Skip to main content
Erschienen in: Breast Cancer Research 1/2009

Open Access 01.06.2009 | Research article

Gene expression profiling of the tumor microenvironment during breast cancer progression

verfasst von: Xiao-Jun Ma, Sonika Dahiya, Elizabeth Richardson, Mark Erlander, Dennis C Sgroi

Erschienen in: Breast Cancer Research | Ausgabe 1/2009

Abstract

Introduction

The importance of the tumor microenvironment in breast cancer has been increasingly recognized. Critical molecular changes in the tumor stroma accompanying cancer progression, however, remain largely unknown. We conducted a comparative analysis of global gene expression changes in the stromal and epithelial compartments during breast cancer progression from normal to preinvasive to invasive ductal carcinoma.

Methods

We combined laser capture microdissection and gene expression microarrays to analyze 14 patient-matched normal epithelium, normal stroma, tumor epithelium and tumor-associated stroma specimens. Differential gene expression and gene ontology analyses were performed.

Results

Tumor-associated stroma undergoes extensive gene expression changes during cancer progression, to a similar extent as that seen in the malignant epithelium. Highly upregulated genes in the tumor-associated stroma include constituents of the extracellular matrix and matrix metalloproteases, and cell-cycle-related genes. Decreased expression of cytoplasmic ribosomal proteins and increased expression of mitochondrial ribosomal proteins were observed in both the tumor epithelium and the stroma. The transition from preinvasive to invasive growth was accompanied by increased expression of several matrix metalloproteases (MMP2, MMP11 and MMP14). Furthermore, as observed in malignant epithelium, a gene expression signature of histological tumor grade also exists in the stroma, with high-grade tumors associated with increased expression of genes involved in immune response.

Conclusions

Our results suggest that the tumor microenvironment participates in tumorigenesis even before tumor cells invade into stroma, and that it may play important roles in the transition from preinvasive to invasive growth. The immune cells in the tumor stroma may be exploited by the malignant epithelial cells in high-grade tumors for aggressive invasive growth.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​bcr2222) contains supplementary material, which is available to authorized users.

Competing interests

XJM and ME are employees of BioTheranostics, Inc. ME, X-JM and DCS are named inventors on a patent application relating to the contents of the manuscript.

Authors' contributions

DCS, XJM and ME conceived of the study, participated in its design and coordination, and helped draft the manuscript. DCS and SD performed microdissection and RNA extraction. XJM and ME performed the microarray experiments and microarray data analysis. ER performed the real-time PCR assays. All authors have read and approved the final manuscript.
Abkürzungen
CXCL
chemokine (C-X-C motif) ligand
DCIS
ductal carcinoma in situ
DCIS-S
DCIS-associated stroma
GREM1
gremlin 1
IDC
invasive ductal carcinoma
IDC-S
IDC-associated stroma
INHBA
inhibin beta A
LCM
laser capture microdissection
PCR
polymerase chain reaction
SFRP1
secreted frizzled-related protein 1
WIF1
WNT inhibitory factor 1.

Introduction

The tumor microenvironment or the stroma hosting the malignant breast epithelial cells is comprised of multiple cell types, including fibroblasts, myoepithelial cells, endothelial cells and various immune cells [14]. One prevailing view is that tumor-associated stroma is activated by the malignant epithelial cells to foster tumor growth – for example, by secreting growth factors, increasing angiogenesis, and facilitating cell migration, ultimately resulting in metastasis to remote organ sites [3]. For example, two chemokines (chemokine (C-X-C motif) ligand (CXCL) 12 and CXCL14) that bind to tumor epithelial cells to promote proliferation, migration and invasion have recently been shown to be overexpressed by the activated tumor fibroblasts and myoepithelial cells [57]. Genes involved in tumor-microenvironment interactions may therefore provide novel targets for diagnostic development and therapeutic intervention. Our understanding of the interactions between epithelial and stromal components of breast cancer, however, remains limited at the molecular level. Using the serial analysis of gene expression technique, Allinen and coworkers performed the first systematic profiling of the various stromal cell types isolated via cell-type-specific cell surface markers and magnetic beads [7]. They demonstrated gene expression alterations in all cell types within the tumor microenvironment accompanying progression from normal breast tissue to ductal carcinoma in situ (DCIS) to invasive ducal carcinoma (IDC) [8], providing evidence that these cell types all participate in tumorigenesis.
Using laser capture microdissection (LCM), we previously performed gene expression analysis of the epithelial compartment of the malignant lesions during breast cancer progression. We discovered that most of the gene expression changes take place prior to local invasion (even in atypical ductal hyperplasia) and that there are no major changes in gene expression accompanying the in situ to invasive growth transition [9]. In the present article we extend this analysis to the tumor stromal microenvironment and demonstrate that, like the tumor epithelium, the tumor stromal microenvironment undergoes extensive gene expression alterations even at the preinvasive stage of DCIS, supporting the view that cell-cell communication via paracrine mechanisms between the two compartments plays an important role in tumor progression.

Materials and methods

Clinical specimen

All breast cancer specimens were fresh-frozen biopsies obtained from the Massachusetts General Hospital between 1998 and 2001. The diagnostic criteria and tumor grading were described previously [9]. Patient and tumor characteristics of the 14 tumor specimens in this study are presented in Table 1. Patients were selected in which patient-matched normal and tumor samples were available and the normal breast lobules did not show fibrocystic change. The research was deemed exempt from informed consent as the samples are unidentifiable to the research team. The study was approved by the Massachusetts General Hospital human research committee in accordance with National Institutes of Health human research study guidelines.
Table 1
Patient and tumor characteristics of samples in the study
Patient number
Age (years)
Grade
Estrogen receptor
Progesterone receptor
Her-2
Size
Nodal status
Tumor type
44
28
III
Positive
Positive
Negative
1
Negative
Ductal
45
36
I
Positive
Positive
Negative
N/A
Negative
Ductal
79
54
I
Positive
Positive
Negative
2.1
Positive
Ductal
96
31
III
Negative
Negative
Negative
3.7
Negative
Ductal
102
55
I
Positive
Negative
Negative
5.2
Positive
Ductal
121
45
II
Positive
Positive
Positive
1.5
Positive
Ductal
131
37
II
Positive
Positive
Positive
1.5
Positive
Ductal
133
44
III
Negative
Negative
Positive
1.5
Positive
Ductal
148
42
II
Positive
Positive
Negative
1.9
Positive
Ductal
153
46
I
Positive
Positive
ND
N/A
Positive
Ductal
169
34
II
Positive
Positive
Negative
2.6
Positive
Ductal
178
43
III
Positive
Positive
Positive
2.8
Positive
Ductal
179
37
III
Negative
Negative
Positive
1.5
Positive
Ductal
180
46
I
Positive
Positive
Negative
1.9
Positive
Ductal
ND, not determined; N/A, not available.

Laser capture microdissection, RNA extraction and microarray analysis

Highly enriched populations of patient-matched normal or malignant epithelial cells and of normal stroma or tumor-associated stroma from the different stages of breast cancer progression were procured by LCM using a PixCell IIe system (Molecular Devices, Mountain View, CA, USA) as previously described [9]. Enrichment for cells of interest was verified by microscopic examination of the LCM cap after microdissection. The microdissected normal stromal compartment consisted of the intralobular, rather than the extralobular, stromal compartment of normal breast tissue that was a minimum 0.3 cm from any premalignant or malignant lesion (Figure 1). The DCIS-associated stroma (DCIS-S) consisted of a 25 μm rim of cells that surrounded the DCIS; for cases in which synchronous DCIS and IDC were present, the DCIS-S was obtained from areas of DCIS that were at least 0.3 cm from the invasive component. The IDC-associated stroma (IDC-S) consists of stromal cells predominantly within the invasive tumor mass.
Total RNA was isolated from captured cells using the Picopure™ RNA isolation kit (Molecular Devices), amplified by T7 RNA amplification (RiboAmp™; Molecular Devices), labeled and hybridized to the whole genome array U133X3P (3'-biased design) according to the manufacturer's instructions (Affymetrix, Santa Clara, CA, USA). The hybridized microarrays were then washed, stained and scanned as per the manufacturer's protocols (Affymetrix).

Data analysis

Raw data from the U133X3P arrays were processed using the Bioconductor rma package with default parameters for background correction, quantile normalization and signal summation [10, 11]. Differential gene expression analyses were performed using linear regression models in the limma package [12]. For comparing normal and tumor samples, we used the patient identification as a blocking variable. For tumor grade comparison, we used the tumor stage (in situ or invasive) as the blocking variable. Statistical significance was corrected for multiple testing using the Benjamini-Hochberg procedure [13]. All procedures were performed in the R statistical environment [14]. For gene ontology analysis, ranked gene lists were first generated according to the moderated t statistics from linear models and then examined for enriched ontology terms using the Gene Set Enrichment Analysis software [15]. The data discussed in this publication have been deposited in the NCBI Gene Expression Omnibus [16] and are accessible [GEO:GSE14548] [17].

