101
|
Abstract
Large interindividual variation is observed in both the response and toxicity associated with anticancer therapy. The etiology of this variation is multifactorial, but is due in part to host genetic variations. Pharmacogenetic and pharmacogenomic studies have successfully identified genetic variants that contribute to this variation in susceptibility to chemotherapy. This review provides an overview of the progress made in the field of pharmacogenetics and pharmacogenomics using a five-stage architecture, which includes 1) determining the role of genetics in drug response; 2) screening and identifying genetic markers; 3) validating genetic markers; 4) clinical utility assessment; and 5) pharmacoeconomic impact. Examples are provided to illustrate the identification, validation, utility, and challenges of these pharmacogenetic and pharmacogenomic markers, with the focus on the current application of this knowledge in cancer therapy. With the advance of technology, it becomes feasible to evaluate the human genome in a relatively inexpensive and efficient manner; however, extensive pharmacogenetic research and education are urgently needed to improve the translation of pharmacogenetic concepts from bench to bedside.
Collapse
Affiliation(s)
- R Stephanie Huang
- Section of Hematology and Oncology, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | | |
Collapse
|
102
|
Maeda K, Sugiyama Y. Impact of genetic polymorphisms of transporters on the pharmacokinetic, pharmacodynamic and toxicological properties of anionic drugs. Drug Metab Pharmacokinet 2008; 23:223-35. [PMID: 18762709 DOI: 10.2133/dmpk.23.223] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
As the importance of drug transporters in the clinical pharmacokinetics of drugs is recognized, genetic polymorphisms of drug transporters have emerged as one of the determinant factors to produce the inter-individual variability of pharmacokinetics. Many clinical studies have shown the influence of genetic polymorphisms of drug transporters on the pharmacokinetics and subsequent pharmacological and toxicological effects of drugs. The functional change in a transporter in clearance organs such as liver and kidney affects the drug concentration in the blood circulation, while that in the pharmacological or toxicological target can alter the local concentration at the target sites without changing its plasma concentration. As for the transporters for organic anions, some single nucleotide polymorphisms (SNPs) or haplotypes occurring with high frequency in organic anion transporting polypeptide (OATP) 1B1, multidrug resistance 1 (MDR1), and breast cancer resistance protein (BCRP) have been extensively investigated in both human clinical studies and in vitro functional assays. We introduce some examples showing the relationship between haplotypes in transporters and pharmacokinetics and pharmacological effects of drugs. We also discuss how to predict the effect of functional changes in drug transporters caused by genetic polymorphisms on the pharmacokinetics of drugs from in vitro data.
Collapse
Affiliation(s)
- Kazuya Maeda
- Department of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Japan
| | | |
Collapse
|
103
|
Ah YM, Kim YM, Kim MJ, Choi YH, Park KH, Son IJ, Kim SG. Drug-induced Hyperbilirubinemia and the Clinical Influencing Factors. Drug Metab Rev 2008; 40:511-37. [DOI: 10.1080/03602530802341133] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
104
|
Kweekel D, Guchelaar HJ, Gelderblom H. Clinical and pharmacogenetic factors associated with irinotecan toxicity. Cancer Treat Rev 2008; 34:656-69. [DOI: 10.1016/j.ctrv.2008.05.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2008] [Revised: 04/29/2008] [Accepted: 05/02/2008] [Indexed: 01/26/2023]
|
105
|
Hamberg P, De Jong FA, Brandsma D, Verweij J, Sleijfer S. Irinotecan-induced central nervous system toxicity. Report on two cases and review of the literature. Acta Oncol 2008; 47:974-8. [PMID: 17924208 DOI: 10.1080/02841860701666089] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
106
|
Seo T, Ishitsu T, Oniki K, Abe T, Shuto T, Nakagawa K. ABCC2 haplotype is not associated with drug-resistant epilepsy. J Pharm Pharmacol 2008; 60:631-5. [PMID: 18416940 DOI: 10.1211/jpp.60.5.0009] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Several studies have investigated the association between the ABCB1 polymorphism and drug-resistant epilepsy. However, the effect of ABCC2 polymorphisms on anti-epileptic drug (AED) responsiveness remains unknown. The ABCC2 polymorphisms have been genotyped in 279 Japanese epileptic patients treated with AEDs. The association between the AED responsiveness and the polymorphisms was estimated by a haplotype-based analysis. No genotype or haplotype was associated with drug-resistant epilepsy. On the other hand, the delGCGC haplotype at G-1774delG, C-24T, G1249A and C3972T was over represented among the epileptic patients with a complication of mental retardation in comparison with those without (32.4% vs 22.0%; P=0.009); and the G-1774delG allele was also associated with mental retardation (P=0.03). No association between the ABCC2 genotypes or haplotypes, and the responsiveness of AEDs was observed, although this finding was inconclusive because of the small sample size.
