We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Research ArticleOpen Accesscc iconby icon

Prognostic predictive value of Ki-67 in stage I–II triple-negative breast cancer

    Fengyan Li

    State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China

    ,
    Xinhui Zhou

    State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China

    ,
    Wendie Hu

    State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China

    ,
    Yujie Du

    State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China

    ,
    Jiayuan Sun

    State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China

    &
    Yaxue Wang

    *Author for correspondence: Tel.: +86 177 6062 4136;

    E-mail Address: wangyx3@sysucc.org.cn

    State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China

    Published Online:https://doi.org/10.2144/fsoa-2023-0129

    Abstract

    Aim: Our research aimed to determine an optimal cutoff value and investigate the prognostic predictive function of Ki-67. Materials & methods: We retrospectively enrolled 1146 patients diagnosed with stage I–II triple-negative breast cancer. Disease-free and overall survival were analyzed using the Kaplan–Meier method and the Cox regression model. Results: We classified Ki-67 >45% as the high group (n = 716). A Ki-67 level of >45% was associated with poorer disease-free survival (p = 0.039) and overall survival (p = 0.029). Lymph node stage, neoadjuvant chemotherapy, and radiotherapy were independent predictive variables of prognosis. Conclusion: Triple-negative breast cancer may be further subcategorized according to the Ki-67 level. Neoadjuvant chemotherapy and postoperative radiotherapy can improve the prognosis of early triple-negative breast cancer.

    Plain language summary

    This study aimed to find the best value of Ki-67, which is a marker used in breast cancer. At last, according to the Ki-67 level over 45%, triple-negative breast cancer may be divided into two groups. Based on the level of Ki-67, treatment decisions may be better. However, we still need more studies to confirm this.

    Tweetable abstract

    Triple-negative breast cancer may be further subcategorized according to the Ki-67 level >45%, which is associated with a poorer prognosis. Treatment decisions based on the level of Ki-67 may be more favorable to the prognosis of patients.

    Breast cancer (BC) is a major threat to women's health and has the highest incidence among women worldwide [1]. Triple-negative breast cancer (TNBC), which is rare, accounts for about 15% of all breast cancer (BC) cases [2]. TNBC is a particular type that is negative for estrogen receptor (ER), progesterone receptor (PR) and HER2. Furthermore, compared with the other subtypes, TNBC is linked to a higher risk of tumor recurrence and mortality [3]. Precise, effective and specialized treatments for TNBC have not yet been discovered and are challenging to explore. In a previous investigation [4], six TNBC subtypes with distinct phenotypes were identified. Each subtype responds differently to treatments; the appropriate adjuvant therapy for patients with TNBC should be determined by the sensitivity of these subtypes to chemotherapy and targeted agents. Therefore, the biological variety of TNBC necessitates the identification of subsets with improved prognoses.

    The proliferation marker Ki-67 has been utilized extensively to identify human tumor cells. According to a recent study [5], Ki-67 plays a role in both interphase and mitotic cells, and its cellular distribution alters significantly as the cell cycle advances. In clinical settings, Ki-67 is typically utilized to assess the prognosis and forecast how luminal BCs respond to neoadjuvant chemotherapy (NACT). According to Arafah et al. [6], the expression of Ki-67 is substantially associated with lymph node (LN) metastases, tumor invasion, high tumor nuclear grade, clinical stage, poor survival results and failure to achieve a complete pathological response. According to this meta-analysis [7], an expression of Ki-67 less than 40% is correlated with a reduced risk of local recurrence (LR) and death. A Ki-67 cutoff value of 14% is a crucial factor in classifying BCs into a favorable prognosis luminal A BC and a bad prognosis luminal B BC [8–10]. However, whether this cutoff of Ki-67 can be applied to early TNBC remains unclear.

    Previous studies [11–13] have shown that TNBC is correlated with high expression of Ki-67, contributing to a poor prognosis. Pan et al. [14] found that 82.8% of patients with TNBC had high Ki-67 scores. Therefore, the cutoff value of Ki-67 may also be higher than that of luminal BC. However, experts have varied opinions on the Ki-67 threshold. The median value was the most common method to divide low and high groups [15]. A median Ki-67 expression level of 40% was used by Wang et al. [12] as the cutoff value in their study cohort. A high Ki-67 index, a big tumor size, and LN positivity were linked to shorter disease-free survival (DFS) and overall survival (OS). According to Zhu X et al. [13], the cutoff Finder determined that the ideal cutoff for Ki-67 was 30%, and the high group's DFS and OS were poorer. Postoperative chemotherapy (POCT) correlated with better OS in patients with N0 (no lymph node metastases) TNBC in the high group than in the low group. In some previous studies [16–18], the cutoff value for Ki-67 has ranged from 10% to 65%, and the value for TNBC is significantly higher than that for other subtypes [16]. A poor prognosis was significantly more probable in patients with TNBC with a high Ki-67 score.

    Although there have been numerous studies on Ki-67 and TNBC, the best cutoff value and applicability in TNBC remain unknown, particularly in stage I–II TNBC. Therefore, this study aimed to determine the best Ki-67 cutoff and its role in predicting TNBC prognosis.

    Materials & methods

    Patients

    Our study used case data from 1519 patients in the Single Disease Database, who underwent preliminary screening for stage I–II TNBC at the Sun Yat-sen University Cancer Center (SYSUCC), between January 2013 and December 2020. The following were the inclusion criteria: Surgery and pathological immunohistochemistry (IHC) of SYSUCC. Both clinically and pathologically confirmed stage I or II TNBC (American Joint Committee on Cancer 8th edition). All participants were aged between 18 and 80 years. Complete and clear clinical data. The exclusion criteria included male patients with BC, history of bilateral BC, lack of follow-up, combination with other malignant tumors and NACT at another institution.

    Pathology & immunohistochemistry

    All participants underwent surgery at SYSUCC, and tissue specimens, including the tumor, sentinel LN and axillary lymph nodes (ALNs), were regularly stained with hematoxylin and eosin (H&E). Tissue slices were embedded in paraffin to obtain tumor samples. All surgical tissues were fixed in formalin buffered with 10% neutral phosphate buffer. H&E were used to color representative tumor chunks that were embedded in 4 m-thick paraffin and cut into slices. Furthermore, IHC was used to evaluate the expression of ER, PR, HER2 and Ki-67. The American Society of Clinical Oncology/College of American Pathologists [19] recommendations on ER and PR statuses were categorized as negative if the values were <1%. When the IHC results were 0, 1, or 2+ without HER2 gene amplification on FI, the HER2 status was considered negative [20]. Three pathologists retrospectively reviewed archived H&E and IHC slides to confirm the diagnosis. With a strict quality assurance and control system to guarantee the validity of the analyses, our center strictly complied with the standardized ‘typewriter’ visual assessment method recommended by the ‘International Ki-67 in Breast Cancer Working Group Assessment Guidelines’ [21]. IHC negative and positive pairs of photos were cut, and each IHC test set negative and positive control quality controls were used to ensure the accuracy and reliability of IHC results.

