You are on page 1of 29

71  Cancer of the Esophagus

Geoffrey Y. Ku and David H. Ilson

S UMMARY OF K EY P OI N T S
Classification Diagnosis and Staging response are modest for most
• Adenocarcinomas and squamous • Symptoms and demographics will patients. The addition of targeted
cell carcinomas (SCCs) comprise strongly suggest the diagnosis. therapies incrementally improves
98% of esophageal tumors. • Endoscopy is the best screening outcomes. The evaluation of immune
• The Siewert classification is now examination, with diagnosis made checkpoint inhibitors reveals a
commonly used to categorize with endoscopy with biopsy and modest but notable benefit in the
gastroesophageal junction (GEJ) cytologic assessment of tumor. metastatic setting and is of high
tumors, although many studies, • Endoscopic ultrasonography (EUS) priority.
especially those for advanced should be used to assess T and N • Surgery is an accepted
disease, have enrolled patients with stage to guide optimal definitive single-modality therapy for patients
esophageal, GEJ, and gastric therapy. with early localized disease
cancers. • Computed tomography (CT) of the (T1–2N0M0) or for patients who may
Incidence chest and abdomen is useful in not tolerate combined modality
• In the United States, approximately screening for metastatic disease. therapy. The selection of surgical
17,290 patients will be diagnosed • Positron emission tomography (PET) approach depends on location and
with esophageal cancer in 2018, with scan is useful to detect additional experience, but no approach has
15,850 deaths. cases of metastatic disease before been demonstrated to lead to
• Historically and internationally, SCC costly and toxic definitive therapy. It superior cure rates.
is the most common histologic type; may be superior to endoscopic EUS • Combined chemoradiation leads to
however, a dramatic increase in the in detecting intraabdominal lymph prolonged median survival and
incidence of adenocarcinoma has nodes, but not periesophageal nodes long-term survival compared with
been documented in the United adjacent to the primary tumor. radiation alone when used as a
States, the United Kingdom, and • Additional studies include definitive nonoperative approach.
Western Europe. laparoscopy, thoracoscopy, bone This represents a potentially curative
scan, and CT of the brain when alternative to surgery for SCCs and
Pathogenesis indicated by clinical circumstances. is appropriate for unresectable
• Esophageal cancers arise as a result lesions of either histologic type.
of chronic irritation from a wide Treatment
• Treatment of premalignant dysplasia • There are now established data
variety of sources, including gastric showing that preoperative
contents in chronic reflux and known is guided by grade of histology.
Low-grade dysplasia should be chemoradiation followed by surgery
carcinogens. improves survival versus surgery
• A strong association between Barrett treated with endoscopic ablative
techniques or observation. alone for both locally advanced
esophagus and adenocarcinoma is (clinically staged T2–T4 or
seen, but a benefit for screening High-grade dysplasia is treated
with endoscopic ablation or node-positive) adenocarcinoma and
endoscopy for those at risk for or SCC. There is less certainty about
with known Barrett esophagus is esophagectomy, although close
follow-up may be appropriate for the value of preoperative
unknown because the overall risk of chemotherapy alone.
cancer-related mortality is low. selected patients.
• Selection of appropriate treatment • Randomized trials have not
Barrett esophagus arises confirmed a survival benefit with
from gastroesophageal reflux for carcinoma depends on tumor
stage and patient performance surgery added to potentially curative
disease (GERD) and other risk chemoradiation in SCC, but there is
factors, including obesity and status.
• Chemotherapy is now routinely used a local control benefit. Therefore
smoking. definitive chemoradiation for patients
• SCC is associated with smoking and in perioperative strategies and is the
mainstay of treatment for recurrent with SCC is an option, with surgery
with alcohol use, and the declining reserved for locally persistent or
incidence has paralleled the decline or metastatic disease, in which
response rates and duration of recurrent disease. There are limited
in smoking. data regarding this approach for

1174 Downloaded for Tabanschii Nicolae (tabanschiirusuhan@gmail.com) at Nicolae Testemiceanu State Medical and Pharmaceutical University from
ClinicalKey.com by Elsevier on November 10, 2022. For personal use only. No other uses without permission. Copyright ©2022. Elsevier Inc. All rights
reserved.
Cancer of the Esophagus  •  CHAPTER 71 1175

patients with adenocarcinomas, but approach has been demonstrated is less well studied in locally
it can be considered in patients who to lead to superior cure rates. advanced esophageal cancer, but
are borderline candidates for • T2–4Nany or TanyN+ Tumors trials in gastric cancer including
esophagectomy. ○ Combined chemoradiation leads to GEJ adenocarcinoma have
• Postoperative adjuvant prolonged median survival and demonstrated a benefit.
chemotherapy or chemoradiation is long-term survival compared with
less well studied in locally advanced radiation alone when used as a Metastatic or Recurrent Disease
esophageal cancer, but trials in definitive nonoperative approach. • Chemotherapy is the mainstay of
gastric cancer including GEJ This represents a potentially treatment for recurrent or metastatic
adenocarcinoma have demonstrated curative alternative to surgery for disease, where response rates and
a benefit. SCCs and is appropriate for duration of response are modest for
• Endoscopic palliative therapy unresectable lesions of either most patients. The standard doublet
includes laser or electrical histologic type. consists of a fluoropyrimidine and a
fulguration, mucosal resection, ○ There are also established data platinum drug. Adding docetaxel to
photodynamic therapy (PDT), or that preoperative chemoradiation this doublet slightly improves
stenting. Excepting very superficial followed by surgery improves outcomes, but at the expense of
lesions, these therapies are not survival compared with surgery significant additional toxicity.
alternatives to surgery because they alone for both locally advanced • The addition of trastuzumab to
do not address deeper disease or adenocarcinoma and SCC. There first-line chemotherapy for
lymphatic spread. is less certainty about the value of HER2-positive disease incrementally
• Radiation and/or chemotherapy preoperative chemotherapy alone. improves outcomes. Similarly,
may be used to palliate local ○ Randomized trials have not ramucirumab with or without
symptoms. confirmed a survival benefit with paclitaxel in the second-line setting
surgery added to potentially is a validated strategy.
Algorithm curative chemoradiation in SCC, • The evaluation of immune
• T1is or T1a Tumors but there is a local control checkpoint inhibitors against the
○ High-grade dysplasia or T1a benefit. Therefore definitive PD-1/PD-L1 axis reveals a modest
tumors are managed with chemoradiation for patients with but notable benefit in the metastatic
endoscopic ablation or SCC is an option, with surgery setting and is of high priority.
esophagectomy. reserved for locally persistent or • Endoscopic palliative therapy
• T1b–2N0 Tumors recurrent disease. There are includes laser or electrical
○ Surgery is an accepted limited data regarding this fulguration, mucosal resection, PDT,
single-modality therapy for approach for patients with or stenting. Excepting very
patients with early localized adenocarcinomas, but it can be superficial lesions, these therapies
disease (T1–2N0M0) or for patients considered in patients who are are not alternatives to surgery
who may not tolerate combined borderline candidates for because they do not address deeper
modality therapy. The selection of esophagectomy. disease or lymphatic spread.
surgical approach depends on ○ Postoperative adjuvant • Radiation and/or chemotherapy may
location and experience, but no chemotherapy or chemoradiation be used to palliate local symptoms.

Esophageal cancer is a devastating disease associated with poor HISTOLOGIC AND MOLECULAR
survival outcome and adverse effects on swallowing and quality of CLASSIFICATION AND LOCATION
life (QoL). Although it is an aggressive malignancy that usually
manifests in a locally advanced stage, significant progress has been Esophageal cancer is classified based on histologic appearance and cell
made in the treatment of this disease, including expanded treatment of origin, as follows: (1) epithelial tumors, (2) metastatic tumors, (3)
options, decreased surgical morbidity and mortality, confirmation lymphomas, and (4) sarcomas. Cancers of epithelial cell origin, pre-
of the benefit of combined modality therapy, and improvements in dominantly SCC and adenocarcinoma, are the most common. Although
identifying patients at risk. These advances are resulting in incremen- SCC and adenocarcinoma were previously treated as similar entities
tal but still quite meaningful improvements in outcome, but there and grouped together, there is increasing recognition that they should
remains considerable controversy over the optimal management in be studied as separate entities whose optimal therapy may diverge in
individual scenarios. The emphasis of this chapter is on selecting the the era of multimodality treatments.
appropriate options in the curative and palliative management of There is now clear-cut molecular evidence that SCCs and adeno-
esophageal cancer. carcinomas are entirely distinct entities. Data from The Cancer Genome
The revised American Joint Committee on Cancer (AJCC) seventh Atlas showed different molecular aberrations between them.1 Specifically,
edition staging system refines the categorization of patients. In par- SCCs showed frequent genomic amplifications of CCND1 and SOX2
ticular, there is now consideration of location within the esophagus, and/or TP63, whereas ERBB2, VEGFA. and GATA4 and GATA6 were
number of positive lymph nodes, and histologic findings and a changed more commonly amplified in adenocarcinomas. Equally notable is
categorization of extent of primary disease. Adenocarcinoma and the fact that esophageal and gastroesophageal junction (GEJ) adeno-
squamous cell carcinoma (SCC) histologic types are also considered carcinomas strongly resembled the chromosomally unstable variant
separately. Survival data based on this staging system are shown of gastric cancer (which is most commonly found in the cardia2),
in Fig. 71.1. suggesting that these tumors should be treated as a single entity.

Downloaded for Tabanschii Nicolae (tabanschiirusuhan@gmail.com) at Nicolae Testemiceanu State Medical and Pharmaceutical University from
ClinicalKey.com by Elsevier on November 10, 2022. For personal use only. No other uses without permission. Copyright ©2022. Elsevier Inc. All rights
reserved.
1176 Part III: Specific Malignancies

adenocarcinoma of the distal esophagus, GEJ, and gastric cardia has


Adenocarcinoma increased 4% to 10% per year among US men since 1976, so it now
100 comprises 75% of all esophageal tumors.5–7 The absolute incidence
in the United States has increased from 3.8 per million in 1973 to
Risk-adjusted survival (%)

80
1975 to 23.3 per million in 2001 based on the National Cancer
0 Institute’s Surveillance, Epidemiology, and End Results (SEER) database.
IA This rate of increase exceeds that of all other cancers, including lung,
60 breast, and prostate cancers and melanoma. Adenocarcinoma is much
IB
less common in African Americans but has increased from 0.4 per
40 IIA
100,000 to 0.9 per 100,000, and 0 to 0.2 per 100,000 in females.
IIB Even as it appears that the rate of increase may be slowing in the
20 IIIA United States,8 the incidence of esophageal adenocarcinoma globally
IIIB
IIIC continues to increase.9
Changing epidemiologic factors account for the increasing incidence
0 of adenocarcinomas, which are now more common because of an
0 2 4 6 8 10 increased incidence of gastroesophageal reflux disease (GERD)10
A Years and obesity.11 Helicobacter pylori, implicated in peptic ulcer disease
and associated with an increased risk of gastric cancer, has not been
Squamous Cell Carcinoma implicated in the pathogenesis of esophageal adenocarcinoma. In
fact, because infection with H. pylori may lead to a reduction in
100 gastric acidity in association with atrophic gastritis, there has been
speculation that a decline in the prevalence of H. pylori infection may
Risk-adjusted survival (%)

80 predispose to an increase in GERD and therefore in the incidence


of GEJ adenocarcinomas.12,13 Similarly, infection with human papil-
lomavirus, while implicated in oropharyngeal SCC, has not been
60 IA
0 observed in esophageal SCC.1,14
IB

40 IIA
IIB
PATHOGENESIS
20 IIIA Data support the hypothesis that esophageal cancer arises as a result
IIIB
IIIC of chronic irritation and inflammation of the esophagus, leading to
a sequence of genetic alterations in the damaged epithelium and
0
histologic changes of dysplasia to carcinoma. Factors that are associated
0 2 4 6 8 10 with an increased risk of SCC include caustic lye ingestion and radiation
B Years therapy, and the interval between injury and the development of
cancer may be considerable. In addition, a history of underlying
Figure 71.1  •  Survival according to American Joint Committee on Cancer/ esophageal disease such as achalasia, Plummer-Vinson syndrome, and
International Union Against Cancer seventh edition staging system for an tylosis is also linked to SCC. In adenocarcinoma, gastroesophageal
international cohort of 4627 patients treated surgically for esophageal neoplasms,
including adenocarcinoma patients (A) and squamous cell carcinoma patients reflux of acid and bile are major risk factors that cause chronic inflam-
(B). (From Rice TW, Rusch VW, Ishwaran H, et al. Worldwide Esophageal mation, which then leads to carcinogenesis.
Cancer Collaboration. Cancer of the esophagus and esophagogastric junction: There are important differences between the risk factors and
data-driven staging for the seventh edition of the American Joint Committee histologic progression that support the concept that SCC and adeno-
on Cancer/International Union Against Cancer cancer staging manuals. Cancer. carcinoma are separate entities with the potential to respond differently
2010;116[16]:3763–3773.) to newer treatment approaches.

SCC usually occurs in the middle third of the esophagus, whereas Clinical Risk Factors
adenocarcinomas are most common in the lower third of the esophagus.
The Siewert classification is now routinely used to further subtype Two major risk factors for esophageal SCC are alcohol use and
GEJ tumors based on their location.3 Siewert type I tumors arise from tobacco smoking, according to epidemiologic studies from various
the distal esophagus and infiltrate the GEJ from above, whereas type countries worldwide. This relationship is dose dependent, and there
III tumors are gastric cardia tumors that infiltrate the GEJ from below; is a multiplicative interaction with alcohol intake and tobacco use.
type II tumors are true tumors of the GEJ. In a prospective cohort study from the Netherlands involving more
than 120,000 people with 16 years of follow-up, the strongest risk
INCIDENCE factor for SCC was alcohol consumption with a 4.6-fold increased
risk, whereas combined exposure with smoking increased the risk more
Because SCCs and adenocarcinomas account for 98% of all esophageal than 8-fold. The risk of SCC decreased with smoking cessation and
tumors, this chapter focuses on these two histologic types. Epidemiologic alcohol abstinence, but only after one to two decades.15
data show that the incidence of esophageal cancer varies considerably In adenocarcinoma, smoking but not alcohol is associated with
from one country to another and often within a single country. This increased risk. A prospective study of tobacco, alcohol, and the risk
geographic diversity underscores the multifactorial etiologies of of esophageal cancer in the United States found an increased risk of
esophageal cancer worldwide, where more than 90% of tumors are SCC among current smokers compared with nonsmokers (hazard ratio
SCCs. The epidemiologic factors responsible for the geographic vari- [HR] 9.27; 95% confidence interval [CI], 4.04–21.29) and also an
ability in incidence of esophageal cancer, including potential dietary increased risk for adenocarcinoma (HR, 3.70; 95% CI, 2.20–6.22).
and environmental carcinogens, remain under active investigation. For people who consumed more than three alcoholic drinks a day
Although cases of SCC have steadily declined in the United States compared with one drink, there was increased risk of esophageal SCC
because of a decrease in tobacco and alcohol abuse,4 the incidence of but not adenocarcinoma.16

Downloaded for Tabanschii Nicolae (tabanschiirusuhan@gmail.com) at Nicolae Testemiceanu State Medical and Pharmaceutical University from
ClinicalKey.com by Elsevier on November 10, 2022. For personal use only. No other uses without permission. Copyright ©2022. Elsevier Inc. All rights
reserved.
Cancer of the Esophagus  •  CHAPTER 71 1177

Nutritional and dietary factors have been found to contribute to in situ carcinoma or dysplasia in the face of a large lesion seen on
the risk of both SCC and adenocarcinoma. Pickled vegetables, processed endoscopic or radiographic studies should not be accepted, and biopsy
meat, and other foods containing nitrates have been linked to an should be repeated to confirm the extent of invasion and guide optimal
increased risk of SCC. Drinking very hot liquids or eating hot foods management.
such as stewed meat frequently is postulated to cause thermal injury Once the pathologic diagnosis has been established, evaluation to
to the esophagus and has been found to be a risk factor for SCC. determine the extent of disease should include a computed tomog-
Low consumption of fruits and vegetables is a risk factor for both raphy (CT) scan of the chest and complete abdomen. The accuracy
SCC and adenocarcinoma.17 of abdominal CT for identification of metastases to the liver and
Obesity, defined as a body mass index of 30 or higher, is a strong celiac axis depends on the bulk of the disease. Small liver metastases,
risk factor for esophageal adenocarcinoma, as demonstrated in a meta- peritoneal studding, and abdominal nodes will often be undetectable.
analysis of epidemiologic studies (HR, 2.78; 95% CI, 1.85–4.16).11 For the patient with an adenocarcinoma of the distal esophagus, GEJ,
This has clear implications regarding the increasing obesity rates in or cardia, a complete abdominopelvic CT scan is recommended because
the United States and could partly explain the rising incidence of these tumors are more likely to metastasize early to periaortic lymph
esophageal adenocarcinoma. The exact mechanism for this association nodes. A complaint of back pain may signal the presence of enlarged
of obesity is not understood but might reflect an increased propensity retroperitoneal nodes.
for gastroesophageal reflux. Obesity does not appear to be a risk Positron emission tomography (PET) scanning enables the
factor for SCC. identification of metastatic disease in patients who might otherwise
inappropriately receive definitive local therapy and therefore is
Adenocarcinoma: Role of Gastroesophageal Reflux now considered a standard staging test. Studies demonstrated that
Disease and Barrett Esophagus PET will detect unsuspected metastatic disease in approximately
15% of patients after all other staging tests have been completed,
There is a clear relationship between GERD and the development of although it is not as useful as other techniques in identifying involved
Barrett esophagus. Esophageal adenocarcinoma frequently arises in regional nodes (based on uptake from the primary tumor obscuring
Barrett esophagus, a condition in which the normal stratified squamous peritumoral lymph nodes). A prospective study of 79 patients found
mucosa of the esophagus is replaced by a metaplastic columnar-lined that the specificity and sensitivity of PET for identifying stage IV
epithelium that extends upward from the GEJ, generally as a result disease were 90% and 74% versus 47% and 78%, respectively, for
of injury from chronic reflux. Barrett esophagus may progress to the combination of CT and endoscopic ultrasonography (EUS),
dysplasia and then malignancy as the dysplastic epithelial cells accu- with an overall accuracy of identification of stage IV disease of 82%
mulate genetic alterations. versus 64% (P = .004).23 Furthermore, when PET was added to
GERD is a well-established risk factor for esophageal adenocarci- CT and EUS, 22% of patients had a change in stage that altered
noma. A population-based case-control study in Sweden demonstrated their planned treatment (15% upstaged to incurable stage IV disease
an odds ratio (OR) of 7.7 (95% CI, 5.3–11.4) for the development and 7% downstaged to a stage at which curative therapy would be
of esophageal cancer in patients with chronic GERD. With long- appropriate). A consensus panel has confirmed the recommendation for
standing, severe symptoms, the OR for esophageal adenocarcinoma PET imaging in possibly localized esophageal cancer to better detect
was 43.5 (95% CI, 18.3–103.5).18 There was no association seen metastatic disease.24
between reflux and SCC. Accurate determination of the extent of disease has a major impact
The prevalence of Barrett esophagus is estimated to be 1% to 2% on therapeutic decision making for single-modality versus multimodal-
of the general population.19 The length of time for progression from ity treatment or curative versus palliative intent, and therefore it is
Barrett esophagus to dysplasia to adenocarcinoma is unknown, although essential that comprehensive staging be performed. A substantial body
several reports of endoscopic surveillance programs have suggested that of literature now also exists regarding EUS and laparoscopy. The
progression of Barrett esophagus to adenocarcinoma is uncommon. largest and earliest experience was with EUS.25,26 A pooled analysis
A nationwide population-based cohort study in Denmark followed of the literature indicates a sensitivity and specificity, respectively,
more than 11,000 patients with Barrett esophagus for a median of for stage T1 of 81.6% and 99.4%, T2 of 81.4% and 96.3%, T3 of
5.2 years and found that the annual risk of adenocarcinoma was 91.4% and 94.4%, and T4 of 92.4% and 97.4%.26 EUS has not
0.12% (95% CI, 0.09–0.15).20 Meanwhile, a meta-analysis of 57 been considered accurate in distinguishing in situ cancer from invasive
studies involving more than 11,000 patients with nondysplastic Barrett (T1) superficial lesions. For nodal staging based on morphologic
esophagus found a pooled annual incidence rate of adenocarcinoma features (size, shape, border, and echo characteristics), the observed
of 0.33%.21 Patients with low-grade dysplasia had an incidence sensitivity and specificity were 84.7% and 84.6%, respectively, which
rate of adenocarcinoma of 0.5% per year, whereas those with high- with the addition of fine-needle aspiration improved to 96.7% and
grade dysplasia has an incidence rate of adenocarcinoma of 3% to 95.5%. EUS is not a reliable technique for diagnosing liver and
5% per year. peritoneal metastases because of the limited depth of penetration of
ultrasound waves.27
DIAGNOSTIC AND STAGING EVALUATION The indications for minimally invasive surgical staging techniques
are not fully defined. Laparoscopic evaluation of abdominal lymph
The most common presentation of esophageal carcinoma is solid food nodes can be achieved with minimal risks and can allow for the
dysphagia and weight loss of several months’ duration. Other presenta- identification of node-positive disease not detected with cross-sectional
tions that occur with esophageal adenocarcinoma in particular include imaging and EUS,28–30 although whether the presence of locoregional
chest pain in the absence of myocardial ischemia and anemia from lymphadenopathy will affect the overall management strategy is unclear.
chronic gastrointestinal bleeding from the mucosal lesion. These clinical Unsuspected findings such as liver metastases or peritoneal studding that
signs and symptoms should prompt endoscopic evaluation and would clearly alter treatment occur in approximately 15% of patients.
diagnostic imaging. The diagnosis is usually evident by the characteristic However, there is not a consensus regarding the use of laparoscopy
narrowing of the esophagus on barium studies, but endoscopy and as part of baseline staging. Whereas the Society of Thoracic Surgeons
biopsy are essential for histopathologic diagnosis. Endoscopic biopsies recommends staging laparoscopy for locally advanced (T3 and T4)
and brushings of the lesion will yield the diagnosis in more than 90% GEJ adenocarcinoma that infiltrates the gastric cardia, National
of patients. Multiple biopsies may be necessary for diagnosis of an Comprehensive Cancer Network guidelines consider laparoscopy an
invasive malignancy that is submucosal or necrotic.22 A diagnosis of optional part of the staging process in patients with GEJ tumors.31

Downloaded for Tabanschii Nicolae (tabanschiirusuhan@gmail.com) at Nicolae Testemiceanu State Medical and Pharmaceutical University from
ClinicalKey.com by Elsevier on November 10, 2022. For personal use only. No other uses without permission. Copyright ©2022. Elsevier Inc. All rights
reserved.
1178 Part III: Specific Malignancies

CHOICE OF THERAPEUTIC OPTIONS: Given the modest activity of single agents in esophageal cancer,
BARRETT ESOPHAGUS AND DYSPLASIA combination chemotherapy of two and even three drugs has been
extensively studied. A Cochrane meta-analysis of 13 trials including
Because of the premalignant nature of Barrett esophagus, it has 1914 patients (which mostly evaluated older chemotherapy regimens)
been recommended that patients with this condition undergo did show modest improvements in RRs (35% versus 18%; OR, 2.91;
regular endoscopic surveillance, primarily to assess for dysplasia. 95% CI, 2.15–3.93), median time to progression (TTP; 5.6 versus
As mentioned in a previous section, it is estimated that the risk of 3.6 months; HR, 0.67; 95% CI, 0.49–0.93), and median OS (8.3
progression from Barrett esophagus to adenocarcinoma is 0.12% versus 6.8 months; HR, 0.82; 95% CI, 0.74–0.90) with combination
to 0.33% annually.20,21 This relatively small risk calls into question over single-agent chemotherapy.54
the value of routine endoscopic surveillance for Barrett metaplasia, Despite incremental advances, the duration of response to both
especially in the absence of dysplasia or specialized intestinal metaplasia, modern single agents and combination regimens is only 4 to 6 months,
given the significant cost and small risk of injury from the follow-up in general, with a median OS of 10 to 12 months.
endoscopies.
The focus in the future may be on better defining the risk of Combination Chemotherapy
progression for individual patients through use of biomarkers and
genetic and epigenetic abnormalities. For example, a retrospective Fluoropyrimidine/Platinum Doublet
assessment of a large multicenter database of patients attempted to The combination of infusional 5-FU–cisplatin has been studied
develop a model of risk assessment based on age and methylation of extensively since the 1980s, and the doublet of a fluoropyrimidine
eight genes previously observed to be associated with progression of with a platinum compound remains a reference regimen in many
Barrett esophagus: p16, HPP1, RUNX3, CDH13, TAC1, NELL1, contemporary trials. Of note, contemporary studies have generally
AKAP12, and SST.32 A model was developed with a sensitivity of 90% enrolled patients with adenocarcinomatous histology, irrespec-
and specificity of 50%, which could create a low-risk group with a tive of whether the tumor is found in the lower esophagus, GEJ,
1.7% rate of progression to high-grade dysplasia or adenocarcinoma or stomach.
over 5 years, a high-risk group with a 27% risk, and intermediate-risk More contemporary trials have evaluated substitutions of both of
groups of patients who would also be good candidates for close these drugs with either an oral 5-FU prodrug (capecitabine55 or S-156)
follow-up. Any study like this requires prospective validation. and/or the newer platinum compound oxaliplatin.55,57 Regimens such
The most recent American Gastroenterological Association medical as S-1–cisplatin,56 capecitabine-cisplatin,58 infusional 5-FU–oxaliplatin,57
position statement on the topic recommends follow-up endoscopy and capecitabine-oxaliplatin (along with the anthracycline epirubicin)55
every 3 to 5 years for patients with Barrett esophagus without dysplasia. appear to have at least comparable efficacy compared with 5-FU–
If low-grade dysplasia is identified, both endoscopic ablative therapy cisplatin and are also mostly associated with decreased toxicity and
and endoscopic surveillance every 12 months are options. Endoscopic increased ease of administration. In fact, an individual patient data
ablation should be offered for patients with high-grade dysplasia. meta-analysis of two randomized trials that compared capecitabine-based
Validated options include endomucosal resection (EMR), radiofrequency with infusional 5-FU–based regimens—the capecitabine-cisplatin versus
ablation, or photodynamic therapy (PDT). The relative benefits and 5-FU–cisplatin trial58 and the REAL-2 study,55 mentioned earlier and
disadvantages of these approaches in this context are beyond the scope discussed in more detail later—suggested that capecitabine-based
of this review. treatments are associated with superior RR and OS than infusional
Esophagectomy is the only treatment option that allows a patient 5-FU regimens.59
to safely stop periodic endoscopic biopsy surveillance. However, other The other platinum analogue—carboplatin—has been associated
alternatives may be optimal for many patients. A meta-analysis including with low single-agent activity in esophagogastric (EG) adenocarcino-
1874 patients who underwent esophagectomy for high-grade dysplasia mas.60 However, phase II trials of carboplatin combined with taxanes
suggested a risk of unsuspected lymph node involvement of only 1% have indicated promising activity,61,62 and the combination of
to 2%, further suggesting that more localized therapies are indeed carboplatin-paclitaxel with concurrent radiation has emerged as an
appropriate.33 However, the patients who choose endoscopic ablative international standard in locally advanced disease.63
therapy must be willing to undergo endoscopic biopsy surveillance One of the only studies that has shown a benefit for a fluoropy-
indefinitely. rimidine doublet compared with fluoropyrimidine alone, although
the former is routinely used, is the phase III SPIRITS (S-1 Plus
Cisplatin Versus S-1 in RCT in the Treatment for Stomach Cancer)
CHEMOTHERAPY study. S-1 is a mixture of tegafur (an oral 5-FU prodrug), gimeracil
Single-Agent Chemotherapy (a dihydropyrimidine dehydrogenate inhibitor that may potentiate
the effect of 5-FU), and oteracil (which may reduce the gastrointestinal
Up to one-half of patients with esophageal cancer have metastatic toxicity of 5-FU). In this Japanese trial in which 298 patients with
disease at presentation, and, despite multimodality therapy in the advanced gastric cancer were randomized to receive S-1 with or without
localized setting, the majority of patients will eventually develop cisplatin, the doublet was associated with a higher RR (54% versus
recurrence. Therefore most patients with esophageal cancer will receive 31%; P = .002) and superior OS (13.0 versus 11.0 months; P = .04).64
chemotherapy at some point during their disease course. Although this study helped to establish S-1–cisplatin as a standard
Until the early 1990s, chemotherapy drugs commonly used to treat therapy in Japan, a trial performed in US and European patients
esophagogastric cancers (EGCs) included 5-fluorouracil (5-FU),34,35 comparing S-1–cisplatin versus 5-FU–cisplatin failed to demonstrate
cisplatin,36–38 and mitomycin,39–41 with single-agent response rates superiority of S-1 over infusional 5-FU.56 Accordingly, S-1 is not
(RRs) ranging from 10% to 25%. Newer drugs with single-agent widely used outside of East Asia.
activity that have since been evaluated include the oral 5-FU pro-
drugs (capecitabine42,43 and S-144–46), the taxanes (paclitaxel47–49 and Moving Beyond 5-Fluorouracil–Cisplatin
docetaxel50–52), and irinotecan,53 with RRs of 15% to 45%. Despite the widespread use in the 1990s of high-dose cisplatin (100 mg/
A Cochrane meta-analysis by Wagner and colleagues that combined m2) with a 4- or 5-day infusion of 5-FU (at 1000 mg/m2/day) as a
three trials of chemotherapy versus best supportive care (BSC) revealed standard therapy, two seminal trials in the late 1990s directed the
a clear survival benefit for treatment (HR, 0.37; 95% CI, 0.24–0.55).54 development of chemotherapy regimens in EGC away from this
This translates into an improvement in median overall survival (OS) schedule of therapy. A European trial compared 5-FU–cisplatin
from 4.3 to 11 months with chemotherapy. with the FAMTX (bolus 5-FU–doxorubicin-methotrexate) and ELF

