toxicogenetic profile and cancer risk in lebanese

32
This article was downloaded by: [University of Windsor] On: 14 November 2014, At: 18:48 Publisher: Taylor & Francis Informa Ltd Registered in England and Wales Registered Number: 1072954 Registered office: Mortimer House, 37-41 Mortimer Street, London W1T 3JH, UK Journal of Toxicology and Environmental Health, Part B: Critical Reviews Publication details, including instructions for authors and subscription information: http://www.tandfonline.com/loi/uteb20 Toxicogenetic Profile and Cancer Risk in Lebanese Hassan R. Dhaini a & Loulou Kobeissi b a Faculty of Health Sciences, University of Balamand, Beirut, Lebanon b Mel & Enid Zuckerman College of Public Health, University of Arizona, Tucson, Arizona, USA Published online: 14 Mar 2014. To cite this article: Hassan R. Dhaini & Loulou Kobeissi (2014) Toxicogenetic Profile and Cancer Risk in Lebanese, Journal of Toxicology and Environmental Health, Part B: Critical Reviews, 17:2, 95-125, DOI: 10.1080/10937404.2013.878679 To link to this article: http://dx.doi.org/10.1080/10937404.2013.878679 PLEASE SCROLL DOWN FOR ARTICLE Taylor & Francis makes every effort to ensure the accuracy of all the information (the “Content”) contained in the publications on our platform. However, Taylor & Francis, our agents, and our licensors make no representations or warranties whatsoever as to the accuracy, completeness, or suitability for any purpose of the Content. Any opinions and views expressed in this publication are the opinions and views of the authors, and are not the views of or endorsed by Taylor & Francis. The accuracy of the Content should not be relied upon and should be independently verified with primary sources of information. Taylor and Francis shall not be liable for any losses, actions, claims, proceedings, demands, costs, expenses, damages, and other liabilities whatsoever or howsoever caused arising directly or indirectly in connection with, in relation to or arising out of the use of the Content. This article may be used for research, teaching, and private study purposes. Any substantial or systematic reproduction, redistribution, reselling, loan, sub-licensing, systematic supply, or distribution in any form to anyone is expressly forbidden. Terms & Conditions of access and use can be found at http:// www.tandfonline.com/page/terms-and-conditions

Upload: loulou

Post on 16-Mar-2017

223 views

Category:

Documents


4 download

TRANSCRIPT

This article was downloaded by: [University of Windsor]On: 14 November 2014, At: 18:48Publisher: Taylor & FrancisInforma Ltd Registered in England and Wales Registered Number: 1072954 Registered office: Mortimer House,37-41 Mortimer Street, London W1T 3JH, UK

Journal of Toxicology and Environmental Health, PartB: Critical ReviewsPublication details, including instructions for authors and subscription information:http://www.tandfonline.com/loi/uteb20

Toxicogenetic Profile and Cancer Risk in LebaneseHassan R. Dhainia & Loulou Kobeissiba Faculty of Health Sciences, University of Balamand, Beirut, Lebanonb Mel & Enid Zuckerman College of Public Health, University of Arizona, Tucson, Arizona,USAPublished online: 14 Mar 2014.

To cite this article: Hassan R. Dhaini & Loulou Kobeissi (2014) Toxicogenetic Profile and Cancer Risk in Lebanese, Journal ofToxicology and Environmental Health, Part B: Critical Reviews, 17:2, 95-125, DOI: 10.1080/10937404.2013.878679

To link to this article: http://dx.doi.org/10.1080/10937404.2013.878679

PLEASE SCROLL DOWN FOR ARTICLE

Taylor & Francis makes every effort to ensure the accuracy of all the information (the “Content”) containedin the publications on our platform. However, Taylor & Francis, our agents, and our licensors make norepresentations or warranties whatsoever as to the accuracy, completeness, or suitability for any purpose of theContent. Any opinions and views expressed in this publication are the opinions and views of the authors, andare not the views of or endorsed by Taylor & Francis. The accuracy of the Content should not be relied upon andshould be independently verified with primary sources of information. Taylor and Francis shall not be liable forany losses, actions, claims, proceedings, demands, costs, expenses, damages, and other liabilities whatsoeveror howsoever caused arising directly or indirectly in connection with, in relation to or arising out of the use ofthe Content.

This article may be used for research, teaching, and private study purposes. Any substantial or systematicreproduction, redistribution, reselling, loan, sub-licensing, systematic supply, or distribution in anyform to anyone is expressly forbidden. Terms & Conditions of access and use can be found at http://www.tandfonline.com/page/terms-and-conditions

Journal of Toxicology and Environmental Health, Part B, 17:95–125, 2014Copyright © Taylor & Francis Group, LLCISSN: 1093-7404 print / 1521-6950 onlineDOI: 10.1080/10937404.2013.878679

TOXICOGENETIC PROFILE AND CANCER RISK IN LEBANESE

Hassan R. Dhaini1, Loulou Kobeissi2

1Faculty of Health Sciences, University of Balamand, Beirut, Lebanon2Mel & Enid Zuckerman College of Public Health, University of Arizona, Tucson, Arizona, USA

An increasing number of genetic polymorphisms in drug-metabolizing enzymes (DME) wereidentified among different ethnic groups. Some of these polymorphisms are associated withan increased cancer risk, while others remain equivocal. However, there is sufficient evi-dence that these associations become significant in populations overexposed to environmentalcarcinogens. Hence, genetic differences in expression activity of both Phase I and Phase IIenzymes may affect cancer risk in exposed populations. In Lebanon, there has been a markedrise in reported cancer incidence since the 1990s. There are also indicators of exposure tounusually high levels of environmental pollutants and carcinogens in the country. This reviewconsiders this high cancer incidence by exploring a potential gene–environment model basedon available DME polymorphism prevalence, and their impact on bladder, colorectal, prostate,breast, and lung cancer in the Lebanese population. The examined DME include glutathioneS-transferases (GST), N-acetyltransferases (NAT), and cytochromes P-450 (CYP). Data suggestthat these DME influence bladder cancer risk in the Lebanese population. Evidence indi-cates that identification of a gene–environment interaction model may help in defining futureresearch priorities and preventive cancer control strategies in this country, particularly forbreast and lung cancer.

OVERVIEW OF CANCER INCIDENCEAND POTENTIAL RISK FACTORS INLEBANESE

Ever since the civil war ended in Lebanon in1990, the number and quality of medical insti-tutions have expanded and improved. Althougha national census has not been conducted since1932, the cancer registration process in thiscountry has evolved. The first data on cancerincidence in Lebanon were based on pathologyreports from select institutions, estimating,for all cancers combined, crude incidencerates of 102.8 and 104.1 per 100,000 inmales and females, respectively (Abou-Daoud,1966). Subsequently, hospital-based registriesstarted emerging. These efforts contributedto the generation of primary information per-taining to cancer incidence and management

Address correspondence to Hassan R. Dhaini, Faculty of Health Sciences, University of Balamand, PO Box 166378 Ashrafieh, Beirut,Lebanon. E-mail: [email protected]

in Lebanon. However, the reported resultsvaried between these registries for differenttypes of cancer (El Saghir et al., 1998; Adibet al., 1998). In 1998, LCEG, a network con-sisting of all hospitals with oncology special-ties and pathology labs in the country, waslaunched. LCEG reported in 1998, for all can-cers combined, overall crude incidence ratesof 141.4 and 126.8 per 100,000 among malesand females, respectively (Shamseddine et al.,2004). In 2005, the Lebanese Ministry of PublicHealth (MOPH) launched the National CancerRegistry (NCR), which published its first andonly report, “Cancer 2003,” in 2006 (Adiband Daniel, 2006). Later, NCR produced can-cer incidence summaries in select years (Adib,Daniel, and Issa, 2008; Lebanese Ministry ofPublic Health and Epidemiology Surveillance

95

Dow

nloa

ded

by [

Uni

vers

ity o

f W

inds

or]

at 1

8:48

14

Nov

embe

r 20

14

96 H. R. DHAINI AND L. KOBEISSI

Program, 2010). In a population of 3.9 mil-lion, 7,197 cases were diagnosed with cancer in2004. Of these, 50.1% of the cancer cases werereported among women, compared to 49.9%among men. The median age at diagnosis forwomen was 56 yr, compared to 63 yr for men.Cancer data of 2004 mark a sharp rise in cancerincidence for both sexes, with an approximate60% increase in the reported cases comparedto those of 1998. The 2004 age-standardizedrate (ASR) for all cancers combined was179.3 per 100,000 among males and 190.3 per100,000 among females (Shamseddine andMusallam, 2010). This increase in cancer inci-dence since 1966 suggests changes in lifestyleand in prevalence of risk factors, coupled withan increase in life expectancy and improvedcase reporting (Shamseddine et al., 2004).In 2004, breast cancer was the most commonlydiagnosed cancer in Lebanon (19.7%) repre-senting 1 in 5 for the entire caseload of allcancers, followed by lung (10.8%) and bladdercancer (9.3%) (Adib et al., 2008).

At the environmental level, Lebanon isan extensively urbanized country with heavytraffic and overcrowding in all its urban areas.Particulate concentrations in air were foundto exceed international standards. Air samplescollected from several locations in the capital,Beirut, revealed that total suspended particu-lates (TSP) levels are reaching up to 291 µg/m3.This exceeds long-term international referencestandards and guidelines on air particulateconcentrations (World Health Organization[WHO]: 60–90 µg/m3; European Union [EU]:150 µg/m3; U.S. Environmental ProtectionAgency [EPA]: 260 µg/m3) (El-Fadel andMassoud, 2000). At the same time, domesticdiesel power generators, compensating for longpower outages, are an important source ofoutdoor air pollution (Salameh et al., 2012).In addition, many reports indicated patterns ofdeterioration in water quality, as well as humanexposure to pollutants by air and throughfood (Houri and El Jeblawi, 2007; Massoudet al., 2010; Korfali and Jurdi, 2007). A studyconducted in Akkar, north of Lebanon, showedexcessive levels of nitrates in sources of drink-ing water reaching as high as 163 ppm (U.S.

EPA primary water quality standard: 10 ppm)(Halwani et al., 1999; U.S. EPA, 2009). In con-trast, soil samples in areas with industrial activ-ities were found to be rich in trace elements,exceeding North American permissible limits(Kassir et al., 2012; Massoud, 2012; CanadianCouncil of Ministers of the Environment[CCME], 1999). Mobile forms of heavy metalssuch as cadmium (Cd), chromium (Cr), zinc(Zn), and nickel (Ni) were identified as poten-tial contaminants of underground water (Kassiret al., 2012). Further, the Lebanese Ministry ofEnvironment reported that the largest industrialsectors in the country—leather, textile, furand clothes dyeing, wood, furniture, and foodindustries—may be polluting the environmentwith heavy metals, pesticides, and variousorganic compounds (Lebanese Ministry ofEnvironment, United Nations EnvironmentProgramme [UNEP], and Global EnvironmentalFacility [GEP], 2005). Pollution is mainly due tounregulated emissions, inappropriate practicesin disposal of domestic and medical wastes,abuse of fertilizers and pesticides, and generallack of environmental monitoring and lawenforcement. Moreover, this country experi-enced the largest oil spill along the easternMediterranean coast during the 2006 Israeliraid (El-Fadel et al., 2012; Coppini et al.,2011). However, despite all these reports,apparent data on environmental levels of manypollutants and carcinogens are still missing.

In addition, smoking in Lebanon is highlyprevalent in public places, particularly as therecent law banning smoking in public areasis not enforced or widely embraced (Nakkashet al., 2010; Saade et al., 2010; Katurji et al.,2010). Approximately 54–60% of the totalLebanese population smokes cigarettes andwater-pipe tobacco (narghile) (Khattab et al.,2012; Waked et al., 2012). Narghile smokingis highly prevalent especially among youth(Nakkash et al., 2011). Sepetdjian et al. (2008)noted that a single narghile smoking sessiondelivers approximately 50-fold more polycyclicaromatic hydrocarbons (PAH) compared toa single smoked cigarette. Many officials andpolicymakers in Lebanon argue that the alarm-ing rise of cancer, particularly that pertaining

Dow

nloa

ded

by [

Uni

vers

ity o

f W

inds

or]

at 1

8:48

14

Nov

embe

r 20

14

TOXICOGENETICS & CANCER IN LEBANESE 97

to bladder and lung cancer, might be directlyattributed to what is known as the new tobaccoepidemic that has been invading the countrysince the 1990s (Shamseddine and Musallam,2010). The proportion of ever-smokers climbedfrom 30% in the 90s to 43% in 2004, and iscurrently estimated to be 54–60% (Chaayaet al., 2004; Waked et al., 2012). Kobeissi et al.(2013) conducted a study among a group ofbladder cancer patients and healthy controlsand showed smoking and occupational expo-sure to diesel as risk factors for this disease inLebanon.

Genetic factors may also contribute to thehigh cancer incidence in Lebanese. Differencesin genetic heterogeneity among different pop-ulations may actually dictate significant differ-ences in cancer risk. In a study of 72 unre-lated Lebanese breast and ovarian patients witha reported family history, BRCA1/2 sequencevariants with deleterious effects were observedin high frequencies above 12% (Jalkh et al.,2012). This is higher than the prevalenceof BRCA1/2 found among breast cancerpatients of African, Asian, European Caucasian,and Hispanic descent (1–5%), and similar tofrequencies reported among ovarian cancerpatients in North America (13–15%) (Ghadirianet al., 2013; Kurian, 2010). In another studyinvestigating matrix metalloproteinases (MMP),a family of enzymes that degrade extracellu-lar matrix components, among 41 Lebaneselung cancer patients and 51 unrelated healthycontrols, a significant association was foundbetween MMP3-1171 5A allele and lung can-cer risk (Fakhoury et al., 2012). Health pro-fessionals are emphasizing the need for moreresearch to produce evidence on such asso-ciations, as well as to elicit genetic and envi-ronmental risk factors, which are potentiallycontributing along with tobacco metabolites tothe continued rise of cancer incidence.

The current review is an attempt to providean evidence-based analysis on how selecteddrug-metabolizing enzymes (DME) andtheir genetic polymorphisms may influencecancer risk in Lebanese following a gene-environment interaction model. This reviewprimarily focused on the DME glutathione

S-transferases (GST), N-acetyltransferases(NAT), and cytochromes P-450 (CYP450), andtheir associations with predominant cancersin this country, mainly bladder, prostate,colorectal, breast, and lung cancer. It is con-ceivable that these DME may serve as potentialuseful biomarkers for an improved preven-tion and a more accurate risk assessment inLebanon.

Cancer Incidence in MalesAccording to latest NCR update “Cancer

2007,” the overall estimated incidence ratefor all cancers combined among men was214.2 per 100,000 (Lebanese Ministry of PublicHealth and Epidemiology Surveillance Program,2010). The highest reported malignancies inLebanese males were prostate (ASR = 39.3 per100,000), bladder (ASR = 32.0 per 100,000),lung (ASR = 30.3 per 100,000), skin (ASR =20.2 per 100,000), and colon cancer (ASR =15.4 per 100,000), and non-Hodgkins lym-phoma (ASR = 14.7 per 100,000). These dataindicate a continuous climb in cancer inci-dence, compared to 1998 and 2004 data, withan unusually high incidence of bladder cancerin the country compared to global incidenceof this disease. The incidence of bladder can-cer among males ranks seventh on a globalscale and accounts for 4.4% of all malignancies(Ferlay et al., 2010). In general, bladder cancerincidence is reported to be higher in indus-trialized countries such as the United States(ASR = 21.1 per 100,000) and the EuropeanUnion (ASR = 6.3 per 100,000), and lower indeveloping countries in Africa and Asia, whereASR is estimated to range between 3.1 and5.2 per 100,000. Egypt presents an excep-tion in Africa with much higher incidence(ASR = 23.7 per 100,000), mainly due toschistosomiasis (Mostafa et al., 1999).

Cancer Incidence in FemalesAmong Lebanese women, the overall crude

incidence rate for all cancers combined in2007 was 213.2 per 100,000. Breast cancermalignancy presented the highest incidence

Dow

nloa

ded

by [

Uni

vers

ity o

f W

inds

or]

at 1

8:48

14

Nov

embe

r 20

14

98 H. R. DHAINI AND L. KOBEISSI

(ASR = 91.8 per 100,000), followed by skin(ASR = 13.3 per 100,000), non-Hodgkins lym-phoma (ASR = 12 per 100,000), colon (ASR =10.3 per 100,000), and bladder cancer (ASR =7.3 per 100,000) (Lebanese Ministry of PublicHealth and Epidemiology Surveillance Program,2010). Incidence of breast cancer in Lebanesefemales is more than twofold higher than theestimated world age-standardized incidence,and more than threefold greater than that ofdeveloping countries, and markedly higher thanthe estimated ASR for Europe (ASR = 66 per100,000) and the United States (ASR = 76 per100,000). In 2004, almost 50% of breast can-cer patients in Lebanon were below the ageof 50 yr, with a median age of 52 yr. Further,breast cancer patients below the age of 40 yrrepresented 22% of cases in Lebanon, versus6% in Western populations (Shamseddine andMusallam, 2010). In fact, breast cancer inci-dence patterns in Lebanon showed some of thehighest age-specific incidence rates globally forthe following age groups: 35 to 39 yr, 40 to44 yr, and 45 to 49 yr (El Saghir et al., 2006,2007; Lakkis et al., 2010). Given that youngerage onset of breast cancer at initial diagnosisis associated with poor prognosis, many breastcancer researchers in Lebanon argue towardmodifying the current screening guidelines toencourage women considered at high risk withstrong familial predisposition to start screeningat age 35 as opposed to age 40 (El Saghir et al.,2006; Lakkis et al., 2010).

