Skip to main content
Erschienen in: Breast Cancer Research and Treatment 3/2014

Open Access 01.12.2014 | Preclinical study

Pathological non-response to chemotherapy in a neoadjuvant setting of breast cancer: an inter-institutional study

verfasst von: D. Balmativola, C. Marchiò, M. Maule, L. Chiusa, L. Annaratone, F. Maletta, F. Montemurro, J. Kulka, P. Figueiredo, Z. Varga, I. Liepniece-Karele, G. Cserni, E. Arkoumani, I. Amendoeira, G. Callagy, A. Reiner-Concin, A. Cordoba, S. Bianchi, T. Decker, D. Gläser, C. Focke, P. van Diest, D. Grabau, E. Lips, J. Wesseling, R. Arisio, E. Medico, C. Wells, A. Sapino

Erschienen in: Breast Cancer Research and Treatment | Ausgabe 3/2014

Abstract

To identify markers of non-response to neoadjuvant chemotherapy (NAC) that could be used in the adjuvant setting. Sixteen pathologists of the European Working Group for Breast Screening Pathology reviewed the core biopsies of breast cancers treated with NAC and recorded the clinico-pathological findings (histological type and grade; estrogen, progesterone receptors, and HER2 status; Ki67; mitotic count; tumor-infiltrating lymphocytes; necrosis) and data regarding the pathological response in corresponding surgical resection specimens. Analyses were carried out in a cohort of 490 cases by comparing the groups of patients showing pathological complete response (pCR) and partial response (pPR) with the group of non-responders (pathological non-response: pNR). Among other parameters, the lobular histotype and the absence of inflammation were significantly more common in pNR (p < 0.001). By ROC curve analyses, cut-off values of 9 mitosis/2 mm2 and 18 % of Ki67-positive cells best discriminated the pNR and pCR + pPR categories (p = 0.018 and < 0.001, respectively). By multivariable analysis, only the cut-off value of 9 mitosis discriminated the different response categories (p = 0.036) in the entire cohort. In the Luminal B/HER2− subgroup, a mitotic count <9, although not statistically significant, showed an OR of 2.7 of pNR. A lobular histotype and the absence of inflammation were independent predictors of pNR (p = 0.024 and <0.001, respectively). Classical morphological parameters, such as lobular histotype and inflammation, confirmed their predictive value in response to NAC, particularly in the Luminal B/HER2− subgroup, which is a challenging breast cancer subtype from a therapeutic point of view. Mitotic count could represent an additional marker but has a poor positive predictive value.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1007/​s10549-014-3192-3) contains supplementary material, which is available to authorized users.

Introduction

Breast cancer is a heterogeneous disease and the different therapeutic modalities used for breast cancer patients reflect this heterogeneity. As highlighted in the 13th St. Gallen International Breast Cancer Conference [1], endocrine therapy (often used alone) is recommended for Luminal A-like breast cancers, anti-HER2 therapy is the most crucial intervention in HER2 positive disease, whereas the only available approach for triple-negative tumors is the use of cytotoxic chemotherapy. Luminal B/HER2− carcinomas represent an intermediate entity from a clinico-therapeutic point of view as endocrine therapy is recommended for all patients and cytotoxic therapy for most of them [1]. Significant efforts are currently being employed to discriminate patients who will benefit from chemotherapy. Multi-gene assays appear to help recognize patients with Luminal disease for whom chemotherapy is not effective, i.e, patients with a low Recurrence Score (RS) by Oncotype DX [2, 3] and those with a “good prognosis signature” with the 70-gene signature assay [4]. Similar results were obtained in the neoadjuvant setting: no or very few pathological complete responses (pCR) to neoadjuvant chemotherapy (NAC) were observed among patients with low risk of recurrence (ROR) based on PAM50 [5]. Additionally, both the 70-gene good prognosis signature and a low 21-gene RS predict a low probability of pCR [6, 7]. The proliferative activity of tumors as assessed with immunohistochemical detection of the cell-cycle-specific antigen Ki67 has been extensively studied [812] and is presently one of the parameters used to address systemic adjuvant therapy for patients with Luminal breast cancers [13]. A recent study has shown a beneficial effect of the addition of chemotherapy to hormone therapy in Luminal B/HER2− carcinomas with a high proliferative index (Ki67 > 32 %) [14]. A significant impact in breast cancer prognosis has also been suggested for the mitotic count [15, 16], but few studies have considered its predictive value in the response to chemotherapy [17, 18]. Cytotoxic drugs, such as taxanes, which target microtubules, are believed to be more effective on cells that proliferate rapidly. However, it is known that the mitotic count is low in many chemosensitive human cancers [19]. Despite this observation, no studies have been carried out to solve this specific issue in breast cancer.
The neoadjuvant setting offers an invaluable opportunity to test the response to therapy, considering that the selection of regimens for NAC generally follows guidelines similar to those applied in the conventional adjuvant setting [20]. However, the majority of studies have focused their attention on pCR as the primary endpoint for response to chemotherapy because of its prognostic value [21, 22]. Thus, statistical analyses performed on neoadjuvant studies tend to group together partial response and non-response to treatment. To our knowledge, studies focusing on the non-responsive breast cancer category are not on record.
Taking all these data together, the main goal of the present study was to identify markers of non-response to NAC that could be used in the adjuvant setting, in particular markers for the Luminal B/HER2− category. The analyses were performed by comparing the group of patients showing some response (from partial to complete) to NAC with the group of patients lacking any response. Furthermore, we investigated for the first time the potential value of the mitotic index in offering additional information about the likelihood of non-response to treatment with taxane-based regimens.

Patients and methods

Study design

Sixteen pathologists of the European Working Group for Breast Screening Pathology from different European Institutions (Città della Salute e della Scienza di Torino, Turin, Italy; 2nd Department of Pathology, Semmelweis University, Budapest, Hungary; Centro Regional De Oncologia De Coimbra, Coimbra, Portugal; University Hospital Zurich, Switzerland; State Pathology Center, Riga, Latvia; Bács-Kiskun County Teaching Hospital, Kecskemét, Hungary; Hospital S. Joao, Porto, Portugal; Clinical Sciences Institute, Galway, Ireland; Donauspital am SMZO, Vienna, Austria; Complejo Hospitalario de Navarra, Pamplona, Navarra, Spain; AOU Careggi, Florence, Italy; Dietrich Bonhoeffer Medical Centre, Neubrandenburg, Germany; University Medical Center Utrecht, Utrecht, The Netherlands; Skane University Hospital, Lund, Sweden; The Netherlands Cancer Institute, Amsterdam, The Netherlands; The Princess Alexandra Hospital, Harlow, Essex, United Kingdom; University College Hospital, London, United Kingdom) were asked to participate to the study. These pathologists reviewed the core biopsy histology slides pertaining to patients with breast cancer treated with NAC at their Institutions. The number of core biopsies available for each patient ranged from 2 to 4, depending on the Institution protocol. The single Institutions recorded a list of clinico-pathological features in a dedicated database, as specified below.

Data collection and definitions

The data recorded were as follows: (i) in the pre-treatment biopsy: the histological type and grade [23], mitotic count, Ki67 proliferation index, presence of inflammation, presence of necrosis, Estrogen Receptor (ER), Progesterone Receptor (PgR), and HER2 status (based on both the immunostaining score and in situ hybridization analysis for score 2+); (ii) in the histological examination of the post-treatment surgical specimens, the degree of response to therapy was categorized following Pinder et al. [24] (Supplementary Table 1) in pathological complete response (pCR) if no residual invasive tumor was found (in situ carcinoma may be present), pathological partial response (pPR) if residual disease or minor signs of response were present on the surgical specimens compared to the tumor cellularity of the pre-treatment core biopsies, pathological non-response (pNR) if no evidence of response to therapy was detected (the presence of lymph node metastasis was not taken into account); and (iii) from the clinical records: the diameter of the tumor before NAC and the type of NAC. The response in lymph nodes was evaluated as detailed in Supplementary Table 1.
With regard to the definition of molecular subtypes, we referred to the St. Gallen recommendations from 2013 [1] that include five categories (Luminal A, Luminal B/HER2−, Luminal B/HER2+, HER2+ and triple negative). In particular, the Luminal B/HER2− category included ER positive carcinomas with >14 % of Ki67 [13] and/or PgR <20 % [25].
To define the methods to assess mitoses and Ki67, the participants had a preliminary meeting. The mitotic figures were counted in 10 high power fields of each core biopsy if possible, and the mitotic count was reported as the mean value. The results were then normalized as the number of mitosis per 2 mm2, i.e., an area equal to 10 fields at high magnification (40×) with a 0.51-mm objective diameter. Ki67 scoring (all centers used the MIB-1 antibody clone) was performed by counting a range of 100–500 cells (depending on the cellularity of the specimen), including also hot spot areas.
Inflammation was defined in two ways: (i) presence/absence of any clearly detectable tumor-infiltrating lymphocytes (TILs) within tumor cells (intra-tumoral) and/or stroma (stromal) at H&E; (ii) percentage of stromal TILs (st-TILs%) as recently recommended [26] (i.e., area occupied by mononuclear inflammatory cells over total intra-tumoral stromal area).
The final cohort comprised 506 cases, 490 of which had information available about their response to NAC.

