HER2+ early breast cancer is a heterogeneous disease, comprising all the intrinsic breast cancer subtypes. The only biomarker available nowadays for anti-HER2 treatment selection is HER2 status itself, but estrogen receptor (ER) status is emerging as a robust predictive marker within HER2+ disease. In this Perspective, we discuss the biological and clinical differences between patients with HER2+/ER-positive (ER+) disease versus those with HER2+/ER-negative (ER-neg) tumors, namely, short-term and long-term (>5 years after diagnosis) prognosis, response to neoadjuvant treatment and benefit from adjuvant anti-HER2–targeted therapies. We also address other possible biomarkers to be used for patient selection in future clinical trials, such as gene signatures, PAM50 subtypes, tumor-infiltrating lymphocytes, PIK3CA mutations, and changes in Ki67 score during treatment and discuss their limitations. Finally, we suggest new clinical trial designs that can have an impact on clinical practice, aiming to test treatment deescalation separately for patients with HER2+/ER+ and HER2+/ER-neg tumors. We also propose an integrated classification of HER2+ disease, comprising DNA, RNA, protein expression, and microenvironment characteristics, in order to identify those tumors that are truly “HER2-addicted” and may benefit the most from anti-HER2 treatment.

Translational Relevance

HER2+ early breast cancer is a heterogeneous disease, in which estrogen receptor (ER) status is emerging as a robust predictive marker. This is related to the fact that the ER and HER2 pathways drive breast cancer proliferation by a complex interaction of cellular signaling processes, including the sharing of intracellular effectors such as MAPK, PI3K, and Src3. Thus, due to the cross-talk between HER2 and ER, treatments targeting one pathway can induce the activation of the other. This leads to a different clinical behavior of HER2+/ER+ disease as compared with HER2+/ER-neg early breast cancer. We discuss how the timing and sequencing of treatments, including anti-HER2 targeted therapy, can influence their benefit according to ER status. We also propose different deescalation trial designs, with potential impact on clinical practice, separately for HER2+/ER+ and HER2+/ER-neg breast cancer, which should be regarded as distinctly different.

It is well known that HER2+ breast cancer is a heterogeneous disease (1, 2). Besides HER2 status itself, there are no validated biomarkers to identify patients likely to benefit from anti-HER2 treatments (3). It has been proposed, however, that the expression of the estrogen receptor (ER) drives variable degrees of benefit from existing anti-HER2 treatment strategies. An increasing number of trials are being developed targeting specifically HER2+/ER-positive (ER+) or HER2+/ER-negative (ER-neg) early breast cancer, and we believe that, in the future, these may be regarded as distinct diseases or we may need additional tools to identify the varying biology within HER2+ disease that is represented by ER status.

Biological differences

Distinct biological characteristics may justify the different clinical behavior observed in HER2+ early breast cancer according to ER status. The ER and the HER2 pathways drive breast cancer proliferation by a complex interaction of cellular signaling processes (4). Due to the intracellular cross-talk between HER2 and ER, treatments interfering with one of the two pathways can have an effect on the other, with reactivation of ER signaling being a known mechanism of resistance to anti-HER2 agents in HER2+/ER+ breast cancer (5).

The distribution of intrinsic (PAM50) breast cancer subtypes within HER2+ tumors also differs according to ER status (6): around 75% of HER2+/ER-neg tumors and 30% of HER2+/ER+ are “HER2-enriched,” which is the subtype associated with HER2/EGFR pathway activation (2), high proliferation rates, and an immune-activated stroma with elevated tumor-infiltrating lymphocytes (TIL) concentrations (Fig. 1). On the other hand, around 70% of HER2+/ER+ tumors are luminal A/B, which are ER-dependent tumors, with low HER2/EGFR pathway activation and a high rate of PIK3CA mutations (1), that are associated with lower response to anti-HER2 treatment (7).

Figure 1.

Current and future classification of HER2+ disease. HER2+ tumors can be divided between estrogen receptor (ER)–positive and ER-negative subgroups, according to the immunohistochemical expression of ER. Yet, at the molecular level, these tumors can be divided into the four molecular subtypes (luminal A, luminal B, HER2-enriched and basal-like), showing different molecular alterations, cell pathway activation, and immune microenvironment. This leads to different responses to anti-HER2 treatment, as well as to chemotherapy and endocrine therapy. ER, estrogen receptor; HER2-E, HER2-enriched molecular subtype; LumA, luminal A molecular subtype; LumB, luminal B molecular subtype; mut, mutation; TILs, tumor-infiltrating lymphocytes; WT, wild-type.

Figure 1.

Current and future classification of HER2+ disease. HER2+ tumors can be divided between estrogen receptor (ER)–positive and ER-negative subgroups, according to the immunohistochemical expression of ER. Yet, at the molecular level, these tumors can be divided into the four molecular subtypes (luminal A, luminal B, HER2-enriched and basal-like), showing different molecular alterations, cell pathway activation, and immune microenvironment. This leads to different responses to anti-HER2 treatment, as well as to chemotherapy and endocrine therapy. ER, estrogen receptor; HER2-E, HER2-enriched molecular subtype; LumA, luminal A molecular subtype; LumB, luminal B molecular subtype; mut, mutation; TILs, tumor-infiltrating lymphocytes; WT, wild-type.

