Abstract

L-Asparaginase-II from Escherichia coli (EcA) is a central component in the treatment of acute lymphoblastic leukemia (ALL). However, the therapeutic efficacy of EcA is limited due to immunogenicity and a short half-life in the patient. Here, we performed rational mutagenesis to obtain EcA variants with a potential to improve ALL treatment. Several variants, especially W66Y and Y176F, killed the ALL cells more efficiently than did wild-type EcA (WT-EcA), although nonleukemic peripheral blood monocytes were not affected. Several assays, including Western blotting, annexin-V/propidium iodide binding, comet, and micronuclei assays, showed that the reduction in viability of leukemic cells is due to the increase in caspase-3, cytochrome c release, poly(ADP-ribose) polymerase activation, down-regulation of anti-apoptotic protein Bcl-XL, an arrest of the cell cycle at the G0/G1 phase, and eventually apoptosis. Both W66Y and Y176F induced significantly more apoptosis in lymphocytes derived from ALL patients. In addition, Y176F and Y176S exhibited greatly decreased glutaminase activity, whereas K288S/Y176F, a variant mutated in one of the immunodominant epitopes, showed reduced antigenicity. Further in vivo immunogenicity studies in mice showed that K288S/Y176F was 10-fold less immunogenic as compared with WT-EcA. Moreover, sera obtained from WT-EcA immunized mice and ALL patients who were given asparaginase therapy for several weeks recognized the K288S/Y176F mutant significantly less than the WT-EcA. Further mechanistic studies revealed that W66Y, Y176F, and K288S/Y176F rapidly depleted asparagine and also down-regulated the transcription of asparagine synthetase as compared with WT-EcA. These highly desirable attributes of these variants could significantly advance asparaginase therapy of leukemia in the future.

Highlights

  • The therapeutic potential of Escherichia coli (EcA) is limited due to immunogenicity and short half-life in patients

  • We focused on the engineering of Escherichia coli L-asparaginase isoenzyme II (EcA) that has been used for decades in the treatment of childhood acute lymphoblastic leukemia (ALL), a disease that affects lymphocytes and lymphocyte precursor cells in the bone marrow [1, 2]

  • The level of the 89-kDa fragment significantly increased after treatment with 0.8 units/ml W66Y and K288S/Y176F. These results suggest that several EcA variants, in particular W66Y and K288S/Y176F, strongly induced PARP-mediated apoptosis, leading to cytochrome c release

Read more

Summary

Introduction

The therapeutic potential of EcA is limited due to immunogenicity and short half-life in patients. Several assays, including Western blotting, annexin-V/propidium iodide binding, comet, and micronuclei assays, showed that the reduction in viability of leukemic cells is due to the increase in caspase-3, cytochrome c release, poly(ADP-ribose) polymerase activation, down-regulation of anti-apoptotic protein Bcl-XL, an arrest of the cell cycle at the G0/G1 phase, and eventually apoptosis Both W66Y and Y176F induced significantly more apoptosis in lymphocytes derived from ALL patients. Further mechanistic studies revealed that W66Y, Y176F, and K288S/Y176F rapidly depleted asparagine and down-regulated the transcription of asparagine synthetase as compared with WT-EcA These highly desirable attributes of these variants could significantly advance asparaginase therapy of leukemia in the future

Objectives
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