Abstract

Simple SummaryThe antimalarial drug artesunate also has anticancer activity. Based on what is known about how artesunate works in malaria, we hypothesized that the kelch-like ECH-associated protein 1 (KEAP1)/nuclear factor erythroid 2-related factor 2 (NRF2) pathway, which is often mutated in non-small-cell lung cancer (NSCLC), would play an important part in determining the sensitivity of NSCLC cell lines to artesunate. We treated cells with increasing doses of artesunate and showed that a cell line with an inactivating mutation in KEAP1 was less responsive to artesunate. Additionally, by modulating the KEAP1/NRF2 pathway we were able to alter the sensitivity of lung cancer cells to artesunate. Taken together, these findings demonstrate that KEAP1 is required for the anticancer activity of artesunate and support the further development of the combination of artesunate and NRF2 inhibitors to treat NSCLC, especially when patients have a mutation in the KEAP1/NRF2 pathway.Artesunate is the most common treatment for malaria throughout the world. Artesunate has anticancer activity likely through the induction of reactive oxygen species, the same mechanism of action utilized in Plasmodium falciparum infections. Components of the kelch-like ECH-associated protein 1 (KEAP1)/nuclear factor erythroid 2-related factor 2 (NRF2) pathway, which regulates cellular response to oxidative stress, are mutated in approximately 30% of non-small-cell lung cancers (NSCLC); therefore, we tested the hypothesis that KEAP1 is required for artesunate sensitivity in NSCLC. Dose response assays identified A549 cells, which have a G333C-inactivating mutation in KEAP1, as resistant to artesunate, with an IC50 of 23.6 µM, while H1299 and H1563 cells were sensitive to artesunate, with a 10-fold lower IC50. Knockdown of KEAP1 through siRNA caused increased resistance to artesunate in H1299 cells. Alternatively, the pharmacological inhibition of NRF2, which is activated downstream of KEAP1 loss, by ML385 partially restored sensitivity of A549 cells to artesunate, and the combination of artesunate and ML385 was synergistic in both A549 and H1299 cells. These findings demonstrate that KEAP1 is required for the anticancer activity of artesunate and support the further development of NRF2 inhibitors to target patients with mutations in the KEAP1/NRF2 pathway.

Highlights

  • Artesunate, first discovered in 1972 by the Chinese scientist Tu Youyou, is a watersoluble semi-synthetic derivative of artemisinin [1]

  • We first investigated the effect of artesunate on three non-small-cell lung cancer (NSCLC) cell lines (A549, H1299, and H1563) by assessing cell viability, using a CellTiter-Glo 2.0 assay, after treatment

  • A549 cells were resistant to artesunate, with a mean IC50 of 23.63 μM ± 8.886 μM from three independent experiments, while H1299 and H1563 cells were sensitive to artesunate, with mean IC50s of 2.36 μM ± 1.275 μM and 3.43 μM ± 1.190 μM, respectively, (Figure 1b)

Read more

Summary

Introduction

Artesunate, first discovered in 1972 by the Chinese scientist Tu Youyou, is a watersoluble semi-synthetic derivative of artemisinin [1]. This Nobel Prize-winning medication has become the WHO-recommended first-line treatment for severe malaria [2]. Artemisinin and its derivative, artesunate, are obtained as extracts from Artemisia annua, or the sweet wormwood plant [3]. In addition to antimalarial activity, artesunate has potential anticancer effects and proposed mechanisms include induction of apoptosis, inhibition of angiogenesis, inhibition of hypoxia-inducible factor-1α (HIF-1α) activation, and direct DNA injury. Artesunate has been clinically investigated in combination with cisplatin and vinorelbine in patients with advanced non-small-cell lung cancer (NSCLC). Both the disease control rate and the time to tumor progression were significantly improved for the artesunate and chemotherapy group when compared to the chemotherapy alone group with no significant differences in toxicity [5]

Objectives
Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call