Skip to main content
Discover Oncology logoLink to Discover Oncology
. 2024 Dec 29;15:845. doi: 10.1007/s12672-024-01719-1

A network dynamic nomogram for predicting overall survival and cancer-specific survival in patients with breast cancer liver metastases: an analysis based on the SEER database

Mengxiang Tian 1,2, Kangtao Wang 1,2, Ming Li 1,
PMCID: PMC11683041  PMID: 39739079

Abstract

The liver stands out as one of the most frequent sites for distant metastasis in breast cancer cases. However, effective risk stratification tools for patients with breast cancer liver metastases (BCLM) are still lacking. We identified BCLM patients from the SEER database spanning from 2010 to 2016. After meticulously filtering out cases with incomplete data, a total of 3179 patients were enrolled and randomly divided into training and validation cohorts at a ratio of 2:1. Leveraging comprehensive patient data, we constructed a nomogram through rigorous evaluation of a Cox regression model. Validation of the nomogram was conducted using a range of statistical measures, including the concordance index (C-index), calibration curves, time-dependent receiver operating characteristic curves, and decision curve analysis (DCA). Both univariable and multivariable analyses revealed significant associations between OS and CSS in BCLM patients and 14 variables, including age, race, and tumor stage, among others. Utilizing these pertinent variables, we formulated nomograms for OS and CSS prediction. Subsequent validation involved rigorous assessment using time-dependent ROC curves, decision curve analysis, C-index evaluations, and calibration curves. Our web-based dynamic nomogram represents a valuable tool for efficiently analyzing the clinical profiles of BCLM patients, thereby aiding in informed clinical decision-making processes.

Supplementary Information

The online version contains supplementary material available at 10.1007/s12672-024-01719-1.

Keywords: Breast cancer liver metastases (BCLM), Web-based dynamic nomogram, Overall survival (OS), Cancer-specific survival (CSS), SEER database

Introduction

Cancer stands as one of the foremost medical challenges globally and ranks as the second leading cause of death in the United States. Among women, breast cancer comprises a significant portion, representing 31% of new cancer cases and ranking as the foremost cancer type, while also claiming the second spot in mortality at 15% [1]. Alarmingly, 90% of cancer-related deaths in breast cancer stem from metastases originating in the primary tumor. Reports indicate that the 5-year overall survival (OS) rate for patients without distant metastasis can exceed 80%, contrasting sharply with the meager 20% 5-year OS rate post-metastasis [2, 3]. This underscores the critical impact of distant metastasis on the prognosis of breast cancer patients, with approximately 10% experiencing distant metastasis during primary tumor diagnosis and treatment, with notable organs such as bone, lung, and liver being primary sites of metastasis [4].

Among these organs, the liver ranks as the third most common site of metastasis in breast cancer cases. Despite its lower incidence compared to bone and lung metastasis, the 5-year survival rate for patients with breast cancer liver metastasis (BCLM) plummets to a mere 8.5%, significantly lower than those with bone or lung metastases [5]. Current treatment modalities for BCLM encompass chemotherapy, endocrine therapy, immunotherapy, target therapies, surgery and radiotherapy [6]. However, the efficacy of these treatments for BCLM patients remains suboptimal due to factors such as chemotherapy and endocrine therapy resistance. Hence, the development of a precise BCLM risk assessment system holds paramount importance in guiding clinical decision-making processes.

Nomograms serve as invaluable tools in assessing tumor patient prognosis [7]. Overcoming the limitations of tumor lymph node metastasis (TNM) staging, nomograms provide a comprehensive numerical probability of individual prognosis by integrating diverse prognostic variables, aligning with the personalized medicine paradigm emphasized in contemporary healthcare [8]. By inputting variables, clinicians can swiftly and accurately calculate patient prognoses based on the cumulative score.

In this study, we have developed two efficient and precise web-based dynamic nomograms tailored for assessing the prognosis of BCLM patients. These nomograms respectively evaluate overall survival (OS) and cancer-specific survival (CSS) in BCLM patients, offering clinicians valuable insights to make informed clinical decisions tailored to individual patient profiles.

Methods

Patient selection, nomogram construction and validation

We utilized the SEER*Stat 8.3.8 program to retrieve and download patient information from the SEER 18 database. This publicly accessible database contains comprehensive cancer incidence data and detailed patient information across the United States.

Our study encompassed adult breast cancer patients diagnosed with liver metastases between 2010 and 2016. The collected patient data included race, age, marital status, detailed TNM stage, history of radical resection at the primary site, receipt of radiation and chemotherapy, ER, PR, and HER2 receptor status in postoperative pathology, as well as the presence of bone, brain, or lung metastases. Patients lacking complete pathological and survival information were excluded. Ultimately, 3197 patients were enrolled in the study, with random allocation into a training cohort (2131 cases) and a verification cohort (1066 cases) at a ratio of 2:1.

