ArticlePDF Available

Abstract and Figures

One of the major obstacles in the treatment of hormone-refractory prostate cancer (HRPC) is the development of chemoresistant tumors. The aim of this study is to evaluate the role of azacitidine as chemosensitizing agent in association with docetaxel (DTX) and cisplatin using two models of aggressive prostate cancer, the 22rv1, and PC3 cell lines. Azacitidine shows antiproliferative effects associated with increased proportion of cells in G0/G1 and evident apoptosis in 22rv1 cells and increased proportion of cells in G2/M phase with the absence of acute cell killing in PC3 cells. In vivo, azacitidine (0.8 mg/kg i.p.) reduced tumor proliferation and induced apoptosis in both xenografts upmodulating the expression of p16INKA, Bax, Bak, p21/WAF1, and p27/KIP1, and inhibiting the activation of Akt activity and the expression of cyclin D1, Bcl-2, and Bcl-XL. In vitro treatments with azacitidine lead to upregulation of cleaved caspase 3 and PARP. BCl2 antagonists, such as HA-14-1, enhanced the effects of azacitidine in these two prostate cancer models. In addition, azacitidine showed synergistic effects with both DTX and cisplatin. In vivo this agent caused tumor growth delay without complete regression in xenograft systems. Azacitidine sensitized PC3 and 22rv1 xenografts to DTX and cisplatin treatments. These combinations were also tolerable in mice and superior to either agent alone. As DTX is the standard first-line chemotherapy for HRPC, the development of DTX-based combination therapies is of great interest in this disease stage. Our results provide a rationale for clinical trials on combination treatments with azacitidine in patients with hormone-refractory and chemoresistant prostate tumors.
Content may be subject to copyright.
Azacitidine improves antitumor effects
of docetaxel and cisplatin in
aggressive prostate cancer models
Claudio Festuccia*, Giovanni Luca Gravina
1
*, Anna Maria D’Alessandro
2
,
Paola Muzi, Danilo Millimaggi
2
, Vincenza Dolo
2
, Enrico Ricevuto
3
,
Carlo Vicentini
2
and Mauro Bologna
4
Department of Experimental Medicine, Chair of General Pathology, University of L’Aquila, Via Vetoio, Coppito-2,
67100 L’Aquila, Italy,
1
Division of Radiotherapy, Department of Experimental Medicine,
2
Department of Health Science,
3
Division of Clinical Oncology,
Department of Experimental Medicine and
4
Department of Basic and Applied Biology, University of L’Aquila, L’Aquila, Italy
(Correspondence should be addressed to C Festuccia; Email: festucci@univaq.it)
*(C Festuccia and G L Gravina contributed equally to this work)
Abstract
One of the major obstacles in the treatment of hormone-refractory prostate cancer (HRPC) is the
development of chemoresistant tumors. The aim of this study is to evaluate the role of azacitidine
as chemosensitizing agent in association with docetaxel (DTX) and cisplatin using two models of
aggressive prostate cancer, the 22rv1, and PC3 cell lines. Azacitidine shows antiproliferative
effects associated with increased proportion of cells in G0/G1 and evident apoptosis in 22rv1 cells
and increased proportion of cells in G2/M phase with the absence of acute cell killing in PC3 cells.
In vivo, azacitidine (0.8 mg/kg i.p.) reduced tumor proliferation and induced apoptosis in both
xenografts upmodulating the expression of p16INKA, Bax, Bak, p21/WAF1, and p27/KIP1, and
inhibiting the activation of Akt activity and the expression of cyclin D1, Bcl-2, and Bcl-XL. In vitro
treatments with azacitidine lead to upregulation of cleaved caspase 3 and PARP. BCl2
antagonists, such as HA-14-1, enhanced the effects of azacitidine in these two prostate cancer
models. In addition, azacitidine showed synergistic effects with both DTX and cisplatin. In vivo this
agent caused tumor growth delay without complete regression in xenograft systems. Azacitidine
sensitized PC3 and 22rv1 xenografts to DTX and cisplatin treatments. These combinations were
also tolerable in mice and superior to either agent alone. As DTX is the standard first-line
chemotherapy for HRPC, the development of DTX-based combination therapies is of great interest
in this disease stage. Our results provide a rationale for clinical trials on combination treatments with
azacitidine in patients with hormone-refractory and chemoresistant prostate tumors.
Endocrine-Related Cancer (2009) 16 401–413
Introduction
Prostate cancer represents a global public health
problem. Worldwide, it is the second most common
noncutaneous cancer in men, accounting for w10% of
male cancers (Haas et al. 2008). In recent years, 5 year
survival rates for prostate cancer have been ranked
the third highest of all cancers (Desireddi et al. 2007,
Svatek et al. 2008). Although chemotherapy histori-
cally has had limited utility in treating advanced
prostate cancers, the results from two recent
randomized clinical trials indicated that docetaxel
(DTX)-based chemotherapy improved survival in
patients with hormone-refractory prostate cancer,
HRPC (Patel et al. 2005,Armstrong et al. 2007).
Docetaxel and mitoxantrone are two food and drug
administration – approved chemotherapeutic agents for
clinical treatment of advanced HRPC and DTX is the
standard first-line chemotherapy in this disease stage.
Endocrine-Related Cancer (2009) 16 401–413
Endocrine-Related Cancer (2009) 16 401–413
1351–0088/09/016–401 q2009 Society for Endocrinology Printed in Great Britain
DOI: 10.1677/ERC-08-0130
Online version via http://www.endocrinology-journals.org
Thus, the development of DTX-based combination
therapies is of great interest for HRPC. Some clinical
studies supported taxane-based therapy, a promising
platform to develop new combinational schedule,
which remains a high priority for many ongoing
studies (Patel et al. 2005,Armstrong et al. 2007 and for
a review, see Calabro
`and Stemberg 2007). In addition
a recent report shows that single-agent satraplatin
improves progression-free survival (PFS) as second-
line chemotherapy for patients with HRPC and
provides further evidence that platinum salts may
have a role in the treatment of HRPC (Nakabayashi
et al.2008,Ross et al. 2008). Nevertheless, improvement
in the efficacy of chemotherapy is urgently needed for
patients with chemotherapy-resistant HRPC.
It is now well established that cancer cells exhibit a
number of genetic and epigenetic defects in the
machinery that governs programmed cell death and
that sabotage of apoptosis is one of the principal factors
aiding in the progression and treatment of neoplastic
diseases. Epigenetic modifications, specifically DNA
hypermethylation, are believed to play an important
role in the downregulation of genes important for
protection against apoptosis. In addition to classical
genetic abnormalities, epigenetic modifications have
emerged as a central driving force in the molecular
pathology of prostate cancer (McCabe et al. 2006,
Murphy et al.2008). Several years ago, Plumb et al.
(2000) showed that resistance to carboplatin in ovarian
cancer cells was mediated by hypermethylation and
loss of function of the MLH1 mismatch repair gene.
Of interest, decitabine, a deoxyderivative of aza-CR, a
cytosine analogue with hypomethylating properties,
reverses the downregulation of expression of some
membrane transporters in vitro (Plumb et al. 2000),
raising the untested possibility that repeated treatment
could lead to a progressive increase in efficacy.
In this study, we used azacitidine and its pharma-
cological formulation (Vidaza) to verify if this agent
was able as single agent to alter tumor cell proliferation
and apoptosis, as well as to improve the sensitivity
against DTX and cisplatin in vitro and in vivo using
two models of aggressive prostate cancer. 22Rv1 is an
androgen-responsive human prostate carcinoma cell
line derived from a primary prostate tumor that
expresses mutant (H874Y) androgen receptors (AR),
whereas PC3 is an androgen irresponsive, AR negative
prostate carcinoma cell line. Our results indicate that
azacitidine has antiproliferative effects in both cell
lines and showed pro-apoptotic effects in 22rv1 cells.
In vitro showed synergistic effects with DTX and
cisplatin. Its pharmacological preparation (Vidaza)
caused tumor growth delay without complete stasis or
regression as single agent, whereas improved anti-
tumor effects of DTX and cisplatin resulting these
combinations well tolerable in mice. Therefore, use of
DNA methylation inhibitors might be a suitable
therapeutic tool for the amplification of pharmacologic
responses of HRPC versus first- and second-line
chemotherapies.
Materials and methods
Reagents
All the materials for tissue culture were purchased
from Hyclone (Cramlington, NE, USA). Antibodies
when not otherwise specified were purchased from
Santa Cruz (Santa Cruz, CA, USA). Plasticware was
obtained from Nunc (Roskilde, Denmark). Azacitidine
(Vidaza) was obtained in collaboration with Celgene
Corporation (Summit, NJ, USA). DNA methyltransfer-
ase activity was evaluated in nuclear cell extracts by a
colorimetric EpiQuik DNA methyltransferase Activity
Assay Kit (BioVision, Mountain View, CA, USA).
Cell lines
The two aggressive prostate cancer models (22rv1 and
PC3 cell lines) were obtained from American Tissue
Culture Collection (ATCC, Rockville, MD, USA) and
grown as recommended.
Cell proliferation inhibition assay
Cell proliferation was evaluated by 3-(4,
5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bro-
mide (MTT; Sigma) assay. Briefly, cells were seeded
in 96-well tissue culture plates (Costar, Corning, NY,
USA) at 3000 cells per well per 150 ml. At 24 h after
seeding, cells were treated with 50 ml serial concen-
trations of azacitidine, DTX, cisplatin or the combina-
tion of azacitidine plus DTX and cisplatin. After 72 h
of drug exposure, MTT (100 ml) solution (2 mg/ml)
was added to each well to incubate for 2 h. Absorbance
at 570 nm was recorded using a 96-well plate reader.
Inhibition curves were drawn by means of values
obtained by OD percentages versus control for each
concentration and IC
50
values were calculated by the
GraFit method (Erithacus Software Limited, Staines,
UK). The Chou and Talalay (1984) combination index
(CI) analysis is a well-established index to determine
the pharmacologic interaction of two drugs. When
CIZ1 (represents an additive effect of the two drugs),
the CI formula is in the same form as a traditional
isobologram equation. Synergism is defined as more
than the expected additive effect with CI !1 and
antagonism is defined as CI O1.
