Open Access

Decoding the heterogeneous landscape in the development prostate cancer (Review)

  • Authors:
    • Yenifer Yamile Segura-Moreno
    • María Carolina Sanabria-Salas
    • Rodolfo Varela
    • Jorge Andrés Mesa
    • Martha Lucia Serrano
  • View Affiliations

  • Published online on: March 15, 2021     https://doi.org/10.3892/ol.2021.12637
  • Article Number: 376
  • Copyright: © Segura-Moreno et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Prostate cancer (PCa) is characterized as being histologically and molecularly heterogeneous; however, this is not only incorrect among individuals, but also at the multiple foci level, which originates in the prostate gland itself. The reasons for such heterogeneity have not been fully elucidated; however, understanding these may be crucial in determining the course of the disease. PCa is characterized by a complex network of chromosomal rearrangements, which simultaneously deregulate multiple genes; this could explain the appearance of exclusive events associated with molecular subtypes, which have been extensively investigated to establish clinical management and the development of therapies targeted to this type of cancer. From a clinical aspect, the prognosis of the patient has focused on the characteristics of the index lesion (the largest focus in PCa); however, a significant percentage of patients (11%) also exhibit an aggressive secondary foci, which may determine the prognosis of the disease, and could be the determining factor of why, in different studies, the classification of the subtypes does not have an association with prognosis. Due to the aforementioned reasons, the analysis of molecular subtypes in several foci, from the same individual could assist in determining the association between clinical evolution and management of patients with PCa. Castration‑resistant PCa (CRPC) has the worst prognosis and develops following androgen ablation therapy. Currently, there are two models to explain the development of CRPC: i) the selection model and ii) the adaptation model; both of which, have been found to include alterations described in the molecular subtypes, such as Enhancer of zeste 2 polycomb repressive complex 2 subunit overexpression, isocitrate dehydrogenase (NAPD+)1 and forkhead box A1 mutations, suggesting that the presence of specific molecular alterations could predict the development of CRPC. This type of analysis could lead to a biological understanding of PCa, to develop personalized medicine strategies, which could improve the response to treatment thus, avoiding the development of resistance. Therefore, the present review discusses the primary molecular factors, to which variable heterogeneity in PCa progress has been attributed.

Introduction

According to the report by the International Agency for Research on Cancer of 2018 (1), prostate cancer (PCa) has the second highest estimated age-standardized incidence rate worldwide (29.3 per 100,000) and is the sixth cause of cancer-associated death in men (7.6 per 100,000). The problem with the management of PCa is due to the difficulty in stratifying between indolent and aggressive cases. Although <5% of patients exhibit advanced disease, up to 40% of patients will eventually develop metastatic disease despite local therapy (2,3). Other patients with PCa undergo hormonal therapy treatment, radical prostatectomy (RP) or radiotherapy, so, numerous cases of PCa only require expectant management; thus, in these patients, an overtreatment may result in significant morbidity.

High Grade Prostatic Intraepithelial Neoplasia (HGPIN) is a prostate preneoplastic lesion, which may develop towards an invasive PCa (25 to 30%) during a process, which may take over 10 years (4), according to a study conducted at Johns Hopkins University School of Medicine, in patients of Baltimore (Maryland, United States of America). Both, HGPIN and PCa are multifocal, and HGPIN foci coexist in adjacent areas of PCa sharing chromosome deletions and interstitial translocations that originate the TMPRSS2-ERG fusion gene and genetic alterations, such as hypermethylation of the π-class glutathione S-transferase (GSTP1) promoter, which suggests a common origin (5). The heterogeneous and multifocality nature of the disease makes it difficult to understand prostate carcinogenesis (6). Currently, there is no adequate method to differentiate patients with poor prognosis of PCa from those with indolent disease, who should only have a controlled follow-up. The primary method of determining the suitable treatment option for a patient with PCa is based on the Gleason classification (5), an assessment of its morphological heterogeneity, which is associated with prognosis. Pathologists can classify each focus of PCa using Gleason patterns (GP) ranging from 1 to 5, and assigning a Gleason score (GS); or using the updated Gleason grade group, which includes the two most representative GPs in the tumor (7,8). Despite the association between Gleason classification and tumor behavior (the degree of differentiation of the neoplastic cells) the association between morphological heterogeneity and molecular heterogeneity has not been elucidated (9).

Molecular studies of PCa have revealed numerous recurrent DNA alterations associated with deregulating genes involved in the development of the prostate, such as the deletions and interstitial translocations that originate the TMPRSS2-ERG fusion gene, chromatin modification, cell cycle regulation and androgen signaling (10,11). Over the last decade (12), the investigation into PCa has focused on identifying the exclusive molecular events in the development of PCa, which could represent early and divergent events and could direct the course of the disease (13); thus, it is crucial to elucidate the carcinogenesis of PCa and utilize the information in the treatment of patients.

The present review will describe an updated review of intratumoral heterogeneity in multifocal PCa, to understand the carcinogenic process and its implication in the management of the disease; as the vast majority of molecular studies in PCa performed are single focused, and do not take into account molecular heterogeneity, which could contribute to limiting the use of molecular subtypes in the prognosis and treatment of the disease.

Molecular heterogeneity of PCa

The origin of PCa could be defined by the occurrence of chromosomal rearrangements that simultaneously, and in a coordinated manner, cause the inactivation of tumor suppressor genes (TSG) and the creation of oncogenic fusions, which would support a model of punctuated development in PCa (11,14). This could in turn be associated with the appearance of canonical alterations, and with the molecular subtypes involved in a broad genomic and transcriptomic diversity within and among intraprostatic PCa foci. Several studies have shown potential for their utilization as prognostic biomarker signatures (1517). Recently published data from The Cancer Genome Atlas (TCGA) (17) supports the division of the major molecular subclasses of localized PCa into erythroblast transformation specific (ETS)-rearrangement PCa [PCa with rearrangements and overexpression of ETS transcription factor ERG (ERG), ETS variant transcription factor (ETV)-1, −4, or other ETS family transcription factors], SPOP-mutated and CHD1-deleted [speckle type BTB/POZ protein (SPOP)/chromodomain helicase DNA binding protein 1] altered cancers (17), and several smaller categories, such as FOXA1 or IDH1deletion, which have been described in Table I. The use of molecular classifiers to personalize treatment shows promise; however, it is still in its infancy and additional validation and optimization are required to ensure it can be used in a clinical setting.

Table I.

Prostate cancer molecular subtypes, prognosis significance and target therapy.

Table I.

Prostate cancer molecular subtypes, prognosis significance and target therapy.

A, ETS(+) subtypes (50–70% prevalence in PCa)

Molecular subtype (%prevalence in PCa)Characterization of the subtypePrognosisTargets for treatments
ERG (46)Overexpression of ERG leads to overexpressionMetastatic phenotype due to anDirected to overcome epigenetic silencing
ETV1 (8)of EZH2, which facilitates formation ofincrease in cellular migration,caused by EZH2:
ETV4 (4)heterochromatin and the decreased expression ofdecrease in anoikis and-DZNep: A global histone methylation inhibitor that
FLI1 (1)NKX3-1 (28).CRPC (24,28).reactivates developmental genes not silenced by
PTENMut is frequently found, and has beenPTENMut has been associated withDNA methylation, in in vitro assays (32).
associated with subsequent activation of Akt,a higher GS and a lower disease--HDAC and DNMT inhibition in in vitro assays (33).
which leads to upregulation of AR and ERGfree survival rate (30,31).Inhibitors of EZH2:
signaling in TMPRSS2-ERG (Fig. 2) (29). -GSK 126: In vitro, overcomes resistance to enzalutamide (antagonist of AR) in CRPC (34).
-CPI-1205: Combined with enzalutamide/abiraterone (inhibitor of androgen biosynthesis) is currently being tested for patients with CRPC (35).
Directed to inhibit ERG function:
-ERG direct inhibition is difficult.
-Blocking function of ERG regulating co-factors, such as PARP1, is more feasible in pre-clinical assays in vitro and in vivo (2527).

