Clinical proteomics: Discovery of cancer biomarkers using mass spectrometry and bioinformatics

Vaccine 25S (2007) B110–B121
Review
Clinical proteomics: Discovery of cancer biomarkers
using mass spectrometry and bioinformatics
approaches—A prostate cancer perspective
Balwir Matharoo-Ball, Graham Ball, Robert Rees ∗
Interdisciplinary Biomedical Research Centre, School of Biomedical and Natural Sciences,
Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, UK
Received 9 May 2007; received in revised form 1 June 2007; accepted 15 June 2007
Abstract
Prostate cancer (PCa) is an intractable disease, where diagnosis and clinical prediction of the disease course and response to treatment is
compromised by the lack of objective and robust biomarker assays. In late stage metastatic disease, treatment options are limited, although it
is recognized that some patients may benefit from immunotherapy and in particular vaccine therapy. However, research into biomarkers that
correlate with the clinical outcome of immunotherapy has lagged behind vaccine development. Thus, proteomic tools are increasingly being
utilized for the discovery of biomarkers which will allow us to make clinical decisions about patient treatment at an earlier stage and should
aid in shortening the development time for vaccines. In this review we will summarize the various proteomic platforms used to investigate
new biomarkers in PCa for better patient diagnosis, prognosis, patient stratification, treatment monitoring and clinical surrogate endpoints.
We will discuss method limitations and highlight the key areas of research required for understanding the etiology of PCa.
© 2007 Elsevier Ltd. All rights reserved.
Keywords: Biomarker; Prostate cancer; Proteomics; Mass Spectrometry; Bioinformatics
Contents
1.
2.
3.
4.
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Clinical proteomics: proteomic methods and their application in prostate cancer research . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
2.1. Two-dimensional electrophoresis techniques . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
2.1.1. Technical limitations of two-dimensional electrophoresis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
2.2. MALDI- and SELDI-MS techniques to identify cancer biomarkers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
2.3. Combination of SELDI and pre-clinical models . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
2.4. Current technology validation challenges . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Complex data management and analysis systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Future applications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
B110
B111
B112
B114
B114
B115
B116
B116
B118
B119
1. Introduction
∗
Corresponding author. Tel.: +44 115 848 6342; fax: +44 115 848 6632.
E-mail address: [email protected] (R. Rees).
0264-410X/$ – see front matter © 2007 Elsevier Ltd. All rights reserved.
doi:10.1016/j.vaccine.2007.06.040
In Europe, prostate cancer is the most common noncutaneous cancer in men after lung cancer. There are
approximately 134,000 new cases every year [1] and approx-
B. Matharoo-Ball et al. / Vaccine 25S (2007) B110–B121
imately 56,000 related deaths. The incidence is increasing
rapidly, as the population of males over the age of 50 grows
and through the increased use of prostate specific antigen
(PSA) assessment. Both hereditary and environmental factors influence the pathogenesis of the disease and there is
evidence that inflammation plays a critical role in its aetiology [2]. It is, however, unclear which cells of the immune
system are implicated in the pre-malignant inflammatory
conditions; prostatic intraepithelial neoplasia (PIN) which
is thought to be the precursor lesion of prostate cancer and
benign prostatic hyperplasia (BPH). Adenocarcinoma of the
prostate is the predominant form of prostate cancer and
most localized prostate cancers can be effectively treated by
surgery or radiation therapy. However, every year, 70,000
men [1] require additional treatment due to recurrence and
sometimes dissemination of the cancer to other parts of the
body (metastasis). After the lymph nodes, the skeleton is the
most common site of metastasis, the majority of which are
osteoblastic, or bone forming. Bone metastases are associated with poor prognosis and are the major cause of prostate
cancer-related morbidity (sickness) and mortality (death).
Androgen ablation therapy is a palliative treatment (reduces
pain and tumour burden) and not curative for patients with
metastases [3]; however, eventually the disease will become
androgen independent (hormone refractory) and re-occur,
at which point other treatment options are limited. For this
reason, there is an urgent requirement to develop new treatment options for prostate cancer, where the identification
of new prostate cancer-associated gene/proteins will aid in
the development of a more comprehensive understanding of
the disease and additional “targeted” therapeutic approaches.
PSA is a tumour marker routinely measured in the blood of
prostate cancer patients. Rising levels of PSA in the blood
of patients receiving androgen ablation therapy provide the
earliest indication of tumour progression to androgen independence. Once the disease is androgen independent, the
average survival time is 2 years [3].
PSA, which was discovered almost 25 years ago, is not
prostate cancer specific and PSA is found in the normal
prostate at equal or higher levels than is found in prostate
cancer. BPH, prostatitis, as well as prostate cancer, can result
in higher PSA levels in the serum as a result of cellular
re-organization allowing leakage into the surrounding blood
vessels. Furthermore, in prostate cancer the blood vessels are
more permeable, making it easier for PSA to enter the bloodstream. Despite these limitations, PSA screening has changed
the course of the disease, although 17% of men with “normal”
blood PSA levels have cancer cells within the prostate [4];
78%, of men that have elevated levels of PSA (in the range
of 4–9.9 ng/ml) do not have prostate cancer [5]. Due to the
lack of accurate biomarkers that detect, monitor, and quantify
significant PCa or distinguish it from benign disease, many
early-stage PCa patients are treated as harbouring significant
PCa, involving physical over treatment with associated emotional morbidity [6]. It is therefore difficult to reliably detect
early stage prostate cancer without histological examination.
B111
Evaluating the Gleason score from a tissue specimen taken
at the time of biopsy is still the most widely used diagnostic and prognostic tool for human prostate cancer [7] and
despite assay improvements, PSA testing does not provide
a prediction of disease outcome in newly diagnosed patients
and cannot, by itself, determine the course of treatment and
increasingly active surveillance is used as an alternative. It
is recognized that some patients with late stage may benefit from immunotherapy and in particular vaccine therapy.
We propose that no single molecular approach will fully
elucidate tumour behaviour, necessitating analysis at multiple levels encompassing integrated data sets to determine
the contributions of genomic alterations, host factors and
environmental factors influencing tumour growth and progression; in addition there is currently no curative treatment
for PCa metastatic disease [8]. For this reason, alternative
therapies are also urgently needed and a growing interest is
focusing on immunotherapeutic approaches. To this extent,
no immunological or other markers currently exist that can
be used as validated surrogate clinical endpoints in trials
for immunotherapy. Potential endpoints include immunological biomarkers (measurement of T-cell and cytokine
responses), autoimmunity as a correlate for treatment outcome, and the possible identification of multi-parametric
(multiple) biomarkers using high-throughput proteomic technologies. High-throughput proteomics technologies should
help in the identification of clinically relevant prostate cancer biomarkers at an earlier stage and should help in the future
development of effective vaccines.
2. Clinical proteomics: proteomic methods and their
application in prostate cancer research
Clinical proteomics can have important applications that
may directly change clinical practice by affecting critical elements of care and management. The ‘proteome’ refers to the
entire repertoire of cellular proteins and because proteins are
involved in almost all biological activities, the proteome is
a rich source of biological information. PCa treatment decisions are complicated by the biologic heterogeneity of the
disease and potential proteomic biomarkers of prostate cancer can benefit not only the early detection of disease, but
can also be used for determining cancer risk, stratifying disease stage and grade, monitoring response to therapy, and
in general assisting in therapeutic decision making. Through
careful sample selection, study design and automation in sample handling and processing, proteomic platforms are fast
becoming powerful tools for deriving protein “signatures”
in a wide range of cancers, including prostate, gastrointestinal, hepatocellular, breast, ovarian, and brain, cancers, where
serum protein patterns can be used to differentiate cancer
from controls [9–14].
Cancer proteomics encompasses the identification and
quantitative analysis of the entire complement of proteins in
a biological sample. The commonly used technologies avail-
B112
B. Matharoo-Ball et al. / Vaccine 25S (2007) B110–B121
able today include 2D-polyacrylamide gel electrophoresis
(2DE), isotope-coded affinity tags, matrix assisted laser desorption ionization-mass spectrometry (MALDI-MS), liquid
chromatography–MS/MS (LC–MS/MS), imaging MS, protein arrays, and autoantibody expression techniques. Chip
based protein and peptide arrays have been used to study
protein–protein, protein–DNA, and protein–RNA interactions [15,16] whilst autoantibody expression studies deal with
tumour-associated antigens shown to be overexpressed in
neoplastic cells and recognized as foreign by the immune
system [17,18]. This method has been successfully used to
identify and track cancer progression and to identify new
“autoantibody signatures” in PCa.
