The use of immunotherapy for esophageal cancer: a review of the current evidence and surgical implications
Review Article

The use of immunotherapy for esophageal cancer: a review of the current evidence and surgical implications

Kai Y. Li1, Mohamed K. Kamel2

1Department of Surgery, Central Michigan University, Mt Pleasant, MI, USA; 2Department of Surgery, Division of Thoracic and Foregut Surgery, University of Rochester Medical Center, Rochester, NY, USA

Contributions: (I) Conception and design: MK Kamel; (II) Administrative support: MK Kamel; (III) Provision of study materials or patients: KY Li; (IV) Collection and assembly of data: KY Li; (V) Data analysis and interpretation: KY Li; (VI) Manuscript writing: Both authors; (VII) Final approval of manuscript: Both authors.

Correspondence to: Mohamed K. Kamel, MD. Department of Surgery, Division of Thoracic and Foregut Surgery, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY 14620, USA. Email: Mohamed_Hussein@URMC.Rochester.edu; dr.m.kamel@Gmail.com.

Abstract: Esophageal cancer (EC) remains a leading cause of cancer-related mortality in the United States and worldwide, with limited therapeutic options for advanced disease. Immunotherapy agents, including immune checkpoint inhibitors (ICIs), have emerged as a promising treatment modality. This review explores the current role of immunotherapy in EC, evaluates its efficacy, safety, and impact on surgical outcomes. We conducted a comprehensive review of the literature, focusing on clinical trials, preclinical data, and emerging therapeutic strategies related to immunotherapy in EC. Studies were selected based on their relevance to ICIs, cancer vaccines, adoptive cell transfer, and novel agents such as T-cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domains (TIGIT) inhibitors. Data from pivotal trials, including CheckMate 577 and KEYNOTE studies, were highlighted. Immunotherapy, particularly with programmed cell death protein 1 (PD-1)/programmed cell death ligand 1 (PD-L1) inhibitors, has shown significant promise in both advanced and early-stage EC. Key trials have demonstrated improved overall survival and progression-free survival with ICIs, especially in patients with high PD-L1 expression. Neoadjuvant immunotherapy has also been explored, showing potential for improving surgical outcomes, including pathologic response rates and long-term survival. However, the effectiveness of immunotherapy varies across EC subtypes, and the long-term durability of response remains uncertain. While these therapies are promising, further research is required to optimize their use and integrate them into clinical practice.

Keywords: Esophageal cancer (EC); immunotherapy; adjuvant; neoadjuvant; perioperative


Received: 17 August 2024; Accepted: 08 April 2025; Published online: 28 April 2025.

doi: 10.21037/ccts-24-33


Introduction

Background

Esophageal cancer (EC) is the 8th most prevalent cancer and the 6th leading cause of cancer-related mortality in the United States (1). Despite a decreasing incidence of EC in the US, its incidence rates have escalated globally and are projected to rise further in the upcoming decades (2). In global hotspots such as East Asia and Africa, EC is currently predicted to claim more than 1 million lives each year (2).

Although EC is often discussed epidemiologically as a single entity, it comprises two main histologic subtypes: esophageal squamous cell carcinoma (eSCC) and esophageal adenocarcinoma (eAC). eSCC constitutes most EC cases worldwide and correlates with lifestyle factors such as tobacco and alcohol consumption, hot food intake, and consumption of nitrogenous compounds like processed meats and fish (3,4). Conversely, eAC is the predominant form of EC in Western countries and is primarily associated with obesity and gastroesophageal reflux disease (GERD) (3,5).

Rationale and knowledge gap

EC carries a grim prognosis, with an overall 5-year survival rate of 21%, plunging to 5.3% for advanced disease (6). This is largely due to the majority of patients presenting with locally advanced or metastatic disease (7). In 2012, the CROSS trial demonstrated a significant survival benefit in patients undergoing pre-operative chemoradiation compared to surgery alone for locally advanced, resectable EC (8). Since the original trial, subsequent studies have demonstrated that this survival benefit persists for at least 10 years (9). Additional efforts have been aimed at determining the optimal regimen, with multiple studies comparing the docetaxel-based fluorouracil + leucovorin + oxaliplatin + docetaxel (FLOT), anthracycline-based Medical Research Council Adjuvant Gastric Infusional Chemotherapy (MAGIC) and Chemoradiotherapy for Oesophageal Cancer followed by Surgery Study (CROSS) chemoradiotherapy regimens. Emerging evidence from recent randomized trials, including Al-Batran et al. and ESOPEC, has demonstrated the favorable perioperative outcomes with neoadjuvant chemotherapy alone (i.e., FLOT regimen) for eAC, with superior overall survival (OS) compared to neoadjuvant chemoradiation of the CROSS regimen (10,11). Additionally, preliminary results from the European Neo-AEGIS trial have suggested that peri-operative chemotherapy may be non-inferior to chemoradiation regimens, such as CROSS (12). More recently, immunotherapy has been increasingly used and has shown promising results in various malignancies including EC (13-15).

Objective

There is increasing interest in developing novel systemic therapy modalities for patients with EC, with immunotherapy emerging as a promising avenue. There have been prior discussions regarding the role of immunotherapy for EC, however, they have not discussed its implication with regard to esophageal surgery and surgical outcomes (16-19). This review aims to provide an overview of the current state of immunotherapy in EC treatment, focusing on its surgical implications.


Mechanisms of action of immunotherapy

Immunotherapy is now considered the fourth pillar of cancer management alongside chemotherapy, radiation, and surgical resection (20). The body naturally has immunologic defense mechanisms that can detect malignant cells and signal them for destruction. Therefore, pre-cancerous cells need to develop necessary mutations to conceal themselves from the immune system before they progress into cancer. Immunotherapy focuses on therapeutic modalities that enhance the patient’s antitumor immune response. There are five major types of immunotherapies: immune checkpoint inhibitors (ICIs), cancer vaccines, adoptive cell transfer (ACT), cytokine therapies, and viro-immunotherapy (21). In the treatment of EC, significant progress has been made in the realm of ICIs, cancer vaccines and ACT.

