Skip to main content
Erschienen in: Breast Cancer Research 3/2007

Open Access 01.06.2007 | Research article

Molecular response to aromatase inhibitor treatment in primary breast cancer

verfasst von: Alan Mackay, Ander Urruticoechea, J Michael Dixon, Tim Dexter, Kerry Fenwick, Alan Ashworth, Suzanne Drury, Alexey Larionov, Oliver Young, Sharon White, William R Miller, Dean B Evans, Mitch Dowsett

Erschienen in: Breast Cancer Research | Ausgabe 3/2007

Abstract

Background

Aromatase inhibitors such as anastrozole and letrozole are highly effective suppressants of estrogen synthesis in postmenopausal women and are the most effective endocrine treatments for hormone receptor positive breast cancer in such women. Little is known of the molecular effects of these agents on human breast carcinomas in vivo.

Methods

We randomly assigned primary estrogen receptor positive breast cancer patients to treatment with anastrozole or letrozole for 2 weeks before surgery. Expression profiling using cDNA arrays was conducted on pretreatment and post-treatment biopsies. Sample pairs from 34 patients provided sufficient RNA for analysis.

Results

Profound changes in gene expression were seen with both aromatase inhibitors, including many classical estrogen-dependent genes such as TFF1, CCND1, PDZK1 and AGR2, but also many other genes that are likely to represent secondary responses; decrease in the expression of proliferation-related genes were particularly prominent. Many upregulated genes are involved in extracellular matrix remodelling, including collagens and members of the small leucine-rich proteoglycan family (LUM, DCN, and ASPN). No significant differences were seen between letrozole and anastrozole in terms of molecular effects. The gene changes were integrated into a Global Index of Dependence on Estrogen (GIDE), which enumerates the genes changing by at least twofold with therapy. The GIDE varied markedly between tumours and related significantly to pretreatment levels of HER2 and changes in immunohistochemically detected Ki67.

Conclusion

Our findings identify the transcriptional signatures associated with aromatase inhibitor treatment of primary breast tumours. Larger datasets using this approach should enable identification of estrogen-dependent molecular changes, which are the determinants of benefit or resistance to endocrine therapy.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​bcr1732) contains supplementary material, which is available to authorized users.
Alan Mackay, Ander Urruticoechea contributed equally to this work.

Competing interests

MD serves on advisory boards and receives grant income from Astrazeneca and Novartis in relation to AIs. The other authors declare that they have no competing interests.

Authors' contributions

AM and AU carried out the microarray profiling, assisted in data analysis and drafted the manuscript. AM and KF produced the microarrays used in this study, TD carried out the Bioinformatic and statistical analyses. SD and AL carried out the RT-PCR validations. OY and SW carried out the immunohistochemistry. JMD, WM, AA, DE and MD participated in the design and conception of the study, and reviewed analyses of the data. MD, JMD and WM designed the study, and AA supported the laboratory work. All authors read and approved the final manuscript.
Abkürzungen
AI
aromatase inhibitor
ER
estrogen receptor
ECM
extracellular matrix
GIDE
Global Index of Dependence on Estrogen
PCR
polymerase chain reaction
PgR
progesterone receptor.

Introduction

Approaching 80% of human breast carcinomas express estrogen receptor (ER)-α protein at clinically significant levels and are considered ER positive. Estrogen deprivation, or antagonism, is an effective treatment for many but not all patients with such tumours. The selective ER modifier tamoxifen has been the predominant treatment for the past two decades and improves survival in ER-positive patients receiving this as adjuvant therapy after surgery [1]. However, in postmenopausal women aromatase inhibition with the nonsteroidal inhibitors anastrozole and letrozole has now been shown to be more effective than tamoxifen as adjuvant therapy [2]. Letrozole and anastrozole are highly specific for the aromatase enzyme and inhibit whole body aromatization by 99% and 97%, respectively [3]. Aromatase inhibitors (AIs) therefore confer highly selective and essentially complete withdrawal of estrogen in postmenopausal patients.
Proliferation of malignant cells, as measured by expression of the nuclear antigen Ki67, is reduced in more than 90% of ER-positive primary breast carcinomas by treatment with AIs [4, 5]. This suggests that almost all ER-positive tumours derive some proliferative stimulus from estrogen and may be considered hormone responsive; in some patients, however, this effect may be only modest. We recently found that the difference in the change in Ki67 after 2 weeks of treatment with anastrozole or tamoxifen, or the two drugs in combination was predictive of relative recurrence-free survival in a parallel adjuvant trial of the same treatments [6]. Additionally, Ki67 levels after 2 weeks of treatment significantly correlated with recurrence-free survival in the same patients in the presurgical study [7]. Both of these findings support the validity of short-term changes in Ki67 as an intermediate marker of the clinical effectiveness of endocrine therapy. It seems likely, however, that Ki67 is an imperfect marker of proliferation and that changes in gene expression other than those related to proliferation may be involved in determining the clinical effectiveness of estrogen deprivation.
Transcriptional profiling of estrogen responses in ER-positive human breast cancer cell lines and model systems in vitro leads to changes in the transcription of large numbers of genes [8, 9], but very little is known of these effects in vivo or how these effects vary between tumours and whether these molecular changes fully encompass the determinants of clinical response. Biopsy of tumours before and during presurgical treatment with an AI allows the study of estrogen-dependent effects across a range of ER-positive breast carcinomas in situ.
We therefore evaluated the effects of estrogen deprivation with letrozole or anastrozole on Ki67 expression and transcriptional profiles in ER-positive breast carcinomas in vivo. Such an approach might provide insights into the mechanisms of clinical benefit and allow the development of a predictor of that benefit.
Our specific aims in the present study were as follows: to determine whether there is a significant difference between letrozole and anastrozole in terms of change in Ki67 (reported elsewhere) and changes in gene transcription; to identify the genes that change with aromatase inhibition and to integrate these as a Global Index of Dependence on Estrogen (GIDE); to assess how the most prominent gene changes relate to those reported in vitro with estrogen stimulation; and to determine the relationship between the GIDE and previously described putative determinants of benefit from endocrine therapy such as HER2 and Ki67 expression.

Materials and methods

Patient samples

Postmenopausal patients with primary ER-positive (Allred scores 2 to 8; note that scores of 2 are conventionally regarded as ER negative [10]) breast cancer were randomly assigned to presurgical treatment for 2 weeks with letrozole (2.5 mg/day orally) or anastrozole (1 mg/day orally). Multiple core-cut biopsies were taken with a 14-guage needle before treatment and at surgery from 54 patients, and were either immediately frozen in liquid nitrogen for RNA analysis or were fixed in neutral buffered formalin for immunohistochemistry. RNA from each frozen biopsy was extracted using Trizol (Invitrogen, Paisley, UK), in accordance with the manufacturer's instructions.

Microarray hybridizations

Total RNA integrity was confirmed on an Agilent 2100 BioAnalyser (Agilent Technologies, South Queensferry, West Lothian, UK) before linear T7 amplification using a RiboAmp kit (Arcturus/Molecular Devices Corporation Sunnyvcale, CA, USA). Amplified aRNA (4 μg) was labeled with either cy3 or cy5 and hybridized to Breakthrough 17K cDNA microarrays in replicate dye swap hybridizations, as previously described [11]. The Breakthrough 17K microarray platform and all primary microarray data have been submitted to Array Express (ArrayExpress submission number E-TABM-180). Annotation of the Breakthrough 17K cDNA microarray based on build 189 of Unigene is provided as Additional file 6.

Analysis of microarray data

Expression values from spots with hybridization artefacts or extremely low intensities were flagged in Genepix 5.1 (Axon Instruments/Molecular Devices Corporation Sunnyvcale, CA, USA) and then converted to missing values and removed from the analysis. The raw intensity values were then converted to log2 ratios of sample to reference (M values) and log2 average spot intensity (A values) for all subsequent pre-processing and analysis. The loess local regression function was used to remove biases resulting from the combined effect of spot intensity and the row group to which the spot belonged, and then to remove the more global bias across the slide. A quantile filter was used to remove data that had average intensity or A values below the 25th percentile in 60% or more of the hybridizations. The M values for each hybridization were rescaled so as to remove the relationship between increasing dispersion of M values with increasing dispersion of A values across the hybridisations. This latter transformation did not involve extensive rescaling of the data and although it clarified the relationships found in this study, these were all apparent without this step. The replicate dye swap hybridizations were then averaged. This left 14,024 genes that were used for paired (pre/post) differential gene expression analysis using SAM version 2.21 [12]. In order to focus on the more extensive gene fluctuations between samples, further reductions in the number of genes used for some analyses were based on filtering out genes with low inter-sample variation. We used the interquartile range as a robust estimate of gene variation and used a stringent threshold at interquartile range = 0.75 (2,418 genes remaining).
In order to map the gross phenotypic changes across the samples, the following supervised analysis was chosen. A core set of genes was selected using the weighted Kolmogorov-Smirnov statistic because of its robustness and flexibility. The maximum number of genes (421) that gave minimum leave-one-out cross-validation in separating pretreatment from post-treatment samples using the k-nearest-neighbor algorithm (k = 7) was identified, and these genes were retained. Agglomerative clustering (see below) was used to separate the 421 selected genes into 10 clusters of co-regulated genes. Each cluster was then represented by the average M value of its genes for each sample (metagene) following centering and rescaling across samples. All clustering used the flexible β agglomerative clustering algorithm with a correlation distance measure for both genes and arrays. Clustering heat maps were produced with Java Treeview 1.0.12 software. Correlations were performed with Spearman rank or Pearson correlations.

