Skip to main content
Erschienen in: BMC Cancer 1/2012

Open Access 01.12.2012 | Research article

Human pancreatic adenocarcinoma contains a side population resistant to gemcitabine

verfasst von: Anke Van den broeck, Lies Gremeaux, Baki Topal, Hugo Vankelecom

Erschienen in: BMC Cancer | Ausgabe 1/2012

Abstract

Background

Therapy resistance remains one of the major challenges to improve the prognosis of patients with pancreatic cancer. Chemoresistant cells, which potentially also display cancer stem cell (CSC) characteristics, can be isolated using the side population (SP) technique. Our aim was to search for a SP in human pancreatic ductal adenocarcinoma (PDAC) and to examine its chemoresistance and CSC(−like) phenotype.

Methods

Human PDAC samples were expanded in immunodeficient mice and first-generation xenografts analyzed for the presence of a Hoechst dye-effluxing SP using flow cytometry (FACS). To investigate chemoresistance of the SP, mice bearing PDAC xenografts were treated with gemcitabine and SP proportion determined. In addition, the SP and the main tumour cell population (MP) were sorted by FACS for RNA extraction to profile gene expression, and for culturing under sphere-forming conditions.

Results

A SP was identified in all PDAC samples, analyzed. This SP was more resistant to gemcitabine than the other tumour cells as examined in vivo. Whole-genome expression profiling of the SP revealed upregulation of genes related to therapy resistance, apoptotic regulation and epithelial-mesenchymal transition. In addition, the SP displayed higher tumourigenic (CSC) activity than the MP as analyzed in vitro by sphere-forming capacity.

Conclusion

We identified a SP in human PDAC and uncovered a chemoresistant and CSC-associated phenotype. This SP may represent a new therapeutic target in pancreatic cancer.

Trial registration

Clinicaltrials.gov NCT00936104
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1471-2407-12-354) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests

Authors’ contributions

AVDB designed and performed the study, analyzed the data and wrote the manuscript. LG trained AVDB in the techniques used and helped in data collection. BT participated in study design and analysis and in writing. HV participated in the design and analysis of the study and wrote the manuscript. All authors read and approved the final manuscript.
Abkürzungen
PDAC
Pancreatic ductal adenocarcinoma
SP
Side population
MP
Main population
FACS
Fluorescence-activated cell sorter
CSC
Cancer stem cell(s)
s.c.
Subcutaneously
SCID
Severe combined immunodeficiency
DAVID
Database for annotation: Visualization and Integrated Discovery
KEGG
Kyoto Encyclopedia of Genes and Genomes
STRING
Search Tool for the Retrieval of Interacting Genes/Proteins
EMT
Epithelial-mesenchymal transition.

Background

Pancreatic cancer or ductal adenocarcinoma (PDAC) remains a highly lethal disease. Despite improvements in medical and surgical care, the overall 5-year survival still has not exceeded 5% [1]. Resistance to chemotherapy is a major cause of treatment failure in pancreatic cancer, both in adjuvant setting after intended curative surgery as well as in advanced inoperable stages [2]. Therefore, PDAC’s chemoresistant cells are highly wanted targets for new therapeutic strategies to eventually improve overall survival.
Therapeutic resistance in pancreatic cancer is caused by low permeability of the tumour micro-environment, as well as by the efficient efflux of toxic agents [3, 4]. One approach to isolate drug-effluxing cells is provided by the side population (SP) technique [5]. SP cells are identified on the basis of Hoechst-dye efflux capacity because of the presence of multidrug resistance transporters. Cells that expel the dye are visualized by dual-wavelength flow cytometry (FACS) as a ‘Hoechst low’ tail of cells, the SP, relative to a larger bulk of ‘Hoechst high’ cells, the main population (MP).
Recently, a SP has been identified in cultured pancreatic cancer cell lines and was found to be chemoresistant to gemcitabine treatment as evaluated in vitro in these cultured cell lines [3, 68]. To date, it is not known whether clinical human PDAC contains a SP and wether this SP is resistant to gemcitabine when assessed in vivo.
In multiple types of cancer the SP is enriched in cells displaying properties of cancer stem cells (CSC) [9, 10]. By definition, CSC (also referred to as tumour-driving cells) represent the tumour’s subpopulation with the highest capacity to drive its growth, invasion and metastasis. CSC are also considered responsible for therapy resistance and disease recurrence [11], and may therefore represent interesting targets for new and more effective treatment strategies [12]. Although still subject of debate, (candidate) CSC populations are being identified in a growing number of cancer types and their functional significance has recently been strongly supported [1315]. In pancreatic cancer, cells based on specific cell-surface markers (i.e. CD24+CD44+ESA+ and CD133+ cells) have been reported to possess CSC characteristics [12, 16, 17].
In the present study, we report for the first time that human PDAC contains a SP and analyzed its resistance to the current standard chemotherapeutic agent for pancreatic cancer, gemcitabine, using a PDAC xenograft in vivo model. In addition, we performed whole-genome expression analysis of PDAC SP cells, which may guide to CSC-associated characteristics and to potential therapeutic targets. Finally, we explored in vitro whether the PDAC SP is enriched in tumourigenic cells as a further characteristic of CSC.

Methods

PDAC samples and xenografts

Between 2007 and 2010, PDAC surgical resection specimens were obtained at the University Hospital Leuven (Belgium) from patients after written informed consent (see Table 1). The study was approved by the KU Leuven ethical committee prior to patient recruitment, and received the study number ML3452. Freshly resected tumours were cut into small pieces (2*2 mm) and implanted subcutaneously (s.c.) in the axilla of severe combined immunodeficiency (SCID) mice (male, 6–10 weeks old) to expand tumour material. Tumour growth was evaluated with a caliper on a weekly basis and volume calculated according to the formula: tumour volume = (length x width2)/2 [18]. Mice bearing tumours with a minimum volume of 150 mm3 were euthanized and tumours were dissected for further analysis. Only first-generation xenograft tumours were used in the experiments described. Hematoxylin-Eosin staining was performed on formalin-fixed sections from original and xenograft tumours.
Table 1
Patients’ characteristics with PDAC used for xenografting
Xenograft no.
Sex
Age (y)
pG
pT
pN
pM
pR
PNI
VI
LVI
Preop RCT
Postop RCT/CT
OS (m)
DFS (m)
SP%
101*
F
45.5
2
4
1
0
1
1
0
0
0
RCT
35.4
18.5
1.5
110*
M
77.8
2
3
0
0
1
1
1
1
0
0
1.1
1.1
6.0
112*
F
77.8
2
3
1
0
0
1
1
1
0
CT
10.1
3.6
6.8
127
M
52.9
2
2
1
0
0
1
0
0
0
CT
37.7
34.8
6.4
128*
M
53.6
2
3
1
0
1
1
0
0
0
CT
11.2
10.2
6.8
136*
M
78.4
2
3
1
0
0
0
0
0
0
0
37.8
24.5
6.1
151°
M
45.9
2
3
1
0
0
1
1
1
0
CT
13.5
10.3
17.6
169
F
80.5
2
3
1
0
0
0
1
1
0
0
10.2
2.4
4.3
174**,°
M
66.9
2
3
0
0
0
1
1
1
0
CT
43.3
19.8
2.7
178**
F
53.2
3
4
1
0
0
1
0
1
0
CT
27.2
27.2
12.0
199
M
52.2
3
3
0
0
1
1
1
1
0
CT
23.5
4.9
5.0
207**,°
F
67.0
2
3
0
0
0
0
0
0
0
CT
7.2
5.5
10.0
218
F
62.6
3
3
1
0
0
1
1
1
0
0
0.3
0.3
1.4
223**,°
F
57.3
2
2
1
0
0
1
1
1
0
0
24.5
7.4
2.0
229**
F
73.2
3
3
1
0
0
1
1
1
0
0
4.5
4.0
5.4
235**,°
F
57.4
1
3
1
0
1
1
0
0
0
CT
19.6
7.7
2.1
241**,°
F
74.6
2
3
0
0
0
1
0
0
0
0
6.9
2.9
2.7
* used for microarray analysis; ** used to investigate gemcitabine resistance; ° used for sphere-formation assay.
Abbreviations: F: female; M: male; y: year; p: pathological; G: histopathological grade; T: tumour size; N: lymph node metastasis; M: metastasis; R: resection margin; PNI: perineural invasion; VI: vascular invasion; LVI: lymphovascular invasion; RCT: radiochemotherapy; CT: chemotherapy; OS: overall survival; DFS: disease-free survival; m: month.

