Skip to main content
Erschienen in: BMC Cancer 1/2022

Open Access 01.12.2022 | Research

Predictive nomogram based on serum tumor markers and clinicopathological features for stratifying lymph node metastasis in breast cancer

verfasst von: Sheng-Kai Geng, Shao-Mei Fu, Hong-Wei Zhang, Yi-Peng Fu

Erschienen in: BMC Cancer | Ausgabe 1/2022

Abstract

Background

This study was aimed to establish the nomogram to predict patients’ axillary node status by using patients’ clinicopathological and tumor characteristic factors.

Methods

A total of 705 patients with breast cancer were enrolled in this study. All patients were randomly divided into a training group and a validation group. Univariate and multivariate ordered logistic regression were used to determine the predictive ability of each variable. A nomogram was performed based on the factors selected from logistic regression results. Receiver operating characteristic curve (ROC) analysis, calibration plots and decision curve analysis (DCA) were used to evaluate the discriminative ability and accuracy of the models.

Results

Logistic regression analysis demonstrated that CEA, CA125, CA153, tumor size, vascular-invasion, calcification, and tumor grade were independent prognostic factors for positive ALNs. Integrating all the predictive factors, a nomogram was successfully developed and validated. The C-indexes of the nomogram for prediction of no ALN metastasis, positive ALN, and four and more ALN metastasis were 0.826, 0.706, and 0.855 in training group and 0.836, 0.731, and 0.897 in validation group. Furthermore, calibration plots and DCA demonstrated a satisfactory performance of our nomogram.

Conclusion

We successfully construct and validate the nomogram to predict patients’ axillary node status by using patients’ clinicopathological and tumor characteristic factors.
Hinweise
Sheng-Kai Geng and Shao-Mei Fu contributed equally to this work.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
Her-2
Human epidermal growth factor receptor-2
TNBC
Triple negative breast cancer
ALND
Axillary lymph node dissection
SLNB
Sentinel lymph node biopsy
ER
Estrogen receptor
PR
Progesterone receptor
DCA
Decision curve analysis
ROC
Receiver operating characteristic curve
BCS
Breast conserving surgery
FNR
False negative rate

Introduction

The standard treatment for breast cancer patients has continually improved with the development of cancer research. Patients with clinically negative node status can avoid axillary lymph node dissection (ALND) if no metastasis is found in the sentinel lymph node (SLN) [1, 2]. In patients with a positive SLN, ALND has been the standard treatment until recent times. Efforts have been made to find if ALND can be avoided in patients with positive SLN to decrease the morbidity of complication including persistent lymphedema, paresthesia in the forearm and axilla and operated arm-weakness.
Approximately 50–70% of patients with positive SLN have no additional disease in the axillary lymph node, some studies [35] and nomograms [59] including MSKCC nomogram [10] have tried to identify the subgroup of breast cancer patients to avoid the ALND. This treatment option was challenged by the International Breast Cancer Study Group (IBCSG) trial 23 − 01 [11], which demonstrate that patients with micro-metastasis in sentinel lymph nodes (SLNs) can be spared from ALND. The Z0011 trial, enrolled with clinically node-negative patients with T1-T2 tumors and 1–2 positive sentinel lymph nodes, demonstrated that ALND did not lead to disease-free survival and overall survival benefit compared with SLNB only [12, 13]. Similarly, the AMAROS trial, the latest ten-year follow-up results also showed that for a comparable patient population, there was no significant difference in the DFS or OS between the ALND and axillary radiotherapy group [14]. Some researches tried to confirm the results in patients underwent mastectomy [15, 16]. In conclusion, accumulating studies have focused on whether we can spare patients from ALND based on the results of sentinel lymph node biopsy (SLNB). However, intraoperative SLNB will prolong the time of surgery and increase the probability of complications. Furthermore, the results of intraoperative SLNB were not as reliable as we thought, previous studies [1721] showed the false negative rate (FNR) was 14-43%. In addition, with the preoperative examination technique improving, the majority of breast cancer patients had a smaller tumor burden and lower incidence of axillary nodal metastasis at the time of diagnosis [22], reducing the benefits of ALNB. It is therefore reasonable to dig further into the question if we can find a way to predict the status of axillary lymph nodes for breast cancer patients to avoid SLNB.
Previous studies [2325] have demonstrated that estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) are predictive factor for nodal metastasis. L. Dihge’s research [26] and several other studies [2730] have tried to develop a nomogram for the prediction of axillary nodal status in breast cancer. However, the axillary lymph node status partially reflects the timeline of tumor development and is affected by the biological characteristic of the primary breast tumor, which can be manifested in terms of tumor marker, vascular-invasion, and calcification. Previous studies [3134] had revealed the diagnostic value of serum tumor markers including CEA, CA125, and CA15-3 in metastatic breast cancer. S. O’Grady et Al’s research also found that calcifications may be associated with tumor invasion into the lymphatic system [3537]. The present studies also revealed that vascular invasion was associated with higher lymph node metastasis possibility [38] and worse patients’ clinical outcome [39]. Therefore, the assessment of tumor characteristics including tumor marker, vascular-invasion, and calcification could be of great importance in revealing the extent of axillary lymph node (ALN) metastasis for patients with breast cancer, which most of studies did not mention. So clinically easily-accessible and reliable nomogram combing patients’ preoperative clinicopathological and tumor biological characteristic factors to predict the status of the axillary lymph node is urgently needed.
In our study, we aimed to establish and independently validate the nomogram to stratify patients’ axillary node status by using patients’ clinicopathological and tumor characteristic factors, in order to decide the best treatment option for patients. Firstly, we aim to predict the presence of lymph node metastasis to avoid unnecessary SLNB considering the postoperative complications. Furthermore, we sought the possibility to predict the stage of axillary lymph nodes metastasis to evaluate whether further ALND is needed in the case of positive SLNB.

Materials and methods

Patients

A total of 705 patients with breast cancer who underwent curative resection and axillary staging were enrolled in this study. After obtaining Ethics Committee’s approval, between January 2002 and December 2019, all patients were collected from the Zhongshan Hospital, Fudan University. Additionally, 705 patients were randomly divided into two groups, training group (509 patients) and validation group (196 patients). The inclusion and exclusion criteria for the patients are as follows: all patients were diagnosed pathologically with breast cancer; all patients enrolled in the study did not receive neoadjuvant chemotherapy; all patients underwent resection defined as a complete resection; all patients were treated with intraoperative SLNB (we use a single tracer, methylene blue, to locate sentinel lymph nodes intraoperatively); clinically N0 and some N1 stage patients enrolled in the training or validation groups were all treated with SLNB; all patients with one or more positive SLN underwent ALND; all the blood samples were obtained within 3 days before operations, all patients had complete records including baseline characteristics (including sex, age, menopause status, stage, molecular type, pre-operation routine blood test, tumor marker). In our study, ER/PR- was defined as less than 1% of breast cancer tissue immunohistochemical (IHC) nuclear staining; ER/PR low expression was defined as 1-10% IHC nuclear staining; PR high expression was defined as > 20% nuclear staining [34]; IHC Ki67 ≤ 14% was defined as low expression and Ki67 > 14% as high expression; Her-2-positive was defined as IHC 3 + or IHC 2 + with fluorescence in situ hybridization (FISH) +; Breast cancer was divided into 4 molecular subtypes : (1) the Luminal A subtype (ER+, PR high expression, HER-2-, Ki-67 low expression), (2) the Luminal B subtype (ER+, PR low expression, HER-2-, Ki-67 high expression, or ER+, PR any, HER-2+, Ki-67 any), (3) the HER-2 enriched subtype (ER-, PR-, Her-2+, Ki-67 any), and (4) the triple negative breast cancer (TNBC) subtype (ER-, PR-, Her-2-, Ki-67 any).