Immunohistochemistry

Conventional immunohistochemistry was performed on each biopsy using antibodies for ER-α clone 6F11 (Novocatsra Laboratories, Newcastle. upon Tyne, UK), progesterone receptor (PgR) clone PgR 636 (Dako Ltd., Ely, Cambridgeshire, UK), and KI67 clone MIB-1 (DAKO), in accordance with the manufacturers' instructions.
ER and PgR immunohistochemistry were quantitated according to Allred score [10]. Ki67 immunohistochemistry is reported as the number of positive cells among 1,000 malignant cells counted and is expressed as a percentage.

Real-time PCR

Quantitative real-time PCR was conducted in five genes of interest (CCND1, PDZK1, FAS, TFF1, and MAN1A1). Total RNA from the same RNA preparations as used for microarray analysis was reverse transcribed using random primers and Superscript III (Invitrogen), in accordance with the manufacturer's instructions. A reverse transcription negative control was included to account for any genomic DNA contamination. cDNA samples were subjected to quantitative PCR using Taqman® (Applied Biosystems, Warrington, UK) on an ABI Prism 7900 HT with primers designed by Primer Express, or Quantitect SYBR green (Qiagen, Crawley, West Sussex, UK) on an Opticon Monitor 2 with primers designed by Primer 3 in two different laboratories. Primer sequences are shown in Additional file 1. Pretreatment/post-treatment changes were estimated after normalization using the geometric mean of the two reference genes that had been shown to be unchanged in expression during treatment with AIs (TBP and KIAA0674).

