Skip to main content
Erschienen in: Breast Cancer Research 2/2012

Open Access 01.04.2012 | Research article

Functional characterization of the 19q12 amplicon in grade III breast cancers

verfasst von: Rachael Natrajan, Alan Mackay, Paul M Wilkerson, Maryou B Lambros, Daniel Wetterskog, Monica Arnedos, Kai-Keen Shiu, Felipe C Geyer, Anita Langerød, Bas Kreike, Fabien Reyal, Hugo M Horlings, Marc J van de Vijver, Jose Palacios, Britta Weigelt, Jorge S Reis-Filho

Erschienen in: Breast Cancer Research | Ausgabe 2/2012

Abstract

Introduction

The 19q12 locus is amplified in a subgroup of oestrogen receptor (ER)-negative grade III breast cancers. This amplicon comprises nine genes, including cyclin E1 (CCNE1), which has been proposed as its 'driver'. The aim of this study was to identify the genes within the 19q12 amplicon whose expression is required for the survival of cancer cells harbouring their amplification.

Methods

We investigated the presence of 19q12 amplification in a series of 313 frozen primary breast cancers and 56 breast cancer cell lines using microarray comparative genomic hybridisation (aCGH). The nine genes mapping to the smallest region of amplification on 19q12 were silenced using RNA interference in phenotypically matched breast cancer cell lines with (MDA-MB-157 and HCC1569) and without (Hs578T, MCF7, MDA-MB-231, ZR75.1, JIMT1 and BT474) amplification of this locus. Genes whose silencing was selectively lethal in amplified cells were taken forward for further validation. The effects of cyclin-dependent kinase 2 (CDK2) silencing and chemical inhibition were tested in cancer cells with and without CCNE1 amplification.

Results

19q12 amplification was identified in 7.8% of ER-negative grade III breast cancer. Of the nine genes mapping to this amplicon, UQCRFS1, POP4, PLEKHF1, C19ORF12, CCNE1 and C19ORF2 were significantly over-expressed when amplified in primary breast cancers and/or breast cancer cell lines. Silencing of POP4, PLEKHF1, CCNE1 and TSZH3 selectively reduced cell viability in cancer cells harbouring their amplification. Cancer cells with CCNE1 amplification were shown to be dependent on CDK2 expression and kinase activity for their survival.

Conclusions

The 19q12 amplicon may harbour more than a single 'driver', given that expression of POP4, PLEKHF1, CCNE1 and TSZH3 is required for the survival of cancer cells displaying their amplification. The observation that cancer cells harbouring CCNE1 gene amplification are sensitive to CDK2 inhibitors provides a rationale for the testing of these chemical inhibitors in a subgroup of patients with ER-negative grade III breast cancers.
Begleitmaterial
Additional file 1: Table S1. Phenotypic characteristics of primary breast tumours analysed by microarray-based comparative genomic hybridisation. aCGH results previously reported, Natrajan et al. [9]., Hunngermann et al. [30], and Turner et al. [19]. Histological grade assessed by the modified Scarff-Bloom-Richardson system [32]. (PDF 1 MB)
Additional file 2: Table S2. Details of the 56 breast cancer cell lines subjected to microarray-based comparative genomic hybridisation. ER status, HER2 gene amplification status and p53 protein levels and mutational status (adapted from Neve et. al. [38], Arriola et al. [20] and [69] *p53 mRNA levels derived from Mackay et al. [16]. p53 protein levels and mutational status (obtained from COSMIC [70] and Neve et al. [38]). "Positive", detectable protein expression; MUT, mutant; "Negative", no detectable protein expression; WT, wild-type. (PDF 612 KB)
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​bcr3154) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

RN and JRF conceived the study. RN, DW, MBK, MA, KK, FCG and AL carried out the experiments. RN and AM analysed the data. HH, MVV, BK, FR and JP provided samples. RN, PW, BW and JRF wrote the manuscript. All authors read and edited the manuscript, and approved its final version.
Abkürzungen
aCGH
microarray comparative genomic hybridisation
BAC
Bacterial Artificial Chromosome
CDK2
cyclin-dependent kinase 2
CHORI
Children's Hospital Oakland Research Institute
BACE
Breakthrough Array CGH Expression
cbs
circular binary segmentation
ER
oestrogen receptor
FACS
fluorescence-activated cell sorting
FISH
fluorescence in situ hybridisation
MTD
Maximum Tolerated Dose
NKI
Netherlands Cancer Institute
PR
progesterone receptor
Mb
megabase(s)
qRT-PCR
Real-time quantitative reverse transcriptase PCR
siRNA
small interfering RNA.

Introduction

Breast cancer is a heterogeneous disease encompassing a number of distinct entities, characterised by distinct biological features and clinical behaviour. Recent evidence has suggested that this heterogeneity is underpinned by distinct patterns of genomic aberrations, which may contribute to the different transcriptomic profiles and clinical phenotypes [14]. Importantly, it has been shown that oestrogen receptor (ER)-positive and -negative breast cancers are fundamentally different diseases, with distinct transcriptomic profiles, gene copy number aberrations and somatic structural rearrangements [58].
Based on the concept of oncogene addiction, we [9, 10] and others [11, 12] have demonstrated that the constellation of genes that are consistently overexpressed when amplified is enriched for genes selectively required for the survival of cancer cells harbouring their amplification and can be exploited as potential therapeutic targets. Using a combination of microarray-based comparative genomic hybridisation (aCGH) and gene expression profiling [1119], previous studies have identified genes which are consistently overexpressed when amplified and suggested potential "amplicon drivers" (for example, FGFR1, FGFR2, GAB2, PPAPDC1B and ZNF703). It should be noted, however, that whilst many potential targets have been postulated, critical molecular drivers of several amplicons remain elusive.
It has now become evident that not all genes within an amplicon are overexpressed when amplified. For example, in the HER2 amplicon, only 7 of the 13 genes that map to the smallest region of amplification are expressed at significantly higher levels when amplified [2022]. Conversely, evidence now suggests that an amplicon may harbour more than one driver [10, 11, 17, 18, 23, 24]. For instance, within the 8p11.2 amplicon, the expression of FGFR1, PPAPDC1B, WHSC1L1, LSM1 and ZNF103 has been shown to be selectively required for the survival of cancer cells harbouring the amplification of these genes [10, 11, 1719, 25].
Amplification of the 19q12 locus has been reported to be found in up to 15% of ER-negative breast cancers [9, 26]. This amplicon often encompasses the cell cycle regulatory gene CCNE1, which has been shown to be overexpressed in a subgroup of ER-negative cancers. Although mRNA and protein expression are more prevalent than gene amplification, CCNE1 has been postulated as the driver gene of this amplicon [9, 2628]. There is evidence, however, that genes within this amplicon other than CCNE1 are consistently overexpressed when amplified [29], including POP4 and C19ORF2.
The aims of this study were (i) to characterise the 19q12 amplicon in breast cancer, (ii) to determine the genes that are overexpressed when amplified in this amplicon, (iii) to investigate which of the genes mapping to this amplicon are selectively required for the survival of cells harbouring their amplification, and (iv) to determine if cancer cells with CCNE1 gene amplification are dependent on CCNE1 cell cycle-related functions for their survival.

Material and methods

Tumour samples

Fresh/frozen primary invasive breast carcinomas from 313 patients were retrieved from independent consecutive cohorts after approval by local Research Ethic Committees from the authors' institutions. None of the patients received pre-operative chemotherapy and/or endocrine therapy. Tumours were either micro-dissected to ensure a tumour cell content > 75% as previously described [9], or one representative section of each tumour was stained with haematoxylin & eosin and only samples with > 70% of neoplastic cells were included, as previously described [19]. A description of the cohort analysed here is presented in Additional file 1 Table S1. Out of the samples included in this study, the aCGH profiles of 95, 24 and 8 cases were previously reported in Natrajan et al. [9], Turner et al. [19], and Hungermann et al. [30], respectively. The remaining samples comprise a series of 31 consecutive HER2-positive breast cancers retrieved from the tissue bank of The Netherlands Cancer Institute (NKI), and a collection of 119 consecutive breast cancers from the tissue bank of the NKI. RNA was available for 48 cases and the gene expression profiles were reported in Natrajan et al. [29]. RNA of sufficient quantity and quality was available for 48 cases and the gene expression profiles were reported in Natrajan et al. [29]. Immunohistochemistry for ER, progesterone receptor (PR) and HER2 was performed as previously described [9, 31]. Histological grade was determined by two pathologists (FCG and JSR-F) using the modified Scarff-Bloom-Richardson system [32]. ER and PR status was determined by immunohistochemical analysis using the 6F11 (1:150) and PgR636 (1:200) antibodies, respectively, as previously described [33]. The Allred scoring system was employed and tumours were considered positive when the score was ≥ 3. HER2 gene amplification was defined based on chromogenic in situ hybridisation analysis using a US Food and Drug Administrator approved probe (SpotLight HER2 amplification probe, Invitrogen, Carlsbad, CA, USA) and/or by inspection of the results of aCGH analysis, as previously described [20]. Tumours were classified into ER-positive/HER2-negative, ER-negative/HER2-negative and HER2-positive subgroups, given i) the results of recent studies demonstrating that the transcriptomic profiles of ER-positive/HER2-negative, ER-negative/HER2-negative and HER2-positive tumours are fundamentally different [34, 35], ii) the technical issues related to the assignment of tumour profiled with different platforms into the 'intrinsic' molecular subtypes [36, 37], and iii) that these subgroups are currently employed to define the systemic therapy for patients with breast cancer.

RNA and DNA extraction

DNA and RNA were extracted as previously described [29]. DNA concentration was measured with Picogreen® (Invitrogen, Paisley, UK) according to the manufacturer's instructions, and RNA was quantified using the Agilent 2100 Bioanalyser with RNA Nano LabChip Kits (Agilent Biosystems, Stockport, UK).

Cell lines

Fifty-six breast cancer cell lines were obtained from ATCC (LGC Standards, Teddington, UK) unless otherwise specified (Additional file 2 Table S2), maintained as previously described [16, 19, 38] and subjected to aCGH analysis as described below. Out of these cell lines, MDA-MB-157, HCC1569, MDA-MB-231, Hs578T, MCF7, ZR75.1, BT474 and JIMT1 were used as models for the functional characterisation of the 19q12 amplicon.

Microarray-based comparative genomic hybridisation (aCGH)

The 32K bacterial artificial chromosome (BAC) re-array collection (CHORI) tiling path aCGH platform was constructed at the Breakthrough Breast Cancer Research Centre, as described previously [33, 39]. This type of BAC array platform has been shown to be as robust as, and to have comparable resolution with, high density oligonucleotide arrays [4042]. DNA labelling, array hybridisations and image acquisition were performed as previously described [9]. aCGH data were pre-processed and analysed using an in-house R script (BACE.R) in R version 2.13.0, as previously described [29, 33]. In brief, raw Log2 ratios of intensity between samples and pooled female genomic DNA were read without background subtraction and normalised in the LIMMA package in R using PrinTipLoess. Outliers were removed based upon their deviation from neighbouring genomic probes using an estimation of the genome-wide median absolute deviation of all probes. After filtering polymorphic BACs a final dataset of 31,367 clones with unambiguous mapping information according to the February 2009 build (hg19) of the human genome [43]. Log2 ratios were rescaled using the genome-wide median absolute deviation in each sample, and then smoothed using circular binary segmentation in the DNACopy package as previously described [29, 33]. Loss was defined as a circular binary segmentation (cbs)-smoothed Log2 ratio < -0.08. Low-level gain was defined as a cbs-smoothed Log2 ratio in the range 0.08 to 0.45, corresponding to approximately three to five copies of the locus, whilst gene amplification was defined as having a Log2 ratio > 0.45, corresponding to more than five copies [9, 33, 39]. Threshold values were determined as previously described [9] and validated empirically by means of in situ hybridisation methods in previous publications [9, 33, 39, 4446]. A categorical analysis was applied to the BACs after classifying them as representing amplification (>0.45), gain (>0.08 and ≤ 0.045), loss (< -0.08), or no-change according to their cbs-smoothed Log2 ratio values [29, 33]. Threshold values were determined and validated as previously described [9]. aCGH data, the analysis history, script and code are publically available online at [47].