Statistical analysis

For the purpose of defining the tumors that will not benefit from chemotherapy, univariate analyses were conducted by dividing the cohort of patients into two sets: pNR and pCR + pPR.
Differences in the distribution of the characteristics between the specimens of the patients with pCR or pPR and the patients with pNR were evaluated using Pearson’s Chi-Square Test and Fisher’s Exact Test for comparison of nominal variables and performing independent sample Nonparametric Test (Mann–Whitney U-Test) for comparison with continuous variables.
The cut-offs for Ki67 %, for the number of mitoses, and for st-TILs% that are able to discriminate the response to treatment (pNR vs pCR + pPR) were assessed for the entire cohort of breast cancer patients by receiver operating characteristic (ROC) curve analysis, in which the sensitivity (SE) is plotted as a function of 1-specificity (1-SP). The Youden Index (J), one of the main summary statistics of the ROC curve, defines the maximum potential effectiveness of a biomarker. J can be formally defined as J = max c {SE (c) + SP (c) − 1}. The cut-off value that achieves this maximum is referred to as the optimal cut-off point that optimizes the biomarker’s discriminating power when the sensitivity and specificity bear equal weight [27, 28].
The interactions between the response to treatment (pNR vs pCR + pPR) and the variables statistically significant at univariate analysis were tested in the entire cohort and in the Luminal B/HER2− cohort in a multivariable binary regression model.
All reported p values (p) were two-sided and p < 0.05 was considered to be statistically significant. All analyses were carried out using SPSS version 20 (SPSS Inc, Chicago, IL, USA) and STATA version 12.0 (Stata Corporation, College Station, Texas) statistical software. This article was written in accordance with the guidelines of the Reporting Recommendations for Tumor Marker Prognostic Studies (REMARK criteria) [29].

Results

Entire cohort analysis

The clinical and histopathological features of the entire population are summarized in Table 1. The analyses of response to treatment were performed either considering primary tumor and lymph node response (454 cases) or tumor response only (490 cases). No statistically significant differences in terms of final distribution in response categories were observed between these separate analyses (Supplementary Tables 2, 3, and 4); therefore, we took into account the response on primary tumors only, which allowed us to perform the analysis in a larger number of cases (Supplementary Table 4).
Table 1
Histopathological characteristics of pre-treatment core biopsies and therapy-related data on the entire cohort of 506 patients with breast cancer treated with neoadjuvant chemotherapy
Histopathological data
Therapy-related data
 
Number of cases
Total %
Valid %
 
Number of cases
Total %
Valid %
Grade
Response
1
35
6.9
7.3
Complete
133
26.3
27.1
2
247
48.8
51.7
Partial
283
55.9
57.8
3
196
38.7
41
Absent
74
14.6
15.1
Unknown
28
5.5
 
Unknown
16
3.2
 
ER score
Antracyclins
Positive
333
65.8
65.8
Yes
412
81.4
88.2
Negative
173
34.2
34.2
No
55
10.9
11.8
Unknown
0
0.0
 
Unknown
39
7.7
 
PGR score
Cyclophosphamide
Positive
280
55.3
55.4
Yes
347
68.6
73.5
Negative
225
44.5
44.6
No
125
24.7
26.2
Unknown
1
0.2
 
Unknown
34
6.7
 
PGR cut-off
Taxanes
<20 %
290
57.3
57.5
Yes
328
64.8
68.2
≥20 %
214
42.3
42.5
No
153
30.2
31.8
Unknown
2
0.4
 
Unknown
25
4.9
 
HER2 score
Herceptin
Positive
104
20.6
21
Yes
90
17,8
18.5
Negative
391
77.3
79
No
397
78.5
81.5
Unknown
11
2.2
 
Unknown
19
3.8
 
Lobular
5-FU
Yes
40
7.9
8
Yes
33
6,5
26.4
No
462
91.3
92
No
92
18.2
73.6
Unknown
4
0.8
 
Unknown
381
75.3
 
Inflammation
Vinca alkaloids
TILs present
258
51
74.1
Yes
15
3.0
3.8
TILs absent
90
17.8
25.9
No
381
75.3
96.2
Unknown
158
31.2
 
Unknown
110
21.7
 
Necrosis
    
Present
128
25.3
36.8
    
Absent
220
43.5
63.2
    
Unknown
158
31.2
     
Molecular categories*
   
Luminal A
70
13.8
14.8
    
Luminal B/HER2−
188
37.2
39.8
    
Luminal B/HER2+
46
9.1
9.7
    
Triple-negative
57
11.3
12.1
    
HER2+
111
21.9
23.5
    
Unknown
34
6.7
  
*following St. Gallen recommendations [1]
To evaluate the impact of the heterogeneity of standard of care in the different Institutions, we first analyzed by Chi-Square Test the difference of response taking into account the different treatment protocols. Twenty-one different treatment schemes were reported. We decided to cluster them in 3 main groups: 259 cases with taxanes, 135 without taxanes, and 87 with Herceptin. The two response categories (pNR vs pCR + pPR) were not significantly different in terms of treatment protocols as reported in Table 2. In particular, by excluding HER2 + carcinomas treated with Herceptin, we found that taxanes were used in association with other cytotoxic drugs in 217 cases (66 %) out of 328 patients with a pCR + pPR. Of 66 patients with a pNR, taxanes were used in the chemotherapy protocols in 42 cases (64 %).
Table 2
Treatment protocols and distribution of histopathological findings obtained from pre-treatment core biopsies in patients with no response (pNR) and with complete and partial response (pCR + pPR) to neoadjuvant chemotherapy in 490 cases
 
pNR (n = 74)
pCR + pPR (n = 416)
p values
Number of cases
Total %
Valid %
Number of cases
Total %
Valid %
Treatment protocols
with taxanes
42
56.8
57.5
217
52.2
53.2
0.693
without taxanes
24
32.4
32.9
111
26.7
27.2
with Herceptin
7
9.5
9.6
80
19.2
19.6
 
Unknown
1
1.4
 
8
1.9
  
Grade
1
6
8.1
8.3
27
6.5
6.9
0.007
2
48
64.9
66.7
189
45.4
48.3
3
18
24.3
25.0
175
42.1
44.8
Unknown
2
2.7
 
25
6.0
  
ER score
Positive
60
81.1
81.1
261
62.7
62.7
0.002
Negative
14
18.9
18.9
155
37.3
37.3
Unknown
0
0.0
 
0
0.0
  
PgR score
Positive
53
71.6
71.6
219
52.6
52.8
0.003
Negative
21
28.4
28.4
196
47.1
47.2
Unknown
0
0.0
 
1
0.2
  
PGR cut-off
<20 %
30
40.5
40.5
249
59.9
60.1
0.002
≥ 20 %
44
59.5
59.5
165
39.7
39.9
Unknown
0
0.0
 
2
0.5
  
HER2 score
Positive
8
10.8
11.1
93
22.4
22.9
0.027
Negative
64
86.5
88.9
314
75.5
77.1
Unknown
2
2.7
 
9
2.2
  
Lobular
Yes
14
18.9
19.2
24
5.8
5.8
<0.001
No
59
79.7
80.8
389
93.5
94.2
Unknown
1
1.4
 
3
0.7
  
Inflammation
TILs present
23
31.1
51.1
229
55.0
77.4
<0.001
TILs absent
22
29.7
48.9
67
16.1
22.6
Unknown
29
39.2
 
120
28.8
  
St-TILs score
>1%
16
21.6
36.4
189
45.4
63.9
0.001
≤1%
28
37.8
63.6
107
25.7
36.1
Unknown
30
40.5
 
120
28.8
  
Necrosis
Present
19
25.7
42.2
107
25.7
36.1
0.508
Absent
26
35.1
57.8
189
45.4
63.9
Unknown
29
39.2
 