SP analysis

Xenograft tumours (n = 17) were dissociated into single cells using collagenase type IV (1 mg/ml in Medium 199; Invitrogen, Grand Island, NY). Cells were incubated with Hoechst33342 (Sigma-Aldrich, Bornem, Belgium) at a final concentration of 5 μg/ml, and the SP was identified as a side branch of ‘Hoechst low’ cells using dual-wavelength FACS analysis (FACSVantage SE, equipped with FACS DIVA software, version 6.0; BD Biosciences, Erembodegem, Belgium; Hoechst red with 675/20 nm filter and Hoechst blue with 424/44 nm filter). Verapamil (100μM; Sigma-Aldrich) was added to verify the SP phenotype, as it results in the reduction of the side branch by blocking the multidrug transporters. Propidium Iodide (2μg/ml; Sigma-Aldrich) was used to exclude dead cells. For further characterization, tumour cells were immunostained for the endothelial marker CD31 and the hematopoietic marker CD45. After Hoechst incubation, fluorescein (FITC)-labeled anti-mouse or anti-human CD31 and phycoerythrin (PE)-labeled anti-mouse or anti-human CD45 antibodies (BD Biosciences), or PE-labeled anti-human CD133 (Miltenyi Biotec, Bergisch Gladbach, Germany) were added using dilutions according to the manufacturer’s recommendations. Sorted SP and MP cells were established as monolayers and subjected to Cyto-Rich Red staining (BD Biosciences).

Treatment of mice bearing xenograft tumours with gemcitabine

To investigate resistance of SP cells to gemcitabine, 7 different human PDAC samples were grown in SCID mice (see Table 1). When the tumour reached a volume of approximately 200 mm3, one group of mice received gemcitabine (Eli Lilly, Brussels, Belgium; 200 mg/kg body weight intraperitoneally, 1 injection every 3 days, 6 injections in total) and the other group (bearing the corresponding tumours) was injected with vehicle (0.9% NaCl; control group). Tumour diameter was measured every 3 days after the first injection. Three days after the last injection, mice were euthanized and tumours analyzed to determine the proportion of SP cells as described above. Gemcitabine was considered effective when tumour volume decreased at least 50%.

Whole-genome expression profiling

For RNA extraction, 25000 SP and 25000 MP cells were sorted by FACS into cold lysis solution (RNeasy Micro Kit; Qiagen, Venlo, The Netherlands). RNA was extracted according to the instructions of the manufacturer. RNA quality and concentration were determined using Picochips on a BioAnalyzer 2100 (Agilent Technologies, Santa Clara, CA). Only samples with RNA Integrity Number (RIN) ≥8.0 were used for gene expression profiling by microarray analysis. After Baugh amplification, Cy3 label was incorporated into the cRNA, which was then hybridized onto whole-genome human 44 K oligonucleotide arrays (G4112F, Agilent) [19, 20].
Expression values were obtained using the Agilent feature extraction software (version 10.1.1.1) and subjected to quantile normalization. Probes lacking a detection call signal (n = 1666) were omitted from further analysis. The log2-ratios for each SP-MP pair were compared with the Limma (Linear Models for Microarra Data) package of Bioconductor [21]. The contrast SP-MP was tested with a moderate t-statistic (implemented in Limma). The resulting p-values were corrected for multiple testing with Benjamini-Hochberg to control false discovery rate. The Database for Annotation, Visualization and Integrated Discovery (DAVID, version 6.7, http://​www.​david.​abcc.​ncifcrf.​org) was used to uncover enriched function-related gene groups by gene-annotation enrichment analysis and to reveal enriched KEGG (Kyoto Encyclopedia of Genes and Genomes) pathways. Significance was tested by the EASE score, a modified Fisher’s exact p-value test to adjust for multiple testing. Gene clusters with an enrichment score of >1.5 were retained. To further visualize gene networks, the Search Tool for the Retrieval of Interacting Genes (STRING, http://​string.​embl.​de) was used.
Gene expression data are available from the Gene Expression Omnibus (GEO, http://​www.​ncbi.​nlm.​nih.​gov/​projects/​geo/​) through series accession number GSE36563.

Tumourigenic (CSC) activity as analyzed by in vitrosphere formation

To investigate sphere-forming capacity, SP and MP subpopulations were sorted by FACS from PDAC xenograft tumours (n = 6; see Table 1), and 40000 cells of each seeded in DMEM/F12 (Invitrogen), supplemented with 0.4% BSA and containing basic fibroblast growth factor (bFGF; 10 ng/ml; R&D Systems, Minneapolis, MN), epidermal growth factor (EGF; 20 ng/ml; R&D Systems), insulin-transferrin-selenium (1:100; Invitrogen) and B27 (1:50; Invitrogen) [22]. The medium was renewed at day 3, and spheres were counted at day 7 to determine the sphere-forming capacity.

Statistical analysis

Statistical analysis was performed using the Visual data discovery software JMP9 (SAS, Cary, NC). A Wilcoxon Test for nonparametric variables was applied to determine the statistical level of difference between values of PDAC SP and MP and to compare the results from the mouse group treated with gemcitabine and the control group. Statistical significance was determined as p < 0.05, and as p < 0.001 for the microarray analysis.

Results

Presence of a SP in human PDAC xenografts

Human PDAC samples (n = 17; Table 1) were expanded by s.c. implantation, each in several SCID mice. All tumours grew in at least one mouse, regardless of the PDAC clinicopathological features or disease stage. Xenograft tumours showed histological resemblance to the original PDAC, although the stromal component was less abundant in the xenograft than in the original tumour (Figure 1A). In all tumours analyzed (n = 17), a SP was identified, ranging from 1.4 to 17.6% of total cells (median: 5.4%; Figure 1B-C; Table 1). Addition of verapamil during incubation with Hoechst decreased the SP proportion to 0.4% (range: 0.1-5.6%), thus confirming the SP phenotype (Figure 1B-C). The SP cells were smaller than the MP cells and had a larger nucleus/cytoplasm ratio, indicative of a poorly differentiated or undifferentiated nature [3] (Figure 1B). No significant correlation was found between the SP size and patients’ survival in this population of 17 PDAC (Table 1). Because it is known that the SP phenotype can co-purify endothelial and hematopoietic cells [20, 23], we further analyzed the xenografts for CD31+ and CD45+ cells. Human epitopes were not detected (data not shown) but mouse CD45+ and CD31+ cells were found, indicating that host cells were attracted and incorporated into the growing human xenograft tumours. Yet, the proportion of CD45+ and CD31+ cells in the SP was low (median: 4.1% and 4.5%, respectively; n = 10; Figure 1D).

