1. Introduction
Upper gastrointestinal (GI) malignancies, including cancers of the esophagus, gastroesophageal junction (GEJ), and stomach, represent a significant global healthcare burden. When combined, they rank as the fourth most common cancer and second leading cause of cancer-related deaths worldwide [
1]. At diagnosis, about half of the patients have potentially curable nonmetastatic disease, but the 5-year overall survival (OS) remains suboptimal at only 21.6% for esophageal cancer and 36.4% for gastric cancer [
2,
3]. Even with the most aggressive curative intent therapy, recurrence rate for locally advanced disease remains high at 36% [
4].
Within the last few years, data has emerged to define the roles of various modalities of therapy including surgery, radiation, and systemic therapies. While progress has been made, the optimal treatment strategy for locally advanced (cT2-4a, cN+/−, cM0) esophageal (EAC), GEJ (GEJ AC), and gastric adenocarcinomas (GAC) remains a subject of active investigation. In this review, we aim to provide a comprehensive overview of the most recent advancements in the management of locally advanced upper GI adenocarcinomas, focusing on multimodal therapies, preoperative/adjuvant/perioperative treatment approaches, and the integration of immunotherapy and targeted therapies.
2. Endoscopic and Surgical Strategies
For patients with very early lesions that have only mucosal invasion (cT1a), endoscopic resection can be considered for curative treatment. Endoscopic mucosal resection (EMR) is the more common technique used in the West, whereas endoscopic submucosal dissection (ESD) is used more commonly in Asia/Japan, though in recent years it is being adopted more in the West as well [
5]. Endoscopic resection can be combined with ablation to eradicate high-grade dysplasia and residual Barrett’s esophagus. If high-risk features are found on pathology, such as positive margins, presence of lymphovascular invasion, and poorly differentiated histology, then additional treatment such as surgery may be needed [
6]. Previously, patients with submucosal tumor extension (cT1b) were thought to have higher risk for lymph node metastases, so endoscopic resection was not used [
7]. However, recent data showed that with appropriate patient selection and more accurate staging via diagnostic mucosal resection, some patients with cT1b disease may be spared from esophagectomy by undergoing ESD [
8,
9,
10].
Surgical resection is the mainstay of curative treatment for most patients with locally advanced upper GI adenocarcinomas. For EAC, the standard surgical approach is esophagectomy with lymphadenectomy, which can be performed using an open, minimally invasive (robotic or laparoscopic), or hybrid technique [
11]. For GAC, the standard surgical approach is subtotal or total gastrectomy with D2 lymph node dissection (extensive removal of lymph nodes in stations 1–12a) [
12,
13]. The surgical approach for GEJ AC is based on the Siewert classification. Siewert type I is defined as tumors with an epicenter 1 to 5 cm above the GEJ, Siewert type II is defined as tumors with an epicenter 1 cm above to 2 cm below the GEJ, and Siewert type III is defined as tumors with an epicenter 2 to 5 cm below the GEJ [
14]. Siewert types I and II tumors are managed as esophageal cancers, while Siewert type III tumors are managed as gastric cancers. The goal of surgeries is to obtain negative margins (R0) and adequate lymph node dissection.
While surgery is an important component of curative-intent therapy, resection alone does not achieve the best outcomes for patients with locally advanced disease. Preoperative and perioperative therapies have both been shown to improve overall survival compared to surgery alone [
15,
16,
17]. Because multimodality therapy is necessary to achieve the best outcomes, cases of locally advanced EAC, GEJ AC, and GAC should be reviewed at multidisciplinary tumor board. While most biomarkers used in the metastatic setting (such as programmed death-ligand 1 [PD-L1], claudin 18.2, and human epidermal growth factor receptor 2 [HER2]) do not currently impact treatment decisions, microsatellite instability (MSI) status should be assessed in all newly diagnosed cases > cT1a to determine the optimal treatment regimen.
3. Radiation
In the context of curative management, radiation is typically administered concurrently with chemotherapy to achieve improved tumor control and reduce risk of locoregional recurrence. Chemoradiation (CRT) has been studied in both the preoperative and adjuvant settings (
Table 1). For treatment of locally advanced EAC and GEJ AC, preoperative CRT was once considered comparable to perioperative chemotherapy. However, recent data from the ESOPEC and TOPGEAR trials have brought the role of preoperative CRT into question [
18,
19]. Moving forward, CRT may be reserved for more limited clinical indications, such as definitive treatment for patients who are not candidates for surgery, or as a salvage option for those who do not respond adequately to chemotherapy.
3.1. Preoperative Chemoradiation
The role for preoperative CRT in locally advanced EAC and GEJ AC was established by the CROSS trial [
15]. This phase 3 trial randomized patients to surgery alone (
n = 188) or CRT followed by surgery (
n = 178). CRT + surgery resulted in a higher R0 resection rate (92% vs. 69%,
p < 0.001), improved median OS (49.4 months vs. 24 months,
p = 0.003), and better disease-free survival (DFS) (not reached vs. 24.2 months,
p < 0.001). These results established preoperative CRT as one of the standard-of-care treatment approaches for locally advanced esophageal and GEJ cancers, and it has remained so over the last decade until recent results from the ESOPEC trial.
For locally advanced GAC, the role for preoperative CRT was explored in the TOPGEAR trial [
19]. TOPGEAR was an international phase 3 trial that randomized patients with resectable GAC to preoperative CRT + perioperative chemotherapy (
n = 286) or perioperative chemotherapy alone (
n = 288). While a higher percentage of patients achieved pathologic complete response (pCR) in the preoperative CRT group (17% vs. 8%), this did not translate to a significant difference in OS between the two groups. The median OS was 46 months in the preoperative CRT + perioperative chemotherapy group and 49 months in the perioperative chemotherapy group (hazard ratio 1.05). These results suggest that preoperative CRT provides no additional benefit over chemotherapy alone and that preoperative radiation has a very limited role in the management of locally advanced GAC.
3.2. Adjuvant Chemoradiation
The ARTIST, ARTIST 2, CRITICS, and MacDonald trials evaluated the role of adjuvant CRT in resected GAC. Both the ARTIST and ARTIST 2 trials compared postoperative chemotherapy versus CRT in GAC patients who underwent complete resection with D2 lymph node dissection [
20,
21]. Results from both trials showed that CRT did not significantly reduce the recurrence rate compared with chemotherapy alone. Likewise, the CRITICS trial, a randomized phase 3 study comparing perioperative chemotherapy (
n = 393) to preoperative chemotherapy + postoperative CRT (
n = 395), found no improvement in OS with postoperative CRT (37 months vs. 43 months, hazard ratio 1.01,
p = 0.90) [
22]. The only trial to demonstrate survival benefit for CRT in the adjuvant setting was the MacDonald trial, which was a phase 3 trial that randomly assigned patients with GAC/GEJ AC to receive either surgery + adjuvant CRT (
n = 281) or surgery alone (
n = 275) [
23]. The addition of adjuvant CRT significantly prolonged the median OS compared with surgery alone (36 months vs. 27 months,
p = 0.005, hazard ratio 1.52). Notably, only 10% of patients on this trial underwent D2 lymph node dissection, whereas 90% of patients had less than a D2 dissection. These results indicate the general lack of benefit of adjuvant CRT in patients who undergo adequate surgical resection, and adjuvant CRT only has a role in patients who undergo suboptimal surgeries.
