PIIS0140673624022268
PIIS0140673624022268
PIIS0140673624022268
Oesophageal cancer
Hong Yang, Feng Wang, Christopher L Hallemeier, Toni Lerut, Jianhua Fu
Oesophageal cancer is the seventh leading cause of cancer mortality worldwide. Two major pathological subtypes ex- Lancet 2024; 404: 1991–2005
ist: oesophageal squamous cell carcinoma and oesophageal adenocarcinoma. Epidemiological studies in the last dec- Department of Thoracic
ade have shown a gradual increase in the incidence of oesophageal adenocarcinoma worldwide. The prognosis of Surgery (Prof H Yang MD PhD,
Prof J Fu MD PhD) and
oesophageal cancer has greatly improved due to breakthroughs in screening, surgical procedures, and novel treat-
Department of Medical
ment modalities. The success achieved with combined modality therapies, including surgery, chemotherapy, and ra- Oncology
diotherapy, to treat locally advanced oesophageal cancer is particularly notable. Immunotherapy has become a crucial (Prof F Wang MD PhD), Sun
treatment for oesophageal cancer, with immune checkpoint inhibitor-based therapies now established as the standard Ya University Cancer
Center, Guangzhou, China;
of care in adjuvant and metastatic first-line settings. This Seminar provides an overview of advances in the screening,
Guangdong Provincial Clinical
diagnosis, and treatment of oesophageal squamous cell carcinoma and oesophageal adenocarcinoma, with a particu- Research Center for Cancer,
lar focus on neoadjuvant therapies for locally advanced oesophageal cancer and immune checkpoint inhibitor-based State Key Laboratory of
therapies. Oncology in South China,
Collaborative Innovation
Centre for Cancer Medicine,
Introduction of scalding foods and beverages are potential risks.5 For Guangzhou, China (Prof H Yang,
Oesophageal cancer, originating from the epithelial oesophageal adenocarcinoma, risk factors include Prof F Wang, Prof J Fu);
cells of the oesophagus, is a major global health burden. smoking, obesity, gastro-oesophageal reflux disease, and Guangdong Esophageal Cancer
Institute, Guangzhou, China
The two major pathological subtypes, oesophageal Barrett’s oesophagus.6 Studies have also shown that (Prof H Yang, Prof J Fu);
squamous cell carcinoma and oesophageal adenocarci eradication of Helicobacter pylori influences the preva Department of Radiation
noma, differ in their geographical distributions and lence of oesophageal adenocarcinoma.7 Oncology, Mayo Clinic,
molecular profiles. Treatment strategies typically involve Modifying risk factors, particularly tobacco use, is Rochester, MN, USA
(Prof C L Hallemeier MD);
a sequential regimen of surgery, chemotherapy, radio considered the most effective strategy to address the Department of Thoracic
therapy, and immunotherapy, under the guidance of growing global burden of oesophageal cancer.2 Some Surgery, University Hospital
a multidisciplinary team. The prognosis of oesophageal medications, such as aspirin, statins, and anti-reflux Leuven, Leuven, Belgium
cancer has improved due to the widespread adoption of agents, could potentially reduce oesophageal adenocarci (Prof T Lerut MD PhD)
combined modality therapies and immune checkpoint noma risk; selenium and other micronutrients could Correspondence to:
Prof Jianhua Fu, Department of
inhibitors. lower the risk of oesophageal squamous cell carcinoma. Thoracic Surgery, Sun Yat-sen
However, the existing evidence supporting these inter University Cancer Center,
Epidemiology, risk factors, and prevention ventions for the primary prevention of oesophageal Guangzhou 510060, China
According to the 2022 Global Cancer Observatory cancer is not yet robust enough for widespread [email protected]
Genomic alterations have been identified as key older than 50 years, White race, tobacco smoking,
drivers of oesophageal carcinoma initiation, progres obesity, and a family history of Barrett’s oesophagus or
sion, and metastasis (figure). Inactivation of aldehyde oesophageal adenocarcinoma in a first-degree relative.15
dehydrogenase 2 (ALDH2) is strongly associated with an Furthermore, non-endoscopic methods, such as
increased prevalence of oesophageal squamous cell sponge cytology sampling, have been shown to be safe
carcinoma.11 Common alterations in oesophageal and feasible for screening of squamous neoplasia.16 The
squamous cell carcinoma include inactivation of trefoil factor 3 test conducted on the basis of sponge
CDKN2A, amplification of CCND1, and loss of RB1. cytology can help detect and monitor Barrett’s oesopha
However, according to reports from The Cancer Genome gus.17 Risk stratification tools incorporating clinical
Atlas Network, CCND1 amplification occurs in 57% of information, sponge cytology, or both, have been
squamous cell carcinoma, but accounts for only 15% of developed for pre-endoscopy screening of oesophageal
oesophageal adenocarcinoma cases,12 highlighting cell cancer.18 The use of artificial intelligence as an auxiliary
cycle kinases as potential therapeutic targets specific for tool has shown potential in detecting oesophageal
oesophageal squamous cell carcinoma. Conversely, squamous cell carcinoma and precancerous lesions.19,20
oesophageal adenocarcinoma often exhibits frequent Moreover, DNA methylation assays of cytosponge
oncogenic amplification—eg, amplification of HER2 samples show promise for improving diagnostic
(also known as ERBB2) in approximately 32% of accuracy of Barrett’s oesophagus;21 however, further
cases13—which is rarely observed in oesophageal research is essential for broader validation and prospec
squamous cell carcinoma, indicating distinct molecular tive studies.
vulnerabilities. Both oesophageal squamous cell
carcinoma and oesophageal adenocarcinoma show Pathology and molecular typing
alterations in chromatin-modifying enzymes. Oesophageal squamous cell carcinoma and oesophageal
Amplifications of chromosome 3q with genes essential adenocarcinoma are typically considered two distinct
for squamous cell maturation are frequent in oesopha disease entities with different tissues of origin
geal squamous cell carcinoma, whereas oesophageal and molecular hallmarks.2,22 Generally, oesophageal
adenocarcinoma usually exhibits amplifications of squamous cell carcinoma arises from epithelial cells,
GATA4 and GATA6, which are associated with gastric and oesophageal adenocarcinoma arises from glandular
See Online for appendix adenocarcinoma (appendix p 1).12,13 Advances in single- cells in the lower part of the oesophagus.
cell sequencing technologies have also provided novel The molecular taxonomy of oesophageal cancer has
insights into the cancer biology of oesophageal cancer made rapid progress; studies in the last decade have
(appendix p 1). uncovered distinct molecular subtypes that have great
potential for prognosis stratification and treatment
Population screening strategy guidance.23 Molecular biomarkers, such as
Studies published in the last decade underscore endo microsatellite instability-high, mismatch repair defi
scopic screening as a key approach for the early detection ciency, and high tumour mutational burden, are
and timely treatment of oesophageal cancer and its associated with favourable responses to immunother
precursor lesions in populations at high risk.14 The apy.23 Microsatellite instability or mismatch repair
criteria to identify populations at high risk for oesopha testing is recommended for oesophagogastric junction
geal squamous cell carcinoma and oesophageal adenocarcinomas but is not yet standard in oesophageal
adenocarcinoma are quite different. Individuals who are squamous cell carcinoma treatment guidelines.
at high risk for oesophageal squamous cell carcinoma Furthermore, the roles of human epidermal growth
include those aged 40 years and older living in high- factor receptor 2 (HER2), programmed death-ligand 1
incidence areas or those with a history of cancer, a family (PD-L1), and other novel genomic markers, are increas
history of oesophageal cancer, or other relevant risk ingly recognised, providing more diverse and
factors, such as smoking, heavy alcohol consumption, a personalised treatment options for patients with
history of squamous cancer of the head and neck, and a oesophageal cancer than previously available.
