early diagnostic biomarkers for esophageal adenocarcinoma ...early diagnostic biomarkers for...

26
Review Early Diagnostic Biomarkers for Esophageal AdenocarcinomaThe Current State of Play Alok Kishorkumar Shah 1 , Nicholas A. Saunders 1 , Andrew P. Barbour 2 , and Michelle M. Hill 1 Abstract Esophageal adenocarcinoma (EAC) is one of the two most common types of esophageal cancer with alarming increase in incidence and very poor prognosis. Aiming to detect EAC early, currently high-risk patients are monitored using an endoscopic-biopsy approach. However, this approach is prone to sampling error and interobserver variability. Diagnostic tissue biomarkers related to genomic and cell-cycle abnormal- ities have shown promising results, although with current technology these tests are difficult to implement in the screening of high-risk patients for early neoplastic changes. Differential miRNA profiles and aberrant protein glycosylation in tissue samples have been reported to improve performance of existing tissue-based diagnostic biomarkers. In contrast to tissue biomarkers, circulating biomarkers are more amenable to population-screening strategies, due to the ease and low cost of testing. Studies have already shown altered circulating glycans and DNA methylation in BE/EAC, whereas disease-associated changes in circulating miRNA remain to be determined. Future research should focus on identification and validation of these circulating biomarkers in large-scale trials to develop in vitro diagnostic tools to screen population at risk for EAC development. Cancer Epidemiol Biomarkers Prev; 22(7); 1185–209. Ó2013 AACR. Introduction After heart disease, cancer is the second leading cause of death globally. Four major cancer sites account for half of the cancer-related mortalities: lung, colorectal, prostate in men, and breast in women. In past 2 decades, a steady decrease in deaths of these 4 major site malignancies led to an overall decrease in cancer-related death rates in men and women (1). In contrast, the incidence of esophageal adenocarcinoma (EAC) is increasing faster than any other cancer type. EAC together with esophageal squamous cell carcinoma (ESCC) is the eighth most-common cancer by prevalence and sixth most-common cause of cancer-relat- ed death globally (2). In 1970s, the incidence of EAC represented less than 5% of total esophageal cancer, and a majority of esophageal cancer cases diagnosed were ESCC. Over a period of 3 decades, EAC incidences have been increasing continuously, especially in western coun- tries among Caucasians. Now almost half of the esoph- ageal malignancy cases diagnosed are EAC (3, 4). EAC and ESCC show marked differences in their geographic spread. EAC is more common in developed countries such as the United Kingdom (8 in 100,000 individuals; ref. 5), Australia, and the United States. Within Europe, southern Europe has the highest EAC incidence (5). On the other side, ESCC is the most common type of esoph- ageal cancer among developing Asian countries (6). Racial disparity also occurs between the 2 types of esophageal cancer. ESCC is more prevalent among Blacks, whereas EAC is at least twice as common in Whites as compared with other ethnic groups (7, 8). Once diagnosed, Black patients showed poorer overall survival than Whites (9, 10). Taken together, strong genetic and environmental factors relating to ethnicity and geographic distribution seem to be playing critical roles in the incidence of esoph- ageal cancer. Studies also suggest possible links between socioeconomic status and the prevalence of esophageal cancer phenotype (6). Risk Factors In the majority of cases, EAC is diagnosed at a late stage, leading to a poor 5-year survival of less than 15% (11). Hence, recent research for EAC has focused on under- standing risk factors and the identification of early diag- nostic biomarkers. Esophageal cancer is unlikely to develop in individuals younger than 40 years of age; however, after that the incidence increases significantly with each decade of life (9). Changing lifestyle and food habits are primarily responsible for the dramatic epidemiologic changes in EAC as described in recent reviews (11–13). Known EAC risk factors include accumulation of visceral fat in the abdomen (14), male gender, high intake of dietary fat and cholesterol with low intake of fruits and vegetables (15), tobacco smoking (16), reduction in Helicobacter pylori Authors' Afliations: 1 The University of Queensland Diamantina Institute; and 2 School of Medicine, The University of Queensland, Woolloongabba, Queensland, Australia Corresponding Author: Michelle M. Hill, The University of Queensland Diamantina Institute, Level 5, Translational Research Institute, 37 Kent Street, Woolloongabba, QLD 4102. Phone: 61-7-3443-7049; Fax: 61-7- 3443-6966; E-mail: [email protected] doi: 10.1158/1055-9965.EPI-12-1415 Ó2013 American Association for Cancer Research. Cancer Epidemiology, Biomarkers & Prevention www.aacrjournals.org 1185 on June 7, 2021. © 2013 American Association for Cancer Research. cebp.aacrjournals.org Downloaded from Published OnlineFirst April 10, 2013; DOI: 10.1158/1055-9965.EPI-12-1415

Upload: others

Post on 28-Jan-2021

2 views

Category:

Documents


0 download

TRANSCRIPT

  • Review

    Early Diagnostic Biomarkers for EsophagealAdenocarcinoma—The Current State of Play

    Alok Kishorkumar Shah1, Nicholas A. Saunders1, Andrew P. Barbour2, and Michelle M. Hill1

    AbstractEsophageal adenocarcinoma (EAC) is one of the two most common types of esophageal cancer with

    alarming increase in incidence and very poor prognosis. Aiming to detect EAC early, currently high-risk

    patients are monitored using an endoscopic-biopsy approach. However, this approach is prone to sampling

    error and interobserver variability. Diagnostic tissue biomarkers related to genomic and cell-cycle abnormal-

    ities have shown promising results, although with current technology these tests are difficult to implement in

    the screening of high-risk patients for early neoplastic changes. Differential miRNA profiles and aberrant

    protein glycosylation in tissue samples have been reported to improve performance of existing tissue-based

    diagnostic biomarkers. In contrast to tissue biomarkers, circulating biomarkers are more amenable to

    population-screening strategies, due to the ease and low cost of testing. Studies have already shown altered

    circulating glycans and DNA methylation in BE/EAC, whereas disease-associated changes in circulating

    miRNA remain to be determined. Future research should focus on identification and validation of these

    circulating biomarkers in large-scale trials to develop in vitro diagnostic tools to screen population at risk for

    EAC development. Cancer Epidemiol Biomarkers Prev; 22(7); 1185–209. �2013 AACR.

    IntroductionAfterheart disease, cancer is the second leading cause of

    death globally. Four major cancer sites account for half ofthe cancer-related mortalities: lung, colorectal, prostatein men, and breast in women. In past 2 decades, a steadydecrease in deaths of these 4 major site malignancies ledto an overall decrease in cancer-related death rates inmenand women (1). In contrast, the incidence of esophagealadenocarcinoma (EAC) is increasing faster than any othercancer type. EAC togetherwith esophageal squamous cellcarcinoma (ESCC) is the eighth most-common cancer byprevalence and sixth most-common cause of cancer-relat-ed death globally (2). In 1970s, the incidence of EACrepresented less than 5% of total esophageal cancer, anda majority of esophageal cancer cases diagnosed wereESCC. Over a period of 3 decades, EAC incidences havebeen increasing continuously, especially inwestern coun-tries among Caucasians. Now almost half of the esoph-ageal malignancy cases diagnosed are EAC (3, 4). EACand ESCC show marked differences in their geographicspread. EAC is more common in developed countriessuch as the United Kingdom (8 in 100,000 individuals;

    ref. 5), Australia, and the United States. Within Europe,southern Europe has the highest EAC incidence (5). Onthe other side, ESCC is the most common type of esoph-ageal cancer amongdevelopingAsian countries (6). Racialdisparity also occurs between the 2 types of esophagealcancer. ESCC is more prevalent among Blacks, whereasEAC is at least twice as common in Whites as comparedwith other ethnic groups (7, 8). Once diagnosed, Blackpatients showed poorer overall survival than Whites(9, 10). Taken together, strong genetic and environmentalfactors relating to ethnicity and geographic distributionseem to be playing critical roles in the incidence of esoph-ageal cancer. Studies also suggest possible links betweensocioeconomic status and the prevalence of esophagealcancer phenotype (6).

    Risk FactorsIn themajority of cases, EAC is diagnosed at a late stage,

    leading to a poor 5-year survival of less than 15% (11).Hence, recent research for EAC has focused on under-standing risk factors and the identification of early diag-nostic biomarkers.

    Esophageal cancer is unlikely to develop in individualsyounger than 40 years of age; however, after that theincidence increases significantly with each decade of life(9). Changing lifestyle and food habits are primarilyresponsible for the dramatic epidemiologic changes inEAC as described in recent reviews (11–13). Known EACrisk factors include accumulation of visceral fat in theabdomen (14), male gender, high intake of dietary fat andcholesterol with low intake of fruits and vegetables (15),tobacco smoking (16), reduction in Helicobacter pylori

    Authors' Affiliations: 1The University of Queensland Diamantina Institute;and 2School of Medicine, The University of Queensland, Woolloongabba,Queensland, Australia

    Corresponding Author: Michelle M. Hill, The University of QueenslandDiamantina Institute, Level 5, Translational Research Institute, 37 KentStreet, Woolloongabba, QLD 4102. Phone: 61-7-3443-7049; Fax: 61-7-3443-6966; E-mail: [email protected]

    doi: 10.1158/1055-9965.EPI-12-1415

    �2013 American Association for Cancer Research.

    CancerEpidemiology,

    Biomarkers& Prevention

    www.aacrjournals.org 1185

    on June 7, 2021. © 2013 American Association for Cancer Research. cebp.aacrjournals.org Downloaded from

    Published OnlineFirst April 10, 2013; DOI: 10.1158/1055-9965.EPI-12-1415

    http://cebp.aacrjournals.org/

  • infections (17), and Barrett’s esophagus (BE), a metaplas-tic change to the esophageal lining. Individuals withBarrett’s esophagus carry almost 30 to 125 times morerisk for EAC development, and 0.5% to 1% of patientswith Barrett’s esophagus are estimated to develop EACeach year (18). Barrett’s esophagus is characterized byreplacement of normal stratified squamous epitheliumwith metaplastic columnar epithelium and is consideredto be a successful adaptation of the distal esophagus inresponse to chronic gastroesophageal reflux disorder(GERD; ref. 19).

    GERD is a very common condition in the western pop-ulation with around 20% reporting weekly symptoms ofheartburn and acid regurgitation (20). Refluxate-contain-ing bile acid, along with gastric acid, is considered to bemore harmful, leading to inflammation, ulceration, Bar-rett’s esophagus, and ultimately EAC. Development ofBarrett’s esophagus is a slow process and distinctivemucus-secreting goblet cell formation can take 5 to 10years (21, 22). Typically, EAC develops through metapla-sia–dysplasia–carcinoma sequence involving genetic andepigenetic modifications, leading to uncontrolled cellproliferation, and is characterized by the presence ofintestinal metaplasiawith low-grade (LGD) to high-gradedysplasia (HGD), which eventuallymay progress to inva-sive carcinoma (20).

    Current Diagnosis ScenarioTo detect pathologic changes leading to EAC develop-

    ment before onset of disease, current clinical practiceinvolves endoscopic screening of patients with high-riskGERD and to characterize the degree of dysplasia inbiopsy samples collected during endoscopy (23, 24).Enrollment of patients into an endoscopic screening pro-grammay be facilitated by a patient questionnaire of self-evaluated symptoms/complications (25, 26). Onceenrolled into the screening program, a patient undergoesendoscopy-biopsy every 3 months to 2 years dependingon the degree of dysplasia, during which 4 quadrantbiopsy samples are taken every 1 to 2 cm and evaluatedfor histologic changes by expert pathologists (23, 24). As asignificant number of patients histologically diagnosedwith HGD develop EAC, endoscopic mucosal ablation oresophageal resection (esophagectomy) are options to stopfurther disease progression in those high-risk patients(27, 28). Significantly improved survival is observed inpatients diagnosed at an early stage during surveillanceendoscopy program as compared with symptomaticallydiagnosed EAC (29–32).

