Skip to main content
Erschienen in: BMC Women's Health 1/2024

Open Access 01.12.2024 | Research

Impact of residual microcalcifcations on prognosis after neoadjuvant chemotherapy in breast cancer patients

verfasst von: Eun Young Kim, Kwan Ho Lee, Ji-Sup Yun, Yong Lai Park, Chan Heun Park, Sung Yoon Jang, Jai Min Ryu, Se Kyung Lee, Byung-Joo Chae, Jeong Eon Lee, Seok Won Kim, Seok Jin Nam, Jong Han Yu

Erschienen in: BMC Women's Health | Ausgabe 1/2024

Abstract

Background

Residual microcalcifications after neoadjuvant chemotherapy (NAC) are challenging for deciding extent of surgery and questionable for impact on prognosis. We investigated changes in the extent and patterns of microcalcifications before and after NAC and correlated them with pathologic response. We also compared prognosis of patients depending on presence of residual microcalcifications after NAC.

Methods

A total of 323 patients with invasive breast carcinoma treated with neoadjuvant chemotherapy at Kangbuk Samsung Hospital and Samsung Medical center from March 2015 to September 2018 were included. Patients were divided into four groups according to pathologic response and residual microcalcifications. Non-pCRw/mic group was defined as breast non-pCR with residual microcalcifications. Non-pCRw/o mic group was breast non-pCR without residual microcalcifications. pCRw/mic group was breast pCR with residual microcalcifications. pCRw/o mic group was breast pCR without residual microcalcifications. The first aim of this study is to investigate changes in the extent and patterns of microcalcifications before and after NAC and to correlate them with pathologic response. The second aim is to evaluate oncologic outcomes of residual microcalcifications according to pathologic response after NAC.

Results

There were no statistical differences in the extent, morphology, and distribution of microcalcifications according to pathologic response and subtype after NAC (all p > 0.05). With a median follow-up time of 71 months, compared to pCRw/o mic group, the hazard ratios (95% confidence intervals) for regional recurrence were 5.190 (1.160–23.190) in non-pCRw/mic group and 5.970 (1.840–19.380) in non-pCRw/o mic group. Compared to pCRw/o mic group, the hazard ratios (95% CI) for distant metastasis were 8.520 (2.130–34.090) in non-pCRw/mic group, 9.120 (2.850–29.200) in non-pCRw/o mic group. Compared to pCRw/o mic, the hazard ratio (95% CI) for distant metastasis in pCRw/mic group was 2.240 (0.230–21.500) without statistical significance (p = 0.486).

Conclusions

Regardless of residual microcalcifications, patients who achieved pCR showed favorable long term outcome compared to non-pCR group.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12905-024-02973-9.

Publisher’s Note

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

Introduction

Neoadjuvant chemotherapy (NAC) is the standard treatment of locally advanced breast cancer. The indication for NAC has been extended to human epidermal growth factor receptor 2 (HER2)-positive and triple-negative breast cancer (TNBC) with clinical T stage ≥ 2 or N stage ≥ 1 due to developments in molecular pathology and targeted therapy. Although similar long-term outcomes were reported compared to adjuvant chemotherapy, NAC has many advantages including increased likelihood of breast conservation and prediction of sensitivity to chemotherapeutic agents. Pathologic complete response (pCR) is associated with better disease-free and overall survival [1, 2]. Although imaging modalities such as mammography, breast sonography, and magnetic resonance imaging (MRI) can predict the extent of a tumor, it is difficult to predict response when tumors are accompanied by microcalcifications.
Breast microcalcifications are calcium deposits within the breast tissue and can be classified according to the Breast Imaging Reporting and Data system (BI-RADS) developed by the American College of Radiology [3]. Most microcalcifications are manifestations of pure ductal carcinoma in situ (DCIS) or intraductal portions of invasive carcinomas [4]. Since DCIS rarely responds to systemic therapy and most microcalcifications do not decrease after NAC, partial mastectomy is often contraindicated even with radiological complete response (CR) on breast MRI [5, 6]. Decisions about surgical extent in patients with remaining extensive microcalcifications after NAC are challenging since residual microcalcifications show low correlation with pathologic response [7, 8]. The definition of pCR varies across studies [9]. Whether pCR is defined as ypTis or ypT0, eradication of residual invasive cancer of the breast is associated with excellent survival outcomes. Since most DCIS manifests as microcalcifications, eradication of all residual microcalcifications is questionable, and the impact of residual microcalcifications on long-term outcome remains unknown.
Therefore, the first aim of this study is to investigate changes in the extent and patterns of microcalcifications before and after NAC and to correlate them with pathologic response. The second aim is to evaluate oncologic outcomes of residual microcalcifications according to pathologic response after NAC.

Methods

Patients

A total of 323 patients with invasive breast carcinoma treated with neoadjuvant chemotherapy at Kangbuk Samsung Hospital and Samsung Medical center from March 2015 to September 2018 were included. Detailed information including patient age, tumor size, and clinical stage as well as estrogen receptor (ER), progesterone receptor, and HER2 positivity were extracted from the database. We excluded patients with stage IV breast cancer, without suspicious microcalcifications on mammography before and after NAC, or without available imaging results (mammography and/or MRI). This retrospective study was approved by the Institutional Review Board of Kangbuk Samsung Hospital (KBSMC 2018–09-033), and the requirement for individual informed consent was waived owing to the retrospective nature of the study. All methods were performed in accordance with the relevant guidelines.

Assessment of mammography and MRI

All microcalcifications on mammography acquired pre- and post-NAC were reassessed by dedicated radiologists with 6–24 years of experience in breast imaging according to the BI-RADS lexicon [10]. Extent, morphology (amorphous, coarse heterogeneous, fine pleomorphic, fine linear or fine-linear branching), distribution (regional, grouped, linear, segmental, diffuse), and location of microcalcifications were evaluated. The categories of breast density were as follows: a. the breasts are almost entirely fatty. b. there are scattered areas of fibroglandular density. c. the breasts are heterogeneously dense, which may obscure small masses. d. the breasts are extremely dense, which lowers the sensitivity of mammography [10].
The extent of microcalcifications was measured in millimeters of the greatest dimensions in the craniocaudal (CC) and mediolateral oblique views. Multifocality was defined as multiple microcalcifications located in the same quadrant, whereas multicentricity was defined as microcalcifications located in more than one quadrant [11]. In cases of multifocal or multicentric microcalcifications, all lesions were individually measured, and the overall extent of disease, including all foci, was measured and used in analysis. “Decreased microcalcifications” was defined as a greater than 25% decrease in extent (not amount nor density) after NAC. “Increased microcalcifications” was defined as a greater than 25% increase in extent (not amount nor density) after NAC. "No change" referred to microcalcifications that neither increased nor decreased. “New” refers to suspicious microcalcifications newly developed after NAC [7]. Tumor response was assessed on MRI after completion of NAC, and the size of residual disease was categorized into four categories according to the response evaluation criteria in solid tumors (RECIST) and the Leeds modified response evaluation criteria in solid tumors, considering tumor size and enhancement characteristics [12, 13]. Radiological CR was defined as no residual enhancement within the original tumor bed or any residual mass. Partial response (PR) was defined as decrease in tumor diameter greater than 30% or improvement in the enhancement curve. Progressive disease (PD) was defined as an increase in the diameter of the tumor greater than 20%. Stable disease was defined as neither PR nor PD.