Close modal

Differences in prognosis between HER2+/ER+ and HER2+/ER-neg early breast cancer in the “trastuzumab era”

In the first 5 years after diagnosis, HER2+/ER-neg patients have worse survival compared with the HER2+/ER+ group. Among 42,887 patients treated between 2010 and 2015, 5-year overall survival (OS) rates were 88.2% in HER2+/ER+ versus 83.9% in HER2+/ER-neg subgroup (P < 0.001; ref. 8). Yet, after 5 years, recurrence risk lowers among HER2+/ER-neg patients, but is maintained among those with HER2+/ER+ tumors (9–11). This leads to similar long-term prognosis: in the HERA trial, 12-year OS in the trastuzumab arms was 81% in the HER2+/ER+ and 78%–79% in the HER2+/ER-neg subgroups (9).

On the other hand, achievement of a pathologic complete response (pCR) after neoadjuvant treatment is a prognostic factor in both groups, but it has a stronger impact on event-free survival (EFS) in the HER2+/ER-neg [hazard ratio (HR), 0.25] compared with the HER2+/ER+ subgroups (HR, 0.58; ref. 12). However, the overall median follow-up for EFS in this meta-analysis was just 5.4 years; thus, the prognostic impact of pCR on late recurrences is currently unknown.

Is ER status predictive of treatment benefit?

It is well documented that HER2+/ER-neg patients treated with chemotherapy and dual anti-HER2 therapy present higher pCR rates (61%–91%) compared with patients with HER2+/ER+ early breast cancer (26%–56%; refs. 13–20); and, compared with HER2+/ER+ tumors, HER2+/ER-neg tumors have consistently demonstrated greater increases in pCR rates with dual anti-HER2 therapy plus chemotherapy compared with chemotherapy/trastuzumab alone (13, 18–20). Nonetheless, duration of anti-HER2 treatment seems to matter in HER2+/ER+ early breast cancer: in TBCRC023, increasing treatment length with trastuzumab/lapatinib and letrozole from 12 to 24 weeks led to an increase in pCR rates from 9% to 33%, whereas no significant difference was observed in HER2+/ER-neg tumors (21). This suggests that in HER2+/ER+ tumors, in the absence of chemotherapy, prolonged inhibition of the HER2 and ER pathways may be necessary not only to decrease proliferation, but to induce apoptosis as well. Yet, adding concomitant endocrine therapy (ET) to anti-HER2 therapy and/or chemotherapy did not yield any advantage in the NSABP B-52 or ADAPT HER2+/HR+ studies (22, 23). This is intriguing, as one would expect that the ER/HER2 pathway blockade would suppress their cross-talk and lead to higher tumor response.

On the opposite end of the spectrum, the high chemosensitivity of HER2+/ER-neg tumors was demonstrated in the ADAPT HER2+/HR study, in which the addition of paclitaxel to trastuzumab/pertuzumab increased pCR rates from 36% to 91% (17). Similarly, the KRISTINE trial showed a significantly higher pCR rate in this group with docetaxel/carboplatin plus trastuzumab/pertuzumab compared with T-DM1/pertuzumab (73% vs. 54%, respectively; ref. 16).

In the adjuvant setting, HER2+/ER+ and HER2+/ER-neg subgroups have a similar degree of proportional risk reduction with trastuzumab (9, 24, 25). Yet, in contrast with the neoadjuvant setting, duration of adjuvant trastuzumab treatment might matter more for HER2+/ER-neg early breast cancer: in a recent meta-analysis, patients receiving 9 weeks to 6 months of trastuzumab had worse EFS compared with patients treated for 12 months (26). Therefore, in HER2+/ER-neg disease, longer trastuzumab treatment is probably more effective than a shorter course. However, the trials included in this meta-analysis had a median follow-up of less than 5 years, so long-term data on HER2+/ER+ patients are needed.

Challenging this idea, the ExteNET trial showed that sequential anti-HER2 blockade (summing 2 years of treatment) was beneficial for HER2+/ER+ patients, but not for HER2+/ER-neg patients (27). On the other hand, updated survival results from APHINITY show that early dual HER2 blockade with trastuzumab/pertuzumab has an effect both in HER2+/ER-neg and HER2+/ER+ diseases (28). Reasons for this difference may lie in these drugs' mechanisms of action, timing of administration, and length of follow-up. After stopping adjuvant trastuzumab, HER2+/ER+ patients continue ET, but the single blocking of the ER pathway may lead to reactivation of the HER2 pathway on residual trastuzumab-resistant malignant cells (4). As neratinib was administered after trastuzumab, it may have prevented the reactivation of the HER2 pathway and, hence, benefited HER2+/ER+ patients, but not those with HER2+/ER-neg tumors. As neratinib irreversibly inhibits several HER-family members (EGFR, HER2, HER4), it may have prevented this cross-talk in a more effective way than administration of trastuzumab for 2 years (which only inhibits HER2; ref. 9). In APHINITY, the early dual blockade has probably eliminated trastuzumab-resistant cells in part of the patients, but the benefit was seen later in HER2+/ER+ patients, as these tend to have later relapses, as compared with HER2+/ER-neg patients.

Recently, the KATHERINE trial showed that in patients with residual disease after neoadjuvant treatment, 1 year of adjuvant T-DM1 was superior to trastuzumab, independently of ER status (29). It should be noted that this trial included selected high-risk patients, for whom suboptimal response to neoadjuvant trastuzumab could have influenced the benefit from a salvage treatment with a different anti-HER2 drug, potentially confounding the effect of baseline ER status.

