Please wait a minute...

中国生物工程杂志

China Biotechnology
China Biotechnology  2016, Vol. 36 Issue (12): 104-110    DOI: DOI:10.13523/j.cb.20161215
    
Advances in Research on Genetic Polymorphism of Cytochrome P450 and Drug Metabolism
LIU Wen-bo1, CHEN Yu-bao2, XING Yu-hua2
1. Huilongsen Europe Technology(Beijing) Co., Ltd, Beijing 101111, China;
2. Beijing Computer Center, Beijing Engineering Research Center for Gene Sequencing & Functional Analysis, Beijing 100094, China
Download: HTML   PDF(408KB) HTML
Export: BibTeX | EndNote (RIS)      

Abstract  

The cytochrome P450(CYP450) play critical roles in the drug metabolism and are responsible for the metabolism of more than 80% of clinical drugs. Genetic polymorphism in different ethnic and population is the main cause for individual variation in drug metabolism. The new advancement between the genetic polymorphism and drug metabolism of CYP1A2, CYP2C9, CYP2C19, CYP2D6 and CYP3A4 were reviewed, which can offer the scientific reference for clinical individual therapy and the development of new drug.



Key wordsCytochrome P450      Genetic polymorphism      Drug metabolism     
Received: 20 June 2016      Published: 25 December 2016
ZTFLH:  Q819  
Cite this article:

LIU Wen-bo, CHEN Yu-bao, XING Yu-hua. Advances in Research on Genetic Polymorphism of Cytochrome P450 and Drug Metabolism. China Biotechnology, 2016, 36(12): 104-110.

URL:

https://manu60.magtech.com.cn/biotech/DOI:10.13523/j.cb.20161215     OR     https://manu60.magtech.com.cn/biotech/Y2016/V36/I12/104

