Skip to main content
Erschienen in: BMC Cancer 1/2014

Open Access 01.12.2014 | Research article

Systematic assessment of prognostic gene signatures for breast cancer shows distinct influence of time and ER status

verfasst von: Xi Zhao, Einar Andreas Rødland, Therese Sørlie, Hans Kristian Moen Vollan, Hege G Russnes, Vessela N Kristensen, Ole Christian Lingjærde, Anne-Lise Børresen-Dale

Erschienen in: BMC Cancer | Ausgabe 1/2014

Abstract

Background

The aim was to assess and compare prognostic power of nine breast cancer gene signatures (Intrinsic, PAM50, 70-gene, 76-gene, Genomic-Grade-Index, 21-gene-Recurrence-Score, EndoPredict, Wound-Response and Hypoxia) in relation to ER status and follow-up time.

Methods

A gene expression dataset from 947 breast tumors was used to evaluate the signatures for prediction of Distant Metastasis Free Survival (DMFS). A total of 912 patients had available DMFS status. The recently published METABRIC cohort was used as an additional validation set.

Results

Survival predictions were fairly concordant across most signatures. Prognostic power declined with follow-up time. During the first 5 years of followup, all signatures except for Hypoxia were predictive for DMFS in ER-positive disease, and 76-gene, Hypoxia and Wound-Response were prognostic in ER-negative disease. After 5 years, the signatures had little prognostic power. Gene signatures provide significant prognostic information beyond tumor size, node status and histological grade.

Conclusions

Generally, these signatures performed better for ER-positive disease, indicating that risk within each ER stratum is driven by distinct underlying biology. Most of the signatures were strong risk predictors for DMFS during the first 5 years of follow-up. Combining gene signatures with histological grade or tumor size, could improve the prognostic power, perhaps also of long-term survival.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1471-2407-14-211) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.
Abkürzungen
70-gene
70-gene gene signature (MammaPrint®)
76-gene
76-gene gene signature
DMFS
Distant Metastasis Free Survival
ER
Estrogen receptor
GGI
Genomic Grade Index
HR
hazard ratio
Hypoxia
Hypoxia gene signature
Intrinsic
Intrinsic signature
PAM50
PAM50 signature
ROR
Risk Of Relapse
RS
21-gene-recurrence-score (Oncotype DX®)
WR
Wound Response signature.

Background

Breast cancer is a heterogeneous disease. Tumors with similar clinico-pathological characteristics can have markedly different clinical courses. Gene signatures developed from genome-wide expression profiling of breast cancer have been shown to provide overlapping clinico-pathological classifications, and more importantly, to add prognostic accuracy and could potentially guide clinical decisions [19].
Despite the fact that a large number of expression-based gene signatures have been developed for breast cancer for prognostic and predictive purpose, the clinical value of these signatures has not been confirmed in prospective studies and the consequence for therapy remains unclear. The 10-year results of ongoing clinical trials [10, 11] for testing the clinical benefit of gene signatures [4, 12] will not be available until 2020. Outcome prediction by gene signatures has been criticized for being inaccurate [13]. Most studies evaluating various signatures [1418] have been carried out on relatively small scales. Compatibility between the signatures and the targeted cohorts with respect to biological and pathological characteristics (Additional file 1: Table S1) is often ignored [16]. Use of validation sets not completely independent of the original training sets may have influenced the results leading to biased interpretation [14]. Furthermore, computing signature scores from inadequately transformed data may have resulted in unreliable or spurious results [19, 20]. It therefore remains desirable to evaluate existing signatures in greater scrutiny on a reasonably sized and representative breast cancer cohort and pinpoint important specifications for more effective use of molecular-based tests in clinical settings.
With this in mind, we investigated nine signatures that have received great interest and been validated in multiple studies. These are Intrinsic signature [13, 21] and PAM50 [9] for classifying breast tumors into five subtypes: luminal A (LumA), luminal B (LumB), HER2-enriched, basal-like, and normal-like; 70-gene profile or MammaPrint® (Agendia, Amsterdam, The Netherlands) [4, 5, 2224] for predicting metastasis free survival over a five-year period; 76-gene signature [6, 25, 26] for predicting distant metastasis within five years for lymph-node-negative breast cancers; genomic grade index (GGI) [7, 27] for reclassifying histologic grade (HG) 2 tumors into HG1-like or HG3-like groups; Wound-Response (WR) signature [28, 29] for classifying tumors into activated or quiescent WR groups; Hypoxia signature [15, 30] for assigning hypoxic or non-hypoxic tumors; 21-gene-recurrence-score (RS) or Oncotype DX® (Genomic Health Inc., Redwood City, CA) [12] for predicting distant recurrence at ten years in adjuvant-tamoxifen-treated patients [12, 31] and EndoPredict (EP) [32], a recently developed 11-gene assay for predicting distant recurrence at ten years in ER-positive and HER2-negative patients who were treated with adjuvant hormonal therapy.
We found that the prognostic effects of signatures declined with follow-up time and were generally better in ER-positive than ER-negative disease. In particular, signatures that had strong predictive power in ER-positive disease, mostly had little predictive power in ER-negative disease, the main exception being WR which had some predictive power also in ER-negative disease; on the other hand, Hypoxia was the only signature with clear predictive power in ER-negative disease, but had no predictive power in ER-positive disease. This illustrates the need for designing robust prognostic tools separately for ER-positive and ER-negative disease.

Methods

Detailed description, together with reproducible code and data, are provided in the Additional files 2, 3 and 4, respectively.

Microarray Data

The gene expression dataset [33] (n = 947) is a collection of six published breast cancer microarray datasets [26, 27, 3437] on Affymetrix Human Genome HG-U133A arrays. The datasets were retrieved from Gene Expression Omnibus [38] (http://​www.​ncbi.​nlm.​nih.​gov/​geo) and ArrayExpress (http://​www.​ebi.​ac.​uk/​arrayexpress) under accession number GSE6532 [27], GSE3494 [34], GSE1456 [35], GSE7390 [26], GSE2603 [36] and E-TABM-158 [37] respectively. Data were processed and RMA-normalized [39] as previously described [33].

Clinical data

We compiled comprehensive clinical information on these 947 samples in addition to what have been collected previously [33]. This includes additional and up-to-date (if available) information on ER status [35], node status [35], tumor size [35], and DMFS follow-ups [34, 35], and treatment information [26, 27, 3437].
Distant Metastasis Free Survival (DMFS: n = 912) was used as clinical endpoint (Additional file 1: Table S2A). Additional file 1: Table S2B summarizes the clinicopathological characteristics with respect to the clinical endpoint. For tumors lacking ER and HER2 status from standard immunohistochemistry (or FISH), the gene expression value for ESR1 and ERBB2, respectively, were used [33]. Among 335 tumors [34, 37] with available TP53 mutation status, 82 tumors were TP53-mutated and 253 were wild-type. Pathological characteristics including tumor size (DMFS: n = 905), lymph node status (DMFS: n = 893) and histological grade (DMFS: n = 781) were recorded. Datasets with adjuvant treatment information [26, 27, 34, 35, 37] included 403 patients (DMFS: n = 395) who did not receive systemic treatment.

Applicability of signatures

We investigated all nine signatures (Table 1; Additional file 1: Table S1) on the full dataset (n = 947), although some of the signatures were originally developed on specific patient subgroups. Most analyses were done separately for ER-positive and ER-negative disease. While RS [12] has only been applied to ER-positive breast cancer, and GGI [7, 27] was developed on ER-positive and only later validated on ER-negative disease (Supplement), for completion we have included both along with the other signatures in the analyses on ER-negative disease.
Table 1
Summary of gene signatures in the study and the annotation mapping coverage
Signature
Description
Validation
Training platform
Coverage (%)
Intrinsic [1, 21]
Intrinsic subtype
Ref. [2, 3]
Stanford cDNA array
410/549 (75%)
PAM50 [9]
PAM50 subtype
Ref. [9]
Agilent Human oligo array
42/50 (84%)
70gene [4]
MammaPrint
Ref. [5, 2224]
Agilent 25 k Human oligo array
46/70 (65.7%)
76gene [6]
Veridex
Ref. [25, 26]
Affymetrix u133a GeneChip
76/76 (100%)
Hypoxia [30]
Hypoxia signature
Ref. [15, 30]
Stanford cDNA array
117/253 (46.2%)a
WR [28]
Wound response
Ref. [15, 29]
Stanford cDNA array
298/380 (78.4%)
GGI [7]
Genomic Grade Index
Ref. [17, 27]
Affymetrix u133a GeneChip
128/128 (100%)
RS [12]
OncoType DX Recurrent Score
Ref. [12, 31]
qRT-PCR
21/21 (100%)
EP [32]
EndoPredict risk score
Ref. [32, 40]
qRT-PCR
11/11 (100%)
aThe original study [30] reported 168 UniGene IDs annotated from 253 clones in the Hypoxia signature, of which 117 clones mapped to the Affy probes (46.2%). Considering these mapped clones represented 116 unique Unigene clusters (under UniGene Build Number 222), the mapping coverage for this signature on the studied data is higher than the percentage reported here.
The 76-gene signature has only been validated in node-negative disease [25, 26], but we found that it was also a valid predictor on node-positive disease and have therefore assessed it on the full dataset. Indeed, several of the signatures were originally developed on node-negative disease, and later validated on node-positive disease (see Additional file 1: Table S1 for details).
The EP signature [32] was originally designed for ER-positive/HER2-negative breast cancer patients for predicting distant recurrence. In this study, it showed significant prognostic power on the complete dataset, ER-positive/HER2-negative treated and untreated subgroups (Figure eight of Additional file 2).