Collapse
Affiliation(s)
- Takayuki Seo
- Division of Pharmacology and Therapeutics, Graduate School of Medical and Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | | | | | | | | | | |
Collapse
|
107
|
GSTP1 Ile105Val polymorphism correlates with progression-free survival in MCRC patients treated with or without irinotecan: a study of the Dutch Colorectal Cancer Group. Br J Cancer 2008; 99:1316-21. [PMID: 18797455 PMCID: PMC2570506 DOI: 10.1038/sj.bjc.6604654] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
A Valine residue at position 105 of the GSTP1 protein results in decreased enzyme activity. As nuclear GSTP1 activity decreases irinotecan cytotoxicity, Val-allele carriers may benefit more from irinotecan chemotherapy. Our aim was to investigate the association of GSTP1 genotype with treatment outcome of irinotecan. Progression-free survival (PFS) and toxicity were determined in 267 metastatic colorectal cancer (MCRC) patients who were treated with first-line capecitabine (CAP) plus irinotecan (CAPIRI), or CAP single agent in a prospective randomised phase III trial (CAIRO). GSTP1 genotype was determined by Pyrosequencing. Patients receiving CAP showed a PFS of 6.6 (Ile/Ile), 6.0 (Ile/Val) and 6.5 months (Val/Val); compared to 7.0 (Ile/Ile), 8.8 (Ile/Val) and 9.2 months (Val/Val) with CAPIRI. Median PFS was 2.7 months longer in Val-allele carriers treated with CAPIRI compared to CAP (P=0.005). Patients with the Ile/Ile genotype showed similar PFS with CAPIRI and CAP (7.0 compared to 6.6 months, P=0.972). Toxicity did not differ significantly among genotypes. GSTP1 codon 105 polymorphism may be predictive for the response to irinotecan-based chemotherapy in patients with MCRC, with the Val-allele being associated with a better outcome. Ile/Ile genotype patients do not appear to benefit from the addition of irinotecan to CAP.
Collapse
|
108
|
Lévesque E, Benoit-Biancamano MO, Delage R, Couture F, Guillemette C. Pharmacokinetics of mycophenolate mofetil and its glucuronide metabolites in healthy volunteers. Pharmacogenomics 2008; 9:869-79. [DOI: 10.2217/14622416.9.7.869] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We previously reported that polymorphisms in the UGT2B7 and UGT1A9 genes are associated with significant alteration in the disposition of mycophenolic acid (MPA) in healthy volunteers. Aim: This study further evaluates the impact of genetic polymorphisms at the UGT1A1, UGT1A7 and ABCC2 loci. Methods: Genetic analyses of five UGT candidate genes and ABCC2 were completed on 47 healthy subjects who received a single dose of 1.5 g mycophenolate mofetil and completed a 12-h pharmacokinetic profile. Results: Multivariate analyses indicate that the ABCC2 -24T promoter polymorphism is associated with a 25% increase in acyl mycophenolic acid phenolic glucuronide level. Subjects with combined ABCC2 -24T and UGT1A9*3 genotypes present a 169% increased exposure to AcMPAG. Homozygosity for UGT1A7 387G/391A (129Lys/131Lys) is associated with a modest but significant 7% reduction in MPA level. When these additional genetic factors are considered in the model, the effects of previously described UGT1A9 and UGT2B7 variations remain significant. No significant effect is observed for UGT1A1*28, UGT1A7 622T/C (Trp208Arg), UGT1A9 -440TC/-331CT, UGT1A9 -118 TA9/10 and seven other ABCC2 SNPs. Conclusion: We demonstrate that MPA disposition is a multigenic process, and that additional studies are required to ascertain the relationship between UGT, ABCC2 genotypes and MPA pharmacokinetics in transplant recipients.
