adenocarcinoma esophageal

download adenocarcinoma esophageal

of 12

Transcript of adenocarcinoma esophageal

  • 8/11/2019 adenocarcinoma esophageal

    1/12

    H. MatthewsMary Koshy, Natia Esiashvilli, Jerome C. Landry, Charles R. Thomas, Jr. and Richard

    Presentation and Progression, Work-up, and Surgical ApproachesMultiple Management Modalities in Esophageal Cancer: Epidemiology,

    doi: 10.1634/theoncologist.9-2-1372004, 9:137-146.The Oncologist

    http://theoncologist.alphamedpress.org/content/9/2/137

    located on the World Wide Web at:The online version of this article, along with updated information and services, is

    http://theoncologist.alphamedpress.org/content/9/2/137http://theoncologist.alphamedpress.org/content/9/2/137http://theoncologist.alphamedpress.org/content/9/2/137
  • 8/11/2019 adenocarcinoma esophageal

    2/12

    Multiple Management Modalities in Esophageal Cancer:Epidemiology, Presentation and Progression, Work-up,

    and Surgical Approaches

    MARY KOSHY,a NATIA ESIASHVILLI,a JEROME C. LANDRY,a CHARLES R. THOMAS JR.,b

    RICHARD H. MATTHEWSc,d

    aEmory University School of Medicine, Department of Radiation Oncology, Atlanta, Georgia, USA;bUTHSCSA/San Antonio Cancer Institute, Department of Radiation Oncology, San Antonio, Texas, USA;

    cBeth Israel/Deaconess Medical Center, Harvard Medical School, Department of Radiation Oncology, Boston,

    Massachusetts, USA; dBoston VA Health Care Radiation Oncology Service, Boston, Massachusetts, USA

    Key Words.Esophageal carcinoma Multimodal treatment Survival Surgeries

    ABSTRACT

    Annually, approximately 13,200 people in the U.S. are

    diagnosed with esophageal cancer and 12,500 die of this

    malignancy. Of new cases, 9,900 occur in men and 3,300

    occur in women. In part I of this two-part series, we

    explore the epidemiology, presentation and progression,

    work-up, and surgical approaches for esophageal cancer.

    In the 1960s, squamous cell cancers made up greater than

    90% of all esophageal tumors. The incidence of

    esophageal adenocarcinomas has risen considerably over

    the past two decades, such that they are now more preva-

    lent than squamous cell cancer in the western hemisphere.

    Despite advances in therapeutic modalities for this dis-

    ease, half the patients are incurable at presentation, and

    overall survival after diagnosis is grim. Evolving knowl-

    edge regarding the etiology of esophageal carcinoma may

    lead to better preventive methods and treatment options

    for early stage superficial cancers of the esophagus. The

    use of endoscopic ultrasound and the developing role of

    positron emission tomography have led to better diagnos-

    tic accuracy in this disease. For years, the standard of care

    for esophageal cancer has been surgery; there are several

    variants of the surgical approach. We will discuss

    combined modality approaches in part II of this series.

    The Oncologist 2004;9:137-146

    The Oncologist 2004;9:137-146 www.TheOncologist.com

    Correspondence: Richard H. Matthews, M.D., Ph.D., Beth Israel/Deaconess Medical Center, Harvard Medical School, 330Brookline Avenue, Boston, Massachusetts 02215, USA. Telephone: 617-232-9500, ext 4457 or ext 5628; Fax: 617-524-0643; e-mail: [email protected] Received May 13, 2003; accepted for publication September 18, 2003.AlphaMed Press 1083-7159/2004/$12.00/0

    TheOncologistGastrointestinalCancer

    LEARNING OBJECTIVES

    After completing this course, the reader will be able to:

    1. Describe the epidemiology, work-up, and staging of esophageal cancer.

    2. Identify the disease presentation, progression, and prognostic factors for esophageal cancer.

    3. Discuss the surgical approach and management of esophageal cancer.

    Access and take the CME test online and receive one hour of AMA PRA category 1 credit at CME.TheOncologist.comCMECME

    This material is protected by U.S. Copyright law.

    Unauthorized reproduction is prohibited.

    For reprints contact: [email protected]

  • 8/11/2019 adenocarcinoma esophageal

    3/12

    Koshy, Esiashvilli, Landry et al. 138

    INTRODUCTION

    It has been estimated that in 2001 there were 13,200

    new cases and 12,500 deaths attributable to esophageal

    carcinoma in the U.S. [1]. Table 1 presents new cancer

    cases and deaths in the U.S. for a few selected sites for

    comparison. We use the ratio of deaths to new cases as a

    rough index of the overall degree of success of treatments

    for various cancers. We can, thus, anticipate that, nation-

    wide and for all presentations and all management

    approaches, the curative success in dealing with cancer of

    the esophagus is less than 10%. Cancer of the esophagus

    occurs much less frequently than cancers of the breast,

    prostate, lung, or rectum (Table 1). With reference to ratios

    of deaths to new cases, roughly 80% of cancers of the

    breast, prostate, and rectum can be treated successfully in

    the U.S. Lung cancer, however, resembles esophageal can-

    cer in that less than 10% of patients in our country are

    treated successfully. Lung cancer occurs less commonly

    than breast or prostate cancers, but accounts for more can-

    cer deaths than the other four sites combined.

    There is an approximate 3:1 male predominance in

    esophageal cancer incidence and death. One factor in the

    poor results obtained for the treatment of esophageal carci-

    nomas is that half the cases are unresectable or metastatic at

    presentation.

    The cervical esophagus begins at the cricopharyngeus

    muscle at the level of the cricoid cartilage and extends 6 cm

    to enter the thoracic inlet. The intrathoracic esophagus

    extends for another 20-25 cm to the gastroesophageal junc-tion. The American Joint Committee on Cancer (AJCC)

    divides the esophagus into four regions: cervical, upper tho-

    racic, midthoracic, and lower thoracic [2].

    The predominant histological types of esophageal car-

    cinoma are squamous cell carcinoma (SCC) and adenocar-

    cinoma (AC). Less common histologies include adenoid

    cystic, mucoepidermoid, adenosquamous, undifferentiated,

    and malignant melanoma, all of which have poor prog-

    noses. Small cell carcinoma can also occur in the esopha-

    gus, and it has a course similar to that in the lung [3].

    Nonepithelial tumors in the esophagus are rare; the most

    common type is leiomyosarcoma and the most common

    metastatic source to the esophagus is the breast [4]. In the

    1960s, more than 90% of all esophageal tumors were SCCs.

    The incidence of esophageal AC has risen considerably

    over the past two decades such that it is now more preva-

    lent than SCC in the western hemisphere. The rise in

    esophageal AC, particularly in the distal esophagus, has

    been paralleled by a rise in gastric cardia AC. This leads to

    the concept of gastroesophageal junction AC.

    The standard of care for esophageal cancer had been

    surgery alone for many years. There are several different

    surgical approaches in use, with various advantages and

    disadvantages and with differences in procedure preference

    in different parts of the world. We review the epidemiol-

    ogy, etiology, presentation and work-up, and surgical

    approaches for esophageal cancer in this paper.

