Skip to main content
Top
Published in: Clinical Pharmacokinetics 5/2000

01-11-2000 | Leading Article

Pharmacogenetics

A Tool for Individualising Antineoplastic Therapy

Authors: Federico Innocenti, Lalitha Iyer, Dr Mark J. Ratain

Published in: Clinical Pharmacokinetics | Issue 5/2000

Login to get access

Abstract

This article reviews the clinical relevance of pharmacogenetics in cancer chemotherapy, with emphasis on drugs for which genetic differences in enzyme metabolism have been demonstrated to affect patient outcome.
About 10% of children with leukaemia are intolerant to mercaptopurine (6-mercaptopurine) because of genetic defects in mercaptopurine inactivation by thiopurine S-methyltransferase. However, mercaptopurine dose intensity, a critical factor for outcome in patients deficient in thiopurine S-methyltransferase, can be maintained by means of thiopurine S-methyltransferase phenotyping or genotyping.
Patients with reduced fluorouracil (5-fluorouracil) catabolism are more likely to be exposed to severe toxicity. The measurement of dihydropyrimidine dehydrogenase activity in patients cannot be considered fully predictive, and the role of dihydropyrimidine dehydrogenase gene variants in this syndrome has yet to be clarified.
With regard to irinotecan, patients with Gilbert’s syndrome phenotype have reduced inactivation of the active topoisomerase I inhibitor 7-ethyl-10-hydroxy-camptothecin (SN-38) caused by a mutation in the UDP-glucuronosyltransferase 1A1 gene promoter. This subset of patients is more likely to be exposed to irinotecan toxicity and could be identified by genotyping for gene promoter variants.
Finally, the experience with amonafide represents a model for dose individualisation approaches that use simple phenotypic probes.
Literature
1.
go back to reference Chabot GG. Factors involved in clinical pharmacology variability in oncology. Anticancer Res 1994; 14: 2269–72.PubMed Chabot GG. Factors involved in clinical pharmacology variability in oncology. Anticancer Res 1994; 14: 2269–72.PubMed
2.
go back to reference Iyer L, Ratain MJ. Pharmacogenetics and cancer chemotherapy. Eur J Cancer 1998; 34: 1493–9.PubMed Iyer L, Ratain MJ. Pharmacogenetics and cancer chemotherapy. Eur J Cancer 1998; 34: 1493–9.PubMed
3.
go back to reference Iyer L. Inherited variations in drug-metabolizing enzymes: significance in clinical oncology. Mol Diagn 1999; 4: 327–33.PubMed Iyer L. Inherited variations in drug-metabolizing enzymes: significance in clinical oncology. Mol Diagn 1999; 4: 327–33.PubMed
4.
go back to reference Shi MM, Bleavins MR, de la Iglesia FA. Technologies for detecting genetic polymorphisms in pharmacogenomics. Mol Diagn 1999; 4: 343–51.PubMed Shi MM, Bleavins MR, de la Iglesia FA. Technologies for detecting genetic polymorphisms in pharmacogenomics. Mol Diagn 1999; 4: 343–51.PubMed
5.
go back to reference Linder MW, Russell AP, Valdes RJ. Pharmacogenetics: a laboratory tool for optimizing therapeutic efficiency. Clin Chem 1997; 43: 254–66.PubMed Linder MW, Russell AP, Valdes RJ. Pharmacogenetics: a laboratory tool for optimizing therapeutic efficiency. Clin Chem 1997; 43: 254–66.PubMed
6.
go back to reference Tidd DM, Paterson AR. A biochemical mechanism for the delayed cytotoxic reaction of 6-mercaptopurine. Cancer Res 1974; 34: 738–46.PubMed Tidd DM, Paterson AR. A biochemical mechanism for the delayed cytotoxic reaction of 6-mercaptopurine. Cancer Res 1974; 34: 738–46.PubMed
7.
go back to reference Deininger M, Szumlanski CL, Otterness DM, et al. Purine substrates for human thiopurine methyltransferase. Biochem Pharmacol 1994; 48: 2135–8.PubMed Deininger M, Szumlanski CL, Otterness DM, et al. Purine substrates for human thiopurine methyltransferase. Biochem Pharmacol 1994; 48: 2135–8.PubMed
8.
go back to reference Woodson LC, Ames MM, Selassie CD, et al. Thiopurine methyltransferase: aromatic thiol substrates and inhibition by benzoic acid derivatives. Mol Pharmacol 1983; 24: 471–8.PubMed Woodson LC, Ames MM, Selassie CD, et al. Thiopurine methyltransferase: aromatic thiol substrates and inhibition by benzoic acid derivatives. Mol Pharmacol 1983; 24: 471–8.PubMed
9.
go back to reference Lennard L, Lilleyman JS, Van Loon J, et al. Genetic variation in response to 6-mercaptopurine for childhood acute lymphoblastic leukaemia. Lancet 1990; 336: 225–9.PubMed Lennard L, Lilleyman JS, Van Loon J, et al. Genetic variation in response to 6-mercaptopurine for childhood acute lymphoblastic leukaemia. Lancet 1990; 336: 225–9.PubMed
10.
go back to reference Lilleyman JS, Lennard L. Mercaptopurine metabolism and risk of relapse in childhood lymphoblastic leukaemia. Lancet 1994; 343: 1188–90.PubMed Lilleyman JS, Lennard L. Mercaptopurine metabolism and risk of relapse in childhood lymphoblastic leukaemia. Lancet 1994; 343: 1188–90.PubMed
11.
go back to reference Andersen JB, Szumlanski C, Weinshilboum RM, et al. Pharmacokinetics, dose adjustments, and 6-mercaptopurine/methotrexate drug interactions in two patients with thiopurine methyltransferase deficiency. Acta Paediatr 1998; 87: 108–11.PubMed Andersen JB, Szumlanski C, Weinshilboum RM, et al. Pharmacokinetics, dose adjustments, and 6-mercaptopurine/methotrexate drug interactions in two patients with thiopurine methyltransferase deficiency. Acta Paediatr 1998; 87: 108–11.PubMed
12.
go back to reference McLeod HL, Coulthard S, Thomas AE, et al. Analysis of thiopurine methyltransferase variant alleles in childhood acute lymphoblastic leukaemia. Br J Haematol 1999; 105: 696–700.PubMed McLeod HL, Coulthard S, Thomas AE, et al. Analysis of thiopurine methyltransferase variant alleles in childhood acute lymphoblastic leukaemia. Br J Haematol 1999; 105: 696–700.PubMed
13.
go back to reference Lennard L, Gibson BE, Nicole T, et al. Congenital thiopurine methyltransferase deficiency and 6-mercaptopurine toxicity during treatment for acute lymphoblastic leukaemia. Arch Dis Child 1993; 69: 577–9.PubMed Lennard L, Gibson BE, Nicole T, et al. Congenital thiopurine methyltransferase deficiency and 6-mercaptopurine toxicity during treatment for acute lymphoblastic leukaemia. Arch Dis Child 1993; 69: 577–9.PubMed
14.