C Festuccia and G L Gravina et al.: Azacitidine effects in advanced prostate cancer models
www.endocrinology-journals.org402
Apoptosis analysis by flow cytometry
Treated and untreated cells were collected and fixed
for 30 min by the addition of 1 ml 70% ethanol. After
30 min, the cells were pelleted by centrifugation
(720 g; 5 min), and resuspended in 1 ml DNA staining
solution (PBS containing 200 mg/ml RNase A,
20 mg/ml propidium iodide plus 0.1% Triton X-100)
and stained by incubation at room temperature for
60 min. All cells were then measured on a FACScan
flow cytometer (Becton Dickinson, Bedford, MA,
USA) and analyzed using Cell Quest software (Becton
Dickinson). Resulting DNA distributions were analy-
zed by Mod.t (Verity Software House, Topsham, ME,
USA) for the proportion of cells in sub-G0 (apoptotic
cells), G1, S, and G2-M phases of the cell cycle.
In vivo treatments
Male CD1 nude mice (Charles River, Milan, Italy)
were maintained under the guidelines established by
our Institution (University of L’Aquila, Medical
School and Science and Technology School Board
Regulations, complying with the Italian government
regulation no. 116, January 27 1992, for the use of
laboratory animals). All mice received s.c. flank
injections of 1!10
6
PC3 and 22v1 cells. Tumor
growth was assessed by bi-weekly measurement
of tumor diameters with a Vernier caliper
(length!width). Tumor weight was calculated accord-
ing to the formula: TW (mg)Ztumor volume (mm
3
)Z
d
2
!D/2, where dand Dare the shortest and the longest
diameters respectively. Treatments used in our in vivo
study were started when tumor volumes reached about
80 mm
3
(Day 0) and were stopped after 28 days. Group
1: 10 mice received i.p. injections of 100 l PBS for
consecutive 7 days. Group 2: 10 mice received i.p.
injections of 100 l of aza-CR (0.8 mg/kg) for consecu-
tive 7 days. Group 3: 10 mice received i.p. injections of
DTX (7.5 mg/kg per week) every week for a maximum
of four administrations. Group 4: 10 mice received i.p.
injections of 100 l of aza-CR (0.8 mg/kg) for consecu-
tive 7 days followed by a dose of DTX (7.5 mg/kg per
week) every week for a maximum of three adminis-
trations. Group 5: 10 mice received i.p. injections of
5.0 mg/kg per day cisplatin for 10 consecutive days.
Group 6: mice received i.p. injections of 100 l of aza-
CR (0.8 mg/kg) for consecutive 7 days followed by i.p.
injections of 5.0 mg/kg per day cisplatin for 10
consecutive days. Experiments were stopped at 28th
day. We replicate the experiments in order to analyze a
total of 20 animals for group. The effect of azacitidine
alone or in combination with cisplatin (CP) or DTX on
tumor growth was evaluated measuring, for all groups,
the mean tumor weight at the end of experiments.
Fractional tumor volume (FTV) for each treatment
group was calculated as the ratio between the mean
tumor volumes of treated and untreated animals. This
was performed for treatment a (FTVa), for treatment
b (FTVb), and for treatment aCb(FTVaCb).
Combination indices were assayed as described
above. Animals were killed by spinal cord dislocation
and tumors were subsequently removed surgically. A
part of the tumor was directly frozen in liquid nitrogen
for protein analysis and the other part was fixed in
paraformaldehyde overnight for histochemical evalu-
ations and immunohistochemical analyses including
endothelial (CD31 and vWF; Dako, Glostrup,
Denmark), proliferative (Ki67; Dako), apoptosis
(bcl2, p21, p27, death receptors and their ligands
Santa Cruz), and signaling (p-Akt; Rockland Immuno-
chemicals, Gilbertsville, PA, USA) marker analysis.
Anti-DNMT1, DNMT3a, and DNMT3b antibodies
were purchased from Biocarta (Hamburg, Germany).
Indirect immunoperoxidase stain of tumor xenografts
was performed on paraffin embedded with four
micrometer tissue sections. Briefly, sections were
incubated with primary antibodies for a night at 4
o
C.
Next, avidin–biotin assays were done using the
Vectastain Elite kit obtained from Vector Laboratories.
Mayer’s hematoxylin was used as nuclear counterstain.
Slides were analyzed separately from CF and PM.
Martius yellow-brilliant crystal scarlet blue
technique
Stains for these techniques were purchased from HD
Supplies (Aylesbury, UK). This technique was used to
analyze the presence of red cells dispersed in tumor
tissue and present in blood vessels as well as to
evaluate better the presence of micro-thrombi and
bleeding zones. Martius yellow, a small molecule dye,
together with phosphotungstic acid in alcoholic
solution stains red cells. Early fibrin deposits may be
colored, but the phosphotungstic acid blocks the
staining of muscle, collagen, and connective tissue
fibers. Brilliant crystal scarlet, a medium-sized
molecule, stains muscle, and mature fibrin. Phospho-
tungstic acid removes any red stain from the collagen.
The large molecule dye aniline blue stains the collagen
and old fibrin.
Hemoglobin assay
Tumor hemoglobin levels were quantified as described
elsewhere (14). Tumors were homogenized in double-
distilled water. Eighty microliters of homogenates
were mixed with 1 ml Drabkin’s solution and
Endocrine-Related Cancer (2009) 16 401–413
www.endocrinology-journals.org 403
incubated for 15 min at room temperature. After
centrifugation at 400 gfor 5 min, the supernatants
were subjected to absorbance measurement at 540 nm.
The absorption that is proportional to hemoglobin
concentration, was divided by tumor weight.
Assessment of global methylation levels
Proteins were extracted from untreated and in vivo
Aza-CR-treated tumors and cells at 7, 14, 21, and
28 days and DNMT activity were assayed. As
described, frozen tumors were pulverized in a liquid
nitrogen-cooled Thermovac pulverizer. The resulting
powders were homogenized in 10 volumes of a 10 mM
Tris–HCl buffer, pH 7.4, containing 1 mM EDTA,
0.5 mM dithiothreitol, 10 mM sodium molybdate,
phosphatase inhibitor cocktail 2 with a dilution 1/100,
and protease inhibition cocktail 2 with a dilution 1/100,
both from Sigma-Aldrich. The homogenates were
centrifuged for 1 h at 105 000 g(C48C) and the
supernatants (cytosols) were used for protein
determination by immunoblotting. Total protein con-
tent was measured using the bicinchoninic acid assay.
Western blot analysis
Cells were washed with cold PBS and immediately
lysed with 1 ml lysis buffer (50 mM HEPES, pH 7.5,
150 mM NaCl, 10% glycerol, 1% Triton X-100, 1 mM
EDTA, 1 mM EGTA, 50 mM NaF, 1 mM sodium
orthovanadate, 30 mM p-nitrophenyl phosphate,
10 mM sodium pyrophosphate, 1 mM phenylmethyl-
sulfonyl fluoride, 10 mg/ml aprotinin, and 10 mg/ml
leupeptin). Flash-frozen tissue samples were crushed
using liquid nitrogen pre-chilled mortar and pestles.
Upon addition of lyses buffer, tissues were further
homogenized with an electric homogenizer and
centrifuged in a microfuge a top spin. Supernatants
were collected and analyzed. Cell lysates and tissue
extracts were electrophoresed in 7% SDS-PAGE, and
separated proteins transferred to nitrocellulose and
probed with the appropriate antibodies using the
conditions recommended by the antibody suppliers.
Statistics analysis
Continuous data were expressed as mean and S.D. and
were compared using an umpired Student’s t-test.
Categorical data were analyzed by the exact Fisher’s
test. Pvalue less than 0.05 were considered statistically
significant.
Results
In vitro experiments: effects of azacitidine
as single agent
Initially, we analyzed the effects of azacitidine on
DNA methylation and expression of DNMT1,
DNMT3a, and DNMT3b. After 48 h of treatment
with azacitidine, the decrease in the DNMT activity
was maximal after 10 days and it was maintained up to
2 weeks (Fig. 1A). Simultaneously, a significant
decrement in DNMT1 and DNMT3b expression levels
occurred with a peak observed at 5–10 days (Fig. 1B).
Conversely, azacitidine treatment did not affect the
protein expression levels of DNMT3a. Antiprolifera-
tive effect of azacitidine treatment was higher in 22rv1
cells than in PC3 cells with IC50 of 0.5 and 2.5 mM
respectively. Treatment of with azacitidine resulted in
a dose- and time-dependent inhibition of growth with
G1 phase arrest associated to apoptosis in 22rv1 cells,
whereas this drug arrested PC3 cells in G2/M phase
without acute cell killing. Also, azacitidine increased
the levels of p21, p27, p53, Bcl-Xs, and Bax and to
reduce Bcl2 and Bcl-Xl expression in 22rv1 cells
(Fig. 2A). This was also associated with an increased
expression of cleaved caspase 3 and PARP. The
expression of p21, Bcl2, caspase 3, and PARP was
Figure 1 DNMT activity (A) and expression of DNMT1,
DNMT3a, and DNMT3b (B) measured at different time after a
48 h 1.0 mM Aza-CR treatment in 22rv1 and PC3 cells. In PC3
cells, a decrement of about twofold in DNMT1 and fivefold in
DNMT3b expression was observed after 5–10 days of
treatment with azacitidine. The levels of DNMT1 were restored
after 15 days, whereas the levels of DNMT3b were maintained
lower up to 20 days (about twofold). In 22rv1 cells, the levels of
DNMT1 and DNMT3b were weakly reduced also if the overall
DNMT activity was significantly decreased.
C Festuccia and G L Gravina et al.: Azacitidine effects in advanced prostate cancer models
www.endocrinology-journals.org404
not changed in PC3 cells following azacitidine
treatment, whereas the expression of protein associated
to G2/M arrest such as p16INKA, cyclin B and cdk1,
cdk2 and cdk4 were differentially modulated (Fig. 2B).