Preoperative evaluation

All patients underwent preoperative imaging examinations (including ultrasound and mammography) to evaluate breast cancer lesions’ calcification; Calcification positive was defined as the calcifications with malignant signs, such as fine sand-like, amorphous, or pleomorphic calcifications found in the same location as the cancer foci on ultrasound or mammography. Patients’ vascular invasion status was evaluated by preoperative tumor core needle biopsy (CNB), vacuum assisted breast biopsy (VABB), or excisional biopsy.

Statistical analysis

The association of clinicopathologic characteristics between testing cohorts and validation cohorts was analyzed by using the X² test or Fisher’s exact test or t-test as appropriate. Univariate ordered logistic regression was performed to determine the association of each variable with the possibility of lymph node metastasis. All variables with p ≤ 0.01 (two tailed) were considered statistically significant and included in the multivariate ordered logistic regression using backward stepwise method. A nomogram for predicting ALN metastasis or four or more ALN metastases was constructed with the remaining variables by means of the “rms” package of the R software in the training group, and the results were validated using patients in the validation group. We used the receiver operating characteristic curve (ROC) analysis and calibration plots to evaluate the discriminative ability and accuracy of the models, respectively. What’s more, decision curve analysis (DCA) was used to assess the actual benefits for clinical patients. All the statistical analyses were performed by using SPSS version 24 (SPSS Inc) and R version 4.1.

Results

Clinicopathological profiles of the patients

All patients (N = 705) were randomly divided into two groups, the training group (N = 509) and the validation group (N = 196). The clinicopathological characteristics of two groups were well balanced and presented in Table 1. The median age of all patients is 55 (26–90), training group 54 (26–90 range), and validation group 56 (27–90 range). There were no significant differences in menopausal status, the count of CEA and CA153, tumor size, histology, biological subtype, tumor grade, vascular invasion, calcification, and location between two groups. Among breast cancer patients in the training group, 187 (36.7%) experienced lymph node metastasis, 74 (14.5%) of whom with 4 or more ALN metastasis. While in the validation group, there were 81 (41.3%) patients experiencing ALN metastasis, 35 (17.8%) of whom with 4 or more ALN metastasis.
Table 1
Patient characteristics
Characteristics
Total (n = 705)
Training group (n = 509)
Validation group (n = 196)
P value
Age, years
 Median (range)
55 (26—90)
54 (26—90)
56 (27—90)
0.033
Menopausal status
 Premenopausal
375 (53.2%)
280 (55.0%)
95 (48.5%)
0.119
 Postmenopausal
330 (46.8%)
229 (45.0%)
101 (51.5%)
 
CEA (Ug/L)
 Average
2.773 ± 3.269
2.619 ± 2.444
3.171 ± 4.775
0.071
CA 125 (Ug/L)
 Average
15.012 ± 12.939
14.701 ± 14.021
15.818 ± 9.560
0.028
CA 153 (Ug/L)
 Average
11.415 ± 9.838
11.061 ± 8.538
12.335 ± 12.584
0.254
Tumor size
 Average
2.620 ± 1.476
2.636 ± 1.480
2.579 ± 1.469
0.478
T category
 T1
330 (46.8%)
228 (44.8%)
102 (52.0%)
0.22
 T2
337 (47.8%)
252 (49.5%)
85 (43.4%)
 
 T3
38 (5.4%)
29 (5.7%)
9 (4.6%)
 
Histology
 Invasive ductal carcinoma
601 (85.2%)
434 (85.3%)
167 (85.2%)
0.177
 Invasive lobular carcinoma
26 (3.7%)
18 (3.5%)
8 (4.1%)
 
 Mixed
30 (4.3%)
25 (4.9%)
3 (2.6%)
 
 Other
48 (6.8%)
32 (6.3%)
16 (8.2%)
 
Biological subtype
 Luminal A
98 (13.9%)
70 (13.8%)
28 (14.3%)
0.604
 Luminal B
449 (63.7%)
325 (63.9%)
124 (63.3%)
 
 Her-2 amplified
71 (10.1%)
55 (10.8%)
16 (8.2%)
 
 TNBC
87 (12.3%)
59 (11.6%)
28 (14.3%)
 
Tumor grade
 Well differentiated, grade 1
78 (11.1%)
51 (10.0%)
27 (13.8%)
0.341
 Moderately differentiated, grade 2
297 (42.7%)
215 (42.2%)
82 (41.8%)
 
 Poorly differentiated, grade 3
330 (46.8%)
243 (47.7%)
87 (44.4%)
 
Vascular invasion
 No
594 (84.3%)
428 (84.1%)
166 (84.7%)
0.843
 Yes
111 (15.7%)
81 (15.9%)
30 (15.3%)
 
Calcification
 No
331 (47.0%)
244 (47.9%)
87 (44.4%)
0.716
 Yes
374 (53.0%)
265 (52.1%)
109 (55.6%)
 
Lymph node metastasis
 No
437 (62.0%)
322 (63.3%)
115 (58.7%)
0.261
 Yes
268 (38.0%)
187 (36.7%)
81 (41.3%)
 
Pathologic N category
 N0
437 (62.0%)
322 (63.3%)
115 (58.7%)
0.328
 N1
159 (22.6%)
113 (22.2%)
46 (23.5%)
 
 N2
65 (9.2%)
41 (8.1%)
24 (12.2%)
 
 N3
44 (6.2%)
33 (6.5%)
11 (5.6%)
 
ER status
 No
160 (22.7%)
116 (22.8%)
44 (22.4%)
0.923
 Yes
545 (77.3%)
393 (77.2%)
152 (77.6%)
 
PR status
 No
261(37.0%)
192 (37.7%)
69 (35.2%)
0.535
 Yes
444(63.0%)
317 (62.3%)
127 (64.8%)
 
HER-2 status
 Negative
510 (72.3%)
361 (70.9%)
149 (76.0%)
0.175
 Overexpressed
195 (27.7%)
148 (29.1%)
47 (24.0%)
 