Proteomic profiles on the other hand, consider all serologic proteins (housekeeping protein, enzymes, antibodies,
etc.), identifying multiparametric biomarkers. Protein profiling, assisted by computational statistical analyses, could
yield a quick and reliable diagnosis of prostate cancer
[9,19–21]. We have conducted research over a number of
years on HLA-peptide expression using ESI-MS/MS (electrospray ionization tandem mass spectrometry) [22,23] as
well as protein expression studies using SEDLI-ToF-MS (surface enhanced desorption laser ionization-time of flight MS)
[24–27] and more recently MALDI-MS in conjunction with
ANNs (artificial neural networks) analysis [28].
2.1. Two-dimensional electrophoresis techniques
The human genome is estimated to contain about 35,000
protein-encoding genes and the goal of clinical proteomics
is to develop technologies for the interrogation of the entire
proteome in a search for proteins that can be used as early
biomarkers of disease, or that may predict response to therapy, or the likelihood of relapse after treatment in blood,
urine, or diseased tissue [29,30]. A number of studies have
attempted to identify and analyze proteins in prostate cancer
from less complex mixtures such as seminal fluids [31,32],
urine [33], laser captured cells from cancer tissues [34–36],
albumin-associated proteins from blood sera [36], glycosylated proteins [37–39] or sub cellular fractions from cancer
cells [40]. However, in order to gain a comprehensive understanding of protein networks, a combinatorial approach is
required.
Protein content has been traditionally measured using
low-throughput techniques such as western blotting, and
immunohistochemical staining [41]. 2DE is by far the most
widely used tool in proteomics approaches for more than 25
years [42]. In 2DE, proteins are separated firstly on the basis
of differences in net charge, through a technique known as
isoelectric focusing (IEF) and secondly on differences in their
molecular mass through polyacrylamide gel electrophoresis.
By use of different staining techniques such as silver staining
[43], Coomasie blue stain, florescent dyes [44], or radiolabels, a few thousand proteins can be visualized in a single
gel. In most proteomic-based work, 2DE gel-matching procedure is used to compare two sets of protein mixtures run
under highly standardized conditions. A minimum of two parallel gels have to be run, to obtain a sound basis for applying
the image technology and to directly compare preparations of
protein mixtures from “disease” and “healthy” states. Ratio
analysis is used to detect quantitative changes in proteins
between two samples. The incorporation of fluorescent dyes
in a process termed 2DE differential in-gel electrophoresis
(DIGE) [45] allows the proteins from different populations
to be labeled and analysed on the same gel. This technique
has made comparative expression analysis between samples
much easier. The major advantage of 2DE is that it enables
the simultaneous separation and visualization of thousands of
unknown proteins. Coupling 2DE, followed by spot protein
digestion using a protease such as trypsin, before mass spectrometric analysis has become a method of choice in recent
years (Fig. 1). This approach allows the identity of the protein through the use of peptide mass fingerprinting (PMF)
and tandem mass spectrometry (Fig.1) to be determined.
PMF is a protein identification technique in which the
mass spectrometer is used to measure the mass of proteolytic peptide fragments. The protein is identified by
matching the experimental peptide mass to the corresponding
PMFs found in the protein or nucleotide sequence databases.
Commonly used databases include SWISSPROT, OWL and
NCBInr databases. However, there remain several limitations
to PMF, including a lack of compete and accurately annotated
genome- and protein-sequence databases for a great number of highly homologous human proteins. Although 2DE
coupled with mass spectrometry has been used to identify
protein changes associated with a variety of human cancers,
until recently, it has had limited applications regarding the
study of early stage prostate cancer. The primary reason for
this limitation is the heterogeneous and infiltrative quality
of prostate cancer that makes it difficult to isolate a pure
population of malignant prostatic epithelium. To overcome
this investigative hurdle, different microdissection techniques
have been developed for procuring pure populations of cells
from human tissue sections.
Laser capture microdissection (LCM) has been used as
a potential tool in proteomic research [46–48]. LCM is a
tool used for the enrichment of homogeneous cells from tissue sections overcoming the problem of tissue heterogeneity.
LCM allows for selection of cells with a precision of 3–5 ␮m
under microscopic visualization. Briefly, a stained slide is
placed under a microscope, and a specific cap with a film
of ethylene vinyl acetate (EVA) film, is placed over the tissue. When the cells of interest are located an infrared laser is
fired, which melts the film in the area of the target. Specific
to prostate cancer studies, LCM has been used to procure
pure populations of patient-matched benign and malignant
prostatic epithelium. Protein expression has been compared
using 2DE and differentially expressed proteins identified by
mass spectrometry [49].
Combinations of the methods described above were used
recently by Lexander et al. [50], who analysed differences
in protein expression in prostate cancer of high and low
B. Matharoo-Ball et al. / Vaccine 25S (2007) B110–B121
B113
Fig. 1. 2DE-gel based approach to clinical proteomics. There are obvious scientific advantages for determining protein complexes and post-translational
modifications when employing this approach. Intact proteins of interest trypsinised which are then subjected to subsequent MS-based analyses using either
single or tandem mass spectrometry. Throughput is usually indirectly proportional to the amount of information gathered. The peptide mass fingerprint (PMF)
generated can then be searched against databases which will provide protein/peptide identity.
aggressiveness, according Gleason score and DNA ploidy
factors that correlate with prognosis. Cell suspensions from
35 prostatectomy specimens, 29 cancer samples and 10
benign samples were prepared by scraping cells from cut
surfaces which were subsequently examined by 2DE. Protein spots that differed quantitatively between sample groups
were identified by MS fingerprinting of tryptic fragments
and MS/MS sequence analysis. They found 39 protein spots
with expression levels that were raised or lowered in correlation with Gleason score and/or DNA ploidy pattern (31
overexpressed in high-malignant cancer, 8 underexpressed).
Of these, 30 were identified by MS. Among overexpressed
proteins were heat-shock, structural, membrane proteins
and enzymes involved in gene silencing, protein synthesis/degradation, mitochondrial protein import (metaxin 2),
detoxification (GST-pi) and energy metabolism. Stromaassociated proteins were generally underexpressed.
In another study, the proteome patterns correlating with
the three anatomical zones of the prostate: the peripheral
(PZ), the transition (TZ) and the central (CZ) zone were investigated [51]. It is proposed that the CZ may be of mesodermal
origin, whereas the other two are of endodermal origin. Cells
were scraped from macroscopically normal areas of PZ, TZ,
and CZ in radical prostatectomy specimens, prepared and
analysed using 2DE. Ten proteins with significant zonal differential expression were identified, via mass spectrometric
fingerprinting of tryptic fragments and selected tandem mass
spectrometry sequence analysis, eight with underexpression
in the CZ versus the PZ and the TZ (arginase II, ATP synthase, cytokeratin 8, lamin A/C, peroxiredoxin 4, protein
disulfide isomerase A3, tropomyosin, and vimentin), and two
with overexpression in the CZ (peroxiredoxin 2 and creatine
kinase B). The PZ and TZ, have epithelium with highly similar major protein expression profiles, whereas the protein
profile of the CZ differed suggesting functional differences.
Despite the effectiveness of hormone therapy in prostate
cancer, all patients with metastatic disease eventually
progress to an androgen-independent state. Once patients
develop androgen-independent cancer, no effective cure
currently exists. Proteins responsive to androgen and
anti-androgen may be involved in the development and
progression of prostate cancer and the ultimate failure of
androgen-ablation therapy. These proteins represent potential
diagnostic and therapeutic targets for improved management
of prostate cancer. Several studies using prostate cancer cell
lines have investigated different aspects of androgen proteins
using proteomic platforms.
