Evolving Therapeutics for Resectable Esophageal Adenocarcinoma

Authors:
Kelsey A. Klute Department of Medicine, University of Nebraska Medical Center, Omaha, NE

Search for other papers by Kelsey A. Klute in
Current site
Google Scholar
PubMed
Close
 MD,
and
Manish A. Shah Department of Medicine, Weill Cornell Medicine, New York, NY

Search for other papers by Manish A. Shah in
Current site
Google Scholar
PubMed
Close
 MD
Full access

Surgical resection remains the backbone of curative treatment for localized esophageal adenocarcinoma. However, resection alone carries a significant risk of recurrence and is no longer considered an acceptable approach for patients with locally advanced disease. Strategies incorporating perioperative chemotherapy, neoadjuvant chemoradiotherapy, and adjuvant immune checkpoint blockade have significantly improved survival. However, despite these advances, approximately half of all patients with locally advanced disease will experience recurrence and ultimately succumb to their disease. To overcome this innate resistance to cytotoxic therapy, novel strategies are being developed. This review discusses both evidence-based and emerging perioperative approaches and explores potential risk-adapted strategies to individualize perioperative therapy.

Resection remains the primary curative treatment for localized esophageal adenocarcinoma (EAC). However, the 5-year survival rate with resection alone remains low at 26% due to a high risk of recurrence.1 Multimodal approaches incorporating perioperative chemotherapy, preoperative chemoradiotherapy (CRT), and adjuvant immunotherapy have improved survival compared with surgery alone.25 These strategies aim to downstage the primary tumor, increase the likelihood of margin-negative resection, eradicate micrometastatic disease, and ultimately improve survival in patients with EAC.

Although the ESOPEC study recently established the benefit of perioperative combination chemotherapy for locally advanced disease, nearly 50% of patients still experience recurrence and succumb to their disease.5 Strategies incorporating targeted therapies and new technology to individualize therapy will be critical in further improving survival.

This review discusses clinical trials that inform the current management of resectable EAC in patients fit for surgery. It will also explore unanswered questions, ongoing trials, and state-of-the-art technology that may inform perioperative therapy in the near future.

Neoadjuvant CRT or Perioperative Chemotherapy—Is There an Optimal Approach?

Randomized clinical trials have established the benefit of preoperative and perioperative therapy using chemotherapy or CRT (Table 1).2,3,57 Until recently, equipoise existed between these strategies, with the choice between perioperative chemotherapy or neoadjuvant CRT largely depending on physician and institutional preference. ASCO, ESMO, and NCCN have historically recommended preoperative CRT or perioperative chemotherapy.810 However, the results of the ESOPEC trial are poised to shift this paradigm.5

Table 1.

Key Clinical Trials Evaluating Preoperative CRT and Perioperative Chemotherapy

Trial Phase Study Population Treatment Arms Endpoints and Efficacy Reference
MAGIC III E/GEJ/G AC Perioperative chemo with ECF + surgery vs surgery alone OS: perioperative chemo superior HR, 0.75; 95% CI, 0.60–0.93; P=.009 7
CROSS III E/GEJ SCC and AC (75% AC) Neoadjuvant CRT with weekly carboplatin + paclitaxel + surgery vs surgery alone OS for AC + SCC: neoadjuvant CRT superior 49.4 vs 24.0 mo HR, 0.657; 95% CI, 0.495–0.871; P=.003

OS for AC subgroup: HR, 0.741 (95% CI, 0.536–1.024; P=.07)
2
FLOT4 II/III GEJ/G AC Perioperative chemo with FLOT vs ECF/ECX pCR (phase II): FLOT superior 15% vs 6%; P=.02

OS (phase III): FLOT superior 35 vs 50 mo HR, 0.77; 95% CI, 0.63–0.94; P=.012
3,54
ESOPEC III E/GEJ AC Perioperative FLOT vs neoadjuvant CRT (CROSS) OS: FLOT superior 66 vs 37 mo HR, 0.72; 95% CI, 0.54–0.96; P=.023 5
TOPGEAR III GEJ/G AC Preoperative chemo (ECF × 2 /FLOT × 3) + neoadjuvant CRT vs preoperative chemo (ECF × 3/FLOT × 4); both groups received postoperative chemo (ECF × 3/FLOT × 4) OS: no difference 46 mo chemo + CRT vs 49 mo chemo HR, 1.04; 95% CI, 0.83–1.30

OS for GEJ subgroup: HR, 0.78; 95% CI, 0.55–1.12
15
Neo-AEGIS III E/GEJ AC CRT vs perioperative ECF/EOX/FLOT (15% FLOT) OS: no difference 48.0 mo perioperative chemo vs 49.2 mo CRT HR, 1.03; 95% CI, 0.77–1.38; P=.82 11
CALGB 80803 II E/GEJ AC Neoadjuvant chemo (FOLFOX or carboplatin + paclitaxel) + CRT with continuation of induction chemo during CRT in PET responders, switch chemo in PET nonresponders pCR in PET nonresponders: 18% (FOLFOX induction) and 20% (carboplatin + paclitaxel induction) 14
CRITICS III GEJ/G AC (17% GEJ) Perioperative chemo (ECX/EOX) vs preoperative chemo and postoperative CRT OS: no difference 43 mo perioperative chemo vs 37 mo postoperative CRT HR, 1.01; 95% CI, 0.84–1.22; P=.90 15

Abbreviations: AC, adenocarcinoma; chemo, chemotherapy; CRT, chemoradiotherapy; DFS, disease-free survival; E, esophagus; ECF/X, epirubicin/cisplatin/5-fluorouracil or capecitabine; EFS, event-free survival; EOX, epirubicin/oxaliplatin/capecitabine; FLOT, 5-FU/leucovorin/oxaliplatin/docetaxel; FOLFOX, 5-fluorouracil/leucovorin/oxaliplatin; G, gastric; GEJ, gastroesophageal junction; HR, hazard ratio; OS, overall survival; pCR, pathologic complete response; SCC, squamous cell carcinoma.

Perioperative Chemotherapy

The phase III MAGIC trial established the benefit of perioperative chemotherapy over surgery alone. Although most patients in the trial had gastric or gastroesophageal junction (GEJ) adenocarcinoma, approximately 25% had EAC.7 Perioperative epirubicin, cisplatin, and 5-FU (ECF) significantly improved overall survival (OS) compared with surgery alone (hazard ratio [HR], 0.75; 95% CI, 0.60–0.93; P=.009), with a 5-year OS rate of 36% versus 23%, respectively. Subsequently, the phase II/III FLOT4 trial compared perioperative ECF to perioperative FLOT (5-FU/leucovorin/oxaliplatin/docetaxel) in patients with resectable GEJ and gastric adenocarcinoma.3 Survival was significantly longer with FLOT (median OS, 50 vs 35 months; HR, 0.75; 95% CI, 0.60–0.93; P=.009), establishing FLOT as the standard perioperative chemotherapy regimen for locally advanced esophageal carcinoma.

Neoadjuvant CRT

The phase III CROSS trial evaluated the benefit of neoadjuvant CRT in patients with resectable esophageal cancer.2 Patients with T2–3 or N+ disease (75% adenocarcinoma) were randomized to neoadjuvant chemotherapy (weekly carboplatin + paclitaxel [CP]) with concurrent radiotherapy (41.4 Gy in 23 fractions) or resection alone. The CROSS regimen was well-tolerated and did not increase postoperative complications. Overall, neoadjuvant CRT doubled median OS to 49.4 months versus 24.0 months with surgery alone (HR, 0.657; 95% CI, 0.495–0.871; P=.003). Although the benefit of neoadjuvant CRT was observed in all patients, the survival benefit was largely driven by those with SCC (HR for death, 0.422; 95% CI, 0.226–0.788; P=.007), compared with a more modest effect in EAC (HR, 0.741; 95% CI, 0.536–1.024; P=.07). CRT reduced the local recurrence rate but did not impact distance recurrence rates (27% vs 28%; HR, 0.76; 95% CI, 0.52–1.13). With long-term follow-up, the benefit of CRT relative to surgery alone in EAC remains marginal, with a 10-year OS rate of 36% versus 26% (HR, 0.75; 95% CI, 0.56–1.00).1 The attenuated benefit in EAC and lack of impact on distant recurrence risk leave the optimal paradigm for resectable EAC in question.

Is There an Optimal Approach?