4. Chemotherapy
4.1. Perioperative Chemotherapy
The MAGIC trial was the first landmark study to establish the perioperative approach for the management of locally advanced upper gastrointestinal adenocarcinomas (
Table 2) [
16]. Patients with resectable EAC, GEJ AC, and GAC were randomized to perioperative epirubicin + cisplatin + fluorouracil (ECF) x 6 cycles (
n = 250) versus surgery alone (
n = 253). In the perioperative arm, patients were treated with ECF three cycles preoperatively and three cycles postoperatively. Perioperative chemotherapy improved 5-year OS (36% vs. 23%,
p = 0.009) as well as progression free survival (PFS) (hazard ratio for progression 0.66,
p < 0.001). This trial demonstrated that perioperative chemotherapy was superior to surgery alone and is a valid treatment approach for patients with locally advanced disease.
FLOT4 was a phase 2/3 trial that compared the MAGIC regimen (
n = 360) with the perioperative fluorouracil + docetaxel + oxaliplatin (FLOT) regimen (
n = 356) [
24]. In the FLOT arm, patients were treated with four cycles of FLOT preoperatively and four cycles of FLOT postoperatively. The FLOT arm significantly improved the median OS (50 months vs. 35 months,
p = 0.012) and median DFS (30 months vs. 18 months,
p = 0.0036) when compared with the MAGIC arm. These results established perioperative FLOT as a new standard-of-care regimen for locally advanced GAC and GEJ AC.
4.2. Adjuvant Chemotherapy
Because perioperative chemotherapy is the current standard of care treatment approach in the West, most patients with locally advanced disease should receive preoperative chemotherapy. However, there are cases where patients undergo upfront surgery (such as patients with gastric outlet obstruction necessitating urgent surgery or patients who were understaged prior to upfront surgery), and adjuvant chemotherapy should be considered. In contrast, the management of gastroesophageal cancers in Asia differs from Western approaches, and upfront surgery remains the preferred strategy for treatment of locally advanced disease in many practices in Asia (Japan/Korea/China/Taiwan); therefore, adjuvant therapy is still widely utilized, and many studies of adjuvant chemotherapy have been conducted in Asia. Notably, because of the differences in practice, outcomes from Western studies should not be directly compared with those from Asian studies.
The role of adjuvant chemotherapy was explored in the ACTS-GC, JACCRO-GC07, and CLASSIC trials. ACTS-GC was a randomized phase 3 trial where patients with locally advanced GAC were treated with surgery + adjuvant S-1 (an oral fluoropyrimidine) (
n = 529) or surgery alone (
n = 530) [
25]. Adjuvant S1 significantly improved the 3-year OS compared with surgery alone (80.1% vs. 70.1%, hazard ratio 0.68,
p = 0.003). The JACCRO-GC07 trial was another phase 3 study in which patients with resected locally advanced GAC were randomized to receive either adjuvant S1 + docetaxel (
n = 453) or adjuvant S-1 alone (
n = 459) [
26]. The addition of docetaxel improved the 3-year relapse-free survival (67.7% vs. 57.4%,
p = 0.0008) and OS (77.7% vs. 71.2%,
p = 0.0076) over S-1 alone. The CLASSIC trial was a randomized phase 3 study that assessed capecitabine + oxaliplatin for 6 months postsurgery (
n = 520) versus surgery alone (
n = 515) [
27]. Adjuvant chemotherapy significantly increased the 3-year DFS compared with surgery only (74% vs. 59%,
p < 0.0001). Notably, all three trials required patients to undergo curative-intent D2 gastrectomy. Together, these trials support the use of adjuvant chemotherapy in patients who did not receive preoperative chemotherapy after undergoing D2 gastrectomy.
5. Preoperative Versus Perioperative Approach
Following the publication of results from the CROSS and MAGIC/FLOT4 trials, there was ongoing debate regarding the superiority of perioperative chemotherapy versus preoperative CRT. The NEO-AEGIS study was a randomized phase 3 trial that attempted to answer this question by enrolling patients with locally advanced EAC/GEJ AC to receive perioperative chemotherapy (MAGIC/FLOT;
n = 184) versus trimodality therapy (CROSS;
n = 178) [
28]. It is important to note that during the course of this trial, the standard of care perioperative chemotherapy regimen changed from ECF to FLOT, but only 15% of the perioperative arm received FLOT. Based on the data that were collected, the OS was similar (median OS 48.0 months vs. 49.2 months; 3-year OS 55% vs. 57%,
p = 0.82) between the perioperative and trimodality groups, but other outcomes (i.e., nodal downstaging, R0 resection, pathological complete response) favored the CROSS regimen. When these results were published, the consensus was that there was equipoise between the two treatment approaches.
However, the long-awaited ESOPEC results were finally published in January 2025 and more definitively addressed the question of FLOT vs. CROSS [
18]. ESOPEC randomized patients with locally advanced EAC/GEJ AC to receive perioperative FLOT (
n = 221) versus preoperative CRT (
n = 217). The 5-year OS (50.6% vs. 38.7%), median OS (66 months vs. 37 months), PFS (51.6% vs. 35.0%), and pCR rate (16.7% vs. 10.1%) were all significantly higher in the FLOT group than in the CROSS group. These results established perioperative FLOT as the preferred treatment approach in patients with resectable locally advanced upper GI adenocarcinoma. A few caveats must be taken into consideration when interpreting these results, including the absence of adjuvant nivolumab in the preoperative CRT group, the lower-than-expected treatment completion rate in the preoperative CRT group (67.7% in ESOPEC vs. 91% in CROSS), and the lower radiation dose used in ESOPEC (41.4 Gy) compared with the North American standard (50.4 Gy). Despite these caveats, the OS data were compelling, and perioperative FLOT has been adopted as the standard-of-care treatment regimen for locally advanced resectable EAC, GEJ AC, and GAC.
The outcomes for patients with locally advanced disease undergoing curative-intent therapy are sobering, as the 5-year OS with perioperative FLOT is still only 50.6%. Efforts are ongoing to identify strategies for enhancing the effectiveness of FLOT. One such strategy draws from the rectal cancer treatment paradigm, where patients receive total neoadjuvant therapy (TNT), combining both systemic chemotherapy and CRT prior to surgery. The CALGB 80803 trial adopted this strategy and used positron emission tomography (PET) scan responses to tailor therapy [
29]. In this phase 2 trial, patients with locally advanced EAC/GEJ AC were randomized to undergo induction chemotherapy with fluorouracil + oxaliplatin (FOLFOX) versus carboplatin/paclitaxel. Repeat PET after induction determined whether patients received CRT with the same chemotherapy (PET responders, defined as ≥ 35% decrease in the standardized uptake value [SUV]) or crossed over to the alternative chemotherapy for CRT (PET non-responders, defined as <35% decrease in SUV). However, the 5-year OS in the best responders was still only 53%. Additionally, a few other trials (including TOPGEAR) also showed no improvement in survival when combining chemotherapy and preoperative CRT [
19,
30]. Thus, there is a need for better strategies to meaningfully improve patient outcomes, and many studies are underway to explore the role of immunotherapy and targeted therapy in the curative setting.