Proteins differently expressed in androgen-sensitive
prostate cancer cell-line LNCaP-FGC and androgen-resistant
line LNCaP-r (a model for development of androgen resistance in prostate cancer) were identified by 2DE and mass
spectrometry [52]. HSP60 was upregulated in LNCaPr, nm23 in LNCaP-FGC, and titin (two isoforms) in
either LNCaP-r or LNCaP-FGC. In non-malignant prostate,
HSP60-staining was in the glandular compartment, particularly basal epithelial cells, whilst in prostate cancer, most
B114
B. Matharoo-Ball et al. / Vaccine 25S (2007) B110–B121
epithelial cells showed moderate–strong staining without
apparent correlation between staining intensity and Gleason grade. This study highlighted that the identification of
HSP60 correlated with clinical results, indicating that this
model can be used for dissection of mechanisms involved
in transformation to androgen resistance and assignment of
protein markers in prostate cancer. Rowland et al. [53], on the
other hand, investigated the effect of androgen (R1881) and
anti-androgen (bicalutamide) on the androgen-responsive
prostate cancer LNCaP cell line using a quantitative gel-based
proteomic approach analysed by 2DE DIGE. Following
androgen supplementation, 108 spots (68 proteins) were
increased and 57 spots (39 proteins) were decreased. Essentially, no difference was observed between control and
anti-androgen-treated samples, confirming the absence of
“off-target” effects of bicalutamide. Identified proteins were
shown to be involved in diverse processes, including the stress
response and intracellular signalling. This study of androgen responses has provided a number of potential candidates
for development as diagnostic/prognostic markers and drug
targets.
Very few studies recently have been carried out using
serum and 2DE approaches and this is primarily due to
the technical challenge of the serum proteome which has
a few proteins are that are so dominant, such as albumin
and immunoglobulins, that they mask detection of other
proteins. Traditionally, serum-based biomarker studies have
used strategies to deplete albumin and immunoglobulins to
increase the sensitivity for the lower abundance proteins
that may represent biomarkers and drug targets. Qin et al.
[54] used this approach, by fractionating serum samples
from patients with prostate cancer and patients with benign
prostate hyperplasia, using anion displacement liquid chromatofocusing chromatography, which separates proteins by
a pH gradient and a positively charged column. Thereafter,
they determined and identified the serum proteins by IEF
gels and 2D DIGE. Several proteins were shown to be differentially expressed in serum from patients with prostate
cancer, which were identified as squamous cell carcinoma
antigen 1 (SCCA1), calgranulin B, and haptoglobin-related
protein. Follow-up studies to determine the specificities of
these findings are still needed.
2.1.1. Technical limitations of two-dimensional
electrophoresis
Clearly, 2DE has limitations when used in proteomics:
one of the weakest points of 2DE is the difficulty in automating the process and its analysis is limited to low-throughput
means, preventing it from being applied to large numbers of
samples in a limited time frame. In addition, 2DE has much
inter-gel variation. 2DE analysis also only resolves the major
components of a protein mixture and the detection of the low
and high molecular mass, of basic and hydrophobic proteins
is inefficient [55,56]. New technologies are now available that
can overcome these limitations and can be adapted for highthroughput processing. The development of techniques such
as multi-dimensional liquid chromatography, combined with
mass spectrometry, to profile peptides from protein digests
allow hundreds of protein to be identified in lysates [57].
MALDI mass spectrometry has also been used for generating patterns of proteins from clinical samples, such as serum
and plasma, and does not rely on protein identity so can be
used to generate a diagnostic fingerprint. MALDI has also
been used for molecular imaging of tissue sections [58] for
generating disease-related protein mass profiles of normal
and malignant breast cells [59].
2.2. MALDI- and SELDI-MS techniques to identify
cancer biomarkers
In the last 10 years or so, MS has increasingly become
the method of choice for analyses of complex protein samples. Two ionization techniques, MALDI and ESI, have had
a major impact on protein biochemistry because they are able
to produce ions in the gas phase without fragmenting the proteins, a problem with older methods. MALDI produces ions
by sublimating (transforming a solid to a gas) and ionizing
the proteins out of a dry, crystalline stage. MALDI mass spectrometry as a high throughput method involves the analysis of
a protein/peptide molecule which requires only a few microliter of sample; the three process steps are: (i) ionization of the
protein and generation of the gas-phase ions; (ii) separation
of ions according to their mass to charge ratio which is based
upon the time it takes for the launched ion to reach the electrode; small ions travel faster and vice versa; (iii) detection of
the ions. In this way a protein/peptide map is generated with
the sensitivity to detect molecules at a few parts per million
comprising tens-of-thousands of protein ions and requires
pattern-recognition type algorithm for analysis. A variant of
MALDI, where the surface of the MALDI target has been
modified, known as SELDI-MS is widely used in cancer proteomics. Protein fragments in the sample bind to the SELDI
surface because they have an affinity for the substances on
the surface [21]. SELDI is an affinity based MS method in
which proteins are selectively adsorbed to a chemically modified surface, and impurities are removed by washing with
buffer. The end product of analysis is a mass spectrum or
chart with a series of spiked peaks, each representing the
ion or charged protein fragment present in a given sample,
where the height of the peak is related to the abundance of
the protein fragment. The size of the peaks and the distance
between them are a fingerprint of the sample and provide a
clue to its identity [21]. Computational artificial intelligence
type systems that learn, adapt and gain experience over time
are uniquely suited for the analysis of mass spectral proteomic data. SELDI-ToF-MS has successfully been used to
detect several disease-associated proteins in complex biological samples, such as tissue/cell lysates, seminal plasma and
serum [31,32,34–36].
LCM from snap frozen prostate cancer serial sections, to
ensure a homogeneous population of cells, in combination
with SELDI-MS has been used in recent studies of prostate
B. Matharoo-Ball et al. / Vaccine 25S (2007) B110–B121
cancer cells for the identification of proteomic alterations
associated with the early stages in the development of prostate
cancer [60,61]. These studies identified a 24-kDa peak which
was observed at low or high intensity in profiles of prostate
cancer cells in 19 of 27 lesions and at low intensity in 3
of 8 hPIN lesions but was not detectable in matched normal cells. SDS-PAGE analysis followed by MS identified
the protein as the dimeric form of mature growth differentiation factor 15 (GDF15). However, the increased expression
of mature GDF15 protein in prostate cancer cells could not
be explained on the basis of up-regulation of GDF15 mRNA
because reverse transcription-PCR analysis showed similar
amounts of transcript in normal, hPIN, and prostate cancer
cells in the same set of serial sections from which the protein
profiles were obtained. Zheng et al. [60] on the other hand
identified a peak at m/z 24,782.56 ± 107.27 that was correlated with the presence of prostate carcinoma. Furthermore,
using LCM, the origin of this protein, which the authors designated PCa-24, was derived from the epithelial cells of the
prostate. PCa-24 expression was detected in 16 of 17 (94%)
prostate carcinoma specimens but not in paired normal cells.
In addition, this protein was not expressed in any of the 12
benign prostatic hyperplasia specimens that were assayed.
The biological significance of all these observations awaits
future investigations.
A different strategy was used by Liu et al. [62] who used
collagenase digestion to achieve single cells from matched
cancer and non-cancer specimens, which were subsequently
digested and pelleted to give a cell-free supernatant. A
reversed phase hydrophobic ProteinChip Array was used to
generate SELDI patterns from 43 primary prostate tumours,
including 26 with matched non-cancer specimens. Quantitative proteomics was applied to one tissue sample which
identified metalloproteinase inhibitor-1 as the biomarker
down-regulated in cancer and was shown to be localized to
secretory cells.