Downloaded for Tabanschii Nicolae (tabanschiirusuhan@gmail.com) at Nicolae Testemiceanu State Medical and Pharmaceutical University from
ClinicalKey.com by Elsevier on November 10, 2022. For personal use only. No other uses without permission. Copyright ©2022. Elsevier Inc. All rights
reserved.
Cancer of the Esophagus  •  CHAPTER 71 1179

(etoposide–leucovorin–5-FU) regimens.65 All regimens performed and median OS was 11.6 versus 11.8 months, respectively. Overall,
poorly, with high rates of toxicity, RRs of 20% or lower, and median cetuximab-FOLFOX appeared to be the least toxic of the three regimens.
OS of only approximately 7 months. These disappointing results Of course, the randomized phase II nature of this study was not
led investigators in continental Europe to pursue better-tolerated designed to detect a survival difference between these regimens, and
colorectal cancer–like schedules of biweekly infusional 5-FU with a the contribution of cetuximab, now thought to be either neutral or
platinum compound57 and to investigate taxane- and irinotecan-based even detrimental, cannot be determined. Nevertheless, the results of
chemotherapy. this trial do support the contention that any benefit of an anthra-
The second key trial was performed in the United Kingdom and cycline, if there is any benefit at all, is likely to be small. Another
compared FAMTX and the ECF regimen, which combines epirubicin notable finding of this study is that cetuximab-cisplatin-irinotecan
with a lower dose of cisplatin (60 mg/m2) and a 21-day infusion of had the lowest RRs and survival outcomes and the highest toxicity
low dose 5-FU (200 mg/m2/day).66 The ECF arm achieved superior rates, which has contributed to a significant decline in the use of
RRs (45% versus 21%; P = .0002), median OS (8.9 versus 5.7 months; this chemotherapy regimen in the first-line setting. Correspondingly,
P = .0009), and QoL at 24 weeks compared with FAMTX. This trial the favorable toxicity profile and activity of the FOLFOX arm have
established ECF as the reference regimen in the United Kingdom. reinforced its established role as the standard first-line regimen in the
United States.
Anthracyclines
After the validation of the ECF regimen, a subsequent study compared Taxanes
it with the MCF regimen, in which mitomycin was substituted for In comparison to the unclear benefit of adding an anthracycline to a
epirubicin (and the infusional 5-FU was administered at a higher dose fluoropyrimidine-platinum doublet, there are randomized data to
of 300 mg/m2/day).67 Although designed to test the superiority of support the addition of a taxane. The phase III V325 randomized
MCF, the study reported no significant differences in RR or median trial in GEJ and gastric adenocarcinomas compared the DCF regimen
survival but found that QoL was better maintained with ECF. Accord- (docetaxel–cisplatin–infusional 5-FU) and infusional 5-FU–cisplatin.69
ingly, ECF remained the preferred regimen, even as some investigators The addition of docetaxel improved RRs (37% versus 25%; P = .01)
questioned whether either of these triplet regimens is actually superior and TTP (5.6 versus 3.7 months; P < .001), but OS was only slightly
to the 5-FU–cisplatin doublet. improved (median OS 9.2 versus 8.6 months; 2-year OS, 18% versus
Subsequently, the REAL-2 (Randomized ECF for Advanced and 9%; P = .02). In addition, the three-drug regimen was associated with
Locally Advanced Esophagogastric Cancer 2) study compared the significantly more toxicity, including a grade 3/4 neutropenia rate of
ECF regimen and the ECX regimen (which involves the substitution 82% (versus 57%) and febrile neutropenia in 29% of patients (versus
of 5-FU with capecitabine), the EOF regimen (substitution of 12%). Fifty percent of patients came off treatment because of either
oxaliplatin for cisplatin) and the EOX regimen (a double substitution severe adverse events or consent withdrawal. Despite these significant
of both capecitabine and oxaliplatin) in patients with advanced EG toxicities, the authors reported a slower decrement in QoL measure-
adenocarcinomas or SCCs.55 All the combinations had similar RRs ments in the DCF arm.70 On the basis of this study, docetaxel was
(40%–48%) and toxicities, and the EOX regimen was associated with approved by the US Food and Drug Administration (FDA) in 2006
improved median OS compared with the ECF regimen (11.2 versus for use with 5-FU–cisplatin in this context.
9.9 months; P = .02), leading the authors to propose that the EOX Because the toxicities seen with the parent DCF regimen are
regimen could replace ECF in future trials. significant and may outweigh its small survival advantage over
Despite the standard use of ECF or one of its derivates in the 5-FU–cisplatin, it has not been widely adopted. It is also unclear if
U.K., the clear superiority of this triplet over a fluoropyrimidine- similar survival benefits would be accrued by the sequential use of
platinum doublet has never been demonstrated in a randomized fashion. first-line 5-FU–cisplatin followed by subsequent docetaxel (or paclitaxel)
One piece of evidence frequently cited to support the incorporation at progression.
of an anthracycline comes from the previously cited Cochrane meta- Several investigators have attempted to modify the regimen to
analysis, which analyzed three individually negative trials (including increase tolerability. For example, our group performed a randomized
the negative evaluation of ECF versus MCF described earlier).54 phase II trial of parent DCF (with prophylactic growth factor support)
Combining all three trials revealed a survival benefit for the addition versus a modified DCF (mDCF) regimen (consisting of reduced doses
of epirubicin (HR, 0.77; 95% CI, 0.62–0.91), which translates into of docetaxel and cisplatin administered with bolus and 2-day infusional
an approximate 2-month survival advantage. However, this conclusion 5-FU and leucovorin every 14 days).71 mDCF was associated with
comes largely from the comparison of ECF versus MCF because that decreased toxicity compared with parent DCF (neutropenic fever rate,
trial contributed two-thirds of the patients to the meta-analysis. Given 9% versus 16%; grade 3/4 nausea/vomiting rate, 2% versus 23%),
the greater toxicity noted on the MCF arm and the fact that the and median OS appeared superior in the mDCF arm (18.8 versus
comparison is not purely between a 5-FU–cisplatin-only arm at identical 12.6 months; P = .007). Nevertheless, 22% of the patients receiving
doses and ECF, a determination of the relative merits of adding mDCF (who had a median age of 59 years) required hospitalization
epirubicin remains difficult to make. in the first 3 months, mostly for treatment-related toxicities (febrile
Continuing questions regarding the benefit of an anthracycline neutropenia and gastrointestinal toxicities), reinforcing the notion
were raised by the results of a randomized phase II trial performed that this remains a relatively difficult regimen to administer and is an
by the US Cancer and Leukemia Group B (CALGB) and Eastern option only for healthy, motivated patients with frequent access to
Cooperative Oncology Group (ECOG). The CALGB 80403/ECOG medical evaluation.
1206 trial randomized 245 patients (222 with adenocarcinomas) to one Another extensively evaluated and commonly used modification
of three chemotherapy regimens—ECF, FOLFOX (biweekly bolus and is the German FLOT regimen, which consists of the substitution of
infusional 5-FU–leucovorin–oxaliplatin), or cisplatin-irinotecan—along oxaliplatin for cisplatin and is built around a 1-day infusion of 5-FU
with cetuximab, a monoclonal antibody against the epidermal growth every 14 days. Encouraging activity was noted in a phase II study of
factor receptor (EGFR), for patients with advanced esophagogastric 59 patients, which reported an RR of 58% and a median OS of 11.1
adenocarcinomas or SCCs.68 In the patients with adenocarcinomas, months.72 The FLOT regimen was subsequently compared with the
both the ECF and FOLFOX regimens plus cetuximab produced RRs 5-FU–oxaliplatin doublet (FLO) in the randomized phase II FLOT65+
of greater than 40% (61% and 54% respectively), which met the study, which enrolled patients who were 65 years of age or older and
primary objective of the trial. However, survival outcomes were very had either locally advanced or metastatic disease.73 No benefit was
similar between the cetuximab-ECF and cetuximab-FOLFOX arms. seen for the triplet combination in patients aged 65 years or older
Median progression-free survival (PFS) was 7.1 versus 6.8 months, with metastatic disease or in any patient aged 70 years or older. This

Downloaded for Tabanschii Nicolae (tabanschiirusuhan@gmail.com) at Nicolae Testemiceanu State Medical and Pharmaceutical University from
ClinicalKey.com by Elsevier on November 10, 2022. For personal use only. No other uses without permission. Copyright ©2022. Elsevier Inc. All rights
reserved.
1180 Part III: Specific Malignancies

study again emphasizes the need for strict patient selection for such two or fewer prior regimens were randomized in a 2:1 ratio to further
three-drug regimens. treatment with either docetaxel or irinotecan versus BSC.84 The patients
who received chemotherapy had a superior OS (5.3 versus 3.8 months;
Irinotecan HR, 0.66; P = .007), and therapy was well tolerated, with manageable
Irinotecan is another active agent in EGC that has been combined hematologic toxicities and comparable rates of nonhematologic toxicities
with mitomycin, 5-FU–leucovorin, and cisplatin with or without in both groups. There were no significant differences between the
docetaxel in phase II evaluations, with RRs ranging from 30% to chemotherapy arms.
65%.74–81 Toxicities in some of these trials (e.g., with cisplatin-docetaxel- The West Japan Oncology Group (WJOG) 4007 study then
irinotecan) have been substantial. randomized 223 patients to second-line paclitaxel versus irinotecan.85
A randomized phase II trial compared the FUFIRI regimen (weekly Median OS was comparable (9.5 versus 8.4 months; P = .38), although
infusional 5-FU–leucovorin–irinotecan) with cisplatin-irinotecan in there appeared to be less toxicity in the paclitaxel arm and more of
patients with advanced GEJ and gastric adenocarcinomas.76 FUFIRI those patients went on to receive third-line chemotherapy than in the
was associated with superior outcomes and less neutropenia than irinotecan-arm. Finally, the UK COUGAR-2 study confirmed the
cisplatin-irinotecan. This led to a subsequent phase III trial of FUFIRI benefit for second-line docetaxel versus BSC in a Western population,
versus 5-FU–cisplatin. Both regimens had comparable efficacy, but with a median OS of 5.2 versus 3.6 months (P = .01).86
there was less neutropenic fever and grade 3/4 stomatitis and nausea Based on these results, patients with good performance statuses
in the FUFIRI arm.82 Only the incidence of grade 3/4 diarrhea was should be offered additional non–cross-resistant treatment beyond
increased in the FUFIRI arm, although more patients withdrew from progression on first-line chemotherapy. As noted later, the combination
the 5-FU–cisplatin arm than the FUFIRI arm (22% versus 10%; P of paclitaxel with ramucirumab, an antibody against vascular endothelial
= .004) for drug-related adverse events. Although there was no clear growth factor receptor–2 (VEGFR2) is now considered the standard
benefit for FUFIRI over 5-FU–cisplatin, the favorable toxicity of this of care in the second-line setting.
combination supports its use as a front-line option, especially in patients
who are not candidates for a platinum compound. Response Rates in Adenocarcinoma and Squamous
The use of first-line 5-FU–irinotecan is now further supported by Cell Carcinoma
the results of a phase III French Fédération Francophone de Can-
cérologie Digestive (FFCD) group study, in which 416 patients were In general, it appears that adenocarcinoma and SCC tumors have
randomized to the FOLFIRI regimen (biweekly bolus and infusional overlapping RRs to combination chemotherapy, similar to the experience
5-FU–leucovorin–irinotecan) versus ECX in patients with advanced with non–small cell lung cancer. Few single agents have been tested
gastric cancer.83 At progression, patients received therapy with the in both cell types, and the number of patients treated in such studies
alternate regimen. This study revealed a superior time to treatment has been small. Phase III studies in SCC patients are also lacking.
failure for FOLFIRI versus ECX (5.1 versus 4.2 months; P = .008) Accordingly, regimens that are active in adenocarcinoma of the
and comparable PFS (5.3 versus 5.8 months; P = .96) and OS (9.5 esophagus, GEJ, and stomach are routinely extrapolated and used to
versus 9.7 months; P = .95). Toxicities were significantly less for treat patients with SCC tumors.
FOLFIRI (e.g., overall grade 3/4 toxicity rate of 69% versus 84%; P
< .001). Time to treatment failure was selected as the primary end TARGETED THERAPY
point because it captured discontinuation of a regimen for both efficacy
and toxicity reasons. Most investigators believe that the potential for making significant
Finally, Boku and colleagues also performed a phase III trial in progress lies in understanding and exploiting the molecular biology
which Japanese patients were randomized to infusional 5-FU versus of these tumors. The focus of recent study has shifted toward testing
cisplatin-irinotecan (a third arm was designed to evaluate and did newer agents that target specific molecular abnormalities known to
confirm noninferiority of S-1 versus infusional 5-FU).44 When compared occur in EGCs.
with infusional 5-FU, cisplatin-irinotecan was associated with an Gene expression profiling of 296 EGCs from a Western population
improved RR (38% versus 9%) but only a nonsignificant trend toward by Dulak and colleagues found amplified genes in 37% of samples,
improved median OS (12.3 versus 10.8 months; P = .055), at the including in several of the therapeutically targetable kinases that are
expense of significantly more grade 3/4 toxicities. As noted earlier, discussed here (specifically, EGFR and HER2), along with fibroblast
results of the CALGB 80403/ECOG 1206 study, which randomized growth factor receptor and K-ras.87 A similar analysis of 193 tumors
patients to cetuximab plus ECF versus FOLFOX versus cisplatin- (mostly from Singapore and East Asia) also identified amplifications
irinotecan, revealed that the cisplatin-irinotecan arm had the lowest of the same five targets.88 On the other hand, mutations in the K-ras
RR and shortest median OS.68 and B-raf oncogenes, which are common in some other solid tumors,
Taken together, these results have led many oncologists to move are rare in EGCs.89
away from using cisplatin-irinotecan as a first-line regimen for advanced Therefore the molecular targets of agents that are currently under
EGCs, although FOLFIRI has recently emerged as a viable first-line active clinical evaluation include HER2 and vascular endothelial growth
option. Some uncertainty about the superiority of irinotecan in the factor (VEGF). Therapies against EGFR were previously intensively
first-line setting was reinforced by the Cochrane meta-analysis of four investigated, but completed phase III studies showed either no benefit
clinical trials, which revealed a non–statistically significant trend toward or even detriment for its addition to standard treatments. Therapies
a small survival benefit for an irinotecan-containing regimen (HR, against HER2 and VEGF have focused exclusively on GEJ and gastric
0.86; 95% CI, 0.73–1.02).54 Notably, the FFCD study discussed adenocarcinoma patients, whereas the studies of anti-EGFR therapies
earlier was not included in this meta-analysis. have also enrolled esophageal SCC patients.

Second-Line Chemotherapy Anti-HER2 Therapy


Until recently, there were no large randomized studies to support a HER2 is a member of the ERBB tyrosine kinase receptor family.
survival benefit for second-line chemotherapy in EGCs. There are Ligand binding to these receptors leads to dimerization of a receptor
now three randomized studies that were performed in patients with either with itself or another member of the ERBB family. At least
gastric cancer to support such a benefit. nine such homodimers and heterodimers exist. In this network, HER2
The first study involved 202 Korean patients with an ECOG plays a major coordinating role because each receptor with a specific
Performance Status score of 1 or lower who had previously received ligand appears to prefer HER2 as its heterodimeric partner. This

Downloaded for Tabanschii Nicolae (tabanschiirusuhan@gmail.com) at Nicolae Testemiceanu State Medical and Pharmaceutical University from
ClinicalKey.com by Elsevier on November 10, 2022. For personal use only. No other uses without permission. Copyright ©2022. Elsevier Inc. All rights
reserved.
Cancer of the Esophagus  •  CHAPTER 71 1181

preference is further biased by overexpression of HER2, as seen in capecitabine-oxaliplatin chemotherapy as first-line treatment in 545
many types of human cancer cells.90 patients whose tumors were HER2 amplified with FISH testing.101
In EGCs, HER2 overexpression has been variably demonstrated Although RRs were higher in the lapatinib and chemotherapy arm
in esophageal SCC (mean, 23%; range, 0%–52%) and GEJ adeno- (53% versus 39%; P = .0031), there was not a statistically significant
carcinoma (mean, 22%; range, 0%–43%).91,92 The wide range of improvement in OS, the primary end point (12.2 versus 10.5 months;
expression reflects the differences in receptor testing based on P = .35). In a preplanned subset analysis, there was a benefit for
immunohistochemistry (IHC) or fluorescence in situ hybridization lapatinib in Asian and younger (<60 years old) patients. There was
(FISH), in addition to the varied cancer stages of patients. no correlation between HER2 positivity by IHC and benefit in this
The significance of HER2 expression as a prognostic or predictive HER2-amplified population.
marker is unclear. In esophageal SCC, HER2 overexpression has been Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate
correlated with extramural invasion and poor response to neoadjuvant of trastuzumab and DM1 (a microtubule inhibitor derived from
chemotherapy.92 Similarly, a meta-analysis of 14 studies involving maytansine) that has been approved by the FDA for use in HER2-
1464 patients with operable esophageal cancer also suggested inferior positive breast cancer that is refractory to trastuzumab and a taxane,
5-year survival for patients with HER2-positive tumors.93 However, based on the phase III EMILIA study.102 T-DM1 binds to HER2,
a more recent study from the Mayo Clinic (which was not part of releasing DM1 into the cytoplasm via receptor-mediated internalization
this meta-analysis) suggested that HER2 overexpression is actually and resulting in apoptosis. The randomized phase II/III GATSBY
associated with a lower tumor grade, fewer malignant lymph nodes, trial compared T-DM1 versus docetaxel-paclitaxel in the second-line
and the presence of Barrett esophagus.94 In patients with esophageal treatment of HER2-positive metastatic gastric cancer that progressed
adenocarcinoma and Barrett esophagus, HER2 expression correlated on trastuzumab-based therapy. Results have been presented only in
with improved disease-specific survival and OS. abstract form but reveal no improvement in OS versus chemotherapy
In gastric and GEJ adenocarcinoma, some studies have demonstrated (7.9 versus 8.6 months; P = .86).103
a correlation between HER2 amplification as determined with FISH Finally, pertuzumab is a humanized monoclonal antibody that
and increasing depth of invasion, lymph node and distant organ binds to the domain II dimerization arm of HER2, leading to inhibition
metastasis, and overall poor survival.95 The worse prognosis conveyed of HER2 dimerization with other ERBB family members and antibody
by HER2 positivity appears to be confirmed by a meta-analysis of 49 dependent cell-mediated cytotoxicity.104,105 Pertuzumab was initially
studies involving 11,337 patients with localized and metastatic tumors.96 FDA approved in combination with trastuzumab and docetaxel for
However, more recent studies not included in this meta-analysis previously untreated HER2-positive breast cancer on the basis of the
have suggested otherwise. A study by our group indicated that HER2 CLEOPATRA study.106
overexpression is more common in intestinal versus diffuse or mixed In the phase II JOSHUA study, 30 HER2-positive EG adenocar-
histologic types (33% versus 8%; P = .001) but that it is not an cinoma patients were randomized to two different doses of pertuzumab
independent prognostic factor in the metastatic setting.97 in combination with capecitabine-cisplatin and trastuzumab.107 Fifteen
In the locally advanced setting, large retrospective reviews also did patients received pertuzumab at a dose of 840 mg every 3 weeks for
not show HER2 to be prognostic or predictive of benefit either from six cycles, and the other 15 patients received 840 mg of pertuzumab
perioperative chemotherapy (in the UK MAGIC study with epirubicin– for cycle 1 followed by 420 mg every 3 weeks for the next five cycles.
cisplatin–5-FU)98 or from adjuvant chemotherapy after up-front surgery Both doses reached the predefined trough concentrations of pertuzumab
(in the Japanese ACTS-GC trial with the oral 5-FU prodrug S-1).99 on day 43. The disease-control rates (complete plus partial response
Part of the discrepancy of these results may arise because of dif- plus stable disease) at the end of six cycles were 82% and 100%,
ferences in IHC staining and scoring for HER2. The more contemporary respectively, in each arm. Toxicities appeared to be similar in both
studies cited here used IHC and FISH techniques similar to those arms, and diarrhea, mostly grade 1/2, was the most common toxicity.
used in the ToGA study (discussed later), which may make their The higher dose of 840 mg every 3 weeks was selected for the phase
conclusions more relevant. III JACOB study (NCT01774786), which has completed accrual and
randomized 780 patients to fluoropyrimidine-cisplatin and trastuzumab
Trastuzumab with or without pertuzumab. These results are eagerly awaited; this
Trastuzumab, a monoclonal antibody against HER2, was the first is the one remaining phase III study that has the potential to further
targeted therapy approved by the FDA for EGCs. In the pivotal ToGA improve treatment in HER2-positive EG adenocarcinoma.
(Trastuzumab for Gastric Cancer) trial, the addition of trastuzumab
to a fluoropyrimidine-cisplatin doublet for patients with GEJ and Anti–Vascular Endothelial Growth Factor Therapy
gastric adenocarcinomas whose tumors were HER2 positive with IHC
(3+) or FISH (HER2/CEP17 ratio ≥2) improved outcomes.100 RRs Therapies directed against VEGF have been the focus of major ongoing
(47% versus 35%; P = .0017), median PFS (6.7 versus 5.5 months; research in solid tumor malignancies. Folkman and others have provided
P = .0002), and OS (13.8 versus 11.1 months; P = .0046) were all compelling evidence linking tumor growth and metastases with
improved with the addition of trastuzumab. The greatest benefit seen angiogenesis.108
for the addition of trastuzumab was in high HER2 overexpressors Of the identified angiogenic factors, VEGF is the most potent and
with IHC 3+ or FISH-positive/IHC 2+ patients. Based on this dif- specific and has been identified as a crucial regulator of both normal
ferential benefit, trastuzumab is approved in the European Union and pathologic angiogenesis. VEGF produces a number of biologic
only for this subgroup of high HER2 overexpressors; in the United effects, including endothelial cell mitogenesis and migration and
States, it is approved for any patient who met the eligibility criteria induction of proteinases, leading to remodeling of the extracellular
for the ToGA study, including patients with FISH-positive status only. matrix, increased vascular permeability, and maintenance of survival
for newly formed blood vessels.109 VEGF exerts its angiogenic effects
Other Anti-HER2 Therapies by binding to several high-affinity transmembrane receptors, most
Building on the results of the ToGA study and mirroring strategies notably VEGFR types 1 (flt-1) and 2 (KDR, flk-1).110 In esophageal
that have proven to be beneficial in HER2-positive breast cancer, an cancer, VEGF is overexpressed in 30% to 60% of patients, with several
anti-HER2 tyrosine kinase inhibitor (TKI) and other antibody strategies studies demonstrating a correlation among high levels of VEGF
have been evaluated. Unfortunately and somewhat unexpectedly, the expression, advanced stage, and poor survival in patients undergoing
results that have been reported show no benefit for these drugs. esophagectomy.111–115 Studies in SCCs have indicated that expression
The first of these negative studies was the LOGiC study, a phase of VEGF in tumors correlates with more advanced tumor stage, the
III evaluation of lapatinib, an anti-HER2 TKI, in combination with presence of nodal and distant metastases, and a poorer survival