ICIs

Immune checkpoint proteins are molecules found on the surface of T- and B-cells that downregulate immune response and promote self-tolerance (22). These molecules, including cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), programmed cell death protein 1 (PD-1) or its ligand (PD-L1), are often overexpressed in cancer cells (23). This overexpression of immune checkpoints leads to a localized suppression of T-cell activity, which cancer cells utilize to grow and metastasize (24). Currently, the U.S. Food and Drug Administration (FDA) has approved two ICIs, nivolumab and pembrolizumab, for use in the treatment of EC.

Nivolumab

The use of nivolumab in EC was studied in the 2019 ATTRACTION-3 trial, where patients with unresectable advanced, recurrent, or metastatic eSCC who had previously undergone chemotherapy were randomized to either receive nivolumab or additional rounds of chemotherapy. In that study, nivolumab was shown to improve OS (10.5 vs. 8.0 months) with a favorable safety profile (25). These results have led to the FDA approving nivolumab as a second-line treatment for patients with unresectable or metastatic EC (26).

With the publication of Checkmate 577 in 2021, nivolumab has become a core component of EC therapy. In this global, randomized control trial, adults who had complete (R0) surgical resection with residual disease (≥ ypT1 and/or ≥ ypN1) after neoadjuvant chemoradiation were assigned to an immunotherapy group with nivolumab treatment or placebo with a primary endpoint of disease-free survival (27). Results showed that median disease-free survival was significantly improved in the nivolumab group compared to placebo (22.4 vs. 11.0 months) (27). Interestingly, benefit seemed to be independent of PD-L1 expression and actually had a greater survival benefit in eSCC than eAC (27). A subsequent study by the same team demonstrated that nivolumab was relatively well tolerated by patients without significant side effects or negative impact on quality of life (13). Based on the results of the CheckMate 577 study, the FDA approved nivolumab for adjuvant treatment in patients with completely resected EC who had residual disease following neoadjuvant chemotherapy (28).

Spurred by these results, the subsequent CheckMate 648 trial was conducted to investigate the use of nivolumab as a potential first-line treatment for EC. In this study, 970 patients with unresectable, advanced, recurrent or metastatic eSCC were randomized to receive nivolumab + chemotherapy, nivolumab + ipilimumab, or traditional chemotherapy to investigate differences in OS. The study demonstrated that both nivolumab + chemotherapy and nivolumab + ipilimumab groups demonstrated increased OS in the overall population compared to chemotherapy alone (13.2 vs. 12.7 vs. 10.7 months) (29). Based on these results, the FDA approved the use of nivolumab in combination with either chemotherapy or ipilimumab for the first-line treatment of advanced or metastatic eSCC (30).

Pembrolizumab

Pembrolizumab, another PD-1 inhibitor, has also been studied. In the phase II KEYNOTE-180 study, researchers studied pembrolizumab as a treatment option for patients with metastatic disease who had received two or more previous therapies. Overall, there was a response rate of 10%, with higher response rates noted in eSCC (31). The follow-up phase III KEYNOTE-181 trial then compared it to chemotherapy for patients with advanced, metastatic disease who had progressed on previous therapy. After 16 months, median survival was noted to be comparable in both groups, with a significantly decreased treatment-related adverse events, in the immunotherapy group (32). Of note, in a subgroup analysis of patients with high PD-L1 expression [combined positive score (CPS) >10], survival was substantially higher in the immunotherapy group (43% vs. 20%) (32). Based on these results, pembrolizumab has received FDA approval as a second line agent for patients with eSCC expressing PD-L1 (CPS >10) (26).

Similar to the CheckMate 648 trial conducted using nivolumab, the KEYNOTE-590 trial was conducted to investigate pembrolizumab + chemotherapy vs. placebo + chemotherapy in patients with previously untreated, locally advanced or metastatic EC that is not amenable to surgical resection or definitive chemoradiation. The results demonstrated that the pembrolizumab group has prolonged OS (6.3 vs. 5.8 months) when compared to placebo, with a more noticeable effect in patients with high PD-L1 expression (CPS >10) (7.5 vs. 5.5 months) (33). The FDA subsequently approved pembrolizumab in combination with chemotherapy for patients with locally advanced or metastatic EC (26).

The FDA-approved immunotherapies have quickly integrated into the National Comprehensive Cancer Network (NCCN) guidelines and are outlined in Table 1.

Table 1

Summary of currently FDA approved immunotherapies for esophageal cancer with their indications

Drug Study Indication
Nivolumab ATTRACTION-3 (25) Preferred second-line therapy for patients with unresectable advanced, recurrent, or metastatic eSCC after chemotherapy
Nivolumab CheckMate 577 (27) Patients with resected eSCC who have residual disease following neoadjuvant chemotherapy
Nivolumab CheckMate 648 (29) First-line therapy in combination with chemotherapy or ipilimumab for patients with advanced eSCC
Pembrolizumab KEYNOTE-181 (32) Second-line therapy for patients with esophageal SCC with high PD-L1 expression levels (CPS >10)
Pembrolizumab KEYNOTE-590 (33) First-line therapy in combination with chemotherapy for patients with advanced EC

CPS, combined positive score; EC, esophageal cancer; eSCC, esophageal squamous cell carcinoma; FDA, Food and Drug Administration; PD-L1, programmed cell death ligand 1.