    Follow-up methods & clinical outcome assessment

    We used outpatient records, telephone calls, and the SYSUCC YIDU CLOUD Intelligent Follow-up Platform to complete follow-up and count from the day of BC diagnosis by pathology. The primary end points were DFS and OS. OS was defined as the time from initial pathological diagnosis to death. The time from the first pathological or radiological diagnosis to LR, metastasis, or BC-related mortality was referred to as DFS. Various tests were utilized to confirm suspected metastases, such as in the bone, using an MRI and bone scan, and in the lung, using a chest computed tomography, puncture biopsy, or PET. All patients were followed up until death or until 30 September 2022.

    Statistical methods

    The optimal margin for Ki-67 was assessed using X-tile (version 3.6.1), a tool developed by Yale University (http://tissuearray.org). It is a bioinformatics tool for biomarker assessment and outcome-based cut-point optimization [22]. The chi-squared test was used to analyze categorical variables. Moreover, using the Cox regression model and the Kaplan-Meier method, DFS and OS were contrasted between the low and high groups. Statistical significance was set at p < 0.05. The computations were performed using the SPSS software (version 27.0).

    Results

    Patients' characteristics & treatments

    Overall, 1146 patients with stage I–II TNBC were recruited; the median follow-up time was 59 months. The optimal Ki-67 cutoff value was 45%, as determined by the x-tile (DFS log-rank p = 0.038, OS log-rank p = 0.028). There were 716 and 430 patients with Ki-67 >45% and <45%, respectively. The median age of patients was 47 years (range: 20–80 years). In the whole cohort, 20.4% and 68.1% had grade II & III, respectively, while the other types included medullary carcinoma and saprophytic carcinoma, totaling 11.5%. In the overall cohort, 23.8% accepted breast-conserving surgery (BCS) and 76.2% underwent a mastectomy. Regarding axillary surgery, 24.9% of the patients underwent sentinel lymph node biopsy (SLNB), and patients with axillary lymph node dissection (ALND) made up 75.1% of the group. According to postoperative pathology, 22.3%, 6.7% and 1.0% of patients had intravascular cancer thrombosis (ICT), nerve tract invasion (NTI), and extra lymph node invasion (ELNI), respectively. There were 65.5% of patients without LN metastasis, 37.6% with tumor dimension ≤2 cm, and 66.2% with tumor size ranging from 2 to 5 cm. Based on the TNM stage, 27.1% and 72.9% of patients had stages I & II tumors, respectively. Regarding adjuvant therapy regimens, 15.2% of the patients received NACT, including regimens based on anthracycline and paclitaxel, and 87.6% received adjuvant POCT. In addition to chemotherapy regimens based on anthracyclines and paclitaxel, 87.6% of patients received adjuvant POCT, platinum-based chemotherapy, and various oral chemotherapy regimens, such as capecitabine. Overall, 41.1% of the patients underwent postoperative radiotherapy (PORT), and the radio sites included the breast, chest wall and supraclavicular LN area; some patients were also irradiated with internal breast LN. The radiotherapy dose was usually 50 Gray/25 fractions, and some patients received tumor bed boost doses or used hyper-fractionation techniques.

    Patients in the high Ki-67 group were younger (p < 0.001) and more likely to be in a premenopausal state (p < 0.001) than those in the low Ki-67 group. Patients in the high group also had a higher histological grade, with 78.1% and 51.4% in the high and low groups, respectively. More participants had POCT in the high group (89.4% vs 84.7%; p = 0.019). Patient characteristics in the different Ki-67 expression groups are presented in Table 1.

    Table 1. Patients' characteristics and treatments.
    Characteristics (n = 1146)Low Ki-67 (≤45) N%High Ki-67 (>45) N%p-valueTotal
    Age  <0.001 
    ≤50235 (54.7%)503 (70.3%) 738 (64.4%)
    >50195 (45.3%)213 (29.7%) 408 (35.6%)
    Menstrual state  <0.001 
    Premenopause235 (54.7%)496 (69.3%) 731 (63.8%)
    Postmenopause195 (45%)220 (30.7%) 415 (36.2%)
    Tumor location  0.952 
    Left223 (51.9%)370 (51.7%) 593 (51.7%)
    Right207 (48.1%)346 (48.3%) 553 (48.3%)
    Breast surgery  0.727 
    Breast-conserving surgery100 (23.2%)173 (24.2%) 273 (23.8%)
    Mastectomy330 (76.7%)543 (75.8%) 873 (76.2%)
    Axillary surgery  0.771 
    Sentinel lymph node biopsy109(25.3%)176(24.6%) 285(24.9%)
    Axillary lymph node dissection321(74.7%)540(75.4%) 861(75.1%)
    Intravascular cancer thrombus  0.877 
    Yes95 (22.1%)161 (22.5%) 256 (22.3%)
    No335 (77.9%)555 (77.5%) 890 (77.7%)
    Nerve tract invasion  0.317 
    Yes33 (7.7%)44 (6.1%) 77 (6.7%)
    No397 (92.3%)672 (93.9%) 1069 (93.3%)
    Extrinsic lymph node invasion  0.766 
    Yes5 (1.2%)7 (1.0%) 12 (1.0%)
    No425 (98.8%)709 (99.0%) 1134 (99.0%)
    Histology stage  <0.001 
    II152 (35.3%)82 (11.4%) 234 (20.4%)
    III221 (51.4%)559 (78.1%) 780 (68.1%)
    Other types57 (13.3%)75 (10.5%) 132 (11.5%)
    Lymph node stage  0.292 
    N0290 (67.4%)461 (64.4%) 751 (65.5%)
    N1140 (32.6%)255 (35.6%) 395 (34.5%)
    Tumor stage  0.178 
    T1175 (40.7%)256 (35.8%) 431 (37.6%)
    T2248 (57.7%)442 (61.7%) 690 (66.2%)
    T37 (1.6%)18 (2.5%) 25 (2.2%)
    TNM stage1  0.082 
    I129 (30.0%)181 (25.3%) 310 (27.1%)
    II301 (70.0%)535 (74.7%) 836 (72.9%)
    Neoadjuvant chemotherapy  0.904 
    Yes66 (15.3%)108 (15.1%) 174 (15.2%)
    No364 (84.7%)608 (84.9%) 972 (84.8%)
    Postoperative chemotherapy  0.019 
    Yes364 (84.7%)640 (89.4%) 1004 (87.6%)
    No66 (15.3%)76 (10.6%) 142 (12.4%)
    Radiotherapy  0.735 
    Yes174 (40.5%)297 (41.5%) 471 (41.1%)
    No256 (59.5%)419 (58.5%) 675 (58.9%)
    Total430716 1146

    1 TNM staging is based on the American Joint Committee on Cancer 8th.

    N0: No lymph node metastases; N1: One to three axillary lymph node metastases; T1: The tumor is ≤2 cm in length; T2: The tumor is >2 cm or ≤5 cm in length; T2: The tumor is >2 cm or ≤5 cm in length.

