Review Open Access
Copyright ©The Author(s) 2023. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Sep 14, 2023; 29(34): 5020-5037
Published online Sep 14, 2023. doi: 10.3748/wjg.v29.i34.5020
Perioperative immunotherapy for esophageal squamous cell carcinoma: Now and future
Yong Liu, Department of Thoracic Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430011, Hubei Province, China
ORCID number: Yong Liu (0000-0002-4048-5837).
Author contributions: Liu Y designed the paper, collected and analyzed the data in the references, and wrote the manuscript.
Conflict-of-interest statement: The author declares no conflicts of interest related to 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: Yong Liu, Doctor, MD, Chief Doctor, Surgeon, Surgical Oncologist, Department of Thoracic Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, No. 26 Shengli Street, Wuhan 430011, Hubei Province, China. liuyong7575@163.com
Received: April 23, 2023
Peer-review started: April 23, 2023
First decision: July 9, 2023
Revised: July 19, 2023
Accepted: August 15, 2023
Article in press: August 15, 2023
Published online: September 14, 2023

Abstract

Esophageal cancer (EC) ranks among the most prevalent malignant tumors affecting the digestive tract. Esophageal squamous cell carcinoma (ESCC) stands as the prevailing pathological subtype, encompassing approximately 90% of all EC patients. In clinical stage II-IVA locally advanced ESCC cases, the primary approach to treatment involves a combination of neoadjuvant therapy and surgical resection. Despite concerted efforts, the long-term outcomes for ESCC patients remain unsatisfactory, with dismal prognoses. However, recent years have witnessed remarkable strides in immunotherapy, particularly in the second- and first-line treatment of advanced or metastatic ESCC, with the development of monoclonal antibodies that inhibit programmed death 1 or programmed death ligand 1 demonstrating encouraging responses and perioperative clinical benefits for various malignancies, including ESCC. This comprehensive review aims to present the current landscape of perioperative immunotherapy for resectable ESCC, focusing specifically on the role of immune checkpoint inhibitors during the perioperative period. Additionally, the review will explore promising biomarkers and offer insights into future prospects.

Key Words: Esophageal squamous cell carcinoma, Immune checkpoint inhibitors, Immunotherapy, Neoadjuvant, Randomized clinical trial

Core Tip: Esophageal cancer ranks among the most prevalent malignant tumors affecting the digestive tract. In locally advanced esophageal squamous cell carcinoma (ESCC), the mainstay of treatment involves neoadjuvant therapy in conjunction with surgical resection. Notably, immunotherapy has achieved significant breakthroughs in the second- and first-line treatment of advanced or metastatic ESCC. This review focuses on the current landscape of perioperative immunotherapy for resectable ESCC and discusses promising biomarkers and future perspectives.



INTRODUCTION

Esophageal cancer (EC), one of the most prevalent malignancies affecting the digestive tract, originates from the epithelial lining of the esophagus. According to the International Agency for Research on Cancer's latest update on the global cancer burden, based on the GLOBOCAN 2020 projections of cancer incidence and mortality, EC ranks seventh in terms of cancer incidence worldwide and sixth in cancer-related mortality[1]. The incidence of EC varies significantly across countries and regions, with East Asia having the highest disease occurrence, being twice the global average (12.2/1000). EC can be classified into two histological subtypes: Esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma (EAC). EAC is the predominant pathological type in relatively low-incidence areas such as Europe and America. However, globally, ESCC accounts for approximately 90% of all EC cases[2], and more than half of the ESCC cases occur in China[3].

For many years, surgery has served as the primary treatment for early-stage ESCC. High-grade dysplasia and very early-stage tumors are amenable to local therapies such as endoscopic resection, ablation, or surgery. Surgery can improve the 5-year survival rate to 60%-85% in patients with early-stage disease[4]. However, due to the subtle nature of early symptoms, many patients present with advanced or metastatic disease at the time of diagnosis. For locally advanced disease, surgery alone has not yielded satisfactory results, with a median survival time of 12 to 18 mo and a 5-year survival rate of 15%-39%[5,6]. Furthermore, local or systemic recurrence is common, with recurrence reported in 35%-50% of patients who had underwent surgery alone[7].

Neoadjuvant therapy combined with surgery stands as a cornerstone in the treatment of locally advanced ESCC, typically encompassing clinical stages II to IVA[8]. The medical community widely acknowledges the value of neoadjuvant therapy due to its efficiency compared to postoperative adjuvant therapy. Moreover, it leads to reductions in tumor and lymph node (LN) volumes, improves the R0 resection rate, and enhances long-term survival rates[9-11]. Additionally, neoadjuvant therapy allows for the evaluation of patient response using resected specimens[12]. Numerous randomized controlled trials (RCTs) have indicated that preoperative or neoadjuvant chemoradiotherapy (nCRT) may result in longer overall survival (OS) compared to surgery alone[13-15]. The CROSS trial established nCRT as the first-line therapeutic choice for resectable locally advanced ESCC, combining radiotherapy with a chemotherapy regimen containing carboplatin and paclitaxel[16]. The NEOCRTEC5010 phase III, multi-center, open-label RCT confirmed the findings of the CROSS trial for ESCC[9]. However, the optimal neoadjuvant therapy for resectable locally advanced ESCC remains a topic of debate. Neoadjuvant chemotherapy showed improved OS over surgery alone only for EAC, whereas nCRT demonstrated considerably better OS than surgery alone for both EAC and ESCC, according to the NewEC study[17]. Several subsequent meta-analyses also supported the utility of preoperative nCRT, showing improved OS compared to other treatment modalities, including surgery alone, neoadjuvant chemotherapy, and neoadjuvant radiotherapy, albeit with increased postoperative mortality[18,19]. Postoperative morbidity was similar between the nCRT and S (surgery) groups (55.6% vs 52.8%; P = 0.720), while in-hospital postoperative mortality was significantly higher in the CRT group (11.1% vs 3.4%; P = 0.049)[20]. A Japanese study[21] examined late complications, revealing grade 2 anastomotic stricture as the most common event, occurring in 30% of the 33 patients. A total of 12 events of grade 3 or worse complications were observed in ten patients, including gastric tube ulcer, cardiac complications, and pulmonary complications. The 5-year incidence rate was 22%, and three patients succumbed to the late complications. A clear advantage of nCRT over neoadjuvant chemotherapy was not established.

Presently, both the National Comprehensive Cancer Network and the Chinese Society of Clinical Oncology guidelines recommend chemoradiotherapy (CRT) as the standard approach for locally advanced ESCC[22-24]. Despite significant efforts made by the medical community, the expected long-term outcomes for ESCC patients have seen limited improvement, remaining poor. The 5-year OS in patients undergoing nCRT and surgery is approximately 50%, and the incidence of local recurrence or distant metastasis remains high[25]. Relapse after nCRT is common and constitutes a major hurdle to overcome[26].

Immunotherapy is a therapeutic approach that involves the use of substances to stimulate or suppress the immune system, aiding the body in combating cancer, infections, and other diseases. It encompasses biologic/targeted agents that aim to enhance and restore the immune system's ability to recognize and eliminate cancer cells by modifying and/or blocking costimulatory signals[27,28]. Over the past few years, immunotherapy has achieved remarkable progress in cancer treatment, particularly with the advent of immune checkpoint inhibitors (ICIs)[29]. The development of ICIs, which inhibit programmed death 1 (PD-1) or programmed death ligand 1 (PD-L1), has shown convincing responses and clinical benefits across various malignancies, including ESCC[30,31].

In the study "KEYNOTE-181", Pembrolizumab demonstrated superior OS, a higher objective response rate, and a lower incidence of grades 3-5 treatment-related adverse events (trAEs) compared to chemotherapy in the second-line setting[32]. Grades 3-5 trAEs occurred in 18.2% of patients treated with Pembrolizumab vs 40.9% in the chemotherapy group. Other trials such as "RATIONALE-302"[33], "ATTRACTION-3"[34], and "ESCORT"[35] have also reported positive outcomes. In these trials, immunotherapy showed lower rates of grades 3-5 trAEs compared to chemotherapy, with percentages of 18.8% vs 55.8% in "RATIONALE-302"[33], 18% vs 63% in "ATTRACTION-3"[34], and 19% vs 39% in "ESCORT"[35].

It is important to note that in studies like "JUPITER-06"[36], "CheckMate-648"[37], "ORIENT-15"[38], "ESCORT-1st"[39], and "KEYNOTE-590"[40], treating patients with advanced EC using PD-1 inhibitors in combination with chemotherapy as first-line therapy resulted in significantly longer OS and progression-free survival (PFS) compared to chemotherapy alone. The occurrence rates of grades 3-5 trAEs in these trials were relatively comparable.

Neoadjuvant immunotherapy has been explored in various other malignancies, such as lung cancer[41,42], melanoma[43], bladder cancer[44], colon cancer[45], and glioblastoma[46]. In a clinical trial (NCT02259621) investigating neoadjuvant Nivolumab, surgery was not delayed, and 45% of resected tumors showed a major pathological response (MPR). In the NADIM trial[42], patients were treated with a neoadjuvant regimen consisting of paclitaxel and carboplatin in combination with Nivolumab. Out of the initial 51 patients deemed eligible, 46 patients received neoadjuvant treatment and subsequently underwent surgery. At the 24-mo mark, the PFS rate was observed to be 77.1%. Among the 27 patients who had melanoma, eight experienced either a complete response or an MPR after receiving a single dose of the anti-PD-1 drug, Pembrolizumab. Importantly, all eight of these patients remained free from the disease[43]. In a single-arm phase II study exploring Atezolizumab before cystectomy in 95 patients with muscle-invasive urothelial cancer (NCT02662309), the pathological complete response (pCR) rate was 31%[44]. In the exploratory NICHE study (NCT03026140)[45], patients with early-stage colon cancers, categorized as mismatch repair-deficient (dMMR) or mismatch repair-proficient (pMMR) tumors, received a single dose of Ipilimumab and two doses of Nivolumab before surgery. Among dMMR tumors, 20 out of 20 displayed a pathological response, with 19 cases of MPR and 12 cases of pCR. In pMMR tumors, 4 out of 15 showed pathological responses, with three cases of MPR and one case of partial response. In a single-arm phase II clinical trial (NCT02550249)[46], a presurgical dose of Nivolumab followed by postsurgical Nivolumab until disease progression or unacceptable toxicity was tested in 30 patients. However, no significant clinical benefit was observed following salvage surgery, although two out of the three patients treated with Nivolumab before and after primary surgery remained alive 33 and 28 mo later.

Based on this background, this review aims to depict the current scenario in the field of perioperative immunotherapy for resectable ESCC, focusing in particular on an overview of the role of ICIs in this field, alongside a discussion of the promising biomarkers and forecast of future perspectives.

PERIOPERATIVE IMMUNOTHERAPY OF ESCC
Drugs for immunotherapy of ESCC

Cancer cells have the ability to evade immune surveillance by disrupting the balance of the tumor microenvironment (TME). This disruption can lead to tumor development by blocking apoptosis, promoting angiogenesis, proliferation, and distant metastasis, and evading immune detection[47]. CD8(+) T cells, also known as cytotoxic T lymphocytes, play a crucial role in killing tumor cells, and their presence in the TME is associated with improved cancer prognosis[48]. For CD8(+) T cells to effectively kill tumor cells, two signals are essential: The recognition of antigens presented by major histocompatibility complexes[49] and the stimulation or suppression of T cell activation. The second signal, acting as a co-stimulatory or co-inhibitory signal, is often referred to as an "immune checkpoint" for CD8(+) T cell function. Immune checkpoints help maintain the balance of T-cell activation, immune tolerance, and immune-mediated tissue damage[50]. Both co-stimulatory and co-inhibitory ligands and their receptors are present on T cells and antigen-presenting cells. Inhibitory checkpoint molecules displayed by CD8(+) T cells can respond to and aid the tumor in evading the immune system[51].

There are four main ICIs named after the corresponding Food and Drug Administration (FDA)-approved monoclonal antibody therapies: Cytotoxic T lymphocyte antigen-4 (CTLA-4), PD-1, PD-L1/L2, and lymphocyte activation gene-3 (LAG-3)[52]. CTLA-4 and PD-1 are members of the CD28 receptor family expressed on T-cells, which bind to their corresponding targets of the B7 family[53]. CTLA-4 is an intracellular protein often found on regulatory T cells, inhibiting CD8(+) T cell activity. Ipilimumab, approved by the FDA in 2011, targets CTLA-4[54]. PD-1 is a transmembrane protein upregulated by repeated stimulation of T cells. PD-1 has two ligands, PD-L1 and PD-L2, which are cell surface proteins expressed on tumor cells and some immune cells within the TME. When PD-1 binds to these ligands, T cell function is inhibited. Overexpression of PD-L1 is associated with tumor progression, as cancer cells exploit the PD-1/PD-L1 and PD-1/PD-L2 pathways to create an immunosuppressive environment[55]. Nivolumab and Pembrolizumab are ICIs that target the PD-1 molecule, both FDA-approved in 2014. In 2018 and 2021, Cemiplimab and Dostarlimab were also approved, respectively[56,57]. Atezolizumab, Avelumab, and Durvalumab are FDA-approved PD-L1 inhibitors[58-60]. LAG-3 is a transmembrane receptor expressed on CD8(+) T cells, further upregulated by T cell activation. Relatlimab is the only FDA-approved LAG-3 inhibitor[61].