preference for very hot or pickled food. For these indi
viduals, chromoendoscopy coupled with biopsy of HER2
suspicious regions is the recommended screening The proportion of patients with HER2-overexpressing
approach to reduce oesophageal squamous cell carci oesophagogastric junction adenocarcinoma ranges
noma-related mortality.14 between 10% and 20% according to different studies.24
For Barrett’s oesophagus and oesophageal adenocarci Patients with HER2-positive oesophageal adenocarci
noma, the American College of Gastroenterology noma—defined as immunohistochemical expression 2+
guidelines advocate a single screening endoscopy for (ie, weak to moderate, complete staining in more than
individuals with chronic gastro-oesophageal reflux 10% of cells) or 3+ (ie, strong, complete membrane
disease symptoms and three or more additional risk staining in more than 10% of cells) combined with
factors for Barrett’s oesophagus, including male sex, age positive fluorescent in-situ hybridisation verification
Middle
thoracic
Oesophagus
30 cm
Lower
thoracic
Healthy epithelium Intraepithelial neoplasia Oesophageal squamous cell carcinoma
40 cm Oesophageal adenocarcinoma
Squamocolumnar
junction
Oesophagogastric
junction
RTK pathway Cell cycle
42 cm • KRAS • CDKN2A
• HER2 • MYC
• EGFR • CCND1
Stomach • PIKSCA • CDK6
Gastrointestinal
development factor
• GATA4
Oncogene • GATA6
Tumour suppressor gene • MUC6
TGF-β Chromatin remodelling
• SMAD4 • ARID1A
• SMARCA4 DNA damage repair Chromatin instability
• ARID2 • TP53 • Hypermethylation
Wnt
• ARID1B • MDM2 • High driving oncogene burden
• APT
(ie, ratio of HER2 copy number and number of chromo proportion score, combined positive score, and tumour
some 17 centromeres within the nucleus ≥2 or average area positivity. Tumour proportion score measures PD-L1
HER2 copy number ≥6)—can benefit from anti-HER2 positivity only in tumour cells, whereas combined
targeted therapies.25 positive score provides a more biologically relevant
assessment by including PD-L1 staining in tumour cells,
PD-L1 lymphocytes, and macrophages (appendix p 10). With its
PD-L1 is one of the most extensively studied biomarkers increasing use in clinical trials, combined positive score
in studies investigating immunotherapy responders. has been endorsed by the National Comprehensive
Currently adopted scoring systems include tumour Cancer Network (NCCN) guidelines and is supported by
the US Food and Drug Administration (FDA), which early-stage oesophageal cancer with increased specificity,
approved PD-L1 immunohistochemical 22C3 pharmDx particularly in the upper oesophagus, where tumours are
(Agilent, Santa Clara, CA, USA) as a surrogate for PD-L1 frequently missed by Lugol’s solution.31
positivity. The widely used eighth edition of the American Joint
Although cutoffs for PD-L1 expression vary across trials, Committee TNM cancer staging system introduces pre
overall survival was consistently prolonged with immune treatment, surgery-alone, and post-treatment
checkpoint inhibitor monotherapy versus chemotherapy pathological TNM stagings to accommodate the use of
as the second-line treatment for patients with high PD-L1 neoadjuvant therapy.32 Endoscopic ultrasound is
expression. However, first-line treatment trials reported a powerful tool to detect tumour invasion depth and
disputable results regarding the predictive value of PD-L1 lymph node metastasis.33,34 One study proposed that
expression for chemoimmunotherapy. As presented in submucosal saline injection can increase the diagnostic
the KEYNOTE-590 and CheckMate-648 trials, patients accuracy of endoscopic ultrasound in discriminating T1a
with high PD-L1 expression had better overall survival (ie, tumours invading the lamina propria or muscularis
with PD-1 inhibitors plus chemotherapy compared with mucosae) and T1b (ie, tumours invading the submucosa)
chemotherapy alone. Conversely, the benefit was absent tumours from 66% to 94%.35 Moreover, endoscopic
in individuals with low PD-L1 expression.26,27 mucosal resection aids diagnosis of T1b tumours. MRI
Furthermore, the post-hoc analysis of the JUPITER-06 is valuable for detecting tumour invasion to adjacent
trial and meta-analysis supported the superiority of organs (T4a and T4b),36 whereas endobronchial ultra
PD-1 antibodies plus chemotherapy over chemotherapy sound can facilitate the diagnosis of airway invasion
alone in patients with low PD-L1 expression.28 These with high sensitivity and specificity,33,37 especially for
findings challenge the notion that PD-L1 expression is tumours situated at the cervical, thoracic superior, and
an unequivocal biomarker for chemoim muno therapy middle segments of the oesophagus. Both CT and
response in oesophageal squamous cell carcinoma, PET-CT, despite their wide use, show poor performance
underscoring the necessity for more reliable molecular for primary tumour staging, with accuracies below 50%.38
typing biomarkers. Antibodies for other therapeutic CT and PET-CT might complement endoscopic ultra
targets, such as claudin-18 isoform 2 (CLDN18.2), show sound in the detection of lymph node metastasis with
promising potential in treating oesophageal adenocarci overall sensitivities of 50% and 57%, and specificities
noma, as evidenced by the SPOTLIGHT and GLOW of 83% and 85% respectively.39 PET-CT is useful for
trials (appendix p 2).29,30 Novel therapeutic targets, evaluation of systemic metastasis, although it has less
including FGFR, and molecular classifications based on clinical value in staging early oesophageal cancer.40
multi-omics technologies are also discussed in the Ultrasound shows high accuracy in diagnosing nodal
appendix (p 2). involvement in the neck, which can facilitate aspiration
biopsy. However, one meta-analysis reported that these
Symptoms and signs of oesophageal cancer imaging techniques are insufficiently accurate for
Because of the inaccessibility of the oesophagus to staging after neoadjuvant treatment.41
clinical examination, obtaining a detailed history from
the patient is of paramount importance. Treatment principles
The most common symptom of oesophageal cancer is On diagnosis, patients should undergo comprehensive
difficulty swallowing (dysphagia). As the oesophageal assessments to determine pathological subtype, TNM
tumour grows, it might progressively impede the passage stage, tumour location, and performance status,
of solid and subsequently semisolid food, and liquids in including cardiorespiratory fitness and treatment toler
later stages. Dysphagia typically manifests insidiously ability. Optimal therapeutic strategies vary by TNM stage
and becomes apparent when the tumour has infiltrated and substantially affect prognosis, making stage-specific
about two-thirds of the circumference of the oesophagus, therapy recommendations essential for both oesophageal
often indicating locally advanced disease. Other squamous cell carcinoma and oesophageal adenocarci
commonly observed symptoms include heartburn and noma patients (table 1).
unintended weight loss (appendix p 3). Superficial oesophageal cancer denotes tumours
limited to the submucosal layer without lymph node
Diagnostic and staging considerations metastasis. Optimal treatment for T1a tumours is endo
As with other solid tumours, therapy recommendations scopic therapy, such as endoscopic mucosal resection or
for oesophageal cancer depend on accurate diagnosis endoscopic submucosal dissection. Radiofrequency
and staging. For symptomatic patients, radioscopy radi ablation is also used for treatment of dysplasia or very
ography with barium swallow contrast helps to identify early-stage adenocarcinoma in high-income countries.57
cancer location and obstruction severity. Endoscopy is However, the role of endoscopic submucosal dissection
performed to ascertain lesion size and location and to in managing T1b oesophageal cancer remains under
obtain tissue for pathological diagnosis. Additionally, debate, given that nodal involvement is observed in
narrow-band imaging has improved the detection of 16·6% of patients, nearly three-fold the rate in
Optimal treatment
cTis-1aN0M0
Oesophageal squamous cell Endoscopic submucosal dissection or endoscopic mucosal resection or endoscopic radiofrequency ablation
carcinoma
Oesophageal adenocarcinoma Endoscopic submucosal dissection or endoscopic mucosal resection or endoscopic radiofrequency ablation
cT1b-2N0M0
Oesophageal squamous cell Oesophagectomy options include two-field lymphadenectomy including recurrent laryngeal nerve lymph node, minimally
carcinoma invasive oesophagectomy,42–44 or robot-assisted minimally invasive oesophagectomy,45 rather than open surgery; adjuvant
chemotherapy if patients staged as pN1–3;46 postoperative radiation for patients without R0 resection
Oesophageal adenocarcinoma Oesophagectomy options include two-field lymphadenectomy including recurrent laryngeal nerve lymph node, minimally
invasive oesophagectomy,42–44 or robot-assisted minimally invasive oesophagectomy,45 rather than open surgery; adjuvant
chemotherapy if patients staged as pN1–3;46 postoperative radiation for patients without R0 resection
cT1-4aN1-3M0 or cT3-4aN0M0
Oesophageal squamous cell Neoadjuvant chemoradiotherapy and surgery (paclitaxel and platinum and 41·4 Gy radiation)47,48 or neoadjuvant triplet
carcinoma chemotherapy (fluorouracil, platinum, and docetaxel);49 adjuvant single-agent immune checkpoint inhibitors for patients
without pathological complete response after neoadjuvant therapy;50 postoperative radiation for patients without R0
resection
Oesophageal adenocarcinoma Perioperative chemotherapy (fluorouracil, leucovorin, oxaliplatin, and docetaxel)51 or neoadjuvant chemoradiotherapy and
surgery (paclitaxel and platinum and 41·4 Gy radiation);48 adjuvant single-agent immune checkpoint inhibitors for patients
without pathological complete response after neoadjuvant therapy;50 postoperative radiation for patients without R0
resection
cT4bN0-3M0
Oesophageal squamous cell Definitive chemoradiotherapy (50·4 Gy radiation, FOLFOX or paclitaxel and platinum);52,53 surgery for persistent and recurrent
carcinoma disease54,55
Oesophageal adenocarcinoma Definitive chemoradiotherapy (50·4 Gy radiation, FOLFOX or paclitaxel and platinum);52,53 surgery for persistent and recurrent
disease54,55
cTanyNanyM1
Oesophageal squamous cell For patients with low PD-L1 expression, chemoimmunotherapy combining paclitaxel, platinum, and immune checkpoint
carcinoma inhibitors remains a recommended approach;28 however, the survival benefit in this group is less pronounced than individuals
with high PD-L1 expression*
Oesophageal adenocarcinoma If HER2-positive and PD-L1-high, anti-HER2 trastuzumab and chemoimmunotherapy (fluorouracil and cisplatin and immune
checkpoint inhibitors);56 if HER2-negative and PD-L1-high, chemoimmunotherapy (fluorouracil and cisplatin and immune
checkpoint inhibitors); if HER2-negative and PD-L1-low: chemotherapy (fluorouracil and cisplatin) and immune checkpoint
inhibitors†
Pathological diagnosis for staging is by endoscopic biopsy, with endoscopic ultrasound to diagnose tumour (T) stage, endoscopic ultrasound or CT to diagnose node (N)
stage, and PET-CT to diagnose metastasis (M) stage. For molecular diagnosis, PD-L1 testing is used for oesophageal squamous cell carcinoma. For oesophageal
adenocarcinoma, PD-L1 and HER2 is used, as well as microsatellite, tumour mutation burden, CLDN18, and FGFR2b testing. FOLFOX=leucovorin, fluorouracil, and oxaliplatin.