Quantitative real-time PCR and immunohistochemistry

TaqMan™ real-time PCR was performed on amplified RNA used for microarray analysis as previously described [9]. Briefly, amplified RNA was converted to double-stranded cDNA, and the cDNA was quantitated with PicoGreen (Molecular Probes, Eugene, OR, USA) using a spectrofluorometer (Molecular Devices). Each gene was analyzed in triplicate in a 96-well plate using ABI 7900 HT (Applied Biosystems, Foster City, CA., USA).
For each gene, the sequences of the PCR primer pairs and the fluorogenic MGB or TAMRA probe (5' to 3'), respectively, are as follows: ESR1, ATGATCAACTGGGCGAAGA, GGTGGACCTGATCATGGA and VIC-TGCCAGGCTTTGTGGA; RRM2, CCTTTAACCAGCACAGCCAGTT, TTATTTGTTTGTAAAGTGCCAGGTTT and VIC-TGCAGCCTCACTGCTTCAACGCA-TAMRA; gremlin 1 (GREM1), ACGGCAAAGAATTATATAGACTATGAGGTA, TTTTATGAGACTATCAACTCCCCTTTC and VIC-CTTGCTGTGTAGGAGGA; and WNT inhibitory factor 1(WIF1), CACTGTGGTAGTGGCATTTAAACAATA, GCCAATGCAAAAAGTTCATACATT and VIC-TTCTAAACACAATGAAATAGGGA.
Estrogen receptor and progesterone receptor immunohistochemistry staining was performed as previously described, using the rabbit monoclonal antibody (SP1) from Lab Vision (Fremont, CA, USA) for the estrogen receptor (1:50 dilution) and using the mouse monoclonal antibody (PgR 636) from Dako (Carpinteria, CA, USA) for the progesterone receptor (1:50 dilution) [18].

Results

Experimental design

The present study included 14 patients with primary ductal breast cancer (Table 1). These patients were primarily estrogen receptor positive (78.6%), lymph node positive (78.6%), and premenopausal (mean age 41 years). We used LCM to isolate the epithelial and stroma compartments separately from each of the 14 fresh-frozen biopsies. In the epithelial compartment, we captured normal and malignant epithelium from DCIS and/or IDC. In the stromal compartment, we captured normal stroma at least 3 mm from the malignant lesion and the DCIS-S and/or IDC-S whenever possible. An example of the microdissected compartments is shown in Figure 1. As shown in Table 2, in the epithelial compartment four cases had all three stages (normal breast epithelium, DCIS, and IDC) available, five cases had normal breast epithelium and IDC only, and five cases had normal breast epithelium and DCIS only; in the stroma, six cases had all three stages available, five cases had normal stromal compartment and DCIS-S, and three cases had the normal stromal compartment and IDC-S. RNA was isolated from the captured cells and interrogated with the Affymetrix whole-genome array U133X3P.
Table 2
Laser capture microdissection of 14 primary breast cancer patients
 
Tumor
Stroma
Patient
Normal
In situ
Invasive
Normal
In situ
Invasive
44
x
x
x
x
x
x
45
x
 
x
x
x
x
79
x
 
x
x
x
x
96
x
x
x
x
x
x
102
x
x
x
x
x
x
121
x
x
x
x
x
x
131
x
x
 
x
x
 
133
x
x
 
x
x
 
148
x
 
x
x
 
x
153
x
 
x
x
 
x
169
x
 
x
x
 
x
178
x
x
 
x
x
 
179
x
x
 
x
x
 
180
x
x
 
x
x
 
x, component captured.

Gene expression changes in the stromal and epithelial compartments during breast cancer progression

We compared the gene expression patterns of the tumor epithelium and stroma at each stage of progression (DCIS or IDC) with their respective normal state using the limma (linear models of microarrays) software package [12]. The resulting P values for differential gene expression in each pair-wise comparison were adjusted for multiple testing [13], and the genes with a significant adjusted P value (P <0.05) were extracted.
The DCIS and IDC stages were each associated with thousands of gene expression alterations relative to their respective normal state in both the tumor epithelium and the stroma (Figure 2). Furthermore, within each compartment, the expression patterns of DCIS-associated and IDC-associated genes were highly similar to each other (Figure 3).
To gain an overview of the biological processes in which these differentially expressed genes are involved, we performed gene set enrichment analysis [19] using the gene ontology database [20]. Table 3 presents the top 20 gene ontology terms significantly enriched within genes upregulated in the invasive stage in the epithelium and the stroma. In the epithelium, the genes were dominated by those associated with the cell cycle (mitosis in particular). In the stroma, the genes prominently featured the components of the extracellular matrix and the matrix metalloproteases responsible for remodeling the extracellular matrix. Additionally, the stromal genes also included those related to the cell cycle, indicating increased proliferation as a common feature in both the tumor epithelium and the stroma.
Table 3
Top 20 gene ontology terms enriched in tumor epithelium and stroma
Name
Size (number of genes)
Normalized enrichment score
False discovery rate q value
Epithelium
   
   SPINDLE
39
2.33
0
   CHROMOSOME_SEGREGATION
28
2.15
0
   CELL_CYCLE_PROCESS
180
2.12
0
MICROTUBULE_CYTOSKELETON_ORGANIZATION_AND_BIOGENESIS
34
2.11
0
   CHROMOSOME__PERICENTRIC_REGION
27
2.11
0
   MICROTUBULE_CYTOSKELETON
142
2.11
0
   PROTEASOME_COMPLEX
22
2.09
1.40 × 10-4
   CONDENSED_CHROMOSOME
30
2.06
2.48 × 10-4
   M_PHASE
105
2.06
2.20 × 10-4
   NUCLEAR_ENVELOPE
71
2.05
1.98 × 10-4
   CELL_CYCLE_PHASE
157
2.05
1.80 × 10-4
   M_PHASE_OF_MITOTIC_CELL_CYCLE
78
2.04
2.49 × 10-4
   CHROMOSOME
115
2.03
2.30 × 10-4
   CYTOSKELETAL_PART
221
2.03
2.14 × 10-4
   MITOSIS
75
2.02
2.66 × 10-4
   MICROTUBULE
32
1.99
2.49 × 10-4
   MITOTIC_CELL_CYCLE
139
1.99
2.35 × 10-4
   CELL_CYCLE_CHECKPOINT_GO_0000075
45
1.98
2.21 × 10-4
   SPINDLE_MICROTUBULE
16
1.97
2.63 × 10-4
   DNA_REPAIR
120
1.94
6.01 × 10-4
   STRUCTURAL_CONSTITUENT_OF_RIBOSOME
74
-3.09
0
Stroma
   
