ArticlePDF Available

Genetic Polymorphism of GSTP1: Prediction of Clinical Outcome to Oxaliplatin/5-FU-based Chemotherapy in Advanced Gastric Cancer

Authors:

Abstract and Figures

The aim of this study was to evaluate the predictive value of the polymorphism Glutathione S-transferase P1 (GSTP1) Ile(105)Val on oxaliplatin/5-FU-based chemotherapy in advanced gastric cancer. Patients with advanced gastric cancer accepted oxaliplatin/5-FU-based chemotherapy as first-line chemotherapy were investigated. GSTP1 Ile(105)Val polymorphism was detected by TaqMan-MGB probe allelic discrimination method. Response to treatment was assessed by disease controlled rate. Time to progression, overall survival and toxicities were recorded. Final patient outcomes were as follows: the allele frequencies of GSTP1 were (105)Ile/(105)Ile 52%, (105)Ile/(105)Val 41% and (105)Val/(105)Val 7%. For patients with (105)Ile/(105)Ile and those with at least one (105)Val allele, disease control rate was 39% and 71% (P=0.026), respectively; median time to progression was 4.0 and 7.0 months (P=0.002); median overall survival time was 7.0 and 9.5 months (P=0.002). Neurological toxicity was more frequently occurred in patients with two (105)Ile alleles (P=0.005). In conclusion, patients with at least one (105)Val allele have better prognosis and response to oxaliplatin/5-FU-based regimen as first-line treatment for patients with advanced gastric cancer.
Content may be subject to copyright.
INTRODUCTION
Gastric cancer is one of the most common adult malignant
tumors. Gastric cancer is the fourth common type of cancer
and the second most frequent cause of death from cancer,
worldwide, with the highest incidence in China, Japan and
Eastern European countries (1). Cisplatin is commonly used
with 5-fluorouracil (5-FU) as chemotherapy doublets in the
treatment of advanced gastric cancer (2). During the last few
years additional drugs were introduced into chemotherapy
regimens for gastric cancer such as oxaliplatin, taxanes and
epirubicin. However, the response rates of these drugs or their
combinations were still less than 50% (3-5). One of the re-
maining challenges is the development of predictive marker
profiles to identify patients who will derive both minimal
toxicity and maximum benefit from certain chemotherapy.
There is a growing body of evidence suggesting that genetic
polymorphisms in genes involved in metabolism, signalling,
DNA-repair and cellular response pathways all contribute to
inter-patient variability of drug response and toxicity (6, 7).
Therefore, pharmacogenetic analyses appear to be a promis-
ing tool to develop individualized treatment plans. Individ-
ual chemotherapy based on pharmacogenetics and pharma-
cogenomics has shown a potentially predictive role to achieve
superior outcome in retrospective and perspective studies in
lung cancer (8, 9). However, little is known in gastric cancer
pharmacogenetics.
Oxaliplatin, carrying a 1,2-diamino-cyclohexane ring, leads
to platinum-DNA adducts, which are more cytotoxic than
adducts formed from other platinum agents and more effec-
tive at blocking DNA replication (10). With the wider use of
oxaliplatin in gastric cancer, predictive markers for clinical
outcome to this platinum agent may help to identify pro-
spectively those patients who are more likely to benefit from
the treatment. Resistance to platinum agents has been attri-
buted to enhanced tolerance to platinum DNA adducts, dec-
reased drug accumulation, or enhanced DNA repair (11). Vari-
able chemosensitivity to oxaliplatin may depend on detoxi-
fication pathways, including the glutathione S-transferase
(GST) family of isoenzymes (12, 13). Glutathione S-trans-
ferase P1 (GSTP1) is a member of a superfamily of dimeric
phase II metabolic enzymes that play an important role in
the cell defense system. GSTP1 directly participates in the
detoxification of platinum compounds and is an important
846
Qing-Fang Li, Ru-Yong Yao,
Ke-wei Liu, Hong-Ying Lv, Tao Jiang,
and Jun Liang
Treatment and Research Center of Oncology, The
Affiliated Hospital of Medical College of Qingdao
University, Qingdao, China
Address for Correspondence
Jun Liang, M.D.
Treatment and Research Center of Oncology, The
Affiliated Hospital of Medical College of Qingdao
University, Jiangsu Road 16, Qingdao 266003, China
Tel : +86-532-82911348, Fax : +86-532-82911999
E-mail : qfmy4812@sina.com
This work was supported in part by grants from
Shandong Province Natural Science Foundation
(Y2008C126).
J Korean Med Sci 2010; 25: 846-52 ISSN 1011-8934
DOI: 10.3346/jkms.2010.25.6.846
Genetic Polymorphism of GSTP1: Prediction of Clinical Outcome
to Oxaliplatin/5-FU-based Chemotherapy in Advanced Gastric Cancer
The aim of this study was to evaluate the predictive value of the polymorphism Glu-
tathione S-transferase P1 (GSTP1) Ile
105
Val on oxaliplatin/5-FU-based chemother-
apy in advanced gastric cancer. Patients with advanced gastric cancer accepted
oxaliplatin/5-FU-based chemotherapy as first-line chemotherapy were investigated.
GSTP1 Ile
105
Val polymorphism was detected by TaqMan-MGB probe allelic discrimi-
nation method. Response to treatment was assessed by disease controlled rate.
Time to progression, overall survival and toxicities were recorded. Final patient out-
comes were as follows: the allele frequencies of GSTP1 were
105
Ile/
105
Ile 52%,
105
Ile/
105
Val 41% and
105
Val/
105
Val 7%. For patients with
105
Ile/
105
Ile and those with at least
one
105
Val allele, disease control rate was 39% and 71% (P=0.026), respectively;
median time to progression was 4.0 and 7.0 months (P=0.002); median overall sur-
vival time was 7.0 and 9.5 months (P=0.002). Neurological toxicity was more fre-
quently occurred in patients with two
105
Ile alleles (P=0.005). In conclusion, patients
with at least one
105
Val allele have better prognosis and response to oxaliplatin/5-FU-
based regimen as first-line treatment for patients with advanced gastric cancer.
Key Words : Polymorphism; Glutathione S-Transferase pi; Oxaliplatin; Stomach Neoplasms
Received : 19 August 2009
Accepted : 23 November 2009
2010 The Korean Academy of Medical Sciences.
This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial
License (http://creativecommons.org/licenses/by-nc/3.0) which permits unrestricted non-commercial use,
distribution, and reproduction in any medium, provided the original work is properly cited.
Polymorphism of GSTP1 as a Prediction of Clinical Outcome 847
mediator of both intrinsic and acquired resistance to platinum
(14). A single nucleotide substitution (AG) at position
313 (resulting in replacing isoleucine with valine at codon
105) has been found to modify the enzyme’s activity and affin-
ity for electrophilic substrates (15). Previous studies showed
the expression level and the polymorphic Ile105Val of GSTP1
had linked with the sensitivity of cancer cells to platinum
(16). Clinical studies also have implicated GSTP1 as a pre-
dictive marker for clinical outcome in patients with cancer
treated with platinum-based chemotherapy (17, 18). How-
ever, the predictive values in advanced gastric cancer with
oxaliplatin-based chemotherapy are still not sufficient.
Given the further biochemical evidence that GSTP1 medi-
ates inactivation of platinum drugs, we analyzed the common
polymorphism in GSTP1-105 in patients with advanced gas-
tric cancer to determine whether the presence of the poly-
morphism is associated with clinical outcome to oxaliplatin/
5-FU-based first-line chemotherapy in Chinese population.
MATERIALS AND METHODS
Study subjects
From July 2006 to December 2008, 163 patients with ad-
vanced gastric cancer were treated at the Cancer Treatment
Center in the Affiliated Hospital of Medical College, Qingdao
University (China). The criteria for acceptance of the patients
were as follows: 1) all patients had bi-dimensionally measur-
able disease; 2) each participant had gastric adenocarcinoma
with stage IV disease, definitely diagnosed by histology; 3)
patients’ performance status was classified according to East-
ern Cooperative Oncology Group (ECOG) criteria, and each
patient’s ECOG status was not greater than 2; 4) patients had
not been treated with chemotherapy previously; 5) each par-
ticipant’s hemogram, hepatic function, renal function, and
electrocardiogram were normal before chemotherapy. Accord-
ing to the criteria mentioned-above, 92 patients were eligi-
ble for the study. All the 92 patients signed an informed con-
sent form before entering the study. Specimens (blood sam-
ples) of the patients were obtained before treatment.
