Minireviews Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Oncol. Jun 15, 2025; 17(6): 106707
Published online Jun 15, 2025. doi: 10.4251/wjgo.v17.i6.106707
Immune checkpoint inhibitors in the first-line treatment of esophageal squamous cell carcinoma: Minireview for a big shift
Giulia Massaro, Alexandra Paulet, School of Human Health Sciences, University of Florence, Florence 50134, Tuscany, Italy
Daniele Lavacchi, Oncology Unit, Careggi University Hospital, Florence 50134, Tuscany, Italy
Marco Brugia, Elisa Giommoni, Oncology Unit, Azienda Universitaria Ospedaliera Careggi, Florence 50134, Tuscany, Italy
Daniele Rossini, Serena Pillozzi, Lorenzo Antonuzzo, Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Tuscany, Italy
Martina Catalano, Giandomenico Roviello, Department of Health Sciences, University of Florence, Florence 50134, Tuscany, Italy
ORCID number: Martina Catalano (0000-0003-1856-2848); Serena Pillozzi (0000-0001-8861-8718); Lorenzo Antonuzzo (0000-0003-3349-5604); Giandomenico Roviello (0000-0001-5504-8237).
Co-first authors: Giulia Massaro and Alexandra Paulet.
Co-corresponding authors: Martina Catalano and Giandomenico Roviello.
Author contributions: Antonuzzo L and Roviello G contributed to the conceptualization of this study; Rossini D, Pillozzi S, and Roviello G were involved in the methodology; Lavacchi D, Rossini D, and Catalano M participated in the validation; Massaro G and Paulet A contributed to the investigation, resources and, writing the original draft of this manuscript; Massaro G curated data; Brugia M, Giommoni E, and Catalano M contributed to reviewing and editing this manuscript; Pillozzi S and Antonuzzo L contributed to the visualization of this study; Lavacchi D, Rossini D, and Catalano M participated in the supervision. Massaro G and Paulet A are co-first authors and contributed equally to this work. Both authors made significant intellectual contributions, including jointly designing the study, acquiring and analyzing experimental data, and co-writing the manuscript. Their collaborative efforts were integral to the development and completion of the research. Catalano M and Roviello G are co-corresponding authors and contributed equally to the supervision and oversight of this work. Both authors were jointly involved in guiding the research design, interpreting results, and revising the manuscript. Roviello G will serve as the primary corresponding author and will take lead responsibility for all communications with the journal throughout the submission, peer review, and publication process, as well as ensuring all journal administrative requirements are met.
Conflict-of-interest statement: All authors report no relevant conflicts of interest for this article.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Giandomenico Roviello, MD, PhD, Associate Professor, Department of Health Sciences, University of Florence, Viale Pieraccini, Florence 50134, Tuscany, Italy. giandomenico.roviello@unifi.it
Received: March 5, 2025
Revised: April 9, 2025
Accepted: April 23, 2025
Published online: June 15, 2025
Processing time: 100 Days and 19.1 Hours

Abstract

Esophageal cancer is an aggressive malignancy often diagnosed at advanced stages, with esophageal squamous cell carcinoma being the predominant subtype worldwide. Standard first-line chemotherapy provides limited survival benefits, with a median overall survival of less than 1 year. Recent advancements in immunotherapy, particularly immune checkpoint inhibitors (ICIs), have transformed the treatment landscape, improving overall survival and progression-free survival. However, response rates remain variable, with programmed death ligand 1 (PD-L1) expression being the primary predictive biomarker. The variability in PD-L1 testing methods and immune microenvironment alterations after prior treatments complicate patient selection for ICIs. Several phase 3 trials, including KEYNOTE-590 and CheckMate 648, have demonstrated the efficacy of ICIs combined with chemotherapy, particularly in patients positive for PD-L1. Despite these advances, long-term survival remains low, emphasizing the need for better biomarkers and novel therapeutic strategies. This review explored current first-line treatment options for esophageal squamous cell carcinoma, challenges in biomarker-based patient selection, and emerging therapeutic approaches.

Key Words: Esophageal squamous cell carcinoma; Immunotherapy; Immune checkpoint inhibitors; Programmed death ligand 1; First-line treatment

Core Tip: This paper examined the evolving landscape of first-line treatments for esophageal squamous cell carcinoma, highlighting the transformative impact of immune checkpoint inhibitors. While immune checkpoint inhibitors combined with chemotherapy have improved survival, response rates remain inconsistent due to variability in programmed death ligand 1 testing and immune microenvironment changes. We critically analyzed landmark phase 3 trials (KEYNOTE-590, CheckMate 648) and discussed the limitations of current biomarkers. Addressing the urgent need for more precise patient selection and novel therapeutic strategies, this review provided insights into emerging approaches that could enhance long-term survival in esophageal squamous cell carcinoma.



INTRODUCTION

Esophageal cancer (EC) is a highly aggressive malignancy that is frequently detected at advanced or metastatic stages. EC is the eleventh most common cancer worldwide with 511000 new cases in 2022 and the seventh-leading cause of cancer-related deaths with 445000 deaths in 2022[1]. There are two main subtypes of EC: Esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma. The risk factors and incidence vary between these two subtypes. ESCC is the most common histological subtype globally, accounting for more than 90% of all cases of EC, with particularly high incidence rates in Eastern Asia and Eastern Africa. The major risk factors for ESCC include smoking and alcohol consumption[2,3]. Spatial transcriptomic studies are exploring the mechanisms of progression from precancerous lesions to ESCC to find new biomarkers for early diagnosis and management[4,5].

The standard first-line chemotherapy regimen for advanced ESCC typically includes a platinum-based fluoropyrimidine doublet that offers a median overall survival (mOS) < 1 year[6]. In recent years, the treatment algorithm for unresectable, locally advanced, or metastatic ESCC has significantly evolved. Multiple randomized controlled trials have demonstrated a remarkable benefit from immune checkpoint inhibitors (ICIs), either alone or in combination with chemotherapy, in improving OS and progression-free survival (PFS)[7-12].

However, long-term survivors remain a small proportion of the total, with a 5-year OS around 10%. For most patients, prognosis remains poor, with a mOS of approximately 1 year[13]. The primary predictive factor for response to ICIs is the expression of programmed death ligand 1 (PD-L1). Nevertheless, for patients with low PD-L1 expression, the benefit of ICIs remains unclear, and a significant subset of these patients does not receive any advantage from immunotherapy[14]. The aim of this review was to explore the challenges in selecting the most appropriate therapeutic strategy for patients with ESCC, with a particular focus on first-line treatment options.

AVAILABLE BIOMARKERS

PD-L1 expression is currently the most established predictive biomarker for determining sensitivity to ICIs. For patients eligible for first-line treatment with ICIs, it is recommended to assess PD-L1 expression using immunohistochemistry (IHC). The clinical implementation of PD-L1 IHC testing is challenging due to the presence of multiple IHC assays, scoring systems, and cutoff thresholds. PD-L1 expression can be assessed using one of three main scoring methods: Tumor proportion score (TPS); combined positive score (CPS); or tumor area positivity (TAP).

TPS is calculated as the percentage of PD-L1-positive tumor cells relative to the total number of tumor cells, multiplied by 100. CPS is a cell-counting method that determines the ratio of the total number of PD-L1-positive cells (including tumor cells, mononuclear lymphocytes, and macrophages) to the total number of viable tumor cells, multiplied by 100. A minimum of 100 viable tumor cells must be present on the PD-L1-stained slide for a valid evaluation. TAP is a visually estimated scoring algorithm developed for the SP263 assay. It calculates the ratio of the area occupied by PD-L1-positive tumor and immune cells to the total tumor area. These scoring systems provide distinct approaches to quantifying PD-L1 expression and help guide therapeutic decisions in clinical practice.