EXTRACELLULAR_MATRIX_STRUCTURAL_CONSTITUENT
25
2.12
0
   COLLAGEN
23
2.07
0.001566
   METALLOENDOPEPTIDASE_ACTIVITY
26
2.06
0.001044
   EXTRACELLULAR_MATRIX
94
1.99
0.001568
   PROTEINACEOUS_EXTRACELLULAR_MATRIX
93
1.97
0.002923
   EXTRACELLULAR_MATRIX_PART
54
1.91
0.007826
   SPINDLE
39
1.89
0.008346
   METALLOPEPTIDASE_ACTIVITY
45
1.82
0.027006
   SKELETAL_DEVELOPMENT
99
1.80
0.032482
   STRUCTURAL_CONSTITUENT_OF_RIBOSOME
74
-3.04
0
In both compartments, the single gene ontology term STRUCTURAL_CONSTITUENT_OF_RIBOSOME was significantly enriched within the downregulated genes (Table 3). To examine this further, we extracted all ribosomal protein-encoding genes that were differentially expressed between DCIS or IDC versus the normal breast in the epithelium and visualized their expression patterns in both compartments. Interestingly, there was an almost complete bipartite partitioning of these genes (Figure 4): while the downregulated genes were all those encoding for the cytoplasmic ribosomal proteins, the upregulated genes were mostly those encoding for the mitochondrial ribosomal proteins.
In addition to these global patterns, Tables 4 and 5 present the top 50 differentially expressed genes in the epithelium and the stroma, respectively. In these tables, besides the dominant features of cell-cycle-related genes in the epithelium and extracellular matrix genes in the stroma discussed earlier, we note several additional genes important in cell signaling pathways. Two antagonists of WNT receptor signaling, WIF1 and secreted frizzled-related protein 1 (SFRP1), were downregulated in both the tumor epithelium and the stroma. In addition, two members of the transforming growth factor beta superfamily, GREM1 and inhibin beta A (INHBA), showed markedly increased expression specifically in the tumor stroma (Table 5).
Table 4
Top 50 genes differentially expressed in tumor epithelium
Probe set
DICS
IDC
Gene description
g5174662_3p_at
5.3
4.0
S100P – S100 calcium binding protein P
g11993936_3p_s_at
4.5
3.4
CYB561 – cytochrome b-561
g7415720_3p_a_at
4.0
3.0
SCD – stearoyl-CoA desaturase (delta-9-desaturase)
Hs.75319.0.S3_3p_at
3.0
3.8
RRM2 – ribonucleotide reductase M2 polypeptide
Hs.106552.0.S3_3p_s_at
4.2
2.3
CNTNAP2 – contactin associated protein-like 2
g12803628_3p_at
3.7
2.6
HIST1H1C – histone cluster 1, H1c
g5031780_3p_at
4.0
2.3
IFI27 – interferon, alpha-inducible protein 27
Hs.180779.1.S1_3p_at
3.1
3.0
HIST1H2BD – histone cluster 1, H2bd
Hs.184572.0.S2_3p_at
2.7
3.3
CDC2 – cell division cycle 2, G1 to S and G2 to M
Hs.223025.0.S2_3p_a_at
2.6
3.4
RAB31 – RAB31, member RAS oncogene family
g12804874_3p_a_at
2.8
3.1
RRM2 – ribonucleotide reductase M2 polypeptide
Hs.239884.0.S1_3p_x_at
3.5
2.4
HIST1H2BC – histone cluster 1, H2bc
g7661973_3p_at
2.7
3.1
MELK – maternal embryonic leucine zipper kinase
g13259549_3p_at
3.2
2.4
IFI6 – interferon, alpha-inducible protein 6
g4504584_3p_at
3.5
2.0
IFIT1 – interferon-induced protein with tetratricopeptide repeats 1
Hs.155956.0.S1_3p_at
2.6
2.8
NAT1 – N-acetyltransferase 1 (arylamine N-acetyltransferase)
Hs.152677.0.S1_3p_at
3.1
2.3
DHRS2 – dehydrogenase/reductase (SDR family) member 2
Hs.239884.0.S1_3p_at
3.3
2.1
HIST1H2BC – histone cluster 1, H2bc
g5803130_3p_a_at
2.2
3.2
RAB31 – RAB31, member RAS oncogene family
g13699814_3p_s_at
2.7
2.7
CYP2B6 – cytochrome P450, family 2, subfamily B, polypeptide 6
g9963780_3p_a_at
2.2
3.2
RAB31 – RAB31, member RAS oncogene family
Hs.72472.0.A1_3p_at
2.3
2.9
-
g13477106_3p_s_at
3.3
1.8
CEACAM6 – carcinoembryonic antigen-related cell adhesion molecule 6 (nonspecific cross-reacting antigen)
Hs.133342.0.S1_3p_x_at
2.9
2.2
GPC1 – glypican 1
Hs.325335.1.S1_3p_at
2.6
2.5
CAPN13 – calpain 13
Hs.34853.0.S2_3p_at
-3.5
-4.0
ID4 – inhibitor of DNA binding 4, dominant negative helix-loop-helix protein
g3387766_3p_a_at
-3.9
-3.6
GPM6B – glycoprotein M6B
Hs.10587.0.S1_3p_at
-3.2
-4.3
DMN – desmuslin
Hs2.348883.1.S1_3p_s_at
-3.9
-3.6
FOXC1 – forkhead box C1
g4758521_3p_at
-4.9
-2.7
SPARCL1 – SPARC-like 1 (mast9, hevin)
g11037715_3p_x_at
-3.9
-3.7
ROPN1 – ropporin, rhophilin associated protein 1
g4504914_3p_at
-3.9
-3.8
KRT15 – keratin 15
Hs.82101.0.S3_3p_at
-3.7
-4.0
PHLDA1 – pleckstrin homology-like domain, family A, member 1
Hs.149356.0.S1_3p_at
-3.8
-3.9
LOC728264 – hypothetical protein LOC728264
g4506856_3p_s_at
-3.3
-4.6
CX3CL1 – chemokine (C-X3-C motif) ligand 1
g4506516_3p_at
-3.9
-4.1
RGS2 – regulator of G-protein signaling 2, 24 kDa
g7657105_3p_at
-4.2
-4.0
GABRP – gamma-aminobutyric acid (GABA) A receptor, pi
Hs.25956.0.S1_3p_at
-4.1
-4.4
SOSTDC1 – sclerostin domain containing 1
Hs.153961.2.S2_3p_at
-4.6
-3.9
BOC – Boc homolog (mouse)
g11991655_3p_at
-4.5
-4.2
C2orf40 – chromosome 2 open reading frame 40
Hs.288850.0.S1_3p_at
-4.1
-4.7
PHLDA1 – pleckstrin homology-like domain, family A, member 1
g7662650_3p_at
-5.1
-3.7
C13orf15 – chromosome 13 open reading frame 15
g4557694_3p_a_at
-4.5
-4.5
KIT – v-kit Hardy-Zuckerman 4 feline sarcoma viral oncogene homolog
Hs.127428.2.S2_3p_a_at
-4.3
-4.9
HOXA9 – homeobox A9
g6005949_3p_at
-4.4
-4.8
WIF1 – WNT inhibitory factor 1
Hs.34853.0.S3_3p_at
-4.8
-4.5
ID4 – inhibitor of DNA binding 4, dominant negative helix-loop-helix protein
g4559274_3p_a_at
-4.1
-5.3
ELF5 – E74-like factor 5 (ets domain transcription factor)
g8400731_3p_a_at
-4.4
-5.2
SFRP1 – secreted frizzled-related protein 1
g5032314_3p_a_at
-4.9
-4.9
DMD – dystrophin (muscular dystrophy, Duchenne and Becker types)
g6005714_3p_at
-4.7
-5.6
SLC6A14 – solute carrier family 6 (amino acid transporter), member 14
Ductal carcinoma in situ (DCIS) and invasive ductal carcinoma (IDC) data presented as log2(fold changes) relative to normal epithelium.
Table 5
Top 50 genes differentially expressed in tumor-associated stroma
Probe set
DCIS
IDC
Gene description
Hs.179729.0.S1_3p_a_at
6.5
7.0
COL10A1 – collagen, type X, alpha 1(Schmid metaphyseal chondrodysplasia)
Hs.28792.0.S1_3p_at
5.9
6.0
NA
4876385_3p_at
4.9
6.3
COL11A1 – collagen, type XI, alpha 1
g7019348_3p_at
4.9
5.7
GREM1 – gremlin 1, cysteine knot superfamily, homolog (Xenopus laevis)
Hs.179729.1.S1_3p_a_at
4.8
5.5
COL10A1 – collagen, type X, alpha 1(Schmid metaphyseal chondrodysplasia)
Hs.297939.3.S1_3p_at
4.6
5.1
FNDC1 – fibronectin type III domain containing 1
37892_3p_at
4.1
5.6
COL11A1 – collagen, type XI, alpha 1
Hs.41271.0.S1_3p_at
4.7
4.9
COL8A1 – collagen, type VIII, alpha 1
g4502938_3p_s_at
3.8
5.4
COL11A1 – collagen, type XI, alpha 1
g186414_3p_a_at
4.5
4.4
INHBA – inhibin, beta A
g8393842_3p_at
4.4
4.4
NOX4 – NADPH oxidase 4
Hs.288467.0.S1_3p_at
3.8
4.8
LRRC15 – leucine rich repeat containing 15
Hs.105700.0.S1_3p_a_at
4.1
4.2
SFRP4 – secreted frizzled-related protein 4
g4481752_3p_at
3.6
4.4
GJB2 – gap junction protein, beta 2, 26 kDa
g8923132_3p_at
3.6
4.3
ASPN – asporin
Hs.287820.2.A1_3p_s_at
3.8
4.1
FN1 – fibronectin 1
g8400733_3p_a_at
3.5
3.9
SFRP4 – secreted frizzled-related protein 4
g10863087_3p_a_at
3.4
3.9
GREM1 – gremlin 1, cysteine knot superfamily, homolog (Xenopus laevis)
g5174662_3p_at
3.4
3.4
S100P – S100 calcium binding protein P
Hs.283713.0.A1_3p_at
3.2
3.5
CTHRC1 – collagen triple helix repeat containing 1
g4502844_3p_at
3.1
3.5
CILP – cartilage intermediate layer protein, nucleotide pyrophosphohydrolase
Hs.76722.2.S1_3p_at
2.8
3.7
-
Hs.70823.0.S3_3p_at
3.3
3.2
SULF1 – sulfatase 1
g4505186_3p_at
3.3
3.0
CXCL9 – chemokine (C-X-C motif) ligand 9
Hs.101302.0.S2_3p_s_at
2.2
3.9
COL12A1 – collagen, type XII, alpha 1
g11415037_3p_at
-3.2
-3.0
SLC22A3 – solute carrier family 22 (extraneuronal monoamine transporter), member 3
Hs.325823.0.A1_3p_at
-2.7
-3.5
CD36 – CD36 molecule (thrombospondin receptor)
g4557418_3p_at
-2.7
-3.6
CD36 – CD36 molecule (thrombospondin receptor)
g4557544_3p_a_at
-2.9
-3.5
EDN3 – endothelin 3
Hs2.147313.1.S1_3p_s_at
-2.9
-3.5
CD300LG – CD300 molecule-like family member g
Hs.106283.4.S1_3p_at
-3.0
-3.4
KLHL13 – kelch-like 13 (Drosophila)
g8400731_3p_a_at
-2.6
-4.0
SFRP1 – secreted frizzled-related protein 1
Hs.250692.0.S4_3p_at
-3.0
-3.7
HLF – hepatic leukemia factor
g4826977_3p_at
-3.3
-3.5
RELN – reelin
Hs.76325.1.A1_3p_x_at
-3.0
-3.9
IGJ – immunoglobulin J polypeptide, linker protein for immunoglobulin alpha and mu polypeptides
Hs.76325.1.A1_3p_at
-3.1
-4.0
IGJ – immunoglobulin J polypeptide, linker protein for immunoglobulin alpha and mu polypeptides
g10835124_3p_a_at
-4.0
-3.1
DCX – doublecortex; lissencephaly, X-linked (doublecortin)
g7657105_3p_at
-3.2
-4.0
GABRP – gamma-aminobutyric acid A receptor, pi
g4506328_3p_at
-3.4
-3.9
PTPRZ1 – protein tyrosine phosphatase, receptor-type, Z polypeptide 1
g4758377_3p_at
-3.9
-3.4
FIGF – c-fos induced growth factor (vascular endothelial growth factor D)
g12707575_3p_at
-3.3
-4.1
OXTR – oxytocin receptor
g13518036_3p_a_at
-2.5
-4.9
MATN2 – matrilin 2
g4559274_3p_a_at
-3.9
-3.6
ELF5 – E74-like factor 5 (ets domain transcription factor)
Hs.10587.0.S1_3p_at
-2.5
-5.1
DMN – desmuslin
Hs.49696.0.A1_3p_at
-3.7
-4.0
SCARA5 – scavenger receptor class A, member 5 (putative)
g4557578_3p_at
-3.4
-4.4
FABP4 – fatty acid binding protein 4, adipocyte
g13186315_3p_a_at
-3.5
-4.3
CAPN6 – calpain 6
g11991655_3p_at
-3.2
-5.3
C2orf40 – chromosome 2 open reading frame 40
g562105_3p_a_at
-4.9
-4.5
DLK1 – delta-like 1 homolog (Drosophila)
g6005949_3p_at
-5.0
-4.8
WIF1 – WNT inhibitory factor 1
Ductal carcinoma in situ (DCIS) and invasive ductal carcinoma (IDC) data presented as log2(fold changes) relative to normal stroma.

