Skip to main content
Erschienen in: BMC Cancer 1/2010

Open Access 01.12.2010 | Research article

Molecular risk assessment of BIG 1-98 participants by expression profiling using RNA from archival tissue

verfasst von: Janine Antonov, Vlad Popovici, Mauro Delorenzi, Pratyaksha Wirapati, Anna Baltzer, Andrea Oberli, Beat Thürlimann, Anita Giobbie-Hurder, Giuseppe Viale, Hans Jörg Altermatt, Stefan Aebi, Rolf Jaggi

Erschienen in: BMC Cancer | Ausgabe 1/2010

Abstract

Background

The purpose of the work reported here is to test reliable molecular profiles using routinely processed formalin-fixed paraffin-embedded (FFPE) tissues from participants of the clinical trial BIG 1-98 with a median follow-up of 60 months.

Methods

RNA from fresh frozen (FF) and FFPE tumor samples of 82 patients were used for quality control, and independent FFPE tissues of 342 postmenopausal participants of BIG 1-98 with ER-positive cancer were analyzed by measuring prospectively selected genes and computing scores representing the functions of the estrogen receptor (eight genes, ER_8), the progesterone receptor (five genes, PGR_5), Her2 (two genes, HER2_2), and proliferation (ten genes, PRO_10) by quantitative reverse transcription PCR (qRT-PCR) on TaqMan Low Density Arrays. Molecular scores were computed for each category and ER_8, PGR_5, HER2_2, and PRO_10 scores were combined into a RISK_25 score.

Results

Pearson correlation coefficients between FF- and FFPE-derived scores were at least 0.94 and high concordance was observed between molecular scores and immunohistochemical data. The HER2_2, PGR_5, PRO_10 and RISK_25 scores were significant predictors of disease free-survival (DFS) in univariate Cox proportional hazard regression. PRO_10 and RISK_25 scores predicted DFS in patients with histological grade II breast cancer and in lymph node positive disease. The PRO_10 and PGR_5 scores were independent predictors of DFS in multivariate Cox regression models incorporating clinical risk indicators; PRO_10 outperformed Ki-67 labeling index in multivariate Cox proportional hazard analyses.

Conclusions

Scores representing the endocrine responsiveness and proliferation status of breast cancers were developed from gene expression analyses based on RNA derived from FFPE tissues. The validation of the molecular scores with tumor samples of participants of the BIG 1-98 trial demonstrates that such scores can serve as independent prognostic factors to estimate disease free survival (DFS) in postmenopausal patients with estrogen receptor positive breast cancer.

Trial Registration

Current Controlled Trials: NCT00004205
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1471-2407-10-37) contains supplementary material, which is available to authorized users.
Janine Antonov, Vlad Popovici contributed equally to this work.

Competing interests

JA, VP, MD, PW, AB, AO, AGH, GV, HJA, SA and RJ declare that they have no competing interest. B.T. holds stocks from Novartis (Ciba Geigy) since 1990.

Authors' contributions

JA, SA and RJ organized the study, planned the experiments and wrote the manuscript. SA and BT organized samples from the International Breast Cancer Study Group. AO and AB carried out RNA isolations, quality controls and gene expression measurements. VP, PW, MD and AGH carried out the statistical analyses. HJA and GV were responsible for histological assessment of stained sections. All authors contributed to the manuscript, they read and approved the final manuscript.
Abkürzungen
AUC
area under the (ROC) curve
CI
confidence interval
DFS
disease-free survival
ER
estrogen receptor
FF
fresh frozen
FFPE
formalin-fixed, paraffin embedded
HR
hazard ratio
IHC
immunohistochemistry
GGI
gene expression grade index
LI
labeling index
LRT
likelihood ratio tests
PCR
polymerase chain reaction
RIN
RNA integrity number
PgR
progesterone receptor
ROC
receiver operating characteristic
RS
recurrence score
TLDA
TaqMan Low Density Arrays.

Background

Clinical and histopathological factors such as lymph node status, tumor size, histological grade, age, and expression of estrogen receptor (ER) and Her2 have traditionally guided treatment decisions of patients with operable breast cancer [1, 2]. Various prognostic models are based on these factors, for example the Nottingham Prognostic Index (NPI) [3, 4], Adjuvant!Online [5, 6] and others [7]. Despite providing excellent estimates of the average risk of recurrence, there remains substantial variation in outcome which may be explained by molecular differences among these tumors [8, 9].
DNA-chip based expression analyses have confirmed the heterogeneity of breast cancer and allowed the development of clinically relevant gene "signatures" or "profiles" [1020]. Such profiles are being implemented widely in routine patient care even though many signatures were developed and validated on heterogeneous patient cohorts with respect to stage of disease and therapy. The utility of gene signatures as part of the decision making process is being validated in ongoing studies (TAILORx [21] and MINDACT [22]). Most profiling studies are based on fresh-frozen (FF) or RNAlater conserved tissue. Such material must be collected and processed separately after surgery, complicating the implementation of molecular analyses into the clinical workflow. Procedures based on formalin-fixed, paraffin-embedded (FFPE) material simplify the acquisition of tumor material and can easily be established as part of the routine pathological procedures. In addition, FFPE tissues collected in the framework of clinical trials could be a valuable resource for future research.
We prospectively selected genes from publicly available microarray data and developed molecular scores representing the ER, progesterone receptor (PgR), Her2 and proliferation (PRO) status, and the overall risk of recurrence (RISK). The reproducibility and robustness of the molecular scores was validated by comparing expression data with RNA from FF and FFPE material of 82 tumors. Molecular scores were determined from 342 ER positive tumor samples of the BIG 1-98 clinical trial. Multivariate Cox proportional hazard models revealed that molecular scores are independent prognostic factors to estimate disease free survival (DFS).

Methods

To assess the quality of expression profiling from FFPE material, matched FF and FFPE samples from 82 human breast cancers were used. Histopathological information was irreversibly anonymized according to Swiss law. Independent FFPE blocks and corresponding clinical data of 437 Swiss participants of the trial BIG 1-98 were provided by the International Breast Cancer Study Group. The ethics committees and required health authorities of each participating institution approved the study protocol, and all patients gave written informed consent (ClinicalTrials.gov number, NCT00004205) [23]. Retrospective tissue collection was carried out in accordance with institutional guidelines and national laws. The patient and tumor characteristics of these patients were similar to the entire BIG 1-98 population (Table 1). BIG 1-98 is a randomized controlled clinical trial of adjuvant hormonal therapy for postmenopausal patients with endocrine-responsive breast cancer comparing 4 arms: 5 years of tamoxifen, 5 years of letrozole, two years of tamoxifen followed by 3 years of letrozole, or vice versa [2426]. All the patients from the BIG 1-98 were treated by mastectomy or breast conserving surgery [2426]. The available paraffin blocks contained material derived from representative tumor regions.
Table 1
Gene Identifications, Categories and Score affiliations
Gene
Category
Accession Nr.
Description
AS
Score
GUSB
Control
NM_000181.1
glucuronidase, beta
81
control
RPLP0
Control
NM_053275.3
NM_001002.3
ribosomal protein, large, P0
105
control
UBB
Control
NM_018955.2
ubiquitin B
120
control
AR
ER
NM_001011645.1
NM_000044.2
androgen receptor (dihydrotestosterone receptor; testicular feminization; spinal and bulbar muscular atrophy; Kennedy disease)
72
ER_8
ERBB4
ER
NM_001042599.1
NM_005235.2
v-erb-a erythroblastic leukemia viral oncogene homolog 4 (avian)
77
ER_8
ESR1
ER
NM_000125.2
estrogen receptor 1
62
ER_8
ER_4
FOXA1
ER
NM_004496.2
forkhead box A1
74
ER_8
GATA3
ER
NM_001002295.1
NM_002051.2
GATA binding protein 3
80
ER_8
MAPT
ER
NM_016834.2
NM_016835.2
NM_016841.2
NM_005910.3
microtubule-associated protein tau
60
ER_8
MYB
ER
NM_005375.2
v-myb myeloblastosis viral oncogene homolog (avian)
96
ER_8
XBP1
ER
NM_005080.2
X-box binding protein 1
60
ER_8
BCL2
ER
NM_000633.2
B-cell CLL/lymphoma 2
81
ER_4
GREB1
PGR
NM_033090.1
NM_148903.1
NM_014668.2
GREB1 protein
77
PGR_5
PGR
PGR
NM_000926.3
progesterone receptor
118
PGR_5 ER_4
RAB31
PGR
NM_006868.2
RAB31, member RAS oncogene family
109
PGR_5
RBBP8
PGR
NM_203291.1
NM_203292.1
NM_002894.2
retinoblastoma binding protein 8
75
PGR_5
SERPINA3
PGR
NM_001085.4
serpin peptidase inhibitor, clade A (alpha-1 antiproteinase, antitrypsin), member 3
70
PGR_5
SCUBE2
PGR
NM_020974.1
CEGP1, signal peptide, CUB domain, EGF-like 2
64
ER_4
ERBB2
HER2
NM_001005862.1
NM_004448.2
v-erb-b2 erythroblastic leukemia viral oncogene homolog 2, neuro/glioblastoma derived oncogene homolog (avian)
120
HER2_2
GRB7
HER2
NM_005310.2
growth factor receptor-bound protein 7
70
HER2_2
CCNB2
Proliferation
NM_004701.2
cyclin B2
73
PRO_10
CCNE2
Proliferation
NM_057735.1
NM_057749.1
cyclin E2
70
PRO_10
CDC2
Proliferation
NM_033379.2 NM_001786.2
cell division cycle 2, G1 to S and G2 to M
92
PRO_10
CENPF
Proliferation
NM_016343.3
centromere protein F, 350/400 ka (mitosin)
99
PRO_10
KIF20A
Proliferation
NM_005733.1
kinesin family member 20A
130
PRO_10
MKI67
Proliferation
NM_002417.3
antigen identified by monoclonal antibody Ki-67
131
PRO_10 PRO_5
ORC6L
Proliferation
NM_014321.2
origin recognition complex, subunit 6 like (yeast)
78
PRO_10
PRC1
Proliferation
NM_199413.1
NM_199414.1
NM_003981.2
protein regulator of cytokinesis 1
66
PRO_10
SPAG5
Proliferation
NM_006461.3
sperm associated antigen 5
114
PRO_10
TOP2A
Proliferation
NM_001067.2
topoisomerase (DNA) II alpha 170 kDa
125
PRO_10
AURKA
Proliferation
NM_003600.2
STK15 aurora kinase A
85
PRO_5
BIRC5
Proliferation
NM_001012271.1
NM_001168.2
baculoviral IAP repeat-containing 5 (survivin)
93
PRO_5
CCNB1
Proliferation
NM_031966.2
cyclin B1
104
PRO_5
MYBL2
Proliferation
NM_002466.2
v-myb myeloblastosis viral oncogene homolog (avian)-like 2
81
PRO_5
Abbreviation: AS, amplicon size