    Although current screeningmethodology shows prom-ise, outcome of endoscopy-biopsy in many cases is non-reproducible due to interobserver variability and sam-pling error (28, 33). Furthermore, histologic dysplasticchanges may be patchy and present heterogeneously inthe tissue sample. This makes the diagnosis challenging,especially in the early stages of transition to LGD (28, 34).In up to 40%of patients, invasive cancer has been found inresected tissue despite negative endoscopic examination

    for the malignancy (35). Moreover, false-positive resultsalso occur, meaning despite intramucosal carcinoma in abiopsy, the subsequently resected tissue has no signs ofcarcinoma (28). These evidence suggest dysplasia gradingis an imperfect measure of cancer risk.

    Despite extensive screening with currently availabletechniques, more than 80% of EACs are diagnosed with-out any prior diagnosis of Barrett’s esophagus or GERD(36, 37). According to an estimate, more than 80% ofBarrett’s esophagus cases are undiagnosed and thereforeare not getting the benefit of the screening program (38).On the other hand, a large proportion of patients under-going routine biopsy screening do not progress to EAC(13). These suggest inability of current methodologies inscreening population to detect high-risk patients and todistinguish between disease progressors from nonpro-gressors. In addition, the screening procedure is not verycost-effective (39). To overcome these challenges, adjunctuse of biomarker has been proposed to stratify the riskassociated with EAC development.

    Biomarkers in EACAccording to United States’ NIH, a biomarker is "a

    characteristic that is objectively measured and evaluatedas an indicator of normal biologic processes, pathogenicprocesses, or pharmacologic responses to a therapeuticintervention (40)."

    In transit from intestinal metaplasia to LGD to HGD toEAC, cells acquire abilities to become self-sufficient forgrowth, evade apoptosis, proliferate uncontrollably, pro-mote angiogenesis, invade underlined epithelium, andstart to metastasize. These changes are accompanied withhistologic changes in tissue architecture, genomic insta-bility, development of tumor microenvironment, modu-lation of immune response, and are therefore reflected inbody fluids (serum/plasma/mucus/urine) or tissue sam-ples and differentiate in terms of their genome/prote-ome/metabolome profile (41). Thus, a biomarker can befrom any of these sources and reflect underlying patho-logic or homeostatic changes. Table 1 summarizes differ-ent classes of biomarkers proposed for BE/EAC.

    National Cancer Institute Early Detection ResearchNetwork (EDRN) guidelines outline biomarker discoveryanddevelopment to a 5-phase process summarized below(42) and depicted in Fig. 1.

    Phase I—Preclinical exploratory study: it comparesnormal versus cancer samples (body fluids/tissue)using technologies such as genomics, microarrayexpression, proteomics, immunohistochemistry, orimmunoblotting to detect significant changes inproteins/genes/metabolites between the groups.

    Phase II—Clinical assay development and validation: it isaimed at developing a clinical assay using a minimallyinvasive sample collection method. The assay is meantto be robust, reproducible, and suitable for storedclinical samples to be used in later phases ofdevelopment. At the end of this phase, one should

    Shah et al.

    Cancer Epidemiol Biomarkers Prev; 22(7) July 2013 Cancer Epidemiology, Biomarkers & Prevention1186

    on June 7, 2021. © 2013 American Association for Cancer Research. cebp.aacrjournals.org Downloaded from

    Published OnlineFirst April 10, 2013; DOI: 10.1158/1055-9965.EPI-12-1415

    http://cebp.aacrjournals.org/

  • expect high specificity and sensitivity for the assay.However, it remains to be determined how early thebiomarker can predict the disease.

    Phase III—Retrospective longitudinal repository studies:the assay is applied on prospectively collected storedsamples to determine the ability of the biomarker todetect the disease before clinical presentation. If so, thencriteria for positive screening is determined for futureuse.

    Phase IV—Prospective screening: the test is prospectivelyapplied to real population to detect the extent andcharacteristic of disease detected by the biomarker. Thisphase gives positive predictive value for the test andgives an idea about feasibility for last phase of controltrials.

    Phase V—Cancer control studies: it comprises large-scaleclinical trial to determine the impact of new screeningprocess on the disease burden in the community.

    With respect to EAC, none of the biomarkers, includinghigh-grade dysplasia, have been evaluated in phase V,whereas very feware evaluated in phase III and IV. Figure1 summarizes proposed EAC biomarkers and how wellthey are characterized in the process of biomarker dis-covery. The following sections will discuss some of theclasses of BE/EAC biomarkers.

    Genomic InstabilityMany groups have studied genomic instability induced

    by aneuploidy, tetraploidy, DNAmethylation, allelic lossand shown some predictive power for these changes. Arole for hypermethylation in the promoter regions oftumor-suppressor genes during the development of EAChas also been well established. Table 2 summarizes DNAmethylation changes associated with metaplasia–dyspla-sia–carcinoma development. In the majority of patients,methylation changes are acquired very early during EACdevelopment, hence these alterations could be used as anearly diagnostic biomarker. Apart from discriminatingpatients at different stages of EAC development, DNAmethylation signatures may be useful as predictors forprogression from Barrett’s esophagus to EAC (43, 44) andfor response to chemotherapy and survival in patientswith EAC (45, 46).

    Although the individual genomic abnormality has thepotential to diagnose disease at different stages, bestresults are obtained when they are used in combination(47–49). LOH at chromosome 9p and 17p locus are con-sidered to be early events during Barrett’s esophaguspathogenesis (50). If present with other chromosomalalterations such as aneuploidy and tetraploidy, itincreases the 10-year risk for development of EAC from12% to approximately 80% (51). However, with the cur-rent flow cytometry technology, it is technically verychallenging for clinical laboratories to assess these geno-mic biomarkers in the samples, which limits widespreaduse of these biomarkers in the clinic.

    Alternatively, genomic alterations canbedetected at theprotein level using immunohistochemistry. One of themost common and earliest genomic abnormality occurs atchromosome 17p, which codes for tumor-suppressor p53protein. Loss of p53 protein expression in tissue samplescorrelates with disease progression (52). However, as p53expression only reflects alterations at one particular gene,it has lower predictive value as comparedwith techniquesmonitoring multiple genomic abnormalities. Further-more, sensitivity drops as mutations or deletions at geno-mic level may not necessarily be detected at the proteinlevel (53).

    In line with the genomic abnormalities described ear-lier, single-nucleotide polymorphism (SNP)–based geno-typing can also stratify cancer risk in patients with Bar-rett’s esophagus. As summarized in Table 3, in the past

    None

    Phase V: Cancer control studies

    Bio

    mar

    ker d

    isco

    very

    and

    dev

    elop

    men

    t

    Phase IV: Prospective screening

    Phase III: Retrospective longitudinal repository studies

    Phase I and II: Preclinical exploration, clinical assay development and validation

    High-gradedysplasis

    DNA methylation, LOH,ploidy, p53 loss, cyclin D1

    PCNA, Ki-67, EGFR, COX-2,miRNA, cMYC, HER2, NF-κB, Bcl-2,VEGF, E-cadherin, p16 abnormalities,

    β-catenin, glycoproteins, etc.

    Figure 1. Summary of current BE/EAC Biomarkers with respect to EDRNclinical phase of development.

    Table 1. Comprehensive summary of differentclasses of BE/EAC biomarkers

    Biomarker class Ref.

    Tissue biomarkersGenomic abnormalities(ploidy and LOH)

    (47–51)

    DNA methylation Refer to Table 2SNPs/expression array studies Refer to Table 3

    Inflammatory markersCOX-2 (69, 72–77)NF-kB (78–81)Cytokines (67, 79, 81–86)MMPs (87–93)Cell-cycle abnormalities (94, 95, 101)miRNA Refer to Table 4Glycosylation changes (121, 123–125)

    Circulatory biomarkersDNA methylation changes (130–132)Glycan alterations (135–138)Metabolic profiling (142–145)

    Biomarkers for Esophageal Adenocarcinoma

    www.aacrjournals.org Cancer Epidemiol Biomarkers Prev; 22(7) July 2013 1187

    on June 7, 2021. © 2013 American Association for Cancer Research. cebp.aacrjournals.org Downloaded from

    Published OnlineFirst April 10, 2013; DOI: 10.1158/1055-9965.EPI-12-1415

    http://cebp.aacrjournals.org/

  • Tab

    le2.

    Sum

    maryof

    hypermethy

    latedge

    nesduringBE/EAC

    dev

    elop

    men

    t Num

    ber

    (%)o

    fsa

    mplessh

    owinghy

    permethy

    lationorstud

    yfind

    ings

    Gen

    eLo

    cation

    Func

    tion

    Metho

    dNorm

    alBE

    LGD

    HGD

    EAC

    Ref.

    p16

    (orCDKN2A

    orINK4A

    )9p

    21Cyc

    lin-dep

    ende

    ntkina

    seinhibitor

    Methy

    latio

    n-sp

    ecificPCR

    5/9(56%

    )14

    /18(77%

    )—

    —18

    /21(85%

    )(146

    )

    Methy

    latio

    n-se

    nsitive

    sing

    le-stran

    dco

    nformationan

    alysis

    0/10

    (0%)

    4/12

    (33%

    )3/11

    (27%

    )3/10

    (30%

    )18

    /22(82%

    )(147

    )

    Methy

    latio

    n-sp

    ecificPCR

    0/17

    (0%)

    14/47(30%

    )9/27

    (32%

    )10

    /18(56%

    )22

    /41(54%

    )(148

    )Methy

    latio

    n-sp

    ecificPCR

    2/64

    (3%)

    14/93(15%

    )—

    —34

    /76(45%

    )(149

    )Methy

    latio

    n-sp

    ecificPCR

    —3/10

    (30%

    )—

    —5/11

    (45%

    )(150

    )Methy

    latio

    n-sp

    ecificPCR

    —27

    /41(66

    %)

    21/45(47%

    )17

    /21(81%

    )65

    /107

    (61%

    )(151

    )Methy

    latio

    n-sp

    ecificPCR

    0%1/15

    (7%)

    4/20

    (20%

    )12

    /20(60%

    )8/15

    (53%

    )(152

    )Methy

    latio

    n-sp

    ecificPCR

    Sep

    aratelyde

    term

    ined

    exon

    1an

    d2methy

    latio

    n.Five

    of16

    (31%

    )exo

    n-1,

    8/16

    (50%

    )exo

    n2in

    EAC

    patient

    samples

    show

    edhy

    permethy

    latio

    n.Exo

    n2

    methy

    latio

    nco

    rrelates

    with

    stag

    eof

    thetumor

    (P¼

    0.01

    )

    (153

    )

    O6-M

    ethy

    lgua

    nine

    -10

    q26

    DNArepa

    irMethy

    light

    tech

    nique

    2/10

    (20%

    )8/13

    (62%

    )—

    —84

    /132

    (64%

    )(154

    )DNA Methy

    ltran

    sferase

    (orMGMT)

    Methy

    latio

    n-sp

    ecificPCR

    6/29

    (21%

    )24

    /27(89%

    )13

    /13(100

    %)

    37/47(79%

    )(155

    )

    APC

    5q21

    -q22

    Wnt/b-caten

    insign

    aling

    Methy

    latio

    n-sp

    ecificPCR

    0/17

    (0%)

    24/48(50%

    )14

    /28(50%

    )14

    /18(78%

    )20

    /32(63%

    )(148

    )

    Methy

    latio

    n-se

    nsitive

    sing

    le-stran

    dco

    nformation

    analysis

    and

    methy

    latio

    n-se

    nsitive

    dot

    blotas

    say

    0/16

    (0%)

    11/11(100

    %)

    ——

    20/21(95%

    )(156

    )Eight

    of14

    histolog

    ically

    norm

    alga

    stric

    muc

    osaad

    jace

    ntto

    EAC

    show

    edsign

    ifica

    ntly

    differen

    tmethy

    latio

    nof

    APC

    promoter.