FISH validation of aCGH results

Fluorescence in situ hybridisation (FISH) analysis for 19q12 amplification was carried out as previously described [9, 48]. Briefly, a FISH probe mapping to 30.10 to 30.25 Mb on chromosome 19 was generated using the BAC RP11-372I05 and biotin labelled as previously described [48]. Pre-treatment and hybridisation were carried out as previously described [9, 48]. Cases were considered amplified if > 50% of neoplastic cells harboured large signal clusters or > 5 signals/nucleus. FISH performed with observers blinded to the results of aCGH analysis (Additional file 3 Figure S1).

TP53 mutation analysis

The entire coding sequence of the TP53 gene (exons 2 to 11) was sequenced as previously described [39, 49]. PCR amplification and Sanger sequencing was carried out in the 16 cases with 19q12 amplification using 5 ng of tumour DNA [50] using the DNA Sequencing Kit BigDye Terminator v 3.1 Cycle Sequencing Ready Reaction Mix (Applied Biosystems, Warrington, UK), as described [39]. Sequences were analysed with Mutation Surveyor software (Softgenetics, State College, PA, USA).

Gene expression analysis

Gene expression profiling of breast cancer samples in the 'Natrajan' dataset was performed using the Illumina human WG6 version 2 expression array as previously described [29]. Raw gene expression values were robust-spline normalised using the Bioconductor lumi package in R (R Foundation, Vienna, Austria) Genes were mapped to their genomic location using the lumiHumanAllv2 annotation database available from Bioconductor. Only Illumina transcript probes with detection P-values < 0.01 in > 25% of samples were included; this resulted in a dataset of 12,699 transcriptionally regulated probes with accurate and unequivocal mapping information. Gene-expression data are publicly available at ArrayExpress (accession number: E-TABM-543 [51]. The final dataset comprises 48 cases, whose clinicopathological characteristics are fully described elsewhere [29].

Identification of genes whose expression correlates with copy number changes

To identify genes whose expression levels correlate with copy number changes, cbs-smoothed Log2 ratios from aCGH data were used to assign the aCGH states for each of the genes in the gene expression dataset using the median values for all BACs that overlap with the genomic position of each gene. This resulted in a 1:1 matrix of expression values and aCGH cbs values, which were used for downstream statistical analysis. To define genes that were overexpressed when amplified, a Mann-Whitney U test was performed using categorical aCGH states (that is, amplification versus no amplification) as the grouping variable and the expression of genes as the dependent variable, as previously described [16, 29]. P-values < 0.05 were considered significant.

Short interfering RNA (siRNA)-mediated silencing

Nine genes that mapped to the smallest region of amplification on 19q12 were selected for functional evaluation, namely CCNE1, UQCRFS1, POP4, PLEKHF1, C19ORF2, C19ORF12, VSTM2B, ZNF536, TSHZ3. Each gene was targeted with four distinct siRNAs (siGENOME SMARTpool) obtained from Thermo Scientific (Epsom, UK): siCCNE1 - pool M-003213-02; siUQCRFS1 - pool M-020100-01; siPOP4 - pool M-020046-00; siPLEKHF1 - pool M-018423-01; siC19ORF2 - pool M-017399-01; siC19ORF12 - pool M-014731-01; siVSTM2B - pool M-023625-01; siZNF536 - pool M-020506-01; siTSHZ3 - pool M-014119-01; and siCDK2 - pool M-003236-04. siGENOME Non-Targeting siRNA Pool #1 and #2 (siCON, D-001206-13 and D-001206-14) were used as controls. As a positive control (that is, a gene whose silencing is lethal) we employed siRNA pools for PLK1 (M-003290-01) as previously described [44]. To identify conditions that maximise transfection efficiency and minimise lipid-mediated toxic effects, multiple transfection conditions for each cell line were tested. Lipofectamine RNAiMax (Invitrogen, Paisley, UK) and Dharmafect 4 (Thermo Scientific, Epsom, UK) were identified as the most efficient and least toxic for the cell lines studied. Breast cancer cells were transfected with target and control siRNAs (50 nmol/L per well in 100 μL total volume) in 96-well plates, according to the manufacturers' instructions as previously described [44, 52]. A total of 1,000 to 5,000 cells were seeded per well that yielded 80 to 90% confluency in the controls at 10 days. Media were replaced every two days and cell viability was assessed using the CellTiter-Glo® assay (Promega, Southampton, UK) as previously described [44]. The average cell survival fraction for each siRNA was calculated relative to that of cells transfected with non-coding control siRNA.

Drug sensitivity assays

For assessment of drug sensitivity, cell lines were plated and transfected with target or control siRNAs in 96-well plates. At 48 hours post transfection, media were supplemented with serial dilutions (10-4M to 10-9M) of doxorubicin, cisplatin, paclitaxel, or the CDK1, CDK2 and CDK9 inhibitor AZD5438 (Tocris Biosciences, Bristol, UK), essentially as previously described [44]. All experiments were performed in triplicate. Survival was assessed with CellTiter-Glo® cell viability assay after seven days of drug treatment and survival fraction compared to siRNA vehicle treated cells. Survival curves and estimated SF50 (the drug concentration used following which 50% of cells survive) were calculated using non-linear regression with GraphPad Prism V5.0 (La Jolla, CA, USA).

Cell cycle analysis

FACS analysis for cell cycle assessment was performed with Propidium Iodide staining five days after transfection as described previously [52].

Western blotting

Total protein lysates (40 μg) from treated and untreated cells were separated by SDS-PAGE according to standard protocols, and immunoblotting was carried out using primary antibodies directed against CCNE1 (HE-12, Abcam ab3927, Cambridge, UK), CDK2 (2546, Cell Signaling Technology, Danvers, MA, USA), phospho-CDK2 (2561, Cell Signaling), PLEKHF1 (20389-1-AP, Proteintech, Chicago, IL, USA), POP4 (Rpp29, sc-23048, Santa Cruz Biotechnology, Santa Cruz, CA, USA), TSHZ3 (sc-134132, Santa Cruz), and CCNB1 (4135, Cell Signaling), and against β-tubulin as loading control (5346, Cell Signaling). Protein expression levels after siRNA silencing were assessed by western blotting 72 hours after transfection of the cells and protein bands quantified using ImageJ 1.44p software (NIH, Bethesda, MD, USA) [53].

Real-time quantitative reverse transcriptase PCR (qRT-PCR)

First strand synthesis was performed as previously described [9], and qRT-PCR was performed using TaqMan chemistry on the ABI Prism 7900HT (Applied Biosystems), using the standard curve method. Assays for CCNE1, PLEKHF1, UQCRFS1, C19ORF12, C19ORF2, VSTM2B, ZNF536, TSHZ3, TFRC and MRPL19 were purchased from Applied Biosystems. Expression levels were normalised to those of TFRC and MRPL19 (Assay on demand ID: Hs00174609_m1-TFRC, Hs00608522_g1-MRPL19, Hs01026536_m1-CCNE1, Hs00759096_s1-PLEKHF1, Hs00705563_s1-UQCRFS1, Hs01026936_m1-C19ORF2, Hs00198357_m1-POP4, Hs01107514_m1-C19ORF12, Hs0416833_m1-VSTM2B, Hs00206981_m1-ZNF536, Hs01583885_m1-TSHZ3, Hs01548894_m1-CDK2, Hs99999188_m1-CCNB1). For the assessment of RNA levels after gene silencing, RNA was extracted using Trizol (Invitrogen, Paisley, UK), and transcript levels were assessed 48 hours after transfection. The level of siRNA silencing was measured relative to that of cells transfected with non-coding control siRNA.

Statistical analysis

All statistical analysis was performed with R version 2.13.0 or GraphPad Prism version 5.0. All statistical tests performed adopted 95% confidence intervals. A two-tailed P-value of < 0.05 was considered significant.

Results

19q12 amplification is associated with grade III ER-negative breast cancers

To determine the clinicopathological correlates of the 19q amplification, 313 frozen breast cancers were subjected to aCGH analysis [9]. Amplification at 19q12 (27.8 to 32.9 Mb) was found in 16/313 (5.1%) of all tumours, and 7.8% (16/188) of grade III cancers (Figure 1A-C; Table 1). Amplification of 19q12 was significantly associated with histological grade III (Fisher's exact test P = 0.018, Table 1), lack of ER expression (Fisher's exact test, P = 0.0042) and ER-negative/HER2-negative subtype (Chi-square test P = 0.0056, Table 1). Given the small number of samples in the subgroup analysis of the clinical subtypes, we have compared the prevalence of 19q12 amplification in ER-negative/HER2-negative vs the other subtypes using the Fisher's exact test, which confirmed a significance association between 19q12 amplification and ER-negative/HER2-negative breast cancers (P = 0.0024).
Table 1
19q12 amplification in 313 breast cancers and clinicopathological associations
  
19q12
Amp n (%)
19q12
Not amp n (%)
P-value
CCNE1
Amp (%)
CCNE1
Not amp (%)
P-value
Whole cohort
 
16 (5.1%)
297 (94.9%)
NA
5 (1.6%)
308 (98.4%)
NA
Histological grade*
   
0.0018 +
  
0.1693+
 
I/II
0 (0%)
109 (100%)
 
0 (0%)
109 (100%)
 
 
III
16 (8%)
188 (92%)
 
5 (2%)
199 (98%)
 
ER
   
0.0042 +
  
0.01984 +
 
Positive
3 (2%)
166 (98%)
 
0 (0%)
169 (100%)
 
 
Negative
13 (9%)
131 (91%)
 
5 (3.5%)
139 (96.5%)
 
HER2
   
0.5398 +
  
0.9801 +
 
Positive
2 (3%)
62 (97%)
 
1(1.6%)
63(98.4%)
 
 
Negative
14 (6%)
235 (94%)
 
4(1.6%)
245(98.4%)
 
Subtypes
   
0.0056**^
  
0.0500**^
 
ER-/HER2-
11 (11%)
90 (89%)
0.0024***
4 (4%)
97 (96%)
0.0399***
 
HER2
2 (3%)
62 (97%)
 
1 (2%)
63 (98%)
 
 
ER+/HER2-
3 (2%)
145 (98%)
 
0 (0%)
148 (100%)
 
Amp, amplified; CCNE1, cyclin E1; ER, oestrogen receptor; NA, not applicable; Not amp, not amplified.
*Histological grade determined by the modified Scarff-Bloom-Richardson system [32].
Significant P-values are highlighted in bold.
+: Fisher's exact test
**: Chi-square test
***: Comparison between ER-/HER2- vs the remaining subtypes by Fisher's exact test.
^: Given that > 20% of the groups have n < 5, the Chi-square P-value should be interpreted with caution.
Amplification of the 19q12 locus was shown to be complex and this amplicon displayed multiple core regions recurrently amplified in primary breast cancers (Figure 1C). Within these cores, nine genes were identified, including CCNE1 (Table 2). Although CCNE1 has been proposed as a driver of this amplicon in breast cancer [14, 26], CCNE1 was amplified in only 5 out of the 16 primary breast cancers harbouring amplification of the 19q12 locus (Figure 1C).
Table 2
Genes mapping to the 19q12 region of amplification
Gene symbol
Gene name
Position (kb)
Copy number and expression Pearson's correlation (r)
Overexpressed when amplified
(MWU) P-value
UQCRFS1
ubiquinol-cytochrome c reductase, Rieske iron-sulfur polypeptide 1
29,698 to 29,704
0.65*
0.0018
VSTM2B
V-set and transmembrane domain containing 2B
30,017 to 30,055
NA
NA
POP4
processing of precursor 4, ribonuclease P/MRP subunit (S. cerevisiae)
30,097 to 30,107
0.77*
0.0018
PLEKHF1
pleckstrin homology domain containing, family F (with FYVE domain) member 1
30,156 to 30,166
0.44*
0.0868
C19ORF12
Uncharacterised protein C19ORF12
30,190 to 30,206
0.63*
0.0018
CCNE1
Cyclin E1
30,303 to 30,315
0.66*
0.0071
C19ORF2
RPB5-mediating protein
30,433 to 30,507
0.81*
0.0001
ZNF536
Zinc finger protein 536
30,863 to 31,049
-0.03
0.6064
TSHZ3
Teashirt zinc finger homeobox 3
31,766 to 31,840
0.08
0.5426
Position refers to the genomic location from the Ensembl genome browser based on the February 2009 hg19 build of the genome
• P < 0.05; significant P-values are highlighted in bold; MWU (Mann-Whitney U test).