    Neoadjuvant chemotherapy information

    In our study, 174 (15.2%) patients underwent preoperative NACT. Since most of the patients in this study had a puncture biopsy performed at other hospitals, preoperative IHC was not available in some cases, so we only counted the pre-treatment Ki-67 values of patients who had a preoperative biopsy and NACT at our institution. One hundred (57.5%, n = 174) patients had Ki-67 values above 45% on preoperative biopsy, while 108 (62.1%, n = 174) patients had Ki-67 values above 45% on postoperative IHC. Ki-67 values decreased in 112 (64.4%, n = 174) cases and increased in 36 (20.7%) cases after NACT. Finally, according to the postoperative pathology report, a total of 94 (54.0%, n = 174) patients reached pCR, including 68.1% (64, n = 94) in the high postoperative Ki-67 group and 31.9% (30, n = 94) in the low Ki-67 group. We performed a chi-square test for the relationship between pCR and postoperative Ki-67 values but did not find a significant difference. Additional information is shown in Table 3.

    Table 2. Neoadjuvant chemotherapy information.
    CharacteristicsLow Ki-67 (≤45%)High Ki-67 (>45%)Chi-square p-valueTotal (n = 174)
    NACTYes66 (37.9%)108 (62.1%)0.904174
    No364 (37.4%)608 (62.6%) 972
    NACT Ki-67 biopsyLow58 (78.4%)16 (21.6%)<0.00174 (42.5%)
    High8 (8.0%)92 (92.0%) 100 (57.5%)
    Ki-67 changeDecrease37 (33.0%)75 (67.0%)0.025112 (64.4%)
    Increase13 (36.1%)23 (63.9%) 36 (20.7%)
    No changes16 (61.5%)10 (38.5%) 26 (14.9%
    NACT pCRYes30 (31.9%)64 (68.1%)0.07694 (54.0%)
    No36 (45.0%)44 (55.0%) 80 (46.0%)

    NACT: Neoadjuvant chemotherapy; pCR: Pathological complete response.

    Table 3. Recurrence, metastasis and death.
     Low Ki-67 (≤45%) N%High Ki-67 (>45%) N%p-valueTotal
    Primary events  0.076 
    Yes80 (18.6%)165 (23.0%) 245 (21.4%)
    No350 (81.4%)551 (77.0%) 901 (78.6%)
    Death    
    Yes59 (13.7%)122 (17.0%)0.136181 (15.8%)
    No371 (86.3%)594 (83.0%) 965 (84.2%)
    Total430716 1146

    Primary events were defined as pathological or radiological diagnoses of recurrence, metastasis, or BC-related death.

    In 174 patients who underwent NACT, we also used the Kaplan-Meier method and log-rank test to analyze the relationship between pCR and prognosis. However, results showed that whether pCR was achieved or not did not affect DFS (p = 0.078) and OS (p = 0.837).

    Recurrence, metastasis & death

    Primary events were defined as pathological or radiological diagnoses of recurrence, metastasis, or BC-related death. A median follow-up period of 59 months later, 245 (21.4%) primary events were observed, with 80 (18.6%) and 165 (23.0%) in the low and high groups, respectively (p = 0.076). Overall, 181 (15.8%) and 122 (17.0%) patients died during the follow-up period in the low and high groups, respectively. The related events are presented in Table 2.

    Ki-67 cutoff at 45% is an independent factor for OS & DFS

    High Ki-67 expression index was remarkably linked with poorer DFS (log-rank p = 0.039; Figure 1A) and OS (log-rank p = 0.029; Figure 1B). We conducted a similar Kaplan–Meier method for the N stage, and the results showed that it was also substantially correlated with worse DFS (log-rank p < 0.001; Figure 1C) and OS (log-rank p < 0.001; Figure 1D).

    Figure 1. Kaplan-Meier survival curve in triple-negative breast cancer patients.

    High Ki-67 expression index was remarkably linked with poorer DFS (log-rank p = 0.039; Figure 1A) and OS (log-rank p = 0.029; Figure 1B). The lymph node stage was also substantially correlated with worse DFS (log-rank p < 0.001; Figure 1C) and OS (log-rank p < 0.001; Figure 1D). (A) Disease-free survival curve of the Ki-67 group; (B) overall Survival curve of the Ki-67 group; (C) disease-free survival curve of the lymph node stage group; (D) overall survival curve of the lymph node stage group.

    DFS: Disease-free survival; N0: no lymph node metastases; N1: One to three axillary lymph node metastases; OS: Overall survival.

    Univariate analysis & multivariate analysis

    In the univariate analysis, high Ki-67 expression, ICT, NTI, ELNI, LN positivity, stage II, no NACT and no POCT were associated with shorter DFS. High Ki-67 expression, ICT, ELNI, LN positivity, stage II, no NACT and no POCT were associated with worse OS in univariate analysis. The prognosis was unaffected by additional clinical or pathological factors such as age at diagnosis, tumor location, POCT and tumor size, among others. The LN stage, TNM stage, NACT, and POCT were predictive variables for DFS and OS as per the multivariate analysis. Additionally, NTI and tumor size >5 cm could somewhat influence the DFS. The results of the univariate and multivariate Cox regression analyses are presented in Tables 4A & B.

    Table 4. The Univariate and Multivariate Analysis of DFS and OS.
    A. Univariate and multivariate analysis of DFS.
    VariablesDFSUnivariateMultivariate
     HR95% CIp-valueHR95% CIp-value
    Age (years)≤50 vs >500.8970.688–1.1700.424   
    Menstrual statePre vs post0.8890.682–1.1590.384   
    Tumor locationLeft vs right1.0180.792–1.3070.892   
    Breast surgeryBCS vs mastectomy0.9370.700–1.2540.661   
    Axillary surgerySLNB vs ALND1.0500.782–1.4090.747   
    Ki-67Low vs high1.3231.013–1.7290.0401.3571.017–1.8100.038
    Intravascular cancer thrombusYes vs no0.7520.567–0.9970.0470.8760.645–1.1890.396
    Nerve tract invasionYes vs no0.5850.387–0.8850.0110.6320.411–0.9730.037
    Extrinsic lymph node invasionYes vs no0.3360.149–0.7560.0080.6610.285–1.5330.335
    Histology stageII vs III0.8170.602–1.1080.1930.6630.486–0.9330.017
    II vs other types1.0700.696–1.6460.7571.0580.680–1.6460.802
    Lymph node stageN0 vs N11.7601,370-2.263<0.0013.8452.684–5.508<0.001
    Tumor stageT1 vs T21.2830.979–1.6800.0701.3010.856–1.9760.218
    T1 vs T31.7180.793–3.7220.1702.9631.228–7.1520.016
    TNM stageI vs II1.4461.062–1.9690.0190.7910.466–1.3430.385
    Neoadjuvant chemotherapyYes vs no1.5731.047–2.3620.0292.2401.434–3.500<0.001
    Postoperative chemotherapyYes vs no1.1630.806–1.6790.4201.1520.774–1.6860.468
    RadiotherapyYes vs no1.4391.103–1.8780.0072.4571.770–3.409<0.001
    B. Univariate and multivariate analysis of OS.
    VariablesOSUnivariateMultivariate
      HR95% CIp-valueHR95% CIp-value
    Age (year)≤50 vs >500.9700.714–1.3180.846   
    Menstrual statePre vs post0.9650.711–1.3080.816   
    Tumor locationLeft vs right1.1000.822–1.4730.520   
    Breast surgeryBCS vs mastectomy1.1330.789–1.6270.498   
    Axillary surgerySLNB vs ALND1.0650.749–1.5130.727   
    Ki-67Low vs high1.4131.035–1.9290.0301.4121.011–1.9720.043
    Intravascular cancer thrombusYes vs no0.6800.493–0.9390.0190.7980.563–1.1310.205
    Nerve tract invasionYes vs no0.6350.390–1.0330.0680.7060.424–1.1730.179
    Extrinsic lymph node invasionyes vs no0.2470.109–0.558<0.0010.6170.263–1.4490.268
    Histology stageII vs III0.8260.578–1.1790.2920.6990.479–1.0210.064
    II vs other types1.0650.647–1.7510.8051.1270.674–1.8850.647
    Lymph node stageN0 vs N12.1391.598–2.863<0.0015.4583.628–8.211<0.001
    Tumor stageT1 vs T21.3410.978–1.8390.0681.4610.909–2.3490.117
    T1 vs T31.2800.465–3.5290.6332.6540.862–8.1690.089
    TNM stageI vs II1.4861.032–2.1410.0330.6580.358–1.2080.177
    Neoadjuvant chemotherapyYes vs no1.9971.195–3.3370.0083.1021.789–5.380<0.001
    Postoperative chemotherapyYes vs no1.1880.779–1.8130.4241.2060.775–1.8770.407
    RadiotherapyYes vs no1.4471.060–1.9740.0202.7881.906–4.077<0.001