The SP is resistant to gemcitabine as assessed in vivo

Seven different PDAC samples were grown in SCID mice (see Table 1). Treatment of mice with gemcitabine affected growth of 4 out of 7 xenograft tumours, resulting in an average volume reduction of 72% in comparison with an average tumour expansion in the vehicle-treated control group of 230% (p = 0.030) (Figure 2A-B). After treatment, tumours were excised and analyzed for SP. The tumour SP proportion was larger in mice treated with gemcitabine than in the corresponding controls (median 6.6% versus 2.7%, respectively; n = 7; p = 0.028; Figure 2B-C). SP enrichment was even higher when only considering the tumours that responded to gemcitabine with tumour shrinkage (of at least 50%) (median: 4.7% versus 1.3%, respectively; n = 4; Figure 2B). CD45+ and CD31+ cells did not rise in the SP in response to gemcitabine (p = 0.21 and p = 0.66, respectively) when compared to control mice.

Whole-genome expression analysis of the PDAC xenograft SP reveals upregulation of genes related to therapy resistance

To characterize the SP at the gene expression level, whole-genome expression analysis was performed on SP and MP cells sorted from 5 different xenograft tumours (see Table 1). Microarrays with human genome oligonucleotide probes were used, thereby excluding the detection of mouse transcripts (such as from the infiltrating CD31+ and CD45+ SP cells). Comparison revealed that 145 probe sets, representing 121 genes, were differentially expressed between the SP and MP (p < 0.001); 80 genes were upregulated in the SP and 41 genes downregulated (complete list in Additional file 1: Table S1; extract of genes in Table 2, selected on the basis of relevance from the literature and from DAVID analysis as below).
Table 2
Selection of genes differentially expressed in the PDAC xenograft SP versus MP
Gene Symbol
Genbank Accession no.
Gene Name
Fold SP/MP
p-value
ZAP70
NM_001079
zeta-chain (TCR) associated protein kinase 70 kDa
24.25
0.00002
PRKCQ
NM_006257
protein kinase C, theta
21.56
0.00008
FASLG
NM_000639
Fas ligand (TNF superfamily, member 6)
19.56
0.00003
LEF1
NM_000639
lymphoid enhancer-binding factor 1
16.45
0.00009
PACSIN1
NM_020804
protein kinase C and casein kinase substrate in neurons 1
12.47
0.00015
MEOX2
NM_005924
mesenchyme homeobox 2
8.22
0.00060
STAT4
NM_003151
signal transducer and activator of transcription 4
7.46
0.00050
EMX2
NM_003151
empty spiracles homeobox 2
6.87
0.00006
SOX11
NM_003108
SRY (sex determining region Y)-box 11
6.19
0.00029
FGF7
NM_002009
fibroblast growth factor 7 (keratinocyte growth factor)
5.74
0.00072
FOXG1B
NM_005249
forkhead box G1B
5.62
0.00019
PKNOX2
NM_022062
PBX/knotted 1 homeobox 2
5.58
0.00017
ITGB3
NM_000212
integrin, beta 3 (platelet glycoprotein IIIa, antigen CD61)
5.35
0.00062
KLF12
NM_007249
Kruppel-like factor 12
5.03
0.00048
SNAI2
NM_003068
snail homolog 2 (Drosophila)
4.99
0.00079
TIE1
NM_005424
tyrosine kinase with immunoglobulin-like and EGF-like domains 1
4.76
0.00019
IRX2
AY335940
iroquois homeobox protein 2
4.53
0.00010
EFNB2
NM_004093
ephrin-B2
4.23
0.00016
ETS1
NM_005238
v-ets erythroblastosis virus E26 oncogene homolog 1 (avian)
3.95
0.00095
BCL2L11
NM_138621
BCL2-like 11
3.68
0.00019
GRB10
NM_001001555
growth factor receptor-bound protein 10
3.68
0.00034
NFIB
NM_005596
nuclear factor I/B
3.46
0.00010
INHBB
NM_002193
inhibin, beta B (activin AB beta polypeptide)
3.32
0.00059
TCF7L1
NM_031283
transcription factor 7-like 1 (T-cell specific, HMG-box)
3.14
0.00073
EPC2
NM_053001
enhancer of polycomb homolog 2 (Drosophila)
2.95
0.00090
GATA1
NM_002049
GATA binding protein 1 (globin transcription factor 1)
2.57
0.00082
KITLG
NM_000899
KIT ligand
2.50
0.00052
ERRFI1
NM_018948
ERBB receptor feedback inhibitor 1
2.45
0.00074
CADM1
NM_014333
cell adhesion molecule 1
−22.84
0.00077
TREM2
NM_018965
triggering receptor expressed on myeloid cells 2
−9.90
0.00017
ALOX5AP
NM_001629
arachidonate 5-lipoxygenase-activating protein
−9.32
0.00017
PLA2G7
NM_005084
phospholipase A2, group VII (platelet-activating factor acetylhydrolase, plasma)
−7.66
0.00007
CD74
NM_004355
CD74 molecule, major histocompatibility complex, class II invariant chain
−7.28
0.00006
MADCAM1
NM_130760
mucosal vascular addressin cell adhesion molecule 1
−6.53
0.00004
CD14
NM_130760
CD14 molecule
−5.94
0.00038
CTSA
NM_000308
cathepsin A
−3.29
0.00068
CTSC
NM_001814
cathepsin C
−2.97
0.00085
Gene-clustering analysis of all differentially expressed genes (p < 0.001) by DAVID showed 3 functionally related groups of genes enriched in the SP: one group of transcription factors, one of adhesion molecules and one of homeobox genes (Table 3). KEGG pathway analysis revealed 3 significantly upregulated pathways in the SP versus the MP, including ‘cancer’ and ‘adherens junctions’ (Table 3). Two KEGG pathways were significantly downregulated in the SP, including ‘cell adhesion molecules’. Visualization of the interaction network of SP-upregulated genes by STRING analysis (Figure 3) reveals that genes involved in chemoresistance [ETS1, KIT ligand ( KITLG) or stem cell factor ( SCF), SNAI2], regulation of apoptosis ( FASLG, GRB10, BCL2L11, ETS1, SNAI2), epithelial-mesenchymal transition (EMT) ( SNAI2, LEF1) and tumourigenesis (oncogenes like FGF7, GATA1, KITLG, ETS1) occupy a central position. Moreover, multidrug transporters, linked to chemoresistance and some considered responsible for the SP phenotype, also show a clear tendency of upregulation in the SP ( ABCG2, 3.63 fold, p = 0.006; ABCA9, 3.66-fold, p = 0.003).
Table 3
Gene-function analysis of all differentially expressed genes between PDAC xenograft SP and MP using DAVID
A. ENRICHED GENE CLUSTERS
Gene group 1: Transcription Factors Enrichment Score: 1.78
1
TCF7L1
transcription factor 7-like 1 (T-cell specific, HMG-box)
2
SNAI2
snail homolog 2 (Drosophila)
3
CNOT4
CCR4-NOT transcription complex, subunit 4
4
ZBTB10
zinc finger and BTB domain containing 10
5
KLF12
Kruppel-like factor 12
6
ZFPM2
zinc finger protein, multitype 2
7
STAT4
signal transducer and activator of transcription 4
8
OSR2
odd-skipped related 2 (Drosophila)
9
IKZF2
IKAROS family zinc finger 2 (Helios)
10
PHF6
PHD finger protein 6
11
ZFHX4
zinc finger homeobox 4
12
SOX11
SRY (sex determining region Y)-box 11
13
CITED4
Cbp/p300-interacting transactivator, with Glu/Asp-rich carboxy-terminal domain, 4
14
ARNTL
aryl hydrocarbon receptor nuclear translocator-like
15
NFIB
nuclear factor I/B
16
GATA1
GATA binding protein 1 (globin transcription factor 1)
17
ETS
v-ets erythroblastosis virus E26 oncogene homolog 1 (avian)
18
LEF1
lymphoid enhancer-binding factor 1
Gene group 2: Adhesion Molecules Enrichment Score: 1.71
1
HLA-DPB1
major histocompatibility complex, class II, DP beta 1
2
MADCAM1
mucosal vascular addressin cell adhesion molecule 1
3
TREM2
triggering receptor expressed on myeloid cells 2
4
EFNB2
ephrin-B2
5
IGSF6
immunoglobulin superfamily, member 6
6
LRFN5
leucine rich repeat and fibronectin type III domain containing 5
7
DSCAM
Down syndrome cell adhesion molecule
8
JAM2
junctional adhesion molecule 2
9
MDGA1
MAM domain containing glycosylphosphatidylinositol anchor 1
Gene group 3: Homeobox Genes Enrichment Score: 1.51
1
PKNOX2
PBX/knotted 1 homeobox 2
2
MEOX2
mesenchyme homeobox 2
3
SPIC
Spi-C transcription factor (Spi-1/PU.1 related)
4
IRX2
iroquois homeobox 2
5
EMX2
empty spiracles homeobox 2
B. KEGG PATHWAYS
Upregulated in SP
 
