Abstract
Esophageal cancer (EC) remains a public health concern with a high mortality and disease burden worldwide. Esophageal squamous cell carcinoma (ESCC) is a predominant histological subtype of EC that has unique etiology, molecular profiles, and clinicopathological features. Although systemic chemotherapy, including cytotoxic agents and immune checkpoint inhibitors, is the main therapeutic option for recurrent or metastatic ESCC patients, the clinical benefits are limited with poor prognosis. Personalized molecular-targeted therapies have been hampered due to the lack of robust treatment efficacy in clinical trials. Therefore, there is an urgent need to develop effective therapeutic strategies. In this review, we summarize the molecular profiles of ESCC based on the findings of pivotal comprehensive molecular analyses, highlighting potent therapeutic targets for establishing future precision medicine for ESCC patients, with the most recent results of clinical trials.
Keywords: chemotherapy, esophageal cancer, esophageal squamous cell carcinoma, immunotherapy, molecular-targeted therapy
Introduction
Esophageal cancer (EC) is the seventh most common cancer and the sixth leading cause of cancer-related deaths worldwide.1 Multimodal treatment with curative intent, combined with surgery, chemotherapy, and/or radiotherapy, has been developed for patients with locally advanced EC.2 However, in addition to the high recurrence rate, EC is still often diagnosed at advanced stages that are not amenable to curative treatment due to the lack of early clinical signs. Although systemic chemotherapy is the main therapeutic option for patients with recurrent or metastatic EC, the prognosis is poor, with a 5-year overall survival (OS) rate of less than 5%.3 Therefore, further development of novel agents is required to improve prognostic outcomes.
EC can be histologically classified as either esophageal squamous cell carcinoma (ESCC) or esophageal adenocarcinoma (EAC), which have distinct epidemiology, molecular profiles, and clinical features.3 –6 ESCC is the most prevalent histological type, accounting for approximately 85% of ECs.7 Geographically, ESCC is the most common histology in Asian and Eastern Europe, while EAC is more common in Northern Europe and Northern America.7 Molecularly, ESCCs share many genetic and epigenetic aberrations specific to the squamous cell lineage,8 –13 while EACs mostly resemble the chromosomal instability (CIN) molecular subtype based on The Cancer Genome Atlas (TCGA) molecular classification of gastric adenocarcinoma.13,14 Clinically, ESCC is associated with a higher sensitivity to radiotherapy,15 and exhibits a higher prevalence of lymphatic spread with poorer survival outcomes than EAC.3,16,17 Thus, ESCC and EAC must be considered separate entities, indicating the need for different therapeutic strategies.
Many molecular-targeted agents have failed to demonstrate significantly improved OS in clinical trials for patients with recurrent or metastatic ESCC, partially due to a lack of selective biomarkers and/or intratumoral heterogeneity. Currently, programmed death-1 (PD-1) is the only clinically validated targeted molecule in ESCC, and immune checkpoint inhibitors (ICIs) targeting PD-1 have changed therapeutic paradigms dramatically because of the durable clinical response in ESCC.18 –21 However, it has clinical benefits for a limited number of ESCC patients, with an overall response rate (ORR) of 16.7–20.3% in monotherapy. Considerable research effort has been invested in characterizing the genomic landscape of ESCC and identifying potential therapeutic targets for precision medicine.8,10,12,13,22 –26
In this review, we summarize the biology and current treatment of ESCC, and provide state-of-the-art knowledge on molecular profiles based on comprehensive molecular analyses. In addition, significant differences to EAC are highlighted where appropriate in terms of clinicopathological and molecular features. Finally, we discuss potential therapeutic targets from both basic and clinical viewpoints.
Clinicopathological and molecular features of ESCC
According to histological classification, EC is mainly divided into two subtypes (ESCC and EAC), which have distinct clinicopathological features and molecular profiles.
Clinicopathological features of ESCC
Clinicopathological features according to histological subtypes are shown in Table 1. ESCC is still the major histological subtype of EC.3,7 The predominant incidence rates of the ESCC subtype are generally observed in developing countries with low socioeconomic status, and risk factors include consumption of tobacco, alcohol, and hot beverages. ESCC develops from the lining of the esophageal squamous epithelium via precursor dysplastic lesions in a multi-step process when exposed to tumorigenesis.27,28 In contrast to the declining incidence rates of ESCC due to economic gains, dietary improvements, and smoking cessation,1,17,29 the rapidly increasing rates of EAC have been attributed to gastroesophageal reflux disease, and obesity in highly developed countries.3,6,7,30 EAC typically originates from Barrett’s mucosa, triggered by persistent gastroesophageal reflux, in which the normal squamous epithelium of the esophagus is replaced by a metaplastic, columnar, or glandular epithelium.6,30,31
Table 1.
Characteristics | Squamous cell carcinoma | Adenocarcinoma |
---|---|---|
Proportion | 85% | 15% |
Incidence trend | Declining | Increasing |
Geography based on the number of estimated cases | Eastern Asia, followed by South-Central Asia, Sub-Saharan Africa, Northern/Western Europe, and South America | Eastern Asia, followed by Northern America, Northern/Western Europe, and South-Central Asia |
Geography based on the incidence rates according to the histological subtypes | Asia, followed by sub-Saharan Africa, Eastern/Western Europe, and South America | Northern Europe, followed by Northern America and Oceania |
Socioeconomic status | Low | High |
Risk factors | Tobacco, alcohol, hot beverages/foods, and regional micronutrient deficiency | Obesity, acid or bile reflux, Barrett’s esophagus, and tobacco consumption |
Race | Black > Caucasian | Caucasian > Black |
Median age | Younger (median 53 years) | Older (median 63 years) |
Location of primary tumor | Upper two-thirds of the esophagus | Lower third of the esophagus and esophagogastric junction |
Precursor lesion | Squamous dysplasia | Barrett’s esophagus |
Radiosensitivity | High | Low |
Survival trend | Stable | Improving |
In the Surveillance, Epidemiology, and End Results (SEER) database of 93,167 patients diagnosed with EC between 1973 and 2009,16 the median OS of EAC improved steadily. However, ESCC remained relatively stable after the 1990s (median OS of 8 months) and was one of the independent unfavorable factors associated with the OS of EC in multivariable Cox regression analyses, with adjustment for relevant clinicopathologic factors and treatment. The median OS was only 10 months for ESCC, even in a recent SEER database of 37,723 patients diagnosed with EC between 2004 and 201532. ESCC also more easily tends toward lymph node metastasis than EAC,17 and the status of metastatic lymph nodes is characteristic of a life-threatening phenotype of ESCC.33 Thus, ESCC cells have aggressive traits, which results in poor prognosis.
In a phase III CROSS trial of neoadjuvant chemoradiotherapy (CRT) plus surgery versus surgery alone for EC or esophagogastric junction,15 neoadjuvant CRT plus surgery showed a significantly favorable OS compared with surgery alone.15 The survival benefit of neoadjuvant CRT was more clinically relevant for patients with ESCC than for those with EAC. In addition, a pathological complete response (pCR) was significantly higher in ESCC than in EAC.15 The high rates of pCR in ESCC versus EAC were also observed in a large cohort study of 895 EC patients who underwent neoadjuvant CRT followed by surgery.34 In contrast, the treatment efficacy of neoadjuvant chemotherapy was similar for both ESCC and EAC in a randomized OEO2 trial.35 Thus, it is evident that ESCC benefits significantly from radiotherapy.15
Aberrant signaling pathways in ESCC
Aberrant molecular profiles during ESCC development
Comprehensive molecular analyses using sequential lesions (normal, precursor lesions, and tumors) derived from the same patient provide the process of ESCC evolution because of their identical germline backgrounds. In physiologically normal esophageal epithelium, clones carrying driver mutations emerge multifocally as early as early infancy.36 These clones expand with aging, which is accelerated by heavy smoking and drinking. In whole-exome sequencing (WES) analyses on multiple samples covering different stages from dysplastic lesions to ESCC derived from the same patients, dysplastic lesions shared most identical mutational characteristics with their matched ESCCs.37 Copy number alterations (CNAs) were also similar between dysplastic lesions and ESCC.37 –39 Importantly, the apolipoprotein B mRNA editing enzyme catalytic polypeptide-like (APOBEC) signatures contribute to mutagenic processes due to their deaminase activity, which confers progression of esophageal normal clones with driver mutations to a more advanced neoplasm with more genomic alterations.9,10,13,22,24,36,38 –41 In fact, a number of truncal/clonal mutations were significantly correlated with the burden of APOBEC mutations in ESCC, indicating that APOBEC-driven mutagenesis is a crucial step in ESCC development.23,24 A phylogeny analysis based on multiregional WES showed that driver mutations had a tendency to be truncal/clonal compared with passenger mutations.23,39 However, approximately 40% of driver mutations were branched/subclonal, indicating relatively late events during the development of ESCC. Most truncal/clonal driver mutations occurred in tumor suppressor genes, such as TP53, lysine methyltransferase 2D (KMT2D, also called MLL2), and zinc finger protein 750 (ZNF750), while the branched/subclonal mutations involved several key oncogenes, such as members of the phosphoinositide 3-kinase (PI3K)/mammalian target of the rapamycin (mTOR) pathway [phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) and MTOR], and the nuclear factor, erythroid 2-like 2 (NRF2) pathway [NFE2 like bZIP transcription factor 2 (NFE2L2) and Kelch-like ECH-associated protein 1 (KEAP1)].23,39 Collectively, the transition from normal squamous epithelium to precursor dysplastic lesions and subsequent ESCC is developed through a process of positive selection and the acquisition of driver mutations, and most of the significantly mutated genes (SMGs) and recurrent CNAs in ESCC are already prevalent alterations in dysplastic lesions neighboring ESCC.
It is well established that the development of EAC is triggered by gastroesophageal reflux and has a stepwise progression of Barrett’s esophagus (BE) to low-grade dysplasia, high-grade-dysplasia, and then EAC.6,30,31,42 The major pathway by which BE develops into EAC is through early TP53 inactivation, which causes genome doubling and facilitates genomic instability, aneuploidy, and oncogene amplification in late events of EAC.43,44 Unlike in ESCC, the spectrum of mutations generally has little overlap between EAC and adjacent BE. Thus, there is different timing between ESCC and EAC for the acquisition of genetic alterations.37
Aberrant molecular profiles in ESCC
Developments in high-throughput genomic technologies have led to a better understanding of the molecular profiles of ESCC and EAC (Table 2). ESCC and EAC exhibit almost mutually exclusive alterations of driver genes.9 ESCCs share multiple SMGs exclusively with SCCs of other tissues, including the head and neck, lung, cervix, and skin, but differ from that of EAC despite the anatomical overlap.8 –13 Although DNA methylation is much less frequent in ESCC than in EAC,13 the SCC histology-driven epigenetic patterns were also similar across organs.11 In contrast, SMGs of EAC closely resemble those of the CIN molecular subtype of stomach adenocarcinoma.13 Thus, two histological subtypes of EC should be considered distinct disease entities, and SCCs that originate from developmentally similar cell lineages may have common pathogenic mechanisms, regardless of their location in different parts of the body. However, ESCC remains a heterogeneous disease with diverse genetic and molecular levels.13,45,46 The complexity of spatial and temporal heterogeneity may be associated with ESCC aggressiveness.23,39,47 In ESCC, several aberrant molecules were categorized according to their functions and pathways (Table 2): (i) receptor tyrosine kinase (RTK)/RAS/PI3K, (ii) cell cycle, (iii) cell adhesion, (iv) cell differentiation and proliferation, (v) chromatin modification, and (vi) oxidative stress response.8 –10,12,13,22,24,41,48
Table 2.
ESCC | EAC | ||
---|---|---|---|
TCGA molecular subtypes | SCC (ESCC1-3) | CIN | |
Molecules | Signaling pathway | Frequency of genetic aberrations (%) | |
RTK/RAS/MAPK and PI3K | |||
EGFR | RTK | 6–26% | 7–18% |
HER2 | RTK | 2–6% | 15–32% |
HER3 | RTK | 1–3% | 3–6% |
HER4 | RTK | 2–13% | 10–12% |
IGF1R | RTK | 2–5% | 1–10% |
FGFR1 | RTK | 11–31% | 2–4% |
MET | RTK | 1–5% | 4–8% |
KRAS | MAPK/PI3K pathways | 4–15% | 13–24% |
PIK3CA | PI3K pathway | 9–45% | 3–10% |
mTOR | PI3K pathway | 2–10% | 6–13% |
PTEN | PI3K pathway | 2–11% | 0–8% |
VEGFA | Angiogenesis | 0–3% | 5–28% |
Cell cycle | |||
TP53 | Regulator of cell cycle | 69–94% | 50–80% |
CDKN2A | Regulator of cell cycle | 15–76% | 13–76% |
CCND1 | Regulator of cell cycle | 33–59% | 10–17% |
CCNE1 | Regulator of cell cycle | 4–7% | 10–14% |
CDK4 | Regulator of cell cycle | 0–4% | 1–3% |
CDK6 | Regulator of cell cycle | 14–23% | 7–17% |
RB1 | Regulator of cell cycle | 2–14% | 0–2% |
MDM2 | Regulator of cell cycle | 2–25% | 5–6% |
Cell adhesion and proliferation | |||
AJUBA | Regulator of cell adhesion | 4–7% | 3% |
FAT1 | Regulator of cell adhesion | 5–16% | 9–13% |
CTNNB1 | Wnt/β-catenin pathway | 0–1% | 2–4% |
AXIN1 | Wnt/β-catenin pathway | 0–2% | 4% |
APC | Wnt/β-catenin pathway | 1–2% | 3–19% |
MYC | Transcription factor | 5–44% | 12–32% |
SMAD4 | TGFβ pathway | 1–8% | 13–59% |
SMAD2 | TGFβ pathway | 0–2% | 7–30% |
PTCH1 | Hedgehog pathway | 3–10% | 0–5% |
Cell differentiation | |||
SOX2/TP63 | Transcription factor | 18–49% | 1–12% |
NOTCH1 | Transcription factor | 7–33% | 0–3% |
FBXW7 | Transcription factor | 4–18% | 0–6% |
ZNF750 | Transcription factor | 7–15% | 0–8% |
GATA4 | Transcription factor | 0–1% | 15–19% |
GATA6 | Transcription factor | 0–3% | 14–21% |
YAP1 | Co-transcription factor | 4–6% | 5% |
Chromatin modification | |||
KDM6A | Histone modifiers | 6–19% | 2–4% |
KMT2D | Histone modifiers | 12–31% | 1–13% |
KMT2C | Histone modifiers | 4–8% | 10–25% |
CREBBP | Histone modifiers | 2–14% | 5% |
EP300 | Histone modifiers | 8–18% | 0–5% |
ARID1A | SWI/SNF | 1–3% | 8–18% |
SMARCA4 | SWI/SNF | 5–10% | 7–17% |
PBRM1 | SWI/SNF | 2–5% | 6% |
Oxidative stress response | |||
NFE2L2 | NRF2 pathway | 10–36% | 0–1% |
KEAP1 | NRF2 pathway | 2–13% | 5% |
CUL3 | NRF2 pathway | 2–5% | 3% |
Immune check points | |||
PD-L1 CPS ≥ 1018,21,53,54 | Immune check points | 31–52% | 25–48% |
cBioPortal for Cancer Genomics (https://www.cbioportal.org/) was also referred.
