Awareness of breast cancer heterogeneity has strikingly increased in the past decade in parallel with the development of high-throughput molecular tests. Beyond the clear usefulness of antiestrogen treatment in luminal tumors and trastuzumab in HER2-positive tumors, breast cancer subtypes may have additional clinical and predictive roles that can be relevant to clinical practice. In this article, we discuss the significance of molecular subtypes in the systemic treatment of early-stage breast tumors smaller than 1 cm (T1a,bN0M0) and suggest new strategies for future treatment recommendations for these patients. Clin Cancer Res; 20(24); 6242–6. ©2014 AACR.

Screening and increased awareness have led to a rise in the detection of T1 breast tumors that are generally estimated to have a low risk of recurrence after locoregional treatment (1–4). However, even small tumors can have an aggressive behavior. This population suffers from low representation in clinical trials, which leads to a situation where medical oncologists lack high-level evidence that can guide them in the treatment of these patients. Tumor size and nodal involvement remain among the most important clinicopathologic prognostic factors in breast cancer, which puts T1a,bN0 (<1 cm) tumors in a group with a generally low risk of recurrence. Information on T1a,bN0 from large population databases of untreated patients demonstrates relatively low cancer-related mortality rates at 15 years (<10%), with much higher death rates from other causes in women older than 50 years (5, 6). However, it has been shown that about 25% of all relapses of small lobular/ductal cancers occur beyond 10 years (7). High grade, young age, high proliferation, and vascular invasion are considered adverse prognostic signs even among T1a,bN0 tumors, suggesting a potential benefit of more aggressive therapy in women diagnosed with tumors harboring any or all of these features (8–11). Furthermore, the classification of breast cancers into different subtypes (luminal A, luminal B, triple negative, and HER2 positive), appears also to affect prognosis and treatment decisions in such early cases (10,12,13). This classification was first based only on histology features [estrogen receptor (ER), progesterone receptor, and HER2] but later it was shown that the differences are also reflected by the mRNA expression profiles (14,15).

Is the threshold for offering adjuvant therapy in T1a,bN0 tumors dependent on the molecular subtype?

There is a lack of high-level evidence regarding the use of chemotherapy in women with T1a,bN0 tumors. However, it is possible to derive prognostic information based on tumor subtype. In a retrospective study conducted by Theriault and colleagues (16), 1,012 patients with T1a,bN0 breast cancer who did not receive chemotherapy or trastuzumab were evaluated. Compared with patients with hormone receptor (HR)–positive disease, patients with HER2-positive and triple-negative breast cancer (TNBC) had 4.9 and 2.7 times greater risk of recurrence, respectively. Another study, conducted by Cancello and colleagues (17), exploring patterns of recurrence in 1,691 patients with T1a,b breast cancers confirmed the same findings. Multivariate analysis showed that women with HER2-positive and TNBC had an increased risk of locoregional relapse by 3-fold and breast cancer–related events by 2-fold, respectively. The luminal B subtype was not associated with a statistically significant increased risk of recurrence when compared with luminal A. The increased recurrence in HER2 patients was demonstrated despite the fact that 44% of HER2-positive patients were treated with chemotherapy compared with only 8% in the luminal B patients. In another recent observational nonrandomized prospective study, included 4,113 women with T1a,bN0 breast cancer, patients with HR-positive disease had the lowest recurrence rates, whereas patients with HR-negative (TNBC or HER2-positive) tumors had the worse outcomes, again, despite the fact that only 8% of HR-positive patients were treated with chemotherapy compared with more than 50% in the HR-negative group (18). On the basis of these data, it appears that women with small size, node-negative breast cancer are at higher risk of relapse if they have HER2-positive or triple-negative breast cancer, whereas for luminal A and B cancers the risk is less clear.

Luminal tumors

In the past decade, several genomic signatures emerged as useful tools to define patients at high risk of recurrence who might benefit from the addition of adjuvant chemotherapy (19). The Oncotype DX 21-gene recurrence score assay is able to predict risk for distant recurrence among ER-positive breast cancers treated with tamoxifen (20). Except from being prognostic, in two retrospective studies involving both node-negative and node-positive patients, Oncotype DX was found to be predictive for the chemotherapy benefit (21,22). Therefore, this system was endorsed by the National Comprehensive Cancer Network for ER-positive tumors >0.5 cm (T1b) as a means to decide whether to give chemotherapy. In the National Surgical Adjuvant Breast and Bowel Project trials that evaluated the Oncotype DX, the distribution of the recurrence scores did not differ according to the size of the tumors and 15% to 16% of the T1a,bN0 tumors were considered at high risk (20,23). In a population-based retrospective study, T1a,bN0 tumors with a high recurrence score (>31) were associated with a 10.1% risk of breast cancer–related death at 10 years compared with 1.3% in the low-risk group (24).