Collapse
Affiliation(s)
- Eric Lévesque
- Laboratory of Pharmacogenomics, Oncology & Molecular Endocrinology Research Center, CHUL Research Center and Faculty of Pharmacy, Laval University, T3–48, 2705 Boul. Laurier, Québec, G1V 4G2, Canada
- Department of Hematology and Oncology, Hôtel-Dieu de Québec Hospital, Faculty of Medicine, Laval University, Québec, Canada
| | - Marie-Odile Benoit-Biancamano
- Laboratory of Pharmacogenomics, Oncology & Molecular Endocrinology Research Center, CHUL Research Center and Faculty of Pharmacy, Laval University, T3–48, 2705 Boul. Laurier, Québec, G1V 4G2, Canada
| | - Robert Delage
- Department of Hematology, Enfant-Jésus Hospital, Faculty of Medicine, Laval University, Québec, Canada
| | - Félix Couture
- Department of Hematology and Oncology, Hôtel-Dieu de Québec Hospital, Faculty of Medicine, Laval University, Québec, Canada
| | - Chantal Guillemette
- Laboratory of Pharmacogenomics, Oncology & Molecular Endocrinology Research Center, CHUL Research Center and Faculty of Pharmacy, Laval University, T3–48, 2705 Boul. Laurier, Québec, G1V 4G2, Canada
| |
Collapse
|
109
|
Abstract
Elucidation of the key mechanisms that confer interindividual differences in drug response remains an important focus of drug disposition and clinical pharmacology research. We now know both environmental and host genetic factors contribute to the apparent variability in drug efficacy or in some cases, toxicity. In addition to the widely studied and recognized genes involved in the metabolism of drugs in clinical use today, we now recognize that membrane-bound proteins, broadly referred to as transporters, may be equally as important to the disposition of a substrate drug, and that genetic variation in drug transporter genes may be a major contributor of the apparent intersubject variation in drug response, both in terms of attained plasma and tissue drug level at target sites of action. Of particular relevance to drug disposition are members of the ATP Binding Cassette (ABC) superfamily of efflux transporters. In this review a comprehensive assessment and annotation of recent findings in relation to genetic variation in the Multidrug Resistance Proteins 1-5 (ABCC1-5) and Breast Cancer Resistance Protein (ABCG2) are described, with particular emphasis on the impact of such transporter genetic variation to drug disposition or efficacy.
Collapse
Affiliation(s)
- Ulrike Gradhand
- Division of Clinical Pharmacology, Department of Medicine, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | | |
Collapse
|
110
|
Williams JA, Andersson T, Andersson TB, Blanchard R, Behm MO, Cohen N, Edeki T, Franc M, Hillgren KM, Johnson KJ, Katz DA, Milton MN, Murray BP, Polli JW, Ricci D, Shipley LA, Vangala S, Wrighton SA. PhRMA white paper on ADME pharmacogenomics. J Clin Pharmacol 2008; 48:849-89. [PMID: 18524998 DOI: 10.1177/0091270008319329] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Pharmacogenomic (PGx) research on the absorption, distribution, metabolism, and excretion (ADME) properties of drugs has begun to have impact for both drug development and utilization. To provide a cross-industry perspective on the utility of ADME PGx, the Pharmaceutical Research and Manufacturers of America (PhRMA) conducted a survey of major pharmaceutical companies on their PGx practices and applications during 2003-2005. This white paper summarizes and interprets the results of the survey, highlights the contributions and applications of PGx by industrial scientists as reflected by original research publications, and discusses changes in drug labels that improve drug utilization by inclusion of PGx information. In addition, the paper includes a brief review on the clinically relevant genetic variants of drug-metabolizing enzymes and transporters most relevant to the pharmaceutical industry.
