+ All Categories
Home > Documents > Association of genetic polymorphism of glutathione S-transferase (GSTM1, GSTT1, GSTP1) with bladder...

Association of genetic polymorphism of glutathione S-transferase (GSTM1, GSTT1, GSTP1) with bladder...

Date post: 18-Dec-2016
Category:
Upload: shiva
View: 220 times
Download: 0 times
Share this document with a friend
11
Original article Association of genetic polymorphism of glutathione S-transferase (GSTM1, GSTT1, GSTP1) with bladder cancer susceptibility Mohammad Reza Safarinejad, M.D.*, Saba Safarinejad, S.B., Nayyer Shafiei, B.Ch.E., Shiva Safarinejad, M.S. Private Clinical Center for Urological Disease Diagnosis and Private Clinic Specialized in Urological and Andrological Genetics, Teheran, Iran Received 13 September 2011; received in revised form 23 October 2011; accepted 11 November 2011 Abstract The glutathione-S-transferases (GSTs) comprise a class of enzymes that detoxify carcinogenic compounds by conjugating glutathione to facilitate their removal. Polymorphisms in GSTM1, GSTT1, and GSTP1 genes have been related to risk for bladder cancer. Studies focusing on GSTs gene variants relationship with the risk of bladder cancer have produced conflicting and inconsistent results. We examine the association between genetic polymorphism of glutathione S-transferase P1, GSTM1, GSTT1 genes and development of bladder transitional cell carcinoma (TCC). The study population consisted of 166 histologically confirmed male bladder TCC cases and 332 healthy male controls. Genotyping was done using the polymerase chain reaction restriction fragment length polymorphism (PCR-RFLP) method and also investigated combined gene interactions. The GSTP1 Val/Val genotype was significantly associated with bladder cancer (OR 4.32, 95% CI: 2.64 – 6.34), whereas the association observed for GSTM1 null (OR 1.32, 95% CI: 0.82–2.62; P 0.67) and GSTT1 null genotype (OR 1.18, 95% CI: 0.79 –1.67; P 0.74) did not reach statistical significance. There was a significant multiple interaction between GSTM1, GSTT1, and GSTP1 genotypes in risk of bladder cancer (P for interaction 0.02). The risk associated with the concurrent presence of GSTM1 positive and GSTP1 Ile/Val or Val/Val (OR 3.71, 95% CI: 2.34 –5.54) and GSTT1 positive and GSTP1 Ile/Val or Val/Val (OR 2.66, 95% CI: 1.54 – 4.72) was statistically significant. Patients carrying GSTP1 Val/Val genotype were at increased risk for developing high-grade (OR 7.68, 95% CI: 4.73–19.25) and muscle invasive (OR 10.67, 95% CI: 6.34 –21.75) bladder cancer. High risk for bladder TCC also was observed with respect to combined GSTT1 null/GSTP1 Ile/Val or Val/Val (OR 4.76, 95% CI: 2.68 –18.72) and GSTM1 null/GSTT1 null/GSTP1 Ile/Val or Val/Val (OR 6.42, 95% CI: 4.76 –14.72) genotype variant. This study suggests that the GSTP1 polymorphism and its combination with GSTM1, and GSTT1 may be associated with bladder cancer susceptibility in the Iranian population. Further confirmation in large population-based studies is needed. © 2013 Elsevier Inc. All rights reserved. Keywords: Bladder neoplasm; Glutathione S-transferase (GST) M1; GSTT1; GSTP1; Polymorphism; Genetics; Epidemiology 1. Introduction Urinary bladder cancer is the fourth most common type of cancer in men and the seventh in women, with an annual incidence of 32 of 100,000 in men and 9 of 100,000 in women [1]. Bladder cancer is usually considered to be caused by environmental carcinogens. Tobacco smoking and occupational exposure to aromatic amines and polycy- clic hydrocarbons have been associated with risk of bladder cancer [2]. There is increasing evidence suggesting that genetic polymorphisms of xenobiotic-metabolizing en- zymes could increase individual susceptibility to various environmental and clinical conditions. The balance between activation and detoxification of carcinogens affects the amount of deoxyribonucleic acid (DNA) damage that oc- curs in cells [3]. Glutathione-S-transferases (GSTs) consti- tute a superfamily of phase II enzymes that are involved in the detoxification of exogenous substrates such as xenobi- otics, environmental substances, and carcinogenic com- pounds [4,5]. To date, human cytosolic GST superfamily contains at least 16 genes subdivided into 8 distinct classes designated as: (alpha), (mu), (kappa), (omega), (pi), (sigma), (theta), and (zeta), which are encoded by the GSTA, GSTM, GSTK, GSTO, GSTP, GSTS, GSTT, and GSTZ genes, respectively [6,7]. Functional polymor- phisms have been identified in the GSTM1, GSTT1, and * Corresponding author. Tel.: 0098-21-22454499; fax: 0098-21- 22456845. E-mail address: [email protected] (M.R. Safarinejad). Urologic Oncology: Seminars and Original Investigations 31 (2013) 1193–1203 1078-1439/$ – see front matter © 2013 Elsevier Inc. All rights reserved. http://dx.doi.org/10.1016/j.urolonc.2011.11.027
Transcript
Page 1: Association of genetic polymorphism of glutathione S-transferase (GSTM1, GSTT1, GSTP1) with bladder cancer susceptibility

C(Gp

Original article

Association of genetic polymorphism of glutathione S-transferase(GSTM1, GSTT1, GSTP1) with bladder cancer susceptibility

Mohammad Reza Safarinejad, M.D.*, Saba Safarinejad, S.B., Nayyer Shafiei, B.Ch.E.,Shiva Safarinejad, M.S.

Private Clinical Center for Urological Disease Diagnosis and Private Clinic Specialized in Urological and Andrological Genetics, Teheran, Iran

Received 13 September 2011; received in revised form 23 October 2011; accepted 11 November 2011

Abstract

The glutathione-S-transferases (GSTs) comprise a class of enzymes that detoxify carcinogenic compounds by conjugating glutathione tofacilitate their removal. Polymorphisms in GSTM1, GSTT1, and GSTP1 genes have been related to risk for bladder cancer. Studies focusingon GSTs gene variants relationship with the risk of bladder cancer have produced conflicting and inconsistent results. We examine theassociation between genetic polymorphism of glutathione S-transferase P1, GSTM1, GSTT1 genes and development of bladder transitionalcell carcinoma (TCC). The study population consisted of 166 histologically confirmed male bladder TCC cases and 332 healthy malecontrols. Genotyping was done using the polymerase chain reaction restriction fragment length polymorphism (PCR-RFLP) method and alsoinvestigated combined gene interactions. The GSTP1 Val/Val genotype was significantly associated with bladder cancer (OR � 4.32, 95%

I: 2.64–6.34), whereas the association observed for GSTM1 null (OR � 1.32, 95% CI: 0.82–2.62; P � 0.67) and GSTT1 null genotypeOR � 1.18, 95% CI: 0.79–1.67; P � 0.74) did not reach statistical significance. There was a significant multiple interaction betweenSTM1, GSTT1, and GSTP1 genotypes in risk of bladder cancer (P for interaction � 0.02). The risk associated with the concurrentresence of GSTM1 positive and GSTP1 Ile/Val or Val/Val (OR � 3.71, 95% CI: 2.34–5.54) and GSTT1 positive and GSTP1 Ile/Val or

Val/Val (OR � 2.66, 95% CI: 1.54–4.72) was statistically significant. Patients carrying GSTP1 Val/Val genotype were at increased riskfor developing high-grade (OR � 7.68, 95% CI: 4.73–19.25) and muscle invasive (OR � 10.67, 95% CI: 6.34–21.75) bladder cancer. Highrisk for bladder TCC also was observed with respect to combined GSTT1 null/GSTP1 Ile/Val or Val/Val (OR � 4.76, 95% CI: 2.68–18.72)and GSTM1 null/GSTT1 null/GSTP1 Ile/Val or Val/Val (OR � 6.42, 95% CI: 4.76–14.72) genotype variant. This study suggests that theGSTP1 polymorphism and its combination with GSTM1, and GSTT1 may be associated with bladder cancer susceptibility in the Iranianpopulation. Further confirmation in large population-based studies is needed. © 2013 Elsevier Inc. All rights reserved.