Location
 UOQ
422 (59.9%)
294(57.8%)
128(65.3%)
0.28
 LOQ
71 (10.1%)
55(10.8%)
16(8.2%)
 
 LIQ
76 (10.8%)
60(11.8%)
16(8.2%)
 
 UIQ
114 (16.2%)
82(16.1%)
32(16.3%)
 
 Central
22 (3.1%)
18(3.5%)
4(2.0%)
 
Her-2 Human epidermal growth factor receptor-2, TNBC Triple negative breast cancer, LIQ Lower-inner quadrant, LOQ Lower-outer quadrant, UIQ Upper-inner quadrant, UOQ Upper-outer quadrant

Independent predictive factors for positive ALNs

In this study, Univariate logistic regression analysis of the clinicopathological characteristics revealed that for the training cohort, age, CEA, CA125, CA153, tumor size, vascular-invasion, calcification, and tumor grade were significantly associated with ALNs metastasis, showed in Table 2. Histology subtype, biological subtype and location were not identified as a predictor of lymph node metastasis in univariable logistic regression analysis and were not included in the multivariable logistic regression analyses. Multivariate logistic regression analysis demonstrated that CEA (Odds ratio 1.085, 95% CI 1.001–1.079, p = 0.046), CA125 (Odds ratio 1.052, 95% CI 1.027–1.077, p < 0.001), CA153 (Odds ratio 1.073, 95% CI 1.042–1.105, p < 0.001), tumor size (Odds ratio 1.145, 95% CI 1.009–1.299, p = 0.035), vascular-invasion (Odds ratio 7.736, 95% CI 3.903–15.332, p < 0.001), calcification (Odds ratio 1.650, 95% CI 1.082–2.514, p = 0.02) and tumor grade (Odds ratio 5.338, 95% CI 1.514–18.840, p = 0.009 for grade 2, Odds ratio 5.864, 95% CI 1.670-20.594, p = 0.006 for grade 3) were independent prognostic factors for positive ALNs.
Table 2
Uni- and multivariate logistic regression analysis
Variable
Univariate analysis
Multi-variate analysis
 
Odds ratio
95% CI
p-value
Odds ratio
95% CI
P value
Age
0.852
0.733–0.999
0.036
0.993
0.975–1.011
0.435
CEA
1.282
1.188–1.384
 < 0.001
1.085
1.001–1.079
0.046
CA125
1.057
1.036–1.078
 < 0.001
1.052
1.027–1.077
 < 0.001
CA153
1.124
1.094–1.154
 < 0.001
1.073
1.042–1.105
 < 0.001
Tumor size
1.313
1.168–1.475
 < 0.001
1.145
1.009–1.299
0.036
Location
 UOQ
Reference
   
 LOQ
0.605
0.327–1.118
0.109
   
 LIQ
0.706
0.398–1.254
0.236
   
 UIQ
0.659
0.395–1.099
0.110
   
 Central
0.398
0.128–1.241
0.112
   
Vascular-invasion
 No
Reference
Reference
 Yes
10.848
6.652–17.690
 < 0.001
7.736
3.903–15.332
 < 0.001
Calcification
 No
Reference
Reference
 Yes
1.900
1.325–2.271
 < 0.001
1.650
1.082–2.514
0.02
Tumor grade
 Well differentiated, grade 1
Reference
   
 Moderately differentiated, grade 2
4.362
1.775–10.718
0.001
5.338
1.514–18.840
0.009
 Poorly differentiated, grade 3
5.657
2.318–13.804
 < 0.001
5.864
1.670–20.594
0.006
Histology subtype
 Invasive ductal carcinoma
Reference
   
 Invasive lobular carcinoma
1.838
0.755–4.477
0.180
   
 Mixed
2.056
0.964–4.379
0.062
   
 Other
0.503
0.215–4.379
0.113
   
ER status
 Negative
0.693
0.447–1.073
0.101
   
 Positive
Reference
   
PR status
 Negative
0.680
0.469–0.986
0.045
   
 Positive
Reference
   
 KI67
1.006
0.998–1.014
0.117
   
Her-2 status
 Negative
0.920
0.626–1.352
0.673
   
 Positive
Reference
   
Biological subtype
 Luminal A
1.147
0.532–2.474
0.726
   
 Luminal B
1.932
1.046–3.567
0.035
   
 Her-2 positive
1.554
0.706–3.421
0.273
   
 TNBC
Reference
   
Abbreviation: Her-2 Human epidermal growth factor receptor-2, TNBC Triple negative breast cancer, LIQ Lower-inner quadrant, LOQ Lower-outer quadrant, UIQ Upper-inner quadrant, UOQ Upper-outer quadrant

Construction of predictive model

All the significant factors were used to create the nomograms. The nomogram for positive ALN and four or more ALN metastasis was shown in Fig. 1. The value of each variable was given a score on the point scale axis. By adding up the scores related to each variable and projecting total scores to the bottom scales, it is easy to calculate the estimated probabilities for positive ALN and four or more ALN metastasis.

Calibration and validation of nomogram

In order to identify the discriminating ability of nomograms, various methods were used in this study, including calibration curves, C-index values and DCA curves.
In our study, calibration curves in the training group for positive ALN and four or more ALN metastasis were shown in Fig. 2a and b. There were no significant deviations between the calibration curves and the reference line, which revealed optimal agreement between model prediction and actual observations for ALN metastasis. In the validation group, the calibration curves for positive ALN and four or more ALN metastasis were shown in Fig. 2c and d. Similarly, good concordance was observed between the model predicted and the actual observations for ALN metastasis. The C-indexes of the nomogram for prediction of no ALN metastasis were 0.826 (95% CI 0.789–0.863, p < 0.001) in training group and 0.836 (95% CI 0.779–0.893, p < 0.001) in validation group, shown in Fig. 3a and d. The C-indexes of the nomogram for prediction of positive ALN were 0.706 (95% CI 0.656–0.756, p < 0.001) in the training group and 0.731 (95% CI 0.635–0.827, p < 0.001) in the validation group, showed in Fig. 3b and e. The C-indexes of the nomogram for prediction of four and more ALN metastasis were 0.855 (95% CI 0.809-0.900, p < 0.001) in the training group and 0.897 (95% CI 0.846–0.947, p < 0.001) in the validation group, showed in Fig. 3c and f. Moreover, in terms of the clinical utility, DCA analysis showed that our nomogram owned superior net benefits and net reduction in interventions. The nomogram was superior to the traditional staging system including tumor size and grade across the range of reasonable threshold probabilities in both the training group and the validation group, shown in Fig. 4a and b.