B, ETS(−) subtypes (50–30% prevalence in PCa)

Molecular subtype (% prevalence in PCa)Characterization of the subtype PrognosisTargets for treatments

Highly expressed EZH2Overexpression of EZH2 can be caused by MYCSimilar to ETS(+), but with worseThe same that for ETS(+), but without ERG inhibition.
but independent of ERGoverexpression, miR-26a, miR-26b and miR-101prognosis (39).In vitro assays with aristeromycin and DZNeP cause
expression (31)loss (3638). growth inhibition of PCa via induction of mir-26a (40)
Inhibitor of MYC (against MYC-driven PCa including CRPC):
-AZD1208 (oral PIM1 kinase inhibitor): Effective at downregulating MYC activity, inhibiting the growth of MYC-driven PCa, inhibits tumorigenesis in tissue recombinants, Myc-PCa, and human PCa xenograft models (41).
-CX-6258 (pan-PIM kinase inhibitor) with CX-5461 (inhibitor of RNA polymerase I): Preclinical efficacy of therapies that combine to target MYC-directed signaling to the ribosome (42).
SPOPMut/-SPOP is a TSG, which codifies for a protein-Less adverse pathologic featuresDirected to SPOP's substrates:
SPINK-1 overexpressioninvolved in both proteasomal degradation andand favorable prognosis (47).-Lestaurtinib: Inhibitor for PRK-1 (serine/threonine
(1115)DNA repair (13).-The high expression of SPINK-1kinase of the PKC family). PRK-1 was shown to play
-Increase of SPOP substrate levels (AR, ERG,could be associated with high GSan important role in the AR complex. Pre-clinical
DAXX, DEK and SRC-3) and the NHEJ pathway (43).and a possible biochemical ormodel in vitro of treatment directed to AR and
-High DNA methylation levels have been associatedclinical recurrence (48,49).SRC-3 (51,52).
with the presence of CHD1 mutations and-SPINK-1 induces EMT, which-Abiraterone (inhibitor of androgen biosynthesis):
MAP3K7 (44,45).increases migratory and invasiveCRPCs with SPOPMut and CHD1 loss appear highly
-SPINK-1 overexpression could act as a growthcapacity in vitro (46).sensitive (53).
factor, stimulating signaling through Androgen-deprivation therapyMab anti-SPINK-1 (54).
EGFR/MAPK (46).induce SPINK1 upregulation associates with neuroendocrine prostate cancer phenotype in mice and patients (50).
FOXA1Mut (3)-FOXA1 is important to differentiate epithelial-It promotes progression of the cell-FOXA1 downregulation induces TGF-β signaling,
cells in the prostate; it binds the enhancer regionscycle in CPRC in pre-clinicalEMT, and cell motility, which is blocked by the TGF-β
in the genome, increases local chromatinassays both in vitro andreceptor I inhibitor, LY2157299. Combination
accessibility, and facilitates the recruitment of AR.in vivo (17,58,59).LY2157299 treatment sensitized PCa cells to
However, due to its dual roles as re-programming-Mutations in FOXA1 Wing2enzalutamide, leading to synergistic effects in inhibiting
factor of AR, as well as functioning as adomains (50% of all mutations)cell invasion in vitro, and xenograft CRPC tumor
collaborating factor, FOXA1 has been shown towere detected in PCa at all stages,growth and metastasis in vivo (58).
exhibit both oncogenic and tumor suppressivewhereas mutations at the highly-In vitro assays show that PARP-2 inhibitors suppress
roles in prostate cancer (55).conserved DNA-contact residueAR signaling and PCa growth through disruption
-FOXA1 mutations have been associated with theR219 (~5%) are enriched inof FOXA1 function (59).
SPOPMut/SPINK-1 subtype (13). mRNA, SCNAmetastatic tumors with
and methylation profiles of this subtype were similar to SPOP (17).neuroendocrine histology (60).
IDH1Mut (1)-IDH1Mut produces R-2-HG, which leads to-Due to its low frequency, there-Use of blocking agents of D-2-hydroxyglutarate and
hypermethylation of DNA by the inhibition ofare no significant conclusionshypomethylating agents in pre-clinical in vitro
members of the α-KG-dependent dioxygenaseon its prognosis; however,assays (62).
protein family. It causes a shift towardsIDH1 mutation is rare in-Knockdown of IDH1 decreased cancer cell
glycolysis and angiogenesis, as an adaptation tolocated PCa (17,61).proliferation and blocked the AR-mediated induction in
hypoxia, and it changes the cellular redox environment by altering the ratio of NADPH to NADP+ (60). IDH metabolic reprogramming in pre-clinical assays in vitro (62).
Biallelic loss of-Characterized by tandem focal duplications,-Associated with CPRC (63).-Immunotherapy by inhibition of the immune control
CDK12 (6.9)gene fusions and a marked differential expression of genes. High load of neoantigens induced by fusion and increased infiltration of tumor T cells by clonal expansion (63). checkpoint (anti-PD1 inhibitor) in a pilot clinical study (63).

[i] Mut, mutated; ETS, erythroblast transformation specific; ERG, ETS transcription factor ERG; ETV, ETS variant transcription factor; EZH2, enhancer of zeste 2 polycomb repressive complex 2 subunit; NKX3-1, NK3 homeobox 1; SPOP, speckle type BTB/POZ protein; SPINK1, serine protease inhibitor Kazal type 1; EGFR, epidermal growth factor receptor; FOXA1, forkhead box A1; IDH1, isocitrate dehydrogenase (NAPD+) 1; CDK12, cyclin dependent kinase 12; TSG, tumor suppressor gene. GS, Gleason Score; CRPC, castration-resistant prostate cancer.

Multiple complex chromosomal rearrangement, as a cause of molecular alterations in PCa

There are several molecular alterations in the PCa, such as copy number changes, gene fusions, single nucleotide mutations and polymorphisms, methylation, microRNAs and long non-coding RNAs, one of the most characteristic involves multiple complex chromosomal rearrangement processes (Fig. 1A), which has been reported in 63% of PCa cases (18). These rearrangements can be classified as chromothripsis or chromoplexy (Fig. 1B and C), and some coordinated structured rearrangements may have intermediate chromothripsis and chromoplexy properties (11), for instance both chromothripsis and chromoplexy display random breakage and fusion of genomic segments with low copy numbers, most likely mediated by non-homologous end-joining.

In addition, Dzamba et al (18) used the CouGaR statistical method, in 63% of PCa, and in 27% of bladder cancers, which is in contrast with other types of cancer such as rectal, breast and thyroid cancer, where these types of alterations have not been identified. The CouGaR method is a novel method for predicting the overall genomic configuration resulting from characterizing the genomic structure of amplified complex genomic rearrangements, leveraging both depth of coverage (DOC) and discordant pair-end mapping techniques, to identify multiple chromosomal rearrangements. The results of Dzamba et al are noteworthy, as it has been reported that bladder and PCa may both develop in the same patient (19).

It has been found that the breaking points of DNA rearrangements are more likely to occur near specific DNA sequences, where the androgen receptor (AR) binds as a transcription factor, known as androgen response elements (ARE), compared with that in other randomly predicted locations, that is anywhere else in the genome (20,21). This finding suggests that AR-ARE complexes may be predisposed to genomic rearrangements through transcriptional stress, since androgen signaling promotes co-recruitment of androgen receptor and topoisomerase II β (TOP2β) to sites of TMPRSS2-ERG genomic breakpoints, triggering recombinogenic TOP2β-mediated DNA double-strand breaks. For example, it has been found that transmembrane serine protease 2 (TMPRSS2)-ERG fusion is induced by the interaction of androgens with AR (14,22).

In the development of PCa, chromothripsis is relatively rare and occurs as one clonal early event; in contrast, chromoplexy is a common and sequential event, which is detected at clonal or sub-clonal level (14,18,23). In a study performed using 57 patients with PCa, Baca et al (11) identified over 5,000 somatic rearrangements associated with this type of chromosomal rearrangements. Several cancer genes were repeatedly deleted or rearranged by chromoplexy, including PTEN, NK3 homeobox 1, cyclin dependent kinase inhibitor 1B, tumor protein p53 (TP53), and RB transcriptional corepressor 1. These multiple complex chromosomal rearrangements are one of the primary reasons for the high molecular heterogeneity in PCa.

