Skip to main content
Erschienen in: Breast Cancer Research and Treatment 1/2016

Open Access 18.06.2016 | Epidemiology

Extent of ductal carcinoma in situ according to breast cancer subtypes: a population-based cohort study

verfasst von: Shusma C. Doebar, Esther C. van den Broek, Linetta B. Koppert, Agnes Jager, Margreet. H. A. Baaijens, Inge-Marie A. M. Obdeijn, Carolien H. M. van Deurzen

Erschienen in: Breast Cancer Research and Treatment | Ausgabe 1/2016

Abstract

Ductal carcinoma in situ (DCIS) is a precursor of invasive breast carcinoma (IBC). The DCIS component is often more extensive than the invasive component, which affects local control. The aim of our study was to analyze features of DCIS within different IBC subtypes, which may contribute to the optimization of personalized approaches for patients with IBC. Patients with IBC reported according to the synoptic reporting module in the Netherlands between 2009 and 2015 were included. Data extraction included characteristics of the invasive component and, if present, several features of the DCIS component. Resection margin status analyses were restricted to patients undergoing breast-conserving surgery (BCS). Differences between subtypes were tested by a Chi-square test, spearman’s Rho test or a one-way ANOVA test. Overall, 36.937 cases of IBC were included. About half of the IBCs (n = 16.014; 43.4 %) were associated with DCIS. Her2+ IBC (irrespective of ER status) was associated with a higher prevalence of adjacent DCIS, a larger extent of DCIS and a higher rate of irradicality of the DCIS component as compared to ER+/Her2− and triple-negative subtypes (P < 0.0001 for all variables). The prevalence of DCIS in triple-negative IBC on the other hand was lowest. In this large population-based cohort study, we showed significant differences between the prevalence and extent of DCIS according to IBC subtypes, which is also reflected in the resection margin status in patients treated with BCS. Our data provide important information regarding the optimization of local therapy according to IBC subtypes.

Introduction

Invasive breast cancer (IBC) is a heterogeneous disease which can be categorized into several histologic or intrinsic subtypes that differ in their biological behavior and clinical outcome [13]. Intrinsic subtypes are most precisely categorized based on multigene expression assays, although each subtype has an immunohistochemical surrogate based on ER, PR, Her2, and Ki-67 index [47]. Ductal carcinoma in situ (DCIS) is seen as a nonobligate precursor of invasive ductal carcinoma (IDC). In the last decades, the detection rate of DCIS increased markedly in the age group of 50–75 years, as a result of the increased use and improved resolution of mammographic mass screening [8, 9]. Synchronous DCIS and adjacent IDC show a high degree of concordance regarding morphology and genetic profiles [1015]. The concordance of receptor expression of ER, PR, and Her2 in DCIS and coexisting IDC is high, with 92 % for ER, 93–97 % for PR, and about 98–100 % for Her2 [1012].
Data regarding the process of progression of DCIS to IBC is limited. Several studies reported frequencies of pure DCIS subtypes based on immunohistochemical surrogates originally described for IBC [5, 13, 14, 16]. In these pure DCIS studies, the distribution of subtypes differs from studies including IBC. In pure DCIS studies, frequencies of Her2−positive subtypes are higher as compared to reported frequencies in IBC; about 15–32 % of pure DCIS cases are Her2 positive, while this frequency is lower in IBC, about 6–14 % [5, 13, 14, 1618]. Reported frequencies of triple-negative pure DCIS on the other hand are lower than reported frequencies in IBC, 6–8 % in pure DCIS versus 11–13 % in IBC [5, 13, 14, 16, 17]. Regarding the Luminal A and Luminal B subgroups, the reported frequencies for pure DCIS and IBC are overlapping (38–63 % in pure DCIS versus 38–73 % in IBC for luminal A and 7–28 % in pure DCIS versus 5–26 % in IBC for luminal B) [5, 13, 14, 16, 17, 19]. Based on these prevalences, a mathematical, hypothetical model has been built, suggesting different speeds of progression according to breast cancer subtypes [20]. This model suggests that Her2+ DCIS has the slowest progression to IBC, while triple-negative DCIS has the fastest progression.
Since the last decades, the proportion of patients undergoing mastectomy decreased and the majority of patients with localized DCIS are treated with breast-conserving surgery (BCS), followed by breast irradiation [21]. Overall, the local recurrence rate (LRR) for patients with DCIS treated with BCS followed by breast irradiation is about 10–17 % within the first 15 years after treatment, of which 50 % concerns IBC [2224]. Recent studies reported that DCIS subtype was an independent predictor for ipsilateral recurrence after treatment by breast surgery alone (BCS or mastectomy) or breast surgery followed by breast irradiation [2528]. The overall LRR in patients with pure DCIS was the highest in Her2-positive and luminal B subgroups (10–48 % and 25–42 % recurring within 10 years of follow-up, respectively) and the lowest in the luminal A subgroup (9–21 %) [2527]. Regarding triple-negative DCIS, no firm conclusion could be drawn from the reported LRRs due to limited numbers of patients. Nevertheless, based on LRRs per subtype, Her2-positive DCIS seems to have an increased risk for LR after breast surgery as compared to Her2-negative DCIS. In line with this, the highest LRR was also observed for Her2−positive IBC following breast surgery and irradiation (LRR of 8–21 % within 10 years of follow-up), as compared to Luminal A and Luminal B type IBC (LRR 1–8 % and 2–10 % respectively) [2931]. These data suggest that adjustment of current treatment guidelines according to breast cancer subtypes, e.g., aggressive local therapy restricted to patients with a high LRR, could result in reduction of complications and costs for low risk patients.
Subtyping of DCIS has the potential to study progression-related features and to identify patients at high risk for LR. However, in daily practice, pure DCIS cases are not routinely analyzed for ER, PR and Her2 status, which limits the opportunity for large-scale retrospective studies. Patients with IBC on the other hand are routinely studied for ER, PR, and Her2 status. This provides the opportunity to indirectly assess adjacent DCIS features, which, as mentioned above, share receptor expression pattern in the vast majority of cases. The aim of this study was to analyze features of DCIS within different IBC subtypes, including the resection margin status in patients treated with BCS, which may contribute to the optimization of personalized approaches for patients with IBC.