Three major commercial IHC assays are commonly used to detect and quantify PD-L1 expression: Ventana PD-L1 SP263 (SP263); Dako PD-L1 IHC 22C3 pharmDx (22C3); and Dako PD-L1 IHC 28-8 pharmDx (28-8). Each assay employs a unique combination of antibody clone, staining platform, scoring algorithm, and cutoff for PD-L1 positivity. The variability in assays and cutoff thresholds (e.g., CPS ≥ 1/≥ 10, TPS ≥ 1%) complicates the identification of patients most likely to benefit from treatment. Additionally, resource limitations in many pathology laboratories and the lack of comparative studies on PD-L1 assays for EC further challenge their clinical implementation[15].

Another challenge of PD-L1 assessment is that immune dynamics are significantly influenced by prior chemoradiotherapy in pretreated patients, both in esophageal adenocarcinoma[16] and ESCC. Single-cell RNA sequencing of ESCC tumors revealed that neochemoradiotherapy enhances CD8+ T cell infiltration and drives their exhaustion, potentially compromising long-term antitumor immunity[17]. The proportion of patients with tumors expressing high levels of PD-L1 in the first-line studies KEYNOTE-590 and CheckMate 649 were 51% with PD-L1 CPS ≥ 10 and 60% with PD-L1 CPS ≥ 5), respectively. They were detected by different anti-programmed cell death protein 1 (anti-PD-1) antibody clones (22C3 and 28-8, respectively) and were higher than observed in the previous second-line study of pembrolizumab in patients with advanced esophageal cancer (KEYNOTE-181) and the third-line study of nivolumab in patients with advanced gastric or gastroesophageal junction cancer (Attraction-2)[8].

Similarly, immune profiling of 41 patients treated with neoadjuvant toripalimab and chemoradiotherapy demonstrated substantial shifts in immune cell composition, particularly an increase in CD86+ macrophage infiltration[18]. These findings highlight the evolving nature of the tumor immune microenvironment after treatment, complicating the accurate timing and interpretation of PD-L1 testing in pretreated patients.

Wang et al[18] conducted a study to evaluate inter-pathologist concordance for four PD-L1 antibodies in ESCC. Sixty-eight pathologists from 19 centers independently assessed the CPS for 22C3, SP263, SP142, and E1 L3N along with the Tumor Cell Positive Score and Immune Cell Positive Score. Fifty paraffin-embedded ESCC samples from patients at clinical stage T2-T4 were used. Results showed that except for SP263 CPS, Tumor Cell Positive Score for 22C3, SP142, and E1 L3N exhibited higher concordance with overall percent agreement values of 0.80, 0.85, and 0.86. The highest concordance was between SP263 and 22C3 (overall percent agreement 0.896). However, interchangeability between SP263 and 22C3 was not supported in standardized PD-L1 expression analyses[18].

EMERGING BIOMARKERS

ESCC is a highly aggressive malignancy with a poor prognosis, highlighting the urgent need for novel biomarkers to enhance early detection and treatment strategies. Recent research has identified several promising biomarkers. MicroRNA signatures have shown great potential, with an 8-microRNA panel demonstrating high diagnostic accuracy[19]. High hsa-miR-3665 promoter methylation levels have been proposed as a potential biomarker for ESCC progression[20]. Gene expression markers, such as PDZ-binding kinase, kinesin-13 family member 2C, and RAD51-associated protein 1, have exhibited high sensitivity in differentiating ESCC from normal tissues, further supporting their diagnostic and prognostic relevance[21].

DNA methylation plays a key role in ESCC by silencing apoptotic genes, impairing DNA repair, and altering the cell cycle, which together contribute to chemotherapy resistance with hypermethylated genes significantly influencing cancer response to chemotherapy, thus serving as valuable predictive markers for treatment outcomes[22]. Furthermore, spatial transcriptomics-derived markers, such as Deltex E3 ubiquitin ligase 3L and bone marrow stromal antigen 2, provide valuable insights into tumor heterogeneity and may serve as therapeutic targets, offering new avenues for precision medicine in ESCC[23]. Although these biomarkers show great potential, additional large-scale studies and clinical validation are necessary to translate these discoveries into better patient outcomes. Unfortunately, they are currently not available in clinical practice, neither from a diagnostic nor a predictive standpoint.

FIRST-LINE THERAPY OPTIONS

The landscape of first-line treatment for ESCC is undergoing rapid advancements, with phase 3 randomized controlled trials confirming the efficacy of anti-PD-1 inhibitors combined with chemotherapy. The main results are shown in Table 1. The first study that provided evidence of the benefit of combining immunotherapy with chemotherapy was the KEYNOTE-590 trial. It demonstrated significant OS and PFS improvements with chemotherapy (5-fluorouracil and cisplatin) plus pembrolizumab compared with the placebo. PD-L1 expression was assessed using the PD-L1 IHC 22C3 assay, with a CPS ≥ 10 considered positive. In patients with ESCC with PD-L1 CPS ≥ 10, mOS was 13.9 months with pembrolizumab vs 8.8 months with placebo [hazard ratio (HR) = 0.57, 95% confidence interval (CI): 0.43-0.75; P < 0.0001]. Similar benefits were observed in the overall study population (12.4 months vs 9.8 months; HR = 0.73, 95%CI: 0.62-0.86; P < 0.0001). PFS was also significantly prolonged in patients with ESCC both with PD-L1 CPS ≥ 10 (7.5 months vs 5.5 months; HR = 0.51, 95%CI: 0.41-0.65; P < 0.0001) and in the overall population (6.3 months vs 5.8 months; HR = 0.65, 95%CI: 0.55-0.76; P < 0.0001). In the intent-to-treat (ITT) population, objective response rate (ORR) was 45.0% in the pembrolizumab plus chemotherapy group compared with 29.3% in the placebo plus chemotherapy group[8]. Based on these results, the Food and Drug Administration (FDA) and European Medicines Agency (EMA) approved pembrolizumab in combination with platinum and fluoropyrimidine-based chemotherapy in the first-line setting.

Table 1 Main results of randomized controlled trials investigating first-line treatment in advanced esophageal squamous cell carcinoma, focusing on the outcomes of the programmed death ligand 1 positive subgroups.

