ArticlePDF Available

Protein profiling of post-prostatic massage urine specimens by surface-enhanced laser desorption/ionization time-of-flight mass spectrometry to discriminate between prostate cancer and benign lesions

Authors:

Abstract and Figures

Post-prostatic massage urine specimens (PMUS) are expected to be rich in proteins originating from the prostatic acini. In this study, we created a PMUS bank consisting of 57 samples obtained from patients with biopsy-proven prostate cancer (PC) and 56 samples from subjects with biopsy-proven benign lesions to analyze protein profiles of PMUS by surface-enhanced laser desorption/ionization time-of-flight mass spectrometry (SELDI-TOF MS). Strong anion-exchange (Q10), weak cation-exchange (CM10) and immobilized metal affinity capture (IMAC30) ProteinChip Arrays were used for protein profiling. In PC samples, single-marker analysis detected 49 mass peaks that were significantly up-regulated and 23 peaks that were significantly down-regulated, compared with peaks obtained from benign lesion samples. To confirm reproducibility we performed additional three rounds of assay using CM10 chip with pH 4.0 binding buffer. Among these significant peaks, a peak of m/z 10788 was significant throughout all 4 rounds of assays. For hierarchical clustering analysis (HCA), we used the 72 peaks which revealed significant differences in single-marker analysis. The heat map discriminated PC from benign lesions with a sensitivity of 91.7% and a specificity of 83.3%. Therefore, SELDI-TOF MS profiling of PMUS can be applied to differentiate patients with PC from cancer-free subjects. However, further investigation is required to verify the usefulness of this method in clinical practice.
Content may be subject to copyright.
Abstract. Post-prostatic massage urine specimens (PMUS) are
expected to be rich in proteins originating from the prostatic
acini. In this study, we created a PMUS bank consisting of
57 samples obtained from patients with biopsy-proven prostate
cancer (PC) and 56 samples from subjects with biopsy-proven
benign lesions to analyze protein profiles of PMUS by
surface-enhanced laser desorption/ionization time-of-flight
mass spectrometry (SELDI-TOF MS). Strong anion-exchange
(Q10), weak cation-exchange (CM10) and immobilized metal
affinity capture (IMAC30) ProteinChip Arrays were used for
protein profiling. In PC samples, single-marker analysis
detected 49 mass peaks that were significantly up-regulated
and 23 peaks that were significantly down-regulated, compared
with peaks obtained from benign lesion samples. To confirm
reproducibility we performed additional three rounds of assay
using CM10 chip with pH 4.0 binding buffer. Among these
significant peaks, a peak of m/z 10788 was significant
throughout all 4 rounds of assays. For hierarchical clustering
analysis (HCA), we used the 72 peaks which revealed
significant differences in single-marker analysis. The heat map
discriminated PC from benign lesions with a sensitivity of
91.7% and a specificity of 83.3%. Therefore, SELDI-TOF MS
profiling of PMUS can be applied to differentiate patients with
PC from cancer-free subjects. However, further investigation
is required to verify the usefulness of this method in clinical
practice.
Introduction
Prostate cancer (PC) is the most common type of cancer in
men and the second highest cause of cancer death in the
United States (1). Recently, mortality rates for PC have been
increasing dramatically in Japan (2). Early detection of PC has
become easier by measuring prostate-specific antigen (PSA);
however, an urgent need exists for novel biomarkers to
improve the specificity of PC detection.
A number of innovations have been made to improve the
specificity of PSA testing. The most successful of these,
measurement of alternative molecular forms of PSA expressed
as the percentage of free PSA, improves the diagnostic
specificity of PSA testing (3,4) and can decrease the number
of false-negative prostate biopsies by ~20-25% (5). Moreover,
PSA velocity, age-specific PSA, PSA density and proPSA
have been postulated to improve the specificity of PSA testing
(6). However, the incidence of PC is shown as high as 22%
among men with a normal PSA range, 2.6-4.0 ng/ml (7).
Furthermore, PSA testing is almost organ-specific, but not
cancer-specific, because elevated serum concentrations are
also found in benign diseases, such as benign prostatic
hypertrophy and prostatitis. Therefore, great emphasis has
been placed on the need to discover novel biomarkers for use
in PC diagnosis.
Proteomic techniques applied to serum or plasma may
provide valuable information regarding protein markers or
patterns of markers that could possibly be used to improve
cancer detection (8). Serum protein profiling with surface
enhanced laser desorption/ionization time-of-flight mass
spectrometry (SELDI-TOF MS) has been shown to detect
cancers, including PC (9). In addition, several case-control
studies have reported excellent validity for PC detection
(10-13).
Serum proteomic approaches have not provided useful
biomarkers for PC in a clinical setting. In order to address this
problem, we conducted a proteomic study on protein
originating from prostate acini obtained by non-invasive
sampling. Post-prostatic massage urine specimens (PMUS),
ONCOLOGY REPORTS 21: 73-79, 2009 73
Protein profiling of post-prostatic massage urine
specimens by surface-enhanced laser desorption/ionization
time-of-flight mass spectrometry to discriminate
between prostate cancer and benign lesions
AKIKO OKAMOTO1, HAYATO YAMAMOTO1, ATSUSHI IMAI1, SHINGO HATAKEYAMA1,
IKUYA IWABUCHI1, TAKAHIRO YONEYAMA1, YASUHIRO HASHIMOTO1, TAKUYA KOIE1,
NORITAKA KAMIMURA1, KAZUYUKI MORI1, KANEMITSU YAMAYA2and CHIKARA OHYAMA1
1Department of Urology, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562;
2Oyokyo Kidney Research Institute, 90 Kozawa Yamazaki, Hirosaki 036-8243, Japan
Received August 5, 2008; Accepted September 29, 2008
DOI: 10.3892/or_00000191
_________________________________________
Correspondence to: Dr Chikara Ohyama, Department of Urology,
Hirosaki University Graduate School of Medicine, 5 Zaifu-cho,
Hirosaki 036-8562, Japan
E-mail: coyama@cc.hirosaki-u.ac.jp
Key words: protein profile, prostate cancer, prostate massage
73-79 3/12/2008 10:18 Ì ™ÂÏ›‰·73
which have been established as diagnostic samples for
prostatitis (14), are expected to be rich in proteins originating
from prostatic acini. Moreover, to our knowledge, this is the
first detailed study describing protein profiling of PMUS by
SELDI-TOF MS.
Materials and methods
Post-prostatic massage urine specimen (PMUS). A flowchart
of this study is illustrated in Fig. 1. PMUS was collected
after digital rectal examination (5 strokes per lobe). Urine
was voided into urine collection cups, briefly centrifuged
(10 min at 2,000 x g), aliquotted, frozen immediately and
stored at -80˚C until protein profile analysis. The PMUS
bank consisted of 57 samples from patients with biopsy-
proven PC and 56 samples from subjects with biopsy-proven
benign lesions. The study was approved by the Institutional
Ethics Committee and a written consent was obtained from
all subjects who participated in the study.
Protein concentration measurement and prostate-specific
antigen assay. Protein concentration of PMUS was
measured by Immage 800 (Beckman Coulter Inc., Brea, CA,
USA) and prostate-specific antigen (PSA) in serum was
measured by Immulite 1000 (Siemens, Deerfield, IL, USA).
Prostate biopsy and pathological diagnosis. After collection
of PMUS, 10 or 12 prostate needle biopsy samples were
transrectally obtained by ultrasound guidance, using an 18 G
needle. The 2002 TNM staging system (15) was used to assign
the stage and the up-dated Gleason grading system from the
International Society of Urological Pathology (ISUP) (16) was
used for tumor grading.
