626
Views
19
CrossRef citations to date
0
Altmetric
Reviews

Ethnic differences in the metabolism, toxicology and efficacy of three anticancer drugs

, PhD & , PhD
Pages 967-988 | Published online: 17 May 2011

Bibliography

  • Iyer L, King CD, Whitington PF, Genetic predisposition to the metabolism of irinotecan (CPT-11). Role of uridine diphosphate glucuronosyltransferase isoform 1A1 in the glucuronidation of its active metabolite (SN-38) in human liver microsomes. J Clin Invest 1998;15:847-54
  • Ciotti M, Basu N, Brangi M, Owens IS. Glucuronidation of 7-ethyl-10-hydroxycamptothecin (SN-38) by the human UDP-glucuronosyltransferases encoded at the UGT1 locus. Biochem Biophys Res Commun 1999;260:199-202
  • Gagne JF, Montminy V, Belanger P, Common human UGT1A polymorphisms and the altered metabolism of irinotecan active metabolite 7-ethyl-10-hydroxycamptothecin (SN-38). Mol Pharmacol 2002;62:608-17
  • Oguri T, Takahashi T, Miyazaki M, UGT1A10 is responsible for SN-38 glucuronidation and its expression in human l ung cancers. Anticancer Res 2004;24:2893-6
  • Santos A, Zanetta S, Cresteil T, Metabolism of irinotecan (CPT-11) by CYP3A4 and CYP3A5 in humans. Clin Cancer Res 2000;6:2012-20
  • Sparreboom A, Danesi R, Ando Y, Pharmacogenomics of ABC transporters and its role in cancer chemotherapy. Drug Resist Updat 2003;6:71-84
  • Nozawa T, Minami H, Sugiura S, Role of organic anion transporter OATP1B1 (OATP-C) in hepatic uptake of irinotecan and its active metabolite, 7-ethyl-10-hydroxycamptothecin: in vitro evidence and effect of single nucleotide polymorphisms. Drug Metab Dispos 2005;33:434-9
  • Chu XY, Suzuki H, Ueda K, Active efflux of CPT-11 and its metabolites in human KB-derived cell lines. J Pharmacol Exp Ther 1999;288:735-41
  • Gupta E, Lestingi TM, Mick R, Metabolic fate of irinotecan in humans: correlation of glucuronidation with diarrhea. Cancer Res 1994;54:3723-5
  • Kehrer DF, Sparreboom A, Verweij J, Modulation of irinotecan-induced diarrhea by cotreatment with neomycin in cancer patients. Clin Cancer Res 2001;7:1136-41
  • Hanioka N, Ozawa S, Jinno H, Human liver UDP-glucuronosyltransferase isoforms involved in the glucuronidation of 7-ethyl-10-hydroxycamptothecin. Xenobiotica 2001;31:687-99
  • Basu NK, Ciotti M, Hwang MS, Differential and special properties of the major human UGT1-encoded gastrointestinal UDP-glucuronosyltransferases enhance potential to control chemical uptake. J Biol Chem 2004;279:1429-41
  • Radominska-Pandya A, Czernik PJ, Little JM, Structural and functional studies of UDP-glucuronosyltransferases. Drug Metab Rev 1999;31:817-99
  • Bosma PJ, Chowdhury JR, Bakker C, The genetic basis of the reduced expression of bilirubin UDP-glucuronosyltransferase 1 in Gilbert's syndrome. N Engl J Med 1995;333:1171-5
  • Beutler E, Gelbart T, Demina A. Racial variability in the UDP-glucuronosyltransferase 1 (UGT1A1) promoter: a balanced polymorphism for regulation of bilirubin metabolism? Proc Natl Acad Sci USA 1998;95:8170-4
  • Girard H, Villeneuve L, Court MH, The novel UGT1A9 intronic I399 polymorphism appears as a predictor of 7-ethyl-10-hydroxycamptothecin glucuronidation levels in the liver. Drug Metab Dispos 2006;34:1220-8
  • Saeki M, Saito Y, Sai K, A combinatorial haplotype of the UDP-glucuronosyltransferase 1A1 gene (#60-#IB) increases total bilirubin levels in Japanese. Clin Chem 2007;53:356-8
  • Saito Y, Maekawa K, Ozawa S, Sawada J. Genetic polymorphisms and haplotypes of major drug metabolizing enzymes in East Asians and their comparison with other ethnic populations. Curr Pharmacogenomics 2007;5:49-78
  • Sandanaraj E, Jada SR, Shu X, Influence of UGT1A9 intronic I399C>T polymorphism on SN-38 glucuronidation in Asian cancer patients. Pharmacogenomics J 2008;8:174-85
  • Cecchin E, Innocenti F, D'Andrea M, Predictive role of the UGT1A1, UGT1A7, and UGT1A9 genetic variants and their haplotypes on the outcome of metastatic colorectal cancer patients treated with fluorouracil, leucovorin, and irinotecan. J Clin Oncol 2009;27:2457-65