Chemotherapy
All participants received the following oxaliplatin/5-FU-
based combination therapy regimen as first-line treatment.
All patients administered oxaliplatin plus 5-FU regimen once
every two weeks: oxaliplatin (Jiangsu Heng-rui Express, China;
H20040817) 85 mg/m2as a 2-hr infusion on day 1, and then
calcium folinate (Jiangsu Heng-rui Express) 200 mg/m2as
a 2-hr infusion on days 1-2, combined with bolus 5-FU (Tian-
jin Jin-yao Express, China; H12020959) 300 mg/m2and con-
tinuous infusion of 5-FU 600 mg/m2on days 1-2. Treatment
was continued until documented disease progression, unac-
ceptable toxic effects or patient’s refusal.
Clinical evaluation
All patients for analyses were required to have completed
at least four cycles of chemotherapy. Computed tomography
imaging was performed every 6 weeks. Objective response to
the treatment corresponded to a decrease in tumor burden
(the sum, over all measurable lesions, of the products of the
largest diameter and its perpendicular diameter) of 50% or
more for at least 6 weeks (partial response, or complete res-
ponse). And disease control includes complete response (CR),
partial response (PR) and stable disease (SD, 25% progres-
sion, <50% shrinkage) for at least 6 weeks. Progressive dis-
ease (PD) was defined as an enlargement in tumor burden of
25% or more (compared with the smallest measurement) or
appearance of new lesions. We calculated the disease control
rate (the percent of CR+PR+SD) to evaluate patients’ response
to chemotherapy. Time to progression (TTP) was calculated
from the time of initial treatment to disease progression. Over-
all survival (OS) was calculated from the time of initial treat-
ment to death or the last follow-up. Neuropathy was scored
according to the oxaliplatin-specific scale reported by Caus-
sanel et al. (19). On this oxaliplatin-specific scale, the grade
of cumulative neuropathy depends on the duration and inten-
sity of symptoms (grade 1, paresthesia, dysesthesia of short
duration; grade 2, paresthesia, dysesthesia persisting between
cycles; grade 3, paresthesia, dysesthesia causing functional
impairment; grade 4, paralysis). Other reliable adverse events
were evaluated using the National Cancer Institute-Common
Toxicity Criteria (NCI-CTC) version 2.0 (grade 1, mild adverse
event; grade 2, moderate adverse event; grade 3, severe and
undesirable adverse event; grade 4, life-threatening or dis-
abling adverse event).
DNA extraction and genotyping
Genomic DNA was isolated from 2 mL peripheral blood,
using Blood Genomic DNA Isolation Kit (Watson Biotech-
nologies, Inc., Taipei, Taiwan). The Ile105Val polymorphism
was analyzed with TaqMan genotyping assays, using the Roter-
gene real time 36-well polymerase chain reaction (PCR) sys-
tem (Gene Company Limited, Hong Kong, China). Probes,
primer and TaqMan universal PCR master mix were purchas-
ed from Applied Biosystems Inc. (ABI). The TaqMan assay
were performed in 25 mL reaction solution containing 1.25 mL
TaqMan SNP Genotyping Assay Mix (consisting of 20×mix
of unlabeled PCR primers and TaqMan MGB probe, FAM and
VIC dye-labeled), 12.5 mL PCR mixture reagent and 20 ng
genomic DNA. The polymerase chain reaction consisted of
an initial step at 95
for 10 min and 48 cycles of denatur-
ing at 92
for 15 sec and annealing 60
for 1 min. SDS
allelic discrimination software provided by Gene Company
Limited was used for analysis of the PCR genotyping results.
848 Q.-F. Li, R.-Y. Yao, K.-W. Liu, et al.
30 randomly selected DNA samples were genotyped at least
twice to confirm the results.
Statistical analysis
Initially, disease controlled rate, time to progression, over-
all survival, and reliable toxicity were the end points consid-
ered in this analysis. Patients’ characteristics were compared
across genotypes using chi-square test. GSTP1 genotype dis-
tribution gained in the study was tested by the Hardy-Wein-
berg equilibrium law. The disease controlled rate were com-
pared by the chi-square test and Fisher exact test between the
patients with 105Ile/105Ile GSTP1 genotype and those with at
least one 105Val allele. The Cox proportional hazards model
was used to determine the association of TTP and OS with
the following baseline prognostic factors: histology, site of
tumor, number of metastatic sites, performance status, age
at enrollment of study, and gender. The odds ratio (OR) and
95% confidence intervals (95% CI) were calculated at the
same time. The log-rank test and Kaplan-Meier plots were
used to evaluate the association of overall survival and time
to progression with GSTP1 polymorphism. Treatment-relat-
ed toxicity was compared by the Mann-Whitney test between
the patients with GSTP1 wild genotype and patients with
GSTP1 mutant genotype at codon 105.
All Pvalues reported here were two-sided, and a probabil-
ity of 0.05 or smaller was considered statistically significant.
Statistical analyses were performed using SPSS software, Ver-
sion 16.0 (SPSS Inc., Chicago, IL, USA).
RESULTS
GSTP1 polymorphism
Ninety-seven percent (89/92) of the patients’ genetic poly-
morphism was determined successfully. GSTP1 genotype was
analyzed for 89 patients, of which 46 (52%) were homozy-
gous for the 105Ile/105Ile GSTP1 genotype, 37 (41%) were
heterozygous for 105Ile/105Val, and 6 (7%) were homozygous
for the 105Val/105Val. The allele frequency for the GSTP1 105Val
allele in our study was 48% (43/89). The allelic distribution
was in Hardy-Weinberg equilibrium (
c
2=0.156, P=0.692).
The 89 patients consisted of 25 females (28%) and 64 males
(72%), with a median age of 55 yr old (range 32 yr old to
77 yr old). No clear patterns for significant associations bet-
ween the polymorphism and any of the other demographic
(gender), clinical (performance status, site of the tumor, and
number of metastases), or pathological characteristics (tumor
differentiation) were observed (Table 1).
Response to chemotherapy
GSTP1-105 genotype analyses were successful for 89 pa-
tients of which 85 were eligible for response analyses. Four
patients withdrew from the study too early to evaluate their
response. The four patients refused to continue chemothera-
*Performance status was classified according to Eastern Cooperative
Oncology Group (ECOG) criteria.
GSTP1, Glutathione S-transferases P1.
GSTP1, Glutathione S-transferases P1; CR, complete response; PR, partial response; SD, stable disease; PD, progressive disease, OR, odds ratio; CI,
confidence intervals.
Characteristics
P
c
2
P
c
2
Chi-square test
GSTP1-
105
Ile/Ile (n)
GSTP1-105
Ile/Val+Val/
Val (n)
Sex 0.822 0.365
Male 35 29
Female 11 14
Age 2.449 0.294
44 5 10
45-59 23 19
>60 18 14
Site of tumor 2.613 0.455
Fundus 6 2
Body 14 18
Sinus 21 19
Other 5 4
Histology 0.182 0.670
Well/moderate 10 11
Poor 36 32
Number of metastasis 0.811 0.847
02019
11315
296
343
Performance status* 0.198 0.656
13837
=2 8 6
Table 1. Demographic, clinical, and pathological characteristics
compared between patients with different GSTP1 genotype
Response to treatment
Genotype
CR+PR+SD OR (95% CI)PD
Chi-square test Logistic regression analysis
P
GSTP1-105 Ile/Val+Val/Val 29 12 1.000
GSTP1-105 Ile/Ile 17 27 9.259 0.026 5.028 (1.750-14.446) 0.003
Table 2. Association between GSTP1 polymorphism and response to treatment
Polymorphism of GSTP1 as a Prediction of Clinical Outcome 849
py. The median cycles of chemotherapy were 5 cycles for the
remaining 85 patients. Among these 85 patients, 12 (14%)
of the patients showed PR, 34 (40%) SD, 39 (46%) showed
PD, and no patients showed CR. The association between the
response to treatment and the GSTP1 polymorphisms was
shown in Table 2.