CIN, chromosomal instability; CPS, combined positive score of PD-L1; EGFR, epidermal growth factor receptor; PTEN, phosphatase and tensine homolog; TCGA, The Cancer Genome Atlas.
RTKs trigger the activation of PI3K/v-akt murine thymoma viral oncogene homolog (Akt) and the mitogen-activated protein kinase (MAPK) signaling pathway, which play crucial roles in tumorigenesis, proliferation, survival, angiogenesis, and metastasis. EAC has a wider range of genetic alterations of RTK/RAS signaling pathways, including human epidermal growth receptor 2 (HER2), KRAS, and vascular endothelial growth factor A (VEGFA), but these are rare events in ESCC.13,25,26,49 –51,55 In fact, the frequency of HER2 amplifications was reported in only 2–6% of ESCCs but 15–32% of EACs.8,10,12,13,22 –26,48 –52,56 In 1391 patients with ECs, including 215 ESCCs and 1176 EACs, HER2 overexpression was observed in 13% of EACs and 1% of ESCCs.55 Among RTKs, epidermal growth factor receptor (EGFR) and fibroblast growth factor receptor 1 (FGFR1) serve as potential therapeutic targets in ESCC because of their frequent gene amplifications. The PI3K signaling pathway is also activated via genetic alterations of PIK3CA and MTOR and the loss of phosphatase and tensine homolog expression occurs in approximately 60% of ESCCs.22
Cyclin D1 encoded by CCND1 can form a complex with cyclin-dependent kinase 4 and 6 (CDK4/6), which may lead to abrogation of both retinoblastoma protein (Rb) and p53 function, resulting in disruption of cell cycle restriction, genome instability, and tumorigenesis.57 Cyclin-dependent kinase inhibitor 2A (CDKN2A) blocks the binding of cyclin D1 to CDK4/6 as a tumor suppressor gene. The cell cycle pathway is dysregulated in almost all ESCCs by mutations or methylations of TP53, CDKN2A, and RB transcriptional corepressor 1 (RB1), and amplifications of CCND1, CDK4/6, and MDM2.10,12,13,24,48,58
Cell adhesion is dysregulated mainly by mutations in FAT atypical cadherin 1 (FAT1) and AJUBA in ESCC.9,10,12,13,22,24,41,48 FAT1 is a member of a cadherin superfamily and regulates cell–cell adhesion, proliferation, migration, and actin dynamics as a tumor suppressor gene.59 The function of FAT1 is frequently disrupted by FAT1 mutation, accounting for 5–16% of ESCCs.10,22,24 In ESCC cell line models, knockdown of FAT1 suppresses cell adhesive force and accelerates cell migration and invasion through activation of the Yes1-associated transcriptional regulator (YAP1) in the Hippo signaling pathway.60,61 AJUBA functions as a scaffold that is implicated in the assembly of multiple protein complexes to regulate cell adhesion, cytoskeletal organization, migration, mitosis, microRNA maturation, and cell differentiation.62,63 Mutation of AJUBA also leads to YAP1 activation by inhibiting large tumor suppressor kinases 1/2 (LATS1/2) in the Hippo pathway.64 Catenin beta 1 (CTNNB1) encodes a β-catenin protein that acts as an essential part of the Wnt signaling pathway. The β-catenin promotes adherens junction formation by binding to E-cadherin, but it can also promote cell proliferation, differentiation, and epithelial–mesenchymal transition (EMT).65 Although genetic aberration of CTNNB1 is rare, altered genes in the Wnt/β-catenin pathway have been reported in 26–86% of ESCCs, including AJUBA and FAT1.24,25,48,66 In addition, this pathway is activated by hypermethylation of its negative regulators66 or methylation-mediated de-repression of WNT2.67 MYC proto-oncogene is a transcription factor acting as a master regulator of genes involved in cell cycle progression, cell proliferation, differentiation, and apoptosis, and it can be activated by several pathways, such as RTK/RAS, Wnt/β-catenin, Notch, and transforming growth factor-β (TGF-β) signaling.68 MYC amplification has been reported in 43% of ESCCs,8 and its overexpression detected in 61% of ESCCs.69 In ESCC, MYC contributes not only to cell proliferation,70 but also to the driver of cancer stem-like cells (CSCs)71 and intrinsic resistance to chemoradiotherapy.72
Squamous cell differentiation is controlled by lineage-specific transcription factors (TFs), including SRY-box transcription factor 2 (SOX2), tumor protein P63 (TP63), ZNF750, and Notch family members.8 –13,58 TP63 and SOX2 are crucial core TFs that orchestrate gene regulatory networks for chromatin accessibility, epigenetic modifications, and gene expression patterns in ESCC.73 SOX2 and TP63 genes are localized on chromosome 3q, which are coamplified in ESCC.13,74 TP63 and SOX2 exhibited overlapping genomic occupancy and cooperatively promoted SCC tumorigenesis and progression through activation of the MAPK and PI3K/Akt signaling pathways.73,75 –77 ZNF750 is a gene related to keratinocyte terminal differentiation and is frequently mutated in ESCC due to APOBEC-driven mutagenesis.13,22 –24 ZNF750 acts as a tumor suppressor by regulating long non-coding RNA and cell cycle activator E2F transcription factor 2 (E2F2).78 –80 Notch signaling regulates squamous differentiation in a normal esophagus.81 The Notch pathway was exclusively activated by recurrent mutations of Notch pathway-related genes, including NOTCH1, NOTCH3, and F-box and WD repeat domain-containing-7 (FBXW7) in ESCC but not in EAC.12,24,26,48,82 The Notch pathway is also activated through histone H4 arginine methylation.83 The activated Notch pathway facilitates not only EMT, but also tumor initiation and CSC phenotype in concert with TGF-β signaling in the tumor microenvironment (TME).84 Importantly, these TFs cooperate to dysregulate the squamous differentiation program, such as the interaction between NOTCH1 and TP6385 and between TP63 and ZNF750.79
Dysregulation of chromatin modification affects the development of both ESCC and EAC. Histone-modifying factors, including KMT2D, lysine methyltransferase 2C (KMT2C), lysine demethylase 6A (KDM6A), CREB binding protein (CREBBP), and E1A binding protein p300 (EP300), were frequently altered in ESCC, while alterations of Switch/sucrose nonfermentable (SWI/SNF)-encoding genes, including AT-rich interaction domain 1A (ARID1A) and SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin, subfamily a, member 4 (SMARCA4), were more common in EAC.12,13,22,24,41
Oxidative stress plays diverse and important roles in carcinogenesis, metabolic reprogramming, aggressive cancer phenotypes, and drug resistance.86 The NFE2L2 gene encodes the NRF2 protein, which is a master transcriptional regulator that induces cytoprotective proteins upon oxidative stress.87 The NRF2 signaling pathway is composed of NF2EL2, KEAP1, and cullin 3 (CUL3) genes, which exhibit mutually exclusive mutations in a manner specific to squamous cell lineage.9,12,13,24,88 Gain-of-function mutations of the NFE2L2 gene confer resistance to stressors, including antitumor therapy, by promoting tumorigenic metabolism.8,87 Loss-of-function mutations of the KEAP1 and CUL3 genes also activate the NRF2 pathway by suppressing ubiquitination of NRF2.9 Thus, ESCCs have activity in the antioxidative pathway.
The most frequently dysregulated biochemical pathways are the cell cycle pathway, followed by chromatin remodeling, Notch, PI3K, Wnt/β-catenin, NRF2, and Hippo pathways in ESCC.8,10 –13,22 –24,48 Dysregulation of the TGF-β pathway, MAPK pathway, E-cadherin signaling, and the SWI/SNF chromatin remodeling complexes are rare in ESCC, but not in EAC.13,51,55
Although cancer genes and biology are dynamically changed through tumor progression, most comprehensive molecular profiles of ESCC were analyzed using the disease without distant metastases.8,10,12,13,22 –26,48 –52 There is a possibility of selection bias that hampers an in-depth understanding of the associations with advanced-stage ESCC. Circulating tumor DNA sequencing is likely a strict approach to reveal the genomic landscape of metastatic ESCC with spatial heterogeneity.89
Biological features of ESCC
Cancer cells are extremely heterogeneous in terms of their malignant potential, drug sensitivity, and potential to metastasize, and relatively rare CSCs are at the top of the cellular hierarchy. CSCs have stemness properties, such as sphere formation, self-renewal, invasion, and cell lineage differentiation, which contribute to the driving force of tumorigenesis and metastasis.90 –92 CSCs are resistant to conventional chemotherapies that eliminate proliferating non-CRCs, not only via activation of drug efflux, DNA damage repair (DDR), drug-resistant proteins, and certain tumorigenic signaling pathways, but also by evading apoptosis and programmed cell death.93 Furthermore, cytotoxic chemotherapies stimulate non-CRCs to gain stemness traits.94 In ESCC, there is accumulating evidence that the biological function of CSCs is activated by various critical signaling pathways, such as Wnt/β-catenin, TGF-β, PI3K, Notch, Hedgehog, YAP1, and Janus kinase (JAK)/signal transducer and activator of transcription 3 (STAT3).48,71,95 –99 In an ESCC patient-derived xenograft model, chemoresistant tumors had high proportions of CSCs with CD90+/CD271+ in line with the overexpression of stemness-associated genes, such as SOX2, OCT4, and ATP-binding cassette superfamily G member 2 transporter (ABCG2), through Wnt/β-catenin and TGF-β signaling pathways.95 CSCs isolated from the ESCC cell line were resistant to genotoxicity through slow-cycling status and avoidance of apoptosis or senescence.100 In addition, a single-cell transcriptome sequencing analysis of ESCC cells with high stemness traits showed high expression of genes related to DNA replication and DDR.101 Thus, the elimination of CSCs is crucial for achieving long-term therapeutic efficacy in ESCC.
EMT represents a morphogenetic process of redifferentiation of epithelial cells into mesenchymal ones, which is implicated in acquiring migratory and invasive abilities, CSC traits, metastatic potential, and chemoresistance as the hallmarks of aggressive phenotype.102 In ESCC, EMT may be promoted by the activation of TGF-β, STAT3, Wnt/β-catenin, Notch, and Hedgehog.84,98,103 –106 ESCC xenograft models showed that TGF-β drove Notch-mediated EMT via activation of the transcription factor Zinc finger E-Box binding homeobox 1 (ZEB1) and concurrently generated CSCs.84 Tumor necrosis factor-associated apoptosis-inducing ligand (TRAIL) promoted EMT via STAT3-dependent expression of programmed death-ligand 1 (PD-L1), an inhibitory immune checkpoint molecule.106 Epigenetic alterations, including regulatory non-coding RNAs (ncRNAs), interact with numerous TFs and act as crucial mechanisms of EMT. The ncRNAs have two types of long non-coding RNAs (lncRNAs) and microRNAs (miRNAs), and these execute a broad repertoire of cellular processes. Emerging studies demonstrate the effects of ncRNAs on EMT, including HOTTIP/miR-30b/Snail axis,107 lncRNA-TTN-AS1/miRNA-133b/Snail axis,108 and miRNA-31/Hippo signaling axis,109 in ESCC.
The TME is composed of diverse cell types, including cancer-associated fibroblasts (CAFs), immune cells, vascular endothelial cells, and stem cells, which play a central role in proliferation, invasion, metastasis, angiogenesis, and chemoresistance in EC.103 As an essential component of the TME, CAFs contribute to EMT as well as angiogenesis through TGF-β and STAT3 pathways in ESCC.98,105 The interaction between ESCCs and CAFs dysregulates the expression of multiple genes encoding growth factors, cytokines, chemokines, matricellular proteins, EMT-related molecules, and components of inflammatory signaling pathways.110 Furthermore, TME creates a CSC niche that is made up of stromal cells, blood vessels, soluble factors, and extracellular matrix proteins, which not only maintains stemness properties, but also creates immune evasion.111,112
Collectively, the aggressive phenotypes of ESCC may be partially explained by the biological features of EMT and CSC phenotypes, establishing a preclinical rationale to target molecules that play a pivotal role in EMT and CSCs in terms of antitumor efficacy and immunity.
Treatment of patients with ESCC
ESCC patients are often diagnosed at advanced stages and are not eligible for curative treatment, and more than half of all patients who are treated with curative intent develop tumor recurrence.6 For such patients, systemic chemotherapy is the main therapeutic option for prolonging survival and improving symptoms and quality of life. Systemic chemotherapy includes two types of treatment: (i) cytotoxic agents, including fluoropyrimidines, platinum compounds, taxanes, and irinotecan, and (ii) ICIs targeting PD-1.
Cytotoxic chemotherapy
The impact of cytotoxic chemotherapy on ESCC has been evaluated in clinical trials (Table 3). Platinum-based chemotherapy remains the backbone of treatment in the first-line setting.2 However, the treatment efficacy of platinum-based doublet chemotherapy is generally modest, with reported ORR of 15–56% and median progression-free survival (PFS) of 4–7 months. The cytotoxic effect of platinum compounds has been attributed to the formation of platinum DNA adducts. Excision repair cross-complementing 1 (ERCC1) is a key rate-limiting enzyme in the nucleotide excision repair system, which can repair these adducts and is consequently associated with platinum resistance.113 The expression of ERCC1 was higher in ESCC than in EAC.55 A pooled analysis demonstrated the status of ERCC1 expression as a valuable predictive biomarker for platinum-based chemotherapy in ESCC, in which the median ORR was 77.5% and 39.3% for patients with low and high expression of ERCC1, respectively.114
Table 3.
Clinical trials of cytotoxic agents in advanced esophageal squamous cell carcinoma.
Regimen | Line | Phase | Histology | No. of pts (No. of ESCC) | ORR (%) | PFS (months) | OS (months) | Ref. |
---|---|---|---|---|---|---|---|---|
CDDP plus 5-FU | ≥1 | II | SCC | 39 | 36 | – | 9.5 | Iizuka et al.115 |
CDDP plus 5-FU versus CDDP | 1 | II | SCC | 88 | 35 versus 19 | – | 8.3 versus 7.0 | Bleiberg et al.116 |
PTX plus CDDP | 1 | II | SCC | 39 | 48.6 | 7 | 13 | Zhang et al.117 |
PTX plus CDDP | 1 | II | SCC | 46 | 56.5 | 5.6 | 17.0 | Huang et al.118 |
PTX plus CBDCA | 1 | II | SCC/AC | 35 (13) | 43 | 3.4* | 9 | El-Rayes et al.119 |
DTX plus CDDP | 1 | II | SCC | 39 | 33 | 5.0 | 8.3 | Kim et al.120 |
GEM plus CDDP | 1 | II | SCC | 38 | 42.1 | 4.1 | 10 | Huang et al.121 |
CPT-11 plus CDDP | 1 | II | SCC | 32 | 31.3 | 4.4 | 9.6 | Lee et al.122 |
CPT-11 plus CDDP | 1 | II | SCC | 27 | 30.0 | 4.5 | 8.8 | Kim et al.123 |
PTX plus L-OHP | 1 | I/II | SCC/AC | 26 (19) | 15 | 4.5 | 12.3 | Kaechele et al.124 |
5-FU plus LV plus L-OHP (FOLFOX) | 1 | II | SCC | 56 | 23.2 | 4.4 | 7.7 | Wang et al.125 |
CPT-11 plus 5FU (AIO) | 1 | II | SCC/AC | 24 (11) | 33 | 6.6** | 13.6 (10.0 for SCC) | Wolff et al.126 |
PTX plus CDDP plus 5-FU | 1 | II | SCC/AC | 61 (31) | 48 | – | 10.8 | Ilson et al.127 |
DTX plus CDDP plus 5-FU | 1 | I/II | SCC/OTH | 55 | 62 | 5.8 | 11.1 | Hironaka et al.128 |
DTX plus CDDP plus 5-FU | 1 | I/II | SCC | 48 | 62.5 | 6 | 13 | Ojima et al.129 |
DTX | ≥1 | II | SCC/AC/OTH | 52 (46) | 20 | – | 8.1 | Muro et al.130 |
PTX | ≥2 | II | SCC/AC | 53 (52) | 44.2 | 3.9** | 10.4 | Kato et al.131 |
CPT-11 | ≥1 | II | SCC/AC | 13 (10) | 22.2 | 3.8** | 6.1 | Mühr-Wilkenshoff et al.132 |
CPT-11 | 2 | II | SCC/AC | 14 (7) | 15.4 | 2 | 5 | Burkart et al.133 |
Median time to progression (TTP) in 27 patients with advanced disease.