The phase III Neo-AEGIS and ESOPEC trials compared neoadjuvant CRT with perioperative chemotherapy in patients with resectable EAC. Neo-AEGIS randomized patients either to neoadjuvant CRT with the CROSS regimen or perioperative chemotherapy, initially using the MAGIC regimen and later incorporating perioperative FLOT.11 The trial design was modified to assess noninferiority after the first interim analysis, but closed prematurely due to slow accrual. Survival was similar between perioperative chemotherapy and CRT (48.0 vs 49.2 months; HR, 1.03; 95% CI, 0.77–1.38; P=.82), leaving clinical equipoise between these approaches. However, because only 15% of the perioperative chemotherapy arm received FLOT, whether perioperative FLOT could be more effective remained an open question.

In contrast, the ESOPEC trial established perioperative FLOT as a more effective strategy than neoadjuvant CRT for fit patients with resectable EAC.5 ESOPEC randomized patients to perioperative FLOT or neoadjuvant CRT with the CROSS regimen, excluding those with SCC or gastric cancer. FLOT significantly improved survival (median, 66 vs 37 months; HR, 0.70; 95% CI, 0.53–0.92; P=.012). Although the pCR rate in the CRT arm was lower than expected based on the pCR rate in the CROSS trial (10% vs 23% in CROSS), direct comparisons are limited because ESOPEC included patients with more advanced tumors [cT4] and a greater proportion of cN+ disease (79.7% vs 64.5% in CROSS). Although FLOT improved survival over CRT in the ESOPEC study, some limitations should be considered. Nearly one-third of patients did not complete planned chemotherapy during radiation. Furthermore, patients in the CRT arm did not receive adjuvant nivolumab, which is the current standard of care for patients with residual disease after neoadjuvant CRT and surgery based on CheckMate 577.4 Thus, the true benefit of perioperative FLOT over standard CRT with adjuvant nivolumab remains unknown.

Together, Neo-AEGIS and ESOPEC suggest that perioperative chemotherapy is at least as effective, and possibly superior, to neoadjuvant CRT in locally advanced EAC. As such, neoadjuvant CRT with adjuvant nivolumab should likely be reserved for patients unable to tolerate FLOT.

Preoperative Chemotherapy + Chemoradiotherapy

Given the proven benefits of perioperative chemotherapy and neoadjuvant CRT individually, there is growing interest in combining these strategies. Total neoadjuvant approaches are particularly appealing for esophagogastric tumors, given the high morbidity of resection and the challenges of delivering postoperative therapy. For example, in the FLOT4 and ESOPEC studies, only 62% and 59% of patients initiated postoperative chemotherapy, and even fewer completed all planned cycles (46% and 52%, respectively).3,7 Retrospective and single-arm studies in EAC support the safety and feasibility of preoperative chemotherapy combined with CRT, demonstrating high adherence rates to planned preoperative therapy.12,13

CALGB 80803

The phase II CALGB 80803 trial evaluated whether FDG-PET following induction chemotherapy could be used to guide neoadjuvant therapy and improve pCR rates in patients receiving neoadjuvant chemotherapy followed by CRT.14 Patients were initially randomized to 6 weeks of modified FOLFOX (5-FU/leucovorin/oxaliplatin) or CP, followed by a PET scan. Based on standard uptake value (SUV) changes, patients were classified as PET responders if the SUV decreased by ≥35%, or nonresponders if the SUV decreased by <35%. Responders continued with the same chemotherapy during CRT, and nonresponders switched to the alternate chemotherapy regimen during CRT.

Results of the primary endpoint, pCR rate in PET nonresponders after switching chemotherapy, are discussed later. Interestingly, a notable finding was the striking difference in pCR rates among PET responders, with a 40.3% pCR rate in FOLFOX responders compared with 14% in CP responders. After a median follow-up of 5.17 years, FOLFOX responders had a median OS that was not reached, with a 5-year OS of 53%, whereas CP responders had a median OS of 38.7 months and a 5-year OS of 42.9%. Although the trial was not designed to directly compare outcomes between chemotherapy regimens, the higher pCR and survival rates in FOLFOX responders suggest a potential advantage over CP.

TOPGEAR

The phase III TOPGEAR trial evaluated the benefit of adding preoperative CRT to perioperative chemotherapy (ECF or FLOT) in patients with gastric or GEJ adenocarcinoma.15 Overall, the addition of preoperative CRT did not improve OS (HR, 1.05; 95% CI, 0.83–1.31; Table 2). Although TOPGEAR primarily focused on gastric cancer, approximately one-third of the cohort had GEJ tumors. Subgroup analyses suggest that preoperative CRT may provide some benefit in the GEJ subgroup (HR, 0.78; 95% CI, 0.55–1.22) and in patients with T1–2 tumors (HR, 0.63; 95% CI, 0.29–1.37). Only one-third of patients received FLOT, and the radiation dose was only 45 Gy, so it is possible that more effective systemic control with FLOT combined with a conventional radiation dose (50.4 Gy) could improve efficacy. Ongoing trials will more specifically evaluate the role of preoperative CRT combined with preoperative chemotherapy in patients with EAC and GEJ adenocarcinoma, often as total neoadjuvant therapy, to determine whether these combined strategies improve outcomes and whether CRT has a role in treating fit patients with resectable EAC (Table 2).1,1518

Table 2.

Selected Clinical Trials Evaluating Strategies Combining Preoperative Chemotherapy With CRT

Trial Phase Study Population Treatment Arms Primary Endpoint and Efficacy Reference
TOPGEAR II/III GEJ/G AC Perioperative chemo (ECF/X × 3 or FLOT × 4 preoperative and postoperative) + preoperative CRT (5-FU infusion) vs perioperative chemo [add RT dose] OS: no difference 46 mo chemo + CRT vs 49 mo chemo alone HR, 1.05; 95% CI, 0.83–1.31 15
TNT-OES 2 II E/GEJ AC TNT with FLOT followed by CRT (CROSS) and CRT (CROSS) followed by FLOT [add RT dose] DFS: ongoing NCT06161818 1
CRITICS-II II E/GEJ AC Preoperative chemo (DOC × 4) vs chemo + CRT vs CRT [add RT dose] EFS: ongoing NCT02931890 16
RACE III GEJ (Siewert I–III) AC Perioperative chemo (FLOT) vs Preoperative chemo + CRT [add RT dose] DFS: ongoing NCT04375605 32
PREACT III GEJ/G (Siewert II/III) AC Perioperative chemo (SOX) + preoperative CRT vs preoperative chemo [add RT dose] DFS: ongoing NCT03013010 18

Abbreviations: AC, adenocarcinoma; chemo, chemotherapy; CRT, chemoradiotherapy; DFS, disease-free survival; DOC, docetaxel/oxaliplatin/capecitabine; E, esophagus; EFS, event-free survival; ECF/X, epirubicin/cisplatin/5-fluorouracil or capecitabine; EFS, event-free survival; FLOT, 5-FU/leucovorin/oxaliplatin/docetaxel; G, gastric; GEJ, gastroesophageal junction; HR, hazard ratio; OS, overall survival; RT, radiotherapy; SCC, squamous cell carcinoma; SOX, S-1/oxaliplatin; TNT, total neoadjuvant therapy.

Notably, no studies to date have definitively demonstrated that trimodality therapy (chemotherapy, radiation, and surgery) is superior to bimodality therapy (eg, perioperative chemotherapy + surgery or preoperative CRT + surgery). This suggests that a subset of patients may have an innate resistance to cytotoxic therapy, and implies that alternative approaches are needed to improve survival.

Role of Immune Checkpoint Inhibitors and Targeted Therapies in Resectable EAC

Despite multimodality therapy, fewer than half of patients with resectable EAC are cured, underscoring the urgent need for more effective strategies.2,3,5 Given the promising activity of immune checkpoint inhibitors (ICIs) and targeted therapies in advanced disease, there is growing enthusiasm for incorporating these novel therapies into the perioperative setting.19,20

Immune Checkpoint Inhibitors

Postoperative Immunotherapy: CheckMate-577

Patients with EAC and residual pathologic disease following neoadjuvant CRT face an exceptionally high risk of recurrence, with 75% developing recurrent disease.2,21 Until recently, there were no proven adjuvant strategies for this high-risk group. The phase III CheckMate-577 trial tested whether 1 year of adjuvant nivolumab in patients who received neoadjuvant CRT and did not experience a pCR could improve disease-free survival (DFS).4 Adjuvant nivolumab doubled DFS compared with placebo (22.4 vs 11 months; HR, 0.69; 96.4% CI, 0.56–0.86; P<.001). In the adenocarcinoma subgroup, adjuvant nivolumab improved median DFS from 11.1 months with placebo to 19.4 months with nivolumab (HR, 0.75; 95% CI, 0.59–0.96). Although OS data are still awaited, adjuvant nivolumab has become the current standard for patients with EAC treated with neoadjuvant CRT who have residual pathologic disease.10

Preoperative ICI: EA2174

EA2174 is a recently completed phase II/III trial evaluating the benefit of adding an ICI to neoadjuvant CRT and comparing the effectiveness of single-agent versus dual checkpoint blockade in the adjuvant setting.22 Patients with EAC were randomized to concurrent CRT with or without nivolumab, followed by surgery. After resection, they were randomized again to receive either nivolumab or nivolumab + ipilimumab. Initial results presented at the 2024 ASCO Annual Meeting showed that the addition of nivolumab to neoadjuvant CRT did not improve the pCR rate in patients with resected EAC or GEJ adenocarcinoma (21% for CRT alone vs 25% for CRT with nivolumab; P=.27). The study is ongoing to analyze the adjuvant endpoint, DFS.

