ArticlePDF AvailableLiterature Review

Gastroesophageal Junction Adenocarcinoma: Is There an Optimal Management?

Authors:

Abstract

The incidence of gastroesophageal junction (GEJ) adenocarcinomas has been rising over the past few decades, creating a need for effective therapeutic strategies. Treatment of locally advanced GEJ tumors, in particular, present a unique challenge because these tumors have generally been approached as either esophageal or gastric cancers, and thus optimal preoperative management remains uncertain. Both neoadjuvant chemoradiation and perioperative chemotherapy have been widely adopted in standard practice; however, it is unclear which approach offers the optimal outcome for the fit patient capable of receiving any planned strategy. In this review, we debate the management of locally advanced GEJ adenocarcinoma, and discuss areas of ongoing investigation which may provide more effective and individualized treatment of patients with GEJ cancers.
GASTROINTESTINAL (NONCOLORECTAL) CANCER
Gastroesophageal Junction Adenocarcinoma: Is
There an Optimal Management?
Daniel Lin, MD, MSc
1
; Uqba Khan, MD
2
; Thorsten O. Goetze, MD
3
; Natalie Reizine, MD
4
; Karyn A. Goodman, MD, MS
5
;
Manish A. Shah, MD
2
; Daniel V. Catenacci, MD
4
; Salah-Eddin Al-Batran, MD
3
; and James A. Posey, MD
1
overview
The incidence of gastroesophageal junction (GEJ) adenocarcinomas has been rising over the past few
decades, creating a need for effective therapeutic strategies. Treatment of locally advanced GEJ tumors, in
particular, present a unique challenge because these tumors have generally been approached as either
esophageal or gastric cancers, and thus optimal preoperative management remains uncertain. Both neo-
adjuvant chemoradiation and perioperative chemotherapy have been widely adopted in standard practice;
however, it is unclear which approach offers the optimal outcome for the t patient capable of receiving any
planned strategy. In this review, we debate the management of locally advanced GEJ adenocarcinoma, and
discuss areas of ongoing investigation which may provide more effective and individualized treatment of
patients with GEJ cancers.
With combined estimates of more than 1.6 million
new cases and more than 1 million related deaths in
2018, cancers of the stomach and esophagus (gas-
troesophageal cancer), which include esophageal
squamous cell carcinoma (SCC), proximal esoph-
agogastric junction (EGJ) adenocarcinomas (esoph-
ageal and gastric cardia adenocarcinomas), and distal
gastric adenocarcinoma, remain important worldwide
public health concern.
1
The incidence of EGJ ade-
nocarcinoma has notably risen in Western countries,
and population analyses in the United States have
reported a nearly 2.5-fold increased incidence since
the 1970s.
2,3
There has been a lack of a single standard system of
classication for EGJ tumors. The Siewert classica-
tion, which establishes three types based on the en-
doscopic location of the epicenter of the tumor and its
relation with the EGJ, allows a tailored surgical ap-
proach and consistency in reporting results associated
with therapeutic interventions.
4,5
Type I EGJ tumors are
located up to 5 cm proximal to the anatomic EGJ, type
II lesions span the anatomic EGJ and have their epi-
center up to 2 cm below the EGJ, and type III cardia
tumors extend up to 5 cm into the stomach.
6
In recent
guidelines from the American Joint Committee on
Cancer 8th edition, EGJ tumors with epicenter in the
distal esophagus or less than 2 cm into the proximal
stomach (Siewert types I and II) based on surgical
resection specimens are included under the esopha-
geal cancer staging classication.
7
However, it is
sometimes difcult for endoscopists to accurately
identify the epicenter of the tumor because the
intraluminal mass often spans across these articial
type I-III boundaries. Moreover, recent molecular
proling studies
8,9
clearly differentiated esophageal
SCC from type I lesions, in which SCCs resembled
SCCs of other organs more than they did type I lesions,
whereas type I lesions predominantly resembled the
chromosomal unstable subtype (CIN) found in the
majority of type II/III lesions and approximately 50% of
distal gastric adenocarcinoma. Therefore, although
there are surgical distinctions between Siewert types
I-III, biologically these are considered quite molecularly
similar.
8,9
Histologically, the large majority of these
tumors are adenocarcinomas.
Surgery remains the primary curative modality for EGJ
adenocarcinomas. However, overall prognosis of this
disease remains poor because of distant and locore-
gional recurrence of disease, with 5-year survival rates
averaging around 30% with surgery alone.
10
Conse-
quently, neoadjuvant or perioperative multimodality
strategies incorporating chemotherapy, radiation, or
the combination of both, have emerged over the past
few decades, with the goal of eradicating occult
micrometastatic disease and improving both surgical
and survival outcomes.
As noted, given its anatomic location joining esophagus
and noncardia stomach, EGJ adenocarcinomas have
often been grouped together with either esophageal
or gastric cancers in large, prospective, randomized
clinical trials, and have rarely been evaluated as
a distinct entity, resulting in trials that have variable
inclusion criteria. The difculty with clearly de-
marcating type I, II, and III EGJ tumors also affects this
Author affiliations
and support
information (if
applicable) appear
at the end of this
article.
Accepted on May 17,
2019 and published
at ascopubs.org on
May 17, 2019:
DOI https://doi.org/
10.1200/EDBK_
236827
e88 2019 ASCO EDUCATIONAL BOOK | asco.org/edbook
Downloaded from ascopubs.org by UNIVERSITY CHICAGO on May 19, 2019 from 128.135.207.229
Copyright © 2019 American Society of Clinical Oncology. All rights reserved.
variability. Several pivotal trials have established paradigms of
multimodality management with perioperative chemotherapy
or preoperative chemoradiation of locally advanced gastro-
esophageal cancers. In particular, the Dutch CROSS trial,
which included mostly type I esophageal adenocarcinoma
and some type II EGJ cancers along with esophageal SCC
(25%), demonstrated superior R0 (microscopically margin-
negative) surgical resection rates and long-term survival
outcomes with neoadjuvant radiotherapy combined with
concurrent weekly carboplatin and paclitaxel, compared with
surgery alone, and established this regimen as a standard of
care.
11
More recently, however, combination perioperative
chemotherapy strategies have been investigated. The FLOT4-
AIO trial, which included a population of EGJ cancers (56%)
along with distal gastric adenocarcinoma (44%), compared
a docetaxel-based triplet chemotherapy FLOT (5-uorouracil
[5-FU], oxaliplatin, docetaxel) with anthracycline-based triplet
epirubicin, cisplatin, and 5-FU or capecitabine (ECF/ECX),
and demonstrated superior survival outcomes with FLOT.
12,13
Given the lack of denitive studies directly comparing che-
motherapy alone or in combination with radiation for locally
advanced EGJ adenocarcinoma, it is difcult to discern which
may be the most effective preoperative strategy for these
patients. This article will therefore debate the optimal man-
agement of locally advanced EGJ adenocarcinoma and will
provide aspects to consider foror againsteither strategy.
We conclude with a discussion of ongoing clinical studies that
may advance and further individualize future strategies for
EGJ cancers.
FOR PRE-/PERIOPERATIVE CHEMOTHERAPY VERSUS
NEOADJUVANT CHEMORADIOTHERAPY
The prognosis of patients with locally advanced EGJ ade-
nocarcinoma is poor with surgery alone. Although the two
approaches of perioperative chemotherapy and neo-
adjuvant chemoradiation have each improved survival
compared with surgery alone and have been widely though
heterogeneously adopted in Western countries, there have
been more consistent data in favor of perioperative che-
motherapy. In this review, we summarize the data in support
of perioperative chemotherapy, and compare this treatment
paradigm with neoadjuvant chemoradiation.
First, all available trials enrolled patients with different pri-
mary tumor locations. Although the trials of perioperative
chemotherapy often enrolled patients with distal gastric and
proximal EGJ adenocarcinomas (type I-III lesions), the
neoadjuvant chemoradiation trials, including the pivotal
CROSS trial,
11
enrolled patients with esophageal SCC as well
as patients with type I and II lesions.
The rst study to predominantly focus on EGJ adenocar-
cinomas was the French perioperative FNCLCC/FFCD 9703
perioperative chemotherapy trial.
14
The majority (75%) of
the 224 patients enrolled to the study had EGJ adenocar-
cinoma, (type I 11%, type II/III 64%, and distal gastric
adenocarcinoma 24%). Patients were randomized to re-
ceive two to three cycles of cisplatin/5-FU followed by
surgery, or to surgery alone. After surgery, patients in the
chemotherapy arm who had no evidence of progressive
disease while on the preoperative therapy received addi-
tional cycles of adjuvant chemotherapy. Treatment in the
perioperative chemotherapy arm resulted in signicantly
improved overall survival (OS; 5-year OS rate 38% vs. 24%;
hazard ratio [HR] 0.69; 95% CI, 0.500.95; p = .02). The
EGJ type II/III tumors derived the highest benet from
perioperative chemotherapy (HR 0.57; 95% CI, 0.390.83).
Perioperative chemotherapy also improved progression-free
survival and the rate of R0 resection.
The MAGIC trial
15
was a landmark phase III trial of gastric
and EGJ adenocarcinoma conducted in the United King-
dom. Focused on distal gastric cancer, the study enrolled
503 patients with clinical stage II or III adenocarcinoma of
the stomach (including type III cardia) (75%), GEJ type II
(11.5%), and lower esophagus type I (14.5%) who were
treated with either three cycles of ECF pre- and postsurgery,
or with surgery alone. Similar to the FNCLCC trial, the
perioperative chemotherapy arm demonstrated a signicant
improvement in progression-free survival and OS (5-year OS
rates 36% vs. 23%; HR 0.75; 95% CI, 0.600.93; p = .009)
compared with surgery alone. All anatomic categories (type
I, type II GEJ, and type III/stomach) derived benet. Despite
fewer patients (11%) in the MAGIC trial having GEJ cancers,
this subgroup seemed to derive the greatest benet from
chemotherapy (HR for death in the EGJ subgroup was
0.49; 95% CI, 0.280.88), similar to the FNCLCC/FFCD
9703 study. Neither the MAGIC nor the FNCLCC/FFCD
9703 trials found that preoperative chemotherapy increased
PRACTICAL APPLICATIONS
Understand the different effective treatment
options for the t patient presenting with po-
tentially operable GEJ adenocarcinoma.