Median TTP.
5-FU, 5-fluorouracil; AC, adenocarcinoma; CBDCA, carboplatin; CDDP, cisplatin; CPT-11, irinotecan; DTX, docetaxel; L-OHP, oxaliplatin; LV, leucovorin; ORR, overall response rate; OS, overall survival; OTH, others; PFS, progression-free survival; Pts, patients; PTX, paclitaxel; Ref, reference; SCC, squamous cell carcinoma.
Single-agent chemotherapy with taxanes or irinotecan is an option for a second- or later-line therapy. 2,130 –133 A retrospective study using a propensity score model showed the clinical benefit of OS in a second-line therapy compared with active symptom control alone for patients with ESCC, with a hazard ratio (HR) of 0.40 [95% confidence interval (CI), 0.24–0.69].134
Immunotherapy
The immune system plays a key role in eliminating cancer cells. However, antitumor immune escape is often promoted by inhibitory immune checkpoint molecules, such as PD-1 and its ligand PD-L1, during the cancer-immunity cycle process.135 –137 PD-1 receptors on T cells bind to PD-L1, and the activated PD-1/PD-L1 signaling axis induces immune tolerance in the TME by disrupting the functioning of both cytotoxic and effector T cells.136,137 ESCC exhibits higher expression of PD-L1 compared with EAC18,21,53,55,138 (Table 2), and high PD-L1 expression shows poor prognostic trends in a meta-analysis of 1350 ESCC patients.139 The treatment of ESCC has changed dramatically following several landmark trials, which have proven the benefit of immunotherapy (Table 4).
Table 4.
Clinical trials of immune checkpoint inhibitors in advanced esophageal squamous cell carcinoma.
Trials | Line | Phase | Target | Inhibitor | Agent | Treatment | No. of pts | Histology | Proportion of ESCC (%) | ORR (%) | PFS (months) | OS (months) | HR (95% CI) of PFS in ESCC | HR (95% CI) of OS in ESCC | Information | Reference |
---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
KEYNOTE-028 | ≥2 | Ib | PD-1 | Ab | Pembrolizumab (Pembro) | mono | 23 | SCC/AC | 78 | 30 (28 for SCC) | 1.8 | 7.0 | – | – | Inclusion criteria: CPS ≥ 1% | Doi et al.140 |
KEYNOTE-180 | ≥3 | II | mono | 121 | SCC/AC | 52 | 9.9 (14.3 for SCC) | 2.0 (2.1 for SCC) | 5.8 (6.8 for SCC) | – | – | ORR: 13.8% and 6.3% for CPS ≥ 10% or <10%, respectively. PFS: both 2.0 months in CPS ≥ 10% or <10% | Shah et al.141 | |||
KEYNOTE-181 | 2 | III | Pembro versus Chemo§ | 628 | SCC/AC | 64 | 13.1 versus 6.7 for ITT (16.7 versus 7.4 for SCC) | 2.1 versus 3.4 for ITT (2.2 versus 3.1 for SCC) | 7.1 versus 7.1 for ITT (8.2 versus 7.1 for SCC) | 0.92 (0.75–1.13) | 0.77 (0.63–0.96) | Primary endpoints: OS in pts with PD-L1 CPS ≥ 10, with ESCC, and in all pts | Kojima et al.18 | |||
KEYNOTE-590 | 1 | III | CF = pembro | 749 | SCC/AC | 73 | 45.0 versus 29.3 for ITT | 6.3 versus 5.8 for ITT (6.3 versus 5.8 for SCC) | 12.4 versus 9.8 for ITT (12.6 versus 9.8 for SCC, and 13·9 versus 8·8 for SCC with CPS ≥ 10) | 0.65 (0.54–0.78) | 0.72 (0.60–0.88) for SCC and 0.57 (0.43–0.75) for SCC with CPS ≥ 10% | Primary endpoints: OS in pts with ESCC and PD-L1 CPS ≥ 10, and OS and PFS in pts with ESCC, PD-L1 CPS ≥ 10, and in all pts | Sun et al.53 | |||
ATTRACTION-1 | ≥2 | II | Nivolumab (Nivo) | Mono | 65 | SCC | 100 | 17 | 1.5 | 10.8 | - | - | Kudo et al.142 | |||
ATTRACTION-3 | 2 | III | Nivo versus Chemo† | 419 | SCC | 100 | 19 versus 22 | 1.7 versus 3.4 | 10.9 versus 8.4 | 1.08 (0.87–1.34) | 0.77 (0.62–0.96) | Kato et al.19 | ||||
CheckMate 648 | 1 | III | CF=Nivo | 970 | SCC | 100 | 47 versus 27 for ITT (53 versus 20 for TPS ≥ 1%) | 5.8 versus 5.6 for ITT (6.9 versus 4.4 for TPS > 1%) | 13.2 versus 10.7 for ITT (15.4 versus 9.1 for TPS > 1%) | 0.81 (0.64–1.04) for ITT and 0.65 (0.46–0.92) for TPS ≥ 1% | 0.74 (0.58–0.96) for ITT and 0.54 (0.37–0.80) for TPS ≥ 1% | Primary endpoints: OS and PFS in TPS PD-L1 ≥ 1% | Doki et al.143 | |||
CTLA-4 | Ipilimumab (Ipi) | Ipi + Nivo versus CF | SCC | 100 | 28 versus 27 for ITT (35 versus 20 for TPS ≥ 1%) | 2.9 versus 5.6 for ITT (4.0 versus 4.4 for TPS > 1%) | 12.8 versus 10.7 for ITT (13.7 versus 9.1 for TPS > 1%) | 1.26 (1.04–1.52) for ITT and 1.02 (0.73–1.43) for TPS ≥ 1% | 0.78 (0.62–0.98) for ITT and 0.64 (0.46–0.90) for TPS ≥ 1% | |||||||
1 | II | PD-1 | Camrelizumab (Camre) | Camre + Apa + Chemo¶ | 30 | SCC | 100 | 80 | 6.85 | 19.43 | – | – | – | Zhang et al.144 | ||
ESCORT | 2 | III | Camre versus Chemo‡ | 457 | SCC | 100 | 20.2 versus 6.4 | 1.9 versus 1.9 | 8.3 versus 6.2 | 0.69 (0.56–0.86) | 0.71 (0.57–0.87) | Primary endpoint: OS for ITT | Huang et al.20 | |||
ESCORT-1st | 1 | III | TP ± Camre | 596 | SCC | 100 | 72.1 versus 62.1 | 6.9 versus 5.6 | 15.3 versus 12.0 | 0.56 (0.46–0.68) | 0.70 (0.56–0.88) | Primary endpoints: OS and PFS for ITT | Luo et al.145 | |||
NCT03469557 | 1 | II | Tislelizumab (Tisle) | CF + Tisle | 15 | SCC | 100 | 46.7 | 10.4 | N/R | – | – | Xu et al.146 | |||
RATIONALE 306 | 1 | III | Chemo†† ± Tisle | 649 | SCC | 100 | 63.5 versus 42.4 | 7.3 versus 5.6 | 17.2 versus 10.6 | 0.62 (0.52–0.75) | 0.66 (0.54–0.80) | Primary endpoint: OS for ITT | Yoon et al.147 | |||
RATIONALE 302 | 2 | III | Tisle versus Chemo§ | 512 | SCC | 100 | 20.3 versus 9.8 | 1.6 versus 2.1 | 8.6 versus 6.3 for ITT (10.3 versus 6.8 for CPS ≥ 10%) | 0.83 (0.67–1.01) | 0.70 (0.57–0.85) for ITT and 0.54 (0.36–0.79) for CPS ≥ 10% | Primary endpoint: OS for ITT | Shen et al.21 | |||
JUPITER-06 | 1 | III | Toripalimab (Tori) | TP ± Tori | 514 | SCC | 100 | 69.3 versus 52.1 | 5.7 versus 5.5 | 17.0 versus 11.0 | 0.58 (0.46–0.74) | 0.58 (0.46–0.74) | Primary endpoints: OS and PFS for ITT | Wang et al.54 | ||
ORIENT-2 | 2 | II | Sintilimab (Sin) | Sin versus chemo║ | 190 | SCC | 100 | 12.6 versus 6.3 | 1.6 versus 2.9 | 7.2 versus 6.2 | 1.00 | 0.70 | Primary endpoint: OS for ITT | Xu et al.148 | ||
ORIENT-15 | 1 | III | Chemo* ± Sin | 659 | SCC | 100 | 75.5 versus 56.9 for ITT (78.7 versus 57.5 for CPS ≥ 10%) | 7.2 versus 5.7 for ITT (8.3 versus 6.4 for CPS ≥ 10%) | 16.7 versus 12.5 for ITT (17.2 versus 13.6 for CPS ≥ 10%) | 0.56 (0.46–0.68) for ITT and 0.58 (0.45–0.75) for CPS ≥ 10% | 0.63 (0.51–0.78) for ITT and 0.64 (0.48–0.85) for CPS ≥ 10% | Primary endpoints: OS in the pts with CPS ≥ 10 and in all pts | Shen et al.149 | |||
KN046-204 | 1 | II | PD-L1/CTLA-4 | BiAb | KN046 | KN046 ± TP | 15 | SCC | 100 | 58.3 | – | – | – | – | Xu et al.150 |
A search for clinical trials included in Table 4 was conducted using the PubMed databases (https://pubmed.ncbi.nlm.nih.gov/). We used the search terms ‘clinical trial’ and ‘medication’ in combination with the term ‘esophageal cancer’. The literature screening process used the following inclusion criteria: (i) prospective phase I–III trials and (ii) studies that evaluated the therapeutic efficacy of medication. The exclusion criteria were as follows: (i) studies for esophageal adenocarcinoma, (ii) studies that evaluated the therapeutic efficacy of perioperative medication for patients with early-staged ESCC, and (iii) studies with results published in languages other than English. Several results of trials, which have been presented at a conference or meeting of cancer specialists but have not yet been published in specialist medical journals, were examined using the European Society for Medical Oncology (ESMO, https://www.esmo.org/) and the American Society of Clinical Oncology (ASCO, https://conferences.asco.org/) website.
5-FU, 5-fluorouracil; 95% CI, 95% confidence interval; Ab, antibody; AC, adenocarcinoma; BiAb, bispecific antibody; CDDP, cisplatin; CF, CDDP plus 5-FU; Chemo§, investigator’s choice (PTX, DTX, or CPT-11); Chemo†, investigator’s choice (PTX or DTX); Chemo††, investigator’s choice (platinum plus fluoropyrimidine or platinum plus PTX); Chemo¶, liposomal PTX and nedaplatin; Chemo‡, investigator’s choice (DTX or CPT-11); Chemo║, investigator’s choice (PTX or CPT-11); Chemo*, TP or CF; CPS, combined positive score; DTX, docetaxel; ESCC, esophageal squamous cell carcinoma; HR, hazard ratio; ITT, intent-to-treat population; mono, monotherapy; ORR, overall response rate; OS, overall survival; PFS, progression-free survival; Pts, patients; PTX, paclitaxel; TPS, tumor proportion score; TP, PTX plus CDDP.
In the second-line setting, monotherapy with anti-PD-1 monoclonal antibody (mAb) demonstrated a survival benefit compared with chemotherapy in phase III ATTRACTION-03,19 KEYNOTE-181,18 ESCORT,20 and RATIONALE-30221 trials. Of note, the survival curves crossed and subsequently sustained separation of the curves in favor of anti-PD-1 mAb monotherapy with duration of response, ranging from 6.9 to 8.5 months.18 –20 The ORR of anti-PD-1 mAb monotherapy was approximately only 20%.18 –21 Collectively, although a subset of patients achieved a durable response, there is still a large proportion of patients who do not benefit from anti-PD-1 mAb treatment. Chemotherapy can promote immune responses by increasing the immunogenicity of cancer cells or inhibiting immunosuppressive circuitries.151,152 Recently, the clinical benefits of anti-PD-1 mAb in combination with first-line chemotherapy for ESCC patients were demonstrated in phase III KEYNOTE-590,53 CheckMate 648,143 ESCORT-lst,145 JUPITER-06,54 ORIENT-15,149 and RATIONALE-306147 trials. The survival curves were separated from the beginning of treatment in favor of anti-PD-1 mAb plus chemotherapy compared with chemotherapy alone, and the combined therapy had a manageable safety profile. Thus, the combination of anti-PD-1 mAb and chemotherapy represents a new standard first-line treatment for ESCC patients. Although the percentage of patients who had an objective response was higher with anti-PD-1 mAb plus chemotherapy than with chemotherapy alone, the absolute difference in ORR was 10.0–21.1% in an intent-to-treat population. Thus, there is an urgent need for potential predictive biomarkers to select the most suitable patients. The status of microsatellite instability-high (MSI-H) is a robust predictive biomarker for treatment with ICIs, but MSI-H is rare in ESCC.55,153 PD-L1 expression status is among the candidate biomarkers of the response to treatment with PD-1 mAb for ESCC patients.53,143,145,147,154 However, the expression of PD-L1 on the cell surface was highly heterogeneous,23 and much more complicated factors arising from cancer cells and the tumor immune microenvironment are likely to contribute to the clinical benefit of ICI treatment.23,155,156 Therefore, further development of predictive biomarkers and more effective immunotherapy are required to improve prognostic outcomes.
Cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) acts as a negative regulator of the initial priming of T cells in the early stage of the immune response process, whereas PD-1/PD-L1 acts in later stages by turning off antitumor T-cell responses.157 Therefore, dual inhibition of PD-1/PD-L1 and CTLA-4 synergistically promotes an antitumor immune response by blocking complementary mechanisms. In a phase III CheckMate 648 trial, the combination of nivolumab plus an anti-CTLA-4 mAb ipilimumab versus chemotherapy alone was also evaluated.143 Treatment with nivolumab plus ipilimumab resulted in a longer OS (median, 12.7 versus 10.7 months) and duration of response (median, 11.1 versus 7.1 months) than chemotherapy alone in ESCC patients, suggesting that the dual inhibitor is also a new standard first-line regimen. However, another primary endpoint PFS was not met, with a crossover of the survival curves. The percentage of patients assessed as having progressive disease in treatment with nivolumab plus ipilimumab was twice as high as the percentage in chemotherapy alone (30 versus 15%), regardless of the high ORR (35 versus 20%). In addition, treatment-related serious adverse events of any grade were more common with nivolumab plus ipilimumab (32%) than with chemotherapy alone (16%), despite chemotherapy-free treatment. Special attention should be paid to treatment with nivolumab plus ipilimumab because of the double-edged sword that has benefits with durable response and risk of rapid progression and serious adverse events.
