ArticlePDF Available

Epithelial-to-Mesenchymal Transition Mediates Docetaxel Resistance and High Risk of Relapse in Prostate Cancer

Authors:

Abstract

Molecular characterization of radical prostatectomy specimens after systemic therapy may identify a gene expression profile for resistance to therapy. This study assessed tumor cells from patients with prostate cancer (PC) participating in a phase-II neoadjuvant docetaxel (D) and androgen deprivation (AD) trial to identify mediators of resistance. Transcriptional level of 93 genes from a D-resistant PC cell lines microarray study was analyzed by Taqman low-density arrays in tumors from patients with high-risk localized PC (36 surgically treated, 28 with neoadjuvant D+AD). Gene expression was compared between groups and correlated with clinical outcome. VIM, AR and P65 were validated by immunohistochemistry. CD44 and ZEB1 expression was tested by immunofluorescence in cells and tumor samples. Parental and D-resistant CRPC cell lines were tested for epithelial-to-mesenchymal transition (EMT) markers before and after D-exposure. Reversion of EMT phenotype was investigated as a D-resistance reversion strategy. Expression of 63 (67.7%) genes differed between groups (P<0.05), including genes related to androgen receptor, NFKB transcription factor, and EMT. Increased EMT markers expression correlated with radiological relapse. D-resistant cells had increased EMT and stem-like cell markers expression. ZEB1 siRNA transfection reverted D-resistance and reduced CD44 expression in DU-145R and PC-3R. Before D-exposure, a selected CD44+ subpopulation of PC-3 cells exhibited EMT phenotype and intrinsic D-resistance; ZEB1/CD44+ subpopulations were found in tumor cell lines and primary tumors; this correlated with aggressive clinical behavior. This study identifies genes potentially related to chemotherapy resistance and supports evidence of the EMT role in docetaxel resistance and adverse clinical behavior in early PC.
2014;13:1270-1284. Published OnlineFirst March 21, 2014.Mol Cancer Ther
Mercedes Marín-Aguilera, Jordi Codony-Servat, Òscar Reig, et al.
Resistance and High Risk of Relapse in Prostate Cancer
Epithelial-to-Mesenchymal Transition Mediates Docetaxel
Updated version
10.1158/1535-7163.MCT-13-0775doi:
Access the most recent version of this article at:
Material
Supplementary
http://mct.aacrjournals.org/content/suppl/2014/03/21/1535-7163.MCT-13-0775.DC1.html
Access the most recent supplemental material at:
Cited Articles
http://mct.aacrjournals.org/content/13/5/1270.full.html#ref-list-1
This article cites by 49 articles, 18 of which you can access for free at:
E-mail alerts related to this article or journal.Sign up to receive free email-alerts
Subscriptions
Reprints and
.pubs@aacr.org
To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at
Permissions
.permissions@aacr.org
To request permission to re-use all or part of this article, contact the AACR Publications Department at
on May 23, 2014. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from
Published OnlineFirst March 21, 2014; DOI: 10.1158/1535-7163.MCT-13-0775
on May 23, 2014. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from
Published OnlineFirst March 21, 2014; DOI: 10.1158/1535-7163.MCT-13-0775
Cancer Biology and Signal Transduction
Epithelial-to-Mesenchymal Transition Mediates Docetaxel
Resistance and High Risk of Relapse in Prostate Cancer
Mercedes Marín-Aguilera
1
, Jordi Codony-Servat
1
,
Oscar Reig
1
, Juan Jos
e Lozano
2
, Pedro Luis Fern
andez
3,5
,
María Ver
onica Pereira
1
, Natalia Jim
enez
1
, Michael Donovan
6
, Pere Puig
6
, Lourdes Mengual
4
,
Raquel Bermudo
3,7
, Albert Font
8
, Enrique Gallardo
9
, María Jos
e Ribal
4
, Antonio Alcaraz
3,4
, Pere Gasc
on
1,3
, and
Bego~
na Mellado
1,3
Abstract
Molecular characterization of radical prostatectomy specimens after systemic therapy may identify a gene
expression profile for resistance to therapy. This study assessed tumor cells from patients with prostate cancer
participating in a phase II neoadjuvant docetaxel and androgen deprivation trial to identify mediators of
resistance. Transcriptional level of 93 genes from a docetaxel-resistant prostate cancer cell lines microarray
study was analyzed by TaqMan low-density arrays in tumors from patients with high-risk localized prostate
cancer (36 surgically treated, 28 with neoadjuvant docetaxel þandrogen deprivation). Gene expression was
compared between groups and correlated with clinical outcome. VIM, AR and RELA were validated by
immunohistochemistry. CD44 and ZEB1 expression was tested by immunofluorescence in cells and tumor
samples. Parental and docetaxel-resistant castration-resistant prostate cancer cell lines were tested for
epithelial-to-mesenchymal transition (EMT) markers before and after docetaxel exposure. Reversion of EMT
phenotype was investigated as a docetaxel resistance reversion strategy. Expression of 63 (67.7%) genes
differed between groups (P<0.05), including genes related to androgen receptor, NF-kB transcription factor,
and EMT. Increased expression of EMT markers correlated with radiologic relapse. Docetaxel-resistant cells
had increased EMT and stem-like cell markers expression. ZEB1 siRNA transfection reverted docetaxel
resistance and reduced CD44 expression in DU-145R and PC-3R. Before docetaxel exposure, a selected CD44
þ
subpopulation of PC-3 cells exhibited EMT phenotype and intrinsic docetaxel resistance; ZEB1/CD44
þ
subpopulations were found in tumor cell lines and primary tumors; this correlated with aggressive clinical
behavior. This study identifies genes potentially related to chemotherapy resistance and supports evi-
dence of the EMT role in docetaxel resistance and adverse clinical behavior in early prostate cancer. Mol
Cancer Ther; 13(5); 1270–84. 2014 AACR.
Introduction
Prostate cancer is the most common malignancy in the
Western world and the second most common cause of
cancer-related mortality in men (1). Although most pat-
ients with metastatic prostate cancer respond to androgen
deprivation therapy, virtually all of them eventually
develop castration-resistant prostate cancer (CRPC). In
2004, the combination of docetaxel and prednisone was
established as the new standard of care for patients with
CRPC (2). More recently, two hormonal agents, abirater-
one and enzalutamide, and a new taxane, cabazitaxel,
have been approved for the treatment of CRPC (3–5).
However, current therapies are not curative and research
is needed to identify predictors of benefit and mechanisms
of resistance for each agent.
To date, several factors have been associated with
docetaxel resistance, including expression of isoforms of
b-tubulin (6), activation of drug efflux pumps (7), PTEN
loss (8), and expression and/or activation of survival
factors (i.e., PI3K/AKT1 and MTOR; refs. 9, 10). Previous
work by our group and others correlated the activation
of NF-kB/interleukin (IL)-6 pathways with docetaxel
resistance in CRPC models and in patients (11–13).
Other studies support a role of JUN/AP-1, SNAI1, and
Authors' Afliations:
1
Laboratory of Translational Oncology and Medical
Oncology Department;
2
Bioinformatics Platform Department, Centro de
Investigaci
on Biom
edica en RedEnfermedades Hep
aticas y Digestivas
(CIBEREHD), Hospital Clínic;
3
Institut d'Investigacions Biom
ediques August
Pi i Sunyer(IDIBAPS);
4
Laboratoryand Departmentof Urology,Hospital Clínic,
Barcelona;
5
Department of Pathology, Hospital Clínic, Universitat de Barce-
lona;
6
Althia;
7
Tumor Bank, Hospital ClínicIDIBAPS Biobank, Barcelona;
8
Medical Oncology Department, Hospital Germans Trias i Pujol, Catalan
Institute of Oncology, Badalona; and
9
Medical Oncology Department, Hos-
pital Parc Taulí, Sabadell, Spain
Note: Supplementary data for this article are available at Molecular Cancer
Therapeutics Online (http://mct.aacrjournals.org/).
M. Marín-Aguilera and J. Codony-Servat contributed equally to this work.
Corresponding Author: Bego~
na Mellado, Medical Oncology Department,
Hospital Clínic de Barcelona, Villarroel 170, Barcelona, 08036, Spain.
Phone: 34-93-227-5400, ext. 2262; Fax: 34-93-454-6520; E-mail:
bmellado@clinic.ub.es
doi: 10.1158/1535-7163.MCT-13-0775
2014 American Association for Cancer Research.
Molecular
Cancer
Therapeutics
Mol Cancer Ther; 13(5) May 2014
1270
on May 23, 2014. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from
Published OnlineFirst March 21, 2014; DOI: 10.1158/1535-7163.MCT-13-0775
NOTCH2/Hedgehog signaling pathways in the devel-
opment of resistance to docetaxel or paclitaxel (14, 15).
Moreover, it has been shown that the inhibition of andro-
gen receptor (AR) nuclear translocation and AR activity
may be an important mechanism of taxane action in
prostate cancer (9).
In previous work, we identified 243 genes with dif-
ferential expression in CRPC docetaxel-sensitive versus
docetaxel-resistant cell lines (16). In the present study,
73 genes from that study together with 20 genes from
the literature were tested in tumor specimens of patients
with high-risk localized prostate cancer included in a
clinical trial of neoadjuvant hormone chemotherapy
(17), and compared with nontreated specimens with
similar clinical characteristics. This approach was based
on the notion that residual tumor cells in prostatectomy
specimens after neoadjuvant systemic therapy are likely
enriched for resistant tumor cells and their molecular
characterization may provide important information on
mechanisms of resistance (18). Our key findings were
then tested in two models of docetaxel-resistant prostate
cancer cell lines.
Patients and Methods
Patients and samples
The study included 28 patients with high-risk localized
prostate cancer from a previously published, multicenter,
phase II trial of neoadjuvant docetaxel plus androgen
deprivation followed by radical prostatectomy (17) and
36 control patients with high-risk prostate cancer treated
with radical prostatectomy without neoadjuvant treat-
ment. Of the 57 participants in the clinical trial (17), 29
were not included in this study: 23 patients did not
consent to participation in the molecular substudy and
insufficient material for molecular analysis was available
for 6 patients, 3 of whom had a pathologic complete
response (pCR) and 3 had microscopic residual tumor
(near pCR) in the prostate specimen.
Inclusion criteria were histologically confirmed adeno-
carcinoma of the prostate with any of the following three
risk criteria: (i) clinical stage T3; (ii) clinical stage T1c or T2
with serum prostate-specific antigen (PSA) >20 ng/mL
and/or Gleason score sum of 8, 9, or 10; or (iii) a Gleason
sum of 7 with a predominant form of 4 (i.e., Gleason score
4þ3). Clinical characteristics are shown in Table 1.
Treatment consisted of three cycles every 28 days of
docetaxel 36 mg /m
2
on days 1, 8, and 15 concomitant with
complete androgen blockade, followed by radical pros-
tatectomy. Patients were followed from the time of study
inclusion until death or last visit. Median follow-up time
was 82 months (range, 10–135). PSA relapse was defined
as two consecutive values of 0.2 ng/mL or greater (19).
Radiologic progression was defined as the progression in
soft tissue lesions measured by computed tomography or
MRI, or by progression to bone (20).
The study was approved by the Institutional Ethics
Committee of each participating hospital and written
informed consent was obtained from all participants.
Formalin-fixed paraffin-embedded (FFPE) specimens
were collected after radical prostatectomy. A representa-
tive tumor area was selected for each block and, according
to its size, between 2 and 12 sections were cut, 10-mm-
thick, and used for RNA isolation. Hematoxylin and eosin
(H&E)–stained sections from tumors and adjacent tissues
were prepared to confirm the histologic diagnosis.
RNA extraction
Total RNA was isolated from tumor specimens using
the RecoverAll Total Nucleic Acid Isolation Kit (Life
Technologies) according to the manufacturer’s protocol.
