ARTICLE

Vol. 125 No. 1353 |

Beyond PSA: are new prostate cancer biomarkers of potential value to New Zealand doctors?

Full article available to subscribers

Prostate cancer is the most commonly registered male cancer in New Zealand making up 25.2% of all registrations, ahead of colorectal cancer and malignant melanoma of the skin, and the third most common cancer registration for both sexes.Prostate cancer was also the third leading cause of male cancer deaths in 20061. Although recent data might be interpreted as suggesting that there has been a decline in the incidence of prostate cancer since the year 20002, this may be an artefact of increased uptake of prostate-specific antigen (PSA) screening at that time. With increased PSA testing comes earlier diagnosis and registration of patients, which in turn will lead to an elevation of diagnosis in younger age groups (giving the pre-2000 increase).The apparent post-2000 decline is thus a result of those patients already being picked up by the test who would have otherwise been diagnosed at that time. The likely result is a paradigm shift in the age distribution of patients with diagnosed prostate cancer, and a return to a steady gradual increase in diagnosed prostate cancer patients, as seen in the pre-PSA years2.PSA testingthe current method of prostate cancer risk and progression assessment a prima facie, falls well short of the performance required of a screen in an age of evidence-based medicine, with sensitivity and specificity of PSA testing being quoted as 74-84% and 90-94% respectively3,4 and a positive predictive value of 21.9% (when using the traditional value of PSA 4.0 ng/mL as a threshold)5.Use of such a test as the basis of clinical decisions for prostate cancer patients renders active surveillance (a programme consisting of regular PSA and DRE (digital rectal examination) testing (in addition to regular biopsy of a patients prostatic tissue) or watchful waiting (where treatment has a stronger palliative element and curative treatments are foregone)116 as the most prudent course of action when a PSA level is shown to be in the grey zone of 2.5 ng/mL-10 ng/mL6.It should be noted, however, that active surveillance and watchful waiting, despite the implication of PSA values, are primarily indicated through key parameters of biopsy results, including Gleason score, clinical grade of disease, number of cores positive upon biopsy and volume of malignant tissue in each positive core. The current dependence on an invasive test for disease prognosis is reflective of the difficulty to differentiate between indolent and aggressive neoplasms with PSA, which is, in essence, a risk-stratification tool.Indeed, this is further underpinned when one observes the high rate of false positive (95 in 1000 men aged 55-69 years who have the PSA test) and a substantial number of false negative results (23 per 1000 men aged 55-69 who have PSA testing and then biopsy)3. As a result, the decisions surrounding treatment become extremely difficult if the sole basis for the decision to treat was a non-invasive test such as PSA (in practice, just as for active surveillance, the decision to treat is primarily indicated through parameters of prostate biopsy).Patients who do not need treatment may opt to be treated and suffer unnecessary side effects. Equally, those who do need treatment may choose not to be treated, and miss the opportunity for an early intervention. It is this dilemma which epitomises the experience of both patient and practitioner in dealing with the inherent uncertainty of PSA testing. Ideally, clinicians would be able to call on an accurate and reliable non-invasive risk-stratification system, whereby patients are empowered with precise knowledge to make more fully informed decisions on their health, and equally have a clearer understanding of the risk of recurrence8.This review discusses novel biomarkers in prostate cancer which have the potential to be incorporated in new risk-stratification systems, and their role in delivering the diagnostic and prognostic precision currently lacking in clinical prostate cancer treatment. We note that this list is not exhaustive, but covers several that would be potentially applicable to the New Zealand clinical situation.PSA testing: the status quoCurrent policy and practiceScreens for genetic susceptibility to breast cancer (BRCA1/2 screening114), or for the presence of early signs of cancer in the cervix (cervical cancer screening113) are both well established in Aotearoa/New Zealand. However, comparable well established methods are not available for screening genetic susceptibility to prostate cancer, despite the similarity in incidences of breast (2572 registrations, 2006) and prostate (2484 registrations, 2006) cancers2.The lack of a well substantiated and non-invasive screening test for early prostate cancer3 (as compared with PAP smear testing in cervical cancer) requires a more aggressive and concerted effort from policymakers, clinicians and researchers to address the uncertainties and errors manifest in the PSA test, which defines the current status of prostate screening and on a more global level, the plight of mens health, in this country.As a reflection of where the New Zealand healthcare system stands with its current prostate screening proceduresout of the eight criteria outlined by the New Zealand National Health Committee (NHC) screening assessment, prostate cancer screening meets only one criterionthat prostate cancer is a condition which is a suitable candidate for screening3. Indeed, PSA and direct rectal examination (DRE) are described as unsuitable tests as:cneither can be described as reliable, accurate, sensitive or specific enough for screening asymptomatic men.d National Health Committee (2004)However, there exists a growing body of evidence which tentatively suggests that screening for prostate cancer is not without its benefits. Specifically, criterion three outlined by the NHCthat there is an effective and accessible treatment or intervention for the condition identified through early detection3would seem to be supported by data presented from the Scandinavian Prostate Cancer Group-4 trial144 demonstrating a reduction in metastatic disease incidence (RR=0.65; p=0.006) and disease-specific death (RR=0.82; p=0.09) for clinically localised prostate cancer specimens after a 12-year follow-up period with radical prostatectomy, as compared to watchful waiting.Additionally, data extracted from a cohort of 7578 men in Sweden, randomised to screening, demonstrated a prostate cancer-specific mortality reduction of almost 50% (RR=0.56; p=0.002) over 14 years compared to non-screened controls145, which would provide randomised controlled trial evidence demanded by the fourth criterion stipulated