ArticlePDF AvailableLiterature Review

Molecular Biomarker in Prostate Cancer: The Role of CpG Island Hypermethylation

Authors:

Abstract and Figures

CpG island hypermethylation may be one of the earliest somatic genome alterations to occur during the development of multiple cancers. Recently, aberrant methylation patterns for different tumors have been reported. We present a comprehensive review of the literature describing the role of CpG island hypermethylation of DNA from prostatic tissue and bodily fluids from men with prostate cancer. We reviewed the literature to evaluate CpG island hypermethylation in tissue and bodily fluids of men with primary and metastatic prostate cancer. Additionally, we reviewed the literature with respect to CpG island hypermethylation patterns in other urological malignancies. Using modern analytic methods, CpG island hypermethylation detection can be achieved. In men with prostate cancer, correlations between specific gene regulatory region hypermethylation analyses and Gleason score, pathologic stage and tumor recurrence have been demonstrated. CpG island hypermethylation may serve as a useful molecular biomarker for the detection and diagnosis of patients with prostate cancer.
Content may be subject to copyright.
Review
Molecular Biomarker in Prostate Cancer: The Role
of CpG Island Hypermethylation
Patrick J. Bastian
a,b,1
, Srinivasan Yegnasubramanian
c
, Ganesh S. Palapattu
a
,
Craig G. Rogers
a
, Xiaohui Lin
b,d
, Angelo M. De Marzo
a,b,d,e
, William G. Nelson
a,b,c,d,e,
*
a
The James Buchanan Brady Urological Institute, Department of Urology, The Johns Hopkins University School of Medicine,
Baltimore, MD 21231-1000, USA
b
Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
c
Department of Pharmacology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
d
The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
e
Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
Accepted 29 July 2004
Available online 1 September 2004
Abstract
CpG island hypermethylation may be one of the earliest somatic genome alterations to occur during the
development of multiple cancers. Recently, aberrant methylation patterns for different tumors have been
reported. We present a comprehensive review of the literature describing the role of CpG island hypermethlytion
of DNA from prostatic tissue and bodily fluids from men with prostate cancer. We reviewed the literature to
evaluate CpG island hypermethylation in tissue and bodily fluids of men with primary and metastatic prostate
cancer. Additionally, we reviewed the literature with respect to CpG island hypermethylation patterns in other
urological malignancies.Using modern analytic methods, CpG island hypermethylation detection can be
achieved. In men with prostate cancer, correlations between specific gene regulatory region hypermethylation
analyses and Gleason score, pathologic stage and tumor recurrence have been demonstrated. CpG island
hypermethylation may serve as a useful molecular biomarker for the detection and diagnosis of patients with
prostate cancer.
#2004 Published by Elsevier B.V.
Keywords: Prostate cancer; DNA methylation; Hypermethylation; CpG island; GSTP1;
Gluthatione S-transferase; Epigenetics
1. I n t r o d u c t i o n
Prostate cancer is the most common serious cancer
in men and the second leading cause of cancer related
deaths in the United States and Western Europe. Since
the beginning of the prostate specific antigen (PSA)
testing era in the early 1990s, the number of men with
non-palpable prostate cancer has significantly
increased [1,2]. Autopsy studies and the recent Prostate
Cancer Prevention Trial (PCPT) have revealed a higher
prevalence of prostatic cancer than anticipated by PSA
screening alone [3–5]. The lifetime risk of developing
prostate cancer is 1 in 6, whereas the lifetime risk of
death due to metastatic prostate cancer is 1 in 30 [6].It
is estimated that 230,110 new cases will be diagnosed
in 2004 and 29,900 men will die of the disease in the
US in 2004 [6].
Because the prevalence of diagnosed prostate cancer
has increased dramatically during the PSA era, many
European
Urology European Urology 46 (2004) 698–708
* Corresponding author. Tel. +1 410 614 1661; Fax: +1 410 502 9817.
E-mail address: patrick.bastian@ukb.uni-bonn.de (P.J. Bastian),
bnelson@jhmi.edu (W.G. Nelson).
1
Co-corresponding author. Klinik und Poliklinik fur Urologie, Univer-
sitatsklinikum Bonn, Rheinische Friedrich-Wilhelms Universitat Bonn,
Sigmund-Freud-Str. 25, 53129 Bonn, Germany. Tel. +49 228 287 5109;
Fax: +49 228 287 4285.
0302-2838/$ – see front matter #2004 Published by Elsevier B.V.
doi:10.1016/j.eururo.2004.07.022
clinicians feared that widespread use of PSA testing for
prostate cancer screening might detect a large number
of clinically insignicant tumors, thus resulting in
potentially unnecessary treatment [7,8]. PSA screen-
ing, regardless of the threshold value, has certain well
documented limitations with regards to sensitivity and
specicity for the detection of prostate cancer [9].
Ultrasound-guided prostate biopsy is the gold standard
method for the diagnosis of prostate cancer. The pre-
cise biopsy strategy in terms of number of samples and
sampled area, however, remains controversial [10,11].
Moreover, non-diagnostic, yet clinically-suspicious,
lesions, such as a small focus of atypical glands (also
referred to as ASAP or atypical, small acinar prolif-
eration) or high grade prostatic intra-epithelial neopla-
sia, necessitate further evaluation in the absence of
obvious cancer in the specimen [12]. Molecular studies
have revealed important information about prostate
cancer development and progression. Multiple immu-
nohistochemistry tools to aid the diagnosis of prostate
cancer have been developed, but as of yet, none of these
alone or in conjunction have been able to denitively
diagnose prostate cancer [1317]. Clearly, more sen-
sitive and specic biomarkers for prostate cancer diag-
nosis would be useful.
Epigenetic alterations, i.e., alterations in gene
expression without changes in the DNA sequence, in
human cancer were rst described in 1983 [18].
Somatic epigenetic genome changes include global
genomic hypomethylation, promotor hypermethyla-
tion of CpG islands and loss of imprinting. The most
common somatic genome alteration during prostate
cancer development appears to be the hypermethyla-
tion in the regulatory region of certain genes, most
commonly in the promoter of the p-class glutathione-
S-transferase (GSTP1) gene [19,20]. Genomic imprint-
ing is an epigenetic alteration in the zygote or gamete
that causes expression of a specic parental allele of a
gene in somatic cells of the offspring. Loss of imprint-
ing can involve activation of the normally silent allele
of a growth promoting gene, or silencing of the nor-
mally expressed allele of a growth inhibitory gene. In
this review we describe the potential use of CpG island
hypermethylation as a molecular marker for prostate
cancer screening, detection and diagnosis.
2. DNA hypermethylation
The dinucleotide sequence CpG often carries the
modied base 5-methyl-cytosine (
5-m
C).
5-m
C can be
maintained in the genome through DNA replication via
the activity of DNA methyltransferases. CpG islands
are clusters (400 to 2000 bp) of CpG dinucleotides
without
5-m
C modications that are present at the
transcriptional regulatory regions of many genes
[21]. Aberrant methylation of CpG islands are now
recognized to be one of the most common soma-
tic alterations in human cancers [2224]. These
potentially reversible epigenetic changes inhibit trans-
cription and lead to gene silencing. DNA methyltrans-
ferases (DNMTs),
5-m
C-binding proteins (MBD) and
histone deactylases couple DNA methylation and tran-
scriptional repression [2224]. In general, only methy-
lation of CpG islands within or surrounding the
transcriptional promotor region is associated with gene
silencing in cancer. That is to say, methylation in other
regions of genes, even within CpG island like
sequences, does not inhibit transcription [24]. Typi-
cally, transcriptional repression occurs as a consec-
quence of the assembly of a repressive chromation
structure containing MBDs, histone deacetylaces,
and other proteins, preventing DNA polymerase at
hypermethylated transcriptional promoter regions
[25].
3. Detection of hypermethylation
As DNA markers of cancer, CpG island hyper-
methylation changes have the advantage of being more
stable than RNA or proteins. Moreover, hypermethyla-
tion changes are common in many types of cancer.
Furthermore, specic CpG island hypermethylation
proles can be observed in several different types of
cancer [26]. Because the position of CpG islands
within regulatory regions is similar across most
patients, detection of hypermethylation at CpG islands
is simpler than that of mutations in cancer. Also,
because of this commonality, a single assay can be
used for detection in all patients [22].
Currently, two major strategies for detection of CpG
island hypermethylation are employed, although new
technologies are constantly emerging. The rst detec-
tion method uses
5-m
C-sensitive restriction enzymes
(Fig. 1), while the second approach uses sodium
bisulte conversion (Fig. 2) to detect CpG island
hypermethylation.
5-m
C-sensitive restriction enzyme
detection assays include Southern blot (SB) analysis
and polymerase chain reaction amplication of DNA
(RE-PCR). Bisulte conversion based assays include
bisulte genomic sequencing (BGS) [27] and bisulte
modication of DNA followed by either selective
polymerase chain reaction amplication (MSP) [28]
or quantative, real-time methylation specic polymer-
ase chain reaction (Q-MSP) [29]. Currently, the SB
P.J. Bastian et al./ European Urology 46 (2004) 698–708 699
analysis has been replaced with more sensitive tests.
The RE-PCR is a very sensitive technique in detecting
single hypermethylated alleles [30], employing treat-
ment with a restriction endonuclease that selectively
cuts its recognition site only if it does not contain
5-m
C
prior to PCR amplication. If the CpG island at the
restriction enzyme recognition site does not contain
a
5-m
C, the DNA is cut and no product can be detected
after PCR amplication. False positive detection of
CpG island hypermethylation with this method may be
the result of incomplete digestion of unmethylated
CpG island targets by restriction enzymes. Currently,
the most common approach is MS-PCR [31]. In this
technique, genomic DNA is rst treated with bisulte,
which deaminates unmethylated C bases to produce U
bases while
5-m
C in CpG islands remains unchanged.
This initial steps converts methylation patterns into
DNA sequence alterations that can be detected with
specically designed PCR primers. One drawback of
this technique is limited sensitivity, as bisulte treat-
ment can damage DNA and yield to inefcient PCR
amplication. BGS is a very labor intensive test and
due to limited sensitivity in detecting CpG island
hypermethylation it is not under development as a
clinical test [32].
4. Hypermethylation in prostate cancer
4.1. CpG island hypermethylation at GSTP1
Multiple studies assessing the CpG island hyper-
methylation status in prostate cancer and other human
cancers have been recently reported. The rst study of
CpG island hypermethylation in prostate cancer found
signicant CpG island hypermethylation at the GSTP1
promotor region [19]. Since then, multiple studies have
reported on the hypermethylation of the GSTP1 reg-
ulatory region in prostate cancer (Table 1 and Fig. 4).