Potential 19q12 amplicon drivers

Genes within an amplicon that are overexpressed when amplified have been shown to constitute potential amplicon drivers and therapeutic targets [912, 1719, 29, 54]. To determine which of the amplified genes in the 19q12 amplicon were significantly overexpressed when amplified, aCGH and microarray expression data from 48 microdissected grade III breast cancers were interrogated [29, 36]. This analysis revealed that out of all the genes mapping to the 19q12 amplification with detectable gene expression, UQCRFS1, POP4, PLEKHF1, C19ORF12, CCNE1 and C19ORF2 had expression levels that significantly correlated with gene copy number (Pearson's correlation r value = 0.65, 0.77, 0.44, 0.63, 0.66, 0.81, respectively; all P < 0.05) and UQCRFS1, POP4, C19ORF12, CCNE1 and C19ORF2 were significantly overexpressed when amplified (Mann-Whitney U test P < 0.05) (Figure 1D, Table 2). VSTM2B expression was undetected in any of the tumours analysed.
To identify models for the study of the genes overexpressed when amplified in the 19q12 amplicon, we first subjected 56 breast cancer cell lines to aCGH analysis and identified two cell lines (that is, HCC1569 and MDA-MB-157), which harbour amplification of 19q12 (minimal amplified region from 29.17 to 32.82 Mb). In a way akin to the majority of primary breast cancers harbouring 19q12 amplification [9], the only cell lines harbouring amplification of this locus were ER-negative/HER2-negative (MDA-MB-157) or ER-negative/HER2-positive (HCC1569) [16, 38]. A panel of phenotypically matched (in terms of ER, PR and HER2 expression) breast cancer cell lines were selected and used as controls (that is, MDA-MB-231, Hs578T, MCF7, ZR75.1, JIMT1 and BT474; Additional file 2 Table S2). Quantitative RT-PCR of cDNA from cancer cells harbouring 19q12 amplification or lacking amplification of this locus revealed that UQCRFS1, POP4, CCNE1 and C19ORF2 genes were overexpressed when amplified (P < 0.05, Mann Whitney U test, Figure 1E), whereas VSTM2B, ZNF536 and TSHZ3 expression could not be detected in the majority of cell lines at the mRNA level.
Taken together, our results suggest that UQCRFS1, POP4, C19ORF12, CCNE1 and C19ORF2 are overexpressed in primary breast cancers and/or breast cancer cell lines harbouring their amplification, and may constitute potential drivers of this amplicon.

CCNE1, POP4, PLEKHF1 and TSHZ3 are selectively required in cells harbouring their amplification

There are several lines of evidence to suggest that one of the characteristics of amplicon drivers is that their expression is selectively required for the survival of cancer cells harbouring their amplification [912, 19]. Given the complexity of amplification at this locus, we chose to assess the potential amplicon drivers by siRNA silencing of all genes mapping to the 19q12 amplicon (that is, UQCRFS1, VSTM2B, POP4, C19ORF12, PLEKHF1, CCNE1, C19ORF2, ZNF536 and TSHZ3) regardless of their expression levels. Silencing of CCNE1, PLEKHF1, POP4, ZNF536 and TSHZ3 expression selectively reduced cell viability in cancer cells harbouring 19q12 gene amplification but had a significantly lower impact on the survival of breast cancer cells lacking amplification of this locus (t-test P < 0.05; Figure 2). Upon siRNA pool deconvolution, the observation that silencing of CCNE1, PLEKHF1, POP4 and TSHZ3 is selectively lethal in cancer cells harbouring their amplification was validated (that is, two or more individual siRNAs were selectively lethal in cancer cells harbouring amplification of the gene silenced; Figure 3A). siRNA silencing of these genes was confirmed at the mRNA and protein levels (Figures 3B, C).
Assessment of the protein levels of POP4, PLEKHF1, CCNE1 and TSHZ3 was performed in the cell lines used in this study. Expression levels of POP4, PLEKHF1 and CCNE1 were significantly higher in cancer cells harbouring amplification of the genes encoding these proteins than in cancer cells devoid of amplification of these loci (Figure 4, Mann-Whitney U test P < 0.05). No significant association between TSHZ3 gene amplification and protein expression was observed, even when corrected for proliferation levels, as defined by CCNB1 expression levels (data not shown).
To test whether silencing of these genes affects cell cycle in cancer cells with or without 19q12 amplification, DNA content was measured upon gene silencing. Upon PLEKHF1, POP4 or TSHZ3 siRNA-mediated gene silencing, slight but not statistically significant increases in the subG1 fraction and decreases in the G2/M fraction were observed in cancer cells with (that is, MDA-MB-157 and HCC1569) or without (that is, MDA-MB-231, JIMT1, ZR75.1 and BT474) 19q12 amplification (Additional file 4 Figure S2). These observations suggest that the silencing of these genes does not significantly alter the cell cycle profile. On the other hand, in cancer cells harbouring 19q12 amplification, CCNE1 silencing resulted in a significant decrease in the fraction of cells in S/G2 and increase in the proportion of cells in G1 and subG1 cell cycle phases (P < 0.0001, Chi-square test, Figure 5A).
To assess whether amplification and overexpression of CCNE1, PLEKHF1, POP4 and TSHZ3 would alter the response of breast cancer cells to common chemotherapeutic agents, we subjected cancer cells with or without 19q12 gene amplification to treatment with doxorubicin, cisplatin and paclitaxel upon silencing of these genes. No significant difference in the sensitivity of these cells to the chemotherapy agents tested was observed upon silencing of PLEKHF1, POP4 or TSHZ3 compared to non-targeting siRNAs (data not shown). CCNE1 siRNA-mediated silencing, however, was found to reduce the sensitivity of cancer cells to doxorubicin, cisplatin and paclitaxel in cancer cells harbouring 19q12 amplification cells but not in cancer cells lacking amplification of this locus (Additional file 5 Figure S3). This is not surprising, given that these chemotherapy agents target cycling cells and, here, we demonstrate that CCNE1 siRNA-mediated silencing leads to G1 arrest.

CDK2 silencing and chemical inhibition is selectively lethal in cancer cells harbouring CCNE1 gene amplification

Given that i) cancer cells harbouring CCNE1 gene amplification depend on CCNE1 expression for their survival, ii) that one of the main functions of CCNE1 is to activate CDK2 in the transition from G1/S phase [55], and iii) that CCNE1 RNAi-mediated silencing led to G1 arrest, we posited that cancer cells harbouring CCNE1 gene amplification would be dependent on CDK2 expression and kinase activity for their survival. siRNA-mediated CDK2 silencing resulted in a significantly higher reduction in cell survival of cancer cells harbouring CCNE1 gene amplification than in cancer cells devoid of amplification of this gene (t-test, P < 0.05, Figures 5B, C). Furthermore, cancer cells harbouring CCNE1 gene amplification displayed a higher sensitivity to the CDK1, CDK2 and CDK9 small molecule inhibitor AZD5438 [56, 57] than cancer cells lacking CCNE1 gene amplification (t-test, P = 0.019, Figure 5D). Moreover, primary breast cancers harbouring CCNE1 amplification were found to have higher levels of CDK2 mRNA expression, adjusted for proliferation using the mRNA levels of CCNB1 [58], than breast cancers devoid of CCNE1 amplification (P = 0.02789, t-test, data not shown).
Taken together, these data provide evidence that tumours harbouring 19q12 amplification have more than one 'driver' and suggest that cancer cells with CCNE1 amplification depend on CDK2 expression and kinase activity for their survival.