    EPIDEMIOLOGY AND ETIOLOGYThe incidence of esophageal cancer varies considerably

    with geographic location and also, to some extent, among

    ethnic groups within a common area. Some of the highest

    rates occur in northern China and northern Iran, where inci-

    dence exceeds 100 in 100,000 individuals; in the U.S., the

    incidence is less than 5 per 100,000, although rates are

    nearly quadruple for African Americans [4]. In Linxian,

    Hunan province, China, esophageal cancer is endemic and

    has been directly related to nitrosamines and inversely

    related to consumption of riboflavin, nicotinic acid, magne-

    sium, and zinc [5]. SCC predominates in African Americans

    over Caucasians by a ratio of 6:1, and AC has the opposite

    preponderance, occurring in Caucasians over African

    Americans at a ratio of 4:1 [6]. Nearly 30 years ago, AC

    accounted for only 15% of cases of esophageal cancer, but

    the incidence of AC has increased more than 350% since

    1970, surpassing SCC since 1990; this increase is also seen,

    to a lesser extent, in African Americans [7, 8]. In the Far

    East, no increase in AC has been observed, and SCC con-

    tinues to be the more common histology [9]. In the western

    world, there is less impact from dietary factors, such as

    nitrosamines, due to different food preservation techniques,

    and the primary etiology of esophageal cancer is the use of

    tobacco and alcohol, which have a synergistic effect; they

    are very strong risk factors for SCC and moderate risk fac-

    tors for AC [10]. Tobacco exposure has been linked to a ten-

    fold higher risk for esophageal SCC in heavy smokers

    relative to nonsmokers, and the risk is directly related to the

    duration of exposure [7, 11]. In contrast, smoking has been

    linked to only a two- to threefold greater risk for esophageal

    AC in smokers relative to nonsmokers [8, 12]. There is a

    greater than multiplicative effect between smoking and

    alcohol consumption that occurs in SCC; for AC, the effect

    Table 1. Comparison of new cases and deaths for selected cancersites, 2001 [109]

    Cancer site New cases Deaths Ratio

    Esophagus 13,200 12,500 0.95

    Breast 193,700 40,600 0.21

    Prostate 198,100 31,500 0.16

    Lung 169,500 157,400 0.93

    Rectum 37,200 8,600 0.23

  • 8/11/2019 adenocarcinoma esophageal

    4/12

    139 Esophageal Carcinomas

    is only additive [13]. The exception to this may be the con-

    sumption of hard liquor, which has been found to have a

    stronger association with AC than with SCC [14]. The rela-

    tive risk of esophageal AC remains high up to 30 years after

    smoking cessation, in contrast to the significant decline in

    risk of SCC within a decade of smoking cessation [15].

    Several studies have linked obesity to the risk for

    esophageal AC. Increasing body mass index (obesity) corre-

    lates with increasing risk for esophageal AC, but decreasing

    risk for esophageal SCC [4]. Obesity promotes gastroe-

    sophageal reflux disease (GERD) by increasing intra-abdom-

    inal pressure; GERD, in turn, promotes the formation of

    Barretts esophagus (BE)a metaplastic precursor of AC

    [16, 17]. Multiple studies have shown that the bacterium

    Helicobacter pylori has an inverse relationship with the risk

    for esophageal cancer but causes a greater risk for gastric

    cancer [18].H. pylori is thought to protect against the risk of

    esophageal AC because it promotes achlorhydria, implyinglower acid production and reflux [15]. As rates of H. pylori

    infection have decreased in the U.S. and Europe, there has

    been a parallel rise in the incidence of GERD (an indepen-

    dent risk factor for esophageal AC) and BE [9]. Prior aerodi-

    gestive tract malignancies predispose to a higher risk for

    esophageal cancer, primarily through field cancerization.

    Ten percent of second primary cancers in patients with prior

    oropharyngeal cancer or lung cancer occur in the esophagus

    [19, 20]. Chronic inflammation and stasis, which occur with

    strictures caused by caustic injury and achalasia, are long-

    term risks for esophageal SCC; in addition, patients withtylosis, which is inherited in an autosomal dominant fashion,

    and Plummer Vinson syndrome have a definite greater risk

    for esophageal SCC [21].

    The definition of BE has evolved from simply a colum-

    nar-lined esophagus to at least 3 cm of columnar lining or

    metaplasia in the esophagus [21, 22]. Approximately 0.5%-

    2% of adults in the western world have BE, with prevalences

    greater in Caucasians and men and with increasing age until

    the eighth decade of life [23, 24]. In a review of 51,311

    patients at the Mayo Clinic, the length of BE epithelium did

    not increase significantly with age, suggesting that BE does

    not progress with age and is latent for several years before

    being discovered [24]. GERD affects up to 44% of the gen-

    eral population in the U.S., but only 10% of people with

    GERD develop BE [25, 26]. Recurrent symptoms of reflux

    have been associated with a 7.7 times greater risk for

    esophageal AC, with more frequent, more severe, and

    longer-lasting reflux resulting in a 43.5 times higher risk for

    esophageal AC [26]. Patients with BE have a 40-fold greater

    risk or 0.5% risk per patient year of developing esophageal

    AC [27]. A middle-aged patient who develops BE has a 10%-

    15% risk of developing esophageal AC during his lifetime

    [28]. Specific risk factors that predispose for the progression

    of BE to AC include hiatal hernia of at least 3 cm in length,

    length of BE, and the presence of dysplasia [29]. In addition,

    increased bile acid exposure is thought to exacerbate

    esophageal mucosal injury and to promote the neoplastic

    process [23, 30].

    Esophageal cancers, both SCC and AC, typically have

    aberrant cell-cycle regulation. Mutations occur in onco-

    genes such as EGFR, erbB-2, and cyclin D1, and in tumor

    suppressors such as 3p(FHIT), Rb, p53, p16, p14ARF, and

    telomerase, which affect the G1 restriction point. Cyclin D1

    directly regulates phosphorylation of the Rb protein at the

    G1 restriction point facilitating G1/S transit. About 40%-

    60% of esophageal carcinomas and 30% of premalignant

    lesions overexpress cyclin D1. Cancers that retain normal

    Rb expression typically overexpress cyclin D1, whereas

    those that lack normal Rb have normal cyclin D1 levels.

    The p53 gene product regulates cell-cycle progression,DNA repair, neovascularization, and apoptosis; 50%-80%

    of esophageal carcinomas havep53 mutations. Malignancy

    is facilitated by inactivation of the growth constraints

    imposed by the Rb and p53 suppressor pathways, but also

    requires activation of telomerase, a ribonucleoprotein that

    adds hexamer DNA repeats to the ends of chromosomes to

    prevent loss of telomere length in DNA replication.

    Elevated telomerase expression is found in high-grade dys-

    plasia as well as virtually all esophageal carcinomas.