Total RNA was quantified with a spectrophotometer
(NanoDrop Technologies).
Gene selection
In total, 93 targe t genes that could potentially be related
to docetaxel resistance and two endogenous control
genes (ACTB and GUSB) were selected for further anal-
ysis in tumors. A set of 73 target genes was selected for
their relative expression in docetaxel-resistant cells
(DU-145R and PC-3R) versus parental cells (DU-145 and
PC-3; ref. 16) using DAVID (21) and Ingenuity Pathway
Analysis software (http://www.ingenuity.com). Twen-
ty genes highlighted in the literature as potential targets
of docetaxel resistance were also selected.
Reverse transcription and preamplification
A High-Capacity cDNA Reverse Transcription Kit (Life
Technologies) was used to reverse transcribe 1 mg of total
RNA in a 50 mL reaction volume. cDNA preamplification
was performed by multiplex PCR with the 93 selected
genes (Supplementary Table S1) and the stem-like cell
markers CD24 and CD44, following the manufacturer’s
instructions for the TaqMan PreAmp Master Mix Kit (Life
Technologies), except that final volume of the reaction was
25 mL.
Gene expression analysis in FFPE samples
Preamplified cDNA was used for gene expression anal-
ysis using 384-Well Microfluidic Cards (Life Technolo-
gies). Preamplified samples were diluted 1:20 in TE 1X
buffer before use. Each card was configured into four
identical 96-gene sets (95 selected genes plus an endog-
enous control gene, RNA18S, by default). The reaction was
carried out following the manufacturer’s instructions on
an ABI 7900HT instrument (Life Technologies). Array
cards were analyzed with RQ Manager Software for
manual data analysis.
Gene expression of CD24 and CD44 markers was studied
by amplifying with TaqMan Gene Expression Master Mix
in a StepOnePlus Real-Time PCR system (Life Technolo-
gies), according to the manufacturer’s recommendations.
Relative gene expression values were calculated on the
basis of the quantification cycle values obtained with SDS
2.4 software (Life Technologies). Expression values were
relative to the GUSB endogenous gene. Samples from
EMT Role in Docetaxel Resistance
www.aacrjournals.org Mol Cancer Ther; 13(5) May 2014 1271
on May 23, 2014. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from
Published OnlineFirst March 21, 2014; DOI: 10.1158/1535-7163.MCT-13-0775
patients who did not receive neoadjuvant treatment were
used for calibration.
Cell culture conditions
The CRPC cell lines DU-145 and PC-3 were purchased
from the American Type Culture Collection in October
2009. The docetaxel-resistant cell lines DU-145R and
PC-3R were developed and maintained as previously
described (12). No further authentication of the cell lines
was done by the authors.
Cell proliferation assays
Cell viability in response to docetaxel was assessed by
an MTT assay with the CellTiter 96 Aqueous Proliferation
Table 1. Clinical characteristics of patients
All
Neoadjuvant
treatment Control
Total number 64 28 36
Median age, y 64 (range, 4674) 64 (range, 4870) 64.5 (range, 4674)
Clinical Stage
T1 19 (29.7%) 4 (14.3%) 15 (41.7%)
T2 33 (51.6%) 14 (50%) 18 (50%)
T3 12 (18.8%) 10 (35.7%) 3 (8.3%)
Pathologic stage
T0 1 (1.6%) 1 (3.6%) 0
T1 1 (1.6%) 1 (3.6%) 0
T2 26 (40.6%) 13 (46.4%) 13 (36.1%)
T3 36 (56.3%) 13 (46.4%) 23 (63.9%)
Gleason score (biopsy)
6 10 (15.6%) 2 (7.1%) 8 (22.2%)
7(3þ4) 22 (34.4%) 7 (25%) 15 (41.7%)
7(4þ3) 16 (25%) 8 (28.6%) 8 (22.2%)
7 (N/A
a
)——1 (2.8%)
8 11 (17.2%) 8 (28.6%) 3 (8.3%)
9 4 (6.3%) 3 (10.7%) 1 (2.8%)
Gleason score (prostatectomy)
N/A
b
18 (28.3%) 18 (64.3%) 0
6 7 (15.2%) 7 (25%) 0
7(3þ4) 13 (20.3%) 1 (3.6%) 12 (33.3%)
7(4þ3) 17 (26.6%) 17 (47.2%)
8 2 (4.3%) 0 2 (5.6%)
9 7 (15.2%) 2 (7.1%) 5 (13.9%)
Median PSA (ng/mL) 8.7 (range, 2.0141) 12.2 (range, 4.741) 8.2 (range, 2.0119.2)
PSA (ng/mL)
<20 56 (87.5) 20 (71.4%) 36 (100%)
>20 8 (12.5%) 8 (28.6%) 0 (0%)
Postoperative radiotherapy
No 35 (58.3%) 13 (54.2%) 22 (61.1%)
Yes 24 (40%) 10 (41.7%) 14 (38.9%)
N/A
a
1 (1.7%) 1 (4.2%) 0 (0%)
Biochemical relapse
No 30 (46.9%) 11 (39.3%) 19 (52.8%)
Yes 34 (53.1%) 17 (60.7%) 17 (47.2%)
Median biochemical relapsefree survival (mo) 31.7 (range, 481) 29.3 (range, 459) 34.1 (range, 881)
Clinical relapse
No 58 (90.6%) 22 (78.6%) 36 (100%)
Yes 6 (9.4%) 6 (21.4%) 0 (0%)
Median clinical relapsefree survival (mo) 51.2 (range, 3184) 51.2 (range, 3184)
Follow-up (mo) 82 (range, 10135) 91 (range, 8196) 69 (range, 10135)
Abbreviation: N/A, not available
a
Missing information.
b
In some cases Gleason score could not be assessed because of tissue changes related to neoadjuvant treatment.
Marín-Aguilera et al.
Mol Cancer Ther; 13(5) May 2014 Molecular Cancer Therapeutics
1272
on May 23, 2014. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from
Published OnlineFirst March 21, 2014; DOI: 10.1158/1535-7163.MCT-13-0775
Assay Kit (Promega) and by the trypan blue exclusion
method using a Neubauer hemocytometer chamber.
Western blot analysis
Whole-cell extracts were prepared and Western blot
analysis performed as described previously (22). Antibo-
dies used were anti-PARP. Antibody was purchased from
Roche (ref. 11835238001); b-catenin (6B3; CTNNB1) anti-
body (ref. 9582), CD44 (156-3C11) Mouse mAb (monoclo-
nal antibody; ref. 3570), E-cadherin (CDH1) antibody (ref.
4065), Snail (C15D3) Rabbit mAb (ref. 3879), TCF8/ZEB1
(D80D3) Rabbit mAb (ref. 3396), and Vimentin (R28; VIM)
antibody (ref. 3932) were purchased from Cell Signaling
Technology. Monoclonal anti–a-tubulin clone B-5-1-2 (ref.
T5168) was purchased from Sigma-Aldrich.
Real-time qRT-PCR in cell lines
Total RNA was isolated from cell lines using the
RNeasy Micro Kit (Qiagen), and quantified with a spec-
trophotometer (Nanodrop Technologies). cDNA was gen-
erated from 1 mg of total RNA using the High Capacity
cDNA Archive Kit (Life Technologies), following the
manufacturer’s instructions. Real-time quantitative rev-
erse transcription PCR (qRT-PCR) was carried out in a
StepOnePlus Real-Time PCR system (Life Technologies)
according to the manufacturer’s recommendations. Data
were acquired using SDS Software 1.4. Amplification
reactions were performed in duplicate. Expression values
were relative to the ACTB endogenous gene. Target genes
were amplified using commercial primers and probes
(Life Technologies; Supplementary Table S1).
Immunohistochemistry
Tissue sections were deparaffinized in xylene and rehy-
drated in graded alcohols. For AR and VIM staining, the
sections were placed in a 97C solution of 0.01 mol/L
EDTA (pH 9.0) for antigen retrieval. Primary mouse
mononuclear antibody for AR (DAKO; Agilent Technol-
ogies) was applied for 20 minutes at room temperature at
dilution 1:150. FLEX Monoclonal Mouse anti-VIM, Clon
V9 (DAKO) was used for VIM staining. Detection was
accomplished with the DAKO Envision System followed
by diaminobenzidine enhancement. For RELA, the sec-
tions were placed in a 97C solution of 0.01 mol/L sodium
citrate (pH 6.0) for antigen retrieval. Then, samples were
incubated with a rabbit polyclonal antibody (Santa Cruz
Biotechnology, Inc.) at dilution 1:400. Detection was per-
formed with Bond Polymer Refine Detection (DAKO;
Agilent Technologies) for the automated Bond system.
AR and RELA were evaluated throughout the semi-
quantitative method histologic score (H-score), which
measures both the intensity and proportion of staining.
The H-score for each sample was calculated by multiply-
ing the percentage of stained tumor cells by the intensity
(0, nonstained; 1, weak; 2, moderate; 3, strong). VIM was
evaluated in the same way but scoring the percentage of
staining on a scale of 0 to 4 (0, 0; 1, <1%; 2, 1%–9%; 3, 10%–
50%; 4, >50%). Nuclear and cytoplasmatic stains were
scored separately for AR and RELA proteins. The assess-
ment of all samples was done by a senior pathologist (P.L.
Fern
andez) who was blinded to all clinical information.
Immunofluorescence staining in cell lines and tumor
samples
Cell pellets were collected in a 1% agarose solution,
fixed in 4% PBS-buffered formaldehyde, and then FFPE.
Sections of 5 mm were analyzed with a multiplex immu-
nofluorescent assay. They were stained with H&E for
histopathologic assessment and stained using immuno-
fluorescence with Alexa fluorochrome–labeled antibo-
dies. Briefly, both control and resistant prostate cancer
cell lines were evaluated with a series of simplex and
duplex immunofluorescence assays to quantify the level
of selected antibody–antigen complexes from specific
regions of interest (ROI).
The FFPE prostate tissue sections also were assessed by
immunofluorescence using a single multiplex assay with
two differentially labeled antibodies (ZEB1 and CD44).
For all specimens the H&E images were used to guide and
register immunofluorescence image capture with a max-
imum of four ROIs per cell pellet and six per tissue section.
Alexa fluorochrome dyes were Vimentin (ref. MO725;
Dako), CD44 (ref. 156-3C11; Cell Signaling Technology),
ZEB1 (ref. sc-25388; Santa Cruz Biotechnology). The ROIs
were acquired from the cells and tumor tissue sections,
blinded to outcome, with a CRI Nuance imaging system,
and then analyzed with fluorescent image analysis soft-
ware to derive quantitative features from cellular/tissue
compartments. Quantitative assessment was performed
using a pixel-area function, normalized to the ROI under
investigation.
siRNA transfection
Dharmacon SMART pool control and ZEB1 siRNA
were used with lipofectamine according to the manufac-
turer’s protocol (Thermo Scientific) to inhibit ZEB1 in DU-
145/R cells. Commercial Silencer Select siRNA of ZEB1
(s229971; Life Technologies) was transfected to PC-3/R
cell lines. Cells were incubated with the siRNA complex
for 24 hours, treated with docetaxel, then harvested to
study protein expression changes of ZEB1 and CDH1 by
Western blot analysis. Apoptosis was studied at 24 and 48
hours by PARP analysis (Western blot analysis), and cell
viability was measured by MTT at 72 hours as described
before.
Fluorescence-activated cell sorting
For flow cytometry, cells were dissociated with Accutase
(Invitrogen) and washed twice in a serum-free medium.