Analysis of protein profiles. PMUS samples were briefly
centrifuged (10 min at 20,000 x g) and the supernatants
were subjected to protein profiling. Protein profiles of the
PMUS samples were obtained by using weak cation-
exchange (CM10), strong anion-exchange (Q10) and
immobilized metal affinity capture (IMAC30) ProteinChip
Arrays (Bio-Rad, Fremont, CA, USA). The ProteinChip
Arrays were assembled into a deep-well type Bioprocessor
assembly (Bio-Rad). Prior to sample loading, Q10 and
CM10 arrays were equilibrated with 150 μl of binding
buffer (for Q10, 50 mM Tris-HCl, pH 8.0; for CM10, 100 mM
sodium acetate, pH 4.0 and 50 mM HEPES, pH 7.0).
Before the samples were loaded, IMAC30 arrays were
charged with Cu2+ by adding 50 μl of 100 mM CuSO4.
After incubation for 5 min, the arrays were quickly rinsed
with water to remove the unbound metal and the surface
was further washed with 50 μl of 100 mM sodium acetate,
pH 4.0. The arrays were then equilibrated with 150 μl of
binding buffer (100 mM sodium phosphate with 0.5 M NaCl,
pH 7.0).
A 10 μl-portion of PMUS was mixed with 30 μl of 2%
CHAPS/9 M urea/50 mM Tris-HCl, pH 9.0 and further diluted
with 60 μl of binding/washing buffer. All arrays were then
incubated with 100 μl of diluted sample for 60 min on a shaker
and washed 3 times with 150 μl of binding buffer. After rinsing
with water, the arrays were removed from the Bioprocessor
assembly and air-dried. After air-drying, a 1.0 μl aliquot of
50% saturated sinapinic acid solution (dissolved in 50%
acetonitrile containing 0.5% trifluoroacetic acid) was added
twice and allowed to dry.
The ProteinChip Arrays were then transferred to the
ProteinChip System Series 4000 (Bio-Rad) which generates
nanosecond laser pulses from a UV-emitting pulsed nitrogen
laser (373 nm). External mass calibration was performed with
protein standards: porcine dynorphin (2148 Da), human
adrenocorticotropic hormone (2934 Da), bovine insulin ß-chain
(3496 Da), human insulin (5808 Da), recombinant hirudin
(6964 Da), bovine cytochrome C (12230 Da), equine
myoglobin (16951 Da), bovine carbonic anhydrase (29023 Da)
and enolase from Saccharomyces cerevisiae (46671 Da). All
assays were repeated twice.
The protein expression patterns were analyzed using
CiphergenExpress Data Manager software, version 3.0
(Bio-Rad), which generates consistent mass peak sets
(clusters) across multiple spectra and enables automatic
comparison. Each cluster was treated as a single protein or
peptide fragment. All data were normalized by the software's
total ion current normalization function, following the
manufacturer's instructions. Spectra between 2500 and
150000 mass-to-charge ratios (m/z) were selected for analysis.
Automatic peak detection was carried out for peaks with
signal/noise ratio >2.5. The Mann-Whitney U test was used to
compare intensities of clustered peaks between the 2 sample
groups.
Single-marker analysis. To identify a candidate peak, we used
CM10, Q10 and IMAC 30 chips. To confirm reproducibility,
we carried out additional 3 rounds of analysis by CM10
chip with pH 4.0 binding buffer. PMUS were randomly
selected from each group for each round. For the first round
analysis, we randomly selected 12 samples each from the
PC-PMUS pool and the benign lesion-PMUS pool. For the
second, third and fourth round analyses, we randomly
selected 10, 8 and 29 samples from each group, respectively
(Fig. 1). Finally, 37 PC samples and 39 benign lesion
samples were examined for single-marker analysis.
Demographic data on the subjects are shown in Table I.
OKAMOTO et al: PROTEIN PROFILE OF URINE AFTER PROSTATE MASSAGE
74
Figure 1. The study design. The PMUS sample bank consisted of 57 samples
from patients with biopsy-proven PC and 56 samples from subjects with
biopsy-proven benign lesions. PMUS samples were randomly selected for
protein profiling. Overall, 39 samples from benign lesions and 37 samples
from PC were subjected to single-marker analysis. Twelve samples from
each pool were subjected to hierarchical clustering analysis (HCA).
73-79 3/12/2008 10:18 Ì ™ÂÏ›‰·74
Hierarchical clustering analysis (HCA). HCA was performed
to create a heat map using CiphergenExpress Data Manager
software, version 3.0 (Bio-Rad). For HCA analysis, we
used the 72 peaks, which revealed significant differences in
single-marker analysis. Clinicopathological data on the
subjects whose PMUS were subjected to HCA are shown in
Table II.
Results
In normal urine samples from healthy subjects, protein cannot
be detected. However, protein concentration of PMUS was
successfully measurable in all specimens as shown in Table I.
Mean protein concentration of PMUS from patients with PC
was 102.3 μg/ml and that from benign lesion was 113.5 μg/ml.
ONCOLOGY REPORTS 21: 73-79, 2009 75
Table I. Demographic data on subjects for single-marker analysis.
–––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––
B (n=39) PC (n=37)
–––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––
Age (years) (mean; range) 68.0 (53-81) 70.3 (53-79)
Serum PSA (ng/ml) (mean; range) 6.9 (2.1-12.5) 15.7 (4.4-111.2)
PMUS protein conc. (μg/ml) (mean; range) 102.3 (27.2-353.5) 113.5 (25.1-338.1)
Gleason score (mean; range) - 7.3 (5-9)
Clinical stage - T1cN0M0-T4N1M1
–––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––
PMUS, post-prostate massage urine specimen; B, benign lesion and PC, prostate cancer.
–––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––
Table II. Clinicopathological data on subjects used for hierarchical clustering analysis (HCA).
–––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––
Case Age Serum PSA Pathology Gleason Clinical PMUS
(year) (ng/ml) score stage concentration (μg/ml)
–––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––
B1 74 6.8 B - - 140.4
B2 70 7.2 B - - 27.2
B3 72 7.3 B - - 123.6
B4 75 8.8 B - - 41.3
B5 61 2.5 B - - 278.1
B6 68 6.6 B - - 57.1
B7 57 6.6 B - - 76.0
B8 60 12.3 B - - 57.1
B9 66 4.1 B - - 38.1
B10 71 4.9 B - - 53.9
B11 70 8.3 B - - 34.9
B12 68 5.2 B - - 70.7
PC1 74 5.2 PC 7 T2aN0M0 45.7
PC2 76 12.2 PC 7 T1cN0M0 291.9
PC3 74 11 PC 7 T2aN0M0 271.3
PC4 67 15.2 PC 7 T3N0M1 25.1
PC5 72 5.8 PC 9 T3N0M0 46.5
PC6 71 76.8 PC 9 T3N0M0 40.7
PC7 76 9.2 PC 7 T2aN0M0 144.8
PC8 68 7.8 PC 9 T2aN0M0 102.3
PC9 75 15.5 PC 7 T2aN0M0 44.4
PC10 77 34.5 PC 7 T3N0M0 86.5
PC11 63 8.6 PC 7 T2aN0M0 86.5
PC12 75 8.8 PC 7 T2bN0M0 138.1
–––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––
PMUS, post-prostate massage urine specimen; B, benign lesion and PC, prostate cancer.
–––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––
73-79 3/12/2008 10:18 Ì ™ÂÏ›‰·75
PMUS protein concentration ranged from 25.1 μg/ml to
353.5 μg/ml. Therefore, we diluted PMUS sample with 2%
CHAPS/9 M urea/50 mM Tris-HCl to obtain final protein
concentration of 25.0 μg/ml.