*The cutoff of high expression of PD-L1 varies across clinical trials; commonly seen criteria include combined positive score of at least 10 or tumour area positivity of at least
1. †The US Food and Drug Administration approved the use of immune checkpoint inhibitors combined with chemotherapy in patients with a combined positivity score less
than 5, whereas the European Medicines Agency did not approve its use for patients with a combined positivity score less than 5.
Table 1: Standard treatment for different stages of the two subtypes of oesophageal cancer
T1a disease.58 Therefore, current evidence and the NCCN In patients with T4 tumours encroaching on adjacent
guidelines prefer oesophagectomy in most patients with organs, including the lung, bronchus, azygos vein, pre
T1b oesophageal cancer. vertebral fascia, inferior pulmonary vein, and
For oesophageal cancer invading the muscular layer pericardium, induction therapies are still encouraged to
without other metastases (ie, T2N0M0), oesophagectomy render borderline-resectable tumours potentially
with lymph node dissection is the standard treatment.34 operable. Compared with chemoradiotherapy alone,
For locally advanced disease characterised by high tumour radical surgery might improve the prognosis for these
burden and nodal involvement (ie, T1-4N1-3M0), pre patients.62 Moreover, for patients who have persistent
operative treatment, including chemotherapy and disease or recurrence after definitive chemoradiotherapy,
chemoradiotherapy, is recommended.47,48,59–61 Neoadjuvant salvage oesophagectomy can have a favourable prognosis
chemoradiotherapy is the preferred scheme for oesophageal if the tumour is resectable.54 Definitive chemoradiother
squamous cell carcinoma based on existing literature,47,48 apy is the primary option for non-surgical candidates
and both preoperative and perioperative chemotherapies with cervical segment oesophageal cancer, supraclavicu
are endorsed for oesophageal adenocarcinoma.59–61 For lar or retroperitoneal lymph node metastasis, or an
those who receive neoadjuvant chemoradiotherapy intolerance for or refusal of surgery. For patients afflicted
plus oesophagectomy without a pathological complete with distant metastasis, systemic therapy should be indi
response, adjuvant therapy with the anti-PD-1 antibody vidualised in the context of performance status and
nivolumab can reduce recurrence risk.50 tumour genetics.
for patients with oesophageal squamous cell carcinoma survival for those who received surgery alone was
(appendix p 6).78 From this perspective, the McKeown 21·1 months, whereas the median overall survival of
oesophagectomy might provide a more comprehensive patients in the chemoradiotherapy group was
lymphadenectomy for the bilateral recurrent laryngeal 81·6 months (HR 0·48, 95% CI 0·28–0·83, p=0·008).
nerve than the Ivor Lewis procedure does. However, the Moreover, compared with surgery alone, preoperative
McKeown procedure poses an increased risk of post chemoradiotherapy did not adversely affect short-term or
operative complications, such as recurrent laryngeal long-term health-related quality of life.93
nerve injury and anastomotic leakage. Further studies In another study, NEOCRTEC5010, 451 patients with
are warranted to compare the two procedures. oesophageal squamous cell carcinoma were randomly
assigned to surgery alone or neoadjuvant chemoradio
Adenocarcinoma therapy followed by surgery. The chemotherapy regimen
The extent of lymphadenectomy for oesophageal adeno of vinorelbine and cisplatin were administered every
carcinoma remains a crucial topic of academic debate. three weeks for two cycles delivered concurrently with a
Transthoracic oesophagectomy with en-bloc (ie, removed radiotherapy regimen of 40 Gy in 20 fractions.47,90
with surrounding adipose tissue) two-field lymphadenec Participants treated with surgery alone had a median
tomy stands as a prevailing consensus, with the NCCN overall survival of 66·5 months, compared with
guidelines recommending resection of at least 15 lymph 100·1 months for participants in the chemoradiotherapy
nodes for adenocarcinoma.34 The optimal extent and group (HR 0·71 [95% CI 0·53–0·96]; p=0·025).90
locations for lymph node dissection are still being inves Building on the NEOCRTEC5010 trial, another study
tigated by the ongoing TIGER trial.81 In the context of showed that neoadjuvant chemoradiotherapy plus
neoadjuvant therapy, some studies underscore the surgery is a cost-effective strategy.94 These results have
importance of dissecting an adequate number of lymph led to the adoption of neoadjuvant chemoradiotherapy
nodes.82 The effects of surgery occuring mostly in high- as the standard of care for numerous patients with
volume hospitals and cancer centres are discussed in the locally advanced oesophageal squamous cell carcinoma
appendix (p 6). scheduled for oesophagectomy.
Chemotherapy is another neoadjuvant alternative.
Neoadjuvant therapy The JCOG9907 trial showed that preoperative chemo
In the last two decades, neoadjuvant therapy has greatly therapy (ie, cisplatin plus fluorouracil) offered superior
reshaped the treatment landscape of locally advanced overall survival than postoperative chemotherapy in
oesophageal cancer (table 2). A meta-analysis of oesophageal squamous cell carcinoma.83 As for direct
26 randomised trials revealed that, compared with comparison between the neoadjuvant chemoradio
surgery alone, both neoadjuvant chemotherapy and therapy and chemotherapy, the CMISG1701 trial
chemoradiotherapy improved overall survival (hazard reported a 9% non-significant increase of 3-year overall
ratio [HR] for chemotherapy 0·86, 95% CI 0·75–0·99, survival rate with the addition of radiotherapy with neo
p=0·03; HR for chemoradiotherapy 0·77, 0·68–0·87, adjuvant chemotherapy (cisplatin and paclitaxel) in
p<0·001).88 According to the findings of the CROSS and cT3-4aN0-1M0 oesophageal cancer;87 the conclusions of
NEOCRTEC5010 trials, preoperative chemoradiotherapy this study might be further compromised due to the
plus surgery is recommended as a standard treatment for small sample size and the exclusion of patients who
locally advanced oesophageal cancer.89,90 To date, potential were staged as cN2–3.
candidates for neoadjuvant therapy are patients with The three-arm JCOG1109 trial49 found that neoadjuvant
stage T1-4N1-3M0 disease. For patients with T1b-T2N0M0 platinum triplet therapy (docetaxel plus cisplatin plus
tumours (ie, tumours invading the muscular layer fluorouracil) significantly improved median overall
without other metastases), the role of neo adjuvant survival compared with the cisplatin plus fluorouracil
therapy is not clear; the FFCD9901 trial showed that pre regimen (docetaxel plus cisplatin plus fluorouracil overall
operative chemoradiotherapy did not improve R0 survival not reached vs cisplatin plus fluorouracil
resection rate and survival, but exacerbated postoperative overall survival of 5·6 years; HR 0·68 [95% CI 0·50–0·92];
mortality in these patients.91 Restaging before p=0·006). There was no statistically significant difference
oesophagectomy is recommended for all patients taking regarding median overall survival between the cisplatin
neoadjuvant treatment.92 plus fluorouracil group (5·6 years) and the chemoradio
therapy group (7·0 years; HR 0·84 [0·63–1·12]; p=0·12).
Squamous cell carcinoma The docetaxel plus cisplatin plus fluorouracil group had
The CROSS randomised controlled trial evaluated the higher rates of grade 3 or 4 haematological adverse
efficacy of a regimen involving five cycles of weekly events—ie, 167 (85%) of 196 patients with neutropenia
chemotherapy (carboplatin and paclitaxel) combined and 32 (16%) of 196 patients with febrile neutropenia.
with concurrent radiotherapy (41·4 Gy over Regarding cause of mortality, the chemoradiotherapy
23 fractions).48,84,89 Of 84 patients diagnosed with oeso group had higher rates of non-cancer-related
phageal squamous cell carcinoma, the median overall deaths (ie, 23 [26%] of 89 patients) than the docetaxel plus
Table 2: Key randomised clinical trials of preoperative or perioperative therapy for resectable oesophageal cancer
cisplatin plus fluorouracil group (ie, seven [9%] of oesophagus and enhance health-related quality of life
74 patients) and the cisplatin plus fluorouracil (appendix p 7).
group (ie, 13 [13%] of 98 patients). This finding might
partly be due to the prophylactic irradiation of the middle Adenocarcinoma
and lower mediastinal lymph nodes, which might increase Since the 1980s, neoadjuvant chemotherapy has become
toxicity. the standard for managing oesophageal adenocarcinoma,
Given the high pathological complete response rates primarily using platinum–fluoropyrimidine
with neoadjuvant chemoradiotherapy, especially in combinations. Advancements have emerged from
oesophageal squamous cell carcinoma populations, three pivotal randomised controlled trials exploring
investigators have explored the potential of a so-called preoperative chemotherapy (OEO2 trial) and
watch-and-wait approach for patients who respond to perioperative chemotherapy (MAGIC and FNCLCC–
treatment. This approach aims to preserve the FFCD trials), which showed improved survival
outcomes.59–61 The FLOT4 trial attempted to identify the radiotherapy, the role of postoperative radiotherapy and
optimal drug combinations between the fluorouracil, chemoradiotherapy has diminished in individuals
leucovorin, oxaliplatin, plus docetaxel (ie, FLOT) regimen receiving radical resection. However, patients with
and the pharmorubicin, cisplatin, plus either fluorouracil positive margins should be considered for postoperative
or capecitabine (ie, ECF/ECX) regimen; the trial showed chemoradiotherapy if radiotherapy was not administered
a statistically significant 9% increase in both the 3-year preoperatively.