Discussion

In this study, we constructed a novel, easy to use and effective nomogram for the prediction of positive ALN and four or more ALN metastasis. After univariant and multivariant logistic regression analysis, CEA (p = 0.046), CA125 (p < 0.001), CA153 (p < 0.001), tumor size (p = 0.035), vascular-invasion (p < 0.001), calcification (p = 0.02) and tumor grade (p = 0.009 for grade 2, p = 0.006 for grade 3) were incorporated as independent predictors of ALN metastasis in patients with breast cancer. In addition, according to the calibration curve and ROC curve analysis in both training group and validation group, the predictive accuracy and concordance of our nomogram were validated. What’s more, DCA analysis also displayed that our nomogram had better a clinical utility and more benefits than the traditional staging system.
ALN status is one of the most important prognostic factors for patients with breast cancer. For breast cancer patients with positive ALNs, it indicates the aggressiveness of the tumor, which may be related to their own biological characteristics. With the in-depth research on tumor molecular mechanisms and tumor microenvironment, the influence of tumor biological characteristics on the prognosis of patients was attached of great importance. Some studies had confirmed the vascular invasion’s ability to discriminate patients’ prognosis [4042]. When concerning breast cancer, serum tumor markers including CEA, CA125, and CA15-3 and calcification had been revealed associated with patients’ clinical outcomes [31, 32, 3537]. Previous researches [59] had tried to construct different nomograms to predict ALN status using patients’ traditional clinicopathological characteristics including tumor size, nuclear grade, and so forth, while most of them did not mention the role of tumor biological characteristics. To our knowledge, this is the first study to construct a nomogram incorporated patients’ clinicopathological and tumor biological characteristic factors. Our predictive model, based on previous studies and combined with tumor biomarkers, can better reflect the role of tumor biological characteristics in the progression of breast cancer. In addition, the results of the calibration curve and ROC curve analysis in both training group and validation group demonstrated the predictive ability and discrimination power of our model. Furthermore, combined the clinical utility with benefit shown in DCA analysis, our nomogram can be utilized effectively to counsel individual patients, thereby helping to personalize the surgical treatment of breast cancer patients.
In patients with positive SLN, present clinically useful predictive models, including the well-known MSKCC nomogram [10] were explicitly designed to predict the non SLN metastasis with patients’ clinicopathological factors. In patients with negative SLN, ALND can safely be avoided. However, M. Klar et Al’s study [43, 44] had shown that side-effects of SLN biopsy without consecutive ALND are not negligible. Furthermore, considering the unreliable results of SLNB caused by the false negative rate [1721] and lower incidence of axillary nodal metastasis at the time of diagnosis [22] due to preoperative examination technique improvement, the benefits of SLNB were reducing. Our novel predictive model incorporated the patient’s clinicopathological and tumor biological characteristics and has better functionality and accuracy. First, our nomogram was proved to be capable of defining 8% of patients as having low-risk (lower than 10% chance) of ALN metastasis (showed in Fig. 4c). The predictive accuracy was high in both training group and validation group (ROC analysis, 0.826, 95% CI 0.789–0.863, p < 0.001 in the training group and 0.836, 95% CI 0.779–0.893, p < 0.001 in the validation group). Second, our nomogram had the ability to define 68.5% of patients as having a low-risk (lower than 10% chance) of four or more ALN metastasis (showed in Fig. 4d). Similarly, the ROC analysis revealed good prediction sensitivity and specificity (ROC analysis, 0.855, 95% CI 0.809-0.900, p < 0.001 in the training group and 0.897, 95% CI 0.846–0.947, p < 0.001 in the validation group). In addition, with the capability to predict breast cancer patients with less than four positive ALN metastasis, our model can improve the utilization efficiency of SLNB and reduce the incidence of complications. At the same time, according to the existing research [1114], it can also provide patients with better clinical treatment decisions based on the results of SLNB.
Some limitations in the present study needs to be considered. Firstly, the number of patients in the validation group enrolled in this study was relatively small. In order to better evaluate and validate the nomogram, more patients need to be enrolled in the study from multiple centers to improve the credibility of the study. Secondly, not all patients in this study underwent ALND. Therefore, it is possible that non-SLN metastasis may have remained. Thirdly, we would incorporate more indexes including the results of images (like mammography or MRI) of axillary lymph node status to improve the establishment of the prediction model in further research.

Conclusion

we successfully construct and independently validate the nomogram to predict patients’ axillary node status by using patients’ clinicopathological and tumor characteristic factors. Furthermore, we sought the possibility to find selected patients to perform SLNB, it would decrease the time of surgery and reduce the postoperative complications, which can give patients better clinical treatment.

Acknowledgements

None.

Declarations

All methods used for this study were performed in accordance with the Declaration of Helsink and our study was approved by the Institutional Review Board of Zhongshan Hospital affiliated to Fudan University. Written informed consent was obtained from all participants included in the study.
Not applicable.

