Skip to main content
Springer logoLink to Springer
. 2024 Apr 29;206(3):527–541. doi: 10.1007/s10549-024-07321-x

Clinico–pathologic factors and survival of patients with breast cancer diagnosed with de novo brain metastasis: a national cancer database analysis

Ali Hijazi 1,, Mohamed Mohanna 1, Saad Sabbagh 1, María Herrán 1, Barbara Dominguez 1, Kaylee Sarna 2, Zeina Nahleh 1
PMCID: PMC11208224  PMID: 38683296

Abstract

Purpose

Patients with Breast Cancer (BC) with Brain Metastasis (BCBM) have poor survival outcomes. We aimed to explore the clinico–pathologic and therapeutic factors predicting the survival in patients with de novo BCBM using the National Cancer Database (NCDB).

Patients and methods

The NCDB was queried for patients with BC between 2010 and 2020. Survival analysis with Kaplan–Meier curves and log rank tests were used to find median overall survival (OS) in months (95% CI) across the different variables. A multivariate cox regression model was computed to identify significant predictors of survival.

Results

Out of n = 2,610,598 patients, n = 9005 (0.34%) had de novo BCBM. A trend of decreasing OS was observed with increasing age, Charlson–Deyo score (CDS), and number of extracranial metastatic sites. The highest median OS was observed in the Triple Positive and the lowest OS in the Triple Negative subgroup. Based on treatment regimen, combination of systemic therapy and local therapy achieved the highest OS. A positive trend in OS was observed in the BC subgroup analysis with targeted therapy demonstrating a survival benefit when added to systemic therapy.

The multivariate cox regression model showed that age, race, ethnicity, insurance, median income, facility type, CDS, BC subtype, metastatic location sites, and treatment combinations received were significantly associated with risk of death. Receiving only local treatment for BM without systemic therapy more than doubled the risk of death compared to combining it with systemic therapy.

Conclusions

This analysis suggests that treatment of systemic disease is the major factor influencing survival in patients with BCBM. Moreover, targeted therapy with anti–HER2 increased survival when added to systemic therapy explaining the highest median OS noted in the Triple Positive subgroup.

Supplementary Information

The online version contains supplementary material available at 10.1007/s10549-024-07321-x.

Keywords: Breast cancer, Brain metastasis, Immunotherapy, Survival, Prognosis, NCDB

Introduction

Breast Cancer (BC) ranks as the most common malignancy among females worldwide with an annual incidence of 2.3 million cases [1, 2]. Specifically, BC with metastasis at diagnosis (de novo metastatic BC) comprises 3–6% of all BC patients and presents a major clinical challenge as these patients have limited–life expectancy [3], with an estimated five–year survival of metastatic BC in women residing in the US limited to 30% [4]. The most common sites of BC metastases include bone, liver, lung, and brain, of which the brain metastatic group has the worst survival outcomes [5]. BC is the second most common source of brain metastases (BM) after lung cancer [6]. The incidence of breast cancer brain metastases (BCBM) has increased steadily over the last several years owing to improved management of the primary disease [7]. Many studies have explored the factors that might predict survival in patients with BCBM, with many factors identified including age, race, marital status, histology, grade, tumor size, molecular subtype, patterns of metastasis, history of chemotherapy, radiotherapy, and surgery of primary cancer [8, 9]. Such studies have led to the development of prognostic scores that help in clinical decision making, such as the well–studied Graded Prognostic Assessment (GPA) scoring tool, which was developed to estimate survival in different BM patients based on the tumor of origin [10]. Some of the significant factors used in the score include age, Karnofsky Performance Status (KPS), extracranial metastases, and number of BM [2]. According to the National Comprehensive Cancer Network (NCCN) guidelines, treatment of BM includes surgery for relief of symptoms, whole brain radiotherapy (WBRT), stereotactic radiosurgery (SRS), and palliative care if applicable [11]. Additionally, BCBM require treatment based on the primary tumor characteristics including chemotherapy, hormonal, and anti–HER2 targeted therapy [12, 13]. There is a growing number of studies and clinical trials investigating newer targeted therapies for BCBM which span different classes such as EGFR receptor modulators, tyrosine kinase inhibitors, and CDK4/6 inhibitors to name a few [1422]. Despite our growing knowledge about BCBM and the many efforts to identify prognostic and therapeutic interventions, large population–based survival studies on de novo BCBM remain lacking. Therefore, we aim to retrospectively analyze the national cancer database (NCDB) to identify factors and therapeutic interventions predicting survival of patients presenting with BCBM (Figs. 1 and 2).

Fig. 1.

Fig. 1

Kaplan–Meier plots of overall survival for breast cancer patients with brain metastases stratified by a age, b facility type, c Charlson-Deyo score, d breast cancer subtype, e number of extracranial metastatic sites, and f location of extracranial metastatic sites

Fig. 2.

Fig. 2

Kaplan–Meier plots of overall survival for breast cancer patients with brain metastases stratified by a brain metastases treatment modality, b breast cancer treatment modality, and c combination of both breast cancer and brain metastases treatments

Materials and methods

Patient data

The NCDB was queried for patients with BC with available data on de novo BM between 2010 and 2020. A total of n = 2,610,598 records of patients with BC were identified, out of whom 9005 had de novo BM. Access to this registry was achieved based on a Participant User File award granted to the principal investigator (N.Z.). The NCDB is a clinical oncology database sourced from hospital registry data collected in more than 1500 Commission on Cancer–accredited facilities (amounting to about 70% of all cancer diagnoses in the country). These data are used to analyze and track patients with malignant neoplastic diseases, their treatments, and outcomes. Variables used from the dataset included facility and patient demographics, BC–specific variables, and treatment modalities. Several variables were computed that are relevant to prognosis in this patient population. Variables for the number and location of extracranial metastatic sites (EMS) were computed by combining five individual metastatic sites: bone, liver, lung, distant lymph nodes, and other sites. A variable on BM treatment modality was computed by combining three individual modalities: Surgery, WBRT, and SRS. A variable on BC treatment modality was computed by combing three individual modalities: chemotherapy, hormonal therapy, and immunotherapy (referring to anti–HER2 therapy and other targeted therapies). Last, a variable on treatment combination was computed by combining the treatment status for BM and BC.

Statistical analysis

Chi–square, fisher exact, independent t, and Mann Whitney U tests were performed to evaluate the association between each categorical variable and treatment combinations received. Kaplan–Meier analyses and log rank tests were performed on the whole dataset to compare median overall survival (OS) across age, facility type, Charlson–Deyo Score (CDS), BC subtype, number of EMS, location of EMS, BM treatment modality, BC treatment modality, and combination of both treatment modalities. Furthermore, the same analysis was conducted on the four BC subgroups to compare OS across the different treatment modalities. Finally Univariate and Multivariate Cox regression models were computed with backwards elimination of 0.1 for both to identify the significant predictors of survival in the patient cohort. The cutoff of statistical significance was set at p < 0.05. SAS version 9.4 and R 4.2.3 were used for data analysis.

Results

Baseline characteristics

Out of n = 2,610,598 patients identified with BC in the NCDB between 2010 and 2020, n = 9005 (0.34%) patients had de novo BM. Table 1 outlines the baseline characteristics of this cohort across the different treatment combinations received. Most patients with de novo BM were in the 61–70 age (30.3%) compared to the lowest proportion in the ≤ 50 age group (20.6%). Most patients were female (98.9%), of White race (76.6%), and non–Hispanic ethnicity (92.4%). Most patients were treated at either Comprehensive Community Cancer Programs (CCCP) (39.2%) or Academic/Research Programs (35.5%). In this database, most patients had insurance with only 7.5% of the cohort being un–insured. Most patients had a CDS of 0 (79.7%) with only 2.6% having a score of ≥ 3. There was a trend of increasing BCBM diagnosis during the 11–year span ranging from 7.7% in 2010 to 10.2% in 2020. Most BC cases had invasive ductal histology (64.9%), were poorly differentiated (43.4%), and ≥ 3 cm in size (62.8%). The BC subtype proportions in this cohort were as follows: 48% HR( +)/HER2( − ), 22.6% HR( − )/HER2( − ), 16.8% HR( +)/HER2( +), and 12.6% HR(−)/HER2( +). Based on the number of EMS, 15.7%, 31.3%, 27.9%, and 25.1% of the cohort had 0, 1, 2, and ≥ 3 EMS, respectively. 17.7% of the patients did not receive treatment for either BC or BM, 9.5% received treatment for BM only, 33.6% received treatment for BC only, and 39.1% received treatment for both BC and BM. All variables except sex, ethnicity, facility type, year of diagnosis, and lympho–vascular invasion were significantly associated with the treatment combination received (p < 0.005).