by the NHCthat a screening programme is effective in reducing morbidity and mortality.Although the inevitable risk of overdiagnosis has been acknowledged by the study authors and elsewhere145,146, these recent developments perhaps signal that it may be pertinent to once again review the current government policy on prostate cancer screening.Strengths and limitationsPSA testing has demonstrable strengths. With 90% of new cases detected early enough for curative treatment115 (where the treatments offer cure rates between 70%-90%) and changes in prostate cancer mortality ranging from 10%-39% in countries in Western Europe, North America and Australia116 we can recognise that, although flawed, PSA is having a positive effect of the clinical treatment of prostate cancer.In addition, when we consider that prostate cancer has a tendency to progress slower than other cancers (and even slower with androgen ablation therapy), the burden associated with the myriad of medical interventions such as radiotherapy, surgery and hospice care will often become more costly than an early, curative intervention administered on the basis of a routine PSA test116.Moreover, the natural course of prostate cancer means that if we were to forego PSA testing and diagnose on the appearance of symptoms, 70% of these cases will already have metastases. It must be acknowledged too, that PSA should only be seen as the initial step in prostate cancer assessmentTRUS (transrectal ultrasound) biopsy remains the gold standard in delivering diagnostic and prognostic data on prostate cancer. Figure 1. Current use of PSA in monitoring progression, diagnosis and prognosis of disease Note: The PSA Grey Zone (2.5ng/uL - 10ng/uL) 6 extends across the whole continuum of prostate cancer progression. These recognised limitations of PSA testing have led to international initiatives towards developing and validating new biomarkers with higher sensitivity and specificity which alone, or in conjunction with current screening methods, are able to deliver more definitive results on the presence and nature of cancer in the prostate, in a fast, cost-effective and non-invasive manner. Through the clinical application of novel biomarkers and effective implementation in the healthcare system, clinicians may aspire to deliver well informed and clear-cut decisions on the course of prostate cancer patients treatments and prognoses, and ultimately deliver better health outcomes for men in Aotearoa/New Zealand. Novel biomarkers: beyond PSA As researchers delve further into the elements underlying sporadic prostate cancer, we begin to unearth increasing evidence of this being a heterogeneous disease18. Unlike the discovery of the Bcr-Abl gene in chronic myeloid leukaemia, it is unlikely that more research will reveal a single specific gene locus that is responsible for prostate cancer. Naturally, such a multifaceted disease demands an equally multifaceted approach to risk-stratification, screening and diagnosis. Novel biomarkers for sporadic prostate cancer have been found on many echelons of the central dogma of genetics: genetic (specifically DNA), epigenetic, transcriptomic, proteomic and metabolomic approaches all show promise for use in clinical medicine in the future. Genomics TMPRSS2-ERGThis marker can be detected using RT-PCR methods, applied to urine samples from subjects whose prostate has been massaged. Discovery of this gene fusion is potentially the most significant advance in the last decade in the molecular pathology of prostate cancer.TMPRSS2 is a prostate specific gene19,20 on chromosome 21 that codes for a transmembrane-bound serine protease20. The protease is predicted to react with a number of proteins on the cell surface, as well as extracellular matrix components, soluble proteins and proteins on nearby cells21.ERG is a member of the ETS family of transcription factors which are able to activate or repress expression of genes involved in cellular proliferation, differentiation and apoptosis22. Figure 2. The potential significance of the TMPRSS2-ERG fusion Note: The androgen-sensitive promoter region of the TMPRSS2 gene, through fusion to ETS family genes, could lead to androgen-driven overexpression of ETS family genes such as ERG. These in turn have been shown to cause downstream effects such as a high expression of the histone deacetylase I (HDAC I) gene, upregulation of Wnt pathways and downregulation of tumour necrosis factor and cell death pathways.23 Genes from the ETS family and TMPRSS2 lie nearby on chromosome 21, and hence fusions typically occur via rearrangements including deletion and translocation24. Cross et al22have suggested the possibility of certain sequences in TMPRSS2 and ERG which make some men more prone to these fusions that are seen in 49% of localised prostate cancers22. Furthermore, the timing of the occurrence of these fusions is particularly significant - TMPRSS2-ERG fusions have not been detected in morpohologically benign prostatic tissue but arise at a very specific point in the pathogenesis of prostate cancer, namely the high-grade prostatic intra-epithelial neoplastic stage (HGPIN) (essentially analogous to carcinoma in situ). In addition, in late-stage androgen receptor-negative cancers, TMPRSS2-ERG fusions were still present in the DNA but were not expressed25, which aligns with the current understanding of the bypass mechanisms involved in androgen-independence and the fact that TMPRSS2 contains an androgen-dependent promoter region22. The clinical significance of these novel discoveries in the TMPRSS2-ERG fusion will be delineated more clearly as further studies are published. In terms of prognostication, there have been groups who have looked at TMPRSS2-ERG fusions in comparison to measures such as Gleason Score, survival data and tumour recurrence. In general, TMPRSS2-ERG fusions were shown to be linked with worse prognoses22: 44% of Gleason pattern 5 contained TMPRSS2-ERG fusions compared with 7% of Gleason pattern 2 tumours26 Non-fusion patients had a 90% survival at 8 years compared with 25% survival at 8 years in those identified having a particular pattern of TMPRSS2 fusion known as 2+ Edel (duplication of TMPRSS2-ERG fusion sequences and interstitial deletion of sequences 5 to ERG)27 Tumours with TMPRSS2-ERG fusions had a higher recurrence rate after radical prostatectomy with an odds ratio of 7.1 (95% confidence interval 1.1-45)28. Despite their prostate specificity and their appearance in Prostatic Intra-epithelial Neoplasia (PIN), TMPRSS2-ERG fusions are unlikely to be suitable for screening as they have been found by Hessels et al29 to show low sensitivity (37% in a cohort of 108). However, in the same study, the fusions were detected with a positive predictive value (PPV) of 94%29, which suggests that