Number of genes
genes
Arrhythmogenic rightventricular cardiomyopathy
4
GJA1, ITGB3, LEF1, TCFL1
Pathways in cancer
6
TCF7L1, KITL, FASL, LEF1, FGF7, ETS1
Adherens junction
3
LEF1, SNAI2, TCF7L1
Downregulated in SP
 
Number of genes
genes
Lysosome
3
CTSA, CTSC, SLC11A1
Cell adhesion molecules (CAMs)
3
CADM1, HLA-DPB1, MADCAM1

The SP is enriched in sphere-forming cells

Tumourigenic (CSC) activity was analyzed in vitro using the sphere-forming assay [22]. SP and MP from xenograft tumours were first depleted from the (murine) endothelial and immune cells by FACS and then seeded in defined culture conditions (see Methods). Viability of the sorted SP and MP cells was identical (data not shown). The CD45-/CD31- SP generated spheres in all experiments (n = 6; median number of spheres: 16; range: 10–35; Figure 4). In contrast, the CD45-/CD31- MP did not consistently generate spheres (not in 2 out of the 6 experiments) and the spheres obtained were lower in number (range: 0–15; median: 8; p = 0.016 versus SP), less well-formed, and smaller in size (Figure 4). These findings indicate that SP cells have a higher sphere-forming capacity than MP cells. It was not possible to assess the propagation (self-renewal) capacity of the sphere-forming cells because the number of spheres obtained was too low and the dispersion did not yield enough (viable) cells.

Discussion

To date, SP analysis in pancreatic cancer has been limited to cultured cell lines [68, 24, 25]. In the present study, we demonstrate for the first time the presence of a SP in human PDAC samples using first-generation xenografts, histologically resemblant to the original tumour samples except for the stromal component. We showed, using a xenograft in vivo model, that the SP is more resistant to gemcitabine than the other tumour cells, and therefore may represent a potential therapeutic target. The response rate of the xenograft tumours to gemcitabine (4 out of 7) was higher than the known response rate in human patients (lower than 20%), which may be due to the reduced stromal component in the xenografts, thus improving drug delivery to the tumour cells (see [3, 4]).
Whole-genome expression profiling of the SP demonstrated the expression of genes involved in cancer pathways, particularly chemoresistance and EMT. In many cancers, the proto-oncogene ETS1 plays a role in chemoresistance and invasion [26]. In PDAC cell lines, ETS1 expression has been linked to gemcitabine resistance and to invasiveness by induction of matrix metalloproteinase (MMP) 2 [27]. We also found upregulation of MMP2 in the PDAC SP, although at the border of statistical significance (3.12 fold; p = 0.003). Equally, KITLG ( SCF) increases invasive capacity in PDAC cell lines, as well as cell proliferation [28, 29]. The zinc finger transcription factor SNAI2 ( SLUG) is involved in chemo- and radioresistance (e.g. through anti-apoptotic mechanisms) [30]. Moreover, SNAI2 is a core regulator of EMT, a key process in cancer pathogenesis and tumour progression by which epithelial cells acquire a mesenchymal phenotype with invasive and migratory properties. EMT is considered to play an important role in tumour resistance and metastasis. The SP of the pancreatic cell line PANC-1 has been shown to possess superior potential for EMT when compared to the MP [24]. Recently, SNAI2 was shown to promote EMT, invasion and metastasis in a pancreatic cancer cell line [31, 32]. EMT involves the downregulation of cell adhesion molecules like E-cadherin. In the present study, E-cadherin was significantly downregulated in 3 of the 5 PDAC samples analyzed (data not shown). SNAI2 can act as a transcriptional repressor of E-cadherin. However, in pancreatic cancer cell lines, SNAI2 was found only a weak suppressor of E-cadherin [32], whereas in human PDAC, no significant correlation could be observed between SNAI2 and E-cadherin [32, 33]. LEF1 (lymphoid enhancer factor 1), a nuclear transducer of the Wnt signaling pathway, also plays a key role in EMT. In addition, LEF1 expression in human pancreatic cancer correlates with advanced tumour stages [34]. TCF7L1 is known to form a complex with LEF1 to achieve DNA-binding ability. Finally, other signaling proteins of the Wnt pathway (Wnt7b, DVL1, FZD1, FZD4, FZD5) as well as components of the TGFβ/BMP pathway ( TGFβ1, BMP1, BMPP2Ra, SMURF2) were upregulated in the SP versus MP, although not reaching statistical significance (0.001 < p < 0.05; data not shown).
Protection against apoptosis represents a further mechanism of therapy resistance. In our current study, BCL2L11 and FASLG are upregulated in the SP. In studies with pancreatic cancer cell lines, BCL2L11 has been correlated with apoptotic resistance as well as with metastatic potential [35, 36]. FASLG may regulate immune evasion of tumour cells by inducing apoptosis in cytotoxic T lymphocytes. FASLG has been reported to play a role in the aggressiveness of PDAC, potentially through this immune escape mechanism [37].
Finally, the multidrug transporter ABCG2 is highly expressed in the SP, suggesting a role in chemoresistance and at the same time supporting the SP phenotype which is, at least partially, linked to activity of this pump. In pancreatic cancer cell lines, ABCG2 was also found to be upregulated in the SP [6, 8] and associated with chemotherapy resistance [38]. In addition, ABCA9, another membrane transporter linked to chemoresistance in some cancers like malignant melanoma [39], was found upregulated in the PDAC SP in our study.
In multiple types of cancer, the SP displays properties reminiscent of CSC. In the present study, we show that the SP of PDAC is enriched in cells that generate spheres. Although sphere formation is regarded as an in-vitro assay for CSC (tumourigenic) activity (see e.g. [22]), it should be noted that this link is not always present as reported, for instance, in high-grade glioma [40]. In order to demonstrate tumourigenic (CSC) activity of the PDAC SP, further study is needed that analyzes in-vivo tumour growth after implantation of the (purified) population in immunodeficient mice. Given the essential interactions of cancer cells with their microenvironment for tumour development, transplantation within the pancreas would be most appropriate but technically highly demanding. Yet, some expression characteristics may already suggest a CSC(−like) phenotype including expression of genes associated with chemoresistance (see above), with the Wnt pathway and with EMT. Indeed, the latter process has recently been uncovered as a key promoter of the generation and activity of CSC [41]. Moreover, KITLG (SCF) has been linked to CSC in prostate [42] and lung cancer [43]. In the present study, expression of the previously proposed CSC membrane markers in pancreatic cancer (i.e. CD24+CD44+ESA+[16, 44] and CD133+[17]) was not found upregulated in the PDAC xenograft SP, neither in microarray analysis, nor in flow-cytometric examination (1.0% CD133+ cells in the SP and 0.7% in the MP; n = 2) (data not shown). Noteworthy, expression of the two previously defined sets of markers (CD24+CD44+ESA+ and CD133+) did also not completely, or only minimally, overlap (10-40%) [16]. Thus, CSC in pancreatic cancer, and the link with the SP, need further investigation.

