Skip to main content
Erschienen in: Journal of Cancer Research and Clinical Oncology 9/2013

Open Access 01.09.2013 | Original Paper

Analysis of UGT1A1*28 genotype and SN-38 pharmacokinetics for irinotecan-based chemotherapy in patients with advanced colorectal cancer: results from a multicenter, retrospective study in Shanghai

verfasst von: Xun Cai, Weiguo Cao, Honghua Ding, Tianshu Liu, Xinli Zhou, Mei Wang, Ming Zhong, Ziyi Zhao, Qing Xu, Liwei Wang

Erschienen in: Journal of Cancer Research and Clinical Oncology | Ausgabe 9/2013

Abstract

Background

The UGT1A1*28 polymorphism, although closely linked with CPT-11-related adverse effects, cannot be used alone to guide individualized treatment decisions. However, CPT-11 dosage can be adjusted according to measured SN-38 pharmacokinetics. Our study is designed to investigate whether there is a relationship between SN-38 peak or valley concentrations and efficacy or adverse effects of CPT-11-based chemotherapy. We retrospectively studied 98 patients treated with advanced colorectal cancer in various UGT1A1*28 genotype groups (mainly (TA)6/(TA)6 and (TA)6/(TA)7 genotypes) treated with CPT-11 as first-line chemotherapy in Shanghai.

Methods

One hundred and sixty-four advanced colorectal cancer patients were enrolled. To understand differences in genotype expression, the frequency of UGT1A1*28 thymine–adenine (TA) repeats in TATA box arrangement was assessed by PCR with genomic DNA extracted from peripheral blood. For ninety-eight cases with the (TA)6/(TA)6 and (TA)6/(TA)7 genotypes treated with CPT-11 as first-line chemotherapy, the plasma concentration of SN-38 was detected by HPLC 1.5 and 49 h after CPT-11 infusion. Efficacy and adverse effects were observed subsequently, and the relationship between SN-38 plasma concentration and efficacy or adverse effects within genotype groups, as well as differences in efficacy and adverse effects between (TA)6/(TA)6 and (TA)6/(TA)7 genotypes were analyzed statistically.

Results

One hundred and fourteen patients (69.51 %) were identified with the (TA)6/(TA)6 genotype, forty-eight patients (29.27 %) with the (TA)6/(TA)7 genotype, and two patients (1.22 %) with the (TA)7/(TA)7 genotype. The average peak and valley concentrations of SN-38 after CPT-11 infusion and plasma bilirubin average levels before and after CPT-11 treatment in the (TA)6/(TA)7 genotype group were all higher than those in (TA)6/(TA)6 group, and the difference was statistically significant (p = 0.00). Stepwise regression analysis showed that SN-38 peak and valley concentration was correlated with PFS in the (TA)6/(TA)6 genotype. In the (TA)6/(TA)7 group, SN-38 peak concentration was correlated with CPT-11 starting dose and OS, valley concentration correlated with plasma bilirubin levels before CPT-11 treatment, delayed diarrhea, and OS. For the (TA)6/(TA)6 genotype, mPFS of the SN-38 peak concentration >43.2 ng/ml subgroup was significantly longer than that of ≤43.2 ng/ml subgroup (8.0 ± 0.35 vs. 6.5 ± 0.79 months, χ 2 = 17.18, p = 0.00) with a relatively high incidence of Grade I/II° myelosuppression; for the (TA)6/(TA)7 genotype, there was no significant difference in mOS between the SN-38 valley concentration >16.83 ng/ml and ≤16.83 subgroups (17.3 ± 0.45 vs. 18.8 ± 0.50 months, χ 2 = 1.38, p = 0.24), but the former had a higher incidence of Grade III/IV° mucositis and delayed diarrhea. For 2 (TA)7/(TA)7 cases, although 25 % dose reduction of CPT-11, which is calculated according to body surface area, Grade IV° bone marrow suppression and Grade III° delayed diarrhea still occurred after CPT-11 treatment, though both adverse effects resolved and did not recur again after a 50 % dose reduction.

Conclusion

The (TA)6/(TA)6 genotype and (TA)6/(TA)7 genotype accounted for the most, and (TA)7/(TA)7 genotype only account for a very small portion of advanced colorectal cancer patients in Shanghai. For the (TA)6/(TA)6 genotype, CPT-11 dosage can be increased gradually to improve efficacy for patients with SN-38 peak concentration ≤43.2 ng/ml after CPT-11 infusion; and for (TA)6/(TA)7 genotype patients, CPT-11 dosage may be lowered appropriately to reduce serious adverse effects such as bone marrow suppression and delayed diarrhea without affecting the efficacy for those with SN-38 valley concentration >16.83 ng/ml. For (TA)7/(TA)7 genotype patients, adverse effects should be closely observed after treatment even if CPT-11 dosage has been reduced.
Abkürzungen
HPLC
High-performance liquid chromatography
PFS
Progression-free survival
mPFS
Median PFS
OS
Overall survival
mOS
Median OS
UGT1A1
Uridine diphosphate glucuronosyl transferase 1A1
SN-38
7-ethyl-10-hydroxycamptothecin
SN-38G
SN-38 glucuronide
AUCSN-38
Area under the curve of SN-38
AUCSN-38G/AUCSN-38
The ratio of area under the curve of SN-38G and SN-38
SAP
Shrimp alkaline phosphatase
IS
Internal standard
RECIST
The response evaluation criteria in solid tumors
CTCAE
National Cancer Institute common terminology criteria for adverse events
hCES
Human carboxylesterase

Introduction

China has a high incidence of colorectal cancer due to a variety of factors including lifestyle changes with 400,000 newly diagnosed cases each year. Colorectal cancer is the second most common malignant tumor in Shanghai, with incidence increasing by 3.67 times since the early 1970s. Between 40 and 70 % patients will experience postoperative recurrence and metastasis following radical surgery for early-stage disease. CPT-11-based chemotherapy has been supported by the research as a standard first-line treatment for advanced colorectal cancer. However, Grade III/IV° bone marrow suppression or delayed diarrhea caused by CPT-11 may occur in some patients during or after treatment, which leads to treatment delays and affect patients’ quality of life (Douillard et al. 2000; Innocenti et al. 2004). Many studies have confirmed that UGT1A1 is the main metabolic enzyme that inactivates SN-38, the active product of CPT-11, into SN-38G. Ethnic differences exist in UGT1A1 enzyme activity and gene polymorphisms (Kaniwa et al. 2005), which are closely related to CPT-11 adverse effects (Innocenti et al. 2004; Massacesi et al. 2006) but they are uncertain with efficacy (Shulman et al. 2011; Liu et al. 2008). In addition, studies have found that AUCSN-38 or AUCSN-38G/AUCSN-38 is associated with neutropenia after CPT-11 treatment (Hirose et al. 2012; Canal et al. 1996), which indicate that CPT-11 or SN-38 pharmacokinetics is as important as UGT1A1 gene polymorphism analysis in predicting CPT-11-related adverse effects, even in dose individualization sets. Presently, data are limited about the role of UGT1A1*28 genotype and SN-38 pharmacokinetics in predicting CPT-11 efficacy and adverse effects, so our study reviewed the plasma SN-38 peak and valley concentration, treatment efficacy, and adverse effects in 98 cases with (TA)6/(TA)6 and (TA)6/(TA)7 genotypes who were treated with CPT-11 as first-line chemotherapy, in order to determine whether there is a relationship between plasma SN-38 peak and valley concentration and efficacy or adverse effects in patients with different genotypes, and provide theoretical basis for CPT-11 treatment individualization based on UGT1A1*28 genotype in combination with SN-38 pharmacokinetics analysis.