GENETIC POLYMORPHISM OF DME

DME Functional Variations and CancerRiskA large number of cancers are associated

with occupational and environmental chemicalexposures (Geller et al., 2008;Mohner et al.,2013;Vlaanderen et al., 2013; Letasiova et al.,2012). Exposure to carcinogens in tobaccoproducts is correlated with cancers of the lung,bladder, colon, and other tissues (Hoffmannand Hoffmann, 1997; Blakely et al., 2013).Chronic exposure to benzene, a gasoline com-ponent and one of many solvents used in the

chemical and drug industries, is also knownto produce cancer, particularly leukemia inhumans (Snyder, 2000; Polychronakis et al.,2013). In various settings involving exposureto fine particles like asbestos, increased ratesof mesothelioma and cancer of the lunghave been consistently observed (Mossmanet al., 2011). In addition, exposure to furansand dioxins like 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), from incineration of domesticand medical wastes, is correlated with higherrisk of lung cancer and non-Hodgkins lym-phoma (National Toxicology Program, 2011).At the same time, reports show increased inci-dence of lung, bladder, and skin cancer inhumans exposed to mixtures of polycyclic aro-matic hydrocarbons (PAH), reported by theInternational Agency for Research on Cancer(IARC, 2012). The primary source of PAH is theburning of carbon-containing compounds suchas fuel and tobacco, in addition to charcoal-broiled food. Human exposure to aromaticamines, such as benzidine or benzidine-baseddyes, is now known to result in urinary blad-der cancer (Rosenman and Reilly, 2004; Golkaet al., 2008). Further, vinyl chloride, a humancarcinogen used by the plastics industry inmany consumer products, including wrappingfilm, water pipes, electrical insulation, flooring,and hoses, is associated with higher risk of lungcancer and angiosarcoma of liver and the brain(National Cancer Institute, 2003). Exposure tometal dust or fume was also related to increasedcancer risk (IARC, 2012). Arsenic compoundsare associated with many forms of skin, lung,bladder, kidney, and liver cancers, particularlywhen high levels are consumed in drinkingwater (Tsai et al., 1998; Bernstam and Nriagu,2000). Studies showed that exposure to Cdfumes and Cd compounds is associated withan increased risk of lung cancer (Lin et al.,2013). Beryllium (Be) is also known to producelung cancer. Occupational exposure to Be mayoccur in the electronic and telecommunicationindustries, while environmental exposure mayprimarily arise from burning of coal and fuel oil.Similarly, Cr compounds, used in electroplatingof metal accessories, are known to induce lungcancer. Workers handling hexavalent Cr are at

Dow

nloa

ded

by [

Uni

vers

ity o

f W

inds

or]

at 1

8:48

14

Nov

embe

r 20

14

TOXICOGENETICS & CANCER IN LEBANESE 99

greater risk compared to the general population(Shi et al., 1999). Aflatoxins are cancer-causingsubstances produced by certain types of fungigrowing on grains and peanuts. Exposure tohigh levels of aflatoxins increases the risk of livercancer (Williams et al., 2004).

Cancer risk is dependent not onlyon potency and intensity of exposure tocarcinogenic compounds, but also on interindi-vidual variation in response to chemicals(Zheng et al., 2011; Lacko et al., 2009;Rossini et al., 2008). Therefore, the geneticmakeup of DME is an important contributorto individual cancer risk. Genetic variants ofDME are now postulated to result in criticalchanges in metabolism of carcinogens andsubsequently to modify individual cancersusceptibility (Rodriguez-Antona et al., 2010).The study of environmental exposures andtheir interactions with genetics identifiedvariations in human genes that, under certainenvironmental exposures, increase cancer riskin a population (Perera, 1998; Rundle et al.,2000). Many chemicals labeled as carcinogenicare bioactivated by Phase I drug-metabolizingenzymes and detoxified by Phase II conju-gation enzymes. The competition betweenbioactivation and detoxification pathways, andthe coordinated level of expression and regu-lation of both types of enzymes are importantfactors in deciding whether an exposure mightresult in a malignant transformation (Stiborovaet al., 2012; Sato et al., 1999).

Functional variation in genes coding forthese enzymes is likely to have an effect oncancer risk for an individual. However, thiseffect may have a large population impact ifrelated polymorphisms are highly prevalent inthat particular population (Brennan, 2002). Thesections that follow review particular geneticpolymorphisms of three types of DME, namely,GST, NAT, and CYP450, and their prevalencein Lebanese. These DME and individual isoen-zymes were selected based on their impor-tance and on data availability in the populationof interest. Other important members of theaforementioned DME families exist but are notaddressed in the context of the current review.

Glutathione S-Transferases (GST)Glutathione S-transferases (GST) are a class

of Phase II enzymes, present in many tissues,and active in defending cells from free radicalsand electrophiles. GST contribute to protec-tion from a broad range of carcinogens andoxidative stress metabolites. In humans, eightdifferent subfamilies of GST enzymes have beendescribed and grouped based on sequence sim-ilarities, termed GST alpha (A), mu (M), pi (P),zeta (Z), theta (T), omega (O), kappa (K), andsigma (S), with one or more genes in each class(Mannervik et al., 1992; Lo and Ali-Osman,2007). Two major genetic polymorphisms forthe GSTM1 gene (chromosome 1p13.3) andGSTT1 gene (chromosome 22q11.23) resultfrom gene deletion and are associated withabsent enzymatic activity in individuals carryingthese deletions (i.e., GST ∗0/∗0 null genotype).Frequencies of GSTM1 and GSTT1 homozy-gous null genotypes were reported to vary indifferent populations (Ginsberg et al., 2009;Cho et al., 2005; Garte et al., 2001; Chen et al.,1997). GSTM1 null genotype had a preva-lence of 53–63% in Caucasians and Asians, and30–40% in Africans (Bailey et al., 1998; Garteet al., 2001), whereas GSTT1 null genotypehas a prevalence of 20–28% in Caucasians andAfricans, and 48–54% in Asians (Strange andFryer, 1999; Chen et al., 1997; Liu et al.,2009; Chowbay et al., 2005). Similarly, theGSTP1 locus was also shown to be polymorphicwith variation in allelic frequencies among dif-ferent ethnicities (Lo and Ali-Osman, 2007).

Two recent studies investigated GST variantfrequency distribution in Lebanese. One studycompared GSTM1 and GSTT1 genotypic fre-quencies in a random sample of 141 Lebaneseresiding in Beirut to two other Arab populations(Salem et al., 2011). GSTM1 and GSTT1 nullgenotype frequencies were found to be52.5 and 37.6%, respectively. Lebanese werefound to be similar to Europeans and Asiansand higher than Africans with regard to thefrequency of GSTM1 null genotype (Table 1).Lebanese also showed GSTT1 null genotypefrequencies lower than Asians and higherthan Caucasians and Africans. In addition, the

Dow

nloa

ded

by [

Uni

vers

ity o

f W

inds

or]

at 1

8:48

14

Nov

embe

r 20

14

TABL

E1.

Glu

tath

ione

S-Tr

ansf

eras

ePo

lym

orph

isms

and

Asso

ciat

edC

ance

rRisk

Leba

nese

Asia

nC

auca

sian

Oth

er

Stud

ies

Alle

les

Risk

Stud

ies

Alle

les

Risk

Stud

ies

Alle

les

Risk

Stud

ies

Alle

les

Risk

Con

trol

popu

latio

ns(fr

eque

n-ci

es)

(Sal

emet

al.,

2011

)G

STM

(52.

5%)

GST

T1ω

(37.

6%)

Dou

ble

null

(16.

3%)

(Cho

wba

yet

al.,

2005

)(L

iuet

al.,

2009

)(G

arte

etal

.,20

01)

GST

M1

(53–

63%

)G

STT1

(48–

54%

)D

oubl

enu

ll(2

4%)

(Bai

ley

etal

.,19

98)

(Gar

teet

al.,

2001

)

GST

M1

(53–

61%

)G

STT1

(20–

27%

)D

oubl

enu

ll(1

0%)

(Bai

ley

etal

.,19

98)

(Che

net

al.,

1997

)

Afri

can

GST

M1

(30-

40%

)G

STT1

(28%

)D

oubl

eN

ull

(4%

)Bl

adde

rca

ncer

N/A�

——

(Wu

etal

.,20

13a)

Dou

ble

Nul

l+G

STP1

Hig

her

(Gon

get

al.,

2012

)D

oubl

eN

ull

Hig

her

(Ber

bere

tal.,

2013

)Tu

rkis

hG

STM

1G

STT1

Non

eH

ighe

rBr

east

canc

er(Z

ghei

bet

al.,

2013

)G

STM

1G

STT1

GST

P1∗

Non

eN

one

Non

e

(Sak

oda

etal

.,20

08)

(Ser

gent

anis

etal

.,20

10)

GST

M1

GST

P1G

STT1

GST

P1

Non

eN

one

Non

eN

one

(Spu

rdle

etal

.,20

10)

GST

M1

GST

T1G

STP1

Non

eN

one

Non

e

(Ram

alhi

nho

etal

.,20

11)

Port

ugue

seG

STM

1G

STT1

Dou

ble

Nul

lD

oubl

eN

ull+

GST

P1

Hig

her

Hig

her

Hig

her

Hig

her

Col

orec

tal

canc

er(D

araz

yet

al.,

2011

)G

STM

1H

ighe

r(E

cono

mop

oulo

set

al.,

2010

)G

STM

1G

STT1

GST

P1

Non

eN

one

Non

e

(Eco

nom

opou

los

etal

.,20

10)

GST

M1

GST

T1G

STP1

Hig

her

Hig

her

Non

e

(Mar

tinez

etal

.,20

06)

(Hez

ova

etal

.,20

12)

Span

ish

GST

M1

GST

T1D

oubl

enu

llC

zech

GST

M1

GST

T1G

STP1

Hig

her

Hig

her

Hig

her

Non

eN

one

Low

erLu

ngca

ncer

N/A

——

(Liu

etal

.,20

12)

(Gu

etal

.,20

07)

(Kiy

ohar

aet

al.,

2012

)

GST

M1

GST

M1

GST

M1

GST

T1G

STP1

Hig

her

Hig

her

Hig

her

Non

eH

ighe

r

(To-

Figu

eras

etal

.,19

97)(

Wan

get

al.,

2003

)(G

erva

sinie

tal.,

2010

)

GST

M1

GST

T1G

STP1

GST

M1

GST

T1G

STP1

Non

eN

one

Hig

her

Non

eN

one

Non

e

(Atin

kaya

etal

.,20

12)

( Ada

etal

.,20

12)

Turk

ish

GST

M1

GST

T1G

STP1

Non

eN

one

Hig

her

Pros

tate

canc

erN

/A

——

(Yan

get

al.,

2013

)(K

won

etal

.,20

11)

(Mo

etal

.,20

09)

GST

T1G

STM

1G

STT1

GST

P1

Hig

her

Hig

her

Non

eN

one

(Yan

get

al.,

2013

)(M

oet

al.,

2009

)G

STT1

GST

M1

GST

T1G

STP1

Hig

her

Hig

her

Non

eN

one

(Tha

kure

tal.,

2011

)(Y

ang

etal

.,20

13)

(Qad

riet

al.,

2011

)

Indi

anG

STM

1G

STT1

GST

P1

Hig

her

Hig

her

Hig

her

�St

udie

sno

tava

ilabl

ear

ein

dica

ted

asN

/A.

∗ GST

P1ge

noty

pelis

ted

refe

rsto

the

A/G

varia

nt.

ωG

STM

1an

dG

STT1

geno

type

slis

ted

refe

rto

the

Hom

ozyg

ote

null,

doub

le-n

ull.

100

Dow

nloa

ded

by [

Uni

vers

ity o

f W

inds

or]

at 1

8:48

14

Nov

embe

r 20

14

TOXICOGENETICS & CANCER IN LEBANESE 101

double null genotype frequency (GSTM1 null +GSTT1 null) in Lebanese (16.3%) is higher thanthat reported in Caucasians and Africans, lowerthan that in Asians (Garte et al., 2001; Choet al., 2005; Moy et al., 2009; Morinobu et al.,1999), and almost similar to other Arab popu-lations studied for GST polymorphisms (Salemet al., 2011; Bu et al., 2004). The secondreport in Lebanese, a case control study inves-tigating colorectal and gastric cancer, showedsimilar frequency distributions for GSTM1 vari-ants (Darazy et al., 2011). In a total sampleof 140 Lebanese individuals, including can-cer patients and matched healthy controls,the GSTM1 null genotype had a frequency of46.2% (Darazy et al., 2011).

N-Acetyltransferases (NAT)NATs are a family of Phase II enzymes,

mainly consisting of two isoenzymes inhumans: N-acetyltransferase 1 (NAT1) andN-acetyltransferase 2 (NAT2). Both enzymesmediate N-acetylation and O-acetylation ofa large number of carcinogens, particularlyheterocyclic and aromatic amine derivatives.NAT2 is mainly expressed in liver and the gas-trointestinal tract (Hein et al., 2000). Morethan 30 single-nucleotide polymorphisms (SNP)were reported for NAT2 in humans (Hein,2009). Many of these polymorphisms arethought to result in altered enzymatic activ-ity (Borlak and Reamon-Buettner, 2006). Mostidentified haplotypes (particularly clusters ofNAT2∗5, ∗6, ∗7, and ∗14) were shown to codefor slow acetylation, while NAT2∗4, designatedas the wild type, codes for fast acetylation(Hein et al., 2000). Frequencies of slow andfast acetylation phenotypes vary among dif-ferent ethnic groups (Sabbagh et al., 2011;Garcia-Martin, 2008). Slow acetylators werereported at frequencies of 67–90% in Arab pop-ulations, 40–60% in Caucasians, and 5–25%in Asians (Lin et al., 1994; Woolhouse et al.,1997; Xie et al., 1997; Karim et al., 1981; Evanset al., 1985; Walker et al., 2009). In Lebanese,only one study examined NAT2 allelic frequen-cies, targeting a group of breast cancer patientsand healthy controls. Results from that study

reported slow acetylators at low frequencies of10% and 18% for NAT2∗6/∗6 and NAT2∗5/∗5genotypes, respectively (Zgheib et al., 2013).

Compared to NAT2, NAT1 was shown tobe expressed in a wider number of tissues(Windmill et al., 2000). It was initially describedto be monomorphic based on its selectivity forp-aminosalicylic acid (Weber and Hein, 1985).The search for a genetic basis to explain dif-ferences in NAT1 enzymatic activity led tothe discovery of a genetic polymorphism, withsignificant variation in the frequency distribu-tion of the different alleles among various eth-nic groups (Walker et al., 2009; Vatsis andWeber, 1993). To-date, 28 NAT1 allelic vari-ants have been identified and characterizedin humans (Arylamine N-acetyltransferase GeneNomenclature Committee, 2011).

In two studies conducted earlier, thefrequency distribution of NAT1 variants inLebanese was reported. The first study col-lected DNA from saliva samples of a small groupof Lebanese living in the Detroit Metropolitanarea in Michigan (Dhaini and Levy, 2000).Results revealed that nearly 50% of the pop-ulation were carriers of the NAT1∗14 allele.NAT1∗14A genetic variant was reported to pro-duce a slow acetylation phenotype based onstudies in recombinant enzyme expressed inEscherichia coli and blood mononuclear celllysate (Butcher et al., 1998; Hughes et al.,1998). Allelic frequency of NAT1∗14A was21.4% among Lebanese-Americans, comparedto 2% or lower among all other populationsand ethnic groups investigated for that partic-ular allele (Butcher et al., 1998; Dhaini andLevy, 2000; Hughes et al., 1998; Cascorbiet al.,2001; Bruhn et al., 1999) (Table 2). Thesecond report was a case-control study con-ducted in a group of 160 Lebanese males,including bladder cancer patients and matchedhealthy controls, all residing in Lebanon (Yassineet al., 2012). The analyzed frequencies weresimilar to those found in the Michigan group;NAT1∗14A was found to have a high allelic fre-quency of 15% in the total sample. At the sametime, a 10.7% allelic frequency was reportedfor NAT1∗10 (Dhaini and Levy, 2000). This isless frequent than in Caucasians, Asians, and

Dow

nloa

ded

by [

Uni

vers

ity o

f W

inds

or]

at 1

8:48

14

Nov

embe

r 20

14

TABL

E2.

N-A

cety

ltran

sfer

ase

Poly

mor

phism

san

dAs

soci

ated

Can

cerR

isk

Leba

nese

Asia

nC

auca

sian

Oth

er

Stud

ies

Alle

les

Risk

Stud

ies

Alle

les

Risk

Stud

ies

Alle

les

Risk

Stud

ies

Alle

les

Risk

Con

trol

popu

latio

ns(fr

eque

n-ci

es)

(Dha

inie

tal.,

2000

)(Z

ghei

bet

al.,

2013

)

NAT

1∗3

(3.6

%)

NAT

1∗4

(56%

)N

AT1∗

10(1

0.7%

)N

AT1∗

14A

(21.

4%)

NAT

1∗1

B(2

.4%

)—

——

—N

AT2∗

5(4

0%)

NAT

2∗6

(32%

)

(Zha

oet

al.,

1998

)

(Lin

etal

.,19

93)

(Zha

oet

al.,

2000

)

NAT

1∗3

(33%

)N

AT1∗

4(3

4%)

NAT

1∗10

(59%

)N

AT1∗

14(0

%)

——

——

NAT

2∗5

(6%

)N

AT2∗

6(2

3-31

%)

(Bru

hnet

al.,

1999

)(C

asco

rbi

etal

.,20

01)

(Bel

leta

l.,19

93)

(Gar

teet

al.,

2001

)

NAT

1∗3

(3%

)N

AT1∗

4(7

0-72

%)

NAT

1∗10

(20-

21%

)N

AT1∗

14(0

.6-2

.2%

)—

——

—N

AT2∗

5(4

4%)

NAT

2∗6

(27-

31%

)

(Wal

kere

tal.,

2009

)(B

elle

tal.,

1993

)(M

illik

anet

al.,

1998

)

Afri

can

NAT

1∗3

(3.6

%)

NAT

1∗4

(50%

)N

AT1∗

10(4

5%)

——

——

—N

AT2∗

5(2

5-31

%)

NAT

2∗6

(22-

28%

)

Blad

der

canc

er(Y

assin

eet

al.,

2012

)(K

obei

ssie

tal.,

2013

)

NAT

1∗10

NAT

1∗14

ALo

wer

Hig

her

(Wu

etal

.,20

13b)

NAT

1∗10

Non

e(C

asco

rbi

etal

.,20

01)

(Wu

etal

.,20

13b)

(Tay

lor

etal

.,19

98)

NAT

1∗10

NAT

1∗14

AN

AT1∗

10N

AT1∗

10

Low

erH

ighe

rN

one

Hig

her

(Wu

etal

.,20

13b)

Afri

can

NAT

1∗10

Non

e

Brea

stca

ncer

�N

/A

for

NAT

1(Z

ghei

bet

al.,

2013

)

——

——

NAT

2∗5

NAT

2∗6

Non

eN

one

(Lee

etal

.,20

03)

(San

graj

rang

etal

.,20

10)

NAT

1∗10

——

——

NAT

2∗5

NAT

2∗6

Non

e

Non

eN

one

(Zhe

nget

al.,

1999

)(K

rajin

ovic

etal

.,20

01)

(Cox

etal

.,20

11)

NAT

1∗10

NAT

1∗11

NAT

1∗10

——

——

NAT

2∗5

NAT

2∗6

NAT

2∗7

Hig

her

Hig

her

Hig

her

Non

eN

one

Non

e

(Mill

ikan

2000

)(O

zbek

etal

.,20

10)

Afri

can

NAT

1∗10

Turk

ish

NAT

2∗5

Non

e

Non

e

Col

orec

tal

canc

erN

/A

——

(Zha

nget

al.,

2002

)N

AT1∗

10H

ighe

r(B

elle

tal.,

1995

)(R

oem

eret

al.,

2008

)(H

ubba

rdet

al.,

1998

)

NAT

1∗10

NAT

1∗10

NAT

1∗11

NAT

1∗14

Hig

her

Non

eN

one

Non

e

(Liu

,Din

g,et

al.,

2012

)

Afri

can

NAT

1∗10

Non

e

Lung

canc

erN

/A

——

(Seo

wet

al.,

1999

)N

AT2

slow

acet

ylat

ors

∗ 5,∗

6,∗ 7

Hig

her

(Bou

char

dyet

al.,

1998

)(W

ikm

anet

al.,

2001

)

NAT

1∗14

NAT

1∗15

NAT

1∗10

NAT

1∗11

Hig

her

Hig

her

Hig

her

Hig

her

(Hei

net

al.,

2000

)

Afri

can

NAT

1∗10

Non

e

Pros

tate

canc

erN

/A

——

(Gon

get

al.,

2011

)(F

ukut

ome

etal

.,19

99)

NAT

1∗10

NAT

1∗10

Non

eH

ighe

r(H

ein

etal

.,20

02)

(Kid

d,H

ein,

etal

.,20

11)

NAT

1∗10

NAT

1∗14

NAT

1∗10

Hig

her

Non

eN

one

(Kid

d,Va

ncle

ave,

etal

.,20

11)

Afri

can

NAT

1∗10

Non

e

�St

udie

sno

tava

ilabl

ear

ein

dica

ted

asN

/A.

