Abstract

Despite recent progress in the treatment, the outcome of adult acute T-cell lymphoblastic leukemia (T-ALL) is poor. Development of novel approach to combat this disease is urgently required. Vorinostat, a pan-histone deacetylase (HDAC) inhibitor, exerts promising anticancer activity in a variety of solid and hematologic malignancies. However, the efficacy of vorinostat monotherapy is unsatisfactory. Here, we show that quinacrine (QC), an anti-malaria drug with potent autophagy inhibitory activity, could synergistically enhance vorinostat-induced cell death at a non-toxic concentration. Compared to the single treatment, QC plus vorinostat significantly induced apoptosis, disrupted the mitochondrial transmembrane potential, and decreased Mcl-1 and Bcl-2/Bax ratio. Interestingly, the application of QC plus vorinostat resulted in mitophagy blockade, as reflected by the increase in the K63-linked ubiquitination of mitochondria protein and the formation of mitochondrial aggresomes. QC plus vorinostat markedly increased the reactive oxygen species (ROS) level in cells. Moreover, the ROS scavenger N-acetylcysteine (NAC) abrogated QC plus vorinostat-induced ROS, decreased the ubiquitination of mitochondria proteins, and cell death. Finally, using a xenograft mouse model, we demonstrated that QC plus vorinostat significantly reduced cell proliferation and induced cell death in vivo. Taken together, our results showed that the combination of QC with vorinostat may represent a novel regimen for the treatment of T-cell acute lymphoblastic leukemia, which deserves clinical evaluation in the future.

Highlights

  • Introduction AcuteT-cell lymphoblastic leukemia (T-ALL) is an aggressive hematologic disease, accounting for about 15Official journal of the Cell Death Differentiation AssociationJing et al Cell Death and Disease (2018)9:589Histone deacetylases (HDACs), which medicate the deacetylation of histone and non-histone proteins, can regulate the activities of transcriptional factors involved in both cancer initiation and progression as well as the posttranslational modification of numerous key proteins, including tumor suppressor genes[3,4]

  • Because QC was revealed to be a multifunctional drug with anticancer activity, we hypothesized that QC may enhance vorinostat-induced cell death in human T-ALL cell lines

  • We showed that the increase of reactive oxygen species (ROS) and the inhibition of mitophagy contribute to the anti-T-ALL effect of the combination of vorinostat and QC (Fig. 6)

Read more

Summary

Introduction

Introduction AcuteT-cell lymphoblastic leukemia (T-ALL) is an aggressive hematologic disease, accounting for about 15Official journal of the Cell Death Differentiation AssociationJing et al Cell Death and Disease (2018)9:589Histone deacetylases (HDACs), which medicate the deacetylation of histone and non-histone proteins, can regulate the activities of transcriptional factors involved in both cancer initiation and progression as well as the posttranslational modification of numerous key proteins, including tumor suppressor genes[3,4]. T-cell lymphoblastic leukemia (T-ALL) is an aggressive hematologic disease, accounting for about 15. Official journal of the Cell Death Differentiation Association. Jing et al Cell Death and Disease (2018)9:589. Histone deacetylases (HDACs), which medicate the deacetylation of histone and non-histone proteins, can regulate the activities of transcriptional factors involved in both cancer initiation and progression as well as the posttranslational modification of numerous key proteins, including tumor suppressor genes[3,4]. HDACs are considered a promising anticancer drug target[5]. Vorinostat, a pan-HDAC inhibitor, has been approved by Food and. Vorinostat exerts powerful anticancer activity in a variety of solid and hematologic malignancies including. Due to the activation of autophagy and other survival pathways, the anticancer effects of vorinostat are often limited[7]. Combinations of vorinostat with other agents are necessary to improve the efficacy of vorinostat

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