Whole-exome sequencing analyses of inflammatory bowel Disease− Associated colorectal cancers

AI Robles, G Traverso, M Zhang, NJ Roberts, MA Khan… - Gastroenterology, 2016 - Elsevier
AI Robles, G Traverso, M Zhang, NJ Roberts, MA Khan, C Joseph, GY Lauwers, FM Selaru
Gastroenterology, 2016Elsevier
Background & Aims A long duration of inflammatory bowel disease (IBD) increases the risk
for colorectal cancer. Mutation analysis of limited numbers of genes has indicated that
colorectal tumors that develop in patients with IBD differ from those of patients without IBD.
We performed whole-exome sequencing analyses to characterize the genetic landscape of
these tumors. Methods We collected colorectal tumor and non-neoplastic tissues from 31
patients with IBD and colorectal cancer (15 with ulcerative colitis, 14 with Crohn's disease …
Background & Aims
A long duration of inflammatory bowel disease (IBD) increases the risk for colorectal cancer. Mutation analysis of limited numbers of genes has indicated that colorectal tumors that develop in patients with IBD differ from those of patients without IBD. We performed whole-exome sequencing analyses to characterize the genetic landscape of these tumors.
Methods
We collected colorectal tumor and non-neoplastic tissues from 31 patients with IBD and colorectal cancer (15 with ulcerative colitis, 14 with Crohn’s disease, and 2 with indeterminate colitis) and performed whole-exome sequencing analyses of the microdissected tumor and matched nontumor tissues. We identified somatic alterations by comparing matched specimens. The prevalence of mutations in sporadic colorectal tumors was obtained from previously published exome-sequencing studies.
Results
Two specimens had somatic mutations in the DNA proofreading or mismatch repair genes POLE, MLH1, and MSH6 and the tumor cells had a hypermutable phenotype. The remaining tumors had, on average, 71 alterations per sample. TP53 was the most commonly mutated gene, with prevalence similar to that of sporadic colorectal tumors (63% of cases). However, tumors from the patients with IBD had a different mutation spectrum. APC and KRAS were mutated at significantly lower rates in tumors from patients with IBD than in sporadic colorectal tumors (13% and 20% of cases, respectively). Several genes were mutated more frequently or uniquely in tumors from patients with IBD, including SOX9 and EP300 (which encode proteins in the WNT pathway), NRG1 (which encodes an ERBB ligand), and IL16 (which encodes a cytokine). Our study also revealed recurrent mutations in components of the Rho and Rac GTPase network, indicating a role for noncanonical WNT signaling in development of colorectal tumors in patients with IBD.
Conclusions
Colorectal tumors that develop in patients with IBD have distinct genetic features from sporadic colorectal tumors. These findings could be used to develop disease-specific markers for diagnosis and treatment of patients with IBD and colorectal cancer.
Elsevier