Histopathologic analysis

Surgery was performed after completion of NAC. Specimens were sent to the mammography unit. Standard compression radiographs were obtained. Radiologists immediately confirmed the presence of microcalcifications inside the specimen. It was surgeon’s discretion to perform further resection of residual microcalcifications, in case specimen did not contain all microcalcifications. Board-certified pathologists with 10–24 years of experience reviewed breast specimens to estimate histopathologic tumor response to NAC. Breast pCR was defined as the absence of residual invasive carcinoma in resected breast specimens. Pathologic tumor response was categorized according to RECIST criteria [12, 14]. In cases of non-pCR, the largest histopathologic diameters of residual tumors were measured. Immunohistochemical staining was performed, and subtypes were determined according to ER, progesterone receptor, and HER2 status. Hormone receptor (HR) positivity was defined as ER and/or progesterone receptor positive.

Survival outcome analysis

Mammographic findings of microcalcifications were reassessed on first follow up after surgery (Fig. 1). Patients were classified into four groups according to pathologic response and residual microcalcifications. Non-pCRw/mic (n = 25) was classified as breast non-pCR with residual microcalcifications. Non-pCRw/o mic (n = 196) was classified as breast non-pCR without residual microcalcifications. pCRw/mic (n = 13) was classified as breast pCR with residual microcalcifications. pCRw/o mic (n = 89) was classified as breast pCR without residual microcalcifications.
Recurrence in any quadrant of the ipsilateral breast was defined as local recurrence. Recurrence in the ipsilateral breast, skin, chest wall, axillary, supraclavicular, internal mammary lymph nodes was defined as regional recurrence. Metastases in distant organs such as bone, lung, liver and brain were defined as distant metastases. Local relapse-free survival (LFS), regional relapse-free survival (RFS), and distant metastasis-free survival (DMFS) were calculated from date of breast cancer diagnosis to the respective event. Overall survival (OS) was defined from the date of breast cancer diagnosis to death from any cause. Patients who did not experience recurrence, distant metastasis, or death were censored at the last follow up.

Statistical analysis

Data were summarized as frequencies and percentages. Descriptive data were tabulated. Changes in the extent, morphology, or distribution of microcalcifications after NAC were correlated using McNemar’s test. Changes in mammographic characteristics were correlated with pathologic response using Pearson's Chi-square test. Differences in tumor characteristics according to subtype were compared using Pearson's Chi-square test.
Univariate and multivariate logistic regression analyses were performed to identify variables predictive of breast pCR. The actuarial rates of recurrence and survival were calculated according to the Kaplan–Meier method, and comparisons were performed using the log-rank test. Univariate and multivariate analyses using the Cox proportional hazards regression model were used to determine predictive factors of recurrence and survival. Any missing data on mammographic characteristics were excluded from statistical analysis. Statistical analyses were performed using PASW software version 18.0 (IBM, Armonk, NY, USA). P-values < 0.05 were considered statistically significant.

Results

A total of 323 patients was diagnosed with invasive breast carcinoma and underwent NAC between March 2015 and September 2018. The clinicopathologic characteristics of the study population before and after NAC are listed in Table 1. The median age of the study population was 47 years (range: 40–54 years). The median tumor size on MRI before NAC was 47.0 mm (interquartile range: 30.0–67.0 mm). Invasive ductal carcinoma, not otherwise specified, was the most common histologic type before NAC (n = 301, 93.2%). Other types included invasive lobular carcinoma (n = 10, 3.1%), mucinous (n = 2, 0.6%), and micropapillary (n = 1, 0.3%). The HR/HER2+ subtype was noted in 31.3% (n = 101) of patients, and HR/HER2 was observed in 15.8% of patients (n = 51). Breast pCR (ypT0 and/or ypTis) was observed in 31.6% (n = 102) of patients. In 3.7% (n = 12) of patients, no residual in situ or invasive lesions (only ypT0) were seen.
Table 1
Clinicopathologic characteristics of the study population before and after neoadjuvant chemotherapy
Characteristics
Total (n = 323)
Non-pCRw/ mic (n = 25)
Non-pCRw/o mic (n = 196)
pCRw/ mic (n = 13)
pCRw/o mic (n = 89)
P value
Age, years
47 (40–54)
45 (37–55)
47 (39–54)
48 (39–56)
48 (41–52)
0.907
Pre-NAC histologic type
     
0.686
 IDC
301 (93.2)
24 (96.0)
181 (92.3)
12 (92.3)
84 (94.4)
 
 ILC
10 (3.1)
1 (4.0)
7 (3.6)
0 (0)
2 (2.2)
 
 Mucinous
2 (0.6)
0 (0)
1 (0.5)
1 (7.7)
0 (0)
 
 Micropapillary
1 (0.3)
0 (0)
1 (0.5)
0 (0)
0 (0)
 
 Other
9 (2.8)
0 (0)
6 (3.1)
0 (0)
3 (3.4)
 
Pre-NAC IHC subtype
     
 < 0.001
 HR + /HER2-
90 (27.9)
6 (24.0)
74 (37.8)
1 (7.7)
9 (10.1)
 
 HR + /HER2 + 
81 (25.1)
5 (20.0)
43 (21.9)
3 (23.1)
30 (33.7)
 
 HR-/HER2 + 
101 (31.3)
10 (40.0)
48 (24.5)
8 (61.5)
35 (39.3)
 
 HR-/HER2-
51 (15.8)
4 (16.0)
31 (15.8)
1 (7.7)
15 (16.9)
 
Pre-NAC tumor size at MRI, mm
47 (30–67)
60 (33–72)
48 (32–67)
50 (27–55)
40 (30–60)
0.1
Pre-NAC Ki-67 ≥ 14%
278 (87.7)
22 (88.0)
161 (84.3)
11 (84.6)
84 (95.5)
0.069
NAC regimen
     
0.001
 AC followed by T
118 (36.6)
6 (24.0)
88 (45.1)
0 (0)
24 (27)
 
 AC followed by TH
133 (41.3)
14 (56.0)
65 (33.3)
9 (69.2)
45 (50.6)
 
 TCHP
27 (8.4)
1 (4.0)
11 (5.6)
1 (7.7)
14 (15.7)
 
 AT
33 (10.2)
3 (12.0)
23 (11.8)
2 (15.4)
5 (5.6)
 
 Other
11 (3.4)
1 (4.0)
8 (4.1)
1 (7.7)
1 (1.1)
 
Radiologic response on MRI
     
 < 0.001
 CR
74 (22.9)
2 (8.0)
22 (11.2)
9 (69.2)
41 (46.1)
 
 Non-CR
249 (77.1)
23 (92.0)
174 (88.8)
4 (30.8)
48 (53.9)
 
Type of surgery
     
 < 0.001
 Total mastectomy
170 (52.6)
0 (0)
134 (68.4)
0 (0)
36 (40.4)
 
 Partial mastectomy
153 (47.4)
25 (100.0)
62 (31.6)
13 (100.0)
53 (59.6)
 
 Post-NAC tumor size at MRI, mm
18 (6–36)
16.50 (9.75–31.75)
22 (12.25–42.5)
0 (0–13)
7 (0–25)
 < 0.001
Post-NAC histologic type
     
 < 0.001
 IDC
145 (45.2)
13 (52.0)
106 (54.1)
4 (30.8)
22 (25.8)
 
 DCIS
65 (20.1)
0 (0)
2 (1.0)
7 (53.8)
56 (62.9)
 