Molecular subtypes in PCa

Over the last decade, several studies have focused on determining the excluding molecular events in the development of PCa, which are able to establish different subtypes, and are associated with the prognosis of the disease, and have the potential to be developed into targeted therapies (11,16,17,24). Therefore, the current status of PCa subtypes, will be subsequently discussed, using TCGA study (17) as a reference for the majority of the comparisons, the prognostic involvement, and the therapies of inhibitors of target oncogenes associated with subtypes, such as EZH2 or ERG (Table I), which have been used or are currently in the experimental phase, in this way some treatments that apply to cases that are ETS(+) will not work for those that are ETS(−), such as blocking function of ERG regulating co-factors, such as PARP1 (2527) (Table I). Table I summarizes the main clinical-pathological characteristics of the subtypes and treatments that have been investigated as a development of personalized medicine (13,17,2426,2863).

ETS(+) subtypes

The ETS family of transcription factors consists of phosphorylated proteins with DNA-binding domains (ETS domain) that act as either activators or repressors of transcription. The family consists of 30 identified genes, 28 of which are found in the human genome. Previous studies have found that between 50 and 70% of patients with PCa overexpress ETS, following gene fusion, which causes ETS to be controlled by ARE (12). Due to the prevalence of these types of rearrangements, efforts to molecularly characterize PCa begin by separating the cases which have gene fusion from those that do not, termed ETS(+) and ETS(−), respectively in numerous studies (20,38,64). TCGA study (17) has characterized four different ETS(+) subtypes, depending on the type of ETS involved in the fusion: ERG, ETV1, ETV4 and Fli-1 proto-oncogene, ETS transcription factor ERG is the most frequently overexpressed ETS. TMPRSS2 the most frequent fusion gene in all ETS fusions; however, fusions with other androgen-regulated genes have also been described, including solute carrier family 45 member 3 and N-myc downstream regulated 1 (65).

The TMPRSS2-ERG gene fusion causes ERG overexpression, which initiates a cascade of events that continues with Enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2) overexpression and decreased NKX3-1 expression (Fig. 2) (20,28). EZH2 is a methyltransferase from the polycomb group, involved in tissue-specific differentiation by histone methylation (H3K27), while NKX3-1 is an androgen-regulated, prostate-specific gene, which encodes a critical transcription factor during prostate development by downregulating epithelial cell growth, and is considered a TSG (20,28,66).

Other common molecular alterations in PCa are deletions and point mutations of PTEN, leading to activation of Akt, which can subsequently lead to over-activation of AR signaling; and in the presence of TMPRSS2-ERG gene fusion, it can increase the activation of a cascade involving ERG, EZH2 and NKX3-1 (Fig. 2) (20,31,33).

ETS(−) subtypes

ETS(−) are PCa subtypes with a canonic alteration, which is different to that in the ETS fusion genes. Whereas rearrangements occurring in ETS(+) tumors display features of chromoplexy, ETS(−) tumors display a higher frequency of chromothripsis (11). Among the ETS(−) subtypes, different studies have found the existence of a highly-expressed EZH2 subtype, which is independent of ERG expression (38,39). The overexpression of EZH2 has been described as well; it is caused by nuclear phosphoprotein MYC, which is involved in the cell cycle progression, apoptosis and cellular transformation. MYC oncogene also represses transcription of microRNA (miR)-26a and miR-26b, which are targets of EZH2, thus contributing to overexpression of EZH2 (36,38). In addition, the genomic or functional loss of miR-101, a TSG whose targets include the EZH2 gene, causes the overexpression of this gene (37) (Fig. 2).

Since epigenetic abnormalities driven by MYC overexpression have been associated with a poorer prognosis of PCa (progression-free survival after radical prostatectomy), therapies targeting these abnormalities may favor patients with EZH2 overexpression (2527) (Table I). Furthermore, TCGA describes 3 ETS(−) subtypes, characterized by mutations in genes, including SPOP, overexpression of serine protease inhibitor Kazal type 1 (SPINK1), forkhead box A1 (FOXA1) or isocitrate dehydrogenase (NAPD+)1 (IDH1) (17) (Table I). Previous studies have reported an overexpression of SPINK, as an independent subtype (66,67); however, its overexpression was associated with the mutated SPOP subtype in TCGA study (17). In 2018, Wu et al (63) reported a novel subtype, characterized by the inactivation of cyclin dependent kinase 12, which could benefit from immune checkpoint inhibition therapy (Table I).

In TCGA study, 26% of PCa cases could not be classified into any of the identified subtypes (17). There were three major groups of prostate cancers in the study, one with mostly unaltered genomes (referred to as quiet), a second group encompassing 50% of all tumors with an intermediate level of SCNAs, and a third group with a high burden of arm level genomic gains and losses (17). These PCa were clinically and genomically heterogeneous; low-pass and high-pass whole-genome sequencing (WGS) on 100 and 19 tumor/normal pairs, some of had numerous somatic copy number alterations (SCNA) and a high GS, which is an indicator of poor prognosis; 33% of them were genomically similar to the SPOP and FOXA1 subtypes, others were enriched for mutations of TP53, KDM6A, and KMT2D (lysine methyltransferase 2D) or specific SCNAs spanning MYC and CCND1 (cyclin D1) and other cases had a low GS (GS 6) with fewer genomic alterations (38% in the ‘quiet’ class vs. 8% in the class with the greatest burden of alterations), such as SCNA and DNA methylation patterns similar to those in normal tissue (17).

Table I organizes the different subtypes with their clinical significance and the treatment options that are being addressed for the future development of personalized medicine; however, this classification has not been clear or reproducible in numerous cases, nor has the response of patients to the treatments either, as ~75% of PCa are multifocal, and more than one subtypes can coexist in the same patient with multifocal PCa (6,68).

Intratumoral molecular heterogeneity

The Gleason classification, using histological methods, that heterogeneity in multifocal PCa can be recognized, which is higher when analyzed from a molecular point of view (6,69). This intratumoral heterogeneity makes it difficult to associate specific molecular alterations, detected in a single focus, with the clinical behavior of a patient with multifocal PCa (6,69). In molecular studies, the pattern of the index tumor is typically obtained to assign molecular alterations and subtypes; however, other foci are not taken into account (6). The index tumor is the largest tumor focus, which in most cases (89%) can be associated with significant pathological parameters, such as the highest GS, the largest tumor volume and extraprostatic extension (70); however, this is not always the case (69). Numerous studies have found that a single clone is responsible for the dispersal of all metastatic foci (13,71,72); which suggests that identifying the ‘deadly’ clone is of utmost importance, and should not necessarily start from the index tumor.

Due to the heterogeneity caused by multifocality in PCa, it is necessary to perform studies, that analyze the molecular alterations in various foci, which would allow the identification of the impact of different molecular subtypes in the same patient (4,64). Some studies using the TMPRSS2-ERG fusion (73,74), PTEN deletion (75), SPINK1, ERG (67), and whole genome sequencing (6,7577) have found a markedly interfocal discordance, which was consistent with the concept that multiple foci of PCa have a multiclonal origins (6). Wei et al (6) also found this heterogeneity when using commercial PCa diagnostic kits (Decipher, Prolaris and Oncotype DX), which are based on the expression of various genes, including immune response genes like testis-specific basic protein and PBX homeobox 1 of Decipher, cell cycle-related genes CDC20 (Cell Division Cycle 2), CDKN3 (Cyclin Dependent Kinase Inhibitor 3), CDC2 (cell division control protein 2 homolog) of Prolaris, genes of androgen signaling AZGP1 (Alpha-2-Glycoprotein 1) and FAM13C (Family With Sequence Similarity 13 Member C) of Oncotype DX. These results suggest that applying a single subtype of the molecular taxonomy of PCa proposed by TCGA to a patient, i.e., studying a single focus, is an over simplified and incorrect view of the molecular landscape of PCa.

Molecular progression to castrate-resistant prostate cancer

As aforementioned, the origins of PCa begins as a pre-neoplastic lesion which progresses to localized cancer, and can subsequently metastasize. Elimination of androgens using surgical or chemical castration, in numerous cases, results in control of PCa (78,79). However, when relapse occurs despite treatment, PCa has progressed to an androgen-independent form of cancer or CRPC, which is considered the most aggressive form of PCa (80).