Time to progression
The 85 patients mentioned-above were eligible for survival
analysis since the four patients took off study too early to eval-
uate their survival time. From July 2006 to April 2009, 83
patients (98%) were followed up, with a median follow-up
period of 11.6 months (range 3.5 to 23 months). The survival
time of the 2 patients who lost follow-up was treated as cen-
sored data.
Among the 85 patients, eighty-seven percent (74/85) of
the participants’ disease had progression during the follow-up
period. Then the TTP of the 9 patients without disease pro-
gression and the 2 patients with lost of follow up was censor-
ed. Compared with patients with a homozygous 105Ile/105Ile
GSTP1 genotype, patients with one or two 105Val alleles had
increased the median time to progression (MTTP). The MTTP
for patients with a homozygous 105Ile/105Ile GSTP1 genotype
was 4.0 months (95% CI 3.649-4.351), whereas that for pa-
Characteristics No.
OR (95% CI)
TTP
COX regression analysis
POR (95% CI)
OS
P
Sex
Female 23 1.000 1.000
Male 62 0.953 (0.529-1.714) 0.871 1.120 (0.584-2.145) 0.733
Age (yr) 0.123 0.318
44 15 1.000 1.000
45-59 40 0.458 (0.215-0.972) 0.042 0.559 (0.257-1.215) 0.142
60 30 0.570 (0.251-1.294) 0.179 0.685 (0.290-1.621) 0.390
Site of tumor 0.457 0.362
Fundus 7 1.000 1.000
Body 31 1.374 (0.518-3.647) 0.523 1.225 (0.458-3.276) 0.686
Sinus 39 1.208 (0.476-3.070) 0.690 1.465 (0.551-3.896) 0.444
Other 8 2.271 (0.724-7.127) 0.160 2.645 (0.809-8.644) 0.108
Histology
Well/moderate 20 1.000 1.000
Poor 65 1.582 (0.892-2.804) 0.117 2.134 (1.153-3.949) 0.016
Number of metastasis 0.153 0.010
0 36 1.000 1.000
1 27 1.080 (0.616-1.894) 0.788 1.446 (0.799-2.684) 0.242
2 15 1.753 (0.864-3.559) 0.120 2.428 (1.120-5.262) 0.025
3 7 2.717 (0.976-7.613) 0.058 5.088 (1.804-14.348) 0.002
Performance status*
1 71 1.000 1.000
=2 14 2.229 (1.096-4.534) 0.027 1.611 (0.775-3.351) 0.202
GSTP1-105 genotype
Ile/Val or Val/Val 1.000 1.000
Ile/Ile 2.359 (1.362-4.087) 0.002 2.268 (1.281-4.017) 0.005
Table 3. Association between patients’ characteristics and TTP, OS after oxaliplatin/5-FU-based chemotherapy
*Performance status was classified according to Eastern Cooperative Oncology Group (ECOG) criteria.
TTP, time to progression; OS, overall survival; OR, odds ratio; CI, confidence intervals; GSTP1, Glutathione S-transferases P1.
Fig. 1. Time to progression curves of patients with advanced gas-
tric cancer according to GSTP1 Ile105Val polymorphism.
Probability of time to progression
1.0
0.8
0.6
0.4
0.2
0.0
2.50 5.00 7.50 10.00 12.50
Time to progression (months)
GSTP1-105 lle/lle
P=0.002
GSTP1-105 lle/Val+Val/Val
Group
1
2
1-censored
2-censored
1, lle/lle
2, lle/Val+Val/Val
850 Q.-F. Li, R.-Y. Yao, K.-W. Liu, et al.
tients with one or two 105Val GSTP1 genotype was 7.0 months
(95% CI 5.360-8.640;
c
2=10.043, P=0.002) (Fig. 1).
The MTTP for all 85 patients was 5.3 months (95% CI
4.431-6.169). Homozygosity for 105Ile/105Ile GSTP1 genotype
was associated with a significantly increased risk of progres-
sion compared with at least one 105Val allele (OR 2.359; 95%
CI 1.362-4.087; P=0.002). Patients with a performance sta-
tus ECOG=2 showed an increased risk of progressing (P=
0.027) when compared with patients who showed a superi-
or performance status (ECOG 0-1). Age, sex, tumor histolog-
ic grade, metastases, as well as site of tumor was not associ-
ated with TTP in this series of patients (Table 3).
Overall survival time
Of the 85 patients, 14 patients were still alive. The survival
time of the 14 patients and the two patients with lost of fol-
low up was treated as censored data. The median overall sur-
vival (MOS) for patients with a homozygous 105Ile/105Ile GS-
TP1 genotype was 7.0 months (95% CI 5.137-8.863), where-
as that for patients with one or two 105Val GSTP1 genotype
was 9.5 months (95% CI 7.847-11.153;
c
2=9.223, P=0.002)
(Fig. 2).
The MOS for all 85 patients was 8.0 months (95% CI
6.623-9.377). Patients with 105Ile/105Ile genotype had a high-
er risk of death compared with patients with one or two 105Val
alleles (OR 2.268; 95% CI 1.281-4.017; P=0.005). Age,
sex, site of tumor, and performance status were not associat-
ed with patient survival. However, patients with more metas-
tases seemed to have a shorter survival time (P=0.010). Oth-
erwise, 65 patients with a poorly differentiated tumor also
tended to have a higher risk of death (P=0.016). The associ-
ation of demographic, clinical, and pathological characteris-
tics and clinical outcome was listed in Table 3.
Toxicity
All 85 patients were evaluated toxicity from the time of
initial chemotherapy to end of follow-up. No treatment-relat-
ed deaths occurred. The most common adverse events of the
treatment were hematological and neurological toxicity. Grade
3 or 4 cumulative peripheral neuropathy was significantly
more common in patients with 105Ile/105Ile GSTP1 genotype
(10 of 44 patients, 23%) compared with patients with 105Ile/
105Val or 105Val/105Val GSTP1 genotype (2 of 41 patients, 5%;
2=5.577, P=0.018). Meanwhile, the overall incidence of neu-
rological toxicity in patients with homozygous 105Ile/105Ile
GSTP1 genotype was significantly higher than that in patients
with one or two 105Val GSTP1 genotype (P=0.005) (Table 4).
DISCUSSION
We attempted to identify predictive genetic polymorphisms
for response and survival to oxaliplatin/5-FU chemotherapy
in patients with advanced gastric cancer. Our analysis of the
common polymorphism for the GSTP1 in patients with ad-
vanced gastric cancer demonstrates that relevant alteration
of a protein’s function can result in differences of defined clin-
ical endpoints. In this study, patients with at least one 105Val
allele had a better prognosis and response to the oxaliplatin/
5-FU-based regimen as first-line treatment for advanced gas-
tric cancer.
The identification of molecular variables that predict either
Adverse events
0 I II III IV
GSTP1-105 Ile/Ile
0 I II III IV
GSTP1-105 Ile/Val+Val/Val Mann-Whit-
ney test
P
Leucocytopenia 24 9 8 3 0 18 10 5 6 2 0.229
Neutrocytopenia 23 8 10 3 0 19 8 4 7 2 0.323
Thrombocytopenia 37 4 2 1 0 36 3 1 0 1 0.626
Erythrocytopenia 35 3 4 2 0 34 4 2 1 0 0.624
Nausea 14 15 12 3 0 18 10 11 2 0 0.399
Vomiting 22 11 9 2 0 21 11 8 1 0 0.807
Neurological toxicity 11 14 9 10 0 19 15 5 2 0 0.005
Table 4. Association between GSTP1 polymorphism and treatment-related toxicity
Fig. 2. Overall survival curves of patients with advanced gastric
cancer according to GSTP1 Ile105Val polymorphism.
Probability of survival
1.0
0.8
0.6
0.4
0.2
0.0
0.0 5.0 10.0 15.0 20.0 25.0
Overall survival time (months)
Group
1
2
1-censored
2-censored
P=0.002
GSTP1-105 lle/lle
GSTP1-105 lle/Val+Val/Val
GSTP1, Glutathione S-transferases P1.