Learn the key discussion point when discussing
management of GEJ adenocarcinoma.
Learn of novel approaches to management in
GEJ adenocarcinoma.
Learn about the factors that inuence the use of
perioperative chemotherapy versus neoadjuvant
chemoradiation for this patient population.
Recognize the need for multidisciplinary dis-
cussion when planning a treatment course for
patients diagnosed with T3N1 adenocarcinoma
of the gastroesphageal junction.
Optimal Management of Gastroesophageal Junction Adenocarcinoma
2019 ASCO EDUCATIONAL BOOK | asco.org/edbook e89
Downloaded from ascopubs.org by UNIVERSITY CHICAGO on May 19, 2019 from 128.135.207.229
Copyright © 2019 American Society of Clinical Oncology. All rights reserved.
morbidity, mortality, or hospitalization time compared with
surgery alone.
The argument for a perioperative chemotherapy approach for
EGJ adenocarcinoma has been further strengthened by the
results of the recent FLOT4-AIO trial.
12,13
In this German trial,
a total of 716 patients with locally advanced, resectable gastric
or EGJ adenocarcinoma were randomly assigned to either the
MAGIC regimen consisting of three preoperative and three
postoperative 3-week cycles of ECF/ECX) or four preoperative
and four postoperative cycles of FLOT. Most patients (56%)
had EGJ adenocarcinoma (24% with Siewert type 1, 32% with
type II/III and 15% with Barrett), whereas 44% had distal
gastric adenocarcinoma. Approximately 80% of patients had
a T3 or greater tumors, with 8% having T4 lesions (whereas
the CROSS study, discussed next, had no T4 lesions). FLOT4-
AIO enrolled 80% of patients having node-positive disease
(the CROSS trial had only 65% with node-positive disease).
The primary study outcome, median OS, was signicantly
improved with FLOT; OS of 35 months was noted in the ECF/
ECXgroupandcomparedwith50monthsintheFLOTgroup
(HR 0.77; 95% CI, 0.630.94; p = .012). Median disease-free
survival was also signicantly improved: 18 months in the ECF/ ECX
group compared with 30 months in the FLOT group (HR
0.75; 95% CI, 0.620.91; p = .004). FLOT was associated
with signicant tumor down-sizing (ypT1 tumors, 25%
with FLOT vs. 15% with ECF/ECX; p = .0008) and increased
rate of R0 resection (85% vs. 78%; p = .0162) as assessed
in the intention-to-treat (ITT) population. As shown in the
phase II part of the FLOT4-AIO trial, FLOT was associated
with signicantly higher proportions of patients achieving
pathologic complete regression (pCR) grade 1a (16%; 95%
CI, 1023 vs. 6%; 95% CI, 311; p = .02), which was more
pronounced in the intestinal histology subgroup (23%) com-
pared with the mixed (0%) and diffuse (3%) types. This is
notable because most EGJ tumors are intestinal CIN type,
whereas most diffuse/mixed types are distal gastric cancers not
EGJ tumors molecularly considered (genomically stable).
8,9
Importantly, the quality of surgerya major point of
criticism generally raised in trials involving multimodality
therapywas excellent in the FLOT4-AIO trial. Nearly all
patients (98%) received at least the protocol-recommended
D2- or two-eld resections; the median number of lymph
nodes removed was 25. The benets observed in the
FLOT4-AIO trial were consistent across the subgroups,
specically with regard to age, anatomic site, histology, and
clinical stage. When assessed for interaction statistically, the
corresponding HRs of death were 0.76 for EGJ and 0.77 for
the stomach, respectively, p = .94. Furthermore, despite the
challenges associated with patients completing planned
post-operative therapy (50%), the FLOT4-AIO trial is the rst
study to show an improvement in OS compared with another
effective bimodal perioperative strategy as opposed to a
surgery alone comparator arm.
In contrast to the studies involving perioperative chemo-
therapy, support for neoadjuvant chemoradiation is mostly
derived from smaller clinical trials with heterogeneity in the
patients. The use of neoadjuvant chemoradiation for EGJ
cancers is based primarily on the CROSS trial.
11
The CROSS
study was the only randomized study evaluating the benet
of neoadjuvant chemoradiotherapy to show a survival
benet from chemoradiation in esophageal and GEJ ade-
nocarcinomas. In this study, 366 patients with esophageal
SCC or esophageal adenocarcinoma type I (73.2%) or EGJ
type II (24%) were randomly assigned to chemotherapy
with weekly paclitaxel and carboplatin combined with radi-
ation therapy followed by surgery or to surgery alone. Al-
though the median OS in the ITT population clearly favored
neoadjuvant chemoradiation over surgery alone (HR 0.66;
95% CI, 0.4950.871; p = .003), the adenocarcinoma sub-
group, the predominant group accounting for 75% of accrual,
beneted to a lesser extent (HR 0.741; 95% CI, 0.5361.
024; p = .07) than the SCC group (HR 0.422; 95% CI,
0.2260.788; p = .007). Moreover, after multivariate
analysis adjusting for known clinicopathologic prognostic
variables, the survival benet in the adenocarcinoma group
was not statistically important. The specic results differ-
entiating the type I and type II adenocarcinoma subgroups
were not presented or published and are therefore un-
known. Further examination of the subgroups of the CROSS
study shows that the relative benet from neoadjuvant
chemoradiation was observed in patients with node-
negative disease (HR 0.422; 95% CI, 0.2390.747; p =
.003), whereas patients with node-positive disease, and
thus signs of an initial systemic spread, did not benet from
neoadjuvant chemoradiation (HR 0.807; 95% CI, 0.576
1.130; p = .21).
16
With only 364 analyzed patients, of which
75% had EGJ adenocarcinoma, the CROSS study is a rel-
atively small and heterogenous study, and its results need
conrmation by other randomized trials. Moreover, it is often
overlooked that the CROSS study compared neoadjuvant
chemoradiation versus surgery only, whereas the FLOT4-
AIO trial compared FLOT against another effective regimen
known to improve survival. The results with FLOT should
therefore be considered and advancement to those achieved
with ECF/ECX.
The only randomized trial to compare neoadjuvant che-
moradiation with chemotherapy alone in EGJ cancer was
the German study by Stahl and colleagues (POET).
17
In this
small study, which closed early because of poor accrual,
126 patients with adenocarcinoma of the esophagus, GEJ,
or gastric cardia were randomly assigned to chemotherapy
followed by surgery or to chemotherapy followed by che-
moradiation and then surgery. There was a statistical trend
toward improved 3-year OS rates (p = .07) in the chemo-
radiation arm. Nonetheless, the POET study was formally
a negative trial and had several substantial limitations,
Lin et al
e90 2019 ASCO EDUCATIONAL BOOK | asco.org/edbook
Downloaded from ascopubs.org by UNIVERSITY CHICAGO on May 19, 2019 from 128.135.207.229
Copyright © 2019 American Society of Clinical Oncology. All rights reserved.
including that the analysis was not performed on an ITT basis
because only 119 of the 126 patients were analyzed, with
most of the dropouts occurring in the chemotherapy arm.
There are other arguments against the routine incorporation
of radiotherapy in EGJ adenocarcinoma patients with locally
advanced disease. Despite the notion that radiotherapy is
requiredto optimize R0 resection, pCR, and as a con-
sequence, local recurrence rates, evidence has suggested
otherwise. The R0 resection rate in the ITT population of the
CROSS study in the EGJ adenocarcinoma subgroup was not
explicitly reported but was approximately 82%85%, and
therefore is similar (84%) to that of the FLOT4-AIO ITT
analysis. The pCR rate (grade 1a by Becker criteria) was
23% in the CROSS study, and similar to the 23% in the
intestinal subtype (of which EGJ is mostly comprised, as
discussed previously, from TCGA proling) of the FLOT4-
AIO study. Moreover, a pCR achieved with chemotherapy
alone implies systemic benet perhaps not derived from
chemoradiotherapycarboplatin/paclitaxel as in CROSS.
Ultimately, the response observed with measurable disease
is believed to impact micrometastatic more so than a dou-
blet with radiation locally. Finally, the predominant pattern
of recurrence of EGJ adenocarcinoma is systemic or distant.
As demonstrated in the FFCD 9703 study,
14
only 24% of
patients had local recurrence (12% with local only and 12%
with both local and distant recurrence), and the remainder
with distant disease alone. Similarly, the CROSS trial re-
ported 22% of patients with localized recurrence (9.6% with
local only, and 12.4% with both local and distant re-
currence). Furthermore, the FLOT4-AIO trial reported
18.5% of patients with local and distant recurrence (12% with
local only, and 6.5% with both local and distant recurrence).
Although cross-trial comparisons should be approached
with caution, the addition of radiotherapy does not seem to
decrease local recurrence to a greater extent.
In each of the contemporary studies discussed, the majority
of patients had clinical T3 and node-positive tumors, which
remains the most common presentation of EGJ adeno-
carcinoma. Thus, until further direct comparative evidence
of perioperative chemotherapy versus neoadjuvant che-
moradiation is available for these patients, given the strong
evidence demonstrating efcacy with consideration of
treatment toxicities, cost, and convenience compared with
chemoradiotherapy, FLOT is emerging as a sound choice for
t patients with locally advanced EGJ adenocarcinoma.