The treatment with HA-14-1, a Bcl2 antagonist, was able
to induce more apoptotic events when co-administrated
with Aza-CR in PC3 (Fig. 3A) and 22rv1 cells
(Fig. 3B). These results suggest that the amount of
apoptosis is related to Bcl2 levels and the low levels of
apoptosis observed in PC3 cells might be partially
associated with the higher expression of Bcl2. Aza-CR
treatment in PC3 cells was also associated with the
phosphorylation of p38 MAP kinase and with the
reduced expression of activated Erk (Fig. 5B) which it
is required for the induction of cell cycle arrest in the
G2/M phase. However, the induction of p38 MAPK
activity was able to start the apoptotic program by
downmodulation of Bcl2 when Aza-CR treated PC3
cells were grown in polyhema-covered Petri dishes
(Fig. 3C). The apoptosis mediated by absence of
adhesion (anoikis) was induced by a p38 MAPK-
dependent Bcl-2 downmodulation (Fig. 3D) because
SB203580, a p38 MAPK inhibitor, treatment caused a
significative slow down in Aza-CR/polyhema-dependent
Bcl2 downmodulation.
In vitro effects of azacitidine in combination with
DTX or cisplatin
We analyzed the effects of combined treatments with
azacitidine (0.5–1 mM for PC3 and 0.1–0.5 mM for
22rv1), DTX (0.5, 1.0, and 10 nM for both cells) and
cisplatin (1, 2.5, and 10 mg/ml for both cells).
Azacitidine sensitized versus DTX and cisplatin in
PC3 and 22rv1 cells (Tables 1 and 2). In PC3 cells, the
stronger antitumor effect was observed with azacitidine
(1.0 mM) and DTX (1.0 nM (CIZ0.29)) and
with azacitidine (1.0 mM) and cisplatin (10 mg/ml
(CIZ0.27)). In 22rv1 cells, the highest antitumor
effects were observed with azacitidine (0.5 mM) and
Figure 2 Western blot analysis on the expression of pro-
apoptotic and antiapoptotic elements in 22rv1 (A) and PC3 (B)
cells treated with 1, 2.5, and 5.0 mM aza-CR. In 22rv1 cells, aza-
CR increased the expression of pro-apoptotic elements such as
p21 (twofold with 2.5 and 5.0 mM aza-CR), p53 (four- to five-fold
with 2.5 and 5.0 mM aza-CR), Bax (two- to four-fold with 2.5 and
5.0 mM aza-CR) and Bcl-XS (twofold with both 2.5 and 5.0 mM
aza-CR) as well as the activity of caspase 3 (four- to six-fold
with 2.5 and 5.0 mM aza-CR). The antiapoptotic element Bcl2
and Bcl-XL was reduced (about tenfold with both 2.5 and 5.0 mM
aza-CR). In PC3 cells, no caspase 3 and PARP activity was
showed and this was associated to no significant modulation of
Bcl2 and p21 expression. Modulation in check point proteins
of cell cycle was, however, shown. P16INK4A levels were
significantly increased (about twofold with 5.0 mM aza-CR),
CdK1 levels were increased of about two with both 1.0 and
2.5 mM aza-CR and of about fourfold with 5.0 mM aza-CR, CdK2
levels were weakly reduced (1.5-fold with 5.0 mM
aza-CR), CdK4 levels were significantly reduced of about
fourfold with 2.5 and 5.0 mM aza-CR and cyclin B1 levels were
not modified. Each lane was loaded with 40 mg proteins and
normalized versus actin. This experiment is representative of
three individual experiments.
Endocrine-Related Cancer (2009) 16 401–413
www.endocrinology-journals.org 405
DTX (1.0 nM (CIZ0.50)) and with azacitidine
(1.0 mM) and cisplatin (2.5 mg/ml (CIZ0.44)).
In vivo effects of azacitidine as single drug or
followed by DTX or cisplatin
The i.p. administration of Vidaza (0.8 mg/kg for 7
consecutive days) on intact nude male mice inoculated
subcutaneously with 22rv1 and PC3 cells resulted in a
statistically significant tumor growth decrement
(P!0.001) in both xenograft models (Fig. 4). In PC3
xenografts (Table 3), following azacitidine treatment
we demonstrated a 36.7% of reduction in tumor
weight respect to controls (P!0.001) with a tumor
growth delay (TGD) of 5.5 days. The proliferation
index was also significantly reduced to 42.6% in
azacitidine treated tumors (P!0.001) with tumor
apoptosis of about 5%. The vessel count was also
reduced to 18.7% (P!0.05). In 22rv1 xenografts
(Table 4), we demonstrated a 65.5% reduction of tumor
weight in aza-CR treated mice respect to controls
(P!0.001), with a tumor growth delay (TGD) of
25.5 days. The proliferation index was also reduced to
74.3% (P!0.001) and apoptosis was evident in about
15% of cells. The vessel count was reduced to 70.4%
Figure 3 Role of Bcl2 in the absence of aza-mediated PC3 cell apoptosis. We used the Bcl2 inhibitor HA-14.1 to demonstrate that
bcl2 overexpression in PC3 cells could be responsible for the absence of acute apoptosis induced by aza-CR treatment. HA-14.1
(10 mM) triggered apoptosis of PC3 cells both in basal culture condition and in the presence of aza-CR pretreatment (1.0 mM).
HA-14.1 was synergistic with aza-CR in PC3 cells, whereas this drug was only additive in 22rv1 cells. These results are
representative of three individual experiments. Each lane was loaded with 40 mg proteins.
Table 1 Pharmacological parameters of combination treatment with azacitidine and docetaxel in PC3 and 22rv1 cells
Treatment A (DTX) Treatment B (aza-CR) Combination treatment
Cells Dose (nM) GI Dose (mM) GI Expected GI Observed GI Increment (CI)
PC-3 0.5 0.95 0.5 0.90 0.85 0.54 1.57 (0.64)
1 0.75 0.70 0.25 2.80 (0.36)
1.0 0.90 0.5 0.90 0.80 0.31 2.58 (0.39)
1 0.75 0.70 0.20 3.50 (0.29)
10.0 0.75 0.5 0.90 0.70 0.25 2.80 (0.36)
1 0.75 0.50 0.20 2.50 (0.40)
22rv1 0.5 0.90 0.1 0.75 0.65 0.40 1.62 (0.62)
0.5 0.50 0.40 0.30 1.33 (0.75)
1.0 0.70 0.1 0.75 0.45 0.25 1.80 (0.55)
0.5 0.50 0.20 0.10 2.00 (0.50)
10.0 0.55 0.1 0.75 0.30 0.20 1.50 (0.67)
0.5 0.50 0.05 0.10 0.50 (2.00)
GIZinhibition (percent versus control).
C Festuccia and G L Gravina et al.: Azacitidine effects in advanced prostate cancer models
www.endocrinology-journals.org406
(P!0.001). Similarly to that observed in vitro, the
levels of DNMT1 and DNMT3b were also lower in
azacitidine treated mice when compared with controls
both in PC3 and 22rv1 xenograft models (data not
shown).
The treatment with azacitidine followed by DTX or
CP was also studied in vivo. An increase in the
effectiveness of DTX and cisplatin was observed after
7 days of co-administration with azacitidine both in
PC3 and 22rv1 xenograft models (Fig. 4 and Tables 3
and 4). The results are as follows:
(i) Docetaxel and cisplatin alone reduced tumor
weight of PC3 xenografts by 41.0% (P!0.001) and
22.2% (P!0.01), with a tumor growth delay of 5.1 and
2.1 days respectively when compared with controls.
The proliferation index was also reduced to 49.9%
(P!0.001) and 23.6% (P!0.01) respectively in DTX-
and cisplatin-treated tumors. Apoptosis was evident
only in DTX-treated tumors at levels of about 7%
(P!0.01). In this xenograft model, 7 days of treatment
with azacitidine followed by DTX or cisplatin resulted
in a significant decrease in tumor weight of 76.4%
Table 2 Pharmacological parameters of combination treatment with azacitidine and cisplatin in PC3 and 22rv1 cells
Treatment A (CP) Treatment B (aza-CR) Combination treatment
Cells Dose (mg/ml) GI Dose (mM) GI Expected GI Observed GI Increment (CI)
PC-3 1 1.00 0.5 0.90 0.90 0.60 1.50 (0.67)
1 0.75 0.75 0.45 1.67 (0.60)
2.5 0.90 0.5 0.90 0.80 0.40 2.00 (0.50)
1 0.75 0.65 0.32 2.03 (0.49)
10.0 0.80 0.5 0.90 0.70 0.25 2.80 (0.36)
1 0.75 0.55 0.15 3.67 (0.27)
22rv1 2.5 0.90 0.1 0.85 0.75 0.60 1.25 (0.80)
0.5 0.67 0.57 0.44 1.30 (0.77)
10.0 0.90 0.1 0.85 0.75 0.45 1.67 (0.60)
0.5 0.67 0.57 0.25 2.28 (0.44)
10.0 0.85 0.1 0.85 0.70 0.35 2.00 (0.50)
0.5 0.67 0.52 0.25 2.08 (0.48)
GIZinhibition.
Figure 4 Tumor proliferation in PC3 and 22rv1 xenografts in the presence of azacitidine alone or in combination with docetaxel (DTX)
and cisplatin (CP). Group 1: 10 mice received (i.p.) injections of 100 ml PBS for consecutive 7 days. Group 2: 10 mice received i.p.
injections of 100 ml of aza-CR (0.8 mg/kg) for consecutive 7 days. Group 3: 10 mice received i.p. injections of docetaxel (7.5 mg/kg
per week) every week for a maximum of four administrations. Group 4: 10 mice received i.p. injections of 100 ml of aza-CR
(0.8 mg/kg) for consecutive 7 days followed by a dose of docetaxel (7.5 mg/kg per week) every week for a maximum of three
administrations. Group 5: 10 mice received i.p. injections of 5.0 mg/kg per day cisplatin for 10 consecutive days. Group 6: mice
received i.p. injections of 100 ml of aza-CR (0.8 mg/kg) for consecutive 7 days followed by i.p. injections of 5.0 mg/kg per day cisplatin
for 10 consecutive days. Proliferation was measured considering the variation of tumor volume (mm
3
) in the time (days). Controls
were administered with saline and treated xenografts were administered with 0.8 mg/kg for the first 7 days of treatment. Animals
were killed at 28th day of treatment.
