Transl Clin Pharmacol. 2014 Jun;22(1):3-7. English.
Published online Jun 30, 2014.
Copyright © 2014 Translational and Clinical Pharmacology
Review

CYP3A5*3 Polymorphism and Its Clinical Implications and Pharmacokinetic Role

Ji-Young Park, Yu-Jung Cha and Kyoung-Ah Kim
    • Department of Clinical Pharmacology and Toxicology, Anam Hospital, Korea University College of Medicine, Seoul 136-705, Korea.
Received April 23, 2014; Accepted April 23, 2014.

It is identical to the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/3.0/).

Abstract

The cytochrome P450 (CYP) 3A subfamily is estimated to participate in the biotransformation of 50% of the currently prescribed drugs. Four members of the CYP3A subfamily have been identified in humans: CYP3A4, CYP3A5, CYP3A7, and CYP3A43. Initial data suggested that CYP3A5 accounts for only a small proportion of the total hepatic CYP3A in about 20% of samples, but it was later revealed that CYP3A5 represents more than 50% of the total CYP3A amount in some individuals. Several genetic variants have been described for the CYP3A5 gene, of which the CYP3A5*3 allele (gA6986G), the most common form and leading to the loss of CYP3A5 activity, has been extensively investigated in the aspect of pharmacokinetics and disease risk. This review summarized the molecular characteristics of the CYP3A5 gene, and discusses the association of the CYP3A5*3 polymorphism with disease risks such as cancer and hypertension, along with its role in the pharmacokinetics of CYP3A substrates.

Keywords
CYP3A5*3; Cytochrome P450; Cancer; Pharmacokinetics; Pharmacogenetics

Introduction

Interindividual variability in pharmacokinetics including drug absorption, distribution, metabolism, and elimination, which can arise from either genetic or non-genetic origins, is a main factor in the development of therapeutic failure or toxicity. Cytochrome P450 is the heme-containing superfamily of enzymes responsible for the metabolism of a variety of diverse substrates, including xenobiotics. Additionally, these enzymes are involved in the metabolism of cholesterol, steroid hormones and other lipids that are important in intraceullar signaling pathways.[1]

The cytochrome P450 (CYP) 3A subfamily is estimated to participate in the biotransformation of 50% of the drugs known to undergo oxidative metabolism.[2] Four members of the CYP3A subfamily have been described in humans-CYP3A4, CYP3A5, CYP3A7, and CYP3A43.[3] CYP3A4 is the most abundant form in the human liver and intestine, followed by CYP3A5.[4] The genes expressing these enzymes are located adjacent to each other in the order CYP3A5, CYP3A7, CYP3A4 and CYP3A43, in a 231-kb cluster on chromosome 7q21-7q22.[5]

CYP3A7 is highly expressed in fetal livers, accounting for up to 50% of total cytochrome P450s. Its expression level decreases rapidly after birth and becomes undetectable in most adult livers, except for the subjects carrying one of two promoter variants, CYP3A7*1B and CYP3A7*1C.[6] CYP3A43 is also expressed at very low levels in adult livers, accounting for only 0.1-0.2% of CYP3A transcripts.[7, 8]

Interestingly, the hepatic expression of CYP3A5 is bimodal, with the high expressor exhibiting 10-30 fold higher CYP3A5 expression levels than the CYP3A5 lower expressor.[4, 9] Kuehl et al. found the causative CYP3A5*3 (g.6986A>G) genetic polymorphism at the position of intron 3, which disrupts the correct splicing of CYP3A5 transcripts to make a truncated CYP3A5 enzyme with no activity.[4]

In this report, we would like to discuss the role of the CYP3A5*3 genetic polymorphism in association with diseases including hypertension and cancer, along with an examination of the pharmacokinetics of CYP3A substrates as a factor causing interindividual variability.