102

Dow

nloa

ded

by [

Uni

vers

ity o

f W

inds

or]

at 1

8:48

14

Nov

embe

r 20

14

TOXICOGENETICS & CANCER IN LEBANESE 103

Africans (Okkels et al., 1997; Zheng et al.,1999; Probst-Hensch et al., 1996; Walker et al.,2009; Zhao et al., 1998) (Table 2). NAT1∗10is reported to code for a rapid acetylatorphenotype (Bell et al., 1995a) and increasedexpression in transfected Chinese hamster ovar-ian cells (Millner et al., 2012). This NAT1 varianthas a substitution in the 3′-untranslated regionthat may alter the polyadenylation signalingleading into a more stable mRNA, and hencean enhanced expression (Bell et al., 1995b).

Cytochromes P-450 (CYP450)The cytochrome P-450-dependent mixed-

function oxidases are a family of heme-containing proteins that play an important rolein the metabolism of a wide variety of endoge-nous compounds active in cellular signalingsuch as steroids, fatty acids, and eicosanoids(Wrighton et al., 1996). CYP are also involvedin bioactivation and detoxification of a largenumber of xenobiotics and carcinogens (Parke,1994; Shi et al., 2010).

Of particular importance is CYP2E1 withseveral reported polymorphisms. Two SNP,CYP2E1∗5B and CYP2E1∗6, have been fre-quently studied (Shahriary et al., 2012).CYP2E1∗5B is characterized by a G to C SNP atposition 1293 in the 5′-flanking region of thegene. G and C alleles are named c1 and c2,respectively. CYP2E1∗5B is reported to alter thetranscription of CYP2E1 gene in vitro (Hayashiet al., 1991). CYP2E1∗6, on the other hand, isbased on a nucleotide substitution T to A atposition 7632 in intron 6. T and A SNP arenamed D and C, respectively. The effect ofCYP2E1∗6 polymorphism on enzyme activityis still not well elucidated. However, Lucaset al. (1995) showed a trend of less efficientinduction in carriers of such allelic variantscompared to wild type.

In Lebanese, two studies explored theallelic frequency distribution of CYP, both ofwhich focused on CYP2E1. The first studyassessed the frequency distribution of CYP2E1among 216 cancer-free Lebanese (100 menand 116 women). The allelic frequencies ofCYP2E1∗5B and CYP2E1∗6 were found to be0.7 and 6.3%, respectively (Zgheib et al., 2010).

For CYP2E1∗5B, 98.6% were c1/c1 and no car-riers were identified for the c2/c2 genotype.For CYP2E1∗6, 89.8% were D/D and only onesubject carried the C/C genotype. All those whocarried the CYP2E1∗5B allele were also carri-ers of the CYP2E1∗6 allele. The second studyexamined the distribution of CYP2E1, as well asthat of CYP1A1, among Lebanese gastrointesti-nal cancer patients and healthy controls (Darazyet al., 2011). Frequencies of the CYP2E1∗6 andCYP1A1∗2A were found to be 4.9 and 11.3% inthe total sample size, respectively.

The reported CYP2E1 allelic frequenciesobserved in Lebanon differed from otherhuman populations (Bolt et al., 2003). Thesefrequencies were lower than those observedin Iranians (1.5 and 16% for CYP2E1∗5Band CYP2E1∗6, respectively) (Shahriary et al.,2012), and lower than those observed inCaucasians (3% for CYP2E1∗5B). However, fre-quencies were similar to those observed inAfrican-Americans (0.3%) for CYP2E1∗5B (Liuet al., 2001), and similar to those observed inCaucasians, Asians, and Turkish for CYP2E1∗6(6.3–7.7%) (Kayaalti and Soylemezoglu, 2010;Brockmoller et al., 1996) (Table 3).

Other CYP were also found to bepolymorphic. CYP2D6, 2C19, 2C9, andCYP3A4/5 polymorphisms account for majorvariations in Phase I drug metabolism, sincealmost 80% of xenobiotics in use are bio-transformed by these enzymes (Belpaire andBogaert, 1996). For example, up to 14% ofCaucasians, 5% of Africans, and 1% of Asianslack CYP2D6 activity, and these are known aspoor metabolizers (Neafsey et al., 2009; Zhouet al., 2009). CYP2D6 polymorphism frequen-cies are not available for Lebanese. Extensivepolymorphisms also occur in other CYP genes,such as 1A1, 1B1, 2A6, 2A13, and 2C8. Severalof these genes play a role in the bioactivationof many carcinogens, and hence contributeto the variable susceptibility to carcinogenesis(Shimada and Guengerich, 1991).

TOXICOGENETICS AND CANCER RISK

In this section, an overview of the literatureevaluating the role of GST, NAT, and CYP2E1 in

Dow

nloa

ded

by [

Uni

vers

ity o

f W

inds

or]

at 1

8:48

14

Nov

embe

r 20

14

TABL

E3.

Cyt

ochr

omes

P450

Poly

mor

phism

san

dAs

soci

ated

Can

cerR

isk

Leba

nese

Asia

nC

auca

sian

Oth

er

Stud

ies

Alle

les

Risk

Stud

ies

Alle

les

Risk

Stud

ies

Alle

les

Risk

Stud

ies

Alle

les

Risk

Con

trol

popu

latio

ns(fr

eque

n-ci

es)

(Zgh

eib

etal

.,20

10)

2E1∗

5B(0

.7%

)2E

1∗6

(6.3

%)

(Bol

teta

l.,20

03)

2E1∗

5B(1

%)

2E1∗

6(6

.3%

)(B

rock

mol

ler

etal

.,19

96)

2E1∗

5B(3

%)

2E1∗

6(6

.5%

)(S

hahr

iary

etal

.,20

12)

Iran

ian

2E1∗

5B(1

.5%

)2E

1∗6

(16%

)Bl

adde

rca

ncer

(Bas

ma

etal

.,20

13)

2E1∗

5Bc1

/c1

Hig

her

(Cho

ieta

l.,20

03)

2E1∗

5Bc1

/c1

Hig

her

(Far

kere

tal.,

1998

)2E

1∗5B

c1/c2

(+)2

E1∗ 6

C

Hig

her

Hig

her

(Can

tore

tal.,

2010

)

Span

ish

2E1∗

5Bc2

/c2

Hig

her

Brea

stca

ncer

(Zgh

eib

etal

.,20

13)

2E1∗

5Bc1

&c2

2E1∗

6D

&C

Non

eN

one

(Wu

etal

.,20

06)

2E1∗

5Bc1

/c1

2E1∗

5Bc2

/c2

Hig

her

Low

er(M

cCar

tyet

al.,

2012

)2E

1∗5B

c1an

dc2

Non

e(Is

can

etal

.,20

01)

Turk

ish

CYP

2E1

Expr

essio

nN

one

Col

orec

tal

canc

er(D

araz

yet

al.,

2011

)2E

1∗6

D&

C1A

1∗2A

Non

eN

one

(Pen

get

al.,

2013

)2E

1∗5B

c2H

ighe

r(K

isset

al.,

2000

)2E

1∗5B

c2H

ighe

r(S

aeed

etal

.,20

13)

Saud

iAra

b2E

1∗6

D&

C1A

1∗2A

Non

eH

ighe

r

Lung

canc

erN

/A�

——

(Zha

net

al.,

2010

)(S

uet

al.,

2011

)(W

ang

etal

.,20

10)

2E1∗

5Bc1

/c1

2E1∗

6D

/D

Hig

her

Hig

her

(Zha

net

al.,

2010

)2E

1∗5B

c1an

dc2

Non

e(P

erez

-Mor

ales

etal

.,20

11)

Mex

ican

2E1∗

5Bc1

&c2

Non

e

Pros

tate

canc

erN

/A

——

(Yan

get

al.,

2006

)2E

1∗5B

c22E

1∗5B

c1/c1

Low

erH

ighe

r(F

erre

iraet

al.,

2003

)2E

1∗5B

c1an

dc2

2E1∗

6D

/D

Non

eH

ighe

r(Jo

shie

tal.,

2012

)

Mul

tieth

nic

(Hisp

anic

&Af

rican

)2E

1∗5B

c1&

c2

Non

e

�St

udie

sno

tava

ilabl

ear

ein

dica

ted

asN

/A.

104

Dow

nloa

ded

by [

Uni

vers

ity o

f W

inds

or]

at 1

8:48

14

Nov

embe

r 20

14

TOXICOGENETICS & CANCER IN LEBANESE 105

susceptibility to bladder, colorectal, prostate,breast, and lung cancer is presented.

Urinary Bladder Cancer (UBC)It is now commonly accepted that urinary

bladder cancer (UBC) is the result of a multifac-torial interaction of environmental triggers, suchas chemical exposure, smoking, and chronicurinary-tract infections, as well as genetic fac-tors. In recent years, a large number of epi-demiologic studies suggested that many geneticpolymorphisms affect UBC risk (Ovsiannikovet al., 2012; Gong et al., 2012; Moore et al.,2011; Cantor et al., 2010). Many studies linkedGSTM1 and GSTT1 null genotypes to differenttypes of cancer including bladder (Chen et al.,2008, 2010; Wu et al., 2013a; Gsur et al.,2001). DNA adducts were significantly higherin subjects with GSTM1 null exposed to PAH(Rojas et al., 1998). In a large meta-analysis,the combination of the GSTP1 genotype witheither GSTM1 null or GSTT1 null was found toincrease UBC risk by more than twofold (Gonget al., 2012). Individuals with all 3 genotypeswere reported to have an increased UBC sus-ceptibility by sixfold in different ethnic groupsincluding Caucasians and Chinese (Wu et al.,2013a) (Table 1). This suggests a gene–geneinteraction that may be explained by varioussubstrates metabolized by different GST result-ing in a combined effect.

A possible role for NAT2 polymorphisms inUBC was suggested more than 30 years agoin view of its role in arylamine metabolism(Lower et al., 1979). A later study reporteda strong association between UBC risk andslow acetylation in groups occupationallyexposed to arylamine (Cartwright et al., 1982).Subsequently, several case-control studies sup-ported these findings with noted UBC can-cer risk reaching as high as 16-fold in slowversus fast acetylators (Hein, 1988; Laderoet al., 1985; Hanke and Krajewska, 1990;Weistenhofer et al., 2008). One striking excep-tion to these results was a cohort study ina group of Chinese workers using benzidine.This cohort reported no association betweenNAT2 genotypes nor phenotypes and UBC risk

(Hayes et al., 1993). More recent studies foundhigher UBC risk in NAT2 slow acetylators (Cuiet al., 2013; Garcia-Closas et al., 2005). In apopulation-based case-control study, an ele-vated risk of UBC was reported among usersof permanent hair dyes who carried NAT2 slowacetylation phenotypes (Koutros et al., 2011).At the molecular level, the role of NAT2 insmoking-related UBC is supported by stud-ies on hemoglobin and DNA adducts (Vineis,1992). Among smokers, NAT2 slow acety-lators had higher levels of 4-aminobiphenylhemoglobin adducts (Yu et al., 1994). Reportedassociations were consistent among differentethnic groups (Green et al., 2000).

NAT1 is the main NAT expressed in bladderepithelia and proximal tubules of the kidneys.NAT1 genetic variants have been associatedwith several cancer types, and overexpressionwas associated with increased survival (Butcherand Minchin, 2012). Some studies suggesteda novel role for this enzyme in cancer cellgrowth through folate homeostasis (Butcherand Minchin, 2012; Stanley et al., 1996).In animal studies, rodents carrying the slowacetylation phenotype showed a higher DNAadducts formation in bladder when exposedto aromatic amines (Jiang et al., 1999; Levyand Weber, 1992). Further, in a case-controlstudy conducted in Lebanese males (54 casesand 106 controls), UBC cases had a higherNAT1∗14A allelic frequency in the total sam-ple with significantly higher clustering amongcases compared to controls, while NAT1∗10had a higher prevalence in controls (Yassineet al., 2012). This is in agreement with a studyconducted in 425 German UBC patients and343 controls, where NAT1∗10 allelic frequencywas lower in UBC patients, while NAT1∗14Awas overrepresented in cases (Cascorbi et al.,2001). Given that NAT1∗14A codes for a slowacetylator phenotype, in NAT1∗14A carriers,acetylation may compete poorly with Phase Ibioactivation pathways for carcinogens. Hence,a population with an overrepresentation ofNAT1∗14A frequency, such as the Lebanesepopulation, may have an enhanced susceptibil-ity for UBC upon chemical exposure. However,findings on NAT1∗10 are still controversial.

Dow

nloa

ded

by [

Uni

vers

ity o

f W

inds

or]

at 1

8:48

14

Nov

embe

r 20

14

106 H. R. DHAINI AND L. KOBEISSI

While some studies reported an associationwith UBC risk (Taylor et al., 1998), other studiesfound no marked association even within simi-lar ethnic groups (Wu et al., 2013b) (Table 2).

Among Phase I enzymes, CYP2E1 metab-olizes a large number of low-molecular-weight carcinogenic compounds includingN-nitrosamines, benzene, and halogenatedhydrocarbons (Tanaka et al, 2000; Hou et al.,2007; Guo et al., 2010; Feng et al., 2012).CYP2E1 is inducible by lifestyle factors includ-ing smoking, alcohol consumption, and expo-sure to various chemicals (Gonzalez, 2007).In Lebanese, Zgheib et al. (2010) noted no car-riers of CYP2E1 genetic variants in 15 Lebanesecancer patients. However, in a preliminarycase-control study conducted in a group of160 Lebanese men, including UBC patients,homozygote carriers of the CYP2E1 c1 allelewere highly clustered in cases compared to con-trols (80 versus 54.1%) (Basma et al., 2013).In the multivariate analysis, a fourfold UBCrisk was observed for carriers of the CYP2E1c1/c1 genotype compared to heterozygote andhomozygote carriers of the c2 allele. Thesefindings are consistent with those of Choiet al. (2003) demonstrating 80% higher UBCrisk in Koreans carrying the c1/c1 genotypecompared to c2 carriers. In contrast, a studyin German Caucasians detected a higherUBC risk in women carrying the CYP2E1∗5Bc2/c2 genotype and/or in combination withthe CYP2E1∗6 C allele (Farker et al., 1998a).Similar findings were reported in Spanish sub-jects exposed to trihalomethanes (Cantor et al.,2010) (Table 3).

In Lebanon, the current UBC incidence isone of the highest in the world (Shamseddineand Musallam, 2010). Incidence patterns indi-cate risk factors that are particular to theLebanese context compared to other coun-tries. In Egypt, the high incidence of UBC isattributed mainly to schistosomiasis (Mostafaet al., 1999). Schistosomiasis is known tobe associated with squamous-cell carcinoma(SCC) (Zheng et al., 2012). UBC incidencein Lebanon is almost double that in Egypt(Zheng et al., 2012). However, in Lebanon,schistosomiasis is extremely rare, and the

majority of reported bladder cancer cases aretransitional-cell carcinoma (NOS), whereas SCCmakes up only 1% of all bladder malignan-cies (Adib et al., 2008). Data suggest that otherunderlying risk factors need to be identifiedin order to further understand the alarminglyhigh UBC incidence in the country. Thus far,the few studies conducted in Lebanese identi-fied smoking, exposure to occupational dieselfumes, prostate-related morbidity, and NAT1acetylation genotype as potential predisposingrisk factors for UBC in men (Yassine et al.,2012; Kobeissi et al., 2013). Risk factors, par-ticularly smoking, are important in this com-munity, given the double burden of the twohighest reported cancer incidences: lung andbladder. UBC in Lebanese may be following agene–environment interaction model. Althoughobserved individual risks associated with N-acetylation genotypes may be small, these doincrease when considered in conjunction withchemical exposures and other metabolizingenzyme genes and susceptibility genes. Morestudies investigating possible etiologies of thisdisease in Lebanese, and modes of interactionbetween the different risk factors, are neededin the future. Specific genetic factors, envi-ronmental factors, and occupational exposuresneed to be examined using more reliable expo-sure biomarkers and more accurate moleculartools.