OPPOSED TO PRE-/PERIOPERATIVE FOR
NEOADJUVANT CHEMORADIOTHERAPY
The rationale for trimodality therapy, which incorporates
chemotherapy and radiation before surgery, stems from
the unique anatomic features of the EGJ.As it spans from the
distal esophagus across the diaphragmatic crus into the
most proximal aspect of the stomach, the cardia, the EGJ
remains anatomically distinct from the more distal stomach.
Because the esophagus is not covered by a serosal lining
and is in close proximity to many organs and structures,
spread by direct extension is frequent. In addition, the rich
submucosal lymphatic network of the esophagus and GEJ
leads to a high risk of lymph node involvement.
18
Indeed,
because the proximal EGJ tumors (Siewerts types I and II,
specically) behave like esophageal cancer, both in terms of
nodal patterns of spread and in terms of survival, the
American Joint Committee on Cancer thus classies EGJ
tumors with esophageal cancer. Anatomically, these tumors
are most similar to distal esophageal adenocarcinoma.
Biologically, distal esophageal tumors are identical to EGJ
tumors and to the CIN subtype of gastric cancer (which is
predominantly seen in the proximal stomach). This is one
reason to consider preoperative chemoradiation for EGJ
tumors: they should be approached similarly to distal
esophageal tumors given their similar behavior.
Furthermore, the data supporting trimodality therapy are
based on the high rate of local failure after esophagectomy
alone, as well as concerns about achieving curative negative
margin resection. For example, the results for patients on
the surgery control arm of the INT-0113 trial,
19
which
randomized patients to preoperative cisplatin/5-FU versus
surgery alone, revealed a 31% local failure rate in patients
who underwent an R0 resection. When all patients entered
on study were examined, an overall 61% rate of failure in
controlling local disease was observed if patients failed to
achieve R0 resection or developed progressive disease on
preoperative therapy are included.
19
In addition, INT-0113
failed to demonstrate differences in R0 resection rate,
treatment mortality, or OS (3-year OS rates of 23% vs. 26%;
p = .53) between patients receiving preoperative chemo-
therapy versus surgery alone, and the failure patterns were
similar.
19
The R0 resection rate was 62% in the preoperative
chemotherapy arm and 59% in the surgery alone group.
Several studies and meta-analyses have demonstrated an
association with positive circumferential resection margin
status and OS.
20,21
Thus, the potential for locoregional
failure and positive circumferential resection margin after
surgery has engendered interest in the use of radia-
tion therapy as part of combined modality surgery-based
therapy.
The landmark study that established the benet of pre-
operative chemoradiotherapy was the Dutch CROSS trial.
11
As noted previously, this study randomized 366 patients
with SCC or adenocarcinoma of the esophagus or EGJ to
treatment with (1) preoperative carboplatin at an area under
the curve of 2 mg per minutes per milliliter and paclitaxel
50 mg/m
2
once weekly for 5 weeks, and concurrent ra-
diotherapy (1.8 Gy daily to 41.4 Gy in 23 fractions, followed
by transthoracic esophagectomy or transhiatal esoph-
agectomy for GEJ cancers), or (2) immediate surgery. All
Optimal Management of Gastroesophageal Junction Adenocarcinoma
2019 ASCO EDUCATIONAL BOOK | asco.org/edbook e91
Downloaded from ascopubs.org by UNIVERSITY CHICAGO on May 19, 2019 from 128.135.207.229
Copyright © 2019 American Society of Clinical Oncology. All rights reserved.
patients were staged by endoscopic ultrasound and com-
puted tomography scan and were required to have either
node-positive or T2-3 disease. The majority of patients
treated had adenocarcinoma (75%) and most tumors in-
volved the distal one-third of the esophagus (58%). The
majority of patients were node positive (65%), and slightly
more patients on the chemoradiotherapy arm had T3 tu-
mors (84%) compared with the surgery-alone arm (78%).
At a median follow up of 45 months, the trial showed
a signicant survival benet for chemoradiotherapy added
to surgery, with median survival increasing from 24 to
49 months (HR, 0.0657; p = .003), and improvement in
2- and 5-year OS (67% and 47% vs. 50% and 34%; HR
0.665). The rate of R0 resection was signicantly improved
on the chemoradiotherapy arm (92% compared with 69%;
p,.001). Pathologic complete responses (pCR) were seen
in 23% of adenocarcinomas. Therapy was well tolerated
with grade 3 or 4 hematologic toxicity seen in 7% patients,
and grade 3 or 4 nonhematologic toxicity seen in less than
13%. There was no difference in either operative morbidity
or mortality, and mortality was below 4% in each arm, which
is considered consistent and appropriate with modern-day
surgical outcomes in high-volume centers. Long-term follow
up indicated a maintained survival benet for preoperative
therapy over surgery alone for both SCC and adenocarci-
noma.
16
At a median follow up of 84.1 months in surviving
patients, benets were maintained for both adenocarci-
noma and SCC patients. For adenocarcinoma, median OS
was increased from 27.1 months to 43.2 months (HR 0.73).
Preoperative chemoradiotherapy signicantly reduced the
risk of both locoregional disease progression (38% with
surgery alone reduced to 22% with chemoradiotherapy)
and distant disease progression (48%39% with chemo-
radiotherapy). Based on these results, a new standard of
care for adenocarcinomas of the distal esophagus or GEJ
was established.
As mentioned earlier, the POET trial
17
was a major phase III
trial designed to compare preoperative chemotherapy and
chemoradiation versus chemotherapy alone, followed by
surgery. This trial only enrolled patients with GEJ cancer, but
was terminated prematurely because of poor accrual, with
anal study enrollment of just 119 eligible patients. Eligible
patients were randomly assigned to receive (1) 2.5 cycles of
a 6-week schedule of weekly 5-FU 2 g/m
2
, 24-hour infusion
and leucovorin 500 mg/m
2
, 2-hour infusion plus biweekly
cisplatin 50 mg/m
2
, or (2) two cycles of the same regimen,
followed by 3 weeks of radiotherapy given in 15 fractions at
a dose of 2 Gy combined with cisplatin 50 mg/m
2
on days 1
and 8 and etoposide 80 mg/m
2
on days 3 to 5. The pCR rate
(16% vs. 2%, p = .03) and lymph nodenegative status
(64% vs. 37%, p = .01) were signicantly higher in the
chemoradiotherapy arm. Although the R0 resection rate in
operated patients was increased from 79% to 88% by
chemoradiotherapy, the ITT probability of complete re-
section was not different between treatment arms. With
a median follow-up time of 45.6 months, 3-year survival
trended toward improvement in the chemoradiotherapy
group (47.4%) compared with the chemotherapy group
(27.7%; p = .07), and freedom from local tumor progression
also favored the chemoradiotherapy group (76.5% vs. 59%;
p = .06). Despite the small sample size, OS also showed
a trend in favor of preoperative chemoradiation (HR 0.65;
95% CI, 0.421.01, p = .055), suggesting that chemo-
radiation may be superior to chemotherapy alone.
Meta-analyses of neoadjuvant chemoradiotherapy versus
surgery-alone trials attempted to synthesize these data, and
one study concluded that chemoradiotherapy was associ-
ated with a 13% absolute benet in survival at 2 years. The
investigators also compared trials of neoadjuvant chemo-
therapy (without radiation) versus surgery alone and found
a smaller survival benet of 7% at 2 years.
22
A more recent
update with more than 4,100 patients further demonstrated
a survival benet of neoadjuvant chemoradiotherapy versus
chemotherapy alone.
23
Given the risk of systemic disease in patients with GEJ and
the importance of better tailoring therapy to tumor biology,
an emerging option for patients with esophageal and EGJ
adenocarcinomas is induction chemotherapy followed by
chemoradiotherapy. The Alliance/CALGB 80803 is a ran-
domized phase II study that was designed to evaluate the
early assessment of chemotherapy responsiveness by
positron emission tomography (PET) imaging to direct
further therapy in patients with esophageal cancer to im-
prove their response to therapy as demonstrated by pCR
rates.
24
A total of 257 patients with resectable T3/4 or node-
positive esophageal adenocarcinoma underwent a baseline
PET/CT scan to determine maximum standardized uptake
value (SUVmax), followed by random assignment to either
modied FOLFOX6 or carboplatin/paclitaxel. After approx-
imately 6 weeks of induction therapy, patients had a repeat
PET scan. If SUVmax decreased by more than 35% from
baseline, as was the case in 50%57% of evaluable pa-
tients, these individuals were deemed PET responders,
continued on their assigned chemotherapy, and received
concurrent radiotherapy. If the decrease in SUVmax did not
meet this threshold, which occurred in 30%38% of
evaluable patients, these patients were considered PET
nonresponders and crossed over to receive the alternative
chemotherapy regimen in conjunction with radiotherapy.
Surgery occurred approximately 6 weeks after the com-
pletion of chemoradiotherapy. The study met its primary
endpoint of increasing pCR rates from 5% to 20% in PET
nonresponders by changing chemotherapy during radio-
therapy. The survival outcomes were also very encouraging.
Although the median survival was higher in the group of
patients that responded to induction chemotherapy than in
Lin et al
e92 2019 ASCO EDUCATIONAL BOOK | asco.org/edbook
Downloaded from ascopubs.org by UNIVERSITY CHICAGO on May 19, 2019 from 128.135.207.229
Copyright © 2019 American Society of Clinical Oncology. All rights reserved.
the group that did not respond (46 months for responders
and 27 months for nonresponders) this was not statistically
signicant (p = .09), suggesting that the change from an
ineffective chemotherapy regimen may have improved
outcomes in the group that was initially not responding.
