american gastroenterological association technical review on

35
American Gastroenterological Association Technical Review on the Role of the Gastroenterologist in the Management of Esophageal Carcinoma KENNETH K. WANG, MICHEL WONGKEESONG, and NAVTEJ S. BUTTAR Barrett’s Esophagus Unit, St. Mary’s Hospital, Mayo Clinic, Rochester, Minnesota T he goal of this evidence-based review was to examine the clinical practice of the gastroenterologist in the management of patients with esophageal carcinoma. The methods for this review were to search and review the literature available on MEDLINE and PREMEDLINE on the topics of esophageal neoplasm, esophageal cancer, and Barrett’s esophagus from 1968 to 2004. Bibliogra- phies of significant reports were also reviewed to ensure that the pertinent literature was reviewed. Recommen- dations are graded as to the level of evidence available on a scale of I–V. Level I evidence is the presence of at least one prospective, randomized, controlled trial, level II evidence is based on well-designed cohort or case-con- trolled studies, level III evidence is based on case series or flawed clinical trials, level IV evidence is based on opin- ions of respected authorities or expert committees, and level V evidence is insufficient evidence to form any opinions. Significance of Esophageal Cancer Esophageal cancer is associated with one of the highest cancer mortality rates in the United States. In 2000, the last year of complete data available from the national cancer Surveillance, Epidemiology, and End Re- sults database, the 5-year relative survival rates from esophageal cancer were the fifth lowest at 15.4%. In addition, the incidence of esophageal cancer is still sig- nificantly trending upward in white men with a 0.4% annual percentage increase from 1992 to 2000. This can be compared with colon cancer, which has actually had a significant decrease of 0.9% annual percentage change over the same period. Esophageal cancer is a predomi- nantly male condition with a male/female incidence of 3.6:1. Esophageal cancers affect older patients, with the peak incidence in those 65–74 years old. It is estimated that, in 2003, there were 13,900 new cases of esophageal cancer and 13,000 deaths due to esophageal cancer. The mortality rate of esophageal cancer is significantly higher in minority populations than in white people. There are 2 major types of esophageal cancer: adeno- carcinoma and squamous cell cancer. The primary known risk factors for adenocarcinoma of the esophagus are smoking, chronic gastroesophageal reflux disease, and Barrett’s esophagus. Known risk factors for squamous cell cancer of the esophagus include smoking, alcohol use, exposure to nitrosamines, ingestion of lye, Fanconi’s anemia, achalasia, Plummer–Vincent webs, and tylosis. Screening and Surveillance for Esophageal Cancer Screening for Esophageal Cancer Adenocarcinoma. Screening for esophageal can- cer depends on the determination of the patient’s risk for cancer, the cost and efficacy of the screening procedure, the stage at which the cancer can be diagnosed, and the treatment options available. At the current time, there is no direct evidence that has validated the use of screening for esophageal cancer in the United States. Screening for esophageal adenocarcinoma has been primarily focused on the detection of Barrett’s esophagus with subsequent surveillance. Although screening for Barrett’s esophagus has not been proven to decrease the risk of cancer, it has become accepted that surveillance for Barrett’s esophagus can detect disease at earlier stages. 1–3 Barrett’s esophagus has traditionally been found to be associated with chronic gastroesophageal reflux disease, and case series have found increasing incidences of Barrett’s esophagus de- pending on the number of years of reflux symptoms. 4 However, a single-center study of 110 subjects found that 7% of asymptomatic individuals had long-segment Abbreviations used in this paper: CI, confidence interval; CT, com- puted tomography; EUS, endoscopic ultrasonography; FDA, Food and Drug Administration; Nd:YAG, neodymium:yttrium-aluminum-garnet; OCT, optical coherence tomography; PAR, population attributable risk; PDT, photodynamic therapy; PET, positron emission tomography; QALY, quality-adjusted life year; SEMS, self-expanding metal stents. © 2005 by the American Gastroenterological Association 0016-5085/05/$30.00 doi:10.1053/j.gastro.2005.03.077 GASTROENTEROLOGY 2005;128:1471–1505

Upload: patrick89

Post on 18-Dec-2014

1.567 views

Category:

Documents


4 download

DESCRIPTION

 

TRANSCRIPT

Page 1: American Gastroenterological Association Technical Review on

AtE

KB

Tmmltaptdaoetflilo

h2nseanabson3ptcmi

GASTROENTEROLOGY 2005;128:1471–1505

merican Gastroenterological Association Technical Review onhe Role of the Gastroenterologist in the Management ofsophageal Carcinoma

ENNETH K. WANG, MICHEL WONGKEESONG, and NAVTEJ S. BUTTAR

arrett’s Esophagus Unit, St. Mary’s Hospital, Mayo Clinic, Rochester, Minnesota

crsBcua

ccttnfeoshbchgfpHt

pDOPQ

he goal of this evidence-based review was to examinethe clinical practice of the gastroenterologist in the

anagement of patients with esophageal carcinoma. Theethods for this review were to search and review the

iterature available on MEDLINE and PREMEDLINE onhe topics of esophageal neoplasm, esophageal cancer,nd Barrett’s esophagus from 1968 to 2004. Bibliogra-hies of significant reports were also reviewed to ensurehat the pertinent literature was reviewed. Recommen-ations are graded as to the level of evidence available onscale of I–V. Level I evidence is the presence of at leastne prospective, randomized, controlled trial, level IIvidence is based on well-designed cohort or case-con-rolled studies, level III evidence is based on case series orawed clinical trials, level IV evidence is based on opin-ons of respected authorities or expert committees, andevel V evidence is insufficient evidence to form anypinions.

Significance of Esophageal Cancer

Esophageal cancer is associated with one of theighest cancer mortality rates in the United States. In000, the last year of complete data available from theational cancer Surveillance, Epidemiology, and End Re-ults database, the 5-year relative survival rates fromsophageal cancer were the fifth lowest at 15.4%. Inddition, the incidence of esophageal cancer is still sig-ificantly trending upward in white men with a 0.4%nnual percentage increase from 1992 to 2000. This cane compared with colon cancer, which has actually had aignificant decrease of 0.9% annual percentage changever the same period. Esophageal cancer is a predomi-antly male condition with a male/female incidence of.6:1. Esophageal cancers affect older patients, with theeak incidence in those 65–74 years old. It is estimatedhat, in 2003, there were 13,900 new cases of esophagealancer and 13,000 deaths due to esophageal cancer. Theortality rate of esophageal cancer is significantly higher

n minority populations than in white people.

There are 2 major types of esophageal cancer: adeno-arcinoma and squamous cell cancer. The primary knownisk factors for adenocarcinoma of the esophagus aremoking, chronic gastroesophageal reflux disease, andarrett’s esophagus. Known risk factors for squamousell cancer of the esophagus include smoking, alcoholse, exposure to nitrosamines, ingestion of lye, Fanconi’snemia, achalasia, Plummer–Vincent webs, and tylosis.

Screening and Surveillance forEsophageal Cancer

Screening for Esophageal Cancer

Adenocarcinoma. Screening for esophageal can-er depends on the determination of the patient’s risk forancer, the cost and efficacy of the screening procedure,he stage at which the cancer can be diagnosed, and thereatment options available. At the current time, there iso direct evidence that has validated the use of screeningor esophageal cancer in the United States. Screening forsophageal adenocarcinoma has been primarily focusedn the detection of Barrett’s esophagus with subsequenturveillance. Although screening for Barrett’s esophagusas not been proven to decrease the risk of cancer, it hasecome accepted that surveillance for Barrett’s esophagusan detect disease at earlier stages.1–3 Barrett’s esophagusas traditionally been found to be associated with chronicastroesophageal reflux disease, and case series haveound increasing incidences of Barrett’s esophagus de-ending on the number of years of reflux symptoms.4

owever, a single-center study of 110 subjects foundhat 7% of asymptomatic individuals had long-segment

Abbreviations used in this paper: CI, confidence interval; CT, com-uted tomography; EUS, endoscopic ultrasonography; FDA, Food andrug Administration; Nd:YAG, neodymium:yttrium-aluminum-garnet;CT, optical coherence tomography; PAR, population attributable risk;DT, photodynamic therapy; PET, positron emission tomography;ALY, quality-adjusted life year; SEMS, self-expanding metal stents.

© 2005 by the American Gastroenterological Association0016-5085/05/$30.00

doi:10.1053/j.gastro.2005.03.077

Page 2: American Gastroenterological Association Technical Review on

(5gsTsebplgfoTod

hwbdsdt

ccfimidtdsimtoistbeaclwtsft

cttstositSdaisacmc

setpuC

e5msseacosb(

BateTtscp

1472 WANG ET AL GASTROENTEROLOGY Vol. 128, No. 5

�3 cm) Barrett’s esophagus.5 A multicenter study of36 subjects found that long-segment Barrett’s esopha-us was only present in 0.36% of subjects, althoughhort-segment Barrett’s esophagus was found in 5.6%.6

his study did find that patients with heartburn had aignificantly higher prevalence of long-segment Barrett’ssophagus than those who did not. This is significantecause past studies have shown that the degree of neo-lasia found in Barrett’s esophagus appears to be corre-ated with the length of Barrett’s esophagus.7 Otherroups that may be at risk would include those withamilial occurrence of adenocarcinoma, but this hasnly been described in a limited number of families.he cost-efficacy of screening family members becausef a history of Barrett’s esophagus has not beenemonstrated.8 –12

Screening methods for detection of Barrett’s esophagusave included standard endoscopy, unsedated endoscopyith ultrathin endoscopes, catheter-based cytology, andalloon cytology.13–16 Although preliminary studies in-icate some promise with these technologies in terms ofcreening for adenocarcinoma, there have not been anyefinitive trials to allow recommendation of theseechniques.

Squamous cell cancer. Screening for squamousell cancer in the general population of the United Statesannot be justified because of the low incidence of thisorm of cancer. However, specific subgroups may bedentified that warrant screening endoscopy for squa-ous cell cancer. The most likely to benefit from screen-

ng would be those who have tylosis, which is a geneticefect in the 17q25 region that is found in patients withhickened palms and soles.17,18 This group is likely toevelop cancer by the age of 65 years and should undergocreening for squamous cell cancer. Patients with lye-nduced or caustic strictures develop cancers approxi-ately 46 years after ingestion and have been described

o have an 8% incidence of cancer.19,20 The developmentf cancer in patients with long-standing achalasia isnfrequent, especially in women, and it is unclear ifurveillance is warranted in these patients.21 Most pa-ients with achalasia are found to have prevalent cancersecause they are usually diagnosed when obstruction andsophageal dilation are found.22 Fanconi’s anemia haslso been associated with the development of esophagealancer and may become more common as patients surviveonger after bone marrow transplantation.23 Patientsith existing aerodigestive tumors, especially those of

he oral cavity, who have had long-term extensive expo-ure to alcohol and tobacco might benefit from screeningor cancers of the esophagus, although the rationale for

his is based on case series.24 Screening for esophageal i

ancer should be performed at the time of diagnosis ofhe head-and-neck cancer. Extensive alcohol consump-ion alone has been found to be an important criterion forcreening in Asian patient populations.25 Partial gastrec-omies have been associated with an increased incidencef squamous cell cancers of the esophagus; these reportstem from areas of the world where squamous cell cancers commonly found, and there is no convincing evidenceo support the screening of patients in the Unitedtates.26 The occurrence of squamous cell cancer is notecreasing in minority populations in the United Statesnd remains the most frequent form of esophageal cancern black and Hispanic populations.27,28 Other conditionsuch as Plummer–Vinson webs have also been identifieds being associated with an increased risk for squamousell cancers, but the decreasing frequency of these websakes this almost a historical footnote in Western

ountries.29

Screening methods such as cytologic balloons orponges for screening for squamous cell cancers have beenxtensively tested in Asia. Balloons appear to be betterhan sponges, with an increased sensitivity of 44% com-ared with 18% for sponges.30 This technology has beensed as the primary screening tool in high-risk areas ofhina.31

Summary of Evidence—There is level III evidence thatndoscopic screening of male white patients older than0 years of age with symptoms of gastroesophageal refluxay be cost-effective. Patients with tylosis, lye-induced

trictures, or Fanconi’s anemia would benefit fromcreening endoscopy for squamous cell cancers (level IIIvidence). In addition, patients with long-term tobaccond alcohol use, achalasia, or prior head-and-neck cancersould be considered for screening, depending on theirther risk factors (level III evidence). The interval forurveillance of these patients has not been established,ut yearly investigations would seem to be reasonablelevel IV evidence).

Surveillance for Esophageal Cancer

Significance of surveillance. The importance ofarrett’s esophagus derives from its association with esoph-geal adenocarcinoma.32,33 Approximately 5%–10% of pa-ients diagnosed with Barrett’s esophagus may developsophageal adenocarcinoma based on older studies.32,34,35

he incidence of cancer will most likely decrease withhe increasing detection of Barrett’s esophagus throughcreening programs. Surveillance of Barrett’s esophagusan detect dysplasia in Barrett’s esophagus, which canredict the risk of future diagnosis of esophageal cancer

n these patients.36,37
Page 3: American Gastroenterological Association Technical Review on

ecfipttBnddmbecioy2Picootpt

ppchaoFcbsw.

to8sagiuwci

c(wpopibeitin

tsptowhpeucwtvdhccrgoeshoSdcopo

cdfst

May 2005 AMERICAN GASTROENTEROLOGICAL ASSOCIATION 1473

The incidence of cancer in patients with Barrett’ssophagus is low in general, which makes surveillanceost-ineffective unless at-risk populations can be identi-ed.38,39 At the current time, dysplasia is used as therimary means to discriminate at-risk patients. A sys-ematic endoscopic biopsy protocol, generally acceptedo be 4 quadrant biopsy specimens taken every 2 cm ofarrett’s mucosa, can provide tissue for histologic diag-osis of dysplasia. The grading system used to classifyysplasia in Barrett’s esophagus is based on the systemeveloped for ulcerative colitis.40–42 Although moleculararkers have shown promise, dysplasia is currently the

est indicator for the risk of cancer. Increasing grade andxtent of dysplasia are associated with increasing risk ofancer.37,43–48 Information from several prospective stud-es and a Barrett’s esophagus registry provide a totalf 783 patients with a follow-up of 2.7–7.3ears.37,43,45,47,49,50 These combined series indicate that% of patients without dysplasia progressed to cancer.rogression from low-grade dysplasia to cancer was noted

n 7% of patients and from high-grade dysplasia toancer in 22% of patients. Unfortunately, it is difficult tobtain true cancer incidence per year of observation basedn the data provided in these series. It is also apparenthat low-grade dysplasia had a wide range of rates ofrogression between the series. This may be because ofhe variability in interpretation of low-grade dysplasia.46

Other findings on endoscopy that have been shown toredict the development of esophageal cancer include theresence of any mucosal abnormalities noted on endos-opy and the extent of high-grade dysplasia noted onistology.48 The presence of mucosal nodularity has beenssociated with a 2.5-fold increased risk of cancer devel-pment in patients with high-grade dysplasia (P � .01).ocal high-grade dysplasia, defined as involvement of �5rypts in one biopsy specimen from the entire set ofiopsy specimens, has been found to be associated withignificantly less risk of cancer development than thoseith greater amounts of high-grade dysplasia (P �

02).48

Unfortunately, the classification of dysplasia is subjec-ive and does have significant observer variation. Inter-bserver pathologic agreement can improve to 85%–7% when the degree of dysplasia is placed in a 2-tierystem. This is done by combining high-grade dysplasiand intramucosal carcinoma in one category and low-rade dysplasia, indefinite for dysplasia, and no dysplasian another.51 This was substantiated by a recent studysing a group of expert pathologists.52 In this study,hen analysis was performed using 2 broad diagnostic

ategories (Barrett’s esophagus without dysplasia, indef-

nite and low-grade dysplasia vs high-grade dysplasia and H

arcinoma), intraobserver agreement was near perfectmean � � 0.82 and 0.80) and interobserver agreementas substantial (� � 0.66–0.70). When the analysis waserformed using the 4 separate classifiers (Barrett’s with-ut dysplasia, indefinite for dysplasia and low-grade dys-lasia, high-grade dysplasia, and carcinoma), the meanntraobserver � was once again substantial (0.64–0.68)ut the mean interobserver � was only 0.43–0.46 (mod-rate agreement). Because of this difficulty in determin-ng the presence of dysplasia, using recent recommenda-ions to screen and perform surveillance only if dysplasias found may be ineffective if histologic interpretation isot accurate.53

Despite the difficulties with surveillance, the detec-ion of high-grade dysplasia or early cancer has beenhown to have the potential to improve survival inatients with Barrett’s esophagus.54,55 Cancers confinedo the esophagus are associated with a 5-year survival ratef 70% compared with a survival rate of 20% for patientsith more invasive cancers.56 Nodal involvement, whichelps determine prognosis, is far less likely to occur inatients who are found to have cancer on surveillancendoscopies compared with those patients who are notndergoing surveillance.1,3,56,57 This leads to signifi-antly improved survival in surveyed patients comparedith those patients who underwent surgery for symp-

omatic disease. Retrospective studies of patients in sur-eillance programs suggest that the patients with inci-ental cancers detected during surveillance are likely toave earlier staged disease than those who present be-ause of symptoms.58,59 Although these case series seemonvincing, there are several concerns. Many of the ret-ospective studies compared patients in surveillanceroups with historical controls. Patients of advanced ager with significant comorbidity are less likely to benrolled in a surveillance program. On the other hand,ubjects enrolled in surveillance programs might haveealthier lifestyles, might have health-seeking behaviors,r were selected because of overall better health status.urvival differences between these 2 groups may well beue to selection bias. In addition, earlier detection ofancer can lead to a false increase in survival time becausef lead-time bias, which results from an early diagnosis ofreclinical cancer rather than any true effect of detectionn subsequent mortality.

In at least 2 small surveillance cohorts, esophagealancer has been reported to be an uncommon cause ofeath in patients with Barrett’s esophagus.60,61 There-ore, the morbidity and mortality from surveillance andubsequent treatment might overshadow any advantagehat surveillance provides by early detection of cancer.

owever, a retrospective population-based cohort study
Page 4: American Gastroenterological Association Technical Review on

oeftlpsdrga

trr5d1fsssmcwgr

sdpts$soSwcwdilaqo

spdai

atasohm4lcbpscsmatssebfahuf

adpon4

bbcnb

ibtacsccfs

1474 WANG ET AL GASTROENTEROLOGY Vol. 128, No. 5

f endoscopic surveillance in 23 patients with Barrett’ssophagus among 589 patients with adenocarcinomaound an improved survival benefit.62 Eleven of 15 pa-ients who were diagnosed with cancer while in surveil-ance programs were alive compared with none of 8atients not under surveillance. None of the deaths in theurveillance group were from cancer compared with 4eaths in the nonsurveillance group that were directlyelated to cancer. Three patients in the surveillanceroup and one patient in the nonsurveillance group dieds a result of complications of surgery.

Allocation of resources based on risk stratification hashe potential to make surveillance more cost-effective. Aecent decision analysis model examined the cost-utilityatio for different surveillance strategies.38 A cohort of0-year-old patients with Barrett’s esophagus withoutysplasia was evaluated for surveillance strategies every–5 years and no surveillance, with esophagectomy per-ormed if high-grade dysplasia was diagnosed. This studyuggested that, with an annual cancer risk of 0.4%,urveillance every 5 years was the only cost-effectivetrategy. More frequent surveillance was associated withore cost and yielded a lower life expectancy. The in-

remental cost-utility ratio for surveillance every 5 yearsas $98,000 per quality-adjusted life year (QALY)ained, comparable to the incremental cost-effectivenessatios of accepted practices.

Another detailed cost-utility analysis assessed thetrategy of surveillance of Barrett’s esophagus detecteduring screening.53 The strategy of surveillance of onlyatients with dysplasia appears to be cost-effective rela-ive to other currently accepted medical practices. Thetrategy yielded an incremental cost-effectiveness ratio of10,440 per QALY saved compared with no screening. Atrategy of no screening would incur direct medical costsf caring for those developing cancer of $16 million.creening and surveillance of patients with dysplasiaould prevent 2580 deaths caused by esophageal adeno-

arcinoma at a cost of $262 million. Fifty-nine patientsould need to be screened and surveyed to prevent oneeath from cancer. Although this model was thorough, its limited by several assumptions, such as that surveil-ance for patients with high-grade dysplasia can ceasefter 2 years and that there are no adverse effects on theuality of life of patients undergoing surveillance, whichther studies have shown to be decreased.63

Biopsy method. Patients who are undergoingurveillance should follow a systematic endoscopic biopsyrotocol after they are free from esophagitis.64 The evi-ence for this is based on expert opinion, limited data,nd the outcome of cohorts of patients managed empir-

cally. Biopsy specimens are first taken from any mucosal w

bnormality, followed by rigorous sampling from each ofhe 4 quadrants of the esophagus every 1–2 cm startingt the gastroesophageal junction and stopping at thequamocolumnar junction using standard or jumbo bi-psy forceps. The goal is to determine if the patient hasigh-grade dysplasia or early cancer so that appropriateanagement can be initiated. The effectiveness of taking

-quadrant biopsy specimens every 1 cm along theength of the Barrett’s segment with jumbo biopsy for-eps instead of the traditional standard biopsy forceps haseen assessed.42 In a retrospective analysis of the Seattlerotocol, it was shown that taking 4-quadrant biopsypecimens every 2 cm would have missed 50% of theancers that were found by taking 4-quadrant biopsypecimens every 1 cm.65 Jumbo biopsy forceps acquireore tissue in a single biopsy than regular biopsy forceps

nd have not been associated with increased complica-ions.66 The use of jumbo biopsy forceps has not beenhown to decrease the number of samples necessary forurveillance, and they require the use of a therapeuticndoscope. The need for use of jumbo biopsy forceps haseen questioned, and one study found no statistical dif-erence in the rate of unsuspected cancers found at esoph-gectomy when patients with Barrett’s esophagus withigh-grade dysplasia underwent preoperative endoscopysing jumbo biopsy forceps versus standard biopsyorceps.67

It is generally accepted that patients with mucosalbnormalities in Barrett’s esophagus should undergo en-oscopic mucosal resection to increase detection of neo-lasia. In one small series, endoscopic mucosal resectionf suspicious lesions diagnosed superficial adenocarci-oma in 13 patients (52%) and high-grade dysplasia in(16%) of 25 patients with Barrett’s esophagus.68

Methods of surveillance. Several methods haveeen studied to perform surveillance. These includerush cytology, chromoendoscopy, magnification endos-opy, and optical imaging techniques. All of these tech-iques have certain potential advantages over routineiopsy.

Brush cytology. Brush cytology has been advocatedn the surveillance of patients with Barrett’s esophagusecause it is faster to perform and it can sample morehan the 1% of the total Barrett’s mucosal area typicallychieved with standard biopsy. The diagnostic value ofytology in patients with Barrett’s esophagus was as-essed using 66 pairs of endoscopic biopsy specimens andytology specimens.69 Cytologic analysis was used toorrectly identify 7 of 8 esophageal cancers, with onealse-negative sample described on analysis as highlyuspicious for cancer. In other studies, cytologic brushing

as found to be a complementary technique in detecting
Page 5: American Gastroenterological Association Technical Review on

slHtwhuttcaqhctsphp

tttccspbatl

iLsfacmmcicwmis9actm

da1itdfe

ebldgwoapdracnipcbTipati(nmdttspep

vviasei

May 2005 AMERICAN GASTROENTEROLOGICAL ASSOCIATION 1475

erious Barrett’s lesions when combined with the histo-ogic examination of esophageal biopsy specimens.70,71

owever, there was only 72% (47/65) concordance be-ween the 2 diagnostic techniques. In the 18 cases thatere discordant, cytologic analysis diagnosed 13 at aigher level of abnormality than the biopsy diagnosis andnderdiagnosed 5. There is potential to further improvehe efficacy of cytology by using objective assessmentools such as digital image analysis, which involves aomputerized analysis of the digitized microscopic im-ge.72,73 This has been demonstrated in China, whereuantitative DNA fluorescence has been found to en-ance detection of squamous cell cancers when applied toytology taken in a high-risk population.74 As men-ioned previously, balloon cytology for the surveillance ofquamous cell cancers has become an accepted part ofractice in high incidence areas, although such devicesave not been shown to be beneficial in Westernopulations.30,75

Chromoendoscopy. Chromoendoscopy involves theopical application of dyes during endoscopy in an efforto enhance the detection of mucosal patterns or lesions onhe basis of their staining characteristics.76 Stains arelassified as vital (absorptive), contrast, or reactive, ac-ording to their mode of action. Vital stains (e.g., Lugol’solution and methylene blue) enter specific cell types byreferential absorption or diffusion across the cell mem-rane. Contrast stains (e.g., indigo carmine and aceticcid) seep into mucosal interstices to highlight mucosalopography. Reactive stains chemically react with cellu-ar constituents.