The GSTP1 hypermethyltion status has since been
investigated in 1071 cases from a total of 24 studies
and has been shown to be hypermethylated in over 81%
of the cases examined. Although most studies used the
same primers, there are differences in the sensitivity
(Fig. 2). This may be due to differences in the tissue
processing and assay conditions. GSTP1 encodes for
the p-class Glutathione S-transferase (GST-p). GSTs
P.J. Bastian et al. / European Urology 46 (2004) 698708700
Fig. 1. Detection of GSTP1 hypermethylation in prostate cancer with
restriction enzyme based assays [19,30,49,52,64]. On top, the regulatory
region of the GSTP1 gene is shown in red from base pair 851 to +239
relative to the transcriptional start site (represented by a green arrow).
Vertical black bars indicate CpG dinucleotides. For Southern blot assay the
restriction enzyme cutting site is shown as a pink vertical bar (base pair
562 and 564) within the southern blot probe. For restriction enzymes
PCR assays, the PCR amplicon is shown along with all restriction enzyme
cutting sites (vertical pink lines) within the amplicon. Fig. 2. Detection of GSTP1 hypermethylation in prostate cancer with
bisulte treatment based assays [3741,45,48,57,6164,6669,71,
7286,88102]. On top, the regulatory region of the GSTP1 gene is shown
in red from base pair 851 to +239 relative to the transcriptional start site
(represented by a green arrow). Vertical black bars indicate CpG dinucleo-
tides. For bisulte genomic sequencing, the amplicons are indicated as
horizontal black lines. For methylation specic PCR arrows indicate the
primer annealing sites. For real time methylation specic PCR, the taqman
probe hybridization site is shown between the specic primers.
are an enzyme family that can detoxify reactive che-
mical species by catalyzing their conjugation to
reduced glutathione [33]. Thus, GSTP1 likely serves
as a ‘‘caretaker’’ gene [34], defending prostate cells
against genomic damage mediated by carcinogens or
various oxidants [20,35,36]. Loss of GSTP1 function
may render prostatic cells sensitive to carcinogenesis
driven by inammation and diet.
4.2. CpG island hypermethylation at various loci
Using all of the currently available techniques to
detect DNA hypermethylation, 45 genes have been
examined in prostate cancer tissue (Table 1). Most
reports have analyzed one gene at a time, but more
recent studies have assayed several genes simulta-
neously and have identied prostate cancer specic
gene hypermethylation proles [3741]. Combining
the literature on CpG island hypermethlyation in pros-
tate cancer with that from data on similar studies from
bladder and renal cancer, a prole that distinguishes
prostate cancer from these other genito-urinary cancers
can be formulated (Fig. 3).
A recent comphrehensive study by Yegansubrama-
nian et al. assesses the extent of hypermethylation in 16
different genes in prostate cancer. Using the quantita-
tive, real-time MSP the authors noted that using various
combinations of GSTP1,APC,RASSF1a,PTGS2 and
MDR1 CpG island hypermethylation can distinguish
primary prostate cancer from benign prostate tissue
with sensitivities of 97.3%100% and a specicity of
92%100% [40]. In contrast to most other studies,
benign prostate tissues were obtained from brain-dead
transplant tissue donors. Although not age-matched to
the study cohort, these tissue samples resembled
healthy prostate upon histopathological analysis. Most
studies use normal prostate tissue adjacent to prostate
cancer as controls, leading to the yet unsolved question
whether normal tissue adjacent to cancer is, although
histopathologically benign, the same in terms of certain
molecular changes.
4.3. CpG island hypermethylation in non-cancerous
tissue
In addition to being methylated in prostatic cancer
tissue, some genes are also methylated in benign
prostatic tissue or benign prostatic hyperplasia
(BPH) (Fig. 4). In non-cancerous prostate tissues,
various groups have reported methylation of CpG
islands at EDNRB (up to 91%) [42],HIC (up to
100%) [38,40],ER (up to 60%80%) [43,44] and
RASSF1a. However, other groups have reported that
methylation at the CpG islands of these genes is highly
P.J. Bastian et al. / European Urology 46 (2004) 698708 701
Fig. 3. A comparison of the overall frequency of hypermethylation at various CpG islands in prostate cancer [19,30,3741,45,49,52,57,61,6371,7286,
88102], renal cell carcinoma [69,103118] and urothelial cancer of the urinary bladder [26,119131]. The overall frequency of methylation at each CpG island
was calculated by dividing the total number of cases studied in the literature by the total number of cases that were methylated at each CpG island. This
comparison suggests that a hypermethylation prole can uniquely identify each urological malignancy.
specic for cancerous tissue and did not nd any
methylation at these genes in benign tissues. These
observed differences in the frequency of CpG island
methylation in benign tissues is probably due to the
differences in the number of CpG dinucleotides inter-
rogated as well as their genomic position. For instance,
Jeronimo et al., using an MSP assay with primers
containing 6 CpG dinucleotides that amplied a region
that was 139 to 9 relative to the translational start
site, found that 91% of benign tissues were methylated
at the EDNRB CpG island [42]. In contrast, Yegnasu-
bramanian et al., using a real time MSP assay with
primers and probe containing 9 CpG dinucleotides that
amplied a region that was 271 to 122 relative to
P.J. Bastian et al. / European Urology 46 (2004) 698708702
Ta b l e 1
A review of hypermethylated genes in prostate cancer tissue
Gene Number
of studies
Number
of samples
a
Number of
hypermethylated samples
b
Overall
frequency (%)
c
Range
(%)
d
References
GSTP1 22 1071 874 81.61 36100 [19,30,3741,45,49,52,57,61,6371]
CDH1 6 367 61 16.62 0100 [37,40,41,63,72,73]
MGMT 5 352 39 11.08 088 [3741]
DAPK 4 320 44 13.75 036 [37,38,40,41]
p16/CDKN2a 8 313 50 15.97 066 [37,3941,7477]
RASSF1a 9 274 199 72.63 5396 [37,40,41,78,79]
RARbeta 3 234 159 67.95 5379 [37,38,80]
APC 3 211 114 54.03 2790 [37,40,41]
CD44 4 191 94 49.21 3277.5 [62,8183]
HIC1 2 182 181 99.45 99100 [38,40]
TIMP3 2 182 6 3.3 06[38,40]
AR 5 181 25 13.81 015 [38,40,8486]
p14 4 158 6 3.8 010.81 [3941,74]
EDNRB 4 147 96 65.31 49100 [40,42,87,88]
PTGS2 2 110 72 65.45 21.6288 [40,41]
Cyclin D2 1 101 32 31.68 n/a [89]
CDH13 1 101 31 30.69 n/a [37]
FHIT 1 101 15 14.85 n/a [37]
MDR1 1 73 64 88 n/a [40]
ESR1 1 73 14 19.18 n/a [40]
p15/CDKN2b 2730 0 0[40]
hMLH1 1 73 0 0 n/a [40]
ER alpha A 2 70 65 92.86 9095 [85,90]
ER alpha B 2 70 64 91.43 9092 [85,90]
PR-B 2700 0 0[85,90]
PR-A 2700 0 0[85,90]
ER alpha C 2700 0 0[85,90]
VEGFR1 1 63 24 38.1 n/a [91]
ER beta 2 61 53 86.89 79100 [44,85]
P27 3 61 10 16.39 638 [39,74,92]
TIG1 1 50 26 52 n/a [93]
RB1 2 48 4 8.33 612.5 [39,74]
P21 2 48 4 8.33 612.5 [39,74]
RUNX3 1 37 1 40.54 n/a [41]
THBS1 1 37 10 27.03 n/a [41]
TNFRSF6 1 32 4 12.5 n/a [94]
ER 1 31 28 90.32 n/a [43]
17p 1 26 25 96 n/a [95]
ZNF185 1 25 11 44 n/a [96]
Inhibin alpha subunit 1 24 7 29.17 n/a [97]
Caveolin-1 1 22 20 90.91 n/a [98]
TSLC1 1 22 7 31.82 n/a [99]
NEP 1 2 3 14.29 n/a [100]
PTEN 1 16 0 0 n/a [74]
P73 1 16 0 0 n/a [74]
p16/p14 1 11 8 72.73 n/a [76]
a
Sum of number of cancerous samples across all studies.
b
Sum of number of cancerous samples that were hypermthylated at each CpG island across all studies.
c
Overall frequency calculated by diversion of number of hypermethylated samples from the toal number of samples.
d
The minimum to maximum frequency reported by all studoes at each CpG island; n/a: not available.
the translational start site, reported that none of the
benign tissues were methylated at the EDNRB CpG
island [40]. Therefore, it appears that the number and
genomic position of CpG dinucleotides interrogated in
the MSP assay may signicantly affect the frequency
of methylation in various prostate tissues.
4.4. Correlation with clinical parameters
In addition, aberrant methylation at EDNRB has
been correlated with Gleason grade and pathological
stage of prostate cancer [40]. The gene encoding for
COX2, PTGS2, has been found to be methylated in
88% of cases and has been correlated with an increased
risk of PSA recurrence independently of Gleason score
or pathological stage [40]. Recently, Kang et al. [41]
described a correlation between hypermethylation at
APC,RASSF1a and RUNX3 with characteristics (PSA
value and Gleason score) associated with a poor prog-
nosis. These studies suggest that assessment of methy-
lation status may be useful as a prognostic marker for
prostate cancer.
4.5. CpG island hypermethylation in precursor
lesions
One explanation for why CpG island hypermethyla-
tion has been observed in non-neoplastic tissue may be
that the examined tissues also contained prostate cancer
or prostatic intraepithelial neoplasia (PIN) cells. To
address this problem, Nakayama et al. used laser cap-
ture microdissection (LCM) to selectively recover cells
from normal prostatic epithelium, PIN, prostate cancer
and proliferative inammatory atrophy (PIA) lesions
[45]. PIA has been implicated to play a role in prostate
carcinogenesis as a potential precursor to PIN and
prostate cancer [46]. Nakayama et al. found GSTP1
hypermethylation in 6% of the PIA lesions, 69% of the
PIN lesions and 91% of the prostate cancer specimens.
Importantly, no hypermethylation was reported in nor-
mal prostate epithelium or benign prostatic hyperplasia
despite the fact these tissue were microdissected from
cancer containing prostates [45] (Fig. 4).