Colorectal Cancer (CRC)Exposure to heterocyclic amines is associ-

ated with colorectal cancer (CRC) in rodents(Layton et al., 1995). GSTM1, GSTT1, andGSTP1 are detoxification enzymes that havebeen shown to metabolize a wide range ofcarcinogens from tobacco smoke, diet, andenvironmental pollutants, including hetero-cyclic amines (Hirvonen, 1995). GST genepolymorphisms were reported to influenceinterindividual susceptibility to smoking-associated CRC (Koh et al., 2011). GSTenzymes are postulated to play a role inthe detoxification of colorectal carcinogensin tobacco smoke, and hence these play arole in CRC susceptibility. In a Spanish study,

Dow

nloa

ded

by [

Uni

vers

ity o

f W

inds

or]

at 1

8:48

14

Nov

embe

r 20

14

TOXICOGENETICS & CANCER IN LEBANESE 107

including 144 CRC patients and 329 healthycontrol individuals, GSTM1 and GSTT1 nulland double null genotypes were associatedwith up to fivefold higher CRC risk (Martinezet al., 2006). Similarly, in a comprehensivemeta-analysis, GSTM1 and GSTT1 null allelescarriers exhibited an increased CRC risk inCaucasian populations. No significant asso-ciations with CRC were detected for bothgenotypes in Chinese subjects (Economopoulosand Sergentanis, 2010). In addition, GSTP1 wasnot found to be associated with CRC risk indifferent ethnic groups, although one studyfound a possible protective effect for this allele(Hezova et al., 2012). Similar to Caucasians,the GSTM1 null genotype in Lebanese was alsoshown to be associated with a significant risein CRC (Darazy et al., 2011) (Table 1). In thissame case-control study, no marked associa-tions were found with either the CYP1A1∗2Aor the CYP2E1∗6 variant. Studies investigat-ing CYP and CRC in other ethnic groupsreport higher risk in carriers of the CYP2E1∗5Bc2 allele (Peng et al., 2013; Kiss et al., 2000)(Table 3).

Although NAT were not investigated forCRC in Lebanese, evidence indicates an asso-ciation between the NAT1 genotype and CRCrisk in many ethnic groups (Hein et al., 2000).In a study investigating NAT1 and NAT2 geneticpolymorphism in a group of 202 CRC patientsand 112 control subjects from Staffordshire,England, NAT1∗10 was found to be associ-ated with higher CRC risk (Bell et al., 1995b).However, rapid acetylation genotypes of NAT2were not a significant risk factor in this Englishpopulation. Similar findings on NAT1∗10 asso-ciation with CRC risk were noted in a studyamong 205 Chinese CRC patients and healthycontrols (Zhang et al., 2002). Another clinic-based case-control study reported a weak asso-ciation between NAT1∗10 and risk of colorectaladenomas in Caucasians exposed to hetero-cyclic amines (Ishibe et al., 2002). In contrast,other studies found no marked associationsbetween NAT1∗10 and CRC risk (Roemer et al.,2008; Katoh et al., 2000; Liu, Ding, et al.,2012) (Table 2).

Prostate Cancer (PCa)In a meta-analysis based on a group of stud-

ies including 9,934 cases and 16,459 controls,a significant association was found betweenthe GSTT1 null genotype and increased risk ofprostate cancer (PCa) in different ethnic groupsincluding Caucasians, Indians, and Asians (Yanget al., 2013). These findings were confirmedby additional studies in Caucasians and Asians(Zhu et al., 2013; Pan et al., 2012). Otherstudies in Indians suggested a role for bothGSTT1 and GSTP1 in PCa (Qadri et al., 2011;Thakur et al., 2011). In contrast, a meta-analysisshowed an increased risk for PCa in Caucasianand Asian carriers of the GSTM1 null genotype,while no associations were found with theGSTT1 and GSTP1 genotypes (Mo et al., 2009).Similar results were observed in a study onKorean men (Kwon et al., 2011) (Table 1).In addition, GSTM1 was linked to PCa suscep-tibility in Japanese (Murata et al., 2001), whileno marked associations between GSTs and PCawere found in Africans (Mo et al., 2009; Yanget al., 2013).

In a study among 225 Chinese PCa patientsand 250 age-matched controls, the PCa riskincreased in smokers and drinkers carryingthe CYP2E1 c1/c1 genotype, and fell in car-riers of the CYP2E1 c2/c2 genotype (Yanget al., 2006, 2009;). In contrast, no associa-tion was noted with CYP2E1∗5B polymorphismin Caucasians and in other studied multiethnicgroups (Ferreira et al., 2003; Joshi et al., 2012).However, the CYP2E1∗6 D allele was associatedwith a twofold elevated PCa risk in a group ofPortuguese (Table 3).

Results for NAT in PCa are also contra-dictory. Several studies explored associationsbetween acetylator genotypes and PCa, butnone in Lebanese. No significant relationshipwas found between NAT2 genotype and PCarisk, and N-acetylation activity levels in prostatetissues were independent of NAT2 genotype(Agundez et al., 1998; Wadelius et al., 1999;Gong et al., 2011). Moreover, overexpressionof NAT2 in rodent prostate tissue did not alterDNA adducts upon chemical exposure (Jianget al., 1999; Purewal et al., 2000). However, for

Dow

nloa

ded

by [

Uni

vers

ity o

f W

inds

or]

at 1

8:48

14

Nov

embe

r 20

14

108 H. R. DHAINI AND L. KOBEISSI

NAT1, an association between the homozygoteNAT1∗10 genotype and PCa risk was reportedin Japanese (Fukutome et al., 1999). Similarly,in Caucasians, a hospital-based matched case-control study showed that NAT1∗10 genotypeis significantly higher in PCa cases comparedto controls (Hein et al., 2002). The study alsoreported an even higher PCa risk for carriersof both the NAT1∗10 allele and the homozy-gous NAT2∗5 acetylator genotype. In contrast,results from a number of other studies in FinnishCaucasians, Asians, Africans, and multiethnicgroups do not support a role for NAT1∗10 inPCa (Gong et al., 2011; Sharma et al., 2010;Kidd et al., 2011a; 2011b) (Table 2).

Breast Cancer (BC)For breast cancer (BC), the younger age

at presentation, higher standardized incidencerates, and a more progressive disease at ini-tial diagnosis emphasize the need to identifypotential risk factors contributing to BC bur-den in Lebanon. Studies investigating a rolefor GST in various types of cancer reportedcontroversial results in BC (Strange and Fryer,1999). In a study including 718 Caucasian BCpatients, GST polymorphism (GSTM1, GSTT1,and GSTP1) did not modify disease risk inBRCA1 and BRCA2 carriers (Spurdle et al.,2010). Similar negative results were found inChinese (Sakoda et al., 2008; Sergentanis andEconomopoulos, 2010). In contrast, all threepolymorphisms, as well as the double nullgenotype, were shown to be risk factors forBC in Portuguese (Ramalhinho et al., 2011).In Iranians, GSTM1 and GSTP1, but not GSTT1,genetic polymorphisms were associated with anelevated risk of BC (Hashemi et al., 2012). In apopulation from western France, GSTP1 wasreported as a significant modifier of BC risk(Maugard et al., 2001). However, in this samestudy, GSTM1 genotypes did not emerge as arisk factor. On the other hand, GSTM1, GSTP1,and GSTT1 genes in Greeks were found to befrequently deregulated in BC (Dialyna et al.,2001). In Lebanese, one study was identified.In 227 BC women and 99 controls, no marked

associations were noted between GST vari-ants (GSTM1, GSTT1, and GSTP1) and BC risk(Zgheib et al., 2013) (Table 1).

Taiwanese women with the CYP2E1∗5Bc2/c2 genotype had a lower BC risk com-pared to those with the c1/c1 genotype(Wu et al., 2006). In contrast, other stud-ies on BC and DME in Caucasians andother ethnicities reported no significant asso-ciation for CYP2E1∗5B polymorphism withBC risk (McCarty et al., 2012; Iscan et al.,2001). Studies in Arab populations, includingLebanese, observed similar results. In a studyconducted among 314 Arab Tunisian BC casesand 246 controls, no marked associations werefound between BC risk and either CYP2E1or GST (Khedhaier et al., 2008). However, asignificant association was reported betweenCYP2D6 G/G wild type, as well as NAT2 slowacetylation, and breast carcinoma risk in post-menopausal patients (Khedhaier et al., 2008).In Lebanese, no significant differences werenoted between BC cases and controls for thedistribution of all three genes NAT2, GST, andCYP2E1 variants (Zgheib et al., 2013) (Tables 2and 3).

NAT1 enzymatic levels were shown to behigh in breast cancers (Adam et al., 2003;Wakefield et al., 2008). The overproductionof NAT1 in normal luminal epithelial breastcells was found to contribute to two of thehallmark traits of cancer: enhanced growthand resistance to drugs. Further, NAT1 wasidentified as an independent prognostic fac-tor of BC relapse in a group of 97 Frenchpostmenopausal patients (Bieche et al., 2004).Patients with strong NAT1 overexpression hada significantly higher relapse-free survival.Another study, using a nested case-controldesign (453 cases and 900 controls) conductedin postmenopausal Caucasian women in Iowa,reported a 30% elevated risk of BC associ-ated with NAT1∗10 allele, and a nearly four-fold elevated risk associated with the NAT1∗11allele (Zheng et al., 1999). The positive asso-ciation of BC with NAT1∗11 allele was moreevident among smokers and those who con-sumed high amounts of red meat. Similarly, inFrench-Canadians, NAT1∗10 allele conferred a

Dow

nloa

ded

by [

Uni

vers

ity o

f W

inds

or]

at 1

8:48

14

Nov

embe

r 20

14

TOXICOGENETICS & CANCER IN LEBANESE 109

fourfold increase in BC risk among women whoconsumed well-done meat (Krajinovic et al.,2001). In this same study, CYP1A1∗4 was alsoobserved to be involved in the susceptibil-ity to breast carcinoma. In contrast, studies inKoreans and Africans reported no marked asso-ciations between NAT1 polymorphisms and BCrisk (Lee et al., 2003; Millikan, 2000). Thusfar, no apparent studies explored NAT1 variantcontribution to BC risk in Lebanese (Table 2).

Lung Cancer (LC)In a case-control study conducted among

360 lung cancer (LC) Chinese patients and360 cancer-free controls, GSTM1-null genotypewas found to be significantly associated withLC risk. Moreover, smokers with GSTM1-nullgenotype who had smoked for more than30 pack-years had a 10-fold higher risk com-pared to nonsmoker wild-type carriers (Liuet al., 2012a). Similarly, another study inChinese demonstrated that heavy smokerswho carry GSTM1-null had a higher LC risk(Gu et al., 2007). In Japanese, a study on841 subjects reported an association betweenGSTM1 and GSTP1 null genotypes and higherLC risk (Kiyohara et al., 2012). The same studyshowed an even higher risk in subjects carry-ing a pertinent combination of multiple “at-risk”genotypes including GSTM1-null and CYP1A1mutants. However, in European Caucasians,no marked associations were found withGSTM1 and GSTT1 null genotypes (Gervasiniet al., 2010; To-Figueras et al., 1997). At thesame time, a GSTP1 genetic variant seems tobe linked to LC in an American Caucasian pop-ulation (Wang et al., 2003). Studies investigatingGST and LC in Turks reported findings similar tothose in Caucasians (Atinkaya et al., 2012; Adaet al., 2012) (Table 1).

Studies investigating CYP2E1 and LC foundcontradictory results among different ethnicgroups. CYP2E1∗5B c1/c1 and CYP2E1∗6 DDgenotypes are reported as predisposing fac-tors for LC in different Asian populations,while c2/c2 and CC genotypes were associatedwith a decreased risk (Su et al., 2011; Wanget al., 2010; Zhan et al., 2010). In contrast,

no associations were found between CYP2E1polymorphisms and LC risk in Caucasian andMexican populations (Perez-Morales et al.,2011; Zhan et al., 2010) (Table 3).

Early studies investigating associationsbetween N-acetylation and LC risk werenegative, or reported weak correlations forrapid NAT2 phenotype in smokers (Rootset al., 1988; Philip et al., 1988; Nyberg et al.,1998). However, one case-control study inFrench-Caucasian smokers noted a signifi-cant association between LC and NAT1 slowacetylator genotypes (Bouchardy et al., 1998).In the same study, no significant associa-tions with NAT2 were reported. Interestingly,NAT1∗14B slow acetylation was recently pos-tulated to be associated with higher risk ofsmoking-induced LC (Millner et al., 2012a).In contrast, a study in German Caucasiansfound a significantly increased risk for lungadenocarcinoma among NAT1 fast acetylators(Wikman et al., 2001). At the same time, studiesin African-Americans reported no marked asso-ciation between NAT1 genetic polymorphismand LC risk (Hein et al., 2000). In Lebanese, noapparent studies investigated the role of any ofthe DME in LC to date (Tables 1–3).

CONCLUSIONS AND FUTUREDIRECTIONS

Lebanon is a small country with a relativelysmall population size, located on the easterncoast of the Mediterranean. Cancer incidencehas increased substantially over the past decadewith little available data on potential etiolo-gies, risk factors, and population genetic profile.Nevertheless, based on the available environ-mental and molecular epidemiology data, anumber of important observations may be high-lighted. First, the Lebanese population is poten-tially exposed to excessive levels of variouschemicals from car emissions, diesel exhaust,fossil fuel power plants, waste incineration,water and soil pollution, war-related chemi-cal exposure, and tobacco smoking. Second,there is recurrent evidence that the populationis characterized by an unusually high frequency

Dow

nloa

ded

by [

Uni

vers

ity o

f W

inds

or]

at 1

8:48

14

Nov

embe

r 20

14

110 H. R. DHAINI AND L. KOBEISSI

of the slow acetylator allele NAT1∗14A. Third,Lebanese present a relatively high frequencyof the GSTM1 and GSTT1 combined nullgenotype compared to other studied popula-tions and ethnic groups. These observationssuggest that high malignancies incidence inLebanese, particularly bladder, lung, and breastcancer, may be following a gene–environmentinteraction model that still needs to be tested.This is based on a high environmental expo-sure and a distinctive genetic predispositionat the level of DME. Evidence with UBC inthe Lebanese population is in agreement withthis model (Dhaini and Levy, 2000; Yassineet al., 2012; Kobeissi et al., 2013). The rela-tively high prevalence of “high-risk alleles” andexcessive chemical exposure may be increasingthe attributable risk in this population.

This review identified a number of gaps inthe literature, as well as future research pri-orities for Lebanon. First, the significance ofthe high frequencies of NAT1∗14A and GSThomozygous null deletions and their contribu-tion to cancer risk in Lebanese requires furtherinvestigation. Large-scale studies targeting NAT1and GST genotypes and phenotypes are neces-sary to clarify their contribution to cancer riskin this country. NAT1 particular contribution tobreast and lung cancer risk in Lebanese requiresinvestigation. Although data suggest a role forNAT1∗10 and NAT1∗11 in BC risk, explor-ing this particular area is limited in Lebanese.Further, investigation of NAT1∗14A in high-riskgroups, such as smokers, may be important forLebanon in view of the double burden of thetwo high cancer incidences: lung and blad-der. However, screening might be useful onlywhen the role of NAT1∗14A in cancer risk isclarified and confirmed in Lebanese. While anindividualized UBC risk prediction nomogramdoes not exist at the moment, NAT1∗14A andother identified genetic polymorphisms in thefuture may provide significant promise for per-sonalized UBC risk assessment, particularly forLebanese men at risk of early-onset disease.These men stand to benefit from these find-ings. The possibility of a personalized UBC riskprediction assessment deserves to be exam-ined further and considered, since it will allow

younger men at increased genetic risk for UBCto follow a preventive lifestyle and concretescreening regimen with appropriate risk coun-seling, and enable other men who are foundnot to carry putative risk factors, includingidentified genetic polymorphisms, to be sparedunnecessary physical and psychological mor-bidity. Second, the complete lack of availabledata on the role of the different Phase I andPhase II DMEs in lung and prostate cancersusceptibility needs to be addressed. In viewof the observed discrepancies in associationsbetween different DME and cancers amongvarious ethnic groups, it becomes imperativeto study risk factors and etiology of diseasein the context of the communities where itarises. Third, in bladder cancer, NAT2 variantcontribution to increased risk requires a thor-ough investigation. A significantly higher riskwas reported earlier in carriers of the combinedNAT2 slow acetylator and NAT1 fast acetylatorin smokers, compared to individual genotypes(Sanderson et al., 2007). This indicates theimportance of considering joint effects betweenmultiple genetic and environmental factors inthe etiology of common complex diseases in apopulation.

At the same time, data on environmentallevels of many carcinogens as well as humanchemical exposure and associated toxicitiesin Lebanese are still scarce and need to beexpanded. Data on exposure to carcinogensneed to be assessed when possible, for amore accurate and reliable associated cancerrisk assessment. More studies are needed toassess health risks from chemical exposures,taking into account gene–environment inter-actions. A recent publication, Public Healthin the Arab World, revealed a clear gap intoxicology data and health risk assessment stud-ies in Arabs including the Lebanese population(Jabbour et al., 2012). Interestingly, the esti-mated lifetime cancer risk in humans fromchemical exposure was recently demonstratedto be inaccurate when genetic polymorphismsare ignored (Simmons and Portier, 2002).

In conclusion, current patterns of cancerincidence and revealed toxicogenetic profilein Lebanese may be associated. This calls for

Dow

nloa

ded

by [

Uni

vers

ity o

f W

inds

or]

at 1

8:48

14

Nov

embe

r 20

14

TOXICOGENETICS & CANCER IN LEBANESE 111

more research in order to confirm these find-ings in larger samples and to identify other riskfactors and potential interactions. Further stud-ies are needed on the way in which geneticpolymorphism informs about cancer risk, par-ticularly for UBC and BC in younger Lebanesemen and women. This also requires sustainingand developing the NCR in order to monitorchanges in cancer incidence. It also neces-sitates launching a periodic national censusfor an accurate enumeration of the popula-tion denominator and subsequent estimationof cancer incidence, despite sensitive religiousand political hindrances. These componentswill set the basis for risk managers and decisionmakers in adopting evidence-based preventivepolicies.

REFERENCES

Abou-Daoud, K. T. 1966. Morbidity from can-cer in Lebanon. Cancer 19: 1293–1300.

Ada, A. O., Kunak, S. C., Hancer, F., Soydas,E., Alpar, S., Gulhan, M., and Iscan, M.2012. Association between GSTM1, GSTT1,and GSTP1 polymorphisms and lung cancerrisk in a Turkish population. Mol. Biol. Rep.39:5985–5993.

Adam, P. J., Berry, J., Loader, J. A., Tyson,K. L., Craggs, G., Smith, P., De Belin, J.,Steers, G., Pezzella, F., Sachsenmeir, K.F., Stamps, A. C., Herath, A., Sim, E.,O’Hare, M. J., Harris, A. L., and Terrett,J. A. 2003. Arylamine N-acetyltransferase-1 is highly expressed in breast cancers andconveys enhanced growth and resistanceto etoposide in vitro. Mol. Cancer Res. 1:826–835.