Computing original gene signature scores

Affymetrix probes were matched against the genes of the signatures (Table 1). Risk scores were then generated using the original algorithms of the signatures and recalibrated on the studied dataset for risk-group assignments.
For Intrinsic and PAM50, subtype classification was performed based on the nearest of the five centroids (distances calculated using correlation to the centroids). Risk score per sample was computed by linear combination of the centroid correlations in ROR-S model (Risk-Of-Relapse scores by Subtype alone) [9]. A pseudo Oncotype DX® Recurrence Score per patient was computed by the unscaled Recurrence Score [12]. Similarly, a pseudo EP Score per patient was obtained by the unscaled risk score for EP [32]. For 76-genes, GGI, RS and EP, rather than assigning risk groups based on published cutoffs, we used a population-based approach in which a fixed proportion of the population was assigned to each risk group. The proportions were derived from previous datasets associated with individual signatures [7, 12, 26, 40]. We found this necessary as our analyses differed from the original methods in technical or methodological manners (Supplement).

Survival analysis

Distant Metastasis Free Survival (DMFS: n = 912) is used as clinical endpoint. Follow-up time was defined as time from diagnosis until distant metastasis, or time of last follow-up if the patient is not known to have distant metastasis. It was noted that DMFS in the Pawitan set [35] was defined as distant metastasis or death, whichever occurs first. Since this only consists of a small portion of the studied cohort, it is unlikely to bias or confound our results.
Continuous risk scores from the original signatures were used instead of categorized risk-groups. For Intrinsic and PAM50, the ROR-S scores were used. For 70-gene, the centroid correlations were reversed to represent the risk.
The concordance index [41] (C-index, an analogy to area under ROC curve) was chosen to compare the predictive strength of the signatures. The contribution of a signature predictor in the univariate setting was evaluated using the proportion of variation explained in the outcome variable (PVE) [42].
Univariate Cox models were fitted for each risk signature. Assessment of the proportional hazard assumption by different methods [4345] indicated clear time-dependencies in the predictive power of the risk signatures and was used to identify suitable time intervals for separate Cox analyses. Standardized hazard ratios (HR) indicate the relative risk associated with a one-standard-deviation increase in the risk score.
Effects of common prognostic factors: tumor size (pT1, pT2 and pT3-pT4), node status (positive versus negative) and histological grade (I-III) were investigated using multivariate Cox models.

METABRIC data

METABRIC [46] expression discovery set (n = 996) was used. Gene annotations on the original IlluminaHT12v3 probes were retrieved using BioMart through R library biomaRt (Ensembl release 68, HG19 human assembly). Disease-specific survival was used as endpoint. Follow-up time was defined as time from diagnosis until death, or time of last follow-up if the patient is not known to have died. Data is available through European Genome-Phenome Archive (http://​www.​ebi.​ac.​uk/​ega/​), under accession number EGAS00000000083.

Results

Subtype signatures comparison

We compared the subtype classification between Intrinsic and PAM50 on the full dataset (n = 947). Overall, their subtype assignments were moderately concordant (Cohen’s kappa [47]κ = 0 · 54). Noticeably, nearly half of the Intrinsic LumA tumors were assigned as LumB by PAM50 (40.7%), while the two signatures appeared to highly agree on classification of basal-like tumors (86.5%; Additional file 5: Figure S1A). Indeed, basal-like was the most concordant subtype with a Pearson correlation of 0 · 94 between Intrinsic and PAM50 (Additional file 5: Figure S1B), followed by normal-like (0.85), LumA (0.68), LumB (0.55) and Her2-enriched (0.42). More specifically, basal-like was the most distinctly classified subtype across these two signatures (Additional file 5: Figure S1C) with disagreement limited to a few borderline classifications. Furthermore, agreements between the subtypes and their immunohistochemistry receptor status counterparts were similar for both signatures. A majority of the 709 IHC ER-positive samples were classified as Luminal tumors (62% for Intrinsic and 64% for PAM50), and half of the IHC HER2-positive samples were classified as HER2-enriched subtype (55% for Intrinsic and 50% for PAM50). The overlap between Basal-like tumors and triple-negative samples was 79% for both PAM50 and Intrinsic (Figure two and three of Additional file 2).
One property that distinguishes these signatures is that proliferation-associated genes were intentionally added when developing PAM50. This may partially explain the disconcordance between PAM50 and Intrinsic in their LumA and LumB classifications. Both signatures were kept for further analysis.

Similarity for risk assessment among gene signatures

The Pearson correlations of the continuous risk scores from individual signatures were generally high (Figure 1A; Additional file 1: Table S3). The correlations were above 0.4, except those involving Hypoxia and between 76-gene and Intrinsic (ρ=0.23), indicating reasonably good concordance across the signatures. The highest correlations were between GGI and PAM50 (0.9), followed by GGI with WR (0.87) and Intrinsic with RS (0.81). Intrinsic and PAM50 ROR-S scores correlated well (ρ=0.61). The Hypoxia signature was negatively associated with the 76-gene classifier (ρ= - 0.02), and 76-gene was also less in agreement with other signatures: correlation with Intrinsic (0.23), 70-gene (0.4) and RS (0.44). Thus, Hypoxia and 76-gene appear distinct from the other signatures.

Comparison of performances of gene signatures for survival prediction

For all signatures except Hypoxia, differences in DMFS between risk groups were highly significant (n = 912; Figure 1B).
Using the continuous risk scores to predict DMFS, PAM50 had the highest C-index of 0.658 with 95% CI [0.64–0.68] (Table 2), followed by GGI (0.656), WR (0.651), RS (0.648), 76-gene (0.642), 70-gene (0.612), Intrinsic (0.598) and Hypoxia (0.525). All signatures received a C-index exceeding the threshold 0.5 for random prediction. The importance of individual signatures in univariate setting as measured by PVE (Table 2) ranked PAM50 (5.74%), GGI (4.87%) and WR (4.83%) as the top three predictors for DMFS, while Hypoxia explained the lowest portion of variation (0.6%). The rankings by C-index and PVE were fairly similar.
Table 2
Assessment of univariate performance of individual gene signatures on Distant Metastasis Free Survival prediction
 
Ca(95% CI)
PVEb(%)
Intrinsic-RORs
0.598 [0.58, 0.62]
1.60
PAM50-RORs
0.658 [0.64, 0.68]
5.74
70-gene
0.612 [0.60, 0.63]
2.76
76-gene
0.642 [0.62, 0.66]
4.58
GGI
0.656 [0.64, 0.67]
4.87
WR
0.651 [0.63, 0.67]
4.83
Hypoxia
0.525 [0.50, 0.55]
0.60
RS
0.648 [0.63, 0.67]
4.05
EP
0.648 [0.63, 0.67]
4.78
aC: concordance index.
b PVE: proportion of variation explained in the outcome variable, comparable with the R 2 in regression modeling.
The variability of the C-index was estimated from 1000 bootstrap iterations.

Time- & ER-dependency of gene signatures for DMFS prediction

The assumption of time-independent proportional hazard was examined for ER-positive group and ER-negative group separately using a univariate Cox model with signature risk scores as covariate. Time-dependency was clearly visible for most of the signatures (Additional file 5: Figure S2A-B; Table 3). In general, signatures seemed to lose their predictive power over time for forecasting DMFS.
Table 3
Time- & ER-dependent effect assessment of individual gene signatures in predicting Distant Metastasis Free Survival (DMFS)
     
ER+ (n= 692)
    
ER– (n= 220)
 
Gene signature
PHaρ (p)
Time
n risk
n event
HR [95% CI]
p
PHaρ (p)
n risk
n event
HR [95% CI]
p
Intrinsic-RORs
-0.23(0.0032)
     
-0.38(0.0069)
    
  
0-5 yr
692
126
1.46 [1.24-1.72]
<0.0001
 
220
61
1.24 [0.94-1.63]
0.1230
  
5-10 yr
566
41
0.83 [0.59-1.15]
0.2576
 
159
7
0.95 [0.49-1.85]
0.8892
  
>10 yr
525
10
0.76 [0.36-1.60]
0.4650
 
152
5
0.49 [0.25-0.98]
0.0440
PAM50-RORs
-0.27(0.0005)
     
-0.13(0.3676)
    
  
0-5 yr
692
126
2.16 [1.78-2.61]
<0.0001
 
220
61
1.11 [0.86-1.44]
0.4312
  
5-10 yr
566
41
1.26 [0.93-1.70]
0.1383
 
159
7
1.6 [0.64-4.03]
0.3161
  
>10 yr
525
10
0.91 [0.48-1.73]
0.7773
 
152
5
0.54 [0.27-1.09]
0.0835
70-gene
-0.18(0.0232)
     
-0.24(0.0702)
    
  
0-5 yr
692
126
1.70 [1.43-2.03]
<0.0001
 
220
61
1.00 [0.79-1.28]
0.9739
  
5-10 yr
566
41
1.16 [0.87-1.55]
0.3054
 
159
7
0.84 [0.43-1.65]
0.6189
  
>10 yr
525
10
1.06 [0.57-1.96]
0.8468
 
152
5
0.40 [0.21-0.78]
0.0066
76-gene
-0.20(0.0092)
     
-0.26(0.0565)
    
  
0-5 yr
692
126
1.83 [1.55-2.17]
<0.0001
 
220
61
1.30 [1.04-1.62]
0.0228
  
5-10 yr
566
41
1.32 [0.99-1.76]
0.0551
 
159
7
1.12 [0.51-2.48]
0.7745
  
>10 yr
525
10
0.58 [0.28-1.20]
0.1446
 
152
5
0.38 [0.09-1.57]
0.1828
GGI
-0.32(<0.0001)
     
-0.29(0.0296)
    
  
0-5 yr
692
126
2.11 [1.77-2.52]
<0.0001
 
220
61
1.07 [0.82-1.38]
0.6288
  
5-10 yr
566
41
1.23 [0.91-1.66]
0.1735
 
159
7
1.22 [0.55-2.71]
0.6170
  
>10 yr
525
10
0.73 [0.37-1.44]
0.3592
 
152
5
0.39 [0.18-0.84]
0.0165
WR
-0.28(0.0001)
     