    Several of the individual aforementioned factors have been

    shown to carry prognostic significance [21].Part of the problem in dealing with esophageal carcino-

    mas is that half the patients present with unresectable or

    metastatic cancers; if a screening program could detect dis-

    ease at an earlier stage, there could be a greater possibility

    of cure. In comparison, there is considerable evidence that

    the combination of physical examination with mammogra-

    phy used as a screening tool decreases the death rate from

    breast cancer [31]. However, several studies have failed to

    demonstrate the benefit of a decreased death rate due to

    lung cancer from screening chest x-rays and physical exam-

    inations [32]. Unfortunately, the value and cost-effective-

    ness of endoscopic surveillance for esophageal cancers has

    not been demonstrated. In 11 screening studies with 1,127

    patients with BE, only 3.5% actually progressed to cancer

    [33]. To date, there is no solid evidence that screening

    reduces the esophageal AC death rate [34].

    Proponents argue that screening, to be productive, should

    be focused on patients with multiple risk factors: GERD,

    Caucasian race, male gender, age greater than 50 years, and

    long duration of symptoms. Small studies have suggested

    that surveillance can identify neoplasms that are at an ear-

    lier stage and, thus, potentially curable [35-38]. However,

  • 8/11/2019 adenocarcinoma esophageal

    5/12

    Koshy, Esiashvilli, Landry et al. 140

    these hopeful results may be confounded by lead time bias,

    length time bias, and pseudodisease [37]. Furthermore,

    even if surveillance were effective, it is unlikely to impact

    mortality soon because few patients with esophageal cancer

    are diagnosed via surveillance programs. Less than 5% of

    patients with esophageal cancer were known to have had BE

    before they sought help for symptoms of cancer, and up to

    40% had no prior history of GERD [39, 40]. Most patients

    with BE die from unrelated causes, and the presence of BE

    does not change life expectancy or overall survival [41, 42].

    Currently, the American College of Gastroenterology recom-

    mends that surveillance endoscopy for BE be guided by the

    presence of dysplasia. If dysplasia is not present, endoscopy

    is recommended every 3 years, and if low-grade dysplasia is

    present, it is recommended that endoscopy be done every 6

    months for 1 year and yearly thereafter if no progression is

    seen [43]. If high-grade dysplasia is present, this finding

    should be confirmed by an experienced pathologist and the

    patient should be offered either esophagectomy or intensive

    surveillance (endoscopy every 3 months) [43]. The technique

    of four-quadrant biopsies taken every 2 cm of BE segment

    during endoscopy is optimal [44]. Dysplasia is currently the

    best pathological predictor of cancer development.

    DISEASE PRESENTATION, PROGRESSION, AND

    PROGNOSTIC FACTORS

    The most common presenting symptoms of esophageal

    cancer are dysphagia and weight loss. Less common symp-

    toms include odynophagia, cachexia, melena, retrosternalpain, and hoarseness [45]. Cancers of the esophagus must

    involve at least 75% of the circumference before the sensa-

    tion of food sticking or blockage is experienced. As noted

    above, about one-half of esophageal cancer patients present

    with locally advanced unresectable disease or distant

    metastasis. The extent of wall penetration and lymph node

    metastases are more important prognosticators of survival

    than tumor length or functional obstruction [21].

    Esophageal AC spreads via transverse penetration

    through the full thickness of the wall, whereas SCC tends to

    spread linearly in a submucosal fashion [46]. Esophageal

    cancer spreads through extensive lymphatic channels with an

    erratic pattern including skip metastases being observed in

    autopsy specimens; in addition, up to 71% of frozen tissue

    sections classified as tumor free by conventional histopathol-

    ogy have been positive for lymphatic micrometastases when

    tested by immunohistochemistry [47]. This micrometastatic

    disease has been shown to be a significant independent

    adverse prognostic factor for relapse-free survival and over-

    all survival [48]. Routine use of immunohistochemistry in

    conjunction with extensive lymph node sampling has been

    advocated as an advance over the traditional staging work-up.

    Histologic tumor type is an independent prognostic fac-

    tor in esophageal cancer patients who have had surgical

    treatment. Siewert et al. analyzed 1,059 patients who had

    undergone resection and found that overall survival at 5 years

    was 46.8% for the AC group versus 37.4% for the SCC

    group; patients with early-stage AC had a much lower inci-

    dence of lymph node involvement than their SCC counter-

    parts, and it was hypothesized this was due to occlusion of

    lymphatic channels secondary to the inflammatory changes

    of GERD in AC [49]. Tumor size less than 5 cm, upper

    third esophageal location, female sex, and age less than 65

    years have all been noted to positively influence outcome

    [50, 51]. Conversely, weight loss, low Karnofsky perfor-

    mance status, deep ulceration of tumor, sinus tract forma-

    tion, and fistula formation have all been found to be poor

    prognostic factors [21, 50].

    STAGING AND WORK-UP

    The AJCC has established a staging system for

    esophageal cancer that is based on the tumor-node-metasta-

    sis (TNM) system and is, in essence, a pathological staging

    system [2]. Table 2 illustrates the AJCC staging classifica-

    tion of esophageal cancer. Only the depth of penetration is

    taken into account for the T staging, not the length of the

    tumor, extent of involved circumference, or degree of

    lumen narrowing. Clinical staging takes into account the

    amount of disease that is present before treatment and is

    based on history, physical examination, biopsy, laboratory

    studies, endoscopic examination, and imaging such asendoscopic ultrasound (EUS), computed tomography (CT)

    scan and positron emission tomography (PET) scan.

    Pathologic staging is based on examination of the surgically

    resected esophagus and lymph nodes. Spread to the celiac

    lymph nodes is considered a site of distant metastasis, as

    are cervical nodes occurring with an esophageal primary

    elsewhere than in the cervical esophagus [2].

    The appropriate work-up for a patient suspected of hav-

    ing esophageal cancer should include a thorough history and

    physical exam with special attention to the supraclavicular

    and cervical lymph nodes. Fine-needle aspiration should be

    performed on any palpable cervical lymph node to rule out

    extrathoracic spread of disease. Blood tests, a chest x-ray,

    and a double-contrast barium swallow should follow. CT

    scans of the chest and upper abdomen should be obtained to

    further characterize the lesion and evaluate for metastatic dis-

    ease. Esophagoscopy, with the intent of tissue biopsy and

    detailed characterization of the lesion, should be pursued

    with an emphasis on how far the lesion is from the incisors

    and the squamocolumnar junction; in addition, the presence

    of esophagitis or BE should be noted [21]. Brushings should

    be obtained before biopsy, and these two procedures together

  • 8/11/2019 adenocarcinoma esophageal

    6/12

    141 Esophageal Carcinomas

    achieve a diagnostic accuracy of 99% [52]. In lesions that

    appear borderline for the endoscopist, the use of Lugols

    iodine can help distinguish normal mucosa by selectively

    staining it and leaving the abnormal mucosa identifiable forbiopsy [53]. Finally, patients with upper- or middle-third

    esophageal cancers or symptoms of hoarseness or hemopty-

    sis should have a bronchoscopy to rule out laryngeal nerve

    involvement or tracheobronchial fistula [21].