Stromal gene expression signature associated with tumor invasion

We next compared the gene expression patterns associated with the DCIS to IDC transition within each compartment. In the tumor epithelium, there were only three genes (POSTN, periostin; SPARC, osteoconectin; SPARCL1, SPARC-like 1) that were significantly upregulated in IDC relative to DCIS. All three genes are known to be specifically expressed in the stroma [2123] and were indeed strongly expressed in the stroma samples in our dataset. Their apparent overexpression in IDC relative to DCIS might therefore be due to contaminating stromal cells in the procured epithelial cell populations in the IDC samples but not in DCIS samples. In the stroma, however, there were more significant changes in comparing IDC-S with DCIS-S, with 76 upregulated genes and 229 downregulated genes (Figure 2). The lack of significant changes in gene expression in the epithelium associated with the DCIS-IDC transition seen here was consistent with that in our previous study [9].
Table 6 presents the top 50 differentially expressed genes between DICS-S and IDC-S (see Additional data file 1). Among genes with increased expression in IDC-S, three matrix metalloproteases (MMP11, MMP2 and MMP14) were notable. In fact, one additional matrix metalloprotease (MMP13) had higher expression in IDC-S than in DCIS-S, with adjusted P = 0.06. These genes have been known to be involved in tumor invasion [3]. On the other hand, genes with decreased expression in IDC-S included many genes involved in vasculature development (for example, EMCN, FLT1, KDR, SELE, MYH11, EDNRB and PODXL), a process expected to increase in invasive cancer. This paradoxical result might reflect the decreased vascular density in the leading invasive front where we microdissected the stroma relative to the stroma surrounding DCIS.
Table 6
Top 50 genes differentially expressed in invasive stroma compared to in situ stroma
Probe set
Log2 (fold change)
Adjusted P value
Gene description
Hs2.434299.1.S1_3p_at
1.61
8.58 × 10-3
-
g13027795_3p_s_at
1.45
1.74 × 10-2
MMP11 – matrix metallopeptidase 11 (stromelysin 3)
Hs.50081.1.S1_3p_a_at
1.36
5.47 × 10-3
KIAA1199 – KIAA1199
g11641276_3p_s_at
1.24
3.71 × 10-2
PDE4DIP – phosphodiesterase 4D interacting protein (myomegalin)
Hs.169517.0.S1_3p_a_at
1.16
5.72 × 10-3
ALDH1B1 – aldehyde dehydrogenase 1 family, member B1
Hs.98523.0.A1_3p_at
1.13
5.32 × 10-3
FAT3 – FAT tumor suppressor homolog 3 (Drosophila)
g10938018_3p_at
1.04
1.74 × 10-2
EPYC – epiphycan
Hs2.350890.1.S1_3p_s_at
1.03
3.72 × 10-2
GABRB2 – gamma-aminobutyric acid (GABA) A receptor, beta 2
g4507922_3p_at
0.98
1.70 × 10-2
WISP2 – WNT1 inducible signaling pathway protein 2
g11342665_3p_at
0.98
4.53 × 10-2
MMP2 – matrix metallopeptidase 2 (gelatinase A, 72kDa gelatinase, 72 kDa type IV collagenase)
g13124890_3p_a_at
0.93
3.28 × 10-2
GALNT1 – UDP-N-acetyl-alpha-D-galactosamine:polypeptide N-acetylgalactosaminyltransferase 1 (GalNAc-T1)
Hs.98523.0.A1_3p_x_at
0.87
3.15 × 10-2
FAT3 – FAT tumor suppressor homolog 3 (Drosophila)
Hs.238532.0.A1_3p_at
0.76
3.71 × 10-2
GALNTL2 – UDP-N-acetyl-alpha-D-galactosamine:polypeptide N-acetylgalactosaminyltransferase-like 2
Hs.42927.0.S1_3p_at
0.75
3.39 × 10-2
ANTXR1 – anthrax toxin receptor 1
Hs2.359399.1.S1_3p_at
0.74
3.71 × 10-2
LOC285758 – hypothetical protein LOC285758
Hs.235795.0.A1_3p_at
0.74
1.38 × 10-2
-
Hs2.46679.2.S1_3p_s_at
0.66
2.11 × 10-3
-
g469044_3p_a_at
0.60
3.22 × 10-2
CNTN1 – contactin 1
g4758607_3p_at
0.53
1.20 × 10-2
-
Hs.288553.0.S1_3p_s_at
0.52
2.82 × 10-2
-
200661_3p_at
0.51
4.16 × 10-2
CTSA – cathepsin A
Hs.2399.1.S1_3p_s_at
0.51
4.48 × 10-2
MMP14 – matrix metallopeptidase 14 (membrane-inserted)
Hs.98183.0.A1_3p_at
0.48
8.58 × 10-3
RSPO4 – R-spondin family, member 4
208756_3p_at
0.48
2.28 × 10-2
EIF3I – eukaryotic translation initiation factor 3, subunit I
Hs.162647.0.S1_3p_at
0.48
2.69 × 10-2
DKFZP547L112 – hypothetical protein DKFZp547L112
Hs.22968.0.S1_3p_a_at
-2.00
8.37 × 10-6
FLT1 – fms-related tyrosine kinase 1 (vascular endothelial growth factor/vascular permeability factor receptor)
g11545907_3p_at
-2.03
2.68 × 10-5
ELTD1 – EGF, latrophilin and seven transmembrane domain containing 1
g5032094_3p_at
-2.03
3.64 × 10-7
SLCO2A1 – solute carrier organic anion transporter family, member 2A1
Hs.8707.0.S1_3p_at
-2.07
1.29 × 10-5
HECW2 – HECT, C2 and WW domain containing E3 ubiquitin protein ligase 2
Hs.134970.0.S1_3p_a_at
-2.07
6.66 × 10-3
KIF26A – kinesin family member 26A
Hs2.420404.1.S1_3p_at
-2.08
7.70 × 10-6
PELO – pelota homolog (Drosophila)
g4504850_3p_a_at
-2.11
3.62 × 10-2
KCNK5 – potassium channel, subfamily K, member 5
Hs.288681.0.S1_3p_at
-2.13
4.98 × 10-4
THSD7A – thrombospondin, type I, domain containing 7A
g4557546_3p_at
-2.15
1.58 × 10-3
EDNRB – endothelin receptor type B
g11321596_3p_at
-2.19
1.64 × 10-3
KDR – kinase insert domain receptor (a type III receptor tyrosine kinase)
Hs.124675.0.A1_3p_at
-2.21
2.21 × 10-4
GIMAP7 – GTPase, IMAP family member 7
Hs.211388.0.S1_3p_at
-2.24
5.72 × 10-3
RUNDC3B – RUN domain containing 3B
g4885556_3p_at
-2.25
2.13 × 10-3
PODXL – podocalyxin-like
Hs.26530.0.S2_3p_at
-2.30
1.60 × 10-3
SDPR – serum deprivation response (phosphatidylserine binding protein)
g13518036_3p_a_at
-2.41
7.19 × 10-3
MATN2 – matrilin 2
Hs.102415.0.S1_3p_at
-2.43
2.67 × 10-8
EMCN – endomucin
Hs.61935.0.S1_3p_at
-2.45
2.68 × 10-5
PCDH17 – protocadherin 17
g8547214_3p_at
-2.46
3.74 × 10-5
EMCN – endomucin
g4520327_3p_at
-2.48
2.67 × 10-3
IL33 – interleukin 33
Hs.10587.0.S1_3p_at
-2.66
2.29 × 10-3
DMN – desmuslin
g3644039_3p_a_at
-2.67
1.43 × 10-2
TP63 – tumor protein p63
Hs.78344.1.S2_3p_a_at
-2.87
2.13 × 10-3
MYH11 – myosin, heavy chain 11, smooth muscle
g6580814_3p_s_at
-2.93
8.90 × 10-5
INMT – indolethylamine N-methyltransferase
Hs.173560.0.S1_3p_at
-2.94
3.58 × 10-2
ODZ2 – odz, odd Oz/ten-m homolog 2 (Drosophila)
g4506870_3p_at
-3.23
1.80 × 10-3
SELE – selectin E (endothelial adhesion molecule 1)

Stromal gene expression signature associated with tumor grade

We have previously shown that tumor grade is associated with a strong gene expression signature in malignant breast epithelial cells [9]. We therefore examined whether a similar signature also exists in the tumor stroma. Comparing grade I (n = 8) and grade III (n = 7) tumor-associated stroma samples (DCIS-S and IDC-S), we identified 526 upregulated genes and 94 downregulated genes in grade III samples (Figure 5; see also Additional data file 2). The gene set enrichment analysis indicated that the tumor stroma in grade III tumors were associated with a strong immune response signature (interferon signaling, activation of leukocytes and T cells) and with increased mitotic activity (Table 7).
Table 7
Top 20 gene sets enriched in grade III-associated stroma
Name
Size (number of genes)
Normalized enrichment score
False discovery rate q value
CELLULAR_DEFENSE_RESPONSE
52
2.31
0
IMMUNE_RESPONSE
220
2.17
0
IMMUNE_SYSTEM_PROCESS
312
2.16
0
T_CELL_ACTIVATION
42
2.14
0
LEUKOCYTE_ACTIVATION
67
2.09
0
JAK_STAT_CASCADE
28
2.05
6.82 × 10-4
LYMPHOCYTE_ACTIVATION
59
2.05
5.85 × 10-4
CELL_ACTIVATION
73
2.04
5.12 × 10-4
M_PHASE_OF_MITOTIC_CELL_CYCLE
78
2.04
4.55 × 10-4
RESPONSE_TO_VIRUS
48
2.04
5.12 × 10-4
SPINDLE
39
2.03
5.60 × 10-4
MITOSIS
75
2.02
5.99 × 10-4
INTERLEUKIN_RECEPTOR_ACTIVITY
20
2.01
6.33 × 10-4
POSITIVE_REGULATION_OF_IMMUNE_RESPONSE
28
2.00
7.35 × 10-4
REGULATION_OF_IMMUNE_SYSTEM_PROCESS
66
1.99
7.54 × 10-4
POSITIVE_REGULATION_OF_IMMUNE_SYSTEM_PROCESS
50
1.99
7.07 × 10-4
RESPONSE_TO_BIOTIC_STIMULUS
112
1.99
6.65 × 10-4
REGULATION_OF_I_KAPPAB_KINASE_NF_KAPPAB_CASCADE
89
1.99
6.85 × 10-4
MRNA_PROCESSING_GO_0006397
67
1.97
0.001135
RESPONSE_TO_OTHER_ORGANISM
76
1.96
0.001282