and 5-year overall survival rates in patients with Single-agent immune checkpoint inhibitors are
adenocarcinoma who received FLOT.51 Preoperative appealing options in the adjuvant setting due to their
chemoradiotherapy is also a standard approach for locally reduced toxicity compared with cytotoxic chemotherapy.
advanced oesophageal adenocarcinoma. In the subgroup The CheckMate-577 trial showed efficacy of a 12-month
analysis of 275 patients with adenocarcinoma from the nivolumab course in patients with residual disease after
CROSS trial, chemoradiotherapy significantly improved neoadjuvant chemoradiotherapy and oesophagectomy.50
overall survival (HR 0·73 [95% CI 0·55–0·98]; p=0·038), Compared with placebo, adjuvant nivolumab signifi
translating to an approximate 10% increase in 5-year and cantly improved disease-free survival (HR 0·69, 95% CI
10-year overall survival rates.48,84,89 0·56–0·86, p<0·001) with manageable toxicity and low
The comparative effectiveness of neoadjuvant chemo impact on health-related quality of life,50 and is now
radiotherapy and chemotherapy has emerged as a focal recommended in this setting. Future analysis of the
topic in oesophageal adenocarcinoma in the last decade. mature overall survival data will further confirm the role
The Neo-AEGIS study compared perioperative chemo of adjuvant immune checkpoint inhibitors in
therapy using the adjusted MAGIC regimen (epirubicin patients with oesophageal cancer receiving trimodality
plus cisplatin or oxaliplatin plus fluorouracil or capecit treatments.
abine) with that of neoadjuvant chemoradiotherapy used
in the CROSS regimen.85 Despite the higher rates of Radiotherapy
pathological complete response, major pathological For over a century, radiotherapy has been a foundational
response, and R0 resection in the neoadjuvant chemora treatment for oesophageal cancer. In the 1920s, Vinson
diotherapy group, median overall survival did not and colleagues pioneered the use of brachytherapy,
significantly differ between the two groups (neoadjuvant placing radium radioisotopes in proximity to the
chemoradiotherapy 48·0 months vs perioperative chem tumours. Modern clinics have used increasingly sophisti
otherapy 49·2 months; HR 1·03, 0·77–1·38, p=0·82) cated external-beam radiotherapy techniques, which
and 3-year overall survival rates were 55% in the neoad have been refined over the past three decades through
juvant chemoradiotherapy group versus 57% in the the evolution of computer-based treatment planning and
perioperative chemotherapy group. The ongoing delivery technologies.
ESOPEC trial is further exploring this comparison For patients with oesophageal cancer ineligible for
(NCT02509286). surgical resection, the standard treatment is definitive
The advent of immune checkpoint inhibitors marks chemoradiotherapy, which consists of external-beam
a new frontier in cancer therapy. Preliminary results radiotherapy (50·0–50·4 Gy in 1·8–2 Gy fractions over
from phase 1/2 trials have shown promising pathological 5–6 weeks) and concurrent chemotherapy. Common
complete response rates, particularly in oesophageal regimens include biweekly fluorouracil and oxaliplatin
squamous cell carcinoma, when immune checkpoint either with or without leucovorin (FOLFOX) or weekly
inhibitors are combined with neoadjuvant therapies carboplatin and paclitaxel.52,53 Initially reported in 1992,
(appendix p 7). Several randomised phase 3 studies are the RTOG 8501 multi-institutional phase 3 randomised
currently under way, including NEOCRTEC2101 controlled trial showed superior efficacy of chemoradio
(NCT05357846), iCROSS (NCT04973306), KEYSTONE- therapy (50 Gy in 25 fractions with concurrent cisplatin
002 (NCT04807673), JUPITER-14 (NCT04848753), and fluorouracil chemotherapy) compared with radio
NCT03221426, NCT03604991, and NCT02743494. therapy alone (64 Gy in 32 fractions) for patients with
localised oesophageal carcinoma.96 Chemoradiotherapy
Adjuvant therapy resulted in reduced local and distant recurrence, with
Postoperative chemotherapy is a key treatment for a 2-year overall survival rate of 38%, compared with the
patients with oesophageal adenocarcinoma. For oeso rate of 10% observed in patients treated with radiotherapy
phageal squamous cell carcinoma, postoperative alone (p<0·001), although chemoradiotherapy resulted
chemotherapy, radiotherapy, and chemoradiotherapy are in more treatment-related adverse events.
common. For patients receiving surgery alone, one pre Subsequent phase 3 randomised controlled trials have
vailing notion suggests that adjuvant chemotherapy or investigated whether escalating the external-beam radio
chemoradiotherapy are potentially only beneficial for therapy dose from 50 Gy to 60–65 Gy with concurrent
node-positive oesophageal squamous cell carcinoma chemotherapy could improve prognosis, but these
populations, as presented in meta-analyses and clinical yielded negative results.52,97 For example, in the
trials.46,95 With the widespread use of preoperative ARTDECO trial conducted in the Netherlands, patients
with locally advanced oesophageal cancer were randomly first-line treatment with fluoropyrimidines (such as
assigned to receive either standard-dose external-beam capecitabine or fluorouracil), platinum agents (such as
radiotherapy (50·4 Gy in 28 fractions) or escalated-dose cisplatin or oxaliplatin), and PD-1 inhibitors has been
external-beam radiotherapy (61·6 Gy in 28 fractions), shown to improve clinical outcomes over chemotherapy
along with concurrent weekly carboplatin and paclitaxel alone, especially in patients with high PD-L1 expression,
chemotherapy. The 3-year local progression-free survival based on the results from CheckMate-649, ORIENT-16,
rates were 70% for the standard-dose group and 73% for and RATIONALE-305.106,108,110 However, whether the
the escalated-dose group, and the overall survival rates benefit of anti-PD-1 antibodies can be extended to patients
were 42% for the standard and 39% for the escalated.52 with low or unknown PD-L1 expression remains
Other topics, including novel radiotherapy techniques, controversial; thus, treatment decisions for these patients
palliative radiotherapy, and oesophagus-preservation should be carefully considered. For HER2-positive
approaches, using definitive chemoradiotherapy are oesophagogastric junction adenocarcinoma, the ToGA
discussed in the appendix (p 8). trial showed that addition of the anti-HER2 antibody
trastuzumab to chemotherapy improved overall survival,
Systemic therapy progression-free survival, and overall response rate.25
First-line systemic therapy Additionally, for HER2-positive oeso phagogastric
The goals of systemic therapy in patients with advanced junction adenocarcinoma accompanied by a combined
oesophageal cancer are to relieve symptoms, improve positive score of at least 1, pembrolizumab, combined
health-related quality of life, and prolong survival. Data with trastuzumab and chemotherapy, was approved by
have evidenced the survival benefit of systemic chemo the US FDA as a first-line treatment.111 For patients with
therapy for patients with advanced oesophageal cancer.98,99 mismatch repair deficiency or microsatellite instability-
Platinum-based combination regimens are the standard high oesophagogastric junction adenocarcinoma,
first-line therapy for oesophageal cancer. However, irrespective of PD-L1 expression, immunotherapy plus
advancements in the last decade in immune checkpoint systemic chemotherapy or pembrolizumab monotherapy
inhibitors have brought hope for potential cures for are both acceptable first-line treatments.
patients with advanced disease.
Chemoimmunotherapy has become a new standard Second-line and subsequent systemic therapy
first-line treatment for inoperable or metastatic oesopha Docetaxel, paclitaxel, or irinotecan are the recommended
geal cancer, including oesophageal squamous cell second-line and subsequent systemic treatments.112
carcinoma and oesophageal adenocarcinoma. Findings Owing to the development of immune checkpoint
from phase 3 clinical trials reported improved patient inhibitors in the last decade, therapeutic options have
survival when PD-1 inhibitors are added to chemotherapy diversified and largely depend on patients’ previous
(table 3).27,100–110 The selection of chemotherapy drugs and treatment history of immunotherapy in first-line therapy.
immune checkpoint inhibitors varies across different For patients with oesophageal squamous cell carcinoma
regions and patient conditions. In the USA and European without previous immunotherapy, PD-1 inhibitors, such
countries, the combination of cisplatin or oxaliplatin as pembrolizumab, nivolumab, and tislelizumab, have
with fluorouracil is preferred as the front-line chemo shown superior efficacy to standard chemotherapy in
therapy for treating adenocarcinoma. However, in Asian second-line treatment, according to the KEYNOTE-181,
countries where oesophageal squamous cell carcinoma ATTRACTION-3, and RATIONALE-302 trials.113–115 For
is more prevalent, particularly in China, the combination patients with advanced oesophagogastric junction ade
of paclitaxel and cisplatin is favoured, as evidenced by nocarcinoma who progress on previous chemotherapy
several first-line studies focused on oesophageal with or without trastuzumab, optional regimens include
squamous cell carcinoma. Currently, the overall impact ramucirumab plus paclitaxel, the FOLFIRI (ie, fluoro
of these distinct chemotherapy regimens on treatment uracil, leucovorin, and irinotecan) regimen with or
efficacy remains unclear and warrants further research. without ramucirumab, and single-agent chemotherapy.