  • Jinno H, Tanaka-Kagawa T, Hanioka N, Glucuronidation of 7-ethyl-10-hydroxycamptothecin (SN-38), an active metabolite of irinotecan (CPT-11), by human UGT1A1 variants, G71R, P229Q, and Y486D. Drug Metab Dispos 2003a;31:108-13
  • International HapMap Project Data (Release #27). Available from: http://hapmap.ncbi.nlm.nih.gov/">http://hapmap.ncbi.nlm.nih.gov/[Last">http://hapmap.ncbi.nlm.nih.gov/ [Last accessed 30 November 2010]
  • Sugatani J, Yamakawa K, Yoshinari K, Identification of a defect in the UGT1A1 gene promoter and its association with hyperbilirubinemia. Biochem Biophys Res Commun 2002;292:492-7
  • Kaniwa N, Kurose K, Jinno H, Racial variability in haplotype frequencies of UGT1A1 and glucuronidation activity of a novel single nucleotide polymorphism 686C> T (P229L) found in an African-American. Drug Metab Dispos 2005;33:458-65
  • Saeki M, Saito Y, Jinno H, Haplotype structures of the UGT1A gene complex in a Japanese population. Pharmacogenomics J 2006;6:63-75
  • Takeuchi K, Kobayashi Y, Tamaki S, Genetic polymorphisms of bilirubin uridine diphosphate-glucuronosyltransferase gene in Japanese patients with Crigler-Najjar syndrome or Gilbert's syndrome as well as in healthy Japanese subjects. J Gastroenterol Hepatol 2004;19:1023-8
  • Guillemette C, Ritter JK, Auyeung DJ, Structural heterogeneity at the UDP-glucuronosyltransferase 1 locus: functional consequences of three novel missense mutations in the human UGT1A7 gene. Pharmacogenetics 2000;10:629-44
  • Han JY, Lim HS, Shin ES, Comprehensive analysis of UGT1A polymorphisms predictive for pharmacokinetics and treatment outcome in patients with non-small-cell lung cancer treated with irinotecan and cisplatin. J Clin Oncol 2006;28:2237-44
  • Kohle C, Mohrle B, Munzel PA, Frequent co-occurrence of the TATA box mutation associated with Gilbert's syndrome (UGT1A1*28) with other polymorphisms of the UDP-glucuronosyltransferase-1 locus (UGT1A6*2 and UGT1A7*3) in Caucasians and Egyptians. Biochem Pharmacol 2003;65:1521-7
  • Lankisch TO, Schulz C, Zwingers T, Gilbert's Syndrome and irinotecan toxicity: combination with UDP-glucuronosyltransferase 1A7 variants increases risk. Cancer Epidemiol Biomarkers Prev 2008;17:695-701
  • Yamanaka H, Nakajima M, Katoh M, A novel polymorphism in the promoter region of human UGT1A9 gene (UGT1A9*22) and its effects on the transcriptional activity. Pharmacogenetics 2004;14:329-32
  • Innocenti F, Liu W, Chen P, Haplotypes of variants in the UDP-glucuronosyltransferase1A9 and 1A1 genes. Pharmacogenet Genomics 2005;15:295-301
  • Girard H, Court MH, Bernard O, Identification of common polymorphisms in the promoter of the UGT1A9 gene: evidence that UGT1A9 protein and activity levels are strongly genetically controlled in the liver. Pharmacogenetics 2004;14:501-15
  • Ramirez J, Liu W, Mirkov S, Lack of association between common polymorphisms in UGT1A9 and gene expression and activity. Drug Metab Dispos 2007;35:2149-53
  • Saito Y, Sai K, Maekawa K, Close association of UGT1A9 IVS1+399C>T with UGT1A1*28, *6, or *60 haplotype and its apparent influence on 7-ethyl-10-hydroxycamptothecin (SN-38) glucuronidation in Japanese. Drug Metab Dispos 2009;37:272-6
  • Innocenti F, Grimsley C, Das S, Haplotype structure of the UDP-glucuronosyltransferase 1A1 promoter in different ethnic groups. Pharmacogenetics 2002;12:725-33
  • Sai K, Saeki M, Saito Y, UGT1A1 haplotypes associated with reduced glucuronidation and increased serum bilirubin in irinotecan-administered Japanese patients with cancer. Clin Pharmacol Ther 2004;75:501-15
  • Ki CS, Lee KA, Lee SY, Haplotype structure of the UDP-glucuronosyltransferase 1A1 (UGT1A1) gene and its relationship to serum total bilirubin concentration in a male Korean population. Clin Chem 2003;49:2078-81
  • Carlini LE, Meropol NJ, Bever J, UGT1A7 and UGT1A9 polymorphisms predict response and toxicity in colorectal cancer patients treated with capecitabine/irinotecan. Clin Cancer Res 2005;11:1226-36