From a diagnostic standpoint differences in the composition of secreted protein species are especially relevant. It has
been hypothesized that the circulation could contain a pattern
of expressed proteins that form a disease-specific signature
which can be identified using high-throughput MS proteomic
analysis with high specificity and sensitivity [13]. The discriminatory power of SELDI-ToF has sparked tremendous
excitement after the most notable serum proteomic report
using this technology identified 100% of all ovarian cancer
samples and correctly assigned 95% of healthy and benign
subject correctly [13]. SELDI serum studies in PCa have been
used as diagnostic tools for identifying discriminate serum
peaks capable of distinguishing between normal, BPH and
PCa patients with sensitivities from 63 to 100% and specificities from 38 to 100% [12–14,20,63,64]. An issue plaguing
the SELDI analytical platform has been the identification of
the discriminatory biomarkers. However, a study by Adam
et al. [20] which had reported a discriminatory peak at 8946
m/z was recently followed up with a study using a prospective population [65] that identified the same peak, but more
B115
importantly was found to retain discriminatory value with an
increased expression in the diseased state. Sequence identification was carried out by liquid chromatography–MS/MS
and subsequent immunoassays verified that an isoform of
apolipoprotein A-II (ApoA-II) was the observed 8946 m/z
SELDI peak. Immunohistochemistry revealed that ApoAII was overexpressed in prostate tumours. SELDI-based
immunoassay also revealed that an 8.9-kDa isoform of ApoAII is specifically overexpressed in serum from individuals
with prostate cancer. ApoA-II was also overexpressed in the
serum of individuals with prostate cancer who have normal prostate-specific antigen (0–4.0 ng/ml). Interestingly; no
other cited studies have reported the same discriminatory
peaks even though the same chromatographic affinity surface
has been used by some groups [14,20]. While these different
groups found different serum proteomic patterns, importantly
all of these patterns showed statistically significant discriminatory power for prostate cancer detection. Clearly, these
studies highlight some of the underlying reproducible and
validation problems associated with SELDI-ToF proteomic
platform.
2.3. Combination of SELDI and pre-clinical models
The application of SELDI-ToF-MS technology is not
only limited to the study of serum for early PCa diagnosis; it has also been used in obtaining proteomic patterns of
the secreted proteins in conditioned medium from LNCAP
cells following stimulation with methyltrienolone (R1881)
an AR-specific agonist, 17beta-estradiol and interleukin-6
[66]. This study reported many peaks which were induced
or repressed following stimulation with R1881 or estradiol.
A peak at 11.8 kDa identified as beta-2-microglobulin (B2M)
was increased upon R1881 stimulation. The investigators further reported an increase of B2M in the serum of mice bearing
human PCa xenograft concluding that this protein is elevated in patients with metastatic and androgen-independent
PCa. This study highlights the potential of preclinical models in biomarker discovery. The human PCa xenograft PC339
was recently used in an immune-incompetent nude mouse
model to identify proteins specifically produced by diseased
cells, rather than as the investigators hypothesized proteins
produced due to secondary body defense mechanisms [67].
The concept of this study was that mouse serum would
contain human proteins which originate from the human
PCa xenograft hence allowing potential biomarkers to be
identified. Using one-dimensional gel electrophoresis, liquid chromatography, and mass spectrometry, tumour-derived
human nm23/nucleoside-diphosphate kinase (NME) in conjunction with six human enzymes involved in glycolysis
(fructose-bisphosphate aldolase A, triose-phosphate isomerase, glyceraldehyde-3-phosphate dehydrogenase, alpha
enolase, and lactate dehydrogenases A and B) were identified in the serum of the tumour-bearing mice. The presence
of human NME and glyceraldehyde-3-phosphate dehydrogenase in the serum of PC339-bearing mice was confirmed
B116
B. Matharoo-Ball et al. / Vaccine 25S (2007) B110–B121
by Western blotting. Although the putative usefulness of
these proteins in predicting prognosis of prostate cancer
remains to be determined, this study illustrates that human
PCa xenograft approach may be a promising tool for the
focused discovery of new prostate cancer biomarkers.
2.4. Current technology validation challenges
By offering a platform that can quickly acquire proteomic
patterns from complex biological samples, SELDI-ToF-MS
has advantages over other conventional analytical techniques.
Like any emerging technology, however, SELDI-ToF-MS
also faces some challenges that can potentially limit its application. One of these challenges is the reproducibility among
proteomic profiling experiments [68]. The chip-to-chip coefficients of variation of peak intensities can vary from 10 to
40% [69]. In 2005 Semmes et al. [70] reported the only systematic study to assess inter-laboratory reproducibility and
validity across populations of prostate cancer patients. They
reported the reproducible detection of quality-control peaks
and the correct classification of nearly all of the same 14
cancer and 14 control samples at six different sites, based
on discriminating peaks identified in these samples 2 years
earlier [71]. As a result, investigators routinely incorporate a
number of approaches to ensure reproducible results. These
include adding a quality control sample on each chip array,
and normalizing spectral data through commercially available or in-house generated computer programs. Undoubtedly,
the appropriate control and assessment of reproducibility
would be required for any future application of SELDI-ToFMS in disease diagnosis. This encouraging study illustrates
that the proteomic platform does indeed have the potential
capability to be used in widespread cancer diagnosis and
prognosis.
The obvious challenge, as stated by many investigators, is
the identification of the important proteins and peptides that
contribute to the proteomic patterns derived from the SELDIbased studies. The low mass resolution and mass accuracy are
inherent to the design of the SELDI system. Some investigators have attempted direct on-chip identification using SELDI
interface on the QqToF instrument, yet achieved little success.
So far the successful identification of proteins and peptides in
the proteomic patterns still relies on other protein separation
and enrichment methodologies.
Every technology has its limitations. The challenges
that SELDI faces would not diminish the demand for
proteomic research in the post genomics era. Our laboratory and others have been employing a combination
of automated robotic chromatographic ZipTip format and
MALDI-ToF-MS to present a powerful and sensitive analysis of pre-fractionated samples [28]. A whole-protein based
top-down separation strategy (Fig. 2) for the identification
of a stage-specific marker in a group comprising 16 patients
with PCa (metastatic and localized disease) and 15 healthy
individuals was used by Lam et al. [72]. MS-profiling, combined with multivariate analysis, yielded 17 serum proteins
specific to metastatic disease. A single protein detected at
m/z 7771 was found to be significantly decreased in the
sera of all the metastatic PCa patients which was isolated
using a C18 prefractionation step, followed by multidimensional liquid chromatography and, finally, two-dimensional
gel electrophoresis. The separation process was monitored by
UV–vis and MALDI-ToF-MS analysis. This strategy identified the unknown protein as platelet factor 4, a chemokine
with prothrombolytic and antiangiogenic activities. Confirmation was achieved using both Western blot analysis
and enzyme-linked immunosorbent assay. This study highlights some of the drawbacks of whole protein based study
design in that additional separation techniques need to be
employed prior to identification. The novelty of the approach
we have adopted is that we can deconvolute the samples
at the protein level for screening biomarkers, followed by
tryptic digestion (bottom-up, Fig. 2) of the same eluted sample which allows us to obtain ANNs directed signatures
and more importantly protein identification with little or
no additional work-up using MALDI-ToF and ESI-MS/MS.
We have used this strategy in our serum studies to investigate new proteome biomarkers for late stage IV melanoma
versus healthy controls [28]. We have developed a reproducible and standardized integrated sample preparation and
mass spectrometry-based proteomic protocol, combined with
ANNs modelling, for protein and tryptic peptide biomarker
discovery and identification in human samples. Another
group have also recently reported [73] the development of
a streamlined, microscale sample preparation protocol that
used either chromatography-based spin columns or 96-well
filtration plates. The approach is based on affinity capture of
albumin (and other carrier proteins) by a novel dye affinity membrane absorber matrix or beads, with subsequent
elution and concentration of the carrier-protein-bound peptides for direct mass spectrometric analysis. Coupled with
the biomarker enrichment protocol, the authors reported that
the high-resolution MALDI O-ToF mass spectra were reproducible for peptide signatures. The raw mass spectra were
then analysed and used to build discriminant disease models
that were challenged with blinded samples for classification
in ovarian cancer patients.
3. Complex data management and analysis systems
The final challenge MALDI/SELDI faces lies largely
in the application of bioinformatics, i.e. the spectral data
management and analysis. The vast amount of spectral
data generated by MALDI/SELDI technology demands
implementation of advanced data management and analysis strategies. The modelling and analysis of data derived
from clinical studies of PCa based on MALDI/SELDI, poses
a significant challenge due to complexity, redundancy and
high dimensionality within these data types [24,74]. Some of
these problems have been overcome by the use of robust computational algorithms such as ANNs, neuro-fuzzy systems
B. Matharoo-Ball et al. / Vaccine 25S (2007) B110–B121
B117
Fig. 2. Top-down and bottom-up proteomics approaches. The top-down approach illustrates a non-gel based approach in which whole proteins are fractionated
via various methods. The fractionated proteins are then subjected to linear MALDI-MS to generate protein mass spectra which can then be analysed by various
bioinformatic statistical tools. The bottom-up approach utilizes primarily non-gel based fractionation of peptides generated from complex protein mixtures.