Tissue samples and data processing

The RNA was isolated from 4 sections (25 μm) of FF material and from 10 paraffin sections (10 μm thick) as described previously [27]. After demodification, the RNA was bound to silica-based columns, DNase I digested and eluted with water. The protocols and reagents for RNA isolation from FF and FFPE tissues were recently incorporated in commercial protocols (RNAready and FFPE RNAready, AmpTec, Hamburg, Germany). RNA qualities were assessed on an Agilent 2100 Bioanalyzer (Agilent Technologies, Inc., Santa Clara, CA, USA). RNA prepared from FF material had a RIN>6 (RNA integrity number), the RIN of RNA from FFPE was 2-3. The percentage of tumor cells in each FFPE block was evaluated on stained tissue sections. From 437 available FFPE samples 43 samples (9.8%) with less than ~30% tumor cells, 10 ER-negative tumor samples and 7 samples (1.6%) with less than 1.5 μg total RNA recovery were excluded from further analysis. Approximately 30% of the sections contained 30-50% tumor cells, and about 60% contained 50-100% tumor cells. Each of the remaining RNAs was tested by quantitative reverse transcription PCR (qRT-PCR) with 3 control genes (GUSB, RPLP0 and UBB). The mean of the three raw Cts (cycle thresholds) was determined. In 35 samples (8%) the mean Ct was >31, indicating poor quality of the RNA. These RNAs were excluded from further analyses. For the remaining 342 RNAs (78.3%), the expression of 34 genes (see Table 1) was measured by qRT-PCR on TaqMan Low Density Arrays (TLDAs) (Applied Biosystems, Foster City, CA, USA) using a one step protocol (Invitrogen, Basel, Switzerland) on an Applied Biosystems 7900HT instrument. Technical replicates were performed for several intact and several partially degraded RNAs from FF and FFPE material, respectively. They revealed Pearson correlation coefficients higher than 0.95 for all 34 assays.
Genes with high correlation to the expression of ER, PgR, Her2 and proliferation related genes were prospectively selected from publicly available microarray data [28]. A complete list of microarray data sets used in the meta-analysis is available at ".http://​breast-cancer-research.​com/​content/​10/​4/​R65/​table/​T1[28] (Additional File 1, Table S1). The scores were defined by giving equal weight to each gene in the four groups (proliferation, estrogen response, progesterone response, Her2 response). Thus, a training set was not used as the scores were based on in silico gene selection.
Raw Ct values were normalized against the mean expression of GUSB, RPLP0 and UBB. Scores for ER (ER_8), PgR (PGR_5), Her2 (HER2_2) and proliferation (PRO_10) were defined as mean expression of all genes in each category (Table 1). A RISK score comprising 25 genes was calculated as follows: RISK_25 = PRO_10+HER2_2-(8 × ER_8+5 × PGR_5)/13. For comparison, ER_4 and PRO_5 scores were calculated based on 4 and 5 genes described previously [27]. The genes corresponding to ER_4 and PRO_5 scores corresponded to the genes used for calculating the recurrence score (RS) [29].

Concordance of molecular scores and pathological parameters

Histopathological data of BIG 1-98 samples were derived from a central review, with the exception of the grade which was locally assessed. The ER and PgR status were dichotomized into positive (≥ 10% immunoreactive cells) or negative (<10%) [30]. Her2 was measured by fluorescence in-situ hybridization or immunohistochemistry (IHC) and tumors were classified according to Rasmussen et al. [31]. The Ki-67 labeling index (LI) was centrally assessed by IHC as described and classified into low or high using the median LI (11%) as cut-off [32]. The same assays and cut-offs were used for the 82 matched samples with the exception of Her2 which was measured using the CB11 monoclonal antibody and using a cut-off of ≥ 50% [33]. Continuous molecular scores were compared to binary IHC parameters using the area under the curve (AUC). The 95% confidence intervals (CI) were estimated by a bootstrap method (100 bootstraps). Two-sided Mann-Whitney tests were used to assess the association between clinicopathological factors and scores.

Statistical analyses

Primary endpoint of survival analyses was DFS as defined previously [25]. Forty-five events were observed in 342 patients with a median follow-up time (estimated by reverse Kaplan-Meier [34]) of 60 months. DFS was estimated by Kaplan Meier analysis. Patients were classified into low and high PRO or RISK scores using the corresponding median score as cut-off. The differences in survival experience between the two resulting groups were assessed with log rank tests. Univariate and multivariate Cox proportional hazard models were used [35] and hazard ratios (HR), CIs and p-values were obtained. The multivariate models were assessed using the log-likelihood and the deviance of residuals. Likelihood ratio tests (LRT) were used to compare different nested multivariate models. No adjustments were made for multiple testing. Univariate Cox proportional hazard models were applied to estimate the rate of events and to produce corresponding plots.

Results

Reliable expression profiling from FFPE tumor tissue

Gene expression was measured from 34 genes using TLDAs with RNA isolated from FF and FFPE material of 82 breast cancers. These data were used solely for the assessment of the expression profiling from FFPE material. Pearson correlation coefficients between FF and FFPE expression values for each tumor and all assays ranged from 0.91 to 0.98. The mean increase of raw Ct values derived of FFPE compared to matched FF tissues was 1.30 units. This Ct shift was mostly compensated by normalization (Additional File 2, Figure S1. and Additional File 3, Figure S2).
Unsupervised hierarchical clustering demonstrated the stability of gene clusters and revealed an excellent agreement between FF- and FFPE-based expression profiles (Additional File 4, Figure S3). Molecular scores were determined for ER, PGR, HER2 and PRO. A linear relationship of scores was found for RNA from FF and RNA from FFPE material (Figure 1). Pearson correlation coefficients for the four scores were 0.968, 0.974, 0.942 and 0.944, respectively. The distributions of ER_8, PGR_5 and HER2_2 scores are shown as histograms together with the fitted mixture of two Gaussian distributions (Additional File 1, Figure S4) used for discriminating the subtypes.
The agreement between molecular scores and corresponding binary IHC variables was assessed by receiver operating characteristic (ROC) curves and AUC. AUCs and 95% CI were calculated for ER_8 (FF = 0.940 (0.835-1.00), FFPE = 0.931 (0.804-1.00)), PGR_5 (FF = 0.919 (0.828-0.986), FFPE = 0.916 (0.806-0.987) and HER2_2 (FF = 0.961 (0.895-1.00), FFPE = 0.963 (0.915-0.993)). PRO_10 was compared with IHC data for Ki-67 using a cut-off of 11% and the resulting AUCs were 0.798 (0.609-0.900) for FF and 0.810 (0.660-0.907) for FFPE, respectively. In conclusion, the agreement of the IHC with FFPE samples was as good as with FF samples.

Concordance between pathological parameters and molecular scores for tumors of the BIG 1-98 clinical trial

Molecular scoring was applied to an independent set of tissue samples from Swiss patients participating in the BIG 1-98 randomized clinical trial and scores were compared to centrally assessed histopathological data by ROC curves. From a total of 437 provided tumor samples 342 ER-positive tumors (78.3%) were suitable for analysis. The AUC was 0.974 (95% CI = 0.946-0.995) for HER2_2 and 0.847 (95% CI = 0.794-0.902) for PGR_5. PRO_10 scores positively correlated with Ki-67 LI (Pearson correlation coefficient 0.51); the AUC was 0.815 (95% CI = 0.768-0.864) for Ki-67 binarized at 11% [32].

The PRO_10 score correlates with histological grade and other clinical factors

The histological grade was assessed according to Elston and Ellis [36]. The PRO_10 score positively correlated with Elston and Ellis scores and with grade (Pearson correlation coefficient 0.453 and 0.409, respectively) (Figure 2). Furthermore, PRO_10 scores were significantly higher in Her2 positive tumors, in tumors larger than 2 cm and in tumors with axillary lymph node metastasis as compared to Her2 negative tumors, T1 tumors and N0 tumors (p ≤ 0.0015, Mann-Whitney tests), respectively (data not shown).