For single-marker analysis, peak intensities detected by
three kinds of chips were compared between the 2 groups.
As a demonstrable example, protein profiles of the first
assay round are presented in Fig. 2. The peak intensity of m/z
4761 in the PC group was significantly higher than peaks in
the benign lesion group (P=0.0010; Fig. 3a). The receiver
OKAMOTO et al: PROTEIN PROFILE OF URINE AFTER PROSTATE MASSAGE
76
Figure 2. Protein profiling using CM10 chip with pH 4.0 buffer. The peak
intensity of m/z 4761, surrounded by line, was markedly higher in the PC
group than in the benign lesion group.
Figure 4. Heat map based on the results of protein profiling, using hierarchical clustering analysis. Horizontal line below the heat map represents case number.
Vertical line represents the 72 significant peaks and chip conditions, which correspond to peak information presented in Table IV. According to the heat map,
we were able to discriminate PC from benign lesions with 91.7% sensitivity and 83.3% specificity. B, benign lesion and PC, prostate cancer.
Figure 3. Difference in peak intensity of m/z 4761 between PC and benign
lesion groups. (a) The peak intensity of m/z 4761 was significantly higher in
the PC group (P=0.0010). (b) The receiver operating characteristic curve
(ROC) was plotted for m/z 4761. The area under the curve (AUC) on the ROC
plot was 0.917.
73-79 3/12/2008 10:18 Ì ™ÂÏ›‰·76
operating characteristic curve (ROC) of m/z 4761 is shown in
Fig. 3b. The area under the curve (AUC) on the ROC was
0.917.
In PC samples, single-marker analysis detected 49 mass
peaks that were significantly up-regulated and 23 peaks that
were significantly down-regulated, compared with peaks
obtained from benign lesion samples. Statistical data and
chip conditions of these peaks are shown in Table III.
To confirm reproducibility, we repeated the assay four
times in total using by CM10 chip with pH 4.0 binding buffer.
Random selection from PMUS bank for repeated single-
marker analysis caused some overlaps of samples. So, finally
we analyzed 37 PC samples and 39 benign lesion samples.
Results with repeated single-marker analysis are summarized
in Table IV. Although the significantly increased or decreased
peaks varied in each assay round, peaks of m/z 10788 showed
significantly lower intensity in the PC group throughout all
assay rounds. The peak of m/z 5384, which showed
significantly lower intensity in 3 assay rounds, is deduced to
be a double charge of the peak of m/z 10788.
ONCOLOGY REPORTS 21: 73-79, 2009 77
Table III. Statistical data and chip conditions of significant
peaks detected in single-marker analysis.
–––––––––––––––––––––––––––––––––––––––––––––––––
M/Z P-value ROC area Chip condition
–––––––––––––––––––––––––––––––––––––––––––––––––
Up-regulated in PC
2670 0.0027 0.861 CM10 pH 4.0
2776 0.0282 0.750 CM10 pH 4.0
2797 0.0243 0.722 CM10 pH 4.0
2978 0.0056 0.806 CM10 pH 4.0
3003 0.0010 0.861 CM10 pH 4.0
3023 0.0005 0.889 CM10 pH 4.0
3174 0.0282 0.778 Q10 pH 8.0
3204 0.0496 0.722 IMAC30
3272 0.0111 0.778 IMAC30
3375 0.0079 0.806 Q10 pH 8.0
3461 0.0012 0.889 CM10 pH 4.0
3496 0.0022 0.889 CM10 pH 4.0
3721 0.0018 0.861 CM10 pH 4.0
3773 0.0027 0.833 CM10 pH 4.0
3786 0.0496 0.722 Q10 pH 8.0
3851 0.0022 0.861 CM10 pH 4.0
3897 0.0377 0.750 CM10 pH 4.0
3938 0.0209 0.750 CM10 pH 4.0
3997 0.0243 0.778 Q10 pH 8.0
4026 0.0079 0.806 CM10 pH 7.0
4028 0.0002 0.917 CM10 pH 4.0
4056 0.0243 0.778 Q10 pH 8.0
4478 0.0377 0.750 CM10 pH 4.0
4544 0.0002 0.944 CM10 pH 4.0
4582 0.0243 0.778 CM10 pH 4.0
4761 0.0010 0.917 CM10 pH 4.0
4763 0.0282 0.778 CM10 pH 7.0
4781 0.0022 0.889 CM10 pH 4.0
4828 0.0153 0.778 Q10 pH 8.0
4862 0.0209 0.778 Q10 pH 8.0
4968 0.0001 0.944 CM10 pH 4.0
5017 0.0047 0.833 Q10 pH 8.0
5817 0.0209 0.778 CM10 pH 4.0
6200 0.0377 0.750 Q10 pH 8.0
6481 0.0027 0.833 CM10 pH 4.0
8030 0.0039 0.833 IMAC30
8037 0.0007 0.889 CM10 pH 4.0
8202 0.0179 0.778 CM10 pH 4.0
8309 0.0179 0.806 CM10 pH 4.0
8871 0.0056 0.806 CM10 pH 4.0
9098 0.0056 0.806 CM10 pH 4.0
9102 0.0327 0.722 IMAC30
9207 0.0433 0.750 CM10 pH 4.0
9281 0.0047 0.833 CM10 pH 4.0
9780 0.0111 0.833 CM10 pH4.0
Table III. Continued.
–––––––––––––––––––––––––––––––––––––––––––––––––
M/Z P-value ROC area Chip condition
–––––––––––––––––––––––––––––––––––––––––––––––––
Up-regulated in PC
9905 0.0056 0.806 CM10 pH 4.0
9990 0.0209 0.750 CM10 pH 4.0
-------------------------------------------------------------------------
Down-regulated in PC
4702 0.0496 0.778 Q10 pH 8.0
4827 0.0496 0.722 IMAC30
5333 0.0496 0.722 CM10 pH 4.0
5339 0.0433 0.750 Q10 pH 8.0
5384 00039 0.833 CM10 pH 4.0
5395 0.0067 0.806 Q10 pH 8.0
7281 0.0153 0.778 Q10 pH 8.0
7589 0.0094 0.778 Q10 pH 8.0
7764 0.0027 0.861 Q10 pH 8.0
10668 0.0111 0.806 Q10 pH 8.0
10677 0.0153 0.778 IMAC30
10678 0.0153 0.806 CM10 pH 4.0
10778 0.0094 0.778 IMAC30
10782 0.0079 0.806 Q10 pH 8.0
10788 0.0067 0.806 CM10 pH 4.0
10888 0.0179 0.806 CM10 pH 4.0
10985 0.0067 0.833 Q10 pH 8.0
10995 0.0111 0.806 CM10 pH 4.0
11201 0.0153 0.778 CM10 pH 4.0
11397 0.0079 0.833 CM10 pH 4.0
13909 0.0111 0.833 IMAC30
28094 0.0433 0.750 IMAC30
38025 0.0111 0.778 IMAC30
–––––––––––––––––––––––––––––––––––––––––––––––––
73-79 3/12/2008 10:18 Ì ™ÂÏ›‰·77
To create a heat map 72 significant peaks which identified
in the first round single-marker analysis were used. According
to the heat map based on the data from these 72 significant
peaks (Fig. 4), we were able to discriminate PC from benign
lesions with a sensitivity of 91.7% and a specificity of
83.3%.
Discussion
SELDI-TOF and matrix-assisted laser desorption/
ionization-TOF MS have been recognized as the most
common techniques for protein profiling (17). These
techniques have been applied to discover a novel biomarker
for PC (11-13,18). However, recent studies emphasize on
the limited usefulness of proteomic approach for identifying
candidates for serum proteins (19). To overcome these
problems, we conducted a proteomic study using PMUS. Most
proteins synthesized in prostatic epithelium are secreted
into prostatic acini and drained into the prostatic duct.