6. Immunotherapy
Immunotherapy (IO) has transformed the treatment landscape for advanced/metastatic gastroesophageal cancers. However, to date, no study has demonstrated an OS benefit from adding IO to chemotherapy or CRT in patients with locally advanced adenocarcinomas. Below, we review the results from recent trials for preoperative, adjuvant, and perioperative immunotherapy, as well as data specific to the microsatellite instability-high/deficient mismatch repair (MSI-H/dMMR) population (
Table 3).
6.1. Preoperative Immunotherapy
The PERFECT trial was a phase 2 feasibility trial for patients with resectable EAC/GEJ AC who were treated with atezolizumab + preoperative CRT (CROSS regimen) [
31]. Forty patients were enrolled, and the pCR rate was 25% (10/40). When compared with historic controls from the Netherlands Cancer Registry after propensity score matching, there was no difference in pCR rate or median OS. Similarly, the ECOG-ACRIN EA2174 trial, which was a phase 2/3 study of perioperative nivolumab and ipilimumab in patients with locoregional EAC and GEJ AC, showed that the addition of nivolumab to preoperative CRT (CROSS regimen) did not improve the pCR rate (24.8% vs. 21.0%,
p = 0.27) [
32].
6.2. Adjuvant Immunotherapy
CheckMate 577, a phase 3 randomized placebo-controlled trial, assigned esophageal/GEJ cancer patients who underwent preoperative CRT followed by R0 resection and had residual pathological disease to receive nivolumab for 1 year (
n = 532) vs. placebo for 1 year (
n = 262). [
33]. Adjuvant nivolumab significantly improved the median DFS (22.4 months vs. 11 months,
p < 0.001). As a result, adjuvant nivolumab became the standard of care for patients undergoing trimodality therapy, though OS data are not yet available.
In contrast to CheckMate 577, the ATTRACTION-5 and VESTIGE trials did not show any benefit to adjuvant immunotherapy. ATTRACTION-5, a phase 3 randomized placebo-controlled trial, enrolled patients with locally advanced GAC/GEJ AC after D2 gastrectomy and randomized them to receive adjuvant nivolumab + chemotherapy (
n = 377) versus placebo + chemotherapy (
n = 378) [
34]. There was no difference in the 3-year relapse-free survival between the nivolumab group and the placebo group (68.4% vs. 65.3%,
p = 0.44). VESTIGE, a phase 2 randomized trial, enrolled patients with high risk EAC, GEJ AC, and GAC who underwent preoperative chemotherapy and surgical resection [
35]. Patients were randomized to either adjuvant chemotherapy or adjuvant nivolumab + ipilimumab. Interim analysis showed that nivolumab + ipilimumab did not improve median DFS over chemotherapy (11.4 months vs. 20.8 months,
p = 0.99) and the trial was stopped because of futility.
6.3. Perioperative Immunotherapy
As perioperative FLOT is currently considered the gold standard treatment for resectable locally advanced upper GI adenocarcinomas, there is significant interest in investigating the potential of combining perioperative chemotherapy with IO. KEYNOTE-585, a phase 3 randomized placebo-controlled trial, assessed perioperative chemotherapy + pembrolizumab versus perioperative chemotherapy + placebo in patients with resectable locally advanced GAC/GEJ AC [
36]. Because the standard chemotherapy regimen changed during the course of the trial, there were two cohorts (main cohort: cisplatin + capecitabine/fluorouracil,
n = 804; FLOT cohort,
n = 203). In the main cohort, pembrolizumab significantly increased the pCR rate when compared with placebo (12.9% vs. 2.0%,
p < 0.00001), but the differences in median event-free survival (44.4 months vs. 25.3 months,
p = 0.0198) and median OS (60.7 months vs. 58.0 months,
p = 0.174) did not reach statistical significance.
In parallel, the MATTERNHORN trial is a phase 3 randomized trial that evaluated perioperative FLOT + durvalumab versus FLOT + placebo in resectable GAC/GEJ AC patients [
37]. Durvalumab significantly increased the pCR rate (19% vs. 7%,
p < 0.00001), but OS data are still pending. Similarly, DANTE is a phase 2/3 randomized trial that assessed perioperative FLOT + atezolizumab versus FLOT + placebo [
38]. Results from the phase 2 portion showed higher histopathologic complete regression rates for the atezolizumab group than the placebo group (24% vs. 15%,
p = 0.032), but we still await the survival data. It is important to acknowledge that pCR rate is not always a surrogate for survival, and whether the pCR improvements seen in these trials will translate to survival benefit is to be seen.
6.4. Immunotherapy for MSI-H/dMMR Patients
Patients with MSI-H/dMMR disease represent a distinct subgroup in which IO can be used without the need for chemotherapy or CRT for the treatment of locally advanced disease. NEONIPIGA was a phase 2 trial that evaluated neoadjuvant nivolumab + ipilimumab and adjuvant nivolumab in localized MSI-H/dMMR GAC/GEJ AC (
n = 32), and INFINITY was a phase 2 trial that assessed neoadjuvant tremelimumab + durvalumab in resectable GAC/GEJ AC (
n = 15) [
39,
40]. While the sample sizes were small, both studies showed remarkable pCR rates of 58.6% (NEONIPIGA) and 60% (INFINITY). Larger confirmatory phase 3 trials are still pending, but these results have already led to an update in the National Comprehensive Cancer Network (NCCN) guidelines for neoadjuvant or perioperative immunotherapy. Because treatment decisions can be impacted, MSI/MMR status should be established in all newly diagnosed cases > cT1a to determine the best treatment approach.
7. Targeted Therapy
Biomarkers such as MSI, PD-L1, HER2, and claudin 18.2 are routinely assessed in patients with advanced/metastatic gastroesophageal cancers to guide therapeutic decisions. However, aside from MSI, these biomarkers currently do not significantly influence treatment decisions for locally advanced disease. HER2-targeted therapies have been well-established in the metastatic setting over the past decade, and more recently, therapies targeting claudin 18.2 have also demonstrated survival benefits. Because of this, targeted therapies are actively being investigated in the context of locally advanced disease, with HER2 being the most extensively studied. Despite numerous trials aimed at advancing the use of HER2-targeted therapies in the locally advanced setting, none have yielded positive results to date (
Table 4). Factors such as alterations in the tumor microenvironment, the development of compensatory pathways, and tumor heterogeneity may all contribute to resistance to HER2-targeted treatments. There is a clear need for the development of more effective strategies to target HER2 and other alterations for more personalized treatment options.
