Elsevier

Biochemical Pharmacology

Volume 91, Issue 2, 15 September 2014, Pages 242-248
Biochemical Pharmacology

CYP2C19 polymorphisms account for inter-individual variability of drug metabolism in cynomolgus macaques

https://doi.org/10.1016/j.bcp.2014.07.004Get rights and content

Abstract

CYP2C19 (formerly known as CYP2C75), highly homologous to human CYP2C19, has been identified in cynomolgus and rhesus macaques, non-human primate species widely used in drug metabolism studies. CYP2C19 is predominantly expressed in liver and encodes a functional drug-metabolizing enzyme. Genetic variants in human CYP2C genes account for the inter-individual variability in drug metabolism; however, genetic variants have not been investigated in macaque CYP2C19. In the present study, re-sequencing of CYP2C19 in 78 cynomolgus and 36 rhesus macaques identified 34 non-synonymous variants. Among these, 6 were located in substrate recognition sites, the domains important for protein function. Eighteen and 6 variants were unique to cynomolgus and rhesus macaques, respectively. Four variants were characterized by site-directed mutagenesis and metabolic assays, and 3 variants (p.Phe100Asn, p.Ala103Val, and p.Ile112Leu) showed substantially reduced activity as compared with wild type in flurbiprofen 4′-hydroxylation, omeprazole 5-hydroxylation, and R-/S-warfarin 7-hydroxylation. These variants, co-segregating in the animals analyzed, influenced metabolic activities because the homozygotes and/or heterozygotes showed significantly reduced catalytic activities in liver toward flurbiprofen 4′-hydroxylation and omeprazole 5-hydroxylation as compared with wild type. Kinetic analysis for R-warfarin 7-hydroxylation and docking simulation indicated that CYP2C19 Ala103Val would change the function and conformation of this enzyme. Ala103Val variation diminished homotropic cooperativity of CYP2C19 with R-warfarin yielding low metabolic capacity. These results indicated that the interindividual variability of CYP2C-dependent drug metabolism is at least partly accounted for by CYP2C19 variants in cynomolgus macaques.

Introduction

Human CYP2Cs, including CYP2C8, CYP2C9, and CYP2C19, are essential drug-metabolizing enzymes, involving the metabolism of approximately 20% of the prescribed drugs such as flurbiprofen and S-mephenytoin [1]. In the human CYP2C genes, a number of genetic variants have been identified (see http://www.cypalleles.ki.se/), including CYP2C9*2 and CYP2C9*3. These alleles, resulting in the reduced activity of the enzyme, are more prevalent in Caucasians than in Africans while Asians do not appear to possess CYP2C9*2 [2]. In human CYP2C19, CYP2C19*2 and CYP2C19*3, accounting for the majority of poor metabolizer phenotypes, are more prevalent in Asians than in Caucasians or Africans, among which CYP2C19*3 is largely found in Asians [2]. These defective alleles of human CYP2C9 and CYP2C19 genes need to be considered in drug metabolism studies during drug development.

Cynomolgus macaques (Macaca fascicularis), and rhesus macaques (Macaca mulatta), are non-human primate species widely used in drug metabolism studies. Cynomolgus CYP2C19, formerly known as CYP2C75, is highly homologous to human CYP2C9 and CYP2C19, and the gene is abundantly expressed in liver [3]. Cynomolgus CYP2C19 metabolizes human CYP2C substrates; S-mephenytoin, flurbiprofen, and tolbutamide [3], [4]. The previous study reported that pharmacokinetics of S-mephenytoin was highly variable among 64 cynomolgus macaques [5], and such inter-animal variations might be accounted for by genetic variants. Genetic polymorphisms have been found in macaque P450 genes [6], [7], [8], [9], [10]: however, the genetic variants have not been investigated in macaque CYP2C19.

In the present study, the genomes of 78 cynomolgus macaques (38 from Indochina and 40 from Indonesia) and 36 rhesus macaques were analyzed to identify CYP2C19 genetic variants. Several identified alleles were analyzed by metabolic assays using flurbiprofen, omeprazole, and S-/R-warfarin as substrate with monkey liver microsomes and the proteins heterologously expressed in Escherichia coli (E. coli).

Section snippets

Preparation of DNA and microsome samples

Whole blood samples were collected from 78 cynomolgus macaques (38 from Indochina and 40 from Indonesia, 4–5 years of age, weighing 3–5 kg) and 36 rhesus macaques (from China, 7 years of age, weighing 3–5 kg). From these samples, genomic DNA was prepared using the Puregene DNA isolation kit (Qiagen, Valencia, CA) according to the manufacturer's instructions. Microsome samples were prepared from the livers of 7 Indochinese cynomolgus macaques as described previously [11]. The study was reviewed

Identification of genetic variants

To identify genetic variants, CYP2C19 was re-sequenced in 78 cynomolgus macaques and 36 rhesus macaques. The analysis identified a total of 34 non-synonymous variants, among which 6 were in substrate recognition sites (SRS), the important regions for protein function; Phe100Asn, Ala103Val, and Ile112Leu in SRS1, Ile237Val and Leu240Met in SRS3, and Leu361Phe in SRS5 (Table 2). Three variants (Phe100Asn, Ala103Val, and Ile112Leu) completely co-segregated and mostly co-segregated in the

Discussion

Cynomolgus macaques are important animal species for drug metabolism studies. However, genetic variants, which might result in the inter-individual variability of catalytic activity, have not been investigated in cynomolgus CYP2C19, which encodes the enzyme metabolizing human CYP2C substrates such as diclofenac, flurbiprofen, omeprazole, and warfarin. In the present study, a total of 34 non-synonymous genetic variants were found by screening the genome samples of 78 cynomolgus and 36 rhesus

Acknowledgements

This work was supported in part by the Ministry of Education, Science, Sports and Culture of Japan and the Takeda Science Foundation, Japan. Authors greatly thank Mr. Masahiro Utoh for his support to this work, Drs. Shotaro Uehara and Makiko Shimizu for their technical assistance, and Mr. Lance Bell for his advice on English writing.

References (21)

There are more references available in the full text version of this article.

Cited by (31)

  • Genetic polymorphisms of drug-metabolizing cytochrome P450 enzymes in cynomolgus and rhesus monkeys and common marmosets in preclinical studies for humans

    2018, Biochemical Pharmacology
    Citation Excerpt :

    Metabolic clearance of human P450 cocktail probes (caffeine, S-warfarin, omeprazole, metoprolol, and midazolam in combination) [28] was compared to those of the same cassette dosing in cynomolgus monkeys [29]. Cynomolgus monkey P450 2C19, highly homologous to human P450 2C19, catalyzed R-warfarin 7-hydroxylation [6,30]. In vivo pharmacokinetics of racemic warfarin and its metabolites after oral and intravenous administrations were studied in male cynomolgus monkeys (n = 11, from Indochina) [17] which had been genotyped for the compound P450 2C19 [F100N; A103V; I112L] variant [10] (Table 2).

View all citing articles on Scopus
View full text