The MammaPrint 70-gene signature has been previously validated as an independent prognostic factor in node-negative and node-positive breast cancer (25). Mook and colleagues (26) evaluated the accuracy of the 70-gene signature in T1 breast cancer irrespective of nodal involvement, and it was found to be an independent prognostic factor for breast cancer survival at 10 years. These data show that the 70-gene signature can also help to individualize adjuvant treatment recommendations in this population.

A more definite proof of the utility of MammaPrint in the setting of T1a,bN0 tumors might come from the MINDACT trial, which compares MammaPrint with the common clinicopathologic criteria in their respective abilities to select patients at high risk for adjuvant chemotherapy. MINDACT has enrolled 6,600 patients, of whom 14% (N = 919) have tumors less than 1 cm in size. The results of this trial are expected to be reported in 2015 to 2016 and will provide high-level evidence about the clinical relevance of applying gene-expression predictors in this setting.

In one of the largest efforts in the field, the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) group has performed an integrated genomic analysis of approximately 2,000 breast cancer primary samples (27). New breast cancer clusters were identified of which an ER-positive subgroup composed of 11q13/14 cis-acting luminal tumors drew specific attention, as it exhibited a steep mortality trajectory. This subgroup showed high frequencies of copy-number variance in the 11q region and amplification of the CCND1 and EMSY genes that reside in the amplicon. Although the predictive role of 11q13-14 amplicon is not clear, the dismal prognosis of these patients may help in tailoring future chemotherapy in T1a,bN0 cancers. For example, patients with amplification of the 11q13-14 amplicon could be recommended to receive chemotherapy, whereas luminal subgroups with copy-number alterations of favorable outcome (1q gain/16q loss) could be spared adjuvant chemotherapy. This concept remains speculative and requires further validation. The fact that different prognostic/predictive gene sets were developed by different groups, but nonetheless do similar jobs as prognostic tools might be attributable to the different technologies used, their implementation, and the nature of the validation tools.

HER2-enriched tumors

Five major randomized studies have shown that trastuzumab improves disease-free survival (DFS) and overall survival in the adjuvant setting when added to backbone chemotherapy in HER2-positive breast cancer (28–31); however, none of these studies recruited patients with T1a,bN0 except the BCIRG006 trial, which randomly assigned 3,222 women with HER2-positive early-stage breast cancer to receive doxorubicin and cyclophosphamide followed by docetaxel every 3 weeks, the same regimen plus trastuzumab, or docetaxel and carboplatin plus trastuzumab (29). Despite this fact, most clinical guidelines and most clinicians, as demonstrated in practice surveys, recommend giving trastuzumab for these small tumors, particularly T1b tumors (32). This is influenced by the significantly higher risk of relapse in HER2-positive patients (13,16). Nevertheless, it is also true that medical oncologists might be overtreating a large fraction of these patients by offering the classic chemotherapy (anthracycline–taxane)-plus-trastuzumab combination. The REMAGUS French breast cancer group retrospectively reviewed the medical charts of patients who had presented with invasive T1a,bN0, HER2-positive breast cancers (33). The records of 97 node-negative patients were retrieved. Forty-one (42%) had been treated with adjuvant trastuzumab. During a median overall follow-up of 29 months, all patients who had received a trastuzumab-based therapy were free of recurrence, even though they generally had more adverse prognostic factors (HR negative and higher grade). The recurrence-free survival rate at 29 months for the 56 patients who had not received an adjuvant trastuzumab-based therapy was estimated at 93.6%. Four of the five recurrences observed in this group were distant metastases and occurred in HR-negative patients, suggesting the poorer outcome of this subgroup. In another recent nonrandomized prospective study performed at Dana-Farber Cancer Institute (Boston, MA; Adjuvant Paclitaxel/Trastuzumab study), 406 women with HER2-positive, node-negative tumors smaller than 3 cm were treated with a regimen that included paclitaxel, 80 mg/m2, plus trastuzumab, 2 mg/kg, for 12 weeks, followed by 9 months of trastuzumab alone at a dose of 6 mg/kg every 3 weeks (34). After a median follow-up of 3.6 years, only four recurrences (0.9%) were observed (34). About 20% of the trial patients had T1a tumors. This suggests that paclitaxel plus trastuzumab can be considered an attractive approach for T1a,bN0 HER2-positive breast cancer in terms of balancing benefits versus risks. The same group is currently planning another clinical trial (ATEMPT), comparing TDM1 with trastuzumab plus paclitaxel in T1 HER2-positive patients, which may provide alternative future regimens for these patients. The Aphinity trial, which is evaluating the usefulness of pertuzumab in addition to trastuzumab and chemotherapy also enrolled a limited number of T1a,bN0 patients at high risk (ER negative or grade 3). The results of this trial are expected to be reported in 2016 and will provide high-level evidence for the treatment of this subgroup of patients.