    TNM staging is based on the American Joint Committee on Cancer 8th.

    ALND: Axillary lymph node dissection; BCS: Breast-conserving surgery; DFS: Disease-free survival; OS: Overall survival; Pre: Premenopause; Post: Postmenopause; SLNB: Sentinel lymph node biopsy.

    Subgroup analysis

    Regarding the subgroup analysis, in the Ki-67 high group, patients with ICT, NTI, ENTI, LN positivity, no NACT, no POCT and no PORT had worse OS in a univariate analysis. However, only the LN stage, NACT and POCT influenced OS in the multivariate analysis. At the same time, patients with NTI, ENTI, LN positivity, no NACT and no PORT had worse DFS in a univariate analysis. While only the LN stage, NACT and POCT affected DFS in the multivariate analysis. In the N0 group, the patients with high Ki-67 and NTI positivity had significantly poorer DFS and OS rates in univariate analysis and multivariate analysis. Because NACT is rarely performed in node-negative patients, the hazard ratio (HR) was <1 in the N0 group. However, in the N1 (one to three axillary lymph node metastases) group, Ki-67 status did not affect prognosis. Overall, the LN stage, NACT and POCT were significant prognostic factors for the Ki-67 high and N0 subgroups. The results of the Ki-67 high and N0 group analyses are shown in Figure 2. All subgroup analyses are presented in the Supplementary Table 1.

    Figure 2. Subgroups of the univariate and multivariate analysis in the Ki-67 high group and N0 group.

    The adjusted p-value and hazard ratio were obtained from the Cox regression model. (A) Analysis for disease-free survival in the Ki-67 high group; (B) analysis for overall survival in the Ki-67 high group; (C) analysis for disease-free survival in the N0 group; (D) analysis for overall survival in the N0 group.

    DFS: Disease-free survival; ELNI: Extra lymph node invasion; HR: Hazard ratio; ICT: Intravascular cancer thrombosis; N0: No lymph node metastases; NACT: Neoadjuvant chemotherapy; NTI: Nerve tract invasion; OS: Overall survival; POCT: Postoperative chemotherapy.

    Discussion

    Due to the aggressive biological behavior of this subtype and the absence of effective targeted therapy options, TNBC has a poor prognosis and is associated with a significant risk of early recurrence. Therefore, there is an urgent need to identify valuable prognostic factors and refine the TNBC classification, which has become a research hotspot recently. IHC monitoring of Ki-67 is an inexpensive and convenient technique that can be extensively used in clinical practice [18]. Based on the available data, the International Ki-67 in Breast Cancer Working Group has suggested criteria for analyzing, reporting, and using this economically effective marker [21]. This makes the detection of Ki-67 more standardized, streamlined, and reasonable.

    We enrolled 1146 participants in this study to examine the prognostic value of Ki-67 in early TNBC. All patients were chosen from a single clinical center to ensure uniformity in the caliber of the pathological marker tests, clinical diagnosis, condition assessment and therapy choices. Ki-67 cutoff levels have been used inconsistently and contentiously in TNBC up to this point, and the baseline Ki-67 level for TNBC is much greater than that for luminal BCs [16]. Recently, many studies [12–15,23,24] have discussed the optimal critical value for Ki-67 expression. Mighri et al. reported a Ki-67 value >20% correlated with early age at diagnosis, LN positivity, tumor grade and a high risk of relapse [23]. According to the ROC curves in Zenzola et al.’s study [24], 60% was the ideal cutoff for Ki-67 expression. For DFS, Ki-67 expression above 60% was an adverse prognostic factor in patients aged >40 years in multivariate analysis. In this study [25], in patients with locally advanced TNBC, Ki-67 protein expression of more than 40% does not necessarily translate into an increased risk of distant metastasis over the first two years following diagnosis. In our study, we concluded that lymph node staging also significantly affects the prognosis of early-stage TNBC and that Ki-67 loses its predictive value when there are metastases in the lymph nodes. Therefore, we believe that the metastasis of the lymph node in locally advanced TNBC is critical to the prognosis, so this study focuses on patients with early-stage TNBC. In our investigation, we used X-tile to determine the optimal outcome and discovered that the best Ki-67 cutoff value for predicting prognosis in TNBC patients was 45%. However, an authoritative cutoff value has not been established; therefore, further studies are needed to explore this.

    Numerous clinical trials [26,27] have shown an association between the degree of Ki-67 expression and potential therapy regimens' success. Previous studies [27] have demonstrated that Ki-67 is one chemosensitivity marker of BC. In contrast, patients with N1 who had significant Ki-67 expression in their TNBC did not benefit from POCT in our study (HR: 1.939; CI: 1.013–3.712; p = 0.046) but did benefit from OS. This phenomenon may be caused by several factors, including that POCT was administered regardless of Ki-67 levels in TNBC. Therefore, survival differences between the high and low groups were difficult to detect. Moreover, the prognosis of patients with N0 is remarkably better than that of patients with N1; therefore, POCT may not benefit patients with N0.

    According to the different classification methods, there are various subtypes of TNBC. Histology, IHC, and FISH assays revealed that TNBCs are primarily basal-like BCs at the gene expression level [28]. Invasive ductal carcinoma is the most prevalent type of TNBC; however, several distinct varieties exist, including adenoid cystic carcinoma, medullary carcinoma and metaplastic carcinoma. TNBC is a category of disorders with varied molecular genetics, and six subtypes of TNBC were identified by Lehmann et al. [4], based on molecular typing, including immunomodulatory, mesenchymal, mesenchymal stem-like, luminal androgen receptor (LAR) and two basal-like subtypes. Burstein et al. [29] also identified four distinct subtypes as follows: LAR, mesenchymal, basal-like immune-suppressed and basal-like immune-activated, supporting the idea that the four subgroups of TNBC can be identified at the transcriptome level. Notably, DFS was the lowest among the basal-like immune-activated subtypes.