Results

RNA of sufficient quality and quantity was obtained from 34 pretreatment/post-treatment pairs of samples. The following findings refer solely to those samples. Patient clinical information is summarized in Additional file 2.
The samples were clustered to determine whether pretreatment and post-treatment biopsies aggregated together as nearest neighbors in clustering dendrograms. Half of pre/post biopsy pairs were found to co-aggregate whether based on all 14,034 measured genes (17/34) or the 2,418 genes remaining following filtering to retain the most variable genes (18/34). Similar proportions of co-aggregating pairs were also identified using other algorithms (for instance, complete linkage and group average linkage; data not shown). Separation of paired biopsies in this analysis contrasts with other studies in which the differences in gene expression among breast tumours is far greater than that observed as a result of treatment with chemotherapeutic agents [11, 13].
A heat map diagram from clustering of the 2,418 most variable genes among the 68 biopsies is shown in Figure 1. Clusters of genes containing some of the most important known markers of breast tumour phenotype are shown in greater detail: ESR1 (Figure 1a), MKI67 (Figure 1b), ERBB2 (Figure 1c), and TFF1 (Figure 1d). Levels of ESR1 and ERBB2 gene expression were inversely correlated in these samples (r = -0.57, P = 0.0005, Pearson correlation), as has been shown in many other studies of breast tumours. The samples with the lowest ESR1 or high ERBB2 invariably had pre/post biopsy pairs that co-aggregated as nearest neighbours and accounted for more than half (11/17) of the co-aggregating pairs. The ERBB2 cluster contained several genes that are present in the ERBB2 amplicon on chromosome 17q12-21, including GRB7, THRAP4, and STARD3, and were highly over-expressed in the four HER2 amplified cases. Data files for Java Treeview are provided as supplementary information (Additional files 7, 8, 9, 10).
To summarize the effects of estrogen deprivation on gene expression, we have derived a Global Index of Dependence on Estrogen (GIDE). This index was defined as the number of genes changing by at least twofold between each pair of biopsies irrespective of the direction of change. This index correlated positively with change in the proliferation marker Ki67 (Spearman rank rho = 0.533, P = 0.0022; Figure 2a) and negatively with the expression of ERBB2 (Spearman rank rho = -0.381, P = 0.0282; Figure 2b). Although no patients with a high GIDE were among the lowest in terms of ESR1 expression, overall there was not a significant correlation between the two. A complete summary of GIDE data is provided in Additional file 3.
The primary end-point of the study was the reduction in tumour proliferation measured as the change in the biomarker Ki67 by conventional immunohistochemistry. The relationship between change in Ki67 immunohistochemistry and microarray expression of ESR1 and ERBB2 is shown in Figure 2c,d. Tumours expressing low levels of ER or high levels of ERBB2 exhibited less reduction in Ki67 staining following AI treatment.
Correlations of the GIDE with immunohistochemical measurements of ER and PgR (Allred score) are shown in Figure 2e,f. In these samples there was a significant correlation of GIDE with pretreatment ER staining but not with that of PgR. There was no significant difference between letrozole and anastrozole in their effects on the GIDE or on Ki67, confirming the result for the whole patient set [14].
A paired SAM statistical analysis identified 1,395 genes that were upregulated and 1,264 genes that were downregulated by AI treatment using a local false discovery rate threshold of 1%. Significantly changing genes were then ranked according to their average fold change; the top 40 downregulated genes are listed in Table 1 and the top 40 upregulated genes are listed in Table 2 (the complete list is shown in Additional file 4). The most consistently downregulated genes included TFF1, PDZK1, AGR2, TFF3, STC2, and CCND1. The most consistently upregulated genes included LUM, CALD1, ASPN, DCN, PDGFRA, VIM, SPARC, MAN1A1, and FAS. Quantitative real-time PCR confirmed significant upregulation of MAN1A1 and FAS (P < 0.05 for each) and downregulation of TFF1, PDZK1, and CCND1 (P < 0.01, P < 0.001 and P < 0.001, respectively; data not shown). The complete list of upregulated and downregulated genes was subjected to Gene Ontology analysis using Onto-Express and Pathway Express [15].
Table 1
Genes downregulated by AI treatment
ID
Symbol
Description
Unigene
Fold
LFDR
HSI182A05
TFF1
Trefoil factor 1 (breast cancer, estrogen-inducible)
Hs.162807
0.26
0.09
HSI054D07
HBB
Haemoglobin, beta
Hs.523443
0.31
0.00
HSI047B01
PDZK1
PDZ domain containing 1
Hs.444751
0.35
0.00
HSI035H02
CYP2B6
Cytochrome P450, family 2B6
Hs.1360
0.41
0.04
HSI075H09
AGR2
Anterior gradient 2 homolog (Xenopus laevis)
Hs.530009
0.41
0.08
HSI031E07
STARD10
START domain containing 10
Hs.188606
0.42
0.00
HSI183G10
TFF3
Trefoil factor 3 (intestinal)
Hs.82961
0.43
0.00
HSI147F09
ZBTB20
Zinc finger and BTB domain containing 20
Hs.570657
0.45
0.00
HSI182A08
STC2
Stanniocalcin 2
Hs.233160
0.47
0.00
HSI147F08
KTN1
Kinectin 1 (kinesin receptor)
Hs.509414
0.47
0.00
HSI059H10
LOC143381
Hypothetical protein LOC143381
Hs.388347
0.47
0.00
HSI147F10
MSI2
Musashi homolog 2 (Drosophila)
Hs.134470
0.49
0.00
HSI177G07
EST
Transcribed locus
Hs.443277
0.50
0.10
HSI053H02
UBE2C
Ubiquitin-conjugating enzyme E2C
Hs.93002
0.52
0.10
HSI096C06
MAPT
Microtubule-associated protein tau
Hs.101174
0.52
0.00
HSI049A02
ERGIC1
ER-golgi intermediate compartment 1
Hs.509163
0.53
0.09
HSI040C08
AZGP1
Alpha-2-glycoprotein 1, zinc
Hs.546239
0.55
0.00
HSI133F06
EST
Transcribed locus
Hs.159264
0.55
0.00
HSI182E08
PLAT
Plasminogen activator, tissue
Hs.491582
0.55
0.00
HSI033B05
LY6E
Lymphocyte antigen 6 complex, locus E
Hs.521903
0.56
0.00
HSI048F12
CCND1
Cyclin D1
Hs.523852
0.56
0.10
HSI085G12
KCNK15
Potassium channel, subfamily K, member 15
Hs.528664
0.57
0.00
HSI177H07
PCBP3
Poly(rC) binding protein 3
Hs.474049
0.57
0.10
HSI032D02
ABCA3
ATP-binding cassette, subfamily A3 (ABC1)
Hs.26630
0.57
0.00
HSI182A02
TFF3
Trefoil factor 3 (intestinal)
Hs.82961
0.58
0.04
HSI025A03
FGD3
FYVE, RhoGEF and PH domain containing 3
Hs.411081
0.58
0.00
HSI070B06
AP1S1
Adaptor-related protein complex 1, sigma 1 subunit
Hs.489365
0.58
0.08
HSI057H12
GNB2
Guanine nucleotide binding protein beta 2
Hs.185172
0.58
0.00
HSI080H11
SEMA3F
Semaphorin 3F
Hs.32981
0.59
0.00
HSI054G06
NUSAP1
Nucleolar and spindle associated protein 1
Hs.511093
0.59
0.00
HSI124D07
RIMS4
Regulating synaptic membrane exocytosis 4
Hs.517065
0.59
0.00
HSI065C09
CNNM2
Cyclin M2
Hs.500903
0.59
0.00
HSI080F02
PREX1
PIP3-dependent RAC exchanger 1
Hs.153310
0.59
0.00
HSI095H09
C6orf97
Chromosome 6 open reading frame 97
Hs.130239
0.59
0.00
HSI161G02
EST
Transcribed locus
Hs.570637
0.60
0.00
HSI045G02
UBE2T
Ubiquitin-conjugating enzyme E2T (putative)
Hs.5199
0.60
0.00
HSI046F10
TOP2A
Topoisomerase (DNA) II alpha 170 kDa
Hs.156346
0.60
0.00
HSI183D04
AR
Androgen receptor
Hs.496240
0.61
0.01
HSI183A08
SLC9A3R1
Solute carrier family 9, member 3 regulator 1
Hs.396783
0.61
0.00
HSI025G02
SHARPIN
SHANK-associated RH domain interactor
Hs.529755
0.61
0.08
Shown are the first 40 downregulated genes from a paired SAM analysis, which identified 1,264 genes to be downregulated by aromatase inhibitor (AI) treatment below a local false discovery rate of 1% (LFDR). The genes are ranked according to their fold change.
Table 2
Genes upregulated by AI treatment
ID
Symbol
Description
Unigene
Fold
LFDR
HSI022G08
LUM
Lumican
Hs.406475
2.87
0.11
HSI101E05
ODF2L
Outer dense fiber of sperm tails 2-like
Hs.149360
2.80
0.07
HSI027H04
IGJ
Immunoglobulin J polypeptide
Hs.381568
2.73
0.07
HSI082D05
RNH1
Ribonuclease/angiogenin inhibitor 1
Hs.530687
2.51
0.00
HSI056B04
COL3A1
Collagen, type III, alpha 1
Hs.443625
2.50
0.00
HSI182D05
MRC1L1
Mannose receptor, C type 1
Hs.461247
2.45
0.06
HSI067F08
C21orf70
Chromosome 21 open reading frame 70
Hs.410830
2.44
0.00
HSI127E04
CALD1
Caldesmon 1
Hs.490203
2.37
0.11
HSI030C06
PTPRC
Protein tyrosine phosphatase, receptor type, C
Hs.192039
2.36
0.11
HSI067H02
ASPN
Asporin (LRR class 1)
Hs.435655
2.34
0.00
HSI066B08
COL14A1
Collagen, type XIV, alpha 1 (undulin)
Hs.409662
2.28
0.11
HSI049B12
COL1A2
Collagen, type I, alpha 2
Hs.489142
2.28
0.00
HSI049G07
DCN
Decorin
Hs.156316
2.28
0.00
HSI067E05
MRC1L1
Mannose receptor, C type 1
Hs.461247
2.22
0.05
HSI101D05
IFT122
Intraflagellar transport 122 homolog (Chlamydomonas)
Hs.477537
2.17
0.03
HSI183E05
PDGFRA
Platelet-derived growth factor receptor, alpha
Hs.74615
2.14
0.09
HSI031A12
FSTL1
Follistatin-like 1
Hs.269512
2.12
0.00
HSI183H01
COL5A2
Collagen, type V, alpha 2
Hs.445827
2.11
0.00
HSI055A11
ECM2
Extracellular matrix protein 2
Hs.117060
2.08
0.09
HSI018G02
SPON1
Spondin 1, extracellular matrix protein
Hs.445818
2.06
0.00
HSI183B01
PDGFRA
Platelet-derived growth factor receptor, alpha
Hs.74615
2.04
0.01
HSI037C02
CPVL
Carboxypeptidase, vitellogenic-like
Hs.233389
2.03
0.06
HSI062B08
SAS10
Disrupter of silencing 10
Hs.322901
2.01
0.00
HSI129E10
ADAM12
ADAM metallopeptidase domain 12 (meltrin alpha)
Hs.386283
2.00
0.00
HSI183G08
RGS1
Regulator of G-protein signalling 1
Hs.75256
1.98
0.18
HSI054F01
VIM
Vimentin
Hs.533317
1.97
0.00
HSI048C08
CTGF
Connective tissue growth factor
Hs.410037
1.97
0.00
HSI183G05
SPARC
Secreted protein, acidic, cysteine-rich (osteonectin)
Hs.111779
1.96
0.00
HSI139G09
ADAMTS2
ADAM metallopeptidase with thrombospondin motif 2
Hs.23871
1.96
0.00
HSI018D05
FBLN1
Fibulin 1
Hs.24601
1.95
0.00
HSI040E08
DUSP1
Dual specificity phosphatase 1
Hs.171695
1.95
0.00
HSI082C05
MAN1A1
Mannosidase, alpha, class 1A, member 1
Hs.102788
1.94
0.01
HSI098G12
RARRES1
Retinoic acid receptor responder 1
Hs.131269
1.94
0.00
HSI044C09
SAT
Spermidine/spermine N1-acetyltransferase
Hs.28491
1.92
0.09
HSI030F05
HTRA1
HtrA serine peptidase 1
Hs.501280
1.92
0.00
HSI088D11
CILP
Cartilage intermediate layer protein
Hs.442180
1.91
0.00
HSI040E09
MME
Membrane metallo-endopeptidase (CALLA, CD10)
Hs.307734
1.91
0.00
HSI028G12
PDGFRA
Platelet-derived growth factor receptor, alpha
Hs.74615
1.89
0.00
HSI060C05
FN1
Fibronectin 1
Hs.203717
1.89
0.00
HSI045G12
CXCL12
Chemokine ligand 12 (stromal cell-derived factor 1)
Hs.522891
1.88
0.00
Shown are the first 40 upregulated genes from a paired SAM analysis, which identified 1,365 genes to be upregulated by aromatase inhibitor (AI) treatment below a local false discovery rate of 1% (LFDR). The genes are ranked according to their fold change.
The change in expression in some of these key index genes in individual patients is shown in Figure 3; the change in Ki67 immunohistochemistry is also shown for comparison. The majority of tumours exhibit large changes in expression of these genes. However, changes in the expression of individual estrogen-responsive genes did not clearly identify tumours with a poor antiproliferative response. Different subsets of tumours exhibited the largest or smallest responses in expression changes for each different gene.
To map the gross phenotypic changes of the tumours in response to AI treatment relative to their initial states, we selected a core set of 421 genes that distinguished pretreatment from post-treatment biopies (see Materials and methods, above). These were used to produce the heat map shown in Figure 4 and separated the biopsies into predominantly pretreatment and post-treatment arms. Three of the four HER2 amplified cases had pretreatment profiles that segregated in the post-treatment arm (216, 228, and 64). The fourth (203) was the only case that expressed high levels of both ESR1 and ERBB2. Seven of the eight pretreatment biopsies that were incorrectly grouped included seven of the 10 biopsies with the lowest pretreatment expression of ESR1 (217, 216, 228, 138, 39, 64, and P3). Data files for Java Treeview are provided as supplementary information (Additional files 11, 12, 13, 14).
Three clusters in this supervised analysis clearly represented distinct pathway related phenotypes based upon the ontology of the genes they contain (Figure 4). Genes in the 'proliferation cluster' exhibited a highly significant overlap with a previously characterized breast cancer proliferation signature [16]. We labelled a cluster containing many genes known to be classically estrogen responsive in breast cancer as an 'estrogen cluster' and one including collagens and other genes involved in extracellular matrix (ECM) deposition as an 'ECM cluster'. Figure 5a shows the combined effect of treatment on the estrogen and proliferation metagenes (mean of each cluster's M values) as a vector diagram in which the pretreatment and post-treatment samples are joined by an arrow. Tumours with extremely low baseline levels of estrogen-dependent gene expression and HER2 amplified tumours exhibit very little change in either cluster (for example, 39, 138 red arrows and 218, 216, 64, green dots, respectively). Perhaps most importantly, this analysis identified a number of cases that had major reductions in expression of the estrogen metagene with minimal impact on the proliferation metagene (for example, 145, 262, 263, blue arrows). Figure 5b shows the interaction of the estrogen metagene and the ECM metagene. The ECM metagene is clearly upregulated in the majority of biopsies irrespective of pretreatment levels of ESR1 and estrogen metagene values (red arrows). The proliferation metagene exhibited the highest positive correlation (r = 0.51, P = 0.000029) with the change in Ki67 immunohistochemistry of any of the nine metagenes (for example, estrogen metagene: r = 0.31, P = 0.102).
Array profiling also identified sets of genes that were both positively and negatively correlated with ER in these biopsies. The intersection of genes associated with ER and those identified as estrogen responsive indicated that only 10% of the genes most highly correlated with high ESR1 expression were downregulated by estrogen deprivation in vivo. A complete list of genes whose expression correlates with ESR1 with a Pearson correlation of greater or less than 0.5 is given in Additional file 5.

