Sunteți pe pagina 1din 4

2 EUROPEAN UROLOGY SUPPLEMEN TS XXX (2017) XXX–XXX

of biologically significant PCA would help further refine the MYC oncogene, a recognized player in PCA pathogenesis
current clinical and pathologic input based management [31] and SNP variants located on chromosomes 19 q13 that
algorithms. Such biomarkers will enhance confidence in harbor kallikrines KLK2 and KLK3 (PSA) genes are of
accurately assigning patients with insignificant PCA catego- particular interest [35,36]. Although each susceptibility
rized as very low risk in National Comprehensive Cancer SNP allele individually carries only a small risk, multiplica-
Network guidelines (http://www.nccn.org/professionals/ tively, a subject SNP profile can be deduced from risk
physician_gls/f_guidelines.asp) to active surveillance ap- algorithms models that will identify individuals in the
proach that is coupled with active monitoring by molecular upper 1% risk tier with approximately 5-fold the risk of the
imaging and ongoing look out for genomic signatures of general population [37,38]. It is such an approach that have
biologic progression using tissue samples. the potential to refine the best groups of men to be targeted
for screening and prevention strategies and help address
2. Pathogenesis of prostate cancer: genetic risk and the current concerns of overdiagnosis and overtreatment of
environmental factors PCA [8,9].
The detection of the more rare but strongly penetrant
The interplay between genetic determinants of risk and germline variants (> 5% frequencies) requires exhaustive
environmental factors is responsible for the long-observed case/control direct sequencing that was only achievable
variation in PCA incidence among geographic populations. with more recently propagated massively parallel next
The higher incidence of PCA in African-Americans compared generation sequencing technologies. Given their rarity, such
with Asian-Americans is thought to be only partially driven variants account for only a minute fraction of PCA incidence
by genetic predisposition [11]—the fact that such risk is but importantly impart a high risk in carrier individuals for
modulated upon migration in a given ethnic group points to early onset (5—7-fold) and at times more aggressive PCA.
environmental and lifestyle factors as additional contribut- Most established among these are germline mutations in
ing determinants of risk [4,5,12,13]. the BRCA 2 tumor suppressor gene [39] and HOXB13 (G84E)
[40,41]. A 5% germline carrier frequency for HOXB13 (G84E)
2.1. Inherited genetic risk factors mutation was shown in families of PCA of mostly European
descent [41]. Men with the BRCA2 germline mutation were
Twins and family pedigree studies have long supported found to be at 5-fold the risk for PCA (7-fold the risk of early
genetic predisposition as a risk factor for PCA disease) in the Breast Cancer Linkage Consortium study
[11,14,15]. Men with a first degree relative affected by [42]. The evidence for BRCA1 [43] and other DNA repair
the disease are at twice the risk for developing PCA. The risk genes such as PALB2, CHECK2, BRIP1, and NBS1 remains less
is even higher (> 4-fold) if a relative was diagnosed with robust [44–47].
PCA at a younger age (< 60 yr) [16,17]. Finally, molecular risk studies support increased sus-
PCA is now recognized as one of the most heritable ceptibility for PCA in Lynch syndrome. A recent meta-
cancer types driven by numerous inherited germline analysis of 12 risk studies showed a 2.28-fold (95%
genetic variants of risk ranging from those that are more confidence interval [CI]: 1.37–3.19) increased risk of PCA
common with a weak risk effect to the rare variants that are for all men from mismatch repair genes mutation-carrying
associated with a stronger risk for PCA. While earlier linkage families. In another study, the relative risk was greatest for
analysis studies pointed to numerous chromosomal loci of MSH2 carriers (5.8, 95% CI: 2.6–20.9) where PCA was the
association, such claims have failed to be consistently first or only diagnosed tumor in 37% of carriers [48–50].
validated [16,18–25]. The same is true for the initial Several multinational consortia (PCA Association Group
suggestion that inflammatory and infection response gene to Investigate Cancer Associated Alterations in the Genome,
loci (ELAC2, RANSEL, and MSR1) were associated with higher http://practical.ccge.medschl.cam.ac.uk/; International
risk of PCA [26,27]. In the largest linkage study performed Consortium for PCA Genetics, http://www.icpcg.org/
by the international consortium of PCA genetics only one ?q=content/about-icpcg; Elucidating Loci Involved in PCA
locus (22q) stood out [28]. Subsequent studies also pointed Susceptibility, http://epi.grants.cancer.gov/gameon/
to 8q24 [29–31] as a region harboring genetic risk variants. personnel.html#ellipse) are investigating the important
In the last decade, the application of Genome Wide clinical impact of genetic susceptibility to PCA in order to
Association Studies that enables assessment of millions of address the potential screening, risk management guide-
single nucleotide polymorphisms (SNPs) in a given indi- lines and functional and treatment implications of the
vidual for disease risk association to PCA [32] has allowed growing list of identified germline genetic variants.
for the detection of far more common germline genetic
variants with only low-to-moderate penetrance. To date, 2.2. Environmental risk factors
over 280 SNPs associations with PCA risk have been
established by 30 such studies (A Catalog of Published A body of evidence points to glandular epithelial cell injury
Genome-Wide Association Studies (https://www.ebi.ac.uk/ resulting from chronic exposure to dietary carcinogens,
gwas/search?query=Prostate%20cancer#association). Col- estrogens, or oxidants as a trigger for a chronic inflamma-
lectively, these SNPs may account for up to one-third of tory milieu that set the stage for cancer development
PCA familial risk. Regions such 8q24 region [33,34] [12,51–55]. Epidemiologic and animal model studies
containing SNPs exerting a modifier effect on neighboring [56,57] strongly support the dietary intake of charred red