Moreover, the diversity in the choice of PD-1 inhibitor In addition, for HER2-positive advanced oesophagogas
presents a challenge, as there are no head-to-head clinical tric junction adenocarcinoma, numerous innovative
trials directly comparing the specific efficacies of various HER2-targeted agents, such as margetuximab, trastu
PD-1 inhibitors. Thus, treatment decisions for specific zumab deruxtecan, and tucatinib, which are potentially
chemotherapy regimens and PD-1 inhibitors in patients more effective at inhibiting the HER2 pathway, have
with advanced oesophageal squamous cell carcinoma been investigated in late-line settings.
should be considered on a patient-specific basis.
Owing to disparities in tumour biology and molecular Immunotherapy for oesophageal cancer
profiles, the treatment of advanced oesophagogastric Immunotherapy, mainly immune checkpoint blockades,
junction adenocarcinoma differs slightly from have become prominent and effective treatments for
that of oesophageal squamous cell carcinoma. For HER2- oesophageal cancer. Following these advancements, we
negative oesophagogastric junction adenocarci noma, discuss several themes, including the chemotherapy
Table 3: Summary of the key phase 3 clinical trials of immune checkpoint inhibitors in advanced oesophageal cancer
regimen, emerging novel immune checkpoint inhibi Palliative treatment and best supportive care
tors, and biomarkers of immunotherapy responses Despite advances in the screening and treatment of
(appendix p 9). oesophageal cancer, a considerable proportion of patients
remain ineligible for curative treatment, and more than
Follow-up and recurrence surveillance half of patients treated with curative intent will inevitably
Given the high recurrence potential of oesophageal have uncontrolled tumour recurrence.47 Palliative therapy
cancer, ongoing surveillance is essential for timely for the purpose of relieving suffering and improving the
treatment on recurrence. Although a previous study has health-related quality of life of patients is often necessary.
established the survival benefits of regular radiological Patients with extensive distant metastasis and poor per
monitoring,116 the optimal surveillance strategy for formance status, especially those with a short life
patients with oesophageal cancer with successful therapy expectancy, are typically candidates for palliative therapy
remains undefined. Typically, follow-up schedules last (appendix p 11).
5 years after treatment, as recurrence beyond this period
is relatively rare in patients with oesophageal cancer.117 Nutritional support
Intensified surveillance should be performed within the Compared with other cancers, patients with oesophageal
first 2 years after treatment since most recurrences occur cancer have a greater risk of malnutrition, because
during this timeframe (appendix p 11). dysphagia exacerbates during disease progression.117
Contributors
Panel: Remaining research questions on oesophageal cancer Conceptualisation: JF and HY. Data curation: HY, FW, CLH, and TL.
Writing (original draft): HY, FW, CLH, and TL. Visualisation:
• What are the genetic determinants in oesophageal cancer development, progression, HY and FW. Funding acquisition: JF and HY. Supervision: JF.
and recurrence?
Declaration of interests
• What is the effective screening approach for individuals with high risk of oesophageal CLH has received travel support to attend the National Comprehensive
cancer? Cancer Network Neuroendocrine and Adrenal Tumor Panel annual
• What is the standard lymph node dissection for oesophageal adenocarcinoma? meeting, the ECOG-ACRIN Data Safety Monitoring Committee annual
meeting, and ASTRO-Veteran’s Health Administration Radiation
• Does immunotherapy in the neoadjuvant setting provide survival benefits? Oncology Quality Surveillance Program–Liver Cancer Panel outside the
• How can we enhance the diagnostic accuracy of clinical complete response after submitted work; and served as a member of the ECOG-ACRIN Data
neoadjuvant therapy? Safety Monitoring Committee and the National Cancer Institute Data
• Is the so-called watch-and-wait strategy appropriate for squamous cell carcinoma Safety Monitoring Board, Imaging and Radiation Therapy Committee
outside the submitted work. All other authors declare no competing
patients with clinical complete response? interests.
• What is the optimal treatment for patients with pathological complete response after
Acknowledgments
neoadjuvant therapy and surgery? Should the treatment be observation or adjuvant We thank Mian Xi (Department of Radiation Oncology, Sun Yat-sen
immunotherapy? University Cancer Center, China) for instruction on the writing of the
• Is adjuvant immunotherapy appropriate for patients who received perioperative radiotherapy section. We thank Changsen Leng (Department of Thoracic
chemotherapy plus surgery? Surgery, Sun Yat-sen University Cancer Center, China) for assisting with
preparation of figure materials. We thank Zihang Mai (Department of
• Is adjuvant immunotherapy appropriate for patients who received surgery alone with Thoracic Surgery, Sun Yat-sen University Cancer Center, China) for
postoperative upstaging? assistance with writing and revising the manuscript and preparing the
• How long should adjuvant immunotherapy last after surgery? data materials.
• Can the addition of immunotherapy into definitive chemoradiotherapy for one year or References
longer improve disease control? 1 Bray F, Laversanne M, Sung H, et al. Global cancer statistics 2022:
GLOBOCAN estimates of incidence and mortality worldwide for
• How can we design individualised combination therapies for patients with advanced 36 cancers in 185 countries. CA Cancer J Clin 2024; 74: 229–63.
disease? 2 Morgan E, Soerjomataram I, Rumgay H, et al. The global landscape
of esophageal squamous cell carcinoma and esophageal
adenocarcinoma incidence and mortality in 2020 and projections to
2040: new estimates from GLOBOCAN 2020. Gastroenterology 2022;
Studies have stressed the roles of active nutritional inter 163: 649–58.
vention in reducing treatment-related complications and 3 Rumgay H, Arnold M, Laversanne M, et al. International trends in
improving treatment outcomes.119,120 Therefore, nutri esophageal squamous cell carcinoma and adenocarcinoma
incidence. Am J Gastroenterol 2021; 116: 1072–76.
tional assessment and support are essential throughout 4 Lin Y, Wang HL, Fang K, Zheng Y, Wu J. International trends in
the whole course of cancer treatment. As for the nutrition esophageal cancer incidence rates by histological subtype
pathway, enteral feeding is the primary choice (appendix (1990–2012) and prediction of the rates to 2030. Esophagus 2022;
19: 560–68.
p 12). 5 Thun M, Linet MS, Cerhan JR, Haiman CA, Schottenfeld D. Cancer
Epidemiology and Prevention. Oxford University Press, 2017.
Conclusion 6 Coleman HG, Xie SH, Lagergren J. The epidemiology of esophageal
Oesophageal cancer remains a formidable global adenocarcinoma. Gastroenterology 2018; 154: 390–405.
7 Arnold M, Laversanne M, Brown LM, Devesa SS, Bray F. Predicting
health challenge, with substantial mortality rates the future burden of esophageal cancer by histological subtype:
despite advancements in treatment modalities. international trends in incidence up to 2030. Am J Gastroenterol
Multidisciplinary approaches, personalised therapies, 2017; 112: 1247–55.
8 Jankowski JAZ, de Caestecker J, Love SB, et al. Esomeprazole and
and innovative technologies have improved patient aspirin in Barrett’s oesophagus (AspECT): a randomised factorial
outcomes, including enhanced survival and quality of trial. Lancet 2018; 392: 400–08.
life. However, further research is needed to uncover 9 Thrift AP. Global burden and epidemiology of Barrett oesophagus
and oesophageal cancer. Nat Rev Gastroenterol Hepatol 2021;
the underlying causes, genetic determinants, and prog 18: 432–43.
nostic indicators of oesophageal cancer. Cancer 10 Kamangar F, Nasrollahzadeh D, Safiri S, et al. The global, regional,
screening, diagnosis, and treatment are crucial to and national burden of oesophageal cancer and its attributable risk
improve patient outcomes, emphasising the need for factors in 195 countries and territories, 1990–2017: a systematic
analysis for the Global Burden of Disease Study 2017.
continuous efforts in these areas (panel). Modifications Lancet Gastroenterol Hepatol 2020; 5: 582–97.
in surgical techniques have the potential to improve 11 Suo C, Yang Y, Yuan Z, et al. Alcohol intake interacts with
perioperative outcomes and postoperative quality of functional genetic polymorphisms of aldehyde dehydrogenase
(ALDH2) and alcohol dehydrogenase (ADH) to increase esophageal
life. The efficacy of immunotherapies, combined with squamous cell cancer risk. J Thorac Oncol 2019; 14: 712–25.
standard treatments, particularly in neoadjuvant 12 Cancer Genome Atlas Research Network. Integrated genomic
settings, necessitates rigorous evaluation through characterization of oesophageal carcinoma. Nature 2017; 541: 169–75.
13 Frankell AM, Jammula S, Li X, et al. The landscape of selection in
extensive clinical trials. With the dedication of 551 esophageal adenocarcinomas defines genomic biomarkers for
researchers, ongoing progress in understanding the clinic. Nat Genet 2019; 51: 506–16.
oesophageal cancer holds the potential to transform it 14 Wei WQ, Chen ZF, He YT, et al. Long-term follow-up of
a community assignment, one-time endoscopic screening study of
from a life-threatening malignancy into a manageable esophageal cancer in China. J Clin Oncol 2015; 33: 1951–57.
disease.