In recent years, several domestic anti-PD-1 monoclonal antibodies have been approved as drugs for various tumors by the China National Medical Product Administration. Notably, Camrelizumab, Sintilimab, Toripalimab, and Tislelizumab are representative drugs that are currently undergoing in-depth research. Camrelizumab, a humanized anti-PD-1 monoclonal antibody developed in China, has shown promising activity and manageable toxicity when combined with Apatinib, an anti-angiogenic drug. This combination may serve as a potential second-line treatment option for patients with advanced ESCC[62] or patients with recurrent or metastatic ESCC[63].

Neoadjuvant immunotherapy of ESCC

As previously mentioned, ICI immunotherapy has demonstrated improved outcomes in terms of OS and disease-free survival (DFS) for both second-line and first-line treatments of ESCC. In the context of resectable locally advanced ESCC, there has been a growing interest in perioperative immunotherapy combined with chemotherapy or radiotherapy, representing a key research direction for this disease. Preoperative neoadjuvant immunotherapy aims to activate the patient's immune system, leading to the formation of immune memory cells, thereby enabling the immune system to assume an immune surveillance role[64,65]. This review will summarize the current status of neoadjuvant immunotherapy as a treatment for ESCC.

Completed and reported clinical studies of neoadjuvant immunotherapy

Numerous clinical studies have investigated the efficacy and safety of immunotherapy for resectable ESCC in the neoadjuvant setting, as detailed in Table 1. Most neoadjuvant immunotherapy trials are conducted in conjunction with chemotherapy or CRT. Given the high incidence of this disease in China, numerous clinical trials on this subject are carried out in the country. Specifically, six ICIs targeting PD-1/PD-L1, namely, Pembrolizumab (4 studies), Camrelizumab (10 studies), Sintilimab (4 studies), Toripalimab (5 studies), and Tislelizumab (1 study), have been studied as neoadjuvant therapy. Additionally, there is an ongoing drug-based neoadjuvant therapy RCT involving Nivolumab, the first ICI used in adjuvant immunotherapy in the CheckMate-577 study[66]. The results of this trial, known as the FRONTiER study (NCT03914443), are eagerly anticipated[67]. In Table 1, four reports were retrospective in nature, while the other 21 reports were prospective, with 20 of them being single-arm trials. Only three studies employed neoadjuvant immunotherapy in combination with concurrent CRT as an intervention. In studies involving neoadjuvant immunotherapy combined with chemotherapy, the chemotherapy drugs used included 5-fluorouracil (5-FU), cisplatin (DDP), carboplatin (CBP), nedaplatin (NDP), paclitaxel (PTX), docetaxel (DTX), albumin-bound paclitaxel (nab-PTX), and PTX liposomes. Over the past few years, the use and dosages of these chemotherapy drugs have demonstrated efficacy for ESCC treatment in clinical settings. Most surgeons have chosen an interval time of 4-6 wk from the end of neoadjuvant therapy to surgery. The primary outcomes of utmost concern to doctors are safety, feasibility, and pCR, while MPR and R0 resection rate are commonly selected as primary or secondary outcomes.

Table 1 Reported clinical results of neoadjuvant immunotherapy for resectable esophageal squamous cell carcinoma.
Drug & Ref.Trial No.PhaseNumber of enrollmentsClinical stageDesignChemotherapy drugsChemotherapy cyclesRadiotherapyInterval time to surgeryPrimary outcomeSafety (rate of grade ≥ 3 trAEs)Feasibility (therapy completion rate)pCRMPRSurgical outcome (R0 rate)
Pembrolizumab[111]Park et al[111] (NR)NA38IA-IVARetrospective5-FU + DDP/PTX + CBPNA41.4 Gy/23 f6-8 wkOperative risk18.75% (3/16)100% (16/16)NANA100%
Pembrolizumab[112]Huang et al[112] (NR)NA54II–IVARetrospectiveDTX + NDP2, Q3W4-6 wkpCR13.04% (4/23)NA30.4% (7/23) NA100%
Pembrolizumab[113]NCT02844075 II28IB-IIISingle- armPTX + CBP5, Q1W41.4 Gy/23 f 5 wkpCRNA92.9% (26/28)46.1% (12/26)NANA
Pembrolizumab[80]Keystone-001/NCT04389177II50IIIA-IIIBSingle-armPTX + DDP3, Q3W4-6 wkpCR, MPR, safety069.0% (29/42)41.4% (12/29) 72.4% (21/29)100%
Camrelizumab[114]Qiao et al[114] (NR)NA254IA-IVARetrospectivePTX, nab-PTX/DTX2, Q3WNApCR6.25% (3/48)NA41.7% (20/48) 60.4 (29/48)NA
Camrelizumab[74]ChiCTR1900026240II60IIIA-IVASingle-armnab-PTX + CBP2, Q3W4-6 wkpCR 56.7% (34/60)85.0% (51/60)39.2% (20/51)NA98.0%
Camrelizumab[115]NCT04506138 I-II46II-IVASingle-armnab-PTX + CBP4-6 wkpCR15.2% (7/46)82.6% (38/46)21.6% (8/37)48.6% (18/37)80.4%
Camrelizumab[82]GASTO1056/ChiCTR2000028900II23II-IIISingle-armnab-PTX + CBP2, Q3W3-6 wkSafety47.8%87.0% (20/23)25% (5/20)50% (10/20)100%
Camrelizumab[116]Yang et al[116] (NR)II12II-IIISingle-armnab-PTX + S1 3, Q3W3-6 wkpCR075.0% (9/12)33.3% (4/12)41.7% (5/12)100%
Camrelizumab[75]NIC-ESCC2019/NCT04225364II56II-IVASingle-armnab-PTX + DDP2, Q3W6 wkpCR10.7% (6/51)91.1% (51/56)35.3% (18/51)23.5% (12/21)100%
Camrelizumab[117]ChiCTR1900023880Ib30IC-IIIBSingle-armnab-PTX + NDP + Apatinib2-4, Q3W4-8 wkSafety, feasibility36.7% (11/30)96.7% (29/30)24.1% (7/29)51.7% (15/29)NA
Camrelizumab[118]ESPRIT/ChiCTR2000033761II48IIA-IIIBSingle-armPTX + NDP2-4, Q3WNApCR4.2% 62.5% (30/48)35.0% (7/20)NANA
Camrelizumab[119]NCT 03917966II40IC-IVASingle-armDTX + NDP2, Q3W4-6 wkMPR3%70.6% (12/17)25.0% (3/12)41.6% (5/12)100%
Camrelizumab[120] ChiCTR2000039170II166Locally advancedSingle-armnab-PTX + NDP NANASafety7.8% (13/166)49.4% (82/166) 18.5% (15/81)63.0% (51/81)97.5% (79/82)
Sintilimab[121]ChiCTR1900026593II47II-IVASingle-armPTX liposome + CBP2, Q3W3-6 wkpCR29.8% (14/47)95.7% (45/47)22.2% (10/45)44.4% (20/45)97.8% (44/45)
Sintilimab[81]SIN-ICE study/ChiCTR2100048917II23IC-IVASingle-armPlatinum3, Q3W4-6 wkpCR, safety30.4% (7/23)73.9% (17/23)35.3%, (6/17) 52.9% (9/17)94.1% (16/17)
Sintilimab[73]ESONICT-1/ChiCTR2100045659II30IIB-IVASingle-armDDP + nab-PTX2, Q3W4-6 wkpCR, safety3.3% (1/30)76.6% (23/30)21.7% (5/23)52.2% (12/23)100%
Sintilimab[88]KEEP-G03/NCT03946969II30IB-IVASingle-armPTX liposom + DDP + S12, Q3WWithin 6 wkSafety, feasibility36.7%100.0% (30/30)20% (6/30)50% (15/30)100%
Toripalimab[122,123]NCT03985670II30II-IVATwo-armPTX + DDP2, Q3-4W4-6 wkpCR8.33% (2/24)80.0% (24/30)20.8% (5/24)NA100%
Toripalimab[124]ChiCTR1900025318II23IIB-IVASingle-armPTX + DDP2, Q3W4-6 wkpCR, R0 rate8.70% (2/23)78.3% (18/23)33.3% (6/18)NA100%
Toripalimab[77]NCT04177797II 20IIIA-IVASingle-armPTX + CBP2, Q3W4-6 wkSafety, feasibility, MPR, pCR20.0% (4/20)80.0% (16/20)18.8% (3/16)43.8% (7/16)87.5% (14/16)
Toripalimab[125]ESONICT-2/ChiCTR2100052784II20IIB-IVASingle-armDTX + DDP2, Q3W4-6 wkpCR, safety15.0% (3/20) 60.0% (12/20)16.7% (2/12)41.7% (5/12)100%
Toripalimab[126]SCALE-1/ChiCTR2100045104Ib20IIB-IVASingle-armPTX + CBP2, Q3W30 Gy/12 f 4-7 wkSafetyNA87.0% (20/23)55% (11/20)80% (16/20)
Tislelizumab[76]TD-NICE/ChiCTR2000037488II45IIIA-IVASingle-armCBP + nab-PTX2, Q3W3-6 wkMPR33.3% (15/45)80.0% (36/45)50% (18/36)72% (26/36)97.2% (35/36)
Multiple[127]CHICTR210004565927IC-IVARetrospectively, two-armPlatinum+ PTX or platinum + 5-Fu2, Q3w4–8 wk30-d major complications11.1% (3/27)NANANA100%

Ensuring the safety of participants is of utmost importance in clinical trials, and the occurrence of trAEs is a key indicator in assessing the safety of neoadjuvant immunotherapy[68]. When ICIs are combined with other anti-cancer therapies, such as chemotherapy, immunotherapy, targeted therapy, or radiotherapy, the incidence of trAEs is significantly higher compared to the use of ICIs alone[69]. These trAEs can affect various organs, with the most common being endocrine (hypothyroidism and hyperthyroidism), gastrointestinal (diarrhea and colitis), pulmonary (pneumonitis), dermatological (rash and pruritus), and hepatic (elevated liver enzymes) complications[70,71]. The majority of trAEs (grades 1-2) are self-limiting or can be managed with immunosuppressive therapy, such as corticosteroids. However, persistent trAEs that do not respond to corticosteroids require close monitoring and appropriate treatment. Fatal trAEs are extremely rare for anti-PD-1 antibodies, with an incidence of less than 0.5% in a meta-analysis of ICI monotherapy studies across various cancer types, most commonly associated with pneumonitis[71]. The incidence of serious trAEs (grade ≥ 3) in neoadjuvant ICI plus chemotherapy ranged from 0% to 36.7% in the studies presented in Table 1. Notably, the combination of chemotherapy with Camrelizumab and Apatinib resulted in the highest incidence of patients experiencing serious trAEs (36.7%). In another meta-analysis comparing the efficacy and safety of various ICIs for patients with advanced or metastatic ESCC, Camrelizumab and Nivolumab were found to have a lower incidence of serious trAEs in the first-line and refractory settings, respectively[72]. Close monitoring and early recognition of relevant symptoms and signs are essential to ensure appropriate management.

Feasibility can be assessed by comparing the completion rate of the trial. In a pooled analysis, the rates of completion of neoadjuvant therapy and surgery ranged from 49.4% (Camrelizumab + nab-PTX + NDP) to 100% (Sintilimab + PTX liposome, DDP, and S-1). Failures in the study were mainly attributed to trAEs[73,74], patient decisions[75-77], or disease progression[78]. As most trials achieved a treatment completion rate of over 60%, interpreting the data in the conference abstract (ChiCTR2000039170) is challenging.

The definition of pCR is the absence of any signs of cancer on a histological resection specimen. The pCR rate is a crucial efficacy-related parameter reported in all studies, with some studies choosing it as a secondary outcome. In a meta-analysis of seven clinical trials involving 815 patients, the pooled pCR rate was 32.4% (95% confidence interval [CI]: 28.2%-36.8%)[79]. Data from Table 1 shows that the pCR rate ranges from 6.7% to 46.1%. The definition of MPR is the presence of less than 10% of the remaining viable tumor cells in the resected primary tumor. In the aforementioned meta-analysis, the pooled MPR rate was 49.4% (95%CI: 42.1%-56.7%)[79]. MPR rate was chosen as the primary outcome in four studies and seemed to be a second primary outcome in the main studies verified in Table 1, with reported rates ranging from 42% to 72.3%[73,76,77,80-83]. The R0 resection rate is defined as a complete resection of the tumor with a negative microscopic edge, indicating no residual tumor. It is another crucial indicator to evaluate the effectiveness of neoadjuvant therapy. The R0 resection rate in Table 1 ranges from 80.4% to 100%, which indicates positive outcomes.