Genetic polymorphism of CYP3A5

Wrighton et al. first reported large interindividual variability of CYP3A5 expression in the liver in 1990,[10] but Jounaidi et al. was the first to report a polymorphism at the 5'-upstream region (CYP3A5*1A) in the aspect of pharmacogenetics.[11] Paulussen et al. then described its genetic basis for the modification of CYP3A5 expression.[12] They showed a clear bimodality in the drug metabolism of the CYP3A5 phenotype, and identified two linked mutations in the promoter region, -45A>G and -368T>G, which were associated with the expression and activity of the CYP3A5 protein. Notably, the mutation at the site of -45A>G is located in the basic transcription binding element (BTE) of CYP3A5. This mutation may interfere Sp1 binding, thus reducing CYP3A5 gene transcription, to result in lower protein expression in the variant allele with -45A. Lee et al. later identified various SNPs in the 5'-upstream region of the CYP3A5 gene.[13] They found 34 SNPs of CYP3A5, including 27 previously unidentified SNPs, by direct sequencing of the exons, intron-exon junctions and 5'-upstream region of CYP3A5 from 92 racially diverse individuals. The majority of these SNPs occurred in African-American, while only one or two were observed in Asians and Caucasians. However, the small sample size, around 24, might have had limitations to finding other variants, and to accurately reveal the occurrence of SNPs in diverse ethnic groups.

The first genetic variant found in the coding region of the CYP3A5 gene was the CYP3A5*2 allele, which was identified in heterozygotes at a frequency of 3 in 10.[14] This mutation is located in exon 11 at the position of 1289C>A, which produces a change in the amino acid at position 398 (threonine → asparagine). As described above, Kuehl et al. first described the role of the CYP3A5*3 polymorphism mechanistically.[4] This SNP was reported in all ethnic groups, and is thus considered the most ancient allele.[15] The allele frequency of CYP3A5*3 among East Asian populations (Korean, Chinese, and Japanese) is 75-79%, and also occurs in Malay (61%), Indian (59%), African-American (34%), Caucasians (91-94%) and Hispanics 76%.[16, 17, 18, 19, 20] In this allele, a guanine at the position of 6986 creates a cryptic splicing site in intron 3, which leads to aberrant splicing of the transcript to cause truncation of the CYP3A5 protein, which results in no enzyme activity.[9, 10] This polymorphic allele is strongly associated with the reduced drug clearance of CYP3A substrates, assuming higher drug levels of the CYP3A substrates in the subjects possessing this allele. Because it has a relatively high frequency and occurs in diverse populations, its roles have been extensively investigated. We discussed it further here.

CYP3A5*4 is a coding SNP located in exon 7, which produces an amino acid substitution from glutamine to arginine at the position of the 200th amino acid (Q200R).[21] In the case of CYP3A5*5, it was first described in a Chinese population, representing 12952T>C substitution located in intron 5, which leads to the formation of an alternatively spliced mRNA like CYP3A5*3, and presumably decreased CYP3A5 protein activity.[21] Both CYP3A5*4 and CYP3A5*5 are rare mutations, present only in Chinese at less than 1% frequency.[21] The CYP3A5*6 allele leads to the skipping of exon 7 in transcripts. It is found with an allele frequency of 17% in African-Americans, but is absent in Asian populations.[1, 21] A single base insertion into exon 11 causes premature termination of the reading frame at the position of the 348th amino acid in the CYP3A5*7 allele, thus producing unstable CYP3A5 proteins. Similar to CYP3A5*6, it is found only in populations of African origin (Zimbabwean, 10% and African-American, 10%), not in Caucasian and Asian populations.[22] The CYP3A5*8 variant, presenting a substitution of nucleotide C to T, leads to an amino acid change from arginine to cysteine at codon 28.[13] It was first observed in African-American (4%), but the frequencies in other ethnic groups have not yet been revealed. CYP3A5*9 is a SNP in the coding region, resulting from a nucleotide substitution from G to A in exon 10, leading to an amino acid change from alanine to threonine at codon 337. It was observed in an Asian population at a frequency of 2%.[13] CYP3A5*10 was identified in one American subject out of a group of 24 diverse Caucasians, with an allelic frequency of 2%.[10] It is a point mutation substituting T to C in exon 12, resulting in an amino acid change at residue 446 from phenylalanine to serine.[10]

In vitro data showed that CYP3A5*1 exhibited the highest maximal clearance for testosterone and the highest Vmax (maximal capacity) for nifedipine oxidation, followed by CYP3A5*9, CYP3A5*8, and CYP3A5*10.[23] In particular, the report also showed that subjects with CYP3A5*10 exhibited a greater than 95% decrease in the intrinsic clearance for both nifedipine and testosterone metabolism.[23]