How can we identify patients who benefit more or less from anti-HER2 treatment?

The biggest question nowadays is how to select patients for escalation/deescalation approaches, maintaining or increasing pCR rates and survival, with minimal toxicity. Therefore, the quest for finding prognostic and predictive biomarkers has been intense (3). One attempt was to quantify gene-expression levels of ESR1 and ERBB2, showing that high levels of ERBB2 (20, 30–32) and low levels of ESR1 (31) are associated with higher pCR rates. Expression levels of genes involved in HER2 and ER pathways were also combined in a predictive model in an analysis of the NSABP B-31 trial. Patients were classified into three groups: (i) “high ESR1”/“low-intermediate ERBB2”: no benefit from trastuzumab (HR, 1.58); (ii) “low ESR1”/“low-intermediate ERBB2”: moderate benefit (HR, 0.60); (iii) “low-intermediate ESR1”/“high ERBB2”: large benefit (HR, 0.28; ref. 33). This eight-gene signature, showing no linear correlation between ERBB2 and ESR1 mRNA levels and benefit from adjuvant trastuzumab, was later validated in the N9831 and FB-7 trials (34, 35). Nonetheless, these intriguing findings have not yet been tested in prospective studies or applied in the clinic.

Other biomarker studies focused on the correlation between PAM50 subtypes and benefit from anti-HER2 treatment. A consistent finding is that HER2-enriched tumors present higher pCR rates (between 28% and 72%) when treated with anti-HER2 therapies (with/without chemotherapy), compared with other subtypes (20, 32, 36, 37), showing that HER2-enriched subtype predicts sensitivity to anti-HER2 treatment. In fact, several studies show that ER status loses its association with pCR when subtype is taken into account, indicating that subtype captures most of the information displayed by ER status and also brings additional information (20, 32, 36, 37). One important aspect to take into account when designing deescalation trials is that HER2-enriched subtype is also chemosensitive, with overall pCR rates in HER2+ disease of around 60% with versus 30% without chemotherapy, respectively (20, 32, 36, 37).

It has also been proposed that high scores of a gene-expression signature of loss of function of RB1 may identify HER2+ tumors more likely to respond to chemotherapy, and this concept has been validated in HER2+/ER+ tumors using a refined 87-gene signature (RBsig; refs. 38, 39). Moreover, higher TIL levels are associated with improved pCR rates and survival in both HER2+/ER-neg and HER2+/ER+ early breast cancer (40, 41), and evidence of immune activation by RNA signatures is independently associated with higher pCR (20, 31). It has also been demonstrated that high TIL levels (>75%) predicted benefit from adjuvant trastuzumab/pertuzumab versus trastuzumab alone (42). Of note, TILs levels tend to be lower in HER2+/ER+ compared with HER2+/ER-neg tumors (40).

A decrease in Ki67 during neoadjuvant treatment is a surrogate for response and prognosis in HER2-negative/ER+ early breast cancer, so its role is being evaluated in HER2+/ER+ disease (43). In ADAPT HER2+/HR+, patients with an early tumor response (Ki67 decrease ≥30% or low-cellular tumor at 3 weeks) had a higher pCR rate compared with nonresponders (36% vs. 20%, respectively; ref. 23). In the PerELISA trial, HER2+/ER+ patients with a Ki67 drop after 2 weeks of letrozole (molecular responders) continued on letrozole, and trastuzumab/pertuzumab were added for another 12 weeks. Nonresponders were switched to paclitaxel with trastuzumab/pertuzumab. The pCR rate was 21% among molecular responders, but even more remarkable was the 81% pCR rate among nonresponders (44). Even though the correlation between an early Ki67 drop and long-term survival is not yet validated in patients with HER2+ tumors, it could become a biomarker for selecting patients with HER2+/ER+ who do not respond to ET and would be candidates to chemotherapy, given the impressive pCR rate achieved in this subgroup.

Thus, a dilemma for future clinical trials is how to select patients: should one be guided by HER2/ER immunohistochemistry status, by molecular subtype or by a combination of both? Based on current evidence, tumors with the highest anti-HER2 sensitivity are those HER2+ by immunohistochemistry/ISH and the PAM50 HER2 enriched. Whether additional biomarkers will help identify higher anti-HER2 sensitivity is currently unknown, although TILs and PIK3CA mutations arise as promising candidates (Fig. 2). Indeed, tumors with high HER2 amplification and an intact PI3K pathway are especially sensitive to HER2-targeted therapies (30). TILs could also potentially be used to select patients, but there are no standardized cutoff points to define “high” versus “low” TIL infiltration (45). Another question is if an early biopsy to assess Ki67 levels should be performed in HER2+/ER+ patients receiving neoadjuvant treatment. This is limited by Ki67 low reproducibility (46) and absence of validation as a surrogate for long-term survival benefit in HER2+ early breast cancer.

Figure 2.

Combining biomarkers in HER2+ disease. Currently, protein expression assessed by immunohistochemistry is the standard method to classify breast cancer into different subtypes, according to tumor's HER2 and estrogen receptor expression. However, an integrated classification of HER2-positive breast tumors, based on their DNA, RNA, protein expression, and microenvironment, is needed to implement more precise and guided treatments and to identify those tumors “addicted” to the HER2 cellular pathway. Here, we propose a definition of “HER2-addicted” tumors that can be further refined in the future, as more data become available. ER, estrogen receptor; HER2-E, HER2-enriched molecular subtype; PR, progesterone receptor; RBsig, RB loss-of-function signature; WT, wild-type.