    (157

    )

    GSTM

    21p

    13.3

    Antioxidan

    tsan

    dprotec

    tionag

    ains

    tDNAdam

    age

    Bisulfite

    pyrose

    quen

    cing

    (sam

    ple

    size

    :EAC-

    100,

    BE-11,

    dys

    plasia-

    11,n

    ormal

    esop

    hage

    al/

    gastric

    muc

    osa-37

    )

  • Tab

    le2.

    Sum

    maryof

    hypermethy

    latedge

    nesduringBE/EAC

    dev

    elop

    men

    t(Con

    t'd)

    Num

    ber

    (%)ofsa

    mplessh

    owinghy

    permethy

    lationorstud

    yfind

    ings

    Gen

    eLo

    cation

    Func

    tion

    Metho

    dNorm

    alBE

    LGD

    HGD

    EAC

    Ref.

    Tach

    ykinin-1

    (TAC1)

    7q21

    -22

    Smoo

    thmus

    cle

    contractility,e

    pith

    elial

    iontran

    sport,

    vasc

    ular

    permea

    bility

    andim

    mun

    efunc

    tion

    Methy

    latio

    n-sp

    ecificPCR

    5/67

    (7.5%)

    38/60(63.3%

    )12

    /19(63.2%

    )11

    /21(52.4%

    )41

    /67(61.2%

    )(162

    )

    Rep

    rimo

    2q23

    Reg

    ulates

    p53

    -med

    iatedce

    ll-cy

    cle

    arrest

    inG2-pha

    se

    Methy

    latio

    n-sp

    ecificPCR

    0/19

    (0%

    )9/25

    (36%

    )—

    7/11

    (64%

    )47

    /75(63%

    )(163

    )

    E-C

    adhe

    rin16

    q22

    .1Caþ

    2-dep

    enden

    tintercellularad

    hesion

    andmaintains

    norm

    altis

    suearch

    itecture

    Methy

    latio

    n-sp

    ecificPCR

    0/4(0%)

    ——

    —26

    /31(84%

    )(164

    )

    SOCS-3

    17q25

    .3Inhibits

    cytokine

    sign

    aling

    Methy

    latio

    n-sp

    ecificPCR

    0%4/30

    (13%

    )6/27

    (22%

    )20

    /29(69%

    )14

    /19(74%

    )(165

    )

    SOCS-1

    16p13

    .13

    0%0/30

    (0%

    )1/27

    (4%

    )6/29

    (21%

    )8/19

    (42%

    )

    Sec

    retedfrizzled

    -related

    proteins(SFR

    P)

    SFR

    P1

    8p11

    .21

    Wnt

    antago

    nist

    Methy

    latio

    n-sp

    ecificPCR

    7/28

    (25%

    )30

    /37(81%

    )—

    —37

    /40(93%

    )(166

    )SFR

    P2

    4q31

    .318

    /28(64%

    )33

    /37(89%

    )—

    —33

    /40(83%

    )SFR

    P1

    8p11

    .21

    Methy

    latio

    n-se

    nsitive

    sing

    le-stran

    dco

    nformationan

    alysis

    andmethy

    latio

    n-se

    nsitive

    dot

    blotas

    say

    1/12

    (8%

    )6/6(100

    %)

    ——

    23/24(96%

    )(156

    )

    SFR

    P2

    4q31

    .311

    /15(73%

    )6/6(100

    %)

    ——

    19/25(76%

    )SFR

    P4

    7p14

    .1Methy

    latio

    n-sp

    ecificPCR

    9/28

    (32%

    )29

    /37(78%

    )—

    —29

    /40(73%

    )(166

    )SFR

    P5

    10q24

    .16/28

    (21%

    )27

    /37(73%

    )—

    —34

    /40(85%

    )Plako

    philin-1

    (PKP1)

    1q32

    Cella

    dhe

    sion

    and

    intrac

    ellularsign

    aling

    Methy

    latio

    n-sp

    ecificPCR

    5/55

    (9.1%)

    5/39

    (12.8%

    )—

    1/4(25%

    )20

    /60(33.3)

    (167

    )

    GATA

    -48p

    23.1-p22

    Tran

    scrip

    tionfactor

    andregu

    late

    cell

    differen

    tiatio

    n

    Methy

    latio

    n-sp

    ecificPCR

    0/17

    (0%

    )—

    ——

    31/44(71%

    )(168

    )

    GATA

    -520

    q13

    .33

    0/17

    (0%

    )—

    ——

    24/44(55%

    )CDH13

    (orH-

    cadhe

    rinor

    T-ca

    dhe

    rin)

    16q24

    Cella

    dhe

    sion

    Methy

    latio

    n-sp

    ecificPCR

    0/66

    (0%

    )42

    /60(70%

    )15

    /19(78.9%

    )16

    /21(76.2)

    51/67(76.1%

    )(169

    )

    (Con

    tinue

    don

    thefollo

    wingpag

    e)

    Biomarkers for Esophageal Adenocarcinoma

    www.aacrjournals.org Cancer Epidemiol Biomarkers Prev; 22(7) July 2013 1189

    on June 7, 2021. © 2013 American Association for Cancer Research. cebp.aacrjournals.org Downloaded from

    Published OnlineFirst April 10, 2013; DOI: 10.1158/1055-9965.EPI-12-1415

    http://cebp.aacrjournals.org/

  • Tab

    le2.

    Sum

    maryof

    hypermethy

    latedge

    nesduringBE/EAC

    dev

    elop

    men

    t(Con

    t'd)

    Num

    ber

    (%)o

    fsa

    mplessh

    owinghy

    permethy

    lationorstud

    yfind

    ings

    Gen

    eLo

    cation

    Func

    tion

    Metho

    dNorm

    alBE

    LGD

    HGD

    EAC

    Ref.

    NELL

    -1(nel-like1)

    11p15

    Tumor

    suppress

    orMethy

    latio

    n-sp

    ecificPCR

    0/66

    (0%)

    28/60(46.7%

    )8/19

    (42.1%

    )13

    /21(61.9%

    )32

    /67(47.8%

    )(170

    )Eye

    sAbse

    nt4

    6q23

    Apop

    tosismod

    ulator

    Methy

    latio

    n-sp

    ecificPCR

    2/58

    (3%)

    27/35(77%

    )—

    —33

    /40(83%

    )(171

    )A-kinasean

    choring

    protein

    12(or

    Gravinor

    AKAP12

    )

    6q24

    -25.2

    Cell-sign

    aling,

    adhe

    sion

    ,mito

    gene

    sis,

    and

    differen

    tiatio

    n

    Methy

    latio

    n-sp

    ecificPCR

    0/66

    (0%)

    29/60(48.3%

    )10

    /19(52.6%

    )11

    /21(52.4%

    )35

    /67(52.2)

    (172

    )

    Vim

    entin

    10p13

    Cytos

    keletonprotein

    Methy

    latio

    n-sp

    ecificPCR

    0/9(0%

    )10

    /11(91%

    )—

    5/5(100

    %)

    21/26(81%

    )(173

    )RUNX3

    1p36

    Tran

    scrip

    tionfactor

    Methy

    latio

    n-sp

    ecificPCR

    1/63

    (2%)

    23/93(25%

    )—

    —37

    /77(48%

    )(149

    )HPP1

    19pter-p13

    .1Tu

    mor

    suppress

    or2/64

    (3%)

    41/93(44%

    )—

    —55

    /77(71%

    )3-OST-2

    16p12

    Sulfotran

    sferas

    een

    zyme

    1/57

    (2%)

    47/60(78%

    )—

    —28

    /73(38%

    )

    Wnt

    inhibitory

    factor-1

    (WIF-1)

    12q14

    .3Wnt

    antago

    nist

    Methy

    latio

    n-sp

    ecificPCR

    81%

    ofpa

    tientswith

    Barrett's

    esop

    hagu

    ssu

    fferingfrom

    EAC

    show

    edhy

    permethy

    latedWIF-1

    asco

    mpared

    with

    20%

    ofpa

    tientswith

    Barrett's

    esop

    hagu

    swith

    outEAC

    (174

    )

    CHFR

    (che

    ckpoint

    with

    forkhe

    adasso

    ciated

    and

    ringfing

    er)

    12q24

    Mito

    sisch

    eckpoint

    protein

    Bisulfite

    pyros

    eque

    ncing

    EAC

    samples31

    %(18/58

    )sho

    wed

    sign

    ifica

    ntly

    high

    erCHFR

    promoter

    methy

    latio

    nas

    compared

    with

    norm

    alsa

    mples(P

    ¼0.01

    ).(175

    )

    Metallothione

    in3

    (orMT3

    )16

    q13

    Metal

    homeo

    stas

    isan

    dprotec

    tion

    agains

    tDNAda

    mag

    e

    Bisulfite

    pyros

    eque

    ncing

    (sam

    ple

    size

    :normal-33,

    BE-5,E

    AC-78)

    Iden

    tified

    2region

    s(R2an

    dR3)

    ofCpG

    nucleo

    tides

    ,which

    show

    edsign

    ifica

    ntly

    high

    ermethy

    latio

    nin

    EACas

    compared

    with

    norm

    alep

    ithelium

    (FDR<0.00

    1).

    Increa

    sedDNAmethy

    latio

    nof

    MT3

    promoter

    R2co

    rrelates

    with

    adva

    nced

    tumor

    stag

    e(P

    ¼0.00

    5)an

    dlymphno

    demetas

    tasis(P

    ¼0.03

    ).DNAmethy

    latio

    nof

    MT3

    promoter

    R3co

    rrelates

    with

    tumor

    stag

    ing(P

    ¼0.03

    )but

    notwith

    lymph

    nodestatus

    (P¼

    0.4).

    (176

    )

    Methy

    lationmarke

    rpan

    el

    Sam

    ple

    size

    Metho

    dFind

    ings

    Ref.

    EAC-35un

    dergo

    ing

    chem

    orad

    iotherap

    yMethy

    latio

    n-sp

    ecificPCR

    Com

    bine

    dmea

    nof

    promoter

    methy

    latio

    nof

    p16

    ,Rep

    rimo,

    p57

    ,p73

    ,RUNX-3,

    CHFR

    ,MGMT,

    TIMP-3,a

    ndHPP1was

    lower

    inpatientswho

    resp

    onded

    toch

    emorad

    iotherap

    y(13/35

    )asco

    mpared

    with

    patientswho

    did

    notresp

    ond

    (22/35

    ;P¼

    0.00

    3).

    (46)

    BE-62(28patientswith

    Barrett's

    esop

    hagu

    sprogres

    sedto

    EAC

    andremaining

    34pa

    tientswith

    Barrett's

    esop

    hagu

    swere

    nonp

    rogres

    sors)

    Methy

    latio

    n-sp

    ecificPCR

    Three-tie

    redstratifi

    catio

    nmod

    elwas

    dev

    elop

    edus

    ingmethy

    latio

    nindex

    (p16

    ,HPP1,

    andRUNX3),B

    arrett's

    esop

    hagu

    sleng

    than

    dpatho

    logy

    .Com

    bine

    dmod

    elbas

    edon

    2-(ROC:0

    .838

    6)an

    d4-ye

    ar(ROC:0

    .791

    0)predictionwas

    able

    toca

    tego

    rizepatientswith

    Barrett'ses

    opha

    gusinto

    low-risk,

    interm

    ediate-risk,

    andhigh

    -riskgrou

    psforEAC

    dev

    elop

    men

    t.

    (44)

    (Con

    tinue

    don

    thefollo

    wingpag

    e)

    Shah et al.

    Cancer Epidemiol Biomarkers Prev; 22(7) July 2013 Cancer Epidemiology, Biomarkers & Prevention1190

    on June 7, 2021. © 2013 American Association for Cancer Research. cebp.aacrjournals.org Downloaded from

    Published OnlineFirst April 10, 2013; DOI: 10.1158/1055-9965.EPI-12-1415

    http://cebp.aacrjournals.org/

  • Tab

    le2.