-0.43(0.0002)
    
  
0-5 yr
692
126
2.07 [1.72-2.48]
<0.0001
 
220
61
1.39 [1.05-1.85]
0.0214
  
5-10 yr
566
41
1.16 [0.86-1.56]
0.3341
 
159
7
0.80 [0.39-1.63]
0.5334
  
>10 yr
525
10
0.63 [0.33-1.20]
0.1612
 
152
5
0.30 [0.12-0.75]
0.0098
Hypoxia
-0.02(0.7717)
     
-0.04(0.7132)
    
  
0-5 yr
692
126
1.06 [0.88-1.26]
0.5483
 
220
61
1.50 [1.20-1.89]
<0.0001
  
5-10 yr
566
41
0.93 [0.68-1.27]
0.6427
 
159
7
1.41 [0.70-2.83]
0.3389
  
>10 yr
525
10
1.10 [0.57-2.10]
0.7773
 
152
5
0.48 [0.17-1.35]
0.1645
RS
-0.25(0.0019)
     
-0.23(0.0628)
    
  
0-5 yr
692
126
1.79 [1.55-2.07]
<0.0001
 
220
61
1.19 [0.92-1.53]
0.1919
  
5-10 yr
566
41
1.06 [0.78-1.43]
0.7311
 
159
7
0.78 [0.39-1.58]
0.4902
  
>10 yr
525
10
0.65 [0.26-1.61]
0.3535
 
152
5
0.57 [0.25-1.30]
0.1798
EP
-0.27(0.0004)
     
-0.18(0.2457)
    
  
0-5 yr
692
126
1.97 [1.66-2.33]
<0.0001
 
220
61
1.11 [0.86-1.45]
0.4199
  
5-10 yr
566
41
1.13 [0.83-1.53]
0.4393
 
159
7
1.04 [0.50-2.15]
0.9198
  
>10 yr
525
10
1.02 [0.55-1.91]
0.9462
 
152
5
0.51 [0.25-1.04]
0.0628
a PH test for time trend: scaled Schoenfeld residuals were tested against transformed time (Kaplan-Meier estimates) for violation of proportional hazard assumption in a univariate Cox model for individual gene signatures. P values are shown.
Analysis was carried out on ER + group and ER- group separately. Preliminary test for time trend was performed by checking proportional hazard assumption in a Cox model§ per signature fitted on all tumors with follow-up time and event status available (column “PH”: correlation and asscoaited p value are reported)). Main effect associated with a signature for DMFS prediction in a certain follow-up time interval was estimated by a Cox model within each ER stratification. The Hazard Ratio (HR) along with its 95% confidence interval and the p value from the Wald test are shown. Numbers of patients at risk (n risk ) were computed at time point 0, 5 and 10 year, respectively.
To investigate the nature of time-dependency in ER-positive tumors, we inspected the cumulative regression plots of the estimate along with 95% confidence intervals from a univariate additive regression model (Additional file 5: Figure S2C). The estimated curve in each plot reflects the cumulative effect of a signature covariate on survival over time, and a time-independent effect should therefore result in a curve with a constant slope. Hypoxia did not seem to have an effect on DMFS prediction. For all the other signatures, there were significant and strong initial positive effects up to around 5 years; these effects tended to disappear after about 10 years. However, the estimates are uncertain towards the end of the time span as few patients remain in the risk set. In ER-negative breast cancers (Additional file 5: Figure S2D), while similar time-dependency was evident for individual signatures, the effects on DMFS predictions were less substantial than in the ER-positive subset, and rather uncertain for most of the signatures. Contrary to its non-predictive behavior in the ER-positive group, Hypoxia predicted DMFS (higher hypoxic scores associated with a shorter survival time) for ER-negative cancers. In addition, WR, 76-gene and Intrinsic also potentially have predictive effect in the early follow-up period.
Based on these results, we divided follow-up time into three intervals: first 5 years, 5–10 years, and beyond 10 years. Patients experiencing an event before the start of the interval were excluded, while those that remained at risk at the end of the time interval were censored. For each time interval, univariate Cox models for each signature were fitted in ER-positive and ER-negative tumors separately. The estimated HRs with 95% confidence interval per time interval and ER status are shown for each signature (Figure 2; Table 3). The HRs were systematically higher at earlier time points and decayed with time; predictions were generally stronger in the ER-positive group than in the ER-negative group. Within the first 5 years, all signatures except for Hypoxia had significant positive effects (p < 0.0001) in the ER-positive group; while in the ER-negative group, Hypoxia (p < 0.0001), WR (p = 0.021) and 76-gene (p = 0.023) were the only classifiers with significant positive effects on DMFS prediction. We observed borderline significant protective effects (HR < 1: higher risk scores had lower risks for distant metastasis) within the last time interval (>10 years) in the ER-negative group for Intrinsic (p = 0.044), 70-gene (p = 0.007), GGI (p = 0.017) and WR (p = 0.01).

Possible effects of cohort differences

Since cohort differences could potentially lead to spurious effects, we ran survival analyses adjusted for cohort differences. However, as cohort differences did not contribute significantly to the models (Figure one and Box five of Additional file 2), it seems unlikely that cohort differences may have biased the results.

Analysis on a systemically untreated subpopulation

To avoid bias introduced by adjuvant treatment, the same analyses were performed on patients that were only treated with surgery with/without radiotherapy (n = 395). Similar indications related to follow-up time and ER status for signatures predicting DMFS hold in this subgroup of patients (Additional file 5: Figure S3), indicating that treatment alone does not explain the effects described above.
Analyses of systemically treated patients confirmed the predictive power of the signatures during the first 5 years of follow-up in the ER-positive group, but had too few events after 5 years for any reliable assessment of time-dependency.

Multivariate analysis on signatures with known prognostic parameters

Node status, tumor size and histological grade all significantly predict DMFS on the complete dataset (n = 912; Additional file 5: Figure S4A). A multivariate Cox model was fitted with node, size, histological grade and individual signatures for the two ER groups separately. In the ER-positive group (Additional file 1: Table S4A), with the exception of Hypoxia (p = 0.7351), signatures remain significant with the presence of size, node and histological grade (Model 1: p < 0.0001 except Intrinsic p = 0.0397). Inclusion of tumor size in the model removed the time trends associated with the signatures (Model 2). The prognostic power of the included predictors were dismal for ER-negative tumors (Additional file 1: Table S4B),

Analysis on prognosis of gene signatures associated with HER2 status for DMFS prediction

We investigated the performance of gene signatures in relation to HER2 status. We observe a decreasing time dependency associated with the prognostic power in the HER2-negative group (Additional file 5: Figure S5A). Due to limited number of events in the 5–10 year followup interval, we cannot draw conclusions about the time trend in the HER2-positive group and the differences in prognostic power between the two HER2 groups (Additional file 5: Figure S5A).
The analysis on groups defined by both HER2 status and ER status revealed a decreasing time trend for the signature’s prognostic power for both the HER2-/ER + and HER2-/ER- groups (Additional file 5: Figure S5B), where at least two events are presented for each time intervals. And HER2-/ER + is generally better than HER2-/ER- in term of prognostic power. This can be largely explained by the ER stratification.

Validation on METABRIC data

We observed similar ER-dependency and similar pattern of gene signatures for the long-term prognosis on the METABRIC complete set, systemically untreated set as well as on the systemically treated set (Additional file 5: Figure S6 & Additional file 1: Table S7). Similarly, including histological grade and tumor size seems to reduce the strength of the time dependency of the signatures (Additional file 1: Table S8).

Discussion

Applicability of individual gene signatures

Growing evidence suggests that expression-based gene signatures are of clinical relevance, especially for identifying patients at high risk of early distant metastasis. One important challenge is to robustly identify patients with low risk, thereby reducing the number of patients receiving cytotoxic treatment. Translating signatures to a new dataset is complicated by differences in microarray platforms and data processing procedures, as well as the clinical differences between cohorts.
Methods based on centroid correlations (e.g. subtype signatures, 70-gene and WR) and methods that transform the data into an invariant scale before computing the risk scores (e.g. GGI) have more consistent performances across different studies. We suspect that summarizing gene expression patterns through weighted averages (e.g. 76-gene, RS, Hypoxia) is more sensitive to data scales and missing gene information. Different normalization procedure from the original study [6] may explain why the original 76-gene signature, prior to the population-based recentering, did not predict any good prognosis in our data. Generally, when the distribution of risk scores depends on platform and normalization procedure, cutoffs for risk group assignment need to be recalibrated. The population-based strategy is more general and applicable for a study with a pure prognostic purpose, but requires the tumors to be representative of the population of breast cancer.