    Conventional CT scans can accurately determine

    resectability 65%-88% of the time [54, 55]. CT accurately

    predicts T stage in 70% of cases and N stage in about 50%-

    70% of cases [54-56]. Secondary to poor sensitivity, CT may

    not be the optimal modality to evaluate celiac lymph nodes

    and T4 disease or response to neoadjuvant chemoradiother-

    apy. The sensitivity and specificity of CT for pathologically

    positive celiac lymph nodes are 30% and 93%, respectively,

    and for T4 disease, are 25% and 94%, respectively [57, 58].

    With respect to assessing response after chemotherapy, no

    correlation has been found between tumor volume reduction

    on serial CT examinations and pathological assessment of

    tumor response or patient survival [55].

    EUS has improved the preoperative staging of

    esophageal cancer, particularly in regard to T and N staging

    [59]. In a meta-analysis of 27 studies, the accuracy of EUS

    for T staging was 90% and for N staging was 80%, and a

    further review of the literature supports an overall accuracy

    for EUS of approximately 85% for T staging and 75% for N

    staging [59-61]. However, the utility of EUS in detecting dis-

    tant metastases other than celiac metastases is low, secondary

    to limited depth of penetration, and therefore, CT is still anecessary and complementary part of the staging work-up

    [62]. EUS detection of celiac lymph node disease correlates

    with overall survival and has a sensitivity of 70%-80% and a

    specificity of 97% [57, 63-65]. EUS has also been useful in

    evaluating recurrence after resection; in a study evaluating 40

    patients with suspected recurrence, the sensitivity and speci-

    ficity of EUS were found to be 95% and 80%, respectively

    [66]. One problem with EUS specificity in that study of pos-

    sible recurrences were the false positives reported for anasto-

    motic thickening because of inflammation. Other limitations

    to EUS include its decreased sensitivity in the presence of

    stenosis and its unreliability in assessment of the response to

    neoadjuvant therapy [67, 68]. In three studies involving 196

    patients, the accuracy of EUS after neoadjuvant therapy

    ranged from 27%-48% for T staging and 38%-71% for N

    staging [68-70]. This significantly lower accuracy is attribut-

    able to the failure of EUS to distinguish between tumor and

    postradiation fibrosis and inflammation.

    Fluorodeoxyglucose (FDG)-PET is rapidly evolving as

    an important tool in the noninvasive staging of patients with

    esophageal cancer. Kole et al. prospectively evaluated 26

    patients and found the diagnostic accuracies in determining

    Table 2. AJCC staging classification of esophageal cancer

    Primary tumor (T) Regional lymph nodes Distant metastasis (M)

    TX: Primary tumor cannot be assessed NX: Regional lymph nodes cannot be assessed MX: Presence of distant metastasis cannotbe assessed

    T0: No evidence of primary tumor N0: No regional lymph node metastasis M0: No distant metastasis

    N1: Regional lymph node metastasis M1: Distant metastasis

    Tis: Carcinoma in situ

    T1: Tumor invades lamina Tumors of the lower thoracic esophagus

    T2: Tumor invades muscularis propria M1a: Metastasis to celiac lymph nodes

    T3: Tumor invades adventitia M1b: Other distant metastasis

    T4: Tumor invades adjacent structures

    Stage grouping Tumors of the midthoracic esophagus

    Stage I: T1N0M0 M1a: Not applicable

    Stage IIA: T2N0M0; T3N0M0 M1b: Nonregional lymph nodes and/orStage IIB: T1N1M0; T2N1M0 other distant metastasis

    Stage III: T3N1M0; T4, any N, M0

    Stage IV: Any T, any N, M1 Tumors of the upper thoracic esophagus

    Stage IVA: Any T, any N, M1a M1a: Metastasis in cervical lymph nodes

    Stage IVB: Any T, any N, M1b M1b: Other distant metastasis

    For the cervical esophagus, the cervical nodes (including the supraclavicular nodes) are considered regional; for the intrathoracic esophagus, themediastinal and perigastric lymph nodes (excluding the celiac nodes) are considered regional.

    T1 has been further subdivided into T1m, cancer confirmed to the mucosa, and T1sm, cancer invading the submucosa.

  • 8/11/2019 adenocarcinoma esophageal

    7/12

    Koshy, Esiashvilli, Landry et al. 142

    resectability to be 65% for CT and 88% for PET; for CT and

    PET together, an accuracy of 92% was achieved [54]. The

    accuracy of PET in detecting primary tumors is 78%; nodal

    metastases are visualized by PET in 86% of cases [71, 72].

    A particular advantage of FDG-PET has been its greater accu-

    racy in detecting distant metastatic disease than CT alone or

    combined with EUS (86% versus 62%), but it has a lower

    accuracy in detecting local nodal disease than CT alone or

    combined with EUS (48% versus 69%) [73-75]. FDG-PET

    has also demonstrated greater accuracy than other methods in

    detecting recurrences other than perianastomotic recurrences

    [76]. The quantitative decrease in FDG uptake seen after

    neoadjuvant therapy has been correlated with histopathologic

    assessment of viable tumor cells, time to disease progression,

    and overall survival [77-79]. FDG-PET may assist in deter-

    mining response to neoadjuvant therapy, which subsequently

    could aid in the selection of patients who will benefit from

    surgery and avoid excess morbidity and mortality in those

    who are unlikely to respond [80]. Minimally invasive staging,

    including thoracoscopic and laparoscopic methods, remains

    the most accurate method to detect distant metastases and

    determine resectability. Although PET had a better accuracy

    than CT in detecting distant metastases (84% versus 63%),

    minimally invasive staging was even better, determining

    resectability with an accuracy of 97%, compared with only

    61% with EUS and CT [81, 82].

    SURGERY

    Esophagectomy remains the standard of care for thetreatment of early-stage tumors confined to the esophagus

    and paraesophageal region. There is controversy about

    whether en bloc resection with extended lymph node dis-

    section confers any advantage in curative management. The

    two most common approaches for definitive resection are

    the transthoracic esophagectomy (TTE) and the transhiatal

    esophagectomy (THE). TTE is the most common approach

    used worldwide, whereas THE is more common in the

    western world. Advantages of TTE include better visual-

    ization, access, and resection of the upper two-thirds of the

    esophagus and mediastinal disease and avoidance of blind

    blunt dissection with tumors of the midthoracic esophagus

    [21]. The rate of resectability of esophageal cancer is

    reported to range from 60%-90%, hospital mortality after

    resection ranges from 1.4%-23%, and the resulting 5-year

    overall survival rate ranges from 10%-25% [83-86]. Five-

    year survival rates reported for stage I esophageal cancer

    range from 80%-94% and, for stage III, rates range from

    10%-14% [83, 84]. Advantages of THE include the avoid-

    ance of morbidity, including the respiratory compromise

    associated with thoracotomy, and the fact that if a leak does

    occur it will be in the neck where it is more accessible [82].

    Operative mortality has ranged from 1% to 4%, and the 5-

    year survival rate has ranged from 20%-25% overall, with

    that of stage I as high as 65% and that of stage III as low as

    10% [87-91]. Two randomized trials and a meta-analysis

    concluded that there were no significant differences

    between TTE and THE with respect to overall morbidity,

    operative mortality, and long-term survival [87, 92].

