Skip to main content
Erschienen in: Breast Cancer Research 1/2014

Open Access 01.02.2014 | Research article

Phosphorylated p-70S6K predicts tamoxifen resistance in postmenopausal breast cancer patients randomized between adjuvant tamoxifen versus no systemic treatment

verfasst von: Karin Beelen, Mark Opdam, Tesa M Severson, Rutger HT Koornstra, Andrew D Vincent, Jelle Wesseling, Jettie J Muris, Els MJJ Berns, Jan B Vermorken, Paul J van Diest, Sabine C Linn

Erschienen in: Breast Cancer Research | Ausgabe 1/2014

Abstract

Introduction

Activation of the phosphatidylinositol-3-kinase (PI3K) and/or mitogen-activated protein kinase (MAPK) pathways results in anti-estrogen resistance in vitro, but a biomarker with clinical validity to predict intrinsic resistance has not been identified. In metastatic breast cancer patients with previous exposure to endocrine therapy, the addition of a mammalian target of rapamycine (mTOR) inhibitor has been shown to be beneficial. Whether or not patients on adjuvant endocrine treatment might benefit from these drugs is currently unclear. A biomarker that predicts intrinsic resistance could potentially be used as companion diagnostic in this setting. We tested the clinical validity of different downstream-activated proteins in the PI3K and/or MAPK pathways to predict intrinsic tamoxifen resistance in postmenopausal primary breast cancer patients.

Methods

We recollected primary tumor tissue from patients who participated in a randomized trial of adjuvant tamoxifen (1–3 years) versus observation. After constructing a tissue micro-array, cores from 563 estrogen receptor α positive were immunostained for p-AKT(Thr308), p-AKT(Ser473), p-mTOR, p-p706SK and p-ERK1/2. Cox proportional hazard models for recurrence free interval were used to assess hazard ratios and interactions between these markers and tamoxifen treatment efficacy.

Results

Interactions were identified between tamoxifen and p-AKT(Thr308), p-mTOR, p-p70S6K and p-ERK1/2. Applying a conservative level of significance, p-p70S6K remained significantly associated with tamoxifen resistance. Patients with p-p70S6K negative tumors derived significant benefit from tamoxifen (HR 0.24, P < 0.0001), while patients whose tumor did express p-p70S6K did not (HR = 1.02, P =0.95), P for interaction 0.004. In systemically untreated breast cancer patients, p-p70S6K was associated with a decreased risk for recurrence.

Conclusions

Patients whose tumor expresses p-p70S6K, as a marker of downstream PI3K and/or MAPK pathway activation, have a favorable prognosis, but do not benefit from adjuvant tamoxifen. A potential benefit from inhibitors of the PI3K/Akt/mTOR pathway in these patients needs to be further explored.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​bcr3598) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

KB, SCL and ADV were responsible for the concept and design of the study. MO, RHTK, JBV, PJvD, JW, and JJM contributed substantially to acquisition of the data. KB, ADV, TMS, EMJJB, SCL, JW and PJvD contributed to the analysis and interpretation of the data. KB, with supervision from SCL, drafted the manuscript. All authors critically revised the manuscript for important intellectual content and approved the final version.
Abkürzungen
CI
confidence interval
ERα
estrogen receptor alpha
ERK
extracellular signal-regulated kinase
HER2
human epidermal growth factor receptor 2
HR
hazard ratio
IHC
immunohistochemistry
MAPK
mitogen-activated protein kinase
mTOR
mammalian target of rapamycine
p
phosphorylated
PgR
progesterone receptor
PI3K
phosphatidylinositol-3-kinase
TMA
tissue micro-array.

Introduction

Activation of the mitogen-activated protein kinase (MAPK) and phosphatidylinositol-3-kinase (PI3K) pathways confers anti-estrogen resistance in vitro[13]. Apart from activation by the canonical pathway drivers (PIK3CA mutations [4], loss of PTEN [5] or overexpression of growth factor receptors such as human epidermal growth factor receptor 2 (HER2) [6] and insulin like growth factor 1 receptor [7]), in vitro data have shown that the PI3K pathway can also be activated in response to estrogen depletion. This results in acquired hormone-resistant breast cancer cells that are sensitive to PI3K/mammalian target of rapamycine (mTOR) inhibition [8]. These preclinical data support the clinical observation that estrogen receptor alpha (ERα)-positive metastatic breast cancer patients with prior exposure to aromatase inhibitors derive substantial benefit from the addition of an mTOR inhibitor [9]. Whether or not patients who are primarily resistant to adjuvant endocrine therapy might benefit from PI3K and/or MAPK pathway inhibition remains to be defined. A biomarker of an activated PI3K and/or MAPK pathway with clinical validity to predict resistance in the adjuvant setting has not been identified [10], but could potentially be used as a companion diagnostic for non-ERα-targeted drugs, such as an mTOR inhibitor.
Several canonical pathway drivers, such as PIK3CA mutations [11, 12], loss of PTEN [12], and HER2 [13], have been studied for their validity to predict resistance. However, none of these drivers significantly predicts lack of benefit from endocrine therapy. An important issue to be considered is that the presence of these drivers in clinical samples may not necessarily result in high activation of downstream proteins [12, 14]. In vitro, PI3K pathway activation leads to phosphorylation of AKT and subsequently of mTOR and p70S6K [10]. Phosphorylation of extracellular signal-regulated kinase (ERK)1/2 is a result of MAPK pathway activation [15] and mediates activation of p70S6K [16]. However, relatively moderate activation of the PI3K pathway was seen in tumors with a PIK3CA exon 20 mutation-associated gene signature [14]. In addition, in a large series of primary breast cancer tumors, reverse-phase protein analysis did not show activation of the typical downstream proteins in the PI3K pathway in PIK3CA mutated luminal A tumors [17]. The activation status of downstream proteins rather than the presence or absence of a canonical driver therefore probably ultimately defines anti-estrogen sensitivity in breast cancer patients.
We hypothesized that activated proteins downstream in the PI3K and/or MAPK kinase pathways could potentially be used as a marker that separates patients who are likely to benefit from adjuvant tamoxifen treatment from those who are primarily resistant to this drug. The aim of our study was therefore to investigate the predictive value of different downstream activated proteins in the PI3K and/or MAPK pathways in a large series of ERα-positive postmenopausal breast cancer patients randomized between adjuvant tamoxifen versus no systemic treatment.