Perioperative ICI

Three phase III trials—KEYNOTE-585, MATTERHORN, DANTE—are currently ongoing to evaluate the combination of ICIs with perioperative chemotherapy (Table 4).17,23,24 Although none of these trials focus specifically on esophageal tumors, their results may provide valuable insights for EAC treatment. All 3 trials demonstrate that adding an ICI to preoperative chemotherapy improves the pCR rate (Table 3). Unfortunately, adding pembrolizumab to perioperative chemotherapy in KEYNOTE-585 did not improve event-free survival (EFS).23 Although the data from DANTE are still maturing, top-line results from MATTERHORN suggest that the study met its primary endpoint of EFS.

Table 3.

Selected Clinical Trials of Immune Checkpoint Inhibitors for Resectable E/GEJ AC

Trial Phase Study Population Treatment Arms Primary Endpoints and Efficacy Reference
CheckMate-577 III E/GEJ SCC and AC (71% AC) without pCR to neoadjuvant CRT Adjuvant nivolumab vs placebo DFS: nivolumab superior 22.4 vs 11.0 mo HR, 0.69; 95% CI, 0.56–0.86; P<.001 4
EA2174 II/III E/GEJ AC Perioperative CRT ± nivolumab

Adjuvant nivolumab ± ipilimumab
pCR rate (neoadjuvant): no difference 21% CRT vs 24.8% CRT + nivolumab (P=.27)

DFS (adjuvant): ongoing NCT03604991
22
KEYNOTE-975 III E/GEJ SCC and AC

Ineligible for curative surgery
Definitive CRT (FOLFOX) + pembrolizumab vs definitive CRT + placebo OS, EFS: ongoing NCT04210115 39
KEYNOTE-585 III GEJ/G AC Perioperative chemo (cisplatin + 5-FU or capecitabine) + pembrolizumab vs chemo + placebo pCR rate: chemo + pembrolizumab superior 12.9% vs 2.0 %; P<.00001

EFS: no benefit 44.4 vs 25.3 mo HR, 0.81; 95% CI, 0.67–0.99; P=.0190

OS: no benefit 60.7 vs 58.0 mo HR, 0.90; 95% CI, 0.73–1.12; P=.174
23
MATTERHORN III GEJ/G AC Perioperative chemo (FLOT) + durvalumab vs chemo EFS (phase III): ongoing

Interim pCR rate: 19% durvalumab + chemo arm vs 7% chemo arm OR, 3.08; P=.00001 NCT04592913
24
DANTE II/III GEJ/G AC Perioperative chemo (FLOT) + atezolizumab vs FLOT pCR or major pathologic response (phase II): chemo + atezolizumab superior 24% vs 15%; P=.032

EFS (phase III): ongoing
17
INFINITY II dMMR/MSI-H GEJ/G AC Neoadjuvant tremelimumab + durvalumab followed by surgery (cohort 1) and nonoperative management (cohort 2) pCR (cohort 1): ongoing

2-y complete clinical response rate (cohort 2): ongoing NCT04817826
28,30

Abbreviations: AC, adenocarcinoma; chemo, chemotherapy; CRT, chemoradiotherapy; DFS, disease-free survival; dMMR, mismatch repair deficient; E, esophagus; ECF/X, epirubicin/cisplatin/5-fluorouracil or capecitabine; EFS, event-free survival; EOX, epirubicin/oxaliplatin/capecitabine; FLOT, 5-FU/leucovorin/oxaliplatin/docetaxel; FOLFOX, 5-fluorouracil/leucovorin/oxaliplatin; G, gastric; GEJ, gastroesophageal junction; HR, hazard ratio; MSI-H, microsatellite instability-high; OR, odds ratio; OS, overall survival; pCR, pathologic complete response; SCC, squamous cell carcinoma.

Table 4.

Selected Clinical Trials of Targeted Therapies for Resectable E/GEJ AC

Trial Phase Study Population Treatment Arms Primary Endpoint and Efficacy Reference
Trials incorporating HER2-targeted therapies
RTOG1010 III HER2+ E/GEJ AC CRT (carboplatin + paclitaxel) with or without trastuzumab DFS: no difference 19.6 mo with CRT + trastuzumab vs 14.2 mo with CRT alone (HR, 0.99; 95% CI, 0.71–1.39; P=.97) 34
PETRARCA II/III HER2+ E/GEJ AC Perioperative chemo (FLOT) + trastuzumab + pertuzumab vs chemo alone pCR rate: favors FLOT + trastuzumab + pertuzumab arm 35% vs 12% (P=.019) Did not proceed to phase III 40
PHERFLOT II HER2 + EG AC Pembrolizumab + FLOT + trastuzumab pCR and 2-y DFS: ongoing NCT05504720 56
EPOC2003 II HER2+ or HER2-low GEJ/G AC Neoadjuvant trastuzumab deruxtecan Major pathologic response: ongoing NCT05034887 58
Trials incorporating EGFR-targeted therapies
RTOG 0436 III E SCC and AC (62% AC) Neoadjuvant CRT with weekly cetuximab vs CRT OS: no benefit HR, 0.90; 95% CI, 0.70–1.16; P=.47 37
SAKK 75/08 III E/GEJ AC and SCC (64% AC) Neoadjuvant chemo (cisplatin + docetaxel) + CRT with pre- and postoperative cetuximab vs chemo + CRT PFS: no benefit 2.9 y cetuximab vs 2.0 y control; HR, 0.79; 95% CI, 0.58–1.07; P=.13 36
Neoadjuvant and perioperative trials incorporating VEGF/VEGFR2-targeted therapies
ST03 II/III GEJ/G AC Perioperative chemo (ECX) + bevacizumab vs chemo alone OS (phase III): no benefit HR, 1.01; 95% CI, 0.61–1.67 38
RAMSES/FLOT7 II/III GEJ/G AC Perioperative chemo (FLOT) vs FLOT + ramucirumab Major pathologic response (phase II): no benefit 29% FLOT vs 26% FLOT + ramucirumab Did not proceed to phase III 57

Abbreviations: AC, adenocarcinoma; chemo, chemotherapy; CRT, chemoradiotherapy; DFS, disease-free survival; E, esophagus; ECX, epirubicin/cisplatin/capecitabine; FLOT, 5-FU/leucovorin/oxaliplatin/docetaxel; G, gastric; GEJ, gastroesophageal junction; HR, hazard ratio; OS, overall survival; pCR, pathologic complete response; PFS, progression-free survival; SCC, squamous cell carcinoma.

Currently, perioperative ICI alone is not recommended, except in the rare subset of patients with mismatch repair deficient (dMMR) or microsatellite instability-high (MSI-H) tumors (<2% EAC).2530 The relatively limited benefit of perioperative chemotherapy, coupled with the activity of ICIs in the advanced setting, provides initial support for this strategy, and underscores the importance of testing all esophagogastric adenocarcinomas for dMMR and/or MSI-H.10,25,26,31 Published data reviewed by Strickland et al27 support neoadjuvant ICI in patients with dMMR/MSI-H esophagogastric cancers, based on pCR rates of approximately 60% and encouraging EFS data.28,29 The ongoing phase II INFINITY trial includes a cohort with a clinical response to preoperative ICI and will evaluate nonoperative management.30 Although available data on perioperative ICI in dMMR/MSI-H tumors are encouraging, the current evidence is insufficient to broadly recommend nonoperative management following neoadjuvant ICI. Ongoing trials in patients with dMMR/MRI-H tumors (ClinicalTrials.gov identifiers: NCT04817826, NCT04062656, NCT04795661, NCT04736485), along with subset analyses from the ongoing KEYNOTE-585, MATTERHORN, and DANTE trials, may further inform the optimal strategy.17,23,24

Targeted Therapies

The therapeutic landscape for advanced esophagogastric cancer has rapidly expanded with the introduction of therapies targeting HER2, VEGF, PD1/PD-L1, and Claudin 18.2.19,3240 However, no targeted therapy has proven beneficial in patients with resectable EAC (Table 4).