Neo-adjuvant immunotherapy

Attention has recently turned towards using immunotherapy in addition to traditional chemoradiotherapy (nCRT) in the neoadjuvant setting. The PALACE-1 trial investigated the utilization of pembrolizumab combined with chemotherapy in the treatment of eSCC and found a pathologic complete response (pCR) rate of 55.6% (34). A similar trial, PEN-ICE, noted a pCR of 46.2% (35). Promising results have been shown with sintilimab in the ESONICT-1 trial, which showed a major pathologic response (MPR) rate of 52.2% and pCR rate of 21.7% in patients with eSCC (36). Spurred by these results, the ESONICT-2 trial showed a similar MPR of 70% and pCR of 16.7% with neoadjuvant toripalimab plus chemotherapy (37). High rates of MPR and pCR in neoadjuvant therapy have been associated with significantly improved outcomes (38). A study by Wong et al. using data from the National Cancer Database (NCDB) was able to show that patients undergoing immunotherapy plus nCRT had increased OS compared to nCRT alone (median OS: 69.1 vs. 56.3 months; 3-year OS of 65.6% vs. 55.0%) (39). On the other hand, in the PERFECT trial, the use of atezolizumab as part of a neo-adjuvant regimen was not associated with improvement in OS compared to chemoradiation alone (40). While studies on the use of neoadjuvant immunotherapy show promising outcomes, those results need to be validated in future long-term studies before such regimens can be integrated into the current practice guidelines (35,41,42).

Implications of immunotherapy on surgical outcomes

Several studies have investigated the implications of receiving neoadjuvant immunotherapy on surgical outcomes (19,43-47). Most of these neoadjuvant trials focused on safety, feasibility, and pathological outcomes, with common end points being MPR and R0 resection. In a pooled study, the completion rate of neoadjuvant therapy followed by surgery ranged from 49% to 100%, with key factors for early failure being treatment-related adverse events, disease progression or patient withdrawal (19). Of those that complete therapy, roughly 26.9% reported adverse events, with a pooled rate of MPR and pCR being 48.9% and 31.4%, respectively (43).

Traditionally, surgical resection is performed 4–6 weeks after completion of neoadjuvant therapy, to optimize pathologic reseponse as well as patients’ short- and long-term outcomes (48,49). There have also been several studies suggesting that a prolonged interval of 6–12 weeks might improve histological outcomes (50,51). However, the NeoRes II trial suggests that a prolonged interval to surgery following nCRT did not demonstrate significant improvement in pCR rate while being associated with a significant decrease in OS (14.2 vs. 26.5 months) (52). In the SCALE-1 trial and Xu et al., surgical resection was performed 4–6 weeks after completion of neoadjuvant immunochemoradiation (44,45). In both studies, the incorporation of neoadjuvant immunotherapy agents did not seem to affect the operative/perioperative outcomes.

Cheng et al. conducted a retrospective cohort study comparing neoadjuvant immunochemotherapy using pembrolizumab, tislelizumab, camrelizumab, sintilimab, or toripalimab to nCRT (46). They were unable to detect any difference in pathologic characteristics such as staging, rates of lympho-vascular invasions or MPR/pCR (46). Interestingly, they did note a statistically significant decrease in adverse operative outcomes, including decreased blood loss, operative time, and perioperative complication rates in the immunotherapy group (46). They attributed these findings to decreased adhesions and tissue edema secondary to a lack of radiation in the immunotherapy group. A similar study conducted by Wu and colleagues demonstrated that patients who received neoadjuvant immunotherapy regimens with camrelizumab, pembrolizumab, or sintilimab have a similar rate of perioperative complications and sustained a higher rate of MPR when compared with nCRT (53).

Reviewing data from the NCDB, immunotherapy did not appear to increase time to surgery, length of hospital stay, or rates of unplanned readmission (39). Data does suggest an association with increased lymph node harvest (17 vs. 15) and N-stage downstaging (49.7% vs. 40.2%) in patients undergoing immunotherapy (39). Researchers were ultimately able to conclude that, in this patient population, neoadjuvant immunotherapy was associated with an increase in long-term OS over 5 years (39). Once patients survive the first 5 years, recurrence is considered rare (54).

A phase I/II retrospective study was conducted by Sihag et al. at Memorial Sloan Kettering to investigate the safety of a durvalumab-based neoadjuvant regimen for EC (47). In terms of safety, they did not detect any difference in rates of major cardiopulmonary complications, anastomotic leak, or 30-/90-day mortality (47). Immunotherapy did not appear to affect the interval time to surgery nor the feasibility of minimally invasive approaches. At the same time, the rate of immune-related adverse events also appeared low, with only one patient requiring withdrawal due to hypersensitivity (47). Unlike Cheng et al.’s study, neither Sihag et al. nor Wu et al were able to demonstrate a statistical difference in operative blood loss or length of hospital stay (46,47,53). While these results may be promising, generalizability is limited by the relatively low sample sizes (<50 patients) and differences in surgeons’ experience and technique. A summary of the major immunotherapy trials for EC is outlined in Table 2.