Ongoing clinical trials in neoadjuvant immunotherapy

This period marks a surge in clinical trials focused on neoadjuvant immunotherapy, particularly for perioperative treatment of ESCC, with a significant number of ongoing RCTs, especially led by Chinese investigators (Table 2). Eight ICIs are currently under investigation, with dozens of trials in progress. PD-1 is the primary target in seven of these trials, while Adebrelimab targets PD-L1. Camrelizumab is the most extensively studied ICI, involved in ten clinical trials, followed by Toripalimab with seven trials. Among these trials, there are 23 single-arm studies, 11 two-arm studies, two three-arm studies, and one four-arm study.

Table 2 Ongoing clinical trials of neoadjuvant immunotherapy for resectable esophageal squamous cell carcinoma.
Drug & targetTrial name/No./Ref.PhaseSample sizeClinical stageDesignChemotherapy drugsChemotherapy cyclesRadiotherapyInterval to surgeryPrimary endpointStart date
NivolumabFRONTiER/NCT03914443[67]I36IC-IVATwo-arm5-FU + DDP2, Q3W12 wkIncidence of dose-limiting toxicities07-May-19
NCT03987815II20NASingle-armNA MPR01-Aug-19
NCT05213312II-III90II-III Two-armPTX/5-FU + DDP2, Q3W4-6 wkpCR01-Jun-22
PembrolizumabPALACE-1/NCT03792347[85]I20IC-IVASingle-armCBP + PTX5, Q1W41.4 Gy/23 f4-6 wkSafety21-Jan-19
PALACE-2/NCT04435197[84] II143IC-IVASingle-armCBP + PTX5, Q1 W41.4 Gy/23 f4-6 wkpCR11-Aug-20
NCT05302011II30IIB/IIIB/IVASingle-armCBP/DDP + DTX4, Q3WTumor response, pCR01-Jun-20
NCT05281003II128IC-IVASingle-armPTX + DDP4, Q4WpCR20-Feb-23
Camrelizumab NCT04520035II60IIB-IVASingle-armPTX + DDP2, Q3WpCR01-Aug-20
NCT04767295II28IA-IVASingle-armnab-PTX + CBP2, Q3W5-8 wkpCR01-Mar-21
NICE-2 Study/NCT 05043688[128] II204Locally advancedThree-armnab-PTX, CBP, PTX2, Q3W41.4 Gy/23 f4-12 wkpCR14-Sep-21
NCT05476380II39IIIB-IVASingle-armPTX + DDP3, Q3WpCR19-Feb-21
NCT05182944II130IIB-IVA Four-armnab-PTX + DDP2, Q3WpCR, 3-yr DFS 15-Jan-22
NCT04937673II40IIB-IVATwo-armPTX/nab-PTX + DDP3, Q3WNABiomarkers related to pCR01-Jul-21
NCT05176002I-II26II-IVASingle-armNANARadiotherapy, NANAEfficacy Safety 23-Sep-21
NCT04666090II42IIA-IVASingle-armnab-PTX + NDP + Apatinib2-3, Q2W4-6wkpCR23-Nov-20
NCT05355168I-II57IC-IVASingle-armNimotuzumab + CRTNApCR, MPR01-Nov-21
NCT03200691II21IIA-IIISingle-arm40 Gy/20 f2-4 wkpCR10-Aug-17
SintilimabNCT03940001I20IIB-IVASingle-armPTX + CBP2, Q3W41.4 Gy/23 fNAUnacceptable toxicity; pCR; MPR01-May-19
(NICCE)NCT05028231 NA46IIB-IVASingle-armnab-PTX + DDP2, Q3WpCR05-Jun-21
NCT05357846III422IIB-IVATwo-armPTX + DDP4, Q1W40 or 45 Gy / 20 f6-8 wkOS01-Nov-22
NCT05244798III420IC-IVAThree-armnab-PTX + CBP2, Q341.4 Gy/23 f6-8 wkpCR01-Nov-22
ToripalimabNCT04280822III400IC-IVATwo-armDDP + PTX2, Q3W2-3 wk3 yr EFS; 5 yr EFS21-Apr-20
NCT04804696II53NASingle-armPTX + DDPNANApCR10-Feb-21
NCT04177875II44IC-IIIBSingle-armDTX/PTX + DDP2, Q3W40 Gy/20 fNAMPR; ORR01-May-19
NCT04888403II45IIB-IVASingle-armnab-PTX + NDP5, Q1W41.4 Gy/23 fWithin 7 wkpCR31-Dec-21
NCT04644250II32IIB-IVASingle-armCBP + PTX liposome5, Q1W41.4 Gy/23 f2-4 wkpCR01-Sep-20
NCT04848753III632IC-IVATwo-armDDP + PTXNANAEFS18-Jun-21
NCT04006041 II44IIB-IVASingle-armPTX + DDP4, Q1W44 Gy/20 f 6-8 wkpCR25-Jun-19
TislelizumabiCROSS/NCT04973306II-III176II-IIITwo-armCBP + PTX5, Q1W41.4 Gy/23 fNApCR; OS02-Mar-22
NCT05323890II15IIB-IVASingle-armnab-PTX + DDP5, Q1W41.4 Gy/23 fNAMPR, pCR20-Apr-22
NCT04974047II70IIB-IVATwo-armPTX/5-FU + DDP2, NA40 or 45 Gy/20 fractionsNApCR17-Aug-21
NCT05189730II80II-IIISingle-armPTX + CBP2, Q3W40 Gy/20 f4-6 wk pCR, incidence of adverse events01-Jul-21
Adebrelimab (PD-L1)NATION1907II/NCT04215471II30ResectableSingle-armORR01-Feb-20
DurvalumabNCT04568200II60IIB-IVATwo-armCBP + PTX4, Q3W41.4 Gy/23 fpCR19-Jun-20
Pembrolizumab (+ Adjuvant)KEYSTONE-002/NCT04807673[86]III342IC-IIIBTwo-armPTX + DDP3, Q3W41.4 Gy/23 f4-6 wkEFS01-Dec-21
PD-1 InhibitorREVO/NCT05007145II92IB-IVATwo-armnab-PTX + DDP2-4, Q3W40 Gy/20 fpCR15-Aug-21
Toripalimab (+ Adjuvant) NCT 04437212II20IIB-IVASingle-armPTX/DDP5, Q3W41.4 Gy/23 f6-8 wkMPR01-Jul-20

An array of combinations of new adjuvant therapies for ESCC are constantly emerging. Most ICIs are being utilized as neoadjuvant adjuncts, combined with chemotherapy in 13 trials or CRT in 18 trials. Adebrelimab, a PD-L1-targeting ICI, is used alone in one trial (NCT04215471), as is Nivolumab in another (NCT03987815). Camrelizumab, on the other hand, is being combined with radiotherapy (NCT05176002 and NCT03200691), or paired with multitargeted small molecule inhibitors and CRT or chemotherapy (NCT04666090), or further combined with both CRT and an anti-EGFR antibody (NCT05355168).

Several large medical centers are now conducting phases II and III clinical studies based on promising results from phases I and II studies. Examples include PALACE-2 (NCT04435197)[84], an advancement of PALACE-1 (NCT03792347)[85], and KEYSTONE-002 (NCT04807673)[86], another advancement based on Keystone-001 (NCT04389177)[80]. Although Pembrolizumab is the ICI being studied in both series of trials, the PALACE trial was designed as neoadjuvant immunotherapy combined with chemotherapy, while the Keystone trial was designed as neoadjuvant combined with chemotherapy and adjuvant immunotherapy. Most chemotherapy regimens include taxols and platinum, with 5-FU being chosen in only two trials. The mainstream radiotherapy method utilizes 41.4 GY divided into 23 fractions, and the interval between neoadjuvant therapy and surgery ranges from 2 to 12 wk, with 4-6 wk being the most common choice. The primary outcome sought by most ongoing trials is pCR.

Ongoing clinical trials of adjuvant immunotherapy

Despite the benefits of neoadjuvant chemoradiation in improving survival compared to surgery alone, achieving a pCR remains challenging, with persistent disease in LNs leading to decreased survival[66]. An RCT involving 346 patients with ESCC compared preoperative and perioperative chemotherapy. Both groups received two cycles of paclitaxel, cisplatin, and 5-fluorouracil before surgery, but only half received two cycles after surgery. The group receiving adjuvant chemotherapy showed an estimated 16% improvement in 5-year survival[87].

In contrast to neoadjuvant methods, adjuvant immunotherapy has received relatively little attention. The global CheckMate-577 trial, a randomized, double-blind, placebo-controlled phase III trial, demonstrated the DFS benefit of Nivolumab for ESCC (29.7 vs 11.0 mo)[66]. Despite the promising results of CheckMate-577 for adjuvant immunotherapy after surgery, there are currently only three ongoing trials in this area. One trial is investigating the effectiveness of Toripalimab (NCT04437212) in both neoadjuvant and adjuvant settings. The other two RCTs are assessing postoperative adjuvant therapy with Tislelizumab, with one in phase II as a single-arm study and the other in phase III with two-arm trials. The phase II trial involves adjuvant immunotherapy combined with CRT, while the phase III trial is adjuvant immunotherapy combined with chemotherapy (Table 3). Further details and results from these ongoing trials are eagerly awaited.

Table 3 Clinical trials of adjuvant immunotherapy for resectable esophageal squamous cell carcinoma.
DrugTrial name/No.PhaseSample sizeClinical stageDesignChemotherapy drugsChemotherapy cyclesRadiotherapyInterval time to surgeryPrimary endpointStart dateOutcome
NivolumabCheckMate-577/NCT02743494III784 (ESCC29%)NATwo-armNADFSCompleted22.1:11.0 mo
Toripalimab (+ Neoadjuvant) NCT 04437212II20IIB-IVASingle-armPTX + DDP5, Q3W41.4 Gy/23 f6-8 wkMPRJul 1, 2020
Tislelizumab AIRE/ChiCTR2100045651III110High-risk resected locally advancedTwo-armPlatinum-based doublets 2, Q3WDFSMay 1, 2021
Tislelizumab CRISEC/NCT04776590II30NASingle-armPTX + CBP5, Q1W41.4 Gy/23 fpCRJan 28, 2021
ISSUES SHOULD BE CONCERNED IN FUTURE

The majority of neoadjuvant immunotherapy trials in ESCC are currently single-arm, phase II clinical trials. These trials explore various combinations of different drugs and different methods of other therapies, leading to a continuous emergence of new clinical trials in this field. However, it is important to acknowledge that progress in some cases has been slow or stagnant.

Ethical conduct should be a primary concern in the development of new therapies. As the development of immunotherapy is often driven by related industries, methodological, legal, and ethical frameworks can sometimes be overlooked. Currently, a significant portion of scientific research in immunotherapy is industry-driven, with various pharmaceutical companies involved in registering RCTs. Despite the existence of international research registries aimed at improving the transparency of medical research, there are still uncertainties and "not applicable" rules and regulations. Consequently, there may be a lack of control over data mining and publication bias. Even if there are significant differences between the research protocol and the reported results, most trial outcomes are still published. To ensure the reliability, quality, and expected clinical benefits of ongoing and future trials in this field, the medical community and relevant stakeholders should focus on curbing the significant increase in "feasibility" trials with unclear expected benefits. Emphasizing a few multi-center phase III trials, conducted by leading centers in ESCC research, could be a crucial approach to prevent unclear or contradictory results and uphold the integrity of the research.

The second issue of concern in clinical trials is safety, which should be carefully considered throughout the perioperative treatment of ESCC. One of the major safety risks is the occurrence of trAEs, especially immune-related adverse events. Perioperative immunotherapy trials for ESCC typically consist of three parts: Preoperative neoadjuvant therapy, surgery, and postoperative adjuvant therapy. Different immunotherapy drugs are administered at varying intervals: Pembrolizumab, Tislelizumab, Atilizumab, Toripalimab, and Sintilimab are usually given at 3-wk intervals, while Nivolumab, Camrelizumab, and Durvalumab are given at 2-wk intervals. The treatment cycles generally involve 2-4 cycles, and multiple factors such as treatment efficacy, surgical timing, economic considerations, and patient compliance are taken into account. Since most RCTs in this field are single-arm or two-arm studies within phase I or II, the safety data obtained may be limited and not fully comprehensive. This might result in an underestimation of the occurrence and severity of trAEs, especially those of grade 3 or above. Additionally, the introduction of new emerging drugs into clinical trials adds uncertain factors that could impact the results. Therefore, researchers should be attentive to the incidence and severity of trAEs, even when they are below grade 2. In cases where the trAEs are grade 3 or higher in severity, special attention should be given to the subjects to avoid fatal consequences. Furthermore, if three or more anti-cancer therapies, including immunotherapy, are administered to a subject simultaneously, researchers should exercise caution due to the higher risk of trAEs for the patient. Vigilance and thorough monitoring are essential to ensure the safety of subjects throughout the course of treatment.