    For tumors below the tracheal bifurcation, radical en bloc

    esophagectomy or two-field dissection has been recom-

    mended [93, 94]. The surgical objective is to achieve exten-

    sive margins, with removal of the upper abdominal,

    retroperitoneal, and mediastinal lymph nodes, the peri-

    cardium, periaortic tissue, bilateral pleura, azygous vein, and

    thoracic duct [93, 95]. Proponents of this radical operation

    argue that the technique is a better attempt to obtain the

    benefits of a resection with no residual disease (R0).

    Achievement of R0 status has been reported to be a prognos-

    tic factor for long-term survival [96, 97]. In a study of 500

    patients undergoing TTE, patients with an R0 resection had

    a 5-year survival rate of 29% in contrast to those with micro-

    scopic residual (R1) or macroscopic residual (R2) disease,

    none of whom survived 5 years [84]. Some have doubted the

    value of the en bloc approach, theorizing that esophageal

    cancer systemically spreads at onset and lymphatic dissec-

    tion is, therefore, palliative rather than curative [98]. The

    counter argument from those viewing the disease progression

    as sequential, from primary site to lymph nodes and then to

    distant sites, is that dramatically low rates of local recurrence

    are better achieved with en bloc resection, which results inbetter long-term survival [94, 99, 100]. Local recurrence

    rates after en bloc resection have ranged from 1%-8%, oper-

    ative mortality rates have ranged from 3%-6%, and the 5-

    year overall survival rate has ranged from 30%-52% [98,

    100, 101]. A survival advantage for patients with advanced

    primary and nodal disease with en bloc resection has been

    claimed in small retrospective studies [94, 101, 102]. The

    superior survival evinced in node-positive patients lends cre-

    dence to the theory that disease progression is sequential and

    that removal of the involved nodal groups is therapeutic and

    potentially curative. Reservations in asserting a superiority

    for the en bloc resection include dependence on retrospective

    rather than randomized study data, with selection bias and

    stage migration as a result of more accurate staging (Will

    Rogers effect). In a study byHagen et al., patients selected

    to undergo en bloc resection were chosen based on the pres-

    ence of limited disease and good general health, while

    those with poor physiologic reserve and more advanced

    disease underwent THE, clearly a selection bias [102]. The

    selection of young patients with limited disease and excel-

    lent performance statuses for en bloc resection has been

    corroborated by others [103].

  • 8/11/2019 adenocarcinoma esophageal

    8/12

    143 Esophageal Carcinomas

    Japanese surgeons routinely extend the en bloc or two-

    field dissection into the neck, mediastinum, and upper

    abdomen, creating a three-field dissection. This is based on

    the finding that cervical nodal metastases occur in up to

    46.3% of upper-third tumors and in up to 27% of lower-

    third tumors in patients undergoing three-field lymph node

    dissection [104]. Two randomized trials have been done in

    Japan comparing three-field radical surgery with two-field

    radical surgery. In one study, the 5-year survival rate was

    significantly better with the three-field procedure (48% ver-

    sus 33%) and, in the other study, a survival difference was

    not noted; however, in both studies, there was a selection

    bias toward younger, earlier stage patients being allocated

    to three-field radical surgery [105, 106]. In the western

    world, three-field surgery has also been investigated in the

    institutional setting.Lerut et al. analyzed 37 patients under-

    going three-field radical resection and found that the extended

    field improved staging accuracy (30% of patients had patho-logic cervical nodes and 17% of gastroesophageal junction

    tumors had positive cervical nodes) but did not change over-

    all survival [107].Altorki et al., in the largest western study of

    three-field radical resection, prospectively analyzed 80

    patients undergoing the procedure and found an operative

    mortality of 5%, cervical nodal involvement of 36%, and a 5-

    year survival rate of 51% (88% in node-negative patients and

    25% in node-positive patients) [108]. Differences in dissec-

    tion technique around the recurrent laryngeal nerve may

    account for the low rates of recurrent laryngeal nerve injury,

    only 6% in that study, dramatically lower than the up to 70%

    observed in Japanese reports [96, 108].

    An impairment to quality of life with respect to speech

    and swallowing has been chronicled in up to 20% of

    patients for up to 5 years postoperatively and must also be

    a serious consideration. Both en bloc dissection and three-

    field resection may offer a survival benefit but, before these

    techniques can be widely adopted, we need more experi-

    ence and randomized studies to substantiate the benefit of

    such radical surgery.

    CONCLUSIONS

    The overall results of treatment for esophageal cancer in

    our country continue to be poor, with over 90% of the new

    cases each year eventually succumbing to this disease. In part

    I of this two-part series we have covered the issues of epi-

    demiology, work-up and staging, prognostic factors, and sur-

    gical treatment. More radical surgical approaches may havethe potential of gaining an advantage in local-regional control,

    and perhaps ultimately survival, although perhaps at a cost in

    terms of the morbidity of the procedure. In part II we will

    explore the integration of chemotherapy and radiation therapy

    into multimodal management approaches to the disease.

    ACKNOWLEDGMENTS

    The authors disclaim any proprietary interest and

    acknowledge that there have been no grants, equipment, or

    drugs provided.

    REFERENCES

    1 Daly JM, Karnell LH, Menck HR. National Cancer Data Base

    report on esophageal carcinoma. Cancer 1996;78:1820-1828.

    2 Greene FL, Page DL, Fleming ID et al., eds. AJCC Cancer

    Staging Manual, Sixth Edition. New York: Springer, 2002:1-294.

    3 Imai T, Sannohe Y, Okano H. Oat cell carcinoma (apudoma)

    of the esophagus: a case report. Cancer 1978;41:358-364.

    4 Fisher S, Brady L. Esophagus. In: Perez CA, Brady LJ, eds.

    Principles and Practice of Radiation Oncology, Third Edition.

    Philadelphia, PA: Lippincott-Raven, 1998:1241-1256.

    5 Cheng KK. The etiology of esophageal cancer in Chinese.Semin Oncol 1994;21:411-415.

    6 Blot WJ, Fraumeni JF Jr. Trends in esophageal cancer mor-

    tality among US blacks and whites. Am J Public Health

    1987;77:296-298.

    7 Blot WJ, McLaughlin JK. The changing epidemiology of

    esophageal cancer. Semin Oncol 1999;26(suppl 15):2-8.

    8 Devesa SS, Blot WF, Fraumeni JF Jr. Changing patterns in

    the incidence of esophageal and gastric carcinoma in the

    United States. Cancer 1998;83:2049-2053.

    9 Law S, Wong J. Changing disease burden and management

    issues for esophageal cancer in the Asia-Pacific region.

    J Gastroenterol Hepatol 2002;17:374-381.

    10 Blot WJ. Esophageal cancer trends and risk factors. Semin

    Oncol 1994;21:403-410.

    11 Doll R, Peto R. Mortality in relation to smoking: 20 years

    observations on male British doctors. Br Med J 1976;2:1525-

    1536.

