메뉴 건너뛰기




Volumn 54, Issue , 2003, Pages 437-452

Pharmacogenetics in Cancer Treatment

Author keywords

Chemotherapy; Drug metabolism; Drug response; Polymorphisms; Toxicity

Indexed keywords

5,10 METHYLENETETRAHYDROFOLATE REDUCTASE (FADH2); 6 MERCAPTOPURINE DERIVATIVE; 7 ETHYL 10 HYDROXYCAMPTOTHECIN; ACYLTRANSFERASE; ANTINEOPLASTIC AGENT; AZATHIOPRINE; CYTOCHROME P450 2D6; DIHYDROPYRIMIDINE DEHYDROGENASE; DOCETAXEL; FC RECEPTOR; FLUOROURACIL; GLUCOSE 6 PHOSPHATE DEHYDROGENASE; GLUCURONOSYLTRANSFERASE; GLUCURONOSYLTRANSFERASE 1A1; GLUTATHIONE TRANSFERASE; IMMUNOGLOBULIN G1; IRINOTECAN; MERCAPTOPURINE; METHOTREXATE; METHYLTRANSFERASE; MULTIDRUG RESISTANCE PROTEIN; OXALIPLATIN; RALTITREXED; RITUXIMAB; SULFOTRANSFERASE; THIOPURINE S METHYLTRANSFERASE; THYMIDYLATE SYNTHASE; TIOGUANINE; UNCLASSIFIED DRUG; UNINDEXED DRUG;

EID: 0038654356     PISSN: 00664219     EISSN: None     Source Type: Book Series    
DOI: 10.1146/annurev.med.54.101601.152352     Document Type: Review
Times cited : (103)

References (76)
  • 1
    • 0032748555 scopus 로고    scopus 로고
    • Pharmacogenetics and pharmacogenomics: Why is this relevant to the clinical geneticist?
    • Nebert DW. 1999. Pharmacogenetics and pharmacogenomics: Why is this relevant to the clinical geneticist? Clin. Genet. 56:247-58
    • (1999) Clin. Genet. , vol.56 , pp. 247-258
    • Nebert, D.W.1
  • 2
    • 0242565795 scopus 로고    scopus 로고
    • http://www.genome.gov/glossary
  • 3
    • 0035016127 scopus 로고    scopus 로고
    • Pharmacogenetics and pharmacogenomics
    • Leeder SJ. 2001. Pharmacogenetics and pharmacogenomics. Pediatr. Clin. North Am. 48(3):765-81
    • (2001) Pediatr. Clin. North Am. , vol.48 , Issue.3 , pp. 765-781
    • Leeder, S.J.1
  • 4
    • 0242423737 scopus 로고
    • Distribution of UDP-glucuronosyl transferases in rat tissue
    • Roy CJ, Novikoff PM, Roy CN, et al. 1985. Distribution of UDP-glucuronosyl transferases in rat tissue. Proc. Natl. Acad. Sci. USA 82(9):2990-94
    • (1985) Proc. Natl. Acad. Sci. USA , vol.82 , Issue.9 , pp. 2990-2994
    • Roy, C.J.1    Novikoff, P.M.2    Roy, C.N.3
  • 5
    • 0034778246 scopus 로고    scopus 로고
    • Human liver UDP-glucuronyltransferase isoforms involved in the glucuronidation of 7-ethyl-10-hydroxycamptothecin
    • Hanioka N, Ozawa S, Jinno H, et al. 2001. Human liver UDP-glucuronyltransferase isoforms involved in the glucuronidation of 7-ethyl-10-hydroxycamptothecin. Xenobiotica 31(10):687-99
    • (2001) Xenobiotica , vol.31 , Issue.10 , pp. 687-699
    • Hanioka, N.1    Ozawa, S.2    Jinno, H.3
  • 6
    • 8544224973 scopus 로고    scopus 로고
    • The UDP-glucuronyltransferase gene superfamily: Recommended nomenclature update based on evolutionary divergence
    • Mackenzie PI, Owens IS, Burchell B, et al. 1997. The UDP- glucuronyltransferase gene superfamily: recommended nomenclature update based on evolutionary divergence. Pharmacogenetics 7:255-69
    • (1997) Pharmacogenetics , vol.7 , pp. 255-269
    • Mackenzie, P.I.1    Owens, I.S.2    Burchell, B.3
  • 7
    • 0028276410 scopus 로고
    • Bilirubin UDP-glucuronyl transferase 1 is the only relevant bilirubin glucuronidating isoform in man
    • Bosma PJ, Seppen J, Goldhoorn B, et al. 1994. Bilirubin UDP-glucuronyl transferase 1 is the only relevant bilirubin glucuronidating isoform in man. J. Biol. Chem. 269:17960-64
    • (1994) J. Biol. Chem. , vol.269 , pp. 17960-17964
    • Bosma, P.J.1    Seppen, J.2    Goldhoorn, B.3
  • 8
    • 0033799997 scopus 로고    scopus 로고
    • Genetic lesions of bilirubin uridine-diphosphoglucuronate- glucuronosyltransferase (UGT1A1) causing Crigler-Najjar and Gilbert syndromes: Correlation of genotype to phenotype
    • Kadakol A, Ghosh SS, Sappal BS, et al. 2000. Genetic lesions of bilirubin uridine-diphosphoglucuronate-glucuronosyltransferase (UGT1A1) causing Crigler-Najjar and Gilbert syndromes: correlation of genotype to phenotype. Mutat. Update 16:297-306
    • (2000) Mutat. Update , vol.16 , pp. 297-306
    • Kadakol, A.1    Ghosh, S.S.2    Sappal, B.S.3
  • 9
    • 0028867826 scopus 로고
    • A sequence abnormality in the promoter region results in reduced expression of bilirubin-UDP-glucuronosyltransferase-1 in Gilbert's syndrome
    • Bosma PJ, Roy CJ, Bakker C, et al. 1995. A sequence abnormality in the promoter region results in reduced expression of bilirubin-UDP- glucuronosyltransferase-1 in Gilbert's syndrome. N. Engl. J. Med. 333:1171-75
    • (1995) N. Engl. J. Med. , vol.333 , pp. 1171-1175
    • Bosma, P.J.1    Roy, C.J.2    Bakker, C.3
  • 10
    • 0030030762 scopus 로고    scopus 로고
    • Genetic variation in bilirubin UDP-glucuronosyltransferase gene promoter and Gilbert's syndrome
    • Monaghan G, Ryan M, Seddon R, et al. 1996. Genetic variation in bilirubin UDP-glucuronosyltransferase gene promoter and Gilbert's syndrome. Lancet 347:578-81
    • (1996) Lancet , vol.347 , pp. 578-581
    • Monaghan, G.1    Ryan, M.2    Seddon, R.3
  • 11
  • 12
    • 0032493441 scopus 로고    scopus 로고
    • Racial variability in the UDP-glucuronosyltransferase 1 (UGT1A1) promoter: A balanced polymorphism for regulation of bilirubin metabolism?