Neoadjuvant immunotherapy presents a dual challenge, aiming to achieve better treatment outcomes while minimizing harm to normal organs caused by immunotherapy. The impact of trAEs during preoperative therapy on surgery must be carefully considered. In the trial "KEEP-G 03"[88], 36.7% (11/30) of patients experienced grades 3-4 TRAEs, but fortunately, these did not result in any surgical delays. However, in another multicenter, single-arm, phase II trial using Camrelizumab and chemotherapy as neoadjuvant treatment for locally advanced ESCC[74], 34 patients (56.7%) experienced adverse events of grade 3 or worse, with one patient (1.7%) experiencing a fatal grade 5 adverse event due to pneumonia and acute respiratory failure. The risk of increased surgical complications after immunotherapy is a concern, emphasizing the importance of forming a multidisciplinary team to address trAEs and conduct comprehensive evaluations during immunotherapy. Early detection and management of trAEs can minimize the impact on subsequent treatment and related complications. Additionally, exploring biomarkers related to trAEs is crucial in the research field.

Surgery itself poses another safety concern. ESCC radical resection is a complex procedure with a high incidence of complications, and surgical team experience significantly influences the rate of complications. Patients undergoing neoadjuvant treatment may face unexpected surgical challenges. High-volume medical units with stable and mature treatment processes, including neoadjuvant therapy and operations, generally have lower complication and mortality rates[89]. Therefore, RCTs involving neoadjuvant treatment should preferably be conducted in high-volume centers to reduce surgical risks and avoid any adverse impacts. Even in multi-center clinical trials, it is advisable to select larger surgical units or include surgeons with significant experience in performing over 100 operations on ESCC, both open and minimally invasive surgeries. Such measures ensure optimal surgical outcomes and minimize potential complications.

The third issue pertains to how we can accurately assess the efficacy of chosen drugs or treatments. Imaging is a crucial tool for preoperative efficacy assessment. Circulating tumor DNA (ct-DNA) has shown promise as an effective predictive method[90]. Positron emission tomography (PET) also has reference value as an assessment method[91,92]. Typically, pCR and MPR rates are used to predict efficacy, based on the examination of residual tumor cells in postoperative pathology. However, this method has certain limitations[93,94]. First, accurately assessing the pathological conditions of ESCC before immunotherapy, especially in cases of LN metastasis, is challenging. Detection can only be done in resected specimens after neoadjuvant immunotherapy, leading to an inability to make accurate comparisons with pathological specimens before surgery. Second, there is currently no universal standard for evaluating pathological response in ESCC after neoadjuvant immunotherapy. Pathologists are relied upon for positioning, measuring, sampling, slicing, and labeling, introducing variability. The different patterns of resection after neoadjuvant treatment, involving thoracic surgeons and pathologists, may also impact ESCC prognosis[95]. Thus, there is a need for a highly sensitive, specific, and preferably repeatable, simple, and feasible biomarker to predict efficacy in clinical practice.

PET is an integral part of the standard staging for ESCC. Its utility and acceptance in initial staging and recurrence detection have led to the hypothesis that PET could be used to differentiate responders from non-responders during neoadjuvant treatment. However, the precise PET parameters with the best predictive values are still a subject of debate. Several traditional PET parameters, such as the maximum and mean standardized uptake value (SUVmax and SUVmean), metabolic tumor volume (MTV), and total lesion glycolysis (TLG)[96], have been studied to correlate with pathological response. In a study of 31 patients with resectable ESCC or EAC, PET was used prospectively during treatment with trimodality therapy, and it was found that baseline TLG and post-chemoradiotherapy TLG were associated with OS[97]. More recently, efforts have been made to develop radiomic signatures and more robust predictive models. Simoni et al[98] investigated multiple traditional PET parameters and identified several radiomic features, as well as tumor regression grade, which correlated with pathological response in a retrospective analysis[98]. However, further research is necessary to develop more reliable predictive models and to validate them in prospective randomized trials. The credibility of PET evaluations in the era of immunotherapy remains largely unknown. Although PET has shown promise in predicting efficacy, especially with regard to OS, its applicability to immunotherapy response assessment is not fully understood. If sufficient high-quality CT or PET data, correlating with pathological response in ESCC, can be obtained, machine learning and artificial intelligence (AI) may emerge as new methods for evaluating neoadjuvant immunotherapy in ESCC. Such advancements could potentially enhance our ability to predict treatment outcomes and optimize patient care.

As of the current writing, specific biomarkers that can precisely determine the efficacy or predict perioperative surgery outcomes of ESCC have not been identified. However, several biomarkers have been explored mainly based on immunological and genetic criteria, including PD-L1 expression, intertumoral lymphoid infiltrates, dMMR/microsatellite instable (MSI), tumor mutation burden (TMB)/tumor neoantigen burden (TNB), and human leukocyte antigen (HLA)[99]. Among these biomarkers, PD-L1 expression is one of the best characterized for anti-PD-1/PD-L1 therapy. It is assessed using immunohistochemistry (IHC) staining and evaluated by the combined positive score (CPS) or tumor proportional score (TPS). CPS is determined by dividing the number of PD-L1-positive cells (tumor cells and other lymphocytes) by the total number of tumor cells[100], while TPS is calculated by dividing the number of PD-L1-positive tumor cells by the total number of tumor cells. In general, high PD-L1 expression usually correlates with an improved objective response to anti-PD-1/PD-L1 therapy[101]. However, some clinical studies have failed to consistently demonstrate this correlation, showing that patients with tumors showing high PD-L1 expression do not always respond to PD-1/PD-L1 blockade[102]. The relationship between PD-L1 expression and clinical outcome in ESCC remains a topic of controversy[103]. While some studies have shown that PD-L1 overexpression is associated with poor clinical outcomes[104], others have indicated a favorable prognosis[105].

Studies have also shown that circulating tumor cells (CTCs) and ct-DNA have a predictive role in evaluating the treatment efficacy of PD-1/PD-L1 inhibition[106,107]. CTCs have been found to be associated with a poor response to PD-L1 inhibitors in non-small cell lung cancer (NSCLC)[106]. Similarly, ct-DNA minimal residual disease (ct-DNA MRD) is an important indicator for monitoring the efficacy of treating NSCLC[108]. Therefore, it is worth exploring whether CTCs and ct-DNA can be introduced into the long-term follow-up of ESCC to help assess treatment response and predict patient outcomes. However, further research is needed to validate the predictive value of these biomarkers in ESCC and determine their potential role in guiding treatment decisions for patients undergoing perioperative immunotherapy.

The fourth issue pertains to the long-term benefits of perioperative immunotherapy. Many clinical trials on perioperative immunotherapy have focused on short-term or mid-term outcomes, such as safety, feasibility, and pCR. However, the true primary outcomes that need to be concerned with are long-term OS and PFS, regardless of the type of immunological drugs selected or the combination with other anti-cancer therapies. For neoadjuvant anti-PD-1 therapy in resectable NSCLC, a trial with a median follow-up of 63 mo (NCT02259621) reported promising long-term outcomes. The 5-year recurrence-free survival (RFS) and OS rates were 60% and 80%, respectively[109]. Similarly, a study evaluating neoadjuvant anti-PD-1 treatment for localized dMMR colorectal cancer (CRC) also reported positive long-term follow-up data. Among patients who underwent surgery or achieved complete response, the 2-year tumor-specific disease-free and OS rates were both 100%[110].

In the context of resectable locally advanced ESCC, neoadjuvant chemotherapy and nCRT remain the standard treatments before surgery. While immune checkpoint-based therapy shows promise, it currently benefits only a small proportion of ESCC patients. Therefore, perioperative immunotherapy should be strictly monitored with ethical confirmation and preferably conducted within clinical trials. Patients must be fully informed about the potential benefits and risks and give informed consent before participating. Accurate screening of target populations and appropriate choice of combination therapy will be crucial for future research in this field. The importance of monitoring and managing trAEs, especially when combining immunotherapies with other anti-cancer therapies, cannot be ignored. Robust predictive and prognostic biomarkers or comprehensive biomarkers need to be identified to optimize treatment strategies and ensure the most effective therapy for patients. The development of clinical consensus or guidelines based on research findings will also be necessary to ensure that patients receive the most applicable and effective immunotherapy treatments.

CONCLUSION

The utilization of immunotherapy is progressively transitioning from a second-line approach for advanced or metastatic cases to a perioperative strategy for resectable locally advanced ESCC. Despite the current results lacking comprehensiveness and robustness, substantial advancements in perioperative immunotherapy for ESCC are evident. With the assurance derived from existing outcomes, numerous EC centers are now engaged in conducting multicenter, multi-arm RCTs. These RCTs hold the promise of providing further insights into the value of perioperative immunotherapy. It is plausible that in the near future, perioperative immunotherapy will emerge as a pivotal component of comprehensive treatment for resectable locally advanced ESCC. However, the determination of the optimal drug or the most effective combination of therapies, as well as the potential role of AI as an assistant, will require further observation and investigation.

Footnotes

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

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country/Territory of origin: China

Peer-review report’s scientific quality classification

Grade A (Excellent): A

Grade B (Very good): B

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Matsuo Y, Japan; Nakano H, Japan; Viswanath Y, United Kingdom S-Editor: Yan JP L-Editor: Wang TQ P-Editor: Cai YX