Most of the molecular alterations in PCa have been described from HGPIN (or associated with HGPIN). However, there are studies, which have found associations between some of these alterations with different stages of PCa progression. ETS fusions and FOXA1 mutations frequently occur in HGPIN (14,23). Overexpression of SPINK1 (~10–25%) and SPOP (~11%), TP53 (~25%), IDH1 (~1%), MAP3K7 and CHD1 (~10–20%) mutations (11,13,17,23,49,81) are more frequent in localized PCa, while the highest and lowest expression levels of EZH2 and NKX3-1, respectively, together with PTEN deletion, have been found in metastatic CRPC (11,23,82). Monoallelic loss of PTEN is present in up to 60% of localized prostate cancers and complete loss of PTEN in prostate cancer is linked to metastasis and androgen-independent progression (86). Alteration of the AR signaling pathway compared with that in other pathways in CRPC suggest that AR signaling continues being the ‘master regulator’ for PCa progression (45,55,84), including AR copy number gain (24% of CRPCs) or AR point mutation (20% of CRPCs). These results assist to define the sequence of the molecular events in the development of PCa, from the origins of the disease through to its progression into metastasis, resistance to treatment and death.

There are 3 types of cells which interact in PCa to survive androgen ablation treatment: Androgen-dependent, androgen producing, and androgen-independent cells (85). The interaction among them determines whether CRPC will develop or not. The pathway which leads from the development of androgen-dependent to androgen-independent cells is still unknown. However, there are two models which have been used to explain this process. Some studies suggest that there is a collection of androgen-independent preexisting cells following therapy (selection model) (86,87); in contrast, other studies postulate that cells acquire novel alterations which allow them to survive in the absence of androgens (adaptation model) (88) for developing CRPC (Fig. 3).

Selection model

In this model, primary PCa consists of a heterogeneous mix of luminal, neuroendocrine and stem cells. When the patient undergoes androgen ablation treatment, most of the androgen-dependent cells undergo apoptosis, while androgen-independent cells persist and survive due to their low androgen requirement (Fig. 3) (86,87,89).

The vast majority of PCa are luminally differentiated adenocarcinomas with the presence of neuroendocrine cells, and respond to hormonal therapy (90); however, there are some tumors, which consist of aggressive and highly proliferating neuroendocrine cells only, for example small cell neuroendocrine PCa, which do not respond to hormone therapy, therefore platinum-based chemotherapy (phase II trial) is used (90,91). These malignant neuroendocrine cells, which share their origin with normal prostatic neuroendocrine cells (93), express epidermal growth factor receptor (EGFR) and receptor tyrosine-protein kinase erbB-2; for these reasons, they are classified as androgen-independent cells (94), and their abundance is considered a promising prognostic marker for the development of CRPC (95).

A previous study compared global transcriptomic profiles of normal basal and luminal epithelial lineages from samples of patients with PCa and PCa cell lines (87). It was found that PCa cells exhibited a gene expression profile similar to that found in a luminal cell and aggressive and neuroendocrine PCa were similar to basal cells (87).

In addition to cell type, in a few cases, point mutations in AR can cause cells, which were originally androgen-dependent, to become androgen-independent cells, and can be resistant to therapy, that is, those pre-existing mutations in the localized disease confer a selective advantage with threapy-resistant cells (84,9597). The S646F mutation within AR, in the hinge region, has been associated with a short response to endocrine therapy, due to a markedly increased transcriptional activity on ARE-containing promoters (95). In addition, AR gene copies (two to four copies), due to polysomy of the X-chromosome, are present in a subgroup of localized PCa, and these specimens may have an advantage in low concentrations of androgens due to therapy (96), since the additional AR copies may be a factor leading to a poor clinical outcome of antiandrogen therapy as there is a compensatory mechanism allowing activation of the AR post-castration. Furthermore, that study concluded that high stage primary prostate cancer may be associated with increased frequencies of aneuploidies of the X chromosome resulting in an increased AR gene copies number.

Adaptation model

The adaptation model suggests that resistance to castration is the result of the acquisition of genetic and/or epigenetic alterations in response to therapy, which allows cells that were previously dependent on androgens to proliferate at low concentrations of androgens due to therapy (97). Adaptations to androgen ablation treatment include the occurrence of mutations with a copy number gain of the AR gene, changes in the expression of AR co-regulation molecules (48,84,98), and deregulation of key molecules in proliferation (99), such as Akt overexpression (29).

The evidence supports the selection model (100102); however, it is difficult to establish either of these models as the definitive or exclusive mechanism. It is also possible that both models independently or cooperatively contribute to the development of CRPC (85,100). Some cells could be independent of androgens and be selected, or they can also gain adaptive advantages to proliferate at low concentrations of androgens; or there could be cells selected for their independence from androgens, and others that adapt and proliferate at low concentrations of androgens (100).

The aforementioned neuroendocrine PCa from small cells may arise de novo, which would support the selection model; however, these generally occur as recurrent tumors in men who have received hormone therapy for prostatic adenocarcinoma, suggesting that the neuroendocrine phenotype is driven by the hostile environment created by hormone therapy, or the adaptation model, and such tumors are composed of pure neuroendocrine cells that are highly proliferative and aggressive (88,90,103), due mutation in P53 (104) (allele of a missense transition converting G to A at position 747, changing negatively charged aspartic acid to hydrophilic amino acid asparagine at amino acid 184) and the inactivation of the interleukin-8-C-X-C motif chemokine receptor 2 signaling pathway (90,104). Differential epigenetic markers between the neuroendocrine and non-neuroendocrine CRPC support a key role of the epigenome in the emergence and/or maintenance of neuroendocrine CRPC (105).

With the widespread use of novel drugs targeting the androgenic axis, such as abiraterone acetate and enzalutamide, there has been a rapid increase in the incidence of small cell neuroendocrine carcinoma (91,105), on the basis of autopsy series and other studies this type of PCa may represent approximately 25% of late stage of PCa (106), which will become a major challenge in the treatment of these patients. The clarification of the determining factors that lead to CRPC will be key to understanding the carcinogenesis process and guiding the clinical management of each patient.

Conclusions

The use of molecular subtypes in PCa to personalize treatment is promising; however, it is necessary to consider multifocality. The lack of an association between subtype and prognosis in PCa may be due to the fact that only the index tumor is investigated. It is important to analyze the subtypes in multiple foci, to elucidate the development of PCa, which could include different molecular subtypes; during the development of tumor foci, these would be selected according to their adaptive advantages, such as resistance to castration and the ability to metastasize.

The presence or absence of a specific alteration in any of the foci may be associated with the potential of PCa to progress to CRPC or to be the target for the development of targeted therapy, which does not necessarily have to be found in the index lesion.

Acknowledgements

Not applicable.

Funding

The present study was funded by the National Institute of Cancerology of Bogota, Colombia (grant no. C190103001-09).

Availability of data and materials

Data sharing is not applicable to this article, as no datasets were generated or analyzed during the current study.

Authors' contributions

YYSM and MLS were responsible for writing the manuscript, editing, acquisition, analysis, interpretation of the data, contributed to data acquisition and edited references. YYSM, MCSS, RV, JAM and MLS contributed to the conception and design of the article and were involved in critical revision of the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

IARC: Global Cancer Observatory https://gco.iarc.fr/today/home: GLOBOCAN, . 2018.The Global Cancer Observatory (GCO) is an interactive web-based platform presenting global cancer statistics to inform cancer control and research.

2 

Beltran H, Yelensky R, Frampton GM, Park K, Downing SR, MacDonald TY, Jarosz M, Lipson D, Tagawa ST, Nanus DM, et al: Targeted next-generation sequencing of advanced prostate cancer identifies potential therapeutic targets and disease heterogeneity. Eur Urol. 63:920–926. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Lu-Yao G, Albertsen PC, Stanford JL, Stukel TA, Walker-Corkery E and Barry MJ: Screening, treatment, and prostate cancer mortality in the Seattle area and Connecticut: Fifteen-year follow-up. J Gen Intern Med. 23:1809–1814. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Partin AW: High-grade prostatic intraepithelial neoplasia on a prostate biopsy-what does it mean? Rev Urol. 4:157–158. 2002.PubMed/NCBI

5 

De Marzo AM, Marchi VL, Epstein JI and Nelson WG: Proliferative inflammatory atrophy of the prostate: Implications for prostatic carcinogenesis. Am J Pathol. 155:1985–1992. 1999. View Article : Google Scholar : PubMed/NCBI