The body of the literature regarding chromoendoscopyn the esophagus consists primarily of staining withugol’s solution for detection of squamous dysplasia anduperficial carcinoma and staining with methylene blueor assessment of Barrett’s esophagus. Lugol’s solution isn iodine-based vital dye that has an affinity for glycogenontained in nonkeratinized squamous epithelium. Chro-oendoscopy with Lugol’s solution has been used pri-arily to screen individuals at high risk for squamous

ell cancer.77–80 The application of Lugol’s solution dur-ng endoscopy is not standardized, with nonuniformoncentrations (1%–3%) and volumes (10–50 mL) asell as variable definitions of “unstained” areas thaterit biopsy.77–80 Despite these differences, these studies

n aggregate show unstained lesions to have a highensitivity (91%–100%) and moderate specificity (40%–5%) for squamous dysplasia and carcinoma.77 Lesionsre found to be more clearly defined and can be signifi-antly larger after staining,81 providing critical informa-ion, particularly if local therapy, such as endoscopic

ucosal resection, is contemplated. Studies of chromoen- a

oscopy with Lugol’s solution in Western populationsre sparse. A prospective analysis of a French cohort of58 alcoholic and/or smoking patients showed no signif-cant advantage of staining with Lugol’s solution relativeo the visual diagnostic accuracy achieved by videoen-oscopy alone.81 Lugol’s-assisted endoscopy may be use-ul in populations living in highly endemic regions forsophageal squamous cell cancer.

Methylene blue stains absorptive cells (intestinal-typepithelium), including specialized intestinal metaplasia,ut does not stain normal squamous or gastric epithe-ium. Methylene blue may be particularly useful in theiagnosis of short segments of columnar-lined esopha-us.82 Other uses include identifying dysplastic fociithin Barrett’s epithelium and identifying residual focif Barrett’s esophagus following mucosal ablative ther-py. Chromoendoscopy with methylene blue requiresretreatment of the mucosa with a mucolytic agent, awell time for the dye to take effect, and a rinse phase toemove excess dye. In general, methylene blue stainingdds 5–10 minutes to the procedure time, but the ac-essories required are inexpensive.83 Specialized intesti-al metaplasia typically stains blue, while a “lighter”ntensity and increased heterogeneity in the stainingattern predict high-grade dysplasia and/or cancer.84 Fo-al unstained areas may represent dysplastic foci within aackground of heterogeneous methylene blue staining.85

hese findings, however, are not consistently replicatedn other studies, and the reproducibility of stainingatterns among Barrett’s tissue types remains debat-ble.86 This technique has been used to allow the gas-roenterologist to perform fewer biopsies to find signif-cantly more dysplasia or cancer in a biopsy specimen12% vs 6%) compared with the random biopsy tech-ique.85 However, 2 prospective trials comparing theethylene blue–guided biopsy technique with the ran-

om biopsy technique found no significant differences inhe detection of dysplasia or early cancer between the 2echniques.87,88 Methylene blue has also recently beenhown to induce oxidative damage of DNA because ofhotoactivation of the dye by the white light of thendoscope, which certainly increases the risk of its use inremalignant tissue.89

Magnification endoscopy. Most new electronicideoendoscopes are equipped with charge-coupled de-ice chips of high pixel density (400K), enabling high-mage resolution (ability to discriminate 2 closelypproximated points). Megapixel density (850K) endo-copes have recently been introduced, which may furthernhance detection. Magnification endoscopy enlarges themage, which enhances mucosal detailing and is gener-

lly used in combination with chromoscopy. Magnifica-
Page 6: American Gastroenterological Association Technical Review on

tamciWfnd

chciSefltttst

ecs(afsiTmyuefoi(atttd

scopaf

tslaanopwsodnoo

is11pmttfitithswsemp

dgstbdoardtiwr

1476 WANG ET AL GASTROENTEROLOGY Vol. 128, No. 5

ion chromoendoscopy has been used primarily to char-cterize Barrett’s esophagus.90–93 Ridged or villousucosal patterns observed under magnification endos-

opy using a variety of stains have been correlated withntestinal metaplasia or dysplasia in case series.91–93

hether magnification endoscopy will decrease the needor endoscopic biopsy or significantly enhance the diag-ostic yield over conventional techniques remains to beetermined in large controlled trials.

Optical biopsy. Optical biopsy is a term that en-ompasses a variety of techniques that use light to en-ance detection of dysplastic lesions. Included in thisategory are spectroscopic techniques and low coherencenterferometry (optical coherence tomography [OCT]).pectroscopy is a light-based diagnostic technique thatxploits properties of light-tissue interactions, such asuorescence, elastic scattering, and Raman scattering, forissue characterization. Collected in the form of spectra,hese optical (light) signals are sensitive to microstruc-ural and/or molecular changes accompanying varioustages of tissue pathology, and spectral differences canhen be correlated to specific histopathologic diagnoses.

Assessment of optical spectroscopic techniques in thesophagus has been limited to small-scale, proof-of-prin-iple studies.94–102 Most studies deal with fluorescencepectroscopy, which shows moderate to high sensitivity75%–100%) and specificity (65%–95%) in differenti-ting high-risk (high-grade dysplasia/adenocarcinoma)rom low-risk (nondysplastic to low-grade dysplasia)amples of Barrett’s esophagus.94–98 False positives occurn the presence of inflammatory or reactive changes.94

he range of diagnostic accuracies primarily relates toethodological differences, because the technique has

et to be optimized for Barrett’s esophagus. It is alsonclear whether fluorescence spectroscopy, aided by anxogenously administered fluorescent agent with affinityor neoplastic tissues (e.g., porfimer sodium or 5-amin-levulinic acid), improves discrimination of Barrett’s ep-thelia over naturally occurring tissue fluorescenceautofluorescence).99–101,103 Future comparative studiesre required to address this issue. Other emerging spec-roscopic techniques, including elastic scattering spec-roscopy,102 Raman spectroscopy,103 and multimodal op-ical spectroscopy,104 have shown initial promise inifferentiating Barrett’s epithelia in feasibility studies.Fluorescence endoscopy is an extension of its spectro-

copic counterpart into an imaging technique, whichonsists of sensitive cameras capturing the emitted flu-rescence emanating from a mucosal field of view ap-roaching that of a conventional endoscope. The obviousdvantage over point spectroscopy is the ability to per-

orm widespread assessment of the mucosa for areas likely t

o harbor dysplastic or early cancerous changes. Recenttudies of fluorescence endoscopy, however, have showedimited diagnostic utility in Barrett’s esophagus.88,105 Insmall prospective study of 35 patients, the sensitivity

nd specificity of autofluorescence imaging for the diag-osis of dysplasia or cancer versus Barrett’s mucosa with-ut dysplasia were 21% and 91%, respectively.88 Arospective, randomized, crossover study of 50 patientsith Barrett’s esophagus, published in abstract form,

howed no significant benefit of fluorescence-guided bi-psies over a standard surveillance biopsy protocol for theetection of high-grade dysplasia and early adenocarci-oma.105 Fluorescence endoscopy is an evolving technol-gy. Optical biopsy and imaging techniques are not yetptimized for clinical use.

OCT is analogous to ultrasonography but uses lightnstead of sound waves to produce high-resolution, cross-ectional imaging of tissue. The spatial resolution of0–25 �m achieved by endoscopic OCT systems is0-fold better than that of high-frequency ultrasonogra-hy but at the expense of a limited sampling depth (�2m).106–109 Current endoscopic OCT devices are cathe-

er based (2–2.7 mm in diameter), allowing scanning ofhe esophagus in a linear,107 transverse,106 or radial108

ashion. Using predetermined OCT criteria for special-zed intestinal metaplasia, the linear scanning OCT sys-em was found to be 97% sensitive and 92% specific fordentifying specialized intestinal metaplasia.110 A quan-itative analysis of the OCT signal seemed to identifyigh-grade dysplasia with high sensitivity (100%) andpecificity (85%) in a small retrospective study, whichill require prospective validation on a larger sample

ize.110 Similar to spectroscopy, OCT is a technique involution, and further refinements are anticipated thatay improve its diagnostic accuracy for the detection of

reneoplastic lesions.Frequency of surveillance. The frequency of en-

oscopic surveillance in patients with Barrett’s esopha-us has been debated without the benefit of well-con-tructed studies. Surveillance intervals recommended inhis section are supported by limited evidence, which isased on our understanding of the natural history ofevelopment of esophageal adenocarcinoma dependingn the grade and extent of dysplasia in Barrett’s esoph-gus. The goal of surveillance is to identify patients atisk and assess them in a timely fashion to prevent oretect cancer at an earlier treatable stage. Summaries ofhe studies that have investigated the incidence of cancern Barrett’s esophagus appear in Tables 1–4. Patientsith nondysplastic Barrett’s esophagus should undergo

epeat endoscopy 1 year after the initial endoscopy. Pa-

ients with persistent nondysplastic Barrett’s esophagus
Page 7: American Gastroenterological Association Technical Review on

Table 1. Diagnosis of Prevalent or Incidental Cancer on Follow-up of Patients With No Dysplasia

Institute or geographicarea Study type

Studyduration

No. ofpatients

Mean ormedianage (y) Duration of follow-up

Totalcancers (%)

Incidentalcancers (%) Comments Reference

University of Chile,Santiago, Chile Prospective 1978–1991 152 52 100 mo (mean) 4 (2.6) 4 (2.6)

LGD developed in 15patients on follow-up

Csendes et al,115

1998University of Otago

Medical School,Dunedin, NewZealand Prospective 1980–1986 52 63 16.4 mo (mean) 0 0 Cooper et al,182 1987

Deutsche Klinik furDiagnostik,Wiesbaden,Germany Prospective 1980–1999 60 61 10 y (mean) 2 (3.3) 2 (3.3)

5 patients developedLGD Eckardt et al,340 2001

University Hospital, ElPalmar, Murcia,Spain Prospective 1982–1993 59 37 287 patient-years 2 (3.3) 2 (3.3) Ortiz et al,341 1996

VA Medical Centerand University ofKansas MedicalCenter Prospective — 80 61

40 mo (mean) or362 patient-years 2 (2.5) 2 (2.5)

One patient developedHGD, 3 hadpersistent LGD, 18had transient LGD,and 56 remainednondysplastic.Patients withprevalent cancerdiagnosed at entry orwithin the first 6months wereexcluded Weston et al,45 1999

Fred HutchinsonCancer ResearchCenter, University ofWashington

Prospectivecohort 1983–1998 129 62

2.4 y (median) or3.9 y (mean) forthose who did notdevelop cancer 5 (3.9) 5 (3.9)

5-year cumulativeincidence of cancerwas reported to be1.7% and 8-yearcumulative incidencewas reported to be8.4% Reid et al,43 2000

LGD, low-grade dysplasia; HGD, high-grade dysplasia.

May

20

05

AM

ERIC

AN

GA

STR

OEN

TERO

LOG

ICA

LA

SS

OC

IATIO

N1

47

7

Page 8: American Gastroenterological Association Technical Review on

Table 2. Diagnosis of Prevalent or Incidental Cancer on Follow-up of Patients in Which the Degree of Dysplasia Was Not Defined or There Were �10 Patients in a SingleDysplasia Category

Institute or geographicarea Study type

Studyduration

No. ofpatients

Mean ormedianage (y)

Duration offollow-up

Totalcancers (%)

Incidentalcancers (%) Comments Reference

Mayo Clinic, Rochester,MN

Retrospective 1960–1983 122 58 8.5 y (mean) 20 (16) 2 (1.9) Cameron et al,342 1985

Boston VA Medical Center,Boston UniversitySchool of Medicine, andTufts University Schoolof Medicine

Retrospective 1962–1983 115 58 3.3 y (mean) 11 (9.5) 2 (1.9) 4 patients had LGD at baseline; one ofthem progressed to HGD and finallyto cancer. HGD developed in 7patients

Spechler et al,34 1984

VA Outcomes Group,Dartmouth-HitchcockMedical Center

Retrospective 1970–1994 102 63 4.8 y (median) 3 (3) 3 (3) 8% of nondysplastic patientsdeveloped dysplasia. In 2 patients,HGD developed before cancer

Katz et al,343 1998

Castle Hill Hospital,Cottingham, UnitedKingdom

Retrospective 1971–1991 26 62 11.5 y (mean) 4 (15.4) 4 (15.4) Moghissi et al,344 1993

Lahey Clinic, Burlington,MA

Retrospective 1973–1989 176 56 3 y (median) 5 (2.8) 5 (2.8) 11% of patients developed dysplasiaon follow-up

Williamson et al,33 1991

University of Rotterdam,Dutch cohort

Retrospectivecohort

1973–1994 155 42 9.3 y (mean)or 1440patient-years

8 (5.3) 8 (5.3) Van der burgh et al,60

1996

Helsinki University, CentralHospital, Finland

Retrospective 1975–1985 32 59 166 patient-years

3 (9.3) 3 (9.3) Ovaska et al,345 1989

Mayo Clinic, Rochester,MN

Retrospective 1975–1994 113 68 6.5 y (median) 3 (2.6) 3 (2.6) Patients in this study underwentfundoplication; one patientdeveloped HGD

McDonald et al,346 1996

University Hospital,Nottingham, England

Prospective 1976–1990 102 63 54 mo (mean) 4 (3.9) 4 (3.9) 2 patients had dysplasia at baselineand a total of 12 had dysplasia onfollow-up

Iftikhar et al,347 1992

Ninewells Hospital andMedical School, Dundee

Retrospective 1976–1986 44 58 9.5 y (mean) 2 (4.5) 2 (4.5) Patients were not in formalsurveillance program

Rana et al,348 2000

Cleveland Clinic Registry 1979–1995 136 58 4.2 y (mean) 2 (1.4) 2 (1.4) One patient without dysplasia and onewith LGD developed cancer. Fivepatients developed HGD, and 24developed LGD.

O’Connor et al,50 1999

14

78

WA

NG

ETA

LG

AS

TRO

ENTER

OLO

GY

Vol.1

28

,N

o.5

Page 9: American Gastroenterological Association Technical Review on

Table 2 (continued). Diagnosis of Prevalent or Incidental Cancer on Follow-up of Patients in Which the Degree of Dysplasia Was Not Defined or There Were �10Patients in a Single Dysplasia Category

Institute or geographicarea Study type

Studyduration

No. ofpatients

Mean ormedianage (y)

Duration offollow-up

Totalcancers (%)

Incidentalcancers (%) Comments Reference

Academic Medical Center,University of Amsterdam

Prospective — 50 59 5.2 y (mean) 5 (10) 5 (10) 6 patients had LGD and one had HGDat baseline. At the end of the study,10 had LGD and 3 had HGD

Hameeteman et al,349

1989

University of Minnesota,Duluth Clinic

Retrospective 1980–1995 149 — 510 patient-years

20 (13.4) 7 (5) 13 prevalent cancers were detected atesophagectomy

Streitz et al,350 1998

Princess AlexandraHospital, Brisbane,Australia

Prospective 1981–1988 81 63 3.6 y (mean) 3 (3.7) 3 (3.7) 2 patients had HGD at baseline, andboth progressed to adenocarcinoma.Ten patients had LGD, and oneprogressed to adenocarcinoma.

Miros et al,37 1991

VA Medical Center, ArizonaHealth Sciences Center

Prospectivecohort

1982–1995 177 62 4.8 y (mean)or 834patient-years

11 (6.2) 4 (2.3) All patients who progressed to cancerdeveloped dysplasia before thedevelopment of cancer

Drewitz et al,351 1997

University of Leeds,Leeds, England

Retrospective 1984–1995 307 58 72 mo (mean) 12 (3.9) 12 (3.9) Patients were excluded if the cancerwas present at baseline or withinthe first 6 months

Bani-Hani et al,59 2000

VA Cooperative StudyGroup

Retrospective 1986–1999 108 58 1037 patient-years

4 (3.7) 4 (3.7) Patients were not in formalsurveillance program

Spechler et al,125 2001

GOSPE, Torino, Italy Prospectivecohort

1987–1995 187 — 36 mo (mean) 3 (1.6) 3 (1.6) 3 patients had LGD at baseline. 5patients developed LGD, and 2developed HGD

Ferraris et al,352 1997

LGD, low-grade dysplasia; HGD, high-grade dysplasia.

May

20

05

AM

ERIC

AN

GA

STR

OEN

TERO

LOG

ICA

LA

SS

OC

IATIO

N1

47

9

Page 10: American Gastroenterological Association Technical Review on

Table 3. Diagnosis of Prevalent or Incidental Cancer on Follow-up of Patients With Low-Grade Dysplasia

Institute or geographicarea Study type

Studyduration

No. ofpatients

Mean ormedianage (y)

Duration offollow-up Total cancers (%) Incidental cancers (%) Comments Reference

VA Outcomes Group,Dartmouth-Hitchcock MedicalCenter Retrospective 1970–1994 24 63 4.8 y (median) * *

3 patients had HGDor cancer. Cancerand HGD werepooled together

Katz et al,343

1998

Fred HutchinsonCancer ResearchCenter, University ofWashington

Prospectivecohort 1983–1998 43 62

2.4 y (median)or 3.9 y(mean) forthose whodid notdevelopcancer 3 (7) 3 (7)

5- and 8-yearcumulativeincidence of cancerwas reported to be12%

Reid et al,43

2000

Cleveland Clinic Retrospective 1986–1997 25 6711 mo

(median) 2 (8) 2 (8)

An additional 5patients developedHGD. Whenpathologists agreedon the diagnosis ofLGD, 7 of 17progressed to HGDor cancer

Skacel etal,46 2000

VA Medical Centerand University ofKansas MedicalCenter Prospective — 48 62 41 mo (mean)

4 (8); one patienthad definite and 3had suspectedintramucosalcancer

4 (8); one patienthad definite and 3had suspectedintramucosalcancer

4 patients progressedto extensive HGD.12 patients hadpersistent LGD. 31patients regressedto no dysplasiaPatients withprevalent cancerdiagnosed at entrywere excluded

Weston etal,353 2001

*Not defined in manuscript, 24 patients developed LGD anytine during follow up, the high grade dysplasia or cancers were pooled; HGD, high-grade dysplasia; LGD, low-grade dysplasia.

14

80

WA

NG

ETA

LG

AS

TRO

ENTER

OLO

GY

Vol.1

28

,N

o.5

Page 11: American Gastroenterological Association Technical Review on

Table 4. Diagnosis of Prevalent or Incidental Cancer on Follow-up of Patients With High-Grade Dysplasia

Institute or geographicarea Study type

Studyduration

No. ofpatients

Mean ormedianage (y)

Duration offollow-up

Totalcancers (%) Incidental cancers (%) Comments Reference

VA Hospital, Hines, IL Prospectivecohort

1979–1996 75 62 7.3 y (mean) 12 (16) 12 (16) 4 patients in whom cancer wasdetected in the first year ofsurveillance were excludedand considered to haveprevalent cancers

Schnell et al,47

2001

Johns Hopkins Retrospective 1982–1994 30 61 — 13 (43) — Esophagectomy. Nosurveillance

Heitmiller etal,354 1996

Fred HutchinsonCancer ResearchCenter, University ofWashington

Prospectivecohort

1983–1998 76 62 2.4y (median)or 3.9y (mean)for thosewho did notdevelopcancer

33 (43.4) 33 (43.4) 5-year cumulative incidence ofcancer was reported to be59%

Reid et al,43

2000

University ofLouisville,Louisville, KY

Retrospective 1985–1995 11 61 — 8 (72.7) — Esophagectomy. Nosurveillance

Edwards etal,355 1996

VA Medical Centerand University ofKansas MedicalCenter

Prospective — 15 61 37mo (mean)

7 (470; 4patientshaddefinitivecancerand 3 hadpossibleintramucosalcancer)

7 (470; 4 patients haddefinitive cancer and 3 hadpossible intramucosalcancer)

Patients had unifocal HGD. In4 patients, the extent ofdysplasia increased. 5patients becamenondysplastic, and 2 hadLGD. Patients with prevalentcancer diagnosed at entrywere excluded

Weston etal,356 2000

Mayo Clinic,Rochester, MN

Retrospectivecohort

1991–1999 100 67.5 30mo (mean)for focalHGD and15mo (mean)for diffuseHGD

32 (32%) 13 (16) after excludingcancers that were detectedin the first 6 months

Patients with cancer at entrywere excluded. 4 patientswith focal and 28 patientswith diffuse HGD werediagnosed with cancer

Buttar et al,200148

HGD, high-grade dysplasia; LGD, low-grade dysplasia.

May

20

05

AM

ERIC

AN

GA

STR

OEN

TERO

LOG

ICA

LA

SS

OC

IATIO

N1

48

1

Page 12: American Gastroenterological Association Technical Review on

sdpainvetscbawpdwmewluyf

pwe2hwlpn1sod

agshapcviigad

rtwsfrsthoiccup

team4B(foptdthomu(tocncep2ncvro(sbc

1482 WANG ET AL GASTROENTEROLOGY Vol. 128, No. 5

hould have periodic surveillance at 5-year intervals,epending on the patient’s health. The rationale of re-eating surveillance twice in the first year is to diagnoseny prevalent dysplasia that may have been missed dur-ng initial endoscopy. Most initial procedures that diag-ose Barrett’s esophagus do not involve complete sur-eillance biopsies because the diagnosis has not beenstablished. The reason to increase the surveillance in-erval to 5 years is based on decision analysis modelshowing that surveillance in this group of patients is notost-effective. The caveat is that this recommendation isased on set assumptions made in the cost-utility model,nd any major change in these assumptions in the futureill require modification of the surveillance interval inatients with nondysplastic Barrett’s esophagus.38 This isifferent from prior gastrointestinal societal guidelines,hich suggested that surveillance should be performed atore frequent intervals, but there has not been sufficient

vidence to support prior recommendations.64 Therefore,e recommend ending surveillance when the anticipated

ife expectancy is limited (�1 year) or the patient isnable to tolerate any therapeutic measures. Evenounger patients may not warrant surveillance if therapyor cancer cannot be performed.

Patients with low-grade dysplasia should undergo re-eat endoscopy twice in the first year, and those patientsho have persistent low-grade dysplasia in Barrett’s

sophagus should have periodic surveillance at 1- to-year intervals until the age of 80 years, depending onealth status. The risk of cancer development in patientsith low-grade dysplasia is intermediate.46 Patients with

ow-grade dysplasia whose diagnosis is agreed on by 2athologists carry a relatively higher risk of cancer diag-osis; therefore, we recommend a surveillance interval ofyear for this group of patients. Patients whose diagno-

is of low-grade dysplasia is not agreed on by 2 pathol-gists but who do not have any evidence of high-gradeysplasia can be surveyed at 2-year intervals.Patients with high-grade dysplasia in Barrett’s esoph-

gus that has been confirmed by 2 experienced patholo-ists should be recommended to proceed with definitiveurgical or endoscopic management, especially if theigh-grade dysplasia is diffuse or mucosal abnormalitiesre found on endoscopy. If the patient does not wish toroceed with definitive treatment until the diagnosis ofancer is made, surveillance should be considered. Sur-eillance endoscopy should be repeated every 3 monthsn the first 2 years, followed by 6-month intervals indef-nitely. The rationale for recommending immediate sur-ical or endoscopic therapy is that many patients maylready have coexisting esophageal cancer that was not

etected due to the limitations of surveillance.111 The y

ationale for repeating surveillance every 3 months forhe first 2 years is to capture any prevalent cancers thatere not detected initially. This surveillance interval

hould be used even if ablative procedures are performedor high-grade dysplasia or cancer because of the risk ofecurrence of the disease.112,113 The reason to increase theurveillance interval to 6 months after the first 2 years ishat the majority of cancers detected in patients withigh-grade dysplasia are within the first year and the riskf incidental cancer after the first 2 years of surveillances low.47,48 After ablative therapy, surveillance should beontinued as per the preablative recommendation be-ause the long-term consequences of ablation are as yetndetermined and because of reports of recurrent neo-lasia after ablation.Summary of Evidence—The use of surveillance in pa-

ients with Barrett’s esophagus is supported by level IIvidence, although it seems that the most cost-effectivepproach is to target patients at higher risk for develop-ent of cancer. Surveillance should be performed using

-quadrant biopsy specimens taken every 1–2 cm ofarrett’s mucosa, depending on the degree of dysplasia

based on level III evidence). The use of jumbo biopsyorceps in routine surveillance is not recommended basedn existing evidence. The use of jumbo biopsy forceps inatients with high-grade dysplasia has not been proveno provide increased benefit. There are as yet insufficientata to recommend chromoendoscopy with Lugol’s solu-ion over conventional videoendoscopy in Western co-orts at risk for esophageal squamous cell cancer (basedn level III evidence). Methylene blue–assisted chro-oendoscopy is not clearly beneficial and should not be

sed in the routine assessment of Barrett’s esophagusbased on level II evidence). Magnification, optical spec-roscopy, and imaging techniques are still being devel-ped and should be considered research tools at theurrent time (based on level III evidence). Surveillance ofondysplastic Barrett’s mucosa established by 2 endos-opies should be at 5-year intervals (based on level IIIvidence). Surveillance of patients with low-grade dys-lasia established by 2 endoscopies should be at 1- to-year intervals, depending on whether or not the diag-osis is confirmed by 2 pathologists. If both pathologistsoncur that low-grade dysplasia is present, 1-year sur-eillance intervals should be used; if agreement cannot beeached (dysplasia is not believed to be present by at leastne pathologist), 2-year intervals should be sufficientbased on level IV evidence). High-grade dysplasiahould be confirmed in a second endoscopy and reviewedy expert pathologists. If this is confirmed, the patientan be offered surveillance at 3-month intervals for 2

ears and then every 6 months (based on level III evi-
Page 13: American Gastroenterological Association Technical Review on

ditpwe

tgap(opicltsspsdaanpnewaoeweparnbdtmwppm

haipepanMthpopmitdtanddchafthp7B3mgslropptcec

May 2005 AMERICAN GASTROENTEROLOGICAL ASSOCIATION 1483

ence). Any mucosal abnormality should be carefullynvestigated and removed for diagnosis. Surgical resec-ion should be recommended and mucosal ablation withhotodynamic therapy (PDT) can be offered for patientsith confirmed high-grade dysplasia (based on level III

vidence).