Clinical implications of CYP3A5 polymorphism

Hypertension

The CYP3A enzymes metabolize cortisol into 6β-hydroxycortisol, and corticosterone into 6β-hydroxycorticosteone.[24, 25] It was shown that the levels of 6α-hydroxycortisol and 6β-hydroxycortisol were higher by an average of 48% in patients with essential hypertension compared to normotensive subje e occurrence of hypertension, or are involved in blood pressure control. Rais et al. reported an association between the urinary 6β-hydroxycortisol/cortisol ratio and the CYP3A5 genotypes in a North Indian normotensive population, and showed the mean 6β-hydro-xycortisol/cortisol ratios to be 110, 76 and 69 in wild-type CYP3A5*1/*1 (n=12), CYP3A5*1/*3 (n=62) and CYP3A5*3/*3 (n=76), respectively, suggesting a substantial role of the polymorphic CYP3A5 genes in the disposition of steroid hormones.[27] Ho et al. reported that the participants of African origin with CYP3A5*3/*3 (146±35 mm Hg) had a higher systolic blood pressure than those with the CYP3A5*1/*3 (119±14.1 mmHg; P=0.0006) and CYP3A5*1/*1 (125±17.4 mmHg; P=0.009) genotypes, but no association was detected between CYP3A5 genotype and blood pressure in Caucasians.[28] They concluded that although blood pressure may be higher in untreated participants of African origin possessing the CYP3A5*3/*3 genotype, the CYP3A5*1 allele may be associated with hypertension that is more refractory to treatment in this ethnic group. However, the current data on the association between CYP3A5 polymorphisms and hypertension or blood pressure control show inconsistencies.[29]

Cancer

CYP3A5 is primarily expressed in the liver and the gastrointestinal tract, and metabolizes endogenous compounds including the steroid hormones testosterone and progesterone, and fatty acids.[30, 31] Based on the assumption that CYP3A5 plays a role in the modulation of steroid hormone levels, many scientists have evaluated the role of CYP3A5 polymorphisms in cancer risk, including in the following cancers: acute or chronic leukemia, prostate cancer, colorectal cancer, gastric cancer, esophageal cancer, testicular cancer, and breast cancer.[16, 32, 33, 34, 35, 36, 37, 38] A recent meta-analysis revealed that CYP3A5*3 polymorphism may increase the risks of acute leukemia, chronic leukemia, and colorectal cancers.[33] However, no evidence was found of association in prostate cancer, liver cancer, and other cancers with CYP3A5*3 polymorphisms. Additionally, the finding was more striking among Asian and Caucasian populations, suggesting ethnic differences in the occurrence of cancers.

Other diseases

Feng et al. studied the role of both CYP2C19*3 and CYP3A5*3 polymorphisms in the susceptibility of Chinese pediatric tuberculosis patients to disease, and found both genetic polymorphisms contributed to protection from tuberculosis.[39] They speculated that this was attributed to the reduction of intracellular M. tuberculosis, caused by successful apoptosis under the influence of reactive oxygen species (ROS) through these genetic polymorphisms.

Du et al. assessed the genetic susceptibility to schizophrenia in association with the CYP3A4/3A5 genes, and concluded that CYP3A4*1G and CYP3A5*3 polymorphisms were strongly associated with the occurrence of schizophrenia.[40] Alterations in dopamine and serotonin (5-HT) levels in the brain have been implicated in the etiology of psychiatric diseases, including schizophrenia and depression. Accordingly, these molecules have been targeted for pharmacotherapy in the field of psychiatry. Cytochrome P450s, including CYP1A1, 1A2, 3A4, and 3A5, are involved in serotonin metabolism, and messenger RNA of both CYP3A4/3A5 is expressed in the brain.[41] These findings indicate that CYP3A5 may play a crucial role in the disposition of dopamine and serotonin in the brain, thereby causing the possible associations of polymorphic expression with the psychiatric diseases, including schizophrenia.