go back to reference Lennard L, Lewis IJ, Michelagnoli M, et al. Thiopurine methyltransferase deficiency in childhood lymphoblastic leukaemia: 6-mercaptopurine dosage strategies. Med Pediatr Oncol 1997; 29: 252–5.PubMed Lennard L, Lewis IJ, Michelagnoli M, et al. Thiopurine methyltransferase deficiency in childhood lymphoblastic leukaemia: 6-mercaptopurine dosage strategies. Med Pediatr Oncol 1997; 29: 252–5.PubMed
15.
go back to reference Evans WE, Horner M, Chu YQ, et al. Altered mercaptopurine metabolism, toxic effects, and dosage requirement in a thiopurine methyltransferase-deficient child with acute lymphocytic leukemia. J Pediatr 1991; 119: 985–9.PubMed Evans WE, Horner M, Chu YQ, et al. Altered mercaptopurine metabolism, toxic effects, and dosage requirement in a thiopurine methyltransferase-deficient child with acute lymphocytic leukemia. J Pediatr 1991; 119: 985–9.PubMed
16.
go back to reference Relling MV, Hancock ML, Boyett JM, et al. Prognostic importance of 6-mercaptopurine dose intensity in acute lymphoblastic leukemia. Blood 1999; 93: 2817–23.PubMed Relling MV, Hancock ML, Boyett JM, et al. Prognostic importance of 6-mercaptopurine dose intensity in acute lymphoblastic leukemia. Blood 1999; 93: 2817–23.PubMed
17.
go back to reference Dibenedetto SP, Guardabasso V, Ragusa R, et al. 6-Mercapto-purine cumulative dose: a critical factor of maintenance therapy in average risk childhood acute lymphoblastic leukemia. Pediatr Hematol Oncol 1994; 11: 251–8.PubMed Dibenedetto SP, Guardabasso V, Ragusa R, et al. 6-Mercapto-purine cumulative dose: a critical factor of maintenance therapy in average risk childhood acute lymphoblastic leukemia. Pediatr Hematol Oncol 1994; 11: 251–8.PubMed
18.
go back to reference Szumlanski C, Otterness D, Her C, et al. Thiopurine methyltransferase pharmacogenetics: human gene cloning and characterization of a common polymorphism. DNA Cell Biol 1996; 15: 17–30.PubMed Szumlanski C, Otterness D, Her C, et al. Thiopurine methyltransferase pharmacogenetics: human gene cloning and characterization of a common polymorphism. DNA Cell Biol 1996; 15: 17–30.PubMed
19.
go back to reference Yates CR, Krynetski EY, Loennechen T, et al. Molecular diagnosis of thiopurine S-methyltransferase deficiency: genetic basis for azathioprine and mercaptopurine intolerance. Ann Intern Med 1997; 126: 608–14.PubMed Yates CR, Krynetski EY, Loennechen T, et al. Molecular diagnosis of thiopurine S-methyltransferase deficiency: genetic basis for azathioprine and mercaptopurine intolerance. Ann Intern Med 1997; 126: 608–14.PubMed
20.
go back to reference Otterness D, Szumlanski C, Lennard L, et al. Human thiopurine methyltransferase pharmacogenetics: gene sequence polymorphisms. Clin Pharmacol Ther 1997; 62: 60–73.PubMed Otterness D, Szumlanski C, Lennard L, et al. Human thiopurine methyltransferase pharmacogenetics: gene sequence polymorphisms. Clin Pharmacol Ther 1997; 62: 60–73.PubMed
21.
go back to reference Tai HL, Krynetski EY, Yates CR, et al. Thiopurine S-methyl-transferase deficiency: two nucleotide transitions define the most prevalent mutant allele associated with loss of catalytic activity in Caucasians. Am J Hum Genet 1996; 58: 694–702.PubMed Tai HL, Krynetski EY, Yates CR, et al. Thiopurine S-methyl-transferase deficiency: two nucleotide transitions define the most prevalent mutant allele associated with loss of catalytic activity in Caucasians. Am J Hum Genet 1996; 58: 694–702.PubMed
22.
go back to reference Otterness DM, Szumlanski CL, Wood TC, et al. Human thiopurine methyltransferase pharmacogenetics: kindred with a terminal exon splice junction mutation that results in loss of activity. J Clin Invest 1998; 101: 1036–44.PubMed Otterness DM, Szumlanski CL, Wood TC, et al. Human thiopurine methyltransferase pharmacogenetics: kindred with a terminal exon splice junction mutation that results in loss of activity. J Clin Invest 1998; 101: 1036–44.PubMed
23.
go back to reference Hon YY, Fessing MY, Pui CH, et al. Polymorphism of the thiopurine S-methyltransferase gene in African-Americans. Hum Mol Genet 1999; 8: 371–6.PubMed Hon YY, Fessing MY, Pui CH, et al. Polymorphism of the thiopurine S-methyltransferase gene in African-Americans. Hum Mol Genet 1999; 8: 371–6.PubMed
24.
go back to reference Collie-Duguid ES, Pritchard SC, Powrie RH, et al. The frequency and distribution of thiopurine methyltransferase alleles in Caucasian and Asian populations. Pharmacogenetics 1999; 9: 37–42.PubMed Collie-Duguid ES, Pritchard SC, Powrie RH, et al. The frequency and distribution of thiopurine methyltransferase alleles in Caucasian and Asian populations. Pharmacogenetics 1999; 9: 37–42.PubMed
25.
go back to reference McLeod HL, Pritchard SC, Githang’a J, et al. Ethnic differences in thiopurine methyltransferase pharmacogenetics: evidence for allele specificity in Caucasian and Kenyan individuals. Pharmacogenetics 1999; 9: 773–6.PubMed McLeod HL, Pritchard SC, Githang’a J, et al. Ethnic differences in thiopurine methyltransferase pharmacogenetics: evidence for allele specificity in Caucasian and Kenyan individuals. Pharmacogenetics 1999; 9: 773–6.PubMed
26.
go back to reference Ameyaw MM, Collie-Duguid ES, Powrie RH, et al. Thiopurine methyltransferase alleles in British and Ghanaian populations. Hum Mol Genet 1999; 8: 367–70.PubMed Ameyaw MM, Collie-Duguid ES, Powrie RH, et al. Thiopurine methyltransferase alleles in British and Ghanaian populations. Hum Mol Genet 1999; 8: 367–70.PubMed
27.
go back to reference Hiratsuka M, Inoue T, Omori F, et al. Genetic analysis of thiopurine methyltransferase polymorphism in a Japanese population. Mutat Res 2000; 448: 91–5.PubMed Hiratsuka M, Inoue T, Omori F, et al. Genetic analysis of thiopurine methyltransferase polymorphism in a Japanese population. Mutat Res 2000; 448: 91–5.PubMed
28.