Urologic Oncology: Seminars and Original Investigations 31 (2013) 1193–1203

Keywords: Bladder neoplasm; Glutathione S-transferase (GST) M1; GSTT1; GSTP1; Polymorphism; Genetics; Epidemiology

ta

1. Introduction

Urinary bladder cancer is the fourth most common typeof cancer in men and the seventh in women, with an annualincidence of 32 of 100,000 in men and 9 of 100,000 inwomen [1]. Bladder cancer is usually considered to becaused by environmental carcinogens. Tobacco smokingand occupational exposure to aromatic amines and polycy-clic hydrocarbons have been associated with risk of bladdercancer [2]. There is increasing evidence suggesting thatgenetic polymorphisms of xenobiotic-metabolizing en-

* Corresponding author. Tel.: �0098-21-22454499; fax: �0098-21-22456845.

pE-mail address: [email protected] (M.R. Safarinejad).

1078-1439/$ – see front matter © 2013 Elsevier Inc. All rights reserved.http://dx.doi.org/10.1016/j.urolonc.2011.11.027

zymes could increase individual susceptibility to variousenvironmental and clinical conditions. The balance betweenactivation and detoxification of carcinogens affects theamount of deoxyribonucleic acid (DNA) damage that oc-curs in cells [3]. Glutathione-S-transferases (GSTs) consti-tute a superfamily of phase II enzymes that are involved inthe detoxification of exogenous substrates such as xenobi-otics, environmental substances, and carcinogenic com-pounds [4,5]. To date, human cytosolic GST superfamilycontains at least 16 genes subdivided into 8 distinct classesdesignated as: � (alpha), � (mu), � (kappa), � (omega), �(pi), � (sigma), � (theta), and � (zeta), which are encoded byhe GSTA, GSTM, GSTK, GSTO, GSTP, GSTS, GSTT,nd GSTZ genes, respectively [6,7]. Functional polymor-

hisms have been identified in the GSTM1, GSTT1, and
Page 2: Association of genetic polymorphism of glutathione S-transferase (GSTM1, GSTT1, GSTP1) with bladder cancer susceptibility

ipigenmdidcowgcd

ladtoaGc

lnGpawGT

2

2

l

1194 M.R. Safarinejad et al. / Urologic Oncology: Seminars and Original Investigations 31 (2013) 1193–1203

GSTP1 genes coding for GSTs enzymes in the �, �, and �classes, respectively.GSTM1 gene is polymorphic, and atleast 4 different alleles exist [8]. The deleted genotypes,which result in the inactive form of the enzyme, have beennamed GSTM1 null [9]. The homozygous deletion ofGSTM1 has been shown to be associated with an increasedrisk of various types of cancer [10–13]. Studies investigat-ng the association between glutathione GSTs polymor-hism and bladder cancer risk report very conflicting andnconsistent results. Three studies reported that GSTM1 nullenotype is a risk factor for bladder cancer [14–16]. How-ver, a recent study found that the GSTM1 polymorphism isot associated with risk of bladder cancer [17]. The poly-orphism in the GSTT1 gene loci is also caused by a gene

eletion [18], and leads to virtual absence of enzyme activ-ty in individuals with the null genotype. A few studiesemonstrated nonsignificant diminished risk of bladder can-er with GSTT1 null genotype [15,19,20] nonetheless; somether studies demonstrated increased risk of bladder cancerith GSTT1 null genotype [21,22]. GSTMI and GSTTI nullenotypes have decreased capacity to detoxify certain car-inogens and have been linked with increased risks foreveloping bladder cancer [23,24].

The GSTP1313 A/G polymorphism at the nucleotideevel results in an amino acid variation of isoleucine/valinet codon 105 in the protein. Valine amino acid leads toiminished enzymes activity [25]. Törüner et al. reportedhat GSTP1Ile/Val or Val/Val genotype increases 1.75 foldf bladder cancer susceptibility [26]. In another study, Sriv-stava et al. [27] reported that the Val/Val genotype of theSTP1 gene polymorphism increases 7.1-fold of bladder

ancer susceptibility.Since Kempkes et al. [20] first presented the potential re-

ationship between GSTT1 and bladder cancer, an increasingumber of studies have investigated the association betweenSTs gene variation and bladder cancer risk in different ethnicopulations. However, the results from epidemiologic studiesre still inconsistent and conflicting. The purpose of this studyas to determine the frequencies of polymorphisms of GSTT1,STM, and GSTP1 genes and their association with bladderCC among Iranian population.

. Materials and methods

.1. Subjects

The study subjects consisted of 166 men with histopatho-ogically confirmed bladder TCC (mean age � SD, 63.8 �

9.2 years) and 332 cancer-free control subjects (mean age �SD, 63.4 � 9.7 years), recruited from our urology clinicbetween October 2006 and October 2009. No patient re-ceived chemotherapy or radiotherapy before recruitment.These patients were carefully paired with controls selectedfrom the blood donors with respect to environmental expo-

sure, diet routine, lifetime occupational history, smoking

history, general health conditions, and previous diseases.All cancer patients were newly diagnosed with transitionalcell carcinoma (TCC) of the bladder. The control groupconsisted of non-related healthy men without history ofmalignant disease who were matched to those in the casegroup for geographic origin, smoking status, and age range(�2 years). For each case, 2 controls were randomly se-lected. All study subjects provided informed consent priorto participating in the study, which was conducted accord-ing to Helsinki declaration (2004).

2.2. Inclusion/exclusion criteria

Patients with previous cancer, with cancer metastasizedto bladder from another origin, and those with previouschemotherapy or radiotherapy were excluded. Selection cri-teria for controls were no evidence of any personal or familyhistory of cancer or other serious illness. Control subjectswho had a previous diagnosis of any type of cancer wereexcluded.

2.3. Evaluations

All individuals underwent a complete physical examina-tion and answered a structured questionnaire that includeddemographic and ethnic background, height, weight, life-time occupational history, smoking habits, use of drugs andmedicines, general health conditions, and personal medicalhistory. Smoking status was defined as: (a) an ‘eversmoker’, who had smoked more than100 cigarettes in hislifetime; and (b) a former smoker, who had stopped smokingmore than 1 year before diagnosis for the patients and morethan 1 year before entry to the study for the control subjects.For each individual, the number of pack-years (PY) was cal-culated to demonstrate the cumulative smoking dose.

PY � (number of cigarettes smoked per day/20 ciga-rettes) � number of years smoked.

The clinical information about tumor size, number, stage,and grade, radiotherapy, and chemotherapy were obtainedfrom medical records. In all of the bladder cancer patients,the tumors were diagnosed histologically as transitional cellcarcinomas (TCC). The tumors were classified as superficial(pTa–pT1) or invasive (pT2–pT4) according to the 1997TNM staging system of the American Joint Committee onCancer. The histologic grades were subdivided into lowgrade and high grade using the grading systems of theWorld health Organization (WHO) and the InternationalSociety of Urological Pathology (ISUP).

2.4. Genotyping

Genotyping was done exactly as the methods we used inour previous study [28].

Genomic DNA was extracted from the peripheral bloodof subjects using the QIAmp blood kit (Qiagen, Chatsworth,

CA) and according to the method describe by Liu et al. [29].
Page 3: Association of genetic polymorphism of glutathione S-transferase (GSTM1, GSTT1, GSTP1) with bladder cancer susceptibility

A

c

Gp6

L

1195M.R. Safarinejad et al. / Urologic Oncology: Seminars and Original Investigations 31 (2013) 1193–1203

For the GSTM1 gene, DNA samples were amplified withthe primers: 5=-CTG CCC TAC TTG ATT GAT GGG-3=and 5=-CTG GAT TGT AGC AGA TCA TGC-3=, usingpolymerase chain reaction (PCR). The paired primers forGSTT1 were: 5=-TTC CTT ACT GGT CCT CAC ATCTC-3= and 5=-TCA CCG GAT CAT GGC CAG CA-3=.

mplification of human -globin (110 bp) with the primers5=-ACA CAA CTG TGT TCA CTA GC-3= and 5=-CAACTT CAT CCA CGT TCA CC-3= was used as an internalcontrol. PCR was performed in a final volume of 50 �l,onsisting of DNA (0.5 �l), and a PCR buffer containing

200 ng of the following reagents: dNTP (0.2 mM each),MgCl2 (1.5 mM), KCL (50 mM), Tris–HCl (10 mM, pH8.3), and 0.1% of bovine serum albumin. Reaction mixtureswere heated at 94°C for 5 minutes followed by 35 cycles ofamplification as follows: a denaturing step at 94°C for 90seconds, an annealing step at 59°C for 1 minute, and finalextension step at 72°C for 1 minute. Reaction products wereanalyzed by 2% agarose gel. The fragment lengths of thePCR products were 273-bp for individuals with one or moreGSTM1 alleles, and 480-bp for individuals with 1 or moreGSTT1 alleles.