Time- and ER-dependency of prognostic gene signatures

Prognostication by gene-expression signatures seemed harder for ER-negative than for ER-positive tumors. It should be noted that most of the signatures have been trained on populations containing a majority of ER-positive tumors. All studied signatures except for Hypoxia showed prognostic power in assessing DMFS in ER-positive breast cancer in the first few years after diagnosis. Only the 76-gene, Wound-Response and Hypoxia signatures were prognostic in the ER-negative group within the first five years. The time-dependent prognostic effect was previously reported for the 76-gene [26] and RS [31].
Most of the signatures were tightly correlated. We believe this may be due to common underlying biological processes. Studies [16, 27, 4850] suggest that cell proliferation is a common characteristic among many signatures (e.g. 76-gene, 70-gene, RS, GGI, PAM50, WR). If the proliferation module drives prognostication in ER-positive tumors, the risk-group separation will be highly comparable to the classification of LumA and LumB tumors within the ER-positive subgroup, as LumB tumors are characterized by higher proliferation. This seemed to be the case for the majority of the signatures (Figure 3). Different signatures essentially detect the low-proliferation subset as low-risk in the ER-positive group [27, 48, 49]. Furthermore, histological grade, which strongly reflects proliferation, shows prognostic value only in the ER-positive subgroup (Additional file 5: Figure S4B; ER + p = 0.0002 vs ER- p = 0.57). This highlights the need for robust prognostic tools designed for each ER subgroup.
The dismal performances in ER-negative tumors of most of the signatures, except 76-gene and Hypoxia, resulted from classifying most of them into the high-risk category [48, 51]. This elevated risk score was predominantly driven by highly proliferative basal-like and Her2-enriched tumors (Figure 3), and left the signatures with poor discriminative power for risk assessment within ER-negative tumors. Clinically, patients with ER-negative tumors are heterogeneous with respect to age as well as treatment received. Most patients with ER-negative tumors receive cytotoxic chemotherapy. All these factors pose difficulties in marker identification and further building prognostic/predictive signatures specific for this subgroup. The ER-specific markers within the 76-gene signature (60 genes from ER + and 16 from ER-) contribute to its prognostic ability in both ER stratifications. Intriguingly, signatures characterizing tumor microenvironment (Hypoxia and Wound-Response) showed prognostic values for ER-negative breast cancer. In line with previous indications [18, 30], Hypoxia seems to carry biological and prognostic information distinct from the other signatures (Figure 1A). More specifically, certain genetic components and the microenvironment of breast tumors are likely to be important for the predictive ability of the Hypoxia signature. Tumors with “high hypoxia response” were more likely to have TP53 mutations and to be ER negative [30]. In this study, TP53-mutated and ER-negative tumors had elevated hypoxic score (one-tailed t-test p = 0.029; Additional file 5: Figure S4C), while no significant differences in the hypoxic score associated with TP53 status were found in the ER-positive tumors (one-tailed t-test p = 0.29). Distinct features of the tumor microenvironment associated with basal-like and luminal tumors [52] possibly underlie the variation in hypoxia responses observed in different ER subgroups.
Proliferation seems to be the common driving force for prognostication in ER-positive breast cancers, while different biological mechanisms such as stress response may be crucial for risk stratification in ER-negative tumors. Additionally, immune-related gene modules have been implicated to be prognostic in high-risk ER-positive breast cancers [53] and ER-negative breast cancers [54, 55].
In most gene expression studies, information on patient treatment is limited and inconsistent. In our combined cohort, treatment data were compiled for systemic adjuvant treatment. Results for patients that did not receive systemic treatment (Additional file 5: Figure S3A-C) were consistent with the main findings. Data on patient cohorts homogeneously treated is important to be able to distinguish between ability to predict treatment response to a specific therapy and prediction of prognosis.
In multivariate analyses on the ER-positive tumors (Additional file 1: Table S4A), signatures remained powerful predictors and added significant information beyond known prognostic parameters, including tumor size, node and histological grade. Histological grade lost much of its prognostic power in models with signature, size and node (Model 1). The signatures’ change in prognostic power over time fell or disappeared in models that included histological grade (Model 3) or tumor size (Model 2). More advanced tumors, grade-3 or large tumor size, tended to experience early relapse, with late relapse more common in less advanced tumors (grade-1 or small tumor size). The inclusion of histological grade or tumor size in the model may thus have captured and masked some of the time-dependency of the signatures’ prognostic power (see Additional file 1: Table S5, Additional file 1: Table S6 and Additional file 5: Figure S4D-E for more detail), although it also indicates that signatures may provide more accurate long-term prognosis when combined with information on histological grade or tumor size. Multivariate analyses on the ER-negative group were not presented because none of the included predicators was significant.
We did not find any notable effect of cohort differences on our analyses (Figure one and Box five of Additional file 2).
The METABRIC set [46] served as an independent validation set for our study. We did not have access to this data until after the original analyses had been performed. The fact that we are able to confirm the observations from the original analyses (based on the meta-cohort; n = 947) in an independent large dataset, undoubtedly validates our study, greatly strengthens the indications and authenticates the conclusions. These findings were confirmed in both the systemically treated and untreated groups, and thus does not seem to be affected by the use of breast cancer specific survival as event instead of DMFS. We did not include the classification for molecular subtype proposed by Curtis et al. [46] as the IC (Integrative Cluster) subgroups are based on clustering on both gene expression and copy number data through a joint latent model [56]. The majority of samples in our main analysis did not have copy number data available, while evaluating the ICs in METABRIC together with other signatures would bias the results since the IC classification was developed using this cohort.
The indications from our study that prognostic power of gene signatures depend on ER-status, has previously been reported by Desmedt et al. [49]. They used a gene module score to estimate HER2 and ER activity, and used this to split the samples by HER2 status, and the HER2-negative were further split by ER status, resulting in three groups. We used ER and HER2 status based on IHC where available, or imputed from gene expression if not. Since we did not see a substantial effect of HER2 status on DMFS or time dependency (Additional file 5: Figure S5), we did not focus on stratification based on the HER2 status.
By the rule of thumb that 10 events per covariate is generally sufficient for Cox analyses [5760], we consider the sample size and number of events sufficient to reliably assess the prognostic power of gene signatures in the different follow-up time intervals for both ER states (Table 3 & Additional file 1: Table S7A), although with some reservations for the last time interval (>10 years) for the ER-negative group which had few events (n = 5 in both studied datasets). However, we consider our results convincing given the consistency across the two datasets and across several signatures.
It is interesting to observe that higher risk scores associated with lower risks for distant metastasis after >10 years follow-up in the ER-negative group. These estimates are based on a small number of events (n = 5 in both datasets), but the fact that it occurs in both datasets lends the finding some credibility. For ER-negative cases still under study after 10 years, high risk signatures tended to correspond to higher histological grade and HER2 positive status.

Compatibility between signatures and target cohort

Some of the signatures had been developed on specific patient subgroups (Additional file 1: Table S1). In particular, several of them were developed on node-negative disease and only later validated on node-positive disease. Signatures developed on one patient subgroup, may be expected to have reduced power on other patient subgroups despite later validation, and so the use of a signature from one patient group extended to a larger group should be done and interpreted with caution.
Specifically in our study, the 76-gene signature is intended for lymph-node-negative cancers. However, since it was predictive for the node-positive patients as well (p = 0.005 for raw relapse scores predicting DMFS, see Supplement), we judged that the 76-gene signature was a valid predictor also for node-positive disease, and could be assessed along with the other signatures without substantial loss of predictive power.
Although the RS was used as a prognostic test in the tamoxifen-treated breast cancers, we found that RS had significant prognostic power for the ER-positive patients in the untreated cohort as well (Additional file 5: Figure S3C). The RS signature was only intended for ER-positive, and so cannot be criticized for performing badly on ER-negative. Indeed, it performed no worse than many of the other signatures, which were intended to cover ER-negative disease.
The EP signature was designed as a prognostic test in ER-positive, HER2-negative breast cancer patients treated with adjuvant endocrine therapy only. We found that the EP had significant prognostic power on the ER-positive, HER2-negative, untreated cancers, as well as the complete set (Supplement). As the treatment information in our main analysis is limited to systemic treatment, the stratified subset for EP is not strictly based on adjuvant endocrine therapy only.

Conclusions

In summary, our study highlights conditions under which it is appropriate to use individual published gene signatures for survival prediction. The distinctions in prognostic behavior of the signatures with respect to ER status suggest that different molecular mechanisms are involved in risk stratifications within each ER stratum. Also, the signatures were primarily able to predict relapse with the first 5 years of follow-up, with little ability to predict later relapses. Incorporating characteristics of the advancement of the tumor might help improve the quality of the prognosis, perhaps also with respect to long-term prognosis. While the majority of the tested signatures are strong risk predictors in the early follow-up time intervals for ER-positive tumors, there are urgent needs to improve risk stratifications for long-term prognosis and ER-negative breast cancers.

Author’s contributions

Conceived of and designed the study: XZ EAR ALBD OCL. Organized and analyzed the data: XZ. Interpreted the results: XZ EAR TS HKMV HGR ALBD OCL. Wrote the paper: XZ EAR TS. Contributed to manuscript and discussions: HKMV HGR VNK OCL ALBD. All authors read and approved the final manuscript.

Grant support

This study was supported by Norwegian Research Council (NFR) FUGE program, Grant 175240 and NFR Cancer program, Grant 193387. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​2.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited.