Patients and methods

Data acquisition

In the Netherlands, all pathology reports are archived in the Dutch Pathology Registry (PALGA) [32]. Since 2009, synoptic reporting modules for reporting several common tumor types including breast cancer became available. In these modules, the parameters are captured in numerous variables instead of free text fields. This offers the unique opportunity to analyze all reports created with the module simultaneously.

Patient and tumor characteristics

For this study, we included all patients with IBC reported according to the protocol module in the Netherlands between January 1, 2009 and September 1, 2015 (n = 36.937 cases). Patients with missing ER, PR, and/or Her2 status; pure DCIS; and patients with IBC after previous treatment (irradical resection, neoadjuvant therapy) were excluded. Patients with bilateral IBC were included as two cases. In case of multiple IBCs in one breast, the largest IBC was included for analysis of tumor characteristics, except for resection margin status, which was assessed for all tumors.
Clinicopathologic characteristics included age, type of surgical procedure (BCS or mastectomy), tumor size (≤2 cm, >2 to ≤5 cm or >5 cm), histological type (according to WHO), grade (according to the modified Bloom and Richardson grading system) [33], ER status, PR status, Her2 status, presence of angioinvasion, presence of DCIS, and nodal status. ER status and PR status were defined as positive in case more than 10 % of the cancer cells that showed nuclear staining, irrespective of density, according to the Dutch Guideline for breast cancer treatment [34]. Her2 status was scored according to the international guidelines [35]. Based on immunohistochemistry, tumors were divided according to the surrogate definitions of intrinsic subtypes as reported in the St Gallen International Expert Consensus 2013 [36]. Low PR expression was defined as ≤20 % [37]. However, the absence of information regarding Ki-67 indexes in our dataset limited the ability to differentiate between Luminal A and Luminal B (Her2−) subtypes, so based on the available information, our cases were subtyped according to the following 5 categories:
1.
ER+/PR high/Her2−,
 
2.
ER+/PR− or low/Her2−,
 
3.
ER+/Her2+,
 
4.
ER−/PR−/Her2+, and
 
5.
ER−/PR−/Her2−.
 
In case DCIS was present, the following features were documented: relation to the invasive component (restricted to invasive component or not), diameter, nuclear grade, and presence of microcalcifications [38]. The overall resection margin status was reported, as well as the margin for both the invasive component and the DCIS component as either free, focally irradical, or more than focally irradical, according to the Dutch Guideline for Breast Cancer Treatment [34]. Focally irradical is defined as tumor (either invasive or DCIS) reaching the ink in a small area (≤4 mm). In case the tumor (either invasive or DCIS) reaches the ink in a larger area or multiple smaller areas, it is defined as more than focally irradical. This distinction has important clinical consequences in the Netherlands, since patients with a focally positive resection margin of IBC or adjacent DCIS do not undergo second surgery (since radiation with a boost dose results in adequate local control), while patients with a more than focally positive resection margin undergo reexcision, according to the Dutch Guideline for Breast Cancer Treatment 2002 [39]. However, these definitions are not applied in most other European and North American countries [40]. Therefore, in this study, we use the term irradicality to describe either focally or more than focally irradical resection margins.

Statistical analysis

Differences between IBC subtypes were tested by means of a Chi-square test (categorical variables) or a one-way ANOVA (continuous variables). Missing values are included in the tables but excluded in the analyses.
Furthermore, the correlation between grade of the invasive component and the DCIS component was tested with Chi-square. The correlation between the extent of the DCIS component and resection margin status of the DCIS component was tested with a spearman’s correlation coefficient. All analyses were performed with SAS Enterprise Guide 7.1.

Results

Baseline characteristics

Overall, we included 36.937 consecutive cases of IBC reported between January 1, 2009 and September 1, 2015. The median age of our patient cohort was 62 years (range 18–100). The majority of patients (60.4 %) underwent BCS. Table 1 provides an overview of clinicopathologic data of all patients. About half of the IBCs (n = 16.014; 43.4 %) were associated with DCIS, either restricted within or outside the invasive component (45.3 and 54.7 %, respectively).
Table 1
Baseline characteristics of all patients with IBC (n = 36937)
Characteristic
N
(%)
Age at diagnosis, years, mean, median (range)
Mean: 61.0
Median: 62.0 (18–100)
 
Type of surgery
 Breast-conserving surgery
22,328
60.45
 Mastectomy
14,609
39.55
Histologic tumor type
 Ductal
29,630
80.22
 Lobular
4703
12.73
 Other
2604
7.05
Tumor size
 ≤2 cm
24,359
65.95
 >2 to ≤5 cm
11,117
30.10
 >5 cm
1461
3.96
Tumor grade
 1
8622
27.13
 2
14,894
46.86
 3
8266
26.01
 Missing
5155
ER status
 Positive
31,662
85.72
 Negative
5275
14.28
PR status
 Positive
25,400
68.77
 Negative
11,487
31.10
Her2 status
 Positive
4140
11.21
 Negative
32,797
88.79
Multiple invasive tumors
 Yes
2650
8.63
 No
28,051
91.37
 Missing
6236
 