1, lle/lle
2, lle/Val+Val/Val
Polymorphism of GSTP1 as a Prediction of Clinical Outcome 851
resistance or sensitivity to chemotherapy is of major interest
in selecting the most likely effective first-line treatment, avoid-
ing adverse side effects of a toxic but inactive therapy. Glu-
tathione S-transferases catalyze the first step in the formation
of mercapturic acids, initiating a pathway that results most-
ly in the elimination of toxic compounds. Previous studies
revealed that substitution of isoleucine with valine at codon
105 alters the function of the GSTP1 protein (20).
It is known that the allele frequencies of metabolic genes are
not equally distributed throughout the human population
but follow diverse ethnic and/or geographic-specific patterns.
An American population-based study reported, for GSTP1,
the prevalence rates of 105Ile/105Val heterozygosity and 105Val/
105Val homozygosity were found to be 30% to 51% and 7%
to 19%, respectively (15). For gastric cancer patients, a Chi-
nese study showed the rates of GSTP1 105Ile/105Val genotype
and 105Val/105Val genotype were 30.6% and 0% (21). In our
study, 41% of the participants were heterozygous for 105Ile/
105Val, and 7% were homozygous for the 105Val/105Val. This
frequency is similar to the frequencies above.
Stoehlmacher et al. (17) reported clinical response was not
statistically associated with GSTP1 genotypes in patients with
colorectal cancer. Ott et al. (18) reported GSTP1 genotypes
were not correlated with response to neoadjuvant chemother-
apy in gastric cancer. Cabelguenne et al. (22) could not observe
a correlation between GSTP1 genotypes and response to cis-
platin neoadjuvant chemotherapy in head and neck squamous
cell cancer (HNSCC) patients. While another study showed
patients possessing the GSTP1-105 Val/Val genotype showed
a significant superior response in gastric cancer (23). Our
results demonstrated patients with at least one 105Val allele
had a better disease control rate compared with patients with
homozygous 105Ile/105Ile genotype. There were some contro-
versial reports about the impact of GSTP1 on clinical res-
ponse, indicating that other potential factors may affect the
response to chemotherapy, such as ethnic background, geo-
graphic pattern or other potential genetic factors.
Our study suggested patients with GSTP1 105Val geno-
type had a better clinical outcome to oxaliplatin/5-FU-based
chemotherapy in advanced gastric cancer patients. GSTP1
Ile105Val polymorphism was studied widely in various tumors,
such as prostate cancer, colorectal cancer, breast cancer, eso-
phageal cancer and gastric cancer. Reports in colorectal can-
cer patients demonstrated a significantly improved time to
progression and overall survival for carriers of the GSTP1 105Val
allele after 5-FU/oxaliplatin chemotherapy (17, 24). Goekkurt
et al. reported that patients with GSTP1 105Val/105Val geno-
type demonstrated significant superior median survival time
after 5-FU/cisplatin chemotherapy for gastric cancer (23). In
the present study, we demonstrated that patients with two
105Ile alleles had a significantly increased risk of progression
and death. In summary, the results we report here are in strong
agreement with the current understanding of GSTP1 involve-
ment in platinum detoxification. Our findings support the
hypothesis that increased platinum sensitivity may in part
be due to impaired GSTP1 enzyme function.
Recent clinical reports provided evidence for an association
between GSTP1 genotype and cytotoxicity of chemothera-
peutic agents (25). A study reported that oxaliplatin-related
cumulative neuropathy scored grade 3 was significantly more
frequent in patients homozygous for the GSTP1 105Ile allele
than in patients homozygous or heterozygous for the GSTP1
105Val allele (OR 5.75; 95% CI 1.08-30.74; P=0.02) (26).
Another report showed Patients with GSTP1 105Ile/105Ile geno-
type had a higher risk of developing a grade >or =2 docetax-
el-induced peripheral neuropathy (25). Similarly, our study
implied patients with two GSTP1 105Ile alleles had a higher
risk of developing neurological toxicity, especially in grade 3
neuropathy. It was reported that individuals with two GSTP1
105Val alleles had a lower catalytic activity compared with indi-
viduals with two GSTP1 105Ile alleles (15). In particular, there
was in vitro evidence that GSTP1 105Val allele is much more
active than the wild-type alleles against cisplatin and carbo-
platin (27). Thus, the increasing rate of grade 3 neuropathy
in patients with 105Ile/105Ile GSTP1 genotype could be exp-
lained by the diminished capacity of detoxifying oxaliplatin.
And the involvement of GSTP1 in cellular apoptosis could
be another hypothesis to explain the protective effect of the
variant genotype.
In conclusion, we demonstrated that among patients with
gastric cancer who received 5-FU/oxaliplatin chemotherapy
as first-line treatment, those possessing the GSTP1 105Val
variant allele show a statistically significant benefit for both
time to progression and overall survival. Our results support
the hypothesis that the effects of chemotherapeutic drugs may
be altered when enzymes that could enhance the elimination
of these drugs show a reduced activity. However, more per-
spective multicentric randomized studies are required to con-
firm our preliminary results for patients with advanced gas-
tric cancer in Chinese population.
REFERENCES
1. Varadhachary G, Ajani JA. Gastric cancer. Clin Adv Hematol Oncol
2005; 3: 118-24.
2. Vanhoefer U, Rougier P, Wilke H, Ducreux MP, Lacave AJ, Van
Cutsem E, Planker M, Santos JG, Piedbois P, Paillot B, Bodenstein
H, Schmoll HJ, Bleiberg H, Nordlinger B, Couvreur ML, Baron B,
Wils JA. Final results of a randomized phase III trial of sequential
high-dose methotrexate, fluorouracil, and doxorubicin versus etopo-
side, leucovorin, and fluorouracil versus infusional fluorouracil and
cisplatin in advanced gastric cancer: a trial of the European Orga-
nization for Research and Treatment of Cancer Gastrointestinal Tract
Cancer Cooperative Group. J Clin Oncol 2000; 18: 2648-57.
3. Sadighi S, Mohagheghi MA, Montazeri A, Sadighi Z. Quality of life
in patients with advanced gastric cancer: a randomized trial com-
paring docetaxel, cisplatin, 5-FU (TCF) with epirubicin, cisplatin,
852 Q.-F. Li, R.-Y. Yao, K.-W. Liu, et al.
5-FU (ECF). BMC Cancer 2006; 6: 274.
4. Sumpter K, Harper-Wynne C, Cunningham D, Rao S, Tebbutt N,
Norman AR, Ward C, Iveson T, Nicolson M, Hickish T, Hill M,
Oates J. Report of two protocol planned interim analyses in a ran-
domised multicentre phase III study comparing capecitabine with
fluorouracil and oxaliplatin with cisplatin in patients with advanced
oesophagogastric cancer receiving ECF. Br J Cancer 2005; 92:
1976-83.
5. Van Cutsem E, Moiseyenko VM, Tjulandin S, Majlis A, Constenla
M, Boni C, Rodrigues A, Fodor M, Chao Y, Voznyi E, Risse ML,
Ajani JA. Phase III study of docetaxel and cisplatin plus fluorouracil
compared with cisplatin and fluorouracil as first-line therapy for
advanced gastric cancer: a report of the V325 Study Group. J Clin
Oncol 2006; 24: 4991-7.
6. Allen WL, Johnston PG. Role of genomic markers in colorectal can-
cer treatment. J Clin Oncol 2005; 23: 4545-52.
7. Yamayoshi Y, Iida E, Tanigawara Y. Cancer pharmacogenomics:
international trends. Int J Clin Oncol 2005; 10: 5-13.
8. Rosell R, Cecere F, Santarpia M, Reguart N, Taron M. Predicting
the outcome of chemotherapy for lung cancer. Curr Opin Pharma-
col 2006; 6: 323-31.
9. Olaussen KA, Dunant A, Fouret P, Brambilla E, Andre F, Haddad V,
Taranchon E, Filipits M, Pirker R, Popper HH, Stahel R, Sabatier L,
Pignon JP, Tursz T, Le Chevalier T, Soria JC. DNA repair by ERCC1
in non-small-cell lung cancer and cisplatin-based adjuvant chemother-
apy. N Engl J Med 2006; 355: 983-91.