go back to reference Relling MV, Hancock ML, Rivera GK, et al. Mercaptopurine therapy intolerance and heterozygosity at the thiopurine S-methyltransferase gene locus. J Natl Cancer Inst 1999; 91: 2001–8.PubMed Relling MV, Hancock ML, Rivera GK, et al. Mercaptopurine therapy intolerance and heterozygosity at the thiopurine S-methyltransferase gene locus. J Natl Cancer Inst 1999; 91: 2001–8.PubMed
29.
go back to reference McLeod HL, Krynetski EY, Relling MV, et al. Genetic polymorphism of thiopurine methyltransferase and its clinical relevance for childhood acute lymphoblastic leukemia. Leukemia 2000; 14: 567–72.PubMed McLeod HL, Krynetski EY, Relling MV, et al. Genetic polymorphism of thiopurine methyltransferase and its clinical relevance for childhood acute lymphoblastic leukemia. Leukemia 2000; 14: 567–72.PubMed
30.
go back to reference Weinshilboum RM, Sladek SL. Mercaptopurine pharmacogenetics: monogenic inheritance of erythrocyte thiopurine methyltransferase activity. Am J Hum Genet 1980; 32: 651–62.PubMed Weinshilboum RM, Sladek SL. Mercaptopurine pharmacogenetics: monogenic inheritance of erythrocyte thiopurine methyltransferase activity. Am J Hum Genet 1980; 32: 651–62.PubMed
31.
go back to reference McLeod HL, Lin JS, Scott EP, et al. Thiopurine methyltransferase activity in American white subjects and black subjects. Clin Pharmacol Ther 1994; 55: 15–20.PubMed McLeod HL, Lin JS, Scott EP, et al. Thiopurine methyltransferase activity in American white subjects and black subjects. Clin Pharmacol Ther 1994; 55: 15–20.PubMed
32.
go back to reference McLeod HL, Relling MV, Liu Q, et al. Polymorphic thiopurine methyltransferase in erythrocytes is indicative of activity in leukemic blasts from children with acute lymphoblastic leukemia. Blood 1995; 85: 1897–902.PubMed McLeod HL, Relling MV, Liu Q, et al. Polymorphic thiopurine methyltransferase in erythrocytes is indicative of activity in leukemic blasts from children with acute lymphoblastic leukemia. Blood 1995; 85: 1897–902.PubMed
33.
go back to reference Lennard L, Van Loon JA, Lilleyman JS, et al. Thiopurine pharmacogenetics in leukemia: correlation of erythrocyte thiopurine methyltransferase activity and 6-thioguanine nucleotide concentrations. Clin Pharmacol Ther 1987; 41: 18–25.PubMed Lennard L, Van Loon JA, Lilleyman JS, et al. Thiopurine pharmacogenetics in leukemia: correlation of erythrocyte thiopurine methyltransferase activity and 6-thioguanine nucleotide concentrations. Clin Pharmacol Ther 1987; 41: 18–25.PubMed
34.
go back to reference Lennard L, Lilleyman JS. Variable mercaptopurine metabolism and treatment outcome in childhood lymphoblastic leukemia. J Clin Oncol 1989; 7: 1816–23.PubMed Lennard L, Lilleyman JS. Variable mercaptopurine metabolism and treatment outcome in childhood lymphoblastic leukemia. J Clin Oncol 1989; 7: 1816–23.PubMed
35.
go back to reference Lennard L, Welch JC, Lilleyman JS. Thiopurine drugs in the treatment of childhood leukaemia: the influence of inherited thiopurine methyltransferase activity on drug metabolism and cytotoxicity. Br J Clin Pharmacol 1997; 44: 455–61.PubMed Lennard L, Welch JC, Lilleyman JS. Thiopurine drugs in the treatment of childhood leukaemia: the influence of inherited thiopurine methyltransferase activity on drug metabolism and cytotoxicity. Br J Clin Pharmacol 1997; 44: 455–61.PubMed
36.
go back to reference Coulthard SA, Howell C, Robson J, et al. The relationship between thiopurine methyltransferase activity and genotype in blasts from patients with acute leukemia. Blood 1998; 92: 2856–62.PubMed Coulthard SA, Howell C, Robson J, et al. The relationship between thiopurine methyltransferase activity and genotype in blasts from patients with acute leukemia. Blood 1998; 92: 2856–62.PubMed
37.
go back to reference Welch JC, Lilleyman JS. 6-Mercaptopurine dose escalation and its effect on drug tolerance in childhood lymphoblastic leukaemia. Cancer Chemother Pharmacol 1996; 38: 113–6.PubMed Welch JC, Lilleyman JS. 6-Mercaptopurine dose escalation and its effect on drug tolerance in childhood lymphoblastic leukaemia. Cancer Chemother Pharmacol 1996; 38: 113–6.PubMed
38.
go back to reference Ho DH, Townsend L, Luna MA, et al. Distribution and inhibition of dihydrouracil dehydrogenase activities in human tissues using 5-fluorouracil as a substrate. Anticancer Res 1986; 6: 781–4.PubMed Ho DH, Townsend L, Luna MA, et al. Distribution and inhibition of dihydrouracil dehydrogenase activities in human tissues using 5-fluorouracil as a substrate. Anticancer Res 1986; 6: 781–4.PubMed
39.
go back to reference Lu Z, Zhang R, Diasio RB. Dihydropyrimidine dehydrogenase activity in human peripheral blood mononuclear cells and liver: population characteristics, newly identified deficient patients, and clinical implication in 5-fluorouracil chemotherapy. Cancer Res 1993; 53: 5433–8.PubMed Lu Z, Zhang R, Diasio RB. Dihydropyrimidine dehydrogenase activity in human peripheral blood mononuclear cells and liver: population characteristics, newly identified deficient patients, and clinical implication in 5-fluorouracil chemotherapy. Cancer Res 1993; 53: 5433–8.PubMed
40.
go back to reference Etienne MC, Lagrange JL, Dassonville O, et al. Population study of dihydropyrimidine dehydrogenase in cancer patients. J Clin Oncol 1994; 12: 2248–53.PubMed Etienne MC, Lagrange JL, Dassonville O, et al. Population study of dihydropyrimidine dehydrogenase in cancer patients. J Clin Oncol 1994; 12: 2248–53.PubMed
41.
go back to reference Harris BE, Song R, Soong SJ, et al. Relationship between dihydropyrimidine dehydrogenase activity and plasma 5-flu-orouracil levels with evidence for circadian variation of enzyme activity and plasma drug levels in cancer patients receiving 5-fluorouracil by protracted continuous infusion. Cancer Res 1990; 50: 197–201.PubMed Harris BE, Song R, Soong SJ, et al. Relationship between dihydropyrimidine dehydrogenase activity and plasma 5-flu-orouracil levels with evidence for circadian variation of enzyme activity and plasma drug levels in cancer patients receiving 5-fluorouracil by protracted continuous infusion. Cancer Res 1990; 50: 197–201.PubMed
42.