Table 2

Summary of relevant esophageal cancer immunotherapy trials

Study Patients Treatment Pathology Endpoint Conclusion
ATTRACTION-3 (25) 419 Nivolumab vs. paclitaxel/docetaxel Advanced/recurrent eSCC OS Nivolumab improved survival compared to standard chemotherapy in patients with previously treated eSCC
CheckMate 577 (27) 532 Nivolumab vs. placebo R0 stage II/III EC + GEJ adenocarcinoma with residual disease DFS Nivolumab significantly increased disease-free survival
CheckMate 648 (29) 2687 Nivolumab + chemotherapy vs. chemotherapy alone Untreated, unresectable EC + GEJ adenocarcinoma OS, PFS Nivolumab + chemotherapy demonstrated superior OS and PFS to chemotherapy alone
KEYNOTE-180 (31) 121 Pembrolizumab Advanced/metastatic EC + GEJ adenocarcinoma resistant to prior therapy ORR Pembrolizumab had persistent antitumor activity with reasonable safety profile in patients with treatment-resistant EC
KEYNOTE-181 (32) 628 Pembrolizumab vs. paclitaxel/docetaxel/irontectan Advanced/metastatic EC + GEJ adenocarcinoma resistant to prior therapy OS Pembrolizumab prolonged survival vs. chemotherapy as second-line therapy in select patient populations
KEYNOTE-590 (33) 749 Pembrolizumab + 5-FU + cisplatin vs. chemotherapy alone Advanced/metastatic EC + GEJ adenocarcinoma OS, PFS Pembrolizumab plus chemotherapy improved overall survival compared to chemotherapy alone
PALACE-1 ( 34) 20 Pembrolizumab + carboplatin + paclitaxel II-IVa eSCC Safety Neoadjuvant immunotherapy was safe, did not delay surgery and offered good pCR
PEN-ICE (35) 18 Pembrolizumab + naplatin + paclitaxel II–IVa eSCC Safety Proposed regimen had few adverse events
ESONICT-1 (36) 30 Sintilimab + cisplatin + paclitaxel II–IV eSCC Safety, pCR Proposed regimen is safe and feasible with high pCR and few post-operative complications
ESONICT-2 (37) 20 Toripalimab + cisplatin + docetaxel III–IVa eSCC pCR, adverse events Proposed regimen had high pCR with few adverse events
SIN-ICE (41) 23 Sintilimab + nedaplatin + paclitaxel II–IVa eSCC Safety, pCR Proposed regimen had high pCR with few adverse events
NICE (42) 60 Camrelizumab + carboplatin + paclitaxel III–IVa eSCC pCR Proposed regimen had high rates of pCR
Sihag et al. (47) 168 Durvalumab + FOLFOX Locally advanced eAC + GEJ adenocarcinoma Safety Proposed regimen had few adverse events and not increase risk of post-operative complications

EC, esophageal cancer; eAC, esophageal adenocarcinoma; eSCC, esophageal squamous cell carcinoma; GEJ, gastroesophageal junction; OS, overall survival; pCR, pathological complete response; PFS, progression-free survival.

Novel immunotherapeutic agents

In addition to those advancements in utilizing ICIs in different clinical settings, promising prospects are also present in the development of novel ICIs. The checkpoint inhibitor T-cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domains (TIGIT) was found to be highly expressed in certain cancers, including EC, and is associated with worse outcomes (55). Novel inhibitors, including domvanalimab, have already been developed to target these receptors. For example, the ongoing STAR-221 trial aims to investigate the utility of these new inhibitors compared to nivolumab in the treatment of treatment-naïve, locally advanced, unresectable eAC (56).

Cancer vaccines

Traditionally, vaccines have been viewed as prophylactic agents that are administered to healthy patients to prevent disease. Recently, there has been a significant push towards the development of therapeutic vaccines, which are given to patients with pre-existing disease to promote a stronger immune response (57). Currently, the FDA has approved three different cancer vaccines: the Bacillus Calmette-Guerin vaccine for bladder cancer, Sipuleucel-T for metastatic prostate cancer, and Talimogene Laherparepvec for metastatic melanoma (58).

In the treatment of EC, two potential targets, New York esophageal squamous cell carcinoma 1 (NY-ESO-1) and melanoma-associated antigen (MAGE-A), have been identified as tumor-specific biomarkers and possible targets for molecular therapy (23,59-61). Kageyama et al. was able to demonstrate in a phase I clinical trial that an NY-ESO-1 based vaccine could be safe and promote an immune response in patients with metastatic EC, with higher doses promoting a stronger response (60). While CD4+ and CD8+ responses were seen in most patients, disease progression was eventually observed (60). Similar results were demonstrated with other agents such as Montanide ISA-51 and Pacibanil OK-432 (62).

A study conducted by Kono et al. suggests that a multi-peptide vaccine could be well-tolerated and help control disease in patients with advanced eSCC (63). In their sample of 10 patients, they were able to achieve temporary disease control in half of the patients, with a couple of patients developing complete response in metastatic lesions for several months (63). Similarly, a study using cancer vaccines in patients with metastatic eSCC demonstrated stable disease in 5 out of 9 patients, although complete/partial response was not observed (64). Promising results have also been shown with adjuvant vaccine therapy, with one phase II study demonstrating therapy in patients with resected eSCC showing improved cancer-specific survival (65).

Despite these findings, a phase II study was unable to detect a difference between NY-ESO-1 vaccine-treated patients who subsequently underwent nCRT and surgical resection vs. unvaccinated controls (66). It appears that, although short-term immune responses are generated, long-term effects and tumor immunosuppressive mechanisms limit vaccination effectiveness (67). Even with setbacks, new immunotherapy mechanisms such as pulse vaccinations and multi-target vaccination regimens are still being developed to overcome such shortcomings and offer promising results (66).

ACT

ACT involves the utilization of a patient’s immune cells to target and eliminate cancer cells (68). Two examples of ACT therapy include chimeric antigen receptor therapy (CAR-T) and T cell receptor therapy (TCR-T). Traditionally, ACT has been used in the treatment of hematologic cancers such as leukemia, lymphomas, and multiple myeloma, with multiple FDA-approved regimens (69-72). Fueled by this success, numerous ongoing studies aim to test the utility of CAR-T and additional ACT methods on solid tumors, including EC. Pre-clinical studies have identified numerous potential targets for ACT therapy for the treatment of EC. This includes the embryologic NY-ESO-1, which has previously been studied in the development of cancer vaccines, as well as ACT treatment of other solid tumors such as synovial sarcoma and melanoma (59,73,74). There are currently several ongoing phase I/II trials testing the efficacy of NY-ESO-1-based ACT, including NCT03638206 and NCT03638206, for the treatment of EC. Additional therapeutic strategies targeting other proteins including human epidermal growth factor receptor 2 (HER-2), epithelial cell adhesion molecule (EpCAM), and Mucin short variant S1 (MUC-1), have shown promising in vitro results as well and are currently being investigated in early phase I trials (75).


