Abstract

Busulphan (Bu) is an alkylating agent used in the conditioning regimen prior to hematopoietic stem cell transplantation (HSCT). Bu is extensively metabolized in the liver via conjugations with glutathione to form the intermediate metabolite (sulfonium ion) which subsequently is degraded to tetrahydrothiophene (THT). THT was reported to be oxidized forming THT-1-oxide that is further oxidized to sulfolane and finally 3-hydroxysulfolane. However, the underlying mechanisms for the formation of these metabolites remain poorly understood. In the present study, we performed in vitro and in vivo investigations to elucidate the involvement of flavin-containing monooxygenase-3 (FMO3) and cytochrome P450 enzymes (CYPs) in Bu metabolic pathway. Rapid clearance of THT was observed when incubated with human liver microsomes. Furthermore, among different recombinant microsomal enzymes, the highest intrinsic clearance for THT was obtained via FMO3 followed by several CYPs including 2B6, 2C8, 2C9, 2C19, 2E1 and 3A4. In Bu- or THT-treated mice, inhibition of FMO3 by phenylthiourea significantly suppressed the clearance of both Bu and THT. Moreover, the simultaneous administration of a high dose of THT (200μmol/kg) to Bu-treated mice reduced the clearance of Bu. Consistently, in patients undergoing HSCT, repeated administration of Bu resulted in a significant up-regulation of FMO3 and glutathione-S-transfrase -1 (GSTA1) genes. Finally, in a Bu-treated patient, additional treatment with voriconazole (an antimycotic drug known as an FMO3-substrate) significantly altered the Bu clearance. In conclusion, we demonstrate for the first time that FMO3 along with CYPs contribute a major part in busulphan metabolic pathway and certainly can affect its kinetics. The present results have high clinical impact. Furthermore, these findings might be important for reducing the treatment-related toxicity of Bu, through avoiding interaction with other concomitant used drugs during conditioning and hence improving the clinical outcomes of HSCT.

Highlights

  • Hematopoietic stem cell transplantation (HSCT) is a curative treatment for several malignant and non-malignant disorders including leukemias, aplastic anemia, thalassemia and inborn errors of metabolism

  • THT is metabolized by microsomal enzymes with flavin-containing monooxygenase-3 (FMO3) as the main enzyme, but with contribution from cytochrome P450 enzymes (CYPs)

  • We have shown that both FMO3 and, to less extent, CYPs are responsible for THT oxidation to THT 1-oxide, may play a role in the downstream oxidation of THT 1-oxide

Read more

Summary

Introduction

Hematopoietic stem cell transplantation (HSCT) is a curative treatment for several malignant and non-malignant disorders including leukemias, aplastic anemia, thalassemia and inborn errors of metabolism. A conditioning regimen is an important treatment prior to HSCT and consists of cytostatics with or without combination with total body irradiation (TBI). Cytostatics are non-selective drugs which eradicate malignant cells as well as normal cells [1]. Conditioning contributes to elimination of the malignant cells, provides a free space for the donor cells and suppresses the host immune system in order to facilitate engraftment and to avoid graft rejection. Busulphan (Bu) is an alkylating agent administered at high doses prior to HSCT. It reacts with DNA to form intra-strand crosslinks via the guanine and adenine nucleotide base pairs. DNA crosslinking causes permanent cell damage and triggers apoptosis

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