Recent Advances in Gastroenterology 13 Chris Probert
INDEX
×
Chapter Notes

Save Clear


Genetic epidemiology of oesophageal cancerChapter 1

Anthony Ellis,
Janet M Risk
 
INTRODUCTION
The majority of oesophageal cancers are carcinomas–either squamous cell (SCC) or adenocarcinoma (AC). Rarer types include small cell cancer and nonepithelial tumours such as sarcomas (e.g. leiomyosarcoma and rhabdomyosarcoma), malignant melanoma and lymphoma. Oesophageal cancer is the 10th commonest cancer in the world overall, but it is the 5th commonest cancer in less developed countries, most of these being SCCs. It has the distinction of having the widest variation in incidence worldwide of any cancer, there being a 15-fold geographical variation in men and a 20-fold variation in women. The highest rates are found in China, Iran and South Africa and lowest rates in central Africa and central America. In these high-risk countries, the majority of oesophageal cancer is due to SCC, but in low-risk countries, particularly certain Western countries, the incidence of AC is rising rapidly and it is the most rapidly increasing of all cancers.
Worldwide, 482,000 people are diagnosed with oesophageal cancer and 407,000 people die from it each year [1]. In the United States, the comparable figures are an estimated 17,990 cases and 15,210 deaths in 2013 [2], whilst in the European Union, for the year 2010, the estimated number of cases was 43,700 and 27,700 deaths. Within Europe, the United Kingdom has the highest rate of oesophageal cancer, with approximately twice the average European age-adjusted rates.
 
RISK FACTORS (TABLE 1.1)
 
Age and gender
Both SCC and AC are associated with increasing age, and are diseases of middle to late life. SCC has roughly an equal gender incidence, particularly in areas of high incidence; however, in areas of low incidence, men predominate just as with AC.
2
Table 1.1   Risk factors for oesophageal cancer
Oesophageal cancer
Factor
Squamous cancer
Adenocarcinoma
Age
> 50
> 50
Gender
M = F
M > F
Race
A-C > Cauc > Hisp
Caucasian
Socioeconomic (lower) status
?
Smoking
Alcohol
x
Increased BMI
Dietary—Reduced fruit and vegetables
?
  Deficiency of Se, Zn and folate
?
Poor oral hygiene
?
Drinking maté
x
Drinking hot liquids
x
Pre-existing diseases
Hiatus hernia/GOR
x
  Barrett's oesophagus
x
  Atrophic gastritis
x
  Achalasia
x
  Caustic injury
x
  Thoracic radiation
x
  Coeliac disease
x
  Previous head and neck cancer
x
  HPV infection
x
  Previous cholecystectomy
x
  H. pylori absence
x
Heredity—Tylosis
x
Alcohol metabolism
x
Medications—Lower LOSP
  NSAIDs/aspirin reduced risk
  Bisphosphonates
x
A-C, Afro-Caribbean; GOR, gastro-oesophageal reflux—increased frequency and duration; Hisp, Hispanic; NSAIDs, nonsteroidal anti-inflammatory drugs.
 
Smoking and alcohol
Both tobacco and alcohol have been identified as major risk factors for oesophageal SCC and are synergistic. Smoking cigarettes increases the risk three- to sevenfold [2], whilst alcohol confers a three- to fivefold risk. Individuals who smoke and drink have a ninefold increased risk. Cigar and pipe smokers have a risk similar to cigarette smokers [2].3
Smoking is less strongly associated with oesophageal AC. Population-based case–control and cohort studies have shown a less than twofold increase, but several of these studies have shown a dose-related effect, increasing the suspicion of a relationship between the two [3]. Alcohol is not a risk factor for oesophageal AC.
 
Race
In the United States, age-adjusted incidence of oesophageal cancer is almost twice as high in African American races than in Caucasians and exceeds that for Hispanics, Asian Americans and Native Americans; however, AC is mainly a disease of Caucasian males [4].
 
Socioeconomic status
SCC was found to be associated with lower socioeconomic status in one study.
 
