Abstract

Cancer cells show higher levels of reactive oxygen species (ROS) than normal cells and increasing intracellular ROS levels are becoming a recognized strategy against tumor cells. Thus, diminishing ROS levels could be also detrimental to cancer cells. We surmise that avoiding ROS generation would be a better option than quenching ROS with antioxidants. Chronic myelogenous leukemia (CML) is triggered by the expression of BCR-ABL kinase, whose activity leads to increased ROS production, partly through NADPH oxidases. Here, we assessed NADPH oxidases as therapeutic targets in CML. We have analyzed the effect of different NADPH oxidase inhibitors, either alone or in combination with BCR-ABL inhibitors, in CML cells and in two different animal models for CML. NADPH oxidase inhibition dramatically impaired the proliferation and viability of BCR-ABL-expressing cells due to the attenuation of BCR-ABL signaling and a pronounced cell-cycle arrest. Moreover, the combination of NADPH oxidase inhibitors with BCR-ABL inhibitors was highly synergistic. Two different animal models underscore the effectiveness of NADPH oxidase inhibitors and their combination with BCR-ABL inhibitors for CML targeting in vivo. Our results offer further therapeutic opportunities for CML, by targeting NADPH oxidases. In the future, it would be worthwhile conducting further experiments to ascertain the feasibility of translating such therapies to clinical practice.

Highlights

  • Chronic myelogenous leukemia (CML) is characterized by the t(9; 22) q(34; q11) translocation encoding the BCRABL oncoprotein, which triggers the chronic phase of the disease [1]

  • Two different animal models underscore the effectiveness of NADPH oxidase inhibitors and their combination with BCR-ABL inhibitors for CML targeting in vivo

  • Similar results were found in acute myelogenous leukemia (AML), and we suggest that this strategy could be applied to other kinase-driven cancers

Read more

Summary

Introduction

Chronic myelogenous leukemia (CML) is characterized by the t(9; 22) q(34; q11) translocation encoding the BCRABL oncoprotein, which triggers the chronic phase of the disease [1]. Each NADPH oxidase seems to have a particular pattern of expression, and most cells express different members of this family [20] The importance of these enzymes in cancer is beginning to be recognized [21] and, interestingly, it has recently been shown that NADPH oxidase inhibitors are efficient at preventing the growth of colon cancer xenografts [22]. This suggests that NADPH oxidases could be exploited as therapeutic targets in the cancer treatment setting

Methods
Results
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