120
28.8
  
With regard to tumor size before NAC, no differences were observed between pNR and pCR + pPR (mean values 35.3 mm (SD: 16.3 mm) and 37.6 mm (SD: 22.2 mm), respectively; p = 0.881). Similarly, tumor necrosis was found at a similar rate in both response groups.
The tumors in the patients with pNR were less frequently of high histological grade (G3) (25 vs 45 %) and more frequently ER + (81 vs 63 %), PgR + (72 vs 53 %) and HER2− (89 vs 77 %) than were the tumors in the pCR + pPR category (Table 2). A lobular histotype was also more common in the pNR than in the pCR + pPR category (19 vs 6 %). On the contrary, the presence of TILs was significantly less common in the pNR (51 %) than in the pCR + pPR (77 %) category (Table 2). Similarly, the st-TILs % was differently distributed within pNR and pCR + pPR categories (mean values: 7 vs 12 %; SDs: 12.42 vs 17.51 %) (p = 0.002) (Supplementary Fig. 1a). Based on ROC curve analysis, the area under the curve was 0.639 (95 % CI 0.544–0.734) and the cut-off values of 1 % of st-TILs (J: 0.297) significantly discriminated the pNR from pCR + pPR group. In particular, to discriminate pNR tumors, a TILs% ≤1 had a sensitivity of 64 %, a specificity of 64 %, a positive predictive value (PPV) of 21 %, and a negative predictive value (NPV) of 92 % (Supplementary Fig. 1c).
Both the mitotic numbers and Ki67 percentages were differently distributed in the pNR and pCR + pPR categories (mean values: 8.5 vs 11.3 for mitosis and 27.9 vs 38.4 % for Ki67; SDs: 10.2 vs 12.1 for mitosis and 14.9 vs 25.7 % for Ki67) (p = 0.046 and 0.001, respectively) (Fig. 1). To determine the cut-offs for proliferation that best discriminate between the pNR and pCR + pPR categories, we performed ROC curve analyses (Fig. 2a, b). The area under the curve was 0.575 (95 % CI 0.506–0.644) for mitoses and 0.635 (95 % CI 0.555–0.715) for Ki67. The cut-off values of 9 mitosis/2 mm2 (J: 0.169) and 18 % of Ki67 positivity (J: 0.253) significantly discriminated the pNR from pCR + pPR groups (Fisher’s exact test p = 0.018 and <0.001, respectively). In particular, to discriminate pNR tumors, the cut-off of 9 mitosis had a sensitivity of 71 %, a specificity of 44 %, a PPV of 18 %, and a NPV of 90 %, whereas the Ki67 cut-off of 18 % showed a sensitivity of 47 %, a specificity of 77 %, a PPV of 26 %, and a NPV of 90 % (Fig. 2c, d). No statistically significant differences in the mitotic index were identified in the pNR and pCR + pPR groups in both the group of patients treated with and without taxanes. On the contrary, the Ki67 percentages were differently distributed in the two response categories, in both the group of patients treated with (p = 0.047) and without taxanes (p = 0.017). For these analyses, patients treated with Herceptin were excluded.

Molecular subtype analysis

According to the St. Gallen recommendations [1], 70 tumors were Luminal A-like, 188 were Luminal B/HER2−, 46 were Luminal B/HER2+, 57 were HER2+, and 111 were triple-negative (Table 1). To evaluate the reliability of our data, we first assessed the distribution of patients with pCR in the different molecular subtypes. As expected, the lowest rate of pCR was observed in Luminal A (3 %) followed by Luminal B/HER2− (16 %). However, the response rate was significantly different between the two subtypes (p = 0.01), and both of them showed a significant difference of response compared with the other subtypes (all of p < 0.01). The highest rates of pCR were achieved in the HER2+ (58 %) and in triple-negative (43 %) subtypes. On the other hand, the Luminal A cancers showed the highest percentage of pNR (31.3 %) (Table 3).
Table 3
Response distribution (a) and statistical correlations (b) in different molecular subtypes
Molecular subtypes
a. Response distribution
pCR vs pPR + pNR
pNR vs pCR + pPR
pCR
pCR %
pPR + pNR
pPR + pNR %
pNR
pNR %
pCR + pPR
pCR + pPR %
Luminal A
2
3
65
97
21
31.3
46
68.7
Luminal B/HER2−
29
16
152
84
27
14.9
154
85.1
Luminal B/HER2+
18
40
27
60
5
11.1
40
88.9
HER2+
32
58.2
23
41.8
3
5.4
52
94.6
Triple-negative
128
42.1
62
56.9
11
10.1
98
89.9
Comparison of molecular subtypes
b. Statistical correlations of the response distribution (p values)
pCR vs pPR + pNR
pNR vs pCR + pPR
Luminal A vs Luminal B/HER2−
0.01
0.006
Luminal A vs Luminal B/HER2+
0.0001
0.002
Luminal A vs HER2+
0.0001
0.0008
Luminal A vs Triple-negative
0.0001
0.0008
Luminal B/HER2− vs Luminal B/HER2+
0.0008
0.67
Luminal B/HER2− vs HER2+
0.0001
0.1
Luminal B/HER2− vs Triple-negative
0.0001
0.3
Luminal B/HER2+ vs HER2+
0.1
0.5
Luminal B/HER2+ vs Triple-negative
0.8
0.8
HER2+ vs Triple-negative
0.09
0.47
pCR pathological complete response, pPR pathological partial response, pNR pathological non-response
Histological grade, radiological tumor diameter pre-CT, necrosis, and mitotic count did not show a significant difference between different response categories (all p > 0.05). The presence of a lobular histotype varied among the different molecular subtypes (15 % in Luminal A; 8.5 % in Luminal B/HER2−; 2.2 % in Luminal B/HER2+; 5.3 % in HER2, and 1.8 % in triple negative) with the highest frequency in the Luminal subtypes. However, the lobular histotype (26 % vs 5 %) significantly differed between the pNR and pCR + pPR tumors (p < 0.00) only in the Luminal B/HER2− cancers (sensitivity: 26 %; specificity: 95 %; PPV: 48 %; NPV: 88 %) (Fig. 3a), although this histotype was more frequent in Luminal A. The absence of TILs was also distributed differently between the pNR and pCR + pPR categories (69 vs 23 %) in the Luminal B/HER2− breast cancers (p < 0.000) (sensitivity: 69 %; specificity: 77 %; PPV: 28 %; NPV: 95 %) (Fig. 3b), and st-TILs % also resulted differentially distributed in pNR and pCR + pPR categories (mean values: 3 vs 10 %; SDs: 5.73 vs 17.23 %) (p = 0.002) in this molecular subtype (Supplementary Fig. 1b). A TILs % ≤1 was significantly related with pNR (p = 0.002) (sensitivity: 81 %; specificity: 61 %; PPV: 22 %; NPV: 96 %) (Supplementary Fig. 1d).
The distribution and cut-off of the percentage of Ki67 were not analyzed in the Luminal subtypes as proliferation is one of the parameters used to define the Luminal A and B categories.
In HER2 + and in triple-negative carcinomas, neither the distribution nor the cut-off value of Ki67 of 18 % were able to discriminate between the different response categories (p > 0.05).

Multivariable analyses

We performed a logistic binary regression on both the entire cohort and the Luminal B/HER2− carcinomas sub vs group. When two different variables of the same parameter (e.g., PgR: score or cut-off; TILs: presence/absence or st-TILS score) were available, the most significant at univariate analysis was used for multivariable test.
In the entire cohort, the cut-off of 9 mitosis was statistically related to different response categories (p = 0.036); in particular, the Odds Ratio (OR) for patients with pNR was 3.3 times higher for carcinomas with ≤9 mitoses than for patients whose tumors had >9 mitoses. A trend for statistical significance was also observed for the histological type (lobular vs non-lobular) (p = 0.071), which provided an OR of 3.6 (Table 4a).
Table 4
Results of multivariable analyses of the entire cohort (a) and the Luminal B/HER2− (b) carcinoma subtype
 