15 Shaheen NJ, Falk GW, Iyer PG, et al. Diagnosis and management of 34 Ajani JA, D’Amico TA, Bentrem DJ, et al. Esophageal and
Barrett’s esophagus: an updated ACG guideline. Am J Gastroenterol esophagogastric junction cancers, version 2.2023, NCCN clinical
2022; 117: 559–87. practice guidelines in oncology. J Natl Compr Canc Netw 2023;
16 Middleton DRS, Mmbaga BT, O’Donovan M, et al. Minimally 21: 393–422.
invasive esophageal sponge cytology sampling is feasible in 35 He L-J, Xie C, Wang Z-X, et al. Submucosal saline injection
a Tanzanian community setting. Int J Cancer 2021; 148: 1208–18. followed by endoscopic ultrasound versus endoscopic ultrasound
17 Fitzgerald RC, di Pietro M, O’Donovan M, et al. Cytosponge-trefoil only for distinguishing between T1a and T1b esophageal cancer.
factor 3 versus usual care to identify Barrett’s oesophagus in Clin Cancer Res 2020; 26: 384–90.
a primary care setting: a multicentre, pragmatic, randomised 36 Qu J, Zhang H, Wang Z, et al. Comparison between free-breathing
controlled trial. Lancet 2020; 396: 333–44. radial VIBE on 3-T MRI and endoscopic ultrasound for preoperative
18 Gao Y, Xin L, Lin H, et al. Machine learning-based automated T staging of resectable oesophageal cancer, with histopathological
sponge cytology for screening of oesophageal squamous cell correlation. Eur Radiol 2018; 28: 780–87.
carcinoma and adenocarcinoma of the oesophagogastric junction: 37 Nishino T, Toba H, Yoshida T, et al. Endobronchial ultrasound
a nationwide, multicohort, prospective study. improves the diagnosis of the tracheobronchial invasion of
Lancet Gastroenterol Hepatol 2023; 8: 432–45. advanced esophageal cancer. Ann Surg Oncol 2021; 28: 6398–406.
19 Yuan XL, Liu W, Lin YX, et al. Effect of an artificial intelligence- 38 Tangoku A, Yamamoto Y, Furukita Y, Goto M, Morimoto M.
assisted system on endoscopic diagnosis of superficial oesophageal The new era of staging as a key for an appropriate treatment for
squamous cell carcinoma and precancerous lesions: a multicentre, esophageal cancer. Ann Thorac Cardiovasc Surg 2012; 18: 190–99.
tandem, double-blind, randomised controlled trial. 39 van Vliet EP, Heijenbrok-Kal MH, Hunink MG, Kuipers EJ,
Lancet Gastroenterol Hepatol 2024; 9: 34–44. Siersema PD. Staging investigations for oesophageal cancer:
20 Gao Y, Xin L, Feng YD, et al. Feasibility and accuracy of artificial a meta-analysis. Br J Cancer 2008; 98: 547–57.
intelligence-assisted sponge cytology for community-based 40 Betancourt Cuellar SL, Carter BW, Macapinlac HA, et al. Clinical
esophageal squamous cell carcinoma screening in China. staging of patients with early esophageal adenocarcinoma: does
Am J Gastroenterol 2021; 116: 2207–15. FDG-PET/CT have a role? J Thorac Oncol 2014; 9: 1202–06.
21 Iyer PG, Taylor WR, Johnson ML, et al. Accurate nonendoscopic 41 de Gouw DJJM, Klarenbeek BR, Driessen M, et al. Detecting
detection of Barrett’s esophagus by methylated DNA markers: pathological complete response in esophageal cancer after
a multisite case control study. Am J Gastroenterol 2020; 115: 1201–09. neoadjuvant therapy based on imaging techniques: a diagnostic
22 Rustgi AK, El-Serag HB. Esophageal carcinoma. N Engl J Med 2014; systematic review and meta-analysis. J Thorac Oncol 2019; 14: 1156–71.
371: 2499–509. 42 Straatman J, van der Wielen N, Cuesta MA, et al. Minimally
23 Liu Z, Zhao Y, Kong P, et al. Integrated multi-omics profiling yields invasive versus open esophageal resection: three-year follow-up of
a clinically relevant molecular classification for esophageal the previously reported randomized controlled trial: the TIME trial.
squamous cell carcinoma. Cancer Cell 2023; 41: 181–95. Ann Surg 2017; 266: 232–36.
24 Janjigian YY, Werner D, Pauligk C, et al. Prognosis of metastatic 43 Mariette C, Markar SR, Dabakuyo-Yonli TS, et al. Hybrid minimally
gastric and gastroesophageal junction cancer by HER2 status: invasive esophagectomy for esophageal cancer. N Engl J Med 2019;
a European and USA international collaborative analysis. Ann Oncol 380: 152–62.
2012; 23: 2656–62. 44 Biere SSAY, van Berge Henegouwen MI, Maas KW, et al. Minimally
25 Bang YJ, Van Cutsem E, Feyereislova A, et al. Trastuzumab in invasive versus open oesophagectomy for patients with oesophageal
combination with chemotherapy versus chemotherapy alone for cancer: a multicentre, open-label, randomised controlled trial.
treatment of HER2-positive advanced gastric or gastro-oesophageal Lancet 2012; 379: 1887–92.
junction cancer (ToGA): a phase 3, open-label, randomised 45 van der Sluis PC, van der Horst S, May AM, et al. Robot-assisted
controlled trial. Lancet 2010; 376: 687–97. minimally invasive thoracolaparoscopic esophagectomy versus open
26 Sun JM, Shen L, Shah MA, et al. Pembrolizumab plus transthoracic esophagectomy for resectable esophageal cancer:
chemotherapy versus chemotherapy alone for first-line treatment of a randomized controlled trial. Ann Surg 2019; 269: 621–30.
advanced oesophageal cancer (KEYNOTE-590): a randomised, 46 Ando N, Iizuka T, Ide H, et al. Surgery plus chemotherapy
placebo-controlled, phase 3 study. Lancet 2021; 398: 759–71. compared with surgery alone for localized squamous cell carcinoma
27 Doki Y, Ajani JA, Kato K, et al. Nivolumab combination therapy in of the thoracic esophagus: a Japan Clinical Oncology Group
advanced esophageal squamous-cell carcinoma. N Engl J Med 2022; Study—JCOG9204. J Clin Oncol 2003; 21: 4592–96.
386: 449–62. 47 Yang H, Liu H, Chen Y, et al. Long-term efficacy of neoadjuvant
28 Wu HX, Pan YQ, He Y, et al. Clinical benefit of first-line chemoradiotherapy plus surgery for the treatment of locally
programmed death-1 antibody plus chemotherapy in low advanced esophageal squamous cell carcinoma: the
programmed cell death ligand 1-expressing esophageal squamous NEOCRTEC5010 randomized clinical trial. JAMA Surg 2021;
cell carcinoma: a post hoc analysis of JUPITER-06 and meta- 156: 721–29.
analysis. J Clin Oncol 2023; 41: 1735–46. 48 Eyck BM, van Lanschot JJB, Hulshof MCCM, et al. Ten-year
29 Shah MA, Shitara K, Ajani JA, et al. Zolbetuximab plus CAPOX in outcome of neoadjuvant chemoradiotherapy plus surgery for
CLDN18.2-positive gastric or gastroesophageal junction esophageal cancer: the randomized controlled CROSS trial.
adenocarcinoma: the randomized, phase 3 GLOW trial. Nat Med J Clin Oncol 2021; 39: 1995–2004.
2023; 29: 2133–41. 49 Kato K, Machida R, Ito Y, et al. Doublet chemotherapy, triplet
30 Shitara K, Lordick F, Bang Y-J, et al. Zolbetuximab plus mFOLFOX6 chemotherapy, or doublet chemotherapy combined with
in patients with CLDN18.2-positive, HER2-negative, untreated, radiotherapy as neoadjuvant treatment for locally advanced
locally advanced unresectable or metastatic gastric or gastro- oesophageal cancer (JCOG1109 NExT): a randomised, controlled,
oesophageal junction adenocarcinoma (SPOTLIGHT): a multicentre, open-label, phase 3 trial. Lancet 2024; 404: 55–66.
randomised, double-blind, phase 3 trial. Lancet 2023; 401: 1655–68. 50 Kelly RJ, Ajani JA, Kuzdzal J, et al. Adjuvant nivolumab in resected
31 Gruner M, Denis A, Masliah C, et al. Narrow-band imaging versus esophageal or gastroesophageal junction cancer. N Engl J Med 2021;
Lugol chromoendoscopy for esophageal squamous cell cancer 384: 1191–203.
screening in normal endoscopic practice: randomized controlled 51 Al-Batran SE, Homann N, Pauligk C, et al. Perioperative
trial. Endoscopy 2021; 53: 674–82. chemotherapy with fluorouracil plus leucovorin, oxaliplatin, and
32 Rice TW, Gress DM, Patil DT, Hofstetter WL, Kelsen DP, docetaxel versus fluorouracil or capecitabine plus cisplatin and
Blackstone EH. Cancer of the esophagus and esophagogastric epirubicin for locally advanced, resectable gastric or gastro-
junction—major changes in the American Joint Committee on oesophageal junction adenocarcinoma (FLOT4): a randomised,
Cancer eighth edition cancer staging manual. CA: Cancer J Clin phase 2/3 trial. Lancet 2019; 393: 1948–57.