    12 Zhang ZF, Kurtz RC, Sun M. Adenocarcinomas of the esoph-

    agus and gastric cardia: medical conditions, tobacco, alcohol,

    and socioeconomic factors. Cancer Epidemiol Biomarkers

    Prev 1996;5:761-768.

    13 Kabat GC, Ng SK, Wynder EL. Tobacco, alcohol intake, anddiet in relation to adenocarcinoma of the esophagus and

    gastric cardia. Cancer Causes Control 1993:4:123-132.

    14 Vaughan TL, Davis S, Kristal A et al. Obesity, alcohol, and

    tobacco as risk factors for cancers of the esophagus and gas-

    tric cardia: adenocarcinoma versus squamous cell carcinoma.

    Cancer Epidemiol Biomarkers Prev 1995;4:85-92.

    15 Heath EI, Limburg PJ, Hawk ET et al. Adenocarcinoma of the

    esophagus: risk factors and prevention. Oncology (Huntingt)

    2000;14:507-514; discussion 518-520, 522-523.

    16 Day JP, Richter JE. Medical and surgical conditions predis-

    posing to gastroesophageal reflux disease. Gastroenterol

    Clin North Am 1990;19:587-607.

  • 8/11/2019 adenocarcinoma esophageal

    9/12

    Koshy, Esiashvilli, Landry et al. 144

    17 Chow WH, Finkle WD, McLaughlin JK et al. The relation of

    gastroesophageal reflux disease and its treatment to adeno-

    carcinomas of the esophagus and gastric cardia. JAMA

    1995;274:474-477.

    18 Chow WH, Blaser MJ, Blot WJ et al. An inverse relation

    between cagA+ strains ofHelicobacter pylori infection and

    risk of esophageal and gastric cardia adenocarcinoma.Cancer Res 1998;58:588-590.

    19 Leon X, Quer M, Diez S et al. Second neoplasm in patients

    with head and neck cancer. Head Neck 1999;21:204-210.

    20 Levi F, Randimbison L, Te VC et al. Second primary cancers

    in patients with lung carcinoma. Cancer 1999;86:186-190.

    21 Schrump D, Altorki N, Forastiere A et al. Cancer of the esopha-

    gus. In: DeVita VT Jr, Hellman S, Rosenberg SA, eds. Cancer:

    Principles and Practice of Oncology, Sixth Edition. Philadelphia,

    PA: Lippincott-Williams & Wilkins, 2001:1319-1342.

    22 Sampliner RE. Updated guidelines for the diagnosis, surveil-

    lance, and therapy of Barretts esophagus. Am J Gastroenterol

    2002;97:1888-1895.

    23 Jankowski JA, Harrison RF, Perry I et al. Barretts metaplasia.

    Lancet 2000;356:2079-2085.

    24 Spechler SJ. Barretts esophagus. Semin Oncol 1994;21:431-

    437.

    25 Gallup Organization. A Gallup Survey on Heartburn Across

    America. Princeton, NJ: Gallup Organization, 1988.

    26 Tharalson EF, Martinez SD, Garewal HS et al. Relationship

    between rate of change in acid exposure along the esophagus

    and length of Barretts epithelium. Am J Gastroenterol

    2002;97;851-856.

    27 Shaheen N, Ransohoff DF. Gastroesophageal reflux, Barrett

    esophagus, and esophageal cancer: scientific review. JAMA

    2002;287:1972-1981.

    28 DeMeester TR. Clinical biology of the Barretts metaplasia,

    dysplasia to carcinoma sequence. Surg Oncol 2001;10:91-102.

    29 Weston AP, Badr AS, Hassanein RS. Prospective multivari-

    ate analysis of clinical, endoscopic, and histological factors

    predictive of the development of Barretts multifocal high-

    grade dysplasia or adenocarcinoma. Am J Gastroenterol

    1999;94:3413-3419.

    30 Jankowski J, Hopwood D, Pringle R et al. Increased expres-

    sion of epidermal growth factor receptors in Barretts esoph-

    agus associated with alkaline reflux: a putative model for

    carcinogenesis. Am J Gastroenterol 1993;88:402-408.

    31 Humphrey LL, Helfand M, Chan BK et al. Breast cancer

    screening: a summary of the evidence for the U.S. Preventive

    Services Task Force. Ann Intern Med 2002;137:347-360.

    32 Berlin NI, Buncher CR, Fontana RS et al. The National

    Cancer Institute Cooperative Early Lung Cancer Detection

    Program. Results of the initial screen (prevalence). Early

    lung cancer detection: Introduction. Am Rev Respir Dis

    1984;130:545-549.

    33 Reid BJ, Levine DS, Longton G et al. Predictors of progres-

    sion to cancer in Barretts esophagus: baseline histology and

    flow cytometry identify low- and high-risk patient subsets.

    Am J Gastroenterol 2000;95:1669-1676.

    34 Sontag SJ. Preventing death of Barretts cancer: does frequent

    surveillance endoscopy do it? Am J Med 2001;111(suppl

    8A):137S-141S.

    35 Ferguson M, Durkin A. Long-term survival after esophagec-

    tomy for Barretts adenocarcinoma in endoscopically surveyed

    and nonsurveyed patients. J Gastrointest Surg 2002;6:29-35;

    discussion 36.

    36 Fitzgerald RC, Saeed IT, Khoo D et al. Rigorous surveillance

    protocol increases detection of curable cancers associated with

    Barretts esophagus. Dig Dis Sci 2001;46:1892-1898.

    37 Corley DA, Levin TR, Habel LA et al. Surveillance and sur-

    vival in Barretts adenocarcinomas: a population-based study.

    Gastroenterology 2002;122:633-640.

    38 Robertson CS, Mayberry JF, Nicholson DA et al. Value of

    endoscopic surveillance in the detection of neoplastic

    change in Barretts oesophagus. Br J Surg 1988;75:760-763.39 Ruol A, Parenti A, Zaninotto G et al. Intestinal metaplasia is

    the probable common precursor of adenocarcinoma in bar-

    rett esophagus and adenocarcinoma of the gastric cardia.

    Cancer 2000;88:2520-2528.

    40 Dulai GS, Guha S, Kahn KL et al. Preoperative prevalence

    of Barretts esophagus in esophageal adenocarcinoma: a

    systematic review. Gastroenterology 2002;122:26-33.

    41 Playford RJ. Endoscopic surveillance of patients with Barretts

    oesophagus. Gut 2002;51:314-315.

    42 Shaheen NJ. Does surveillance endoscopy improve life

    expectancy in those with Barretts esophagus? Gastroenterology

    2001;121:1516-1518.

    43 Spechler SJ. Clinical practice. Barretts esophagus. N Engl

    J Med 2002;346:836-842.

    44 Levine DS, Haggitt RC, Blount PL et al. An endoscopic

    biopsy protocol can differentiate high-grade dysplasia

    from early adenocarcinoma in Barretts esophagus.

    Gastroenterology 1993;105:40-50.

    45 Ojala K, Sorri M, Jokinen K et al. Symptoms of carcinoma

    of the oesophagus. Med J Aust 1982;1:384-385.

    46 Danoff B, Cooper J, Klein M. Primary adenocarcinoma of

    the upper oesophagus. Clin Radiol 1978;29:519-522.