PRO and RISK scores predict disease free survival in lymph node positive patients and patients with grade II breast cancer

The prognostic values of PRO_10 and RISK_25 scores were assessed by their ability to assign patients to low and high risk groups. Patients were stratified according to histological grade and low or high PRO_10 and RISK_25 scores using the corresponding medians as cut-offs (Figure 3). As expected, patients with grade III tumors had poorer DFS than patients with grade I or grade II tumors (p = 0.0019, panel A). High PRO_10 scores correlated with poorer DFS compared to low scores in all (p = 0.0043, panel B) and in histological grade II tumors (p = 0.0024, panel C). Similarly, RISK_25 discriminated between favorable and poor DFS in all (p = 0.0005, panel D) and in node positive tumors (p = 0.0009, panel E). Univariate Cox proportional hazards regression analysis confirmed these observations.
The PGR_5, PRO_10 and RISK_25 scores were all significant predictors of DFS (p < 0.05) as were histological grade, tumor size, number of positive lymph nodes and Ki-67 LI (Table 2). The PRO_5 score was also a significant predictor of DFS but PRO_10 score was numerically better than PRO_5 in terms of log-likelihood (L) and deviance of residuals (D) (PRO_10: L = -223.35, D = 225.83; PRO_5: L = -224.16, D = 227.57).
Table 2
Baseline characteristics.
Characteristic
Patients with FFPE profiles from Swiss participants used in the study (N = 342)
Provided material of Swiss participants
(N = 437)
Patients of the BIG 1-98 population not used in the study
(N = 7573)
Overall BIG 1-98 population
(N = 8010)
Menopausal category - N (%)
    
Postmen. before chemo
321 (93.9)
413 (94.5)
7279 (96.1)
7692 (96.0)
Postmen. after chemo
10 (2.9)
11 (2.5)
181 (2.4)
192 (2.4)
Premenopausal (ineligible)
0 (0.0)
2 (0.5)
21 (0.3)
23 (0.3)
Uncertain status
10 (2.9)
10 (2.3)
92 (1.2)
102 (1.3)
Unknown/missing
1 (0.3)
1 (0.2)
0
1 (<0.1)
Age at randomization - years
    
Median
62
62
61
61
Range
41-86
41-86
38-90
38-90
Tumor size - N (%)
    
≤ 2 cm
195 (57.0)
251 (57.4)
4706 (62.1)
4957 (61.9)
> 2 cm
144 (42.1)
179 (41.0)
2794 (36.9)
2973 (37.1)
Unknown/missing
3 (0.9)
7 (1.6)
73 (1.0)
80 (1.0)
Tumor grade - N (%)
    
Grade 1
94 (27.5)
124 (28.4)
2007 (26.5)
2131 (26.6)
Grade 2
196 (57.3)
251 (57.4)
3649 (48.2)
3900 (38.7)
Grade 3
49 (14.3)
59 (13.5)
1166 (15.4)
1225 (15.3)
Unknown/missing
3 (0.9)
3 (0.7)
751 (9.9)
754 (9.4)
Nodal status - N (%)
    
Negative (including Nx)
186 (54.4)
245 (56.1)
4342 (57.3)
4587 (57.3)
Positive
152 (44.4)
188 (43.0)
3123 (41.2)
3311 (41.3)
Unknown/missing
4 (1.2)
4 (1.0)
108 (1.4)
112 (1.4)
ER and PgR status - N (%)
    
ER pos and PgR pos.
268 (78.4)
340 (77.8)
4715 (62.3)
5055 (63.1)
ER pos and PgR neg.
66 (19.3)
87 (19.9)
1544 (20.4)
1631 (20.4)
ER pos and PgR unknown
1 (0.3)
1 (0.2)
1153 (15.2)
1154 (14.4)
ER neg and PgR pos.
5 (1.5)
7 (1.6)
136 (1.8)
143 (1.8)
ER unknown, PGR pos.
0
0
7 (0.1)
7 (0.1)
Other
2 (0.6)
2 (0.5)
18 (0.3)
20 (0.2)
Local therapy - N (%)
    
BCS and RT
236 (69.0)
310 (70.9)
3987 (52.7)
4297 (53.7)
BCS and no RT
13 (3.8)
16 (3.7)
228 (3.0)
244 (3.0)
Mastectomy and RT
24 (7.0)
25 (5.7)
1415 (18.7)
1440 (18.0)
Mastectomy and no RT.
68 (19.9)
85 (19.5)
1926 (25.4)
2011 (25.1)
Other
1 (0.3)
1 (0.2)
17 (0.2)
18 (0.2)
Adjuvant or neoadjuvant
    
chemo (or both) - N (%)
    
Yes
133 (38.9)
159 (36.4)
1865 (24.6)
2024 (25.3)
No
209 (61.1)
278 (63.6)
5708 (75.4)
5986 (74.7)
Abbreviations: BCS, breast conserving surgery; Nx, nodal status unknown; postmen., postmenopausal; RT, radiotherapy; PgR, progesterone receptor; pos., positive; neg., negative
Figure 4 shows the estimated rate of recurrence as a function of PRO_10, PGR_5 and RISK_25 scores. The PRO_5, PRO_10 and the RISK_25 scores remained significant predictors of DFS when applied to patients with grade II breast cancer.

PRO_10 and PGR_5 scores are independent risk factors in multivariate analyses

The impact of the molecular scores PRO_10 and PGR_5 was further documented in multivariate models comprising clinicopathologic predictors and molecular scores that were significant in univariate analyses.
Multivariate analyses revealed that PRO_10 is a predictor of DFS independent of tumor size (T), number of positive lymph nodes (N), grade (G) and Ki-67 LI. PRO_10 represents proliferation-related genes and it was of interest to compare it to Ki-67. Table 2 shows the results of multivariate analyses including T, N, G and either Ki-67 (model 1) or PRO_10 (model 3) in comparison with a model containing both markers (model 2). The full model (model 2) was significantly better than model 1 (LRT p = 0.0071). No significant difference was found for PRO_10 between models 2 and 3 (LRT p = 0.8075). Thus, adding PRO_10 to T, N, G and Ki-67 significantly improved the model. In contrast, adding Ki-67 to T, N, G and PRO_10 did not bring additional information.
The same procedure was used to evaluate whether PGR_5 further improved model 6 containing T, N, G and PRO_10 (Table 2). The full model including all 5 variables (model 5) performed better than model 4 (T, N, G, PGR_5; LRT p = 0.0089) and model 6 (T, N, G, PRO_10; LRT p = 0.0339). Both, PGR_5 and PRO_10 remained significant in model 5 suggesting that the two scores contain independent information with respect to prognosis and outcome.

Discussion

Gene expression profilings define clinically relevant gene signatures [15, 17, 37, 38]. For the present work, we selected genes correlating with the ER, PgR, Her2 and proliferative status using a meta-analysis of gene expression profiles [28]. The prognostic power of resulting gene expression scores for ER, PgR, proliferation and overall risk of recurrence was validated using tissues and clinical data from a representative subset of participants of trial BIG 1-98 confirming the correlation structure of these genes and their association with clinical and outcome variables.
Multiple genes representing each score were quantified by qRT-PCR. RNA from 82 matched FF and FFPE tissues were compared by qRT-PCR on TLDAs. The mean increase of raw Ct values between RNA from FF and FFPE tissues was 1.3 units. This is similar to the findings of Cronin and co-workers (+2.0 units) in a comparable setting [39]. Duration of formalin fixation, storage time and conditions influence the quality of RNA derived of FFPE tissues with direct effects on the sensitivity of subsequent PCR reactions [40]. However, normalization effectively compensated for this shift of Ct values (Additional File 2, Fig S1 and Additional File 3, Figure S2).
The mean expression of eight genes related to ER and five genes related to PgR were used to calculate the ER_8 and PGR_5 scores. Scores representing different functional categories were combined in RISK_25 score. The molecular scores determined from 82 paired samples of FF and FFPE tumors were highly concordant, as were molecular scores and immunohistochemically assessed parameters demonstrating the reliability of the procedure.
Molecular scores were validated in an independent set of tumor tissues from 342 participants of trial BIG 1-98. In contrast to histological analyses which can also be performed from tissue sections that contain considerable normal, stromal or fat components the architecture of the tissue is completely lost during work up for molecular analyses and therefore, it was important to exclude samples with inadequate tumor content. A histological section was taken from the immediate vicinity of each sample that was used for molecular analyses. Each section was assessed by an experienced pathologist (H.J.A.) and molecular analyses were restricted to samples containing at least 30% tumor cells. For comparison, RNA was also isolated from tumor-surrounding cells which led to rather poor RNA recoveries from comparable tissue areas (data not shown). However, this does not exclude that tumor-surrounding cells may have a limited impact on molecular scores in such analyses. Contamination by non-tumor cells may be reduced by macrodissecting tumors before RNA isolation and molecular assessment. The same procedure would also make tumors accessible to molecular analysis when sections contain less than 30% tumor cells.
Classification of patients by low and high PRO_10 and RISK_25 scores corresponded to low and high risk of recurrence. PRO, RISK and PGR scores were prognostic for DFS not only in the entire patient population but also in a subpopulation of patients with node positive disease (Figure 3D and 3E). We provide evidence independent of Genomic Health™ that a RISK score based on similar biological processes as the recurrence score (RS), but with other genes selected through a different procedure, can predict DFS [29, 41, 42]. In contrast to the RS which was validated with tamoxifen-treated patients, PRO_10, RISK_25 and PGR_5 scores were validated with patients treated with tamoxifen, letrozole or a sequence of both drugs; therefore, they may apply to patients who received either of these drugs.
Histological grading is an important factor in estimating the risk of recurrence of patients with breast cancer [2, 43]. Recently, Sortiriou and colleagues have developed the gene expression grade index (GGI) based on the expression of 97 genes related to proliferation. They demonstrated that grade II cancers are comprised of tumors which are similar to genomic grade I or grade III with corresponding clinical outcomes [16, 44]. Our findings agree with these observations as grade II tumors could be further classified into low and high risk of recurrence by 10 genes (PRO_10) (Figure 3C) or even by 5 genes (PRO_5 score) (data not shown). Seven of the PRO_10 and three of the PRO_5 genes are also part of GGI. The PRO_5 genes (Table 1) corresponded to the proliferation-related genes of the RS [29]. The assessment of gene signatures related to proliferation such as GGI or PRO scores is of special interest in ER positive, grade II breast cancer for whom therapeutic decisions are often difficult. Both, GGI and RS were shown to be associated with response to chemotherapy [45, 46]. In contrast to GGI which requires FF tumor material, PRO scores or RS can be determined from a few microtome slices or cores such as used for tissue microarrays [47]. Material for molecular analysis can be taken from the same FFPE tissue block used for histological and immunohistochemical analyses without interfering with clinicopathological workflow.
The prognostic value of Ki-67 in early breast cancer was recently confirmed [48]. However, Ki-67 is not used uniformly in clinical practice [49, 50] as it appears to be difficult to agree on cut-off values separating high and low proliferation tumors or on its value in assisting the choice of adjuvant therapy [50, 51]. Therefore, instead of dichotomizing Ki-67 it may be more feasible to use Ki-67 as continuous variable [52]. Here, we made a comparison between centrally assessed Ki-67 LI and a qRT-PCR based proliferation signature. The PRO_10 score correlated with Ki-67 LI, and both were significant predictors of DFS in univariate Cox analyses. In multivariate models however, PRO_10 offered superior prognostic value and outperformed Ki-67 LI (Table 3). Moreover, the PRO_10 score added independent prognostic information to anatomical staging.
Table 3
Cox Proportional Hazard Analyses.
Covariate
P- value
HR (95% CI)
Univariate Analyses*
  