Competing interests

The authors declare that they have no competing interests.
Literatur
1.
Zurück zum Zitat Perou CM, Sørlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, Ross DT, Johnsen H, Akslen LA, Fluge O, Pergamenschikov A, Williams C, Zhu SX, Lønning PE, Børresen-Dale AL, Brown PO, Botstein D: Molecular portraits of human breast tumours. Nature. 2000, 406 (6797): 747-752. 10.1038/35021093.CrossRefPubMed Perou CM, Sørlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, Ross DT, Johnsen H, Akslen LA, Fluge O, Pergamenschikov A, Williams C, Zhu SX, Lønning PE, Børresen-Dale AL, Brown PO, Botstein D: Molecular portraits of human breast tumours. Nature. 2000, 406 (6797): 747-752. 10.1038/35021093.CrossRefPubMed
2.
Zurück zum Zitat Sørlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, Hastie T, Eisen MB, van de Rijn M, Jeffrey SS, Thorsen T, Quist H, Matese JC, Brown PO, Botstein D, Lønning PE, Børresen-Dale AL: Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci USA. 2001, 98 (19): 10869-10874. 10.1073/pnas.191367098.CrossRefPubMedPubMedCentral Sørlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, Hastie T, Eisen MB, van de Rijn M, Jeffrey SS, Thorsen T, Quist H, Matese JC, Brown PO, Botstein D, Lønning PE, Børresen-Dale AL: Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci USA. 2001, 98 (19): 10869-10874. 10.1073/pnas.191367098.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Sørlie T, Tibshirani R, Parker J, Hastie T, Marron JS, Nobel A, Deng S, Johnsen H, Pesich R, Geisler S, Demeter J, Perou CM, Lønning PE, Brown PO, Børresen-Dale AL, Botstein D: Repeated observation of breast tumor subtypes in independent gene expression data sets. Proc Natl Acad Sci USA. 2003, 100 (14): 8418-8423. 10.1073/pnas.0932692100.CrossRefPubMedPubMedCentral Sørlie T, Tibshirani R, Parker J, Hastie T, Marron JS, Nobel A, Deng S, Johnsen H, Pesich R, Geisler S, Demeter J, Perou CM, Lønning PE, Brown PO, Børresen-Dale AL, Botstein D: Repeated observation of breast tumor subtypes in independent gene expression data sets. Proc Natl Acad Sci USA. 2003, 100 (14): 8418-8423. 10.1073/pnas.0932692100.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Veer LJ V‘t, Dai H, van de Vijver MJ, He YD, Hart AA, Mao M, Peterse HL, Van der Kooy K, Marton MJ, Witteveen AT, Schreiber GJ, Kerkhoven RM, Roberts C, Linsley PS, Bernards R, Friend SH: Gene expression profiling predicts clinical outcome of breast cancer. Nature. 2002, 415 (6871): 530-536. 10.1038/415530a.CrossRef Veer LJ V‘t, Dai H, van de Vijver MJ, He YD, Hart AA, Mao M, Peterse HL, Van der Kooy K, Marton MJ, Witteveen AT, Schreiber GJ, Kerkhoven RM, Roberts C, Linsley PS, Bernards R, Friend SH: Gene expression profiling predicts clinical outcome of breast cancer. Nature. 2002, 415 (6871): 530-536. 10.1038/415530a.CrossRef
5.
Zurück zum Zitat van de Vijver MJ, He YD, Van’t Veer LJ, Dai H, Hart AA, Voskuil DW, Schreiber GJ, Peterse JL, Roberts C, Marton MJ, Parrish M, Atsma D, Witteveen A, Glas A, Delahaye L, van der Velde T, Bartelink H, Rodenhuis S, Rutgers ET, Friend SH, Bernards R: A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med. 2002, 347 (25): 1999-2009. 10.1056/NEJMoa021967.CrossRefPubMed van de Vijver MJ, He YD, Van’t Veer LJ, Dai H, Hart AA, Voskuil DW, Schreiber GJ, Peterse JL, Roberts C, Marton MJ, Parrish M, Atsma D, Witteveen A, Glas A, Delahaye L, van der Velde T, Bartelink H, Rodenhuis S, Rutgers ET, Friend SH, Bernards R: A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med. 2002, 347 (25): 1999-2009. 10.1056/NEJMoa021967.CrossRefPubMed
6.
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, Jatkoe T, Berns EM, Atkins D, Foekens JA: Gene-expression profiles to predict distant metastasis of lymph-node-negative primary breast cancer. Lancet. 2005, 365 (9460): 671-679. 10.1016/S0140-6736(05)17947-1.CrossRefPubMed Wang Y, Klijn JG, Zhang Y, Sieuwerts AM, Look MP, Yang F, Talantov D, Timmermans M, Meijer-van Gelder ME, Yu J, Jatkoe T, Berns EM, Atkins D, Foekens JA: Gene-expression profiles to predict distant metastasis of lymph-node-negative primary breast cancer. Lancet. 2005, 365 (9460): 671-679. 10.1016/S0140-6736(05)17947-1.CrossRefPubMed
7.
Zurück zum Zitat Sotiriou C, Wirapati P, Loi S, Harris A, Fox S, Smeds J, Nordgren H, Farmer P, Praz V, Haibe-Kains B, Desmedt C, Larsimont D, Cardoso F, Peterse H, Nuyten D, Buyse M, Van de Vijver MJ, Bergh J, Piccart M, Delorenzi M: Gene expression profiling in breast cancer: understanding the molecular basis of histologic grade to improve prognosis. J Natl Cancer Inst. 2006, 98 (4): 262-272. 10.1093/jnci/djj052.CrossRefPubMed Sotiriou C, Wirapati P, Loi S, Harris A, Fox S, Smeds J, Nordgren H, Farmer P, Praz V, Haibe-Kains B, Desmedt C, Larsimont D, Cardoso F, Peterse H, Nuyten D, Buyse M, Van de Vijver MJ, Bergh J, Piccart M, Delorenzi M: Gene expression profiling in breast cancer: understanding the molecular basis of histologic grade to improve prognosis. J Natl Cancer Inst. 2006, 98 (4): 262-272. 10.1093/jnci/djj052.CrossRefPubMed
8.
Zurück zum Zitat Langerød A, Zhao H, Borgan O, Nesland J, Bukholm I, Ikdahl T, Karesen R, Borresen-Dale A, Jeffrey S: TP53 mutation status and gene expression profiles are powerful prognostic markers of breast cancer. Breast Cancer Res. 2007, 9 (3): R30-10.1186/bcr1675.CrossRefPubMedPubMedCentral Langerød A, Zhao H, Borgan O, Nesland J, Bukholm I, Ikdahl T, Karesen R, Borresen-Dale A, Jeffrey S: TP53 mutation status and gene expression profiles are powerful prognostic markers of breast cancer. Breast Cancer Res. 2007, 9 (3): R30-10.1186/bcr1675.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Parker JS, Mullins M, Cheang MCU, Leung S, Voduc D, Vickery T, Davies S, Fauron C, He X, Hu Z: Supervised risk predictor of breast cancer based on intrinsic subtypes. J Clin Oncol. 2009, 27 (8): 1160-10.1200/JCO.2008.18.1370.CrossRefPubMedPubMedCentral Parker JS, Mullins M, Cheang MCU, Leung S, Voduc D, Vickery T, Davies S, Fauron C, He X, Hu Z: Supervised risk predictor of breast cancer based on intrinsic subtypes. J Clin Oncol. 2009, 27 (8): 1160-10.1200/JCO.2008.18.1370.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Cardoso F, Van’t Veer L: Clinical application of the 70-gene profile: the MINDACT trial. J Clin Oncol. 2008, 26 (5): 729-10.1200/JCO.2007.14.3222.CrossRefPubMed Cardoso F, Van’t Veer L: Clinical application of the 70-gene profile: the MINDACT trial. J Clin Oncol. 2008, 26 (5): 729-10.1200/JCO.2007.14.3222.CrossRefPubMed
11.
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 (5): 721-10.1200/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 (5): 721-10.1200/JCO.2007.15.1068.CrossRefPubMed
12.
Zurück zum Zitat Paik S, Shak S, Tang G, Kim C, Baker J, Cronin M, Baehner FL, Walker MG, Watson D, Park T, Hiller W, Fisher ER, Wickerham DL, Bryant J, Wolmark N: A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. N Engl J Med. 2004, 351 (27): 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, Hiller W, Fisher ER, Wickerham DL, Bryant J, Wolmark N: A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. N Engl J Med. 2004, 351 (27): 2817-2826. 10.1056/NEJMoa041588.CrossRefPubMed
13.
Zurück zum Zitat Koscielny S: Why Most Gene Expression Signatures of Tumors Have Not Been Useful in the Clinic. Sci Transl Med. 2010, 2 (14): 14ps2-CrossRefPubMed Koscielny S: Why Most Gene Expression Signatures of Tumors Have Not Been Useful in the Clinic. Sci Transl Med. 2010, 2 (14): 14ps2-CrossRefPubMed
14.
Zurück zum Zitat Fan C, Oh DS, Wessels L, Weigelt B, Nuyten DS, Nobel AB, Van’t Veer LJ, Perou CM: Concordance among gene-expression-based predictors for breast cancer. N Engl J Med. 2006, 355 (6): 560-569. 10.1056/NEJMoa052933.CrossRefPubMed Fan C, Oh DS, Wessels L, Weigelt B, Nuyten DS, Nobel AB, Van’t Veer LJ, Perou CM: Concordance among gene-expression-based predictors for breast cancer. N Engl J Med. 2006, 355 (6): 560-569. 10.1056/NEJMoa052933.CrossRefPubMed
15.
Zurück zum Zitat Nuyten DSA, Hastie T, Chi JTA, Chang HY, van de Vijver MJ: Combining biological gene expression signatures in predicting outcome in breast cancer: An alternative to supervised classification. Eur J Cancer. 2008, 44 (15): 2319-2329. 10.1016/j.ejca.2008.07.015.CrossRefPubMed Nuyten DSA, Hastie T, Chi JTA, Chang HY, van de Vijver MJ: Combining biological gene expression signatures in predicting outcome in breast cancer: An alternative to supervised classification. Eur J Cancer. 2008, 44 (15): 2319-2329. 10.1016/j.ejca.2008.07.015.CrossRefPubMed
16.