it could be a useful risk-assessment tool whereby a clinician could request further biopsies in the cases where patients have a negative initial biopsy but persistently elevated PSA and positive test for the gene fusion product. A similar pattern of low sensitivity but a high positive predictive value is seen in TMPRSS2-ERG fusions and their association with five key histological features30: Blue-tinged mucin Cribriform growth pattern Intraductal tumour spread Macronucleoli Signet-ring cell features Ninety-three percent of cases in 253 prostate cancers with three of more of these features were TMPRSS2-ERG fusion positive (high PPV) but equally, 24% of TMPRSS2-ERG fusions did not show any of these features (low sensitivity) 30. Its positive predictive value is comparable to the morphological features of HNPCC and BRCA-associated breast cancers, but the link between genotype and phenotype is not yet fully understood. Tumour morphology and association between TMPRSS2-ERG fusions thus stands as a potentially useful addition to the current armoury of diagnostic and risk-stratification methods, but further research is required in the field before we see collaboration between clinicians and histopathologic and cytogenetic services in New Zealand. Urinary 8-hydroxydeoxyguanosine (8-OHdG)It is widely agreed that reactive oxygen species (ROS) are direct causes of DNA damage. 8-hydroxydeoxyguanosine (8-OHdG), an oxidised nucleoside of DNA, is a frequently detected lesion where mismatch repair plays a key role43.Upon DNA repair, 8-OHdG is excreted in the urine and thus can not only be a measure of DNA repair capacity, but also a biomarker for oxidative stress and potential carcinogenic initiation44, 45. Increased urinary DNA lesions were detected by Chiou et al43 in both prostate and bladder cancer patients (58.5ng/mg creatinine of urinary DNA lesions in prostate cancer patients compared with 36.1ng/mg creatinine of Urinary DNA lesions in healthy patients) with a sensitivity of 31% and a specificity of 100%. Although their study population was small (and the fact that a biomarker of oxidative stress is not prostate-specific), the specificity of the test and the non-invasive nature of it suggests that with further investigation urinary 8-OHdG has potential as a biomarker which can allow for risk-stratification in those who have elevated serum PSA or a strong family history of prostate cancer. 8-OHdG is frequently detected in both non-malignant and malignant tissue. However, in non-malignant tissues extensive oxidative DNA damage drives cells to cell-cycle arrest (metabolic blockage), while in neoplastic prostate cancer cells it activates repair mechanisms favouring the escape from senescence and the expansion of DNA-damaged clones133.The combination of 8-OHdG in urine, measured along with cell-cycle check point evaluators such as CDKN1A, a cyclin-dependent kinase inhibitor and the product of the growth-arrested and DNA damage inducible gene Gadd45, from a parallel blood sample, may provide a greater understanding of the progression towards malignancy134, 135. Transcriptomics HepsinHepsin is a type II membrane associated serine protease whose structure and similarity to other serine proteases suggests that hepsin is involved in tumour growth, and hence hepsin stands as an attractive target in cancer biomarker development. Its prostate-specificity is best demonstrated through evidence of overexpression of hepsin (median 46.1-fold) in cancerous prostate tissue in 90% of prostate cancer samples (n=90)46. These findings have been confirmed through the work of Magee et al47 in an analysis of 4712 genes. In the same analysis, Hepsin was found to be over-expressed in prostatic intra-epithelial neoplasia in comparison to BPH which points to a relationship between Hepsin and neoplastic transformation. In addition, one can propose that such a biomarker can aid in the prognostication of Gleason 4 and 5 tumours with the discovery of a correlation between increased Hepsin expression and higher Gleason score46. The major shortcoming of the use of Hepsin is the fact that it can only be detected in tissue specimens and, despite attempts to use RNA extracted from urine for quantitating hepsin136 is not currently detectable from urine or serum samples48. Thus, the arrival of Hepsin as a prognostic tool for differentiation of indolent from aggressive tumours depends firmly on the discovery of novel methods of detection that will render it more accessible to clinical practice. Prostate cancer antigen 3 (DD3PCA3)DD3PCA3 is a novel, prostate-specific gene found to be up-regulated in cancerous prostate cells and over-expressed in >95% of clinical specimens31,33. PCA3 is more specific for prostate cancer than serum prostate-specific antigen (PSA), which is prostate-specific but not cancer-specific41. The proof of its prostate specificity has been shown through RT-PCR methodologies, in which PCA3 mRNA expression was low but quantifiable in benign prostatic tissue, but undetectable in normal and malignant tissue from other organs32. Equally, proof of over-expression of DD3PCA3in malignant prostate tissue with a median 66-fold up-regulation (compared to expression in benign tissue) has been demonstrated by Northern Blot analyses31. DD3PCA3 has been concluded to express non-coding mRNA (defined through the presence of alternative splicing, polyadenylation, lack of an extended open reading frame and numerous stop codons) for which there is no discrete cytoplasmic protein productdespite overexpression of the mRNA transcript31. The function of the DD3PCA3 gene and its non-coding mRNA transcript are currently undefined; hence, there is equally little known about the role of the DD3PCA3 gene in pathogenesis of prostate cancer. The magnitude of overexpression of the DD3PCA3 gene in malignant specimens when compared to the near-negligible amounts of DD3PCA3 expression in benign prostatic tissue confirms that the ultimate cause of the lack of a cytoplasmic protein product from PCA3 mRNA expression lies in the transcription as opposed to translation of other processing steps31. Although conflicting literature does exist on the subject of the DD3PCA3 genes clinical utility, the majority pertaining to the matter confirm that DD3PCA3 has strong diagnostic value, particularly in differentiating early-stage prostate cancer from benign prostatic hyperplasia (BPH)34,35,36. PPV of 52.2% in men with PCA3 \u2265100 is reported by Roobol et al 2010a and Robool et al 2010b. This marker stands as one of the most attractive risk-stratification tools to detect early prostate cancer for a gamut of reasons: The DD3PCA3 test does not require a biopsy- the mRNA is collected from urine after DRE and prostatic massage34. DD3PCA3 levels are directly reflective of tumour