go back to reference Fleming RA, Milano G, Thyss A, et al. Correlation between dihydropyrimidine dehydrogenase activity in peripheral mononuclear cells and systemic clearance of fluorouracil in cancer patients. Cancer Res 1992; 52: 2899–902.PubMed Fleming RA, Milano G, Thyss A, et al. Correlation between dihydropyrimidine dehydrogenase activity in peripheral mononuclear cells and systemic clearance of fluorouracil in cancer patients. Cancer Res 1992; 52: 2899–902.PubMed
43.
go back to reference Johnson MR, Wang K, Tillmanns S, et al. Structural organization of the human dihydropyrimidine dehydrogenase gene. Cancer Res 1997; 57: 1660–3.PubMed Johnson MR, Wang K, Tillmanns S, et al. Structural organization of the human dihydropyrimidine dehydrogenase gene. Cancer Res 1997; 57: 1660–3.PubMed
44.
go back to reference Wei X, Elizondo G, Sapone A, et al. Characterization of the human dihydropyrimidine dehydrogenase gene. Genomics 1998; 51: 391–400.PubMed Wei X, Elizondo G, Sapone A, et al. Characterization of the human dihydropyrimidine dehydrogenase gene. Genomics 1998; 51: 391–400.PubMed
45.
go back to reference Meinsma R, Fernandez-Salguero P, Van Kuilenburg AB, et al. Human polymorphism in drug metabolism: mutation in the dihydropyrimidine dehydrogenase gene results in exon skipping and thymine uracilurea. DNA Cell Biol 1995; 14: 1–6.PubMed Meinsma R, Fernandez-Salguero P, Van Kuilenburg AB, et al. Human polymorphism in drug metabolism: mutation in the dihydropyrimidine dehydrogenase gene results in exon skipping and thymine uracilurea. DNA Cell Biol 1995; 14: 1–6.PubMed
46.
go back to reference Wei X, McLeod HL, McMurrough J, et al. Molecular basis of the human dihydropyrimidine dehydrogenase deficiency and 5-fluorouracil toxicity. J Clin Invest 1996; 98: 610–5.PubMed Wei X, McLeod HL, McMurrough J, et al. Molecular basis of the human dihydropyrimidine dehydrogenase deficiency and 5-fluorouracil toxicity. J Clin Invest 1996; 98: 610–5.PubMed
47.
go back to reference McLeod HL, Collie-Duguid ES, Vreken P, et al. Nomenclature for human DPYD alleles. Pharmacogenetics 1998; 8: 455–9.PubMed McLeod HL, Collie-Duguid ES, Vreken P, et al. Nomenclature for human DPYD alleles. Pharmacogenetics 1998; 8: 455–9.PubMed
48.
go back to reference Collie-Duguid ES, Etienne MC, Milano G, et al. Known variant DPYD alleles do not explain DPD deficiency in cancer patients. Pharmacogenetics 2000; 10: 217–23.PubMed Collie-Duguid ES, Etienne MC, Milano G, et al. Known variant DPYD alleles do not explain DPD deficiency in cancer patients. Pharmacogenetics 2000; 10: 217–23.PubMed
49.
go back to reference Vreken P, Van Kuilenburg AB, Meinsma R, et al. A point mutation in an invariant splice donor site leads to exon skipping in two unrelated Dutch patients with dihydropyrimidine dehydrogenase deficiency. J Inherit Metab Dis 1996; 19: 645–54.PubMed Vreken P, Van Kuilenburg AB, Meinsma R, et al. A point mutation in an invariant splice donor site leads to exon skipping in two unrelated Dutch patients with dihydropyrimidine dehydrogenase deficiency. J Inherit Metab Dis 1996; 19: 645–54.PubMed
50.
go back to reference Ridge SA, Sludden J, Brown O, et al. Dihydropyrimidine dehydrogenase pharmacogenetics in Caucasian subjects. Br J Clin Pharmacol 1998; 46: 151–6.PubMed Ridge SA, Sludden J, Brown O, et al. Dihydropyrimidine dehydrogenase pharmacogenetics in Caucasian subjects. Br J Clin Pharmacol 1998; 46: 151–6.PubMed
51.
go back to reference Van Kuilenburg AB, Vreken P, Beex LV, et al. Severe 5-fluorouracil toxicity caused by reduced dihydropyrimidine dehydrogenase activity due to heterozygosity for a G→A point mutation. J Inherit Metab Dis 1998; 21: 280–4.PubMed Van Kuilenburg AB, Vreken P, Beex LV, et al. Severe 5-fluorouracil toxicity caused by reduced dihydropyrimidine dehydrogenase activity due to heterozygosity for a G→A point mutation. J Inherit Metab Dis 1998; 21: 280–4.PubMed
52.
go back to reference Johnson MR, Hageboutros A, Wang K, et al. Life-threatening toxicity in a dihydropyrimidine dehydrogenase-deficient patient after treatment with topical 5-fluorouracil. Clin Cancer Res 1999; 5: 2006–11.PubMed Johnson MR, Hageboutros A, Wang K, et al. Life-threatening toxicity in a dihydropyrimidine dehydrogenase-deficient patient after treatment with topical 5-fluorouracil. Clin Cancer Res 1999; 5: 2006–11.PubMed
53.
go back to reference Ridge SA, Sludden J, Wei X, et al. Dihydropyrimidine dehydrogenase pharmacogenetics in patients with colorectal cancer. Br J Cancer 1998; 77: 497–500.PubMed Ridge SA, Sludden J, Wei X, et al. Dihydropyrimidine dehydrogenase pharmacogenetics in patients with colorectal cancer. Br J Cancer 1998; 77: 497–500.PubMed
54.
go back to reference Kouwaki M, Hamajima N, Sumi S, et al. Identification of novel mutations in the dihydropyrimidine dehydrogenase gene in a Japanese patient with 5-fluorouracil toxicity. Clin Cancer Res 1998; 4: 2999–3004.PubMed Kouwaki M, Hamajima N, Sumi S, et al. Identification of novel mutations in the dihydropyrimidine dehydrogenase gene in a Japanese patient with 5-fluorouracil toxicity. Clin Cancer Res 1998; 4: 2999–3004.PubMed
55.
go back to reference Collie-Duguid ES, Johnston SJ, Powrie RH, et al. Cloning and initial characterization of the human DPYD gene promoter. Biochem Biophys Res Commun 2000; 271: 28–35.PubMed Collie-Duguid ES, Johnston SJ, Powrie RH, et al. Cloning and initial characterization of the human DPYD gene promoter. Biochem Biophys Res Commun 2000; 271: 28–35.PubMed
56.
go back to reference Fleming RA, Milano GA, Gaspard MH, et al. Dihydropyrimidine dehydrogenase activity in cancer patients. Eur J Cancer 1993; 29A: 740–4.PubMed Fleming RA, Milano GA, Gaspard MH, et al. Dihydropyrimidine dehydrogenase activity in cancer patients. Eur J Cancer 1993; 29A: 740–4.PubMed
57.