 ILC
99 (30.7)
12 (48.0)
87 (44.4)
0 (0)
0 (0)
 
 LCIS
1 (0.3)
0
0
0
1 (1.1)
 
 Other
1 (0.3)
0 (0)
1 (0.5)
0 (0)
0 (0)
 
none (only ypT0)
12 (3.7)
0 (0)
0 (0)
2 (15.4)
10 (11.2)
 
Breast pCR (ypT0 and/or ypTis)
102 (31.6)
0 (0)
0 (0)
13 (100)
89 (100)
 < 0.001
Post-NAC tumor size (invasive), mm
16 (4.50–37)
12 (3–18)
21 (9–45)
0 (0–0)
0 (0–0)
 < 0.001
Post-NAC tumor size (in situ), mm
24.50 (7–50)
35 (9.5–50)
30 (15–54.25)
20 (7.75–36)
10 (2–30)
0.002
Post-NAC extensive intraductal component
69 (28.2)
6 (26.1)
63 (32.3)
0 (0)
0 (0)
0.001
Post-NAC lymphovascular invasion
101 (34.1)
7 (28.0)
89 (45.4)
0 (0)
5 (7.5)
 < 0.001
Post-NAC Ki-67 ≥ 14%
108 (50)
12 (63.2)
74 (48.7)
4 (80)
18 (45)
0.305
Follow-up (months)
74.6 (68.85–86.44)
72.7 (69.39–78.26)
75.2 (68.29–84.73)
71.8 (68.99–86.37)
74.7 (68.93–87.19)
0.816
Local recurrence
39 (12.1)
5 (20.0)
31 (15.8)
0 (0)
3 (3.4)
0.007
Regional recurrence
43 (13.3)
4 (16.0)
36 (18.4)
0 (0)
3 (3.4)
0.003
Distant metastasis
62 (19.2)
6 (24.0)
52 (26.5)
1 (7.7)
3 (3.4)
 < 0.001
Death
38 (11.8)
4 (16.0)
31 (15.8)
0 (0)
3 (3.4)
0.010
Data are presented as median (interquartile range) or number (%)
Abbreviations: NAC Neoadjuvant chemotherapy, IDC Invasive ductal carcinoma, ILC Invasive lobular carcinoma, DCIS Ductal carcinoma in situ, HR Hormone receptor, HER2 Human epidermal growth factor receptor 2, AC Adriamycin/cyclophosphamide, CR Complete response, T Docetaxel, TH Docetaxel/trastuzumab, TCHP Docetaxel/carboplatin/trastuzumab/pertuzumab, AT Adriamycin/docetaxel, LCIS Lobular carcinoma in situ, pCR Pathologic complete response
Mammographic findings before and after NAC are described in Supplementary Table 1. Multifocal/multicentric microcalcifications were noted in 110 patients (34.1%) before NAC. Our results showed that 126 (39.3%) and 148 (45.7%) of patients had pleomorphic microcalcifications before and after NAC, respectively. In addition, 69 (21.3%) and 74 (22.9%) patients showed fine linear/linear branching microcalcifications before and after NAC, respectively. After NAC, there was no change in extent of microcalcifications in 221 (68.4%) patients. Decrease of extent was noted in 74 (23.0%) patients, while increase was evident in 18 (5.6%) patients. The median extent of microcalcifications was 41.0 [interquartile range (IQR) 25.0–66.5] mm before NAC and 43.0 (IQR 22.0–70.0) mm after NAC. The majority of microcalcifications did not present changes in morphology (72.5%) after NAC.
Changes in mammographic and MRI characteristics after NAC according to pathologic response are described in Supplementary Table 2. Changes in extent, morphology of microcalcifications, and mammographic density after NAC did not differ significantly according to pathologic response (all p > 0.05). Changes in sizes of tumors measured on MRI after NAC were significantly different between the pCR and non-pCR groups (p < 0.001). Fifty patients (49.0%) in the pCR group showed radiological CR on MRI after NAC. In contrast, 153 patients (69.2%) in the non-pCR group showed PR and 21 patients (9.5%) in the non-pCR group exhibited SD on MRI. This implies that changes in size of tumor on MRI was correlated with pathologic response, however changes in the extent and morphology of microcalcifications after NAC were not correlated with pathologic response.
Changes in mammographic and MRI characteristics after NAC differed according to subtype (Table 2; Figs. 2 and 3). The extent of microcalcifications after NAC was largest in HR HER2+ subtype and smallest in HR HER2 subtype (48.0 mm vs. 23.5 mm, p = 003). There were no differences in changes in extent of microcalcifications according to subtype (p = 0.147), though there was a statistically significant difference in the change in morphology of microcalcifications after NAC according to subtype (p < 0.001). About 22–24% of the HR+ HER2+ and HR HER2+ subtypes showed changes in morphology of microcalcifications. In addition, the pathologic response differed according to subtype (p < 0.001). HR HER2+ subtype showed the highest pCR rate (42.6%), and the HR+ HER2 subtype showed the lowest pCR rate (11.1%).
Table 2
Changes in mammographic and MRI characteristics after NAC according to subtype
 
HR+ HER2 (n = 90)
HR HER2 (n = 51)
HR+ HER2+ (n = 81)
HR HER2+ (n = 101)
P value
Post-NAC tumor size (invasive), mm
25.0 (12.0, 50.0)
19.5 (6.3, 33.5)
13.0 (3.3, 25.0)
14.0 (3.3, 25.0)
0.004
Post-NAC tumor size (in situ), mm
30.0 (11.5, 60.0)
25.0 (7.0, 40.0)
20.0 (5.3, 46.5)
25.0 (6.0, 43.5)
0.346
Post-NAC extent of microcalcifications, mm
44.0 (27.0, 71.0)
23.5 (13.3, 35.8)
41.0 (20.5, 70.0)
48.0 (28.8, 70.3)
0.003
Changes in extent of microcalcifications
    
0.147
 No change
60 (66.7)
31 (60.8)
50 (61.7)
67 (66.3)
 
 Decreased
18 (20.0)
8 (15.7)
19 (23.5)
25 (24.8)
 
 Increased
5 (5.6)
2 (3.9)
6 (7.4)
4 (4.0)
 
 New
2 (2.2)
2 (3.9)
1 (1.2)
4 (4.0)
 
 Not specified
5 (5.6)
8 (15.7)
5 (6.2)
1 (1.0)
 
Change in morphology of microcalcifications
    
 < 0.001
 No change
46 (51.1)
18 (35.3)
35 (43.2)
51 (50.5)
 
 Change
14 (15.6)
1 (2.0)
20 (24.7)
22 (21.8)
 
 Not specified
30 (33.3)
32 (62.7)
26 (32.1)
28 (27.7)
 
Radiologic response of breast cancer on MRI
    
0.073
 CR
11 (12.2)
10 (19.6)
26 (32.1)
27 (26.7)
 
 Non-CR
78 (86.7)
41 (80.4)
54 (66.7)
73 (72.3)
 
 Not specified
1 (1.1)
0 (0.0)
1 (1.2)
1 (1.0)
 
Pathologic response of breast cancer
    
 < 0.001
 pCR
10 (11.1)
16 (31.4)
33 (40.7)
43 (42.6)
 
 Non-pCR
80 (88.9)
35 (68.6)
48 (59.3)
58 (57.4)
 