Methods

Patients and materials

We have recollected primary tumor tissue blocks from stage I to III postmenopausal breast cancer patients who were randomized (2:1) between 1 year of tamoxifen (30 mg/day) versus no adjuvant therapy (IKA trial, 1982 to 1994) [18, 19]. Study data were part of the Oxford meta-analysis [20]. After 1989, based on two interim analyses showing a significant improvement in recurrence-free survival in lymph node-positive patients, node-positive patients in this trial skipped the first randomization and all received 1 year of tamoxifen. After 1 year a second randomization was performed to receive another 2 years of tamoxifen or to stop further treatment. In total, 1,662 patients were included. None of these patients received adjuvant chemotherapy. The patient characteristics and clinical outcome of the original study group (1,662 patients) have been presented elsewhere [19].
Sufficient tumor material was available for 739 patients, who did not differ in prognostic factors from the total group (Table S1 in Additional file 1). After revision of ERα status as assessed with immunohistochemistry (IHC), a total of 563 ERα-positive tumors were used for subsequent analysis. We used a cutoff value ≥10% of positive tumor cells for ERα positivity, since this is common practice in the Netherlands and also this would avoid the potential inclusion of basal-like tumors [21] in our analysis. The original trial was approved by the central ethics committee of the Netherlands Cancer Institute and informed consent was obtained from all study participants. For this retrospective translational study, no additional consent was required according to Dutch legislation [22] since the use of archival pathology left-over material does not interfere with patient care. Tumor tissue was handled according to the Dutch code of conduct for dealing responsibly with human tissue in the context of health research [23].

Immunohistochemistry

Tissue microarrays (TMAs) were constructed using formalin-fixed paraffin-embedded tumor blocks. A total of three (0.6 mm) cores per tumor were embedded in the TMAs that were stained for ERα, progesterone receptor (PgR) and HER2. ERα and PgR were considered positive when ≥10% of invasive cells showed nuclear reactivity. HER2 was considered positive when membranous staining was DAKO score 3 [24]. In the case of DAKO score 2, chromogenic in situ hybridization was performed. For tumors without sufficient cores in the TMA, whole slides were cut and assessed for ERα (n = 60), PgR (n = 55) and HER2 (n = 36). The tumor grade was scored on a hematoxylin and eosin-stained slide using the modified Bloom–Richardson score [25].
Antibodies used for immunohistochemistry of downstream phosphorylated (p) proteins are shown in Table S2 in Additional file 1. For p-AKT(Ser473), antigen retrieval was performed using citrate buffer and slides were incubated overnight with antibody (dilution 1:50). All other phospho-protein stainings (p-AKT(Thr308), p-mTOR, p-ERK1/2 and p-p70S6K) were performed using a standardized protocol on the Ventana Benchmark® Ultra system (Ventana Medical Systems, Tucson, USA). To ensure phospho-specificity of the antibodies, for each antibody a test TMA containing positive cores was dephosphorylated by λ-phosphatase before staining, resulting in disappearance of the positive staining (Figure S1 in Additional file 1).
Cytoplasmic intensity (0 to 3) was assessed for p-AKT(Ser473), p-AKT(Thr308) and p-p70S6K. The percentage of tumor cells with submembranous staining was scored for p-mTOR, and the proportion of positive nuclei was scored for p-ERK1/2. For each staining, one of the TMAs was quantified independently in a blinded manner by a second observer to calculate inter-observer variability. For further analyses, we used the scores produced by the first observer (MO).
Since the stability of phospho-proteins is a matter of debate [26], we tested whether the relative age of tumor samples (divided in quartiles) was associated with quantitative phospho-protein staining for each of these markers. In addition, to test for a possible effect of different fixation procedures, we tested whether the inclusion center was associated with differences in quantitative phospho-protein expression.

Statistical analysis

The recurrence-free interval was defined as the time from the date of first randomization until the occurrence of a local, regional or distant recurrence or breast cancer-specific death [27]. Since a secondary contralateral breast tumor cannot be inferred from the molecular make-up of the primary tumor, while the other type of events can be inferred in relation to tamoxifen resistance of the primary tumor, this was not considered an event and these patients were censored at the date of this occurrence. The association between expression of downstream activated proteins and known prognostic factors was tested using Fisher’s exact test.
We hypothesized that high expression of downstream activated proteins is associated with tamoxifen resistance. Our primary analysis was therefore to test whether tamoxifen benefit was dependent on any of the downstream activated proteins in the PI3K and/or MAPK pathway. We analyzed these markers as a binary factor, using the median level as the cutoff value. Adjusted Cox proportional hazard regression analyses were performed including an interaction variable. Covariates included age (≥65 vs. <65), grade (grade 3 vs. grade 1 to 2), tumor size (T3 to T4 vs. T1 to T2), HER2 status (positive vs. negative), and PgR status (positive vs. negative). All survival analyses were stratified for nodal status. Owing to the multiple co-primary endpoints of this study, we apply a more conservative level of significance (α = 0.01) when assessing the interactions.
Further exploratory analyses examined tamoxifen benefit when the markers were implemented as continuous linear variables. For those continuous linear variables that showed an interaction with tamoxifen treatment, we explored which level of dichotomization best predicted tamoxifen benefit, by comparing Akaike’s information criteria of the Cox proportional hazards models for all possible cutoff values. In addition, based on knowledge derived from preclinical studies [1, 3], we explored whether a composed variable of either high p-ERK1/2 or high p-mTOR – indicating the activation of either the MAPK pathway or the PI3K pathway – was associated with tamoxifen resistance. To assess the prognostic value of the phospho-proteins, we analyzed their putative prognostic potential in the subgroup of patients who were randomized to the control arm. The reason why we did not use all patients and corrected for tamoxifen treatment is that this correction would assume that all ERα-positive breast cancer patients would derive similar benefit from tamoxifen. Since the phospho-protein might be associated with tamoxifen resistance, simply correcting for the assumed tamoxifen benefit without a correction for a potential interaction between treatment and phospho-protein could bias the analysis for prognostic potential. Survival curves were constructed using the Kaplan–Meier method. This study complied with reporting recommendations for tumor marker prognostic studies (REMARK) criteria [28] as outlined in Table S3 in Additional file 1.

Results

Association of downstream activated proteins in the PI3K and/or MAPK pathways with clinico-pathological factors

The inter-observer variability analyzed using the (weighted) Cohen’s kappa coefficient is depicted in Table S4 in Additional file 1. Figure S2 in Additional file 1 shows the number of evaluable cases for each downstream protein. We did not find a significant difference in median phospho-protein expression and relative age of tumor samples (Figure S3a,b,c,d,e in Additional file 1). Median phospho-protein expression was not significantly different among inclusion centers (Figure S4a,b,c,d,e in Additional file 1).
We did not find a significant association between p-p70S6K and any of the known clinico-pathological variables (Table 1). Significant associations were found between p-p70S6K and all other downstream activated proteins (Table 1). The association between other downstream activated proteins and known prognostic variables is shown in Table S5 in Additional file 1. High p-mTOR and positive p-ERK1/2 staining were both associated with a positive PgR status and low tumor grade. In addition, high expression of p-AKT(Ser473) was associated with a positive PgR status.
Table 1
Associations between p-p70S6K and clinico-pathological variables as well as other PI3K and/or MAPK pathway proteins
 
p-p70S6K (n = 438)
 