The role that the immune system plays in the development and clinical behavior of breast cancer can differ according to the subtypes. A way to define the immune response against cancer is the quantification of tumor-infiltrating lymphocytes (TIL). TILs were found to be in greater numbers in the HER2 and TNBC compared with the luminal subtypes (35). In the FinHER trial, which randomized patients with HER2-positive breast cancers to 9 weeks of trastuzumab or no trastuzumab in the adjuvant setting, each 10% increase in lymphocytic infiltration was significantly associated with decreased distant recurrence in patients randomized to trastuzumab arm (36). These data support the role of antitumor immunity in the efficacy of trastuzumab. In addition, patients with HER2-positive disease and high TILs were found to benefit more from adjuvant anthracycline-based chemotherapy as observed in the BIG02-98 randomized phase III trial (35). This suggests that TILs may be used to define patients with T1a,bN0 that would derive the highest benefit of trastuzumab and chemotherapy in the adjuvant setting.

Triple-negative breast cancer

TNBCs, which account for 15% to 20% of all invasive breast cancers, have an aggressive nature and increased risk of relapse and breast cancer–related deaths compared with other subtypes (37). Although from a clinical point of view, TNBCs are managed as one group, recent studies have shown that TNBCs are composed of a heterogeneous group of diseases with different biologies and clinical behaviors (38–40). Current guidelines suggest adjuvant chemotherapy for patients with tumor size >0.5 cm (T1b; ref. 41). As no targeted therapy is currently available for the treatment of TNBCs, chemotherapy remains the only option.

Using gene-expression profiling, seven distinct TNBC subtypes were identified, and later on, a bioinformatics tool (TNBC type) was developed to determine the TNBC molecular subtype from gene-expression profiles (42). This tool was further validated in the Tumor Cancer Genome Atlas data. TNBC subtype-specific differences in survival were demonstrated; for instance, although the immune-modulator subtype had a median DFS duration of 20.1 months, the luminal-androgen-receptor subtype (not to be confused with luminal ER-positive tumors) had a median DFS of 4.4 months. This may also have clinical relevance as there is progress in the development of androgen receptor-targeted therapies (43). The METABRIC consortium has also identified a large subgroup among TNBC basal-like tumors characterized by high-genomic instability (5 loss/8q gain/10p gain/12p gain) and relatively good long-term outcomes (27). These molecular findings may guide future differential use of chemotherapy in T1a,bN0 cancers.

As described for HER2-positive patients, TILs were also found to be high in the TNBC breast cancer group (35). Increasing 10% increments of infiltration were in fact associated with better prognosis, supporting a possible prognostic role of TILs in patients with small tumors.

The T1a-T1b cutoff

The classical staging system discriminates between T1a and T1b tumors (below or above 0.5 cm) based on large longitudinal cohorts that demonstrated minimal differences in prognosis between these groups with relatively no impact on rate of breast cancer–related deaths (24). As it is becoming clearer that the aggressiveness of tumors is biology driven rather than influenced by tumor size alone, it may not be advisable to artificially dichotomize patients using the 0.5-cm cutoff for diagnostic tests and treatment decisions. Moreover, there is evidence that tissue fixation and handling may affect tumor size (44, 45). This includes the impact of preoperative biopsies, the effect of paraffin temperature, the pathologic observer discrepancies, and the tendency for rounding results that have been shown to influence the correct estimation of tumor size (46). In two recent reports there were no clear differences in outcomes between patients with T1a and T1b in HER2-positive tumors (18, 47). Therefore, oncologists should be cautious when making decisions solely based on whether the tumor is T1a or T1b. It is possible that in the near future assays such as Oncotype DX or MammaPrint and newer prognostic tools such as TNBCtype will be helpful in defining the best treatment strategy for small tumors.