    Recently, the largest TNBC cohort in the world was mapped by Professor Shao Zhimin and his team [30] at the Cancer Hospital at Fudan University. The team used four IHC markers to simplify TNBC typing and aid in the clinical molecular typing of the disease. The authors classified TNBCs into four transcriptome-based subtypes: LAR, immunomodulatory, basal-like immunosuppressed and mesenchymal. The international system, based on the multidimensional omics big data system, which explores a new route for determining the target of TNBC, has also recommended this as the first TNBC classification standard. Other experts [31] have found that folate receptor alpha (FRα) is necessary for the biosynthesis of amino acids and nucleotide bases. Given that FRα is highly expressed in TNBC and correlated with clinicopathological factors, it may be possible to identify a subpopulation of patients who could serve as prospective targets for novel therapeutic approaches in managing this type of BC. However, genetic profiling and multiplex IHC tests are not commonly used in clinical practice. For instance, our center does not currently perform pathology testing for further molecular typing of TNBC. Therefore, we can attempt to stage TNBC based on commonly used IHC markers, such as Ki-67. I believe that if the classification of TNBC can indeed be further classified according to Ki-67, we can combine Ki-67 with the subtypes of TNBC for further discussion. In addition, the sensitivity of these new subtypes of chemotherapy, radiotherapy, targeted drugs, and endocrine drugs is still unknown; therefore, larger clinical trials are needed to explore the subtype's therapeutic sensitivity.

    One study [32] showed that TNBC is regularly correlated with young groups, and most cases are poorly differentiated. Similarly, Vihervuori et al. demonstrated that age at diagnosis is one of the most important predictors of disease-specific death in TNBC [33]. This is consistent with our findings that 64.4% of patients were <50 years old in the entire cohort, 70.3% in the Ki-67 high group were <50 years old, and 54.7% were <50 years old in the low group (p < 0.001). Among them, 78.1% of the patients in the high group were histologically graded as grade III, whereas only 51.4% in the Ki-67 low expression group were grade III (p < 0.001). However, in our study, patient age at BC diagnosis and menopausal status did not affect DFS or OS, contrary to the findings of these studies [34,35].

    In addition, the N stage is still a prominent prognostic element for TNBC [12,13]. In our study, DFS (HR: 3.931, 95% CI: 2.749–5.622; p < 0.001) and OS (HR: 5.550, 95% CI: 3.698–8.330; p < 0.001) were better in LN negative patients than in LN positive ones. Furthermore, a large-scale clinical study [35] of patients with TNBC confirmed that the LN ratio among LN-positive predicts the LR rate and survival rate, while the Ki-67 score appears to predict mortality rather than recurrence.

    Although TNBC is characterized by high rates of disease recurrence and poor survival, it is noticeably more susceptible to chemotherapy than the other BC subtypes [18]. Consequently, chemotherapy remains the mainstay of treatment. Sequential anthracycline-and taxane-based NACT, with a full pathological response that strongly correlates with long-term survival outcomes, is a standard therapeutic strategy for many patients with early TNBC. However, compared with nab-paclitaxel plus gemcitabine in TNBC, this first sizable randomized trial [36] supports great efficacy and tolerability of the neoadjuvant nab-paclitaxel plus carboplatin regimen. Zhang et al. [37] suggested NACT + BCS as a successful treatment for TNBC, with a favorable long-term outlook and an obvious short-term impact. Moreover, high Ki-67 expression and clinical stage I were independent protective variables against NACT effectiveness. High Ki-67 scores and non-declining Ki-67 scores were powerful risk factors for outcomes. In our study, NACT was an independent predictor of DFS (HR: 2.263, 95% CI: 1.449–3.534; p < 0.001) and OS (HR: 3.131, 95% CI: 1.806–5.429; p < 0.001) in the whole group of patients. Additionally, NACT significantly affected prognosis in the high Ki-67 group rather than in the low Ki-67 group. Therefore, the effectiveness and outcomes of NACT in patients may be predicted using Ki-67 expression. Simultaneously, patients with TNBC without a pathological complete response (pCR) after NACT are more likely to relapse, and their prognosis is worse, which is different from the results of our studies. Overall, the most often mentioned biomarkers linked to a pCR were elevated Ki-67 expression [38]. Significant correlations between pCR and Ki-67 expression over 20% were found by Toss et al. [39]. However, the best threshold for pCR prediction in this study [40] was 50%.

    In addition to Ki-67's ability to predict the efficacy of NACT to a certain extent, some scholars have recently found that the condition of the androgen receptor (AR) can also affect the therapeutic effect. According to Zhu et al. [41], lack of expression was an independent predictor of pCR, whereas high histological grade, low Ki-67 expression and incomplete NACT treatment courses were shown to be independent indicators of a progressive illness. Some scholars [42] also demonstrated that AR is a strong positive prognostic factor in BCs expressing ERs. According to Mohammed et al. [43], compared with quadruple-negative BC, AR + TNBC exhibits a lower rate of pCR, which may indicate that this subtype has some chemoresistance. However, AR may not be a useful biomarker or therapeutic target in Iranian patients with TNBC [44]. Additionally, a diverse collection of TNBC was identified using AR. TNBCs are classified into three risk categories by Astvatsaturyan K et al. [45]. High-risk (AR-EGFR+) TNBCs, which represent the basal molecular subtype with the worst prognosis, may benefit most from POCT.

    Many researchers have examined factors other than Ki-67 expression. The p53 and Ki-67 were shown to have predictive significance for chemotherapy sensitivity and prognosis in patients with TNBC by Zhang G et al. [46]. In this study [47], the cluster of differentiation 117 (CD117+)/tumor protein P53 (TP53) missense mutation was associated with the markers of proliferation, Ki-67, ICT and recurrence in TNBC. Other studies have found that these factors can also affect the prognosis of TNBC, including tumor-infiltrating lymphocytes (TILs), CD103 iTIL density [48], positive prolactin receptor [49], adipophilin [50], erythropoietin receptor [51] and increased Bcl-2 expression [52]. TILs, as the principal body of the tumor immune microenvironment, have been confirmed by previous large-scale clinical experiments [53–55] as powerful influencing factors for the prognosis and survival of TNBC. TILs are strong predictive indicators of the response to NACT and immunotherapy [56,57] for TNBC patients. Furthermore, in addition to sTILs in the training set, High Ki-67 (cutoff >35%) was found to be the only predictor of pCR [58]. However, certain challenges are still associated with the usual clinical application of genetic tests and other factors. Conversely, the Ki-67 test is more practical, affordable and viable. Additionally, new detection techniques should be developed and widely used in daily clinical practice to make the detection of TNBC subtypes more convenient, efficient, and accurate.

    Regarding microscopic pathology, according to a meta-analysis [59], the sentinel LNs of ELNI were associated with a higher risk of both mortality and tumor recurrence in BC. In Aziz et al.’s study [60], the ELNI was an independent prognostic factor and was measured along with cancer burden in the therapeutic regimen of ALN-positive patients. Additionally, ICT has been considered an important link to metastasis in BC for a long time [61,62]; there are few studies on NTI currently. Although in our study, ELNI and ICT's effects on prognosis were insignificant, NTI is one of the crucial factors influencing the DFS in early TNBC, particularly in LN-negative patients.