 
Negative (n = 188)
Positive (n = 250)
P value
Treatment
No tamoxifen
40 (21)
55 (22)
0.92a
Tamoxifen 1 year
89 (47)
113 (45)
Tamoxifen 3 years
59 (31)
82 (33)
Age
<65
82 (44)
128 (51)
0.12b
≥65
106 (56)
122 (49)
Lymph node
Negative
92 (49)
141 (56)
0.12b
Positive
96 (51)
109 (44)
T stage
T1 to T2
167 (89)
221 (88)
1.00b
T3 to T4
21 (11)
29 (12)
Grade
Grade 1 to 2
123 (65)
153 (61)
0.37b
Grade 3
65 (35)
97 (39)
Progesterone receptor
Negative
98 (52)
111 (44)
0.10b
Positive
88 (47)
139 (56)
Missing
2 (1)
0 (0)
HER2
Negative
168 (89)
223 (89)
0.17b
Positive
12 (6)
26 (10)
Missing
8 (4)
1 (0)
p-AKT(Ser473)c
Low (0 to 1)
109 (71)
53 (24)
<0.0001b
High (2 to 3)
44 (29)
172 (76)
p-AKT(Thr308)c
Negative
143 (81)
87 (36)
<0.0001b
Positive
34 (20)
153 (64)
p-mTORc
Low (0 to 59%)
156 (87)
176 (73)
0.0003b
High (≥60%)
23 (13)
66 (27)
p-ERK1/2c
Negative
114 (65)
61 (26)
<0.0001b
 
Positive
61 (35)
177 (74)
 
Data presented as n (%). ERK, extracellular signal-regulated kinase; HER2, human epidermal growth factor receptor 2; MAPK, mitogen-activated protein kinase; mTOR, mammalian target of rapamycine; PI3K, phosphatidylinositol-3-kinase; p, phosphorylated. aLinear by linear test. bFisher’s exact test (analysis based on cases without missing values). cMissing data not shown.

Downstream activated proteins in the PI3K and/or MAPK pathways predict resistance to tamoxifen

In the total group of ERα-positive patients (n = 563), a total of 132 recurrence-free interval events occurred (Table S6 in Additional file 1). The number of patients in each treatment arm pre and post interim analysis is shown in Figure S5 in Additional file 1. The median follow-up of patients without a recurrence event is 7.8 years. When stratified by nodal status, the hazard ratio (HR) for tamoxifen versus control in this cohort was 0.54 (95% confidence interval (CI) = 0.36 to 0.83, P = 0.004). In our primary analysis, using the median expression levels as the cutoff value for a binary factor, we did not find a significant interaction between either p-AKT(Ser473) or p-mTOR and tamoxifen. In addition, the interactions between both p-AKT(Thr308) and p-ERK1/2 and tamoxifen were nonsignificant (adjusted P for interaction 0.09 and 0.06 respectively) (Table 2). However, we observed a significant interaction for p-p70S6K with tamoxifen (P = 0.004). Patients whose tumor did not express p-p70S6K derived significant benefit from tamoxifen (HR = 0.24, 95% CI = 0.12 to 0.47, P < 0.0001), while patients whose tumor did express p-p70S6K had no benefit (multivariate HR = 1.02, 95% CI = 0.48 to 2.21, P = 0.95) (Figure 1A; Table S7 in Additional file 1). Performing the same analysis using a 1% cutoff value for ERα positivity added four extra patients and did not substantially change these results (data not shown).
Table 2
Adjusted hazard ratios and tests for interaction between PI3K and/or MAPK pathway markers and tamoxifen
 
Variable level
Adjusted hazard ratioafor tamoxifen versus control (95% confidence interval)
Adjusted P value for interaction
p-AKT (Ser473)
Below median (0 to 1)
0.34 (0.17 to 0.69)
0.16
Above median (2 to 3)
0.68 (0.33 to 1.38)
p-AKT (Thr308)
Below median (0)
0.42 (0.24 to 0.74)
0.09
Above median (1 to 3)
1.03 (0.43 to 2.50)
p-mTOR
Below median (0 to 20%)
0.41 (0.23 to 0.71)
0.25
Above median (21 to 100%)
0.74 (0.31 to 1.76)
p-ERK1/2
Below median (0)
0.34 (0.18 to 0.63)
0.06
Above median (1 to 100%)
0.87 (0.40 to 1.87)
p-p70S6K
Below median (0)
0.24 (0.13 to 0.47)
0.004
 
Above median (1 to 3)
1.02 (0.48 to 2.21)
 
ERK, extracellular signal-regulated kinase; MAPK, mitogen-activated protein kinase; mTOR, mammalian target of rapamycine; PI3K, phosphatidylinositol-3-kinase; p, phosphorylated. a Recurrence-free interval survival. Models were adjusted for age, T stage, grade, progesterone receptor status and human epidermal growth factor receptor 2 status. Models were stratified for nodal status.
In our exploratory analyses, analyzing the expression of downstream activated proteins as a continuous variable, we observed an interaction with tamoxifen for p-AKT(Thr308) (adjusted P = 0.03), p-mTOR (adjusted P = 0.03) as well as p-p70S6K (adjusted P = 0.006) (Table S8 in Additional file 1). Examining Akaike’s information criteria values indicated that a dichotomization of p-mTOR submembranous staining into <60% and ≥60% provided the best fit to the data (Table S9 and Figure S6 in Additional file 1). For p-AKT(Thr308), p-ERK1/2 and p-p70S6K, a dichotomization in any positive versus negative staining (which corresponded with dichotomization with median expression levels as the cutoff values) provided the best fit to the data (Tables S10,S11 and Figures S7,S8 in Additional file 1). Tables S12,S13,S14,S15 in Additional file 1 present the distribution of known prognostic factors over the treatment arms both for patients who were positive and for patients who were negative for these markers.
Based on preclinical knowledge that p-ERK1/2 and p-mTOR represent activation of, respectively, the MAPK pathway and the PI3K pathway, we explored the interaction with tamoxifen for the combination of these markers. Patients with a tumor that expressed either high p-mTOR or positive p-ERK1/2 protein did not benefit from tamoxifen (adjusted HR = 1.00, 95% CI = 0.48 to 2.08, P = 1.00), while patients whose tumor was negative for these two markers did derive significant benefit (adjusted HR = 0.25, 95% CI = 0.13 to 0.48, P < 0.0001) (P for interaction = 0.004) (Figure 1B; Table S16 in Additional file 1).

Downstream activated proteins in the PI3K and/or MAPK pathways are associated with good prognosis in the absence of adjuvant systemic treatment

When systemically untreated breast cancer patients were dichotomized according to p-AKT(Ser473), p-AKT(Thr308), p-mTOR or p-p70S6K status, the group that was positive for the marker (dichotomized according to Akaike’s information criteria) exhibited a decreased risk for recurrence compared with patients whose tumor had no or low expression of the marker (Figure 2 and Table 3; Tables S17,S18,S19,S20 in Additional file 1). A similar trend was seen for patients whose tumor did express p-ERK1/2 (P = 0.14). Hierarchical clustering of the downstream activated proteins in the PI3K and/or MAPK pathway is shown in Figure 3.
Table 3
Adjusted hazard ratios for recurrence-free interval according to PI3K and/or MAPK pathway markers in patients who did not receive adjuvant systemic treatment
 
Number of cases in analysis (events)
Hazard ratioa
95% confidence interval
P value
p-AKT(Ser 473), (2 to 3) versus (0 to 1)
86 (27)
0.30
0.12 to 0.76
0.01
p-AKT(Thr308), positive versus negative
100 (26)
0.30
0.11 to 0.80
0.02
p-mTOR, high (≥60% ) versus low (0 to 59% )
94 (26)
0.11
0.02 to 0.55
0.007
p-ERK1/2, positive versus negative
94 (26)
0.52
0.22 to 1.24
0.14
p-p70S6K, positive versus negative
94 (25)
0.11
0.04 to 0.32
<0.0001
ERK, extracellular signal-regulated kinase; MAPK, mitogen-activated protein kinase; mTOR, mammalian target of rapamycine; PI3K, phosphatidylinositol-3-kinase; p, phosphorylated. aFor each marker, Cox proportional hazard models were performed with co-variables: lymph node status, age, tumor size, grade and progesterone receptor.