OR
95% CI for OR
p value
Lower
Upper
a. Multivariable analysis—entire cohort
 Maximum radiological diameter
0.991
0.968
1.014
0.442
 Lobular carcinoma
3.591
0.895
14.408
0.071
 TILs absent
1.906
0.693
5.239
0.211
 Necrosis absent
2.011
0.722
5.599
0.181
 Low grade (G1 + G2)
0.594
0.191
1.851
0.369
 ER positive (>1%)
0.940
0.256
3.457
0.926
 PgR positive (>20 %)
1.710
0.549
5.330
0.355
 HER2 negative
1.739
0.518
5.833
0.370
 Mitotic number ≤ 9
3.323
1.081
10.211
0.036
 Ki-67 < 18 %
0.626
0.192
2.042
0.437
b. Multivariable analysis – Luminal B/HER2− subtype
 Maximum radiological diameter
0.979
0.938
1.022
0.331
 Lobular carcinoma
12.503
1.390
112.445
0.024
 TILs absent
6.254
1.343
29.126
0.020
 Necrosis absent
1.096
0.204
5.888
0.915
 Low grade (G1 + G2)
0.167
0.026
1.062
0.058
 Mitotic number ≤ 9
2.695
0.503
14.425
0.247
OR odds ratio, CI confidence interval
Within the Luminal B/HER2− subgroup, the analysis was performed excluding the ER score, the PgR cut-off, and the HER2 score because these variables are used to define this category [1]. In Luminal B/HER2− tumors, the lobular histotype and the absence of inflammation were independent predictors of pNR (p = 0.024 and 0.020, respectively). The ORs for pNR were 12.5 times higher for carcinomas of lobular histological type than non-lobular cancers and 6.2 times higher for carcinomas with no evidence of TILs than cancers with any clearly detectable intra-tumoral or stromal lymphocyte infiltrates. The mitotic cut-off showed an OR of 2.7 in the Luminal B/HER2− subgroup; however, in the multivariable model, it did not reach statistical significance (p = 0.247) (Table 4b).

Discussion

With the goal of identifying patients for whom chemotherapy is unlikely to yield a beneficial effect, we analyzed the differences in the distribution of common histopathological features between non-responder patients and patients who show some response (either partial or complete) to NAC in a neoadjuvant cohort. As in previous reports, which focused on achieving a pCR as a primary endpoint, our univariate analysis demonstrated that ER and PgR positivity are associated with a lack of response, as was the lobular histological type and the absence of inflammation. However, for the multivariable analyses neither the expression of ER nor PgR were independent variables for discriminating the pNR category from the pCR + pPR category. As recently suggested by Delpech et al. [27] for pCR, our result supports the idea that pNR to NAC is more related to intrinsic tumor characteristics than to ER expression. Many studies have shown that the response to NAC is lower in terms of pCR in locally advanced lobular carcinomas than in invasive ductal carcinomas [3034]. The presence of inflammation has instead been proposed as a predictive factor of response to chemotherapy in breast cancer in general [1, 3538]. A recent study for harmonization of the evaluation of TILs recommends considering only the stromal inflammation [26]; however, the authors specify that “this recommendation is based on the methodology used in published phase III studies, implying that there is room for future refinement as evidence accumulates to show the validity of alternative parameters and/or methodologies that improve upon this practice.” In the present study, the correlation between pNR and the absence of TILs (both intra-tumoral and/or stromal) was slightly stronger (p < 0.000) than between pNR and st-TILs% ≤1 (p = 0.001). However, in the pNR category the number of cases with the absence of TILs (22 cases, 48.9 %) was lower than that with st-TILs% ≤1 (28 cases, 63.6 %). This may influence the multivariable analysis results, though it may also indicate a better selection of pNR cases.
By considering specific intrinsic subtypes, Loi et al. observed a significant association between the presence of TILs and a good prognosis in triple-negative and HER2 + breast cancers [39]. Recent evidence indicates that in triple-negative breast cancer, both stromal as well as intra-tumoral TILs are predictive of pathological response to platinum-based NAC [40]. In our study, we showed that the lobular histotype and the absence of TILs were associated with pNR in the entire cohort using univariate analysis. However, using multivariable analyses, these two parameters correlated with pNR only in the Luminal B/HER2− breast cancers. In other subtypes, it is possible that other factors rather than histological type and inflammation concur to the pNR, in particular in Luminal A breast carcinomas where the pNR was high and the lobular histotype was more frequent than in Luminal B/HER2−.
Luminal B is the most challenging molecular subtype of breast cancers in terms of treatment. By being more aggressive than Luminal A, Luminal B/HER2− breast cancers are generally treated with both endocrine therapy and chemotherapy, but this approach is not always effective [41]. Being able to recognize a priori which Luminal B/HER2− carcinomas will not respond to chemotherapy would help avoid the toxic adverse effects of the treatment and plan a more effective anti-hormone treatment.
When evaluating markers of proliferation, our data showed significant differences for both the distribution of the mitotic numbers and the percentage of Ki67 within different response categories using univariate analyses on the entire cohort of patients. The main limitation of our study is related to the analysis of core biopsies, which may not completely represent the heterogeneous expression of proliferation markers within a given tumor. Nevertheless, the measurement of Ki67 is poorly standardized across laboratories even on whole tissue sections [4244]. On the other hand, Lehr et al. [45] have recently shown that while the percentages of Ki67 (evaluated by MIB-1 immunostaining) were comparable in biopsies and resection specimens irrespective of the method of quantification, the mitotic count was significantly overestimated in resection specimens of invasive breast carcinomas most likely as a result of a delay of tissue fixation. Another critical issue may be the high standard deviation values for both mitoses and Ki67; however, with ROC curve analysis, we identified two cut-off values (9 for mitosis count and 18 % for Ki67 expression) that performed well in differentiating the pNR and pPR + pCR categories using univariate analyses. In addition, the mitotic count was the only independent variable in the entire cohort of patients but not in any of the molecular subtypes. This finding could stem from the low number of cases available for each category, and larger studies are warranted to investigate further the utility of mitotic counts within distinct molecular subgroups. The hypothesis that a low mitotic count would characterize non-responsive patients treated with taxanes was not supported by our results. This finding is consistent with studies that suggest that drugs damaging DNA or microtubules are active through non-mitotic mechanisms [19].
In conclusion, although the pNR category has been neglected in the literature, the non-responsive patients could merit further investigation by using genetic signatures, particularly in breast cancers that do have the option of chemotherapy, such as Luminal B/HER2− breast carcinomas.

Acknowledgments

This work was supported by Ricerca Sanitaria Finalizzata (RF-2010-2310674 to AS) and by the Italian Association for Cancer Research—AIRC (MFAG13310 to CM).

Conflict of interest

The authors declare that they have no conflict of interest.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution Noncommercial License which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and the source are credited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Gynäkologie

Kombi-Abonnement

Mit e.Med Gynäkologie erhalten Sie Zugang zu CME-Fortbildungen der beiden Fachgebiete, den Premium-Inhalten der Fachzeitschriften, inklusive einer gedruckten gynäkologischen oder urologischen Zeitschrift Ihrer Wahl.