Potent molecular-targeted therapy for patients with ESCC
ESCC and EAC have different features in terms of pathogenesis, epidemiology, molecular profile, and tumor biology, emphasizing the need for the development of separate therapeutic strategies for individual histology subtypes. Although many molecular-targeted agents are currently approved for various tumor types, none of the targeted therapies, apart from ICIs, have been established for the clinical management of ESCC. Therefore, novel therapies tailored to the molecular composition are urgently required to improve prognosis. A growing number of comprehensive molecular analyses have provided potential targets for precision medicine for ESCC8,10,12,13,22 –26 (Table 2), which may lead to therapeutic breakthroughs with a personalized approach. In this section, we summarize the results of previous trials (Table 5) and discuss potential therapeutic targets (Figure 1), as well as ongoing trials (Table 6).
Table 5.
Clinical trials of molecular-targeted agents in advanced esophageal squamous cell carcinoma.
Trials | Target | Inhibitor | Agent | Line | Phase | Treatment | Histology | No. of pts (No. of ESCC) | ORR (%) | PFS (months) | OS (months) | Information | Reference |
---|---|---|---|---|---|---|---|---|---|---|---|---|---|
EMR200637-001 | EGFR | SMI | Sym004 | ≥2 | I | Mono | SCC | 30 | 16.7 | 2.3 | – | Kojima et al.158 | |
NCT03888092 | Larotinib | ≥2 | I | Mono | SCC | 81 | 13.7 | 2.9 | 5.9 | EGFR Amp or Over | Liu et al.159 | ||
NCT01855854 | Icotinib | ≥2 | II | Mono | SCC | 54 | 16.7 | 1.7 | 5.1 | EGFR Amp or Over | Huang et al.160 | ||
NCT02353936 | Afatinib | ≥1 | II | Mono | SCC | 49 | 14.3 | 3.4 | 6.3 | Hong et al.161 | |||
Ilson et al. | Erlotinib | ≥1 | II | Mono | SCC/AC | 30 (13) | 6.7 (15% for SCC) | 3.3 for SCC | 10.3 (8.2 for SCC) | Ilson et al.162 | |||
1839IL/0059 | Gefitinib (Gefi) | 2 | II | Mono | SCC/AC/OTH | 36 (9) | 2.8 | 2.0 | 5.5 | Janmaat et al.163 | |||
COG | ≥1 | III | Gefi versus placebo | SCC/AC/OTH | 449 (106) | 2.7 versus 0.4 | 1.6 versus 1.2 | 3.7 versus 3.7 | HR of PFS; 0.72 in ESCC and 0.81 in EAC | Dutton et al.164 | |||
HR = 0.80 (95% CI, 0.66–0.96) | HR = 0.90 (95% CI, 0.74–1.09) | ||||||||||||
NCT01336049 | Ab | Nimotuzumab (Nimo) | 1 | II | TP + Nimo | SCC | 56 | 51.8 | 10.8 | 20.2 | OS: 14.0 months in 27 pts with metastatic disease | Lu et al.165 | |
AIO | Cetuximab (Cetu) |
1 | II | CF ± Cetu | SCC | 62 | 19 versus 13 | 5.9 versus 3.6 | 9.5 versus 5.5 | EGFR Over | Lorenzen et al.166 | ||
POWER | Panitumumab (Pani) | 1 | III | CF ± Pani | SCC | 146 | 35.6 versus 37.0 | 5.3 versus 5.8 | 9.4 versus 10.2 | Trial stopped early based on interim efficacy results | Moehler et al.167 | ||
HR = 1.21 (95% CI, 0.85–1.73) | HR = 1.17 (95% CI, 0.79–1.75) | ||||||||||||
NCT01608022 | pan-HER§ | SMI | Dacomitinib | ≥2 | II | Mono | SCC | 49 | 12.5 | 3.3 | 6.4 | Kim et al.168 | |
NCT01142388 | IGF-IR | Ab | Cixutumumab (Cixu) | ≥2 | II | PTX ± Cixu | SCC/AC | 94 (15) | 14 versus 12 | 2.3 versus 2.6 | 6.4 versus 6.5 | Cohen et al.169 | |
EPOC1303 | PI3K | SMI | BKM120 | ≥2 | II | Mono | SCC | 42 | 4.9 | 2.0 | 9.0 | Kato et al.170 | |
ALTER1102 | multi† | Anlotinib (Anlo) | ≥2 | II | Anlo versus placebo | SCC | 165 | 7.3 versus 3.6 | 3.0 versus 1.4 | 6.1 versus 7.2 | Huang et al.171 | ||
HR = 0.46 (95% CI, 0.32–0.66) | HR = 1.18 (95% CI, 0.79–1.75) | ||||||||||||
ESO-Shanghai 11 | VEGFR2 | Apatinib (Apa) | ≥2 | II | Mono | SCC | 40 | 7.5 | 3.8 | 5.8 | Chu et al.172 | ||
NCT03285906 | ≥2 | II | Mono | SCC/AC | 30 (2) | 7.7 | 4.6 | 6.6 | Yanwei et al.173 | ||||
NCT03603756 | 1 | II | Apa + Camre + Chemo* | SCC | 30 | 80 | 6.9 | 19.4 | Zhang et al.144 | ||||
NCT03997448 | CDK4/6 | Palbociclib | ≥2 | II | Mono | SCC/AC/OTH | 21 (5) | 0 | 1.8 | 3.0 | Karasic et al.174 |
A search for clinical trials included in Table 5 was conducted using the PubMed databases (https://pubmed.ncbi.nlm.nih.gov/). We used the search terms ‘clinical trial’ and ‘medication’ in combination with the term ‘esophageal cancer’. The literature screening process used the following inclusion criteria: (i) prospective phase I–III trials and (ii) studies that evaluated the therapeutic efficacy of medication. The exclusion criteria were as follows: (i) studies for esophageal adenocarcinoma, (ii) studies that evaluated the therapeutic efficacy of perioperative medication for patients with early-staged ESCC, and (iii) studies with results published in languages other than English. Several results of trials, which have been presented at a conference or meeting of cancer specialists but have not yet been published in specialist medical journals, were examined using the European Society for Medical Oncology (ESMO, https://www.esmo.org/) and the American Society of Clinical Oncology (ASCO, https://conferences.asco.org/) website.
95% CI, 95% confidence interval; Ab, antibody; AC, adenocarcinoma; Amp, amplification; Camre, Camrelizumab (anti-PD-1 antibody); CF, CDDP plus 5-fluorouracil; Chemo*, liposomal PTX plus nedaplatin; ESCC, esophageal squamous cell carcinoma; HR, hazard ratio; mono, monotherapy; multi†, VEGFR1-3/FGFR1-4/PDGFRα-β/RET/c-KIT; pan-HER§, EGFR/ERBB2/ERBB4; PFS, progression-free survival; Pts, patients; ORR, overall response rate; OS, overall survival; OTH, others; Over, overexpression; PTX, paclitaxel; SMI, small molecule inhibitor; TP, PTX plus cisplatin.
Figure 1.
Potent inhibitors directed against pivotal signaling pathways, including EGFR, FGFR1, VEGFR2, NRF2, YAP1, and cyclin D1-CDK4/6, in ESCC. EGFR, FGFR1, and VEGFR2 are members of the RTK family. Binding the ligands to these RTK receptors leads to cytoplasmic tyrosine phosphorylation, resulting in the activation of MAPK and PI3K/Akt signaling. The PI3K/Akt pathway is also activated by FAK and loss of PTEN. YAP1 is activated through inhibition of LATS1/2 via loss of function mutations of FAT1 as upstream activators of NF2/merlin, which forms transcription complexes with TFs, including TEAD, AP-1, BRD4, and E2F. Cyclin D1 forms a complex with CDK4/6, resulting in disruption of cell cycle restriction. NRF2 signaling pathway is activated by alterations of NF2EL2, KEAP1, and CUL3 genes, which induces cytoprotective proteins upon oxidative stress. NRF2-addicted cancer cells have dependence on glutaminase (GLS)-mediated glutamine metabolism, including glutathione (GSH).
mAb, monoclonal antibody; PTEN, phosphatase and tensine homolog; SMI, small molecule inhibitor; TKIs, tyrosine kinase inhibitors.
Table 6.
Ongoing clinical trials of molecular-targeted agents in advanced esophageal squamous cell carcinoma.
Trials | Target | Inhibitor | Agents | Phase | Treatment | Inclusion criteria |
---|---|---|---|---|---|---|
NCT05164848 | EGFR | TKI | Afatinib (Afa) | I | Afa + JMT101 | EGFR Over or Amp |
NCT04880811 | II | Afa + Tori | ||||
NCT04839471 | II | Afa + BI-754091 | ||||
NCT04415853 | Larotinib (Laro) | III | Laro versus CPT-11/S-1 | EGFR Over | ||
NCT04229537 | Ab | SCT200 | I | SCT200 + SCT-I10A | ||
NCT05164848 | JMT101 | I | JMT101 + Afa | |||
NCT03766178 | Nimotuzumab (Nimo) | II | Nimo + Camre | |||
NCT04945733 | EGFR/MET | BiAb | Amivantamab, JNJ-61186372 | II | Mono | EGFR Over or MET Over |
NCT05117931 | II | Mono | EGFR or MET Amp | |||
NCT05022654 | EGFR/HER3 | BiAb | SI-B001 | II | SI-B001 + CPT-11 | Prior ICIs |
NCT02052778 | FGFR | TKI | Futibatinib, TAS-120 (Futi) | I/II | mono | FGF/FGFR aberrations |
NCT04721223 | SHP2 | Allo | JAB-3068 | I | JAB-3068 + Tori | |
NCT04000529 | TNO155 | I | TNO155 + Spa | |||
NCT02645864 | VEGFR2 | TKI | Apatinib (Apa) | I | Apa + CPT-11 | |
NCT05049681 | III | Camre ± Apa | ||||
NCT03762564 | Ab | Ramucirumab (Ram) | II | PTX ± Ram | ||
NCT05038813 | multi§ | TKI | Anlotinib, AL3818 (Anlo) | II | Anlo + TQB2450 | |
NCT04063683 | II | Anlo + TP | ||||
NCT05013697 | II | TQB2450 + TP ± Anlo | ||||
NCT03387904 | II | CPT-11 ± Anlo | ||||
NCT04797507 | II | Anlo + Camre | ||||
NCT04503967 | II | Anlo + Nivo | ||||
NCT04866381 | multi† | TKI | Famitinib, SHR-1020 (Fami) | II | Fami + Camre | Prior ICIs |
NCT05007613 | multi║ | TKI | Cabozantinib (Cabo) | II | Cabo + Atezo | |
NCT04949256 | multi‡ | TKI | Lenvatinib (Lenva) | III | Pembro + Chemo* ± Lenva | |
NCT04984018 | HDAC1–3 and 10 | Benza | Tucidinostat, Chidamide (Tucid) | II | Tucid + Camre | Prior ICIs |
NCT05163483 | II | Tucid + Tori | ||||
NCT04866381 | CDK4/6 | SMI | Dalpiciclib, SHR-6390 (Dalp) | II | Mono | Prior ICIs |
NCT04866381 | II | Dalp + Camre | Prior ICIs | |||
NCT02915432 | PD-1 | Ab | Toripalimab, JS001 (Tori) | Ib/II | Mono | |
NCT03785496 | Spartalizumab, PDR001 (Spa) | II | Mono | |||
NCT03958890 | HLX10 | III | CF ± HLX10 | |||
NCT04187352 | PD-L1 | Ab | Sugemalimab, CS1001 (Suge) | III | CF ± Suge | |
NCT03925870 | PD-L1/CTLA-4 | BiAb | KN046 | II | Mono | |
NCT04785820 | PD-1/TIM3 | BiAb | RO7121661 | II | RO7121661 versus Nivo | |
NCT04785820 | PD-1/LAG3 | BiAb | RO7247669 | II | RO7247669 versus Nivo | |
NCT04540211 | TIGIT | Ab | Tiragolumab (Tirago) | III | TP ± Tirago plus Atezo | |
NCT04732494 | Ociperlimab, BGB-A1217 (Ocip) | II | Tisle ± Ocip | |||
NCT05104567 | IL-2 | Recom | THOR-707 (SAR444245) | II | THOR-707 + Pembro |
A search for ongoing clinical trials that had been included in Table 6 was conducted using ClinicalTrials.gov (https://clinicaltrials.gov/). Search approaches were performed by entering a keyword such as ‘esophageal squamous cell carcinoma’ in the ‘condition or disease’ field and by selecting ‘adult’ in the age field and ‘interventional (clinical trial)’ in the ‘study type’ field using the basic and advanced interfaces in the resource. In addition, specific targeted molecules were searched using the ‘other terms’ field. National Clinical Trial (NCT) identifier number was listed for each specific trial in each table.
Ab, antibody; Allo, bioavailable allosteric inhibitor; Alpe, Alpelisib (BYL719, selective PI3Kα inhibitor); Amp, amplification; Atezo, Atezolizumab (anti-PD-L1 antibody); BI-754091, anti-PD-1 antibody; Benza, benzamide-type inhibitor; BiAb, bispecific antibody; Camre, Camrelizumab (anti-PD-1 antibody); CDDP, cisplatin; Chemo*, CF or FOLFOX; CF, CDDP plus 5-fluorouracil; CPT-11, irinotecan; ICIs, immune checkpoint inhibitors; IP, CPT-11 plus CDDP; L-OHP, oxaliplatin; Mono, monotherapy; multi†, VEGFR-2/PDGFR/c-KIT; multi‡, VEGFR1-3/PDGFR α/FGFR1-4/KIT/RET; multi§, VEGFR1-3/FGFR1-4/PDGFRα-β/RET/c-KIT; multi║, VEGFR1-3/MET/AXL; NCT number, ClinicalTrials.gov Identifier; NDP, nedaplatin; Nivo, Nivolumab (anti-PD-1 antibody); Over, overexpression; Pembro, Pembrolizumab (anti-PD-1 antibody); PTX, paclitaxel; Recom, pegylated recombinant IL-2 receptor beta/gamma complex; SCT-I10A, anti-PD-1 antibody; SMI, small molecule inhibitor; TKI, tyrosine kinase inhibitor; Tori, Toripalimab (JS001, anti-PD-1 antibody); TP, PTX plus CDDP; TQB2450, anti-PD-1 antibody.
EGFR-targeted therapy
EGFR overexpression and amplification are reported in 18–89% and 6–26% of ESCCs, respectively.8,10,12,13,22 –26,48,52,55,160,175 –177 A meta-analysis including 802 patients with ESCC showed that EGFR overexpression was associated with lymph node metastatic status and poor OS.178 In addition, there was a trend toward a worse prognosis for ESCC patients with a high EGFR gene copy number than for those with a low copy number.179
Based on the frequent EGFR overexpression of gene amplification in ESCC, several phase II/III trials have been conducted to evaluate the treatment efficacy of EGFR inhibitors, including mAb and tyrosine kinase inhibitors (TKIs). Unfortunately, these trials have shown limited efficacy (Table 5). A phase III COG trial of EGFR TKI gefitinib versus placebo in previously treated patients with EC, including 106 patients with ESCC, was conducted.164 Although gefitinib showed the limited benefits of PFS (1.6 versus 1.2 months), the primary endpoint, OS, was not met. Another phase III POWER trial of first-line chemotherapy with or without EGFR mAb panitumumab in ESCC patients was terminated early because of potential safety concerns on interim analysis.167 There was no improvement in OS in addition to panitumumab, with more frequent treatment-related deaths. Similar findings were found in a randomized phase II AIO trial of the addition of EGFR mAb cetuximab to first-line chemotherapy in ESCC patients, which resulted in no significant improvement in terms of OS, PFS, and ORR.166 Of note, only a subset of patients had marked clinical responses.160,161,163,164,168,180 The identification of predictive biomarkers is vital for improving the clinical outcomes of patients treated with EGFR inhibitors. In a phase Ib trial of EGFR TKI larotinib in pretreated advanced ESCC with EGFR overexpression or amplification, larotinib demonstrated promising antitumor activity, with an ORR of 20.0% and median PFS of 3.4 months at a dose of 350 mg 149. A phase III trial of larotinib versus chemotherapy is ongoing for ESCC with EGFR overexpression (NCT04415853; Table 6).