HER2

HER2 overexpression or amplification occurs in approximately 23% and 11% of esophagogastric cancers. Trastuzumab, a monoclonal antibody targeting HER2, combined with frontline chemotherapy, improves survival in HER2-positive advanced EAC.33,41 Two trials testing HER2-directed therapy in resectable EAC showed no significant benefit (Table 4). RTOG 1010 compared neoadjuvant CRT with trastuzumab, followed by adjuvant trastuzumab, versus CRT alone in patients with HER2-positive resectable EAC.34 DFS was numerically longer in the trastuzumab group, but the benefit was not statistically significant. PETRARCA added trastuzumab and pertuzumab, another monoclonal antibody against HER2, to perioperative FLOT.35 This strategy improved pCR rates (35% vs 12%; P=.019), but the trial was stopped early due to slow accrual. Ongoing studies will further explore strategies incorporating HER2-directed therapy into the perioperative setting for patients with localized HER2-positive esophagogastric adenocarcinoma.

EGFR, VEGF

Two phase III trials, RTOG 0436 and SAKK 75/08, examined the benefit of adding the anti-EGFR antibody cetuximab to neoadjuvant chemoradiation in esophageal cancer; however, neither demonstrated a survival benefit.36,37 The addition of the anti-VEGFA antibody bevacizumab to perioperative chemotherapy in patients with resectable gastric and esophageal tumors in the phase II STO trial did not improve OS.38

Individualizing Multimodality Management of Resectable EAC

Although the pivotal trials discussed herein inform the current approach to the management of EAC, several key questions remain, particularly for underrepresented groups in clinical trials. Racial and ethnic disparities exist in the presentation, treatment, and outcomes of patients with EAC. Non-White patients often present with more advanced disease, and stage-for-stage, they are less likely to undergo surgery.42 Notably, neither CROSS, TOPGEAR, nor ESOPEC report on the race and ethnicity of the subjects enrolled.2,5,15 Given the geographic locations of these studies (Western Europe, Canada, Australia, and New Zealand), African American, American Indian, and Hispanic patients are likely underrepresented. In the US-based CALGB 80803, 93% of the 225 patients were White, and 97% were non-Hispanic.14 This underrepresentation limits the generalizability of the results. Expanding enrollment to a more diverse patient population to better represent the population affected by the disease is critical, as are strategies that ensure all patients have access to appropriate multidisciplinary care.

Although ESOPEC establishes perioperative FLOT as the more effective regimen for fit patients undergoing resection for EAC, several important clinical questions remain. The most effective approach for patients who cannot tolerate FLOT is unknown. Neo-AEGIS11 suggests that CRT is more effective than perioperative ECX, but based on CALGB 80803,14 FOLFOX may be a more effective chemotherapy backbone to consider for patients who cannot tolerate FLOT. ESOPEC5 does not address the management of tumors that do not respond to preoperative therapy, as all patients in the perioperative chemotherapy group were assigned to receive postoperative FLOT, regardless of response. ESOPEC also highlights the challenge of delivering postoperative therapy following esophagectomy. Only 63% of patients in the perioperative FLOT arm began postoperative therapy, raising the question of whether a total neoadjuvant approach could further improve efficacy.

Another open question is the role of nonoperative management for patients who may not tolerate resection due to advanced age, frailty, or medical comorbidities, or for those who experience a complete clinical response to preoperative therapy. Given the increasing uptake of organ preservation approaches in rectal cancer and the risk of morbidity and impact on quality of life of esophagectomy, there is increasing interest in the nonoperative management of esophageal cancer. Although growing evidence supports the nonoperative management of esophageal SCC, there are currently fewer data for EAC. Definitive CRT should be considered for patients unable to undergo resection, and the benefit of incorporating pembrolizumab into the CRT paradigm is being evaluated in the ongoing KEYNOTE-975 study.39

Another major challenge in treating esophagogastric cancers is managing patients whose tumors do not respond to preoperative therapy. In the MAGIC trial,7 patients with node-positive disease at resection survived a median of 16 months. However, in practice, the standard paradigm is to resume the same chemotherapy regimen used in the preoperative setting.43 Conversely, a proportion of patients who have exceptional responses to neoadjuvant therapy may be overtreated with adjuvant therapy. FDG-PET and circulating tumor DNA (ctDNA) are proven prognostic tools that could inform adaptive perioperative strategies. The ability to detect the presence of residual disease noninvasively in the preoperative and postoperative settings could help guide risk-adapted approaches for resectable EAC.

FDG-PET

FDG-PET imaging is routinely used to stage and assess the response to neoadjuvant therapy for esophageal cancer.44 In esophagogastric cancer treated with neoadjuvant therapy, a reduction in the SUV ≥35% from baseline is associated with a histologic response and improved survival.4547 Metabolic nonresponders represent a very-high-risk group, with an expected pCR rate of only 4%.48 Using the study design described earlier,14 CALGB 80803 evaluated whether adapting preoperative therapy based on PET response to induction chemotherapy could improve the pCR rate in PET nonresponders. The study met its primary endpoint (pCR rate in PET nonresponders), showing a pCR rate of 18% for FOLFOX to CP and 20% for CP to FOLFOX, compared with the expected rate of only 4%.48 These results demonstrate that response assessment using PET following neoadjuvant chemotherapy and adapting therapy based on treatment response is a viable and promising strategy.

ctDNA

ctDNA is an emerging tool with significant potential to dynamically evaluate cancer. However, available evidence in esophageal cancer is largely limited to retrospective data and post hoc analyses, often from heterogeneous cohorts. Two prospective studies of patients with EAC have shown that ctDNA detection following neoadjuvant CRT completion was associated with shorter DFS.49,50 In patients with EAC treated with neoadjuvant chemotherapy and resection, 90% of those with detectable ctDNA following resection went on to develop recurrence, with significantly shorter survival in patients with detectable ctDNA (10.0 vs 20.9 months; HR, 5.55; 95% CI, 2.42–12.71; P=.0003).50 In another analysis of patients with stage I–III esophagogastric cancer (adenocarcinoma and SCC), ctDNA detection within 16 weeks of resection was associated with significantly shorter DFS in all patients, including the subset with esophageal cancer (HR, 55.6; 95% CI, 6.9–7,198.7; P<.001).51

Though largely focused on gastric and GEJ adenocarcinoma, additional studies show a significant association between ctDNA dynamics and clinical and pathologic outcomes.52,53 In a cohort of patients with gastric and GEJ adenocarcinoma, early ctDNA clearance during neoadjuvant therapy was associated with the longest DFS and OS. Conversely, ctDNA persistence at mid-neoadjuvant therapy, postneoadjuvant therapy, and postoperative time points was associated with incrementally shorter recurrence-free survival and OS.52 In an analysis of ctDNA dynamics among a subset of patients treated with preoperative chemotherapy in the CRITICS trial, detection of ctDNA following resection was significantly associated with recurrence (median EFS, 18.7 months vs not reached; HR, 21.8; P<.001).53 In both studies, the presence or absence of ctDNA following neoadjuvant therapy strongly correlated with the degree of histologic regression in the resected tumor.

Although preliminary, these data suggest that ctDNA has significant potential to inform risk-adapted perioperative strategies for patients with localized EAC. However, data are insufficient to recommend the routine use of ctDNA for guiding treatment decisions in EAC at this time. This caution stems from the lack of evidence demonstrating that ctDNA-guided strategies translate to improved patient outcomes. Furthermore, the added cost of ctDNA testing without established clinical benefit should preclude its routine clinical use. Therefore, well-designed, prospective clinical trials that are specifically powered to evaluate the clinical impact of ctDNA-guided approaches are essential for validating its utility.

A phase II trial in development will evaluate a ctDNA-guided approach to postoperative therapy in patients with locally advanced esophagogastric adenocarcinoma who received preoperative chemotherapy and resection. Following resection, patients with undetectable ctDNA will receive de-escalated postoperative therapy with capecitabine, whereas those with detectable ctDNA will be randomized to either standard postoperative FLOT or salvage therapy. The salvage therapy regimen will consist of a proven regimen used in the frontline setting for advanced esophagogastric adenocarcinoma tailored to the individual patient’s tumor, based on PD-L1, HER2, and Claudin 18.2 expression. The primary objectives are to demonstrate that in patients with undetectable ctDNA, de-escalated therapy does not compromise DFS, and in patients with detectable ctDNA, salvage therapy improves DFS compared with standard postoperative FLOT therapy.