Data are mean (standard deviation) or number (percentage)
Abbreviations: NAC Neoadjuvant chemotherapy, HR Hormone receptor, HER2 Human epidermal growth factor receptor 2, CR Complete response, pCR Pathologic complete response
Predictive factors of pCR (ypT0 or ypTis) are listed in Supplementary Table 3. The univariate-adjusted odds ratio for pCR was 3.046 (95% confidence interval [CI]: 1.475–6.290) for HER2+ subtype, 5.091 (95% CI: 2.900–8.939) for tumor size < 10 mm on MRI after NAC, 41.667 (95% CI: 5.276–329.047) for radiologic CR on MRI, and 2.824 (95% CI: 1.734–4.598) for partial mastectomy. Only HER2+ subtype remained significant after multivariate analysis. Changes in extent and morphology of microcalcifications were not predictive factors in either univariate or multivariate regression.
Univariate and multivariate analyses were performed to identify variables for LFS, RFS, DMFS, and OS (Tables 3, 4, 5 and 6). HER2+ positivity showed negative correlations with local and regional recurrence, and lymphovascular invasion and Ki-67 ≥ 14% after NAC were positively correlated with local and regional recurrence, as well as DMFS and OS. Other factors including extent of microcalcifications, presence of residual microcalcifciaiton (regardless of pCR) after NAC did not predict recurrence or survival. Since there were only few cases of local, regional recurrences, distant metastasis and death in patients who achieved radiologic CR on MRI, it was difficult to calculate odds ratio for the variable “radiologic response of MRI” in multivariate analysis. We could not provide odds ratio for that variable in Tables 3, 4, 5 and 6.
Table 3
Univariate and multivariate analyses of local recurrence
Characteristics
Univariate
Multivariate
Odds ratio (95% CI)
P value
Odds ratio (95% CI)
P value
Age at diagnosis, years (ref =  < 40)
  ≥ 40
0.712 (0.350–1.530)
0.363
  
Clinical T stage before NAC (ref = cT1)
 cT2-4
1.387 (0.250- 25.810)
0.758
  
Clinical N stage before NAC (ref = cT0)
 cN1-3
1.232 (0.340–7.950)
0.785
  
Lymphovascular invasion (ref = absent)
 Present
6.416 (3.110- 14.090)
 < 0.001
5.950 (2.430–16.130)
 < 0.001
HR + (ref = negative)
 Positive
0.459 (0.220–0.910)
0.028
0.490 (0.170–1.300)
0.159
HER2+(ref = negative)
 Positive
0.462 (0.210- 0.970)
0.046
0.340 (0.130- 0.860)
0.026
Ki-67 before NAC (ref =  < 14%)
  ≥ 14%
1.728 (0.580- 7.430)
0.383
  
Ki-67 after NAC (ref =  < 14%)
  ≥ 14%
4.810 (2.090- 12.500)
 < 0.001
5.010 (1.760- 16.120)
0.004
Radiologic response of MRI (ref = CR)
 Non-CR
6.283 (1.860–39.240)
0.013
N/A
0.988
Extent of surgery (ref = total mastectomy)
 Partial mastectomy
0.661 (0.330–1.300)
0.237
  
Pre-NAC extent of microcalcifications, mm (ref =  < 10)
  ≥ 10
0.500 (0.120–3.440)
0.397
  
Post-NAC extent of microcalcifications, mm (ref =  < 10)
  ≥ 10
0.590 (0.140- 3.990)
0.512
  
Residual microcalcification (ref = absent)
 Present
1.119 (0.364–2.839)
0.827
1.740 (0.410–6.410)
0.423
Abbreviations: CI Confidence interval, HER2 Human epidermal growth factor receptor 2, NAC Neoadjuvant chemotherapy, N/A Not applicable, CR Complete response, PR Partial response, ref Reference, SD Stable disease, PD Progressive disease
Table 4
Univariate and multivariate analyses of regional recurrence
Characteristics
Univariate
Multivariate
Odds ratio (95% CI)
P value
Odds ratio (95% CI)
P value
Age at diagnosis, years (ref =  < 40)
  ≥ 40
0.642 (0.320 -1.320)
0.211
  
Clinical T stage before NAC (ref = cT1)
 cT2-4
1.556 (0.290- 28.910)
0.677
  
Clinical N stage before NAC (ref = cT0)
 cN1-3
1.390 (0.380 -8.950)
0.667
  
Lymphovascular invasion (ref = absent)
 Present
7.758 (3.810- 16.910)
 < 0.001
6.000 (2.510- 15.650)
 < 0.001
HR + (ref = negative)
 Positive
0.606 (0.310- 1.160)
0.131
  
HER2+(ref = negative)
 Positive
0.373 (0.170- 0.770)
0.009
0.280 (0.110- 0.690)
0.007
Ki-67 before NAC (ref =  < 14%)
  ≥ 14%
1.958 (0.660- 8.390)
0.282
  
Ki-67 after NAC (ref =  < 14%)
  ≥ 14%
4.808 (2.180 -11.790)
 < 0.001
7.940 (3.050- 23.400)
 < 0.001
Radiologic response of MRI (ref = CR)
 Non-CR
14.812 (3.140- 264.940)
0.008
N/A
0.988
Extent of surgery (ref = total mastectomy)
 Partial mastectomy
0.490 (0.240- 0.950)
0.039
1.400 (0.510- 3.800)
0.504
Pre-NAC extent of microcalcifications, mm (ref =  < 10)
  ≥ 10
0.545 (0.130- 3.740)
0.458
  
Post-NAC extent of microcalcifications, mm (ref =  < 10)
  ≥ 10
0.619 (0.150- 4.180)
0.550
  
Residual microcalcification (ref = absent)
 Present
0.742 (0.213–1.992)
0.592
0.600 (0.100- 2.880)
0.543
Abbreviations: CI Confidence interval, HER2 Human epidermal growth factor receptor 2, NAC Neoadjuvant chemotherapy, N/A Not applicable, CR Complete response, PR Partial response, ref Reference, SD Stable disease, PD Progressive disease
Table 5
Univariate and multivariate analyses of distant metastasis
Characteristics
Univariate
Multivariate
Odds ratio (95% CI)
P value
Odds ratio (95% CI)
P value
Age at diagnosis, years (ref =  < 40)
  ≥ 40
0.694 (0.380- 1.300)
0.241
  
Clinical T stage before NAC (ref = cT1)
 cT2-4
1.071 (0.270- 7.150)
0.931
  
Clinical N stage before NAC (ref = cT0)
 cN1-3
2.166 (0.600- 13.870)
0.309
  
Lymphovascular invasion (ref = absent)
 Present
6.594 (3.610- 12.450)
 < 0.001
4.590 (2.230- 9.830)
 < 0.001
HR + (ref = negative)
 Positive
0.878 (0.500- 1.530)
0.644
  
HER2+(ref = negative)
 Positive
0.516 (0.270- 0.960)
0.038
0.530 (0.250- 1.110)
0.099
Ki-67 before NAC (ref =  < 14%)
  ≥ 14%
1.357 (0.580- 3.740)
0.515
  
Ki-67 after NAC (ref =  < 14%)
  ≥ 14%
2.606 (1.390- 5.050)
0.004
3.230 (1.520- 7.160)
0.003
Radiologic response of MRI (ref = CR)
 Non-CR
11.429 (3.440- 70.820)
0.001
N/A
0.988
Extent of surgery (ref = total mastectomy)
 Partial mastectomy
0.546 (0.300- 0.960)
0.039
1.400 (0.610- 3.210)
0.426
Pre-NAC extent of microcalcifications, mm (ref =  < 10)
  ≥ 10
0.891 (0.210- 6.050)
0.887
  