Materials and methods

Patient eligibility

A total of 164 hospitalized patients with local advanced and metastatic colorectal cancer were eligible for the study. Disease was confirmed with pathological and imaging data in Zhongshan Hospital, Huashan Hospital affiliated with Fudan University, Ruijin hospital, Renji hospital affiliated with Shanghai Jiaotong University Medical College, Changhai hospital affiliated with the Second Military University, Shanghai No. 1 and No. 10 People’s Hospital, and the Central Hospital of Jing’an District from April 2010 to January 2012. Of the eligible patients, 117 were males and 47 were females, aged from 26 to 75 years with a median age of 60 years, and included 100 cases of colon cancer and 64 cases of rectal cancer. Twenty cases had stage IIIb disease and 144 cases had stage IV disease according to the AJCC Cancer Staging standard (6th edition). Inclusion criteria were as follows: ECOG physical status scores from 0 to 2, with measurable disease not treated and life expectancy of ≥3 months; of all the 164 cases, 98 cases were assigned to receive a modified FOLFIRI chemotherapy regimen for at least 4 cycles as first-line palliative chemotherapy after excluding chemotherapy contraindications and obtaining informed consent. Plasma bilirubin and transaminase level could not exceed 1.5 times and 5 times the normal upper limit for patients with liver metastases. Exclusion criteria such as pregnant or lactating women, patients with complete or incomplete intestinal obstruction or with a history of chronic enteritis or extensive bowel resection, or with central nervous system metastases, having been seriously allergic to drugs and its excipients used in the treatment, evaluable lesions which had received radiotherapy or other treatments, vital organ dysfunction, history of other malignancies (cervical carcinoma in situ or skin basal cell carcinoma excluded) and poor compliance.
About 2 ml of peripheral blood sample (anticoagulated with EDTA) was obtained from each patient before chemotherapy for UGT1A1*28 genotype analysis, and then the patients received FOLFIRI chemotherapy 1.5-h infusion of CPT-11 (180 mg/m2) and 2-h infusion of leucovorin (400 mg/m2), followed by 5-fluorouracil bolus (400 mg/m2) on day 1 and a 46-h continuous infusion (2,400 mg/m2) for each cycle, repeated every 2 weeks. About 2 ml of peripheral blood sample was taken 1.5 and 49 h after CPT-11 infusion for SN-38 plasma concentration detection by HPLC (Hirose et al. 2012; Schoemaker et al. 2003), and CPT-11 dosage was lowered by 20–25 % in case of Grade III° adverse effects.

UGT1A1*28 genotype analysis

Genomic DNA was extracted with DNA extraction kit (Qiagen Inc, Valencia, CA, USA) for PCR amplification. Prime sequences were designed as follows: Upstream: 5′-TCCCTGCTACCTTTGTGGAC-3′, downstream: 5′-AGCAGGCCCAGGACAAGT-3′. The PCR mix was (25 μl): 2 μl of 10× PCR buffer with 15 mM MgCl2, 2 μl of dNTP (2.5 mM), 1 μl of Primers (10 μm), 1 μl of DNA templates, 0.2 μl of DNA Taq polymerase (5U/μl), and 18.8 μl of ddH2O. Amplification procedure was as follows: a initial denaturation step at 94 °C for 5 min; followed by 40 cycles of denaturation (94 °C for 15 s), annealing (55 °C for 25 s) and extension (72 °C for 50 s), and finally, extension at 72 °C for 7 min. If the PCR product band is clear by electrophoresis, take 5 μl of eligible specimen, mixed by adding 2 μl SAP, kept at 37 °C for 60 min and 80 °C for 15 min, and then saved at 4 °C. Took 3 μl positive PCR hydrolysates, 1 μl sequencing reagent (bigdye), and 2 μl sequencing primer for PCR amplification: initial denaturation at 96 °C for 1 min, followed by 25 cycles of denaturation (96 °C for 10 s), annealing (50 °C for 5 s) and extension (60 °C for 4 min), and then stored at 4 °C. The sequencing product was sequenced in DNA sequencing instrument (ABI-373, PE corp., USA) after being purified. Sequencing results were displayed and analyzed with GeneMapper software.

SN-38 plasma concentration detection

SN-38 and IS (acetone) were dissolved with dimethyl sulfoxide into 1.0 mg/ml stock solution stored at −80 °C. SN-38 reference standard plasma was prepared at a concentration range of 2–500 ng/ml with blank plasma. We then took 200 μl of the mentioned plasma, vortexed with acetonitrile (100 ng/ml) for 1 min, and centrifuged at 9,500 rpm for 10 min, and then tested after taking 200 μl supernatant vortexed with 100 μl hydrochloric acid (1 mol/L) for 30 s. The standard curve and linear regression were prepared by taking the ratio of peak area of SN-38 and IS as the vertical axis. Chromatographic condition: Waters®Nova-Pak C18 Guard column (20 mm × 3.9 mm, 5 μm of particle diameter); mobile phase: disodium hydrogen phosphate (0.05 mol/L, pH = 4.0, containing 0.05 mol/L 1-heptanesulfonic acid)-acetonitrile (75:25, v/v). Testing condition: flow rate of 1 ml/min, column temperature at 25 °C, excitation wavelength at 370 nm, and emission wavelength at 470 nm. The retention time for SN-38 and IS was 15.4 and 17.8 min under the experimental condition.

Evaluation criteria and follow-up

The first efficacy follow-up was performed for all patients by imaging scans after 3 cycles of chemotherapy, and partial remission required reconfirmation at least 4 weeks after initial assessment. Re-evaluation was to be done every 3 cycles until disease progression and patients to be followed for overall survival every 3 months. Patients could receive other chemotherapy or best supportive care after disease progression, but not include targeted therapy. The median follow-up was 16 month. Tumor efficacy was evaluated using RECIST, and toxicity was graded according to CTCAE version 3.0.

Statistical analyses

Data were expressed as mean ± standard deviation. Prognostic factors such as mPFS and mOS were calculated using the Kaplan–Meier method, and survival differences were analyzed by log rank test. A t test was used to compare the difference between the groups and a chi-square test to compare count data. Degree of adverse effects, plasma concentration, and efficacy was accessed by variance analysis. All values were two-sided, and statistical significance was accepted at the p < 0.05 level. SPSS version 16.0 software (SPSS Inc., USA) was used for all statistical analyses.