References
1.  Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71:209-249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50630]  [Cited by in F6Publishing: 43963]  [Article Influence: 14654.3]  [Reference Citation Analysis (47)]
2.  Rustgi AK, El-Serag HB. Esophageal carcinoma. N Engl J Med. 2014;371:2499-2509.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 817]  [Cited by in F6Publishing: 905]  [Article Influence: 90.5]  [Reference Citation Analysis (0)]
3.  Yan Y, Feng X, Li C, Lerut T, Li H. Treatments for resectable esophageal cancer: from traditional systemic therapy to immunotherapy. Chin Med J (Engl). 2022;135:2143-2156.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 6]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
4.  Plum PS, Hölscher AH, Pacheco Godoy K, Schmidt H, Berlth F, Chon SH, Alakus H, Bollschweiler E. Prognosis of patients with superficial T1 esophageal cancer who underwent endoscopic resection before esophagectomy-A propensity score-matched comparison. Surg Endosc. 2018;32:3972-3980.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 18]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
5.  Omloo JM, Lagarde SM, Hulscher JB, Reitsma JB, Fockens P, van Dekken H, Ten Kate FJ, Obertop H, Tilanus HW, van Lanschot JJ. Extended transthoracic resection compared with limited transhiatal resection for adenocarcinoma of the mid/distal esophagus: five-year survival of a randomized clinical trial. Ann Surg. 2007;246:992-1000; discussion 1000.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 519]  [Cited by in F6Publishing: 497]  [Article Influence: 31.1]  [Reference Citation Analysis (0)]
6.  Oppedijk V, van der Gaast A, van Lanschot JJ, van Hagen P, van Os R, van Rij CM, van der Sangen MJ, Beukema JC, Rütten H, Spruit PH, Reinders JG, Richel DJ, van Berge Henegouwen MI, Hulshof MC. Patterns of recurrence after surgery alone versus preoperative chemoradiotherapy and surgery in the CROSS trials. J Clin Oncol. 2014;32:385-391.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 280]  [Cited by in F6Publishing: 331]  [Article Influence: 33.1]  [Reference Citation Analysis (0)]
7.  Chen G, Wang Z, Liu XY, Liu FY. Recurrence pattern of squamous cell carcinoma in the middle thoracic esophagus after modified Ivor-Lewis esophagectomy. World J Surg. 2007;31:1107-1114.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 79]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
8.  Leng XF, Daiko H, Han YT, Mao YS. Optimal preoperative neoadjuvant therapy for resectable locally advanced esophageal squamous cell carcinoma. Ann N Y Acad Sci. 2020;1482:213-224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 48]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
9.  Yang H, Liu H, Chen Y, Zhu C, Fang W, Yu Z, Mao W, Xiang J, Han Y, Chen Z, Yang H, Wang J, Pang Q, Zheng X, Li T, Lordick F, D'Journo XB, Cerfolio RJ, Korst RJ, Novoa NM, Swanson SJ, Brunelli A, Ismail M, Fernando HC, Zhang X, Li Q, Wang G, Chen B, Mao T, Kong M, Guo X, Lin T, Liu M, Fu J; AME Thoracic Surgery Collaborative Group. Neoadjuvant Chemoradiotherapy Followed by Surgery Versus Surgery Alone for Locally Advanced Squamous Cell Carcinoma of the Esophagus (NEOCRTEC5010): A Phase III Multicenter, Randomized, Open-Label Clinical Trial. J Clin Oncol. 2018;36:2796-2803.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 362]  [Cited by in F6Publishing: 505]  [Article Influence: 84.2]  [Reference Citation Analysis (0)]
10.  Lewis S, Lukovic J. Neoadjuvant Therapy in Esophageal Cancer. Thorac Surg Clin. 2022;32:447-456.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
11.  Shah MA, Kennedy EB, Catenacci DV, Deighton DC, Goodman KA, Malhotra NK, Willett C, Stiles B, Sharma P, Tang L, Wijnhoven BPL, Hofstetter WL. Treatment of Locally Advanced Esophageal Carcinoma: ASCO Guideline. J Clin Oncol. 2020;38:2677-2694.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 145]  [Article Influence: 36.3]  [Reference Citation Analysis (0)]
12.  Puetz K, Bollschweiler E, Semrau R, Mönig SP, Hölscher AH, Drebber U. Neoadjuvant chemoradiation for patients with advanced oesophageal cancer - which response grading system best impacts prognostic discrimination? Histopathology. 2019;74:731-743.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 18]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
13.  Shapiro J, van Lanschot JJB, Hulshof MCCM, van Hagen P, van Berge Henegouwen MI, Wijnhoven BPL, van Laarhoven HWM, Nieuwenhuijzen GAP, Hospers GAP, Bonenkamp JJ, Cuesta MA, Blaisse RJB, Busch ORC, Ten Kate FJW, Creemers GM, Punt CJA, Plukker JTM, Verheul HMW, Bilgen EJS, van Dekken H, van der Sangen MJC, Rozema T, Biermann K, Beukema JC, Piet AHM, van Rij CM, Reinders JG, Tilanus HW, Steyerberg EW, van der Gaast A; CROSS study group. Neoadjuvant chemoradiotherapy plus surgery versus surgery alone for oesophageal or junctional cancer (CROSS): long-term results of a randomised controlled trial. Lancet Oncol. 2015;16:1090-1098.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1292]  [Cited by in F6Publishing: 1578]  [Article Influence: 175.3]  [Reference Citation Analysis (0)]
14.  Sun HB, Xing WQ, Liu XB, Zheng Y, Yang SJ, Wang ZF, Liu SL, Ba YF, Zhang RX, Liu BX, Fan CC, Chen PN, Liang GH, Yu YK, Liu Q, Wang HR, Li HM, Li ZX; written on Henan Cancer Hospital Thoracic Oncology Group (HCHTOG). Neoadjuvant chemotherapy versus neoadjuvant chemoradiotherapy for locally advanced oesophageal squamous cell carcinoma: a single-Centre, open-label, randomized, controlled, clinical trial (HCHTOG1903). BMC Cancer. 2020;20:303.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 8]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
15.  Tepper J, Krasna MJ, Niedzwiecki D, Hollis D, Reed CE, Goldberg R, Kiel K, Willett C, Sugarbaker D, Mayer R. Phase III trial of trimodality therapy with cisplatin, fluorouracil, radiotherapy, and surgery compared with surgery alone for esophageal cancer: CALGB 9781. J Clin Oncol. 2008;26:1086-1092.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 941]  [Cited by in F6Publishing: 1000]  [Article Influence: 62.5]  [Reference Citation Analysis (0)]
16.  van Hagen P, Hulshof MC, van Lanschot JJ, Steyerberg EW, van Berge Henegouwen MI, Wijnhoven BP, Richel DJ, Nieuwenhuijzen GA, Hospers GA, Bonenkamp JJ, Cuesta MA, Blaisse RJ, Busch OR, ten Kate FJ, Creemers GJ, Punt CJ, Plukker JT, Verheul HM, Spillenaar Bilgen EJ, van Dekken H, van der Sangen MJ, Rozema T, Biermann K, Beukema JC, Piet AH, van Rij CM, Reinders JG, Tilanus HW, van der Gaast A; CROSS Group. Preoperative chemoradiotherapy for esophageal or junctional cancer. N Engl J Med. 2012;366:2074-2084.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3288]  [Cited by in F6Publishing: 3653]  [Article Influence: 304.4]  [Reference Citation Analysis (0)]
17.  Zhou HY, Zheng SP, Li AL, Gao QL, Ou QY, Chen YJ, Wu ST, Lin DG, Liu SB, Huang LY, Li FS, Zhu HY, Qiao GB, Lanuti M, Yao HR, Yu YF. Clinical evidence for association of neoadjuvant chemotherapy or chemoradiotherapy with efficacy and safety in patients with resectable esophageal carcinoma (NewEC study). EClinicalMedicine. 2020;24:100422.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 11]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
18.  Chan KKW, Saluja R, Delos Santos K, Lien K, Shah K, Cramarossa G, Zhu X, Wong RKS. Neoadjuvant treatments for locally advanced, resectable esophageal cancer: A network meta-analysis. Int J Cancer. 2018;143:430-437.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 68]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
19.  Pasquali S, Yim G, Vohra RS, Mocellin S, Nyanhongo D, Marriott P, Geh JI, Griffiths EA. Survival After Neoadjuvant and Adjuvant Treatments Compared to Surgery Alone for Resectable Esophageal Carcinoma: A Network Meta-analysis. Ann Surg. 2017;265:481-491.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 124]  [Article Influence: 17.7]  [Reference Citation Analysis (0)]
20.  Mariette C, Dahan L, Mornex F, Maillard E, Thomas PA, Meunier B, Boige V, Pezet D, Robb WB, Le Brun-Ly V, Bosset JF, Mabrut JY, Triboulet JP, Bedenne L, Seitz JF. Surgery alone versus chemoradiotherapy followed by surgery for stage I and II esophageal cancer: final analysis of randomized controlled phase III trial FFCD 9901. J Clin Oncol. 2014;32:2416-2422.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 354]  [Cited by in F6Publishing: 418]  [Article Influence: 41.8]  [Reference Citation Analysis (0)]
21.  Murakami Y, Hamai Y, Emi M, Hihara J, Imano N, Takeuchi Y, Takahashi I, Nishibuchi I, Kimura T, Okada M, Nagata Y. Long-term results of neoadjuvant chemoradiotherapy using cisplatin and 5-fluorouracil followed by esophagectomy for resectable, locally advanced esophageal squamous cell carcinoma. J Radiat Res. 2018;59:616-624.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 13]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
22.  Ajani JA, D'Amico TA, Bentrem DJ, Chao J, Corvera C, Das P, Denlinger CS, Enzinger PC, Fanta P, Farjah F, Gerdes H, Gibson M, Glasgow RE, Hayman JA, Hochwald S, Hofstetter WL, Ilson DH, Jaroszewski D, Johung KL, Keswani RN, Kleinberg LR, Leong S, Ly QP, Matkowskyj KA, McNamara M, Mulcahy MF, Paluri RK, Park H, Perry KA, Pimiento J, Poultsides GA, Roses R, Strong VE, Wiesner G, Willett CG, Wright CD, McMillian NR, Pluchino LA. Esophageal and Esophagogastric Junction Cancers, Version 2.2019, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2019;17:855-883.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 568]  [Cited by in F6Publishing: 550]  [Article Influence: 110.0]  [Reference Citation Analysis (0)]
23.  Ajani JA, D'Amico TA, Bentrem DJ, Cooke D, Corvera C, Das P, Enzinger PC, Enzler T, Farjah F, Gerdes H, Gibson M, Grierson P, Hofstetter WL, Ilson DH, Jalal S, Keswani RN, Kim S, Kleinberg LR, Klempner S, Lacy J, Licciardi F, Ly QP, Matkowskyj KA, McNamara M, Miller A, Mukherjee S, Mulcahy MF, Outlaw D, Perry KA, Pimiento J, Poultsides GA, Reznik S, Roses RE, Strong VE, Su S, Wang HL, Wiesner G, Willett CG, Yakoub D, Yoon H, McMillian NR, Pluchino LA. Esophageal and Esophagogastric Junction Cancers, Version 2.2023, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2023;21:393-422.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 40]  [Reference Citation Analysis (0)]
24.  Muro K, Lordick F, Tsushima T, Pentheroudakis G, Baba E, Lu Z, Cho BC, Nor IM, Ng M, Chen LT, Kato K, Li J, Ryu MH, Zamaniah WIW, Yong WP, Yeh KH, Nakajima TE, Shitara K, Kawakami H, Narita Y, Yoshino T, Van Cutsem E, Martinelli E, Smyth EC, Arnold D, Minami H, Tabernero J, Douillard JY. Pan-Asian adapted ESMO Clinical Practice Guidelines for the management of patients with metastatic oesophageal cancer: a JSMO-ESMO initiative endorsed by CSCO, KSMO, MOS, SSO and TOS. Ann Oncol. 2019;30:34-43.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 80]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
25.  Zhang W, Wang P, Pang Q. Immune checkpoint inhibitors for esophageal squamous cell carcinoma: a narrative review. Ann Transl Med. 2020;8:1193.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 10]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
26.  Eyck BM, van Lanschot JJB, Hulshof MCCM, van der Wilk BJ, Shapiro J, van Hagen P, van Berge Henegouwen MI, Wijnhoven BPL, van Laarhoven HWM, Nieuwenhuijzen GAP, Hospers GAP, Bonenkamp JJ, Cuesta MA, Blaisse RJB, Busch OR, Creemers GM, Punt CJA, Plukker JTM, Verheul HMW, Spillenaar Bilgen EJ, van der Sangen MJC, Rozema T, Ten Kate FJW, Beukema JC, Piet AHM, van Rij CM, Reinders JG, Tilanus HW, Steyerberg EW, van der Gaast A; CROSS Study Group. Ten-Year Outcome of Neoadjuvant Chemoradiotherapy Plus Surgery for Esophageal Cancer: The Randomized Controlled CROSS Trial. J Clin Oncol. 2021;39:1995-2004.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 257]  [Article Influence: 85.7]  [Reference Citation Analysis (0)]
27.  Groom JR. Regulators of T-cell fate: Integration of cell migration, differentiation and function. Immunol Rev. 2019;289:101-114.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 39]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
28.  Masopust D, Schenkel JM. The integration of T cell migration, differentiation and function. Nat Rev Immunol. 2013;13:309-320.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 382]  [Cited by in F6Publishing: 419]  [Article Influence: 38.1]  [Reference Citation Analysis (0)]