Discussion

In this retrospective analysis of tumor samples from ERα-positive postmenopausal breast cancer patients, we have shown the clinical validity of p-p70S6K, as a marker of PI3K and/or MAPK pathway activation, to predict adjuvant tamoxifen resistance. Patients whose tumor expressed p-p70S6K did not benefit from adjuvant tamoxifen, confirming previous in vitro findings [1, 2].
We are aware of the discussion that the use of phospho-specific antibodies for IHC may be challenging, since preanalytic variables (such as fixation technique and duration) may critically affect the signal [26]. Although we did not observe an association between phospho-protein expression and either tumor size, inclusion center or relative age of the tumor sample and confirmed the phospho-specificity of the antibodies, we cannot exclude that unknown preanalytic variables might have affected the results of IHC. Nevertheless, since these unknown variables would have affected both tumor samples from control patients as well as tamoxifen-treated patients, the observed predictive value of p-p70S6K markers is probably not biased by these preanalytic variables. Unlike others [29, 30], we have used automatic immunostainings to improve robustness and reproducibility of these IHC-based tests. Nevertheless, the visual interpretation of IHC-based tests is still a subjective, time-consuming and variable process, with an inherent intra-observer and inter-observer variability [31, 32]. The technical validity of IHC-based tests may therefore be further improved by image analysis applications on digital slides [33]. We deemed a direct comparison of the observed frequencies of breast cancers scoring positive for the downstream-activated proteins analyzed with those observed in other studies [30, 34, 35] not to be appropriate due to differences in study population, antibodies and staining techniques as well as interpretation of these stainings.
Very few studies have tested the predictive validity of downstream-activated proteins in the PI3K and/or MAPK pathway in the context of a randomized clinical trial. Perez-Tenorio and colleagues observed a decreased benefit from tamoxifen only in those patients with PIK3CA mutations who also expressed AKT activation [12], but the test for interaction was not significant. In a subgroup analysis of ERα-positive and PgR-positive patients, Bostner and colleagues recently observed a reduced benefit from tamoxifen in those patients whose tumor expressed high p-mTOR [29]. These results support our data and suggest that downstream activated proteins have a superior clinical validity to predict endocrine resistance compared with the presence or absence of a pathway driver [36]. The strength of our study is that we pre-specified the cutoff value (median expression level) that was used for the primary analysis of the putative marker as a binary factor. In addition, we tested the markers as continuous linear variables and used Aikake’s information criteria to define the optimal cutoff, which coincided with the median for most putative biomarkers. p-p70S6K (as a marker of activation of either the PI3K pathway and/or the MAPK pathway) was the only significant marker according to our conservative definition of significance, although most of the other downstream activated proteins showed a similar trend. In addition, we found a significant interaction for the composed variable of either high p-ERK1/2 and/or p-mTOR expression, representing activated MAPK and PI3K pathways, respectively.
In our study, interestingly, most of the markers of PI3K and/or MAPK activation were associated with a favorable prognosis in the absence of systemic treatment. Although an association with low-grade and positive PgR status was shown, the favorable outcome was independent of these factors in multivariate analysis. The question arises of why these tumors have a favorable prognosis. Although others have studied the association of these markers with prognosis in series of patients treated with endocrine therapy [8, 30, 37], none of these studies discerned prognosis from prediction. In case a putative biomarker under investigation results in a reduced treatment efficacy, this would cancel out a potential prognostic effect if the biomarker is analyzed in series of patients who are all treated with the drug [10]. A direct comparison between our data and these studies is therefore not possible.
Our study has several limitations. First of all, for retrospective biomarker analysis we could only use the subgroup of 563 ERα-positive patients from whom sufficient tumor material was available. However, this subgroup did not differ from the total study population. Furthermore, the duration of tamoxifen therapy differs from the current duration of at least 5 years. However, we anticipate that the relative effects of the biomarkers analyzed in this study will be similar for shorter and longer duration of endocrine therapy. The patients in our study randomized to adjuvant treatment received tamoxifen only (and no aromatase inhibitors), while currently most ERα-positive postmenopausal breast cancer patients receive an aromatase inhibitor preceding or following tamoxifen treatment. Since the putative predictive biomarkers studied are considered to cause escape from hormone dependency [8], our data are thought to be applicable to current clinical practice as well. Furthermore, the patients in our trial had not received adjuvant chemotherapy and thereby the observed effects of the putative biomarkers were not biased by endocrine-resistant patients who were cured by adjuvant chemotherapy. In current clinical practice, patients who relapse despite adjuvant endocrine therapy and adjuvant chemotherapy are both endocrine therapy resistant and chemotherapy resistant. The adjuvant endocrine therapy resistance may be explained by activation of the PI3K/MAPK pathway and our data suggest that p-p70S6K may identify those patients.
It would be clinically relevant to determine whether p-p70S6K could be used as a companion diagnostic for the addition of non-ERα-targeted drugs (like PI3K/mTOR inhibitors) to endocrine therapy in the adjuvant setting. In metastatic breast cancer patients, who had been randomized between tamoxifen alone or in combination with everolimus, a trend for a better treatment efficacy was observed for patients with a tumor with high p-p70S6K [38]. Randomized clinical trials in the adjuvant setting testing the addition of everolimus to hormonal therapy are currently recruiting patients [39], and these studies are of great importance to test the treatment predictive value of p-p70S6K in this setting.

Conclusion

With the approval of everolimus for metastatic breast cancer patients with acquired endocrine therapy resistance, the first non-ERα-targeted drug that can overcome endocrine therapy resistance has become clinically available. Whether or not inhibitors of the PI3K/Akt/mTOR pathway may overcome endocrine therapy resistance in the adjuvant setting is currently under investigation. A biomarker of an activated PI3K and/or MAPK pathway with clinical validity to predict endocrine therapy resistance in the adjuvant setting has not been identified [10], but such a biomarker could potentially be used as a companion diagnostic for these non-ERα-targeted drugs. In our series of ERα-positive postmenopausal breast cancer patients, p-p70S6K is significantly associated with intrinsic tamoxifen resistance. Patients whose tumor expresses p-p70S6K, as a marker of downstream PI3K and/or MAPK pathway activation, have a favorable prognosis but do not benefit from adjuvant tamoxifen. It would be of great importance to validate these findings in other randomized series with endocrine therapy and to further explore this marker as a potential companion diagnostic.