Downloaded for Tabanschii Nicolae (tabanschiirusuhan@gmail.com) at Nicolae Testemiceanu State Medical and Pharmaceutical University from
ClinicalKey.com by Elsevier on November 10, 2022. For personal use only. No other uses without permission. Copyright ©2022. Elsevier Inc. All rights
reserved.
1182 Part III: Specific Malignancies

outcome.113,116 In esophageal adenocarcinoma, increasing expression 6 months or longer, and fewer metastatic sites. The RRs of Japanese
of VEGF correlates with the transition from Barrett esophagus to patients who received ramucirumab-paclitaxel versus paclitaxel were
high-grade dysplasia and with the transition from microinvasive to 41% versus 19%, respectively (P = .0035), compared with 27% versus
locally advanced cancer.117,118 Similarly, increased VEGF expression 13%, respectively (P = .0004), for Western patients. Similarly, the
on tumors and increased serum VEGF levels have been correlated median PFS for Japanese patients who received ramucirumab-paclitaxel
with worse prognoses in gastric cancer.119,120 versus paclitaxel was 5.6 versus 2.8 months (HR, 0.50; P = .0002)
compared with 4.2 versus 2.8 months in Western patients (HR,
Bevacizumab 0.63; P < .0001). However, there was no OS benefit in the Japanese
The phase III AVAGAST trial evaluated the addition of bevacizumab, patients (11.4 versus 11.5 months; P = .51), whereas Western patients
a monoclonal antibody against VEGF-A, to capecitabine-cisplatin as did have improved OS (8.6 versus 5.9 months; P = .0050). This
first-line therapy for patients with advanced EG adenocarcinoma.121 may have been because of the increased use of postdiscontinuation
Although RRs and PFS were improved, the primary end point of an therapy in Japanese patients compared with Western patients (75%
OS improvement was not met, although there was a non–statistically versus 37%). When only Japanese patients who did not receive
significant trend toward benefit for the bevacizumab-containing arm additional therapy at progression were analyzed, those who received
(12.1 versus 10.1 months; P = .1002). In a planned subset analysis, ramucirumab did have superior OS (9.6 versus 4.3 months; HR,
there did appear to be more benefit for European and Pan-American 0.338; 95% CI, 0.124–0.922). Whether Japanese patients were able
patients than Asian patients. to receive more second-line and beyond therapy because of more
The contention that gastric cancer in East Asia is fundamentally favorable tumor biologic features that led to a slower decline in
different from that in the rest of the world was bolstered by a biomarker performance status, superior oncologic care, or some other factor is
analysis of the AVAGAST study.122 Baseline VEGF-A levels and baseline not known.
levels of neuropilin-1, transmembrane glycoproteins involved in Based on its activity in the second-line setting, a phase III study
angiogenesis and tumor growth, were found to be prognostic and evaluating first-line fluoropyrimidine-platinum with or without
predictive. In patients who received chemotherapy alone, higher ramucirumab has completed accrual (RAINFALL; ClinicalTrials.gov
VEGF-A level and lower neuropilin-1 expression were associated with NCT02314117).
shorter OS. In patients who received bevacizumab and chemotherapy,
this same pattern was associated with greater benefit from the addition Anti-Vascular Endothelial Growth Factor Receptor–2 Tyrosine
of bevacizumab. Noticeably, the relationship between baseline VEGF-A
levels and benefit from bevacizumab was seen only in non-Asian Kinase Inhibitors
patients. Additional evidence to suggest that bevacizumab may offer Apatinib, an anti-VEGFR2 TKI, was evaluated in a phase III study
greater benefits to a Western population comes from the two phase in 267 Chinese GEJ and gastric adenocarcinoma patients who had
II studies performed by our group that paved the way for the AVAGAST received two or more prior chemotherapy regimens.128 They were
study.123,124 In both of these studies, the addition of bevacizumab to randomized to receive either apatinib or placebo. Median OS was
different chemotherapy regimens improved survival outcomes signifi- significantly improved in the apatinib group (6.5 versus 4.7 months;
cantly, compared with historical controls. HR, 0.71; P = .0149), as was PFS (2.6 versus 1.8 months; HR,
0.444; P < .001). Responses in the apatinib group were rare (2.84%
Ramucirumab versus 0% for placebo-treated patients). Toxicities were consistent with
Momentum for the evaluation of anti-VEGF therapies in EGC was those of other anti-VEGFR TKIs and included hypertension (35.2%
halted after the AVAGAST trial until the results of the phase III all-grade, 4.5% grade 3/4), hand-foot syndrome (27.8% all-grade,
REGARD study were made available.125 Three hundred fifty-five 8.5% grade 3/4), and elevated liver transaminases (27.8% all-grade, 8%
patients with EG adenocarcinoma with prior progression on first-line grade 3/4). On the basis of this study, apatinib was approved in China
fluoropyrimidine and/or platinum-based chemotherapy were random- for this indication.
ized 2:1 to receive ramucirumab, an antibody against VEGFR2, versus Additional evidence that anti-VEGFR TKIs are active in non-Asian
placebo. The patients who received ramucirumab had improvement patients with EG adenocarcinoma comes from the randomized phase
in PFS (2.1 versus 1.3 months; HR, 0.48; P < .0001) and OS (5.2 II INTEGRATE study of regorafenib, another VEGFR2 TKI, in
versus 3.8 months, HR, 0.78; P = .047). Responses were uncommon which 152 GEJ and gastric adenocarcinoma patients from Australia,
(RR was 3% in both arms). The only toxicity more common in the New Zealand, South Korea, and Canada who had received two or
ramucirumab arm was grade 3 hypertension in 8% of patients. On fewer prior regimens were randomized in a 2:1 fashion to receive
the basis of this study, the FDA approved ramucirumab as second-line regorafenib versus placebo.129 The primary end point was PFS, which
monotherapy in April 2014. was improved in the regorafenib arm (2.6 versus 0.9 months; HR,
In addition, a subsequent phase III study of second-line paclitaxel 0.40; P < .001). There was a trend toward improved OS (5.8 versus
with or without ramucirumab (the RAINBOW study) showed benefit 4.5 months; HR, 0.74; P = .147) and an OS benefit may have been
for the combination.126 Median OS was superior for the ramucirumab- obscured by the fact that the study permitted crossover to regorafenib
paclitaxel arm (9.6 versus 7.4 months; P = .0169); RRs (28% versus at the time of progression. The grade 3/4 toxicity rate appears to have
16%; P = .0001) and PFS (4.4 versus 2.9 months; P < .0001) were been similar to that of apatinib. Interesting to note, the benefit for
also improved. When ramucirumab was combined with paclitaxel, regorafenib seemed to be greater in the South Korean than in other
the only other toxicity observed in addition to hypertension was a patients (HR, 0.12 versus 0.61; P < .001). On the basis of this study,
higher incidence of grade 3/4 neutropenia (41% versus 19%) but not a phase III study (INTEGRATEII, ClinicalTrials.gov NCT02773524)
a higher rate of neutropenic fever (3.1% versus 2.4%). The activity will shortly open.
and tolerability of this regimen led to its approval in November 2014 It is worth noting that the HR for PFS and OS are virtually
and has established it as the standard of care in the second-line setting. identical for apatinib and regorafenib (and very similar for single-agent
A subsequent subset analysis that compared 140 Japanese patients second-line ramucirumab), suggesting a robust antiangiogenic class
in this study with 398 patients from Western countries showed that effect for these drugs in EG adenocarcinoma. It is also important to
the Japanese patients appeared to derive more benefit in terms of note that none of the patients in these studies received prior ramu-
improvements in RR and PFS with the addition of ramucirumab.127 cirumab, so it is unclear if VEGFR TKIs will have activity in this
In general, these patients had more favorable characteristics than increasingly common group of patients. INTEGRATEII will permit
Western patients, including a higher proportion of patients with prior use of ramucirumab and will, it is hoped, definitively answer
ECOG Performance Status 0, a TTP on first-line chemotherapy of this question. For now, especially given its superior toxicity profile

Downloaded for Tabanschii Nicolae (tabanschiirusuhan@gmail.com) at Nicolae Testemiceanu State Medical and Pharmaceutical University from
ClinicalKey.com by Elsevier on November 10, 2022. For personal use only. No other uses without permission. Copyright ©2022. Elsevier Inc. All rights
reserved.
Cancer of the Esophagus  •  CHAPTER 71 1183

relative to the VEGFR TKIs, ramucirumab in the second-line setting increased in the cetuximab arm, including grade 3/4 diarrhea, rash,
remains standard. hand-foot syndrome, and hypomagnesemia.
Although all of these studies have focused on adenocarcinoma
Anti-Epidermal Growth Factor Receptor Therapy histologic type, a German randomized phase II study evaluated
5-FU–cisplatin with or without cetuximab in 62 patients with
EGFR or ERBB1 is another member of the ERBB transmembrane esophageal SCC.142 The RR rate was not improved with the addition
growth factor receptor family. The known ligands of the EGFR are of cetuximab (19% versus 13%; P = .79), and there was only a trend
epidermal growth factor and transforming growth factor-α. Binding toward improved time to treatment failure (3.4 versus 1.6 months; P
of a ligand to the EGFR causes it to dimerize either with itself or = .25) and OS (9.5 versus 5.5 months; P = .32). Despite these equivocal
with another member of the ERBB family. Dimerization then leads results, a phase III study in Germany in which patients were randomized
to activation of the TK and the downstream phosphorylation and to receive 5-FU–cisplatin with or without panitumumab was initiated.
activation of other effector signals.130 The study closed because of slow accrual, and results have yet to be
In EGCs, EGFR overexpression demonstrated by IHC or gene presented (ClinicalTrials.gov NCT01627379).
amplification by FISH occurs in 30% to 90% of tumors and correlates In the locally advanced setting, two studies have evaluated the
with increased invasion, a more advanced stage, more poorly differenti- addition of cetuximab to chemoradiation for locally advanced esophageal
ated histologic features, and a worse prognosis.131–134 and GEJ tumors. The UK SCOPE-1 study treated 258 patients with
The findings of four completed phase III trials of the anti-EGFR capecitabine-cisplatin and radiation with or without cetuximab.143
antibodies cetuximab and panitumumab have been published. Individu- Seventy-three percent of patients had SCC histologic type, and 60%
ally, each of these studies has been negative; collectively, they have had stage III disease. The original design of the study was to recruit
dampened enthusiasm for further evaluation of these drugs in an 420 patients. However, it was stopped after a preplanned analysis of
unselected population. the first 180 patients who had completed 24 weeks of follow-up
In the metastatic setting, the results of two studies have been occurred because it met predetermined criteria for futility. The primary
published. In the REAL3 study 553 patients with advanced EG end point of being free of treatment failure at 24 weeks was lower in
adenocarcinoma were randomized to EOC (epirubicin-oxaliplatin- the cetuximab arm (66.4%; 90% CI, 58.6–73.6) versus the standard
capecitabine) chemotherapy alone or with the addition of panitu- arm (76.9%; 90% CI, 69.7–83.0). The patients who received cetuximab
mumab.135 Unfortunately, the addition of panitumumab resulted in also had inferior OS (22.1 versus 25.4 months; HR, 1.45; P = .035).
a statistically significant inferior OS (11.3 versus 8.8 months; HR, Patients who received cetuximab were less likely to receive or complete
1.37; P = .013), which was the primary end point. The addition standard chemoradiation and also had higher rates of dermatologic
of panitumumab also resulted in a trend toward inferior PFS (6.0 and metabolic toxicities.
versus 7.4 months; HR, 1.22; P = .068) and did not improve RRs Finally and most recently, the results of the Radiation Therapy
(46% versus 42%; P = .42). Toxicities were also increased with the Oncology Group (RTOG) 0436 study were presented in abstract
addition of panitumumab and included higher rates of grade 3/4 form.144 This study evaluated cisplatin-paclitaxel and radiation with
diarrhea, mucositis, rash, and hypomagnesemia, all class effects of or without cetuximab in the nonoperative setting for locally advanced
anti-EGFR antibodies. esophageal adenocarcinomas and SCCs. There was no difference in
These disappointing results cannot be easily explained. One pos- outcomes between both arms, confirming a lack of benefit for cetuximab
sibility is that increased toxicities seen when panitumumab was initially combined with chemoradiation.
added to full-dose EOC chemotherapy led to dose reductions in
oxaliplatin (by 23%) and capecitabine (by 20%). These dose reductions Immunotherapy
in chemotherapy—coupled with a lack of biologic benefit or even
harm from the addition of panitumumab—could have resulted in the There has been growing excitement among oncologists and patients
inferior OS in the experimental arm. alike regarding the use of immunotherapy or, more specifically,
A second possibility is that the combination of anti-EGFR therapy immune checkpoint inhibitors. Since the landmark approval by
with the 5-FU prodrug capecitabine may be associated with an the FDA of the anti–cytotoxic T-lymphocyte antigen-4 (CTLA-4)
unsuspected and inexplicable negative interaction. We previously antibody ipilimumab in advanced melanoma,145,146 these and other
performed a meta-analysis of anti-EGFR antibodies in the first-line antibodies (namely, antagonists of the programmed death [PD]-1/
therapy of colorectal cancer and noted that the addition of these PD-ligand 1 [PD-L1] pathway) that derepress the immune system have
antibodies to regimens containing capecitabine or bolus 5-FU is undergone extensive evaluation in multiple other solid tumors, includ-
associated with reduced or absent benefit compared with infusional ing EGC. These studies have led to the FDA approval of additional
5-FU–based regimens.136 Although the biologic basis for such a proposed immune checkpoint inhibitors in several solid tumor malignancies
negative interaction is not known, there are emerging data that anti- and, in EGC, have culminated in one recently reported positive
EGFR therapy can reduce the expression of thymidylate synthase, phase III study.147
which is a main target for 5-FU.137–139 Such reduced expression may
mean that the choice of infusional 5-FU as opposed to capecitabine Immune Checkpoints
is important when anti-EGFR therapies are being added. The generation of an effective cellular immune response by T cells
Finally, the REAL3 investigators also published a companion against a cancer cell requires a primary signal and cosignals.148 The
manuscript of biomarker analysis focusing on genes previously identified primary signal comes from recognition of a cancer-associated antigen
to correlate with response or resistance. Consistent with other published by a T-cell receptor with very high specificity for the antigen, presented
data, the rates of mutations in K-ras, B-raf, and PIK3CA and loss of in the context of a class I/II major histocompatibility complex molecule,
PTEN expression were low (5.7%, 0%, 2.5%, and 15.0%, respectively) which is expressed on so-called antigen-presenting cells (APCs) such
and did not predict response to panitumumab.140 as dendritic cells or on the tumor cell itself. In addition to this primary
Similarly, EXPAND, a phase III trial of 904 patients with advanced signal, a secondary signal is required, in the absence of which the T
EG adenocarcinoma treated with capecitabine-cisplatin with or without cell may be rendered anergic or nonfunctional.
cetuximab, also failed to show a benefit in PFS, the primary end point It is now well established that the interactions between multiple
(4.4 versus 5.6 months; HR, 1.09; P = .32).141 RRs (30% versus 29%; molecules at the interface between the T cell and tumor cell create
P = .77) and OS (9.4 versus 10.7 months; P = .95) were also not multiple secondary signals, some of which are costimulatory and
improved. There was no difference in the primary outcome based on some of which are inhibitory. The ultimate activation or quiescence
EGFR expression demonstrated with IHC. Again, toxicities were of the immune response depends on the complex interplay and net

Downloaded for Tabanschii Nicolae (tabanschiirusuhan@gmail.com) at Nicolae Testemiceanu State Medical and Pharmaceutical University from
ClinicalKey.com by Elsevier on November 10, 2022. For personal use only. No other uses without permission. Copyright ©2022. Elsevier Inc. All rights
reserved.
1184 Part III: Specific Malignancies

signal—positive or negative—of these diverse interactions, which occur 11.2% (versus 0% for the placebo group) in this group, in which
at different times in the process of successful T-cell engagement and patients were enrolled irrespective of PD-L1 status. Based on this
activation. study, regulatory approval for the use of nivolumab in this setting is
Under normal physiologic conditions, immune checkpoints are pending in Japan.
crucial for the maintenance of self-tolerance (and the avoidance Overall, these results confirm the notable but modest activity of
of autoimmunity) and also for protection of tissues from excessive single-agent immune checkpoint inhibition in EGC. Ongoing and
damage if the immune system were to respond too exuberantly to an future efforts are focused on combinatorial strategies—with chemo-
infection. However, many of these molecules have become co-opted therapy, other immune checkpoint inhibitors, tumor vaccines, and
by cancer cells (in the process of achieving equilibrium and escape locoregional therapies such as radiation or chemoradiation—and will,
from the immune system). Their identification and targeting with it is hoped, transform the treatment of this disease in the next decade.
neutralizing (or agonist) antibodies now form the basis of modern
era immunotherapy. CHOICE OF THERAPEUTIC OPTIONS:
In 1995, James Allison and colleagues were one of two groups that
simultaneously characterized the function of CTLA-4, a protein that LOCALIZED ESOPHAGEAL CANCER
has high homology with CD28, which was already known to be a Surgery Alone
costimulatory molecule expressed on T cells necessary to provide the
secondary signal for T-cell activation.149 Just as CD28 does, CTLA-4 Esophagectomy with reconstruction has a clear goal of both achieving
also binds cognate ligands, the B7 molecules (which are found on local tumor control and restoring swallowing function. Esophageal
APCs), but with much higher affinity. However, unlike with CD28, surgical intraoperative risks, postoperative complications, and length
CTLA-4 expression is induced only when a T cell becomes activated. It of hospitalization have all decreased to acceptable levels that are now
then competes with CD28 for binding to the B7 molecules but leads comparable with other major oncologic resections.154–156 Whether
to downregulation and eventual abrogation of the immune response. surgical resection is performed as the sole therapy or as part of a
Subsequently, PD-1 was also identified as another negative immune combined approach, the surgical principles and techniques are the
checkpoint molecule.150 PD-1 has two ligands, PD-L1 and PD-L2. same, with the goal of achieving a gross total resection. Surgery as a
PD-L2 is mostly expressed on APCs, whereas PD-L1 is expressed on single modality is generally only appropriate for curable patients with
numerous tissues, including immune and tumor cells. In the tumor very early lesions or those who are medically unfit for combined
microenvironment, PD-L1 expressed on tumor cells binds to PD-1 modality therapy. Cancer cure rates do not appear related to the surgical
on activated T cells reaching the tumor. This delivers an inhibitory technique used.
signal to those T cells, preventing them from killing target cancer When surgery is used, an R0 resection—gross total resection with
cells and protecting the tumor from immune elimination.151 Unlike negative margins—is critical for long-term outcome, even with the
CTLA-4, which is thought to be necessary for T-cell activation, the availability of adjuvant or salvage chemoradiation. Results from a
PD-1/PD-L1/2 pathway is thought to protect cells from T-cell attack.152 data set of 1602 patients undergoing resection of distal esophageal
and GEJ adenocarcinoma demonstrated 5- and 10-year survival rates
Anti-PD-1 Antibodies for R0 resected patients of 43.2% and 32.7%, respectively, whereas
A full discussion of immune checkpoint inhibitors is beyond the scope the corresponding figures for patients with microscopic or macro-
of this chapter; the breathtaking pace at which studies are being scopic residual disease (R1 and R2 resection) were 11.1% and 6.2%
presented—and the existing data used to design the next generation (P < .0001).157
of studies—would also make this discussion rapidly outdated. A so-called palliative esophagectomy to restore swallowing function
However, two pivotal studies warrant discussion. At this time, the but with little or no change in long-term survival because of the extent
only study with results that have been published—and the first major of disease is of highly uncertain benefit relative to the risks and morbid-
study to ignite enthusiasm for this field in EGC—is the KEYNOTE-012 ity and any delay in administering systemic therapy and should be
study, a phase Ib dose-expansion study that evaluated 39 patients with contemplated only under very limited circumstances.
GEJ or gastric adenocarcinomas.153 All patients had tumors that were The standard operation in the United States includes resecting the
found to be PD-L1 positive with an experimental IHC assay that involved portion of esophagus, the proximal stomach, and the regional
used the Merck 22C3 antibody. Based on the cutoff for positivity of lymph nodes, as illustrated in Fig. 71.2. The surgical resection is
1% or greater membrane staining of tumor or peritumoral mono- therefore properly termed a partial esophagogastrectomy with regional
nuclear inflammatory cells, 40% of tumors were noted to be PD-L1 (or one field) lymphadenectomy. The resected esophagus is replaced
positive. with a conduit, usually the stomach or segments of the small or large
Nineteen patients were from Asia, and the rest were from the rest intestine, which are, in turn, mobilized as a vascularized pedicle and
of the world. Patients were heavily pretreated, and two-thirds had anastomosed to the remaining proximal esophagus.
received two or more prior therapies. The confirmed RR was 22% A number of incisional approaches can be successfully used to
for all patients; four of these eight patients had ongoing responses at perform a partial esophagogastrectomy, including the transhiatal, Ivor
the time of data analysis, and the median duration of response was Lewis, left thoracoabdominal, and three-incision techniques (Fig. 71.3).
40 weeks. Median PFS was 1.9 months, and median OS was 11.4 Less common techniques or modifications of the approaches are listed.
months; the 6- and 12-month OS rates were 66% and 42%, respectively. The specific incisional approach used generally determines how much
Toxicities appeared to be in line with the known side effects of esophagus is removed and where the esophageal anastomosis will be
pembrolizumab and included grade 3 or greater pneumonitis, pem- located (see Fig. 71.2). In the past, various surgeons have argued in
phigoid, peripheral neuropathy, and hypothyroidism in one patient support of their preferred techniques, giving the impression that all
(3%) each. of these approaches were uniquely different procedures. It is now
The benefit of anti-PD-1 therapy was confirmed by the presentation accepted that all of these incisional techniques use partial esophago-
of the results of a phase III study of nivolumab, another anti-PD-1 gastrectomy (except segmental esophagectomy with free jejunal grafting,
antibody, in 493 East Asian patients who had received two or more which is discussed separately), and outcomes are similar. Ultimately,
prior chemotherapy regimens.147 They were randomized in a 2:1 fashion long-term disease-specific survival after esophagectomy is related to
to nivolumab versus placebo. The study revealed a very modest pathologic tumor stage and is not dependent on the surgical esopha-
improvement in PFS (1.65 versus 1.45 months; HR, 0.60; P < .0001) gectomy technique.156,158,159 The data continue to demonstrate no
and OS (5.32 versus 4.14 months; HR, 0.63; P < .0001) but a difference in mortality or survival between transthoracic and transhiatal
landmark 12-month OS rate of 26.6% (versus 10.9%). The RR was esophagectomy (THE) approaches.160–162 The main variables when

Downloaded for Tabanschii Nicolae (tabanschiirusuhan@gmail.com) at Nicolae Testemiceanu State Medical and Pharmaceutical University from
ClinicalKey.com by Elsevier on November 10, 2022. For personal use only. No other uses without permission. Copyright ©2022. Elsevier Inc. All rights
reserved.
Cancer of the Esophagus  •  CHAPTER 71 1185

Transhiatal

Ivor-Lewis

Left thoracoabdominal
Transhiatal approach Ivor Lewis approach

Left thoracoabdominal
Three-incision approach approach

Figure 71.2  •  Regardless of the surgical approach used, a partial esopha- Figure 71.3  •  Different incisions are used to perform partial esophago-
gogastrectomy is performed to resect esophageal tumors. Depending on the gastrectomy. (From Reichle RI, Fishman EK, Nison MS, et al. Evaluation of
approach, different lengths of esophagus are removed. (From Heitmiller RF. the post-surgical esophagus after partial esophagogastrectomy for esophageal
Carcinoma of the esophagus. In: Bayless TM, ed. Current Therapy in Gastro- cancer. Invest Radiol. 1993;28:247.)
enterology and Liver Disease. 4th ed. St. Louis: Mosby; 1994;81.)
more extensive therapy for adequate curative management. This is
appropriate therapy for disease confined to the mucosa, where there
is a probability of lymph node involvement that approaches 0%. This
performing a partial esophagogastrectomy are which incision(s) to approach may also be appropriate for submucosa involvement up to
use, the length of esophagus to resect, what to use to replace the one-third of the thickness: lymph node involvement is observed in
esophagus, and which route through the chest this conduit will take. 45% (23 of 51) of cases with deep submucosal invasion versus 10%
(3 of 29) of middle-third and 7.5% (3 of 40) of inner-third cancers.163
CHOICE OF THERAPEUTIC OPTIONS: EARLY Therefore for lesions with invasion beyond the submucosa, standard
ESOPHAGEAL CANCER management including surgical resection is warranted, although
EMR may be appropriate under select circumstances with minimal
Early esophageal cancers (T1–2N0) may be managed with single- submucosal invasion.
modality therapy with the goal of curing the tumor while minimizing
morbidity of treatment. Endoscopic approaches should optimally be Esophagectomy for Stage I and IIa Tumors
considered only for tumors confirmed to invade no further than
one-third of the depth of the submucosa (Tis, Ia). When deeper Esophagectomy is the standard therapy. The outcome is favorable,
invasion has occurred, there is an otherwise high rate of undetected especially for T1–2N0 disease, and is demonstrated in Fig. 71.1. This
nodal involvement that would necessitate esophagectomy and result procedure has the advantage of pathologic confirmation of the extent
in a change to a combined modality approach. Single-modality therapy of disease, which offers prognostic information and which may inform
is also appropriate for the rare group of patients with locally advanced adjuvant therapy. For patients with T3N0 or TanyN+ tumors, combined
disease who are not medically appropriate candidates for combined modality therapy should be considered because the long-term survival
modality therapy. is less than 50% and clinical staging may frequently overlook involved
lymph nodes.
Nonsurgical Management of Early-Stage (Tis, Ia)
Esophageal Cancer CHOICE OF THERAPEUTIC OPTIONS:
EMR may be the optimal endoscopic minimally invasive approach LOCALLY ADVANCED ESOPHAGEAL CANCER
for early carcinoma. In contrast to PDT and radiofrequency ablation, Transhiatal Resection
this approach allows pathologic confirmation of depth of invasion.
This confirmation is critical because the greater the depth of invasion, THE is a frequently used approach, “rediscovered” in 1976 by Dr.
the higher is the associated risk of nodal involvement, which requires Mark Orringer, in which the intrathoracic esophagus is mobilized

Downloaded for Tabanschii Nicolae (tabanschiirusuhan@gmail.com) at Nicolae Testemiceanu State Medical and Pharmaceutical University from
ClinicalKey.com by Elsevier on November 10, 2022. For personal use only. No other uses without permission. Copyright ©2022. Elsevier Inc. All rights
reserved.
1186 Part III: Specific Malignancies

distally through the esophageal hiatus and proximally through a cervical Lewis in 1945.164 This procedure was designed to optimize exposure
incision. The increased prevalence of distal esophagus and GEJ adeno- of the intrathoracic esophagus, which passes through the upper two-
carcinoma has largely been responsible for the widespread popularity thirds of the chest along the right posterior mediastinum. Once the
amongst surgeons of the transhiatal approach. Because of their distal involved intrathoracic esophagus has been mobilized, a partial
esophageal location, these tumors are invariably near the GEJ and esophagogastrectomy is performed and the esophagus is replaced with
readily accessible for direct-vision dissection through the hiatus. stomach, colon, or (less frequently) jejunum, which is passed into the
Moreover, the regional lymph nodes for these distal tumors are in the chest along the esophageal bed and anastomosed to the proximal
parahiatal and proximal lesser curvature regions, both accessible via esophagus, usually at or above the level of the azygos arch. The
laparotomy. The resected esophagus is reconstructed by using the advantages of the technique are the excellent exposure of the mid to
greater curvature of the stomach, vascular pedicle based on the right upper intrathoracic esophagus, dissection of esophageal pathologic
gastroepiploic artery, or long-segment colon, which is passed up into structures from the surrounding mediastinal structures, and the ability
the neck as vascularized graft to be anastomosed to the proximal to perform a mediastinal lymph node dissection. The disadvantages
cervical esophagus. Although reports exist regarding the use of jejunum are related to the use of a thoracotomy and its associated morbidity,
for long-segment esophageal replacement, small bowel is generally with limits on the proximal resection margin and the potential for
not an ideal option for esophageal replacement because of its mesenteric an intrathoracic esophageal anastomotic leak, which is a more difficult
vascular anatomy, unless specialized techniques with vascular augmenta- management problem than a cervical anastomotic leak. Reported
tion are used. The esophageal replacement conduit is passed through complications include respiratory problems in 11% to 20%, anastomotic
the chest into the neck by one of three routes: (1) subcutaneous, (2) leak in 3% to 7%, and wound infection in 5%. Operative mortality
substernal, or (3) posterior mediastinum. The posterior mediastinum ranges from none to 4%.165–167
is the preferred route when possible. The advantages of THE include
avoidance of postthoracotomy discomfort, wide proximal esophageal Left Thoracoabdominal Approach
margins to ensure complete resection of tumor and Barrett mucosa,
cervical anastomosis where the consequences of anastomotic leak are The left thoracoabdominal approach uses a single incision extending
minimized, and an esophageal reconstruction that results in an excellent from the left side of the chest onto the abdomen; it provides excellent
quality of swallowing. exposure of the lower third of the esophagus and left upper quadrant
The disadvantages of this approach include an inability to visualize of the abdomen.168 This technique is ideal for patients with limited
middle or proximal third tumors and to perform extensive intrathoracic tumors near the GEJ, especially when the extent of gastric invasion
regional lymphadenectomy, the potential for injury to intrathoracic is unclear, because it yields superb exposure and maximizes reconstruc-
and cervical structures, and the need for long-segment esophageal tive options of the lower third of the esophagus. Respiratory complica-
replacement. Randomized trials have confirmed similar long-term tions are the most common postoperative complications with this
survival with the use of a transhiatal approach despite a more limited approach. At least some degree of atelectasis, usually involving the
mediastinal lymph node dissection than can be accomplished via left lower lung, occurs in most patients. Pneumonia is reported to
thoracotomy. occur in up to 24% of cases. Anastomotic leaks occur in as many as
THE is safe, well tolerated, and associated with infrequent major 12% of cases, leading to a higher mortality rate. Other complications
complications in experienced hands.161 In a large series of more than include atrial fibrillation in 10%, wound infection in 1.5% to 5.2%,
2000 patients, the mortality rate was only 1% in those patients who and (infrequently) empyema and subphrenic abscess. The reported
underwent surgery in 1998 and later; anastomotic leaks and pulmonary operative mortality is none to 6.2%.168,169
complications occurred in 9% and 2% of patients, respectively.
A Dutch randomized trial evaluated the experimental arm of THE Multiple Incisions
with reconstruction with the gastric remnant compared with a right
thoracoabdominal approach.156 In this study, lymph node dissection Multiple-incision surgical approaches combine the incisional strategies
varied with the two surgical approaches. For all patients, periesophageal of the standard techniques. Of these, the three-incision approach
lymph nodes were removed, as was the lymph node–bearing region using a cervical incision (right or left), right thoracotomy, and midline
along the left gastric artery. However, in the right thoracoabdominal laparotomy, as described by McKeown, is the most common and is
group, a much more extensive dissection was performed, including also referred to as the three-incision, three-hole, total esophagectomy,
lower and middle mediastinal, subcarinal, and right-sided paratracheal or modified McKeown approach.170 It combines the exposure of the
lymph nodes (dissected en bloc). Through a midline laparotomy, the thoracotomy approach for esophageal mobilization or nodal dissection
paracardial, lesser curvature, left gastric artery (along with lesser with the advantages of a cervical esophageal anastomosis. Patient
curvature), celiac trunk, common hepatic artery, and splenic artery outcome results with this technique are similar to those of other
nodes were dissected, and a gastric tube was constructed. Despite the approaches, with reported mortality of 3% to 4% and esophageal
theoretical benefit of a more extensive nodal resection, the pattern of anastomotic leak rates of 5% or less.171
recurrence was similar: locoregional recurrence occurred in 14% and
12% of patients, respectively; distant recurrence in 25% and 18%; Radical Resections
and both in 18% and 19% (P = .60). Perioperative morbidity was
substantially less with transhiatal resection, especially pulmonary The majority of patients with esophageal cancer are first seen with
complications, but in-hospital mortality was similar. locally advanced disease (T3–4N0 or TanyN+ tumors). In these patients,
Intriguingly, a subgroup analysis of long-term outcome suggested survival results are poor with surgery alone. An approach to improve
a possible benefit based on lymph node status. Although 5-year OS outcomes involves adding an en bloc, wide-field lymphadenectomy to
did not differ between the THE and transthoracic groups (34% versus the standard esophagectomy technique. The esophagus has an extensive
36%; P = .71), 5-year locoregional disease-free survival (DFS) was regional lymphatic drainage. Arbitrarily, the lymphatic drainage has
improved in the transthoracic group for patients with one to eight been divided into three zones or fields—cervical, intrathoracic, and
positive lymph nodes (23% versus 64%; P = .02).159 abdominal. Standard esophagectomy techniques involve regional, or
one-field, lymphadenectomy. Radical approaches advocate two- or
Ivor Lewis Approach three-field lymphadenectomy in conjunction with esophageal resection
and replacement. Hagen and colleagues believe that proximal hemigas-
Partial esophagogastrectomy with an abdominal and right thoracotomy trectomy should also be included as part of an en bloc approach, using
approach was originally described by Welsh thoracic surgeon Ivor colon to replace the resected esophagus.172,173 Radical esophagectomy is