Endocrine-Related Cancer (2009) 16 401–413
www.endocrinology-journals.org 407
(P!0.001 versus both Vidaza and DTX single
treatments) and of 58.3% (P!0.01 versus Vidaza
and P!0.001 versus cisplatin) respectively. Moreover,
sequential regimens of azacitidine CDTX or
azacitidine Ccisplatin were respectively associated
with tumor growth delay values of 25.5 and 15.2 days:
reductions in the proliferation index of 96.5%
(P!0.001 versus both single treatments) and 81.5%
(P!0.001 versus both single treatments); significant
increases in apoptosis of 18% (P!0.001 versus both
single treatments) and 14.3% (P!0.001 versus both
single treatments); and reductions in vessel counts of
w75% (P!0.001 versus both single treatments) and
55% (P!0.001 versus Vidaza and P!0.01 versus
cisplatin treatments). Additionally, 15.0% (3/20, NS)
and 25.0% (5/20, (PZ0.056, NS)) of mice were tumor-
free after 28 days of treatment.
(ii) In 22rv1 xenografts, DTX or cisplatin alone
reduced tumor weight of 46.8% (P!0.001) and 30.9%
(P!0.001), with a tumor growth delay of 17.3 and
7.5 days respectively when compared with controls.
The proliferation index was also reduced of 66.1%
(P!0.001) and 53.2% (P!0.001) respectively in
DTX- and cisplatin-treated animals. Apoptosis was
evident in 10% (P!0.05) and 25% (P!0.01) of cells
in DTX- and cisplatin-treated animals respectively.
In this xenograft model, 7 days of treatment with
azacitidine followed by DTX or cisplatin resulted
in a significant decrease in tumor weight of 68.5%
(P!0.05 versus DTX treatment, whereas not signi-
ficant versus Vidaza single treatment) and 77.5%
(P!0.001 versus both single treatments) respectively.
Additionally, sequential regimens of azacitidine CDTX
or azacitidine Ccisplatin were respectively associated
with tumor growth delay values of 28.0 and 30.2 days;
reductions in the proliferation index of 79.6% (P!0.01
versus Vidaza and P!0.001 versus DTX single
treatments) and 83.5% (P!0.001 versus both single
treatments); significant increases in apoptosis of
18.2 and 34.3% (P!0.001 versus all single treat-
ments); reductions in vessel counts of about
75% (P!0.05 versus Vidaza and P!0.001 versus
Table 3 Antitumor activity of Vidaza in PC-3 tumors alone or in combination with docetaxel (DTX) or cisplatin (CP)
Treatment
Drug
Dose
mg/kg Mice
Weight
of mice
(grGS.D.)
Tumor
weight
(mgGS.D.)
TGD
(days)
PI
(Ki67 %) Apoptosis Vessels
Tumor-free
mice
Saline 20 25.0G2.1 585G135 37.3G4.5 !2 30.0G4.3 0/20
Vidaza 0.8 20 23.4G2.3 370G183 5.5 21.4G2.7 5.4G2.6 24.4G2.7 0/20
DTX 7.5 20 25.0G1.7 345G132 5.1 18.7G3.3 7.2G2.7 27.5G4.5 0/20
VidazaCDTX 0.8C7.5 20 21.8G2.6 138G84 25.5 1.3G1.7 18.2G3.2 7.5G2.5 3/20
CP 5 20 25.1G1.5 455G161 2.1 28.5G3.1 !2 28.5G3.5 0/20
VidazaCCP 0.8C5 20 20.8G2.8 244G58 15.2 6.9G0.6 14.3G2.1 13.4G4.4 5/20
TGD (tumor growth delay) was determined by the difference in time taken for the mean tumor volume to double in treated versus
control animals. PI (proliferation index) was measured considering the mean of Ki67 positive cell percentageGS.D. measured on five
random fields at !100. Apoptosis was measured as the percentage of tunnel positive cells GS.D. measured on five random fields
(!400). Vessels were measured as the vessel number per field at !100.
Table 4 Antitumor activity of Vidaza in 22rv1 tumors alone or in combination with docetaxel (DTX) or cisplatin (CP)
Treatment
Drug
Dose
mg/kg Mice
Weight of
mice
(grGS.D.)
Tumor
weight
(mgGS.D.)
TGD
(days)
PI
(Ki67 %) Apoptosis Vessels
Tumor-free
mice
Saline 20 25.3G1.8 652G220 45.5G6.5 !2 38.5G5.0 0/20
Vidaza 0.8 20 24.1G2.0 225G55 25.5 11.7G0.7 15.4G3.0 11.4G0.8 1/20
DTX 7.5 20 25.0G1.7 305G121 17.3 15.4G5.1 10.2G1.5 17.5G3.0 0/20
VidazaCDTX 0.8C7.5 20 21.8G2.6 205G125 28.0 9.3G1.4 18.2G2.5 9.5G1.0 2/20
CP 5 20 25.1G1.5 450G101 7.5 21.3G3.9 25.5G2.5 7.5G1.5 1/20
VidazaCCP 0.8C5 20 20.8G2.8 147G66 30.2 7.5G2.0 34.3G8.0 7.4G4.4 6/20
TGD (tumor growth delay) was determined by the difference in time taken for the mean tumor volume to double in treated versus
control animals. PI (proliferation index) was measured considering the mean of Ki67 positive cell percentage GS.D. measured on
five random fields at !100. Apoptosis was measured as the percentage of tunnel positive cells GS.D. measured on five random
fields (!400). Vessels were measured as the vessel number per field at !100.
C Festuccia and G L Gravina et al.: Azacitidine effects in advanced prostate cancer models
www.endocrinology-journals.org408
DTX single treatments) and 81% (P!0.001 versus
Vidaza and NS versus cisplatin single treatments).
Moreover, 10.0% (2/20) and 30.0% (6/20) of mice
treated with the Vidaza plus DTX and Vidaza plus
cisplatin combined treatments were tumor-free after
28 days, although only the second combined treatment
was statistically significant versus control (P!0.05)
and the statistic analysis showed not significant values
for the comparisons versus Vidaza or cisplatin
(PZ0.096 for both comparisons).
(iii) Sequential treatments with either azacitidine
CDTX or azacitidine Ccisplatin resulted in a
significant reduction in tumor blood vessels compared
with controls (Tables 3 and 4) both in PC3 and 22rv1
xenografts. This antiangiogenetic effect was further
supported by quantification of hemoglobin content in
Figure 5 (A) Western blot analysis showing the increment of DNMT1 andDNMT3b expression without modulation of Akt activity after
CP treatment in PC3 and 22rv1 tissues. Forty micrograms of proteins were loaded in each lane. (B). Immunohistochemical appearance
of PC3 tumors harvested from control andCP treated mice showing increased DNMT1 expression after CP treatment. (C). Microscopic
appearance of PC3 and 22rv1 tumors stained withMartius yellow-brilliant crystal scarlet blue technique. We show the presence of large
blood vessels (stained in yellow/orange)near to cancer cell nests of untreated 22rv1 tumors. Untreated PC3 tumors show the presence
of numerous and small blood vessels scatteredin the tumor mass and delineating single tumor cell nests. Vidaza reduces blood vessel
number both in 22rv1 and PC3 xenografts. Vessels appear dilated with unstructured capillary bed dispersed in collagen I deposits
(azure staining) primarily in 22rv1 tumors. DTX treated 22rv1 tissues appear with large unstructured capillary bed and abundant
hemorrhagic areas. The presence of numerousphagocytes (neutrophilic granulocytes and monocytes) dispensed in collagen I deposit
suggests that a previous colliquative necrosis was present. In the combined treatment (Vidaza plus DTX), the above-mentioned
appearance was amplified with deposition also of fibrin clots (pale pink-staining) evident in 22rv1 tissues and formation of dense
collagen I deposits (fibrosis) which envelop tumor cell nests. Single pictures are at !400. (B) Angiogenesis was evaluated measuring
hemoglobin presence in tissue extracts or counting CD34/von Willebrand double positive endothelial cells forming blood vessels. Full
colour version of this figure available via http://dx.doi.org/10.1677/ERC-08-0130.
Endocrine-Related Cancer (2009) 16 401–413
www.endocrinology-journals.org 409
tumor tissues, used as a marker of the number of
blood vessels. To specifically assess vascular density
within the tumors, sections were stained with Masson
trichromic and BMBS. Blue-stained spiral strands of
collagen were detected in peripheral areas, whereas
PC3 cells grow to form large cell masses that were
surrounded by red-stained blood vessels that often
seemed dilated. Control xenografts exhibited numer-
ous dilated and even sinusoid-looking vessels as
compared with treated xenografts. The antiangiogenic
effects of azacitidine followed by either DTX or
cisplatin were further confirmed by immunohisto-
chemical localization of von Willebrand factor that is
a marker for endothelial cells and the presence of
microvessels. However, a reduction in microvessel
density was only observed in tumors treated with
azacitidine Ccisplatin compared with controls or
with azacitidine alone. Results also showed anti-
angiogenetic effects with the combination of azaciti-
dine and DTX, including significant reductions in the
vessels analyzed with the presence of fibrin clots and
red dispersion in the tumor parenchyma, and the
amount of hemoglobin analyzed in tissue extracts
(Fig. 5A and B).
(iv) In addition, we found that treatments with
cisplatin were able to increase the levels of DNMT1
in PC3 but not in 22rv1 cells. This is in agreement
with the higher effects of combination azacitidine
plus cisplatin in PC3 when compared with those
observed in 22rv1. In Fig. 5C, we show western blot
analysis performed on PC3 and 22rv1 tissue extracts
whereas in Fig. 5D, we show the expression of
DNMT1 in PC3 xenografts with or without cisplatin
treatment. DTX was not able to modify DNMT
expression in PC3 and 22rv1 xenografts (data not
shown) supporting the high effects of combination
treatment with azacitidine and cisplatin observed in
both PCa models.
Discussion
One of the major obstacles in curing locally advanced
and metastatic prostate cancer is the development of
resistance to therapy. Historically, chemotherapy has
had limited utility in treating castration-resistant
locally advanced and metastatic prostate cancer.