go back to reference McMurrough J, McLeod HL. Analysis of the dihydropyrimidine dehydrogenase polymorphism in a British population. Br J Clin Pharmacol 1996; 41: 425–7.PubMed McMurrough J, McLeod HL. Analysis of the dihydropyrimidine dehydrogenase polymorphism in a British population. Br J Clin Pharmacol 1996; 41: 425–7.PubMed
58.
go back to reference Grem JL, Yee LK, Venzon DJ, et al. Inter- and intraindividual variation in dihydropyrimidine dehydrogenase activity in peripheral blood mononuclear cells. Cancer Chemother Pharmacol 1997; 40: 117–25.PubMed Grem JL, Yee LK, Venzon DJ, et al. Inter- and intraindividual variation in dihydropyrimidine dehydrogenase activity in peripheral blood mononuclear cells. Cancer Chemother Pharmacol 1997; 40: 117–25.PubMed
59.
go back to reference Van Kuilenburg AB, Van Lenthe H, Tromp A, et al. Pitfalls in the diagnosis of patients with a partial dihydropyrimidine dehydrogenase deficiency. Clin Chem 2000; 46: 9–17.PubMed Van Kuilenburg AB, Van Lenthe H, Tromp A, et al. Pitfalls in the diagnosis of patients with a partial dihydropyrimidine dehydrogenase deficiency. Clin Chem 2000; 46: 9–17.PubMed
60.
go back to reference Van Kuilenburg AB, van Lenthe H, Blom MJ, et al. Profound variation in dihydropyrimidine dehydrogenase activity in human blood cells: major implications for the detection of partly deficient patients. Br J Cancer 1999; 79: 620–6.PubMed Van Kuilenburg AB, van Lenthe H, Blom MJ, et al. Profound variation in dihydropyrimidine dehydrogenase activity in human blood cells: major implications for the detection of partly deficient patients. Br J Cancer 1999; 79: 620–6.PubMed
61.
go back to reference Lu Z, Zhang R, Carpenter JT, et al. Decreased dihydropyrimidine dehydrogenase activity in a population of patients with breast cancer: implication for 5-fluorouracil-based chemotherapy. Clin Cancer Res 1998; 4: 325–9.PubMed Lu Z, Zhang R, Carpenter JT, et al. Decreased dihydropyrimidine dehydrogenase activity in a population of patients with breast cancer: implication for 5-fluorouracil-based chemotherapy. Clin Cancer Res 1998; 4: 325–9.PubMed
62.
go back to reference Chazal M, Etienne MC, Renee N, et al. Link between dihydropyrimidine dehydrogenase activity in peripheral blood mononuclear cells and liver. Clin Cancer Res 1996; 2: 507–10.PubMed Chazal M, Etienne MC, Renee N, et al. Link between dihydropyrimidine dehydrogenase activity in peripheral blood mononuclear cells and liver. Clin Cancer Res 1996; 2: 507–10.PubMed
63.
go back to reference Stephan F, Etienne MC, Wallays C, et al. Depressed hepatic dihydropyrimidine dehydrogenase activity and fluorouracilrelated toxicities. Am JMed 1995; 99: 685–8. Stephan F, Etienne MC, Wallays C, et al. Depressed hepatic dihydropyrimidine dehydrogenase activity and fluorouracilrelated toxicities. Am JMed 1995; 99: 685–8.
64.
go back to reference Milano G, Etienne MC, Pierrefite V, et al. Dihydropyrimidine dehydrogenase deficiency and fluorouracil-related toxicity. Br J Cancer 1999; 79: 627–30.PubMed Milano G, Etienne MC, Pierrefite V, et al. Dihydropyrimidine dehydrogenase deficiency and fluorouracil-related toxicity. Br J Cancer 1999; 79: 627–30.PubMed
65.
go back to reference Katona C, Kralovanszky J, Rosta A, et al. Putative role of dihydropyrimidine dehydrogenase in the toxic side effect of 5-fluorouracil in colorectal cancer patients. Oncology 1998; 55: 468–74.PubMed Katona C, Kralovanszky J, Rosta A, et al. Putative role of dihydropyrimidine dehydrogenase in the toxic side effect of 5-fluorouracil in colorectal cancer patients. Oncology 1998; 55: 468–74.PubMed
66.
go back to reference Etienne MC, Cheradame S, Fischel JL, et al. Response to fluorouracil therapy in cancer patients: the role of tumoral dihydropyrimidine dehydrogenase activity. J Clin Oncol 1995; 13: 1663–70.PubMed Etienne MC, Cheradame S, Fischel JL, et al. Response to fluorouracil therapy in cancer patients: the role of tumoral dihydropyrimidine dehydrogenase activity. J Clin Oncol 1995; 13: 1663–70.PubMed
67.
go back to reference Salonga D, Danenberg KD, Johnson M, et al. Colorectal tumors responding to 5-fluorouracil have low gene expression levels of dihydropyrimidine dehydrogenase, thymidylate synthase, and thymidine phosphorylase. Clin Cancer Res 2000; 6: 1322–7.PubMed Salonga D, Danenberg KD, Johnson M, et al. Colorectal tumors responding to 5-fluorouracil have low gene expression levels of dihydropyrimidine dehydrogenase, thymidylate synthase, and thymidine phosphorylase. Clin Cancer Res 2000; 6: 1322–7.PubMed
68.
go back to reference Huang CL, Yokomise H, Kobayashi S, et al. Intratumoral expression of thymidylate synthase and dihydropyrimidine dehydrogenase in non-small cell lung cancer patients treated with 5-FU-based chemotherapy. Int J Oncol 2000; 17: 47–54.PubMed Huang CL, Yokomise H, Kobayashi S, et al. Intratumoral expression of thymidylate synthase and dihydropyrimidine dehydrogenase in non-small cell lung cancer patients treated with 5-FU-based chemotherapy. Int J Oncol 2000; 17: 47–54.PubMed
69.
go back to reference McLeod HL, Sludden J, Murray GI, et al. Characterization of dihydropyrimidine dehydrogenase in human colorectal tumours. Br J Cancer 1998; 77: 461–5.PubMed McLeod HL, Sludden J, Murray GI, et al. Characterization of dihydropyrimidine dehydrogenase in human colorectal tumours. Br J Cancer 1998; 77: 461–5.PubMed
70.
go back to reference Diasio RB, Beavers TL, Carpenter JT. Familial deficiency of dihydropyrimidine dehydrogenase: biochemical basis for familial pyrimidinemia and severe 5-fluorouracil-induced toxicity. J Clin Invest 1988; 81: 47–51.PubMed Diasio RB, Beavers TL, Carpenter JT. Familial deficiency of dihydropyrimidine dehydrogenase: biochemical basis for familial pyrimidinemia and severe 5-fluorouracil-induced toxicity. J Clin Invest 1988; 81: 47–51.PubMed
71.