Zurück zum Zitat Reyal F, Van Vliet M, Armstrong N, Horlings H, De Visser K, Kok M, Teschendorff A, Mook S, Van’t Veer L, Caldas C: A comprehensive analysis of prognostic signatures reveals the high predictive capacity of the proliferation, immune response and RNA splicing modules in breast cancer. Breast Cancer Res. 2008, 10 (6): R93-10.1186/bcr2192.CrossRefPubMedPubMedCentral Reyal F, Van Vliet M, Armstrong N, Horlings H, De Visser K, Kok M, Teschendorff A, Mook S, Van’t Veer L, Caldas C: A comprehensive analysis of prognostic signatures reveals the high predictive capacity of the proliferation, immune response and RNA splicing modules in breast cancer. Breast Cancer Res. 2008, 10 (6): R93-10.1186/bcr2192.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Haibe-Kains B, Desmedt C, Piette F, Buyse M, Cardoso F, Van’t Veer L, Piccart M, Bontempi G, Sotiriou C: Comparison of prognostic gene expression signatures for breast cancer. BMC Genomics. 2008, 9 (1): 394-10.1186/1471-2164-9-394.CrossRefPubMedPubMedCentral Haibe-Kains B, Desmedt C, Piette F, Buyse M, Cardoso F, Van’t Veer L, Piccart M, Bontempi G, Sotiriou C: Comparison of prognostic gene expression signatures for breast cancer. BMC Genomics. 2008, 9 (1): 394-10.1186/1471-2164-9-394.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Zhao X, Rødland EA, Sørlie T, Naume B, Langerød A, Frigessi A, Kristensen VN, Børresen-Dale AL, Lingjærde OC: Combining Gene Signatures Improves Prediction of Breast Cancer Survival. PLoS One. 2011, 6 (3): e17845-10.1371/journal.pone.0017845.CrossRefPubMedPubMedCentral Zhao X, Rødland EA, Sørlie T, Naume B, Langerød A, Frigessi A, Kristensen VN, Børresen-Dale AL, Lingjærde OC: Combining Gene Signatures Improves Prediction of Breast Cancer Survival. PLoS One. 2011, 6 (3): e17845-10.1371/journal.pone.0017845.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Weigelt B, Mackay A, A’hern R, Natrajan R, Tan D, Dowsett M, Ashworth A, Reis-Filho JS: Breast cancer molecular profiling with single sample predictors: a retrospective analysis. Lancet Oncol. 2010, 11 (4): 339-10.1016/S1470-2045(10)70008-5.CrossRefPubMed Weigelt B, Mackay A, A’hern R, Natrajan R, Tan D, Dowsett M, Ashworth A, Reis-Filho JS: Breast cancer molecular profiling with single sample predictors: a retrospective analysis. Lancet Oncol. 2010, 11 (4): 339-10.1016/S1470-2045(10)70008-5.CrossRefPubMed
20.
Zurück zum Zitat Sørlie T, Borgan E, Myhre S, Vollan HK, Russnes H, Zhao X, Nilsen G, Lingjærde OC, Børresen-Dale AL, Rødland E: The importance of gene-centring microarray data. Lancet Oncol. 2010, 11 (8): 719-720. 10.1016/S1470-2045(10)70174-1.CrossRefPubMed Sørlie T, Borgan E, Myhre S, Vollan HK, Russnes H, Zhao X, Nilsen G, Lingjærde OC, Børresen-Dale AL, Rødland E: The importance of gene-centring microarray data. Lancet Oncol. 2010, 11 (8): 719-720. 10.1016/S1470-2045(10)70174-1.CrossRefPubMed
21.
Zurück zum Zitat Perou CM, Jeffrey SS, van de Rijn M, Rees CA, Eisen MB, Ross DT, Pergamenschikov A, Williams CF, Zhu SX, Lee JCF, Lashkari D, Shalon D, Brown PO, Botstein D, Lashkari D, Shalon D, Brown PO, Botstein D: Distinctive gene expression patterns in human mammary epithelial cells and breast cancers. Proc Natl Acad Sci. 1999, 96 (16): 9212-9217. 10.1073/pnas.96.16.9212.CrossRefPubMedPubMedCentral Perou CM, Jeffrey SS, van de Rijn M, Rees CA, Eisen MB, Ross DT, Pergamenschikov A, Williams CF, Zhu SX, Lee JCF, Lashkari D, Shalon D, Brown PO, Botstein D, Lashkari D, Shalon D, Brown PO, Botstein D: Distinctive gene expression patterns in human mammary epithelial cells and breast cancers. Proc Natl Acad Sci. 1999, 96 (16): 9212-9217. 10.1073/pnas.96.16.9212.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Mook S, Schmidt MK, Viale G, Pruneri G, Eekhout I, Floore A, Glas AM, Bogaerts J, Cardoso F, Piccart-Gebhart MJ, Rutgers ET, Van't Veer LJ, TRANSBIG Consortium: The 70-gene prognosis-signature predicts disease outcome in breast cancer patients with 1–3 positive lymph nodes in an independent validation study. Breast Cancer Res Treat. 2009, 116 (2): 295-302. 10.1007/s10549-008-0130-2. doi:10.1007/s10549-008-0130-2. Epub 2008 Jul 27CrossRefPubMed Mook S, Schmidt MK, Viale G, Pruneri G, Eekhout I, Floore A, Glas AM, Bogaerts J, Cardoso F, Piccart-Gebhart MJ, Rutgers ET, Van't Veer LJ, TRANSBIG Consortium: The 70-gene prognosis-signature predicts disease outcome in breast cancer patients with 1–3 positive lymph nodes in an independent validation study. Breast Cancer Res Treat. 2009, 116 (2): 295-302. 10.1007/s10549-008-0130-2. doi:10.1007/s10549-008-0130-2. Epub 2008 Jul 27CrossRefPubMed
23.
Zurück zum Zitat Buyse M, Loi S, Van’t Veer L, Viale G, Delorenzi M, Glas AM, Saghatchian A: Validation and clinical utility of a 70-gene prognostic signature for women with node-negative breast cancer. JNCI Cancer Spectr. 2006, 98 (17): 1183-CrossRef Buyse M, Loi S, Van’t Veer L, Viale G, Delorenzi M, Glas AM, Saghatchian A: Validation and clinical utility of a 70-gene prognostic signature for women with node-negative breast cancer. JNCI Cancer Spectr. 2006, 98 (17): 1183-CrossRef
24.
Zurück zum Zitat Espinosa E, Vara J, Redondo A, Sanchez J, Hardisson D, Zamora P, Pastrana FG, Cejas P, Martinez B, Suarez A: Breast cancer prognosis determined by gene expression profiling: a quantitative reverse transcriptase polymerase chain reaction study. J Clin Oncol. 2005, 23 (29): 7278-10.1200/JCO.2005.01.4746.CrossRefPubMed Espinosa E, Vara J, Redondo A, Sanchez J, Hardisson D, Zamora P, Pastrana FG, Cejas P, Martinez B, Suarez A: Breast cancer prognosis determined by gene expression profiling: a quantitative reverse transcriptase polymerase chain reaction study. J Clin Oncol. 2005, 23 (29): 7278-10.1200/JCO.2005.01.4746.CrossRefPubMed
25.
Zurück zum Zitat Foekens JA, Atkins D, Zhang Y, Sweep FCGJ, Harbeck N, Paradiso A, Cufer T, Sieuwerts AM, Talantov D, Span PN: Multicenter validation of a gene expression-based prognostic signature in lymph node-negative primary breast cancer. J Clin Oncol. 2006, 24 (11): 1665-10.1200/JCO.2005.03.9115.CrossRefPubMed Foekens JA, Atkins D, Zhang Y, Sweep FCGJ, Harbeck N, Paradiso A, Cufer T, Sieuwerts AM, Talantov D, Span PN: Multicenter validation of a gene expression-based prognostic signature in lymph node-negative primary breast cancer. J Clin Oncol. 2006, 24 (11): 1665-10.1200/JCO.2005.03.9115.CrossRefPubMed
26.
Zurück zum Zitat Desmedt C, Piette F, Loi S, Wang Y, Lallemand F, Haibe-Kains B, Viale G, Delorenzi M, Zhang Y, D’Assignies M: TRANSBIG Consortium. 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 (11): 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 M: TRANSBIG Consortium. 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 (11): 3207-3214. 10.1158/1078-0432.CCR-06-2765.CrossRefPubMed
27.
Zurück zum Zitat Loi S, Haibe-Kains B, Desmedt C, Lallemand F, Tutt AM, Gillet C, Ellis P, Harris A, Bergh J, Foekens JA: Definition of clinically distinct molecular subtypes in estrogen receptor-positive breast carcinomas through genomic grade. J Clin Oncol. 2007, 25 (10): 1239-10.1200/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: Definition of clinically distinct molecular subtypes in estrogen receptor-positive breast carcinomas through genomic grade. J Clin Oncol. 2007, 25 (10): 1239-10.1200/JCO.2006.07.1522.CrossRefPubMed
28.