Anhänge

Electronic supplementary material

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat Goldhirsch A, Winer EP, Coates AS, Gelber RD, Piccart-Gebhart M, Thurlimann B, Senn HJ (2013) 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):2206–2223PubMedCentralPubMedCrossRef Goldhirsch A, Winer EP, Coates AS, Gelber RD, Piccart-Gebhart M, Thurlimann B, Senn HJ (2013) 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):2206–2223PubMedCentralPubMedCrossRef
2.
Zurück zum Zitat Albain KS, Barlow WE, Shak S, Hortobagyi GN, Livingston RB, Yeh IT, Ravdin P, Bugarini R, Baehner FL, Davidson NE, Sledge GW, Winer EP, Hudis C, Ingle JN, Perez EA, Pritchard KI, Shepherd L, Gralow JR, Yoshizawa C, Allred DC, Osborne CK, Hayes DF (2010) Prognostic and predictive value of the 21-gene recurrence score assay in postmenopausal women with node-positive, oestrogen-receptor-positive breast cancer on chemotherapy: a retrospective analysis of a randomised trial. Lancet Oncol 11(1):55–65PubMedCentralPubMedCrossRef Albain KS, Barlow WE, Shak S, Hortobagyi GN, Livingston RB, Yeh IT, Ravdin P, Bugarini R, Baehner FL, Davidson NE, Sledge GW, Winer EP, Hudis C, Ingle JN, Perez EA, Pritchard KI, Shepherd L, Gralow JR, Yoshizawa C, Allred DC, Osborne CK, Hayes DF (2010) Prognostic and predictive value of the 21-gene recurrence score assay in postmenopausal women with node-positive, oestrogen-receptor-positive breast cancer on chemotherapy: a retrospective analysis of a randomised trial. Lancet Oncol 11(1):55–65PubMedCentralPubMedCrossRef
3.
Zurück zum Zitat Paik S, Tang G, Shak S, Kim C, Baker J, Kim W, Cronin M, Baehner FL, Watson D, Bryant J, Costantino JP, Geyer CE Jr, Wickerham DL, Wolmark N (2006) Gene expression and benefit of chemotherapy in women with node-negative, estrogen receptor-positive breast cancer. J Clin Oncol 24(23):3726–3734PubMedCrossRef Paik S, Tang G, Shak S, Kim C, Baker J, Kim W, Cronin M, Baehner FL, Watson D, Bryant J, Costantino JP, Geyer CE Jr, Wickerham DL, Wolmark N (2006) Gene expression and benefit of chemotherapy in women with node-negative, estrogen receptor-positive breast cancer. J Clin Oncol 24(23):3726–3734PubMedCrossRef
4.
Zurück zum Zitat Hassett MJ, Silver SM, Hughes ME, Blayney DW, Edge SB, Herman JG, Hudis CA, Marcom PK, Pettinga JE, Share D, Theriault R, Wong YN, Vandergrift JL, Niland JC, Weeks JC (2012) Adoption of gene expression profile testing and association with use of chemotherapy among women with breast cancer. J Clin Oncol 30(18):2218–2226PubMedCentralPubMedCrossRef Hassett MJ, Silver SM, Hughes ME, Blayney DW, Edge SB, Herman JG, Hudis CA, Marcom PK, Pettinga JE, Share D, Theriault R, Wong YN, Vandergrift JL, Niland JC, Weeks JC (2012) Adoption of gene expression profile testing and association with use of chemotherapy among women with breast cancer. J Clin Oncol 30(18):2218–2226PubMedCentralPubMedCrossRef
5.
Zurück zum Zitat Parker JS, Mullins M, Cheang MC, Leung S, Voduc D, Vickery T, Davies S, Fauron C, He X, Hu Z, Quackenbush JF, Stijleman IJ, Palazzo J, Marron JS, Nobel AB, Mardis E, Nielsen TO, Ellis MJ, Perou CM, Bernard PS (2009) Supervised risk predictor of breast cancer based on intrinsic subtypes. J Clin Oncol 27(8):1160–1167PubMedCentralPubMedCrossRef Parker JS, Mullins M, Cheang MC, Leung S, Voduc D, Vickery T, Davies S, Fauron C, He X, Hu Z, Quackenbush JF, Stijleman IJ, Palazzo J, Marron JS, Nobel AB, Mardis E, Nielsen TO, Ellis MJ, Perou CM, Bernard PS (2009) Supervised risk predictor of breast cancer based on intrinsic subtypes. J Clin Oncol 27(8):1160–1167PubMedCentralPubMedCrossRef
6.
Zurück zum Zitat Gianni L, Zambetti M, Clark K, Baker J, Cronin M, Wu J, Mariani G, Rodriguez J, Carcangiu M, Watson D, Valagussa P, Rouzier R, Symmans WF, Ross JS, Hortobagyi GN, Pusztai L, Shak S (2005) Gene expression profiles in paraffin-embedded core biopsy tissue predict response to chemotherapy in women with locally advanced breast cancer. J Clin Oncol 23(29):7265–7277PubMedCrossRef Gianni L, Zambetti M, Clark K, Baker J, Cronin M, Wu J, Mariani G, Rodriguez J, Carcangiu M, Watson D, Valagussa P, Rouzier R, Symmans WF, Ross JS, Hortobagyi GN, Pusztai L, Shak S (2005) Gene expression profiles in paraffin-embedded core biopsy tissue predict response to chemotherapy in women with locally advanced breast cancer. J Clin Oncol 23(29):7265–7277PubMedCrossRef
7.
Zurück zum Zitat Straver ME, Glas AM, Hannemann J, Wesseling J, van de Vijver MJ, Rutgers EJ, Vrancken Peeters MJ, van Tinteren H, Van’t Veer LJ, Rodenhuis S (2010) The 70-gene signature as a response predictor for neoadjuvant chemotherapy in breast cancer. Breast Cancer Res Treat 119(3):551–558PubMedCrossRef Straver ME, Glas AM, Hannemann J, Wesseling J, van de Vijver MJ, Rutgers EJ, Vrancken Peeters MJ, van Tinteren H, Van’t Veer LJ, Rodenhuis S (2010) The 70-gene signature as a response predictor for neoadjuvant chemotherapy in breast cancer. Breast Cancer Res Treat 119(3):551–558PubMedCrossRef
8.
Zurück zum Zitat Denkert C, Loibl S, Muller BM, Eidtmann H, Schmitt WD, Eiermann W, Gerber B, Tesch H, Hilfrich J, Huober J, Fehm T, Barinoff J, Jackisch C, Prinzler J, Rudiger T, Erbstosser E, Blohmer JU, Budczies J, Mehta KM, von Minckwitz G (2013) Ki67 levels as predictive and prognostic parameters in pretherapeutic breast cancer core biopsies: a translational investigation in the neoadjuvant GeparTrio trial. Ann Oncol 24(11):2786–2793PubMedCrossRef Denkert C, Loibl S, Muller BM, Eidtmann H, Schmitt WD, Eiermann W, Gerber B, Tesch H, Hilfrich J, Huober J, Fehm T, Barinoff J, Jackisch C, Prinzler J, Rudiger T, Erbstosser E, Blohmer JU, Budczies J, Mehta KM, von Minckwitz G (2013) Ki67 levels as predictive and prognostic parameters in pretherapeutic breast cancer core biopsies: a translational investigation in the neoadjuvant GeparTrio trial. Ann Oncol 24(11):2786–2793PubMedCrossRef
9.
Zurück zum Zitat Fasching PA, Heusinger K, Haeberle L, Niklos M, Hein A, Bayer CM, Rauh C, Schulz-Wendtland R, Bani MR, Schrauder M, Kahmann L, Lux MP, Strehl JD, Hartmann A, Dimmler A, Beckmann MW, Wachter DL (2011) Ki67, chemotherapy response, and prognosis in breast cancer patients receiving neoadjuvant treatment. BMC Cancer 11:486PubMedCentralPubMedCrossRef Fasching PA, Heusinger K, Haeberle L, Niklos M, Hein A, Bayer CM, Rauh C, Schulz-Wendtland R, Bani MR, Schrauder M, Kahmann L, Lux MP, Strehl JD, Hartmann A, Dimmler A, Beckmann MW, Wachter DL (2011) Ki67, chemotherapy response, and prognosis in breast cancer patients receiving neoadjuvant treatment. BMC Cancer 11:486PubMedCentralPubMedCrossRef
10.
Zurück zum Zitat Niikura N, Iwamoto T, Masuda S, Kumaki N, Xiaoyan T, Shirane M, Mori K, Tsuda B, Okamura T, Saito Y, Suzuki Y, Tokuda Y (2012) Immunohistochemical Ki67 labeling index has similar proliferation predictive power to various gene signatures in breast cancer. Cancer Sci 103(8):1508–1512PubMedCrossRef Niikura N, Iwamoto T, Masuda S, Kumaki N, Xiaoyan T, Shirane M, Mori K, Tsuda B, Okamura T, Saito Y, Suzuki Y, Tokuda Y (2012) Immunohistochemical Ki67 labeling index has similar proliferation predictive power to various gene signatures in breast cancer. Cancer Sci 103(8):1508–1512PubMedCrossRef