Angioinvasion
 Yes
3715
14.03
 No
22,773
85.97
 Missing
10,449
Presence of DCIS component
 Yes
16,014
43.35
 No
20,923
56.65
Overall resection margin status (invasive component and/or DCIS component)a
 Free
18,552
83.09
 Focally irradical
2286
10.24
 More than focally irradical
1490
6.67
Resection margin status of invasive component onlya
 Free
19,755
88.48
 Focally irradical
1621
7.26
 More than focally irradical
952
4.26
Nodal status
 Negative
11,428
60.22
 Positive
7550
39.78
 Missing
17,959
aAnalysis restricted to patients with BCS (n = 22328)
Table 2 provides details of all patients with IBC and adjacent DCIS. Overall, there was a strong correlation between grade of the DCIS component and grade of the invasive component (p < 0.0001, Chi-square test). Both the extent of DCIS and DCIS extending beyond the invasive component correlated with irradicality of the DCIS component (spearman’s rho = 0.3, p < 0.0001 and P < 0.0001, Chi-square test, respectively). The frequency of multiple IBCs was significantly higher in IBC cases with adjacent DCIS (10.2 %) as compared to IBC cases without adjacent DCIS (7.4 %) (p < 0.0001, Chi-square test).
Table 2
DCIS characteristics of all patients with IBC and adjacent DCIS (n = 16014)
Characteristic
N
(%)
DCIS grade
 1
2598
16.33
 2
7896
49.64
 3
5414
34.03
 Missing
106
DCIS restricted to invasive component
 Yes
4452
45.29
 No
5377
54.71
 Missing
6185
Diameter of DCIS, cm, mean, median (range)
Mean: 2.08
Median: 1.50 (0–20)
Presence of DCIS-associated microcalcifications
 Yes
4400
49.97
 No
4406
50.03
 Missing
7208
Resection margin status of DCIS component onlya
 Free
8323
83.67
 Focally irradical
1168
11.74
 More than focally irradical
456
4.58
 Missing
34
 
aAnalysis restricted to patients with BCS (n = 9981)

Clinicopathologic features according to breast cancer subtypes

Based on immunohistochemical stainings, IBCs were categorized into the following 5 categories: ER+/PR high/Her2− (n = 21315; 57.7 %), ER+/PR− or low/Her2− (n = 7541; 20.4 %), ER+/Her2+ (n = 2806; 7.6 %), ER−/PR−/Her2+ (n = 1334; 3.6 %), or ER−/PR−/Her2− (n = 3941; 10.7 %). Table 3 provides an overview of patient and tumor characteristics according to different IBC subtypes.
Table 3
Clinicopathologic characteristics according to different subtypes of IBC (n = 36937)
Characteristic
ER+, PR high, Her2−
(n = 21315)
ER+, PR− or low
Her2−
(n = 7541)
ER+, Any PR,
Her2+
(n = 2806)
ER−, PR−,
Her2+
(n = 1334)
ER−, PR−,
Her2−
(n = 3941)
P-value
Age at diagnosis, years, mean, median and range
Mean: 61.1
Median: 62
Range: 18–99
Mean: 63.4
Median: 64
Range: 21–97
Mean: 57.6
Median: 57
Range: 19–100
Mean: 59.3
Median: 59 Range: 24–97
Mean: 59.0;
Median: 59
Range: 22–98
<0.0001
Type of surgery, no (%)
<0.0001
 Breast-conserving surgery
13,507
63.37
4476
59.36
1514
53.96
599
44.90
2232
56.64
 Mastectomy
7808
36.63
3065
40.64
1292
46.04
735
55.10
1709
43.36
Tumor type, no (%)
<0.0001
 Ductal
16,695
78.33
5753
76.29
2548
90.81
1255
94.08
3379
85.74
 Lobular
3130
14.68
1319
17.49
147
5.24
20
1.50
87
2.21
 Other
1490
6.99
469
6.22
111
3.96
59
4.42
475
12.05
Tumor size, no (%)
<0.0001
 ≤2 cm
14,849
69.66
4931
65.39
1719
61.26
729
54.65
2131
54.07
 >2 to ≤5 cm
5771
27.07
2267
30.06
964
34.35
528
39.58
1587
40.27
 >5 cm
695
3.26
343
4.55
123
4.38
77
5.77
223
5.66
Tumor grade, no (%)
<0.0001
 1
6513
35.17
1786
27.41
203
8.63
29
2.62
91
2.77
 2
9559
51.62
3379
51.86
1018
43.28
273
24.66
665
20.21
 3
2445
13.20
1351
20.73
1131
48.09
805
72.72
2534
77.02
 Missing
2798
1025
454
227
651
Multiple invasive tumors, no (%)
<0.0001
 Yes
1624
9.02
510
8.25
216
9.45
106
9.97
194
5.97
 No
16,296
90.94
5671
91.75
2070
90.55
957
90.03
3057
94.03
 Missing
3395
1360
520
271
690
Angioinvasion, no (%)
<0.0001
 Yes
1734
11.20
727
13.46
427
21.72
267
29.28
560
20.52
 No
13,747
88.80
4673
86.54
1539
78.28
645
70.72
2169
79.48
 Missing
5834
2141
840
422
1212
Overall resection margin status (invasive and/or DCIS component)a
<0.0001
 Free
11,243
83.24
3676
82.13
1205
79.59
459
76.63
1969
88.22
 Focally irradical
1366
10.11
492
10.99
178
11.76
92
15.36
158
7.08
 More than focally irradical
898
6.65
308
6.88
131
8.65
48
8.01
105
4.70
Resection margin status of invasive componenta, no (%)
<0.0001
 Free
11,879
87.95
3910
87.35
1344
88.77
540
90.15
2082
93.28
 Focally irradical
1028
7.61
356
7.95
112
7.40
41
6.84
84
3.76
 More than focally irradical
600
4.44
210
4.69
58
3.83
18
3.01
66
2.96
Nodal status, no (%)
<0.0001
 Negative
6717
62.17
2293
59.19
774
53.20
359
47.61
1285
61.45
 Positive
4087
37.83
1581
40.81
681
46.80
395
52.39
806
38.55
 Missing
10,511
3667
1351
580
1850
aAnalysis restricted to patients with BCS (n = 22328)
Overall, regarding the invasive component, the ER−/Her2+ and triple-negative subgroups showed the most aggressive biological features. The ER+/Her2− subgroups showed the most favorable biological features while the ER+/Her2+ subgroup showed intermediate results. Regarding the ER+/Her2− subgroups, the presence of a high PR expression was associated with more favorable tumor characteristics as compared to those cases with absence or low PR expression.
In general, patients with Her2+ (irrespective of ER status) and triple-negative IBC were younger as compared to patients with ER+/Her2− IBC (P < 0.0001). Besides, median tumor size of these subtypes was larger (P < 0.0001), which was in line with the higher proportion of patients undergoing a mastectomy (P < 0.0001). Histologically, these tumors were more often of ductal type (p < 0.0001) and of higher grade (P < 0.0001). The frequency of angioinvasion and nodal involvement was highest in the ER−/Her2+ subgroup (P < 0.0001).
There was a strong correlation between the presence of DCIS and breast cancer subtype (P < 0.0001). Table 4 provides an overview of all DCIS characteristics according to different subtypes of IBC. DCIS was most often present adjacent to IBCs with overexpression of Her2 (irrespective of ER status) with a frequency of 59.1 % in the ER+/Her2+ subgroup and 57.4 % in the ER−/Her2+ subgroup. The frequency of a DCIS component was lowest in the triple-negative subgroup (34.1 %).
Table 4
DCIS characteristics according to different subtypes of IBC (n = 16014)
Characteristic
ER+, PR high, Her2−
(n = 21315)
ER+, PR− or low
Her2−
(n = 7541)
ER+, Any PR,
Her2+
(n = 2806)
ER−, PR−,
Her2+
(n = 1334)
ER−, PR−,
Her2−
(n = 3941)
P-value
Presence of DCIS, no (%)
<0.0001
 Yes
9168
43.01
3078
40.82
1658
59.09
766
57.42
1344
34.10
 No
12,147
56.99
4463
59.18
1148
40.91
568
42.58
2597
65.90
DCIS grade, no (%)
<0.0001
 1
1983
21.77
512
16.72
61
3.70
4
0.53
38
2.86
 2
5310
58.32
1613
52.66
572
34.73
106
13.93
294
21.11
 3
1813
19.91
938
30.62
1014
61.57
651
85.55
998
75.04
 Missing
61
15
11
5
14
Presence of DCIS-associated microcalcifications, no (%)
<0.0001
 Yes
2464
48.21
876
52.02
512
59.26
240
60.91
308
40.90
 No
2647
51.79
808
47.98
352
40.74
154
39.09
445
59.10
 Missing
4057
1394
794
372
591
DCIS restricted to invasive component, no (%)
<0.0001
 Yes
2774
48.52
823
43.82
383
39.24
127
29.13
345
41.97
 No
2943
51.48
1055
56.18
593
60.76
309
70.87
477
58.03
 Missing
3451
1200
682
330
522
Diameter of DCIS, cm, mean, median and range
Mean: 1.9; Median: 1.4 Range: 0–20
Mean: 1.9; Median: 1.4 Range: 0–19
Mean: 2.6; Median: 2.0 Range: 0–20
Mean: 3.2; Median: 2.3 Range: 0–20
Mean: 2.1; Median: 1.5 Range: 0–15
<0.0001
Resection margin status of DCIS componenta
 