Cells were stained live in the staining solution containing
bovine serum albumin andfluorescein isothiocyanate-con-
jugated monoclonal anti-CD44 (15 minat 4C). A minimum
of 500,000 viable cells per sample were analyzed on a
cytometer. For fluorescence-activated cell sorting (FACS),
2to510
7
cells were similarly stained for CD44 and used
to sort out CD44
þ
and CD44
cells. For the positive
EMT Role in Docetaxel Resistance
www.aacrjournals.org Mol Cancer Ther; 13(5) May 2014 1273
on May 23, 2014. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from
Published OnlineFirst March 21, 2014; DOI: 10.1158/1535-7163.MCT-13-0775
Table 2. Multivariate analysis of gene expression and patient outcomes
Gene FC Progression
Multivariate
a
HR (95% CI) Gene FC Progression
Multivariate
a
HR (95% CI) Gene FC Progression
Multivariate
a
HR (95% CI)
Differentially expressed genes
TGFBR3 4.48 rPFS ABCB1 2.46 NFKB1 1.76 bPFS
SERPINB5 4.43 CDH2 2.45 AR 1.73 rPFS
CST6 4.19 LTB 2.40 PTPRM 1.71
CLDN11 3.69 rPFS, bPFS 3.056
(1.1697.988)
rPFS
TIMP2 2.38 rPFS REL 1.68 bPFS
GPR87 3.65 bPFS ID2 2.38 KLF9 1.50 rPFS
AREG 3.42 EFEMP1 2.35 rPFS BRCA1 1.47
SCD5 3.32 rPFS FRMD3 2.32 SMAD4 1.36 bPFS 0.163
(0.0580.457)
TMEM45A 3.30 rPFS HTRA1 2.31 FRMD4A 1.31
MAP7D3 3.23 LAMC2 2.22 bPFS 0.131
(0.0270.634)
GSPT2 1.29
VIM 3.23 rPFS SLC1A3 2.17 rPFS, bPFS 2.555
(1.0885.999)
rPFS
FN1 1.20
BCL2A1 3.09 ZEB1 2.16 rPFS GOSR2 1.16
PLSCR4 3.01 rPFS IFI16 2.15 NDRG1 1.27
SCARA3 3.01 EGFR 2.11 BTBD11 1.41 bPFS 0.321
(0.1430.720)
ITGB2 2.92 SAMD9 2.09 CCNB1 1.41
SAMD12 2.80 rPFS, bPFS FBN1 2.03 ESRP1 1.46
S100A4 2.80 rPFS FAS 1.91 FBP1 1.53
G0S2 2.79 TACSTD2 1.83 EPCAM 1.73
SLCO4A1 2.75 rPFS RELA 1.81 rPFS AIM1 1.98
SNAI1 2.71 TXNIP 1.77 rPFS FLJ27352 2.09 rPFS, bPFS 0.257
(0.1130.584)
rPFS
IL6 2.64 rPFS 0.112
(0.0150.856)
rPFS
KLHL24 1.77 rPFS ST14 2.19 rPFS
LOC401093 2.49 EML1 1.76 rPFS C1orf116 6.08 rPFS
(Continued on the following page)
Marín-Aguilera et al.
Mol Cancer Ther; 13(5) May 2014 Molecular Cancer Therapeutics
1274
on May 23, 2014. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from
Published OnlineFirst March 21, 2014; DOI: 10.1158/1535-7163.MCT-13-0775
population, only the top 10% mostly brightly stained cells
were selected. The CD44
þ
cells selected were culturedas an
individual clone in 96-well plates and expanded.
Statistical analysis
TaqMan low-density arrays (TLDA) gene expression
data were evaluated by the Wilcoxon rank-sum test and
receiver operating characteristic (ROC) analysis. Time to
PSA progression and radiologic progression were calcu-
lated from the time of prostate cancer diagnosis until PSA
or radiologic progression, respectively. The log-rank test
was used in univariate survival analyses. Multivariate
analysis of gene expression was evaluated by Cox pro-
portional hazards regression, including stage, Gleason,
PSA, and neoadjuvant treatment as clinical covariates;
backward stepwise likelihood was used for selection.
Real-time qRT-PCR experimental data were expressed as
mean SEM and were analyzed by the Student ttest. All
the statistical tests were conducted at the two-sided 0.05
level of significance.
Results
Differential gene expression between treated and
nontreated tumors
Among the 93 genes analyzed (Supplementary Table
S1), we observed differential expression (P<0.05) in 63
(67.7%) genes (Table 2); 53 genes were overexpressed and
10 underexpressed in tumor specimens from patients
treated with neoadjuvant docetaxel plus androgen depri-
vation. Genes of the NF-kB pathway (such as NFKB1,REL,
and RELA), AR, and epithelial-to-mesenchymal transition
(EMT)–related genes (such as ZEB1,VIM,CDH2, and
TGFBR3) were overexpressed in treated tumors. Among
the downregulated genes in treated tumors, were the
metastasis-suppressor gene NDRG1 (23) and the adhesion
molecule EPCAM, a regulator of the alternative splicing of
CD44 (ESRP1; ref. 24) and ST14 (a negative regulator of the
EMT mediator ZEB1; Table 2; Fig. 1A; ref. 25).
Gene expression and clinical outcome
We tested the possible prognostic impact of the 93 genes
studied by TLDAs (Supplementary Table S1). Individu-
ally, the expression of several genes was related to time-to-
PSA and/or clinical relapse (Table 2). Time to radiologic
progression and PSA progression curves are shown
in Fig. 1B and C and Supplementary Figs. S1 and S2. Of
note, the overexpression of AR, and the EMT-related
genes TGFBR3,ZEB1, and VIM was correlated with a
shorter time of radiologic progression (Fig. 1B).
We then performed a multivariate analysis, including
the genes with individual prognostic value, clinical prog-
nostic factors (PSA, Gleason, and clinical stage), and
neoadjuvant treatment. Results are shown in Table 2A
and B. In the multivariate analysis, the reduced expression
of CLDN7 was an adverse-independent prognostic factor
for clinical relapse. Loss of CLDN7 has been correlated
with adverse prognostic variables in prostate cancer and
Table 2. Multivariate analysis of gene expression and patient outcomes (Cont'd )
Gene FC Progression
Multivariate
a
HR (95% CI) Gene FC Progression
Multivariate
a
HR (95% CI) Gene FC Progression
Multivariate
a
HR (95% CI)
Nondifferentially expressed genes
CBLB ns bPFS 0.315
(0.1430.695)
CSCR7 ns bPFS 0.241
(0.0770.755)
RAB40B ns bPFS 0.314
(0.1340.739)
CCPG1 ns bPFS 0.263
(0.1180.585)
EPS8L1 ns bPFS SCEL ns rPFS
CDH1 ns bPFS 0.446
(0.2170.917)
IGF1R ns bPFS SERPINA1 ns rPFS 0.032
(0.0020.632)
CDK19 ns rPFS LOC401093 ns rPFS TP53INPL ns rPFS
CLDN7 ns rPFS 0.054
(0.0040.699)
MALAT1 ns rPFS, bPFS 0.361
(0.1410.921)
rPFS
NOTE: False discovery rate for differentially expressed genes was <0.074 in all cases.
Abbreviations: bPFS, biochemical progression-free survival; FC, fold change; rPFS, radiologic progression-free survival.
a
Signicant Cox regression analysis; HR (95% condence interval, CI); P<0.05.
EMT Role in Docetaxel Resistance
www.aacrjournals.org Mol Cancer Ther; 13(5) May 2014 1275
on May 23, 2014. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from
Published OnlineFirst March 21, 2014; DOI: 10.1158/1535-7163.MCT-13-0775
ANontreated Treated
C
0 20 40 60 80 100 120
Low expression
CD24
High expression
P= 0.01
1.0
0.8
0.6
0.4
0.2
0.0
Time (mo)
Proportion of biochemical relapse-free survival
B
Proportion of radiologic progression-free survival
P = 0.009
VIM Low expression
High expression
1.0
0.8
0.6
0.4
0.2
0.0
P = 0.015
AR Low expression
High expression
0 25 50 75 100 125 0 25 50 75 100 125
0 25 50 75 100 125 0 25 50 75 100 125
Time (mo) Time (mo)
Time (mo) Time (mo)
1.0
0.8
0.6
0.4
0.2
0.0
P = 0.044
ZEB1 Low expression
High expression
1.0
0.8
0.6
0.4
0.2
0.0
P = 0.024
TGFB3R Low expression
High expression
1.0
0.8
0.6
0.4
0.2
0.0
Proportion of radiologic progression-free survival
E Low expression
Cytoplasmatic AR
High expression
P = 0.005
1.0
0.8
0.6
0.4
0.2
0.0
P = 0.035
Low expression
High expression
Nuclear RELA
0 25 50 75 100 125
Time (mo)
0 25 50 75 100 125
Time (mo)
1.0
0.8
0.6
0.4
0.2
0.0
Proportion of radiologic progression-free survival
High cytoplasmatic AR
60×
20×
20×
60×
High nuclear RELA
60×
20×
Low nuclear RELA
60×
20×
Low cytoplasmatic AR
F
D
Nuclear AR Cytoplasmic AR VIM Nuclear RELA Cytoplasmic RELA
IHC score (arbitrary units)
**
Controls (nontreated) Treated patients
300
250
200
50
0
100
150
Marín-Aguilera et al.
Mol Cancer Ther; 13(5) May 2014 Molecular Cancer Therapeutics
1276
on May 23, 2014. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from
Published OnlineFirst March 21, 2014; DOI: 10.1158/1535-7163.MCT-13-0775
with EMT (26). Of note, the low expression of CDH1 was
an independent prognostic factor for time to PSA relapse.
We also analyzed the prognostic impact of the stem-like
cell markers CD24 and CD44, which were underexpressed
and overexpressed, respectively, in treated tumors (FC
CD24: 0.59, P¼0.07; FC CD44: 1.63, P<0.000; Supple-
mentary Table S1 and Supplementary Figs. S1 and S2). Of
note, low expression of CD24 was correlated with shorter
time of biochemical progression (Fig. 1C).
Immunohistochemistry in treated versus nontreated
tumors
We explored the expression of VIM and both cytoplas-
matic and nuclear RELA and AR in tumor samples from
neoadjuvant-treated and -nontreated patients. Staining of
cytoplasmatic RELA was significatively higher in the trea-
ted versus nontreated patients [immunohistochemistry
(IHC) score 181.9 vs. 148.3, respectively; Fig. 1D and F].
Moreover, nuclear RELA was significantly related to worse
clinical relapse (Fig. 1E). Vimentin expression was nonsig-
nificantly higher in treated tumors (IHC score 2 vs. 1,
respectively; Fig. 1D). No differences were found in the
expression of nuclear AR; however, cytoplasmatic AR
expression was significantly higher in the treated tumors
(IHC score 102.5 vs. 14.5) and correlated with radiologic
progression survival (Fig. 1D–F).
Docetaxel-resistant prostate cancer cells express
EMT and stem-like cell markers
On the basis of the results described above, we studied
the link between EMT and docetaxel resistance in four
prostate cancer cell lines models (parental DU-145 and
PC-3R cells, and their docetaxel-resistant partners DU-
145R and PC-3R, respectively). As shown in Fig. 2A and B,
the docetaxel-resistant cells phenotype was consistent
with EMT, i.e., decreased expression of epithelial markers
(CDH1 and CTNNB1) and increased expression of mes-
enchymal markers (VIM and ZEB1) at the protein level.
Consistent results were found at mRNA level, except for
CTNNB1 (data not shown).
Recent studies have shown that cells with EMT pheno-
type share characteristics of stem-like cancer cells (14, 27).
For that reason, we tested the expression of stem-like cell
markers and showed that docetaxel-resistant cells, both
DU-145R and PC-3R, exhibit transcriptional features of
cancer-stem cells, such as increased expression of CD44
and the loss of CD24 (Fig. 2C).