    Sum

    maryof

    hypermethy

    latedge

    nesduringBE/EAC

    dev

    elop

    men

    t(Con

    t'd)

    Num

    ber

    (%)ofsa

    mplessh

    owinghy

    permethy

    lationorstud

    yfind

    ings

    Gen

    eLo

    cation

    Func

    tion

    Metho

    dNorm

    alBE

    LGD

    HGD

    EAC

    Ref.

    BE-195

    (145

    patientswith

    Barrett's

    esop

    hagu

    sprog

    ressed

    toEAC

    andremaining

    50patientswith

    Barrett's

    esop

    hagu

    swere

    nonp

    rogres

    sors)

    Methy

    latio

    n-sp

    ecificPCR

    HPP1(P

    ¼0.00

    25),p16

    (P¼

    0.00

    66),an

    dRUNX3(P

    ¼0.00

    02)w

    eresign

    ifica

    ntly

    hypermethy

    latedin

    progres

    sors

    asco

    mpared

    with

    nonp

    rogres

    sors.In

    combination,

    pan

    elof

    8methy

    latio

    nmarke

    rs(p16

    ,HPP1,

    RUNX3,

    CDH13

    ,TA

    C1,

    NELL

    1,AKAP12

    ,and

    SST)

    show

    edse

    nsitivitie

    sof

    0.44

    3an

    d0.62

    9at

    spec

    ificity

    of0.9an

    d0.8forEAC

    progres

    sion

    inpatientswith

    Barrett's

    esop

    hagu

    sus

    ingco

    mbined

    mod

    eldes

    igne

    don

    thebas

    isof

    2an

    d4ye

    arsof

    follo

    w-up.

    (43)

    EAC-41(adjace

    ntno

    rmal

    samples

    asco

    ntrol)

    Methy

    latio

    n-sp

    ecificPCR

    Patientsha

    ving

    morethan

    50%

    oftheirge

    nesmethy

    lated(APC,E

    -cad

    herin

    ,MGMT,

    ER,p

    16,D

    AP-kinase,

    andTIMP3)

    show

    edsign

    ifica

    ntly

    poo

    r2-ye

    arsu

    rvival(P

    ¼0.04

    )and

    2-ye

    arrelapse

    -freesu

    rvival(P

    ¼0.03

    )asco

    mpared

    with

    thepa

    tientsha

    ving

    less

    than

    50%

    methy

    latio

    n.

    (45)

    BE-18,

    EAC-38(m

    ultip

    lebiopsies

    weretake

    nan

    dclas

    sified

    into

    norm

    al,B

    arrett's

    esop

    hagu

    s,HGD,a

    ndEAC)

    Bisulfite-m

    odified

    DNAwith

    PCR

    Themethy

    latio

    nfreq

    uenc

    iesof

    9ge

    nes(APC,C

    DKN2A

    ,ID4,

    MGMT ,

    RBP1,

    RUNX3,

    SFR

    P1,

    TIMP3,

    andTM

    EFF

    2)foun

    dto

    be95

    %,5

    9%,7

    6%,5

    7%,7

    0%,

    73%

    ,95%

    ,74%

    ,and

    83%

    ,res

    pec

    tively,inEACsa

    mples,whe

    reas

    95%

    ,28%

    ,78

    %,4

    8%,5

    8%,4

    8%,9

    3%,8

    8%,a

    nd75

    %,res

    pec

    tively,

    inBarrett's

    esop

    hagu

    ssa

    mples,

    which

    was

    sign

    ifica

    ntly

    high

    eras

    compared

    with

    norm

    alsq

    uamou

    sep

    ithelium.T

    hemethy

    latio

    nfreq

    uenc

    yforC

    DKN2A

    andRUNX3was

    sign

    ifica

    ntly

    high

    erforEAC

    asco

    mpared

    with

    Barrett's

    esop

    hagu

    sbiopsy

    samples

    .

    (177

    )

    Normal-30,

    BE-29,

    HGD-8,E

    AC-29

    Illum

    inaGolden

    Gatemethy

    latio

    nbea

    darray

    Ove

    rallmed

    ianmethy

    latio

    nat

    thetotal706

    numbersof

    mos

    tinformativeCpG

    sites

    grad

    ually

    increa

    sedfrom

    norm

    al-B

    E-H

    GD/EAC

    (P<0.00

    1).T

    heau

    thors

    differen

    tiatedbetwee

    nEACvs.normal,H

    GDvs.normal,B

    arrett'ses

    opha

    gusvs.

    norm

    al,E

    ACvs

    .Barrett's

    esop

    hagu

    s,an

    dHGDvs

    .Barrett's

    esop

    hagu

    sbas

    edon

    422,

    225,

    195,

    17,a

    nd3nu

    mbersof

    CpG

    sites,which

    issh

    owingdifferen

    tial

    methy

    latio

    nbetwee

    nresp

    ectiv

    egrou

    ps.

    (178

    )

    Iden

    tifica

    tionpha

    se(BE-22,

    EAC-

    24);retros

    pec

    tiveva

    lidation

    pha

    se(BE-60,

    LGD/H

    GD-36,

    EAC-90);p

    rosp

    ectiv

    eva

    lidation

    pha

    se(98pa

    tientsun

    der

    surveillanc

    e).

    Iden

    tifica

    tionpha

    se:IlluminaInfinium

    assa

    y;retros

    pec

    tive/prosp

    ectiv

    eva

    lidationpha

    se:

    pyros

    eque

    ncing

    Onthebas

    isof

    initial

    iden

    tifica

    tionpha

    se,7

    gene

    s(SLC

    22A18

    ,ATP

    2B4,

    PIGR,

    GJA

    12,R

    IN2,

    RGN,a

    ndTC

    EAL7

    )sho

    wingmos

    tprominen

    tmethy

    latio

    nch

    ange

    swerese

    lected

    forva

    lidation.

    Com

    binationof

    4ge

    nes(ROC

    0.98

    8)SLC

    22A18

    ,PIG

    R,G

    JA12

    ,and

    RIN2sh

    owed

    sens

    itivityof

    94%

    andsp

    ecificityof

    97%.T

    hispa

    nelo

    f4ge

    nessh

    owingdifferen

    tialm

    ethy

    latio

    n,stratifi

    edpatients

    into

    low-,interm

    ediate-,an

    dhigh

    -riskgrou

    psforEAC

    dev

    elop

    men

    tin

    prosp

    ectiv

    eva

    lidation.

    (179

    )

    Non

    dys

    plasticBarrett'ses

    opha

    gus

    (not

    progres

    sedto

    EAC)-16

    ,Barrett's

    esop

    hagu

    smuc

    osa

    from

    patientsprogres

    sedto

    EAC-12

    Methy

    latio

    n-se

    nsitive

    sing

    le-stran

    dco

    nformation

    analysis

    andmethy

    latio

    n-se

    nsitive

    dot

    blot

    assa

    yBarrett'ses

    opha

    gussa

    mplesco

    llected

    from

    patientswho

    prog

    ressed

    toEACin12

    mon

    thstim

    epe

    riodsh

    owed

    100%

    ,91%

    ,and

    92%

    hypermethy

    latio

    nof

    APC,

    TIMP-3,a

    ndTE

    RT,

    resp

    ectiv

    ely,

    asco

    mpared

    with

    36%

    ,23%

    ,and

    17%

    inBarrett's

    esop

    hagu

    smuc

    osaco

    llected

    from

    patientswho

    did

    notprogres

    sto

    EAC.

    (180

    )

    Methy

    lationmarke

    rpan

    el

    Sam

    ple

    size

    Metho

    dFind

    ings

    Ref.

    Biomarkers for Esophageal Adenocarcinoma

    www.aacrjournals.org Cancer Epidemiol Biomarkers Prev; 22(7) July 2013 1191

    on June 7, 2021. © 2013 American Association for Cancer Research. cebp.aacrjournals.org Downloaded from

    Published OnlineFirst April 10, 2013; DOI: 10.1158/1055-9965.EPI-12-1415

    http://cebp.aacrjournals.org/

  • Tab

    le3.

    Sum

    maryof

    gene

    expressionprofilingstud

    iesforBE/EAC

    Sam

    ple

    size

    Array

    des

    criptio

    nOutco

    me

    Find

    ings

    Externa

    lvalidation

    Ref.

    BE-21(pairedno

    rmal

    esop

    hage

    alan

    dga

    stric

    samplesas

    control)

    Seriala

    nalysisof

    gene

    express

    ion,

    PCRan

    dim

    mun

    oblotting

    Disea

    seprog

    ression

    Ofno

    te,5

    34tags

    weresign

    ifica

    ntly

    differen

    tially

    expressed

    betwee

    nno

    rmales

    opha

    gealsq

    uamou

    sep

    ithelium

    andBarrett's

    esop

    hagu

    s.Th

    emos

    tup

    regu

    latedge

    nesin

    Barrett's

    esop

    hagu

    sas

    compared

    with

    norm

    alep

    ithelium

    wereiden

    tified

    tobetrefoilfac

    tors,a

    nnex

    inA10

    andga

    lectin-4

    with

    each

    differen

    ttypeof

    tissu

    esh

    owed

    anun

    ique

    cytoke

    ratin

    expres

    sion

    .

    No

    (181

    )

    Barrett's

    esop

    hagu

    san

    dHGD-11

    (match

    edbiopsy

    samples)

    cDNAmicroarray

    Disea

    seprog

    ression

    Using

    2.5-fold

    cutoff,iden

    tified

    131up

    regu

    latedan

    d16

    dow

    nreg

    ulated

    gene

    sinHGD.Twen

    ty-fou

    rof28

    mos

    tsign

    ifica

    ntly

    differen

    tge

    nessh

    owed

    similar

    chan

    gesduringva

    lidation.

    Rea

    l-tim

    ePCR

    (182

    )

    EAC-91

    Oligo-microarray

    Disea

    seprog

    ression

    A4-ge

    nepan

    elco

    nsists

    ofdeo

    xycy

    tidinekina

    se,3

    0 -pho

    spho

    aden

    osine50-pho

    spho

    sulfa

    tesy

    ntha

    se2,

    sirtuin-2,

    andtripartitemotif-co

    ntaining

    44predicted

    5-ye

    arsu

    rvival.

    Immun

    ohistoch

    emistry

    (183

    )

    Twen

    ty-three

    pairedBarrett's

    esop

    hagu

    san

    dno

    rmal

    epith

    elium

    samples

    Tran

    scrip

    tiona

    lprofiling

    andproteo

    mics

    Disea

    seprog

    ression

    Iden

    tified

    2,82

    2ge

    nesto

    bedifferen

    tially

    expressed

    betwee

    nBarrett's

    esop

    hagu

    san

    dno

    rmal

    epith

    elium.S

    ignifica

    ntly

    overex

    press

    edge

    nes

    duringBarrett's

    esop

    hagu

    sbe

    long

    edto

    cytokine

    san

    dgrow

    thfactors,

    cons

    titue

    ntsof

    extrac

    ellular

    matrix

    ,bas

    emen

    tmem

    brane

    andtig

    htjunc

    tions

    ,proteinsinvo

    lved

    inprostag

    land

    inan

    dpho

    spho

    inos

    itolm

    etab

    olism,n

    itric

    oxide

    produc

    tionan

    dbioen

    erge

    tics.

    While

    gene

    sen

    codingHSPan

    dva

    rious

    kina

    seswere

    dow

    nreg

    ulated

    .

    No

    (184

    )

    Lymphno

    demetas

    tatic

    (n¼

    55)

    andno

    nmetas

    tatic

    (n¼

    22)E

    AC

    samples

    Oligo-microarray

    Disea

    seprog

    ression

    Lymphno

    de–

    pos

    itive

    samplessh

    owed

    sign

    ifica

    ntdow

    nreg

    ulationof

    arginino

    succ

    inatesynthe

    tase

    asco

    mpared

    with

    lymphno

    deno

    nmetas

    tatic

    samples(P

    ¼0.04

    8).