Table 1.

Baseline demographics and breast cancer-related variables with group comparisons across the different treatments received

Variable Categories Overall No treatment for both Treatment for BM only Treatment for BC only Treatment for both p-value
N (%) 9005 (100) 1594 (17.7%) 859 (9.5%) 3027 (33.6%) 3525 (39.1%)

Age (years)

N (%)

 ≤ 50 1854 (20.6) 187 (11.7) 123 (14.3) 691 (22.8) 853 (24.2)  < 0.001
51–60 2510 (27.9) 341 (21.4) 232 (27.0) 873 (28.8) 1064 (30.2)
61–70 2732 (30.3) 470 (29.5) 276 (32.1) 912 (30.1) 1074 (30.5)
 ≥ 70 1909 (21.2) 596 (37.4) 228 (26.5) 551 (18.2) 534 (15.1)

Sex

N (%)

Female 8904 (98.9) 1577 (98.9) 852 (99.2) 2993 (98.9) 3482 (98.8) 0.7821
Male 101 (1.1) 17 (1.1) 7 (0.8) 34 (1.1) 43 (1.2)

Race

N (%), n = 8919

Black 1671 (18.7) 329 (20.9) 187 (22.1) 522 (17.4) 633 (18.1) 0.0071
Other 415 (4.7) 69 (4.4) 32 (3.8) 154 (5.1) 160 (4.6)
White 6833 (76.6) 1177 (74.7) 628 (74.1) 2329 (77.5) 2699 (77.3)

Ethnicity

N (%), n = 8764

Hispanic 664 (7.6) 106 (6.9) 61 (7.3) 241 (8.2) 256 (7.5) 0.4304
Non-Hispanic 8100 (92.4) 1434 (93.1) 778 (92.7) 2707 (91.8) 3181 (92.5)
Insurance status N (%), n = 8768 Medicaid 1439 (16.4) 206 (13.4) 114 (13.7) 512 (17.4) 607 (17.6)  < 0.0001
Medicare 3332 (38.0) 800 (51.9) 400 (48.0) 1025 (34.9) 1107 (32.0)
Not insured 655 (7.5) 144 (9.3) 67 (8.0) 215 (7.3) 229 (6.6)
Private insurance/managed care 3342 (38.1) 391 (25.4) 253 (30.3) 1183 (40.3) 1515 (43.8)
Facility type N (%), n = 8449 Academic/research program 3003 (35.5) 495 (32.1) 300 (35.8) 998 (35.8) 1210 (36.8) 0.0976
Community cancer program 742 (8.8) 149 (9.7) 79 (9.4) 253 (9.1) 261 (8.0)
Comprehensive community cancer program 3315 (39.2) 643 (41.7) 323 (38.6) 1085 (38.9) 1264 (38.5)
Integrated network cancer program 1389 (16.4) 254 (16.5) 135 (16.1) 451 (16.2) 549 (16.7)

Median income quartiles 2012–2016

N (%), n = 8092

 < $40,227 1758 (21.7) 337 (23.3) 179 (23.2) 581 (21.2) 661 (21.0) 0.0096
$4022–$50,353 1810 (22.4) 327 (22.6) 186 (24.1) 580 (21.3) 717 (22.8)
$50,354–$63,332 1901 (23.5) 354 (24.5) 168 (21.8) 609 (22.3) 770 (24.5)
 > $63,333 2623 (32.4) 428 (29.6) 238 (30.9) 960 (35.2) 997 (31.7)
Percent no high school degree quartiles 2012–2016 N (%), n = 8112  < 6.3% 1675 (20.6) 265 (18.3) 138 (17.8) 602 (22.0) 670 (21.2) 0.0025
6.3%–10.8% 2204 (27.2) 382 (26.4) 188 (24.3) 751 (27.5) 883 (28.0)
10.9%–17.5% 2189 (27.0) 402 (27.8) 224 (28.9) 730 (26.7) 833 (26.4)
 > 17.6% 2044 (25.2) 398 (27.5) 224 (28.9) 651 (23.8) 771 (24.4)

Year of diagnosis

N (%)

2010 696(7.7) 127(8.0) 72(8.4) 240(7.9) 257(7.3) 0.1797
2011 741(8.2) 135(8.5) 80(9.3) 238(7.9) 288(8.2)
2012 728(8.1) 132(8.3) 73(8.5) 261(8.6) 262(7.4)
2013 756(8.4) 117(7.3) 74(8.6) 271(9.0) 294(8.3)
2014 806(9.0) 123(7.7) 71(8.3) 293(9.7) 319(9.1)
2015 851(9.5) 150(9.4) 77(9.0) 308(10.2) 316(9.0)
2016 845(9.4) 163(10.2) 60(7.0) 269(8.9) 353(10.0)
2017 824(9.2) 156(9.8) 71(8.3) 282(9.3) 315(8.9)
2018 916(10.1) 171(10.7) 88(10.2) 289(9.5) 368(10.4)
2019 923(10.2) 157(9.9) 96(11.2) 285(9.4) 385(10.9)
2020 919(10.2) 163(10.2) 97(11.2) 291(9.6) 368(10.4)

Histology

N (%)

Ductal 5844 (64.9) 851 (53.4) 518 (60.3) 2044 (67.5) 2431 (69.0)  < 0.001
Lobular 590 (6.6) 90 (5.6) 39 (4.5) 241 (8.0) 220 (6.2)
Other 2571 (28.6) 653 (41.0) 302 (35.2) 742 (24.5) 874 (24.8)

Grade

N (%), n = 6085

1 916(15.1) 131(14.8) 61(11.8) 383(17.4) 341(13.7)  < 0.001
2 2525(41.5) 348(39.4) 186(36.1) 924(42.0) 1067(43.0)
3 2644(43.4) 405(45.8) 269(52.1) 894(40.6) 1076(43.3)
Tumor size N (%), n = 6561  < 1 cm 538 (8.2) 74 (7.2) 53 (8.8) 175 (7.8) 236 (8.8) 0.0002
1–2 cm 909 (13.9) 145 (14.0) 105 (17.4) 279 (12.3) 380 (14.2)
2–3 cm 989 (15.1) 148 (14.3) 116 (19.3) 360 (16.0) 365 (13.7)
 > 3 cm 4125 (62.8) 666 (64.5) 328 (54.5) 1443 (63.9) 1688 (63.2)

Lympho-vascular invasion

N (%), n = 2711

0 1742(64.3) 237(63.5) 134(61.2) 655(66.0) 716(63.5) 0.4583
1 969(35.7) 136(36.5) 85(38.8) 337(34.0) 411(36.5)

Charlson Deyo score

N (%)

0 7178 (79.7) 1196 (75.0) 637 (74.2) 2477 (81.8) 2868 (81.4)  < 0.001
1 1222 (13.6) 232 (14.6) 138 (16.1) 401 (13.2) 451 (12.8)
2 370 (4.1) 91 (5.7) 54 (6.3) 90 (3.0) 135 (3.8)
 ≥ 3 235 (2.6) 75 (4.7) 30 (3.5) 59 (1.9) 71 (2.0)

Breast cancer subtype

N (%), n = 7563

HR ( − )/HER2 ( − ) 1708 (22.6) 319 (30.0) 285 (42.3) 422 (15.7) 682 (21.7)  < 0.001
HR ( − )/HER2 ( +) 956 (12.6) 122 (11.5) 91 (13.5) 285 (10.6) 458 (14.6)
HR ( +)/HER2 ( − ) 3627 (48.0) 482 (45.3) 208 (30.9) 1514 (56.4) 1423 (45.3)
HR ( +)/HER2 ( +) 1272 (16.8) 140 (13.2) 90 (13.3) 464 (17.3) 578 (18.4)