The frequency of T1a,bN0 tumors is increasing sharply, especially in the Western world as a result of mammographic screening. The need to balance risks and benefits when establishing a treatment strategy in this relatively good prognostic group of patients is not trivial. The recent molecular classification of breast cancers, together with the development of new prognostic tools and -omics-based clinical trials, may facilitate more accurate tailoring of treatment recommendations for this population of breast cancer patients (Table 1). Instead of differentially dichotomizing treatment decisions based on tumor size (>0.5 cm), it is hoped that the future will bring more individualized treatment strategies that will take into account the biologic aggressiveness of the tumor, the clinical parameters (age and comorbidities) of the patient, and the patient's preferences.

Table 1.

Diagnostic and treatment options for T1a,b breast cancer

SubtypeCurrent recommendationsFuture possible recommendations
Luminal T1a—No chemotherapy T1a,b—Oncotype DX MammaPrint 
 T1b—Oncotype DX 11q13/14, 1q gain/16q loss 
HER2 positive T1a—No adjuvant therapy T1a,b—tolerable regimens 
 T1b—Trastuzumab + aggressive chemotherapy New drugsTILs 
TNBC T1a—No chemotherapy T1a,b—TNBCtype, 5 loss/8q gain/10p gain/12p gain 
 T1b—Adjuvant chemotherapy TILs 
SubtypeCurrent recommendationsFuture possible recommendations
Luminal T1a—No chemotherapy T1a,b—Oncotype DX MammaPrint 
 T1b—Oncotype DX 11q13/14, 1q gain/16q loss 
HER2 positive T1a—No adjuvant therapy T1a,b—tolerable regimens 
 T1b—Trastuzumab + aggressive chemotherapy New drugsTILs 
TNBC T1a—No chemotherapy T1a,b—TNBCtype, 5 loss/8q gain/10p gain/12p gain 
 T1b—Adjuvant chemotherapy TILs 

H.A. Azim is a consultant/advisory board member for Celgene, GlaxoSmithKline, Nanostring, and Novartis. M. Piccart is a board member for PharmaMar and a consultant/advisory board member for Amgen, Astellas, AstraZeneca, Bayer, Eli Lilly, Invivis, Merck, Novartis, Pfizer, Roche/Genentech, Sanofi-Aventis, Symphogen, Synthon, and Verastem. No potential conflicts of interest were disclosed by the other authors.

Conception and design: A. Sonnenblick, D. Fumagalli, H.A. Azim Jr, M. Piccart

Development of methodology: M. Piccart

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): A. Sonnenblick, D. Fumagalli

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): M. Piccart

Writing, review, and/or revision of the manuscript: A. Sonnenblick, D. Fumagalli, H.A. Azim Jr, C. Sotiriou, M. Piccart

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): A. Sonnenblick, D. Fumagalli

The authors thank Carolyn Straehle for her editorial work.