  • Ando Y, Saka H, Ando M, Polymorphisms of UDP-glucuronosyltransferase gene and irinotecan toxicity: a pharmacogenetic analysis. Cancer Res 2000;60:6921-6
  • Iyer L, Das S, Janisch L, UGT1A1*28 polymorphism as a determinant of irinotecan disposition and toxicity. Pharmacogenomics J 2002;2:43-7
  • Innocenti F, Undevia SD, Iyer L, Genetic variants in the UDP-glucuronosyltransferase 1A1 gene predict the risk of severe neutropenia of irinotecan. J Clin Oncol 2004;22:1382-8
  • Marcuello E, Altes A, Menoyo A, UGT1A1 gene variations and irinotecan treatment in patients with metastatic colorectal cancer. Br J Cancer 2004;91:678-82
  • Rouits E, Boisdron-Celle M, Dumont A, Relevance of different UGT1A1 polymorphisms in irinotecan-induced toxicity: a molecular and clinical study of 75 patients. Clin Cancer Res 2004;10:5151-9
  • Rouits E, Charasson V, Petain A, Pharmacokinetic and pharmacogenetic determinants of the activity and toxicity of irinotecan in metastatic colorectal cancer patients. Br J Cancer 2008;99:1239-45
  • McLeod HL, Sargent DJ, Marsh S, Pharmacogenetic predictors of adverse events and response to chemotherapy in metastatic colorectal cancer: results from North American Gastrointestinal Intergroup Trial N9741. J Clin Oncol 2010;28:3227-33
  • Toffoli G, Cecchin E, Corona G, The role of UGT1A1*28 polymorphism in the pharmacodynamics and pharmacokinetics of irinotecan in patients with metastatic colorectal cancer. J Clin.Oncol 2006;24:3061-8
  • Massacesi C, Terrazzino S, Marcucci F, Uridine diphosphate glucuronosyl transferase 1A1 promoter polymorphism predicts the risk of gastrointestinal toxicity and fatigue induced by irinotecan-based chemotherapy. Cancer 2006;106:1007-16
  • Cote JF, Kirzin S, Kramar A, UGT1A1 polymorphism can predict hematologic toxicity in patients treated with irinotecan. Clin Cancer Res 2007;13:3269-75
  • Rhodes KE, Zhang W, Yang D, ABCB1, SLCO1B1 and UGT1A1 gene polymorphisms are associated with toxicity in metastatic colorectal cancer patients treated with first-line irinotecan. Drug Metab Lett 2007;1:23-30
  • Kweekel DM, Gelderblom H, Van der Straaten T, Dutch Colorectal Cancer Group study. UGT1A1*28 genotype and irinotecan dosage in patients with metastatic colorectal cancer: a Dutch Colorectal Cancer Group study. Br J Cancer 2008;99:275-82
  • Glimelius B, Garmo H, Berglund A, Prediction of irinotecan and 5-fluorouracil toxicity and response in patients with advanced colorectal cancer. Pharmacogenomics J 2010; published online 23 February 2010, doi: 10.1038/tpj.2010.10.
  • Martinez-Balibrea E, Abad A, Martinez-Cardus A, UGT1A and TYMS genetic variants predict toxicity and response of colorectal cancer patients treated with first-line irinotecan and fluorouracil combination therapy. Br J Cancer 2010;103:581-9
  • Minami H, Sai K, Saeki M, Irinotecan pharmacokineitcs/pharmacodynamics and UGT1A genetic polymorphisms in Japanese: Roles of UGT1A1*6 and *28. Pharmacogenet Genomics 2007;17:497-504
  • Araki K, Fujita K, Ando Y, Pharmacogenetic impact of polymorphisms in the coding region of the UGT1A1 gene on SN-38 glucuronidation in Japanese patients with cancer. Cancer Sci 2006;97:1255-9
  • Takano M, Kato M, Yoshikawa T, Clinical significance of UDP-glucuronosyltransferase 1A1*6 for toxicities of combination chemotherapy with irinotecan and cisplatin in gynecologic cancers: a prospective multi-institutional study. Oncology 2009;76:315-21
  • Jada SR, Lim R, Wong CI, Role of UGT1A1*6, UGT1A1*28 and ABCG2 c.421C>A polymorphisms in irinotecan-induced neutropenia in Asian cancer patients. Cancer Sci 2007;98:1461-7
  • Sai K, Saito Y, Sakamoto H, Importance of UDP-glucuronosyltransferase 1A1 *6 for irinotecan toxicities in Japanese cancer patients. Cancer Lett 2008;261:165-7
  • Onoue M, Terada T, Kobayashi M, UGT1A1*6 polymorphism is most predictive of severe neutropenia induced by irinotecan in Japanese cancer patients. Int J Clin Oncol 2009;14:136-42