    The results of one study [63] have shown that PORT benefits patients with T1/T2 TNBC with more than four positive ALNs. The prognosis of patients with TNBC without LN metastasis treated with BCS plus PORT was comparable to that of those treated with MCM. This finding [64] also showed a significantly higher risk of LR in T1–2N0 TNBC treated with MCM without PORT than in those treated with BCS. In our study, PORT also improved DFS and OS in the entire cohort, regardless of the TNM stage. We were not able to explore the combined effects of surgical modalities and the LN stage on radiotherapy; this presents an opportunity for future research. Conversely, in a review [65], TNBC showed resistance to radiotherapy and did not change the LR rate. However, further exploration is required to understand the relevance of PORT in TNBC.

    At the same time, our research has some limitations. First, we used the X-tile tool to determine the cutoff value in the whole cohort. Hence, to ensure the consistency of the tool's result from our entire cohort, we did not perform propensity score matching, and as a result, the results may be partially biased. Second, we did not analyze the subgroups of TNM stage or surgical methods; therefore, no further data support the efficacy of radiotherapy. Finally, regarding the novelty and innovation of this paper, although many studies cited here have concluded that high Ki-67 has a worse prognosis in TNBC, they have also given their cutoff values. However, there are still no official guidelines or treatment strategies to define this threshold in TNBC, so we believe that more exploration is necessary to define the Ki-67's cutoff point. Although this is a very simple study, the investigators believe that the results of many simple studies may ultimately influence patient treatment decisions. As we described, TNBC is highly heterogeneous, and maybe the value of Ki-67 alone could not completely and powerfully determine the novel TNBC category in previous studies. However, until the guidelines clarify the indicators for the further staging of TNBC, could Ki-67, a widely used and easily detectable immunohistochemical indicator in clinical practice, be given priority? This was the original intention of our study.

    Conclusion

    In conclusion, early TNBC patients may be further subcategorized according to the Ki-67 level at 45%, which is associated with a poorer prognosis, particularly in the N0 group. Lymph node staging remains a strong prognostic factor for prognosis. Additionally, NACT and radiotherapy can improve the prognosis for early TNBC patients. Based on the results of this study, we believe that Ki-67 can be considered a reference indicator for TNBC subgroup analysis because of its wide clinical application and its effect on prognosis. Therefore, there is still a need for prospective studies with large sample sizes, specialized assay technicians, and standard operating procedures to explore the optimal detection procedure for Ki-67. Overall, when combined, Ki-67 expression and clinicopathological factors may help predict the prognosis of TNBC and could be used as a guide when recommending individual treatment plans. We still need a large number of prospective and retrospective clinical studies to validate this. Therefore, future studies should focus on identifying efficient biomarkers to direct treatment and prognosis.

    Summary points
    • This research aimed to determine an optimal cutoff value and investigate the prognostic predictive function of Ki-67, which is a proliferation-related biomarker used in luminal breast cancer.

    • We retrospectively enrolled 1146 patients diagnosed with stage I–II triple-negative breast cancer, between Jan 2013 and Dec 2020, at the Sun Yat-sen University Cancer Center.

    • The best cutoff point for Ki-67 was 45%, which was found in the X-tile.

    • For the 1146 patients, we classified Ki-67 >45% as the high group and Ki-67 ≤ 45% as the low group.

    • Lymph node stage, neoadjuvant chemotherapy, and postoperative radiotherapy were independent predictive variables of prognosis.

    • Triple-negative breast cancer may be further subcategorized according to the Ki-67 level >45%, which is associated with a poorer prognosis, particularly in the lymph node-negative group.

    • Neoadjuvant chemotherapy and postoperative radiotherapy can improve the prognosis.

    • Therefore, there is still a need for prospective studies with large sample sizes, specialized assay technicians, and standard operating procedures to explore the optimal detection procedure for Ki-67.

    Supplementary data

    To view the supplementary data that accompany this paper please visit the journal website at: www.future-science.com/doi/suppl/10.4155/fsoa-2023-0129

    Author contributions

    The planning and coordination of the study were performed by F Li and Y Wang. X Zhou and Y Wang performed the data analysis and drafted the manuscript. W Hu and Y Du contributed to the writing of the manuscript and data analysis. For significant intellectual content, Y Wang J Sun and F Li revised the manuscript and offered final approval and revision of the work. The final manuscript has been read and approved by all authors.

    Acknowledgments

    We gratefully acknowledge all the staff members of the Department of Pathology, SYSUCC, Guangzhou, China, for their help and cooperation. This work was supported by the SYSUCC Single Disease Database, YIDU CLOUD Intelligent Follow-up Platform, and the Research Data Deposit platform, which provided data support and convenience for our research.

    Financial disclosure

    The authors have no financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

    Competing interests disclosure

    The authors have no competing interests or relevant affiliations with any organization or entity with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

    Writing disclosure

    No writing assistance was utilized in the production of this manuscript.

    Ethical conduct of research

    The SYSUCC Ethics Committee approved this project (Ethical Consent Letter No.: B2022-638-01). As per the requirements of our Ethics Committee, patients in retrospective clinical studies did not need to sign informed consent. We state that we have followed the principles outlined in the Declaration of Helsinki.