Nevertheless, DTX is the standard first-line chemo-
therapy for this disease. The responses to DTX are,
however, partial and associated with increased resis-
tance to apoptosis, thus indicating a clear need for new
therapies in HRPC patients. A recent report shows that
single-agent satraplatin improves PFS as second-line
chemotherapy for patients with HRPC (Nakabayashi
et al. 2008,Ross et al. 2008). The resistance of tumor
cells to anticancer agents remains a major cause of
failure in the treatment of patients with cancer. The
classic mechanism for the evolution of a multidrug-
resistant phenotype was believed to involve a single
molecular mechanism, such as overexpression of
P-glycoprotein. However, it now seems that the
multidrug-resistant phenotype represents a complex,
multifactor process in which epigenetic mechanisms
may play a crucial role. Epigenetic alterations,
including aberrant promoter methylation, are respon-
sible for the silencing of tumor suppressor genes and
for increased resistance to chemotherapy. This high-
lights a potential role for DNMT as potential molecular
targets in cancer therapy (Mishra et al. 2008).
Exploiting gene reactivation by epigenetic acting
agents in combination with cytotoxic therapies has
been a strategy that holds much clinical promise (for
review, see Lu et al. 2006). Although azacitidine has
shown significant promise in the treatment of hemato-
poietic malignancies, and is approved for the treatment
of myelodysplastic syndrome (MDS; Issa et al. 2005,
Abdulhaq & Rossetti 2007), research into its activity in
solid tumors has been less impressive and have been
similarly disappointing (Schwartsmann et al. 2000,
Pohlmann et al. 2002,Appleton et al. 2007). However,
it is important to note that the majority of these studies
used dose and schedule combinations that are now
recognized to be suboptimal. In addition, the major
issue involves either drug uptake or tumor cell
proliferation rate because the analysis of tumor tissues
showed a disappointingly small decrease of methyl-
ation (3%). This has been shown to be below the
threshold necessary for resensitization to chemother-
apy (Appleton et al. 2007). Our research demonstrates
that tumor levels of DNMT1 and DNMT3b were lower
in azacitidine-treated conditions both in vitro and in
PC3 and 22rv1 xenograft models. Our in vitro data
indicate that DNA methylation activity reaches
maximum inhibition of inhibition at 10 days after a
48 h of azacitidine treatment. This observed inhibition
remained at high levels for a maximum 2 weeks.
In parallel, a significant downregulation in DNMT1
and DNMT3b expression occurred and peaked at
5–10 days, and persisted even after 14 days. This
reduction of DNMT activity was associated with the
in vitro and in vivo re-expression of markers of
differentiation, including AR (Jarrard et al. 1998,
Gravina et al.2008), and is responsible for the
restoration of responsiveness to hormonal therapy
(Izbicka et al. 1999,Sonpavde et al. 2007,Gravina
et al. 2008). This reduction in DNMT activity is also
likely responsible for the delay in progression to
C Festuccia and G L Gravina et al.: Azacitidine effects in advanced prostate cancer models
www.endocrinology-journals.org410
androgen independent disease and for the improvement
in survival in the transgenic adenocarcinoma of mouse
prostate mouse model of prostate cancer (Zorn et al.
2007). In the present report, we observed also that
DNMT inhibition with azacitidine was able to induce
the in vitro and in vivo expression of p16INKA, Bax,
Bak, p21/WAF1, and p27/KIP1. This was also
responsible for inhibiting the activation of Akt, the
expression of cyclin D1, Bcl-2, and Bcl-XL associated
with the processing of caspases-3 with increased
apoptosis. Nevertheless, we observed an increased
Bcl2 mediated apoptosis by culturing aza-CR treated
cells in presence of polyhema which impedes cell
adhesion. This condition induces a specialized apopto-
tic mechanism termed anoikis that is normally
suppressed in tumor cells (Diaz-Montero et al. 2006,
Glinsky 2006,Kupferman et al. 2007) including PC3
cells. Although the reactivation of anoikis by aza-CR
can represent an important therapeutic approach to
increase tumor cell death present in the blood stream
and could slow down the metastatic process and
contribute to antimetastatic treatments of advanced
tumors, ad hoc experiments are required.
In addition, the downmodulation of p-Akt and
members of Bcl-2 family could indeed sensitize PC3
and 22rv1 cells versus DTX and cisplatin increasing
the antiproliferative and pro-apoptotic effects of single
drugs both in vitro and in vivo. The in vivo experiments
revealed synergistic effects with DTX and cisplatin
both in PC3 and 22rv1 xenografts. Immunocyto-
chemistry and enzymatic assays revealed also that
in vivo DNMT1 expression was significantly increased
in cisplatin treated 22rv1 and PC3 xenografts, whereas
the induction of DNMT1 levels was lower in DTX
treated tumors. This is in agreement with previous
reports showing that an increased DNMT1 expression
is a common event in a multimodality-resistant
phenotype in tumor cells (Misrha et al. 2008), in
cisplatin and taxane (Segura-Pacheco et al. 2006)
resistance mechanisms. Taken together, our study
indicate the DNMT downmodulation represents an
useful therapeutic approach for enhancing chemosen-
sitivity of prostate cancers to DTX and cisplatin and
provide a rationale for clinical trials on combination
treatments with azacitidine in patients with advanced
prostate tumors.
Declaration of interest
There is no conflict of interest that could be perceived as
prejudicing the impartiality of the research reported.
Funding
This research did not receive any specific grant from any
funding agency in the public, commercial or not-for-profit
sector.
References
Abdulhaq H & Rossetti JM 2007 The role of azacitidine in
the treatment of myelodysplastic syndromes. Expert
Opinion on Investigational Drugs 16 1967–1975.
Appleton K, Mackay HJ, Judson I, Plumb JA, McCormick C,
Strathdee G, Lee C, Barrett S, Reade S, Jadayel D et al.
2007 Phase I and pharmacodynamic trial of the DNA
methyltransferase inhibitor decitabine and carboplatin
in solid tumors. Journal of Clinical Oncology 25
4603–4609.
Armstrong AJ, Garrett-Mayer ES, Yang YC, de Wit R,
Tannock IF & Eisenberger M 2007 A contemporary
prognostic nomogram for men with hormone-refractory
metastatic prostate cancer: a TAX327 study analysis.
Clinical Cancer Research 13 6396–6403.
Brown R & Strathdee G 2002 Epigenomics and epigenetic
therapy of cancer. Trends in Molecular Medicine 8
S43–S48.
Bastola DR, Pahwa GS, Lin MF & Cheng PW 2002
Downregulation of PTEN/MMAC/TEP1 expression
in human prostate cancer cell line DU145 by growth
stimuli. Molecular and Cellular Biochemistry 236
75–81.
Bertram J, Peacock JW, Fazli L, Mui AL, Chung SW, Cox
ME, Monia B, Gleave ME & Ong CJ 2006 Loss of PTEN
is associated with progression to androgen independence.
Prostate 66 895–902.
Calabro
`F & Sternberg CN 2007 Current indications for
chemotherapy in prostate cancer patients. European
Urology 51 17–26.
Chou TC & Talalay P 1984 Quantitative analysis of dose-
effect relationships: the combined effects of multiple
drugs or enzyme inhibitors. Advances in Enzyme
Regulation 22 27–55.
Desireddi NV, Roehl KA, Loeb S, Yu X, Griffin CR,
Kundu SK, Han M & Catalona WJ 2007 Improved
stage and grade-specific progression-free survival rates
after radical prostatectomy in the PSA era. Urology 70
950–955.
Diaz-Montero CM, Wygant JN & McIntyre BW 2006
PI3-K/Akt-mediated anoikis resistance of human
osteosarcoma cells requires Src activation. European
Journal of Cancer 42 1491–1500.
Glinsky 2006 Genomic models of metastatic cancer:
functional analysis of death-from-cancer signature genes
reveals aneuploid, anoikis-resistant, metastasis-enabling
phenotype with altered cell cycle control and activated
Polycomb Group (PcG) protein chromatin silencing
pathway. Cell Cycle 51208–1216.
Gravina GL, Festuccia C, Millimaggi D, Dolo V, Tombolini V,
de Vito M, Vicentini C & Bologna M 2008 Chronic
Endocrine-Related Cancer (2009) 16 401–413
www.endocrinology-journals.org 411
azacitidine treatment results in differentiating effects,
sensitizes against bicalutamide in androgen-independent
prostate cancer cells. Prostate 68 793–801.
Hellebrekers DM, Jair KW, Vire
´E, Eguchi S, Hoebers NT,
Fraga MF, Esteller M, Fuks F, Baylin SB, van Engeland M
& Griffioen AW 2006 Angiostatic activity of DNA methyl-
transferase inhibitors. Molecular Cancer Therapeutics 5
467–475.
Haas GP, Delongchamps N, Brawley OW, Wang CY & de la
Roza G 2008 The worldwide epidemiology of prostate
cancer: perspectives from autopsy studies. Canadian
Journal of Urology 15 3866–3871.
Izbicka E, MacDonald JR, Davidson K, Lawrence RA,
Gomez L & Von Hoff DD 1999 5,6-dihydro-50-
azacytidine (DHAC) restores androgen responsiveness in
androgen-insensitive prostate cancer cells. Anticancer
Research 19 1285–1291.
Issa JP, Gharibyan V, Cortes J, Jelinek J, Morris G,
Verstovsek S, Talpaz M, Garcia-Manero G & Kantarjian
HM 2005 Phase II study of low-dose decitabine in patients
with chronic myelogenous leukemia resistant to imatinib
mesylate. Journal of Clinical Oncology 23 3948–3956.
Jarrard DF, Kinoshita H, Shi Y, Sandefur C, Hoff D,
Meisner LF, Chang C, Herman JG, Isaacs WB & Nassif N
1998 Methylation of the androgen receptor promoter CpG
island is associated with loss of androgen receptor
expression in prostate cancer cells. Cancer Research
58 5310–5314.
Kim KM & Lee YJ 2005 Amiloride augments TRAIL-
induced apoptotic death by inhibiting phosphorylation of
kinases and phosphatases associated with the P13K-Akt
pathway. Oncogene 24 355–366.
Kupferman ME, Patel V, Sriuranpong V, Amornphimoltham
P, Jasser SA, Mandal M, Zhou G, Wang J, Coombes K,
Multani A et al. 2007 Molecular analysis of anoikis
resistance in oral cavity squamous cell carcinoma. Oral
Oncology 43 440–454.