Although miRNA-338-5p inhibits the signaling pathways of MET and EGFR by directly targeting the 3′ untranslated regions of these genes,181 its expression is downregulated in the majority of ESCC cases.182 Amivantamab is a bispecific antibody (BiAb) targeting EGFR and MET, with immune cell-directing activity.183 The U.S. Food and Drug Administration granted accelerated approval to amivantamab for patients with non-small-cell lung cancer harboring the EGFR exon 20 insertion mutation, based on the results of the CHRYSALIS trial.184 The activity of amivantamab is assessed in ESCC with overexpression or amplification of EGFR and MET (NCT04945733 and NCT05117931) (Table 6). The EGFR signaling pathway is activated via hetero-dimerization with other HER families as well as EGFR homo-dimerization.185 A phase II trial of an irreversible pan-HER TKI dacomitinib was conducted to assess whether combined inhibition of all HER family kinases has more potent antitumor activity than EGFR inhibition alone in ESCC patients.168 The ORR was 12.5% (6 of 48 patients), with a median PFS of 3.3 months (Table 5). There are ongoing early trials of EGFR/HER3, a bispecific antibody (BiAb) SI-B001 (NCT05022654), and a combined blockade of EGFR using TKI afatinib and mAb JMT101 (NCT05164848) (Table 6). In addition, aberrations of cell cycle-related genes coexist in the majority of EGFR-amplified ESCC, and the blockade of CDK4/6 may enhance the efficacy of the EGFR inhibitor.186
FGFR-targeted therapy
The FGFR family consists of four highly conserved receptors (FGFR1, FGFR2, FGFR3, and FGFR4). The FGFR pathway is cancer specifically dysregulated by its overexpression and genetic alterations, driving cancer development and progression.187 –189 FGFR genomic alterations were detected in 7% of the 4853 solid tumors, of which gene amplifications were the most common alteration, with 66% of the cases having FGFR aberrations.188 In ESCC, FGFR1 has the most frequent genetic alteration among FGFR family members, and FGFR1 gene amplification was detected in 14.3%.188 A meta-analysis including 2326 ESCC patients showed that FGFR1 amplification was significantly correlated with lymph node metastasis, poorer differentiation, and unfavorable OS with an HR of 1.50 (95% CI: 1.25–1.81).190 In addition, FGFR ligands FGF3, FGF4, and FGF19 genes are more frequently amplified in ESCC.55 Futibatinib (TAS-120) is an orally bioavailable, highly selective, and irreversible FGFR1–4 inhibitor.191 A phase I/II trial of futibatinib is ongoing in patients with advanced solid tumors harboring FGF/FGFR aberrations (NCT02052778; Table 6).
PI3K-targeted therapy
PI3K/Akt signaling is one of the most frequently dysregulated biochemical pathways in ESCC.8,10 –13,22 –24,48 Class I PI3Ks consist of a p110 catalytic subunit and a p85 regulatory subunit, and they are divided into four catalytic isoforms, p110α, p110β, p110γ, and p110δ, which are encoded by PIK3CA, PIK3CB, PIK3CG, and PIK3CD, respectively.192 BKM120 is an oral pan-class I PI3K inhibitor targeting all four catalytic isoforms of class I PI3K. In a phase II EPOC1303 trial of BKM120 in 41 ESCC patients, the primary endpoint was met, with a disease control rate (DCR) of 51.2%170 (Table 5). Although pan-class I PI3K inhibitors have the advantage of on-target inhibition of each of the class I PI3K catalytic isoforms, they have the risk of broader off-target effects, resulting in an unfavorable toxicity profile and limited clinical utility.193 PI3K isoform-specific inhibitors may offer a superior therapeutic window and less toxicity than pan-class I PI3K inhibitors.192 Double PIK3CA mutations in cis may be a predictive biomarker for selecting patients who would benefit from PI3Kα inhibitors.194
There are alternative strategies for the blockade of the PI3K/Akt signaling pathway. The insulin-like growth factor-II (IGF-II)/IGF-I Receptor (IGF-IR) signaling pathway plays an important role in ESCC progression by activating Akt, and treatment with anti-IGF-IR mAb cixutumumab resulted in the enhanced antitumor effect and chemosensitivity in a preclinical study.195 However, in a phase II trial of second-line paclitaxel plus cixutumumab in patients with metastatic EC (NCT01142388), the addition of cixutumumab did not improve clinical outcomes169 (Table 5). Focal adhesion kinase (FAK) is a cytoplasmic tyrosine kinase that activates the PI3K/Akt pathway as a mediator of cell signaling downstream of growth factor and cytokine receptors.196 In a preclinical study, a specific FAK inhibitor defactinib dose and time dependently induced the dissociation of PI3K, which led to the blockade of Akt signaling and the expression of several oncogenes, such as SOX2, MYC, EGFR, or MET.197 Furthermore, treatment with defactinib suppressed tumor growth and metastatic ability in ESCC xenograft models. AXL is a RTK belonging to the Tyro3/Axl/Mer (TAM) family,198 and it is frequently overexpressed in ESCC.199 AXL dimerizes with EGFR to activate the protein kinase C (PKC), leading to the activation of mTOR in an PI3K-independent manner.200 Consequently, AXL overexpression mediates resistance to PI3K inhibitors by activating the EGFR/PKC/mTOR axis in ESCC, and the blockade of AXL may revert resistance to PI3Ka inhibitors. Because several other signaling pathways connect with the PI3K/Akt signaling network,193 the major targets downstream of the PI3K pathway may be compensated for via bypass or crosstalk signaling when PI3K is blocked.
Angiogenesis-targeted therapy
Angiogenesis contributes to the progression of tumorigenesis and metastasis by providing nutrition, growth factors, and an oxygen supply.201 In addition, tumor vasculature suppresses the delivery of chemotherapy agents to the tumor site due to its leaky properties.202 VEGF and its receptor VEGFR are key mediators responsible for angiogenesis, and therapies directed against VEGF/VEGFR are the focus of major molecular-targeted research in solid tumors. In ESCC, high levels of VEGF expression were associated with poor survival in a meta-analysis that included 2043 patients with ESCC,203 and several trials of anti-VEGFR inhibitors were conducted.
Apatinib is a novel VEGFR2 tyrosine kinase inhibitor, and treatment with apatinib resulted in inhibited proliferation and induced apoptosis in ESCC cell line models.204 Two phase II trials showed similar clinical activity for apatinib, with ORR of 7.5–7.7%, DCR of 61.5–65.0%, and PFS of 3.8 months–4.6 months, as a second- or later-line therapy in ESCC patients172,173 (Table 5). Anlotinib is an oral small-molecule TKI targeting VEGFR 1–3, FGFR 1–4, platelet-derived growth factor receptor (PDGFR) α and β, RET, and c-KIT.205 In a randomized, double-blind, placebo-controlled phase II trial to evaluate the antitumor activity and safety of anlotinib as a second- or later-line therapy in ESCC patients, the PFS as a primary endpoint was significantly improved in anlotinib compared with placebo, although there was no significant difference in terms of ORR (7.3 versus 3.6%)171 (Table 5). Currently, several early phase trials of agents targeting angiogenesis in combination with chemotherapy are ongoing: (i) TKIs, including apatinib (NCT02645864) and anlotinib (NCT04063683 and NCT03387904), and (ii) mAb directed against VEGFR2 (ramucirumab, NCT03762564) (Table 6).
CDK4/6-targeted therapy
The cyclin D1-CDK4/6 pathway is triggered by amplifications of CCND1 and CDK4/6, and losses of CDKN2A and RB1 in ESCC,10,12,13,24,48,58 suggesting a rational target for ESCC. CDK4/6 has proven to be an effective target in hormone receptor-positive advanced breast cancer.206 A phase II trial of palbociclib, a SMI of CDK4/6, was conducted with patients with EC and gastric cancer, among which one of five ESCC patients had stable disease lasting for more than 4 months174 (Table 5). Dalpiciclib (SHR6390) is an orally bioavailable, small-molecule CDK4/6 inhibitor, and treatment with dalpiciclib suppressed cell proliferation and tumor growth by inducing cell cycle arrest at the G1 phase in ESCC cell lines and xenograft models.207 An exploratory clinical trial of dalpiciclib is ongoing in ESCC patients (NCT04866381; Table 6).
Because CDK4/6 inhibitor alone may limit clinical efficacy, the combination with other therapeutic strategies can be of potential benefit. Therefore, the identification of a therapeutic vulnerability in ESCC harboring cell cycle dysregulation is required. Aberration of the cyclin D1-CDK4 pathway also leads to reprogramming of cellular metabolism, which contributes to glutamine addiction and mitochondrial dysfunction.208 Dual blockade of glutaminase and mitochondrial respiration may effectively reduce tumor burden by interfering with cell proliferation and survival.
NRF2-targeted therapy
Transcription factor NRF2, encoded by the NFE2L2 gene, regulates cellular antioxidant, detoxification, inflammation, and metabolic pathways by modulating the constitutive and inducible expression of over 200 genes that contain antioxidant response elements in their regulatory regions.209 –211 NRF2 also induces upregulation of multidrug resistance-associated proteins, which are a major drug efflux pump.212 Consequently, the NRF2 signaling pathway confers cancer cells with a proliferative advantage, as well as resistance to antitumor drugs and radiotherapies. In ESCC, the NRF2 pathway is aberrantly activated by genetic and epigenetic alterations, including gain-of-function mutations of the NFE2L2 gene and loss-of-function mutations of negative regulators KEAP1 and CUL3 genes.9,12,13,24,88 ESCC cells with mutations of the NFE2L2 gene showed increased cell proliferation and resistance to 5-fluorouracil and γ-irradiation,87 and blockage of NRF2 repressed the migration and invasion of ESCC cells in the hypoxic microenvironment.213 NRF2 overexpression was associated with lymph node metastases, unfavorable response to neoadjuvant chemoradiation, and poor postoperative outcomes in ESCC patients.87,214 Thus, the NRF2 signaling pathway may contribute to the hallmarks of cancer through cellular metabolic processes leading to ‘NRF2 addiction’ in ESCC, and have potential as a relevant therapeutic target for NRF2-addicted ESCC patients.
Several strategies have been proposed to target the NRF2 signaling pathway.215 Potent inhibitors of NRF2, including brusatol,216 halofuginone,217 AEM1,218 ML385,219 and trigonelline,220 have been identified by high-throughput screening.221 Many flavonoids, such as luteolin, chrysin, and apigenin, have been reported as regulators of NRF2 mRNA expression in cancer cells.222 NRF2 downstream pathways can also be targeted. Nuclear receptor subfamily 0 group B member 1 (NR0B1) was identified as a druggable vulnerability by chemical proteomics using NRF2-addicted lung cancer cells, and NR0B1 protein complexes were disrupted by novel covalent SMIs, such as BPK-26 and BPK-29.223 As an important safety concern, NRF2 inhibitors may cause ‘off-target’ side effects on cancer-bearing hosts because of the essential role of NRF2 in cytoprotection.
Metabolic rewiring evokes cellular mechanisms that reduce therapeutic mightiness. Cancer cells avidly consume glutamine to fuel biosynthesis and proliferation, and glutaminases regulate glutathione and oxidative stress, which are activated through NRF2-driven metabolic adaptations.211,224 NRF2-addicted cancer cells depend on glutamine metabolism and show vulnerability to glutaminase inhibition via depletion of glutathione and increased replication stress.225 –228 Telaglenastat (CB-839) is a first-in-class, oral, highly selective glutaminase inhibitor that has demonstrated preclinical activity in NRF2-addicted models225 –228 and antitumor activity in patients with previously treated renal cell carcinoma in the phase II ENTRATA trial.229 In addition, a combined therapy of glutaminase inhibitor and the poly (ADP-ribose) polymerase (PARP) inhibitor may be an effective synthetic lethal approach.226 Competition between cancer cells and immune cells for glutamine as a critical nutrient creates a metabolic checkpoint that induces immunosuppressive status in TME. Telaglenastat may enhance the antitumor effect of ICIs through the activation of immune cell function via blocking tumor glutamine consumption.230
YAP1-targeted therapy
The Hippo pathway controls organ development and tissue homeostasis.231 YAP1 is a downstream transcription coactivator of the Hippo pathway and forms transcription complexes with TFs, including the transcriptional enhancer activator domain (TEAD), activator protein 1 (AP-1), bromodomain-containing protein 4 (BRD4), and E2 factor (E2F).231 –234 YAP1 signaling is abnormally dysregulated in various tumor types, which is implicated in tumorigenesis, EMT, and CSC traits, a central bypass mechanism that results in resistance to EGFR and RAS/MAPK inhibitors, and plays a protective role against chemotherapy-induced apoptosis.231 –239
In ESCC, YAP1 is constitutively activated due to YAP1 gene amplification, mutations in FAT families as upstream activators of neurofibromin 2 (NF2)/merlin in the Hippo pathway, and deletion of vestigial-like family member 4 (VGLL4)/autophagy related 7 (ATG7) as a direct competitor with YAP for binding to TEADs.6,12,240 In addition, YAP1 is frequently overexpressed in 56–81% of ESCC cases, which are associated with aggressive phenotype.241,242 YAP1 controls the squamous differentiation of esophageal basal cells through crosstalk with Notch signaling, and ectopic nuclear YAP1 activation causes a hyperplastic phenotype of the esophageal epithelium in gain-of-function murine models.243 Knockdown of YAP1 results in increased apoptosis and decreased properties of proliferation, invasion, chemoresistance, and CSCs in ESCC.240,244 –246 Thus, YAP is a promising target for ESCC.
Currently, there are potentially viable agents targeting YAP1 signaling, including TEAD inhibitors (CA3, MYF-01–37, K-975, and IK-930),238,247 –249 BRD4 inhibitor (JQ1) as blockade of both YAP1 transcription and direct interaction of BRD4 with YAP1,239,250 tankyrase inhibitor (XAV939) as an indirect inhibitor of angiomotin-mediated YAP1 activity,251 and potent next-generation constrained ethyl antisense oligonucleotides to selectively target YAP1 (ON-537).252 As the YAP1-TEAD transcription complex controls the expression of CDK6, which leads to cellular senescence,253 dual blockade of YAP1 and CDK6 may also be effective in ESCC.254 IK-930 is a selective TEAD inhibitor that disrupts the TEAD-dependent transcription of key genes involved in cancer progression, metastases, and therapeutic resistance. A preclinical study showed the antitumor activity of IK-930 both as a monotherapy and in combination with targeted agents, such as MEK and EGFR inhibitors, in multiple tumor models.249 A phase I first-in human trial of IK-930 is ongoing to evaluate the safety and preliminary antitumor activity in advanced solid tumors (NCT05228015). ION-537 is a next-generation antisense oligonucleotide inhibitor of YAP1, which has resulted not only in a markedly decreased expression of YAP1 protein and preclinical treatment efficacy, but also in enhanced immune activation, such as an increased number of infiltrating T cells and myeloid lineage cells in the tumors.252 A phase I trial of ION537 is ongoing in molecularly selected advanced solid tumors (NCT04659096). Although YAP1 downstream pathway-targeted therapy represents a rational approach, it is unlikely that targeting one downstream effector will fully recapitulate the effects of the direct blockade of YAP1 because of the diverse functions and escape mechanisms of YAP1-targeted molecules.