Strengths and limitations

Fortunately, while discussing EC, there is a strong body of literature to draw from. Given its global presence, there is ongoing discussion in multiple different patient populations and countries, notably in East Asia and the United States. In this study, we reviewed multiple recent pivotal trials which have contributed significantly to the integration of immunotherapy in clinical practice, as well as a discussion regarding emerging therapeutic agents. With that said, this analysis does have some limitations. First, the studies that review outcomes regarding neoadjuvant chemoimmunotherapy vs. chemoradiation vs. chemotherapy are often retrospective, which makes them vulnerable to selection bias (46,47,53). Additionally, many of the studies discussed are subject to subgroup variability. For example, multiple studies have noted that patients with high levels of PD-L1 receptors have improved outcomes regarding immunotherapy (26,32,33). Similarly, immunotherapy appears to have more favorable outcomes with eSCC compared to eAC (35-40). It is thought that this may be secondary to different tumor microenvironments with increased infiltration of CD8+/CD45RO+ cells and increased presence of lymphoid structures such as lymphoid tissue in eSCC (76,77). On the other hand, eAC tumors often have lower levels of CD8+ T cells and increased immune suppressive cell populations (78). The presence of such differences makes the generalizability of such discussions difficult.


Conclusions

Immunotherapy has emerged as a promising modality in the treatment of EC, offering new hope for patients. ICIs such as nivolumab and pembrolizumab have demonstrated improved outcomes in the neoadjuvant and adjuvant treatment of EC as well as cases of metastatic disease, prolonging survival. The success of these agents has led to their integration into NCCN guidelines. Beyond ICIs, ongoing research into cancer vaccines and ACT holds promise for expanding treatment options, with promising early preclinical trials.

The integration of immunotherapy into the multidisciplinary management of EC presents a significant opportunity for surgeons. Multiple studies have demonstrated an increase in pathologic response. At the same time, preliminary data suggest that inclusion of immunotherapy does not significantly impact surgical parameters such as leak rate and other post-operative complications. Future trials are essential to further refine these treatment strategies, expand their indications and improve outcomes for patients with this challenging disease.


Acknowledgments

None.


Footnote

Peer Review File: Available at https://ccts.amegroups.com/article/view/10.21037/ccts-24-33/prf

Funding: None.