Acknowledgements

The authors thank all pathology departments throughout the Netherlands for submission of formalin-fixed paraffin-embedded tumor blocks. They thank Philip Schouten for his support with preparation of Figure 3. This work was supported by grants from TI Pharma (project number T3-502) and from A Sister’s Hope.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

KB, SCL and ADV were responsible for the concept and design of the study. MO, RHTK, JBV, PJvD, JW, and JJM contributed substantially to acquisition of the data. KB, ADV, TMS, EMJJB, SCL, JW and PJvD contributed to the analysis and interpretation of the data. KB, with supervision from SCL, drafted the manuscript. All authors critically revised the manuscript for important intellectual content and approved the final version.
Anhänge

Authors’ original submitted files for images

Literatur
1.
Zurück zum Zitat Campbell RA, Bhat-Nakshatri P, Patel NM, Constantinidou D, Ali S, Nakshatri H: Phosphatidylinositol 3-kinase/AKT-mediated activation of estrogen receptor alpha: a new model for anti-estrogen resistance. J Biol Chem. 2001, 276: 9817-9824. 10.1074/jbc.M010840200.CrossRefPubMed Campbell RA, Bhat-Nakshatri P, Patel NM, Constantinidou D, Ali S, Nakshatri H: Phosphatidylinositol 3-kinase/AKT-mediated activation of estrogen receptor alpha: a new model for anti-estrogen resistance. J Biol Chem. 2001, 276: 9817-9824. 10.1074/jbc.M010840200.CrossRefPubMed
2.
Zurück zum Zitat Clark AS, West K, Streicher S, Dennis PA: Constitutive and inducible Akt activity promotes resistance to chemotherapy, trastuzumab, or tamoxifen in breast cancer cells. Mol Cancer Ther. 2002, 1: 707-717.PubMed Clark AS, West K, Streicher S, Dennis PA: Constitutive and inducible Akt activity promotes resistance to chemotherapy, trastuzumab, or tamoxifen in breast cancer cells. Mol Cancer Ther. 2002, 1: 707-717.PubMed
3.
Zurück zum Zitat Shou J, Massarweh S, Osborne CK, Wakeling AE, Ali S, Weiss H, Schiff R: Mechanisms of tamoxifen resistance: increased estrogen receptor-HER2/neu cross-talk in ER/HER2-positive breast cancer. J Natl Cancer Inst. 2004, 96: 926-935. 10.1093/jnci/djh166.CrossRefPubMed Shou J, Massarweh S, Osborne CK, Wakeling AE, Ali S, Weiss H, Schiff R: Mechanisms of tamoxifen resistance: increased estrogen receptor-HER2/neu cross-talk in ER/HER2-positive breast cancer. J Natl Cancer Inst. 2004, 96: 926-935. 10.1093/jnci/djh166.CrossRefPubMed
4.
Zurück zum Zitat Isakoff SJ, Engelman JA, Irie HY, Luo J, Brachmann SM, Pearline RV, Cantley LC, Brugge JS: Breast cancer-associated PIK3CA mutations are oncogenic in mammary epithelial cells. Cancer Res. 2005, 65: 10992-11000. 10.1158/0008-5472.CAN-05-2612.CrossRefPubMed Isakoff SJ, Engelman JA, Irie HY, Luo J, Brachmann SM, Pearline RV, Cantley LC, Brugge JS: Breast cancer-associated PIK3CA mutations are oncogenic in mammary epithelial cells. Cancer Res. 2005, 65: 10992-11000. 10.1158/0008-5472.CAN-05-2612.CrossRefPubMed
5.
Zurück zum Zitat Parsons R, Simpson L: PTEN and cancer. Methods Mol Biol. 2003, 222: 147-166.PubMed Parsons R, Simpson L: PTEN and cancer. Methods Mol Biol. 2003, 222: 147-166.PubMed
6.
Zurück zum Zitat Stancovski I, Peles E, Ben LR, Lemprecht R, Kelman Z, Goldman-Michael R, Hurwitz E, Bacus S, Sela M, Yarden Y: Signal transduction by the neu/erbB-2 receptor: a potential target for anti-tumor therapy. J Steroid Biochem Mol Biol. 1992, 43: 95-103. 10.1016/0960-0760(92)90192-L.CrossRefPubMed Stancovski I, Peles E, Ben LR, Lemprecht R, Kelman Z, Goldman-Michael R, Hurwitz E, Bacus S, Sela M, Yarden Y: Signal transduction by the neu/erbB-2 receptor: a potential target for anti-tumor therapy. J Steroid Biochem Mol Biol. 1992, 43: 95-103. 10.1016/0960-0760(92)90192-L.CrossRefPubMed
7.
Zurück zum Zitat Zhang Y, Moerkens M, Ramaiahgari S, de Bont H, Price L, Meerman J, van de Water B: Elevated insulin-like growth factor 1 receptor signaling induces anti-estrogen resistance through the MAPK/ERK and PI3K/Akt signaling routes. Breast Cancer Res. 2011, 13: R52-10.1186/bcr2883.CrossRefPubMedPubMedCentral Zhang Y, Moerkens M, Ramaiahgari S, de Bont H, Price L, Meerman J, van de Water B: Elevated insulin-like growth factor 1 receptor signaling induces anti-estrogen resistance through the MAPK/ERK and PI3K/Akt signaling routes. Breast Cancer Res. 2011, 13: R52-10.1186/bcr2883.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Miller TW, Hennessy BT, Gonzalez-Angulo AM, Fox EM, Mills GB, Chen H, Higham C, Garcia-Echeverria C, Shyr Y, Arteaga CL: Hyperactivation of phosphatidylinositol-3 kinase promotes escape from hormone dependence in estrogen receptor-positive human breast cancer. J Clin Invest. 2010, 120: 2406-2413. 10.1172/JCI41680.CrossRefPubMedPubMedCentral Miller TW, Hennessy BT, Gonzalez-Angulo AM, Fox EM, Mills GB, Chen H, Higham C, Garcia-Echeverria C, Shyr Y, Arteaga CL: Hyperactivation of phosphatidylinositol-3 kinase promotes escape from hormone dependence in estrogen receptor-positive human breast cancer. J Clin Invest. 2010, 120: 2406-2413. 10.1172/JCI41680.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Baselga J, Campone M, Piccart M, Burris HA, Rugo HS, Sahmoud T, Noguchi S, Gnant M, Pritchard KI, Lebrun F, Beck JT, Ito Y, Yardley D, Deleu I, Perez A, Bachelot T, Vittori L, Xu Z, Mukhopadhyay P, Lebwohl D, Hortobagyi GN: Everolimus in postmenopausal hormone-receptor-positive advanced breast cancer. N Engl J Med. 2011, 366: 520-529.CrossRefPubMed Baselga J, Campone M, Piccart M, Burris HA, Rugo HS, Sahmoud T, Noguchi S, Gnant M, Pritchard KI, Lebrun F, Beck JT, Ito Y, Yardley D, Deleu I, Perez A, Bachelot T, Vittori L, Xu Z, Mukhopadhyay P, Lebwohl D, Hortobagyi GN: Everolimus in postmenopausal hormone-receptor-positive advanced breast cancer. N Engl J Med. 2011, 366: 520-529.CrossRefPubMed