We employed univariable Cox proportional hazards regression analysis to identify independent prognostic factors, upon which prognostic factor models were constructed. Subsequently, based on the results of univariable analysis, we conducted multivariable Cox proportional risk regression analysis to formulate a nomogram incorporating significant variables from the training cohort. The nomogram was designed to analyze 1-year, 3-year, and 5-year overall survival (OS) and cancer-specific survival (CSS) rates. Calibration curves, C-index, and decision curve analyses (DCA) were utilized to assess the predictive performance of the nomograms. Furthermore, time-dependent ROC analysis was employed to evaluate the efficacy of the OS and CSS models developed in our study (Fig. 1).

Fig. 1.

Fig. 1

Flow charts of selected patients in this study. We included a total of 4974 BCLM patients from the SEER database and excluded 1777 patients according to the exclusion criteria described earlier. For the remaining 3197 patients who met the requirements, we randomly divided them into the training cohort and the verification cohort on a 2:1 basis. The training queue is used to construct the Norman diagram, and the effectiveness of the validation queue is used to evaluate the architectural model. We validated the constructed Norman plots using calibration curves, C-index decision curve analysis (DAC), and time-varying ROC methods

Statistical analysis

All of these statistical analyses were conducted using R version 4.2.0. Univariable and multivariable Cox regression analyses were employed to assess the independent factors influencing overall survival (OS) and cancer-specific survival (CSS) in BCLM patients. Statistically significant differences were determined with a threshold of P < 0.1 in the univariable analysis and P < 0.05 in the multivariable analysis.

Web-based dynamic nomogram publication

Following validation, the web-based dynamic nomograms for BCLM OS and CSS were published online via the R package “DynNom” and hosted on shinyapps.io (https://www.shinyapps.io/) [9], accessed on 9 February 2024.

Ethical consideration

This is an observational study, therefore it does not necessitate review by the Xiangya Hospital Ethics Committee.

Result

Basic patient information

According to the process outlined in Fig. 1 above, we excluded patients lacking detailed pathological diagnosis, immunohistochemical results, and survival information, resulting in a total of 3197 patients being included in our study. The basic information of all patients is presented in Table 1. In summary, 41% of the patients in the study were aged between 51 and 65, 99.6% were female, 53.6% were unmarried, and 73.9% were white. Regarding pathological information, 100% of patients were diagnosed with de novo stage IV, 34.4% had T4 tumors, and 78.7% had lymph node metastasis. Concerning treatment, while 74.1% of patients received chemotherapy, the proportion of patients who underwent surgery was significantly lower, at only 32.1%. Regarding organ metastasis, bone remained the most common site for breast cancer patients with liver metastasis, affecting 56.6% of patients, followed by the lung at 32.7%. The percentages of patients testing positive for ER, PR, and HER-2 were 65.3%, 50.4%, and 40.9%, respectively. The mean OS and CSS for patients included in the study were 32.9 months and 35.1 months, respectively.

Table 1.

Basic information for all patients included in this study

Variables Total Cohort (n = 3197) Training cohort (n = 2131) Validation cohort(n = 1066)
No % No % No %
Age
 ≤ 50 982 30.7 640 30 342 32.1
 51–65 1313 41 907 42.6 406 38.1
 > 65 902 28.3 584 27.4 318 29.8
Gender
 Female 3183 99.6 2122 99.6 1061 99.5
 Male 14 0.4 9 0.4 5 0.5
Marital status
 Married 1482 46.4 977 45.8 505 47.4
 Unmarried 1715 53.6 1154 54.2 561 52.6
Race
 White 2361 73.9 1574 73.9 787 73.8
 Black 570 17.8 379 17.8 191 17.9
 Other 266 8.3 178 8.3 88 8.3
Grade
 Grade I/II 1299 40.6 872 40.9 427 40
 Grade III 1898 59.4 1259 59.1 639 60
T stage
 T1 426 13.3 269 12.6 157 14.7
 T2 1094 34.2 721 33.8 373 35
 T3 577 18.1 408 19.2 169 15.9
 T4 1100 34.4 733 34.4 367 34.4
N stage
 N0 680 21.3 459 21.5 221 20.7
 N+ 2517 78.7 1672 78.5 845 79.3
Primary surgery
 No 2170 67.9 1437 67.4 733 68.8
 Yes 1027 32.1 694 32.6 333 31.2
Chemotherapy
 No 828 25.9 560 26.3 268 25.1
 Yes 2369 74.1 1571 73.7 798 74.9
Radiation
 No 2335 73 1553 72.9 782 73.4
 Yes 862 27 578 27.1 284 26.6
Bone metastatic
 No 1386 43.4 932 43.7 454 42.6
 Yes 1811 56.6 1199 56.3 612 57.4
Brain metastatic
 No 2951 92.3 1969 92.4 982 92.1
 Yes 246 7.7 162 7.6 84 7.9
Lung metastatic
 No 2153 67.3 1432 67.2 721 67.6
 Yes 1044 32.7 699 32.8 345 32.4
ER
 Negative 1108 34.7 721 33.8 387 36.3
 Positive 2089 65.3 1410 66.2 679 63.7
PR
 Negative 1586 49.6 1055 49.5 531 49.8
 Positive 1611 50.4 1076 50.5 535 50.2
HER2
 Negative 1891 59.1 1264 59.3 627 58.8
 Positive 1306 40.9 867 40.7 439 41.2
 OS (months) 32.9 33.7 31.4
 CSS (months) 35.1 35.9 33.7