Discussion

Anastrozole and letrozole are highly specific and efficient inhibitors of the aromatase enzyme, leading to profound estrogen deprivation in postmenopausal women [17]. These agents are also the most effective treatment for breast cancer in postmenopausal patients and have become the standard of care over recent years [2]. Here, we have used gene expression profiling by microarray to identify the longitudinal differences in gene expression between matched pretreatment and post-treatment biopsies of tumours from patients treated with AIs. The data generated in this study are biologically relevant in terms of identifying genes that respond to estrogen withdrawal in primary breast tumours in vivo, and are clinically relevant in identifying genes or groups of genes that may be used to understand and predict the response of patients to AI treatment. Although many reports have examined estrogen-regulated gene expression in breast cancer cells and model systems, generating a comprehensive genome-wide catalogue of estrogen-responsive genes [18], there are as yet few reported studies in which an AI was used as a biological probe of estrogen-dependent expression profiles in human breast carcinomas in vivo [19, 20]. The number of patients included in our study was too small for confidence in matters of detail, but important broad messages may be developed.
Several studies have been reported over the past few years utilizing expression profiling of breast tumours, which demonstrated that the expression of ER by breast carcinomas is a consistently dominant feature in their transcriptional profile [13, 21, 22]. Although these studies have identified many hundreds of genes that are significantly associated with ER expression, it is has not been clear which of these genes are directly responsible for estrogen responses in tumour cells. The present study indicates that only a small proportion of the genes correlating with ER status are estrogen-responsive in vivo.
In the present study we included only ER-positive tumours (plus three tumours with Allred scores of 2, conventionally considered ER negative) [10]. Correlations between gene expression and ER in the current dataset were therefore made in relation to degree of ER expression rather than to ER positivity or negativity. Nonetheless, we observed strong correlations between ER and many genes that have previously been shown to be strongly associated with ER positivity, including GATA3, FOXA1, AGR2, AR, and STC2, in microarray profiling studies of mixed ER-positive and ER-negative tumours [2123]. The present study indicates that only a small proportion of the genes correlating with ER status are estrogen responsive in vivo.
The GIDE may be a useful approach to characterizing the overall biological reactivity of a tumour to and dependence on estrogen. The data indicate that there is a continuum of such dependence, with 3,304 genes changing in one tumour by more than twofold over the 2-week treatment period, whereas only 105 changed in another. These data recapitulate the continuum of change shown by Ki67 immunohistochemistry, which indicates that almost all ER-positive tumours exhibit an antiproliferative response to estrogen deprivation, although this is highly variable between patients. The data from the GIDE similarly suggest that few ER-positive tumours are completely nonresponsive to estrogen deprivation. There was only a modestly significant relationship between the GIDE and the pretreatment level of ER based on immunohistochemistry; the current data suggest that high ER expression may be necessary for a tumour to be highly responsive (high GIDE) but that some tumours with a high ER have only a moderate or poor biological response.
The GIDE may be a useful end-point for investigating the mechanisms of resistance to hormonal therapy. One putative mechanism is through over-expression of growth factor receptors such as HER2. Although HER2 was associated with a low GIDE, in all but one case these tumours also had low ER, as has previously been observed [24]. PgR positivity has generally been regarded as indicative of an intact ER mechanism. An association with a higher GIDE might have been anticipated, and although there was a trend toward a positive association with higher PgR expression, this was not significant, possibly because of the limited numbers of samples. Although the GIDE would benefit from a proven association with clinical outcome, we recently showed that 2-week change in Ki67 was predictive of long-term outcome after treatment with endocrine agents in the adjuvant setting [6, 25, 26], and in this study the GIDE was found to be significantly associated with change in Ki67. The profound changes in transcriptional profiles found in some but not all tumours in this study suggest that it is possible that predicting clinical response to an AI by transcriptional profiling may, as with Ki67, be more precise when conducted in tumours shortly after starting treatment.
There have been many reports of the transcriptional profiling of estrogen responses in breast cancer cell lines in vitro, including those in MCF-7 [8, 9], T47D [27] and ZR75.1 [28] breast cancer cell lines and their derivatives [29, 30], as well as those using model systems in experimental animals [31, 32]. These studies identified many hundreds of genes that were upregulated and downregulated by estrogen treatments. Computational and experimental attempts have also been made to integrate these data and catalogue all of the estrogen-responsive genes and estrogen response elements in the genome [18, 33]. Many of the genes upregulated by estrogen in vitro were downregulated by AI treatment in our study including the majority of classically estrogen-responsive genes (TFF1, TFF3, CYP2B6, PDZK1, and AGR2). TFF1 (pS2) is one of the best characterized estrogen-responsive genes in breast cancer [8, 3436]. CYP2B6 is dramatically upregulated by estrogen in ZR75.1 cells, although it is not expressed in MCF-7 cells [37]. PDZK1 has consistently been identified as one of the genes most highly upregulated by oestradiol in MCF-7 cells [8, 9]. AGR2 is another classically estrogen-responsive gene that is expressed in both cell lines and ER-positive breast tumours [38] and that has been associated with a poor response to hormonal therapy [39]. One of the genes that we found to be significantly downregulated by AIs was that encoding aromatase itself (CYP19A1). This finding supports earlier evidence of a positive autocrine feedback loop [40].
In contrast to the genes downregulated by AI treatment, the upregulated genes are not represented by those that are directly downregulated by estrogen in cell lines in vitro. Gene Ontology analysis of the upregulated genes identified pathways involved with the regulation of the actin cytoskeleton, cytokine-receptor interactions and focal adhesion to be more commonly associated with the functions of stromal components than of epithelial cells (VIM, CTGF, FN1, and SPARC). The genes most highly upregulated by AI treatment include several members of the of the small leucine-rich proteoglycan family (LUM, ASPN, and DCN), which regulate matrix remodelling. Lumican is not expressed in cancer cells in breast cancers but in fibroblasts, and it is associated with high tumour grade, low ER levels and young age [41]. Decorin is preferentially expressed in stromal areas in proliferating endometrium, is directly upregulated by estrogens in stromal endometrial cells in vitro [42] and is upregulated in mouse uterus by estrogen treatment [43]. Asporin is closely related to biglycan, which has been shown to be downregulated by estrogen in the stroma of normal human breast tissue in a mouse xenograft model [32]. The effect of this stromal signature on patient survival is unclear but reduced small leucine-rich proteoglycan family expression has been observed in poor prognosis ER-negative breast cancer [44].
There are several possible mechanisms for this upregulation of a stromal signature clearing response to AI treatment. It was notable that the genes representing this stromal signature were upregulated independent of high level ER-α expression in tumour cells. It is possible that upregulation may result from an interaction with stromally expressed ER-β [45]. For example, CD36 has been shown to be directly upregulated by estrogen via ER-β [46], and both lumican and PDGFRA were induced by the selective ER modulator raloxifene in U2OS cells transfected with ER-β [30]. Among the other genes upregulated by AI treatment in the present study are genes representative of the normal profiles of luminal and myoepithelial phenotypes [47, 48] that are not driven by high-level ER over-expression, including RARRES1, MME, TCF4, SFN, and CAV1. This represents a joint upregulation in post-treatment biopsies of a basal/stromal phenotype, which has also been shown in estrogen treatments of normal human breast tissue in xenograft studies [32].
Taken together, these findings highlight the fact that studies identifying estrogen-responsive genes in cell lines do not take into account the diversity of responsiveness, composition and genetic backgrounds seen in primary ER-positive breast tumours. Although many of the gene changes are likely to be directly transcriptionally regulated by estrogen, it is also likely that the majority are a secondary consequence of estrogen deprivation and the resulting inhibition of breast tumour proliferation by AI treatment. Recently, Oh and coworkers [38] have attempted to integrate data on estrogen responsiveness in MCF-7 cells in vitro with gene expression and clinical outcome data from 65 ER-positive and/or PgR-positive breast cancer patients to predict outcome for hormone responsive breast cancer. The study used only the 383 genes that were upregulated by estrogen treatment in this single cell line but, a very high dose (1 μmol/l) of oestradiol was used and dosage differences have been suggested to compromise comparisons of transcriptional signatures [49]. The identification of a comprehensive profile of estrogen-responsive genes in tumours deprived of estrogen in vivo may be expected to provide a much better basis on which to classify the estrogen response of breast tumours than in vitro studies.
Robust gene selection methods were used to identify genes that together best separated pretreatment from post-treatment samples. Cluster analysis using these genes identified groups associated with proliferation and estrogen responses. The 32 genes that constitute the 'proliferation cluster' contain 17 of those reported by Dai and coworkers [16] as a proliferation signature containing critical genes predicting the long-term clinical outcome of patients with ER-positive breast tumours. To summarize both the 'estrogen cluster' and the 'proliferation cluster', we used metagene values to depict the relative changes in tumour phenotype in response to AI treatment. In most tumours there was a coordinated decrease in both of these clusters, but we observed that in some tumours these facets of phenotype change were uncoupled. A better understanding of the mechanisms that lead to a poor antiproliferative response in the presence of a good response in the 'estrogen cluster' of genes is likely to provide a guide to additional treatments for ER-positive breast cancer and may be possible with an extension of this study to larger numbers of tumours.