Dietary factors
 
Carcinogens
 
N-Nitroso compounds
N-Nitroso compounds are known carcinogens, thought to act by causing the development of alkyl adducts in DNA. In animals, they have been implicated in the development of several cancers, including in the nasal cavity, oesophagus and stomach [5], although direct evidence for human cancers is lacking. Furthermore, 45–75% of exposure comes from endogenous synthesis of ingested nitrites or nitrates and the remainder from tobacco smoking, occupational exposure and food sources such as vegetables. Pickled vegetables and other foodstuffs may be a source of nitroso compounds, particularly in endemic areas such as China, either directly or secondary to contamination by toxin-producing fungi.
 
Polycyclic aromatic hydrocarbons
These compounds are contained in a drink called maté which is made by making infusions of the herb Ilex paraguayensis with hot water–a practice common in certain parts of South America, which also has high rates of oesophageal cancer [5]. It is not certain whether it is the polycyclic aromatic hydrocarbons (PAH) alone or the fact that the maté is often drunk very hot (see below) which is the important factor.
 
Nutritional deficiencies
 
Fresh fruit and vegetables
Diets low in fresh fruit and vegetables have been postulated to be conducive to the development of oesophageal cancer, most of the evidence coming from case–control studies. Conversely, diets high in fresh fruit and vegetables have been shown to lower the risk of oesophageal cancer although the vast majority of the evidence has been in relation to SCC [6]. What evidence there is does not support a reduced risk of AC with diets high in fruit and vegetables.
 
Vitamins and minerals
Low levels of selenium, zinc and folic acid have been associated with increased risk of SCC. Diets high in folic acid have been shown to reduce the risk of oesophageal cancer.4
 
Poor oral hygiene
Initial evidence supporting a link between oesophageal cancer and poor oral hygiene appear to be confirmed by later studies. However, most of the studies did not take account of other, potentially confounding, factors.
 
Repetitive thermal injury
Repetitive thermal injury to the oesophageal mucosa from the ingestion of hot food and drinks has been considered a cause of oesophageal cancer. More than half of 59 case–control and cohort studies have shown a significantly increased risk with ingestion of hot liquids although the methodology has been criticised and, in the majority of studies, oesophageal SCC and AC were not differentiated.
 
Occupation
Traditionally, oesophageal SCC was said to be increased in certain occupations related to alcohol production, such as draymen and publicans, although the evidence for this was contaminated by confounding factors which were not adjusted for in the analysis. Occupations characterised by exposure to asbestos, silica and other substances such as sulphuric acid have been associated with an increased risk of this cancer [6].
 
Oesophageal disease
 
Achalasia
Case–control studies in patients with achalasia have indicated that oesophageal SCC occurs in 3–7% of this population, compared with 2% in the general population [7].
 
Gastro-oesophageal reflux
Symptomatic gastro-oesophageal reflux is one of the strongest risk factors for oesophageal AC. The risk is increased depending on the length of the history, the frequency of symptoms and presence of nocturnal reflux [8].
 
Barrett's oesophagus
Barrett's oesophagus (BO) is a condition in which the normal squamous mucosa is replaced with columnar epithelium in the distal oesophagus as a result of chronic gastro-oesophageal reflux and is strongly associated with the development of oesophageal AC. The prevalence of BO is 1.6% in the general population and 10–15% in patients with reflux who have undergone upper gastrointestinal endoscopy. The risk of AC in patients with Barrett's oesophagus is 0.5% per year. Barrett's oesophagus is most commonly diagnosed in middle to late life, with the average age at diagnosis being 55. More men are diagnosed than women and it is most common in Caucasian populations, less common in black and Hispanic populations and least common in Asian populations. Barrett's oesophagus is associated with smoking and obesity. The risk of AC in BO is approximately 1% [9].
 
Obesity
The mechanism whereby obesity increases the risk of oesophageal AC is probably indirect, through the promotion of gastro-oesophageal reflux as a result of increased intra-5abdominal pressure and the subsequent development of BO. A meta-analysis showed an increased odds ratio (OR) of 2.78 (1.85–4.16) for AC in patients with a body mass index greater than 30 [10,11].
 
Coeliac disease
Coeliac disease is associated with an increased risk for a number of malignancies including oesophageal SCC for which the risk is increased eightfold [12].
 