29.  Baba Y, Nomoto D, Okadome K, Ishimoto T, Iwatsuki M, Miyamoto Y, Yoshida N, Baba H. Tumor immune microenvironment and immune checkpoint inhibitors in esophageal squamous cell carcinoma. Cancer Sci. 2020;111:3132-3141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in F6Publishing: 132]  [Article Influence: 33.0]  [Reference Citation Analysis (0)]
30.  Janjigian YY, Bendell J, Calvo E, Kim JW, Ascierto PA, Sharma P, Ott PA, Peltola K, Jaeger D, Evans J, de Braud F, Chau I, Harbison CT, Dorange C, Tschaika M, Le DT. CheckMate-032 Study: Efficacy and Safety of Nivolumab and Nivolumab Plus Ipilimumab in Patients With Metastatic Esophagogastric Cancer. J Clin Oncol. 2018;36:2836-2844.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 407]  [Cited by in F6Publishing: 473]  [Article Influence: 78.8]  [Reference Citation Analysis (0)]
31.  Kudo T, Hamamoto Y, Kato K, Ura T, Kojima T, Tsushima T, Hironaka S, Hara H, Satoh T, Iwasa S, Muro K, Yasui H, Minashi K, Yamaguchi K, Ohtsu A, Doki Y, Kitagawa Y. Nivolumab treatment for oesophageal squamous-cell carcinoma: an open-label, multicentre, phase 2 trial. Lancet Oncol. 2017;18:631-639.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 238]  [Cited by in F6Publishing: 278]  [Article Influence: 39.7]  [Reference Citation Analysis (0)]
32.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 299]  [Cited by in F6Publishing: 507]  [Article Influence: 126.8]  [Reference Citation Analysis (0)]
33.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 77]  [Article Influence: 38.5]  [Reference Citation Analysis (0)]
34.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 428]  [Cited by in F6Publishing: 645]  [Article Influence: 129.0]  [Reference Citation Analysis (0)]
35.  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, 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 170]  [Cited by in F6Publishing: 317]  [Article Influence: 79.3]  [Reference Citation Analysis (0)]
36.  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, 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, Zhu Z, Chen R, Jia L, Li W, Liao W, Liu HX, Ma D, Ma J, Qin Y, Shi Z, Wei Q, Xiao K, Chen X, Dai G, He J, Li J, Li G, Liu Z, Yuan X, Zhang J, Fu Z, He Y, Ju F, Tang P, Wang T, Wang W, 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 141]  [Article Influence: 70.5]  [Reference Citation Analysis (0)]
37.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 174]  [Cited by in F6Publishing: 344]  [Article Influence: 172.0]  [Reference Citation Analysis (0)]
38.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 108]  [Article Influence: 54.0]  [Reference Citation Analysis (0)]
39.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 137]  [Cited by in F6Publishing: 266]  [Article Influence: 88.7]  [Reference Citation Analysis (0)]
40.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 310]  [Cited by in F6Publishing: 534]  [Article Influence: 178.0]  [Reference Citation Analysis (0)]
41.  Forde PM, Chaft JE, Smith KN, Anagnostou V, Cottrell TR, Hellmann MD, Zahurak M, Yang SC, Jones DR, Broderick S, Battafarano RJ, Velez MJ, Rekhtman N, Olah Z, Naidoo J, Marrone KA, Verde F, Guo H, Zhang J, Caushi JX, Chan HY, Sidhom JW, Scharpf RB, White J, Gabrielson E, Wang H, Rosner GL, Rusch V, Wolchok JD, Merghoub T, Taube JM, Velculescu VE, Topalian SL, Brahmer JR, Pardoll DM. Neoadjuvant PD-1 Blockade in Resectable Lung Cancer. N Engl J Med. 2018;378:1976-1986.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1051]  [Cited by in F6Publishing: 1251]  [Article Influence: 208.5]  [Reference Citation Analysis (0)]
42.  Provencio M, Nadal E, Insa A, García-Campelo MR, Casal-Rubio J, Dómine M, Majem M, Rodríguez-Abreu D, Martínez-Martí A, De Castro Carpeño J, Cobo M, López Vivanco G, Del Barco E, Bernabé Caro R, Viñolas N, Barneto Aranda I, Viteri S, Pereira E, Royuela A, Casarrubios M, Salas Antón C, Parra ER, Wistuba I, Calvo V, Laza-Briviesca R, Romero A, Massuti B, Cruz-Bermúdez A. Neoadjuvant chemotherapy and nivolumab in resectable non-small-cell lung cancer (NADIM): an open-label, multicentre, single-arm, phase 2 trial. Lancet Oncol. 2020;21:1413-1422.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 283]  [Cited by in F6Publishing: 397]  [Article Influence: 99.3]  [Reference Citation Analysis (0)]
43.  Huang AC, Orlowski RJ, Xu X, Mick R, George SM, Yan PK, Manne S, Kraya AA, Wubbenhorst B, Dorfman L, D'Andrea K, Wenz BM, Liu S, Chilukuri L, Kozlov A, Carberry M, Giles L, Kier MW, Quagliarello F, McGettigan S, Kreider K, Annamalai L, Zhao Q, Mogg R, Xu W, Blumenschein WM, Yearley JH, Linette GP, Amaravadi RK, Schuchter LM, Herati RS, Bengsch B, Nathanson KL, Farwell MD, Karakousis GC, Wherry EJ, Mitchell TC. A single dose of neoadjuvant PD-1 blockade predicts clinical outcomes in resectable melanoma. Nat Med. 2019;25:454-461.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 323]  [Cited by in F6Publishing: 409]  [Article Influence: 81.8]  [Reference Citation Analysis (0)]
44.  Powles T, Kockx M, Rodriguez-Vida A, Duran I, Crabb SJ, Van Der Heijden MS, Szabados B, Pous AF, Gravis G, Herranz UA, Protheroe A, Ravaud A, Maillet D, Mendez MJ, Suarez C, Linch M, Prendergast A, van Dam PJ, Stanoeva D, Daelemans S, Mariathasan S, Tea JS, Mousa K, Banchereau R, Castellano D. Clinical efficacy and biomarker analysis of neoadjuvant atezolizumab in operable urothelial carcinoma in the ABACUS trial. Nat Med. 2019;25:1706-1714.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 285]  [Cited by in F6Publishing: 354]  [Article Influence: 70.8]  [Reference Citation Analysis (0)]
45.  Chalabi M, Fanchi LF, Dijkstra KK, Van den Berg JG, Aalbers AG, Sikorska K, Lopez-Yurda M, Grootscholten C, Beets GL, Snaebjornsson P, Maas M, Mertz M, Veninga V, Bounova G, Broeks A, Beets-Tan RG, de Wijkerslooth TR, van Lent AU, Marsman HA, Nuijten E, Kok NF, Kuiper M, Verbeek WH, Kok M, Van Leerdam ME, Schumacher TN, Voest EE, Haanen JB. Neoadjuvant immunotherapy leads to pathological responses in MMR-proficient and MMR-deficient early-stage colon cancers. Nat Med. 2020;26:566-576.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 341]  [Cited by in F6Publishing: 625]  [Article Influence: 156.3]  [Reference Citation Analysis (0)]
46.  Schalper KA, Rodriguez-Ruiz ME, Diez-Valle R, López-Janeiro A, Porciuncula A, Idoate MA, Inogés S, de Andrea C, López-Diaz de Cerio A, Tejada S, Berraondo P, Villarroel-Espindola F, Choi J, Gúrpide A, Giraldez M, Goicoechea I, Gallego Perez-Larraya J, Sanmamed MF, Perez-Gracia JL, Melero I. Neoadjuvant nivolumab modifies the tumor immune microenvironment in resectable glioblastoma. Nat Med. 2019;25:470-476.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 331]  [Cited by in F6Publishing: 408]  [Article Influence: 81.6]  [Reference Citation Analysis (0)]
47.  Whiteside TL. The tumor microenvironment and its role in promoting tumor growth. Oncogene. 2008;27:5904-5912.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1334]  [Cited by in F6Publishing: 1568]  [Article Influence: 98.0]  [Reference Citation Analysis (0)]
48.  Blessin NC, Li W, Mandelkow T, Jansen HL, Yang C, Raedler JB, Simon R, Büscheck F, Dum D, Luebke AM, Hinsch A, Möller K, Menz A, Bernreuther C, Lebok P, Clauditz T, Sauter G, Marx A, Uhlig R, Wilczak W, Minner S, Krech T, Fraune C, Höflmayer D, Burandt E, Steurer S. Prognostic role of proliferating CD8(+) cytotoxic Tcells in human cancers. Cell Oncol (Dordr). 2021;44:793-803.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 9]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
49.  Zhang N, Bevan MJ. CD8(+) T cells: foot soldiers of the immune system. Immunity. 2011;35:161-168.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 561]  [Cited by in F6Publishing: 632]  [Article Influence: 48.6]  [Reference Citation Analysis (0)]
50.  Nurieva RI, Liu X, Dong C. Yin-Yang of costimulation: crucial controls of immune tolerance and function. Immunol Rev. 2009;229:88-100.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 111]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
51.  Vinay DS, Ryan EP, Pawelec G, Talib WH, Stagg J, Elkord E, Lichtor T, Decker WK, Whelan RL, Kumara HMCS, Signori E, Honoki K, Georgakilas AG, Amin A, Helferich WG, Boosani CS, Guha G, Ciriolo MR, Chen S, Mohammed SI, Azmi AS, Keith WN, Bilsland A, Bhakta D, Halicka D, Fujii H, Aquilano K, Ashraf SS, Nowsheen S, Yang X, Choi BK, Kwon BS. Immune evasion in cancer: Mechanistic basis and therapeutic strategies. Semin Cancer Biol. 2015;35 Suppl:S185-S198.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 758]  [Cited by in F6Publishing: 938]  [Article Influence: 104.2]  [Reference Citation Analysis (0)]
52.  Baldwin XL, Spanheimer PM, Downs-Canner S. A Review of Immune Checkpoint Blockade for the General Surgeon. J Surg Res. 2023;281:289-298.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 3]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
53.  Buchbinder EI, Desai A. CTLA-4 and PD-1 Pathways: Similarities, Differences, and Implications of Their Inhibition. Am J Clin Oncol. 2016;39:98-106.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1427]  [Cited by in F6Publishing: 1423]  [Article Influence: 177.9]  [Reference Citation Analysis (0)]
54.  Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science. 2018;359:1350-1355.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2848]  [Cited by in F6Publishing: 3716]  [Article Influence: 619.3]  [Reference Citation Analysis (0)]
55.  Sharpe AH, Pauken KE. The diverse functions of the PD1 inhibitory pathway. Nat Rev Immunol. 2018;18:153-167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 774]  [Cited by in F6Publishing: 808]  [Article Influence: 115.4]  [Reference Citation Analysis (0)]
56.  Geng Q, Rohondia SO, Khan HJ, Jiao P, Dou QP. Small molecules as antagonists of co-inhibitory pathways for cancer immunotherapy: a patent review (2018-2019). Expert Opin Ther Pat. 2020;30:677-694.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 4]  [Reference Citation Analysis (0)]
57.  Baumeister SH, Freeman GJ, Dranoff G, Sharpe AH. Coinhibitory Pathways in Immunotherapy for Cancer. Annu Rev Immunol. 2016;34:539-573.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 528]  [Cited by in F6Publishing: 628]  [Article Influence: 78.5]  [Reference Citation Analysis (0)]
58.  Yi M, Zheng X, Niu M, Zhu S, Ge H, Wu K. Combination strategies with PD-1/PD-L1 blockade: current advances and future directions. Mol Cancer. 2022;21:28.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 400]  [Cited by in F6Publishing: 371]  [Article Influence: 185.5]  [Reference Citation Analysis (0)]
59.  Awadasseid A, Wu Y, Zhang W. Advance investigation on synthetic small-molecule inhibitors targeting PD-1/PD-L1 signaling pathway. Life Sci. 2021;282:119813.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 18]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
60.  Wu Q, Jiang L, Li SC, He QJ, Yang B, Cao J. Small molecule inhibitors targeting the PD-1/PD-L1 signaling pathway. Acta Pharmacol Sin. 2021;42:1-9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 197]  [Cited by in F6Publishing: 148]  [Article Influence: 49.3]  [Reference Citation Analysis (0)]
61.  Maruhashi T, Sugiura D, Okazaki IM, Okazaki T. LAG-3: from molecular functions to clinical applications. J Immunother Cancer. 2020;8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 224]  [Article Influence: 56.0]  [Reference Citation Analysis (0)]
62.  Meng X, Wu T, Hong Y, Fan Q, Ren Z, Guo Y, Yang X, Shi P, Yang J, Yin X, Luo Z, Xia J, Zhou Y, Xu M, Liu E, Jiang G, Li S, Zhao F, Ma C, Hou Z, Li J, Wang J, Wang F. Camrelizumab plus apatinib as second-line treatment for advanced oesophageal squamous cell carcinoma (CAP 02): a single-arm, open-label, phase 2 trial. Lancet Gastroenterol Hepatol. 2022;7:245-253.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 11]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
63.  Huang J, Xu B, Mo H, Zhang W, Chen X, Wu D, Qu D, Wang X, Lan B, Yang B, Wang P, Zhang H, Yang Q, Jiao Y. Safety, Activity, and Biomarkers of SHR-1210, an Anti-PD-1 Antibody, for Patients with Advanced Esophageal Carcinoma. Clin Cancer Res. 2018;24:1296-1304.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 133]  [Article Influence: 22.2]  [Reference Citation Analysis (0)]
64.  O'Donnell JS, Hoefsmit EP, Smyth MJ, Blank CU, Teng MWL. The Promise of Neoadjuvant Immunotherapy and Surgery for Cancer Treatment. Clin Cancer Res. 2019;25:5743-5751.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 89]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