Collapse
Affiliation(s)
- J Andrew Williams
- Pfizer Global Research and Development, 10646 Science Center Drive (CB10), San Diego, CA 92121, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
111
|
Abstract
The purpose of this study was to evaluate the affinity of docetaxel for 14 transporter proteins and assess the functional significance of 17 variants in five genes involved in drug elimination. Among the transfected models investigated, OATP1B3 (SLCO1B3) was identified as the most efficient influx transporter for docetaxel. None of the observed genotypes (SLCO1B3, ABCB1, and ABCC2) was related with docetaxel clearance in 92 white patients (P > 0.17). However, the simultaneous presence of the CYP3A4*1B and CYP3A5*1A alleles was associated with a 64% increase in docetaxel clearance (P = 0.0015), independent of both sex and CYP3A activity (as determined using the erythromycin breath test). This haplotype was also associated with increased midazolam clearance in another population (P = 0.0198). An analysis of the CYP3A locus among CEPH-HapMap samples revealed that CYP3A4*1B is present exclusively among a subset of CYP3A5 expressors. Therefore, future studies should first stratify the population on the basis of CYP3A5 genotype and then compare CYP3A activity between individuals with and without the CYP3A4*1B allele.
Collapse
|
112
|
Influence of genetic polymorphisms on intestinal expression and rifampicin-type induction of ABCC2 and on bioavailability of talinolol. Pharmacogenet Genomics 2008; 18:357-65. [PMID: 18334920 DOI: 10.1097/fpc.0b013e3282f974b7] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES To evaluate whether ABCC2 gene polymorphisms are associated with expression and/or function of the efflux pump. METHODS We investigated the allele frequency of ABCC2 -24C>T, -23G>A, c.1249G>A, c.1446C>G, c.1457C>T, c.2302C>T, c.2366C>T, c.3542G>T, c.3561G>A, c.3563T>A, c.3972C>T, c.4348G>A, and 4544G>A in 374 nonrelated German healthy volunteers and determined the impact on duodenal mRNA and protein content of ABCC2. For functional analysis, the disposition of intravenously (30 mg) and orally administered talinolol (100 mg) was measured among 31 individuals. Moreover, the effects of rifampicin-type induction (600 mg, 8 days) of duodenal ABCC2 were quantified in 22 participants with regard to genetic polymorphisms. RESULTS The allele frequencies were 18.3% (-24T), 21.1% (1249A), 1.4% (1446G), 0.1% (3542T), 4.5% (3563A), 34.2% (3972T), and 4.4% (4544A); carriers of -23G>A, 1457C>T, 2302C>T, 2366C>T, 3561G>A, and 4348G>A were not identified. The -24T allele was in strong linkage with 3972T, and 3563A with 4544A, whereas 1249A was weakly linked with other variant alleles. None of the single nucleotide polymorphisms investigated influenced significantly intestinal ABCC2 mRNA and protein content. The variant ABCC2 1249G>A (V417I), however, was associated with lower oral bioavailability (P=0.001), and increased residual clearance of intravenous talinolol (P=0.021). Intestinal ABCC2 mRNA and protein expression were upregulated by rifampicin treatment, a genetic influence could be detected in only four cases heterozygote for 3563T>A or 4544G>A. CONCLUSION The 1249G>A (V417I) polymorphism is obviously associated with higher activity of the intestinal transporter.