Chemoprevention for EsophagealCancer

Acid Inhibition

Because esophageal cancer is uncommon and in-erventions are usually only warranted in high-riskroups, strategies for chemoprevention of this conditionre being advocated. These include the use of acid sup-ression, nonsteroidal anti-inflammatory drugsNSAIDs), and lifestyle modifications. Antireflux surgeryr prolonged acid suppression using high doses of protonump inhibitors result in the appearance of squamousslands in Barrett’s esophageal segment but have notonsistently been shown to regress metaplastic epithe-ium or prevent esophageal adenocarcinoma.114–117 Al-hough the rationale for intensive antireflux therapyeems reasonable, there is not good clinical evidence toupport this approach. The primary rationale for theseroposals is that proton pump inhibitors incompletelyuppress gastric acid and fail to completely reduce duo-enal-esophageal bile reflux.118,119 Acid and bile salts canctivate several key cellular pathways in Barrett’s esoph-gus that are normally associated with progression ofeoplasia in Barrett’s esophagus, and inhibition of theseathways may prevent the development of adenocarci-oma of the esophagus in an animal model of Barrett’ssophagus.120–123 In patients with Barrett’s esophagusho were asymptomatic on acid suppressive therapy and

lso had normalization of intraesophageal acid exposuren pH monitoring, there was significantly more differ-ntiation and less proliferation compared with patientsho continued to have abnormal intraesophageal acid

xposure.124 However, even on high dosages of protonump inhibitors, acid inhibition may be difficult tochieve, with 4 of 25 patients having pathologic acideflux on 80 mg/day of omeprazole.119 Fundoplication isot successful in controlling acid reflux on a long-termasis and did not prevent development of cancer, asemonstrated by a follow-up to a randomized prospectiverial comparing medical with surgical therapy.125 Epide-iologic studies involving more than 66,000 patientsith reflux treated medically compared with 11,000atients treated with fundoplication in Sweden found norotective effect of surgery in decreasing the develop-

ent of adenocarcinoma of the esophagus.126 c

Ablation Therapy

Ablation therapy in conjunction with acid controlas been promoted for the treatment of Barrett’s esoph-gus to decrease the risk for esophageal cancer.127,128 Thiss based on the observation that elimination of the meta-lastic epithelium can result in squamous mucosa regen-ration.129 Multiple ablation therapies have been pro-osed and involve the use of multipolar coagulation,rgon plasma coagulation, neodymium:yttrium-alumi-um-garnet (Nd:YAG) laser coagulation, and PDT.ultipolar coagulation has been performed in a prospec-

ive multicenter study with 58 patients that eliminatedistologic evidence of Barrett’s mucosa in 78% of treatedatients.130 Argon plasma coagulation has been used inne randomized, nonblinded, prospective trial in 40atients after fundoplication for acid control.131 Barrett’sucosa was initially found underneath squamous mucosa

n this study in more than 35% of patients 1 month afterreatment but decreased to 5% at 1 year. Patients whoid not achieve complete ablation had 95% regression inhe amount of Barrett’s mucosa. Multiple case series ofrgon plasma coagulation have been reported with sig-ificant reduction in Barrett’s mucosa and decrease inysplasia.132–139 Many ablation trials emphasize that re-uction in the length or area of Barrett’s mucosa shouldorrespond with a decreased risk for cancer, although thisas not been shown in a prospective study. PDT has beenpproved by the Food and Drug Administration (FDA)or use in patients with high-grade dysplasia to decreasehe risk of cancer formation. Case series of patients withigh-grade dysplasia treated with PDT using sodiumorfimer have found that dysplasia can be eliminated in8% of patients with elimination of 75%–80% of thearrett’s mucosa, but esophageal strictures occurred in4% of patients.140 Unfortunately, the residual Barrett’sucosa after ablation therapy has been found to contain

enetic mutations similar to those found before ablation,uggesting that histologic improvement may not corre-ate with elimination of cancer risk.113 A prospectiveandomized trial studied PDT using 2 different dosagesf 5-aminolevulinic acid (a less potent agent than sodiumorfimer) followed by argon plasma coagulation com-ared with argon plasma coagulation alone in 40 pa-ients.141 These investigators found that argon plasmaoagulation alone or in combination with PDT was onlyffective in eliminating about 70% of the Barrett’s mu-osa.

NSAIDs

The rationale for the use of NSAIDs to prevent

ancer in Barrett’s esophagus is based on experimental
Page 14: American Gastroenterological Association Technical Review on

atcdpawwpenooaBsn

pCaulhecewcfttcpgcpe

cifcgbacoap

ar1l17ctb

deflttaoaeNsbmtele

ldpuphmoCat

flaelHe

1484 WANG ET AL GASTROENTEROLOGY Vol. 128, No. 5

nd epidemiologic evidence. No randomized controlledrials have found aspirin to be effective for prevention ofancer. It has been shown that bile salts in a pH-ependent manner can activate the arachidonic acidathway and may promote neoplasia in Barrett’s esoph-gus.121,122 The activation of the arachidonic acid path-ay leads to increased production of prostaglandin E2,hich increases cellular proliferation.142,143 Inhibition ofrostaglandin E2 normalizes proliferation in Barrett’spithelial cells and decreases it to a level that is seen inormal squamous epithelium.143 Inhibition of the cyclo-xygenase enzyme with selective and nonselective cyclo-xygenase inhibitors has been shown to reduce the risk ofdenocarcinoma of the esophagus in an animal model ofarrett’s esophagus.123 Finally, cyclooxygeanse-2 expres-

ion has also been shown to parallel the progression ofeoplasia in human Barrett’s esophagus.144

Epidemiologic studies have found that NSAIDsrotect against the risk of esophageal cancer.145–149

ase-controlled studies have shown a 36%–90% rel-tive risk reduction for esophageal cancer in patientssing aspirin or other NSAIDs occasionally or on aong-term basis.145–147,149 A decision analysis modelas been used to evaluate the feasibility and cost-ffectiveness of NSAIDs to prevent esophageal adeno-arcinoma. It suggested that the high incidence ofsophageal adenocarcinoma in Barrett’s esophagusith high-grade dysplasia renders chemoprevention a

ost-effective option.150 NSAIDs were not a cost-ef-ective measure in the general population of all pa-ients with Barrett’s esophagus, but this was sensitiveo variations in the cost of chemoprevention, the in-idence of cancer, and the efficacy of the NSAID inrevention of cancer. In patients with gastroesopha-eal reflux, a chemopreventive approach will only beost-effective if the agents are very safe and effective torevent the development of adenocarcinoma of thesophagus.

Lifestyle

Lifestyle factors for the development of esophagealancer have been identified through epidemiologic stud-es. Obesity seems to be one of the most important riskactors associated with esophageal cancer.151,152 In a re-ently completed study of patients with esophageal andastric cancer, smoking and dietary habits were found toe important risk factors.152 Smoking was found to havepopulation attributable risk (PAR) of 39.7% (95%

onfidence interval [CI], 25.6%–55.8%). The frequencyf gastroesophageal reflux disease symptoms also showedpositive trend in the PAR, with symptoms at least once

er day accounting for almost one half of the PAR n

ssociated with the presence of any gastroesophagealeflux disease symptoms (29.7%; 95% CI,9.5%–42.3%). Consumption of fruits and vegetablesess than twice a day on average had a modest PAR of5.3% (95% CI, 5.8%–34.6%). In this population,8.7% (95% CI, 66.5%–87.3%) of esophageal adeno-arcinoma cases could be attributed to one or more ofhese well-established risk factors, with smoking andody mass index contributing the most.Summary of Evidence—Based on level II evidence, fun-

oplication should not be recommended solely to preventsophageal cancer in patients with gastroesophageal re-ux disease. The use of high-dose proton pump inhibi-ors to prevent esophageal cancers in patients with gas-roesophageal reflux disease has also not been establishednd cannot be recommended in clinical practice (basedn level III evidence). Level II evidence suggests thatblation therapy may decrease dysplasia in Barrett’ssophagus and could potentially decrease cancer risk.SAIDs should not be recommended at the current time

olely for the prevention of esophageal adenocarcinomaut have promise as a chemopreventative agent, whichight be beneficial if required for other medical indica-

ions (based on level II evidence). Patients at risk forsophageal cancer should be counseled to adapt healthierifestyles, in particular attaining normal weight andliminating tobacco use (based on level II evidence).

Diagnosis and Staging ofEsophageal Cancer

Diagnosis

Most patients with esophageal cancer present at aate stage when the diagnosis of cancer is made, withysphagia as the cardinal symptom.153 In particular,ersistent dysphagia that progresses from solids to liq-ids should heighten suspicion for esophageal cancer andrompt an endoscopic evaluation. Up to 75% of patientsave experienced anorexia and weight loss when seekingedical attention. Patients may also present with

dynophagia, chest pain, or gastrointestinal bleeding.ough aggravated by swallowing raises the possibility ofn esophagopulmonary fistula, a devastating complica-ion associated with a high 30-day mortality rate.154

The diagnosis of esophageal cancer is established byexible endoscopy with biopsy. The features, location,nd size of the tumor can be more accurately assessed byndoscopy than by radiographic studies.155 Barium swal-ow as an initial diagnostic test is of limited value.156,157

owever, it may be useful to confirm the presence ofsophagopulmonary fistulas or document complete lumi-

al obstruction when clinically suspected. In patients
Page 15: American Gastroenterological Association Technical Review on

wqttt7

tsfamfhmtouddsb

gcbsamiatcb

a2iiwdwagc

aumttsOd(nodwttinpccl

mpas

May 2005 AMERICAN GASTROENTEROLOGICAL ASSOCIATION 1485

ith advanced cancers, esophageal dilation may be re-uired to allow for a standard (OD, 9.8 mm) endoscopeo traverse the obstructed lumen. Alternatively, an ul-rathin (OD, 5.3–6 mm) endoscope may pass throughhe stenosis and allow completion of the examination in5% of cases.158

Biopsy specimens are required for histologic confirma-ion. The diagnostic yield reaches 100% when 6 or moreamples are obtained using a standard endoscopic biopsyorceps.159,160 Biopsy specimens of necrotic or fibroticreas should be avoided. The adequacy of biopsy speci-ens obtained via ultrathin endoscopes has not been

ormally assessed. As an adjunct, brush cytology can beelpful in sampling tight malignant strictures, whichay not be easily accessible by conventional biopsy

echniques.161 Brushings should be collected before bi-psy to maximize the diagnostic yield.162 Endoscopicltrasonography (EUS) should be considered when stan-ard biopsy and/or brush cytology fail to confirm theiagnosis in the setting of high clinical suspicion (e.g.,ubmucosal tumors).163 Fine-needle aspiration or Tru-cutiopsy can be performed at the time of EUS.

Staging

The staging of esophageal cancer is critical touide further therapy for the patient. Patients with can-er confined to the mucosa or superficial submucosa cane treated using surgical resection or potentially endo-copic therapy.137,164 However, patients who have moredvanced disease will require surgical resection or che-oradiation.165,166 None of the currently available stag-

ng technologies have been shown to be able to stage allspects of the tumor. The selection of the best stagingests depends on the probability of detecting metastaticancer. Although endoscopy is not a staging tool, it can

Figure 1. Algorithm for staging of advanced cancers.

e helpful to guide the gastroenterologist toward which

dditional staging studies are needed. In one series of09 patients, endoscopy was 89% accurate in determin-ng tumor staging compared with surgical or EUS stag-ng.167 Patients with large bulky circumferential tumorsho present with dysphagia are likely to have advancedisease and will require tests to confirm this. Patientsho have cancers that are �2 cm in diameter and are

symptomatic are more likely to have early disease. Al-orithms for evaluation of advanced and early esophagealancers are found in Figures 1 and 2.

The most commonly used staging procedure in esoph-geal cancer is computed tomography (CT). The partic-lar strength of this test is the ability to detect distantetastatic disease.168 The primary reason this is used as

he first staging study is because of its availability andhe change in the treatment course that would occurhould metastasis or invasion of other organs be detected.n initial presentation, most patients with metastaticisease had involvement of abdominal lymph nodes45%), liver (35%), lung (20%), and cervical lymphodes (18%) based on findings at esophagectomy.169 CTf the chest and abdomen correctly identified metastaticisease in only 69% of the cases. In this series, patientsithout metastasis on CT did not have involvement of

he bones or brain. CT has limited accuracy in staginghe extent of tumor invasion, with correct staging foundn 50%–90%.170–172 CT has been shown to produce falseegatives in 30%–60% of patients.173 Even with im-rovements in technology such as the helical CT scanner,omparisons with EUS and fine-needle aspiration indi-ate that it is still not as sensitive for detecting celiacymph node disease or T4 carcinoma.174

EUS consists of 3 fundamentally different devices thatay not be available to all gastroenterologists. There are

robes that can be placed within the endoscope or useds stand-alone instruments, radial scanning echoendo-copes, and linear array echoendoscopes that permit fine-

Figure 2. Algorithm for staging of early cancers.

Page 16: American Gastroenterological Association Technical Review on

nmhrMuiTmpup97baTbSinewTahcdcctctdcb

tnimHtfnsTcpPaa

aas

iiposptbPntrisfscaratvsarpa

iospCsibeEeh

mT

1486 WANG ET AL GASTROENTEROLOGY Vol. 128, No. 5

eedle aspiration. EUS can be directed primarily at theucosa with the use of probes that usually involve

igh-frequency scanning at 20–30 MHz or can be di-ected to more distant structures by scanning at 7.5–12Hz with the radial echoendoscopes. The diagnostic

tility of EUS is in determining the depth of tumornvasion and the presence of local-regional adenopathy.his technology is limited in terms of detecting distantetastasis because visualization of other organ systems,

articularly the lung, is not possible because air disruptsltrasound conduction. Identification of abnormal-ap-earing lymph nodes has been found to be specific (85%–7%) for nodal metastasis but not very sensitive (72%–7%) in case series.175,176 If abnormalities are found,iopsies can be performed on the lesions using a smallspiration needle positioned in a linear array instrument.he sensitivity of this technique in tumor staging haseen reported in clinical series between 59% and 90%.ome of this variation can be attributed to the difficultyn passing the ultrasound instrument through a malig-ant stricture.177 Histologic confirmation of nodal dis-ase altered planned therapy in only 13% of patientsith esophageal cancer in one retrospective case series.178

his may be because the majority of esophageal cancersre found at an advanced stage. Miniprobes that haveigher resolution are the best at defining early-stageancer.179 Decision analysis models have been used toetermine the value of EUS in assessment of esophagealancer. These models have found that EUS is a moreost-effective initial staging strategy if the probabilityhat EUS can find advanced disease is �30% or if theost of EUS is less than 3.5 times that of CT.180 Addi-ional analysis based on the use of EUS in a nationalatabase found that preoperative use of this techniqueould prevent 26% of patients from unnecessary com-ined-modality therapy.181

Positron emission tomography (PET) is a technologyhat uses 18F-fluorodeoxyglucose for the detection ofodal or distant metastasis in esophageal cancer.182 Thiss used to detect neoplastic tissues because they normallyetabolize glucose at a faster rate than normal tissues.owever, inflammatory tissues are also fast glucose me-

abolizers and metastases often have to be differentiatedrom inflamed tissue, leading to false positives.183 Aumber of case series have found that PET is not asensitive as EUS or CT for locoregional disease.184–189

his technology cannot define the tumor stage because itannot resolve the layers of the esophagus. In addition,atients with hyperglycemia are not good candidates forET. Because of these factors, PET cannot be envisioneds an initial staging tool in esophageal cancer. New

dvances in PET technology include fusion PET, which C

ctually combines the CT image with the PET image tollow better tumor localization. This may increase thepecificity of the test.

Other staging procedures that have been investigatednclude minimally invasive surgery or laparoscopic stag-ng before esophagectomy. Case series have found thaterformance of laparoscopic- or thoracoscopic-guided bi-psies of lymph nodes increases the detection of meta-tatic disease and can change management in 17% ofatients.171,190–196 Thoracoscopy has a significant advan-age in detecting thoracic lymph nodes and when com-ined with laparoscopy appears to be superior to EUS,ET, and CT staging.171,192,197 These series generally doot have uniform staging procedures performed beforehe laparoscopy, which makes it difficult to assess theole of these procedures in relation to the standard stag-ng modalities. Bronchoscopy has been suggested as ataging procedure for patients with tumors that areound above the tracheal bifurcation. In one prospectivetudy, almost 10% of patients were found to have tra-heal involvement on the basis of bronchoscopy alone,lthough EUS was not used in this study.198 Magneticesonance endoscopy has also been used in staging esoph-geal cancer but as yet has not been shown to be superioro existing tests.199,200 Recent cost analyses of thesearious staging modalities alone and in combination haveuggested that CT combined with EUS with fine-needlespiration if appropriate offered the least expensive andeasonable efficacy in terms of QALYs.201 PET and EUSlus fine-needle aspiration offered more QALYs but waslso more expensive at $60,544 per QALY.

Summary of Evidence—Given the state of the currentnformation, we recommend the following strategy. CTf the chest and upper abdomen should be the firsttaging procedure, followed by EUS and fine-needle as-iration if no evidence of distant metastasis is found onT and the procedure is available. If surgical resection is

till considered, PET can be considered if available due tots increased sensitivity for distant metastasis. This isased on level III evidence. In patients with potentiallyarly-stage disease (tumors �2 cm and nonobstructing),US with endoscopic mucosal resection may be consid-red as an alternative staging procedure if available foristologic staging of the cancer and potential therapy.

Treatment of Esophageal Cancer

Treatment of Early Cancers

Early esophageal cancers, those confined in theucosa or upper submucosa of the esophagus, are termed1, N0, M0 by the American Joint Commission on

ancer terminology (see Table 5). There have not been
Page 17: American Gastroenterological Association Technical Review on

atdfTiT(osmeriJititewh

camwmcw7vtmf

rpOtHTf2tmdtr

nAgcbywi5i9mtamfttitl

p

T

T

N

M

S

May 2005 AMERICAN GASTROENTEROLOGICAL ASSOCIATION 1487

ny randomized treatment trials for these cancers becausehey are rare, accounting for �5% of esophageal cancersiagnosed in most series. In Japan, there has been aurther division in T1 lesions. An early cancer is termed1m if confined to the mucosa and T1sm if submucosal

nvasion is found. There has even been further division of1sm lesions into penetration into the upper third

sm1), the middle third (sm2), or the lower third (sm3)f the submucosa.202 In the Japanese experience withquamous cell cancers, penetration of the superficial sub-ucosa is associated with a 6% risk of metastasis. How-

ver, if deeper penetration of the submucosa is found, theisk of metastasis increased to 47%. There is a problemn the interpretation of some of the literature fromapan.203 Overall, Japanese pathologists will diagnosentramucosal carcinoma based primarily on patterns ofhe cell nucleus rather than on the demonstration ofnvasion as required by Western pathologists. Some ofhe data concerning the lack of metastatic potential forarly cancers may be biased by the inclusion of cases ofhat would be classified by Western pathologists as

able 5. Staging According to the American JointCommission on Cancer

umor staging for esophageal cancerT0 No evidence of primary tumorTis Carcinoma in situT1 Tumor invades lamina propria or submucosaT2 Tumor invades muscularis propriaT3 Tumor invades adventitiaT4 Tumor invades adjacent structures

odal stagingN0 No evidence of lymph nodesN1 Evidence of regional lymph nodesetastasisM0 No evidence of distant metastasisM1 Evidence of distant metastasisM1a For lesions in the lower thorax indicates metastasis to the

celiac nodesM1a For lesions in the upper thorax indicates cervical lymph

nodesM1b For lesions in the midthorax indicates nonregional or other

nodal groupsM1b For lower esophagus or upper thorax lesions indicates

distant metastasisX designation in any area indicates that the lesion was unable to

be assessed.taging of esophageal cancersStage 0 Tis,N0,M0Stage 1 T1,N0,M0Stage 2a T2-3,N0,M0Stage 2b T1-2,N1,M0Stage 3T3, N1,M0

T4, any N,M0Stage 4 Any T, any N, M1Stage 4a Any T, any N, M1aStage 4b Any T, any N, M1b

igh-grade or even low-grade dysplasia. p

The traditional approach for these squamous cell can-ers has been surgical resection because cure can bechieved in �90% of T1m cancers.204,205 A survey ofajor medical centers in Europe found that 253 patientsith early cancers treated with esophagectomy had aortality rate of 9.1%.205 Patients with intraepithelial

ancers had a 5-year survival rate of 93%, while thoseith intramucosal cancers had a decreased survival rate of3%. If cancer progressed into the submucosa, the sur-ival rate further decreased to 44%. Twenty of 21 pa-ients with recurrent disease had submucosal involve-ent with cancer. These results are similar to those

ound in Japan.There is less information available regarding surgical

esection of early adenocarcinoma. Early cancers haverimarily been reported as part of larger surgical series.verall, limited reports have results with a 100% rate of

otal excision without any operative mortality.206–211

owever, this is likely to be influenced by reporting bias.he reported mortality rate for esophagectomy per-

ormed on patients with high-grade dysplasia is between% and 6%.212,213 The major concern with surgicalherapy for high-grade dysplasia is the 40% incidence oforbidity associated with the procedure. Possible proce-

ure-related complications include anastomotic stric-ures, leaks, chronic aspiration, infection, and chylotho-ax.