Previous aerodigestive cancers
Approximately 2% of patients who have had a SCC of the upper aerodigestive tract will go on to develop a metachronous SCC of the oesophagus [13].
 
Caustic injury
This is due to the ingestion of strong alkalis such as sodium or potassium hydroxide contained in drain-cleaning fluid and other household cleaning products. Most cases occur in children under the age 5 when they are accidental and the rest in adults as a result of psychotic or suicidal behaviour. It results in scarring and stricture formation in the oesophagus. SCC develops after approximately 40 years [14].
 
Thoracic radiation
Radiotherapy to the mediastinum for the treatment of a number of cancers including breast cancer, lung cancer and lymphoma increases the risk of both types of oesophageal cancer [15].
 
Plummer—Vinson/Patterson—Kelly syndrome
Patients with this condition, which comprises a triad of features, namely dysphagia, iron deficiency anaemia and oesophageal webs, have a greater risk of SCC [16] although some cases of this syndrome may be due to undiscovered coeliac disease.
 
Helicobacter pylori infection and atrophic gastritis
A meta-analysis of 19 studies suggested an inverse relationship between the presence of CagA-positive Helicobacter pylori (H. pylori) and the risk of AC [17]. It has been suggested that the decline of H. pylori infection may be partly responsible for the increase in AC over the past decades.
 
Previous cholecystectomy
A population-based, cohort study of cholecystectomised patients in Sweden found an increased risk of AC (standardized incidence ratio, 1.3; 95% CI, 1.0–1.8) but not of SCC (SIR, 0.9; 95% CI, 0.7–1.1) [18]. The rationale behind the study was that duodenogastric reflux of bile occurred after cholecystectomy and gastro-oesophageal reflux of bile is associated with Barrett's oesophagus.
 
Human papilloma virus
Human papilloma virus (HPV) has been implicated in the development of both benign and malignant squamous cell tumours of the oesophagus; 21.3% of squamous cell papillomas and 22.9% of SCCs of the oesophagus were positive for HPV infection [19].6
 
Medications
 
Medicines that reduce the tone of the lower oesophageal sphincter
A number of medications may increase the risk of AC by causing relaxation of the lower oesophageal sphincter, thereby promoting gastro-oesophageal reflux. These include β-adrenergic agonists (bronchodilators), calcium channel blockers, nitrates, phosphodiesterase inhibitors (aminophylline), anticholinergics, γ-aminobutyric acid agonists (baclofen), benzodiazepines and hormone replacement therapy (HRT). The available evidence has been conflicting.
 
Bisphosphonates
Early studies on the possible link between bisphosphonate use and oesophageal cancer showed conflicting results. In a series of population-based case–control studies in two large primary care databases, exposure to bisphosphonates was not associated with increased risk [20], although a limitation of this study was that no attempt was made to differentiate the two main types of oesophageal cancer.
 
Medications that may protect against oesophageal cancer
 
Nonsteroidal anti-inflammatory drugs and aspirin
There is epidemiological evidence to indicate that these medications may reduce the risk of oesophageal cancer, particularly AC, by approximately 40%, although one study found that using the selective COX inhibitor, celecoxib, did not appear to have any effect [21]. The mechanism of this action is thought to be by the inhibition of the enzyme cyclooxygenase.
 
Proton pump inhibitors and H2 receptor antagonists
Unfortunately, studies looking at the effects of these medications in oesophageal cancer have not produced any consistent results. Theoretically, they could work by reducing the acid content of the gastric refluxate, the stimulant to mucosal change in Barrett's oesophagus. Alternatively, by reducing gastric acid, they may encourage gastric colonisation by bacteria that are capable of producing gastric carcinogens such as N-nitrosamines.
 
Hormone replacement therapy
It has been speculated that HRT may be implicated in a number of cancers including cancers of the oropharynx, stomach and colon. A meta-analysis of three case–control studies from Europe provided evidence for a beneficial effect of HRT in oesophageal cancer. This result is all the more surprising considering the fact that HRT has been shown to increase the risk of reflux (vide supra).
 