Conclusions

Our study revealed the presence of a SP in human PDAC, displaying chemoresistance and CSC-associated activity, as well as expression of genes involved in both processes. SP cells thus may represent interesting targets for new and more efficient therapeutic strategies. Chemoresistance, anti-apoptosis and EMT genes identified may guide us to potential molecular targets.

Acknowledgements

AVDB and LG acknowledge support by PhD Fellow grants from the Fund for Scientific Research – Flanders (FWO-Vlaanderen) and BT acknowledges support by a research grant of the FWO.
The authors are very grateful to Vik Van Duppen (Hematology, KU Leuven) for his help with FACS analysis and sorting, and to the VIB-MAF (KU Leuven) for the microarray analysis.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests

Authors’ contributions

AVDB designed and performed the study, analyzed the data and wrote the manuscript. LG trained AVDB in the techniques used and helped in data collection. BT participated in study design and analysis and in writing. HV participated in the design and analysis of the study and wrote the manuscript. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Jemal A, Siegel R, Ward E, Hao Y, Xu J, Murray T, Thun MJ: Cancer statistics, 2008. CA Cancer J Clin. 2008, 58 (2): 71-96. 10.3322/CA.2007.0010.CrossRefPubMed Jemal A, Siegel R, Ward E, Hao Y, Xu J, Murray T, Thun MJ: Cancer statistics, 2008. CA Cancer J Clin. 2008, 58 (2): 71-96. 10.3322/CA.2007.0010.CrossRefPubMed
2.
Zurück zum Zitat Van den Broeck A, Sergeant G, Ectors N, Van Steenbergen W, Aerts R, Topal B: Patterns of recurrence after curative resection of pancreatic ductal adenocarcinoma. Eur J Surg Oncol. 2009, 35 (6): 600-604. 10.1016/j.ejso.2008.12.006.CrossRefPubMed Van den Broeck A, Sergeant G, Ectors N, Van Steenbergen W, Aerts R, Topal B: Patterns of recurrence after curative resection of pancreatic ductal adenocarcinoma. Eur J Surg Oncol. 2009, 35 (6): 600-604. 10.1016/j.ejso.2008.12.006.CrossRefPubMed
3.
Zurück zum Zitat Olive KP, Jacobetz MA, Davidson CJ, Gopinathan A, McIntyre D, Honess D, Madhu B, Goldgraben MA, Caldwell ME, Allard D, Frese KK, Denicola G, Feig C, Combs C, Winter SP, Ireland-Zecchini H, Reichelt S, Howat WJ, Chang A, Dhara M, Wang L, Rückert F, Grützmann R, Pilarsky C, Izeradjene K, Hingorani SR, Huang P, Davies SE, Plunkett W, Egorin M, et al: Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science. 2009, 324 (5933): 1457-1461. 10.1126/science.1171362.CrossRefPubMedPubMedCentral Olive KP, Jacobetz MA, Davidson CJ, Gopinathan A, McIntyre D, Honess D, Madhu B, Goldgraben MA, Caldwell ME, Allard D, Frese KK, Denicola G, Feig C, Combs C, Winter SP, Ireland-Zecchini H, Reichelt S, Howat WJ, Chang A, Dhara M, Wang L, Rückert F, Grützmann R, Pilarsky C, Izeradjene K, Hingorani SR, Huang P, Davies SE, Plunkett W, Egorin M, et al: Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science. 2009, 324 (5933): 1457-1461. 10.1126/science.1171362.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Olson P, Hanahan D: Cancer. Breaching the cancer fortress. Science. 2009, 324 (5933): 1400-1401. 10.1126/science.1175940.CrossRefPubMed Olson P, Hanahan D: Cancer. Breaching the cancer fortress. Science. 2009, 324 (5933): 1400-1401. 10.1126/science.1175940.CrossRefPubMed
5.
Zurück zum Zitat Goodell MA: Stem cell identification and sorting using the Hoechst 33342 side population (SP). Curr Protoc Cytom. 2005, suppl 9: 18.1-18.11. Goodell MA: Stem cell identification and sorting using the Hoechst 33342 side population (SP). Curr Protoc Cytom. 2005, suppl 9: 18.1-18.11.
6.
Zurück zum Zitat Yao J, Cai HH, Wei JS, An Y, Ji ZL, Lu ZP, Wu JL, Chen P, Jiang KR, Dai CC, Qian ZY, Xu ZK, Miao Y: Side population in the pancreatic cancer cell lines SW1990 and CFPAC-1 is enriched with cancer stem-like cells. Oncol Rep. 2010, 23 (5): 1375-1382.PubMed Yao J, Cai HH, Wei JS, An Y, Ji ZL, Lu ZP, Wu JL, Chen P, Jiang KR, Dai CC, Qian ZY, Xu ZK, Miao Y: Side population in the pancreatic cancer cell lines SW1990 and CFPAC-1 is enriched with cancer stem-like cells. Oncol Rep. 2010, 23 (5): 1375-1382.PubMed
7.
Zurück zum Zitat Zhang SN, Huang FT, Huang YJ, Zhong W, Yu Z: Characterization of a cancer stem cell-like side population derived from human pancreatic adenocarcinoma cells. Tumori. 2010, 96 (6): 985-992.PubMed Zhang SN, Huang FT, Huang YJ, Zhong W, Yu Z: Characterization of a cancer stem cell-like side population derived from human pancreatic adenocarcinoma cells. Tumori. 2010, 96 (6): 985-992.PubMed
8.
Zurück zum Zitat Zhou J, Wang CY, Liu T, Wu B, Zhou F, Xiong JX, Wu HS, Tao J, Zhao G, Yang M, Gou SM: Persistence of side population cells with high drug efflux capacity in pancreatic cancer. World J Gastroenterol. 2008, 14 (6): 925-930. 10.3748/wjg.14.925.CrossRefPubMedPubMedCentral Zhou J, Wang CY, Liu T, Wu B, Zhou F, Xiong JX, Wu HS, Tao J, Zhao G, Yang M, Gou SM: Persistence of side population cells with high drug efflux capacity in pancreatic cancer. World J Gastroenterol. 2008, 14 (6): 925-930. 10.3748/wjg.14.925.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Haraguchi N, Utsunomiya T, Inoue H, Tanaka F, Mimori K, Barnard GF, Mori M: Characterization of a side population of cancer cells from human gastrointestinal system. Stem Cells. 2006, 24 (3): 506-513. 10.1634/stemcells.2005-0282.CrossRefPubMed Haraguchi N, Utsunomiya T, Inoue H, Tanaka F, Mimori K, Barnard GF, Mori M: Characterization of a side population of cancer cells from human gastrointestinal system. Stem Cells. 2006, 24 (3): 506-513. 10.1634/stemcells.2005-0282.CrossRefPubMed
10.