burden (as it is mRNA from cancer cells) and are not affected by prostate size, unlike PSA (which is a surrogate serum marker). This reduces the number of false positives detected in BPH cases and hence increases overall specificity32. DD3PCA3 mRNA expression adds the most value to current diagnostic tools at PSA values between 2.5ng/ml and 4.0ng/ml34. The quantitative PCA3 score has been found to correlate to the frequency of prostate cancer-positive biopsythus it can act as a means to stratify patients into categories of prostate cancer risk32. In theory, it has all the hallmarks of a test which can deliver the much sought after specificity that is currently lacking in determining whether to biopsy or not. However, current validation studies have struggled to produce definitive results confirming DD3PCA3 mRNA as a clinically applicable biomarker. Five studies which look the performance of DD3PCA3 which use \u22652.5ng/ml or \u22653.0ng/ml as PSA cut-off values gave the following values (as an average across the five studies)37, 38,39,40,41: PPV: 28.3% Sensitivity: 62.6% Specificity: 74.8% (Sample Size [average]: 303) Values for sensitivity have been quoted as high as 82% at 2.5ng/ml PSA cut-off42 and for specificity. Mearini et al34 claim 100% sensitivity (when PSA and DD3PCA3 are combined) in a tPSA range <4ng/ml. It must also be noted that PCA3 scores and PSA cut-offs can be varied to change the specificity and sensitivity, whereby a higher PCA3/PSA cut-off will produce very high specificity (i.e. very few false positive results) but much compromised sensitivity (high number of false negative results) and vice versa with lowered cut-off values. In addition, the means by which PCA3 is assayed for (i.e. the technology used) can also alter these results. What these values demonstrate is a classic teething issue of a novel biomarker; the lack of consistency in the type of assay used to identify the marker as well as small sample sizes hampers the production of consistent results and ultimately prevents the attainment of a definitive answer on the applicability of DD3PCA3 as a prostate cancer biomarker. This being said, its prostate-specificity and its potential to differentiate between indolent neoplasms and early malignant tumours ensures that further extensive research will be conducted into the utility of DD3PCA3 as a biomarker aiding clinicians in early diagnosis of prostate cancer. Epigenomics Glutathione-S-transferase P1 (GSTP1)From the family of Glutathione-S-transferases, GSTP1 conjugates chemically reactive electrophiles with glutathione, thus preventing DNA damage from reactive oxygen species and carcinogens which release reactive electrophilic metabolites49. Promoter hypermethylation of the region expressing GSTP1 has been directly linked to the loss of GSTP1 expression in prostate cancer50,51,52; indeed, this somatic genomic alteration is manifest in over 90% of prostate cancersmaking it the most frequent epigenetic event reported in prostate cancer51,52,53. With respect to its role in cancer pathogenesis, GSTP1 hypermethylation and the resulting loss of expression is a process presently considered as a promoter of cancer (as opposed to an initiator), with loss of GSTP1 increasing susceptibility of DNA to oxidants and free radicals54. GSTP1 hypermethylation is an attractive target for more intensive investigation into its role as a prostate cancer biomarker for many reasons: Its role in the pathogenesis of prostate cancer has been elucidated and the mechanism is well understood. GSTP1 hypermethylation is not frequently observed in normal prostate tissue50,53(although there have been reports of GSTP1 hypermethylation in high grade prostatic intra-epithelial neoplasia). GSTP1 hypermethylation is less frequent in non-prostate genitourinary malignancies (e.g. renal and bladder cancer) 54. GSTP1 is not limited by the accessibility of sample collection; it can be identified in a range of body fluids: urine, serum, and ejaculate 54. Although non-invasive procedures including collection of urine and ejaculate are held as the ideal means of attaining diagnostic information, there are key shortcomings with the use of these tissues. It has been shown that GSTP1 methylation levels are higher in plasma compared to urine, suggesting that prostate cancer is preferentially disseminated into the bloodstream rather than the prostatic ductal system54. With ejaculate, the inherent nature of such a collection procedure, particularly with older men, renders this avenue as one unlikely to see significant clinical exposure. Solutions such as prostatic massage to release cancer cells into the prostatic urethra before collection have so far delivered mixed results48,58,59. The difficulties faced in attaining clinically applicable detection rates through non-invasive methods remains a barrier yet to be surmounted. Currently, the most promising results portraying GSTP1 hypermethylation have been produced from tissue samples. The use of quantitative methylation specific PCR (QMSP) in screening for GSTP1 methylation has been reported to deliver 85.5% sensitivity and 96.8% specificity (n=128)56. When further tests were conducted on the same set of tissue specimens to assess the capacity for differentiation between non-cancerous tissue and histologically-proven adenocarcinoma (n=21), the QMSP assay correctly diagnosed the specimens with 90.9% sensitivity and 100% specificity and 100% positive predictive value. In addition, Harden et al57 demonstrate a 15% increase in specificity of the gold-standard of prostate diagnosishistopathologic assessmentthrough combining histopathologic assessment with QMSP for GSTP1. Furthermore, there is evidence that this method may be complemented with a measure of ENT SCTR methylation137. These results highlight the potential for GSTP1 hypermethylation as a means of complementing histopathological diagnosis of prostate samples and furthermore, a means of differentiating indolent and malignant neoplasms in cases where PSA levels alone are unable to discriminate56. Wnt signalling and methylationWnt signalling and its subsequent pathways are known to be crucial in mammalian and embryonic development60, 61. Its role in the pathogenesis of cancer can be summarised by the following diagram (modified from van der Poel HG60): Figure 3. Potential involvement of the Wnt pathway in the development of malignancy. The steps portrayed are: Binding of Wnt ligand to the frizzled transmembrane receptor. Decreased phosphorylation of B-catenin by GSK3-B. Therefore, stabilised B-catenin now accumulates in the nucleus. Nuclear B-catenin converts the TCF/LEF DNA binding complex from a transcriptional repressor into a transcriptional activator. Transcriptional activation of many cancer-related genes61. In the case of