10.
Zurück zum Zitat Beelen K, Zwart W, Linn SC: Can predictive biomarkers in breast cancer guide adjuvant endocrine therapy?. Nat Rev Clin Oncol. 2012, 9: 529-541. 10.1038/nrclinonc.2012.121.CrossRefPubMed Beelen K, Zwart W, Linn SC: Can predictive biomarkers in breast cancer guide adjuvant endocrine therapy?. Nat Rev Clin Oncol. 2012, 9: 529-541. 10.1038/nrclinonc.2012.121.CrossRefPubMed
11.
Zurück zum Zitat Ellis MJ, Lin L, Crowder R, Tao Y, Hoog J, Snider J, Davies S, DeSchryver K, Evans DB, Steinseifer J, Bandaru R, Liu W, Gardner H, Semiglazov V, Watson M, Hunt K, Olson J, Baselga J: Phosphatidyl-inositol-3-kinase alpha catalytic subunit mutation and response to neoadjuvant endocrine therapy for estrogen receptor positive breast cancer. Breast Cancer Res Treat. 2010, 119: 379-390. 10.1007/s10549-009-0575-y.CrossRefPubMedPubMedCentral Ellis MJ, Lin L, Crowder R, Tao Y, Hoog J, Snider J, Davies S, DeSchryver K, Evans DB, Steinseifer J, Bandaru R, Liu W, Gardner H, Semiglazov V, Watson M, Hunt K, Olson J, Baselga J: Phosphatidyl-inositol-3-kinase alpha catalytic subunit mutation and response to neoadjuvant endocrine therapy for estrogen receptor positive breast cancer. Breast Cancer Res Treat. 2010, 119: 379-390. 10.1007/s10549-009-0575-y.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Perez-Tenorio G, Alkhori L, Olsson B, Waltersson MA, Nordenskjold B, Rutqvist LE, Skoog L, Stal O: PIK3CA mutations and PTEN loss correlate with similar prognostic factors and are not mutually exclusive in breast cancer. Clin Cancer Res. 2007, 13: 3577-3584. 10.1158/1078-0432.CCR-06-1609.CrossRefPubMed Perez-Tenorio G, Alkhori L, Olsson B, Waltersson MA, Nordenskjold B, Rutqvist LE, Skoog L, Stal O: PIK3CA mutations and PTEN loss correlate with similar prognostic factors and are not mutually exclusive in breast cancer. Clin Cancer Res. 2007, 13: 3577-3584. 10.1158/1078-0432.CCR-06-1609.CrossRefPubMed
13.
Zurück zum Zitat Dowsett M, Houghton J, Iden C, Salter J, Farndon J, A’Hern R, Sainsbury R, Baum M: Benefit from adjuvant tamoxifen therapy in primary breast cancer patients according oestrogen receptor, progesterone receptor, EGF receptor and HER2 status. Ann Oncol. 2006, 17: 818-826. 10.1093/annonc/mdl016.CrossRefPubMed Dowsett M, Houghton J, Iden C, Salter J, Farndon J, A’Hern R, Sainsbury R, Baum M: Benefit from adjuvant tamoxifen therapy in primary breast cancer patients according oestrogen receptor, progesterone receptor, EGF receptor and HER2 status. Ann Oncol. 2006, 17: 818-826. 10.1093/annonc/mdl016.CrossRefPubMed
14.
Zurück zum Zitat Loi S, Haibe-Kains B, Majjaj S, Lallemand F, Durbecq V, Larsimont D, Gonzalez-Angulo AM, Pusztai L, Symmans WF, Bardelli A, Ellis P, Tutt AN, Gillett CE, Hennessy BT, Mills GB, Phillips WA, Piccart MJ, Speed TP, McArthur GA, Sotiriou C: PIK3CA mutations associated with gene signature of low mTORC1 signaling and better outcomes in estrogen receptor-positive breast cancer. Proc Natl Acad Sci USA. 2010, 107: 10208-10213. 10.1073/pnas.0907011107.CrossRefPubMedPubMedCentral Loi S, Haibe-Kains B, Majjaj S, Lallemand F, Durbecq V, Larsimont D, Gonzalez-Angulo AM, Pusztai L, Symmans WF, Bardelli A, Ellis P, Tutt AN, Gillett CE, Hennessy BT, Mills GB, Phillips WA, Piccart MJ, Speed TP, McArthur GA, Sotiriou C: PIK3CA mutations associated with gene signature of low mTORC1 signaling and better outcomes in estrogen receptor-positive breast cancer. Proc Natl Acad Sci USA. 2010, 107: 10208-10213. 10.1073/pnas.0907011107.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Roskoski R: ERK1/2 MAP kinases: structure, function, and regulation. Pharmacol Res. 2012, 66: 105-143. 10.1016/j.phrs.2012.04.005.CrossRefPubMed Roskoski R: ERK1/2 MAP kinases: structure, function, and regulation. Pharmacol Res. 2012, 66: 105-143. 10.1016/j.phrs.2012.04.005.CrossRefPubMed
16.
Zurück zum Zitat Iijima Y, Laser M, Shiraishi H, Willey CD, Sundaravadivel B, Xu L, McDermott PJ, Kuppuswamy D: c-Raf/MEK/ERK pathway controls protein kinase C-mediated p70S6K activation in adult cardiac muscle cells. J Biol Chem. 2002, 277: 23065-23075. 10.1074/jbc.M200328200.CrossRefPubMed Iijima Y, Laser M, Shiraishi H, Willey CD, Sundaravadivel B, Xu L, McDermott PJ, Kuppuswamy D: c-Raf/MEK/ERK pathway controls protein kinase C-mediated p70S6K activation in adult cardiac muscle cells. J Biol Chem. 2002, 277: 23065-23075. 10.1074/jbc.M200328200.CrossRefPubMed
17.
Zurück zum Zitat Network CGA: Comprehensive molecular portraits of human breast tumours. Nature. 2012, 490: 61-70. 10.1038/nature11412.CrossRef Network CGA: Comprehensive molecular portraits of human breast tumours. Nature. 2012, 490: 61-70. 10.1038/nature11412.CrossRef
18.
Zurück zum Zitat Michalides R, van Tinteren H, Balkenende A, Vermorken JB, Benraadt J, Huldij J, van Diest P: Cyclin A is a prognostic indicator in early stage breast cancer with and without tamoxifen treatment. Br J Cancer. 2002, 86: 402-408. 10.1038/sj.bjc.6600072.CrossRefPubMedPubMedCentral Michalides R, van Tinteren H, Balkenende A, Vermorken JB, Benraadt J, Huldij J, van Diest P: Cyclin A is a prognostic indicator in early stage breast cancer with and without tamoxifen treatment. Br J Cancer. 2002, 86: 402-408. 10.1038/sj.bjc.6600072.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Vermorken JB, Burgers JMV, Taat CW, van de Slee PHT, Hennipman A, Norman JWR, Rozendaal KJ, Van Tinteren H, Huldij J, Benraadt J: Adjuvant tamoxifen in breast cancer: interim results of a comprehensive cancer center Amsterdam trial [Abstract 329, poster session III]. Breast Cancer Res Treat. 1998, 50: 283- Vermorken JB, Burgers JMV, Taat CW, van de Slee PHT, Hennipman A, Norman JWR, Rozendaal KJ, Van Tinteren H, Huldij J, Benraadt J: Adjuvant tamoxifen in breast cancer: interim results of a comprehensive cancer center Amsterdam trial [Abstract 329, poster session III]. Breast Cancer Res Treat. 1998, 50: 283-