Moreover, the median survival of 50 months and 4-year OS
of 52% in the patients who had FOLFOX induction che-
motherapy and were responders demonstrates that by
enriching the population for good responders using PET as
a biomarker results in more favorable outcomes in this high-
risk population.
25
A follow up to this study is to evaluate the
use of FLOT induction chemotherapy followed by a CROSS
chemoradiotherapy regimen to address both the high risk of
systemic and local recurrence. By combining these ap-
proaches in the neoadjuvant setting, there is also the benet
that patients will receive all planned therapy, rather than in
the postoperative setting, where many patients are not able
to receive adjuvant chemotherapy.
DISCUSSION
Although several well-conducted randomized clinical trials
over the past couple of decades have clearly demonstrated
that multidisciplinary management with neoadjuvant che-
moradiation or perioperative chemotherapy for locally ad-
vanced EGJ cancers improve both surgical and survival
outcomes, the optimal strategy remains in question. How-
ever, several ongoing clinical trials may further elucidate
a preferred approach. The multicenter German ESOPEC
study is a randomized phase III trial that directly com-
pares perioperative chemotherapy according to the FLOT
protocol with preoperative chemoradiation according to
the CROSS regimen in patients with adenocarcinoma of
the esophagus with clinical stage cT1N+ M0 or cT2-4a
N0/N+, M0 (NCT02509286). Similarly, the Neo-AGIS
ICORG 10-14 trial is randomizing patients with T2 or
node-positive adenocarcinoma of the esophagus or EGJ to
preoperative chemoradiation per CROSS protocol or peri-
operative chemotherapy with ECF or ECX per the MAGIC
approach (NCT01726452). Finally, the TOPGEAR study, an
international phase III trial led by the Australasian Gastro-
Intestinal Trials Group, randomizes patients with adenocar-
cinoma of the stomach or EGJ to receive either perioperative
chemotherapy alone (ECF, or recently changed to FLOT) or
preoperative chemotherapy (ECF or FLOT) followed by pre-
operative chemoradiation with 5-FU or capecitabine (NCT
01924819).
In addition to treatment with standard cytotoxic chemo-
therapy, immune checkpoint inhibitors have emerged as
a therapeutic option in the advanced or metastatic setting,
and have the potential to enhance preoperative therapeutic
strategies. At present, checkpoint inhibitors for gastro-
esophageal cancers are U.S. Food and Drug Administration
approved for all patients with tumors that are mismatch
repair decient as characterized by microsatellite instability
high, after failure of rst-line cytotoxic chemotherapy, and
for those with tumors that have positive programmed cell
death ligand 1 (PD-L1) expression, after two lines of stan-
dard therapy. Recent studies have demonstrated single
agent antiPD-1 or antiPD-L1 activity of up to 20%30% in
patients with gastroesophageal cancer (particularly those
with PD-L1+ expression, dened as 1% staining of tumor
cells) who progressed after two or more lines of prior
therapy.
26-28
Moreover, trials evaluating the efcacy of
immunotherapy in earlier lines of therapy have shown
encouraging signals. The phase III KEYNOTE 181 trial
compared pembrolizumab with systemic chemotherapy
(investigators choice of paclitaxel, docetaxel, or irinotecan)
in 628 patients with esophageal or EGJ (Siewert type I)
cancers who had progressed after one line of standard
therapy. Although there was no important difference in OS
between the two treatment arms in the entire ITT population,
in those patients whose tumors had with high PD-L1 ex-
pression, combined positive staining (CPS 10), pem-
brolizumab demonstrated superior OS compared with
chemotherapy (9.3 vs. 6.7 months; HR 0.69; 95% CI,
0.520.93; p = .0074). Furthermore, preliminary analyses
from the phase II KEYNOTE 059 trial
29
demonstrated that in
a cohort of patients with previously untreated gastric and
GEJ cancer, uoropyrimidine, and cisplatin plus pem-
brolizumab yielded response rates of 60% in all patients and
around 69% in PD-L1+ tumor patients.
29
Several randomized
phase III trials are investigating the role of immunotherapy in
the preoperative setting for gastroesophageal cancers. The
ICONIC trial is evaluating perioperative antiPD-L1 anti-
body, avelumab, with chemotherapy (FLOT) in patients with
gastric, GEJ, or esophageal cancers (NCT03399071). An-
other phase II/III trial will evaluate antiPD-1 and anti
CTLA-4 agents, nivolumab and ipilimumab, respectively,
in patients with esophageal and EGJ cancers who are un-
dergoing surgery, with an initial randomization to pre-
operative chemoradiation with or without nivolumab, and
a second randomization to either adjuvant nivolumab or
nivolumab/ipilimumab postresection (NCT03604991). The
DANTE trial evaluates atezolizumab with FLOT in the per-
ioperative thera py of gastric and EGJ cance rs
(NCT03421288). Finally, the PANDA trial will evaluate
neoadjuvant capecitabine, oxaliplatin, and docetaxel in
combination with antiPD-L1 antibody, atezolizumab, in
resectable gastric or GEJ cancers (NCT03448835).
Amplied or overexpressed in about one-third of gastro-
esophageal adenocarcinomas, human epidermal growth
factor receptor 2 (HER2) has remained a key therapeutic
target in advanced or metastatic disease. The benetof
trastuzumab, a humanized monoclonal antibody targeting
the HER2 receptor, was rst established in the ToGA trial,
a phase III study that randomized patients with gastric or
Optimal Management of Gastroesophageal Junction Adenocarcinoma
2019 ASCO EDUCATIONAL BOOK | asco.org/edbook e93
Downloaded from ascopubs.org by UNIVERSITY CHICAGO on May 19, 2019 from 128.135.207.229
Copyright © 2019 American Society of Clinical Oncology. All rights reserved.
GEJ adenocarcinoma that were HER2-positive, to receive
standard chemotherapy (infusional 5-FU or capecitabine
with cisplatin) with or without trastuzumab. The addition of
trastuzumab improved response rates (47% vs. 35%) as
well as the primary endpoint of OS, from 11.1 months
without trastuzumab to 13.8 months with trastuzumab (HR
0.74; 95% CI, 0.600.91; p = .0046).
30
Furthermore, the
benet of adding trastuzumab to perioperative chemo-
therapy has been preliminarily demonstrated in small,
single- arm phase II trials using a CAPOX or FLOT che-
motherapy backbone, with reported pCR of up to 20% in
these cohorts.
31,32
Moreover, Schokker and colleagues
33
demonstrated the feasibility of adding dual anti-HER2
therapy with trastuzumab and pertuzumab to preoperative
chemoradiation per CROSS regimen in the phase IB TRAP
study, with reported pCR rate of 33%.
33
Ongoing randomized
clinical trials may clarify the benet of adding HER2-targeted
therapy in the preoperative setting. These studies include the
randomized phase III RTOG 1010 trial evaluating neoadjuvant
chemoradiation using the CROSS approach with or without
trastuzumab in esophageal cancers (NCT01196390). PET-
RARCA is a randomized phase II/III trial investigating peri-
operative FLOT with or without trastuzumab/pertuzumab
(NCT02581462). The INNOVATION trial is a randomized
phase II study comparing perioperative uoropyrimidine and
platinum-based chemotherapy withthesamechemotherapy
plus trastuzumab with or without pertuzumab in patients with
gastric or EGJ adenocarcinoma (NCT02205047).
Ultimately, the right approach should involve a multidisci-
plinary team assessment and consideration of patient fac-
tors that may inuence choice of therapeutics. As we
acquire nal study results that should help rene the
therapeutic approach in the node-positive disease patient,
we still need to consider the margin of benet, quality of life,
and best pratice for all patients with the diagnosis of EGJ
tumors. What we have thus far is a strong signal for systemic
therapy prior to operation for most patients. What we need,
as in many malignancies, is predictive noninvasive surro-
gates of response and outcome. We are getting close with
PET imaging to assess response during chemotherapy. We
are still steps away from identifying targetable, differentially
expressed pathways that yield higher response rates and
resullt in improvements in overall survival. Therefore, clin-
ical trial participation is highly necessary toward advancing
our understanding.
AFFILIATIONS
1
Thomas Jefferson University Hospital, Sidney Kimmel Cancer Center,
Philadelphia, PA
2
Weill Cornell Medicine, NU Presbyterian Hospital, New York, NY
3
Institute of Clinical Cancer Research, UCT University Cancer Center,
Krankenhaus Nordwest, Frankfurt, Germany
4
University of Chicago Medical Center and Biological Sciences,
Chicago, IL
5
University of Colorado, Denver, CO
CORRESPONDING AUTHOR
James A. Posey, MD, Thomas Jefferson University Hospital, Sidney
Kimmel Cancer Center, 1025 Walnut St. Suite 700, College Building,
Philadelphia, PA 19107; email: james.poseyIII@jefferson.edu.
AUTHORSDISCLOSURES OF POTENTIAL CONFLICTS OF
INTERESTAND DATA AVAILABILITY STATEMENT
Disclosures provided by the authors and data availability statement (if
applicable) are available with this article at DOI https://doi.org/10.1200/
EDBK_236827.
REFERENCES
1. Bray F, Ferlay J, Soerjomataram I, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185
countries. CA Cancer J Clin. 2018;68:394-424.
2. Blot WJ, Devesa SS, Kneller RW, et al. Rising incidence of adenocarcinoma of the esophagus and gastric cardia. JAMA. 1991;265:1287-1289.
3. Buas MF, Vaughan TL. Epidemiology and risk factors for gastroesophageal junction tumors: understanding the rising incidence of this disease. Semin Radiat
Oncol. 2013;23:3-9.
4. Siewert JR, H¨
olscher AH, Becker K, et al. [Cardia cancer: attempt at a therapeutically relevant classication]. Chirurg. 1987;58:25-32.