Phase
Experimental treatment
Enrollment
PD-L1 assay
PD-L1 cutoff
Median follow-up
Median OS PD-L1 positive subgroups
Median OS in overall population
Median PFS PD-L1 positive subgroups
Median PFS in overall population
ORR PD-L1 positive subgroups
ORR in overall population
KEYNOTE-590[8], n = 7493Pembrolizumab + 5FU + cisplatinGlobal22C3 assayPD-L1 CPS ≥ 10 (n = 383)22.6 months (IQR: 19.6-27.1)13.5 months vs 9.4 months (HR = 0.62, 95%CI: 0.49-0.78, P < 0.0001)12.4 months vs 9.8 months (HR = 0.73, 95%CI: 0.62-0.86, P < 0.0001)7.5 months vs 5.5 months (HR = 0.51, 95%CI: 0.41-0.65, P < 0.0001)6.3 months vs 5.8 months (HR = 0.65, 95%CI: 0.55-0.76, P < 0.0001)51.1% vs 26.9%45.0% vs 29.3%
CheckMate 648[24], n = 6453Nivolumab + 5FU + cisplatinGlobal28-8 assayPD-L1 TPS ≥ 1% (n = 97)13 months115.4 months vs 9.1 months (HR = 0.54, 95%CI: 0.37-0.80, P < 0.001)13.2 months vs 10.7 months (HR = 0.74, 95%CI: 0.58-0.96, P = 0.002)6.9 months vs 4.4 months (HR = 0.65, 95%CI: 0.46-0.92, P = 0.002)5.8 months vs 5.6 months (HR = 0.81, 95%CI: 0.64-1.04, P = 0.04)53% vs 20%47% vs 27%
CheckMate 648[24], n = 6493Nivolumab + ipilimumab + 5FU + cisplatinGlobal28-8 assayPD-L1 TPS ≥ 1% (n = 97)13 months113.7 months vs 9.1 months (HR = 0.64, 95%CI: 0.46-0.90, P = 0.001)12.7 months vs 10.7 months (HR = 0.78, 95%CI: 0.62-0.98, P = 0.01)4.0 months vs 4.4 months (HR = 1.02, 95%CI: 0.73-1.43, P = 0.90)35% vs 20%28% vs 27%
ESCORT-1st[11], n = 5963Camrelizumab + paclitaxel + cisplatinChina6E8 antibodyPD-L1 TPS ≥ 1% (n = 329)10.8 months (IQR: 7.3-14.3)15.3 months vs 11.5 months (HR = 0.59, 95%CI: 0.43-0.80, P = 0.32)15.3 months vs 12.0 months (HR = 0.70, 95%CI: 0.56-0.88, P = 0.001)6.9 months vs 5.6 months (HR = 0.51, 95%CI: 0.39-0.67, P = 0.38)6.9 months vs 5.6 months (HR = 0.56, 95%CI: 0.46-0.68, P < 0.001)74.1% vs 65.6%72.1% vs 62.1%
ESCORT-1st[25], n = 5963Camrelizumab + paclitaxel + cisplatinChina6E8 antibodyPD-L1 TPS ≥ 1% (n = 329)24 months2/515.6 months vs 12.6 months (HR = 0.70, 95%CI: 0.58-0.84, P = 0.0001)/57.6 months vs 5.8 months (HR = 0.54, 95%CI: 0.45-0.65, P = 0.0001)/5/5
JUPITER-06[26], n = 5143Toripalimab + paclitaxel + cisplatinChinaJS311 assayPD-L1 CPS ≥ 1 (n = 401)7.1 months215.2 months vs 10.9 months (HR = 0.61, 95%CI: 0.44-0.87, P = 0.0056)17 months vs 11 months (HR = 0.58, 95%CI: 0.43-0.78, P = 0.0004)5.7 months vs 5.5 months (HR = 0.58, 95%CI: 0.44-0.75, P < 0.0001)5.7 months vs 5.5 months (HR = 0.58, 95%CI: 0.46-0.74, P < 0.0001)69.3% vs 52.1%
ORIENT-15[27], n = 6593Sintilimab + paclitaxel + cisplatinGlobal (> 97% China)22C3 assayPD-L1 CPS ≥ 10 (n = 381)16 months (IQR: 12.3-19.4)17.2 months vs 13.6 months (HR = 0.64, 95%CI: 0.48-0.85, P = 0.002)16.7 months vs 12.5 months (HR = 0.63, 95%CI: 0.51-0.78, P < 0.001)8.3 months vs 6.4 months (HR = 0.58 95%CI: 0.45-0.75, P < 0.001)7.2 months vs 5.7 months (HR = 0.56, 95%CI: 0.46-0.68, P < 0.001)68% vs 49%66% vs 45%
ASTRUM-007[29]3Serplulimab +5FU + cisplatinChina/3PD-L1 CPS ≥ 1 (n = 383)14.9 months (IQR: 8.8-19.7)15.3 months vs 11.8 months (HR = 0.68, 95%CI: 0.53-0.87, P = 0.0020)3/35.6 months vs 5.3 months (HR = 0.60, 95%CI: 0.48-0.75, P < 0.0001)3/357.6% vs 42.1%
RATIONALE 306[28], n = 6453Tislelizumab + cisplatin (or oxaliplatin) + capecitabine (or paclitaxel or 5FU)GlobalSP263 assayPD-L1 TAP score ≥ 10% (n = 223)16.3 months (IQR: 8.6-21.8)16.6 months vs 10.0 months (HR = 0.62, 95%CI: 0.44-0.87, P = 0.0029)17.2 months vs 10.6 months (HR = 0.66, 95%CI: 0.54-0.80, P < 0.0001)7.3 months vs 5.6 months (HR = 0.62, 95%CI: 0.52-0.75, P < 0.0001)63% vs 42%
SKYSCRAPER-08[30], n = 4613Tiragolumab + atezolizumab + paclitaxel + cisplatinAsiaSP263 assayPD-L1 TAP score ≥ 10% (n = 199)14.6 months418.9 months vs 10.5 months (HR = 0.70, 95%CI: 0.49-1.01)15.7 months vs 11.1 months (HR = 0.70, 95%CI: 0.55-0.88, P = 0.0024)6.2 months vs 5.4 months (HR = 0.56; 95%CI: 0.45-0.70, P = 0.0001)59.7% vs 45.5%
MORPHEUS-EC[31], n = 1521/2bTiragolumab + atezolizumab + 5FU + cisplatinAsiaSP263 assayPD-L1 TAP score ≥ 10% (n = 40)10.9 months416 months vs 9.9 months (HR = 0.64, 95%CI: 0.35-1.20)6.9 months vs 4.1 months (HR = 0.52, 95%CI: 0.30-0.91)67.7% vs 47.8%
MORPHEUS-EC[31], n = 1521/2bAtezolizumab + 5FU + cisplatinAsiaSP263 assayPD-L1 TAP score ≥ 10% (n = 40)11.4 months413.1 months vs 9.9 months (HR = 0.75, 95%CI: 0.40-1.39)6.8 months vs 4.1 months (HR = 0.75, 95%CI: 0.44-1.26)53.8% vs 47.8%

The CheckMate 648 trial assessed the efficacy of nivolumab combined with chemotherapy (fluorouracil and cisplatin), nivolumab with ipilimumab, or chemotherapy alone in patients with advanced, unresectable ESCC regardless of PD-L1 expression that was determined using the PD-L1 IHC 28-8 pharmDx assay, with a PD-L1 ≥ 1% considered positive. Among patients with PD-L1 ≥ 1%, mOS was 15.4 months for patients receiving nivolumab plus chemotherapy compared with 9.1 months for those treated with chemotherapy alone (HR = 0.54, 95%CI: 0.37-0.80; P < 0.001). Similarly, nivolumab plus ipilimumab improved survival, with a mOS of 13.7 months vs 9.1 months for chemotherapy alone (HR = 0.64, 95%CI: 0.46-0.90; P = 0.001). In the overall study population, mOS was 13.2 months for nivolumab plus chemotherapy compared to 10.7 months for chemotherapy alone (HR = 0.74, 99.1%CI: 0.58-0.96; P = 0.002), while nivolumab plus ipilimumab achieved a mOS of 12.7 months vs 10.7 months (HR = 0.78, 95%CI: 0.62-0.98; P = 0.01). PFS was significantly improved with nivolumab plus chemotherapy in patients with PD-L1 ≥ 1% with a median PFS (mPFS) of 6.9 months compared with 4.4 months with chemotherapy alone (HR = 0.65, 95%CI: 0.46-0.92; P = 0.002). In contrast, no significant difference in PFS was observed between the nivolumab plus ipilimumab group and the chemotherapy-alone group (4.0 months vs 4.4 months, HR = 1.02, 95%CI: 0.73-1.43; P = 0.90). ORR was notably higher with nivolumab plus chemotherapy than with chemotherapy alone, reaching 53% vs 20% in patients with PD-L1 ≥ 1% and 47% vs 27% in the overall population. Furthermore, in the nivolumab plus ipilimumab cohort, the ORR for patients with PD-L1 ≥ 1% was 35%[24].