prostate cancer, there are a handful of epigenetic changes which are thought to alter the Wnt signalling pathway: Hypermethylation of the APC (adenomatous polyposis coli) gene is increased 8-fold in prostate tumours relative to samples of benign prostatic hypertrophy61. It has been proposed that DNA hypermethylation of the APC gene, an important component of the B-catenin degradation complex, may lead to the nuclear accumulation of B-catenin and hence the activation of the Wnt signalling pathway activating various oncogenes61. Equally, E-cadherin is a cell-membrane protein, which is known to both interact with B-catenin and be involved in the process of epithelial-to-mesenchymal transmission (EMT), a key step in the development of malignancy63. When the promoter for the E-cadherin gene is silenced by methylation, it not only promotes EMT but also the release of B-catenin away from the cell membrane and into the cytoplasmic and nuclear compartments. The presence of B-catenin in the nucleus will hence activate Wnt signalling61,62. Secreted-frizzled related proteins (SFRPs) and Wnt inhibitory factor-1 (Wif-1) are antagonists for Wnt signalling. Thus, silencing of genes which express SFRPs and Wif-1 through hypermethylation will lead to aberrant Wnt signalling and cancer progression. Although silencing of genes encoding SFRPs and Wif-1 has been identified in many cancers, including colorectal, lung, and bladder cancers and lymphocytic leukaemia64, there is insufficient evidence to definitively claim that Wnt antagonist genes play a key role in prostate cancer development. Despite the extensive elucidation of the Wnt signalling pathway, there remain questions over its relevance to prostate cancer and whether assays for hypermethylation of any of the aforementioned genes will aid the delineation of a diagnostic landscape. However, the role of potential cancer promoters, exemplified by Wnt signalling

Authors

Lance Ng, Research Student, Auckland Cancer Society Research Centre, University of Auckland; Nishi Karunasinghe; Research Technician, Auckland Cancer Society Research Centre, University of Auckland; Challaraj S Benjamin, Professor, Consultant Oncologist, Auckland Cancer Society Research Centre, University of Auckland and Department of Clinical Oncology, Auckland Hospital, Auckland; Lynnette R Ferguson; Professor, Head of Department, Nutrition, School of Medical Sciences, Auckland Cancer Society Research Centre, University of Auckland

Correspondence

Lance Ng, Research Student, Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland, New Zealand.

Correspondence email

lng036@aucklanduni.ac.nz

Competing interests

None declared.

Ministry of Health. Cancer: New Registrations and Deaths 2006. Wellington, New Zealand, Ministry of Health 2010.Ministry of Health. Cancer Projections: Incidence 2004-08 to 2014-18. Wellington, Ministry of Health 2010.National Advisory Committee on Health and Disability. Prostate Cancer Screening in New Zealand. Wellington, National Health Committee 2004.Durham J. Population Screening for Prostate Cancer. A Systematic Review, New Zealand Guidelines Group 2002.Schroder FH, Bangma CH, Roobol MJ. \"Is it Necessary to Detect All Prostate Cancers in men with Serum PSA Levels <3.0 ng/ml? A Comparison of Biopsy Results of PCPT and Outcome-Related Information from ERSPC.\" Eur Urol 2008. 53: 901-908.Klecka J, Holubec L, Pesta M, et al. \"Differential Display Code 3 (DD3/PCA3) in Prostate Cancer Diagnosis.\" Anticancer Res 2010. 30: 665-670.Loeb S and Schaeffer EM. \"Risk Factors, Prevention and Early Detection of Prostate Cancer.\" Primary Care: Clinics in Office Practice 2009. 36(3): 603-621.Benedettini E, Nguyen P, Loda M. \"The pathogenesis of prostate cancer: from molecular to metabolic alterations.\" Diagn Histopathol 2008. 14(5): 195-201.Roehl KA, Loeb S, Antenor JAV et al. \"Characteristics of Patients with Familial Versus Sporadic Prostate Cancer.\" J Urol 2006. 176: 2438-2442.Gronberg H, Smith J, Emanuelsson M, et al. \"In Swedish Families with Hereditary Prostate Cancer, Linkage to the HPC1 Locus on Chromosome 1q24-25 is Restricted to Families with Early-Onset Prostate Cancer.\" A J Hum Genet 1999. 65(1): 134-140.Bratt O. \"Hereditary Prostate Cancer: Clinical Aspects.\" J Urol 2002. 168(3): 906-913.Lichtenstein P, Holm NV, Verkasalo PK et al. \"Environmental and heritable factors in the causation of cancer: analyses of cohorts of twins from Sweden, Denmark and Finland.\" N Engl J Med 2000. 343: 78.Valeri A, Azzouzi R, Drelon E, et al. \"Early-onset hereditary prostate cancer is not associated with specific clinical features.\" Prostate 2000. 45: 66.Bratt O, Damber J.-E, Emanuelsson M, Gronberg H. \"Hereditary prostate cancer: clinical characteristics and survival.\" J Urol 2002. 167: 2423.Xu J and the International Consortium for Prostate Cancer Genetics. \"Combined Analysis of Hereditary Prostate Cancer Linkage to 1q24-25: Results from 772 Hereditary Prostate Cancer Families from the International Consortium for Prostate Cancer Genetics.\" Am J Hum Genet 2000. 66(3): 945-957.Freedman ML, Haiman CA, Patterson N et al. \"Admixture mapping identifies 8q24 as a prostate cancer risk locus in African American men.\" Proc Nat Acad Sci USA 2006. 103: 14068-73.Meyer A, Schurmann P, Ghahremani M, et al. \"Association of chromosomal locus 8q24 and risk of prostate cancer: A hospital based study of German patients treated with brachytherapy.\" Urol Oncol 2008. 27(4): 373-376.Hessels D, Rittenhouse HG, Schalken JA. \"Molecular Diagnostics in Prostate Cancer.\" Eur Urol: EAU Update Series 2005. 3(4): 200-213.Afar DE, Vivanco I, Hubert RS, et al. \"Catalytic cleavage of the androgen-regulated TMPRSS2 protease results in its secretion by prostate and prostate cancer epithelia.\" Cancer Res 2001. 61: 1686-92.Paolini-Giacobino A, Chen H, Peitsch MC, et al. \"Cloning of the TMPRSS2 gene, which encodes a novel serine protease with transmembrane, LDLRA and SRCR domains and maps to 21q22.3.\" Genomics 1997. 44: 309-20.Wilson S, Greer B, Hooper J, et al. \"The membrane-anchored serine protease, TMPRSS2, activates PAR-2 in prostate cancer cells.\" Biochem J 2005. 388: 967-72.Cross SS, Rehman I, Hamdy FC, et al. \"TMPRSS2 Fusions in Prostate Cancer.\" Diagn Histopathol 2008. 14(3): 1.Iljin K, Wolf M, Edgren H, et al. \"TMPRSS2 fusions with oncogenic ETS factors in prostate cancer involve unbalanced genomic rearrangements and are associated with HDAC1 and epigenetic programming.\" Cancer Res 2006. 66: 10242-6.Tomlins SA, Rhodes DR, Perner S, et al. \"Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer.\" Science 2005.310: 644-648.Hermans KG, van Marion R, van Dekken H, et al. \"TMPRSS2-ERG fusion by translocation or interstitial deletion is highly relevant in androgen-dependent prostate cancer, but is bypassed in late-stage androgen receptor-negative prostate cancer.