20.
Zurück zum Zitat Early Breast Cancer Trialists’ Collaborative Group: Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials. Lancet. 2005, 365: 1687-1717.CrossRef Early Breast Cancer Trialists’ Collaborative Group: Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials. Lancet. 2005, 365: 1687-1717.CrossRef
21.
Zurück zum Zitat Iwamoto T, Booser D, Valero V, Murray JL, Koenig K, Esteva FJ, Ueno NT, Zhang J, Shi W, Qi Y, Matsuoka J, Yang EJ, Hortobagyi GN, Hatzis C, Symmans WF, Pusztai L: Estrogen receptor (ER) mRNA and ER-related gene expression in breast cancers that are 1% to 10% ER-positive by immunohistochemistry. J Clin Oncol. 2012, 30: 729-734. 10.1200/JCO.2011.36.2574.CrossRefPubMed Iwamoto T, Booser D, Valero V, Murray JL, Koenig K, Esteva FJ, Ueno NT, Zhang J, Shi W, Qi Y, Matsuoka J, Yang EJ, Hortobagyi GN, Hatzis C, Symmans WF, Pusztai L: Estrogen receptor (ER) mRNA and ER-related gene expression in breast cancers that are 1% to 10% ER-positive by immunohistochemistry. J Clin Oncol. 2012, 30: 729-734. 10.1200/JCO.2011.36.2574.CrossRefPubMed
25.
Zurück zum Zitat Elston CW, Ellis IO: 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. 1991, 19: 403-410. 10.1111/j.1365-2559.1991.tb00229.x.CrossRefPubMed Elston CW, Ellis IO: 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. 1991, 19: 403-410. 10.1111/j.1365-2559.1991.tb00229.x.CrossRefPubMed
26.
Zurück zum Zitat Siddiqui S, Rimm DL: Pre-analytic variables and phospho-specific antibodies: the Achilles heel of immunohistochemistry. Breast Cancer Res. 2010, 12: 113-10.1186/bcr2782.CrossRefPubMedPubMedCentral Siddiqui S, Rimm DL: Pre-analytic variables and phospho-specific antibodies: the Achilles heel of immunohistochemistry. Breast Cancer Res. 2010, 12: 113-10.1186/bcr2782.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Hudis CA, Barlow WE, Costantino JP, Gray RJ, Pritchard KI, Chapman JA, Sparano JA, Hunsberger S, Enos RA, Gelber RD, Zujewski JA: Proposal for standardized definitions for efficacy end points in adjuvant breast cancer trials: the STEEP system. J Clin Oncol. 2007, 25: 2127-2132. 10.1200/JCO.2006.10.3523.CrossRefPubMed Hudis CA, Barlow WE, Costantino JP, Gray RJ, Pritchard KI, Chapman JA, Sparano JA, Hunsberger S, Enos RA, Gelber RD, Zujewski JA: Proposal for standardized definitions for efficacy end points in adjuvant breast cancer trials: the STEEP system. J Clin Oncol. 2007, 25: 2127-2132. 10.1200/JCO.2006.10.3523.CrossRefPubMed
28.
Zurück zum Zitat McShane LM, Altman DG, Sauerbrei W, Taube SE, Gion M, Clark GM: Reporting recommendations for tumor marker prognostic studies (REMARK). J Natl Cancer Inst. 2005, 97: 1180-1184. 10.1093/jnci/dji237.CrossRefPubMed McShane LM, Altman DG, Sauerbrei W, Taube SE, Gion M, Clark GM: Reporting recommendations for tumor marker prognostic studies (REMARK). J Natl Cancer Inst. 2005, 97: 1180-1184. 10.1093/jnci/dji237.CrossRefPubMed
29.
Zurück zum Zitat Bostner J, Karlsson E, Pandiyan MJ, Westman H, Skoog L, Fornander T, Nordenskjold B, Stal O: Activation of Akt, mTOR, and the estrogen receptor as a signature to predict tamoxifen treatment benefit. Breast Cancer Res Treat. 2013, 137: 397-406. 10.1007/s10549-012-2376-y.CrossRefPubMed Bostner J, Karlsson E, Pandiyan MJ, Westman H, Skoog L, Fornander T, Nordenskjold B, Stal O: Activation of Akt, mTOR, and the estrogen receptor as a signature to predict tamoxifen treatment benefit. Breast Cancer Res Treat. 2013, 137: 397-406. 10.1007/s10549-012-2376-y.CrossRefPubMed
30.
Zurück zum Zitat Kim EK, Kim HA, Koh JS, Kim MS, Kim KI, Lee JI, Moon NM, Ko E, Noh WC: Phosphorylated S6K1 is a possible marker for endocrine therapy resistance in hormone receptor-positive breast cancer. Breast Cancer Res Treat. 2011, 126: 93-99. 10.1007/s10549-010-1315-z.CrossRefPubMed Kim EK, Kim HA, Koh JS, Kim MS, Kim KI, Lee JI, Moon NM, Ko E, Noh WC: Phosphorylated S6K1 is a possible marker for endocrine therapy resistance in hormone receptor-positive breast cancer. Breast Cancer Res Treat. 2011, 126: 93-99. 10.1007/s10549-010-1315-z.CrossRefPubMed
31.
Zurück zum Zitat Parker RL, Huntsman DG, Lesack DW, Cupples JB, Grant DR, Akbari M, Gilks CB: Assessment of interlaboratory variation in the immunohistochemical determination of estrogen receptor status using a breast cancer tissue microarray. Am J Clin Pathol. 2002, 117: 723-728. 10.1309/PEF8-GL6F-YWMC-AG56.CrossRefPubMed Parker RL, Huntsman DG, Lesack DW, Cupples JB, Grant DR, Akbari M, Gilks CB: Assessment of interlaboratory variation in the immunohistochemical determination of estrogen receptor status using a breast cancer tissue microarray. Am J Clin Pathol. 2002, 117: 723-728. 10.1309/PEF8-GL6F-YWMC-AG56.CrossRefPubMed
32.
Zurück zum Zitat Rexhepaj E, Brennan DJ, Holloway P, Kay EW, McCann AH, Landberg G, Duffy MJ, Jirstrom K, Gallagher WM: Novel image analysis approach for quantifying expression of nuclear proteins assessed by immunohistochemistry: application to measurement of oestrogen and progesterone receptor levels in breast cancer. Breast Cancer Res. 2008, 10: R89-10.1186/bcr2187.CrossRefPubMedPubMedCentral Rexhepaj E, Brennan DJ, Holloway P, Kay EW, McCann AH, Landberg G, Duffy MJ, Jirstrom K, Gallagher WM: Novel image analysis approach for quantifying expression of nuclear proteins assessed by immunohistochemistry: application to measurement of oestrogen and progesterone receptor levels in breast cancer. Breast Cancer Res. 2008, 10: R89-10.1186/bcr2187.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Al-Janabi S, Huisman A, van Diest PJ: Digital pathology: current status and future perspectives. Histopathology. 2011, 61: 1-9.CrossRefPubMed Al-Janabi S, Huisman A, van Diest PJ: Digital pathology: current status and future perspectives. Histopathology. 2011, 61: 1-9.CrossRefPubMed