Conflicts of Interest: Both authors have completed the ICMJE uniform disclosure form (available at https://ccts.amegroups.com/article/view/10.21037/ccts-24-33/coif). M.K.K. serves as an unpaid editorial board member of Current Challenges in Thoracic Surgery from March 2025 to February 2027. The other author has no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Sung H, Ferlay J, Siegel RL, 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. [Crossref] [PubMed]
  2. Liu CQ, Ma YL, Qin Q, et al. Epidemiology of esophageal cancer in 2020 and projections to 2030 and 2040. Thorac Cancer 2023;14:3-11. [Crossref] [PubMed]
  3. Wheeler JB, Reed CE. Epidemiology of esophageal cancer. Surg Clin North Am 2012;92:1077-87. [Crossref] [PubMed]
  4. Keszei AP, Goldbohm RA, Schouten LJ, et al. Dietary N-nitroso compounds, endogenous nitrosation, and the risk of esophageal and gastric cancer subtypes in the Netherlands Cohort Study. Am J Clin Nutr 2013;97:135-46. [Crossref] [PubMed]
  5. Coleman HG, Xie SH, Lagergren J. The Epidemiology of Esophageal Adenocarcinoma. Gastroenterology 2018;154:390-405. [Crossref] [PubMed]
  6. Institute NC. Cancer Stat Facts: Esophageal Cancer 2024. Available online: https://seer.cancer.gov/statfacts/html/esoph.html
  7. van Rossum PSN, Mohammad NH, Vleggaar FP, et al. Treatment for unresectable or metastatic oesophageal cancer: current evidence and trends. Nat Rev Gastroenterol Hepatol 2018;15:235-49. [Crossref] [PubMed]
  8. van Hagen P, Hulshof MC, van Lanschot JJ, et al. Preoperative chemoradiotherapy for esophageal or junctional cancer. N Engl J Med 2012;366:2074-84. [Crossref] [PubMed]
  9. Eyck BM, van Lanschot JJB, Hulshof MCCM, 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. [Crossref] [PubMed]
  10. Al-Batran SE, Homann N, Pauligk C, et al. Perioperative chemotherapy with fluorouracil plus leucovorin, oxaliplatin, and docetaxel versus fluorouracil or capecitabine plus cisplatin and epirubicin for locally advanced, resectable gastric or gastro-oesophageal junction adenocarcinoma (FLOT4): a randomised, phase 2/3 trial. Lancet 2019;393:1948-57. [Crossref] [PubMed]
  11. Hoeppner J, Brunner T, Lordick F, et al. Prospective randomized multicenter phase III trial comparing perioperative chemotherapy (FLOT protocol) to neoadjuvant chemoradiation (CROSS protocol) in patients with adenocarcinoma of the esophagus (ESOPEC trial). J Clin Oncol 2024;42:LBA1. [Crossref]
  12. Reynolds JV, Preston SR, O'Neill B, et al. Neo-AEGIS (Neoadjuvant trial in Adenocarcinoma of the Esophagus and Esophago-Gastric Junction International Study): Preliminary results of phase III RCT of CROSS versus perioperative chemotherapy (Modified MAGIC or FLOT protocol).(NCT01726452). J Clin Oncol 2021;41:295. [Crossref]
  13. Kelly RJ, Ajani JA, Kuzdzal J, et al. Adjuvant Nivolumab in Resected Esophageal or Gastroesophageal Junction Cancer. N Engl J Med 2021;384:1191-1203. Erratum in: N Engl J Med 2023;388:672. [Crossref] [PubMed]
  14. Shah MA, Kennedy EB, Alarcon-Rozas AE, et al. Immunotherapy and Targeted Therapy for Advanced Gastroesophageal Cancer: ASCO Guideline. J Clin Oncol 2023;41:1470-91. [Crossref] [PubMed]
  15. Park D, Jeon WJ, Yang C, et al. Advancing Esophageal Cancer Treatment: Immunotherapy in Neoadjuvant and Adjuvant Settings. Cancers (Basel) 2024;16:318. [Crossref] [PubMed]
  16. Liu Y, Bao Y, Yang X, et al. Efficacy and safety of neoadjuvant immunotherapy combined with chemoradiotherapy or chemotherapy in esophageal cancer: A systematic review and meta-analysis. Front Immunol 2023;14:1117448. [Crossref] [PubMed]
  17. Shoji Y, Koyanagi K, Kanamori K, et al. Immunotherapy for esophageal cancer: Where are we now and where can we go. World J Gastroenterol 2024;30:2496-501. [Crossref] [PubMed]
  18. Wei DD, Fang JM, Wang HZ, et al. Perioperative immunotherapy for esophageal squamous cell carcinoma. Front Immunol 2024;15:1330785. [Crossref] [PubMed]
  19. Liu Y. Perioperative immunotherapy for esophageal squamous cell carcinoma: Now and future. World J Gastroenterol 2023;29:5020-37. [Crossref] [PubMed]
  20. Hunter P. The fourth pillar: Despite some setbacks in the clinic, immunotherapy has made notable progress toward becoming an additional therapeutic option against cancer. EMBO Rep 2017;18:1889-92. [Crossref] [PubMed]
  21. Zhang Y, Zhang Z. The history and advances in cancer immunotherapy: understanding the characteristics of tumor-infiltrating immune cells and their therapeutic implications. Cell Mol Immunol 2020;17:807-21. [Crossref] [PubMed]
  22. Francisco LM, Sage PT, Sharpe AH. The PD-1 pathway in tolerance and autoimmunity. Immunol Rev 2010;236:219-42. [Crossref] [PubMed]
  23. Zhang C, Peng Y, Hublitz P, et al. Genetic abrogation of immune checkpoints in antigen-specific cytotoxic T-lymphocyte as a potential alternative to blockade immunotherapy. Sci Rep 2018;8:5549. [Crossref] [PubMed]
  24. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011;144:646-74. [Crossref] [PubMed]
  25. Kato K, Cho BC, Takahashi M, et al. Nivolumab versus chemotherapy in patients with advanced oesophageal squamous cell carcinoma refractory or intolerant to previous chemotherapy (ATTRACTION-3): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol 2019;20:1506-17. [Crossref] [PubMed]
  26. Ajani JA, D'Amico TA, Bentrem DJ, et al. Esophageal and Esophagogastric Junction Cancers, Version 2.2023, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2023;21:393-422. [Crossref] [PubMed]
  27. Kelly RJ, Ajani JA, Kuzdzal J, et al. Adjuvant Nivolumab in Resected Esophageal or Gastroesophageal Junction Cancer. N Engl J Med 2021;384:1191-203. [Crossref] [PubMed]
  28. Horiba MN, Casak SJ, Mishra-Kalyani PS, et al. FDA Approval Summary: Nivolumab for the Adjuvant Treatment of Adults with Completely Resected Esophageal/Gastroesophageal Junction Cancer and Residual Pathologic Disease. Clin Cancer Res 2022;28:5244-8. [Crossref] [PubMed]