Downloaded for Tabanschii Nicolae (tabanschiirusuhan@gmail.com) at Nicolae Testemiceanu State Medical and Pharmaceutical University from
ClinicalKey.com by Elsevier on November 10, 2022. For personal use only. No other uses without permission. Copyright ©2022. Elsevier Inc. All rights
reserved.
Cancer of the Esophagus  •  CHAPTER 71 1187

more complex surgery than standard techniques. Nonetheless, 30-day and 30-day surgical mortality improved from 8.3% to 3.1%.182 In
mortality rates as low as 1.6% to 4.3% are reported.172,174–176 However, addition, better patient selection may play an important role in
survival data using radical esophagectomy techniques are conflicting, improved outcome. For example, Steyerberg and colleagues proposed
and therefore it is unknown whether there is sufficient benefit to a simple scale based on important comorbidities, age, preoperative
outweigh the increased operative morbidity. therapies, and esophagectomy volume at the treating hospital, which
divided patients into groups with predicted 30-day mortality of under
Minimally Invasive Esophagectomy 4% to approximately 20%.183 It is also expected that more accurate
preoperative staging with PET scan and EUS may improve surgical
With the advent of minimally invasive surgical techniques, an interest outcome by removing some patients with existing gross metastatic
developed in applying thoracoscopic and laparoscopic techniques to disease.
esophagectomy.177 Certainly, the techniques of gastric and esophageal Overall, despite advances in surgical techniques, postesophagectomy
mobilization have been well established for other complex minimally survival has remained remarkably stable over time, emphasizing that
invasive surgeries. Minimally invasive esophagectomy (MIE) required improvements would likely require effective combined modality
that these individual techniques be “spliced” together. Not unexpectedly, therapies.
a steep learning curve exists for these surgeries.
Approaches used have included laparoscopic transhiatal resec- PERIOPERATIVE CHEMOTHERAPY
tion, combined laparoscopic–thoracoscopic procedures, and AND SURGERY
laparoscopic creation of a gastric tube with thoracotomy and other
combinations. The need to convert to an open surgical procedure A strategy of perioperative chemotherapy is the predominant approach
has been uncommon. The number of lymph nodes removed also in Europe, based primarily on the phase III MAGIC (Medical Research
appears similar to that achieved with open surgery.178 Luketich from Council Adjuvant Gastric Infusional Chemotherapy) trial performed
the University of Pittsburgh has described results for more than in the United Kingdom.184 In this trial, 503 patients with gastric
a thousand patients, 76% with adenocarcinoma and 93% with cancer (26% of whom had tumors in the lower esophagus or GEJ)
distal and GEJ tumors.179 In this group, 98% underwent resection were randomized to three cycles (9 weeks) each of preoperative and
with negative margins, and nodal dissection recovered a median of postoperative ECF (epirubicin–cisplatin–5-FU) and surgery or surgery
21 nodes. alone. Perioperative chemotherapy resulted in significant improvement
There has been a single randomized trial examining the role of in 5-year OS (36% versus 23%; P = .009), establishing this regimen
MIE.155 In this randomized trial, 115 patients were randomized to as a standard of care.
MIE versus standard approaches, with the primary objectives including A similar degree of benefit was also noted in the contemporaneous
the short-term outcomes of pulmonary infections, pain, and QoL French FFCD 9703 trial of 224 patients with EG adenocarcinoma.185
end points.180 In this trial, 93% underwent preoperative therapy before Patients were randomized to six cycles (18 weeks) of perioperative
resection. The end points of infection, pain, and QoL were superior 5-FU–cisplatin followed by surgery versus surgery alone. Perioperative
in the group undergoing MIE. The R0 resection rates, number of chemotherapy in this trial was associated with a significant improvement
lymph nodes retrieved, and pathologic stage were similar in both in 5-year DFS (34% versus 19%; P = .003) and OS (38% versus
groups. Three-year survival data were recently published and show 24%; P = .02). Although comparisons among different clinical trials
no difference in DFS (37% versus 43%; P = .602) or OS (41% versus must be made cautiously, the survival benefit seen with 5-FU–cisplatin
43%; P = .633) for the open surgery versus MIE groups. in this trial appears to be nearly identical to that seen with ECF in
Overall, when comparing MIE with open procedures, anastomotic the MAGIC trial.
leaks, stricture rates, and survival seem to be comparable; however, The benefit of anthracycline and the duration of preoperative
MIE may be associated with a lower intraoperative blood loss, shorter therapy have now been definitively disputed by the MRC OEO-5
length of stay, and less morbidity, but offset by longer operative times study, which has so far been presented only in abstract form.186 In
and costs. this study, 897 patients with esophageal or GEJ adenocarcinomas
were randomized to preoperative chemotherapy with either 6 weeks of
SURVIVAL AFTER SURGERY ALONE 5-FU–cisplatin or 12 weeks of ECX (epirubicin-cisplatin-capecitabine)
chemotherapy. Although the pathologic RR was improved in the
Cancer-related survival after surgical resection is discussed separately ECX group versus the 5-FU–cisplatin group (including a pathologic
from the description of individual techniques to emphasize the fact complete response [pCR] rate of 11% versus 3% in the patients who
that postesophagectomy survival is a function of stage and not of underwent surgery), there was no difference in median PFS (1.78 versus
surgical approach. 1.53 years; P = .058) or OS (2.15 versus 2.02 years; P = .86) between
Regardless of whether a thoracotomy or nonthoracotomy technique the groups.
is used, long-term OS is similar, at approximately 20% to 25%. This The OEO-5 study has therefore also raised the provocative sug-
fact has been underscored most graphically by Muller and associates, gestion that as little as 6 weeks of preoperative chemotherapy conveys
who reviewed the world literature to compare overall postesophagectomy the same OS benefit as 12 weeks of chemotherapy. Although this
survival by technique and found no significant difference.155 Hulscher may be counterintuitive based on the findings of the MAGIC and
and colleagues performed a meta-analysis of the English language FFCD studies and other studies in gastric cancer in which 6 to 12
literature of transthoracic and transhiatal resection of esophageal cancer months of adjuvant chemotherapy have been administered,187,188
and found a higher risk of pulmonary morbidity and mortality with these results are not without precedent. As will be discussed later,
the transthoracic procedure but a similar 5-year OS of approximately the CROSS study found benefit for preoperative chemoradiation in
20%.181 These investigators also compared limited transhiatal resection which patients received only five weekly treatments of carboplatin-
with an extended en bloc lymphadenectomy in 220 patients with paclitaxel as the entirety of their systemic chemotherapy.63,189 The
esophageal adenocarcinomas. No significant difference was found in absolute improvement in OS seen in the CROSS study is also very much
survival or operative mortality. in the range of 10% to 15% seen in other positive phase III studies
Although the surgical procedure does not affect outcome, large discussed here.
retrospective series have suggested a modest improvement in long-term It is also important to remember that although the MAGIC and
outlook in recent years, probably the result of lower operative mortality. FFCD studies aimed to deliver 18 weeks of perioperative chemotherapy,
For example, Gockel and colleagues found that for the periods 1985 in both studies only 50% of patients who underwent preoperative
to 1995 and 1995 to 2005, 5-year OS increased from 15% to 25%, chemotherapy and surgery were able to receive or complete adjuvant

Downloaded for Tabanschii Nicolae (tabanschiirusuhan@gmail.com) at Nicolae Testemiceanu State Medical and Pharmaceutical University from
ClinicalKey.com by Elsevier on November 10, 2022. For personal use only. No other uses without permission. Copyright ©2022. Elsevier Inc. All rights
reserved.
1188 Part III: Specific Malignancies

chemotherapy, further suggesting that most patients derived the survival CHEMOTHERAPY AFTER SURGERY
benefit of chemotherapy from receiving significantly less than 18 weeks
of treatment. The generally poor ability to deliver therapy after surgery In comparison to chemoradiation, trials in East Asia of resectable
was also seen in the recently presented Dutch CRITICS study (to be gastric cancer have frequently focused on postoperative chemotherapy
discussed later) and would suggest, especially given the absence of a alone. To date, two large phase III trials have demonstrated a benefit
biologic rationale to split up the same systemic treatment with surgery, for this approach. These data support the use of adjuvant fluoropy-
that future experimental strategies should focus on exclusively preopera- rimidines as monotherapy and combination chemotherapy with a
tive approaches. fluoropyrimidine plus a platinum agent. The results are summarized
Aside from the MAGIC and FFCD studies, other contemporary in Table 71.2 but should be interpreted with caution because these
phase III evaluations of preoperative or perioperative chemotherapy trials have exclusively enrolled patients with gastric adenocarcinoma.
in esophagogastric adenocarcinomas have either been negative or had In East Asia, less than 10% of tumors occur in the proximal stomach
more marginal benefit. The North American Intergroup 113 trial or GEJ, making it unclear if they are applicable to patients with
failed to show a survival benefit for perioperative 5-FU–cisplatin in esophageal or GEJ adenocarcinoma.
440 patients with esophageal cancer (approximately half of whom ACTS-GC (Adjuvant Chemotherapy Trial of TS-1 for Gastric
had adenocarcinomas; eligibility limited extension of the tumor to Cancer) was performed in Japan. In this study of 1059 patients with
2 cm beyond the GEJ into the stomach).190 The MRC OEO-2 trial, stage II/III gastric cancer who had undergone D2 resections, patients
in which 802 patients were randomized to surgery alone versus two were randomized to 1 year of adjuvant S-1 versus observation.187
cycles of preoperative 5-FU–cisplatin, reported a modest improvement Five-year outcomes for this trial were updated in 2011, confirming
in 5-year OS with chemotherapy (23% versus 17%; P = .03).191 that adjuvant S-1 is associated with significant improvements in 5-year
Two-thirds of patients had adenocarcinomas and three-quarters of relapse-free survival (RFS) (65.4% versus 53.1%; HR, 0.65; 95% CI,
tumors were in the lower esophagus or gastric cardia. Most recently, 0.54–0.79) and OS (71.7% versus 61.1%; HR, 0.67; 95% CI,
the European EORTC 40954 trial evaluated a strategy of preoperative 0.54–0.83) compared with observation alone.194 Subgroup analyses
5-FU–leucovorin-cisplatin in 144 patients with GE and gastric adeno- revealed benefit for all groups, including by stage and histologic type.
carcinoma.192 The trial was stopped because of poor accrual, which The second of these trials is the CLASSIC trial (capecitabine and
limited the power of the study, and no differences in survival were oxaliplatin adjuvant study in stomach cancer), which was performed
detected. in 1035 East Asian patients who had undergone a D2 resection of
These data are summarized in Table 71.1. An updated meta-analysis stage II–IIIB gastric cancer.188 Patients were randomized to 6 months
by Sjoquist and colleagues of 10 randomized trials involving preoperative of adjuvant capecitabine-oxaliplatin versus observation. Updated survival
chemotherapy for esophageal and GEJ cancers suggested a 13% data confirmed improved 5-year OS for patients who received che-
decreased risk of all-cause mortality for this approach in patients with motherapy (78% versus 69%; HR, 0.66; P = .0015); 5-year DFS was
adenocarcinomas versus surgery alone (HR, 0.87; 95% CI, 0.79–0.96; also improved (68% versus 53%; HR, 0.58; P < .0001).195
P < .005).193 In this meta-analysis, both the MAGIC and EORTC The possibility of benefit for an adjuvant chemotherapy-only
40954 trials were excluded because outcomes were not stratified based approach for esophageal or GEJ adenocarcinoma was suggested by
on gastric versus GEJ tumors. a phase II study performed by ECOG of four cycles of postopera-
In comparison with a benefit for adenocarcinoma tumors, there tive cisplatin-paclitaxel in patients with pT3–4N+ adenocarcinomas
was only a trend toward benefit for preoperative chemotherapy in of the esophagus, GEJ, and cardia.196 The results confirmed the
SCC tumors (HR, 0.92; 95% CI, 0.81–1.04; P = .18). feasibility of the approach. Comparison with historical controls

Table 71.1  Esophageal Cancer Preoperative Chemotherapy Phase III Trials


SURVIVAL
Histologic No. of R0 Resection Pathologic Local
Treatment Type Patients Rate CR Rate Median Overall Failure Reference
Perioperative ECF + surgery Adenocarcinoma 250 69% 0% 24 mo 5 yr, 36% 14% Cunningham
Surgery 253 66% N/A 20 mo 5 yr, 23% 21% et al184
Perioperative 5-FU/cisplatin Adenocarcinoma 109 87% NS NS 5 yr, 38% 24% Ychou et al185
+ surgery
Surgery 110 74% N/A NS 5 yr, 24% 26%
Preoperative ECX + surgery Adenocarcinoma 446 67% 11% 25.8 3 yr, 42% NS Alderson et al186
Preoperative 5-FU/cisplatin 451 60% 3% 24.2 3 yr, 39% NS
+ surgery
Perioperative 5-FU/cisplatin Adenocarcinoma 213 62% 2.5% 14.9 mo 3 yr, 23% 32% Kelsen et al190
+ surgery (54%) + SCC
Surgery 227 59% N/A 16.1 mo 3 yr, 26% 31%
Preoperative 5-FU/cisplatin Adenocarcinoma 400 60% NS 16.8 mo 5 yr, 23% 19% Medical Research
+ surgery (66%) + SCC Council,260 Allum
Surgery 402 54% N/A 13.3 mo 5 yr, 17% 17% et al191
Preoperative 5-FU/LV/ Adenocarcinoma 72 82% 7.1% 64.6 mo 2 yr, 73% NS Schuhmacher
cisplatin + surgery et al192
Surgery 72 66.7% N/A 52.5 mo 2 yr, 69.9% NS

Boldface indicates statistically significant difference.


CR, Complete response; ECF, epirubicin, cisplatin, 5-fluorouracil; ECX, epirubicin, cisplatin, capecitabine; 5-FU, 5-fluorouracil; LV, leucovorin; N/A, not applicable; NS, not stated;
SCC, squamous cell carcinoma.

Downloaded for Tabanschii Nicolae (tabanschiirusuhan@gmail.com) at Nicolae Testemiceanu State Medical and Pharmaceutical University from
ClinicalKey.com by Elsevier on November 10, 2022. For personal use only. No other uses without permission. Copyright ©2022. Elsevier Inc. All rights
reserved.
Cancer of the Esophagus  •  CHAPTER 71 1189

Table 71.2  Results of Phase III Postoperative Chemotherapy Trials in Esophageal and Gastric Cancer
SURVIVAL
No. of Local
Treatment Histologic Type Patients Median Overall Failure Reference
Surgery Adenocarcinoma 530 NR 5 yr, 61% 2.8% Sakuramoto et al,187
Surgery + S-1 (gastric) 529 NR 5 yr, 72% 1.3% Sasako et al194
Surgery Adenocarcinoma 515 NRa 5 yr, 78% 44% Bang et al.188,195
Surgery + Capeox (gastric) 520 NRa 5 yr, 69% 21%
Surgery SCC 100 NS 5 yr, 45% 30% Ando et al197
Surgery + cisplatin/vindesine 105 NS 5 yr, 48% 30%
Surgery SCC 122 NS 5 yr, 52% 46% Ando et al198
Surgery + 5-FU/cisplatin 120 NS 5 yr, 61% 8%
5-FU/cisplatin + surgery SCC 164 NS 5 yr, 55% NS Ando et al199
Surgery + 5-FU/cisplatin 166 NS 5 yr, 43% NS

a
Disease-free survival. Numbers in bold indicate statistically significant differences.
Capeox, Capecitabine-oxaliplatin; CR, complete response; 5-FU, 5-fluorouracil; NR, not reached; NS, not stated; SCC, squamous cell carcinoma.

suggested an improved survival (60% versus 38% 2-year survival; chemoradiation was established as the standard of care in the nonsurgical
P = .0008). management of locally advanced esophageal SCC.
In comparison, postoperative chemotherapy for resected esophageal Since then, preoperative chemoradiation has been evaluated
SCC has not clearly been shown to be beneficial. This approach was extensively in trials of esophageal or GEJ cancer. Six contemporary
studied in two Japanese randomized trials in which patients with SCC randomized trials have compared preoperative chemoradiation followed
histology were randomized to receive two cycles of chemotherapy by surgery versus surgery alone.63,202–206 Of these, three have been
with cisplatin-vindesine197 or 5-FU–cisplatin,198 respectively. Although positive and revealed a survival benefit for this approach. These results
the trial with cisplatin-vindesine did not show any survival benefit, are summarized in Table 71.3.
an unplanned subset analysis of the trial with 5-FU–cisplatin revealed A potential new standard of care was established by the rigorously
a survival benefit for patients with lymph node involvement (5-year conducted Dutch CROSS trial.63 In this study of 366 evaluable patients
DFS of 52% versus 38%). with esophageal tumors (of which 75% and 65%, respectively, were
The possible benefit of postoperative therapy suggested by the adenocarcinomas and lymph node positive at EUS), patients were
aforementioned trial led to a subsequent Japanese trial in which 330 randomized to preoperative carboplatin-paclitaxel combined with
patients with SCC histologic type were randomized to surgery and 41.4 Gy of radiation versus surgery alone. Preoperative chemoradiation
either two cycles of preoperative or postoperative 5-FU–cisplatin.199 The resulted in an improvement in R0 resection rates (92% versus 67%;
results showed improved 5-year OS for the preoperative chemotherapy P < .001), in a pCR rate of 29% (23% for adenocarcinoma and 49%
group (55% versus 43%; P = .04), further raising questions about for SCC), and in improved OS compared with surgery alone (median
the value of adjuvant chemotherapy in these patients. However, OS, 49.4 versus 24.0 months; 3-year OS, 58% versus 44%; P = .003).
interpretation of these results is confounded by the fact that only Preoperative therapy was also relatively well tolerated, with mostly
96 of the 166 patients randomized to postoperative chemotherapy grade 3 toxicities noted in only 20% of patients (13% nonhematologic,
received any treatment; 38 patients who had pN0 disease were not 7% hematologic). There did appear to be a greater degree of benefit
treated per protocol because of the prior observation that adjuvant for patients with SCC versus adenocarcinoma histologic type (univariate
chemotherapy benefited only patients with lymph node involvement. HR for death, 0.45 versus 0.73), but all patients derived benefit.
Furthermore, this study observed that a survival benefit for preoperative Long-term follow-up of this study confirmed an OS benefit for both
chemotherapy was seen only in N0 patients, which contrasted with adenocarcinoma and SCC patients, and a 9% reduction in distant
the prior postoperative adjuvant study that claimed a benefit only in metastases was reported for patients who received preoperative
N1 patients. chemoradiation.189
Although this study demonstrated a clear benefit for chemoradiation,
PREOPERATIVE CHEMORADIATION it is not possible to definitively conclude that carboplatin-paclitaxel
is the preferred regimen when combined with radiation relative to
AND SURGERY standard 5-FU–cisplatin used in other trials. Nevertheless, the pCR
Standard Approaches rate of 49% in SCC is the highest ever reported in a phase III trial,
and the pCR rate of 23% for adenocarcinomas compares favorably
The seminal phase III US RTOG 85-01 study demonstrated the with the findings of other phase II/III studies. Coupled with the ease
superiority of chemoradiation over radiation alone.200 This nonoperative of administration and tolerability, carboplatin-paclitaxel has become
study compared standard fractionation radiation (64 Gy) versus the new standard of care and has become the reference regimen for
radiation (50 Gy) plus concurrent 5-FU–cisplatin. The trial was stopped future trial design. However, initial results of the CALGB 80803
when data from 121 patients showed an improved median OS in study suggest that a 5-FU–platinum regimen may be comparable or
favor of chemoradiation (12.5 versus 8.9 months). Two-year survival even possibly superior (see later).
was also improved in the chemoradiation group (38% versus 10%), Overall, many of the other randomized trials conducted were
as was 5-year survival (21% versus 0%).201 Although the majority of associated with methodologic concerns (including the lack of rig-
patients treated in this trial had SCC, long-term survival was also orous pretherapy staging with EUS and/or laparoscopy) and were
seen in the small number of adenocarcinoma patients in the trial, significantly smaller than randomized preoperative chemotherapy trials
with 13% of patients alive at 5 years. Based on this study, (e.g., the positive CALGB 9781 study enrolled only 56 patients).

Downloaded for Tabanschii Nicolae (tabanschiirusuhan@gmail.com) at Nicolae Testemiceanu State Medical and Pharmaceutical University from
ClinicalKey.com by Elsevier on November 10, 2022. For personal use only. No other uses without permission. Copyright ©2022. Elsevier Inc. All rights
reserved.
1190 Part III: Specific Malignancies

Table 71.3  Results of Phase III Preoperative Chemoradiation Trials in Esophageal Cancer
SURVIVAL
No. of R0 Resection Pathologic Local
Treatment Histologic Type Patients Rate CR Rate Median Overall Failure Reference
Preoperative CRT Adenocarcinoma (76%) 50 45% 24% 16.9 mo 3 yr, 30% 19% Urba et al204
Surgery + SCC 50 45% N/A 17.6 mo 3 yr, 16% 42%
Preoperative CRT Adenocarcinoma 58 NS 25% 16 mo 3 yr, 32% NS Walsh et al205
Surgery 55 N/A 11 mo 3 yr, 6%
Preoperative CRT SCC 143 81% 26% 18.6 m 5 yr, 26% NS Bosset et al.202
Surgery 139 69% N/A 18.6 mo 5 yr, 26%
Preoperative CRT Adenocarcinoma (63%) 128 80% 9% 22.2 mo NS 15% Burmeister et al203
Surgery + SCC + other 128 59% N/A 19.3 mo 26%
Preoperative CRT Adenocarcinoma (75%) 30 NS 40% 4.5 yr 5 yr, 39% NS Tepper et al206
Surgery + SCC 26 N/A 1.8 yr 5 yr, 16%
Preoperative CRT Adenocarcinoma (74%) 178 92% 29% 49.4 mo 3 yr, 58% NS van Hagen et al63
Surgery + SCC 188 69% N/A 24.0 mo 3 yr, 44%

CR, Complete response; CRT, chemoradiation; N/A, not applicable; NS, not stated; SCC, squamous cell carcinoma.

Although the results of these trials are conflicting, they do at a radiation or chemoradiation.209 Although there are technical and
minimum suggest improved curative resection rates and decreased local radiobiologic differences between high-dose and low-dose rates, there
recurrence. are no clear therapeutic advantages.
A benefit for preoperative chemoradiation is supported by the In the RTOG 92-07 trial, 75 patients (92% with SCC) received
previously discussed meta-analysis in which 13 randomized trials of the RTOG 85-01 combined modality regimen (5-FU–cisplatin/50 Gy)
preoperative chemoradiation (including the five aforementioned trials) followed by a boost during cycle 3 of chemotherapy with either low–
were analyzed.193 Preoperative chemoradiation was associated with a dose rate or high–dose rate brachytherapy.210 Although the complete
decreased risk of all-cause mortality of 25% (HR, 0.75; 95% CI, RR was 73% with a median follow-up of only 11 months, local
0.59–0.95; P = .02) in patients with adenocarcinoma histologic type failure as the first site of failure occurred in 27%. Acute toxicity
versus surgery alone. There also appeared to be benefit for SCC tumors included 58% grade 3 toxicities, 26% grade 4 toxicities, and 8%
(HR, 0.80; 95% CI, 0.68–0.93; P = .004). treatment-related deaths. The cumulative incidence of fistula was 18%
per year, and the crude incidence was 14%. Of the six treatment-related
Intensification of Combined Modality Therapy fistulas, three were fatal. Given this significant toxicity and a lack
of clear improvement in outcomes, this approach has been largely
Neoadjuvant Chemotherapy abandoned.
The administration of systemic chemotherapy before chemoradiation
has several theoretical benefits. In a disease that is moderately che- External-Beam Radiotherapy
mosensitive, more chemotherapy may better control micrometastatic
disease. The approximately 70% dysphagia improvement rates of INT 0123 (RTOG 94-05) was the follow-up trial to RTOG 85-01.
chemotherapy may also reduce the need for feeding tube placement In this trial, patients with either SCCs (85%) or adenocarcinomas
during chemoradiation. (15%) who were selected for a nonsurgical approach were randomized
Despite these theoretical benefits, there does not appear to be a to a slightly modified RTOG 85-01 combined modality regimen with
benefit to such an approach. To date, the largest study that has evaluated 50.4 Gy versus the same chemotherapy with 64.8 Gy.211
this approach is a randomized phase II study performed at MD The study was closed when an interim analysis revealed that it was
Anderson Cancer Center.207 One hundred and twenty-six patients unlikely that the high-dose arm would achieve a superior survival
were randomized to receive either induction infusional 5-FU–oxaliplatin compared with the standard-dose arm. For the 218 eligible patients,
followed by chemoradiation with 5-FU–oxaliplatin or chemoradiation there was no significant difference in median OS (13.0 versus 18.1
alone, followed by surgery in both groups. There was a nonsignificant months) or 2-year survival (31% versus 40%) in the high-dose versus
trend toward an increased pCR rate in the patients who received standard-dose arm. Eleven treatment-related deaths occurred in the
induction chemotherapy (13% versus 26%; P = .094) but absolutely high-dose arm compared with two in the standard-dose arm. However,
no difference in survival outcomes (median OS, 43.7 versus 45.6 7 of the 11 deaths occurred in patients who had received less than
months; P = .69). 50.4 Gy, making it unclear if the higher dose of radiation was
Although induction chemotherapy may not be beneficial in responsible for the apparent increased mortality. Based on the findings
and of itself, it can allow for early assessment by PET scan, to be of the INT 123 trial, the standard dose of external-beam radiation
discussed later. remains 50.4 Gy.