Competing interests

The authors declared no conflicts of interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Krag DN, Anderson SJ, Julian TB, Brown AM, Harlow SP, Costantino JP, Ashikaga T, Weaver DL, Mamounas EP, Jalovec LM, et al. Sentinel-lymph-node resection compared with conventional axillary-lymph-node dissection in clinically node-negative patients with breast cancer: overall survival findings from the NSABP B-32 randomised phase 3 trial. Lancet Oncol. 2010;11(10):927–33.CrossRef Krag DN, Anderson SJ, Julian TB, Brown AM, Harlow SP, Costantino JP, Ashikaga T, Weaver DL, Mamounas EP, Jalovec LM, et al. Sentinel-lymph-node resection compared with conventional axillary-lymph-node dissection in clinically node-negative patients with breast cancer: overall survival findings from the NSABP B-32 randomised phase 3 trial. Lancet Oncol. 2010;11(10):927–33.CrossRef
2.
Zurück zum Zitat Veronesi U, Paganelli G, Viale G, Luini A, Zurrida S, Galimberti V, Intra M, Veronesi P, Maisonneuve P, Gatti G, et al. Sentinel-lymph-node biopsy as a staging procedure in breast cancer: update of a randomised controlled study. Lancet Oncol. 2006;7(12):983–90.CrossRef Veronesi U, Paganelli G, Viale G, Luini A, Zurrida S, Galimberti V, Intra M, Veronesi P, Maisonneuve P, Gatti G, et al. Sentinel-lymph-node biopsy as a staging procedure in breast cancer: update of a randomised controlled study. Lancet Oncol. 2006;7(12):983–90.CrossRef
3.
Zurück zum Zitat Zhu L, Jin L, Li S, Chen K, Jia W, Shan Q, Walter S, Song E, Su F. Which nomogram is best for predictingnon-sentinel lymph node metastasis in breast cancer patients? A meta-analysis. Breast Cancer Res Treat. 2013;137(3):783–95.CrossRef Zhu L, Jin L, Li S, Chen K, Jia W, Shan Q, Walter S, Song E, Su F. Which nomogram is best for predictingnon-sentinel lymph node metastasis in breast cancer patients? A meta-analysis. Breast Cancer Res Treat. 2013;137(3):783–95.CrossRef
4.
Zurück zum Zitat Coutant C, Olivier C, Lambaudie E, Fondrinier E, Marchal F, Guillemin F, Seince N, Thomas V, Levцque J, Barranger E, et al. Comparison of models to predict nonsentinel lymph node status in breast cancer patients with metastatic sentinel lymph nodes: a prospective multicenter study. J Clin Oncol. 2009;27(17):2800–8.CrossRef Coutant C, Olivier C, Lambaudie E, Fondrinier E, Marchal F, Guillemin F, Seince N, Thomas V, Levцque J, Barranger E, et al. Comparison of models to predict nonsentinel lymph node status in breast cancer patients with metastatic sentinel lymph nodes: a prospective multicenter study. J Clin Oncol. 2009;27(17):2800–8.CrossRef
5.
Zurück zum Zitat Barranger E, Coutant C, Flahault A, Delpech Y, Darai E, Uzan S. An axilla scoring system to predict non-sentinel lymph node status in breast cancer patients with sentinel lymph node involvement. Breast Cancer Res Treat. 2005;91(2):113–9.CrossRef Barranger E, Coutant C, Flahault A, Delpech Y, Darai E, Uzan S. An axilla scoring system to predict non-sentinel lymph node status in breast cancer patients with sentinel lymph node involvement. Breast Cancer Res Treat. 2005;91(2):113–9.CrossRef
6.
Zurück zum Zitat Degnim AC, Reynolds C, Pantvaidya G, Zakaria S, Hoskin T, Barnes S, Roberts MV, Lucas PC, Oh K, Koker M, et al. Nonsentinel node metastasis in breast cancer patients: assessment of an existing and a new predictive nomogram. Am J Surg. 2005;190(4):543–50.CrossRef Degnim AC, Reynolds C, Pantvaidya G, Zakaria S, Hoskin T, Barnes S, Roberts MV, Lucas PC, Oh K, Koker M, et al. Nonsentinel node metastasis in breast cancer patients: assessment of an existing and a new predictive nomogram. Am J Surg. 2005;190(4):543–50.CrossRef
7.
Zurück zum Zitat Pal A, Provenzano E, Duffy SW, Pinder SE, Purushotham AD. A model for predicting non-sentinel lymph node metastatic disease when the sentinel lymph node is positive. Br J Surg. 2008;95(3):302–9.CrossRef Pal A, Provenzano E, Duffy SW, Pinder SE, Purushotham AD. A model for predicting non-sentinel lymph node metastatic disease when the sentinel lymph node is positive. Br J Surg. 2008;95(3):302–9.CrossRef
8.
Zurück zum Zitat Kohrt HE, Olshen RA, Bermas HR, Goodson WH, Wood DJ, Henry S, Rouse RV, Bailey L, Philben VJ, Dirbas FM, et al. New models and online calculator for predicting non-sentinel lymph node status in sentinel lymph node positive breast cancer patients. BMC Cancer. 2008;8:66.CrossRef Kohrt HE, Olshen RA, Bermas HR, Goodson WH, Wood DJ, Henry S, Rouse RV, Bailey L, Philben VJ, Dirbas FM, et al. New models and online calculator for predicting non-sentinel lymph node status in sentinel lymph node positive breast cancer patients. BMC Cancer. 2008;8:66.CrossRef
9.
Zurück zum Zitat Van Zee KJ, Manasseh DM, Bevilacqua JL, Boolbol SK, Fey JV, Tan LK, Borgen PI, Cody HS 3rd, Kattan MW. A nomogram for predicting the likelihood of additional nodal metastases in breast cancer patients with a positive sentinel node biopsy. Ann Surg Oncol. 2003;10(10):1140–51.CrossRef Van Zee KJ, Manasseh DM, Bevilacqua JL, Boolbol SK, Fey JV, Tan LK, Borgen PI, Cody HS 3rd, Kattan MW. A nomogram for predicting the likelihood of additional nodal metastases in breast cancer patients with a positive sentinel node biopsy. Ann Surg Oncol. 2003;10(10):1140–51.CrossRef
10.
Zurück zum Zitat Chue KM, Yong WS, Thike AA, Ahmed SS, Li HH, Wong CY, Ho GH, Madhukumar P, Tan BK, Ong KW, et al. Predicting the likelihood of additional lymph node metastasis in sentinel lymph node positive breast cancer: validation of the Memorial Sloan-Kettering Cancer Centre (MSKCC) nomogram. J Clin Pathol. 2014;67(2):112–9.CrossRef Chue KM, Yong WS, Thike AA, Ahmed SS, Li HH, Wong CY, Ho GH, Madhukumar P, Tan BK, Ong KW, et al. Predicting the likelihood of additional lymph node metastasis in sentinel lymph node positive breast cancer: validation of the Memorial Sloan-Kettering Cancer Centre (MSKCC) nomogram. J Clin Pathol. 2014;67(2):112–9.CrossRef
11.
