Back to Journals » Drug Design, Development and Therapy » Volume 11

Pharmacokinetic interactions between glimepiride and rosuvastatin in healthy Korean subjects: does the SLCO1B1 or CYP2C9 genetic polymorphism affect these drug interactions? Observations and introspection of the bioanalysis

Authors Thakkar D, Dash RP

Received 15 March 2017

Accepted for publication 16 March 2017

Published 20 April 2017 Volume 2017:11 Pages 1263—1265

DOI https://doi.org/10.2147/DDDT.S137116

Checked for plagiarism Yes

Editor who approved publication: Professor Anastasios Lymperopoulos



Disha Thakkar,1 Ranjeet Prasad Dash2,3

1Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat, India; 2Drug Metabolism and Pharmacokinetics, Johns Hopkins Drug Discovery Program, Johns Hopkins University, 3Department of Neurology, Johns Hopkins University, Baltimore, MD, USA                                                                                                 

The topic of polymorphism in drug metabolizing enzymes and drug transporters with their impact on pharmacotherapy is of great interest. The clinical relevance depends on the vectorial movement, the therapeutic index of the substrates and inherent interindividual variability.1 With respect to the variability, various polymorphisms associated with the drug transporters have been reported that led to the alteration in the pharmacokinetic and pharmacodynamic profile of the drugs.2 Many preclinical and clinical studies have provided the evidence for the application of genetic information for the development of individualized therapies.3                                                                    


Author’s reply

Min Soo Park1,2


1
Department of Clinical Pharmacology, Severance Hospital, 2Department of Pediatrics, Yonsei University College of Medicine, Seoul, South Korea


First of all, we would like to thank Thakkar and Dash for their interest in our paper “Pharmacokinetic interactions between glimepiride and rosuvastatin in healthy Korean subjects: does the SLCO1B1 or CYP2C9 genetic polymorphism affect these drug interactions?” Regarding their concerns about the sample processing of rosuvastatin and N-desmethyl rosuvastatin, they seem to have misunderstood because detailed contents were missed.

View the original paper by Kim CO and colleagues.

 

Dear editor

The topic of polymorphism in drug metabolizing enzymes and drug transporters with their impact on pharmacotherapy is of great interest. The clinical relevance depends on the vectorial movement, the therapeutic index of the substrates and inherent interindividual variability.1 With respect to the variability, various polymorphisms associated with the drug transporters have been reported that led to the alteration in the pharmacokinetic and pharmacodynamic profile of the drugs.2 Many preclinical and clinical studies have provided the evidence for the application of genetic information for the development of individualized therapies.3

Recently, Kim et al demonstrated a well-planned and meticulously executed clinical investigation to understand the role of SLCO1B1 or CYP2C9 genetic polymorphism on the pharmacokinetics of glimepiride and rosuvastatin in healthy Korean subjects.4 Most importantly, the pharmacokinetic interaction study consisted of two parts: parts 1 and 2. Part 1 was designed to evaluate the effect of rosuvastatin on the pharmacokinetics of glimepiride and part 2 was designed to determine the effect of glimepiride on the pharmacokinetics of rosuvastatin. Blood samples were collected at predetermined time points and samples were analyzed using liquid chromatography-mass spectrometer to determine the drug concentration in the plasma samples.4

The intent of this letter is to highlight the concerns related to the sample processing method adopted by the authors for bioanalysis. For analysis of rosuvastatin and N-desmethyl rosuvastatin, the authors took 50 μL of plasma and mixed with 10 μL of valsartan, an internal standard (100 ng/mL in 50% acetonitrile).4 The mixture was centrifuged, followed by the collection of supernatant, which was evaporated till dryness and reconstituted in 200 μL of 50% acetonitrile for analysis.4 From the process detailed by the authors, it is evident that the authors wanted to deproteinize the plasma samples using 50% acetonitrile containing the internal standard. However, the concern is the amount and concentration of acetonitrile used for protein precipitation. An ideal amount for organic solvents (at 100% concentration) used for deproteinization should be more than 3–4 volumes of the plasma sample to achieve optimum precipitation.5,6 However, the authors have used only 10 μL of organic solvent (one-fifth of the plasma volume) at 50% concentration4, which raises the concern regarding the insufficient protein precipitation and the amount recovered for the subsequent process. The same sample extraction process has been adopted for glimepiride as well.