10. Galanski M. Recent developments in the field of anticancer platinum
complexes. Recent Pat Anticancer Drug Discov 2006; 1: 285-95.
11. Raymond E, Faivre S, Woynarowski JM, Chaney SG. Oxaliplatin:
mechanism of action and antineoplastic activity. Semin Oncol 1998;
25 (2 Suppl 5): 4-12.
12. Marsh S, McLeod HL. Cancer pharmacogenetics. Br J Cancer 2004;
90: 8-11.
13. Nagasubramanian R, Innocenti F, Ratain MJ. Pharmacogenetics in
cancer treatment. Annu Rev Med 2003; 54: 437-52.
14. Goto S, Iida T, Cho S, Oka M, Kohno S, Kondo T. Overexpression
of glutathione S-transferase pi enhances the adduct formation of cis-
platin with glutathione in human cancer cells. Free Radic Res 1999;
31: 549-58.
15. Watson MA, Stewart RK, Smith GB, Massey TE, Bell DA. Human
glutathione S-transferase P1 polymorphisms: relationship to lung
tissue enzyme activity and population frequency distribution. Car-
cinogenesis 1998; 19: 275-80.
16. Booton R, Ward T, Heighway J, Ashcroft L, Morris J, Thatcher N.
Glutathione-S-transferase P1 isoenzyme polymorphisms, platinum-
based chemotherapy, and non-small cell lung cancer. J Thorac Oncol
2006; 1: 679-83.
17. Stoehlmacher J, Park DJ, Zhang W, Groshen S, Tsao-Wei DD, Yu
MC, Lenz HJ. Association between glutathione S-transferase P1, T1,
and M1 genetic polymorphism and survival of patients with metastat-
ic colorectal cancer. J Natl Cancer Inst 2002; 94: 936-42.
18. Ott K, Lordick F, Becker K, Ulm K, Siewert J, Hofler H, Keller G.
Glutathione-S-transferase P1, T1 and M1 genetic polymorphisms in
neoadjuvant-treated locally advanced gastric cancer: GSTM1-pre-
sent genotype is associated with better prognosis in completely resect-
ed patients. Int J Colorectal Dis 2008; 23: 773-82.
19. Caussanel JP, Levi F, Brienza S, Misset JL, Itzhaki M, Adam R,
Milano G, Hecquet B, Mathe G. Phase I trial of 5-day continuous
venous infusion of oxaliplatin at circadian rhythm-modulated rate
compared with constant rate. J Natl Cancer Inst 1990; 82: 1046-50.
20. Srivastava SK, Singhal SS, Hu X, Awasthi YC, Zimniak P, Singh
SV. Differential catalytic efficiency of allelic variants of human glu-
tathione S-transferase Pi in catalyzing the glutathione conjugation
of thiotepa. Arch Biochem Biophys 1999; 366: 89-94.
21. Liu B, Wei J, Zou Z, Qian X, Nakamura T, Zhang W, Ding Y, Feng
J, Yu L. Polymorphism of XRCC1 predicts overall survival of gas-
tric cancer patients receiving oxaliplatin-based chemotherapy in
Chinese population. Eur J Hum Genet 2007; 15: 1049-53.
22. Cabelguenne A, Loriot MA, Stucker I, Blons H, Koum-Besson E,
Brasnu D, Beaune P, Laccourreye O, Laurent-Puig P, De Waziers I.
Glutathione-associated enzymes in head and neck squamous cell car-
cinoma and response to cisplatin-based neoadjuvant chemotherapy.
Int J Cancer 2001; 93: 725-30.
23. Goekkurt E, Hoehn S, Wolschke C, Wittmer C, Stueber C, Hossfeld
DK, Stoehlmacher J. Polymorphisms of glutathione S-transferases
(GST) and thymidylate synthase (TS)--novel predictors for response
and survival in gastric cancer patients. Br J Cancer 2006; 94: 281-6.
24. Stoehlmacher J, Park DJ, Zhang W, Yang D, Groshen S, Zahedy S,
Lenz HJ. A multivariate analysis of genomic polymorphisms: predic-
tion of clinical outcome to 5-FU/oxaliplatin combination chemother-
apy in refractory colorectal cancer. Br J Cancer 2004; 91: 344-54.
25. Mir O, Alexandre J, Tran A, Durand JP, Pons G, Treluyer JM, Gold-
wasser F. Relationship between GSTP1 Ile(105)Val polymorphism
and docetaxel-induced peripheral neuropathy: clinical evidence of
a role of oxidative stress in taxane toxicity. Ann Oncol 2009; 20:
736-40.
26. Lecomte T, Landi B, Beaune P, Laurent-Puig P, Loriot MA. Glu-
tathione S-transferase P1 polymorphism (Ile
105
Val) predicts cumula-
tive neuropathy in patients receiving oxaliplatin-based chemothera-
py. Clin Cancer Res 2006; 12: 3050-6.
27. Ishimoto TM, Ali-Osman F. Allelic variants of the human glutathione
S-transferase P1 gene confer differential cytoprotection against anti-
cancer agents in Escherichia coli. Pharmacogenetics 2002; 12: 543-53.
... 66,67 The I150V polymorphism of GSTP1 with a phenotype of reduced enzymatic capacity has been correlated with better therapeutic outcomes in gastric cancer patients receiving oxaliplatin-based chemotherapy. 68,69 Besides platinum drugs, the sensitivity to doxorubicin and various alkylating agents in cancer may also be affected by the detoxification process mediated by GSTA subclass, GSTP1, and GSTM1 enzymes. 70,71 However, additional mechanisms other than GSH conjugation may be involved in GST-related chemoresistance to doxorubicin and alkylators, such as free radical scavenging and apoptosis suppression via the inhibition of the mitogenactivated protein kinase (MAPK) pathway. ...
Article
Full-text available
Resistance to cancer therapies has been a commonly observed phenomenon in clinical practice, which is one of the major causes of treatment failure and poor patient survival. The reduced responsiveness of cancer cells is a multifaceted phenomenon that can arise from genetic, epigenetic, and microenvironmental factors. Various mechanisms have been discovered and extensively studied, including drug inactivation, reduced intracellular drug accumulation by reduced uptake or increased efflux, drug target alteration, activation of compensatory pathways for cell survival, regulation of DNA repair and cell death, tumor plasticity, and the regulation from tumor microenvironments (TMEs). To overcome cancer resistance, a variety of strategies have been proposed, which are designed to enhance the effectiveness of cancer treatment or reduce drug resistance. These include identifying biomarkers that can predict drug response and resistance, identifying new targets, developing new targeted drugs, combination therapies targeting multiple signaling pathways, and modulating the TME. The present article focuses on the different mechanisms of drug resistance in cancer and the corresponding tackling approaches with recent updates. Perspectives on polytherapy targeting multiple resistance mechanisms, novel nanoparticle delivery systems, and advanced drug design tools for overcoming resistance are also reviewed.
... To date, several prognostic biomarkers have been proposed to risk stratify GC patients. HER2 (9), FGFR2 (10), Ecadherin (11), the PI3K signaling axis (12), VEGF (13), and p53 (14) are among the most important prognostic markers that have been introduced for GC. However, the controversial prognostic values and lack of validation evidence of these biomarkers for clinical use has led to ongoing research to identify novel, effective, and more precise biomarkers. ...
Article
Full-text available
Aim: The present study aimed to evaluate the expression profile, prognostic value, and possible correlation of TRIM29 with β-catenin, Cyclin D, and Bcl2 in Iranian patients with GC. Background: Tripartite Motif Containing 29 (TRIM29) has been reported to function as an oncogene or a tumor suppressor depending on the tumor type. This contextual function has created a controversial situation that needs to be fully delineated in various cancers. Although few studies have reported an elevated TRIM29 expression in gastric cancer (GC), its clinicopathological and prognostic values as well as possible molecular mechanisms are yet to be re-evaluated in different populations. Methods: Real-time quantitative PCR was used to detect TRIM29, β-catenin, Cyclin D, and Bcl-2 expression in 40 GC and their adjacent normal tissues. Patients were further stratified into high and low expression subgroups based on their TRIM29 expression levels. The association of TRIM29 expression level with β-catenin, Cyclin D, BCL2, some clinicopathological features, and patients' overall survival (OS) was assessed using appropriate statistical analyses. Results: The results showed a significantly higher TRIM29 expression level in GC tissues compared with their corresponding normal tissues (fold change=2.94, p=0.003). Patients with high TRIM29 expression levels exhibited poorer OS (HR=1.25, 95% CI: 1.06-1.47, p=0.007). High expression of TRIM29 was also associated with increased levels of β-catenin, Cyclin D, and Bcl-2 genes expression. Conclusion: Overexpression of TRIM29 is associated with poor prognosis in patients with GC and is probably mediated through β-catenin/Cyclin D/Bcl2 pathway and can be considered as a potential independent prognostic marker.