Figure 2.

Combining biomarkers in HER2+ disease. Currently, protein expression assessed by immunohistochemistry is the standard method to classify breast cancer into different subtypes, according to tumor's HER2 and estrogen receptor expression. However, an integrated classification of HER2-positive breast tumors, based on their DNA, RNA, protein expression, and microenvironment, is needed to implement more precise and guided treatments and to identify those tumors “addicted” to the HER2 cellular pathway. Here, we propose a definition of “HER2-addicted” tumors that can be further refined in the future, as more data become available. ER, estrogen receptor; HER2-E, HER2-enriched molecular subtype; PR, progesterone receptor; RBsig, RB loss-of-function signature; WT, wild-type.

Close modal

Deescalating strategies: a plea for separate trials according to ER status

Despite distinct biological and clinical characteristics between HER2+/ER-neg and HER2+/ER+ early breast cancer, they are still treated similarly, except for ET. Nevertheless, different deescalation strategies for HER2+ early breast cancer might be explored according to ER status: (i) to decrease anti-HER2 treatment duration, (ii) to avoid anthracyclines, and (iii) to design chemotherapy-free regimens.

As detailed before, a meta-analysis showed a worse outcome with a shorter treatment duration of adjuvant trastuzumab in patients with HER2+/ER-neg disease, but this comparison was nonsignificant in patients with HER2+/ER+ tumors. Moreover, in the PERSEPHONE trial (the only trial so far proving its noninferiority hypothesis of 6 months vs 1 year of trastuzumab), the HR for the comparison of the two strategies was very close to 1 in the HER2+/ER+ subgroup (HR, 0.91; 95% confidence interval, 0.68–1.21; ref. 47). Therefore, in patients with HER2+/ER+ tumors, a shorter duration of trastuzumab treatment may be seen as a safe option by some—nonetheless, due to the late relapse profile of these tumors, longer follow-up data are needed before adopting this strategy.

The APT trial (in which patients with ≤3 cm, node-negative tumors received 12 weeks of paclitaxel and 1 year of trastuzumab) showed a 7-year invasive disease-free survival of 94.6% for HER2+/ER+ disease and of 90.7% for HER2+/ER-neg disease, with just four distant recurrences (1%; ref. 48). Thus, this treatment deescalation seems very safe for patients with low-burden tumors. Given the high chemosensitivity of HER2+/ER-neg tumors, leading to a higher rate of pCR, a possible strategy for avoiding anthracyclines in patients with somewhat larger tumors would be to assess the tumor's sensitivity upfront with neoadjuvant double HER2-blockade plus a taxane. The DeCrescendo trial will test this hypothesis in patients with cT1c-T2N0 HER2+/ER-neg tumors, who will receive 12 weeks of taxanes plus trastuzumab/pertuzumab. Those who achieve pCR will be spared from adjuvant anthracyclines and will only receive double HER2-blockade until completion of 1 year of treatment. The study is powered to test 3-year relapse-free survival (RFS) rate, with a narrow confidence interval, primarily in patients with PAM50 HER2-enriched tumors and then in the overall population.

The reduced chemosensitivity of HER2+/ER+ early breast cancer fuels the interest in alternative, targeted approaches without chemotherapy. One is the use of CDK4/6 inhibitors, which synergize with trastuzumab in HER2+/ER+ tumor models (49). In the small NA-PHER2 study, 20 weeks of palbociclib/ET/trastuzumab/pertuzumab provided a significant reduction in tumor proliferation, but led to a modest pCR rate of 27% (50). Long-term survival results and efficacy according to molecular subtypes are awaited. There are ongoing trials [PALTAN (NCT02907918); TOUCH (NCT03644186)] exploring this approach. In particular, TOUCH is assessing RBsig (39) as a potential biomarker to identify HER2+/ER+ patients who could be spared from chemotherapy and treated with ET/anti-HER2/palbociclib. On the other hand, a recent joint analysis of neoadjuvant “chemotherapy-free” trials (SOLTI-PAMELA, TBCRC023/TBCRC006, and PerELISA) has shown that the combination of the HER2-enriched subtype with a high mRNA expression of ERBB2 leads to the identification of a subgroup of patients exquisitely sensitive to double HER2 blockade—with pCR rates ranging from 44.5% to 66.7% (51). Among HER2+/ER+ patients, only 32% were “HER2-enriched/ERBB2-high,” whereas among the HER2+/ER-neg subgroup, there were 68% of patients with this profile. Hence, as the HER2+/ER-neg subgroup is clearly enriched with these HER2-addicted tumors, one could envision a trial restricted to HER2+/ER-neg patients: they would be screened upfront and those with a HER2-enriched/ERBB2-high expression tumor would be proposed to receive neoadjuvant trastuzumab/pertuzumab only. Like in the DeCrescendo trial, patients achieving pCR would be spared of any adjuvant chemotherapy and the primary endpoint would be a high RFS rate with a narrow confidence interval.