65.  Topalian SL, Taube JM, Pardoll DM. Neoadjuvant checkpoint blockade for cancer immunotherapy. Science. 2020;367.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 443]  [Cited by in F6Publishing: 512]  [Article Influence: 128.0]  [Reference Citation Analysis (0)]
66.  Kelly RJ, Ajani JA, Kuzdzal J, Zander T, Van Cutsem E, Piessen G, Mendez G, Feliciano J, Motoyama S, Lièvre A, Uronis H, Elimova E, Grootscholten C, Geboes K, Zafar S, Snow S, Ko AH, Feeney K, Schenker M, Kocon P, Zhang J, Zhu L, Lei M, Singh P, Kondo K, Cleary JM, Moehler M; CheckMate 577 Investigators. Adjuvant Nivolumab in Resected Esophageal or Gastroesophageal Junction Cancer. N Engl J Med. 2021;384:1191-1203.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 469]  [Cited by in F6Publishing: 704]  [Article Influence: 234.7]  [Reference Citation Analysis (0)]
67.  Yamamoto S, Kato K, Daiko H, Kojima T, Hara H, Abe T, Tsubosa Y, Nagashima K, Aoki K, Mizoguchi Y, Kitano S, Yachida S, Shiba S, Kitagawa Y. Feasibility study of nivolumab as neoadjuvant chemotherapy for locally esophageal carcinoma: FRONTiER (JCOG1804E). Future Oncol. 2020;16:1351-1357.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 34]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
68.  Jia XH, Xu H, Geng LY, Jiao M, Wang WJ, Jiang LL, Guo H. Efficacy and safety of neoadjuvant immunotherapy in resectable nonsmall cell lung cancer: A meta-analysis. Lung Cancer. 2020;147:143-153.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 56]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
69.  Zhou X, Yao Z, Bai H, Duan J, Wang Z, Wang X, Zhang X, Xu J, Fei K, Zhang Z, Tan F, Xue Q, Gao S, Gao Y, Wang J, He J. Treatment-related adverse events of PD-1 and PD-L1 inhibitor-based combination therapies in clinical trials: a systematic review and meta-analysis. Lancet Oncol. 2021;22:1265-1274.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 86]  [Article Influence: 28.7]  [Reference Citation Analysis (0)]
70.  Wang Y, Zhou S, Yang F, Qi X, Wang X, Guan X, Shen C, Duma N, Vera Aguilera J, Chintakuntlawar A, Price KA, Molina JR, Pagliaro LC, Halfdanarson TR, Grothey A, Markovic SN, Nowakowski GS, Ansell SM, Wang ML. Treatment-Related Adverse Events of PD-1 and PD-L1 Inhibitors in Clinical Trials: A Systematic Review and Meta-analysis. JAMA Oncol. 2019;5:1008-1019.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 378]  [Cited by in F6Publishing: 462]  [Article Influence: 92.4]  [Reference Citation Analysis (0)]
71.  Khoja L, Day D, Wei-Wu Chen T, Siu LL, Hansen AR. Tumour- and class-specific patterns of immune-related adverse events of immune checkpoint inhibitors: a systematic review. Ann Oncol. 2017;28:2377-2385.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 390]  [Cited by in F6Publishing: 518]  [Article Influence: 86.3]  [Reference Citation Analysis (0)]
72.  Gao TT, Shan JH, Yang YX, Zhang ZW, Liu SL, Xi M, Liu MZ, Zhao L. Comparative efficacy and safety of immunotherapy for patients with advanced or metastatic esophageal squamous cell carcinoma: a systematic review and network Meta-analysis. BMC Cancer. 2022;22:992.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
73.  Zhang Z, Hong ZN, Xie S, Lin W, Lin Y, Zhu J, Yang X, Lin Z, Lin J, Kang M. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 36]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
74.  Liu J, Yang Y, Liu Z, Fu X, Cai X, Li H, Zhu L, Shen Y, Zhang H, Sun Y, Chen H, Yu B, Zhang R, Shao J, Zhang M, Li Z. Multicenter, single-arm, phase II trial of camrelizumab and chemotherapy as neoadjuvant treatment for locally advanced esophageal squamous cell carcinoma. J Immunother Cancer. 2022;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 70]  [Article Influence: 35.0]  [Reference Citation Analysis (0)]
75.  Liu J, Li J, Lin W, Shao D, Depypere L, Zhang Z, Li Z, Cui F, Du Z, Zeng Y, Jiang S, He P, Gu X, Chen H, Zhang H, Lin X, Huang H, Lv W, Cai W, Liang W, Liang H, Jiang W, Wang W, Xu K, Wu K, Lerut T, Fu J, He J. Neoadjuvant camrelizumab plus chemotherapy for resectable, locally advanced esophageal squamous cell carcinoma (NIC-ESCC2019): A multicenter, phase 2 study. Int J Cancer. 2022;151:128-137.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 61]  [Article Influence: 30.5]  [Reference Citation Analysis (0)]
76.  Yan X, Duan H, Ni Y, Zhou Y, Wang X, Qi H, Gong L, Liu H, Tian F, Lu Q, Sun J, Yang E, Zhong D, Wang T, Huang L, Wang J, Chaoyang Wang, Wang Y, Wan Z, Lei J, Zhao J, Jiang T. Tislelizumab combined with chemotherapy as neoadjuvant therapy for surgically resectable esophageal cancer: A prospective, single-arm, phase II study (TD-NICE). Int J Surg. 2022;103:106680.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 57]  [Article Influence: 28.5]  [Reference Citation Analysis (0)]
77.  He W, Leng X, Mao T, Luo X, Zhou L, Yan J, Peng L, Fang Q, Liu G, Wei X, Wang K, Wang C, Zhang S, Zhang X, Shen X, Huang D, Yi H, Bei T, She X, Xiao W, Han Y. Toripalimab Plus Paclitaxel and Carboplatin as Neoadjuvant Therapy in Locally Advanced Resectable Esophageal Squamous Cell Carcinoma. Oncologist. 2022;27:e18-e28.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 40]  [Article Influence: 20.0]  [Reference Citation Analysis (0)]
78.  van den Ende T, de Clercq NC, van Berge Henegouwen MI, Gisbertz SS, Geijsen ED, Verhoeven RHA, Meijer SL, Schokker S, Dings MPG, Bergman JJGHM, Haj Mohammad N, Ruurda JP, van Hillegersberg R, Mook S, Nieuwdorp M, de Gruijl TD, Soeratram TTD, Ylstra B, van Grieken NCT, Bijlsma MF, Hulshof MCCM, van Laarhoven HWM. Neoadjuvant Chemoradiotherapy Combined with Atezolizumab for Resectable Esophageal Adenocarcinoma: A Single-arm Phase II Feasibility Trial (PERFECT). Clin Cancer Res. 2021;27:3351-3359.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 119]  [Article Influence: 39.7]  [Reference Citation Analysis (0)]
79.  Ge F, Huo Z, Cai X, Hu Q, Chen W, Lin G, Zhong R, You Z, Wang R, Lu Y, Huang Q, Zhang H, Song A, Li C, Wen Y, Jiang Y, Liang H, He J, Liang W, Liu J. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 26]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
80.  Shang X, Zhao G, Liang F, Zhang C, Zhang W, Liu L, Li R, Duan X, Ma Z, Yue J, Chen C, Meng B, Ren X, Jiang H. Safety and effectiveness of pembrolizumab combined with paclitaxel and cisplatin as neoadjuvant therapy followed by surgery for locally advanced resectable (stage III) esophageal squamous cell carcinoma: a study protocol for a prospective, single-arm, single-center, open-label, phase-II trial (Keystone-001). Ann Transl Med. 2022;10:229.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 21]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
81.  Duan H, Wang T, Luo Z, Wang X, Liu H, Tong L, Dong X, Zhang Y, Valmasoni M, Kidane B, Almhanna K, Wiesel O, Pang S, Ma J, Yan X. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 25]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
82.  Yang W, Xing X, Yeung SJ, Wang S, Chen W, Bao Y, Wang F, Feng S, Peng F, Wang X, Chen S, He M, Zhang N, Wang H, Zeng B, Liu Z, Kidane B, Seder CW, Koyanagi K, Shargall Y, Luo H, Peng S, Cheng C. Neoadjuvant programmed cell death 1 blockade combined with chemotherapy for resectable esophageal squamous cell carcinoma. J Immunother Cancer. 2022;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 76]  [Article Influence: 38.0]  [Reference Citation Analysis (0)]
83.  Duan H, Shao C, Pan M, Liu H, Dong X, Zhang Y, Tong L, Feng Y, Wang Y, Wang L, Newman NB, Sarkaria IS, Reynolds JV, De Cobelli F, Ma Z, Jiang T, Yan X. Neoadjuvant Pembrolizumab and Chemotherapy in Resectable Esophageal Cancer: An Open-Label, Single-Arm Study (PEN-ICE). Front Immunol. 2022;13:849984.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 21]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
84.  Zheng Y, Li C, Yu B, Zhao S, Li J, Chen X, Li H. Preoperative pembrolizumab combined with chemoradiotherapy for esophageal squamous cell carcinoma: Trial design. JTCVS Open. 2022;9:293-299.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
85.  Li C, Zhao S, Zheng Y, Han Y, Chen X, Cheng Z, Wu Y, Feng X, Qi W, Chen K, Xiang J, Li J, Lerut T, Li H. Preoperative pembrolizumab combined with chemoradiotherapy for oesophageal squamous cell carcinoma (PALACE-1). Eur J Cancer. 2021;144:232-241.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 121]  [Article Influence: 30.3]  [Reference Citation Analysis (0)]
86.  Shang X, Zhang W, Zhao G, Liang F, Zhang C, Yue J, Duan X, Ma Z, Chen C, Pang Q, Liu L, Ren X, Meng B, Zhang P, Ma Y, Zhang L, Li H, Kang X, Li Y, Jiang H. Pembrolizumab Combined With Neoadjuvant Chemotherapy Versus Neoadjuvant Chemoradiotherapy Followed by Surgery for Locally Advanced Oesophageal Squamous Cell Carcinoma: Protocol for a Multicentre, Prospective, Randomized-Controlled, Phase III Clinical Study (Keystone-002). Front Oncol. 2022;12:831345.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 14]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
87.  Zhao Y, Dai Z, Min W, Sui X, Kang H, Zhang Y, Ren H, Wang XJ. Perioperative versus Preoperative Chemotherapy with Surgery in Patients with Resectable Squamous Cell Carcinoma of Esophagus: A Phase III Randomized Trial. J Thorac Oncol. 2015;10:1349-1356.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 31]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
88.  Chen X, Xu X, Wang D, Liu J, Sun J, Lu M, Wang R, Hui B, Li X, Zhou C, Wang M, Qiu T, Cui S, Sun N, Li Y, Wang F, Liu C, Shao Y, Luo J, Gu Y. Neoadjuvant sintilimab and chemotherapy in patients with potentially resectable esophageal squamous cell carcinoma (KEEP-G 03): an open-label, single-arm, phase 2 trial. J Immunother Cancer. 2023;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 8]  [Reference Citation Analysis (0)]
89.  Feo CV, Villaflor VM, Patti MG. Should esophageal resections for cancer be performed in high-volume centers only? Updates Surg. 2011;63:147-150.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
90.  Forschner A, Battke F, Hadaschik D, Schulze M, Weißgraeber S, Han CT, Kopp M, Frick M, Klumpp B, Tietze N, Amaral T, Martus P, Sinnberg T, Eigentler T, Keim U, Garbe C, Döcker D, Biskup S. Tumor mutation burden and circulating tumor DNA in combined CTLA-4 and PD-1 antibody therapy in metastatic melanoma - results of a prospective biomarker study. J Immunother Cancer. 2019;7:180.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 121]  [Article Influence: 24.2]  [Reference Citation Analysis (0)]
91.  Jreige M, Letovanec I, Chaba K, Renaud S, Rusakiewicz S, Cristina V, Peters S, Krueger T, de Leval L, Kandalaft LE, Nicod-Lalonde M, Romero P, Prior JO, Coukos G, Schaefer N. (18)F-FDG PET metabolic-to-morphological volume ratio predicts PD-L1 tumour expression and response to PD-1 blockade in non-small-cell lung cancer. Eur J Nucl Med Mol Imaging. 2019;46:1859-1868.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 48]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
92.  Lv G, Sun X, Qiu L, Sun Y, Li K, Liu Q, Zhao Q, Qin S, Lin J. PET Imaging of Tumor PD-L1 Expression with a Highly Specific Nonblocking Single-Domain Antibody. J Nucl Med. 2020;61:117-122.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 104]  [Article Influence: 20.8]  [Reference Citation Analysis (0)]
93.  Sholl LM. Understanding patterns of pathologic response following neoadjuvant immunotherapy for solid tumors. Ann Oncol. 2018;29:1630-1632.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
94.  Cottrell TR, Thompson ED, Forde PM, Stein JE, Duffield AS, Anagnostou V, Rekhtman N, Anders RA, Cuda JD, Illei PB, Gabrielson E, Askin FB, Niknafs N, Smith KN, Velez MJ, Sauter JL, Isbell JM, Jones DR, Battafarano RJ, Yang SC, Danilova L, Wolchok JD, Topalian SL, Velculescu VE, Pardoll DM, Brahmer JR, Hellmann MD, Chaft JE, Cimino-Mathews A, Taube JM. Pathologic features of response to neoadjuvant anti-PD-1 in resected non-small-cell lung carcinoma: a proposal for quantitative immune-related pathologic response criteria (irPRC). Ann Oncol. 2018;29:1853-1860.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 160]  [Cited by in F6Publishing: 285]  [Article Influence: 57.0]  [Reference Citation Analysis (0)]
95.  Tang H, Jiang D, Zhang S, Zeng Z, Tan L, Hou Y, Wang Q; Zhongshan NCRT-EC working group, Wang H, Zhu J, Shen Y, Yin J, Ge D. Residual tumor characteristics of esophageal squamous cell carcinoma after neoadjuvant chemoradiotherapy. J Thorac Cardiovasc Surg. 2021;162:1632-1641.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 6]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
