Oral Presentation Advances in Neuroblastoma Research Congress 2016

Whole exome and deep targeted sequencing of clinically aggressive neuroblastomas reveal recurrent somatic mutations in pathways involved in cancer progression (#111)

Vito A Lasorsa 1 , Daniela Formicola 1 , Piero Pignataro 1 , Flora Cimmino 1 , Francesco M Calabrese 2 , Jaume Mora 3 , Maria Rosaria Esposito 4 , Marcella Pantile 4 , Carlo Zanon 4 , Marilena De Mariano 5 , Luca Longo 5 , Michael D Hogarty 6 , Carmen de Torres 3 , Gian Paolo Tonini 4 , Achille Iolascon 1 , Mario Capasso 1
  1. University of Naples Federico II, Napoli, NA, Italy
  2. University of Bari, Bari, Italy
  3. Hospital Sant Joan de Déu, Barcelona
  4. Pediatric Research Institute (IRP) – Fondazione Città della Speranza, Padua, Italy
  5. IRCCS AOU San Martino-IST National Cancer Research Institute, Genoa, Italy
  6. Children’s Hospital of Philadelphia, Philadelphia, USA

The complete spectrum of somatic mutations of the most aggressive forms of neuroblastoma is still to be defined. Here we sought to identify additional potential cancer drivers in high-risk and ultra-high-risk (high-risk patients with any adverse event within 36 months from diagnosis) neuroblastoma.

Whole exome sequencing was performed for 17 ultra-high-risk germline and tumor pairs to identify somatic mutations and deep targeted sequencing of 134 genes selected from the initial screening in an additional set of 48 germline and tumor pairs (62.5% were ultra-high-risk and high-risk), 17 ultra-high-risk tumors and 17 human-derived neuroblastoma cell lines.

Combining both cohorts we found 22 significantly mutated genes, many of which implicated in cancer progression processes. Of these, fifteen (68.2%) were highly expressed in neuroblastoma supporting the biological rationale for their involvement in this malignancy. CHD9, annotated as cancer driver in public databases, was the most significantly altered gene (4.0% of cases) after ALK. Other genes (PTK2, NAV3, NAV1, LRRC17, PXDN, FZD1, ARHGEF10L and ATRX) expressed in neuroblastoma and involved in cell invasiveness and migration were mutated at frequencies between 4% and 2%. Pathways implicated in cell survival, proliferation and motility (focal adhesion and regulation of actin cytoskeleton) were the most frequently disrupted affecting 14.1% of cases, suggesting potential novel therapeutic strategies to prevent disease progression. Rare potentially pathogenic germline variants were significantly enriched in BARD1, CHEK2 and AXIN2.

To conclude, the combination of whole exome and deep targeted sequencing in a discovery and validation cohort experimental design, identified novel cancer genes in clinically aggressive neuroblastomas. Our analyses demonstrate that infrequently mutated genes may have pathway-level implications in leading tumor progression and suggests possible novel strategies for therapeutic interventions in aggressive forms of neuroblastoma.