    • Beutler E, Gelbart T, Demina A. 1998. Racial variability in the UDP-glucuronosyltransferase 1 (UGT1A1) promoter: a balanced polymorphism for regulation of bilirubin metabolism? Proc. Natl. Acad. Sci. USA 95:8170-74
    • (1998) Proc. Natl. Acad. Sci. USA , vol.95 , pp. 8170-8174
    • Beutler, E.1    Gelbart, T.2    Demina, A.3
  • 13
    • 0030053274 scopus 로고    scopus 로고
    • The genetic basis of Gilbert's syndrome
    • Sato H, Adachi Y, Koiwai O. 1996. The genetic basis of Gilbert's syndrome. Lancet 347:557-58
    • (1996) Lancet , vol.347 , pp. 557-558
    • Sato, H.1    Adachi, Y.2    Koiwai, O.3
  • 14
    • 0034162687 scopus 로고    scopus 로고
    • Characterization of CPT-11 hydrolysis by human liver carboxylesterase isoforms hCE-1 and hCE-2
    • Humerickhouse R, Lohrbach K, Li L, et al. 2000. Characterization of CPT-11 hydrolysis by human liver carboxylesterase isoforms hCE-1 and hCE-2. Cancer Res. 60:1189-92
    • (2000) Cancer Res. , vol.60 , pp. 1189-1192
    • Humerickhouse, R.1    Lohrbach, K.2    Li, L.3
  • 15
    • 0028022585 scopus 로고
    • Metabolic fate of irinotecan in humans: Correlation of glucuronidation with diarrhea
    • Gupta E, Lestingi TM, Mick R, et al. 1994. Metabolic fate of irinotecan in humans: correlation of glucuronidation with diarrhea. Cancer Res. 54:3723-25
    • (1994) Cancer Res. , vol.54 , pp. 3723-3725
    • Gupta, E.1    Lestingi, T.M.2    Mick, R.3
  • 16
    • 0032934383 scopus 로고    scopus 로고
    • Phenotype-genotype correlation of in vitro SN-38 (active metabolite of irinotecan) and bilirubin glucuronidation in human liver tissue with UGT1A1 promoter polymorphism
    • Iyer L, Hall D, Das S, et al. 1999. 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. 65(5):576-82
    • (1999) Clin. Pharmacol. Ther. , vol.65 , Issue.5 , pp. 576-582
    • Iyer, L.1    Hall, D.2    Das, S.3
  • 17
    • 0030716924 scopus 로고    scopus 로고
    • Severe CPT-11 toxicity in patients with Gilbert's syndrome: Two case reports
    • Wasserman E, Myara A, Lokiec F, et al. 1997. Severe CPT-11 toxicity in patients with Gilbert's syndrome: two case reports. Ann. Oncol. 9:1049-51
    • (1997) Ann. Oncol. , vol.9 , pp. 1049-1051
    • Wasserman, E.1    Myara, A.2    Lokiec, F.3
  • 18
    • 0034671387 scopus 로고    scopus 로고
    • Polymorphisms of UDP-glucuronosyltransferase gene and irinotecan toxicity: A pharmacogenetic analysis
    • Ando Y, Saka H, Ando M, et al. 2000. Polymorphisms of UDP-glucuronosyltransferase gene and irinotecan toxicity: a pharmacogenetic analysis. Cancer Res. 60:6921-26
    • (2000) Cancer Res. , vol.60 , pp. 6921-6926
    • Ando, Y.1    Saka, H.2    Ando, M.3
  • 19
    • 0036025450 scopus 로고    scopus 로고
    • UGT1A1*28 polymorphism as a determinant of irinotecan disposition and toxicity
    • Iyer L, Das S, Janish L, et al. 2002. UGT1A1*28 polymorphism as a determinant of irinotecan disposition and toxicity. Pharmacogenom. J. 2:43-47
    • (2002) Pharmacogenom. J. , vol.2 , pp. 43-47
    • Iyer, L.1    Das, S.2    Janish, L.3
  • 20
    • 0025161416 scopus 로고
    • Studies on the expression and metabolic capabilities of human liver cytochrome P450IIIA5
    • Wrighton SA, Brian WR, Sari MA, et al. 1990. Studies on the expression and metabolic capabilities of human liver cytochrome P450IIIA5. Mol. Pharmacol. 38:207-13
    • (1990) Mol. Pharmacol. , vol.38 , pp. 207-213
    • Wrighton, S.A.1    Brian, W.R.2    Sari, M.A.3
  • 21
    • 0036089341 scopus 로고    scopus 로고
    • Co-regulation of CYP3A4 and CYP3A5 and contribution to hepatic and intestinal midazolam metabolism
    • Yvonne SL, Amy LS, Dowling SD, et al. 2002. Co-regulation of CYP3A4 and CYP3A5 and contribution to hepatic and intestinal midazolam metabolism. Mol. Pharmacol. 62:162-72
    • (2002) Mol. Pharmacol. , vol.62 , pp. 162-172
    • Yvonne, S.L.1    Amy, L.S.2    Dowling, S.D.3
  • 22
    • 0034791777 scopus 로고    scopus 로고
    • Polymorphisms in human CYP2C8 decrease metabolism of the anticancer drug paclitaxel and arachidonic acid
    • Dai D, Zeldin DC, Blaisdell JA, et al. 2001. Polymorphisms in human CYP2C8 decrease metabolism of the anticancer drug paclitaxel and arachidonic acid. Pharmacogenetics 11(7):597-607