4.6. CpG island hypermethylation in biopsy tissue
The ultimate proof of a diagnosis of prostate cancer
is the needle core biopsy. Although the biopsy is widely
used and a standard urological procedure, no standard
has been set in terms of sampling number of cores and
the regions of the prostate to be biopsied [47]. False
negative results, possibly from sampling error, can
further cloud patient management. In addition, the
diagnosis of prostate cancer from biopsy material
can be challenging for pathologists, as several entities
can imitate the histological appearance of prostate
cancer histologically [32]. To circumvent these issues,
Harden et al. (Q-MS-PCR) and Chu et al. (RE-PCR)
have examined the use of CpG island hypermethylation
at GSTP1 in small tissue samples obtained by prostate
biopsy [48,49]. They found that by using PCR
based detection techniques they were able to clearly
distinguish neoplastic prostate from non-neoplastic
prostate tissue. In another report, Goessl et al. used
MSP to detect GSTP1 CpG island hypermethylation
from biopsy needle washes and was able to detect it in
70% of the prostate cancer cases surveyed [50]. Gon-
P.J. Bastian et al. / European Urology 46 (2004) 698708 703
Fig. 4. A comparison of the overall frequence of CpG island hpermethylation in normal prostate, benign prostatic hyperplasia (BPH), prostatic intraepithelial
neoplasia (PIN)and prostate cancer (PCA) [19,30,3741,45,49,52,57,61,6386,88102]. The overall frequency was determined as described in the legendof Fig. 3.
zalgo et al. examined urine collected after prostate
biopsy and detected GSTP1 hypermethylation in 50%
of the patients with histologically proven prostate
cancer [51]. In this study 33% of patients without
prostate cancer or PIN also exhibited GSPT1 hyper-
methylation in post-biopsy urine DNA [51]. Interest-
ingly, two patients with a negative initial biopsy and
positive post-biopsy urine analysis for GSTP1 hyper-
methylation had a subsequent biopsy that was positive
for prostate cancer.
4.7. CpG island hypermethylation in metastatic
disease
Relatively few studies have examined the extent of
aberrant hypermethylation in metastatic sites of pros-
tate cancer [40,52,53]. In one such report, Yegnasubra-
manian et al. examined prostate cancer specimens that
included metastatic disease to multiple regions such as
lymph nodes, lung, liver and bone and found that the
hypermethylation pattern in prostate cancer metastases
mimicked that of the primary tumor and tended to show
greater differences within patients with multiple sites of
metastases than within sites across methylated patients
[40].Ko
¨llermann et al. studied lymph nodes obtained
during standard radical prostatectomy and pelvic lym-
phadenectomy for organ-conned prostate cancer in
patients with subsequent PSA relapse [53]. Although all
of these men had histologically benign lymph nodes,
GSTP1 hypermethylation was detected in 90% of the
lymph nodes at the time of surgery.
5. Hypermethylation in bodily fluids of
prostate cancer patients
For any markerto become clinically useful, it must be
present and detetctable in easily accessible sites such as
peripheral blood,urine, ejaculate or prostatic secretions.
To date, only CpG island hypermethylation of the
GSTP1 promotor has been examined in a small number
of studies in bodily uids from patients with prostate
cancer (Table 2). Peripheral blood specimens are easy
available and are a part of current prostate cancer
screening and detection modalities. Although cell-free
circulating DNA has been detected in the plasma and
serum as early as 1948 [54], circulating DNA in the
plasma and serum of patients with urological malig-
nancies was only recently described [55]. Aside from
free circulating prostate DNA, prostate cancer DNA
may be present in the circulation as a result of intravas-
cular cell death of prostate cancer cells or circulating
phagocytic cells that have ingested prostate cancer cells
P.J. Bastian et al. / European Urology 46 (2004) 698708704
Ta b l e 2
Studies of GSTP1 hypermethylation in bodily uids of prostate cancer patients
Study Detection method
a
Frequency (%) Reference
Normal BPH
b
PIN
c
PCA
d
Urine
Carins et al. MS-PCR 27 [61]
Goessl et al. MS-PCR 2 29 73 [62]
Goessl et al. MS-PCR 3 76 [58]
Jeronimo et al. MS-PCR 3.2 30 [56]
Gonzalgo et al. MS-PCR (after biopsy) 33 67 58 [51]
Ejaculate
Goessl et al. MS-PCR 0 50 [58]
Suh et al. RE-PCR 44 [60]
Plasma
Goessl et al. MS-PCR 0 72 [57]
Goessl et al. MS-PCR 0 56 (T2-3) [58]
Goessl et al. MS-PCR 0 93 (T4 or mets) [58]
Jeronimo et al. MS-PCR 0 36 [56]
Prostatic secretion
Gonzalgo et al. MS-PCR
Primer Set A 76 [59]
Primer Set B 54 [59]
Biopsy washings
Goessl et al. MS-PCR 0 67 100 [50]
a
MS-PCR: methylation specic PCR; RE-PCR:
5-m
CpG-sensitive restriction enzyme PCR.
b
Benign prostatic hyperplasia.
c
Prostatic intraepithilial neoplasia.
d
Prostate cancer.
[32]. Jeronimo et al. detected GSTP1 hypermethylation
in the plasma of 36% of men with organ conned
prostate cancer [56]. Goessl et al. found GSTP1 hyper-
methylation in plasma of 56% of pathological stage T2
3N0M0 and in 93% of pathological stage T4N+ or M+
prostate cancer patients [57,58]. The clinical use of these
studies remains unclear, but appears promising.
Other bodily uids that can be collected and tested
for CpG island hypermethylation include urine, ejacu-
late and prostatic secretions. DNA can appear in uids
of the urinary tract by cell shedding into the prostatic
ducts. Gonzalgo et al. recently reported that GSTP1
hypermethylation was detectable in 86% of prostatic
secretion specimens from men undergoing radical
prostatectomy [59]. Suh et al. was the rst to demon-
strate the usefulness of GSTP1 hypermethylation for
detecting prostate cancer in the ejaculate [60]. They
found GSTP1 hypermethylation in the ejaculate of
approximately 50% men with prostate cancer [60].
Cairns et al. demonstrated GSTP1 hypermethylation
in urine of 27% of men with early stage prostate cancer
[61]. After one minute of prostatic massage, Goessl et
al. were able to detect GSTP1 hypermethylation in 73%
of men with prostate cancer [62]. By presumably
enriching the urine with prostate cells, prostatic mas-
sage appears to increase the detection rate of hyper-
methylated GSTP1 in men with prostate cancer in
comparison to a routine urine sample. This would also
seem to explain the high detection rates observed by
Gonzalgo et al. in post-biopsy urine specimens [51].A
prospective evaluation of GSTP1 hypermethylation in
bodily uids for prostate cancer has yet to be published.
Additional studies using prostate specic marker
panels are also warranted to distinguish prostate cancer
from other cancers such as kidney, urinary bladder or
gastrointestinal cancers.
6. Conclusion
The hypermethylation of CpG islands represents a
somatic, epigenetic event that almost uniformly arises
during prostate caricnogenesis. Using modern detection
assays, CpG island hypermethylation of multiple pros-
tate cancer specic genes has become a promising
molecular marker for prostate cancer diagnosis and
detection. By applying these techniques to readily avail-
able clinical specimens such as urine or blood the current
ability to diagnosis prostate cancer may be improved.
Acknowledgements
This work was supported by NIH/NCI grant R01
CA70196 and NIH/NCI SPORE grant P50 CA58236.
W.G. Nelson has a patent (United States patent
5.552.277) titled ‘‘Genetic Diagnosis of Prostate
Cancer’’.
References
[1] Soh S, Kattan MW, Berkman S, Wheeler TM, Scardino PT. Has there
been a recent shift in the pathological features and prognosis of
patients treated with radical prostatectomy? J Urol 1997;157:
2212.
[2] Lerner SE, Seay TM, Blute ML, Bergstralh EJ, Barrett D, Zincke H.
Prostate specic antigen detected prostate cancer (clinical stage T1c):
an interim analysis. J Urol 1996;155:821.
[3] Thompson IM, Goodman PJ, Tangen CM, Lucia MS, Miller GJ, Ford
LG, et al. The inuence of nasteride on the development of prostate
cancer. N Engl J Med 2003;349:215.
[4] Sakr WA, Grignon DJ, Crissman JD, Heilbrun LK, Cassin BJ, Pontes
JJ, et al. High grade prostatic intraepithelial neoplasia (HGPIN) and
prostatic adenocarcinoma between the ages of 2069: an autopsy
study of 249 cases. In Vivo 1994;8:439.
[5] Thompson IM, Pauler DK, Goodman PJ, Tangen CM, Lucia MS,
Parnes HL, et al. Prevalence of prostate cancer among men with a
prostate-specic antigen level <or = 4.0 ng per milliliter. N Engl J
Med 2004;350:2239.
[6] Jemal A, Tiwari RC, Murray T, Ghafoor A, Samuels A, Ward E, et al.
Cancer statistics. CA Cancer J Clin 2004;54:8.
[7] Draisma G, Boer R, Otto SJ, van der Cruijsen IW, Damhuis RAM,
Schroder FH, et al. Lead times and overdetection due to prostate-
specic antigen screening: estimates from the European randomized
study of screening for prostate cancer. J Natl Cancer Inst
2003;95:868.
[8] Chen C, Lamharzi N, Weiss NS, Etzioni R, Dightman DA, Barnett M,
et al. Androgen receptor polymorphisms and the incidence of pros-
tate cancer. Cancer Epidemiol Biomarkers Prev 2002;11:1033.
[9] Nelson WG, De Marzo AM, Isaacs WB. Prostate cancer. N Engl J
Med 2003;349:366.
[10] Makhlouf AA, Krupski TL, Kunkle D, Theodorescu D. The effect of
sampling more cores on the predictive accuracy of pathological grade
and tumour distribution in the prostate biopsy. BJU Int 2004;93:
271.
[11] de la Taille A, Antiphon P, Salomon L, Cherfan M, Porcher R,
Hoznek A, et al. Prospective evaluationof a 21-sample needle biopsy
procedure designed to improve the prostate cancer detection rate.
Urology 2003;61:1181.
[12] De Marzo AM, Meeker AK, Zha S, Luo J, Nakayama M, Platz EA,
et al. Human prostate cancer precursors and pathobiology. Urology
2003;62:55.
[13] Signoretti S, Waltregny D, Dilks J, Isaac B, Lin D, Garraway L, et al.
p63 is a prostate basal cell marker and is required for prostate
development. Am J Pathol 2000;157:1769.
[14] Jiang Z, Iczkowski KA, Woda BA, Tretiakova M, Yang XJ. P504S
immunostaining boosts diagnostic resolution of ‘‘suspicious’’ foci in
prostatic needle biopsy specimens. Am J Clin Pathol 2004;121:99.
[15] Parsons JK, Gage WR, Nelson WG, De Marzo AM. p63 protein
expression is rare in prostate adenocarcinoma: implications for
cancer diagnosis and carcinogenesis. Urology 2001;58:619.
P.J. Bastian et al. / European Urology 46 (2004) 698708 705
[16] Luo J, Dunn TA, Ewing CM, Walsh PC, Isaacs WB. Decreased gene
expression of steroid 5 alpha-reductase 2 in human prostate cancer:
implications for nasteride therapy of prostate carcinoma. Prostate
2003;57:134.