Adib, S., and Daniel, J. 2006. Cancer inLebanon 2003. Lebanese Ministry of PublicHealth. http://www.cancer.org.lb/public/NationalCancerRegistryReport2003.pdf

Adib, S., Daniel, J., and Issa, G. 2008.Cancer in Lebanon 2004, with an update ofCancer 2003. Lebanese Ministry of PublicHealth. http://www.moph.gov.lb/Prevention/Surveillance/Pages/Cancer.aspx

Adib, S. M., Mufarrij, A. A., Shamseddine, A.I., Kahwaji, S. G., Issa, P., and el-Saghir, N.S. 1998. Cancer in Lebanon: An epidemi-ological review of the American Universityof Beirut Medical Center Tumor Registry(1983–1994). Ann. Epidemiol. 8: 46–51.

Agundez, J. A., Martinez, C., Olivera, M.,Gallardo, L., Ladero, J. M., Rosado, C., Prados,J., Rodriguez-Molina, J., Resel, L., andBenitez,J. 1998. Expression in human prostate ofdrug- and carcinogen-metabolizing enzymes:Association with prostate cancer risk. Br. J.Cancer 78: 1361–1367.

Arylamine N-acetyltransferase GeneNomenclature Committee. 2011. HumanNAT1 alleles (haplotypes). http://n-acetyltransferasenomenclature.louisville.edu(accessed May 8, 2012).

Atinkaya, C., Taspinar, M., Sakiragaoglu, O.,Oz, G., Yazici, U., Oztuna, D., Tastepe,I., and Sunguroglu, A. 2012. The effect ofCYP1A1, GSTT1 and GSTM1 polymorphismson the risk of lung cancer: A case-controlstudy. Hum. Exp. Toxicol. 31: 107–1080.

Bailey, L. R., Roodi, N., Verrier, C. S., Yee,C. J., Dupont, W. D., and Parl, F. F. 1998.Breast cancer and CYPIA1, GSTM1, andGSTT1 polymorphisms: Evidence of a lackof association in Caucasians and AfricanAmericans. Cancer Res. 58: 65–70.

Basma, H. A., Kobeissi, L. H., Jabbour, M. E.,Moussa, M. A., and Dhaini, H. R. 2013.CYP2E1 and NQO1 genotypes and bladdercancer risk in a Lebanese population. Int. J.Mol. Epidemiol. Genet. 4: 207–217.

Bell, D. A., Badawi, A. F., Lang, N. P., Ilett, K.F., Kadlubar, F. F., and Hirvonen, A. 1995a.Polymorphism in the N-acetyltransferase 1(NAT1) polyadenylation signal: Associationof NAT1∗10 allele with higher N-acetylationactivity in bladder and colon tissue. CancerRes. 55: 5226–5229.

Bell, D. A., Stephens, E. A., Castranio, T.,Umbach, D. M., Watson, M., Deakin, M.,Elder, J., Hendrickse, C., Duncan, H., andStrange, R. C. 1995b. Polyadenylation poly-morphism in the acetyltransferase 1 gene(NAT1) increases risk of colorectal cancer.Cancer Res. 55: 3537–3542.

Dow

nloa

ded

by [

Uni

vers

ity o

f W

inds

or]

at 1

8:48

14

Nov

embe

r 20

14

112 H. R. DHAINI AND L. KOBEISSI

Bell, D. A., Taylor, J. A., Butler, M. A.,Stephens, E. A., Wiest, J., Brubaker, L.H., Kadlubar, F. F., and Lucier, G. W.1993. Genotype/phenotype discordancefor human arylamine N-acetyltransferase(NAT2) reveals a new slow-acetylatorallele common in African-Americans.Carcinogenesis 14: 1689–1692.

Belpaire, F. M., and Bogaert, M. G. 1996.Cytochrome P450: Genetic polymorphismand drug interactions. Acta Clin. Belg. 51:254–260.

Berber, U., Yilmaz, I., Yilmaz, O., Haholu,A., Kucukodaci, Z., Ates, F., and Demirel,D. 2013. CYP1A1 (Ile462Val), CYP1B1(Ala119Ser and Val432Leu), GSTM1 (null),and GSTT1 (null) polymorphisms and blad-der cancer risk in a Turkish population. AsianPac. J. Cancer Prev. 14: 3925–3329.

Bernstam, L., and Nriagu, J. 2000. Molecularaspects of arsenic stress. J. Toxicol. Environ.Health B 3: 293–322.

Bieche, I., Girault, I., Urbain, E., Tozlu, S., andLidereau, R. 2004. Relationship betweenintratumoral expression of genes codingfor xenobiotic-metabolizing enzymes andbenefit from adjuvant tamoxifen in estro-gen receptor alpha-positive postmenopausalbreast carcinoma. Breast Cancer Res. 6:R252–R263.

Blakely, T., Barendregt, J. J., Foster, R. H., Hill,S., Atkinson, J., Sarfati, D., and Edwards, R.2013. The association of active smoking withmultiple cancers: National census-cancerregistry cohorts with quantitative bias anal-ysis. Cancer Causes Control 24: 1243–1255.

Bolt, H. M., Roos, P. H., and Thier, R. 2003. Thecytochrome P-450 isoenzyme CYP2E1 in thebiological processing of industrial chemicals:consequences for occupational and environ-mental medicine. Int. Arch. Occup. Environ.Health 76: 174–185.

Borlak, J., and Reamon-Buettner, S. M.2006. N-Acetyltransferase 2 (NAT2) genepolymorphisms in colon and lung cancerpatients. BMC Med. Genet. 7: 58.

Bouchardy, C., Mitrunen, K., Wikman,H., Husgafvel-Pursiainen, K., Dayer, P.,

Benhamou, S., and Hirvonen, A. 1998. N-Acetyltransferase NAT1 and NAT2 genotypesand lung cancer risk. Pharmacogenetics 8:291–298.

Brennan, P. 2002. Gene–environment interac-tion and aetiology of cancer: What doesit mean and how can we measure it?Carcinogenesis 23: 381–387.

Brockmoller, J., Cascorbi, I., Kerb, R., andRoots, I. 1996. Combined analysis ofinherited polymorphisms in arylamine N-acetyltransferase 2, glutathione-S-transferaseM1, and T1, microsomal epoxide hydrolase,and cytochrome P450 enzymes as modula-tors of bladder cancer risk. Cancer Res. 56:3915–3925.

Bruhn, C., Brockmoller, J., Cascorbi, I., Roots,I., and Borchert, H. H. 1999. Correlationbetween genotype and phenotype ofthe human arylamine N-acetyltransferasetype 1 (NAT1). Biochem. Pharmacol. 58:1759–1764.

Bu, R., Gutierrez, M. I., Al-Rasheed, M.,Belgaumi, A., and Bhatia, K. 2004. Variabledrug metabolism genes in Arab population.Pharmacogenomics J. 4: 260–266.

Butcher, N., Ilett, K., and Minchin, R. 1998.Functional polymorphism of the humanarylamine N-acetyltransferase type 1 genecaused by C190T and G560A mutations.Pharmacogenetics 8:67–72.

Butcher, N. J., and Minchin, R. F. 2012.Arylamine N-acetyltransferase 1: A noveldrug target in cancer development.Pharmacol. Rev. 64: 147–165.

Canadian Council of Ministers of theEnvironment. 1999. Canadian SoilQuality Guidelines for the Protection ofEnvironmental and Human Health. http://ceqg-rcqe.ccme.ca

Cantor, K. P., Villanueva, C. M., Silverman,D. T., Figueroa, J. D., Real, F. X., Garcia-Closas, M., Malats, N., Chanock, S., Yeager,M., Tardon, A., Garcia-Closas, R., Serra, C.,Carrato, A., Castano-Vinyals, G., Samanic,C., Rothman, N., and Kogevinas, M. 2010.Polymorphisms in GSTT1, GSTZ1, andCYP2E1, disinfection by-products, and risk

Dow

nloa

ded

by [

Uni

vers

ity o

f W

inds

or]

at 1

8:48

14

Nov

embe

r 20

14

TOXICOGENETICS & CANCER IN LEBANESE 113

of bladder cancer in Spain. Environ. HealthPerspect. 118:1545–1550.

Cartwright, R. A., Glashan, R. W., Rogers,H. J., Ahmad, R. A., Barham-Hall, D.,Higgins, E., and Kahn, M. A. 1982. Roleof N-acetyltransferase phenotypes in bladdercarcinogenesis: A pharmacogenetic epidemi-ological approach to bladder cancer. Lancet2: 842–845.

Cascorbi, I., Roots, I., and Brockmöller,J. 2001. Association of NAT1 andNAT2 polymorphisms to urinary blad-der cancer: Significantly reduced risk insubjects with NAT1∗10. Cancer Res. 61:5051–5056.

Chaaya, M., El-Roueiheb, Z., Chemaitelly, H.,Azar, G., Nasr, J., and Al-Sahab, B. 2004.Argileh smoking among university students:A new tobacco epidemic. Nicotine TobaccoRes. 6: 457–463.

Chen, B., Zhou, Y., Yang, P., and Wu, X. T. 2010.Glutathione S-transferase M1 gene polymor-phism and gastric cancer risk: An updatedanalysis. Arch. Med. Res. 41: 558–566.

Chen, C. L., Liu, Q., Pui, C. H., Rivera, G.K., Sandlund, J. T., Ribeiro, R., Evans, W. E.,and Relling, M. V. 1997. Higher frequencyof glutathione S-transferase deletions in blackchildren with acute lymphoblastic leukemia.Blood 89: 1701–1707.

Chen, H. C., Hu, W. X., Liu, Q. X., Li, W. K.,Chen, F. Z., Rao, Z. Z., Liu, X. F., Luo, Y. P.,and Cao, Y. F. 2008. Genetic polymorphismsof metabolic enzymes CYP1A1, CYP2D6,GSTM1 and GSTT1 and leukemia suscepti-bility. Eur. J. Cancer Prev. 17: 251–258.

Cho, H. J., Lee, S. Y., Ki, C. S., andKim, J. W. 2005. GSTM1, GSTT1 andGSTP1 polymorphisms in the Korean popu-lation. J. Korean Med. Sci. 20: 1089–1092.

Choi, J. Y., Lee, K. M., Cho, S. H., Kim, S. W.,Choi, H. Y., Lee, S. Y., Im, H. J., Yoon, K.J., Choi, H., Choi, I., Hirvonen, A., Hayes,R. B., and Kang, D. 2003. CYP2E1 andNQO1 genotypes, smoking and bladder can-cer. Pharmacogenetics 13: 349–355.

Chowbay, B., Zhou, S., and Lee, E. J. 2005. Aninterethnic comparison of polymorphismsof the genes encoding drug-metabolizing

enzymes and drug transporters: Experiencein Singapore. Drug Metab. Rev. 37:327–378.

Coppini, G., De Dominicis, M., Zodiatis, G.,Lardner, R., Pinardi, N., Santoleri, R., Colella,S., Bignami, F., Hayes, D. R., Soloviev, D.,Georgiou, G., and Kallos, G. 2011. Hindcastof oil-spill pollution during the Lebanon cri-sis in the eastern Mediterranean, July–August2006. Mar. Pollut. Bull. 62: 140–153.

Cox, D. G., Dostal, L., Hunter, D. J., LeMarchand, L., Hoover, R., Ziegler, R. G.,and Thun, M. J. 2011. N-Acetyltransferase2 polymorphisms, tobacco smoking, andbreast cancer risk in the breast and prostatecancer cohort consortium. Am. J. Epidemiol.174: 1316–1322.

Cui, X., Lu, X., Hiura, M., Omori, H., Miyazaki,W., and Katoh, T. 2013. Association of geno-types of carcinogen-metabolizing enzymesand smoking status with bladder cancer ina Japanese population. Environ. Health Prev.Med. 18: 136–142.

Darazy, M., Balbaa, M., Mugharbil, A., Saeed,H., Sidani, H., and Abdel-Razzak, Z. 2011.CYP1A1, CYP2E1, and GSTM1 genepolymorphisms and susceptibility tocolorectal and gastric cancer amongLebanese. Genet. Test Mol. Biomarkers 15:423–429.

Dhaini, H., and Levy, G. 2000. Arylamine N-acetyltransferase 1 (NAT1) genotypes in aLebanese population. Pharmacogenetics 10:79–83.

Dialyna, I. A., Arvanitis, D. A., and Spandidos,D. A. 2001. Genetic polymorphisms andtranscriptional pattern analysis of CYP1A1,AhR, GSTM1, GSTP1 and GSTT1 genes inbreast cancer. Int. J. Mol. Med. 8: 79–87.

Economopoulos, K. P., and Sergentanis, T. N.2010. GSTM1, GSTT1, GSTP1, GSTA1 andcolorectal cancer risk: A comprehensivemeta-analysis. Eur. J. Cancer 46: 1617–1631.

El Saghir, N. S., Adib, S., Mufarrij, A., Kahwaji,S., Taher, A., Issa, P., and Shamseddine, A.I. 1998. Cancer in Lebanon: Analysis of10,220 cases from the American Universityof Beirut Medical Center. J. Med. Liban. 46:4–11.

Dow

nloa

ded

by [

Uni

vers

ity o

f W

inds

or]

at 1

8:48

14

Nov

embe

r 20

14

114 H. R. DHAINI AND L. KOBEISSI

El Saghir, N. S., Khalil, M. K., Eid, T., El Kinge, A.R., Charafeddine, M., Geara, F., Seoud, M.,and Shamseddine, A. I. 2007. Trends in epi-demiology and management of breast cancerin developing Arab countries: A literatureand registry analysis. Int. J. Surg. 5:225–233.

El Saghir, N. S., Seoud, M., Khalil, M. K.,Charafeddine, M., Salem, Z. K., Geara, F.B., and Shamseddine, A. I. 2006. Effectsof young age at presentation on survival inbreast cancer. BMC Cancer 6: 194.

El-Fadel, M., Abdallah, R., and Rachid,G. 2012. A modeling approach towardoil spill management along the easternMediterranean. J. Environ. Manage. 113:93–102.

El-Fadel, M., and Massoud, M. 2000.Particulate matter in urban areas: Health-based economic assessment. Sci. TotalEnviron. 257: 133–146.

Evans, D. A., Paterson, S., Francisco, P., andAlvarez, G. 1985. The acetylator phenotypesof Saudi Arabian diabetics. J. Med. Genet.22: 479–483.

Fakhoury, H. M., Noureddine, S., Tamim,H., Chmaisse, H., and Makki, R. 2012.Association of MMP3-1171(5A>6A) poly-morphism with lung cancer in Lebanon.Genet. Test Mol. Biomarkers 16: 988–990.

Farker, K., Lehmann, M. H., Kastner, R.,Hoffmann, A., Janitzky, V., Schubert, J., Matz,U., and Hofmann, W. 1998a. CYP2E1 geno-typing in renal cell/urothelial cancer patientsin comparison with control populations. Int.J. Clin. Pharmacol. Ther. 36: 463–468.

Farker, K., Lehmann, M. H., Oelschlagel,B., Haerting, J., Hoffmann, A., Janitzky,V., and Schubert, J. 1998b. Impact ofCYP2E1 genotype in renal cell and urothe-lial cancer patients. Exp. Toxicol. Pathol. 50:425–431.

Feng, J., Pan, X., Yu, J., Chen, Z., Xu, H., El-Rifai,W., Zhang, G., and Xu, Z. 2012. FunctionalPstI/RsaI polymorphism in CYP2E1 is associ-ated with the development, progression andpoor outcome of gastric cancer. PLoS One 7:e44478.

Ferlay, J., Shin, H., Bray, F., Forman, D.,Mathers, C., and Parkin, D. 2010. Estimates

of worldwide burden of cancer in 2008:GLOBOCAN 2008. Int. J. Cancer 127:2893–2917.

Ferreira, P. M., Medeiros, R., Vasconcelos, A.,Costa, S., Pinto, D., Morais, A., Oliveira, J.,and Lopes, C. 2003. Association betweenCYP2E1 polymorphisms and susceptibility toprostate cancer. Eur. J. Cancer Prev. 12:205–211.

Fukutome, K., Watanabe, M., Shiraishi, T.,Murata, M., Uemura, H., Kubota, Y.,Kawamura, J., Ito, H., and Yatani, R. 1999.N-Acetyltransferase 1 genetic polymorphisminfluences the risk of prostate cancer devel-opment. Cancer Lett. 136: 83–87.

Garcia-Closas, M., Malats, N., Silverman, D.,Dosemeci, M., Kogevinas, M., Hein, D. W.,Tardon, A., Serra, C., Carrato, A., Garcia-Closas, R., Lloreta, J., Castano-Vinyals,G., Yeager, M., Welch, R., Chanock, S.,Chatterjee, N., Wacholder, S., Samanic, C.,Tora, M., Fernandez, F., Real, F. X., andRothman, N. 2005. NAT2 slow acetylation,GSTM1 null genotype, and risk of bladdercancer: Results from the Spanish BladderCancer Study and meta-analyses. Lancet366: 649–659.

Garcia-Martin, E. 2008. Interethnic andintraethnic variability of NAT2 singlenucleotide polymorphisms. Curr. DrugMetab. 9: 487–497.

Garte, S., Gaspari, L., Alexandrie, A. K.,Ambrosone, C., Autrup, H., Autrup, J. L.,Baranova, H., Bathum, L., Benhamou, S.,Boffetta, P., Bouchardy, C., Breskvar, K.,Brockmoller, J., Cascorbi, I., Clapper, M.L., Coutelle, C., Daly, A., Dell’Omo, M.,Dolzan, V., Dresler, C. M., Fryer, A., Haugen,A., Hein, D. W., Hildesheim, A., Hirvonen,A., Hsieh, L. L., Ingelman-Sundberg, M.,Kalina, I., Kang, D., Kihara, M., Kiyohara,C., Kremers, P., Lazarus, P., Le Marchand, L.,Lechner, M. C., van Lieshout, E. M., London,S., Manni, J. J., Maugard, C. M., Morita, S.,Nazar-Stewart, V., Noda, K., Oda, Y., Parl,F. F., Pastorelli, R., Persson, I., Peters, W. H.,Rannug, A., Rebbeck, T., Risch, A., Roelandt,L., Romkes, M., Ryberg, D., Salagovic, J.,Schoket, B., Seidegard, J., Shields, P. G., Sim,

Dow

nloa

ded

by [

Uni

vers

ity o

f W

inds

or]

at 1

8:48

14

Nov

embe

r 20

14

TOXICOGENETICS & CANCER IN LEBANESE 115

E., Sinnet, D., Strange, R. C., Stucker, I.,Sugimura, H., To-Figueras, J., Vineis, P., Yu,M. C., and Taioli, E. 2001. Metabolic genepolymorphism frequencies in control popu-lations. Cancer Epidemiol. Biomarkers Prev.10: 1239–12348.