Fig. 1. Representative screening for the GST genotypes. Patterns for eacSTM1 (273-bp fragment) genes, detected by PCR-RLFP analysis are shoroducts samples: 100 bp ladder (lane L); absence of null genotypes (lane, 7, 8, 10, 11).

Fig. 2. Cleavage of PCR products of GSTP1 gene by the Alw26I restriction

), Ile/Ile allele (lanes 2, 3, 5, 6); Ile/Val allele (lanes 1, 7, 8, 9), and Val/Val al

The GSTP1 genotype was also determined by themethod of Liu et al. [29] using a PCR-restriction fragmentlength polymorphism (RFLP) technique. An 177 bp frag-ment of the GSTP1 gene containing Ile to Val substitutionin exon 5 (codon 105) was amplified using the primer pair5=-ACC CCA GGG CTC TAT GGG AA-3= and 5=-TGAGGG CAC AAG AAG CCC CT-3=. The amplification cy-cles included an initial denaturing at 94°C for 30 seconds,followed by an annealing step at 61°C for 30 seconds, anda final extension step at 72°C for 30 seconds. The PCRproducts were then digested with Alw26I. HomozygousIle/Ile individuals had a single fragment of 177-bp, andhomozygous Val/Val individuals had both 92- and 85-bpfragments (Figs. 1, and 2). The presence of all 3 fragmentscorresponded to heterozygous Ile/Val individuals.

2.5. Quality control

Quality control was maintained by genotyping 10% du-plicates for cases and controls with 100% concordance tothe genotype by PCR-RFLP. Laboratory personnel were

genotypes, GSTT1 (480 bp fragment), -globin (268-bp fragment) andsence of the PCR product indicates the null genotype. Representative PCR9); GSTT1-null allele (lanes 2, 5), and GSTM1-null allele (lanes 1, 2, 5,

uclease. Representative PCR-RFLP products samples: 100 bp ladder (lane

h of thewn. Abs 3, 4,

endon

lele (lane 4).
Page 4: Association of genetic polymorphism of glutathione S-transferase (GSTM1, GSTT1, GSTP1) with bladder cancer susceptibility

oaTtfpb

gmnaaaV

cteidPI

OO

E

M

M

S

TT

1196 M.R. Safarinejad et al. / Urologic Oncology: Seminars and Original Investigations 31 (2013) 1193–1203

unable to distinguish among case, control, and quality con-trol samples.

2.6. Statistical analysis

The power of the study was determined using Quantosoftware, ver. 1.0 (available from: http://hydra.usc.edu/gxe)with consideration of following variables: case-controlstudy design, significance level (�) � 0.05 (2-sided), modelf inheritance was log additive, allele frequency was 0.18,nd the genetic effect for odds ratio (OR) was 1.8 or greater.he present study achieved 80% of the statistical power for

he Val allele of GSTP1, which showed the lowest allelerequency among the 3 polymorphisms. Results are ex-ressed as mean � standard deviation (SD). Hardy-Wein-erg equilibrium was examined using the goodness-of-fit 2

test to compare the observed allele frequencies with theexpected frequencies determined from control subjects. The2 or Fisher’s (F) exact test was used to assess the homo-eneity between cases and controls regarding ethnicity,edication use, cigarette smoking, disease extent, and ge-

otypes. Associations of polymorphisms with susceptibilitynd clinicopathologic characteristics of bladder cancer werenalyzed by unconditional logistic regression to estimatedjusted odds ratios and 95% confidence intervals (CI).

Table 1Distribution of selected demographic variables and risk factors among the

Characteristic Cancer patients

Age (year) 63.8 � 9.2BMI (kg/m2) 28.2 � 4.6

bese no. (%) 13 (7.8)ccupational status no. (%)Employed 60 (36.1)Unemployed 28 (16.9)Retired 78 (47.0)

ducational levelNone 8 (4.8)Primary school 59 (35.6)High school 90 (54.2)Graduate 9 (5.4)ean � SD biochemistry:Blood glucose (mg/dl) 99 � 11Alanine aminotransferase (IU/l) 28 � 14Aspartate aminotransferase (IU/l) 28 � 12Alkaline phosphatase (IU/l) 251 � 49Total bilirubin (mg/dl) 0.9 � 0.2Blood urea nitrogen (mg/dl) 14.4 � 4.2Creatinine (mg/dl) 0.9 � 0.2ean � SD hematological parameter:Hemoglobin (mg/dl) 14.6 � 0.34White blood cells (103) 7.78 � 2.11Red blood cells (109) 8.17 � 0.51Platelets (103) 874 � 117

moking statusNever 50 (30.1)Former 44 (26.5)Current 72 (43.4)

ariables that were significantly associated with bladder

ancer incidence or outcome by univariate analysis wereested by multiple logistic regression model in order toxamine the effect of all genotypes and clinical risk factors,ncluding, age, and cigarette smoking as independent pre-ictors. Statistical analysis was performed by Statisticalackage for the Social Sciences (SPSS) ver. 17.0 (SPSSnc., Chicago, IL).

cases and healthy controls

66) Controls (n � 332) P value

63.4 � 9.7 0.7128.4 � 4.2 0.7227 (8.1) 0.82

120 (36.1) 0.9458 (17.5) 0.64

154 (46.4) 0.78

17 (5.1) 0.82115 (34.7) 0.43183 (55.1) 0.3417 (5.1) 0.84

101 � 11 0.6228 � 16 0.8727 � 12 0.32

246 � 50 0.410.8 � 0.2 0.81

14.4 � 4.3 0.790.9 � 0.1 0.84

14.4 � 0.37 0.647.74 � 2.15 0.628.21 � 0.54 0.77877 � 120 0.73

105 (31.6) 0.7488 (26.5) 0.97

139 (41.9) 0.08

able 2he clinicopathologic characteristics in 166 patients with bladder TCC

Variable Mean � SD or n (%)

Age (years) 63.8 � 9.2Primary tumor stage

Superficial tumor (pTa–pT1) 107 (64.5)Invasive tumor (pT2–pT4) 59 (35.5)

Histopathologic gradingLow grade 119 (71.7)High grade 47 (28.3)

Tumor numberSingle 114 (68.7)Multiple 52 (31.3)

Tumor size (cm)�1 56 (33.7)1–3 50 (30.1)

cancer

(n � 1

�3 60 (36.2)

Page 5: Association of genetic polymorphism of glutathione S-transferase (GSTM1, GSTT1, GSTP1) with bladder cancer susceptibility

e

3b

HoaAGtC

3p

e

1197M.R. Safarinejad et al. / Urologic Oncology: Seminars and Original Investigations 31 (2013) 1193–1203

3. Results

3.1. Characteristics of the study population

The frequency distributions of demographic and clin-ical characteristics of the cases and controls are presentedin Tables 1 and 2. The cases and controls appeared to bewell matched on age: mean age was 63.8 � 9.2 years forcases and 63.4 � 9.7 years for controls (P � 0.71).Throughout our analyses, we consistently found no im-portant confounding by age, BMI, smoking status, oreducation. In the following paragraphs all of the reportedORs are adjusted for confounding factors in multivariateanalysis.