go back to reference Harris BE, Carpenter JT, Diasio RB. Severe 5-fluorouracil toxicity secondary to dihydropyrimidine dehydrogenase deficiency: a potentially more common pharmacogenetic syndrome. Cancer 1991; 68: 499–501.PubMed Harris BE, Carpenter JT, Diasio RB. Severe 5-fluorouracil toxicity secondary to dihydropyrimidine dehydrogenase deficiency: a potentially more common pharmacogenetic syndrome. Cancer 1991; 68: 499–501.PubMed
72.
go back to reference Houyau P, Gay C, Chatelut E, et al. Severe fluorouracil toxicity in a patient with dihydropyrimidine dehydrogenase deficiency. J Natl Cancer Inst 1993; 85: 1602–3.PubMed Houyau P, Gay C, Chatelut E, et al. Severe fluorouracil toxicity in a patient with dihydropyrimidine dehydrogenase deficiency. J Natl Cancer Inst 1993; 85: 1602–3.PubMed
73.
go back to reference Lyss AP, Lilenbaum RC, Harris BE, et al. Severe 5-fluorouracil toxicity in a patient with decreased dihydropyrimidine dehydrogenase activity. Cancer Invest 1993; 11: 239–40.PubMed Lyss AP, Lilenbaum RC, Harris BE, et al. Severe 5-fluorouracil toxicity in a patient with decreased dihydropyrimidine dehydrogenase activity. Cancer Invest 1993; 11: 239–40.PubMed
74.
go back to reference Van Kuilenburg AB, Vreken P, Beex LV, et al. Heterozygosity for a point mutation in an invariant splice donor site of dihydropyrimidine dehydrogenase and severe 5-fluorouracil related toxicity. Eur J Cancer 1997; 33: 2258–64.PubMed Van Kuilenburg AB, Vreken P, Beex LV, et al. Heterozygosity for a point mutation in an invariant splice donor site of dihydropyrimidine dehydrogenase and severe 5-fluorouracil related toxicity. Eur J Cancer 1997; 33: 2258–64.PubMed
75.
go back to reference Takimoto CH, Lu ZH, Zhang R, et al. Severe neurotoxicity following 5-fluorouracil-based chemotherapy in a patient with dihydropyrimidine dehydrogenase deficiency. Clin Cancer Res 1996; 2: 477–81.PubMed Takimoto CH, Lu ZH, Zhang R, et al. Severe neurotoxicity following 5-fluorouracil-based chemotherapy in a patient with dihydropyrimidine dehydrogenase deficiency. Clin Cancer Res 1996; 2: 477–81.PubMed
76.
go back to reference Shehata N, Pater A, Tang SC. Prolonged severe 5-fluorouracil-associated neurotoxicity in a patient with dihydropyrimidine dehydrogenase deficiency. Cancer Invest 1999; 17: 201–5.PubMed Shehata N, Pater A, Tang SC. Prolonged severe 5-fluorouracil-associated neurotoxicity in a patient with dihydropyrimidine dehydrogenase deficiency. Cancer Invest 1999; 17: 201–5.PubMed
77.
go back to reference Lu Z, Zhang R, Diasio RB. Population characteristics of hepatic dihydropyrimidine dehydrogenase activity, a key metabolic enzyme in 5-fluorouracil chemotherapy. Clin Pharmacol Ther 1995; 58: 512–22.PubMed Lu Z, Zhang R, Diasio RB. Population characteristics of hepatic dihydropyrimidine dehydrogenase activity, a key metabolic enzyme in 5-fluorouracil chemotherapy. Clin Pharmacol Ther 1995; 58: 512–22.PubMed
78.
go back to reference Tuchman M, Stoeckeler JS, Kiang DT, et al. Familial pyrimidinemia and pyrimidinuria associated with severe fluorouracil toxicity. N Engl J Med 1985; 313: 245–9.PubMed Tuchman M, Stoeckeler JS, Kiang DT, et al. Familial pyrimidinemia and pyrimidinuria associated with severe fluorouracil toxicity. N Engl J Med 1985; 313: 245–9.PubMed
79.
go back to reference Gamelin E, Boisdron-Celle M, Guerin-Meyer V, et al. Correlation between uracil and dihydrouracil plasma ratio, fluorouracil (5-FU) pharmacokinetic parameters, and tolerance in patients with advanced colorectal cancer: a potential interest for predicting 5-FU toxicity and determining optimal 5-FU dosage. J Clin Oncol 1999; 17: 1105–10.PubMed Gamelin E, Boisdron-Celle M, Guerin-Meyer V, et al. Correlation between uracil and dihydrouracil plasma ratio, fluorouracil (5-FU) pharmacokinetic parameters, and tolerance in patients with advanced colorectal cancer: a potential interest for predicting 5-FU toxicity and determining optimal 5-FU dosage. J Clin Oncol 1999; 17: 1105–10.PubMed
80.
go back to reference Bocci G, Danesi R, Di Paolo A, et al. Comparative pharmacokinetic analysis of 5-fluorouracil and its major metabolite 5-fluoro-5,6-dihydrouracil after conventional and reduced test dose in cancer patients. Clin Cancer Res 2000; 6: 3032–7.PubMed Bocci G, Danesi R, Di Paolo A, et al. Comparative pharmacokinetic analysis of 5-fluorouracil and its major metabolite 5-fluoro-5,6-dihydrouracil after conventional and reduced test dose in cancer patients. Clin Cancer Res 2000; 6: 3032–7.PubMed
81.
go back to reference Humerickhouse R, Lohrbach K, Li L, et al. Characterization of CPT-11 hydrolysis by human liver carboxylesterase isoforms hCE-1 andhCE-2. Cancer Res 2000; 60: 1189–92.PubMed Humerickhouse R, Lohrbach K, Li L, et al. Characterization of CPT-11 hydrolysis by human liver carboxylesterase isoforms hCE-1 andhCE-2. Cancer Res 2000; 60: 1189–92.PubMed
82.
go back to reference Kawato Y, Furuta T, Aonuma M, et al. Antitumor activity of a camptothecin derivative, CPT-11, against human tumor xenografts in nude mice. Cancer Chemother Pharmacol 1991; 28: 192–8.PubMed Kawato Y, Furuta T, Aonuma M, et al. Antitumor activity of a camptothecin derivative, CPT-11, against human tumor xenografts in nude mice. Cancer Chemother Pharmacol 1991; 28: 192–8.PubMed
83.
go back to reference Rivory LP, Riou JF, Haaz MC, et al. Identification and properties of a major plasma metabolite of irinotecan (CPT-11) isolated from the plasma of patients. Cancer Res 1996; 56: 3689–94.PubMed Rivory LP, Riou JF, Haaz MC, et al. Identification and properties of a major plasma metabolite of irinotecan (CPT-11) isolated from the plasma of patients. Cancer Res 1996; 56: 3689–94.PubMed
84.
