Inhibitory effects of canagliflozin on pancreatic cancer are mediated via the downregulation of glucose transporter‑1 and lactate dehydrogenase A

  • Authors:
    • Duiyue Xu
    • Yiran Zhou
    • Xin Xie
    • Liangyuan He
    • Jialu Ding
    • Shuyang Pang
    • Baiyong Shen
    • Changlin Zhou
  • View Affiliations

  • Published online on: September 8, 2020     https://doi.org/10.3892/ijo.2020.5120
  • Pages: 1223-1233
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Pancreatic cancer is one of the most lethal solid malignancies, with a poor prognosis and a high mortality rate. Pancreatic cancer cells exhibit enhanced glycolysis to maintain their rapid growth. Canagliflozin (CANA) is a sodium‑glucose co‑transporter 2 inhibitor used for the clinical treatment of diabetes. Recent studies have demonstrated the potential ability of CANA to suppress hepatocellular carcinoma, whereas its therapeutic effects on and mechanisms in pancreatic cancer have rarely been reported. In the present study, the antitumor effects of CANA on pancreatic cancer were investigated. The data obtained indicated that pancreatic cancer growth was effectively suppressed by CANA in a dose‑dependent manner, with peak inhibition rates of 54.3 and 57.6% in cultured Capan‑1 and PANC‑1 cells respectively. The tumor inhibitory rate reached 45.2% in nude mice with PANC‑1‑derived tumors, suggesting its effective antitumor activity against pancreatic cancer in vitro and/or in vivo. In addition, the combined treatment of Capan‑1 and PANC‑1 cells with gemcitabine and CANA exhibited a greater efficacy compared with that of treatment with gemcitabine alone. Moreover, glucose uptake and lactate production were decreased, and the mRNA levels of the glycolysis‑associated genes, including glucose transporter‑1 and lactate dehydrogenase A were decreased, indicating the inhibitory effects caused by the combination treatment on the metabolism of glucose in pancreatic cancer cells. Furthermore, CANA induced apoptosis, notably early apoptosis, and decreased the protein levels of PI3K, p‑AKT, p‑mTOR and HIF‑1α, which indicated that the PI3K/AKT/mTOR signaling pathway was involved in the glycolytic process. These results demonstrated that pancreatic cancer growth was effectively inhibited by CANA via the suppression of glycolysis. This was mediated primarily by the PI3K/AKT/mTOR signaling pathway, revealing the underlying role and potential of this pathway for the clinical treatment of pancreatic cancer. Novel applications for the existing drug CANA can be explored, which could reduce the cost and time required for drug development in the field of drug discovery.
View Figures
View References

Related Articles

Journal Cover

November-2020
Volume 57 Issue 5

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Xu D, Zhou Y, Xie X, He L, Ding J, Pang S, Shen B and Zhou C: Inhibitory effects of canagliflozin on pancreatic cancer are mediated via the downregulation of glucose transporter‑1 and lactate dehydrogenase A. Int J Oncol 57: 1223-1233, 2020
APA
Xu, D., Zhou, Y., Xie, X., He, L., Ding, J., Pang, S. ... Zhou, C. (2020). Inhibitory effects of canagliflozin on pancreatic cancer are mediated via the downregulation of glucose transporter‑1 and lactate dehydrogenase A. International Journal of Oncology, 57, 1223-1233. https://doi.org/10.3892/ijo.2020.5120
MLA
Xu, D., Zhou, Y., Xie, X., He, L., Ding, J., Pang, S., Shen, B., Zhou, C."Inhibitory effects of canagliflozin on pancreatic cancer are mediated via the downregulation of glucose transporter‑1 and lactate dehydrogenase A". International Journal of Oncology 57.5 (2020): 1223-1233.
Chicago
Xu, D., Zhou, Y., Xie, X., He, L., Ding, J., Pang, S., Shen, B., Zhou, C."Inhibitory effects of canagliflozin on pancreatic cancer are mediated via the downregulation of glucose transporter‑1 and lactate dehydrogenase A". International Journal of Oncology 57, no. 5 (2020): 1223-1233. https://doi.org/10.3892/ijo.2020.5120