Thus, PMUS is expected to be rich in proteins originating
from prostatic epithelium. As demonstrated in the present
study, protein concentration in PMUS was much higher than
in urine. Moreover, this finding suggests that an abundant
source of proteins in PMUS originates from the prostatic
acini.
Lack of reproducibility in SELDI-TOF MS whole-serum
proteomic profiling has also been noted (20). To overcome the
weak point in reproducibility, we repeated our assays. For
single-marker analysis we performed 4 rounds of SELDI-TOF
MS analysis.
In the first single-marker analysis, we found 49 peaks that
were significantly up-regulated in the patients with PC and
23 peaks that were significantly down-regulated. During the
4 rounds of assays, significant peaks varied from round to
round. Among the significant peaks, a peak of m/z 10788
remained significant throughout all the rounds. Furthermore,
the peak of m/z 5384, which showed significantly lower
intensity in 3 assay rounds, may be a double charge of the peak
of m/z 10788. Therefore, we believe that the peak of m/z
10788 could be a promising single-marker for early detection
of PC. Further study is required focusing on the structural
analysis for this peak.
For HCA analysis, we used 72 peaks that proved significant
in the first-round assay. In spite of the small sample size, we
were able to discriminate biopsy-proven PC from benign
lesions with high sensitivity and specificity by using the
heat map. Especially, its high specificity of 83.3% is
remarkably higher than that of PSA test for the detection of
PC (6,7). However, this method along with single-marker
analysis, requires further investigation with a large number of
samples.
In this preliminary study, we postulated that promising
urine markers, originating from prostatic acini, can be obtained
by prostatic massage. Our assays identified a potential marker
to differentiate patients with PC from cancer-free subjects.
However, as stated previously (21), all candidate markers must
be strictly evaluated through multi-step checkpoints including
accurate methods for detecting markers, single institutional
pilot studies and rigorous validation in retrospective and
prospective studies.
Acknowledgements
This study was supported by a grant from the CREST (Core
Research for Evolutional Science and Technology) project of
the Japan Science and Technology Agency.
OKAMOTO et al: PROTEIN PROFILE OF URINE AFTER PROSTATE MASSAGE
78
Table IV. Significant peaks detected by repeated single-marker analysis using by CM10 with pH 4.0 binding buffer.
–––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––
Assay round
1st 2nd 3rd 4th
–––––––––––––––– –––––––––––––––– –––––––––––––––– ––––––––––––––––
P-value AUC P-value AUC P-value AUC P-value AUC
–––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––
Up-regulated
(m/z)
4761 0.0012 0.917 0.0112 0.861 NS - NS -
5817 0.0209 0.778 0.0413 0.742 NS - 0.0338 0.643
8037 0.0007 0.889 NS - NS - 0.0351 0.609
8871 0.0056 0.806 NS - NS - 0.017 0.679
9098 0.0218 0.806 NS - NS - 0.0218 0.648
9780 0.0111 0.833 NS - NS - NS -
Down-regulated
(m/z)
5384 0.0039 0.833 NS - 0.0157 0.844 0.0393 0.617
10788 0.0067 0.806 0.0162 0.783 0.0274 0.797 0.0474 0.644
–––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––
NS, not significant and AUC, area under the curve.
–––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––––
73-79 3/12/2008 10:18 Ì ™ÂÏ›‰·78
References
1. Stephenson RA: Prostate cancer trends in the era of prostate-
specific antigen. An update of incidence, mortality, and clinical
factors from the SEER database. Urol Clin North Am 29:
173-181, 2002.
2. Marugame T and Mizuno S: Comparison of prostate cancer
mortality in five countries: France, Italy, Japan, UK and USA
from the WHO mortality database (1960-2000). Jpn J Clin Oncol
35: 690-691, 2005.
3. Lilja H, Christensson A, Dahlen U, Matikainen MT, Nilsson O,
Pettersson K and Lovgren T: Prostate-specific antigen in serum
occurs predominantly in complex with ·1-antichymotrypsin.
Clin Chem 37: 1618-1625,1991.
4. Stenman UH, Leinonen J, Alfthan H, Rannikko S, Tuhkanen K
and Alfthan O: A complex between prostate-specific antigen
and ·1-antichymotrypsin is the major form of prostate-specific
antigen in serum of patients with prostatic cancer: assay of the
complex improves clinical sensitivity for cancer. Cancer Res 51:
222-226, 1991.
5. Catalona WJ, Partin AW, Slawin KM, et al: Use of the percentage
of free prostate-specific antigen to enhance differentiation of
prostate cancer from benign prostatic disease: a prospective
multicenter clinical trial. JAMA 279: 1542-1547, 1998.
6. Gretzer MB and Partin AW: PSA markers in prostate cancer
detection. Urol Clin North Am 30: 677-686, 2003.
7. Catalona WJ, Smith DS and Ornstein DK: Prostate cancer
detection in men with serum PSA concentrations of 2.6 to 4.0 ng/
mL and benign prostate examination. Enhancement of specificity
with free PSA measurements. JAMA 277:1452-1455, 1997.
8. Ornstein DK and Petricoin EF: Proteomics to diagnose human
tumors and provide prognostic information. Oncology 18:
521-519, 2004.
9. Bons JAP, Wodzig WKWH and van Dieijen-Visser MP: Protein
profiling as a diagnostic tool in clinical chemistry: a review. Clin
Chem Lab Med 43: 1281-1290, 2005.
10. Adam BL, Qu Y, Davis JW, et al: Serum protein fingerprinting
coupled with a pattern-matching algorithm distinguishes prostate
cancer from benign prostate hyperplasia and healthy men.
Cancer Res 62: 3609-3614, 2002.
11. Qu Y, Adam BL, Yasui Y, et al: Boosted decision tree analysis
of surface-enhanced laser desorption/ionization mass spectral
serum profiles discriminates prostate cancer from noncancer
patients. Clin Chem 148: 1835-1843, 2002.
12. Petricoin EF III, Ornstein DK, Paweletz CP, et al: Serum
proteomic patterns for detection of prostate cancer. J Natl Cancer
Inst 94: 1576-1578, 2002.
13. Ornstein DK, Rayford W, Fusaro VA, et al: Serum proteomic
profiling can discriminate prostate cancer from benign prostates
in men with total prostate specific antigen levels between 2.5 and
15 ng/ml. J Urol 172: 1302-1305, 2004.
14. Nickel JC: The Pre and Post Massage Test (PPMT), a simple
screen for prostatitis. Tech Urol 3: 38-43, 1997.
15. Greene FL, Page DL, Fleming ID, Fritz A, Balch CM, Haller DG
and Morrow M: AJCC. In: Cancer Staging Manual (eds). 6th
edition. Springer-Verlag, New York, pp317-312, 2002.
16. Epstein JI, Allsbrook WC Jr, Amin MB, Egevad LL and ISUP
Grading Committee, The 2005 International Society of Urological
Pathology (ISUP) Consensus Conference on Gleason Grading
of Prostatic Carcinoma. Am J Surg Pathol 29: 1228-1242,
2005.
17. Wulfkuhle JD, Liotta LA and Petricoin EF: Proteomic
applications for the early detection of cancer. Nat Rev Cancer 3:
267-275, 2003.
18. Partin AW and Chan DW: Detection of prostate cancer using
serum proteomics pattern in a histologically confirmed
population. J Urol 171: 1782-1787, 2004 .
19. McLerran D, Grizzle WE, Feng Z, et al: SELDI-TOF MS whole
serum proteomic profiling with IMAC surface does not reliably
detect prostate cancer. Clin Chem 54: 53-60, 2008.