Collapse
|
113
|
Sato T, Hatate K, Ikeda A, Yamanashi T, Ozawa H, Onosato W, Nakamura T, Ihara A, Watanabe M. Treatment of advanced or recurrent colorectal cancer with irinotecan in Japan and elsewhere. Expert Opin Pharmacother 2008; 9:1223-8. [DOI: 10.1517/14656566.9.7.1223] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
114
|
Abstract
Although approximately half of the administered dose of irinotecan is recovered in urine, scarce data are available on the association of renal function with irinotecan pharmacokinetics and toxicity. Here, these relationships are investigated in 187 patients treated with irinotecan in a three-weekly schedule. No significant effects on irinotecan pharmacokinetics were found in these patients. However, in 131 patients treated with the registered dose, categorized renal function was related to hematological toxicity. The incidence of grade 3-4 neutropenia decreased as function of creatinine clearance, particularly in nonsmoking patients (P < 0.01). Patients with slower creatinine clearance (35-66 ml/min) had a four-times higher risk of grade 3-4 neutropenia (58% vs. 14%; P < 0.001). This study suggests that pretreatment renal function values are associated with irinotecan-induced neutropenia. A confirmatory analysis is warranted to determine whether measures of renal function should be incorporated in future attempts toward individualized treatment with irinotecan.
Collapse
|
115
|
Integrated pharmacogenetic prediction of irinotecan pharmacokinetics and toxicity in patients with advanced non-small cell lung cancer. Lung Cancer 2008; 63:115-20. [PMID: 18221820 DOI: 10.1016/j.lungcan.2007.12.003] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2007] [Revised: 11/29/2007] [Accepted: 12/03/2007] [Indexed: 12/24/2022]
Abstract
To define an integrated pharmacogenetic model for predicting irinotecan pharmacokinetic (PK) and severe toxicity, we evaluated multivariate analysis using 15 polymorphisms within seven genes with putative influence on metabolism and transport of irinotecan. A total of 107 NSCLC patients treated with irinotecan were evaluated for PK and genotyped for the UGT1A1*6, UGT1A1*28, UGT1A9*22, ABCB11236C>T, 2677G>T/A, 3435C>T, ABCC2-24C>T, 1249G>A, 3972C>T, ABCG234G>A, 421C>A, and SLCO1B1 -11187G>A, 388A>G, and 521T>C, and CYP3A5*3 polymorphisms. Multivariate linear and logistic regression analyses including genotypes and clinicopathologic factors were performed. SN-38 AUC was significantly correlated with ANCs (r=-0.3, p=0.009) and grade 4 neutropenia (p=0.01). The UGT1A1*6/*6, UGT1A9*1/*1 or *1/*22, and SLCO1B1 521TC or CC genotypes, and female-gender were predictive for higher AUC(SN-38) in multivariate analysis. Among them, SLCO1B1 521TC or CC and UGT1A1*6/*6 genotypes were independently predictive for grade 4 neutropenia in multivariate analysis (OR=3.8 and 7.4, respectively). Although no significant association was observed between PK parameters and grade 3 diarrhea, UGT1A9*1/*1, ABCC23972CC, and ABCG234GA or AA genotypes were independently predictive for grade 3 diarrhea in multivariate analysis (OR=6.3, 5.6, and 5.1, respectively). Patient selection based on integrated pharmacogenetic model would be helpful for predicting irinotecan-PK and severe toxicities in NSCLC patients.
Collapse
|
116
|
Abstract
Paclitaxel is widely used in many cancers including ovarian, breast, lung, head and neck and primary unknown. Paclitaxel is extensively metabolized by cytochrome P450s and excreted in bile. The cytochromes involved include 2C8 and 3A4. This is a review of the pharmacokinetics, pharmacodynamics, drug interactions, metabolism and pharmacogenomics of paclitaxel.