[17] Zhou M, Aydin H, Kanane H, Epstein JI. How often does alpha-
methylacyl-CoA-racemase contribute to resolving an atypical diag-
nosis on prostate needle biopsy beyond that provided by basal cell
markers? Am J Surg Pathol 2004;28:239.
[18] Feinberg AP, Vogelstein B. Hypomethylation distinguishes genes of
some human cancers from their normal counterparts. Nature
1983;301:89.
[19] Lee WH, Morton RA, Epstein JI, Brooks JD, Campbell PA, Bova GS,
et al. Cytidine methylation of regulatory sequences near the pi-class
glutathione S-transferase gene accompanies human prostatic carci-
nogenesis. Proc Natl Acad Sci USA 1994;91:11733.
[20] Nelson WG, De Marzo AM, DeWeese TL. The molecular pathogen-
esis of prostate cancer: Implications for prostate cancer prevention.
Urology 2001;57:39.
[21] Bird AP. DNA methylation and the frequency of CpG in animal
DNA. Nucleic Acids Res 1980;8:1499.
[22] Herman JG, Baylin SB. Gene silencing in cancer in association with
promoter hypermethylation. N Engl J Med 2003;349:2042.
[23] Attwood JT, Yung RL, Richardson BC. DNA methylation and
the regulation of gene transcription. Cell Mol Life Sci 2002;59:
241.
[24] Jones PA, Baylin SB. The fundamental role of epigenetic events in
cancer. Nat Rev Genet 2002;3:415.
[25] Bird A, Taggart M, Frommer M, Miller OJ, Macleod D. A fraction of
the mouse genome that is derived from islands of nonmethylated,
CpG-rich DNA. Cell 1985;40:91.
[26] Esteller M, Corn PG, Baylin SB, Herman JG. A gene hypermethyla-
tion prole of human cancer. Cancer Res 2001;61:3225.
[27] Clark SJ, Harrison J, Paul CL, Frommer M. High sensitivity mapping
of methylated cytosines. Nucleic Acids Res 1994;22:2990.
[28] Herman JG, Graff JR, Myohanen S, Nelkin BD, Baylin SB. Methy-
lation-specic PCR: a novel PCR assay for methylation status of CpG
islands. Proc Natl Acad Sci USA 1996;93:9821.
[29] Eads CA, Danenberg KD, Kawakami K, Saltz LB, Blake C, Shibata
D, et al. MethylLight: a high-troughput assay to measure DNA
methylation. Nucleic Acids Res 2000;28:E32.
[30] Lee WH, Isaacs WB, Bova GS, Nelson WG. CG island methylation
changes near the GSTP1 gene in prostatic carcinoma cells detected
using the polymerase chain reaction: a new prostate cancer biomar-
ker. Cancer Epidemiol Biomarkers Prev 1997;6:443.
[31] Herman JG, Jen J, Merlo A, Baylin SB. Hypermethylation-associated
inactivation indicates a tumor suppressor role for p15INK4B. Cancer
Res 1996;56:722.
[32] Nakayama M, Gonzalgo ML, Yegnasubramanian S, Lin X, De Marzo
AM, Nelson WG. GSTP1 CpG island hypermethylation as a mole-
cular biomarker for prostate cancer. J Cell Biochem 2004;91:540.
[33] Hayes JD, Pulford DJ. Te glutathione S-transferase supergene family:
Regulation of GST and the contribution of the isoenzymes to cancer
chemoprotection and drug resistance. Crit Rev Biochem Mol Biol
1995;30:445.
[34] Kinzler KW, Vogelstein B. Cancer-susceptibility genes: gatekeepers
and caretakers. Nature 1997;386:761.
[35] Shirai T, Sano M, Tamano S, Takahashi S, Hirose M, Futakuchi M,
et al. The prostate: a target for carcinogenicity of 2-amino-1-methyl-
6-phenylimidazo[4,5-b]pyridine (PhIP) derived from cooked foods.
Cancer Res 1997;57:195.
[36] Chan JM, Giovannucci EL. Vegetables, fruits, associated micro-
nutrients, and risk of prostate cancer. Epidemiol Rev 2001;23: 81.
[37] Maruyama R, Toyooka S, Toyooka KO, Virmani AK, Zochbauer-
Muller S, Farinas AJ, et al. Aberrant promoter methylation prole of
prostate cancers and its relationship to clinicopathological features.
Clin Cancer Res 2002;8:514.
[38] Yamanaka M, Watanabe M, Yamada Y, Takagi A, Murata T, Taka-
hashi H, et al. Altered methylation of multiple genes in carcinogen-
esis of the prostate. Int J Cancer 2003;106:382.
[39] Konishi N, Nakamura M, Kishi M, Nishimine M, Ishida E, Shimada
K. DNA hypermethylation status of multiple genes in prostate
adenocarcinomas. Jpn J Cancer Res 2002;93:767.
[40] Yegnasubramanian S, Kowalski J, Gonzalgo ML, Zahurak M, Pian-
tadosi S, Walsh PC, et al. Hypermethylation of CpG islands in
primary and metastatic human prostate cancer. Cancer Res
2004;64:1975.
[41] Kang GH, Lee S, Lee HJ, Hwang KS. Aberrant CpG island hyper-
methylation of multiple genes in prostate cancer and prostatic
intraepithelial neoplasia. J Pathol 2004;202:233.
[42] Jeronimo C, Henrique R, Campos PF, Oliveira J, Caballero OL,
Lopes C, et al. Endothelin B receptor gene hypermethylation in
prostate adenocarcinoma. J Clin Pathol 2003;56:52.
[43] Li LC, Chui R, Nakajima K, Oh BR, Au HC, Dahiya R. Frequent
methylation of estrogen receptor in prostate cancer: correlation with
tumor progression. Cancer Res 2000;60:702.
[44] Nojima D, Li LC, Dharia A, Perinchery G, Ribeiro-Filho L, Yen TS,
et al. CpG hypermethylation of the promoter region inactivates the
estrogen receptor-beta gene in patients with prostate carcinoma.
Cancer 2001;92:2076.
[45] Nakayama M, Bennett CJ, Hicks JL, Epstein JI, Platz EA, Nelson
WG, et al. Hypermethylation of the human glutathione S-transferase-
pi gene (GSTP1) CpG island is present in a subset of proliferative
inammatory atrophy lesions but not in normal or hyperplastic
epithelium of the prostate: a detailed study using laser-capture
microdissection. Am J Pathol 2003;163:923.
[46] De Marzo AM, Marchi VL, Epstein JI, Nelson WG. Proliferative
inammatory atrophy of the prostate: implications for prostatic
carcinogenesis. Am J Pathol 1999;155:1985.
[47] Paul R, Scho
¨ler S, van Randenborgh H, Ku
¨bler H, Alschibaja M,
Busch R, et al. Morbidity of prostattic biopsy for different biopsy
strategies: is there a relation to core number and sampling region?
Eur Urol 2004;45:450.
[48] Harden SV, Sanderson H, Goodman SN, Partin AA, Walsh PC,
Epstein JI, et al. Quantitative GSTP1 methylation and the detection
of prostate adenocarcinoma in sextant biopsies. J Natl Cancer Inst
2003;95:1634.
[49] Chu DC, Chuang CK, Fu JB, Huang HS, Tseng CP, Sun CF. The use
of real-time quantitative polymerase chain reaction to detect hyper-
methylation of the CpG islands in the promoter region anking
the GSTP1 gene to diagnose prostate carcinoma. J Urol 2002;
167:1854.
[50] Goessl C, Muller M, Heicappell R, Krause H, Schostak M, Straub B,
et al. Methylation-specic PCR for detection of neoplastic DNA in
biopsy washings. J Pathol 2002;196:331.
[51] Gonzalgo ML, Pavlovich CP, Lee SM, Nelson WG. Prostate cancer
detection by GSTP1 methylation analysis of postbiopsy urine speci-
mens. Clin Cancer Res 2003;9:2673.
[52] Santourlidis S, Florl A, Ackermann R, Wirtz HC, Schulz WA. High
frequency of alterations in DNA methylation in adenocarcinoma of
the prostate. Prostate 1999;39:166.
[53] Kollermann J, Muller M, Goessl C, Krause H, Helpap B, Pantel K,
et al. Methylation-specic PCR for DNA-based detection of occult
tumor cells in lymph nodes of prostate cancer patients. Eur Urol
2003;44:533.
[54] Mandel P, Metais P. Les acids nucleiques du plasma saguin chez
lhomme. CR Acad Aci Paris: 241, 1948.
[55] Goessl C, Heicappell R, Munker R, Anker P, Stroun M, Krause H,
et al. Microsatellite analysis of plasma DNA from patients with clear
cell renal carcinoma. Cancer Res 1998;58:4728.
[56] Jeronimo C, Usadel H, Henrique R, Silva C, Oliveira J, Lopes C, et al.
Quantitative GSTP1 hypermethylation in bodily uids of patients
with prostate cancer. Urology 2002;60:1131.
P.J. Bastian et al. / European Urology 46 (2004) 698708706
[57] Goessl C, Krause H, Muller M, Heicappell R, Schrader M, Sach-
singer J, et al. Fluorescent methylation-specic polymerase chain
reaction for DNA-based detection of prostate cancer in bodily uids.
Cancer Res 2000;60:5941.
[58] Goessl C, Muller M, Heicappell R, Krause H, Miller K. DNA-based
detection of prostate cancer in blood, urine, and ejaculates. Ann NY
Acad Sci 2001;945:51.
[59] Gonzalgo ML, Nakayama M, Lee SM, De Marzo AM, Nelson WG.
Detection of GSTP1 methylation in prostatic secretions using com-
binatorial MSP analysis. Urology 2004;63:414.
[60] Suh CI, Shanafelt T, May DJ, Shroyer KR, Bobak JB, Crawford ED,
et al. Comparison of telomerase activity and GSTP1 promoter
methylation in ejaculate as potential screening tests for prostate
cancer. Mol Cell Probes 2000;14:211.
[61] Cairns P, Esteller M, Herman JG, Schoenberg M, Jeronimo C,
Sanchez-Cespedes M, et al. Molecular detection of prostate cancer
in urine by GSTP1 hypermethylation. Clin Cancer Res 2001;7:2727.
[62] Goessl C, Muller M, Heicappell R, Krause H, Straub B, Schrader M,
et al. DNA-based detection of prostate cancer in urine after prostatic
massage. Urology 2001;58:335.
[63] Woodson K, Hayes R, Wideroff L, Villaruz L, Tangrea J. Hyper-
methylation of GSTP1, CD44, and E-cadherin genes in prostate
cancer among US Blacks and Whites. Prostate 2003;55:199.