... Thirty-nine records were excluded due to the following reasons: not involving toxicity outcome (n = 8), not involving rs1695 (n = 7), pharmacokinetic studies (n = 5), abstracts, or case reports (n = 7), or unable to extract data (n = 12). Therefore, this meta-analysis included ten studies [3,7,8,[11][12][13][14][15][16][17]. Figure 2 presents the OR and 95% CIs relating to the rs1695 polymorphism in the GSTP1 gene with the risks of various platinum-associated toxicities, such as gastrointestinal toxicity, hematological toxicity, neutropenia, and neurotoxicity. Gastrointestinal toxicity involved four studies and suggested that the rs1695 polymorphism was associated with an approximately 44% decrease in the odds of toxicity compared to the wild-type Table 1 shows the baseline characteristics of the included studies. ...
Article
Full-text available
Background: Platinum-based chemotherapy regimens have been proven to be effective in various cancers; however, considerable toxicities may develop and can even lead to treatment discontinuation. Diverse factors may influence adverse treatment events, with pharmacogenetic variations being one prime example. Polymorphisms within the glutathione S-transferase pi 1 (GSTP1) gene may especially alter enzyme activity and, consequently, various toxicities in patients receiving platinum-based chemotherapy. Due to a lack of consistency in the degree of elevated complication risk, we performed a systematic literature review and meta-analysis to determine the level of platinum-associated toxicity in patients with the GSTP1 rs1695 polymorphism. Methods: We conducted a systematic search for eligible studies published before January 2022 from PubMed, Web of Science, and EMBASE based on Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Odds ratios (ORs) and 95% confidence intervals (CIs) were calculated to evaluate the strength of the association between the rs1695 polymorphism and various toxicities. Results: Ten eligible studies met the inclusion criteria. The pooled ORs for hematological toxicity and neutropenia in the patients with the variant (G) allele were 1.7- and 2.6-times higher than those with the AA genotype (95% CI 1.06–2.73 and 1.07–6.35), respectively. In contrast, the rs1695 polymorphism resulted in a 44% reduced gastrointestinal toxicity compared to wild-type homozygotes. Conclusion: Our study found that the GSTP1 rs1695 polymorphism was significantly correlated with platinum-induced toxicities. The study also revealed that rs1695 expression exhibited tissue-specific patterns and thus yielded opposite effects in different tissues. A personalized chemotherapy treatment based on these polymorphisms may be considered for cancer patients in the future.
Article
Advanced colorectal cancer (CRC) patients with peritoneal metastasis (PM) are highly aggressive malignancies with poor prognosis. Systematic chemotherapy and local treatments are the primary therapeutic approaches. However, systemic chemotherapy is...
Article
Purpose Majority of the gallbladder cancer (GBC) cases are diagnosed at an advanced stage where chemotherapy alone (or in combination with other treatment methods) is mainly opted as therapeutic approach. However, success or failure of this approach largely depends on the interindividual genetic differences. Careful consideration on the genetic association could assist in the evaluation of patient’s treatment response and survival rate. Hence, the present study aims to investigate the survival of patients with GBC and their treatment response to gemcitabine and cisplatin/carboplatin-based chemotherapy in association with Glutathione S-transferase (GSTs) gene polymorphism. Material and Methods A total of 216 histologically confirmed cases of gallbladder cancer were recruited. A total of 180 patients were treated with gemcitabine and cisplatin/carboplatin-based chemotherapy. GSTM1, GSTT1, and GSTP1 genotypes were determined by multiplex PCR and by PCR restriction fragment length polymorphism (PCR-RFLP), respectively. The influence of genetic polymorphism on overall survival was analyzed by Kaplan–Meier method, survival rate difference was analyzed by log-rank test, and hazard ratio for mortality outcomes was estimated using Cox regression method. Results GBC patients having genotype GSTP1 ( AG + GG ) showed poor 3-year survival rate of 0.8% compared to 10.9% of GSTP1 ( AA ) genotype (χ ² = 6.456, P = 0.011). The multivariate Cox regression results showed that the death risk was significantly higher in GSTP1 (AG + GG) genotype (HR = 3.858, P = 0.050). We found no association of GSTM1 and GSTT1 gene polymorphism with the survival; however, the combined genotypes of GSM1/GSTP1, GSTT1/GSTP1, and GSTM1/GSTT1/GSTP1 were associated with survival ( P = 0.053, 0.006, and 0.058, respectively). Increased death hazard was noted by the genotype combinations of GSTM1+/GSTP1AG + GG (HR = 3.484, P = 0.024), GSTM1-/GSTP1AG + GG (HR = 2.721, P = 0.014), GSTT1+/GSTP1AG + GG (HR = 20.690, P = 0.001), and GSTT1-/GSTP1AA (HR = 26.111, P < 0.0001). Our findings indicate that chemotherapy treatment response of GSTP1 (AG + GG ) has 1.62-fold increased risk for progression compared to GSTP1 (AA ) genotype (p = 0.018); however, none of the genotypes showed association with overall survival and death risk after chemotherapeutic treatment. Conclusion We found that the presence of GSTP1 ( AG + GG) genotype showed survival disadvantage and poor treatment outcomes in response to gemcitabine and cisplatin/carboplatin-based chemotherapy. This could serve as biomarker, and future research in pharmacogenomics will definitely pave the way for the development of better treatment approach for GBC.
Chapter
Pharmacogenomics is a powerful tool to predict individual response to treatment, in order to personalize therapy, and it has been explored extensively in oncology practice. Not only efficacy on the malignant disease has been investigated but also the possibility to predict adverse effects due to drug administration. Chemotherapy-induced peripheral neurotoxicity (CIPN) is one of those. This potentially severe and long-lasting/permanent side effect of commonly administered anticancer drugs can severely impair quality of life (QoL) in a large cohort of long survival patients. So far, a pharmacogenomics-based approach in CIPN regard has been quite delusive, making a methodological improvement warranted in this field of interest: even the most refined genetic analysis cannot be effective if not applied correctly. Here we try to devise why it is so, suggesting how THE “bench-side” (pharmacogenomics) might benefit from and should cooperate with THE “bed-side” (clinimetrics), in order to make genetic profiling effective if applied to CIPN.Key words Pharmacogenomics Chemotherapy-induced peripheral neurotoxicityPersonalized medicine
Conference Paper
Full-text available
Sheet metal parts are widely used in the white goods industry, especially in oven manufacturing. After the baking trays are produced in progressive molds, they are exposed to temperatures of 800-860 °C for enamel coating. Due to the thinness of the trays or the different form structures, distortion problems are observed in the trays during the enameling process. In this study, thermal analysis was applied to unembossed and embossed trays in Ansys program. According to the results of the analysis, the maximum stress value of the embossed trays exposed to 850 °C temperature decreased from 11293 MPa to 9460 MPa compared to the unembossed trays resulting in an improvement of % 16,23. Maximum strain value decreased from 0,061 to 0,051 resulting in an improvement of % 16,39.
Conference Paper
Full-text available
Sheet metal materials are used in parts of white appliances that require strength. In order for the products to be durable and light, the most appropriate sheet thickness is important in terms of cost and quality. Sheet metal parts are produced in presses and molds, so they are exposed to different drawing, cutting and drilling forces. In this study, the deformations of the formed and unformed trays used in the ovens as a result of the applied force were investigated. While the maximum total deformation value of the formless tray was 6,6747 mm, this value decreased to 6,3842 mm with the addition of the form, resulting in an improvement of % 4,35. The maximum equivalent stress value of the formed tray decreased from 761,23 MPa to 742,39 MPa compared to the unformed tray, resulting in an improvement of % 2,47. According to these results, the embossed baking tray was found to be more resistant to mechanical forces than the unembossed trays.