Clinicopathological Variables
  
   HER2
0.7816
1.18 (0.36 - 3.84)
   PgR
0.5147
0.78 (0.36 - 1.66)
   Histological grade
0.0032
1.99 (1.26 - 3.14)
   Ki-67 LI
0.0226
1.02 (1.00 - 1.04)
   Tumor size
0.0047
1.22 (1.06 - 1.39)
   Number of positive nodes
<0.0001
1.13 (1.08 - 1.18)
   Treatment (4 categories)
0.1540
-
Molecular scores
  
   HER2_2
0.1080
1.20 (0.96 - 1.51)
   PGR_5
0.0344
0.66 (0.44 - 0.97)
   PRO_5
0.0003
2.14 (1.42 - 3.22)
   PRO_10
<0.0001
2.09 (1.45 - 3.00)
   RISK_25
0.0001
1.54 (1.24 - 1.91)
Multivariate Analyses: Comparison of PRO_10 and Ki-67 LI**
Model 1: log-likelihood = -179.38, Deviance = 188.11
   Number of positive nodes
<0.0001
1.19 (1.12 - 1.27)
   Tumor size
0.0370
1.19 (1.01 - 1.39)
   Grade
0.4200
1.25 (0.72 - 2.17)
   Ki-67 LI
0.1300
1.02 (1.00 - 1.04)
Model 2: log-likelihood = -175.75, Deviance = 180.71
   Number of positive nodes
<0.0001
1.19 (1.12 - 1.27)
   Tumor size
0.1300
1.14 (0.96 - 1.34)
   Grade
0.9600
0.99 (0.55 - 1.76)
   PRO_10
0.0092
2.12 (1.20 - 3.72)
   Ki-67 LI
0.8100
1.00 (0.97 - 1.03)
Model 3: log-likelihood = -175.78, Deviance = 180.77
   Number of positive nodes
<0.0001
1.19 (1.12 - 1.27)
   Tumor size
0.1200
1.14 (0.97 - 1.34)
   Grade
0.9400
0.98 (0.55 - 1.74)
   PRO_10
0.0026
2.03 (1.28 - 3.23)
Multivariate Analyses: Role of PGR_5***
Model 4: log-likelihood = -215.27, Deviance = 214.30
   Number of positive nodes
<0.0001
1.12 (1.07 - 1.16)
   Tumor size
0.2000
1.11 (0.95 - 1.30)
   Grade
0.0170
1.78 (1.11 - 2.87)
   PGR_5
0.0570
0.68 (0.45 - 1.01)
Model 5: log-likelihood = -211.85, Deviance = 208.03
   Number of positive nodes
<0.0001
1.06 (1.06 - 1.16)
   Tumor size
0.4300
1.07 (0.91 - 1.26)
   Grade
0.3000
1.32 (0.78 - 2.23)
   PRO_10
0.0092
1.73 (1.15 - 2.62)
   PGR_5
0.0360
0.65 (0.43 - 0.97)
Model 6: log-likelihood = -214.10, Deviance = 211.25
   Number of positive nodes
<0.0001
1.11 (1.06 - 1.16)
   Tumor size
0.1700
1.13 (0.95 - 1.34)
   Grade
0.2100
1.40 (0.83 - 2.37)
   PRO_10
0.0150
1.71 (1.11 - 2.62)
*Histological grading was analyzed according to three categories (histological grade I, II or III). Number of lymph node metastases and tumor size were continuous variables. PgR and Her2 were centrally assessed and binary IHC data were included in the analyses [30, 31]. Centrally assessed Ki-67 labeling index and molecular scores were included as continuous variables.
**Data of 299 patients with available Ki-67 LI were included in model 1, 2 and 3, respectively.
***Data of all 342 patients were included in model 4, 5 and 6, respectively.
Abbreviations: HR, hazard ratio; CI, confidence interval; LRT, likelihood ratio test; Ki-67 LI, Ki-67 labeling index.
Models 3 and 6 should not be compared directly as they were fitted on different sample sizes, due to missing data in Ki-67 LI.
PgR, as measured by immunohistochemistry [30] or microarray analysis [53], was shown to positively correlate with prognosis. Here we show that the molecular PGR_5 score was also positively associated with DFS (Figure 4) and added independent prognostic information to anatomical staging and PRO_10 (Table 3). Thus, PGR_5 and PRO_10 scores independently predict prognosis in the BIG 1-98 population.
Compared to immunohistochemically assessed parameters, qRT-PCR based scores are quantitative, relatively independent on operator expertise and less affected by inter-observer variability. The procedure is simple, economical and can be standardized easily with good control genes, reference samples and quality control procedures.
The results of this study are based on a limited number of patients and follow-up time (60 months). Similar analyses with independent, larger sample sizes and more mature follow-up data are planned to further consolidate the prognostic and possibly predictive value of the proposed scores in each treatment arm separately.
Gene expression profiling has improved the understanding of molecular subtypes of breast cancer. FFPE material is not widely used although it may facilitate and speed up the development and validation of novel gene signatures due to the availability of well-characterized tissues from numerous clinical trials [54, 55]. The same material can be used for molecular diagnostics. The investigation of gene signatures may become more important in the future as an increasing proportion of agents under development for breast cancer treatment have defined molecular targets. Early integration of biomarker analysis in the drug development process has the potential to improve the specificity and efficiency of novel therapeutics. This opens the possibility to further individualize therapy of patients with breast cancer.

Conclusions

We define four molecular scores based on quantitative measurement of gene expression with RNA derived of FFPE tissues. The genes for each score were selected from a large meta-analysis of microarrays. The genes do not coincide with genes used for other molecular scores like the RS (except genes that were previously used as immunohistochemical markers such as ER, PgR or Her2). Two of the described scores are shown to be independent predictors of disease-free survival of postmenopausal patients with operable, estrogen receptor positive breast cancer. The proliferation-associated score outperforms the Ki-67 labeling index measured by immunohistochemistry.

Acknowledgements

We thank all patients, physicians and pathologists who participated in the BIG 1-98 trial. We thank the IBCSG for providing material and corresponding clinicopathological data. We thank Dr. M. Schobesberger and K. Price for helpful discussions and critical reading of the manuscript and R. Kammler for preparing FFPE material. This work was supported by Cancer Research Switzerland/Oncosuisse, the NCCR "Molecular Oncology" and the Bernese Cancer League. The IBCSG Statistical Center receives funding from the US NCI (CA-75362). The BIG 1-98 trial was financed by Novartis and coordinated by the International Breast Cancer Study Group (funded by the Swedish Cancer Society, The Cancer Council Australia, Australian New Zealand Breast Cancer Trials Group, Frontier Science and Technology Research Foundation, Swiss Group for Clinical Cancer Research (SAKK), Cancer Research Switzerland/Oncosuisse and the Foundation for Clinical Cancer Research of Eastern Switzerland (OSKK).
BIG 1-98 Steering Committee: B. Thürlimann (Chair), S. Aebi, L. Blacher, M. Castiglione, A. S. Coates, T. Cufer, P. Dinh, J. F. Forbes, R. D. Gelber, A. Giobbie-Hurder, A. Goldhirsch, A. Hiltbrunner, S. B. Holmberg, R. Maibach, A. Martoni, L. Mauriac, G. McGrogan, H. T. Mouridsen, R. Paridaens, K. N. Price, M. Rabaglio, B.B. Rasmussen, M.M. Regan, A. Santoro, I. E. Smith, A. Wardley, G. Viale. Novartis: H. A. Chaudri-Ross, R. Dias, D. B. Evans, C. Sguotti, U. Trostmann
Swiss Group for Clinical Cancer Research (SAKK): A. Goldhirsch (up to January 2004), R. Herrmann (from June 2004): Kantonsspital Aarau, Zentrum f. Onkologie, Aarau: A. Schönenberger, W. Mingrone, Ch. Honegger, E. Bärtschi, M. Neter, M. Rederer, G. Schär; University Hospital Basel, Basel: C. Rochlitz, R. Herrmann, D. Oertli, E. Wight, H. Moch; Institute of Oncology of Southern Switzerland: Ospedale San Giovanni, Bellinzona: J. Bernier, L. Bronz, F. Cavalli, E. Gallerani, A. Richetti, A. Franzetti; Ospedale Regionale di Lugano (Civico & Italiano), Lugano: M. Conti-Beltraminelli, M. Ghielmini, T. Gyr, S. Mauri, P. C. Saletti; Ospedale Regionale Beata Vergine, Mendrisio: A. Goldhirsch, O. Pagani, R. Graffeo, M. Locatelli, S. Longhi, P.C. Rey, M. Ruggeri; Ospedale Regionale La Carità, Locarno: E. Zucca, D. Wyss; Istituto Cantonale di Patologia, Locarno: L. Mazzucchelli, E. Pedrinis, T. Rusca; Inselspital, Berne: S. Aebi, M. F. Fey, M. Castiglione, M. Rabaglio; Kantonsspital Olten, Olten: S. Aebi, M. F. Fey, M. Zuber, G. Beck; Bürgerspital, Solothurn: S. Aebi, M. F. Fey, R. Schönenberger;Spital Thun-Simmental AG Thun: J.M. Lüthi, D. Rauch; Hôpital Cantonal Universitaire HCUG, Geneva: H. Bonnefoi; Rätisches Kantons- und Regionalspital, Chur: F. Egli, R. Steiner, P. Fehr; Centre Pluridisciplinaire d'Oncologie, Lausanne: L. Perey, P. de Grandi, W. Jeanneret, S. Leyvraz, J.-F. Delaloye; Kantonsspital St. Gallen, St. Gallen: B. Thürlimann, D. Köberle, F. Weisser, S., Mattmann, A. Müller, T. Cerny, B. Späti, M. Höfliger, G. Fürstenberger, B. Bolliger, C. Öhlschlegel, U. Lorenz, M. Bamert, J. Kehl-Blank, E. Vogel; Kantonales Spital Herisau, Herisau: B. Thürlimann, D. Hess, I. Senn, D. Köberle, A. Ehrsam, C. Nauer, C. Öhlschlegel, J. Kehl-Blank, E. Vogel; Stadtspital Triemli, Zürich: L. Widmer, M. Häfner; Universitätsspital Zürich, Zürich: B. C. Pestalozzi, M. Fehr, R. Caduff, Z. Varga, R. Trüb, D. Fink.
Swiss Private MDs: Private Praxis, Zürich: B. A. Bättig; Sonnenhof-Klinik Engeried, Berne: K. Buser; Frauenklinik Limmattalspital, Schlieren: N. Bürki; Private Praxis, Birsfelden: A. Dieterle; Private Praxis, Biel: L. Hasler; Private Praxis, Baar: M. Mannhart-Harms; Brust-Zentrum, Zürich: C. Rageth; Private Praxis, Berne: J. Richner; Private Praxis, Bellinzona: V. Spataro; Private Praxis, Winterthur: M. Umbricht.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