Results

UGT1A1 polymorphism for patients with advanced colorectal cancer in Shanghai

One hundred and fourteen patients (69.51 %) were identified with the (TA)6/(TA)6 genotype, forty-eight patients (29.27 %) with the (TA)6/(TA)7 genotype, and two patients (1.22 %) with the (TA)7/(TA)7 genotype (for sequencing results, see Fig. 1), and there were no statistical differences in age, gender, ECOG performance score, primary tumor site, TMN staging, and chemotherapy order between (TA)6/(TA)6 and (TA)6/(TA)7 groups except for CPT-11 dosage reduction in the (TA)6/(TA)7 group (see Table 1).
Table 1
Comparison of clinical characteristics between (TA)6/(TA)6 and (TA)6/(TA)7 groups
Clinical characteristics
6/6 genotype (n = 114)
6/7 genotype (n = 48)
F
P
ECOG performance score
  
2.34
0.27
 0′
34
10
  
 1′
80
38
  
Gender
  
1.87
0.31
 Male
82
34
  
 Female
32
14
  
Age (year)
57.54 ± 10.38
58.35 ± 10.06
0.21
0.65
Primary tumor site
  
0.66
0.50
 Colon
66
34
  
 Rectum
48
14
  
TMN staging
  
3.86
0.19
 IIIb
12
8
  
 IV
102
40
  
Chemotherapy order
  
0.42
0.58
 First line
74
24
  
 Second line
40
24
  
CPT-11 starting dosage (mg)
297.72 ± 35.30
299.17 ± 43.46
0.05
0.83
CPT-11 dosage reduction
  
23.67
0.00
 Yes
8
17
  
 No
106
31
  

SN-38 peak and valley concentration after CPT-11 infusion and plasma bilirubin level before and after treatment for (TA)6/(TA)6 and (TA)6/(TA)7 groups treated with first-line chemotherapy

SN-38 average peak and valley concentration after CPT-11 infusion was 45.57 ± 19.38 and 8.67 ± 5.45 ng/ml, respectively, for the (TA)6/(TA)6 genotype group, and 71.80 ± 9.15 and 15.39 ± 7.25 ng/ml for the (TA)6/(TA)7 genotype, and the difference was statistically significant (t = 6.39, p = 0.00; t = 4.82, p = 0.00). Plasma bilirubin levels were 9.42 ± 2.43 and 9.56 ± 2.26 μmol/L for (TA)6/(TA)6 group, and 13.91 ± 3.90 and 14.44 ± 2.99 μmol/L for (TA)6/(TA)7 group before and after treatment, respectively, with significant difference between the groups (t = 6.69, p = 0.00; t = 8.46, p = 0.0) but without significant difference before and after treatment within groups (t = 0.35, p = 0.72; t = 0.53, p = 0.60), see Fig. 2.

Stepwise regression analysis between SN-38 peak and valley concentration and efficacy, adverse effects

Taking SN-38 peak and valley concentration as dependent variables, and adverse reactions, short-term effect, PFS, and OS as independent variables, stepwise regression analysis showed that, in the (TA)6/(TA)6 group, SN-38 peak and valley concentration was related to PFS, but in the (TA)6/(TA)7 genotype, SN-38 peak concentration was related to CPT-11 starting dose and OS, and the valley concentration was related to plasma bilirubin levels before CPT-11 treatment, delayed diarrhea incidence, and OS (regression equation coefficients are shown in Table 2).
Table 2
Stepwise regression of SN-38 peak, valley concentrations with efficacy, and adverse effects
 
Unstandardized coefficients
Standardized coefficients
t
p
B
SE
Beta
(TA)6/(TA)6 genotype
 SN-38 peak concentration
  Constant
−9.53
7.47
 
−1.28
0.21
  PFS
8.48
1.12
0.67
7.58
0.00
 SN-38 valley concentration
  Constant
−3.28
2.42
 
−1.36
0.18
  PFS
1.84
0.36
0.51
5.08
0.00
(TA)6/(TA)7 genotype
 SN-38 peak concentration
  Constant
17.78
9.70
 
1.83
0.08
  CPT-11 starting dosage
0.08
0.03
0.45
2.86
0.01
  OS
1.78
0.64
0.44
2.80
0.01
 SN-38 valley concentration
  Constant
−5.93
6.21
 
−0.95
0.35
  Delayed diarrhea
3.64
1.05
0.47
3.46
0.00
  OS
1.56
0.43
0.49
3.60
0.00
  Bilirubin level before treatment
−0.70
0.25
−0.38
−2.82
0.01

Analysis of SN-38 peak, valley concentration correlation with efficacy and adverse effects of the subgroups in (TA)6/(TA)6 and (TA)6/(TA)7 genotypes