Geller, F., Urfer, W. and Golka, K. 2008.Bladder cancer and occupational exposuresin North Rhine-Westphalia, Germany. J.Toxicol. Environ. Health A 71: 856–858.

Gervasini, G., San Jose, C., Carrillo, J. A.,Benitez, J., and Cabanillas, A. 2010. GSTpolymorphisms interact with dietary fac-tors to modulate lung cancer risk: Studyin a high-incidence area. Nutr. Cancer 62:750–758.

Ghadirian, P., Robidoux, A., Nassif, E., Martin,G., Potvin, C., Patocskai, E., Younan,R., Larouche, N., Venne, A., Zhang, S.,Royer, R., and Narod, S. 2014. Screeningfor BRCA1 and BRCA2 mutations amongFrench-Canadian breast cancer cases attend-ing an outpatient clinic in Montreal. Clin.Genet. 85: 31–35.

Ginsberg, G., Smolenski, S., Hattis, D., Guyton,K. Z., Johns, D. O., and Sonawane, B.2009. Genetic polymorphism in glutathionetransferases (GST): Population distribution ofGSTM1, T1, and P1 conjugating activity. J.Toxicol. Environ. Health B 12: 389–439.

Golka, K., Heitmann, P., Giesler, F., Hodzic,J., Masche, N., Bolt, H. M., and Geller,F. 2008. Elevated bladder cancer risk dueto colorants—A statewide case-control studyin North Rhine-Westphalia, Germany. J.Toxicol. Environ. Health A 71: 851–855.

Gong, C., Hu, X., Gao, Y., Cao, Y., Gao,F., and Mo, Z. 2011. A meta-analysis ofthe NAT1 and NAT2 polymorphisms andprostate cancer: A HUgE review. Med.Oncol. 28: 365–376.

Gong, M., Dong, W., and An, R. 2012.Glutathione S-transferase T1 polymorphismcontributes to bladder cancer risk: A meta-analysis involving 50 studies. DNA Cell Biol.31: 1187–1197.

Gonzalez, F. J. 2007. The 2006 Bernard B.Brodie Award lecture. CYP2E1. Drug Metab.Dispos. 35: 1–8.

Green, J., Banks, E., Berrington, A., Darby,S., Deo, H., and Newton, R. 2000. N-Acetyltransferase 2 and bladder cancer: Anoverview and consideration of the evi-dence for gene-environment interaction. Br.J. Cancer 83: 412–417.

Gsur, A., Haidinger, G., Hinteregger, S.,Bernhofer, G., Schatzl, G., Madersbacher,S., Marberger, M., Vutuc, C., and Micksche,M. 2001. Polymorphisms of glutathione-S-transferase genes (GSTP1, GSTM1 andGSTT1) and prostate-cancer risk. Int. J.Cancer 95: 152–155.

Gu, Y. F., Zhang, Z. D., Zhang, S. C., Zheng,S. H., Jia, H. Y., and Gu, S. X. 2007.[Combined effects of genetic polymorphismsin cytochrome P450s and GSTM1 on lungcancer susceptibility]. Zhonghua Yi Xue Za Zhi87: 3064–3068.

Guo, X., Zeng, Y., Deng, H., Liao, J., Zheng, Y.,Li, J., Kessing, B., and O’Brien, S. J. 2010.Genetic polymorphisms of CYP2E1, GSTP1,NQO1 and MPO and the risk of nasopharyn-geal carcinoma in a Han Chinese populationof Southern China. BMC Res. Notes 3: 212.

Halwani, J., Baroudi, B. O., and Wartel,M. 1999. [Nitrate contamination of thegroundwater of the Akkar Plain in northernLebanon]. Sante 9: 219–223.

Hanke, J., and Krajewska, B. 1990. Acetylationphenotypes and bladder cancer. J. Occup.Med. 32: 917–918.

Hashemi, M., Eskandari-Nasab, E., Fazaeli,A., Taheri, M., Rezaei, H., Mashhadi, M.,Arbabi, F., Kaykhaei, M. A., Jahantigh, M.,and Bahari, G. 2012. Association betweenpolymorphisms of glutathione S-transferasegenes (GSTM1, GSTP1 and GSTT1) andbreast cancer risk in a sample Iranian pop-ulation. Biomarkers Med. 6: 797–803.

Hayashi, S., Watanabe, J., and Kawajiri,K. 1991. Genetic polymorphisms in the5′-flanking region change transcriptionalregulation of the human cytochromeP450IIE1 gene. J. Biochem. 110: 559–565.

Hayes, R. B., Bi, W., Rothman, N., Broly, F.,Caporaso, N., Feng, P., You, X., Yin, S.,Woosley, R. L., and Meyer, U. A. 1993.N-Acetylation phenotype and genotype and

Dow

nloa

ded

by [

Uni

vers

ity o

f W

inds

or]

at 1

8:48

14

Nov

embe

r 20

14

116 H. R. DHAINI AND L. KOBEISSI

risk of bladder cancer in benzidine-exposedworkers. Carcinogenesis 14: 675–678.

Hein, D., Doll, M., Fretland, A., Leff, M., Webb,S., Xiao, G., Devanaboyina, U., Nangju,N., and Feng, Y. 2000. Molecular genet-ics and epidemiology of the NAT1 andNAT2 acetylation polymorphism. CancerEpidemiol. Biomarkers Prev. 9: 29–42.

Hein, D., Grant, D., and Sim, E. 2000. Updateon consensus arylamine N-acetyltransferasegene nomenclature. Pharmacogenetics 10:291–292.

Hein, D., Leff, M., Ishibe, N., Sinha, R., Frazier,H., Doll, M., Xiao, G., Weinrich, M., andCaporaso, N. 2002. Association of prostatecancer with rapid N-acetyltransferase 1(NAT1∗10) in combination with slow N-acetyltransferase 2 acetylator genotypes ina pilot case-control study. Environ. Mol.Mutagen. 40: 161–167.

Hein, D. W. 1988. Acetylator genotype andarylamine-induced carcinogenesis. Biochim.Biophys. Acta 948: 37–66.

Hein, D. W. 2009. N-Acetyltransferase SNPs:Emerging concepts serve as a paradigm forunderstanding complexities of personalizedmedicine. Expert Opin. Drug Metab. Toxicol.5: 353–366.

Hezova, R., Bienertova-Vasku, J., Sachlova,M., Brezkova, V., Vasku, A., Svoboda, M.,Radova, L., Kiss, I., Vyzula, R., and Slaby, O.2012. Common polymorphisms in GSTM1,GSTT1, GSTP1, GSTA1 and susceptibility tocolorectal cancer in the Central Europeanpopulation. Eur. J. Med. Res. 17: 17.

Hirvonen, A. 1995. Genetic factors in individ-ual responses to environmental exposures. J.Occup. Environ. Med. 37: 37–43.

Hoffmann, D., and Hoffmann, I. 1997. Thechanging cigarette, 1950–1995. J. Toxicol.Environ. Health 50: 307–364.

Hou, D. F., Wang, S. L., He, Z. M., Yang, F., andChen, Z. C. 2007. Expression of CYP2E1 inhuman nasopharynx and its metaboliceffect in vitro. Mol. Cell. Biochem. 298:93–100.

Houri, A., and El Jeblawi, S. W. 2007. Waterquality assessment of Lebanese coastal riversduring dry season and pollution load into

the Mediterranean Sea. J. Water Health 5:615–623.

Hubbard, A. L., Moyes, C., Wyllie, A. H., Smith,C. A., and Harrison, D. J. 1998. N-Acetyltransferase 1: Two polymorphisms in cod-ing sequence identified in colorectal cancerpatients. Br. J. Cancer 77: 913–916.

Hughes, N., Janezic, S., McQueen, K., Jewett,M., Castranio, T., Bell, D., and Grant,D. 1998. Identification and characteriza-tion of variant alleles of human acetyltran-ferase NAT1 with defective function usingp-aminosalicylate as an in-vivo and in-vitroprobe. Pharmacogenetics 8: 55–66.

International Agency for Research on Cancer.2012. IARC Monographs on the Evaluationof Carcinogenic Risks to Humans, vol. 100.World Health Organization Press. http://apps.who.int/bookorders/anglais/detart1.jsp?codlan=1&codcol=72&codcch=100

Iscan, M., Klaavuniemi, T., Coban, T.,Kapucuoglu, N., Pelkonen, O., and Raunio,H. 2001. The expression of cytochromeP450 enzymes in human breast tumoursand normal breast tissue. Breast Cancer Res.Treat. 70: 47–54.

Ishibe, N., Sinha, R., Hein, D. W., Kulldorff, M.,Strickland, P., Fretland, A. J., Chow, W. H.,Kadlubar, F. F., Lang, N. P., and Rothman,N. 2002. Genetic polymorphisms in hetero-cyclic amine metabolism and risk of colorectaladenomas. Pharmacogenetics 12: 145–150.

Jabbour, S., Giacaman, R., Khawaja, M.,Nuwayhid, I., and Yamout, R. 2012. Publichealth in the Arab world, vol. 1. Cambridge,UK: Cambridge University Press.

Jalkh, N., Nassar-Slaba, J., Chouery, E.,Salem, N., Uhrchammer, N., Golmard,L., Stoppa-Lyonnet, D., Bignon, Y. J.,and Megarbane, A. 2012. Prevalance ofBRCA1 and BRCA2 mutations in familialbreast cancer patients in Lebanon. Hered.Cancer Clin. Pract. 10: 7.

Jiang, W., Feng, Y., and Hein, D. 1999.Higher DNA adduct levels in urinarybladder and prostate of slow acetylatorinbred rats administered 3,2’-dimethyl-4-aminobiphenyl. Toxicol. Appl. Pharmacol.156: 187–194.

Dow

nloa

ded

by [

Uni

vers

ity o

f W

inds

or]

at 1

8:48

14

Nov

embe

r 20

14

TOXICOGENETICS & CANCER IN LEBANESE 117

Joshi, A. D., Corral, R., Catsburg, C., Lewinger,J. P., Koo, J., John, E. M., Ingles, S. A.,and Stern, M. C. 2012. Red meat andpoultry, cooking practices, genetic suscep-tibility and risk of prostate cancer: Resultsfrom a multiethnic case-control study.Carcinogenesis 33: 2108–2118.

Karim, A. K., Elfellah, M. S., and Evans, D.A. 1981. Human acetylator polymorphism:Estimate of allele frequency in Libya anddetails of global distribution. J. Med. Genet.18: 325–330.

Kassir, L. N., Lartiges, B., and Ouaini, N. 2012.Effects of fertilizer industry emissions on localsoil contamination: A case study of a phos-phate plant on the east Mediterranean coast.Environ. Technol. 33: 873–885.

Katoh, T., Boissy, R., Nagata, N., Kitagawa,K., Kuroda, Y., Itoh, H., Kawamoto, T., andBell, D. A. 2000. Inherited polymorphismin the N-acetyltransferase 1 (NAT1) and 2(NAT2) genes and susceptibility to gastric andcolorectal adenocarcinoma. Int. J. Cancer85: 46–49.

Katurji, M., Daher, N., Sheheitli, H., Saleh, R.,and Shihadeh, A. 2010. Direct measurementof toxicants inhaled by water pipe users inthe natural environment using a real-time insitu sampling technique. Inhal. Toxicol. 22:1101–1109.

Kayaalti, Z., and Soylemezoglu, T. 2010.Distribution of ADH1B, ALDH2, CYP2E1 ∗6,and CYP2E1 ∗7B genotypes in Turkish popu-lation. Alcohol 44: 415–423.

Khattab, A., Javaid, A., Iraqi, G., Alzaabi, A.,Ben Kheder, A., Koniski, M. L., Shahrour, N.,Taright, S., Idrees, M., Polatli, M., Rashid, N.,and El Hasnaoui, A. 2012. Smoking habits inthe Middle East and North Africa: Results ofthe BREATHE study. Respir. Med. 106(suppl.2): S16–S24.

Khedhaier, A., Hassen, E., Bouaouina, N.,Gabbouj, S., Ahmed, S. B., and Chouchane,L. 2008. Implication of xenobiotic metab-olizing enzyme gene (CYP2E1, CYP2C19,CYP2D6, mEH and NAT2) polymorphisms inbreast carcinoma. BMC Cancer 8: 109.

Kidd, L. C., Vancleave, T. T., Doll, M. A.,Srivastava, D. S., Thacker, B., Komolafe,

O., Pihur, V., Brock, G. N., and Hein, D.W. 2011a. No association between variantN-acetyltransferase genes, cigarette smok-ing and prostate cancer susceptibility amongmen of African descent. Biomarkers Cancer2011: 1–13.

Kidd, L. R., Hein, D. W., Woodson, K.,Taylor, P. R., Albanes, D., Virtamo, J., andTangrea, J. A. 2011b. Lack of association ofthe N-acetyltransferase NAT1∗10 allele withprostate cancer incidence, grade, or stageamong smokers in Finland. Biochem. Genet.49: 73–82.

Kiss, I., Sandor, J., Pajkos, G., Bogner, B.,Hegedus, G., and Ember, I. 2000. Colorectalcancer risk in relation to genetic poly-morphism of cytochrome P450 1A1, 2E1,and glutathione-S-transferase M1 enzymes.Anticancer Res. 20:519–522.

Kiyohara, C., Horiuchi, T., Takayama, K., andNakanishi, Y. 2012. Genetic polymorphismsinvolved in carcinogen metabolism and DNArepair and lung cancer risk in a Japanesepopulation. J. Thorac. Oncol. 7: 954–962.

Kobeissi, L. H., Yassine, I. A., Jabbour, M.E., Moussa, M. A., and Dhaini, H. R.2013. Urinary bladder cancer risk factors:A Lebanese case-control study. Asian Pac. J.Cancer Prev. 14: 3205–3211.

Koh, W. P., Nelson, H. H., Yuan, J. M., Vanden Berg, D., Jin, A., Wang, R., and Yu,M. C. 2011. Glutathione S-transferase (GST)gene polymorphisms, cigarette smoking andcolorectal cancer risk among Chinese inSingapore. Carcinogenesis 32: 1507–1511.

Korfali, S. I., and Jurdi, M. 2007. Assessmentof domestic water quality: Case study,Beirut, Lebanon. Environ. Monit. Assess. 135:241–251.

Koutros, S., Silverman, D. T., Baris, D., Zahm,S. H., Morton, L. M., Colt, J. S., Hein, D.W., Moore, L. E., Johnson, A., Schwenn,M., Cherala, S., Schned, A., Doll, M. A.,Rothman, N., and Karagas, M. R. 2011. Hairdye use and risk of bladder cancer in theNew England bladder cancer study. Int. J.Cancer 129: 2894–2904.

Krajinovic, M., Ghadirian, P., Richer, C., Sinnett,H., Gandini, S., Perret, C., Lacroix, A.,

Dow

nloa

ded

by [

Uni

vers

ity o

f W

inds

or]

at 1

8:48

14

Nov

embe

r 20

14

118 H. R. DHAINI AND L. KOBEISSI

Labuda, D., and Sinnett, D. 2001. Geneticsusceptibility to breast cancer in French-Canadians: Role of carcinogen-metabolizingenzymes and gene–environment interac-tions. Int. J. Cancer 92: 220–225.

Kurian, A. W. 2010. BRCA1 and BRCA2 muta-tions across race and ethnicity: Distributionand clinical implications. Curr. Opin. Obstet.Gynecol. 22: 72–78.

Kwon, D. D., Lee, J. W., Han, D. Y., Seo, I. Y.,Park, S. C., Jeong, H. J., Yang, Y. S., Chae, S.C., Na, K. S., Mo, K. J., Kim, J. J., and Rim, J. S.2011. Relationship between the glutathione-S-transferase P1, M1, and T1 genotypesand prostate cancer risk in Korean subjects.Korean J. Urol. 52: 247–252.

Lacko, M., Oude Ophuis, M. B., Peters,W. H., and Manni, J. J. 2009. Geneticpolymorphisms of smoking-related carcino-gen detoxifying enzymes and head and neckcancer susceptibility. Anticancer Res. 29:753–761.

Ladero, J. M., Kwok, C. K., Jara, C., Fernandez,L., Silmi, A. M., Tapia, D., and Uson, A.C. 1985. Hepatic acetylator phenotype inbladder cancer patients. Ann. Clin. Res. 17:96–99.

Lakkis, N. A., Adib, S. M., Osman, M. H.,Musharafieh, U. M., and Hamadeh, G. N.2010. Breast cancer in Lebanon: Incidenceand comparison to regional and Westerncountries. Cancer Epidemiol. 34: 221–325.

Layton, D. W., Bogen, K. T., Knize, M. G.,Hatch, F. T., Johnson, V. M., and Felton, J. S.1995. Cancer risk of heterocyclic amines incooked foods: An analysis and implicationsfor research. Carcinogenesis 16: 39–52.

Lebanese Ministry of Environment, UnitedNations Environment Programme (UNEP),and Global Environment Facility (GEF).2005. National profile on management ofchemicals in Lebanon. http://test.moe.gov.lb/pdf/Pops/NationalProfileonChemicalsManagementinLebanon.pdf

Lebanese Ministry of Public Health, andEpidemiology Surveillance Program. 2010.National Cancer Registry Report cancerdata 2007. MOPH. http://www.moph.gov.lb/Prevention/Surveillance/Pages/Cancer.aspx

Lee, K. M., Park, S. K., Kim, S. U.,Doll, M. A., Yoo, K. Y., Ahn, S. H.,Noh, D. Y., Hirvonen, A., Hein, D. W.,and Kang, D. 2003. N-Acetyltransferase(NAT1, NAT2) and glutathione S-transferase(GSTM1, GSTT1) polymorphisms in breastcancer. Cancer Lett. 196: 179–186.

Letasiova, S., Medvéova, A., Sovcikova, A.,Dusinska, M., Volkovova, K., Mosoiu, C., andBartonova, A. 2012. Bladder cancer, a reviewof the environmental risk factors. Environ.Health 11(suppl. 1): S11.

Levy, G., and Weber, W. 1992. 2-Aminofluorene–DNA adducts in mouseurinary bladder: Effect of age, sex andacetylator phenotype. Carcinogenesis 13:159–164.