Combined immunotherapy
Although ESCC represents a potential anti-tumorigenic immune environment through activated T cells and increased dendritic cells (DCs), the infiltration of cytotoxic T cells into tumors is hindered by exclusionary pressures.255 In a single-cell transcriptome analysis of the TME, ESCCs had profound heterogeneity in the cell composition of individual tumors and their cellular status, leading to distinct immune status and tumor biology.155,156 In addition, different subsets of T cells express distinct inhibitory immune checkpoint molecules, such as CTLA-4, T-cell immunoreceptor with Ig and ITIM domains (TIGIT), CD96, and lymphocyte activation gene 3 protein (LAG3), in the TME of ESCC.155,156 Collectively, most ESCCs may be ‘cold’ tumors, with low TILs and high expression of co-inhibitory molecules, and the immunologically ignorant phenotype may have a poor response to ICIs. Therefore, several treatment strategies have been examined to turn immunologically ‘cold’ tumors with poor immune activation into ‘hot’ tumors with strong immune infiltration in clinical trials combining the anti-PD-1/PD-L1 mAb with other immune-modulating treatments, including other ICIs, anti-angiogenetic inhibitors, and other agents (Table 6).
KN046 is a novel recombinant humanized PD-L1/CTLA-4 bispecific single-domain antibody-Fc fusion protein that can bind effectively to both PD-L1 and CTLA-4.256 A phase II KN046-204 trial (NCT03925870) of KN046 monotherapy or combined with chemotherapy in ESCC is ongoing, in which the ORR and DCR are 58.3% and 91.6%, respectively, in a cohort with KN046 plus first-line chemotherapy.150 The treatment efficacy of a fully human immunoglobin G4 anti-PD-L1 mAb sugemalimab (CS1001) in combination with first-line chemotherapy is also assessing in a phase III trial (NCT04187352) (Table 6).
The PD-1/PD-L1 interaction is not the only immune checkpoint pathway regulating T-cell activation in the TME. TIGIT, LAG3, and T-cell immunoglobulin mucin receptor 3 (TIM3) are overexpressed on effector CD4+ and CD8+ T cells, regulatory T cells (Tregs), and natural killer (NK) cells, which act as inhibitory immune checkpoint modulators.257 TIGIT binds to CD155 with high affinity and competes with its activating counter-receptor CD226, which suppresses antitumor immunity through inactivating immune cell effector functions, either directly, or indirectly through Treg-mediated suppression.258 The anti-TIGIT mAb tiragolumab plus the anti-PD-L1 mAb atezolizumab showed a clinically meaningful benefit of ORR and PFS compared with atezolizumab alone as a first-line treatment for patients with PD-L1-positive lung cancer.259 In ESCC, a phase III SKYSCRAPER-08 trial (NCT04540211) is ongoing to evaluate the efficacy of tiragolumab plus atezolizumab in combination with first-line chemotherapy versus chemotherapy alone (Table 6). In a randomized phase II AdvanTIG-203 (NCT04732494), the clinical benefit of anti-TIGIT mAb ociperlimab (BGB-A1217) plus anti-PD-1 mAb tislelizumab versus tislelizumab alone was assessed in a second-line setting. LAG-3 has high-affinity binding to the major histocompatibility complex class II, which acts as an inhibitory immune checkpoint via the activation of Tregs and the suppression of DCs and CD8 + T cells.260 LAG-3 and PD-1 synergistically regulate T-cell function in promoting immune escape.261 A phase I trial of anti-PD-1 mAb BI 754,091 alone and in combination with anti-LAG-3 mAb BI 754,111 was conducted in Asian patients with advanced solid tumors, including ESCCs.262 In a cohort of BI 754,091 plus BI 754,111 in EC patients who had received ≥1 line of prior systemic therapy and no prior anti-PD-1/PD-L1 therapy, the ORR was 21.6% (8/37). A randomized phase II trial (NCT04785820) of a PD-1/LAG-3 BiAb RO7247669 versus anti-PD-1 mAb nivolumab in ESCC is ongoing. T-cell immunoglobulin mucin-3 (TIM-3) expressed on innate immune cells is a crucial checkpoint that can induce T-cell apoptosis and exhaust type 1 CD4+ T cells and type 1 CD8+ T cells by binding to its ligand glycan-binding protein-9 expressed on cancer cells.257,263 The blockade of TIM-3 preclinically restored the functions of TILs,264 overcame resistance to an anti-PD-1 mAb 234, and promoted a synergistic effect in combination with anti-PD-1 mAb.265 The randomized phase II trial (NCT04785820) is assessing the efficacy of a PD-1/TIM-3 BiAb RO7121661 versus nivolumab in ESCC (Table 6).
As VEGF has been found to exert immunosuppressive effects via several mechanisms, such as decreasing the number of TILs, activating immune checkpoint molecules, inhibiting DC differentiation, and downregulating MHC,136,266 antiangiogenic agents could have immunomodulatory effects. In fact, the most compatible partners of ICIs were an anti-angiogenetic inhibitor and platinum chemotherapy in a cross-sectional study from 98 clinical trials including 24,915 patients.267 Thus, the combination of antiangiogenetic agents with ICI has emerged as an alternative strategy. In a phase II multiple cohort trial of regorafenib, a novel oral multi-kinase inhibitor including angiogenic kinases, in combination with nivolumab in advanced solid tumors, the ESCC cohort showed an ORR of 43% and median PFS of 6.9 months in 30 patients.268 Currently, there are several ongoing trials combining ICI with apatinib (NCT05049681), anlotinib (NCT05038813, NCT05013697, NCT04797507, and NCT04503967), famitinib (NCT04866381), and cabozantinib (NCT05007613; Table 6). Lenvatinib targets VEGFR 1, 2, and 3, PDGFR α, FGFR, c-KIT, and RET tyrosine kinases. A systematic review showed more promising antitumor activity in solid cancers with lenvatinib plus pembrolizumab than with either lenvatinib or pembrolizumab alone.269 A phase III LEAP-014 trial (NCT04949256) of lenvatinib plus pembrolizumab versus pembrolizumab in combination with first-line chemotherapy for ESCC patients is ongoing.
The EGFR signaling pathway establishes the non-inflamed and immunosuppressive TME, with recruitment and activated function of Tregs, downregulated MHC expression, and decreased numbers of effector CD8+ T cells, DCs, and M1-like tumor-associated macrophages.270 Therefore, blockade of EGFR signaling may synergistically enhance the efficacy of PD-1/PD-L1 inhibitors in ESCC (NCT04880811, NCT04839471, NCT03766178, and NCT04229537) (Table 6).
Activation of FGFR suppresses cancer immunity through inhibition of interferon-gamma-stimulated JAK/STAT signaling pathway271 and downregulation of MHC class I and MHC class II.272 In preclinical models, the combination of FGFR-TKIs with anti-PD-1/PD-L1 mAb augmented antitumor immunity in renal cell carcinoma271 and head and neck squamous cell carcinoma.272 As FGFR is frequently activated and associated with poor prognosis in ESCC,188,190 the treatment strategy targeting both FGFR and PD-1/PD-L1 may be a novel immunologic approach for treating patients with FGFR-aberrant ESCC. In a phase Ib trial of futibatinib plus pembrolizumab in advanced or metastatic EC harboring FGFR mRNA overexpression, the interim analysis showed acceptable safety, with an ORR of 44% in ICIs naïve patients and 20% in ICI refractory patients.273
Src homology 2 domain-containing phosphatase (SHP2) is a non-receptor ubiquitous protein tyrosine phosphatase,274 which serves as an important hub to connect several intracellular oncogenic signaling pathways.275 It not only suppresses T-cell activation, but also causes T-cell anergy as a downstream molecule in the PD-1 signaling pathway.276,277 In preclinical studies, SHP2 inhibition reversed immunosuppression in the TME via restored Th1 immunity and activated T-cell function.277 Of note, the combined inhibition of SHP2 and anti-PD-1 had a higher antitumor activity than either inhibition alone.278 Thus, the dual blockade of SHP2 and PD-1/PD-L1 may enhance immune-mediated responses. The rationale for dual blockade has been assessed in phase I trials of anti-PD-1 mAb plus allosteric SHP2 inhibitor JAB-0368 (NCT04721223) or TNO155 (NCT04000529; Table 6).
Interleukin-2 (IL-2) is an immunoregulatory cytokine that drives effector T and NK cell expansion through the IL-2 receptor beta/gamma complex-JAK/STAT signaling axis.279 In contrast, IL-2 is essential for generation of Tregs, which are more sensitive to IL-2 stimulation than the other immune cell populations, such as conventional T cells and NK cells, due to constitutive expression of IL-2 receptor alpha.280 These conflicting functions suggest that a more selective approach is required for therapeutic applications. THOR-707 is a recombinant human IL-2 molecule irreversibly bound to a pegylated chain to block alpha-binding while retaining affinity for beta/gamma IL-2 receptor subunits.281 Treatment with THOR-707 resulted in enhanced tumor infiltration of CD8+ T cells and NK cells, without Treg expansion, and a reduction of tumor growth in vitro. In an ongoing multicohort phase II trial, the clinical benefit of THOR-707 plus anti-PD-1 mAb was assessed in a subcohort of ESCC (NCT05104567; Table 6).
Histone deacetylase (HDAC) inhibitors enhance immunogenicity and antitumor immune responses through the reduced number and function of myeloid-derived suppressor cells and Tregs,282 –284 enhanced tumor antigens,285 and increased MHC presentation.285 On the other hand, treatment with HDAC inhibitors resulted in upregulation of PD-L1,286 and nuclear acetylated PD-L1 stimulated antigen presentation.287 These preclinical data provide a rationale for combining HDAC inhibitors with anti-PD-1/PD-L1 therapies. Several early clinical trials have shown encouraging effects of combination treatment with the HDAC inhibitor and the PD-1/PD-L1 inhibitor in melanoma, lung cancer, or head and neck cancer.288 –290 Tucidinostat is an orally bioavailable benzamide-type inhibitor of HDAC isoenzymes 1, 2, 3, and 10. In ESCC, two phase II trials of tucidinostat plus PD-1 inhibitors are ongoing (NCT04984018 and NCT05163483; Table 6).
Potent strategies of definitive chemoradiation
Definitive chemoradiotherapy (dCRT) is the standard treatment for locally advanced, unresectable ESCC,291 but it is associated with poor local control and higher locoregional recurrence, ranging from 27% to 83%.292 –294 Therefore, several intensified treatment modalities have been attempted to improve treatment outcomes, such as the addition of agents targeting EGFR and ICI. A phase II/III SCOPE1 trial was conducted to evaluate the addition of anti-EGFR mAb cetuximab to dCRT.295 The futility criteria in the planned interim analysis were met, and this trial was stopped early without continuation to phase III. Similarly, a phase III NRG Oncology RTOG 0436 trial has failed to demonstrate OS improvement with the addition of cetuximab to dCRT in EC patients as well as in ESCC patients as a histologic analysis.296 In contrast, two phase III trials demonstrated promising results of EGFR TKI erlotinib297,298 and EGFR humanized mAb nimotuzumab.299 In a phase III trial of erlotinib in combination with dCRT in ESCC, the addition of erlotinib significantly decreased locoregional recurrence and improved OS, especially in patients with EGFR overexpressing ESCC.297,298 A phase III NXCEL1311 trial was conducted to compare the treatment efficacy of dCRT plus nimotuzumab with dCRT plus a placebo in unresectable locally advanced ESCC.299 An interim analysis for short-term efficacy showed a significantly higher ORR (93.8 versus 72.0%) and CR rate (32.5 versus 12.2%) in nimotuzumab than the placebo. An analysis of the primary endpoint of OS is planned.
It has been shown that chemotherapy and radiotherapy could cause immunogenic cell death in cancer cells, which is recognized by DC and activates cytotoxic T cells.300 Radiotherapy also induces antitumor immune response through the enhanced diversity of the T-cell receptor repertoire of intra-tumoral T cells via DNA damage301 and the infiltration of effector T cells into tumors via the release of chemokines.302 In non-small cell lung cancer, synergistic effects between CRT and ICIs have been demonstrated,303 indicating the rationale for this combinational strategy. Two phase II trials showed the promising efficacy of consolidation ICI following dCRT in patients with unresectable locally advanced ESCC. In a treatment with atezolizumab following dCRT, the confirmed CR was 40%, with median PFS of 3.2 months and OS of 31.0 months, in 40 patients.304 In an interim analysis of anti-PD-1 mAb camrelizumab as a consolidation therapy after dCRT, 13 of 15 patients had stable disease. After a median follow-up period of 17 months, the median PFS and OS were not reached.305 Currently, several phase III trials have investigated the combination of definitive chemoradiotherapy with ICIs, including durvalumab (NCT04550260), pembrolizumab (NCT04210115), tislelizumab (NCT03957590), and tiragolumab plus atezolizumab (NCT04543617).
Conclusion
ESCC is a predominant histological subtype of EC, and the unsatisfying prognosis highlights the clinical need for effective therapeutic compounds. Since ESCC shares molecular features specific to the squamous cell lineage, ESCC may benefit from agents effective against other SCCs across organs such as the lung, cervix, and head and neck. In addition, the complexity of spatial and intratumoral heterogeneity likely hampers the efficacy of targeted therapies. Therefore, the identification of a population vulnerable to specific molecular inhibition may pave the way for establishing personalized molecular-targeted medicine for ESCC patients.
Acknowledgments
None.
Footnotes
ORCID iDs: Akira Ooki
https://orcid.org/0000-0001-7618-5775
Hiroki Osumi
https://orcid.org/0000-0002-4742-0446
Contributor Information
Akira Ooki, Department of Gastroenterological Chemotherapy, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan.
Hiroki Osumi, Department of Gastroenterological Chemotherapy, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan.
Keisho Chin, Department of Gastroenterological Chemotherapy, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan.
Masayuki Watanabe, Department of Gastroenterological Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan.
Kensei Yamaguchi, Department of Gastroenterological Chemotherapy, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan.
Declarations
Ethics approval and consent to participate: Not applicable.
Consent for publication: Not applicable.
Author contributions: Akira Ooki: Conceptualization; Investigation; Writing – original draft.
Hiroki Osumi: Writing – review & editing.
Keisho Chin: Writing – review & editing.
Masayuki Watanabe: Writing – review & editing.
Kensei Yamaguchi: Writing – review & editing.
Funding: The authors received no financial support for the research, authorship, and/or publication of this article.