Endoscopic mucosal resection has been performed on aumber of patients with early cancers, particularly insia, where the technique has been established for earlyastric cancers. Studies have shown that squamous cellancers confined to the mucosa or upper submucosa coulde treated successfully with endoscopic therapy.214 Five-ear cure rates for patients with intramucosal cancersere 100%, while those who had cancers that penetrated

nto the deep submucosa had 5-year survival rates of9%–54% in a series of 152 cases. Reports from othernstitutions for superficial cancers in Japan reported a5%–100% 5-year survival rate with recurrences esti-ated at 3%–7%. Even these recurrences were believed

o be treatable with additional endoscopic therapy.215 Insurvey of 143 Japanese medical institutions, endoscopicucosal resection was regarded as the treatment of choice

or intramucosal cancers.216 Complications in 2418 pa-ients were primarily stenosis, hemorrhage, or perfora-ion, which were found in 7% of patients. The mostmportant predictors of stenosis were resection of morehan three fourths of the circumference of the esophagealumen or if the lesion was �3 cm in length.217

Early mucosal resections were performed using a trans-arent overtube through which an endoscope could be

ositioned.218 The lesion was first identified visually, and
Page 18: American Gastroenterological Association Technical Review on

titrbbNtukrtTldSasiosdsttws

sTmPahiaomBcaeauytBmh

aT

eshctofwathcutEcplot

fitts1sotmahi

wrutceiracevpb

1488 WANG ET AL GASTROENTEROLOGY Vol. 128, No. 5

hen a fluid cushion was created under the lesion bynjecting saline and epinephrine submucosally beneathhe lesion. If the lesion could not be elevated from theemainder of the esophageal wall, the resection could note performed. Later developments involved the use ofand ligation and eventually the cap-fitted endoscope.219

ew variations of this technique have been described andermed endoscopic submucosal dissection, including these of an insulated tipped electrocautery knife, needlenife, or triangular tipped knife that can completelyesect the lesion with the use of a special snare designedo resect the lesion without any need for suction.220,221

hese techniques were all designed for the relatively flatesions associated with early squamous cell cancers. En-oscopic mucosal resection performed in the Unitedtates involves the use of variceal band ligation, whichllows the formation of a pseudopolyp that can be re-ected by a regular snare. A second common techniquenvolves the use of a specialized cap that can be fittednto an endoscope. This requires the use of a crescentnare that is designed to form a loop around the lip of theistal end of the cap. The targeted tissue can then beuctioned into the cap and the snare closed to performhe resection. A prospective comparison study of these 2echniques in 100 resections performed in 72 patientsith early cancers did not find any differences in terms of

ize of the specimen or complication rates.222

Early esophageal adenocarcinomas have been endo-copically treated in the setting of Barrett’s esophagus.hese have been described in large case series usingucosal resection alone or in combination withDT.223,224 Lesions most amenable to mucosal resectionre those that are polypoid, elevated, �2 cm in size, andaving a low-grade cancer.223 Mucosal resection was usedn 115 patients (83% with early cancers) and resulted in3-year overall survival rate of 88%.137 Complications

ccurred in 9% of patients, including decreases in he-oglobin and stricture formation. However, becausearrett’s esophagus without high-grade dysplasia or can-er was left untreated, 30% of the patients developeddditional neoplastic lesions. For this reason, it is gen-rally advised that the remainder of the Barrett’s esoph-gus be treated after resection of a cancer. PDT has beensed for this purpose, with 94% initial success rates at 1ear.224 Although not widely performed, mucosal resec-ion has been performed to completely eliminate thearrett’s segment in 2 reports.225,226 Circumferentialucosal resection does tend to generate strictures; this

as been reported in 3 of the 13 reported cases.Endoscopic therapy using PDT, laser therapy, and

rgon plasma coagulation has also been reported.227–233

hese treatments have had limited success in completely r

liminating Barrett’s esophagus and cancer, with re-ponse rates of 25%–94%. These small case reports havead a wide variation in the depth of invasion of theseancers (T0–T2) and the type of therapy delivered. Laserherapy with Nd:YAG, which operates at a wavelengthf 1063 nm, penetrates quite deeply and has been favoredor tumor ablation. Lasers that operate in the 540-nmavelength, such as the argon or KTP-YAG lasers, havemuch more limited depth of penetration. Thermal

herapies, such as argon plasma coagulation applied atigh-power settings, have also been used to treat earlyancers. PDT is influenced by the type of photosensitizersed for therapy. Aminolevulinic acid is not approved inhe United States at this time, but it has found favor inurope because it is not associated with the prolongedutaneous photosensitivity that is found with the sodiumorfimer that is used in the United States. Due to its veryimited depth of penetration, aminolevulinic acid hasnly been found to be useful for cancer therapy if thehickness of the cancer is �2 mm.230

Other treatments that have been reported in case seriesor the treatment of patients with superficial cancersnclude radiation therapy and brachytherapy. Radiationherapy has been used in Japan as a single-modalityherapy for superficial squamous cell cancers. Five-yearurvival rates after treatment in 2 series with a total of83 patients ranged from 39% to 45%.234,235 Thesetudies have also shown a trend toward nodal recurrencef cancer in patients who have had submucosal penetra-ion.234 Patients with cancer strictly confined to theucosa did not have nodal recurrence. Brachytherapy

lone and in combination with external beam radiationas also been reported, although the 3-year survival raten these patients is only 14%.236

Treatment of Advanced Cancers

The gastroenterologist is often asked to determinehether there is a role for oncologists before surgical

esection. There has been substantial controversy over these of neoadjuvant therapy before esophagectomy for thereatment of patients with locally advanced esophagealarcinoma, primarily patients with stage IIb or III dis-ase (Table 5). Primary surgical therapy for cancers lim-ted to the esophagus, stage I or IIa disease, has had goodesults without the need for or morbidity of chemother-py.205,208,237,238 Surgical therapy for more advanced can-er has been performed as either a limited resection orn-bloc removal of lymph nodes, which can be performedia a transhiatal or transthoracic (Ivor–Lewis) ap-roach.239,240 Extensive resection of lymph nodes haseen more commonly practiced in Asia and involves

emoving nodes in the neck, chest, and abdomen in a
Page 19: American Gastroenterological Association Technical Review on

3popsaatfcsssfessisi

dcrgaeAhlstcabbmcvh

ottcspc

awppgathihactdh

tsGdtpttT0ta

cs

T

NLAWBKU

May 2005 AMERICAN GASTROENTEROLOGICAL ASSOCIATION 1489

-field lymph node dissection.241 This procedure is rarelyracticed in the United States because of the prolongedperating times and the morbidity associated with therocedure. An en-bloc lymph node dissection of thetomach and chest is practiced in the United States in anttempt to achieve complete surgical removal of tumornd appears in small series to eliminate recurrence ofumor in the resected area.242 In general, esophagectomyor squamous cell cancer seems to have been more suc-essful in Asia than in Western countries in terms ofurvival and operative mortality.243,244 Minimally inva-ive esophagectomy has recently been promoted for re-ection of esophageal cancer with the use of laparoscopyor gastric mobilization and resection.245,246 The gastro-sophageal anastomosis is accomplished with a thoraco-cope or manually. Although the procedure has not beenhown to improve patient outcomes in terms of morbid-ty or mortality, it has been attractive to patients withuperficial cancer or high-grade dysplasia who desire anmproved cosmetic result.

It has been well recognized that survival is related toisease stage.240,247 Thus, it is not surprising that theoncept of neoadjuvant (preoperative) chemotherapy andadiation had significant appeal to oncologists and sur-eons. By reducing the number of involved lymph nodesnd decreasing cancer stage, neoadjuvant therapy couldnhance the ability of surgical resection to cure patients.

number of prospective, randomized, controlled trialsave investigated the use of neoadjuvant therapy fol-owed by surgery versus surgery alone, and these areummarized in Table 6.248–254 Only one of these studies,hat by Walsh et al., actually showed an advantage tohemotherapy and radiation with an odds ratio of 8.44nd a 95% CI that does not cross 1.251 This study haseen criticized for its high surgical mortality rate thatiases against the surgical therapy alone group. A recenteta-analysis combined all of these studies and con-

luded that neoadjuvant therapy increased patient sur-ival at 3 years with an odds ratio of 2.5 (P � .04) and

able 6. Summary of Randomized Controlled Trials ComparinSurgical Resection With Surgical Resection Alone

ReferenceNo. of

patientsSurvival

period (y)Surgery

alone (%)

ygaard et al248 186 3 9e Prise et al249 86 3 13.8pinop et al250 69 5 10alsh et al251 113 3 6osset et al252 282 5 32elsen et al253 440 2 35rba et al254 100 3 16

ad a decreased risk of locoregional recurrence with an c

dds ratio of 0.83 (P � .01).255 This study found thathere was a nonsignificant trend toward an increase inreatment mortality. The concurrent administration ofhemotherapy and radiation therapy was believed to beignificantly better than sequential therapy. A significantercentage (21%) of the patients in these series had aomplete pathologic response at the time of resection.

With these high pathologic response rates with radi-tion and chemotherapy alone, it has been questionedhether surgical therapy is needed in the treatment ofatients with more advanced cancers. Initial studies com-aring neoadjuvant regimens found that the smallroups of patients who refused surgery after chemother-py and radiation had a 5-year survival rate of 18% ifhey had squamous cancers but no 5-year survival if theyad adenocarcinomas.256 The Intergroup 0123 trial stud-ed a nonsurgical approach to esophageal cancer usingigh-dose radiation (64.8 Gy) versus standard-dose radi-tion (50.4 Gy) in combination with 5-fluorouracil andis-platinum in 216 evaluable patients.257 The trial waserminated after interim analysis because the results in-icated that 50.5-Gy radiation was as effective as theigher dose, with 2-year survival rates of 40%.The need for combined chemotherapy and radiation

herapy has been best established in a randomized pro-pective trial from the Radiation Therapy Oncologyroup 85-01 study, which examined 134 patients ran-omized to radiation alone versus radiation in combina-ion with chemotherapy with 5-fluorouracil and cis-latinum.258 This trial clearly established that combinedherapy with a 5-year survival rate of 26% was superioro radiation therapy alone with a 0% 5-year survival rate.he odds ratio of this study was 0.02 (95% CI, 0.00–.38). One caveat from this study was that only 68% ofhe patients who planned to undergo chemotherapy wereble to complete the treatment course.

Summary of Evidence—Patients with early esophagealancers confined to the mucosa should be treated withurgical resection, with consideration of endoscopic mu-

oadjuvant Chemotherapy and Radiation in Addition to

Chemoradiationand surgery (%) P value

Oddsratio

95% Confidenceinterval

17 .30 2.81 0.61–12.9519.2 .56 1.57 0.50–5.0024 .40 3.1 0.74–12.832 .01 8.44 2.33–30.5733 .78 1.06 0.64–1.7437 .53 1.1 0.74–1.6330 .15 2.25 0.85–5.93

g Ne

osal resection with adjuvant mucosal treatment for any

Page 20: American Gastroenterological Association Technical Review on

r(ass(Ialwr(vat

vsuTdPota

ptedaav

tlflidtcoimftc

h

bispImiapes1daerf0badm

Ilnbhmhe

tdu(ccflamrsevtsutts

1490 WANG ET AL GASTROENTEROLOGY Vol. 128, No. 5

emaining preneoplastic tissue (ie, Barrett’s esophagus)based on level III evidence). Patients with early esoph-geal cancers that penetrate into the upper third of theubmucosa can be treated with endoscopic mucosal re-ection if surgical mortality is anticipated to be �6%based on level III evidence). Patients with stage IIb andII disease may benefit from concomitant chemotherapynd radiation therapy before surgical therapy (based onevel II evidence). Patients with stage I and IIa diseaseho are good candidates for surgical therapy do not

equire neoadjuvant therapy before esophagectomybased on level II evidence). Patients with more ad-anced-stage cancer may be treated with chemotherapynd radiation therapy or be considered for palliativeherapy.

Palliation of Esophageal Cancer

Esophageal cancer is usually diagnosed at an ad-anced and incurable stage. Patients with locally unre-ectable cancer or patients who are surgically unfit mayndergo palliation by a variety of nonoperative means.he goals of palliation in this setting are to alleviateysphagia, aid in nutrition, and improve quality of life.alliation can be achieved by external beam radiotherapyr intraluminal brachytherapy, with or without chemo-herapy. However, patients may not tolerate these ther-pies or improvement in dysphagia may be slow.259

Endoscopic therapy plays an important role in thealliation of malignant dysphagia. Several endoscopicechniques have been applied, but limited informationxists as to the optimal approach. The choice is primarilyictated by tumor characteristics, patient preference, andvailable expertise. Treatment must be individualized,nd different methods of palliation may be appropriate atarious stages of the patient’s illness.

Malignant esophageal stenoses can be dilated usinghrough-the-scope balloons or wire-guided polyvinyl di-ators (e.g., Savary dilators), with or without the aid ofuoroscopy. Dilation can be complicated by perforationn up to 10% of cases.260–262 Blind passage of Maloneyilators is not recommended in complex malignant stric-ures due to a higher risk of perforation.263 Most patientsan be dilated to a luminal diameter that allows passagef a liquid to soft diet (�9–12 mm), but improvements brief and typically measured in days. Dilation is usedostly as an adjunct to other modalities, such as in

acilitating placement of an esophageal stent or in pa-ients awaiting improvement in swallowing followinghemoradiation therapy.264

Among endoscopic modalities, esophageal stenting

as assumed a primary role as a palliative technique s

ecause of its effectiveness in rapidly relieving dysphagian one procedure. In the United States and abroad,elf-expanding metal stents (SEMS) have largely sup-lanted conventional plastic semirigid prostheses.265,266

n contrast to plastic semirigid stents, SEMS requireinimal to no preplacement dilation and are easier to

nsert.267–269 SEMS also expand the obstructed lumen togreater degree (18–23 mm) than that provided by

lastic semirigid stents bounded by fixed internal diam-ters (10–12 mm). Both SEMS and plastic semirigidtents have a highly successful rate of insertion (90%–00%). They are also similarly effective at alleviatingysphagia in �90% of patients; most are able to toleratet least liquids following stent placement.270–272 How-ver, the collective experience gathered from prospectiveandomized trials has shown SEMS to be associated withewer procedure-related complications (10%–43% vs%–16%), shorter hospital stays, and trends towardetter quality of life and survival.269–274 Although SEMSre more expensive than plastic semirigid stents, this costifference was not found to be significant in the overallanagement of dysphagia.274

A self-expanding plastic stent (Polyflex; Boston Scientificnc, Natick, MA) has been approved by the FDA for pal-iation of malignant dysphagia and, more recently, for be-ign disease. The potential advantage is its removability,ut it is priced similar to available SEMS. Initial studiesave shown the Polyflex stent to be efficacious in relievingalignant dysphagia.275–278 Prospective randomized trials,

owever, are needed to assess the cost-effectiveness of self-xpanding plastic stents versus SEMS.

Three FDA-approved SEMS are currently available inhe United States. These SEMS differ in design andelivery systems, with additional refinements for partic-lar indications (Table 7). The EsophaCoil stentMedtronic InStent Inc, Eden Prairie, MN) is no longerommercially available. Endoscopic placement of SEMSan be performed with or without the assistance ofuoroscopy.279,280 The majority of SEMS currently in usere partially covered with a thin silicone or polyurethaneembrane to prevent tumor ingrowth. In a prospective

andomized study of 62 patients, membrane-coveredtents offered significantly better palliation than uncov-red stents due to decreased rates of tumor ingrowth (3%s 30%) and decreased need for endoscopic reinterven-ions (0% vs 27%) for recurrent dysphagia.281 Coveredtents, however, tend to migrate more frequently thanncovered ones (26% vs 0%), particularly when placed inhe distal esophagus.282 Modifications in design, such ashe conical-shaped Flamingo Wallstent and Ultraflextent with proximal flaring, have improved anchoring of

tents to tissue and have made covered SEMS more
Page 21: American Gastroenterological Association Technical Review on

msw

cdiwthotiadarfa

wrs1sto1ati(t5fi(l

ptiaau

mpslpnpsps1oimafbtbd

Fibttat

T

MDCRDLFSDF

Aa

b

May 2005 AMERICAN GASTROENTEROLOGICAL ASSOCIATION 1491

igration resistant.283,284 These 2 stents have been useduccessfully for palliation of distal esophageal cancersith an overall migration rate of 6%.284

There are limited prospective randomized studiesomparing various SEMS for palliation of malignantysphagia.284,285 In a study of 100 patients, success ratesn alleviating dysphagia were similar among patientsho received the Z-stents, Flamingo Wallstents, or Ul-

raflex stents.285 Although complication rates wereigher in the Z-stents group (36%) than in the Ultraflexr Flamingo Wallstent groups (24% and 18%, respec-ively), these differences did not reach statistical signif-cance. In a study of 53 patients, Flamingo Wallstentsnd Ultraflex stents were equally effective at palliatingysphagia with similar complication rates.284 There ares yet insufficient data that convincingly show the supe-iority of one SEMS over another. Currently, the choiceor a particular SEMS primarily depends on device avail-bility, familiarity, and personal preference.286

Types and rates of SEMS-related complications varyidely among studies. Results of a national survey and

eview of complications from compiled case serieshowed immediate technical complication rates of 5%–7%, including misplacement (0.3%–5%), failed expan-ion (4%–7%), failed deployment (1%–3%), and migra-ion (0.3%–2%).268 Immediate patient complicationsccurred in 7%–15% of cases, including chest pain (6%–2%), bleeding (0.2%–0.6%), perforation (0.6%–1%),nd death (0.5%–1.4%). Delayed technical complica-ions occurred in 9%–18% of cases, including tumorngrowth/overgrowth (6%–11%) and stent migration3%–7%). Delayed patient complications occurred in upo 27% of patients, including reflux symptoms (4%–%), recurrent dysphagia (8%–9%), tracheoesophagealstulas (1%–3%), bleeding (0.5%–4%), perforation0.5%–0.8%), and death as a result of underlying ma-

able 7. FDA-Approved SEMS

Ultraflexa

aterial Nickel titanium (nitinoesign Meshovered Yesadial force �elivery system (F) 16ength (cm) 10, 12, 15lare-end diameter (mm) 23, 28haft diameter (mm) 18, 23egree of shortening (%) 30–40istula closure Yes

dapted and reprinted with permission from Baron.286

Microvasive/Boston Scientific Inc (Natick, MA).Wilson-Cook Medical (Winston-Salem, NC).

ignancy occurring within 30 days (7%). r

Tumor ingrowth or overgrowth causing recurrent dys-hagia can be managed by placement of another stent orumor ablation using a variety of endoscopic modalities,ncluding laser therapy, argon plasma coagulation ther-py, and PDT.287–290 Care must be taken to avoid dam-ging the indwelling stent when thermal modalities aresed.There are conflicting reports on whether stent-relatedorbidity is increased in patients who have been treated

reviously with radiation and/or chemotherapy. Sometudies have shown an increased rate of stent-relatedife-threatening complications in patients who have hadrior chemoradiation therapy, whereas others haveot.272,285,291–295 Data are limited regarding the use ofalliative chemoradiation therapy after insertion oftents. In a small study of 29 patients, the group ofatients undergoing chemoradiation therapy followingtent placement survived longer (median, 331 days vs57 days; P � .05) than the stent-only group.296 On thether hand, a high risk of major complications was notedn patients undergoing radiotherapy after stent place-ent.297 The major complications found were creation oftracheal-esophageal fistula and massive hematemesis

rom stent erosion into the aorta. Placement of SEMSefore radiation therapy should be performed with cau-ion. Comparisons between available studies are difficultecause of differences in types of stents used and studyesigns.Stents are ideal for obstructing midesophageal cancers.

ollowing stent placement, patients are advised to mod-fy their diet and avoid solid boluses (e.g., meat andread) that could potentially become impacted withinhe stent. Stents that straddle the esophagogastric junc-ion may lead to severe reflux symptoms. Therefore, strictntireflux lifestyle precautions and administration of an-isecretory medications (ie, proton pump inhibitors) are

Wallstent IIa Z-Stentb

Elgiloy Stainless steelMesh ZigzagYes Yes��� ��18 3110, 15 8, 10, 12, 1428 2520 1830 0–10Yes Yes

l)

equired. Clinical experience with the recently intro-

Page 22: American Gastroenterological Association Technical Review on

dvpwastSSecbcmec

acfieccaoeefitwp

twsupsisratotsApbrgl

de

abaCdel

cidit5d(1pabpcia

plsrbrtlssstrrubpcmtcei

1492 WANG ET AL GASTROENTEROLOGY Vol. 128, No. 5

uced Dua stent (Z-stent with a windsock-like antirefluxalve) is limited, but the device seems beneficial inalliating dysphagia and controlling reflux in patientsith distal esophageal cancers.298,299 Stenting of esoph-

geal cancers in proximity of the upper esophagealphincter can be problematic, although tumors involvinghe cervical esophagus occur in �5% of all patients.300

mall case series have reported successful placement ofEMS to palliate cancers within 2 cm of the uppersophageal sphincter, although this can be technicallyhallenging.292,301 Complications may include foreignody sensation, oropharyngeal aspiration, and trachealompression with airway compromise.285 Stent place-ent in this setting should only be attempted by an

xperienced operator, or alternative modalities should beonsidered.

Stents can be effective for palliating dysphagia second-ry to malignant extrinsic compression. Covered stentsan also be used to treat malignant tracheoesophagealstulas and constitute a unique advantage over otherndoscopic modalities in this setting. Covered SEMS areonsidered a primary form of therapy for this ominousondition. Successful closure of malignant tracheoesoph-geal fistulas using covered SEMS is seen in 90%–100%f patients.293,295,301,302 Patients with persistent tracheo-sophageal fistulas despite esophageal stenting may ben-fit from airway stenting (double stenting) to close thestula303 or esophageal bypass in surgically fit pa-ients.304 Malignant esophageal obstruction associatedith perforation during endoscopic procedures may bealliated successfully with covered SEMS.305

Intratumoral injection of absolute alcohol results inissue necrosis and sloughing. The technique is cheap,idely available, and relatively simple to perform. The

clerosant is typically injected in 0.5- to 1-mL aliquotssing a standard sclerotherapy needle, targeting exo-hytic parts of the tumor. Experience accrued from caseeries shows an initial success rate of 80%–100% inmproving dysphagia. However, the palliative effect ishort-term (�1 month), and repeated sessions are usuallyequired in most patients.306–310 Chest pain is commonfter therapy. Serious complications, including medias-initis and tracheoesophageal fistulas, occur in up to 5%f cases.308 Standardized dosimetry and local control ofherapy are problematic due to leaking at the injectionite or tracking of the sclerosant along tissue planes.lcohol injection is probably best reserved for short,rotuberant, and nonfibrotic tumors that are not amena-le to other endoscopic palliative therapies. Preliminaryeports of intratumoral injection of cisplatin/epinephrineel have shown limited and short-lived efficacy of the

ocally administered chemotherapeutic agent in relieving fi

ysphagia.311,312 This form of injection therapy remainsxperimental at this time.

Similar to injection therapy, short exophytic tumorsre more conducive to thermal modalities, which includeipolar electrocoagulation, argon plasma coagulation,nd laser therapy. The use of bipolar electrocautery (BI-AP probe) for esophageal tumor ablation has beenescribed but made unpopular by more user-friendly andqually effective thermal modalities such as the Nd:YAGaser.313–315

Argon plasma coagulation therapy is a form of non-ontact monopolar electrocautery delivered to tissue viaonized, electrically conductive argon gas. Palliation ofysphagia can initially be achieved in most patients withnoperable cancer, but the median range between rein-erventions is approximately 1 month, with an average of

treatment sessions per patient.316 Despite the limitedepth of injury achieved by argon plasma coagulation�2 mm), perforations have been reported to occur in%–2% of treatments. In one study, one third of theatients eventually required esophageal stenting as anlternative mode of palliative therapy.316 There is limitedenefit in treating advanced bulky tumors with argonlasma coagulation. On the other hand, argon plasmaoagulation may be helpful in controlling tumor bleed-ng and in staving off tumor ingrowth or overgrowthssociated with metal stents.

The Nd:YAG laser causes deeper injury than argonlasma coagulation because it vaporizes tissue to recana-ize the obstructed lumen. Short-segment exophytic le-ions are ideal for laser therapy. The procedure is usuallyepeated 48 hours later for further treatment and de-ridement. Laser application is preferably performed in aetrograde fashion, which usually requires prelaser dila-ion for passage of the endoscope through the obstructedumen.317 Tumors that are close to the upper esophagealphincter may be more amenable to laser therapy thantenting. However, laser therapy is less successful thantenting for tumors involving the esophagogastric junc-ion and cardia.318 Laser therapy is 70%–95% effective atelieving dysphagia.317,319,320 The duration of responseanges from 1 to 2 months, but multiple sessions aresually required due to tumor regrowth.321 Response cane enhanced by external beam radiation or brachythera-y.322–324 Minor complications of laser therapy includehest pain, transient worsening of dysphagia from treat-ent-related edema, and leukocytosis. Major complica-

ions include bleeding, perforation (0%–5%), and tra-heoesophageal fistula (0%–6%). Laser equipment isxpensive and may not be widely available. Unlike stent-ng, laser therapy is contraindicated in the presence of

stulas; is generally not suitable for long, tortuous, cir-
Page 23: American Gastroenterological Association Technical Review on

cfs

tnaPetptsoeistaatidfilSs

pisrsrtctod(sSsopdigstws

tittttloP

ptptafworaantctatcapttvws

cagtsacftsabp

May 2005 AMERICAN GASTROENTEROLOGICAL ASSOCIATION 1493

umferentially narrowed tumors; and is not applicableor dysphagia caused by malignant extrinsic compres-ion.