The rationale is that the digested peptides will reflect the native proteins and that the peptides behave more uniformly in both fractionation and detection.
Digest-generated peptides are then subjected to mass spectrometry analysis. The approach can involve simple peptide mass profiling as well as quantitative
tandem mass spectrometry to yield protein identification and relative protein concentration.
[75,76], support vector machines [77] independent components analysis [78], Bayesian methodologies [79] or decision
trees [80]. Artificial neural networks (ANNs) are a form of
machine learning capable of accurately modelling biological
systems and identifying biomarkers. They have been applied
to a number of diverse areas for the identification of “biologically relevant” molecules, including mass spectrometry
[24,81,82] and genomic micro-array analysis of tumour tissue [83]. It is well known that ANNs are a powerful tool
for the analysis of complex data [84–86] can cope with data
containing a high level of background noise and can be used
to identify the influence of many interacting factors [87]; all
positives which that makes it highly suitable for the study of
mass spectrometry derived data. A number of studies have
indicated the approach can produce generalized models with
a greater accuracy than conventional statistical techniques
in medical diagnostics [88,89] without relying on predetermined relationships as in other computational modelling
techniques. One commonly used algorithm is the backpropagation algorithm applied to the multi-layer perceptron (MLP)
architecture (Fig. 3) [90,91]. The MLP consists of an input
layer, a hidden layer or layers and an output layer. The input
layer represents the independent variables; the hidden layer
mathematically represents features within the data and the
output layer represents the dependant variables or classes.
Each layer comprises a number of nodes interconnected by
weighted links. The weighted links feed values from the previous layers where they are summed and the resulting value
applied to a transfer function (commonly a sigmoid). Thus
the neural network mathematically modifies input values to
produce predicted output values. The algorithm is applied to
the architecture in order to update the weights in the architecture such that the ANN model is able to make accurate
predictions.
Fig. 3. Structure of a multilayer perceptron showing a 3 layer structure,
interconnected nodes and a sigmoidal transfer function. The input layer represents the independent variables – each node representing a parameter. The
hidden layer represents a mathematical feature detection layer – each node
represents a feature. The output layer represents the dependant variables –
each node represents a variable or class.
B118
B. Matharoo-Ball et al. / Vaccine 25S (2007) B110–B121
In this field we have made significant advances developing
and applying ANN based approaches based on MLP ANNs
trained with back propagation algorithms, for the analysis
of highly dimensional biomedical data. While others have
applied ANNs to the analysis of complex biomedical data,
the level of cross validation is frequently less robust [75,92]
and these models are likely to suffer from overtraining [93].
We have developed and applied extensive random sample
cross-validation approaches that exceed the standards outlined by Michiels et al. [94]. This procedure randomly splits
the data into training (60%), selection (20%) and test (20%)
data sets. The training set is used to stop training, the selection set is used to prevent overtraining and the test set is used
to assess the generalization of the model, i.e. its performance
on blind data. This process is repeated for 50 different random data splits for the same global data set. These validation
approaches are incorporated within our model development
process and have been shown to produce excellent predictive performance with good sensitivity and specificity for
secondary blind data sets [82] and allow the calculation of
confidence intervals for predictions. Within this cross validation framework, we have incorporated a piece-wise additive
approach to model development and parameterization which
is able to identify the most representative subset of biomarkers associated with the classes being modelled in the system
[28]. In this way using this stepwise approach to model
development we define an optimum set of mass spectrometry
derived markers that may be used to define a clinical question.
Other earlier studies have frequently used sensitivity analysis
[e.g. 82,95,96] to identify features within the trained ANN
model. These however suffer from issues associated with
the analysis of high dimensionality data. We have demonstrated that we can successfully overcome issues associated
with complexity, non-linearity and the high dimensionality
of the data, while providing a clear indication of the relationship between biomarker parameters and clinical class. ANNs
have been criticized by some citing that they have limited
transparency and no insight can be gained into the way they
make predictions [93]. The methods we have developed have
made significant advances in gaining information on biological systems by interrogation of ANN models overcoming this
criticism.
To address issues around the biological variability of samples within given class populations we have developed an
approach for the investigation of structures within the population, based on the median predictions across a number of
models for individuals, ranked by value. By examination of
these values and their resulting distribution we may assign
a probability to an individual falling within a given class
(e.g. cancer or non-cancer) and indicate the likelihood of
that individual belonging to that class. Ranking the predictions of the models using this approach allows us to define
a fuzzy boundary between classes [28,97]. We have successfully applied these approaches (as a part of EU Framework 6
ENACT and other programmes) to the analysis of immunological data [98] for the modelling and prediction of clinical
classes. In this instance immunological parameters have been
used to characterize responders and non-responders following whole tumour cell vaccine therapy. We were able through
the creation of an ANN based predictive model to identify and characterize a subset of predictive cellular immune
markers that were associated with response in hormonerefractory prostate cancer patients; these included expression
of cytokines, such as IFN-␥, TNF-␣ and IL-2, from patient
peripheral blood lymphocytes following in vitro stimulation with calcium ionophore, plus T cell proliferation in
response to vaccine antigen [98]. Through understanding
the mechanisms underlying the response to immunotherapy, the potential exists to predict whether an individual
will respond to vaccination prior to treatment or to monitor
patient’s immune response on therapy and adjust the treatment accordingly to ensure appropriate therapy. In all of the
cases where our ANN approaches have been applied the features identified as predictive have been biologically relevant
and subsequently been validated on new cases [28,82]; the
immunological markers identified in [98] shall be validated
using samples from an ongoing Phase IIb clinical trial in the
same patient population.
The approaches described above have had some successful application in the field of prostate cancer [87,99]. Some
studies have utilized ANN approaches to increase the sensitivity and specificity of PSA tests by inclusion of factors such
as percentage free PSA, age and digital rectal examination
results [100,101]. Others have adopted a similar approach
using PSA, age, stage, bone scan results, and treatment
[102]. Additionally they have been utilized to model recurrence post radical prostatectomy [103], to predict response
to immunotherapy [98] and to predict lymph node spread
[104]. Few of these studies use wide ranging cross platform
“omic” studies from multiple points in the disease progression pathway. Many have relied on existing bimolecular
tests, histopathology or clinical features. The need for integrated, multiplatform multicentre studies was highlighted by
Stephan et al. [99]. We have demonstrated through encouraging preliminary results that we can identify response to
therapy based on immunological and proteomic parameters.
We have also conducted preliminary work using ANNs to
integrate data and predicative models across multiple platforms.
4. Future applications
A number of groups are trying to identify proteins/peptides
and analyze from less complex fluids such as urine [33,105].
Proteomics may hold promise for early detection of disease
using proteomic patterns of body fluid samples, diagnosis as
a complement to histopathology based on proteomic patterns,
individualized selection of therapeutic combinations that best
target the entire disease-specific protein network, real-time
assessment of therapeutic efficacy and toxicity, and change
of therapy dependent upon the diseased protein network asso-
B. Matharoo-Ball et al. / Vaccine 25S (2007) B110–B121
ciated with drug resistance. Continued efforts at improving
the sensitivity of mass-spectrometry analyses will allow this
technology to be used on smaller tissue samples of prostate
cancer. An integrated approach to identifying prostate cancer
biomarkers using “Omic” technologies will prove to be of
value at the key clinical assessment points, strongly associating with disease progression status and treatment response.
In particular, we propose that it is imperative to adopt an integrated and complementary technologies and bioinformatics
analysis to elucidate profiles of stage specific disease markers
that can clearly discriminate patient sub-populations and predict treatment outcome and provide innovative biomarkers for
prostate cancer. This presents a unique opportunity to apply
this methodology to identify prostate cancer biomarkers and
determine their clinical utility.
[15]
[16]
[17]
[18]
[19]
[20]
References
[1] http://www.cancerworld.org/mediacentre/Prostate Cancer Factsheet.
aspx#top.