AO received speaker honoraria from Merck Serono, Bristol-Myers Squibb, Ono Pharmaceutical, Daiichi Sankyo, Chugai Pharmaceutical Co. Ltd, and Takeda Pharmaceutical Co. Ltd. KC received speaker honoraria from Taiho Pharmaceutical Co., Ono Pharmaceutical, Merck Serono, Bristol-Myers Squibb, and Chugai Pharmaceutical Co. Ltd. KY received speaker honoraria from Chugai Pharmaceutical Co. Ltd., Bristol-Myers Squibb, Merck Serono, Taiho Pharmaceutical Co., Takeda, and Eli Lilly; a consultant fee from Takeda Pharmaceutical Co. Ltd.; honoraria from Tsumura Co. Ltd., Nihon Kayaku Co. Ltd., and Chugai Pharmaceutical Co. Ltd; research grants from Sumitomo Dainippon Pharma, Gilead Sciences, MSD, and Boehringer Ingelheim, Daiichi Sankyo, and Chugai Pharmaceutical Co. Ltd; Speaker honoraria, research grants, and scholarship grant from Ono Pharmaceutical, Yakult Honsha Co., Ltd., and Sanofi.
Availability of data and materials: Not applicable.
References
- 1. Sung H, Ferlay J, Siegel RL, et al. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2021; 71: 209–249. [DOI] [PubMed] [Google Scholar]
- 2. The National Comprehensive Cancer Network. NCCN clinical practice guidelines in oncology, Esophageal and Esophagogastric Junction Cancer. Version 1. https://www.nccn.org/professionals/physician_gls/pdf/esophageal.pdf (2022, accessed 1 May 2022).
- 3. Uhlenhopp DJ, Then EO, Sunkara T, et al. Epidemiology of esophageal cancer: update in global trends, etiology and risk factors. Clin J Gastroenterol 2020; 13: 1010–1021. [DOI] [PubMed] [Google Scholar]
- 4. Abnet CC, Arnold M, Wei WQ. Epidemiology of esophageal squamous cell carcinoma. Gastroenterology 2018; 154: 360–373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Coleman HG, Xie SH, Lagergren J. The epidemiology of esophageal adenocarcinoma. Gastroenterology 2018; 154: 390–405. [DOI] [PubMed] [Google Scholar]
- 6. Smyth EC, Lagergren J, Fitzgerald RC, et al. Oesophageal cancer. Nat Rev Dis Primers 2017; 3: 17048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Arnold M, Ferlay J, van Berge Henegouwen MI, et al. Global burden of oesophageal and gastric cancer by histology and subsite in 2018. Gut 2020; 69: 1564–1571. [DOI] [PubMed] [Google Scholar]
- 8. Chang J, Tan W, Ling Z, et al. Genomic analysis of oesophageal squamous-cell carcinoma identifies alcohol drinking-related mutation signature and genomic alterations. Nat Commun 2017; 8: 15290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Lin DC, Dinh HQ, Xie JJ, et al. Identification of distinct mutational patterns and new driver genes in oesophageal squamous cell carcinomas and adenocarcinomas. Gut 2018; 67: 1769–1779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Lin DC, Hao JJ, Nagata Y, et al. Genomic and molecular characterization of esophageal squamous cell carcinoma. Nat Genet 2014; 46: 467–473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Lin EW, Karakasheva TA, Lee DJ, et al. Comparative transcriptomes of adenocarcinomas and squamous cell carcinomas reveal molecular similarities that span classical anatomic boundaries. PLoS Genet 2017; 13: e1006938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Gao YB, Chen ZL, Li JG, et al. Genetic landscape of esophageal squamous cell carcinoma. Nat Genet 2014; 46: 1097–1102. [DOI] [PubMed] [Google Scholar]
- 13. The Cancer Genome Atlas Research Network. Integrated genomic characterization of oesophageal carcinoma. Nature 2017; 541: 169–175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature 2014; 513: 202–209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. van Hagen P, Hulshof MC, van Lanschot JJ, et al. Preoperative chemoradiotherapy for esophageal or junctional cancer. New Engl J Med 2012; 366: 2074–2084. [DOI] [PubMed] [Google Scholar]
- 16. Njei B, McCarty TR, Birk JW. Trends in esophageal cancer survival in United States adults from 1973 to 2009: a SEER database analysis. J Gastroenterol Hepatol 2016; 31: 1141–1146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Siewert JR, Ott K. Are squamous and adenocarcinomas of the esophagus the same disease? Semin Radiat Oncol 2007; 17: 38–44. [DOI] [PubMed] [Google Scholar]
- 18. Kojima T, Shah MA, Muro K, et al. Randomized Phase III KEYNOTE-181 study of pembrolizumab versus chemotherapy in advanced esophageal cancer. J Clin Oncol 2020; 38: 4138–4148. [DOI] [PubMed] [Google Scholar]
- 19. Kato K, Cho BC, Takahashi M, et al. Nivolumab versus chemotherapy in patients with advanced oesophageal squamous cell carcinoma refractory or intolerant to previous chemotherapy (ATTRACTION-3): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol 2019; 20: 1506–1517. [DOI] [PubMed] [Google Scholar]
- 20. Huang J, Xu J, Chen Y, et al. 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. [DOI] [PubMed] [Google Scholar]
- 21. Shen L, Kato K, Kim S-B, et al. RATIONALE 302: randomized, phase 3 study of tislelizumab versus chemotherapy as second-line treatment for advanced unresectable/metastatic esophageal squamous cell carcinoma. J Clin Oncol 2021; 39: 4012–4012. [Google Scholar]
- 22. Zhang L, Zhou Y, Cheng C, et al. Genomic analyses reveal mutational signatures and frequently altered genes in esophageal squamous cell carcinoma. Am J Hum Genet 2015; 96: 597–611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Yan T, Cui H, Zhou Y, et al. Multi-region sequencing unveils novel actionable targets and spatial heterogeneity in esophageal squamous cell carcinoma. Nat Commun 2019; 10: 1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Sawada G, Niida A, Uchi R, et al. Genomic landscape of esophageal squamous cell carcinoma in a Japanese population. Gastroenterology 2016; 150: 1171–1182. [DOI] [PubMed] [Google Scholar]
- 25. Booka E, Tsubosa Y, Yokota T, et al. Whole exome sequencing and deep sequencing of esophageal squamous cell carcinoma and adenocarcinoma in Japanese patients using the Japanese version of the Genome Atlas, JCGA. Esophagus 2021; 18: 743–752. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Wang K, Johnson A, Ali SM, et al. Comprehensive genomic profiling of advanced esophageal squamous cell carcinomas and esophageal adenocarcinomas reveals similarities and differences. Oncologist 2015; 20: 1132–1139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Wang GQ, Abnet CC, Shen Q. Histological precursors of oesophageal squamous cell carcinoma: results from a 13 year prospective follow up study in a high risk population. Gut 2005; 54: 187–192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Wei WQ, Chen ZF, He YT, et al. Long-term Follow-Up of a community assignment, One-Time endoscopic screening study of esophageal cancer in China. J Clin Oncol 2015; 33: 1951–1957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Wang QL, Xie SH, Li WT, et al. Smoking cessation and risk of esophageal cancer by histological type: systematic review and meta-analysis. J Natl Cancer Inst 2017; 109: 1–10. [DOI] [PubMed] [Google Scholar]
- 30. Anderson LA, Watson RG, Murphy SJ. Risk factors for Barrett’s oesophagus and oesophageal adenocarcinoma: results from the FINBAR study. World J Gastroenterol 2007; 13: 1585–1594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Sharma P. Clinical practice. Barrett’s esophagus. New Engl J Med 2009; 361: 2548–2556. [DOI] [PubMed] [Google Scholar]
- 32. Then EO, Lopez M, Saleem S, et al. Esophageal cancer: an updated surveillance epidemiology and end results database analysis. World J Oncol 2020; 11: 55–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Ooki A, Yamashita K, Kobayashi N, et al. Lymph node metastasis density and growth pattern as independent prognostic factors in advanced esophageal squamous cell carcinoma. World J Surg 2007; 31: 2184–2191. [DOI] [PubMed] [Google Scholar]
- 34. Xi M, Yang Y, Zhang L, et al. Multi-institutional analysis of recurrence and survival after neoadjuvant chemoradiotherapy of esophageal cancer: impact of histology on recurrence patterns and outcomes. Ann Surg 2019; 269: 663–670. [DOI] [PubMed] [Google Scholar]
- 35. Allum WH, Stenning SP, Bancewicz J, et al. Long-term results of a randomized trial of surgery with or without preoperative chemotherapy in esophageal cancer. J Clin Oncol 2009; 27: 5062–5067. [DOI] [PubMed] [Google Scholar]
- 36. Yokoyama A, Kakiuchi N, Yoshizato T, et al. Age-related remodelling of oesophageal epithelia by mutated cancer drivers. Nature 2019; 565: 312–317. [DOI] [PubMed] [Google Scholar]
- 37. Chen XX, Zhong Q, Liu Y, et al. Genomic comparison of esophageal squamous cell carcinoma and its precursor lesions by multi-region whole-exome sequencing. Nat Commun 2017; 8: 524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Liu X, Zhang M, Ying S, et al. Genetic alterations in esophageal tissues from squamous dysplasia to carcinoma. Gastroenterology 2017; 153: 166–177. [DOI] [PubMed] [Google Scholar]
- 39. Hao JJ, Lin DC, Dinh HQ, et al. Spatial intratumoral heterogeneity and temporal clonal evolution in esophageal squamous cell carcinoma. Nat Genet 2016; 48: 1500–1507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Moody S, Senkin S, Islam SMA. Mutational signatures in esophageal squamous cell carcinoma from eight countries with varying incidence. Nat Genet 2021; 53: 1553–1563. [DOI] [PubMed] [Google Scholar]
- 41. Li XC, Wang MY, Yang M, et al. A mutational signature associated with alcohol consumption and prognostically significantly mutated driver genes in esophageal squamous cell carcinoma. Ann Oncol 2018; 29: 938–944. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Paulson TG, Reid BJ. Focus on Barrett’s esophagus and esophageal adenocarcinoma. Cancer Cell 2004; 6: 11–16. [DOI] [PubMed] [Google Scholar]
- 43. Stachler MD, Taylor-Weiner A, Peng S, et al. Paired exome analysis of Barrett’s esophagus and adenocarcinoma. Nat Genet 2015; 47: 1047–1055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Scott SJ, Li X, Jammula S, et al. Evidence that polyploidy in esophageal adenocarcinoma originates from mitotic slippage caused by defective chromosome attachments. Cell Death Differ 2021; 28: 2179–2193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Xiong T, Wang M, Zhao J, et al. An esophageal squamous cell carcinoma classification system that reveals potential targets for therapy. Oncotarget 2017; 8: 49851–49860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Ma M, Chen Y, Chong X, et al. Integrative analysis of genomic, epigenomic and transcriptomic data identified molecular subtypes of esophageal carcinoma. Aging 2021; 13: 6999–7019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Chen Z, Zhao M, Liang J, et al. Dissecting the single-cell transcriptome network underlying esophagus non-malignant tissues and esophageal squamous cell carcinoma. EBioMedicine 2021; 69: 103459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Song Y, Li L, Ou Y, et al. Identification of genomic alterations in oesophageal squamous cell cancer. Nature 2014; 509: 91–95. [DOI] [PubMed] [Google Scholar]
- 49. Secrier M, Li X, de Silva N, et al. Mutational signatures in esophageal adenocarcinoma define etiologically distinct subgroups with therapeutic relevance. Nat Genet 2016; 48: 1131–1141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Dulak AM, Stojanov P, Peng S, et al. Exome and whole-genome sequencing of esophageal adenocarcinoma identifies recurrent driver events and mutational complexity. Nat Genet 2013; 45: 478–486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Frankell AM, Jammula S, Li X, et al. The landscape of selection in 551 esophageal adenocarcinomas defines genomic biomarkers for the clinic. Nat Genet 2019; 51: 506–516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Naseri A, Salehi-Pourmehr H, Majidazar R, et al. Systematic Review and meta-analysis of the most common genetic mutations in esophageal squamous cell carcinoma. J Gastrointest Cancer 2022; 53: 1040–1049. [DOI] [PubMed] [Google Scholar]
- 53. Sun JM, Shen L, Shah MA, et al. Pembrolizumab plus chemotherapy versus chemotherapy alone for first-line treatment of advanced oesophageal cancer (KEYNOTE-590): a randomised, placebo-controlled, phase 3 study. Lancet 2021; 398: 759–771. [DOI] [PubMed] [Google Scholar]
- 54. Wang ZX, Cui C, Yao J. 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.e273. [DOI] [PubMed] [Google Scholar]
- 55. Salem ME, Puccini A, Xiu J, et al. Comparative molecular analyses of esophageal squamous cell carcinoma, esophageal adenocarcinoma, and gastric adenocarcinoma. Oncologist 2018; 23: 1319–1327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Gibault L, Metges JP, Conan-Charlet V, et al. Diffuse EGFR staining is associated with reduced overall survival in locally advanced oesophageal squamous cell cancer. Br J Cancer 2005; 93: 107–115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Musgrove EA, Caldon CE, Barraclough J, et al. Cyclin D as a therapeutic target in cancer. Nat Rev Cancer 2011; 11: 558–572. [DOI] [PubMed] [Google Scholar]
- 58. Lin DC, Wang MR, Koeffler HP. Genomic and epigenomic aberrations in esophageal squamous cell carcinoma and implications for patients. Gastroenterology 2018; 154: 374–389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Valletta D, Czech B, Spruss T, et al. Regulation and function of the atypical cadherin FAT1 in hepatocellular carcinoma. Carcinogenesis 2014; 35: 1407–1415. [DOI] [PubMed] [Google Scholar]
- 60. Hu X, Zhai Y, Shi R, et al. FAT1 inhibits cell migration and invasion by affecting cellular mechanical properties in esophageal squamous cell carcinoma. Oncol Rep 2018; 39: 2136–2146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Lu Y, Wang Z, Zhou L, et al. FAT1 and PTPN14 regulate the malignant progression and chemotherapy resistance of esophageal cancer through the Hippo Signaling Pathway. Anal Cell Pathol 2021; 2021: 9290372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. James V, Zhang Y, Foxler DE, et al. LIM-domain proteins, LIMD1, ajuba, and WTIP are required for microRNA-mediated gene silencing. Proc Natl Acad Sci USA 2010; 107: 12499–12504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Schleicher K, Schramek D. AJUBA: a regulator of epidermal homeostasis and cancer. Exp Dermatol 2021; 30: 546–559. [DOI] [PubMed] [Google Scholar]
- 64. Tanaka I, Osada H, Fujii M, et al. LIM-domain protein AJUBA suppresses malignant mesothelioma cell proliferation via Hippo signaling cascade. Oncogene 2015; 34: 73–83. [DOI] [PubMed] [Google Scholar]
- 65. Lei Y, Chen L, Zhang G, et al. MicroRNAs target the Wnt/β-catenin signaling pathway to regulate epithelial-mesenchymal transition in cancer (Review). Oncol Rep 2020; 44: 1299–1313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Kishino T, Niwa T, Yamashita S, et al. Integrated analysis of DNA methylation and mutations in esophageal squamous cell carcinoma. Mol Carcinog 2016; 55: 2077–2088. [DOI] [PubMed] [Google Scholar]