3.2. Genotyping

3.2.1. Genotype distributions of the GSTM1polymorphisms between the cases and controls

The genotype frequencies of GSTM1 polymorphismsamong the controls were in agreement with the Hardy-Weinberg equilibrium (2 test: P � 0.842). Genotype fre-quencies of the GSTM1 polymorphisms among the casepatients and control subjects and their associations with riskof bladder cancer are shown in Table 3. There was no mainffect on risk of bladder cancer.

.2.2. Genotype distributions of the GSTT1 polymorphismsetween the cases and controls

Distribution of GSTT1 genotype among our controls metardy-Weinberg equilibrium (2 test: P � 0.232). Theverall prevalence of the GSTT1 null genotype was similarmong cases and controls (21.1% vs. 20.8%, respectively).

tendency of increased risk was also observed betweenSTT1 null genotype and bladder cancer, but this associa-

ion failed to reach statistical significance (OR � 1.18, 95%

Table 3The distribution, as n (%), of GSTM1, GSTT1, and GSTP1 genotypes in

Genotype and allele frequency Control (n � 332) (%)

GSTM1Present 239 (72.0)Null 93 (28.0)

GSTT1Present 263 (79.2)Null 69 (20.8)

GSTP1Ile/Ile 172 (51.8)Ile/Val 152 (45.8)Val/Val 8 (2.4)Ile/Val or Val/Val 160 (48.2)

a Adjusted OR: adjusted in multivariate logistic regression models inclgenotypes.

I: 0.79–1.76; P � 0.74) (Table 3).

.2.3. Genotype distributions of the GSTP1olymorphisms between the cases and controls

The GSTP1 genotypes were also in Hardy-Weinbergquilibrium for the controls (2 test: P � 0.228).

The Ile/Val genotype was more frequent in cancer pa-tients with a significant trend for an OR of 2.32 (95% CI:1.24–3.65; P � 0.007). The proportion of the individualswith GSTP1 Val/Val genotype was higher in cases (14.5%vs. 2.4%), the ORs being 4.32 (95% CI: 2.64–6.34; P �0.001).

3.3. Distribution of double GST genotypes among cancerpatients and controls

Significant linkage disequilibrium was found among allpairs of GSTs gene. A total of 11 double combinations wereobserved in the participants. Combinations of these poly-morphisms were also evaluated (Table 4). There was astatistically significant multiple interaction betweenGSTM1, GSTT1, and GSTP1 genotypes in risk of bladdercancer (P for interaction � 0.02). The frequency of theGSTM1 positive � GSTP1 Ile/Ile or Val/Val combination,was higher in patients (45.2%) compared with controls(33.7%; OR � 3.71, 95% CI: 2.34–5.54; P � 0.004; Table 4).A comparison between individuals with GSTT1 positive �GSTP1 Ile/Ile or Val/Val combination and those withGSTT1 positive � GSTP1 Ile/Ile combination indicated anOR of 2.66 (95% CI: 1.45–4.72; P � 0.007). The combi-nation of GSTM1 null genotype with GSTP1 Ile/Val orVal/Val genotype, also resulted in 4.76-fold increased riskfor bladder TCC (95% CI: 2.68–8.72; P � 0.002). Theopposite was the case for GSTP1 Ile/Ile genotype. Thecombination with GSTP1 Ile/Ile genotype was associatedwith a 66% lower decreased risk of bladder TCC forGSTM1 null (OR � 0.44; 95% CI: 0.23–0.74; P � 0.001),and a 35% lower decreased risk of bladder TCC for GSTT1null (OR � 0.65; 95% CI: 0.42–0.84; P � 0.007) (for

ients with bladder cancer and healthy controls

es (n � 166) (%) Adjusted ORa (95% CI) P value

(69.9) 1.0 (referent)(30.1) 1.32 (0.82–2.62) 0.67

(78.9) 1.0 (referent)(21.1) 1.18 (0.79–1.76) 0.74

(32.5) 1.0 (referent)(53.0) 2.32 (1.24–3.65) 0.007(14.5) 4.32 (2.64–6.34) 0.001(67.5) 3.48 (1.34–5.61) 0.002

ge, BMI, occupational status, educational level, smoking status and GST

the pat

Cas

11650

13135

548824

112

uding a

details see Table 4).

Page 6: Association of genetic polymorphism of glutathione S-transferase (GSTM1, GSTT1, GSTP1) with bladder cancer susceptibility

1198 M.R. Safarinejad et al. / Urologic Oncology: Seminars and Original Investigations 31 (2013) 1193–1203

3.4. Distribution of triple GST. genotypes among cancerpatients and controls

Since none of the null genotypes alone seem to representa significant risk factor, associations between the mutantgenotypes were also evaluated. Individuals with a combinedgenotype GSTM1 positive/GSTT1 positive/GSTP1 Ile/Valor Ile/Val exhibited an elevated risk for bladder cancer withan OR of 2.2 (95% CI 1.10 � 4.57 and 1.12 � 4.20,respectively). The combination with highest risk wasGSTM1 null/GSTT1 null/GSTP1 Ile/Val or Val/Val (OR �6.42, 95% CI: 4.76–14.72; P � 0.0001). Any combinationwith GSTP1 Ile/Ile, irrespective of GSTM1 and GSTT1genotype, resulted in a decreased risk for bladder TCC(Table 5). Lowest risk was among subjects with GSTM1present/GSTT1 null/GSTP1Ile/Ile (OR � 0.66, 95% CI:0.47–0.79; P � 0.02).

3.5. Association of GSTs genotypes with tumorstage/grade

After stratification for grade and stage, neither was theGSTM1 null genotype frequency statistically different be-

Table 4Distribution of double GST genotypes among cancer patients and control

Double GST genotypes Cases (n � 166) (%)

GSTM1 and GSTT1Both present 79 (47.6)Either Null 49 (29.5)Both null 38 (22.9)

GSTM1 and GSTP1M1(�/�) and P1(Ile/Ile) 41 (24.7)M1(�/�) and P1(Ile/val or Val/Val) 75 (45.2)M1(–/–) and P1(Ile/Ile) 13 (7.8)M1(–/–) and P1(Ile/Val or Val/Val) 37 (22.3)

GSTT1 and GSTP1T1(�/�) and P1(Ile/Ile) 43 (25.9)T1(�/�) and P1(Ile/Val or Val/Val) 88 (53.0)T1(–/–) and P1(Ile/Ile) 11 (6.6)T1(–/–) and P1(Ile/Val or Val/Val) 24 (14.5)

a Adjusted OR: adjusted in multivariate logistic regression models inclgenotypes.

Table 5Distribution of triple GST genotypes among cancer patients and controls

Triple GST genotypes Cases (n � 166)

M1 (�/�) and T1(�/�) and P1(Ile/Ile) 22 (13.3)M1 (�/�) and T1(�/�) and P1(Ile/Val or Val/Val) 74 (44.6)M1(–/–), T1(�/�), and P1(Ile/Ile) 12 (7.2)M1(–/–), T1(�/�), and P1(lIe/Val or Val/Val) 23 (13.9)M1(�/�), T1(–/–), and P1(Ile/Ile) 18 (10.8)M1(�/�), T1(–/–), and P1(Ile/Val or Val/Val) 2 (1.2)M1(–/–), T1(–/–), and P1(Ile/Ile) 2 (1.2)M1(–/–), T1(–/–), and P1(Ile/Val or Val/Val) 13 (7.8)

a Adjusted OR: adjusted in multivariate logistic regression models incl

genotypes.

tween lower and higher grade and stage of diseases (Fig. 3).Similarly to GSTM1 null genotype, the frequency of theGSTT1 null genotype did not significantly differ betweenvarious subgroups of cases (Fig. 3). The GSTP1 Val/Valgenotype was observed to be associated significantly withan increased risk of having high-grade (OR � 7.68, 95% CI:4.73–19.25; P � 0.0001) and invasive (OR � 10.67, 95%CI: 6.34–21.75; P � 0.0001) tumors of bladder cancer(Table 6). Individuals with both null genotypes showed asignificantly increased risk of developing high grade (OR �7.48, 95% CI: 4.58–14.62; P � 0.0001), and muscle inva-sive (OR � 6.84, 95% CI: 4.45–12.18; P � 0.0001) tumorsof the bladder (Table 7).