A. Sonnenblick is an ESMO translational research fellow.

1.
Fracheboud
J
,
Otto
SJ
,
van Dijck
JAAM
,
Broeders
MJM
,
Verbeek
ALM
,
de Koning
HJ
, et al
Decreased rates of advanced breast cancer due to mammography screening in the Netherlands
.
Br J Cancer
2004
;
91
:
861
7
.
2.
Schootman
M
,
Jeffe
D
,
Reschke
A
,
Aft
R
. 
The full potential of breast cancer screening use to reduce mortality has not yet been realized in the United States
.
Breast Cancer Res Treat
2004
;
85
:
219
22
.
3.
Youlden
DR
,
Cramb
SM
,
Dunn
NAM
,
Muller
JM
,
Pyke
CM
,
Baade
PD
. 
The descriptive epidemiology of female breast cancer: an international comparison of screening, incidence, survival, and mortality
.
Cancer Epidemiol
2012
;
36
:
237
48
.
4.
Bland
KI
,
Menck
HR
,
Scott-Conner
CE
,
Morrow
M
,
Winchester
DJ
,
Winchester
DP
. 
The National Cancer Data Base 10-year survey of breast carcinoma treatment at hospitals in the United States
.
Cancer
1998
;
83
:
1262
73
.
5.
Tabár
L
,
Fagerberg
G
,
Day
NE
,
Duffy
SW
,
Kitchin
RM
. 
Breast cancer treatment and natural history: new insights from results of screening
.
Lancet
1992
;
339
:
412
4
.
6.
Hanrahan
EO
,
Gonzalez-Angulo
AM
,
Giordano
SH
,
Rouzier
R
,
Broglio
KR
,
Hortobagyi
GN
, et al
Overall survival and cause-specific mortality of patients with stage T1a,bN0M0 breast carcinoma
.
J Clin Oncol
2007
;
25
:
4952
60
.
7.
Rosen
PP
,
Saigo
PE
,
Braun
DW
 Jr
,
Weathers
E
,
DePalo
A
. 
Predictors of recurrence in stage I (T1N0M0) breast carcinoma
.
Ann Surg
1981
;
193
:
15
25
.
8.
Houvenaeghel
G
,
Goncalves
A
,
Classe
JM
,
Garbay
JR
,
Giard
S
,
Charytensky
H
, et al
Characteristics and clinical outcome of T1 breast cancer: a multicenter retrospective cohort study
.
Ann Oncol
2014
;
25
:
623
8
.
9.
Leitner
SP
,
Swern
AS
,
Weinberger
D
,
Duncan
LJ
,
Hutter
RV
. 
Predictors of recurrence for patients with small (one centimeter or less) localized breast cancer (T1a,b N0 M0)
.
Cancer
1995
;
76
:
2266
74
.
10.
Hanrahan
EO
,
Valero
V
,
Gonzalez-Angulo
AM
,
Hortobagyi
GN
. 
Prognosis and management of patients with node-negative invasive breast carcinoma that is 1 cm or smaller in size (stage 1; T1a,bN0M0): a review of the literature
.
J Clin Oncol
2006
;
24
:
2113
22
.
11.
Fisher
B
,
Dignam
J
,
Tan-Chiu
E
,
Anderson
S
,
Fisher
ER
,
Wittliff
JL
, et al
Prognosis and treatment of patients with breast tumors of one centimeter or less and negative axillary lymph nodes
.
J Natl Cancer Inst
2001
;
93
:
112
20
.
12.
Joensuu
H
,
Isola
J
,
Lundin
M
,
Salminen
T
,
Holli
K
,
Kataja
V
, et al
Amplification of erbB2 and erbB2 expression are superior to estrogen receptor status as risk factors for distant recurrence in pT1N0M0 breast cancer: a nationwide population-based study
.
Clin Cancer Res
2003
;
9
:
923
30
.
13.
Curigliano
G
,
Viale
G
,
Bagnardi
V
,
Fumagalli
L
,
Locatelli
M
,
Rotmensz
N
, et al
Clinical relevance of HER2 overexpression/amplification in patients with small tumor size and node-negative breast cancer
.
J Clin Oncol
2009
;
27
:
5693
9
.
14.
Sørlie
T
,
Perou
CM
,
Tibshirani
R
,
Aas
T
,
Geisler
S
,
Johnsen
H
, et al
Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications
.
Proc Natl Acad Sci U S A
2001
;
98
:
10869
74
.
15.
Perou
CM
,
Sørlie
T
,
Eisen
MB
,
vande Rijn
M
,
Jeffrey
SS
,
Rees
CA
, et al
Molecular portraits of human breast tumours
.
Nature
2000
;
406
:
747
52
.
16.
Theriault
RL
,
Litton
JK
,
Mittendorf
EA
,
Chen