According to the corrected predictive values and standard deviations of SN-38 peak and valley concentration (45.49 ± 2.29 and 8.67 ± 0.74 ng/ml; 71.58 ± 2.07 and 15.25 ± 1.58 ng/ml), the (TA)6/(TA)6 genotype was divided into subgroups for the analysis by peak and valley concentration >43.2 or ≤43.2 ng/ml and >9.41 or ≤9.41 ng/ml, respectively. The (TA)6/(TA)7 genotype was divided similarly with boundaries at >73.65 or ≤73.65 ng/ml and >16.83 or ≤16.83 ng/ml, respectively. We found that mPFS for the SN-38 peak concentration >43.2 ng/ml subgroup was significantly higher than that for the ≤43.2 ng/ml subgroup (8.0 ± 0.35 vs. 6.5 ± 0.79 months, χ 2 = 17.18, p = 0.00) only with a relatively high incidence of Grade I/II° myelosuppression in the former, but there was no significant difference in mPFS between the >9.41 and ≤9.41 ng/ml subgroups (7.5 ± 0.25 vs. 7.0 ± 0.49 months, χ 2 = 0.75, p = 0.39) for the (TA)6/(TA)6 genotype; no significant difference was found in mOS between SN-38 peak concentration >73.65, ≤73.65 ng/ml subgroups and valley concentration >16.83 ng/ml, ≤16.83 subgroups (18.6 ± 1.30 vs. 16.3 ± 0.51 months, χ 2 = 2.83, p = 0.09; 17.3 ± 0.45 vs. 18.8 ± 0.50 months, χ 2 = 1.38, p = 0.24), but the >16.83 ng/ml subgroup had a higher incidence of Grade III/IV° mucositis and delayed diarrhea than ≤16.83 subgroup (F = 5.58, p = 0.03; F = 19.60, p = 0.00, see Fig. 3; Table 3) for the (TA)6/(TA)7 genotype.
Table 3
Adverse effects of each subgroup with different SN-38 peak concentration and valley concentration for (TA)6/(TA)6 and (TA)6/(TA)7 genotype
Adverse effects in (TA)6/(TA)6 genotype
SN-38 peak concentration (ng/ml)
F
p
SN-38 trough concentration (ng/ml)
F
p
>43.2 (n = 41)
≤43.2 (n = 33)
>9.41 (n = 28)
≤9.41 (n = 46)
A (%)
I/II°
28/41 (68.29)
23/33 (69.70)
1.34
0.72
19/28 (67.86)
32/46 (69.57)
1.30
0.72
III/IV°
0/41 (0)
1/33 (3.03)
0/28 (0)
1/46 (2.17)
B (%)
I/II°
7/41 (17.07)
4/33 (12.12)
5.63
0.02
4/28 (14.29)
7/46 (15.22)
3.44
0.07
III/IV°
0/41 (0)
0/33 (0)
0/28 (0)
0/46 (0)
C (%)
I/II°
26/41 (63.41)
24/33 (72.73)
1.16
0.29
18/28 (64.29)
32/46 (69.57)
0.12
0.73
III/IV°
1/41 (2.44)
0/33 (0)
0/28 (0)
1/46 (2.17)
D (%)
I/II°
11/41 (26.83)
16/33 (48.49)
0.06
0.82
6/28 (21.43)
21/46 (45.65)
0.59
0.45
III/IV°
0/41 (0)
0/33 (0)
0/28 (0)
0/46 (0)
E (%)
I/II°
4/41 (9.76)
1/33 (3.03)
1.30
0.26
2/28 (7.14)
7/46 (15.22)
0.01
0.92
III/IV°
0/41 (0)
0/33 (0)
0/28 (0)
0/46 (0)
Adverse effects in (TA)6/(TA)7 genotype
SN-38 peak concentration (ng/ml)
F
p
SN-38 trough concentration (ng/ml)
F
p
>73.65 (n = 9)
≤73.65 (n = 15)
>16.83 (n = 8)
≤16.83 (n = 16)
A (%)
I/II°
3/9 (33.33)
9/15 (60.0)
0.28
0.60
2/8 (25.0)
10/16 (62.50)
3.79
0.06
III/IV°
5/9 (55.56)
5/15 (33.33)
6/8 (75.0)
4/16 (25.0)
B (%)
I/II°
2/9 (22.22)
5/15 (33.33)
0.31
0.58
5/8 (62.50)
2/16 (12.50)
5.58
0.03
III/IV°
0/9 (0)
0/15 (0)
0/8 (0)
0/16 (0)
C (%)
I/II°
3/9 (33.33)
9/15 (60.0)
1.24
0.28
5/8 (62.50)
7/16 (439.75)
1.24
0.28
III/IV°
0/9 (0)
0/15 (0)
0/8 (0)
0/16 (0)
D (%)
I/II°
7/9 (77.78)
10/15 (66.67)
1.05
0.32
5/8 (62.50)
11/16 (68.75)
19.60
0.00
III/IV°
1/9 (11.11)
2/15 (13.33)
3/8 (37.50)
0/16 (0)
E (%)
I/II°
0/9 (0)
4/15 (26.67)
3.0
0.10
1/8 (12.50)
3/16 (18.75)
0.30
0.59
III/IV°
0/9 (0)
0/15 (0)
0/8 (0)
0/16 (0)
A: bone marrow suppression; B: mucositis; C: nausea and vomiting; D: delayed diarrhea; E: liver function injury

SN-38 peak, valley concentration, plasma bilirubin, and adverse effects for (TA)7/(TA)7 genotype

SN-38 average peak and valley concentration after CPT-11 infusion were 113.4 and 33.18 ng/ml, and the average plasma bilirubin levels before and after treatment were 20.9 and 24.1 μmol/L. Although 25 % dose reduction of CPT-11, which is calculated according to the body surface area, Grade IV° bone marrow suppression and Grade III° delayed diarrhea still occurred after CPT-11 treatment in 2 (TA)7/(TA)7 cases, both adverse effects resolved and did not recur after a 50 % dose reduction.

Discussion

Clinical administration dosage is generally calculated according to the body surface area or weight presently (Reilly and Workman 1993), which is the group average dose, but in fact, only a part of drugs with low toxicity and conventional administration, which doses calculated as above, may get satisfactory effects. The plasma concentration may be affected by drug absorption, distribution, metabolism, and excretion, even small changes in plasma concentration may cause efficacy differences and lead to serious adverse effects. Anti-tumor therapy is entering the era of individualized treatment, through the use of biological markers, drug metabolism genes, or pharmacokinetics detection, etc. The purpose of individualized treatment for advanced colorectal cancer is to improve efficacy and/or to avoid and reduce the incidence of adverse effects (Gamelin et al. 2008; Ychou et al. 2003; Fety et al. 1998; Kleibl et al. 2009; Van Kuilenburg et al. 2002; Chang et al. 2009; Liu et al. 2011). CPT-11 is metabolized to its active product SN-38 by hCES in vivo and mainly inactivated to SN-38G by UGT1A1. It is generally believed that SN-38 determines activity and toxicity; however, most studies have not found a correlation