GSTM1 present/GSTP1 Ile/Val or Val/Val, and GSTT1present/GSTP1 Ile/Val or Val/Val patients had significantlylower risk for high grade (OR � 0.21, 95% CI: 0.14–0.37;P � 0.0001, and OR � 0.51, 95% CI: 0.32–0.77; P �0.0006, respectively), and invasive (OR � 0.27, 95% CI:0.18–0.47; P � 0.0001, and OR � 0.47, 95% CI: 0.36–0.72; P � 0.001, respectively) bladder TCC; a similarassociation was also found for GSTM1 null/GSTP1 Ile/Ile, and GSTT1 null/GSTP1 Ile/Ile patients (for detailssee Table 7).

ls (n � 332) (%) Odds ratioa (OR) 95% CI P value

.7) 1.0 (referent)

.3) 1.49 0.81–2.34 0.66

.0) 1.16 0.84–1.76 0.79

.3) 1.0 (referent)

.7) 3.71 2.34–5.54 0.004

.5) 0.44 0.23–0.74 0.001

.5) 4.76 2.68–8.72 0.002

.6) 1.0 (referent)

.7) 2.66 1.45–4.72 0.007

.2) 0.65 0.42–0.84 0.007

.5) 3.88 2.86–5.82 0.001

ge, BMI, occupational status, educational level, smoking status and GST

Controls (n � 332) (%) Odds ratioa (OR) 95% CI P value

95 (28.6) 1.0 (referent)106 (31.9) 3.48 1.64–5.63 0.007

31 (9.3) 0.74 0.58–2.32 0.6331 (9.3) 4.36 2.57–7.65 0.00225 (15.1) 0.66 0.47–0.79 0.0213 (3.9) 0.86 0.74–2.47 0.2321 (6.3) 0.82 0.71–2.76 0.4510 (3.0) 6.42 4.76–14.72 0.0001

ge, BMI, occupational status, educational level, smoking status and GST

s

Contro

165 (4994 (2873 (22

127 (38112 (33

45 (1348 (14

138 (41125 (37

34 (1035 (10

uding a

(%)

uding a

Page 7: Association of genetic polymorphism of glutathione S-transferase (GSTM1, GSTT1, GSTP1) with bladder cancer susceptibility

wGe

GGaa

3p

cops

le6

just

edod

dsra

tios

for

rela

tion

betw

een

GST

geno

type

san

ddi

ffer

ent

grad

esan

dcl

inic

alst

ages

atdi

agno

sis

ofbl

adde

rca

ncer

ymor

phis

mG

rade

no.

(%)

OR

a(9

5%C

I)(n

�59

)P

valu

eC

linic

alst

ages

no.(

%)

OR

a(9

5%C

I)P

valu

e

Low

grad

eH

igh

grad

e(n

�10

7)Su

perfi

cial

(n�

119)

Inva

sive

(n�

47)

TM

1re

sent

95(8

1.9)

21(1

8.1)

1.0

(Ref

eren

t)83

(71.

6)33

(28.

4)1.

0(R

efer

ent)

ull

24(4

8.0)

26(5

2.0)

1.83

(0.6

5–2.

47)

0.64

24(4

8.0)

26(5

2.0)

1.82

(0.6

7–2.

57)

0.66

TT

1re

sent

103

(78.

6)28

(21.

4)1.

0(R

efer

ent)

90(6

8.7)

41(3

1.3)

1.0

(Ref

eren

t)ul

l16

(45.

7)19

(54.

3)1.

77(0

.62–

2.72

)0.

0817

(48.

6)18

(51.

4)1.

81(0

.71–

2.42

)0.

56T

P1 le/I

le49

(90.

7)5

(9.3

)1.

0(r

efer

ent)

40(7

4.1)

14(2

5.9)

1.0

(ref

eren

t)le

/Val

63(7

1.6)

25(2

8.4)

0.42

(0.2

7–0.

61)

0.00

0165

(73.

9)23

(26.

1)0.

40(0

.18–

0.62

)0.

0001

al/V

al7

(29.

2)17

(70.

8)7.

68(4

.73–

9.25

)0.

0001

2(8

.3)

22(9

1.7)

10.6

7(6

.34–

21.7

5)0.

0001

le/V

alor

Val

/Val

70(6

2.5)

42(3

7.5)

0.62

(0.4

8–0.

79)

0.00

267

(59.

8)45

(40.

2)0.

72(0

.51–

0.87

)0.

002

Adj

uste

dO

R:

adju

sted

inm

ultiv

aria

telo

gist

icre

gres

sion

mod

els

incl

udin

gag

e,B

MI,

occu

patio

nal

stat

us,

educ

atio

nal

leve

l,sm

okin

gst

atus

and

GST

geno

type

s.

1199M.R. Safarinejad et al. / Urologic Oncology: Seminars and Original Investigations 31 (2013) 1193–1203

Combinations of GSTP1, GSTM1, and GSTT1 showed asignificant linear gene-dosage relationship with grade (P fortrend � 0.001) and stage (P for trend � 0.001), withincrease in number of putative risk genotypes (GSTM1null, GSTT1 null, GSTP1 Ile/Val or Val/Val) (Tables 7,and 8).

Subjects carrying the GSTM1 positive/GSTT1 positive/GSTP1 Ile/Val or Val/Val combined genotype were pro-tected against high grade (OR � 0.35, 95% CI � 0.22–0.64; P � 0.0001) and muscle invasive (OR � 0.44, 95%CI � 0.23–0.67; P � 0.0005) bladder cancer development(Table 8). After stratification for grade and stage, a statis-tically significant increased risk of invasive (OR � 8.62,95% CI � 6.41–17.39; P � 0.0001) and high-grade (OR �8.21, 95% CI � 6.37–17.34; P � 0.0001) bladder cancer

as found for carries of the GSTM1 null/GSTT1 null/STP1 Ile/Val or Val/Val genotype, compared with refer-

nt genotype (Table 8).Due to small numbers of subjects in GSTM1 present/

STT1 null/GSTP1 Ile/Val or Val/Val, and GSTM1 null/STT1 null/GSTP1 Ile/Ile, category, we were unable to

nalyze the association between these combined genotypesnd tumor behaviors.

.6. Association of GSTM1, GSTT1, and GSTP1olymorphisms with smoking status

The median number of smoking pack-years among theases and controls was 14.7 and 14.1, respectively. More-ver, as with age and BMI, none of the 3 GST polymor-hisms was correlated with the number of pack-yearsmoked (all 3 Kruskal-Wallis test P values � 0.34),

indicating that the differences in smoking history be-tween cancer cases and controls cannot confound ourgenotypic association results. This finding was furtherconfirmed by multivariable logistic regression analyses

Fig. 3. Distribution of GSTM1 null, GSTT1 null, GSTP1 Ile/Val, andGDTP1 Val/Val genotypes in different grade and stage of bladder tumors.

(data not shown). The GSTP1 Val/Val genotype showed

Tab

Ad

Pol

GS P N

GS P N

GS I I V I a

Page 8: Association of genetic polymorphism of glutathione S-transferase (GSTM1, GSTT1, GSTP1) with bladder cancer susceptibility

Table 7Adjusted odds ratios for relation between double GST genotypes and different grades and clinical stages at diagnosis of bladder cancer

Double GST genotype Grade no. (%) ORa (95% CI) P value Clinical stages no. (%) ORa (95% CI) P value

Low grade (n � 119) High grade (n � 47) Superficial (n � 107) Invasive (n � 59)

GSTM1 and GSTT1Both present 74 (93.7%) 5 (6.3%) 1.0 (referent) 64 (81.0%) 15 (19.0%) 1.0 (referent)Either null 35 (71.4%) 14 (28.6%) 0.52 (0.26–0.75) 0.006 31 (63.3%) 18 (36.7%) 0.62 (0.31–0.83) 0.008Both null 10 (26.3%) 28 (73.7%) 7.48 (4.58–14.62) 0.0001 12 (31.6%) 26 (68.4%) 6.84 (4.45–12.18) 0.0001