Conclusion

In summary, short-term estrogen deprivation with AIs leads to profound changes in transcriptional profiles. Although many of the genes were previously described in cell culture studies as responsive to estrogen stimulation, many additional estrogen-responsive genes were identified that responded to estrogen deprivation in vivo, particularly those that are repressed by estrogen. The study revealed complex changes in estrogen-responsive pathways, proliferation and matrix remodelling, which cannot be simply summarized by the ER status of the tumours or completely recapitulated in cell line studies. The global changes in gene expression can be integrated into a GIDE that we found to be associated with previously established correlates with clinical outcome. Studies of this type, linked with clinical outcome, should enable the key genes that underpin clinical response/benefit to be established and may be expected to reveal the molecular features of tumours responsible for sensitivity and resistance to estrogen deprivation.

Acknowledgements

This work was funded by unconditional grants from Novartis to MD and JMD, and by Breakthrough Breast Cancer and the Evelyn Lauder Breast Cancer Research Fund. The authors would like to thank Dr Anita Grigoriadis for her help with the submission of microarray data to ArrayExpress. We would also like to thank Dr Ian Giddings and the members of the CRUK microarray facility for the joint use of IMAGE clone collections in the production of the 17K Breakthrough cDNA microarrays used in this study.

Competing interests

MD serves on advisory boards and receives grant income from Astrazeneca and Novartis in relation to AIs. The other authors declare that they have no competing interests.