\" Cancer Res 2006. 66: 10658-63.Rajput AB, Miller MA, De Luca A, et al. \"Frequency of the TMPRSS2-ERG gene fusion is increased in moderate to poorly differentiated prostate cancers.\" J Clin Pathol 2007.60: 1238-43.Demichelis F, Fall K, Perner S, et al. \"TMPRSS2-ERG gene fusion associated with lethal prostate cancer in a watchful waiting cohort.\" Oncogene 2007. 26: 4596-9.Attard G, Clark J, et al. \"Duplication of the fusion of TMPRSS2 to ERG sequences identifies fatal human prostate cancer.\" Oncogene 2007. 27(3): 253-63.Hessels D, Smit FP, Verhaegh GW, et al. \"Detection of TMPRSS2-ERG fusion transcripts and prostate cancer antigen 3 in urinary sediments may improve diagnosis of prostate cancer.\" Clin Cancer Res 2007. 13: 5103-8.Mosquera JM, Perner S, Demichelis F, et al. \"Morphological features of TMPRSS2-ERG gene fusion prostate cancer.\" J Pathol 2007. 212: 91-101.Schalken JA, Hessels D, Verhaegh G. \"New Targets for Therapy in Prostate Cancer: Differential Display Code 3 (DD3PCA3), a Highly Prostate Cancer-Specific Gene.\" Urology 2003. 62: 34-43.Schalken JA. \"Towards Early and More Specific Diagnosis of Prostate Cancer? Beyond PSA: New Biomarkers Ready for the Prime Time.\" Eur Urol Suppl 2009. 8: 97-102.Bussemakers MJ, van Bokhoven A, Verhaegh GW, et al. \"DD3: A New Prostate-Specific Gene, Highly Over-Expressed in Prostate Cancer.\" Cancer Res 1999. 59: 5975-5979Mearini E, Antognelli C, Del Buono C, et al. \"The combination of urine DD3PCA3 mRNA and PSA mRNA as molecular markers of prostate cancer.\" Biomarkers 2009. 14(4): 235-243.Groskopf J, Aubin SMJ, Deras IL, et al. \"APTIMA PCA3 Molecular urine Test: Development of a Method to Aid in the Diagnosis of Prostate Cancer.\" Clin Chem 2006. 52(6): 1089-1095.Vaananen RM, Rissanen M, Kauko O, et al. \"Quantitative real-time RT-PCR assay for PCA3.\" Clin Biochem 2008. 41: 103-8.Hessels D, Klein Gunnewiek JMT, van Oort I, et al. \"DD3PCA3-based molecular urine analysis for the diagnosis of prostate cancer.\" Eur Urol 2003. 44: 8-16.van Gils MP, Cornel EB, Hessels D, et al. \"Molecular PCA3 diagnostics on prostatic fluid.\" Prostate 2007. 67: 881-7.Kranse R, Beemsterboer PMM, Rietbergen JBW, et al. \"Predictors for biopsy outcome in the European Randomized Study of Screening for Prostate Cancer (Rotterdam region).\" Prostate 1999. 39: 316-22.Marks LS, Fradet Y, Deras IL, et al. \"PCA3 molecular urine assay for prostate cancer in men undergoing repeat biopsy.\" Urology 2007. 69: 532-5.Deras IL, Aubin SMJ, Blase A, et al. \"PCA3: a molecular urine assay for predicting prostate biopsy outcome.\" J Urol 2008. 179: 1587-92.Tinzl M, Marberger M, Horvath S, Chypre C. \"DD3PCA3 RNA Analysis in Urine - A new Perspective for Detecting Prostate Cancer.\" Eur Urol 2004. 46: 182-187.Chiou C, Chang P, Chan E, et al. \"Urinary 8-hydroxydeoxyguanosine and its analogs as DNA marker of oxidative stress: development of an ELISA and measurement in both bladder and prostate cancers.\" Clin Chim Acta 2003. 334: 87-94.Cooke MS, Evans MD, Herbert KE, Lunec J. \"Urinary 8-oxo-2'-deoxyguanosine-source, significance and supplements.\" Free Radic Res 2000. 32: 381-97.Helbock HJ, Beckman KB, Ames BN. \"8-Hydroxydeoxyguanosine and 8-hyroxyguanine as biomarkers of oxidative DNA damage.\" Methods Enzymol 1999. 300: 156-66.Stephan C, Yousef GM, Scorilas A, et al. \"Hepsin is highly over expressed in and a new candidate for a prognostic indicator in prostate cancer.\" J Urol 2004. 171: 187-191.Magee JA, Araki T, Patil S, et al. \"Expression profiling reveals hepsin overexpression in prostate cancer.\" Cancer Res 2001. 61: 5692.Parekh DJ, Ankerst DP, Troyer D, et al. \"Biomarkers for Prostate Cancer Detection.\" J Urol 2007. 178: 2252-2259.Stangelberger A, Margreiter M, Seitz C, Djavan B. \"Prostate Cancer Screening Markers.\" JMHG 2007. 4(3): 233-244.Millar DS, Ow KK, Paul CL, et al. \"Detailed methylation analysis of the glutathione S-transferase pi (GSTP1) gene in prostate cancer.\" Oncogene 1999. 18: 1313-24.Lin X, Tascilar M, Lee WH, et al. \"GSTP1 CpG island hypermethylation is responsible for the absence of GSTP1 expression in human prostate cancer cells.\" Am J Pathol 2001. 159: 1815-26.Jeronimo C, Varzim G, Henrique R, et al. \"1105V polymorphism and promoter methylation of the GSTP1 gene in prostate adenocarcinoma.\" Cancer Epidemiol. Biomarkers Prev 2002. 11: 445-50.Brooks JD, Weinstein M, Lin X, et al. \"CG island methylation changes near the GSTP1 gene in prostatic intraepithelial neoplasia.\" Cancer Epidemiol. Biomarkers Prev. 1998. 7: 531-6.Henrique R and Jeronimo C. \"Molecular Detection of Prostate Cancer: A Role for GSTP1 Hypermethylation.\" Eur Urol 2004. 46: 660-669.Lin X, Asgari K, Putzi MJ, et al. \"Reversal of GSTP1 CpG island by hypermethylation and reactivation of pi-class glutathione-S-transferase (GSTP1) expression in human prostate cancer cells by treatment with procainamide.\" Cancer Res. 2001. 61: 8611-6.Jeronimo C, Usadel H, Henrique R, et al. \"Quantitation of GSTP1 methylation in non-neoplastic prostatic tissue and organ-confined prostate adenocarcinoma.\" J Natl Cancer Inst 2001. 93: 1747-52.Harden SV, Sanderson H, Goodman SN, et al. \"Quantitative GSTP! methylation and the detection of prostate adenocarcinoma in sextant biopsies.\" J Natl Cancer Inst 2003. 95: 1634-7.Crocitto LE, Korns D, Kretzner L, et al. \"Prostate Cancer Molecular Markers GSTP1 and hTERT in expressed prostatic secretions as predictors of biopsy results.\" Urology 2004. 64: 821.Goessl C, Muller M, Heicappell R, et al. \"DNA-based detection of prostate cancer in urine after prostatic massage.\" Urology 2001. 58: 335.van der Poel HG. \"Molecular markers in the diagnosis of prostate cancer.\" Crit Rev Oncol Hematol 2007. 61: 104-139.Greene KL, Li L, Okino ST, Carroll PR. \u201cMolecular Basis of Prostate Cancer\u201d. The Molecular Basis of Cancer (Third Ed.) 2008, Elsevier: 431-440.Verras M and Sun Z. \"Roles and regulation of Wnt signalling and beta-catenin in prostate cancer.\" Cancer Lett 2006. 237: 22.Li LC, Okino ST, Dahiya R. \"DNA methylation in prostate cancer.\" Biochim et Biophys Acta 2004. 1704: 87.Urakami S, Shiina H, Enokida H, et al. \"Epigenetic inactivation of Wnt inhibitory factor-1 plays an important role in bladder cancer through aberrant canonical Wnt/beta-catenin signalling pathway.\" Clin Cancer Res 2006. 12: 383.Ministry of Health. Tobacco Trends 2007: A brief update on monitoring indicators. Wellington, Ministry of Health 2008.Rodriguez C, Tatham LM, Thun MJ, et al. \"Smoking and fatal prostate cancer in a large cohort of adult men.\" Am J Epidemiol 1997. 145: 466.Rohrmann S, Genkinger JM, Burke A, et al. \"Smoking and risk of fatal prostate cancer in a prospective US study.\" Urology 2007. 