    No

    (185

    )

    EAC-6

    andga

    stric

    cardiaca

    ncer-8

    aCGH

    Disea

    seprog

    ression

    Iden

    tified

    HGF(45%

    )and

    BCAS1(27%

    )tobemos

    tfreq

    uently

    overex

    pressed

    gene

    sresp

    ectiv

    elyat

    7q21

    and20

    q13

    locu

    s.

    No

    (186

    )

    Eleve

    nmatch

    edsa

    mplese

    ts(hea

    lthy-BE-EAC

    match

    ed-6,

    norm

    al-B

    Ematch

    ed-4

    and

    norm

    al-EAC

    match

    ed-1)

    SNPmicroarray

    Disea

    seprog

    ression

    60%

    ofBarrett's

    esop

    hagu

    san

    d57

    %of

    EAC

    samplesco

    ntaine

    dat

    leas

    ton

    eof

    thege

    nomic

    alteratio

    nsin

    theform

    ofdeletions

    ,dup

    lications

    ,am

    plifica

    tions

    ,cop

    ynu

    mber

    chan

    ges,

    andne

    utral

    LOH.

    No

    (187

    )

    (Con

    tinue

    don

    thefollo

    wingpag

    e)

    Shah et al.

    Cancer Epidemiol Biomarkers Prev; 22(7) July 2013 Cancer Epidemiology, Biomarkers & Prevention1192

    on June 7, 2021. © 2013 American Association for Cancer Research. cebp.aacrjournals.org Downloaded from

    Published OnlineFirst April 10, 2013; DOI: 10.1158/1055-9965.EPI-12-1415

    http://cebp.aacrjournals.org/

  • Tab

    le3.

    Sum

    maryof

    gene

    expressionprofilingstud

    iesforBE/EAC

    (Con

    t'd)

    Sam

    ple

    size

    Array

    des

    cription

    Outco

    me

    Find

    ings

    Externa

    lvalidation

    Ref.

    Normal-39,

    BE-25,

    EAC-38,

    and

    ESCC-26

    cDNAmicroarray

    Disea

    seprogres

    sion

    Clusteringsh

    owed

    these

    parationof

    samples

    into

    4distinct

    grou

    ps.

    Ofno

    te,2

    ,158

    clon

    eswere

    differen

    tially

    expressed

    betwee

    nno

    rmal

    and

    Barrett's

    esop

    hagu

    ssa

    mples,

    whe

    reas

    1,30

    6be

    twee

    nBarrett's

    esop

    hagu

    san

    dEAC.B

    E/EAC

    samplessh

    owed

    differen

    tiale

    xpressionof

    hydrolase

    s,lyso

    zyme,

    fuco

    sidas

    e,tran

    scrip

    tion

    factors,

    muc

    ins,

    andthetrefoilfac

    tors.

    No

    (188

    )

    BE-20,

    LGD-19,

    HGD-20an

    dEAC-42

    SNPmicroarray

    Disea

    seprogres

    sion

    Increa

    sing

    numbersof

    SNPsan

    dloss

    ofch

    romos

    omes

    with

    disea

    seprogres

    sion

    .Chrom

    osom

    aldisruptio

    nwas

    iden

    tified

    intheFH

    IT,

    WWOX,R

    UNX1,

    KIF26

    B,M

    GC48

    628,

    PDE4D

    ,C20

    orf133

    ,GMDS,D

    MD,a

    ndPARK2ge

    nesin

    EAC.

    No

    (189

    )

    EAC-75sp

    ecim

    ensfrom

    64pa

    tients,ad

    jace

    ntpairedno

    rmal

    tissu

    efrom

    patientswith

    EAC-

    28

    DNAmicroarray

    Disea

    seprogres

    sion

    Iden

    tified

    AKR1B

    10,C

    D93

    ,CSPG2,

    DKK3,

    LUM,

    MMP1,

    SOX21

    ,SPP1,

    SPARC,a

    ndTW

    IST1

    gene

    sas

    biomarke

    rbas

    edon

    tran

    scrip

    tomicsdata.

    Qua

    ntita

    tivereal-tim

    ePCRiden

    tified

    SPARC

    and

    SPP1ge

    nesto

    beas

    sociated

    with

    EAC

    patient

    survival

    (P<0.02

    4).

    Rea

    l-tim

    ePCR

    (190

    )

    EAC-8,g

    astric

    cardia

    canc

    er-3

    aCGH

    andcD

    NA

    microarray

    Disea

    seprogres

    sion

    Tran

    scrip

    tomicsdataiden

    tified

    11ge

    nesto

    be

    differen

    tially

    expressed

    (ELF

    3,SLC

    45A3,CLD

    N12

    ,CDK6,

    SMURF1

    ,ARPC1B

    ,ZKSCAN1,

    MCM7,

    COPS6,

    FDFT

    1 ,an

    dCTS

    B).IHCan

    alys

    isreve

    aled

    sign

    ifica

    ntov

    erex

    pressionof

    CDK6ace

    ll-cy

    cle

    regu

    latorin

    tumor

    samples.

    No

    (191

    )

    BE-20

    aCGH

    arrays

    andhigh

    den

    sity

    SNP

    geno

    typing

    Disea

    seprogres

    sion

    Cop

    ynu

    mber

    loss

    esweredetec

    tedat

    FRA3B

    (81%

    ),FR

    A9A

    /C(71.4%

    ),FR

    A5E

    (52.4%

    ),an

    dFR

    A4D

    (52.4%

    )site

    sin

    early

    Barrett's

    esop

    hagu

    s.Validationstud

    yco

    nfirm

    edloss

    ofFR

    A3B

    and

    FRA16

    Din

    early

    Barrett's

    esop

    hagu

    ssa

    mples.

    Rea

    l-tim

    ePCRan

    dpyros

    eque

    ncing

    (192

    )

    BE-11,

    gastroes

    opha

    geal

    junc

    tion

    (GEJ)

    aden

    ocarcino

    ma-11

    aCGH

    with

    awho

    lech

    romos

    ome8q

    contig

    array

    Disea

    seprogres

    sion

    Ove

    rexp

    ress

    ionof

    MYC

    andEXT1

    ,while

    downreg

    ulationof

    MTS

    S1,

    FAM84

    B,a

    ndC8o

    rf17

    issign

    ifica

    ntly

    asso

    ciated

    with

    GEJ

    aden

    ocarcino

    ma.

    (193

    )

    BE-14,

    EAC-5,E

    SCC-3

    cDNAmicroarray

    Disea

    seprogres

    sion

    Iden

    tified

    160ge

    nesthat

    candifferen

    tiate

    betwee

    nBarrett's

    esop

    hagu

    san

    des

    opha

    geal

    canc

    er.

    No

    (194

    )

    Twen

    ty-fou

    rpa

    iredsa

    mplesof

    norm

    al,B

    arrett's

    esop

    hagu

    s,an

    dEAC

    phen

    otyp

    e

    cDNAmicroarray

    Disea

    seprogres

    sion

    Ofno

    te,2

    14differen

    tially

    regu

    latedge

    nesco

    uld

    differen

    tiate

    betwee

    nno

    rmal,B

    arrett'ses

    opha

    gus,

    andEACphe

    notype.

    Gen

    esinvo

    lved

    inep

    idermal

    No

    (195

    )

    (Con

    tinue

    don

    thefollo

    wingpag

    e)

    Biomarkers for Esophageal Adenocarcinoma

    www.aacrjournals.org Cancer Epidemiol Biomarkers Prev; 22(7) July 2013 1193

    on June 7, 2021. © 2013 American Association for Cancer Research. cebp.aacrjournals.org Downloaded from

    Published OnlineFirst April 10, 2013; DOI: 10.1158/1055-9965.EPI-12-1415

    http://cebp.aacrjournals.org/

  • Tab

    le3.

    Sum

    maryof

    gene

    expressionprofilingstud

    iesforBE/EAC

    (Con

    t'd)

    Sam

    ple

    size

    Array

    des

    criptio

    nOutco

    me

    Find

    ings

    Externa

    lvalidation

    Ref.

    differen

    tiatio

    nareun

    derex

    pressed

    inEAC

    asco

    mpared

    with

    Barrett's

    esop

    hagu

    s.Exp

    ress

    ion

    ratio

    ofGATA

    6to

    SPRR3ca

    ndifferen

    tiate

    betwee

    n3ph

    enotyp

    esstud

    ied.

    Poo

    ledbiosp

    ysa

    mplesfrom

    Barrett's

    esop

    hagu

    s,es

    opha

    geal

    squa

    mou

    s,ga

    stric

    ,an

    dduo

    den

    um

    Oligo-microarray

    Disea

    seprog

    ression

    Differen

    tiate

    differen

    ttis

    sueclus

    ters

    bas

    edon

    gene

    expressionprofile.Iden

    tified

    38ge

    nesthat

    are

    upregu

    latedin

    Barrett's

    esop

    hagu

    stis

    sueclus

    ter,

    which

    belong

    toce

    llcy

    cle(P1c

    dc4

    7,PCM-1),ce

    llmigratio

    n(urokina

    se-typ

    eplasm

    inog

    enrece

    ptor,

    LUCA-1/H

    YAL1

    ),grow

    thregu

    latio

    n(TGF-b

    superfamily

    protein,a

    mphiregu

    lin,C

    yr61

    ),stress

    resp

    onse

    s(calcy

    clin,A

    TF3,

    TR3orpha

    nrece

    ptor),

    epith

    elialc

    ells

    urface

    antig

    ens(epsilon-BP,E

    SA,

    integrin

    b4,m

    esothe

    linCAK-1

    antig

    enprecu

    rsor),

    and4muc

    ins.

    No

    (196

    )

    Normal-24,

    BE-18,

    EAC-9

    cDNAmicroarray

    Disea

    seprog

    ression

    Iden

    tified

    457,

    295,

    and36

    differen

    tially

    expres

    sed

    gene

    s,resp

    ectiv

    ely,betwee

    nno

    rmal-EAC,normal-

    Barrett's

    esop

    hagu

    s,an

    dBE–EAC

    grou

    ps.

    No

    (197

    )

    89-EAC

    cDNA-m

    ediated

    anne

    aling,

    selection,

    extens

    ion,

    andlig

    ation

    assa

    ywith

    502kn

    own

    canc

    er-related

    gene

    s

    Disea

    seprog

    ression

    Iden

    tified

    differen

    tialg

    eneex

    pressionbe

    twee

    nea

    rlystag

    esof

    EAC(T1an

    dT2

    )vs.late

    (T3an

    dT4

    ).Gen

    eex

    pressionprofile

    reve

    aled

    ERBB4,

    ETV

    1,TN

    FSF6

    ,MPLge

    nesto

    beco

    mmon

    betwee

    nad

    vanc

    edtumor

    stag

    ean

    dlymphno

    demetas

    tasis.

    No

    (198

    )

    Normal

    esop

    hage

    almuc

    osa-9,

    esop

    hagitis

    -6,B

    E-10,

    EAC-5,

    GEJad

    enoc

    arcino

    ma-9,

    stom

    achsa

    mples-32

    (normal

    muc

    osa-11

    ,IM-9,intes

    tinal-

    typead

    enoc

    arcino

    ma-7,

    and

    diffus

    eca

    rcinom

    a-5)

    cDNAmicroarray

    Disea

    seprog

    ression

    Onthebas

    isof

    theex

    pressionprofile,g

    enes

    asso

    ciated

    with

    thelip

    idmetab

    olism

    andcy

    tokine

    nodulearefoun

    dto

    besign

    ifica

    ntly

    altered

    betwee

    nEAC

    andothe

    rgrou

    ps.