Tobacco smoke contains polycyclic aromatic hydrocarbons (PAHs), which become activated in the body by action of the cytochrome P450s.[42] Based on the assumption that the activated PAHs are associated with decreased lung function, for which CYP3A5 is one of the PHA-metabolizing enzymes,[43, 44, 45] Kaur-Knudsen et al. studied whether CYP3A5*3 influences lung function and the risk of chronic obstructive pulmonary disease (COPD) in smokers.[46] However, no association could be identified in the study between CYP3A5*3 and decreased lung function or risk of COPD among those who have smoked.

Pharmacokinetics of CYP3A substrates

The CYP3A enzymes have very broad substrate specificity, and an extremely large number of structurally diverse drugs are metabolized by a variety of different pathways through the enzymes.[5, 15] Considering that CYP3A enzymes including CYP3A5 participate in the biotransformation of 50% of the pharmaceutical drugs currently in use,[2] it is obvious that the polymorphic CYP3A5*3 gene must play a crucial role in the pharmacokinetics of these CYP3A substrates. It is reasonable to assume that polymorphisms in the CYP3A5 gene should lead to interindividual variability, which has been observed in the pharmacokinetics of CYP3A substrates. Similar to other studies evaluating the role of CYP3A5*3 polymorphism, when we assessed the effect of CYP3A5*3 polymorphism on the pharmacokinetics of various CYP3A substrates, we found increased drug exposure or plasma drug levels for the following: simvastatin, alprazolam, risperidone, and carbamazepine.[47, 48, 49, 50] In the case of alprazolam,[50] after a 1 mg dose was administered to healthy individuals, the area under the plasma concentration-time curve (AUC) for alprazolam was significantly greater in subjects with the CYP3A5*3/*3 polymorphism (830.5 ng*h/mL) than in those with CYP3A5*1/*1 (599.9 ng*h/mL) (P=0.030), while the oral clearance was also significantly different between the CYP3A5*1/*1 (3.5 L/h) and CYP3A5*3/*3 groups (2.5 L/h) (P=0.036). Simvastatin also showed similar results: after 20 mg simvastatin medication in healthy subjects, the AUC in the CYP3A5*1/*1 carriers (4.94 ng*h/mL) was significantly lower than in the CYP3A5*3/*3 carriers (16.35 ng*h/mL; P=0.013). The oral clearance was also significantly different between CYP3A5*1/*1 carriers (4.80 L/h) and CYP3A5*3/*3 carriers (1.35 L/h; P<0.05). Besides these studies, many scientists have evaluated the role of CYP3A5*3 polymorphisms in the pharmacokinetics of various CYP3A substrates.[9, 15, 51, 52]

Despite the clear evidence that the polymorphic CYP3A5*3 gene leads to decreased enzyme activity, intriguingly, a contradictory finding showing a higher clearance in subjects with the CYP3A5*3 allele than those with normal allele was also observed. In the case of amlodipine, the oral clearance was lower in the CYP3A5*1 carriers (27.0 L/h) than the CYP3A5*3/*3 (32.4 L/h) carriers (P=0.063), but the mean AUC of amlodipine was higher in the CYP3A5*1 (200.9 L/h) carriers than in the CYP3A5*3/*3 (167.6 L/h) carriers (P=0.029).[53] This finding indicates that despite the molecular evidence that CYP3A5*3 has a dysfunctional enzyme activity, the CYP3A5*3 enzyme can still have a higher activity than the CYP3A5*1 enzyme for some CYP3A substrates; however, the mechanism(s) for CYP3A5*3 carriers to exhibit a higher clearance rate than normal CYP3A5*1 carriers could not be clearly explained. Another recent study also observed consistent findings to ours when investigating amlodipine pharmacokinetics.[29]