    Open access

    This work is licensed under the Creative Commons Attribution 4.0 License. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Harbeck N, Gnant M. Breast cancer. Lancet 389(10074), 1134–1150 (2017).
    • 2. Gelmon K, Dent R, Mackey JR, Laing K, McLeod D, Verma S. Targeting triple-negative breast cancer: optimising therapeutic outcomes. Ann. Oncol. 23(9), 2223–2234 (2012).
    • 3. Blows FM, Driver KE, Schmidt MK et al. Subtyping of breast cancer by immunohistochemistry to investigate a relationship between subtype and short and long term survival: a collaborative analysis of data for 10,159 cases from 12 studies. PLoS Med. 7, e1000279 (2010).
    • 4. Lehmann BD, Bauer JA, Chen X et al. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J. Clin. Invest. 121(7), 2750–2767 (2011).
    • 5. Sun X, Kaufman PD. Ki-67: more than a proliferation marker. Chromosoma. 127(2), 175–186 (2018).
    • 6. Arafah MA, Ouban A, Ameer OZ, Quek KJ. KI-67 LI expression in triple-negative breast cancer patients and its significance. Breast Cancer (Auckl). 15, 11782234211016977 (2021).
    • 7. Wu Q, Ma G, Deng Y et al. Prognostic value of Ki-67 in patients with resected triple-negative breast cancer: a meta-analysis. Front Oncol. 9, 1068 (2019). •• This study points out that the cutoff of ki-67 ≥40% is associated with a greater risk of recurrence and death compared with lower expression rates, despite the Ki-67 threshold with the greatest prognostic significance is as yet unknown.
    • 8. Goldhirsch A, Winer EP, Coates AS et al. Personalizing the treatment of women with early breast cancer: highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2013. Ann. Oncol. 24(9), 2206–2223 (2013).
    • 9. Penault-Llorca F, Radosevic-Robin N. Ki67 assessment in breast cancer: an update. Pathology 49(2), 166–171 (2017).
    • 10. Focke CM, Bürger H, van Diest PJ et al. Interlaboratory variability of Ki67 staining in breast cancer. Eur. J. Cancer 84, 219–227 (2017).
    • 11. Husni Cangara M, Miskad UA, Masadah R, Nelwan BJ, Wahid S. Gata-3 and KI-67 expression in correlation with molecular subtypes of breast cancer. Breast Dis. 40(Suppl. 1), S27–S31 (2021).
    • 12. Wang W, Wu J, Zhang P et al. Prognostic and predictive value of Ki-67 in triple-negative breast cancer. Oncotarget. 7(21), 31079–31087 (2016). •• This study is very similar to ours, while the exact methodology and the final results are slightly different. Patients in the high Ki-67 group seem to benefit more from treatment with carboplatin, but this needs to be further verified.
    • 13. Zhu X, Chen L, Huang B et al. The prognostic and predictive potential of Ki-67 in triple-negative breast cancer. Sci Rep. 10(1), 225 (2020). •• This study is also similar to ours, yet the exact methodology and the final results are slightly different. Ki-67 cutoff at 30% had the early independent prognostic and predictive potential for OS and DFS in TNBCs, and Ki-67 > 30% was significantly associated with a worse prognosis, especially for stage I patients.
    • 14. Pan Y, Yuan Y, Liu G, Wei Y. P53 and Ki-67 as prognostic markers in triple-negative breast cancer patients. PLoS ONE 12(2), e0172324 (2017).
    • 15. Yerushalmi R, Woods R, Ravdin PM, Hayes MM, Gelmon KA. Ki67 in breast cancer: prognostic and predictive potential. Lancet Oncol. 11(2), 174–183 (2010).
    • 16. Aleskandarany MA, Green AR, Benhasouna AA et al. Prognostic value of proliferation assay in the luminal, HER2-positive, and triple-negative biologic classes of breast cancer. Breast Cancer Res. 14(1), R3 (2012).
    • 17. Munzone E, Botteri E, Sciandivasci A et al. Prognostic value of Ki-67 labeling index in patients with node-negative, triple-negative breast cancer. Breast Cancer Res. Treat. 134(1), 277–282 (2012).
    • 18. Mrklić I, Ćapkun V, Pogorelić Z, Tomić S. Prognostic value of Ki-67 proliferating index in triple negative breast carcinomas. Pathol. Res. Pract. 209(5), 296–301 (2013).
    • 19. Hammond ME, Hayes DF, Dowsett M et al. American Society of Clinical Oncology/College Of American Pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer. J. Clin. Oncol. 28(16), 2784–2795 (2010).
    • 20. Wolff AC, Hammond MEH, Allison KH et al. Human epidermal growth factor receptor 2 testing in breast cancer: american society of clinical oncology/college of american pathologists clinical practice guideline focused update. J. Clin. Oncol. 36(20), 2105–2122 (2018).
    • 21. Nielsen TO, Leung SCY, Rimm DL et al. Assessment of Ki67 in breast cancer: updated recommendations from the international ki67 in breast cancer working group. J. Natl Cancer Inst. 113(7), 808–819 (2021).
    • 22. Camp RL, Dolled-Filhart M, Rimm DL. X-tile: a new bio-informatics tool for biomarker assessment and outcome-based cut-point optimization. Clin Cancer Res. 10(21), 7252–7259 (2004).
    • 23. Mighri N, Mejri N, Boujemaa M et al. Association between epidemiological and clinico-pathological features of breast cancer with prognosis, family history, Ki-67 proliferation index and survival in Tunisian breast cancer patients. PLoS One. 17(9), e0269732 (2022).
    • 24. Zenzola V, Cabezas-Quintario MA, Arguelles M et al. Prognostic value of Ki-67 according to age in patients with triple-negative breast cancer. Clin Transl Oncol. 20(11), 1448–1454 (2018).
    • 25. Setiawan K, Suryawisesa IB, Widiana IK, Sudarsa IW. Does a 40% cutoff point for Ki-67 expression have a role in identifying the development of distant metastasis within 2 years in locally advanced triple negative breast cancer patients? Eur J Breast Health. 19(4), 274–278 (2023).
    • 26. Goetz MP, Kalari KR, Suman VJ et al. Tumor sequencing and patient-derived xenografts in the neoadjuvant treatment of breast cancer. J. Natl Cancer Inst. 109(7), djw306 (2017).
    • 27. Wang RX, Chen S, Jin X, Shao ZM. Value of Ki-67 expression in triple-negative breast cancer before and after neoadjuvant chemotherapy with weekly paclitaxel plus carboplatin. Sci Rep. 6, 30091 (2016).
    • 28. Reis-Filho JS, Pusztai L. Gene expression profiling in breast cancer: classification, prognostication, and prediction. Lancet 378(9805), 1812–1823 (2011).
    • 29. Burstein MD, Tsimelzon A, Poage GM et al. Comprehensive genomic analysis identifies novel subtypes and targets of triple-negative breast cancer. Clin Cancer Res. 21(7), 1688–1698 (2015).
    • 30. Jiang YZ, Ma D, Suo C et al. Genomic and transcriptomic landscape of triple-negative breast cancers: subtypes and treatment strategies. Cancer Cell. 35(3), 428–440.e5 (2019).
    • 31. Zagorac I, Lončar B, Dmitrović B, Kralik K, Kovačević A. Correlation of folate receptor alpha expression with clinicopathological parameters and outcome in triple negative breast cancer. Ann Diagn Pathol. 48, 151596 (2020).
    • 32. Shetty J, Rao C. Expression of E cadherin and Ki 67: emerging prognostic markers in triple-negative breast cancer. Indian J Surg Oncol. 10(2), 377–381 (2019).
    • 33. Vihervuori H, Korpinen K, Autere TA, Repo H, Talvinen K, Kronqvist P. Varying outcomes of triple-negative breast cancer in different age groups-prognostic value of clinical features and proliferation. Breast Cancer Res. Treat. 196(3), 471–482 (2022). • This study proposes that increased proliferation, as evaluated using Ki-67 or geminin immunohistochemistry, showed potential in detecting survival differences in subgroups of TNBC.