Lin, H. J., Han, C. Y., Lin, B. K., andHardy, S. 1993. Slow acetylator mutations inthe human polymorphic N-acetyltransferasegene in 786 Asians, blacks, Hispanics, andwhites: Application to metabolic epidemiol-ogy. Am. J. Hum. Genet. 52: 827–834.

Lin, H. J., Han, C. Y., Lin, B. K., and Hardy,S. 1994. Ethnic distribution of slowacetylator mutations in the polymorphicN-acetyltransferase (NAT2) gene.Pharmacogenetics 4: 125–134.

Lin, Y.-S., Caffrey, J. L., Lin, J.-W., Bayliss,D., Faramawi, M. F., Bateson, T. F., andSonawane, B. 12013. Increased risk of can-cer mortality associated with cadmium expo-sures in older Americans with low zincintake. J. Toxicol. Environ. Health A 76: 1–15.

Liu, D., Wang, F., Wang, Q., Guo, X., Xu,H., Wang, W., and Zhang, L. 2012a.Association of glutathione S-transferaseM1 polymorphisms and lung cancer risk in aChinese population. Clin. Chim. Acta 414:188–190.

Liu, J., Ding, D., Wang, X., Chen, Y., Li,R., Zhang, Y., and Luo, R. 2012b. N-Acetyltransferase polymorphism and risk ofcolorectal adenoma and cancer: A pooledanalysis of variations from 59 studies. PLoSOne 7: e42797.

Liu, L., Yang, L., Zhang, Y. C., Ai, X. F., Wang,J. X., and Xiao, Z. J. 2009. [Polymorphisms ofdrug-metabolizing enzymes genes in a Han

Dow

nloa

ded

by [

Uni

vers

ity o

f W

inds

or]

at 1

8:48

14

Nov

embe

r 20

14

TOXICOGENETICS & CANCER IN LEBANESE 119

Chinese population]. Zhonghua Yi Xue Za Zhi89: 2675–2681.

Liu, S., Park, J. Y., Schantz, S. P., Stern, J. C., andLazarus, P. 2001. Elucidation of CYP2E1 5′regulatory RsaI/Pstl allelic variants and theirrole in risk for oral cancer. Oral Oncol. 37:437–445.

Lo, H. W., and Ali-Osman, F. 2007. Geneticpolymorphism and function of glutathione S-transferases in tumor drug resistance. Curr.Opin. Pharmacol. 7: 367–374.

Lower, G. J., Nilsson, T., Nelson, C., Wolf,H., Gamsky, T., and Bryan, G. 1979. N-Acetyltransferase phenotype and risk in uri-nary bladder cancer: Approaches in molec-ular epidemiology. Preliminary results inSweden and Denmark. Environ. HealthPerspect. 29: 71–79.

Lucas, D., Menez, C., Girre, C., Berthou,F., Bodenez, P., Joannet, I., Hispard, E.,Bardou, L. G., and Menez, J. F. 1995.Cytochrome P450 2E1 genotype and chlor-zoxazone metabolism in healthy and alco-holic Caucasian subjects. Pharmacogenetics5: 298–304.

Mannervik, B., Awasthi, Y. C., Board, P.G., Hayes, J. D., Di Ilio, C., Ketterer,B., Listowsky, I., Morgenstern, R.,Muramatsu, M., Pearson, W. R., et al.1992. Nomenclature for human glutathionetransferases. Biochem. J. 282: 305–306.

Martinez, C., Martin, F., Fernandez, J. M.,Garcia-Martin, E., Sastre, J., Diaz-Rubio, M.,Agundez, J. A., and Ladero, J. M. 2006.Glutathione S-transferases mu 1, theta 1, pi1, alpha 1 and mu 3 genetic polymorphismsand the risk of colorectal and gastric cancersin humans. Pharmacogenomics 7: 711–718.

Massoud, M. A. 2012. Assessment of waterquality along a recreational section of theDamour River in Lebanon using the waterquality index. Environ. Monit. Assess. 184:4151–4160.

Massoud, M. A., Al-Abady, A., Jurdi, M., andNuwayhid, I. 2010. The challenges of sus-tainable access to safe drinking water inrural areas of developing countries: Case ofZawtar El-Charkieh, Southern Lebanon. J.Environ. Health 72:24–30.

Maugard, C. M., Charrier, J., Pitard, A.,Campion, L., Akande, O., Pleasants, L.,and Ali-Osman, F. 2001. Genetic polymor-phism at the glutathione S-transferase (GST)P1 locus is a breast cancer risk modifier. Int.J. Cancer 91: 334–339.

McCarty, C. A., Reding, D. J., Commins,J., Williams, C., Yeager, M., Burmester, J.K., Schairer, C., and Ziegler, R. G. 2012.Alcohol, genetics and risk of breast cancerin the prostate, lung, colorectal and ovarian(PLCO) cancer screening trial. Breast CancerRes. Treat. 133: 785–792.

Millikan, R. C. 2000. NAT1∗10 andNAT1∗11 polymorphisms and breast cancerrisk. Cancer Epidemiol. Biomarkers Prev. 9:217–219.

Millikan, R. C., Pittman, G. S., Newman,B., Tse, C. K., Selmin, O., Rockhill,B., Savitz, D., Moorman, P. G., andBell, D. A. 1998. Cigarette smoking, N-acetyltransferases 1 and 2, and breast cancerrisk. Cancer Epidemiol. Biomarkers Prev. 7:371–378.

Millner, L. M., Doll, M. A., Cai, J., States,J. C., and Hein, D. W. 2012a. Phenotypeof the most common “slow acetylator”arylamine N-acetyltransferase 1 genetic vari-ant (NAT1∗14B) is substrate-dependent.Drug Metab. Dispos. 40: 198–204.

Millner, L. M., Doll, M. A., Stepp, M. W., States,J. C., and Hein, D. W. 2012b. Functionalanalysis of arylamine N-acetyltransferase 1(NAT1) NAT1∗10 haplotypes in a completeNATb mRNA construct. Carcinogenesis 33:348–355.

Mo, Z., Gao, Y., Cao, Y., Gao, F., and Jian,L. 2009. An updating meta-analysis of theGSTM1, GSTT1, and GSTP1 polymorphismsand prostate cancer: A HuGE review.Prostate 69: 662–688.

Mohner, M., Kersten, N., and Gellissen, J. 2013.Diesel motor exhaust and lung cancer mor-tality: Reanalysis of a cohort study in potashminers. Eur. J. Epidemiol. 28: 159–168.

Moore, L. E., Baris, D. R., Figueroa, J. D.,Garcia-Closas, M., Karagas, M. R., Schwenn,M. R., Johnson, A. T., Lubin, J. H., Hein,D. W., Dagnall, C. L., Colt, J. S., Kida, M.,

Dow

nloa

ded

by [

Uni

vers

ity o

f W

inds

or]

at 1

8:48

14

Nov

embe

r 20

14

120 H. R. DHAINI AND L. KOBEISSI

Jones, M. A., Schned, A. R., Cherala, S.S., Chanock, S. J., Cantor, K. P., Silverman,D. T., and Rothman, N. 2011. GSTM1 nulland NAT2 slow acetylation genotypes, smok-ing intensity and bladder cancer risk: Resultsfrom the New England bladder cancer studyand NAT2 meta-analysis. Carcinogenesis 32:182–189.

Morinobu, A., Kanagawa, S., Koshiba, M.,Sugai, S., and Kumagai, S. 1999. Associationof the glutathione S-transferase M1 homozy-gous null genotype with susceptibility toSjogren’s syndrome in Japanese individuals.Arthritis Rheum. 42: 2612–2615.

Mossman, B. T., Lippmann, M., Hesterberg,T. W., Kelsey, K. T., Barchowsky, A., andBonner, J. C. 2011. Pulmonary endpoints(lung carcinomas and asbestosis) followinginhalation exposure to asbestos. J. Toxicol.Environ. Health B 14: 76–121.

Mostafa, M. H., Sheweita, S. A., andO’Connor, P. J. 1999. Relationship betweenschistosomiasis and bladder cancer. Clin.Microbiol. Rev. 12: 97–111.

Moy, K. A., Yuan, J. M., Chung, F. L.,Wang, X. L., Van Den Berg, D., Wang,R., Gao, Y. T., and Yu, M. C. 2009.Isothiocyanates, glutathione S-transferaseM1 and T1 polymorphisms and gastriccancer risk: A prospective study of menin Shanghai, China. Int. J. Cancer 125:2652–2659.

Murata, M., Watanabe, M., Yamanaka, M.,Kubota, Y., Ito, H., Nagao, M., Katoh, T.,Kamataki, T., Kawamura, J., Yatani, R., andShiraishi, T. 2001. Genetic polymorphismsin cytochrome P450 (CYP) 1A1, CYP1A2,CYP2E1, glutathione S-transferase (GST)M1 and GSTT1 and susceptibility to prostatecancer in the Japanese population. CancerLett. 165: 171–177.

Nakkash, R. T., Khalil, J., and Afifi, R. A. 2011.The rise in narghile (shisha, hookah) water-pipe tobacco smoking: A qualitative studyof perceptions of smokers and non smokers.BMC Public Health 11: 315.

Nakkash, R. T., Khalil, J., Chaaya, M., andAfifi, R. A. 2010. Building research evi-dence for policy advocacy: A qualitative

evaluation of existing smoke-free policies inLebanon. Asian Pac. J. Public Health 22:168S–174S.

National Cancer Institute, National Institute ofEnvironmental Health Sciences, Agency forToxic Substances & Disease Registry, andU.S. Department of Health and HumanServices. 2003. Cancer and the environ-ment. U.S. Department of Health andHuman Services. http://www.atsdr.cdc.gov/risk/cancer/cancer-substances.html

National Toxicology Program. 2011. 2,3,7,8-Tetrachlorodibenzo-p-dioxin. Rep. Carcino-gens 12: 396–398.

Neafsy, P., Ginsberg, G., Hattis, D., Johns, D.O., Guyton, K. Z., and Sonawane, B. 2009.Genetic polymorphism in cytochrome P4502D6 (CYP2D6): Population distribution ofCYP2D6 activity. J. Toxicol. Environ. Health B12: 362–388.

Nyberg, F., Hou, S. M., Hemminki, K., Lambert,B., and Pershagen, G. 1998. GlutathioneS-transferase mu1 and N-acetyltransferase2 genetic polymorphisms and exposure totobacco smoke in nonsmoking and smokinglung cancer patients and population con-trols. Cancer Epidemiol. Biomarkers Prev. 7:875–883.

Okkels, H., Sigsgaard, T., Wolf, H., and Autrup,H. 1997. Arylamine N-acetyltransferase 1(NAT1) and 2 (NAT2) polymorphisms in sus-ceptibility to bladder cancer: The influenceof smoking. Cancer Epidemiol. BiomarkersPrev. 6: 225–231.

Ovsiannikov, D., Selinski, S., Lehmann, M. L.,Blaszkewicz, M., Moormann, O., Haenel, M.W., Hengstler, J. G., and Golka, K. 2012.Polymorphic enzymes, urinary bladder can-cer risk, and structural change in the localindustry. J. Toxicol. Environ. Health A 75:557–565.

Ozbek, Y. K., Ozturk, T., Tuzuner, B. M., Calay,Z., Ilvan, S., Seyhan, F. M., Kisakesen, H. I.,Ozturk, O., and Isbir, T. 2010. Combinedeffect of CYP1B1 codon 432 polymorphismand N-acetyltransferase 2 slow acetylatorphenotypes in relation to breast cancer inthe Turkish population. Anticancer Res. 30:2885–2889.

Dow

nloa

ded

by [

Uni

vers

ity o

f W

inds

or]

at 1

8:48

14

Nov

embe

r 20

14

TOXICOGENETICS & CANCER IN LEBANESE 121

Pan, Z. J., Huang, W. J., Zou, Z. H., and Gao,X. C. 2012. The GSTT1 null genotype con-tributes to increased risk of prostate cancer inAsians: A meta-analysis. Asian Pac. J. CancerPrev. 13: 2635–2638.

Parke, D. V. 1994. The cytochromes P450 andmechanisms of chemical carcinogenesis.Environ. Health Perspect. 102: 852–853.

Peng, H., Xie, S. K., Huang, M. J., andRen, D. L. 2013. Associations of CYP2E1rs2031920 and rs3813867 polymorphismswith colorectal cancer risk: A systemicreview and meta-analysis. Tumour Biol. 34:2389–2395.

Perera, F. P. 1998. Molecular epidemiol-ogy of environmental carcinogenesis. RecentResults Cancer Res. 154: 39–46.

Perez-Morales, R., Mendez-Ramirez, I., Castro-Hernandez, C., Martinez-Ramirez, O. C.,Gonsebatt, M. E., and Rubio, J. 2011.Polymorphisms associated with the risk oflung cancer in a healthy Mexican Mestizopopulation: Application of the additivemodel for cancer. Genet. Mol. Biol. 34:546–552.

Philip, P. A., Fitzgerald, D. L., Cartwright, R.A., Peake, M. D., and Rogers, H. J. 1988.Polymorphic N-acetylation capacity in lungcancer. Carcinogenesis 9: 491–493.

Polychronakis, I., Dounias, G., Makropoulos, V.,Riza, E., and Linos, A. 2013. Work-relatedleukemia: A systematic review. J. Occup.Med. Toxicol. 8: 14.

Probst-Hensch, N., Haile, R., Li, D., Sakamoto,G., Louie, A., Lin, B., Frankl, H., Lee,E., and Lin, H. 1996. Lack of associa-tion between the polyadenylation polymor-phism in the NAT1 (acetyltransferase 1) geneand colorectal adenomas. Carcinogenesis 17:2125–2129.

Purewal, M., Fretland, A. J., Schut, H. A.,Hein, D. W., and Wargovich, M. J.2000. Association between acetylatorgenotype and 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) DNAadduct formation in colon and prostate ofinbred Fischer 344 and Wistar Kyoto rats.Cancer Lett. 149: 53–60.

Qadri, Q., Sameer, A. S., Shah, Z. A.,Hamid, A., Alam, S., Manzoor, S., and

Siddiqi, M. A. 2011. Genetic polymorphismof the glutathione-S-transferase P1 gene(GSTP1) and susceptibility to prostate cancerin the Kashmiri population. Genet. Mol. Res.10: 3038–3045.

Ramalhinho, A. C., Fonseca-Moutinho, J. A.,and Breitenfeld, L. 2011. Glutathione S-transferase M1, T1, and P1 genotypesand breast cancer risk: a study in aPortuguese population. Mol. Cell. Biochem.355:265–271.

Rodriguez-Antona, C., Gomez, A., Karlgren,M., Sim, S. C., and Ingelman-Sundberg, M.2010. Molecular genetics and epigenetics ofthe cytochrome P450 gene family and its rel-evance for cancer risk and treatment. Hum.Genet. 127: 1–17.

Roemer, H. C., Weistenhofer, W., Lohlein, D.,Geller, F., Blomeke, B., and Golka, K. 2008.N-Acetyltransferase 1 in colon and rectalcancer cases from an industrialized area. J.Toxicol. Environ. Health A 71: 902–905.

Rojas, M., Alexandrov, K., Cascorbi, I.,Brockmoller, J., Likhachev, A., Pozharisski, K.,Bouvier, G., Auburtin, G., Mayer, L., Kopp-Schneider, A., Roots, I., and Bartsch, H. 1998.High benzo[a]pyrene diol-epoxide DNAadduct levels in lung and blood cells from indi-viduals with combined CYP1A1 MspI/Msp-GSTM1∗0/∗0 genotypes. Pharmacogenetics8: 109–118.

Roots, I., Drakoulis, N., Ploch, M., Heinemeyer,G., Loddenkemper, R., Minks, T., Nitz, M.,Otte, F., and Koch, M. 1988. Debrisoquinehydroxylation phenotype, acetylationphenotype, and ABO blood groups asgenetic host factors of lung cancer risk. Klin.Wochenschr. 66(suppl. 11): 87–97.

Rosenman, K. D., and Reilly, M. J. 2004. Cancermortality and incidence among a cohort ofbenzidine and dichlorobenzidine dye man-ufacturing workers. Am. J. Ind. Med. 46:505–512.

Rossini, A., de Almeida Simao, T.,Albano, R. M., and Pinto, L. F. 2008.CYP2A6 polymorphisms and risk fortobacco-related cancers. Pharmacogenomics9: 1737–1752.

Rundle, A., Tang, D., Hibshoosh, H., Estabrook,A., Schnabel, F., Cao, W., Grumet, S., and

Dow

nloa

ded

by [

Uni

vers

ity o

f W

inds

or]

at 1

8:48

14

Nov

embe

r 20

14

122 H. R. DHAINI AND L. KOBEISSI

Perera, F. P. 2000. The relationship betweengenetic damage from polycyclic aromatichydrocarbons in breast tissue and breast can-cer. Carcinogenesis 21: 1281–1289.

Saade, G., Seidenberg, A. B., Rees, V. W.,Otrock, Z., and Connolly, G. N. 2010.Indoor secondhand tobacco smoke emissionlevels in six Lebanese cities. Tobacco Control19: 138–142.

Sabbagh, A., Darlu, P., Crouau-Roy, B.,and Poloni, E. S. 2011. Arylamine N-acetyltransferase 2 (NAT2) genetic diversityand traditional subsistence: A worldwidepopulation survey. PLoS One 6: e18507.

Saeed, H. M., Alanazi, M. S., Nounou, H.A., Salaby, M. A., Semlali, A., Azzam, N.,Aljebreen,A.,Alharby,O.,Parine,N.R.,Shaik,J., and Arafaha, M. 2013. Cytochrome P4501A1, 2E1 and GSTM1 gene polymorphismsand susceptibility to colorectal cancer in theSaudi population. Asian Pac. J. Cancer Prev.14: 3761–3768.

Sakoda, L. C., Blackston, C. R., Xue, K.,Doherty, J. A., Ray, R. M., Lin, M. G.,Stalsberg, H., Gao, D. L., Feng, Z., Thomas,D. B., and Chen, C. 2008. Glutathione S-transferase M1 and P1 polymorphisms andrisk of breast cancer and fibrocystic breastconditions in Chinese women. Breast CancerRes. Treat. 109: 143–155.

Salameh, P., Salame, J., Khayat, G., Akhdar, A.,Ziadeh, C., Azizi, S., Khoury, F., Akiki, Z.,Nasser, Z., Abou Abbass, L., Saadeh, D., andWaked, M. 2012. Exposure to outdoor airpollution and chronic bronchitis in adults:A case-control study. Int. J. Occup. Environ.Med. 3: 165–177.