Given the predictive value of ER status, it would be reasonable for the next wave of clinical trials to target separately HER2+/ER+ and HER2+/ER-neg patients. This would lead to a better tailoring of deescalation treatment strategies, and hence to a greater chance of “success.” In addition, this could greatly reduce the morbidity and costs associated with the long and intense systemic regimens that patients with HER2+ tumors still receive, despite years of biomarker research. Thus, besides the use of ER status, we feel that an integrated classification of HER2+ tumors, based on their DNA, RNA, protein expression, and immune microenvironment, would also be useful for further treatment refinement in the future.

M. Brandão reports receiving other commercial research support and speakers bureau honoraria from Roche/GNE. R. Caparica reports receiving speakers bureau honoraria from AstraZeneca, Boehringer Ingelheim, and Janssen, and is an unpaid consultant/advisory board member for Pfizer and AstraZeneca. L. Malorni reports receiving commercial research grants from Pfizer and Novartis, and reports receiving speakers bureau honoraria from Novartis. A. Prat is a paid consultant for Roche, Oncolytics Biotech, Daiichi Sankyo, Novartis, Pfizer, and Nanostring Technologies, and reports receiving commercial research grants from Novartis, Nanostring Technologies, Incyte, and Puma. L.A. Carey is an unpaid consultant/advisory board member for Roche/Genentech, Novartis, and Seattle Genetics. No potential conflicts of interest were disclosed by the other authors.

Conception and design: M. Brandão, R. Caparica, M. Piccart

Development of methodology: Not applicable

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): Not applicable

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): Not applicable

Writing, review, and/or revision of the manuscript: M. Brandão, R. Caparica, L. Malorni, A. Prat, L.A. Carey, M. Piccart