PDT has also been applied as a palliative technique forhe management of malignant dysphagia.325,326 PDT isot limited to bulky tumors; infiltrative or flat tumoralreas as well as long-segment tumors may be amenable toDT. In addition, PDT using sodium porfimer may beasier to apply in cervical esophageal cancers than laserherapy or stenting. The response rate in dysphagia im-rovement ranges from 60% to 90%. Acute complica-ions (5%–20%) include chest pain, odynophagia, nau-ea, fever, leukocytosis, and pleural effusion.259 A majorbstacle to the use of sodium porfimer/PDT in advancedsophageal cancers is the duration of skin photosensitiv-ty (4–6 weeks) associated with the therapy. This is aignificant problem in a palliative setting. Costs relatedo initial purchasing of equipment and photosensitizersre additional drawbacks. PDT using aminolevuliniccid is attractive because of its limited duration of pho-osensitivity (48 hours), but this therapy has more lim-ted depth of tissue necrosis. PDT with hematoporphyrinerivative (which is a compound similar to sodium por-mer) was significantly more effective than aminolevu-inic acid in a nonrandomized study of 49 patients,327

odium porfimer is the only currently approved photo-ensitizer for gastrointestinal use in the United States.

Studies comparing various endoscopic modalities foralliation of malignant dysphagia are few and conflict-ng. Retrospective studies comparing laser therapy withtenting have shown similar outcomes with regard toelief of dysphagia but higher complication rates in thetent group.328,329 These studies are limited by theiretrospective design and heterogeneous patient popula-ions. On the other hand, a prospective, randomized,ontrolled trial of laser therapy versus SEMS in 60 pa-ients with previously untreated esophageal cancer dem-nstrated a significant improvement in the degree ofysphagia and a reduction in the reintervention rate35.7% vs 100%) in favor of SEMS.282 A smaller pro-pective randomized study of 39 patients evaluatingEMS versus laser therapy combined with radiotherapyhowed the former to be more cost-effective.330 A studyf 65 patients, however, showed longer survival rates inatients undergoing thermal ablative therapy (laser pre-ominantly) compared with those who received stent-ng.331 Stenting cost less but was associated with areater deterioration of health-related quality of life. In 2mall prospective randomized trials comparing laserherapy with injection therapy, no significant differencesere noted with regard to improvement in dysphagia

core or complication rates.332,333 a

In a prospective randomized study involving 236 pa-ients, PDT and laser therapy were similarly efficaciousn terms of dysphagia relief, although there was a trendoward a better response with PDT for tumors located inhe upper and middle third of the esophagus and for longumors.325 PDT was associated with fewer perforationshan with laser therapy (1% vs 7%). However, PDT wasimited by photosensitivity in 19% of cases. Terminationf sessions due to adverse events occurred in 3% withDT versus 19% with laser therapy.Nutritional support is often required in support of

atients with esophageal carcinoma. There is evidencehat enteral nutrition is beneficial in patients with dys-hagia and in those receiving radiation therapy.334 En-eral nutrition is generally preferred to the parenteralpproach. A randomized prospective study was per-ormed using these 2 approaches in patients who under-ent surgery for gastrointestinal tumors.335 Althoughnly 10% of the 257 patients had esophageal cancer, theesults showed that enteral nutrition was less expensivend improved gut perfusion. Enteral nutrition can beccomplished by feeding tubes that can be placed withasogastric or nasojejunal approaches, although theseypes of tubes are associated with increased bodily con-erns by patients and could limit their social interac-ions.336 Percutaneous gastrostomy or jejunostomy tubesre preferred because there is less social stigma and theubes are better tolerated. A retrospective study of per-utaneous gastrostomy tubes in 229 patients with esoph-geal cancer found that they could be placed in 97% ofatients, although dilation was required in 45% of pa-ients, with 2 having fatal adverse events from perfora-ions.337 Although only anecdotal, radiologists have ad-ocated radiologic placement of fielding tubes in patientsith oropharyngeal cancers to decrease the possibility of

tomal metastasis.338

Finally, palliative care of patients with esophagealancer requires the involvement of physicians who arettentive to both physical and emotional needs. Dyspha-ia is a symptom that has significant impact on a pa-ient’s perception of health, as demonstrated in a recenttudy of health state utilities in 50 patients with esoph-geal cancer.339 Many patients feel abandoned by physi-ians after medical therapy has been unsuccessful andurther interventions are no longer indicated. These pa-ients require support from hospice personnel and de-erve physician attention and compassion. Until esoph-geal cancer can be effectively treated, compassion maye one of the most important “therapies” we can offer foralliation of this disease.Summary of Evidence—Endoscopic therapy is effective

t alleviating dysphagia. However, the selection of a

Page 24: American Gastroenterological Association Technical Review on

pmptmswubSspat

dcofirsbhlwtvoetaascqTimBtetdemnqc

rb

agwsEcTecrmarcbTttt

1494 WANG ET AL GASTROENTEROLOGY Vol. 128, No. 5

articular endoscopic technique should be based on tu-or characteristics, patient preference, and available ex-

ertise. Esophageal dilation results in short-lived pallia-ion and is best used as an adjunct to other palliativeodalities (based on level III evidence). Esophageal

tenting is the preferred endoscopic modality in patientsith long malignant strictures more than 2 cm from thepper esophageal sphincter and/or with fistulas (alsoased on level III evidence). There is level I evidence thatEMS are superior to conventional semirigid plastictents in the management of malignant dysphagia. Otheralliative methods such as alcohol injection, laser ther-py, and PDT are similarly efficacious as tumor ablativeherapies (based on level II evidence).

Future Directions

It is clear that further research is needed in theetection, prevention, and treatment of esophageal car-inoma. The cause of the continued increase in incidencef adenocarcinoma of the esophagus still remains unde-ned. The need to stratify patients for esophageal cancerisk is essential to implement treatment and preventiontrategies. Although histologic evidence of dysplasia haseen the gold standard to define cancer risk, this is veryard to reproduce and is difficult to detect withoutaborious biopsy protocols. Biomarkers for cancer riskould be very attractive but will require large prospec-

ive studies to validate. Chemoprevention would be aiable strategy for patients at lower risk for developmentf cancer, and NSAIDs might a fruitful candidate. How-ver, due to the low risks for cancer, very large patientrials will be required to demonstrate benefit. In patientst high risk for cancer, ablative therapies have beenpplied with some initial promising results. The clinicalignificance of reduction of the amount of Barrett’s mu-osa, elimination of dysplasia, and development of subs-uamous areas of Barrett’s mucosa needs to be defined.he detection and staging of esophageal cancers could be

mproved with the advent of higher-resolution imagingodalities such as OCT and fluorescence spectroscopy.oth of these entities require further development before

hey can be clinically applied. Endoscopic therapy ofarly cancers seems to be achievable, but large prospec-ive studies are needed to define the best methods and toetermine the long-term outcome. Surgical therapy isvolving toward less invasive approaches, but improve-ents in chemotherapy and radiation might decrease the

eed for resection in advanced cancers. Palliation is re-uired for the majority of patients with esophageal can-

er, and the development of newer stents that prevent

eflux and conform better to the esophagus would beeneficial to patients.

Summary

Esophageal cancer is increasing in incidence and isssociated with a high mortality rate. The ability ofastroenterologists to increase survival in this diseaseill depend on earlier detection through screening and

urveillance strategies. Early cancers can be staged withUS and endoscopic mucosal resection. More advancedancers will require the addition of CT and possible PET.reatment of early cancers is increasingly shifting towardndoscopic treatment. More advanced but localized can-ers can be treated by surgical resection. Cancers withegional lymphadenopathy may require neoadjuvant che-otherapy and radiation therapy. Patients with more

dvanced cancers may respond to primary therapy withadiation and chemotherapy. Palliation of esophagealancers is based on stent therapy, but physicians shoulde attentive to the emotional needs of the dying patient.he future of therapy for esophageal cancer may rest with

he development of chemoprevention methods, althoughhere is not substantial evidence to support its use at thisime.

References1. Streitz JM Jr, Andrews CW Jr, Ellis FH Jr. Endoscopic surveillance

of Barrett’s esophagus. Does it help? J Thorac Cardiovasc Surg1993;105:383–387; discussion 387–388.

2. Atkinson M, Iftikhar SY, James PD, Robertson CS, Steele RJ.The early diagnosis of oesophageal adenocarcinoma by endo-scopic screening. Eur J Cancer Prev 1992;1:327–330.

3. Peters JH, Clark GW, Ireland AP, Chandrasoma P, Smyrk TC,DeMeester TR. Outcome of adenocarcinoma arising in Barrett’sesophagus in endoscopically surveyed and nonsurveyed pa-tients. J Thorac Cardiovasc Surg 1994;108:813–821; discus-sion 821–822.

4. Lieberman DA, Oehlke M, Helfand M. Risk factors for Barrett’sesophagus in community-based practice. GORGE consortium.Gastroenterology Outcomes Research Group in Endoscopy. Am JGastroenterol 1997;92:1293–1297.

5. Gerson LB, Shetler K, Triadafilopoulos G. Prevalence of Barrett’sesophagus in asymptomatic individuals. Gastroenterology2002;123:461–467.

6. Rex DK, Cummings OW, Shaw M, Cumings MD, Wong RK,Vasudeva RS, Dunne D, Rahmani EY, Helper DJ. Screening forBarrett’s esophagus in colonoscopy patients with and withoutheartburn. Gastroenterology 2003;125:1670–1677.

7. Iftikhar SY, James PD, Steele RJ, Hardcastle JD, Atkinson M.Length of Barrett’s oesophagus: an important factor in thedevelopment of dysplasia and adenocarcinoma (comment). Gut1992;33:1155–1158.

8. Poynton AR, Walsh TN, O’Sullivan G, Hennessy TP. Carcinomaarising in familial Barrett’s esophagus. Am J Gastroenterol1996;91:1855–1856.

9. Eng C, Spechler SJ, Ruben R, Li FP. Familial Barrett esophagus andadenocarcinoma of the gastroesophageal junction (published erra-tum appears in Cancer Epidemiol Biomarkers Prev 1993;2:505).

Cancer Epidemiol Biomarkers Prev 1993;2:397–399.
Page 25: American Gastroenterological Association Technical Review on

May 2005 AMERICAN GASTROENTEROLOGICAL ASSOCIATION 1495

10. Jochem VJ, Fuerst PA, Fromkes JJ. Familial Barrett’s esophagusassociated with adenocarcinoma. Gastroenterology 1992;102:1400–1402.

11. Chak A, Lee T, Kinnard MF, Brock W, Faulx A, Willis J, CooperGS, Sivak MV Jr, Goddard KA. Familial aggregation of Barrett’soesophagus, oesophageal adenocarcinoma, and oesophago-gastric junctional adenocarcinoma in Caucasian adults. Gut2002;51:323–328.

12. Trudgill N. Familial factors in the etiology of gastroesophagealreflux disease, Barrett’s esophagus, and esophageal adenocar-cinoma. Chest Surg Clin North Am 2002;12:15–24.

13. Saeian K, Staff DM, Vasilopoulos S, Townsend WF, Almagro UA,Komorowski RA, Choi H, Shaker R. Unsedated transnasal en-doscopy accurately detects Barrett’s metaplasia and dysplasia.Gastrointest Endosc 2002;56:472–478.

14. Falk GW, Chittajallu R, Goldblum JR, Biscotti CV, Geisinger KR,Petras RE, Birgisson S, Rice TW, Richter JE. Surveillance ofpatients with Barrett’s esophagus for dysplasia and cancer withballoon cytology (comment). Gastroenterology 1997;112:1787–1797.

15. Rader AE, Faigel DO, Ditomasso J, Magaret N, Burm M, FennertyMB. Cytological screening for Barrett’s esophagus using a pro-totype flexible mesh catheter. Dig Dis Sci 2001;46:2681–2686.

16. Fennerty MB, DiTomasso J, Morales TG, Peterson D, KarmakarA, Fernandez T, Sampliner R. Screening for Barrett’s esophagusby balloon cytology. Am J Gastroenterol 1995;90:1230–1232.

17. Risk JM, Mills HS, Garde J, Dunn JR, Evans KE, Hollstein M,Field JK. The tylosis esophageal cancer (TOC) locus: more thanjust a familial cancer gene. Dis Esophagus 1999;12:173–176.

18. Riddell RH. Early detection of neoplasia of the esophagus andgastroesophageal junction. Am J Gastroenterol 1996;91:853–863.

19. Csikos M, Horvath O, Petri A, Petri I, Imre J. Late malignanttransformation of chronic corrosive oesophageal strictures. Lan-genbecks Arch Chir 1985;365:231–238.

20. Ti TK. Oesophageal carcinoma associated with corrosive inju-ry—prevention and treatment by oesophageal resection. Br JSurg 1983;70:223–225.

21. Sandler RS, Nyren O, Ekbom A, Eisen GM, Yuen J, Josefsson S.The risk of esophageal cancer in patients with achalasia. Apopulation-based study. JAMA 1995;274:1359–1362.

22. Loviscek LF, Cenoz MC, Badaloni AE, Agarinakazato O. Earlycancer in achalasia. Dis Esophagus 1998;11:239–247.

23. Rosenberg PS, Greene MH, Alter BP. Cancer incidence in per-sons with Fanconi anemia (comment). Blood 2003;101:822–826.

24. Maisel RH, Vermeersch H. Panendoscopy for second primariesin head and neck cancer. Ann Otol Rhinol Laryngol 1981;90:460–464.

25. Yokoyama A, Ohmori T, Makuuchi H, Maruyama K, Okuyama K,Takahashi H, Yokoyama T, Yoshino K, Hayashida M, Ishii H.Successful screening for early esophageal cancer in alcoholicsusing endoscopy and mucosa iodine staining (comment). Can-cer 1995;76:928–934.

26. Kikuchi S, Sato M, Katada N, Sakuramoto S, Shimao H, KakitaA, Ohida M, Saigenji K. Efficacy of endoscopic surveillance ofthe upper gastrointestinal tract following distal gastrectomy forearly gastric cancer. Hepatogastroenterology 2003;50:1704–1707.

27. Sharma VK, Chockalingam H, Hornung CA, Vasudeva R, HowdenCW. Changing trends in esophageal cancer: a 15-year experi-ence in a single center. Am J Gastroenterol 1998;93:702–705.

28. Firoozi B, Vega KJ, Holland BK, Koliver MG, Trotman BW. Epide-miologic pattern of esophageal cancer at an inner-city university

hospital. J Assoc Acad Minor Phys 1999;10:44–47.

29. Larsson LG, Sandstrom A, Westling P. Relationship of Plummer-Vinson disease to cancer of the upper alimentary tract in Swe-den. Cancer Res 1975;35:3308–3316.

30. Roth MJ, Liu SF, Dawsey SM, Zhou B, Copeland C, Wang GQ,Solomon D, Baker SG, Giffen CA, Taylor PR. Cytologic detectionof esophageal squamous cell carcinoma and precursor lesionsusing balloon and sponge samplers in asymptomatic adults inLinxian, China. Cancer 1997;80:2047–2059.

31. Yang H, Berner A, Mei Q, Giercksky KE, Warloe T, Yang G, Cui J,Suo Z, Zhang S, Nesland JM. Cytologic screening for esopha-geal cancer in a high-risk population in Anyang County, China.Acta Cytol 2002;46:445–452.

32. Cameron AJ. Epidemiology of columnar-lined esophagus andadenocarcinoma. Gastroenterol Clin North Am 1997;26:487–494.

33. Williamson WA, Ellis FH Jr, Gibb SP, Shahian DM, Aretz HT,Heatley GJ, Watkins E Jr. Barrett’s esophagus. Prevalence andincidence of adenocarcinoma. Arch Intern Med 1991;151:2212–2216.

34. Spechler SJ, Robbins AH, Rubins HB, Vincent ME, Heeren T,Doos WG, Colton T, Schimmel EM. Adenocarcinoma and Bar-rett’s esophagus. An overrated risk? Gastroenterology 1984;87:927–933.

35. Reid BJ, Weinstein WM. Barrett’s esophagus and adenocarci-noma. Ann Rev Med 1987; 38:477–492.

36. Hamilton SR, Smith RR. The relationship between columnarepithelial dysplasia and invasive adenocarcinoma arising in Bar-rett’s esophagus. Am J Clin Pathol 1987;87:301–312.

37. Miros M, Kerlin P, Walker N. Only patients with dysplasiaprogress to adenocarcinoma in Barrett’s oesophagus (com-ment). Gut 1991;32:1441–1446.

38. Provenzale D, Schmitt C, Wong JB. Barrett’s esophagus: a newlook at surveillance based on emerging estimates of cancerrisk. Am J Gastroenterol 1999;94:2043–2053.

39. Shaheen NJ, Provenzale D, Sandler RS. Upper endoscopy as ascreening and surveillance tool in esophageal adenocarcinoma:a review of the evidence. Am J Gastroenterol 2002;97:1319–1327.

40. Reid BJ, Haggitt RC, Rubin CE, Rabinovitch PS. Barrett’s esoph-agus. Correlation between flow cytometry and histology in de-tection of patients at risk for adenocarcinoma. Gastroenterology1987;93:1–11.

41. Haggitt RC, Reid BJ, Rabinovitch PS, Rubin CE. Barrett’s esoph-agus. Correlation between mucin histochemistry, flow cytom-etry, and histologic diagnosis for predicting increased cancerrisk. Am J Pathol 1988;131:53–61.

42. Levine DS, Haggitt RC, Blount PL, Rabinovitch PS, Rusch VW,Reid BJ. An endoscopic biopsy protocol can differentiate high-grade dysplasia from early adenocarcinoma in Barrett’s esoph-agus (comment). Gastroenterology 1993;105:40–50.

43. Reid BJ, Levine DS, Longton G, Blount PL, Rabinovitch PS.Predictors of progression to cancer in Barrett’s esophagus:baseline histology and flow cytometry identify low- and high-riskpatient subsets. Am J Gastroenterol 2000;95:1669–1676.

44. Teodori L, Gohde W, Persiani M, Ferrario F, Tirindelli Danesi D,Scarpignato C, Di Tondo U, Alo P, Capurso L. DNA/protein flowcytometry as a predictive marker of malignancy in dysplasia-freeBarrett’s esophagus: thirteen-year follow-up study on a cohort ofpatients. Cytometry 1998;34:257–263.

45. Weston AP, Badr AS, Hassanein RS. Prospective multivariateanalysis of clinical, endoscopic, and histological factors predic-tive of the development of Barrett’s multifocal high-grade dys-plasia or adenocarcinoma. Am J Gastroenterol 1999;94:3413–3419.

46. Skacel M, Petras RE, Gramlich TL, Sigel JE, Richter JE, Goldblum

JR. The diagnosis of low-grade dysplasia in Barrett’s esophagus
Page 26: American Gastroenterological Association Technical Review on

1496 WANG ET AL GASTROENTEROLOGY Vol. 128, No. 5

and its implications for disease progression. Am J Gastroenterol2000;95:3383–3387.

47. Schnell TG, Sontag SJ, Chejfec G, Aranha G, Metz A, O’ConnellS, Seidel UJ, Sonnenberg A. Long-term nonsurgical manage-ment of Barrett’s esophagus with high-grade dysplasia. Gastro-enterology 2001;120:1607–1619.

48. Buttar NS, Wang KK, Sebo TJ, Riehle DM, Krishnadath KK,Lutzke LS, Anderson MA, Petterson TM, Burgart LJ. Extent ofhigh-grade dysplasia in Barrett’s esophagus correlates with riskof adenocarcinoma. Gastroenterology 2001;120:1630–1639.

49. Robertson CS, Mayberry JF, Nicholson DA, James PD, AtkinsonM. Value of endoscopic surveillance in the detection of neoplas-tic change in Barrett’s oesophagus. Br J Surg 1988;75:760–763.

50. O’Connor JB, Falk GW, Richter JE. The incidence of adenocarci-noma and dysplasia in Barrett’s esophagus: report on the Cleve-land Clinic Barrett’s Esophagus Registry. Am J Gastroenterol1999;94:2037–2042.

51. Reid BJ, Haggitt RC, Rubin CE, Roth G, Surawicz CM, Van BelleG, Lewin K, Weinstein WM, Antonioli DA, Goldman H, et al.Observer variation in the diagnosis of dysplasia in Barrett’sesophagus. Hum Pathol 1988;19:166–178.

52. Montgomery E, Goldblum JR, Greenson JK, Haber MM, LampsLW, Lauwers GY, Lazenby AJ, Lewin DN, Robert ME, WashingtonK, Zahurak ML, Hart J. Dysplasia as a predictive marker forinvasive carcinoma in Barrett esophagus: a follow-up studybased on 138 cases from a diagnostic variability study. HumPathol 2001;32:379–388.

53. Inadomi JM, Sampliner R, Lagergren J, Lieberman D, FendrickAM, Vakil N. Screening and surveillance for Barrett esophagusin high-risk groups: a cost-utility analysis (comment). Ann InternMed 2003;138:176–186.

54. Lerut T, Coosemans W, Van Raemdonck D, Dillemans B, DeLeyn P, Marnette JM, Geboes K. Surgical treatment of Barrett’scarcinoma. Correlations between morphologic findings and prog-nosis. J Thorac Cardiovasc Surg 1994;107:1059–1065; dis-cussion 1065–1066.

55. Paraf F, Flejou JF, Pignon JP, Fekete F, Potet F. Surgical pathol-ogy of adenocarcinoma arising in Barrett’s esophagus. Analysisof 67 cases. Am J Surg Pathol 1995;19:183–191.

56. Farrow DC, Vaughan TL. Determinants of survival following thediagnosis of esophageal adenocarcinoma (United States). Can-cer Causes Control 1996;7:322–327.

57. van Sandick JW, van Lanschot JJ, Kuiken BW, Tytgat GN, Offer-haus GJ, Obertop H. Impact of endoscopic biopsy surveillance ofBarrett’s oesophagus on pathological stage and clinical out-come of Barrett’s carcinoma. Gut 1998;43:216–222.

58. Wright TA, Gray MR, Morris AI, Gilmore IT, Ellis A, Smart HL,Myskow M, Nash J, Donnelly RJ, Kingsnorth AN. Cost effective-ness of detecting Barrett’s cancer. Gut 1996;39:574–579.

59. Bani-Hani K, Sue-Ling H, Johnston D, Axon AT, Martin IG. Bar-rett’s oesophagus: results from a 13-year surveillance pro-gramme. Eur J Gastroenterol Hepatol 2000;12:649–654.

60. van der Burgh A, Dees J, Hop WC, van Blankenstein M. Oesoph-ageal cancer is an uncommon cause of death in patients withBarrett’s oesophagus. Gut 1996;39:5–8.

61. Macdonald CE, Wicks AC, Playford RJ. Final results from 10 yearcohort of patients undergoing surveillance for Barrett’s oesoph-agus: observational study. BMJ 2000;321:1252–1255.

62. Corley DA, Levin TR, Habel LA, Weiss NS, Buffler PA. Surveil-lance and survival in Barrett’s adenocarcinomas: a population-based study. Gastroenterology 2002;122:633–640.

63. Fisher D, Jeffreys A, Bosworth H, Wang J, Lipscomb J, ProvenzaleD. Quality of life in patients with Barrett’s esophagus undergoingsurveillance. Am J Gastroenterol 2002;97:2193–2200.

64. Sampliner RE, The Practice Parameters Committee of the Amer-

ican College of Gastroenterology. Updated guidelines for the

diagnosis, surveillance, and therapy of Barrett’s esophagus.Am J Gastroenterol 2002;97:1888–1895.

65. Reid BJ, Blount PL, Feng Z, Levine DS. Optimizing endoscopicbiopsy detection of early cancers in Barrett’s high-grade dyspla-sia. Am J Gastroenterol 2000;95:3089–3096.

66. Levine DS, Reid BJ. Endoscopic biopsy technique for acquiringlarger mucosal samples. Gastrointest Endosc 1991;37:332–337.

67. Falk GW, Rice TW, Goldblum JR, Richter JE. Jumbo biopsyforceps protocol still misses unsuspected cancer in Barrett’sesophagus with high-grade dysplasia. Gastrointest Endosc1999;49:170–176.

68. Nijhawan PK, Wang KK. Endoscopic mucosal resection for le-sions with endoscopic features suggestive of malignancy andhigh-grade dysplasia within Barrett’s esophagus. GastrointestEndosc 2000;52:328–332.

69. Robey SS, Hamilton SR, Gupta PK, Erozan YS. Diagnostic valueof cytopathology in Barrett esophagus and associated carci-noma. Am J Clin Pathol 1988;89:493–498.