5. R ¨
udiger Siewert J, Feith M, Werner M. Adenocarcinoma of the esophagogastric junction: results of surgical therapy based on anatomical/topographic
classication in 1,002 consecutive patients. Ann Surg. 2000;232:353-361.
6. Siewert JR, Stein HJ. Classication of adenocarcinoma of the oesophagogastric junction. Br J Surg. 1998;85:1457-1459.
7. Amin MB, Edge SB, Greene FL. AJCC Cancer Staging Manual, 8th Ed. New York: Springer; 2017.
8. Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014;513:202-209.
9. Cancer Genome Atlas Research Network; Analysis Working Group: Asan University; BC Cancer Agency, et al. Integrated genomic characterization of oeso-
phageal carcinoma. Nature. 2017;541:169-175.
10. Njei B, McCarty TR, Birk JW. Trends in esophageal cancer survival in United States adults from 1973 to 2009: A SEER database analysis. J Gastroenter ol Hepatol.
2016;31:1141-1146.
Lin et al
e94 2019 ASCO EDUCATIONAL BOOK | asco.org/edbook
Downloaded from ascopubs.org by UNIVERSITY CHICAGO on May 19, 2019 from 128.135.207.229
Copyright © 2019 American Society of Clinical Oncology. All rights reserved.
11. van Hagen P, Hulshof MC, van Lanschot JJ, et al.; CROSS Group. Preoperative chemoradiotherapy for esophageal or junctional cancer. N Engl J Med. 2012;
366:2074-2084.
12. Al-Batran SE, Homann N, Schmalenberg H, et al. Perioperative chemotherapy with docetaxel, oxaliplatin, and uorouracil/leucovorin (FLOT) versus epirubicin,
cisplatin, and uorouracil or capecitabine (ECF/ECX) for resectable gastric or gastroesophageal junction (GEJ) adenocarcinoma (FLOT4-AIO): a multicenter,
randomized phase 3 trial (abstract). J Clin Oncol. 2017;35 (suppl; abstr 4004).
13. Al-Batran SE, Homann N, Pauligk C, et al. Perioperative chemotherapy with uorouracil plus leucovorin, oxaliplatin, and docetaxel versus uorouracil or
capecitabine plus cisplatin and epirubicin for locally advanced, resectable gastric or gastro-oesophageal junction adenocarcinoma (FLOT4): a multicentre, open-
label, randomised, phase 2/3 trial. Lancet. In press.
14. Ychou M, Boige V, Pignon JP, et al. Perioperative chemotherapy compared with surgery alone for resectable gastroesophageal adenocarcino ma: an FNCLCC and
FFCD multicenter phase III trial. J Clin Oncol. 2011;29:1715-1721.
15. Cunningham D, Allum WH, Stenning SP, et al.; MAGIC Trial Participants. Perioperative chemotherapy versus surgery alone for resectable gastroesophageal
cancer. N Engl J Med. 2006;355:11-20.
16. Shapiro J, van Lanschot JJB, Hulshof MCCM, et al.; CROSS study group. Neoadjuvant chemoradiotherapy plus surgery versus surgery alone for oesophageal or
junctional cancer (CROSS): long-term results of a randomised controlled trial. Lancet Oncol. 2015;16:1090-1098.
17. Stahl M, Walz MK, Stuschke M, et al. Phase III comparison of preoperative chemotherapy compared with chemoradiotherapy in patients with locally advanced
adenocarcinoma of the esophagogastric junction. J Clin Oncol. 2009;27:851-856.
18. Leers JM, DeMeester SR, Chan N, et al. Clinical characteristics, biologic behavior, and survival after esophagectomy are similar for adenocarcinoma of the
gastroesophageal junction and the distal esophagus. J Thorac Cardiovasc Surg. 2009;138:594-602, discussion 601-602.
19. Kelsen DP, Winter KA, Gunderson LL, et al.; USA Intergroup. Long-term results of RT OG trial 8911 (USA Intergroup 113): a random assignment trial comparison
of chemotherapy followed by surgery compared with surgery alone for esophageal cancer. J Clin Oncol. 2007;25:3719-3725.
20. Chan DS, Reid TD, Howell I, et al. Systematic review and meta-analysis of the inuence of circumfere ntial resection margin involvement on survival in patients
with operable oesophageal cancer. Br J Surg. 2013;100:456-464.
21. Gilbert S, Martel AB, Seely AJ, et al. Prognostic signicance of a positive radial margin after esophageal cancer resection. J Thorac Cardiovasc Surg. 2015;
149:548-555, discussion 555.
22. Gebski V, Burmeister B, Smithers BM, et al.; Australasian Gastro -Intestinal Trials Group. Survival benets from neoadjuvant chemoradiotherapy or chemotherapy
in oesophageal carcinoma: a meta-analysis. Lancet Oncol. 2007;8:226-234.
23. Sjoquist KM, Burmeister BH, Smithers BM, et al.; Australasian Gastro-Intestinal Trial s Group. Survival after neoadjuvant chemotherapy or chemoradiotherapy for
resectable oesophageal carcinoma: an updated meta-analysis. Lancet Oncol. 2011;12:681-692.
24. Goodman KA, Niedzwiecki D, Hall N, et al. Initial results of CALGB 80803 (Alliance): a randomized phase II trial of PET scan-directed combin ed modality therapy
for esophageal cancer. J Clin Oncol. 2017;35 (suppl 4; abstr 1).
25. Goodman KA, Hall N, Bekai-Saab T, et al. Survival Outcomes in CALGB 80803 (Alliance): a randomized phase II Trial of PET scan-directed combined modality
therapy for esophageal cancer. J Clin Oncol. 2018;36(suppl; abstr 4012).
26. Muro K, Chung HC, Shankaran V, et al. Pembrolizumab for patients with PD-L1-positive advanced gastric cancer (KEYNOTE-012): a multicentre, open-label,
phase 1b trial. Lancet Oncol. 2016;17:717-726.
27. Doi T, Piha-Paul SA, Jalal SI, et al. Safety and antitumor activity of the anti-programmed death-1 antibody pembrolizumab in patients with advanced esophageal
carcinoma. J Clin Oncol. 2018;36:61-67.
28. Kang YK, Boku N, Satoh T, et al. Nivolumab in patients with advanced gastric or gastro-oesophageal junction cancer refractory to, or intolerant of,atleast
two previous chemotherapy regimens (ONO-4538-12, ATTRACTION-2): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2017;
390:2461-2471.
29. Bang YJ, Muro K, Fuchs CS, et al. KEYNOTE-059 cohort 2: safety and efcacy of pembrolizumab (pembro) plus 5-uorouracil (5-FU) and cisplatin for rst-line
(1L) treatment of advanced gastric cancer. J Clin Oncol. 2017;35 (suppl 15; abstr 4012).
30. Bang YJ, Van Cutsem E, Feyereislova A, et al; ToGA Trial Investigators. Trastuzumab in combination with chemotherapy versus chemotherapy alone for
treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet. 2010;
376:687-697.
31. Rivera F, Jim ´
enez-Fonseca P, Garcia Alfonso P, et al. NeoHx study: perioperative treatment with trastuzum ab in combination with capecitabine and oxaliplatin
(XELOX-T) in patients with HER2 resectable stomach or esophagogastric junction (EGJ) adenocarcinomaR0 resection, pCR, and toxicity analysis. J Clin Oncol.
2013;31 (suppl 15; abstr 4098).
32. Hofheinz R, Hegewisch-Becker S, Thuss-Patience PC, et al. HER-FLOT: trastuzumab in combination with FLOT as perioperative treatment for patients with
HER2-positive locally advanced esophagogastric adenocarcinoma: a phase II trial of the AIO gastric cancer study group (abstract). J Clin Oncol. 2014;36
(15_suppl; abstr 4073).
33. Schokker S, Molenaar RJ, Meijer SL, et al. Feasibility study of trastuzumab (T) and pertuzumab (P) added to neoadjuvant chemoradiotherapy (nCRT) in
resectable HER2+ esophageal adenocarcinoma (EAC) patients (pts): the TRAP study. J Clin Oncol. 2018;36 (15_suppl; abstr 4057).
Optimal Management of Gastroesophageal Junction Adenocarcinoma
2019 ASCO EDUCATIONAL BOOK | asco.org/edbook e95
Downloaded from ascopubs.org by UNIVERSITY CHICAGO on May 19, 2019 from 128.135.207.229
Copyright © 2019 American Society of Clinical Oncology. All rights reserved.
... There are three types of GEJ tumors: the Siewert classification of a GEJ mass is based on the location of the epicenter of the tumors and its relationship with the GEJ. Type I lesions are located 5 cm proximal to the GEJ, type II lesions span the GEJ and have their focal point up to 2 cm below the GEJ, and type III cardiac tumors extend up to 5 cm into the stomach [4]. It is important to differentiate between the three to have optimal surgical planning and specified preoperative and postoperative care [5]. ...
... Proximal gastric cancer (PGC) was also found in 17% cases. In previous reports, increased incidence of tumor occurrence in proximal end has also been noted [15,16,17]. This may be related to many factors, including Helicobacter pylori infection and eating habits. ...
Article
Full-text available
Background: Gastric cancer (GC), is a common malignancy that poses a significant health burden in India. Although clinicopathological studies of GC helps to generate baseline data and guide future health care strategies and management but there is limited literature on this regard, particularly in this geographical part. In this study, we designed to evaluate the clinicopathological profile of gastric cancer with an aim to detect the cancer early for reducing the morbidity and mortality. Materials and Methods: During this hospital-based retrospective study, clinicopathological information was extracted from hospital records of GC patients who underwent subtotal or total gastrectomy between the years 2018 and 2020 Results: A total 279 cases of gastric carcinoma were included out of which male-to-female ratio was 2.4:1. The mean age of the study population was 54.47±12.2 years with range 18 to 82 years. The frequency of gastric cancer was highest in the antrum. Adenocarcinoma was the most frequent histologic subtype. The majority of our patients presented at an advanced stage locally. Conclusion: The present study confirms that the incidence of gastric cancer surges between the fourth and sixth decades. Males are disproportionately afflicted. The most prevalent symptom is abdominal pain, which is frequently vague and therefore disregarded. The majority of patients exhibited advanced disease. Raising public awareness can help us detect the disease earlier and develop a more effective treatment.