 Free
5103
85.33
1622
83.44
707
78.82
251
71.10
640
82.79
<0.0001
 Focally  irradical
640
10.70
245
12.60
127
14.16
73
20.68
83
10.74
 More than  focally  irradical
237
3.96
77
3.96
63
7.02
29
8.22
50
6.47
 Missing
22
5
2
0
5
aAnalysis restricted to patients with BCS and presence of DCIS
Besides a higher prevalence of DCIS in the Her2+ groups, DCIS was more often located outside the invasive component and the DCIS component was more extensive (P < 0.0001 for all variables). DCIS-associated microcalcifications were most often seen adjacent to Her2+ IBC, while the frequency was lowest in the triple-negative group (p < 0.0001).
Analysis of resection margin status was restricted to patients treated with BCS. Overall, the frequency of irradicality (of either the invasive or the DCIS component) was highest in the Her2+ subgroups and lowest in the triple-negative subgroup (P < 0.0001). Analysis of irradicality of the invasive component separately showed the highest frequency of irradicality in the ER+/Her2− subgroups and the lowest in the triple-negative subgroup (P < 0.0001). Analysis of irradicality of the DCIS component however showed another distribution as compared to the irradicality of the invasive component; the frequency of irradicality of the DCIS component was highest in the Her2+ subgroups (P < 0.0001).