ER estrogen receptor, PR progesterone receptor, HER2 human epidermal growth factor receptor 2

Selection of variables in our nomogram

We conducted univariable and multivariable Cox regression analyses for all the variables listed in Table 1, determining their respective weights in OS and CSS. Initially, we performed a univariable analysis of the 16 variables, revealing that 14 of them displayed statistical significance. Subsequently, a multivariable analysis was conducted on these 14 variables to identify the final variables associated with OS and CSS, respectively (refer to Tables 2 and 3). The findings indicated that, with the exception of patient gender and whether radiotherapy was administered, all other variables demonstrated significant correlations with the OS and CSS of BCLM patients, thus warranting inclusion in the subsequent construction of the nomogram.

Table 2.

Univariable and multivariable Cox regression analysis of OS in BCLM patients

Characteristics Univariable Analysis Multivariable Analysis
OR 95%CI p OR 95%CI p
Age < 0.001 < 0.001
 < 50 Reference 1 Reference 1
 51–65 1.273 1.106–1.466 0.001 1.173 1.016–1.354 0.03
 > 65 2.071 1.785–2.403 < 0.001 1.7 1.446–1.998 < 0.001
Gender 0.903 NA
 Female Reference 1
 Male 0.947 0.393–2.28 0.903
Marital status < 0.001 < 0.001
 Married Reference 1 Reference 1
 Unmarried 1.386 1.236–1.553  < 0.001 1.207 1.073–1.358 0.002
Race < 0.001 0.001
 White Reference 1 Reference 1
 Black 1.312 1.139–1.512 < 0.001 1.306 1.128–1.512 < 0.001
 Other 0.887 0.71–1.109 0.293 0.953 0.761–1.193 0.672
Grade < 0.001 < 0.001
 I/II Reference 1 Reference 1
 III 1.263 1.125–1.419 < 0.001 1.441 1.272–1.634 < 0.001
T stage < 0.001 0.019
 T1 Reference 1 Reference 1
 T2 1.005 0.832–1.215 0.955 1.087 0.898–1.317 0.391
 T3 1.076 0.873–1.325 0.494 1.172 0.946–1.452 0.147
 T4 1.375 1.142–1.657 0.001 1.305 1.074–1.586 0.007
N stage 0.023 0.001
 N0 Reference 1 Reference 1
 N+ 0.857 0.751–0.979 0.023 0.821 0.714–0.944 0.006
Primary surgery < 0.001 < 0.001
 Yes Reference 1 Reference 1
 No 1.495 1.322–1.691 < 0.001 1.417 1.246–1.611 < 0.001
Chemotherapy < 0.001 < 0.001
 Yes Reference 1 Reference 1
 No 1.816 1.612–2.047 < 0.001 1.57 1.372–1.796 < 0.001
Radiation 0.441 NA
 Yes Reference 1
 No 1.055 0.929–1.197 0.441
Bone metastatic < 0.001 < 0.001
 No Reference 1 Reference 1
 Yes 1.321 1.178–1.482 < 0.001 1.231 1.091–1.39 0.001
Brain metastatic < 0.001 < 0.001
 No Reference 1 Reference 1
 Yes 2.068 1.709–2.502 < 0.001 1.789 1.467–2.182 < 0.001
Lung metastatic < 0.001 < 0.001
 No Reference 1 Reference 1
 Yes 1.597 1.422–1.793 < 0.001 1.272 1.127–1.435 < 0.001
ER < 0.001 < 0.001
 Positive Reference 1 Reference 1
 Negative 1.43 1.273–1.607 < 0.001 1.457 1.243–1.707 < 0.001
PR < 0.001 < 0.001
 Positive Reference 1 Reference 1
 Negative 1.44 1.286–1.612 < 0.001 1.389 1.192–1.619 < 0.001
HER-2 < 0.001 < 0.001
 Positive Reference 1 Reference 1
 Negative 1.773 1.571–2 < 0.001 1.931 1.695–2.199 < 0.001

ER estrogen receptor, PR progesterone receptor, HER2 human epidermal growth factor receptor 2, OR odds ratio, CI confidence interval, NA not available

Table 3.