Notes

Nothing to declare

References

    1. Wang K, Chen S, Xie W, Wan YJ. Retinoids induce cytochrome P450 3A4 through RXR/VDR-mediated pathway. Biochem Pharmacol 2008;75:2204–2213.
    1. Evans WE, Relling MV. Pharmacogenomics: translating functional genomics into rational therapeutics. Science 1999;286:487–491.
    1. Nelson DR, Koymans L, Kamataki T, Stegeman JJ, Feyereisen R, Waxman DJ, et al. P450 superfamily: update on new sequences, gene mapping, accession numbers and nomenclature. Pharmacogenetics 1996;6:1–42.
    1. Kuehl P, Zhang J, Lin Y, Lamba J, Assem M, Schuetz J, et al. Sequence diversity in CYP3A promoters and characterization of the genetic basis of polymorphic CYP3A5 expression. Nat Genet 2001;27:383–391.
    1. Shimada T, Yamazaki H, Mimura M, Inui Y, Guengerich FP. Interindividual variations in human liver cytochrome P-450 enzymes involved in the oxidation of drugs, carcinogens and toxic chemicals: studies with liver microsomes of 30 Japanese and 30 Caucasians. J Pharmacol Exp Ther 1994;270:414–423.
    1. Burk O, Tegude H, Koch I, Hustert E, Wolbold R, Glaeser H, et al. Molecular mechanisms of polymorphic CYP3A7 expression in adult human liver and intestine. J Biol Chem 2002;277:24280–24288.
    1. Westlind A, Malmebo S, Johansson I, Otter C, Andersson TB, Ingelman-Sundberg M, et al. Cloning and tissue distribution of a novel human cytochrome p450 of the CYP3A subfamily, CYP3A43. Biochem Biophys Res Commun 2001;281:1349–1355.
    1. Gellner K, Eiselt R, Hustert E, Arnold H, Koch I, Haberl M, et al. Genomic organization of the human CYP3A locus: identification of a new, inducible CYP3A gene. Pharmacogenetics 2001;11:111–121.
    1. Hustert E, Haberl M, Burk O, Wolbold R, He YQ, Klein K, et al. The genetic determinants of the CYP3A5 polymorphism. Pharmacogenetics 2001;11:773–779.
    1. Wrighton SA, Brian WR, Sari MA, Iwasaki M, Guengerich FP, Raucy JL, et al. Studies on the expression and metabolic capabilities of human liver cytochrome P450IIIA5 (HLp3). Mol Pharmacol 1990;38:207–213.
    1. Jounaïdi Y, Guzelian PS, Maurel P, Vilarem MJ. Sequence of the 5'-flanking region of CYP3A5: comparative analysis with CYP3A4 and CYP3A7. Biochem Biophys Res Commun 1994;205:1741–1747.
    1. Paulussen A, Lavrijsen K, Bohets H, Hendrickx J, Verhasselt P, Luyten W, et al. Two linked mutations in transcriptional regulatory elements of the CYP3A5 gene constitute the major genetic determinant of polymorphic activity in humans. Pharmacogenetics 2000;10:415–424.
    1. Lee SJ, Usmani KA, Chanas B, Ghanayem B, Xi T, Hodgson E, et al. Genetic findings and functional studies of human CYP3A5 single nucleotide polymorphisms in different ethnic groups. Pharmacogenetics 2003;13:461–472.
    1. Jounaïdi Y, Hyrailles V, Gervot L, Maurel P. Detection of CYP3A5 allelic variant: a candidate for the polymorphic expression of the protein? Biochem Biophys Res Commun 1996;221:466–470.
    1. Lakhman SS, Ma Q, Morse GD. Pharmacogenomics of CYP3A: considerations for HIV treatment. Pharmacogenomics 2009;10:1323–1339.
    1. Plummer SJ, Conti DV, Paris PL, Curran AP, Casey G, Witte JS. CYP3A4 and CYP3A5 genotypes, haplotypes, and risk of prostate cancer. Cancer Epidemiol Biomarkers Prev 2003;12:928–932.
    1. Sinues B, Vicente J, Fanlo A, Vasquez P, Medina JC, Mayayo E, et al. CYP3A5*3 and CYP3A4*1B allele distribution and genotype combinations: differences between Spaniards and Central Americans. Ther Drug Monit 2007;29:412–416.