Number of extracranial metastatic sites

N (%), n = 8979

Brain + 1 metastatic site 2808 (31.3) 476 (30.1) 241 (28.3) 1037 (34.3) 1054 (29.9)  < 0.001
Brain + 2 metastatic sites 2504 (27.9) 397 (25.1) 211 (24.8) 947 (31.3) 949 (27.0)
Brain +  ≥ 3 metastatic sites 2258 (25.1) 450 (28.5) 175 (20.5) 776 (25.7) 857 (24.3)
Only brain 1409 (15.7) 258 (16.3) 225 (26.4) 265 (8.7) 661 (18.8)

Location of extracranial metastatic sites

N (%), n = 8979

Bone 1786 (19.8) 283 (17.9) 109 (12.8) 799 (26.4) 595 (16.9)  < 0.001
Bone + liver 696 (7.8) 102 (6.4) 53 (6.2) 314 (10.4) 227 (6.5)
Bone + liver + lung 908 (10.1) 183 (11.6) 63 (7.4) 331 (10.9) 331 (9.4)
Bone + lung 1038 (11.6) 169 (10.7) 71 (8.3) 380 (12.6) 418 (11.9)
Liver 184 (2.1) 30 (1.9) 18 (2.1) 61 (2.0) 75 (2.1)
Liver + lung 264 (2.9) 50 (3.2) 36 (4.2) 75 (2.5) 103 (2.9)
Lung 689 (7.7) 141 (8.9) 91 (10.7) 137 (4.5) 320 (9.1)
Only brain 1409 (15.7) 258 (16.3) 225 (26.4) 265 (8.8) 661 (18.8)
Other 2005 (22.3) 365 (23.1) 186 (21.8) 663 (21.9) 791 (22.4)
Brain metastasis treatment modality N (%), n = 9004 No treatment 4620 (51.3) 1594 (100.0) 0 (0.0) 3026 (100.0) 0 (0.0)  < 0.001
SRS 827 (9.2) 0 (0.0) 122 (14.2) 0 (0.0) 705 (20.0)
WBRT 2596 (28.8) 0 (0.0) 505 (58.7) 0 (0.0) 2091 (59.3)
Surgery 445 (4.9) 0 (0.0) 150 (17.5) 0 (0.0) 295 (8.4)
Surgery + SRS 311 (3.5) 0 (0.0) 53 (6.2) 0 (0.0) 258 (7.3)
Surgery + WBRT 205 (2.3) 0 (0.0) 29 (3.4) 0 (0.0) 176 (5.0)
Breast cancer treatment modality N (%), n = 8991 No treatment 2439 (27.1) 1586 (100.0) 853 (100.0) 0 (0.0) 0 (0.0) < 0.001
Immunotherapy 98 (1.1) 0 (0.0) 0 (0.0) 49 (1.6) 49 (1.4)
Chemotherapy 2101 (23.4) 0 (0.0) 0 (0.0) 894 (29.5) 1207 (34.2)
Hormonal therapy 1434 (16.0) 0 (0.0) 0 (0.0) 753 (24.9) 681 (19.3)
Immunotherapy + hormonal therapy 199 (2.2) 0 (0.0) 0 (0.0) 112 (3.7) 87 (2.5)
Chemotherapy + hormonal therapy 1390 (15.5) 0 (0.0) 0 (0.0) 695 (23.0) 695 (19.7)
Chemotherapy + immunotherapy 912 (10.1) 0 (0.0) 0 (0.0) 340 (11.2) 572 (16.2)
Chemotherapy + hormonal therapy + immunotherapy 418 (4.6) 0 (0.0) 0 (0.0) 184 (6.1) 234 (6.6)

Median OS of the Cohort across different variables

The median OS of the 9005–patient cohort was 10.9 months (95% CI, 10.3–11.5). OS decreased significantly with increasing age, with highest OS observed in the ≤ 50 age group at 18.96 months (16.92–20.86) and the lowest in the ≥ 70 age group at 4.70 months (4.07–5.29) (log rank test, p < 0.0001). Patients treated at an Academic/Research Program had the highest OS amongst the different facilities at 13.63 months (12.19–15.00) (log rank test, p < 0.0001). OS decreased significantly with increasing CDS, with the highest OS in the group with a score of 0 at 12.42 months (11.76–13.17) and the lowest in the group with a score of ≥ 3 at 2.86 months (2.17–3.78) (log rank test, p < 0.0001). Across the four BC subgroups, the HR( +)/HER2( +) group had the highest OS at 22.05 months (18.73–24.67) compared to the lowest in the HR(−)/HER2(−) at 5.62 months (5.19–6.18) (log rank test, p < 0.0001). The HR( +)/HER2(–) and HR(–)/HER2( +) subgroups had similar OS at 15.80 months (14.46–17.15) and 14.59 months (11.79–16.95), respectively. There was a trend of worsening survival with increasing number of EMS, with the 1 EMS group having the highest OS at 13.17 months (12.02–14.36) compared to the group of with ≥ 3 EMS with lowest OS at 7.59 months (6.70–8.84) (log rank test, p < 0.0001). Based on the location of the EMS, bone metastasis conferred the highest OS amongst all combinations at 16.53 months (14.82–18.40) with the combined Liver and Lung group having the lowest OS at 5.22 months (3.09–6.34) (log rank test, p < 0.0001). Across the different local BM treatment modalities, patients without any treatment had the lowest OS at 9.26 months (8.31–10.02) compared to Surgery + WBRT group which had the highest OS at 32.33 months (23.98–40.44) (log rank test, p < 0.0001). Across the different BC treatment modalities, patients without any treatment had the lowest OS at 2.1 months (1.97–2.23) compared to the Chemotherapy + Hormonal Therapy + Immunotherapy group which had the highest OS at 42.35 months (35.48–54.14) (log rank test, p < 0.0001). Last, across the treatment combinations, the lowest OS was observed in the subgroup without any treatment at 1.77 months (1.64–1.97) followed by local treatment for BM only at 2.63 months (2.33–2.96). The subgroups that received BC treatment only and combination treatment for both brain and breast entities had similar OS at 16.92 months (16.00–18.27) and 16.30 months (15.11–17.38), respectively (log rank test, p < 0.0001). Table 2 summarizes the OS across the different variables, and Figs. 12 show the Kaplan–Meier curves with risk tables.

Table 2.

Median overall survival (OS) across age, facility type, Charlson-Deyo score, breast cancer subtype, number of extracranial metastatic sites, location of extracranial metastatic sites, brain metastasis treatment modality, breast cancer treatment modality, and treatment combinations