Moreover, in cell lines, we detected by immunofluores-
cence analysis a subset of cells coexpressing CD44 and
ZEB1. Scattered cells with these features were detectable
in the parental cell lines; however, this population was
highly enriched in the resistant cells (Fig. 2D). By FACS,
we then isolated from the parental PC-3 cells a subpop-
ulation of cells with high expression of CD44. We selected
a derived CD44
þ
/PC-3 clone that showed an increased
expression of VIM and ZEB1 and decreased CDH1 expres-
sion (Fig. 2E). This clone from the parental cells was
significantly more resistant to docetaxel than the parental
cell line, PC-3 (Fig. 2F).
Dose–response experiments in both parental and resis-
tant cells showed that docetaxel exposure significantly
increased the expression of VIM in PC-3 and PC-3R cells,
of ZEB1 in PC-3 cells, and of SNAI1 in DU-145, PC-3 and
PC-3R cells. TWIST1 expression increased in all cell lines
after docetaxel treatment. In contrast, no significant dif-
ferences were observed in the expression of SNAI2 and
CDH1 with docetaxel exposure (Fig. 3A). About stem-like
cell markers, inconsistent results were obtained for CD24
expression after docetaxel exposure because CD24 exp-
ression increased in PC-3 cells but decrease in DU-145
cells. In contrast, CD44 significantly increased in PC-3
cells with docetaxel treatment (Fig. 3B).
EMT mediates docetaxel resistance in prostate
cancer cells
To test whether inhibition of EMT could revert doce-
taxel resistance, we downmodulated the expression of
ZEB1, a key inducer of EMT. siRNA ZEB1 transfected DU-
145R and PC-3R cells had an increased expression of
CDH1 (Fig. 4A) and decreased CD44 (Fig. 4B), confirm-
ing the link between EMT and stem-like cell phenotype.
Moreover, siRNA ZEB1 transfected cells showed signif-
icantly increased sensitivity to docetaxel compared with
control cells (P<0.05; Fig. 4B and C). The magnitude of
the reversion of chemoresistance was more pronounced
in DU-145R and PC-3R cells than in the parental cells.
Docetaxel-induced apoptosis was more pronounced in
the ZEB1–siRNA transfected cells (Fig. 4B).
ZEB1/CD44 expression in tumor samples
On the basis of preclinical findings, we decided to
investigate whether CD44
þ
/ZEB1
þ
cells were present in
primary prostate cancer specimens. Twenty-two FFPE
tumors from patients with high-risk prostate cancer
Figure 1. The gene expression prole and related outcome of patients treated with neoadjuvancy versus nontreated patients. A, heatmap of differentially
expressed genes in tumor samples from neoadjuvant-treated patients, compared with those without treatment (P<0.05). Rows, genes; columns,
samples. Red pixels, upregulated genes; green pixels, downregulated genes. B, radiologic progression-free survival analysis of patients according to gene
expression of AR and the EMT-related markers TGFBR3,ZEB1, and VIM. High and low expression was established according to receiver operating
characteristic (ROC) curve analysis. The log-rank test was used to assess the statistical difference between the two groups (P<0.05). C, KaplanMeier curve
representing biochemical progression-free survival analysis of patients according to gene expression of the stem-like cell marker CD24. High and low
expression was established according to ROC curve analysis. The log-rank test was used to assess the statistical difference between the two groups
(P<0.05). D, IHC of VIM and nuclear and cytoplasmatic RELA and AR. Box plot, IHC scores for each protein. ,P<0.05. E, Kaplan Meier graphs representing
radiologic progression-free survival analysis of patients according to cytoplasmatic AR and RELA nuclear staining by IHC. The log-rank test was used
to assess the statistical difference between the two groups (P<0.05). F, images show representative immunohistoche mical staining for nuclear RELA and
cytoplasmatic AR protein in prostate cancer tumors. Magnications illustrate high and low staining of cells.
EMT Role in Docetaxel Resistance
www.aacrjournals.org Mol Cancer Ther; 13(5) May 2014 1277
on May 23, 2014. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from
Published OnlineFirst March 21, 2014; DOI: 10.1158/1535-7163.MCT-13-0775
treated with docetaxel and androgen suppression and 15
control patients with sufficient remaining material were
available for immunofluorescence studies. All samples
were positive for CD44 staining but only 7 of 15 controls
(46.7%) and 7 of 22 treated patients (31.8%) had a ZEB1
signal. Overall, there were no differences between the
control and treated groups in the expression of ZEB1
(0.0059 vs. 0.013 mean intensity, respectively) or CD44
(1.27 vs. 1.01 mean intensity, respectively). Tumor cells
that coexpressed ZEB1 and CD44
þ
were observed in 3
(13.6%) of the 22 patients in the neoadjuvant group.
However, none of the control patients presented with
coexpression of both markers (Fig. 4D). Notably, ZEB1/
CD44 coexpression was associated with aggressive clin-
ical behavior: At the time of outcome analysis, all patients
had relapsed, 2 had developed liver metastasis, and 1 had
died due to disease progression (Fig. 4E).
Discussion
In this study, we confirm that some of the molecular
alterations associated with docetaxel resistance in a pre-
viously described in vitro model of CRPC cell lines are
present in residual cells of prostatectomy specimens trea-
ted with neoadjuvant docetaxel plus androgen depriva-
tion. Our findings may be especially relevant in clinical
practice because most patients receive androgen depriva-
tion prior and concomitantly to the administration of
docetaxel. The observed deregulated pathways may trans-
late common mechanisms of resistance to both therapies.
DU-145 DU-145R
PC-3 PC-3R
D
F
0
1
1.8
2.5
5
10
0
50
100
150
PC-3
PC-3 clon
*
**
*
Docetaxel (nmol/L)
% of Viability
E
CDH1
VIM
ZEB1
Epithelial markers
Mesenchymal marker
Stem cell marker
PC-3 Clon
CD44
C
CD24
CD44
0
1
2
3
DU-145
DU-145R
PC-3
PC3-R
** **
Fold change
Stem cell markers
A
α-Tubulin
VIM
Mesenchymal
markers
ZEB1
DU-145 DU-145R PC-3 PC-3R
Epithelial
markers
DU-145 DU-145R
CDH1
CTNNB1
α-Tubulin
α-Tubulin
PC-3 PC-3R
CD44
ZEB1
CD44+ZEB1
CD44
ZEB1
CD44+ZEB1
CD44
ZEB1
CD44+ZEB1
CD44
ZEB1
CD44+ZEB1
B
CDH1
CDH2
VIM
SNAI2
SNAI1
TWIST
ZEB1
0.0
0.2
0.4
0.6
0.8
1.0
5
10
15
DU-145R
PC3-R
DU-145
PC-3
** **
*
*
*
*
EMT markers
Fold change
(compared with parental cells)
Figure 2. EMT and stem cell
markers in parental and docetaxel-
resistant cell lines. A, Western blot
analysis in DU-145, DU-145R, PC-
3, and PC-3R cell lines of epithelial
markers (CDH1 and CTNNB1) and
mesenchymal markers (VIM and
ZEB1). Tubulin was used as a load
control. B, gene expression of EMT
markers by qRT-PCR in DU-145,
DU-145R, PC-3, and PC-3R cell
lines. Data shown are the
mean SEM of cell lines from
triplicate experiments (,P<0.05;
,P<0.001). C, gene expression of
stem cell markers by qRT-PCR
in DU-145, DU-145R, PC-3, and
PC-3R cell lines. Data, mean
SEM of cell lines from triplicate
experiments (,P<0.001). D,
confocal immunouorescence of
CD44 (red) and ZEB1 (green) in
DU-145, DU-145R, PC-3, and PC-
3R lines. Colocalization of ZEB1
and CD44 results are in yellow.
Nuclei are stained with 40,6-
diamidino-2-phenylindole (blue). E,
Western blot analysis in parental
PC-3 and a subpopulation of
parental PC-3 cells (clone) sorted
by CD44 marker. Tubulin was used
as a load control. F, viability assay
of PC-3 and PC-3 clone under
docetaxel treatment performed by
the Tripan blue method (,P<0.05).
Marín-Aguilera et al.
Mol Cancer Ther; 13(5) May 2014 Molecular Cancer Therapeutics
1278
on May 23, 2014. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from
Published OnlineFirst March 21, 2014; DOI: 10.1158/1535-7163.MCT-13-0775
Different neoadjuvant studies have been designed to
identify pathways involved in resistance to androgen
deprivation or chemotherapy in prostate cancer. In one
study of neoadjuvant androgen deprivation, the authors
observed that many androgen-responsive genes, includ-
ing AR and PSA, were not suppressed; this suggests that
suboptimal suppression of tumoral androgen activity
may lead to adaptive cellular changes to allow prostate
cancer cells survival in a low-androgen environment (28).
Another group analyzed prostate tumors removed by
radical prostatectomy after 3 months of androgen depri-
vation. Gene expression analysis revealed that PSA and
other androgen-responsive genes were overexpressed in
tumors from patients who relapsed (29). Our data are in
concordance with these reports. We observed that the
expression of AR and several AR-regulated genes (i.e.,
ZEB1,IL6,TGFBR3,KLF9) increased in treated tumors,
even though serum PSA levels decreased under therapy
in most cases, as we previously reported (17). Moreover,
high levels of AR correlated with high risk of clinical
relapse. These data suggest that persistence of AR signal-
ing may be related to treatment resistance and/or to
eventual disease progression.
We observed no differences in AR nuclear staining
between treated and nontreated samples. However, cyto-
plasmatic expression was significantly higher in residual
tumor cells after androgen deprivation and docetaxel
exposure. Prior reports have shown that taxanes inhibit
AR nuclear translocation and that patients treated with
taxanes may have lower nuclear expression than treat-
ment-na
ve patients (30). This was not observed in our
study, likely because our patients were treated with com-
bined therapy. Prior studies have shown that androgen
deprivation increased full-length AR protein levels in
CRPC cells, but decreased its nuclear localization (31).
Other studies have used a similar approach in patients
treated with neoadjuvantchemotherapy alone (32, 33). One
group performed microarray analysis of tumor specimens
from 31 patients treated with docetaxel plus mitoxantrone
(33). The comparison of pre- and posttreatment samples
showed increased expression of cytokines regulated by the
NF-kB pathway. These data are in concordance with our
A
VIM
0
1
5
10
0
5
10
15 DU-145
DU-145R
PC-3
PC-3R
Docetaxel (nmol/L)
Fold change
SNAI1
0
1
2.5
5
10
0
5
10
15
20 DU-145
DU-145R
PC-3
PC-3R
Fold change
TWIST1
0
1
2.5
5
10
0
1
2
3
4DU-145
DU-145R
PC-3
PC-3R
Docetaxel (n
mol/L
)
Fold change
SNAI2
0
1
2.5
5
10
0
5
10
15 DU-145
DU-145R
PC-3
PC-3R
Fold change
ZEB1
0
1
2.5
5
10
0
10
20
30 DU-145
DU-145R
PC-3
PC-3R
Docetaxel (n
mol/L
)
Docetaxel (nmol/L)
Docetaxel (n
mol/L
)
Docetaxel (nmol/L)
Docetaxel (n
mol/L
)
Docetaxel (n
mol/L
)
Fold change
CDH1
0
1
2.5
5
10
0.000
0.005
0.5
1.0
1.5
2.0
2.5 DU-145
DU-145R
PC-3
PC-3R
Fold change
B
CD24
0
1
2.5
5
10
0
1
2
3DU-145
DU-145R
PC-3
PC-3R
Fold change
CD44
0
1
2.5
5
10
0
2
4
6
8DU-145
DU-145R
PC-3
PC-3R
Fold change
Figure 3. Effect of docetaxel exposure on EMT and stem-like gene expression markers in prostate cancer cell lines. A, EMT markers gene expression in a
docetaxel doseresponse manner. B, stem-like cell markers gene expression in a docetaxel doseresponse manner. Geometric al symbols represent
signicant differences in the corresponding cell line; data from DU-145 0 nmol/L were considered the reference for all the other measures (i.e., fold change,1).