Intensification of the Radiation Dose Preoperative Chemoradiation for


Intraluminal Brachytherapy Early-Stage Disease
Another approach to the dose intensification of chemoradiation is Although the aforementioned studies focused on locally advanced
increasing the radiation dose, through either intraluminal brachytherapy tumors (which, by contemporary standards, would include staging
and/or external beam radiotherapy. with EUS and consist mostly of uT3–4N+ tumors), in the French
Intraluminal brachytherapy has been used both as primary therapy FFCD 9901 study, 195 patients with early-stage cT1–2Nany or cT3N0
(usually as a palliative modality)208 and as a boost after external beam tumors were treated with preoperative 5-FU–cisplatin and radiation

Downloaded for Tabanschii Nicolae (tabanschiirusuhan@gmail.com) at Nicolae Testemiceanu State Medical and Pharmaceutical University from
ClinicalKey.com by Elsevier on November 10, 2022. For personal use only. No other uses without permission. Copyright ©2022. Elsevier Inc. All rights
reserved.
Cancer of the Esophagus  •  CHAPTER 71 1191

and surgery versus surgery alone.212 Seventy-two percent of patients randomized to adjuvant chemotherapy and chemoradiation with bolus
in this study had SCCs, and 24% and 74%, respectively, had cT1 5-FU–leucovorin versus observation alone after surgery. Patients who
and cN0 tumors. This study revealed a strikingly high 93% R0 resection received adjuvant chemoradiation had an improvement in RFS (3-year
rate in the surgery-alone arm, which was not improved with preoperative RFS, 48% versus 31%; P < .001; and 3-year OS, 51% versus 40%;
chemoradiation. Similarly, median DFS and OS were not significantly P = .005). Despite these positive results, this trial is frequently criticized
different in both arms, but in-hospital postoperative mortality was because of the relatively inadequate surgical resections that were
significantly increased in the chemoradiation arm (11.1% versus 3.4%; performed; 54% of patients had less than a D1 or D2 resection, which
P = .049). is less than an optimal resection of the involved lymph nodes. It has
These results are somewhat surprising. Despite the lack of been argued that radiation in this setting potentially compensated for
improvement in R0 resection rates, locoregional recurrence was inadequate surgery because the greatest impact of adjuvant chemoradia-
reduced in the chemoradiation arm (22.1% versus 28.9%; P = .02); tion was a reduction in local recurrence of cancer. Such benefits may
on the other hand, the rate of distant recurrence was not signifi- not be seen for radiotherapy if a more complete D1 or D2 surgical
cantly different between both arms (22.5% versus 28.9%; P = .31). resection is undertaken.
In addition, the unexpectedly high postoperative mortality in the Based on the results of the Intergroup trial, the CALGB launched
chemoradiation arm, compared with 4% in both treatment arms of and completed the 80101 trial. A total of 546 gastric cancer patients
the CROSS study, might have obscured a small survival benefit from (30% of whom had tumors involving the GEJ and proximal stomach)
chemoradiation. were enrolled. The standard arm consisted of systemic bolus 5-FU–
Nevertheless, these results are not necessarily inconsistent with leucovorin preceding and following chemoradiation with infusional
other published data. An accepted approach, discussed later, is definitive 5-FU; the experimental arm intensified the systemic chemotherapy
chemoradiation without surgery for patients with SCC who achieve by replacing the bolus 5-FU–leucovorin with the ECF regimen.
a clinical complete response (CR). Because 72% of patients in this Results have been presented in abstract form and reveal no improve-
study had SCC tumors, it suggests the complementary conclusion ment in 3-year DFS (47% versus 46%) or OS (52% versus 50%)
that SCC patients who do undergo chemoradiation should not undergo with the addition of an anthracycline and platinum compound to
mandatory surgery. Similarly, these results are concordant with current 5-FU.215 These results are also virtually identical to the outcomes in
guidelines from the National Comprehensive Cancer Network, which the adjuvant chemoradiation arm of the Intergroup 116 trial. These
recommend surgery alone for patients with cT1N0 tumors. results indicate that 5-FU monotherapy, combined with radiation,
remains a standard of care and that adding cisplatin and epirubicin
Preoperative Chemoradiation Versus Chemotherapy to adjuvant chemotherapy failed to improve survival. ECF should
not be used as an adjuvant chemotherapy regimen, although preop-
The possible superiority of preoperative chemoradiation over preopera- erative and postoperative ECF without radiation therapy remains a
tive chemotherapy was suggested by the German POET (Preoperative care standard.
Chemotherapy or Radiochemotherapy in Esophagogastric Adenocar- Finally, the Dutch CRITICS study evaluated an alternative to the
cinoma Trial) study, in which patients with GEJ adenocarcinomas perioperative ECF approach by randomizing patients (38% of whom
were randomized to either 5-FU–leucovorin–cisplatin followed by had GEJ or proximal gastric adenocarcinomas) to perioperative ECX or
surgery or 5-FU–leucovorin–cisplatin followed by chemoradiation EOX (epirubicin-oxaliplatin-capecitabine) chemotherapy (the control
with cisplatin-etoposide and then surgery.213 One hundred nineteen arm) versus preoperative ECX-EOX and adjuvant chemoradiation
eligible patients were randomized before the trial was closed because with capecitabine. The results revealed no difference in PFS and
of poor accrual, limiting the power of this study to detect a difference OS for either treatment arm.216 Although subgroup analyses are
between the treatment groups. Nevertheless, patients who received planned, the nearly superimposable Kaplan-Meier survival curves
preoperative chemoradiation had a higher pCR rate (15.6% versus confirm, for now at least, that patients who have received preopera-
2%; P = .03) and tumor-free lymph node status (ypN0 64.4% versus tive chemotherapy should not receive adjuvant chemoradiation in a
36.7%; P = .01) than those who received preoperative chemotherapy. standard fashion.
There were also trends toward an improvement in local control (76.5% These results are shown in Table 71.4.
versus 59%; P = .06) and in 3-year OS (47.4% versus 27.7%; P =
.07) for the chemoradiation group that nearly approached statistical IS SURGERY NECESSARY AFTER COMBINED
significance. MODALITY THERAPY?
A similar nonsignificant trend toward improved outcomes with
preoperative chemoradiation over chemotherapy for adenocarcinoma Two randomized European trials compared definitive chemoradiation
histologic type was also suggested in the meta-analysis by Sjoquist versus chemoradiation followed by surgery in (mostly) esophageal
and colleagues, which revealed an all-cause mortality HR of 0.88 SCC patients.217,218 Taken together, both studies suggest that local
(95% CI, 0.76–1.01; P = .07) favoring chemoradiation.193 control is improved by subsequent surgery but that there is no clear
A major benefit of preoperative chemoradiation over chemotherapy improvement in survival.
alone may stem from an improvement in R0 resection rates for tumors In the setting of definitive chemoradiation, FOLFOX (5-FU–
that involve the GEJ. For example, the R0 resection rates in the 1400 leucovorin–oxaliplatin) also appears to be a comparable option to
patients treated in the contemporary MAGIC and OEO-5 studies 5-FU–cisplatin based on the French PRODIGE5/ACCORD17
were consistently below 70%; in the OEO-5 study, the majority of study, in which 267 patients were randomized to either regimen with
patients were assessed to be surgical candidates based on preoperative radiation as definitive therapy.219 Eighty-five percent of the patients
EUS and PET scan. The addition of radiation to preoperative che- in this study had SCC. Survival and toxicities were comparable in
motherapy in the CROSS study improved the R0 resection rate to both arms.
greater than 90%. An interesting question that arises from one of these studies (the
FFCD 9102 trial) is whether patients who do not respond to initial
POSTOPERATIVE CHEMORADIATION therapy benefit from subsequent surgery. In this study, patients
received initial treatment with 5-FU–cisplatin and radiation; only
In the United States, a standard of care is postoperative chemoradiation responders were subsequently randomized to surgery versus additional
for GEJ and gastric cancers undergoing up-front resection, based chemoradiation. A total of 192 of the 451 registered patients were
primarily on the results of the Intergroup 116 trial.214 In this trial, not randomized to further protocol therapy after initial chemoradia-
556 patients (20% of whom had tumors that involved the GEJ) were tion, primarily because of a lack of response (111 patients) but also

Downloaded for Tabanschii Nicolae (tabanschiirusuhan@gmail.com) at Nicolae Testemiceanu State Medical and Pharmaceutical University from
ClinicalKey.com by Elsevier on November 10, 2022. For personal use only. No other uses without permission. Copyright ©2022. Elsevier Inc. All rights
reserved.
1192 Part III: Specific Malignancies

Table 71.4  Results of Phase III Postoperative Chemoradiation Trials in Esophageal Cancer
DISEASE-FREE
SURVIVAL OVERALL SURVIVAL
No. of Local
Treatment Patients Median Overall Median Overall Failurea Reference
Surgery 275 19 mo 3 yr, 31% 27 mo 3 yr, 41% 29% MacDonald et al214
Postoperative 5FU/LV → 5-FU/RT → 5-FU/LV 281 30 mo 3 yr, 48% 36 mo 3 yr, 50% 19%
Postoperative 5-FU/LV → 5-FU/RT → 5-FU/LV 280 30 mo 3 yr, 46% 36.6 mo 3 yr, 50% NS Fuchs et al215
5 yr, 41%
Postoperative ECF → 5-FU/RT → ECF 266 28 mo 3 yr, 47% 37.8 mo 3 yr, 52% NS
5 yr, 44%
ECX/EOX → Surgery → ECX/EOX 393 27.6 mo 5 yr, 38.5% 42 mo 5 yr, 40.8% NS Verheij et al216
ECX/EOX → Surgery → Chemoradiation 395 30 mo 5 yr, 39.5% 39.6 mo 5 yr, 40.9% NS

a
Local failure with or without distant recurrence; entries in bold indicate statistically significant differences.
ECF, Epirubicin/cisplatin/infusional 5-fluorouracil; ECX, epirubicin/cisplatin/capecitabine; EOX, epirubicin/oxaliplatin/capecitabine; 5-FU, 5-fluorouracil; LV, leucovorin; NS, not
stated.

because of medical contraindication or patient refusal.220 Of these findings did suggest that postoperative mortality was not increased
nonrandomized patients, 112 subsequently underwent surgery. The by delaying surgery. Therefore such an approach can be considered
median OS for the nonresponding patients who underwent surgery for select patients at institutions with significant experience with
was significantly superior to the median OS of those who did not (17.0 this strategy.
versus 5.5 months; P < .0001) and was comparable to the median
OS of the patients who were randomized (18.9 months; P = .40). POSITRON EMISSION TOMOGRAPHY–
Although there are clear limitations and potential strong confounders DIRECTED THERAPY
to such an analysis, these data suggest that salvage esophagectomy may
be beneficial in a subset of patients who do not respond to initial Fluorodeoxyglucose–positron emission tomography (FDG-PET)
therapy. scanning is emerging as an important tool to investigate response to
On the other hand, there are no randomized data in patients with therapy. Several studies in esophagogastric tumors have demonstrated
adenocarcinomas to suggest that definitive chemoradiation is comparable that the degree of response detected with PET after preoperative
to chemoradiation and surgery. However, given the significant morbidity chemoradiation224,225 or chemotherapy226,227 is highly correlated with
and mortality associated with esophagectomy even at high-volume pathologic response at surgery and with patient survival.
institutions, one option is to closely follow adenocarcinoma patients The German MUNICON trial evaluated the strategy of taking
who achieve a clinical CR to preoperative chemoradiation—especially patients with locally advanced GEJ adenocarcinomas with a suboptimal
those who are relatively frail—with repeat endoscopy and imaging. response to 2 weeks of induction chemotherapy with 5-FU–cisplatin,
Those patients who develop locoregional failure without distant as determined with serial PET scans, directly to surgery, instead of
metastases may then be evaluated for salvage esophagectomy. continuing with presumably ineffective chemotherapy. Patients with
A major concern about such an approach is that postoperative a metabolic response demonstrated with PET (defined as ≥35%
complications and deaths may significantly increase when surgery is reduction in standard uptake value between baseline and repeat PET
delayed beyond the standard 6- to 8-week break after chemoradiation. scan) continued with an additional 12 weeks of chemotherapy before
The group at MD Anderson Cancer Center reported their experience surgery.228 This trial revealed a significantly improved R0 resection
in 65 patients with esophageal adenocarcinoma who underwent salvage rate (96% versus 74%; P = .002), major pathologic RR (58%
esophagectomies a median of 216 days after chemoradiation.221 When versus 0%; P = .001), median event-free survival (29.7 versus 14.1
compared with matched patients who underwent planned esophagec- months; P = .002), and median OS (median not reached versus 25.8
tomy after chemoradiation, postoperative complications and survival months; P = .015) for PET responders versus PET nonresponders.
did not appear to be different. The outcome for PET nonresponders referred for immediate surgery
Similar conclusions were drawn from a retrospective French was similar to the outcome of such patients in an earlier trial who
review, which examined this approach in 848 patients, 540 of whom completed 3 months of preoperative chemotherapy,226 indicating
underwent esophagectomy after preoperative chemoradiation and that nonresponding patients were not compromised by referral to
308 who underwent salvage surgery after chemoradiation for either immediate surgery. These results therefore support the early discontinu-
persistent cancer (234 patients) or recurrent cancer (74 patients).222 ation of inactive preoperative chemotherapy in PET nonresponder
Although the rates of anastomotic leak and surgical infection were patients.
increased in the salvage versus immediate surgery groups, in-hospital Building on the results of the MUNICON trial, the MUNICON-2
mortality rates were similar and relatively high in both groups (8.4% trial attempted to improve outcome in the PET nonresponders to the
versus 9.3%; P = .688). Three-year DFS (39.2% versus 32.8%; P = same regimen of preoperative 5-FU–cisplatin by treating them with
.232) and OS (43.3% versus 40.1%; P = .542) were also similar in “salvage” chemoradiation with cisplatin before surgery.229 When
both groups. compared with the PET responders who completed 3 months of
In addition, the RTOG 0246 study evaluated induction che- 5-FU–cisplatin before surgery, the PET nonresponders had inferior
motherapy with 5-FU–cisplatin-paclitaxel and chemoradiation 2-year PFS (64% versus 33%; P = .035) and a trend toward inferior
with 5-FU–cisplatin in 43 patients with locally advanced esopha- 2-year OS (71% versus 42%; P = .10). These results likely speak to
geal cancer; salvage surgery was reserved for patients with locally the underlying unfavorable biologic features of the tumors of PET
persistent or recurrent disease.223 Although the study did not meet nonresponders but do not rule out the possibility that such patients
its primary end point of improving 1-year survival to 77.5%, the can receive effective salvage therapy. In this trial, the chemotherapy

Downloaded for Tabanschii Nicolae (tabanschiirusuhan@gmail.com) at Nicolae Testemiceanu State Medical and Pharmaceutical University from
ClinicalKey.com by Elsevier on November 10, 2022. For personal use only. No other uses without permission. Copyright ©2022. Elsevier Inc. All rights
reserved.
Cancer of the Esophagus  •  CHAPTER 71 1193

administered with radiation (cisplatin) had already been assessed to incidence of airway invasion with or without fistulas also appears to
be associated with suboptimal outcomes at PET scanning when have decreased.
administered as induction therapy. Irrespective of its incidence, the treatment of esophageal cancer
Another possible strategy would be to use PET assessment after with airway invasion with or without fistula continues to present
induction chemotherapy to dictate subsequent chemotherapy during difficult management decisions. Primary surgical resection is generally
concurrent radiation. Responding patients can continue with the same considered inadvisable because of the substantial treatment-related
chemotherapy regimen during concurrent radiation, whereas nonre- mortality and morbidity. In a series of 28 patients with known airway
sponding patients can be switched to alternative, non–cross-resistant invasion without fistulas who underwent surgical exploration with or
chemotherapy during radiation. Our group has reported long-term without esophagectomy, the operative mortality was 17%.235 Other
DFS in patients whose disease progressed on induction chemotherapy approaches have included palliative treatments, such as the placement
but who were changed to alternative chemotherapy during subsequent of endoesophageal stents to relieve malignant dysphagia or to occlude
chemoradiation.230 a fistula or single-modality radiation therapy.236 Left untreated, patients
We have also since retrospectively reviewed our experience of with airway fistulas have a median survival of only 1 to 6 weeks.236,237
changing to alternative chemotherapy during radiation for some PET Small Japanese studies have focused on the use of combined
nonresponders to induction chemotherapy in 201 patients with chemoradiation to treat patients with T4 tumors with or without
esophageal or GEJ adenocarcinomas.231 Our data suggest that improve- fistulas.238–240 Although these studies have demonstrated long-term
ments in pCR rate and PFS are possible and that there is also a trend survival in a minority of patients, they have also been associated with
toward improved OS. the worsening or development of new fistulas and treatment-related
Based on this concept, the CALGB completed the 80803 trial, deaths in a significant proportion of patients. For example, in a phase
which enrolled patients with esophageal or GEJ adenocarcinomas. I/II evaluation, Nishimura and colleagues treated 28 patients with
Participants were randomized to receive induction chemotherapy esophageal squamous cell cancers with airway invasion with or without
with either carboplatin-paclitaxel or a modification of the FOLFOX6 fistulas.239 Patients received protracted infusion 5-FU–cisplatin, along
regimen (infusional 5-FU–leucovorin–oxaliplatin). Responses to induc- with split-dose radiation to a total dose of 60 Gy over 30 fractions.
tion chemotherapy were then adjudicated with an early PET scan This study reported an encouraging 2-year survival rate of 27% for
performed after induction chemotherapy. Although PET responders the patients with stage III (T4N+M0) disease.241 However, this was
continued with the same regimen during concurrent radiation, PET at the expense of significant toxicity, including the development or
nonresponders were changed to the alternative regimen with radiation worsening of fistulas in 18% of patients and treatment-related deaths
before surgery. The primary end point was to improve the pCR rate in 7% of patients.
in PET nonresponder patients by changing chemotherapy during Given the significant potential toxicities associated with up-front
combined chemoradiation. chemoradiation, one option is induction chemotherapy in an attempt
Results have very recently been presented in abstract form and to downstage the primary tumor and to potentially reduce the risk
indicate an improvement in the pCR rate in PET nonresponders who of fistula formation or progression with subsequent chemoradiation.
change chemotherapy to 17% PET non-responders to carboplatin/ We previously published a case report of a patient with a T4 tumor
paclitaxel and 19% FOLFOX (compared with a historical rate of 3% and tracheoesophageal fistula who underwent induction chemotherapy
in our retrospective analysis), meeting the primary end point of the and reassessment with PET scan and bronchoscopy.242 These revealed
study.232 It is also of significant interest that the pCR rate in the PET significant response in the tumor and complete healing of the fistula.
responders to induction FOLFOX who received 5-FU–oxaliplatin He then received chemoradiation and achieved a prolonged clinical
with radiation is 37.5%, whereas the pCR rate in the PET responders complete response.
to induction carboplatin-paclitaxel who received that regimen with
radiation is 12.5%. The number of patients in these two groups is CERVICAL ESOPHAGEAL CANCER
relatively small (64 and 56, respectively) and the study was not designed
to detect a difference in outcomes between the two induction regimens. Cervical esophageal cancer (almost always SCC) is a particular treatment
These results also seem to suggest the opposite conclusion of the challenge. The surgery required is often quite extensive, requiring a
UK NEOSCOPE study, in which 85 patients were randomized to total laryngopharyngoesophagectomy, an operation associated with
induction capecitabine-oxaliplatin, followed by concurrent chemoradia- significant dysfunction and morbidity. This has led to the use of
tion with carboplatin-paclitaxel versus capecitabine-oxaliplatin. Results definitive chemoradiation as the preferred alternative in many patients.
presented in abstract form revealed a pCR rate of 28% versus 12% Definitive radiation is fraught with technical difficulty in achieving
for the carboplatin-paclitaxel versus capecitabine-oxaliplatin group.233 an adequate dose, given the complexity of treating a structure in
Of course, the lack of PET assessment in the NEOSCOPE study and proximity to the spinal cord, the varying shape of the thorax and
the administration of induction capecitabine-oxaliplatin to all patients upper neck, and the bilateral lymph nodes at risk. The optimal dose
make it impossible to infer any conclusions. is unknown, with some radiation oncologists advocating for a higher
The results of the CALGB 80803 study are hypothesis generat- dose (approximately 70 Gy versus the 50.4 Gy that was used in the
ing, and clinical equipoise continues to exist about what the optimal RTOG 85-01 study200) than commonly used for treatment of head
chemotherapy regimen to combine with radiation is, especially and neck SCCs in or bordering the cervical esophagus. Although
because treatment was well tolerated in both arms. Survival data this seems reasonable given the similar biologic characteristics and
are pending. behavior that may underlie all aerodigestive SCCs and given that
tumors of the head and neck and cervical esophagus are in reality
MANAGEMENT OF TRACHEOESOPHAGEAL separated by only several centimeters, enthusiasm for such an approach
FISTULA is also dampened by the negative results of studies in esophageal
cancer in which the dose of radiation was augmented, namely
In the United States, esophageal cancer with airway invasion is fairly RTOG 94-05.211
uncommon. In a series from the Washington, DC Veterans Affairs Data for higher doses of radiation in this context are extremely
hospital from 1985 to 1998, 20% of patients with established esophageal limited; a retrospective analysis of 44 patients has suggested a benefit
cancer had airway involvement, with 9.4% having fistulous connec- for higher doses of radiation (68.1 versus 60 Gy),243 although there
tions.234 The vast majority (88%) of these patients had SCC, consistent is always a concern that short- and long-term toxicity could be higher.
with the location of these tumors in the cervical and proximal esophagus. In another series, 34 patients were treated with 5-FU–cisplatin che-
As the incidence of the SCC histology has steadily decreased, the motherapy and radiation to 61.2 Gy.244 The local control rate was

Downloaded for Tabanschii Nicolae (tabanschiirusuhan@gmail.com) at Nicolae Testemiceanu State Medical and Pharmaceutical University from
ClinicalKey.com by Elsevier on November 10, 2022. For personal use only. No other uses without permission. Copyright ©2022. Elsevier Inc. All rights
reserved.
1194 Part III: Specific Malignancies

88%, and the 5-year OS estimate was 55%. Acute toxicity was after brachytherapy.208 One potential benefit is that brachytherapy
acceptable, but two patients died from complications of strictures. can be used in patients previously treated with external beam radio-
In the absence of clear-cut data, treatment needs to be individualized therapy.250 Such a situation may arise in a patient with medically
and based on multidisciplinary evaluation and consensus. inoperable disease with local persistence or recurrence after definitive
chemoradiation.
The choice of palliative radiation and/or chemotherapy to treat
TREATMENT OF DYSPHAGIA dysphagia has to be carefully made based on patient factors, including
Chemotherapy and Radiation the burden of metastatic disease. For patients with significant metastatic
disease, an initial trial of chemotherapy is warranted, given its significant
Esophageal obstruction due to malignancy can lead to dysphagia that dysphagia improvement rates and the imperative to rapidly control
can be the source of very significant morbidity. Therefore the restoration systemic disease and to stabilize the patient’s performance status. On
of the ability to swallow is a key objective of both curative and palliative the other hand, initial treatment with palliative radiation is an option
treatments. The management of malignant esophageal obstruction for patients with small-volume metastatic disease and significant
should not compromise or interfere with curative therapy. Placement dysphagia. The endoscopic procedures discussed in the following section
of a percutaneous gastric or jejunostomy feeding tube and esophageal can also be incorporated into treatment decisions, hopefully offering
stents may be useful in temporizing symptoms, improving QoL, and patients several different opportunities and modalities to reduce this
optimizing the patient’s nutritional status while curative therapy is extremely debilitating symptom.
initiated. Other endoluminal treatments such as laser therapy, PDT,
or brachytherapy are generally not appropriate in situations in which Endoscopic Techniques
curative treatment is the goal. For patients who are not candidates
for curative therapy, the palliative treatment selected often depends The available endoscopic options for management of malignant
on the likelihood of success given the length and location of the lesion, dysphagia have shifted from the use of rigid funnel-shaped tubes to
life expectancy, the burden of metastatic disease, and the expertise of the use of expandable stents, laser fulguration, and PDT. Because
the treating team. there are no large studies directly comparing the efficacy of the available
Chemotherapy trials have reported significant palliation of patient endoscopic treatment modalities, the treatment modality chosen for
dysphagia with chemotherapy alone.64,92,114,119 Spiridonidis and col- each patient depends on the length, location, and angulation of the
leagues treated patients who had unresectable or metastatic esophageal stricture, the patient’s overall health status, the cost of the technique,
cancer with cisplatin and etoposide.261 Of 18 evaluable patients with the experience of the endoscopist, and the presence or absence of a
dysphagia, 89% experienced relief of dysphagia within 3 weeks of tracheoesophageal (TE) fistula.
initiating chemotherapy. Of 25 patients with dysphagia before therapy Because of the relatively high incidence of complications, rigid
with cisplatin and paclitaxel, 18 showed complete resolution (72%) funnel tubes have been replaced by self-expanding metallic stents
and 2 had partial resolution of dysphagia (8%) with chemotherapy. (SEMSs) and self-expanding plastic stents (SEPSs) that are compressible,
In a trial using a combination of weekly irinotecan and cisplatin, 20 are easier to place, and have fewer complications.251–253 The first-
patients had evaluable dysphagia; 14 patients (70%) had complete generation SEMSs were uncovered and were associated with rapid
resolution; and 4 patients (20%) experienced improvement of dysphagia, tumor ingrowth, resulting in stent obstruction. Up to 78% of patients
with improvement occurring after a median of one treatment cycle.119 who survived at least 4 months required repeat endoscopic intervention
In an older phase III trial, Ross and McKenna treated nearly 600 for palliation from tumor ingrowth. Newer SEMSs are covered with
patients with esophageal or gastric cancer with either the ECF regimen polyethylene, polyurethane, or silicone sheaths to delay or prevent
or mitomycin combined with cisplatin and 5-FU infusion.92 In patients ingrowth of tumor, substantially reducing the need for repeat endoscopic
evaluable for dysphagia, in both treatment arms 77% of patients procedures. SEMSs require less tumor dilation for insertion, expand
reported improvement in dysphagia symptoms. The rate of dysphagia to greater luminal diameter than rigid stents, and have thinner walls
relief reported in these trials correlated with antitumor RRs ranging to minimize erosion and hemorrhage. SEMSs may be “stacked” to
from 40% to 50%. manage tumor growth over the top of a previously placed expandable
There are relatively limited data on the ability of chemoradiation stent. Successful insertion of an SEMS is reported to occur in more
to improve dysphagia. Coia and colleagues reported a series of patients than 90% of patients. Initial complications are less frequent and less
treated in the 1980s; within 2 weeks after the start of chemoradiation, morbid than those reported with the use of rigid tubes.
45% had improvement in dysphagia, and by the completion of the In a series of 127 stent placements in 100 patients, researchers
sixth week, 83% had improvement.245 Overall, 88% had an improve- reported that no fatal complications occurred, 1.6% of patients had
ment in dysphagia, with a median time to maximum improvement severe pain necessitating removal, 3.1% of patients had inadequate
of 4 weeks. Histologic features and stage had no impact on the rate deployment, 0.8% of patients experienced perforation, 7.9% of patients
of palliation. In another series from the 1990s, 106 patients were had a food impaction, 11% of patients reported severe reflux, and
treated and 51% maintained improved swallowing until the time of 8.7% of patients developed stent migration requiring removal. Two
last follow-up or death.246 of 16 patients who underwent subsequent chemotherapy after stent
One of the few randomized studies to compare palliative radiation placement developed stent erosion through the esophageal wall. Studies
to 30 to 35 Gy alone versus chemoradiation with cisplatin is the suggest an increased rate of stent-related complications in patients
TROG 03.01/NCIC CTG ES2 study performed in the United who have undergone prior chemoradiation.252
Kingdom, Canada, Australia, and New Zealand.247 The abstract Patients will have severe reflux when the esophageal stent traverses
presentation shows that there was only a nonsignificant trend in favor the GEJ, and this may result in life-threatening aspiration. There is
of improvement in dysphagia at 9 weeks maintained at 13 weeks for an SEMS with an antireflux valve that may reduce risk of reflux;
chemoradiation versus radiation (47% versus 41%; P = .42), at the however, its success in clinical practice is muted, and widespread use
expense of increased toxicity. of the antireflux stent is not practiced.
Intraluminal brachytherapy is also an effective, albeit more Stents placed distally into the gastric lumen may be prone to
limited, method of achieving palliation of dysphagia in 40% to migration, and/or the distal end may cause gastric mucosal ulceration
90% of patients.248,249 In a randomized trial from the Dutch SIREC resulting in hemorrhage. This can often be managed endoscopically.
Study Group of stent versus one 12-Gy fraction of brachytherapy, SEMSs cannot be placed within 2 cm of the cricopharyngeal muscle
dysphagia, as measured by a variety of QoL scales, improved more because patients cannot tolerate the SEMS because of severe pain and
rapidly after stent placement; however, long-term relief was superior the sensation of a foreign body in the upper esophagus.