Collapse
Affiliation(s)
- Helen Steed
- University of Alberta, Department of Gyne-oncology, Department of Obstetrics and Gynecology, Cross Cancer Institute, Edmonton, Alberta, Canada
| | | |
Collapse
|
117
|
Sai K, Saito Y, Itoda M, Fukushima-Uesaka H, Nishimaki-Mogami T, Ozawa S, Maekawa K, Kurose K, Kaniwa N, Kawamoto M, Kamatani N, Shirao K, Hamaguchi T, Yamamoto N, Kunitoh H, Ohe Y, Yamada Y, Tamura T, Yoshida T, Minami H, Matsumura Y, Ohtsu A, Saijo N, Sawada JI. Genetic Variations and Haplotypes of ABCC2 Encoding MRP2 in a Japanese Population. Drug Metab Pharmacokinet 2008; 23:139-47. [DOI: 10.2133/dmpk.23.139] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
118
|
Marsh S, Phillips MS. Integrating pharmacogenomics into oncology clinical practice. Expert Rev Clin Pharmacol 2008; 1:73-80. [PMID: 24410511 DOI: 10.1586/17512433.1.1.73] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Oncology pharmacogenomics has seen a great deal of progress in the past 10 years. The release of the Human Genome Project data and the availability of fast, affordable genotyping platforms has allowed the field to expand and has provided invaluable data for pharmacogenomics research. The introduction of US FDA-approved targeted therapy (trastuzumab), package insert changes (irinotecan and tamoxifen) and the initiation of a genotype-guided clinical trial for cancer therapy (TYMS TSER in rectal cancer), along with panels of DNA and expression markers (Roche AmpliChip(®) and Oncotype Dx™ panel) are paving the way towards the integration of pharmacogenomics into clinical practice.
Collapse
Affiliation(s)
- Sharon Marsh
- Washington University in St Louis, Division of Oncology, 660 South Euclid Ave - Campus Box 8069, St Louis, MO 63110, USA.
| | | |
Collapse
|
119
|
Affiliation(s)
- S Marsh
- Washington University, Division of Molecular Oncology, St Louis, USA
| |
Collapse
|
120
|
Flieger D, Klassert C, Hainke S, Keller R, Kleinschmidt R, Fischbach W. Phase II Clinical Trial for Prevention of Delayed Diarrhea with Cholestyramine/Levofloxacin in the Second-Line Treatment with Irinotecan Biweekly in Patients with Metastatic Colorectal Carcinoma. Oncology 2007; 72:10-6. [DOI: 10.1159/000111083] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2007] [Accepted: 06/21/2007] [Indexed: 11/19/2022]
|
121
|
Gamelin L, Capitain O, Morel A, Dumont A, Traore S, Anne LB, Gilles S, Boisdron-Celle M, Gamelin E. Predictive Factors of Oxaliplatin Neurotoxicity: The Involvement of the Oxalate Outcome Pathway. Clin Cancer Res 2007; 13:6359-68. [DOI: 10.1158/1078-0432.ccr-07-0660] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
122
|
Sparreboom A, Wolff AC, Mathijssen RH, Chatelut E, Rowinsky EK, Verweij J, Baker SD. Evaluation of Alternate Size Descriptors for Dose Calculation of Anticancer Drugs in the Obese. J Clin Oncol 2007; 25:4707-13. [DOI: 10.1200/jco.2007.11.2938] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Despite the rising prevalence of obesity, there is paucity of information describing how doses of anticancer drugs should be adjusted in clinical practice. Here, we assessed the pharmacokinetics of eight anticancer drugs in adults and evaluated the potential utility of alternative weight descriptors in dose calculation for the obese. Patients and Methods A total of 1,206 adult cancer patients were studied, of whom 162 (13.4%) were obese (body mass index ≥ 30). Pharmacokinetic parameters were calculated using noncompartmental analysis, and compared between lean (body mass index ≤ 25) and obese patients. Results The absolute clearance of cisplatin, paclitaxel, and troxacitabine was significantly increased in the obese (P < .023), but this was not observed for carboplatin, docetaxel, irinotecan, or topotecan (P < .17). For doxorubicin, the systemic clearance was statistically significantly reduced in obese women (P = .013), but not in obese men (P = .52). Evaluation of alternate weight descriptors for dose calculation in the obese, including predicted normal weight, lean body mass, (adjusted) ideal body weight, and the mean of ideal and actual body weight, indicated that, for most of the evaluated drugs, weight scalars used to calculate body-surface area should consider actual body weight regardless of size. Conclusion The results suggest that a number of widely used empiric strategies for dose adjustments in obese patients, including a priori dose reduction or dose capping, should be discouraged.