  • Liu CY, Chen PM, Chiou TJ, UGT1A1*28 polymorphism predicts irinotecan-induced severe toxicities without affecting treatment outcome and survival in patients with metastatic colorectal carcinoma. Cancer 2008;112:1932-40
  • Nakamura Y, Soda H, Oka M, Randomized phase II trial of irinotecan with paclitaxel or gemcitabine for non-small cell lung cancer: association of UGT1A1*6 and UGT1A1*27 with severe neutropenia. J Thorac Oncol 2011;6:121-7
  • Kitagawa C, Ando M, Ando Y, Genetic polymorphism in the phenobarbital-responsive enhancer module of the UDP-glucuronosyltransferase 1A1 gene and irinotecan toxicity. Pharmacogenet Genomics 2005;15:35-41
  • Lara PN Jr, Natale R, Crowley J, Phase III trial of irinotecan/cisplatin compared with etoposide/cisplatin in extensive-stage small-cell lung cancer: clinical and pharmacogenomic results from SWOG S0124. J Clin Oncol 2009;27:2530-5
  • Ando M, Ando Y, Sekido Y, Genetic polymorphisms of the UDP-glucuronosyltransferase 1A7 gene and irinotecan toxicity in japanese cancer patients. Jpn J Cancer Res 2002;93:591-7
  • Hoffmeyer S, Burk O, von Richter O, Functional polymorphisms of the human multidrug-resistance gene: multiple sequence variations and correlation of one allele with P-glycoprotein expression and activity in vivo. Proc Natl Acad Sci USA 2000;97:3473-8
  • Marzolini C, Paus E, Buclin T, Polymorphisms in human MDR1 (P-glycoprotein): recent advances and clinical relevance. Clin Pharmacol Ther 2004;75:13-33
  • Kim RB, Leake BF, Choo EF, Identification of functionally variant MDR1 alleles among European Americans and African Americans. Clin Pharmacol Ther 2001;70:189-99
  • Kroetz DL, Pauli-Magnus C, Hodges LM, Pharmacogenetics of Membrane Transporters Investigators. Sequence diversity and haplotype structure in the human ABCB1 (MDR1, multidrug resistance transporter) gene. Pharmacogenetics 2003;13:481-94
  • Sai K, Itoda M, Saito Y, Genetic variations and haplotype structures of the ABCB1 gene in a Japanese population: an expanded haplotype block covering the distal promoter region, and associated ethnic differences. Ann Hum Genet 2006;70:605-22
  • Sai K, Kaniwa N, Itoda M, Haplotype analysis of ABCB1/MDR1 blocks in a Japanese population reveals genotype-dependent renal clearance of irinotecan. Pharmacogenetics 2003;13:741-57
  • Sai K, Saito Y, Maekawa K, Additive effects of drug transporter genetic polymorphisms on irinotecan pharmacokinetics/pharmacodynamics in Japanese cancer patients. Cancer Chemother Pharmacol 2010;66:95-105
  • Choi JH, Ahn BM, Yi J, MRP2 haplotypes confer differential susceptibility to toxic liver injury. Pharmacogenet Genomics 2007;17:403-15
  • Sai K, Saito Y, Itoda M, Genetic variations and haplotypes of ABCC2 encoding MRP2 in a Japanese population. Drug Metab Pharmacokinet 2008;23:139-47
  • Bosch TM, Doodeman VD, Smits PH, Pharmacogenetic screening for polymorphisms in drug-metabolizing enzymes and drug transporters in a Dutch population. Mol Diagn Ther 2006;10:175-85
  • Han JY, Lim HS, Yoo YK, Associations of ABCB1, ABCC2, and ABCG2 polymorphisms with irinotecan-pharmacokinetics and clinical outcome in patients with advanced non-small cell lung cancer. Cancer 2007;110:138-47
  • Innocenti F, Kroetz DL, Schuetz E, Comprehensive pharmacogenetic analysis of irinotecan neutropenia and pharmacokinetics. J Clin Oncol 2009;27(16):2604-14
  • Haenisch S, Zimmermann U, Dazert E, Influence of polymorphisms of ABCB1 and ABCC2 on mRNA and protein expression in normal and cancerous kidney cortex. Pharmacogenomics J 2007;7:56-65
  • Innocenti F, Undevia SD, Chen PX, Pharmacogenetic analysis of interindividual irinotecan (CPT-11) pharmacokinetic (PK) variability: evidence for a functional variant of ABCC2 [abstract no: 2010]. Volume 22. No 14S 2004 ASCO Annual Meeting Proceedings (Post-Meeting Edition)
  • Han JY, Lim HS, Park YH, Integrated pharmacogenetic prediction of irinotecan pharmacokinetics and toxicity in patients with advanced non-small cell lung cancer. Lung Cancer 2009;63:115-20