Authors' contributions

AM and AU carried out the microarray profiling, assisted in data analysis and drafted the manuscript. AM and KF produced the microarrays used in this study, TD carried out the Bioinformatic and statistical analyses. SD and AL carried out the RT-PCR validations. OY and SW carried out the immunohistochemistry. JMD, WM, AA, DE and MD participated in the design and conception of the study, and reviewed analyses of the data. MD, JMD and WM designed the study, and AA supported the laboratory work. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Early Breast Cancer Trialists' Collaborative Group: Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials. Lancet. 2005, 365: 1687-1717. 10.1016/S0140-6736(05)66544-0.CrossRef Early Breast Cancer Trialists' Collaborative Group: Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials. Lancet. 2005, 365: 1687-1717. 10.1016/S0140-6736(05)66544-0.CrossRef
2.
Zurück zum Zitat Smith IE, Dowsett M: Aromatase inhibitors in breast cancer. N Engl J Med. 2003, 348: 2431-2442. 10.1056/NEJMra023246.CrossRefPubMed Smith IE, Dowsett M: Aromatase inhibitors in breast cancer. N Engl J Med. 2003, 348: 2431-2442. 10.1056/NEJMra023246.CrossRefPubMed
3.
Zurück zum Zitat Geisler J, Haynes B, Anker G, Dowsett M, Lonning PE: Influence of letrozole and anastrozole on total body aromatization and plasma estrogen levels in postmenopausal breast cancer patients evaluated in a randomized, cross-over study. J Clin Oncol. 2002, 20: 751-757. 10.1200/JCO.20.3.751.CrossRefPubMed Geisler J, Haynes B, Anker G, Dowsett M, Lonning PE: Influence of letrozole and anastrozole on total body aromatization and plasma estrogen levels in postmenopausal breast cancer patients evaluated in a randomized, cross-over study. J Clin Oncol. 2002, 20: 751-757. 10.1200/JCO.20.3.751.CrossRefPubMed
4.
Zurück zum Zitat Ellis MJ, Tao Y, Young O, White S, Proia AD, Murray J, Renshaw L, Faratian D, Thomas J, Dowsett M, et al: Estrogen-independent proliferation is present in estrogen-receptor HER2-positive primary breast cancer after neoadjuvant letrozole. J Clin Oncol. 2006, 24: 3019-3025. 10.1200/JCO.2005.04.3034.CrossRefPubMed Ellis MJ, Tao Y, Young O, White S, Proia AD, Murray J, Renshaw L, Faratian D, Thomas J, Dowsett M, et al: Estrogen-independent proliferation is present in estrogen-receptor HER2-positive primary breast cancer after neoadjuvant letrozole. J Clin Oncol. 2006, 24: 3019-3025. 10.1200/JCO.2005.04.3034.CrossRefPubMed
5.
Zurück zum Zitat Burcombe R, Wilson GD, Dowsett M, Khan I, Richman PI, Daley F, Detre S, Makris A: Evaluation of Ki-67 proliferation and apoptotic index before, during and after neoadjuvant chemotherapy for primary breast cancer. Breast Cancer Res. 2006, 8: R31-10.1186/bcr1508.CrossRefPubMedPubMedCentral Burcombe R, Wilson GD, Dowsett M, Khan I, Richman PI, Daley F, Detre S, Makris A: Evaluation of Ki-67 proliferation and apoptotic index before, during and after neoadjuvant chemotherapy for primary breast cancer. Breast Cancer Res. 2006, 8: R31-10.1186/bcr1508.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Dowsett M, Smith IE, Ebbs SR, Dixon JM, Skene A, Griffith C, Boeddinghaus I, Salter J, Detre S, Hills M, et al: Proliferation and apoptosis as markers of benefit in neoadjuvant endocrine therapy of breast cancer. Clin Cancer Res. 2006, 12: 1024s-1030s. 10.1158/1078-0432.CCR-05-2127.CrossRefPubMed Dowsett M, Smith IE, Ebbs SR, Dixon JM, Skene A, Griffith C, Boeddinghaus I, Salter J, Detre S, Hills M, et al: Proliferation and apoptosis as markers of benefit in neoadjuvant endocrine therapy of breast cancer. Clin Cancer Res. 2006, 12: 1024s-1030s. 10.1158/1078-0432.CCR-05-2127.CrossRefPubMed
7.
Zurück zum Zitat Dowsett M, Smith IE, Ebbs RS, Dixon JM, Skene A, A'Hern R, Salter J, Detre S, Hills M, Walsh G, et al: Prognostic value of Ki67 expression after short-term presurgical endocrine therapy for primary breast cancer. J Natl Cancer Inst. Dowsett M, Smith IE, Ebbs RS, Dixon JM, Skene A, A'Hern R, Salter J, Detre S, Hills M, Walsh G, et al: Prognostic value of Ki67 expression after short-term presurgical endocrine therapy for primary breast cancer. J Natl Cancer Inst.
8.
Zurück zum Zitat Frasor J, Danes JM, Komm B, Chang KC, Lyttle CR, Katzenellenbogen BS: Profiling of estrogen up- and down-regulated gene expression in human breast cancer cells: insights into gene networks and pathways underlying estrogenic control of proliferation and cell phenotype. Endocrinology. 2003, 144: 4562-4574. 10.1210/en.2003-0567.CrossRefPubMed Frasor J, Danes JM, Komm B, Chang KC, Lyttle CR, Katzenellenbogen BS: Profiling of estrogen up- and down-regulated gene expression in human breast cancer cells: insights into gene networks and pathways underlying estrogenic control of proliferation and cell phenotype. Endocrinology. 2003, 144: 4562-4574. 10.1210/en.2003-0567.CrossRefPubMed
9.
Zurück zum Zitat Frasor J, Stossi F, Danes JM, Komm B, Lyttle CR, Katzenellenbogen BS: Selective estrogen receptor modulators: discrimination of agonistic versus antagonistic activities by gene expression profiling in breast cancer cells. Cancer Res. 2004, 64: 1522-1533. 10.1158/0008-5472.CAN-03-3326.CrossRefPubMed Frasor J, Stossi F, Danes JM, Komm B, Lyttle CR, Katzenellenbogen BS: Selective estrogen receptor modulators: discrimination of agonistic versus antagonistic activities by gene expression profiling in breast cancer cells. Cancer Res. 2004, 64: 1522-1533. 10.1158/0008-5472.CAN-03-3326.CrossRefPubMed
10.
Zurück zum Zitat Harvey JM, Clark GM, Osborne CK, Allred DC: Estrogen receptor status by immunohistochemistry is superior to the ligand-binding assay for predicting response to adjuvant endocrine therapy in breast cancer. J Clin Oncol. 1999, 17: 1474-1481.PubMed Harvey JM, Clark GM, Osborne CK, Allred DC: Estrogen receptor status by immunohistochemistry is superior to the ligand-binding assay for predicting response to adjuvant endocrine therapy in breast cancer. J Clin Oncol. 1999, 17: 1474-1481.PubMed
11.
Zurück zum Zitat Cleator SJ, Powles TJ, Dexter T, Fulford L, Mackay A, Smith IE, Valgeirsson H, Ashworth A, Dowsett M: The effect of the stromal component of breast tumours on prediction of clinical outcome using gene expression microarray analysis. Breast Cancer Res. 2006, 8: R32-10.1186/bcr1506.CrossRefPubMedPubMedCentral Cleator SJ, Powles TJ, Dexter T, Fulford L, Mackay A, Smith IE, Valgeirsson H, Ashworth A, Dowsett M: The effect of the stromal component of breast tumours on prediction of clinical outcome using gene expression microarray analysis. Breast Cancer Res. 2006, 8: R32-10.1186/bcr1506.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Tusher VG, Tibshirani R, Chu G: Significance analysis of microarrays applied to the ionizing radiation response. Proc Natl Acad Sci USA. 2001, 98: 5116-5121. 10.1073/pnas.091062498.CrossRefPubMedPubMedCentral Tusher VG, Tibshirani R, Chu G: Significance analysis of microarrays applied to the ionizing radiation response. Proc Natl Acad Sci USA. 2001, 98: 5116-5121. 10.1073/pnas.091062498.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Sorlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, Hastie T, Eisen MB, van de Rijn M, Jeffrey SS, et al: Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci USA. 2001, 98: 10869-10874. 10.1073/pnas.191367098.CrossRefPubMedPubMedCentral Sorlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, Hastie T, Eisen MB, van de Rijn M, Jeffrey SS, et al: Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci USA. 2001, 98: 10869-10874. 10.1073/pnas.191367098.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Murray J, Young O, Renshaw L, White S, Prescot RJ, Krause A, Evans DB, Salem R, Cameron D, Dowsett M, et al: Letrozole and anastrozole: a pre-operative study of their effects on ER positive breast cancers in postmenopausal women. Breast Cancer Res Treat. 2004, S37-Suppl 1 Murray J, Young O, Renshaw L, White S, Prescot RJ, Krause A, Evans DB, Salem R, Cameron D, Dowsett M, et al: Letrozole and anastrozole: a pre-operative study of their effects on ER positive breast cancers in postmenopausal women. Breast Cancer Res Treat. 2004, S37-Suppl 1
15.
Zurück zum Zitat Draghici S, Khatri P, Bhavsar P, Shah A, Krawetz SA, Tainsky MA: Onto-Tools, the toolkit of the modern biologist: Onto-Express, Onto-Compare, Onto-Design and Onto-Translate. Nucleic Acids Res. 2003, 31: 3775-3781. 10.1093/nar/gkg624.CrossRefPubMedPubMedCentral Draghici S, Khatri P, Bhavsar P, Shah A, Krawetz SA, Tainsky MA: Onto-Tools, the toolkit of the modern biologist: Onto-Express, Onto-Compare, Onto-Design and Onto-Translate. Nucleic Acids Res. 2003, 31: 3775-3781. 10.1093/nar/gkg624.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Dai H, van't Veer L, Lamb J, He YD, Mao M, Fine BM, Bernards R, van de Vijver M, Deutsch P, Sachs A, et al: A cell proliferation signature is a marker of extremely poor outcome in a subpopulation of breast cancer patients. Cancer Res. 2005, 65: 4059-4066. 10.1158/0008-5472.CAN-04-3953.CrossRefPubMed Dai H, van't Veer L, Lamb J, He YD, Mao M, Fine BM, Bernards R, van de Vijver M, Deutsch P, Sachs A, et al: A cell proliferation signature is a marker of extremely poor outcome in a subpopulation of breast cancer patients. Cancer Res. 2005, 65: 4059-4066. 10.1158/0008-5472.CAN-04-3953.CrossRefPubMed