6 

Wei L, Wang J, Lampert E, Schlanger S, DePriest AD, Hu Q, Gomez EC, Murakam M, Glenn ST, Conroy J, et al: Intratumoral and intertumoral genomic heterogeneity of multifocal localized prostate cancer impacts molecular classifications and genomic prognosticators. Eur Urol. 71:183–192. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Epstein JI, Allsbrook WC, Amin MB and Egevad LL; ISUP Grading Committee, : The 2005 International society of urological pathology (ISUP) consensus conference on gleason grading of prostatic carcinoma. Am J Surg Pathol. 29:1228–1242. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Epstein JI, Egevad L, Amin MB, Delahunt B, Srigley JR and Humphrey PA; Grading Committee, : The 2014 International Society of Urological Pathology (ISUP) Consensus Conference on Gleason Grading of Prostatic Carcinoma: Definition of Grading Patterns and Proposal for a New Grading System. Am J Surg Pathol. 40:244–252. 2016. View Article : Google Scholar : PubMed/NCBI

9 

De Nunzio C, Pastore AL, Lombardo R, Simone G, Leonardo C, Mastroianni R, Collura D, Muto G, Gallucci M, Carbone A, et al: The new Epstein gleason score classification significantly reduces upgrading in prostate cancer patients. Eur J Surg Oncol. 44:835–839. 2018. View Article : Google Scholar : PubMed/NCBI

10 

Baca SC and Garraway LA: The genomic landscape of prostate cancer. Front Endocrinol (Lausanne). 3:692012. View Article : Google Scholar : PubMed/NCBI

11 

Baca SC, Prandi D, Lawrence MS, Mosquera JM, Romanel A, Drier Y, Park K, Kitabayashi N, MacDonald TY, Ghandi M, et al: Punctuated evolution of prostate cancer genomes. Cell. 153:666–677. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Tomlins SA, Rhodes DR, Perner S, Dhanasekaran SM, Mehra R, Sun XW, Varambally S, Cao X, Tchinda J, Kuefer R, et al: Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science. 310:644–648. 2005. View Article : Google Scholar : PubMed/NCBI

13 

Barbieri CE, Baca SC, Lawrence MS, Demichelis F, Blattner M, Theurillat JP, White TA, Stojanov P, Van Allen E, Stransky N, et al: Exome sequencing identifies recurrent SPOP, FOXA1 and MED12 mutations in prostate cancer. Nat Genet. 44:685–689. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Berger MF, Lawrence MS, Demichelis F, Drier Y, Cibulskis K, Sivachenko AY, Sboner A, Esgueva R, Pflueger D, Sougnez C, et al: The genomic complexity of primary human prostate cancer. Nature. 470:214–220. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Tomlins SA, Laxman B, Dhanasekaran SM, Helgeson BE, Cao X, Morris DS, Menon A, Jing X, Cao Q, Han B, et al: Distinct classes of chromosomal rearrangements create oncogenic ETS gene fusions in prostate cancer. Nature. 448:595–599. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Tomlins SA, Alshalalfa M, Davicioni E, Erho N, Yousefi K, Zhao S, Haddad Z, Den RB, Dicker AP, Trock BJ, et al: Characterization of 1577 primary prostate cancers reveals novel biological and clinicopathologic insights into molecular subtypes. Eur Urol. 68:555–567. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Cancer Genome Atlas Research Network, . The Molecular Taxonomy of Primary Prostate Cancer. Cell. 163:1011–1025. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Dzamba M, Ramani AK, Buczkowicz P, Jiang Y, Yu M, Hawkins C and Brudno M: Identification of complex genomic rearrangements in cancers using CouGaR. Genome Res. 27:107–117. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Chun TY: Coincidence of bladder and prostate cancer. J Urol. 157:65–67. 1997. View Article : Google Scholar : PubMed/NCBI

20 

Yu J, Mani RS, Cao Q, Brenner CJ, Cao X, Wang X, Wu L, Li J, Hu M, Gong Y, et al: An integrated network of androgen receptor, polycomb, and TMPRSS2-ERG gene fusions in prostate cancer progression. Cancer Cell. 17:443–454. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Lin C, Yang L, Tanasa B, Hutt K, Ju BG, Ohgi K, Zhang J, Rose DW, Fu XD, Glass CK and Rosenfeld MG: Nuclear receptor-induced chromosomal proximity and DNA breaks underlie specific translocations in cancer. Cell. 139:1069–1083. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Mani RS, Tomlins SA, Callahan K, Ghosh A, Nyati MK, Varambally S, Palanisamy N and Chinnaiyan AM: Induced chromosomal proximity and gene fusions in prostate cancer. Science. 326:12302009. View Article : Google Scholar : PubMed/NCBI

23 

Jung SH, Shin S, Kim MS, Baek IP, Lee JY, Lee SH, Kim TM, Lee SH and Chung YJ: Genetic progression of high grade prostatic intraepithelial neoplasia to prostate cancer. Eur Urol. 69:823–830. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Lapointe J, Li C, Higgins JP, van de Rijn M, Bair E, Montgomery K, Ferrari M, Egevad L, Rayford W, Bergerheim U, et al: Gene expression profiling identifies clinically relevant subtypes of prostate cancer. Proc Natl Acad Sci USA. 101:811–816. 2004. View Article : Google Scholar : PubMed/NCBI

25 

Brenner JC, Ateeq B, Li Y, Yocum AK, Cao Q, Asangani IA, Patel S, Wang X, Liang H, Yu J, et al: Mechanistic rationale for inhibition of poly(ADP-ribose) polymerase in ETS gene fusion-positive prostate cancer. Cancer Cell. 19:664–678. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Williamson SR and Cheng L: Potential for targeted therapy in prostate cancers with ERG abnormalities. Asian J Androl. 13:781–782. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Karpova Y, Wu C, Divan A, McDonnell ME, Hewlett E, Makhov P, Gordon J, Ye M, Reitz AB, Childers WE, et al: Non-NAD-like PARP-1 inhibitors in prostate cancer treatment. Biochem Pharmacol. 167:149–162. 2019. View Article : Google Scholar : PubMed/NCBI

28 

Kunderfranco P, Mello-Grand M, Cangemi R, Pellini S, Mensah A, Albertini V, Malek A, Chiorino G, Catapano CV and Carbone GM: ETS transcription factors control transcription of EZH2 and epigenetic silencing of the tumor suppressor gene Nkx3.1 in prostate cancer. PLoS One. 5:e105472010. View Article : Google Scholar : PubMed/NCBI

29 

Wang S, Gao J, Lei Q, Rozengurt N, Pritchard C, Jiao J, Thomas GV, Li G, Roy-Burman P, Nelson PS, et al: Prostate-specific deletion of the murine Pten tumor suppressor gene leads to metastatic prostate cancer. Cancer Cell. 4:209–221. 2003. View Article : Google Scholar : PubMed/NCBI

30 

Gao T, Mei Y, Sun H, Nie Z, Liu X and Wang S: The association of Phosphatase and tensin homolog (PTEN) deletion and prostate cancer risk: A meta-analysis. Biomed Pharmacother. 83:114–121. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Leinonen KA, Saramäki OR, Furusato B, Kimura T, Takahashi H, Egawa S, Suzuki H, Keiger K, Ho Hahm S, Isaacs WB, et al: Loss of PTEN is associated with aggressive behavior in ERG-positive prostate cancer. Cancer Epidemiol Biomarkers Prev. 22:2333–2344. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Ngollo M, Lebert A, Dagdemir A, Judes G, Karsli-Ceppioglu S, Daures M, Kemeny JL, Penault-Llorca F, Boiteux JP, Bignon YJ, et al: The association between histone 3 lysine 27 trimethylation (H3K27me3) and prostate cancer: Relationship with clinicopathological parameters. BMC Cancer. 14:9942014. View Article : Google Scholar : PubMed/NCBI

33 

Ishigami-Yuasa M, Ekimoto H and Kagechika H: Class IIb HDAC inhibition enhances the inhibitory effect of Am80, a synthetic retinoid, in prostate cancer. Biol Pharm Bull. 42:448–452. 2019. View Article : Google Scholar : PubMed/NCBI

34 

Bai Y, Zhang Z, Cheng L, Wang R, Chen X, Kong Y, Feng F, Ahmad N, Li L and Liu X: Inhibition of enhancer of zeste homolog 2 (EZH2) overcomes enzalutamide resistance in castration-resistant prostate cancer. J Biol Chem. 294:9911–9923. 2019. View Article : Google Scholar : PubMed/NCBI