JA, VP, MD, PW, AB, AO, AGH, GV, HJA, SA and RJ declare that they have no competing interest. B.T. holds stocks from Novartis (Ciba Geigy) since 1990.

Authors' contributions

JA, SA and RJ organized the study, planned the experiments and wrote the manuscript. SA and BT organized samples from the International Breast Cancer Study Group. AO and AB carried out RNA isolations, quality controls and gene expression measurements. VP, PW, MD and AGH carried out the statistical analyses. HJA and GV were responsible for histological assessment of stained sections. All authors contributed to the manuscript, they read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Carlson RW, Jahanzeb M, Kiel K, Marks LB, Mc Cromick B, Pierce LJ, Ward JH, Topham NS: NCCN Clinical Practice Guidelines in Oncology V.2.2008. Book NCCN Clinical Practice Guidelines in Oncology V.2. 2008, [http://www.nccn.org] Carlson RW, Jahanzeb M, Kiel K, Marks LB, Mc Cromick B, Pierce LJ, Ward JH, Topham NS: NCCN Clinical Practice Guidelines in Oncology V.2.2008. Book NCCN Clinical Practice Guidelines in Oncology V.2. 2008, [http://​www.​nccn.​org]
2.
Zurück zum Zitat Goldhirsch A, Wood WC, Gelber RD, Coates AS, Thurlimann B, Senn HJ: Progress and promise: highlights of the international expert consensus on the primary therapy of early breast cancer 2007. Ann Oncol. 2007, 18: 1133-1144. 10.1093/annonc/mdm271.CrossRefPubMed Goldhirsch A, Wood WC, Gelber RD, Coates AS, Thurlimann B, Senn HJ: Progress and promise: highlights of the international expert consensus on the primary therapy of early breast cancer 2007. Ann Oncol. 2007, 18: 1133-1144. 10.1093/annonc/mdm271.CrossRefPubMed
3.
Zurück zum Zitat Blamey RW, Pinder SE, Ball GR, Ellis IO, Elston CW, Mitchell MJ, Haybittle JL: Reading the prognosis of the individual with breast cancer. Eur J Cancer. 2007, 43: 1545-1547. 10.1016/j.ejca.2007.01.003.CrossRefPubMed Blamey RW, Pinder SE, Ball GR, Ellis IO, Elston CW, Mitchell MJ, Haybittle JL: Reading the prognosis of the individual with breast cancer. Eur J Cancer. 2007, 43: 1545-1547. 10.1016/j.ejca.2007.01.003.CrossRefPubMed
4.
Zurück zum Zitat Galea MH, Blamey RW, Elston CE, Ellis IO: The Nottingham Prognostic Index in primary breast cancer. Breast Cancer Res Treat. 1992, 22: 207-219. 10.1007/BF01840834.CrossRefPubMed Galea MH, Blamey RW, Elston CE, Ellis IO: The Nottingham Prognostic Index in primary breast cancer. Breast Cancer Res Treat. 1992, 22: 207-219. 10.1007/BF01840834.CrossRefPubMed
5.
Zurück zum Zitat Olivotto IA, Bajdik CD, Ravdin PM, Speers CH, Coldman AJ, Norris BD, Davis GJ, Chia SK, Gelmon KA: Population-based validation of the prognostic model ADJUVANT! for early breast cancer. J Clin Oncol. 2005, 23: 2716-2725. 10.1200/JCO.2005.06.178.CrossRefPubMed Olivotto IA, Bajdik CD, Ravdin PM, Speers CH, Coldman AJ, Norris BD, Davis GJ, Chia SK, Gelmon KA: Population-based validation of the prognostic model ADJUVANT! for early breast cancer. J Clin Oncol. 2005, 23: 2716-2725. 10.1200/JCO.2005.06.178.CrossRefPubMed
6.
Zurück zum Zitat Ravdin PM, Siminoff LA, Davis GJ, Mercer MB, Hewlett J, Gerson N, Parker HL: Computer program to assist in making decisions about adjuvant therapy for women with early breast cancer. J Clin Oncol. 2001, 19: 980-991.PubMed Ravdin PM, Siminoff LA, Davis GJ, Mercer MB, Hewlett J, Gerson N, Parker HL: Computer program to assist in making decisions about adjuvant therapy for women with early breast cancer. J Clin Oncol. 2001, 19: 980-991.PubMed
7.
Zurück zum Zitat Ellis MJ, Tao Y, Luo J, A'Hern R, Evans DB, Bhatnagar AS, Chaudri Ross HA, von Kameke A, Miller WR, Smith I, et al: Outcome prediction for estrogen receptor-positive breast cancer based on postneoadjuvant endocrine therapy tumor characteristics. J Natl Cancer Inst. 2008, 100: 1380-1388. 10.1093/jnci/djn309.CrossRefPubMedPubMedCentral Ellis MJ, Tao Y, Luo J, A'Hern R, Evans DB, Bhatnagar AS, Chaudri Ross HA, von Kameke A, Miller WR, Smith I, et al: Outcome prediction for estrogen receptor-positive breast cancer based on postneoadjuvant endocrine therapy tumor characteristics. J Natl Cancer Inst. 2008, 100: 1380-1388. 10.1093/jnci/djn309.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Andre F, Pusztai L: Molecular classification of breast cancer: implications for selection of adjuvant chemotherapy. Nat Clin Pract Oncol. 2006, 3: 621-632. 10.1038/ncponc0636.CrossRefPubMed Andre F, Pusztai L: Molecular classification of breast cancer: implications for selection of adjuvant chemotherapy. Nat Clin Pract Oncol. 2006, 3: 621-632. 10.1038/ncponc0636.CrossRefPubMed
9.
Zurück zum Zitat Pusztai L, Mazouni C, Anderson K, Wu Y, Symmans WF: Molecular classification of breast cancer: limitations and potential. Oncologist. 2006, 11: 868-877. 10.1634/theoncologist.11-8-868.CrossRefPubMed Pusztai L, Mazouni C, Anderson K, Wu Y, Symmans WF: Molecular classification of breast cancer: limitations and potential. Oncologist. 2006, 11: 868-877. 10.1634/theoncologist.11-8-868.CrossRefPubMed
10.
Zurück zum Zitat Brenton JD, Carey LA, Ahmed AA, Caldas C: Molecular classification and molecular forecasting of breast cancer: ready for clinical application?. J Clin Oncol. 2005, 23: 7350-7360. 10.1200/JCO.2005.03.3845.CrossRefPubMed Brenton JD, Carey LA, Ahmed AA, Caldas C: Molecular classification and molecular forecasting of breast cancer: ready for clinical application?. J Clin Oncol. 2005, 23: 7350-7360. 10.1200/JCO.2005.03.3845.CrossRefPubMed
11.
Zurück zum Zitat Buyse M, Loi S, van't Veer L, Viale G, Delorenzi M, Glas AM, d'Assignies MS, Bergh J, Lidereau R, Ellis P, et al: Validation and clinical utility of a 70-gene prognostic signature for women with node-negative breast cancer. J Natl Cancer Inst. 2006, 98: 1183-1192.CrossRefPubMed Buyse M, Loi S, van't Veer L, Viale G, Delorenzi M, Glas AM, d'Assignies MS, Bergh J, Lidereau R, Ellis P, et al: Validation and clinical utility of a 70-gene prognostic signature for women with node-negative breast cancer. J Natl Cancer Inst. 2006, 98: 1183-1192.CrossRefPubMed
12.
Zurück zum Zitat Desmedt C, Piette F, Loi S, Wang Y, Lallemand F, Haibe-Kains B, Viale G, Delorenzi M, Zhang Y, d'Assignies MS, et al: Strong time dependence of the 76-gene prognostic signature for node-negative breast cancer patients in the TRANSBIG multicenter independent validation series. Clin Cancer Res. 2007, 13: 3207-3214. 10.1158/1078-0432.CCR-06-2765.CrossRefPubMed Desmedt C, Piette F, Loi S, Wang Y, Lallemand F, Haibe-Kains B, Viale G, Delorenzi M, Zhang Y, d'Assignies MS, et al: Strong time dependence of the 76-gene prognostic signature for node-negative breast cancer patients in the TRANSBIG multicenter independent validation series. Clin Cancer Res. 2007, 13: 3207-3214. 10.1158/1078-0432.CCR-06-2765.CrossRefPubMed
13.
Zurück zum Zitat Perou CM, Sorlie T, Eisen MB, Rijn van de M, Jeffrey SS, Rees CA, Pollack JR, Ross DT, Johnsen H, Akslen LA, et al: Molecular portraits of human breast tumours. Nature. 2000, 406: 747-752. 10.1038/35021093.CrossRefPubMed Perou CM, Sorlie T, Eisen MB, Rijn van de M, Jeffrey SS, Rees CA, Pollack JR, Ross DT, Johnsen H, Akslen LA, et al: Molecular portraits of human breast tumours. Nature. 2000, 406: 747-752. 10.1038/35021093.CrossRefPubMed
14.
