Poster Presentation 32nd Lorne Cancer 2020

The tumor immune microenvironment of germline BRCA1/2 and sporadic prostate cancer (#360)

Anna S Trigos 1 , Anu Pasam 1 , Patricia D Banks 1 , Roslyn Wallace 1 , Simon Keam 1 , Heather Thorne 1 , kConFab kConFab 1 , Catherine Mitchell 1 , David Clouston 2 , Declan G Murphy 1 , Nathan Lawrentschuk 1 , Damien M Bolton 3 , Daniel Moon 4 , Elena Castro 5 , David Olmos 5 , Phil K Darcy 1 , Ygal Haupt 1 , Scott Williams 1 , David L Goode 1 , Paul Neeson 1 , Shahneen K Sandhu 1
  1. Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
  2. TissuPath, Melbourne, Vic, Australia
  3. Austin Hospital, Melbourne, VIC, Australia
  4. Epworth Healthcare, Melbourne, VIC, Australia
  5. Spanish National Cancer Research Centre, Madrid, Spain

Prostate cancer (PC) is considered an immunologically ‘cold’ tumor and therefore patients are generally not considered good candidates for immunotherapy. Tumors arising in germline (g) BRCA1/2 mutation carriers are associated with higher genomic instability and levels of immune infiltration in breast and ovarian cancer. We investigated how germline mutations in DNA repair genes affect the tumor immune microenvironment (TME) of PC.

Archival primary tumor samples from 26 patients with gBRCA2 mutations, 5 with gBRCA1, 5 with mutations in other DNA repair genes (ATMCHEK2FANCIPALB2 or BRCA2+MSH2), and 26 sporadic patients were analyzed. OPAL multiplex immunohistochemistry was used to detect 7 markers (CD3, CD4, CD8, FOXP3, PDL1, AMACR, DAPI) and identify immune subsets. Novel computational distance-based methods to characterize the spatial distribution of cells were developed. Gene expression was evaluated with the Nanostring immune gene panel.

gBRCA1/2 carriers showed lower levels of T cells (9.73% of the tumor stroma) compared to sporadic tumors (14.8%). In both cohorts the T cell population was dominated by CD4+ cells (69.5%), with CD8+ cells representing only 25.6%. Sporadic PCs displayed aggregation of T cells into large clusters in the stroma dominated by CD4+ cells and few CD8+ cells, while 77% of high-grade gBRCA2 patients were enriched in high levels of free, non-aggregated CD8+ T-cells in the tumor area. HLA-A expression was 2.37 times higher in gBRCA2 patients (= 3.4x10-8). gBRCA2 patients with larger T-cell aggregates had an overall poorer prognosis compared to patients without (time to metastasis 55.1 vs. 85.3 months, survival time 67.3 vs. 85.0 months).

gBRCA2 carriers displayed higher levels of HLA-A and higher number of free CD8+ T cells infiltrating tumor regions, suggesting an immune system that could potentially be harnessed. The degree of clustering of T cells (free vs. aggregated) within the TME may provide valuable prognostic information that warrants further validation.