Downloaded for Tabanschii Nicolae (tabanschiirusuhan@gmail.com) at Nicolae Testemiceanu State Medical and Pharmaceutical University from
ClinicalKey.com by Elsevier on November 10, 2022. For personal use only. No other uses without permission. Copyright ©2022. Elsevier Inc. All rights
reserved.
Cancer of the Esophagus  •  CHAPTER 71 1195

There is no difference in efficacy or complication rates among any (monopolar or bipolar) with Nd:YAG laser techniques have shown
of the currently available SEMSs. SEPSs are also available and indicated comparable results in terms of morbidity, mortality, and relief of
for malignant dysphagia, but there are no comparative studies between dysphagia.
SEPSs and SEMSs. SEPSs can be better tolerated if placed near the PDT is a technology that has generated a great deal of interest,
cricopharyngeal muscle, and they are removable. However, the migration although it is not currently widely used (for reasons that will be
rate may be increased when compared with SEMSs. Despite these discussed shortly). This technique involves the administration of a
complications, placement of esophageal stents for malignant dysphagia chemical sensitizer, usually hematoporphyrin derivative or dihemato-
does improve swallowing in 88% to 100% of patients. porphyrin ethers, that accumulates preferentially in the target tumor.
A clear indication for placement of an SEMS is the presence of a The tumor is then exposed to a specific wavelength of low-power
TE fistula. As compared with other endoscopic options, placement laser light that activates the accumulated sensitizing chemical, resulting
of an SEMS is often a successful palliative measure. In small series, in tumor necrosis mediated by production of singlet oxygen radicals.
SEMSs were found effective in sealing a TE fistula, with low complica- The light penetrates only several millimeters, so this therapy is most
tion rates.250,254,255 Consideration should be given to placing a respiratory useful for palliative benefit because only superficial tumors would be
stent as well. treated in their entirety.
Other endoscopic options for palliation of malignant dysphagia Studies have documented that PDT is at least equally as effective
include neodymium:yttrium-aluminum-garnet (Nd:YAG) laser, as laser therapy in palliation in patients with esophageal cancer and
electrofulguration with monopolar or bipolar (BICAP) coagulation, is associated with a lower incidence of perforation.257,258 In a series of
and PDT.256–258 215 patients undergoing palliative PDT, the mean dysphagia-free
Electrofulguration techniques use either a monopolar probe or a interval was 66 days.259 Complications included perforation (2%),
BICAP device. The monopolar probe directly coagulates the obstructing stricture (2%), Candida esophagitis (2%), pleural effusions (4%), and
endoluminal tumor under endoscopic control. The BICAP tumor sunburn (6%). The procedure-related mortality rate was 1.8%.
probe is passed through the narrowed esophageal lumen over a Disadvantages of PDT relative to other methods of endoscopic pal-
guidewire. The mechanism of action of the BICAP probe includes liation, which have severely limited its use, include a prolonged period
dilation of the narrowed lumen, tumor coagulation, hyperthermia, of photosensitivity and significant cost (for both the laser and the
and tissue necrosis with delayed sloughing. Because the BICAP probe chemical agents).
coagulates blindly and circumferentially, it is not recommended for
noncircumferential lesions, because injury to the normal esophageal The complete reference list is available online at
mucosa may result in perforation. Comparisons of electrocoagulation ExpertConsult.com.

KEY REFERENCES
1. Cancer Genome Atlas Research Network, Analysis plus cetuximab in metastatic esophageal and 100. Bang YJ, Van Cutsem E, Feyereislova A, et al. Trastu-
Working Group: Asan University, BC Cancer Agency, gastroesophageal junction cancers. J Clin Oncol. zumab in combination with chemotherapy versus
et al. Integrated genomic characterization of oesopha- 2016;34(23):2736–2742. chemotherapy alone for treatment of HER2-positive
geal carcinoma. Nature. 2017;541(7636):169–175. 69. Van Cutsem E, Moiseyenko VM, Tjulandin S, advanced gastric or gastro-oesophageal junction
2. Cancer Genome Atlas Research Network, Analysis et al. Phase III study of docetaxel and cisplatin plus cancer (ToGA): a phase 3, open-label, randomised
Working Group: Dana-Farber Cancer Institute, fluorouracil compared with cisplatin and fluorouracil controlled trial. Lancet. 2010;376(9742):687–697.
Institute for Systems Biology, et al. Comprehensive as first-line therapy for advanced gastric cancer: a 125. Fuchs CS, Tomasek J, Yong CJ, et al. Ramucirumab
molecular characterization of gastric adenocarcinoma. report of the V325 Study Group. J Clin Oncol. monotherapy for previously treated advanced gastric
Nature. 2014;513(7517):202–209. 2006;24(31):4991–4997. or gastro-oesophageal junction adenocarcinoma
18. Lagergren J, Bergstrom R, Lindgren A, Nyren O. 71. Shah MA, Janjigian YY, Stoller R, et al. Ran- (REGARD): an international, randomised, mul-
Symptomatic gastroesophageal reflux as a risk factor domized multicenter phase II study of modified ticentre, placebo-controlled, phase 3 trial. Lancet.
for esophageal adenocarcinoma. N Engl J Med. docetaxel, cisplatin, and fluorouracil (DCF) versus 2014;383(9911):31–39.
1999;340(11):825–831. DCF plus growth factor support in patients with 126. Wilke H, Muro K, Van Cutsem E, et al. Ramuci-
20. Hvid-Jensen F, Pedersen L, Drewes AM, Sorensen metastatic gastric adenocarcinoma: a study of the rumab plus paclitaxel versus placebo plus paclitaxel
HT, Funch-Jensen P. Incidence of adenocarcinoma US Gastric Cancer Consortium. J Clin Oncol. in patients with previously treated advanced gastric
among patients with Barrett’s esophagus. N Engl J 2015;33(33):3874–3879. or gastro-oesophageal junction adenocarcinoma
Med. 2011;365(15):1375–1383. 72. Al-Batran SE, Hartmann JT, Hofheinz R, et al. (RAINBOW): a double-blind, randomised phase
23. Flamen P, Lerut A, Van Cutsem E, et al. Utility Biweekly fluorouracil, leucovorin, oxaliplatin, and 3 trial. Lancet Oncol. 2014;15(11):1224–1235.
of positron emission tomography for the staging docetaxel (FLOT) for patients with metastatic 128. Li J, Qin S, Xu J, et al. Randomized, double-blind,
of patients with potentially operable esophageal adenocarcinoma of the stomach or esophagogastric placebo-controlled phase III trial of apatinib in
carcinoma. J Clin Oncol. 2000;18(18):3202–3210. junction: a phase II trial of the Arbeitsgemeinschaft patients with chemotherapy-refractory advanced or
54. Wagner AD, Unverzagt S, Grothe W, et al. Che- Internistische Onkologie. Ann Oncol. 2008;19(11): metastatic adenocarcinoma of the stomach or gas-
motherapy for advanced gastric cancer. Cochrane 1882–1887. troesophageal junction. J Clin Oncol. 2016;34(13):
Database Syst Rev. 2010;(3):CD004064. 84. Kang JH, Lee SI, Lim DH, et al. Salvage chemo- 1448–1454.
55. Cunningham D, Starling N, Rao S, et  al. therapy for pretreated gastric cancer: a randomized 147. Kang Y, Satoh T, Ryu MH, et al. Nivolumab
Capecitabine and oxaliplatin for advanced esopha- phase III trial comparing chemotherapy plus best (ONO-4538/BMS-936558) as salvage treatment
gogastric cancer. N Engl J Med. 2008;358(1):36–46. supportive care with best supportive care alone. after second or later-line chemotherapy for advanced
57. Al-Batran SE, Hartmann JT, Probst S, et al. J Clin Oncol. 2012;30(13):1513–1518. gastric or gastro-esophageal junction cancer (AGC):
Phase III trial in metastatic gastroesophageal 85. Hironaka S, Ueda S, Yasui H, et al. Randomized, a double-blinded, randomized, phase III trial. J Clin
adenocarcinoma with fluorouracil, leucovorin open-label, phase III study comparing irinotecan Oncol. 2017;35:2 [abstr].
plus either oxaliplatin or cisplatin: a study of the with paclitaxel in patients with advanced gastric 148. Pardoll DM. The blockade of immune checkpoints in
Arbeitsgemeinschaft Internistische Onkologie. J Clin cancer without severe peritoneal metastasis after cancer immunotherapy. Nat Rev Cancer. 2012;12(4):
Oncol. 2008;26(9):1435–1442. failure of prior combination chemotherapy using 252–264.
63. van Hagen P, Hulshof MC, van Lanschot fluoropyrimidine plus platinum: WJOG 4007 Trial. 153. Muro K, Chung HC, Shankaran V, et  al.
JJ, et  al. Preoperative chemoradiotherapy for J Clin Oncol. 2013;31(35):4438–4444. Pembrolizumab for patients with PD-L1-positive
esophageal or junctional cancer. N Engl J Med. 2012; 86. Ford HE, Marshall A, Bridgewater JA, et al. advanced gastric cancer (KEYNOTE-012): a mul-
366(22):2074–2084. Docetaxel versus active symptom control for ticentre, open-label, phase 1b trial. Lancet Oncol.
68. Enzinger PC, Burtness BA, Niedzwiecki D, et al. refractory oesophagogastric adenocarcinoma 2016;17(6):717–726.
CALGB 80403 (Alliance)/E1206: a randomized (COUGAR-02): an open-label, phase 3 randomised 156. Hulscher JB, van Sandick JW, de Boer AG, et al.
phase II study of three chemotherapy regimens controlled trial. Lancet Oncol. 2014;15(1):78–86. Extended transthoracic resection compared with

Downloaded for Tabanschii Nicolae (tabanschiirusuhan@gmail.com) at Nicolae Testemiceanu State Medical and Pharmaceutical University from
ClinicalKey.com by Elsevier on November 10, 2022. For personal use only. No other uses without permission. Copyright ©2022. Elsevier Inc. All rights
reserved.
1196 Part III: Specific Malignancies

limited transhiatal resection for adenocarcinoma of the of the esophagus. N Engl J Med. 1992;326(24): using epirubicin, cisplatin, and infusional (CI) 5-FU
esophagus. N Engl J Med. 2002;347(21):1662–1669. 1593–1598. (ECF) before and after CI 5-FU and radiotherapy
161. Orringer MB, Marshall B, Chang AC, Lee J, Pickens 201. Cooper JS, Guo MD, Herskovic A, et al. Chemo- (CRT) compared with bolus 5-FU/LV before and
A, Lau CL. Two thousand transhiatal esophagecto- radiotherapy of locally advanced esophageal cancer: after CRT: intergroup trial CALGB 80101. J Clin
mies: changing trends, lessons learned. Ann Surg. long-term follow-up of a prospective randomized Oncol. 2011;29:4003 [abstr].
2007;246(3):363–372, discussion 372–374. trial (RTOG 85-01). Radiation Therapy Oncology 216. Verheij M, Jansen E, Cats A, et al. A multicenter
181. Hulscher JB, Tijssen JG, Obertop H, van Lanschot Group. JAMA. 1999;281(17):1623–1627. randomized phase III trial of neo-adjuvant chemo-
JJ. Transthoracic versus transhiatal resection for 207. Ajani JA, Xiao L, Roth JA, et al. A phase II ran- therapy followed by surgery and chemotherapy or by
carcinoma of the esophagus: a meta-analysis. Ann domized trial of induction chemotherapy versus no surgery and chemoradiotherapy in resectable gastric
Thorac Surg. 2001;72(1):306–313. induction chemotherapy followed by preoperative cancer: first results from the CRITICS study. J Clin
184. Cunningham D, Allum WH, Stenning SP, et al. chemoradiation in patients with esophageal cancer. Oncol. 2016;34:4000 [abstr].
Perioperative chemotherapy versus surgery alone for Ann Oncol. 2013;24(11):2844–2849. 217. Stahl M, Stuschke M, Lehmann N, et al. Chemo-
resectable gastroesophageal cancer. N Engl J Med. 210. Gaspar LE, Winter K, Kocha WI, Coia LR, radiation with and without surgery in patients with
2006;355(1):11–20. Herskovic A, Graham M. A phase I/II study of locally advanced squamous cell carcinoma of the
185. Ychou M, Boige V, Pignon JP, et al. Perioperative external beam radiation, brachytherapy, and con- esophagus. J Clin Oncol. 2005;23(10):2310–2317.
chemotherapy compared with surgery alone for current chemotherapy for patients with localized 218. Bedenne L, Michel P, Bouche O, et al. Chemoradia-
resectable gastroesophageal adenocarcinoma: an carcinoma of the esophagus (Radiation Therapy tion followed by surgery compared with chemoradia-
FNCLCC and FFCD multicenter phase III trial. Oncology Group Study 9207): final report. Cancer. tion alone in squamous cancer of the esophagus:
J Clin Oncol. 2011;29(13):1715–1721. 2000;88(5):988–995. FFCD 9102. J Clin Oncol. 2007;25(10):1160–
186. Alderson D, Langley R, Nankivell M, et al. Neo- 211. Minsky BD, Pajak TF, Ginsberg RJ, et al. INT 0123 1168.
adjuvant chemotherapy for resectable oesophageal (Radiation Therapy Oncology Group 94-05) phase 221. Marks JL, Hofstetter W, Correa AM, et al. Salvage
and junctional adenocarcinoma: results from the III trial of combined-modality therapy for esophageal esophagectomy after failed definitive chemoradiation
UK Medical Research Council randomised OEO5 cancer: high-dose versus standard-dose radiation for esophageal adenocarcinoma. Ann Thorac Surg.
trial (ISRCTN 01852072). J Clin Oncol. 2015;33: therapy. J Clin Oncol. 2002;20(5):1167–1174. 2012;94(4):1126–1132, discussion 1132–1133.
4002. 212. Mariette C, Dahan L, Mornex F, et al. Surgery alone 222. Markar S, Gronnier C, Duhamel A, et al. Salvage
187. Sakuramoto S, Sasako M, Yamaguchi T, et al. Adju- versus chemoradiotherapy followed by surgery for surgery after chemoradiotherapy in the management
vant chemotherapy for gastric cancer with S-1, an stage I and II esophageal cancer: final analysis of of esophageal cancer: is it a viable therapeutic option?
oral fluoropyrimidine. N Engl J Med. 2007;357(18): randomized controlled phase III trial FFCD 9901. J Clin Oncol. 2015;33(33):3866–3873.
1810–1820. J Clin Oncol. 2014;32(23):2416–2422. 226. Ott K, Weber WA, Lordick F, et al. Metabolic
188. Bang YJ, Kim YW, Yang HK, et al. Adjuvant 213. Stahl M, Walz MK, Stuschke M, et al. Phase III imaging predicts response, survival, and recurrence
capecitabine and oxaliplatin for gastric cancer after comparison of preoperative chemotherapy compared in adenocarcinomas of the esophagogastric junction.
D2 gastrectomy (CLASSIC): a phase 3 open-label, with chemoradiotherapy in patients with locally J Clin Oncol. 2006;24(29):4692–4698.
randomised controlled trial. Lancet. 2012;379(9813): advanced adenocarcinoma of the esophagogastric 228. Lordick F, Ott K, Krause BJ, et al. PET to assess
315–321. junction. J Clin Oncol. 2009;27(6):851–856. early metabolic response and to guide treatment
193. Sjoquist KM, Burmeister BH, Smithers BM, 214. Macdonald JS, Smalley SR, Benedetti J, et al. of adenocarcinoma of the oesophagogastric junc-
et al. Survival after neoadjuvant chemotherapy Chemoradiotherapy after surgery compared with tion: the MUNICON phase II trial. Lancet Oncol.
or chemoradiotherapy for resectable oesophageal surgery alone for adenocarcinoma of the stomach 2007;8(9):797–805.
carcinoma: an updated meta-analysis. Lancet Oncol. or gastroesophageal junction. N Engl J Med. 232. Goodman K, Niedzwiecki D, Hall N, et al. Initial
2011;12(7):681–692. 2001;345(10):725–730. results of CALGB 80803 (Alliance): a randomized
200. Herskovic A, Martz K, al-Sarraf M, et al. Com- 215. Fuchs C, Tepper J, Niedzwiecki D, et al. Post- phase II trial of PET scan-directed combined
bined chemotherapy and radiotherapy compared operative adjuvant chemoradiation for gastric or modality therapy for esophageal cancer. J Clin
with radiotherapy alone in patients with cancer gastroesophageal junction (GEJ) adenocarcinoma Oncol. 2017;35(4_suppl):1.

Downloaded for Tabanschii Nicolae (tabanschiirusuhan@gmail.com) at Nicolae Testemiceanu State Medical and Pharmaceutical University from
ClinicalKey.com by Elsevier on November 10, 2022. For personal use only. No other uses without permission. Copyright ©2022. Elsevier Inc. All rights
reserved.
Cancer of the Esophagus  •  CHAPTER 71 1196.e1
1196.e1

REFERENCES
1. Cancer Genome Atlas Research Network, Analysis 20. Hvid-Jensen F, Pedersen L, Drewes AM, Sorensen 38. Kantarjian H, Ajani JA, Karlin DA. cis-Diaminod-
Working Group: Asan University, BC Cancer Agency, HT, Funch-Jensen P. Incidence of adenocarcinoma ichloroplatinum (II) chemotherapy for advanced
et al. Integrated genomic characterization of oesopha- among patients with Barrett’s esophagus. N Engl J adenocarcinoma of the upper gastrointestinal tract.
geal carcinoma. Nature. 2017;541(7636):169–175. Med. 2011;365(15):1375–1383. Oncology. 1985;42(2):69–71.
2. Cancer Genome Atlas Research Network, Analysis 21. Desai TK, Krishnan K, Samala N, et al. The 39. Desai PB, Borges EJ, Vohra VG, Paymaster JC.
Working Group: Dana-Farber Cancer Institute, incidence of oesophageal adenocarcinoma in non- Carcinoma of the esophagus in India. Cancer.
Institute for Systems Biology, et al. Comprehensive dysplastic Barrett’s oesophagus: a meta-analysis. Gut. 1969;23(4):979–989.
molecular characterization of gastric adenocarcinoma. 2012;61(7):970–976. 40. Engstrom PF, Lavin PT, Klaassen DJ. Phase II evalu-
Nature. 2014;513(7517):202–209. 22. Kasugai T, Kobayashi S, Kuno N. Endoscopic ation of mitomycin and cisplatin in advanced esopha-
3. Rudiger Siewert J, Feith M, Werner M, Stein HJ. cytology of the esophagus, stomach and pancreas. geal carcinoma. Cancer Treat Rep. 1983;67(7–8):
Adenocarcinoma of the esophagogastric junction: Acta Cytol. 1978;22(5):327–330. 713–715.
results of surgical therapy based on anatomical/ 23. Flamen P, Lerut A, Van Cutsem E, et al. Utility 41. Whittington RM, Close HP. Clinical experience
topographic classification in 1,002 consecutive of positron emission tomography for the staging with mitomycin C (NSC-26980). Cancer Chemother
patients. Ann Surg. 2000;232(3):353–361. of patients with potentially operable esophageal Rep. 1970;54(3):195–198.
4. Enzinger PC, Mayer RJ. Esophageal cancer. N Engl carcinoma. J Clin Oncol. 2000;18(18):3202– 42. Hong YS, Song SY, Lee SI, et al. A phase II trial of
J Med. 2003;349(23):2241–2252. 3210. capecitabine in previously untreated patients with
5. Crew KD, Neugut AI. Epidemiology of upper gastro- 24. Wong R, Walker-Dilks C, Raifu A. Evidence-based advanced and/or metastatic gastric cancer. Ann Oncol.
intestinal malignancies. Semin Oncol. 2004;31(4): guideline recommendations on the use of positron 2004;15(9):1344–1347.
450–464. emission tomography imaging in oesophageal cancer. 43. Koizumi W, Saigenji K, Ujiie S, Terashima M, Sakata
6. Devesa SS, Fraumeni JF Jr. The rising incidence Clin Oncol (R Coll Radiol). 2012;24(2):86–104. Y, Taguchi T. A pilot phase II study of capecitabine
of gastric cardia cancer. J Natl Cancer Inst. 25. Botet JF, Lightdale CJ, Zauber AG, Gerdes H, in advanced or recurrent gastric cancer. Oncology.
1999;91(9):747–749. Urmacher C, Brennan MF. Preoperative staging of 2003;64(3):232–236.
7. Pohl H, Welch HG. The role of overdiagnosis and esophageal cancer: comparison of endoscopic US 44. Boku N, Yamamoto S, Fukuda H, et al. Fluorouracil
reclassification in the marked increase of esophageal and dynamic CT. Radiology. 1991;181(2):419–425. versus combination of irinotecan plus cisplatin versus
adenocarcinoma incidence. J Natl Cancer Inst. 26. Puli SR, Reddy JB, Bechtold ML, Antillon D, S-1 in metastatic gastric cancer: a randomised phase
2005;97(2):142–146. Ibdah JA, Antillon MR. Staging accuracy of 3 study. Lancet Oncol. 2009;10(11):1063–1069.
8. Buas MF, Vaughan TL. Epidemiology and risk factors esophageal cancer by endoscopic ultrasound: a meta- 45. Chollet P, Schoffski P, Weigang-Kohler K, et al. Phase
for gastroesophageal junction tumors: understanding analysis and systematic review. World J Gastroenterol. II trial with S-1 in chemotherapy-naive patients with
the rising incidence of this disease. Semin Radiat 2008;14(10):1479–1490. gastric cancer. A trial performed by the EORTC
Oncol. 2013;23(1):3–9. 27. Pfau PR, Perlman SB, Stanko P, et al. The role Early Clinical Studies Group (ECSG). Eur J Cancer.
9. Kong CY, Kroep S, Curtius K, et al. Exploring and clinical value of EUS in a multimodality 2003;39(9):1264–1270.
the recent trend in esophageal adenocarcinoma esophageal carcinoma staging program with CT 46. Sakata Y, Ohtsu A, Horikoshi N, Sugimachi K,
incidence and mortality using comparative simula- and positron emission tomography. Gastrointest Mitachi Y, Taguchi T. Late phase II study of novel
tion modeling. Cancer Epidemiol Biomarkers Prev. Endosc. 2007;65(3):377–384. oral fluoropyrimidine anticancer drug S-1 (1 M
2014;23(6):997–1006. 28. Luketich JD, Schauer P, Landreneau R, et al. tegafur-0.4 M gimestat-1 M otastat potassium)
10. Rubenstein JH, Taylor JB. Meta-analysis: the Minimally invasive surgical staging is superior to in advanced gastric cancer patients. Eur J Cancer.
association of oesophageal adenocarcinoma with endoscopic ultrasound in detecting lymph node 1998;34(11):1715–1720.
symptoms of gastro-oesophageal reflux. Aliment metastases in esophageal cancer. J Thorac Cardiovasc 47. Ilson D, Wadleigh R, Leichman L, Kelsen D.
Pharmacol Ther. 2010;32(10):1222–1227. Surg. 1997;114(5):817–821, discussion 821–823. Paclitaxel given by a weekly 1-h infusion in advanced
11. Hampel H, Abraham NS, El-Serag HB. Meta- 29. Kaushik N, Khalid A, Brody D, Luketich J, McGrath esophageal cancer. Ann Oncol. 2007;18(5):898–902.
analysis: obesity and the risk for gastroesophageal K. Endoscopic ultrasound compared with laparos- 48. Ajani JA, Ilson DH, Daugherty K, Pazdur R,
reflux disease and its complications. Ann Intern Med. copy for staging esophageal cancer. Ann Thorac Surg. Lynch PM, Kelsen DP. Activity of taxol in
2005;143(3):199–211. 2007;83(6):2000–2002. patients with squamous cell carcinoma and adeno-
12. Xia HH, Talley NJ. Helicobacter pylori infection, 30. Krasna MJ, Reed CE, Nedzwiecki D, et al. CALGB carcinoma of the esophagus. J Natl Cancer Inst.
reflux esophagitis, and atrophic gastritis: an unex- 9380: a prospective trial of the feasibility of thora- 1994;86(14):1086–1091.
plored triangle. Am J Gastroenterol. 1998;93(3):394– coscopy/laparoscopy in staging esophageal cancer. 49. Anderson SE, O’Reilly EM, Kelsen DP, Ilson DH.
400. Ann Thorac Surg. 2001;71(4):1073–1079. Phase II trial of 96-hour paclitaxel in previously
13. Ye W, Held M, Lagergren J, et al. Helicobacter pylori 31. Mehta K, Bianco V, Awais O, Luketich JD, treated patients with advanced esophageal cancer.
infection and gastric atrophy: risk of adenocarcinoma Pennathur A. Minimally invasive staging of Cancer Invest. 2003;21(4):512–516.
and squamous-cell carcinoma of the esophagus and esophageal cancer. Ann Cardiothorac Surg. 2017;6(2): 50. Einzig AI, Neuberg D, Remick SC, et al. Phase
adenocarcinoma of the gastric cardia. J Natl Cancer 110–118. II trial of docetaxel (Taxotere) in patients with
Inst. 2004;96(5):388–396. 32. Jin Z, Cheng Y, Gu W, et al. A multicenter, double- adenocarcinoma of the upper gastrointestinal
14. Bucchi D, Stracci F, Buonora N. Masanotti G. Human blinded validation study of methylation biomarkers tract previously untreated with cytotoxic chemo-
papillomavirus and gastrointestinal cancer: a review. for progression prediction in Barrett’s esophagus. therapy: the Eastern Cooperative Oncology Group
World J Gastroenterol. 2016;22(33):7415–7430. Cancer Res. 2009;69(10):4112–4115. (ECOG) results of protocol E1293. Med Oncol.
15. Steevens J, Schouten LJ, Goldbohm RA, van 33. Dunbar KB, Spechler SJ. The risk of lymph-node 1996;13(2):87–93.
den Brandt PA. Alcohol consumption, cigarette metastases in patients with high-grade dysplasia or 51. Heath EI, Urba S, Marshall J, Piantadosi S, Forastiere
smoking and risk of subtypes of oesophageal and intramucosal carcinoma in Barrett’s esophagus: a AA. Phase II trial of docetaxel chemotherapy in
gastric cancer: a prospective cohort study. Gut. systematic review. Am J Gastroenterol. 2012;107(6): patients with incurable adenocarcinoma of the
2010;59(1):39–48. 850–862, quiz 863. esophagus. Invest New Drugs. 2002;20(1):95–99.
16. Freedman ND, Abnet CC, Leitzmann MF, et al. 34. Ezdinli EZ, Gelber R, Desai DV, Falkson G, 52. Muro K, Hamaguchi T, Ohtsu A, et al. A phase
A prospective study of tobacco, alcohol, and the Moertel CG, Hahn RG. Chemotherapy of advanced II study of single-agent docetaxel in patients with
risk of esophageal and gastric cancer subtypes. Am esophageal carcinoma: Eastern Cooperative Oncology metastatic esophageal cancer. Ann Oncol. 2004;15(6):
J Epidemiol. 2007;165(12):1424–1433. Group experience. Cancer. 1980;46(10):2149–2153. 955–959.
17. Steevens J, Schouten LJ, Goldbohm RA, van den 35. Lokich JJ, Shea M, Chaffey J. Sequential infusional 53. Enzinger PC, Kulke MH, Clark JW, et al. A phase
Brandt PA. Vegetables and fruits consumption 5-fluorouracil followed by concomitant radiation II trial of irinotecan in patients with previously
and risk of esophageal and gastric cancer subtypes for tumors of the esophagus and gastroesophageal untreated advanced esophageal and gastric adeno-
in the Netherlands Cohort Study. Int J Cancer. junction. Cancer. 1987;60(3):275–279. carcinoma. Dig Dis Sci. 2005;50(12):2218–2223.
2011;129(11):2681–2693. 36. Davis S, Shanmugathasa M, Kessler W. cis- 54. Wagner AD, Unverzagt S, Grothe W, et al. Che-
18. Lagergren J, Bergstrom R, Lindgren A, Nyren O. Dichlorodiammineplatinum(II) in the treatment of motherapy for advanced gastric cancer. Cochrane
Symptomatic gastroesophageal reflux as a risk factor esophageal carcinoma. Cancer Treat Rep. 1980;64(4–5): Database Syst Rev. 2010;(3):CD004064.
for esophageal adenocarcinoma. N Engl J Med. 709–711. 55. Cunningham D, Starling N, Rao S, et  al.
1999;340(11):825–831. 37. Murthy SK, Prabhakaran PS, Chandrashekar M, Capecitabine and oxaliplatin for advanced esopha-
19. Ronkainen J, Aro P, Storskrubb T, et  al. Deshpande R, Doval DC, Gopinath KS. Neoadju- gogastric cancer. N Engl J Med. 2008;358(1):36–46.
Prevalence of Barrett’s esophagus in the general vant cis-DDP in esophageal cancers: an experience 56. Ajani JA, Rodriguez W, Bodoky G, et al. Multicenter
population: an endoscopic study. Gastroenterology. at a regional cancer centre, India. J Surg Oncol. phase III comparison of cisplatin/S-1 with cisplatin/
2005;129(6):1825–1831. 1990;45(3):173–176. infusional fluorouracil in advanced gastric or