Zurück zum Zitat Sorlie T, Tibshirani R, Parker J, Hastie T, Marron JS, Nobel A, Deng S, Johnsen H, Pesich R, Geisler S, et al: Repeated observation of breast tumor subtypes in independent gene expression data sets. Proc Natl Acad Sci USA. 2003, 100: 8418-8423. 10.1073/pnas.0932692100.CrossRefPubMedPubMedCentral Sorlie T, Tibshirani R, Parker J, Hastie T, Marron JS, Nobel A, Deng S, Johnsen H, Pesich R, Geisler S, et al: Repeated observation of breast tumor subtypes in independent gene expression data sets. Proc Natl Acad Sci USA. 2003, 100: 8418-8423. 10.1073/pnas.0932692100.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Sotiriou C, Piccart MJ: Taking gene-expression profiling to the clinic: when will molecular signatures become relevant to patient care?. Nat Rev Cancer. 2007, 7: 545-553. 10.1038/nrc2173.CrossRefPubMed Sotiriou C, Piccart MJ: Taking gene-expression profiling to the clinic: when will molecular signatures become relevant to patient care?. Nat Rev Cancer. 2007, 7: 545-553. 10.1038/nrc2173.CrossRefPubMed
16.
Zurück zum Zitat Sotiriou C, Wirapati P, Loi S, Harris A, Fox S, Smeds J, Nordgren H, Farmer P, Praz V, Haibe-Kains B, et al: Gene expression profiling in breast cancer: understanding the molecular basis of histologic grade to improve prognosis. J Natl Cancer Inst. 2006, 98: 262-272.CrossRefPubMed Sotiriou C, Wirapati P, Loi S, Harris A, Fox S, Smeds J, Nordgren H, Farmer P, Praz V, Haibe-Kains B, et al: Gene expression profiling in breast cancer: understanding the molecular basis of histologic grade to improve prognosis. J Natl Cancer Inst. 2006, 98: 262-272.CrossRefPubMed
17.
Zurück zum Zitat Stadler ZK, Come SE: Review of gene-expression profiling and its clinical use in breast cancer. Crit Rev Oncol Hematol. 2008, 1-11. Stadler ZK, Come SE: Review of gene-expression profiling and its clinical use in breast cancer. Crit Rev Oncol Hematol. 2008, 1-11.
18.
Zurück zum Zitat Vijver van de MJ, He YD, van't Veer LJ, Dai H, Hart AA, Voskuil DW, Schreiber GJ, Peterse JL, Roberts C, Marton MJ, et al: A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med. 2002, 347: 1999-2009. 10.1056/NEJMoa021967.CrossRefPubMed Vijver van de MJ, He YD, van't Veer LJ, Dai H, Hart AA, Voskuil DW, Schreiber GJ, Peterse JL, Roberts C, Marton MJ, et al: A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med. 2002, 347: 1999-2009. 10.1056/NEJMoa021967.CrossRefPubMed
19.
Zurück zum Zitat van 't Veer LJ, Dai H, Vijver van de MJ, He YD, Hart AA, Mao M, Peterse HL, Kooy van der K, Marton MJ, Witteveen AT, et al: Gene expression profiling predicts clinical outcome of breast cancer. Nature. 2002, 415: 530-536. 10.1038/415530a.CrossRefPubMed van 't Veer LJ, Dai H, Vijver van de MJ, He YD, Hart AA, Mao M, Peterse HL, Kooy van der K, Marton MJ, Witteveen AT, et al: Gene expression profiling predicts clinical outcome of breast cancer. Nature. 2002, 415: 530-536. 10.1038/415530a.CrossRefPubMed
20.
Zurück zum Zitat Wang Y, Klijn JG, Zhang Y, Sieuwerts AM, Look MP, Yang F, Talantov D, Timmermans M, Meijer-van Gelder ME, Yu J, et al: Gene-expression profiles to predict distant metastasis of lymph-node-negative primary breast cancer. Lancet. 2005, 365: 671-679.CrossRefPubMed Wang Y, Klijn JG, Zhang Y, Sieuwerts AM, Look MP, Yang F, Talantov D, Timmermans M, Meijer-van Gelder ME, Yu J, et al: Gene-expression profiles to predict distant metastasis of lymph-node-negative primary breast cancer. Lancet. 2005, 365: 671-679.CrossRefPubMed
21.
Zurück zum Zitat Sparano JA, Paik S: Development of the 21-gene assay and its application in clinical practice and clinical trials. J Clin Oncol. 2008, 26: 721-728. 10.1200/JCO.2007.15.1068.CrossRefPubMed Sparano JA, Paik S: Development of the 21-gene assay and its application in clinical practice and clinical trials. J Clin Oncol. 2008, 26: 721-728. 10.1200/JCO.2007.15.1068.CrossRefPubMed
22.
Zurück zum Zitat Cardoso F, Van't Veer L, Rutgers E, Loi S, Mook S, Piccart-Gebhart MJ: Clinical application of the 70-gene profile: the MINDACT trial. J Clin Oncol. 2008, 26: 729-735. 10.1200/JCO.2007.14.3222.CrossRefPubMed Cardoso F, Van't Veer L, Rutgers E, Loi S, Mook S, Piccart-Gebhart MJ: Clinical application of the 70-gene profile: the MINDACT trial. J Clin Oncol. 2008, 26: 729-735. 10.1200/JCO.2007.14.3222.CrossRefPubMed
23.
Zurück zum Zitat Viale G, Giobbie-Hurder A, Regan MM, Coates AS, Mastropasqua MG, Dell'Orto P, Maiorano E, MacGrogan G, Braye SG, Ohlschlegel C, et al: Prognostic and predictive value of centrally reviewed Ki-67 labeling index in postmenopausal women with endocrine-responsive breast cancer: results from Breast International Group Trial 1-98 comparing adjuvant tamoxifen with letrozole. J Clin Oncol. 2008, 26: 5569-5575. 10.1200/JCO.2008.17.0829.CrossRefPubMedPubMedCentral Viale G, Giobbie-Hurder A, Regan MM, Coates AS, Mastropasqua MG, Dell'Orto P, Maiorano E, MacGrogan G, Braye SG, Ohlschlegel C, et al: Prognostic and predictive value of centrally reviewed Ki-67 labeling index in postmenopausal women with endocrine-responsive breast cancer: results from Breast International Group Trial 1-98 comparing adjuvant tamoxifen with letrozole. J Clin Oncol. 2008, 26: 5569-5575. 10.1200/JCO.2008.17.0829.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Coates AS, Keshaviah A, Thurlimann B, Mouridsen H, Mauriac L, Forbes JF, Paridaens R, Castiglione-Gertsch M, Gelber RD, Colleoni M, et al: Five years of letrozole compared with tamoxifen as initial adjuvant therapy for postmenopausal women with endocrine-responsive early breast cancer: update of study BIG 1-98. J Clin Oncol. 2007, 25: 486-492. 10.1200/JCO.2006.08.8617.CrossRefPubMed Coates AS, Keshaviah A, Thurlimann B, Mouridsen H, Mauriac L, Forbes JF, Paridaens R, Castiglione-Gertsch M, Gelber RD, Colleoni M, et al: Five years of letrozole compared with tamoxifen as initial adjuvant therapy for postmenopausal women with endocrine-responsive early breast cancer: update of study BIG 1-98. J Clin Oncol. 2007, 25: 486-492. 10.1200/JCO.2006.08.8617.CrossRefPubMed
25.
Zurück zum Zitat Thurlimann B, Keshaviah A, Coates AS, Mouridsen H, Mauriac L, Forbes JF, Paridaens R, Castiglione-Gertsch M, Gelber RD, Rabaglio M, et al: A comparison of letrozole and tamoxifen in postmenopausal women with early breast cancer. N Engl J Med. 2005, 353: 2747-2757. 10.1056/NEJMoa052258.CrossRefPubMed Thurlimann B, Keshaviah A, Coates AS, Mouridsen H, Mauriac L, Forbes JF, Paridaens R, Castiglione-Gertsch M, Gelber RD, Rabaglio M, et al: A comparison of letrozole and tamoxifen in postmenopausal women with early breast cancer. N Engl J Med. 2005, 353: 2747-2757. 10.1056/NEJMoa052258.CrossRefPubMed
26.
Zurück zum Zitat Mouridsen H, Giobbie-Hurder A, Goldhirsch A, Thurlimann B, Paridaens R, Smith I, Mauriac L, Forbes JF, Price KN, Regan MM, et al: Letrozole therapy alone or in sequence with tamoxifen in women with breast cancer. N Engl J Med. 2009, 361: 766-776. 10.1056/NEJMoa0810818.CrossRefPubMed Mouridsen H, Giobbie-Hurder A, Goldhirsch A, Thurlimann B, Paridaens R, Smith I, Mauriac L, Forbes JF, Price KN, Regan MM, et al: Letrozole therapy alone or in sequence with tamoxifen in women with breast cancer. N Engl J Med. 2009, 361: 766-776. 10.1056/NEJMoa0810818.CrossRefPubMed
27.
Zurück zum Zitat Oberli A, Popovici V, Delorenzi M, Baltzer A, Antonov J, Matthey S, Aebi S, Altermatt HJ, Jaggi R: Expression profiling with RNA from formalin-fixed, paraffin-embedded material. BMC Med Genomics. 2008, 1: 1-9. 10.1186/1755-8794-1-9.CrossRef Oberli A, Popovici V, Delorenzi M, Baltzer A, Antonov J, Matthey S, Aebi S, Altermatt HJ, Jaggi R: Expression profiling with RNA from formalin-fixed, paraffin-embedded material. BMC Med Genomics. 2008, 1: 1-9. 10.1186/1755-8794-1-9.CrossRef
28.