Heredity
 
Molecular studies
Oesophageal SCC, like most solid tumours, has been shown to harbour many somatic mutations, which may be driver or bystander events. Early whole genome searches for causative genes used the technologies of the time, namely, restriction fragment-length polymorphisms (RFLP) and loss of heterozygosity (LOH) or allelic imbalance (AI) of microsatellite alleles, to identify commonly deleted or duplicated regions. However, by today's standards, these studies used widely spaced markers and fairly modest tumour 7numbers. The most commonly identified regions by these methods were 3p (35–41%), 5q (36–53%), 9p (36–65%), 9q (31–60%), 13q (43–53%), 17p (55–62%), 17q (33–71%), 18q (38–46%) and 21q (65%). Studies investigating the progressive accumulation of genetic defects, ranging from low-grade to high-grade dysplasias and carcinoma, demonstrated early loss of genetic regions at 3p21, 9p22 and 17p13 in dysplastic lesions that mirrored similar evidence from head and neck cancers [22,23] and suggests some commonality in the mutational pathway of these squamous cell aerodigestive tract cancers.
The identification of chromosomal areas of genetic alteration in oesophageal SCC led to an explosion of targeted AI studies investigating those regions most commonly altered in an attempt to localise the causative genes, but with limited initial success owing to the infancy of the human genome project. More definitive results were obtained by targeting candidate genes previously implicated in other SCCs, such as APC (5q), p16 (CDKN2A; 9p), Rb (13q), BRCA2 (13q) and p53 (17p), which have all been demonstrated to be altered in many oesophageal SCCs.
The advent of high-density microarray-based genome-wide scanning methods such as comparative genome hybridisation (CGH) and single-nucleotide polymorphism (SNP) arrays has revolutionised the field of genomics. Several relatively large-scale CGH studies in oesophageal SCC have been undertaken and have confirmed the previous allelic imbalance studies in identifying losses at 3p, 5q, 9p and 18q and gains at 13q [24]. Additional amplifications have also been frequently observed at 3q and 8q. Interestingly, chromosome 17 alterations are rarely observed by CGH, implying that no net loss or gain of genomic material results during AI on this chromosome.
The use of SNP arrays has been much more extensive, given the additional, gene-specific information that can be gleaned from such studies. Indeed, over 5000 cases and more than 7000 controls have been analysed in this way, with validation in a further 30,000 individuals [25,26]. These studies, together with meta-analyses, [27] have identified SNPs having highly significant associations with oesophageal SCC and allowed the identification of candidate genes on 10q23 (PLCE1), 20p13 (C20orf54; SLC52A3) and loci located at 2q33, 5q11, 6p21, 12q24 and 21q22.
Much of this more recent work has been undertaken in Asian populations, where the incidence of the disease is relatively high; therefore, it is gratifying that there is a degree of crossover in the chromosomal regions identified by both AI (predominantly in Caucasian populations) and CGH or SNP arrays (mainly Asian populations) and in the identification of PLCE1 and C20orf54 as candidate oesophageal SCC-specific aberrations [28].
 
Family history
In areas of high incidence, such as Iran and China, familial aggregation of squamous cancer has been recorded [29], but this is not observed in areas of low incidence suggesting that common environmental rather than hereditary risk factors are responsible.
There has been a steady accumulation of evidence pointing to a genetic component in hiatus hernia, reflux oesophagitis, Barrett's oesophagus and oesophageal AC. Familial clustering of cases of hiatus hernia, Barrett's oesophagus and gastro-oesophageal reflux have all been documented [30], with each condition cosegregating in different families. In some families, the familial tendency has been so strong as to suggest autosomal dominant inheritance [31]. Twin studies have confirmed significantly greater concordance rates in monozygotic, compared with dizygotic, twin pairs–implying that genetic factors are playing an important role in these conditions.8
 