between single-nucleotide polymorphisms and enzyme activity in UGT1A1 (Wu et al. 2004; Charasson et al. 2004), However, the UGT1A1 gene polymorphism can cause decline or absence of enzyme activity. A number of studies have indicated that the UGT1A1*28 homozygous genotype [(TA)7/(TA)7 genotype] may lead to weakening of SN-38 glucuronidation, which is an important risk factor for CPT-11-associated adverse effects such as delayed diarrhea and myelosuppression (Marcuello et al. 2004; Iyer et al. 2002; Paoluzzi et al. 2004). Our results showed that (TA)6/(TA)6 and (TA)6/(TA)7 genotypes accounted for 69.51 and 29.27 % of patients, respectively, and the (TA)7/(TA)7 genotype only for 1.22 % of the 164 enrolled patients in Shanghai, and that genotype had no association with age, gender, ECOG performance score, primary tumor site, TMN staging, or chemotherapy background, which was similar to other domestic studies (Yang et al. 2009; Zhang et al. 2007), but the proportion was lower for (TA)6/(TA)6 and (TA)6/(TA)7 genotypes than foreign studies (Iyer et al. 2002; Côté et al. 2007; Toffoli et al. 2006; Braun et al. 2008); it is also explained that CPT-11-related adverse effects are lower in Chinese than in foreigners.
Although the UGT1A1*28 heterozygous genotype [(TA)6/(TA)7 genotype] can lead to decrease in UGT1A1 activity, gene polymorphism analysis alone cannot be used to direct individualized treatment of CPT-11 because changes also occur in enzyme activity for this genotype with individual difference. Pharmacokinetic studies have shown that (TA)7/(TA)7 genotype has higher AUCSN-38 than (TA)6/(TA)6 and (TA)6/(TA)7 genotypes (Iyer et al. 2002), with CPT-11 reaching peak concentration 1.5 h after infusion, terminal half-life of 10.8 h, and fall to valley level of about 30 ng/ml 25.5 h roughly after infusion (Sumiyoshi et al. 1995); in addition, 5-fluorouracil dose not change CPT-11 metabolism in vivo in a certain range of dosage (Ducreux et al. 1999; Saltz et al. 1996). These results can help to give guidance on CPT-11 individualized treatment by plasma concentration detection. Despite (TA)6/(TA)6 and (TA)6/(TA)7 genotypes accounting for most cases, there are no detailed reports before about the differences in terms of efficacy and adverse effects after first-line treatment with CPT-11. Our study found that the average peak and valley concentrations of SN-38 in (TA)6/(TA)6 genotype were higher than those in (TA)6/(TA)7 genotype after CPT-11 infusion; although average plasma bilirubin was at normal levels in both genotypes, the levels in (TA)6/(TA)7 genotype are also higher than that in (TA)6/(TA)6 genotype before and after treatment with significant difference, which was consistent with the findings of Rouits et al. (2008). The cause for this phenomenon is that bilirubin and SN-38 are both substrates of UGT1A1, and abnormality in bilirubin glucuronidation leaves, bilirubin level elevated when UGT1A1 activity decreases, while CPT-11 metabolism may also be abnormal because of the substrate competition, which leads to elevation of SN-38 concentration.
Stepwise regression analysis showed that SN-38 average peak and valley concentration were associated with PFS in the (TA)6/(TA)6 genotype; however, SN-38 peak concentration was associated with CPT-11 starting dosage and OS, and valley concentration was associated with plasma bilirubin levels before CPT-11 treatment, delayed diarrhea, and OS in (TA)6/(TA)7 genotype. So we divided the two genotypes into four subgroups according to the upper and lower limit of the corrected predictive values and standard deviations for SN-38 average peak and valley concentrations, in order to analyze the relationship between plasma concentration and efficacy or adverse effects in the subgroups. The results showed that the mPFS of the SN-38 peak concentration >43.2 ng/ml subgroup was significantly higher than that of ≤43.2 ng/ml subgroup with a relatively high incidence of Grade I/II° myelosuppression in the (TA)6/(TA)6 genotype, while there was no significant difference in mOS between SN-38 valley concentration >16.83 ng/ml and ≤16.83 subgroups, but the former had a higher incidence of Grade III/IV° mucositis and delayed diarrhea in (TA)6/(TA)7 genotype. This suggested that CPT-11 dosage can be gradually increased to improve treatment efficacy in the SN-38 peak concentration ≤43.2 ng/ml subgroup after CPT-11 infusion without serious adverse effects for patients with the (TA)6/(TA)6 genotype (in our study, 55.41 %(41/74) of (TA)6/(TA)7 genotype with SN-38 peak concentration ≤43.2 ng/ml, which means more than half of the patients require dose adjustment), while CPT-11 dosage may be appropriately lowered to reduce the incidence of serious adverse effects without affecting the efficacy in the SN-38 valley concentration >16.83 ng/ml subgroup for the (TA)6/(TA)7 genotype. SN-38 peak and valley concentration and plasma bilirubin level in the (TA)7/(TA)7 genotype were significantly higher than that in the (TA)6/(TA)7 genotype, while the incidence of serious adverse effects was consistent with other studies (Marcuello et al. 2004; Iyer et al. 2002a, b; Paoluzzi et al. 2004).