Univariable and multivariable Cox regression analysis of CSS in BCLM patients

Characteristics Univariable analysis Multivariable analysis
OR 95%CI p OR 95%CI p
Age < 0.001 < 0.001
 < 50 Reference 1 Reference 1
 51–65 1.261 1.08–1.473 0.003 1.16 0.991–1.359 0.065
 > 65 1.935 1.637–2.287 < 0.001 1.535 1.279–1.843 < 0.001
Gender 0.889 NA
 Female Reference 1
 Male 0.933 0.349–2.49 0.889
Marital status < 0.001 < 0.001
 Married Reference 1 Reference 1
 Unmarried 1.379 1.212–1.569 < 0.001 1.211 1.06–1.384 0.005
Race 0.001 0.018
 White Reference 1 Reference 1
 Black 1.274 1.087–1.493 0.003 1.236 1.049–1.457 0.011
 Other 0.808 0.626–1.044 0.103 0.883 0.682–1.144 0.346
Grade < 0.001 < 0.001
 I/II Reference 1 Reference 1
 III 1.347 1.18–1.538 < 0.001 1.553 1.347–1.791 < 0.001
T stage < 0.001 0.05
 T1 Reference 1 Reference 1
 T2 0.961 0.764–1.21 0.737 1.041 0.825–1.314 0.733
 T3 1.144 0.896–1.46 0.281 1.213 0.944–1.559 0.131
 T4 1.368 1.095–1.71 0.006 1.265 1.003–1.595 0.047
N stage 0.023 0.008
 N0 Reference 1 Reference 1
 N+ 0.837 0.717–0.976 0.023 0.803 0.683–0.943 0.008
Primary surgery < 0.001 < 0.001
 Yes Reference 1 Reference 1
 No 1.573 1.336–1.768 < 0.001 1.466 1.266–1.696 < 0.001
Chemotherapy < 0.001 < 0.001
 Yes Reference 1 Reference 1
 No 1.837 1.603–2.106 < 0.001 1.617 1.386–1.886 < 0.001
Radiation 0.397 NA
 Yes Reference 1
 No 1.063 0.923–1.225 0.397
Bone metastatic < 0.001 < 0.001
 No Reference 1 Reference 1
 Yes 1.429 1.253–1.628 < 0.001 1.326 1.155–1.523 < 0.001
Brain metastatic < 0.001 < 0.001
 No Reference 1 Reference 1
 Yes 2.099 1.691–2.606 < 0.001 1.679 1.341–2.102 < 0.001
Lung metastatic < 0.001 < 0.001
 No Reference 1 Reference 1
 Yes 1.666 1.463–1.897 < 0.001 1.328 1.16–1.519 < 0.001
ER < 0.001 < 0.001
 Positive Reference 1 Reference 1
 Negative 1.396 1.225–1.592 < 0.001 1.462 1.225–1.745 < 0.001
PR < 0.001 < 0.001
 Positive Reference 1 Reference 1
 Negative 1.453 1.279–1.651 < 0.001 1.438 1.212–1.707 < 0.001
HER-2 < 0.001 < 0.001
 Positive Reference 1 Reference 1
 Negative 1.868 1.631–2.14 < 0.001 2.097 1.81–2.43 < 0.001

ER estrogen receptor, PR progesterone receptor, HER2 human epidermal growth factor receptor 2, OR odds ratio, CI confidence interval, NA not available

Nomogram construction and validation

Following univariable and multivariable Cox regression analyses, we identified a total of 14 variables suitable for constructing the nomogram. These variables were found to significantly correlate with both OS and CSS. Among them are age, marital status, race, tumor grade, T stage, lymph node metastasis, surgery, chemotherapy, bone metastasis, lung metastasis, brain metastasis, ER status, PR status, and HER2 status. Utilizing these variables, we developed 1-year, 3-year, and 5-year nomograms for OS and CSS, respectively. These nomograms include individual scores and total scores, with the total score determined based on the individual scores computed using the nomogram. These scores enable the evaluation of patients’ 1-year, 3-year, and 5-year OS and CSS outcomes (refer to Figs. 2A and 3A).

Fig. 2.

