- 1Department of Gastroenterology, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
- 2Department of Thoracic Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
- 3Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
Neoadjuvant treatments play a crucial role in improving survival rates for patients with locally advanced resectable esophageal cancer. The CROSS and NEOCRTEC5010 trials have shown that neoadjuvant concurrent chemoradiotherapy significantly enhances survival compared to surgery alone. In contrast, the NeoRes and CMISG1701 trials indicate that while neoadjuvant chemoradiotherapy yields a higher histological complete response rate, it does not confer survival benefits over neoadjuvant chemotherapy. The recent JCOG1109 trial has demonstrated that neoadjuvant triplet chemotherapy offers a statistically significant overall survival advantage compared to doublet chemotherapy. However, combining doublet chemotherapy with radiotherapy did not show notable survival improvement compared to doublet chemotherapy alone. Additionally, neoadjuvant immunotherapy in conjunction with chemotherapy has shown a greater histological complete response rate compared to neoadjuvant chemotherapy, with comparable rates to neoadjuvant chemoradiotherapy. These findings have sparked debate regarding the necessity of radiotherapy in neoadjuvant treatment protocols. This review aims to elucidate the role of radiotherapy based on the current evidence and to assess ongoing and future trials that may address existing knowledge gaps. It will also underscore the challenges in making definitive recommendations about radiotherapy, particularly as technologies and treatment modalities continue to advance.
Introduction
Esophageal cancer is the sixth most commonly diagnosed cancer and the fifth leading cause of cancer-related deaths globally, representing a significant public health burden (1). Esophageal squamous cell carcinoma predominates in East Asia, while adenocarcinoma is more common in Western countries. Unfortunately, nonspecific symptoms often lead to diagnosis at an advanced stage (2). Consequently, many patients with locally advanced disease are initially deemed unresectable. However, a subset with localized disease can achieve cure through neoadjuvant therapy followed by surgery.
Neoadjuvant chemotherapy as an alternative to chemoradiotherapy
Neoadjuvant chemoradiotherapy followed by surgery is the established standard for locally advanced resectable esophageal cancer, regardless of histological subtype (3). The CROSS trial demonstrated that neoadjuvant chemoradiotherapy significantly improves overall survival compared to surgery alone in patients with locally advanced adenocarcinoma or squamous cell carcinoma (hazard ratio [HR] = 0.70, 95% confidence interval [CI]: 0.55-0.89; P = 0.004) (4, 5). Similarly, the NEOCRTEC5010 trial confirmed the survival benefit of neoadjuvant chemoradiotherapy over surgery alone in locally advanced esophageal squamous cell carcinoma (HR = 0.74, 95% CI: 0.57-0.97; P = 0.03) (6, 7).
Despite this evidence, clinical adoption of neoadjuvant chemoradiotherapy remains limited, with fewer than 5% of patients in China receiving this treatment (8). Several factors contribute to this low uptake. Firstly, while over 50% of esophageal cancer patients in high-income countries receive radiotherapy for local control, access is significantly constrained in low- and middle-income countries (9). Radiotherapy infrastructure is often underdeveloped in these regions, limiting global accessibility for esophageal cancer patients (10).
Secondly, multidisciplinary team discussions are crucial for managing esophageal cancer, particularly in formulating radiotherapy-based strategies. However, only a small proportion of patients are reviewed in multidisciplinary team meetings, which are vital for optimizing staging, ensuring timely surgery, and improving adherence to clinical guidelines (11–13).
Thirdly, some surgeons express concern that neoadjuvant chemoradiotherapy may increase toxicity, potentially leading to greater operative difficulty, higher postoperative morbidity, and mortality (14–18). Additionally, disease progression during neoadjuvant chemoradiotherapy or patient preference may prevent some patients from proceeding to surgery (19).
Given these challenges, neoadjuvant chemotherapy is increasingly recognized as a viable alternative to chemoradiotherapy, offering comparable survival outcomes. The NeoRes trial found no significant difference in 3-year overall survival between the two approaches for locally advanced esophageal or junctional cancer (HR = 1.11, 95% CI: 0.74-1.67; P = 0.77) (20). Similarly, the CMISG1701 trial revealed no significant survival difference between the two therapies in esophageal squamous cell carcinoma patients (HR = 0.82, 95% CI: 0.58-1.18; P = 0.28) (21). Based on this evidence, neoadjuvant radiotherapy may not be essential (22).
Despite the use of neoadjuvant chemotherapy followed by esophagectomy, the prognosis for locally advanced resectable esophageal cancer remains poor, with 5-year survival rates around 50-60%. Furthermore, many patients undergoing radical esophagectomy experience postoperative complications, hindering the completion of adjuvant chemotherapy (23). Consequently, more intensive neoadjuvant strategies are needed to improve long-term outcomes.
Summary: Neoadjuvant chemotherapy may be an alternative to standard chemoradiotherapy, particularly where radiotherapy access or multidisciplinary coordination is limited, as trials show comparable survival. However, despite this alternative approach, overall survival remains poor and postoperative complications hinder further treatment, necessitating more intensive neoadjuvant regimens.
Intensive neoadjuvant chemotherapy enhances survival outcomes
The JCOG1109 trial evaluated the efficacy and safety of triplet chemotherapy (fluorouracil, cisplatin, and docetaxel) compared to doublet chemotherapy (fluorouracil and cisplatin) and chemoradiotherapy (41.4 Gy combined with fluorouracil and cisplatin) in patients with locally advanced esophageal squamous cell carcinoma (24). Results showed that triplet chemotherapy significantly improved the 3-year overall survival rate compared to doublet chemotherapy (HR = 0.68, 95% CI: 0.50-0.92; P = 0.006). However, no significant difference was observed between chemoradiotherapy and doublet chemotherapy (HR = 0.84, 95% CI: 0.63-1.12; P = 0.12). Although triplet chemotherapy showed a numerically better survival trend compared to chemoradiotherapy, this difference was not statistically significant (HR = 0.80, 95% CI: 0.59-1.10; P > 0.05).