Zurück zum Zitat Wu C, Alman BA: Side population cells in human cancers. Cancer Lett. 2008, 268 (1): 1-9. 10.1016/j.canlet.2008.03.048.CrossRefPubMed Wu C, Alman BA: Side population cells in human cancers. Cancer Lett. 2008, 268 (1): 1-9. 10.1016/j.canlet.2008.03.048.CrossRefPubMed
11.
Zurück zum Zitat Clarke MF, Dick JE, Dirks PB, Eaves CJ, Jamieson CH, Jones DL, Visvader J, Weissman IL, Wahl GM: Cancer stem cells–perspectives on current status and future directions: AACR Workshop on cancer stem cells. Cancer Res. 2006, 66 (19): 9339-9344. 10.1158/0008-5472.CAN-06-3126.CrossRefPubMed Clarke MF, Dick JE, Dirks PB, Eaves CJ, Jamieson CH, Jones DL, Visvader J, Weissman IL, Wahl GM: Cancer stem cells–perspectives on current status and future directions: AACR Workshop on cancer stem cells. Cancer Res. 2006, 66 (19): 9339-9344. 10.1158/0008-5472.CAN-06-3126.CrossRefPubMed
12.
Zurück zum Zitat Sergeant G, Vankelecom H, Gremeaux L, Topal B: Role of cancer stem cells in pancreatic ductal adenocarcinoma. Nat Rev Clin Oncol. 2009, 6 (10): 580-586. 10.1038/nrclinonc.2009.127.CrossRefPubMed Sergeant G, Vankelecom H, Gremeaux L, Topal B: Role of cancer stem cells in pancreatic ductal adenocarcinoma. Nat Rev Clin Oncol. 2009, 6 (10): 580-586. 10.1038/nrclinonc.2009.127.CrossRefPubMed
13.
Zurück zum Zitat Boman BM, Wicha MS: Cancer stem cells: a step toward the cure. J Clin Oncol. 2008, 26 (17): 2795-2799. 10.1200/JCO.2008.17.7436.CrossRefPubMed Boman BM, Wicha MS: Cancer stem cells: a step toward the cure. J Clin Oncol. 2008, 26 (17): 2795-2799. 10.1200/JCO.2008.17.7436.CrossRefPubMed
14.
Zurück zum Zitat Eaves CJ: Cancer stem cells: Here, there, everywhere?. Nature. 2008, 456 (7222): 581-582. 10.1038/456581a.CrossRefPubMed Eaves CJ: Cancer stem cells: Here, there, everywhere?. Nature. 2008, 456 (7222): 581-582. 10.1038/456581a.CrossRefPubMed
15.
Zurück zum Zitat Eppert K, Takenaka K, Lechman ER, Waldron L, Nilsson B, van Galen P, Metzeler KH, Poeppl A, Ling V, Beyene J, Canty AJ, Danska JS, Bohlander SK, Buske C, Minden MD, Golub TR, Jurisica I, Ebert BL, Dick JE: Stem cell gene expression programs influence clinical outcome in human leukemia. Nat Med. 2011, 17 (9): 1086-1093. 10.1038/nm.2415.CrossRefPubMed Eppert K, Takenaka K, Lechman ER, Waldron L, Nilsson B, van Galen P, Metzeler KH, Poeppl A, Ling V, Beyene J, Canty AJ, Danska JS, Bohlander SK, Buske C, Minden MD, Golub TR, Jurisica I, Ebert BL, Dick JE: Stem cell gene expression programs influence clinical outcome in human leukemia. Nat Med. 2011, 17 (9): 1086-1093. 10.1038/nm.2415.CrossRefPubMed
16.
Zurück zum Zitat Li C, Heidt DG, Dalerba P, Burant CF, Zhang L, Adsay V, Wicha M, Clarke MF, Simeone DM: Identification of pancreatic cancer stem cells. Cancer Res. 2007, 67 (3): 1030-1037. 10.1158/0008-5472.CAN-06-2030.CrossRefPubMed Li C, Heidt DG, Dalerba P, Burant CF, Zhang L, Adsay V, Wicha M, Clarke MF, Simeone DM: Identification of pancreatic cancer stem cells. Cancer Res. 2007, 67 (3): 1030-1037. 10.1158/0008-5472.CAN-06-2030.CrossRefPubMed
17.
Zurück zum Zitat Hermann PC, Huber SL, Herrler T, Aicher A, Ellwart JW, Guba M, Bruns CJ, Heeschen C: Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell Stem Cell. 2007, 1 (3): 313-323. 10.1016/j.stem.2007.06.002.CrossRefPubMed Hermann PC, Huber SL, Herrler T, Aicher A, Ellwart JW, Guba M, Bruns CJ, Heeschen C: Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell Stem Cell. 2007, 1 (3): 313-323. 10.1016/j.stem.2007.06.002.CrossRefPubMed
18.
Zurück zum Zitat Tomayko MM, Reynolds CP: Determination of subcutaneous tumor size in athymic (nude) mice. Cancer Chemother Pharmacol. 1989, 24 (3): 148-154. 10.1007/BF00300234.CrossRefPubMed Tomayko MM, Reynolds CP: Determination of subcutaneous tumor size in athymic (nude) mice. Cancer Chemother Pharmacol. 1989, 24 (3): 148-154. 10.1007/BF00300234.CrossRefPubMed
19.
Zurück zum Zitat Baugh LR, Hill AA, Brown EL, Hunter CP: Quantitative analysis of mRNA amplification by in vitro transcription. Nucleic Acids Res. 2001, 29 (5): E29-10.1093/nar/29.5.e29.CrossRefPubMedPubMedCentral Baugh LR, Hill AA, Brown EL, Hunter CP: Quantitative analysis of mRNA amplification by in vitro transcription. Nucleic Acids Res. 2001, 29 (5): E29-10.1093/nar/29.5.e29.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Chen J, Gremeaux L, Fu Q, Liekens D, Van Laere S, Vankelecom H: Pituitary progenitor cells tracked down by side population dissection. Stem Cells. 2009, 27 (5): 1182-1195. 10.1002/stem.51.CrossRefPubMed Chen J, Gremeaux L, Fu Q, Liekens D, Van Laere S, Vankelecom H: Pituitary progenitor cells tracked down by side population dissection. Stem Cells. 2009, 27 (5): 1182-1195. 10.1002/stem.51.CrossRefPubMed
21.
Zurück zum Zitat Smyth GK: Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol. 2004, 3 (1): 1544-6115. Smyth GK: Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol. 2004, 3 (1): 1544-6115.
22.
Zurück zum Zitat Gou S, Liu T, Wang C, Yin T, Li K, Yang M, Zhou J: Establishment of clonal colony-forming assay for propagation of pancreatic cancer cells with stem cell properties. Pancreas. 2007, 34 (4): 429-435. 10.1097/MPA.0b013e318033f9f4.CrossRefPubMed Gou S, Liu T, Wang C, Yin T, Li K, Yang M, Zhou J: Establishment of clonal colony-forming assay for propagation of pancreatic cancer cells with stem cell properties. Pancreas. 2007, 34 (4): 429-435. 10.1097/MPA.0b013e318033f9f4.CrossRefPubMed
23.