Lu Q, Qiu X, Hu N, Wen H, Su Y & Richardson BC 2006
Epigenetics, disease, and therapeutic interventions.
Ageing Research Reviews 5449–467.
McCabe MT, Low JA, Daignault S, Imperiale MJ, Wojno KJ
& Day ML 2006 Inhibition of DNA methyltransferase
activity prevents tumorigenesis in a mouse model of
prostate cancer. Cancer Research 66 385–392.
Mishra MV, Bisht KS, Sun L, Muldoon-Jacobs K, Awwad R,
Kaushal A, Nguyen P, Huang L, Pennington JD &
Markovina S 2008 DNMT1 as a molecular target in
a multimodality-resistant phenotype in tumor cells.
Molecular Cancer Research 6243–249.
Mottet D, Bellahce
`ne A, Pirotte S, Waltregny D, Deroanne C,
Lamour V, Lidereau R & Castronovo V 2007 Histone
deacetylase 7 silencing alters endothelial cell migration, a key
step in angiogenesis. Circulation Research 101 1237–1246.
Murphy TM, Perry AS & Lawler M 2008 The emergence of
DNA methylation as a key modulator of aberrant cell
death in prostate cancer. Endocrine-Related Cancer 15
11–25.
Nakabayashi M, Sartor O, Jacobus S, Regan MM,
McKearn D, Ross RW, Kantoff PW, Taplin ME &
Oh WK 2008 Response to docetaxel/carboplatin-based
chemotherapy as first- and second-line therapy in
patients with metastatic hormone-refractory prostate
cancer. BJU International 101 308–312.
Patel AR, Sandler HM & Pienta KJ 2005 Radiation Therapy
Oncology Group 0521: a phase III randomized trial of
androgen suppression and radiation therapy versus androgen
suppression and radiation therapy followed by chemotherapy
with docetaxel/prednisone for localized, high-risk prostate
cancer. Clinical Genitourinary Cancer 4212–214.
Pohlmann P,DiLeone LP, Cancella AI, Caldas AP, Dal Lago L,
Campos O Jr, Monego E, Rivoire W & Schwartsmann G
2002 Phase II trial of cisplatin plus decitabine, a new DNA
hypomethylating agent,in patients with advancedsquamous
cell carcinoma of the cervix. American Journal of Clinical
Oncology 25 496–501.
Plumb JA, Strathdee G, Sludden J, Kaye SB & Brown R 2000
Reversal of drug resistance in human tumor xenografts
by 20-deoxy-5-azacytidine-induced demethylation of
the hMLH1 gene promoter. Cancer Research 60
6039–6044.
Pulukuri SM & Rao JS 2005 Activation of p53/p21Waf1/
Cip1 pathway by 5-aza-20-deoxycytidine inhibits cell
proliferation, induces pro-apoptotic genes and
mitogen-activated protein kinases in human prostate
cancer cells. International Journal of Oncology 26
863–871.
Ross RW, Beer TM, Jacobus S, Bubley GJ, Taplin ME, Ryan
CW, Huang J & Oh WK 2008 Prostate cancer clinical trials
consortium. A phase 2 study of carboplatin plus docetaxelin
men with metastatic hormone-refractory prostate cancer
who are refractory to docetaxel. Cancer 112 521–526.
Schwartsmann G, Schunemann H, Gorini CN, Filho AF,
Garbino C, Sabini G, Muse I, DiLeone L & Mans DR
2000 A phase I trial of cisplatin plus decitabine, a new
DNA-hypomethylating agent, in patients with advanced
solid tumors and a follow-up early phase II evaluation in
patients with inoperable non-small cell lung cancer.
Investigational New Drugs 18 83–91.
Segura-Pacheco B, Perez-Cardenas E, Taja-Chayeb L,
Chavez-Blanco A, Revilla-Vazquez A, Benitez-
Bribiesca L & Duenas-Gonza
´lez A 2006 Global DNA
hypermethylation-associated cancer chemotherapy
resistance and its reversion with the demethylating agent
hydralazine. Journal of Translational Medicine 432.
Sonpavde G, Aparicio A, Guttierez I, Boehm KA, Hutson TE,
Berry WR, Asmar L & von Hoff DD 2007 Phase II study of
azacitidine to restore responsiveness of prostate cancer to
hormonal therapy. Clinical Genitourinary Cancer 5457–459.
Svatek RS, Lee JJ, Roehrborn CG, Lippman SM & Lotan Y
2008 Cost-effectiveness of prostate cancer chemopre-
vention: a quality of life-years analysis. Cancer 112
1058–1065.
Thakkar H, Chen X, Tyan F, Gim S, Robinson H, Lee C,
Pandey SK, Nwokorie C, Onwudiwe N & Srivastava RK
C Festuccia and G L Gravina et al.: Azacitidine effects in advanced prostate cancer models
www.endocrinology-journals.org412
2001 Pro-survival function of Akt/protein kinase B in
prostate cancer cells. Relationship with TRAIL resist-
ance. Journal Biological Chemistry 276 38361–38369.
Verhagen PC, van Duijn PW, Hermans KG, Looijenga LH,
van Gurp RJ, Stoop H, van der Kwast TH & Trapman J
2006 The PTEN gene in locally progressive prostate
cancer is preferentially inactivated by bi-allelic gene
deletion. Journal of Pathology 208 699–707.
Whang YE, Yuan XJ, Liu Y, Majumder S & Lewis TD 2004
Regulation of sensitivity to TRAIL by the PTEN tumor
suppressor. Vitamins and Hormone 67 409–426.
Xu J, Langefeld CD, Zheng SL, Gillanders EM, Chang BL,
Isaacs SD, Williams AH, Wiley KE, Dimitrov L, Meyers
DA et al. 2004 Interaction effect of PTEN and CDKN1B
chromosomal regions on prostate cancer linkage. Human
Genetics 115 255–262.
Zorn CS, Wojno KJ, McCabe MT, Kuefer R, Gschwend JE &
Day ML 2007 5-aza-20-deoxycytidine delays androgen-
independent disease and improves survival in the
transgenic adenocarcinoma of the mouse prostate mouse
model of prostate cancer. Clinical Cancer Research 13
2136–2143.
Endocrine-Related Cancer (2009) 16 401–413
www.endocrinology-journals.org 413
Article
Full-text available
Nowadays, prostate cancer is one of the most common forms of malignant neoplasms in men all over the world. Against the background of increasing incidence, there is a high mortality rate from prostate cancer, which is associated with an inadequate treatment strategy. Such a high prevalence of prostate cancer requires the development of methods that can ensure early detection of the disease, improve the effectiveness of treatment, and predict the therapeutic effect. Under these circumstances, it becomes crucial to focus on the development of effective diagnostic and therapeutic approaches. Due to the development of molecular genetic methods, a large number of studies have been accumulated on the role of epigenetic regulation of gene activity in cancer development, since it is epigenetic changes that can be detected at the earliest stages of cancer development. The presence of epigenetic aberrations in tumor tissue and correlations with drug resistance suggest new therapeutic approaches. Detection of epigenetic alterations such as CpG island methylation, histone modification, and microRNAs as biomarkers will improve the diagnosis of the disease, and the use of these strategies as targets for therapy will allow for greater personalization of prostate cancer treatment.
Article
Full-text available
Epigenetic modifications, such as DNA methylation, is widely studied in cancer. DNA methylation patterns have been shown to distinguish between benign and malignant tumors in various cancers, including prostate cancer. It may also contribute to oncogenesis, as it is frequently associated with downregulation of tumor suppressor genes. Aberrant patterns of DNA methylation, in particular the CpG island hypermethylator phenotype (CIMP), have shown associative evidence with distinct clinical features and outcomes, such as aggressive subtypes, higher Gleason score, prostate-specific antigen (PSA), and overall tumor stage, overall worse prognosis, as well as reduced survival. In prostate cancer, hypermethylation of specific genes is significantly different between tumor and normal tissues. Methylation patterns could distinguish between aggressive subtypes of prostate cancer, including neuroendocrine prostate cancer (NEPC) and castration resistant prostate adenocarcinoma. Further, DNA methylation is detectable in cell-free DNA (cfDNA) and is reflective of clinical outcome, making it a potential biomarker for prostate cancer. This review summarizes recent advances in understanding DNA methylation alterations in cancers with the focus on prostate cancer. We discuss the advanced methodology used for evaluating DNA methylation changes and the molecular regulators behind these changes. We also explore the clinical potential of DNA methylation as prostate cancer biomarkers and its potential for developing targeted treatment of CIMP subtype of prostate cancer.
Article
Discovering novel cancer therapies has attracted extreme interest in the last decade. In this regard, multidrug resistance (MDR) to chemotherapies is the primary challenge in cancer treatment. Cancerous cells are growingly become resistant to existing chemotherapeutics by employing diverse mechanisms, highlighting the significance of discovering approaches to overcome MDR. One promising strategy is utilizing aptamers as unique tools to target elements or signalings incorporated in resistance mechanisms or develop active targeted drug delivery systems or chimeras enabling the precise delivery of novel agents to inhibit the conventionally undruggable resistance elements. Further, due to their advantages over their proteinaceous counterparts, particularly antibodies, including improved targeting action, enhanced thermal stability, easier production, and superior tumor penetration, aptamers are emerging and have frequently been considered for developing cancer therapeutics. Here, we highlighted significant chemoresistance pathways and thoroughly discussed using aptamers as prospective tools to surmount cancer MDR.