Post-NAC extent of microcalcifications, mm (ref =  < 10)
  ≥ 10
1.056 (0.260- 7.050)
0.946
  
Residual microcalcification (ref = absent)
 Present
0.944 (0.366–2.145)
0.897
0.810 (0.200–2.920)
0.755
Abbreviations: CI Confidence interval, HER2 Human epidermal growth factor receptor 2, NAC Neoadjuvant chemotherapy, N/A Not applicable, CR Complete response, PR Partial response, ref Reference, SD Stable disease, PD Progressive disease
Table 6
Univariate and multivariate analyses of death
Characteristics
Univariate
Multivariate
Odds ratio (95% CI)
P value
Odds ratio (95% CI)
P value
Age at diagnosis, years (ref =  < 40)
  ≥ 40
0.788 (0.380- 1.730)
0.534
  
Clinical T stage before NAC (ref = cT1)
 cT2-4
1.345 (0.250- 25.040)
0.780
  
Clinical N stage before NAC (ref = cT0)
 cN1-3
0.727 (0.230- 3.230)
0.625
  
Lymphovascular invasion (ref = absent)
 Present
7.096 (3.380- 16.050)
 < 0.001
5.230 (2.190- 13.620)
 < 0.001
HR + (ref = negative)
 Positive
0.482 (0.230 -0.960)
0.041
0.420 (0.150- 1.110)
0.082
HER2+(ref = negative)
 Positive
0.436 (0.200- 0.910)
0.031
0.320 (0.120- 0.780)
0.015
Ki-67 before NAC (ref =  < 14%)
  ≥ 14%
1.672 (0.560- 7.190)
0.413
  
Ki-67 after NAC (ref =  < 14%)
  ≥ 14%
4.167 (1.870- 10.270)
0.001
4.030 (1.480- 12.090)
0.009
Radiologic response of MRI (ref = CR)
 Non-CR
12.741 (2.680- 228.220)
0.013
N/A
0.988
Extent of surgery (ref = total mastectomy)
 Partial mastectomy
0.613 (0.300- 1.220)
0.169
  
Pre-NAC extent of microcalcifications, mm (ref =  < 10)
  ≥ 10
0.433 (0.100- 2.990)
0.309
  
Post-NAC extent of microcalcifications, mm (ref =  < 10)
  ≥ 10
0.533 (0.130- 3.620)
0.436
  
Residual microcalcification (ref = absent)
 Present
0.869 (0.248–2.351)
0.801
1.670 (0.400–6.010)
0.447
Abbreviations: CI Confidence interval, HER2 Human epidermal growth factor receptor 2, NAC Neoadjuvant chemotherapy, N/A Not applicable, CR Complete response, PR Partial response, ref Reference, SD Stable disease, PD Progressive disease
We evaluated long-term outcomes of residual microcalcifications after NAC according to pathologic response (Fig. 4a-d) (See Supplementary Table 4). The median follow-up period was 74.6 months. Compared to pCRw/o mic, the hazard ratios (95% CI) for regional recurrence in non-pCRw/mic group, non-pCRw/o mic group were 5.190 (1.160–23.190), 5.970 (1.840–19.380), respectively. Compared to pCRw/o mic, the hazard ratios (95% CI) for distant metastasis in non-pCRw/mic group, non-pCRw/o mic group were 8.520 (2.130–34.090), 9.120 (2.850–29.200), respectively. Compared to pCRw/o mic, the hazard ratio (95% CI) for distant metastasis in pCRw/mic group was 2.240 (0.230–21.500) without statistical significance (p = 0.486). We could not perform statistical analysis for LFS, RFS, OS in pCRw/ mic group owing to small number of events. This association was consistently observed in terms of LFS and OS, in which non-pCR were associated with an increased risk regardless of residual microcalcifications.

Discussion

In the present study, we aimed to determine whether residual microcalcifications reflect the chemotherapeutic response of breast cancer and to clarify whether changes in the characteristics of microcalcifications after NAC predict pathologic response and affect long-term outcomes. We demonstrated that extent of residual microcalcifications after NAC were not correlated with extent of residual cancer. Only HER2+ subtype and radiologic CR on MRI were predictors of pCR.
Past studies demonstrated no correlation between changes in microcalcifications and pCR [15, 16] and recommended excision of indeterminate or suspicious-looking microcalcifications after NAC [17]. Yim et al. found that changes in microcalcifications after NAC were correlated with tumor response to NAC [18]. They reported more frequent decreases in microcalcifications in cancers showing CR on MRI or Miller-Payne grade 5 and more frequent increases of microcalcifications in cancers showing PD on MRI or Miller-Payne grade 1. They insisted upon complete removal of all microcalcifications since residual microcalcifications can be problematic as they increase in number on subsequent mammograms. In contrast, some authors have argued that residual microcalcifications are not always indications for total mastectomy. Some insisted that decisions about surgical methods should rely on immunohistochemical subtypes and evidence of radiologic CR on MRI. Mazari et al. reported that patients with HER2+ subtype can achieve pCR even with residual mammographic microcalcifications [19]. This result is not surprising since higher pCR rate in the HER2+ subtype was reported in past trials such as the NeoSphere and TRYPHAENA studies [20, 21]. In our study, we also found that radiologic response on MRI and immunohistochemical subtype were correlated with pCR. However, changes in the extent and distribution of microcalcifications were not correlated with pathologic response. Because of these conflicting results, there is no consensus regarding management of residual microcalcifications after NAC.
Currently, there is no standardized definition of pCR. Some studies have included noninvasive cancer residuals [14], whereas others have identified pCR as complete eradication of all invasive and noninvasive cancer [22]. It remains questionable whether patients with residual noninvasive cancer after NAC should be considered as having achieved pCR. Von Minckwitz et al. proposed that pCR should be defined only as no invasive and no in situ residuals in breasts and nodes since these discriminate favorable and unfavorable outcomes [2]. Others argued that a residual DCIS component is not a worse prognosis than complete eradication of DCIS [2325]. Thus, it is unclear, in terms of prognosis such as recurrence or survival, whether all microcalcifications should be eradicated since they can be correlated with residual DCIS. Some researchers insist that malignant microcalcifications are more strongly associated with invasive breast carcinomas since they contain larger hydroxyapatite particles [26]. However, such reports solely reported cases of microcalcifications without NAC. Kim et al. reported that 37.5% of residual microcalcifications (36/96) were associated with only in situ component in final pathology after NAC [8]. They further stated that the extent of microcalcifications on mammography after NAC did not correlate with the extent of residual cancer in 38.5% of the patients. Further research is necessary to analyze the subgroup of patients who are likely to achieve pCR regardless of the extent of residual microcalcifications.
Residual microcalcifications with pCR in the breast do not affect long-term outcomes. In our study, 102 (31.6%) achieved breast pCR, 65 of whom had residual DCIS. Of the patients with breast pCR, there were four cases of distant metastases. Two patients from the pCRw/o mic group developed simultaneous local recurrence and metastatic disease eventually leading to death. One patient in the pCRw/o mic group developed distant recurrence as a first event, eventually leading to death. One patient from the pCRw/mic group developed distant metastases. With small number of patients and few events in each group, this study is underpowered to detect anything but a difference in outcome between the non-pCR and pCR groups. Consequently, we reached no definitive conclusion regarding difference in long term outcome between pCRw/o mic group and pCRw/mic group.
In contrast to our results, JY Lee et al. found that extent of surgery in cases of tumor regression without change in extent of microcalcifications must be determined based on residual microcalcifications [27]. In their study, five (10.2%) patients in the RESMIN (reduced mass with no change in residual microcalcifications) group experienced locoregional recurrence, and 80% reported residual cancers. In line with our study, JH Cheun et al. reported that margin involvement did not affect the risk of local recurrence in patients showing pCR with residual DCIS in the breast [28]. They demonstrated that the prognosis of patients with residual DCIS was not significantly affected by resection margin status, concluding that residual DCIS after NAC is clinically and pathologically different from usual DCIS. Thus, there may be no need to worry about residual DCIS on resection margin.
There are several limitations of our study. First, it was a retrospectively designed multicenter study that could involve bias owing to the heterogeneity of data. For example, the decision to perform partial mastectomy with remaining microcalcifications depended on surgeon preference. Therefore, large prospective randomized trials are needed. Second, we did not assess other types of pathologic responses such as residual cancer burden [29], grading by Miller-Payne [30], or CPS + EG score [31]. However, Miller-Payne grade evaluates tumor response based on reduction of tumor cellularity not the size of invasive or in situ carcinoma [30], and CPS + EG score does not include information of the invasive or in situ component. Therefore, the commonly used yp TNM stage by AJCC used in this study is a reasonable criterion to assess pathologic response. Last, we could not correlate residual microcalcifications with histopathological findings of residual tumours whether these microcalcifications were correlated with benign, in situ or invasive component. Prospective study is needed to find out the correlation between microcalcifications and histopathology in the future.
In conclusion, changes in the extent of microcalcifications were not correlated with pathologic response. HER2+ subtype and post-NAC findings on MRI were significant predictive factors for pCR. In patients who achieved pCR, residual microcalcifications did not increase the risk of recurrence, distant metastases, or death. Until standard guidelines are established, the eradication of residual microcalcifications remains challenging.