[2] Denis L, Murphy GP. Overview of phase III trials on combined androgen treatment in patients with metastatic prostate cancer. Cancer
1993;72:3888–95.
[3] Miles AK, Rogers A, Li G, Seth R, Powe D, McArdle SE, et al.
Identification of a novel prostate cancer-associated tumor antigen.
Prostate 2007;67(3):274–87.
[4] Thompson IM, Pauler DK, Goodman PJ, Tangen CM, Lucia MS,
Parnes HL, et al. Prevalence of prostate cancer among men with a
prostate-specific antigen level < or =4.0 ng per millilitre. N Engl J
Med 2004;350:2239–46.
[5] Catalona WJ, Smith DS, Ratliff TL, Dodds KM, Coplen DE, Yuan JJ,
et al. Measurement of prostate-specific antigen in serum as a screening
test for prostate cancer. N Engl J Med 1991;324:1156–61.
[6] Etzioni R, Penson DF, Legler JM, di Tommaso D, Boer R, Gann PH,
et al. Overdiagnosis due to prostate-specific antigen lessons from U.S.
prostate cancer incidence trends. J Natl Cancer 2002;94(13):981–90.
[7] Bradley EB, Bissonette EA, Theodorescu D. Determinants of quality
of life and voiding function of patients treated with radical prostectomy or permanent brachytherapy for prostate cancer. BJU Int
2004;94(7):1003–9.
[8] Ichikawa T, Suzuki H, Ueda T, Komiya A, Imamoto T, Kojima S.
Hormone treatment for prostate cancer: current issues and future
directions. Cancer Chemother Pharmacol 2005;56:58–63.
[9] Qu Y, Adam BL, Yasui Y, Ward MD, Cazares LH, Schellhammer PF,
et al. Boosted decision tree analysis of surface-enhanced laser desorption/ionization mass spectral serum profiles discriminates prostate
cancer from noncancer patients. Clin Chem 2002;48:1835–43.
[10] Feldman AL, Espina V, Petricoin EF, Liotta LA, Rosenblatt KP. Use
of proteomic patterns to screen for gastrointestinal malignancies.
Surgery 2004;135:243–7.
[11] Poon TC, Yip TT, Chan AT, Yip C, Yip V, Mok TS, et al. Comprehensive proteomic profiling identifies serum proteomic signatures for
detection of hepatocellular carcinoma and its subtypes. Clin Chem
2003;49:752–60.
[12] Li J, Zhang Z, Rosenzweig J, Wang YY, Chan DW. Proteomics and
bioinformatics approaches for identification of serum biomarkers to
detect breast cancer. Clin Chem 2002;48:1296–304.
[13] Petricoin EF, Ardekani AM, Hitt BA, Levine PJ, Fusaro VA, Steinberg
SM, et al. Use of proteomic patterns in serum to identify ovarian
cancer. Lancet 2002;359:572–7.
[14] Villanueva J, Philip J, Entenberg D, Chaparro CA, Tanwar MK, Holland EC, et al. Serum peptide profiling by magnetic particle-assisted,
[21]
[22]
[23]
[24]
[25]
[26]
[27]
[28]
[29]
[30]
[31]
[32]
[33]
B119
automated sample processing and MALDI-ToF mass spectrometry.
Anal Chem 2004;76:1560–70.
Grubb RL, Calvert VS, Wulkuhle JD, Paweletz CP, Linehan WM,
Phillips JL, et al. Signal pathway profiling of prostate cancer using
reverse phase protein arrays. Proteomics 2003;3(11):2142–6.
Paweletz CP, Charboneau L, Bichsel VE, Simone NL, Chen T, Gillespie JW, et al. Reverse phase protein microarrays which capture
disease progression show activation of pro-survival pathways at the
cancer invasion front. Oncogene 2001;20(16):1981–9.
Chen YT, Güre AO, Tsang S, Stockert E, Jäger E, Knuth A, et al.
Identification of multiple cancer/testis antigens by allogeneic antibody screening of a melanoma cell line library. Proc Natl Acad Sci
USA 1998;95(12):6919–23.
Bradford TJ, Wang X, Chinnaiyan AM. Cancer immunomics: using
autoantibody signatures in the early detection of prostate cancer. Urol
Oncol 2006;24(3):237–42.
Petricoin EF, Ornstein DK, Paweletz CP, Ardekani A, Hackett PS, Hitt
BA, et al. Serum proteomic patterns for detection of prostate cancer.
J Natl Cancer Inst 2002;94(20):1576–8, 16.
Adam BL, Qu Y, Davis JW, Ward MD, Clements MA, Cazares LH, et
al. Serum protein fingerprinting coupled with a pattern-matching algorithm distinguishes prostate cancer from benign prostate hyperplasia
and healthy men. Cancer Res 2002;62:3609–14.
Aebersold R, Mann M. Mass spectrometry-based proteomics. Nature
2003;422:198–207.
Clark RE, Dodi IA, Hill SC, Lill JR, Aubert G, Macintyre AR, et al.
Direct evidence that leukemic cells present HLA-associated immunogenic peptides derived from the BCR-ABL b3a2 fusion protein. Blood
2001;98(10):2887–93.
Bonner PL, Lill JR, Hill S, Creaser CS, Rees RC. Electrospray mass
spectrometry for the identification of MHC class I-associated peptides
expressed on cancer cells. J Immunol Meth 2002;262(1–2):5–19.
Ball G, Mian S, Holding F, Allibone RO, Lowe J, Ali S, et al. An integrated approach utilising artificial neural networks and SELDI mass
spectrometry for the classification of human tumours and rapid identification of potential biomarkers. Bioinformatics 2002;18:395–404.
Mian S, Ball G, Hornbuckle J, Holding F, Carmichael J, Ellis I, et al.
A prototype methodology combining surface-enhanced laser desorption/ionization protein chip technology and artificial neural network
algorithms to predict the chemoresponsiveness of breast cancer cell
lines exposed to Paclitaxel and Doxorubicin under in vitro conditions.
Cancer Immunol Immunother 2004;53(3):256–61.
Lotze MT, Rees RC. Identifying biomarkers and surrogates of tumors
(cancer biometrics): correlation with immunotherapies and immune
cells. Cancer Immunol Immunother 2004;53(3):256–61.
Mian S, Ugurel S, Parkinson E, Schlenzka I, Dryden I, Lancashire L,
et al. Serum proteomic fingerprinting discrimonates between clinical
stages and predicts disease progression in melanoma patients. J Clin
Oncol 2005;23:5088–93.
Matharoo-Ball B, Ratcliffe L, Lancashire L, Ugurel S, Miles AK,
Weston DJ, et al. Diagnostic biomarkers differentiating metastatic
melanoma patients from healthy controls identified by an integrated
MALDI-ToF mass spectrometry/bioinformatic approach. Proteomics
2007;1:605–20.
Clarke W, Zhang Z, Chan DW. The application of clinical proteomics to cancer and other diseases. Clin Chem Lab Med
2003;41(12):1562–70.
Liotta LA, Espina V, Mehta AI, Calvert V, Rosenblatt K, Geho D,
et al. Protein microarrays: meeting analytical challenges for clinical
applications. Cancer Cell 2003;3(4):317–25.
Utleg AG, Yi EC, Xie T, Shannon P, White JT, Goodlett DR, et al.
Proteomic analysis of human prostasomes. Prostate 2003;56:150–61.
Fung KY, Glode LM, Green S, Duncan MW. A comprehensive characterization of the peptide and protein constituents of human seminal
fluid. Prostate 2004;61:171–81.
M’Koma AE, Blum DL, Norris JL, Koyama T, Billheimer D, Motley
S, et al. Detection of pre-neoplastic and neoplastic prostate disease
B120
[34]
[35]
[36]
[37]
[38]
[39]
[40]
[41]
[42]
[43]
[44]
[45]
[46]
[47]
[48]
[49]
[50]
[51]
[52]
B. Matharoo-Ball et al. / Vaccine 25S (2007) B110–B121
by MALDI profiling of urine. Biochem Biophys Res Commun 2007
Feb 16;353(3):829–34.