Validation of selected differentially expressed genes

We next used quantitative real-time PCR to validate selected genes differentially expressed in the various comparisons presented above. Quantitative real-time PCR analysis of the same samples as used in the microarray analysis confirmed the marked downregulation of WIF1 in both neoplastic epithelium and tumor stroma (Figure 6a) and the marked upregulation of GREM1 in both DCIS-associated and IDC-associated stroma (Figure 6b). In addition, two representative genes (ESR1, estrogen receptor alpha; and RRM2, ribonucleotide reductase M2 subunit) differentially expressed in the stroma between grade III and grade I tumors (see Additional data file 2) were also confirmed by quantitative real-time PCR. In both the epithelium and stroma, RRM2, a cell proliferation marker, was more highly expressed in grade III tumors (Figure 6c), whereas ESR1 was more highly expressed in grade I tumors (Figure 6d). Although expression of estrogen receptor alpha is thought to be restricted to the tumor epithelial cells in human breast cancer [24], we confirmed the low but detectable levels of estrogen receptor alpha expression in stromal fibroblasts by immunohistochemical staining (Figure 6e).

Discussion

Exploratory genome-wide analysis of the tumor microenvironment in breast cancer has been limited to date. Using serial analysis of gene expression coupled with antibody-based ex vivo tissue fractionation, Allinen and colleagues identified a limited set of 417 cell-type-specific genes among the most prominent cell types in breast cancer (epithelial, myoepithelial, and endothelial cells, fibroblasts, and leukocytes) [7]. Finak and colleagues more recently obtained gene expression profiles of both epithelial and stromal compartments from the same tumor biopsy via LCM [25]. These workers only analyzed the morphologically normal epithelium and normal stroma, however, leaving the gene expression changes in the tumor-activated stroma unexplored. Our work therefore provides the first comprehensive comparative analysis of in vivo gene expression changes in the tumor epithelium and its stromal microenvironment during breast cancer progression from normal to DCIS to IDC.
We observed extensive gene expression changes in the stroma associated with DCIS and IDC, suggesting that tumor-adjacent stroma coevolves with the tumor epithelium, even before tumor invasion occurs. These alterations included many components of the extracellular matrix and the extracellular-matrix-remodeling matrix metalloproteases. Increased mitotic gene expression occurred both in the malignant epithelium and adjacent stroma, which may reflect the often observed desmoplastic reaction around the tumor cells. Expression of cytoplasmic ribosomal proteins was generally decreased in both compartments during cancer progression. While this result may seem paradoxical in that increased protein synthesis is considered a hallmark of cancer, it is supported by several different lines of studies. First, decreased expression of many ribosomal proteins has also been observed in colorectal cancer compared with normal mucosal epithelium [26]. Secondly, many ribosomal protein genes have been found to be haploinsufficient tumor suppressors in zebrafish [27]. Thirdly, the oncogenic activity of c-Myc is inhibited by the ribosomal protein L11, and inactivation of the L11 gene by small interfering RNA increases c-Myc-induced transcription and cell proliferation [28].
The mechanism by which ribosomal proteins contribute to tumorigenesis is unknown. Decreased expression of ribosomal proteins in cancer may reflect a qualitative change in ribosomal structure, which may allow differential translation of gene products required for rapid tumor growth. Alternatively, it may reflect some unknown nonribosomal functions by these proteins. In contrast to the decreased expression of these cytoplasmic ribosomal protein genes, we observed increased expression of a number of mitochondrial ribosomal protein genes in both the tumor epithelium and the stroma. The human mitochondrial ribosomes are responsible for the production of several key proteins in bioenergetics including subunits of the ATP synthase. Given the importance of mitochondria in cancer [29, 30], our novel finding suggests that the mitochondrial ribosome may be a potential therapeutic target and thus warrants further study.
The top differentially expressed genes between tumor-associated stroma and the adjacent normal stroma included several signaling molecules known to be important for tumorigenesis. Two antagonists of WNT receptor signaling, WIF1 and SFRP1, were consistently downregulated both in the tumor epithelium and stroma. The WNT signaling pathway plays an important role in development and tissue homeostasis, and its aberrant activation by loss of expression WIF1 or SFRP1 has been shown to be an important early event in breast cancer progression [3133]. Two transforming growth factor beta superfamily members (GREM1 and INHBA) are strongly induced in the tumor-associated stroma. GREM1 is a bone morphogenetic protein antagonist, and it is overexpressed in cancer-associated stromal cells in many solid tumors [34]. It has been hypothesized that bone morphogenetic proteins and bone morphogenetic protein antagonists may play opposing roles in the maintenance of a niche of self-renewing stem cells, with bone morphogenetic protein antagonists such as GREM1 blocking cell differentiation [34]. WNT3A was recently demonstrated in human fibroblasts to markedly increase the expression of GREM2, a close paralog of GREM1 – raising the possibility that the significant downregulation of WNT antagonists (WIF1 and SFRP1) and upregulation of GREM1 in the stroma [35] we observed here may be functionally linked.
INHBA is the gene for the beta A subunit of inhibin and activin, which are pleiotropic growth factors regulating the growth and differentiation of many cell types via autocrine and paracrine mechanisms [36]. Although its role in breast cancer remains unclear, circulating levels of INHBA has been shown to be higher in breast cancer patients with bone metastasis [37]. These signaling molecules could serve as key messengers between the tumor and its microenvironment, as shown for CXCL12 and CXCL14, which are overexpressed in tumor-associated myoepithelial cells and myofibroblasts [6, 7, 38]. We note that in our dataset, however, CXCL12 and CXCL14 were also expressed in normal stroma. This discrepancy could be due to the fact that Allinen and colleagues used purified stromal cell types [7] and we used the whole stroma compartment in our study.
A watershed event in breast cancer progression is the invasion of tumor cells into the stromal compartment. The only morphological diagnostic criterion distinguishing DCIS from IDC is the association of DCIS with a complete basement membrane. Understanding the molecular events that drive the DCIS-IDC transition has been of great interest. We have previously shown [9], and confirm in the present study, that the malignant epithelium of DCIS and IDC are very similar without significant differences at the transcriptome level. This conclusion is supported by the recent demonstration that MCFDCIS cells, a cell line model for DCIS, make the DCIS-IDC transition spontaneously without further molecular changes in the malignant epithelial cells themselves [39]. Instead, this transition is driven by fibroblasts and blocked by myoepithelial cells.
In the present article we demonstrated that the stromal compartment is associated with a relatively small number of significant changes accompanying the DCIS-IDC transition. In particular, several matrix metalloproteases (MMP2, MMP11 and MMP14) showed significantly increased expression in IDC-associated stroma. MMP14, a membrane-type matrix metalloprotease, can activate MMP2 protease activity, which degrades type IV collagen, the major structural component of the basement membrane [40, 41]. MMP11 has recently been shown to exhibit protease activity towards type VI collagen and to promote tumor progression [42]. MMP11 has been shown to be differentially expressed in IDC relative to DCIS in two other studies. Schuetz and colleagues conducted a study similar to ours, using LCM and microarrays to profile the epithelium of patient-matched DCIS and IDC, and found MMP11 to be upregulated in IDC relative to DCIS [43]. Their result differs from ours, however, in that we observed upregulation of MMP11 in the IDC-associated stroma but not in the epithelium. A stromal origin of MMP11 expression had been established previously [44]. The result by Schuetz and coworkers might be due to contaminating nonepithelial cells in their LCM samples, a possibility acknowledged by these authors [43]. In another study, Hannemann and colleagues identified a gene expression signature including MMP11 to be able to distinguish IDC from DCIS [45]. Since no microdissection was performed in that study, the gene expression profiles they obtained were from mixtures of tumor epithelium and stroma. Nevertheless, our results together with these other studies support the notion that stroma-produced matrix metalloproteases may be key players driving the DCIS-IDC transition.
Finally, we showed that – like the epithelial compartment [9] – tumor stroma also exhibited a robust gene expression signature correlating with the histological tumor grade. These genes are primarily involved in immune response and cell-cycle progression. The association of an immune response signature with the more aggressive high-grade tumors is seemingly paradoxical. The interactions between tumor cells and the various immune cells are complex, however, ranging from tumor growth-suppressing effects to tumor growth-promoting effects [4648]. Perhaps the immune response signature associated with high-grade tumors represents the escape phase [48], when the cancer cells become resistant to immune attack and hijack the abundant cytokines and chemokines made by the immune cells to grow, invade and spread to distant organs.