Acknowledgements

None.

Declarations

This retrospective study was approved by the Institutional Review Board of KBSMC (KBSMC 2018–09-033), and the requirement for individual consent was waived owing to the retrospective nature of the study.
Not applicable.

Competing interests

The authors declare no competing interests.
Open Access This 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 Cortazar P, Zhang L, Untch M, Mehta K, Costantino JP, Wolmark N, et al. Pathological complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis. Lancet. 2014;384(9938):164–72.CrossRefPubMed Cortazar P, Zhang L, Untch M, Mehta K, Costantino JP, Wolmark N, et al. Pathological complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis. Lancet. 2014;384(9938):164–72.CrossRefPubMed
2.
Zurück zum Zitat von Minckwitz G, Untch M, Blohmer JU, Costa SD, Eidtmann H, Fasching PA, et al. Definition and impact of pathologic complete response on prognosis after neoadjuvant chemotherapy in various intrinsic breast cancer subtypes. J Clin Oncol. 2012;30(15):1796–804.CrossRef von Minckwitz G, Untch M, Blohmer JU, Costa SD, Eidtmann H, Fasching PA, et al. Definition and impact of pathologic complete response on prognosis after neoadjuvant chemotherapy in various intrinsic breast cancer subtypes. J Clin Oncol. 2012;30(15):1796–804.CrossRef
3.
Zurück zum Zitat Cox RF, Morgan MP. Microcalcifications in breast cancer: lessons from physiological mineralization. Bone. 2013;53(2):437–50.CrossRefPubMed Cox RF, Morgan MP. Microcalcifications in breast cancer: lessons from physiological mineralization. Bone. 2013;53(2):437–50.CrossRefPubMed
4.
Zurück zum Zitat Kopans DB. Interpreting the mammogram. Breast imaging. Boston: Lippincott Williams Wilkins; 2007. pp. 365–480. Kopans DB. Interpreting the mammogram. Breast imaging. Boston: Lippincott Williams Wilkins; 2007. pp. 365–480.
5.
Zurück zum Zitat Doebar SC, van den Broek EC, Koppert LB, Jager A, Baaijens MHA, Obdeijn IAM, et al. Extent of ductal carcinoma in situ according to breast cancer subtypes: a population-based cohort study. Breast Cancer Res Treat. 2016;158(1):179–87.CrossRefPubMedPubMedCentral Doebar SC, van den Broek EC, Koppert LB, Jager A, Baaijens MHA, Obdeijn IAM, et al. Extent of ductal carcinoma in situ according to breast cancer subtypes: a population-based cohort study. Breast Cancer Res Treat. 2016;158(1):179–87.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Holmes D, Colfry A, Czerniecki B, Dickson-Witmer D, Francisco Espinel C, Feldman E, et al. Performance and practice Guideline for the Use of Neoadjuvant systemic therapy in the management of breast Cancer. Ann Surg Oncol. 2015;22(10):3184–90.CrossRefPubMed Holmes D, Colfry A, Czerniecki B, Dickson-Witmer D, Francisco Espinel C, Feldman E, et al. Performance and practice Guideline for the Use of Neoadjuvant systemic therapy in the management of breast Cancer. Ann Surg Oncol. 2015;22(10):3184–90.CrossRefPubMed
7.
Zurück zum Zitat Adrada BE, Huo L, Lane DL, Arribas EM, Resetkova E, Yang W. Histopathologic correlation of residual mammographic microcalcifications after neoadjuvant chemotherapy for locally advanced breast cancer. Ann Surg Oncol. 2015;22(4):1111–7.CrossRefPubMed Adrada BE, Huo L, Lane DL, Arribas EM, Resetkova E, Yang W. Histopathologic correlation of residual mammographic microcalcifications after neoadjuvant chemotherapy for locally advanced breast cancer. Ann Surg Oncol. 2015;22(4):1111–7.CrossRefPubMed
8.
Zurück zum Zitat Kim EY, Do SI, Yun JS, Park YL, Park CH, Moon JH, et al. Preoperative evaluation of mammographic microcalcifications after neoadjuvant chemotherapy for breast cancer. Clin Radiol. 2020;75(8):641. e19- e27.CrossRef Kim EY, Do SI, Yun JS, Park YL, Park CH, Moon JH, et al. Preoperative evaluation of mammographic microcalcifications after neoadjuvant chemotherapy for breast cancer. Clin Radiol. 2020;75(8):641. e19- e27.CrossRef
9.
Zurück zum Zitat Pennisi A, Kieber-Emmons T, Makhoul I, Hutchins L. Relevance of Pathological Complete Response after neoadjuvant therapy for breast Cancer. Breast Cancer (Auckl). 2016;10:103–6.PubMed Pennisi A, Kieber-Emmons T, Makhoul I, Hutchins L. Relevance of Pathological Complete Response after neoadjuvant therapy for breast Cancer. Breast Cancer (Auckl). 2016;10:103–6.PubMed
10.
Zurück zum Zitat American College of Radiology, Committee BI-RADS. ACR BI-RADS® atlas: breast imaging reporting and data system. 5th ed. Reston, VA: American College of Radiology; 2013. American College of Radiology, Committee BI-RADS. ACR BI-RADS® atlas: breast imaging reporting and data system. 5th ed. Reston, VA: American College of Radiology; 2013.
11.
Zurück zum Zitat Zhou MR, Tang ZH, Li J, Fan JH, Pang Y, Yang HJ, et al. Clinical and pathologic features of multifocal and multicentric breast cancer in Chinese women: a retrospective cohort study. J Breast Cancer. 2013;16(1):77–83.CrossRefPubMedPubMedCentral Zhou MR, Tang ZH, Li J, Fan JH, Pang Y, Yang HJ, et al. Clinical and pathologic features of multifocal and multicentric breast cancer in Chinese women: a retrospective cohort study. J Breast Cancer. 2013;16(1):77–83.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45(2):228–47.CrossRefPubMed Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45(2):228–47.CrossRefPubMed
13.
Zurück zum Zitat Fatayer H, Sharma N, Manuel D, Kim B, Keding A, Perren T, et al. Serial MRI scans help in assessing early response to neoadjuvant chemotherapy and tailoring breast cancer treatment. Eur J Surg Oncol. 2016;42(7):965–72.CrossRefPubMed Fatayer H, Sharma N, Manuel D, Kim B, Keding A, Perren T, et al. Serial MRI scans help in assessing early response to neoadjuvant chemotherapy and tailoring breast cancer treatment. Eur J Surg Oncol. 2016;42(7):965–72.CrossRefPubMed
14.
Zurück zum Zitat Green MC, Buzdar AU, Smith T, Ibrahim NK, Valero V, Rosales MF, et al. Weekly paclitaxel improves pathologic complete remission in operable breast cancer when compared with paclitaxel once every 3 weeks. J Clin Oncol. 2005;23(25):5983–92.CrossRefPubMed Green MC, Buzdar AU, Smith T, Ibrahim NK, Valero V, Rosales MF, et al. Weekly paclitaxel improves pathologic complete remission in operable breast cancer when compared with paclitaxel once every 3 weeks. J Clin Oncol. 2005;23(25):5983–92.CrossRefPubMed
15.
Zurück zum Zitat Feliciano Y, Mamtani A, Morrow M, Stempel MM, Patil S, Jochelson MS. Do calcifications seen on Mammography after Neoadjuvant Chemotherapy for breast Cancer always need to be excised? Ann Surg Oncol. 2017;24(6):1492–8.CrossRefPubMed Feliciano Y, Mamtani A, Morrow M, Stempel MM, Patil S, Jochelson MS. Do calcifications seen on Mammography after Neoadjuvant Chemotherapy for breast Cancer always need to be excised? Ann Surg Oncol. 2017;24(6):1492–8.CrossRefPubMed
16.
Zurück zum Zitat Weiss A, Lee KC, Romero Y, Ward E, Kim Y, Ojeda-Fournier H, et al. Calcifications on mammogram do not correlate with tumor size after neoadjuvant chemotherapy. Ann Surg Oncol. 2014;21(10):3310–6.CrossRefPubMed Weiss A, Lee KC, Romero Y, Ward E, Kim Y, Ojeda-Fournier H, et al. Calcifications on mammogram do not correlate with tumor size after neoadjuvant chemotherapy. Ann Surg Oncol. 2014;21(10):3310–6.CrossRefPubMed
18.
Zurück zum Zitat Yim H, Ha T, Kang DK, Park SY, Jung Y, Kim TH. Change in microcalcifications on mammography after neoadjuvant chemotherapy in breast cancer patients: correlation with tumor response grade and comparison with lesion extent. Acta Radiol. 2019;60(2):131–9.CrossRefPubMed Yim H, Ha T, Kang DK, Park SY, Jung Y, Kim TH. Change in microcalcifications on mammography after neoadjuvant chemotherapy in breast cancer patients: correlation with tumor response grade and comparison with lesion extent. Acta Radiol. 2019;60(2):131–9.CrossRefPubMed
19.