69(4): 721-725.Huncharek M, Haddock S, Reid R, Kupelnick B. \"Smoking as a Risk Factor for Prostate Cancer: A Meta-Analysis of 24 Prospective Cohort Studies.\" Am J Public Health 2010. 100: 693-701.Ferdinandusse S, Denis S, Ijist L, et al. \"Subcellular localization and physiological role of alpha-methylacyl-CoA racemase.\" J Lipid Res 2000. 41: 1890-6.Kotti TJ, Savolainen K, Helander HM, et al. \"In mouse alpha-methylacyl-CoA racemase, the same gene product is simultaneously located in mitochondria and peroxisomes.\" J Biol Chem 2000. 275: 20887-95.Rubin M, Zhou M, Dhanasekaran SM, et al. \"alpha-methylacyl Coenzyme A Racemase as a Tissue Biomarker for Prostate Cancer.\" JAMA 2002. 287(13): 1662-1670.Jiang Z, Woda BA, Rosk KL, et al. \"P504S: a new molecular marker for the detection of prostate carcinoma.\" Am J Surg Pathol 2001. 25: 1397-404.Suzue K, Montag AG, Tretiakova M, et al. \"Altered expression of alpha-methylacyl-coenzyme A racemase in prostatic adenocarcinoma following hormone therapy.\" Am J Clin Pathol 2005. 123: 553-6Jiang Z, Woda BA, Wu CL, et al. \"Discovery and clinical application of a novel prostate cancer marker: alpha-methylacyl CoA racemase (P504S).\" Am J Clin Pathol 2004. 122: 275-89.Rogers CG, Yan G, Zha S, et al. \"Prostate cancer detection on urinalysis for alpha methylacyl coenzyme a racemase protein.\" J Urol 2004. 172(4 Pt 1): 1501-3.Zielie PJ, Mobley JA, Ebb RG, et al. \"A Novel Diagnostic Test for Prostate Cancer Emerges from the Determination of a-methylacyl-coenzyme a racemase in Prostatic Secretions.\" J Urol 2004. 172: 1130-1133.Darson MF, Pacelli A, Roche P, et al. \"Human glandular kallikrein 2 expression in prostate adenocarcinoma and lymph node metastases.\" Urology 1998. 53: 939-44.Lintula S, Stenman J, Bjartell A, et al. \"Relative concentrations of hK2/PSA mRNA in benign and malignant prostatic tissue.\" Prostate 2005. 63: 324-9.Scorilas A, Plebani M, Mazza S, et al. \"Serum Human Glandular Kallikrein (hK2) and Insulin-Like Growth Factor I (IGF-I) Improve the Discrimination Between Prostate Cancer and Benign Prostatic Hyperplasia in Combination With Total and % Free PSA.\" Prostate 2003. 54: 220-229.Steuber T, Vickers AJ, Serio AM, et al. \"Comparison of Free and Total Forms of Serum Human Kallikrein 2 and Prostate-Specific Antigen for Prediction of Locally Advanced and Recurrent Prostate Cancer.\" Clin Chem 2007. 53(2): 233-240.Steuber T, O'Brien MF, Lilja H, et al. \"Serum Markers for Prostate Cancer: A Rational Approach to the Literature.\" Eur Urol 2008. 54: 31-40.Logothetis CJ and Lin S. \"Osteoblasts in Prostate Cancer Metastasis to Bone.\" Nat Rev Cancer 2005. 5(1): 21-28.Horowitz MC, Xi Y, Wilson K, et al.. \"Control of osteoclastogenesis and bone resorption by members of the TNF family of receptors and ligands.\" Cytokine and Growth Factor Rev 2001. 12: 9-18.Brown JM, Vessella RL, Kostenuik PJ, et al. \"Serum osteoprotegerin levels are increased in patients with advanced prostate cancer.\" Clin Cancer Res 2001. 7: 2977-83.Eaton CL, Wells JM, Holen I, et al. \"Serum osteoprotegerin (OPG) levels are associated with disease progression and response to androgen ablation in patients with prostate cancer.\" Prostate 2004. 59: 304-10.Jung K, Lein M, Von Hosslin K, et al. \"Osteoprotegerin in serum as a novel marker of bone metastatic spread in prostate cancer.\" Clin Chem 2001. 47: 2061-3.Lipton A, Ali SM, Leitzel K, et al. \"Serum osteoprotegerin levels in healthy controls and cancer patients.\" Clin Cancer Res 2002. 8: 2306-10.Hofbauer LC and Heufelder AE. \"The role of osteoprotegerin and receptor activator of nuclear factor kappaB ligand in the pathogenesis and treatment of rheumatoid arthritis.\" Arthritis Rheum 2001. 44: 253-9.Feuerherm AJ, Borset M, Seidel C, et al. \"Elevated levels of osteoprotegerin (OPG) and hepatocyte growth factor (HGF) in rheumatoid arthritis.\" Scand J Rheumatol 2001. 30: 229-34.Sommerfeld HJ, Meeker AK, Piatyszek MA, et al. \"Telomerase Activity: a prevalent marker of malignant human prostate tissue.\" Cancer Res 1996. 56: 218-22.Kallakury BV, Brien TP, Lowry CV, et al. \"Telomerase activity in human benign prostate tissue and prostatic adenocarcinomas.\" Diagn Mol Pathol 1997. 6: 192-8.Meid FH, Gygi CM, Leisinger H, et al. \"The use of telomerase activity for the detection of prostate cancer cells after prostatic massage.\" J Urol 2001. 165: 1802-1805.Vicentini C, Gravina GL, Angelucci A, et al. \"Detection of telomerase activity in prostate massage samples improves differentiating prostate cancer from benign prostatic hyperplasia.\" J Cancer Res Clin Oncol 2004. 130: 217-21.Botchkina GI, Kim RH, Botchkina IL, et al. \"Noninvasive detection of prostate cancer by quantitative analysis of telomerase activity.\" Clin Cancer Res 2005. 11: 3243-9.Schick F, Bongers H, Kurz S, et al. \"Localized proton MR spectroscopy of citrate in vitro and of the human prostate in vivo at 1.5T.\" Magn Reson Med 1993. 29(1): 38-43.Kurhanewicz J, Vigneron DB, Hricak H, et al. \"Three-dimensional H-1 MR spectroscopic imaging of the in situ human prostate with high (0.24-0.7cm3) spatial resolution.\" Radiology 1996. 198(3): 795-805.Kurhanewicz J, Vigneron DB, Hricak H, et al. \"Prostate Cancer: metabolic response to cryosurgery as detected with 3D H-1 MR spectroscopic imaging.\" Radiology 1996. 200(2): 489-96.Costello LC, Franklin RB, Narayan P, et al. \"Citrate in the diagnosis of prostate cancer.\" Prostate 1999. 38(3): 237-45.Cheng LL, Burns MA, Taylor JL, et al. \"Metabolic characterisation of human prostate cancer with tissue magnetic resonance spectroscopy.\" Cancer Res 2005. 65(8): 3030-4.Glunde K and Serkova NJ. \"Therapeutic targets and biomarkers identified in cancer choline phospholipid metabolism.\" Pharmacogenomics 2006. 7(7): 1109-23.Kurhanewicz J, Dahiya R, MacDonald JM, et al. \"Citrate alterations in primary and metastatic human prostatic adenocarcinomas: 1H magnetic resonance spectroscopy and biochemical study.\" Magn Reson Med 1993. 29(2): 149-57.Costello LC, Franklin RB, Feng P, et al. \"Mitochondrial function, zinc, and intermediary metabolism relationships in normal prostate and prostate cancer.\" Mitochondrion 2005. 5(3): 143-53.Teahan O, Bevan CL, Waxman J, Keun HC. (2010). \"Metabolic signatures of malignant progression in prostate epithelial cells.\" Int J Biochem Cell Biol.Swanson MG, Vigneron DB, Tabatabai ZL, et al. \"Proton HR-MAS spectroscopy and quantitative pathologic analysis of MRI/3D-MRSI-targeted postsurgical prostate tissues.\" Magn Reson Med 2003. 50(5): 944-54.Kurhanewicz J, Vigneron DB, Nelson SJ, et al. \"Three-dimensional magnetic resonance spectroscopic imaging of brain and prostate cancer.\" Neoplasia 2000. 2(1-2): 166-89.Swanson MG, Zektzer AS, Tabatabai ZL, et al. \"Quantitative analysis of prostate metabolites using 1H HR-MAS spectroscopy.