Age # of Cases Median OS 95% CI
  ≤ 50 years 1022/1501 18.96 16.92, 20.86
 51–60 years 1538/2054 12.98 11.89, 14.13
 61–70 years 1651/2183 10.55 9.50, 11.83
  ≥ 70 years 1266/1523 4.70 4.07, 5.29
 Total 5477/7261
 Log-rank test p-value  < .0001
Facility type
 Academic/research program 1755/2445 13.63 12.19, 15.00
 Community cancer program 472/611 9.89 7.75, 11.37
 Comprehensive community cancer program 2097/2650 9.26 8.38, 9.89
 Integrated network cancer program 864/1117 9.26 8.02, 10.55
 Total 5188/6823
 Log-rank test p-value  < .0001
Charlson Deyo score
 0 4251/5775 12.42 11.76, 13.17
 1 821/1014 8.08 6.77, 9.56
 2 249/291 4.90 3.38, 7.06
  ≥ 3 156/181 2.86 2.17, 3.78
 Total 5477/7261
 Log-rank test p-value  < .0001
Breast cancer subtype
 HR( − )/HER2( − ) 1194/1367 5.62 5.19, 6.18
 HR( − )/HER2( +) 547/767 14.59 11.79, 16.95
 HR( +)/HER2( − ) 2132/2936 15.80 14.46, 17.15
 HR( +)/HER2( +) 655/998 22.05 18.73, 24.67
 Total 4528/6068
 Log-rank test p-value  < .0001
Number of extracranial metastatic sites
 Only brain 868/1200 12.00 10.38, 13.83
 Brain + 1 metastatic site 1798/2374 13.17 12.02, 14.36
 Brain + 2 metastatic sites 1562/2053 10.48 9.53, 11.83
 Brain +  ≥ 3 metastatic sites 1232/1613 7.59 6.70, 8.84
 Total 5460/7240
 Log-rank test p-value  < .0001
Location of extracranial metastatic sites
 Only brain 868/1200 12.00 10.38, 13.83
 Bone 1127/1529 16.53 14.82, 18.40
 Liver 133/161 6.37 4.57, 13.80
 Lung 470/586 8.38 7.29, 9.63
 Bone + liver 467/595 10.12 7.79, 12.09
 Bone + lung 712/931 11.99 10.28, 13.73
 Liver + lung 200/232 5.22 3.09, 6.34
 Bone + liver + lung 671/805 6.83 5.49, 8.05
 Other 812/1201 9.56 8.08, 10.91
 Total 5460/7240
 Log-rank test p-value  < .0001
Brain metastasis treatment modality
 No treatment 2816/3729 9.26 8.31, 10.02
 WBRT 1725/2125 10.25 9.56, 11.10
 SRS 442/651 15.41 13.24, 18.53
 Surgery 246/358 19.81 14.78, 25.43
 Surgery + SRS 165/249 20.5 16.26, 23.98
 Surgery + WBRT 83/149 32.33 23.98, 40.44
 Total 5477/7261
 Log-rank test p-value  < .0001
Breast cancer treatment modality
 No treatment 1678/1955 2.10 1.97, 2.23
 Immunotherapy 60/74 5.00 3.32, 8.41
 Chemotherapy 1507/1817 10.50 9.86, 11.24
 Hormonal therapy 967/1225 13.54 12.02, 15.11
 Immunotherapy + hormonal therapy 103/154 23.69 17.74, 27.56
 Chemotherapy + hormonal therapy 680/1071 26.38 24.71, 28.55
 Chemotherapy + immunotherapy 345/647 27.56 24.77, 33.31
 Chemotherapy + hormonal therapy + immunotherapy 133/307 42.35 35.48, 54.14
 Total 5473/7250
 Log-rank test p-value  < .0001
Treatment combination
 No treatment for both 1082/1275 1.77 1.64, 1.97
 Treatment for brain metastasis only 600/691 2.63 2.33, 2.96
 Treatment for breast cancer only 1734/2454 16.92 16.00, 18.27
 Treatment for both 2061/2841 16.30 15.11, 17.38
 Total 5477/7261
 Log-rank test p-value  < .0001

Median OS by treatment modality across BC subtypes

Based on BM treatment modality, the Surgery + WBRT groups achieved the highest OS across three BC subgroups at 33.35 (24.48–40.87), 48.85 (10.41–), and 15.8 (6.31–21.98), in the HR( +)/HER2( − ), HR( − )/HER2( +), and HR( − )/HER2( − ) subgroups, respectively. For the HR( +)/HER2( +) subgroup, computing the Surgery + WBRT value was not possible, and Surgery + SRS achieve the highest OS at 42.25 (12.98–) (log rank test, p < 0.0001). Based on BC treatment modality, the Chemotherapy + Hormonal therapy + Immunotherapy groups achieved the highest OS across three BC subgroups at 55.13 (35.58–), 42.35 (36.04–55.36), and 31.34 (7.82–), for the HR( +)/HER2( − ), HR( +)/HER2( +), and HR( − )/HER2( +) subgroups, respectively. For the HR( − )/HER2( − ) subgroup, Chemotherapy + Immunotherapy achieved the highest OS at 11.7 (9.46–16.72) (log rank test, p < 0.0001). Based on treatment combinations, receiving local and systemic treatment combined for both BM and BC achieved the highest OS at 19.02 (17.08–20.70), 28.94 (24.77–35.29), 19.42 (16.95–23.36), and 8.84 (7.85–9.79) for the HR( +)/HER2( − ), HR( +)/HER2( +), HR( − )/HER2( +), and HR( − )/HER2( − ) subgroups, respectively (log rank test, p < 0.0001). Table 3 summarizes the OS across the four BC subgroups, and supplementary Figs. 3–6 show the Kaplan–Meier curves with risk tables.

Table 3.

Median overall survival (OS) for the four breast cancer subgroups across the different treatment modalities

BC subtype HR( +)/HER2( − ) HR( +)/HER2( +) HR( − )/HER2( +) HR( − )/HER2( − )
BM treatment modality # of Cases OS 95% CI # of Cases OS 95% CI # of Cases OS 95% CI # of Cases OS 95% CI
No treatment 1155/1615 14.88 13.17, 17.02 323/477 17.74 14.09, 22.34 237/329 11.56 7.85, 16.20 528/597 4.5 3.81, 5.03
WBRT 613/767 13.08 10.87, 14.39 240/331 18.83 14.23, 22.77 208/278 15.74 12.58, 18.43 452/498 6.08 5.36, 6.87
SRS 174/257 18.53 13.86, 22.77 48/97 39.79 28.75, 51/84 16.82 10.45, 30.65 107/134 8.5 5.49, 12.35
Surgery 109/159 25.43 19.35, 30.72 21/39 35.98 14.36, 53.26 25/34 10.02 4.80, 19.55 41/51 6.7 4.27, 11.37
Surgery + SRS 52/87 28.12 19.84, 44.09 17/33 42.25 12.98, 18/25 24.08 5.30, 59.17 43/53 7.46 4.44, 16.16
Surgery + WBRT 29/51 33.35 24.48, 40.87 6/21 29.11, 8/17 48.85 10.41, 23/34 15.8 6.31, 21.98
Total 2132/2936 655/998 547/767 1194/1367
Log-rank test p-value  < .0001  < .0001 0.0209  < 0.0001
BC treatment modality
 No treatment 463/565 2.69 2.43, 3.32 143/174 2.56 1.94, 3.02 130/152 1.77 1.45, 2.20 441/481 2.07 1.91, 2.30
 Immunotherapy (I) 4/6 18.34 1.87, 19/21 4.17 2.14, 14.75 26/32 3.94 2.90, 6.05 3/6 1.45,
 Chemotherapy (C) 340/433 12.48 10.87, 14.88 110/137 18.83 11.53,23.49 205/254 15.51 12.16, 17.77 683/792 8.51 7.66, 9.23
 Hormonal therapy (H) 746/957 15.38 13.54,18.20 63/70 5.6 3.22, 9.03 5/6 0.81 0.39, 11/12 3.33 0.90, 16.46
 I + H 46/73 31.54 24.51, 35.38 44/65 18 10.15, 23.36 3/4 7.36 1.35,
 C + H 481/785 26.91 25.36, 29.14 92/134 31.57 20.96, 40.31 13/13 14.16 6.01, 24.90 17/22 10.68 6.77, 27.24
 C + I 20/31 14.42 8.90, 27.07 104/209 36.37 27.50, 51.52 156/291 30.65 22.97, 35.45 39/53 11.7 9.46, 16.72
 C + H + I 31/83 55.13 35.58, 77/184 42.35 36.04, 55.36 14-Sep 31.34 7.82,
 Total 2131/2933 652/994 547/766 1194/1366
 Log-rank test p-value  < .0001  < .0001  < .0001  < .0001
Treatment combination
 Neither BM nor BC 321/398 2.56 2.10, 3.32 85/106 2.27 1.74, 2.70 77/90 1.51 1.15, 2.0 232/248 1.87 1.58, 2.07
 Only BM 143/170 3.25 2.50, 4.57 61/72 3.15 1.87, 5.26 53/63 2.07 1.64, 3.48 209/234 2.3 2.07, 2.63
 Only BC 834/1217 22.24 18.96, 24.77 238/371 24.67 21.19, 28.68 160/239 19.02 14.88, 23.16 296/349 8.57 7.33, 9.89
 BM + BC 834/1151 19.02 17.08, 20.70 271/449 28.94 24.77, 35.29 257/375 19.42 16.95, 23.36 457/536 8.84 7.85, 9.79
 Total 2132/2936 655/998 547/767 1194/1367
 Log-rank test p-value  < .0001  < .0001  < .0001  < .0001