Nevertheless, intensive neoadjuvant chemotherapy is associated with increased adverse events. Grade ≥3 febrile neutropenia occurred in 1%, 16%, and 5% of patients in the doublet chemotherapy, triplet chemotherapy, and chemoradiotherapy groups, respectively. Grade ≥2 postoperative pneumonia, anastomotic leak, and recurrent laryngeal nerve paralysis were reported in the doublet group (10%, 10%, 15%), triplet group (10%, 9%, 10%), and chemoradiotherapy group (13%, 13%, 10%).
Similarly, the ESOPEC trial, a two-arm randomized phase III study, compared perioperative chemotherapy (5-FU, leucovorin, oxaliplatin, and docetaxel) with neoadjuvant chemoradiation (41.4 Gy plus carboplatin and paclitaxel) (25, 26). After a median follow-up of 55 months, the 3-year overall survival rate was significantly higher in the chemotherapy group (57.4% vs. 50.7%). This survival advantage persisted at 5 years (50.6% vs. 38.7%). The chemotherapy regimen reduced the risk of death by 30% (HR = 0.70, 95% CI: 0.53-0.92; P = 0.012).
Collectively, these trials suggest that intensive neoadjuvant chemotherapy could become the new standard of care for locally advanced esophageal cancer, potentially obviating the need for radiotherapy in neoadjuvant strategies.
Summary: The JCOG1109 and ESOPEC trials indicates that intensive neoadjuvant chemotherapy regimens significantly enhance survival outcomes compared to standard doublet chemotherapy or chemoradiotherapy, despite increased toxicity risks, suggesting its potential as a new standard that may obviate the need for radiotherapy.
Reasons for survival improvement with intensive neoadjuvant chemotherapy
Intensive neoadjuvant chemotherapy facilitates tumor downstaging, improves R0 resection rates, and helps eliminate potential micrometastases and occult distant lesions. The CROSS trial reported comparable distant metastasis rates between surgery alone and neoadjuvant chemoradiotherapy (27% vs. 28%) over 10 years of follow-up (5). In contrast, the JCOG1109 trial demonstrated a lower incidence of distant-only recurrences with triplet chemotherapy versus chemoradiotherapy (31.6% vs. 49.3%) (24).
Notably, locoregional-only recurrences occurred more frequently in the triplet chemotherapy group (43.4% vs. 22.7%). Patients with locoregional recurrence in this cohort were more likely to receive subsequent chemoradiotherapy (50.0% vs. 17.1%) or radiotherapy alone (28.4% vs. 21.4%), indicating better access to curative-intent salvage therapies. Consequently, locoregional-only recurrence patients in the triplet group experienced longer survival (18.9 vs. 9.9 months).
These findings suggest that while locoregional recurrence may be manageable with curative radiotherapy, long-term survival depends more critically on controlling distant metastases, a goal potentially better achieved through intensive systemic therapy. In JCOG1109, patients with distant metastases received salvage chemotherapy. However, the absence of immune checkpoint inhibitors, which enhance efficacy in metastatic esophageal cancer when combined with chemotherapy (27–32), likely limited survival benefits in the chemoradiotherapy group (33, 34).
Additionally, neoadjuvant chemoradiotherapy may increase postoperative and non-cancer-related mortality, contributing to its modest survival improvements. The JCOG1109 trial reported significantly higher non-cancer-related deaths with chemoradiotherapy versus triplet chemotherapy (25.8% vs. 9.5%), consistent with prior studies (20, 35).
Elevated non-cancer mortality, particularly from lung and cardiac causes, may stem from radiation fields. The JCOG1109 trial employed elective lymph node irradiation covering most mediastinal lymph nodes for middle and lower thoracic tumors, increasing radiation exposure to adjacent organs and associated risks (36, 37). Conversely, restricted fields for upper thoracic disease likely reduced cardiac exposure, contributing to a favorable HR of 0.68 for this subgroup.
Current practice favors involved field irradiation for esophageal cancer, as it minimizes radiation to organs at risk while maintaining survival outcomes comparable to elective nodal irradiation (38–40). When involved-field irradiation was used, no significant differences emerged in mortality due to neoadjuvant therapy side effects (7.0% vs. 3.1%, P = 0.684), postoperative complications (12.3% vs. 6.2%, P = 0.355), other diseases (5.3% vs. 1.6%, P = 0.622), or unknown causes (3.5% vs. 6.2%, P = 0.684) between chemoradiotherapy and chemotherapy cohorts (41).
Critically, JCOG1109 showed no significant overall survival improvement for triplet chemotherapy over chemoradiotherapy (HR = 0.80, 95% CI: 0.59-1.10). Limitations in the chemoradiotherapy group, including elective nodal irradiation protocols, surgical techniques, and salvage therapies, further complicate its clinical application. Among these challenges, integrating immunotherapy emerges as particularly significant.
Summary: Intensive neoadjuvant chemotherapy enhances survival through superior systemic control of distant metastases and lower non-cancer mortality compared to chemoradiotherapy, despite higher locoregional recurrence rates which are treatable with salvage radiotherapy. Limitations of radiation fields and lacking of immunotherapy in salvage settings further impacted chemoradiotherapy outcomes.