... The surgical approach involves the removal of a portion of the stomach, the segment of the esophagus affected by cancer, and approximately 3 to 4 inches (about 7.6 to 10 cm) of healthy esophagus above it if the cancer is located in the lower part of the esophagus, close to the stomach, or at the gastroesophageal (GE) junction where the esophagus and stomach meet. Afterward, the remaining esophagus is connected to the stomach either high in the chest or in the neck [17]. ...
Article
Full-text available
Esophageal cancer is a highly aggressive and deadly disease, ranking as the sixth leading cause of cancer-related deaths worldwide. Despite advances in treatment, the prognosis remains poor. A multidisciplinary approach is crucial for achieving complete remission, with treatment options varying based on disease stage. Surgical intervention and endoscopic treatment are used for localized cancer, while systemic treatments like chemoradiotherapy and targeted drug therapy play a crucial role. Molecular markers such as HER2 and EGFR can be targeted with drugs like trastuzumab and cetuximab, and immunotherapy drugs like pembrolizumab and nivolumab show promise by targeting immune checkpoint proteins. Epigenetic modifications offer new avenues for targeted therapy. Treatment selection depends on factors like stage, tumor location, and patient health, with post-operative and rehabilitation care being essential. Early diagnosis, appropriate treatment, and supportive care are key to improving outcomes. Continued research is needed to develop effective targeted drugs with minimal side effects. This review serves as a valuable resource for clinicians and researchers dedicated to enhancing esophageal cancer treatment outcomes.
... Adenocarcinoma of the esophagogastric junction (AEG) is relatively rare but highly aggressive, 1 being a great health threat worldwide. The classification of AEG lacks a single standard system due to its special anatomical location, 2 and each subtype exerts different clinicopathological properties. 3 The Siewert classification divides AEG into three types based on the endoscopic location of tumor epicenter relative to the esophagogastric junction (EGJ): Siewert type I (5 to 1 cm above the EGJ), type II (1 cm above to 2 cm below the EGJ), and type III (2 to 5 cm below the EGJ). ...
Article
Full-text available
Objective It remains unclear whether primary tumor resection improves survival in patients with metastatic Siewert type II adenocarcinoma of the esophagogastric junction (AEG). Therefore, our study attempted to investigate the prognostic value of primary tumor resection on metastatic AEG. Methods In total, 4200 patients diagnosed with metastatic AEG were retrieved from the Surveillance, Epidemiology, and End Results (SEER) database from 2004 to 2015. Patients were categorized into two groups according to the performance of primary tumor resection. Pearson’s chi-square test, Kaplan–Meier survival curve, and Cox regression analysis were conducted in this study. In addition, propensity-score matching was conducted to match 323 patients who received primary tumor resection and another 323 patients without. Results Multivariate Cox regression analysis demonstrated that primary tumor resection was a significant prognostic factor in patients with metastatic AEG before matching. Moreover, in the matched cohort, metastatic AEG patients receiving primary tumor resection had significantly longer overall survival (hazard ratio [HR]: .54, 95% confidence interval [CI]: .46–.64, P < .001) and cancer-specific survival (HR: .53, 95% CI: .45–.63, P < .001). Subgroup analysis similarly revealed that primary tumor resection was significantly associated with better survival in most subgroups. Conclusion The present population-based study identified that primary tumor resection led to significantly superior survival in patients with metastatic AEG. These findings are likely to contribute to the development of individualized therapy in metastatic AEG.
... Gastric adenocarcinoma (GAC) and gastroesophageal junction adenocarcinoma (GEJA) are prevalent and dangerous malignancies worldwide, with more than 1 million newly diagnosed cases and over 0.7 million deaths in 2020 (1)(2)(3). It has been reported that the incidence of GAC has declined, while that of GEJA has steadily increased in recent years (4)(5)(6). Moreover, there is a large proportion of GAC and GEJA patients with advanced disease at diagnosis who lose the opportunity for potentially curative surgical resection (7,8). ...
Article
Full-text available
Objective Apatinib and irinotecan are used as systematic therapies for advanced gastric adenocarcinoma (GAC) and gastroesophageal junction adenocarcinoma (GEJA), while the evidence for their combination as second-line therapy in these patients is limited. This study aimed to evaluate the efficacy and safety of second-line apatinib plus irinotecan for the treatment of GAC and GEJA. Methods In this prospective, multicenter phase II clinical study, 28 patients with advanced GAC or GEJA who received second-line apatinib plus irinotecan were recruited. Results In total, 1 (3.6%) patient achieved complete response, 7 (25.0%) patients achieved partial response, 13 (46.4%) patients had stable disease, and 4 (14.3%) patients showed progressive disease, while clinical response was not evaluable or not assessed in 3 (10.7%) patients. The objective response rate and disease control rate were 28.6% and 75.0%, respectively. Meanwhile, the median (95% confidence interval (CI)) progression-free survival (PFS) was 4.5 (3.9-5.1) months, and the median (95% CI) overall survival (OS) was 11.3 (7.4-15.1) months. By multivariate Cox regression analysis, male sex, liver metastasis, and peritoneal metastasis were independently associated with worse PFS or OS, while treatment duration ≥5 months was independently associated with better OS. In terms of the safety profile, 89.3% of patients experienced treatment-emergent adverse events of any grade, among which 82.1% of patients had grade 1-2 adverse events and 64.3% of patients had grade 3-4 adverse events. Conclusion Apatinib plus irinotecan as second-line therapy achieves a good treatment response and satisfactory survival with tolerable safety in patients with advanced GAC or GEJA.
... At present, there is controversy regarding the treatment of advanced, unresectable, and metastatic EAC/GEA (27). Although current platinum-based chemotherapy regimens have made some progress in improving the OS of patients with advanced, unresectable, and metastatic EAC/GEA, the 5-year survival rate of patients is still unsatisfactory. ...
Article
Full-text available
Background There is increasing evidence that immunotherapy (programmed cell death-1 (PD-1) inhibitor) combined with chemotherapy is superior to chemotherapy alone in neoadjuvant therapy for patients with previously untreated, unresectable advanced, or metastatic esophageal adenocarcinoma (EAC)/gastric/gastroesophageal junction adenocarcinoma (GEA). However, the results of recent studies have been contradictory. Therefore, the aim of this article is to evaluate the efficacy and safety of PD-1 inhibitors combined with chemotherapy in neoadjuvant therapy through meta-analysis. Method We comprehensively reviewed the literature and clinical randomized controlled trials (RCTs) by February 2022 by searching Medical Subject Headings (MeSH) and keywords such as “esophageal adenocarcinoma” or “immunotherapy” in several databases, including the Embase, Cochrane, PubMed, and ClinicalTrials.gov websites. Two authors independently selected studies, extracted data, and assessed the risk of bias and quality of evidence by using standardized Cochrane Methods procedures. The primary outcomes were 1-year overall survival (OS) and 1-year progression-free survival (PFS), estimated by calculating the 95% confidence interval (CI) for the combined odds ratio (OR) and hazard ratio (HR). Secondary outcomes estimated using OR were disease objective response rate (DORR) and incidence of adverse events. Results Four RCTs with a total of 3,013 patients researching the efficacy of immunotherapy plus chemotherapy versus chemotherapy alone on gastrointestinal cancer were included in this meta-analysis. The results showed that immune checkpoint inhibitor plus chemotherapy treatment was associated with an increased risk of PFS (HR = 0.76 [95% CI: 0.70–0.83]; p < 0.001), OS (HR = 0.81 [95% CI: 0.74–0.89]; p < 0.001), and DORR (relative ratio (RR) = 1.31 [95% CI: 1.19–1.44]; p < 0.0001) when compared with chemotherapy alone in advanced, unresectable, and metastatic EAC/GEA. However, immunotherapy combined with chemotherapy increased the incidence of adverse reactions such as alanine aminotransferase elevation (OR = 1.55 [95% CI: 1.17–2.07]; p = 0.003) and palmar-plantar erythrodysesthesia (PPE) syndrome (OR = 1.30 [95% CI: 1.05–1.63]; p = 0.02). Nausea (OR = 1.24 [95% CI: 1.07–1.44]; p = 0.005) and white blood cell count decreased (OR = 1.40 [95% CI: 1.13–1.73]; p = 0.002), and so on. Fortunately, toxicities were within acceptable limits. Meanwhile, for patients with a combined positive score (CPS) ≥1, compared with chemotherapy alone, immunotherapy combined with chemotherapy had a better overall survival rate (HR = 0.81 [95% CI: 0.73–0.90]; p = 0.0001). Conclusion Our study shows that immunotherapy plus chemotherapy has an obvious benefit for patients with previously untreated, unresectable advanced, or metastatic EAC/GEA when compared with chemotherapy alone. However, a high risk of adverse reactions may occur during immunotherapy plus chemotherapy, and more studies focusing on the treatment strategies of untreated, unresectable advanced, or metastatic EAC/GEA are warranted. Systematic review registration www.crd.york.ac.uk , identifier CRD42022319434.
... Gastroesophageal junction cancers are generally classified according to the American Joint Committee on Cancer as either gastric or esophageal cancer, depending on the location of its epicentre relative to the cardia [3]. In the locally advanced and metastatic settings, however, gastric and gastroesophageal junction cancers are managed similarly [4,5]. ...