Zurück zum Zitat Mazari FAK, Sharma N, Dodwell D, Horgan K. Human epidermal growth factor 2-positive breast Cancer with Mammographic Microcalcification: relationship to Pathologic Complete Response after Neoadjuvant Chemotherapy. Radiology. 2018;288(2):366–74.CrossRefPubMed Mazari FAK, Sharma N, Dodwell D, Horgan K. Human epidermal growth factor 2-positive breast Cancer with Mammographic Microcalcification: relationship to Pathologic Complete Response after Neoadjuvant Chemotherapy. Radiology. 2018;288(2):366–74.CrossRefPubMed
20.
Zurück zum Zitat Gianni L, Pienkowski T, Im YH, Tseng LM, Liu MC, Lluch A, et al. 5-year analysis of neoadjuvant pertuzumab and trastuzumab in patients with locally advanced, inflammatory, or early-stage HER2-positive breast cancer (NeoSphere): a multicentre, open-label, phase 2 randomised trial. Lancet Oncol. 2016;17(6):791–800.CrossRefPubMed Gianni L, Pienkowski T, Im YH, Tseng LM, Liu MC, Lluch A, et al. 5-year analysis of neoadjuvant pertuzumab and trastuzumab in patients with locally advanced, inflammatory, or early-stage HER2-positive breast cancer (NeoSphere): a multicentre, open-label, phase 2 randomised trial. Lancet Oncol. 2016;17(6):791–800.CrossRefPubMed
21.
Zurück zum Zitat Schneeweiss A, Chia S, Hickish T, Harvey V, Eniu A, Hegg R, et al. Pertuzumab plus Trastuzumab in combination with standard neoadjuvant anthracycline-containing and anthracycline-free chemotherapy regimens in patients with HER2-positive early breast cancer: a randomized phase II cardiac safety study (TRYPHAENA). Ann Oncol. 2013;24(9):2278–84.CrossRefPubMed Schneeweiss A, Chia S, Hickish T, Harvey V, Eniu A, Hegg R, et al. Pertuzumab plus Trastuzumab in combination with standard neoadjuvant anthracycline-containing and anthracycline-free chemotherapy regimens in patients with HER2-positive early breast cancer: a randomized phase II cardiac safety study (TRYPHAENA). Ann Oncol. 2013;24(9):2278–84.CrossRefPubMed
22.
Zurück zum Zitat von Minckwitz G, Rezai M, Loibl S, Fasching PA, Huober J, Tesch H, et al. Capecitabine in addition to anthracycline- and taxane-based neoadjuvant treatment in patients with primary breast cancer: phase III GeparQuattro study. J Clin Oncol. 2010;28(12):2015–23.CrossRef von Minckwitz G, Rezai M, Loibl S, Fasching PA, Huober J, Tesch H, et al. Capecitabine in addition to anthracycline- and taxane-based neoadjuvant treatment in patients with primary breast cancer: phase III GeparQuattro study. J Clin Oncol. 2010;28(12):2015–23.CrossRef
23.
Zurück zum Zitat Mazouni C, Peintinger F, Wan-Kau S, Andre F, Gonzalez-Angulo AM, Symmans WF, et al. Residual ductal carcinoma in situ in patients with complete eradication of invasive breast cancer after neoadjuvant chemotherapy does not adversely affect patient outcome. J Clin Oncol. 2007;25(19):2650–5.CrossRefPubMed Mazouni C, Peintinger F, Wan-Kau S, Andre F, Gonzalez-Angulo AM, Symmans WF, et al. Residual ductal carcinoma in situ in patients with complete eradication of invasive breast cancer after neoadjuvant chemotherapy does not adversely affect patient outcome. J Clin Oncol. 2007;25(19):2650–5.CrossRefPubMed
24.
Zurück zum Zitat Jones RL, Lakhani SR, Ring AE, Ashley S, Walsh G, Smith IE. Pathological complete response and residual DCIS following neoadjuvant chemotherapy for breast carcinoma. Br J Cancer. 2006;94(3):358–62.CrossRefPubMedPubMedCentral Jones RL, Lakhani SR, Ring AE, Ashley S, Walsh G, Smith IE. Pathological complete response and residual DCIS following neoadjuvant chemotherapy for breast carcinoma. Br J Cancer. 2006;94(3):358–62.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Mazouni C, Peintinger F, Wan-Kau S, Andre F, Gonzalez-Angulo AM, Symmans F, et al. Effect on patient outcome of residual DCIS in patients with complete eradication of invasive breast cancer after neoadjuvant chemotherapy. J Clin Oncol. 2007;25(18suppl):530.CrossRef Mazouni C, Peintinger F, Wan-Kau S, Andre F, Gonzalez-Angulo AM, Symmans F, et al. Effect on patient outcome of residual DCIS in patients with complete eradication of invasive breast cancer after neoadjuvant chemotherapy. J Clin Oncol. 2007;25(18suppl):530.CrossRef
26.
Zurück zum Zitat Karamouzis MV, Likaki-Karatza E, Ravazoula P, Badra FA, Koukouras D, Tzorakoleftherakis E, et al. Non-palpable breast carcinomas: correlation of mammographically detected malignant-appearing microcalcifications and molecular prognostic factors. Int J Cancer. 2002;102(1):86–90.CrossRefPubMed Karamouzis MV, Likaki-Karatza E, Ravazoula P, Badra FA, Koukouras D, Tzorakoleftherakis E, et al. Non-palpable breast carcinomas: correlation of mammographically detected malignant-appearing microcalcifications and molecular prognostic factors. Int J Cancer. 2002;102(1):86–90.CrossRefPubMed
27.
Zurück zum Zitat Lee J, Park NJ, Park HY, Kim WW, Kang B, Keum H, et al. Oncologic necessity for the complete removal of residual microcalcifications after neoadjuvant chemotherapy for breast cancer. Sci Rep. 2022;12(1):21535.ADSCrossRefPubMedPubMedCentral Lee J, Park NJ, Park HY, Kim WW, Kang B, Keum H, et al. Oncologic necessity for the complete removal of residual microcalcifications after neoadjuvant chemotherapy for breast cancer. Sci Rep. 2022;12(1):21535.ADSCrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Cheun JH, Lee YJ, Lee JH, Shin Y, Chun JW, Baek SY, et al. Surgical margin status and survival outcomes of breast cancer patients treated with breast-conserving surgery and whole-breast irradiation after neoadjuvant chemotherapy. Breast Cancer Res Treat. 2022;194(3):683–92.CrossRefPubMed Cheun JH, Lee YJ, Lee JH, Shin Y, Chun JW, Baek SY, et al. Surgical margin status and survival outcomes of breast cancer patients treated with breast-conserving surgery and whole-breast irradiation after neoadjuvant chemotherapy. Breast Cancer Res Treat. 2022;194(3):683–92.CrossRefPubMed
29.
Zurück zum Zitat Symmans WF, Peintinger F, Hatzis C, Rajan R, Kuerer H, Valero V, et al. Measurement of residual breast cancer burden to predict survival after neoadjuvant chemotherapy. J Clin Oncol. 2007;25(28):4414–22.CrossRefPubMed Symmans WF, Peintinger F, Hatzis C, Rajan R, Kuerer H, Valero V, et al. Measurement of residual breast cancer burden to predict survival after neoadjuvant chemotherapy. J Clin Oncol. 2007;25(28):4414–22.CrossRefPubMed
30.
Zurück zum Zitat Ogston KN, Miller ID, Payne S, Hutcheon AW, Sarkar TK, Smith I, et al. A new histological grading system to assess response of breast cancers to primary chemotherapy: prognostic significance and survival. Breast. 2003;12(5):320–7.CrossRefPubMed Ogston KN, Miller ID, Payne S, Hutcheon AW, Sarkar TK, Smith I, et al. A new histological grading system to assess response of breast cancers to primary chemotherapy: prognostic significance and survival. Breast. 2003;12(5):320–7.CrossRefPubMed
31.
Zurück zum Zitat Jeruss JS, Mittendorf EA, Tucker SL, Gonzalez-Angulo AM, Buchholz TA, Sahin AA, et al. Combined use of clinical and pathologic staging variables to define outcomes for breast cancer patients treated with neoadjuvant therapy. J Clin Oncol. 2008;26(2):246–52.CrossRefPubMed Jeruss JS, Mittendorf EA, Tucker SL, Gonzalez-Angulo AM, Buchholz TA, Sahin AA, et al. Combined use of clinical and pathologic staging variables to define outcomes for breast cancer patients treated with neoadjuvant therapy. J Clin Oncol. 2008;26(2):246–52.CrossRefPubMed
Metadaten
Titel
Impact of residual microcalcifcations on prognosis after neoadjuvant chemotherapy in breast cancer patients
verfasst von
Eun Young Kim
Kwan Ho Lee
Ji-Sup Yun
Yong Lai Park
Chan Heun Park
Sung Yoon Jang
Jai Min Ryu
Se Kyung Lee
Byung-Joo Chae
Jeong Eon Lee
Seok Won Kim
Seok Jin Nam
Jong Han Yu
Publikationsdatum
01.12.2024
Verlag
BioMed Central
Erschienen in
BMC Women's Health / Ausgabe 1/2024
Elektronische ISSN: 1472-6874
DOI
https://doi.org/10.1186/s12905-024-02973-9