    47 Hosch SB, Stoecklein NH, Pichlmeier U et al. Esophageal

    cancer: the mode of lymphatic tumor cell spread and its

    prognostic significance. J Clin Oncol 2001;19:1970-1975.

    48 Izbicki JR, Hosch SB, Pichlmeier U et al. Prognostic value

    of immunohistochemically identifiable tumor cells in lymph

    nodes of patients with completely resected esophageal

    cancer. N Engl J Med 1997;337:1188-1194.

    49 Siewert JR, Stein HJ, Feith M et al. Histologic tumor type is

    an independent prognostic parameter in esophageal cancer:

    lessons from more than 1,000 consecutive resections at a sin-

    gle center in the Western world. Ann Surg 2001;234:360-367;

    discussion 368-369.

  • 8/11/2019 adenocarcinoma esophageal

    10/12

    145 Esophageal Carcinomas

    50 Hussey DH, Barakley T, Bloedorn F. Carcinoma of the

    esophagus. In: Fletcher GH, ed. Textbook of Radiotherapy,

    Third Edition. Philadelphia, PA: Lea & Febiger, 1980:688.

    51 Pearson JG. The present status and future potential of radio-

    therapy in the management of esophageal cancer. Cancer

    1977;39(suppl 2):882-890.

    52 Zargar SA, Khuroo MS, Jan GM et al. Prospective compar-

    ison of the value of brushings before and after biopsy in the

    endoscopic diagnosis of gastroesophageal malignancy. Acta

    Cytol 1991;35:549-552.

    53 Mori M, Adachi Y, Matsushima T et al. Lugol staining pat-

    tern and histology of esophageal lesions. Am J Gastroenterol

    1993;88:701-705.

    54 Kole AC, Plukker JT, Nieweg OE et al. Positron emission

    tomography for staging of oesophageal and gastroesophageal

    malignancy. Br J Cancer 1998;78:521-527.

    55 Griffith JF, Chan AC, Chow LT et al. Assessing chemother-

    apy response of squamous cell oesophageal carcinoma with

    spiral CT. Br J Radiol 1999;72:678-684.

    56 Rankin SC, Taylor H, Cook GJ et al. Computed tomography

    and positron emission tomography in the pre-operative stag-

    ing of oesophageal carcinoma. Clin Radiol 1998;53:659-665.

    57 Reed CE, Mishra G, Sahai AV et al. Esophageal cancer staging:

    improved accuracy by endoscopic ultrasound of celiac lymph

    nodes. Ann Thorac Surg 1999;67:319-321; discussion 322.

    58 Romagnuolo J, Scott J, Hawes RH et al. Helical CT versus

    EUS with fine needle aspiration for celiac nodal assessment

    in patients with esophageal cancer. Gastrointest Endosc

    2002;55:648-654.

    59 Rosch T. Endosonographic staging of esophageal cancer: areview of literature results. Gastrointest Endosc Clin N Am

    1995;5:537-547.

    60 Kelly S, Harris KM, Berry E et al. A systematic review of

    the staging performance of endoscopic ultrasound in gastro-

    oesophageal carcinoma. Gut 2001;49:534-539.

    61 Heidemann J, Schilling MK, Schmassmann A et al. Accuracy

    of endoscopic ultrasonography in preoperative staging of

    esophageal carcinoma. Dig Surg 2000;17:219-224.

    62 Lightdale CJ. Esophageal cancer. American College of

    Gastroenterology. Am J Gastroenterol 1999;94:20-29.

    63 Eloubeidi MA, Wallace MB, Hoffman BJ et al. Predictors of

    survival for esophageal cancer patients with and withoutceliac axis lymphadenopathy: impact of staging endosonogra-

    phy. Ann Thorac Surg 2001;72:212-219; discussion 219-220.

    64 Catalano MF, Alcocer E, Chak A et al. Evaluation of metasta-

    tic celiac axis lymph nodes in patients with esophageal carci-

    noma: accuracy of EUS. Gastrointest Endosc 1999;50:352-356.

    65 Vickers J. Role of endoscopic ultrasound in the preoperative

    assessment of patients with oesophageal cancer. Ann R Coll

    Surg Engl 1998;80:233-239.

    66 Lightdale CJ, Botet JF, Kelsen DP et al. Diagnosis of recurrent

    upper gastrointestinal cancer at the surgical anastomosis by

    endoscopic ultrasound. Gastrointest Endosc 1989;35:407-412.

    67 Tio LT, Blank LE, Wijers OB et al. Staging and prognosis

    using endosonography in patients with inoperable esophageal

    carcinoma treated with combined intraluminal and external

    irradiation. Gastrointest Endos 1994:40:304-310.

    68 Zuccaro G Jr, Rice TW, Goldblum J et al. Endoscopic ultra-

    sound cannot determine suitability for esophagectomy after

    aggressive chemoradiotherapy for esophageal cancer. AmJ Gastroenterol 1999;94:906-912.

    69 Laterza E, de Manzoni G, Guglielmi A et al. Endoscopic

    ultrasonography in the staging of esophageal carcinoma after

    preoperative radiotherapy and chemotherapy. Ann Thorac

    Surg 1999;67:1466-1469.

    70 Beseth BD, Bedford R, Isacoff WH et al. Endoscopic ultra-

    sound does not accurately assess pathologic stage of esophageal

    cancer after neoadjuvant chemoradiotherapy. Am Surg

    2000;66:827-831.

    71 Choi JY, Lee KH, Shim YM et al. Improved detection of

    individual nodal involvement in squamous cell carcinoma of

    the esophagus by FDG PET. J Nucl Med 2000;41:808-815.

    72 Kato H, Kuwano H, Nakajima M et al. Comparison between

    positron emission tomography and computed tomography in

    the use of the assessment of esophageal carcinoma. Cancer

    2002;94:921-928.

    73 Flamen P, Lerut A, Van Cutsem E et al. Utility of positron

    emission tomography for the staging of patients with poten-

    tially operable esophageal carcinoma. J Clin Oncol

    2000;18:3202-3210.

    74 Lerut T, Flamen P, Ectors N et al. Histopathologic validation of

    lymph node staging with FDG-PET scan in cancer of the esoph-

    agus and gastroesophageal junction: a prospective study based

    on primary surgery with extensive lymphadenectomy. AnnSurg 2000;232:743-752.

    75 Meltzer CC, Luketich JD, Friedman D et al. Whole-body

    FDG positron emission tomographic imaging for staging

    esophageal cancer comparison with computed tomography.

    Clin Nucl Med 2000;25:882-887.

    76 Kim K, Park SJ, Kim BT et al. Evaluation of lymph node

    metastases in squamous cell carcinoma of the esophagus

    with positron emission tomography. Ann Thorac Surg

    2001;71:290-294.

    77 Brucher BL, Weber W, Bauer M et al. Neoadjuvant therapy of

    esophageal squamous cell carcinoma: response evaluation by

    positron emission tomography. Ann Surg 2001;233:300-309.