96.  van Rossum PSN, Fried DV, Zhang L, Hofstetter WL, Ho L, Meijer GJ, Carter BW, Court LE, Lin SH. The value of (18)F-FDG PET before and after induction chemotherapy for the early prediction of a poor pathologic response to subsequent preoperative chemoradiotherapy in oesophageal adenocarcinoma. Eur J Nucl Med Mol Imaging. 2017;44:71-80.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 23]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
97.  Elimova E, Wang X, Etchebehere E, Shiozaki H, Shimodaira Y, Wadhwa R, Planjery V, Charalampakis N, Blum MA, Hofstetter W, Lee JH, Weston BR, Bhutani MS, Rogers JE, Maru D, Skinner HD, Macapinlac HA, Ajani JA. 18-fluorodeoxy-glucose positron emission computed tomography as predictive of response after chemoradiation in oesophageal cancer patients. Eur J Cancer. 2015;51:2545-2552.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 35]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
98.  Simoni N, Rossi G, Benetti G, Zuffante M, Micera R, Pavarana M, Guariglia S, Zivelonghi E, Mengardo V, Weindelmayer J, Giacopuzzi S, de Manzoni G, Cavedon C, Mazzarotto R. (18)F-FDG PET/CT Metrics Are Correlated to the Pathological Response in Esophageal Cancer Patients Treated With Induction Chemotherapy Followed by Neoadjuvant Chemo-Radiotherapy. Front Oncol. 2020;10:599907.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
99.  Fan J, Shen X, Wang Y, Zhou HL, Liu G, Li YL, Xu ZX. Biomarkers for immune checkpoint therapy targeting programmed death 1 and programmed death ligand 1. Biomed Pharmacother. 2020;130:110621.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 8]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
100.  Shah MA, Kojima T, Hochhauser D, Enzinger P, Raimbourg J, Hollebecque A, Lordick F, Kim SB, Tajika M, Kim HT, Lockhart AC, Arkenau HT, El-Hajbi F, Gupta M, Pfeiffer P, Liu Q, Lunceford J, Kang SP, Bhagia P, Kato K. 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;5:546-550.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 300]  [Cited by in F6Publishing: 316]  [Article Influence: 63.2]  [Reference Citation Analysis (0)]
101.  Aguiar PN Jr, De Mello RA, Hall P, Tadokoro H, Lima Lopes G. PD-L1 expression as a predictive biomarker in advanced non-small-cell lung cancer: updated survival data. Immunotherapy. 2017;9:499-506.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 121]  [Cited by in F6Publishing: 145]  [Article Influence: 24.2]  [Reference Citation Analysis (0)]
102.  Paz-Ares L, Spira A, Raben D, Planchard D, Cho BC, Özgüroğlu M, Daniel D, Villegas A, Vicente D, Hui R, Murakami S, Spigel D, Senan S, Langer CJ, Perez BA, Boothman AM, Broadhurst H, Wadsworth C, Dennis PA, Antonia SJ, Faivre-Finn C. Outcomes with durvalumab by tumour PD-L1 expression in unresectable, stage III non-small-cell lung cancer in the PACIFIC trial. Ann Oncol. 2020;31:798-806.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 122]  [Article Influence: 30.5]  [Reference Citation Analysis (0)]
103.  Doroshow DB, Bhalla S, Beasley MB, Sholl LM, Kerr KM, Gnjatic S, Wistuba II, Rimm DL, Tsao MS, Hirsch FR. PD-L1 as a biomarker of response to immune-checkpoint inhibitors. Nat Rev Clin Oncol. 2021;18:345-362.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 203]  [Cited by in F6Publishing: 572]  [Article Influence: 190.7]  [Reference Citation Analysis (0)]
104.  Yagi T, Baba Y, Ishimoto T, Iwatsuki M, Miyamoto Y, Yoshida N, Watanabe M, Baba H. PD-L1 Expression, Tumor-infiltrating Lymphocytes, and Clinical Outcome in Patients With Surgically Resected Esophageal Cancer. Ann Surg. 2019;269:471-478.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 117]  [Article Influence: 23.4]  [Reference Citation Analysis (0)]
105.  Wakita A, Motoyama S, Nanjo H, Sato Y, Yoshino K, Sasaki T, Kawakita Y, Liu J, Imai K, Saito H, Minamiya Y. PD-L1 Expression Is a Prognostic Factor in Patients with Thoracic Esophageal Cancer Treated Without Adjuvant Chemotherapy. Anticancer Res. 2017;37:1433-1441.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 24]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
106.  Tamminga M, de Wit S, Hiltermann TJN, Timens W, Schuuring E, Terstappen LWMM, Groen HJM. Circulating tumor cells in advanced non-small cell lung cancer patients are associated with worse tumor response to checkpoint inhibitors. J Immunother Cancer. 2019;7:173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 69]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
107.  Lipson EJ, Velculescu VE, Pritchard TS, Sausen M, Pardoll DM, Topalian SL, Diaz LA Jr. Circulating tumor DNA analysis as a real-time method for monitoring tumor burden in melanoma patients undergoing treatment with immune checkpoint blockade. J Immunother Cancer. 2014;2:42.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 150]  [Cited by in F6Publishing: 171]  [Article Influence: 17.1]  [Reference Citation Analysis (0)]
108.  Pellini B, Chaudhuri AA. Circulating Tumor DNA Minimal Residual Disease Detection of Non-Small-Cell Lung Cancer Treated With Curative Intent. J Clin Oncol. 2022;40:567-575.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 71]  [Article Influence: 35.5]  [Reference Citation Analysis (0)]
109.  Rosner S, Reuss JE, Zahurak M, Zhang J, Zeng Z, Taube J, Anagnostou V, Smith KN, Riemer J, Illei PB, Broderick SR, Jones DR, Topalian SL, Pardoll DM, Brahmer JR, Chaft JE, Forde PM. Five-Year Clinical Outcomes after Neoadjuvant Nivolumab in Resectable Non-Small Cell Lung Cancer. Clin Cancer Res. 2023;29:705-710.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Reference Citation Analysis (0)]
110.  Xiao BY, Zhang X, Cao TY, Li DD, Jiang W, Kong LH, Tang JH, Han K, Zhang CZ, Mei WJ, Xiao J, Pan ZZ, Li YF, Zhang XS, Ding PR. Neoadjuvant Immunotherapy Leads to Major Response and Low Recurrence in Localized Mismatch Repair-Deficient Colorectal Cancer. J Natl Compr Canc Netw. 2023;21:60-66.e5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 12]  [Reference Citation Analysis (0)]
111.  Park SY, Hong MH, Kim HR, Lee CG, Cho JH, Cho BC, Kim DJ. The feasibility and safety of radical esophagectomy in patients receiving neoadjuvant chemoradiotherapy with pembrolizumab for esophageal squamous cell carcinoma. J Thorac Dis. 2020;12:6426-6434.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 25]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
112.  Huang B, Shi H, Gong X, Yu J, Xiao C, Zhou B, Liang Z, Li X. Comparison of efficacy and safety between pembrolizumab combined with chemotherapy and simple chemotherapy in neoadjuvant therapy for esophageal squamous cell carcinoma. J Gastrointest Oncol. 2021;12:2013-2021.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 19]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
113.  Lee S, Ahn BC, Park SY, Kim DJ, Lee CG, Cho J, Kim JH, Kim HR, Kim YH, Park SR, Chun YJ, Hong MH, Cho B. A phase II trial of preoperative chemoradiotherapy and pembrolizumab for locally advanced esophageal squamous cell carcinoma (ESCC). Ann Oncol. 2019;30:754.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 5]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
114.  Qiao Y, Zhao C, Li X, Zhao J, Huang Q, Ding Z, Zhang Y, Jiao J, Zhang G, Zhao S. Efficacy and safety of camrelizumab in combination with neoadjuvant chemotherapy for ESCC and its impact on esophagectomy. Front Immunol. 2022;13:953229.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 13]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
115.  Xu WL, Jiang YH, Wang CC, Wu J, Li JQ, Hu YQ, Lu WS, Shen DJ, Wang YJ, Chen QX. The efficacy and safety of neoadjuvant camrelizumab and chemotherapy for locally advanced thoracic esophageal squamous cell carcinoma. J Clin Oncol. 2022;40:278.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
116.  Yang G, Su X, Yang H, Luo G, Gao C, Zheng Y, Xie W, Huang M, Bei T, Bai Y, Wang Z, Cai P, He H, Xiang J, Cai M, Zhang Y, Qu C, Fu J, Liu Q, Hu Y, Zhong J, Huang Y, Guo Q, Zhang X. Neoadjuvant programmed death-1 blockade plus chemotherapy in locally advanced esophageal squamous cell carcinoma. Ann Transl Med. 2021;9:1254.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 29]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
117.  Wang Z. Neoadjuvant camrelizumab combined with chemotherapy and apatinib for locally advanced thoracic esophageal squamous cell carcinoma (ESCC): A single-arm, open-label, phase Ib study. J Clin Oncol. 2021;39:4047.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 4]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
118.  Ma JQ, Zhang JF, Yang YN, Zheng DY, Wang XY, Liang H, Zhang LQ, Xin YZ, Ling XD, Fang CY, Jiang H, Zheng W. Camrelizumab combined with paclitaxel and nedaplatin as neoadjuvant therapy for locally advanced esophageal squamous cell carcinoma (ESPRIT): A phase II, single-arm, exploratory research. J Clin Oncol. 2021;39:e16033.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
119.  Wang F, Yu Q, Meng XG, Fan QX. Camrelizumab in combination with preoperative chemotherapy for locally advanced esophageal squamous cell carcinoma: A single-arm, open-label, phase II study. J Clin Oncol. 2021;39:222.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 4]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
120.  Zhang Y, Shen GY, Xu RY, Huang GZ, Yang SS, Zheng QF, Huang ZJ, Duan HB, Ma LY, Yang LB, Yi YF, Liu RX, Li KZ, Chen SG. Real-world effectiveness and safety of camrelizumab-based neoadjuvant therapy in resectable esophageal cancer: Initial results of a prospective multicenter observational study. J Clin Oncol. 2022;40:250.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
121.  Zhang Z, Ye J, Li H, Gu D, Du M, Ai D, Chen W, Fang Y, Xu X, Bai C, Zhao K, Zhou G. Neoadjuvant sintilimab and chemotherapy in patients with resectable esophageal squamous cell carcinoma: A prospective, single-arm, phase 2 trial. Front Immunol. 2022;13:1031171.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 14]  [Reference Citation Analysis (0)]
122.  Xing W, Zhao L, Zheng Y, Liu B, Liu X, Li T, Zhang Y, Ma B, Yang Y, Shang Y, Fu X, Liang G, Yuan D, Qu J, Chai X, Zhang H, Wang Z, Lin H, Liu L, Ren X, Zhang J, Gao Q. The Sequence of Chemotherapy and Toripalimab Might Influence the Efficacy of Neoadjuvant Chemoimmunotherapy in Locally Advanced Esophageal Squamous Cell Cancer-A Phase II Study. Front Immunol. 2021;12:772450.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 40]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
123.  Xing W, Zhao L, Fu X, Liang G, Zhang Y, Yuan D, Li Z, Gao Q, Zheng Y; written on Henan Cancer Hospital Thoracic Oncology Group (HCHTOG). A phase II, single-centre trial of neoadjuvant toripalimab plus chemotherapy in locally advanced esophageal squamous cell carcinoma. J Thorac Dis. 2020;12:6861-6867.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 10]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
124.  Liu DL, Zhang Q, Zhu JH, Qian T, Yin R, Fan ZH, Jiang F, Li HH, Lu JW, Cao GC. Phase-II study of toripalimab combined with neoadjuvant chemotherapy for the treatment of resectable esophageal squamous cell carcinoma. J Clin Oncol. 2021;39:e16029.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
125.  Gao L, Lu J, Zhang P, Hong ZN, Kang M. 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;13:478-487.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 19]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
126.  Jiang N, Jiang M, Zhu XZ, Ren BH, Zhang JY, Guo Z, Wu YA, Song X, Zhao LJ, Kong C, Cao GC, Qian PD, Zhao Y, Li S, Luo SF. SCALE-1: Safety and efficacy of short course neoadjuvant chemo-radiotherapy plus toripalimab for locally advanced resectable squamous cell carcinoma of esophagus. J Clin Oncol. 2022;40:4063.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
127.  Hong ZN, Zhang Z, Chen Z, Weng K, Peng K, Lin J, Kang M. Safety and feasibility of esophagectomy following combined neoadjuvant immunotherapy and chemotherapy for locally advanced esophageal cancer: a propensity score matching. Esophagus. 2022;19:224-232.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 6]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
128.  Yang Y, Zhu L, Cheng Y, Liu Z, Cai X, Shao J, Zhang M, Liu J, Sun Y, Li Y, Yi J, Yu B, Jiang H, Chen H, Yang H, Tan L, Li Z. Three-arm phase II trial comparing camrelizumab plus chemotherapy versus camrelizumab plus chemoradiation versus chemoradiation as preoperative treatment for locally advanced esophageal squamous cell carcinoma (NICE-2 Study). BMC Cancer. 2022;22:506.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 18]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]