    • (2001) Pharmacogenetics , vol.11 , Issue.7 , pp. 597-607
    • Dai, D.1    Zeldin, D.C.2    Blaisdell, J.A.3
  • 23
    • 0018822866 scopus 로고
    • Mercaptopurine pharmacogenetics: Monogenic inheritance of erythrocyte thiopurine methyltransferase activity
    • Weinshilboum RM, Sladek S. 1980. Mercaptopurine pharmacogenetics: monogenic inheritance of erythrocyte thiopurine methyltransferase activity. Am. J. Hum. Genet. 32:651-52
    • (1980) Am. J. Hum. Genet. , vol.32 , pp. 651-652
    • Weinshilboum, R.M.1    Sladek, S.2
  • 24
    • 0030048791 scopus 로고    scopus 로고
    • Thiopurine methyltransferase pharmacogenetics: Human gene cloning and characterization of a common polymorphism
    • Szumlanski C, Otterness D, Her C, et al. 1996. Thiopurine methyltransferase pharmacogenetics: human gene cloning and characterization of a common polymorphism. DNA Cell. Biol. 15:17-30
    • (1996) DNA Cell. Biol. , vol.15 , pp. 17-30
    • Szumlanski, C.1    Otterness, D.2    Her, C.3
  • 25
    • 0030986251 scopus 로고    scopus 로고
    • Enhanced proteolysis of thiopurine S-methyltransferase (TPMT) encoded by mutant alleles in humans (TPMT*3A, TPMT*20): Mechanisms for the genetic polymorphism of TPMT activity
    • Tai HL, Krynetski EY, Schuetz EG, et al. 1997. Enhanced proteolysis of thiopurine S-methyltransferase (TPMT) encoded by mutant alleles in humans (TPMT*3A, TPMT*20): mechanisms for the genetic polymorphism of TPMT activity. Proc. Natl. Acad. Sci. USA 94:6444-49
    • (1997) Proc. Natl. Acad. Sci. USA , vol.94 , pp. 6444-6449
    • Tai, H.L.1    Krynetski, E.Y.2    Schuetz, E.G.3
  • 26
    • 0033837392 scopus 로고    scopus 로고
    • Thiopurine methyltransferase (TPMT) promoter variable number tandem repeat (VNTR) polymorphism: Genotype-phenotype correlation analysis for 1211 patients
    • Yan L, Zhang S, Eiff B, et al. 2000. Thiopurine methyltransferase (TPMT) promoter variable number tandem repeat (VNTR) polymorphism: genotype-phenotype correlation analysis for 1211 patients. Clin. Pharmacol. Ther. 68:210-19
    • (2000) Clin. Pharmacol. Ther. , vol.68 , pp. 210-219
    • Yan, L.1    Zhang, S.2    Eiff, B.3
  • 27
    • 0025837127 scopus 로고
    • Altered mercaptopurine metabolism, toxic effects and dosage requirement in a thiopurine methyltransferase deficient child with acute lymphoblastic leukemia
    • Evans WE, Horner M, Chu YQ, et al. 1991. Altered mercaptopurine metabolism, toxic effects and dosage requirement in a thiopurine methyltransferase deficient child with acute lymphoblastic leukemia. J. Pediatr. 119:985-89
    • (1991) J. Pediatr. , vol.119 , pp. 985-989
    • Evans, W.E.1    Horner, M.2    Chu, Y.Q.3
  • 28
    • 0027401302 scopus 로고
    • Azathiopurine-induced myelosuppression in thiopurine methyltransferase deficient heart transplant recipients
    • Schutz E, Gummert J, Mohr F, et al. 1993. Azathiopurine-induced myelosuppression in thiopurine methyltransferase deficient heart transplant recipients. Lancet 341:436
    • (1993) Lancet , vol.341 , pp. 436
    • Schutz, E.1    Gummert, J.2    Mohr, F.3
  • 29
    • 0030656430 scopus 로고    scopus 로고
    • Thiopurine drugs in the treatment of childhood leukemia: The influence of inherited thiopurine methyltransferase activity on drug metabolism and cytotoxicity
    • Lennard L, Welch JC, Lilleyman JS, et al. 1997. Thiopurine drugs in the treatment of childhood leukemia: the influence of inherited thiopurine methyltransferase activity on drug metabolism and cytotoxicity. Br. J. Clin. Pharmacol. 44:455-61
    • (1997) Br. J. Clin. Pharmacol. , vol.44 , pp. 455-461
    • Lennard, L.1    Welch, J.C.2    Lilleyman, J.S.3
  • 30
    • 0035871560 scopus 로고    scopus 로고
    • Preponderance of thiopurine S-methyltransferase deficiency and heterozygosity among patients intolerant to mercaptopurine or azathiopurine
    • Evans WE, Hon YY, Bomgaars L, et al. 2001. Preponderance of thiopurine S-methyltransferase deficiency and heterozygosity among patients intolerant to mercaptopurine or azathiopurine. J. Clin. Oncol. 19(8):2293-301
    • (2001) J. Clin. Oncol. , vol.19 , Issue.8 , pp. 2293-2301
    • Evans, W.E.1    Hon, Y.Y.2    Bomgaars, L.3
