Login / Signup

Interplays of glucose metabolism and KRAS mutation in pancreatic ductal adenocarcinoma.

Yu-Huei LiuChun-Mei HuYuan-Sheng HsuWen-Hwa Lee
Published in: Cell death & disease (2022)
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive and deadliest cancer worldwide. The primary reasons for this are the lack of early detection methods and targeted therapy. Emerging evidence highlights the metabolic addiction of cancer cells as a potential target to combat PDAC. Oncogenic mutations of KRAS are the most common triggers that drive glucose uptake and utilization via metabolic reprogramming to support PDAC growth. Conversely, high glucose levels in the pancreatic microenvironment trigger genome instability and de novo mutations, including KRAS G12D , in pancreatic cells through metabolic reprogramming. Here, we review convergent and diverse metabolic networks related to oncogenic KRAS mutations between PDAC initiation and progression, emphasizing the interplay among oncogenic mutations, glucose metabolic reprogramming, and the tumor microenvironment. Recognizing cancer-related glucose metabolism will provide a better strategy to prevent and treat the high risk PDAC population.
Keyphrases
  • high glucose
  • wild type
  • stem cells
  • endothelial cells
  • blood glucose
  • blood pressure
  • adipose tissue
  • papillary thyroid
  • dna methylation
  • skeletal muscle
  • drug induced