Fig. 2

Nomogram and model assessment for predicting 1-, 3-, and 5-year overall survival (OS) in BCLM patients. A The OS nomogram for BCLM patients. B The calibration plots in the training cohort for 1-year, 3-year, and 5-year OS. AUC values of ROCs of the nomograms for 1-year, 3-year, and 5-year OS. C The ROC curves of training cohort. D The decision curve analyses (DCA) of the nomogram and other factors’ overall survival for the training cohort. E The calibration plots in the verification cohort for 1-year, 3-year, and 5-year OS. F The ROC curves of validation cohort. G The decision curve analyses (DCA) of the nomogram and other factors’ overall survival for the validation cohort. AUC Area under Curve, ROC Receiver Operating Characteristic

Fig. 3.

Fig. 3

Nomogram and model assessment for predicting 1-, 3-, and 5-year cancer-specific survival (CSS) in BCLM patients. A The CSS nomogram for BCLM patients. B The calibration plots in the training cohorts for 1-year, 3-year, and 5-year CSS. AUC values of ROCs of the nomograms for 1-year, 3-year, and 5-year CSS. C The ROC curves of training cohort. D The decision curve analyses (DCA) of the nomogram and other factors’ cancer-specific survival for the training cohort. E The calibration plots in the verification cohorts for 1-year, 3-year, and 5-year CSS. F The ROC curves of verification cohort. G The decision curve analyses (DCA) of the nomogram and other factors’ cancer-specific survival for the verification cohort. AUC Area under Curve, ROC Receiver Operating Characteristic

Following the construction of the nomogram, we assessed its accuracy using various methods, including the concordance index (C-index), calibration curves, time-dependent receiver operating characteristic [10] curves, and decision curve analysis (DCA). For OS, the C-index values were 0.709 (95% confidence interval (CI]) 0.693–0.725) in the training cohort and 0.726 (95% CI 0.705–0.747) in the verification cohort. For CSS, the C-index values were 0.710 (95% CI 0.693–0.728) in the training cohort and 0.721 (95% CI 0.697–0.745) in the verification cohort (refer to Table 4). The calibration curves of the nomogram (Figs. 2B, E, 3B, and E) demonstrate a high level of agreement between predicted and observed survival probabilities in both the training and verification cohorts for OS and CSS, respectively. Furthermore, the time-dependent area under the curve [11] values of OS and CSS predictions at 1, 3, and 5 years are all greater than 0.75 in both the training and verification cohorts (Figs. 2C, F, 3C, and F), indicating a favorable discriminatory ability of our developed nomogram.

Table 4.

The C-indices for predictions of OS and CSS

Group OS CSS
C-Index 95%CI C-Index 95%CI
Training group 0.709 0.693–0.725 0.710 0.693–0.728
Validation group 0.726 0.705–0.747 0.721 0.697–0.745

C-index index of concordance, CI confidence interval

DCA curves are now widely utilized to compare nomograms with other clinically relevant factors. Figures 2D, G, 3D, and G depict DCA curves for OS and CSS of the nomogram and other clinical factors in BCLM patients. Our findings reveal that, in comparison with other clinical factors, the DCA curve of our designed nomogram exhibits significant superiority, suggesting its enhanced suitability for clinical use in aiding physicians to make informed decisions.

Risk stratification and their subgroup survival analysis

We aggregated the scores for each variable of BCLM patients to derive the total score and subsequently determined the cutoff value using X-tile software (version 3.6.1; Yale University, New Haven, CT, USA) (refer to Fig. 4A and C). Based on the X-tile software analysis results, we categorized the OS and CSS of BCLM patients into three risk levels: high-risk, medium-risk, and low-risk. Specifically, for OS, the high-risk, medium-risk, and low-risk groups for BCLM patients correspond to scores > 450, 305–460, and < 305, respectively (see Fig. 4B). Similarly, for CSS, the high-risk, medium-risk, and low-risk categories for BCLM patients are defined as scores > 437, 289–437, and < 289, respectively (refer to Fig. 4D).

Fig. 4.

Fig. 4

Risk stratification by X-tile software, the subgroup and their survival analysis. A The cut-off values of BCLM patients for OS. The black dot means the OS distinction point. B Risk stratification of OS in BCLM patients. The blue color is the low-risk group, the gray color is the medium-risk group, and the purple color is the high-risk group. C The cut-off values of BCLM patients for CSS. The black dot means the CSS distinction point. D Risk stratification of CSS in BCLM patients. The blue color is the low-risk group, the gray color is the medium-risk group, and the purple color is the high-risk group. E The Kaplan–Meier survival curves of three risk subgroups of BCLM patients for OS. F The Kaplan–Meier survival curves of three risk subgroups of BCLM patients for CSS

Utilizing this risk stratification, we generated Kaplan–Meier survival curves for BCLM patients across different risk levels. The 5-year OS rates were 35.0%, 11.3%, and 3.02% for high-risk, medium-risk, and low-risk BCLM patients, respectively (see Fig. 4E). Likewise, the 5-year CSS rates were 39.9%, 14.1%, and 1.6% for high-risk, medium-risk, and low-risk BCLM patients, respectively (refer to Fig. 4F). Notably, the differences among the three risk subgroups for both OS and CSS were statistically significant.