Paweletz CP, Liotta LA, Petricoin III EF. New technologies for
biomarker analysis of prostate cancer progression: Laser capture
microdissection and tissue proteomics. Urology 2001;57:160–3.
Cazares LH, Adam BL, Ward MD, Nasim S, Schellhammer PF,
Semmes OJ, et al. Normal, benign, preneoplastic, and malignant
prostate cells have distinct protein expression profiles resolved by
surface enhanced laser desorption/ionization mass spectrometry. Clin
Cancer Res 2002;8:2541–52.
Diaz JI, Cazares LH, Corica A, John SO. Selective capture of prostatic
basal cells and secretory epithelial cells for proteomic and genomic
analysis. Urol Oncol 2004;22:329–36.
Lowenthal MS, Mehta AI, Frogale K, Bandle RW, Araujo RP, Hood
BL, et al. Analysis of albumin-associated peptides and proteins from
ovarian cancer patients. Clin Chem 2005;51:1933–45.
Manning JC, Seyrek K, Kaltner H, Andre S, Sinowatz F, Gabius HJ.
Glycomic profiling of developmental changes in bovine testis by lectin
histochemistry and further analysis of the most prominent alteration
on the level of the glycoproteome by lectin blotting and lectin affinity
chromatography. Histol Histopathol 2004;19:1043–60.
Gretzer MB, Chan DW, van Rootselaar CL, Rosenzweig JM, Dalrymple S, Mangold LA, et al. Proteomic analysis of dunning prostate
cancer cell lines withvariable metastatic potential using SELDI-ToF.
Prostate 2004;60:325–31.
Weinschenk T, Gouttefangeas C, Schirle M, Obermayr F, Walter
S, Schoor O, et al. Integrated functional genomics approach for
the design of patient-individual antitumour vaccines. Cancer Res
2002;62:5818–27.
Michener CM, Ardekani AM, Petricoin III EF, Liotta LA, Kohn
EC. Genomics and proteomics: application of novel technology
to early detection and prevention of cancer. Cancer Detect Prev
2002;26(4):249–55.
O’Farrell PZ, Gold LM. Transcription and translation of prereplicative
bacteriophage T4 genes in vitro. J Biol Chem 1973;248(15):5512–9.
Merril CR, Switzer RC, Van Keuren ML. Trace polypeptides in cellular extracts and human body fluids detected by two-dimensional
electrophoresis and a highly sensitive silver stain. Proc Natl Acad Sci
USA 1979;76(9):4335–9.
Patton WF. A thousand points of light: the application of fluorescence
detection technologies to two-dimensional gel electrophoresis and
proteomics. Electrophoresis 2000;21(6):1123–44.
Unlu M, Morgan ME, Minden JS. Difference gel electrophoresis:
a single gel method for detecting changes in protein extracts. Electrophoresis 1997;18(11):2071–7.
Ornstein DK, Gillespie JW, Paweletz CP, Duray PH, Herring J,
Vocke CD, et al. Proteomic analysis of laser capture microdissected
human prostate cancer and in vitro prostate cell lines. Electrophoresis
2000;21(11):2235–42.
Best CJ, Emmert-Buck MR. Molecular profiling of tissue samples using laser capture microdissection. Expert Rev Mol Diagn
2001;1(1):53–60.
Paweletz CP, Liotta LA, Petricoin III EF. New technologies for
biomarker analysis of prostate cancer progression: laser capture microdissection and tissue proteomics. Urology 2001;57(4):
160–3.
Ahram M, Best CJ, Flaig MJ, Gillespie JW, Leiva IM, Chuaqui RF,
et al. Proteomic analysis of human prostate cancer. Mol Carcinog
2002;33(1):9–15.
Lexander H, Palmberg C, Hellman U, Auer G, Hellstrom M, Franzen
B, et al. Correlation of protein expression, Gleason score and DNA
ploidy in prostate cancer. Proteomics 2006;6(15):4370–80.
Lexander H, Franzen B, Hirschberg D, Becker S, Hellstrom M,
Bergman T, et al. Differential protein expression in anatomical zones
of the prostate. Proteomics 2005;5(10):2570–6.
Johansson B, Pourian MR, Chuan YC, Byman I, Bergh A, Pang ST,
et al. Proteomic comparison of prostate cancer cell lines LNCaP-FGC
[53]
[54]
[55]
[56]
[57]
[58]
[59]
[60]
[61]
[62]
[63]
[64]
[65]
[66]
[67]
[68]
[69]
[70]
and LNCaP-r reveals heatshock protein 60 as a marker for prostate
malignancy. Prostate 2006;66(12):1235–44.
Rowland JG, Robson JL, Simon WJ, Leung HY, Slabas AR. Evaluation of an in vitro model of androgen ablation and identification
of the androgen responsive proteome in LNCaP cells. Proteomics
2007;7(1):47–63.
Qin S, Ferdinand AS, Richie JP, O’Leary MP, Mok SC, Liu
BC. Chromatofocusing fractionation and two-dimensional difference
gel electrophoresis for low abundance serum proteins. Proteomics
2005;5(12):3183–92.
Fountoulakis M, Gasser R. Proteomic analysis of the cell envelope
fraction of Escherichia coli. Amino Acids 2003;24(1–2):19–41.
Gygi SP, Corthals GL, Zhang Y, Rochon Y, Aebersold R. Evaluation of two-dimensional gel electrophoresis-based proteome analysis
technology. Proc Natl Acad Sci USA 2000;97(17):9390–5.
Washburn MP, Wolters D, Yates III JR. Large-scale analysis of the
yeast proteome by multidimensional protein identification technology. Nat Biotechnol 2001;19(3):242–7.
Stoeckli M, Chaurand P, Hallahan DE, Caprioli RM. Imaging mass
spectrometry: a new technology for the analysis of protein expression
in mammalian tissues. Nat Med 2001;7(4):493–6.
Xu BJ, Caprioli RM, Sanders ME, Jensen RA. Direct analysis of laser
capture microdissected cells by MALDI mass spectrometry. J Am Soc
Mass Spectrom 2002;13(11):1292–7.
Zheng Y, Xu Y, Ye B, Lei J, Weinstein MH, O’Leary MP, et al.
Prostate carcinoma tissue proteomics for biomarker discovery. Cancer
2003;98(12):2576–82.
Cheung PK, Woolcock B, Adomat H, Sutcliffe M, Bainbridge TC,
Jones EC, et al. Protein profiling of microdissected prostate tissue
links growth differentiation factor 15 to prostate carcinogenesis. Cancer Res 2004;64(17):5929–33.
Liu AY, Zhang H, Sorensen CM, Diamond DL. Analysis of
prostate cancer by proteomics using tissue specimens. J Urol
2005;173(1):73–8.
Ornstein DK, Rayford W, Fusaro VA, Conrads TP, Ross SJ, Hitt BA, et
al. Serum proteomic profiling can discriminate prostate cancer from
benign prostates in men with total prostate specific antigen levels
between 2.5 and 15.0 ng/ml. J Urol 2004;172:1302–5.
Pan YZ, Xiao XY, Zhao D, Zhang L, Ji GY, Li Y, et al. Application
of surface-enhanced laser desorption/ionization time-of-flight-based
serum proteomic array technique for the early diagnosis of prostate
cancer. Asian J Androl 2006;8(1):45–51.
Malik G, Ward MD, Gupta SK, Trosset MW, Grizzle WE, Adam BL,
et al. Serum levels of an isoform of apolipoprotein A-II as a potential
marker for prostate cancer. Clin Cancer Res 2005;11(3):1073–85.
Gross M, Top I, Laux I, Katz J, Curran J, Tindell C, et al. Beta-2microglobulin is an androgen-regulated secreted protein elevated in
serum of patients with advanced prostate cancer. Clin Cancer Res
2007;13(7):1979–86.
van den Bemd GJ, Krijgsveld J, Luider TM, van Rijswijk AL, Demmers JA, Jenster G. Mass spectrometric identification of human
prostate cancer-derived proteins in serum of xenograft-bearing mice.
Mol Cell Proteomics 2006;5(10):1830–9.
Baggerly KA, Morris JS, Coombes KR, Baggerly KA, Morris JS,
Coombes KR. Reproducibility of SELDI-ToF protein patterns in
serum: comparing datasets from different experiments. Bioinformatics 2004;20(5):777–85.