  • Tamura A, Wakabayashi K, Onishi Y, Re-evaluation and functional classification of non-synonymous single nucleotide polymorphisms of the human ATP-binding cassette transporter ABCG2. Cancer Sci 2007;98:231-9
  • Furukawa T, Wakabayashi K, Tamura A, Major SNP (Q141K) variant of human ABC transporter ABCG2 undergoes lysosomal and proteasomal degradations. Pharm Res 2009;26:469-79
  • Bosch TM, Kjellberg LM, Bouwers A, Detection of single nucleotide polymorphisms in the ABCG2 gene in a Dutch population. Am J Pharmacogenomics 2005;5:123-31
  • Maekawa K, Itoda M, Sai K, Genetic variation and haplotype structure of the ABC transporter gene ABCG2 in a Japanese population. Drug Metab Pharmacokinet 2006;21:109-21
  • de Jong FA, Marsh S, Mathijssen RH, ABCG2 pharmacogenetics: ethnic differences in allele frequency and assessment of influence on irinotecan disposition. Clin Cancer Res 2004;10:5889-94
  • Cha PC, Mushiroda T, Zembutsu H, Single nucleotide polymorphism in ABCG2 is associated with irinotecan-induced severe myelosuppression. J Hum Genet 2009;54:572-80
  • Tirona RG, Leake BF, Merino G, Kim RB. Polymorphisms in OATP-C: identification of multiple allelic variants associated with altered transport activity among European- and African-Americans. J Biol Chem 2001;276:35669-75
  • Maeda K, Ieiri I, Yasuda K, Effects of organic anion transporting polypeptide 1B1 haplotype on pharmacokinetics of pravastatin, valsartan, and temocapril. Clin Pharmacol Ther 2006;79:427-39
  • Xiang X, Jada SR, Li HH, Pharmacogenetics of SLCO1B1 gene and the impact of *1b and *15 haplotypes on irinotecan disposition in Asian cancer patients. Pharmacogenet Genomics 2006;16:683-91
  • Kim SR, Saito Y, Sai K, Genetic variations and frequencies of major haplotypes in SLCO1B1 encoding the transporter OATP1B1 in Japanese subjects: SLCO1B1*17 is more prevalent than *15. Drug Metab Pharmacokinet 2007;22:456-61
  • Takane H, Miyata M, Burioka N, Severe toxicities after irinotecan-based chemotherapy in a patient with lung cancer: a homozygote for the SLCO1B1*15 allele. Ther Drug Monit 2007;29:666-8
  • Takane H, Kawamoto K, Sasaki T, Life-threatening toxicities in a patient with UGT1A1*6/*28 and SLCO1B1*15/*15 genotypes after irinotecan-based chemotherapy. Cancer Chemother Pharmacol 2009;63:1165-9
  • Sakaguchi S, Garcia-Bournissen F, Kim R, Prolonged neutropenia after irinotecan-based chemotherapy in a child with polymorphisms of UGT1A1 and SLCO1B1. Arch Dis Child 2009;94:981-2
  • Mathijssen RH, de Jong FA, van Schaik RH, Prediction of irinotecan pharmacokinetics by use of cytochrome P450 3A4 phenotyping probes. J Natl Cancer Inst 2004;96:1585-92
  • Mathijssen RH, Marsh S, Karlsson MO, Irinotecan pathway genotype analysis to predict pharmacokinetics. Clin Cancer Res 2003;9:3246-53
  • Pillot GA, Read WL, Hennenfent KL, A phase II study of irinotecan and carboplatin in advanced non-small cell lung cancer with pharmacogenomic analysis: final report. J Thorac Oncol 2006;1:972-8
  • Sai K, Saito Y, Fukushima-Uesaka H, Impact of CYP3A4 haplotypes on irinotecan pharmacokinetics in Japanese cancer patients. Cancer Chemother Pharmacol 2008;62:529-37
  • Murayama N, Nakamura T, Saeki M, CYP3A4 gene polymorphisms influence testosterone 6beta-hydroxylation. Drug Metab Pharmacokinet 2002;17:150-6
  • Maekawa K, Harakawa N, Yoshimura T, CYP3A4*16 and CYP3A4*18 Alleles Found in East Asians Exhibit Differential Catalytic Activities for Seven CYP3A4 Substrate Drugs. Drug Metab Dispos 2010;38:2100-4
  • Dai D, Tang J, Rose R, Identification of variants of CYP3A4 and characterization of their abilities to metabolize testosterone and chlorpyrifos. J Pharmacol Exp Ther 2001;299:825-31
  • van der Bol JM, Mathijssen RH, Creemers GJ, A CYP3A4 phenotype-based dosing algorithm for individualized treatment of irinotecan. Clin Cancer Res 2010;16:736-42
  • Humerickhouse R, Lohrbach K, Li L, Characterization of CPT-11 hydrolysis by human liver carboxylesterase isoforms hCE-1 and hCE-2. Cancer Res 2000;60:1189-92