Discussion

Our national registration system for pathology reporting provided a unique opportunity for this large-scale population-wide cohort study describing the presence and extent of DCIS according to breast cancer subtypes, in relation to other clinicopathologic features.
In our study, we showed substantial differences between immunohistochemical breast cancer subtypes regarding age, type of surgery, histology, tumor grade, and tumor size, which is consistent with literature [41, 42]. Briefly, Her2+ and triple-negative tumors are associated with younger age, larger size, and higher grade compared to luminal subtypes. However, on the other side of the spectrum, ER+/Her2− IBC showed the most favorable tumor characteristics, especially in the case of a high PR expression. This is in line with recent work of Prat et al. in which they concluded that the addition of a PR expression of more than 20 % adds prognostic value within the current immunohistochemical-based luminal A definition by improving the identification of IBCs with a good prognosis [37]. The ER+/Her2+ group seems to be an intermediate subgroup.
Regarding DCIS, we showed that Her2+ IBC is associated with a higher prevalence of adjacent DCIS and a larger extent of DCIS as compared to other IBC subtypes. In line with this, we reported a relatively high rate of irradicality of the DCIS component in Her2+ IBC. These findings are consistent with previous studies reporting a relatively high rate of Her2 positivity in pure DCIS cases, presence of extensive DCIS adjacent to Her2+ IBC, and a high LRR after BCS for Her2+ IBC [13, 29, 30, 42, 43]. Since the risk of an irradical resection is higher for IBCs that are associated with an extensive DCIS component as compared to those with a limited in situ component [41, 44], it seems likely that the DCIS component adjacent to Her2+ IBC is responsible for the high LRR. Therefore, preoperative knowledge regarding the extent of DCIS according to breast cancer subtypes may result in adjustment of local therapy and consequently local control. This may reduce undertreatment in those patients with a large DCIS component, including fewer secondary surgeries and local recurrences. On the other hand, it may result in less overtreatment in those patients with a low prevalence and/or limited extent of DCIS, e.g., by reduction of excision volume which affects cosmetic outcome. In recent years, there is an increased number of pathology laboratories performing the ER, PR, and Her2 status on preoperative needle biopsies on a routine basis, mainly as a result of the increased use of neoadjuvant treatment, which provides a better understanding of tumor growth patterns preoperatively. The presence of DCIS-associated microcalcifications adjacent to the majority of Her2+ IBCs, as shown in this study, may provide important preoperative information regarding imaging by mammography. Besides, since the DCIS component adjacent to Her2+ IBCs is mainly of high grade, a preoperative MRI could be beneficial for these patients, particularly for those without microcalcifications, since this imaging technique is considered to be the most sensitive modality in detecting the presence and extent of intermediate- and high-grade DCIS [45, 46].
According to our knowledge, our study includes the largest series of patients ever published regarding the presence and extent of DCIS adjacent to breast cancer subtypes, thanks to our national protocolled registration of breast cancer pathology reports. However, our study also has several weaknesses including the missing data regarding receptor expression of the DCIS component. However, since several studies reported a very high concordance (90–100 %) of ER, PR, and Her2 expression between DCIS and adjacent IBC, it is highly unlikely that this has affected our results. The second limitation is the lack of information regarding proliferation, because Ki-67 is not routinely performed in our pathology laboratories. This limited an accurate categorization of luminal A versus luminal B subtypes, which is partly based on a low versus a high Ki-67 index. A third limitation of our study is the lack of clinical follow-up regarding local control. In this study, we used data from 2009 (in this year we started registering according to standard pathology protocols) until 2015, resulting in inadequate follow-up time.
In conclusion, in this large population-based cohort study, we showed significant differences between the prevalence and extent of DCIS according to breast cancer subtypes. Her2+ IBC was associated with the highest prevalence and extent of DCIS, while on the other side of the spectrum, triple-negative IBC had the lowest prevalence of DCIS of all IBC subtypes. Since the extent of DCIS was also reflected in the resection margin status in patients treated with BCS, these data provide important information regarding the optimization of local therapy.

Compliance with ethical standards

Conflict of interest

None declared.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution-NonCommercial 4.0 International License (http://creativecommons.org/licenses/by-nc/4.0/), which permits any noncommercial use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

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.