Downloaded for Tabanschii Nicolae (tabanschiirusuhan@gmail.com) at Nicolae Testemiceanu State Medical and Pharmaceutical University from
ClinicalKey.com by Elsevier on November 10, 2022. For personal use only. No other uses without permission. Copyright ©2022. Elsevier Inc. All rights
reserved.
1196.e2 Part III: Specific Malignancies

gastroesophageal adenocarcinoma study: the FLAGS docetaxel, cisplatin, and fluorouracil (DCF) versus cancer without severe peritoneal metastasis after
trial. J Clin Oncol. 2010;28(9):1547–1553. DCF plus growth factor support in patients with failure of prior combination chemotherapy using
57. Al-Batran SE, Hartmann JT, Probst S, et al. metastatic gastric adenocarcinoma: a study of the fluoropyrimidine plus platinum: WJOG 4007 Trial.
Phase III trial in metastatic gastroesophageal US Gastric Cancer Consortium. J Clin Oncol. J Clin Oncol. 2013;31(35):4438–4444.
adenocarcinoma with fluorouracil, leucovorin 2015;33(33):3874–3879. 86. Ford HE, Marshall A, Bridgewater JA, et al.
plus either oxaliplatin or cisplatin: a study of the 72. Al-Batran SE, Hartmann JT, Hofheinz R, et al. Docetaxel versus active symptom control for refrac-
Arbeitsgemeinschaft Internistische Onkologie. J Clin Biweekly fluorouracil, leucovorin, oxaliplatin, and tory oesophagogastric adenocarcinoma (COUGAR-
Oncol. 2008;26(9):1435–1442. docetaxel (FLOT) for patients with metastatic 02): an open-label, phase 3 randomised controlled
58. Kang YK, Kang WK, Shin DB, et al. Capecitabine/ adenocarcinoma of the stomach or esophagogastric trial. Lancet Oncol. 2014;15(1):78–86.
cisplatin versus 5-fluorouracil/cisplatin as first-line junction: a phase II trial of the Arbeitsgemeinschaft 87. Dulak AM, Schumacher SE, van Lieshout J, et al.
therapy in patients with advanced gastric cancer: Internistische Onkologie. Ann Oncol. 2008;19(11): Gastrointestinal adenocarcinomas of the esophagus,
a randomised phase III noninferiority trial. Ann 1882–1887. stomach, and colon exhibit distinct patterns of
Oncol. 2009;20(4):666–673. 73. Al-Batran SE, Pauligk C, Homann N, et al. The genome instability and oncogenesis. Cancer Res.
59. Okines AF, Norman AR, McCloud P, Kang YK, feasibility of triple-drug chemotherapy combina- 2012;72(17):4383–4393.
Cunningham D. Meta-analysis of the REAL-2 tion in older adult patients with oesophagogastric 88. Deng N, Goh LK, Wang H, et al. A comprehensive
and ML17032 trials: evaluating capecitabine- cancer: a randomised trial of the Arbeitsgemeinschaft survey of genomic alterations in gastric cancer reveals
based combination chemotherapy and infused Internistische Onkologie (FLOT65+). Eur J Cancer. systematic patterns of molecular exclusivity and
5-fluorouracil-based combination chemotherapy 2013;49(4):835–842. co-occurrence among distinct therapeutic targets.
for the treatment of advanced oesophago-gastric 74. Gold P, Carter G, Livingston R. Phase II trial of Gut. 2012;61(5):673–684.
cancer. Ann Oncol. 2009;20(9):1529–1534. irinotecan (CPT-11) and mitomycin C (MMC) in 89. van Grieken NC, Aoyama T, Chambers PA, et al.
60. Einzig A, Kelsen DP, Cheng E, et al. Phase II trial the treatment of metastatic esophageal and gastric KRAS and BRAF mutations are rare and related
of carboplatin in patients with adenocarcinomas of cancers. Abstract 644. Proc Am Soc Clin Oncol. 2001; to DNA mismatch repair deficiency in gastric
the upper gastrointestinal tract. Cancer Treat Rep. 22. cancer from the East and the West: results from a
1985;69(12):1453–1454. 75. Ajani JA, Baker J, Pisters PW, et al. CPT-11 plus large international multicentre study. Br J Cancer.
61. El-Rayes BF, Shields A, Zalupski M, et al. A phase cisplatin in patients with advanced, untreated gastric 2013;108(7):1495–1501.
II study of carboplatin and paclitaxel in esophageal or gastroesophageal junction carcinoma: results of 90. Casalini P, Iorio MV, Galmozzi E, Menard S. Role
cancer. Ann Oncol. 2004;15(6):960–965. a phase II study. Cancer. 2002;94(3):641–646. of HER receptors family in development and dif-
62. Evans D, Miner T, Iannitti D, et al. Docetaxel, 76. Pozzo C, Barone C, Szanto J, et al. Irinotecan in ferentiation. J Cell Physiol. 2004;200(3):343–350.
capecitabine and carboplatin in metastatic esophago- combination with 5-fluorouracil and folinic acid 91. al-Kasspooles M, Moore JH, Orringer MB, Beer DG.
gastric cancer: a phase II study. Cancer Invest. 2007; or with cisplatin in patients with advanced gastric Amplification and over-expression of the EGFR and
25(6):445–448. or esophageal-gastric junction adenocarcinoma: erbB-2 genes in human esophageal adenocarcinomas.
63. van Hagen P, Hulshof MC, van Lanschot JJ, et al. results of a randomized phase II study. Ann Oncol. Int J Cancer. 1993;54(2):213–219.
Preoperative chemoradiotherapy for esophageal or 2004;15(12):1773–1781. 92. Ross JS, McKenna BJ. The HER-2/neu oncogene
junctional cancer. N Engl J Med. 2012;366(22): 77. Slater S, Shamash J, Wilson P, Gallagher CJ, Slevin in tumors of the gastrointestinal tract. Cancer Invest.
2074–2084. ML. Irinotecan, cisplatin and mitomycin in inoper- 2001;19(5):554–568.
64. Koizumi W, Narahara H, Hara T, et al. S-1 plus able gastro-oesophageal and pancreatic cancers—a 93. Chan DS, Twine CP, Lewis WG. Systematic review
cisplatin versus S-1 alone for first-line treatment of new active regimen. Br J Cancer. 2002;87(8): and meta-analysis of the influence of HER2 expres-
advanced gastric cancer (SPIRITS trial): a phase III 850–853. sion and amplification in operable oesophageal
trial. Lancet Oncol. 2008;9(3):215–221. 78. Ilson DH, Saltz L, Enzinger P, et al. Phase II trial of cancer. J Gastrointest Surg. 2012;16(10):1821–1829.
65. Vanhoefer U, Rougier P, Wilke H, et al. Final results weekly irinotecan plus cisplatin in advanced esopha- 94. Yoon HH, Shi Q, Sukov WR, et al. Association
of a randomized phase III trial of sequential high- geal cancer. J Clin Oncol. 1999;17(10):3270–3275. of HER2/ErbB2 expression and gene amplifica-
dose methotrexate, fluorouracil, and doxorubicin 79. Burtness B, Gibson M, Egleston B, et al. Phase tion with pathologic features and prognosis in
versus etoposide, leucovorin, and fluorouracil II trial of docetaxel-irinotecan combination in esophageal adenocarcinomas. Clin Cancer Res.
versus infusional fluorouracil and cisplatin in advanced esophageal cancer. Ann Oncol. 2009;20(7): 2012;18(2):546–554.
advanced gastric cancer: a trial of the European 1242–1248. 95. Brien TP, Odze RD, Sheehan CE, McKenna BJ, Ross
Organization for Research and Treatment of Cancer 80. Jatoi A, Tirona MT, Cha SS, et al. A phase II trial JS. HER-2/neu gene amplification by FISH predicts
Gastrointestinal Tract Cancer Cooperative Group. of docetaxel and CPT-11 in patients with metastatic poor survival in Barrett’s esophagus-associated
J Clin Oncol. 2000;18(14):2648–2657. adenocarcinoma of the esophagus, gastroesophageal adenocarcinoma. Hum Pathol. 2000;31(1):35–39.
66. Webb A, Cunningham D, Scarffe JH, et al. Ran- junction, and gastric cardia. Int J Gastrointest Cancer. 96. Chua TC, Merrett ND. Clinicopathologic factors
domized trial comparing epirubicin, cisplatin, and 2002;32(2–3):115–123. associated with HER2-positive gastric cancer and its
fluorouracil versus fluorouracil, doxorubicin, and 81. Enzinger PC, Ryan DP, Clark JW, et al. Weekly impact on survival outcomes—a systematic review.
methotrexate in advanced esophagogastric cancer. docetaxel, cisplatin, and irinotecan (TPC): results of Int J Cancer. 2012;130(12):2845–2856.
J Clin Oncol. 1997;15(1):261–267. a multicenter phase II trial in patients with metastatic 97. Janjigian YY, Werner D, Pauligk C, et al. Prognosis
67. Ross P, Nicolson M, Cunningham D, et al. Pro- esophagogastric cancer. Ann Oncol. 2009;20(3): of metastatic gastric and gastroesophageal junction
spective randomized trial comparing mitomycin, 475–480. cancer by HER2 status: a European and USA
cisplatin, and protracted venous-infusion fluorouracil 82. Dank M, Zaluski J, Barone C, et al. Randomized International collaborative analysis. Ann Oncol.
(PVI 5-FU) with epirubicin, cisplatin, and PVI 5-FU phase III study comparing irinotecan combined 2012;23(10):2656–2662.
in advanced esophagogastric cancer. J Clin Oncol. with 5-fluorouracil and folinic acid to cisplatin 98. Okines AF, Thompson LC, Cunningham D, et al.
2002;20(8):1996–2004. combined with 5-fluorouracil in chemotherapy Effect of HER2 on prognosis and benefit from peri-
68. Enzinger PC, Burtness BA, Niedzwiecki D, et al. naive patients with advanced adenocarcinoma of operative chemotherapy in early oesophago-gastric
CALGB 80403 (Alliance)/E1206: a randomized the stomach or esophagogastric junction. Ann Oncol. adenocarcinoma in the MAGIC trial. Ann Oncol.
phase II study of three chemotherapy regimens plus 2008;19(8):1450–1457. 2013;24(5):1253–1261.
cetuximab in metastatic esophageal and gastroesopha- 83. Guimbaud R, Louvet C, Ries P, et al. Prospective, 99. Terashima M, Kitada K, Ochiai A, et al. Impact
geal junction cancers. J Clin Oncol. 2016;34(23): randomized, multicenter, phase III study of fluoro- of expression of human epidermal growth factor
2736–2742. uracil, leucovorin, and irinotecan versus epirubicin, receptors EGFR and ERBB2 on survival in stage
69. Van Cutsem E, Moiseyenko VM, Tjulandin S, cisplatin, and capecitabine in advanced gastric II/III gastric cancer. Clin Cancer Res. 2012;18(21):
et al. Phase III study of docetaxel and cisplatin plus adenocarcinoma: a French intergroup (Federation 5992–6000.
fluorouracil compared with cisplatin and fluorouracil Francophone de Cancerologie Digestive, Federation 100. Bang YJ, Van Cutsem E, Feyereislova A, et al. Trastu-
as first-line therapy for advanced gastric cancer: a Nationale des Centres de Lutte Contre le Cancer, and zumab in combination with chemotherapy versus
report of the V325 Study Group. J Clin Oncol. Groupe Cooperateur Multidisciplinaire en Oncolo- chemotherapy alone for treatment of HER2-positive
2006;24(31):4991–4997. gie) study. J Clin Oncol. 2014;32(31):3520–3526. advanced gastric or gastro-oesophageal junction
70. Ajani JA, Moiseyenko VM, Tjulandin S, et al. 84. Kang JH, Lee SI, Lim DH, et al. Salvage chemo- cancer (ToGA): a phase 3, open-label, randomised
Clinical benefit with docetaxel plus fluorouracil and therapy for pretreated gastric cancer: a randomized controlled trial. Lancet. 2010;376(9742):687–
cisplatin compared with cisplatin and fluorouracil phase III trial comparing chemotherapy plus best 697.
in a phase III trial of advanced gastric or gastro- supportive care with best supportive care alone. J 101. Hecht JR, Bang YJ, Qin SK, et al. Lapatinib in
esophageal cancer adenocarcinoma: the V-325 Study Clin Oncol. 2012;30(13):1513–1518. combination with capecitabine plus oxaliplatin in
Group. J Clin Oncol. 2007;25(22):3205–3209. 85. Hironaka S, Ueda S, Yasui H, et al. Randomized, human epidermal growth factor receptor 2-positive
71. Shah MA, Janjigian YY, Stoller R, et al. Ran- open-label, phase III study comparing irinotecan advanced or metastatic gastric, esophageal, or gastro-
domized multicenter phase II study of modified with paclitaxel in patients with advanced gastric esophageal adenocarcinoma: TRIO-013/LOGiC—a

Downloaded for Tabanschii Nicolae (tabanschiirusuhan@gmail.com) at Nicolae Testemiceanu State Medical and Pharmaceutical University from
ClinicalKey.com by Elsevier on November 10, 2022. For personal use only. No other uses without permission. Copyright ©2022. Elsevier Inc. All rights
reserved.
Cancer of the Esophagus  •  CHAPTER 71 1196.e3
1196.e3

randomized phase III Trial. J Clin Oncol. 2016;34(5): gastric carcinoma. Cancer Lett. 2000;153(1–2): of fluoropyrimidine matters. Cancer Chemother
443–451. 7–12. Pharmacol. 2012;70(2):231–238.
102. Verma S, Miles D, Gianni L, et al. Trastuzumab 121. Ohtsu A, Shah MA, Van Cutsem E, et al. 137. Bijnsdorp IV, Kruyt FA, Fukushima M, Smid
emtansine for HER2-positive advanced breast cancer. Bevacizumab in combination with chemotherapy K, Gokoel S, Peters GJ. Molecular mechanism
N Engl J Med. 2012;367(19):1783–1791. as first-line therapy in advanced gastric cancer: a underlying the synergistic interaction between
103. Kang Y, Shah M, Ohtsu A, et al. A randomized, randomized, double-blind, placebo-controlled phase trifluorothymidine and the epidermal growth factor
open-label, multicenter, adaptive phase 2/3 study III study. J Clin Oncol. 2011;29(30):3968–3976. receptor inhibitor erlotinib in human colorectal
of trastuzumab emtansine (T-DM1) versus a taxane 122. Van Cutsem E, de Haas S, Kang YK, et al. Bevaci- cancer cell lines. Cancer Sci. 2010;101(2):440–447.
(TAX) in patients (pts) with previously treated zumab in combination with chemotherapy as first- 138. Kim HP, Yoon YK, Kim JW, et al. Lapatinib, a
HER2-positive locally advanced or metastatic line therapy in advanced gastric cancer: a biomarker dual EGFR and HER2 tyrosine kinase inhibitor,
gastric/gastroesophageal junction adenocarcinoma evaluation from the AVAGAST randomized phase downregulates thymidylate synthase by inhibiting
(LA/MGC/GEJC). J Clin Oncol. 2016;4(4_suppl):5. III trial. J Clin Oncol. 2012;30(17):2119–2127. the nuclear translocation of EGFR and HER2. PLoS
104. Franklin MC, Carey KD, Vajdos FF, Leahy DJ, de 123. Shah MA, Jhawer M, Ilson DH, et al. Phase II ONE. 2009;4(6):e5933.
Vos AM, Sliwkowski MX. Insights into ErbB signal- study of modified docetaxel, cisplatin, and fluoro- 139. Skvortsov S, Sarg B, Lindner H, et al. Cetuximab
ing from the structure of the ErbB2-pertuzumab uracil with bevacizumab in patients with metastatic inhibits thymidylate synthase in colorectal cells
complex. Cancer Cell. 2004;5(4):317–328. gastroesophageal adenocarcinoma. J Clin Oncol. expressing epidermal growth factor receptor.
105. Scheuer W, Friess T, Burtscher H, Bossenmaier B, 2011;29(7):868–874. Proteomics Clin Appl. 2008;2(6):908–914.
Endl J, Hasmann M. Strongly enhanced antitumor 124. Shah MA, Ramanathan RK, Ilson DH, et al. 140. Okines AF, Gonzalez de Castro D, Cunningham D,
activity of trastuzumab and pertuzumab combination Multicenter phase II study of irinotecan, cisplatin, et al. Biomarker analysis in oesophagogastric cancer:
treatment on HER2-positive human xenograft tumor and bevacizumab in patients with metastatic gastric results from the REAL3 and TransMAGIC trials.
models. Cancer Res. 2009;69(24):9330–9336. or gastroesophageal junction adenocarcinoma. J Clin Eur J Cancer. 2013;49(9):2116–2125.
106. Baselga J, Cortes J, Kim SB, et al. Pertuzumab plus Oncol. 2006;24(33):5201–5206. 141. Lordick F, Kang YK, Chung HC, et al. Capecitabine
trastuzumab plus docetaxel for metastatic breast 125. Fuchs CS, Tomasek J, Yong CJ, et al. Ramucirumab and cisplatin with or without cetuximab for patients
cancer. N Engl J Med. 2012;366(2):109–119. monotherapy for previously treated advanced gastric with previously untreated advanced gastric cancer
107. Kang YK, Rha SY, Tassone P, et al. A phase IIa or gastro-oesophageal junction adenocarcinoma (EXPAND): a randomised, open-label phase 3 trial.
dose-finding and safety study of first-line pertuzumab (REGARD): an international, randomised, mul- Lancet Oncol. 2013;14(6):490–499.
in combination with trastuzumab, capecitabine and ticentre, placebo-controlled, phase 3 trial. Lancet. 142. Lorenzen S, Schuster T, Porschen R, et al. Cetuximab
cisplatin in patients with HER2-positive advanced 2014;383(9911):31–39. plus cisplatin-5-fluorouracil versus cisplatin-
gastric cancer. Br J Cancer. 2014;111(4):660–666. 126. Wilke H, Muro K, Van Cutsem E, et al. Ramuci- 5-fluorouracil alone in first-line metastatic squamous
108. Folkman J. Angiogenesis and angiogenesis inhibition: rumab plus paclitaxel versus placebo plus paclitaxel cell carcinoma of the esophagus: a randomized phase
an overview. EXS. 1997;79:1–8. in patients with previously treated advanced gastric II study of the Arbeitsgemeinschaft Internistische
109. Ferrara N, Davis-Smyth T. The biology of vascular or gastro-oesophageal junction adenocarcinoma Onkologie. Ann Oncol. 2009;20(10):1667–1673.
endothelial growth factor. Endocr Rev. 1997;18(1): (RAINBOW): a double-blind, randomised phase 143. Crosby T, Hurt CN, Falk S, et al. Chemoradiotherapy
4–25. 3 trial. Lancet Oncol. 2014;15(11):1224–1235. with or without cetuximab in patients with oesopha-
110. Eskens FA, Verweij J. The clinical toxicity profile 127. Shitara K, Muro K, Shimada Y, et al. Subgroup geal cancer (SCOPE1): a multicentre, phase 2/3
of vascular endothelial growth factor (VEGF) and analyses of the safety and efficacy of ramucirumab randomised trial. Lancet Oncol. 2013;14(7):627–637.
vascular endothelial growth factor receptor (VEGFR) in Japanese and Western patients in RAINBOW: 144. Ilson D, Moughan J, Suntharalingam M, et al.
targeting angiogenesis inhibitors; a review. Eur J a randomized clinical trial in second-line treatment RTOG 0436: a phase III trial evaluating the addition
Cancer. 2006;42(18):3127–3139. of gastric cancer. Gastric Cancer. 2016;19(3):927– of cetuximab to paclitaxel, cisplatin, and radiation
111. Inoue K, Ozeki Y, Suganuma T, Sugiura Y, Tanaka 938. for patients with esophageal cancer treated without
S. Vascular endothelial growth factor expression 128. Li J, Qin S, Xu J, et al. Randomized, double-blind, surgery. J Clin Oncol. 2014;32:4007 [abstr].
in primary esophageal squamous cell carcinoma. placebo-controlled phase III trial of apatinib in 145. Hodi FS, O’Day SJ, McDermott DF, et al. Improved
Association with angiogenesis and tumor progression. patients with chemotherapy-refractory advanced survival with ipilimumab in patients with metastatic
Cancer. 1997;79(2):206–213. or metastatic adenocarcinoma of the stomach melanoma. N Engl J Med. 2010;363(8):711–723.
112. Kitadai Y, Haruma K, Tokutomi T, et al. Significance or gastroesophageal junction. J Clin Oncol. 146. Robert C, Thomas L, Bondarenko I, et al. Ipilimumab
of vessel count and vascular endothelial growth factor 2016;34(13):1448–1454. plus dacarbazine for previously untreated metastatic
in human esophageal carcinomas. Clin Cancer Res. 129. Pavlakis N, Sjoquist KM, Martin AJ, et al. Rego- melanoma. N Engl J Med. 2011;364(26):2517–2526.
1998;4(9):2195–2200. rafenib for the treatment of advanced gastric cancer 147. Kang Y, Satoh T, Ryu MH, et al. Nivolumab
113. Kleespies A, Guba M, Jauch KW, Bruns CJ. Vascular (INTEGRATE): a multinational placebo-controlled (ONO-4538/BMS-936558) as salvage treatment
endothelial growth factor in esophageal cancer. J phase II trial. J Clin Oncol. 2016;34(23):2728–2735. after second or later-line chemotherapy for advanced
Surg Oncol. 2004;87(2):95–104. 130. Yarden Y, Ullrich A. Growth factor receptor tyrosine gastric or gastro-esophageal junction cancer (AGC):
114. Shih CH, Ozawa S, Ando N, Ueda M, Kitajima kinases. Annu Rev Biochem. 1988;57:443–478. a double-blinded, randomized, phase III trial. J Clin
M. Vascular endothelial growth factor expression 131. Itakura Y, Sasano H, Shiga C, et al. Epidermal Oncol. 2017;35:2 [abstr].
predicts outcome and lymph node metastasis in growth factor receptor overexpression in esophageal 148. Pardoll DM. The blockade of immune check-
squamous cell carcinoma of the esophagus. Clin carcinoma. An immunohistochemical study cor- points in cancer immunotherapy. Nat Rev Cancer.
Cancer Res. 2000;6(3):1161–1168. related with clinicopathologic findings and DNA 2012;12(4):252–264.
115. Imdahl A, Bognar G, Schulte-Monting J, Schoffel amplification. Cancer. 1994;74(3):795–804. 149. Krummel MF, Allison JP. CD28 and CTLA-4
U, Farthmann EH, Ihling C. Predictive factors 132. Kitagawa Y, Ueda M, Ando N, Ozawa S, Shimizu have opposing effects on the response of T cells to
for response to neoadjuvant therapy in patients N, Kitajima M. Further evidence for prognostic stimulation. J Exp Med. 1995;182(2):459–465.
with oesophageal cancer. Eur J Cardiothorac Surg. significance of epidermal growth factor receptor gene 150. Freeman GJ, Long AJ, Iwai Y, et al. Engage-
2002;21(4):657–663. amplification in patients with esophageal squamous ment of the PD-1 immunoinhibitory receptor
116. Shimada H, Hoshino T, Okazumi S, et al. Expression cell carcinoma. Clin Cancer Res. 1996;2(5):909–914. by a novel B7 family member leads to negative
of angiogenic factors predicts response to chemora- 133. Gibault L, Metges JP, Conan-Charlet V, et al. Diffuse regulation of lymphocyte activation. J Exp Med.
diotherapy and prognosis of oesophageal squamous EGFR staining is associated with reduced overall 2000;192(7):1027–1034.
cell carcinoma. Br J Cancer. 2002;86(4):552– survival in locally advanced oesophageal squamous 151. Zou W, Chen L. Inhibitory B7-family molecules in
557. cell cancer. Br J Cancer. 2005;93(1):107–115. the tumour microenvironment. Nat Rev Immunol.
117. Mobius C, Stein HJ, Becker I, et al. The ‘angiogenic 134. Wilkinson NW, Black JD, Roukhadze E, et al. Epi- 2008;8(6):467–477.
switch’ in the progression from Barrett’s metaplasia dermal growth factor receptor expression correlates 152. Sharma P, Allison JP. The future of immune
to esophageal adenocarcinoma. Eur J Surg Oncol. with histologic grade in resected esophageal adeno- checkpoint therapy. Science. 2015;348(6230):56–61.
2003;29(10):890–894. carcinoma. J Gastrointest Surg. 2004;8(4):448–453. 153. Muro K, Chung HC, Shankaran V, et  al.
118. Vallbohmer D, Peters JH, Kuramochi H, et al. 135. Waddell T, Chau I, Cunningham D, et al. Epiru- Pembrolizumab for patients with PD-L1-positive
Molecular determinants in targeted therapy for bicin, oxaliplatin, and capecitabine with or without advanced gastric cancer (KEYNOTE-012): a mul-
esophageal adenocarcinoma. Arch Surg. 2006;141(5): panitumumab for patients with previously untreated ticentre, open-label, phase 1b trial. Lancet Oncol.
476–481, discussion 481–482. advanced oesophagogastric cancer (REAL3): a 2016;17(6):717–726.
119. Maeda K, Chung YS, Ogawa Y, et al. Prognostic randomised, open-label phase 3 trial. Lancet Oncol. 154. Ellis FH Jr, Heatley GJ, Krasna MJ, Williamson WA,
value of vascular endothelial growth factor expression 2013;14(6):481–489. Balogh K. Esophagogastrectomy for carcinoma of the
in gastric carcinoma. Cancer. 1996;77(5):858–863. 136. Ku GY, Haaland BA, de Lima Lopes G Jr. Cetux- esophagus and cardia: a comparison of findings and
120. Yoshikawa T, Tsuburaya A, Kobayashi O, et al. Plasma imab in the first-line treatment of K-ras wild-type results after standard resection in three consecutive
concentrations of VEGF and bFGF in patients with metastatic colorectal cancer: the choice and schedule eight-year intervals with improved staging criteria.