Cox regression model

Univariate analyses were performed on 14 explanatory variables, and significant variables were computed to a multivariate cox regression model to find hazard ratios [HR (95% CI), p–value]. On multivariate analysis, older age was associated with increased risk of death. Compared to ≤ 50–year age–group, the 51–60 year and ≥ 70–year age groups had higher risk of death [1.17(1.04–1.31), p = 0.0099)] and [1.53(1.31–1.79), p < 0.0001], respectively. Patients with races other than White had lower risk of death compared to White patients [0.78(0.63–0.96), p = 0.0216]. Hispanic patients had lower risk of death compared to non–Hispanic patients [0.72(0.60–0.86), p = 0.0003]. Compared to patients with private insurance, those who were un–insured [1.38(1.18–1.61), p < 0.0001], on Medicaid [1.28(1.14–1.43), p < 0.0001], and on Medicare [1.20(1.07–1.34), p = 0.0013] had higher risks of death. Patients with a median income of < $40,227 had higher risk of death compared to ˃$63,333 [1.22(1.06–1.40), p = 0.0058], while high school degree was not significantly associated with survival. Compared to academic/research program facilities, CCCP [1.15(1.05–1.26), p = 0.0018], and integrated network cancer programs [1.21(1.08–1.36), p = 0.0012] had higher risks of death. Compared to patients with no comorbidities, higher CDS correlated with higher risks of death at [1.13(1.02–1.26), p = 0.0249], [1.32(1.09–1.60), p = 0.0041], and [1.74(1.39–2.18), p < 0.0001] for the 1, 2, and ≥ 3 score groups, respectively. Compared to patients diagnosed in 2018–2020, those diagnosed earlier in 2010–2011 [1.25(1.08–1.45), p = 0.0029] and 2014–2015 [1.20(1.03–1.39), p = 0.0164] had higher risk of death. The three BC subgroups had lower risk of death compared to the triple negative group, with the HR( +)/HER2( +) group having the best outcome with the lowest risk [0.43(0.38–0.49), p < 0.0001]. The location and number of EMS was significantly correlated with survival. Compared to only brain, bone + liver + lung [2.06(1.78–2.38), p < 0.0001] had the highest risk of death, followed by liver + lung [1.97(1.59–2.44), p < 0.0001], bone + liver [1.96(1.67–2.31), p < 0.0001], liver [1.88(1.45–2.45), p < 0.0001], other combinations [1.85(1.58–2.18), p < 0.0001], bone + lung [1.41(1.21–1.63), p < 0.0001], lung [1.31(1.12–1.53), p = 0.0009], and bone [1.31(1.15–1.49), p < 0.0001]. Compared to patients who received treatment for both breast and brain entities, patients who had no treatment for either [2.65(2.36–2.98), p < 0.0001] and treatment for BM only [2.30(2.00–2.63), p < 0.0001] were significantly more likely to die. Treatment for BC only was not statistically significant (p = 0.0920). Table 4 summarizes the results of the univariate and multivariate cox regression modelsa.

Table 4.

Univariate and multivariate cox regression models for variables predicting risk of death in the patient cohort

Cox regression model Univariate Multivariate
Variable HR (95% CI) p-value HR (95% CI) p-value
Age
  ≤ 50 years (ref) 1 1
 51–60 1.25(1.16–1.36)  < 0.0001 1.17(1.04–1.31) 0.0099
 61–70 1.36(1.26–1.47)  < 0.0001 1.11(0.98–1.27) 0.0935
  ≥ 70 2.05(1.88–2.22)  < 0.0001 1.53(1.31–1.79)  < 0.0001
Sex
 Female (ref.) 1
 Male 1.15(0.90–1.46) 0.2751
Race
 White (ref.) 1 1
 Black 1.09(1.02–1.16) 0.0167 1.04(0.94–1.15) 0.4849
 Other 0.76(0.66–0.88) 0.0002 0.78(0.63–0.96) 0.0216
Ethnicity
 Non-Hispanic (ref.) 1 1
 Hispanic 0.65(0.58–0.73)  < 0.0001 0.72(0.60–0.86) 0.0003
Insurance status
 Private insurance/managed care (ref.) 1 1
 Not insured 1.37(1.23–1.52)  < 0.0001 1.38(1.18–1.61)  < 0.0001
 Medicaid 1.17(1.08–1.27) 0.0002 1.28(1.14–1.43)  < 0.0001
 Medicare 1.57(1.48–1.68)  < 0.0001 1.20(1.07–1.34) 0.0013
Median income quartiles (2012–2016)
 > $63,333 (ref.) 1 1
 $50,354–$63,332 1.12(1.04–1.21) 0.0022 1.10(0.98–1.23) 0.1031
 $40,227–$50,353 1.16(1.07–1.25) 0.0002 1.11(0.98–1.26) 0.0924
 < $40,227 1.17(1.08–1.26)  < 0.0001 1.22(1.06–1.40) 0.0058
Percent no high school degree quartiles (2012–2016)
 < 6.3% (ref.) 1 1
 6.3%–10.8% 1.08(1.00–1.17) 0.053 1.02(0.90–1.14) 0.7999
 10.9%–17.5% 1.16(1.07–1.26) 0.0004 1.05(0.92–1.20) 0.4946
 > 17.6% 1.05(0.97–1.14) 0.2211 0.90(0.77–1.05) 0.1804
Facility type
 Academic/research program (ref.) 1 1
 Community cancer program 1.21(1.09–1.34) 0.0002 1.06(0.92–1.22) 0.3907
 Comprehensive community cancer program 1.24(1.16–1.32)  < 0.0001 1.15(1.05–1.26) 0.0018
 Integrated network cancer program 1.26(1.16–1.37)  < 0.0001 1.21(1.08–1.36) 0.0012
Charlson Deyo score
 0 (ref.) 1 1
 1 1.26(1.17–1.36)  < 0.0001 1.13(1.02–1.26) 0.0249
 2 1.65(1.45–1.88)  < 0.0001 1.32(1.09–1.60) 0.0041
  ≥ 3 1.92(1.63–2.25)  < 0.0001 1.74(1.39–2.18)  < 0.0001
Year of diagnosis
 2010 1.32(1.18–1.49)  < 0.0001
 2011 1.23(1.10–1.38) 0.0004
 2012 1.16(1.04–1.31) 0.0108
 2013 1.15(1.03–1.30) 0.0156
 2014 1.19(1.06–1.33) 0.0033
 2015 1.10(0.98–1.24) 0.0965
 2016 1.10(0.98–1.24) 0.1067
 2017 1.00(1.00–1.00)
 2018 1.13(1.01–1.27) 0.0385
 2019 1.00(1.00–1.00)
 2020 (ref.) 1
Year of diagnosis (regrouped)
 2010–2011 1.20(1.10–1.30)  < 0.0001 1.25(1.08–1.45) 0.0029
 2012–2013 1.09(1.00–1.18) 0.044 1.05(0.91–1.22) 0.4896
 2014–2015 1.08(0.99–1.16) 0.0774 1.20(1.03–1.39) 0.0164
 2016–2017 1.04(0.94–1.15) 0.5058 1.02(0.87–1.19) 0.8482
 2018–2020 (ref.) 1 1
Histology
 Ductal (ref.) 1
 Lobular 1.03(0.92–1.14) 0.6592
 Other 1.20(1.13–1.27)  < 0.0001
Grade
 1 (ref.) 1 1
 2 1.15(1.03–1.28) 0.0124 1.04(0.91–1.19) 0.547
 3 1.40(1.26–1.56)  < 0.0001 1.16(1.00–1.35) 0.0551
Tumor size
  > 3 cm(ref.) 1
 2–3 cm 1.02(0.93–1.11) 0.73
 1–2 cm 1.02(0.93–1.12) 0.6889
  < 1 cm 0.99(0.89–1.11) 0.8897
Lympho-vascular invasion
 0 (ref.) 1
 1 1.01(0.92–1.11) 0.8672
Breast cancer subtype
 HR( − )/HER2( − ) (ref.) 1 1 -
 HR( − )/HER2( +) 0.52(0.47–0.58)  < 0.0001 0.58(0.51–0.66)  < 0.0001
 HR( +)/HER2( − ) 0.51(0.48–0.55)  < 0.0001 0.54(0.49–0.60)  < 0.0001
 HR( +)/HER2( +) 0.41(0.37–0.45)  < 0.0001 0.43(0.38–0.49)  < 0.0001
Location of extracranial metastatic sites
 Only brain (ref.) 1 1
 Bone 0.96(0.88–1.05) 0.3799 1.31(1.15–1.49)  < 0.0001
 Bone + liver 1.22(1.09–1.37) 0.0005 1.96(1.67–2.31)  < 0.0001
 Bone + liver + lung 1.44(1.30–1.60)  < 0.0001 2.06(1.78–2.38)  < 0.0001
 Bone + lung 1.07(0.97–1.19) 0.1576 1.41(1.21–1.63)  < 0.0001
 Liver 1.27(1.06–1.52) 0.0107 1.88(1.45–2.45)  < 0.0001
 Liver + lung 1.73(1.48–2.02)  < 0.0001 1.97(1.59–2.44)  < 0.0001
 Lung 1.34(1.20–1.50)  < 0.0001 1.31(1.12–1.53) 0.0009
 Other 1.22(1.11–1.35)  < 0.0001 1.85(1.58–2.18)  < 0.0001
Treatment combination
 Treatment for both (ref.) 1 1
 Treatment for breast cancer only 0.99(0.93–1.06) 0.7528 0.93(0.85–1.01) 0.092
 Treatment for brain metastasis only 2.42(2.21–2.65)  < 0.0001 2.30(2.00–2.63)  < 0.0001
 No treatment for both 3.14(2.91–3.38)  < 0.0001 2.65(2.36–2.98)  < 0.0001