Conclusions

Multimodality therapy has led to improved survival for patients with EAC, with recent data from landmark trials establishing the importance of effective systemic therapy in improving survival for patients with EAC. The ESOPEC trial represents a significant milestone in the evolution of treatment for resectable EAC, highlighting the importance of effective systemic control in this disease. Despite these advancements, fewer than half of patients are cured with multimodality therapy, underscoring the urgent need for more effective strategies. Targeted therapies have significantly improved survival in the metastatic setting and are currently being explored in the localized, curative setting. Incorporating these advancements and novel approaches that individualize perioperative treatment to individual patients offers considerable promise for the future management of EAC.

References

  • 1.

    van der Zijden CJ, Eyck BM, van der Gaast A, et al. ChemoTherapy aNd chemoradioTherapy for adenocarcinoma of the OESophagus and esophagogastric junction with oligometastases: protocol of the TNT-OES-1 trial. Contemp Clin Trials Commun 2022;28:100934.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 2.

    van Hagen P, Hulshof MCCM, van Lanschot JJB, et al. Preoperative chemoradiotherapy for esophageal or junctional cancer. N Engl J Med 2012;366:20742084.

  • 3.

    Al-Batran SE, Homann N, Pauligk C, et al. Perioperative chemotherapy with fluorouracil plus leucovorin, oxaliplatin, and docetaxel versus fluorouracil or capecitabine plus cisplatin and epirubicin for locally advanced, resectable gastric or gastro-oesophageal junction adenocarcinoma (FLOT4): a randomised, phase 2/3 trial. Lancet 2019;393:19481957.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 4.

    Kelly RJ, Ajani JA, Kuzdzal J, et al. Adjuvant nivolumab in resected esophageal or gastroesophageal junction cancer. N Engl J Med 2021;384:11911203.

  • 5.

    Hoeppner J, Brunner T, Schmoor C, et al. Perioperative chemotherapy or preoperative chemoradiotherapy in esophageal cancer. N Engl J Med 2025;392:323335.

  • 6.

    Allum WH, Stenning SP, Bancewicz J, et al. Long-term results of a randomized trial of surgery with or without preoperative chemotherapy in esophageal cancer. J Clin Oncol 2009;27:50625067.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 7.

    Cunningham D, Allum WH, Stenning SP, et al. Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N Engl J Med 2006;355:1120.

  • 8.

    Shah MA, Kennedy EB, Catenacci DV, et al. Treatment of locally advanced esophageal carcinoma: ASCO guideline. J Clin Oncol 2020;38:26772694.

  • 9.

    Obermannová R, Alsina M, Cervantes A, et al. Oesophageal cancer: ESMO clinical practice guideline for diagnosis, treatment and follow-up. Ann Oncol 2022;33:9921004.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 10.

    Ajani JA, D’Amico TA, Bentrem DJ, et al. NCCN Clinical Practice Guidelines in Oncology: Esophageal and Esophagogastric Junction Cancers. Version 2.2023. Accessed March 24, 2024. To view the most recent version, visit https://www.nccn.org

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 11.

    Reynolds JV, Preston SR, O’Neill B, et al. Trimodality therapy versus perioperative chemotherapy in the management of locally advanced adenocarcinoma of the oesophagus and oesophagogastric junction (Neo-AEGIS): an open-label, randomised, phase 3 trial. Lancet Gastroenterol Hepatol 2023;8:10151027.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 12.

    Schulz C, Kullmann F, Kunzmann V, et al. NeoFLOT: multicenter phase II study of perioperative chemotherapy in resectable adenocarcinoma of the gastroesophageal junction or gastric adenocarcinoma—very good response predominantly in patients with intestinal type tumors. Int J Cancer 2015;137:678685.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 13.

    Wo JY, Clark JW, Eyler CE, et al. Results and molecular correlates from a pilot study of neoadjuvant induction FOLFIRINOX followed by chemoradiation and surgery for gastroesophageal adenocarcinomas. Clin Cancer Res 2021;27:63436353.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 14.

    Goodman KA, Ou FS, Hall NC, et al. Randomized phase II study of PET response–adapted combined modality therapy for esophageal cancer: mature results of the CALGB 80803 (Alliance) trial. J Clin Oncol 2021;39:28032815.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 15.

    Leong T, Smithers BM, Michael M, et al. Preoperative chemoradiotherapy for resectable gastric cancer. N Engl J Med 2024;391:18101821.

  • 16.

    Slagter AE, Jansen EPM, van Laarhoven HWM, et al. CRITICS-II: a multicentre randomised phase II trial of neo-adjuvant chemotherapy followed by surgery versus neo-adjuvant chemotherapy and subsequent chemoradiotherapy followed by surgery versus neo-adjuvant chemoradiotherapy followed by surgery in resectable gastric cancer. BMC Cancer 2018;18:877.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 17.

    Lorenzen S, Götze TO, Thuss-Patience P, et al. Perioperative atezolizumab plus fluorouracil, leucovorin, oxaliplatin, and docetaxel for resectable esophagogastric cancer: interim results from the randomized, multicenter, phase II/III DANTE/IKF-s633 trial. J Clin Oncol 2024;42:410420.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 18.

    Liu X, Jin J, Cai H, et al. Study protocol of a randomized phase III trial of comparing preoperative chemoradiation with preoperative chemotherapy in patients with locally advanced gastric cancer or esophagogastric junction adenocarcinoma: PREACT. BMC Cancer 2019;19:606.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 19.

    Janjigian YY, Ajani JA, Moehler M, et al. First-line nivolumab plus chemotherapy for advanced gastric, gastroesophageal junction, and esophageal adenocarcinoma: 3-year follow-up of the phase III CheckMate 649 trial. J Clin Oncol 2024;42:20122020.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 20.

    Sun JM, Shen L, Shah MA, et al. Pembrolizumab plus chemotherapy versus chemotherapy alone for first-line treatment of advanced oesophageal cancer (KEYNOTE-590): a randomised, placebo-controlled, phase 3 study. Lancet 2021;398:759771.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 21.

    Blum Murphy M, Xiao L, Patel VR, et al. Pathological complete response in patients with esophageal cancer after the trimodality approach: the association with baseline variables and survival—the University of Texas MD Anderson Cancer Center experience. Cancer 2017;123:41064113.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 22.

    Eads JR, Graham N, Gibson MK, et al. A phase II/III study of peri-operative nivolumab (nivo) and ipilimumab (ipi) in patients (pts) with locoregional esophageal (E) and gastroesophageal junction (GEJ) adenocarcinoma: results of the neoadjuvant pathologic complete response (pCR) rate (ECOG-ACRIN EA2174). J Clin Oncol 2024;42(Suppl 16):Abstract 4000.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 23.

    Shitara K, Rha SY, Wyrwicz LS, et al. Neoadjuvant and adjuvant pembrolizumab plus chemotherapy in locally advanced gastric or gastro-oesophageal cancer (KEYNOTE-585): an interim analysis of the multicentre, double-blind, randomised phase 3 study. Lancet Oncol 2024;25:212224.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 24.

    Oh DY, Janjigian YY, Al-Batran SE, et al. Pathological complete response (pCR) to durvalumab plus 5-fluorouracil, leucovorin, oxaliplatin and docetaxel (FLOT) in resectable gastric and gastroesophageal junction cancer (GC/GEJC): interim results of the global, phase III MATTERHORN study. Ann Oncol 2023;34:S15201521.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 25.

    Smyth EC, Wotherspoon A, Peckitt C, et al. Mismatch repair deficiency, microsatellite instability, and survival: an exploratory analysis of the Medical Research Council Adjuvant Gastric Infusional Chemotherapy (MAGIC) trial. JAMA Oncol 2017;3:11971203.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 26.

    Choi YY, Kim H, Shin SJ, et al. Microsatellite instability and programmed cell death-ligand 1 expression in stage II/III gastric cancer: post hoc analysis of the CLASSIC randomized controlled study. Ann Surg 2019;270:309316.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 27.

    Strickland MR, Lander EM, Gibson MK, et al. Gastroesophageal adenocarcinomas with defective mismatch repair: current knowledge and clinical management. J Natl Compr Canc Netw 2024;22:e237103.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 28.

    André T, Tougeron D, Piessen G, et al. Neoadjuvant nivolumab plus ipilimumab and adjuvant nivolumab in localized deficient mismatch repair/microsatellite instability–high gastric or esophagogastric junction adenocarcinoma: the GERCOR NEONIPIGA phase II study. J Clin Oncol 2023;41:255265.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 29.