    No

    (199

    )

    Sev

    enteen

    pairedsa

    mples

    ofno

    rmal,B

    E/EAC

    cDNAmicroarray

    Disea

    seprog

    ression

    Eac

    htis

    suetype

    expresses

    distin

    ctse

    tof

    gene

    s,which

    candifferen

    tiate

    betwee

    ntheirph

    enotyp

    es.

    Barrett'ses

    opha

    gusan

    dEACex

    presses

    similarset

    ofstromal

    gene

    sthat

    aredifferen

    tfrom

    norm

    alep

    ithelium.

    No

    (200

    )

    BE-19,

    EAC-20(98tis

    sue

    spec

    imen

    swereco

    llected

    and

    catego

    rized

    into

    differen

    tgrou

    ps)

    Onthebas

    isof

    previou

    smicroarraystud

    ies23

    gene

    swereva

    lidated

    usingreal-tim

    ePCR

    Disea

    seprog

    ression

    Out

    of23

    gene

    s,pa

    nelo

    f3ge

    nes(BFT

    ,TSPAN,a

    ndTP

    )was

    able

    todiscrim

    inatebetwee

    nBarrett's

    esop

    hagu

    san

    dEACin

    internal

    valid

    ationwith

    0%clas

    sifica

    tionerror.

    N.A.

    (201

    )

    (Con

    tinue

    don

    thefollo

    wingpag

    e)

    Shah et al.

    Cancer Epidemiol Biomarkers Prev; 22(7) July 2013 Cancer Epidemiology, Biomarkers & Prevention1194

    on June 7, 2021. © 2013 American Association for Cancer Research. cebp.aacrjournals.org Downloaded from

    Published OnlineFirst April 10, 2013; DOI: 10.1158/1055-9965.EPI-12-1415

    http://cebp.aacrjournals.org/

  • Tab

    le3.

    Sum

    maryof

    gene

    expressionprofilingstud

    iesforBE/EAC

    (Con

    t'd)

    Sam

    ple

    size

    Array

    des

    cription

    Outco

    me

    Find

    ings

    Externa

    lvalidation

    Ref.

    Normal-30,

    BE-31,

    gastric

    muc

    osa-34

    ,duo

    den

    um-18

    Biomarke

    rsforBarrett's

    esop

    hagu

    swere

    iden

    tified

    using3

    pub

    licly

    available

    microarraydatas

    ets

    andva

    lidated

    using

    real-tim

    ePCRan

    dim

    mun

    ohistoch

    emistry.

    Disea

    seprogres

    sion

    Out

    of14

    gene

    siden

    tified

    ,dop

    ade

    carbox

    ylas

    e(DDC)

    andTrefoilfac

    tor3(TFF

    3)wereva

    lidated

    tobe

    upregu

    latedin

    Barrett's

    esop

    hagu

    s.

    N.A.

    (202

    )

    EAC-56

    Olig

    onuc

    leotide

    microarrayan

    daC

    GH

    Disea

    seprogres

    sion

    Iden

    tified

    4ne

    wge

    nes(EGFR

    ,WT1

    ,NEIL2,

    and

    MTM

    R9)to

    beov

    erex

    pressed

    in10

    %to

    25%

    EAC.

    Exp

    ress

    ionleve

    lsof

    thes

    e4ge

    nesdifferen

    tiated

    patie

    ntswith

    EAC

    into

    3grou

    psna

    melygo

    od,

    averag

    e,an

    dpoo

    rdep

    endingup

    ontheirp

    rogn

    osis

    (P<0.00

    8)

    Immun

    ohistoch

    emistry

    (203

    )

    BE/LGD-72,

    HGD-11,

    EAC-15

    Bac

    teria

    lartificial

    chromos

    omeaC

    GH

    Disea

    seprogres

    sion

    Cop

    ynu

    mber

    chan

    gesweremoreco

    mmon

    and

    larger

    asdisea

    seprogres

    sto

    laters

    tage

    s.Patients

    having

    copynu

    mber

    alteratio

    nsinvo

    lvingmore

    than

    70Mbpwereat

    increa

    sedris

    kof

    progres

    sion

    toEAC

    (P¼

    0.00

    47)

    No

    (60)

    EAC-30,

    BE-6,L

    GD-9,H

    GD-10

    Gen

    ome-wideCGH

    Disea

    seprogres

    sion

    Loss

    of7q

    33-q35

    was

    foun

    din

    HGDas

    compared

    with

    precu

    rsor

    LGD(P

    ¼0.01

    ).Lo

    ssof

    16q21

    -q22

    andga

    inof

    20q1

    1.2-q1

    3.1was

    sign

    ifica

    ntly

    differen

    tbetwee

    nHGDan

    dEAC

    (P¼

    0.02

    and

    0.03

    ,res

    pec

    tively).

    No

    (56)

    EAC-30,

    lymphno

    demetas

    tasis-

    8,HGD-11,

    LGD-8,a

    ndBE-6

    from

    30EAC

    patient

    biopsy

    samples

    CGH

    Disea

    seprogres

    sion

    Iden

    tified

    region

    sun

    dergo

    ingco

    pynu

    mber

    loss

    and

    amplifica

    tionduringea

    chstag

    eof

    tran

    sitio

    n.Ave

    rage

    number

    ofch

    romos

    omal

    imba

    lanc

    ese

    que

    ntially

    increa

    sedfrom

    BE–LG

    D–HGD–EAC–

    lymphno

    demetas

    tasis.

    No

    (54)

    Forty-tw

    opatientsreprese

    ntdifferen

    tstag

    esof

    disea

    seSNParray

    Disea

    seprogres

    sion

    SNPab

    norm

    alities

    increa

    sesfrom

    2%to

    morethan

    30%

    asthedise

    aseprog

    ress

    from

    Barrett's

    esop

    hagu

    sto

    EAC.T

    otalnu

    mbe

    rofS

    NPalteratio

    nsin

    tissu

    esa

    mples

    istig

    htlyco

    rrelated

    with

    DNA

    abno

    rmalities

    such

    asan

    euploidy

    andLO

    H.

    No

    (57)

    EAC-27an

    dmatch

    edno

    rmal-14

    SNParray

    Disea

    seprogres

    sion

    Con

    firm

    edprev

    ious

    lydes

    cribed

    geno

    mic

    alteratio

    nssu

    chas

    amplifica

    tionon

    8qan

    d20

    q13

    ordeletion/

    LOH

    on3p

    and9p

    .Alsoiden

    tified

    alteratio

    nsin

    seve

    raln

    ovel

    gene

    san

    dDNAregion

    sin

    EAC

    samples

    .

    No

    (58)

    (Con

    tinue

    don

    thefollo

    wingpag

    e)

    Biomarkers for Esophageal Adenocarcinoma

    www.aacrjournals.org Cancer Epidemiol Biomarkers Prev; 22(7) July 2013 1195

    on June 7, 2021. © 2013 American Association for Cancer Research. cebp.aacrjournals.org Downloaded from

    Published OnlineFirst April 10, 2013; DOI: 10.1158/1055-9965.EPI-12-1415

    http://cebp.aacrjournals.org/

  • Tab

    le3.

    Sum

    maryof

    gene

    expressionprofilingstud

    iesforBE/EAC

    (Con

    t'd)

    Sam

    ple

    size

    Array

    des

    cription

    Outco

    me

    Find

    ings

    Externa

    lvalidation

    Ref.

    EAC-26

    SNParray

    Disea

    seprogres

    sion

    Con

    firm

    edpreviou

    slyreportedfreq

    uent

    chan

    gesto

    FHIT,C

    DKN2A

    ,TP53

    ,and

    MYC

    gene

    sin

    EAC.

    Iden

    tified

    PDE4D

    andMGC48

    628as

    tumor-

    suppress

    orge

    nes.

    No

    (59)

    EAC-35

    cDNAmicroarray

    Res

    pon

    seto

    chem

    othe

    rapy

    Iden

    tified

    165differen

    tially

    expres

    sedge

    nesbetwee

    npo

    or(n

    ¼17

    )and

    good

    outcom

    e(n

    ¼18

    )patient

    grou

    ps.

    Topfunc

    tiona

    lpathw

    aybas

    edon

    differen

    tialg

    eneex

    pressionwas

    iden

    tified

    tobe

    Toll-rece

    ptorsign

    aling.

    No

    (204

    )

    EAC-47(lo

    cally

    adva

    nced

    tumor)

    cDNAmicroarray

    Res

    pon

    seto

    chem

    othe

    rapy

    Iden

    tified

    86ge

    nessh

    owingat

    leas

    t2-folddifferen

    cebe

    twee

    nch

    emothe

    rapy

    resp

    onders(n

    ¼28

    )and

    nonres

    pon

    ders(n

    ¼19

    ).EphrinB3rece

    ptor,w

    hich

    show

    edhigh

    estdifferen

    cebetwee

    nthegrou

    ps,

    show

    edstrong

    mem

    brane

    staining

    inch

    emothe

    rapyresp

    ondingtumorsus

    ing

    immun

    ohistoch

    emistry.

    No

    (205

    )

    Patientswith

    EAC-19un

    dergo

    ing

    chem

    orad

    iotherap

    yOlig

    o-microarray

    Res

    pon

    seto

    chem

    orad

    iotherap

    yRed

    uced

    expressionof

    IVL,

    CRNN,N

    ICE-1,S

    100A

    2,an

    dSPPR3ge

    nesco

    rrelated

    with

    poo

    rsurviva

    land

    nonres

    pon

    seto

    chem

    othe

    rapy.

    No

    (206

    )

    19patients(EAC-16,

    ESCC-2

    and

    aden

    osqua

    mou

    sca

    rcinom

    a-1)

    undergo

    ingch

    emorad

    iotherap

    y

    Olig

    o-microarray

    Res

    pon

    seto

    chem

    orad

    iotherap

    yLo

    wer

    expressionforp

    anelof

    gene

    sPERP,S

    100A

    2,an

    dSPRR3was

    asso

    ciated

    with

    nonres

    pon

    seto

    therap

    y.Pathw

    ayan

    alysis

    iden

    tified

    downreg

    ulationof

    apop

    tosisin

    nonres

    pon

    ders.

    No

    (207

    )

    EAC-174

    ,ESCC-36

    SNPsas

    sociated

    with

    the

    chem

    othe

    rapy

    drug

    actio

    npa

    thway

    Res

    pon

    seto

    chem

    orad

    iotherap

    yIden

    tified

    asso

    ciationbetwee

    nge

    netic

    polymorphism

    san

    dresp

    onse

    topreop

    erative

    chem

    othe

    rapy(fluo

    rourac

    ilan

    dplatinum

    compou

    nds)

    andradiotherap

    y.

    No

    (208

    )

    NOTE

    :Majority

    ofstud

    iesdes

    cribed

    inthis

    table

    includ

    eva

    lidationof

    resu

    ltsin

    thesa

    mepatient

    coho

    rtas

    used

    indisco

    very

    pha

    se.Onlystud

    iesthat

    includ

    edva

    lidationus

    ing

    indep

    enden

    tpatient

    coho

    rtarede

    scrib

    edas

    external

    valid

    ation.

    Shah et al.