70. Geisinger KR, Teot LA, Richter JE. A comparative cytopathologicand histologic study of atypia, dysplasia, and adenocarcinomain Barrett’s esophagus. Cancer 1992;69:8–16.

71. Geisinger KR. Endoscopic biopsies and cytologic brushings ofthe esophagus are diagnostically complementary. Am J ClinPathol 1995;103:295–299.

72. Sebo TJ. Digital image analysis. Mayo Clin Proc 1995;70:81–82.

73. Pacifico RJ, Wang KK, Buttar NS, et al. Surveillance of Barrett’sesophagus using brush cytology and digital image analysis: arandomized prospective trial (abstr). Gastrointest Endosc 2003;57:AB91.

74. Hu N, Taylor PR, Rao JY, Hemstreet GP, Liu SF, Zou XN, MarkSD, Dawsey SM. Quantitative fluorescence image analysis ofDNA content and nuclear morphology on esophageal ballooncytology smears and subsequent development of esophagealand gastric cardia cancer in Linxian, China. Cancer EpidemiolBiomarkers Prev 1998;7:59–64.

75. Dawsey SM, Shen Q, Nieberg RK, Liu SF, English SA, Cao J,Zhou B, Wang GQ, Lewin KJ, Liu FS, Roth MJ, Taylor PR. Studiesof esophageal balloon cytology in Linxian, China. Cancer Epide-miol Biomarkers Prev 1997;6:121–130.

76. Connor MJ, Sharma P. Chromoendoscopy and magnificationendoscopy in Barrett’s esophagus. Gastrointest Endosc ClinNorth Am 2003;13:269–277.

77. Dawsey SM, Fleischer DE, Wang GQ, Zhou B, Kidwell JA, Lu N,Lewin KJ, Roth MJ, Tio TL, Taylor PR. Mucosal iodine stainingimproves endoscopic visualization of squamous dysplasia andsquamous cell carcinoma of the esophagus in Linxian, China.Cancer 1998;83:220–231.

78. Muto M, Hironaka S, Nakane M, Boku N, Ohtsu A, Yoshida S.Association of multiple Lugol-voiding lesions with synchronousand metachronous esophageal squamous cell carcinoma inpatients with head and neck cancer. Gastrointest Endosc 2002;56:517–521.

79. Tincani AJ, Brandalise N, Altemani A, Scanavini RC, Valerio JB,Lage HT, Molina G, Martins AS. Diagnosis of superficial esoph-ageal cancer and dysplasia using endoscopic screening with a2% lugol dye solution in patients with head and neck cancer.Head Neck 2000;22:170–174.

80. Freitag CP, Barros SG, Kruel CD, Putten AC, Dietz J, Gruber AC,Diehl AS, Meurer L, Breyer HP, Wolff F, Vidal R, Arruda CA, Luz LP,Fagundes RB, Prolla JC. Esophageal dysplasias are detected byendoscopy with Lugol in patients at risk for squamous cell carci-noma in southern Brazil. Dis Esophagus 1999;12:191–195.

81. Meyer V, Burtin P, Bour B, Blanchi A, Cales P, Oberti F, PersonB, Croue A, Dohn S, Benoit R, Fabiani B, Boyer J. Endoscopic

detection of early esophageal cancer in a high-risk population:
Page 27: American Gastroenterological Association Technical Review on

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

May 2005 AMERICAN GASTROENTEROLOGICAL ASSOCIATION 1497

does Lugol staining improve videoendoscopy? GastrointestEndosc 1997;45:480–484.

82. Sharma P, Topalovski M, Mayo MS, Weston AP. Methylene bluechromoendoscopy for detection of short-segment Barrett’sesophagus. Gastrointest Endosc 2001;54:289–293.

83. Canto MI. Methylene blue chromoendoscopy for Barrett’sesophagus: coming soon to your GI unit? (letter; comment).Gastrointest Endosc 2001;54:403–409.

84. Canto MI, Setrakian S, Willis JE, Chak A, Petras RE, Sivak MV.Methylene blue staining of dysplastic and nondysplastic Bar-rett’s esophagus: an in vivo and ex vivo study. Endoscopy2001;33:391–400.

85. Canto MI, Setrakian S, Willis J, Chak A, Petras R, Powe NR,Sivak MV Jr. Methylene blue-directed biopsies improve detec-tion of intestinal metaplasia and dysplasia in Barrett’s esopha-gus. Gastrointest Endosc 2000;51:560–568.

86. Wong RK, Horwhat JD, Maydonovitch CL. Sky blue or murkywaters: the diagnostic utility of methylene blue. GastrointestEndosc 2001;54:409–413.

87. Wo JM, Ray MB, Mayfield-Stokes S, Al-Sabbagh G, Gebrail F,Slone SP, Wilson MA. Comparison of methylene blue-directedbiopsies and conventional biopsies in the detection of intestinalmetaplasia and dysplasia in Barrett’s esophagus: a preliminarystudy. Gastrointest Endosc 2001;54:294–301.

88. Egger K, Werner M, Meining A, Ott R, Allescher HD, Hofler H,Classen M, Rosch T. Biopsy surveillance is still necessary inpatients with Barrett’s oesophagus despite new endoscopicimaging techniques. Gut 2003;52:18–23.

89. Olliver JR, Wild CP, Sahay P, Dexter S, Hardie LJ. Chromoendos-copy with methylene blue and associated DNA damage in Bar-rett’s oesophagus. Lancet 2003;362:373–374.

90. Stevens PD, Lightdale CJ, Green PH, Siegel LM, Garcia-Car-rasquillo RJ, Rotterdam H. Combined magnification endoscopywith chromoendoscopy for the evaluation of Barrett’s esopha-gus. Gastrointest Endosc 1994;40:747–749.

91. Guelrud M, Herrera I, Essenfeld H, Castro J. Enhanced magni-fication endoscopy: a new technique to identify specialized in-testinal metaplasia in Barrett’s esophagus. Gastrointest En-dosc 2001;53:559–565.

92. Endo T, Awakawa T, Takahashi H, Arimura Y, Itoh F, YamashitaK, Sasaki S, Yamamoto H, Tang X, Imai K. Classification ofBarrett’s epithelium by magnifying endoscopy. Gastrointest En-dosc 2002;55:641–647.

93. Sharma P, Weston AP, Topalovski M, Cherian R, BhattacharyyaA, Sampliner RE. Magnification chromoendoscopy for the detec-tion of intestinal metaplasia and dysplasia in Barrett’s oesoph-agus (comment). Gut 2003;52:24–27.

94. Panjehpour M, Overholt BF, Vo-Dinh T, Haggitt RC, Edwards DH,Buckley FP III. Endoscopic fluorescence detection of high-gradedysplasia in Barrett’s esophagus (comment). Gastroenterology1996;111:93–101.

95. Bourg-Heckly G, Blais J, Padilla JJ, Bourdon O, Etienne J, Guil-lemin F, Lafay L. Endoscopic ultraviolet-induced autofluores-cence spectroscopy of the esophagus: tissue characterizationand potential for early cancer diagnosis. Endoscopy 2000;32:756–765.

96. Wang KK, Buttar NS, WongKeeSong LM, Lutzke LS, Mirhej A,Krishnadath KK. The use of an optical biopsy system in Bar-rett’s esophagus (abstr). Gastroenterology 2001;120:A413.

97. Mayinger B, Horner P, Jordan M, Gerlach C, Horbach T, Hohen-berger W, Hahn EG. Light-induced autofluorescence spectros-copy for the endoscopic detection of esophageal cancer. Gas-trointest Endosc 2001;54:195–201.

98. Pfefer TJ, Paithankar DY, Poneros JM, Schomacker KT, NishiokaNS. Temporally and spectrally resolved fluorescence spectros-copy for the detection of high grade dysplasia in Barrett’s esoph-

agus. Lasers Surg Med 2003;32:10–16.

99. von Holstein CS, Nilsson AM, Andersson-Engels S, Willen R,Walther B, Svanberg K. Detection of adenocarcinoma in Bar-rett’s oesophagus by means of laser induced fluorescence. Gut1996;39:711–716.

00. Brand S, Wang TD, Schomacker KT, Poneros JM, Lauwers GY,Compton CC, Pedrosa MC, Nishioka NS. Detection of high-gradedysplasia in Barrett’s esophagus by spectroscopy measure-ment of 5-aminolevulinic acid-induced protoporphyrin IX fluores-cence. Gastrointest Endosc 2002;56:479–487.

01. Ortner MA, Ebert B, Hein E, Zumbusch K, Nolte D, Sukowski U,Weber-Eibel J, Fleige B, Dietel M, Stolte M, Oberhuber G, Por-schen R, Klump B, Hortnagl H, Lochs H, Rinneberg H. Timegated fluorescence spectroscopy in Barrett’s oesophagus. Gut2003;52:28–33.

02. Wallace M, Van Dam J. Enhanced gastrointestinal diagnosis:light-scattering spectroscopy and optical coherence tomogra-phy. Gastrointest Endosc Clin North Am 2000;10:71–80, vi.

03. WongKeeSong LM, Marcon N. Fluorescence and Raman spec-troscopy. Gastrointest Endosc Clin North Am 2003;13:279–296.

04. Georgakoudi I, Jacobson BC, Van Dam J, Backman V, WallaceMB, Muller MG, Zhang Q, Badizadegan K, Sun D, Thomas GA,Perelman LT, Feld MS. Fluorescence, reflectance, and light-scattering spectroscopy for evaluating dysplasia in patientswith Barrett’s esophagus. Gastroenterology 2001;120:1620–1629.

05. Bergman JJ, Kara MA, Smits ME, Rosmolen WD, Ten Kate F,Tytgat G. A randomized cross-over study comparing light-in-duced fluorescence endoscopy (LIFE) with standard endoscopyfor detection of early neoplasia in Barrett’s esophagus (abstr).Gastroenterology 2003;124:A49.

06. Sergeev AM, Gelikonov VM, Gelikonov GV, Feldchtein FI,Kuranov RV, Gladkova ND. In vivo endoscopic OCT imaging ofprecancer and cancer of human mucosa. Opt Exp 1997;1:432–440.

07. Bouma BE, Tearney GJ, Compton CC, Nishioka NS. High-reso-lution imaging of the human esophagus and stomach in vivousing optical coherence tomography. Gastrointest Endosc2000;51:467–474.

08. Sivak MV, Kobayashi K, Izatt JA, Rollins AM, Ung-Runyawee R,Chak A, Wong RC, Isenberg GA, Willis J. High-resolution endo-scopic imaging of the GI tract using optical coherence tomog-raphy. Gastrointest Endosc 2000;51:474–479.

09. Jackle S, Gladkova N, Feldchtein F, Terentieva A, Brand B,Gelikonov G, Gelikonov V, Sergeev A, Fritscher-Ravens A,Freund J, Seitz U, Schroder S, Soehendra N. In vivo endoscopicoptical coherence tomography of esophagitis, Barrett’s esoph-agus, and adenocarcinoma of the esophagus. Endoscopy2000;32:750–755.

10. Poneros JM, Nishioka NS. Diagnosis of Barrett’s esophagususing optical coherence tomography. Gastrointest Endosc ClinNorth Am 2003;13:309–323.

11. Falk GW. Barrett’s esophagus. Gastroenterology 2002;122:1569–1591.

12. Van Laethem JL, Peny MO, Salmon I, Cremer M, Deviere J.Intramucosal adenocarcinoma arising under squamous re-epi-thelialisation of Barrett’s oesophagus. Gut 2000;46:574–577.

13. Krishnadath K, Wang K, Taniguchi K, Sebo TJ, Buttar NS, Ander-son MA, Lutzke LS, Liu W. Persistent genetic abnormalities inBarrett’s esophagus after photodynamic therapy. Gastroenter-ology 2000;119:624–630.

14. Sampliner RE, Garewal HS, Fennerty MB, Aickin M. Lack ofimpact of therapy on extent of Barrett’s esophagus in 67 pa-tients. Dig Dis Sci 1990;35:93–96.

15. Csendes A, Braghetto I, Burdiles P, Puente G, Korn O, Diaz JC,Maluenda F. Long-term results of classic antireflux surgery in

152 patients with Barrett’s esophagus: clinical, radiologic, en-
Page 28: American Gastroenterological Association Technical Review on

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1498 WANG ET AL GASTROENTEROLOGY Vol. 128, No. 5

doscopic, manometric, and acid reflux test analysis before andlate after operation. Surgery 1998;126:645–657.

16. Peters FT, Ganesh S, Kuipers EJ, Sluiter WJ, Klinkenberg-KnolEC, Lamers CB, Kleibeuker JH. Endoscopic regression of Bar-rett’s oesophagus during omeprazole treatment; a randomiseddouble blind study. Gut 1999;45:489–494.

17. Yau P, Watson DI, Devitt PG, Game PA, Jamieson GG. Laparo-scopic antireflux surgery in the treatment of gastroesophagealreflux in patients with Barrett esophagus. Arch Surg 2000;135:801–805.

18. Ouatu-Lascar R, Triadafilopoulos G. Complete elimination ofreflux symptoms does not guarantee normalization of intra-esophageal acid reflux in patients with Barrett’s esophagus.Am J Gastroenterol 1998;93:711–716.

19. Fass R, Sampliner RE, Malagon IB, Hayden CW, Camargo L,Wendel CS, Garewal HS. Failure of oesophageal acid control incandidates for Barrett’s oesophagus reversal on a very highdose of proton pump inhibitor. Aliment Pharmacol Ther 2000;14:597–602.

20. Fitzgerald RC, Omary MB, Triadafilopoulos G. Dynamic effects ofacid on Barrett’s esophagus. An ex vivo proliferation and differ-entiation model. J Clin Invest 1996;98:2120–2128.

21. Kaur BS, Ouatu-Lascar R, Omary MB, Triadafilopoulos G. Bilesalts induce or blunt cell proliferation in Barrett’s esophagus inan acid-dependent fashion. Am J Physiol Gastrointest LiverPhysiol 2000;278:G1000–G1009.

22. Zhang F, Altorki NK, Wu YC, Soslow RA, Subbaramaiah K,Dannenberg AJ. Duodenal reflux induces cyclooxygenase-2 inthe esophageal mucosa of rats: evidence for involvement of bileacids. Gastroenterology 2001;121:1391–1399.

23. Buttar NS, Wang KK, Leontovich O, Westcott JY, Pacifico RJ,Anderson MA, Krishnadath KK, Lutzke LS, Burgart LJ. Chemo-prevention of esophageal adenocarcinoma by COX-2 inhibitorsin an animal model of Barrett’s esophagus. Gastroenterology2002;122:1101–1112.

24. Ouatu-Lascar R, Fitzgerald RC, Triadafilopoulos G. Differentia-tion and proliferation in Barrett’s esophagus and the effects ofacid suppression. Gastroenterology 1999;117:327–335.

25. Spechler SJ, Lee E, Ahnen D, Goyal RK, Hirano I, Ramirez F,Raufman JP, Sampliner R, Schnell T, Sontag S, Vlahcevic ZR,Young R, Williford W. Long-term outcome of medical and surgi-cal therapies for gastroesophageal reflux disease: follow-up of arandomized controlled trial (comment). JAMA 2001;285:2331–2338.

26. Ye W, Chow WH, Lagergren J, Yin L, Nyren O. Risk of adenocar-cinomas of the esophagus and gastric cardia in patients withgastroesophageal reflux diseases and after antireflux surgery.Gastroenterology 2001;121:1286–1293.

27. Wang KK, Sampliner RE. Mucosal ablation therapy of barrettesophagus. Mayo Clin Proc 2001;76:433–437.

28. Pacifico RJ, Wang KK. Role of mucosal ablative therapy in thetreatment of the columnar-lined esophagus. Chest Surg ClinNorth Am 2002;12:185–203.

29. Berenson MM, Johnson TD, Markowitz NR, Buchi KN, SamowitzWS. Restoration of squamous mucosa after ablation of Bar-rett’s esophageal epithelium. Gastroenterology 1993;104:1686–1691.

30. Sampliner RE, Faigel D, Fennerty MB, Lieberman D, Ippoliti A,Lewin K, Weinstein WM. Effective and safe endoscopic reversalof nondysplastic Barrett’s esophagus with thermal electrocoag-ulation combined with high-dose acid inhibition: a multicenterstudy. Gastrointest Endosc 2001;53:554–558.

31. Ackroyd R, Tam W, Schoeman M, Devitt PG, Watson DI. Pro-spective randomized controlled trial of argon plasma coagula-tion ablation vs. endoscopic surveillance of patients with Bar-rett’s esophagus after antireflux surgery (comment).

Gastrointest Endosc 2004;59:1–7.

32. Pagani M, Granelli P, Chella B, Antoniazzi L, Bonavina L, Perac-chia A. Barrett’s esophagus: combined treatment using argonplasma coagulation and laparoscopic antireflux surgery. DisEsophagus 2003;16:279–283.

33. Van Laethem JL, Cremer M, Peny MO, Delhaye M, Deviere J.Eradication of Barrett’s mucosa with argon plasma coagulationand acid suppression: immediate and mid term results (com-ment). Gut 1998;43:747–751.

34. Pereira-Lima JC, Busnello JV, Saul C, Toneloto EB, Lopes CV,Rynkowski CB, Blaya C. High power setting argon plasma coag-ulation for the eradication of Barrett’s esophagus (comment).Am J Gastroenterol 2000;95:1661–1668.

35. Morris CD, Byrne JP, Armstrong GR, Attwood SE. Prevention ofthe neoplastic progression of Barrett’s oesophagus by endo-scopic argon beam plasma ablation (comment). Br J Surg 2001;88:1357–1362.

36. Basu KK, Pick B, Bale R, West KP, de Caestecker JS. Efficacyand one year follow up of argon plasma coagulation therapy forablation of Barrett’s oesophagus: factors determining persis-tence and recurrence of Barrett’s epithelium (comment). Gut2002;51:776–780.

37. May A, Gossner L, Pech O, Fritz A, Gunter E, Mayer G, Muller H,Seitz G, Vieth M, Stolte M, Ell C. Local endoscopic therapy forintraepithelial high-grade neoplasia and early adenocarcinomain Barrett’s oesophagus: acute-phase and intermediate resultsof a new treatment approach (comment). Eur J GastroenterolHepatol 2002;14:1085–1091.

38. Van Laethem JL, Jagodzinski R, Peny MO, Cremer M, Deviere J.Argon plasma coagulation in the treatment of Barrett’s high-grade dysplasia and in situ adenocarcinoma. Endoscopy 2001;33:257–261.

39. Schulz H, Miehlke S, Antos D, Schentke KU, Vieth M, Stolte M,Bayerdorffer E. Ablation of Barrett’s epithelium by endoscopicargon plasma coagulation in combination with high-dose ome-prazole. Gastrointest Endosc 2000;51:659–663.

40. Overholt BF, Panjehpour M, Haydek JM. Photodynamic therapyfor Barrett’s esophagus: follow-up in 100 patients. GastrointestEndosc 1999;49:1–7.

41. Hage M, Siersema PD, van Dekken H, Steyerberg EW, Har-ingsma J, van de Vrie W, Grool TE, van Veen RL, Sterenborg HJ,Kuipers EJ. 5-aminolevulinic acid photodynamic therapy versusargon plasma coagulation for ablation of Barrett’s oesophagus:a randomised trial. Gut 2004;53:785–790.

42. Buttar NS, Wang KK, Anderson MA, Dierkhising RA, Pacifico RJ,Krishnadath KK, Lutzke LS. The effect of selective cyclooxygen-ase-2 inhibition in Barrett’s esophagus epithelium: an in vitrostudy. J Natl Cancer Inst 2002;94:422–429.

43. Kaur BS, Triadafilopoulos G. Acid- and bile-induced PGE(2) re-lease and hyperproliferation in Barrett’s esophagus are COX-2and PKC-epsilon dependent. Am J Physiol Gastrointest LiverPhysiol 2002;283:G327–G334.

44. Wilson KT, Fu S, Ramanujam KS, Meltzer SJ. Increased expres-sion of inducible nitric oxide synthase and cyclooxygenase-2 inBarrett’s esophagus and associated adenocarcinomas. CancerRes 1998;58:2929–2934.

45. Thun MJ, Namboodiri MM, Calle EE, Flanders WD, Heath CW Jr.Aspirin use and risk of fatal cancer (comment). Cancer Res1993;53:1322–1327.

46. Funkhouser EM, Sharp GB. Aspirin and reduced risk of esoph-ageal carcinoma. Cancer 1995;76:1116–1119.

47. Farrow DC, Vaughan TL, Hansten PD, Stanford JL, Risch HA,Gammon MD, Chow WH, Dubrow R, Ahsan H, Mayne ST,Schoenberg JB, West AB, Rotterdam H, Fraumeni JF Jr, Blot WJ.Use of aspirin and other nonsteroidal anti-inflammatory drugsand risk of esophageal and gastric cancer. Cancer Epidemiol

Biomarkers Prev 1998;7:97–102.
Page 29: American Gastroenterological Association Technical Review on

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

May 2005 AMERICAN GASTROENTEROLOGICAL ASSOCIATION 1499

48. Langman MJ, Cheng KK, Gilman EA, Lancashire RJ. Effect ofanti-inflammatory drugs on overall risk of common cancer: case-control study in general practice research database (comment).BMJ 2000;320:1642–1646.

49. Coogan PF, Rosenberg L, Palmer JR, Strom BL, Zauber AG,Stolley PD, Shapiro S. Nonsteroidal anti-inflammatory drugs andrisk of digestive cancers at sites other than the large bowel.Cancer Epidemiol Biomarkers Prev 2000;9:119–123.

50. Sonnenberg A, Fennerty MB. Medical decision analysis of che-moprevention against esophageal adenocarcinoma. Gastroen-terology 2003;124:1758–1766.

51. Caygill CP, Johnston DA, Lopez M, Johnston BJ, Watson A, ReedPI, Hill MJ. Lifestyle factors and Barrett’s esophagus. Am JGastroenterol 2002;97:1328–1331.

52. Engel LS, Chow WH, Vaughan TL, Gammon MD, Risch HA,Stanford JL, Schoenberg JB, Mayne ST, Dubrow R, Rotterdam H,West AB, Blaser M, Blot WJ, Gail MH, Fraumeni JF Jr. Populationattributable risks of esophageal and gastric cancers. J NatlCancer Inst 2003;95:1404–1413.

53. McKeown KC. Clinical presentation of carcinoma of the oesoph-agus. J Roy Coll Surg Edinbh 1986;31:199–206.

54. Burt M. Management of malignant esophagorespiratory fistula.Chest Surg Clin North Am 1996;6:765–776.

55. Dooley CP, Larson AW, Stace NH, Renner IG, Valenzuela JE,Eliasoph J, Colletti PM, Halls JM, Weiner JM. Double-contrastbarium meal and upper gastrointestinal endoscopy. A compar-ative study. Ann Intern Med 1984;101:538–545.

56. Esfandyari T, Potter JW, Vaezi MF. Dysphagia: a cost analysis ofthe diagnostic approach. Am J Gastroenterol 2002;97:2733–2737.

57. Spechler SJ. AGA technical review on treatment of patients withdysphagia caused by benign disorders of the distal esophagus(comment). Gastroenterology 1999;117:233–254.

58. Mulcahy HE, Fairclough PD. Ultrathin endoscopy in the assess-ment and treatment of upper and lower gastrointestinal tractstrictures. Gastrointest Endosc 1998;48:618–620.

59. Graham DY, Schwartz JT, Cain GD, Gyorkey F. Prospective eval-uation of biopsy number in the diagnosis of esophageal andgastric carcinoma. Gastroenterology 1982;82:228–231.

60. Lal N, Bhasin DK, Malik AK, Gupta NM, Singh K, Mehta SK.Optimal number of biopsy specimens in the diagnosis of carci-noma of the oesophagus. Gut 1992;33:724–726.

61. Jacobson BC, Hirota W, Baron TH, Leighton JA, Faigel DO. Therole of endoscopy in the assessment and treatment of esoph-ageal cancer. Gastrointest Endosc 2003;57:817–822.

62. Zargar SA, Khuroo MS, Jan GM, Mahajan R, Shah P. Prospectivecomparison of the value of brushings before and after biopsy inthe endoscopic diagnosis of gastroesophageal malignancy.Acta Cytol 1991;35:549–552.

63. Faigel DO, Deveney C, Phillips D, Fennerty MB. Biopsy-negativemalignant esophageal stricture: diagnosis by endoscopic ultra-sound. Am J Gastroenterol 1998;93:2257–2260.

64. May A, Gossner L, Pech O, Muller H, Vieth M, Stolte M, Ell C.Intraepithelial high-grade neoplasia and early adenocarcinomain short-segment Barrett’s esophagus (SSBE): curative treat-ment using local endoscopic treatment techniques. Endoscopy2002;34:604–610.