Zurück zum Zitat Galimberti V, Cole BF, Zurrida S, Viale G, Luini A, Veronesi P, Baratella P, Chifu C, Sargenti M, Intra M, et al. Axillary dissection versus no axillary dissection in patients with sentinel-node micrometastases (IBCSG 23 – 01): a phase 3 randomised controlled trial. Lancet Oncol. 2013;14(4):297–305.CrossRef Galimberti V, Cole BF, Zurrida S, Viale G, Luini A, Veronesi P, Baratella P, Chifu C, Sargenti M, Intra M, et al. Axillary dissection versus no axillary dissection in patients with sentinel-node micrometastases (IBCSG 23 – 01): a phase 3 randomised controlled trial. Lancet Oncol. 2013;14(4):297–305.CrossRef
12.
Zurück zum Zitat Giuliano AE, Ballman K, McCall L, Beitsch P, Whitworth PW, Blumencranz P, Leitch AM, Saha S, Morrow M, Hunt KK. Locoregional Recurrence after Sentinel Lymph Node Dissection with or without Axillary dissection in patients with Sentinel Lymph Node Metastases: long-term follow-up from the American College of Surgeons Oncology Group (Alliance) ACOSOG Z0011 Randomized Trial. Ann Surg. 2016;264(3):413–20.CrossRef Giuliano AE, Ballman K, McCall L, Beitsch P, Whitworth PW, Blumencranz P, Leitch AM, Saha S, Morrow M, Hunt KK. Locoregional Recurrence after Sentinel Lymph Node Dissection with or without Axillary dissection in patients with Sentinel Lymph Node Metastases: long-term follow-up from the American College of Surgeons Oncology Group (Alliance) ACOSOG Z0011 Randomized Trial. Ann Surg. 2016;264(3):413–20.CrossRef
13.
Zurück zum Zitat Giuliano AE, Ballman KV, McCall L, Beitsch PD, Brennan MB, Kelemen PR, Ollila DW, Hansen NM, Whitworth PW, Blumencranz PW, et al. Effect of Axillary Dissection vs no Axillary dissection on 10-Year overall survival among women with invasive breast Cancer and Sentinel Node Metastasis: the ACOSOG Z0011 (Alliance) Randomized Clinical Trial. JAMA. 2017;318(10):918–26.CrossRef Giuliano AE, Ballman KV, McCall L, Beitsch PD, Brennan MB, Kelemen PR, Ollila DW, Hansen NM, Whitworth PW, Blumencranz PW, et al. Effect of Axillary Dissection vs no Axillary dissection on 10-Year overall survival among women with invasive breast Cancer and Sentinel Node Metastasis: the ACOSOG Z0011 (Alliance) Randomized Clinical Trial. JAMA. 2017;318(10):918–26.CrossRef
14.
Zurück zum Zitat Donker M, van Tienhoven G, Straver ME, Meijnen P, van de Velde CJ, Mansel RE, Cataliotti L, Westenberg AH, Klinkenbijl JH, Orzalesi L, et al. Radiotherapy or surgery of the axilla after a positive sentinel node in breast cancer (EORTC 10981–22023 AMAROS): a randomised, multicentre, open-label, phase 3 non-inferiority trial. Lancet Oncol. 2014;15(12):1303–10.CrossRef Donker M, van Tienhoven G, Straver ME, Meijnen P, van de Velde CJ, Mansel RE, Cataliotti L, Westenberg AH, Klinkenbijl JH, Orzalesi L, et al. Radiotherapy or surgery of the axilla after a positive sentinel node in breast cancer (EORTC 10981–22023 AMAROS): a randomised, multicentre, open-label, phase 3 non-inferiority trial. Lancet Oncol. 2014;15(12):1303–10.CrossRef
15.
Zurück zum Zitat Fu Y, Chung D, Cao MA, Apple S, Chang H. Is axillary lymph node dissection necessary after sentinel lymph node biopsy in patients with mastectomy and pathological N1 breast cancer? Ann Surg Oncol. 2014;21(13):4109–23.CrossRef Fu Y, Chung D, Cao MA, Apple S, Chang H. Is axillary lymph node dissection necessary after sentinel lymph node biopsy in patients with mastectomy and pathological N1 breast cancer? Ann Surg Oncol. 2014;21(13):4109–23.CrossRef
16.
Zurück zum Zitat Crawford JD, Ansteth M, Barnett J, Glissmeyer M, Johnson NG. Routine completion axillary lymph node dissection for positive sentinel nodes in patients undergoing mastectomy is not associated with improved local control. Am J Surg. 2013;205(5):581–4. discussion 584.CrossRef Crawford JD, Ansteth M, Barnett J, Glissmeyer M, Johnson NG. Routine completion axillary lymph node dissection for positive sentinel nodes in patients undergoing mastectomy is not associated with improved local control. Am J Surg. 2013;205(5):581–4. discussion 584.CrossRef
17.
Zurück zum Zitat Chao C, Wong SL, Ackermann D, Simpson D, Carter MB, Brown CM, Edwards MJ, McMasters KM. Utility of intraoperative frozen section analysis of sentinel lymph nodes in breast cancer. Am J Surg. 2001;182(6):609–15.CrossRef Chao C, Wong SL, Ackermann D, Simpson D, Carter MB, Brown CM, Edwards MJ, McMasters KM. Utility of intraoperative frozen section analysis of sentinel lymph nodes in breast cancer. Am J Surg. 2001;182(6):609–15.CrossRef
18.
Zurück zum Zitat Tanis PJ, Boom RP, Koops HS, Faneyte IF, Peterse JL, Nieweg OE, Rutgers EJ, Tiebosch AT, Kroon BB. Frozen section investigation of the sentinel node in malignant melanoma and breast cancer. Ann Surg Oncol. 2001;8(3):222–6.CrossRef Tanis PJ, Boom RP, Koops HS, Faneyte IF, Peterse JL, Nieweg OE, Rutgers EJ, Tiebosch AT, Kroon BB. Frozen section investigation of the sentinel node in malignant melanoma and breast cancer. Ann Surg Oncol. 2001;8(3):222–6.CrossRef
19.
Zurück zum Zitat Weiser MR, Montgomery LL, Susnik B, Tan LK, Borgen PI, Cody HS. Is routine intraoperative frozen-section examination of sentinel lymph nodes in breast cancer worthwhile? Ann Surg Oncol. 2000;7(9):651–5.CrossRef Weiser MR, Montgomery LL, Susnik B, Tan LK, Borgen PI, Cody HS. Is routine intraoperative frozen-section examination of sentinel lymph nodes in breast cancer worthwhile? Ann Surg Oncol. 2000;7(9):651–5.CrossRef
20.
Zurück zum Zitat Rahusen FD, Pijpers R, Van Diest PJ, Bleichrodt RP, Torrenga H, Meijer S. The implementation of the sentinel node biopsy as a routine procedure for patients with breast cancer. Surgery. 2000;128(1):6–12.CrossRef Rahusen FD, Pijpers R, Van Diest PJ, Bleichrodt RP, Torrenga H, Meijer S. The implementation of the sentinel node biopsy as a routine procedure for patients with breast cancer. Surgery. 2000;128(1):6–12.CrossRef
21.
Zurück zum Zitat Veronesi U, Paganelli G, Viale G, Galimberti V, Luini A, Zurrida S, Robertson C, Sacchini V, Veronesi P, Orvieto E, et al. Sentinel lymph node biopsy and axillary dissection in breast cancer: results in a large series. J Natl Cancer Inst. 1999;91(4):368–73.CrossRef Veronesi U, Paganelli G, Viale G, Galimberti V, Luini A, Zurrida S, Robertson C, Sacchini V, Veronesi P, Orvieto E, et al. Sentinel lymph node biopsy and axillary dissection in breast cancer: results in a large series. J Natl Cancer Inst. 1999;91(4):368–73.CrossRef