20. Rogers MA, Clarke P, Noble J, Munro NP, Paul A, Selby PJ
and Banks RE: Proteomic profiling of urinary proteins in renal
cancer by surface enhanced laser desorption ionization and
neural-network analysis: identification of key issues affecting
potential clinical utility. Cancer Res 63: 6971-6983, 2003.
21. Parekh DJ, Ankerst DP, Troyer D, Srivastava S and
Thompson IM: Biomarkers for prostate cancer detection. J Urol
178: 2252-2259, 2007.
ONCOLOGY REPORTS 21: 73-79, 2009 79
73-79 3/12/2008 10:18 Ì ™ÂÏ›‰·79
... (177), it allows a hypothesis-free approach to novel biomarker discovery and subsequent evaluation. This approach has been used to identify proteomic patterns to differentiate diabetic from non-diabetic nephropathy (178,179), early acute kidney injury (180)(181)(182), acute renal transplant rejection (183,184), lupus nephritis (185,186) and urogenital malignancy (187)(188)(189). ...
... Although these limitations have generally limited the use of SELDI as a clinical tool (177), it allows a hypothesis-free approach to novel biomarker discovery and subsequent evaluation. This approach has been used to identify proteomic patterns to differentiate diabetic from non-diabetic nephropathy (178,179), early acute kidney injury (180)(181)(182), acute renal transplant rejection (183,184), lupus nephritis (185,186) and urogenital malignancy (187)(188)(189). Validation of these findings have led to the identification of candidate biomarkers for clinical utilitisation such as EN-2 in prostate cancer (220) or a panel of urine markers in lupus nephritis (221). ...
... Cancer cells secret proteases that can act on proteins present in the urine, ultimately leading to a differential abundance of urinary peptides in individuals with cancer (18,32). Previous studies have shown that endogenous urinary peptide signatures have diagnostic value for PCa, especially in discriminating between PCa and BPH, resulting in high sensitivity (67.4 to 91.7%) and specificity (71.2 to 90.5%) (33)(34)(35). Our urinary peptide panel outperformed PSA in discriminating between PCa and BPH, showing high levels of sensitivity (87.88%) and specificity (88%). ...
Article
Full-text available
Prostate cancer (PCa) is one of the most prevalent types of cancer in men worldwide; however, the main diagnostic tests available for PCa have limitations and a biopsy is required for histopathological confirmation of the disease. Prostate-specific antigen (PSA) is the main biomarker used for the early detection of PCa, but an elevated serum concentration is not cancer-specific. Therefore, there is a need for the discovery of new non-invasive biomarkers that can accurately diagnose PCa. The present study used trichloroacetic acid-induced protein precipitation and liquid chromatography-mass spectrometry to profile endogenous peptides in urine samples from patients with PCa (n=33), benign prostatic hyperplasia (n=25) and healthy individuals (n=28). Receiver operating characteristic curve analysis was performed to evaluate the diagnostic performance of urinary peptides. In addition, Proteasix tool was used for in silico prediction of protease cleavage sites. Five urinary peptides derived from uromodulin were revealed to be significantly altered between the study groups, all of which were less abundant in the PCa group. This peptide panel showed a high potential to discriminate between the study groups, resulting in area under the curve (AUC) values between 0.788 and 0.951. In addition, urinary peptides outperformed PSA in discriminating between malignant and benign prostate conditions (AUC=0.847), showing high sensitivity (81.82%) and specificity (88%). From in silico analyses, the proteases HTRA2, KLK3, KLK4, KLK14 and MMP25 were identified as potentially involved in the degradation of uromodulin peptides in the urine of patients with PCa. In conclusion, the present study allowed the identification of urinary peptides with potential for use as non-invasive biomarkers in PCa diagnosis.
... A number of clinical studies were performed using the SELDI-TOF-MS technique through urine analysis to validate the levels of common molecular markers for the major GI tumors [84][85][86]. In contrast, our study emphasized on only bioinformatics tools for prediction of specifically, miRNA biomarkers given their significance in the said sub-types and so no clinical validation process was further carried out unlike the aforementioned studies on urine. ...
Article
Full-text available
Background miRNAs are endogenous, non-coding and evolutionarily conserved RNA molecules. They have been found to be involved in the progression and proliferation of various cancers due to their contribution in post-transcriptional regulation. Stomach Adenocarcinoma (STAD) and Liver Hepatocellular Carcinoma (LIHC) are the two most common cancers of the upper intestinal tract. Our study aimed to evaluate the circulating miRNAs from both STAD and LIHC samples and to identify commonly dysregulated miRNAs as biomarkers to detect both cancers at the same time. Methods and Materials Common differentially expressed miRNAs (DEMs) from GEO and Bioexpress datasets were considered for initial processing in the analysis. Pathway analysis of the selected miRNAs through DIANA-miRpath tool, followed by survival analysis based on prognostic values through OncoLnc server led to the final biomarker candidates for diagnosis and prognosis of STAD and LIHC. An elaborate miRNA-gene-cancer network was set up for a specialized understanding of the selected DEMs corresponding to the specifically unique target genes and the cancer types. The gene ontology analysis was performed using BINGO to determine functional connotations of the differentially expressed genes (DEGs). Results After a thorough analysis, we found that the 4 miRNAs: miR-183-5p, miR-203-3p, miR-126-3p and miR-25-3p could be potential prognostic biomarkers against both STAD and LIHC. The differentially expressed genes (DEGs) for these miRNAs were inferred through GEPIA and miRwalk v2.0. The miRNA-gene-cancer network revealed that the commonly deregulated miRNAs could influence the same genes and pathways altered by multiple cancers at the same time- in our case, STAD and LIHC. To support our claim, we showed the gene ontology analysis by BINGO, attesting the functional assignment of the DEGs behind the metastasis and development of both the cancers. Conclusion Our study evaluated a particularly effective avenue of identifying novel miRNA for both early diagnostic and prognostic purposes against more than one cancer.
... In the clinical setting, urine is an optimal sample source, as it is easy to obtain, the collection is non-invasive, and is relatively stable in terms of sample integrity. A substantial number of previous studies have found the SELDI-TOF-MS technique ideally suited for urine analysis, with a combination of high throughput, speed and relatively low cost (40)(41)(42). However, a main drawback of this technique is the comparatively medium resolution of the spectra obtained, but this is adequate to resolve peaks in the 1-25-kDa range from spectra with <500 peaks. ...
Article
Full-text available
Several potential urinary biomarkers exhibiting an association with upper gastrointestinal tumour growth have been previously identified, of which S100A6, S100A9, rabenosyn‑5 and programmed cell death 6‑interacting protein (PDCD6IP) were further validated and found to be upregulated in malignant tumours. The cancer cohort from our previous study was subclassified to assess whether distinct molecular markers can be identified for each individual cancer type using a similar approach. Urine samples from patients with cancers of the stomach, oesophagus, oesophagogastric junction or pancreas were analysed by surface‑enhanced laser desorption/ionization‑time‑of‑flight mass spectrometry using both CM10 and IMAC30 (Cu2+‑complexed) chip types and LC‑MS/MS‑based mass spectrometry after chromatographic enrichment. This was followed by protein identification, pattern matching and validation by western blotting. We found 8 m/z peaks with statistical significance for the four cancer types investigated, of which m/z 2447 and 2577 were identified by pattern matching as fragments of cathepsin‑B (CTSB) and cystatin‑B (CSTB); both molecules are indicative of pancreatic cancer. Additionally, we observed a potential association of upregulated α‑1‑antichymotrypsin with pancreatic and gastric cancers, of PDCD6IP, vitelline membrane outer layer protein 1 homolog (VMO1) and triosephosphate isomerase (TPI1) with oesophagogastric junctional cancers, and of complement C4‑A, prostatic acid phosphatase, azurocidin and histone‑H1 with oesophageal cancer. Furthermore, the potential pancreatic cancer biomarkers CSTB and CTSB were validated independently by western blotting. Therefore, the present study identified two new potential urinary biomarkers that appear to be associated with pancreatic cancer. This may provide a simple, non‑invasive screening test for use in the clinical setting.