65. Stein HJ, Brucher BL, Sendler A, Siewert JR. Esophageal cancer:patient evaluation and pre-treatment staging. Surg Oncol 2001;10:103–111.

66. Rice TW, Blackstone EH, Adelstein DJ. N1 esophageal carci-noma: the importance of staging and downstaging. J ThoracCardiovasc Surg 2001;121:454–462.

67. Dittler HJ, Pesarini AC, Siewert JR. Endoscopic classification ofesophageal cancer: correlation with the T stage. Gastrointest

Endosc 1992;38:662–668.

68. Saunders HS, Wolfman NT, Ott DJ. Esophageal cancer. Radio-logic staging. Radiol Clin North Am 1997;35:281–294.

69. Quint LE, Hepburn LM, Francis IR, Whyte RI, Orringer MB. Inci-dence and distribution of distant metastases from newly diag-nosed esophageal carcinoma. Cancer 1995;76:1120–1125.

70. Botet JF, Lightdale CJ, Zauber AG, Gerdes H, Urmacher C,Brennan MF. Preoperative staging of esophageal cancer: com-parison of endoscopic US and dynamic CT (comment). Radiol-ogy 1991;181:419–425.

71. Krasna MJ, Reed CE, Nedzwiecki D, Hollis DR, Luketich JD,DeCamp MM, Mayer RJ, Sugarbaker DJ; CALGB Thoracic Sur-geons. CALGB 9380: a prospective trial of the feasibility ofthoracoscopy/laparoscopy in staging esophageal cancer. AnnThorac Surg 2001;71:1073–1079.

72. Choi JY, Lee KH, Shim YM, Lee KS, Kim JJ, Kim SE, Kim BT.Improved detection of individual nodal involvement in squa-mous cell carcinoma of the esophagus by FDG PET. J Nucl Med2000;41:808–815.

73. Rice TW. Clinical staging of esophageal carcinoma. CT, EUS,and PET. Chest Surg Clin North Am 2000;10:471–485.

74. Shami VM, Waxman I. Endoscopic ultrasound-guided fine nee-dle aspiration in esophageal cancer. Min Chir 2002;57:811–818.

75. Reed CE, Mishra G, Sahai AV, Hoffman BJ, Hawes RH. Esoph-ageal cancer staging: improved accuracy by endoscopic ultra-sound of celiac lymph nodes. Ann Thorac Surg 1999;67:319–321; discussion 322.

76. Eloubeidi MA, Wallace MB, Reed CE, Hadzijahic N, Lewin DN,Van Velse A, Leveen MB, Etemad B, Matsuda K, Patel RS,Hawes RH, Hoffman BJ. The utility of EUS and EUS-guided fineneedle aspiration in detecting celiac lymph node metastasis inpatients with esophageal cancer: a single-center experience.Gastrointest Endosc 2001;54:714–719.

77. Kallimanis GE, Gupta PK, al-Kawas FH, Tio LT, Benjamin SB,Bertagnolli ME, Nguyen CC, Gomes MN, Fleischer DE. Endo-scopic ultrasound for staging esophageal cancer, with or with-out dilation, is clinically important and safe (comment). Gastroi-ntest Endosc 1995;41:540–546.

78. Mortensen MB, Pless T, Durup J, Ainsworth AP, Plagborg GJ,Hovendal C. Clinical impact of endoscopic ultrasound-guidedfine needle aspiration biopsy in patients with upper gastrointes-tinal tract malignancies. A prospective study. Endoscopy 2001;33:478–483.

79. Wu LF, Wang BZ, Feng JL, Cheng WR, Liu GR, Xu XH, Zheng ZC.Preoperative TN staging of esophageal cancer: comparison ofminiprobe ultrasonography, spiral CT and MRI. World J Gastro-enterol 2003;9:219–224.

80. Hadzijahic N, Wallace MB, Hawes RH, VanVelse A, LeVeen M,Marsi V, Hoffman BJ, Sahai AV. CT or EUS for the initial stagingof esophageal cancer? A cost minimization analysis (comment).Gastrointest Endosc 2000;52:715–720.

81. Shumaker DA, De Garmo P, Faigel DO. Potential impact ofpreoperative EUS on esophageal cancer management and cost.Gastrointest Endosc 2002;56:391–396.

82. Flanagan FL, Dehdashti F, Siegel BA, Trask DD, Sundaresan SR,Patterson GA, Cooper JD. Staging of esophageal cancer with18F-fluorodeoxyglucose positron emission tomography. Am JRoentgenol 1997;168:417–424.

83. van Westreenen HL, Heeren PA, Jager PL, van Dullemen HM,Groen H, Plukker JT. Pitfalls of positive findings in stagingesophageal cancer with F-18-fluorodeoxyglucose positron emis-sion tomography. Ann Surg Oncol 2003;10:1100–1105.

84. Block MI, Patterson GA, Sundaresan RS, Bailey MS, FlanaganFL, Dehdashti F, Siegel BA, Cooper JD. Improvement in stagingof esophageal cancer with the addition of positron emissiontomography. Ann Thorac Surg 1997;64:770–776; discussion

776–777.
Page 30: American Gastroenterological Association Technical Review on

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

1500 WANG ET AL GASTROENTEROLOGY Vol. 128, No. 5

85. Luketich JD, Schauer PR, Meltzer CC, Landreneau RJ, Urso GK,Townsend DW, Ferson PF, Keenan RJ, Belani CP. Role ofpositron emission tomography in staging esophageal cancer.Ann Thorac Surg 1997;64:765–769.

86. Meltzer CC, Luketich JD, Friedman D, Charron M, Strollo D,Meehan M, Urso GK, Dachille MA, Townsend DW. Whole-bodyFDG positron emission tomographic imaging for staging esoph-ageal cancer comparison with computed tomography. Clin NuclMed 2000;25:882–887.

87. Flamen P, Lerut A, Van Cutsem E, De Wever W, Peeters M,Stroobants S, Dupont P, Bormans G, Hiele M, De Leyn P, VanRaemdonck D, Coosemans W, Ectors N, Haustermans K, Mor-telmans L. Utility of positron emission tomography for the stag-ing of patients with potentially operable esophageal carcinoma.J Clin Oncol 2000;18:3202–3210.

88. Wren SM, Stijns P, Srinivas S. Positron emission tomography inthe initial staging of esophageal cancer. Arch Surg 2002;137:1001–1007.

89. Rasanen JV, Sihvo EI, Knuuti MJ, Minn HR, Luostarinen ME,Laippala P, Viljanen T, Salo JA. Prospective analysis of accuracyof positron emission tomography, computed tomography, andendoscopic ultrasonography in staging of adenocarcinoma ofthe esophagus and the esophagogastric junction. Ann SurgOncol 2003;10:954–960.

90. Stein HJ, Kraemer SJ, Feussner H, Fink U, Siewert JR. Clinicalvalue of diagnostic laparoscopy with laparoscopic ultrasound inpatients with cancer of the esophagus or cardia. J GastrointestSurg 1997;1:167–173.

91. Kassis ES, Nguyen N, Shriver SP, Siegfried JM, Schauer PR,Luketich JD. Detection of occult lymph node metastases inesophageal cancer by minimally invasive staging combined withmolecular diagnostic techniques. J Soc Laparoendosc Surg1998;2:331–336.

92. Krasna MJ, Mao YS, Sonett J, Gamliel Z. The role of thoraco-scopic staging of esophageal cancer patients. Eur J Cardiotho-rac Surg 1999;16(Suppl 1):S31–S33.

93. van Dijkum EJ, de Wit LT, van Delden OM, Kruyt PM, vanLanschot JJ, Rauws EA, Obertop H, Gouma DJ. Staging laparos-copy and laparoscopic ultrasonography in more than 400 pa-tients with upper gastrointestinal carcinoma. J Am Coll Surg1999;189:459–465.

94. Krasna MJ, Jiao X. Thoracoscopic and laparoscopic staging foresophageal cancer. Semin Thorac Cardiovasc Surg 2000;12:186–194.

95. Heath EI, Kaufman HS, Talamini MA, Wu TT, Wheeler J, Heit-miller RF, Kleinberg L, Yang SC, Olukayode K, Forastiere AA. Therole of laparoscopy in preoperative staging of esophageal can-cer (comment). Surg Endosc 2000;14:495–499.

96. Luketich JD, Meehan M, Nguyen NT, Christie N, Weigel T,Yousem S, Keenan RJ, Schauer PR. Minimally invasive surgicalstaging for esophageal cancer. Surg Endosc 2000;14:700–702.

97. Krasna MJ, Jiao X, Mao YS, Sonett J, Gamliel Z, Kwong K,Burrows W, Flowers JL, Greenwald B, White C. Thoracoscopy/laparoscopy in the staging of esophageal cancer: Marylandexperience. Surg Laparosc Endosc Percutan Tech 2002;12:213–218.

98. Riedel M, Hauck RW, Stein HJ, Mounyam L, Schulz C, SchomigA, Siewert JR. Preoperative bronchoscopic assessment of air-way invasion by esophageal cancer: a prospective study. Chest1998;113:687–695.

99. Feldman DR, Kulling DP, Hawes RH, Kay CL, Muckenfuss VR,Cotton PB, Bohning DE, Young JW. MR endoscopy: preliminaryexperience in human trials (erratum appears in Radiology 1997;203:580). Radiology 1997;202:868–870.

00. Kulling D, Feldman DR, Kay CL, Hoffman BJ, Reed CE, Young

JW, Hawes RH. Local staging of esophageal cancer using en-

doscopic magnetic resonance imaging: prospective comparisonwith endoscopic ultrasound. Endoscopy 1998;30:745–749.

01. Wallace MB, Nietert PJ, Earle C, Krasna MJ, Hawes RH, Hoff-man BJ, Reed CE. An analysis of multiple staging managementstrategies for carcinoma of the esophagus: computed tomogra-phy, endoscopic ultrasound, positron emission tomography,and thoracoscopy/laparoscopy. Ann Thorac Surg 2002;74:1026–1032.

02. Yoshida M, Momma K. Endoscopic evaluation of the depth ofinvasion in cases of superficial esophageal cancer in determin-ing indications for endoscopic mucosal resection (in Japanese).Nippon Geka Gakkai Zasshi 2002;103:337–342.

03. Schlemper RJ, Dawsey SM, Itabashi M, Iwashita A, Kato Y,Koike M, Lewin KJ, Riddell RH, Shimoda T, Sipponen P, StolteM, Watanabe H. Differences in diagnostic criteria for esopha-geal squamous cell carcinoma between Japanese and Westernpathologists (comment). Cancer 2000;88:996–1006.

04. Froelicher P, Miller G. The European experience with esopha-geal cancer limited to the mucosa and submucosa. Gastroin-test Endosc 1986;32:88–90.

05. Bonavina L. Early oesophageal cancer: results of a Europeanmulticentre survey. Group Europeen pour l’Etude des Maladiesde l’Oesophage. Br J Surg 1995;82:98–101.

06. Thomson BN, Cade RJ. Oesophagectomy for early adenocarci-noma and dysplasia arising in Barrett’s oesophagus. Aust N ZJ Surg 2003;73:121–124.

07. Ferguson MK, Durkin A. Long-term survival after esophagectomyfor Barrett’s adenocarcinoma in endoscopically surveyed andnonsurveyed patients. J Gastrointest Surg 2002;6:29–36.

08. Stein HJ, Feith M, Mueller J, Werner M, Siewert JR. Limitedresection for early adenocarcinoma in Barrett’s esophagus. AnnSurg 2000;232:733–742.

09. Holscher AH, Bollschweiler E, Schneider PM, Siewert JR. Earlyadenocarcinoma in Barrett’s oesophagus. Br J Surg 1997;84:1470–1473.

10. Streitz JM Jr, Ellis FH Jr, Gibb SP, Balogh K, Watkins E Jr.Adenocarcinoma in Barrett’s esophagus. A clinicopathologicstudy of 65 cases. Ann Surg 1991;213:122–125.

11. Hagen JA, DeMeester SR, Peters JH, Chandrasoma P, De-Meester TR. Curative resection for esophageal adenocarcino-ma: analysis of 100 en bloc esophagectomies. Ann Surg 2001;234:520–530; discussion 530–531.

12. Tseng EE, Wu TT, Yeo CJ, Heitmiller RF. Barrett’s esophaguswith high grade dysplasia: surgical results and long-term out-come—an update. J Gastrointest Surg 2003;7:164–170; dis-cussion 170–171.

13. Rice TW, Falk GW, Achkar E, Petras RE. Surgical management ofhigh-grade dysplasia in Barrett’s esophagus (comment). Am JGastroenterol 1993;88:1832–1836.

14. Makuuchi H, Machimura T, Mizutani K, Shimada H, Kanno K,Tajima T, Mitomi T. Controversy in the treatment of superficialesophageal carcinoma—indications and problems of the proce-dures (in Japanese). Nippon Geka Gakkai Zasshi 1992;93:1059–1062.

15. Endo M, Takeshita K, Inoue H. Endoscopic mucosal resection ofesophageal cancer (in Japanese). Gan To Kagaku Ryoho 1995;22:192–195.

16. Kodama M, Kakegawa T. Treatment of superficial cancer of theesophagus: a summary of responses to a questionnaire onsuperficial cancer of the esophagus in Japan. Surgery 1998;123:432–439.

17. Katada C, Muto M, Manabe T, Boku N, Ohtsu A, Yoshida S.Esophageal stenosis after endoscopic mucosal resection ofsuperficial esophageal lesions. Gastrointest Endosc 2003;57:165–169.

18. Inoue H, Endo M. Endoscopic esophageal mucosal resection

using a transparent tube. Surg Endosc 1990;4:198–201.
Page 31: American Gastroenterological Association Technical Review on

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

May 2005 AMERICAN GASTROENTEROLOGICAL ASSOCIATION 1501

19. Inoue M, Shiozaki H, Tamura S, Monden M. Endoscopic muco-sal resection for early esophageal cancer (in Japanese). NipponRinsho 1996;54:1286–1291.

20. Miyamoto S, Muto M, Hamamoto Y, Boku N, Ohtsu A, Baba S,Yoshida M, Ohkuwa M, Hosokawa K, Tajiri H, Yoshida S. A newtechnique for endoscopic mucosal resection with an insulated-tip electrosurgical knife improves the completeness of resec-tion of intramucosal gastric neoplasms. Gastrointest Endosc2002;55:576–581.

21. Soehendra N, Binmoeller KF, Bohnacker S, Seitz U, Brand B,Thonke F, Gurakuqi G. Endoscopic snare mucosectomy in theesophagus without any additional equipment: a simple tech-nique for resection of flat early cancer. Endoscopy 1997;29:380–383.

22. May A, Gossner L, Behrens A, Kohnen R, Vieth M, Stolte M, EllC. A prospective randomized trial of two different endoscopicresection techniques for early stage cancer of the esophagus.Gastrointest Endosc 2003;58:167–175.

23. Ell C, May A, Gossner L, Pech O, Gunter E, Mayer G, Henrich R,Vieth M, Muller H, Seitz G, Stolte M. Endoscopic mucosalresection of early cancer and high-grade dysplasia in Barrett’sesophagus. Gastroenterology 2000;118:670–677.

24. Buttar NS, Wang KK, Lutzke LS, Krishnadath KK, Anderson MA.Combined endoscopic mucosal resection and photodynamictherapy for esophageal neoplasia within Barrett’s esophagus.Gastrointest Endosc 2001;54:682–688.

25. Seewald S, Akaraviputh T, Seitz U, Brand B, Groth S, MendozaG, He X, Thonke F, Stolte M, Schroeder S, Soehendra N. Cir-cumferential EMR and complete removal of Barrett’s epithe-lium: a new approach to management of Barrett’s esophaguscontaining high-grade intraepithelial neoplasia and intramuco-sal carcinoma. Gastrointest Endosc 2003;57:854–859.

26. Satodate H, Inoue H, Yoshida T, Usui S, Iwashita M, Fukami N,Shiokawa A, Kudo SE. Circumferential EMR of carcinoma arisingin Barrett’s esophagus: case report. Gastrointest Endosc2003;58:288–292.

27. Sharma P, Jaffe PE, Bhattacharyya A, Sampliner RE. Laser andmultipolar electrocoagulation ablation of early Barrett’s adeno-carcinoma: long-term follow-up. Gastrointest Endosc 1999;49:442–446.

28. Gossner L, May A, Stolte M, Seitz G, Hahn EG, Ell C. KTP laserdestruction of dysplasia and early cancer in columnar-linedBarrett’s esophagus. Gastrointest Endosc 1999;49:8–12.

29. Gossner L, Stolte M, Sroka R, Rick K, May A, Hahn EG, Ell C.Photodynamic ablation of high-grade dysplasia and early cancerin Barrett’s esophagus by means of 5-aminolevulinic acid (com-ment). Gastroenterology 1998;114:448–455.

30. Ell C, Gossner L, May A, Schneider HT, Hahn EG, Stolte M,Sroka R. Photodynamic ablation of early cancers of the stomachby means of mTHPC and laser irradiation: preliminary clinicalexperience (comment). Gut 1998;43:345–349.

31. May A, Gossner L, Gunter E, Stolte M, Ell C. Local treatment ofearly cancer in short Barrett’s esophagus by means of argonplasma coagulation: initial experience. Endoscopy 1999;31:497–500.

32. Overholt BF, Panjehpour M. Photodynamic therapy in Barrett’sesophagus: reduction of specialized mucosa, ablation of dys-plasia, and treatment of superficial esophageal cancer. SeminSurg Oncol 1995;11:372–376.

33. Weston AP, Sharma P. Neodymium:yttrium-aluminum garnetcontact laser ablation of Barrett’s high grade dysplasia andearly adenocarcinoma. Am J Gastroenterol 2002;97:2998–3006.

34. Nemoto K, Matsumoto Y, Yamakawa M, Jo S, Ito Y, Oguchi M,Kokubo N, Nishimura Y, Yamada S, Okawa T. Treatment of

superficial esophageal cancer by external radiation therapy

alone: results of a multi-institutional experience. Int J RadiatOncol Biol Phys 2000;46:921–925.

35. Okawa T, Tanaka M, Kita-Okawa M, Nishio M, Kikuchi Y, ShiratoH, Yamada S, Asakawa H, Niibe H, Dokiya T, et al. Superficialesophageal cancer: multicenter analysis of results of definitiveradiation therapy in Japan. Radiology 1995;196:271–274.

36. Maingon P, d’Hombres A, Truc G, Barillot I, Michiels C, BedenneL, Horiot JC. High dose rate brachytherapy for superficial cancerof the esophagus. Int J Radiat Oncol Biol Phys 2000;46:71–76.

37. Rice TW, Adelstein DJ. Precise clinical staging allows treatmentmodification of patients with esophageal carcinoma. Oncology1997;11:58–62.

38. Tachibana M, Kinugasa S, Dhar DK, Tabara H, Masunaga R,Kotoh T, Kubota H, Nagasue N. Prognostic factors in T1 and T2squamous cell carcinoma of the thoracic esophagus. Arch Surg1999;134:50–54.

39. Skinner DB, Little AG, Ferguson MK, Soriano A, Staszak VM.Selection of operation for esophageal cancer based on staging.Ann Surg 1986;204:391–401.

40. Gertsch P, Vauthey JN, Lustenberger AA, Friedlander-Klar H.Long-term results of transhiatal esophagectomy for esophagealcarcinoma. A multivariate analysis of prognostic factors. Cancer1993;72:2312–2319.

41. Tachibana M, Kinugasa S, Yoshimura H, Dhar DK, Nagasue N.Extended esophagectomy with 3-field lymph node dissection foresophageal cancer. Arch Surg 2003;138:1383–1389; discus-sion 1390.

42. Nigro JJ, DeMeester SR, Hagen JA, DeMeester TR, Peters JH,Kiyabu M, Campos GM, Oberg S, Gastal O, Crookes PF, Brem-ner CG. Node status in transmural esophageal adenocarcinomaand outcome after en bloc esophagectomy. J Thorac CardiovascSurg 1999;117:960–968.

43. Zhang DW, Cheng GY, Huang GJ, Zhang RG, Liu XY, Mao YS,Wang YG, Chen SJ, Zhang LZ, Wang LJ, et al. Operable squa-mous esophageal cancer: current results from the East. WorldJ Surg 1994;18:347–354.

44. Watson A. Operable esophageal cancer: current results fromthe West. World J Surg 1994;18:361–366.

45. Pierre AF, Luketich JD. Technique and role of minimally invasiveesophagectomy for premalignant and malignant diseases of theesophagus. Surg Oncol Clin North Am 2002;11:337–350.

46. Luketich JD, Schauer PR, Christie NA, Weigel TL, Raja S, Fer-nando HC, Keenan RJ, Nguyen NT. Minimally invasive esopha-gectomy. Ann Thorac Surg 2000;70:906–911; discussion911–912.

47. Vogel SB, Mendenhall WM, Sombeck MD, Marsh R, WoodwardER. Downstaging of esophageal cancer after preoperative radi-ation and chemotherapy. Ann Surg 1995;221:685–693; dis-cussion 693–695.

48. Nygaard K, Hagen S, Hansen HS, Hatlevoll R, Hultborn R,Jakobsen A, Mantyla M, Modig H, Munck-Wikland E, RosengrenB, et al. Pre-operative radiotherapy prolongs survival in operableesophageal carcinoma: a randomized, multicenter study of pre-operative radiotherapy and chemotherapy. The second Scandi-navian trial in esophageal cancer. World J Surg 1992;16:1104–1109; discussion 1110.

49. Le Prise E, Etienne PL, Meunier B, Maddern G, Ben Hassel M,Gedouin D, Boutin D, Campion JP, Launois B. A randomizedstudy of chemotherapy, radiation therapy, and surgery versussurgery for localized squamous cell carcinoma of the esopha-gus. Cancer 1994;73:1779–1784.

50. Apinop C, Puttisak P, Preecha N. A prospective study of com-bined therapy in esophageal cancer. Hepatogastroenterology1994;41:391–393.

51. Walsh TN, Noonan N, Hollywood D, Kelly A, Keeling N, Hennessy

TP. A comparison of multimodal therapy and surgery for esoph-
Page 32: American Gastroenterological Association Technical Review on

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

1502 WANG ET AL GASTROENTEROLOGY Vol. 128, No. 5

ageal adenocarcinoma (comment). N Engl J Med1996;335:462–467.

52. Bosset JF, Gignoux M, Triboulet JP, Tiret E, Mantion G, Elias D,Lozach P, Ollier JC, Pavy JJ, Mercier M, Sahmoud T. Chemora-diotherapy followed by surgery compared with surgery alone insquamous-cell cancer of the esophagus. N Engl J Med 1997;337:161–167.

53. Kelsen DP, Ginsberg R, Pajak TF, Sheahan DG, Gunderson L,Mortimer J, Estes N, Haller DG, Ajani J, Kocha W, Minsky BD,Roth JA. Chemotherapy followed by surgery compared with sur-gery alone for localized esophageal cancer (comment). N EnglJ Med 1998;339:1979–1984.

54. Urba S, Orringer MB, Turrisi A. Randomized trial of preoperativechemoradiation versus surgery alone in patients with locore-gional esophageal carcinoma. J Clin Oncol 2001;12:305–313.

55. Urschel JD, Vasan H. A meta-analysis of randomized controlledtrials that compared neoadjuvant chemoradiation and surgeryto surgery alone for resectable esophageal cancer. Am J Surg2003;185:538–543.

56. Wolfe WG, Vaughn AL, Seigler HF, Hathorn JW, Leopold KA,Duhaylongsod FG. Survival of patients with carcinoma of theesophagus treated with combined-modality therapy. J ThoracCardiovasc Surg 1993;105:749–755; discussion 755–756.

57. Minsky BD, Pajak TF, Ginsberg RJ, Pisansky TM, Martenson J,Komaki R, Okawara G, Rosenthal SA, Kelsen DP. INT 0123(Radiation Therapy Oncology Group 94-05) phase III trial ofcombined-modality therapy for esophageal cancer: high-doseversus standard-dose radiation therapy. J Clin Oncol 2002;20:1167–1174.

58. Cooper JS, Guo MD, Herskovic A, Macdonald JS, Martenson JAJr, Al-Sarraf M, Byhardt R, Russell AH, Beitler JJ, Spencer S,Asbell SO, Graham MV, Leichman LL. Chemoradiotherapy oflocally advanced esophageal cancer: long-term follow-up of aprospective randomized trial (RTOG 85-01). Radiation TherapyOncology Group. JAMA 1999;281:1623–1627.