Chapter
The term epigenetics dates back to the 1940s, when Conrad Waddington introduced it to refer to gene expression changes that occur during development and do not involve alterations in the DNA sequence. Subsequently the definition expanded beyond development, and the field became one of the most rapidly developing ones in life sciences. Advances in epigenetics transformed our understanding of cellular and molecular events that occur during development, homeostasis, and disease and helped explain processes that have long fascinated and puzzled scientists, such as the link between inflammation and disease, the intricacies of memory formation and maintenance, and the connection between the social environment/social adversity and chronic disease risk. Epigenetically mediated gene expression changes were described in a broad group of medical conditions, including cancer and neurodegenerative, metabolic, autoimmune, psychiatric, and cardiovascular diseases and, of these, the most advanced understanding of their contribution, so far, has occurred for cancer. Changes in DNA methylation, histone posttranslational modifications, and microRNA alterations, described in a broad group of human cancers, were implicated in all stages of carcinogenesis, including initiation, progression, invasion, and metastasis. The discovery of epigenetic biomarkers facilitated novel strategies for the early detection of disease, helped better monitor progression, therapeutic response, and prognosis, and revolutionized personalized medicine. Moreover, the reversible nature of epigenetic marks opened the possibility to therapeutically reverse aberrant gene expression patterns and catalyzed the emergence of epigenetic drugs. Besides their promise as monotherapies, epigenetic drugs show considerable interest thanks to the possibility to combine them with other cancer therapeutic modalities, such as chemotherapy, hormone therapy, and radiation therapy.KeywordsCancerDNA methylationEpigeneticsGene expressionHistone post-translational modificationsRNA interference
Chapter
Prostate cancer (PCa) is the second most common cancer worldwide with an estimated 1.3 million new cases in 2018 and 359,000 associated deaths, making it a leading cause of cancer-related deaths in men. Early detection of PCa allows for curative therapies such as surgery and radiation or, depending on disease aggressiveness, patient’s age and co-morbidities, active surveillance, which avoids overtreatment for men with indolent disease. Despite intense efforts to improve survival following androgen deprivation therapy and chemotherapy, virtually all metastatic patients still progress to the lethal castration resistant stage of the disease. Over the last decade, we have come to appreciate that epigenetic alterations play a central role in PCa development and progression, with the androgen receptor and its signaling pathway being heavily controlled through epigenetic modifications and engagement with various epigenetic enzymes. Here, we give an overview of the potential use of epigenetic therapies as an attractive avenue for the management of advanced PCa, as well as some preclinical evidence of the usefulness of epigenetic alterations as predictive biomarkers.
Article
Introduction: Neuroendocrine transdifferentiation (NED) of prostate cancer (PC) cells is associated with the development of resistance to antiandrogen therapy and poor prognosis in patients with castration-resistant PC (CRPC). Many of the molecular events, involved in NED, appear to be mediated by epigenetic mechanisms. In this study, we evaluated the antitumor activity and epigenetic modulation of 2 epigenetic drugs, such as the demethylating agent 5-aza-2'-deoxycytidine (AZA) and the methyl donor S-adenosylmethionine (SAM), in 2 human CRPC cell lines with NED (DU-145 and PC-3). Methods: The effects of AZA and SAM on cell viability, cell cycle, apoptosis, migration, and genome-wide DNA methylation profiling have been evaluated. Results: Both drugs showed a prominent antitumor activity in DU-145 and PC-3 cells, through perturbation of cell cycle progression, induction of apoptosis, and inhibition of cell migration. AZA and SAM reversed NED in DU-145 and PC-3, respectively. Moreover, AZA treatment modified DNA methylation pattern in DU-145 cells, sustaining a pervasive hypomethylation of the genome, with a relevant effect on several pathways involved in the regulation of cell proliferation, apoptosis, and cell migration, in particular Wnt/β-catenin. Conclusions: A relevant antitumor activity of these epigenetic drugs on CRPC cell lines with NED opens a new scenario in the therapy of this lethal variant of PC.
Chapter
DNA methylation is one of the most characterized epigenetic modifications, which plays a vital role in regulating gene transcription, chromatin remodeling, and genome stability. Aberrant DNA methylation promotes cancer progression and drug resistance. Inhibitors targeting DNA methyltransferases (DNMTs), the key catalytic enzymes in DNA methylation, have evidently shown therapeutic efficacy in many malignancies. Chemotherapy is one of the major treatment regimens for cancer patients. However, acquired drug resistance invariably hinders the clinical effect. Increasing evidence has shown that aberrant DNA methylation is involved in chemoresistance and DNMTis have the promising potential to reverse the chemoresistance. Here we summarize recent advances in DNMTis as novel sensitizers to overcome the resistance to chemotherapy.
Article
Background/aim: Cancers are selectively sensitive to methionine (MET) restriction (MR) due to their addiction to MET which is overused for elevated methylation reactions. MET addiction of cancer was discovered by us 45 years ago. MR of cancer results in depletion of S-adenosylmethionine (SAM) for transmethylation reactions, resulting in selective cancer-growth arrest in the late S/G2-phase of the cell cycle. The aim of the present study was to determine if blockade of the MET-methylation axis is a highly-effective strategy for cancer chemotherapy. Materials and methods: In the present study, we demonstrated the efficacy of MET-methylation-axis blockade using MR by oral-recombinant methioninase (o-rMETase) combined with decitabine (DAC), an inhibitor of DNA methylation, and an inhibitor of SAM synthesis, cycloleucine (CL). We determined a proof-of-concept of the efficacy of the MET-methylation-axis blockade on a recalcitrant undifferentiated/unclassified soft-tissue sarcoma (USTS) patient-derived orthotopic xenograft (PDOX) mouse model. Results: The o-rMETase-CL-DAC combination regressed the USTS PDOX with extensive cancer necrosis. Conclusion: The new concept of combination MET-methylation-axis blockade is effective and can now be tested on many types of recalcitrant cancer.
Article
Prostate cancer is a multifactorial disease that mainly occurs due to the accumulation of somatic, genetic and epigenetic changes, resulting in the inactivation of tumor-suppressor genes and activation of oncogenes. Mutations in genes, specifically those that control cell growth and division or the repair of damaged DNA, make the cells grow and divide uncontrollably to form a tumor. The risk of developing prostate cancer depends upon the gene that has undergone the mutation. Identifying such genetic risk factors for prostate cancer pose a challenge for the researchers. Besides genetic mutations, many epigenetic alterations including DNA methylation, histone modifications (methylation, acetylation, ubiquitylation, sumoylation, and phosphorylation) nucleosomal remodelling, and chromosomal looping, have been significantly contributed to the onset of prostate cancer as well as the prognosis, diagnosis, and treatment of prostate cancer. Chronic inflammation also plays a major role in the onset and progression of human cancer, via. modifications in the tumor microenvironment by initiating epithelial-mesenchymal transition and remodelling the extracellular matrix. In this article, the authors present a brief history of the mechanisms and potential links between the genetic aberrations, epigenetic changes, inflammation and inflammasomes that are known to contribute to the prognosis of prostate cancer. Furthermore, the authors examine and discuss clinical potential of prostate carcinogenesis in relation to epigenetics and inflammation for its diagnosis and treatment.
Article
Full-text available
The development of resistance to anti-cancer therapeutics remains one of the core issues preventing the improvement of survival rates in cancer. Therapy resistance can arise in a multitude of ways, including the accumulation of epigenetic alterations in cancer cells. By remodeling DNA methylation patterns or modifying histone proteins during oncogenesis, cancer cells reorient their epigenomic landscapes in order to aggressively resist anti-cancer therapy. To combat these chemoresistant effects, epigenetic modifiers such as DNA hypomethylating agents, histone deacetylase inhibitors, histone demethylase inhibitors, along with others have been used. While these modifiers have achieved moderate success when used either alone or in combination with one another, the most positive outcomes were achieved when they were used in conjunction with conventional anti-cancer therapies. Epigenome modifying drugs have succeeded in sensitizing cancer cells to anti-cancer therapy via a variety of mechanisms: disrupting pro-survival/anti-apoptotic signaling, restoring cell cycle control and preventing DNA damage repair, suppressing immune system evasion, regulating altered metabolism, disengaging pro-survival microenvironmental interactions and increasing protein expression for targeted therapies. In this review, we explore different mechanisms by which epigenetic modifiers induce sensitivity to anti-cancer therapies and encourage the further identification of the specific genes involved with sensitization to facilitate development of clinical trials.
Article
Full-text available
Tumor necrosis factor superfamily member TRAIL/Apo-2L has recently been shown to induce apoptosis in transformed and cancer cells. Some prostate cancer cells express constitutively active Akt/protein kinase B due to a complete loss of lipid phosphatase PTEN gene, a negative regulator of phosphatidylinositol 3-kinase pathway. Constitutively active Akt promotes cellular survival and resistance to chemotherapy and radiation. We have recently noticed that some human prostate cancer cells are resistant to TRAIL. We therefore examined the intracellular mechanisms of cellular resistance to TRAIL. The cell lines expressing the highest level of constitutively active Akt were more resistant to undergo apoptosis by TRAIL than those expressing the lowest level. Down-regulation of constitutively active Akt by phosphatidylinositol 3-kinase inhibitors, wortmannin and LY294002, reversed cellular resistance to TRAIL. Treatment of resistant cells with cycloheximide (a protein synthesis inhibitor) rendered cells sensitive to TRAIL. Transfecting dominant negative Akt decreased Akt activity and increased TRAIL-induced apoptosis in cells with high Akt activity. Conversely, transfecting constitutively active Akt into cells with low Akt activity increased Akt activity and attenuated TRAIL-induced apoptosis. Inhibition of TRAIL sensitivity occurs at the level of BID cleavage, as caspase-8 activity was not affected. Enforced expression of anti-apoptotic protein Bcl-2 or Bcl-X(L) inhibited TRAIL-induced mitochondrial dysfunction and apoptosis. We therefore identify Akt as a constitutively active kinase that promotes survival of prostate cancer cells and demonstrate that modulation of Akt activity, by pharmacological or genetic approaches, alters the cellular responsiveness to TRAIL. Thus, TRAIL in combination with agents that down-regulate Akt activity can be used to treat prostate cancer.
Article
Full-text available
A generalized method for analyzing the effects of multiple drugs and for determining summation, synergism and antagonism has been proposed. The derived, generalized equations are based on kinetic principles. The method is relatively simple and is not limited by 1) whether the dose-effect relationships are hyperbolic or sigmoidal, 2) whether the effects of the drugs are mutually exclusive or nonexclusive, 3) whether the ligand interactions are competitive, noncompetitive or uncompetitive, 4) whether the drugs are agonists or antagonists, or 5) the number of drugs involved.