    Ludford K, Ho WJ, Thomas JV, et al. Neoadjuvant pembrolizumab in localized microsatellite instability high/deficient mismatch repair solid tumors. J Clin Oncol 2023;41:21812190.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 30.

    Pietrantonio F, Raimondi A, Lonardi S, et al. INFINITY: a multicentre, single-arm, multi-cohort, phase II trial of tremelimumab and durvalumab as neoadjuvant treatment of patients with microsatellite instability-high (MSI) resectable gastric or gastroesophageal junction adenocarcinoma (GAC/GEJAC). J Clin Oncol 2023;41(Suppl 4):Abstract 358.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 31.

    Bang YJ, Van Cutsem E, Fuchs CS, et al. KEYNOTE-585: phase III study of perioperative chemotherapy with or without pembrolizumab for gastric cancer. Future Oncol 2019;15:943952.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 32.

    Lorenzen S, Biederstädt A, Ronellenfitsch U, et al. RACE-trial: neoadjuvant radiochemotherapy versus chemotherapy for patients with locally advanced, potentially resectable adenocarcinoma of the gastroesophageal junction – a randomized phase III joint study of the AIO, ARO and DGAV. BMC Cancer 2020;20:886.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 33.

    Janjigian YY, Kawazoe A, Bai Y, et al. Pembrolizumab plus trastuzumab and chemotherapy for HER2-positive gastric or gastro-oesophageal junction adenocarcinoma: interim analyses from the phase 3 KEYNOTE-811 randomised placebo-controlled trial. Lancet 2023;402:21972208.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 34.

    Safran HP, Winter K, Ilson DH, et al. Trastuzumab with trimodality treatment for oesophageal adenocarcinoma with HER2 overexpression (NRG Oncology/RTOG 1010): a multicentre, randomised, phase 3 trial. Lancet Oncol 2022;23:259269.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 35.

    Swain SM, Baselga J, Kim SB, et al. Pertuzumab, trastuzumab, and docetaxel in HER2-positive metastatic breast cancer. N Engl J Med 2015;372:724734.

  • 36.

    Ruhstaller T, Thuss-Patience P, Hayoz S, et al. Neoadjuvant chemotherapy followed by chemoradiation and surgery with and without cetuximab in patients with resectable esophageal cancer: a randomized, open-label, phase III trial (SAKK 75/08). Ann Oncol 2018;29:13861393.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 37.

    Suntharalingam M, Winter K, Ilson D, et al. Effect of the addition of cetuximab to paclitaxel, cisplatin, and radiation therapy for patients with esophageal cancer: the NRG Oncology RTOG 0436 phase 3 randomized clinical trial. JAMA Oncol 2017;3:15201528.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 38.

    Cunningham D, Stenning SP, Smyth EC, et al. Peri-operative chemotherapy with or without bevacizumab in operable oesophagogastric adenocarcinoma (UK Medical Research Council ST03): primary analysis results of a multicentre, open-label, randomised phase 2-3 trial. Lancet Oncol 2017;18:357370.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 39.

    Shah MA, Bennouna J, Doi T, et al. KEYNOTE-975 study design: a phase III study of definitive chemoradiotherapy plus pembrolizumab in patients with esophageal carcinoma. Future Oncol 2021;17:11431153.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 40.

    Hofheinz RD, Haag GM, Ettrich TJ, et al. Perioperative trastuzumab and pertuzumab in combination with FLOT versus FLOT alone for HER2-positive resectable esophagogastric adenocarcinoma: final results of the PETRARCA multicenter randomized phase II trial of the AIO. J Clin Oncol 2020;38(Suppl 15):Abstract 4502.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 41.

    Bang YJ, Van Cutsem E, Feyereislova A, et al. 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;376:687697.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 42.

    Okereke IC, Westra J, Tyler D, et al. Disparities in esophageal cancer care based on race: a National Cancer Database analysis. Dis Esophagus 2022;35:doab083.

  • 43.

    Smyth EC, Fassan M, Cunningham D, et al. Effect of pathologic tumor response and nodal status on survival in the Medical Research Council Adjuvant Gastric Infusional Chemotherapy trial. J Clin Oncol 2016;34:27212727.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 44.

    Kurnit KC, Dumbrava EEI, Litzenburger B, et al. Precision oncology decision support: current approaches and strategies for the future. Clin Cancer Res 2018;24:27192731.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 45.

    Ott K, Weber WA, Lordick F, et al. Metabolic imaging predicts response, survival, and recurrence in adenocarcinomas of the esophagogastric junction. J Clin Oncol 2006;24:46924698.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 46.

    Ilson DH, Minsky BD, Ku GY, et al. Phase 2 trial of induction and concurrent chemoradiotherapy with weekly irinotecan and cisplatin followed by surgery for esophageal cancer. Cancer 2012;118:28202827.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 47.

    Weber WA, Ott K, Becker K, et al. Prediction of response to preoperative chemotherapy in adenocarcinomas of the esophagogastric junction by metabolic imaging. J Clin Oncol 2001;19:30583065.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 48.

    Zum Büschenfelde CM, Herrmann K, Schuster T, et al. (18)F-FDG PET-guided salvage neoadjuvant radiochemotherapy of adenocarcinoma of the esophagogastric junction: the MUNICON II trial. J Nucl Med 2011;52:11891196.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 49.

    Azad TD, Chaudhuri AA, Fang P, et al. Circulating tumor DNA analysis for detection of minimal residual disease after chemoradiotherapy for localized esophageal cancer. Gastroenterology 2020;158:494505.e6.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 50.

    Ococks E, Frankell AM, Masque Soler N, et al. Longitudinal tracking of 97 esophageal adenocarcinomas using liquid biopsy sampling. Ann Oncol 2021;32:522532.

  • 51.

    Huffman BM, Aushev VN, Budde GL, et al. Analysis of circulating tumor DNA to predict risk of recurrence in patients with esophageal and gastric cancers. JCO Precis Oncol 2022;6:e2200420.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 52.

    Zaanan A, Didelot A, Broudin C, et al. Longitudinal circulating tumor DNA (ctDNA) analysis during treatment (Tx) of locally advanced resectable (LAR) gastric or gastroesophageal junction (G/GEJ) adenocarcinoma (ADENOCA): the PLAGAST prospective biomarker study. J Clin Oncol 2024;42(Suppl 16):Abstract 4028.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 53.

    Leal A, van Grieken NCT, Palsgrove DN, et al. White blood cell and cell-free DNA analyses for detection of residual disease in gastric cancer. Nat Commun 2020;11:525.

  • 54.

    Al-Batran SE, Hofheinz RD, Pauligk C, et al. Histopathological regression after neoadjuvant docetaxel, oxaliplatin, fluorouracil, and leucovorin versus epirubicin, cisplatin, and fluorouracil or capecitabine in patients with resectable gastric or gastro-oesophageal junction adenocarcinoma (FLOT4-AIO): results from the phase 2 part of a multicentre, open-label, randomised phase 2/3 trial. Lancet Oncol 2016;17:16971708.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 55.

    Cats A, Jansen EPM, van Grieken NCT, et al. Chemotherapy versus chemoradiotherapy after surgery and preoperative chemotherapy for resectable gastric cancer (CRITICS): an international, open-label, randomised phase 3 trial. Lancet Oncol 2018;19:616628.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 56.

    Tintelnot J, Stein A, Al-Batran SE, et al. Pembrolizumab and trastuzumab in combination with FLOT in the perioperative treatment of HER2-positive, localized esophagogastric adenocarcinoma—a phase II trial of the AIO study group (AIO STO 0321). Front Oncol 2023;13:1272175.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 57.

    Goetze TO, Hofheinz RD, Schmalenberg H, et al. Perioperative ramucirumab in combination with FLOT versus FLOT alone for resectable esophagogastric adenocarcinoma (RAMSES/FLOT7) with high rate of signet cell component: final results of the multicenter, randomized phase II/III trial of the German AIO and Italian GOIM. J Clin Oncol 2022;40(Suppl 16):Abstract 4042.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 58.

    Takahari D, Kawazoe A, Machida N, et al. Phase 2 study of trastuzumab deruxtecan in the neoadjuvant treatment for patients with HER2-positive gastric and gastroesophageal junction adenocarcinoma (EPOC2003). J Clin Oncol 2022;40(Suppl 16):Abstract TPS4161.

    • PubMed
    • Search Google Scholar
    • Export Citation

Submitted November 11, 2024; final revision received March 19, 2025; accepted for publication March 20, 2025.