Zurück zum Zitat Bleau AM, Hambardzumyan D, Ozawa T, Fomchenko EI, Huse JT, Brennan CW, Holland EC: PTEN/PI3K/Akt pathway regulates the side population phenotype and ABCG2 activity in glioma tumor stem-like cells. Cell Stem Cell. 2009, 4 (3): 226-235. 10.1016/j.stem.2009.01.007.CrossRefPubMedPubMedCentral Bleau AM, Hambardzumyan D, Ozawa T, Fomchenko EI, Huse JT, Brennan CW, Holland EC: PTEN/PI3K/Akt pathway regulates the side population phenotype and ABCG2 activity in glioma tumor stem-like cells. Cell Stem Cell. 2009, 4 (3): 226-235. 10.1016/j.stem.2009.01.007.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Kabashima A, Higuchi H, Takaishi H, Matsuzaki Y, Suzuki S, Izumiya M, Iizuka H, Sakai G, Hozawa S, Azuma T, Hibi T: Side population of pancreatic cancer cells predominates in TGF-beta-mediated epithelial to mesenchymal transition and invasion. Int J Cancer. 2009, 124 (12): 2771-2779. 10.1002/ijc.24349.CrossRefPubMed Kabashima A, Higuchi H, Takaishi H, Matsuzaki Y, Suzuki S, Izumiya M, Iizuka H, Sakai G, Hozawa S, Azuma T, Hibi T: Side population of pancreatic cancer cells predominates in TGF-beta-mediated epithelial to mesenchymal transition and invasion. Int J Cancer. 2009, 124 (12): 2771-2779. 10.1002/ijc.24349.CrossRefPubMed
25.
Zurück zum Zitat Wang YH, Li F, Luo B, Wang XH, Sun HC, Liu S, Cui YQ, Xu XX: A side population of cells from a human pancreatic carcinoma cell line harbors cancer stem cell characteristics. Neoplasma. 2009, 56 (5): 371-378. 10.4149/neo_2009_05_371.CrossRefPubMed Wang YH, Li F, Luo B, Wang XH, Sun HC, Liu S, Cui YQ, Xu XX: A side population of cells from a human pancreatic carcinoma cell line harbors cancer stem cell characteristics. Neoplasma. 2009, 56 (5): 371-378. 10.4149/neo_2009_05_371.CrossRefPubMed
27.
Zurück zum Zitat Khanna A, Mahalingam K, Chakrabarti D, Periyasamy G: Ets-1 expression and gemcitabine chemoresistance in pancreatic cancer cells. Cell Mol Biol Lett. 2011, 16 (1): 101-113. 10.2478/s11658-010-0043-z.CrossRefPubMed Khanna A, Mahalingam K, Chakrabarti D, Periyasamy G: Ets-1 expression and gemcitabine chemoresistance in pancreatic cancer cells. Cell Mol Biol Lett. 2011, 16 (1): 101-113. 10.2478/s11658-010-0043-z.CrossRefPubMed
28.
Zurück zum Zitat Zhang M, Ma Q, Hu H, Zhang D, Li J, Ma G, Bhat K, Wu E: Stem cell factor/c-kit signaling enhances invasion of pancreatic cancer cells via HIF-1alpha under normoxic condition. Cancer Lett. 2011, 303 (2): 108-117. 10.1016/j.canlet.2011.01.017.CrossRefPubMed Zhang M, Ma Q, Hu H, Zhang D, Li J, Ma G, Bhat K, Wu E: Stem cell factor/c-kit signaling enhances invasion of pancreatic cancer cells via HIF-1alpha under normoxic condition. Cancer Lett. 2011, 303 (2): 108-117. 10.1016/j.canlet.2011.01.017.CrossRefPubMed
29.
Zurück zum Zitat Yasuda A, Sawai H, Takahashi H, Ochi N, Matsuo Y, Funahashi H, Sato M, Okada Y, Takeyama H, Manabe T: The stem cell factor/c-kit receptor pathway enhances proliferation and invasion of pancreatic cancer cells. Mol Cancer. 2006, 5: 46-10.1186/1476-4598-5-46.CrossRefPubMedPubMedCentral Yasuda A, Sawai H, Takahashi H, Ochi N, Matsuo Y, Funahashi H, Sato M, Okada Y, Takeyama H, Manabe T: The stem cell factor/c-kit receptor pathway enhances proliferation and invasion of pancreatic cancer cells. Mol Cancer. 2006, 5: 46-10.1186/1476-4598-5-46.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Cobaleda C, Perez-Caro M, Vicente-Duenas C, Sanchez-Garcia I: Function of the zinc-finger transcription factor SNAI2 in cancer and development. Annu Rev Genet. 2007, 41: 41-61. 10.1146/annurev.genet.41.110306.130146.CrossRefPubMed Cobaleda C, Perez-Caro M, Vicente-Duenas C, Sanchez-Garcia I: Function of the zinc-finger transcription factor SNAI2 in cancer and development. Annu Rev Genet. 2007, 41: 41-61. 10.1146/annurev.genet.41.110306.130146.CrossRefPubMed
31.
Zurück zum Zitat Nishioka R, Itoh S, Gui T, Gai Z, Oikawa K, Kawai M, Tani M, Yamaue H, Muragaki Y: SNAIL induces epithelial-to-mesenchymal transition in a human pancreatic cancer cell line (BxPC3) and promotes distant metastasis and invasiveness in vivo. Exp Mol Pathol. 2010, 89 (2): 149-157. 10.1016/j.yexmp.2010.05.008.CrossRefPubMed Nishioka R, Itoh S, Gui T, Gai Z, Oikawa K, Kawai M, Tani M, Yamaue H, Muragaki Y: SNAIL induces epithelial-to-mesenchymal transition in a human pancreatic cancer cell line (BxPC3) and promotes distant metastasis and invasiveness in vivo. Exp Mol Pathol. 2010, 89 (2): 149-157. 10.1016/j.yexmp.2010.05.008.CrossRefPubMed
32.
Zurück zum Zitat Hotz B, Arndt M, Dullat S, Bhargava S, Buhr HJ, Hotz HG: Epithelial to mesenchymal transition: expression of the regulators snail, slug, and twist in pancreatic cancer. Clin Cancer Res. 2007, 13 (16): 4769-4776. 10.1158/1078-0432.CCR-06-2926.CrossRefPubMed Hotz B, Arndt M, Dullat S, Bhargava S, Buhr HJ, Hotz HG: Epithelial to mesenchymal transition: expression of the regulators snail, slug, and twist in pancreatic cancer. Clin Cancer Res. 2007, 13 (16): 4769-4776. 10.1158/1078-0432.CCR-06-2926.CrossRefPubMed
33.
Zurück zum Zitat Zhang K, Chen D, Jiao X, Zhang S, Liu X, Cao J, Wu L, Wang D: Slug enhances invasion ability of pancreatic cancer cells through upregulation of matrix metalloproteinase-9 and actin cytoskeleton remodeling. Lab Invest. 2011, 91 (3): 426-438. 10.1038/labinvest.2010.201.CrossRefPubMedPubMedCentral Zhang K, Chen D, Jiao X, Zhang S, Liu X, Cao J, Wu L, Wang D: Slug enhances invasion ability of pancreatic cancer cells through upregulation of matrix metalloproteinase-9 and actin cytoskeleton remodeling. Lab Invest. 2011, 91 (3): 426-438. 10.1038/labinvest.2010.201.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Jesse S, Koenig A, Ellenrieder V, Menke A: Lef-1 isoforms regulate different target genes and reduce cellular adhesion. Int J Cancer. 2010, 126 (5): 1109-1120.PubMed Jesse S, Koenig A, Ellenrieder V, Menke A: Lef-1 isoforms regulate different target genes and reduce cellular adhesion. Int J Cancer. 2010, 126 (5): 1109-1120.PubMed