Based on CheckMate 648, the FDA approved nivolumab in combination with fluoropyrimidine-based and platinum-based chemotherapy and nivolumab in combination with ipilimumab for the first-line treatment of patients with advanced or metastatic ESCC, irrespective of tumor PD-L1 status. EMA approval is restricted to patients with tumor cell PD-L1 expression of ≥ 1%.

The fully Asian ESCORT-1st trial showed efficacy of camrelizumab in combination with chemotherapy (paclitaxel and cisplatin) compared with placebo plus chemotherapy. PD-L1 expression was evaluated using a PD-L1 IHC kit (6E8 antibody: Abcam) and categorized based on the TPS. At a minimum follow-up of 24 months the results demonstrated a statistically significant improvement in OS (15.3 months vs 12.0 months, HR = 0.70, 95%CI: 0.56-0.88; P < 0.0001) and in PFS (6.9 months vs 5.6 months, HR = 0.51, 95%CI: 0.39-0.67; P < 0.0001) in the ITT population. At a median follow up of 10.8 months, HR for death was below 1 in those patients with PD-L1 expression ≥ 1%, both for mOS (15.3 months vs 11.5 months, HR = 0.59, 95%CI: 0.43-0.80; P = 0.32) and mPFS (6.9 months vs 5.6 months, HR = 0.51, 95%CI: 0.39-0.67; P = 0.38). This suggests a potential survival benefit also for patients with PD-L1 < 1%, although the result was not statistically significant. ORR were observed in 72.1% of patients in the camrelizumab-chemotherapy group compared with 62.1% of patients in the placebo-chemotherapy group, showing a 10.1% difference between the groups (95%CI: 2.6%-7.6%; P = 0.009)[11,25]. Camrelizumab combination therapy for advanced or metastatic ESCC has been approved by National Medical Products Administration in China.

Similarly, the JUPITER-06 trial found toripalimab combined with paclitaxel and cisplatin vs placebo with paclitaxel and cisplatin significantly improved survival in Asian patients. PD-L1 expression was measured using the JS311 IHC assay, with PD-L1 CPS ≥ 1 considered positive and CPS ≥ 10 indicating high expression. PFS was significantly better in the toripalimab group, with a mPFS of 5.7 months compared to 5.5 months in the placebo group (HR = 0.58, 95%CI: 0.46-0.74; P < 0.0001). In the PD-L1 CPS ≥ 10 subgroup (HR = 0.65, 95%CI: 0.45-0.92; P = 0.02), and in the CPS < 10 subgroup (HR = 0.56, 95%CI: 0.41-0.78). In the overall population, OS was significantly improved in the toripalimab group, with 17 months in the experimental arm vs 11 months in the placebo group (HR = 0.58, 95%CI: 0.43-0.78; P = 0.0004), maintaining a positive trend in PD-L1 CPS ≥ 10 subgroup, though without reaching statistical significance (17.0 months vs 10.9 months, HR = 0.64, 95%CI: 0.40-1.03; P = 0.06)[26]. Toripalimab has received approval in China and Europe for the first-line treatment of advanced ESCC in combination with chemotherapy, demonstrating efficacy across various PD-L1 expression levels. However, it is not currently approved for this indication in the United States.

The ORIENT-15 trial demonstrated superiority of sintilimab combined with chemotherapy (cisplatin plus paclitaxel or cisplatin plus 5-fluorouracil) compared with placebo plus chemotherapy. PD-L1 expression was evaluated using the 22C3 pharmDx assay, considering both the TPS and CPS scoring systems. The primary endpoint was OS in both the overall population and the subgroup with PD-L1 CPS ≥ 10. In the overall population, results demonstrated a significant improvement in OS for the sintilimab-chemotherapy group compared with the placebo-chemotherapy group, with a mOS of 16.7 months vs 12.5 months (HR = 0.63, 95%CI: 0.51-0.78; P < 0.001). Among patients with PD-L1 CPS ≥ 10, mOS was 17.2 months in the sintilimab-chemotherapy group vs 13.6 months in the placebo group (HR = 0.64, 95%CI: 0.48-0.85; P = 0.002). Similarly, sintilimab-chemotherapy demonstrated significantly improved PFS (7.2 months vs 5.7 months (HR = 0.56, 95%CI: 0.46-0.68; P < 0.001), especially in PD-L1 CPS ≥ 10 subgroup (8.3 months vs 6.4 months, HR = 0.58, 95%CI: 0.45-0.75, P < 0.001)[27]. In summary, sintilimab has received approval from the National Medical Products Administration in China for use in combination with chemotherapy as a first-line treatment for ESCC, applicable to patients regardless of PD-L1 expression status. However, it has not yet been approved by the FDA or EMA for this indication.

Another trial that investigated a combination immunochemotherapy strategy was the RATIONAL306 trial comparing tislelizumab plus chemotherapy (oxaliplatin or cisplatin plus fluoropyrimidine or paclitaxel) vs placebo plus chemotherapy. PD-L1 expression status by TAP score was tested using the VENTANA PD-L1 (SP263) assay. mOS was 17.2 months in tislelizumab group vs 10.6 months in the placebo group (HR = 0.66, 95%CI: 0.54-0.80; P < 0.000). PD-L1 TAP score positivity thresholds of 10% showed significant differences in OS benefit (16.6 months vs 10.0 months, HR = 0.62, 95%CI: 0.44-0.87; P = 0·0029). Significant improvement in PFS was observed with tislelizumab plus chemotherapy vs placebo plus chemotherapy in the overall population (7.3 months vs 5.6 months, HR = 0.62, 95%CI: 0.52-0.75; P < 0·0001)[28].

Astrum-007 is a phase 3, double-blind, multicenter trial that assessed the effectiveness and safety of serplulimab combined with chemotherapy (fluoropyrimidine and cisplatin) vs chemotherapy alone in patients with advanced or metastatic ESCC and PD-L1 CPS ≥ 1. The study demonstrated that mOS was significantly longer in the serplulimab plus chemotherapy group compared with the placebo plus chemotherapy group (15.3 months vs 11.8 months, HR = 0.68, 95%CI: 0.53-0.87; P = 0.0020). The mPFS was 5.8 months in the serplulimab group and 5.3 months in the control group (HR = 0.60, 95%CI: 0.48-0.75, P < 0.0001). In patients with PD-L1 CPS ≥ 10, adding serplulimab to chemotherapy resulted in even greater survival benefits, both in mPFS and in mOS. Antitumor responses were also enhanced by serplulimab in both the PD-L1 CPS ≥ 10 and 1 ≤ CPS < 10 subgroups[29].