Weitere Artikel der Ausgabe 1/2024

BMC Women's Health 1/2024 Zur Ausgabe

Blutdrucksenkung könnte Uterusmyome verhindern

Frauen mit unbehandelter oder neu auftretender Hypertonie haben ein deutlich erhöhtes Risiko für Uterusmyome. Eine Therapie mit Antihypertensiva geht hingegen mit einer verringerten Inzidenz der gutartigen Tumoren einher.

Antikörper-Wirkstoff-Konjugat hält solide Tumoren in Schach

16.05.2024 Zielgerichtete Therapie Nachrichten

Trastuzumab deruxtecan scheint auch jenseits von Lungenkrebs gut gegen solide Tumoren mit HER2-Mutationen zu wirken. Dafür sprechen die Daten einer offenen Pan-Tumor-Studie.

Mammakarzinom: Senken Statine das krebsbedingte Sterberisiko?

15.05.2024 Mammakarzinom Nachrichten

Frauen mit lokalem oder metastasiertem Brustkrebs, die Statine einnehmen, haben eine niedrigere krebsspezifische Mortalität als Patientinnen, die dies nicht tun, legen neue Daten aus den USA nahe.

S3-Leitlinie zur unkomplizierten Zystitis: Auf Antibiotika verzichten?

15.05.2024 Harnwegsinfektionen Nachrichten

Welche Antibiotika darf man bei unkomplizierter Zystitis verwenden und wovon sollte man die Finger lassen? Welche pflanzlichen Präparate können helfen? Was taugt der zugelassene Impfstoff? Antworten vom Koordinator der frisch überarbeiteten S3-Leitlinie, Prof. Florian Wagenlehner.

Update Gynäkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert – ganz bequem per eMail.