7.1. Preoperative Targeted Therapy
RTOG 1010, a randomized phase 3 trial, enrolled patients with HER2-positive locally advanced EAC to receive preoperative CRT with (
n = 102) or without trastuzumab (
n = 101) [
41]. Median DFS was 19.6 months in the CRT + trastuzumab group versus 14.2 months in the CRT group (
p = 0.97). The addition of trastuzumab did not improve outcomes for patients undergoing preoperative CRT.
7.2. Perioperative Targeted Therapy
HER-FLOT was an exploratory phase 2 trial of perioperative FLOT + trastuzumab in patients with HER2-positive locally advanced GAC/GEJ AC [
42]. Fifty-six patients were enrolled, and the pCR rate was 21.4%. PETRARCA, a randomized phase 2/3 trial for patients with HER2-positive resectable GAC/GEJ AC, assigned patients to receive either perioperative FLOT alone (
n = 41) or perioperative FLOT + trastuzumab + pertuzumab (
n = 40) [
43]. The addition of trastuzumab + pertuzumab significantly increased the pCR rate compared with the FLOT alone (35% vs. 12%,
p = 0.02). However, this trial was prematurely closed prior to transition to phase 3 because of the negative results and lack of survival benefit from the JACOB study (chemotherapy + trastuzumab + pertuzumab for metastatic HER2-positive GAC/GEJ AC). Additionally, the regimen of FLOT + trastuzumab + pertuzumab was also associated with considerable grade ≥3 diarrhea (41%), so it was difficult to tolerate. Finally, results from the INNOVATION trial were presented at the 2025 American Society of Clinical Oncology (ASCO) Gastrointestinal Cancers Symposium. INNOVATION, a randomized phase 2 trial, assigned patients with HER2-positive GAC/GEJ AC to receive perioperative chemotherapy alone (
n = 35) or perioperative chemotherapy + trastuzumab (
n = 67) or perioperative chemotherapy + trastuzumab + pertuzumab (
n = 70) [
44]. Cisplatin + capecitabine was initially used as the chemotherapy regimen, but it was later switched to FLOT after FLOT became the new standard of care regimen. The 5-year PFS was not significantly different between the groups (51.9% vs. 61.0% vs. 47.9%), and the 5-year OS also was not significantly different between the groups (60.5% vs. 67.5% vs. 62.6%). Because of these results, the standard-of-care treatment for HER2-positive locally advanced gastroesophageal adenocarcinomas remains FLOT.
8. Future Directions
Advancements in the treatment of locally advanced EAC, GEJ AC, and GAC have proven to be challenging. Despite multiple trials evaluating multimodality therapy and combinations of chemotherapy, immunotherapy, and targeted therapy, most of the trials have yielded negative results. Empiric trials have mostly failed to significantly improve patient outcomes. There is a scarcity of data on targeted therapies beyond those targeting HER2 (such as drugs targeting claudin 18.2) for locally advanced disease, and there is a clear need to find better predictive biomarkers and develop a more personalized approach for the management of locally advanced gastroesophageal adenocarcinomas.
In addition to establishing better targeted therapies, there is also increasing interest in the possibility of organ preservation and nonoperative management (“watchful waiting”) in patients with clinical complete response after preoperative therapy. This approach has been more commonly used in esophageal squamous cell carcinoma, which has a much higher pathologic complete response rate after preoperative therapy [
15]. This approach is also being evaluated in MSI-H/dMMR patients (NCT06059495). In order to assess both biomarker-driven targeted therapies and the possibility of organ preservation, the American Association for Cancer Research (AACR) is developing a unique platform neoadjuvant biomarker-driven trial in patients with gastroesophageal adenocarcinoma, and more details will be available in the coming months.
Cellular therapies such as chimeric antigen receptor T-cell (CAR-T) therapy have emerged as a promising treatment modality in various cancers. While CAR-T therapy has been successful in hematologic malignancies, its applications have been limited in solid tumors [
45]. For gastroesophageal cancers, CAR-T therapy remains experimental and is being studied in early-phase clinical trials. Common antigens targeted by CAR-T for gastroesophageal cancers include claudin 18.2 (NCT05539430, NCT04404595) and CEA (NCT06010862, NCT06821048). All ongoing trials are for advanced/metastatic disease, not for locally advanced disease with curative-intent treatment. Challenges for adapting CAR-T therapy for GI cancers include the heterogeneity of tumor target antigen expression, the complex immune microenvironment, and the risk of severe side effects such as cytokine release syndrome and neurotoxicity [
46,
47]. More data from ongoing trials are needed to fully understand the safety, efficacy, and long-term benefits of CAR-T therapy for curative treatment in these cancers.
Circulating tumor DNA (ctDNA) has shown potential as a prognostic biomarker and a tool for monitoring treatment response and detecting recurrence in patients with locally advanced gastroesophageal cancers. One exploratory study found that pretreatment ctDNA status more accurately stratified DFS than nodal status on PET [
48]. Additionally, clearance of ctDNA after neoadjuvant treatment was associated with a favorable response to therapy [
48]. Another observational study found that preoperative ctDNA detection was associated with high risk of recurrence [
49]. Recent research has also explored the use of ctDNA as a strategy for watchful waiting in locally advanced gastroesophageal cancer, with clearance of ctDNA during neoadjuvant chemoradiation potentially indicating a lower risk of relapse [
50]. However, while these findings are promising, they are very preliminary, and ctDNA is currently used only as a research tool. Larger prospective studies are needed to validate the application of ctDNA as a biomarker and to determine its potential role in guiding treatment decisions.
9. Conclusions
Over the last decade, significant advancements have been made in the treatment of locally advanced gastroesophageal adenocarcinoma. However, despite these developments, patient outcomes following curative-intent therapy remain suboptimal. Currently, perioperative chemotherapy is the standard of care for resectable locally advanced EAC, GEJ AC, and GAC regardless of biomarkers, with the exception of MSI-H/dMMR. Ongoing efforts to enhance treatment options include investigating the roles of immunotherapy and targeted therapies. As this is a rapidly evolving field, the standard of care is likely to shift in the near future. There is a critical need for more reliable predictive biomarkers and the development of novel targeted therapies to enable more personalized treatment strategies and, ultimately, improve patient outcomes.
Author Contributions
Investigation, J.J.L., J.E.R., R.E.W., Q.G., M.B.M. and J.A.A.; writing—original draft preparation, J.J.L., J.E.R., R.E.W., Q.G., M.B.M. and J.A.A.; writing—review and editing, J.J.L., J.E.R., R.E.W., Q.G., M.B.M. and J.A.A. All authors have read and agreed to the published version of the manuscript.
Funding
This research received no external funding.
Conflicts of Interest
The authors declare no conflicts of interest.
References
- Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global Cancer Statistics 2022: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA A Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef] [PubMed]
- Cancer of the Stomach—Cancer Stat Facts. Available online: https://seer.cancer.gov/statfacts/html/stomach.html (accessed on 22 January 2025).