Literatur
1.
Zurück zum Zitat Perou CM, Sorlie T, Eisen MB et al (2000) Molecular portraits of human breast tumours. Nature 406:747–752CrossRefPubMed Perou CM, Sorlie T, Eisen MB et al (2000) Molecular portraits of human breast tumours. Nature 406:747–752CrossRefPubMed
2.
Zurück zum Zitat Sorlie T, Perou CM, Tibshirani R et al (2001) Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci 98:10869–10874CrossRefPubMedPubMedCentral Sorlie T, Perou CM, Tibshirani R et al (2001) Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci 98:10869–10874CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Louwman MW, Vriezen M, van Beek MW et al (2007) Uncommon breast tumors in perspective: incidence, treatment and survival in the Netherlands. Int J Cancer 121:127–135CrossRefPubMed Louwman MW, Vriezen M, van Beek MW et al (2007) Uncommon breast tumors in perspective: incidence, treatment and survival in the Netherlands. Int J Cancer 121:127–135CrossRefPubMed
4.
Zurück zum Zitat Nielsen TO, Perou CM (2015) CCR 20th Anniversary Commentary: the Development of Breast Cancer Molecular Subtyping. Clin Cancer Res 21:1779–1781CrossRefPubMed Nielsen TO, Perou CM (2015) CCR 20th Anniversary Commentary: the Development of Breast Cancer Molecular Subtyping. Clin Cancer Res 21:1779–1781CrossRefPubMed
5.
Zurück zum Zitat Ng CK, Schultheis AM, Bidard FC, Wiegelt B, Reis-Filho JS et al (2015) Breast cancer genomics from microarrays to massively parallel sequencing: paradigms and new insights. J Natl Cancer Inst 107:djv015CrossRefPubMed Ng CK, Schultheis AM, Bidard FC, Wiegelt B, Reis-Filho JS et al (2015) Breast cancer genomics from microarrays to massively parallel sequencing: paradigms and new insights. J Natl Cancer Inst 107:djv015CrossRefPubMed
6.
7.
Zurück zum Zitat Guedj M, Marisa L, de Reynies A et al (2012) A refined molecular taxonomy of breast cancer. Oncogene 31:1196–1206CrossRefPubMed Guedj M, Marisa L, de Reynies A et al (2012) A refined molecular taxonomy of breast cancer. Oncogene 31:1196–1206CrossRefPubMed
8.
Zurück zum Zitat Van Steenbergen LN, Voogd AC, Roukema JA et al (2009) Screening caused rising incidence rates of ductal carcinoma in situ of the breast. Breast Cancer Res Treat 115:181–183CrossRefPubMed Van Steenbergen LN, Voogd AC, Roukema JA et al (2009) Screening caused rising incidence rates of ductal carcinoma in situ of the breast. Breast Cancer Res Treat 115:181–183CrossRefPubMed
9.
Zurück zum Zitat Louwman WJ, Voogd AC, van Dijck JA et al (2008) On the rising trends of incidence and prognosis for breast cancer patients diagnosed 1975–2004: a long-term population-based study in southeastern Netherlands. Cancer Causes Control 19:97–106CrossRefPubMed Louwman WJ, Voogd AC, van Dijck JA et al (2008) On the rising trends of incidence and prognosis for breast cancer patients diagnosed 1975–2004: a long-term population-based study in southeastern Netherlands. Cancer Causes Control 19:97–106CrossRefPubMed
10.
Zurück zum Zitat Steinman S, Wang J, Bourne P, Yang Q, Tang P (2007) Expression of cytokeratin markers, ER-alpha, PR, HER-2/neu, and EGFR in pure ductal carcinoma in situ (DCIS) and DCIS with co-existing invasive ductal carcinoma (IDC) of the breast. Ann Clin Lab Sci 37:127–134PubMed Steinman S, Wang J, Bourne P, Yang Q, Tang P (2007) Expression of cytokeratin markers, ER-alpha, PR, HER-2/neu, and EGFR in pure ductal carcinoma in situ (DCIS) and DCIS with co-existing invasive ductal carcinoma (IDC) of the breast. Ann Clin Lab Sci 37:127–134PubMed
11.
Zurück zum Zitat Park K, Han S, Kim H, Kim J, Shin E (2006) HER2 status in pure ductal carcinoma in situ and in the intraductal and invasive components of invasive ductal carcinoma determined by fluorescence in situ hybridization and immunohistochemistry. Histopathology 48:702–707CrossRefPubMed Park K, Han S, Kim H, Kim J, Shin E (2006) HER2 status in pure ductal carcinoma in situ and in the intraductal and invasive components of invasive ductal carcinoma determined by fluorescence in situ hybridization and immunohistochemistry. Histopathology 48:702–707CrossRefPubMed
12.
Zurück zum Zitat Steinman S, Wang J, Bourne P, Kim J, Shin E (2007) Expression of cytokeratin markers, ER-alpha, PR, HER-2/neu, and EGFR in pure ductal carcinoma in situ (DCIS) and DCIS with co-existing invasive ductal carcinoma (IDC) of the breast. Ann Clin Lab Sci 37:127–134PubMed Steinman S, Wang J, Bourne P, Kim J, Shin E (2007) Expression of cytokeratin markers, ER-alpha, PR, HER-2/neu, and EGFR in pure ductal carcinoma in situ (DCIS) and DCIS with co-existing invasive ductal carcinoma (IDC) of the breast. Ann Clin Lab Sci 37:127–134PubMed
13.
Zurück zum Zitat Meijnen P, Peterse JL, Antonini N, Rutgers EJ, van de Vijver MJ (2008) Immunohistochemical categorisation of ductal carcinoma in situ of the breast. Br J Cancer 98:137–142CrossRefPubMed Meijnen P, Peterse JL, Antonini N, Rutgers EJ, van de Vijver MJ (2008) Immunohistochemical categorisation of ductal carcinoma in situ of the breast. Br J Cancer 98:137–142CrossRefPubMed
14.
Zurück zum Zitat Clark SE, Warwick J, Carpenter R, Bowen RL, Duffy SW, Jones JL (2011) Molecular subtyping of DCIS: heterogeneity of breast cancer reflected in pre-invasive disease. Br J Cancer 104:120–127CrossRefPubMed Clark SE, Warwick J, Carpenter R, Bowen RL, Duffy SW, Jones JL (2011) Molecular subtyping of DCIS: heterogeneity of breast cancer reflected in pre-invasive disease. Br J Cancer 104:120–127CrossRefPubMed
15.
Zurück zum Zitat Zhou W, Jirström K, Amini RM et al (2013) Molecular subtypes in ductal carcinoma in situ of the breast and their relation to prognosis: a population-based cohort study. BMC Cancer 13:512CrossRefPubMedPubMedCentral Zhou W, Jirström K, Amini RM et al (2013) Molecular subtypes in ductal carcinoma in situ of the breast and their relation to prognosis: a population-based cohort study. BMC Cancer 13:512CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Tamimi RM, Baer HJ, Marotti J et al (2008) Comparison of molecular phenotypes of ductal carcinoma in situ and invasive breast cancer. Breast Cancer Res 10:R67CrossRefPubMedPubMedCentral Tamimi RM, Baer HJ, Marotti J et al (2008) Comparison of molecular phenotypes of ductal carcinoma in situ and invasive breast cancer. Breast Cancer Res 10:R67CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Su Y, Zheng Y, Zheng W et al (2011) Distinct distribution and prognostic significance of molecular subtypes of breast cancer in Chinese women: a population-based cohort study. BMC Cancer 11:292CrossRefPubMedPubMedCentral Su Y, Zheng Y, Zheng W et al (2011) Distinct distribution and prognostic significance of molecular subtypes of breast cancer in Chinese women: a population-based cohort study. BMC Cancer 11:292CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Bauer KR, Brown M, Cress RD, Parise CA, Caggiano V (2007) Descriptive analysis of estrogen receptor (ER)-negative, progesterone receptor (PR)-negative, and HER2-negative invasive breast cancer, the so-called triple-negative phenotype: a population-based study from the California cancer Registry. Cancer 109:1721–1728CrossRefPubMed Bauer KR, Brown M, Cress RD, Parise CA, Caggiano V (2007) Descriptive analysis of estrogen receptor (ER)-negative, progesterone receptor (PR)-negative, and HER2-negative invasive breast cancer, the so-called triple-negative phenotype: a population-based study from the California cancer Registry. Cancer 109:1721–1728CrossRefPubMed