Zurück zum Zitat Chang HY, Sneddon JB, Alizadeh AA, Sood R, West RB, Montgomery K, Chi JT, van de Rijn M, Botstein D, Brown PO: Gene expression signature of fibroblast serum response predicts human cancer progression: similarities between tumors and wounds. PLoS Biol. 2004, 2 (2): E7-10.1371/journal.pbio.0020007.CrossRefPubMedPubMedCentral Chang HY, Sneddon JB, Alizadeh AA, Sood R, West RB, Montgomery K, Chi JT, van de Rijn M, Botstein D, Brown PO: Gene expression signature of fibroblast serum response predicts human cancer progression: similarities between tumors and wounds. PLoS Biol. 2004, 2 (2): E7-10.1371/journal.pbio.0020007.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Chang HY, Nuyten DSA, Sneddon JB, Hastie T, Tibshirani R, Sørlie T, Dai H, He YD, Van’t Veer LJ, Bartelink H: Robustness, scalability, and integration of a wound-response gene expression signature in predicting breast cancer survival. Proc Natl Acad Sci USA. 2005, 102 (10): 3738-10.1073/pnas.0409462102.CrossRefPubMedPubMedCentral Chang HY, Nuyten DSA, Sneddon JB, Hastie T, Tibshirani R, Sørlie T, Dai H, He YD, Van’t Veer LJ, Bartelink H: Robustness, scalability, and integration of a wound-response gene expression signature in predicting breast cancer survival. Proc Natl Acad Sci USA. 2005, 102 (10): 3738-10.1073/pnas.0409462102.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Chi JT, Wang Z, Nuyten DS, Rodriguez EH, Schaner ME, Salim A, Wang Y, Kristensen GB, Helland A, Borresen-Dale AL, Giaccia A, Longaker MT, Hastie T, Yang GP, van de Vijver MJ, Brown PO: Gene expression programs in response to hypoxia: cell type specificity and prognostic significance in human cancers. PLoS Med. 2006, 3 (3): e47-10.1371/journal.pmed.0030047.CrossRefPubMedPubMedCentral Chi JT, Wang Z, Nuyten DS, Rodriguez EH, Schaner ME, Salim A, Wang Y, Kristensen GB, Helland A, Borresen-Dale AL, Giaccia A, Longaker MT, Hastie T, Yang GP, van de Vijver MJ, Brown PO: Gene expression programs in response to hypoxia: cell type specificity and prognostic significance in human cancers. PLoS Med. 2006, 3 (3): e47-10.1371/journal.pmed.0030047.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Albain KS, Barlow WE, Shak S, Hortobagyi GN, Livingston RB, Yeh I: Prognostic and predictive value of the 21-gene recurrence score assay in postmenopausal women with node-positive, oestrogen-receptor-positive breast cancer on chemotherapy: a retrospective analysis of a randomised trial. Lancet Oncol. 2010, 11 (1): 55-65. 10.1016/S1470-2045(09)70314-6.CrossRefPubMed Albain KS, Barlow WE, Shak S, Hortobagyi GN, Livingston RB, Yeh I: Prognostic and predictive value of the 21-gene recurrence score assay in postmenopausal women with node-positive, oestrogen-receptor-positive breast cancer on chemotherapy: a retrospective analysis of a randomised trial. Lancet Oncol. 2010, 11 (1): 55-65. 10.1016/S1470-2045(09)70314-6.CrossRefPubMed
32.
Zurück zum Zitat Filipits M, Rudas M, Jakesz R, Dubsky P, Fitzal F, Singer CF, Dietze O, Greil R, Jelen A, Sevelda P: A new molecular predictor of distant recurrence in ER-positive, HER2-negative breast cancer adds independent information to conventional clinical risk factors. Clin Cancer Res. 2011, 17 (18): 6012-6020. 10.1158/1078-0432.CCR-11-0926.CrossRefPubMed Filipits M, Rudas M, Jakesz R, Dubsky P, Fitzal F, Singer CF, Dietze O, Greil R, Jelen A, Sevelda P: A new molecular predictor of distant recurrence in ER-positive, HER2-negative breast cancer adds independent information to conventional clinical risk factors. Clin Cancer Res. 2011, 17 (18): 6012-6020. 10.1158/1078-0432.CCR-11-0926.CrossRefPubMed
33.
Zurück zum Zitat van Vliet MH, Reyal F, Horlings HM, van de Vijver MJ, Reinders MJ, Wessels LF: Pooling breast cancer datasets has a synergetic effect on classification performance and improves signature stability. BMC Genomics. 2008, 9: 375-10.1186/1471-2164-9-375.CrossRefPubMedPubMedCentral van Vliet MH, Reyal F, Horlings HM, van de Vijver MJ, Reinders MJ, Wessels LF: Pooling breast cancer datasets has a synergetic effect on classification performance and improves signature stability. BMC Genomics. 2008, 9: 375-10.1186/1471-2164-9-375.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Miller LD, Smeds J, George J, Vega VB, Vergara L, Ploner A, Pawitan Y, Hall P, Klaar S, Liu ET: An expression signature for p53 status in human breast cancer predicts mutation status, transcriptional effects, and patient survival. Proc Natl Acad Sci USA. 2005, 102 (38): 13550-10.1073/pnas.0506230102.CrossRefPubMedPubMedCentral Miller LD, Smeds J, George J, Vega VB, Vergara L, Ploner A, Pawitan Y, Hall P, Klaar S, Liu ET: An expression signature for p53 status in human breast cancer predicts mutation status, transcriptional effects, and patient survival. Proc Natl Acad Sci USA. 2005, 102 (38): 13550-10.1073/pnas.0506230102.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Pawitan Y, Bjöhle J, Amler L, Borg AL, Egyhazi S, Hall P, Han X, Holmberg L, Huang F, Klaar S: Gene expression profiling spares early breast cancer patients from adjuvant therapy: derived and validated in two population-based cohorts. Breast Cancer Res. 2005, 7 (6): R953-R964. 10.1186/bcr1325.CrossRefPubMedPubMedCentral Pawitan Y, Bjöhle J, Amler L, Borg AL, Egyhazi S, Hall P, Han X, Holmberg L, Huang F, Klaar S: Gene expression profiling spares early breast cancer patients from adjuvant therapy: derived and validated in two population-based cohorts. Breast Cancer Res. 2005, 7 (6): R953-R964. 10.1186/bcr1325.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Minn AJ, Gupta GP, Siegel PM, Bos PD, Shu W, Giri DD, Viale A, Olshen AB, Gerald WL, Massague J: Genes that mediate breast cancer metastasis to lung. Nature. 2005, 436 (7050): 518-524. 10.1038/nature03799.CrossRefPubMedPubMedCentral Minn AJ, Gupta GP, Siegel PM, Bos PD, Shu W, Giri DD, Viale A, Olshen AB, Gerald WL, Massague J: Genes that mediate breast cancer metastasis to lung. Nature. 2005, 436 (7050): 518-524. 10.1038/nature03799.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Chin K, DeVries S, Fridlyand J, Spellman PT, Roydasgupta R, Kuo WL, Lapuk A, Neve RM, Qian Z, Ryder T: Genomic and transcriptional aberrations linked to breast cancer pathophysiologies. Cancer Cell. 2006, 10 (6): 529-541. 10.1016/j.ccr.2006.10.009.CrossRefPubMed Chin K, DeVries S, Fridlyand J, Spellman PT, Roydasgupta R, Kuo WL, Lapuk A, Neve RM, Qian Z, Ryder T: Genomic and transcriptional aberrations linked to breast cancer pathophysiologies. Cancer Cell. 2006, 10 (6): 529-541. 10.1016/j.ccr.2006.10.009.CrossRefPubMed
38.
Zurück zum Zitat Edgar R, Domrachev M, Lash AE: Gene Expression Omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 2002, 30 (1): 207-10.1093/nar/30.1.207.CrossRefPubMedPubMedCentral Edgar R, Domrachev M, Lash AE: Gene Expression Omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 2002, 30 (1): 207-10.1093/nar/30.1.207.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Irizarry RA, Bolstad BM, Collin F, Cope LM, Hobbs B, Speed TP: Summaries of Affymetrix GeneChip probe level data. Nucleic Acids Res. 2003, 31 (4): e15-10.1093/nar/gng015.CrossRefPubMedPubMedCentral Irizarry RA, Bolstad BM, Collin F, Cope LM, Hobbs B, Speed TP: Summaries of Affymetrix GeneChip probe level data. Nucleic Acids Res. 2003, 31 (4): e15-10.1093/nar/gng015.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Dubsky P, Filipits M, Jakesz R, Rudas M, Singer C, Greil R, Dietze O, Luisser I, Klug E, Sedivy R: EndoPredict improves the prognostic classification derived from common clinical guidelines in ER-positive, HER2-negative early breast cancer. Ann Oncol. 2012, 24: 640-647.CrossRefPubMedPubMedCentral Dubsky P, Filipits M, Jakesz R, Rudas M, Singer C, Greil R, Dietze O, Luisser I, Klug E, Sedivy R: EndoPredict improves the prognostic classification derived from common clinical guidelines in ER-positive, HER2-negative early breast cancer. Ann Oncol. 2012, 24: 640-647.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Harrell F, Lee K, Mark D: Tutorial in biostatistics multivariable prognostic models: issues in developing models, evaluating assumptions and adequacy, and measuring and reducing errors. Stat Med. 1996, 15: 361-387. 10.1002/(SICI)1097-0258(19960229)15:4<361::AID-SIM168>3.0.CO;2-4.CrossRefPubMed Harrell F, Lee K, Mark D: Tutorial in biostatistics multivariable prognostic models: issues in developing models, evaluating assumptions and adequacy, and measuring and reducing errors. Stat Med. 1996, 15: 361-387. 10.1002/(SICI)1097-0258(19960229)15:4<361::AID-SIM168>3.0.CO;2-4.CrossRefPubMed
42.
Zurück zum Zitat Schemper M: The relative importance of prognostic factors in studies of survival. Stat Med. 1993, 12 (24): 2377-2382. 10.1002/sim.4780122413.CrossRefPubMed Schemper M: The relative importance of prognostic factors in studies of survival. Stat Med. 1993, 12 (24): 2377-2382. 10.1002/sim.4780122413.CrossRefPubMed
43.
Zurück zum Zitat Grambsch PM, Therneau TM: Proportional hazards tests and diagnostics based on weighted residuals. Biometrika. 1994, 81 (3): 515-10.1093/biomet/81.3.515.CrossRef Grambsch PM, Therneau TM: Proportional hazards tests and diagnostics based on weighted residuals. Biometrika. 1994, 81 (3): 515-10.1093/biomet/81.3.515.CrossRef
44.
Zurück zum Zitat Aalen OO: A linear regression model for the analysis of life times. Stat Med. 1989, 8 (8): 907-925. 10.1002/sim.4780080803.CrossRefPubMed Aalen OO: A linear regression model for the analysis of life times. Stat Med. 1989, 8 (8): 907-925. 10.1002/sim.4780080803.CrossRefPubMed
45.
Zurück zum Zitat Aalen OO, Borgan Ø, Gjessing HK: Survival and event history analysis: a process point of view. 2008, New York, USA: SpringerCrossRef Aalen OO, Borgan Ø, Gjessing HK: Survival and event history analysis: a process point of view. 2008, New York, USA: SpringerCrossRef
