Page 70 - Read Online
P. 70

Fraser. J Transl Genet Genom 2018;2:21. I  https://doi.org/10.20517/jtgg.2018.27                                                        Page 7 of 15

               Table 2. Summary of whole genome sequencing studies of localized prostate cancer
                Ref.                 Number and type of whole              Major significance
                                       genomes sequenced
                Berger et al. [69] , 2011  7 primary high risk tumours  First whole-genome sequencing of localized prostate cancer. Identification of
                                                          closed-loop chain rearrangements
                Baca et al. , 2012  55 primary tumours, 2 neuroendo-  Characterization of temporal changes in prostate cancer structural variation
                      [71]
                                   crine metastases       (“chromoplexy”)
                Weischenfeldt et al. [72] , 2013 11 early-onset primary tumours  Androgen-dependent structural variation enriched in prostate cancers aris-
                                   7 elderly-onset primary tumours  ing in men < 50 years of age
                Cancer Genome Atlas Re-  19 primary tumours  Molecular subclasses of localized prostate cancers
                search Network [29] , 2015
                Boutros et al. [49] , 2015  23 malignant foci from 5 primary   Spatial heterogeneity of localized prostate cancer
                                   tumours
                Cooper et al. [77] , 2015  12 malignant foci from 3 primary   Spatial-temporal heterogeneity of localized prostate cancer. Identification of
                                   tumours                aberrations in morphologically-normal prostate epithelium
                Fraser et al. [10] , 2017  200 intermediate risk primary   Largest study of prostate cancer whole genomes to date. Identification of
                                   tumours                recurrent driver aberrations linked to adverse clinical outcome
                Taylor et al. , 2017  19 disease foci from 14 germline   Tumour genomes of BRCA2 mutation carriers closely resemble those of
                       [15]
                                   BRCA2 mutation carriers  castration-resistant metastatic disease. MED12/MED12L pathway as driver
                                                          of clinical aggression
                Camacho et al. [88] , 2017  103 primary tumours  Assessment of somatic genome-wide copy number aberrations and mecha-
                                                          nism of copy number loss
                Ren et al. [89] , 2018  65 primary tumours from Chinese   Low frequency of TMPRSS2:ERG fusion in Chinese prostate cancers. Identifi-
                                   men                    cation of novel tumour suppressor genes
                Espiritu et al. , 2018  93 intermediate risk primary   Analysis of the temporal evolution of prostate cancer. Development of a
                        [9]
                                   tumours                clonality-aware multi-modal biomarker of adverse clinical outcome
                Wedge et al. [79] , 2018  87 primary tumours, 20 metastatic  Temporal evolution of prostate cancer. Identification of potential druggable
                                   lesions                targets in localized disease
                Su et al. [84] , 2018  17 nuclei from 2 primary tumours  First report of single nucleus whole-genome sequencing in prostate cancer.
                                                          Significant spatial heterogeneity within the same gland

                                                   [71]
               A subsequent study from the same group  showed that these complex closed-loop chain rearrangements
               occur through a process termed “chromoplexy”. Using simulated genome data, the authors demonstrated
               that chromoplexy is an important mechanism of tumour suppressor gene inactivation in prostate cancer,
               and further demonstrated that chromoplexy occurs throughout the subclonal evolution of prostate cancer in
               a “punctuated equilibrium” of tumour evolution.

               The Cancer Genome Atlas (TCGA) program sequenced the whole exomes of 333 primary prostate cancers,
                                                                         [29]
               with whole-genome sequencing of 19 cases of low mutational burden , representing the first well-powered
               survey of protein coding variants in localized prostate cancer. Using associated CNA, RNA expression, DNA
               methylation, and protein phosphorylation arrays, the TCGA team identified several consensus clusters of
               localized prostate cancer, including tumours harbouring an ETS fusion (~60%) and SPOP, FOXA1, or IDH1
               mutation (~15%, collectively). Approximately 25% of localized prostate cancers did not sort into one of these
               clusters, perhaps suggesting the existence of additional molecular subtypes that could not be detected given
               the statistical power of the TCGA study and the relatively low mutational burden in localized prostate can-
               cer. While TCGA identified molecular aberrations in several clinically-relevant pathways (e.g., PI3K, DNA
               repair, and other), the putative effects of these aberrations on clinical outcome could not be assessed due to
               the limited clinical follow up associated with the genomic findings. One potential clinical utility of Weisch-
                          [72]
               enfeldt et al.  surveyed the whole genomes of eleven prostate cancers arising in men 50 years of age or
               younger and compared these with seven propensity-matched prostate cancers of elderly-onset. While rare,
               these cancers in younger men represent a unique clinical challenge, and both the long natural life expectan-
               cy of these men and the increased clinical aggression of these early-onset cancers necessitates definitive local
               therapy. Interestingly, these cancers harbour a lower overall burden of structural variation, perhaps owing to
               the reduced time for acquisition of mutations, but possess a strong bias toward androgen-induced GRs - such
               as T2E - relative to cancers arising in older men. This was associated with increased AR mRNA abundance
               in early-onset prostate cancer. These findings suggest a unique biology of early-onset prostate cancer and
   65   66   67   68   69   70   71   72   73   74   75