H
,
Meric-Bernstam
F
,
Chavez-Macgregor
M
, et al
Age and survival estimates in patients who have node-negative T1ab breast cancer by breast cancer subtype
.
Clin Breast Cancer
2011
;
11
:
325
31
.
17.
Cancello
G
,
Maisonneuve
P
,
Rotmensz
N
,
Viale
G
,
Mastropasqua
MG
,
Pruneri
G
, et al
Prognosis in women with small (T1mic,T1a,T1b) node-negative operable breast cancer by immunohistochemically selected subtypes
.
Breast Cancer Res Treat
2011
;
127
:
713
20
.
18.
Vaz-Luis
I
,
Ottesen
RA
,
Hughes
ME
,
Mamet
R
,
Burstein
HJ
,
Edge
SB
, et al
Outcomes by tumor subtype and treatment pattern in women with small, node-negative breast cancer: a multi-institutional study
.
J Clin Oncol
2014
;
32
:
2142
50
.
19.
Azim
HA
 Jr
,
Michiels
S
,
Zagouri
F
,
Delaloge
S
,
Filipits
M
,
Namer
M
, et al
Utility of prognostic genomic tests in breast cancer practice: the IMPAKT 2012 Working Group Consensus Statement
.
Ann Oncol
2013
;
24
:
647
54
.
20.
Paik
S
,
Tang
G
,
Shak
S
,
Kim
C
,
Baker
J
,
Kim
W
, et al
Gene expression and benefit of chemotherapy in women with node-negative, estrogen receptor-positive breast cancer
.
J Clin Oncol
2006
;
24
:
3726
34
.
21.
Dowsett
M
,
Cuzick
J
,
Wale
C
,
Forbes
J
,
Mallon
EA
,
Salter
J
, et al
Prediction of risk of distant recurrence using the 21-gene recurrence score in node-negative and node-positive postmenopausal patients with breast cancer treated with anastrozole or tamoxifen: a TransATAC study
.
J Clin Oncol
2010
;
28
:
1829
34
.
22.
Albain
KS
,
Barlow
WE
,
Shak
S
,
Hortobagyi
GN
,
Livingston
RB
,
Yeh
I-T
, et al
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
2010
;
11
:
55
65
.
23.
Paik
S
,
Shak
S
,
Tang
G
,
Kim
C
,
Baker
J
,
Cronin
M
, et al
A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer
.
N Engl J Med
2004
;
351
:
2817
26
.
24.
Habel
LA
,
Shak
S
,
Jacobs
MK
,
Capra
A
,
Alexander
C
,
Pho
M
, et al
A population-based study of tumor gene expression and risk of breast cancer death among lymph node-negative patients
.
Breast Cancer Res
2006
;
8
:
R25
.
25.
Buyse
M
,
Loi
S
,
van't Veer
L
,
Viale
G
,
Delorenzi
M
,
Glas
AM
, et al
Validation and clinical utility of a 70-gene prognostic signature for women with node-negative breast cancer
.
J Natl Cancer Inst
2006
;
98
:
1183
92
.
26.
Mook
S
,
Knauer
M
,
Bueno-de-Mesquita
JM
,
Retel
VP
,
Wesseling
J
,
Linn
SC
, et al
Metastatic potential of T1 breast cancer can be predicted by the 70-gene MammaPrint signature
.
Ann Surg Oncol
2010
;
17
:
1406
13
.
27.
Curtis
C
,
Shah
SP
,
Chin
S-F
,
Turashvili
G
,
Rueda
OM
,
Dunning
MJ
, et al
The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups
.
Nature
2012
;
486
:
346
52
.
28.
Joensuu
H
,
Kellokumpu-Lehtinen
P-L
,
Bono
P
,
Alanko
T
,
Kataja
V
,
Asola
R
, et al
Adjuvant docetaxel or vinorelbine with or without trastuzumab for breast cancer
.
N Engl J Med
2006
;
354
:
809
20
.
29.
Slamon
D
,
Eiermann
W
,
Robert
N
,
Pienkowski
T
,
Martin
M
,
Press
M
, et al
Adjuvant trastuzumab in HER2-positive breast cancer
.
N Engl J Med
2011
;
365
:
1273
83
.
30.
Piccart-Gebhart
MJ
,
Procter
M
,
Leyland-Jones
B
,
Goldhirsch
A
,
Untch
M
,
Smith
I
, et al
Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer
.
N Engl J Med
2005
;
353
:
1659
72
.
31.
Romond
EH
,
Perez
EA
,
Bryant
J
,
Suman
VJ
,
Geyer
CE
 Jr
,
Davidson
NE
, et al
Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer
.
N Engl J Med
2005
;
353
:
1673
84
.
32.
Zardavas
D
,