Conclusions

In summary, our study suggests that the UGT1A1*28 (TA)6/(TA)6 and (TA)6/(TA)7 genotypes account for most patients, while the homozygous mutant genotype only accounts for a very small portion of patients with advanced colorectal cancer in Shanghai. For the (TA)6/(TA)6 and (TA)6/(TA)7 genotypes, UGT1A1*28 gene polymorphism in combination with SN-38 pharmacokinetics analysis provides a good theoretical basis for CPT-11 individualized treatment; however, it should be verified by further expanding sample sizes and repeated determination of SN-38 plasma concentration after dosage adjustment, in order to improve efficacy and meanwhile avoid or reduce serious adverse effects. For the (TA)7/(TA)7 genotype, despite the reduction in initial treatment dose, further adjustment of CPT-11 dosage still should be done according to adverse effects after treatment.

Acknowledgments

The author thanks the phase I clinical research at Shanghai First People’s Hospital, Shanghai Jiaotong University, for pharmacokinetics works and Shanghai Yuanqi Biomedical Science and Technology Co. for gene polymorphism testing works.

Conflict of interest

None.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution License which permits any use, distribution, and reproduction in any medium, provided the original author(s) and the source are credited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
Zurück zum Zitat Braun MS, Richman SD, Quirke P, Daly C, Adlard JW, Elliott F, Barrett JH et al (2008) Predictive biomarkers of chemotherapy efficacy in colorectal cancer: results from the UK MRC FOCUS trial. J Clin Oncol 26(16):2690–2698PubMedCrossRef Braun MS, Richman SD, Quirke P, Daly C, Adlard JW, Elliott F, Barrett JH et al (2008) Predictive biomarkers of chemotherapy efficacy in colorectal cancer: results from the UK MRC FOCUS trial. J Clin Oncol 26(16):2690–2698PubMedCrossRef
Zurück zum Zitat Canal P, Gay C, Dezeuze A, Douillard JY, Bugat R, Brunet R, Adenis A, Herait P, Lokiec F, Mathieu-Boue A (1996) Pharmacokinetics and pharmacodynamics of irinotecan during a phase II clinical trial in colorectal cancer. Pharmacology and Molecular Mechanisms Group of the European Organization for Research and Treatment of Cancer. J Clin Oncol 14(10):2688–2695PubMed Canal P, Gay C, Dezeuze A, Douillard JY, Bugat R, Brunet R, Adenis A, Herait P, Lokiec F, Mathieu-Boue A (1996) Pharmacokinetics and pharmacodynamics of irinotecan during a phase II clinical trial in colorectal cancer. Pharmacology and Molecular Mechanisms Group of the European Organization for Research and Treatment of Cancer. J Clin Oncol 14(10):2688–2695PubMed
Zurück zum Zitat Chang DZ, Kumar V, Ma Y, Li K, Kopetz S (2009) Individualized therapies in colorectal cancer: KRAS as a marker for response to EGFR-targeted therapy. J Hematol Oncol 2:18PubMedCrossRef Chang DZ, Kumar V, Ma Y, Li K, Kopetz S (2009) Individualized therapies in colorectal cancer: KRAS as a marker for response to EGFR-targeted therapy. J Hematol Oncol 2:18PubMedCrossRef
Zurück zum Zitat Charasson V, Bellott R, Meynard D, Longy M, Gorry P, Robert J (2004) Pharmacogenetics of human carboxylesterase 2, an enzyme involved in the activation of irinotecan into SN-38. Clin Pharmacol Ther 76(6):528–535PubMedCrossRef Charasson V, Bellott R, Meynard D, Longy M, Gorry P, Robert J (2004) Pharmacogenetics of human carboxylesterase 2, an enzyme involved in the activation of irinotecan into SN-38. Clin Pharmacol Ther 76(6):528–535PubMedCrossRef
Zurück zum Zitat Côté JF, Kirzin S, Kramar A, Mosnier JF, Diebold MD, Soubeyran I, Thirouard AS, Selves J, Laurent-Puig P, Ychou M (2007) UGT1A1 polymorphism can predict hematologic toxicity in patients treated with irinotecan. Clin Cancer Res 13(11):3269–3275PubMedCrossRef Côté JF, Kirzin S, Kramar A, Mosnier JF, Diebold MD, Soubeyran I, Thirouard AS, Selves J, Laurent-Puig P, Ychou M (2007) UGT1A1 polymorphism can predict hematologic toxicity in patients treated with irinotecan. Clin Cancer Res 13(11):3269–3275PubMedCrossRef
Zurück zum Zitat Douillard JY, Cunningham D, Roth AD, Navarro M, James RD, Karasek P, Jandik P, Iveson T, Carmichael J, Alakl M, Gruia G, Awad L, Rougier P (2000) Irinotecan combined with fluorouracil compared withfluorouracil alone as first-line treatment for metastaticcolorectal cancer: a muhicentre randomised trial. Lancet 355(9209):1041–1047PubMedCrossRef Douillard JY, Cunningham D, Roth AD, Navarro M, James RD, Karasek P, Jandik P, Iveson T, Carmichael J, Alakl M, Gruia G, Awad L, Rougier P (2000) Irinotecan combined with fluorouracil compared withfluorouracil alone as first-line treatment for metastaticcolorectal cancer: a muhicentre randomised trial. Lancet 355(9209):1041–1047PubMedCrossRef
Zurück zum Zitat Ducreux M, Ychou M, Seitz JF, Bonnay M, Bexon A, Armand JP, Mahjoubi M, Méry-Mignard D, Rougier P (1999) Irinotecan combined with bolus fluorouracil, continuous infusion fluorouracil, and high-dose leucovorin every two weeks (LV5FU2 regimen): a clinical dose-finding and pharmacokinetic study in patients with pretreated metastatic colorectal cancer. J Clin Oncol 17(9):2901–2908PubMed Ducreux M, Ychou M, Seitz JF, Bonnay M, Bexon A, Armand JP, Mahjoubi M, Méry-Mignard D, Rougier P (1999) Irinotecan combined with bolus fluorouracil, continuous infusion fluorouracil, and high-dose leucovorin every two weeks (LV5FU2 regimen): a clinical dose-finding and pharmacokinetic study in patients with pretreated metastatic colorectal cancer. J Clin Oncol 17(9):2901–2908PubMed
Zurück zum Zitat Fety R, Rolland F, Barberi-Heyob M, Hardouin A, Campion L, Conroy T, Merlin JL, Rivière A, Perrocheau G, Etienne MC, Milano G (1998) Clinical impact of pharmacokinetically-guided dose adaptation of 5-fluorouracil: results from a multicentric randomized trial in patients with locally advanced head and neck carcinomas. Clin Cancer Res 4(9):2039–2045PubMed Fety R, Rolland F, Barberi-Heyob M, Hardouin A, Campion L, Conroy T, Merlin JL, Rivière A, Perrocheau G, Etienne MC, Milano G (1998) Clinical impact of pharmacokinetically-guided dose adaptation of 5-fluorouracil: results from a multicentric randomized trial in patients with locally advanced head and neck carcinomas. Clin Cancer Res 4(9):2039–2045PubMed
Zurück zum Zitat Gamelin E, Delva R, Jacob J, Merrouche Y, Raoul JL, Pezet D, Dorval E, Piot G, Morel A, Boisdron-Celle M (2008) Individual fluorouracil dose adjustment based on pharmacokinetic follow-up compared with conventional dosage: results of a multicenter randomized trial of patients with metastatic colorectal cancer. J Clin Oncol 26(13):2099–2105PubMedCrossRef Gamelin E, Delva R, Jacob J, Merrouche Y, Raoul JL, Pezet D, Dorval E, Piot G, Morel A, Boisdron-Celle M (2008) Individual fluorouracil dose adjustment based on pharmacokinetic follow-up compared with conventional dosage: results of a multicenter randomized trial of patients with metastatic colorectal cancer. J Clin Oncol 26(13):2099–2105PubMedCrossRef