Web-based dynamic nomogram publication

We have officially released our web-based nomograms for predicting OS in BCLM patients (Fig. 5A) and CSS (Fig. 5B). These nomograms are accessible at the following links: (https://osnomogram-xyhospital.shinyapps.io/BCLM-OSnomogram/) (accessed on 9 February 2024) and (https://cssnomogram-xyhospital.shinyapps.io/BCLM-CSSnomogram/) (accessed on 9 February 2024). You can utilize these platforms to perform data calculations and conduct risk analysis.

Fig. 5.

Fig. 5

The web-based dynamic nomogram. A Web-based dynamic nomogram of OS of BCLM patients (https://osnomogram-xyhospital.shinyapps.io/BCLM-OSnomogram/) (accessed on 9 February 2024). B Web-based dynamic nomogram of CSS of BCLM patients (https://cssnomogram-xyhospital.shinyapps.io/BCLM-CSSnomogram/) (accessed on 9 February 2024)

Discussion

Breast cancer holds the highest incidence and ranks as the second leading cause of mortality among female malignancies. It’s estimated that over 150,000 breast cancer survivors in the United States are living with metastatic disease, with distant metastasis being a primary cause of mortality [12, 13]. Liver metastasis stands as the third most prevalent site for solid tumor metastasis in breast cancer, with the survival rates of breast cancer liver metastasis (BCLM) patients notably lower compared to those with bone or lung metastasis [14]. Despite the existence of prognostic nomograms for bone and lung metastasis of breast cancer established by Lu et al. [15] and Wang et al. [16], respectively, there has been a notable absence of prognostic nomograms specifically tailored for BCLM patients. Hence, we have developed a dynamic web-based nomogram leveraging fundamental patient information, pathological classifications, and treatment protocols of BCLM patients. This tool facilitates rapid and accurate evaluation of both overall survival (OS) and cancer-specific survival (CSS) statuses across diverse patient profiles. Clinicians can seamlessly input patient-specific information into our nomogram to assess prognosis and guide clinical treatment decisions effectively.

The typical cascade of tumor metastasis involves tumor cells invading blood or lymphatic vessels, transitioning into circulating tumor cells (CTCs), adapting to the microenvironment as disseminated tumor cells (DTCs), and eventually evolving into metastatic initiation cells upon reaching the site of metastasis to establish metastatic tumors [17, 18]. However, the molecular mechanism underlying liver metastasis in breast cancer remains a relatively unexplored area. The influence of liver-specific sinus structures and the liver microenvironment on breast cancer cells remains ambiguous [5]. Contrary to common belief, our findings challenge the notion that patients with breast cancer liver metastasis (BCLM) and positive lymph node metastasis exhibit better overall survival (OS) and cancer-specific survival (CSS) compared to those without metastasis. Instead, our results suggest that breast cancer cells may have a predilection for escaping into blood vessels rather than lymphatics to form CTCs, ultimately leading to liver metastases. Consequently, future research on the mechanism of liver metastasis in breast cancer may benefit from focusing on the mechanism of blood-derived tumor cell dissemination.

Our study revealed a correlation between the racial category of BCLM patients and their overall survival (OS) and cancer-specific survival (CSS), with black women demonstrating a tendency toward poorer prognoses. This disparity may stem from socioeconomic disparities, as black women often face economic challenges leading to lower levels of healthcare access and disparities in receiving standard treatment protocols. Reports indicate notable differences in treatment patterns between black and white breast cancer patients [19, 20]. For instance, for stage I and II breast cancer, a higher percentage of white patients receive breast-conserving surgery compared to black women. Similarly, for stage III breast cancer, black women are less likely to receive radiation and chemotherapy alone, with a higher propensity for mastectomy compared to white women. Furthermore, black women with hormone receptor (ER, PR) positive breast cancer are less likely to undergo endocrine therapy compared to their white counterparts [21]. Hence, it is imperative, based on our findings and similar reports, to ensure equitable access to high-quality treatment and health screening services for breast cancer patients, especially those with BCLM, across all racial backgrounds. Specifically, for black women, initiatives should focus on ensuring appropriate patients receive standardized treatments such as hormone therapy or mastectomy [22, 23]. Additionally, efforts to provide comprehensive, equitable, and affordable health insurance coverage for individuals of all racial backgrounds are crucial [24].