Please
Pleasecite
citethis
thisarticle
articleininpress
pressas:
as:Netto
NettoGJ,
GJ,etetal.
al.Prostate
ProstateCancer:
Cancer:An
AnUpdate
Updateon
onMolecular
MolecularPathology
Pathologywith
withClinical
ClinicalImplications.
Implications.
Eur
EurUrol
UrolSuppl
Suppl(2017),
(2017),https://doi.org/10.1016/j.eursup.2017.10.001
https://doi.org/10.1016/j.eursup.2017.10.001
EUROPEAN UROLOGY SUPPLEMEN TS XXX (2017) XXX–XXX 3

Fig. 1 – Etiological factors implicated in prostate cancer development. Chronic inflammation triggered by environmental and lifestyle exposures leads
to persistent prostate epithelial cell damage. Inherited genetic predisposition also plays a determining factor in promoting oncogenesis.
PIA = proliferative inflammatory atrophy; PIN = prostatic intraepithelial neoplasia; SNP = single-nucleotide polymorphism; STD = sexually transmitted
disease.

meats and animal fats as risk factors for PCA through extensive list of biomarkers has been evaluated in the last
formation of heterocyclic aromatic amine (eg, 2-amino-1- 2 decades for their potential role in predicting disease
methyl-6-phenylimidazopyrine) and polycyclic aromatic outcome and as therapeutic targets [4,5,12,72–75]. They
hydrocarbon carcinogens [58–60]. Likewise, animal data include markers of proliferation index (ki67) [76–79],
link estrogen exposure to prostate epithelial cell damage tumor suppression genes (eg, p53, p21, p27, NKX3.1, PTEN,
and inflammation potentially through induction of autoim- retinoblastoma gene Rb), oncogenes (eg, Bcl2, c-myc, EZH2,
munity [61,62]. Less robust evidence implicates sexually and HER2/neu), adhesion molecules (CD44, E-Cadherin),
transmitted infectious agents (eg, trichomonas, chlamydia, PI3K/akt/mTOR pathway members [80,81], apoptosis reg-
and gonorrhea) as potential initiators of chronic inflamma- ulators (eg, survivin and transforming growth factor b1),
tion of the prostate (Fig. 1) [63–66]. Epithelial damage and androgen receptor status, and prostate tissue lineage
ensuing inflammation is the common pathogenic link specific markers (PSA, prostatic-specific acid phosphatase,
between all the above environmental carcinogens and and prostate-specific membrane antigen).
PCA development. The persistent oxidative stress exerted The role of p53 expression in predicting prognosis in PCA
upon prostatic epithelial cells is counteracted by a genome has been extensively studied. Brewster et al [82] found p53
damage defense and cell survival response through expression and Gleason score in needle biopsy to be
expression of a and p class glutathione S-transferases, independent predictors of biochemical recurrence follow-
cyclooxygenase-2, and others mediators [54,67–69]. Subse- ing radical prostatectomy (RP) [82]. Retrospective studies
quently, epigenetic silencing of hundreds of genes including evaluating prostatectomy specimens also found p53 to be of
the crucial caretaker gene GSTP1 take hold in the damaged prognostic significance independent of grade, stage, margin
epithelium and persists throughout subsequent cancer status [83–85]. The later has been further illustrated in
progression phases. Such changes are detectable in the more recent genome wide studies [86]. A prognostic role of
earliest histologic manifestation of injury response, namely p27 in predicting progression following prostatectomy have
proliferative inflammatory atrophy that seems to share also been shown while less robust evidence exists for the
many of the somatic genetic and epigenetic alterations that prognostic role of p21 [87] and NKX3.1 [88,89]. Several
are exhibited by prostatic intraepithelial neoplasm and PCA studies have supported a prognostic role for Bcl2 [82,84] and
[70–72]. myc oncogenes [90] as potential adjuncts to histologic
prognostic parameters. With the exception of tumor
3. Somatic molecular alterations in PCA suppressor gene PTEN and myc oncogene, none of the
above individual biomarkers have transitioned into clinical
3.1. Genetic alterations use.
Assessment of loss of PTEN alone or in combination with
PCA is a complex and heterogeneous disease. Prostate ERG fusion have recently gained significant momentum
tumor development and progression involves alterations in in routine practice as an ancillary test to help better
numerous genetic pathways. As our understanding of these delineate indolent lower grade disease that can be managed
pathways and key driver molecular alterations evolved, an by active surveillance. The following sections further