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): Not applicable

Study supervision: M. Piccart

M. Brandão and R. Caparica acknowledge the support of Institut Jules Bordet, which funded their research fellowship.

1.
Ferrari
A
,
Vincent-Salomon
A
,
Pivot
X
,
Sertier
A-S
,
Thomas
E
,
Tonon
L
, et al
A whole-genome sequence and transcriptome perspective on HER2-positive breast cancers
.
Nat Commun
2016
;
7
:
12222
.
2.
Prat
A
,
Carey
LA
,
Adamo
B
,
Vidal
M
,
Tabernero
J
,
Cortés
J
, et al
Molecular features and survival outcomes of the intrinsic subtypes within HER2-positive breast cancer
.
J Natl Cancer Inst
2014
;
106
. pii: dju152.
3.
Gingras
I
,
Gebhart
G
,
de Azambuja
E
,
Piccart-Gebhart
M
. 
HER2-positive breast cancer is lost in translation: time for patient-centered research
.
Nat Rev Clin Oncol
2017
;
14
:
669
81
.
4.
Giuliano
M
,
Trivedi
MV
,
Schiff
R
. 
Bidirectional crosstalk between the estrogen receptor and human epidermal growth factor receptor 2 signaling pathways in breast cancer: molecular basis and clinical implications
.
Breast Care
2013
;
8
:
256
62
.
5.
Wang
Y-C
,
Morrison
G
,
Gillihan
R
,
Guo
J
,
Ward
RM
,
Fu
X
, et al
Different mechanisms for resistance to trastuzumab versus lapatinib in HER2-positive breast cancers – role of estrogen receptor and HER2 reactivation
.
Breast Cancer Res
2011
;
13
:
R121
.
6.
Cejalvo
JM
,
Pascual
T
,
Fernández-Martínez
A
,
Adamo
B
,
Chic
N
,
Vidal
M
, et al
1727PDistribution of the PAM50 breast cancer subtypes within each pathology-based group: a combined analysis of 15,339 patients across 29 studies
.
Ann Oncol
2017
;
28
(suppl_5): v595–v604.
7.
Loibl
S
,
Majewski
I
,
Guarneri
V
,
Nekljudova
V
,
Holmes
E
,
Bria
E
, et al
PIK3CA mutations are associated with reduced pathological complete response rates in primary HER2-positive breast cancer: pooled analysis of 967 patients from five prospective trials investigating lapatinib and trastuzumab
.
Ann Oncol
2016
;
27
:
1519
25
.
8.
Hwang
K-T
,
Kim
J
,
Jung
J
,
Chang
JH
,
Chai
YJ
,
Oh
SW
, et al
Impact of breast cancer subtypes on prognosis of women with operable invasive breast cancer: a population-based study using SEER database
.
Clin Cancer Res
2019
;
25
:
1970
9
.
9.
Cameron
D
,
Piccart-Gebhart
MJ
,
Gelber
RD
,
Procter
M
,
Goldhirsch
A
,
de Azambuja
E
, et al
11 years' follow-up of trastuzumab after adjuvant chemotherapy in HER2-positive early breast cancer: final analysis of the HERceptin Adjuvant (HERA) trial
.
Lancet
2017
;
389
:
1195
205
.
10.
Chumsri
S
,
Li
Z
,
Serie
DJ
,
Mashadi-Hossein
A
,
Colon-Otero
G
,
Song
N
, et al
Incidence of late relapses in patients with HER2-positive breast cancer receiving adjuvant trastuzumab: combined analysis of NCCTG N9831 (Alliance) and NRG oncology/NSABP B-31
.
J Clin Oncol
2019
;
37
:
3425
35
.
11.
Lambertini
M
,
Campbell
C
,
Gelber
RD
,
Viale
G
,
McCullough
A
,
Hilbers
F
, et al
Dissecting the effect of hormone receptor status in patients with HER2-positive early breast cancer: exploratory analysis from the ALTTO (BIG 2-06) randomized clinical trial
.
Breast Cancer Res Treat
2019
;
177
:
103
14
.
12.
Cortazar
P
,
Zhang
L
,
Untch
M
,
Mehta
K
,
Costantino
JP
,
Wolmark
N
, et al
Pathological complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis
.
Lancet
2014
;
384
:
164
72
.
13.
Gianni
L
,
Pienkowski
T
,
Im
Y-H
,
Roman
L
,
Tseng
L-M
,
Liu
M-C
, et al
Efficacy and safety of neoadjuvant pertuzumab and trastuzumab in women with locally advanced, inflammatory, or early HER2-positive breast cancer (NeoSphere): a randomised multicentre, open-label, phase 2 trial
.
Lancet Oncol
2012
;
13
:
25
32
.
14.
Schneeweiss
A
,
Chia
S
,
Hickish
T
,
Harvey
V
,
Eniu
A
,
Hegg
R
, et al
Pertuzumab plus trastuzumab in combination with standard neoadjuvant anthracycline-containing and anthracycline-free chemotherapy regimens in patients with HER2-positive early breast cancer: a randomized phase II cardiac safety study (TRYPHAENA)
.
Ann Oncol
2013
;
24
:
2278
84
.
15.
Loibl
S
,
Jackisch
C
,
Schneeweiss
A
,
Schmatloch
S
,
Aktas
B
,
Denkert
C
, et al
Dual HER2-blockade with pertuzumab and trastuzumab in HER2-positive early breast cancer: a subanalysis of data from the randomized phase III GeparSepto trial
.
Ann Oncol
2017
;
28
:
497
504
.
16.
Hurvitz
SA
,
Martin
M
,
Symmans
WF
,
Jung
KH
,
Huang
C-S
,
Thompson
AM
, et al
Neoadjuvant trastuzumab, pertuzumab, and chemotherapy versus trastuzumab emtansine plus pertuzumab in patients with HER2-positive breast cancer (KRISTINE): a randomised, open-label, multicentre, phase 3 trial
.
Lancet Oncol
2018
;
19
:
115
26
.
17.
Nitz
UA
,
Gluz
O
,
Christgen
M
,
Grischke
E-M
,
Augustin
D
,
Kuemmel
S
, et al
De-escalation strategies in HER2-positive early breast cancer (EBC): final analysis of the WSG-ADAPT HER2+/HR- phase II trial: efficacy, safety, and predictive markers for 12 weeks of neoadjuvant dual blockade with trastuzumab and pertuzumab ± weekly paclitaxel
.
Ann Oncol
