Poster Presentation Clinical Oncology Society of Australia Annual Scientific Meeting 2024

Epithelial cell adhesion molecule induces induces Wnt receptor transcription to promote colorectal cancer progression (#218)

Han-Chung Wu 1 2 , Sushree Shankar Panda 1 , Chi-Chiu Lee 1 , Chia-Ning Shen 2 3 , Wen-Shan Li 2 4 , Hsing-Pang Hsieh 4 5
  1. Institute of Cellular and Organismic Biology,, Academia Sinica, Taipei, Taiwan
  2. Biomedical Translation Research Center, Academia Sinica, Taiwan
  3. Genomics Research Center, Academia Sinica, Taipei, Taiwan
  4. Institute of Chemistry, Academia Sinica, Taiwan
  5. Institute of Biotech and Pharma Research, National Health Research Institutes, Miaoli County, Taiwan

Epithelial cell adhesion molecule (EpCAM) has been widely studied as a tumor antigen due to its expression in varieties of solid tumors. Moreover, the glycoprotein contributes to critical cancer-associated cellular functionalities via its extracellular (EpEX) and intracellular (EpICD) domains. In colorectal cancer (CRC), EpCAM has been implicated in the Wnt signaling pathway, as EpICD and β-Catenin are coordinately translocated to the nucleus. Once in the nucleus, EpICD transcriptionally regulates EpCAM target genes. Here, we studied the role of EpCAM in colorectal cancer (CRC) stemness. We found that the EpEX-induced Wnt signaling activates TACE and γ-secretase enzymes to augment shedding of EpEX and EpICD, establishing a positive feedback loop. Importantly, we show that the EpICD interacted with the promoters of Wnt receptors (FZD6 and LRP5/6) thus upregulated their transcriptional activity inducing Wnt signaling. Furthermore, activation of Wnt-pathway-associated kinases in the β-Catenin destruction complex (GSK3β and CK1) induced γ-secretase activity to augment EpICD shedding, establishing a positive-feedback loop. Our EpCAM-neutralizing antibody (EpAb2-6) and a porcupine inhibitor (LGK974) each partially attenuated cancer stemness, while their combination abolished stemness-related endpoints, induced apoptosis in vitro and markedly diminished tumor progression in animal models of human CRC. From these findings, we conclude that EpCAM stimulates Wnt signaling to promote cancer stemness, and the combination of EpAb2-6 and porcupine inhibitors may represent an effective CRC treatment, including for KRAS-mutant cancers. Thus, the mechanistic insights gained from our study may be useful to improve existing treatments or to develop novel anticancer therapeutics.