- Cancer of the Esophagus—Cancer Stat Facts. Available online: https://seer.cancer.gov/statfacts/html/esoph.html (accessed on 22 January 2025).
- Glatz, T.; Verst, R.; Kuvendjiska, J.; Bronsert, P.; Becker, H.; Hoeppner, J.; Kulemann, B. Pattern of Recurrence and Patient Survival After Perioperative Chemotherapy with 5-FU, Leucovorin, Oxaliplatin and Docetaxel (FLOT) for Locally Advanced Esophagogastric Adenocarcinoma in Patients Treated Outside Clinical Trials. J. Clin. Med. 2020, 9, 2654. [Google Scholar] [CrossRef] [PubMed]
- Ahmed, O.; Ajani, J.A.; Lee, J.H. Endoscopic Management of Esophageal Cancer. World J. Gastrointest. Oncol. 2019, 11, 830–841. [Google Scholar] [CrossRef]
- Forbes, N.; Elhanafi, S.E.; Al-Haddad, M.A.; Thosani, N.C.; Draganov, P.V.; Othman, M.O.; Ceppa, E.P.; Kaul, V.; Feely, M.M.; Sahin, I.; et al. American Society for Gastrointestinal Endoscopy Guideline on Endoscopic Submucosal Dissection for the Management of Early Esophageal and Gastric Cancers: Summary and Recommendations. Gastrointest. Endosc. 2023, 98, 271–284. [Google Scholar] [CrossRef]
- Leers, J.M.; DeMeester, S.R.; Oezcelik, A.; Klipfel, N.; Ayazi, S.; Abate, E.; Zehetner, J.; Lipham, J.C.; Chan, L.; Hagen, J.A.; et al. The Prevalence of Lymph Node Metastases in Patients With T1 Esophageal Adenocarcinoma: A Retrospective Review of Esophagectomy Specimens. Ann. Surg. 2011, 253, 271–278. [Google Scholar] [CrossRef]
- Joseph, A.; Draganov, P.V.; Maluf-Filho, F.; Aihara, H.; Fukami, N.; Sharma, N.R.; Chak, A.; Yang, D.; Jawaid, S.; Dumot, J.; et al. Outcomes for Endoscopic Submucosal Dissection of Pathologically Staged T1b Esophageal Cancer: A Multicenter Study. Gastrointest. Endosc. 2022, 96, 445–453. [Google Scholar] [CrossRef]
- McCarty, J.C.; Parker, R.K.; Vidri, R.J.; Robinson, K.A.; Lipsitz, S.; Gangadharan, S.P.; Iyer, P.G. A Retrospective Cohort Study of Endoscopic Therapy and Esophagectomy for Stage 1 Esophageal Cancer: Less Is More. Gastrointest. Endosc. 2020, 92, 23–30. [Google Scholar] [CrossRef]
- Fan, X.; Wang, J.; Xia, L.; Qiu, H.; Tian, Y.; Zhangcai, Y.; Luo, X.; Gao, Y.; Li, C.; Wu, Y.; et al. Efficacy of Endoscopic Therapy for T1b Esophageal Cancer and Construction of Prognosis Prediction Model: A Retrospective Cohort Study. Int. J. Surg. 2023, 109, 1708–1719. [Google Scholar] [CrossRef]
- Ramji, S. Esophagectomy 1: Basics. Available online: https://www.aats.org/tsra-primer-esophagectomy-1-basics (accessed on 22 January 2025).
- Degiuli, M.; Reddavid, R.; Tomatis, M.; Ponti, A.; Morino, M.; Sasako, M.; Rebecchi, F.; Garino, M.; Vigano, L.; Scaglione, D.; et al. D2 Dissection Improves Disease-Specific Survival in Advanced Gastric Cancer Patients: 15-Year Follow-Up Results of the Italian Gastric Cancer Study Group D1 versus D2 Randomised Controlled Trial. Eur. J. Cancer 2021, 150, 10–22. [Google Scholar] [CrossRef]
- Songun, I.; Putter, H.; Kranenbarg, E.M.-K.; Sasako, M.; van de Velde, C.J. Surgical Treatment of Gastric Cancer: 15-Year Follow-Up Results of the Randomised Nationwide Dutch D1D2 Trial. Lancet Oncol. 2010, 11, 439–449. [Google Scholar] [CrossRef]
- Siewert, J.R.; Stein, H.J. Classification of Adenocarcinoma of the Oesophagogastric Junction. Br. J. Surg. 1998, 85, 1457–1459. [Google Scholar] [CrossRef] [PubMed]
- van Hagen, P.; Hulshof, M.C.C.M.; van Lanschot, J.J.B.; Steyerberg, E.W.; Henegouwen, M.I.; van Berge Wijnhoven, B.P.L.; Richel, D.J.; Nieuwenhuijzen, G.A.P.; Hospers, G.A.P.; Bonenkamp, J.J.; et al. Preoperative Chemoradiotherapy for Esophageal or Junctional Cancer. N. Engl. J. Med. 2012, 366, 2074–2084. [Google Scholar] [CrossRef] [PubMed]
- Cunningham, D.; Allum, W.H.; Stenning, S.P.; Thompson, J.N.; Van de Velde, C.J.; Nicolson, M.; Scarffe, J.H.; Lofts, F.J.; Falk, S.J.; Iveson, T.J.; et al. Perioperative Chemotherapy Versus Surgery Alone for Resectable Gastroesophageal Cancer. N. Engl. J. Med. 2006, 355, 11–20. [Google Scholar] [CrossRef] [PubMed]
- Ychou, M.; Boige, V.; Pignon, J.-P.; Conroy, T.; Bouché, O.; Lebreton, G.; Ducourtieux, M.; Bedenne, L.; Fabre, J.-M.; Saint-Aubert, B.; et al. Perioperative Chemotherapy Compared With Surgery Alone for Resectable Gastroesophageal Adenocarcinoma: An FNCLCC and FFCD Multicenter Phase III Trial. J. Clin. Oncol. 2011, 29, 1715–1721. [Google Scholar] [CrossRef]
- Hoeppner, J.; Brunner, T.; Schmoor, C.; Bronsert, P.; Kulemann, B.; Claus, R.; Utzolino, S.; Izbicki, J.R.; Gockel, I.; Gerdes, B.; et al. Perioperative Chemotherapy or Preoperative Chemoradiotherapy in Esophageal Cancer. N. Engl. J. Med. 2025, 392, 323–335. [Google Scholar] [CrossRef]
- Leong, T.; Smithers, B.M.; Michael, M.; Haustermans, K.; Wong, R.; Gebski, V.; O’connell, R.L.; Zalcberg, J.; Boussioutas, A.; Findlay, M.; et al. Preoperative Chemoradiotherapy for Resectable Gastric Cancer. N. Engl. J. Med. 2024, 391, 1810–1821. [Google Scholar] [CrossRef]