2017
;
28
:
2768
72
.
18.
Baselga
J
,
Bradbury
I
,
Eidtmann
H
,
Di Cosimo
S
,
de Azambuja
E
,
Aura
C
, et al
Lapatinib with trastuzumab for HER2-positive early breast cancer (NeoALTTO): a randomised, open-label, multicentre, phase 3 trial
.
Lancet
2012
;
379
:
633
40
.
19.
Robidoux
A
,
Tang
G
,
Rastogi
P
,
Geyer
CE
,
Azar
CA
,
Atkins
JN
, et al
Lapatinib as a component of neoadjuvant therapy for HER2-positive operable breast cancer (NSABP protocol B-41): an open-label, randomised phase 3 trial
.
Lancet Oncol
2013
;
14
:
1183
92
.
20.
Carey
LA
,
Berry
D
,
Cirrincione
CT
,
Barry
WT
,
Pitcher
B
,
Harris
LN
, et al
Molecular heterogeneity and response to neoadjuvant human epidermal growth factor receptor 2 targeting in CALGB 40601, a randomized phase III trial of paclitaxel plus trastuzumab with or without lapatinib
.
J Clin Oncol
2016
;
34
:
542
9
.
21.
Rimawi
MF
,
Niravath
PA
,
Wang
T
,
Rexer
B
,
Forero
A
,
Wolff
AC
, et al
Abstract S6-02: TBCRC023: a randomized multicenter phase II neoadjuvant trial of lapatinib plus trastuzumab, with endcorine therapy and without chemotherapy, for 12 vs. 24 weeks in patients with HER2 overexpressing breast cancer
.
Cancer Res
2015
;
75
:
S6
02
.
22.
Rimawi
MF
,
Cecchini
RS
,
Rastogi
P
,
Geyer
CE
,
Fehrenbacher
L
,
Stella
PJ
, et al
Abstract S3-06: a phase III trial evaluating pCR in patients with HR+, HER2-positive breast cancer treated with neoadjuvant docetaxel, carboplatin, trastuzumab, and pertuzumab (TCHP) ± estrogen deprivation: NRG Oncology/NSABP B-52
.
Cancer Res
2017
;
77
:
S3
06
.
23.
Harbeck
N
,
Gluz
O
,
Christgen
M
,
Kates
RE
,
Braun
M
,
Küemmel
S
, et al
De-escalation strategies in human epidermal growth factor receptor 2 (HER2)-positive early breast cancer (BC): final analysis of the West German study group adjuvant dynamic marker-adjusted personalized therapy trial optimizing risk assessment and therapy response prediction in early BC HER2- and hormone receptor-positive phase II randomized trial-efficacy, safety, and predictive markers for 12 weeks of neoadjuvant trastuzumab emtansine with or without endocrine therapy (ET) versus trastuzumab plus ET
.
J Clin Oncol
2017
;
35
:
3046
54
.
24.
Perez
EA
,
Romond
EH
,
Suman
VJ
,
Jeong
J-H
,
Sledge
G
,
Geyer
CE
, et al
Trastuzumab plus adjuvant chemotherapy for human epidermal growth factor receptor 2-positive breast cancer: planned joint analysis of overall survival from NSABP B-31 and NCCTG N9831
.
J Clin Oncol
2014
;
32
:
3744
52
.
25.
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
.
26.
Niraula
S
,
Gyawali
B
. 
Optimal duration of adjuvant trastuzumab in treatment of early breast cancer: a meta-analysis of randomized controlled trials
.
Breast Cancer Res Treat
2019
;
173
:
103
9
.
27.
Martin
M
,
Holmes
FA
,
Ejlertsen
B
,
Delaloge
S
,
Moy
B
,
Iwata
H
, et al
Neratinib after trastuzumab-based adjuvant therapy in HER2-positive breast cancer (ExteNET): 5-year analysis of a randomised, double-blind, placebo-controlled, phase 3 trial
.
Lancet Oncol
2017
;
18
:
1688
700
.
28.
Piccart
M
,
Procter
M
,
Fumagalli
D
,
de Azambuja
E
,
Clark
E
,
Ewer
MS
, et al
Interim overall survival analysis of APHINITY (BIG 4-11): a randomized multicenter, double-blind, placebo-controlled trial comparing chemotherapy plus trastuzumab plus pertuzumab versus chemotherapy plus trastuzumab plus placebo as adjuvant therapy in patients with operable HER2-positive early breast cancer
.
San Antonio Breast Cancer Symposium
2019
;
80
:
GS1
04
.
29.
von Minckwitz
G
,
Huang
C-S
,
Mano
MS
,
Loibl
S
,
Mamounas
EP
,
Untch
M
, et al
Trastuzumab emtansine for residual invasive HER2-positive breast cancer
.
N Engl J Med
2019
;
380
:
617
28
.
30.
Veeraraghavan
J
,
Angelis
CD
,
Mao
R
,
Wang
T
,
Herrera
S
,
Pavlick
AC
, et al
A combinatorial biomarker predicts pathologic complete response to neoadjuvant lapatinib and trastuzumab without chemotherapy in patients with HER2+ breast cancer
.
Ann Oncol
2019
;
30
:
927
33
.
31.
Fumagalli
D
,
Venet
D
,
Ignatiadis
M
,
Azim
HA
,
Maetens
M
,
Rothé
F
, et al
RNA sequencing to predict response to neoadjuvant anti-HER2 therapy: a secondary analysis of the NeoALTTO randomized clinical trial
.
JAMA Oncol
2016
;
3
:
227
34
.
32.
Llombart-Cussac
A
,
Cortés
J
,
Paré
L
,
Galván
P
,
Bermejo
B
,
Martínez
N
, et al
HER2-enriched subtype as a predictor of pathological complete response following trastuzumab and lapatinib without chemotherapy in early-stage HER2-positive breast cancer (PAMELA): an open-label, single-group, multicentre, phase 2 trial
.
Lancet Oncol
2017
;
18
:
545
54
.
33.
Pogue-Geile
KL
,
Kim
C
,
Jeong
J-H
,
Tanaka
N
,
Bandos
H
,
Gavin
PG
, et al
Predicting degree of benefit from adjuvant trastuzumab in NSABP trial B-31
.
J Natl Cancer Inst
2013
;
105
:
1782
8
.
34.
Pogue-Geile
KL
,
Song
N
,
Serie
DJ
,
Thompson
EA
. 
Abstract PD3-18: The NSABP/NRG 8-gene signature accurately predicts degree of benefit from trastuzumab in Alliance/NCCTG N9831: validation of the 8-gene signature in an independent clinical trial