Zurück zum Zitat Wirapati P, Sotiriou C, Kunkel S, Farmer P, Pradervand S, Haibe-Kains B, Desmedt C, Ignatiadis M, Sengstag T, Schutz F, et al: Meta-analysis of gene-expression profiles in breast cancer: toward a unified understanding of breast cancer sub-typing and prognosis signatures. Breast Cancer Res. 2008, 10: R65-10.1186/bcr2124.CrossRefPubMedPubMedCentral Wirapati P, Sotiriou C, Kunkel S, Farmer P, Pradervand S, Haibe-Kains B, Desmedt C, Ignatiadis M, Sengstag T, Schutz F, et al: Meta-analysis of gene-expression profiles in breast cancer: toward a unified understanding of breast cancer sub-typing and prognosis signatures. Breast Cancer Res. 2008, 10: R65-10.1186/bcr2124.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Paik S, Shak S, Tang G, Kim C, Baker J, Cronin M, Baehner FL, Walker MG, Watson D, Park T, et al: A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. N Engl J Med. 2004, 351: 2817-2826. 10.1056/NEJMoa041588.CrossRefPubMed Paik S, Shak S, Tang G, Kim C, Baker J, Cronin M, Baehner FL, Walker MG, Watson D, Park T, et al: A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. N Engl J Med. 2004, 351: 2817-2826. 10.1056/NEJMoa041588.CrossRefPubMed
30.
Zurück zum Zitat Viale G, Regan MM, Maiorano E, Mastropasqua MG, Dell'Orto P, Rasmussen BB, Raffoul J, Neven P, Orosz Z, Braye S, et al: Prognostic and predictive value of centrally reviewed expression of estrogen and progesterone receptors in a randomized trial comparing letrozole and tamoxifen adjuvant therapy for postmenopausal early breast cancer: BIG 1-98. J Clin Oncol. 2007, 25: 3846-3852. 10.1200/JCO.2007.11.9453.CrossRefPubMed Viale G, Regan MM, Maiorano E, Mastropasqua MG, Dell'Orto P, Rasmussen BB, Raffoul J, Neven P, Orosz Z, Braye S, et al: Prognostic and predictive value of centrally reviewed expression of estrogen and progesterone receptors in a randomized trial comparing letrozole and tamoxifen adjuvant therapy for postmenopausal early breast cancer: BIG 1-98. J Clin Oncol. 2007, 25: 3846-3852. 10.1200/JCO.2007.11.9453.CrossRefPubMed
31.
Zurück zum Zitat Rasmussen BB, Regan MM, Lykkesfeldt AE, Dell'Orto P, Del Curto B, Henriksen KL, Mastropasqua MG, Price KN, Mery E, Lacroix-Triki M, et al: Adjuvant letrozole versus tamoxifen according to centrally-assessed ERBB2 status for postmenopausal women with endocrine-responsive early breast cancer: supplementary results from the BIG 1-98 randomised trial. Lancet Oncol. 2008, 9: 23-28. 10.1016/S1470-2045(07)70386-8.CrossRefPubMed Rasmussen BB, Regan MM, Lykkesfeldt AE, Dell'Orto P, Del Curto B, Henriksen KL, Mastropasqua MG, Price KN, Mery E, Lacroix-Triki M, et al: Adjuvant letrozole versus tamoxifen according to centrally-assessed ERBB2 status for postmenopausal women with endocrine-responsive early breast cancer: supplementary results from the BIG 1-98 randomised trial. Lancet Oncol. 2008, 9: 23-28. 10.1016/S1470-2045(07)70386-8.CrossRefPubMed
32.
Zurück zum Zitat Viale G, Regan MM, Mastropasqua MG, Maffini F, Maiorano E, Colleoni M, Price KN, Golouh R, Perin T, Brown RW, et al: Predictive value of tumor Ki-67 expression in two randomized trials of adjuvant chemoendocrine therapy for node-negative breast cancer. J Natl Cancer Inst. 2008, 100: 207-212. 10.1093/jnci/djm289.CrossRefPubMed Viale G, Regan MM, Mastropasqua MG, Maffini F, Maiorano E, Colleoni M, Price KN, Golouh R, Perin T, Brown RW, et al: Predictive value of tumor Ki-67 expression in two randomized trials of adjuvant chemoendocrine therapy for node-negative breast cancer. J Natl Cancer Inst. 2008, 100: 207-212. 10.1093/jnci/djm289.CrossRefPubMed
33.
Zurück zum Zitat Hayes DF, Thor AD, Dressler LG, Weaver D, Edgerton S, Cowan D, Broadwater G, Goldstein LJ, Martino S, Ingle JN, et al: HER2 and response to paclitaxel in node-positive breast cancer. N Engl J Med. 2007, 357: 1496-1506. 10.1056/NEJMoa071167.CrossRefPubMed Hayes DF, Thor AD, Dressler LG, Weaver D, Edgerton S, Cowan D, Broadwater G, Goldstein LJ, Martino S, Ingle JN, et al: HER2 and response to paclitaxel in node-positive breast cancer. N Engl J Med. 2007, 357: 1496-1506. 10.1056/NEJMoa071167.CrossRefPubMed
34.
Zurück zum Zitat Schemper M, Smith TL: A note on quantifying follow-up in studies of failure time. Control Clin Trials. 1996, 17: 343-346. 10.1016/0197-2456(96)00075-X.CrossRefPubMed Schemper M, Smith TL: A note on quantifying follow-up in studies of failure time. Control Clin Trials. 1996, 17: 343-346. 10.1016/0197-2456(96)00075-X.CrossRefPubMed
35.
Zurück zum Zitat Cox DR: Regression models and life-tables. J R Stat Soc B. 1972, 34: 187-220. Cox DR: Regression models and life-tables. J R Stat Soc B. 1972, 34: 187-220.
36.
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
37.
Zurück zum Zitat Ma XJ, Hilsenbeck SG, Wang W, Ding L, Sgroi DC, Bender RA, Osborne CK, Allred DC, Erlander MG: The HOXB13:IL17BR expression index is a prognostic factor in early-stage breast cancer. J Clin Oncol. 2006, 24: 4611-4619. 10.1200/JCO.2006.06.6944.CrossRefPubMed Ma XJ, Hilsenbeck SG, Wang W, Ding L, Sgroi DC, Bender RA, Osborne CK, Allred DC, Erlander MG: The HOXB13:IL17BR expression index is a prognostic factor in early-stage breast cancer. J Clin Oncol. 2006, 24: 4611-4619. 10.1200/JCO.2006.06.6944.CrossRefPubMed
38.
Zurück zum Zitat Ma XJ, Salunga R, Dahiya S, Wang W, Carney E, Durbecq V, Harris A, Goss P, Sotiriou C, Erlander M, Sgroi D: A five-gene molecular grade index and HOXB13:IL17BR are complementary prognostic factors in early stage breast cancer. Clin Cancer Res. 2008, 14: 2601-2608. 10.1158/1078-0432.CCR-07-5026.CrossRefPubMed Ma XJ, Salunga R, Dahiya S, Wang W, Carney E, Durbecq V, Harris A, Goss P, Sotiriou C, Erlander M, Sgroi D: A five-gene molecular grade index and HOXB13:IL17BR are complementary prognostic factors in early stage breast cancer. Clin Cancer Res. 2008, 14: 2601-2608. 10.1158/1078-0432.CCR-07-5026.CrossRefPubMed
39.
Zurück zum Zitat Cronin M, Pho M, Dutta D, Stephans JC, Shak S, Kiefer MC, Esteban JM, Baker JB: Measurement of gene expression in archival paraffin-embedded tissues: development and performance of a 92-gene reverse transcriptase-polymerase chain reaction assay. Am J Pathol. 2004, 164: 35-42.CrossRefPubMedPubMedCentral Cronin M, Pho M, Dutta D, Stephans JC, Shak S, Kiefer MC, Esteban JM, Baker JB: Measurement of gene expression in archival paraffin-embedded tissues: development and performance of a 92-gene reverse transcriptase-polymerase chain reaction assay. Am J Pathol. 2004, 164: 35-42.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat von Ahlfen S, Missel A, Bendrat K, Schlumpberger M: Determinants of RNA quality from FFPE samples. PLoS ONE. 2007, 2: e1261-10.1371/journal.pone.0001261.CrossRefPubMedPubMedCentral von Ahlfen S, Missel A, Bendrat K, Schlumpberger M: Determinants of RNA quality from FFPE samples. PLoS ONE. 2007, 2: e1261-10.1371/journal.pone.0001261.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Goldstein LJ, Gray R, Badve S, Childs BH, Yoshizawa C, Rowley S, Shak S, Baehner FL, Ravdin PM, Davidson NE, et al: Prognostic Utility of the 21-Gene Assay in Hormone Receptor-Positive Operable Breast Cancer Compared With Classical Clinicopathologic Features. J Clin Oncol. 2008, 26: 4063-4071. 10.1200/JCO.2007.14.4501.CrossRefPubMedPubMedCentral Goldstein LJ, Gray R, Badve S, Childs BH, Yoshizawa C, Rowley S, Shak S, Baehner FL, Ravdin PM, Davidson NE, et al: Prognostic Utility of the 21-Gene Assay in Hormone Receptor-Positive Operable Breast Cancer Compared With Classical Clinicopathologic Features. J Clin Oncol. 2008, 26: 4063-4071. 10.1200/JCO.2007.14.4501.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Paik S: Methods for gene expression profiling in clinical trials of adjuvant breast cancer therapy. Clin Cancer Res. 2006, 12: 1019s-1023s. 10.1158/1078-0432.CCR-05-2296.CrossRefPubMed Paik S: Methods for gene expression profiling in clinical trials of adjuvant breast cancer therapy. Clin Cancer Res. 2006, 12: 1019s-1023s. 10.1158/1078-0432.CCR-05-2296.CrossRefPubMed
43.