GSTM1 and GSTP1M1(�/�) and P1(Ile/Ile) 36 (78.8%) 5 (12.2%) 1.0 (referent) 26 (63.4%) 15 (36.6%) 1.0 (referent)M1(�/�) and P1(Ile/val or Val/Val) 65 (86.7%) 10 (13.3%) 0.21 (0.14–0.37) 0.0001 63 (84.0%) 12 (16.0%) 0.27 (0.18–0.47) 0.0001M1(–/–) and P1(Ile/Ile) 8 (61.5%) 5 (38.5%) 0.58 (0.34–0.79) 0.004 9 (69.2%) 4 (30.8%) 0.38 (0.18–0.72) 0.002M1(–/–) and P1(Ile/Val or Val/Val) 10 (27.0%) 27 (73.0%) 7.32 (4.47–14.22) 0.0001 9 (24.3%) 28 (75.7%) 7.72 (6.64–15.11) 0.0001

GSTT1 and GSTP1T1(�/�) and P1(Ile/Ile) 37 (86.0%) 6 (14.0%) 1.0 (referent) 27 (62.8%) 16 (37.2%) 1.0 (referent)T1(�/�) and P1(Ile/Val or Val/Val) 68 (77.3%) 20 (22.7%) 0.51 (0.36–0.72) 0.001 66 (75.0%) 22 (25.0%) 0.47 (0.32–0.77) 0.0006T1(–/–) and P1(Ile/Ile) 6 (54.5%) 5 (45.5%) 0.84 (0.65–2.84) 0.08 7 (63.6%) 4 (36.4%) 0.41 (0.37–0.78) 0.003T1(–/–) and P1(Ile/Val or Val/Val) 8 (33.3%) 16 (66.7%) 4.82 (2.56–9.76) 0.001 7 (29.2%) 17 (70.8%) 7.15 (5.87–14.82) 0.0002

a Adjusted OR: adjusted in multivariate logistic regression models including age, BMI, occupational status, educational level, smoking status and GST genotypes.

Table 8Adjusted odds ratios for relation between triple GST genotypes and different grades and clinical stages at diagnosis of bladder cancer

Triple GST genotype Grade no. (%) ORa (95% CI) P value Clinical stages no. (%) ORa (95% CI) P value

Low grade(n � 119)

High grade(n � 47)

Superficial(n � 107)

Invasive(n � 59)

M1 (�/�) and T1(�/�) and P1(Ile/Ile) 20 (90.9) 2 (9.1) 1.0 (referent) 15 (68.2) 7 (31.8) 1.0 (referent)M1 (�/�) and T1(�/�) and P1(Ile/Val or Val/Val) 60 (81.1) 14 (18.9) 0.35 (0.22–0.64) 0.0001 53 (71.6) 21 (28.4) 0.44 (0.23–0.67) 0.0005M1(–/–), T1(�/�), and P1(Ile/Ile) 8 (66.7) 4 (33.3) 0.54 (0.36–0.86) 0.008 9 (75.0) 3 (25.0) 0.37 (0.21–0.76) 0.002M1(–/–), T1(�/�), and P1(lIe/Val or Val/Val) 11 (47.8) 12 (52.2) 1.82 (0.84–2.47) 0.24 10 (43.5) 13 (56.5) 1.82 (0.87–2.61) 0.27M1(�/�), T1(–/–), and P1(Ile/Ile) 13 (72.2) 5 (27.8) 0.42 (0.37–0.79) 0.003 13 (72.2) 5 (27.8) 0.39 (0.26–0.77) 0.003M1(�/�), T1(–/–), and P1(Ile/Val or Val/Val) 2 (100.0) 0 (0.0) NA 2 (100.0) 0 (0.0) NAM1(–/–), T1(–/–), and P1(Ile/Ile) 2 (100.0) 0 (0.0) NA 2 (100.0) 0 (0.0) NAM1(–/–), T1(–/–), and P1(Ile/Val or Val/Val) 3 (23.1) 10 (76.9) 8.21 (6.37–17.34) 0.0001 3 (23.1) 10 (76.9) 8.62 (6.41–17.39) 0.0001

a Adjusted OR: adjusted in multivariate logistic regression models including age, BMI, occupational status, educational level, smoking status and GST genotypes.

1200M

.R.

Safarinejadet

al./

Urologic

Oncology:

Seminars

andO

riginalInvestigations

31(2013)

1193–1203

Page 9: Association of genetic polymorphism of glutathione S-transferase (GSTM1, GSTT1, GSTP1) with bladder cancer susceptibility

1201M.R. Safarinejad et al. / Urologic Oncology: Seminars and Original Investigations 31 (2013) 1193–1203

an increased risk among non-smokers (OR � 3.22, 95%CI � 1.76 –5.37; P � 0.016), smokers (OR � 5.72, 95%CI � 3.28 –7.36; P � 0.003), former smokers (OR �6.14, 95% CI � 4.26 – 8.57; P � 0.001) and currentsmokers (OR � 4.34, 95% CI � 2.41– 6.76; P � 0.002).No association of GSTT1 and GSTM1 polymorphismswas found with smoking. In particular, light smokers(�20 pack-years) had a 3.17-fold (95% CI � 1.82– 4.79)and heavy smokers (�20 pack-years) had a 4.76-fold(95% CI � 2.59 – 6.82) increased risk, compared withnonsmokers.

4. Discussion

We detected no association between GSTM1 and GSTT1and risk for bladder cancer; however, the GSTP1 Ile/Val orVal/Val genotypes were associated with a significantly in-creased risk. In our study population, the bladder TCC riskwas found to increase 4.3-fold in individuals with the ho-mozygous GSTP1 Val/Val variant genotype. Our finding isconsistent with studies of bladder cancer conducted in In-dian [27], Turkish [26], and British population [30]. Con-versely, Steinhoff et al. [31] in the German population andKatoh et al. [32] in the Japanese population found no asso-ciation for GSTP1 Ile/Val or Val/Val genotype with sus-ceptibility to bladder cancer. Also, significant associationwas found between the GSTP1 Val/Val genotype and thegrade and stage of cancer with an OR of 7.68 and 10.67,respectively. In the present study, we did not observe sig-nificant increased risk for bladder cancer with GSTT1 nullgenotypes compared with the controls. This finding agreeswith previous studies [27,31]; nonetheless, in studies per-formed in Slovakian and Egyptian [21,33] populations, sta-tistically significant risk of bladder cancer for GSTT1 nullgenotype was reported. Two meta-analyses demonstratedthat the GSTM1 null genotype associated with an overall40%–50% increased risk for developing bladder cancer risk[34,35]. Decreased risk for bladder cancer has also beenreported with GSTT1 null genotype [19]. Results pertainingto the GSTT1 polymorphism and the risk of bladder cancerare less consistent [33,36]. Salagovic et al. [21] and Lee etal. [22] found an increased risk of bladder cancer with theGSTT1 null genotype. Kempkes et al. [20] and Brockmolleret al. [19] reported a significantly increased risk of bladdercancer associated with GSTT1 null genotype in nonsmok-ers. In contrast, Sobti et al. demonstrated significant asso-ciation between GSTT1 null genotype as a risk to bladdercancer in the light smoker group with an OR of 2.19 [17]. Inthe most recent meta analysis, Zeng et al. suggest thatGSTT1 null status is associated with a modest increase inthe risk of bladder cancer [37].

We also found a nonsignificant risk for the GSTM1 nullgenotype. Our results concur with the observations made byKim et al. in the Korean population [38], and by Srivastava

et al. in the Indian population [27]. Conversely, several

other studies reported significant association of GSTM1 inbladder cancer [21,31].

Bladder TCC with different grades and stages may rep-resent entities with different etiology and prognosis, anddisparities in the tumor molecular genetics [39]. In thecurrent study, the results did not show a significant associ-ation between GSTT1 and GSTM1 polymorphisms andgrade and stage of the cancer. Our findings concur with theprevious studies that showed nonsignificant association ofGSTM1 genotypes with grade of the disease [27,40]. How-ever, when we analyzed the relation between double GSTgenotypes and different grade and clinical stages, we founda statistically significant association between both GSTM1/GSTT1 null genotype variant.