[64] Lin X, Tascilar M, Lee WH, Vles WJ, Lee BH, Veeraswamy R, et al.
GSTP1 CpG island hypermethylation is responsible for the absence
of GSTP1 expression in human prostate cancer cells. Am J Pathol
2001;159:1815.
[65] Brooks JD, Weinstein M, Lin X, Sun Y, Pin SS, Bova GS, et al. CG
island methylation changes near the GSTP1 gene in prostatic intrae-
pithelial neoplasia. Cancer Epidemiol Biomarkers Prev 1998;7:531.
[66] Jeronimo C, Usadel H, Henrique R, Oliveira J, Lopes C, Nelson WG,
et al. Quantitation of GSTP1 methylation in non-neoplastic prostatic
tissue and organ-conned prostate adenocarcinoma. J Natl Cancer
Inst 2001;93:1747.
[67] Jeronimo C, Varzim G, Henrique R, Oliveira J, Bento MJ, Silva C,
et al. I105V polymorphism and promoter methylation of the GSTP1
gene in prostate adenocarcinoma. Cancer Epidemiol Biomarkers
Prev 2002;11:445.
[68] Millar DS, Ow KK, Paul CL, Russell PJ, Molloy PL, Clark SJ.
Detailed methylation analysis of the glutathione S-transferase pi
(GSTP1) gene in prostate cancer. Oncogene 1999;18:1313.
[69] Esteller M, Corn PG, Urena JM, Gabrielson E, Baylin SB, Herman
JG. Inactivation of glutathione S-transferase P1 gene by promoter
hypermethylation in human neoplasia. Cancer Res 1998;58:4515.
[70] Goessl C. Laser-uorescence microsatellite analysis and new results
in microsatellite analysis of plasma/serum DNA of cancer patients.
Ann NY Acad Sci 2000;906:63.
[71] Harden SV, Guo Z, Epstein JI, Sidransky D. Quantitative GSTP1
methylation clearly distinguishes benign prostatic tissue and limited
prostate adenocarcinoma. J Urol 2003;169:1138.
[72] Li LC, Zhao H, Nakajima K, Oh BR, Filho LA, Carroll P, et al.
Methylation of the E-cadherin gene promoter correlates with pro-
gression of prostate cancer. J Urol 2001;166:705.
[73] Kallakury BV, Sheehan CE, Winn-Deen E, Oliver J, Fisher HA,
Kaufman Jr RP, et al. Decreased expression of catenins (alpha and
beta), p120 CTN, and E-cadherin cell adhesion proteins and E-
cadherin gene promoter methylation in prostatic adenocarcinomas.
Cancer 2001;92:2786.
[74] Konishi N, Nakamura M, Kishi M, Nishimine M, Ishida E, Shimada
K. Heterogeneous methylation and deletion patterns of the INK4a/
ARF locus within prostate carcinomas. Am J Pathol 2002;160:1207.
[75] Gu K, Mes-Masson AM, Gauthier J, Saad F. Analysis of the p16
tumor suppressor gene in early-stage prostate cancer. Mol Carcinog
1998;21:164.
[76] Nguyen TT, Nguyen CT, Gonzales FA, Nichols PW, Yu MC, Jones
PA. Analysis of cyclin-dependent kinase inhibitor expression and
methylation patterns in human prostate cancers. Prostate 2000;
43:233.
[77] Jarrard DF, Bova GS, Ewing CM, Pin SS, Nguyen SH, Baylin SB,
et al. Deletional, mutational, and methylation analyses of CDKN2
(p16/MTS1) in primary and metastatic prostate cancer. Genes Chro-
mosomes Cancer 1997;19:90.
[78] Kuzmin I, Gillespie JW, Protopopov A, Geil L, Dreijerink K, Yang Y,
et al. The RASSF1A tumor suppressor gene is inactivated in prostate
tumors and suppresses growth of prostate carcinoma cells. Cancer
Res 2002;62:3498.
[79] Liu L, Yoon JH, Dammann R, Pfeifer GP. Frequent hypermethylation
of the RASSF1A gene in prostate cancer. Oncogene 2002;21:6835.
[80] Nakayama T, Watanabe M, Yamanaka M, Hirokawa Y, Suzuki H, Ito
H, et al. The role of epigenetic modications in retinoic acid receptor
beta2 gene expression in human prostate cancers. Lab Invest 2001;
81:1049.
[81] Kito H, Suzuki H, Ichikawa T, Sekita N, Kamiya N, Akakura K, et al.
Hypermethylation of the CD44 gene is associated with progression
and metastasis of human prostate cancer. Prostate 2001;49:110.
[82] Verkaik NS, van Steenbrugge GJ, van Weerden WM, Bussemakers
MJ, van der Kwast TH. Silencing of CD44 expression in prostate
cancer by hypermethylation of the CD44 promoter region. Lab Invest
2000;80:1291.
[83] Lou W, Krill D, Dhir R, Becich MJ, Dong JT, Frierson Jr HF, et al.
Methylation of the CD44 metastasis suppressor gene in human
prostate cancer. Cancer Res 1999;59:2329.
[84] Nakayama T, Watanabe M, Suzuki H, Toyota M, Sekita N, Hirokawa
Y, et al. Epigenetic regulation of androgen receptor gene expression
in human prostate cancers. Lab Invest 2000;80:1789.
[85] Sasaki M, Tanaka Y, Perinchery G, Dharia A, Kotcherguina I,
Fujimoto S, et al. Methylation and inactivation of estrogen, proges-
terone, and androgen receptors in prostate cancer. J Natl Cancer Inst
2002;94:384.
[86] Kinoshita H, Shi Y, Sandefur C, Meisner LF, Chang C, Choon A,
et al. Methylation of the androgen receptor minimal promoter
silences transcription in human prostate cancer. Cancer Res
2000;60:3623.
[87] Pao MM, Tsutsumi M, Liang G, Uzvolgyi E, Gonzales FA, Jones PA.
The endothelin receptor B (EDNRB) promoter displays heteroge-
neous, site specic methylation patterns in normal and tumor cells.
Hum Mol Genet 2001;10:903.
[88] Nelson JB, Lee WH, Nguyen SH, Jarrard DF, Brooks JD, Magnuson
SR, et al. Methylation of the 50CpG island of the endothelin B
receptor gene is common in human prostate cancer. Cancer Res
1997;57:35.
[89] Padar A, Sathyanarayana UG, Suzuki M, Maruyama R, Hsieh JT,
Frenkel EP, et al. Inactivation of cyclin D2 gene in prostate cancers
by aberrant promoter methylation. Clin Cancer Res 2003;9:4730.
[90] Sasaki M, Kaneuchi M, Fujimoto S, Tanaka Y, Dahiya R. Hyper-
methylation can selectively silence multiple promoters of steroid
receptors in cancers. Mol Cell Endocrinol 2003;202:201.
[91] Yamada Y, Watanabe M, Yamanaka M, Hirokawa Y, Suzuki H,
Takagi A, et al. Aberrant methylation of the vascular endothelial
growth factor receptor-1 gene in prostate cancer. Cancer Sci
2003;94:536.
[92] Kibel AS, Christopher M, Faith DA, Bova GS, Goodfellow PJ, Isaacs
WB. Methylation and mutational analysis of p27(kip1) in prostate
carcinoma. Prostate 2001;48:248.
[93] Zhang J, Liu L, Pfeifer GP. Methylation of the retinoid response gene
TIG1 in prostate cancer correlates with methylation of the retinoic
acid receptor beta gene. Oncogene 2004;23:2241.
[94] Santourlidis S, Warskulat U, Florl AR, Maas S, Pulte T, Fischer J,
et al. Hypermethylation of the tumor necrosis factor receptor super-
family 6 (APT1, Fas, CD95/Apo-1) gene promoter at rel/nuclear
factor kappaB sites in prostatic carcinoma. Mol Carcinog
2001;32:36.
P.J. Bastian et al. / European Urology 46 (2004) 698708 707
[95] Morton Jr RA, Watkins JJ, Bova GS, Wales MM, Baylin SB, Isaacs
WB. Hypermethylation of chromosome 17P locus D17S5 in human
prostate tissue. J Urol 1996;156:512.
[96] Vanaja DK, Cheville JC, Iturria SJ, Young CY. Transcriptional silen-
cing of zinc nger protein 185 identied by expression proling is
associatedwith prostate cancer progression. Cancer Res 2003;63:3877.
[97] Schmitt JF, Millar DS, Pedersen JS, Clark SL, Venter DJ, Frydenberg
M, et al. Hypermethylation of the inhibin alpha-subunit gene in
prostate carcinoma. Mol Endocrinol 2002;16:213.
[98] Cui J, Rohr LR, Swanson G, Speights VO, Maxwell T, Brothman AR.
Hypermethylation of the caveolin-1 gene promoter in prostate cancer.
Prostate 2001;46:249.
[99] Fukuhara H, Kuramochi M, Fukami T, Kasahara K, Furuhata M,
Nobukuni T, et al. Promoter methylation of TSLC1 and tumor
suppression by its gene product in human prostate cancer. Jpn J
Cancer Res 2002;93:605.
[100] Usmani BA, Shen R, Janeczko M, Papandreou CN, Lee WH, Nelson
WG, et al. Methylation of the neutral endopeptidase gene promoter in
human prostate cancers. Clin Cancer Res 2000;6:1664.
[101] Gao AC, Lou W, Sleeman JP, Isaacs JT. Metastasis suppression by the
standard CD44 isoform does not require the binding of prostate
cancer cells to hyaluronate. Cancer Res 1998;58:2350.
[102] Gao AC, Lou W, Dong JT, Isaacs JT. CD44 is a metastasis suppressor
gene for prostatic cancer located on human chromosome 11p13.
Cancer Res 1997;57:846.
[103] Esteller M, Cordon-Cardo C, Corn PG, Meltzer SJ, Pohar KS,
Watkins DN, et al. p14ARF silencing by promoter hypermethylation
mediates abnormal intracellular localization of MDM2. Cancer Res
2001;61:2816.
[104] Morris MR, Hesson LB, Wagner KJ, Morgan NV, Astuti D, Lees RD,
et al. Multigene methylation analysis of Wilmstumour and adult
renal cell carcinoma. Oncogene 2003;22:6794.
[105] Nojima D, Nakajima K, Li LC, Franks J, Ribeiro-Filho L, Ishii N,
et al. CpG methylation of promoter region inactivates E-cadherin
gene in renal cell carcinoma. Mol Carcinog 2001;32:19.
[106] Maestro ML, Del Barco V, Sanz-Casla MT, Moreno J, Zanna I,
Redondo E, et al. 50CpG island methylation of the suppressor gene
p16/CDKN2/MTS1 in renal carcinoma. Arch Esp Urol 2000;53:417.