To overcome resistance to chemotherapy plus anti-PD-L1/anti-PD-1 therapies, studies, such as SKYSCRAPER-08 and MORPHEUS-EC trials, have been designed to incorporate new immunological targets. The phase 3 SKYSCRAPER-08 study evaluated the combination of tiragolumab (anti-TIGIT) plus atezolizumab with chemotherapy as a first-line treatment for advanced ESCC in an Asian patient population. TIGIT is an emerging inhibitory checkpoint expressed on activated T cells and natural killer cells, and tiragolumab may have the potential to enhance immune responses by working in synergy with other immunotherapeutic agents. A total of 461 patients were randomized to receive either tiragolumab and atezolizumab with chemotherapy or placebo with chemotherapy for up to six cycles, followed by maintenance therapy. At a minimum follow-up of 6.5 months, median independent review facility-assessed PFS was 6.2 months with tiragolumab vs 5.4 months with placebo (HR = 0.56, 95%CI: 0.45-0.70; P < 0.0001). The final OS analysis at 14.5 months showed a mOS of 15.7 months in the tiragolumab arm vs 11.1 months in the placebo arm (HR = 0.70, 95%CI: 0.55-0.88; P = 0.0024), confirming a significant and clinically meaningful survival benefit. These benefits were observed consistently across subgroups, regardless of PD-L1 expression levels[30].

In the MORPHEUS-EC phase Ib/II study a total of 152 patients were randomly assigned to one of three groups: Chemotherapy alone; atezolizumab plus chemotherapy; or tiragolumab plus atezolizumab with chemotherapy. mPFS was 4.1 months for chemotherapy, 6.8 months for atezolizumab plus chemotherapy, and 6.9 months for tiragolumab plus atezolizumab with chemotherapy. The survival benefit was consistent across various subgroups, including those based on PD-L1 status, with a trend toward improved OS in the tiragolumab arm[31].

The statistical analysis methods were similar across the studies. The Kaplan-Meier method was used to estimate OS, PFS, and duration of response. Differences in OS and PFS between groups were assessed using a stratified log-rank test, while HRs and 95%CIs were estimated using a Cox proportional hazards model. In KN590 the ORR differences were evaluated with the stratified Miettinen-Nurminen method[8]. In ORIENT-15 ORR and disease control rate were calculated using the normal approximation method with corresponding 95%CIs, and ORR differences between treatment groups were assessed using the Miettinen-Nurminen method[27]. In the RATIONAL-306 and ESCORT-1 study the Cochran-Mantel-Haenszel test was used to assess the ORR, with Clopper-Pearson 95%CIs[28,25].

The choice of chemotherapy varied across first-line chemoimmunotherapy trials, with paclitaxel plus cisplatin being more commonly used in studies focused on the Chinese population, whereas fluoropyrimidine plus cisplatin was the preferred regimen in global trials. Despite variations in chemotherapy protocols and PD-1 inhibitors, the survival benefits associated with adding a PD-1 inhibitor remained comparable across these studies[8,11,24-31]. Moreover, findings from the RATIONALE-306 trial indicated no significant difference in OS HR between patients treated with fluoropyrimidine plus cisplatin and those receiving paclitaxel plus cisplatin (0.66 vs 0.69)[28]. That said, key efficacy measures appeared to be more favorable in trials evaluating paclitaxel plus cisplatin plus PD-1 blockade compared with those assessing fluoropyrimidine plus cisplatin plus PD-1 blockade. While geographic factors (China vs other regions) may have influenced these results, they do not fully account for the observed differences.

Interestingly, in the Chinese subgroup of the KEYNOTE-590 trial, fluoropyrimidine plus cisplatin plus PD-1 blockade achieved a strong OS HR of 0.51, yet its median OS remained lower than that seen in Chinese trials using paclitaxel plus cisplatin[8]. One possible explanation is that paclitaxel in contrast to 5-fluorouracil has a greater potential to induce immunogenic cell death and enhance anti-tumor immune responses[32]. This characteristic may contribute to the superior efficacy observed with paclitaxel plus cisplatin plus PD-1 blockade. Furthermore, paclitaxel plus cisplatin is more convenient to administer than fluoropyrimidine plus cisplatin, making it a compelling option for further research in populations beyond China.

Regardless of the chemotherapy backbone, given these different treatment options, several researchers have focused on evaluating the cost-effectiveness of chemotherapy-immunotherapy combinations compared with monotherapy among currently approved regimens. The findings vary depending on the type of immunotherapy used. While the combination of toripalimab, sintilimab, and camrelizumab with chemotherapy has been shown to be cost-effective, the addition of serplulimab, pembrolizumab, or nivolumab to chemotherapy has not been considered a cost-effective option[33,34].

SAFETY PROFILE

The analysis of adverse events (AEs) across multiple studies shows no significant differences between treatment arms in terms of overall toxicity. In KN590, nearly all patients experienced AE, with grade 3 or higher events occurring at similar rates between the pembrolizumab group and the placebo group (86% and 83%, respectively)[8]. Likewise, treatment-related events were comparable in frequency and severity. The most common immune-mediated AEs were hypothyroidism, pneumonitis, and hyperthyroidism[8]. In CM648, grade 3-4 AEs were more common in the nivolumab plus chemotherapy arm (47%) than in the nivolumab plus ipilimumab (32%) arm or chemotherapy arm (36%), but treatment-related mortality remained low across all groups (about 2%)[24].

In patients receiving nivolumab plus chemotherapy, AEs were mainly chemotherapy-related, with nausea, decreased appetite, and stomatitis being the most common, along with some immune-mediated events. In contrast, nivolumab plus ipilimumab primarily led to immune-mediated AEs, most commonly rash, pruritus, and hypothyroidism[24]. The ESCORT-1 and JUPITER-06 studies reported high rates of treatment-related AEs, but severe toxicities (grade ≥ 3) were comparable between immunotherapy and placebo arms[11,25,26]. The most common immune-related AE with camrelizumab was reactive capillary endothelial proliferation (79.9% vs 10.8%)[11]. Similarly, in sintilimab and tislelizumab trials, AEs rates were similar between treatment groups, with immune-related toxicities being more frequent in immunotherapy arms but mostly mild (grade 1-2)[27,28].

In SKYSCRAPER-08, 98.2% of patients experienced treatment-related AEs, with a slightly higher incidence of grade 3/4 events in the tiragolumab + atezolizumab + chemotherapy group (59.6% vs 56.4%)[30]. However, grade 5 events were more frequent with tiragolumab (2.6% vs 0.9%). MORPHEUS-EC showed a similar trend, with grade 3-5 AEs occurring in 82.6% (chemotherapy), 83.1% (atezolizumab + chemotherapy), and 85.5% (tiragolumab + atezolizumab + chemotherapy), indicating a modest increase in serious AEs (SAEs) with the addition of tiragolumab[31]. Overall, while ICIs are generally associated with several immune-related toxicities, their safety profile remains comparable with chemotherapy, with most SAEs occurring at similar rates across treatment groups.

In the ASTRUM-007 study, the incidence of AEs of any grade was similar between the ITT population (98%) and the control population (98%). Immune-related AEs occurred in 35% of patients in the serplulimab plus chemotherapy group and 18% in the placebo plus chemotherapy group. The most frequent immune-related AEs were hypothyroidism, dermatitis, and hyperthyroidism[31].