Ades
F
,
Spasojevic
IB
,
Pugliano
L
,
Capelan
M
,
Paesmans
M
, et al
Controversial issues in early-stage breast cancer: a global collaborative survey, supported by the European Society for Medical Oncology (ESMO)
.
Ann Oncol
2014
;
25
:
1558
62
.
33.
Rodrigues
MJ
,
Wassermann
J
,
Albiges
L
,
Brain
E
,
Delaloge
S
,
Stevens
D
, et al
Trastuzumab treatment in t1ab, node-negative, human epidermal growth factor receptor 2-overexpressing breast carcinomas
.
J Clin Oncol
2010
;
28
:
e541
2
.
34.
Tolaney
SM
,
Barry
WT
,
Dang
CT
,
Yardley
DA
,
Moy
B
,
Marcom
PK
, et al
A phase II study of adjuvant paclitaxel and trastuzumab (apt trial) for node-negative, Her2-positive breast cancer. a phase ii study of adjuvant paclitaxel and trastuzumab (apt trial) for node-negative, HER2-positive breast cancer [abstract]
.
Cancer Res
2013
;
73
:
S1
04
.
35.
Loi
S
,
Sirtaine
N
,
Piette
F
,
Salgado
R
,
Viale
G
,
Van Eenoo
F
, et al
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
2013
;
31
:
860
7
.
36.
Loi
S
,
Michiels
S
,
Salgado
R
,
Sirtaine
N
,
Jose
V
,
Fumagalli
D
, et al
Tumor infiltrating lymphocytes is prognostic and predictive for trastuzumab benefit in early breast cancer: results from the FinHER trial
.
Ann Oncol
2014
;
25
:
1544
50
.
37.
Metzger-Filho
O
,
Giobbie-Hurder
A
,
Mallon
EA
,
Viale
G
,
Winer
EP
,
Thurlimann
BJK
, et al
Relative effectiveness of letrozole alone or in sequence with tamoxifen for patients diagnosed with invasive lobular carcinoma
.
J Clin Oncol
31
, 
2013
.
38.
Yu
K-D
,
Zhu
R
,
Zhan
M
,
Rodriguez
AA
,
Yang
W
,
Wong
S
, et al
Identification of prognosis-relevant subgroups in patients with chemoresistant triple-negative breast cancer
.
Clin Cancer Res
2013
;
19
:
2723
33
.
39.
Masuda
H
,
Baggerly
KA
,
Wang
Y
,
Zhang
Y
,
Gonzalez-Angulo
AM
,
Meric-Bernstam
F
, et al
Differential response to neoadjuvant chemotherapy among 7 triple-negative breast cancer molecular subtypes
.
Clin Cancer Res
2013
;
19
:
5533
40
.
40.
Lehmann
BD
,
Bauer
JA
,
Chen
X
,
Sanders
ME
,
Chakravarthy
AB
,
Shyr
Y
, et al
Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies
.
J Clin Invest
2011
;
121
:
2750
67
.
41.
Gradishar
WJ
,
Anderson
BO
,
Blair
SL
,
Burstein
HJ
,
Cyr
A
,
Elias
AD
, et al
Breast cancer version 3.2014
.
J Natl Compr Cancer Netw
2014
;
12
:
542
90
.
42.
Chen
X
,
Li
J
,
Gray
WH
,
Lehmann
BD
,
Bauer
JA
,
Shyr
Y
, et al
TNBCtype: a subtyping tool for triple-negative breast cancer
.
Cancer Inform
2012
;
11
:
147
56
.
43.
Gucalp
A
,
Traina
TA
. 
Triple-negative breast cancer: role of the androgen receptor
.
Cancer J
2010
;
16
:
62
5
.
44.
Werner
M
,
Chott
A
,
Fabiano
A
,
Battifora
H
. 
Effect of formalin tissue fixation and processing on immunohistochemistry
.
Am J Surg Pathol
2000
;
24
:
1016
9
.
45.
Pritt
B
,
Tessitore
JJ
,
Weaver
DL
,
Blaszyk
H
. 
The effect of tissue fixation and processing on breast cancer size
.
Hum Pathol
2005
;
36
:
756
60
.
46.
Provencher
L
,
Diorio
C
,
Hogue
J-C
,
Doyle
C
,
Jacob
S
. 
Does breast cancer tumor size really matter that much?
Breast Edinb Scotl
2012
;
21
:
682
5
.
47.
Fehrenbacher
L
,
Capra
AM
,
Quesenberry
CP
,
Fulton
R
,
Shiraz
P
,
Habel
LA
. 
Distant invasive breast cancer recurrence risk in human epidermal growth factor receptor 2-positive T1a and T1b node-negative localized breast cancer diagnosed from 2000 to 2006: a cohort from an integrated health care delivery system.
J Clin Oncol
2014
;
32
:
2151
8
.