    Cancer Epidemiol Biomarkers Prev; 22(7) July 2013 Cancer Epidemiology, Biomarkers & Prevention1196

    on June 7, 2021. © 2013 American Association for Cancer Research. cebp.aacrjournals.org Downloaded from

    Published OnlineFirst April 10, 2013; DOI: 10.1158/1055-9965.EPI-12-1415

    http://cebp.aacrjournals.org/

  • decade, several studies conducted using advanced geno-mic techniques such as array-comparative genomichybridization (aCGH) and SNP arrays confirmed previ-ously reported copy number alterations and identifiednovel genomic loci undergoing changes during process ofmetaplasia–dysplasia–carcinoma development (54–60). Ithas been shown that as the disease progresses from earlyto late stages, SNP abnormalities increase from approxi-mately 2% to 30% (54, 57). The total number of SNPalterations in tissue samples is tightly correlated withpreviously reported DNA abnormalities such as aneu-ploidy, copy number alterations, and LOH highlightingthe application of SNP-based genotyping to assess geno-mic abnormalities (54–60). Thus, SNP-based genotypingprovides an alternative way to assess genomic abnormal-ities during EAC pathogenesis.Studies on gene expression changes in EAC have been

    propelled by recent progress in genomic technologies,each identifying unique sets of gene expression profile,which can be used as a biomarker panel for diseasediagnosis, prognosis, or to predict response to therapy(Table 3).Moreover, determination of the gene expressionchanges has been extremely helpful to understanddetailed pathogenesis and will form basis for developingfuture therapies. However, future validation using inde-pendent sample cohorts will be necessary for themajorityof these potential biomarkers.Apart from genomic abnormalities associated with the

    disease progression, inheriting genetic factors are alsoimplicated for EAC development. Risk for BE/EAC andGERD is increased by 2- to 4-fold when a first-degreerelative is already affected by any of these conditions (61).Recently, a study conducted by The Esophageal Adeno-carcinoma Genetics Consortium and TheWellcome TrustCase Control Consortium identified link between SNPs atthe MHC locus and chromosome 16q24.1 with risk forBarrett’s esophagus (62). They also identified SNPs asso-ciated with body weight measures that were present withmore than expected frequency in Barrett’s esophagussamples supporting epidemiologic findings about obesityas a risk factor for Barrett’s esophagus and EAC (62). Wuand colleagues examined the relationship between pres-ence of risk genotypes and the onset of EAC. They iden-tified 10 SNPs associatedwith the age of EAConset. Genesassociated with 5 of 10 SNPs identified were known to beinvolved in apoptosis (63).Recently, published cancer genome–sequencing stud-

    ies have given deeper insights into the genomic abnor-malities associated with the EAC pathogenesis. The com-parative genomic analysis between EAC and ESCCreported by Agrawal and colleagues (64) confirmed pre-viously verywell-described association of p53 genemuta-tions with esophageal cancer development. The authorsalso conducted comparative genome-wide analysisbetween matched Barrett’s esophagus and EAC patienttissue samples and concluded that the majority of geno-mic changes occur early during EAC development, at thestageofBarrett’s esophagus (64). Similar conclusionswere

    made by next-generation sequencing of biopsy samplesobtained from the same patient at the stage of Barrett’sesophagus and EAC (65). The authors also identifiedARID1A as novel tumor-suppressor gene and around15% of patientswith EAC showed loss of ARID1Aproteinin tissue samples. In vitro studies suggested it to beassociated with cell growth, proliferation, and invasion(65). Very recently published high-resolution methylomeanalysis has provided first evidence for methylationchanges at genomic regions that encodenoncodingRNAs.The authors identified longnoncodingRNA,AFAP1-AS1,to be severely hypomethylated in Barrett’s esophagus andEAC tissue samples, silencing of which significantlyreduced aggressiveness of EAC cell lines OE33 andSKGT4 (66).

    Taken together, genomic abnormalities play key rolesduring each stage of transformation from normal squa-mous epithelium to EAC.

    Cancer-Related InflammationGastric and bile acid exposure in the esophageal epi-

    thelium leads to the development of chronic inflamma-tory conditions mainly driven by elevated levels of proin-flammatory cytokines. Chronic inflammatory responsesinduce cell survival and increase cell proliferation, henceplay key roles in the development of EAC (67, 68). Expres-sions of various inflammatory molecules such as COX-2,NF-kB, interleukin (IL)-6, IL-8, and matrix metalloprotei-nases (MMP) have been evaluated as prognostic biomar-kers for BE/EAC development.

    Exposure to gastric/bile acid and cytokines leads toincreased COX-2 expression (69). COX-2 is a rate-limitingenzyme that regulates synthesis of prostaglandins fromarachidonic acid. COX-2 directly increases cell prolifera-tion and promotes tumor invasion (69), andCOX-2–medi-ated increase in prostaglandin synthesis could result intumor growth and angiogenesis (70). COX-2 expressionhas been detected in disease-free esophageal tissue homo-genates using immunoblotting (69). In comparison withGERD, patients suffering from erosive reflux show slight-ly higher gene expressions of this enzyme in tissue sam-ples (71). Several studies have shown significantlyincreased COX-2 expression correlating with the diseaseprogression from Barrett’s esophagus to dysplasia andEAC (69, 72–75). Furthermore, expression levels of COX-2have been shown to have a prognostic value in EAC withhigher levels associated with poor survival and increasedchances of tumor relapse (76, 77).

    Another well-studied inflammatory biomarker NF-kBis activated in response to exposure with bile acid andelevated NF-kB expression levels are found during Bar-rett’s esophagus, dysplasia, and adenocarcinoma (78–80).Activated NF-kB translocates from cytoplasm to nucleusand upregulates transcription of the genes involvedin inflammatory processes. Moreover, nuclear NF-kBexpression has been shown to be correlated with thepatient response to chemoradiotherapy.All of thepatientswho showed complete response to chemoradiotherapy

    Biomarkers for Esophageal Adenocarcinoma

    www.aacrjournals.org Cancer Epidemiol Biomarkers Prev; 22(7) July 2013 1197

    on June 7, 2021. © 2013 American Association for Cancer Research. cebp.aacrjournals.org Downloaded from

    Published OnlineFirst April 10, 2013; DOI: 10.1158/1055-9965.EPI-12-1415

    http://cebp.aacrjournals.org/

  • had elevated NF-kB levels pretreatment and showed lackof active NF-kB posttreatment (81).

    In line with NF-kB and COX-2, expression of indi-vidual or combinations of proinflammatory cytokinesIL-1b, IL-6, IL-8, and TNF-a is significantly increased inBarrett’s esophagus and EAC as compared with squa-mous epithelium (82–84). IL-1b and IL-8 expressionlevels also correlate with the stage of EAC (79). Patientswho responded to neoadjuvant chemotherapy treat-ment showed significantly reduced expressions of IL-8 and IL-1b in postchemotherapy esophageal tissuesections (81). IL-6 is activated in response to reflux andthe IL-6/STAT3 antiapoptotic pathway may underliethe development of dysplasia and tumor (85). Serum IL-6 levels were reported to provide 87% sensitivity and92% specificity for EAC diagnosis in a recent retrospec-tive study (86). However, the study only comparedbetween healthy and EAC groups. It would be interest-ing to see how early it can diagnose EAC during theprocess of metaplasia–dysplasia. Combination of cyto-kines IFN-g , IL-1a, IL-8, IL-21, and IL-23 along withplatelet proteoglycan and miRNA-375 expression pro-filing has been shown to build an inflammatory riskmodel, which has clinical use to determine prognosis forpatients with EAC (67).

    MMPs are a family of proteolytic enzymes involved inthe degradation of extracellular matrix components.MMPs play a role in both inflammation and tumormetas-tasis. Immunohistochemical staining forMMP-1, MMP-2,MMP-7, andMMP-9 has been reported to be significantlyhigher in EAC as compared with healthy individuals (87,88). Higher level of MMP-1 expression has been associ-ated with the lymph node metastases and possibly poorpatient survival (89). Expression ofMMP-9 is shown to bean early event during the EAC transformation and itsexpression levels are correlated with the progression ofthe disease (90–92). Activity of MMPs is inhibited by afamily of proteins called tissue inhibitors of metallopro-teinases (TIMP). Specifically, TIMP-3 gene is methylatedin EAC development and its reduced expression is asso-ciated with stage of the tumor and patient survival (93).On contrary, Salmela and colleagues described elevatedTIMP-1 and TIMP-3 expression in EAC tumor samples(88).

    Although the underlying tissue inflammation is veryclosely associated with EAC development and severalinflammation-related biomarkers have been identified,these remain to be validated in large-scale biomarkerstudies.

    Cell Cycle–Related AbnormalitiesTo compensate for the tissue damage induced by gas-

    tric/bile acid, the underlying epithelium starts to prolif-erate rapidly and become uncontrolled resulting in neo-plasia. To meet the proliferation requirements, the cellshave to overcome cell-cycle checkpoints. Cyclin D1 over-expression is one such means by which cells overcomeG1–S checkpoint, and cyclin D1 immunohistochemical

    staining has been proposed to identify patients with Bar-rett’s esophagus with an increased risk for EAC (94). Incontrast to cyclin D1, expression of p16 protein results incell-cycle arrest in G1 phase as it has been shown to inhibitcyclin-dependent kinase–induced phosphorylation ofretinoblastoma protein. Early genomic abnormalities dur-ing EAC development significantly affect p16 proteinexpression,which can bedeterminedusing immunostain-ing and implemented as a potential biomarker (95). Fur-ther large-scale trials are required to confirm cell-cycleabnormalities during EAC development to implementthem as a biomarker.

    Bottom of the pyramid in Fig. 1 represents list ofbiomarkers in the initial stages of development. Tumorsharboring overexpression of growth factor receptors [EGFreceptor (EGFR) and HER-2] are associated with poorpatient survival (96, 97), whereas those overexpressingapoptosis regulator Bcl-2 protein showed prolonged sur-vival (98). Incipient angiogenesis is a marked feature ofBarrett’s esophagus and underlining tissue expressesangiogenesis markers VEGF and its receptors (99). Neo-vascularization continues as the disease progresses fromBarrett’s esophagus to EAC. Measuring the degree ofneovascularization correlated with histopathologic gradeof the tumor and associated with the patient survival(100). Expression of 2 prominent cell proliferation mar-kers, PCNA and Ki-67, has been described to be alteredduring BE–EAC development (101).

    miRNAmiRNA was first discovered in Caenorhabditis elegans

    (102) and since then it has beenwidely studied in a varietyof biologic phenomena. These short stretches of approx-imately 21 nucleotides do not code for protein but playimportant roles in gene regulation by either suppressingprotein synthesis or causing mRNA cleavage. UnlikesiRNA, miRNA can target multiple genes on remote lociand therefore control diverse group of proteins. Severalkey properties of carcinogenesis have been shown to beregulated via miRNA, for example, angiogenesis andmetastasis (103).

    With increased biologic understanding ofmiRNAs andtheir role in cancer, they have been proposed in severaldifferent clinical applications including cancer diagnosisand tumor prognosis, tumor classification, and also as atherapeutic target for disease intervention. Differentialtissue miRNA expression has been observed in severaldifferent malignancies and these changes can be used fordiagnosis and classification of the tumors (103). miRNAbioarrays were first used to show differential miRNAexpression in healthy, Barrett’s esophagus, and EACtissue samples (104). Since then, a number of differentstudies have identified miRNA changes associated withthe development of the BE–EAC. Table 4 summarizesprimary findings of miRNA expression profiling studiesalong with statistical significance and fold-change values.Biologic significance for some of the miRNA-relatedchanges is discussed later.

    Shah et al.