ONGOING TRIALS

In the context of these findings, ongoing trials continue to explore combination therapies aimed at improving treatment outcomes in advanced ESCC. Among the ongoing studies, we highlight a series of promising trials from a therapeutic perspective as they combine immunotherapy with chemotherapy alongside other molecules, potentially overcoming the primary resistance to current treatment regimens. The LEAP-014 (NCT04949256) is a phase 3 randomized trial assessing the efficacy and safety of pembrolizumab plus lenvatinib and chemotherapy compared with pembrolizumab plus chemotherapy alone.

Another multicenter phase II trial (NCT05013697) is investigating the combination of benmelstobart (a novel humanized anti-PD-L1 monoclonal antibody) with anlotinib (a multitargeted tyrosine kinase inhibitor) or placebo, in combination with paclitaxel and cisplatin as a first-line treatment for advanced ESCC. A phase II clinical study (NCT05322499) is evaluating the efficacy and safety of camrelizumab combined with chemotherapy or anlotinib in patients with advanced ESCC who have previously received first-line immunotherapy.

A two-arm, open-label, multicenter study (NCT05522894) is assessing the efficacy and safety of AK104 (anti-PD-1/CTLA-4 bispecific antibody) alone or combined with cisplatin and paclitaxel in treating advanced ESCC without prior systemic therapy. A phase I/II study (NCT06532799) is examining the safety and efficacy of autologous tumor-infiltrating lymphocyte therapy combined with pembrolizumab immunotherapy in patients with advanced or metastatic refractory gastric and EC. Lastly, the ANSWER study (NCT05214222) is an open-label, phase II trial investigating the combination of penpulimab (an anti-PD-1 antibody) and chemotherapy with or without anlotinib as a first-line treatment for advanced ESCC.

CONCLUSION

Choosing the optimal first-line treatment for metastatic ESCC is becoming increasingly complex due to the expanding range of therapeutic options. The decision-making process involves a careful evaluation of biomarker assessments, response rates, mechanisms of resistance, and the integration of next-generation ICIs. One of the current challenges is the variability in the PD-L1 scoring methods, TPS, CPS, and TAP. These scoring systems are not always concordant, making it difficult to consistently stratify patients and select the most effective therapy. Furthermore, response rates among patients who are positive for PD-L1 vary widely, reinforcing the need for improved biomarker-driven treatment approaches[15].

Resistance to PD-1/PD-L1 inhibitors remains a significant hurdle in ESCC treatment, requiring novel strategies to enhance immune responses. The inhibition of TIGIT, an emerging checkpoint target, has shown the potential to strengthen anti-tumor immunity by complementing the PD-L1/PD-1 axis. Early clinical data suggest that adding anti-TIGIT agents to existing regimens can lead to superior responses compared with both chemotherapies alone and traditional chemoimmunotherapy involving PD-L1 blockade[35].

Another key factor influencing treatment selection is the choice of chemotherapy backbone. There is no globally accepted standard regimen, and geographical preferences vary between platinum/fluoropyrimidine and platinum/taxane combinations. Preclinical evidence indicates that these regimens exert different immunomodulatory effects, potentially altering the efficacy of concurrent PD-1 inhibition[32]. Fluoropyrimidines influence dendritic cell maturation, while paclitaxel promotes the depletion of myeloid-derived suppressor cells, both of which may impact the immune response against tumors. However, direct comparisons between these chemotherapy backbones in the context of immunotherapy remain limited. Most clinical trials have allowed only one platinum doublet, with the notable exception of the ORIENT-15 study, where both paclitaxel and fluorouracil-based regimens were included. However, since 93% of patients in that trial received paclitaxel, it remains difficult to draw definitive conclusions about the comparative efficacy of these regimens[27].

Despite concerns about immune-related toxicities, the overall safety profile of chemoimmunotherapy appears to be comparable with chemotherapy alone in terms of SAEs. Across multiple studies, the incidence of AEs greater than grade 3 do not seem to be significantly higher in arms incorporating ICIs. This suggests that while checkpoint inhibitors introduce distinct toxicity profiles, they do not necessarily add to the overall burden of SAEs, if patients receive appropriate monitoring and management.

Given these complexities, selecting the most appropriate first-line treatment for metastatic ESCC requires a personalized approach that considers patient characteristics, biomarker status, resistance mechanisms, chemotherapy backbone, and safety concerns. The emergence of anti-TIGIT agents offers an exciting opportunity to improve patient outcomes by enhancing immune responses beyond PD-1/PD-L1 blockade. However, further studies are necessary to refine treatment strategies and establish the most effective therapeutic combinations in this rapidly evolving landscape. The ongoing clinical trials are essential to explore these innovative therapies and to provide more insight into their potential to overcome current treatment limitations and improve patient survival in metastatic ESCC.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Oncology

Country of origin: Italy

Peer-review report’s classification

Scientific Quality: Grade B, Grade C

Novelty: Grade A, Grade C

Creativity or Innovation: Grade A, Grade D

Scientific Significance: Grade A, Grade C

P-Reviewer: Wang RT; Zhou Y S-Editor: Wang JJ L-Editor: Filipodia P-Editor: Zhang XD