19.
Zurück zum Zitat Cheang MC, Chia SK, Voduc D et al (2009) Ki67 index, HER2 status, and prognosis of patients with luminal B breast cancer. J Natl Cancer Inst 101:736–750CrossRefPubMedPubMedCentral Cheang MC, Chia SK, Voduc D et al (2009) Ki67 index, HER2 status, and prognosis of patients with luminal B breast cancer. J Natl Cancer Inst 101:736–750CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Kurbel S (2013) In search of triple-negative DCIS: tumor-type dependent model of breast cancer progression from DCIS to the invasive cancer. Tumour Biol 4:1–7CrossRef Kurbel S (2013) In search of triple-negative DCIS: tumor-type dependent model of breast cancer progression from DCIS to the invasive cancer. Tumour Biol 4:1–7CrossRef
21.
Zurück zum Zitat Van Steenbergen LN, Voogd AC, Roukema JA et al (2014) Time trends and inter-hospital variation in treatment and axillary staging of patients with ductal carcinoma in situ of the breast in the era of screening in Southern Netherlands. Breast 23:63–68CrossRefPubMed Van Steenbergen LN, Voogd AC, Roukema JA et al (2014) Time trends and inter-hospital variation in treatment and axillary staging of patients with ductal carcinoma in situ of the breast in the era of screening in Southern Netherlands. Breast 23:63–68CrossRefPubMed
22.
Zurück zum Zitat Vidali C, Caffo O, Aristei C et al (2012) Conservative treatment of breast ductal carcinoma in situ: results of an Italian multi-institutional retrospective study. Radiat Oncol 7:177CrossRefPubMedPubMedCentral Vidali C, Caffo O, Aristei C et al (2012) Conservative treatment of breast ductal carcinoma in situ: results of an Italian multi-institutional retrospective study. Radiat Oncol 7:177CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Rakovitch E, Nofech-Mozes S, Hanna W et al (2012) HER2/neu and Ki-67 expression predict non-invasive recurrence following breast-conserving therapy for ductal carcinoma in situ. Br J Cancer 13(106):1160–1165CrossRef Rakovitch E, Nofech-Mozes S, Hanna W et al (2012) HER2/neu and Ki-67 expression predict non-invasive recurrence following breast-conserving therapy for ductal carcinoma in situ. Br J Cancer 13(106):1160–1165CrossRef
24.
Zurück zum Zitat Donker M, Litiere S, Werutsky G et al (2013) Breast-conserving treatment with or without radiotherapy in ductal carcinoma in situ: 15-year recurrence rates and outcome after a recurrence, from the EORTC 10853 randomized phase III trial. J Clin Oncol 31:4054CrossRefPubMed Donker M, Litiere S, Werutsky G et al (2013) Breast-conserving treatment with or without radiotherapy in ductal carcinoma in situ: 15-year recurrence rates and outcome after a recurrence, from the EORTC 10853 randomized phase III trial. J Clin Oncol 31:4054CrossRefPubMed
25.
Zurück zum Zitat Han K, Nofech-Mozes S, Narod S et al (2012) Expression of HER2neu in ductal carcinoma in situ is associated with local recurrence. J Clin Oncol 24:183–189CrossRef Han K, Nofech-Mozes S, Narod S et al (2012) Expression of HER2neu in ductal carcinoma in situ is associated with local recurrence. J Clin Oncol 24:183–189CrossRef
26.
Zurück zum Zitat Lazzeroni M, Guerrieri-Gonzaga A, Botteri E et al (2013) Tailoring treatment for ductal intraepithelial neoplasia of the breast according to Ki-67 and molecular phenotype. Br J Cancer 108:1593–1601CrossRefPubMedPubMedCentral Lazzeroni M, Guerrieri-Gonzaga A, Botteri E et al (2013) Tailoring treatment for ductal intraepithelial neoplasia of the breast according to Ki-67 and molecular phenotype. Br J Cancer 108:1593–1601CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Williams KE, Barnes NLP, Cramer A et al (2015) Molecular phenotypes of DCIS predict overall and invasive recurrence. Ann Oncol 26:1019–1025CrossRefPubMed Williams KE, Barnes NLP, Cramer A et al (2015) Molecular phenotypes of DCIS predict overall and invasive recurrence. Ann Oncol 26:1019–1025CrossRefPubMed
28.
Zurück zum Zitat Curigliano G, Disalvatore D, Esposito A et al (2015) Risk of subsequent in situ and invasive breast cancer in human epidermal growth factor receptor 2-positive ductal carcinoma in situ. Ann Oncol 26:682–687CrossRefPubMed Curigliano G, Disalvatore D, Esposito A et al (2015) Risk of subsequent in situ and invasive breast cancer in human epidermal growth factor receptor 2-positive ductal carcinoma in situ. Ann Oncol 26:682–687CrossRefPubMed
29.
Zurück zum Zitat Voduc KD, Cheang MC, Tyldesley S, Gelmon K, Nielsen TO, Kennecke H (2010) Breast cancer subtypes and the risk of local and regional relapse. J Clin Oncol 28:1684–1691CrossRefPubMed Voduc KD, Cheang MC, Tyldesley S, Gelmon K, Nielsen TO, Kennecke H (2010) Breast cancer subtypes and the risk of local and regional relapse. J Clin Oncol 28:1684–1691CrossRefPubMed
30.
Zurück zum Zitat Nguyen PL, Taghian AG, Katz MS et al (2008) Breast cancer subtype approximated by estrogen receptor, progesterone receptor, and HER-2 is associated with local and distant recurrence after breast-conserving therapy. J Clin Oncol 26:2373–2378CrossRefPubMed Nguyen PL, Taghian AG, Katz MS et al (2008) Breast cancer subtype approximated by estrogen receptor, progesterone receptor, and HER-2 is associated with local and distant recurrence after breast-conserving therapy. J Clin Oncol 26:2373–2378CrossRefPubMed
31.
Zurück zum Zitat Rezai M, Kellersmann S, Knispel S, Lax H, Kimmig R, Kern P (2015) Translating the concept of intrinsic subtypes into an oncoplastic cohort of more than 1000 patients—predictors of recurrence and survival. Breast 24:384–390CrossRefPubMed Rezai M, Kellersmann S, Knispel S, Lax H, Kimmig R, Kern P (2015) Translating the concept of intrinsic subtypes into an oncoplastic cohort of more than 1000 patients—predictors of recurrence and survival. Breast 24:384–390CrossRefPubMed