Yet another concern is the column temperature mentioned in the bioanalytical method to be 500°C,4 which is expected to be too high for any kind of chromatographic columns. Do the authors want to say 50°C or are they indicating about the ion-source temperature of the mass spectrometer as 500°C? Although the authors have tried to understand the mechanistic aspects of genetic polymorphism on pharmacokinetic interaction of rosuvastatin and glimepiride, the bioanalytical method adopted for pharmacokinetic studies needs further clarification.

This communication was prepared to promote an exchange of thoughts on a topic of great interest in genetic polymorphism and pharmacokinetic interactions.

Disclosure

The authors report no conflicts of interest in this communication.


References

1.

Ieiri I, Takane H, Hirota T, Otsubo K, Higuchi S. Genetic polymorphisms of drug transporters: pharmacokinetic and pharmacodynamic consequences in pharmacotherapy. Expert Opin Drug Metab Toxicol. 2006;2(5):651–674.

2.

Fukasawa T, Suzuki A, Otani K. Effects of genetic polymorphism of cytochrome P450 enzymes on the pharmacokinetics of benzodiazepines. J Clin Pharm Ther. 2007;32(4):333–341.

3.

Wei CY, Lee MT, Chen YT. Pharmacogenomics of adverse drug reactions: implementing personalized medicine. Hum Mol Genet. 2012;21(R1):R58–R65.

4.

Kim CO, Oh ES, Kim H, Park MS. Pharmacokinetic interactions between glimepiride and rosuvastatin in healthy Korean subjects: does the SLCO1B1 or CYP2C9 genetic polymorphism affect these drug interactions? Drug Des Devel Ther. 2017;11:503–512.

5.

[no authors listed]. Mass Spectrometry in Bioanalysis. Particle Sciences – Technical Brief. 2009;4:1–2. Available from: http://www.particlesciences.com/news/technical-briefs/2009/mass-spectrometry-bioanalysis.html Accessed March 14, 2017.

6.

Pucci V, Di Palma S, Alfieri A, Bonelli F, Monteagudo E. A novel strategy for reducing phospholipids-based matrix effect in LC-ESI-MS bioanalysis by means of HybridSPE. J Pharm Biomed Anal. 2009;50(5):867–871.

Author’s reply

Min Soo Park1,2

1Department of Clinical Pharmacology, Severance Hospital, 2Department of Pediatrics, Yonsei University College of Medicine, Seoul, South Korea

Correspondence: Min Soo Park, Department of Clinical Pharmacology, Severance Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea, Email [email protected]

Dear editor

First of all, we would like to thank Thakkar and Dash for their interest in our paper “Pharmacokinetic interactions between glimepiride and rosuvastatin in healthy Korean subjects: does the SLCO1B1 or CYP2C9 genetic polymorphism affect these drug interactions?” Regarding their concerns about the sample processing of rosuvastatin and N-desmethyl rosuvastatin, they seem to have misunderstood because detailed contents were missed. As mentioned by Thakkar and Dash, higher amounts of organic solvents are used for deproteinization. Before the mixture of plasma and internal standard was centrifuged, we added 800 μL of 0.1% formic acid – diethyl ether (1:7, v/v) to deproteinize. We consider that our sample extraction process for rosuvastatin and N-desmethyl rosuvastatin analysis was appropriate because organic solvents were added at more than 10 times the volume of the plasma sample.

Similar to the sample processing of rosuvastatn and N-desmethyl rosuvastatin, 800 μL of 0.1% formic acid – diethyl ether (1:7, v/v) were added to the mixture with plasma and internal standard for glimepiride analysis, and then the mixture was centrifuged. After centrifugation, 100 μL of supernatant was diluted with 100 μL of water, and 10 μL of the resultant solution was injected directly into the column heated at 40°C not 500°C. Unfortunately, we confirm that the column temperature for glimepiride assay was a typo. We are pleased to be able to correct our paper.

Disclosure

The authors report no conflicts of interest in this communication.

Dove Medical Press encourages responsible, free and frank academic debate. The content of the Drug Design, Development and Therapy ‘letters to the editor’ section does not necessarily represent the views of Dove Medical Press, its officers, agents, employees, related entities or the Drug Design, Development and Therapy editors. While all reasonable steps have been taken to confirm the content of each letter, Dove Medical Press accepts no liability in respect of the content of any letter, nor is it responsible for the content and accuracy of any letter to the editor.

Creative Commons License © 2017 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.