Conclusions

The present study provides the first comparative analysis of the in situ gene expression profiles of patient-matched normal and neoplastic breast epithelial and stromal compartments of both preinvasive and invasive stages of human breast cancer progression. This study of the breast cancer microenvironment at the transcriptome level and previous studies at the genomic [49, 50] and epigenetic [51, 52] levels support the view that the tumor microenvironment is an important co-conspirator rather than a passive bystander during tumorigenesis. Molecular alterations within the stroma offer novel avenues for therapeutic interventions and disease prognosis [53]. This gene expression dataset of carefully procured in situ tumor epithelium and stroma should be a timely and valuable addition to the resources for the breast cancer research community.

Acknowledgements

The present study was supported by NIH RO1-1CA112021-01 (to DCS), the Department of Defense grants W81XWH-04-1-0606 and DAMD170310428 (to DCS), Susan G. Komen Breast Cancer Foundation grant BCTR0402932 (to DCS), and the Avon Foundation (to DCS). The authors thank Erica Chatfield for technical assistance in microdissection and RNA extraction, and they thank Sridhar Ramaswamy and E. Schmidt for helpful discussions and reading of the manuscript.

Competing interests

XJM and ME are employees of BioTheranostics, Inc. ME, X-JM and DCS are named inventors on a patent application relating to the contents of the manuscript.