46.
Zurück zum Zitat Curtis C, Shah S, Chin S, Turashvili G, Rueda O, Dunning M, Speed D, Lynch A, Samarajiwa S, Yuan Y, Gräf S, Ha G, Haffari G, Bashashati A, Russell R, McKinney S, Langerød A, Green A, Provenzano E, Wishart G, Pinder S, Watson P, Markowetz F, Murphy L, Ellis I, Purushotham A, Børresen-Dale AL, Brenton JD, Tavaré S, Caldas C, Aparicio S, METABRIC Group, et al: The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature. 2012, 486 (7403): 346-352. doi:10.1038/nature10983PubMedPubMedCentral Curtis C, Shah S, Chin S, Turashvili G, Rueda O, Dunning M, Speed D, Lynch A, Samarajiwa S, Yuan Y, Gräf S, Ha G, Haffari G, Bashashati A, Russell R, McKinney S, Langerød A, Green A, Provenzano E, Wishart G, Pinder S, Watson P, Markowetz F, Murphy L, Ellis I, Purushotham A, Børresen-Dale AL, Brenton JD, Tavaré S, Caldas C, Aparicio S, METABRIC Group, et al: The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature. 2012, 486 (7403): 346-352. doi:10.1038/nature10983PubMedPubMedCentral
47.
Zurück zum Zitat Cohen J: A coefficient of agreement for nominal scales. Educ Psychol Meas. 1960, 20 (1): 37-46. 10.1177/001316446002000104.CrossRef Cohen J: A coefficient of agreement for nominal scales. Educ Psychol Meas. 1960, 20 (1): 37-46. 10.1177/001316446002000104.CrossRef
48.
Zurück zum Zitat Wirapati P, Sotiriou C, Kunkel S, Farmer P, Pradervand S, Haibe-Kains B, Desmedt C, Ignatiadis M, Sengstag T, Schutz F: Meta-analysis of gene expression profiles in breast cancer: toward a unified understanding of breast cancer subtyping and prognosis signatures. Breast Cancer Res. 2008, 10 (4): 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: Meta-analysis of gene expression profiles in breast cancer: toward a unified understanding of breast cancer subtyping and prognosis signatures. Breast Cancer Res. 2008, 10 (4): R65-10.1186/bcr2124.CrossRefPubMedPubMedCentral
49.
Zurück zum Zitat Desmedt C, Haibe-Kains B, Wirapati P, Buyse M, Larsimont D, Bontempi G, Delorenzi M, Piccart M, Sotiriou C: Biological processes associated with breast cancer clinical outcome depend on the molecular subtypes. Clin Cancer Res. 2008, 14 (16): 5158-10.1158/1078-0432.CCR-07-4756.CrossRefPubMed Desmedt C, Haibe-Kains B, Wirapati P, Buyse M, Larsimont D, Bontempi G, Delorenzi M, Piccart M, Sotiriou C: Biological processes associated with breast cancer clinical outcome depend on the molecular subtypes. Clin Cancer Res. 2008, 14 (16): 5158-10.1158/1078-0432.CCR-07-4756.CrossRefPubMed
50.
Zurück zum Zitat Drier Y, Domany E: Do Two Machine-Learning Based Prognostic Signatures for Breast Cancer Capture the Same Biological Processes?. PLoS One. 2011, 6 (3): e17795-10.1371/journal.pone.0017795.CrossRefPubMedPubMedCentral Drier Y, Domany E: Do Two Machine-Learning Based Prognostic Signatures for Breast Cancer Capture the Same Biological Processes?. PLoS One. 2011, 6 (3): e17795-10.1371/journal.pone.0017795.CrossRefPubMedPubMedCentral
51.
Zurück zum Zitat Sotiriou C, Pusztai L: Gene-expression signatures in breast cancer. N Engl J Med. 2009, 360 (8): 790-800. 10.1056/NEJMra0801289.CrossRefPubMed Sotiriou C, Pusztai L: Gene-expression signatures in breast cancer. N Engl J Med. 2009, 360 (8): 790-800. 10.1056/NEJMra0801289.CrossRefPubMed
52.
Zurück zum Zitat Camp JT, Elloumi F, Roman-Perez E, Rein J, Stewart DA, Harrell JC, Perou CM, Troester MA: Interactions with Fibroblasts Are Distinct in Basal-Like and Luminal Breast Cancers. Mol Cancer Res. 2011, 9 (1): 3-10.1158/1541-7786.MCR-10-0372.CrossRefPubMed Camp JT, Elloumi F, Roman-Perez E, Rein J, Stewart DA, Harrell JC, Perou CM, Troester MA: Interactions with Fibroblasts Are Distinct in Basal-Like and Luminal Breast Cancers. Mol Cancer Res. 2011, 9 (1): 3-10.1158/1541-7786.MCR-10-0372.CrossRefPubMed
53.
Zurück zum Zitat Rody A, Holtrich U, Pusztai L, Liedtke C, Gaetje R, Ruckhaeberle E, Solbach C, Hanker L, Ahr A, Metzler D, Engels K, Karn T, Kaufmann M: T-cell metagene predicts a favorable prognosis in estrogen receptor-negative and HER2-positive breast cancers. Breast Cancer Res. 2009, 11 (2): R15-10.1186/bcr2234.CrossRefPubMedPubMedCentral Rody A, Holtrich U, Pusztai L, Liedtke C, Gaetje R, Ruckhaeberle E, Solbach C, Hanker L, Ahr A, Metzler D, Engels K, Karn T, Kaufmann M: T-cell metagene predicts a favorable prognosis in estrogen receptor-negative and HER2-positive breast cancers. Breast Cancer Res. 2009, 11 (2): R15-10.1186/bcr2234.CrossRefPubMedPubMedCentral
54.
Zurück zum Zitat Teschendorff AE, Caldas C: A robust classifier of high predictive value to identify good prognosis patients in ER-negative breast cancer. Breast Cancer Res. 2008, 10 (4): R73-10.1186/bcr2138.CrossRefPubMedPubMedCentral Teschendorff AE, Caldas C: A robust classifier of high predictive value to identify good prognosis patients in ER-negative breast cancer. Breast Cancer Res. 2008, 10 (4): R73-10.1186/bcr2138.CrossRefPubMedPubMedCentral
55.
Zurück zum Zitat Hallett RM, Dvorkin-Gheva A, Bane A, Hassell JA: A Gene Signature for Predicting Outcome in Patients with Basal-like Breast Cancer. Sci Rep. 2012, 2: 227-CrossRefPubMedPubMedCentral Hallett RM, Dvorkin-Gheva A, Bane A, Hassell JA: A Gene Signature for Predicting Outcome in Patients with Basal-like Breast Cancer. Sci Rep. 2012, 2: 227-CrossRefPubMedPubMedCentral
56.
Zurück zum Zitat Shen R, Olshen AB, Ladanyi M: Integrative clustering of multiple genomic data types using a joint latent variable model with application to breast and lung cancer subtype analysis. Bioinformatics. 2009, 25 (22): 2906-2912. 10.1093/bioinformatics/btp543.CrossRefPubMedPubMedCentral Shen R, Olshen AB, Ladanyi M: Integrative clustering of multiple genomic data types using a joint latent variable model with application to breast and lung cancer subtype analysis. Bioinformatics. 2009, 25 (22): 2906-2912. 10.1093/bioinformatics/btp543.CrossRefPubMedPubMedCentral
57.
Zurück zum Zitat Concato J, Peduzzi P, Holford TR, Feinstein AR: Importance of events per independent variable in proportional hazards analysis I. Background, goals, and general strategy. J Clin Epidemiol. 1995, 48 (12): 1495-1501. 10.1016/0895-4356(95)00510-2.CrossRefPubMed Concato J, Peduzzi P, Holford TR, Feinstein AR: Importance of events per independent variable in proportional hazards analysis I. Background, goals, and general strategy. J Clin Epidemiol. 1995, 48 (12): 1495-1501. 10.1016/0895-4356(95)00510-2.CrossRefPubMed
58.
Zurück zum Zitat Peduzzi P, Concato J, Feinstein AR, Holford TR: Importance of events per independent variable in proportional hazards regression analysis II. Accuracy and precision of regression estimates. J Clin Epidemiol. 1995, 48 (12): 1503-1510. 10.1016/0895-4356(95)00048-8.CrossRefPubMed Peduzzi P, Concato J, Feinstein AR, Holford TR: Importance of events per independent variable in proportional hazards regression analysis II. Accuracy and precision of regression estimates. J Clin Epidemiol. 1995, 48 (12): 1503-1510. 10.1016/0895-4356(95)00048-8.CrossRefPubMed
59.
Zurück zum Zitat Vittinghoff E, McCulloch CE: Relaxing the rule of ten events per variable in logistic and Cox regression. Am J Epidemiol. 2007, 165 (6): 710-718. 10.1093/aje/kwk052.CrossRefPubMed Vittinghoff E, McCulloch CE: Relaxing the rule of ten events per variable in logistic and Cox regression. Am J Epidemiol. 2007, 165 (6): 710-718. 10.1093/aje/kwk052.CrossRefPubMed
60.
Zurück zum Zitat Nielsen TO, Parker JS, Leung S, Voduc KD, Ebbert M, Vickery TL, Davies SR, Snider JE, Stijleman IJ, Reed J: A comparison of PAM50 intrinsic subtyping with immunohistochemistry and clinical prognostic factors in tamoxifen-treated estrogen receptor positive breast cancer. Clin Cancer Res. 2010, 5222-5232. Nielsen TO, Parker JS, Leung S, Voduc KD, Ebbert M, Vickery TL, Davies SR, Snider JE, Stijleman IJ, Reed J: A comparison of PAM50 intrinsic subtyping with immunohistochemistry and clinical prognostic factors in tamoxifen-treated estrogen receptor positive breast cancer. Clin Cancer Res. 2010, 5222-5232.
Metadaten
Titel
Systematic assessment of prognostic gene signatures for breast cancer shows distinct influence of time and ER status
verfasst von
Xi Zhao
Einar Andreas Rødland
Therese Sørlie
Hans Kristian Moen Vollan
Hege G Russnes
Vessela N Kristensen
Ole Christian Lingjærde
Anne-Lise Børresen-Dale
Publikationsdatum
01.12.2014
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2014
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/1471-2407-14-211

Weitere Artikel der Ausgabe 1/2014

BMC Cancer 1/2014 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.