  • Kubo T, Kim SR, Sai K, Functional characterization of three naturally occurring single nucleotide polymorphisms in the CES2 gene encoding carboxylesterase 2 (HCE-2). Drug Metab Dispos 2005;33:1482-7
  • Kim SR, Sai K, Tanaka-Kagawa T, Haplotypes and a novel defective allele of CES2 found in a Japanese population. Drug Metab Dispos 2007;35:1865-72
  • Hosokawa M. Structure and catalytic properties of carboxylesterase isozymes involved in metabolic activation of prodrugs. Molecules 2008;13:412-31
  • Tanimoto K, Kaneyasu M, Shimokuni T, Human carboxylesterase 1A2 expressed from carboxylesterase 1A1 and 1A2 genes is a potent predictor of CPT-11 cytotoxicity in vitro. Pharmacogenet Genomics 2007;17:1-10
  • Fukami T, Nakajima M, Maruichi T, Structure and characterization of human carboxylesterase 1A1, 1A2, and 1A3 genes. Pharmacogenet Genomics 2008;18:911-20
  • Sai K, Saito Y, Tatewaki N, Association of carboxylesterase 1A genotypes with irinotecan pharmacokinetics in Japanese cancer patients. Br J Clin Pharmacol 2010;70:222-33
  • Toffoli G, Cecchin E, Gasparini G, Genotype-driven phase I study of irinotecan administered in combination with fluorouracil/leucovorin in patients with metastatic colorectal cancer. J Clin Oncol 2010;28:866-71
  • Hoskins JM, Marcuello E, Altes A, Irinotecan pharmacogenetics: influence of pharmacodynamic genes. Clin Cancer Res 2008;14:1788-96
  • Artac M, Bozcuk H, Pehlivan S, The value of XPD and XRCC1 genotype polymorphisms to predict clinical outcome in metastatic colorectal carcinoma patients with irinotecan-based regimens. J Cancer Res Clin Oncol 2010;136:803-9
  • Hoskins JM, Goldberg RM, Qu P, UGT1A1*28 genotype and irinotecan-induced neutropenia: dose matters. J Natl Cancer Inst 2007;99:1290-5
  • Hu ZY, Yu Q, Pei Q, Guo C. Dose-dependent association between UGT1A1*28 genotype and irinotecan-induced neutropenia: low doses also increase risk. Clin Cancer Res 2010;16:3832-42
  • Noble S, Goa KL. Gemcitabine. A review of its pharmacology and clinical potential in non-small cell lung cancer and pancreatic cancer. Drugs 1997;54:447-72
  • Ueno H, Kiyosawa K, Kaniwa N. Pharmacogenomics of gemcitabine: can genetic studies lead to tailor-made therapy? Br J Cancer 2007;97:145-51
  • Green MR. Gemcitabine safety overview. Semin Oncol 1996;23:32-5
  • Gilbert JA, Salavaggione OE, Ji Y, Gemcitabine pharmacogenomics: cytidine deaminase and deoxycytidylate deaminase gene resequencing and functional genomics. Clin Cancer Res 2006;12:1794-803
  • Fukunaga AK, Marsh S, Murry DJ, Identification and analysis of single-nucleotide polymorphisms in the gemcitabine pharmacologic pathway. Pharmacogenomics J 2004;4:307-14
  • Yue LJ, Chen XW, Li CR, Single-nucleotide polymorphisms of the cytidine deaminase gene in childhood with acute leukemia and normal Chinese children. Zhonghua Yi Xue Yi Chuan Xue Za Zhi 2007;24:699-702
  • Sugiyama E, Lee SJ, Lee SS, Ethnic differences of two non-synonymous single nucleotide polymorphisms in CDA gene. Drug Metab Pharmacokinet 2009;24:553-6
  • Giovannetti E, Laan AC, Vasile E, Correlation between cytidine deaminase genotype and gemcitabine deamination in blood samples. Nucleosides Nucleotides Nucleic Acids 2008;27:720-5
  • . Tibaldi C, Giovannetti E, Vasile E, Correlation of CDA. ERCC1, and XPD polymorphisms with response and survival in gemcitabine/cisplatin-treated advanced non-small cell lung cancer patients. Clin Cancer Res 2008;14:1797-803
  • Chew HK, Doroshow JH, Frankel P, Phase II studies of gemcitabine and cisplatin in heavily and minimally pretreated metastatic breast cancer. J Clin Oncol 2009;27:2163-9
  • Maring JG, Wachters FM, Slijfer M, Pharmacokinetics of gemcitabine in non-small-cell lung cancer patients: impact of the 79A>C cytidine deaminase polymorphism. Eur J Clin Pharmacol 2010;66:611-17