35 

Taplin ME, Hussain A, Shah S, Neal D. Shore, Manish Agrawal, William Clark, et al: ProSTAR: A phase Ib/II study of CPI-1205, a small molecule inhibitor of EZH2, combined with enzalutamide (E) or abiraterone/prednisone (A/P) in patients with metastatic castration-resistant prostate cancer (mCRPC). J Clin Oncol. 2019. View Article : Google Scholar : PubMed/NCBI

36 

Koh CM, Iwata T, Zheng Q, Bethel C, Yegnasubramanian S and De Marzo AM: Myc enforces overexpression of EZH2 in early prostatic neoplasia via transcriptional and post-transcriptional mechanisms. Oncotarget. 2:669–683. 2011. View Article : Google Scholar : PubMed/NCBI

37 

Varambally S, Cao Q, Mani RS, Shankar S, Wang X, Ateeq B, Laxman B, Cao X, Jing X, Ramnarayanan K, et al: Genomic loss of microRNA-101 leads to overexpression of histone methyltransferase EZH2 in cancer. Science. 322:1695–699. 2008. View Article : Google Scholar : PubMed/NCBI

38 

Börno ST, Fischer A, Kerick M, Fälth M, Laible M, Brase JC, Kuner R, Dahl A, Grimm C, Sayanjali B, et al: Genome-wide DNA methylation events in TMPRSS2-ERG fusion-negative prostate cancers implicate an EZH2-dependent mechanism with miR-26a hypermethylation. Cancer Discov. 2:1024–1035. 2012. View Article : Google Scholar : PubMed/NCBI

39 

Melling N, Thomsen E, Tsourlakis MC, Kluth M, Hube-Magg C, Minner S, Koop C, Graefen M, Heinzer H, Wittmer C, et al: Overexpression of enhancer of zeste homolog 2 (EZH2) characterizes an aggressive subset of prostate cancers and predicts patient prognosis independently from pre- and postoperatively assessed clinicopathological parameters. Carcinogenesis. 36:1333–1340. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Uchiyama N, Tanaka Y and Kawamoto T: Aristeromycin and DZNeP cause growth inhibition of prostate cancer via induction of mir-26a. Eur J Pharmacol. 812:138–146. 2017. View Article : Google Scholar : PubMed/NCBI

41 

Kirschner AN, Wang J, van der Meer R, Anderson PD, Franco-Coronel OE, Kushner MH, Everett JH, Hameed O, Keeton EK, Ahdesmaki M, et al: PIM kinase inhibitor AZD1208 for treatment of MYC-driven prostate cancer. J Natl Cancer Inst. 107:dju4072015. View Article : Google Scholar : PubMed/NCBI

42 

Rebello RJ, Kusnadi E, Cameron DP, Pearson HB, Lesmana A, Devlin JR, Drygin D, Clark AK, Porter L, Pedersen J, et al: The dual inhibition of RNA Pol I transcription and PIM kinase as a new therapeutic approach to treat advanced prostate cancer. Clin Cancer Res. 22:5539–5552. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Boysen G, Barbieri CE, Prandi D, Blattner M, Chae SS, Dahija A, Nataraj S, Huang D, Marotz C, Xu L, et al: SPOP mutation leads to genomic instability in prostate cancer. Elife. 4:e092072015. View Article : Google Scholar : PubMed/NCBI

44 

Rodrigues LU, Rider L, Nieto C, Romero L, Karimpour-Fard A, Loda M, Lucia MS, Wu M, Shi L, Cimic A, et al: Coordinate loss of MAP3K7 and CHD1 promotes aggressive prostate cancer. Cancer Res. 75:1021–1034. 2015. View Article : Google Scholar : PubMed/NCBI

45 

Grasso CS, Wu YM, Robinson DR, Cao X, Dhanasekaran SM, Khan AP, Quist MJ, Jing X, Lonigro RJ, Brenner JC, et al: The mutational landscape of lethal castration-resistant prostate cancer. Nature. 487:239–243. 2012. View Article : Google Scholar : PubMed/NCBI

46 

Shen C, Zhang J, Qi M, Chang YWY and BH: Roles of serine protease inhibitor kazal type 1 (SPINK1) in prostate cancer. Med chem. 4:725–728. 2014. View Article : Google Scholar

47 

Liu D, Takhar M, Alshalalfa M, Erho N, Shoag J, Jenkins RB, Karnes RJ, Ross AE, Schaeffer EM, Rubin MA, et al: Impact of the SPOP mutant subtype on the interpretation of clinical parameters in prostate cancer. JCO Precis Oncol. 2018:102018.

48 

Johnson MH, Ross AE, Alshalalfa M, Erho N, Yousefi K, Glavaris S, Fedor H, Han M, Faraj SF, Bezerra SM, et al: SPINK1 Defines a molecular subtype of prostate cancer in men with more rapid progression in an at risk, natural history radical prostatectomy cohort. J Urol. 196:1436–1444. 2016. View Article : Google Scholar : PubMed/NCBI

49 

Yun SJ, Kim SK, Kim J, Cha EJ, Kim JS, Kim SJ, Ha YS, Kim YH, Jeong P, Kang HW, et al: Transcriptomic features of primary prostate cancer and their prognostic relevance to castration-resistant prostate cancer. Oncotarget. 8:114845–114855. 2017. View Article : Google Scholar : PubMed/NCBI

50 

Tiwari R, Manzar N, Bhatia V, Yadav A, Nengroo MA, Datta D, Carskadon S, Gupta N, Sigouros M, Khani F, et al: Androgen deprivation upregulates SPINK1 expression and potentiates cellular plasticity in prostate cancer. Nat Commun. 11:3842020. View Article : Google Scholar : PubMed/NCBI

51 

Geng C, Rajapakshe K, Shah SS, Shou J, Eedunuri VK, Foley C, Fiskus W, Rajendran M, Chew SA, Zimmermann M, et al: Androgen receptor is the key transcriptional mediator of the tumor suppressor SPOP in prostate cancer. Cancer Res. 74:5631–5643. 2014. View Article : Google Scholar : PubMed/NCBI

52 

Lu D, Lee J, Lee A and Lee R: Development of a new approach for the therapy of prostate cancer with SPOP mutations. J Cancer Therapy. 6:841–848. 2015. View Article : Google Scholar

53 

Boysen G, Rodrigues DN, Rescigno P, Seed G, Dolling D, Riisnaes R, Crespo M, Zafeiriou Z, Sumanasuriya S, Bianchini D, et al: SPOP-Mutated/CHD1-deleted lethal prostate cancer and abiraterone sensitivity. Clin Cancer Res. 24:5585–5593. 2018. View Article : Google Scholar : PubMed/NCBI

54 

Ateeq B, Tomlins SA, Laxman B, Asangani IA, Cao Q, Cao X, Li Y, Wang X, Feng FY, Pienta KJ, et al: Therapeutic targeting of SPINK1-positive prostate cancer. Sci Transl Med. 3:72ra172011. View Article : Google Scholar : PubMed/NCBI

55 

Stelloo S, Nevedomskaya E, Kim Y, Schuurman K, Valle-Encinas E, Lobo J, Krijgsman O, Peeper DS, Chang SL, Feng FY, et al: Integrative epigenetic taxonomy of primary prostate cancer. Nat Commun. 9:49002018. View Article : Google Scholar : PubMed/NCBI

56 

Imamura Y, Sakamoto S, Endo T, Utsumi T, Fuse M, Suyama T, Kawamura K, Imamoto T, Yano K, Uzawa K, et al: FOXA1 promotes tumor progression in prostate cancer via the insulin-like growth factor binding protein 3 pathway. PLoS One. 7:e424562012. View Article : Google Scholar : PubMed/NCBI

57 

Adams EJ, Karthaus WR, Hoover E, Liu D, Gruet A, Zhang Z, Cho H, DiLoreto R, Chhangawala S, Liu Y, et al: FOXA1 mutations alter pioneering activity, differentiation and prostate cancer phenotypes. Nature. 571:408–412. 2019. View Article : Google Scholar : PubMed/NCBI

58 

Song B, Park SH, Zhao JC, Fong KW, Li S, Lee Y, Yang YA, Sridhar S, Lu X, Abdulkadir SA, et al: Targeting FOXA1-mediated repression of TGF-β signaling suppresses castration-resistant prostate cancer progression. J Clin Invest. 129:569–582. 2019. View Article : Google Scholar : PubMed/NCBI