Disclosures: Dr. Klute has disclosed receiving grant/research support from and serving as a scientific advisor for Bristol Myers Squibb; and serving as a consultant for Daiichi Sankyo, Pfizer, Genus Oncology, and Oncolys BioPharma. Dr. Shah has disclosed receiving grant/research support from Merck, Bristol Myers Squibb, Oncolys BioPharma; serving as a principal investigator for Merck; serving as a scientific advisor for Merck and Bristol Myers Squibb; and serving as a consultant for Takeda Pharmaceuticals and Astellas Pharmaceuticals.

Correspondence: Manish A. Shah, MD, Department of Medicine, Weill Cornell Medicine, 1305 York Avenue, Y1247, New York, NY 10021. Email: mas9313@med.cornell.edu
  • Collapse
  • Expand
  • 1.

    van der Zijden CJ, Eyck BM, van der Gaast A, et al. ChemoTherapy aNd chemoradioTherapy for adenocarcinoma of the OESophagus and esophagogastric junction with oligometastases: protocol of the TNT-OES-1 trial. Contemp Clin Trials Commun 2022;28:100934.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 2.

    van Hagen P, Hulshof MCCM, van Lanschot JJB, et al. Preoperative chemoradiotherapy for esophageal or junctional cancer. N Engl J Med 2012;366:20742084.

  • 3.

    Al-Batran SE, Homann N, Pauligk C, et al. Perioperative chemotherapy with fluorouracil plus leucovorin, oxaliplatin, and docetaxel versus fluorouracil or capecitabine plus cisplatin and epirubicin for locally advanced, resectable gastric or gastro-oesophageal junction adenocarcinoma (FLOT4): a randomised, phase 2/3 trial. Lancet 2019;393:19481957.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 4.

    Kelly RJ, Ajani JA, Kuzdzal J, et al. Adjuvant nivolumab in resected esophageal or gastroesophageal junction cancer. N Engl J Med 2021;384:11911203.

  • 5.

    Hoeppner J, Brunner T, Schmoor C, et al. Perioperative chemotherapy or preoperative chemoradiotherapy in esophageal cancer. N Engl J Med 2025;392:323335.

  • 6.

    Allum WH, Stenning SP, Bancewicz J, et al. Long-term results of a randomized trial of surgery with or without preoperative chemotherapy in esophageal cancer. J Clin Oncol 2009;27:50625067.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 7.

    Cunningham D, Allum WH, Stenning SP, et al. Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N Engl J Med 2006;355:1120.

  • 8.

    Shah MA, Kennedy EB, Catenacci DV, et al. Treatment of locally advanced esophageal carcinoma: ASCO guideline. J Clin Oncol 2020;38:26772694.

  • 9.

    Obermannová R, Alsina M, Cervantes A, et al. Oesophageal cancer: ESMO clinical practice guideline for diagnosis, treatment and follow-up. Ann Oncol 2022;33:9921004.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 10.

    Ajani JA, D’Amico TA, Bentrem DJ, et al. NCCN Clinical Practice Guidelines in Oncology: Esophageal and Esophagogastric Junction Cancers. Version 2.2023. Accessed March 24, 2024. To view the most recent version, visit https://www.nccn.org

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 11.

    Reynolds JV, Preston SR, O’Neill B, et al. Trimodality therapy versus perioperative chemotherapy in the management of locally advanced adenocarcinoma of the oesophagus and oesophagogastric junction (Neo-AEGIS): an open-label, randomised, phase 3 trial. Lancet Gastroenterol Hepatol 2023;8:10151027.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 12.

    Schulz C, Kullmann F, Kunzmann V, et al. NeoFLOT: multicenter phase II study of perioperative chemotherapy in resectable adenocarcinoma of the gastroesophageal junction or gastric adenocarcinoma—very good response predominantly in patients with intestinal type tumors. Int J Cancer 2015;137:678685.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 13.

    Wo JY, Clark JW, Eyler CE, et al. Results and molecular correlates from a pilot study of neoadjuvant induction FOLFIRINOX followed by chemoradiation and surgery for gastroesophageal adenocarcinomas. Clin Cancer Res 2021;27:63436353.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 14.

    Goodman KA, Ou FS, Hall NC, et al. Randomized phase II study of PET response–adapted combined modality therapy for esophageal cancer: mature results of the CALGB 80803 (Alliance) trial. J Clin Oncol 2021;39:28032815.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 15.

    Leong T, Smithers BM, Michael M, et al. Preoperative chemoradiotherapy for resectable gastric cancer. N Engl J Med 2024;391:18101821.

  • 16.

    Slagter AE, Jansen EPM, van Laarhoven HWM, et al. CRITICS-II: a multicentre randomised phase II trial of neo-adjuvant chemotherapy followed by surgery versus neo-adjuvant chemotherapy and subsequent chemoradiotherapy followed by surgery versus neo-adjuvant chemoradiotherapy followed by surgery in resectable gastric cancer. BMC Cancer 2018;18:877.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 17.

    Lorenzen S, Götze TO, Thuss-Patience P, et al. Perioperative atezolizumab plus fluorouracil, leucovorin, oxaliplatin, and docetaxel for resectable esophagogastric cancer: interim results from the randomized, multicenter, phase II/III DANTE/IKF-s633 trial. J Clin Oncol 2024;42:410420.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 18.

    Liu X, Jin J, Cai H, et al. Study protocol of a randomized phase III trial of comparing preoperative chemoradiation with preoperative chemotherapy in patients with locally advanced gastric cancer or esophagogastric junction adenocarcinoma: PREACT. BMC Cancer 2019;19:606.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 19.

    Janjigian YY, Ajani JA, Moehler M, et al. First-line nivolumab plus chemotherapy for advanced gastric, gastroesophageal junction, and esophageal adenocarcinoma: 3-year follow-up of the phase III CheckMate 649 trial. J Clin Oncol 2024;42:20122020.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 20.

    Sun JM, Shen L, Shah MA, et al. Pembrolizumab plus chemotherapy versus chemotherapy alone for first-line treatment of advanced oesophageal cancer (KEYNOTE-590): a randomised, placebo-controlled, phase 3 study. Lancet 2021;398:759771.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 21.

    Blum Murphy M, Xiao L, Patel VR, et al. Pathological complete response in patients with esophageal cancer after the trimodality approach: the association with baseline variables and survival—the University of Texas MD Anderson Cancer Center experience. Cancer 2017;123:41064113.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 22.

    Eads JR, Graham N, Gibson MK, et al. A phase II/III study of peri-operative nivolumab (nivo) and ipilimumab (ipi) in patients (pts) with locoregional esophageal (E) and gastroesophageal junction (GEJ) adenocarcinoma: results of the neoadjuvant pathologic complete response (pCR) rate (ECOG-ACRIN EA2174). J Clin Oncol 2024;42(Suppl 16):Abstract 4000.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 23.

    Shitara K, Rha SY, Wyrwicz LS, et al. Neoadjuvant and adjuvant pembrolizumab plus chemotherapy in locally advanced gastric or gastro-oesophageal cancer (KEYNOTE-585): an interim analysis of the multicentre, double-blind, randomised phase 3 study. Lancet Oncol 2024;25:212224.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 24.

    Oh DY, Janjigian YY, Al-Batran SE, et al. Pathological complete response (pCR) to durvalumab plus 5-fluorouracil, leucovorin, oxaliplatin and docetaxel (FLOT) in resectable gastric and gastroesophageal junction cancer (GC/GEJC): interim results of the global, phase III MATTERHORN study. Ann Oncol 2023;34:S15201521.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 25.

    Smyth EC, Wotherspoon A, Peckitt C, et al. Mismatch repair deficiency, microsatellite instability, and survival: an exploratory analysis of the Medical Research Council Adjuvant Gastric Infusional Chemotherapy (MAGIC) trial. JAMA Oncol 2017;3:11971203.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 26.

    Choi YY, Kim H, Shin SJ, et al. Microsatellite instability and programmed cell death-ligand 1 expression in stage II/III gastric cancer: post hoc analysis of the CLASSIC randomized controlled study. Ann Surg 2019;270:309316.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 27.

    Strickland MR, Lander EM, Gibson MK, et al. Gastroesophageal adenocarcinomas with defective mismatch repair: current knowledge and clinical management. J Natl Compr Canc Netw 2024;22:e237103.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 28.

    André T, Tougeron D, Piessen G, et al. Neoadjuvant nivolumab plus ipilimumab and adjuvant nivolumab in localized deficient mismatch repair/microsatellite instability–high gastric or esophagogastric junction adenocarcinoma: the GERCOR NEONIPIGA phase II study. J Clin Oncol 2023;41:255265.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 29.