References
1.  Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, Jemal A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2024;74:229-263.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5690]  [Cited by in RCA: 6593]  [Article Influence: 6593.0]  [Reference Citation Analysis (1)]
2.  Rustgi AK, El-Serag HB. Esophageal carcinoma. N Engl J Med. 2014;371:2499-2509.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 817]  [Cited by in RCA: 989]  [Article Influence: 89.9]  [Reference Citation Analysis (0)]
3.  Abnet CC, Arnold M, Wei WQ. Epidemiology of Esophageal Squamous Cell Carcinoma. Gastroenterology. 2018;154:360-373.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 935]  [Cited by in RCA: 1135]  [Article Influence: 162.1]  [Reference Citation Analysis (1)]
4.  Liu X, Zhao S, Wang K, Zhou L, Jiang M, Gao Y, Yang R, Yan S, Zhang W, Lu B, Liu F, Zhao R, Liu W, Zhang Z, Liu K, Li X, Dong Z. Spatial transcriptomics analysis of esophageal squamous precancerous lesions and their progression to esophageal cancer. Nat Commun. 2023;14:4779.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 25]  [Reference Citation Analysis (0)]
5.  Li R, Li N, Yang Q, Tong X, Wang W, Li C, Zhao J, Jiang D, Huang H, Fang C, Xie K, Yuan J, Chen S, Li G, Luo H, Gao Z, Wu D, Cui X, Jiang W, Guo L, Ma H, Feng Y. Spatial transcriptome profiling identifies DTX3L and BST2 as key biomarkers in esophageal squamous cell carcinoma tumorigenesis. Genome Med. 2024;16:148.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
6.  Moehler M, Maderer A, Thuss-Patience PC, Brenner B, Meiler J, Ettrich TJ, Hofheinz RD, Al-Batran SE, Vogel A, Mueller L, Lutz MP, Lordick F, Alsina M, Borchert K, Greil R, Eisterer W, Schad A, Slotta-Huspenina J, Van Cutsem E, Lorenzen S. Cisplatin and 5-fluorouracil with or without epidermal growth factor receptor inhibition panitumumab for patients with non-resectable, advanced or metastatic oesophageal squamous cell cancer: a prospective, open-label, randomised phase III AIO/EORTC trial (POWER). Ann Oncol. 2020;31:228-235.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 30]  [Cited by in RCA: 70]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
7.  Kojima T, Shah MA, Muro K, Francois E, Adenis A, Hsu CH, Doi T, Moriwaki T, Kim SB, Lee SH, Bennouna J, Kato K, Shen L, Enzinger P, Qin SK, Ferreira P, Chen J, Girotto G, de la Fouchardiere C, Senellart H, Al-Rajabi R, Lordick F, Wang R, Suryawanshi S, Bhagia P, Kang SP, Metges JP; KEYNOTE-181 Investigators. Randomized Phase III KEYNOTE-181 Study of Pembrolizumab Versus Chemotherapy in Advanced Esophageal Cancer. J Clin Oncol. 2020;38:4138-4148.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 299]  [Cited by in RCA: 669]  [Article Influence: 133.8]  [Reference Citation Analysis (0)]
8.  Sun JM, Shen L, Shah MA, Enzinger P, Adenis A, Doi T, Kojima T, Metges JP, Li Z, Kim SB, Cho BC, Mansoor W, Li SH, Sunpaweravong P, Maqueda MA, Goekkurt E, Hara H, Antunes L, Fountzilas C, Tsuji A, Oliden VC, Liu Q, Shah S, Bhagia P, Kato K; KEYNOTE-590 Investigators. 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;398:759-771.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 310]  [Cited by in RCA: 857]  [Article Influence: 214.3]  [Reference Citation Analysis (0)]
9.  Kato K, Cho BC, Takahashi M, Okada M, Lin CY, Chin K, Kadowaki S, Ahn MJ, Hamamoto Y, Doki Y, Yen CC, Kubota Y, Kim SB, Hsu CH, Holtved E, Xynos I, Kodani M, Kitagawa Y. 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;20:1506-1517.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 428]  [Cited by in RCA: 780]  [Article Influence: 130.0]  [Reference Citation Analysis (0)]
10.  Shen L, Kato K, Kim SB, Ajani JA, Zhao K, He Z, Yu X, Shu Y, Luo Q, Wang J, Chen Z, Niu Z, Zhang L, Yi T, Sun JM, Chen J, Yu G, Lin CY, Hara H, Bi Q, Satoh T, Pazo-Cid R, Arkenau HT, Borg C, Lordick F, Li L, Ding N, Tao A, Shi J, Van Cutsem E; RATIONALE-302 Investigators. Tislelizumab Versus Chemotherapy as Second-Line Treatment for Advanced or Metastatic Esophageal Squamous Cell Carcinoma (RATIONALE-302): A Randomized Phase III Study. J Clin Oncol. 2022;40:3065-3076.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 70]  [Cited by in RCA: 162]  [Article Influence: 54.0]  [Reference Citation Analysis (0)]
11.  Luo H, Lu J, Bai Y, Mao T, Wang J, Fan Q, Zhang Y, Zhao K, Chen Z, Gao S, Li J, Fu Z, Gu K, Liu Z, Wu L, Zhang X, Feng J, Niu Z, Ba Y, Zhang H, Liu Y, Zhang L, Min X, Huang J, Cheng Y, Wang D, Shen Y, Yang Q, Zou J, Xu RH; ESCORT-1st Investigators. 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;326:916-925.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 137]  [Cited by in RCA: 438]  [Article Influence: 109.5]  [Reference Citation Analysis (0)]
12.  Huang J, Xu J, Chen Y, Zhuang W, Zhang Y, Chen Z, Chen J, Zhang H, Niu Z, Fan Q, Lin L, Gu K, Liu Y, Ba Y, Miao Z, Jiang X, Zeng M, Chen J, Fu Z, Gan L, Wang J, Zhan X, Liu T, Li Z, Shen L, Shu Y, Zhang T, Yang Q, Zou J; ESCORT Study Group. Camrelizumab versus investigator's choice of chemotherapy as second-line therapy for advanced or metastatic oesophageal squamous cell carcinoma (ESCORT): a multicentre, randomised, open-label, phase 3 study. Lancet Oncol. 2020;21:832-842.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 170]  [Cited by in RCA: 404]  [Article Influence: 80.8]  [Reference Citation Analysis (0)]
13.  Yap DWT, Leone AG, Wong NZH, Zhao JJ, Tey JCS, Sundar R, Pietrantonio F. Effectiveness of Immune Checkpoint Inhibitors in Patients With Advanced Esophageal Squamous Cell Carcinoma: A Meta-analysis Including Low PD-L1 Subgroups. JAMA Oncol. 2023;9:215-224.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 15]  [Cited by in RCA: 44]  [Article Influence: 22.0]  [Reference Citation Analysis (0)]
14.  Smyth EC, Gambardella V, Cervantes A, Fleitas T. Checkpoint inhibitors for gastroesophageal cancers: dissecting heterogeneity to better understand their role in first-line and adjuvant therapy. Ann Oncol. 2021;32:590-599.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 51]  [Cited by in RCA: 94]  [Article Influence: 23.5]  [Reference Citation Analysis (0)]
15.  Klempner SJ, Cowden ES, Cytryn SL, Fassan M, Kawakami H, Shimada H, Tang LH, Wagner DC, Yatabe Y, Savchenko A, Salcius J, Johng D, Chen J, Montenegro G, Moehler M. PD-L1 Immunohistochemistry in Gastric Cancer: Comparison of Combined Positive Score and Tumor Area Positivity Across 28-8, 22C3, and SP263 Assays. JCO Precis Oncol. 2024;8:e2400230.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 6]  [Cited by in RCA: 8]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
16.  Kelly RJ, Zaidi AH, Smith MA, Omstead AN, Kosovec JE, Matsui D, Martin SA, DiCarlo C, Werts ED, Silverman JF, Wang DH, Jobe BA. The Dynamic and Transient Immune Microenvironment in Locally Advanced Esophageal Adenocarcinoma Post Chemoradiation. Ann Surg. 2018;268:992-999.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 48]  [Cited by in RCA: 84]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
17.  Wen J, Fang S, Hu Y, Xi M, Weng Z, Pan C, Luo K, Ling Y, Lai R, Xie X, Lin X, Lin T, Chen J, Liu Q, Fu J, Yang H. Impacts of neoadjuvant chemoradiotherapy on the immune landscape of esophageal squamous cell carcinoma. EBioMedicine. 2022;86:104371.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 23]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