Article
Full-text available
Background: Gastric cancer mortality remains among the highest of all cancers. Trifluridine/tipiracil (FTD/TPI) represents Canada's first standard-of-care, third-line, systemic therapy for metastatic gastric/gastroesophageal cancer. We characterized real-world treatment patterns in patients enrolled to receive FTD/TPI through Taiho Pharma Canada's Patient Support Program. Methods: Demographic and clinical information were collected from November 2019 to November 2021 for adult patients with refractory metastatic gastric/gastroesophageal cancer throughout Canada. We examined all variables using descriptive statistics and performed survival and association analyses. Results: 162 patients enrolled to receive FTD/TPI with a median age of 65 years, 12 of whom had HER2 positive disease. Among 123 patients who started FTD/TPI, median follow-up was 3.1 months and median progression-free survival (PFS) was 3.5 months (95% CI 3.2-4.0). Among 121 patients who discontinued FTD/TPI, median treatment duration was 2.39 cycles (IQR 1.14-3.86). A total of 52% discontinued treatment due to disease progression, and 27% had a dose reduction or delay. On multivariable logistic regression, prior FOLFIRI was a statistically significant predictor of treatment modification. Conclusions: Through the Patient Support Program, FTD/TPI is an actively utilized treatment option in heavily pretreated metastatic gastric/gastroesophageal cancer, despite its recent introduction. With longer-than-expected treatment duration and PFS, FTD/TPI likely addresses an important unmet need for effective and tolerable therapies in this setting.
... Overall prognosis and survival for EGAC patients remain poor (3). Despite advancements in multimodal therapies, EGAC is difficult to treat due to advanced disease at the time of diagnosis necessitating research into molecular mechanisms which control tumor behavior to improve its detection and treatment options (4). Glypican-1 (GPC1) is a membrane bound heparan sulfate proteoglycans (HSPG) (5). ...
Article
Full-text available
Background: Glypican 1 (GPC1) is a heparan sulphate proteoglycan cell membrane protein. It is implicated in driving cancers of the breast, brain, pancreas, and prostate; however, its role in esophagogastric cancer (EGAC) remains unexplored. The aim of the study was to investigate and elucidate the molecular mechanistic of GPC1 in human EGAC. Methods: Thirty tissue and 120 microarray sections of EGAC were evaluated with Anti-GPC1 immunohistochemistry. Loss and gain of GPC1 function were performed using lentivirus transfection in EGAC cell lines. Mechanistically, AKT/GSK/β-catenin pathway was evaluated using AKT inhibitor MK-2206 and Wnt/β-catenin stimulant LiCl. Results: GPC1 overexpression was found in 102 cases (68%). Overexpression of GPC1 correlated with lymph node metastasis, poor differentiation and decreased overall survival. Lentivirus mediated GPC1 knockdown resulted in decreased cell proliferation, migration, invasion, and colony formation. Knockdown caused G0/G1 cell cycle arrest, increased apoptosis, and reduced epithelial mesenchymal transition (EMT). GPC1 mediated its effects by activation of AKT/GSK/β-catenin pathway. Conclusions: This is the first descriptive study to decipher the role of GPC1 in EGAC. Our results suggest that GPC1 regulates cell proliferation and growth and may serve as an attractive oncotarget in EGAC.
... The development of HER-2 inhibitors has changed the landscape of treatment for patients suffering HER-2 positive tumors [8]. Adding HER-2-directed agents to the standard chemotherapeutic regimen has demonstrated survival benefits in several malignant tumors, including BC, gastric cancer, and gastroesophageal junction adenocarcinoma [9][10][11]. Trastuzumab is a prototype of humanized anti-HER-2 monoclonal therapeutic antibodies that selectively target the extracellular domain of the HER-2 receptor [12]. ...
Article
Background: Around 20% of breast cancers (BCs) overexpress Human Epidermal Growth Factor Receptor 2 (HER-2). HER-2 overexpression is associated with increased tumor aggressiveness and poor prognosis. Trastuzumab (an anti-HER2 monoclonal antibody) has been reported to improve overall survival in early-stage and metastatic BCs, but at the expense of increasing cardiac morbidity. In the current review study, we aims to discuss the pathogenesis of trastuzumab-induced cardiotoxicity and the potential ameliorating role of spironolactone in this regard. Methods: The search strategy aimed to identify both published and unpublished studies. First off, we identified keywords and index terms, including trastuzumab, cardiotoxicity, heart failure, and spironolactone to conduct a broad search in PubMed, Embase, Scopus, and Web of Science, using the aforementioned keywords either individually or in combination. Lastly, the reference list of all identified articles was also evaluated. Our study included observational and interventional studies, case-reports, and systematic reviews and meta-analyses. Results: Trastuzumab could deteriorate mitochondrial function and subsequently leads to the accumulation of Reactive Oxygen Species (ROS) in cardiomyocytes. Published clinical studies offered conflicting results regarding the efficacy of angiotensin-converting enzyme inhibitors and beta-blockers in respect of trastuzumab-induced cardiotoxicity. On the other hand, spironolactone was found to have both antioxidant and anti-inflammatory properties. Recent in-vivo studies supported the cardioprotective effect of spironolactone through maintaining mitochondrial ultrastructure and reducing ROS production. Conclusion: Although spironolactone mitigates oxidative stress and mitochondrial dysfunction, there is a lack of clinical evidence to support the effectiveness of spironolactone in trastuzumab-induced cardiotoxicity. Design and implementation of clinical trials are recommended to determine the potential beneficial effects of spironolactone on trastuzumab-induced cardiotoxicity.
Article
Full-text available
Anestesi umum adalah suatu tindakan yang ditujukan untuk menghilangkan rasa nyeri, membuat tidak sadarkan diri dan menimbulkan amnesia yang reversibel dan dapat diprediksi. Metode atau teknik anestesi umum terbagi menjadi 3 yaitu teknik anestesi umum inhalasi, anestesi umum intravena dan anestesi umum seimbang.
Article
Full-text available
Purpose The anti–programmed death-1 antibody pembrolizumab was evaluated in KEYNOTE-028, a multicohort, phase IB study of patients with programmed death ligand-1 (PD-L1)–positive advanced solid tumors. Results from the esophageal carcinoma cohort are reported herein. Patients and Methods Eligible patients with squamous cell carcinoma or adenocarcinoma of the esophagus or gastroesophageal junction in whom standard therapy failed and who had PD-L1–positive tumors received pembrolizumab 10 mg/kg every 2 weeks for up to 2 years or until confirmed disease progression or intolerable toxicity. Response was assessed every 8 weeks up to 6 months and every 12 weeks thereafter. Primary end points were safety and overall response rate, determined by investigator review per Response Evaluation Criteria in Solid Tumors (version 1.1). Results Among 83 patients with esophageal carcinoma and samples evaluable for PD-L1 expression, 37 (45%) had PD-L1–positive tumors, and 23 were enrolled. Median age was 65 years; 78% had squamous histology; and 87% received ≥ two prior therapies for advanced/metastatic disease. As of the data cutoff (February 20, 2017), median follow-up was 7 months (range, 1 to 33 months). Nine patients (39%) experienced treatment-related adverse events, most commonly decreased appetite, decreased lymphocyte count, generalized rash, and rash (two patients [9%] each). No grade 4 adverse events or deaths were attributed to pembrolizumab. Overall response rate was 30% (95% CI, 13% to 53%); median duration of response was 15 months (range, 6 to 26 months). A six-gene interferon-γ gene expression signature analysis suggested that delayed progression and increased response occur among pembrolizumab-treated patients with higher interferon-γ composite scores. Conclusion Pembrolizumab demonstrated manageable toxicity and durable antitumor activity in patients with heavily pretreated, PD-L1–positive advanced esophageal carcinoma.
Article
Background: Docetaxel-based chemotherapy is effective in metastatic gastric and gastro-oesophageal junction adenocarcinoma. This study reports on the safety and efficacy of the docetaxel-based triplet FLOT (fluorouracil plus leucovorin, oxaliplatin and docetaxel) as a perioperative therapy for patients with locally advanced, resectable tumours. Methods: In this controlled, open-label, phase 2/3 trial, we randomly assigned 716 patients with histologically-confirmed advanced clinical stage cT2 or higher or nodal positive stage (cN+), or both, resectable tumours, with no evidence of distant metastases, via central interactive web-based-response system, to receive either three pre-operative and three postoperative 3-week cycles of 50 mg/m2 epirubicin and 60 mg/m2 cisplatin on day 1 plus either 200 mg/m2 fluorouracil as continuous intravenous infusion or 1250 mg/m2 capecitabine orally on days 1 to 21 (ECF/ECX; control group) or four preoperative and four postoperative 2-week cycles of 50 mg/m2 docetaxel, 85 mg/m2 oxaliplatin, 200 mg/m2 leucovorin and 2600 mg/m2 fluorouracil as 24-h infusion on day 1 (FLOT; experimental group). The primary outcome of the trial was overall survival (superiority) analysed in the intention-to-treat population. This trial is registered with ClinicalTrials.gov, number NCT01216644. Findings: Between Aug 8, 2010, and Feb 10, 2015, 716 patients were randomly assigned to treatment in 38 German hospitals or with practice-based oncologists. 360 patients were assigned to ECF/ECX and 356 patients to FLOT. Overall survival was increased in the FLOT group compared with the ECF/ECX group (hazard ratio [HR] 0·77; 95% confidence interval [CI; 0.63 to 0·94]; median overall survival, 50 months [38·33 to not reached] vs 35 months [27·35 to 46·26]). The number of patients with related serious adverse events (including those occurring during hospital stay for surgery) was similar in the two groups (96 [27%] in the ECF/ECX group vs 97 [27%] in the FLOT group), as was the number of toxic deaths (two [<1%] in both groups). Hospitalisation for toxicity occurred in 94 patients (26%) in the ECF/ECX group and 89 patients (25%) in the FLOT group. Interpretation: In locally advanced, resectable gastric or gastro-oesophageal junction adenocarcinoma, perioperative FLOT improved overall survival compared with perioperative ECF/ECX. Funding: The German Cancer Aid (Deutsche Krebshilfe), Sanofi-Aventis, Chugai, and Stiftung Leben mit Krebs Foundation.