Downloaded for Tabanschii Nicolae (tabanschiirusuhan@gmail.com) at Nicolae Testemiceanu State Medical and Pharmaceutical University from
ClinicalKey.com by Elsevier on November 10, 2022. For personal use only. No other uses without permission. Copyright ©2022. Elsevier Inc. All rights
reserved.
1196.e4 Part III: Specific Malignancies

J Thorac Cardiovasc Surg. 1997;113(5):836–846, esophagectomies. Ann Surg. 2001;234(4):520–530, 192. Schuhmacher C, Gretschel S, Lordick F, et al.
discussion 846–848. discussion 530–531. Neoadjuvant chemotherapy compared with surgery
155. Muller JM, Erasmi H, Stelzner M, Zieren U, 174. Nishimaki T, Suzuki T, Suzuki S, Kuwabara S, alone for locally advanced cancer of the stomach
Pichlmaier H. Surgical therapy of oesophageal Hatakeyama K. Outcomes of extended radical and cardia: European Organisation for Research and
carcinoma. Br J Surg. 1990;77(8):845–857. esophagectomy for thoracic esophageal cancer. J Treatment of Cancer randomized trial 40954. J Clin
156. Hulscher JB, van Sandick JW, de Boer AG, et al. Am Coll Surg. 1998;186(3):306–312. Oncol. 2010;28(35):5210–5218.
Extended transthoracic resection compared with 175. Altorki NK, Girardi L, Skinner DB. En bloc 193. Sjoquist KM, Burmeister BH, Smithers BM,
limited transhiatal resection for adenocarcinoma esophagectomy improves survival for stage III et al. Survival after neoadjuvant chemotherapy
of the esophagus. N Engl J Med. 2002;347(21): esophageal cancer. J Thorac Cardiovasc Surg. or chemoradiotherapy for resectable oesophageal
1662–1669. 1997;114(6):948–955, discussion 955–956. carcinoma: an updated meta-analysis. Lancet Oncol.
157. Feith M, Stein HJ, Siewert JR. Adenocarcinoma of 176. Bumm R, Feussner H, Bartels H, et al. Radical 2011;12(7):681–692.
the esophagogastric junction: surgical therapy based transhiatal esophagectomy with two-field lymph- 194. Sasako M, Sakuramoto S, Katai H, et al. Five-year
on 1602 consecutive resected patients. Surg Oncol adenectomy and endodissection for distal esophageal outcomes of a randomized phase III trial comparing
Clin N Am. 2006;15(4):751–764. adenocarcinoma. World J Surg. 1997;21(8):822–831. adjuvant chemotherapy with S-1 versus surgery
158. Gockel I, Heckhoff S, Messow CM, Kneist W, 177. Luketich JD, Alvelo-Rivera M, Buenaventura PO, alone in stage II or III gastric cancer. J Clin Oncol.
Junginger T. Transhiatal and transthoracic resection et al. Minimally invasive esophagectomy: outcomes 2011;29(33):4387–4393.
in adenocarcinoma of the esophagus: does the opera- in 222 patients. Ann Surg. 2003;238(4):486–494, 195. Noh SH, Park SR, Yang HK, et al. Adjuvant
tive approach have an influence on the long-term discussion 494–495. capecitabine plus oxaliplatin for gastric cancer after
prognosis? World J Surg Oncol. 2005;3:40. 178. Bottger T, Terzic A, Muller M, Rodehorst A. Mini- D2 gastrectomy (CLASSIC): 5-year follow-up of an
159. Omloo JM, Lagarde SM, Hulscher JB, et al. Extended mally invasive transhiatal and transthoracic esopha- open-label, randomised phase 3 trial. Lancet Oncol.
transthoracic resection compared with limited tran- gectomy. Surg Endosc. 2007;21(10):1695–1700. 2014;15(12):1389–1396.
shiatal resection for adenocarcinoma of the mid/distal 179. Luketich JD, Pennathur A, Awais O, et al. Outcomes 196. Armanios M, Xu R, Forastiere AA, Haller DG,
esophagus: five-year survival of a randomized clinical after minimally invasive esophagectomy: review of Kugler JW, Benson AB 3rd. Adjuvant chemotherapy
trial. Ann Surg. 2007;246(6):992–1000, discussion over 1000 patients. Ann Surg. 2012;256(1):95– for resected adenocarcinoma of the esophagus,
1000–1001. 103. gastro-esophageal junction, and cardia: phase II
160. Rentz J, Bull D, Harpole D, et al. Transthoracic 180. Cuesta MA, Biere SS, van Berge Henegouwen MI, trial (E8296) of the Eastern Cooperative Oncology
versus transhiatal esophagectomy: a prospective van der Peet DL. Randomised trial, minimally inva- Group. J Clin Oncol. 2004;22(22):4495–4499.
study of 945 patients. J Thorac Cardiovasc Surg. sive oesophagectomy versus open oesophagectomy 197. Ando N, Iizuka T, Kakegawa T, et al. A randomized
2003;125(5):1114–1120. for patients with resectable oesophageal cancer. J trial of surgery with and without chemotherapy
161. Orringer MB, Marshall B, Chang AC, Lee J, Pickens Thorac Dis. 2012;4(5):462–464. for localized squamous carcinoma of the thoracic
A, Lau CL. Two thousand transhiatal esophagecto- 181. Hulscher JB, Tijssen JG, Obertop H, van Lanschot esophagus: the Japan Clinical Oncology Group Study.
mies: changing trends, lessons learned. Ann Surg. JJ. Transthoracic versus transhiatal resection for J Thorac Cardiovasc Surg. 1997;114(2):205–209.
2007;246(3):363–372, discussion 372–374. carcinoma of the esophagus: a meta-analysis. Ann 198. Ando N, Iizuka T, Ide H, et al. Surgery plus chemo-
162. Morgan MA, Lewis WG, Hopper AN, et al. Prospec- Thorac Surg. 2001;72(1):306–313. therapy compared with surgery alone for localized
tive comparison of transthoracic versus transhiatal 182. Gockel I, Sultanov FS, Domeyer M, Goenner U, squamous cell carcinoma of the thoracic esophagus: a
esophagectomy following neoadjuvant therapy Junginger T. Developments in esophageal surgery Japan Clinical Oncology Group Study–JCOG9204.
for esophageal cancer. Dis Esophagus. 2007;20(3): for adenocarcinoma: a comparison of two decades. J Clin Oncol. 2003;21(24):4592–4596.
225–231. BMC Cancer. 2007;7:114. 199. Ando N, Kato H, Igaki H, et al. A randomized
163. Raja S, Rice TW, Goldblum JR, et al. Esophageal 183. Steyerberg EW, Neville BA, Koppert LB, et al. trial comparing postoperative adjuvant chemotherapy
submucosa: the watershed for esophageal cancer. Surgical mortality in patients with esophageal cancer: with cisplatin and 5-fluorouracil versus preoperative
J Thorac Cardiovasc Surg. 2011;142(6):1403–1411, development and validation of a simple risk score. chemotherapy for localized advanced squamous cell
e1. J Clin Oncol. 2006;24(26):4277–4284. carcinoma of the thoracic esophagus (JCOG9907).
164. Lewis I. Carcinoma of the oesophagus. Radical resec- 184. Cunningham D, Allum WH, Stenning SP, et al. Ann Surg Oncol. 2012;19(1):68–74.
tion with oesophago-gastrostomy for a midthoracic Perioperative chemotherapy versus surgery alone for 200. Herskovic A, Martz K, al-Sarraf M, et al. Combined
growth by a right transpleural approach. Proc R Soc resectable gastroesophageal cancer. N Engl J Med. chemotherapy and radiotherapy compared with
Med. 1945;38(9):483–484. 2006;355(1):11–20. radiotherapy alone in patients with cancer of the
165. Mathisen DJ, Grillo HC, Wilkins EW Jr, Moncure 185. Ychou M, Boige V, Pignon JP, et al. Perioperative esophagus. N Engl J Med. 1992;326(24):1593–1598.
AC, Hilgenberg AD. Transthoracic esophagectomy: chemotherapy compared with surgery alone for 201. Cooper JS, Guo MD, Herskovic A, et al. Chemo-
a safe approach to carcinoma of the esophagus. Ann resectable gastroesophageal adenocarcinoma: an radiotherapy of locally advanced esophageal cancer:
Thorac Surg. 1988;45(2):137–143. FNCLCC and FFCD multicenter phase III trial. long-term follow-up of a prospective randomized
166. Nichols FC 3rd, Allen MS, Deschamps C, Cassivi J Clin Oncol. 2011;29(13):1715–1721. trial (RTOG 85-01). Radiation Therapy Oncology
SD, Pairolero PC. Ivor Lewis esophagogastrectomy. 186. Alderson D, Langley R, Nankivell M, et al. Neo- Group. JAMA. 1999;281(17):1623–1627.
Surg Clin North Am. 2005;85(3):583–592. adjuvant chemotherapy for resectable oesophageal 202. Bosset JF, Gignoux M, Triboulet JP, et al. Chemo-
167. Griffin SM, Shaw IH, Dresner SM. Early com- and junctional adenocarcinoma: results from the UK radiotherapy followed by surgery compared with
plications after Ivor Lewis subtotal esophagectomy Medical Research Council randomised OEO5 trial surgery alone in squamous-cell cancer of the
with two-field lymphadenectomy: risk factors and (ISRCTN 01852072). J Clin Oncol. 2015;33:4002. esophagus. N Engl J Med. 1997;337(3):161–167.
management. J Am Coll Surg. 2002;194(3):285–297. 187. Sakuramoto S, Sasako M, Yamaguchi T, et al. Adju- 203. Burmeister BH, Smithers BM, Gebski V, et al.
168. Heitmiller RF. Results of standard left thora- vant chemotherapy for gastric cancer with S-1, an Surgery alone versus chemoradiotherapy followed
coabdominal esophagogastrectomy. Semin Thorac oral fluoropyrimidine. N Engl J Med. 2007;357(18): by surgery for resectable cancer of the oesophagus: a
Cardiovasc Surg. 1992;4(4):314–319. 1810–1820. randomised controlled phase III trial. Lancet Oncol.
169. Shahian DM, Neptune WB, Ellis FH Jr, Watkins E 188. Bang YJ, Kim YW, Yang HK, et al. Adjuvant 2005;6(9):659–668.
Jr. Transthoracic versus extrathoracic esophagectomy: capecitabine and oxaliplatin for gastric cancer after 204. Urba SG, Orringer MB, Turrisi A, Iannettoni M,
mortality, morbidity, and long-term survival. Ann D2 gastrectomy (CLASSIC): a phase 3 open-label, Forastiere A, Strawderman M. Randomized trial of
Thorac Surg. 1986;41(3):237–246. randomised controlled trial. Lancet. 2012;379(9813): preoperative chemoradiation versus surgery alone in
170. McKeown KC. Total three-stage oesophagec- 315–321. patients with locoregional esophageal carcinoma.
tomy for cancer of the oesophagus. Br J Surg. 189. Shapiro J, van Lanschot JJ, Hulshof MC, et al. J Clin Oncol. 2001;19(2):305–313.
1976;63(4):259–262. Neoadjuvant chemoradiotherapy plus surgery versus 205. Walsh TN, Noonan N, Hollywood D, Kelly
171. Swanson SJ, Sugarbaker DJ. The three-hole surgery alone for oesophageal or junctional cancer A, Keeling N, Hennessy TP. A comparison of
esophagectomy. The Brigham and Women’s Hospital (CROSS): long-term results of a randomised con- multimodal therapy and surgery for esophageal
approach (modified McKeown technique). Chest trolled trial. Lancet Oncol. 2015;16(9):1090–1098. adenocarcinoma. N Engl J Med. 1996;335(7):462–
Surg Clin N Am. 2000;10(3):531–552. 190. Kelsen DP, Ginsberg R, Pajak TF, et al. Chemo- 467.
172. Hagen JA, Peters JH, DeMeester TR. Superiority therapy followed by surgery compared with surgery 206. Tepper J, Krasna MJ, Niedzwiecki D, et al. Phase
of extended en bloc esophagogastrectomy for car- alone for localized esophageal cancer. N Engl J Med. III trial of trimodality therapy with cisplatin,
cinoma of the lower esophagus and cardia. J Thorac 1998;339(27):1979–1984. fluorouracil, radiotherapy, and surgery compared
Cardiovasc Surg. 1993;106(5):850–858, discussion 191. Allum WH, Stenning SP, Bancewicz J, Clark PI, with surgery alone for esophageal cancer: CALGB
858–859. Langley RE. Long-term results of a randomized trial 9781. J Clin Oncol. 2008;26(7):1086–1092.
173. Hagen JA, DeMeester SR, Peters JH, Chand- of surgery with or without preoperative chemotherapy 207. Ajani JA, Xiao L, Roth JA, et al. A phase II ran-
rasoma P, DeMeester TR. Curative resection for in esophageal cancer. J Clin Oncol. 2009;27(30): domized trial of induction chemotherapy versus no
esophageal adenocarcinoma: analysis of 100 en bloc 5062–5067. induction chemotherapy followed by preoperative

Downloaded for Tabanschii Nicolae (tabanschiirusuhan@gmail.com) at Nicolae Testemiceanu State Medical and Pharmaceutical University from
ClinicalKey.com by Elsevier on November 10, 2022. For personal use only. No other uses without permission. Copyright ©2022. Elsevier Inc. All rights
reserved.
Cancer of the Esophagus  •  CHAPTER 71 1196.e5
1196.e5

chemoradiation in patients with esophageal cancer. for esophageal adenocarcinoma. Ann Thorac 239. Nishimura Y, Suzuki M, Nakamatsu K, Kanamori
Ann Oncol. 2013;24(11):2844–2849. Surg. 2012;94(4):1126–1132, discussion 1132– S, Yagyu Y, Shigeoka H. Prospective trial of concur-
208. Homs MY, Essink-Bot ML, Borsboom GJ, Steyerberg 1133. rent chemoradiotherapy with protracted infusion
EW, Siersema PD, Dutch SSG. Quality of life after 222. Markar S, Gronnier C, Duhamel A, et al. Salvage of 5-fluorouracil and cisplatin for T4 esophageal
palliative treatment for oesophageal carcinoma—a surgery after chemoradiotherapy in the management cancer with or without fistula. Int J Radiat Oncol
prospective comparison between stent placement of esophageal cancer: is it a viable therapeutic option? Biol Phys. 2002;53(1):134–139.
and single dose brachytherapy. Eur J Cancer. J Clin Oncol. 2015;33(33):3866–3873. 240. Ohtsu A, Boku N, Muro K, et al. Definitive
2004;40(12):1862–1871. 223. Swisher SG, Winter KA, Komaki RU, et al. A chemoradiotherapy for T4 and/or M1 lymph node
209. Okawa T, Dokiya T, Nishio M, Hishikawa Y, Morita Phase II study of a paclitaxel-based chemoradiation squamous cell carcinoma of the esophagus. J Clin
K. Multi-institutional randomized trial of external regimen with selective surgical salvage for resectable Oncol. 1999;17(9):2915–2921.
radiotherapy with and without intraluminal brachy- locoregionally advanced esophageal cancer: initial 241. Sobin LH, Fleming ID. TNM Classification of
therapy for esophageal cancer in Japan. Japanese reporting of RTOG 0246. Int J Radiat Oncol Biol Malignant Tumors, fifth edition (1997). Union
Society of Therapeutic Radiology and Oncology Phys. 2012;82(5):1967–1972. Internationale Contre le Cancer and the American
(JASTRO) Study Group. Int J Radiat Oncol Biol 224. Downey RJ, Akhurst T, Ilson D, et al. Whole body Joint Committee on Cancer. Cancer. 1997;80(9):
Phys. 1999;45(3):623–628. 18FDG-PET and the response of esophageal cancer 1803–1804.
210. Gaspar LE, Winter K, Kocha WI, Coia LR, to induction therapy: results of a prospective trial. 242. Ku GY, Goodman KA, Rusch VW, Ilson DH. Suc-
Herskovic A, Graham M. A phase I/II study of J Clin Oncol. 2003;21(3):428–432. cessful treatment of esophageal cancer with airway
external beam radiation, brachytherapy, and con- 225. Flamen P, Van Cutsem E, Lerut A, et al. Positron invasion with induction chemotherapy and concur-
current chemotherapy for patients with localized emission tomography for assessment of the response rent chemoradiotherapy. J Thorac Oncol. 2009;4(3):
carcinoma of the esophagus (Radiation Therapy to induction radiochemotherapy in locally advanced 432–434.
Oncology Group Study 9207): final report. Cancer. oesophageal cancer. Ann Oncol. 2002;13(3):361–368. 243. Zhu WG, Zhou K, Yu CH, Han JH, Li T, Chen XF.
2000;88(5):988–995. 226. Ott K, Weber WA, Lordick F, et al. Metabolic Efficacy analysis of simplified intensity-modulated
211. Minsky BD, Pajak TF, Ginsberg RJ, et al. INT 0123 imaging predicts response, survival, and recurrence radiotherapy with high or conventional dose and
(Radiation Therapy Oncology Group 94-05) phase in adenocarcinomas of the esophagogastric junction. concurrent chemotherapy for patients with neck and
III trial of combined-modality therapy for esophageal J Clin Oncol. 2006;24(29):4692–4698. upper thoracic esophageal carcinoma. Asian Pac J
cancer: high-dose versus standard-dose radiation 227. Weber WA, Ott K, Becker K, et al. Prediction of Cancer Prev. 2012;13(3):803–807.
therapy. J Clin Oncol. 2002;20(5):1167–1174. response to preoperative chemotherapy in adenocar- 244. Burmeister BH, Dickie G, Smithers BM, Hodge
212. Mariette C, Dahan L, Mornex F, et al. Surgery alone cinomas of the esophagogastric junction by metabolic R, Morton K. Thirty-four patients with carcinoma
versus chemoradiotherapy followed by surgery for imaging. J Clin Oncol. 2001;19(12):3058–3065. of the cervical esophagus treated with chemora-
stage I and II esophageal cancer: final analysis of 228. Lordick F, Ott K, Krause BJ, et al. PET to assess diation therapy. Arch Otolaryngol Head Neck Surg.
randomized controlled phase III trial FFCD 9901. early metabolic response and to guide treatment 2000;126(2):205–208.
J Clin Oncol. 2014;32(23):2416–2422. of adenocarcinoma of the oesophagogastric junc- 245. Coia LR, Soffen EM, Schultheiss TE, Martin EE,
213. Stahl M, Walz MK, Stuschke M, et al. Phase III tion: the MUNICON phase II trial. Lancet Oncol. Hanks GE. Swallowing function in patients with
comparison of preoperative chemotherapy compared 2007;8(9):797–805. esophageal cancer treated with concurrent radiation
with chemoradiotherapy in patients with locally 229. zum Buschenfelde CM, Herrmann K, Schuster T, and chemotherapy. Cancer. 1993;71(2):281–
advanced adenocarcinoma of the esophagogastric et al. (18)F-FDG PET-guided salvage neoadjuvant 286.
junction. J Clin Oncol. 2009;27(6):851–856. radiochemotherapy of adenocarcinoma of the 246. Harvey JA, Bessell JR, Beller E, et al. Chemo-
214. Macdonald JS, Smalley SR, Benedetti J, et al. Chemo- esophagogastric junction: the MUNICON II trial. radiation therapy is effective for the palliative
radiotherapy after surgery compared with surgery J Nucl Med. 2011;52(8):1189–1196. treatment of malignant dysphagia. Dis Esophagus.
alone for adenocarcinoma of the stomach or gastro- 230. Ilson DH, Bains M, Kelsen DP, et al. Phase I 2004;17(3):260–265.
esophageal junction. N Engl J Med. 2001;345(10): trial of escalating-dose irinotecan given weekly 247. Penniment M. Full report of the TROG 03.01,
725–730. with cisplatin and concurrent radiotherapy in NCIC CTG ES2 multinational phase III study in
215. Fuchs C, Tepper J, Niedzwiecki D, et al. Post- locally advanced esophageal cancer. J Clin Oncol. advanced esophageal cancer comparing palliation of
operative adjuvant chemoradiation for gastric or 2003;21(15):2926–2932. dysphagia and quality of life in patients treated with
gastroesophageal junction (GEJ) adenocarcinoma 231. Ku GY, Kriplani A, Janjigian YY, et al. Change in radiotherapy or chemoradiotherapy. J Clin Oncol.
using epirubicin, cisplatin, and infusional (CI) 5-FU chemotherapy during concurrent radiation followed 2015;33(3_suppl):6.
(ECF) before and after CI 5-FU and radiotherapy by surgery after a suboptimal positron emission 248. Sur RK, Singh DP, Sharma SC, et al. Radiation
(CRT) compared with bolus 5-FU/LV before and tomography response to induction chemotherapy therapy of esophageal cancer: role of high dose
after CRT: intergroup trial CALGB 80101. J Clin improves outcomes for locally advanced esophageal rate brachytherapy. Int J Radiat Oncol Biol Phys.
Oncol. 2011;29:4003 [abstr]. adenocarcinoma. Cancer. 2016;122(13):2083–2090. 1992;22(5):1043–1046.
216. Verheij M, Jansen E, Cats A, et al. A multicenter 232. Goodman K, Niedzwiecki D, Hall N, et al. Initial 249. Sur RK, Donde B, Levin VC, Mannell A. Fraction-
randomized phase III trial of neo-adjuvant chemo- results of CALGB 80803 (Alliance): a randomized ated high dose rate intraluminal brachytherapy in
therapy followed by surgery and chemotherapy or by phase II trial of PET scan-directed combined modal- palliation of advanced esophageal cancer. Int J Radiat
surgery and chemoradiotherapy in resectable gastric ity therapy for esophageal cancer. J Clin Oncol. 2017; Oncol Biol Phys. 1998;40(2):447–453.
cancer: first results from the CRITICS study. J Clin 35(4_suppl):1. 250. Gaspar LE, Nag S, Herskovic A, Mantravadi R,
Oncol. 2016;34:4000 [abstr]. 233. Mukherjee S, Hurt C, Gwynne S, et al. NEOSCOPE: Speiser B. American Brachytherapy Society (ABS)
217. Stahl M, Stuschke M, Lehmann N, et al. Chemo- a randomised phase II study of induction chemo- consensus guidelines for brachytherapy of esophageal
radiation with and without surgery in patients therapy followed by either oxaliplatin/capecitabine cancer. Clinical Research Committee, American
with locally advanced squamous cell carcinoma of (OXCAP) or carboplatin/paclitaxel (CarPac) based Brachytherapy Society, Philadelphia, PA. Int J Radiat
the esophagus. J Clin Oncol. 2005;23(10):2310– chemoradiation (CRT) as pre-operative regimen for Oncol Biol Phys. 1997;38(1):127–132.
2317. resectable oesophageal adenocarcinoma. Eur J Cancer. 251. Sgourakis G, Gockel I, Radtke A, et al. The use of
218. Bedenne L, Michel P, Bouche O, et al. Chemoradia- 2017;74:38–46. self-expanding stents in esophageal and gastroesopha-
tion followed by surgery compared with chemoradia- 234. Alexander EP, Trachiotis GD, Lipman TO, Wadleigh geal junction cancer palliation: a meta-analysis and
tion alone in squamous cancer of the esophagus: RG. Evolving management and outcome of esopha- meta-regression analysis of outcomes. Dig Dis Sci.
FFCD 9102. J Clin Oncol. 2007;25(10):1160–1168. geal cancer with airway involvement. Ann Thorac 2010;55(11):3018–3030.
219. Conroy T, Galais MP, Raoul JL, et al. Definitive Surg. 2001;71(5):1640–1644. 252. Iraha Y, Murayama S, Toita T, et al. Self-expandable
chemoradiotherapy with FOLFOX versus fluoro- 235. Altorki NK, Migliore M, Skinner DB. Esophageal metallic stent placement for patients with inoperable
uracil and cisplatin in patients with oesophageal carcinoma with airway invasion. Evolution and esophageal carcinoma: investigation of the influence
cancer (PRODIGE5/ACCORD17): final results choices of therapy. Chest. 1994;106(3):742–745. of prior radiotherapy and chemotherapy. Radiat Med.
of a randomised, phase 2/3 trial. Lancet Oncol. 236. Burt M, Diehl W, Martini N, et al. Malignant 2006;24(4):247–252.
2014;15(3):305–314. esophagorespiratory fistula: management options and 253. Hirdes MM, Siersema PD, Vleggaar FP. A new fully
220. Vincent J, Mariette C, Pezet D, et al. Early surgery survival. Ann Thorac Surg. 1991;52(6):1222–1228, covered metal stent for the treatment of benign and
for failure after chemoradiation in operable thoracic discussion 1228–1229. malignant dysphagia: a prospective follow-up study.
oesophageal cancer. Analysis of the non-randomised 237. Reed MF, Mathisen DJ. Tracheoesophageal fistula. Gastrointest Endosc. 2012;75(4):712–718.
patients in FFCD 9102 phase III trial: chemora- Chest Surg Clin N Am. 2003;13(2):271–289. 254. Tomaselli F, Maier A, Sankin O, Woltsche M, Pinter
diation followed by surgery versus chemoradiation 238. Koike R, Nishimura Y, Nakamatsu K, Kanamori H, Smolle-Juttner FM. Successful endoscopical
alone. Eur J Cancer. 2015;51(13):1683–1693. S, Shibata T. Concurrent chemoradiotherapy for sealing of malignant esophageotracheal fistulae by
221. Marks JL, Hofstetter W, Correa AM, et al. Salvage esophageal cancer with malignant fistula. Int J Radiat using a covered self-expandable stenting system. Eur
esophagectomy after failed definitive chemoradiation Oncol Biol Phys. 2008;70(5):1418–1422. J Cardiothorac Surg. 2001;20(4):734–738.

Downloaded for Tabanschii Nicolae (tabanschiirusuhan@gmail.com) at Nicolae Testemiceanu State Medical and Pharmaceutical University from
ClinicalKey.com by Elsevier on November 10, 2022. For personal use only. No other uses without permission. Copyright ©2022. Elsevier Inc. All rights
reserved.
1196.e6 Part III: Specific Malignancies

255. Abadal JM, Echenagusia A, Simo G, Camunez F. cancer: light dosimetry and randomized comparison cancer: experience in 215 patients. Ann Thorac Surg.
Treatment of malignant esophagorespiratory fistulas with Nd:YAG laser therapy. Gastroenterology. 2003;76(5):1687–1692, discussion 1692–1693.
with covered stents. Abdom Imaging. 2001;26(6): 1995;109(1):63–72. 260. Medical Research Council Oesophageal Cancer
565–569. 258. Lightdale CJ, Heier SK, Marcon NE, et al. Pho- Working Group. Surgical resection with or
256. Jensen DM, Machicado G, Randall G, Tung LA, todynamic therapy with porfimer sodium versus without preoperative chemotherapy in oesophageal
English-Zych S. Comparison of low-power YAG laser thermal ablation therapy with Nd:YAG laser for cancer: a randomised controlled trial. Lancet.
and BICAP tumor probe for palliation of esophageal palliation of esophageal cancer: a multicenter ran- 2002;359(9319):1727–1733.
cancer strictures. Gastroenterology. 1988;94(6): domized trial. Gastrointest Endosc. 1995;42(6):507– 261. Spiridonidis CH, Laufman LR, Jones JJ, et al.
1263–1270. 512. A phase II evaluation of high dose cisplatin and
257. Heier SK, Rothman KA, Heier LM, Rosenthal WS. 259. Litle VR, Luketich JD, Christie NA, et al. Pho- etoposide in patients with advanced esophageal
Photodynamic therapy for obstructing esophageal todynamic therapy as palliation for esophageal adenocarcinoma. Cancer. 1996;78:2070.

Downloaded for Tabanschii Nicolae (tabanschiirusuhan@gmail.com) at Nicolae Testemiceanu State Medical and Pharmaceutical University from
ClinicalKey.com by Elsevier on November 10, 2022. For personal use only. No other uses without permission. Copyright ©2022. Elsevier Inc. All rights
reserved.

You might also like