    1. Dally H, Bartsch H, Jäger B, Edler L, Schmezer P, Spiegelhalder B, et al. Genotype relationships in the CYP3A locus in Caucasians. Cancer Lett 2004;207:95–99.
    1. Balram C, Zhou Q, Cheung YB, Lee EJ. CYP3A5*3 and *6 single nucleotide polymorphisms in three distinct Asian populations. Eur J Clin Pharmacol 2003;59:123–126.
    1. Fukuen S, Fukuda T, Maune H, Ikenaga Y, Yamamoto I, Inaba T, et al. Novel detection assay by PCR-RFLP and frequency of the CYP3A5 SNPs, CYP3A5*3 and *6, in a Japanese population. Pharmacogenetics 2002;12:331–334.
    1. Chou FC, Tzeng SJ, Huang JD. Genetic polymorphism of cytochrome P450 3A5 in Chinese. Drug Metab Dispos 2001;29:1205–1209.
    1. Roy JN, Lajoie J, Zijenah LS, Barama A, Poirier C, Ward BJ, et al. CYP3A5 genetic polymorphisms in different ethnic populations. Drug Metab Dispos 2005;33:884–887.
    1. Lee SJ, Goldstein JA. Functionally defective or altered CYP3A4 and CYP3A5 single nucleotide polymorphisms and their detection with genotyping tests. Pharmacogenomics 2005;6:357–371.
    1. Watlington CO, Kramer LB, Schuetz EG, Zilai J, Grogan WM, Guzelian P, et al. Corticosterone 6 beta-hydroxylation correlates with blood pressure in spontaneously hypertensive rats. Am J Physiol 1992;262:F927–F931.
    1. Ghosh S, Grogan WM, Basu A, Watlington C. Renal corticosterone 6 beta-hydroxylase in the spontaneously hypertensive rat. Biochim Biophys Acta 1993;1182:152–156.
    1. Cui JS, Hopper JL, Harrap SB. Antihypertensive treatments obscure familial contributions to blood pressure variation. Hypertension 2003;41:207–210.
    1. Rais N, Hussain A, Chawla YK, Kohli KK. Association between urinary 6b-hydroxycortisol/cortisol ratio and CYP3A5 genotypes in a normotensive population. Exp Ther Med 2013;5:527–532.
    1. Ho H, Pinto A, Hall SD, Flockhart DA, Li L, Skaar TC, et al. Association between the CYP3A5 genotype and blood pressure. Hypertension 2005;45:294–298.
    1. Zhang YP, Zuo XC, Huang ZJ, Cai JJ, Wen J, Duan DD, et al. CYP3A5 polymorphism, amlodipine and hypertension. J Hum Hypertens 2014;28:145–149.
    1. Willrich MA, Hirata MH, Hirata RD. Statin regulation of CYP3A4 and CYP3A5 expression. Pharmacogenomics 2009;10:1017–1024.
    1. Lamba JK, Lin YS, Schuetz EG, Thummel KE. Genetic contribution to variable human CYP3A-mediated metabolism. Adv Drug Deliv Rev 2002;54:1271–1294.
    1. Islam MS, Mostofa AG, Ahmed MU, Bin Sayeed MS, Hassan MR, et al. Association of CYP3A4, CYP3A5 polymorphisms with lung cancer risk in Bangladeshi population. Tumour Biol 2014;35:1671–1678.
    1. Wang BS, Liu Z, Xu WX, Sun SL. CYP3A5*3 polymorphism and cancer risk: a meta-analysis and meta-regression. Tumour Biol 2013;34:2357–2366.
    1. Kristiansen W, Haugen TB, Witczak O, Andersen JM, Fosså SD, Aschim EL. CYP1A1, CYP3A5 and CYP3A7 polymorphisms and testicular cancer susceptibility. Int J Androl 2011;34:77–83.
    1. Gervasini G, García-Martín E, Ladero JM, Pizarro R, Sastre J, Martínez C, et al. Genetic variability in CYP3A4 and CYP3A5 in primary liver, gastric and colorectal cancer patients. BMC Cancer 2007;7:118.
    1. Dandara C, Ballo R, Parker MI. CYP3A5 genotypes and risk of oesophageal cancer in two South African populations. Cancer Lett 2005;225:275–282.
    1. Rao DN, Manjula G, Sailaja K, Surekha D, Raghunadharao D, Rajappa S, et al. Association of CYP3A5*3 polymorphism with development of acute leukemia. Indian J Hum Genet 2011;17:175–178.