Our study also revealed that the receipt of radiotherapy did not significantly impact the overall survival (OS) and cancer-specific survival (CSS) of BCLM patients. Several randomized clinical trials have indicated that older patients with hormone-receptor-positive breast cancer who undergo breast-conserving surgery may not experience survival disadvantages without radiation therapy [2527]. Therefore, drawing from these clinical trials and our own findings, we suggest that for BCLM patients who opt out of radiotherapy or have contraindications to it, omitting radiotherapy from the treatment regimen may not adversely affect distant recurrence or long-term overall survival. Furthermore, our findings suggest that surgical intervention is associated with improved survival outcomes in BCLM patients, aligning with conclusions drawn from multiple retrospective studies and randomized clinical trials [2831]. Possible explanations for this phenomenon include: the potential for enhanced effectiveness of systemic treatments by reducing tumor burden through primary tumor removal; removal of necrotic and non-vascularized tumor regions, potentially inaccessible to drugs, thereby enhancing chemotherapy efficacy; elimination of the primary tumor as a source of systemic seeding; and alteration of the immunosuppressive tumor microenvironment, potentially rendering systemic therapy more effective [32, 33]. To date, the relationship between surgical removal of the primary tumor and prognosis in stage IV breast cancer patients has remained inconclusive. Nonetheless, our study findings may provide support for the notion that primary tumor removal significantly enhances both OS and CSS in BCLM patients.

Limitations

Our study also carries several limitations that warrant consideration. Firstly, certain variables that could potentially impact the prognosis of BCLM patients, such as nutritional status and the Ki67 index, were not included in our analysis. Incorporating these variables could provide further insights into patient outcomes. Secondly, our study relied on a retrospective analysis of data from the SEER database, which inherently introduces selection bias. Future studies employing prospective designs could help mitigate this limitation. Lastly, our verification of the nomogram was conducted using cases from within the SEER database rather than external cases, which may affect the generalizability of our findings. Utilizing external cases for validation would enhance the accuracy and reliability of our nomogram.

Supplementary Information

Acknowledgements

No.

Abbreviations

BCLM

Breast cancer liver metastases

OS

Overall survival

CSS

Cancer-specific survival

DCA

Decision curve analysis

TNM

Tumor lymph node metastasis

ER

Estrogen receptor

PR

Progesterone receptor

HER2

Human epidermal growth factor receptor 2

OR

Odds ratio

CI

Confidence interval

CTC

Circulating tumor cells

DTC

Disseminated tumor cells

Author contributions

Conception and design: M.T., K.W. and M.L.; administrative support: M.T. and K.W.; provision of study materials or patients: M.T.; collection and assembly of data: M.T., K.W. and M.L.; data analysis and Interpretation: K.W., and M.L.; writing—original draft: M.T., K.W. and M.L.: writing—review and editing: all authors final approval of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by funds from the Natural Scientific Foundation of China (No. 30771122) to Ming Li.

Data availability

Data is provided within the manuscript or supplementary information files.

Declarations

Ethics approval and consent to participate

Not applicable.

Informed consent

Not applicable.

Competing interests

The authors declare no competing interests.