[107] Romanenko A, Morell-Quadreny L, Lopez-Guerrero JA, Pellin A,
Nepomnyaschy V, Vozianov A, et al. P16INK4A and p15INK4B
gene alteration associated with oxidative stress in renal cell carci-
nomas after the chernobyl accident (pilot study). Diagn Mol Pathol
2002;11:163.
[108] Dreijerink K, Braga E, Kuzmin I, Geil L, Duh FM, Angeloni D, et al.
The candidate tumor suppressor gene, RASSF1A, from human
chromosome 3p21.3 is involved in kidney tumorigenesis. Proc Natl
Acad Sci USA 2001;98:7504.
[109] Morrissey C, Martinez A, Zatyka M, Agathanggelou A, Honorio S,
Astuti D, et al. Epigenetic inactivation of the RASSF1A 3p21.3
tumor suppressor gene in both clear cell and papillary renal cell
carcinoma. Cancer Res 2001;61:7277.
[110] Yoon JH, Dammann R, Pfeifer GP. Hypermethylation of the CpG
island of the RASSF1A gene in ovarian and renal cell carcinomas. Int
J Cancer 2001;94:212.
[111] Bachman KE, Herman JG, Corn PG, Merlo A, Costello JF, Cavenee
WK, et al. Methylation-associated silencing of the tissue inhibitor of
metalloproteinase-3 gene suggest a suppressor role in kidney, brain,
and other human cancers. Cancer Res 1999;59:798.
[112] Clifford SC, Prowse AH, Affara NA, Buys CH, Maher ER. Inactiva-
tion of the von Hippel-Lindau (VHL) tumour suppressor gene and
allelic losses at chromosome arm 3p in primary renal cell carcinoma:
evidence for a VHL-independent pathway in clear cell renal tumour-
igenesis. Genes Chromosomes Cancer 1998;22:200.
[113] Girolami F, Passerini I, Gargano D, Frusconi S, Villari D, Nicita G,
et al. Microsatellite analysis of chromosome 3p region in sporadic
renal cell carcinomas. Pathol Oncol Res 2002;8:241.
[114] Herman JG, Latif F, Weng Y, Lerman MI, Zbar B, Liu S, et al.
Silencing of the VHL tumor-suppressor gene by DNA methylation in
renal carcinoma. Proc Natl Acad Sci USA 1994;91:9700.
[115] Oh RR, Park JY, Lee JH, Shin MS, Kim HS, Lee SK, et al. Expression
of HGF/SF and Met protein is associated with genetic alterations of
VHL gene in primary renal cell carcinomas. Apmis 2002;110:229.
[116] Prowse AH, Webster AR, Richards FM, Richard S, Olschwang S,
Resche F, et al. Somatic inactivation of the VHL gene in Von Hippel-
Lindau disease tumors. Am J Hum Genet 1997;60:765.
[117] Brauch H, Weirich G, Brieger J, Glavac D, Rodl H, Eichinger M,
et al. VHL alterations in human clear cell renal cell carcinoma:
association with advanced tumor stage and a novel hot spot mutation.
Cancer Res 2000;60:1942.
[118] Kondo K, Yao M, Yoshida M, Kishida T, Shuin T, Miura T, et al.
Comprehensive mutational analysis of the VHL gene in sporadic
renal cell carcinoma: relationship to clinicopathological parameters.
Genes Chromosomes Cance 2002;34:58.
[119] Maruyama R, Toyooka S, Toyooka KO, Harada K, Virmani AK,
Zochbauer-Muller S, et al. Aberrant promoter methylation prole of
bladder cancer and its relationship to clinicopathological features.
Cancer Res 2001;61:8659.
[120] Florl AR, Franke KH, Niederacher D, Gerharz CD, Seifert HH,
Schulz WA. DNA methylation and the mechanisms of CDKN2A
inactivation in transitional cell carcinoma of the urinary bladder. Lab
Invest 2000;80:1513.
[121] Chan MW, Chan LW, Tang NL, Tong JH, Lo KW, Lee TL, et al.
Hypermethylation of multiple genes in tumor tissues and voided
urine in urinary bladder cancer patients. Clin Cancer Res 2002;8:464.
[122] Akao T, Kakehi Y, Itoh N, Ozdemir E, Shimizu T, Tachibana A, et al.
A high prevalence of functional inactivation by methylation mod-
ication of p16INK4A/CDKN2/MTS1 gene in primary urothelial
cancers. Jpn J Cancer Res 1997;88:1078.
[123] Chang LL, Yeh WT, Yang SY, Wu WJ, Huang CH. Genetic altera-
tions of p16INK4a and p14ARF genes in human bladder cancer. J
Urol 2003;170:595.
[124] Tada Y, Wada M, Taguchi K, Mochida Y, Kinugawa N, Tsuneyoshi
M, et al. The association of death-associated protein kinase hyper-
methylation with early recurrence in supercial bladder cancers.
Cancer Res 2002;62:4048.
[125] Tsutsumi M, Tsai YC, Gonzalgo ML, Nichols PW, Jones PA. Early
acquisition of homozygous deletions of p16/p19 during squamous
cell carcinogenesis and genetic mosaicism in bladder cancer. Onco-
gene 1998;17:3021.
[126] Valenzuela MT, Galisteo R, Zuluaga A, Villalobos M, Nunez MI,
Oliver FJ, et al. Assessing the use of p16(INK4a) promoter gene
methylation in serum for detection of bladder cancer. Eur Urol
2002;42:622.
[127] Wu Y, Wu H, Ma T, Wu C. The mechanism of p16 gene inactivation
in human bladder transitional cell carcinoma. Zhonghua Bing Li Xue
Za Zhi 1999;28:39.
[128] Muto S, Horie S, Takahashi S, Tomita K, Kitamura T. Genetic and
epigenetic alterations in normal bladder epithelium in patients with
metachronous bladder cancer. Cancer Res 2000;60:4021.
[129] Bornman DM, Mathew S, Alsruhe J, Herman JG, Gabrielson E.
Methylation of the E-cadherin gene in bladder neoplasia and in
normal urothelial epithelium from elderly individuals. Am J Pathol
2001;159:831.
[130] Ribeiro-Filho LA, Franks J, Sasaki M, Shiina H, Li LC, Nojima D,
et al. CpG hypermethylation of promoter region and inactivation of
E-cadherin gene in human bladder cancer. Mol Carcinog 2002;34:
187.
[131] Horikawa Y, Sugano K, Shigyo M, Yamamoto H, Nakazono M,
Fujimoto H, et al. Hypermethylation of an E-cadherin (CDH1)
promoter region in high grade transitional cell carcinoma of
the bladder comprising carcinoma in situ. J Urol 2003;169:
1541.
P.J. Bastian et al. / European Urology 46 (2004) 698708708
... Most of the abnormal hypomethylation occurred in the open sea area (59%). The high enrichment of abnormal hypermethylation in the promoter CpG island region has been demonstrated by previous studies [46]. ...
... In biological process, the second-ranked activation of innate immune response (covering 10 cancers) was closely related to the immune response. The first step in fighting cancer cells was activating innate immune cells, and they could also work together with adaptive immune cells [46,47]. In hallmark, the first-ranked interferon gamma response (covering 11 cancers) and the second-ranked inflammatory response (covering 9 cancers) were closely related to the immune response. ...
Article
Full-text available
Abnormal DNA methylation is a fundamental characterization of epigenetics in cancer. Here we demonstrate that aberrant DNA methylating can modulate the tumour immune microenvironment in 16 cancer types. Differential DNA methylation in promoter region can regulate the transcriptomic pattern of immune-related genes and DNA hypomethylation mainly participated in the processes of immunity, carcinogenesis and immune infiltration. Moreover, many cancer types shared immune-related functions, like activation of innate immune response, interferon gamma response and NOD-like receptor signalling pathway. DNA methylation can further help identify molecular subtypes of kidney renal clear cell carcinoma. These subtypes are characterized by DNA methylation pattern, major histocompatibility complex, cytolytic activity and cytotoxic t lymphocyte and tumour mutation burden, and subtype with hypomethylation pattern shows unstable immune status. Then, we investigate the DNA methylation pattern of exhaustion-related marker genes and further demonstrate the role of hypomethylation in tumour immune microenvironment. In summary, our findings support the use of hypomethylation as a biomarker to understand the mechanism of tumour immune environment.
... Hypermethylation of its promotor thus leads to the loss of its function in cells and thus increased vulnerability of cells to DNA damage. In some studies, it has been found that this gene's promotor has been methylated in as many as 75% of pre invasive high grade prostatic intraepithelial neoplasms and in over 90% of prostate tumours [38]. ...
Article
Prostate cancer is the most common form of non-skin cancer in men and the second leading cause of cancer death among them. The estimated incidence of prostate cancer worldwide was 1,414,259 in 2020, making it the fourth most commonly diagnosed malignancy, with the majority of cases seen in those over 65 years of age. Several genes have been implicated in prostate cancer development using many molecular methods to elucidate genes related to its development and progressions. Prostate cancer gene variants can be categorised based on many factors, such as the type of gene they affect (tumour suppressor, oncogene) or the type of mutation observed in the development of the illness (SNP’s, gene fusions, translocations, deletions, duplications etc) epigenetic changes and even alternative splice variants are also observed in the development of prostate cancer. Understanding these various genetic connotations can then have implications for diagnosis and treatment.
... In the aspect of immune microenvironment, one of the characteristics of prostate atrophy was the hypermethylation of CpG island in glutathione S-transferase-pi (GSTP1) gene. GSTP1 gene CpG island hypermethylation was thought to be the early change in the tumorigenesis of PCa [38], and was barely observed in normal prostate epithelial cells and hyperplastic cells [39]. Normally, the product of GSTP1 gene is glutathione S-transferase, an anti-oxidative enzyme releasing the inflammatory oxidative stress (OS) from reactive oxygen species (ROS). ...
Article
Full-text available
Currently, medication for benign prostate hyperplasia (BPH) and prostate cancer (PCa) are mainly based on modulating the hormone and nervous systems. However, side effects often affect patients, and might decrease their commitment to continuing the medication and lower their quality of life. Some studies have indicated that chronic inflammation might be the cause of BPH and PCa. Based on this hypothesis, the effect of phloretin, a potent anti-inflammatory and anti-oxidative flavonoid, has been researched since 2010. Results from animal and in-vitro studies, obtained from databases, also indicate that the use of phloretin in treating BPH and PCa is promising. Due to its effect on inflammatory cytokines, apoptosis or anti-apoptosis, reactive oxygen species, anti-oxidant enzymes and oxidative stress, phloretin is worthy of further study in human clinical trials regarding safety and effective dosages.