2017; 67: 304–17. 52 Hulshof MCCM, Geijsen ED, Rozema T, et al. Randomized study
33 Yang J, Luo G-Y, Liang R-B, et al. Efficacy of endoscopic on dose escalation in definitive chemoradiation for patients with
ultrasonography for determining clinical T category for esophageal locally advanced esophageal cancer (ARTDECO study). J Clin Oncol
squamous cell carcinoma: data from 1434 surgical cases. 2021; 39: 2816–24.
Ann Surg Oncol 2018; 25: 2075–82.
53 Conroy T, Galais MP, Raoul JL, et al. Definitive chemoradiotherapy 74 Hulscher JB, van Sandick JW, de Boer AG, et al. Extended
with FOLFOX versus fluorouracil and cisplatin in patients with transthoracic resection compared with limited transhiatal resection
oesophageal cancer (PRODIGE5/ACCORD17): final results of for adenocarcinoma of the esophagus. N Engl J Med 2002;
a randomised, phase 2/3 trial. Lancet Oncol 2014; 15: 305–14. 347: 1662–69.
54 Kamarajah SK, Phillips AW, Hanna GB, Low D, Markar SR. Definitive 75 Tagkalos E, van der Sluis PC, Berlth F, et al. Robot-assisted
chemoradiotherapy compared to neoadjuvant chemoradiotherapy minimally invasive thoraco-laparoscopic esophagectomy versus
with esophagectomy for locoregional esophageal cancer: national minimally invasive esophagectomy for resectable esophageal
population-based cohort study. Ann Surg 2022; 275: 526–33. adenocarcinoma, a randomized controlled trial (ROBOT-2 trial).
55 Markar S, Gronnier C, Duhamel A, et al. Salvage surgery after BMC Cancer 2021; 21: 1060.
chemoradiotherapy in the management of esophageal cancer: is it a 76 Gottlieb-Vedi E, Kauppila JH, Mattsson F, et al. Long-term survival
viable therapeutic option? J Clin Oncol 2015; 33: 3866–73. in esophageal cancer after minimally invasive esophagectomy
56 Janjigian YY, Kawazoe A, Bai Y, et al. Pembrolizumab plus compared to open esophagectomy. Ann Surg 2022; 276: e744–48.
trastuzumab and chemotherapy for HER2-positive gastric or 77 Hagens ERC, van Berge Henegouwen MI, Gisbertz SS. Distribution
gastro-oesophageal junction adenocarcinoma: interim analyses of lymph node metastases in esophageal carcinoma patients
from the phase 3 KEYNOTE-811 randomised placebo-controlled undergoing upfront surgery: a systematic review. Cancers 2020;
trial. Lancet 2023; 402: 2197–208. 12: 1592.
57 Bennett C, Green S, DeCaestecker J, et al. Surgery versus radical 78 Xu S, Chen D, Liu Z, et al. Impact of the extent of recurrent
endotherapies for early cancer and high-grade dysplasia in Barrett’s laryngeal nerve lymphadenectomy on thoracic esophageal
oesophagus. Cochrane Database Syst Rev 2020; 5: CD007334. squamous cell carcinoma: a real-world multicentre study.
58 Merkow RP, Bilimoria KY, Keswani RN, et al. Treatment trends, risk Eur J Cardiothorac Surg 2023; 63: ezad168.
of lymph node metastasis, and outcomes for localized esophageal 79 Li B, Hu H, Zhang Y, et al. Extended right thoracic approach
cancer. J Natl Cancer Inst 2014; 106: dju133. compared with limited left thoracic approach for patients with
59 Allum WH, Stenning SP, Bancewicz J, Clark PI, Langley RE. middle and lower esophageal squamous cell carcinoma. Ann Surg
Long-term results of a randomized trial of surgery with or without 2018; 267: 826–32.
preoperative chemotherapy in esophageal cancer. J Clin Oncol 2009; 80 Li B, Zhang Y, Miao L, et al. Esophagectomy with three-field versus
27: 5062–67. two-field lymphadenectomy for middle and lower thoracic
60 Ychou M, Boige V, Pignon JP, et al. Perioperative chemotherapy esophageal cancer: long-term outcomes of a randomized clinical
compared with surgery alone for resectable gastroesophageal trial. J Thorac Oncol 2021; 16: 310–17.
adenocarcinoma: an FNCLCC and FFCD multicenter phase III trial. 81 Hagens ERC, van Berge Henegouwen MI, van Sandick JW, et al.
J Clin Oncol 2011; 29: 1715–21. Distribution of lymph node metastases in esophageal carcinoma
61 Cunningham D, Allum WH, Stenning SP, et al. Perioperative [TIGER study]: study protocol of a multinational observational
chemotherapy versus surgery alone for resectable gastroesophageal study. BMC Cancer 2019; 19: 662.
cancer. N Engl J Med 2006; 355: 11–20. 82 Raja S, Rice TW, Murthy SC, et al. Value of lymphadenectomy in
62 Yamasaki M, Miyata H, Yamashita K, et al. Chemoradiotherapy patients receiving neoadjuvant therapy for esophageal
versus triplet chemotherapy as initial therapy for T4b esophageal adenocarcinoma. Ann Surg 2021; 274: e320–27.
cancer: survival results from a multicenter randomized phase 2 trial. 83 Ando N, Kato H, Igaki H, et al. A randomized trial comparing
Br J Cancer 2023; 129: 54–60. postoperative adjuvant chemotherapy with cisplatin and
63 Shaheen NJ, Sharma P, Overholt BF, et al. Radiofrequency ablation in 5-fluorouracil versus preoperative chemotherapy for localized
Barrett’s esophagus with dysplasia. N Engl J Med 2009; 360: 2277–88. advanced squamous cell carcinoma of the thoracic esophagus
64 Sun F, Yuan P, Chen T, Hu J. Efficacy and complication of endoscopic (JCOG9907). Ann Surg Oncol 2012; 19: 68–74.
submucosal dissection for superficial esophageal carcinoma: a 84 Shapiro J, van Lanschot JJB, Hulshof MCCM, et al. Neoadjuvant
systematic review and meta-analysis. J Cardiothorac Surg 2024; 9: 78. chemoradiotherapy plus surgery versus surgery alone for
65 Guo H-M, Zhang X-Q, Chen M, Huang S-L, Zou X-P. Endoscopic oesophageal or junctional cancer (CROSS): long-term results of
submucosal dissection vs endoscopic mucosal resection for a randomised controlled trial. Lancet Oncol 2015; 16: 1090–98.
superficial esophageal cancer. World J Gastroenterol 2014; 20: 5540–47. 85 Reynolds JV, Preston SR, O’Neill B, et al. Trimodality therapy
66 Sferrazza S, Crispino F, Vieceli F, Fiorentino A, Michielan A, versus perioperative chemotherapy in the management of locally
de Pretis G. Circumferential endoscopic submucosal dissection for advanced adenocarcinoma of the oesophagus and oesophagogastric
long-segment Barrett’s adenocarcinoma: the double-tunnel and junction (Neo-AEGIS): an open-label, randomised, phase 3 trial.
single clip-and-loop traction method. Endoscopy 2023; 55: E645–46. Lancet Gastroenterol Hepatol 2023; 8: 1015–27.
67 Zhang Y, Ding H, Chen T, et al. Outcomes of endoscopic submucosal 86 Klevebro F, Alexandersson von Döbeln, Wang N, et al.
dissection vs esophagectomy for T1 esophageal squamous cell A randomized clinical trial of neoadjuvant chemotherapy versus
carcinoma in a real-world cohort. Clin Gastroenterol Hepatol 2019; neoadjuvant chemoradiotherapy for cancer of the oesophagus or
17: 73–81. gastro-oesophageal junction. Ann Oncol 2016; 27: 660–67.
68 Ishihara R, Arima M, Iizuka T, et al. Endoscopic submucosal 87 Tang H, Wang H, Fang Y, et al. Neoadjuvant chemoradiotherapy
dissection/endoscopic mucosal resection guidelines for esophageal versus neoadjuvant chemotherapy followed by minimally invasive
cancer. Dig Endosc 2020; 32: 452–93. esophagectomy for locally advanced esophageal squamous cell
carcinoma: a prospective multicenter randomized clinical trial.
69 Minashi K, Nihei K, Mizusawa J, et al. Efficacy of endoscopic
Ann Oncol 2023; 34: 163–72.
resection and selective chemoradiotherapy for stage I esophageal
squamous cell carcinoma. Gastroenterology 2019; 157: 382–90. 88 Faron M, Cheugoua-Zanetsie M, Tierney J, et al. Individual
participant data network meta-analysis of neoadjuvant
70 Yamashita H, Kadota T, Minamide T, et al. Efficacy and safety of
chemotherapy or chemoradiotherapy in esophageal or
second photodynamic therapy for local failure after salvage
gastroesophageal junction carcinoma. J Clin Oncol 2023; 41: 4535–47.
photodynamic therapy for esophageal cancer. Dig Endosc 2022;
34: 488–96. 89 van Hagen P, Hulshof MC, van Lanschot JJ, et al. Preoperative
chemoradiotherapy for esophageal or junctional cancer.