Article
Full-text available
Glutathione-S-transferases (GST) regulate the cellular response to oxidative stress. We previously highlighted the importance of oxidative stress in taxane toxicity and therefore investigated the relationship between the GST isoforms M1, T1 and P1 gene polymorphisms and docetaxel (Taxotere)-induced peripheral neuropathy (DIPN). The GSTM1 (null), GSTT1 (null) and GSTP1 (Ile(105)Val and Ala(114)Val) polymorphisms were determined in a cohort of cancer patients treated with docetaxel and entered in a clinical trial database. The relationship between GST polymorphisms and grade > or = 2 DIPN as primary end point was studied. Fifty-eight patients (median age 61 years) received a total of 261 cycles of docetaxel given as single agent. Patients with GSTP1 (105)Ile/(105)Ile genotype had a higher risk of developing a grade > or = 2 DIPN than did those with other GSTP1 genotypes (8 of 27 versus 2 of 31, respectively, odds ratio 6.11; 95% confidence interval 1.17-31.94; P = 0.03). In multivariate analysis, grade > or = 2 DIPN was strongly correlated with GSTP1 (105)Ile/(105)Ile genotype (P = 0.01) and the number of cycles (P = 0.03). We found a significant correlation between GSTP1 (105)Ile/(105)Ile genotype and the development of grade > or = 2 DIPN. This finding strongly suggests a role of oxidative stress in the pathophysiology of DIPN.
Article
The toxic effects and tissue uptake of both cisplatin and oxaliplatin--[(1R, 2R)-1,2-cyclohexanediamine-N,N'] [oxalato(2-)-O,O']platinum--were previously shown to vary similarly according to dosing time in mice. A 4-hour infusion of cisplatin resulted in fewer side effects and allowed administration of higher doses at 16 hours than at 4 hours in patients with cancer. We hypothesized that the continuous venous infusion of oxaliplatin for 5 days would be less toxic and would deliver a higher dose to the patient if the drug were infused at a circadian rhythm-modulated rate (peak at 16 hr; schedule B) rather than at a constant rate (schedule A). We tested this hypothesis in a randomized phase I trial. We escalated the dose of oxaliplatin to the patient by 25 mg/m2 per course. Courses were repeated every 3 weeks. An external, multichannel, programmable-in-time pump was used for the infusions. Toxicity was assessable for 94 courses in 23 patients (12 patients with breast carcinoma, nine with hepatocellular carcinoma, and two with cholangiocarcinoma). The incidence of neutropenia of World Health Organization grades II-IV and the incidence of distal paresthesias were 10 or more times higher (P less than .05) with schedule A than with schedule B. In addition, vomiting was 55% higher (P = .15) with schedule A than with schedule B. Furthermore, with schedule B, the mean dose of oxaliplatin (P less than .001) and its maximum tolerated dose (P = .06) could be increased by 15% over those doses with schedule A. An objective response was achieved in two of the 12 patients with previously treated breast cancer. We recommend that the dose of oxaliplatin for phase II trials be 175 mg/m2, delivered according to the circadian rhythm-modulated rate.
Article
The association between glutathione S-transferase (GST) activity as measured by 1-chloro-2,4-dinitrobenzene (CDNB) conjugation and genotype at exon 5 and exon 6 of the human GSTP1 gene was investigated in normal lung tissue obtained from 34 surgical patients. These samples were genotyped for previously identified polymorphisms in exon 5 (Ile105Val) and exon 6 (Ala114Val) by PCR-RFLP and direct sequencing. GST enzyme activity was significantly lower among individuals with the 105 Val allele. Homozygous Ile/Ile samples (n = 18) had a mean cytosolic CDNB conjugating activity of 74.9 +/- 3.8 nmol/mg per min; heterozygotes (n = 13) had a mean specific activity of 62.1 +/- 4.2 nmol/mg per min and homozygous Val/Val (n = 3) had a mean specific activity of 52.5 +/- 4.5 nmol/mg per min. The CDNB conjugating activity measured for the Ile/Ile genotype group was significantly different from that observed in the Ile/Val group (P = 0.03), and from Ile/Val and Val/Val genotypes combined (P = 0.009). Mean GST activity values were consistently lower in individuals with genotypes containing the 105 valine allele, regardless of smoking exposure. Genotypes at codon 114 were also assessed but the mean GST activity was not significantly lower in individuals with the 114 valine allele. A new haplotype, present in two samples who were homozygous 105Ile and had a 114Val, was identified and proposed as GSTP1*D. Frequencies of the exon 5 and exon 6 polymorphisms were determined in samples obtained from European-Americans, African-Americans and Taiwanese. The differences observed were highly significant suggesting the possibility of GSTP1 genotype-associated, ethnic differences in cancer susceptibility and chemotherapeutic response.
Article
Oxaliplatin, a platinum-based chemotherapeutic agent with a 1,2-diaminocyclohexane (DACH) carrier ligand, has shown in vitro and in vivo efficacy against many tumor cell lines, including some that are resistant to cisplatin and carboplatin. The retention of the bulky DACH ring by activated oxaliplatin is thought to result in the formation of platinum-DNA adducts, which appear to be more effective at blocking DNA replication and are more cytotoxic than adducts formed from cisplatin. Studies by the National Cancer Institute (NCI) have suggested that oxaliplatin has a spectrum of activity different from that of either cisplatin or carboplatin, suggesting that it has different molecular targets and/or mechanisms of resistance. Oxaliplatin has been demonstrated to differ in some mechanisms associated with the development of cisplatin resistance. Compared with cisplatin-conditioned cells, deficiencies in mismatch repair (MMR) and increases in replicative bypass, which appear to contribute to cisplatin resistance, have not been shown to induce a similar resistance to oxaliplatin. A decreased likelihood of resistance development makes oxaliplatin a good candidate for first-line therapy. Studies also demonstrate additive and/or synergistic activity with a number of other compounds, however, suggesting the possible use of oxaliplatin in combination therapies.
Article
Alkylating agents are extensively used in the treatment of cancer. The clinical usefulness of this class of anticancer drugs, however, is often limited by the emergence of drug-resistant tumor cells. Increased glutathione (GSH) conjugation through catalysis by GSH S-transferases (GSTs) is believed to be an important mechanism in tumor cell resistance to alkylating agents. In the present study, we report that the allelic variants of human Pi class GST (hGSTP1-1), which differ in their primary structures at amino acids in positions 104 and/or 113, exhibit significant differences in their activity in the GSH conjugation of alkylating anticancer drug thiotepa. Mass spectrometry revealed that the major product of the reaction between thiotepa and GSH was the monoglutathionyl-thiotepa conjugate. While nonenzymatic formation of monoglutathionyl-thiotepa was negligible, the formation of this conjugate was increased significantly in the presence of hGSTP1-1 protein. The hGSTP1-1-catalyzed GSH conjugation of thiotepa was time and protein dependent and followed Michaelis-Menten kinetics. The catalytic efficiency of hGSTP1-1(I104, A113) variant was approximately 1.9- and 2.6-fold higher compared with hGSTP1-1(V104,A113) and hGSTP1-1(V104,V113) isoforms, respectively. The results of the present study indicate that the hGSTP1-1 polymorphism may be an important factor in GST-mediated tumor cell resistance to thiotepa, and that subjects homozygous for the hGSTP1-1(I104,A113) allele, which is most frequent in human populations, are likely to be at a greater risk for developing GST-mediated resistance to thiotepa than heterozygotes or homozygotes with valine 104 background.