... Other examples are S100P and MAGE in pancreatic cancer and melanoma, respectively (Futscher et al., 2004). Unlike the hypomethylation, hypermethylation is a property shown by the specific CpG region only (Bastian et al., 2004;Ellinger et al., 2008;Kvasha et al., 2008;Xi et al., 2013;Fujii et al., 2015;Skvortsova et al., 2019). Due to that transcriptional inactivation of promoter genes involved in cell repair, cell cycle mechanism, and apoptosis process occurs. ...
Book
This book provides information about different types and stages of cancer and their subtypes with their respective molecular mechanisms, etiology, histopathology, and cellular origins. This book also provides detailed information about cancer incidence, mortality, and different types of technologies both bio and nano employed in cancer diagnosis and screening, and their applications in cancer therapies. This book informs readers about molecular mechanisms of cancer, diagnosis, and therapies along with different computational techniques used on a single platform. The chapters include a broad and integrated perspective on cancer-related topics. This book covers both conventional and emerging techniques employed in cancer screening and diagnosis, including imaging, biomarker, and electrochemical nanosensor-based approaches with detailed information on sensor development. Similarly, this book also covers the mechanisms of different conventional and emerging herbal and nano therapies used in cancer treatment. The authors discuss applications of different computational and mathematical tools, such as machine-learning methods, that can be employed in cancer diagnosis and therapy at the level of personalized medicine. Features: Offers an integrated approach to provide information about all aspects of cancer biology, diagnosis, and therapy Focuses on both conventional and emerging tools/techniques applicable in cancer screening and diagnosis Covers the mechanisms of conventional and emerging anticancer drugs and therapies Provides insights about a personalized medicine-based approach in cancer diagnosis and therapy. This book is essential for university students, course lecturers, researchers, and industrialists working in the fields of cancer biology, medicine, and pharmacology.
Chapter
Epigenetics refers to changes in gene expression that are not caused by changes in the underlying DNA sequence. It is now clear that epigenetic changes may play a significant role in prostate cancer (PC) development and progression and can also be used to personalize treatment for PC patients. DNA methylation involves the addition of a methyl group to a cytosine or adenine base in the DNA molecule, which can lead to changes in gene expression. CpG dinucleotides are found in clusters called CpG islands, which are commonly found in promoter regions of genes. DNA methylation in promoter regions is associated with gene silencing, as it blocks the binding of transcription factors and RNA polymerase. Histone modifications, such as acetylation, can also influence gene expression by altering the accessibility of DNA to transcription factors and other regulatory proteins. Regulation of gene transcription is one of many antagonistic roles linked to H2A.Z nucleosome, and its overexpression has been associated with poor prognosis and increased metastasis. Among various histone modifications, H3K9, H3K18, H3K27, and H4K12 have been the focus of extensive research. A number of miRNA dysregulations have been identified in PC, and these dysregulated miRNAs have been implicated in the regulation of key pathways involved in cell proliferation, apoptosis, angiogenesis, and tumor metastasis.
Article
Full-text available
Simple Summary Prostate cancer (PCa) is characterized by various genomic alterations that play a pivotal role in carcinogenesis. Efforts in precision medicine aimed at improving diagnosis, prevention, and surveillance based on genetic alterations are advancing. Notably, no tumor markers surpass prostate-specific antigen in specificity, and existing treatments primarily target the androgen receptor axis, with exceptions for patients with alterations in homologous recombination repair-related genes, such as BRCA1/2 and ATM, who may benefit from poly (ADP-ribose) polymerase inhibitors. In order to delineate the current state of research on PCa, we provide an overview of cutting-edge genomic research, including genome alterations, cancer immunology, and non-coding RNAs, in PCa. These aspects are relevant to comprehending the molecular mechanisms underlying health disparities, PCa initiation and progression, and drug resistance in the precision medicine era. Abstract The progression of prostate cancer (PCa) relies on the activation of the androgen receptor (AR) by androgens. Despite efforts to block this pathway through androgen deprivation therapy, resistance can occur through several mechanisms, including the abnormal activation of AR, resulting in castration-resistant PCa following the introduction of treatment. Mutations, amplifications, and splicing variants in AR-related genes have garnered attention in this regard. Furthermore, recent large-scale next-generation sequencing analysis has revealed the critical roles of AR and AR-related genes, as well as the DNA repair, PI3K, and cell cycle pathways, in the onset and progression of PCa. Moreover, research on epigenomics and microRNA has increasingly become popular; however, it has not translated into the development of effective therapeutic strategies. Additionally, treatments targeting homologous recombination repair mutations and the PI3K/Akt pathway have been developed and are increasingly accessible, and multiple clinical trials have investigated the efficacy of immune checkpoint inhibitors. In this comprehensive review, we outline the status of PCa research in genomics and briefly explore potential future developments in the field of epigenetic modifications and microRNAs.
Article
Background: Oncologic outcomes for patients with localized prostate cancer (PCa) undergoing radical prostatectomy (RP) can vary widely. Hypermethylation of tumor-associated genes has potential as a novel diagnostic tool and predictive biomarker in PCa. We investigated the methylation status of tumor-associated genes in patients who underwent RP. Methods: Patients who underwent RP during 2004 to 2008 were matched retrospectively based on post-operative D'Amico risk stratification. Quantitative pyrosequencing was used to analyze methylation status of 10 gene loci in cancerous and adjacent benign tissue from histological specimen. Follow-up was performed according to EAU guideline recommendations. Statistical analyses were performed to correlate methylation levels in cancerous and benign tissue with risk profiles and biochemical recurrence (BCR). Results: The cohort included 71 patients: 22 low-risk, 22 intermediate-risk, and 27 high-risk. Mean follow-up time was 74 months. Methylation status differed significantly between cancerous and adjacent benign tissue for the 5 gene loci GSTP1, APC, RASSF1, TNFRFS10c, and RUNX3 (each P < 0.001). Also, the methylation level was significantly higher in high-risk than in low-risk patients for Endoglin2 and APC (P = 0.026; P = 0.032). Using ROC analysis, hypermethylation of APC in PCa tissue was associated with higher risk of BCR (P = 0.005). Conclusion: Methylation status of various gene loci holds diagnostic and predictive potential in PCa. Hypermethylation of APC, RASSF1, TNFRFS10c and RUNX3 were identified as novel PCa-specific biomarkers. Furthermore, increased methylation levels of APC and Endoglin2 were associated with high-risk PCa. Additionally, hypermethylation of APC was associated with increased risk of BCR after RP.
Article
The comparative analysis of the methylation status of promoter regions of eleven different suppressor genes has been carried out in the groups of patients with benign prostatic hyperplasia, prostate cancer and in patients without pathology with the use of methylation-specific polymerase chain reaction. The group without pathology revealed no methylation. The group of the patients with benign prostatic hyperplasia in most cases revealed either no methylation or it occurred in only one of the eleven genes. Methylation of two or more genes was found in five cases which in further histological examination showed morphological evidence of in situ carcinoma . As a result of the comparative analysis of the study groups five candidate genes were selected for the genetic markers panel for differential prostate pathology diagnosis.
Article
Clinical genomic testing is becoming routine in prostate cancer, as biomarker‐driven therapies such as poly‐ADP ribose polymerase (PARP) inhibitors and anti‐PD1 immunotherapy are now approved for select men with castration‐resistant prostate cancer harboring alterations in DNA repair genes. Challenges for precision medicine in prostate cancer include an overall low prevalence of actionable genomic alterations and a still limited understanding of the impact of tumor heterogeneity and co‐occurring alterations on treatment response and outcomes across diverse patient populations. Expanded tissue‐based technologies such as whole‐genome sequencing, transcriptome analysis, epigenetic analysis, and single‐cell RNA sequencing have not yet entered the clinical realm and could potentially improve upon our understanding of how molecular features of tumors, intratumoral heterogeneity, and the tumor microenvironment impact therapy response and resistance. Blood‐based technologies including cell‐free DNA, circulating tumor cells (CTCs), and extracellular vesicles (EVs) are less invasive molecular profiling resources that could also help capture intraindividual tumor heterogeneity and track dynamic changes that occur in the context of specific therapies. Furthermore, molecular imaging is an important biomarker tool within the framework of prostate cancer precision medicine with a capability to detect heterogeneity across metastases and potential therapeutic targets less invasively. Here, we review recent technological advances that may help promote the future implementation and value of precision oncology testing for patients with advanced prostate cancer.
Article
Background Hypermethylation of CpG islands in the promoter regions of tumor suppressor genes is one mechanism of tumorigenesis. Caveolin‐1 (Cav‐1), a gene coding for the structural component of cellular caveolae, is involved in cell signaling and has been proposed to be a tumor suppressor gene in several malignancies. This gene maps to 7q31.1, a site known to be deleted in some prostate tumors. We chose to examine the methylation status of the promoter region of Cav‐1 to determine whether this gene could function as a tumor suppressor in prostate cancer Methods Genomic DNA from both tumor and normal prostate epithelial cells was obtained from paraffin‐embedded prostate sections by laser capture microdissection (LCM). The methylation status of 24 CpG sites at the 5′ promoter region of Cav‐1 was analyzed by bisulfite‐direct‐sequencing after amplification by PCR using primers specific for bisulfate modified DNA. Immunohistochemistry staining with a cav‐1‐specific antibody was also performed to evaluate the expression of the gene Results Twenty of the 22 (90.9%) informative cases showed promoter hypermethylation in the tumor cell population when compared with adjacent normal prostate cells with an average Methylation Index (potential frequency of total possible methylated Cs) from tumor cells equal to 0.426 vs. 0.186 for normal cells (P = 0.001). While no association with Gleason grade was found, overall increased methylation correlated with PSA failure (P = 0.016), suggestive of clinical recurrence. Elevated immunoreactivity with a Cav‐1 antibody was observed in tumor cells from 7 of 26 prostate samples tested; this was associated with a Gleason score but not correlated with PSA failure or Methylation Index Conclusions CpG sites at the 5′ promoter of Cav‐1 are more methylated in tumor than in adjacent normal prostate cells. Hypermethylation of the Cav‐1 promoter supports the notion that Cav‐1 may function as a tumor suppressor gene in prostate cancer and evidence is presented suggesting that methylation status of this gene is not only a marker for cancer but also may be predictive of outcome. Prostate 46:249–256, 2001. © 2001 Wiley‐Liss, Inc.