71 Low DE, Kuppusamy MK, Alderson D, et al. Benchmarking
N Engl J Med 2012; 366: 2074–84.
complications associated with esophagectomy. Ann Surg 2019;
269: 291–98. 90 Yang H, Liu H, Chen Y, et al. Neoadjuvant chemoradiotherapy
followed by surgery versus surgery alone for locally advanced
72 Low DE, Allum W, De Manzoni G, et al. Guidelines for perioperative
squamous cell carcinoma of the esophagus (NEOCRTEC5010):
care in esophagectomy: Enhanced Recovery After Surgery (ERAS)
a phase III multicenter, randomized, open-label clinical trial.
Society recommendations. World J Surg 2019; 43: 299–330.
J Clin Oncol 2018; 36: 2796–803.
73 Kutup A, Nentwich MF, Bollschweiler E, Bogoevski D, Izbicki JR,
91 Mariette C, Dahan L, Mornex F, et al. Surgery alone versus
Hölscher AH. What should be the gold standard for the surgical
chemoradiotherapy followed by surgery for stage I and II
component in the treatment of locally advanced esophageal cancer:
esophageal cancer: final analysis of randomized controlled phase III
transthoracic versus transhiatal esophagectomy. Ann Surg 2014;
trial FFCD 9901. J Clin Oncol 2014; 32: 2416–22.
260: 1016–22.
92 Lordick F, Mariette C, Haustermans K, Obermannová R, Arnold D. 107 Kang Y-K, Chen L-T, Ryu M-H, et al. Nivolumab plus chemotherapy
Oesophageal cancer: ESMO Clinical Practice Guidelines for versus placebo plus chemotherapy in patients with HER2-negative,
diagnosis, treatment and follow-up. Ann Oncol 2016; untreated, unresectable advanced or recurrent gastric or gastro-
27 (suppl 5): v50–57. oesophageal junction cancer (ATTRACTION-4): a randomised,
93 Noordman BJ, Verdam MGE, Lagarde SM, et al. Effect of multicentre, double-blind, placebo-controlled, phase 3 trial.
neoadjuvant chemoradiotherapy on health-related quality of life in Lancet Oncol 2022; 23: 234–47.
esophageal or junctional cancer: results from the randomized 108 Xu J, Jiang H, Pan Y, et al. Sintilimab plus chemotherapy for
CROSS trial. J Clin Oncol 2018; 36: 268–75. unresectable gastric or gastroesophageal junction cancer: the
94 Zhan M, Zheng H, Yang Y, Xu T, Li Q. Cost-effectiveness analysis of ORIENT-16 randomized clinical trial. JAMA 2023; 330: 2064–74.
neoadjuvant chemoradiotherapy followed by surgery versus surgery 109 Rha SY, Oh D-Y, Yañez P, et al. Pembrolizumab plus chemotherapy
alone for locally advanced esophageal squamous cell carcinoma versus placebo plus chemotherapy for HER2-negative advanced
based on the NEOCRTEC5010 trial. Radiother Oncol 2019; gastric cancer (KEYNOTE-859): a multicentre, randomised, double-
141: 27–32. blind, phase 3 trial. Lancet Oncol 2023; 24: 1181–95.
95 Lee Y, Samarasinghe Y, Lee MH, et al. Role of adjuvant therapy in 110 Qiu MZ, Oh DY, Kato K, et al. Tislelizumab plus chemotherapy
esophageal cancer patients after neoadjuvant therapy and versus placebo plus chemotherapy as first line treatment for
esophagectomy: a systematic review and meta-analysis. Ann Surg advanced gastric or gastro-oesophageal junction adenocarcinoma:
2022; 275: 91–98. RATIONALE-305 randomised, double blind, phase 3 trial. BMJ
96 Herskovic A, Martz K, al-Sarraf M, et al. Combined chemotherapy 2024; 385: e078876.
and radiotherapy compared with radiotherapy alone in patients with 111 Janjigian YY, Kawazoe A, Yañez P, et al. The KEYNOTE-811 trial of
cancer of the esophagus. N Engl J Med 1992; 326: 1593–98. dual PD-1 and HER2 blockade in HER2-positive gastric cancer.
97 Minsky BD, Pajak TF, Ginsberg RJ, et al. INT 0123 (radiation Nature 2021; 600: 727–30.
therapy oncology group 94-05) phase III trial of combined-modality 112 Chen YX, Wang ZX, Jin Y, et al. An immunogenic and oncogenic
therapy for esophageal cancer: high-dose versus standard-dose feature-based classification for chemotherapy plus PD-1 blockade in
radiation therapy. J Clin Oncol 2002; 20: 1167–74. advanced esophageal squamous cell carcinoma. Cancer Cell 2023;
98 Wagner AD, Grothe W, Haerting J, Kleber G, Grothey A, Fleig WE. 41: 919–932.e5.
Chemotherapy in advanced gastric cancer: a systematic review and 113 Kojima T, Shah MA, Muro K, et al. Randomized phase III
meta-analysis based on aggregate data. J Clin Oncol 2006; KEYNOTE-181 study of pembrolizumab versus chemotherapy in
24: 2903–09. advanced esophageal cancer. J Clin Oncol 2020; 38: 4138–48.
99 van Kleef JJ, Ter Veer E, van den Boorn HG, et al. Quality of life 114 Kato K, Cho BC, Takahashi M, et al. Nivolumab versus
during palliative systemic therapy for esophagogastric cancer: chemotherapy in patients with advanced oesophageal squamous
systematic review and meta-analysis. J Natl Cancer Inst 2020; cell carcinoma refractory or intolerant to previous chemotherapy
112: 12–29. (ATTRACTION-3): a multicentre, randomised, open-label, phase 3
100 Luo H, Lu J, Bai Y, et al. Effect of camrelizumab vs placebo added trial. Lancet Oncol 2019; 20: 1506–17.
to chemotherapy on survival and progression-free survival in 115 Shen L, Kato K, Kim SB, et al. Tislelizumab versus chemotherapy as
patients with advanced or metastatic esophageal squamous cell second-line treatment for advanced or metastatic esophageal
carcinoma: the ESCORT-1st randomized clinical trial. JAMA 2021; squamous cell carcinoma (RATIONALE-302): a randomized
326: 916–25. phase III study. J Clin Oncol 2022; 40: 3065–76.
101 Wang Z-X, Cui C, Yao J, et al. Toripalimab plus chemotherapy in 116 Elliott JA, Markar SR, Klevebro F, et al. An international
treatment-naïve, advanced esophageal squamous cell carcinoma multicenter study exploring whether surveillance after esophageal
(JUPITER-06): a multi-center phase 3 trial. Cancer Cell 2022; cancer surgery impacts oncological and quality of life outcomes
40: 277–88. (ENSURE). Ann Surg 2023; 277: e1035–44.
102 Lu Z, Wang J, Shu Y, et al. Sintilimab versus placebo in 117 Lou F, Sima CS, Adusumilli PS, et al. Esophageal cancer recurrence
combination with chemotherapy as first line treatment for locally patterns and implications for surveillance. J Thorac Oncol 2013;
advanced or metastatic oesophageal squamous cell carcinoma 8: 1558–62.
(ORIENT-15): multicentre, randomised, double blind, phase 3 trial. 118 Zheng RS, Sun KX, Zhang SW, et al. Report of cancer epidemiology
BMJ 2022; 377: e068714. in China, 2015. Zhonghua Zhong Liu Za Zhi 2019; 41: 19–28
103 Xu J, Kato K, Raymond E, et al. Tislelizumab plus chemotherapy (in Chinese).
versus placebo plus chemotherapy as first-line treatment for 119 Zylstra J, Whyte GP, Beckmann K, et al. Exercise prehabilitation
advanced or metastatic oesophageal squamous cell carcinoma during neoadjuvant chemotherapy may enhance tumour regression
(RATIONALE-306): a global, randomised, placebo-controlled, phase in oesophageal cancer: results from a prospective non-randomised
3 study. Lancet Oncol 2023; 24: 483–95. trial. Br J Sports Med 2022; 56: 402–09.
104 Song Y, Zhang B, Xin D, et al. First-line serplulimab or placebo plus 120 Lu Z, Fang Y, Liu C, et al. Early interdisciplinary supportive care in
chemotherapy in PD-L1-positive esophageal squamous cell patients with previously untreated metastatic esophagogastric
carcinoma: a randomized, double-blind phase 3 trial. Nat Med 2023; cancer: a phase III randomized controlled trial. J Clin Oncol 2021;
29: 473–82. 39: 748–56.
105 Li J, Chen Z, Bai Y, et al. First-line sugemalimab with chemotherapy
for advanced esophageal squamous cell carcinoma: a randomized Copyright © 2024 Elsevier Ltd. All rights reserved, including those for
phase 3 study. Nat Med 2024; 30: 740–48. text and data mining, AI training, and similar technologies.
106 Janjigian YY, Shitara K, Moehler M, et al. First-line nivolumab plus
chemotherapy versus chemotherapy alone for advanced gastric,
gastro-oesophageal junction, and oesophageal adenocarcinoma
(CheckMate 649): a randomised, open-label, phase 3 trial. Lancet
2021; 398: 27–40.