    1. Liu TC, Lin SF, Chen TP, Chang JG. Polymorphism analysis of CYP3A5 in myeloid leukemia. Oncol Rep 2002;9:327–329.
    1. Feng WX, Liu F, Gu Y, Jiao WW, Sun L, Xiao J, et al. Functional polymorphisms in CYP2C19 & CYP3A5 genes associated with decreased susceptibility for paediatric tuberculosis. Indian J Med Res 2012;135:642–649.
    1. Du J, Xu Y, Duan S, Zhang A, Xuan J, Wang L, et al. A case-control association study between the CYP3A4 and CYP3A5 genes and schizophrenia in the Chinese Han population. Prog Neuropsychopharmacol Biol Psychiatry 2009;33:1200–1204.
    1. Jia Y, Yu X, Zhang B, Yuan Y, Xu Q, Shen Y, et al. No association between polymorphisms in three genes of cytochrome p450 family and paranoid schizophrenia in northern Chinese Han population. Eur Psychiatry 2004;19:374–376.
    1. Shimada T, Martin MV, Pruess-Schwartz D, Marnett LJ, Guengerich FP. Roles of individual human cytochrome P-450 enzymes in the bioactivation of benzo(a)pyrene, 7,8-dihydroxy-7,8-dihydrobenzo(a)pyrene, and other dihydrodiol derivatives of polycyclic aromatic hydrocarbons. Cancer Res 1989;49:6304–6312.
    1. Le Moual N, Orlowski E, Schenker MB, Avignon M, Brochard P, Kauffmann F. Occupational exposures estimated by means of job exposure matrices in relation to lung function in the PAARC survey. Occup Environ Med 1995;52:634–643.
    1. Piipari R, Savela K, Nurminen T, Hukkanen J, Raunio H, Hakkola J, et al. Expression of CYP1A1, CYP1B1 and CYP3A, and polycyclic aromatic hydrocarbon-DNA adduct formation in bronchoalveolar macrophages of smokers and non-smokers. Int J Cancer 2000;86:610–616.
    1. Roberts-Thomson SJ, McManus ME, Tukey RH, Gonzalez FF, Holder GM. The catalytic activity of four expressed human cytochrome P450s towards benzo[a]pyrene and the isomers of its proximate carcinogen. Biochem Biophys Res Commun 1993;192:1373–1379.
    1. Kaur-Knudsen D, Bojesen SE, Nordestgaard BG. CHRNA3 and CYP3A5*3 genotype, lung function and chronic obstructive pulmonary disease in the general population. Pharmacogenet Genomics 2014;24:220–229.
    1. Park PW, Seo YH, Ahn JY, Kim KA, Park JY. Effect of CYP3A5*3 genotype on serum carbamazepine concentrations at steady-state in Korean epileptic patients. J Clin Pharm Ther 2009;34:569–574.
    1. Kang RH, Jung SM, Kim KA, Lee DK, Cho HK, Jung BJ, et al. Effects of CYP2D6 and CYP3A5 genotypes on the plasma concentrations of risperidone and 9-hydroxyrisperidone in Korean schizophrenic patients. J Clin Psychopharmacol 2009;29:272–277.
    1. Kim KA, Park PW, Lee OJ, Kang DK, Park JY. Effect of polymorphic CYP3A5 genotype on the single-dose simvastatin pharmacokinetics in healthy subjects. J Clin Pharmacol 2007;47:87–93.
    1. Park JY, Kim KA, Park PW, Lee OJ, Kang DK, Shon JH, et al. Effect of CYP3A5*3 genotype on the pharmacokinetics and pharmacodynamics of alprazolam in healthy subjects. Clin Pharmacol Ther 2006;79:590–599.
    1. Wojnowski L, Kamdem LK. Clinical implications of CYP3A polymorphisms. Expert Opin Drug Metab Toxicol 2006;2:171–182.
    1. Utecht KN, Hiles JJ, Kolesar J. Effects of genetic polymorphisms on the pharmacokinetics of calcineurin inhibitors. Am J Health Syst Pharm 2006;63:2340–2348.
    1. Kim KA, Park PW, Lee OJ, Choi SH, Min BH, Shin KH, et al. Effect of CYP3A5*3 genotype on the pharmacokinetics and pharmacodynamics of amlodipine in healthy Korean subjects. Clin Pharmacol Ther 2006;80:646–656.

Metrics
Share
PERMALINK