Zurück zum Zitat Hirose K, Kozu C, Yamashita K, Maruo E, Kitamura M, Hasegawa J, Omoda K, Murakami T, Maeda Y (2012) Correlation between plasma concentration ratios of SN-38 glucuronide and SN-38 and neutropenia induction in patients with colorectal cancer and wild-type UGT1A1 gene. Oncol Lett 3(3):694–698PubMed Hirose K, Kozu C, Yamashita K, Maruo E, Kitamura M, Hasegawa J, Omoda K, Murakami T, Maeda Y (2012) Correlation between plasma concentration ratios of SN-38 glucuronide and SN-38 and neutropenia induction in patients with colorectal cancer and wild-type UGT1A1 gene. Oncol Lett 3(3):694–698PubMed
Zurück zum Zitat Innocenti F, Undevia SD, Iyer L, Chen PX, Das S, Kocherginsky M, Karrison T, Janisch L, Ramírez J, Rudin CM, Vokes EE, Ratain MJ (2004) Genetic variants in the UDP-glucuronosyltransferase 1A1 gene predict the risk of severe neutropenia of irinotecan. J Clin Oncol 22(8):1382–1388PubMedCrossRef Innocenti F, Undevia SD, Iyer L, Chen PX, Das S, Kocherginsky M, Karrison T, Janisch L, Ramírez J, Rudin CM, Vokes EE, Ratain MJ (2004) Genetic variants in the UDP-glucuronosyltransferase 1A1 gene predict the risk of severe neutropenia of irinotecan. J Clin Oncol 22(8):1382–1388PubMedCrossRef
Zurück zum Zitat Iyer L, Das S, Janisch L, Wen M, Ramírez J, Karrison T, Fleming GF, Vokes EE, Schilsky RL, Ratain MJ (2002a) UGT1A1* 28 polymorphism as a determinant of irinotecan disposition and toxicity. Pharmacogenomics J 2(1):43–47PubMedCrossRef Iyer L, Das S, Janisch L, Wen M, Ramírez J, Karrison T, Fleming GF, Vokes EE, Schilsky RL, Ratain MJ (2002a) UGT1A1* 28 polymorphism as a determinant of irinotecan disposition and toxicity. Pharmacogenomics J 2(1):43–47PubMedCrossRef
Zurück zum Zitat Iyer L, Das S, Janisch L, Wen M, Ramírez J, Karrison T, Fleming GF, Vokes EE, Schilsky RL, Ratain MJ (2002b) UGT1A1*28 polymorphism as a determinant of irinotecan disposition and toxicity. Pharmacogenomics J 2:43–47PubMedCrossRef Iyer L, Das S, Janisch L, Wen M, Ramírez J, Karrison T, Fleming GF, Vokes EE, Schilsky RL, Ratain MJ (2002b) UGT1A1*28 polymorphism as a determinant of irinotecan disposition and toxicity. Pharmacogenomics J 2:43–47PubMedCrossRef
Zurück zum Zitat Kaniwa N, Kurose K, Jinno H, Tanaka-Kagawa T, Saito Y, Saeki M, Sawada J, Tohkin M, Hasegawa R (2005) Racial variability in haplotype frequencies of UGT1A1 and glucuronidation activity of a novel single nucleotide polymorphism 686C > T (P229L) found in an African-American. Drug Metab Dispos 33(3):458–465PubMedCrossRef Kaniwa N, Kurose K, Jinno H, Tanaka-Kagawa T, Saito Y, Saeki M, Sawada J, Tohkin M, Hasegawa R (2005) Racial variability in haplotype frequencies of UGT1A1 and glucuronidation activity of a novel single nucleotide polymorphism 686C > T (P229L) found in an African-American. Drug Metab Dispos 33(3):458–465PubMedCrossRef
Zurück zum Zitat Kleibl Z, Fidlerova J, Kleiblova P, Kormunda S, Bilek M, Bouskova K, Sevcik J, Novotny J (2009) Influence of dihydropyrimidine dehydrogenase gene (DPYD) coding sequence variants on the development of fluoropyrimidine-related toxicity in patients with high-grade toxicity and patients with excellent tolerance of fluoropyrimidine-based chemotherapy. Neoplasma 56(4):303–316PubMedCrossRef Kleibl Z, Fidlerova J, Kleiblova P, Kormunda S, Bilek M, Bouskova K, Sevcik J, Novotny J (2009) Influence of dihydropyrimidine dehydrogenase gene (DPYD) coding sequence variants on the development of fluoropyrimidine-related toxicity in patients with high-grade toxicity and patients with excellent tolerance of fluoropyrimidine-based chemotherapy. Neoplasma 56(4):303–316PubMedCrossRef
Zurück zum Zitat Liu CY, Chen PM, Chiou TJ, Liu JH, Lin JK, Lin TC, Chen WS, Jiang JK, Wang HS, Wang WS (2008) UGT1A1*28 polymorphism predicts irinotecan-induced severe toxicities without affecting treatment outcome and survival in patients with metastatic colorectal carcinoma. Cancer 112(9):1932–1940PubMedCrossRef Liu CY, Chen PM, Chiou TJ, Liu JH, Lin JK, Lin TC, Chen WS, Jiang JK, Wang HS, Wang WS (2008) UGT1A1*28 polymorphism predicts irinotecan-induced severe toxicities without affecting treatment outcome and survival in patients with metastatic colorectal carcinoma. Cancer 112(9):1932–1940PubMedCrossRef
Zurück zum Zitat Liu Y, Xia Q, Jia Y, Guo H, Wei B, Hua Y, Yang S (2011) Significance of differential expression of thymidylate synthase in normal and primary tumor tissues from patients with colorectal cancer. J Hematol Oncol 4:33PubMedCrossRef Liu Y, Xia Q, Jia Y, Guo H, Wei B, Hua Y, Yang S (2011) Significance of differential expression of thymidylate synthase in normal and primary tumor tissues from patients with colorectal cancer. J Hematol Oncol 4:33PubMedCrossRef
Zurück zum Zitat Marcuello E, Altés A, Menoyo A, Del Rio E, Gómez-Pardo M, Baiget M (2004) UGT1A1 gene variations and irinotecan treatment in patients with metastatic colorectal cancer. Br J Cancer 91(4):678–682PubMed Marcuello E, Altés A, Menoyo A, Del Rio E, Gómez-Pardo M, Baiget M (2004) UGT1A1 gene variations and irinotecan treatment in patients with metastatic colorectal cancer. Br J Cancer 91(4):678–682PubMed
Zurück zum Zitat Massacesi C, Terrazzino S, Marcucci F, Rocchi MB, Lippe P, Bisonni R, Lombardo M, Pilone A, Mattioli R, Leon A (2006) Uridine diphosphate glucuronosyl transferase 1 A 1 promoter polymorphism predicts the risk of gastrointestinal toxicity and fatigue induced by irinotecan-based chemotherapy. Cancer 106(5):1007–1016PubMedCrossRef Massacesi C, Terrazzino S, Marcucci F, Rocchi MB, Lippe P, Bisonni R, Lombardo M, Pilone A, Mattioli R, Leon A (2006) Uridine diphosphate glucuronosyl transferase 1 A 1 promoter polymorphism predicts the risk of gastrointestinal toxicity and fatigue induced by irinotecan-based chemotherapy. Cancer 106(5):1007–1016PubMedCrossRef
Zurück zum Zitat Paoluzzi L, Singh AS, Price DK, Danesi R, Mathijssen RH, Verweij J, Figg WD, Sparreboom A (2004) Influence of genetic variants in UGT1A1 and UGT1A9 on the in vivo glucuronidation of SN-38. J Clin Pharmacol 44(8):854–860PubMedCrossRef Paoluzzi L, Singh AS, Price DK, Danesi R, Mathijssen RH, Verweij J, Figg WD, Sparreboom A (2004) Influence of genetic variants in UGT1A1 and UGT1A9 on the in vivo glucuronidation of SN-38. J Clin Pharmacol 44(8):854–860PubMedCrossRef
Zurück zum Zitat Reilly JJ, Workman P (1993) Normalization of anti-cancer drug dosage using body weight and surface area: is it worthwhile? A review of theoretical and practical considerations. Cancer Chemother Pharmacol 32(6):411–418PubMedCrossRef Reilly JJ, Workman P (1993) Normalization of anti-cancer drug dosage using body weight and surface area: is it worthwhile? A review of theoretical and practical considerations. Cancer Chemother Pharmacol 32(6):411–418PubMedCrossRef