Neoadjuvant immunotherapy improves pathological complete response rates
The ESCORT-NEO/NCCES01 trial compared neoadjuvant camrelizumab plus chemotherapy versus neoadjuvant chemotherapy alone (42). Results revealed that camrelizumab with albumin-bound paclitaxel and cisplatin significantly improved pathological complete response (pCR) rates (28.0% vs. 4.7%, P < 0.0001). Although this pCR exceeded the 19.8% reported for triplet chemotherapy (fluorouracil, cisplatin, and docetaxel), direct comparison was not performed. Grade ≥3 treatment-related adverse events during neoadjuvant treatment occurred in 34.1% versus 28.8% between immunochemotherapy and chemotherapy groups, with postoperative complication rates of 34.2% and 32.0%.
The REVO trial further compared pCR rates between immunochemotherapy (camrelizumab, albumin-bound paclitaxel, and cisplatin) and chemoradiotherapy (36–40 Gy plus albumin-bound paclitaxel and cisplatin). The immunochemotherapy cohort achieved higher pCR (40.6% vs. 35.7%). Grade ≥3 treatment-related adverse events were 22% versus 31.8% between immunochemotherapy and chemoradiotherapy groups before surgery, and 28.1% vs. 21.4% after surgery. Notably, the 40.6% pCR rate of the REVO trial exceeded the 28.0% of the ESCORT-NEO/NCCES01 trial (42). This enhanced pCR correlated with superior 2-year overall survival (81.3% vs 71.3%; HR = 1.57, 95% CI: 1.26-1.96; P < 0.001) and disease-free survival (73.9% vs 63.4%; HR = 1.37, 95% CI: 1.11-1.69; P < 0.001) (43).
However, pCR rates for immunochemotherapy vary considerably (16.7%-57.1%) (44, 45), with 26.9% (95% CI, 16.7%-38.3%) treatment-related severe adverse events. This heterogeneity prompts questions about the impact of immunotherapy when combined with chemoradiotherapy. The Palace-1 trial evaluated preoperative pembrolizumab plus chemoradiotherapy (41.4 Gy plus carboplatin and paclitaxel) for resectable esophageal squamous cell carcinoma (46). This regimen proved safe, did not delay surgery, and achieved 55.6% pCR. Similarly, NEOCRTEC1901 reported higher pCR with toripalimab plus chemoradiotherapy (44 Gy plus paclitaxel and cisplatin) versus chemoradiotherapy alone (50% vs. 36%, P = 0.19) (47). Collectively, neoadjuvant immunotherapy plus chemoradiotherapy consistently achieve pCR >50%, which is higher than immunochemotherapy (48, 49). Safety profiles remain comparable, with no significant differences in grade 3/4 adverse events or postoperative complications versus neoadjuvant chemoradiotherapy.
Summary: Neoadjuvant immunotherapy significantly enhances pCR rates, whether added to chemotherapy or chemoradiotherapy, while maintaining comparable safety profiles to standard neoadjuvant approaches.
Pathological complete response rate is not a prognostic factor
Despite the high pCR rates observed with immunotherapy combined with chemoradiotherapy, these rates did not correlate with improved survival outcomes. Although the immunochemotherapy demonstrated a significantly lower pCR rate compared to immunotherapy plus chemoradiotherapy (32.3% vs. 52.1%, P = 0.004), the 2-year overall survival rates were similar (84.42% vs. 81.70%, P = 0.860), as were the 2-year disease-free survival rates (83.21% vs. 80.47%, P = 0.839) (50).
The JCOG1109 trial reinforced these findings, reporting a substantially higher pCR rate in the neoadjuvant chemoradiotherapy group (38.5%) compared to the triplet chemotherapy (19.8%) and doublet chemotherapy (2.0%) groups (24). Nevertheless, this elevated pCR rate did not translate into improved overall survival relative to either the triplet or doublet chemotherapy cohorts. In contrast, the triplet chemotherapy group did exhibit an overall survival benefit over the doublet group.
Similarly, the NeoRes trial revealed minimal differences in long-term survival between the neoadjuvant chemoradiotherapy and chemotherapy groups, despite a significant disparity in pCR rates (28% vs. 9%) (20). The CMISG1701 study also documented a higher pCR rate in the chemoradiotherapy group (27.7% vs. 2.9%), yet no significant difference in survival outcomes was observed (HR = 0.82, P = 0.28) (21).
Collectively, these studies suggest that pCR may not be a reliable prognostic indicator for long-term outcomes when comparing different preoperative treatment strategies (51). In the absence of comprehensive long-term survival data, the pCR rates associated with various neoadjuvant therapies fail to clarify whether chemoradiotherapy combined with immunotherapy is superior to triplet chemotherapy or immunochemotherapy. This leads to a pivotal question: What is the clinical significance of achieving pCR rates exceeding 50% through the combination of chemoradiotherapy and immunotherapy?
Summary: Superior pCR rates achieved with various neoadjuvant chemoradiotherapy or immunotherapy combinations do not correlate with improved overall survival compared to regimens yielding lower pCR rates, challenging the value of pCR as a prognostic indicator for long-term outcomes.
Organ preservation after neoadjuvant immunotherapy plus chemoradiotherapy
The clinical significance of achieving high pCR rates following neoadjuvant immunotherapy combined with chemoradiotherapy lies in organ preservation. The SANO trial, a phase III multicenter stepped-wedge cluster randomized controlled trial, compared active surveillance with standard surgical intervention for locally advanced esophageal cancer patients who achieved a clinical complete response (cCR) after neoadjuvant chemoradiotherapy (41.1 Gy plus carboplatin and paclitaxel) (52). Patients with cCR were randomized to either active surveillance (with salvage surgery upon local recurrence) or immediate standard surgical treatment. Of the 309 patients evaluated, 198 were allocated to active surveillance and 111 underwent standard surgery. With a median follow-up of 38 months, overall survival in the active surveillance cohort was non-inferior to that in the surgery group (HR = 1.14, 95% CI: 0.74-1.78; P = 0.55), indicating that esophageal preservation is a feasible alternative for patients attaining cCR after neoadjuvant chemoradiotherapy.