22.
Zurück zum Zitat Cady B, Stone MD, Schuler JG, Thakur R, Wanner MA, Lavin PT. The new era in breast cancer. Invasion, size, and nodal involvement dramatically decreasing as a result of mammographic screening. Arch Surg 1996;131(3):301–8.CrossRef Cady B, Stone MD, Schuler JG, Thakur R, Wanner MA, Lavin PT. The new era in breast cancer. Invasion, size, and nodal involvement dramatically decreasing as a result of mammographic screening. Arch Surg  1996;131(3):301–8.CrossRef
23.
Zurück zum Zitat Crabb SJ, Cheang MC, Leung S, Immonen T, Nielsen TO, Huntsman DD, Bajdik CD, Chia SK. Basal breast cancer molecular subtype predicts for lower incidence of axillary lymph node metastases in primary breast cancer. Clin Breast Cancer. 2008;8(3):249–56.CrossRef Crabb SJ, Cheang MC, Leung S, Immonen T, Nielsen TO, Huntsman DD, Bajdik CD, Chia SK. Basal breast cancer molecular subtype predicts for lower incidence of axillary lymph node metastases in primary breast cancer. Clin Breast Cancer. 2008;8(3):249–56.CrossRef
24.
Zurück zum Zitat Wiechmann L, Sampson M, Stempel M, Jacks LM, Patil SM, King T, Morrow M. Presenting features of breast cancer differ by molecular subtype. Ann Surg Oncol. 2009;16(10):2705–10.CrossRef Wiechmann L, Sampson M, Stempel M, Jacks LM, Patil SM, King T, Morrow M. Presenting features of breast cancer differ by molecular subtype. Ann Surg Oncol. 2009;16(10):2705–10.CrossRef
25.
Zurück zum Zitat Ugras S, Stempel M, Patil S, Morrow M. Estrogen receptor, progesterone receptor, and HER2 status predict lymphovascular invasion and lymph node involvement. Ann Surg Oncol. 2014;21(12):3780–6.CrossRef Ugras S, Stempel M, Patil S, Morrow M. Estrogen receptor, progesterone receptor, and HER2 status predict lymphovascular invasion and lymph node involvement. Ann Surg Oncol. 2014;21(12):3780–6.CrossRef
26.
Zurück zum Zitat Dihge L, Bendahl PO, Rydцn L. Nomograms for preoperative prediction of axillary nodal status in breast cancer. Br J Surg. 2017;104(11):1494–505.CrossRef Dihge L, Bendahl PO, Rydцn L. Nomograms for preoperative prediction of axillary nodal status in breast cancer. Br J Surg. 2017;104(11):1494–505.CrossRef
27.
Zurück zum Zitat Ahn SK, Kim MK, Kim J, Lee E, Yoo TK, Lee HB, Kang YJ, Kim J, Moon HG, Chang JM, et al. Can we skip intraoperative evaluation of Sentinel Lymph Nodes? Nomogram Predicting involvement of three or more axillary lymph nodes before breast Cancer surgery. Cancer Res Treat. 2017;49(4):1088–96.CrossRef Ahn SK, Kim MK, Kim J, Lee E, Yoo TK, Lee HB, Kang YJ, Kim J, Moon HG, Chang JM, et al. Can we skip intraoperative evaluation of Sentinel Lymph Nodes? Nomogram Predicting involvement of three or more axillary lymph nodes before breast Cancer surgery. Cancer Res Treat. 2017;49(4):1088–96.CrossRef
28.
Zurück zum Zitat Chen K, Liu J, Li S, Jacobs L. Development of nomograms to predict axillary lymph node status in breast cancer patients. BMC Cancer. 2017;17(1):561.CrossRef Chen K, Liu J, Li S, Jacobs L. Development of nomograms to predict axillary lymph node status in breast cancer patients. BMC Cancer. 2017;17(1):561.CrossRef
29.
Zurück zum Zitat Bevilacqua JL, Kattan MW, Fey JV, Cody HS 3rd, Borgen PI, Van Zee KJ. Doctor, what are my chances of having a positive sentinel node? A validated nomogram for risk estimation. J Clin Oncol. 2007;25(24):3670–9.CrossRef Bevilacqua JL, Kattan MW, Fey JV, Cody HS 3rd, Borgen PI, Van Zee KJ. Doctor, what are my chances of having a positive sentinel node? A validated nomogram for risk estimation. J Clin Oncol. 2007;25(24):3670–9.CrossRef
30.
Zurück zum Zitat Viale G, Zurrida S, Maiorano E, Mazzarol G, Pruneri G, Paganelli G, Maisonneuve P, Veronesi U. Predicting the status of axillary sentinel lymph nodes in 4351 patients with invasive breast carcinoma treated in a single institution. Cancer. 2005;103(3):492–500.CrossRef Viale G, Zurrida S, Maiorano E, Mazzarol G, Pruneri G, Paganelli G, Maisonneuve P, Veronesi U. Predicting the status of axillary sentinel lymph nodes in 4351 patients with invasive breast carcinoma treated in a single institution. Cancer. 2005;103(3):492–500.CrossRef
31.
Zurück zum Zitat Wang W, Xu X, Tian B, Wang Y, Du L, Sun T, Shi Y, Zhao X, Jing J. The diagnostic value of serum tumor markers CEA, CA19-9, CA125, CA15-3, and TPS in metastatic breast cancer. Clin Chim Acta. 2017;470:51–5.CrossRef Wang W, Xu X, Tian B, Wang Y, Du L, Sun T, Shi Y, Zhao X, Jing J. The diagnostic value of serum tumor markers CEA, CA19-9, CA125, CA15-3, and TPS in metastatic breast cancer. Clin Chim Acta. 2017;470:51–5.CrossRef
32.
Zurück zum Zitat Li G, Hu J, Hu G. Biomarker studies in early detection and prognosis of breast Cancer. Adv Exp Med Biol. 2017;1026:27–39.CrossRef Li G, Hu J, Hu G. Biomarker studies in early detection and prognosis of breast Cancer. Adv Exp Med Biol. 2017;1026:27–39.CrossRef
33.
Zurück zum Zitat Radenkovic S, Milosevic Z, Konjevic G, Karadzic K, Rovcanin B, Buta M, Gopcevic K, Jurisic V. Lactate dehydrogenase, catalase, and superoxide dismutase in tumor tissue of breast cancer patients in respect to mammographic findings. Cell Biochem Biophys. 2013;66(2):287–95.CrossRef Radenkovic S, Milosevic Z, Konjevic G, Karadzic K, Rovcanin B, Buta M, Gopcevic K, Jurisic V. Lactate dehydrogenase, catalase, and superoxide dismutase in tumor tissue of breast cancer patients in respect to mammographic findings. Cell Biochem Biophys. 2013;66(2):287–95.CrossRef
34.
Zurück zum Zitat Prat A, Cheang MC, Martцn M, Parker JS, Carrasco E, Caballero R, Tyldesley S, Gelmon K, Bernard PS, Nielsen TO, et al. Prognostic significance of progesterone receptor-positive tumor cells within immunohistochemically defined luminal a breast cancer. J Clin Oncol. 2013;31(2):203–9.CrossRef Prat A, Cheang MC, Martцn M, Parker JS, Carrasco E, Caballero R, Tyldesley S, Gelmon K, Bernard PS, Nielsen TO, et al. Prognostic significance of progesterone receptor-positive tumor cells within immunohistochemically defined luminal a breast cancer. J Clin Oncol. 2013;31(2):203–9.CrossRef
35.