35.
Zurück zum Zitat Ruckert F, Samm N, Lehner AK, Saeger HD, Grutzmann R, Pilarsky C: Simultaneous gene silencing of Bcl-2, XIAP and Survivin re-sensitizes pancreatic cancer cells towards apoptosis. BMC Cancer. 2010, 10: 379-10.1186/1471-2407-10-379.CrossRefPubMedPubMedCentral Ruckert F, Samm N, Lehner AK, Saeger HD, Grutzmann R, Pilarsky C: Simultaneous gene silencing of Bcl-2, XIAP and Survivin re-sensitizes pancreatic cancer cells towards apoptosis. BMC Cancer. 2010, 10: 379-10.1186/1471-2407-10-379.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Bold RJ, Virudachalam S, McConkey DJ: BCL2 expression correlates with metastatic potential in pancreatic cancer cell lines. Cancer. 2001, 92 (5): 1122-1129. 10.1002/1097-0142(20010901)92:5<1122::AID-CNCR1429>3.0.CO;2-H.CrossRefPubMed Bold RJ, Virudachalam S, McConkey DJ: BCL2 expression correlates with metastatic potential in pancreatic cancer cell lines. Cancer. 2001, 92 (5): 1122-1129. 10.1002/1097-0142(20010901)92:5<1122::AID-CNCR1429>3.0.CO;2-H.CrossRefPubMed
37.
Zurück zum Zitat Ohta T, Elnemr A, Kitagawa H, Kayahara M, Takamura H, Fujimura T, Nishimura G, Shimizu K, Yi SQ, Miwa K: Fas ligand expression in human pancreatic cancer. Oncol Rep. 2004, 12 (4): 749-754.PubMed Ohta T, Elnemr A, Kitagawa H, Kayahara M, Takamura H, Fujimura T, Nishimura G, Shimizu K, Yi SQ, Miwa K: Fas ligand expression in human pancreatic cancer. Oncol Rep. 2004, 12 (4): 749-754.PubMed
38.
Zurück zum Zitat Chen M, Xue X, Wang F, An Y, Tang D, Xu Y, Wang H, Yuan Z, Gao W, Wei J, Zhang J, Miao Y: Expression and promoter methylation analysis of ATP-binding cassette. Oncol Rep. 2012, 27 (1): 265-269.PubMed Chen M, Xue X, Wang F, An Y, Tang D, Xu Y, Wang H, Yuan Z, Gao W, Wei J, Zhang J, Miao Y: Expression and promoter methylation analysis of ATP-binding cassette. Oncol Rep. 2012, 27 (1): 265-269.PubMed
39.
Zurück zum Zitat Chen KG, Valencia JC, Gillet JP, Hearing VJ, Gottesman MM: Involvement of ABC transporters in melanogenesis and the development of multidrug resistance of melanoma. Pigment Cell Melanoma Res. 2009, 22 (6): 740-749. 10.1111/j.1755-148X.2009.00630.x.CrossRefPubMedPubMedCentral Chen KG, Valencia JC, Gillet JP, Hearing VJ, Gottesman MM: Involvement of ABC transporters in melanogenesis and the development of multidrug resistance of melanoma. Pigment Cell Melanoma Res. 2009, 22 (6): 740-749. 10.1111/j.1755-148X.2009.00630.x.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Barrett LE, Granot Z, Coker C, Iavarone A, Hambardzumyan D, Holland EC, Nam HS, Benezra R: Self-renewal does not predict tumor growth potential in mouse models of high-grade glioma. Cancer Cell. 2012, 21 (1): 11-24. 10.1016/j.ccr.2011.11.025.CrossRefPubMed Barrett LE, Granot Z, Coker C, Iavarone A, Hambardzumyan D, Holland EC, Nam HS, Benezra R: Self-renewal does not predict tumor growth potential in mouse models of high-grade glioma. Cancer Cell. 2012, 21 (1): 11-24. 10.1016/j.ccr.2011.11.025.CrossRefPubMed
41.
Zurück zum Zitat Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, Brooks M, Reinhard F, Zhang CC, Shipitsin M, Campbell LL, Polyak K, Brisken C, Yang J, Weinberg RA: The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell. 2008, 133 (4): 704-715. 10.1016/j.cell.2008.03.027.CrossRefPubMedPubMedCentral Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, Brooks M, Reinhard F, Zhang CC, Shipitsin M, Campbell LL, Polyak K, Brisken C, Yang J, Weinberg RA: The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell. 2008, 133 (4): 704-715. 10.1016/j.cell.2008.03.027.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Ma Y, Liang D, Liu J, Axcrona K, Kvalheim G, Giercksky KE, Nesland JM, Suo Z: Synergistic effect of SCF and G-CSF on stem-like properties in prostate cancer cell lines. Tumour Biol. 2012, 33 (4): 967-978. 10.1007/s13277-012-0325-3.CrossRefPubMedPubMedCentral Ma Y, Liang D, Liu J, Axcrona K, Kvalheim G, Giercksky KE, Nesland JM, Suo Z: Synergistic effect of SCF and G-CSF on stem-like properties in prostate cancer cell lines. Tumour Biol. 2012, 33 (4): 967-978. 10.1007/s13277-012-0325-3.CrossRefPubMedPubMedCentral
43.
Zurück zum Zitat Levina V, Marrangoni A, Wang T, Parikh S, Su Y, Herberman R, Lokshin A, Gorelik E: Elimination of human lung cancer stem cells through targeting of the stem cell factor-c-kit autocrine signaling loop. Cancer Res. 2010, 70 (1): 338-346. 10.1158/0008-5472.CAN-09-1102.CrossRefPubMed Levina V, Marrangoni A, Wang T, Parikh S, Su Y, Herberman R, Lokshin A, Gorelik E: Elimination of human lung cancer stem cells through targeting of the stem cell factor-c-kit autocrine signaling loop. Cancer Res. 2010, 70 (1): 338-346. 10.1158/0008-5472.CAN-09-1102.CrossRefPubMed
44.
Zurück zum Zitat Simeone DM: Pancreatic cancer stem cells: implications for the treatment of pancreatic cancer. Clin Cancer Res. 2008, 14 (18): 5646-5648. 10.1158/1078-0432.CCR-08-0584.CrossRefPubMed Simeone DM: Pancreatic cancer stem cells: implications for the treatment of pancreatic cancer. Clin Cancer Res. 2008, 14 (18): 5646-5648. 10.1158/1078-0432.CCR-08-0584.CrossRefPubMed
Metadaten
Titel
Human pancreatic adenocarcinoma contains a side population resistant to gemcitabine
verfasst von
Anke Van den broeck
Lies Gremeaux
Baki Topal
Hugo Vankelecom
Publikationsdatum
01.12.2012
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2012
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/1471-2407-12-354

Weitere Artikel der Ausgabe 1/2012

BMC Cancer 1/2012 Zur Ausgabe

Update Onkologie

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