Molecular Oncology (Nov 2023)

Canagliflozin mediates tumor suppression alone and in combination with radiotherapy in non‐small cell lung cancer (NSCLC) through inhibition of HIF‐1α

  • Olga‐Demetra Biziotis,
  • Evangelia Evelyn Tsakiridis,
  • Amr Ali,
  • Elham Ahmadi,
  • Jianhan Wu,
  • Simon Wang,
  • Bassem Mekhaeil,
  • Kanwaldeep Singh,
  • Gabe Menjolian,
  • Thomas Farrell,
  • Bassam Abdulkarim,
  • Ranjan K. Sur,
  • Aruz Mesci,
  • Peter Ellis,
  • Tobias Berg,
  • Jonathan L Bramson,
  • Paola Muti,
  • Gregory R Steinberg,
  • Theodoros Tsakiridis

DOI
https://doi.org/10.1002/1878-0261.13508
Journal volume & issue
Vol. 17, no. 11
pp. 2235 – 2256

Abstract

Read online

Non‐small cell lung cancer (NSCLC) has a poor prognosis, and effective therapeutic strategies are lacking. The diabetes drug canagliflozin inhibits NSCLC cell proliferation and the mammalian target of rapamycin (mTOR) pathway, which mediates cell growth and survival, but it is unclear whether this drug can enhance response rates when combined with cytotoxic therapy. Here, we evaluated the effects of canagliflozin on human NSCLC response to cytotoxic therapy in tissue cultures and xenografts. Ribonucleic acid sequencing (RNA‐seq), real‐time quantitative PCR (RT‐qPCR), metabolic function, small interfering ribonucleic acid (siRNA) knockdown, and protein expression assays were used in mechanistic analyses. We found that canagliflozin inhibited proliferation and clonogenic survival of NSCLC cells and augmented the efficacy of radiotherapy to mediate these effects and inhibit NSCLC xenograft growth. Canagliflozin treatment alone moderately inhibited mitochondrial oxidative phosphorylation and exhibited greater antiproliferative capacity than specific mitochondrial complex‐I inhibitors. The treatment downregulated genes mediating hypoxia‐inducible factor (HIF)‐1α stability, metabolism and survival, activated adenosine monophosphate‐activated protein kinase (AMPK) and inhibited mTOR, a critical activator of hypoxia‐inducible factor‐1α (HIF‐1α) signaling. HIF‐1α knockdown and stabilization experiments suggested that canagliflozin mediates antiproliferative effects, in part, through suppression of HIF‐1α. Transcriptional regulatory network analysis pinpointed histone deacetylase 2 (HDAC2), a gene suppressed by canagliflozin, as a key mediator of canagliflozin's transcriptional reprogramming. HDAC2 knockdown eliminated HIF‐1α levels and enhanced the antiproliferative effects of canagliflozin. HDAC2‐regulated genes suppressed by canagliflozin are associated with poor prognosis in several clinical NSCLC datasets. In addition, we include evidence that canagliflozin also improves NSCLC response to chemotherapy. In summary, canagliflozin may be a promising therapy to develop in combination with cytotoxic therapy in NSCLC.

Keywords