18.  Wang X, He J, Li J, Wu C, Yue M, Niu S, Jia Y, Jia Z, Cai L, Liu Y. Concordance of assessments of four PD-L1 immunohistochemical assays in esophageal squamous cell carcinoma (ESCC). J Cancer Res Clin Oncol. 2024;150:43.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 7]  [Reference Citation Analysis (0)]
19.  Liu F, Tian T, Xia LL, Ding Y, Cormier RT, He Y. Circulating miRNAs as novel potential biomarkers for esophageal squamous cell carcinoma diagnosis: a meta-analysis update. Dis Esophagus. 2017;30:1-9.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 6]  [Cited by in RCA: 11]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
20.  Zhou J, Liu S, Zhang J, Zeng Q, Lin Z, Fu R, Lin Y, Hu Z. Discovery and validation of Hsa-microRNA-3665 promoter methylation as a potential biomarker for the prognosis of esophageal squaous cell carcinoma. Int J Clin Oncol. 2025;30:309-319.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
21.  Zheng L, Li L, Xie J, Jin H, Zhu N. Six Novel Biomarkers for Diagnosis and Prognosis of Esophageal squamous cell carcinoma: validated by scRNA-seq and qPCR. J Cancer. 2021;12:899-911.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 10]  [Cited by in RCA: 41]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
22.  Li Z, Chen X, Li Y, Xu Y, Zhou Y. Aberrant DNA Methylation in Esophageal Squamous Cell Carcinoma and its Clinical Implications in Systemic Chemotherapy. Int J Med Sci. 2025;22:1002-1014.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
23.  Doki Y, Ajani JA, Kato K, Xu J, Wyrwicz L, Motoyama S, Ogata T, Kawakami H, Hsu CH, Adenis A, El Hajbi F, Di Bartolomeo M, Braghiroli MI, Holtved E, Ostoich SA, Kim HR, Ueno M, Mansoor W, Yang WC, Liu T, Bridgewater J, Makino T, Xynos I, Liu X, Lei M, Kondo K, Patel A, Gricar J, Chau I, Kitagawa Y; CheckMate 648 Trial Investigators. Nivolumab Combination Therapy in Advanced Esophageal Squamous-Cell Carcinoma. N Engl J Med. 2022;386:449-462.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 174]  [Cited by in RCA: 609]  [Article Influence: 203.0]  [Reference Citation Analysis (2)]
24.  Luo H, Lu J, Bai Y, Mao T, Wang J, Fan Q, Zhang Y, Zhao K, Chen Z, Gao S, Li J, Fu Z, Gu K, Liu Z, Wu L, Zhang X, Feng J, Sheng Z, Zeng W, Xu R. Final analysis of the randomized phase 3 ESCORT-1st trial: Camrelizumab plus chemotherapy as first-line treatment for advanced or metastatic esophageal squamous cell carcinoma. J Clin Oncol. 2024;42:4055.  [PubMed]  [DOI]  [Full Text]
25.  Wang ZX, Cui C, Yao J, Zhang Y, Li M, Feng J, Yang S, Fan Y, Shi J, Zhang X, Shen L, Shu Y, Wang C, Dai T, Mao T, Chen L, Guo Z, Liu B, Pan H, Cang S, Jiang Y, Wang J, Ye M, Chen Z, Jiang D, Lin Q, Ren W, Wang J, Wu L, Xu Y, Miao Z, Sun M, Xie C, Liu Y, Wang Q, Zhao L, Li Q, Huang C, Jiang K, Yang K, Li D, Liu Y, Zhu Z, Chen R, Jia L, Li W, Liao W, Liu HX, Ma D, Ma J, Qin Y, Shi Z, Wei Q, Xiao K, Zhang Y, Zhang Y, Chen X, Dai G, He J, Li J, Li G, Liu Y, Liu Z, Yuan X, Zhang J, Fu Z, He Y, Ju F, Liu Z, Tang P, Wang T, Wang W, Zhang J, Luo X, Tang X, May R, Feng H, Yao S, Keegan P, Xu RH, Wang F. Toripalimab plus chemotherapy in treatment-naïve, advanced esophageal squamous cell carcinoma (JUPITER-06): A multi-center phase 3 trial. Cancer Cell. 2022;40:277-288.e3.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 293]  [Cited by in RCA: 251]  [Article Influence: 83.7]  [Reference Citation Analysis (0)]
26.  Xu J, Kato K, Raymond E, Hubner RA, Shu Y, Pan Y, Park SR, Ping L, Jiang Y, Zhang J, Wu X, Yao Y, Shen L, Kojima T, Gotovkin E, Ishihara R, Wyrwicz L, Van Cutsem E, Jimenez-Fonseca P, Lin CY, Wang L, Shi J, Li L, Yoon HH. 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;24:483-495.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 38]  [Cited by in RCA: 114]  [Article Influence: 57.0]  [Reference Citation Analysis (0)]
27.  Lu Z, Wang J, Shu Y, Liu L, Kong L, Yang L, Wang B, Sun G, Ji Y, Cao G, Liu H, Cui T, Li N, Qiu W, Li G, Hou X, Luo H, Xue L, Zhang Y, Yue W, Liu Z, Wang X, Gao S, Pan Y, Galais MP, Zaanan A, Ma Z, Li H, Wang Y, Shen L; ORIENT-15 study group. 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.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 58]  [Cited by in RCA: 204]  [Article Influence: 68.0]  [Reference Citation Analysis (0)]
28.  Song Y, Zhang B, Xin D, Kou X, Tan Z, Zhang S, Sun M, Zhou J, Fan M, Zhang M, Song Y, Li S, Yuan Y, Zhuang W, Zhang J, Zhang L, Jiang H, Gu K, Ye H, Ke Y, Li J, Wang Q, Zhu J, Huang J; ASTRUM-007 investigators. First-line serplulimab or placebo plus chemotherapy in PD-L1-positive esophageal squamous cell carcinoma: a randomized, double-blind phase 3 trial. Nat Med. 2023;29:473-482.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 36]  [Cited by in RCA: 90]  [Article Influence: 45.0]  [Reference Citation Analysis (0)]
29.  Hsu C, Lu Z, Gao S, Wang J, Sun J, Liu T, Fan Q, Cai J, Ge F, Li S, Zhang L, Cha E, Shen L. SKYSCRAPER-08: A phase III, randomized, double-blind, placebo-controlled study of first-line (1L) tiragolumab (tira) + atezolizumab (atezo) and chemotherapy (CT) in patients (pts) with esophageal squamous cell carcinoma (ESCC). J Clin Oncol. 2024;42:245.  [PubMed]  [DOI]  [Full Text]
30.  Sun J, Chao Y, Kim S, Rha SY, Evans TJ, Strickland A, Wainberg ZA, Chau I, Pelles-avraham S, Ajani JA, Malhotra R, Liu Q, Li S, Cha E, Rahalkar S, Allen S, Hsu C. MORPHEUS-EC: A phase Ib/II open-label, randomized study of first-line tiragolumab (tira) + atezolizumab (atezo) + chemotherapy (CT) in patients (pts) with esophageal cancer (EC). J Clin Oncol. 2024;42:324.  [PubMed]  [DOI]  [Full Text]
31.  Galluzzi L, Humeau J, Buqué A, Zitvogel L, Kroemer G. Immunostimulation with chemotherapy in the era of immune checkpoint inhibitors. Nat Rev Clin Oncol. 2020;17:725-741.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 285]  [Cited by in RCA: 847]  [Article Influence: 169.4]  [Reference Citation Analysis (0)]
32.  Xu K, Yu M, Sun Q, Zhang L, Qian X, Su D, Gong J, Shang J, Lin Y, Li X. Cost-effectiveness of PD-1 inhibitors combined with chemotherapy for first-line treatment of oesophageal squamous cell carcinoma in China: a comprehensive analysis. Ann Med. 2025;57:2482019.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
33.  Liu S, Dou L, Li S. Cost-effectiveness analysis of PD-1 inhibitors combined with chemotherapy as first-line therapy for advanced esophageal squamous-cell carcinoma in China. Front Pharmacol. 2023;14:1055727.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 10]  [Cited by in RCA: 15]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
34.  Zhang P, Liu X, Gu Z, Jiang Z, Zhao S, Song Y, Yu J. Targeting TIGIT for cancer immunotherapy: recent advances and future directions. Biomark Res. 2024;12:7.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 32]  [Reference Citation Analysis (0)]
35.  Wu M, Huang Q, Xie Y, Wu X, Ma H, Zhang Y, Xia Y. Improvement of the anticancer efficacy of PD-1/PD-L1 blockade via combination therapy and PD-L1 regulation. J Hematol Oncol. 2022;15:24.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 49]  [Cited by in RCA: 256]  [Article Influence: 85.3]  [Reference Citation Analysis (0)]