    • 34. Nishimura R, Osako T, Okumura Y et al. Triple negative breast cancer: an analysis of the subtypes and the effects of menopausal status on invasive breast cancer. J Clin Med. 11(9), 2331 (2022).
    • 35. Urru SAM, Gallus S, Bosetti C et al. Clinical and pathological factors influencing survival in a large cohort of triple-negative breast cancer patients. BMC Cancer. 18(1), 56 (2018).
    • 36. Gluz O, Nitz U, Liedtke C et al. Comparison of neoadjuvant nab-paclitaxel + carboplatin vs nab-paclitaxel + gemcitabine in triple-negative breast cancer: randomized WSG-ADAPT-TN trial results. J. Natl Cancer Inst. 110(6), 628–637 (2018).
    • 37. Zhang H, Wang Z, Liu W, Wang P, Zhang X. Breast-conserving surgery in triple-negative breast cancer: a retrospective cohort study. Evid Based Complement Alternat Med. 5431563 (2023).
    • 38. van den Ende NS, Nguyen AH, Jager A, Kok M, Debets R, van Deurzen CHM. Triple-negative breast cancer and predictive markers of response to neoadjuvant chemotherapy: a systematic review. Int J Mol Sci. 24(3), 2969 (2023).
    • 39. Toss A, Venturelli M, Civallero M et al. Predictive factors for relapse in triple-negative breast cancer patients without pathological complete response after neoadjuvant chemotherapy. Front Oncol. 12, 1016295 (2022).
    • 40. Gamucci T, Pizzuti L, Sperduti I et al. Neoadjuvant chemotherapy in triple-negative breast cancer: a multicentric retrospective observational study in real-life setting. J. Cell. Physiol. 233(3), 2313–2323 (2018).
    • 41. Zhu M, Yu Y, Shao X, Zhu L, Wang L. Predictors of response and survival outcomes of triple negative breast cancer receiving neoadjuvant chemotherapy. Chemotherapy 65(3–4), 101–109 (2020).
    • 42. Riaz N, Idress R, Habib S, Lalani EN. Lack of androgen receptor expression selects for basal-like phenotype and is a predictor of poor clinical outcome in non-metastatic triple negative breast cancer. Front Oncol. 10, 1083 (2020).
    • 43. Mohammed AA, Elsayed FM, Algazar M, Rashed HE, Anter AH. Neoadjuvant chemotherapy in triple negative breast cancer: correlation between androgen receptor expression and pathological response. Asian Pac J Cancer Prev. 21(2), 563–568 (2020).
    • 44. Naimi A, Soltan M, Amjadi E, Goli P, Kefayat A. Androgen receptor expression and its correlation with clinicopathological parameters in iranian patients with triple negative breast cancer. Iran J Pathol. 15(3), 239–244 (2020).
    • 45. Astvatsaturyan K, Yue Y, Walts AE, Bose S. Androgen receptor positive triple negative breast cancer: clinicopathologic, prognostic, and predictive features. PLoS One. 13(6), e0197827 (2018).
    • 46. Zhang G, Shi Z, Liu L et al. The prognostic relevance of p53 and Ki-67 to chemotherapy sensitivity and prognosis in triple-negative breast cancer. Transl Cancer Res. 10(2), 1082–1087 (2021).
    • 47. Luo Y, Huang W, Zhang H, Liu G. Prognostic significance of CD117 expression and TP53 missense mutations in triple-negative breast cancer. Oncol Lett. 15(5), 6161–6170 (2018).
    • 48. Park MH, Kwon SY, Choi JE, Gong G, Bae YK. Intratumoral CD103-positive tumour-infiltrating lymphocytes are associated with favourable prognosis in patients with triple-negative breast cancer. Histopathology 77(4), 560–569 (2020).
    • 49. Motamedi B, Rafiee-Pour HA, Khosravi MR et al. Prolactin receptor expression as a novel prognostic biomarker for triple negative breast cancer patients. Ann Diagn Pathol. 46, 151507 (2020). • This study proposes a new receptor that is the prolactin receptor (PRLR). PRLR is a novel emerging prognostic biomarker in different cancers, especially in breast cancer, which plays a crucial role in TNBC and has to be considered as an independent prognostic biomarker for TNBC patients.
    • 50. Yoshikawa K, Ishida M, Yanai H, Tsuta K, Sekimoto M, Sugie T. Adipophilin expression is an independent marker for poor prognosis of patients with triple-negative breast cancer: an immunohistochemical study. PLoS One. 15(11), e0242563 (2020).
    • 51. Badowska-Kozakiewicz AM, Budzik MP, Liszcz A et al. Clinicopathological factors associated with novel prognostic markers for patients with triple negative breast cancer. Arch Med Sci. 15(6), 1433–1442 (2019).
    • 52. Ozretic P, Alvir I, Sarcevic B et al. Apoptosis regulator Bcl-2 is an independent prognostic marker for worse overall survival in triple-negative breast cancer patients. Int J Biol Markers. 33(1), 109–115 (2018).
    • 53. Loi S, Michiels S, Salgado R et al. Tumor infiltrating lymphocytes are prognostic in triple negative breast cancer and predictive for trastuzumab benefit in early breast cancer: results from the FinHER trial. Ann. Oncol. 25(8), 1544–1550 (2014).
    • 54. Adams S, Gray RJ, Demaria S et al. Prognostic value of tumor-infiltrating lymphocytes in triple-negative breast cancers from two phase III randomized adjuvant breast cancer trials: ECOG 2197 and ECOG 1199. J. Clin. Oncol. 32(27), 2959–2966 (2014).
    • 55. Pruneri G, Vingiani A, Bagnardi V et al. Clinical validity of tumor-infiltrating lymphocytes analysis in patients with triple-negative breast cancer. Ann. Oncol. 27(2), 249–256 (2016).
    • 56. Yu H, Meng X, Chen H et al. Correlation between mammographic radiomics features and the level of tumor-infiltrating lymphocytes in patients with triple-negative breast cancer. Front Oncol. 10, 412 (2020).
    • 57. Zhang L, Wang XI, Zhang S. Tumor-infiltrating lymphocyte volume is a better predictor of neoadjuvant therapy response and overall survival in triple-negative invasive breast cancer. Hum. Pathol. 80, 47–54 (2018).
    • 58. Abuhadra N, Sun R, Yam C et al. Predictive roles of baseline stromal tumor-infiltrating lymphocytes and ki-67 in pathologic complete response in an early-stage triple-negative breast cancer prospective trial. Cancers (Basel). 15(13), 3275 (2023).
    • 59. Nottegar A, Veronese N, Senthil M et al. Extra-nodal extension of sentinel lymph node metastasis is a marker of poor prognosis in breast cancer patients: a systematic review and an exploratory meta-analysis. Eur. J. Surg. Oncol. 42(7), 919–925 (2016).
    • 60. Aziz S, Wik E, Knutsvik G et al. Extra-nodal extension is a significant prognostic factor in lymph node positive breast cancer. PLoS ONE 12(2), e0171853 (2017).
    • 61. Gujam FJ, Going JJ, Edwards J, Mohammed ZM, McMillan DC. The role of lymphatic and blood vessel invasion in predicting survival and methods of detection in patients with primary operable breast cancer. Crit. Rev. Oncol. Hematol. 89(2), 231–241 (2014).
    • 62. Zhang S, Zhang D, Yi S et al. The relationship of lymphatic vessel density, lymphovascular invasion, and lymph node metastasis in breast cancer: a systematic review and meta-analysis. Oncotarget. 8(2), 2863–2873 (2017).
    • 63. Chen L, Zhang J, Chen J et al. Post-operative radiotherapy is beneficial for T1/T2 triple negative breast cancer patients with four or more positive lymph nodes. Oncotarget. 8(26), 42917–42925 (2017).
    • 64. Abdulkarim BS, Cuartero J, Hanson J, Deschênes J, Lesniak D, Sabri S. Increased risk of locoregional recurrence for women with T1–2N0 triple-negative breast cancer treated with modified radical mastectomy without adjuvant radiation therapy compared with breast-conserving therapy. J. Clin. Oncol. 29(21), 2852–2858 (2011).
    • 65. He MY, Rancoule C, Rehailia-Blanchard A et al. Radiotherapy in triple-negative breast cancer: current situation and upcoming strategies. Crit. Rev. Oncol. Hematol. 131, 96–101 (2018).