Discussion

Here we demonstrate that the 19q12 locus is amplified in 5.1% of all invasive breast cancers and that this amplification is preferentially found in grade III ER-negative breast cancers. This is consistent with previous observations that suggested that 19q12 amplification is found in 5 to 15% of breast cancers and is associated with ER-negative disease [9, 26, 27, 29]. Furthermore, through a combination of genomic profiling of primary breast cancers and breast cancer cell lines and RNAi experiments, we have demonstrated that the 19q12 amplicon may contain more than one 'driver'.
Previous studies have suggested that CCNE1 is the likeliest driver of the 19q12 amplicon [26, 27]. Although our results support the contention that CCNE1 is one of the drivers of this amplicon, in this study CCNE1 was amplified only in 5 out of 16 breast cancers harbouring 19q12 amplification. These observations are in agreement with those from studies of the 19q12 amplicon in gastric cancer, where CCNE1 was amplified only in a subset of cases harbouring 19q12 amplification [59], and with those of previous studies that have found a prevalence of CCNE1 amplification in 1.2% to 1.4% of primary breast cancers [14, 60]. Taken together, these lines of evidence suggest that CCNE1 may not be the sole driver of this amplicon and that other genes within the 19q12 amplicon may also constitute drivers. Consistent with this hypothesis, we observed that cancer cells harbouring 19q12 amplification require the expression of not only CCNE1, but also PLEKHF1, POP4 and TSHZ3 for their survival.
PLEKHF1 (pleckstrin homology domain containing, family F (with FYVE domain) member 1) encodes a protein that is known to induce apoptosis through the lysosomal-mitochondrial pathway and triggers caspase-independent apoptosis [61]. Recent evidence suggests that this process involves the recruitment of phosphorylated p53 and that silencing of endogenous p53 impairs its function [62]. Despite its reported role in apoptosis, PLEKHF1 siRNA-mediated silencing in cancer cells harbouring its amplification did not lead to an increase in the sub-G0 proportion of cells. It could be speculated that in a TP53 mutant background (as was the case of HCC1569 and MDA-MB-157; Additional file 2 Table S2), PLEKHF1 gene amplification and overexpression may confer a selective advantage through mechanisms other than through the recruitment of phosphorylated p53. Regrettably, no breast cancer cell lines harbouring PLEKHF1 gene amplification and wild-type TP53 were available to test this hypothesis. All primary breast cancers with PLEKHF1 amplification, however, were found to harbour TP53 mutations, suggesting that PLEKHF1 amplification and TP53 gene mutations may have epistatic interactions and that TP53 mutational status should be taken into account in the evaluation of the potential role of PLEKHF1 as a therapeutic target. POP4 (processing of precursor 4, ribonuclease P/MRP subunit (S. cerevisiae)) encodes one of the protein subunits of the small nucleolar ribonucleoprotein complexes and is involved in the processing of precursor RNAs. Further studies investigating the mechanisms that lead to a survival advantage in cancer cells harbouring amplification and overexpression of PLEKHF1 and POP4 are warranted. TSHZ3 (teashirt zinc finger homeobox 3) is a zinc finger transcription factor and has been shown to be involved in muscle cell differentiation [63, 64]. Given that the TSHZ3 gene promoter is reported to be frequently methylated in primary breast cancers and breast cancer cell lines [65], this gene has been suggested to display tumour suppressor function. Our results do not corroborate this hypothesis and suggest that THSZ3 is one of the drivers of the 19q12 amplicon, given that we demonstrate here that THSZ3 is amplified in 2.6% of breast cancers, and that its silencing is selectively lethal in cancer cells harbouring its amplification. Although protein expression is not directly correlated with amplification, the breast cancer cell lines harbouring 19q12 amplification displayed either the highest levels of THSZ3 protein expression (that is, MDA-MB-157) or the presence of a THSZ3 isoform (HCC1569, Figure 4).
Although CCNE1 amplification was restricted to a subset of cancers harbouring 19q12 amplification, we set out to investigate if breast cancer cells harbouring CCNE1 amplification would be selectively dependent on the expression of this gene for their survival. siRNA-mediated silencing of CCNE1 had a significantly greater effect on the survival of cancer cells harbouring CCNE1 amplification than in those lacking its amplification. This is in agreement with a recent study in ovarian cancer that demonstrated that reduction of CCNE1 expression significantly inhibited cell growth in CCNE1 expressing cells, with a more profound effect in ovarian cancer cells harbouring CCNE1 gene amplification [66]. Moreover, forced expression of CCNE1 in cells with low expression has been previously shown to result in increased cell proliferation [66]. Here we demonstrate that CCNE1 siRNA silencing in breast cancer cells harbouring CCNE1 gene amplification, but not in those lacking this amplification, resulted in a significant arrest in G1 (Figure 5 and Additional file 4 Figure S2). These findings provide a rationale for the apparent selective reduction in sensitivity to chemotherapy agents caused by CCNE1 siRNA silencing reported in ovarian cancer cells harbouring CCNE1 gene amplification [67], and described here in breast cancer cells harbouring CCNE1 gene amplification (Additional file 5 Figure S3).
Progression through the G1/S phase of the cell cycle is regulated through the partnership of CDK2 with its regulatory subunit CCNE1 [55]. Given that CCNE1 silencing in cancer cells harbouring CCNE1 gene amplification leads to G1 arrest, we tested whether these cells would be dependent on CDK2 expression and kinase activity for their survival. CDK2 siRNA silencing and inhibition of CDK2 kinase activity using a CDK1, CDK2 and CDK9 inhibitor (AZD5438) resulted in significantly higher reduction in survival of cancer cells harbouring CCNE1 gene amplification. This is in agreement with a recent study based on the analysis of the conditional mouse models MMTV-Low Molecular Weight (LMW)-Ccne1; Tp53+/-; Cdk2+/+, MMTV-LMW-Ccne1; Tp53+/-; Cdk2+/- and MMTV-LMW-Ccne1; Tp53+/-; Cdk2-/-. While mice with at least one functional copy of Cdk2 consistently developed mammary gland tumours, Cdk2-/- mice did not develop tumours through 24 months. Furthermore, administration of two Cdk inhibitors delayed the progression of mammary gland tumours in MMTV-LMW-Ccne1; Tp53+/-; Cdk2+/+ mice [68]. It should be noted that although MCF7 cells showed a similar sensitivity to the CDK1, CDK2 and CKD9 inhibitor AZD5438, CDK2 siRNA silencing had a significantly more limited impact on the viability of these cells, suggesting that the sensitivity of MCF7 cells to AZD5438 is unlikely to be caused by inhibition of CDK2. Taken together, these results demonstrate that breast cancer cells harbouring CCNE1 gene amplification are dependent on CDK2 expression and kinase activity for their survival and suggest that CCNE1 amplification may constitute a potential biomarker of sensitivity to CDK2 inhibitors. It should be noted, however, that CDK1, CDK2 and CDK9 inhibitors may also be efficacious in a subgroup of ER-positive breast cancers, given that MCF7 cells also show sensitivity to AZD5438. Analysis of the results of clinical trials testing CDK inhibitors in breast cancer patients (for example, "Maximum Tolerated Dose (MTD) of Liposomal Doxorubicin in Combination With Seliciclib for Patients With Metastatic Triple Negative Breast Cancer" trial; http://​clinicaltrial.​gov identifier NCT01333423) are warranted.

Conclusion

Here we have demonstrated that CCNE1 is a driver of the 19q12 amplicon and that cancer cells harbouring CCNE1 gene amplification display an increased sensitivity to CDK2 RNAi-mediated silencing and chemical inhibition. It should be noted, however, that CCNE1 was shown to be amplified only in a subset of breast cancers harbouring 19q12 amplification. siRNA silencing of all genes mapping to the 19q12 amplicon revealed the existence of genes other than CCNE1 whose expression is selectively required for the survival of cancer cells harbouring amplification of this locus. Our results suggest that drivers other than CCNE1 may exist in the 19q12 amplicon.

Acknowledgements

We thank Radost Vatcheva and Frederik Wallberg for technical assistance. This work was supported by Breakthrough Breast Cancer. BW is funded by a Cancer Research UK postdoctoral fellowship. Jorge S Reis-Filho is a recipient of the 2010 CRUK Future Leaders Prize. We acknowledge NHS funding to the NIHR Biomedical Research Centre.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

RN and JRF conceived the study. RN, DW, MBK, MA, KK, FCG and AL carried out the experiments. RN and AM analysed the data. HH, MVV, BK, FR and JP provided samples. RN, PW, BW and JRF wrote the manuscript. All authors read and edited the manuscript, and approved its final version.
Anhänge