Footnotes

Publisher's Note

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

References

  • 1.Siegel RL, et al. Cancer statistics, 2023. CA Cancer J Clin. 2023;73(1):17–48. [DOI] [PubMed] [Google Scholar]
  • 2.Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumours. Nature. 2012;490(7418):61–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Allemani C, et al. Global surveillance of trends in cancer survival 2000–14 (CONCORD-3): analysis of individual records for 37 513 025 patients diagnosed with one of 18 cancers from 322 population-based registries in 71 countries. Lancet. 2018;391(10125):1023–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Ng CKY, et al. Genetic heterogeneity in therapy-naïve synchronous primary breast cancers and their metastases. Clin Cancer Res. 2017;23(15):4402–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Liang Y, et al. Metastatic heterogeneity of breast cancer: molecular mechanism and potential therapeutic targets. Semin Cancer Biol. 2020;60:14–27. [DOI] [PubMed] [Google Scholar]
  • 6.Diamond JR, Finlayson CA, Borges VF. Hepatic complications of breast cancer. Lancet Oncol. 2009;10(6):615–21. [DOI] [PubMed] [Google Scholar]
  • 7.Grimes DA. The nomogram epidemic: resurgence of a medical relic. Ann Intern Med. 2008;149(4):273–5. [DOI] [PubMed] [Google Scholar]
  • 8.Balachandran VP, et al. Nomograms in oncology: more than meets the eye. Lancet Oncol. 2015;16(4):e173–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Jalali A, et al. Visualising statistical models using dynamic nomograms. PLoS ONE. 2019;14(11): e0225253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Peery AF, et al. Burden and cost of gastrointestinal, liver, and pancreatic diseases in the United States: update 2018. Gastroenterology. 2019;156(1):254-272.e11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.King KR, et al. IRF3 and type I interferons fuel a fatal response to myocardial infarction. Nat Med. 2017;23(12):1481–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Gogate A, et al. Projecting the prevalence and costs of metastatic breast cancer from 2015 through 2030. JNCI Cancer Spectr. 2021;5(4):pkab063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Mariotto AB, et al. Estimation of the number of women living with metastatic breast cancer in the United States. Cancer Epidemiol Biomarkers Prev. 2017;26(6):809–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Miller KD, et al. Cancer treatment and survivorship statistics, 2022. CA Cancer J Clin. 2022;72(5):409–36. [DOI] [PubMed] [Google Scholar]
  • 15.Lyu X, Luo B. Prognostic factors and survival prediction in HER2-positive breast cancer with bone metastases: a retrospective cohort study. Cancer Med. 2021;10(22):8114–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Wang K, et al. Web-based dynamic nomograms for predicting overall survival and cancer-specific survival in breast cancer patients with lung metastases. J Pers Med. 2022;13(1):43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Mohme M, Riethdorf S, Pantel K. Circulating and disseminated tumour cells - mechanisms of immune surveillance and escape. Nat Rev Clin Oncol. 2017;14(3):155–67. [DOI] [PubMed] [Google Scholar]
  • 18.Pantel K, Alix-Panabières C. Crucial roles of circulating tumor cells in the metastatic cascade and tumor immune escape: biology and clinical translation. J Immunother Cancer. 2022;10(12): e005615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Ellis L, et al. Racial and ethnic disparities in cancer survival: the contribution of tumor, sociodemographic, institutional, and neighborhood characteristics. J Clin Oncol. 2018;36(1):25–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Allgood KL, et al. Validating self-reported mammography use in vulnerable communities: findings and recommendations. Cancer Epidemiol Biomarkers Prev. 2014;23(8):1649–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Lum SS, et al. Disruption of national cancer database data models in the first year of the COVID-19 pandemic. JAMA Surg. 2023;158(6):643–50. [DOI] [PubMed] [Google Scholar]
  • 22.Jemal A, et al. Factors that contributed to black-white disparities in survival among nonelderly women with breast cancer between 2004 and 2013. J Clin Oncol. 2018;36(1):14–24. [DOI] [PubMed] [Google Scholar]
  • 23.DeSantis CE, et al. Breast cancer statistics, 2015: convergence of incidence rates between black and white women. CA Cancer J Clin. 2016;66(1):31–42. [DOI] [PubMed] [Google Scholar]
  • 24.Sineshaw HM, et al. Factors that contribute to differences in survival of black vs white patients with colorectal cancer. Gastroenterology. 2018;154(4):906-915.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Kunkler IH, et al. Breast-conserving surgery with or without irradiation in women aged 65 years or older with early breast cancer (PRIME II): a randomised controlled trial. Lancet Oncol. 2015;16(3):266–73. [DOI] [PubMed] [Google Scholar]
  • 26.Kunkler IH, et al. Breast-conserving surgery with or without irradiation in early breast cancer. N Engl J Med. 2023;388(7):585–94. [DOI] [PubMed] [Google Scholar]
  • 27.Sidaway P. Radiotherapy can be omitted for older patients with low-risk disease. Nat Rev Clin Oncol. 2023;20(4):210. [DOI] [PubMed] [Google Scholar]
  • 28.Soran A, et al. Randomized trial comparing resection of primary tumor with no surgery in stage IV breast cancer at presentation: protocol MF07-01. Ann Surg Oncol. 2018;25(11):3141–9. [DOI] [PubMed] [Google Scholar]
  • 29.Soran A, et al. Primary surgery with systemic therapy in patients with de novo stage IV breast cancer: 10-year follow-up; protocol MF07-01 randomized clinical trial. J Am Coll Surg. 2021;233(6):742-751.e5. [DOI] [PubMed] [Google Scholar]
  • 30.Arciero C, et al. Surgery and survival in patients with stage IV breast cancer. Breast J. 2019;25(4):644–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Zheng A, et al. Clinical information and management status of de novo stage IV breast cancer patients: a Chinese multicenter investigation (CSBrS-002). Chin Med J (Engl). 2021;134(13):1569–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Israel I, Margenthaler JA. Surgical extirpation of the primary tumor in stage IV breast cancer: the debate continues. J Am Coll Surg. 2021;233(6):751–2. [DOI] [PubMed] [Google Scholar]
  • 33.Ruiterkamp J, Ernst MF. The role of surgery in metastatic breast cancer. Eur J Cancer. 2011;47(Suppl 3):S6-22. [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

Data is provided within the manuscript or supplementary information files.


Articles from Discover Oncology are provided here courtesy of Springer

RESOURCES