Alcohol-metabolising enzymes
Many gastro-oesophageal cancers are strongly associated with environmental and life style risk factors. For oesophageal SCC, alcohol is an important risk factor and, in Asian populations, even light alcohol use appears to be associated with susceptibility [32]. Because exposure to high levels of acetaldehyde, the principal metabolite of alcohol, may be the cause, it is not a surprise that genome-wide SNP array studies identified chromosomes 4q23 and 12q24, where the major alcohol-metabolising enzymes, ADH cluster and ALDH2 genes, respectively, are located.
Several large studies have been carried out investigating genetic susceptibility in Asian and Western populations, including meta-analyses, and have identified increased risk of oesophageal cancer in individuals with polymorphisms in ADH1B and ALDH2 that code for proteins with lower metabolising activity. Thus, the less active G allele of rs1229984 in ADH1B is associated with an increased risk of oesophageal SCC, with the OR increasing from a modest effect to an OR of 20 in heavy drinkers, whilst the A allele of rs671 in ALDH2 shows a protective effect in nondrinkers but is strongly associated with oesophageal SCC in heavy drinkers [3335].
Similarly, polymorphisms in other detoxifying enzymes, such as NAD(P)H dehydrogenase, quinone 1 (NQO1), which has been proposed to stabilise p53, have been shown to confer an increased risk in Asian populations but not in Europeans [36].
 
Hereditary diseases with oesophageal cancer
 
Clarke–Howel–Evans–McConnell syndrome (OMIM 1485000)
Tylosis (hyperkeratosis palmaris et plantaris) is a focal, nonepidermolytic form of keratoderma. It is inherited as an autosomal dominant condition. Two types of tylosis are recognised. In type B, the hyperkeratosis appears in the first year of life but, in type A, it does not appear until about puberty. Type B is not associated with malignancy, but type A is associated with SCC of the oesophagus that usually manifests in middle to later life. Type A is also associated with other features such as oral leucokeratosis and follicular hyperkeratosis. The gene for type A tylosis has been localised to chromosome 17q25 and has been identified as RHBDF2 [37]. This 30.5-kb gene encodes a protein of 856 amino acids that is a member of a class of proteins known as rhomboids. Rhomboid proteins are intramembrane serine proteases that cleave substrates in or near the transmembrane domains. The protein product of RHBDF2 is classified as an inactive or irhomboid due to the fact that it lacks the typical proteolytic site made up of a serine, a histidine and an aspartic acid residue. Altered expression and localisation of this protein have subsequently also been shown in sporadically occurring oesophageal SCC, thus validating the AI data at 17q and implicating the gene product, iRhom2, in many of these cancers (Risk, unpublished data, 2013). Further, the iRhom2 protein has been shown to control processing of the major ‘sheddase,’ ADAM17, which controls the membrane shedding of many epidermal growth factor receptor ligands, inflammatory cytokines and adhesion molecules [38]. Indeed, in cell lines derived from tylotic skin, increased ADAM17 processing is detected together with increased production of ADAM17 substrates in response to inflammatory stimuli, whilst increased EGF signalling and cell migration are also observed (Kelsell, personal communication, 2014). Five families have been documented and, in four of these families, a mutation has been found in the RHBBF2 gene. The risk of developing oesophageal SCC in the Liverpool family has been calculated to be 95% at the age of 65 [39].9
 
Dyskeratosis congenital (OMIM 305000)
This condition is characterised by cutaneous pigmentation, premature greying, nail dystrophy, leucoplakia, thrombocytopenia, anaemia, testicular atrophy and a predisposition to cancer, including oesophageal cancer.
 
Fanconi's anaemia (OMIM 227650)
Fanconi's anaemia is a rare, mainly autosomal recessive disorder, resulting in failure of DNA repair. It, therefore, leads to the development of haematological (acute myeloid leukaemia) and nonhaematological (head and neck, oesophageal, gastrointestinal, vulvar and anal) malignancies.
 
Recessive dystrophic epidermolysis bullosa (OMIM 226600)
This is one of a number of inherited disorders resulting in the formation of skin blisters on minimal trauma. Patients have an increased of cancer of the skin and oesophagus.
 