... An extension of MALDI-TOF that is widely used in proteomics is SELDI-TOF. A protein profiling study of post-prostatic massage urine specimens via SELDI-TOF revealed 49 mass peaks that were significantly up-regulated and 23 peaks that were significantly downregulated in CaP compared with non-cancerous controls (Okamoto et al., 2009). Since only the mass to charge (m/z) values can be obtained, combining urinary analysis of SELDI-TOF and follow-up identification approaches such as LC-MS/MS will most likely help in discovery of urine-based biomarker for CaP. ...
Article
Prostate cancer (CaP) is the most common cancer in men and the second leading cause of cancer deaths in males in Australia. Although serum prostate-specific antigen (PSA) has been the most widely used biomarker in CaP detection for decades, PSA screening has limitations such as low specificity and potential association with over-diagnosis. Current biomarkers used in the clinic are not useful for the early detection of CaP, or monitoring its progression, and have limited value in predicting response to treatment. Urine is an ideal body fluid for the detection of protein markers of CaP and is emerging as a potential source for biomarker discovery. Gene-based biomarkers in urine such as prostate cancer antigen-3 (PCA3), and genes for transmembrane protease serine-2 (TMPRSS2), and glutathione S-transferase P (GSTP1) have been developed and evaluated in the past decades. Among these biomarkers, urinary PCA3 is the only one approved by the FDA in the USA for clinical use. The study of urine microRNAs (miRNAs) is another burgeoning area for investigating biomarkers to achieve a pre-biopsy prediction of CaP to contribute to early detection. The development of mass spectrometry (MS)-based proteomic techniques has sparked new searches for novel protein markers for many diseases including CaP. Urinary biomarkers for CaP represent a promising alternative or an addition to traditional biomarkers. Future success in biomarker discovery will rely on collaboration between clinics and laboratories. In addition, research efforts need to be moved from biomarker discovery to validation in a large cohort or separate population of patients and translation of these findings to clinical practice. In this review, we discuss urine as a potential source for CaP biomarker discovery, summarise important genetic urine biomarkers in CaP and focus on MS-based proteomic approaches as well as other recent developments in quantitative techniques for CaP urine biomarker discovery.
... While MALDI platforms are the most frequent MS instruments for resolving the proteomic composition, another similar platform, SELDI (Matrix Assisted Laser Desorption Ionization) was also used in proteomics [32]; a major distinct feature of SELDI is the resolution of integral proteins, and not of peptide fragments. Such studies were applied for serum samples [33,34] or on urine samples [35]. ...
Article
Full-text available
The clinical and fundamental research in prostate cancer - the most common urological cancer in men - is currently entering the proteomic and genomic era. The focus has switched from one single marker (PSA) to panels of biomarkers (including proteins involved in ribosomal function and heat shock proteins). Novel genetic markers (such as Transmembrane protease serine 2 (TMPRSS2)-ERG fusion gene mRNA) or prostate cancer gene 3 (PCA3) had already entered the clinical practice, raising the question whether subsequent protein changes impact the evolution of the disease and the response to treatment. Proteomic technologies such as MALDI-MS, SELDI-MS, i-TRAQ allow a qualitative/quantitative analysis of the proteome variations, in both serum and tumor tissue. A new trend in prostate cancer research is proteomic analysis of prostasomes (prostate-specific exosomes), for the discovery of new biomarkers. This paper provides an update of novel clinical tests used in research and clinical diagnostic, as well as of potential tissue or fluid biomarkers provided by extensive proteomic research data.
... SELDI-TOF MS profiling of post-prostatic massage urine specimens was also applied to differentiate patients with PC from cancer-free subjects. In a study using 57 samples obtained from patients with biopsy-proven PCa and 56 samples from subjects with biopsy-proven BPH, 72 peaks revealed significant differences between groups [100]. This set was reported to discriminate PC from BPH with sensitivity of 91.7% and specificity of 83.3%. ...
Article
Full-text available
Prostate cancer (PCa) is the second most frequently diagnosed malignancy in men worldwide. The introduction of prostate specific antigen (PSA) has greatly increased the number of men diagnosed with PCa but at the same time, as a result of the low specificity, led to overdiagnosis, resulting to unnecessary biopsies and high medical cost treatments. The primary goal in PCa research today is to find a biomarker or biomarker set for clear and effecttive diagnosis of PCa as well as for distinction between aggressive and indolent cancers. Different proteomic technologies such as 2-D PAGE, 2-D DIGE, MALDI MS profiling, shotgun proteomics with label-based (ICAT, iTRAQ) and label-free (SWATH) quantification, MudPIT, CE-MS have been applied to the study of PCa in the past 15 years. Various biological samples, including tumor tissue, serum, plasma, urine, seminal plasma, prostatic secretions and prostatic-derived exosomes were analyzed with the aim of identifying diagnostic and prognostic biomarkers and developing a deeper understanding of the disease at the molecular level. This review is focused on the overall analysis of expression proteomics studies in the PCa field investigating all types of human samples in the search for diagnostics biomarkers. Emphasis is given on proteomics platforms used in biomarker discovery and characterization, explored sources for PCa biomarkers, proposed candidate biomarkers by comparative proteomics studies and the possible future clinical application of those candidate biomarkers in PCa screening and diagnosis. In addition, we review the specificity of the putative markers and existing challenges in the proteomics research of PCa.
... SELDI-TOF MS profiling of post-prostatic massage urine specimens was also applied to differentiate patients with PC from cancer-free subjects. In a study using 57 samples obtained from patients with biopsy-proven PCa and 56 samples from subjects with biopsy-proven BPH, 72 peaks revealed significant differences between groups [100]. This set was reported to discriminate PC from BPH with sensitivity of 91.7% and specificity of 83.3%. ...
Article
Full-text available
Prostate cancer (PCa) is the second most frequently diagnosed malignancy in men worldwide. The introduction of prostate specific antigen (PSA) has greatly increased the number of men diagnosed with PCa but at the same time, as a result of the low specificity, led to overdiagnosis, resulting to unnecessary biopsies and high medical cost treatments. The primary goal in PCa research today is to find a biomarker or biomarker set for clear and effect-tive diagnosis of PCa as well as for distinction between aggressive and indolent cancers. Different proteomic technologies such as 2-D PAGE, 2-D DIGE, MALDI MS profiling, shotgun proteomics with label-based (ICAT, iTRAQ) and label-free (SWATH) quantification, MudPIT, CE-MS have been applied to the study of PCa in the past 15 years. Various biological samples, including tumor tissue, serum, plasma, urine, seminal plasma, prostatic secretions and prostatic-derived exosomes were analyzed with the aim of identifying diagnostic and prognostic biomarkers and developing a deeper understanding of the disease at the molecular level. This review is focused on the overall analysis of expression proteomics studies in the PCa field investigating all types of human samples in the search for diagnostics biomarkers. Emphasis is given on proteomics platforms used in biomarker discovery and characterization, explored sources for PCa biomarkers, proposed candidate biomarkers by comparative proteomics studies and the possible future clinical application of those candidate biomarkers in PCa screening and diagnosis. In addition, we review the specificity of the putative markers and existing challenges in the proteomics research of PCa.