EMT Role in Docetaxel Resistance
www.aacrjournals.org Mol Cancer Ther; 13(5) May 2014 1279
on May 23, 2014. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from
Published OnlineFirst March 21, 2014; DOI: 10.1158/1535-7163.MCT-13-0775
C Lipo si C Lipo si
DU-145 DU-145R
A
C Lipo si C Lipo si
PC-3 PC-3R
C
0
2
3.5
0
50
100
150
PC-3 lip
PC-3 siRNA ZEB1
PC-3R lip
PC-3R siRNA ZEB1
**
**
*
*
*
**
Docetaxel (nmol/L)
Cell viability (%)
0
5
10
0
50
100
150
DU-145 lip
DU-145 siRNA ZEB1
DU-145R lip
DU-145R siRNA ZEB1
*
*
*
*
*
*
*
*
*
**
*
*
Docetaxel (nmol/L)
Cell viability (%)
B
Docetaxel 10 nmol/L Docetaxel 10 nmol/L
DU-145 DU-145R
C Lipo si C Lipo si C Lipo si C Lipo si
CD44
PARP
TUB
Docetaxel 3 nmol/L Docetaxel 3 nmol/L
PC-3 PC-3R
C Lipo si C Lipo si C Lipo si C Lipo si
D
1.0
0.8
0.4
0.6
0.2
0.0
ZEB1 and CD44 Colocalization
No colocalization
Colocalization
Time (mo)
P = 0.371
0 20 40 60 80 100 120
Proportion of radiologic
progression-free survival
ZEB1 and CD44 Colocalization
No colocalization
Colocalization
P = 0.273
1.0
0.8
0.4
0.6
0.2
0.0
0 20 40 60 80 100
Proportion of radiologic
progression-free survival
E
CDH1
ZEB1
TUB
Time (mo)
Marín-Aguilera et al.
Mol Cancer Ther; 13(5) May 2014 Molecular Cancer Therapeutics
1280
on May 23, 2014. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from
Published OnlineFirst March 21, 2014; DOI: 10.1158/1535-7163.MCT-13-0775
results showing an increased expression in treated tumors
of NF-kB subunits and NF-kB–regulated cytokines, such as
IL-6, adding support to a body of evidence on the involve-
ment of this pathway in resistance to chemotherapy in
prostate cancer (11). On the other hand, NF-kB activation
may induce EMT in prostate cancer (34). Although our
study did not investigate the potential causal relationship
between NF-kB activation and EMT, this last phenomenon
was found to be highly relevant in resistance to therapy.
Moreover, increased nuclear NF-kB (RELA staining) cor-
related with a shorter time to clinical relapse, confirming
the prognostic value of this pathway activation in prostate
cancer (22).
In the present study, we analyze the transcriptional
profile of residual tumor cells after combined neoadjuvant
androgen deprivation and docetaxel treatment. Because
macrodissected tumor tissues were used for gene expres-
sion studies, our results may translate expression patterns
from both tumor and surrounding nontumor cells. How-
ever, a prior study using the macrodissection strategy
reported only minor interference of nontumor cells with
the overall gene expression profile (35). Moreover, we
considered stroma and benign cells contamination to be
homogeneous in both the treated and nontreated patient
groups. Among the 93 genes analyzed, we observed
differential expression between treated and nontreated
tumors in 63 (67.7%) genes. Of note, the over expression of
the EMT genes correlated with a shorter time to clinical
relapse.
In the EMT process, cells lose epithelial characteristics
and gain mesenchymal properties to increase motility and
invasion, allowing tumor cells to acquire the capacity to
infiltrate surrounding tissues and to metastasize in distant
sites. EMT is typically characterized by the loss of epithe-
lial (i.e., CDH1) and the gain of mesenchymal (i.e., VIM,
CDH2) markers expression (36). Several reports suggest
that AR activation, as well as androgen deprivation ther-
apy, may induce changes characteristic of EMT that may
be involved in prostate cancer progression (37–39). The
expression of the transcription factor ZEB1 may be
induced by dihydrotestosterone and is mediated by two
androgen-response elements (40). Recently, Sun and
colleagues showed that androgen deprivation causes
EMT in animal models and in tumor samples of patients
treated with hormone therapy (41). Moreover, the pres-
ence of AR-truncated isoforms, which are increased in
the castration-resistant progression, regulate the expres-
sion of EMT (42).
On the other hand, there are molecular similarities
between cancer stem-like cells and EMT phenotypic cells.
Moreover, cells with an EMT phenotype induced by
different factors are rich sources for stem-like cancer cells
(14, 27). We observed in the DU-145 in vitro model that
docetaxel-resistant cells expressed high levels of the stem
cell marker CD44 and decreased levels of CD24. More-
over, docetaxel treatment increased CD44 expression in
tumor cells. Likewise, RT-PCR results in tumor samples
showed an increased expression of CD44 and a decreased
expression of CD24 in tumors treated with neoadjuvant
androgen deprivation plus docetaxel. Our results are in
accordance with those of Puhr and colleagues, who
detected an increased CD24
low
–CD44
high
cell population
in docetaxel-resistant prostate cancer models (43). Simi-
larly, Li and colleagues detected CD24
low
–CD44
high
breast
cancer cells that were resistant to neoadjuvant chemo-
therapy (44). In a preclinical study, CD44 and CD147
enhanced metastatic capacity and chemoresistance of
prostate cancer cells, potentially mediated by activation
of the phosphoinositide 3-kinase and mitogen-activated
protein kinase pathways (45).
In the present work, we identified a population of
prostate cancer cells exhibiting an EMT phenotype that
are primarily resistant to docetaxel. The presence of an
intrinsic resistant cell population was supported by the
isolation of docetaxel-resistant clonal cells in the paren-
tal cell line PC-3, before docetaxel exposure, with a high
expression of CD44 and EMT markers and the loss of
CDH1. ZEB1
þ
/CD44
þ
cells were identified at a very low
frequency in the two parental cell lines, DU-145 and
PC3, before docetaxel exposure but their frequency
massively increased in docetaxel-resistant cells. Simi-
larly, a small percentage of ZEB1
þ
/CD44
þ
cells were
also observed in primary high-risk localized prostate
cancer tumors. ZEB1
þ
/CD44
þ
cells were present only in
tumors that had previously received neoadjuvant
androgen deprivation plus docetaxel (13.6%). Both
in vitro and tumor sample findings support the presence
of primary resistant cells harboring EMT/stem cell–like
characteristics and suggest that the exposure to doce-
taxel may eliminate sensitive cells resulting, however, in
the selective out-growth of this resistant cell population.
In our model, docetaxel also induced EMT changes in
the parental and resistant cell lines. On the basis of our
findings, both mechanisms, the existence of a primary
resistant cell with an EMT phenotype and the induction of
EMT changes induced by docetaxel, are possible. In recent
work on docetaxel-resistant PC-3– and DU-145–derived
cell lines, the authors reported that docetaxel-resistant
cells underwent an EMT transition associated with a
reduction of microRNA (miR)-200c and miR-205, which
Figure 4. Inhibition of ZEB1 in parental and docetaxel-resistant cell lines. ZEB1CD44 staining in prostate tumor specimens. A, Western blot analysis
of CDH1 and ZEB1 in the four cell lines (DU-145, DU-145R, PC-3, and PC-3R) when ZEB1 was inhibited by siRNA. B, Western blot analysis of CD44
and PARP in the four cell lines (DU-145, DU-145R, PC-3, and PC-3R transfected cells) treated with docetaxel; the band of CD44 in PC-3 and PC-3R
corresponds to the variant CD44v6. C, MTT of ZEB1siRNA transfected cells. Data, mean SEM of triplicate experiments. ,P<0.05. D, CD44 and ZEB1
immunouorescence image of a prostate tumor biopsy from a patient treated with neoadjuvant docetaxel and androgen deprivation. E, KaplanMeier
according to immunouorescence intensities of CD44ZEB1 colocalization and clinical/biochemical relapse of patients treated with neoadjuvant docetaxel
and controls without neoadjuvant treatment. C, nontrasnfected cells; Lipo, control lipofectamine; si, siRNAZEB1.
www.aacrjournals.org Mol Cancer Ther; 13(5) May 2014 1281
EMT Role in Docetaxel Resistance
on May 23, 2014. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from
Published OnlineFirst March 21, 2014; DOI: 10.1158/1535-7163.MCT-13-0775
regulate the epithelial phenotype. Their study also
showed reduced CDH1 expression in tumors after neoad-
juvant chemotherapy (43). Another study showed that
paclitaxel DU-145–resistant cells have greater ZEB1,VIM,
and SNAI1 expression (46).
We tested whether EMT played a causal role in
docetaxel chemoresistance by interfering with the
expression of the transcription factor ZEB1, a key medi-
ator of EMT, in prostate cancer cell lines. We observed
that ZEB1 genetic downmodulation restored CDH1 but
suppressed CD44 expression, which was consistent
with a reversion of EMT and stem-like cell features.
We also observed that ZEB1 inhibition caused prostate
cancer cell mortality independently of docetaxel. This
effect was previously described and is consistent with
the known role of ZEB1 in cell proliferation related,
which is related to the expression of cell cycle inhibitory
cyclin-dependent kinase inhibitors (47). Furthermore,
ZEB1 inhibition restored sensitivity to docetaxel, sup-
porting a mechanistic role of EMT and stem-like cell
phenotype in resistance to therapy. In a previous study
of an adenocarcinoma lung cancer model, inhibition of
ZEB1 significantly enhanced the chemosensitivity of
docetaxel-resistant cells in vitro,andin vivo the ectopic
expression of ZEB1 increased chemoresistance (48).
Several reports have provided evidence that EMT is
critical for invasionand migration and is involved in tumor
recurrence, which is believed to be tightly linked to cancer
stem cells. CD44 and VIM expression in primary tumors
has been correlated with adverse prognosis (34, 49). Nota-
bly, the few patients in our series with ZEB1
þ
/CD44
þ
tumor cells in primary tumors showed extremely aggres-
sive clinical behavior.
In summary, we observed a differential expression of
NF-kB, AR, EMT, and stem-like cell markers between
treated and nontreated tumors. Moreover, they were
related to a higher risk of PSA and/or clinical relapse.
Because the neoadjuvant population may be of higher risk
than the surgical patients, we cannot exclude the possi-
bility that the expression of these markers is more related
to the characteristics of the disease than to the therapy.
However, none of the clinical factors (PSA, Gleason,
clinical stage, or the presence of prior neoadjuvant ther-
apy) correlated with clinical outcome in the univariate or
multivariate analysis in our series.
Overall, our findings support a role of EMT in resistance
to prostate cancer therapy and progression. Our clinical
data were generated in the neoadjuvant setting and can-
not be extrapolated to patients with CRPC. However, both
in vitro and clinical results support the investigation of the
role of EMT in resistance to chemotherapy in CRPC.
Moreover, novel strategies to revert or prevent EMT are
warranted to improve the outcome of CRPC or to increase
the probabilities of cure for patients with high-risk pros-
tate cancer.
Disclosure of Potential Conicts of Interest
M. Donovan is a consultant/advisory board member for Althia. No
potential conflicts of interest were disclosed by the other authors.
Authors' Contributions
Conception and design: M. Marı
´n-Aguilera, J. Codony-Servat, L. Men-
gual, B. Mellado
Development of methodology: M. Marı
´n-Aguilera, J. Codony-Servat,
N. Jim
enez, P. Puig, L. Mengual, M.J. Ribal, B. Mellado
Acquisition of data (provided animals, acquired and managed patients,
provided facilities, etc.): M. Marı
´n-Aguilera, J. Codony-Servat, P.L.