  • 31
    • 0033486029 scopus 로고    scopus 로고
    • Mercaptopurine therapy intolerance and heterozygosity at the thiopurine methyltransferase gene locus
    • Relling MV, Hancock ML, Rivera GK, et al. 1999. Mercaptopurine therapy intolerance and heterozygosity at the thiopurine methyltransferase gene locus. J. Natl. Cancer. Inst. 91(23):2001-8
    • (1999) J. Natl. Cancer. Inst. , vol.91 , Issue.23 , pp. 2001-2008
    • Relling, M.V.1    Hancock, M.L.2    Rivera, G.K.3
  • 32
    • 0033135738 scopus 로고    scopus 로고
    • Prognostic importance of 6-mercaptopurine dose intensity in acute lymphoblastic leukemia
    • Relling MV, Hancock ML, Boyett JM, et al. 1999. Prognostic importance of 6-mercaptopurine dose intensity in acute lymphoblastic leukemia. Blood 93(9): 2817-23
    • (1999) Blood , vol.93 , Issue.9 , pp. 2817-2823
    • Relling, M.V.1    Hancock, M.L.2    Boyett, J.M.3
  • 33
    • 0025331198 scopus 로고
    • Genetic variation in response to 6-mercaptopurine for childhood acute lymphoblastic leukemia
    • Lennard L, Lilleyman JS, Van Loon J, et al. 1990. Genetic variation in response to 6-mercaptopurine for childhood acute lymphoblastic leukemia. Lancet 336:225-29
    • (1990) Lancet , vol.336 , pp. 225-229
    • Lennard, L.1    Lilleyman, J.S.2    Van Loon, J.3
  • 34
    • 0035033019 scopus 로고    scopus 로고
    • Pharmacogenetics: Unlocking the human genome for better drug therapy
    • 33a. McLeod HL, Evans WE. 2001. Pharmacogenetics: unlocking the human genome for better drug therapy. Annu. Rev. Pharmacol. Toxicol. 41:101-21
    • (2001) Annu. Rev. Pharmacol. Toxicol. , vol.41 , pp. 101-121
    • McLeod, H.L.1    Evans, W.E.2
  • 35
    • 0018894446 scopus 로고
    • Degradation of pyrimidines and pyrimidine analogs - Pathways and mutual influences
    • Wasternack C. 1980. Degradation of pyrimidines and pyrimidine analogs - pathways and mutual influences. Pharmacol. Ther. 8:629-52
    • (1980) Pharmacol. Ther. , vol.8 , pp. 629-652
    • Wasternack, C.1
  • 36
    • 0023187085 scopus 로고
    • Clinical pharmacokinetics of 5-fluorouracil and its metabolism in plasma, urine and bile
    • Heggie GD, Sommadossi J-P, Cross DS, et al. 1987. Clinical pharmacokinetics of 5-fluorouracil and its metabolism in plasma, urine and bile. Cancer Res. 47: 2203-306
    • (1987) Cancer Res. , vol.47 , pp. 2203-2306
    • Heggie, G.D.1    Sommadossi, J.-P.2    Cross, D.S.3
  • 37
    • 0033975358 scopus 로고    scopus 로고
    • Phase I and pharmacologic study of oral fluorouracil on a chronic daily schedule in combination with the dihydropyrimidine dehydrogenase inactivator eniluracil
    • Baker SD, Diasio RB, O'Reilly S, et al. 2000. Phase I and pharmacologic study of oral fluorouracil on a chronic daily schedule in combination with the dihydropyrimidine dehydrogenase inactivator eniluracil. J.Clin.Oncol. 4:915-26
    • (2000) J. Clin. Oncol. , vol.4 , pp. 915-926
    • Baker, S.D.1    Diasio, R.B.2    O'Reilly, S.3
  • 38
    • 0022263834 scopus 로고
    • Familial pyrimidinemia and pyrimidinuria associated with severe fluorouracil toxicity
    • Tuchman M, Stoeckeler JS, Kiang DT, et al. 1985. Familial pyrimidinemia and pyrimidinuria associated with severe fluorouracil toxicity. N. Engl. J. Med. 313 (4):245-49
    • (1985) N. Engl. J. Med. , vol.313 , Issue.4 , pp. 245-249
    • Tuchman, M.1    Stoeckeler, J.S.2    Kiang, D.T.3
  • 39
    • 0025990479 scopus 로고
    • Severe 5-fluorouracil toxicity secondary to dihydropyrimidine dehydrogenase deficiency: A potentially more common pharmacogenetic syndrome
    • Harris BE, Carpenter JT, Diaso RB. 1991. Severe 5-fluorouracil toxicity secondary to dihydropyrimidine dehydrogenase deficiency: a potentially more common pharmacogenetic syndrome. Cancer 69:499-501
    • (1991) Cancer , vol.69 , pp. 499-501
    • Harris, B.E.1    Carpenter, J.T.2    Diaso, R.B.3
  • 40
    • 0029973215 scopus 로고    scopus 로고
    • Molecular basis of the human dihydropyrimidine dehydrogenase deficiency and 5-fluorouracil toxicity
    • Wei X, McLeod HD, McMurrough J, et al. 1996. Molecular basis of the human dihydropyrimidine dehydrogenase deficiency and 5-fluorouracil toxicity. J. Clin. Invest. 98:610-15