Quality of life was significantly better in the active surveillance group at both 6 months (P = 0.002) and 9 months (P = 0.007). Surgical outcomes were comparable, with R1 resection rates of 2% in both groups and 90-day postoperative mortality rates of 4% and 5%, respectively. Therefore, organ preservation has emerged as a shared goal for clinicians and patients, particularly as neoadjuvant chemoradiotherapy combined with immunotherapy achieves pCR rates exceeding 50%, suggesting that a substantial proportion of patients may avoid esophagectomy.
The critical challenge for esophagus preservation is accurately identifying patients with cCR. The preSANO study outlined a multimodal assessment protocol (1): Deep bite-on-bite biopsies under endoscopic ultrasonography to assess pathological regression in the primary tumor (2), Fine-needle aspiration of suspicious lymph nodes to evaluate regional lymph node status, and (3) 18-fluorodeoxyglucose positron emission tomography/computed tomography to detect potential distant metastases (53).
These methods yielded false-negative rates of 10% (95% CI: 4%–23%) for locoregional recurrence and 15% (95% CI: 7%-28%) for distant metastases. To improve accuracy in assessing primary tumor regression and lymph node status, incorporating magnetic resonance imaging alongside 18-fluorodeoxyglucose positron emission tomography/computed tomography is recommended, as observed morphological changes can aid in identifying pCR (54–56). Additionally, circulating tumor DNA analysis may enhance the performance of positron emission tomography/computed tomography, given that circulating tumor DNA-positive patients exhibit higher rates of distant metastases (15.1% vs. 3.3%) (53).
While effective, the complexity and cost of these detection methods make them impractical for all patients following chemoradiotherapy plus immunotherapy. Selective application is therefore advised for populations with a high likelihood of achieving cCR, necessitating the identification of predictive markers for treatment response.
Currently, programmed cell death ligand 1 expression is a commonly used biomarker. However, its predictive value for cCR in esophageal cancer clinical trials remains inconsistent (27–32). Furthermore, optimal cutoff values for tumor proportion score and combined positive score have yet to be firmly established, warranting caution in relying solely on programmed cell death ligand 1 to predict cCR.
Summary: High pCR rates from neoadjuvant immunotherapy plus chemoradiotherapy enable organ preservation strategies, as evidenced by the SANO trial where active surveillance was non-inferior to surgery for cCR patients and improved quality of life. However, accurately identifying candidates for preservation remains challenging due to limitations in current cCR assessment methods and the absence of validated biomarkers.
Future perspectives
Based on the highest current level of evidence, neoadjuvant chemoradiotherapy remains the recommended standard approach (4–7). Following this treatment, patients are advised to undergo esophagectomy. For those seeking organ preservation, a comprehensive efficacy assessment should be conducted. If cCR is confirmed, organ preservation may be considered; otherwise, surgical intervention is indicated. Postoperatively, patients achieving should transition to surveillance, while those without pCR require adjuvant immunotherapy.
In patients failing to achieve pCR after esophagectomy, adjuvant immunotherapy is essential. The CheckMate 577 trial demonstrated this principle by randomizing patients without pCR after neoadjuvant chemoradiotherapy and surgery (2:1 ratio) to receive nivolumab or placebo (57). Nivolumab significantly improved median disease-free survival versus placebo (22.4 vs. 11.0 months; HR = 0.69, 96.4% CI = 0.56-0.86; P < 0.001), with consistent benefits across all prespecified subgroups.
However, neoadjuvant immunotherapy is increasingly utilized in both trials and clinical practice, demonstrating substantial efficacy. Given the inconsistent outcomes associated with various neoadjuvant regimens (chemotherapy, radiotherapy, and immunotherapy), optimal strategies require further evaluation. The ongoing SCIENCE trial, a prospective, multicenter, randomized phase III study, aims to address this by enrolling 420 patients with locally advanced thoracic esophageal squamous cell carcinoma (58). Participants will be randomized (1:1:1) into three groups (1): neoadjuvant chemotherapy plus immunotherapy (2), neoadjuvant chemoradiotherapy plus immunotherapy, or (3) neoadjuvant chemoradiotherapy alone. This trial is expected to provide high-level evidence for this clinical dilemma.
Future research should prioritize identifying robust predictive biomarkers. Emerging evidence indicates that CD8+ Tex-SPRY1 cells enhance antitumor immunity by promoting a pro-inflammatory macrophage phenotype and supporting B cell function (59). Moreover, elevated levels of CD8+ and CD4+ tumor-infiltrating lymphocytes are associated with improved therapeutic outcomes (60). Additionally, patients with a high tumor mutation burden may derive greater survival benefits from immunotherapy (61). These biomarkers hold promise for predicting treatment response and enabling risk stratification.
Summary: The future treatment involves refining the current standard (chemoradiotherapy followed by surgery) by integrating organ preservation for cCR patients and adjuvant immunotherapy for non-pCR cases. While the SCIENCE trial seeks to define the optimal neoadjuvant approach incorporating immunotherapy; crucially, identifying reliable predictive biomarkers is essential for advancing personalized therapy.