Article
Omics are wide concepts used to define metadata in life sciences and are increasing in number with the development of science. Thus the era of omics started with genomics, which studies the genome; followed by proteomics which focuses on proteins and metabolomics which covers the field of metabolites. Cellomics deals with "the study of cells" or "the knowledge of cellular phenotype and function". One of the main areas of cellomics application is cancer. As there is considerable amount of overlapping between these concepts, we studied papers published between 2005-2013, dealing with cellomics and cancer and found 192 articles, and further classified them in 3 main categories: A: wide genomic and proteomic data analyses; B: cellomics pattern analyses in different kinds of oncologic diseases; C: general cell behavior applied in oncology or studies of atypical cancer cells in different kinds of tumors. 83 papers were clustered in cluster A, 65 in cluster B and 60 in cluster C, 16 papers were included in more than one cluster as they focused on theme of overlapping. Cluster B and C were further sub clustered regarding cancer type and research field. The papers covered a wide range of oncologic diseases: colon-rectal, gastric, breast cancer, ovarian, lung, all types of leukemia, prostate cancer, brain tumors etc; a wide range of cellular mechanisms: cell proliferation, cell migration, cell death, cell adhesion, cell counting, apoptose, phosphorylation, DNA damage, DNA ploidity, DNA sequencing, free circulating DNA etc. Samples used for the study were mainly plasma and tissue but saliva, feces, urine, exhaled breath concentrate etc were also reported for the study. Regarding techniques, papers focused on: flow cytometry, tissue microarray, antibody microarray, reverse phase protein microarray, single nucleotide polymorphism; and also on mass spectrometry: MALDI-TOF-MS, SELDI-TOF-MS, liquid chromatography-tandem mass spectrometry (LC-MS/MS) etc. The article emphasizes the need for standardization. With the emergence of omics fields, there are now no boundary between cellomics, genomics and proteomics. The genes codify processes and proteins as well as cells; as cells are under DNA control and are influenced through RNA protein synthesis, therefore the determination of proteins as the cells as well as the determination of DNA might be referred to as being genomic, proteomic or cellomic.
Article
Full-text available
Context: The percentage of free prostate-specific antigen (PSA) in serum has been shown to enhance the specificity of PSA testing for prostate cancer detection, but earlier studies provided only preliminary cutoffs for clinical use. Objective: To develop risk assessment guidelines and a cutoff value for defining abnormal percentage of free PSA in a population of men to whom the test would be applied. Design: Prospective blinded study using the Tandem PSA and free PSA assays (Hybritech Inc, San Diego, Calif). Setting: Seven nationwide university medical centers. Participants: A total of 773 men (379 with prostate cancer, 394 with benign prostatic disease) 50 to 75 years of age with a palpably benign prostate gland, PSA level of 4.0 to 10.0 ng/mL, and histologically confirmed diagnosis. Main outcome measures: A percentage of free PSA cutoff that maintained 95% sensitivity for prostate cancer detection, and probability of cancer for individual patients. Results: The percentage of free PSA may be used in 2 ways: as a single cut-off (ie, perform a biopsy for all patients at or below a cutoff of 25% free PSA) or as an individual patient risk assessment (ie, base biopsy decisions on each patient's risk of cancer). The 25% free PSA cutoff detected 95% of cancers while avoiding 20% of unnecessary biopsies. The cancers associated with greater than 25% free PSA were more prevalent in older patients, and generally were less threatening in terms of tumor grade and volume. For individual patients, a lower percentage of free PSA was associated with a higher risk of cancer (range, 8%-56%). In the multivariate model used, the percentage of free PSA was an independent predictor of prostate cancer (odds ratio [OR], 3.2; 95% confidence interval [CI], 2.5-4.1; P < .001) and contributed significantly more than age (OR, 1.2; 95% CI, 0.92-1.55) or total PSA level (OR, 1.0; 95% CI, 0.92-1.11) in this cohort of subjects with total PSA values between 4.0 and 10.0 ng/mL. Conclusions: Use of the percentage of free PSA can reduce unnecessary biopsies in patients undergoing evaluation for prostate cancer, with a minimal loss in sensitivity in detecting cancer. A cutoff of 25% or less free PSA is recommended for patients with PSA values between 4.0 and 10.0 ng/mL and a palpably benign gland, regardless of patient age or prostate size. To our knowledge, this study is the largest series to date evaluating the percentage of free PSA in a population representative of patients in whom the test would be used in clinical practice.
Article
Full-text available
We have studied the forms of prostate-specific antigen (PSA) in serum of patients with prostatic cancer and benign prostatic hyperplasia. Fractionation of serum by gel filtration and assay of the fractions for PSA showed that a considerable part of the PSA immunoreactivity in serum consisted of complexes that were larger than PSA. The complexes were assayed by time-resolved immunofluorometric assays based on an antibody against PSA on the solid phase and europium-labeled antibodies against various protease inhibitors as indicator antibodies. In addition to its monomeric form, PSA was found to occur in complex with alpha 1-antichymotrypsin. The proportion of the alpha 1-antichymotrypsin complex was a major form of PSA and it increased with increasing PSA concentrations, being over 85% at PSA levels exceeding 1000 micrograms/liter. A complex with alpha 1-protease inhibitor was also observed in serum of patients with prostatic cancer and very high levels of PSA. Complexes with alpha 2-macroglobulin and inter-alpha-trypsin inhibitor were detected, but their concentrations were low and similar in sera of cancer patients, normal men, and normal women, suggesting that they were not prostate derived. Commercial immunoradiometric assays for PSA were found to measure free PSA and its complexes with alpha 1-antichymotrypsin but not the complexes with alpha 2-macroglobulin and inter-alpha-trypsin inhibitor. The proportion of the PSA-alpha 1-antichymotrypsin complex was higher in patients with prostatic cancer than in those with benign hyperplasia. Therefore, assay of the complex had a higher sensitivity for cancer than assay of total PSA immunoreactivity.
Article
Full-text available
Immunologic measurements of the serum concentration of prostate-specific antigen (PSA), an abundant prostatic-secreted serine proteinase, are frequently used to monitor patients with prostate cancer, though it has not been ascertained whether this immunoreactivity represents a PSA zymogen, the active proteinase, or PSA complexed to extracellular proteinase inhibitors. To characterize the PSA immunoreactivity in serum, we used monoclonal antibodies produced against PSA and a polyclonal rabbit IgG against alpha 1-antichymotrypsin in the design of three noncompetitive PSA assays: assay T, which detected PSA both when present as the active proteinase and when complexed to alpha 1-antichymotrypsin; assay F, which recognized the active proteinase but most poorly detected PSA complexed to alpha 1-antichymotrypsin; and assay C, which was specific for PSA complexed to alpha 1-antichymotrypsin. We used the three assays to measure PSA immunoreactivity in 64 patients' sera and in the effluent after gel chromatography of sera from four patients. This identified an 80- to 90-kDa complex between PSA and alpha 1-antichymotrypsin as the predominant fraction of the PSA immunoreactivity in blood plasma; an immunoreactive 25- to 40-kDa compound was the minor fraction.