.
Cancer Res
2018
;
78
:
PD3-18
PD3-18
.
35.
Pogue-Geile
KL
,
Wang
Y
,
Srinivasan
A
,
Gavin
PG
,
Kim
RS
,
Song
N
, et al
Abstract P3-10-04: the fully validated NSABP/NRG 8-gene signature which predicted the degree of benefit in the adjuvant setting (B-31 and NCCTG N9831) associates with pCR in the neoadjuvant setting in NSABP clinical trial FB-7
.
Cancer Res
2019
;
79
:
P3-10-04
P3-10-04
.
36.
Prat
A
,
Angelis
CD
,
Pascual
T
,
Gutierrez
C
,
Wang
T
,
Paré
L
, et al
Abstract P2-09-12: Independent validation of the HER2-enriched subtype as a predictor of pathological complete response following trastuzumab and lapatinib without chemotherapy in early-stage HER2-positive breast cancer
.
Cancer Res
2018
;
78
:
P2-09-12
P2-09-12
.
37.
Prat
A
,
Slamon
D
,
Hurvitz
SA
,
Press
MF
,
Phillips
GL
,
Valverde
VL
, et al
Abstract PD3-06: association of intrinsic subtypes with pathological complete response (pCR) in the KRISTINE neoadjuvant phase 3 clinical trial in HER2-positive early breast cancer (EBC)
.
Cancer Res
2018
;
78
:
PD3-06
PD3-06
.
38.
Witkiewicz
AK
,
Ertel
A
,
McFalls
J
,
Valsecchi
ME
,
Schwartz
G
,
Knudsen
ES
. 
RB-pathway disruption is associated with improved response to neoadjuvant chemotherapy in breast cancer
.
Clin Cancer Res
2012
;
18
:
5110
22
.
39.
Risi
E
,
Grilli
A
,
Migliaccio
I
,
Biagioni
C
,
McCartney
A
,
Guarducci
C
, et al
A gene expression signature of retinoblastoma loss-of-function predicts resistance to neoadjuvant chemotherapy in ER-positive/HER2-positive breast cancer patients
.
Breast Cancer Res Treat
2018
;
170
:
329
41
.
40.
Salgado
R
,
Denkert
C
,
Campbell
C
,
Savas
P
,
Nuciforo
P
,
Nucifero
P
, et al
Tumor-infiltrating lymphocytes and associations with pathological complete response and event-free survival in HER2-positive early-stage breast cancer treated with lapatinib and trastuzumab: a secondary analysis of the NeoALTTO trial
.
JAMA Oncol
2015
;
1
:
448
54
.
41.
Denkert
C
,
von Minckwitz
G
,
Darb-Esfahani
S
,
Lederer
B
,
Heppner
BI
,
Weber
KE
, et al
Tumour-infiltrating lymphocytes and prognosis in different subtypes of breast cancer: a pooled analysis of 3771 patients treated with neoadjuvant therapy
.
Lancet Oncol
2018
;
19
:
40
50
.
42.
Krop
IE
,
Paulson
J
,
Campbell
C
,
Kiermaier
AC
,
Andre
F
,
Fumagalli
D
, et al
Genomic correlates of response to adjuvant trastuzumab (H) and pertuzumab (P) in HER2+ breast cancer (BC): biomarker analysis of the APHINITY trial
.
J Clin Oncol
2019
;
37
:
1012
.
43.
Dowsett
M
,
Smith
IE
,
Ebbs
SR
,
Dixon
JM
,
Skene
A
,
A'Hern
R
, et al
Prognostic value of Ki67 expression after short-term presurgical endocrine therapy for primary breast cancer
.
J Natl Cancer Inst
2007
;
99
:
167
70
.
44.
Guarneri
V
,
Dieci
MV
,
Bisagni
G
,
Frassoldati
A
,
Bianchi
GV
,
De Salvo
GL
, et al
De-escalated treatment with trastuzumab-pertuzumab-letrozole in patients with HR+/HER2+ operable breast cancer with Ki67 response after 2 weeks letrozole: final results of the PerELISA neoadjuvant study
.
J Clin Oncol
2018
;
36
:
507
.
45.
Salgado
R
,
Denkert
C
,
Demaria
S
,
Sirtaine
N
,
Klauschen
F
,
Pruneri
G
, et al
The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: recommendations by an international TILs working group 2014
.
Ann Oncol
2015
;
26
:
259
71
.
46.
Dowsett
M
,
Nielsen
TO
,
A'Hern
R
,
Bartlett
J
,
Coombes
RC
,
Cuzick
J
, et al
Assessment of Ki67 in breast cancer: recommendations from the international Ki67 in breast cancer working group
.
J Natl Cancer Inst
2011
;
103
:
1656
64
.
47.
Earl
HM
,
Hiller
L
,
Vallier
A-L
,
Loi
S
,
Howe
D
,
Higgins
HB
, et al
PERSEPHONE: 6 versus 12 months of adjuvant trastuzumab in patients (pts) with HER2 positive (+) early breast cancer (EBC): randomised phase 3 non-inferiority trial with definitive 4-year (yr) disease-free survival (DFS) results
.
J Clin Oncol
2018
;
36
:
506
.
48.
Earl
HM
,
Hiller
L
,
Vallier
A-L
,
Loi
S
,
McAdam
K
,
Hughes-Davies
L
, et al
6 versus 12 months of adjuvant trastuzumab for HER2-positive early breast cancer (PERSEPHONE): 4-year disease-free survival results of a randomised phase 3 non-inferiority trial
.
Lancet
2019
;
393
:
2599
612
.
49.
Goel
S
,
Wang
Q
,
Watt
AC
,
Tolaney
SM
,
Dillon
DA
,
Li
W
, et al
Overcoming therapeutic resistance in HER2-positive breast cancers with CDK4/6 inhibitors
.
Cancer Cell
2016
;
29
:
255
69
.
50.
Gianni
L
,
Bisagni
G
,
Colleoni
M
,
Del Mastro
L
,
Zamagni
C
,
Mansutti
M
, et al
Neoadjuvant treatment with trastuzumab and pertuzumab plus palbociclib and fulvestrant in HER2-positive, ER-positive breast cancer (NA-PHER2): an exploratory, open-label, phase 2 study
.
Lancet Oncol
2018
;
19
:
249
56
.
51.
Prat
A
,
Pascual
T
,
De Angelis
C
,
Gutierrez
C
,
Llombart-Cussac
A
,
Wang
T
, et al
HER2-enriched subtype and ERBB2 expression in HER2-positive breast cancer treated with dual HER2 blockade
.
J Natl Cancer Inst
2010
;
112
:
46
54
.