Zurück zum Zitat Carlson RW, Allred DC, Anderson BO, Burstein HJ, Carter WB, Edge SB, Erban JK, Farrar WB, Goldstein LJ, Gradishar WJ, et al: NCCN Practice Guidelines in Oncology: Breast Cancer. 2009, v.1 Carlson RW, Allred DC, Anderson BO, Burstein HJ, Carter WB, Edge SB, Erban JK, Farrar WB, Goldstein LJ, Gradishar WJ, et al: NCCN Practice Guidelines in Oncology: Breast Cancer. 2009, v.1
44.
Zurück zum Zitat Desmedt C, Giobbie-Hurder A, Neven P, Paridaens R, Christiaens MR, Smeets A, Lallemand F, Haibe-Kains B, Viale G, Gelber RD, et al: The Gene expression Grade Index: a potential predictor of relapse for endocrine-treated breast cancer patients in the BIG 1-98 trial. BMC Med Genomics. 2009, 2: 40-10.1186/1755-8794-2-40.CrossRefPubMedPubMedCentral Desmedt C, Giobbie-Hurder A, Neven P, Paridaens R, Christiaens MR, Smeets A, Lallemand F, Haibe-Kains B, Viale G, Gelber RD, et al: The Gene expression Grade Index: a potential predictor of relapse for endocrine-treated breast cancer patients in the BIG 1-98 trial. BMC Med Genomics. 2009, 2: 40-10.1186/1755-8794-2-40.CrossRefPubMedPubMedCentral
45.
Zurück zum Zitat Paik S, Tang G, Shak S, Kim C, Baker J, Kim W, Cronin M, Baehner FL, Watson D, Bryant J, et al: Gene expression and benefit of chemotherapy in women with node-negative, estrogen receptor-positive breast cancer. J Clin Oncol. 2006, 24: 3726-3734. 10.1200/JCO.2005.04.7985.CrossRefPubMed Paik S, Tang G, Shak S, Kim C, Baker J, Kim W, Cronin M, Baehner FL, Watson D, Bryant J, et al: Gene expression and benefit of chemotherapy in women with node-negative, estrogen receptor-positive breast cancer. J Clin Oncol. 2006, 24: 3726-3734. 10.1200/JCO.2005.04.7985.CrossRefPubMed
46.
Zurück zum Zitat Liedtke C, Hatzis C, Symmans WF, Desmedt C, Haibe-Kains B, Valero V, Kuerer H, Hortobagyi GN, Piccart-Gebhart M, Sotiriou C, Pusztai L: Genomic grade index is associated with response to chemotherapy in patients with breast cancer. J Clin Oncol. 2009, 27: 3185-3191. 10.1200/JCO.2008.18.5934.CrossRefPubMedPubMedCentral Liedtke C, Hatzis C, Symmans WF, Desmedt C, Haibe-Kains B, Valero V, Kuerer H, Hortobagyi GN, Piccart-Gebhart M, Sotiriou C, Pusztai L: Genomic grade index is associated with response to chemotherapy in patients with breast cancer. J Clin Oncol. 2009, 27: 3185-3191. 10.1200/JCO.2008.18.5934.CrossRefPubMedPubMedCentral
47.
Zurück zum Zitat Schobesberger M, Baltzer A, Oberli A, Kappeler A, Gugger M, Burger H, Jaggi R: Gene expression variation between distinct areas of breast cancer measured from paraffin-embedded tissue cores. BMC Cancer. 2008, 8: 343-10.1186/1471-2407-8-343.CrossRefPubMedPubMedCentral Schobesberger M, Baltzer A, Oberli A, Kappeler A, Gugger M, Burger H, Jaggi R: Gene expression variation between distinct areas of breast cancer measured from paraffin-embedded tissue cores. BMC Cancer. 2008, 8: 343-10.1186/1471-2407-8-343.CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Viale G, Giobbie-Hurder A, BIG 1-98 Collaborative Group and International Breast Cancer Study Group (IBCSG): Value of centrally-assessed Ki-67 labeling index as a marker of prognosis and predictor of response to adjuvant endocrine therapy in the BIG 1-98 trial of postmenopausal women with estrogen receptor-positive breast cancer. Breast Cancer Res Treat. 2007, 106 (Supplement 1): S17-Abstract 64 Viale G, Giobbie-Hurder A, BIG 1-98 Collaborative Group and International Breast Cancer Study Group (IBCSG): Value of centrally-assessed Ki-67 labeling index as a marker of prognosis and predictor of response to adjuvant endocrine therapy in the BIG 1-98 trial of postmenopausal women with estrogen receptor-positive breast cancer. Breast Cancer Res Treat. 2007, 106 (Supplement 1): S17-Abstract 64
49.
Zurück zum Zitat Carlson RW, Allred DC, Anderson BO, Burstein HJ, Carter WB, Edge SB, Erban JK, Farrar WB, Goldstein LJ, Gradishar WJ, et al: Breast cancer. Clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2009, 7: 122-192.PubMed Carlson RW, Allred DC, Anderson BO, Burstein HJ, Carter WB, Edge SB, Erban JK, Farrar WB, Goldstein LJ, Gradishar WJ, et al: Breast cancer. Clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2009, 7: 122-192.PubMed
50.
Zurück zum Zitat de Azambuja E, Cardoso F, de Castro G, Colozza M, Mano MS, Durbecq V, Sotiriou C, Larsimont D, Piccart-Gebhart MJ, Paesmans M: Ki-67 as prognostic marker in early breast cancer: a meta-analysis of published studies involving 12,155 patients. Br J Cancer. 2007, 96: 1504-1513. 10.1038/sj.bjc.6603756.CrossRefPubMedPubMedCentral de Azambuja E, Cardoso F, de Castro G, Colozza M, Mano MS, Durbecq V, Sotiriou C, Larsimont D, Piccart-Gebhart MJ, Paesmans M: Ki-67 as prognostic marker in early breast cancer: a meta-analysis of published studies involving 12,155 patients. Br J Cancer. 2007, 96: 1504-1513. 10.1038/sj.bjc.6603756.CrossRefPubMedPubMedCentral
51.
Zurück zum Zitat Whitfield ML, George LK, Grant GD, Perou CM: Common markers of proliferation. Nat Rev Cancer. 2006, 6: 99-106. 10.1038/nrc1802.CrossRefPubMed Whitfield ML, George LK, Grant GD, Perou CM: Common markers of proliferation. Nat Rev Cancer. 2006, 6: 99-106. 10.1038/nrc1802.CrossRefPubMed
52.
Zurück zum Zitat Urruticoechea A, Smith IE, Dowsett M: Proliferation marker Ki-67 in early breast cancer. J Clin Oncol. 2005, 23: 7212-7220. 10.1200/JCO.2005.07.501.CrossRefPubMed Urruticoechea A, Smith IE, Dowsett M: Proliferation marker Ki-67 in early breast cancer. J Clin Oncol. 2005, 23: 7212-7220. 10.1200/JCO.2005.07.501.CrossRefPubMed
53.
Zurück zum Zitat Loi S, Haibe-Kains B, Desmedt C, Lallemand F, Tutt AM, Gillet C, Ellis P, Harris A, Bergh J, Foekens JA, et al: Definition of clinically distinct molecular subtypes in estrogen receptor-positive breast carcinomas through genomic grade. J Clin Oncol. 2007, 25: 1239-1246. 10.1200/JCO.2006.07.1522.CrossRefPubMed Loi S, Haibe-Kains B, Desmedt C, Lallemand F, Tutt AM, Gillet C, Ellis P, Harris A, Bergh J, Foekens JA, et al: Definition of clinically distinct molecular subtypes in estrogen receptor-positive breast carcinomas through genomic grade. J Clin Oncol. 2007, 25: 1239-1246. 10.1200/JCO.2006.07.1522.CrossRefPubMed
54.
Zurück zum Zitat Chang JC, Makris A, Gutierrez MC, Hilsenbeck SG, Hackett JR, Jeong J, Liu ML, Baker J, Clark-Langone K, Baehner FL, et al: Gene expression patterns in formalin-fixed, paraffin-embedded core biopsies predict docetaxel chemosensitivity in breast cancer patients. Breast Cancer Res Treat. 2008, 108: 233-240. 10.1007/s10549-007-9590-z.CrossRefPubMed Chang JC, Makris A, Gutierrez MC, Hilsenbeck SG, Hackett JR, Jeong J, Liu ML, Baker J, Clark-Langone K, Baehner FL, et al: Gene expression patterns in formalin-fixed, paraffin-embedded core biopsies predict docetaxel chemosensitivity in breast cancer patients. Breast Cancer Res Treat. 2008, 108: 233-240. 10.1007/s10549-007-9590-z.CrossRefPubMed
55.
Zurück zum Zitat Paik S: Molecular assays to predict prognosis of breast cancer. Clin Adv Hematol Oncol. 2007, 5: 681-682.PubMed Paik S: Molecular assays to predict prognosis of breast cancer. Clin Adv Hematol Oncol. 2007, 5: 681-682.PubMed
Metadaten
Titel
Molecular risk assessment of BIG 1-98 participants by expression profiling using RNA from archival tissue
verfasst von
Janine Antonov
Vlad Popovici
Mauro Delorenzi
Pratyaksha Wirapati
Anna Baltzer
Andrea Oberli
Beat Thürlimann
Anita Giobbie-Hurder
Giuseppe Viale
Hans Jörg Altermatt
Stefan Aebi
Rolf Jaggi
Publikationsdatum
01.12.2010
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2010
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/1471-2407-10-37

Weitere Artikel der Ausgabe 1/2010

BMC Cancer 1/2010 Zur Ausgabe

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

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