Page 64 - Read Online
P. 64

de Kouchkovsky et al. J Transl Genet Genom 2021;5:265-77  https://dx.doi.org/10.20517/jtgg.2021.32                              Page 269

               Table 1. Summary of key genomic alterations associated with t-SCNC
                Genes       Summary of findings                                         Comments
                RB1/TP53    RB1 loss and inactivating TP53 mutations are key genomic hallmarks of t-SCNC. Their frequency  Combined RB1 loss and inactivating TP53 mutations have been implicated with ADT resistance
                            increases as PC progresses from primary adenocarcinoma to mCRPC-adenocarcinoma. Both   and lineage plasticity through EZH2-dependent epigenetic reprogramming and upregulation of
                                                      [3,11]                                                       [23-26]
                            alterations are further enriched in t-SCNC                  the “stemness” transcriptional factor SOX2
                AR/AR enhancer/  t-SCNC is associated with a relative paucity of genomic alterations (e.g., amplifications,   Specific R219 mutations in the AR coactivator and pioneer transcription factor FOXA1 have been
                                                                         [3,14]                                              [30]
                AR co-activators  activating point mutations) involving the AR gene locus and AR enhancer  associated with upregulation of EMT programs and t-SCNC
                SETD2, CYLD  Loss of SETD2 and CYLD has also been associated with t-SCNC [3]  Genomic loss of CYLD is detected in up to half of t-SCNC cases and associated with decreased
                                                                                                      [3]
                                                                                        canonical AR signaling
               t-SCNC: Treatment-associated small cell neuroendocrine prostate cancer; PC: prostate cancer; mCRPC: metastatic castration-resistant prostate cancer; ADT: androgen deprivation therapy; EZH2: enhancer of zeste
               homolog 2; AR: androgen receptor; EMT: epithelial-mesenchymal-transition.


                                                                                                            [40]
               mediated in part by lysine-specific demethylase 1 (LSD1), thereby altering chromatin structure and accessibility .
               MYC (encoded by the MYCN proto-oncogene) is another key transcription factor upregulated in t-SCNC. In normal human development, N-MYC promotes
               the proliferation of neuronal progenitor cells and plays a critical role in organogenesis. MYCN upregulation has been implicated in the development of various
               malignancies and is associated with shortened overall survival in prostate cancer patients . MYCN expression is upregulated in RB1 deficient LNCaP cell
                                                                                            [41]
               lines  and in t-SCNC samples  and is thought to play a direct causal role in the emergence of neuroendocrine differentiation. Indeed, N-MYC has been
                                          [42]
                   [21]
               shown to cooperate with EZH2 to downregulate AR transcriptional programs, and forced MYCN expression in PTEN deficient PC xenografts recapitulates a
               poorly-differentiated carcinoma with variable AR expression levels and epithelial-mesenchymal transition phenotype . MYCN expression has also been
                                                                                                                     [42]
               implicated in increased resistance to AR-targeted therapy, in part through upregulation of DNA damage response pathway genes such as ATM [42-44] .
               Interestingly, N-MYC transcriptional activity appears to be modulated by circulating androgen levels. Thus, following androgen depletion, a shift in the N-
               MYC cistrome (i.e., genome-wide binding sites) towards neural development and lineage pathways genes (e.g., SOX2, CHGA) has been described . This
                                                                                                                                              [42]
               mechanism offers a potential link between exposure to androgen deprivation therapy and the emergence of t-SCNC.

               Forkhead box A1 (encoded by the FOXA1 gene) is a pioneer transcription factor and important AR co-regulator. It is the third most commonly altered gene in
               prostate cancer, with gene rearrangements or point mutations detected in up to a third of patients with mCRPC [30,45] . Canonical FOXA1 signaling maintains
               epithelial differentiation in prostate and prostate cancer cells, in part through the transcriptional suppression of interleukin-8 - which has itself been implicated
                                                                                                            [46]
               in neuroendocrine differentiation via upregulation of enolase 2 expression and the MAPK/ERK pathway . Conversely, FOXA1 knockdown in LNCaP
               prostate adenocarcinoma cells induces neuroendocrine differentiation, and FOXA1 gene expression is significantly lower among t-SCNC samples when
               compared to prostatic adenocarcinoma . However, the exact role of FOXA1 signaling in lineage plasticity and neuroendocrine differentiation has not been
                                                 [46]
               fully established, as some evidence suggests that FOXA1 reprogramming - rather than downregulation - contributes to the emergence of t-SCNC. Mutations in
   59   60   61   62   63   64   65   66   67   68   69