Article
This article provides a status report on the global burden of cancer worldwide using the GLOBOCAN 2018 estimates of cancer incidence and mortality produced by the International Agency for Research on Cancer, with a focus on geographic variability across 20 world regions. There will be an estimated 18.1 million new cancer cases (17.0 million excluding nonmelanoma skin cancer) and 9.6 million cancer deaths (9.5 million excluding nonmelanoma skin cancer) in 2018. In both sexes combined, lung cancer is the most commonly diagnosed cancer (11.6% of the total cases) and the leading cause of cancer death (18.4% of the total cancer deaths), closely followed by female breast cancer (11.6%), prostate cancer (7.1%), and colorectal cancer (6.1%) for incidence and colorectal cancer (9.2%), stomach cancer (8.2%), and liver cancer (8.2%) for mortality. Lung cancer is the most frequent cancer and the leading cause of cancer death among males, followed by prostate and colorectal cancer (for incidence) and liver and stomach cancer (for mortality). Among females, breast cancer is the most commonly diagnosed cancer and the leading cause of cancer death, followed by colorectal and lung cancer (for incidence), and vice versa (for mortality); cervical cancer ranks fourth for both incidence and mortality. The most frequently diagnosed cancer and the leading cause of cancer death, however, substantially vary across countries and within each country depending on the degree of economic development and associated social and life style factors. It is noteworthy that high‐quality cancer registry data, the basis for planning and implementing evidence‐based cancer control programs, are not available in most low‐ and middle‐income countries. The Global Initiative for Cancer Registry Development is an international partnership that supports better estimation, as well as the collection and use of local data, to prioritize and evaluate national cancer control efforts. CA: A Cancer Journal for Clinicians 2018;0:1‐31. © 2018 American Cancer Society
Article
4012 Background: Preliminary analyses from the global, multicohort, phase 2 KEYNOTE-059 (NCT02335411) study suggested that safety of pembro + 5-FU + cisplatin is manageable as 1L therapy in pts with advanced gastric or gastroesophageal junction (G/GEJ) cancer (cohort 2). We present efficacy and updated safety data from KEYNOTE-059 cohort 2. Methods: Cohort 2 enrolled pts ≥18 y with HER2 – recurrent or metastatic G/GEJ adenocarcinoma, measurable disease, no prior therapy for metastatic/advanced disease, and ECOG PS 0-1. Pts received pembro 200 mg on day 1 of each 21-day cycle + cisplatin 80 mg/m ² for 6 cycles + 5-FU 800 mg/m ² (or capecitabine 1000 mg/m ² in Japan) Q3W for up to 2 y or until disease progression, investigator/pt decision to withdrawal, or unacceptable toxicity. PD-L1 ⁺ pts had expression in ≥1% tumor or stromal cells using IHC (22C3 antibody). End points were safety and tolerability (primary), ORR (RECIST v1.1, by central review), DOR, PFS, and OS (secondary). Results: Of 25 enrolled pts, 64% were men, 68% were Asian, and 64% had PD-L1 ⁺ tumors. Median age was 64 y. At data cutoff (Oct 19, 2016), median duration of follow-up was 12.2 mo (range, 1.8 to 19.6) and 84% of pts had discontinued treatment, mainly owing to clinical or radiologic disease progression (64%). ORR (CR + PR) was 60% (95% CI, 38.7-78.9) in all pts. Overall, 32% of pts had SD (95% CI, 14.9-53.5), 4% had PD (95% CI, 0.1-20.4), and 4% were not evaluable (95% CI, 0.1-20.4). ORR was 68.8% (95% CI, 41.3-89.0) in PD-L1 ⁺ pts and 37.5% (95% CI, 8.5-75.5) in PD-L1 – pts. Median DOR (range) was 4.6 mo (2.6 to 14.4+) in all pts, 4.6 mo (3.2 to 14.4+) in PD-L1 ⁺ pts, and 5.4 mo (2.8 to 8.3+) in PD-L1 – pts. Median PFS was 6.6 mo (95% CI, 5.9-10.6); median OS was 13.8 mo (95% CI, 7.3-not estimable). Grade 3-4 treatment-related adverse events (TRAEs) occurred in 76% of pts. TRAEs led to discontinuation in 3 pts (grade 3 stomatitis, grade 2 hypoacusis, and grade 1 creatinine increase). No TRAEs were fatal. Conclusions: Pembro + 5-FU + cisplatin showed manageable safety and encouraging antitumor activity as 1L therapy for pts with advanced G/GEJ cancer. Further exploration of pembro + 5-FU + cisplatin in this setting is warranted. Clinical trial information: NCT02335411.
Article
Background: Patients with advanced gastric or gastro-oesophageal junction cancer refractory to, or intolerant of, two or more previous regimens of chemotherapy have a poor prognosis, and current guidelines do not recommend any specific treatments for these patients. We assessed the efficacy and safety of nivolumab, a fully human IgG4 monoclonal antibody inhibitor of programmed death-1 (PD-1), in patients with advanced gastric or gastro-oesophageal junction cancer who had been previously been treated with two or more chemotherapy regimens. Methods: In this randomised, double-blind, placebo-controlled, phase 3 trial done at 49 clinical sites in Japan, South Korea, and Taiwan, eligible patients (aged ≥20 years with unresectable advanced or recurrent gastric or gastro-oesophageal junction cancer refractory to, or intolerant of, standard therapy [including two or more previous chemotherapy regimens], with an Eastern Cooperative Oncology Group [ECOG] performance status of 0-1, and naive to anti-PD-1 therapy or other therapeutic antibodies and pharmacotherapies for the regulation of T cells) were recruited. Patients were randomly assigned (2:1) using an interactive web response system to receive 3 mg/kg nivolumab or placebo intravenously every 2 weeks, stratified by country, ECOG performance status, and number of organs with metastases. Study treatment was continued until progressive disease per investigator assessment or onset of toxicities requiring permanent discontinuation. Patients and investigators were masked to group assignment. The primary endpoint was overall survival in the intention-to-treat population. Safety was analysed in all patients who received at least one dose of study treatment. This study is ongoing but not recruiting new patients, and is registered with ClinicalTrials.gov, number NCT02267343. Findings: Between Nov 4, 2014, and Feb 26, 2016, we randomly assigned 493 patients to receive nivolumab (n=330) or placebo (n=163). At the data cutoff (Aug 13, 2016), median follow-up in surviving patients was 8·87 months (IQR 6·57-12·37) in the nivolumab group and 8·59 months (5·65-11·37) in the placebo group. Median overall survival was 5·26 months (95% CI 4·60-6·37) in the nivolumab group and 4·14 months (3·42-4·86) in the placebo group (hazard ratio 0·63, 95% CI 0·51-0·78; p<0·0001). 12-month overall survival rates were 26·2% (95% CI 20·7-32·0) with nivolumab and 10·9% (6·2-17·0) with placebo. Grade 3 or 4 treatment-related adverse events occurred in 34 (10%) of 330 patients who received nivolumab and seven (4%) of 161 patients who received placebo; treatment-related adverse events led to death in five (2%) of 330 patients in the nivolumab group and two (1%) of 161 patients in the placebo group. No new safety signals were observed. Interpretation: In this phase 3 study, the survival benefits indicate that nivolumab might be a new treatment option for heavily pretreated patients with advanced gastric or gastro-oesophageal junction cancer. Ongoing trials that include non-Asian patients are investigating nivolumab for advanced gastric or gastro-oesophageal junction cancer in various settings and earlier treatment lines. Funding: Ono Pharmaceutical and Bristol-Myers Squibb.
Article
1 Background: To determine whether changing chemotherapy (CT) during pre-op chemoradiation (CRT) based on response to induction CT by ¹⁸ F-fluoro-deoxyglucose PET imaging can lead to improved pathologic complete response (pCR) in patients (pts) with resectable esophageal and gastroesophageal junction (GEJ) adenocarcinomas. Methods: 257 eligible pts were enrolled, underwent baseline PET scan, and were randomized to one of 2 induction CT arms: Modified FOLFOX-6 (oxaliplatin, leucovorin, 5-FU), days 1, 15, 29 or Carboplatin/Paclitaxel (CP), days 1, 8, 22, 29. Repeat PET was performed days 36-42; change in max standardized uptake value (SUV) from baseline was assessed. PET non-responders (≤35% decrease in SUV – PET-NR) crossed over to alternative CT regimen during CRT (50.4Gy/28 fractions). PET responders (>35% decrease in SUV – PET-R) continued on same CT during CRT. Pts underwent surgery 6 wks post-CRT. Pts evaluable if had surgery, disease progression (PD), death due to disease, were unresectable or had adverse event (AE). Primary endpoint was pCR in PET-NR who crossed over to alternative CT (expected 5% under H 0 to 20% under H a ). Results: Pre-audit PET response data after induction CT and pCR rates after CRT and surgery are shown in the table. For PET-NR who crossed over to alternative CT during CRT, pCR was 15.6%; 95% CI (0.08, 0.26). Conclusions: Efficacy criteria were met for an improvement in pCR rates among pts who were PET-NR after induction CT and received alternative CT during CRT for esophageal and GEJ adenoca. Pts receiving induction and concurrent CP had an unexpectedly low pCR. Support: U10CA180821, U10CA180882 Clinical trial information: NCT01333033. [Table: see text]