Electronic supplementary material

Additional file 1: Table S1. Phenotypic characteristics of primary breast tumours analysed by microarray-based comparative genomic hybridisation. aCGH results previously reported, Natrajan et al. [9]., Hunngermann et al. [30], and Turner et al. [19]. Histological grade assessed by the modified Scarff-Bloom-Richardson system [32]. (PDF 1 MB)
Additional file 2: Table S2. Details of the 56 breast cancer cell lines subjected to microarray-based comparative genomic hybridisation. ER status, HER2 gene amplification status and p53 protein levels and mutational status (adapted from Neve et. al. [38], Arriola et al. [20] and [69] *p53 mRNA levels derived from Mackay et al. [16]. p53 protein levels and mutational status (obtained from COSMIC [70] and Neve et al. [38]). "Positive", detectable protein expression; MUT, mutant; "Negative", no detectable protein expression; WT, wild-type. (PDF 612 KB)
Literatur
1.
Zurück zum Zitat Reis-Filho JS, Pusztai L: Gene expression profiling in breast cancer: classification, prognostication, and prediction. Lancet. 2011, 378: 1812-1823. 10.1016/S0140-6736(11)61539-0.CrossRefPubMed Reis-Filho JS, Pusztai L: Gene expression profiling in breast cancer: classification, prognostication, and prediction. Lancet. 2011, 378: 1812-1823. 10.1016/S0140-6736(11)61539-0.CrossRefPubMed
2.
Zurück zum Zitat Sotiriou C, Pusztai L: Gene-expression signatures in breast cancer. N Engl J Med. 2009, 360: 790-800. 10.1056/NEJMra0801289.CrossRefPubMed Sotiriou C, Pusztai L: Gene-expression signatures in breast cancer. N Engl J Med. 2009, 360: 790-800. 10.1056/NEJMra0801289.CrossRefPubMed
3.
Zurück zum Zitat Weigelt B, Baehner FL, Reis-Filho JS: The contribution of gene expression profiling to breast cancer classification, prognostication and prediction: a retrospective of the last decade. J Pathol. 2010, 220: 263-280.CrossRefPubMed Weigelt B, Baehner FL, Reis-Filho JS: The contribution of gene expression profiling to breast cancer classification, prognostication and prediction: a retrospective of the last decade. J Pathol. 2010, 220: 263-280.CrossRefPubMed
4.
Zurück zum Zitat Colombo PE, Milanezi F, Weigelt B, Reis-Filho JS: Microarrays in the 2010s: the contribution of microarray-based gene expression profiling to breast cancer classification, prognostication and prediction. Breast Cancer Res. 2011, 13: 212-10.1186/bcr2890.CrossRefPubMedPubMedCentral Colombo PE, Milanezi F, Weigelt B, Reis-Filho JS: Microarrays in the 2010s: the contribution of microarray-based gene expression profiling to breast cancer classification, prognostication and prediction. Breast Cancer Res. 2011, 13: 212-10.1186/bcr2890.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Iwamoto T, Bianchini G, Booser D, Qi Y, Coutant C, Shiang CY, Santarpia L, Matsuoka J, Hortobagyi GN, Symmans WF, Holmes FA, O'Shaughnessy J, Hellerstedt B, Pippen J, Andre F, Simon R, Pusztai L: Gene pathways associated with prognosis and chemotherapy sensitivity in molecular subtypes of breast cancer. J Natl Cancer Inst. 2011, 103: 264-272. 10.1093/jnci/djq524.CrossRefPubMed Iwamoto T, Bianchini G, Booser D, Qi Y, Coutant C, Shiang CY, Santarpia L, Matsuoka J, Hortobagyi GN, Symmans WF, Holmes FA, O'Shaughnessy J, Hellerstedt B, Pippen J, Andre F, Simon R, Pusztai L: Gene pathways associated with prognosis and chemotherapy sensitivity in molecular subtypes of breast cancer. J Natl Cancer Inst. 2011, 103: 264-272. 10.1093/jnci/djq524.CrossRefPubMed
6.
Zurück zum Zitat Weigelt B, Pusztai L, Ashworth A, Reis-Filho JS: Challenges translating breast cancer gene signatures into the clinic. Nat Rev Clin Oncol. 2011, 9: 58-64. 10.1038/nrclinonc.2011.125.CrossRefPubMed Weigelt B, Pusztai L, Ashworth A, Reis-Filho JS: Challenges translating breast cancer gene signatures into the clinic. Nat Rev Clin Oncol. 2011, 9: 58-64. 10.1038/nrclinonc.2011.125.CrossRefPubMed
7.
Zurück zum Zitat Jonsson G, Staaf J, Vallon-Christersson J, Ringner M, Holm K, Hegardt C, Gunnarsson H, Fagerholm R, Strand C, Agnarsson BA, Kilpivaara O, Luts L, Heikkila P, Aittomaki K, Blomqvist C, Loman N, Malmstrom P, Olsson H, Johannsson OT, Arason A, Nevanlinna H, Barkardottir RB, Borg A: Genomic subtypes of breast cancer identified by array-comparative genomic hybridization display distinct molecular and clinical characteristics. Breast Cancer Res. 2010, 12: R42-10.1186/bcr2596.CrossRefPubMedPubMedCentral Jonsson G, Staaf J, Vallon-Christersson J, Ringner M, Holm K, Hegardt C, Gunnarsson H, Fagerholm R, Strand C, Agnarsson BA, Kilpivaara O, Luts L, Heikkila P, Aittomaki K, Blomqvist C, Loman N, Malmstrom P, Olsson H, Johannsson OT, Arason A, Nevanlinna H, Barkardottir RB, Borg A: Genomic subtypes of breast cancer identified by array-comparative genomic hybridization display distinct molecular and clinical characteristics. Breast Cancer Res. 2010, 12: R42-10.1186/bcr2596.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Stephens PJ, McBride DJ, Lin ML, Varela I, Pleasance ED, Simpson JT, Stebbings LA, Leroy C, Edkins S, Mudie LJ, Greenman CD, Jia M, Latimer C, Teague JW, Lau KW, Burton J, Quail MA, Swerdlow H, Churcher C, Natrajan R, Sieuwerts AM, Martens JW, Silver DP, Langerod A, Russnes HE, Foekens JA, Reis-Filho JS, van 't Veer L, Richardson AL, Borresen-Dale AL, et al: Complex landscapes of somatic rearrangement in human breast cancer genomes. Nature. 2009, 462: 1005-1010. 10.1038/nature08645.CrossRefPubMedPubMedCentral Stephens PJ, McBride DJ, Lin ML, Varela I, Pleasance ED, Simpson JT, Stebbings LA, Leroy C, Edkins S, Mudie LJ, Greenman CD, Jia M, Latimer C, Teague JW, Lau KW, Burton J, Quail MA, Swerdlow H, Churcher C, Natrajan R, Sieuwerts AM, Martens JW, Silver DP, Langerod A, Russnes HE, Foekens JA, Reis-Filho JS, van 't Veer L, Richardson AL, Borresen-Dale AL, et al: Complex landscapes of somatic rearrangement in human breast cancer genomes. Nature. 2009, 462: 1005-1010. 10.1038/nature08645.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Natrajan R, Lambros MB, Rodriguez-Pinilla SM, Moreno-Bueno G, Tan DS, Marchio C, Vatcheva R, Rayter S, Mahler-Araujo B, Fulford LG, Hungermann D, Mackay A, Grigoriadis A, Fenwick K, Tamber N, Hardisson D, Tutt A, Palacios J, Lord CJ, Buerger H, Ashworth A, Reis-Filho JS: Tiling path genomic profiling of grade 3 invasive ductal breast cancers. Clin Cancer Res. 2009, 15: 2711-2722. 10.1158/1078-0432.CCR-08-1878.CrossRefPubMed Natrajan R, Lambros MB, Rodriguez-Pinilla SM, Moreno-Bueno G, Tan DS, Marchio C, Vatcheva R, Rayter S, Mahler-Araujo B, Fulford LG, Hungermann D, Mackay A, Grigoriadis A, Fenwick K, Tamber N, Hardisson D, Tutt A, Palacios J, Lord CJ, Buerger H, Ashworth A, Reis-Filho JS: Tiling path genomic profiling of grade 3 invasive ductal breast cancers. Clin Cancer Res. 2009, 15: 2711-2722. 10.1158/1078-0432.CCR-08-1878.CrossRefPubMed
10.
Zurück zum Zitat Reis-Filho JS, Simpson PT, Turner NC, Lambros MB, Jones C, Mackay A, Grigoriadis A, Sarrio D, Savage K, Dexter T, Iravani M, Fenwick K, Weber B, Hardisson D, Schmitt FC, Palacios J, Lakhani SR, Ashworth A: FGFR1 emerges as a potential therapeutic target for lobular breast carcinomas. Clin Cancer Res. 2006, 12: 6652-6662. 10.1158/1078-0432.CCR-06-1164.CrossRefPubMed Reis-Filho JS, Simpson PT, Turner NC, Lambros MB, Jones C, Mackay A, Grigoriadis A, Sarrio D, Savage K, Dexter T, Iravani M, Fenwick K, Weber B, Hardisson D, Schmitt FC, Palacios J, Lakhani SR, Ashworth A: FGFR1 emerges as a potential therapeutic target for lobular breast carcinomas. Clin Cancer Res. 2006, 12: 6652-6662. 10.1158/1078-0432.CCR-06-1164.CrossRefPubMed
11.
Zurück zum Zitat Bernard-Pierrot I, Gruel N, Stransky N, Vincent-Salomon A, Reyal F, Raynal V, Vallot C, Pierron G, Radvanyi F, Delattre O: Characterization of the recurrent 8p11-12 amplicon identifies PPAPDC1B, a phosphatase protein, as a new therapeutic target in breast cancer. Cancer Res. 2008, 68: 7165-7175. 10.1158/0008-5472.CAN-08-1360.CrossRefPubMed Bernard-Pierrot I, Gruel N, Stransky N, Vincent-Salomon A, Reyal F, Raynal V, Vallot C, Pierron G, Radvanyi F, Delattre O: Characterization of the recurrent 8p11-12 amplicon identifies PPAPDC1B, a phosphatase protein, as a new therapeutic target in breast cancer. Cancer Res. 2008, 68: 7165-7175. 10.1158/0008-5472.CAN-08-1360.CrossRefPubMed
12.
Zurück zum Zitat Bocanegra M, Bergamaschi A, Kim YH, Miller MA, Rajput AB, Kao J, Langerod A, Han W, Noh DY, Jeffrey SS, Huntsman DG, Borresen-Dale AL, Pollack JR: Focal amplification and oncogene dependency of GAB2 in breast cancer. Oncogene. 2010, 29: 774-779. 10.1038/onc.2009.364.CrossRefPubMed Bocanegra M, Bergamaschi A, Kim YH, Miller MA, Rajput AB, Kao J, Langerod A, Han W, Noh DY, Jeffrey SS, Huntsman DG, Borresen-Dale AL, Pollack JR: Focal amplification and oncogene dependency of GAB2 in breast cancer. Oncogene. 2010, 29: 774-779. 10.1038/onc.2009.364.CrossRefPubMed
13.
Zurück zum Zitat Andre F, Job B, Dessen P, Tordai A, Michiels S, Liedtke C, Richon C, Yan K, Wang B, Vassal G, Delaloge S, Hortobagyi GN, Symmans WF, Lazar V, Pusztai L: Molecular characterization of breast cancer with high-resolution oligonucleotide comparative genomic hybridization array. Clin Cancer Res. 2009, 15: 441-451. 10.1158/1078-0432.CCR-08-1791.CrossRefPubMed Andre F, Job B, Dessen P, Tordai A, Michiels S, Liedtke C, Richon C, Yan K, Wang B, Vassal G, Delaloge S, Hortobagyi GN, Symmans WF, Lazar V, Pusztai L: Molecular characterization of breast cancer with high-resolution oligonucleotide comparative genomic hybridization array. Clin Cancer Res. 2009, 15: 441-451. 10.1158/1078-0432.CCR-08-1791.CrossRefPubMed
14.
Zurück zum Zitat Chin K, DeVries S, Fridlyand J, Spellman PT, Roydasgupta R, Kuo WL, Lapuk A, Neve RM, Qian Z, Ryder T, Chen F, Feiler H, Tokuyasu T, Kingsley C, Dairkee S, Meng Z, Chew K, Pinkel D, Jain A, Ljung BM, Esserman L, Albertson DG, Waldman FM, Gray JW: Genomic and transcriptional aberrations linked to breast cancer pathophysiologies. Cancer Cell. 2006, 10: 529-541. 10.1016/j.ccr.2006.10.009.CrossRefPubMed Chin K, DeVries S, Fridlyand J, Spellman PT, Roydasgupta R, Kuo WL, Lapuk A, Neve RM, Qian Z, Ryder T, Chen F, Feiler H, Tokuyasu T, Kingsley C, Dairkee S, Meng Z, Chew K, Pinkel D, Jain A, Ljung BM, Esserman L, Albertson DG, Waldman FM, Gray JW: Genomic and transcriptional aberrations linked to breast cancer pathophysiologies. Cancer Cell. 2006, 10: 529-541. 10.1016/j.ccr.2006.10.009.CrossRefPubMed
15.