Effect modification by smoking status has been exam-ined in some studies, but with conflicting results [21,41].Our study revealed that the effects of GSTM1 and GSTT1polymorphisms in developing bladder cancer are indepen-dent of smoking status. This finding was in agreement with2 German studies [20,42], but disagrees with finding ofSobti et al. in the Indian population [17].

Data from our study do not support a substantial as-sociation between GSTM1 null and GSTT1 null geno-types and bladder cancer risk. One possible explanationis that the effect of the GSTM1 null and GSTT1 nullgenotypes may be detectable only in combination withother “at-risk” genotypes. Confirmation of this would,however, have required much larger study groups thanthe present ones.

When GSTM1 null and GSTP1 Ile/Val or Val/Val ge-notype were present together, risk increased by 4.76-fold incomparison to referent group. Cases with GSTP1 Ile/Val orVal/Val genotype along with GSTT1 null genotype were at3.88-times more risk of having TCC of bladder. This issuggestive of a synergistic function only between GSTM1null and GSTT1 null and GSTP1 Ile/Val genotype in theetiology of bladder cancer. Of importance, when we com-bined the 3-risk genotypes, risk increased more than 6-fold.

Some studies also evaluated the combination effects ofrisk genotypes (GSTM1 null, GSTT1 null, GSTP1 Ile/Valor Val/Val), and significant results could be found in mostof the studies [26,31,43]. The present study indicated esca-lation of risk with multiple risk alleles of GSTs. This findingsuggests that gene–gene interactions may be partially in-volved in genetic susceptibility for bladder cancer, whichcould be explained by various substrates used by differentGSTs inducing resulting in combined action.

We also attempted to examine the correlation betweenclinical stage and/or pathologic grade with GSTs genotypesfor the risk of bladder cancer, but no association was foundbetween GSTM1 null and GATT1 null genotypes (Figure).This finding agrees with previous study by Srivastava et al.[27]. In contrast, our findings showed a significant associ-ation of GSTM1/GSTT1 double null genotypes with tumorgrade and stage of bladder cancer. However, there is dis-

agreement in the published literature about this finding [27].
Page 10: Association of genetic polymorphism of glutathione S-transferase (GSTM1, GSTT1, GSTP1) with bladder cancer susceptibility

[

[

[

[

[

[

[

[

[

[

1202 M.R. Safarinejad et al. / Urologic Oncology: Seminars and Original Investigations 31 (2013) 1193–1203

The prevalence of GSTM1 and GSTT1 null genotypes varysignificantly among ethnic groups. The reported frequency ofthe GSTM1-null genotype is around 50% of the Caucasianpopulation in Europe [44,45]. In the present study, the fre-quency of the GSTM1-null genotype in the Iranian populationwas 28%. Therefore, the frequency of the homozygotic dele-tion of the gene (GSTM1 null) in the Iranian population ismuch lower than in all the European Caucasian population.The GSTT1 null allele is one of the most widely studied GSTpolymorphisms with a frequency of about 10%–20% in Cau-casians and 50%–60% in Asians [46]. The GSTP1 polymor-phism had strongest associations with bladder cancer. Thefrequency of GSTP1 genotypes in our controls is comparableto other larger studies with Caucasian populations [31,47],which demonstrate that potential selection bias may be relatedwith lifestyle factors, but not polymorphisms of GSTP1. Afteradjustment for disease stage or grade, GSTP1 Ile/Val genotypealone conferred protective effects against high-grade and high-stage tumor. The chance of having high-grade and muscleinvasive tumors decreases about 60%, in individuals harboringGSTP1 Ile/Val genotype.

In the current study, we only recruited male patients. Aninteraction between gender, the GSTM1 variant, smoking, andlung cancer has been reported [48]. With respect to genderdifferences, a hospital-based study from Germany of 157 blad-der cancer cases and 223 controls reported that the GSTT1polymorphism was unrelated to bladder cancer risk in men andnonsignificantly inversely related in women. However, gender-specific effects were not observed in the meta- and pooled-analysis of GSTM1 genotype and bladder cancer risk [49].

Our study has some limitations. Of risk factors for bladderTCC, we only addressed the smoking history. Because smok-ing is a well known risk factor for bladder cancer, we madeevery effort to match both groups regarding smoking history.Control subjects did not undergo any investigation regardingpresence of bladder cancer. Therefore, some of them (espe-cially smoker men) might have carcinoma in situ or bladdercarcinoma when recruited into study, and perhaps some ofthem will develop bladder cancer in the future. Despite thefairly large number of subjects included in the study, thenumbers were still regrettably small in some of the subgroupanalyses. In addition, multiple comparisons may cause a ten-dency towards findings by chance alone. Finally, since thefindings from the present study were only from an Iranianpopulation, it is uncertain whether these results are relevant toother ethnic groups.

5. Conclusion

The results obtained in the present study show that individ-uals with a particular combination of genotypes (GSTM1 null,GSTT1 null, and GSTP1 Ile/Val or Val/Val) have a signifi-cantly increased risk for bladder TCC, particularly for high-grade and muscle-invasive tumors. The interesting findings of

this study needs confirmation from larger studies.

Acknowledgments

The authors are indebted to the participants.

References

[1] Ferlay J, Bray F, Pisani P, et al. GLOBOCAN 2000: Cancer Inci-dence, Mortality, and Prevalence Worldwide. Ver. 1.0. IARC CancerBase No. 5. Lyon (France): IARC Press, 2001.

[2] Cohen SM, Shirai T, Steineck G. Epidemiology and etiology ofpremalignant and malignant urothelial changes. Scand J Urol Nephrol2000;(Suppl):105–15.

[3] Stern MC, Johnson LR, Bell DA, et al. XPD Codon 751 polymor-phism, metabolism genes, smoking, and bladder cancer risk. CancerEpidemiol Biomarkers Prev 2002;11:1004–11.

[4] Sanyal S, Festa F, Sakano S, et al. Polymorphisms in DNA repair andmetabolic genes in bladder cancer. Carcinogenesis 2004;25:729–34.

[5] Schnakenberg E, Breuer R, Werdin R, et al. Susceptibility genes:GSTM1 and GSTM3 as genetic risk factors in bladder cancer. Cyto-genet Cell Genet 2000;91:234–8.

[6] Lo HW, Ali-Osman F. Genetic polymorphism and function of gluta-thione S-transferases in tumor drug resistance. Curr Opin Pharmacol2007;7:367–74.

[7] Strange RC, Spiteri MA, Ramachandran S, et al. Glutathione-S-transferase family of enzymes. Mutat Res 2001;482:21–6.

[8] Hoffmann D, Hecht SS. Advances in tobacco carcinogenesis, In:Cooper CS, Grover PL, editors. Handbook of Experimental Pharma-cology. Heidelberg: Springer-Verlag, 1990. p. 63–102.

[9] Landi S. Mammalian class � GST and differential susceptibility tocarcinogens: A review. Mutat Res 2000;463:247–83.

10] Benhamou S, Lee WJ, Alexandrie AK, et al. Meta- and pooledanalyses of the effects of glutathione S-transferase M1 polymor-phisms and smoking on lung cancer risk. Carcinogenesis 2002;23:1343–50.

11] Singh M, Shah PP, Singh AP, et al. Association of genetic polymor-phisms in glutathione S-transferases and susceptibility to head andneck cancer. Mutat Res 2008;638:184–94.

12] Smits KM, Gaspari L, Weijenberg MP, et al. Interaction betweensmoking, GSTM1 deletion, and colorectal cancer: Results from theGSEC study. Biomarkers 2003;8:299–310.

13] Ates NA, Tamer L, Ates C, et al. Glutathione S transferase M1, T1,P1 genotypes and risk for development of colorectal cancer. BiochemGenet 2005;43:149–63.

14] Srivastava DS, Kumar A, Mittal B, et al. Polymorphism of GSTM1and GSTT1 genes in bladder cancer: A study from North India. ArchToxicol 2004;78:430–4.

15] Katoh T, Inatomi H, Kim H, et al. Effects of glutathione S-transferase(GST) M1 and GSTT1 genotypes on urothelial cancer risk. CancerLett 1998;132:147–52.

