ettl J, et al. Talazoparib in Patients with Advanced Breast Cancer and a Germline BRCA Mutation. N Engl J Med 2018;379:753-63. [Crossref] [PubMed]
  94. Robson ME, Tung N, Conte P, et al. OlympiAD final overall survival and tolerability results: Olaparib versus chemotherapy treatment of physician's choice in patients with a germline BRCA mutation and HER2-negative metastatic breast cancer. Ann Oncol 2019;30:558-66. [Crossref] [PubMed]
  95. Wang Y, Wang E, Pan L, et al. A new strategy of CyberKnife treatment system based radiosurgery followed by early use of adjuvant bevacizumab treatment for brain metastasis with extensive cerebral edema. J Neurooncol 2014;119:369-76. [Crossref] [PubMed]
  96. Lu YS, Chen TW, Lin CH, et al. Bevacizumab preconditioning followed by Etoposide and Cisplatin is highly effective in treating brain metastases of breast cancer progressing from whole-brain radiotherapy. Clin Cancer Res 2015;21:1851-8. [Crossref] [PubMed]
  97. Leone JP, Emblem KE, Weitz M, et al. Phase II trial of carboplatin and bevacizumab in patients with breast cancer brain metastases. Breast Cancer Res 2020;22:131. [Crossref] [PubMed]
  98. Zagouri F, Zoumpourlis P, Le Rhun E, et al. Intrathecal administration of anti-HER2 treatment for the treatment of meningeal carcinomatosis in breast cancer: A metanalysis with meta-regression. Cancer Treat Rev 2020;88:102046. [Crossref] [PubMed]
  99. Glantz MJ, Jaeckle KA, Chamberlain MC, et al. A randomized controlled trial comparing intrathecal sustained-release cytarabine (DepoCyt) to intrathecal methotrexate in patients with neoplastic meningitis from solid tumors. Clin Cancer Res 1999;5:3394-402. [PubMed]
  100. Grossman SA, Finkelstein DM, Ruckdeschel JC, et al. Randomized prospective comparison of intraventricular methotrexate and thiotepa in patients with previously untreated neoplastic meningitis. Eastern Cooperative Oncology Group. J Clin Oncol 1993;11:561-9. [Crossref] [PubMed]
  101. Le Rhun E, Guckenberger M, Smits M, et al. EANO-ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up of patients with brain metastasis from solid tumours. Ann Oncol 2021;32:1332-47. [Crossref] [PubMed]
  102. Pace A, Dirven L, Koekkoek JAF, et al. European Association for Neuro-Oncology (EANO) guidelines for palliative care in adults with glioma. Lancet Oncol 2017;18:e330-40. [Crossref] [PubMed]
  103. Levin VA, Bidaut L, Hou P, et al. Randomized double-blind placebo-controlled trial of bevacizumab therapy for radiation necrosis of the central nervous system. Int J Radiat Oncol Biol Phys 2011;79:1487-95. [Crossref] [PubMed]
  104. Le Rhun E, Weller M, Brandsma D, et al. EANO-ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up of patients with leptomeningeal metastasis from solid tumours. Ann Oncol 2017;28:iv84-99. [Crossref] [PubMed]
  105. Oehrlich NE, Spineli LM, Papendorf F, et al. Clinical outcome of brain metastases differs significantly among breast cancer subtypes. Oncol Lett 2017;14:194-200. [Crossref] [PubMed]
  106. Kim YJ, Kim JS, Kim IA. Molecular subtype predicts incidence and prognosis of brain metastasis from breast cancer in SEER database. J Cancer Res Clin Oncol 2018;144:1803-16. [Crossref] [PubMed]
  107. Laakmann E, Riecke K, Goy Y, et al. Comparison of nine prognostic scores in patients with brain metastases of breast cancer receiving radiotherapy of the brain. J Cancer Res Clin Oncol 2016;142:325-32. [Crossref] [PubMed]
  108. Kufel-Grabowska J, Niwińska A, Radecka BS, et al. The Usefulness of Prognostic Tools in Breast Cancer Patients with Brain Metastases. Cancers (Basel) 2022;14:1099. [Crossref] [PubMed]
  109. Subbiah IM, Lei X, Weinberg JS, et al. Validation and Development of a Modified Breast Graded Prognostic Assessment As a Tool for Survival in Patients With Breast Cancer and Brain Metastases. J Clin Oncol 2015;33:2239-45. [Crossref] [PubMed]
  110. Sperduto PW, Mesko S, Li J, et al. Beyond an Updated Graded Prognostic Assessment (Breast GPA): A Prognostic Index and Trends in Treatment and Survival in Breast Cancer Brain Metastases From 1985 to Today. Int J Radiat Oncol Biol Phys 2020;107:334-43. [Crossref] [PubMed]
  111. Franzoi MA, Hortobagyi GN. Leptomeningeal carcinomatosis in patients with breast cancer. Crit Rev Oncol Hematol 2019;135:85-94. [Crossref] [PubMed]

(English Language Editor: Hau Pak Yui Christopher)

doi: 10.21037/tbcr-22-30
Cite this article as: Wang T, Chen J, Yang J, Fu M, Hua W, Jia W, Liu Y, Wang B, Yan M, Zhou J, Hao C, Chen J, Ou D, Jiang T, Mao Y, Jiang Z; the CSCO expert panel of breast cancer. CSCO expert consensus on the diagnosis and treatment of breast cancer brain metastasis. Transl Breast Cancer Res 2022;3:22.

Download Citation