Zurück zum Zitat Rouits E, Charasson V, Pétain A, Boisdron-Celle M, Delord JP, Fonck M, Laurand A, Poirier AL, Morel A, Chatelut E, Robert J, Gamelin E (2008) Pharmacokinetic and pharmacogenetic determinants of the activity and toxicity of irinotecan in metastatic colorectal cancer patients. Br J Cancer 99(8):1239–1245PubMedCrossRef Rouits E, Charasson V, Pétain A, Boisdron-Celle M, Delord JP, Fonck M, Laurand A, Poirier AL, Morel A, Chatelut E, Robert J, Gamelin E (2008) Pharmacokinetic and pharmacogenetic determinants of the activity and toxicity of irinotecan in metastatic colorectal cancer patients. Br J Cancer 99(8):1239–1245PubMedCrossRef
Zurück zum Zitat Saltz LB, Kanowitz J, Kemeny NE, Schaaf L, Spriggs D, Staton BA, Berkery R, Steger C, Eng M, Dietz A, Locker P, Kelsen DP (1996) Phase I clinical and pharmacokinetic study of irinotecan, fluorouracil, and leucovorin in patients with advanced solid tumors. J Clin Oncol 14(11):2959–2967PubMed Saltz LB, Kanowitz J, Kemeny NE, Schaaf L, Spriggs D, Staton BA, Berkery R, Steger C, Eng M, Dietz A, Locker P, Kelsen DP (1996) Phase I clinical and pharmacokinetic study of irinotecan, fluorouracil, and leucovorin in patients with advanced solid tumors. J Clin Oncol 14(11):2959–2967PubMed
Zurück zum Zitat Schoemaker NE, Rosing H, Jansen S, Schellens JH, Beijnen JH (2003) High-performance liquid chromatographic analysis of the anticancer drug irinotecan (CPT-11) and its active metabolite SN-38 in human plasma. Ther Drug Monit 25(1):120–124PubMedCrossRef Schoemaker NE, Rosing H, Jansen S, Schellens JH, Beijnen JH (2003) High-performance liquid chromatographic analysis of the anticancer drug irinotecan (CPT-11) and its active metabolite SN-38 in human plasma. Ther Drug Monit 25(1):120–124PubMedCrossRef
Zurück zum Zitat Shulman K, Cohen I, Barnett-Griness O, Kuten A, Gruber SB, Lejbkowicz F, Rennert G (2011) Clinical implications of UGT1A1*28 genotype testing in colorectal cancer patients. Cancer 117(14):3156–3162PubMedCrossRef Shulman K, Cohen I, Barnett-Griness O, Kuten A, Gruber SB, Lejbkowicz F, Rennert G (2011) Clinical implications of UGT1A1*28 genotype testing in colorectal cancer patients. Cancer 117(14):3156–3162PubMedCrossRef
Zurück zum Zitat Sumiyoshi H, Fujiwara Y, Ohune T, Yamaoka N, Tamura K, Yamakido M (1995) High performance liquid chromatographic determination of irinotecan (CPT-11) and its active metabolite (SN-38) in human plasma. J Chromatogr B Biomed Appl 670(2):309–316PubMedCrossRef Sumiyoshi H, Fujiwara Y, Ohune T, Yamaoka N, Tamura K, Yamakido M (1995) High performance liquid chromatographic determination of irinotecan (CPT-11) and its active metabolite (SN-38) in human plasma. J Chromatogr B Biomed Appl 670(2):309–316PubMedCrossRef
Zurück zum Zitat Toffoli G, Cecchin E, Corona G, Russo A, Buonadonna A, D’Andrea M, Pasetto LM et al (2006) The role of UGT1A1*28 polymorphism in the pharmacodynamics and pharmacokinetics of irinotecan in patients with metastatic colorectal cancer. J Clin Oncol 24(19):3061–3068PubMedCrossRef Toffoli G, Cecchin E, Corona G, Russo A, Buonadonna A, D’Andrea M, Pasetto LM et al (2006) The role of UGT1A1*28 polymorphism in the pharmacodynamics and pharmacokinetics of irinotecan in patients with metastatic colorectal cancer. J Clin Oncol 24(19):3061–3068PubMedCrossRef
Zurück zum Zitat Van Kuilenburg AB, Meinsma R, Zoetekouw L, Van Gennip AH (2002) Increased risk of grade IV neutropenia after administration of 5-fluorouracil due to a dihydropyrimidine dehydrogenase deficiency: high prevalence of the IVS14+1G> a mutation. Int J Cancer 101(3):253–258PubMedCrossRef Van Kuilenburg AB, Meinsma R, Zoetekouw L, Van Gennip AH (2002) Increased risk of grade IV neutropenia after administration of 5-fluorouracil due to a dihydropyrimidine dehydrogenase deficiency: high prevalence of the IVS14+1G> a mutation. Int J Cancer 101(3):253–258PubMedCrossRef
Zurück zum Zitat Wu MH, Chen P, Wu X, Liu W, Strom S, Das S, Cook EH Jr, Rosner GL, Dolan ME (2004) Determination and analysis of single nucleotide polymorphisms and haplotype structure of the human carboxylesterase 2 gene. Pharmacogenetics 14(9):595–605PubMedCrossRef Wu MH, Chen P, Wu X, Liu W, Strom S, Das S, Cook EH Jr, Rosner GL, Dolan ME (2004) Determination and analysis of single nucleotide polymorphisms and haplotype structure of the human carboxylesterase 2 gene. Pharmacogenetics 14(9):595–605PubMedCrossRef
Zurück zum Zitat Yang LX, Ma T, Zhang J, Ye ZB, Xiang M, Zhu ZG (2009) A study on the relationship between the adverse events of irinotecan-based chemotherapy and UGT1A1*28 gene polymorphism. J Intern Med Concepts. Pract. (Chinese) 4(4):300–304 Yang LX, Ma T, Zhang J, Ye ZB, Xiang M, Zhu ZG (2009) A study on the relationship between the adverse events of irinotecan-based chemotherapy and UGT1A1*28 gene polymorphism. J Intern Med Concepts. Pract. (Chinese) 4(4):300–304
Zurück zum Zitat Ychou M, Duffour J, Kramar A, Debrigode C, Gourgou S, Bressolle F, Pinguet F (2003) Individual 5-FU dose adaptation in metastatic colorectal cancer: results of a phase II study using a bimonthly pharmacokinetically intensified LV5FU2 regimen. Cancer Chemother Pharmacol 52(4):282–290PubMedCrossRef Ychou M, Duffour J, Kramar A, Debrigode C, Gourgou S, Bressolle F, Pinguet F (2003) Individual 5-FU dose adaptation in metastatic colorectal cancer: results of a phase II study using a bimonthly pharmacokinetically intensified LV5FU2 regimen. Cancer Chemother Pharmacol 52(4):282–290PubMedCrossRef
Zurück zum Zitat Zhang A, Xing Q, Qin S, Du J, Wang L, Yu L, Li X, Xu L, Xu M, Feng G, He L (2007) Intra-ethnic differences in genetic variants of the UGT-glucuronosyltransferase 1A1 gene in Chinese populations. Pharmacogenomics J 7(5):333–338PubMedCrossRef Zhang A, Xing Q, Qin S, Du J, Wang L, Yu L, Li X, Xu L, Xu M, Feng G, He L (2007) Intra-ethnic differences in genetic variants of the UGT-glucuronosyltransferase 1A1 gene in Chinese populations. Pharmacogenomics J 7(5):333–338PubMedCrossRef
Metadaten
Titel
Analysis of UGT1A1*28 genotype and SN-38 pharmacokinetics for irinotecan-based chemotherapy in patients with advanced colorectal cancer: results from a multicenter, retrospective study in Shanghai
verfasst von
Xun Cai
Weiguo Cao
Honghua Ding
Tianshu Liu
Xinli Zhou
Mei Wang
Ming Zhong
Ziyi Zhao
Qing Xu
Liwei Wang
Publikationsdatum
01.09.2013
Verlag
Springer Berlin Heidelberg
Erschienen in
Journal of Cancer Research and Clinical Oncology / Ausgabe 9/2013
Print ISSN: 0171-5216
Elektronische ISSN: 1432-1335
DOI
https://doi.org/10.1007/s00432-013-1480-7

Weitere Artikel der Ausgabe 9/2013

Journal of Cancer Research and Clinical Oncology 9/2013 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.