go back to reference Dodds HM, Haaz MC, Riou JF, et al. Identification of a new metabolite of CPT-11 (irinotecan): pharmacological properties and activation to SN-38. J Pharmacol Exp Ther 1998; 286: 578–83.PubMed Dodds HM, Haaz MC, Riou JF, et al. Identification of a new metabolite of CPT-11 (irinotecan): pharmacological properties and activation to SN-38. J Pharmacol Exp Ther 1998; 286: 578–83.PubMed
85.
go back to reference Gupta E, Lestingi TM, Mick R, et al. Metabolic fate of irinotecan inhumans: correlation of glucuronidation with diarrhea. Cancer Res 1994; 54: 3723–5.PubMed Gupta E, Lestingi TM, Mick R, et al. Metabolic fate of irinotecan inhumans: correlation of glucuronidation with diarrhea. Cancer Res 1994; 54: 3723–5.PubMed
86.
go back to reference Iyer L, King CD, Whitington PF, et al. 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; 101: 847–54.PubMed Iyer L, King CD, Whitington PF, et al. 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; 101: 847–54.PubMed
87.
go back to reference Sampietro M, Iolascon A. Molecular pathology of Crigler-Najjar type I and II and Gilbert’s syndromes. Haematologica 1999; 84: 150–7.PubMed Sampietro M, Iolascon A. Molecular pathology of Crigler-Najjar type I and II and Gilbert’s syndromes. Haematologica 1999; 84: 150–7.PubMed
88.
go back to reference Bosma PJ, Chowdhury JR, Bakker C, et al. The genetic basis of the reduced expression of bilirubin UDP-glucuronosyltransferase 1 in Gilbert’s syndrome. N Engl J Med 1995; 333: 1171–5.PubMed Bosma PJ, Chowdhury JR, Bakker C, et al. The genetic basis of the reduced expression of bilirubin UDP-glucuronosyltransferase 1 in Gilbert’s syndrome. N Engl J Med 1995; 333: 1171–5.PubMed
89.
go back to reference Monaghan G, Ryan M, Seddon R, et al. Genetic variation in bilirubin UDP-glucuronyltransferase gene promoter and Gilbert’s syndrome. Lancet 1996; 347: 578–81.PubMed Monaghan G, Ryan M, Seddon R, et al. Genetic variation in bilirubin UDP-glucuronyltransferase gene promoter and Gilbert’s syndrome. Lancet 1996; 347: 578–81.PubMed
90.
go back to reference Monaghan G, Foster B, Jurima-Romet M, et al. UGT1*1 genotyping in a Canadian Inuit population. Pharmacogenetics 1997; 7: 153–6.PubMed Monaghan G, Foster B, Jurima-Romet M, et al. UGT1*1 genotyping in a Canadian Inuit population. Pharmacogenetics 1997; 7: 153–6.PubMed
91.
go back to reference Owens D, Evans L. Population studies on Gilbert’s syndrome. J Med Genet 1975; 12: 152–6.PubMed Owens D, Evans L. Population studies on Gilbert’s syndrome. J Med Genet 1975; 12: 152–6.PubMed
92.
go back to reference Hall D, Ybazeta G, Destro-Bisol G, et al. Variability at the uridine diphosphate glucuronosyltransferase 1A1 promoter in human populations and primates. Pharmacogenetics 1999; 9: 591–9.PubMed Hall D, Ybazeta G, Destro-Bisol G, et al. Variability at the uridine diphosphate glucuronosyltransferase 1A1 promoter in human populations and primates. Pharmacogenetics 1999; 9: 591–9.PubMed
93.
go back to reference Ando Y, Chida M, Nakayama K, et al. The UGT1A1*28 allele is relatively rare in a Japanese population. Pharmacogenetics 1998; 8: 357–60.PubMed Ando Y, Chida M, Nakayama K, et al. The UGT1A1*28 allele is relatively rare in a Japanese population. Pharmacogenetics 1998; 8: 357–60.PubMed
94.
go back to reference 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.PubMed 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.PubMed
95.
go back to reference Lampe JW, Bigler J, Homer NK, et al. UDP-glucuronosyltransferase (UGT1A1*28 and UGT1A6*2) polymorphisms in Caucasians and Asians: relationships to serum bilirubin concentrations. Pharmacogenetics 1999; 9: 341–9.PubMed Lampe JW, Bigler J, Homer NK, et al. UDP-glucuronosyltransferase (UGT1A1*28 and UGT1A6*2) polymorphisms in Caucasians and Asians: relationships to serum bilirubin concentrations. Pharmacogenetics 1999; 9: 341–9.PubMed
96.
go back to reference DiRienzo A, Hall D, Iyer L, et al. Two new alleles in the promoter of the bilirubin UDP-glucuronosyl transferase 1 (UGT1A1) gene [abstract]. Clin Pharmacol Ther 1998; 63: 207. DiRienzo A, Hall D, Iyer L, et al. Two new alleles in the promoter of the bilirubin UDP-glucuronosyl transferase 1 (UGT1A1) gene [abstract]. Clin Pharmacol Ther 1998; 63: 207.
97.
go back to reference Iolascon A, Faienza MF, Centra M, et al. (TA)8 allele in the UGT1A1 gene promoter of a Caucasian with Gilbert’s syndrome. Haematologica 1999; 84: 106–9.PubMed Iolascon A, Faienza MF, Centra M, et al. (TA)8 allele in the UGT1A1 gene promoter of a Caucasian with Gilbert’s syndrome. Haematologica 1999; 84: 106–9.PubMed
98.
go back to reference Akaba K, Kimura T, Sasaki A, et al. Neonatal hyperbilirubinemia and a common mutation of the bilirubin uridine diphosphate-glucuronosyltransferase gene in Japanese. J Hum Genet 1999; 44: 22–5.PubMed Akaba K, Kimura T, Sasaki A, et al. Neonatal hyperbilirubinemia and a common mutation of the bilirubin uridine diphosphate-glucuronosyltransferase gene in Japanese. J Hum Genet 1999; 44: 22–5.PubMed
99.
go back to reference Akaba K, Kimura T, Sasaki A, et al. Neonatal hyperbilirubinemia and mutation of the bilirubin uridine diphosphate-glucuronosyltransferase gene: a common missense mutation among Japanese, Koreans and Chinese. Biochem Mol Biol Int 1998; 46: 21–6.PubMed Akaba K, Kimura T, Sasaki A, et al. Neonatal hyperbilirubinemia and mutation of the bilirubin uridine diphosphate-glucuronosyltransferase gene: a common missense mutation among Japanese, Koreans and Chinese. Biochem Mol Biol Int 1998; 46: 21–6.PubMed
100.
go back to reference Huang C-S, Luo G-A, Huang M-J, et al. Variations of the bilirubin uridine-diphosphoglucuronosyl transferase 1A1 gene in healthy Taiwanese. Pharmacogenetics 2000; 10: 539–44.PubMed Huang C-S, Luo G-A, Huang M-J, et al. Variations of the bilirubin uridine-diphosphoglucuronosyl transferase 1A1 gene in healthy Taiwanese. Pharmacogenetics 2000; 10: 539–44.PubMed
101.