The model included fourteen explanatory variables (age, race, ethnicity, insurance status, median household income quartile 2012–2016, percent of no high school degree, Charlson Deyo Score, histology, grade, breast cancer subtype, metastasis location sites, treatment combinations, and year of diagnosis)

aUnivariate logistic regressions ran first. Sex, tumor size, and lympho-vascular invasion all not significant so not included in multivariate model. Histology (p = 0.1024) was eliminated by backward elimination. Model set at 0.1 cutoff

Discussion

In this analysis, we identified several factors contributing to prognosis of patients presenting with de novo BCBM including age, facility type, CDS, BC subtype, number and location of EMS, and local and systemic treatment modalities. Younger age, treatment at an academic/research program, lower CDS, triple positive BC status, having only one EMS, receiving surgery and WBRT, receiving Chemotherapy + Hormonal Therapy + Immunotherapy, and receiving combined BM and BC therapies were all associated with improved OS.

This data is consistent with another retrospective analysis including n = 1366 patients with de novo BCBM patients from the Surveillance, Epidemiology, and End Results (SEER) database between 2015 and 2019, by Yaning et al. finding median OS to be 12.0 months (10.4–13.6), which is very similar to our cohort’s value of 10.9 months [23]. Furthermore, the authors identified similar trends in subgroup survival, with the HR( +)/HER2( +) group having the best OS at 19.0 months (11.8–26.2) and the HR( − )/HER2( − ) having the worst OS at 7.0 months (5.4–8.6), both of which overlap with our results. Moreover, there was a similar trend in the OS of patients based on the metastatic sites with the bone only group having the longest OS (17.0 vs 16.5 months in our cohort) and all three sites (bone + liver + lung) having the lowest OS at 8.0 months (5.4–10.6) compared to 6.8 months in our cohort. Lastly, the OS decreased with increasing number of EMS like what was observed in our cohort. Similar trends were also observed in another study conducted on 248 patients with de novo BCBM between 2010 and 2018 from the SEER database [24]. In our analysis, OS decreased with increasing age, number of comorbidities, and number of EMS, which is in line with previously noted studies.

Overall, Surgery + WBRT yielded the best survival benefit amongst BM treatments, and these findings were also consolidated in the BC subgroup analysis. This is in line with the findings of the GPA study by Sperduto et al. which found that Surgery + WBRT treatment achieved the highest OS amongst all other combinations in BCBM patients at 25 months [2]. On the other hand, a recent systematic review on radiation therapy for BM identified five randomized trials conducted on post–surgical radiotherapy (SRS or WBRT) and found no differences in OS in the pooled results [25]. A growing number of clinical trials are ongoing to explore the best treatment modality for the local treatment of BCBM patients [6].

Overall, Chemotherapy + Hormonal Therapy + Immunotherapy yielded the best survival benefit amongst all BC treatments, findings also observed in the BC subgroup analysis. Of note, immunotherapy consistently improved survival across all the BC subtypes when added to systemic therapy. For example, in the HR( +)/HER2( − ) subgroup, adding targeted therapy more than doubled survival when added to the hormonal therapy alone group (from 15.38 to 31.54 months) and to the Chemotherapy + Hormonal therapy group (from 26.91 to 55.13 months). There is a growing number of studies and clinical trials that are investigating promising targeted and biologic therapies to target BCBM and shown survival benefits [12] which could explain the improved survival outcomes in our analysis with the addition of anti–HER2 therapy and other targeted therapies. Some of the drugs being explored include the anti HER2 targeting antibodies including: Trastuzumab [26, 27], Trastuzumab Emtansine [28, 29], Trastuzumab Deruxtecan [30], and Pertuzumab [31]; tyrosine kinase inhibitors including: Lapatinib [3235], Neratinib [3638], Afatinib [39], Tucatinib [40], Taselisib [41], Alpelisib [42], Buparlisib [43]; and CDK 4/6 inhibitors including: Palbociclib [44], Ribociclib [45], and Abemaciclib [46]; among other classes of targeted therapies. Unfortunately, the biologic agents used in treatment of the BCBM patient cohort are not available in the NCDB, but the trend of improved survival speaks to the rapid development of new targeted therapies that are currently under study. One example is the approval of Pembrolizumab for neoadjuvant and adjuvant treatment of patients with high–risk early–stage triple–negative BC in 2021 [47]. The study at hand is limited to 2020 and hence outcomes may improve even more for triple negative breast cancer in the coming years with more targeted therapies approved.

In the combined treatment analysis, receiving treatment for BM alone did not seem to prolong survival. Furthermore, treating BC alone achieved similar survival to treating both BC and BM. This suggests that the major therapeutic contributor to OS in de novo BCBM patients is the treatment of the underlying primary tumor rather than the BM itself. This finding is further supported by the findings of the multivariable cox regression model which integrates all the variables to identify and validate the individual survival benefits. In the model, treatment of BM alone increased the risk of death 2.3 folds compared to receiving dual treatment, which suggests that it is the BC treatment that confers any survival benefit.

Limitations

The study at hand has several limitations by virtue of it being conducted on a retrospective database which impedes control of certain variables. Furthermore, the NCDB does not provide information about relevant prognostic indicators identified in many studies such as number and size of BM, KPS, and the type of chemotherapy and targeted therapy received. Additionally, it was not possible to delineate the extent of BM surgery, and the radiation dose and number of treatment fractions to the BM in the analysis. Last, the NCDB provides information only about de novo BM and not recurrent BM. Recurrent BM constitutes a bigger percentage of BM and remains an important factor to consider when predicting prognosis. Despite these limitations, this remains, to the best of our knowledge, the biggest cohort of de novo BCBM patients to date and provides valuable information for clinical practice.

Conclusion

We retrospectively analyzed the biggest cohort of de novo BCBM patients exploring clinical and therapeutic factors associated with survival. Our results maintain the short survival of BCBM patients while also providing subgroup specific values that can guide clinical decision making. The BM–specific treatment that yielded the best survival outcomes was surgery combined with WBRT, and targeted therapy improved survival when added to systemic therapy across all subgroups. Further analysis showed that treating BM alone may decrease survival compared to receiving treatment for both BM and BC indicating that the primary disease is the main predictor of survival, and the BM management may serve a palliative role. Prospective studies are needed to consolidate these findings and to further highlight the role of targeted personalized therapy in improving survival of patients with BCBM.

Supplementary Information

Below is the link to the electronic supplementary material.

Author contributions

A.H., M.M., and Z.N. wrote the main manuscript text. S.S. and K.S. conducted the data management and statistical analyses. M.H. and B.D. prepared the tables and figures. All authors reviewed the manuscript.

Funding

The authors declare that no funds, grants, or other support were received during the preparation of this manuscript.

Data Availability

No datasets were generated or analysed during the current study.

Declarations

Competing interests

The authors have no relevant financial or non-financial interests to disclose.