11.
Zurück zum Zitat Sheri A, Dowsett M (2012) Developments in Ki67 and other biomarkers for treatment decision making in breast cancer. Ann Oncol 23(Suppl 10):x219–x227PubMedCrossRef Sheri A, Dowsett M (2012) Developments in Ki67 and other biomarkers for treatment decision making in breast cancer. Ann Oncol 23(Suppl 10):x219–x227PubMedCrossRef
12.
Zurück zum Zitat Sueta A, Yamamoto Y, Hayashi M, Yamamoto S, Inao T, Ibusuki M, Murakami K, Iwase H (2014) Clinical significance of pretherapeutic Ki67 as a predictive parameter for response to neoadjuvant chemotherapy in breast cancer; is it equally useful across tumor subtypes? Surgery 155(5):927–935PubMedCrossRef Sueta A, Yamamoto Y, Hayashi M, Yamamoto S, Inao T, Ibusuki M, Murakami K, Iwase H (2014) Clinical significance of pretherapeutic Ki67 as a predictive parameter for response to neoadjuvant chemotherapy in breast cancer; is it equally useful across tumor subtypes? Surgery 155(5):927–935PubMedCrossRef
13.
Zurück zum Zitat Cheang MC, Chia SK, Voduc D, Gao D, Leung S, Snider J, Watson M, Davies S, Bernard PS, Parker JS, Perou CM, Ellis MJ, Nielsen TO (2009) Ki67 index, HER2 status, and prognosis of patients with luminal B breast cancer. J Natl Cancer Inst 101(10):736–750PubMedCentralPubMedCrossRef Cheang MC, Chia SK, Voduc D, Gao D, Leung S, Snider J, Watson M, Davies S, Bernard PS, Parker JS, Perou CM, Ellis MJ, Nielsen TO (2009) Ki67 index, HER2 status, and prognosis of patients with luminal B breast cancer. J Natl Cancer Inst 101(10):736–750PubMedCentralPubMedCrossRef
14.
Zurück zum Zitat Criscitiello C, Disalvatore D, De Laurentiis M, Gelao L, Fumagalli L, Locatelli M, Bagnardi V, Rotmensz N, Esposito A, Minchella I, De Placido S, Santangelo M, Viale G, Goldhirsch A, Curigliano G (2014) High Ki-67 score is indicative of a greater benefit from adjuvant chemotherapy when added to endocrine therapy in Luminal B HER2 negative and node-positive breast cancer. Breast 23(1):69–75PubMedCrossRef Criscitiello C, Disalvatore D, De Laurentiis M, Gelao L, Fumagalli L, Locatelli M, Bagnardi V, Rotmensz N, Esposito A, Minchella I, De Placido S, Santangelo M, Viale G, Goldhirsch A, Curigliano G (2014) High Ki-67 score is indicative of a greater benefit from adjuvant chemotherapy when added to endocrine therapy in Luminal B HER2 negative and node-positive breast cancer. Breast 23(1):69–75PubMedCrossRef
15.
Zurück zum Zitat Klintman M, Strand C, Ahlin C, Beglerbegovic S, Fjallskog ML, Grabau D, Gudlaugsson E, Janssen EA, Lovgren K, Skaland I, Bendahl PO, Malmstrom P, Baak JP, Ferno M (2013) The prognostic value of mitotic activity index (MAI), phosphohistone H3 (PPH3), cyclin B1, cyclin A, and Ki67, alone and in combinations, in node-negative premenopausal breast cancer. PLoS One 8(12):e81902PubMedCentralPubMedCrossRef Klintman M, Strand C, Ahlin C, Beglerbegovic S, Fjallskog ML, Grabau D, Gudlaugsson E, Janssen EA, Lovgren K, Skaland I, Bendahl PO, Malmstrom P, Baak JP, Ferno M (2013) The prognostic value of mitotic activity index (MAI), phosphohistone H3 (PPH3), cyclin B1, cyclin A, and Ki67, alone and in combinations, in node-negative premenopausal breast cancer. PLoS One 8(12):e81902PubMedCentralPubMedCrossRef
16.
Zurück zum Zitat Lende TH, Janssen EA, Gudlaugsson E, Voorhorst F, Smaaland R, van Diest P, Soiland H, Baak JP (2011) In patients younger than age 55 years with lymph node-negative breast cancer, proliferation by mitotic activity index is prognostically superior to adjuvant! J Clin Oncol 29(7):852–858PubMedCrossRef Lende TH, Janssen EA, Gudlaugsson E, Voorhorst F, Smaaland R, van Diest P, Soiland H, Baak JP (2011) In patients younger than age 55 years with lymph node-negative breast cancer, proliferation by mitotic activity index is prognostically superior to adjuvant! J Clin Oncol 29(7):852–858PubMedCrossRef
17.
Zurück zum Zitat Penault-Llorca F, Abrial C, Raoelfils I, Chollet P, Cayre A, Mouret-Reynier MA, Thivat E, Mishellany F, Gimbergues P, Durando X (2008) Changes and predictive and prognostic value of the mitotic index, Ki-67, cyclin D1, and cyclo-oxygenase-2 in 710 operable breast cancer patients treated with neoadjuvant chemotherapy. Oncologist 13(12):1235–1245PubMedCrossRef Penault-Llorca F, Abrial C, Raoelfils I, Chollet P, Cayre A, Mouret-Reynier MA, Thivat E, Mishellany F, Gimbergues P, Durando X (2008) Changes and predictive and prognostic value of the mitotic index, Ki-67, cyclin D1, and cyclo-oxygenase-2 in 710 operable breast cancer patients treated with neoadjuvant chemotherapy. Oncologist 13(12):1235–1245PubMedCrossRef
18.
Zurück zum Zitat Vincent-Salomon A, Rousseau A, Jouve M, Beuzeboc P, Sigal-Zafrani B, Freneaux P, Rosty C, Nos C, Campana F, Klijanienko J, Al Ghuzlan A, Sastre-Garau X (2004) Proliferation markers predictive of the pathological response and disease outcome of patients with breast carcinomas treated by anthracycline-based preoperative chemotherapy. Eur J Cancer 40(10):1502–1508PubMedCrossRef Vincent-Salomon A, Rousseau A, Jouve M, Beuzeboc P, Sigal-Zafrani B, Freneaux P, Rosty C, Nos C, Campana F, Klijanienko J, Al Ghuzlan A, Sastre-Garau X (2004) Proliferation markers predictive of the pathological response and disease outcome of patients with breast carcinomas treated by anthracycline-based preoperative chemotherapy. Eur J Cancer 40(10):1502–1508PubMedCrossRef
20.
Zurück zum Zitat von Minckwitz G, Fontanella C (2013) Selecting the neoadjuvant treatment by molecular subtype: how to maximize the benefit? Breast 22(Suppl 2):S149–S151CrossRef von Minckwitz G, Fontanella C (2013) Selecting the neoadjuvant treatment by molecular subtype: how to maximize the benefit? Breast 22(Suppl 2):S149–S151CrossRef
21.
Zurück zum Zitat Marchio C, Sapino A (2011) The pathologic complete response open question in primary therapy. J Natl Cancer Inst Monogr 43:86–90CrossRef Marchio C, Sapino A (2011) The pathologic complete response open question in primary therapy. J Natl Cancer Inst Monogr 43:86–90CrossRef
22.
Zurück zum Zitat Penault-Llorca F, Abrial C, Raoelfils I, Cayre A, Mouret-Reynier MA, Leheurteur M, Durando X, Achard JL, Gimbergues P, Chollet P (2008) Comparison of the prognostic significance of Chevallier and Sataloff’s pathologic classifications after neoadjuvant chemotherapy of operable breast cancer. Hum Pathol 39(8):1221–1228PubMedCrossRef Penault-Llorca F, Abrial C, Raoelfils I, Cayre A, Mouret-Reynier MA, Leheurteur M, Durando X, Achard JL, Gimbergues P, Chollet P (2008) Comparison of the prognostic significance of Chevallier and Sataloff’s pathologic classifications after neoadjuvant chemotherapy of operable breast cancer. Hum Pathol 39(8):1221–1228PubMedCrossRef
23.
Zurück zum Zitat Elston CW, Ellis IO (1991) Pathological prognostic factors in breast cancer. I. The value of histological grade in breast cancer: experience from a large study with long-term follow-up. Histopathology 19(5):403–410PubMedCrossRef Elston CW, Ellis IO (1991) Pathological prognostic factors in breast cancer. I. The value of histological grade in breast cancer: experience from a large study with long-term follow-up. Histopathology 19(5):403–410PubMedCrossRef