    78 Weber WA, Ott K, Becker K et al. Prediction of response to

    preoperative chemotherapy in adenocarcinomas of the esoph-

    agogastric junction by metabolic imaging. J Clin Oncol

    2001;19:3058-3065.

    79 Kato H, Kuwano H, Nakajima M et al. Usefulness of

    positron emission tomography for assessing the response of

    neoadjuvant chemoradiotherapy in patients with esophageal

    cancer. Am J Surg 2002;184:279-283.

    80 Stein HJ, Brucher BL, Sendler A et al. Esophageal cancer:

    patient evaluation and pre-treatment staging. Surg Oncol

    2001;10:103-111.

  • 8/11/2019 adenocarcinoma esophageal

    11/12

    Koshy, Esiashvilli, Landry et al. 146

    81 Luketich JD, Friedman DM, Weigel TL et al. Evaluation of

    distant metastases in esophageal cancer: 100 consecutive

    positron emission tomography scans. Ann Thorac Surg

    1999;68:1133-1136; discussion 1136-1137.

    82 Nguyen NT, Roberts PF, Follette DM et al. Evaluation of

    minimally invasive surgical staging for esophageal cancer.

    Am J Surg 2001;182:702-706.

    83 Visbal AL, Allen MS, Miller DL et al. Ivor Lewis esophago-

    gastrectomy for esophageal cancer. Ann Thorac Surg

    2001;71:1803-1808.

    84 Ellis FH Jr. Standard resection for cancer of the esophagus

    and cardia. Surg Oncol Clin N Am 1999;8:279-294.

    85 Chu KM, Law SY, Fok M et al. A prospective randomized

    comparison of transhiatal and transthoracic resection for lower-

    third esophageal carcinoma. Am J Surg 1997;174:320-324.

    86 Law SY, Fok M, Cheng SW et al. A comparison of outcome

    after resection for squamous cell carcinomas and adenocar-

    cinomas of the esophagus and cardia. Surg Gynecol Obstet1992;175:107-112.

    87 Barbier PA, Luder PJ, Schupfer G et al. Quality of life and

    patterns of recurrence following transhiatal esophagectomy

    for cancer: results of a prospective follow-up in 50 patients.

    World J Surg 1988;12:270-276.

    88 Orringer MB, Marshall B, Iannettoni MD. Transhiatal

    esophagectomy: clinical experience and refinements. Ann

    Surg 1999;230:392-400; discussion 400-403.

    89 Gelfand GA, Finley RJ, Nelems B et al. Transhiatal

    esophagectomy for carcinoma of the esophagus and cardia.

    Experience with 160 cases. Arch Surg 1992;127:1164-1167;

    discussion 1167-1168.

    90 Gertsch P, Vauthey JN, Lustenberger AA et al. Long-term

    results of transhiatal esophagectomy for esophageal carci-

    noma. A multivariate analysis of prognostic factors. Cancer

    1993;72:2312-2319.

    91 Vigneswaran WT, Trastek VF, Pairolero PC et al. Transhiatal

    esophagectomy for carcinoma of the esophagus. Ann Thorac

    Surg 1993;56:838-844; discussion 844-846.

    92 Goldminc M, Maddern G, Le Prise E et al. Oesophagectomy

    by a transhiatal approach or thoracotomy: a prospective

    randomized trial. Br J Surg 1993;80:367-370.

    93 Skinner DB. En bloc resection for neoplasms of the esophagusand cardia. J Thorac Cardiovasc Surg 1983;85:59-71.

    94 Altorki NK, Girardi L, Skinner DB. En bloc esophagectomy

    improves survival for stage III esophageal cancer. J Thorac

    Cardiovasc Surg 1997;114:948-955; discussion 955-956.

    95 Logan A. The surgical treatment of carcinoma of the esophagus

    and cardia. J Thorac Cardiovasc Surg 1963;46:150-161.

    96 Law S, Wong J. What is appropriate treatment for carcinoma

    of the thoracic esophagus? World J Surg 2001;25:189-195.

    97 Hermanek P. pTNM and residual tumor classifications: prob-

    lems of assessment and prognostic significance. World J Surg

    1995;19:184-190.

    98 Orringer MB, Marshall B, Stirling MC. Transhiatal

    esophagectomy for benign and malignant disease. J Thorac

    Cardiovasc Surg 1993;105:265-276; discussion 276-277.

    99 Hagen JA, DeMeester SR, Peters JH et al. Curative resection

    for esophageal adenocarcinoma: analysis of 100 en bloc

    esophagectomies. Ann Surg 2001;234:520-530; discussion

    530-531.

    100 Altorki N, Skinner D. Should en bloc esophagectomy be the

    standard of care for esophageal carcinoma? Ann Surg

    2001;234:581-587.

    101 Lerut T, De Leyn P, Coosemans W et al. Surgical strategies inesophageal carcinoma with emphasis on radical lymphadenec-

    tomy. Ann Surg 1992;216:583-590.

    102 Hagen JA, Peters JH, DeMeester TR. Superiority of extended

    en bloc esophagogastrectomy for carcinoma of the lower esoph-

    agus and cardia. J Thorac Cardiovasc Surg 1993;106:850-858;

    discussion 858-859.

    103 DeMeester TR, Zaninotto G, Johansson KE. Selective thera-

    peutic approach to cancer of the lower esophagus and cardia.

    J Thorac Cardiovasc Surg 1988;95:42-54.

    104 Akiyama H, Tsurumaru M, Udagawa H et al. Radical lymph

    node dissection for cancer of the thoracic esophagus. Ann

    Surg 1994;220:364-372; discussion 372-373.105 Kato H, Watanabe H, Tachimori Y et al. Evaluation of neck

    lymph node dissection for thoracic esophageal carcinoma.

    Ann Thorac Surg 1991;51:931-935.

    106 Nishihira T, Hirayama K, Mori S. A prospective randomized

    trial of extended cervical and superior mediastinal lym-

    phadenectomy for carcinoma of the thoracic esophagus. Am

    J Surg 1998;175:47-51.

    107 Lerut T, Coosemans W, De Leyn P et al. Reflections on

    three field lymphadenectomy in carcinoma of the esophagus

    and gastroesophageal junction. Hepatogastroenterology

    1999;46:717-725.

    108 Altorki NK, Skinner DB. Occult cervical nodal metastasis inesophageal cancer: preliminary results of three-field lym-

    phadenectomy. J Thorac Cardiovasc Surg 1997;113:540-544.

    109 Greenlee RT, Hill-Harmon MB, Murray T et al. Cancer

    statistics 2001. CA Cancer J Clin 2001;51:15-36.

  • 8/11/2019 adenocarcinoma esophageal

    12/12

    ionshttp://theoncologist.alphamedpress.org/content/9/2/137#otherarticles

    This article has been cited by 6 HighWire-hosted articles:

    http://theoncologist.alphamedpress.org/content/9/2/137#otherarticleshttp://theoncologist.alphamedpress.org/content/9/2/137#otherarticleshttp://theoncologist.alphamedpress.org/content/9/2/137#otherarticleshttp://theoncologist.alphamedpress.org/content/9/2/137#otherarticles