Footnotes

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

References

  • 1.Łukasiewicz S, Czeczelewski M, Forma A, Baj J, Sitarz R, Stanisławek A. Breast cancer-epidemiology, risk factors, classification, prognostic markers, and current treatment strategies-an updated review. Cancers (Basel) 2021;13(17):4287. doi: 10.3390/cancers13174287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Sperduto PW, Mesko S, Li J, Cagney D, Aizer A, Lin NU, Nesbit E, Kruser TJ, Chan J, Braunstein S, et al. Survival in patients with brain metastases: summary report on the updated diagnosis-specific graded prognostic assessment and definition of the eligibility quotient. J Clin Oncol. 2020;38(32):3773–3784. doi: 10.1200/JCO.20.01255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Daily K, Douglas E, Romitti PA, Thomas A. Epidemiology of de novo metastatic breast cancer. Clin Breast Cancer. 2021;21(4):302–308. doi: 10.1016/j.clbc.2021.01.017. [DOI] [PubMed] [Google Scholar]
  • 4.Mariotto AB, Etzioni R, Hurlbert M, Penberthy L, Mayer M. Estimation of the number of women living with metastatic breast cancer in the United States. Cancer Epidemiol Biomarkers Prev. 2017;26(6):809–815. doi: 10.1158/1055-9965.EPI-16-0889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Conquer Cancer the ASCO Foundation (2022) Board CNE: breast cancer—metastatic: introduction.
  • 6.Watase C, Shiino S, Shimoi T, Noguchi E, Kaneda T, Yamamoto Y, Yonemori K, Takayama S, Suto A. Breast cancer brain metastasis-overview of disease state, treatment options and future perspectives. Cancers (Basel) 2021;13(5):1078. doi: 10.3390/cancers13051078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Kuksis M, Gao Y, Tran W, Hoey C, Kiss A, Komorowski AS, Dhaliwal AJ, Sahgal A, Das S, Chan KK, et al. The incidence of brain metastases among patients with metastatic breast cancer: a systematic review and meta-analysis. Neuro Oncol. 2021;23(6):894–904. doi: 10.1093/neuonc/noaa285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Li R, Zhang K, Siegal GP, Wei S. Clinicopathological factors associated with survival in patients with breast cancer brain metastasis. Hum Pathol. 2017;64:53–60. doi: 10.1016/j.humpath.2017.03.022. [DOI] [PubMed] [Google Scholar]
  • 9.Wang R, Zhu Y, Liu X, Liao X, He J, Niu L. The clinicopathological features and survival outcomes of patients with different metastatic sites in stage IV breast cancer. BMC Cancer. 2019;19(1):1091. doi: 10.1186/s12885-019-6311-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Sperduto PW, Kased N, Roberge D, Xu Z, Shanley R, Luo X, Sneed PK, Chao ST, Weil RJ, Suh J, et al. 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(4):419–425. doi: 10.1200/JCO.2011.38.0527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Horbinski C, Nabors LB, Portnow J, Baehring J, Bhatia A, Bloch O, Brem S, Butowski N, Cannon DM, Chao S. NCCN guidelines insights: central nervous system cancers, version 2.2022: featured updates to the NCCN guidelines. J Natl Compr Canc Netw. 2023;21(1):12–20. doi: 10.6004/jnccn.2023.0002. [DOI] [PubMed] [Google Scholar]
  • 12.Bailleux C, Eberst L, Bachelot T. Treatment strategies for breast cancer brain metastases. Br J Cancer. 2021;124(1):142–155. doi: 10.1038/s41416-020-01175-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Raghunath A, Desai K, Ahluwalia MS. Current treatment options for breast cancer brain metastases. Curr Treat Options Oncol. 2019;20(3):19. doi: 10.1007/s11864-019-0618-5. [DOI] [PubMed] [Google Scholar]
  • 14.Corti C, Antonarelli G, Criscitiello C, Lin NU, Carey LA, Cortés J, Poortmans P, Curigliano G. Targeting brain metastases in breast cancer. Cancer Treat Rev. 2022;103:102324. doi: 10.1016/j.ctrv.2021.102324. [DOI] [PubMed] [Google Scholar]
  • 15.Fares J, Kanojia D, Rashidi A, Ulasov I, Lesniak MS. Landscape of combination therapy trials in breast cancer brain metastasis. Int J Cancer. 2020;147(7):1939–1952. doi: 10.1002/ijc.32937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Nasrazadani A, Brufsky A. Neratinib: the emergence of a new player in the management of HER2+ breast cancer brain metastasis. Future Oncol. 2020;16(7):247–254. doi: 10.2217/fon-2019-0719. [DOI] [PubMed] [Google Scholar]
  • 17.Anwar M, Chen Q, Ouyang D, Wang S, Xie N, Ouyang Q, Fan P, Qian L, Chen G, Zhou E, et al. Pyrotinib treatment in patients with HER2-positive metastatic breast cancer and brain metastasis: exploratory final analysis of real-world. Multicenter Data Clin Cancer Res. 2021;27(16):4634–4641. doi: 10.1158/1078-0432.CCR-21-0474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Nader-Marta G, Martins-Branco D, Agostinetto E, Bruzzone M, Ceppi M, Danielli L, Lambertini M, Kotecki N, Awada A, de Azambuja E. Efficacy of tyrosine kinase inhibitors for the treatment of patients with HER2-positive breast cancer with brain metastases: a systematic review and meta-analysis. ESMO Open. 2022;7(3):100501. doi: 10.1016/j.esmoop.2022.100501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Sun H, Xu J, Dai S, Ma Y, Sun T. Breast cancer brain metastasis: current evidence and future directions. Cancer Med. 2023;12(2):1007–1024. doi: 10.1002/cam4.5021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Mills MN, King W, Soyano A, Pina Y, Czerniecki BJ, Forsyth PA, Soliman H, Han HS, Ahmed KA. Evolving management of HER2+ breast cancer brain metastases and leptomeningeal disease. J Neurooncol. 2022;157(2):249–269. doi: 10.1007/s11060-022-03977-x. [DOI] [PubMed] [Google Scholar]
  • 21.Ivanova M, Porta FM, Giugliano F, Frascarelli C, Sajjadi E, Venetis K, Cursano G, Mazzarol G, Guerini-Rocco E, Curigliano G, et al. Breast cancer with brain metastasis: molecular insights and clinical management. Genes (Basel) 2023;14(6):1160. doi: 10.3390/genes14061160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Epaillard N, Bassil J, Pistilli B. Current indications and future perspectives for antibody-drug conjugates in brain metastases of breast cancer. Cancer Treat Rev. 2023;119:102597. doi: 10.1016/j.ctrv.2023.102597. [DOI] [PubMed] [Google Scholar]
  • 23.He Y, Shao Y, Chen Q, Liu C, Zhu F, Liu H. Brain metastasis in de novo stage IV breast cancer. Breast. 2023;71:54–59. doi: 10.1016/j.breast.2023.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Sun MS, Yun YY, Liu HJ, Yu ZH, Yang F, Xu L. Brain metastasis in de novo breast cancer: an updated population-level study from SEER database. Asian J Surg. 2022;45(11):2259–2267. doi: 10.1016/j.asjsur.2021.12.037. [DOI] [PubMed] [Google Scholar]
  • 25.Garsa A, Jang JK, Baxi S, Chen C, Akinniranye O, Hall O, Larkin J, Motala A, Hempel S. Radiation therapy for brain metastases: a systematic review. Pract Radiat Oncol. 2021;11(5):354–365. doi: 10.1016/j.prro.2021.04.002. [DOI] [PubMed] [Google Scholar]
  • 26.Park YH, Park MJ, Ji SH, Yi SY, Lim DH, Nam DH, Lee JI, Park W, Choi DH, Huh SJ, et al. Trastuzumab treatment improves brain metastasis outcomes through control and durable prolongation of systemic extracranial disease in HER2-overexpressing breast cancer patients. Br J Cancer. 2009;100(6):894–900. doi: 10.1038/sj.bjc.6604941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Pestalozzi BC, Holmes E, de Azambuja E, Metzger-Filho O, Hogge L, Scullion M, Láng I, Wardley A, Lichinitser M, Sanchez RI, et al. 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(3):244–248. doi: 10.1016/S1470-2045(13)70017-2. [DOI] [PubMed] [Google Scholar]