Zurück zum Zitat O’Grady S, Morgan MP. Microcalcifications in breast cancer: from pathophysiology to diagnosis and prognosis. Biochim et Biophys Acta Rev Cancer. 2018;1869(2):310–20.CrossRef O’Grady S, Morgan MP. Microcalcifications in breast cancer: from pathophysiology to diagnosis and prognosis. Biochim et Biophys Acta Rev Cancer. 2018;1869(2):310–20.CrossRef
36.
Zurück zum Zitat Zheng K, Tan JX, Li F, Wei YX, Yin XD, Su XL, Li HY, Liu QL, Ma BL, Ou JH, et al. Relationship between mammographic calcifications and the clinicopathologic characteristics of breast cancer in Western China: a retrospective multi-center study of 7317 female patients. Breast Cancer Res Treat. 2017;166(2):569–82.CrossRef Zheng K, Tan JX, Li F, Wei YX, Yin XD, Su XL, Li HY, Liu QL, Ma BL, Ou JH, et al. Relationship between mammographic calcifications and the clinicopathologic characteristics of breast cancer in Western China: a retrospective multi-center study of 7317 female patients. Breast Cancer Res Treat. 2017;166(2):569–82.CrossRef
37.
Zurück zum Zitat Ling H, Liu ZB, Xu LH, Xu XL, Liu GY, Shao ZM. Malignant calcification is an important unfavorable prognostic factor in primary invasive breast cancer. Asia-Pac J Clin Oncol. 2013;9(2):139–45.CrossRef Ling H, Liu ZB, Xu LH, Xu XL, Liu GY, Shao ZM. Malignant calcification is an important unfavorable prognostic factor in primary invasive breast cancer. Asia-Pac J Clin Oncol. 2013;9(2):139–45.CrossRef
38.
Zurück zum Zitat Agarwal S, Singh A, Bagga PK. Immunohistochemical evaluation of lymphovascular invasion in carcinoma breast with CD34 and D2-40 and its correlation with other prognostic markers. Indian J Pathol Microbiol. 2018;61(1):39–44.CrossRef Agarwal S, Singh A, Bagga PK. Immunohistochemical evaluation of lymphovascular invasion in carcinoma breast with CD34 and D2-40 and its correlation with other prognostic markers. Indian J Pathol Microbiol. 2018;61(1):39–44.CrossRef
39.
Zurück zum Zitat Panagiotopoulos N, Lagoudianakis E, Pappas A, Filis K, Salemis N, Manouras A, Kontzoglou K, Zografos G. Lymphovascular infiltration in the tumor bed is a useful marker of biological behavior in breast cancer. J BUON. 2016;21(5):1082–9. Panagiotopoulos N, Lagoudianakis E, Pappas A, Filis K, Salemis N, Manouras A, Kontzoglou K, Zografos G. Lymphovascular infiltration in the tumor bed is a useful marker of biological behavior in breast cancer. J BUON. 2016;21(5):1082–9.
40.
Zurück zum Zitat Mokdad AA, Singal AG, Marrero JA, Zhu H, Yopp AC. Vascular invasion and metastasis is predictive of outcome in Barcelona clinic liver cancer stage C hepatocellular carcinoma. J Natl Compr Cancer Network. 2017;15(2):197–204.CrossRef Mokdad AA, Singal AG, Marrero JA, Zhu H, Yopp AC. Vascular invasion and metastasis is predictive of outcome in Barcelona clinic liver cancer stage C hepatocellular carcinoma. J Natl Compr Cancer Network. 2017;15(2):197–204.CrossRef
41.
Zurück zum Zitat Shen YN, Bai XL, Li GG, Liang TB. Review of radiological classifications of pancreatic cancer with peripancreatic vessel invasion: are new grading criteria required? Cancer Imaging. 2017;17(1):14.CrossRef Shen YN, Bai XL, Li GG, Liang TB. Review of radiological classifications of pancreatic cancer with peripancreatic vessel invasion: are new grading criteria required? Cancer Imaging. 2017;17(1):14.CrossRef
42.
Zurück zum Zitat Wechman SL, Emdad L, Sarkar D, Das SK, Fisher PB. Vascular mimicry: triggers, molecular interactions and in vivo models. Adv Cancer Res. 2020;148:27–67.CrossRef Wechman SL, Emdad L, Sarkar D, Das SK, Fisher PB. Vascular mimicry: triggers, molecular interactions and in vivo models. Adv Cancer Res. 2020;148:27–67.CrossRef
43.
Zurück zum Zitat Klar M, Foeldi M, Markert S, Gitsch G, Stickeler E, Watermann D. Good prediction of the likelihood for sentinel lymph node metastasis by using the MSKCC nomogram in a german breast cancer population. Ann Surg Oncol. 2009;16(5):1136–42.CrossRef Klar M, Foeldi M, Markert S, Gitsch G, Stickeler E, Watermann D. Good prediction of the likelihood for sentinel lymph node metastasis by using the MSKCC nomogram in a german breast cancer population. Ann Surg Oncol. 2009;16(5):1136–42.CrossRef
44.
Zurück zum Zitat Langer I, Guller U, Berclaz G, Koechli OR, Schaer G, Fehr MK, Hess T, Oertli D, Bronz L, Schnarwyler B, et al. Morbidity of sentinel lymph node biopsy (SLN) alone versus SLN and completion axillary lymph node dissection after breast cancer surgery: a prospective swiss multicenter study on 659 patients. Ann Surg. 2007;245(3):452–61.CrossRef Langer I, Guller U, Berclaz G, Koechli OR, Schaer G, Fehr MK, Hess T, Oertli D, Bronz L, Schnarwyler B, et al. Morbidity of sentinel lymph node biopsy (SLN) alone versus SLN and completion axillary lymph node dissection after breast cancer surgery: a prospective swiss multicenter study on 659 patients. Ann Surg. 2007;245(3):452–61.CrossRef
Metadaten
Titel
Predictive nomogram based on serum tumor markers and clinicopathological features for stratifying lymph node metastasis in breast cancer
verfasst von
Sheng-Kai Geng
Shao-Mei Fu
Hong-Wei Zhang
Yi-Peng Fu
Publikationsdatum
01.12.2022
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2022
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-022-10436-3

Weitere Artikel der Ausgabe 1/2022

BMC Cancer 1/2022 Zur Ausgabe

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Erhöhtes Risiko fürs Herz unter Checkpointhemmer-Therapie

28.05.2024 Nebenwirkungen der Krebstherapie Nachrichten

Kardiotoxische Nebenwirkungen einer Therapie mit Immuncheckpointhemmern mögen selten sein – wenn sie aber auftreten, wird es für Patienten oft lebensgefährlich. Voruntersuchung und Monitoring sind daher obligat.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Perioperative Checkpointhemmer-Therapie verbessert NSCLC-Prognose

28.05.2024 NSCLC Nachrichten

Eine perioperative Therapie mit Nivolumab reduziert das Risiko für Rezidive und Todesfälle bei operablem NSCLC im Vergleich zu einer alleinigen neoadjuvanten Chemotherapie um über 40%. Darauf deuten die Resultate der Phase-3-Studie CheckMate 77T.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.