Zurück zum Zitat Chin SF, Teschendorff AE, Marioni JC, Wang Y, Barbosa-Morais NL, Thorne NP, Costa JL, Pinder SE, van de Wiel MA, Green AR, Ellis IO, Porter PL, Tavare S, Brenton JD, Ylstra B, Caldas C: High-resolution aCGH and expression profiling identifies a novel genomic subtype of ER negative breast cancer. Genome Biol. 2007, 8: R215-10.1186/gb-2007-8-10-r215.CrossRefPubMedPubMedCentral Chin SF, Teschendorff AE, Marioni JC, Wang Y, Barbosa-Morais NL, Thorne NP, Costa JL, Pinder SE, van de Wiel MA, Green AR, Ellis IO, Porter PL, Tavare S, Brenton JD, Ylstra B, Caldas C: High-resolution aCGH and expression profiling identifies a novel genomic subtype of ER negative breast cancer. Genome Biol. 2007, 8: R215-10.1186/gb-2007-8-10-r215.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Mackay A, Tamber N, Fenwick K, Iravani M, Grigoriadis A, Dexter T, Lord CJ, Reis-Filho JS, Ashworth A: A high-resolution integrated analysis of genetic and expression profiles of breast cancer cell lines. Breast Cancer Res Treat. 2009, 118: 481-498. 10.1007/s10549-008-0296-7.CrossRefPubMed Mackay A, Tamber N, Fenwick K, Iravani M, Grigoriadis A, Dexter T, Lord CJ, Reis-Filho JS, Ashworth A: A high-resolution integrated analysis of genetic and expression profiles of breast cancer cell lines. Breast Cancer Res Treat. 2009, 118: 481-498. 10.1007/s10549-008-0296-7.CrossRefPubMed
17.
Zurück zum Zitat Holland DG, Burleigh A, Git A, Goldgraben MA, Perez-Mancera PA, Chin SF, Hurtado A, Bruna A, Ali HR, Greenwood W, Dunning MJ, Samarajiwa S, Menon S, Rueda OM, Lynch AG, McKinney S, Ellis IO, Eaves CJ, Carroll JS, Curtis C, Aparicio S, Caldas C: ZNF703 is a common Luminal B breast cancer oncogene that differentially regulates luminal and basal progenitors in human mammary epithelium. EMBO Mol Med. 2011, 3: 167-180. 10.1002/emmm.201100122.CrossRefPubMedPubMedCentral Holland DG, Burleigh A, Git A, Goldgraben MA, Perez-Mancera PA, Chin SF, Hurtado A, Bruna A, Ali HR, Greenwood W, Dunning MJ, Samarajiwa S, Menon S, Rueda OM, Lynch AG, McKinney S, Ellis IO, Eaves CJ, Carroll JS, Curtis C, Aparicio S, Caldas C: ZNF703 is a common Luminal B breast cancer oncogene that differentially regulates luminal and basal progenitors in human mammary epithelium. EMBO Mol Med. 2011, 3: 167-180. 10.1002/emmm.201100122.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Sircoulomb F, Nicolas N, Ferrari A, Finetti P, Bekhouche I, Rousselet E, Lonigro A, Adelaide J, Baudelet E, Esteyries S, Wicinski J, Audebert S, Charafe-Jauffret E, Jacquemier J, Lopez M, Borg JP, Sotiriou C, Popovici C, Bertucci F, Birnbaum D, Chaffanet M, Ginestier C: ZNF703 gene amplification at 8p12 specifies luminal B breast cancer. EMBO Mol Med. 2011, 3: 153-166. 10.1002/emmm.201100121.CrossRefPubMedPubMedCentral Sircoulomb F, Nicolas N, Ferrari A, Finetti P, Bekhouche I, Rousselet E, Lonigro A, Adelaide J, Baudelet E, Esteyries S, Wicinski J, Audebert S, Charafe-Jauffret E, Jacquemier J, Lopez M, Borg JP, Sotiriou C, Popovici C, Bertucci F, Birnbaum D, Chaffanet M, Ginestier C: ZNF703 gene amplification at 8p12 specifies luminal B breast cancer. EMBO Mol Med. 2011, 3: 153-166. 10.1002/emmm.201100121.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Turner N, Pearson A, Sharpe R, Lambros M, Geyer F, Lopez-Garcia MA, Natrajan R, Marchio C, Iorns E, Mackay A, Gillett C, Grigoriadis A, Tutt A, Reis-Filho JS, Ashworth A: FGFR1 amplification drives endocrine therapy resistance and is a therapeutic target in breast cancer. Cancer Res. 2010, 70: 2085-2094. 10.1158/0008-5472.CAN-09-3746.CrossRefPubMedPubMedCentral Turner N, Pearson A, Sharpe R, Lambros M, Geyer F, Lopez-Garcia MA, Natrajan R, Marchio C, Iorns E, Mackay A, Gillett C, Grigoriadis A, Tutt A, Reis-Filho JS, Ashworth A: FGFR1 amplification drives endocrine therapy resistance and is a therapeutic target in breast cancer. Cancer Res. 2010, 70: 2085-2094. 10.1158/0008-5472.CAN-09-3746.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Arriola E, Marchio C, Tan DS, Drury SC, Lambros MB, Natrajan R, Rodriguez-Pinilla SM, Mackay A, Tamber N, Fenwick K, Jones C, Dowsett M, Ashworth A, Reis-Filho JS: Genomic analysis of the HER2/TOP2A amplicon in breast cancer and breast cancer cell lines. Lab Invest. 2008, 88: 491-503. 10.1038/labinvest.2008.19.CrossRefPubMed Arriola E, Marchio C, Tan DS, Drury SC, Lambros MB, Natrajan R, Rodriguez-Pinilla SM, Mackay A, Tamber N, Fenwick K, Jones C, Dowsett M, Ashworth A, Reis-Filho JS: Genomic analysis of the HER2/TOP2A amplicon in breast cancer and breast cancer cell lines. Lab Invest. 2008, 88: 491-503. 10.1038/labinvest.2008.19.CrossRefPubMed
21.
Zurück zum Zitat Orsetti B, Nugoli M, Cervera N, Lasorsa L, Chuchana P, Ursule L, Nguyen C, Redon R, du Manoir S, Rodriguez C, Theillet C: Genomic and expression profiling of chromosome 17 in breast cancer reveals complex patterns of alterations and novel candidate genes. Cancer Res. 2004, 64: 6453-6460. 10.1158/0008-5472.CAN-04-0756.CrossRefPubMed Orsetti B, Nugoli M, Cervera N, Lasorsa L, Chuchana P, Ursule L, Nguyen C, Redon R, du Manoir S, Rodriguez C, Theillet C: Genomic and expression profiling of chromosome 17 in breast cancer reveals complex patterns of alterations and novel candidate genes. Cancer Res. 2004, 64: 6453-6460. 10.1158/0008-5472.CAN-04-0756.CrossRefPubMed
22.
Zurück zum Zitat Staaf J, Jonsson G, Ringner M, Vallon-Christersson J, Grabau D, Arason A, Gunnarsson H, Agnarsson BA, Malmstrom PO, Johannsson OT, Loman N, Barkardottir RB, Borg A: High-resolution genomic and expression analyses of copy number alterations in HER2-amplified breast cancer. Breast Cancer Res. 2010, 12: R25-10.1186/bcr2568.CrossRefPubMedPubMedCentral Staaf J, Jonsson G, Ringner M, Vallon-Christersson J, Grabau D, Arason A, Gunnarsson H, Agnarsson BA, Malmstrom PO, Johannsson OT, Loman N, Barkardottir RB, Borg A: High-resolution genomic and expression analyses of copy number alterations in HER2-amplified breast cancer. Breast Cancer Res. 2010, 12: R25-10.1186/bcr2568.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Kao J, Pollack JR: RNA interference-based functional dissection of the 17q12 amplicon in breast cancer reveals contribution of coamplified genes. Genes Chromosomes Cancer. 2006, 45: 761-769. 10.1002/gcc.20339.CrossRefPubMed Kao J, Pollack JR: RNA interference-based functional dissection of the 17q12 amplicon in breast cancer reveals contribution of coamplified genes. Genes Chromosomes Cancer. 2006, 45: 761-769. 10.1002/gcc.20339.CrossRefPubMed
24.
Zurück zum Zitat Shiu KK, Natrajan R, Geyer FC, Ashworth A, Reis-Filho JS: DNA amplifications in breast cancer: genotypic-phenotypic correlations. Future Oncol. 2010, 6: 967-984. 10.2217/fon.10.56.CrossRefPubMed Shiu KK, Natrajan R, Geyer FC, Ashworth A, Reis-Filho JS: DNA amplifications in breast cancer: genotypic-phenotypic correlations. Future Oncol. 2010, 6: 967-984. 10.2217/fon.10.56.CrossRefPubMed
25.
Zurück zum Zitat Streicher KL, Yang ZQ, Draghici S, Ethier SP: Transforming function of the LSM1 oncogene in human breast cancers with the 8p11-12 amplicon. Oncogene. 2007, 26: 2104-2114. 10.1038/sj.onc.1210002.CrossRefPubMed Streicher KL, Yang ZQ, Draghici S, Ethier SP: Transforming function of the LSM1 oncogene in human breast cancers with the 8p11-12 amplicon. Oncogene. 2007, 26: 2104-2114. 10.1038/sj.onc.1210002.CrossRefPubMed
26.
Zurück zum Zitat Adelaide J, Finetti P, Bekhouche I, Repellini L, Geneix J, Sircoulomb F, Charafe-Jauffret E, Cervera N, Desplans J, Parzy D, Schoenmakers E, Viens P, Jacquemier J, Birnbaum D, Bertucci F, Chaffanet M: Integrated profiling of basal and luminal breast cancers. Cancer Res. 2007, 67: 11565-11575. 10.1158/0008-5472.CAN-07-2536.CrossRefPubMed Adelaide J, Finetti P, Bekhouche I, Repellini L, Geneix J, Sircoulomb F, Charafe-Jauffret E, Cervera N, Desplans J, Parzy D, Schoenmakers E, Viens P, Jacquemier J, Birnbaum D, Bertucci F, Chaffanet M: Integrated profiling of basal and luminal breast cancers. Cancer Res. 2007, 67: 11565-11575. 10.1158/0008-5472.CAN-07-2536.CrossRefPubMed
27.
Zurück zum Zitat Moelans CB, de Weger RA, Monsuur HN, Vijzelaar R, van Diest PJ: Molecular profiling of invasive breast cancer by multiplex ligation-dependent probe amplification-based copy number analysis of tumor suppressor and oncogenes. Mod Pathol. 2010, 23: 1029-1039. 10.1038/modpathol.2010.84.CrossRefPubMed Moelans CB, de Weger RA, Monsuur HN, Vijzelaar R, van Diest PJ: Molecular profiling of invasive breast cancer by multiplex ligation-dependent probe amplification-based copy number analysis of tumor suppressor and oncogenes. Mod Pathol. 2010, 23: 1029-1039. 10.1038/modpathol.2010.84.CrossRefPubMed
28.
Zurück zum Zitat Sieuwerts AM, Look MP, Meijer-van Gelder ME, Timmermans M, Trapman AM, Garcia RR, Arnold M, Goedheer AJ, de Weerd V, Portengen H, Klijn JG, Foekens JA: Which cyclin E prevails as prognostic marker for breast cancer? Results from a retrospective study involving 635 lymph node-negative breast cancer patients. Clin Cancer Res. 2006, 12: 3319-3328. 10.1158/1078-0432.CCR-06-0225.CrossRefPubMed Sieuwerts AM, Look MP, Meijer-van Gelder ME, Timmermans M, Trapman AM, Garcia RR, Arnold M, Goedheer AJ, de Weerd V, Portengen H, Klijn JG, Foekens JA: Which cyclin E prevails as prognostic marker for breast cancer? Results from a retrospective study involving 635 lymph node-negative breast cancer patients. Clin Cancer Res. 2006, 12: 3319-3328. 10.1158/1078-0432.CCR-06-0225.CrossRefPubMed
29.
Zurück zum Zitat Natrajan R, Weigelt B, Mackay A, Geyer FC, Grigoriadis A, Tan DS, Jones C, Lord CJ, Vatcheva R, Rodriguez-Pinilla SM, Palacios J, Ashworth A, Reis-Filho JS: An integrative genomic and transcriptomic analysis reveals molecular pathways and networks regulated by copy number aberrations in basal-like, HER2 and luminal cancers. Breast Cancer Res Treat. 2010, 121: 575-589. 10.1007/s10549-009-0501-3.CrossRefPubMed Natrajan R, Weigelt B, Mackay A, Geyer FC, Grigoriadis A, Tan DS, Jones C, Lord CJ, Vatcheva R, Rodriguez-Pinilla SM, Palacios J, Ashworth A, Reis-Filho JS: An integrative genomic and transcriptomic analysis reveals molecular pathways and networks regulated by copy number aberrations in basal-like, HER2 and luminal cancers. Breast Cancer Res Treat. 2010, 121: 575-589. 10.1007/s10549-009-0501-3.CrossRefPubMed
30.
Zurück zum Zitat Hungermann D, Schmidt H, Natrajan R, Tidow N, Poos K, Reis-Filho JS, Brandt B, Buerger H, Korsching E: Influence of whole arm loss of chromosome 16q on gene expression patterns in oestrogen receptor-positive, invasive breast cancer. J Pathol. 2011, 224: 517-528. 10.1002/path.2938.CrossRefPubMed Hungermann D, Schmidt H, Natrajan R, Tidow N, Poos K, Reis-Filho JS, Brandt B, Buerger H, Korsching E: Influence of whole arm loss of chromosome 16q on gene expression patterns in oestrogen receptor-positive, invasive breast cancer. J Pathol. 2011, 224: 517-528. 10.1002/path.2938.CrossRefPubMed
31.