59. Nash CL, Gerdes H. Methods of palliation of esophageal andgastric cancer. Surg Oncol Clin North Am 2002;11:459–483.

60. Moses FM, Peura DA, Wong RKH, Johnson LF. Palliative dilationof esophageal carcinoma. Gastrointest Endosc 1985;31:61–63.

61. Aste H, Munizzi F, Martines H, Pugliese V. Esophageal dilationin malignant dysphagia. Cancer 1985;56:2713–2715.

62. Lundell L, Leth R, Lind T, Lonroth H, Sjovall M, Olbe L. Palliativeendoscopic dilatation in carcinoma of the esophagus andesophagogastric junciton. Acta Chir Scand 1989;155:179–184.

63. Hernandez LJ, Jacobson JW, Harris MS. Comparison among theperforation rates of Maloney, balloon, and savary dilation ofesophageal strictures. Gastrointest Endosc 2000;51:460–462.

64. Anand BS, Saeed ZA, Michaletz PA, Winchester CB, Doherty MA,Liem JH, Graham DY. A randomized comparison of dilatationalone versus dilatation plus laser in patients receiving chemo-therapy and external beam radiation for esophageal carcinoma.Dig Dis Sci 1998;43:2255–2260.

65. Segalin A, Bonavina L, Carazzone A, Ceriani C, Peracchia A.Improving results of esophageal stenting: a study on 160 con-secutive unselected patients. Endoscopy 1997;29:701–709.

66. Baron TH. Expandable metal stents for the treatment of cancer-ous obstruction of the gastrointestinal tract. N Engl J Med2001;344:1681–1687.

67. Kozarek RA, Ball TJ, Brandabur JJ, Patterson DJ, Low D, Hill L,Raltz S. Expandable versus conventional esophageal prosthe-ses: easier insertion may not preclude subsequent stent-re-lated problems. Gastrointest Endosc 1996;43:204–208.

68. Ramirez F, Dennert B, Zierer ST, Sanowski RA. Esophageal

self-expandable metallic stents–indications, practice, tech-

niques, and complications: results of a national survey.Gastrointest Endosc 1997;45:360–364.

69. Sanyika C, Corr P, Haffejee A. Palliative treatment of oesopha-geal carcinoma—efficacy of plastic versus self-expandablestents. South Afr Med J 1999;89:640–643.

70. Knyrim K, Wagner HJ, Bethge N, Keymling M, Vakil N. A con-trolled trial of an expansile metal stent for palliation of esoph-ageal obstruction due to inoperable cancer. N Engl J Med 1993;329:1302–1307.

71. De Palma GD, Di Matteo E, Romano G, Fimmano A, RondinoneG, Catanzano C. Plastic prosthesis versus expandable metalstents for palliation of inoperable esophageal thoracic carci-noma: a controlled prospective study. Gastrointest Endosc1996;43:478–482.

72. Siersema PD, Hop WC, Dees J, Tilanus HW, van BlankensteinM. Coated self-expanding metal stents versus latex prosthesesfor esophagogastric cancer with special reference to prior radi-ation and chemotherapy: a controlled, prospective study. Gas-trointest Endosc 1998;47:113–120.

73. Roseveare CD, Patel P, Simmonds N, Goggin PM, Kimble J,Shepherd HA. Metal stents improve dysphagia, nutrition andsurvival in malignant oesophageal stenosis: a randomized con-trolled trial comparing modified Gianturco Z-stents with plasticAtkinson tubes. Eur J Gastroenterol Hepatol 1998;10:653–657.

74. O’Donnell CA, Fullarton GM, Watt E, Lennon K, Murrary GD,Moss JG. Randomized clinical trial comparing self-expandingmetallic stents with plastic endoprostheses in the palliation ofoesophageal cancer. Br J Surg 2002;89:985–992.

75. Bethge N, Vakil N. A prospective trial of a new self-expandingplastic stent for malignant esophageal obstruction. Am J Gas-troenterol 2001;96:1350–1354.

76. Decker P, Lippler J, Decker D, Hirner A. Use of the Polyflex stentin the palliative therapy of esophageal carcinoma: results in 14cases and review of the literature. Surg Endosc 2001;15:1444–1447.

77. Dormann AJ, Eisendrath P, Wigginghaus B, Huchzermeyer H,Deviere J. Palliation of esophageal carcinoma with a new self-expanding plastic stent. Endoscopy 2003;35:207–211.

78. Costamagna G, Shah SK, Tringali A, Mutignani M, Perri V,Riccioni ME. Prospective evaluation of a new self-expandingplastic stent for inoperable esophageal strictures. Surg Endosc2003;17:891–895.

79. White RE, Mungatana C, Topazian M. Esophageal stent place-ment without fluoroscopy. Gastrointest Endosc 2001;53:348–351.

80. Austin AS, Khan Z, Cole AT, Freeman JG. Placement of esoph-ageal self-expanding metallic stents without fluoroscopy. Gas-trointest Endosc 2001;54:357–359.

81. Vakil N, Morris AI, Marcon N, Segalin A, Peracchia A, Bethge N,Zuccaro G, Bosco JJ, Jones WF. A prospective, randomized,controlled trial of covered expandable metal stents in the palli-ation of malignant esophageal obstruction at the gastroesoph-ageal junction. Am J Gastroenterol 2001;96:1791–1796.

82. Adam A, Ellul J, Watkinson AF, Tan BS, Morgan RA, SaundersMP, Mason RC. Palliation of inoperable esophageal carcinoma:a prospective randomized trial of laser therapy and stent place-ment. Radiology 1997;202:344–348.

83. Siersema P, Hop W, van Blankenstein M, Dees J. A new designmetal stent (Flamingo stent) for palliation of malignant dyspha-gia: a prospective study. Gastrointest Endosc 2000;51:139–145.

84. Sabharwal T, Hamady MS, Chui S, Atkinson S, Mason R, AdamA. A randomised prospective comparison of the Flamingo Wall-stent and Ultraflex stent for palliation of dysphagia associatedwith lower third oesophageal carcinoma. Gut 2003;52:922–

926.
Page 33: American Gastroenterological Association Technical Review on

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

May 2005 AMERICAN GASTROENTEROLOGICAL ASSOCIATION 1503

85. Siersema P, Hop W, van Blankenstein M, van Tilburg AJ, Bac DJ,Homs MY, Kuipers EJ. A comparison of 3 types of covered metalstents for the palliation of patients with dysphagia caused byesophagogastric carcinoma: a prospective, randomized study.Gastrointest Endosc 2001;54:145–153.

86. Baron TH. A practical guide for choosing an expandable metalstent for GI malignancies: is a stent by any other name still astent? Gastrointest Endosc 2001;54:269–272.

87. Ell C, May A, Hahn EG. Gianturco-Z stents in the palliativetreatment of malignant esophageal obstruction and esophago-tracheal fistulas. Endoscopy 1995;27:495–500.

88. Lagattolla NR, Rowe PH, Anderson H, Dunk AA. Restentingmalignant oesophageal strictures. Br J Surg 1998;85:261–263.

89. Simsek H, Oksuzoghi G, Akhan O. Endoscopic Nd:YAG lasertherapy for esophageal wallstent occlusion due to tumor in-growth. Endoscopy 1996;28:400.

90. Scheider DM, Siemens M, Cirocco M, Haber GB, Kandel G,Kortan P, Marcon NE. Photodynamic therapy for the treatmentof tumor ingrowth in expandable esophageal stents. Endoscopy1997;29:271–274.

91. Kinsman KJ, DeGregorio BT, Katon RM, Morrison K, Saxon RR,Keller FS, Rosch J. Prior radiation and chemotherapy increasethe risk of life-threatening complications after insertion of me-tallic stents for esophagogastric malignancy. Gastrointest En-dosc 1996;43:196–203.

92. Bethge N, Sommer N, Vakil N. A prospective trial of self-expand-ing metal stents in the palliation of malignant esophageal stric-tures near the upper esophageal sphincter. Gastrointest En-dosc 1997;45:300–303.

93. Sumiyoshi T, Gotoda T, Muro K, Rembacken B, Goto M, Sum-iyoshi Y, Ono H, Saito D. Morbidity and mortality after self-expandable metallic stent placement in patients with progres-sive or recurrent esophageal cancer after chemoradiotherapy.Gastrointest Endoscopy 2003;57:882–885.

94. Raijman I, Siddique I, Lynch P. Does chemoradiation therapyincrease the incidence of complications with self-expandingcoated stents in the management of malignant esophagealstrictures. Am J Gastroenterol 1997;92:2192–2196.

95. Raijman I, Siddique I, Ajani J, Lynch P. Palliation of malignantdysphagia and fistulae with coated expandable metal stents:experience with 101 patients. Gastrointest Endosc 1998;48:172–179.

96. Ludwig D, Dehne A, Burmester E, Wiedemann GJ, Stange EF.Treatment of unresectable carcinoma of the esophagus or thegastroesophageal junction by mesh stents with or without ra-diochemotherapy. Int J Oncol 1998;13:583–588.

97. Nishimura Y, Nagata K, Katano S, Hirota S, Nakamura K,Higuchi F, Soejima T, Sai H; Japanese Society for EsophagealDiseases. Severe complications in advanced esophagealcancer treated with radiotherapy after intubation of esopha-geal stents: a questionnaire survey of the Japanese Societyfor Esophageal Diseases. Int J Radiat Oncol Biol Phys 2003;56:1327–1332.

98. Dua K, Kozarek RA, Kim J, Evans J, Medda BK, Lang I, HoganWJ, Shaker R. Self-expanding metal esophageal stent withanti-reflux mechanism. Gastrointest Endosc 2001;53:603–613.

99. Laasch HU, Marriott A, Wilbraham L, Tunnah S, England RE,Martin DF. Effectiveness of open versus antireflux stents forpalliation of distal esophageal carcinoma and prevention ofsymptomatic gastroesophageal reflux. Radiology 2002;225:359–365.

00. Sawant D, Moghissi K. Management of unresectable oesopha-geal cancer: a review of 537 patients. Eur J Cardiothorac Surg

1994;8:113–116.

01. Siersema P, Schrauwen SL, van Blankenstein M, SteyerbergEW, van der Gaast A, Tilanus HW, Dees J; Rotterdam Esopha-geal Tumor Study Group. Self-expanding metal stents for com-plicated and recurrent esophagogastric cancer. GastrointestEndosc 2001;54:579–586.

02. Morgan RA, Ellul J, Denton ER, Glynos M, Mason RC, Adam A.Malignant esophageal fistulas and perforations: managementwith plastic-covered metallic endoprostheses. Radiology 1997;204:527–532.

03. Freitag L, Tekolf E, Steveling H, Donovan TJ, Stamatis G. Man-agement of malignant esophagotracheal fistulas with airwaystenting and double stenting. Chest 1996;110:1155–1160.

04. Low DE, Kozarek RA. Comparison of conventional and wiremesh expandable prostheses and surgical bypass in patientswith malignant esophagorespiratory fistulas. Ann Thorac Surg1998;65:919–923.

05. Watkinson A, Ellul J, Entwisle K, Farrugia M, Mason R, Adam A.Plastic-covered metallic endoprostheses in the management ofoesophageal perforation in patients with oesophageal carci-noma. Clin Radiol 1995;50:304–309.

06. Payne-James JJ, Spiller RC, Misiewicz JJ, Silk DB. Use of etha-nol-induced tumor necrosis to palliate dysphagia in patientswith esophagogastric cancer. Gastrointest Endosc 1990;36:43–46.

07. Nwokolo CU, Payne-James JJ, Silk DB, Misiewicz JJ, Loft DE.Palliation of malignant dysphagia by ethanol induced tumournecrosis. Gut 1994;35:299–203.

08. Chung SC, Leong HT, Choi CY, Leung JW, Li AK. Palliation ofmalignant oesophageal obstruction by endoscopic alcohol in-jection. Endoscopy 1994;26:275–277.

09. Moreira LS, Coelho RC, Sadala RU, Dani R. The use of ethanolinjection under endoscopic control to palliate dysphagia causedby esophagogastric cancer. Endoscopy 1994;26:311–314.

10. Guitron A, Adalid R, Huerta F, Macias M, Sanchez-Navarrete M.Palliative treatment of esophageal cancer with transendoscopicinjection of alcohol (in Spanish). Rev Gastroenterol Mex 1996;61:208–211.

11. Monga SP, Wadleigh R, Sharma A, Adib H, Strader D, Singh G,Harmon JW, Berlin M, Monga DK, Mishra L. Intratumoral therapyof cisplatin/epinephrine injectable gel for palliation in patientswith obstructive esophageal cancer. Am J Clin Oncol 2000;23:386–392.

12. Harbord M, Dawes RF, Barr H, Giovannini M, Viens P, EysseleinV, Mishra L, Orenberg EK, Bown SG. Palliation of patients withdysphagia due to advanced esophageal cancer by endoscopicinjection of cisplatin/epinephrine injectable gel. GastrointestEndosc 2002;56:644–651.

13. Johnston J, Fleischer D, Petrini J, Nord HJ. Palliative bipolarelectrocoagulation therapy of obstruction of esophageal cancer.Gastrointest Endosc 1987;33:349–353.

14. Jensen DM, Machicado G, Randall G, Tung LA, English-Zych S.Comparison of low-power YAG laser and BICAP tumor probe forpalliation of esophageal cancer strictures. Gastroenterology1988;94:1263–1270.

15. McIntyre AS, Morris DL, Sloan RL, Robertson CS, Harrison J,Burnham WR, Atkinson M. Palliative therapy of malignantesophageal stricture with the bipolar tumor probe and pros-thetic tube. Gastrointest Endosc 1989;35:531–535.

16. Heindorff H, Wojdemann M, Bisgaard T, Svendsen LB. Endo-scopic palliation of inoperable cancer of the oesophagus orcardia by argon electrocoagulation. Scand J Gastroenterol1998;33:21–23.

17. Mitty RD, Cave DR, Birkett DH. One-stage retrograde approachto Nd:YAG laser palliation of esophageal carcinoma. Endoscopy1996;28:350–355.

18. Loizou LA, Grigg D, Atkinson M, Robertson CS, Bown S. A

prospective comparison of laser therapy and intubation in en-
Page 34: American Gastroenterological Association Technical Review on

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

1504 WANG ET AL GASTROENTEROLOGY Vol. 128, No. 5

doscopic palliation for malignant dysphagia. Gastroenterology1991;100:1303–1310.

19. Lightdale CJ, Zimbalist E, Winawer SJ. Outpatient managementof esophageal cancer with endoscopic Nd:YAG laser. Am JGastroenterol 1987;82:46–50.

20. Naveau S, Chiesa A, Poynard T, Chaput JC. Endoscopic Nd-YAGlaser therapy as palliative treatment for esophageal and cardialcancer. Parameters affecting long-term outcome. Dig Dis Sci1990;35:294–301.

21. Bourke MJ, Hope RL, Chu G, Gillespie PE, Bull C, O’Rourke I,Williams SJ. Laser palliation of inoperable malignant dysphagia:initial and at death. Gastrointest Endosc 1996;43:29–32.

22. Spencer GM, Thorpe SM, Sargeant IR, Blackman GM, Solano J,Tobias JS, Bown SG. Laser and brachytherapy in the palliationof adenocarcinoma of the oesophagus and cardia. Gut 1996;39:726–731.

23. Sargeant IR, Tobias JS, Blackman G, Thorpe S, Glover JR, BownSG. Radiotherapy enhances laser palliation of malignant dys-phagia: a randomised study. Gut 1997;40:362–369.

24. Tan CC, Freeman JG, Holmes GK, Benghiat A. Laser therapycombined with brachytherapy for the palliation of malignantdysphagia. Singapore Med J 1998;39:202–207.

25. Lightdale CJ, Heier SK, Marcon NE, McCaughan JS Jr, Gerdes H,Overholt BF, Sivak MV Jr, Stiegmann GV, Nava HR. Photody-namic therapy with porfimer sodium versus thermal ablationtherapy with Nd:YAG laser for palliation of esophageal cancer: amulticenter randomized trial. Gastrointest Endosc 1995;42:507–512.

26. Heier SK, Rothman KA, Heier LM, Rosenthal WS. Photodynamictherapy for obstructing esophageal cancer: light dosimetry andrandomized comparison with Nd:YAG laser therapy. Gastroen-terology 1995;109:63–72.

27. Maier A, Tomaselli F, Matzi V, Woltsche M, Anegg U, Fell B,Rehak P, Pinter H, Smolle-Juttner FM. Comparison of 5-amin-olaevulinic acid and porphyrin photosensitization for photody-namic therapy of malignant bronchial stenosis: a clinical pilotstudy. Lasers Surg Med 2002;30:12–17.

28. Gevers AM, Macken E, Hiele M, Rutgeerts P. A comparison oflaser therapy, plastic stents, and expandable metal stents forpalliation of malignant dysphagia in patients without a fistula.Gastrointest Endosc 1998;48:383–388.

29. Sihvo EI, Pentikainen T, Luostarinen ME, Ramo JO, Salo J.Inoperable adenocarcinoma of the oesophagogastric junction: acomparative clinical study of laser coagulation versus self-ex-panding metallic stents with special reference to cost analysis.Eur J Surg Oncol 2002;28:711–715.

30. Konigsrainer A, Riedmann B, De Vries A, Ofner D, Spechten-hauser B, Aigner F, Fritsch E, Margreiter R. Expandable metalstents versus laser combined with radiotherapy for palliation ofunresectable esophageal cancer: a prospective randomizedtrial. Hepatogastroenterology 2000;47:724–727.

31. Dallal HJ, Smith GD, Grieve DC, Ghosh S, Penman ID, PalmerKR. A randomized trial of thermal ablative therapy versus ex-pandable metal stents in the palliative treatment of patientswith esophageal carcinoma. Gastrointest Endosc 2001;54:549–557.

32. Angelini G, Pasini AF, Ederle A, Castagnini A, Talamini G, Bu-lighin G. Nd:YAG laser versus polidocanol injection for palliationof esophageal malignancy: a prospective, randomized study.Gastrointest Endosc 1991;37:607–610.

33. Carazzone A, Bonavina L, Segalin A, Ceriani C, Peracchia A.Endoscopic palliation of oesophageal cancer: results of a pro-spective comparison of Nd:YAG laser and ethanol injection. EurJ Surg 1999;165:351–356.

34. Schattner M. Enteral nutritional support of the patient with

cancer: route and role. J Clin Gastroenterol 2003;36:297–302.

35. Braga M, Gianotti L, Gentilini O, Parisi V, Salis C, Di CV. Earlypostoperative enteral nutrition improves gut oxygenation andreduces costs compared with total parenteral nutrition. seecomment. Crit Care Med 2001;29:242–248.

36. Roberge C, Tran M, Massoud C, Poiree B, Duval N, Damecour E,Frout D, Malvy D, Joly F, Lebailly P, Henry-Amar M. Quality of lifeand home enteral tube feeding: a French prospective study inpatients with head and neck or oesophageal cancer. Br J Cancer2000;82:263–269.

37. Stockeld D, Fagerberg J, Granstrom L, Backman L. Percutane-ous endoscopic gastrostomy for nutrition in patients with oe-sophageal cancer. Eur J Surg 2001;167:839–844.

38. Pickhardt PJ, Rohrmann CA Jr, Cossentino MJ. Stomal metas-tases complicating percutaneous endoscopic gastrostomy: CTfindings and the argument for radiologic tube placement. Am JRoentgenol 2002;179:735–739.

39. Wildi SM, Cox MH, Clark LL, Turner R, Hawes RH, Hoffman BJ,Wallace MB. Assessment of health state utilities and quality oflife in patients with malignant esophageal dysphagia. Am JGastroenterol 2004;99:1044–1049.

40. Eckardt VF, Kanzler G, Bernhard G. Life expectancy and cancerrisk in patients with Barrett’s esophagus: a prospective con-trolled investigation. Am J Med 2001;111:33–37.

41. Ortiz A, Martinez de Haro LF, Parrilla P, Morales G, Molina J,Bermejo J, Liron R, Aguilar J. Conservative treatment versusantireflux surgery in Barrett’s oesophagus: long-term results ofa prospective study. Br J Surg 1996;83:274–278.

42. Cameron AJ, Ott BJ, Payne WS. The incidence of adenocarci-noma in columnar-lined esophagus. N Engl J Med 1985;313:857–859.

43. Katz D, Rothstein R, Schned A, Dunn J, Seaver K, Antonioli D.The development of dysplasia and adenocarcinoma during en-doscopic surveillance of Barrett’s esophagus. Am J Gastroen-terol 1998;93:536–541.

44. Moghissi K, Sharpe DA, Pender D. Adenocarcinoma and Bar-rett’s oesophagus. A clinico-pathological study. Eur J Cardiotho-rac Surg 1993;7:126–131.

45. Ovaska J, Miettinen M, Kivilaakso E. Adenocarcinoma arising inBarrett’s esophagus. Dig Dis Sci 1989;34:1336–1339.

46. McDonald ML, Trastek VF, Allen MS, Deschamps C, PairoleroPC. Barretts’s esophagus: does an antireflux procedure reducethe need for endoscopic surveillance? J Thorac Cardiovasc Surg1996;111:1135–1138; discussion 1139–1140.

47. Iftikhar SY, James PD, Steele RJ, Hardcastle JD, Atkinson M.Length of Barrett’s oesophagus: an important factor in thedevelopment of dysplasia and adenocarcinoma (see com-ments). Gut 1992; 33:1155–1158.

48. Rana PS, Johnston DA. Incidence of adenocarcinoma and mor-tality in patients with Barrett’s oesophagus diagnosed between1976 and 1986: implications for endoscopic surveillance. DisEsophagus 2000;13:28–31.

49. Hameeteman W, Tytgat GN, Houthoff HJ, van den Tweel JG.Barrett’s esophagus: development of dysplasia and adenocar-cinoma (see comments). Gastroenterology 1989;96:1249–1256.

50. Streitz JM Jr, Ellis FH Jr, Tilden RL, Erickson RV. Endoscopicsurveillance of Barrett’s esophagus: a cost-effectiveness com-parison with mammographic surveillance for breast cancer.Am J Gastroenterol 1998;93:911–915.

51. Drewitz DJ, Sampliner RE, Garewal HS. The incidence of ade-nocarcinoma in Barrett’s esophagus: a prospective study of170 patients followed 4.8 years (see comments). Am J Gastro-enterol 1997;92:212–215.

52. Ferraris R, Bonelli L, Conio M, Fracchia M, Lapertosa G, Aste H.Incidence of Barrett’s adenocarcinoma in an Italian population:

an endoscopic surveillance programme. Gruppo Operativo per
Page 35: American Gastroenterological Association Technical Review on

3

3

3

3

CR

ptmM

fv

May 2005 AMERICAN GASTROENTEROLOGICAL ASSOCIATION 1505

lo Studio delle Precancerosi Esofagee (GOSPE). Eur JGastroenterol Hepatol 1997;9:881–885.

53. Weston AP, Banerjee SK, Sharma P, Tran TM, Richards R,Cherian R. p53 protein overexpression in low grade dysplasia(LGD) in Barrett’s esophagus: immunohistochemical markerpredictive of progression. Am J Gastroenterol 2001;96:1355–1362.

54. Heitmiller RF, Redmond M, Hamilton SR. Barrett’s esophaguswith high-grade dysplasia. An indication for prophylactic esoph-agectomy. Ann Surg 1996;224:66–71.

55. Edwards MJ, Gable DR, Lentsch AB, Richardson JD. The ratio-nale for esophagectomy as the optimal therapy for Barrett’sesophagus with high-grade dysplasia. Ann Surg 1996;223:585–589; discussion 589–591.

56. Weston AP, Sharma P, Topalovski M, Richards R, Cherian R, K

Dixon A. Long-term follow-up of Barrett’s high-grade dysplasia.Am J Gastroenterol 2000;95:1888–1893.

Address requests for reprints to: Chair, Clinical Practice and Economicsommittee, AGA National Office, c/o Membership Department, 4930 Delay Avenue, Bethesda, Maryland 20814. Fax: (301) 654-5920.This literature review and the recommendations herein were pre-

ared for the American Gastroenterological Association Clinical Prac-ice and Economics Committee. The paper was approved by the Com-ittee on January 31, 2005, and by the AGA Governing Board onarch 20, 2005.The Clinical Practice and Economics Committee acknowledges the

ollowing individuals whose critiques of this review paper providedaluable guidance to the authors: David E. Fleischer, MD, Richard A.

ozarek, MD, and Gottumukkala S. Raju, MD, MRCP.