Conclusion
Neoadjuvant triplet chemotherapy demonstrates superior systemic oncological control, reducing the incidence of distant metastases and thereby contributing to improved overall survival. In contrast, neoadjuvant chemoradiotherapy enhances locoregional control, evidenced by reduced local recurrent lesions, higher pCR rates, increased R0 resection rates, and decreased lymph node metastasis frequency, though without conferring overall survival benefits. Neoadjuvant immunotherapy combined with either chemotherapy or chemoradiotherapy achieves significantly higher pCR rates compared to chemotherapy or chemoradiotherapy alone. Critically, however, this elevated pCR rate does not correlate with improved overall survival or disease-free survival.
Author contributions
YAL: Data curation, Investigation, Writing – original draft. S-FW: Formal Analysis, Methodology, Writing – original draft. H-WL: Investigation, Writing – original draft. YL: Writing – original draft, Methodology. WH: Validation, Writing – original draft. X-BP: Conceptualization, Writing – review & editing.
Funding
The author(s) declare that no financial support was received for the research and/or publication of this article.
Conflict of interest
The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
Generative AI statement
The author(s) declare that no Generative AI was used in the creation of this manuscript.
Publisher’s note
All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.
Abbreviations
HR, hazard ratio; CI, confidence interval; pCR, pathological complete response; cCR, clinical complete response.
References
1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. (2021) 71:209–49. doi: 10.3322/caac.21660
2. Lagergren J, Smyth E, Cunningham D, and Lagergren P. Oesophageal cancer. Lancet. (2017) 390:2383–96. doi: 10.1016/S0140-6736(17)31462-9
3. Obermannova R, Alsina M, Cervantes A, Leong T, Lordick F, Nilsson M, et al. Oesophageal cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol. (2022) 33:992–1004. doi: 10.1016/j.annonc.2022.07.003
4. van Hagen P, Hulshof MC, van Lanschot JJ, Steyerberg EW, van Berge Henegouwen MI, Wijnhoven BP, et al. Preoperative chemoradiotherapy for esophageal or junctional cancer. N Engl J Med. (2012) 366:2074–84. doi: 10.1056/NEJMoa1112088
5. Eyck BM, van Lanschot JJB, Hulshof M, van der Wilk BJ, Shapiro J, van Hagen P, et al. Ten-year outcome of neoadjuvant chemoradiotherapy plus surgery for esophageal cancer: the randomized controlled CROSS trial. J Clin Oncol. (2021) 39:1995–2004. doi: 10.1200/JCO.20.03614
6. Yang H, Liu H, Chen Y, Zhu C, Fang W, Yu Z, et al. Long-term efficacy of neoadjuvant chemoradiotherapy plus surgery for the treatment of locally advanced esophageal squamous cell carcinoma: the NEOCRTEC5010 randomized clinical trial. JAMA Surg. (2021) 156:721–9. doi: 10.1001/jamasurg.2021.2373
7. Yang H, Liu H, Chen Y, Zhu C, Fang W, Yu Z, et al. Neoadjuvant chemoradiotherapy followed by surgery versus surgery alone for locally advanced squamous cell carcinoma of the esophagus (NEOCRTEC5010): A phase III multicenter, randomized, open-label clinical trial. J Clin Oncol. (2018) 36:2796–803. doi: 10.1200/JCO.2018.79.1483
8. Wang Z, Sun S, Li K, Huang C, Liu X, Zhang G, et al. Feasibility analysis of combined surgery for esophageal cancer. World J Surg Oncol. (2023) 21:41. doi: 10.1186/s12957-023-02930-0
9. Chandra RA, Keane FK, Voncken FEM, and Thomas CR Jr. Contemporary radiotherapy: present and future. Lancet. (2021) 398:171–84. doi: 10.1016/S0140-6736(21)00233-6
10. Atun R, Jaffray DA, Barton MB, Bray F, Baumann M, Vikram B, et al. Expanding global access to radiotherapy. Lancet Oncol. (2015) 16:1153–86. doi: 10.1016/S1470-2045(15)00222-3
11. Huang YC, Kung PT, Ho SY, Tyan YS, Chiu LT, and Tsai WC. Effect of multidisciplinary team care on survival of oesophageal cancer patients: a retrospective nationwide cohort study. Sci Rep. (2021) 11:13243. doi: 10.1038/s41598-021-92618-w
12. Lindblad M, Jestin C, Johansson J, Edholm D, and Linder G. Multidisciplinary team meetings improve survival in patients with esophageal cancer. Dis Esophagus. (2024) 37(11):doae061. doi: 10.1093/dote/doae061
13. Zhao S, Qi W, and Chen J. Role of a multidisciplinary team in administering radiotherapy for esophageal cancer. BMC Cancer. (2020) 20:974. doi: 10.1186/s12885-020-07467-z
14. Lv J, Cao XF, Zhu B, Ji L, Tao L, and Wang DD. Effect of neoadjuvant chemoradiotherapy on prognosis and surgery for esophageal carcinoma. World J Gastroenterol. (2009) 15:4962–8. doi: 10.3748/wjg.15.4962