  • 28.Krop IE, Lin NU, Blackwell K, Guardino E, Huober J, Lu M, Miles D, Samant M, Welslau M, Diéras V. Trastuzumab emtansine (T-DM1) versus lapatinib plus capecitabine in patients with HER2-positive metastatic breast cancer and central nervous system metastases: a retrospective, exploratory analysis in EMILIA. Ann Oncol. 2015;26(1):113–119. doi: 10.1093/annonc/mdu486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Montemurro F, Delaloge S, Barrios CH, Wuerstlein R, Anton A, Brain E, Hatschek T, Kelly CM, Peña-Murillo C, Yilmaz M, et al. Trastuzumab emtansine (T-DM1) in patients with HER2-positive metastatic breast cancer and brain metastases: exploratory final analysis of cohort 1 from KAMILLA, a single-arm phase IIIb clinical trial. Ann Oncol. 2020;31(10):1350–1358. doi: 10.1016/j.annonc.2020.06.020. [DOI] [PubMed] [Google Scholar]
  • 30.Modi S, Saura C, Yamashita T, Park YH, Kim SB, Tamura K, Andre F, Iwata H, Ito Y, Tsurutani J, et al. Trastuzumab deruxtecan in previously treated HER2-positive breast cancer. N Engl J Med. 2020;382(7):610–621. doi: 10.1056/NEJMoa1914510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Swain SM, Baselga J, Miles D, Im YH, Quah C, Lee LF, Cortés J. Incidence of central nervous system metastases in patients with HER2-positive metastatic breast cancer treated with pertuzumab, trastuzumab, and docetaxel: results from the randomized phase III study CLEOPATRA. Ann Oncol. 2014;25(6):1116–1121. doi: 10.1093/annonc/mdu133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Lin NU, Diéras V, Paul D, Lossignol D, Christodoulou C, Stemmler HJ, Roché H, Liu MC, Greil R, Ciruelos E, et al. Multicenter phase II study of lapatinib in patients with brain metastases from HER2-positive breast cancer. Clin Cancer Res. 2009;15(4):1452–1459. doi: 10.1158/1078-0432.CCR-08-1080. [DOI] [PubMed] [Google Scholar]
  • 33.Bachelot T, Romieu G, Campone M, Diéras V, Cropet C, Dalenc F, Jimenez M, Le Rhun E, Pierga JY, Gonçalves A, et al. Lapatinib plus capecitabine in patients with previously untreated brain metastases from HER2-positive metastatic breast cancer (LANDSCAPE): a single-group phase 2 study. Lancet Oncol. 2013;14(1):64–71. doi: 10.1016/S1470-2045(12)70432-1. [DOI] [PubMed] [Google Scholar]
  • 34.Geyer CE, Forster J, Lindquist D, Chan S, Romieu CG, Pienkowski T, Jagiello-Gruszfeld A, Crown J, Chan A, Kaufman B, et al. Lapatinib plus capecitabine for HER2-positive advanced breast cancer. N Engl J Med. 2006;355(26):2733–2743. doi: 10.1056/NEJMoa064320. [DOI] [PubMed] [Google Scholar]
  • 35.Lin NU, Freedman RA, Ramakrishna N, Younger J, Storniolo AM, Bellon JR, Come SE, Gelman RS, Harris GJ, Henderson MA, et al. A phase I study of lapatinib with whole brain radiotherapy in patients with Human Epidermal Growth Factor Receptor 2 (HER2)-positive breast cancer brain metastases. Breast Cancer Res Treat. 2013;142(2):405–414. doi: 10.1007/s10549-013-2754-0. [DOI] [PubMed] [Google Scholar]
  • 36.Awada A, Colomer R, Inoue K, Bondarenko I, Badwe RA, Demetriou G, Lee SC, Mehta AO, Kim SB, Bachelot T, et al. Neratinib plus paclitaxel vs trastuzumab plus paclitaxel in previously untreated metastatic ERBB2-positive breast cancer: the NEfERT-T randomized clinical trial. JAMA Oncol. 2016;2(12):1557–1564. doi: 10.1001/jamaoncol.2016.0237. [DOI] [PubMed] [Google Scholar]
  • 37.Freedman RA, Gelman RS, Anders CK, Melisko ME, Parsons HA, Cropp AM, Silvestri K, Cotter CM, Componeschi KP, Marte JM, 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(13):1081–1089. doi: 10.1200/JCO.18.01511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Saura C, Oliveira M, Feng YH, Dai MS, Chen SW, Hurvitz SA, Kim SB, Moy B, Delaloge S, Gradishar W, et al. Neratinib plus capecitabine versus lapatinib plus capecitabine in HER2-positive metastatic breast cancer previously treated with ≥ 2 HER2-directed regimens: phase III NALA trial. J Clin Oncol. 2020;38(27):3138–3149. doi: 10.1200/JCO.20.00147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Cortés J, Dieras V, Ro J, Barriere J, Bachelot T, Hurvitz S, Le Rhun E, Espié M, Kim SB, Schneeweiss A, et al. Afatinib alone or afatinib plus 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(16):1700–1710. doi: 10.1016/S1470-2045(15)00373-3. [DOI] [PubMed] [Google Scholar]
  • 40.Murthy RK, Loi S, Okines A, Paplomata E, Hamilton E, Hurvitz SA, Lin NU, Borges V, Abramson V, Anders C, et al. Tucatinib, trastuzumab, and capecitabine for HER2-positive metastatic breast cancer. N Engl J Med. 2020;382(7):597–609. doi: 10.1056/NEJMoa1914609. [DOI] [PubMed] [Google Scholar]
  • 41.Dent S, Cortés J, Im YH, Diéras V, Harbeck N, Krop IE, Wilson TR, Cui N, Schimmoller F, Hsu JY, et al. Phase III randomized study of taselisib or placebo with fulvestrant in estrogen receptor-positive, PIK3CA-mutant, HER2-negative, advanced breast cancer: the SANDPIPER trial. Ann Oncol. 2021;32(2):197–207. doi: 10.1016/j.annonc.2020.10.596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.André F, Ciruelos E, Rubovszky G, Campone M, Loibl S, Rugo HS, Iwata H, Conte P, Mayer IA, Kaufman B, et al. Alpelisib for PIK3CA-mutated, hormone receptor-positive advanced breast cancer. N Engl J Med. 2019;380(20):1929–1940. doi: 10.1056/NEJMoa1813904. [DOI] [PubMed] [Google Scholar]
  • 43.Di Leo A, Johnston S, Lee KS, Ciruelos E, Lønning PE, Janni W, O'Regan R, Mouret-Reynier MA, Kalev D, Egle D, et al. Buparlisib plus fulvestrant in postmenopausal women with hormone-receptor-positive, HER2-negative, advanced breast cancer progressing on or after mTOR inhibition (BELLE-3): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2018;19(1):87–100. doi: 10.1016/S1470-2045(17)30688-5. [DOI] [PubMed] [Google Scholar]
  • 44.Shah AN, Santa-Maria CA, Mukhija D, Shah N, Kang AK, Kumthekar P, Burdett K, Chandra S, Chang J, Tsarwhas D, et al. A phase II single-arm study of palbociclib in patients with HER2-positive breast cancer with brain metastases and analysis of ctDNA in patients with active brain metastases. Clin Breast Cancer. 2023;23(3):324–329. doi: 10.1016/j.clbc.2022.12.006. [DOI] [PubMed] [Google Scholar]
  • 45.Hortobagyi GN, Stemmer SM, Burris HA, Yap YS, Sonke GS, Paluch-Shimon S, Campone M, Blackwell KL, André F, Winer EP, et al. Ribociclib as first-line therapy for HR-positive, advanced breast cancer. N Engl J Med. 2016;375(18):1738–1748. doi: 10.1056/NEJMoa1609709. [DOI] [PubMed] [Google Scholar]
  • 46.Tolaney SM, Sahebjam S, Le Rhun E, Bachelot T, Kabos P, Awada A, Yardley D, Chan A, Conte P, Diéras V, et al. A phase II study of abemaciclib in patients with brain metastases secondary to hormone receptor-positive breast cancer. Clin Cancer Res. 2020;26(20):5310–5319. doi: 10.1158/1078-0432.CCR-20-1764. [DOI] [PubMed] [Google Scholar]
  • 47.Shah M, Osgood CL, Amatya AK, Fiero MH, Pierce WF, Nair A, Herz J, Robertson KJ, Mixter BD, Tang S, et al. FDA approval summary: pembrolizumab for neoadjuvant and adjuvant treatment of patients with high-risk early-stage triple-negative breast cancer. Clin Cancer Res. 2022;28(24):5249–5253. doi: 10.1158/1078-0432.CCR-22-1110. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Data Availability Statement

No datasets were generated or analysed during the current study.


Articles from Breast Cancer Research and Treatment are provided here courtesy of Springer

RESOURCES