Zurück zum Zitat Kreike B, Hart G, Bartelink H, van de Vijver MJ: Analysis of breast cancer related gene expression using natural splines and the Cox proportional hazard model to identify prognostic associations. Breast Cancer Res Treat. 2010, 122: 711-720. 10.1007/s10549-009-0588-6.CrossRefPubMed Kreike B, Hart G, Bartelink H, van de Vijver MJ: Analysis of breast cancer related gene expression using natural splines and the Cox proportional hazard model to identify prognostic associations. Breast Cancer Res Treat. 2010, 122: 711-720. 10.1007/s10549-009-0588-6.CrossRefPubMed
32.
Zurück zum Zitat Elston CW, Ellis IO: Pathological prognostic factors in breast cancer. I. The value of histological grade in breast cancer: experience from a large study with long-term follow-up. Histopathology. 2002, 41: 154-161.CrossRefPubMed Elston CW, Ellis IO: Pathological prognostic factors in breast cancer. I. The value of histological grade in breast cancer: experience from a large study with long-term follow-up. Histopathology. 2002, 41: 154-161.CrossRefPubMed
33.
Zurück zum Zitat Lacroix-Triki M, Suarez PH, MacKay A, Lambros MB, Natrajan R, Savage K, Geyer FC, Weigelt B, Ashworth A, Reis-Filho JS: Mucinous carcinoma of the breast is genomically distinct from invasive ductal carcinomas of no special type. J Pathol. 2010, 222: 282-298. 10.1002/path.2763.CrossRefPubMed Lacroix-Triki M, Suarez PH, MacKay A, Lambros MB, Natrajan R, Savage K, Geyer FC, Weigelt B, Ashworth A, Reis-Filho JS: Mucinous carcinoma of the breast is genomically distinct from invasive ductal carcinomas of no special type. J Pathol. 2010, 222: 282-298. 10.1002/path.2763.CrossRefPubMed
34.
Zurück zum Zitat Wirapati P, Sotiriou C, Kunkel S, Farmer P, Pradervand S, Haibe-Kains B, Desmedt C, Ignatiadis M, Sengstag T, Schutz F, Goldstein DR, Piccart M, Delorenzi M: Meta-analysis of gene expression profiles in breast cancer: toward a unified understanding of breast cancer subtyping and prognosis signatures. Breast Cancer Res. 2008, 10: R65-10.1186/bcr2124.CrossRefPubMedPubMedCentral Wirapati P, Sotiriou C, Kunkel S, Farmer P, Pradervand S, Haibe-Kains B, Desmedt C, Ignatiadis M, Sengstag T, Schutz F, Goldstein DR, Piccart M, Delorenzi M: Meta-analysis of gene expression profiles in breast cancer: toward a unified understanding of breast cancer subtyping and prognosis signatures. Breast Cancer Res. 2008, 10: R65-10.1186/bcr2124.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Desmedt C, Haibe-Kains B, Wirapati P, Buyse M, Larsimont D, Bontempi G, Delorenzi M, Piccart M, Sotiriou C: Biological processes associated with breast cancer clinical outcome depend on the molecular subtypes. Clin Cancer Res. 2008, 14: 5158-5165. 10.1158/1078-0432.CCR-07-4756.CrossRefPubMed Desmedt C, Haibe-Kains B, Wirapati P, Buyse M, Larsimont D, Bontempi G, Delorenzi M, Piccart M, Sotiriou C: Biological processes associated with breast cancer clinical outcome depend on the molecular subtypes. Clin Cancer Res. 2008, 14: 5158-5165. 10.1158/1078-0432.CCR-07-4756.CrossRefPubMed
36.
Zurück zum Zitat Weigelt B, Mackay A, A'Hern R, Natrajan R, Tan DS, Dowsett M, Ashworth A, Reis-Filho JS: Breast cancer molecular profiling with single sample predictors: a retrospective analysis. Lancet Oncol. 2010, 11: 339-349. 10.1016/S1470-2045(10)70008-5.CrossRefPubMed Weigelt B, Mackay A, A'Hern R, Natrajan R, Tan DS, Dowsett M, Ashworth A, Reis-Filho JS: Breast cancer molecular profiling with single sample predictors: a retrospective analysis. Lancet Oncol. 2010, 11: 339-349. 10.1016/S1470-2045(10)70008-5.CrossRefPubMed
37.
Zurück zum Zitat Guedj M, Marisa L, de Reynies A, Orsetti B, Schiappa R, Bibeau F, Macgrogan G, Lerebours F, Finetti P, Longy M, Bertheau P, Bertrand F, Bonnet F, Martin AL, Feugeas JP, Bieche I, Lehmann-Che J, Lidereau R, Birnbaum D, Bertucci F, de Thé H, Theillet C: A refined molecular taxonomy of breast cancer. Oncogene. 2012, 31: 1196-1206. 10.1038/onc.2011.301.CrossRefPubMed Guedj M, Marisa L, de Reynies A, Orsetti B, Schiappa R, Bibeau F, Macgrogan G, Lerebours F, Finetti P, Longy M, Bertheau P, Bertrand F, Bonnet F, Martin AL, Feugeas JP, Bieche I, Lehmann-Che J, Lidereau R, Birnbaum D, Bertucci F, de Thé H, Theillet C: A refined molecular taxonomy of breast cancer. Oncogene. 2012, 31: 1196-1206. 10.1038/onc.2011.301.CrossRefPubMed
38.
Zurück zum Zitat Neve RM, Chin K, Fridlyand J, Yeh J, Baehner FL, Fevr T, Clark L, Bayani N, Coppe JP, Tong F, Speed T, Spellman PT, DeVries S, Lapuk A, Wang NJ, Kuo WL, Stilwell JL, Pinkel D, Albertson DG, Waldman FM, McCormick F, Dickson RB, Johnson MD, Lippman M, Ethier S, Gazdar A, Gray JW: A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes. Cancer Cell. 2006, 10: 515-527. 10.1016/j.ccr.2006.10.008.CrossRefPubMedPubMedCentral Neve RM, Chin K, Fridlyand J, Yeh J, Baehner FL, Fevr T, Clark L, Bayani N, Coppe JP, Tong F, Speed T, Spellman PT, DeVries S, Lapuk A, Wang NJ, Kuo WL, Stilwell JL, Pinkel D, Albertson DG, Waldman FM, McCormick F, Dickson RB, Johnson MD, Lippman M, Ethier S, Gazdar A, Gray JW: A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes. Cancer Cell. 2006, 10: 515-527. 10.1016/j.ccr.2006.10.008.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Geyer FC, Weigelt B, Natrajan R, Lambros MB, de Biase D, Vatcheva R, Savage K, Mackay A, Ashworth A, Reis-Filho JS: Molecular analysis reveals a genetic basis for the phenotypic diversity of metaplastic breast carcinomas. J Pathol. 2010, 220: 562-573. 10.1002/path.2675.CrossRefPubMed Geyer FC, Weigelt B, Natrajan R, Lambros MB, de Biase D, Vatcheva R, Savage K, Mackay A, Ashworth A, Reis-Filho JS: Molecular analysis reveals a genetic basis for the phenotypic diversity of metaplastic breast carcinomas. J Pathol. 2010, 220: 562-573. 10.1002/path.2675.CrossRefPubMed
40.
Zurück zum Zitat Coe BP, Ylstra B, Carvalho B, Meijer GA, Macaulay C, Lam WL: Resolving the resolution of array CGH. Genomics. 2007, 89: 647-653. 10.1016/j.ygeno.2006.12.012.CrossRefPubMed Coe BP, Ylstra B, Carvalho B, Meijer GA, Macaulay C, Lam WL: Resolving the resolution of array CGH. Genomics. 2007, 89: 647-653. 10.1016/j.ygeno.2006.12.012.CrossRefPubMed
41.
Zurück zum Zitat Gunnarsson R, Staaf J, Jansson M, Ottesen AM, Goransson H, Liljedahl U, Ralfkiaer U, Mansouri M, Buhl AM, Smedby KE, Hjalgrim H, Syvanen AC, Borg A, Isaksson A, Jurlander J, Juliusson G, Rosenquist R: Screening for copy-number alterations and loss of heterozygosity in chronic lymphocytic leukemia--a comparative study of four differently designed, high resolution microarray platforms. Genes Chromosomes Cancer. 2008, 47: 697-711. 10.1002/gcc.20575.CrossRefPubMed Gunnarsson R, Staaf J, Jansson M, Ottesen AM, Goransson H, Liljedahl U, Ralfkiaer U, Mansouri M, Buhl AM, Smedby KE, Hjalgrim H, Syvanen AC, Borg A, Isaksson A, Jurlander J, Juliusson G, Rosenquist R: Screening for copy-number alterations and loss of heterozygosity in chronic lymphocytic leukemia--a comparative study of four differently designed, high resolution microarray platforms. Genes Chromosomes Cancer. 2008, 47: 697-711. 10.1002/gcc.20575.CrossRefPubMed
42.
Zurück zum Zitat Tan DS, Lambros MB, Natrajan R, Reis-Filho JS: Getting it right: designing microarray (and not 'microawry') comparative genomic hybridization studies for cancer research. Lab Invest. 2007, 87: 737-754. 10.1038/labinvest.3700593.CrossRefPubMed Tan DS, Lambros MB, Natrajan R, Reis-Filho JS: Getting it right: designing microarray (and not 'microawry') comparative genomic hybridization studies for cancer research. Lab Invest. 2007, 87: 737-754. 10.1038/labinvest.3700593.CrossRefPubMed
44.
Zurück zum Zitat Wilkerson PM, Dedes KJ, Wetterskog D, Mackay A, Lambros MB, Mansour M, Frankum J, Lord CJ, Natrajan R, Ashworth A, Reis-Filho JS: Functional characterization of EMSY gene amplification in human cancers. J Pathol. 2011, 225: 29-42. 10.1002/path.2944.CrossRefPubMed Wilkerson PM, Dedes KJ, Wetterskog D, Mackay A, Lambros MB, Mansour M, Frankum J, Lord CJ, Natrajan R, Ashworth A, Reis-Filho JS: Functional characterization of EMSY gene amplification in human cancers. J Pathol. 2011, 225: 29-42. 10.1002/path.2944.CrossRefPubMed
45.
Zurück zum Zitat Marchio C, Lambros MB, Gugliotta P, Di Cantogno LV, Botta C, Pasini B, Tan DS, Mackay A, Fenwick K, Tamber N, Bussolati G, Ashworth A, Reis-Filho JS, Sapino A: Does chromosome 17 centromere copy number predict polysomy in breast cancer? A fluorescence in situ hybridization and microarray-based CGH analysis. J Pathol. 2009, 219: 16-24. 10.1002/path.2574.CrossRefPubMed Marchio C, Lambros MB, Gugliotta P, Di Cantogno LV, Botta C, Pasini B, Tan DS, Mackay A, Fenwick K, Tamber N, Bussolati G, Ashworth A, Reis-Filho JS, Sapino A: Does chromosome 17 centromere copy number predict polysomy in breast cancer? A fluorescence in situ hybridization and microarray-based CGH analysis. J Pathol. 2009, 219: 16-24. 10.1002/path.2574.CrossRefPubMed
46.
Zurück zum Zitat Duprez R, Wilkerson PM, Lacroix-Triki M, Lambros MB, Mackay A, Hern RA, Gauthier A, Pawar V, Colombo PE, Daley F, Natrajan R, Ward E, Macgrogan G, Arbion F, Michenet P, Weigelt B, Vincent-Salomon A, Reis-Filho JS: Immunophenotypic and genomic characterization of papillary carcinomas of the breast. J Pathol. 2012, 226: 427-441. 10.1002/path.3032.CrossRefPubMed Duprez R, Wilkerson PM, Lacroix-Triki M, Lambros MB, Mackay A, Hern RA, Gauthier A, Pawar V, Colombo PE, Daley F, Natrajan R, Ward E, Macgrogan G, Arbion F, Michenet P, Weigelt B, Vincent-Salomon A, Reis-Filho JS: Immunophenotypic and genomic characterization of papillary carcinomas of the breast. J Pathol. 2012, 226: 427-441. 10.1002/path.3032.CrossRefPubMed
48.
Zurück zum Zitat Lambros MB, Simpson PT, Jones C, Natrajan R, Westbury C, Steele D, Savage K, Mackay A, Schmitt FC, Ashworth A, Reis-Filho JS: Unlocking pathology archives for molecular genetic studies: a reliable method to generate probes for chromogenic and fluorescent in situ hybridization. Lab Invest. 2006, 86: 398-408. 10.1038/labinvest.3700390.CrossRefPubMed Lambros MB, Simpson PT, Jones C, Natrajan R, Westbury C, Steele D, Savage K, Mackay A, Schmitt FC, Ashworth A, Reis-Filho JS: Unlocking pathology archives for molecular genetic studies: a reliable method to generate probes for chromogenic and fluorescent in situ hybridization. Lab Invest. 2006, 86: 398-408. 10.1038/labinvest.3700390.CrossRefPubMed
49.
Zurück zum Zitat Zhou W, Muggerud AA, Vu P, Due EU, Sørlie T, Børresen-Dale AL, Warnberg F, Langerod A: Full sequencing of TP53 identifies identical mutations within in situ and invasive components in breast cancer suggesting clonal evolution. Mol Oncol. 2009, 3: 214-219. 10.1016/j.molonc.2009.03.001.CrossRefPubMed Zhou W, Muggerud AA, Vu P, Due EU, Sørlie T, Børresen-Dale AL, Warnberg F, Langerod A: Full sequencing of TP53 identifies identical mutations within in situ and invasive components in breast cancer suggesting clonal evolution. Mol Oncol. 2009, 3: 214-219. 10.1016/j.molonc.2009.03.001.CrossRefPubMed