Fern
andez, N. Jim
enez, M. Donovan, R. Bermudo, A. Font, E. Gallardo,
M.J. Ribal, B. Mellado
Analysis and interpretation of data (e.g., statistical analysis, biostatis-
tics, computational analysis): M. Marı
´n-Aguilera, J. Codony-Servat,
O. Reig, J.J. Lozano, P.L. Fern
andez, N. Jim
enez, M. Donovan, L. Mengual,
A. Font, P. Gasc
on, B. Mellado
Writing, review, and/or revision of the manuscript: M. Marı
´n-Aguilera,
J. Codony-Servat,
O. Reig, P.L. Fern
andez, M. Donovan, A. Font, E. Gal-
lardo, P. Gasc
on, B. Mellado
Administrative, technical, or material support (i.e., reporting or orga-
nizing data, constructing databases): M. Marı
´n-Aguilera, J. Codony-
Servat, M.V. Pereira, N. Jim
enez, L. Mengual
Study supervision: M. Marı
´n-Aguilera, J. Codony-Servat, M. Donovan,
A. Alcaraz, B. Mellado
Acknowledgments
The authors thank Instituto de Salud Carlos III and Cellex Foundation
for funding the project. The authors also would like to thank M
onica Marı
´n
and Laura Gelabert for their excellent technical assistance and Elaine Lilly,
for review of the English text.
Grant Support
This work was supported by Cellex Foundation, by the Instituto de
Salud Carlos III—Subdirecci
on General de Evaluaci
on y Fomento de la
Investigaci
on (grants number PI07/0388 and PI12/01226; to B. Mellado),
and by the Ministerio de Economı
´a y competitividad (grant number
SAF2012-40017-C02-02; to P.L. Fern
andez). This study was also cofinanced
by Fondo Europeo de Desarrollo Regional. Uni
on Europea. Una manera de
hacer Europa. M. Marı
´n-Aguilera received a grant from Cellex Foundation.
This work was developed at the Centro Esther Koplowitz, Barcelona,
Spain.
The costs of publication of this article were defrayed in part by the
payment of page charges. This article must therefore be hereby marked
advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate
this fact.
Received September 16, 2013; revised February 13, 2014; accepted March
7, 2014; published OnlineFirst March 21, 2014.
References
1. Siegel R, Naishadham D, Jemal A. Cancer statistics for Hispanics/
Latinos, 2012. CA Cancer J Clin 2012;62:28398.
2. Tannock IF, de WR, Berry WR, Horti J, Pluzanska A, Chi KN,
et al. Docetaxel plus prednisone or mitoxantrone plus prednisone
for advanced prostate cancer. N Engl J Med 2004;351:150212.
3. Ryan CJ, Smith MR, de Bono JS, Molina A, Logothetis CJ, de SP, et al.
Abiraterone in metastatic prostate cancer without previous chemo-
therapy. N Engl J Med 2013;368:13848.
4. Scher HI, Fizazi K, Saad F, Taplin ME, Sternberg CN, Miller K, et al.
Increased survival with enzalutamide in prostate cancer after chemo-
therapy. N Engl J Med 2012;367:118797.
5. de Bono JS, Oudard S, Ozguroglu M, Hansen S, Machiels JP, Kocak
I, et al. Prednisone plus cabazitaxel or mitoxantrone for metastatic
castration-resistant prostate cancer progressing after docetaxel
treatment: a randomised open-label trial. Lancet 2010;376:
114754.
Mol Cancer Ther; 13(5) May 2014 Molecular Cancer Therapeutics
1282
Marín-Aguilera et al.
on May 23, 2014. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from
Published OnlineFirst March 21, 2014; DOI: 10.1158/1535-7163.MCT-13-0775
6. Ploussard G, Terry S, Maille P, Allory Y, Sirab N, Kheuang L, et al. Class
III beta-tubulin expression predicts prostate tumor aggressiveness
and patient response to docetaxel-based chemotherapy. Cancer Res
2010;70:925364.
7. Fojo T, Menefee M. Mechanisms of multidrug resistance: the potential
role of microtubule-stabilizing agents. Ann Oncol 2007;18 Suppl 5:
v3v8.
8. Antonarakis ES, Keizman D, Zhang Z, Gurel B, Lotan TL, Hicks JL, et al.
An immunohistochemical signature comprising PTEN, MYC, and Ki67
predicts progression in prostate cancer patients receiving adjuvant
docetaxel after prostatectomy. Cancer 2012;118:606371.
9. Darshan MS, Loftus MS, Thadani-Mulero M, Levy BP, Escuin D, Zhou
XK, et al. Taxane-induced blockade to nuclear accumulation of the
androgen receptor predicts clinical responses in metastatic prostate
cancer. Cancer Res 2011;71:601929.
10. Guertin DA, Sabatini DM. Dening the role of mTOR in cancer. Cancer
Cell 2007;12:922.
11. Domingo-Domenech J, Oliva C, Rovira A, Codony-Servat J, Bosch M,
Filella X, et al. Interleukin 6, a nuclear factor-kappaB target, predicts
resistance to docetaxel in hormone-independent prostate cancer and
nuclear factor-kappaB inhibition by PS-1145 enhances docetaxel
antitumor activity. Clin Cancer Res 2006;12:557886.
12. Codony-Servat J, Marin-Aguilera M, Visa L, Garcia-Albeniz X, Pineda
E, Fernandez PL, et al. Nuclear factor-kappa B and interleukin-6 related
docetaxel resistance in castration-resistant prostate cancer. Prostate
2013;73:51221.
13. Shen MM, bate-Shen C. Molecular genetics of prostate cancer: new
prospects for old challenges. Genes Dev 2010;24:19672000.
14. Kurrey NK, Jalgaonkar SP, Joglekar AV, Ghanate AD, Chaskar PD,
Doiphode RY, et al. Snail and slug mediate radioresistance and
chemoresistance by antagonizing p53-mediated apoptosis and
acquiring a stem-like phenotype in ovarian cancer cells. Stem Cells
2009;27:205968.
15. Domingo-Domenech J, Vidal SJ, Rodriguez-Bravo V, Castillo-Martin
M, Quinn SA, Rodriguez-Barrueco R, et al. Suppression of acquired
docetaxel resistance in prostate cancer through depletion of notch-
and hedgehog-dependent tumor-initiating cells. Cancer Cell 2012;
22:37388.
16. Marin-Aguilera M, Codony-Servat J, Kalko SG, Fernandez PL, Ber-
mudo R, Buxo E, et al. Identication of docetaxel resistance genes in
castration-resistant prostate cancer. Mol Cancer Ther 2012;11:
32939.
17. Mellado B, Font A, Alcaraz A, Aparicio LA, Veiga FJ, Areal J, et al.
Phase II trial of short-term neoadjuvant docetaxel and complete
androgen blockade in high-risk prostate cancer. Br J Cancer
2009;101:124852.
18. Seruga B, Ocana A, Tannock IF. Drug resistance in metastatic cas-
tration-resistant prostate cancer. Nat Rev Clin Oncol 2011;8:1223.
19. Boccon-Gibod L, Djavan WB, Hammerer P, Hoeltl W, Kattan MW,
Prayer-Galetti T, et al. Management of prostate-specic antigen
relapse in prostate cancer: a European Consensus. Int J Clin Pract
2004;58:38290.
20. Scher HI, Halabi S, Tannock I, Morris M, Sternberg CN, Carducci MA,
et al. Design and end points of clinical trials for patients with progres-
sive prostate cancer and castrate levels of testosterone: recommen-
dations of the Prostate Cancer Clinical Trials Working Group. J Clin
Oncol 2008;26:114859.
21. Huang dW, Sherman BT, Lempicki RA. Systematic and integrative
analysis of large gene lists using DAVID bioinformatics resources. Nat
Protoc 2009;4:4457.
22. Domingo-Domenech J, Mellado B, Ferrer B, Truan D, Codony-Servat
J, Sauleda S, et al. Activation of nuclear factor-kappaB in human
prostate carcinogenesis and association to biochemical relapse. Br J
Cancer 2005;93:128594.
23. Sun J, Zhang D, Bae DH, Sahni S, Jansson P, Zheng Y, et al. Metastasis
suppressor, NDRG1, mediates its activity through signaling pathways
and molecular motors. Carcinogenesis 2013;34:194354.
24. Yae T, Tsuchihashi K, Ishimoto T, Motohara T, Yoshikawa M, Yoshida
GJ, et al. Alternative splicing of CD44 mRNA by ESRP1 enhances lung
colonization of metastatic cancer cell. Nat Commun 2012;3:883.
25. Gemmill RM, Roche J, Potiron VA, Nasarre P, Mitas M, Coldren CD,
et al. ZEB1-responsive genes in nonsmall cell lung cancer. Cancer
Lett 2011;300:6678.
26. Sheehan GM, Kallakury BV, Sheehan CE, Fisher HA, Kaufman RP Jr,
Ross JS. Loss of claudins-1 and -7 and expression of claudins-3 and -4
correlate with prognostic variables in prostatic adenocarcinomas.
Hum Pathol 2007;38:5649.
27. Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, et al. The
epithelialmesenchymal transition generates cells with properties of
stem cells. Cell 2008;133:70415.
28. Mostaghel EA, Page ST, Lin DW, Fazli L, Coleman IM, True LD,
et al. Intraprostatic androgens and androgen-regulated gene
expression persist after testosterone suppression: therapeutic
implications for castration-resistant prostate cancer. Cancer Res
2007;67:503341.
29. Ryan CJ, Smith A, Lal P, Satagopan J, Reuter V, Scardino P, et al.
Persistent prostate-specic antigen expression after neoadjuvant
androgen depletion: an early predictor of relapse or incomplete andro-
gen suppression. Urology 2006;68:8349.
30. Zhu ML, Horbinski CM, Garzotto M, Qian DZ, Beer TM, Kyprianou N.
Tubulin-targeting chemotherapy impairs androgen receptor activity in
prostate cancer. Cancer Res 2010;70:79928002.
31. Gowda PS, Deng JD, Mishra S, Bandyopadhyay A, Liang S, Lin S, et al.
Inhibition of hedgehog and androgen receptor signaling pathways
produced synergistic suppression of castration-resistant prostate
cancer progression. Mol Cancer Res 2013;11:144861.
32. Febbo PG, Richie JP, George DJ, Loda M, Manola J, Shankar S,
et al. Neoadjuvant docetaxel before radical prostatectomy in
patients with high-risk localized prostate cancer. Clin Cancer Res
2005;11:523340.
33. Huang CY, Beer TM, Higano CS, True LD, Vessella R, Lange PH, et al.
Molecular alterations in prostate carcinomas that associate with in vivo
exposure to chemotherapy: identication of a cytoprotective mecha-
nism involving growth differentiation factor 15. Clin Cancer Res
2007;13:582533.
34. Zhang Q, Helfand BT, Jang TL, Zhu LJ, Chen L, Yang XJ, et al. Nucle ar
factor-kappaB-mediated transforming growth factor-beta-induced
expression of vimentin is an independent predictor of biochemical
recurrence after radical prostatectomy. Clin Cancer Res 2009;15:
355767.
35. de Bruin EC, van de PS, Lips EH, van ER, van der Zee MM, Lombaerts
M, et al. Macrodissection versus microdissection of rectal carcinoma:
minor inuence of stroma cells to tumor cell gene expression proles.
BMC Genomics 2005;6:142.
36. Nauseef JT, Henry MD. Epithelial-to-mesenchymal transition in pros-
tate cancer: paradigm or puzzle? Nat Rev Urol 2011;8:42839.
37. Jennbacken K, Tesan T, Wang W, Gustavsson H, Damber JE, Welen K.
N-cadherin increases after androgen deprivation and is associated
with metastasis in prostate cancer. Endocr Relat Cancer 2010;17:
46979.
38. Zhu ML, Kyprianou N. Role of androgens and the androgen receptor in
epithelialmesenchymal transition and invasion of prostate cancer
cells. FASEB J 2010;24:76977.