Lewczuk P, Esselmann H, Groemer TW, Bibl M, Maler JM, Steinacker
P, et al. Amyloid beta peptides in cerebrospinal fluid as profiled
with surface enhanced laser desorption/ionization time-of-flight mass
spectrometry: evidence of novel biomarkers in Alzheimer’s disease.
Biol Psychiatry 2004;55(5):524–30.
Semmes OJ, Feng Z, Adam BL, Banez LL, Bigbee WL, Campos
D, et al. Evaluation of serum protein profiling by surface-enhanced
laser desorption/ionization time-of-flight mass spectrometry for the
detection of prostate cancer. I. Assessment of platform reproducibility.
Clin Chem 2005;51(1):102–12.
B. Matharoo-Ball et al. / Vaccine 25S (2007) B110–B121
[71] Grizzle WE, Adam BL, Bigbee WL, Conrads TP, Carroll C, Feng Z,
et al. Serum protein expression profiling for cancer detection: validation of a SELDI-based approach for prostate cancer. Dis Markers
2003–2004;19(4–5):185–95.
[72] Lam YW, Mobley JA, Evans JE, Carmody JF, Ho SM. Mass
profiling-directed isolation and identification of a stage-specific serologic protein biomarker of advanced prostate cancer. Proteomics
2005;5(11):2927–38.
[73] Lopez MF, Mikulskis A, Kuzdzal S, Golenko E, Petricoin EF, Liotta
LA, et al. A novel, high-throughput workflow for discovery and identification of serum carrier protein-bound peptide biomarker candidates
in ovarian cancer samples. Clin Chem 2007;53(6):1067–74.
[74] Robson B. The dragon on the gold: myths and realities for data mining in biomedicine and biotechnology using digital and molecular
libraries. J Proteome Res 2004;3(6):1113–9.
[75] Lisboa PJ, Taktak AFG. The use of artificial neural networks in
decision support in cancer: a systematic review. Neural Networks
2006;19:408–15.
[76] Catto JWF, Linkens DA, Abbod MF, Chen M, Burton JL, Feeley KM,
et al. Artificial Intelligence in predicting bladder cancer outcome: a
comparison of neuro-fuzzy modelling and artificial neural networks.
Clin Cancer Res 2003;9:4172–7.
[77] Wagner M, Naik DN, Pothen A, Kasukurti S, Devineni RR, Adam
BL, et al. Computational protein biomarker prediction: a case study
for prostate cancer. BMC Bioinf 2004;5:26.
[78] Saidi S, Holland CM, Kreil DP, MacKay DJC, Charnock-Jones DS,
Print CG, et al. Independent component analysis of microarray data
in the study of endometrial cancer. Oncogene 2004;23(39):6677–83.
[79] Chu W, Ghahramani Z, Falciani F, Wild DL. Biomarker discovery in
microarray gene expression data with Gaussian processes. Bioinformatics 2005;21:3385–93, 16.
[80] Menard C, Johann D, Lowenthal M, Muanza T, Sproull M, Ross S,
et al. Discovering clinical biomarkers of ionizing radiation exposure
with serum proteomic analysis. Cancer Res 2006;66(3):1844–50.
[81] Chen Y-D, Zheng S, Yu J-K, Hu X. Artificial neural networks analysis of surface-enhanced laser desorption/ionization mass spectra of
serum protein pattern distinguishes colorectal cancer from healthy
population. Clin Cancer Res 2004;10:8380–5.
[82] Lancashire L, Schmid O, Shah H, Ball G. Classification of bacterial species from proteomic data using combinatorial approaches
incorporating artificial neural networks, cluster analysis and principal
components analysis. Bioinformatics 2005;21(10):2191–9.
[83] Khan J, Wei JS, Ringner M, Saal LH, Ladanyi M, Westermann F, et al.
Classification and diagnostic prediction of cancers using gene expression profiling and artificial neural networks. Nat Med 2001;7:673–9.
[84] Wei JT, Zhang Z, Barnhill SD, Madyastha KR, Zhang H, Oesterling
JE. Understanding artificial neural networks and their application in
urology. Urology 1998;52:161–72.
[85] Balls GR, Sanders GE, Palmer-Brown D. Investigating microclimatic influences on ozone injury in clover (Trifolium subterraneum
L. cv. Geraldton) using artificial neural networks. New Phytol
1996;132:271–80.
[86] Desilva CJS, Choong PI, Attikiouzel Y. Artificial neural networks and
breast cancer prognosis. Aust Comput J 1994;26:78–81.
[87] Anagnostou T, Remzi M, Djavan B. Artificial neural networks for
decision-making in urologic. Oncol Rev Urol 2003;5(1):15–21.
B121
[88] Tafeit E, Reibnegger G. Artificial neural networks in laboratory
medicine and medical outcome prediction. Clin Chem Lab Med
1999;37(9):845–53.
[89] Reckwitz T, Potter SR, Partin AW. Prediction of locoregional extension and metastatic disease in prostate cancer: a review. World J Urol
2000;18(3):165–72.
[90] Rumelhart DE, McClelland JL. Parallel distribution processing:
explorations in the microstructure of cognition, vol. 1: foundations.
Cambridge, MA: MIT Press; 1986.
[91] Bishop CM. Neural networks for pattern recognition. New York, NY:
Oxford University Press Inc.; 1995.
[92] Lisboa PJ. A review of evidence of health benefit from artificial neural networks in medical intervention. Neural Networks
2002;15(1):11–39.
[93] Asyali MH, Colak D, Demirkaya O, Inan MS. Gene expression profile
classification: a review. Curr Bioinformat 2006;1:55–73.
[94] Michiels S, Koscielny S, Hill C. Prediction of cancer outcome
with microarrays: a multiple random validation strategy. Lancet
2005;365:488–92, 2005.
[95] Sato F, Shimada Y, Selaru FM, Shibata D, Maeda M, Watanabe G, et
al. Prediction of survival in patients with esophageal carcinoma using
artificial neural networks. Cancer 2005;103:1596–605.
[96] Delen D, Walker G, Kadam A. Predicting breast cancer survivability.
Artif Intell Med 2005;34(2):113–27.
[97] Iversen C, Lancashire L, Waddington M, Forsyth S, Ball G. Identification of Enterobacter sakazakii from closely related species: the use
of artificial neural networks in the analysis of biochemical and 16S
rDNA data. BMC Microbiol 2006;6:28.
[98] Michael A, Ball G, Quatan N, Wushishi F, Russell N, Whelan J, et
al. Delayed disease progression after allogeneic cell vaccination in
hormone-resistant prostate cancer and correlation with immunologic
variables. Clin Cancer Res 2005;11(12):4469.
[99] Stephan C, Cammann H, Jung K. Artificial neural networks: has the
time come for their use in prostate cancer patients? Nat Clin Pract
Urol 2005;2(6):262–3.
[100] Stephan C, Meyer HA, Kwiatkowski M, Recker F, Cammann H, Loening SA, et al. A (−5, −7) proPSA based artificial neural network to
detect prostate cancer. Eur J Urol 2006;50(5):1014–20.
[101] Djavan B, Remzi M, Zlotta A, Seitz C, Snow P, Marberger M. Novel
artificial neural network for early detection of prostate cancer. J Clin
Oncol 2002;4:921–9.
[102] Naguib RN, Robinson MC, Neal DE, Hamdy FC. Neural network analysis of combined conventional and experimental prognostic
markers in prostate cancer: a pilot study. Br J Cancer 1998;78(2):
246–50.
[103] Mattfeldt T, Kestler HA, Hautmann R, Gottfried HW. Prediction
of prostatic cancer progression after radical prostatectomy using
artificial neural networks: a feasibilty study. BJU Int 1999;84:
316–23.
[104] Crawford ED, Batuello JT, Snow PB, et al. The use of artificial intelligence technology to predict lymph node spread in men with clinically
localized prostate cancer. Cancer 2000;88(9):2105–9.
[105] Rehman I, Azzouzi AR, Catto JW, Allen S, Cross SS, Feeley
K, et al. Proteomic analysis of voided urine after prostatic massage from patients with prostate cancer: a pilot study. Urology
2004;64(6):1238–43.