- Lee, J.; Lim, D.H.; Kim, S.; Park, S.H.; Park, J.O.; Park, Y.S.; Lim, H.Y.; Choi, M.G.; Sohn, T.S.; Noh, J.H.; et al. Phase III Trial Comparing Capecitabine Plus Cisplatin Versus Capecitabine Plus Cisplatin with Concurrent Capecitabine Radiotherapy in Completely Resected Gastric Cancer with D2 Lymph Node Dissection: The ARTIST Trial. J. Clin. Oncol. 2012, 30, 268–273. [Google Scholar] [CrossRef]
- Park, S.H.; Lim, D.H.; Sohn, T.S.; Lee, J.; Zang, D.Y.; Kim, S.T.; Kang, J.H.; Oh, S.Y.; Hwang, I.G.; Ji, J.H.; et al. A Randomized Phase III Trial Comparing Adjuvant Single-Agent S1, S-1 with Oxaliplatin, and Postoperative Chemoradiation with S-1 and Oxaliplatin in Patients with Node-Positive Gastric Cancer After D2 Resection: The ARTIST 2 Trial☆. Ann. Oncol. 2021, 32, 368–374. [Google Scholar] [CrossRef]
- Cats, A.; Jansen, E.P.M.; van Grieken, N.C.T.; Sikorska, K.; Lind, P.; Nordsmark, M.; Meershoek-Klein Kranenbarg, E.; Boot, H.; Trip, A.K.; Swellengrebel, H.A.M.; 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, 616–628. [Google Scholar] [CrossRef]
- Macdonald, J.S.; Smalley, S.R.; Benedetti, J.; Hundahl, S.A.; Estes, N.C.; Stemmermann, G.N.; Haller, D.G.; Ajani, J.A.; Gunderson, L.L.; Jessup, J.M.; et al. Chemoradiotherapy After Surgery Compared with Surgery Alone for Adenocarcinoma of the Stomach or Gastroesophageal Junction. N. Engl. J. Med. 2001, 345, 725–730. [Google Scholar] [CrossRef]
- Al-Batran, S.-E.; Homann, N.; Pauligk, C.; Goetze, T.O.; Meiler, J.; Kasper, S.; Kopp, H.-G.; Mayer, F.; Haag, G.M.; Luley, K.; 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, 1948–1957. [Google Scholar] [CrossRef] [PubMed]
- Sakuramoto, S.; Sasako, M.; Yamaguchi, T.; Kinoshita, T.; Fujii, M.; Nashimoto, A.; Furukawa, H.; Nakajima, T.; Ohashi, Y.; Imamura, H.; et al. Adjuvant Chemotherapy for Gastric Cancer with S-1, an Oral Fluoropyrimidine. N. Engl. J. Med. 2007, 357, 1810–1820. [Google Scholar] [CrossRef]
- Kakeji, Y.; Yoshida, K.; Kodera, Y.; Kochi, M.; Sano, T.; Ichikawa, W.; Lee, S.-W.; Shibahara, K.; Shikano, T.; Kataoka, M.; et al. Three-Year Outcomes of a Randomized Phase III Trial Comparing Adjuvant Chemotherapy with S-1 plus Docetaxel Versus S-1 Alone in Stage III Gastric Cancer: JACCRO GC-07. Gastric Cancer 2022, 25, 188–196. [Google Scholar] [CrossRef] [PubMed]
- Bang, Y.-J.; Kim, Y.-W.; Yang, H.-K.; Chung, H.C.; Park, Y.-K.; Lee, K.H.; Lee, K.-W.; Kim, Y.H.; Noh, S.-I.; Cho, J.Y.; et al. Adjuvant Capecitabine and Oxaliplatin for Gastric Cancer After D2 Gastrectomy (CLASSIC): A Phase 3 Open-Label, Randomised Controlled Trial. Lancet 2012, 379, 315–321. [Google Scholar] [CrossRef]
- Reynolds, J.V.; Preston, S.R.; O’Neill, B.; Lowery, M.A.; Baeksgaard, L.; Crosby, T.; Cunningham, M.; Cuffe, S.; Griffiths, G.O.; Parker, I.; 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, 1015–1027. [Google Scholar] [CrossRef]
- Goodman, K.A.; Ou, F.-S.; Hall, N.C.; Bekaii-Saab, T.; Fruth, B.; Twohy, E.; Meyers, M.O.; Boffa, D.J.; Mitchell, K.; Frankel, W.L.; 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, 2803–2815. [Google Scholar] [CrossRef]
- Ajani, J.A.; Xiao, L.; Roth, J.A.; Hofstetter, W.L.; Walsh, G.; Komaki, R.; Liao, Z.; Rice, D.C.; Vaporciyan, A.A.; Maru, D.M.; et al. A Phase II Randomized Trial of Induction Chemotherapy Versus No Induction Chemotherapy Followed by Preoperative Chemoradiation in Patients with Esophageal Cancer. Ann. Oncol. 2013, 24, 2844–2849. [Google Scholar] [CrossRef]
- van den Ende, T.; de Clercq, N.C.; van Berge Henegouwen, M.I.; Gisbertz, S.S.; Geijsen, E.D.; Verhoeven, R.H.A.; Meijer, S.L.; Schokker, S.; Dings, M.P.G.; Bergman, J.J.G.H.M.; et al. Neoadjuvant Chemoradiotherapy Combined with Atezolizumab for Resectable Esophageal Adenocarcinoma: A Single-Arm Phase II Feasibility Trial (PERFECT). Clin. Cancer Res. 2021, 27, 3351–3359. [Google Scholar] [CrossRef]
- Eads, J.R.; Graham, N.; Gibson, M.K.; Rajdev, L.; Chakravarthy, A.B.; Khullar, O.V.; Lin, S.H.; Wistuba, I.I.; Agarwal, R.; Blazar, M.; 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, 4000. [Google Scholar] [CrossRef]
- Kelly, R.J.; Ajani, J.A.; Kuzdzal, J.; Zander, T.; Van Cutsem, E.; Piessen, G.; Mendez, G.; Feliciano, J.; Motoyama, S.; Lièvre, A.; et al. Adjuvant Nivolumab in Resected Esophageal or Gastroesophageal Junction Cancer. N. Engl. J. Med. 2021, 384, 1191–1203. [Google Scholar] [CrossRef]
- Kang, Y.-K.; Terashima, M.; Kim, Y.-W.; Boku, N.; Chung, H.C.; Chen, J.-S.; Ji, J.; Yeh, T.-S.; Chen, L.-T.; Ryu, M.-H.; et al. Adjuvant Nivolumab Plus Chemotherapy Versus Placebo Plus Chemotherapy for Stage III Gastric or Gastro-Oesophageal Junction Cancer After Gastrectomy with D2 or More Extensive Lymph-Node Dissection (ATTRACTION-5): A Randomised, Multicentre, Double-Blind, Placebo-Controlled, Phase 3 Trial. Lancet Gastroenterol. Hepatol. 2024, 9, 705–717. [Google Scholar] [CrossRef] [PubMed]