  • Ciccolini J, Dahan L, Andre N, Cytidine deaminase residual activity in serum is a predictive marker of early severe toxicities in adults after gemcitabine-based chemotherapies. J Clin Oncol 2010;28:160-5
  • Sugiyama E, Kaniwa N, Kim SR, Pharmacokinetics of gemcitabine in Japanese cancer patients: the impact of a cytidine deaminase polymorphism. J Clin Oncol 2007;25(8):32-42
  • Giovannetti E, Tibaldi C, Falcone A, Impact of cytidine deaminase polymorphisms on toxicity after gemcitabine: the question is still ongoing. J Clin Oncol 2010;28:e221-2
  • Sugiyama E, Kaniwa N, Kim SR, Population pharmacokinetics of gemcitabine and its metabolite in Japanese cancer patients: impact of genetic polymorphisms. Clin Pharmacokinet 2010;49:549-58
  • Yonemori K, Ueno H, Okusaka T, Severe drug toxicity associated with a single-nucleotide polymorphism of the cytidine deaminase gene in a Japanese cancer patient treated with gemcitabine plus cisplatin. Clin Cancer Res 2005;11:2620-4
  • Ueno H, Kaniwa N, Okusaka T, Homozygous CDA*3 is a major cause of life-threatening toxicities in gemcitabine-treated Japanese cancer patients. Br J Cancer 2009;100:870-3
  • Mercier C, Raynal C, Dahan L, Toxic death case in a patient undergoing gemcitabine-based chemotherapy in relation with cytidine deaminase downregulation. Pharmacogenet Genomics 2007;17:841-4
  • Soo RA, Wang LZ, Ng SS, Distribution of gemcitabine pathway genotypes in ethnic Asians and their association with outcome in non-small cell lung cancer patients. Lung Cancer 2009;63:121-7
  • Gray JH, Mangravite LM, Owen RP, Functional and genetic diversity in the concentrative nucleoside transporter, CNT1, in human populations. Mol Pharmacol 2004;65:512-19
  • Okazaki T, Javle M, Tanaka M, Single nucleotide polymorphisms of gemcitabine metabolic genes and pancreatic cancer survival and drug toxicity. Clin Cancer Res 2010;16:320-9
  • Tanaka M, Javle M, Dong X, Gemcitabine metabolic and transporter gene polymorphisms are associated with drug toxicity and efficacy in patients with locally advanced pancreatic cancer. Cancer 2010;116:5325-35
  • Rha SY, Jeung HC, Choi YH, An association between RRM1 haplotype and gemcitabine-induced neutropenia in breast cancer patients. Oncologist 2007;12:622-30
  • Dong S, Guo AL, Chen ZH, RRM1 single nucleotide polymorphism -37C>A correlates with progression-free survival in NSCLC patients after gemcitabine-based chemotherapy. J Hematol Oncol 2010;3:10
  • Rodriguez J, Boni V, Hernandez A, Association of RRM1 -37A>C polymorphism with clinical outcome in colorectal cancer patients treated with gemcitabine-based chemotherapy. Eur J Cancer 2011; published online 8 January 2011, doi:10.1016/j.ejca.2010.11.032.
  • Riggs BL, Hartmann LC. Selective estrogen-receptor modulators -- mechanisms of action and application to clinical practice. N Engl J Med 2003;348:618-29
  • Lien EA, Solheim E, Lea OA, Distribution of 4-hydroxy-N-desmethyltamoxifen and other tamoxifen metabolites in human biological fluids during tamoxifen treatment. Cancer Res 1989;49:2175-83
  • Ingelman-Sundberg M. Genetic polymorphisms of cytochrome P450 2D6 (CYP2D6): clinical consequences, evolutionary aspects and functional diversity. Pharmacogenomics J 2005;5:6-13
  • Bradford LD. CYP2D6 allele frequency in European Caucasians, Asians, Africans and their descendants. Pharmacogenomics 2002;3:229-43
  • Sistonen J, Sajantila A, Lao O, CYP2D6 worldwide genetic variation shows high frequency of altered activity variants and no continental structure. Pharmacogenet Genomics 2007;17:93-101
  • Qin S, Shen L, Zhang A, Systematic polymorphism analysis of the CYP2D6 gene in four different geographical Han populations in mainland China. Genomics 2008;92:152-8
  • Lee SJ, Lee SS, Jung HJ, Discovery of novel functional variants and extensive evaluation of CYP2D6 genetic polymorphisms in Koreans. Drug Metab Dispos 2009;37:1464-70
  • Iwashima K, Yasui-Furukori N, Kaneda A, No association between CYP2D6 polymorphisms and personality trait in Japanese. Br J Clin Pharmacol 2007;64:96-9