Article
Full-text available
The authors describe a phase I trial of cisplatin plus decitabine, a novel DNA-hypomethylating agent, in patients with advanced solid tumors, which was followed by an early phase II evaluation of the combination in patients with inoperable non-small cell lung cancer (NSCLC). In the phase I trial, cisplatin was studied at a fixed dose of 33 mg/m2, while decitabine was escalated in four (I-IV) dose escalation levels (45, 67, 90 to 120 mg/m2, respectively) in consecutive groups of at least 3 patients per dose level. Decytabine was administered to the patients as a two-hour intravenous infusion, while cisplatin was given intravenously immediately after the end of decitabine infusion. Both agents were given on days 1-3 every 21 days. Twenty-one patients were included in the phase I trial. Dose level IV (120 mg/m2 decitabine) was considered the maximum tolerated dose (MTD), while the dose-limiting toxicities were neutropenia, thrombocytopenia and mucositis. The recommended doses for phase II trials in good- and poor-risk patients were 90 (level III) and 67 mg/m2 (level II), respectively. One short-lasting partial response was observed in a patient with cervical cancer, while two minor regression were documented in a patients with NSCLC and cervical cancer, respectively. Dose level II was selected for the phase II trial in patients with inoperable NSCLC. Fourteen consecutive patients were included in this part of the study. The median age of the patients was 57 years (range, 39-75), male/female ratio of 11/3 and a median WHO performance status 1 (0-2). The stage of disease were IIIB (5) and IV (9). Prior irradiation to the chest was given in one case. A total of 30 treatment courses were evaluable for toxicity and response, with a median of 2 courses per patient (1-4). Grade 3-4 neutropenia and thrombocytopenia were observed in about half of the cases. Mucositis, diarrhea, nausea and vomiting, and skin rash were also observed in some patients. Three minor responses were documented, which lasted for 4, 16 and 36 weeks. Median survival of patients was 15 weeks (4-38). In conclusion, the cisplatin plus decitabine combination did not exhibit significant antitumor activity in patients with NSCLC at the dose and schedule applied in this trial to justify its further evaluation in this patient population.
Article
Full-text available
Loss of DNA mismatch repair because of hypermethylation of the hMLH1 gene promoter occurs at a high frequency in a number of human tumors. A role for loss of mismatch repair (MMR) in resistance to a number of clinically important anticancer drugs has been shown. We have investigated whether the demethylating agent 2'-deoxy-5-azacytidine (DAC) can be used in vivo to sensitize MMR-deficient, drug-resistant ovarian (A2780/cp70) and colon (SW48) tumor xenografts that are MLH1 negative because of gene promoter hypermethylation. Treatment of tumor-bearing mice with the demethylating agent DAC at a nontoxic dose induces MLH1 expression. Re-expression of MLH1 is associated with a decrease in hMLH1 gene promoter methylation. DAC treatment alone has no effect on the growth rate of the tumors. However, DAC treatment sensitizes the xenografts to cisplatin, carboplatin, temozolomide, and epirubicin. Sensitization is comparable with that obtained by reintroduction of the hMLH1 gene by chromosome 3 transfer. Consistent with loss of MMR having no effect on sensitivity in vitro to Taxol, DAC treatment has no effect on the Taxol sensitivity of the xenografts. DAC treatment does not sensitize xenografts of HCT116, which lacks MMR because of hMLH1 mutation. Because there is emerging data on the role of loss of MMR in clinical drug resistance, DAC could have a role in increasing the efficacy of chemotherapy for patients whose tumors lack MLH1 expression because of hMLH1 promoter methylation.
Article
We have previously shown that 5-aza-2'-deoxycytidine (5-aza) is an effective chemopreventive agent capable of preventing early disease progression in the transgenic adenocarcinoma of the mouse prostate (TRAMP) model. The purpose of this study was to determine the effect of 5-aza on preexisting TRAMP prostate cancers and prevention of androgen-independent prostate cancer. TRAMP mice with established prostate cancers were treated with 5-aza, castration, castration + 5-aza, or vehicle control (PBS). One cohort of 22 mice per treatment was euthanized after 10 weeks of treatment, whereas a second cohort of 14 mice per group was followed until death to determine survival. Histologic sections of prostate, pelvic lymph nodes, lung, and liver were blinded and analyzed by a certified genitourinary pathologist (K.J.W.). Combined treatment (castration + 5-aza) provided significant survival benefits over either single treatment (combined versus castration P = 0.029, combined versus 5-aza P = 0.036). At 24 weeks of age, 86% of mice within the PBS cohort exhibited histologic evidence of prostate cancer, whereas only 47% of the combined cohort exhibited malignant disease (P < 0.0001). Additionally, whereas 43% of the PBS treatment group exhibited lymph node metastases, these were only observed in 21% of the combined treatment mice. This is the first study to examine the effect of 5-aza and combined castration + 5-aza on preexisting prostate cancer in an animal model. Based on these preclinical findings, we suggest that 5-aza treatment may prolong the time to an androgen-independent status and thus survival in a hormone-deprived setting in prostate cancer.
Article
Myelodysplastic syndromes (MDS) are a group of clonal disturbances with defective cellular differentiation. Vitamin D3 (VD) analogues can act on the differentiation and maturity of different cell lines. We studied the effects of VD on a series of patients with MDS in an open-design trial. Nineteen patients, 12 men and seven women, with MDS were included. Patients were 74.8 +/- 5.6 years (mean +/- SD), seven had refractory anaemia with ringed sideroblasts, five had refractory anaemia, one had refractory anaemia with excess of blasts and six had chronic myelomonocytic leukaemia. All the patients were in a low to intermediate risk group. Mean follow-up period was 26.21 months, range 9-75. Responders were defined as follows: granulocyte or platelet count increase by 50%, or haemoglobin increase of 1.5 g/dl or transfusion needs decrease by 50%. The first five patients received 266 microg of calcifediol three times a week and the other 14 received calcitriol (0.25-0.75 microg/d). Response was observed in 11 patients. In the calcifediol-treated group, one case responded, three were nonresponders, and one showed progression. In the calcitriol group, 10 were responders (two with major response), and four were non-responders. No correlation was observed between baseline levels of vitamin D metabolites and the presence of response. No hypercalcaemia was observed. Treatment with vitamin D3 metabolites could induce a long-standing response of the haematological disturbance in some low-intermediate risk MDS patients without inducing hypercalcaemia.
Article
Androgen-independent metastatic prostate cancer is characterized by a heterogeneous loss of androgen receptor (AR) expression among tumor cells. In this study, we evaluate DNA hypermethylation as a potential transcriptional regulatory mechanism in AR-negative prostate cancer cell lines. Nucleotide sequence analysis demonstrates an approximately 15-kb CpG island in the AR gene that encompasses the transcription start site and exon 1. Using Southern blotting with methylation-sensitive restriction enzymes and methylation-specific PCR, we find aberrant methylation in the AR expression-negative cell lines Du145, DuPro, TSU-PR1, and PPC1. Incomplete methylation in the AR CpG island is also seen in normal female breast and ovarian tissues consistent with the inactivation of one X chromosome by hypermethylation. In contrast, prostate cancer cell lines LNCaP and PC3 express AR and are unmethylated. Normal prostate epithelial cell strains demonstrate no methylation. Exposure of AR-negative prostate cancer cell lines to 5-aza-2' deoxycytidine, a demethylating agent, induces the reexpression of AR RNA in DuPro and TSU-PR1. This reexpression is associated with a demethylation of this region. Prostate-specific antigen, an androgen-responsive gene, is also specifically induced in these lines after AR reexpression. Therefore, in vitro DNA methylation of the 5' CpG AR island may be associated with the loss of AR expression. Furthermore, our results demonstrate that treatment with demethylating agents may engender the reexpression and function of the androgen receptor in AR-negative cell lines.
Article
The androgen resistance of some prostate cancer patients may be due to transcriptional inactivation of the androgen receptor (AR) gene catalyzed by cytosine DNA methyltransferase. To determine if an inhibitor of cytosine DNA methyltransferase, 5,6-dihydro-5'-azacytidine (DHAC), can restore the androgen sensitivity in androgen-insensitive human prostate carcinoma cell lines in vitro, we cultured androgen-insensitive (PC3, DU-145, and TSUPrl) and androgen-responsive (LNCaP) cells with subcytotoxic concentrations (< or = IC50) of DHAC for 14 days followed by exposure to dihydrotestosterone (DHT) or to hydroxyflutamide for 7 days. Only DHAC-treated DU-145 cells showed growth stimulation by 10(-11) to 10(-9) M DHT and a partial inhibition by 10(-5) and 10(-6) M hydroxyflutamide. However, since DU-145 is the only cell line tested that is known to have a hypermethylated AR promoter, the observed effects may be due to a partial demethylation of the AR by DHAC. Our data provide an evidence that cytosine DNA methyltransferase inhibitors can restore androgen responsiveness in androgen-refractory tumor cells, which are then sensitive to growth inhibition by antiandrogens.
Article
Epigenetic inactivation of genes that are crucial for the control of normal cell growth is a hallmark of cancer cells. These epigenetic mechanisms include crosstalk between DNA methylation, histone modification and other components of chromatin higher-order structure, and lead to the regulation of gene transcription. Re-expression of genes epigenetically inactivated can result in the suppression of tumour growth or sensitization to other anticancer therapies. Small molecules that reverse epigenetic inactivation are now undergoing clinical trials in cancer patients. This, together with epigenomic analysis of chromatin alterations such as DNA methylation and histone acetylation, opens up the potential both to define epigenetic patterns of gene inactivation in tumours and to use drugs that target epigenetic silencing.
Article
Genetic alterations and/or deletion of the tumor suppressor gene PTEN/MMAC/TEP1 occur in many types of human cancer including prostate cancer. We describe the production of monoclonal antibody against recombinant human PTEN and the study of PTEN gene and protein expression in three commercially available human prostate cancer cell lines, PC-3, LNCaP, and DU 145. Northern blotting analyses showed that LNCaP and DU145 but not PC-3 cells expressed PTEN mRNA. However, Western blotting analyses using a monoclonal antibody against PTEN demonstrated the expression of PTEN protein in DU145 but not LNCaP cells. In DU 145 cells, PTEN expression at both the mRNA and protein levels inversely correlated with serum concentrations and levels of PKB/Akt phosphorylation. In addition, the basal activity of PKB/Akt as indicated by level of phosphorylation was higher in prostate cancer cells which do not express PTEN than that in the cells expressing wild type PTEN. Thus, PTEN may play a critical role in regulating cellular signaling in prostate cancer cells.