[1] Klingenberg M. Pigments of rat liver microsomes. Archs Biochem Biophys, 1958, 75:376-386.
[2] Pedersen R S, Damkier P, Christensen M M, et al. A cytochrome P450 phenotyping cocktail causing unexpected adverse reactions in female volunteers. Eur J Clin Pharmacol, 2013,69(12):1997-1999.
[3] Venkatakrishnan K, Von M L, Greenblatt D J. Human drug metabolism and the cytochromes P450:application and relevance of in vitro models. J Clin Pharmacol, 2001, 41(11):1149-1179.
[4] Donzelli M, Derungs A, Serratore M G, et al. The basel cocktail for simultaneous phenotyping of human cytochrome P450 isoforms in plasma, saliva and dried blood spots. Clin Pharmacokinet, 2014,53(3):271-282.
[5] Kirchheiner J, Nickchen K, Bauer M, et al. Pharmacogenetics of antidepressants and antipsychotics:the contribution of allelic variations to the phenotype of drug response. Mol Psychiatry, 2004,9:442-473.
[6] Danielson P B.The cytochrome P450 superfamily:biochemistry, evolution and drug metabolism in humans. Current Drug Metabolism, 2002, 3(6):561-597.
[7] David Ra. Comparison of cytochrome P450(CYP) genes from the mouse and human genomes, including nomenclature recommendations for genes, pseudogenes and alternative-splice variants. Pharmacogenetics, 2004,14:1-18.
[8] Ma C, Adjei A, Salavaggione O, et al. Human aromatase (CYP19) pharmacogenomics:Gene resequencing and functional genomics. Clinical Pharmacology & Therapeutics, 2005, 77(2):22.
[9] Zanger U, Turpeinen M, Klein K, et al. Functional pharmacogenetics/genomics of human cytocromes P450 involved in drug biotransformation. Anal Bioanal Chem,2008, 392,1093-1108.
[10] Bradford L D. CYP2D6 allele frequency in European caucasians, Asians, Africans, and their descendants. Pharmacogenomics, 2002,3:229-243.
[11] Satomi Y, Nishino H. Inhibition of the enzyme activity of cytochrome P4501A1, 1A2 and 3A4 by fucoxanthin, a marine carotenoid. Oncol Lett, 2013,6(3):860-864.
[12] Yoshinari K, Ntoriyabe Y. Constitutive androstane receptor transcriptionally activates human CYP1A1 and CYP1A2 genes through a common regulatory element in the 5'-flanking region. Biochemical Pharmacology, 2010,79(2):261-269.
[13] Sansen S, Yano J K, Reynald R L, et al. Adaptations for the oxidation of polycyclic aromatic hydrocarbons exhibited by the structure of human P4501A2. Journal of Biological Chemistry, 2007, 282(19):14348-14355.
[14] Gunes A, Dahl M L. Variation in CYP1A2 activity and its clinical implications:influence of environmental factors and genetic polymorphisms. Pharmacogenomics, 2008, 9(5):625-637..
[15] Shao W, He J. CYP1A2 rs2069514 polymorphism and lung cancer susceptibility:a meta-analysis. Annals of Translational Medicine, 2015, 3(7):93.
[16] Allorge D, Chevalier D, Lo-Guidice J M, et al. Identification of a novel splice-site mutation in the CYP1A2 gene. Br J Clin Pharmacol, 2003,56:341-344.
[17] Linder M W, Looney S, Iii J E, et al. Warfarin dose adjustments based on CYP2C9 genetic polymorphisms. Journal of Thrombosis and Thrombolysis, 2002, 14(3):227-232.
[18] Allabi A C, Gala J L, Horsmans Y, et al. Functional impact of CYP2C9*5, CYP2C9*6, CYP2C9*8, and CYP2C9*11 in vivo among black Africans. Clin Pharmacol Ther, 2004, 76:113-118.
[19] Si D, Guo Y, Zhang Y, et al.Identification of a novel variant CYP2C9 allele in Chinese. Pharmacogenetics, 2004, 14:465-469.
[20] Dai D P, Li C B, Wang S H, et al. Identification and characterization of a novel CYP2C9 allelic variant in a warfarin-sensitive patient. Pharmacogenomics, 2015,16(13):1475-1486.
[21] Lee S J, Kim W Y, Kim H, et al. Identification of new CYP2C19 variants exhibiting decreased enzyme activity in the metabolism of S-mephenytoin and omeprazole. Drug Metab Dispos, 2009, 37(11):2262-2269.
[22] Klotz U. Clinical impact of CYP2C19 polymorphism on the action of proton pump inhibitors:a review of a special problem. Int J Clin Pharmacol Ther, 2006, 44(7):297-302.
[23] Sim S C, Risinger C, Dahl M L, et al. A common novel CYP2C19 gene variant causes ultrarapid drug metabolism relevant for the drug response to proton pump inhibitors and antidepressants. Clin Pharmacol Ther, 2006, 79:103-113.
[24] Chaudhry A S, Prasad B, Shirasaka Y, et al. The CYP2C19 intron 2 branch point SNP is the ancestral polymorphism contributing to the poor metabolizer phenotype in livers with CYP2C19*35 and CYP2C19*2 alleles. Drug Metab Dispos, 2015, 43(8):1226-1335.
[25] Sheng H H,Zeng A P,Zhu W X,et al.Allelic distributions of CYP2D6 gene copy number variation in the eastern Han Chinese population. Acta Phamacol Sin, 2007, 28:279-286.
[26] Shen H, He M M, Liu H, et al. Comparative metabolic capabilities and inhibitory profiles of CYP2D6.1, CYP2D6.10, and CYP2D6.17. Drug Metab Dispos,2007, 35:1292-1300.
[27] Borba M A, Meloneto R P, Leitão G M, et al. Evaluating the impact of missenses mutations in CYP2D6*7 and CYP2D6*14A:does it compromise tamoxifen metabolism.Pharmacogenomics, 2016, 17(6):573-582.
[28] Lamba J K, Lin Y S, Schuetz E G, et al. Genetic contribution to variable human CYP3A-mediated metabolism. Adv Drug Deliv Rev, 2002, 54:1271-1294.
[29] Du J, Yu L,Wang L, et al. Differences in CYP3A4*1G genotype distribution and haplotypes of CYP3A4, CYP3A5 and CYP3A7 in 3 Chinese populations. Clin Chim Acta, 2007, 383(4):172-174.
[30] Hsieh K P, Lin Y Y, Cheng C L, et al. Novel mutations of CYP3A4 in Chinese. Drug Metab Dispos, 2001, 29:268-273.
[31] Dai D, Tang J, Rose R, et al. Identification of variants of CYP3A4 and characterization of their abilities to metabolize testosterone and chlorpyrifos. J Pharmacol Exp Ther, 2001,299:825-831.
[32] Werk A N, Lefeldt S, Bruckmueller H, et al. Identification and characterization of a defective CYP3A4 genotype in a kidney transplant patient with severely diminished tacrolimus clearance. Clin Pharmacol Ther, 2014, 95(4):416-422.
[33] Wang X D, Li J L. A pharmacogenetic study of pregnane X receptor (NR1I2) in Han Chinese. Curr Drug Metab, 2007, 8:778-786.

[1] ZHANG Yu-ting,LI Wei-guo,LIANG Dong-mei,QIAO Jian-jun,CAI YIN Qing-ge-le. Research Progress in Synthetic Biology of P450s in Terpenoid Synthesis[J]. China Biotechnology, 2020, 40(8): 84-96.
[2] WU Xue-long, YANG Xiao-hui, WANG Jun-qing, WANG Rui-ming. Expression and Characteristics of Apis mellifera NADPH-cytochrome P450 Reductase Gene in Escherichia coli[J]. China Biotechnology, 2016, 36(12): 28-35.