  • Soyama A, Saito Y, Kubo T, Sequence-based analysis of the CYP2D6*36-CYP2D6*10 tandem-type arrangement, a major CYP2D6*10 haplotype in the Japanese population. Drug Metab Pharmacokinet 2006;21:208-16
  • Borges S, Desta Z, Li L, Quantitative effect of CYP2D6 genotype and inhibitors on tamoxifen metabolism: implication for optimization of breast cancer treatment. Clin Pharmacol Ther 2006;80:61-74
  • Lim HS, Ju Lee H, Seok Lee K, Clinical implications of CYP2D6 genotypes predictive of tamoxifen pharmacokinetics in metastatic breast cancer. J Clin Oncol 2007;25:3837-45
  • Kiyotani K, Mushiroda T, Imamura CK, Significant effect of polymorphisms in CYP2D6 and ABCC2 on clinical outcomes of adjuvant tamoxifen therapy for breast cancer patients. J Clin Oncol 2010;28:1287-93
  • Schroth W, Goetz MP, Hamann U, Association between CYP2D6 polymorphisms and outcomes among women with early stage breast cancer treated with tamoxifen. JAMA 2009;302:1429-36
  • Goetz MP, Rae JM, Suman VJ, Pharmacogenetics of tamoxifen biotransformation is associated with clinical outcomes of efficacy and hot flashes. J Clin Oncol 2005;23:9312-18
  • Schroth W, Antoniadou L, Fritz P, Breast cancer treatment outcome with adjuvant tamoxifen relative to patient CYP2D6 and CYP2C19 genotypes. J Clin Oncol 2007;25:5187-93
  • Nowell SA, Ahn J, Rae JM, Association of genetic variation in tamoxifen-metabolizing enzymes with overall survival and recurrence of disease in breast cancer patients. Breast Cancer Res Treat 2005;91:249-58
  • Wegman P, Vainikka L, Stal O, Genotype of metabolic enzymes and the benefit of tamoxifen in postmenopausal breast cancer patients. Breast Cancer Res 2005;7:R284-90
  • Wegman P, Elingarami S, Carstensen J, Genetic variants of CYP3A5, CYP2D6, SULT1A1, UGT2B15 and tamoxifen response in postmenopausal patients with breast cancer. Breast Cancer Res 2007;9:R7
  • Schroth W, Hamann U, Fasching PA, CYP2D6 polymorphisms as predictors of outcome in breast cancer patients treated with tamoxifen: expanded polymorphism coverage improves risk stratification. Clin Cancer Res 2010;16.4468-77
  • Kiyotani K, Mushiroda T, Hosono N, Lessons for pharmacogenomics studies: association study between CYP2D6 genotype and tamoxifen response. Pharmacogenet Genomics 2010;20:565-8
  • Thompson AM, Johnson A, Quinlan P, Comprehensive CYP2D6 genotype and adherence affect outcome in breast cancer patients treated with tamoxifen monotherapy. Breast Cancer Res Treat 2011;125:279-87
  • Dezentje VO, van Blijderveen NJ, Gelderblom H, Effect of concomitant CYP2D6 inhibitor use and tamoxifen adherence on breast cancer recurrence in early-stage breast cancer. J Clin Oncol 2010;28:2423-9
  • Osborne CK. Tamoxifen in the treatment of breast cancer. N Engl J Med 1998;339:1609-18
  • Mortimer JE, Flatt SW, Parker BA, Tamoxifen, hot flashes and recurrence in breast cancer. Breast Cancer Res Treat 2008;108:421-6
  • Henry NL, Rae JM, Li L, Association between CYP2D6 genotype and tamoxifen-induced hot flashes in a prospective cohort. Breast Cancer Res Treat 2009;117:571-5
  • Bonanni B, Macis D, Maisonneuve P, Polymorphism in the CYP2D6 tamoxifen-metabolizing gene influences clinical effect but not hot flashes: data from the Italian Tamoxifen Trial. J Clin Oncol 2006;24:3708-9
  • Lara PN Jr, Chansky K, Shibata T, Common arm comparative outcomes analysis of phase 3 trials of cisplatin + irinotecan versus cisplatin + etoposide in extensive stage small cell lung cancer1: final patient-level results from Japan Clinical Oncology Group 9511 and Southwest Oncology Group 0124. Cancer 2010; published on line 24 August 2010, doi: 10.1002/cncr.25532.
  • Sanoff HK, Sargent DJ, Green EM, Racial differences in advanced colorectal cancer outcomes and pharmacogenetics: a subgroup analysis of a large randomized clinical trial. J Clin Oncol 2009;27:4109-15
  • Kiyotani K, Mushiroda T, Nakamura Y, Reply to T. Lang et al. J Clin Oncol 2010;28:e449

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.