    • (1996) J. Clin. Invest. , vol.98 , pp. 610-615
    • Wei, X.1    McLeod, H.D.2    McMurrough, J.3
  • 41
    • 0034984031 scopus 로고    scopus 로고
    • Importance of dihydrpyrimidine dehydrogenase (DPD) deficiency in patients exhibiting toxicity following treatment with 5-fluorouracil
    • Johnson MR, Diasio RB. 2001. Importance of dihydrpyrimidine dehydrogenase (DPD) deficiency in patients exhibiting toxicity following treatment with 5-fluorouracil. Adv. Enzyme. Regul. 41: 151-57
    • (2001) Adv. Enzyme. Regul. , vol.41 , pp. 151-157
    • Johnson, M.R.1    Diasio, R.B.2
  • 42
    • 0028577327 scopus 로고
    • Dihydropyrimidine dehydrogenase (DPD) and clinical pharmacology of 5-fluorouracil
    • Milano G, Etienne MC. 1994. Dihydropyrimidine dehydrogenase (DPD) and clinical pharmacology of 5-fluorouracil. Anticancer Res. 14:2295-97
    • (1994) Anticancer Res. , vol.14 , pp. 2295-2297
    • Milano, G.1    Etienne, M.C.2
  • 43
    • 0000163968 scopus 로고    scopus 로고
    • Dihydropyrimidine dehydrogenase deficiency and fluorouracil-related toxicity
    • Milano G, Etienne MC, Pierrefite V, et al. 1999. Dihydropyrimidine dehydrogenase deficiency and fluorouracil-related toxicity. Br. J. Cancer 79(3-4):627-30
    • (1999) Br. J. Cancer , vol.79 , Issue.3-4 , pp. 627-630
    • Milano, G.1    Etienne, M.C.2    Pierrefite, V.3
  • 44
    • 0034488052 scopus 로고    scopus 로고
    • Clinical implications of dihydropyrimidine dehydrogenase (DPD) deficiency in patients with severe 5-fluorouracil-asociated toxicity: Identification of new mutations in the DPD gene
    • van Kuilenberg AB, Haasjes J, Richel DJ, et al. 2000. Clinical implications of dihydropyrimidine dehydrogenase (DPD) deficiency in patients with severe 5-fluorouracil-asociated toxicity: identification of new mutations in the DPD gene. Clin. Cancer Res. 6:4705-12
    • (2000) Clin. Cancer Res. , vol.6 , pp. 4705-4712
    • Kuilenberg, A.B.1    Haasjes, J.2    Richel, D.J.3
  • 45
    • 0032143631 scopus 로고    scopus 로고
    • Characterization of the human dihydropyrimidine dehydrogenase gene
    • Wei X, Elizondo G, Sapone A, et al. 1996. Characterization of the human dihydropyrimidine dehydrogenase gene. Genomics 51:391-400
    • (1996) Genomics , vol.51 , pp. 391-400
    • Wei, X.1    Elizondo, G.2    Sapone, A.3
  • 46
    • 0034901306 scopus 로고    scopus 로고
    • Lethal outcome of a patient with a complete dihydropyrimidine dehydrogenase (DPD) deficiency after administration of 5-fluorouracil. Frequency of the common IVS14 + IG > a mutation causing DPD deficiency
    • van Kuilenberg AB, Muller EW, Haasjes J, et al. 2001. Lethal outcome of a patient with a complete dihydropyrimidine dehydrogenase (DPD) deficiency after administration of 5-fluorouracil. Frequency of the common IVS14 + IG > A mutation causing DPD deficiency. Clin. Cancer Res. 7(5):1149-53
    • (2001) Clin. Cancer Res. , vol.7 , Issue.5 , pp. 1149-1153
    • Kuilenberg, A.B.1    Muller, E.W.2    Haasjes, J.3
  • 47
    • 0034808681 scopus 로고    scopus 로고
    • Prevalence of a common point mutation in the dihydropyrimidine dehydrogenase (DPD) gene within the 5'-splice donor site of intron 14 in patients with severe 5-fluorouracil (5-FU)-related toxicity compared to controls
    • Raida M, Schwabe W, Hausler P, et al. 2001. Prevalence of a common point mutation in the dihydropyrimidine dehydrogenase (DPD) gene within the 5'-splice donor site of intron 14 in patients with severe 5-fluorouracil (5-FU)-related toxicity compared to controls. Clin. Cancer Res. 7:2832-39
    • (2001) Clin. Cancer Res. , vol.7 , pp. 2832-2839
    • Raida, M.1    Schwabe, W.2    Hausler, P.3
  • 48
    • 0034050905 scopus 로고    scopus 로고
    • Known variant DPYD alleles do not explain DPD deficiency in cancer patients
    • Collie-Duguid ES, Etienne MC, Milano G, et al. 2000. Known variant DPYD alleles do not explain DPD deficiency in cancer patients. Pharmacogenetics 10(30):217-23
    • (2000) Pharmacogenetics , vol.10 , Issue.30 , pp. 217-223
    • Collie-Duguid, E.S.1    Etienne, M.C.2    Milano, G.3
  • 49
    • 0242565958 scopus 로고    scopus 로고
    • Deleted in proof
    • Deleted in proof