Salem, A. H., Yaqoob, A., Ali, M., Handu,S., Fadel, R., Abu-Hijleh, M., and Almawi,W. 2011. Genetic polymorphism of theglutathione S-transferase M1 and T1 genes inthree distinct Arab populations. Dis. Markers31: 311–316.

Sanderson, S., Salanti, G., and Higgins, J. 2007.Joint effects of the N-acetyltransferase 1 and2 (NAT1 and NAT2) genes and smoking onbladder carcinogenesis: A literature-basedsystematic HuGE review and evidence syn-thesis. Am. J. Epidemiol. 166: 741–751.

Sangrajrang, S., Sato, Y., Sakamoto, H.,Ohnami, S., Khuhaprema, T., and Yoshida, T.2010. Genetic polymorphisms in folate andalcohol metabolism and breast cancer risk:A case-control study in Thai women. BreastCancer Res. Treat. 123: 885–893.

Sato, M., Sato, T., Izumo, T., and Amagasa,T. 1999. Genetic polymorphism of drug-metabolizing enzymes and susceptibility tooral cancer. Carcinogenesis 20: 1927–1931.

Seow, A., Zhao, B., Poh, W. T., Teh, M., Eng, P.,Wang, Y. T., Tan, W. C., Lee, E. J., and Lee,H. P. 1999. NAT2 slow acetylator genotypeis associated with increased risk of lung can-cer among non-smoking Chinese women inSingapore. Carcinogenesis 20: 1877–1881.

Sepetdjian, E., Shihadeh, A., and Saliba, N. A.2008. Measurement of 16 polycyclic aro-matic hydrocarbons in narghile waterpipetobacco smoke. Food Chem. Toxicol. 46:1582–1590.

Sergentanis, T. N., and Economopoulos, K. P.2010. GSTT1 and GSTP1 polymorphismsand breast cancer risk: A meta-analysis.Breast Cancer Res. Treat. 121: 195–202.

Shahriary, G. M., Galehdari, H., Jalali, A.,Zanganeh, F., Alavi, S. M., and Aghanoori,M. R. 2012. CYP2E1∗5B, CYP2E1∗6,CYP2E1∗7B, CYP2E1∗2, and CYP2E1∗3 allelefrequencies in Iranian populations. AsianPac. J. Cancer Prev. 13:6505–6510.

Shamseddine, A., and Musallam, K. 2010.Cancer epidemiology in Lebanon. MiddleEast J. Cancer 1: 41–44.

Shamseddine, A., Sibai, A., Gehchan, N., Rahal,B., El-Saghir, N., Ghosn, M., Aftimos, G.,Chamsuddine, N., and Seoud, M. 2004.Cancer incidence in postwar Lebanon:Findings from the first national population-based registry, 1998. Ann. Epidemiol. 14:663–668.

Sharma, S., Cao, X., Wilkens, L. R., Yamamoto,J., Lum-Jones, A., Henderson, B. E., Kolonel,L. N., and Le Marchand, L. 2010. Well-done meat consumption, NAT1 andNAT2 acetylator genotypes and prostatecancer risk: The multiethnic cohort study.Cancer Epidemiol. Biomarkers Prev. 19:1866–1870.

Dow

nloa

ded

by [

Uni

vers

ity o

f W

inds

or]

at 1

8:48

14

Nov

embe

r 20

14

TOXICOGENETICS & CANCER IN LEBANESE 123

Shi, X., Chiu, A., Chen, C.T., Halliwell, B.,Castranova, V. and Vallyathan, V. 1999.Reduction of chromium VI and its relation-ship to carcinogenesis. J. Toxicol. Environ.Health B 1: 87–104.

Shi, Z., Dragin, N., Galvez-Peralta, M., Jorge-Nebert, L. F., Miller, M. L., Wang, B., andNebert, D. W. 2010. Organ-specific rolesof CYP1A1 during detoxication of dietarybenzo[a]pyrene. Mol. Pharmacol. 78: 46–57.

Shimada, T., and Guengerich, F. 1991.Oxidation of toxic and carcinogenic chemi-cals by human cytochrome P-450 enzymes.Chem. Res. Toxicol. 4: 391–407.

Simmons, P. T., and Portier, C. J. 2002.Toxicogenomics: The new frontier in riskanalysis. Carcinogenesis 23: 903–905.

Snyder, R. 2000.Overview of the toxicology ofbenzene. J. Toxicol. Environ. Health A 61:339–346.

Spurdle, A. B., Fahey, P., Chen, X., McGuffog,L., Easton, D., Peock, S., Cook, M., Simard,J., Rebbeck, T. R., Antoniou, A. C., andChenevix-Trench, G. 2010. Pooled anal-ysis indicates that the GSTT1 deletion,GSTM1 deletion, and GSTP1 Ile105Valpolymorphisms do not modify breast cancerrisk in BRCA1 and BRCA2 mutation carriers.Breast Cancer Res. Treat. 122: 281–285.

Stanley, L., Coroneos, E., Cuff, R., Hickman,D., Ward, A., and Sim, E. 1996.Immunochemical detection of arylamineN-acetyltransferase in normal and neoplas-tic bladder. J. Histochem. Cytochem. 44:1059–1067.

Stiborova, M., Levova, K., Barta, F., Shi, Z., Frei,E., Schmeiser, H. H., Nebert, D. W., Phillips,D. H., and Arlt, V. M. 2012. Bioactivationversus detoxication of the urothelial carcino-gen aristolochic acid I by human cytochromeP450 1A1 and 1A2. Toxicol. Sci. 125:345–358.

Strange, R. C., and Fryer, A. A. 1999. Theglutathione S-transferases: Influence of poly-morphism on cancer susceptibility. IARC Sci.Publ. 148: 231–249.

Su, X. L., Bin, B., Cui, H. W., and Ran, M. R.2011. Cytochrome P450 2E1 RsaI/PstI andDraI polymorphisms are risk factors for lung

cancer in Mongolian and Han populationin Inner Mongolia. Chin. J. Cancer Res. 23:107–111.

Tanaka, E., Terada, M., and Misawa, S. 2000.Cytochrome P450 2E1: Its clinical and tox-icological role. J. Clin. Pharm. Ther. 25:165–175.

Taylor, J., Umbach, D., Stephens, E., Castranio,T., Paulson, D., Robertson, C., et al. 1998.The role of N-acetylation polymorphismsin smoking-associated bladder cancer:Evidence of a gene–gene–exposurethree-way interaction. Cancer Res. 58:3603–3610.

Thakur, H., Gupta, L., Sobti, R. C., Janmeja,A. K., Seth, A., and Singh, S. K. 2011.Association of GSTM1T1 genes with COPDand prostate cancer in north Indian popula-tion. Mol. Biol. Rep. 38: 1733–1739.

To-Figueras, J., Gene, M., Gomez-Catalan, J.,Galan, M. C., Fuentes, M., Ramon, J. M.,Rodamilans, M., Huguet, E., and Corbella,J. 1997. Glutathione S-transferase M1(GSTM1) and T1 (GSTT1) polymorphismsand lung cancer risk among NorthwesternMediterraneans. Carcinogenesis 18:1529–1533.

Tsai, S.-M., Wang, T.-N., and Ko, Y.-C. 1998.Cancer mortality trends in a blackfoot diseaseendemic community of Taiwan followingwater source replacement. J. Toxicol. Environ.Health A 55: 389–404.

U.S. Environmental Protection Agency. 2009.National primary drinking water regulations.http://www.epa.gov/safewater/consumer/pdf/mcl.pdf

Vatsis, K., and Weber, W. 1993. Structural het-erogeneity of Caucasians N-acetlytranferanseat the NAT1 gene locus. Arch. Biochem.Biophys. 301: 71–76.

Vineis, P. 1992. Epidemiological models ofcarcinogenesis: The example of bladder can-cer. Cancer Epidemiol. Biomarkers Prev. 1:149–153.

Vlaanderen, J., Straif, K., Pukkala, E.,Kauppinen, T., Kyyronen, P., Martinsen,J. I., Kjaerheim, K., Tryggvadottir, L., Hansen,J., Sparen, P., and Weiderpass, E. 2013.Occupational exposure to trichloroethylene

Dow

nloa

ded

by [

Uni

vers

ity o

f W

inds

or]

at 1

8:48

14

Nov

embe

r 20

14

124 H. R. DHAINI AND L. KOBEISSI

and perchloroethylene and the risk oflymphoma, liver, and kidney cancer in fourNordic countries. Occup. Environ. Med. 70:393–401.

Wadelius, M., Autrup, J. L., Stubbins, M. J.,Andersson, S. O., Johansson, J. E., Wadelius,C., Wolf, C. R., Autrup, H., and Rane, A.1999. Polymorphisms in NAT2, CYP2D6,CYP2C19 and GSTP1 and their associationwith prostate cancer. Pharmacogenetics 9:333–340.

Waked, M., Khayat, G., and Salameh, P. 2012.Cigarette smokers’ profile in Lebanese adults.J. Res. Health Sci. 12: 75–80.

Wakefield, L., Robinson, J., Long, H., Ibbitt, J.C., Cooke, S., Hurst, H. C., and Sim, E. 2008.Arylamine N-acetyltransferase 1 expressionin breast cancer cell lines: A potential markerin estrogen receptor-positive tumors. GenesChromosomes Cancer 47: 118–126.

Walker, K., Ginsberg, G., Hattis, D., Johns, D.,Guyton, K., and Sonawane, B. 2009. Geneticpolymorphism in N-acetyltransferase (NAT):Population distribution of NAT1 andNAT2 activity. J. Toxicol. Environ. Health B12: 440–472.

Wang, Y., Spitz, M. R., Schabath, M. B.,Ali-Osman, F., Mata, H., and Wu, X.2003. Association between glutathione S-transferase p1 polymorphisms and lung can-cer risk in Caucasians: a case-control study.Lung Cancer 40: 25–32.

Wang, Y., Yang, H., Li, L., Wang, H., Zhang,C., Yin, G., and Zhu, B. 2010. Associationbetween CYP2E1 genetic polymorphismsand lung cancer risk: A meta-analysis. Eur. J.Cancer 46: 758–764.

Weber, W., and Hein, D. 1985. N-Acetylationpharmacogenetics. Pharmacol. Rev. 37:25–79.

Weistenhofer, W., Blaszkewicz, M., Bolt, H.M., and Golka, K. 2008. N-Acetyltransferase-2 and medical history in bladder cancer caseswith a suspected occupational disease (BK1301) in Germany. J. Toxicol. Environ. HealthA 71: 906–910.

Wikman, H., Thiel, S., Jager, B., Schmezer,P., Spiegelhalder, B., Edler, L., Dienemann,H., Kayser, K., Schulz, V., Drings, P.,

Bartsch, H., and Risch, A. 2001. Relevanceof N-acetyltransferase 1 and 2 (NAT1,NAT2) genetic polymorphisms in non-small cell lung cancer susceptibility.Pharmacogenetics 11: 157–168.

Williams, J. H., Phillips, T. D., Jolly, P. E., Stiles,J. K., Jolly, C. M., and Aggarwal, D. 2004.Human aflatoxicosis in developing countries:A review of toxicology, exposure, potentialhealth consequences, and interventions. Am.J. Clin. Nutr. 80: 1106–1122.

Windmill, K., Gaedigk, A., Hall, P.,Samaratunga, H., Grant, D., and McManus,M. 2000. Localization of N-acetyltransferasesNAT1 and NAT2 in human tissues. Toxicol.Sci. 54: 19–29.

Woolhouse, N. M., Qureshi, M. M., Bastaki,S. M., Patel, M., Abdulrazzaq, Y., andBayoumi, R. A. 1997. Polymorphic N-acetyltransferase (NAT2) genotyping ofEmiratis. Pharmacogenetics 7: 73–82.

Wrighton, S. A., VandenBranden, M., and Ring,B. J. 1996. The human drug metaboliz-ing cytochromes P450. J. Pharmacokinet.Biopharm. 24: 461–473.

Wu, K., Wang, X., Xie, Z., Liu, Z., and Lu,Y. 2013a. Glutathione S-transferase P1 genepolymorphism and bladder cancer suscepti-bility: An updated analysis. Mol. Biol. Rep.40: 687–695.

Wu, K., Wang, X., Xie, Z., Liu, Z., and Lu, Y.2013b. N-acetyltransferase 1 polymorphismand bladder cancer susceptibility: A meta-analysis of epidemiological studies. J. Int.Med. Res. 41: 31–37.

Wu, S. H., Tsai, S. M., Hou, M. F., Lin, H. S.,Hou, L. A., Ma, H., Lin, J. T., Yeh, F. L.,and Tsai, L. Y. 2006. Interaction of geneticpolymorphisms in cytochrome P450 2E1 andglutathione S-transferase M1 to breast can-cer in Taiwanese woman without smokingand drinking habits. Breast Cancer Res. Treat.100: 93–98.

Xie, H. G., Xu, Z. H., Ou-Yang, D. S., Shu, Y.,Yang, D. L., Wang, J. S., Yan, X. D., Huang,S. L., Wang, W., and Zhou, H. H. 1997.Meta-analysis of phenotype and genotypeof NAT2 deficiency in Chinese populations.Pharmacogenetics 7: 503–514.

Dow

nloa

ded

by [

Uni

vers

ity o

f W

inds

or]

at 1

8:48

14

Nov

embe

r 20

14

TOXICOGENETICS & CANCER IN LEBANESE 125

Yang, J., Gu, M., Song, N. H., Feng, N. H., Hua,L. X., Ju, X. B., Sui, Y. G., Wang, X. R., andWu, H. F. 2009. [Correlation of prostate can-cer susceptibility with genetic polymorphismof cytochrome P450 2E1, smoking and drink-ing: A case-control study in the populationof Nanjing area]. Zhonghua Nan Ke Xue 15:7–11.

Yang, J., Qian, L. X., Wu, H. F., Xu, Z. Q., Sui,Y. G., Wang, X. R., and Zhang, W. 2006.Genetic polymorphisms in the cytochromeP450 1A1 and 2E1 genes, smoking, drinkingand prostate cancer susceptibility: A case-control study in a Han nationality populationin Southern China. Int. J. Urol. 13: 773–780.

Yang, Q., Du, J., and Yao, X. 2013. Significantassociation of glutathione S-transferaseT1 null genotype with prostate cancer risk: Ameta-analysis of 26,393 subjects. PLoS One8: e53700.

Yassine, I. A., Kobeissi, L., Jabbour, M. E., andDhaini, H. R. 2012. N-Acetyltransferase 1(NAT1) genotype: A risk factor for urinarybladder cancer in a Lebanese population. J.Oncol. 2012: 512976.

Yu, M. C., Skipper, P. L., Taghizadeh, K.,Tannenbaum, S. R., Chan, K. K., Henderson,B. E., and Ross, R. K. 1994. Acetylatorphenotype, aminobiphenyl–hemoglobinadduct levels, and bladder cancer risk inwhite, black, and Asian men in Los Angeles,California. J. Natl. Cancer Inst. 86: 712–716.

Zgheib, N. K., Mitri, Z., Geryess, E., andNoutsi, P. 2010. Cytochrome P4502E1(CYP2E1) genetic polymorphisms in aLebanese population: Frequency distributionand association with morbid diseases. Genet.Test Mol. Biomarkers 14: 393–397.

Zgheib, N. K., Shamseddine, A. A., Geryess,E., Tfayli, A., Bazarbachi, A., Salem, Z.,Shamseddine, A., Taher, A., and El-Saghir,N. 2013. Genetic polymorphisms of CYP2E1,GST, and NAT2 enzymes are not asso-ciated with risk of breast cancer in asample of Lebanese women. Mutat. Res.747–748:40–47.

Zhan, P., Wang, J., Zhang, Y., Qiu, L. X., Zhao,S. F., Qian, Q., Wei, S. Z., Yu, L. K., and Song,

Y. 2010. CYP2E1 Rsa I/Pst I polymorphismis associated with lung cancer risk amongAsians. Lung Cancer 69: 19–25.

Zhang, Y., Shi, Z., Deng, C., Zhu, Y., andZhou, Y. 2002. [The research the relation-ship between N-acetyltransferase 1 genepolymorphisms and genetic susceptibilityto sporadic colorectal adenocarcinoma].Zhonghua Nei Ke Za Zhi 41: 746–749.

Zhao, B., Lee, E. J., Yeoh, P. N., and Gong,N. H. 1998. Detection of mutations andpolymorphism of N-acetyltransferase 1 genein Indian, Malay and Chinese populations.Pharmacogenetics 8: 299–304.

Zhao, B., Seow, A., Lee, E. J., and Lee, H.P. 2000. Correlation between acetylationphenotype and genotype in Chinese women.Eur. J. Clin. Pharmacol. 56: 689–692.

Zheng, J., Chen, S., Jiang, L., You, Y., Wu,D., and Zhou, Y. 2011. Functional geneticvariations of cyclooxygenase-2 and suscep-tibility to acute myeloid leukemia in aChinese population. Eur. J. Haematol. 87:486–493.

Zheng, W., Deitz, A. C., Campbell, D. R.,Wen, W. Q., Cerhan, J. R., Sellers, T. A.,Folsom, A. R., and Hein, D. W. 1999. N-Acetyltransferase 1 genetic polymorphism,cigarette smoking, well-done meat intake,and breast cancer risk. Cancer EpidemiolBiomarkers Prev 8: 233–239.

Zheng, Y. L., Amr, S., Saleh, D. A., Dash, C.,Ezzat, S., Mikhail, N. N., Gouda, I., Loay, I.,Hifnawy, T., Abdel-Hamid, M., Khaled, H.,Wolpert, B., Abdel-Aziz, M. A., and Loffredo,C. A. 2012. Urinary bladder cancer riskfactors in Egypt: A multicenter case-controlstudy. Cancer Epidemiol. Biomarkers Prev.21: 537–546.

Zhou, S. F., Liu, J. P., and Chowbay, B.2009. Polymorphism of human cytochromeP450 enzymes and its clinical impact. DrugMetab. Rev. 41: 89–95.

Zhu, J., Chen, W., Xu, S., Wang, Y., and Sun,Z. 2013. GSTT1 null genotype is associatedwith an increased risk of prostate cancer inCaucasians: A meta-analysis. Urol. Int. 91:113–120.

Dow

nloa

ded

by [

Uni

vers

ity o

f W

inds

or]

at 1

8:48

14

Nov

embe

r 20

14