17.
Zurück zum Zitat Geisler J, Lonning PE: Endocrine effects of aromatase inhibitors and inactivators in vivo: review of data and method limitations. J Steroid Biochem Mol Biol. 2005, 95: 75-81. 10.1016/j.jsbmb.2005.04.015.CrossRefPubMed Geisler J, Lonning PE: Endocrine effects of aromatase inhibitors and inactivators in vivo: review of data and method limitations. J Steroid Biochem Mol Biol. 2005, 95: 75-81. 10.1016/j.jsbmb.2005.04.015.CrossRefPubMed
18.
Zurück zum Zitat Carroll JS, Meyer CA, Song J, Li W, Geistlinger TR, Eeckhoute J, Brodsky AS, Keeton EK, Fertuck KC, Hall GF, et al: Genome-wide analysis of estrogen receptor binding sites. Nat Genet. 2006, 38: 1289-1297. 10.1038/ng1901.CrossRefPubMed Carroll JS, Meyer CA, Song J, Li W, Geistlinger TR, Eeckhoute J, Brodsky AS, Keeton EK, Fertuck KC, Hall GF, et al: Genome-wide analysis of estrogen receptor binding sites. Nat Genet. 2006, 38: 1289-1297. 10.1038/ng1901.CrossRefPubMed
19.
Zurück zum Zitat Miller WR, Krause A, Evans DB, Renshaw L, Murray J, Larionov A, Anderson TJ, White S, Hampton G, Walker JR, et al: Phenotypes for endocrine resisitance can be identified by RNA microarray of sequential biopsies and are more variable than those predicting for tumour response. Breast Cancer Res Treat. 2005, 94: S17-10.1007/s10549-005-6653-x.CrossRef Miller WR, Krause A, Evans DB, Renshaw L, Murray J, Larionov A, Anderson TJ, White S, Hampton G, Walker JR, et al: Phenotypes for endocrine resisitance can be identified by RNA microarray of sequential biopsies and are more variable than those predicting for tumour response. Breast Cancer Res Treat. 2005, 94: S17-10.1007/s10549-005-6653-x.CrossRef
20.
Zurück zum Zitat Kristensen VN, Sorlie T, Geisler J, Yoshimura N, Linegjaerde OC, Glad I, Frigessi A, Harada N, Lonning PE, Borresen-Dale AL: Effects of anastrozole on the intratumoral gene expression in locally advanced breast cancer. J Steroid Biochem Mol Biol. 2005, 95: 105-111. 10.1016/j.jsbmb.2005.04.028.CrossRefPubMed Kristensen VN, Sorlie T, Geisler J, Yoshimura N, Linegjaerde OC, Glad I, Frigessi A, Harada N, Lonning PE, Borresen-Dale AL: Effects of anastrozole on the intratumoral gene expression in locally advanced breast cancer. J Steroid Biochem Mol Biol. 2005, 95: 105-111. 10.1016/j.jsbmb.2005.04.028.CrossRefPubMed
21.
Zurück zum Zitat Gruvberger S, Ringner M, Chen Y, Panavally S, Saal LH, Borg A, Ferno M, Peterson C, Meltzer PS: Estrogen receptor status in breast cancer is associated with remarkably distinct gene expression patterns. Cancer Res. 2001, 61: 5979-5984.PubMed Gruvberger S, Ringner M, Chen Y, Panavally S, Saal LH, Borg A, Ferno M, Peterson C, Meltzer PS: Estrogen receptor status in breast cancer is associated with remarkably distinct gene expression patterns. Cancer Res. 2001, 61: 5979-5984.PubMed
22.
Zurück zum Zitat van 't Veer LJ, Dai H, van de Vijver MJ, He YD, Hart AA, Mao M, Peterse HL, van der Kooy 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, van de Vijver MJ, He YD, Hart AA, Mao M, Peterse HL, van der Kooy 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
23.
Zurück zum Zitat Perou CM, Sorlie T, Eisen MB, van de Rijn 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, van de Rijn 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
24.
Zurück zum Zitat Konecny GE, Thomssen C, Luck HJ, Untch M, Wang HJ, Kuhn W, Eidtmann H, du Bois A, Olbricht S, Steinfeld D, et al: Her-2/neu gene amplification and response to paclitaxel in patients with metastatic breast cancer. J Natl Cancer Inst. 2004, 96: 1141-1151.CrossRefPubMed Konecny GE, Thomssen C, Luck HJ, Untch M, Wang HJ, Kuhn W, Eidtmann H, du Bois A, Olbricht S, Steinfeld D, et al: Her-2/neu gene amplification and response to paclitaxel in patients with metastatic breast cancer. J Natl Cancer Inst. 2004, 96: 1141-1151.CrossRefPubMed
25.
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
26.
Zurück zum Zitat Dowsett M, Smith IE, Ebbs SR, Dixon JM, Skene A, Griffith C, Boeddinghaus I, Salter J, Detre S, Hills M, et al: Short-term changes in Ki-67 during neoadjuvant treatment of primary breast cancer with anastrozole or tamoxifen alone or combined correlate with recurrence-free survival. Clin Cancer Res. 2005, 11: 951s-958s.PubMed Dowsett M, Smith IE, Ebbs SR, Dixon JM, Skene A, Griffith C, Boeddinghaus I, Salter J, Detre S, Hills M, et al: Short-term changes in Ki-67 during neoadjuvant treatment of primary breast cancer with anastrozole or tamoxifen alone or combined correlate with recurrence-free survival. Clin Cancer Res. 2005, 11: 951s-958s.PubMed
27.
Zurück zum Zitat Mrusek S, Classen-Linke I, Vloet A, Beier HM, Krusche CA: Estradiol and medroxyprogesterone acetate regulated genes in T47D breast cancer cells. Mol Cell Endocrinol. 2005, 235: 39-50. 10.1016/j.mce.2005.01.009.CrossRefPubMed Mrusek S, Classen-Linke I, Vloet A, Beier HM, Krusche CA: Estradiol and medroxyprogesterone acetate regulated genes in T47D breast cancer cells. Mol Cell Endocrinol. 2005, 235: 39-50. 10.1016/j.mce.2005.01.009.CrossRefPubMed
28.
Zurück zum Zitat Cicatiello L, Scafoglio C, Altucci L, Cancemi M, Natoli G, Facchiano A, Iazzetti G, Calogero R, Biglia N, De Bortoli M, et al: A genomic view of estrogen actions in human breast cancer cells by expression profiling of the hormone-responsive transcriptome. J Mol Endocrinol. 2004, 32: 719-775. 10.1677/jme.0.0320719.CrossRefPubMed Cicatiello L, Scafoglio C, Altucci L, Cancemi M, Natoli G, Facchiano A, Iazzetti G, Calogero R, Biglia N, De Bortoli M, et al: A genomic view of estrogen actions in human breast cancer cells by expression profiling of the hormone-responsive transcriptome. J Mol Endocrinol. 2004, 32: 719-775. 10.1677/jme.0.0320719.CrossRefPubMed
29.
Zurück zum Zitat Itoh T, Karlsberg K, Kijima I, Yuan YC, Smith D, Ye J, Chen S: Letrozole-, anastrozole-, and tamoxifen-responsive genes in MCF-7aro cells: a microarray approach. Mol Cancer Res. 2005, 3: 203-218.PubMed Itoh T, Karlsberg K, Kijima I, Yuan YC, Smith D, Ye J, Chen S: Letrozole-, anastrozole-, and tamoxifen-responsive genes in MCF-7aro cells: a microarray approach. Mol Cancer Res. 2005, 3: 203-218.PubMed
30.
Zurück zum Zitat Kian Tee M, Rogatsky I, Tzagarakis-Foster C, Cvoro A, An J, Christy RJ, Yamamoto KR, Leitman DC: Estradiol and selective estrogen receptor modulators differentially regulate target genes with estrogen receptors alpha and beta. Mol Biol Cell. 2004, 15: 1262-1272. 10.1091/mbc.E03-06-0360.CrossRefPubMedCentral Kian Tee M, Rogatsky I, Tzagarakis-Foster C, Cvoro A, An J, Christy RJ, Yamamoto KR, Leitman DC: Estradiol and selective estrogen receptor modulators differentially regulate target genes with estrogen receptors alpha and beta. Mol Biol Cell. 2004, 15: 1262-1272. 10.1091/mbc.E03-06-0360.CrossRefPubMedCentral
31.
Zurück zum Zitat Harvell DM, Richer JK, Allred DC, Sartorius CA, Horwitz KB: Estradiol regulates different genes in human breast tumor xenografts compared with the identical cells in culture. Endocrinology. 2006, 147: 700-713. 10.1210/en.2005-0617.CrossRefPubMed Harvell DM, Richer JK, Allred DC, Sartorius CA, Horwitz KB: Estradiol regulates different genes in human breast tumor xenografts compared with the identical cells in culture. Endocrinology. 2006, 147: 700-713. 10.1210/en.2005-0617.CrossRefPubMed
32.
Zurück zum Zitat Wilson CL, Sims AH, Howell A, Miller CJ, Clarke RB: Effects of oestrogen on gene expression in epithelium and stroma of normal human breast tissue. Endocr Relat Cancer. 2006, 13: 617-628. 10.1677/erc.1.01165.CrossRefPubMed Wilson CL, Sims AH, Howell A, Miller CJ, Clarke RB: Effects of oestrogen on gene expression in epithelium and stroma of normal human breast tissue. Endocr Relat Cancer. 2006, 13: 617-628. 10.1677/erc.1.01165.CrossRefPubMed
33.
Zurück zum Zitat Jin VX, Leu YW, Liyanarachchi S, Sun H, Fan M, Nephew KP, Huang TH, Davuluri RV: Identifying estrogen receptor alpha target genes using integrated computational genomics and chromatin immunoprecipitation microarray. Nucleic Acids Res. 2004, 32: 6627-6635. 10.1093/nar/gkh1005.CrossRefPubMedPubMedCentral Jin VX, Leu YW, Liyanarachchi S, Sun H, Fan M, Nephew KP, Huang TH, Davuluri RV: Identifying estrogen receptor alpha target genes using integrated computational genomics and chromatin immunoprecipitation microarray. Nucleic Acids Res. 2004, 32: 6627-6635. 10.1093/nar/gkh1005.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Nunez AM, Jakowlev S, Briand JP, Gaire M, Krust A, Rio MC, Chambon P: Characterization of the estrogen-induced pS2 protein secreted by the human breast cancer cell line MCF-7. Endocrinology. 1987, 121: 1759-1765.CrossRefPubMed Nunez AM, Jakowlev S, Briand JP, Gaire M, Krust A, Rio MC, Chambon P: Characterization of the estrogen-induced pS2 protein secreted by the human breast cancer cell line MCF-7. Endocrinology. 1987, 121: 1759-1765.CrossRefPubMed