Article
Full-text available
The prostate-specific antigen test has been a major factor in increasing awareness and better patient management of prostate cancer (PCA), but its lack of specificity limits its use in diagnosis and makes for poor early detection of PCA. The objective of our studies is to identify better biomarkers for early detection of PCA using protein profiling technologies that can simultaneously resolve and analyze multiple proteins. Evaluating multiple proteins will be essential to establishing signature proteomic patterns that distinguish cancer from noncancer as well as identify all genetic subtypes of the cancer and their biological activity. In this study, we used a protein biochip surface enhanced laser desorption/ionization mass spectrometry approach coupled with an artificial intelligence learning algorithm to differentiate PCA from noncancer cohorts. Surface enhanced laser desorption/ionization mass spectrometry protein profiles of serum from 167 PCA patients, 77 patients with benign prostate hyperplasia, and 82 age-matched unaffected healthy men were used to train and develop a decision tree classification algorithm that used a nine-protein mass pattern that correctly classified 96% of the samples. A blinded test set, separated from the training set by a stratified random sampling before the analysis, was used to determine the sensitivity and specificity of the classification system. A sensitivity of 83%, a specificity of 97%, and a positive predictive value of 96% for the study population and 91% for the general population were obtained when comparing the PCA versus noncancer (benign prostate hyperplasia/healthy men) groups. This high-throughput proteomic classification system will provide a highly accurate and innovative approach for the early detection/diagnosis of PCA.
Article
Proteomics is a rapidly emerging scientific discipline that holds great promise in identifying novel diagnostic and prognostic biomarkers for human cancer. Technologic improvements have made it possible to profile and compare the protein composition within defined populations of cells. Laser capture microdissection is a tool for procuring pure populations of cells from human tissue sections to be used for downstream proteomic analysis. Two-dimensional polyacrylamide gel electrophoresis (2D-PAGE) has been used traditionally to separate complex mixtures of proteins. Improvements in this technology have greatly enhanced resolution and sensitivity providing a more reproducible and comprehensive survey. Image analysis software and robotic instrumentation have been developed to facilitate comparisons of complex protein expression patterns and isolation of differentially expressed proteins spots. Differential in-gel electrophoresis (DIGS) facilitates protein expression by labeling different populations of proteins with fluorescent dyes. Isotope-coded affinity tagging (ICAT) uses mass spectroscopy for protein separation and different isotope tags for distinguishing populations of proteins. Although in the past proteomics has been primarily used for discovery, significant efforts are being made to develop proteomic technologies into clinical tools. Reverse phase protein arrays offer a robust new method of quantitatively assessing expression levels and the activation status of a panel of proteins. Surface-enhanced laser-desorption/ionization time-of-flight (SELDI-TOF) mass spectroscopy rapidly assesses complex protein mixtures in tissue or serum. Combined with artificial intelligence-based pattern recognition algorithms, this emerging technology can generate highly accurate diagnostic in = formation. It is likely that mass spectroscopy-based serum proteomics will evolve into useful clinical tools for the detection and treatment of human cancers.
Article
Pathologic states within the prostate may be reflected by changes in serum proteomic patterns. To test this hypothesis, we analyzed serum proteomic mass spectra with a bioinformatics tool to reveal the most fit pattern that discriminated the training set of sera of men with a histopathologic diagnosis of prostate cancer (serum prostate-specific antigen [PSA] ≥4 ng/mL) from those men without prostate cancer (serum PSA level <1 ng/mL). Mass spectra of blinded sera (N = 266) from a test set derived from men with prostate cancer or men without prostate cancer were matched against the discriminating pattern revealed by the training set. A predicted diagnosis of benign disease or cancer was rendered based on similarity to the discriminating pattern discovered from the training set. The proteomic pattern correctly predicted 36 (95%, 95% confidence interval [CI] = 82% to 99%) of 38 patients with prostate cancer, while 177 (78%, 95% CI = 72% to 83%) of 228 patients were correctly classified as having benign conditions. For men with marginally elevated PSA levels (4–10 ng/mL; n = 137), the specificity was 71%. If validated in future series, serum proteomic pattern diagnostics may be of value in deciding whether to perform a biopsy on a man with an elevated PSA level.
Article
To determine the detection rate of prostate cancer in a screening population of men with prostate-specific antigen (PSA) concentrations of 2.6 to 4.0 ng/mL and a benign prostate examination, to assess the clinicopathological features of the cancers detected, and to assess the usefulness of measuring the ratio of free to total PSA to reduce the number of prostatic biopsies. A community-based study of serial screening for prostate cancer with serum PSA measurements and prostate examinations. University medical center. A total of 914 consecutive screening volunteers aged 50 years or older with serum PSA levels of 2.6 to 4.0 ng/mL who had a benign prostate examination and no prior screening tests suspicious for prostate cancer, 332 (36%) of whom underwent biopsy of the prostate. Cancer detection rate, clinical and pathological features of cancers detected, and specificity for cancer detection using measurements of percentage of free PSA. Cancer was detected in 22% (73/332) of men who underwent biopsy. All cancers detected were clinically localized, and 81% (42/52) that were surgically staged were pathologically organ confined. Ten percent of the cancers were clinically low-volume and low-grade tumors, and 17% of those surgically staged were low-volume and low-grade or moderately low-grade tumors (possibly harmless). Using a percentage of free PSA cutoff of 27% or less as a criterion for performing prostatic biopsy would have detected 90% of cancers, avoided 18% of benign biopsies, and yielded a positive predictive value of 24% in men who underwent biopsy. There is an appreciable rate of detectable prostate cancer in men with serum PSA levels of 2.6 to 4.0 ng/mL. The great majority of cancers detected have the features of medically important tumors. Free serum PSA measurements may reduce the number of additional biopsies required by the lower PSA cutoff.
Article
The segmented quantitative culture technique originally described more than 25 years ago is acknowledged as the best test to diagnose prostatitis. However, it, is not widely used in clinical practice. This is especially true in primary care settings, but even most urologists appear to have abandoned the procedure. Herein is proposed a simple and cost-effective screen for prostatitis, which involves the culture and microscopic examination of urine before and after prostatic massage. This Pre and Post Massage Test (PPMT) was applied to a personal series of 53 patients and 59 patients from the literature in whom the results of the segmented cultures are available and the results were reevaluated. In this selected patient population the PPMT alone led to the same diagnosis in 102 (91.1%). Within the expected limitations of this retrospective review, the calculated sensitivity and specificity of the PPMT were both 91%. This report should provoke researchers to review their prostatitis data, stimulate discussion, and hopefully convince physicians that adoption of a simpler diagnostic plan for prostatitis is far superior to doing no workup at all.
Article
Concurrent with the successful life-saving efforts in terms of prostate cancer diagnosis and treatment, some men who do not need treatment are receiving it. These are men destined to die of causes other than prostate cancer. Unfortunately, at diagnosis, men needing treatment for prostate cancer cannot be differentiated from men who do not. To make such decisions correctly for individual patients would require extremely precise measures of the time to death from prostate cancer versus when the patient would die from a competing cause. Predictive tools with this level of accuracy will never be available given the inherent uncertainty of life. At the time of prostate cancer diagnosis, the date and the cause of death for the patient are matters of weak statistical speculation. Unless the date of death from prostate cancer and the date of death from non-prostate cancer causes can be precisely determined for each patient, some men will always be overtreated or undertreated. Conservative strategies result in the undertreatment of some patients who would benefit from treatment while sparing other patients unneeded treatment. Aggressive strategies result in the overtreatment of patients who do not need therapy while curing other men of prostate cancer. Both strategies are correct, but only some of the time. Better methods of determining the length of life and cause of death may improve this situation, but not by much. [figure: see text] Dramatic shifts in the incidence, grade, stage, and age of men with prostate cancer have been observed with the advent of widespread PSA-based cancer detection in the United States. Grade and stage trends suggest that more biologically relevant (the shift from well-differentiated to moderately differentiated tumors) and yet therapeutically amenable (earlier stage) tumors have been identified in large numbers of patients during the PSA era. Clearly many men have been diagnosed and treated who will not benefit from such treatment. The relative mix of these two groups of men is not known. Given the long delay between treatment and mortality that is inherent in prostate cancer (Fig. 14), the full effects of treatment on prostate cancer mortality are probably not yet seen in prostate cancer mortality data.