16] Shao J, Gu M, Zhang Z, et al. Genetic variants of the cytochromeP450 and glutathione S-transferase associated with risk of bladdercancer in a south-eastern Chinese population. Int J Urol 2008;15:216–21.

17] Sobti RC, Al-Badran AI, Sharma S, et al. Genetic polymorphisms ofCYP2D6, GSTM1, and GSTT1 genes and bladder cancer risk inNorth India. Cancer Genet Cytogenet 2005;156:68–73.

18] Pemble S, Schroeder KR, Spencer SR, et al. Human glutathioneS-transferase � (GSTT1): cDNA cloning and the characterization of agenetic polymorphism. Biochem J 1994;300:271–6.

19] Brockmoller J, Cascorbi I, Kerb R, et al. Combined analysis ofinherited polymorphism in arylamine N-acetyltransferases, glutathi-one S-transferases M1 and T1, microsomal epoxide hydrolase, andCYP 450 enzyme as modulator of bladder cancer risk. Cancer Res

1996;56:3915–25.
Page 11: Association of genetic polymorphism of glutathione S-transferase (GSTM1, GSTT1, GSTP1) with bladder cancer susceptibility

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

1203M.R. Safarinejad et al. / Urologic Oncology: Seminars and Original Investigations 31 (2013) 1193–1203

[20] Kempkes M, Golka K, Reich S, et al. Glutathione S-transferaseGSTM1 and STT1 null genotypes as potential risk factors for urothe-lial cancer of the bladder. Arch Toxicol 1996;71:123–6.

[21] Salagovic J, Kalina I, Stubna J, et al. Genetic polymorphism ofglutathione S-transferase M1 and T1 and risk factor in lung andbladder cancer. Neoplasma 1998;45:312–7.

[22] Lee SJ, Cho SH, Park SK, et al. Association of genetic polymorphismof glutathione S-transferase M1 and T1 and bladder cancer. J KoreanCancer Assoc 1999;31:548–55.

[23] Bell DA, Taylor JA, Paulson DF, et al. Genetic risk and carcinogenexposure: A common inherited defect of the carcinogen-metabolismgene glutathione S-transferase M1 (GSTM1) that increases susceptibilityto bladder cancer. J Natl Cancer Inst 1993;85:1159–64.

24] Lafuente A, Pujol F, Carretero P, et al. Human glutathione S-trans-ferase m (GST m) deficiency as a marker for the susceptibility tobladder and larynx cancer among smokers. Cancer Lett 1993;68:49 –54.

25] Ali-Osman F, Akande N, Mao J. Molecular cloning, characterization,and expression in Escherichia coli of full-length cDNA of threehuman glutathione S-transferase � gene variants. Evidence for dif-ferential catalytic activity of the encoded proteins. J Biol Chem1997;272:10004–12.

26] Törüner GA, Akyerli C, Uçar A, et al. Polymorphism of glutathioneS-transferase genes (GSTM1, GSTP1, and GSTT1) and bladder can-cer susceptibility in the Turkish population. Arch Toxicol 2001;75:459–64.

27] Srivastava DS, Mishra DK, Mandhani A, et al. Association of geneticpolymorphism of glutathione S-transferase M1, T1, P1, and suscep-tibility to bladder cancer. Eur Urol 2005;48:339–44.

28] Safarinejad MR, Shafiei N, Safarinejad S. The association of gluta-thione-S-transferase gene polymorphisms (GSTM1, GSTT1, GSTP1)with idiopathic male infertility. J Hum Genet 2010;55:565–70.

29] Liu YJ, Huang PL, Chang YF, et al. GSTP1 genetic polymorphism isassociated with a higher risk of DNA damage in pesticide exposedfruit growers. Cancer Epidemiol Biomarkers Prev 2006;15:659–66.

30] Harries LW, Stubbins MJ, Forman D, et al. Identification of geneticpolymorphisms at the glutathione S-transferase � locus and associa-tion with susceptibility to bladder, testicular, and prostate cancer.Carcinogenesis 1997;18:641–4.

31] Steinhoff C, Franke KH, Golka K, et al. Glutathione transferaseisozyme genotypes in patients with prostate and bladder carcinoma.Arch Toxicol 2000;74:521–6.

32] Katoh T, Kaneko S, Takasawa S, et al. Human glutathione S-trans-ferase P1 polymorphism and susceptibility to smoking related epi-thelial cancer, oral, lung, gastric, colorectal, and urothelial cancer.Pharmacogenetics 1999;9:165–9.

33] Abdel-Rahman SZ, Anwar WA, Abdel-Aal, et al.GSTM1 and GSTT1genes are potential risk modifiers for bladder cancer. Cancer DetectPrev 1998;22:129–38.

34] García-Closas M, Malats N, Silverman D, et al. NAT2 slow acety-

lation, GSTM1 null genotype, and risk of bladder cancer: Results

from the Spanish Bladder Cancer Study and meta-analyses. Lancet2005;366:649–59.

35] Johns LE, Houlston RS. Glutathione S-transferase �1 (GSTM1) sta-tus and bladder cancer risk: A meta-analysis. Mutagenesis 2000;15:399–404.

36] Guengerich FP, Their R, Persmark M, et al. Conjugation of carcin-ogen by � class glutathione S-transferase; mechanism and relevanceto variations in human risk. Pharmacogenetics 1995;5:103–7.

37] Zeng FF, Liu SY, Wei W, et al. Genetic polymorphisms of glutathi-one S-transferase T1 and bladder cancer risk: A meta-analysis. ClinExp Med 2010;10:59–68.

38] Kim WJ, Kim H, Kim CH, et al. GSTT1-null genotype is a protectivefactor against bladder cancer. Urology 2002;60:913–8.

39] Brockmoller J, Kerb R, Drakoulis N, et al. Glutathione S-transferaseM1 and its variants A and B as host factors of bladder cancersusceptibility: A case-control study. Cancer Res 1994;54:4103–11.

40] Georgiou I, Filiadis IF, Alamanos Y, et al. Glutathione S-transferasenull genotypes in transitional cell bladder cancer: A case-controlstudy. Eur Urol 2000;37:660–4.

41] Schnakenberg E, Lustig M, Breuer R, et al. Gender-specific effects ofNAT2 and GSTM1 in bladder cancer. Clin Genet 2000;57:270–7.

42] Anwar WA, Abdel-Rahman SZ, El-Zein RA, et al. Genetic polymor-phisms of GSTM1, CYP2E1, and CYP2D6 in Egyptian bladdercancer patients. Carcinogenesis 1996;17:1923–9.

43] Hung RJ, Boffetta P, Brennan P, et al. GST, NAT, SULT1A1,CYP1B1 genetic polymorphisms, interactions with environmentalexposures and bladder cancer risk in a high-risk population. Int JCancer 2004;110:598–604.

44] Lafunte A, Pujol F, Carretero P, et al. Human glutathione S-trans-ferase m (GST�) deficiency as a marker for the susceptibility tobladder and larynx cancer among smokers. Cancer Lett 1993;68:49 –54.

45] Gabbani G, Hou SM, Nardini B, et al. GSTM1 and NAT2 genotypesand urinary mutagens in coke oven workers. Carcinogenesis 1996;17:1677–81.

46] Nelson HH, Wiencke JK, Christiani DC, et al. Ethnic differences inthe prevalence of the homozygous deleted genotype of glutathioneS-transferase �. Carcinogenesis 1995;16:1243–5.

[47] Shepard TF, Platz EA, Kantoff PW, et al. No association between theI105V polymorphism of the glutathione S-transferase P1 gene(GSTP1) and prostate cancer risk: A prospective study. Cancer Epi-demiol Biomarkers Prev 2000;9:1267–318.

[48] Tang DL, Rundle A, Warburton D, et al. Associations between bothgenetic and environmental biomarkers and lung cancer: Evidence ofa greater risk of lung cancer in women smokers. Carcinogenesis1998;19:1949–53.

[49] Engel LS, Taioli E, Pfeiffer R, et al. Pooled analysis and meta-analysis of glutathione S-transferase M1 and bladder cancer: A HuGE

review. Am J Epidemiol 2002;156:95–109.

Recommended