15. Kumagai K, Rouvelas I, Tsai JA, Mariosa D, Klevebro F, Lindblad M, et al. Meta-analysis of postoperative morbidity and perioperative mortality in patients receiving neoadjuvant chemotherapy or chemoradiotherapy for resectable oesophageal and gastro-oesophageal junctional cancers. Br J Surg. (2014) 101:321–38. doi: 10.1002/bjs.9418
16. Jin HL, Zhu H, Ling TS, Zhang HJ, and Shi RH. Neoadjuvant chemoradiotherapy for resectable esophageal carcinoma: a meta-analysis. World J Gastroenterol. (2009) 15:5983–91. doi: 10.3748/wjg.15.5983
17. Yuan MX, Cai QG, Zhang ZY, Zhou JZ, Lan CY, and Lin JB. Application of neoadjuvant chemoradiotherapy and neoadjuvant chemotherapy in curative surgery for esophageal cancer: A meta-analysis. World J Gastrointest Oncol. (2024) 16:214–33. doi: 10.4251/wjgo.v16.i1.214
18. Csontos A, Fazekas A, Szako L, Farkas N, Papp C, Ferenczi S, et al. Effects of neoadjuvant chemotherapy vs chemoradiotherapy in the treatment of esophageal adenocarcinoma: A systematic review and meta-analysis. World J Gastroenterol. (2024) 30:1621–35. doi: 10.3748/wjg.v30.i11.1621
19. Lee JL, Park SI, Kim SB, Jung HY, Lee GH, Kim JH, et al. A single institutional phase III trial of preoperative chemotherapy with hyperfractionation radiotherapy plus surgery versus surgery alone for resectable esophageal squamous cell carcinoma. Ann Oncol. (2004) 15:947–54. doi: 10.1093/annonc/mdh219
20. Klevebro F, Alexandersson von Dobeln G, Wang N, Johnsen G, Jacobsen AB, Friesland S, et al. A randomized clinical trial of neoadjuvant chemotherapy versus neoadjuvant chemoradiotherapy for cancer of the oesophagus or gastro-oesophageal junction. Ann Oncol. (2016) 27:660–7. doi: 10.1093/annonc/mdw010
21. Tang H, Wang H, Fang Y, Zhu JY, Yin J, Shen YX, et al. Neoadjuvant chemoradiotherapy versus neoadjuvant chemotherapy followed by minimally invasive esophagectomy for locally advanced esophageal squamous cell carcinoma: a prospective multicenter randomized clinical trial. Ann Oncol. (2023) 34:163–72. doi: 10.1016/j.annonc.2022.10.508
22. Stiles BM and Altorki NK. The NeoRes trial: questioning the benefit of radiation therapy as part of neoadjuvant therapy for esophageal adenocarcinoma. J Thorac Dis. (2017) 9:3465–8. doi: 10.21037/jtd.2017.08.146
23. Ando N, Kato H, Igaki H, Shinoda M, Ozawa S, Shimizu H, et al. A randomized trial comparing postoperative adjuvant chemotherapy with cisplatin and 5-fluorouracil versus preoperative chemotherapy for localized advanced squamous cell carcinoma of the thoracic esophagus (JCOG9907). Ann Surg Oncol. (2012) 19:68–74. doi: 10.1245/s10434-011-2049-9
24. Kato K, Machida R, Ito Y, Daiko H, Ozawa S, Ogata T, et al. Doublet chemotherapy, triplet chemotherapy, or doublet chemotherapy combined with radiotherapy as neoadjuvant treatment for locally advanced oesophageal cancer (JCOG1109 NExT): a randomised, controlled, open-label, phase 3 trial. Lancet. (2024) 404:55–66. doi: 10.1016/S0140-6736(24)00745-1
25. Hoeppner J, Lordick F, Brunner T, Glatz T, Bronsert P, Rothling N, et al. ESOPEC: prospective randomized controlled multicenter phase III trial comparing perioperative chemotherapy (FLOT protocol) to neoadjuvant chemoradiation (CROSS protocol) in patients with adenocarcinoma of the esophagus (NCT02509286). BMC Cancer. (2016) 16:503. doi: 10.1186/s12885-016-2564-y
26. Hoeppner J, Brunner T, Schmoor C, Bronsert P, Kulemann B, Claus R, et al. Perioperative chemotherapy or preoperative chemoradiotherapy in esophageal cancer. N Engl J Med. (2025) 392:323–35. doi: 10.1056/NEJMoa2409408
27. Doki Y, Ajani JA, Kato K, Xu J, Wyrwicz L, Motoyama S, et al. Nivolumab combination therapy in advanced esophageal squamous-cell carcinoma. N Engl J Med. (2022) 386:449–62. doi: 10.1056/NEJMoa2111380
28. Sun JM, Shen L, Shah MA, Enzinger P, Adenis A, Doi T, 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) 398:759–71. doi: 10.1016/S0140-6736(21)01234-4
29. Luo H, Lu J, Bai Y, Mao T, Wang J, Fan Q, et al. Effect of camrelizumab vs placebo added to chemotherapy on survival and progression-free survival in patients with advanced or metastatic esophageal squamous cell carcinoma: the ESCORT-1st randomized clinical trial. JAMA. (2021) 326:916–25. doi: 10.1001/jama.2021.12836
30. Lu Z, Wang J, Shu Y, Liu L, Kong L, Yang L, et al. Sintilimab versus placebo in combination with chemotherapy as first line treatment for locally advanced or metastatic oesophageal squamous cell carcinoma (ORIENT-15): multicentre, randomised, double blind, phase 3 trial. BMJ. (2022) 377:e068714. doi: 10.1136/bmj-2021-068714
31. Xu J, Kato K, Raymond E, Hubner RA, Shu Y, Pan Y, et al. Tislelizumab plus chemotherapy versus placebo plus chemotherapy as first-line treatment for advanced or metastatic oesophageal squamous cell carcinoma (RATIONALE-306): a global, randomised, placebo-controlled, phase 3 study. Lancet Oncol. (2023) 24:483–95. doi: 10.1016/S1470-2045(23)00108-0