SUMMARY
The demography of oesopahgeal cancer has been changing over the past few decades. Not only has adenocarcinoma overtaken squamous cancer as the most frequent oesophageal cancer in Western countries, but there has been an improvement in the 5-year survival rate as a result of the earlier recognition and treatment of less advanced lesions. Several predisposing environmental risk factors have been identified, the removal of which should potentially result in further reductions in incidence and mortality, although at present this remains to be seen.
A significant compilation of genetic information now exists for oesophageal SCC, but the challenge of realizing this information in clinical practice remains. The identification and implementation of genetic biomarkers for this disease remains problematic. Perhaps the genetic analysis of this disease should be focussed towards the identification of novel druggable targets to increase treatment options which can then be offered to targeted patient populations of the basis of the genetic analysis of the tumour.
REFERENCES
  1. European age-standardised rates calculated by the Statistical Information Team at Cancer Research UK, 2011 using data from GLOBOCAN, IARC, version 1.2.
  1. Seigel R, Naishadham D, Jemal A. Cancer statistics, 2013. CA Cancer J Clin 2013; 63:11.
  1. Kamangar F, Chow W-H, Abnet CC, et al. Environmental causes of esophageal cancer. In: Gastroenterol Clin North Am. Esophageal Cancer 2009; 38:27–57.
  1. Baquet CR, Commiskey P, Mack K, et al. Esophageal cancer epidemiology in blacks and whites: racial and gender disparities in incidence, mortality, survival rates and histology. J Natl Med Assoc 2005; 97:1471–1478.
  1. Roshandel G, Semnani S, Malekzadeh R, et al. Polycyclic aromatic hydrocarbons and esophageal squamous cell carcinoma. Arch Iranian Med 2012; 15:713–722.
  1. Rushton L, Hutchings SJ, Fortunato L, et al. Occupational cancer burden in Great Britain. Br J Cancer 2012; 107:S3–S7.
  1. Zendehdel K, Nyren O, Edberg A, et al. Risk of esophageal adenocarcinoma in achalasia patients, a retrospective cohort study in Sweden. Am J Gastroenterol 2011; 106:57–61.
  1. Lagergren J, Bergström R, Lindgren A, et al. Symptomatic gastroesophageal reflux as a risk factor for esophageal adenocarcinoma. N Engl J Med 1999; 340:825–831.
  1. Hongo M, Nagasaki Y, Shoji T. Epidemiology of esophageal cancer: Orient to Occident. Effects of chronology, geography and ethnicity. J Gastroenterol Hepatol 2009; 24:729–735.
  1. Tutian R. Obesity and GERD: pathophysiology and effect of bariatric surgery. Curr Gastroenterol Rep 2011; 13:205–212.
  1. Merry AHH, Schouten LJ, Goldbohm R, et al. Body mass index, height and risk of adenocarcinoma of the oesophagus and gastric cardia: a prospective cohort study. Gut 2007; 56:1503–1511.
  1. Green PH, Fleischauer AT, Bhagat G, et al. Risk of malignancy in patients with celiac disease. Am J Med 2003; 115:191–195.
  1. Haldun S, Mendenhall WM, Amdur RJ, et al. Synchronous and metachronous squamous cell carcinomas of the head and neck mucosal sites. J Clin Oncol 2001; 19:1358–1362.
  1. Ruol A, Rampado S, Parente A, et al. Caustic ingestion and oesophageal cancer: intra- and peri-tumoural fibrosis is associated with a better prognosis. Eur J Cardiothoracic Surg 2010; 38:659–664.
  1. Micke O, Schäfer U, Glashorster M, et al. Radiation-induced oesophageal carcinoma 30 years after mediastinal irradiation: case report and review of the literature. Jap J Clin Oncol 1999; 29(3):164–170.
  1. Chisholm M. The association between webs, iron and post-cricoid carcinoma. Postgrad Med J 1974:215–219.
  1. Islami F, Kamangar F. Helicobacter pylori and esophageal cancer risk: a meta-analysis. Cancer Prev Res (Phila) 2008; 1:329–338.