- Lordick, F.; Mauer, M.E.; Stocker, G.; Cella, C.A.; Ben-Aharon, I.; Piessen, G.; Wyrwicz, L.; Al-Haidari, G.; Fleitas-Kanonnikoff, T.; Boige, V.; et al. Adjuvant Immunotherapy in Patients with Resected Gastric and Oesophagogastric Junction Cancer Following Preoperative Chemotherapy with High Risk for Recurrence (ypN+ and/or R1): European Organisation of Research and Treatment of Cancer (EORTC) 1707 VESTIGE Study. Ann. Oncol. 2025, 36, 197–207. [Google Scholar] [CrossRef] [PubMed]
- Shitara, K.; Rha, S.Y.; Wyrwicz, L.S.; Oshima, T.; Karaseva, N.; Osipov, M.; Yasui, H.; Yabusaki, H.; Afanasyev, S.; Park, Y.-K.; 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, 212–224. [Google Scholar] [CrossRef] [PubMed]
- Janjigian, Y.Y.; Al-Batran, S.-E.; Wainberg, Z.A.; Van Cutsem, E.; Molena, D.; Muro, K.; Hyung, W.J.; Wyrwicz, L.S.; Oh, D.-Y.; Omori, T.; et al. Pathological Complete Response (pCR) to 5-Fluorouracil, Leucovorin, Oxaliplatin and Docetaxel (FLOT) with or Without Durvalumab (D) in Resectable Gastric and Gastroesophageal Junction Cancer (GC/GEJC): Subgroup Analysis by Region from the Phase 3, Randomized, Double-Blind MATTERHORN Study. J. Clin. Oncol. 2024, 42, LBA246. [Google Scholar] [CrossRef]
- Lorenzen, S.; Götze, T.O.; Thuss-Patience, P.; Biebl, M.; Homann, N.; Schenk, M.; Lindig, U.; Heuer, V.; Kretzschmar, A.; Goekkurt, E.; 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, 410–420. [Google Scholar] [CrossRef]
- André, T.; Tougeron, D.; Piessen, G.; de la Fouchardière, C.; Louvet, C.; Adenis, A.; Jary, M.; Tournigand, C.; Aparicio, T.; Desrame, J.; 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, 255–265. [Google Scholar] [CrossRef]
- Pietrantonio, F.; Raimondi, A.; Lonardi, S.; Murgioni, S.; Cardellino, G.G.; Tamberi, S.; Strippoli, A.; Palermo, F.; Prisciandaro, M.; Randon, G.; 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, 358. [Google Scholar] [CrossRef]
- Safran, H.P.; Winter, K.; Ilson, D.H.; Wigle, D.; DiPetrillo, T.; Haddock, M.G.; Hong, T.S.; Leichman, L.P.; Rajdev, L.; Resnick, M.; 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, 259–269. [Google Scholar] [CrossRef]
- Hofheinz, R.-D.; Hegewisch-Becker, S.; Kunzmann, V.; Thuss-Patience, P.; Fuchs, M.; Homann, N.; Graeven, U.; Schulte, N.; Merx, K.; Pohl, M.; et al. Trastuzumab in Combination with 5-Fluorouracil, Leucovorin, Oxaliplatin and Docetaxel as Perioperative Treatment for Patients with Human Epidermal Growth Factor Receptor 2-Positive Locally Advanced Esophagogastric Adenocarcinoma: A Phase II Trial of the Arbeitsgemeinschaft Internistische Onkologie Gastric Cancer Study Group. Int. J. Cancer 2021, 149, 1322–1331. [Google Scholar] [CrossRef]
- Hofheinz, R.-D.; Merx, K.; Haag, G.M.; Springfeld, C.; Ettrich, T.; Borchert, K.; Kretzschmar, A.; Teschendorf, C.; Siegler, G.; Ebert, M.P.; et al. FLOT Versus FLOT/Trastuzumab/Pertuzumab Perioperative Therapy of Human Epidermal Growth Factor Receptor 2–Positive Resectable Esophagogastric Adenocarcinoma: A Randomized Phase II Trial of the AIO EGA Study Group. J. Clin. Oncol. 2022, 40, 3750–3761. [Google Scholar] [CrossRef]
- Wagner, A.D.; Grabsch, H.I.; Mauer, M.E.; Lorenzen, S.; Bouche, O.; Thuss-Patience, P.C.; Elme, A.; Moehler, M.H.; Fumagalli Romario, U.; Kang, Y.-K.; et al. EORTC-1203 GITC “INNOVATION”: Integration of Trastuzumab (T), with or Without Pertuzumab (P), into Perioperative Chemotherapy of HER-2 Positive Stomach Cancer: Overall Survival Results. J. Clin. Oncol. 2025, 43, LBA331. [Google Scholar] [CrossRef]
- Hupperetz, C.; Lah, S.; Kim, H.; Kim, C.H. CAR T Cell Immunotherapy Beyond Haematological Malignancy. Immune Netw. 2022, 22, e6. [Google Scholar] [CrossRef] [PubMed]
- Xu, N.; Wu, Z.; Pan, J.; Xu, X.; Wei, Q. CAR-T Cell Therapy: Advances in Digestive System Malignant Tumors. Mol. Ther. Oncol. 2024, 32, 200872. [Google Scholar] [CrossRef] [PubMed]
- Chen, T.; Wang, M.; Chen, Y.; Liu, Y. Current Challenges and Therapeutic Advances of CAR-T Cell Therapy for Solid Tumors. Cancer Cell Int. 2024, 24, 133. [Google Scholar] [CrossRef]
- Cabalag, C.S.; Yates, M.; Corrales, M.B.; Yeh, P.; Wong, S.Q.; Zhang, B.Z.; Fujihara, K.M.; Chong, L.; Hii, M.W.; Dawson, S.-J.; et al. Potential Clinical Utility of a Targeted Circulating Tumor DNA Assay in Esophageal Adenocarcinoma. Ann. Surg. 2022, 276, e120–e126. [Google Scholar] [CrossRef]
- Hofste, L.S.M.; Geerlings, M.J.; von Rhein, D.; Tolmeijer, S.H.; Weiss, M.M.; Gilissen, C.; Hofste, T.; Garms, L.M.; Janssen, M.J.R.; Rütten, H.; et al. Circulating Tumor DNA-Based Disease Monitoring of Patients with Locally Advanced Esophageal Cancer. Cancers 2022, 14, 4417. [Google Scholar] [CrossRef]
- To, H.Q.Q.; Menon, H.; Green, G.B. Analysis of ctDNA Assay as a Strategy for Watchful Waiting in Locally Advanced Esophageal Cancer. J. Clin. Oncol. 2024, 42, e16126. [Google Scholar] [CrossRef]
Table 1.
Summary of major trials of chemoradiation in locally advanced gastroesophageal adenocarcinoma.
Table 1.
Summary of major trials of chemoradiation in locally advanced gastroesophageal adenocarcinoma.