59 

Gui B, Gui F, Takai T, Feng C, Bai X, Fazli L, Dong X, Liu S, Zhang X, Zhang W, et al: Selective targeting of PARP-2 inhibits androgen receptor signaling and prostate cancer growth through disruption of FOXA1 function. Proc Natl Acad Sci USA. 116:14573–14582. 2019. View Article : Google Scholar : PubMed/NCBI

60 

Xu W, Yang H, Liu Y, Yang Y, Wang P, Kim SH, Ito S, Yang C, Wang P, Xiao MT, et al: Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of α-ketoglutarate-dependent dioxygenases. Cancer Cell. 19:17–30. 2011. View Article : Google Scholar : PubMed/NCBI

61 

Ghiam AF, Cairns RA, Thoms J, Dal Pra A, Ahmed O, Meng A, Mak TW and Bristow RG: IDH mutation status in prostate cancer. Oncogene. 31:38262012. View Article : Google Scholar : PubMed/NCBI

62 

Mondesir J, Willekens C, Touat M and de Botton S: IDH1 and IDH2 mutations as novel therapeutic targets: Current perspectives. J Blood Med. 7:171–180. 2016. View Article : Google Scholar : PubMed/NCBI

63 

Wu YM, Cieślik M, Lonigro RJ, Vats P, Reimers MA, Cao X, Ning Y, Wang L, Kunju LP, de Sarkar N, et al: Inactivation of CDK12 delineates a distinct immunogenic class of advanced prostate cancer. Cell. 173:1770–1782.e14. 2018. View Article : Google Scholar : PubMed/NCBI

64 

Tomlins SA, Rhodes DR, Yu J, Varambally S, Mehra R, Perner S, Demichelis F, Helgeson BE, Laxman B, Morris DS, et al: The role of SPINK1 in ETS rearrangement-negative prostate cancers. Cancer Cell. 13:519–528. 2008. View Article : Google Scholar : PubMed/NCBI

65 

Hermans KG, van Marion R, van Dekken H, Jenster G, van Weerden WM and Trapman J: 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. 66:10658–10663. 2006. View Article : Google Scholar : PubMed/NCBI

66 

Thangapazham R, Saenz F, Katta S, Mohamed AA, Tan SH, Petrovics G, Srivastava S and Dobi A: Loss of the NKX3.1 tumorsuppressor promotes the TMPRSS2-ERG fusion gene expression in prostate cancer. BMC Cancer. 14:162014. View Article : Google Scholar : PubMed/NCBI

67 

Fontugne J, Davis K, Palanisamy N, Udager A, Mehra R, McDaniel AS, Siddiqui J, Rubin MA, Mosquera JM and Tomlins SA: Clonal evaluation of prostate cancer foci in biopsies with discontinuous tumor involvement by dual ERG/SPINK1 immunohistochemistry. Mod Pathol. 29:157–165. 2016. View Article : Google Scholar : PubMed/NCBI

68 

Løvf M, Zhao S, Axcrona U, Johannessen B, Bakken AC, Carm KT, Hoff AM, Myklebost O, Meza-Zepeda LA and Lie AK: Multifocal primary prostate cancer exhibits high degree of genomic heterogeneity. Eur Urol. 75:498–505. 2019. View Article : Google Scholar : PubMed/NCBI

69 

Huang CC, Deng FM, Kong MX, Ren Q, Melamed J and Zhou M: Re-evaluating the concept of ‘dominant/index tumor nodule’ in multifocal prostate cancer. Virchows Arch. 464:589–594. 2014. View Article : Google Scholar : PubMed/NCBI

70 

McNeal JE, Price HM, Redwine EA, Freiha FS and Stamey TA: Stage A versus stage B adenocarcinoma of the prostate: Morphological comparison and biological significance. J Urol. 139:61–65. 1988. View Article : Google Scholar : PubMed/NCBI

71 

Liu W, Laitinen S, Khan S, Vihinen M, Kowalski J, Yu G, Chen L, Ewing CM, Eisenberger MA, Carducci MA, et al: Copy number analysis indicates monoclonal origin of lethal metastatic prostate cancer. Nat Med. 15:559–565. 2009. View Article : Google Scholar : PubMed/NCBI

72 

Haffner MC, Mosbruger T, Esopi DM, Fedor H, Heaphy CM, Walker DA, Adejola N, Gürel M, Hicks J, Meeker AK, et al: Tracking the clonal origin of lethal prostate cancer. J Clin Invest. 123:4918–4922. 2013. View Article : Google Scholar : PubMed/NCBI

73 

Barry M, Perner S, Demichelis F and Rubin MA: TMPRSS2-ERG fusion heterogeneity in multifocal prostate cancer: clinical and biologic implications. Urology. 70:630–633. 2007. View Article : Google Scholar : PubMed/NCBI

74 

Furusato B, Gao CL, Ravindranath L, Chen Y, Cullen J, McLeod DG, Dobi A, Srivastava S, Petrovics G and Sesterhenn IA: Mapping of TMPRSS2-ERG fusions in the context of multi-focal prostate cancer. Mod Pathol. 21:67–75. 2008. View Article : Google Scholar : PubMed/NCBI

75 

Yoshimoto M, Ding K, Sweet JM, Ludkovski O, Trottier G, Song KS, Joshua AM, Fleshner NE, Squire JA and Evans AJ: PTEN losses exhibit heterogeneity in multifocal prostatic adenocarcinoma and are associated with higher Gleason grade. Mod Pathol. 26:435–447. 2013. View Article : Google Scholar : PubMed/NCBI

76 

Boutros PC, Fraser M, Harding NJ, de Borja R, Trudel D, Lalonde E, Meng A, Hennings-Yeomans PH, McPherson A, Sabelnykova VY, et al: Spatial genomic heterogeneity within localized, multifocal prostate cancer. Nat Genet. 47:736–745. 2015. View Article : Google Scholar : PubMed/NCBI

77 

Cooper CS, Eeles R, Wedge DC, Van Loo P, Gundem G, Alexandrov LB, Kremeyer B, Butler A, Lynch AG, Camacho N, et al: Analysis of the genetic phylogeny of multifocal prostate cancer identifies multiple independent clonal expansions in neoplastic and morphologically normal prostate tissue. Nat Genet. 47:367–372. 2015. View Article : Google Scholar : PubMed/NCBI

78 

Crawford ED, Heidenreich A, Lawrentschuk N, Tombal B, Pompeo ACL, Mendoza-Valdes A, Miller K, Debruyne FMJ and Klotz L: Androgen-targeted therapy in men with prostate cancer: Evolving practice and future considerations. Prostate Cancer Prostatic Dis. 22:24–38. 2019. View Article : Google Scholar : PubMed/NCBI

79 

Vickers AJ, Bianco FJ, Serio AM, Eastham JA, Schrag D, K EA, Reuther AM, Kattan MW, Pontes JE and Scardino PT: The surgical learning curve for prostate cancer control after radical prostatectomy. J Natl Cancer Inst. 99:1171–1177. 2007. View Article : Google Scholar : PubMed/NCBI

80 

Yu EY, Gulati R, Telesca D, Jiang P, Tam S, Russell KJ, Nelson PS, Etzioni RD and Higano CS: Duration of first off-treatment interval is prognostic for time to castration resistance and death in men with biochemical relapse of prostate cancer treated on a prospective trial of intermittent androgen deprivation. J Clin Oncol. 28:2668–2673. 2010. View Article : Google Scholar : PubMed/NCBI

81 

Huang KC, Evans A, Donnelly B and Bismar TA: SPINK1 Overexpression in localized prostate cancer: A rare event inversely associated with ERG expression and exclusive of homozygous PTEN deletion. Pathol Oncol Res. 23:399–407. 2017. View Article : Google Scholar : PubMed/NCBI

82 

Green SM, Mostaghel EA and Nelson PS: Androgen action and metabolism in prostate cancer. Mol Cell Endocrinol. 360:3–13. 2012. View Article : Google Scholar : PubMed/NCBI

83 

Phin S, Moore MW and Cotter PD: Genomic rearrangements of PTEN in prostate cancer. Front Oncol. 3:2402013. View Article : Google Scholar : PubMed/NCBI

84 

Taylor BS, Schultz N, Hieronymus H, Gopalan A, Xiao Y, Carver BS, Arora VK, Kaushik P, Cerami E, Reva B, et al: Integrative genomic profiling of human prostate cancer. Cancer Cell. 18:11–22. 2010. View Article : Google Scholar : PubMed/NCBI