  • 50
    • 0031014682 scopus 로고    scopus 로고
    • Thymidylate synthase inhibitors in cancer therapy: Direct and indirect inhibitors
    • Rutsum YM, Harstrick A, Cao S, et al. 1997. Thymidylate synthase inhibitors in cancer therapy: direct and indirect inhibitors. J. Clin. Oncol. 15:389-400
    • (1997) J. Clin. Oncol. , vol.15 , pp. 389-400
    • Rutsum, Y.M.1    Harstrick, A.2    Cao, S.3
  • 51
    • 19244381642 scopus 로고    scopus 로고
    • Thymidylate synthase levels as a therapeutic and prognostic predictor in breast cancer
    • Nishimura R, Nagao K, Miyayama H, et al. 1999. Thymidylate synthase levels as a therapeutic and prognostic predictor in breast cancer. Anticancer Res. 19:5621-26
    • (1999) Anticancer Res. , vol.19 , pp. 5621-5626
    • Nishimura, R.1    Nagao, K.2    Miyayama, H.3
  • 52
    • 0030883708 scopus 로고    scopus 로고
    • Quantitation of intratumoral thymidylate synthase expression predicts for disseminated colorectal cancer response and resistance to protracted-infusion fluorouracil and weekly leucovorin
    • Leichman CG, Lenz HJ, Leichman L, et al. 1997. Quantitation of intratumoral thymidylate synthase expression predicts for disseminated colorectal cancer response and resistance to protracted-infusion fluorouracil and weekly leucovorin. J. Clin. Oncol. 15:3223-29
    • (1997) J. Clin. Oncol. , vol.15 , pp. 3223-3229
    • Leichman, C.G.1    Lenz, H.J.2    Leichman, L.3
  • 53
    • 0032994840 scopus 로고    scopus 로고
    • Thymidylate synthase level as the main predictive parameter for sensitivity to 5-fluorouracil, but not for folate-based thymidylate synthase inhibitors, in 13 nonselected colon cancer cell lines
    • van Triest B, Pinedo HM, van Hensbergen Y, et al. 1999. Thymidylate synthase level as the main predictive parameter for sensitivity to 5-fluorouracil, but not for folate-based thymidylate synthase inhibitors, in 13 nonselected colon cancer cell lines. Clin. Cancer Res. 5:643-54
    • (1999) Clin. Cancer Res. , vol.5 , pp. 643-654
    • Van Triest, B.1    Pinedo, H.M.2    Van Hensbergen, Y.3
  • 54
    • 0033427561 scopus 로고    scopus 로고
    • Polymorphic tandem repeats in the thymidylate synthase gene is associated with its protein expression in human gastrointestinal cancers
    • Kawakami K, Omura K, Kanehira E, et al. 1999. Polymorphic tandem repeats in the thymidylate synthase gene is associated with its protein expression in human gastrointestinal cancers. Anticancer Res. 19(4B):3249-52
    • (1999) Anticancer Res. , vol.19 , Issue.4 B , pp. 3249-3252
    • Kawakami, K.1    Omura, K.2    Kanehira, E.3
  • 55
    • 0033564283 scopus 로고    scopus 로고
    • Ethnic variation in the thymidylate synthase enhancer region polymorphism among Caucasian and Asian populations
    • Marsh S, Collie-Duguid ES, Li T, et al. 1999. Ethnic variation in the thymidylate synthase enhancer region polymorphism among Caucasian and Asian populations. Genomics 58:310-12
    • (1999) Genomics , vol.58 , pp. 310-312
    • Marsh, S.1    Collie-Duguid, E.S.2    Li, T.3
  • 56
    • 0035430487 scopus 로고    scopus 로고
    • Polymorphism in the thymidylate synthase promoter enhancer region in colorectal cancer
    • Marsh S, McKay JA, Cassidy J, et al. 2001. Polymorphism in the thymidylate synthase promoter enhancer region in colorectal cancer. Int. J. Clin. Oncol. 19:383-86
    • (2001) Int. J. Clin. Oncol. , vol.19 , pp. 383-386
    • Marsh, S.1    McKay, J.A.2    Cassidy, J.3
  • 57
    • 0035750549 scopus 로고    scopus 로고
    • Thymidylate synthase gene polymorphism determines response and toxicity of 5-FU chemotherapy
    • Pullarkat ST, Stoehlmacher J, Ghaderi V, et al. 2001. Thymidylate synthase gene polymorphism determines response and toxicity of 5-FU chemotherapy. Pharmacogenom. J. 1:65-70
    • (2001) Pharmacogenom. J. , vol.1 , pp. 65-70
    • Pullarkat, S.T.1    Stoehlmacher, J.2    Ghaderi, V.3
  • 58
    • 0035860142 scopus 로고    scopus 로고
    • A polymorphism in the enhancer region of the thymidylate synthase gene influences the survival of colorectal cancer patients treated with 5-fluorouracil