35.
Zurück zum Zitat Coser KR, Chesnes J, Hur J, Ray S, Isselbacher KJ, Shioda T: Global analysis of ligand sensitivity of estrogen inducible and suppressible genes in MCF7/BUS breast cancer cells by DNA microarray. Proc Natl Acad Sci USA. 2003, 100: 13994-13999. 10.1073/pnas.2235866100.CrossRefPubMedPubMedCentral Coser KR, Chesnes J, Hur J, Ray S, Isselbacher KJ, Shioda T: Global analysis of ligand sensitivity of estrogen inducible and suppressible genes in MCF7/BUS breast cancer cells by DNA microarray. Proc Natl Acad Sci USA. 2003, 100: 13994-13999. 10.1073/pnas.2235866100.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Carroll JS, Brown M: Estrogen receptor target gene: an evolving concept. Mol Endocrinol. 2006, 20: 1707-1714. 10.1210/me.2005-0334.CrossRefPubMed Carroll JS, Brown M: Estrogen receptor target gene: an evolving concept. Mol Endocrinol. 2006, 20: 1707-1714. 10.1210/me.2005-0334.CrossRefPubMed
37.
Zurück zum Zitat Soulez M, Parker MG: Identification of novel oestrogen receptor target genes in human ZR75-1 breast cancer cells by expression profiling. J Mol Endocrinol. 2001, 27: 259-274. 10.1677/jme.0.0270259.CrossRefPubMed Soulez M, Parker MG: Identification of novel oestrogen receptor target genes in human ZR75-1 breast cancer cells by expression profiling. J Mol Endocrinol. 2001, 27: 259-274. 10.1677/jme.0.0270259.CrossRefPubMed
38.
Zurück zum Zitat Oh DS, Troester MA, Usary J, Hu Z, He X, Fan C, Wu J, Carey LA, Perou CM: Estrogen-regulated genes predict survival in hormone receptor-positive breast cancers. J Clin Oncol. 2006, 24: 1656-1664. 10.1200/JCO.2005.03.2755.CrossRefPubMed Oh DS, Troester MA, Usary J, Hu Z, He X, Fan C, Wu J, Carey LA, Perou CM: Estrogen-regulated genes predict survival in hormone receptor-positive breast cancers. J Clin Oncol. 2006, 24: 1656-1664. 10.1200/JCO.2005.03.2755.CrossRefPubMed
39.
Zurück zum Zitat Innes HE, Liu D, Barraclough R, Davies MP, O'Neill PA, Platt-Higgins A, de Silva Rudland S, Sibson DR, Rudland PS: Significance of the metastasis-inducing protein AGR2 for outcome in hormonally treated breast cancer patients. Br J Cancer. 2006, 94: 1057-1065. 10.1038/sj.bjc.6603065.CrossRefPubMedPubMedCentral Innes HE, Liu D, Barraclough R, Davies MP, O'Neill PA, Platt-Higgins A, de Silva Rudland S, Sibson DR, Rudland PS: Significance of the metastasis-inducing protein AGR2 for outcome in hormonally treated breast cancer patients. Br J Cancer. 2006, 94: 1057-1065. 10.1038/sj.bjc.6603065.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Prosperi JR, Robertson FM: Cyclooxygenase-2 directly regulates gene expression of P450 Cyp19 aromatase promoter regions pII, pI.3 and pI.7 and estradiol production in human breast tumor cells. Prostaglandins Other Lipid Mediat. 2006, 81: 55-70. 10.1016/j.prostaglandins.2006.07.003.CrossRefPubMed Prosperi JR, Robertson FM: Cyclooxygenase-2 directly regulates gene expression of P450 Cyp19 aromatase promoter regions pII, pI.3 and pI.7 and estradiol production in human breast tumor cells. Prostaglandins Other Lipid Mediat. 2006, 81: 55-70. 10.1016/j.prostaglandins.2006.07.003.CrossRefPubMed
41.
Zurück zum Zitat Naito Z: Role of the small leucine-rich proteoglycan (SLRP) family in pathological lesions and cancer cell growth. J Nippon Med Sch. 2005, 72: 137-145. 10.1272/jnms.72.137.CrossRefPubMed Naito Z: Role of the small leucine-rich proteoglycan (SLRP) family in pathological lesions and cancer cell growth. J Nippon Med Sch. 2005, 72: 137-145. 10.1272/jnms.72.137.CrossRefPubMed
42.
Zurück zum Zitat Yanaihara A, Otsuka Y, Iwasaki S, Aida T, Tachikawa T, Irie T, Okai T: Differences in gene expression in the proliferative human endometrium. Fertil Steril. 2005, 1206-1215. 10.1016/j.fertnstert.2004.11.032. Suppl 1 Yanaihara A, Otsuka Y, Iwasaki S, Aida T, Tachikawa T, Irie T, Okai T: Differences in gene expression in the proliferative human endometrium. Fertil Steril. 2005, 1206-1215. 10.1016/j.fertnstert.2004.11.032. Suppl 1
43.
Zurück zum Zitat Hong SH, Nah HY, Lee JY, Gye MC, Kim CH, Kim MK: Analysis of estrogen-regulated genes in mouse uterus using cDNA microarray and laser capture microdissection. J Endocrinol. 2004, 181: 157-167. 10.1677/joe.0.1810157.CrossRefPubMed Hong SH, Nah HY, Lee JY, Gye MC, Kim CH, Kim MK: Analysis of estrogen-regulated genes in mouse uterus using cDNA microarray and laser capture microdissection. J Endocrinol. 2004, 181: 157-167. 10.1677/joe.0.1810157.CrossRefPubMed
44.
Zurück zum Zitat Troup S, Njue C, Kliewer EV, Parisien M, Roskelley C, Chakravarti S, Roughley PJ, Murphy LC, Watson PH: Reduced expression of the small leucine-rich proteoglycans, lumican, and decorin is associated with poor outcome in node-negative invasive breast cancer. Clin Cancer Res. 2003, 9: 207-214.PubMed Troup S, Njue C, Kliewer EV, Parisien M, Roskelley C, Chakravarti S, Roughley PJ, Murphy LC, Watson PH: Reduced expression of the small leucine-rich proteoglycans, lumican, and decorin is associated with poor outcome in node-negative invasive breast cancer. Clin Cancer Res. 2003, 9: 207-214.PubMed
45.
Zurück zum Zitat Choi Y, Pinto M: Estrogen receptor beta in breast cancer: associations between ERbeta, hormonal receptors, and other prognostic biomarkers. Appl Immunohistochem Mol Morphol. 2005, 13: 19-24. 10.1097/00129039-200503000-00004.CrossRefPubMed Choi Y, Pinto M: Estrogen receptor beta in breast cancer: associations between ERbeta, hormonal receptors, and other prognostic biomarkers. Appl Immunohistochem Mol Morphol. 2005, 13: 19-24. 10.1097/00129039-200503000-00004.CrossRefPubMed
46.
Zurück zum Zitat Hsieh YC, Choudhry MA, Yu HP, Shimizu T, Yang S, Suzuki T, Chen J, Bland KI, Chaudry IH: Inhibition of cardiac PGC-1alpha expression abolishes ERbeta agonist-mediated cardioprotection following trauma-hemorrhage. FASEB J. 2006, 20: 1109-1117. 10.1096/fj.05-5549com.CrossRefPubMed Hsieh YC, Choudhry MA, Yu HP, Shimizu T, Yang S, Suzuki T, Chen J, Bland KI, Chaudry IH: Inhibition of cardiac PGC-1alpha expression abolishes ERbeta agonist-mediated cardioprotection following trauma-hemorrhage. FASEB J. 2006, 20: 1109-1117. 10.1096/fj.05-5549com.CrossRefPubMed
47.
Zurück zum Zitat Jones C, Mackay A, Grigoriadis A, Cossu A, Reis-Filho JS, Fulford L, Dexter T, Davies S, Bulmer K, Ford E, et al: Expression profiling of purified normal human luminal and myoepithelial breast cells: identification of novel prognostic markers for breast cancer. Cancer Res. 2004, 64: 3037-3045. 10.1158/0008-5472.CAN-03-2028.CrossRefPubMed Jones C, Mackay A, Grigoriadis A, Cossu A, Reis-Filho JS, Fulford L, Dexter T, Davies S, Bulmer K, Ford E, et al: Expression profiling of purified normal human luminal and myoepithelial breast cells: identification of novel prognostic markers for breast cancer. Cancer Res. 2004, 64: 3037-3045. 10.1158/0008-5472.CAN-03-2028.CrossRefPubMed
48.
Zurück zum Zitat Grigoriadis A, Mackay A, Reis-Filho JS, Steele D, Iseli C, Stevenson B, Jongeneel CV, Valgeirsson H, Fenwick K, Iravani M, et al: Establishment of the epithelial-specific transcriptome of normal and malignant human breast cells based on MPSS and array expression data. Breast Cancer Res. 2006, 8: R56-10.1186/bcr1604.CrossRefPubMedPubMedCentral Grigoriadis A, Mackay A, Reis-Filho JS, Steele D, Iseli C, Stevenson B, Jongeneel CV, Valgeirsson H, Fenwick K, Iravani M, et al: Establishment of the epithelial-specific transcriptome of normal and malignant human breast cells based on MPSS and array expression data. Breast Cancer Res. 2006, 8: R56-10.1186/bcr1604.CrossRefPubMedPubMedCentral
49.
Zurück zum Zitat Shioda T, Chesnes J, Coser KR, Zou L, Hur J, Dean KL, Sonnenschein C, Soto AM, Isselbacher KJ: Importance of dosage standardization for interpreting transcriptomal signature profiles: evidence from studies of xenoestrogens. Proc Natl Acad Sci USA. 2006, 103: 12033-12038. 10.1073/pnas.0605341103.CrossRefPubMedPubMedCentral Shioda T, Chesnes J, Coser KR, Zou L, Hur J, Dean KL, Sonnenschein C, Soto AM, Isselbacher KJ: Importance of dosage standardization for interpreting transcriptomal signature profiles: evidence from studies of xenoestrogens. Proc Natl Acad Sci USA. 2006, 103: 12033-12038. 10.1073/pnas.0605341103.CrossRefPubMedPubMedCentral
Metadaten
Titel
Molecular response to aromatase inhibitor treatment in primary breast cancer
verfasst von
Alan Mackay
Ander Urruticoechea
J Michael Dixon
Tim Dexter
Kerry Fenwick
Alan Ashworth
Suzanne Drury
Alexey Larionov
Oliver Young
Sharon White
William R Miller
Dean B Evans
Mitch Dowsett
Publikationsdatum
01.06.2007
Verlag
BioMed Central
Erschienen in
Breast Cancer Research / Ausgabe 3/2007
Elektronische ISSN: 1465-542X
DOI
https://doi.org/10.1186/bcr1732

Weitere Artikel der Ausgabe 3/2007

Breast Cancer Research 3/2007 Zur Ausgabe

Update Onkologie

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