Please
Pleasecite
citethis
thisarticle
articleininpress
pressas:
as:Netto
NettoGJ,
GJ,etetal.
al.Prostate
ProstateCancer:
Cancer:An
AnUpdate
Updateon
onMolecular
MolecularPathology
Pathologywith
withClinical
ClinicalImplications.
Implications.
Eur
EurUrol
UrolSuppl
Suppl(2017),
(2017),https://doi.org/10.1016/j.eursup.2017.10.001
https://doi.org/10.1016/j.eursup.2017.10.001
4 EUROPEAN UROLOGY SUPPLEMEN TS XXX (2017) XXX–XXX

expand on the important role of ETS gene fusions and showed absence of PTEN gene deletion and 66% of cases
mammalian target of rapamycin (mTOR) pathway in PCA. with PTEN protein loss by IHC showed PTEN gene deletion
by FISH. Among cases with intact PTEN by IHC, homozygous
3.2. ETS gene fusions (p = 0.04) but not heterozygous (p = 0.10) PTEN gene
deletion was weakly associated with reduced recurrence-
In 2005, Tomlins et al [91,92] identified a recurrent free survival. The data support the utility of PTEN IHC and
chromosomal rearrangement in over one-half of their PTEN FISH as complementary screening tools for PTEN loss
analyzed PCA cases. The rearrangements lead predomi- in PCA [108].
nantly to the fusion of the androgen-responsive promoter The same group has previously shown IHC screening for
elements of the TMPRSS2 gene (21q22) to one of three PTEN loss to be particularly useful to identify cases with
members of the ETS transcription factors family members heterogeneous PTEN gene deletion especially when present
ERG, ETV1, and ETV4 located at chromosomes 21q22, 7p21, in a subset of tumor glands. Mutations, small insertions, or
and 17q21, respectively. Although the prognostic role of deletions, and/or epigenetic or microRNA-mediated mech-
assessing TMPRSS2-ETS rearrangements in PCA tissue anisms may lead to PTEN protein loss in tumors with
samples, as a standalone marker, was subsequently not normal or hemizygous PTEN gene copy number [109].
substantiated in well-designed large cohort studies [93,94], In a large nested case control tissue microarray, we
the discovery had great implications in terms of furthering initially demonstrated loss of PTEN immunoexpression to
our understanding of PCA pathogenesis and provided a new be an independent predictor of biochemical recurrence
marker for molecular diagnosis in PCA [95–99]. Further- (BCR) following RP [110]. Our group subsequently demon-
more, as shown below, recent genome wide studies have strated [111] the prognostic role of assessing PTEN
repointed to the prognostic significance of TMPRSS2-ERG alteration in a surgical cohort of high-risk PCA patients
fusion status as part of genomic signatures that could where its loss was also predictive of decreased time to
stratify disease aggressiveness. The potential diagnostic and metastatic spread. Assessing PTEN in a large cohort of PCA
prognostic role of detecting TMPRSS2-ERG fusion in post- (over 4750), Krohn et al [112] demonstrated that biallelic
prostate massage urine samples is also very promising PTEN inactivation, by either homozygous deletion or
[100–102]. deletion of one allele and mutation of the other, occurs in
The development of commercial anti-ERG monoclonal most PTEN-defective cancers and characterizes a particu-
antibodies facilitated the use immunohistochemistry (IHC) larly aggressive subset of metastatic and hormone-refrac-
for the evaluation of ERG protein expression as a simpler, less tory PCA. PTEN deletions were present in 20% of PCA (8%
costly, surrogate approach to fluorescent in situ hybridiza- heterozygous and 12% homozygous). PTEN deletions were
tion (FISH) for detection of TMPRSS2-ERG fusion. Several associated with early BCR, advanced tumor stage, high
groups were able to demonstrate a strong correlation Gleason grade, presence of lymph node metastasis,
between ERG overexpression by IHC and ERG fusion status hormone-refractory disease, presence of ERG gene fusion,
with over 86% sensitivity and specificity rates [103,104]. and nuclear p53 accumulation. The prognostic impact of