    Ludford K, Ho WJ, Thomas JV, et al. Neoadjuvant pembrolizumab in localized microsatellite instability high/deficient mismatch repair solid tumors. J Clin Oncol 2023;41:21812190.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 30.

    Pietrantonio F, Raimondi A, Lonardi S, et al. INFINITY: a multicentre, single-arm, multi-cohort, phase II trial of tremelimumab and durvalumab as neoadjuvant treatment of patients with microsatellite instability-high (MSI) resectable gastric or gastroesophageal junction adenocarcinoma (GAC/GEJAC). J Clin Oncol 2023;41(Suppl 4):Abstract 358.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 31.

    Bang YJ, Van Cutsem E, Fuchs CS, et al. KEYNOTE-585: phase III study of perioperative chemotherapy with or without pembrolizumab for gastric cancer. Future Oncol 2019;15:943952.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 32.

    Lorenzen S, Biederstädt A, Ronellenfitsch U, et al. RACE-trial: neoadjuvant radiochemotherapy versus chemotherapy for patients with locally advanced, potentially resectable adenocarcinoma of the gastroesophageal junction – a randomized phase III joint study of the AIO, ARO and DGAV. BMC Cancer 2020;20:886.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 33.

    Janjigian YY, Kawazoe A, Bai Y, et al. Pembrolizumab plus trastuzumab and chemotherapy for HER2-positive gastric or gastro-oesophageal junction adenocarcinoma: interim analyses from the phase 3 KEYNOTE-811 randomised placebo-controlled trial. Lancet 2023;402:21972208.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 34.

    Safran HP, Winter K, Ilson DH, et al. Trastuzumab with trimodality treatment for oesophageal adenocarcinoma with HER2 overexpression (NRG Oncology/RTOG 1010): a multicentre, randomised, phase 3 trial. Lancet Oncol 2022;23:259269.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 35.

    Swain SM, Baselga J, Kim SB, et al. Pertuzumab, trastuzumab, and docetaxel in HER2-positive metastatic breast cancer. N Engl J Med 2015;372:724734.

  • 36.

    Ruhstaller T, Thuss-Patience P, Hayoz S, et al. Neoadjuvant chemotherapy followed by chemoradiation and surgery with and without cetuximab in patients with resectable esophageal cancer: a randomized, open-label, phase III trial (SAKK 75/08). Ann Oncol 2018;29:13861393.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 37.

    Suntharalingam M, Winter K, Ilson D, et al. Effect of the addition of cetuximab to paclitaxel, cisplatin, and radiation therapy for patients with esophageal cancer: the NRG Oncology RTOG 0436 phase 3 randomized clinical trial. JAMA Oncol 2017;3:15201528.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 38.

    Cunningham D, Stenning SP, Smyth EC, et al. Peri-operative chemotherapy with or without bevacizumab in operable oesophagogastric adenocarcinoma (UK Medical Research Council ST03): primary analysis results of a multicentre, open-label, randomised phase 2-3 trial. Lancet Oncol 2017;18:357370.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 39.

    Shah MA, Bennouna J, Doi T, et al. KEYNOTE-975 study design: a phase III study of definitive chemoradiotherapy plus pembrolizumab in patients with esophageal carcinoma. Future Oncol 2021;17:11431153.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 40.

    Hofheinz RD, Haag GM, Ettrich TJ, et al. Perioperative trastuzumab and pertuzumab in combination with FLOT versus FLOT alone for HER2-positive resectable esophagogastric adenocarcinoma: final results of the PETRARCA multicenter randomized phase II trial of the AIO. J Clin Oncol 2020;38(Suppl 15):Abstract 4502.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 41.

    Bang YJ, Van Cutsem E, Feyereislova A, et al. 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;376:687697.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 42.

    Okereke IC, Westra J, Tyler D, et al. Disparities in esophageal cancer care based on race: a National Cancer Database analysis. Dis Esophagus 2022;35:doab083.

  • 43.

    Smyth EC, Fassan M, Cunningham D, et al. Effect of pathologic tumor response and nodal status on survival in the Medical Research Council Adjuvant Gastric Infusional Chemotherapy trial. J Clin Oncol 2016;34:27212727.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 44.

    Kurnit KC, Dumbrava EEI, Litzenburger B, et al. Precision oncology decision support: current approaches and strategies for the future. Clin Cancer Res 2018;24:27192731.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 45.

    Ott K, Weber WA, Lordick F, et al. Metabolic imaging predicts response, survival, and recurrence in adenocarcinomas of the esophagogastric junction. J Clin Oncol 2006;24:46924698.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 46.

    Ilson DH, Minsky BD, Ku GY, et al. Phase 2 trial of induction and concurrent chemoradiotherapy with weekly irinotecan and cisplatin followed by surgery for esophageal cancer. Cancer 2012;118:28202827.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 47.

    Weber WA, Ott K, Becker K, et al. Prediction of response to preoperative chemotherapy in adenocarcinomas of the esophagogastric junction by metabolic imaging. J Clin Oncol 2001;19:30583065.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 48.

    Zum Büschenfelde CM, Herrmann K, Schuster T, et al. (18)F-FDG PET-guided salvage neoadjuvant radiochemotherapy of adenocarcinoma of the esophagogastric junction: the MUNICON II trial. J Nucl Med 2011;52:11891196.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 49.

    Azad TD, Chaudhuri AA, Fang P, et al. Circulating tumor DNA analysis for detection of minimal residual disease after chemoradiotherapy for localized esophageal cancer. Gastroenterology 2020;158:494505.e6.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 50.

    Ococks E, Frankell AM, Masque Soler N, et al. Longitudinal tracking of 97 esophageal adenocarcinomas using liquid biopsy sampling. Ann Oncol 2021;32:522532.

  • 51.

    Huffman BM, Aushev VN, Budde GL, et al. Analysis of circulating tumor DNA to predict risk of recurrence in patients with esophageal and gastric cancers. JCO Precis Oncol 2022;6:e2200420.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 52.

    Zaanan A, Didelot A, Broudin C, et al. Longitudinal circulating tumor DNA (ctDNA) analysis during treatment (Tx) of locally advanced resectable (LAR) gastric or gastroesophageal junction (G/GEJ) adenocarcinoma (ADENOCA): the PLAGAST prospective biomarker study. J Clin Oncol 2024;42(Suppl 16):Abstract 4028.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 53.

    Leal A, van Grieken NCT, Palsgrove DN, et al. White blood cell and cell-free DNA analyses for detection of residual disease in gastric cancer. Nat Commun 2020;11:525.

  • 54.

    Al-Batran SE, Hofheinz RD, Pauligk C, et al. Histopathological regression after neoadjuvant docetaxel, oxaliplatin, fluorouracil, and leucovorin versus epirubicin, cisplatin, and fluorouracil or capecitabine in patients with resectable gastric or gastro-oesophageal junction adenocarcinoma (FLOT4-AIO): results from the phase 2 part of a multicentre, open-label, randomised phase 2/3 trial. Lancet Oncol 2016;17:16971708.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 55.

    Cats A, Jansen EPM, van Grieken NCT, et al. Chemotherapy versus chemoradiotherapy after surgery and preoperative chemotherapy for resectable gastric cancer (CRITICS): an international, open-label, randomised phase 3 trial. Lancet Oncol 2018;19:616628.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 56.

    Tintelnot J, Stein A, Al-Batran SE, et al. Pembrolizumab and trastuzumab in combination with FLOT in the perioperative treatment of HER2-positive, localized esophagogastric adenocarcinoma—a phase II trial of the AIO study group (AIO STO 0321). Front Oncol 2023;13:1272175.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 57.

    Goetze TO, Hofheinz RD, Schmalenberg H, et al. Perioperative ramucirumab in combination with FLOT versus FLOT alone for resectable esophagogastric adenocarcinoma (RAMSES/FLOT7) with high rate of signet cell component: final results of the multicenter, randomized phase II/III trial of the German AIO and Italian GOIM. J Clin Oncol 2022;40(Suppl 16):Abstract 4042.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 58.

    Takahari D, Kawazoe A, Machida N, et al. Phase 2 study of trastuzumab deruxtecan in the neoadjuvant treatment for patients with HER2-positive gastric and gastroesophageal junction adenocarcinoma (EPOC2003). J Clin Oncol 2022;40(Suppl 16):Abstract TPS4161.

    • PubMed
    • Search Google Scholar
    • Export Citation

Metrics

All Time Past Year Past 30 Days
Abstract Views 0 0 0
Full Text Views 913 913 913
PDF Downloads 682 682 682
EPUB Downloads 0 0 0