39. Matuszak EA, Kyprianou N. Androgen regulation of epithelialmesen-
chymal transition in prostate tumorigenesis. Expert Rev Endocrinol
Metab 2011;6:46982.
40. Anose BM, Sanders MM. Androgen receptor regulates transcription of
the ZEB1 transcription factor. Int J Endocrinol 2011;2011:903918.
41. Sun Y, Wang BE, Leong KG, Yue P, Li L, Jhunjhunwala S, et al.
Androgen deprivation causes epithelialmesenchymal transition in the
prostate: implications for androgen-deprivation therapy. Cancer Res
2012;72:52736.
42. Cottard F, Asmane I, Erdmann E, Bergerat JP, Kurtz JE, Ceraline J.
Constitutively active androgen receptor variants upregulate expres-
sion of mesenchymal markers in prostate cancer cells. PLoS ONE
2013;8:e63466.
43. Puhr M, Hoefer J, Schafer G, Erb HH, Oh SJ, Klocker H, et al. Epithelial-
to-mesenchymal transition leads to docetaxel resistance in prostate
cancer and is mediated by reduced expression of miR-200c and miR-
205. Am J Pathol 2012;181:2188201.
www.aacrjournals.org Mol Cancer Ther; 13(5) May 2014 1283
EMT Role in Docetaxel Resistance
on May 23, 2014. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from
Published OnlineFirst March 21, 2014; DOI: 10.1158/1535-7163.MCT-13-0775
44. Li M, Knight DA, Smyth MJ, Stewart TJ. Sensitivity of a novel model of
mammary cancer stem cell-like cells to TNF-related death pathways.
Cancer Immunol Immunother 2012;61:125568.
45. Hao J, Madigan MC, Khatri A, Power CA, Hung TT, Beretov J, et al. In
vitro and in vivo prostate cancer metastasis and chemoresistance can
be modulated by expression of either CD44 or CD147. PLoS ONE
2012;7:e40716.
46. Kim JJ, Yin B, Christudass CS, Terada N, Rajagopalan K, Fabry B, et al.
Acquisition of paclitaxel resistance is associated with a more aggres-
sive and invasive phenotype in prostate cancer. J Cell Biochem 2013;
114:128693.
47. Liu Y, Sanchez-Tillo E, Lu X, Huang L, Clem B, Telang S, et al. The ZEB1
transcription factor acts in a negative feedback loop with miR200
downstream of Ras and Rb1 to regulate Bmi1 expression. J Biol Chem
2014;289:411625.
48. Ren J, Chen Y, Song H, Chen L, Wang R. Inhibition of ZEB1 reverses
EMT and chemoresistance in docetaxel-resistant human lung adeno-
carcinoma cell line. J Cell Biochem 2013;114:1395403.
49. Patrawala L, Calhoun T, Schneider-Broussard R, Li H, Bhatia B, Tang
S, et al. Highly puried CD44
þ
prostate cancer cells from xenograft
human tumors are enriched in tumorigenic and metastatic progenitor
cells. Oncogene 2006;25:1696708.
Mol Cancer Ther; 13(5) May 2014 Molecular Cancer Therapeutics
1284
Marín-Aguilera et al.
on May 23, 2014. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from
Published OnlineFirst March 21, 2014; DOI: 10.1158/1535-7163.MCT-13-0775
... However, the involvement of Gln metabolism in docetaxel-resistant CRPC cells with mesenchymal phenotype has not yet been investigated. Cancer cells with mesenchymal phenotypes have been linked to therapy resistance and stem cell features in multiple cancers, including PCa [9,[26][27][28][29]. Ippolito and colleagues demonstrated that docetaxel-resistant PC3 and DU145 cells shift their metabolism to OXPHOS and that the metabolic adaptation is related to the mesenchymal cell phenotype, which has been associated with increased features of the Gln metabolism [14,30]. ...
Article
Full-text available
Docetaxel (DX) serves as a palliative treatment option for metastatic prostate cancer (PCa). Despite initial remission, acquired DX resistance is inevitable. The mechanisms behind DX resistance have not yet been deciphered, but a mesenchymal phenotype is associated with DX resistance. Mesenchymal phenotypes have been linked to metabolic rewiring, obtaining most ATP production by oxidative phosphorylation (OXPHOS) powered substantially by glutamine (Gln). Likewise, Gln is known to play an essential role in modulating bioenergetic, redox homeostasis and autophagy. Herein, investigations of Gln deprivation on DX-sensitive and -resistant (DR) PCa cells revealed that the DR cell sub-lines were susceptible to Gln deprivation. Mechanistically, Gln deprivation reduced OXPHOS and ATP levels, causing a disturbance in cell cycle progression. Genetic and chemical inhibition of the Gln-metabolism key protein GLS1 could validate the Gln deprivation results, thereby representing a valid therapeutic target. Moreover, immunohistological investigation of GLS1 revealed a high-expressing GLS1 subgroup post-docetaxel failure, exhibiting low overall survival. This subgroup presents an intriguing opportunity for targeted therapy focusing on glutamine metabolism. Thus, these findings highlight a possible clinical rationale for the chemical inhibition of GLS1 as a therapeutic strategy to target mesenchymal DR PCa cells, thereby delaying accelerated tumour progression.
... In order to mediate EMT in PCa cells and impart enzalutamide resistance, ISL1 stimulates Akt phosphorylation [124]. According to intriguing research, activating the EMT process may cause PCa to relapse and acquire docetaxel resistance [125]. Castration resistance in PCa may also be caused by overexpression of the oncogenic FOX2 and development of EMT [126]. ...
Article
Full-text available
Epithelial-mesenchymal transition (EMT) is a complicated molecular process that governs cellular shape and function changes throughout tissue development and embryogenesis. In addition, EMT contributes to the development and spread of tumors. Expanding and degrading the surrounding microenvironment, cells undergoing EMT move away from the main location. On the basis of the expression of fibroblast-specific protein-1 (FSP1), fibroblast growth factor (FGF), collagen, and smooth muscle actin (-SMA), the mesenchymal phenotype exhibited in fibroblasts is crucial for promoting EMT. While EMT is not entirely reliant on its regulators like ZEB1/2, Twist, and Snail proteins, investigation of upstream signaling (like EGF, TGF-β, Wnt) is required to get a more thorough understanding of tumor EMT. Throughout numerous cancers, connections between tumor epithelial and fibroblast cells that influence tumor growth have been found. The significance of cellular crosstalk stems from the fact that these events affect therapeutic response and disease prognosis. This study examines how classical EMT signals emanating from various cancer cells interfere to tumor metastasis, treatment resistance, and tumor recurrence.
... In addition to CTC enumeration, deeper molecular interrogation to further appreciate the phenotypic CTC heterogeneity offers another promising tool for prognostication. To date, most studies have focused on tumor cell heterogeneity and the impact of detecting specific markers on clinical behavior, therapy resistance and survival of patients with PCa [26,27]. However, in the past decade, emerging technologies for CTC isolation and characterization have permitted research on the biology of CTCs. ...
Article
Full-text available
Rational: Circulating tumor cells (CTCs) appear to be a promising tool for predicting the clinical outcome and monitoring the response to treatment in patients with solid tumors. The current study assessed the clinical relevance of monitoring CTCs in patients with metastatic castration resistant prostate cancer (mCRPC) treated with cabazitaxel. Patients and methods: Patients with histologically confirmed mCRPC who were previously treated with a docetaxel-containing regimen and experienced disease progression were enrolled in this multicenter prospective study. CTC counts were enumerated using the CellSearch system at baseline (before cabazitaxel initiation), after one cabazitaxel cycle (post 1st cycle) and at disease progression (PD). Patients were stratified into predetermined CTC-positive and CTC-negative groups. The phenotypic characterization was performed using double immunofluorescence staining with anti-CKs and anti-Ki67, anti-M30 or anti-vimentin antibodies. Results: The median PFS and OS were 4.0 (range, 1.0-17.9) and 14.5 (range, 1.2-33.9) months, respectively. At baseline, 48 out of 57 (84.2%) patients had ≥1 CTCs/7.5 mL of peripheral blood (PB) and 37 (64.9%) had ≥5 CTCs/7.5 mL of PB. After one treatment cycle, 30 (75%) out of the 40 patients with available measurements had ≥1 detectable CTC/7.5 mL of PB and 24 (60%) ≥ 5CTCs/7.5 mL of PB; 12.5% of the patients with detectable CTCs at the baseline sample had no detectable CTCs after one treatment cycle. The detection of ≥5CTCs/7.5 mL of PB at baseline and post-cycle 1 was associated with shorter PFS and OS (p = 0.002), whereas a positive CTC status post-cycle 1 strongly correlated with poorer OS irrespective of the CTC cut-off used. Multivariate analysis revealed that the detection of non-apoptotic (CK+/M30-) CTCs at baseline is an independent predictor of shorter OS (p = 0.005). Conclusions: In patients with mCRPC treated with cabazitaxel, CTC counts both at baseline and after the first cycle retain their prognostic significance, implying that liquid biopsy monitoring might serve as a valuable tool for predicting treatment efficacy and survival outcomes.
Article
Unlabelled: Currently, clinical biomarkers are urgently needed to improve patient management to guide personal therapy for cancer. In this study, we investigate the deregulation of Zeb-1 in prostate cancer (PC) Tunisian patients. Expression patterns of the Zeb-1 were investigated in prostate adenocarcinoma and benign prostate biopsies using quantitative real-time reverse transcription-polymerase chain reaction (RT-qPCR) and 2-ΔΔCt method. Statistical analysis was used to identify differences across groups depending on gene expression level. Furthermore, we exploited a follow-up over 15 years to correlate Zeb-1 deregulation and clinical outcomes in PC patients. Based on ROC curve analyses, the AUC was found in discriminating PC patients from controls (AUC = 0.757; p < 0.001). In addition, the higher expression level was significantly associated with PSA, Digital Rectal Examination, Gleason score, tumor stage, and distant lymph node metastases. Moreover, Zeb-1 overexpression was correlated with shorter overall survival (OS) (p = 0.042), poor progression-free survival (PFS) (p = 0.007), and with resistance to taxanes (p = 0.012). Our data provide the aberrant expression of Zeb-1 in PC patients suggesting its potential diagnostic, prognostic, and theranostic role. Further functional studies are mandatory to strengthen these results and to uncover the molecular mechanism of this neoplasm. Supplementary information: The online version contains supplementary material available at 10.1007/s13205-024-03941-8.
Article
Docetaxel has been the standard first-line chemotherapy for lethal metastatic prostate cancer (mPCa) since 2004, but resistance to docetaxel treatment is common. The molecular mechanisms of docetaxel resistance remain largely unknown and could be amenable to interventions that mitigate resistance. We have recently discovered that several docetaxel-resistant mPCa cell lines exhibit lower uptake of cellular copper and uniquely express higher levels of a copper exporter protein ATP7B. Knock-down of ATP7B by silencing RNAs (siRNAs) sensitized docetaxel resistant-mPCa cells to the growth inhibitory and apoptotic effects of docetaxel. Importantly, deletions of ATP7B in human mPCa tissues predict significantly better survival of patients after their first chemotherapy than those with wild-type ATP7B (P = 0.0006). In addition, disulfiram (DSF), an FDA-approved drug for the treatment of alcohol dependence, in combination with copper, significantly enhanced the in vivo antitumor effects of docetaxel in a docetaxel-resistant xenograft tumor model. Our analyses also revealed that DSF and copper engaged with ATP7B to decrease protein levels of COMM domain-containing protein 1 (COMMD1), S-phase kinase-associated protein 2 (Skp2), and clusterin and markedly increase protein expression of cyclin-dependent kinase inhibitor 1 (p21/WAF1). Taken together, our results indicate a copper-dependent nutrient vulnerability through ATP7B exporter in docetaxel-resistant PCa for improving the therapeutic efficacy of docetaxel.