Authors' contributions

DCS, XJM and ME conceived of the study, participated in its design and coordination, and helped draft the manuscript. DCS and SD performed microdissection and RNA extraction. XJM and ME performed the microarray experiments and microarray data analysis. ER performed the real-time PCR assays. All authors have read and approved the final manuscript.
Literatur
2.
Zurück zum Zitat de Visser KE, Coussens LM: The inflammatory tumor microenvironment and its impact on cancer development. Contrib Microbiol. 2006, 13: 118-137.CrossRefPubMed de Visser KE, Coussens LM: The inflammatory tumor microenvironment and its impact on cancer development. Contrib Microbiol. 2006, 13: 118-137.CrossRefPubMed
3.
Zurück zum Zitat Liotta LA, Kohn EC: The microenvironment of the tumour-host interface. Nature. 2001, 411: 375-379.CrossRefPubMed Liotta LA, Kohn EC: The microenvironment of the tumour-host interface. Nature. 2001, 411: 375-379.CrossRefPubMed
4.
5.
Zurück zum Zitat Muller A, Homey B, Soto H, Ge N, Catron D, Buchanan ME, McClanahan T, Murphy E, Yuan W, Wagner SN, Barrera JL, Mohar A, Verastegui E, Zlotnik A: Involvement of chemokine receptors in breast cancer metastasis. Nature. 2001, 410: 50-56.CrossRefPubMed Muller A, Homey B, Soto H, Ge N, Catron D, Buchanan ME, McClanahan T, Murphy E, Yuan W, Wagner SN, Barrera JL, Mohar A, Verastegui E, Zlotnik A: Involvement of chemokine receptors in breast cancer metastasis. Nature. 2001, 410: 50-56.CrossRefPubMed
6.
Zurück zum Zitat Orimo A, Gupta PB, Sgroi DC, Arenzana-Seisdedos F, Delaunay T, Naeem R, Carey VJ, Richardson AL, Weinberg RA: Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell. 2005, 121: 335-348.CrossRefPubMed Orimo A, Gupta PB, Sgroi DC, Arenzana-Seisdedos F, Delaunay T, Naeem R, Carey VJ, Richardson AL, Weinberg RA: Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell. 2005, 121: 335-348.CrossRefPubMed
7.
Zurück zum Zitat Allinen M, Beroukhim R, Cai L, Brennan C, Lahti-Domenici J, Huang H, Porter D, Hu M, Chin L, Richardson A, Schnitt S, Sellers WR, Polyak K: Molecular characterization of the tumor microenvironment in breast cancer. Cancer Cell. 2004, 6: 17-32.CrossRefPubMed Allinen M, Beroukhim R, Cai L, Brennan C, Lahti-Domenici J, Huang H, Porter D, Hu M, Chin L, Richardson A, Schnitt S, Sellers WR, Polyak K: Molecular characterization of the tumor microenvironment in breast cancer. Cancer Cell. 2004, 6: 17-32.CrossRefPubMed
8.
Zurück zum Zitat Allred DC, Mohsin SK, Fuqua SA: Histological and biological evolution of human premalignant breast disease. Endocr Relat Cancer. 2001, 8: 47-61.CrossRefPubMed Allred DC, Mohsin SK, Fuqua SA: Histological and biological evolution of human premalignant breast disease. Endocr Relat Cancer. 2001, 8: 47-61.CrossRefPubMed
9.
Zurück zum Zitat Ma XJ, Salunga R, Tuggle JT, Gaudet J, Enright E, McQuary P, Payette T, Pistone M, Stecker K, Zhang BM, Zhou YX, Varnholt H, Smith B, Gadd M, Chatfield E, Kessler J, Baer TM, Erlander MG, Sgroi DC: Gene expression profiles of human breast cancer progression. Proc Natl Acad Sci USA. 2003, 100: 5974-5979.CrossRefPubMedPubMedCentral Ma XJ, Salunga R, Tuggle JT, Gaudet J, Enright E, McQuary P, Payette T, Pistone M, Stecker K, Zhang BM, Zhou YX, Varnholt H, Smith B, Gadd M, Chatfield E, Kessler J, Baer TM, Erlander MG, Sgroi DC: Gene expression profiles of human breast cancer progression. Proc Natl Acad Sci USA. 2003, 100: 5974-5979.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Gentleman RC, Carey VJ, Bates DM, Bolstad B, Dettling M, Dudoit S, Ellis B, Gautier L, Ge Y, Gentry J, Hornik K, Hothorn T, Huber W, Iacus S, Irizarry R, Leisch F, Li C, Maechler M, Rossini AJ, Sawitzki G, Smith C, Smyth G, Tierney L, Yang JY, Zhang J: Bioconductor: open software development for computational biology and bioinformatics. Genome Biol. 2004, 5: R80-CrossRefPubMedPubMedCentral Gentleman RC, Carey VJ, Bates DM, Bolstad B, Dettling M, Dudoit S, Ellis B, Gautier L, Ge Y, Gentry J, Hornik K, Hothorn T, Huber W, Iacus S, Irizarry R, Leisch F, Li C, Maechler M, Rossini AJ, Sawitzki G, Smith C, Smyth G, Tierney L, Yang JY, Zhang J: Bioconductor: open software development for computational biology and bioinformatics. Genome Biol. 2004, 5: R80-CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Bolstad BM, Irizarry RA, Astrand M, Speed TP: A comparison of normalization methods for high density oligonucleotide array data based on variance and bias. Bioinformatics. 2003, 19: 185-193.CrossRefPubMed Bolstad BM, Irizarry RA, Astrand M, Speed TP: A comparison of normalization methods for high density oligonucleotide array data based on variance and bias. Bioinformatics. 2003, 19: 185-193.CrossRefPubMed
12.
Zurück zum Zitat Wettenhall JM, Smyth GK: limmaGUI: a graphical user interface for linear modeling of microarray data. Bioinformatics. 2004, 20: 3705-3706.CrossRefPubMed Wettenhall JM, Smyth GK: limmaGUI: a graphical user interface for linear modeling of microarray data. Bioinformatics. 2004, 20: 3705-3706.CrossRefPubMed
13.
Zurück zum Zitat Benjamin RS, Hochberg Y: Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Ser B. 1995, 57: 289-300. Benjamin RS, Hochberg Y: Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Ser B. 1995, 57: 289-300.
15.
Zurück zum Zitat Subramanian A, Kuehn H, Gould J, Tamayo P, Mesirov JP: GSEA-P: a desktop application for Gene Set Enrichment Analysis. Bioinformatics. 2007, 23: 3251-3253.CrossRefPubMed Subramanian A, Kuehn H, Gould J, Tamayo P, Mesirov JP: GSEA-P: a desktop application for Gene Set Enrichment Analysis. Bioinformatics. 2007, 23: 3251-3253.CrossRefPubMed
16.
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: 207-210.CrossRefPubMedPubMedCentral Edgar R, Domrachev M, Lash AE: Gene Expression Omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 2002, 30: 207-210.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Ma XJ, Wang Z, Ryan PD, Isakoff SJ, Barmettler A, Fuller A, Muir B, Mohapatra G, Salunga R, Tuggle JT, Tran Y, Tran D, Tassin A, Amon P, Wang W, Enright E, Stecker K, Estepa-Sabal E, Smith B, Younger J, Balis U, Michaelson J, Bhan A, Habin K, Baer TM, Brugge J, Haber DA, Erlander MG, Sgroi DC: A two-gene expression ratio predicts clinical outcome in breast cancer patients treated with tamoxifen. Cancer Cell. 2004, 5: 607-616.CrossRefPubMed Ma XJ, Wang Z, Ryan PD, Isakoff SJ, Barmettler A, Fuller A, Muir B, Mohapatra G, Salunga R, Tuggle JT, Tran Y, Tran D, Tassin A, Amon P, Wang W, Enright E, Stecker K, Estepa-Sabal E, Smith B, Younger J, Balis U, Michaelson J, Bhan A, Habin K, Baer TM, Brugge J, Haber DA, Erlander MG, Sgroi DC: A two-gene expression ratio predicts clinical outcome in breast cancer patients treated with tamoxifen. Cancer Cell. 2004, 5: 607-616.CrossRefPubMed
19.
Zurück zum Zitat Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES, Mesirov JP: Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA. 2005, 102: 15545-15550.CrossRefPubMedPubMedCentral Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES, Mesirov JP: Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA. 2005, 102: 15545-15550.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS, Eppig JT, Harris MA, Hill DP, Issel-Tarver L, Kasarskis A, Lewis S, Matese JC, Richardson JE, Ringwald M, Rubin GM, Sherlock G: Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat Genet. 2000, 25: 25-29.CrossRefPubMedPubMedCentral Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS, Eppig JT, Harris MA, Hill DP, Issel-Tarver L, Kasarskis A, Lewis S, Matese JC, Richardson JE, Ringwald M, Rubin GM, Sherlock G: Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat Genet. 2000, 25: 25-29.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Kanno A, Satoh K, Masamune A, Hirota M, Kimura K, Umino J, Hamada S, Satoh A, Egawa S, Motoi F, Unno M, Shimosegawa T: Periostin, secreted from stromal cells, has biphasic effect on cell migration and correlates with the epithelial to mesenchymal transition of human pancreatic cancer cells. Int J Cancer. 2008, 122: 2707-2718.CrossRefPubMed Kanno A, Satoh K, Masamune A, Hirota M, Kimura K, Umino J, Hamada S, Satoh A, Egawa S, Motoi F, Unno M, Shimosegawa T: Periostin, secreted from stromal cells, has biphasic effect on cell migration and correlates with the epithelial to mesenchymal transition of human pancreatic cancer cells. Int J Cancer. 2008, 122: 2707-2718.CrossRefPubMed
22.
Zurück zum Zitat Coutu DL, Wu JH, Monette A, Rivard GE, Blostein MD, Galipeau J: Periostin, a member of a novel family of vitamin K-dependent proteins, is expressed by mesenchymal stromal cells. J Biol Chem. 2008, 283: 17991-18001.CrossRefPubMed Coutu DL, Wu JH, Monette A, Rivard GE, Blostein MD, Galipeau J: Periostin, a member of a novel family of vitamin K-dependent proteins, is expressed by mesenchymal stromal cells. J Biol Chem. 2008, 283: 17991-18001.CrossRefPubMed
23.
Zurück zum Zitat Framson PE, Sage EH: SPARC and tumor growth: where the seed meets the soil?. J Cell Biochem. 2004, 92: 679-690.CrossRefPubMed Framson PE, Sage EH: SPARC and tumor growth: where the seed meets the soil?. J Cell Biochem. 2004, 92: 679-690.CrossRefPubMed
24.
Zurück zum Zitat Jensen EV, Cheng G, Palmieri C, Saji S, Makela S, Van Noorden S, Wahlstrom T, Warner M, Coombes RC, Gustafsson JA: Estrogen receptors and proliferation markers in primary and recurrent breast cancer. Proc Natl Acad Sci USA. 2001, 98: 15197-15202.CrossRefPubMedPubMedCentral Jensen EV, Cheng G, Palmieri C, Saji S, Makela S, Van Noorden S, Wahlstrom T, Warner M, Coombes RC, Gustafsson JA: Estrogen receptors and proliferation markers in primary and recurrent breast cancer. Proc Natl Acad Sci USA. 2001, 98: 15197-15202.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Finak G, Sadekova S, Pepin F, Hallett M, Meterissian S, Halwani F, Khetani K, Souleimanova M, Zabolotny B, Omeroglu A, Park M: Gene expression signatures of morphologically normal breast tissue identify basal-like tumors. Breast Cancer Res. 2006, 8: R58-CrossRefPubMedPubMedCentral Finak G, Sadekova S, Pepin F, Hallett M, Meterissian S, Halwani F, Khetani K, Souleimanova M, Zabolotny B, Omeroglu A, Park M: Gene expression signatures of morphologically normal breast tissue identify basal-like tumors. Breast Cancer Res. 2006, 8: R58-CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Kasai H, Nadano D, Hidaka E, Higuchi K, Kawakubo M, Sato TA, Nakayama J: Differential expression of ribosomal proteins in human normal and neoplastic colorectum. J Histochem Cytochem. 2003, 51: 567-574.CrossRefPubMed Kasai H, Nadano D, Hidaka E, Higuchi K, Kawakubo M, Sato TA, Nakayama J: Differential expression of ribosomal proteins in human normal and neoplastic colorectum. J Histochem Cytochem. 2003, 51: 567-574.CrossRefPubMed
27.
Zurück zum Zitat Amsterdam A, Sadler KC, Lai K, Farrington S, Bronson RT, Lees JA, Hopkins N: Many ribosomal protein genes are cancer genes in zebrafish. PLoS Biol. 2004, 2: E139-CrossRefPubMedPubMedCentral Amsterdam A, Sadler KC, Lai K, Farrington S, Bronson RT, Lees JA, Hopkins N: Many ribosomal protein genes are cancer genes in zebrafish. PLoS Biol. 2004, 2: E139-CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Hagland H, Nikolaisen J, Hodneland LI, Gjertsen BT, Bruserud O, Tronstad KJ: Targeting mitochondria in the treatment of human cancer: a coordinated attack against cancer cell energy metabolism and signalling. Expert Opin Ther Targets. 2007, 11: 1055-1069.CrossRefPubMed Hagland H, Nikolaisen J, Hodneland LI, Gjertsen BT, Bruserud O, Tronstad KJ: Targeting mitochondria in the treatment of human cancer: a coordinated attack against cancer cell energy metabolism and signalling. Expert Opin Ther Targets. 2007, 11: 1055-1069.CrossRefPubMed