  29. Doki Y, Ajani JA, Kato K, et al. Nivolumab Combination Therapy in Advanced Esophageal Squamous-Cell Carcinoma. N Engl J Med 2022;386:449-62. [Crossref] [PubMed]
  30. Administration USFaD. FDA approvals of Opdivo in combination with chemotherapy and Opdivo in combination with Yervoy for first-line advanced or metastatic esophageal squamous cell carcinoma, and Kymriah (tisagenlecleucel) for relapsed or refractory follicular lymphoma 2022. Available online: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-disco-burst-edition-fda-approvals-opdivo-combination-chemotherapy-and-opdivo-combination-yervoy
  31. Shah MA, Kojima T, Hochhauser D, et al. Efficacy and Safety of Pembrolizumab for Heavily Pretreated Patients With Advanced, Metastatic Adenocarcinoma or Squamous Cell Carcinoma of the Esophagus: The Phase 2 KEYNOTE-180 Study. JAMA Oncol 2019;5:546-50. [Crossref] [PubMed]
  32. Kojima T, Shah MA, Muro K, et al. Randomized Phase III KEYNOTE-181 Study of Pembrolizumab Versus Chemotherapy in Advanced Esophageal Cancer. J Clin Oncol 2020;38:4138-48. [Crossref] [PubMed]
  33. Sun JM, Shen L, Shah MA, et al. Pembrolizumab plus chemotherapy versus chemotherapy alone for first-line treatment of advanced oesophageal cancer (KEYNOTE-590): a randomised, placebo-controlled, phase 3 study. Lancet 2021;398:759-71. [Crossref] [PubMed]
  34. Li C, Zhao S, Zheng Y, et al. Preoperative pembrolizumab combined with chemoradiotherapy for oesophageal squamous cell carcinoma (PALACE-1). Eur J Cancer 2021;144:232-41. [Crossref] [PubMed]
  35. Duan H, Shao C, Pan M, et al. Neoadjuvant Pembrolizumab and Chemotherapy in Resectable Esophageal Cancer: An Open-Label, Single-Arm Study (PEN-ICE). Front Immunol 2022;13:849984. [Crossref] [PubMed]
  36. Zhang Z, Hong ZN, Xie S, et al. Neoadjuvant sintilimab plus chemotherapy for locally advanced esophageal squamous cell carcinoma: a single-arm, single-center, phase 2 trial (ESONICT-1). Ann Transl Med 2021;9:1623. [Crossref] [PubMed]
  37. Gao L, Lu J, Zhang P, et al. Toripalimab combined with docetaxel and cisplatin neoadjuvant therapy for locally advanced esophageal squamous cell carcinoma: a single-center, single-arm clinical trial (ESONICT-2). J Gastrointest Oncol 2022;13:478-87. [Crossref] [PubMed]
  38. Berger AC, Farma J, Scott WJ, et al. Complete response to neoadjuvant chemoradiotherapy in esophageal carcinoma is associated with significantly improved survival. J Clin Oncol 2005;23:4330-7. [Crossref] [PubMed]
  39. Wong LY, Liou DZ, Backhus LM, et al. The impact of neoadjuvant immunotherapy on perioperative outcomes and survival after esophagectomy for esophageal cancer. JTCVS Open 2023;14:547-60. [Crossref] [PubMed]
  40. van den Ende T, de Clercq NC, van Berge Henegouwen MI, et al. Neoadjuvant Chemoradiotherapy Combined with Atezolizumab for Resectable Esophageal Adenocarcinoma: A Single-arm Phase II Feasibility Trial (PERFECT). Clin Cancer Res 2021;27:3351-9. [Crossref] [PubMed]
  41. Duan H, Wang T, Luo Z, et al. A multicenter single-arm trial of sintilimab in combination with chemotherapy for neoadjuvant treatment of resectable esophageal cancer (SIN-ICE study). Ann Transl Med 2021;9:1700. [Crossref] [PubMed]
  42. Li Z, Liu J, Zhang M, et al. A phase II study of neoadjuvant immunotherapy combined with chemotherapy (camrelizumab plus albumin paclitaxel and carboplatin) in resectable thoracic esophageal squamous cell cancer (NICE study): Interim results. J Clin Oncol 2021;39:4060. [Crossref]
  43. Ge F, Huo Z, Cai X, 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. [Crossref] [PubMed]
  44. Xu X, Sun Z, Liu Q, et al. Neoadjuvant chemoradiotherapy combined with sequential perioperative toripalimab in locally advanced esophageal squamous cell cancer. J Immunother Cancer 2024;12:e008631. [Crossref] [PubMed]
  45. Jiang N, Zhang J, Guo Z, et al. Short-course neoadjuvant radiotherapy combined with chemotherapy and toripalimab for locally advanced esophageal squamous cell carcinoma (SCALE-1): a single-arm phase Ib clinical trial. J Immunother Cancer 2024;12:e008229. [Crossref] [PubMed]
  46. Cheng J, Guo M, Yang Y, et al. Perioperative Outcomes of Minimally Invasive Esophagectomy After Neoadjuvant Immunotherapy for Patients With Locally Advanced Esophageal Squamous Cell Carcinoma. Front Immunol 2022;13:848881. [Crossref] [PubMed]
  47. Sihag S, Ku GY, Tan KS, et al. Safety and feasibility of esophagectomy following combined immunotherapy and chemoradiotherapy for esophageal cancer. J Thorac Cardiovasc Surg 2021;161:836-843.e1. [Crossref] [PubMed]
  48. Xiao X, Cheng C, Cheng L, et al. Longer Time Interval from Neoadjuvant Chemoradiation to Surgery is Associated with Poor Survival for Patients Without Clinical Complete Response in Oesophageal Cancer. Ann Surg Oncol 2023;30:886-96. [Crossref] [PubMed]
  49. Ranney DN, Mulvihill MS, Yerokun BA, et al. Surgical resection after neoadjuvant chemoradiation for oesophageal adenocarcinoma: what is the optimal timing? Eur J Cardiothorac Surg 2017;52:543-51. [Crossref] [PubMed]
  50. Shapiro J, van Hagen P, Lingsma HF, et al. Prolonged time to surgery after neoadjuvant chemoradiotherapy increases histopathological response without affecting survival in patients with esophageal or junctional cancer. Ann Surg 2014;260:807-13; discussion 813-4. [Crossref] [PubMed]
  51. Klevebro F, Nilsson K, Lindblad M, et al. Association between time interval from neoadjuvant chemoradiotherapy to surgery and complete histological tumor response in esophageal and gastroesophageal junction cancer: a national cohort study. Dis Esophagus 2020;33:doz078. [Crossref] [PubMed]
  52. Nilsson K, Klevebro F, Sunde B, et al. Oncological outcomes of standard versus prolonged time to surgery after neoadjuvant chemoradiotherapy for oesophageal cancer in the multicentre, randomised, controlled NeoRes II trial. Ann Oncol 2023;34:1015-24. [Crossref] [PubMed]