In certain settings, ERG immunostaining could be of PTEN deletion was seen in both ERG fusion-positive and ERG
utility in establishing the diagnosis of carcinoma when a fusion-negative tumors [112] (Fig. 3). An evaluation of PTEN
pathologist is faced with a small focus of atypical glands on and ERG in 1044 incident PCA cases in the Health
needle biopsy. In such cases, positive ERG staining in Professionals Follow-up Study and Physicians’ Health Study
combination with other immunomarkers such as racemase showed PTEN loss to be associated with lethal progression
(AMACR), p63, and PTEN could be very helpful. As discussed of disease; however, this was true only in tumors that were
below, a prognostic role for ERG IHC expression has also also negative for ERG fusion [113].
recently evolved in combination with PTEN. As mentioned above, assessment of PTEN loss is gaining
an important role as an adjunct biomarker to stratify
3.3. PTEN loss patients with pathologically low risk disease (Grade Group
1 and Grade group 2; Gleason grade 3 + 3 = 6 and 3 + 4 = 7,
The PI3K/mTOR pathway plays an important role in cell respectively) that are in consideration for active surveil-
growth, proliferation, and oncogenesis in PCA [105–107]. lance. Loss of PTEN on needle biopsies in such patients
PTEN is the master negative regulator of the pathway. would argue for a definitive therapy over active surveil-
Several well-designed retrospective studies have revealed lance. This is based on multiple studies showing PTEN
that loss of PTEN tumor suppressor gene activity, and the loss on needle biopsy to be associated with a higher
ensuing mTOR pathway activation, is associated with poor likelihood of tumor upgrade and the presence of nonorgan
prognosis in PCA. confined disease on RP [114,115]. In one such study, Guedes
PTEN loss in PCA tissue samples can be evaluated either et al [116] showed PTEN loss in a Gleason score
by FISH to detect deletion of PTEN gene allele(s) in tumor 3 + 4 = 7 biopsy to be independently associated with an
cells or more recently by IHC (Fig. 2). Lotan et al [108] increased risk of nonorgan confined disease at prostatec-
compared PTEN assessment by IHC and FISH in the largest tomy (52% vs 27%).
existing RP cohort with clinical follow up (13 665 patients). The mTOR pathway is also a potential target of therapy in
High concordance between IHC and FISH was documented. PCA. Several rapamycin analogs have been assessed as
Ninety-three percent of tumors with intact PTEN IHC potential therapeutic agents for PCA [107,117,118].

Please
Pleasecite
citethis
thisarticle
articleininpress
pressas:
as:Netto
NettoGJ,
GJ,etetal.
al.Prostate
ProstateCancer:
Cancer:An
AnUpdate
Updateon
onMolecular
MolecularPathology
Pathologywith
withClinical
ClinicalImplications.
Implications.
Eur
EurUrol
UrolSuppl
Suppl(2017),
(2017),https://doi.org/10.1016/j.eursup.2017.10.001
https://doi.org/10.1016/j.eursup.2017.10.001
EUROPEAN UROLOGY SUPPLEMEN TS XXX (2017) XXX–XXX 5

Fig. 2 – Phosphatase and tensin homolog (PTEN) immunohistochemistry in prostate cancer. (A) PTEN intact in tumor cells ‘‘T’’, note equivalent staining
in adjacent benign glands labeled ‘‘B’’. Lower frame: higher magnification inset shows positive ‘‘P’’ staining tumor glands. (B) PTEN homogeneous loss
in tumor glands ‘‘T’’, with intact staining in nearby benign glands ‘‘B’’ and stroma. Lower frame: higher magnification inset shows negative ‘‘N’’ staining
tumor glands. (C) PTEN heterogeneous loss, with staining loss in some but not all tumor cells ‘‘T’’. Higher magnification inset below shows positive
‘‘P’’staining and negative ‘‘N’’ staining tumor glands. (D) PTEN uninterpretable staining. PTEN stain is weaker but not lost in tumor glands and absence
of background benign glands for comparison makes it difficult to interpret. Lower frame: higher magnification inset shown below. Adapted with
permission from Lotan et al [108].