31.
Zurück zum Zitat Ugolini F, Charafe-Jauffret E, Bardou VJ, Geneix J, Adelaide J, Labat-Moleur F, Penault-Llorca F, Longy M, Jacquemier J, Birnbaum D, Pebusque MJ: WNT pathway and mammary carcinogenesis: loss of expression of candidate tumor suppressor gene SFRP1 in most invasive carcinomas except of the medullary type. Oncogene. 2001, 20: 5810-5817.CrossRefPubMed Ugolini F, Charafe-Jauffret E, Bardou VJ, Geneix J, Adelaide J, Labat-Moleur F, Penault-Llorca F, Longy M, Jacquemier J, Birnbaum D, Pebusque MJ: WNT pathway and mammary carcinogenesis: loss of expression of candidate tumor suppressor gene SFRP1 in most invasive carcinomas except of the medullary type. Oncogene. 2001, 20: 5810-5817.CrossRefPubMed
32.
Zurück zum Zitat Wissmann C, Wild PJ, Kaiser S, Roepcke S, Stoehr R, Woenckhaus M, Kristiansen G, Hsieh JC, Hofstaedter F, Hartmann A, Knuechel R, Rosenthal A, Pilarsky C: WIF1, a component of the Wnt pathway, is down-regulated in prostate, breast, lung, and bladder cancer. J Pathol. 2003, 201: 204-212.CrossRefPubMed Wissmann C, Wild PJ, Kaiser S, Roepcke S, Stoehr R, Woenckhaus M, Kristiansen G, Hsieh JC, Hofstaedter F, Hartmann A, Knuechel R, Rosenthal A, Pilarsky C: WIF1, a component of the Wnt pathway, is down-regulated in prostate, breast, lung, and bladder cancer. J Pathol. 2003, 201: 204-212.CrossRefPubMed
33.
Zurück zum Zitat Klopocki E, Kristiansen G, Wild PJ, Klaman I, Castanos-Velez E, Singer G, Stohr R, Simon R, Sauter G, Leibiger H, Essers L, Weber B, Hermann K, Rosenthal A, Hartmann A, Dahl E: Loss of SFRP1 is associated with breast cancer progression and poor prognosis in early stage tumors. Int J Oncol. 2004, 25: 641-649.PubMed Klopocki E, Kristiansen G, Wild PJ, Klaman I, Castanos-Velez E, Singer G, Stohr R, Simon R, Sauter G, Leibiger H, Essers L, Weber B, Hermann K, Rosenthal A, Hartmann A, Dahl E: Loss of SFRP1 is associated with breast cancer progression and poor prognosis in early stage tumors. Int J Oncol. 2004, 25: 641-649.PubMed
34.
Zurück zum Zitat Sneddon JB, Zhen HH, Montgomery K, Rijn van de M, Tward AD, West R, Gladstone H, Chang HY, Morganroth GS, Oro AE, Brown PO: Bone morphogenetic protein antagonist gremlin 1 is widely expressed by cancer-associated stromal cells and can promote tumor cell proliferation. Proc Natl Acad Sci USA. 2006, 103: 14842-14847.CrossRefPubMedPubMedCentral Sneddon JB, Zhen HH, Montgomery K, Rijn van de M, Tward AD, West R, Gladstone H, Chang HY, Morganroth GS, Oro AE, Brown PO: Bone morphogenetic protein antagonist gremlin 1 is widely expressed by cancer-associated stromal cells and can promote tumor cell proliferation. Proc Natl Acad Sci USA. 2006, 103: 14842-14847.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Klapholz-Brown Z, Walmsley GG, Nusse YM, Nusse R, Brown PO: Transcriptional program induced by Wnt protein in human fibroblasts suggests mechanisms for cell cooperativity in defining tissue microenvironments. PLoS ONE. 2007, 2: e945-CrossRefPubMedPubMedCentral Klapholz-Brown Z, Walmsley GG, Nusse YM, Nusse R, Brown PO: Transcriptional program induced by Wnt protein in human fibroblasts suggests mechanisms for cell cooperativity in defining tissue microenvironments. PLoS ONE. 2007, 2: e945-CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Reis FM, Luisi S, Carneiro MM, Cobellis L, Federico M, Camargos AF, Petraglia F: Activin, inhibin and the human breast. Mol Cell Endocrinol. 2004, 225: 77-82.CrossRefPubMed Reis FM, Luisi S, Carneiro MM, Cobellis L, Federico M, Camargos AF, Petraglia F: Activin, inhibin and the human breast. Mol Cell Endocrinol. 2004, 225: 77-82.CrossRefPubMed
37.
Zurück zum Zitat Mylonas I, Jeschke U, Shabani N, Kuhn C, Friese K, Gerber B: Inhibin/activin subunits (inhibin-alpha, -betaA and -betaB) are differentially expressed in human breast cancer and their metastasis. Oncol Rep. 2005, 13: 81-88.PubMed Mylonas I, Jeschke U, Shabani N, Kuhn C, Friese K, Gerber B: Inhibin/activin subunits (inhibin-alpha, -betaA and -betaB) are differentially expressed in human breast cancer and their metastasis. Oncol Rep. 2005, 13: 81-88.PubMed
38.
Zurück zum Zitat Burger JA, Kipps TJ: CXCR4: a key receptor in the crosstalk between tumor cells and their microenvironment. Blood. 2006, 107: 1761-1767.CrossRefPubMed Burger JA, Kipps TJ: CXCR4: a key receptor in the crosstalk between tumor cells and their microenvironment. Blood. 2006, 107: 1761-1767.CrossRefPubMed
39.
Zurück zum Zitat Hu M, Yao J, Carroll DK, Weremowicz S, Chen H, Carrasco D, Richardson A, Violette S, Nikolskaya T, Nikolsky Y, Bauerlein EL, Hahn WC, Gelman RS, Allred C, Bissell MJ, Schnitt S, Polyak K: Regulation of in situ to invasive breast carcinoma transition. Cancer Cell. 2008, 13: 394-406.CrossRefPubMedPubMedCentral Hu M, Yao J, Carroll DK, Weremowicz S, Chen H, Carrasco D, Richardson A, Violette S, Nikolskaya T, Nikolsky Y, Bauerlein EL, Hahn WC, Gelman RS, Allred C, Bissell MJ, Schnitt S, Polyak K: Regulation of in situ to invasive breast carcinoma transition. Cancer Cell. 2008, 13: 394-406.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Rozanov DV, Savinov AY, Williams R, Liu K, Golubkov VS, Krajewski S, Strongin AY: Molecular signature of MT1-MMP: transactivation of the downstream universal gene network in cancer. Cancer Res. 2008, 68: 4086-4096.CrossRefPubMedPubMedCentral Rozanov DV, Savinov AY, Williams R, Liu K, Golubkov VS, Krajewski S, Strongin AY: Molecular signature of MT1-MMP: transactivation of the downstream universal gene network in cancer. Cancer Res. 2008, 68: 4086-4096.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Egeblad M, Werb Z: New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer. 2002, 2: 161-174.CrossRefPubMed Egeblad M, Werb Z: New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer. 2002, 2: 161-174.CrossRefPubMed
42.
Zurück zum Zitat Motrescu ER, Blaise S, Etique N, Messaddeq N, Chenard MP, Stoll I, Tomasetto C, Rio MC: Matrix metalloproteinase-11/stromelysin-3 exhibits collagenolytic function against collagen VI under normal and malignant conditions. Oncogene. 2008, 27: 6347-55.CrossRefPubMed Motrescu ER, Blaise S, Etique N, Messaddeq N, Chenard MP, Stoll I, Tomasetto C, Rio MC: Matrix metalloproteinase-11/stromelysin-3 exhibits collagenolytic function against collagen VI under normal and malignant conditions. Oncogene. 2008, 27: 6347-55.CrossRefPubMed
43.
Zurück zum Zitat Schuetz CS, Bonin M, Clare SE, Nieselt K, Sotlar K, Walter M, Fehm T, Solomayer E, Riess O, Wallwiener D, Kurek R, Neubauer HJ: Progression-specific genes identified by expression profiling of matched ductal carcinomas in situ and invasive breast tumors, combining laser capture microdissection and oligonucleotide microarray analysis. Cancer Res. 2006, 66: 5278-5286.CrossRefPubMed Schuetz CS, Bonin M, Clare SE, Nieselt K, Sotlar K, Walter M, Fehm T, Solomayer E, Riess O, Wallwiener D, Kurek R, Neubauer HJ: Progression-specific genes identified by expression profiling of matched ductal carcinomas in situ and invasive breast tumors, combining laser capture microdissection and oligonucleotide microarray analysis. Cancer Res. 2006, 66: 5278-5286.CrossRefPubMed
44.
Zurück zum Zitat Basset P, Bellocq JP, Wolf C, Stoll I, Hutin P, Limacher JM, Podhajcer OL, Chenard MP, Rio MC, Chambon P: A novel metalloproteinase gene specifically expressed in stromal cells of breast carcinomas. Nature. 1990, 348: 699-704.CrossRefPubMed Basset P, Bellocq JP, Wolf C, Stoll I, Hutin P, Limacher JM, Podhajcer OL, Chenard MP, Rio MC, Chambon P: A novel metalloproteinase gene specifically expressed in stromal cells of breast carcinomas. Nature. 1990, 348: 699-704.CrossRefPubMed
45.
Zurück zum Zitat Hannemann J, Velds A, Halfwerk JB, Kreike B, Peterse JL, Vijver van de MJ: Classification of ductal carcinoma in situ by gene expression profiling. Breast Cancer Res. 2006, 8: R61-CrossRefPubMedPubMedCentral Hannemann J, Velds A, Halfwerk JB, Kreike B, Peterse JL, Vijver van de MJ: Classification of ductal carcinoma in situ by gene expression profiling. Breast Cancer Res. 2006, 8: R61-CrossRefPubMedPubMedCentral
46.
Zurück zum Zitat de Visser KE, Eichten A, Coussens LM: Paradoxical roles of the immune system during cancer development. Nat Rev Cancer. 2006, 6: 24-37.CrossRefPubMed de Visser KE, Eichten A, Coussens LM: Paradoxical roles of the immune system during cancer development. Nat Rev Cancer. 2006, 6: 24-37.CrossRefPubMed
47.
Zurück zum Zitat Clement JF, Meloche S, Servant MJ: The IKK-related kinases: from innate immunity to oncogenesis. Cell Res. 2008, 18: 889-899.CrossRefPubMed Clement JF, Meloche S, Servant MJ: The IKK-related kinases: from innate immunity to oncogenesis. Cell Res. 2008, 18: 889-899.CrossRefPubMed
49.
Zurück zum Zitat Fukino K, Shen L, Matsumoto S, Morrison CD, Mutter GL, Eng C: Combined total genome loss of heterozygosity scan of breast cancer stroma and epithelium reveals multiplicity of stromal targets. Cancer Res. 2004, 64: 7231-7236.CrossRefPubMed Fukino K, Shen L, Matsumoto S, Morrison CD, Mutter GL, Eng C: Combined total genome loss of heterozygosity scan of breast cancer stroma and epithelium reveals multiplicity of stromal targets. Cancer Res. 2004, 64: 7231-7236.CrossRefPubMed
50.
Zurück zum Zitat Patocs A, Zhang L, Xu Y, Weber F, Caldes T, Mutter GL, Platzer P, Eng C: Breast-cancer stromal cells with TP53 mutations and nodal metastases. N Engl J Med. 2007, 357: 2543-2551.CrossRefPubMed Patocs A, Zhang L, Xu Y, Weber F, Caldes T, Mutter GL, Platzer P, Eng C: Breast-cancer stromal cells with TP53 mutations and nodal metastases. N Engl J Med. 2007, 357: 2543-2551.CrossRefPubMed
51.
Zurück zum Zitat Fiegl H, Millinger S, Goebel G, Muller-Holzner E, Marth C, Laird PW, Widschwendter M: Breast cancer DNA methylation profiles in cancer cells and tumor stroma: association with HER-2/neu status in primary breast cancer. Cancer Res. 2006, 66: 29-33.CrossRefPubMed Fiegl H, Millinger S, Goebel G, Muller-Holzner E, Marth C, Laird PW, Widschwendter M: Breast cancer DNA methylation profiles in cancer cells and tumor stroma: association with HER-2/neu status in primary breast cancer. Cancer Res. 2006, 66: 29-33.CrossRefPubMed
52.
Zurück zum Zitat Hu M, Yao J, Cai L, Bachman KE, Brule van den F, Velculescu V, Polyak K: Distinct epigenetic changes in the stromal cells of breast cancers. Nat Genet. 2005, 37: 899-905.CrossRefPubMed Hu M, Yao J, Cai L, Bachman KE, Brule van den F, Velculescu V, Polyak K: Distinct epigenetic changes in the stromal cells of breast cancers. Nat Genet. 2005, 37: 899-905.CrossRefPubMed
53.
Zurück zum Zitat Finak G, Bertos N, Pepin F, Sadekova S, Souleimanova M, Zhao H, Chen H, Omeroglu G, Meterissian S, Omeroglu A, Hallett M, Park M: Stromal gene expression predicts clinical outcome in breast cancer. Nat Med. 2008, 14: 518-527.CrossRefPubMed Finak G, Bertos N, Pepin F, Sadekova S, Souleimanova M, Zhao H, Chen H, Omeroglu G, Meterissian S, Omeroglu A, Hallett M, Park M: Stromal gene expression predicts clinical outcome in breast cancer. Nat Med. 2008, 14: 518-527.CrossRefPubMed
Metadaten
Titel
Gene expression profiling of the tumor microenvironment during breast cancer progression
verfasst von
Xiao-Jun Ma
Sonika Dahiya
Elizabeth Richardson
Mark Erlander
Dennis C Sgroi
Publikationsdatum
01.06.2009
Verlag
BioMed Central
Erschienen in
Breast Cancer Research / Ausgabe 1/2009
Elektronische ISSN: 1465-542X
DOI
https://doi.org/10.1186/bcr2222

Weitere Artikel der Ausgabe 1/2009

Breast Cancer Research 1/2009 Zur Ausgabe

Update Onkologie

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