32. Wang ZX, Cui C, Yao J, Zhang Y, Li M, Feng J, et al. Toripalimab plus chemotherapy in treatment-naive, advanced esophageal squamous cell carcinoma (JUPITER-06): A multi-center phase 3 trial. Cancer Cell. (2022) 40:277–88 e3. doi: 10.1016/j.ccell.2022.02.007
33. Zhao XH, Gao HM, Wen JY, Wang HS, Wu LY, Song CY, et al. Immune checkpoint inhibitors combined with or without radio(chemo)therapy for locally advanced or recurrent/metastatic esophageal squamous cell carcinoma. Discov Oncol. (2023) 14:165. doi: 10.1007/s12672-023-00783-3
34. Hu HH, Xu X, Li XY, Zeng Y, Li Y, Song XY, et al. The value of intervention with radiotherapy after first-line chemo-immunotherapy in locally advanced or metastatic esophageal squamous cell carcinoma: A multi-center retrospective study. Clin Transl Radiat Oncol. (2024) 48:100818. doi: 10.1016/j.ctro.2024.100818
35. Ychou M, Boige V, Pignon JP, Conroy T, Bouche O, Lebreton G, et al. Perioperative chemotherapy compared with surgery alone for resectable gastroesophageal adenocarcinoma: an FNCLCC and FFCD multicenter phase III trial. J Clin Oncol. (2011) 29:1715–21. doi: 10.1200/JCO.2010.33.0597
36. Dai Y, Huang D, Zhao W, and Wei J. A comparative study of elective nodal irradiation and involved field irradiation in elderly patients with advanced esophageal cancer. Front Oncol. (2023) 13:1323908. doi: 10.3389/fonc.2023.1323908
37. Lyu J, Yisikandaer A, Li T, Zhang X, Wang X, Tian Z, et al. Comparison between the effects of elective nodal irradiation and involved-field irradiation on long-term survival in thoracic esophageal squamous cell carcinoma patients: A prospective, multicenter, randomized, controlled study in China. Cancer Med. (2020) 9:7460–8. doi: 10.1002/cam4.3409
38. Nakatani Y, Kato K, Shoji H, Iwasa S, Honma Y, Takashima A, et al. Comparison of involved field radiotherapy and elective nodal irradiation in combination with concurrent chemotherapy for T1bN0M0 esophageal cancer. Int J Clin Oncol. (2020) 25:1098–104. doi: 10.1007/s10147-020-01652-7
39. Liu M, Zhao K, Chen Y, and Jiang GL. Evaluation of the value of ENI in radiotherapy for cervical and upper thoracic esophageal cancer: a retrospective analysis. Radiat Oncol. (2014) 9:232. doi: 10.1186/s13014-014-0232-4
40. Chen X, Zhang Y, Zhou X, Wang M, Na F, Zhou L, et al. Involved-field irradiation or elective-nodal irradiation in neoadjuvant chemo-radiotherapy for locally-advanced esophageal cancer: comprehensive analysis for dosimetry, treatment-related complications, impact on lymphocyte, patterns of failure and survival. Front Oncol. (2023) 13:1274924. doi: 10.3389/fonc.2023.1274924
41. Wang H, Tang H, Fang Y, Tan L, Yin J, Shen Y, et al. Morbidity and mortality of patients who underwent minimally invasive esophagectomy after neoadjuvant chemoradiotherapy vs neoadjuvant chemotherapy for locally advanced esophageal squamous cell carcinoma: A randomized clinical trial. JAMA Surg. (2021) 156:444–51. doi: 10.1001/jamasurg.2021.0133
42. Qin J, Xue L, Hao A, Guo X, Jiang T, Ni Y, et al. Neoadjuvant chemotherapy with or without camrelizumab in resectable esophageal squamous cell carcinoma: the randomized phase 3 ESCORT-NEO/NCCES01 trial. Nat Med. (2024) 30:2549–57. doi: 10.1038/s41591-024-03064-w
43. Guo X, Chen C, Zhao J, Wang C, Mei X, Shen J, et al. Neoadjuvant chemoradiotherapy vs chemoimmunotherapy for esophageal squamous cell carcinoma. JAMA Surg. (2025) 160:565–74. doi: 10.1001/jamasurg.2025.0220
44. Ge F, Huo Z, Cai X, Hu Q, Chen W, Lin G, et al. Evaluation of clinical and safety outcomes of neoadjuvant immunotherapy combined with chemotherapy for patients with resectable esophageal cancer: A systematic review and meta-analysis. JAMA Netw Open. (2022) 5:e2239778. doi: 10.1001/jamanetworkopen.2022.39778
45. Zhang J, Zhao P, Xu R, Han L, Chen W, and Zhang Y. Comparison of the efficacy and safety of perioperative immunochemotherapeutic strategies for locally advanced esophageal cancer: a systematic review and network meta-analysis. Front Immunol. (2024) 15:1478377. doi: 10.3389/fimmu.2024.1478377
46. Li C, Zhao S, Zheng Y, Han Y, Chen X, Cheng Z, et al. Preoperative pembrolizumab combined with chemoradiotherapy for oesophageal squamous cell carcinoma (PALACE-1). Eur J Cancer. (2021) 144:232–41. doi: 10.1016/j.ejca.2020.11.039
47. Chen R, Liu Q, Li Q, Zhu Y, Zhao L, Liu S, et al. A phase II clinical trial of toripalimab combined with neoadjuvant chemoradiotherapy in locally advanced esophageal squamous cell carcinoma (NEOCRTEC1901). EClinicalMedicine. (2023) 62:102118. doi: 10.1016/j.eclinm.2023.102118