50.
Zurück zum Zitat Armstrong AB, Wang M, Eble JN, MacLennan GT, Montironi R, Tan PH, Lopez-Beltran A, Zhang S, Baldridge LA, Spartz H, Cheng L: TP53 mutational analysis supports monoclonal origin of biphasic sarcomatoid urothelial carcinoma (carcinosarcoma) of the urinary bladder. Mod Pathol. 2009, 22: 113-118. 10.1038/modpathol.2008.176.CrossRefPubMed Armstrong AB, Wang M, Eble JN, MacLennan GT, Montironi R, Tan PH, Lopez-Beltran A, Zhang S, Baldridge LA, Spartz H, Cheng L: TP53 mutational analysis supports monoclonal origin of biphasic sarcomatoid urothelial carcinoma (carcinosarcoma) of the urinary bladder. Mod Pathol. 2009, 22: 113-118. 10.1038/modpathol.2008.176.CrossRefPubMed
52.
Zurück zum Zitat Turner NC, Lord CJ, Iorns E, Brough R, Swift S, Elliott R, Rayter S, Tutt AN, Ashworth A: A synthetic lethal siRNA screen identifying genes mediating sensitivity to a PARP inhibitor. EMBO J. 2008, 27: 1368-1377. 10.1038/emboj.2008.61.CrossRefPubMedPubMedCentral Turner NC, Lord CJ, Iorns E, Brough R, Swift S, Elliott R, Rayter S, Tutt AN, Ashworth A: A synthetic lethal siRNA screen identifying genes mediating sensitivity to a PARP inhibitor. EMBO J. 2008, 27: 1368-1377. 10.1038/emboj.2008.61.CrossRefPubMedPubMedCentral
54.
Zurück zum Zitat Cheng KW, Lahad JP, Kuo WL, Lapuk A, Yamada K, Auersperg N, Liu J, Smith-McCune K, Lu KH, Fishman D, Gray JW, Mills GB: The RAB25 small GTPase determines aggressiveness of ovarian and breast cancers. Nat Med. 2004, 10: 1251-1256. 10.1038/nm1125.CrossRefPubMed Cheng KW, Lahad JP, Kuo WL, Lapuk A, Yamada K, Auersperg N, Liu J, Smith-McCune K, Lu KH, Fishman D, Gray JW, Mills GB: The RAB25 small GTPase determines aggressiveness of ovarian and breast cancers. Nat Med. 2004, 10: 1251-1256. 10.1038/nm1125.CrossRefPubMed
55.
Zurück zum Zitat Malumbres M, Barbacid M: Cell cycle, CDKs and cancer: a changing paradigm. Nat Rev Cancer. 2009, 9: 153-166. 10.1038/nrc2602.CrossRefPubMed Malumbres M, Barbacid M: Cell cycle, CDKs and cancer: a changing paradigm. Nat Rev Cancer. 2009, 9: 153-166. 10.1038/nrc2602.CrossRefPubMed
56.
Zurück zum Zitat Anderson M, Andrews DM, Barker AJ, Brassington CA, Breed J, Byth KF, Culshaw JD, Finlay MR, Fisher E, McMiken HH, Green CP, Heaton DW, Nash IA, Newcombe NJ, Oakes SE, Pauptit RA, Roberts A, Stanway JJ, Thomas AP, Tucker JA, Walker M, Weir HM: Imidazoles: SAR and development of a potent class of cyclin-dependent kinase inhibitors. Bioorg Med Chem Lett. 2008, 18: 5487-5492. 10.1016/j.bmcl.2008.09.024.CrossRefPubMed Anderson M, Andrews DM, Barker AJ, Brassington CA, Breed J, Byth KF, Culshaw JD, Finlay MR, Fisher E, McMiken HH, Green CP, Heaton DW, Nash IA, Newcombe NJ, Oakes SE, Pauptit RA, Roberts A, Stanway JJ, Thomas AP, Tucker JA, Walker M, Weir HM: Imidazoles: SAR and development of a potent class of cyclin-dependent kinase inhibitors. Bioorg Med Chem Lett. 2008, 18: 5487-5492. 10.1016/j.bmcl.2008.09.024.CrossRefPubMed
57.
Zurück zum Zitat Byth KF, Thomas A, Hughes G, Forder C, McGregor A, Geh C, Oakes S, Green C, Walker M, Newcombe N, Green S, Growcott J, Barker A, Wilkinson RW: AZD5438, a potent oral inhibitor of cyclin-dependent kinases 1, 2, and 9, leads to pharmacodynamic changes and potent antitumor effects in human tumor xenografts. Mol Cancer Ther. 2009, 8: 1856-1866. 10.1158/1535-7163.MCT-08-0836.CrossRefPubMed Byth KF, Thomas A, Hughes G, Forder C, McGregor A, Geh C, Oakes S, Green C, Walker M, Newcombe N, Green S, Growcott J, Barker A, Wilkinson RW: AZD5438, a potent oral inhibitor of cyclin-dependent kinases 1, 2, and 9, leads to pharmacodynamic changes and potent antitumor effects in human tumor xenografts. Mol Cancer Ther. 2009, 8: 1856-1866. 10.1158/1535-7163.MCT-08-0836.CrossRefPubMed
58.
Zurück zum Zitat Turner NC, Reis-Filho JS, Russell AM, Springall RJ, Ryder K, Steele D, Savage K, Gillett CE, Schmitt FC, Ashworth A, Tutt AN: BRCA1 dysfunction in sporadic basal-like breast cancer. Oncogene. 2007, 26: 2126-2132. 10.1038/sj.onc.1210014.CrossRefPubMed Turner NC, Reis-Filho JS, Russell AM, Springall RJ, Ryder K, Steele D, Savage K, Gillett CE, Schmitt FC, Ashworth A, Tutt AN: BRCA1 dysfunction in sporadic basal-like breast cancer. Oncogene. 2007, 26: 2126-2132. 10.1038/sj.onc.1210014.CrossRefPubMed
59.
Zurück zum Zitat Leung SY, Ho C, Tu IP, Li R, So S, Chu KM, Yuen ST, Chen X: Comprehensive analysis of 19q12 amplicon in human gastric cancers. Mod Pathol. 2006, 19: 854-863.PubMed Leung SY, Ho C, Tu IP, Li R, So S, Chu KM, Yuen ST, Chen X: Comprehensive analysis of 19q12 amplicon in human gastric cancers. Mod Pathol. 2006, 19: 854-863.PubMed
60.
Zurück zum Zitat Courjal F, Cuny M, Simony-Lafontaine J, Louason G, Speiser P, Zeillinger R, Rodriguez C, Theillet C: Mapping of DNA amplifications at 15 chromosomal localizations in 1875 breast tumors: definition of phenotypic groups. Cancer Res. 1997, 57: 4360-4367.PubMed Courjal F, Cuny M, Simony-Lafontaine J, Louason G, Speiser P, Zeillinger R, Rodriguez C, Theillet C: Mapping of DNA amplifications at 15 chromosomal localizations in 1875 breast tumors: definition of phenotypic groups. Cancer Res. 1997, 57: 4360-4367.PubMed
61.
Zurück zum Zitat Chen W, Li N, Chen T, Han Y, Li C, Wang Y, He W, Zhang L, Wan T, Cao X: The lysosome-associated apoptosis-inducing protein containing the pleckstrin homology (PH) and FYVE domains (LAPF), representative of a novel family of PH and FYVE domain-containing proteins, induces caspase-independent apoptosis via the lysosomal-mitochondrial pathway. J Biol Chem. 2005, 280: 40985-40995. 10.1074/jbc.M502190200.CrossRefPubMed Chen W, Li N, Chen T, Han Y, Li C, Wang Y, He W, Zhang L, Wan T, Cao X: The lysosome-associated apoptosis-inducing protein containing the pleckstrin homology (PH) and FYVE domains (LAPF), representative of a novel family of PH and FYVE domain-containing proteins, induces caspase-independent apoptosis via the lysosomal-mitochondrial pathway. J Biol Chem. 2005, 280: 40985-40995. 10.1074/jbc.M502190200.CrossRefPubMed
62.
Zurück zum Zitat Li N, Zheng Y, Chen W, Wang C, Liu X, He W, Xu H, Cao X: Adaptor protein LAPF recruits phosphorylated p53 to lysosomes and triggers lysosomal destabilization in apoptosis. Cancer Res. 2007, 67: 11176-11185. 10.1158/0008-5472.CAN-07-2333.CrossRefPubMed Li N, Zheng Y, Chen W, Wang C, Liu X, He W, Xu H, Cao X: Adaptor protein LAPF recruits phosphorylated p53 to lysosomes and triggers lysosomal destabilization in apoptosis. Cancer Res. 2007, 67: 11176-11185. 10.1158/0008-5472.CAN-07-2333.CrossRefPubMed
63.
Zurück zum Zitat Caubit X, Lye CM, Martin E, Core N, Long DA, Vola C, Jenkins D, Garratt AN, Skaer H, Woolf AS, Fasano L: Teashirt 3 is necessary for ureteral smooth muscle differentiation downstream of SHH and BMP4. Development. 2008, 135: 3301-3310. 10.1242/dev.022442.CrossRefPubMed Caubit X, Lye CM, Martin E, Core N, Long DA, Vola C, Jenkins D, Garratt AN, Skaer H, Woolf AS, Fasano L: Teashirt 3 is necessary for ureteral smooth muscle differentiation downstream of SHH and BMP4. Development. 2008, 135: 3301-3310. 10.1242/dev.022442.CrossRefPubMed
64.
Zurück zum Zitat Faralli H, Martin E, Core N, Liu QC, Filippi P, Dilworth FJ, Caubit X, Fasano L: Teashirt-3, a novel regulator of muscle differentiation, associates with BRG-1 associated factor 57 (BAF57) to inhibit myogenin gene expression. J Biol Chem. 2011 Faralli H, Martin E, Core N, Liu QC, Filippi P, Dilworth FJ, Caubit X, Fasano L: Teashirt-3, a novel regulator of muscle differentiation, associates with BRG-1 associated factor 57 (BAF57) to inhibit myogenin gene expression. J Biol Chem. 2011
65.
Zurück zum Zitat Yamamoto M, Cid E, Bru S, Yamamoto F: Rare and frequent promoter methylation, respectively, of TSHZ2 and 3 genes that are both downregulated in expression in breast and prostate cancers. PLoS One. 2011, 6: e17149-10.1371/journal.pone.0017149.CrossRefPubMedPubMedCentral Yamamoto M, Cid E, Bru S, Yamamoto F: Rare and frequent promoter methylation, respectively, of TSHZ2 and 3 genes that are both downregulated in expression in breast and prostate cancers. PLoS One. 2011, 6: e17149-10.1371/journal.pone.0017149.CrossRefPubMedPubMedCentral
66.
Zurück zum Zitat Nakayama N, Nakayama K, Shamima Y, Ishikawa M, Katagiri A, Iida K, Miyazaki K: Gene amplification CCNE1 is related to poor survival and potential therapeutic target in ovarian cancer. Cancer. 2010, 116: 2621-2634.CrossRefPubMed Nakayama N, Nakayama K, Shamima Y, Ishikawa M, Katagiri A, Iida K, Miyazaki K: Gene amplification CCNE1 is related to poor survival and potential therapeutic target in ovarian cancer. Cancer. 2010, 116: 2621-2634.CrossRefPubMed
67.
Zurück zum Zitat Etemadmoghadam D, George J, Cowin PA, Cullinane C, Kansara M, Gorringe KL, Smyth GK, Bowtell DD: Amplicon-dependent CCNE1 expression is critical for clonogenic survival after cisplatin treatment and is correlated with 20q11 gain in ovarian cancer. PLoS One. 2010, 5: e15498-10.1371/journal.pone.0015498.CrossRefPubMedPubMedCentral Etemadmoghadam D, George J, Cowin PA, Cullinane C, Kansara M, Gorringe KL, Smyth GK, Bowtell DD: Amplicon-dependent CCNE1 expression is critical for clonogenic survival after cisplatin treatment and is correlated with 20q11 gain in ovarian cancer. PLoS One. 2010, 5: e15498-10.1371/journal.pone.0015498.CrossRefPubMedPubMedCentral
68.
Zurück zum Zitat Akli S, Van Pelt CS, Bui T, Meijer L, Keyomarsi K: Cdk2 is required for breast cancer mediated by the low-molecular-weight isoform of Cyclin E. Cancer Res. 2011, 71: 3377-3386. 10.1158/0008-5472.CAN-10-4086.CrossRefPubMedPubMedCentral Akli S, Van Pelt CS, Bui T, Meijer L, Keyomarsi K: Cdk2 is required for breast cancer mediated by the low-molecular-weight isoform of Cyclin E. Cancer Res. 2011, 71: 3377-3386. 10.1158/0008-5472.CAN-10-4086.CrossRefPubMedPubMedCentral
Metadaten
Titel
Functional characterization of the 19q12 amplicon in grade III breast cancers
verfasst von
Rachael Natrajan
Alan Mackay
Paul M Wilkerson
Maryou B Lambros
Daniel Wetterskog
Monica Arnedos
Kai-Keen Shiu
Felipe C Geyer
Anita Langerød
Bas Kreike
Fabien Reyal
Hugo M Horlings
Marc J van de Vijver
Jose Palacios
Britta Weigelt
Jorge S Reis-Filho
Publikationsdatum
01.04.2012
Verlag
BioMed Central
Erschienen in
Breast Cancer Research / Ausgabe 2/2012
Elektronische ISSN: 1465-542X
DOI
https://doi.org/10.1186/bcr3154

Weitere Artikel der Ausgabe 2/2012

Breast Cancer Research 2/2012 Zur Ausgabe

Update Onkologie

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