Article
In this paper, we provide direct evidence that glutathione S-transferase pi (GSTpi) detoxifies cisplatin (CDDP). We used human colonic cancer HCT8 cells sensitive and resistant to CDDP, the level of cisplatin-glutathione adduct (DDP-GSH) being higher in the resistant cells. There was an overexpression of GSTpi mRNA in these CDDP-resistant cells. Incubation of the cells with CDDP resulted in the formation of DDP-GSH dependent on the CDDP concentration and the incubation time. The formation of DDP-GSH was abolished when the cells were pre-treated with ethacrynic acid or ketoprofen, inhibitors of GSTpi. Purified GSTpi also catalyzed the formation of DDP-GSH in vitro, with an apparent Km of 0.23 mM for CDDP and an apparent Vmax of 4.9 nmol/min/mg protein. The increase in DDP-GSH produced by GSTpi was linear with incubation time up to 3 h and optimal of pH 7.4. A GSTpi transfectant cell line was constructed in HCT8 cells using a pcDNA3.1 (-)/Myc-His B with an expression vector containing cDNA for GSTpi. Transfection of GSTpi cDNA into HCT8 cells resulted in an increase in the expression of GSTpi by 1.4-fold in parallel with an augmentation of the formation of DDP-GSH. These results suggest that GSTpi plays a role in the formation of DDP-GSH and the acquisition of resistance to CDDP in cancer cells.
Article
To compare the efficacy and tolerability of etoposide, leucovorin, and bolus fluorouracil (ELF) or infusional fluorouracil plus cisplatin (FUP) with that of the reference protocol of fluorouracil, doxorubicin, and methotrexate (FAMTX) in advanced gastric cancer. A total of 399 patients with advanced adenocarcinoma of the stomach were randomized and analyzed for toxicity, tumor response, and progression-free and overall survival. Only reviewed and confirmed responses were considered. The analysis of remission was based on assessable patients with documented measurable lesions. The intent-to-treat principle, log-rank test, and Cox regression model were used for the statistical analysis of time-to-event end points. The overall response rate for 245 eligible patients with measurable disease was 9% with ELF, 20% with FUP, and 12% with FAMTX, with no significant differences. One hundred twelve patients were eligible for efficacy in assessable, nonmeasurable disease. No change was observed in 66% of patients treated with ELF, 56% with FUP, and 55% with FAMTX. Two patients achieved a complete tumor regression (one each for ELF and FAMTX). With a median follow-up time of 4.5 years, the median survival times were 7.2 months with ELF, 7.2 months with FUP, and 6.7 months with FAMTX, respectively, with no significant differences. Nonhematologic and hematologic toxicities of ELF, FUP, and FAMTX were acceptable, with neutropenia being the major toxicity for all three regimens. Seven treatment-related deaths occurred (two with FUP and five with FAMTX). All three investigated regimens demonstrate modest clinical efficacy and should not be regarded as standard treatment for advanced gastric cancer. New strategies should be considered to achieve a better clinical efficacy in the treatment of advanced gastric cancer.
Article
Glutathione S-transferases (GSTs) are metabolic phase II enzymes that promote reactive metabolite elimination by conjugating them to glutathione (GSH). Because of their important role in xenobiotic metabolism and detoxification, they have been implicated in carcinogenesis processes, especially epithelium transformation. Moreover, their influence on response to chemotherapy in cancer patients has been demonstrated. Genetic polymorphisms for GSTM1, GSTT1 and GSTP1 have been found in human populations and have been shown to have phenotypic consequences. To investigate the role of GST enzymes in carcinogenesis and in response to chemotherapy in patients with head and neck squamous cell carcinoma (HNSCC), GSTP1, GSTM1 and GSTT1 were studied prospectively in a large series of HNSCC patients. Correlations between GST alterations, p53 mutation status and clinical response to chemotherapy were investigated. We showed that the risk of developing laryngeal cancer was increased by 2.6-fold [95% CI 1.6--6.1] in patients with the GSTM1 null genotype and by 2.8-fold [95% CI 0.9--8.1] in patients with the homozygous GSTP1 val105 genotype. Furthermore, individuals with this latter genotype were over-represented in the p53 mutation group (p = 0.05). After storage duration and hemolysis adjustment, a significantly lower plasmatic GSTP1 level was observed in complete responders compared with partial and non-responders (mean: 4.4 +/- 0.06 microg/l, 4.7 +/- 0.06 microg/l and 4.7 +/- 0.07 microg/l; p = 0.05), respectively. The prevalence of p53-mutated tumors was significantly higher in the group of non-responders (81%) compared with partial (60%) and complete responders (64%) (p = 0.05). Two types of multivariate analysis were performed including parameters that have been shown to influence response to chemotherapy significantly in univariate analysis. p53 mutations and high tumor stage are independent factors of non-response to chemotherapy, whereas plasmatic GSTP1 levels and low tumor stage are independent factors of complete response. Our data suggest that GST enzymes are associated with larynx cancer and that their use as predictive factors and treatment targets should be further explored.
Article
Members of the glutathione S-transferase (GST) superfamily are important in cellular defense mechanisms. These enzymes attach reduced glutathione to electrophilic groups in a wide variety of toxic compounds, including chemotherapeutic agents. Certain polymorphisms in GSTs are associated with changes in enzyme activity, sensitivity to chemotherapy, and overall patient survival. In a retrospective study, we investigated associations between common polymorphisms in genes for several GST subclasses (GSTP1, GSTT1, GSTM1) and survival of patients with metastatic colorectal cancer receiving 5-fluorouracil (5-FU)/oxaliplatin chemotherapy. During 1998-2000, 107 previously treated patients with advanced colorectal cancer received 5-FU/oxaliplatin combination chemotherapy. Associations between deletion polymorphisms in GSTM1 and GSTT1 genes and between a polymorphism in the GSTP1 gene that generates an Ile(105)Val in the GSTP1 protein and survival were evaluated using relative risks (RRs) of dying and the log-rank test. All statistical tests were two-sided. Patients heterozygous for the GSTP1 polymorphism had an RR = 0.47 (95% confidence interval [CI] = 0.27 to 0.81) compared with patients homozygous for the GSTP1 (105)Ile allele. Patients homozygous for the mutant polymorphism had an RR = 0.16 (95% CI = 0.04 to 0.63). After adjustment for performance status and tumor site, the stratified RRs were 0.28 (95% CI = 0.07 to 1.10) for patients with two (105)Val alleles and 0.64 (95% CI = 0.36 to 1.16) for those with one (105)Val allele (P =.042). Patients with the (105)Val/(105)Val genotype survived a median of 24.9 months, those with the (105)Ile/(105)Ile genotype a median of 7.9 months, and those with the (105)Ile/(105)Val genotype a median of 13.3 months (P<.001). The GSTM1 and GSTT1 genotypes were not associated with survival or clinical response. The GSTP1 Ile(105)Val polymorphism is associated in a dose-dependent fashion with increased survival of patients with advanced colorectal cancer receiving 5-FU/oxaliplatin chemotherapy.
Article
The polymorphic human GSTP1 gene locus encodes proteins that differentially metabolize electrophilic substrates, including, many chemotherapeutic agents used in clinical cancer therapy. In this study, we used XL1-Blue MRF strain, transformed with phagemid expression vectors carrying cDNAs of three GSTP1 alleles, to investigate the cytoprotective abilities of the different GSTP1 alleles against four clinically active anticancer agents, namely, carboplatin, cisplatin, thiotepa, and 4-hydroperoxyifosfamide. Following induction of protein expression with isopropyl-beta-d-thiogalactoside, the cells were treated with each drug for 3 h (1 h for 4-hydroperoxyifosfamide). Surviving fractions were determined and used to compute a cytoprotective factor for each allele against each drug. The results showed all the GSTP1 alleles to be cytoprotective, albeit, to different degrees. For cisplatin and carboplatin, the allele was most protective, with CPs of 5.58 and 3.76, respectively, compared with 1.21 and 1.61 for and 2.50 and 2.79 for. In contrast, protection against thiotepa was highest for the allele, with a cytoprotective factor of 1.56, compared to 1.32 for and 1.1 for. For 4-hydroperoxyifosfamide, the CP for and was the same, 1.45, compared with 1.18 for. These data demonstrate significant differences in the ability of the different GSTP1 alleles to protect against the cytotoxicity of electrophilic anticancer agents. The level of protection differs significantly between different GSTP1 alleles, and between different anticancer agents. The optimized prokaryotic system described provides a useful and rapid tool for pharmacogenetic analysis of the effects of genes on drug sensitivity.