Trial Name | Phase | Disease Site | Setting | Intervention | Outcomes | Reference |
---|
CROSS | III | Esophagus, GEJ | Preoperative | Preoperative CRT + surgery vs. surgery alone | R0 resection 92% vs. 69%, p < 0.001 mOS 49.4 m vs. 24.0 m, HR 0.657, p = 0.003 | [11] |
TOPGEAR | III | GEJ, Gastric | Preoperative | Preoperative CRT + perioperative chemotherapy (ECF, FLOT) vs. perioperative chemotherapy alone | pCR 17% vs. 8% mOS 46 m vs. 49 m, HR 1.05 | [14] |
ARTIST | III | Gastric | Adjuvant | Adjuvant chemotherapy (XP) + CRT vs. adjuvant chemotherapy | 3-year DFS 78.2% vs. 74.2%, p = 0.0862 | [15] |
ARTIST 2 | III | Gastric | Adjuvant | Adjuvant S-1 vs. SOX vs. CRT (SOX + RT) | 3-year DFS 64.8% vs. 74.3% vs. 72.8%, p = 0.879 (SOX vs. CRT) | [16] |
CRITICS | III | GEJ, Gastric | Adjuvant | Perioperative chemotherapy vs. preoperative chemotherapy + postoperative CRT | mOS 43 m vs. 37 m, HR 1.01, p = 0.90 | [17] |
MacDonald | III | GEJ, Gastric | Adjuvant | Surgery alone vs. surgery + adjuvant CRT | mOS 27 vs. 36 m, HR 1.35, p = 0.005 | [18] |
Table 2.
Summary of major trials of chemotherapy in locally advanced gastroesophageal adenocarcinoma.
Table 2.
Summary of major trials of chemotherapy in locally advanced gastroesophageal adenocarcinoma.
Trial Name | Phase | Disease Site | Setting | Intervention | Outcomes | Reference |
---|
MAGIC | III | Esophagus, GEJ, Gastric | Perioperative | Perioperative ECF vs. surgery alone | 5-year OS 36% vs. 23%, HR 0.75, p = 0.009 | [12] |
FLOT4-AIO | II/III | GEJ, Gastric | Perioperative | Perioperative FLOT vs. perioperative ECF/ECX | mOS 50 m vs. 35 m, HR 0.77, p = 0.012 mDFS 30 m vs. 18 m, HR 0.75, p = 0.0036 | [19] |
ACTS-GC | III | Gastric | Adjuvant | Surgery + adjuvant S-1 vs. surgery alone | 3-year OS 80.1% vs. 70.1%, HR 0.68, p = 0.003 | [20] |
JACCRO-GC07 | III | Gastric | Adjuvant | Adjuvant S-1 + docetaxel vs. adjuvant S-1 alone | 3-year RFS 67.7% vs. 57.4%, HR 0.715, p = 0.0008 3-year OS 77.7% vs. 71.2%, HR 0.742, p = 0.0076 | [21] |
CLASSIC | III | Gastric | Adjuvant | Surgery + adjuvant CAPOX vs. surgery alone | 3-year DFS 74% vs. 59%, HR 0.56, p < 0.0001 | [22] |
Table 3.
Summary of major trials of immunotherapy in locally advanced gastroesophageal adenocarcinoma.
Table 3.
Summary of major trials of immunotherapy in locally advanced gastroesophageal adenocarcinoma.
Trial Name | Phase | Disease Site | Setting | Intervention | Outcomes | Reference |
---|
PERFECT | II | Esophagus | Neoadjuvant | Preoperative CRT + atezolizumab | pCR 25% | [27] |
ECOG-ACRIN EA2174 | II/III | Esophagus, GEJ | Neoadjuvant | Preoperative CRT + nivolumab vs. preoperative CRT alone | pCR 24.8% vs. 21.0%, p = 0.27 | [28] |
CHECKMATE-577 | III | Esophagus, GEJ | Adjuvant | Adjuvant nivolumab vs. placebo | mDFS 22.4 m vs. 11.0 m, HR 0.69, p < 0.001 | [29] |
ATTRACTION-05 | III | GEJ, Gastric | Adjuvant | Adjuvant chemotherapy (S-1, CAPOX) + nivolumab vs. adjuvant chemotherapy alone | 3-year RFS 68.4% vs. 65.3%, HR 0.90, p = 0.44 | [30] |
VESTIGE | II | Esophagus, GEJ, Gastric | Adjuvant | Adjuvant nivolumab + ipilimumab vs. adjuvant chemotherapy | mDFS 11.4 m vs. 20.8 m, HR 1.55, p = 0.99 | [31] |
KEYNOTE-585 | III | GEJ, Gastric | Perioperative | Perioperative chemotherapy (cisplatin + capecitabine/fluorouracil, FLOT) + pembrolizumab vs. perioperative chemotherapy alone | Main cohort: pCR 12.9% vs. 2.0%, p < 0.00001 mEFS 44.4 m vs. 25.3 m, HR 0.81, p = 0.0198 mOS 60.7 m vs. 58.0 m, HR 0.90, p = 0.174 | [32] |
MATTERHORN | III | GEJ, Gastric | Perioperative | FLOT + durvalumab vs. FLOT + placebo | pCR 19% vs. 7%, p < 0.00001 | [33] |
DANTE | II/III | GEJ, Gastric | Perioperative | FLOT + atezolizumab vs. FLOT + placebo | CRR 24% vs. 15%, p = 0.032 | [34] |
NEONIPIGA | II | GEJ, Gastric | Perioperative | Preoperative ipilimumab + nivolumab and postoperative nivolumab | pCR 58.6% | [35] |
INFINITY | II | GEJ, Gastric | Neoadjuvant | Neoadjuvant tremelimumab + durvalumab | pCR 60% | [36] |
Table 4.
Summary of major trials of HER2-targeted therapy in locally advanced gastroesophageal adenocarcinoma.
Table 4.
Summary of major trials of HER2-targeted therapy in locally advanced gastroesophageal adenocarcinoma.
Trial Name | Phase | Disease Site | Setting | Intervention | Outcomes | Reference |
---|
RTOG1010 | III | Esophagus | Neoadjuvant | Preoperative CRT + trastuzumab vs. CRT alone | mDFS 19.6 m vs. 14.2 m, HR 0.99, p = 0.97 | [37] |
HER-FLOT | II | GEJ, Gastric | Perioperative | FLOT + trastuzumab | pCR 21.4% | [38] |
PETRARCA | II | GEJ, Gastric | Perioperative | FLOT + trastuzumab + pertuzumab vs. FLOT alone | pCR 35% vs. 12%, p = 0.019 | [39] |
INNOVATION | II | GEJ, Gastric | Perioperative | Perioperative chemotherapy (cisplatin + capecitabine, FLOT) vs. chemotherapy + trastuzumab vs. chemotherapy + trastuzumab + pertuzumab | 5-year PFS 51.9% vs. 61.0% vs. 47.9% 5-year OS 60.5% vs. 67.5% vs. 62.6% | [40] |
| Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).