Collapse
Affiliation(s)
- Alex Sparreboom
- From the Department of Medical Oncology, Erasmus MC–Daniel den Hoed Cancer Center, Rotterdam, the Netherlands; The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Institut Claudius-Regaud, Toulouse, France; and the Institute for Drug Development, Cancer Therapy and Research Center, San Antonio, TX
| | - Antonio C. Wolff
- From the Department of Medical Oncology, Erasmus MC–Daniel den Hoed Cancer Center, Rotterdam, the Netherlands; The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Institut Claudius-Regaud, Toulouse, France; and the Institute for Drug Development, Cancer Therapy and Research Center, San Antonio, TX
| | - Ron H.J. Mathijssen
- From the Department of Medical Oncology, Erasmus MC–Daniel den Hoed Cancer Center, Rotterdam, the Netherlands; The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Institut Claudius-Regaud, Toulouse, France; and the Institute for Drug Development, Cancer Therapy and Research Center, San Antonio, TX
| | - Etienne Chatelut
- From the Department of Medical Oncology, Erasmus MC–Daniel den Hoed Cancer Center, Rotterdam, the Netherlands; The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Institut Claudius-Regaud, Toulouse, France; and the Institute for Drug Development, Cancer Therapy and Research Center, San Antonio, TX
| | - Eric K. Rowinsky
- From the Department of Medical Oncology, Erasmus MC–Daniel den Hoed Cancer Center, Rotterdam, the Netherlands; The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Institut Claudius-Regaud, Toulouse, France; and the Institute for Drug Development, Cancer Therapy and Research Center, San Antonio, TX
| | - Jaap Verweij
- From the Department of Medical Oncology, Erasmus MC–Daniel den Hoed Cancer Center, Rotterdam, the Netherlands; The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Institut Claudius-Regaud, Toulouse, France; and the Institute for Drug Development, Cancer Therapy and Research Center, San Antonio, TX
| | - Sharyn D. Baker
- From the Department of Medical Oncology, Erasmus MC–Daniel den Hoed Cancer Center, Rotterdam, the Netherlands; The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Institut Claudius-Regaud, Toulouse, France; and the Institute for Drug Development, Cancer Therapy and Research Center, San Antonio, TX
| |
Collapse
|
123
|
Abstract
Irinotecan is widely used in the treatment of metastatic colorectal cancer and extensive small-cell lung cancer. Its use is limited by severe toxicities such as neutropenia and delayed-type diarrhea. Irinotecan is converted to its active metabolite SN-38. SN-38 is further metabolized to SN-38G by various hepatic and extrahepatic UGT1A isozymes, mainly UGT1A1. Impaired glucuronidation activity of the UGT1A1 enzyme has been linked with elevated levels of SN-38, leading to toxicities. UGT1A1*28 involves an extra TA repeat in the UGT1A1 promoter region and is the variant most frequently contributing to interpatient variability in irinotecan pharmacokinetics and toxicities. This information led to the revision of the irinotecan label by the US Food and Drug Administration. Recently, UGT1A1*6 seems to contribute to the risk of toxicity of irinotecan in Asian patients. The pharmacogenetics of irinotecan (irinogenetics) is one of few promising examples of the application of pharmacogenetics to individualized drug therapy. This review summarizes ongoing studies and unanswered questions on irinogenetics.