go back to reference Koiwai O, Yasui Y, Hasada K, et al. Three Japanese patients with Crigler-Najjar syndrome type I carry an identical nonsense mutation in the gene for UDP-glucuronosyltransferase. Jpn J Hum Genet 1995; 40: 253–7.PubMed Koiwai O, Yasui Y, Hasada K, et al. Three Japanese patients with Crigler-Najjar syndrome type I carry an identical nonsense mutation in the gene for UDP-glucuronosyltransferase. Jpn J Hum Genet 1995; 40: 253–7.PubMed
102.
go back to reference Sato H, Adachi Y, Koiwai O. The genetic basis of Gilbert’s syndrome. Lancet 1996; 347: 557–8.PubMed Sato H, Adachi Y, Koiwai O. The genetic basis of Gilbert’s syndrome. Lancet 1996; 347: 557–8.PubMed
103.
go back to reference Iyer L, Hall D, Das S, et al. Phenotype-genotype correlation of in vitro SN-38 (active metabolite of irinotecan) and bilirubin glucuronidation in human liver tissue with UGT1A1 promoter polymorphism. Clin Pharmacol Ther 1999; 65: 576–82.PubMed Iyer L, Hall D, Das S, et al. Phenotype-genotype correlation of in vitro SN-38 (active metabolite of irinotecan) and bilirubin glucuronidation in human liver tissue with UGT1A1 promoter polymorphism. Clin Pharmacol Ther 1999; 65: 576–82.PubMed
104.
go back to reference Iyer L, Bergseth AC, Ybazeta G, et al. High variability in SN-38 glucuronidation in liver samples from African population as predicted by UGT1A1 genotypes [abstract]. Clin Pharmacol Ther 1999; 65: 197. Iyer L, Bergseth AC, Ybazeta G, et al. High variability in SN-38 glucuronidation in liver samples from African population as predicted by UGT1A1 genotypes [abstract]. Clin Pharmacol Ther 1999; 65: 197.
105.
go back to reference Ando Y, Saka H, Asai G, et al. UGT1A1 genotypes and glucuronidation of SN-38, the active metabolite of irinotecan. Ann Oncol 1998; 9: 845–7.PubMed Ando Y, Saka H, Asai G, et al. UGT1A1 genotypes and glucuronidation of SN-38, the active metabolite of irinotecan. Ann Oncol 1998; 9: 845–7.PubMed
106.
go back to reference Wasserman E, Myara A, Lokiec F, et al. Severe CPT-11 toxicity in patients with Gilbert’s syndrome: two case report. Ann Oncol 1997; 8: 1049–51.PubMed Wasserman E, Myara A, Lokiec F, et al. Severe CPT-11 toxicity in patients with Gilbert’s syndrome: two case report. Ann Oncol 1997; 8: 1049–51.PubMed
107.
go back to reference Iyer L, Janisch L, Das S, et al. UGT1A1 promoter genotype correlates with pharmacokinetics of irinotecan (CPT-11) [abstract]. Proc Am Soc Clin Oncol 2000; 19: 178a. Iyer L, Janisch L, Das S, et al. UGT1A1 promoter genotype correlates with pharmacokinetics of irinotecan (CPT-11) [abstract]. Proc Am Soc Clin Oncol 2000; 19: 178a.
108.
go back to reference De Isabella P, Zunino F, Capranico G. Base sequence determinants of amonafide stimulation of topoisomerase II DNA cleavage. Nucleic Acids Res 1995; 23: 223–9.PubMed De Isabella P, Zunino F, Capranico G. Base sequence determinants of amonafide stimulation of topoisomerase II DNA cleavage. Nucleic Acids Res 1995; 23: 223–9.PubMed
109.
go back to reference Costanza ME, Berry D, Henderson IC, et al. Amonafide: an active agent in the treatment of previously untreated advanced breast cancer — a cancer and leukemia group B study (CALGB 8642). Clin Cancer Res 1995; 1: 699–704.PubMed Costanza ME, Berry D, Henderson IC, et al. Amonafide: an active agent in the treatment of previously untreated advanced breast cancer — a cancer and leukemia group B study (CALGB 8642). Clin Cancer Res 1995; 1: 699–704.PubMed
110.
go back to reference Felder TB, McLean MA, Vestal ML, et al. Pharmacokinetics and metabolism of the antitumor drug amonafide (NSC-308847) in humans. Drug Metab Dispos 1987; 15: 773–7.PubMed Felder TB, McLean MA, Vestal ML, et al. Pharmacokinetics and metabolism of the antitumor drug amonafide (NSC-308847) in humans. Drug Metab Dispos 1987; 15: 773–7.PubMed
111.
go back to reference Price Evans DA, Manley KA, McKusick VA. Genetic control of isoniazid metabolism in man. BMJ 1960; 2: 485–91. Price Evans DA, Manley KA, McKusick VA. Genetic control of isoniazid metabolism in man. BMJ 1960; 2: 485–91.
112.
go back to reference Ratain MJ, Mick R, Berezin F, et al. Paradoxical relationship between acetylator phenotype and amonafide toxicity. Clin Pharmacol Ther 1991; 50: 573–9.PubMed Ratain MJ, Mick R, Berezin F, et al. Paradoxical relationship between acetylator phenotype and amonafide toxicity. Clin Pharmacol Ther 1991; 50: 573–9.PubMed
113.
go back to reference Ratain MJ, Mick R, Berezin F, et al. Phase I study of amonafide dosing based on acetylator phenotype. Cancer Res 1993; 53: 2304–8.PubMed Ratain MJ, Mick R, Berezin F, et al. Phase I study of amonafide dosing based on acetylator phenotype. Cancer Res 1993; 53: 2304–8.PubMed
114.
go back to reference Ratain MJ, Mick R, Janish L, et al. Individualized dosing of amonafide based on a pharmacodynamic model incorporating acetylator phenotype and gender. Pharmacogenetics 1996; 6: 93–101.PubMed Ratain MJ, Mick R, Janish L, et al. Individualized dosing of amonafide based on a pharmacodynamic model incorporating acetylator phenotype and gender. Pharmacogenetics 1996; 6: 93–101.PubMed
Metadata
Title
Pharmacogenetics
A Tool for Individualising Antineoplastic Therapy
Authors
Federico Innocenti
Lalitha Iyer
Dr Mark J. Ratain
Publication date
01-11-2000
Publisher
Springer International Publishing
Published in
Clinical Pharmacokinetics / Issue 5/2000
Print ISSN: 0312-5963
Electronic ISSN: 1179-1926
DOI
https://doi.org/10.2165/00003088-200039050-00001

Other articles of this Issue 5/2000

Clinical Pharmacokinetics 5/2000 Go to the issue