  1. Freedman J, Ye W, Näslund E, et al. Association between cholecystectomy and adenocarcinoma of the esophagus. Gastroenterology 2001; 121(3):548–553.
  1. Syrjänen KJ. HPV infections and oesophageal cancer. J Clin Pathol 2002; 55:721–728.11
  1. Vinogradova Y, Hippisley-Cox J. Exposure to biphosphates and risk of gastrointestinal cancers: series of nested, case-control studies with QResearch and CPRD data. BMJ 2013; 346:f114.
  1. Corley DA, Kerlikowske K, Verma R, et al. Protective association of aspirin/NSAIDs and esophageal cancer: a systematic review and meta-analysis. Gastroenterology, 2003; 124:47–56.
  1. Roth MJ, Hu N, Emmert-Buck MR, et al. Genetic progression and heterogeneity associated with the development of esophageal squamous cell carcinoma. Cancer Res 2001; 61:4098–4104.
  1. Zhang L, Poh CF, Williams M, et al. Loss of heterozygosity (LOH) profiles—validated risk predictors for progression to oral cancer. Cancer Prev Res 2012; 5:1081–1089.
  1. Pack SD, Karkera JD, Zhuang ZP, et al. Molecular cytogenetic fingerprinting of esophageal squamous cell carcinoma by comparative genomic hybridization reveals a consistent pattern of chromosomal alterations. Genes Chrom Cancer 1999; 25:160–168.
  1. Wang LD, Zhou FY, Li XM, et al. Genome-wide association study of esophageal squamous cell carcinoma in Chinese subjects identifies susceptibility loci at PLCE1 and C20orf54. Nat Genet 2010; 42:759–763.
  1. Wu C, Kraft P, Zhai K, et al. Genome-wide association analyses of esophageal squamous cell carcinoma in Chinese identify multiple susceptibility loci and gene-environment interactions. Nat Genet 2012; 44:1090–1097.
  1. Abnet CC, Wang Z, Song X, et al. Genotypic variants at 2q33 and risk of esophageal squamous cell carcinoma in China: a meta-analysis of genome-wide association studies. Hum Mol Genet 2012; 21:2132–2141.
  1. Palmer AJ, Lochhead P, Hold GL, et al. Genetic variation in C20orf54, PLCE1 and MUC1 and the risk of upper gastrointestinal cancers in Caucasian populations. Eur J Cancer Prev 2012; 21:541–544.
  1. Robertson EV, Jankowski JA. Genetics of gastroesophageal cancer: paradigms, paradoxes, and prognostic utility. Am J Gastroenterol 2008; 103:443–449.
  1. Sappati Biyyani RS, Chessler L, McCain E, et al. Familial trends of inheritance in gastro-oesophageal reflux disease, Barrett's oesophagus and Barrett's adenocarcinoma: 20 families. Dis Esophagus 2007; 20:53–57.
  1. Islami F, Fedirko V, Tramacere I, et al. Alcohol drinking and esophageal squamous cell carcinoma with focus on light-drinkers and never-smokers: a systematic review and meta-analysis. Int J Cancer 2011; 129:2473–2484.
  1. Fang P, Jiao S, Zhang X, et al. Meta-analysis of ALDH2 variants and esophageal cancer in Asians. Asian Pac J Cancer Prev 2011; 12:2623–2627.
  1. Zhang G, Mai R, Huang B. ADH1B Arg47His polymorphism is associated with esophageal cancer risk in high-incidence Asian population: evidence from a meta-analysis. PLoS One 2010; 5:e13679.
  1. Tanaka F, Yamamoto K, Suzuki S, et al. Strong interaction between the effects of alcohol consumption and smoking on oesophageal squamous cell carcinoma among individuals with ADH1B and/or ALDH2 risk alleles. Gut 2010; 59:1457–1464.
  1. Wang Z, Hu J, Zhong J. Meta-analysis of the NAD(P)H:quinine oxidoreductase 1 gene 609 C>T polymorphism with esophageal cancer risk. DNA Cell Biol 2012; 31:560–567.
  1. Blaydon DC, Etheridge SL, Risk JM, et al. RHBDF2 mutations are associated with tylosis, a familial esophageal cancer syndrome. Am J Hum Genet. 2012; 90:340–346.
  1. Adrain C, Freeman M. New lives for old: evolution of pseudoenzyme function illustrated by iRhoms. Nat Rev Mol Cell Biol 2012; 13:489–498.
  1. Ellis A, Field JK, Field AE, et al. Tylosis associated with carcinoma of the oesophagus and oral leukoplakia in a large Liverpool family—a review of six generations. Eur J Cancer, Oral Oncol 1994; 30B:102–112.12