    • Iacopetta B, Grieu F, Joseph D, et al. 2001. A polymorphism in the enhancer region of the thymidylate synthase gene influences the survival of colorectal cancer patients treated with 5-fluorouracil. Br. J. Cancer 85:827-30
    • (2001) Br. J. Cancer , vol.85 , pp. 827-830
    • Iacopetta, B.1    Grieu, F.2    Joseph, D.3
  • 59
    • 0037161062 scopus 로고    scopus 로고
    • Polymorphism of the thymidylate synthase gene and outcome of acute lymphoblastic leukemia
    • Krajinovic M, Costea I, Chiasson S. 2002. Polymorphism of the thymidylate synthase gene and outcome of acute lymphoblastic leukemia. Lancet 359:1033-34
    • (2002) Lancet , vol.359 , pp. 1033-1034
    • Krajinovic, M.1    Costea, I.2    Chiasson, S.3
  • 60
    • 0028057250 scopus 로고
    • Depletion of B-cells in vivo by a chimeric mouse human monoclonal antibody to CD20
    • Reff ME, Carner K, Chambers KS, et al. 1994. Depletion of B-cells in vivo by a chimeric mouse human monoclonal antibody to CD20. Blood 83:435-45
    • (1994) Blood , vol.83 , pp. 435-445
    • Reff, M.E.1    Carner, K.2    Chambers, K.S.3
  • 61
    • 0032030699 scopus 로고    scopus 로고
    • Apoptosis of malignant human B cells by ligation of CD20 with monoclonal antibodies
    • Shan D, Ledbetter JA, Press OW. 1998. Apoptosis of malignant human B cells by ligation of CD20 with monoclonal antibodies. Blood 91:1644-52
    • (1998) Blood , vol.91 , pp. 1644-1652
    • Shan, D.1    Ledbetter, J.A.2    Press, O.W.3
  • 62
    • 0036464719 scopus 로고    scopus 로고
    • Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor FcγRIIIa gene
    • Cartron G, Dacheux L, Salles G, et al. 2002. Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor FcγRIIIa gene. Blood 99:754-58
    • (2002) Blood , vol.99 , pp. 754-758
    • Cartron, G.1    Dacheux, L.2    Salles, G.3
  • 63
    • 0030611643 scopus 로고    scopus 로고
    • FcγRIIIa-158V/F polymorphism influences the binding of IgG by natural killer cell FcγRIIIa, independently of the FcγRIIIa-48L/R/H phenotype
    • Koene HR, Kleijer M, Algra J, et al. 1997. FcγRIIIa-158V/F polymorphism influences the binding of IgG by natural killer cell FcγRIIIa, independently of the FcγRIIIa-48L/R/H phenotype. Blood 90:1109-14
    • (1997) Blood , vol.90 , pp. 1109-1114
    • Koene, H.R.1    Kleijer, M.2    Algra, J.3
  • 64
    • 0029049553 scopus 로고
    • A candidate genetic risk factor for vascular disease; a common mutation in methylenetetrahydrofolate reductase
    • Frosst P, Blom HJ, Milos R, et al. 1995. A candidate genetic risk factor for vascular disease; a common mutation in methylenetetrahydrofolate reductase. Nat. Genet. 10:111-13
    • (1995) Nat. Genet. , vol.10 , pp. 111-113
    • Frosst, P.1    Blom, H.J.2    Milos, R.3
  • 65
    • 0031961002 scopus 로고    scopus 로고
    • Worldwide distribution of a common methylenetetrahydrofolate reductase mutation
    • Schneider JA, Rees DC, Liu YT, et al. 1998. Worldwide distribution of a common methylenetetrahydrofolate reductase mutation. Am. J. Hum. Genet. 62:1258-60
    • (1998) Am. J. Hum. Genet. , vol.62 , pp. 1258-1260
    • Schneider, J.A.1    Rees, D.C.2    Liu, Y.T.3
  • 66
    • 0035412398 scopus 로고    scopus 로고
    • Pharmacogenetics of methotrexate: Toxicity among marrow transplantation patients varies with the methylenetetrahydrofolate reductase C677T polymorphism
    • Ulrich CM, Yasui Y, Storb R, et al. 2001. Pharmacogenetics of methotrexate: toxicity among marrow transplantation patients varies with the methylenetetrahydrofolate reductase C677T polymorphism. Blood 98:231-34
    • (2001) Blood , vol.98 , pp. 231-234
    • Ulrich, C.M.1    Yasui, Y.2    Storb, R.3
  • 67
    • 0033998678 scopus 로고    scopus 로고
    • MTHFR gene polymorphism and severe toxicity during adjuvant treatment of early breast cancer with cyclophosphamide, methotrexate and fluorouracil (CMF)
    • Toffoli G, Veronesi A, Boiocchi M, et al. 2000. MTHFR gene polymorphism and severe toxicity during adjuvant treatment of early breast cancer with cyclophosphamide, methotrexate and fluorouracil (CMF). Ann. Oncol. 11:373-75
    • (2000) Ann. Oncol. , vol.11 , pp. 373-375
    • Toffoli, G.1    Veronesi, A.2    Boiocchi, M.3
  • 68
    • 0035886707 scopus 로고    scopus 로고