48. Liu Y, Men Y, Bao Y, Ma Z, Wang J, Pang Q, et al. Neoadjuvant immunochemoradiation therapy versus chemoradiation therapy in esophageal cancer: A systematic review and meta-analysis of reconstructed individual patient data. Int J Radiat Oncol Biol Phys. (2025) S0360-3016(25)00387-6. doi: 10.1016/j.ijrobp.2025.04.020
49. Wang H, Li S, Liu T, Chen J, and Dang J. Neoadjuvant immune checkpoint inhibitor in combination with chemotherapy or chemoradiotherapy in resectable esophageal cancer: A systematic review and meta-analysis. Front Immunol. (2022) 13:998620. doi: 10.3389/fimmu.2022.998620
50. Yang G, Yue H, Zhang X, Zeng C, Tan L, and Zhang X. Comparison of neoadjuvant chemotherapy or chemoradiotherapy plus immunotherapy for locally resectable esophageal squamous cell carcinoma. Front Immunol. (2024) 15:1336798. doi: 10.3389/fimmu.2024.1336798
51. Su F, Yang X, Yin J, Shen Y, and Tan L. Validity of using pathological response as a surrogate for overall survival in neoadjuvant studies for esophageal cancer: A systematic review and meta-analysis. Ann Surg Oncol. (2023) 30:7461–71. doi: 10.1245/s10434-023-13778-9
52. Noordman BJ, Wijnhoven BPL, Lagarde SM, Boonstra JJ, Coene P, Dekker JWT, et al. Neoadjuvant chemoradiotherapy plus surgery versus active surveillance for oesophageal cancer: a stepped-wedge cluster randomised trial. BMC Cancer. (2018) 18:142. doi: 10.1186/s12885-018-4034-1
53. Bondzi-Simpson A, Ribeiro T, Grant A, Ko M, Coburn N, Hallet J, et al. Patients with complete clinical response after neoadjuvant chemoradiotherapy for locally advanced esophageal cancer: A Markov decision analysis of esophagectomy versus active surveillance. J Thorac Cardiovasc Surg. (2024) 168(6):1538–1549.e1. doi: 10.1016/j.jtcvs.2024.04.020
54. Withey SJ, Owczarczyk K, Grzeda MT, Yip C, Deere H, Green M, et al. Association of dynamic contrast-enhanced MRI and (18)F-Fluorodeoxyglucose PET/CT parameters with neoadjuvant therapy response and survival in esophagogastric cancer. Eur J Surg Oncol. (2023) 49:106934. doi: 10.1016/j.ejso.2023.05.009
55. Xu X, Sun ZY, Wu HW, Zhang CP, Hu B, Rong L, et al. Diffusion-weighted MRI and (18)F-FDG PET/CT in assessing the response to neoadjuvant chemoradiotherapy in locally advanced esophageal squamous cell carcinoma. Radiat Oncol. (2021) 16:132. doi: 10.1186/s13014-021-01852-z
56. Borggreve AS, Goense L, van Rossum PSN, Heethuis SE, van Hillegersberg R, Lagendijk JJW, et al. Preoperative prediction of pathologic response to neoadjuvant chemoradiotherapy in patients with esophageal cancer using (18)F-FDG PET/CT and DW-MRI: A prospective multicenter study. Int J Radiat Oncol Biol Phys. (2020) 106:998–1009. doi: 10.1016/j.ijrobp.2019.12.038
57. Kelly RJ, Ajani JA, Kuzdzal J, Zander T, Van Cutsem E, Piessen G, et al. Adjuvant nivolumab in resected esophageal or gastroesophageal junction cancer. N Engl J Med. (2021) 384:1191–203. doi: 10.1056/NEJMoa2032125
58. He W, Bai H, Lv J, Tang P, Hu T, Zhou H, et al. Neoadjuvant chemotherapy or chemoradiotherapy plus sintilimab versus neoadjuvant chemoradiotherapy for locally advanced oesophageal squamous cell carcinoma: a study protocol of a multicentre, randomised, controlled, phase III trial (SCIENCE study). BMJ Open. (2025) 15:e095828. doi: 10.1136/bmjopen-2024-095828
59. Liu Z, Zhang Y, Ma N, Yang Y, Ma Y, Wang F, et al. Progenitor-like exhausted SPRY1(+)CD8(+) T cells potentiate responsiveness to neoadjuvant PD-1 blockade in esophageal squamous cell carcinoma. Cancer Cell. (2023) 41:1852–70 e9. doi: 10.1016/j.ccell.2023.09.011
60. Kiyozumi Y, Baba Y, Okadome K, Yagi T, Ishimoto T, Iwatsuki M, et al. IDO1 expression is associated with immune tolerance and poor prognosis in patients with surgically resected esophageal cancer. Ann Surg. (2019) 269:1101–8. doi: 10.1097/SLA.0000000000002754
Keywords: esophageal cancer, surgery, radiotherapy, chemotherapy, immunotherapy
Citation: Lin Y, Wang S-F, Liang H-W, Liu Y, Huang W and Pan X-B (2025) Preoperative radiotherapy in patients with locally advanced esophageal squamous cell carcinoma: a narrative review. Front. Oncol. 15:1613954. doi: 10.3389/fonc.2025.1613954
Received: 18 April 2025; Accepted: 07 July 2025;
Published: 22 July 2025.
Edited by:
Xuefeng Leng, Sichuan Cancer Hospital, ChinaReviewed by:
Alessio Vagliasindi, Oncological Center of Basilicata (IRCCS), ItalyRamon Mohanlal, Independent Researcher, New York, NY, United States
Copyright © 2025 Lin, Wang, Liang, Liu, Huang and Pan. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
*Correspondence: Xin-Bin Pan, cGFueGluYmluQGd4bXUuZWR1LmNu