Collapse
Affiliation(s)
- Tae Won Kim
- Section of Oncology, Department of Medicine, Asan Medical Center, University of Ulsan, Seoul, Korea
| | | |
Collapse
|
124
|
van der Bol JM, Mathijssen RHJ, Loos WJ, Friberg LE, van Schaik RHN, de Jonge MJA, Planting AST, Verweij J, Sparreboom A, de Jong FA. Cigarette smoking and irinotecan treatment: pharmacokinetic interaction and effects on neutropenia. J Clin Oncol 2007; 25:2719-26. [PMID: 17563393 DOI: 10.1200/jco.2006.09.6115] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
PURPOSE Several constituents of cigarette smoke are known to interact with drug metabolizing enzymes and potentially affect treatment outcome with substrate drugs. The purpose of this study was to determine the effects of cigarette smoking on the pharmacokinetics and adverse effects of irinotecan. PATIENTS AND METHODS A total of 190 patients (49 smokers, 141 nonsmokers) treated with irinotecan (90-minute intravenous administration on a 3-week schedule) were evaluated for pharmacokinetics. Complete toxicity data were available in a subset of 134 patients receiving 350 mg/m2 or 600 mg flat-fixed dose irinotecan. RESULTS In smokers, the dose-normalized area under the plasma concentration-time curve of irinotecan was significantly lower (median, 28.7 v 33.9 ng x h/mL/mg; P = .001) compared with nonsmokers. In addition, smokers showed an almost 40% lower exposure to SN-38 (median, 0.54 v 0.87 ng x h/mL/mg; P < .001) and a higher relative extent of glucuronidation of SN-38 into SN-38G (median, 6.6 v 4.5; P = .006). Smokers experienced considerably less hematologic toxicity. In particular, the incidence of grade 3 to 4 neutropenia was 6% in smokers versus 38% in nonsmokers (odds ratio [OR], 0.10; 95% CI, 0.02 to 0.43; P < .001). There was no significant difference in incidence of delayed-onset diarrhea (6% v 15%; OR, 0.34; 95% CI, 0.07 to 1.57; P = .149). CONCLUSION This study indicates that smoking significantly lowers both the exposure to irinotecan and treatment-induced neutropenia, indicating a potential risk of treatment failure. Although the underlying mechanism is not entirely clear, modulation of CYP3A and uridine diphosphate glucuronosyltransferase isoform 1A1 may be part of the explanation. The data suggest that additional investigation is warranted to determine whether smokers are at increased risk for treatment failure.
Collapse
Affiliation(s)
- Jessica M van der Bol
- Department of Medical Oncology, Erasmus MC University Medical Center, Daniel den Hoed Cancer Center, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
125
|
Engels FK, de Jong FA, Sparreboom A, Mathot RAA, Loos WJ, Kitzen JJEM, de Bruijn P, Verweij J, Mathijssen RHJ. Medicinal Cannabis Does Not Influence the Clinical Pharmacokinetics of Irinotecan and Docetaxel. Oncologist 2007; 12:291-300. [PMID: 17405893 DOI: 10.1634/theoncologist.12-3-291] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE To date, data regarding the potential of cannabinoids to modulate cytochrome P450 isozyme 3A (CYP3A) activity are contradictory. Recently, a standardized medicinal cannabis product was introduced in The Netherlands. We anticipated an increased use of medicinal cannabis concurrent with anticancer drugs, and undertook a drug-interaction study to evaluate the effect of concomitant medicinal cannabis on the pharmacokinetics of irinotecan and docetaxel, both subject to CYP3A-mediated biotransformation. PATIENTS AND METHODS Twenty-four cancer patients were treated with i.v. irinotecan (600 mg, n = 12) or docetaxel (180 mg, n = 12), followed 3 weeks later by the same drugs concomitant with medicinal cannabis (200 ml herbal tea, 1 g/l) for 15 consecutive days, starting 12 days before the second treatment. Blood samples were obtained up to 55 hours after dosing and analyzed for irinotecan and its metabolites (SN-38, SN-38G), respectively, or docetaxel. Pharmacokinetic analyses were performed during both treatments. Results are reported as the mean ratio (95% confidence interval [CI]) of the observed pharmacokinetic parameters with and without concomitant medicinal cannabis. RESULTS Medicinal cannabis administration did not significantly influence exposure to and clearance of irinotecan (1.04; CI, 0.96-1.11 and 0.97; CI, 0.90-1.05, respectively) or docetaxel (1.11; CI, 0.94-1.28 and 0.95; CI, 0.82-1.08, respectively). CONCLUSION Coadministration of medicinal cannabis, as herbal tea, in cancer patients treated with irinotecan or docetaxel does not significantly influence the plasma pharmacokinetics of these drugs. The evaluated variety of medicinal cannabis can be administered concomitantly with both anticancer agents without dose adjustments.
Collapse
Affiliation(s)
- Frederike K Engels
- Department of Medical Oncology, Erasmus MC University Medical Center Rotterdam - Daniel den Hoed Cancer Center, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|