3.4. Genomic classifications of PCA with a more favorable outcome. The findings were validated
in an independent clinical cohort at Memorial Sloan-
In one of the earliest genomic studies, using gene expression Kettering Cancer Center.
profiling (complementary DNA microarrays), Lapointe et al In one of the first studies on whole genome sequencing
[2] were able to identify three subclasses of PCA based on (WGS) of PCA, Berger et al [121] analyzed the entire genome
their distinct patterns of gene expression. The authors of seven PCA samples obtained from warm autopsy
subsequently used array-based comparative genomic hy- procedures. The daunting effort brought to light the
bridization [119] to identify recurrent copy number genetic occurrence of complex chains of balanced (ie, copy-neutral)
alterations that corresponded to three prognostically distinct rearrangements within or adjacent to known cancer genes
groups: (1) deletions at 5q21 and 6q15 group associated with that on average include 90 rearrangements per genome. This
favorable outcome group, (2) a 8p21 (NKX3-1) and 21q22 process of complex rearrangements, termed ‘‘chromoplexy,’’
(resulting in TMPRSS2-ERG fusion) deletion group, and (3) was further confirmed in a subsequent larger WGS study
8q24 (MYC) and 16p13 gains, and loss at 10q23 (PTEN) and performed by the same investigators on 57 PCA samples
16q23 groups correlating with metastatic disease and [122]. Distinctive patterns of chromoplexy appeared in (ETS
neg
aggressive outcome. More recent integrated genome-wide CHD1del) tumors compared with (ETS pos CHD1wt) PCA
assessment of PCA has further defined various chromosomal (Fig. 4). Tumors with a deletion of CHD1 demonstrated an
rearrangements and copy number gains and losses, including excess of intrachromosomal chained rearrangements and
ETS gene family fusions, PTEN loss, and androgen receptor gene deletions clustered in one or two chromosome(s) with
(AR) amplification, that drive PCA development and progres- breakpoints concentrated in GC-poor nonexpressed DNA
sion to lethal, metastatic castration-resistant PCA (CRPC). sites. In contrast, in ETS pos tumors, many single chromoplexy
Taylor et al [120], using DNA copy number, messenger events joined DNA from dispersed regions of six or more
RNA (mRNA) expression, and focused exon resequencing chromosomes. In the latter group, the chain rearrangements
were able to stratify clusters of low- and high-risk disease primarily involved breakpoints enriched near open (actively
beyond the ability of risk stratification by traditional transcribed) chromatin, AR, and ERG DNA binding sites. The
pathologic prognostic parameters such as Gleason grade. latter suggests a link between chromatin and transcriptional
Six clusters of PCA are identified by unsupervised hierar- regulation and the genesis of genomic aberrations. Among
chical clustering with distinct risk for BCR. Markert et al [86] the genes that were most frequently disrupted by the
evaluated mRNA microarray signature profiles in a large rearrangements are adhesion molecule gene CADM2 and not
Swedish watchful-waiting cohort of PCA patients and found surprisingly PTEN and MAGI2, a gene that encodes for the
mRNA stem-like signatures in combination with p53 and PTEN interacting protein.
PTEN inactivation to be associated with very poor survival The sequence of oncogenic events in PCA progression has
outcome. TMPRSS2-ERG fusion group had an intermediate been further delineated with WGS studies. Genome-wide
survival outcome compared with the remaining groups germline SNPs coverage permitted the identification of DNA

Please
Pleasecite
citethis
thisarticle
articleininpress
pressas:
as:Netto
NettoGJ,
GJ,etetal.
al.Prostate
ProstateCancer:
Cancer:An
AnUpdate
Updateon
onMolecular
MolecularPathology
Pathologywith
withClinical
ClinicalImplications.
Implications.
Eur
EurUrol
UrolSuppl
Suppl(2017),
(2017),https://doi.org/10.1016/j.eursup.2017.10.001
https://doi.org/10.1016/j.eursup.2017.10.001

S-ar putea să vă placă și