    • Phase I clinical and pharmacogenetic trial of irinotecan and raltitrexed administered every 21 days to patients with cancer
    • Stevenson JP, Redlinger M, Kluijtmans LAJ, et al. 2001. Phase I clinical and pharmacogenetic trial of irinotecan and raltitrexed administered every 21 days to patients with cancer. J. Clin. Oncol. 19(20):4081-87
    • (2001) J. Clin. Oncol. , vol.19 , Issue.20 , pp. 4081-4087
    • Stevenson, J.P.1    Redlinger, M.2    Kluijtmans, L.A.J.3
  • 69
    • 0031791029 scopus 로고    scopus 로고
    • Platinum-DNA adduct, nucleotide excision repair and platinum based anti-cancer chemotherapy
    • Reed E. 1998. Platinum-DNA adduct, nucleotide excision repair and platinum based anti-cancer chemotherapy. Cancer Treat. Rev. 24(5):331-44
    • (1998) Cancer Treat. Rev. , vol.24 , Issue.5 , pp. 331-344
    • Reed, E.1
  • 70
    • 0035893755 scopus 로고    scopus 로고
    • A xeroderma pigmentosum group D gene polymorphism predicts clinical outcome to platinum-based chemotherapy in patients with advanced colorectal cancer
    • Park DJ, Stoehlmacher J, Zhang W, et al. 2001. A xeroderma pigmentosum group D gene polymorphism predicts clinical outcome to platinum-based chemotherapy in patients with advanced colorectal cancer. Cancer Res. 61(24):8654-58
    • (2001) Cancer Res. , vol.61 , Issue.24 , pp. 8654-8658
    • Park, D.J.1    Stoehlmacher, J.2    Zhang, W.3
  • 71
    • 0033955346 scopus 로고    scopus 로고
    • XRCC1 keeps DNA from getting stranded
    • Thompson LH, West MG. 2002. XRCC1 keeps DNA from getting stranded. Mutat. Res. 459:1-18
    • (2002) Mutat. Res. , vol.459 , pp. 1-18
    • Thompson, L.H.1    West, M.G.2
  • 72
    • 0035174375 scopus 로고    scopus 로고
    • A polymorphism of the XRCC1 gene predicts for response to platinum based treatment in advanced colorectal cancer
    • Stoehlmacher J, Ghaderi V, Iobal S, et al. 2001. A polymorphism of the XRCC1 gene predicts for response to platinum based treatment in advanced colorectal cancer. Anticancer Res. 21(4B):3075-79
    • (2001) Anticancer Res. , vol.21 , Issue.4 B , pp. 3075-3079
    • Stoehlmacher, J.1    Ghaderi, V.2    Iobal, S.3
  • 73
    • 0033010781 scopus 로고    scopus 로고
    • Werner syndrome lymphoblastoid cells are sensitive to camptothecin-induced apoptosis in S-phase
    • Poot M, Gollahon KA, Rabinovitch PS. 1999. Werner syndrome lymphoblastoid cells are sensitive to camptothecin-induced apoptosis in S-phase. Hum. Genet. 104(1):10-14
    • (1999) Hum. Genet. , vol.104 , Issue.1 , pp. 10-14
    • Poot, M.1    Gollahon, K.A.2    Rabinovitch, P.S.3
  • 74
    • 0033582750 scopus 로고    scopus 로고
    • Polymorphisms at the Werner locus. I. Newly identified polymorphisms, ethnic variability of 1367Cys/Arg, and its stability in a population of Finnish centenarians
    • Castro E, Ogburn CE, Hunt KE, et al. 1999. Polymorphisms at the Werner locus. I. Newly identified polymorphisms, ethnic variability of 1367Cys/Arg, and its stability in a population of Finnish centenarians. Am. J. Med. Genet. 82(5):399-403
    • (1999) Am. J. Med. Genet. , vol.82 , Issue.5 , pp. 399-403
    • Castro, E.1    Ogburn, C.E.2    Hunt, K.E.3
  • 75
    • 0036016317 scopus 로고    scopus 로고
    • A naturally occurring mutation in MRP1 results in a selective decrease in organic anion transport and in increased doxorubicin resistance
    • Conrad S, Kauffmann HM, Ito K, et al. 2002. A naturally occurring mutation in MRP1 results in a selective decrease in organic anion transport and in increased doxorubicin resistance. Pharmacogenetics 12(4):321-30
    • (2002) Pharmacogenetics , vol.12 , Issue.4 , pp. 321-330
    • Conrad, S.1    Kauffmann, H.M.2    Ito, K.3
  • 76
    • 0035051412 scopus 로고    scopus 로고
    • MDR1 pharmacogenetics: Frequency of the C3435T mutation in exon 26 is significantly influenced by ethnicity
    • Ameyaw MM, Regateiro F, Li T, et al. 2001. MDR1 pharmacogenetics: frequency of the C3435T mutation in exon 26 is significantly influenced by ethnicity. Pharmacogenetics 11(3):217-21
    • (2001) Pharmacogenetics , vol.11 , Issue.3 , pp. 217-221
    • Ameyaw, M.M.1    Regateiro, F.2    Li, T.3


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.