85 

Zhang J, Cunningham JJ, Brown JS and Gatenby RA: Integrating evolutionary dynamics into treatment of metastatic castrate-resistant prostate cancer. Nat Commun. 8:18162017. View Article : Google Scholar : PubMed/NCBI

86 

Lawson DA, Zong Y, Memarzadeh S, Xin L, Huang J and Witte ON: Basal epithelial stem cells are efficient targets for prostate cancer initiation. Proc Natl Acad Sci USA. 107:2610–2615. 2010. View Article : Google Scholar : PubMed/NCBI

87 

Zhang D, Park D, Zhong Y, Lu Y, Rycaj K, Gong S, Chen X, Liu X, Chao HP, Whitney P, et al: Stem cell and neurogenic gene-expression profiles link prostate basal cells to aggressive prostate cancer. Nat Commun. 7:107982016. View Article : Google Scholar : PubMed/NCBI

88 

Nouri M, Caradec J, Lubik AA, Li N, Hollier BG, Takhar M, Altimirano-Dimas M, Chen M, Roshan-Moniri M, Butler M, et al: Therapy-induced developmental reprogramming of prostate cancer cells and acquired therapy resistance. Oncotarget. 8:18949–18967. 2017. View Article : Google Scholar : PubMed/NCBI

89 

Wu C, Wyatt AW, Lapuk AV, McPherson A, McConeghy BJ, Bell RH, Anderson S, Haegert A, Brahmbhatt S, Shukin R, et al: Integrated genome and transcriptome sequencing identifies a novel form of hybrid and aggressive prostate cancer. J Pathol. 227:53–61. 2012. View Article : Google Scholar : PubMed/NCBI

90 

Lipianskaya J, Cohen A, Chen CJ, Hsia E, Squires J, Li Z, Zhang Y, Li W, Chen X, Xu H and Huang J: Androgen-deprivation therapy-induced aggressive prostate cancer with neuroendocrine differentiation. Asian J Androl. 16:541–544. 2014. View Article : Google Scholar : PubMed/NCBI

91 

Aparicio AM, Harzstark AL, Corn PG, Wen S, Araujo JC, Tu SM, Pagliaro LC, Kim J, Millikan RE, Ryan C, et al: Platinum-based chemotherapy for variant castrate-resistant prostate cancer. Clin Cancer Res. 19:3621–3630. 2013. View Article : Google Scholar : PubMed/NCBI

92 

Bonkhoff H and Remberger K: Differentiation pathways and histogenetic aspects of normal and abnormal prostatic growth: A stem cell model. Prostate. 28:98–106. 1996. View Article : Google Scholar : PubMed/NCBI

93 

Cortés MA, Cariaga-Martinez AE, Lobo MV, Martín Orozco RM, Motiño O, Rodríguez-Ubreva FJ, Angulo J, López-Ruiz P and Colás B: EGF promotes neuroendocrine-like differentiation of prostate cancer cells in the presence of LY294002 through increased ErbB2 expression independent of the phosphatidylinositol 3-kinase-AKT pathway. Carcinogenesis. 33:1169–1177. 2012. View Article : Google Scholar : PubMed/NCBI

94 

Abrahamsson PA, Wadström LB, Alumets J, Falkmer S and Grimelius L: Peptide-hormone- and serotonin-immunoreactive tumour cells in carcinoma of the prostate. Pathol Res Pract. 182:298–307. 1987. View Article : Google Scholar : PubMed/NCBI

95 

Thompson J, Hyytinen ER, Haapala K, Rantala I, Helin HJ, Jänne OA, Palvimo JJ and Koivisto PA: Androgen receptor mutations in high-grade prostate cancer before hormonal therapy. Lab Invest. 83:1709–1713. 2003. View Article : Google Scholar : PubMed/NCBI

96 

Röpke A, Erbersdobler A, Hammerer P, Palisaar J, John K, Stumm M and Wieacker P: Gain of androgen receptor gene copies in primary prostate cancer due to X chromosome polysomy. Prostate. 59:59–68. 2004. View Article : Google Scholar : PubMed/NCBI

97 

Nouri M, Ratther E, Stylianou N, Nelson CC, Hollier BG and Williams ED: Androgen-targeted therapy-induced epithelial mesenchymal plasticity and neuroendocrine transdifferentiation in prostate cancer: An opportunity for intervention. Front Oncol. 4:3702014. View Article : Google Scholar : PubMed/NCBI

98 

Han G, Buchanan G, Ittmann M, Harris JM, Yu X, Demayo FJ, Tilley W and Greenberg NM: Mutation of the androgen receptor causes oncogenic transformation of the prostate. Proc Natl Acad Sci USA. 102:1151–1156. 2005. View Article : Google Scholar : PubMed/NCBI

99 

Kaarbø M, Mikkelsen OL, Malerød L, Qu S, Lobert VH, Akgul G, Halvorsen T, Maelandsmo GM and Saatcioglu F: PI3K-AKT-mTOR pathway is dominant over androgen receptor signaling in prostate cancer cells. Cell Oncol. 32:11–27. 2010.PubMed/NCBI

100 

Terry S and Beltran H: The many faces of neuroendocrine differentiation in prostate cancer progression. Front Oncol. 4:602014. View Article : Google Scholar : PubMed/NCBI

101 

Choi N, Zhang B, Zhang L, Ittmann M and Xin L: Adult murine prostate basal and luminal cells are self-sustained lineages that can both serve as targets for prostate cancer initiation. Cancer Cell. 21:253–265. 2012. View Article : Google Scholar : PubMed/NCBI

102 

Germann M, Wetterwald A, Guzmán-Ramirez N, vander Pluijm G, Culig Z, Cecchini MG, Williams ED and Thalmann GN: Stem-like cells with luminal progenitor phenotype survive castration in human prostate cancer. Stem Cells. 30:1076–1086. 2012. View Article : Google Scholar : PubMed/NCBI

103 

Evans AJ, Humphrey PA, Belani J, van der Kwast TH and Srigley JR: Large cell neuroendocrine carcinoma of prostate: A clinicopathologic summary of 7 cases of a rare manifestation of advanced prostate cancer. Am J Surg Pathol. 30:684–693. 2006. View Article : Google Scholar : PubMed/NCBI

104 

Chen H, Sun Y, Wu C, Magyar CE, Li X, Cheng L, Yao JL, Shen S, Osunkoya AO, Liang C and Huang J: Pathogenesis of prostatic small cell carcinoma involves the inactivation of the P53 pathway. Endocr Relat Cancer. 19:321–331. 2012. View Article : Google Scholar : PubMed/NCBI

105 

Beltran H, Prandi D, Mosquera JM, Benelli M, Puca L, Cyrta J, Marotz C, Giannopoulou E, Chakravarthi BV, Varambally S, et al: Divergent clonal evolution of castration-resistant neuroendocrine prostate cancer. Nat Med. 22:298–305. 2016. View Article : Google Scholar : PubMed/NCBI

106 

Aparicio A, Logothetis CJ and Maity SN: Understanding the lethal variant of prostate cancer: Power of examining extremes. Cancer Discov. 1:466–468. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2021
Volume 21 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Segura-Moreno YY, Sanabria-Salas MC, Varela R, Mesa JA and Serrano ML: Decoding the heterogeneous landscape in the development prostate cancer (Review). Oncol Lett 21: 376, 2021
APA
Segura-Moreno, Y.Y., Sanabria-Salas, M.C., Varela, R., Mesa, J.A., & Serrano, M.L. (2021). Decoding the heterogeneous landscape in the development prostate cancer (Review). Oncology Letters, 21, 376. https://doi.org/10.3892/ol.2021.12637
MLA
Segura-Moreno, Y. Y., Sanabria-Salas, M. C., Varela, R., Mesa, J. A., Serrano, M. L."Decoding the heterogeneous landscape in the development prostate cancer (Review)". Oncology Letters 21.5 (2021): 376.
Chicago
Segura-Moreno, Y. Y., Sanabria-Salas, M. C., Varela, R., Mesa, J. A., Serrano, M. L."Decoding the heterogeneous landscape in the development prostate cancer (Review)". Oncology Letters 21, no. 5 (2021): 376. https://doi.org/10.3892/ol.2021.12637