32.
Zurück zum Zitat Casparie M, Tiebosch AT, Burger G et al (2007) Pathology databanking and biobanking in The Netherlands, a central role for PALGA, the nationwide histopathology and cytopathology data network and archive. Cell Oncol 29:19–24PubMedPubMedCentral Casparie M, Tiebosch AT, Burger G et al (2007) Pathology databanking and biobanking in The Netherlands, a central role for PALGA, the nationwide histopathology and cytopathology data network and archive. Cell Oncol 29:19–24PubMedPubMedCentral
33.
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 19:403–410CrossRefPubMed 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 19:403–410CrossRefPubMed
35.
Zurück zum Zitat Wolff AC, Hammond ME, Hicks DG et al (2013) American society of clinical oncology; college of American pathologists. recommendations for human epidermal growth factor 2 testing in breast cancer: american society of clinical oncology/college of American pathologists practice guideline update. J Clin Oncol 31:3997–4013CrossRefPubMed Wolff AC, Hammond ME, Hicks DG et al (2013) American society of clinical oncology; college of American pathologists. recommendations for human epidermal growth factor 2 testing in breast cancer: american society of clinical oncology/college of American pathologists practice guideline update. J Clin Oncol 31:3997–4013CrossRefPubMed
36.
Zurück zum Zitat Goldhirsch A, Winer EP, Coates AS et al (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 24:2206–2223CrossRefPubMedPubMedCentral Goldhirsch A, Winer EP, Coates AS et al (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 24:2206–2223CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Prat A, Cheang MC, Martín M et al (2013) Prognostic significance of progesterone receptor-positive tumor cells within immunohistochemically defined luminal A breast cancer. J Clin Oncol 31:203–209CrossRefPubMed Prat A, Cheang MC, Martín M et al (2013) Prognostic significance of progesterone receptor-positive tumor cells within immunohistochemically defined luminal A breast cancer. J Clin Oncol 31:203–209CrossRefPubMed
38.
Zurück zum Zitat Nuclear dcis: the consensus conference committee (1997) Consensus conference on the classification of ductal carcinoma. Cancer 80:1798–1802CrossRef Nuclear dcis: the consensus conference committee (1997) Consensus conference on the classification of ductal carcinoma. Cancer 80:1798–1802CrossRef
40.
Zurück zum Zitat Buchholz TA, Somerfield MR, Griggs JJ et al (2014) Margins for breast-conserving surgery with whole-breast irradiation in stage I and II invasive breast cancer: american Society of Clinical Oncology endorsement of the Society of Surgical Oncology/American Society for Radiation Oncology consensus guideline. J Clin Oncol 32:1502–1506CrossRefPubMed Buchholz TA, Somerfield MR, Griggs JJ et al (2014) Margins for breast-conserving surgery with whole-breast irradiation in stage I and II invasive breast cancer: american Society of Clinical Oncology endorsement of the Society of Surgical Oncology/American Society for Radiation Oncology consensus guideline. J Clin Oncol 32:1502–1506CrossRefPubMed
41.
Zurück zum Zitat Jia H, Jia W, Yang Y et al (2014) HER-2 positive breast cancer is associated with an increased risk of positive cavity margins after initial lumpectomy. World J Surg Oncol 12:289CrossRefPubMedPubMedCentral Jia H, Jia W, Yang Y et al (2014) HER-2 positive breast cancer is associated with an increased risk of positive cavity margins after initial lumpectomy. World J Surg Oncol 12:289CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Parise CA, Bauer KR, Brown MM, Caggiano V (2009) Breast cancer subtypes as defined by the estrogen receptor (ER), progesterone receptor (PR), and the human epidermal growth factor receptor 2 (HER2) among women with invasive breast cancer in California, 1999-2004. Breast J 15:593–602CrossRefPubMed Parise CA, Bauer KR, Brown MM, Caggiano V (2009) Breast cancer subtypes as defined by the estrogen receptor (ER), progesterone receptor (PR), and the human epidermal growth factor receptor 2 (HER2) among women with invasive breast cancer in California, 1999-2004. Breast J 15:593–602CrossRefPubMed
43.
Zurück zum Zitat Tot T (2015) Early (<10 mm) HER2-Positive Invasive Breast Carcinomas are Associated with Extensive Diffuse High-Grade DCIS: implications for Preoperative Mapping, Extent of Surgical Intervention, and Disease-Free Survival. Ann Surg Oncol 22:2532–2539CrossRefPubMed Tot T (2015) Early (<10 mm) HER2-Positive Invasive Breast Carcinomas are Associated with Extensive Diffuse High-Grade DCIS: implications for Preoperative Mapping, Extent of Surgical Intervention, and Disease-Free Survival. Ann Surg Oncol 22:2532–2539CrossRefPubMed
44.
Zurück zum Zitat Vicini FA, Eberlein TJ, Connolly JL et al (1991) The optimal extent of resection for patients with stages I or II breast cancer treated with conservative surgery and radiotherapy. Ann Surg 214:200–205CrossRefPubMedPubMedCentral Vicini FA, Eberlein TJ, Connolly JL et al (1991) The optimal extent of resection for patients with stages I or II breast cancer treated with conservative surgery and radiotherapy. Ann Surg 214:200–205CrossRefPubMedPubMedCentral
45.
Zurück zum Zitat Greenwood HI, Heller SL, Kim S, Sigmund EE, Shaylor SD, Moy L (2013) Ductal carcinoma in situ of the breasts: review of MR imaging features. Radiographics 33:1569–1588CrossRefPubMed Greenwood HI, Heller SL, Kim S, Sigmund EE, Shaylor SD, Moy L (2013) Ductal carcinoma in situ of the breasts: review of MR imaging features. Radiographics 33:1569–1588CrossRefPubMed
46.
Zurück zum Zitat Yamada T, Mori N, Watanabe M et al (2010) Radiologic-pathologic correlation of ductal carcinoma in situ. Radiographics 30:1183–1198CrossRefPubMed Yamada T, Mori N, Watanabe M et al (2010) Radiologic-pathologic correlation of ductal carcinoma in situ. Radiographics 30:1183–1198CrossRefPubMed
Metadaten
Titel
Extent of ductal carcinoma in situ according to breast cancer subtypes: a population-based cohort study
verfasst von
Shusma C. Doebar
Esther C. van den Broek
Linetta B. Koppert
Agnes Jager
Margreet. H. A. Baaijens
Inge-Marie A. M. Obdeijn
Carolien H. M. van Deurzen
Publikationsdatum
18.06.2016
Verlag
Springer US
Erschienen in
Breast Cancer Research and Treatment / Ausgabe 1/2016
Print ISSN: 0167-6806
Elektronische ISSN: 1573-7217
DOI
https://doi.org/10.1007/s10549-016-3862-4

Weitere Artikel der Ausgabe 1/2016

Breast Cancer Research and Treatment 1/2016 Zur Ausgabe

Update Onkologie

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