    Cancer Epidemiol Biomarkers Prev; 22(7) July 2013 Cancer Epidemiology, Biomarkers & Prevention1198

    on June 7, 2021. © 2013 American Association for Cancer Research. cebp.aacrjournals.org Downloaded from

    Published OnlineFirst April 10, 2013; DOI: 10.1158/1055-9965.EPI-12-1415

    http://cebp.aacrjournals.org/

  • Smith and colleagues identified reduced expression ofmiR-200 and miR-141 in Barrett’s esophagus and EACtissue samples. They conducted bioinformatics analysisand correlated these miRNA expression changes withcellular processes such as cell cycle, cell proliferation,apoptosis, and cell migration (105). miR-196a, which isdescribedas amarker of progression fromBarrett’s esoph-agus to EAC, can increase cell proliferation and anchor-age-independent growth and inhibit apoptosis in EACcell lines in vitro (106). The downstream targets for miR-196a are verified to be Annexin A1, S100 calcium-bindingprotein A9, small proline-rich protein 2C, and Keratin 5,which showed reduced expression in EAC patient tissuesamples as compared with normal epithelium (106, 107).Several studies described in Table 4 report overexpressionof miR-192 during EAC carcinogenesis. miR-192 has beenreported tobe a target of p53 andhas been able to suppresscancer progression in osteosarcoma and colon cancer celllines throughp21 accumulation and cell-cycle arrest (108).As shown in Table 4,miR-21 is overexpressed during BE/EAC and it can function as an oncogene as shown intumors of breast, brain, lung, prostate, pancreas, colon,liver, and chronic lymphocytic leukemia. It negativelyregulates tumor- and metastasis-suppressor genes PTEN,TPM1, PDCD4, and Sprouty2 (109–112). miR-194 expres-sion is regulated by hepatocyte nuclear factor (HNF)-1atranscription factor, which is induced during BE/EACand may lead to upregulation of miR-194 (109). Higherexpression of miR-194 is also observed in metastaticpancreatic cell lines (113). Among miRNAs found to bedownregulated during EAC development, let-7 family ofmiRNAs is tumor-suppressive and negatively regulatesRas oncogene. Fassan and colleagues confirmed upregu-lation of HMGA2, which is one of the target of let-7miRNA, using immunohistochemistry in tissue samples(110, 112, 114). Further studies in the regards of miRNAandmiRNA target geneswill improve the biologic under-standing of EAC pathogenesis and may also providenovel molecular targets for disease intervention.Notably, miRNAs are found to be stable in serum

    encapsulated in microvesicles and can be accessed easily(115). In fact, circulating miRNA profiling has showndistinct expression patterns in a number of cancers, otherthan EAC (116). This opens up new avenues for circulat-ing miRNA changes as a potential biomarker for EAC.

    GlycoproteinsProtein glycosylation is a common posttranslational

    modification with almost half of the proteins synthesizedundergoing 1 of the 2 major types either N-linked or O-linked glycan modifications. The biosynthetic process ofglycosylation is regulated by the expression and localiza-tion of glycosyltransferases/glycosidases and the avail-ability of substrate glycans (117).Aberrant glycosylation changes have previously been

    reported in several different cancers namelybreast cancer,prostate cancer, melanoma, pancreatic cancer, ovariancancer, etc. (118, 119). These changes include truncated

    forms of O-glycans, increased degree of branching in N-glycans, and elevated sialylation, sulfation, and fucosyla-tion with a range of other possible variations (119). Thedifferential glycosylation can alter protein interactions,stability, trafficking, immunogenicity, and function (118).Tumor-specific glycosylation changes are activelyinvolved in neoplastic progression, namely metastasis,as glycoproteins are found abundantly on cell surfacesand extracellularmatrices and therefore play a vital role incellular interactions.

    Lectins are a family of glycan-binding proteins exten-sively used in glycobiology due to preferential binding ofeach lectin to recognize specific glycan structures (119,120). The first effort to identify differential glycosylationin the progression to Barrett’s esophagus and EAC wasmade in 1987 by Shimamoto and colleagues using differ-ential binding pattern to 5 lectins in tissue specimens(121). The glycoconjugate expression profile in Barrett’sesophagus was found to be significantly different fromnormal esophageal epithelium. Interestingly, glycoconju-gate expression between Barrett’s esophagus and normalduodenum was quite similar. There were minimal glyco-conjugate expression changes between Barrett’s esopha-gus and LGD. However, EAC tissue samples showedsignificantly different lectin-binding pattern than BE/LGD (121). Using rabbit esophageal epithelium, Poor-khalkali and colleagues showeddifferential lectin bindingin response to acid/pepsin exposure suggesting acidexposure can induce cell surface glycosylation changes(122). In 2008, Neumann and colleagues used 4 differentlectins to identify pathologic mucosal changes (123). Theyobserved 2 distinct lectin-binding patterns. Onewas asso-ciated with the GERD, whereas the other pattern wascharacteristic for Barrett’s esophagus mucosa. Specifical-ly,UEA (Ulex europaeus) lectin bindingwasupregulated inBarrett’s esophagus tissue sections, which suggests pos-sible increase in fucosylation during the disease progress(123). A recently published study has concluded thatdysplasia can alter glycan expression and lectin bindingto the tissue samples. Fluorescently labeled WGA (wheatgerm agglutinin) lectin-binding intensity was found to beinversely related to the degree of dysplasia (124). Further-more, the authors used fluorescent-capable endoscope exvivo in the study and followed all the protocols in amanner that exactly mimics a clinical study in vivo. Fol-lowed by topical fluorescein-labeled WGA spray, theauthors measured fluorescence in the tissue samples.Measurement of lectin fluorescence was a more sensitiveapproach to identify dysplastic lesions as compared withwhite light endoscopic technique. Their data show clinicaluse of such a lectin-based endoscopic technique if devel-oped further (124). In a phase III biomarker clinical trialstudy, Bird-Liberman and colleagues combined 3 differ-ent abnormalities to predict EAC progression in patientswith Barrett’s esophagus. Along with using conventionalLGD and DNA content abnormalities they used AOL(Aspergillus oryzae) lectin binding to the tissue samples,which detects presence of a1-6 fucose on the cell surface

    Biomarkers for Esophageal Adenocarcinoma

    www.aacrjournals.org Cancer Epidemiol Biomarkers Prev; 22(7) July 2013 1199

    on June 7, 2021. © 2013 American Association for Cancer Research. cebp.aacrjournals.org Downloaded from

    Published OnlineFirst April 10, 2013; DOI: 10.1158/1055-9965.EPI-12-1415

    http://cebp.aacrjournals.org/

  • Table 4. Summary of literature describing miRNA expression changes in BE/EAC

    Sample size Upregulated in BE/EAC Downregulated in BE/EAC Ref.

    71 (BE-12, Barrett'sesophagus withoutdysplasia-20, LGD-27,EAC/HGD-12)

    miR-192 (P < 0.00001), miR-196a (P < 0.05):upregulated in Barrett's esophagus ascompared with healthy tissue.miR-196aexpression is correlated with progressionfrom IM-LGD-HGD-EAC (P < 0.005).

    miR203 (P < 0.00001): downregulation inBarrett's esophagus as compared withhealthy tissue.

    (209)

    22 (Barrett's esophaguswithout dysplasia-11,Barrett's esophagus withdysplasia-11)

    miR-15b (3.3-fold; P < 0.05), miR-203 (5.7-fold; P < 0.05): upregulated in dysplasia ascompared with nondysplastic Barrett'sesophagus.

    miR-486-5p (4.8-fold; P < 0.05), miR-let-7a(3.3-fold; P < 0.05): downregulated indysplasia as compared with nondysplasticBarrett's esophagus.

    (110)

    100 (EAC-100, adjacentnormal tissue as control)

    miR-21 (�3-fold; P < 0.05), miR-223 (�2-fold;P < 0.05), miR-192 (�3.5-fold; P < 0.05),and miR-194 (�3.5-fold; P < 0.05):upregulated in EAC as compared withadjacent normal tissue.

    miR-203 (�3-fold; P < 0.05): downregulatedin EAC as compared with adjacent normaltissue.

    (111)

    25 (Healthy-9, BE-5,HGD-1, EAC-10)

    miR-192 (1.7-fold; FDR < 1 e�07), miR-194(2-fold; FDR < 1e�07), miR-21 (3.7-fold;FDR ¼ 0.0003), miR-200c (1.9-fold; FDR ¼0.0015), miR-93 (1.3-fold; FDR ¼ 0.0108):upregulated in EAC as compared withBarrett's esophagus.

    miR-27b (1.43-fold; FDR ¼ 0.0003), miR-342(1.25-fold; FDR ¼ 0.0015), miR-125b (2-fold; FDR ¼ 0.0108), miR-100 (1.25-fold;FDR ¼ 0.011): downregulated in EAC ascompared with Barrett's esophagus.

    (104)

    75 (Healthy-15, BE-15,LGD-15, HGD-15,EAC-15)

    miR-215 (62.8-fold; P < 1e�07), miR-192(6.34-fold; P < 1e�07): upregulated inBarrett's esophagus in comparison withnormal tissue and remained at similar levelswith disease progress.

    miR-205 (10-fold; P ¼ 1.39e�0.5), let-7c(2.04-fold; P ¼ 3.11e�05), miR-203 (6.67-fold; P ¼ 3.2e�0.5): downregulated inBarrett's esophagus in comparison withnormal tissue and remained at similar levelsas disease progresses.

    (114)

    91 (LGD-31, HGD-29, EAC-31, In all cases adjacentnormal tissue used as acontrol)

    miR-200a (13.5-fold; P ¼ 0.02), miR-513(1.58-fold; P ¼ 0.03), miR-125b (9.2-fold; P¼ 0.04), miR-101 (1.83-fold; P¼ 0.04), miR-197 (1.61-fold; P ¼ 0.04): upregulated inLGD to HGD transition.

    miR-23b (1.45-fold; P ¼ 0.007), miR-20b(1.56-fold; P¼ 0.01), miR-181b (2.22-fold;P¼ 0.03), miR-203 (1.49-fold; P¼ 0.03), miR-193b (2.70-fold; P ¼ 0.04), miR-636 (4.17-fold; P ¼ 0.04): downregulated in LGD toHGD transition. let-7a (1.75-fold; P ¼ 0.01),let-7b (1.59-fold; P ¼ 0.009), let-7c (1.69-fold; P ¼ 0.03), let-7f (1.69-fold; P ¼ 0.03),miR-345 (2-fold; P ¼ 0.02), miR-494 (1.72-fold; P ¼ 0.03), miR-193a (2.27-fold; P ¼0.05): downregulated in HGD-EACdevelopment process.

    (112)

    48 (BE-19, EAC-29) miR-21 (�2.8-fold; P < 0.05), miR-143(�11.3-fold;P

  • (125). Thus, monitoring tissue glycan changes can becombined with existing biomarkers to improve the pre-dictive power of the currently used biomarkers.A potential mechanism responsible for these changes is

    considered to be bile acid exposure-induced gene expres-sion and secretory pathway changes in esophageal epi-thelium. Using carbohydrate-specific lectins that detectN- and O-linked glycosylation and core fucosylation,Byrne and colleagues have shown differential lectin bind-ing to the cell surface and differential intracellular local-ization when normal squamous and Barrett’s metaplasticcell lines were treated with deoxycholic acid (126). Nan-carrowand colleagues profiledwhole-genome expressionin normal squamous esophageal epithelium, Barrett’sesophagus, and EAC and concluded that Barrett’s esoph-

    agus is a tissue with enhanced glycoprotein synthesismachinery to provide strong mucosal defense againstacid exposure (127).

    Outlook—Circulating BiomarkersLast 3 decades showed continuously increased EAC

    incidences and similar trend is expected in future becauseof rising incidences of obesity and GERD in the popula-tion.Current endoscopic screeningprogrammightbenefitthe highest risk population to monitor disease progres-sion. Monitoring dysplasia in the tissue samples has notprovided fruitful outcome for early diagnosis; however,inclusion of the genomic and cell-cycle biomarkers hasshown definite improvement in the predictive powerover currently used histologic technique. Any biomarker

    Table 4. Summary of literature describing miRNA expression changes in BE/EAC (Cont'd)

    Sample size Upregulated in BE/EAC Downregulated in BE/EAC Ref.

    11 (EAC-11, differentlesions were collectedfrom these patients andclassified into Barrett'sesophagus, LGD, HGD,and EAC)

    miR-196a is overexpressed in early EAC(151-fold) > HGD (62.2-fold; P ¼ 0.00002) >LGD (31.1-fold; P ¼ 0.0005) > Barrett'sesophagus (28.9-fold; P ¼ 0.00001). Foldchanges are calculated as compared withnormal epithelium.

    — (107)

    45 (patients with EACundergoing surgery)

    miR-143 (P ¼ 0.0148), miR-199a_3p (P ¼0.0009), miR-199a_5p (P ¼ 0.0129), miR-100 (P ¼ 0.0022) and miR-145 (P ¼ 0.1176)expression