34.
Zurück zum Zitat Bostner J, Skoog L, Fornander T, Nordenskjold B, Stal O: Estrogen receptor-alpha phosphorylation at serine 305, nuclear p21-activated kinase 1 expression, and response to tamoxifen in postmenopausal breast cancer. Clin Cancer Res. 2010, 16: 1624-1633. 10.1158/1078-0432.CCR-09-1733.CrossRefPubMed Bostner J, Skoog L, Fornander T, Nordenskjold B, Stal O: Estrogen receptor-alpha phosphorylation at serine 305, nuclear p21-activated kinase 1 expression, and response to tamoxifen in postmenopausal breast cancer. Clin Cancer Res. 2010, 16: 1624-1633. 10.1158/1078-0432.CCR-09-1733.CrossRefPubMed
35.
Zurück zum Zitat Gutierrez MC, Detre S, Johnston S, Mohsin SK, Shou J, Allred DC, Schiff R, Osborne CK, Dowsett M: Molecular changes in tamoxifen-resistant breast cancer: relationship between estrogen receptor, HER-2, and p38 mitogen-activated protein kinase. J Clin Oncol. 2005, 23: 2469-2476. 10.1200/JCO.2005.01.172.CrossRefPubMed Gutierrez MC, Detre S, Johnston S, Mohsin SK, Shou J, Allred DC, Schiff R, Osborne CK, Dowsett M: Molecular changes in tamoxifen-resistant breast cancer: relationship between estrogen receptor, HER-2, and p38 mitogen-activated protein kinase. J Clin Oncol. 2005, 23: 2469-2476. 10.1200/JCO.2005.01.172.CrossRefPubMed
36.
Zurück zum Zitat Beelen K, Opdam M, Severson T, Koornstra RHT, Vincent A, Wesseling J, Murris J, Berns EMJJ, Vermorken JB, van Diest PJ, Linn SC: PIK3CA mutations, phosphatase and tensin homolog, human epidermal growth 3 factor receptor 2 and insulin-like growth factor 1 receptor and adjuvant tamoxifen resistance in postmenopausal breast cancer patients. Breast Cancer Res. 2013, in press Beelen K, Opdam M, Severson T, Koornstra RHT, Vincent A, Wesseling J, Murris J, Berns EMJJ, Vermorken JB, van Diest PJ, Linn SC: PIK3CA mutations, phosphatase and tensin homolog, human epidermal growth 3 factor receptor 2 and insulin-like growth factor 1 receptor and adjuvant tamoxifen resistance in postmenopausal breast cancer patients. Breast Cancer Res. 2013, in press
37.
Zurück zum Zitat Generali D, Fox SB, Brizzi MP, Allevi G, Bonardi S, Aguggini S, Milani M, Bersiga A, Campo L, Dionisio R, Vergoni F, Giardini R, Dogliotti L, Bottini A, Harris AL, Berruti A: Down-regulation of phosphatidylinositol 3′-kinase/AKT/molecular target of rapamycin metabolic pathway by primary letrozole-based therapy in human breast cancer. Clin Cancer Res. 2008, 14: 2673-2680. 10.1158/1078-0432.CCR-07-1046.CrossRefPubMed Generali D, Fox SB, Brizzi MP, Allevi G, Bonardi S, Aguggini S, Milani M, Bersiga A, Campo L, Dionisio R, Vergoni F, Giardini R, Dogliotti L, Bottini A, Harris AL, Berruti A: Down-regulation of phosphatidylinositol 3′-kinase/AKT/molecular target of rapamycin metabolic pathway by primary letrozole-based therapy in human breast cancer. Clin Cancer Res. 2008, 14: 2673-2680. 10.1158/1078-0432.CCR-07-1046.CrossRefPubMed
38.
Zurück zum Zitat Bachelot T, Ferrero J, Cropet C, Bourgier C, Abadie-Lacourtoisie , Levy C: Translational studies within the TAMRAD randomized GINECO trial: evidence for TORC1 activation marker as a predictive factor for everolimus efficacy in metastatic breast cancer (mBC). Ann Oncol. 2012, 23 (Suppl 2): ii17-ii24. Bachelot T, Ferrero J, Cropet C, Bourgier C, Abadie-Lacourtoisie , Levy C: Translational studies within the TAMRAD randomized GINECO trial: evidence for TORC1 activation marker as a predictive factor for everolimus efficacy in metastatic breast cancer (mBC). Ann Oncol. 2012, 23 (Suppl 2): ii17-ii24.
39.
Zurück zum Zitat Chavez-Mac GM, Barlow WE, Gonzalez-Angulo AM, Rastogi P, Mamounas EP, Ganz PA, Schott AF, Paik S, Lew DL, Bandos H, Hortobagyi GN: A phase III randomized, placebo-controlled clinical trial evaluating the use of adjuvant endocrine therapy +/− one year of everolimus in patients with high-risk, hormone receptor- (HR) positive and HER2-negative breast cancer: SWOG/NSABP S1207 [Abstract OT2-2-04]. Cancer Res. 2012, 72 (24): Supplement 3-doi: 10.1158/0008-5472 Chavez-Mac GM, Barlow WE, Gonzalez-Angulo AM, Rastogi P, Mamounas EP, Ganz PA, Schott AF, Paik S, Lew DL, Bandos H, Hortobagyi GN: A phase III randomized, placebo-controlled clinical trial evaluating the use of adjuvant endocrine therapy +/− one year of everolimus in patients with high-risk, hormone receptor- (HR) positive and HER2-negative breast cancer: SWOG/NSABP S1207 [Abstract OT2-2-04]. Cancer Res. 2012, 72 (24): Supplement 3-doi: 10.1158/0008-5472
Metadaten
Titel
Phosphorylated p-70S6K predicts tamoxifen resistance in postmenopausal breast cancer patients randomized between adjuvant tamoxifen versus no systemic treatment
verfasst von
Karin Beelen
Mark Opdam
Tesa M Severson
Rutger HT Koornstra
Andrew D Vincent
Jelle Wesseling
Jettie J Muris
Els MJJ Berns
Jan B Vermorken
Paul J van Diest
Sabine C Linn
Publikationsdatum
01.02.2014
Verlag
BioMed Central
Erschienen in
Breast Cancer Research / Ausgabe 1/2014
Elektronische ISSN: 1465-542X
DOI
https://doi.org/10.1186/bcr3598

Weitere Artikel der Ausgabe 1/2014

Breast Cancer Research 1/2014 Zur Ausgabe

Update Onkologie

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