Article
BACKGROUND Downregulation of genes which negatively control cell cycle progression represents a possible mechanism for prostate tumorigenesis. We examined the expression levels of the p16, p15, p14, and retinoblastoma‐susceptibility (RB) genes in primary prostate cancers and human prostate cancer cell lines, and correlated this with the DNA methylation levels of two loci in p16 . METHODS The mRNA levels of p16, p15, and p14 were examined by reverse transcriptase‐PCR (RT‐PCR). DNA methylation of the p16 5′ CpG island was determined by bisulfite genomic sequencing, while methylation of exon 2 shared by the p16 and p14 genes was measured by a quantitative bisulfite‐based technique, methylation‐sensitive single‐nucleotide primer extension (Ms‐SNuPE). RB protein levels were assessed by immunohistochemical staining of histologic sections of normal and tumor prostate tissues, using a monoclonal antibody (mAB). RESULTS Overexpression of p16 mRNA was found in 6/9 (67) of prostate tumors compared to the adjacent normal prostate, whereas elevated p14 and p15 levels were only observed in 2/9 (22) and 1/6 (17) of prostate cases, respectively. There was no statistically significant association of grade (P = 0.18) and stage (P = 1.00) of prostate cancer to the elevated p16 levels in the tumors. The p16 5′ CpG island was completely unmethylated in these tissues. In contrast, exon 2 of p16/p14 was methylated in both the tumor and normal adjacent prostates, and was increased in 8/11 (73) of tumors relative to normal tissues. There was no association between p16 overexpression and increased p16/p14 exon 2 methylation in these tumors (P = 1.00). Diminished RB levels in prostate tumors that had upregulated p16 mRNA were found, although absent RB was also detected in tumors without elevated p16 levels. The expression levels of the two genes, RB and p16 , were not correlated statistically (P = 0.16). CONCLUSIONS Our studies show that although the levels of the cell cycle regulators p16, p15, p14 , and Rb are altered in prostate cancers, there is no apparent correlation to grade, stage, or any pattern of regulation between the related genes. Exon 2 of p16/p14 is methylated in a majority of prostate tumors compared to the unmethylated upstream 5′ region, and may be a potential tumor marker for human prostate cancer. Prostate 43:233–242, 2000. © 2000 Wiley‐Liss, Inc.
Article
BACKGROUND Alterations of DNA methylation have been reported in many human cancers. In prostatic carcinoma, hypermethylation of the GST P gene promoter and an overall decrease in methylcytosine content have been reported. The aim of the present study was to investigate the frequency and extent of these alterations in relation to tumor stage and grade, in order to explore their clinical relevance and to determine their relationship to each other. METHODS DNA from 32 histologically verified adenocarcinomas of the prostate was analyzed for GST P hypermethylation by a semiquantitative PCR method and for overall DNA methylation by quantitative Southern blot analysis or LM‐PCR of LINE‐1 repetitive sequence methylation. RESULTS GST P hypermethylation was detected in 24/32 (75%) specimens, and LINE‐1 hypomethylation in 17/32 (53%). Both alterations tended to increase in frequency and extent with tumor stage. All but 1 of 8 carcinomas with lymph node involvement were positive for GST P hypermethylation. Six of these as compared to 2 out of 24 showed strong hypomethylation (P = 0.005). Hypermethylation and hypomethylation did not show a quantitative correlation, but all except two samples with weak LINE‐1 hypomethylation also displayed GST P hypermethylation. CONCLUSIONS GST P hypermethylation is an extremely frequent change in prostatic carcinoma which most probably precedes genome‐wide hypomethylation. It appears useful for sensitive detection of prostatic carcinoma, whereas pronounced LINE‐1 hypomethylation may be associated with progressive tumors. Prostate 39:166–174, 1999. © 1999 Wiley‐Liss, Inc.
Article
The GSTP1 gene encodes for an enzyme, glutathione S-transferase pi (GSTpi), involved in detoxification of carcinogens. An amino acid substitution (I105V) in GSTP1 produces a variant enzyme with lower activity and less capability of effective detoxification. This variant GSTP*B allele has been associated with a propensity to develop several neoplasms. Because GSTP1 promoter hypermethylation and inactivation of GSTpi, expression is a frequent alteration in prostate carcinoma, we hypothesized that this somatic epigenetic modification could obviate any reduced enzyme activity caused by the germ-line polymorphism. We tested for the GSTP1 genotype in a population of prostate cancer patients, and in a control group composed of patients with benign prostatic hyperplasia (BPH) and healthy blood donors. Tissue samples from the 105 prostate cancer cases (105 adenocarcinomas and 34 prostatic intraepithelial neoplasia lesions), and from 43 BPH patients were tested for GSTP1 hypermethylation by methylation-specific PCR. GSTpi protein expression was assessed by immunohistochemistry. No significant effect on prostate cancer risk was detectable for GSTP1 genotype compared with the control population (odds ratio, 1.02; 95% confidence interval, 0.59-1.75). Moreover, no association was found between this genotype and tumor or BPH methylation status. Patients with unmethylated carcinomas did not disclose significant differences in genotypic distribution compared with the control population. In adenocarcinoma, a strong association (P < 0.00001) between GSTP1 promoter hypermethylation and loss of GSTπ expression was observed; however, this trend was not retained in prostatic intraepithelial neoplasia or BPH lesions. Although the GSTP1 polymorphism is not associated with altered susceptibility to prostate cancer, somatic promoter hypermethylation is an effective, but not the only, cause of decreased GSTπ function.
Article
Inactivation of tumour suppressor gene(s) (TSGs) on 3p appears to be a critical event in the pathogenesis of clear cell renal cell carcinoma (CC-RCC). Analysis of loss of heterozygosity (LOH) in sporadic RCC samples has implicated roles for TSGs in three specific regions of 3p in RCC development: (1) 3p12-p14, which includes the breakpoint of the familial t(3;8) constitutional translocation involved in hereditary RCC development and a recently cloned putative TSG, the FHIT gene; (2) 3p21.2-p21.3, a common region of deletion in many cancers including lung; and (3) 3p25-p26, which contains the von Hippel–Lindau (VHL) disease TSG. We and others have shown that most primary sporadic CC-RCCs contain somatic VHL gene mutations, clearly implicating inactivation of the VHL gene in the pathogenesis of CC-RCC. It is not known if CC-RCC without VHL gene mutations have alternative mechanisms of VHL gene inactivation or result from an alternative non-VHL pathway to RCC, e.g., inactivation of TSGs in 3p12-p21. We and others have reported hypermethylation and silencing of the VHL TSG in RCC from patients with VHL disease and in CC-RCC cell lines. However, the incidence and specificity of VHL methylation in primary sporadic RCC has not been defined. Therefore, we analysed methylation of the VHL, CDKN2, MYC, and H19 genes in primary RCC samples. Hypermethylation of the VHLpromoter region was detected in 11% (11/99) of the primary RCCs analysed. In 10 of these tumours, there was no evidence of concomitant VHLgene mutation. VHL methylation was specific to CC-RCC (15%, 7/45) but was not detected in any non-CC tumours (n = 16). None of the 11 RCCs methylated at VHL had evidence of methylation at either CDKN2 or MYC (methylation at CDKN2 was, however, detected in 3%, or 1/33, of RCCs without VHL methylation). A normal methylation pattern at H19 was demonstrated in the three RCCs with methylated VHL analysed. Previous studies have suggested that, in addition to VHL, other 3p TSGs at 3p12-p14 and 3p21 may be involved in CC-RCC tumourigenesis. However, the interpretation of these studies has been difficult because information on VHL gene status has not been available for these data sets. Therefore, we investigated a subset of 55 sporadic RCCs (of known VHL gene methylation and mutation status) for LOH at polymorphic markers close to candidate TSG loci in the 3p14.2 and 3p21.2-p21.3 regions. Among tumours with LOH at one or more 3p markers, the incidence of 3p25 allele loss was higher in tumours with VHL alterations (mutation or methylation) than in those without. For tumours without detectable VHL alterations, the frequency of 3p14-p21 LOH was significantly higher than the frequency of 3p25-p26 LOH (93%, 13/14 vs. 43%, 6/14; P = 0.013), whereas, in RCC samples with VHL methylation or mutation, the frequency of 3p14-p21 LOH did not differ from that of 3p25-p26 (72%, 18/25 vs. 59%, 13/22; P = 0.376). None of the 11 RCCs with 3p25 allele loss that were informative at 3p21 and 3p14 showed LOH at 3p25 only. These findings suggest that (1) VHL methylation is a specific and important event in the pathogenesis of CC-RCC; (2) in CC-RCC with 3p LOH but without VHL inactivation, mutations in TSGs at 3p14-p21 appear to have a primary role in tumourigenesis; and (3) inactivation of other 3p TSGs in addition to VHL may also be required for malignant transformation in tumours with VHL gene inactivation. Genes Chromosomes Cancer 22:200–209, 1998.
Article
To investigate the role of epigenetic gene silencing in the pathogenesis of Wilms' tumour and renal cell carcinoma (RCC), we determined their methylation profile using a candidate gene approach. Thus, 40 Wilms' tumours and up to 49 adult RCC were analysed by methylation-specific PCR for promoter methylation at CASP8, CDH1, CDH13, DAPK, MGMT, NORE1A, p14ARF and RARB2 in primary Wilms' tumours and CASP8, CDH1, CDH13, CRBP1, DAPK, MGMT, MT1G, NORE1A, P16INK4a, SDHB and RARB2 in primary RCC. Both tumour sample sets had previously been analysed for RASSF1A promoter methylation, and p16 INK4a methylation results were also available for the Wilms' tumour samples. Wilms' tumours demonstrated a high incidence of methylation at CASP8 (43%) and MGMT (30%), intermediate frequencies at NORE1A (15%), p14 ARF (15%), p16INK4a (10%), DAPK (11%) and CRBP1 (9%), but promoter methylation was rare or absent at RARB2 (0%), CDH13 (0%) and CDH1 (3%). No association was detected between methylation of RASSF1A, CASP8 or MGMT in individual tumours. The frequency of MGMT methylation was higher in stage 1 and 2 tumours (50%) than in stage 3 and 4 tumours (17%) but this did not reach statistical significance (P = 0.06). RCC were most frequently methylated at DAPK (24%), MT1G (20%), NORE1A (19%), CDH1 (16%) and MGMT (9%) and not or rarely at SDHB (4%), RARB2 (0%), p16INK4a (0%) and CDH13 (3%). There were no associations between methylation of RASSF1A, DAPK and CDH1 in individual tumours. Papillary RCC demonstrated a higher frequency of DAPK methylation (43%) than clear cell tumours (19%) (P = 0.14). We have demonstrated that de novo promoter methylation is frequent in Wilms' tumour and RCC, and these data enable methylation profiles to be constructed for each tumour type. Thus, combining our results with data published previously, it appears that promoter methylation occurs frequently (≥20% of primary tumours) at CASP8, SLIT2 and RASSF1A in Wilms' tumour and at RASSF1A, TIMP3, DAPK, SLIT2, MT1G and GSTP1 in RCC.