25.
Zurück zum Zitat Prat A, Cheang MC, Martin M, Parker JS, Carrasco E, Caballero R, Tyldesley S, Gelmon K, Bernard PS, Nielsen TO, Perou CM (2013) Prognostic significance of progesterone receptor-positive tumor cells within immunohistochemically defined luminal A breast cancer. J Clin Oncol 31(2):203–209. doi:10.1200/JCO.2012.43.4134 PubMedCentralPubMedCrossRef Prat A, Cheang MC, Martin M, Parker JS, Carrasco E, Caballero R, Tyldesley S, Gelmon K, Bernard PS, Nielsen TO, Perou CM (2013) Prognostic significance of progesterone receptor-positive tumor cells within immunohistochemically defined luminal A breast cancer. J Clin Oncol 31(2):203–209. doi:10.​1200/​JCO.​2012.​43.​4134 PubMedCentralPubMedCrossRef
26.
Zurück zum Zitat Salgado R, Denkert C, Demaria S, Sirtaine N, Klauschen F, Pruneri G, Wienert S, Van den Eynden G, Baehne FL, Penault-Llorca F, Perez EA, Thompson EA, Symmans WF, Richardson AL, Brock J, Criscitiello C, Bailey H, Ignatiadis M, Floris G, Sparano J, Kos Z, Nielsen T, Rimm DL, Allison KH, Reis-Filho JS, Loibl S, Sotiriou C, Viale G, Badve S, Adams S, Willard-Gallo K, Loi S (2014) Harmonization of the evaluation of tumor infiltrating lymphocytes (TILs) in breast cancer: recommendations by an international TILs-working group 2014. Ann Oncol. doi:10.1093/annonc/mdu450 Salgado R, Denkert C, Demaria S, Sirtaine N, Klauschen F, Pruneri G, Wienert S, Van den Eynden G, Baehne FL, Penault-Llorca F, Perez EA, Thompson EA, Symmans WF, Richardson AL, Brock J, Criscitiello C, Bailey H, Ignatiadis M, Floris G, Sparano J, Kos Z, Nielsen T, Rimm DL, Allison KH, Reis-Filho JS, Loibl S, Sotiriou C, Viale G, Badve S, Adams S, Willard-Gallo K, Loi S (2014) Harmonization of the evaluation of tumor infiltrating lymphocytes (TILs) in breast cancer: recommendations by an international TILs-working group 2014. Ann Oncol. doi:10.​1093/​annonc/​mdu450
27.
Zurück zum Zitat Faraggi D (2000) The effect of random measurement error on receiver operating characteristic (ROC) curves. Stat Med 19(1):61–70PubMedCrossRef Faraggi D (2000) The effect of random measurement error on receiver operating characteristic (ROC) curves. Stat Med 19(1):61–70PubMedCrossRef
28.
Zurück zum Zitat Reiser B (2000) Measuring the effectiveness of diagnostic markers in the presence of measurement error through the use of ROC curves. Stat Med 19(16):2115–2129PubMedCrossRef Reiser B (2000) Measuring the effectiveness of diagnostic markers in the presence of measurement error through the use of ROC curves. Stat Med 19(16):2115–2129PubMedCrossRef
29.
Zurück zum Zitat McShane LM, Altman DG, Sauerbrei W, Taube SE, Gion M, Clark GM (2006) Reporting recommendations for tumor marker prognostic studies (REMARK). Breast Cancer Res Treat 100(2):229–235PubMedCrossRef McShane LM, Altman DG, Sauerbrei W, Taube SE, Gion M, Clark GM (2006) Reporting recommendations for tumor marker prognostic studies (REMARK). Breast Cancer Res Treat 100(2):229–235PubMedCrossRef
30.
Zurück zum Zitat Cocquyt VF, Blondeel PN, Depypere HT, Praet MM, Schelfhout VR, Silva OE, Hurley J, Serreyn RF, Daems KK, Van Belle SJ (2003) Different responses to preoperative chemotherapy for invasive lobular and invasive ductal breast carcinoma. Eur J Surg Oncol 29(4):361–367PubMedCrossRef Cocquyt VF, Blondeel PN, Depypere HT, Praet MM, Schelfhout VR, Silva OE, Hurley J, Serreyn RF, Daems KK, Van Belle SJ (2003) Different responses to preoperative chemotherapy for invasive lobular and invasive ductal breast carcinoma. Eur J Surg Oncol 29(4):361–367PubMedCrossRef
31.
Zurück zum Zitat Cristofanilli M, Gonzalez-Angulo A, Sneige N, Kau SW, Broglio K, Theriault RL, Valero V, Buzdar AU, Kuerer H, Buchholz TA, Hortobagyi GN (2005) Invasive lobular carcinoma classic type: response to primary chemotherapy and survival outcomes. J Clin Oncol 23(1):41–48PubMedCrossRef Cristofanilli M, Gonzalez-Angulo A, Sneige N, Kau SW, Broglio K, Theriault RL, Valero V, Buzdar AU, Kuerer H, Buchholz TA, Hortobagyi GN (2005) Invasive lobular carcinoma classic type: response to primary chemotherapy and survival outcomes. J Clin Oncol 23(1):41–48PubMedCrossRef
32.
Zurück zum Zitat Delpech Y, Coutant C, Hsu L, Barranger E, Iwamoto T, Barcenas CH, Hortobagyi GN, Rouzier R, Esteva FJ, Pusztai L (2013) Clinical benefit from neoadjuvant chemotherapy in oestrogen receptor-positive invasive ductal and lobular carcinomas. Br J Cancer 108(2):285–291PubMedCentralPubMedCrossRef Delpech Y, Coutant C, Hsu L, Barranger E, Iwamoto T, Barcenas CH, Hortobagyi GN, Rouzier R, Esteva FJ, Pusztai L (2013) Clinical benefit from neoadjuvant chemotherapy in oestrogen receptor-positive invasive ductal and lobular carcinomas. Br J Cancer 108(2):285–291PubMedCentralPubMedCrossRef
33.
Zurück zum Zitat Loibl S, Volz C, Mau C, Blohmer JU, Costa SD, Eidtmann H, Fasching PA, Gerber B, Hanusch C, Jackisch C, Kummel S, Huober J, Denkert C, Hilfrich J, Konecny GE, Fett W, Stickeler E, Harbeck N, Mehta KM, Nekljudova V, von Minckwitz G, Untch M (2014) Response and prognosis after neoadjuvant chemotherapy in 1,051 patients with infiltrating lobular breast carcinoma. Breast Cancer Res Treat 144(1):153–162PubMedCrossRef Loibl S, Volz C, Mau C, Blohmer JU, Costa SD, Eidtmann H, Fasching PA, Gerber B, Hanusch C, Jackisch C, Kummel S, Huober J, Denkert C, Hilfrich J, Konecny GE, Fett W, Stickeler E, Harbeck N, Mehta KM, Nekljudova V, von Minckwitz G, Untch M (2014) Response and prognosis after neoadjuvant chemotherapy in 1,051 patients with infiltrating lobular breast carcinoma. Breast Cancer Res Treat 144(1):153–162PubMedCrossRef
34.
Zurück zum Zitat Petrelli F, Barni S (2013) Response to neoadjuvant chemotherapy in ductal compared to lobular carcinoma of the breast: a meta-analysis of published trials including 1,764 lobular breast cancer. Breast Cancer Res Treat 142(2):227–235PubMedCrossRef Petrelli F, Barni S (2013) Response to neoadjuvant chemotherapy in ductal compared to lobular carcinoma of the breast: a meta-analysis of published trials including 1,764 lobular breast cancer. Breast Cancer Res Treat 142(2):227–235PubMedCrossRef
35.
Zurück zum Zitat Denkert C, Loibl S, Noske A, Roller M, Muller BM, Komor M, Budczies J, Darb-Esfahani S, Kronenwett R, Hanusch C, von Torne C, Weichert W, Engels K, Solbach C, Schrader I, Dietel M, von Minckwitz G (2010) Tumor-associated lymphocytes as an independent predictor of response to neoadjuvant chemotherapy in breast cancer. J Clin Oncol 28(1):105–113PubMedCrossRef Denkert C, Loibl S, Noske A, Roller M, Muller BM, Komor M, Budczies J, Darb-Esfahani S, Kronenwett R, Hanusch C, von Torne C, Weichert W, Engels K, Solbach C, Schrader I, Dietel M, von Minckwitz G (2010) Tumor-associated lymphocytes as an independent predictor of response to neoadjuvant chemotherapy in breast cancer. J Clin Oncol 28(1):105–113PubMedCrossRef
36.