Chapter
In this chapter entitled “Cancer Metastasis, ROS/Redox Signaling, and PCD Resistance/Redox Metabolism,” initially metastasis link to cell adhesion marker kinetics and resistance has been discussed during cancer development. Metastasis development steps have been discussed with the help of EMT/MET changes. The role of the TME has been discussed in the contribution of stroma cells to EMT. The role of matrix metalloproteinases (MMPs) in cell proliferation, survival, immune response, and angiogenesis in addition to invasion has been discussed. Dormancy and reactivation phase have been taken up at the molecular level, and the involvement of CSCs to metastasis has also been discussed. Redox regulation in metastasis development has been discussed with the involvement and regulation of specially HIF-1, TGF-b, NFkB factors. Metastatic cell survival and PCD resistance have been addressed in cancer resistance development. Redox metabolic characteristics of the cancer cell link to metastasis resistance have been discussed.
Article
Epithelial-mesenchymal transition (EMT) is a cellular process by which epithelial cells lose their characteristics and acquire mesenchymal traits to promote cell movement. This program is aberrantly activated in human cancers and endows tumor cells with increased abilities in tumor initiation, cell migration, invasion, metastasis, and therapy resistance. The EMT program in tumors is rarely binary and often leads to a series of gradual or intermediate epithelial-mesenchymal states. Functionally, epithelial-mesenchymal plasticity (EMP) improves the fitness of cancer cells during tumor progression and in response to therapies. Here, we discuss the most recent advances in our understanding of the diverse roles of EMP in tumor initiation, progression, metastasis, and therapy resistance and address major clinical challenges due to EMP-driven phenotypic heterogeneity in cancer. Uncovering novel molecular markers and key regulators of EMP in cancer will aid the development of new therapeutic strategies to prevent cancer recurrence and overcome therapy resistance. Expected final online publication date for the Annual Review of Pathology: Mechanisms of Disease, Volume 19 is January 2024. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Article
Full-text available
DAVID bioinformatics resources consists of an integrated biological knowledgebase and analytic tools aimed at systematically extracting biological meaning from large gene/protein lists. This protocol explains how to use DAVID, a high-throughput and integrated data-mining environment, to analyze gene lists derived from high-throughput genomic experiments. The procedure first requires uploading a gene list containing any number of common gene identifiers followed by analysis using one or more text and pathway-mining tools such as gene functional classification, functional annotation chart or clustering and functional annotation table. By following this protocol, investigators are able to gain an in-depth understanding of the biological themes in lists of genes that are enriched in genome-scale studies.
Article
Full-text available
Ras mutations are frequent in cancer cells where they drive proliferation and resistance to apoptosis. However in primary cells, mutant Ras instead can cause oncogene-induced senescence, a tumor suppressor function linked to repression of the polycomb factor Bmi1, which normally regulates cell cycle inhibitory cyclin dependent kinase inhibitors (cdki). It is unclear how Ras causes repression of Bmi1 in primary cells to suppress tumor formation while inducing the gene in cancer cells to drive tumor progression. Ras also induces the EMT transcription factor ZEB1 to trigger tumor invasion and metastasis. Beyond its well-documented role in EMT, ZEB1 is important for maintaining repression of cdki. Indeed, heterozygous mutation of ZEB1 is sufficient for elevated cdki expression, leading to premature senescence of primary cells. A similar phenotype is evident with Bmi1 mutation. We show that activation of Rb1 in response to mutant Ras causes dominant repression of ZEB1 in primary cells, but loss of the Rb1 pathway is a hallmark of cancer cells and in the absence of such Rb1 repression Ras induces ZEB1 in cancer cells. ZEB1 represses miR-200 in the context of a mutual repression loop. Because miR-200 represses Bmi1, induction of ZEB1 leads to induction of Bmi1. Rb1 pathway status then dictates the opposing effects of mutant Ras on the ZEB1-miR-200 loop in primary vs. cancer cells. This loop not only triggers EMT, surprisingly we show it acts downstream of Ras to regulate Bmi1 expression and thus the critical decision between oncogene-induced senescence and tumor initiation.
Article
Full-text available
The metastasis suppressor, N-myc downstream regulated gene 1 (NDRG1), is negatively correlated with tumor progression in multiple neoplasms, being a promising new target for cancer treatment. However, the precise molecular effects of NDRG1 remain unclear. Herein, we summarize recent advances in understanding the impact of NDRG1 on cancer metastasis with emphasis on its interactions with the key oncogenic nuclear factor-kappaB, phosphatidylinositol-3 kinase/phosphorylated AKT/mammalian target of rapamycin and Ras/Raf/mitogen-activated protein kinase kinase/extracellular signal-regulated kinase signaling pathways. Recent studies demonstrating the inhibitory effects of NDRG1 on the epithelial–mesenchymal transition, a key initial step in metastasis, TGF-β pathway and the Wnt/β-catenin pathway are also described. Furthermore, NDRG1 was also demonstrated to regulate molecular motors in cancer cells, leading to inhibition of F-actin polymerization, stress fiber formation and subsequent reduction of cancer cell migration. Collectively, this review summarizes the underlying molecular mechanisms of the antimetastatic effects of NDRG1 in cancer cells.
Article
Full-text available
Androgen receptor (AR) signaling pathway remains the main target of novel therapeutics for castration-resistant prostate cancer (CRPC). However, constitutively active AR variants lacking the carboxy-terminal region in CRPC lead to therapy inefficacy. Moreover, recent studies suggest that AR variants are expressed in primary prostate tumors and may contribute to tumor progression. The aim of this study was to investigate the impact of AR variants on tumor progression. N-cadherin expression was analyzed in LNCaP cells overexpressing the wild type AR or a constitutively active AR variant by qRT-PCR, Western blot and immunofluorescence. We showed here for the first time that N-cadherin expression was increased in the presence of constitutively active AR variants. Moreover, this increased expression of N-cadherin in LNCaP overexpressing AR variants was negatively regulated by androgens. Although N-cadherin expression is often associated with a downregulation of E-cadherin, this phenomenon was not observed in our model. Nevertheless, in addition to the increased expression of N-cadherin, an upregulation of other mesenchymal markers expression such as Vimentin, SNAIL and ZEB1 was observed in the presence of constitutively active AR variants. In conclusion, our findings highlight novel consequences of constitutively active AR variants on the regulation of mesenchymal markers in prostate cancer. Citation Format: Félicie COTTARD, Irène ASMANE, Eva ERDMANN, Jean-Pierre BERGERAT, Jean-Emmanuel KURTZ, Jocelyn CERALINE. Constitutively active androgen receptor variants upregulate expression of mesenchymal markers in prostate cancer cells. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 2117. doi:10.1158/1538-7445.AM2014-2117
Article
Full-text available
Background: Abiraterone acetate, an androgen biosynthesis inhibitor, improves overall survival in patients with metastatic castration-resistant prostate cancer after chemotherapy. We evaluated this agent in patients who had not received previous chemotherapy. Methods: In this double-blind study, we randomly assigned 1088 patients to receive abiraterone acetate (1000 mg) plus prednisone (5 mg twice daily) or placebo plus prednisone. The coprimary end points were radiographic progression-free survival and overall survival. Results: The study was unblinded after a planned interim analysis that was performed after 43% of the expected deaths had occurred. The median radiographic progression-free survival was 16.5 months with abiraterone-prednisone and 8.3 months with prednisone alone (hazard ratio for abiraterone-prednisone vs. prednisone alone, 0.53; 95% confidence interval [CI], 0.45 to 0.62; P<0.001). Over a median follow-up period of 22.2 months, overall survival was improved with abiraterone-prednisone (median not reached, vs. 27.2 months for prednisone alone; hazard ratio, 0.75; 95% CI, 0.61 to 0.93; P=0.01) but did not cross the efficacy boundary. Abiraterone-prednisone showed superiority over prednisone alone with respect to time to initiation of cytotoxic chemotherapy, opiate use for cancer-related pain, prostate-specific antigen progression, and decline in performance status. Grade 3 or 4 mineralocorticoid-related adverse events and abnormalities on liver-function testing were more common with abiraterone-prednisone. Conclusions: Abiraterone improved radiographic progression-free survival, showed a trend toward improved overall survival, and significantly delayed clinical decline and initiation of chemotherapy in patients with metastatic castration-resistant prostate cancer. (Funded by Janssen Research and Development, formerly Cougar Biotechnology; ClinicalTrials.gov number, NCT00887198.).
Article
The molecular determinants of carcinogenesis, tumor progression and patient prognosis can be deduced from simultaneous comparison of thousands of genes by microarray analysis. However, the presence of stroma cells in surgically excised carcinoma tissues might obscure the tumor cell-specific gene expression profiles of these samples. To circumvent this complication, laser microdissection can be performed to separate tumor epithelium from the surrounding stroma and healthy tissue. In this report, we compared RNAs isolated from macrodissected, of which only surrounding healthy tissue had been removed, and microdissected rectal carcinoma samples by microarray analysis in order to determine the most reliable approach to detect the expression of tumor cell-derived genes by microarray analysis.
Article
Unlabelled: Metastatic prostate cancer is initially treated with androgen ablation therapy, which causes regression of androgen-dependent tumors. However, these tumors eventually relapse resulting in recurrent castration-resistant prostate cancer (CRPC). Currently, there is no effective therapy for CRPC and the molecular mechanisms that lead to the development of CRPC are not well understood. Here, we evaluated the hypothesis that combined inhibition of Hedgehog (Hh) and androgen receptor (AR) signaling will synergistically attenuate the growth of CRPC in vitro and in vivo. Androgen deprivation induced full-length androgen receptor protein levels in CRPC cells, but decreased its nuclear localization and transcriptional activity. However, androgen deprivation also increased a truncated form of androgen receptor (lacking ligand-binding domain) that possessed transcriptional activity in CRPC cells. Androgen deprivation also promoted the expression of Hh signaling components in CRPC cells, xenograft tumors, and the prostate glands of castrated mice. Importantly, although inhibition of either Hh or androgen receptor signaling alone was only moderately effective in blocking CRPC cell growth, combination of an Hh pathway inhibitor and a noncompetitive androgen receptor inhibitor synergistically suppressed the growth of CRPC cells in vitro and in vivo. Finally, noncompetitive inhibition of androgen receptor, but not competitive inhibition, was effective at limiting the activity of truncated androgen receptor leading to the inhibition of CRPC. Implications: Combined therapy using Hh inhibitors and a non-competitive AR inhibitor may limit CRPC growth.
Article
Docetaxel has been used as one of the first-line chemotherapies in solid tumors including advanced non-small cell lung cancer (NSCLC). However, limited responses to chemotherapy are observed in clinic and the molecular mechanisms have not been fully understood. Emerging evidence suggests that epithelial-mesenchymal transition (EMT) plays an important role in the processes of tumor metastasis as well as resistance towards anticancer agents. In this study, it was observed that docetaxel-resistant human lung adenocarcinoma cell line (SPC-A1/DTX) was typical of mesenchymal phenotype. SPC-A1/DTX cell line has increased migratory and invasive capacity both in vitro and in vivo. Among the master EMT-inducing transcriptional factors, ZEB1 was found to be significantly increased in SPC-A1/DTX cell line. ZEB1 knockdown with RNA interference could reverse the EMT phenotype and inhibit the migratory ability of SPC-A1/DTX cells. Furthermore, inhibition of ZEB1 significantly enhanced the chemosensitivity of SPC-A1/DTX cells to docetaxel in vitro and in vivo and ectopic expression of ZEB1 increased the chemoresistance of SPC-A1 cells to docetaxel. All these results provide experimental evidence that ZEB1 might be an attractive target for the treatment of human NSCLC. J. Cell. Biochem. © 2012 Wiley Periodicals, Inc.