\" Magn Reson Med 2006. 55(6): 1257-64.Griffin JL and Shockcor JP \"Metabolic profiles of cancer cells.\" Nat Rev Cancer 2004. 4(7): 551-61.Garcia-Segura JM, Sanchez Chapado M, Ibarburn C, et al. \"In vivo proton magnetic resonance spectroscopy of diseased prostate: spectroscopic features of malignant versus benign pathology.\" Magn Reson Imaging 1999. 17(5): 755-65.Browett P. Familial Cancer. MBCHB209B Cancer Lectures July 2010. Auckland, University of Auckland: Faculty of Medical and Health Sciences; School of Medicine; Dept. Molecular Medicine and Pathology.Pepe MS, Etzioni R, Feng Z, et al. \"Phases of Biomarker Development for Early Detection of Cancer.\" J Natl Cancer Inst 2001. 93(14): 1054-1061.Cancer Control Taskforce. The New Zealand Cancer Control Strategy: Action Plan 2005-2010. Wellington, Ministry of Health 2005.Ministry of Health. The Primary Healthcare Strategy. Wellington, Ministry of Health 2001.Ministry of Health. Initial Response to the New Zealand Cervical Cancer Audit. Wellington, Ministry of Health 2004.Chamberlain J. Breastscreen Aotearoa: An independent review. Wellington, Ministry of Health 2002.Smart RF and Tulloch DN. \"New Zealand should introduce population screening for prostate cancer using PSA testing.\" J Primary Health Care 2009. 1(4): 319-322.Smart RF. \"PSA testing and DRE, TRUS scanning with sector biopsy, improved histology, curative treatments, and active surveillance for prostate cancer: a success story for men\u2019s health.\" NZ Med J 2008. 121(1287).Lamb DS, Slaney D, Smart RF, et al. \"Prostate cancer: the new evidence base for diagnosis and treatment.\" Pathology 2007. 39(6): 537-544.Lamb DS and Delahunt B. \"Prostate cancer screening - finding the middle road forward.\" NZ Med J 2005. 118(1209): 1-3.Lamb DS, Denham JW, Delahunt B, et al. \"Prostate cancer screening in Australasia.\" Clin Oncol (R Coll Radiol) 2005. 17(4): 231-3.Vasen HFA, Mecklin J-P, Meera Khan P, Lynch HT. \"The International Collaborative Group on Hereditary Non-Polyposis Colorectal Cancer (IGC-HNPCC).\" Dis Colon Rectum 1991. 34(5): 424-5.McClellan J and King M-C. \"Genetic Heterogeneity in Human Disease.\" Cell 2010. 141(2): 210-217.Paynter NP, Chasman DI, Pare G, et al. \"Association between a literature-based genetic risk score and cardiovascular events in women.\" JAMA 2010. 303(7): 631-7.Manolio TA, Collins FS, Cox NJ, et al. \"Finding the missing heritability of complex diseases.\" Nature 2009. 461(7265): 747-53.Bratt O, Kristoffersson U, Lundgren R, Olsson H. \"Familial and Hereditary Prostate Cancer in Southern Sweden. A Population-based Case-Control Study.\" Eur J Cancer 1999. 35(2): 272-7.Dijkman GA and Debruyne FM. \"Epidemiology of prostate cancer.\" Eur Urol 1996. 30(3): 281-95.Gray M, Borman B, Crampton P, et al. \"Elevated serum prostate-specific antigen levels and public health implications in three New Zealand ethnic groups: European, Maori and Pacific Island men.\" NZ Med J 2005. 118(1209).Salmond C and Crampton P. Deprivation and Health. In Howden-Chapman P, Tobias M, ed. Social Inequalities in Health: New Zealand 1999. Wellington, Ministry of Health 2000.Sporle A, Pearce NE, Davis PB, et al. \"Social class mortality differences in Maori and non-Maori men aged 15-64 during the last two decades.\" NZ Med J 2002. 115(1150): 127-31.Lamb DS, Bupha-Intr O, Bethwaite P, et al. \"Prostate Cancer - Are Ethnic Minorities Disadvantaged?\" Anticancer Res 2008. 28: 3891-3896.Loeb S, Gashti SN, et al. \"Exclusion of inflammation in the differential diagnosis of an elevated prostate-specific antigen (PSA).\" Urol Oncol: Seminars and Original Investigations 2009. 27: 64-66.Oefelein MG, Smith N, Carter M, et al. \"The incidence of prostate cancer progression with undetectable serum prostate specific antigen in a series of 394 radical prostatectomies.\" J Urol 1995. 154: 2128-2131.Lee DK, Park JH, Kim JH, et al. \"Progression of Prostate Cancer Despite an Extremely Low Serum Level of Prostate-Specific Antigen.\" Korean J Urol 2010. 51(5): 358-61.Giovannini C, Chieco P, Bertaccini A, et al. \"Checkpoint effectors CDKN1A and Gadd45 corellate with oxidative DNA damage in human prostate carcinoma.\" Anticancer Res 2004. 24(6): 3955-3960.Kabacik S, Mackay A, Tamber N, et al. \"Gene expression following ionising radiation: Identification of biomarkers for dose estimation and prediction of individual response.\" Int J Radiat Biol 2010. [Epub ahead of print].Hanova M, Vodickova L, Vaclavikova R, et al. \"DNA damage, DNA repair rates and mRNA expression levels of cell cycle genes (TP53, p21(CDKN1A), BCL2 and BAX) with respect to occupational exposure to styrene.\" Carcinogenesis 2011. 32(1): 74-9.Talesa VN, Antognelli C, Del Buono C, et al. \"Diagnostic potential in prostate cancer of a panel of urinary tumour markers.\" Cancer Biomarkers 2009. 5(6): 241-245.Devaney J, Stirzaker C, Qu W, et al. \"Epigenetic Deregulation Across Chromosome 2q14.2 Differentiates Normal from Prostate Cancer and Provides a Regional Panel of Novel DNA Methylation Cancer Biomarkers.\" Cancer Epidemiol. Biomarkers Prev 2011. 20(1): 148-159.Clines GA, Mohammad KS, Bao Y, et al. \"Dickkopf homolog 1 mediates endothelin-1-stimulated new bone formation.\" Mol Endocrinol 2007. 21: 486-498.Li ZG, Yang J, Vasquez ES, et al. \"Low-density lipoprotein receptor-related protein 5 (LRP5) mediates the prostate cancer-induced formation of new bone.\" Oncogene 2008. 27: 596-603.Jentzmik F, Stephan C, Lein M, et al. \"Sarcosine in prostate cancer tissue is not a different metabolite for prostate cancer aggressiveness and biochemical progression.\" J Urol 2011. 185(2): 706-11.Struys EA, Heijboer AC, van Moorselaar J, et al. \"Serum sarcosine is not a marker for prostate cancer.\" Ann Clin Biochem 2010. 47: 282.Sreekumar A, Poisson LM, Rajendiran TM, et al. \"Metabolomic profiles delineate potential role for sarcosine in prostate cancer progression.\" Nature 2009. 457(7231): 910-4.Cao DL, Ye DW, Zhang HL, et al. \"A multiplex model of combining gene-based, protein-based, and metabolite-based with positive and negative markers in urine for the early diagnosis of prostate cancer.\" Prostate 2010. [Epub ahead of print].Bill-Axelson A, Holmberg L, Filen F, et al. \u201cRadical Prostatectomy Versus Watchful Waiting in Localised Prostate Cancer: the Scandinavian Prostate Cancer Group-4 Randomized Trial.\u201d J Natl Cancer Inst 2008; 100: 1144 \u2013 1154.Hugosson J, Carlsson S, Aus G, et al. \u201cMortality results from the Goteborg randomised population-based prostate-cancer screening trial.\u201d Lancet Oncol 2010; 11(8): 725-732.Schroder FH, Hugosson J, Roobol MJ, et al. \u201cScreening and prostate-cancer related mortality in a randomized European study.\u201d N Engl Med J 2009; 360(13): 1320-8.