Zurück zum Zitat Gu-Trantien C, Loi S, Garaud S, Equeter C, Libin M, de Wind A, Ravoet M, Le Buanec H, Sibille C, Manfouo-Foutsop G, Veys I, Haibe-Kains B, Singhal SK, Michiels S, Rothe F, Salgado R, Duvillier H, Ignatiadis M, Desmedt C, Bron D, Larsimont D, Piccart M, Sotiriou C, Willard-Gallo K (2013) CD4(+) follicular helper T cell infiltration predicts breast cancer survival. J Clin Investig 123(7):2873–2892PubMedCentralPubMedCrossRef Gu-Trantien C, Loi S, Garaud S, Equeter C, Libin M, de Wind A, Ravoet M, Le Buanec H, Sibille C, Manfouo-Foutsop G, Veys I, Haibe-Kains B, Singhal SK, Michiels S, Rothe F, Salgado R, Duvillier H, Ignatiadis M, Desmedt C, Bron D, Larsimont D, Piccart M, Sotiriou C, Willard-Gallo K (2013) CD4(+) follicular helper T cell infiltration predicts breast cancer survival. J Clin Investig 123(7):2873–2892PubMedCentralPubMedCrossRef
37.
38.
Zurück zum Zitat Loi S, Sirtaine N, Piette F, Salgado R, Viale G, Van Eenoo F, Rouas G, Francis P, Crown JP, Hitre E, de Azambuja E, Quinaux E, Di Leo A, Michiels S, Piccart MJ, Sotiriou C (2013) Prognostic and predictive value of tumor-infiltrating lymphocytes in a phase III randomized adjuvant breast cancer trial in node-positive breast cancer comparing the addition of docetaxel to doxorubicin with doxorubicin-based chemotherapy: BIG 02-98. J Clin Oncol 31(7):860–867PubMedCrossRef Loi S, Sirtaine N, Piette F, Salgado R, Viale G, Van Eenoo F, Rouas G, Francis P, Crown JP, Hitre E, de Azambuja E, Quinaux E, Di Leo A, Michiels S, Piccart MJ, Sotiriou C (2013) Prognostic and predictive value of tumor-infiltrating lymphocytes in a phase III randomized adjuvant breast cancer trial in node-positive breast cancer comparing the addition of docetaxel to doxorubicin with doxorubicin-based chemotherapy: BIG 02-98. J Clin Oncol 31(7):860–867PubMedCrossRef
39.
Zurück zum Zitat Loi S, Michiels S, Salgado R, Sirtaine N, Jose V, Fumagalli D, Kellokumpu-Lehtinen PL, Bono P, Kataja V, Desmedt C, Piccart MJ, Loibl S, Denkert C, Smyth MJ, Joensuu H, Sotiriou C (2014) 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):1544–1550PubMedCrossRef Loi S, Michiels S, Salgado R, Sirtaine N, Jose V, Fumagalli D, Kellokumpu-Lehtinen PL, Bono P, Kataja V, Desmedt C, Piccart MJ, Loibl S, Denkert C, Smyth MJ, Joensuu H, Sotiriou C (2014) 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):1544–1550PubMedCrossRef
40.
Zurück zum Zitat Vinayak S, Gray RJ, Adams S, Jensen KC, Manola J, Afghahi A, Goldstein LJ, Ford JM, Badve SS, Telli ML (2014) Association of increased tumor-infiltrating lymphocytes (TILs) with immunomodulatory (IM) triple-negative breast cancer (TNBC) subtype and response to neoadjuvant platinum-based therapy in PrECOG0105. J Clin Oncol 32:S1000 Vinayak S, Gray RJ, Adams S, Jensen KC, Manola J, Afghahi A, Goldstein LJ, Ford JM, Badve SS, Telli ML (2014) Association of increased tumor-infiltrating lymphocytes (TILs) with immunomodulatory (IM) triple-negative breast cancer (TNBC) subtype and response to neoadjuvant platinum-based therapy in PrECOG0105. J Clin Oncol 32:S1000
41.
Zurück zum Zitat Turner N, Biganzoli L, Malorni L, Migliaccio I, Moretti E, Pestrin M, Sanna G, Siclari O, Di Leo A (2013) Adjuvant chemotherapy: which patient? What regimen? Am Soc Clin Oncol Educ Book 4:3-8 Turner N, Biganzoli L, Malorni L, Migliaccio I, Moretti E, Pestrin M, Sanna G, Siclari O, Di Leo A (2013) Adjuvant chemotherapy: which patient? What regimen? Am Soc Clin Oncol Educ Book 4:3-8
42.
Zurück zum Zitat Dowsett M, Nielsen TO, A’Hern R, Bartlett J, Coombes RC, Cuzick J, Ellis M, Henry NL, Hugh JC, Lively T, McShane L, Paik S, Penault-Llorca F, Prudkin L, Regan M, Salter J, Sotiriou C, Smith IE, Viale G, Zujewski JA, Hayes DF (2011) Assessment of Ki67 in breast cancer: recommendations from the international Ki67 in breast cancer working group. J Natl Cancer Inst 103(22):1656–1664PubMedCentralPubMedCrossRef Dowsett M, Nielsen TO, A’Hern R, Bartlett J, Coombes RC, Cuzick J, Ellis M, Henry NL, Hugh JC, Lively T, McShane L, Paik S, Penault-Llorca F, Prudkin L, Regan M, Salter J, Sotiriou C, Smith IE, Viale G, Zujewski JA, Hayes DF (2011) Assessment of Ki67 in breast cancer: recommendations from the international Ki67 in breast cancer working group. J Natl Cancer Inst 103(22):1656–1664PubMedCentralPubMedCrossRef
43.
Zurück zum Zitat Luporsi E, Andre F, Spyratos F, Martin PM, Jacquemier J, Penault-Llorca F, Tubiana-Mathieu N, Sigal-Zafrani B, Arnould L, Gompel A, Egele C, Poulet B, Clough KB, Crouet H, Fourquet A, Lefranc JP, Mathelin C, Rouyer N, Serin D, Spielmann M, Haugh M, Chenard MP, Brain E, de Cremoux P, Bellocq JP (2012) Ki-67: level of evidence and methodological considerations for its role in the clinical management of breast cancer: analytical and critical review. Breast Cancer Res Treat 132(3):895–915PubMedCentralPubMedCrossRef Luporsi E, Andre F, Spyratos F, Martin PM, Jacquemier J, Penault-Llorca F, Tubiana-Mathieu N, Sigal-Zafrani B, Arnould L, Gompel A, Egele C, Poulet B, Clough KB, Crouet H, Fourquet A, Lefranc JP, Mathelin C, Rouyer N, Serin D, Spielmann M, Haugh M, Chenard MP, Brain E, de Cremoux P, Bellocq JP (2012) Ki-67: level of evidence and methodological considerations for its role in the clinical management of breast cancer: analytical and critical review. Breast Cancer Res Treat 132(3):895–915PubMedCentralPubMedCrossRef
44.
Zurück zum Zitat Polley MY, Leung SC, McShane LM, Gao D, Hugh JC, Mastropasqua MG, Viale G, Zabaglo LA, Penault-Llorca F, Bartlett JM, Gown AM, Symmans WF, Piper T, Mehl E, Enos RA, Hayes DF, Dowsett M, Nielsen TO (2013) An international Ki67 reproducibility study. J Natl Cancer Inst 105(24):1897–1906PubMedCrossRef Polley MY, Leung SC, McShane LM, Gao D, Hugh JC, Mastropasqua MG, Viale G, Zabaglo LA, Penault-Llorca F, Bartlett JM, Gown AM, Symmans WF, Piper T, Mehl E, Enos RA, Hayes DF, Dowsett M, Nielsen TO (2013) An international Ki67 reproducibility study. J Natl Cancer Inst 105(24):1897–1906PubMedCrossRef
45.
Zurück zum Zitat Lehr HA, Rochat C, Schaper C, Nobile A, Shanouda S, Vijgen S, Gauthier A, Obermann E, Leuba S, Schmidt M, CCR, Delaloye JF, Simiantonaki N, Schaefer SC (2013) Mitotic figure counts are significantly overestimated in resection specimens of invasive breast carcinomas. Mod Pathol 26(3):336–342PubMedCrossRef Lehr HA, Rochat C, Schaper C, Nobile A, Shanouda S, Vijgen S, Gauthier A, Obermann E, Leuba S, Schmidt M, CCR, Delaloye JF, Simiantonaki N, Schaefer SC (2013) Mitotic figure counts are significantly overestimated in resection specimens of invasive breast carcinomas. Mod Pathol 26(3):336–342PubMedCrossRef
Metadaten
Titel
Pathological non-response to chemotherapy in a neoadjuvant setting of breast cancer: an inter-institutional study
verfasst von
D. Balmativola
C. Marchiò
M. Maule
L. Chiusa
L. Annaratone
F. Maletta
F. Montemurro
J. Kulka
P. Figueiredo
Z. Varga
I. Liepniece-Karele
G. Cserni
E. Arkoumani
I. Amendoeira
G. Callagy
A. Reiner-Concin
A. Cordoba
S. Bianchi
T. Decker
D. Gläser
C. Focke
P. van Diest
D. Grabau
E. Lips
J. Wesseling
R. Arisio
E. Medico
C. Wells
A. Sapino
Publikationsdatum
01.12.2014
Verlag
Springer US
Erschienen in
Breast Cancer Research and Treatment / Ausgabe 3/2014
Print ISSN: 0167-6806
Elektronische ISSN: 1573-7217
DOI
https://doi.org/10.1007/s10549-014-3192-3

Weitere Artikel der Ausgabe 3/2014

Breast Cancer Research and Treatment 3/2014 Zur Ausgabe

Update Onkologie

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