  53. Wu Z, Zheng Q, Chen H, et al. Efficacy and safety of neoadjuvant chemotherapy and immunotherapy in locally resectable advanced esophageal squamous cell carcinoma. J Thorac Dis 2021;13:3518-28. [Crossref] [PubMed]
  54. Abou Chaar MK, Godin A, Harmsen WS, et al. Determinants of Long-term Survival Decades After Esophagectomy for Esophageal Cancer. Ann Thorac Surg 2023;116:1036-44. [Crossref] [PubMed]
  55. Xu D, Zhao E, Zhu C, et al. TIGIT and PD-1 may serve as potential prognostic biomarkers for gastric cancer. Immunobiology 2020;225:151915. [Crossref] [PubMed]
  56. Klempner SJ, Shitara K, Sison A, et al. STAR-221: A randomized, open-label, multicenter, phase 3 trial of domvanalimab, zimberelimab, and chemotherapy versus nivolumab and chemotherapy in previously untreated, locally advanced, unresectable or metastatic gastric, gastroesophageal junction, and esophageal adenocarcinoma. J Clin Oncol 2023;41:TPS4206. [Crossref]
  57. Lin MJ, Svensson-Arvelund J, Lubitz GS, et al. Cancer vaccines: the next immunotherapy frontier. Nat Cancer 2022;3:911-26. [Crossref] [PubMed]
  58. Hargrave A, Mustafa AS, Hanif A, et al. Recent Advances in Cancer Immunotherapy with a Focus on FDA-Approved Vaccines and Neoantigen-Based Vaccines. Vaccines (Basel) 2023;11:1633. [Crossref] [PubMed]
  59. Oshima Y, Shimada H, Yajima S, et al. NY-ESO-1 autoantibody as a tumor-specific biomarker for esophageal cancer: screening in 1969 patients with various cancers. J Gastroenterol 2016;51:30-4. [Crossref] [PubMed]
  60. Kageyama S, Wada H, Muro K, et al. Dose-dependent effects of NY-ESO-1 protein vaccine complexed with cholesteryl pullulan (CHP-NY-ESO-1) on immune responses and survival benefits of esophageal cancer patients. J Transl Med 2013;11:246. [Crossref] [PubMed]
  61. Forghanifard MM, Gholamin M, Farshchian M, et al. Cancer-testis gene expression profiling in esophageal squamous cell carcinoma: identification of specific tumor marker and potential targets for immunotherapy. Cancer Biol Ther 2011;12:191-7. [Crossref] [PubMed]
  62. Kakimi K, Isobe M, Uenaka A, et al. A phase I study of vaccination with NY-ESO-1f peptide mixed with Picibanil OK-432 and Montanide ISA-51 in patients with cancers expressing the NY-ESO-1 antigen. Int J Cancer 2011;129:2836-46. [Crossref] [PubMed]
  63. Kono K, Mizukami Y, Daigo Y, et al. Vaccination with multiple peptides derived from novel cancer-testis antigens can induce specific T-cell responses and clinical responses in advanced esophageal cancer. Cancer Sci 2009;100:1502-9. [Crossref] [PubMed]
  64. Iwahashi M, Katsuda M, Nakamori M, et al. Vaccination with peptides derived from cancer-testis antigens in combination with CpG-7909 elicits strong specific CD8+ T cell response in patients with metastatic esophageal squamous cell carcinoma. Cancer Sci 2010;101:2510-7. [Crossref] [PubMed]
  65. Yasuda T, Nishiki K, Hiraki Y, et al. Phase II Adjuvant Cancer-specific Vaccine Therapy for Esophageal Cancer Patients Curatively Resected After Preoperative Therapy With Pathologically Positive Nodes; Possible Significance of Tumor Immune Microenvironment in its Clinical Effects. Ann Surg 2022;275:e155-62. [Crossref] [PubMed]
  66. Ishikawa T, Kageyama S, Miyahara Y, et al. Safety and antibody immune response of CHP-NY-ESO-1 vaccine combined with poly-ICLC in advanced or recurrent esophageal cancer patients. Cancer Immunol Immunother 2021;70:3081-91. [Crossref] [PubMed]
  67. Gu YM, Zhuo Y, Chen LQ, et al. The Clinical Application of Neoantigens in Esophageal Cancer. Front Oncol 2021;11:703517. [Crossref] [PubMed]
  68. Farkona S, Diamandis EP, Blasutig IM. Cancer immunotherapy: the beginning of the end of cancer? BMC Med 2016;14:73. [Crossref] [PubMed]
  69. Wang K, Wei G, Liu D. CD19: a biomarker for B cell development, lymphoma diagnosis and therapy. Exp Hematol Oncol 2012;1:36. [Crossref] [PubMed]
  70. Tettamanti S, Biondi A, Biagi E, et al. CD123 AML targeting by chimeric antigen receptors: A novel magic bullet for AML therapeutics? Oncoimmunology 2014;3:e28835. [Crossref] [PubMed]
  71. Heffner LT, Jagannath S, Zimmerman TM, et al. BT062, an antibody-drug conjugate directed against CD138, given weekly for 3 weeks in each 4 week cycle: safety and further evidence of clinical activity. Blood 2012;120:4042. [Crossref]
  72. Ramos CA, Ballard B, Zhang H, et al. Clinical and immunological responses after CD30-specific chimeric antigen receptor-redirected lymphocytes. J Clin Invest 2017;127:3462-71. [Crossref] [PubMed]
  73. Ramachandran I, Lowther DE, Dryer-Minnerly R, et al. Systemic and local immunity following adoptive transfer of NY-ESO-1 SPEAR T cells in synovial sarcoma. J Immunother Cancer 2019;7:276. [Crossref] [PubMed]
  74. Robbins PF, Kassim SH, Tran TL, et al. A pilot trial using lymphocytes genetically engineered with an NY-ESO-1-reactive T-cell receptor: long-term follow-up and correlates with response. Clin Cancer Res 2015;21:1019-27. [Crossref] [PubMed]
  75. Zhu YG, Xiao BF, Zhang JT, et al. Genetically Modified T Cells for Esophageal Cancer Therapy: A Promising Clinical Application. Front Oncol 2021;11:763806. [Crossref] [PubMed]
  76. Conroy MJ, Kennedy SA, Doyle SL, et al. A study of the immune infiltrate and patient outcomes in esophageal cancer. Carcinogenesis 2021;42:395-404. [Crossref] [PubMed]
  77. Zhang D, Jiang D, Jiang L, et al. HLA-A(+) tertiary lymphoid structures with reactivated tumor infiltrating lymphocytes are associated with a positive immunotherapy response in esophageal squamous cell carcinoma. Br J Cancer 2024;131:184-95. [Crossref] [PubMed]
  78. Groen-van Schooten TS, Harrasser M, Seidel J, et al. Phenotypic immune characterization of gastric and esophageal adenocarcinomas reveals profound immune suppression in esophageal tumor locations. Front Immunol 2024;15:1372272. [Crossref] [PubMed]
doi: 10.21037/ccts-24-33
Cite this article as: Li KY, Kamel MK. The use of immunotherapy for esophageal cancer: a review of the current evidence and surgical implications. Curr Chall Thorac Surg 2025;7:8.

Download Citation