- 67. Cao W, Lee H, Wu W, et al. Multi-faceted epigenetic dysregulation of gene expression promotes esophageal squamous cell carcinoma. Nat Commun 2020; 11: 3675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Dang CV. MYC on the path to cancer. Cell 2012; 149: 22–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Lian Y, Niu X, Cai H, et al. Clinicopathological significance of c-MYC in esophageal squamous cell carcinoma. Tumour Biol 2017; 39: 1010428317715804. [DOI] [PubMed] [Google Scholar]
- 70. Li J, Liang Y, Lv H, et al. miR-26a and miR-26b inhibit esophageal squamous cancer cell proliferation through suppression of c-MYC pathway. Gene 2017; 625: 1–9. [DOI] [PubMed] [Google Scholar]
- 71. Zhang HF, Wu C, Alshareef A, et al. The PI3K/AKT/c-MYC Axis promotes the acquisition of cancer stem-like features in esophageal squamous cell carcinoma. Stem Cells 2016; 34: 2040–2051. [DOI] [PubMed] [Google Scholar]
- 72. Hirata H, Niida A, Kakiuchi N, et al. The evolving genomic landscape of esophageal squamous cell carcinoma under chemoradiotherapy. Cancer Res 2021; 81: 4926–4938. [DOI] [PubMed] [Google Scholar]
- 73. Jiang YY, Jiang Y, Li CQ, et al. TP63, SOX2, and KLF5 establish a core regulatory circuitry that controls epigenetic and transcription patterns in esophageal squamous cell carcinoma cell lines. Gastroenterology 2020; 159: 1311–1327.e19. [DOI] [PubMed] [Google Scholar]
- 74. Bass AJ, Watanabe H, Mermel CH, et al. SOX2 is an amplified lineage-survival oncogene in lung and esophageal squamous cell carcinomas. Nat Genet 2009; 41: 1238–1242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Jiang Y, Jiang YY, Xie JJ, et al. Co-activation of super-enhancer-driven CCAT1 by TP63 and SOX2 promotes squamous cancer progression. Nat Commun 2018; 9: 3619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Watanabe H, Ma Q, Peng S, et al. SOX2 and p63 colocalize at genetic loci in squamous cell carcinomas. J Clin Investig 2014; 124: 1636–1645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Gen Y, Yasui K, Nishikawa T, et al. SOX2 promotes tumor growth of esophageal squamous cell carcinoma through the AKT/mammalian target of rapamycin complex 1 signaling pathway. Cancer Sci 2013; 104: 810–816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Yang HL, Xu C, Yang YK, et al. ZNF750 exerted its antitumor action in oral squamous cell carcinoma by regulating E2F2. J Cancer 2021; 12: 7266–7276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Hazawa M, Lin DC, Handral H, et al. ZNF750 is a lineage-specific tumour suppressor in squamous cell carcinoma. Oncogene 2017; 36: 2243–2254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Ana Choi SS, Ko JM, Yu VZ, et al. Differentiation-related zinc finger protein 750 suppresses cell growth in esophageal squamous cell carcinoma. Oncol Lett 2021; 22: 513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Ohashi S, Natsuizaka M, Yashiro-Ohtani Y, et al. NOTCH1 and NOTCH3 coordinate esophageal squamous differentiation through a CSL-dependent transcriptional network. Gastroenterology 2010; 139: 2113–2123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Wu H, Yu J, Li Y, et al. Single-cell RNA sequencing reveals diverse intratumoral heterogeneities and gene signatures of two types of esophageal cancers. Cancer Lett 2018; 438: 133–143. [DOI] [PubMed] [Google Scholar]
- 83. Zhao Y, Lu Q, Li C, et al. PRMT1 regulates the tumour-initiating properties of esophageal squamous cell carcinoma through histone H4 arginine methylation coupled with transcriptional activation. Cell Death Dis 2019; 10: 359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Natsuizaka M, Whelan KA, Kagawa S, et al. Interplay between Notch1 and notch3 promotes EMT and tumor initiation in squamous cell carcinoma. Nat Commun 2017; 8: 1758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Nguyen BC, Lefort K, Mandinova A, et al. Cross-regulation between Notch and p63 in keratinocyte commitment to differentiation. Genes Dev 2006; 20: 1028–1042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Gall Trošelj K, Tomljanović M, Jaganjac M, et al. Oxidative stress and cancer heterogeneity orchestrate NRF2 roles relevant for therapy response. Molecules 2022; 27: 1468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Shibata T, Kokubu A, Saito S, et al. NRF2 mutation confers malignant potential and resistance to chemoradiation therapy in advanced esophageal squamous cancer. Neoplasia 2011; 13: 864–873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Kim YR, Oh JE, Kim MS, et al. Oncogenic NRF2 mutations in squamous cell carcinomas of oesophagus and skin. J Pathol 2010; 220: 446–451. [DOI] [PubMed] [Google Scholar]
- 89. Nakamura Y, Taniguchi H, Ikeda M. Clinical utility of circulating tumor DNA sequencing in advanced gastrointestinal cancer: SCRUM-Japan GI-SCREEN and GOZILA studies. Nat Med 2020; 26: 1859–1864. [DOI] [PubMed] [Google Scholar]
- 90. Clarke MF. Clinical and therapeutic implications of Cancer Stem Cells. N Engl J Med 2019; 380: 2237–2245. [DOI] [PubMed] [Google Scholar]
- 91. Prasetyanti PR, Medema JP. Intra-tumor heterogeneity from a cancer stem cell perspective. Mol Cancer 2017; 16: 41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Beck B, Blanpain C. Unravelling cancer stem cell potential. Nat Rev Cancer 2013; 13: 727–738. [DOI] [PubMed] [Google Scholar]
- 93. Holohan C, Van Schaeybroeck S, Longley DB, et al. Cancer drug resistance: an evolving paradigm. Nat Rev Cancer 2013; 13: 714–726. [DOI] [PubMed] [Google Scholar]
- 94. Batlle E, Clevers H. Cancer stem cells revisited. Nat Med 2017; 23: 1124–1134. [DOI] [PubMed] [Google Scholar]
- 95. Liu A, Zhu J, Wu G, et al. Antagonizing miR-455-3p inhibits chemoresistance and aggressiveness in esophageal squamous cell carcinoma. Mol Cancer 2017; 16: 106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Zhao Y, Zhu J, Shi B, et al. The transcription factor LEF1 promotes tumorigenicity and activates the TGF-β signaling pathway in esophageal squamous cell carcinoma. J Exp Clin Cancer Res 2019; 38: 304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Qiao Y, Zhang C, Li A, et al. IL6 derived from cancer-associated fibroblasts promotes chemoresistance via CXCR7 in esophageal squamous cell carcinoma. Oncogene 2018; 37: 873–883. [DOI] [PubMed] [Google Scholar]
- 98. Chen MF, Chen PT, Lu MS, et al. IL-6 expression predicts treatment response and outcome in squamous cell carcinoma of the esophagus. Mol Cancer 2013; 12: 26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Long A, Giroux V, Whelan KA, et al. WNT10A promotes an invasive and self-renewing phenotype in esophageal squamous cell carcinoma. Carcinogenesis 2015; 36: 598–606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Chen Y, Li D, Wang D, et al. Quiescence and attenuated DNA damage response promote survival of esophageal cancer stem cells. J Cell Biochem 2012; 113: 3643–3652. [DOI] [PubMed] [Google Scholar]
- 101. Wu H, Li Y, Hou Q, et al. Single-cell intratumoral stemness analysis reveals the involvement of cell cycle and DNA damage repair in two different types of esophageal cancer. Oncol Rep 2019; 41: 3201–3208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Dongre A, Weinberg RA. New insights into the mechanisms of epithelial-mesenchymal transition and implications for cancer. Nat Rev Mol Cell Biol 2019; 20: 69–84. [DOI] [PubMed] [Google Scholar]
- 103. Lin EW, Karakasheva TA, Hicks PD, et al. The tumor microenvironment in esophageal cancer. Oncogene 2016; 35: 5337–5349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Isohata N, Aoyagi K, Mabuchi T, et al. Hedgehog and epithelial-mesenchymal transition signaling in normal and malignant epithelial cells of the esophagus. Int J Cancer 2009; 125: 1212–1221. [DOI] [PubMed] [Google Scholar]
- 105. Noma K, Smalley KS, Lioni M, et al. The essential role of fibroblasts in esophageal squamous cell carcinoma-induced angiogenesis. Gastroenterology 2008; 134: 1981–1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Zhang H, Qin G, Zhang C, et al. TRAIL promotes epithelial-to-mesenchymal transition by inducing PD-L1 expression in esophageal squamous cell carcinomas. J Exp Clin Cancer Res 2021; 40: 209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Lin C, Wang Y, Wang Y, et al. Transcriptional and posttranscriptional regulation of HOXA13 by lncRNA HOTTIP facilitates tumorigenesis and metastasis in esophageal squamous carcinoma cells. Oncogene 2017; 36: 5392–5406. [DOI] [PubMed] [Google Scholar]
- 108. Lin C, Zhang S, Wang Y, et al. Functional role of a novel long noncoding RNA TTN-AS1 in esophageal squamous cell carcinoma progression and metastasis. Clin Cancer Res 2018; 24: 486–498. [DOI] [PubMed] [Google Scholar]
- 109. Gao Y, Yi J, Zhang K, et al. Downregulation of MiR-31 stimulates expression of LATS2 via the hippo pathway and promotes epithelial-mesenchymal transition in esophageal squamous cell carcinoma. J Exp Clin Cancer Res 2017; 36: 161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Karakasheva TA, Lin EW, Tang Q, et al. IL-6 mediates cross-talk between tumor cells and activated fibroblasts in the tumor microenvironment. Cancer Res 2018; 78: 4957–4970. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Scadden DT. The stem-cell niche as an entity of action. Nature 2006; 441: 1075–1079. [DOI] [PubMed] [Google Scholar]
- 112. Vahidian F, Duijf PHG, Safarzadeh E, et al. Interactions between cancer stem cells, immune system and some environmental components: friends or foes? Immunol Lett 2019; 208: 19–29. [DOI] [PubMed] [Google Scholar]
- 113. Bellmunt J, Paz-Ares L, Cuello M, et al. Gene expression of ERCC1 as a novel prognostic marker in advanced bladder cancer patients receiving cisplatin-based chemotherapy. Ann Oncol 2007; 18: 522–528. [DOI] [PubMed] [Google Scholar]
- 114. Zhuo ZG, Zhu YK, Deng HY, et al. Predictive value of excision repair cross-complementation Group 1 in the response to platinum-based chemotherapy in esophageal cancer: a meta-analysis. Oncol Res Treat 2020; 43: 160–169. [DOI] [PubMed] [Google Scholar]
- 115. Iizuka T, Kakegawa T, Ide H, et al. Phase II evaluation of cisplatin and 5-fluorouracil in advanced squamous cell carcinoma of the esophagus: a Japanese esophageal oncology group trial. Jpn J Clin Oncol 1992; 22: 172–176. [PubMed] [Google Scholar]
- 116. Bleiberg H, Conroy T, Paillot B, et al. Randomised phase II study of cisplatin and 5-fluorouracil (5-FU) versus cisplatin alone in advanced squamous cell oesophageal cancer. Eur J Cancer 1997; 33: 1216–1220. [DOI] [PubMed] [Google Scholar]
- 117. Zhang X, Shen L, Li J, et al. A phase II trial of paclitaxel and cisplatin in patients with advanced squamous-cell carcinoma of the esophagus. Am J Clin Oncol 2008; 31: 29–33. [DOI] [PubMed] [Google Scholar]
- 118. Huang J, Zhou Y, Zhang H, et al. A phase II study of biweekly paclitaxel and cisplatin chemotherapy for recurrent or metastatic esophageal squamous cell carcinoma: ERCC1 expression predicts response to chemotherapy. Med Oncol 2013; 30: 343. [DOI] [PubMed] [Google Scholar]
- 119. El-Rayes BF, Shields A, Zalupski M, et al. A phase II study of carboplatin and paclitaxel in esophageal cancer. Ann Oncol 2004; 15: 960–965. [DOI] [PubMed] [Google Scholar]
- 120. Kim JY, Do YR, Park KU, et al.; Do YR. A multi-center phase II study of docetaxel plus cisplatin as first-line therapy in patients with metastatic squamous cell esophageal cancer. Cancer Chemother Pharmacol 2010; 66: 31–36. [DOI] [PubMed] [Google Scholar]
- 121. Huang J, Fan QX, Chen L, et al. Long-term outcomes of gemcitabine and cisplatin in patients with recurrent or metastatic esophageal squamous cell carcinoma: a phase II trial. Chin Med J 2011; 124: 4012–4017. [PubMed] [Google Scholar]
- 122. Lee DH, Kim HT, Han JY, et al. A phase II trial of modified weekly irinotecan and cisplatin for chemotherapy-naïve patients with metastatic or recurrent squamous cell carcinoma of the esophagus. Cancer Chemother Pharmacol 2008; 61: 83–88. [DOI] [PubMed] [Google Scholar]
- 123. Kim M, Keam B, Kim TM, et al. Phase II study of irinotecan and cisplatin combination chemotherapy in metastatic, unresectable esophageal cancer. Cancer Res Treat 2017; 49: 416–422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Kaechele V, Moehler M, Lutz MP, et al. A phase I/II study of oxaliplatin and paclitaxel in patients with non-resectable cancer of the oesophagus and adenocarcinoma of the gastro-oesophageal junction: a study of the Arbeitsgemeinschaft internistische Onkologie. Cancer Chemother Pharmacol 2010; 66: 191–195. [DOI] [PubMed] [Google Scholar]
- 125. Wang J, Chang J, Yu H, et al. A phase II study of oxaliplatin in combination with leucovorin and fluorouracil as first-line chemotherapy in patients with metastatic squamous cell carcinoma of esophagus. Cancer Chemother Pharmacol 2013; 71: 905–911. [DOI] [PubMed] [Google Scholar]
- 126. Wolff K, Wein A, Reulbach U, et al. Weekly high-dose 5-fluorouracil as a 24-h infusion and sodium folinic acid (AIO regimen) plus irinotecan in patients with locally advanced nonresectable and metastatic adenocarcinoma or squamous cell carcinoma of the oesophagus: a phase II trial. Anticancer Drugs 2009; 20: 165–173. [DOI] [PubMed] [Google Scholar]
- 127. Ilson DH, Ajani J, Bhalla K, et al. Phase II trial of paclitaxel, fluorouracil, and cisplatin in patients with advanced carcinoma of the esophagus. J Clin Oncol 1998; 16: 1826–1834. [DOI] [PubMed] [Google Scholar]
- 128. Hironaka S, Tsubosa Y, Mizusawa J, et al. Phase I/II trial of 2-weekly docetaxel combined with cisplatin plus fluorouracil in metastatic esophageal cancer (JCOG0807). Cancer Sci 2014; 105: 1189–1195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Ojima T, Nakamori M, Nakamura M, et al. Phase I/II study of divided-dose docetaxel, cisplatin and fluorouracil for patients with recurrent or metastatic squamous cell carcinoma of the esophagus. Dis Esophagus 2017; 30: 1–7. [DOI] [PubMed] [Google Scholar]
- 130. Muro K, Hamaguchi T, Ohtsu A, et al. A phase II study of single-agent docetaxel in patients with metastatic esophageal cancer. Ann Oncol 2004; 15: 955–959. [DOI] [PubMed] [Google Scholar]
- 131. Kato K, Tahara M, Hironaka S, et al. A phase II study of paclitaxel by weekly 1-h infusion for advanced or recurrent esophageal cancer in patients who had previously received platinum-based chemotherapy. Cancer Chemother Pharmacol 2011; 67: 1265–1272. [DOI] [PubMed] [Google Scholar]
- 132. Mühr-Wilkenshoff F, Hinkelbein W, Ohnesorge I, et al. A pilot study of irinotecan (CPT-11) as single-agent therapy in patients with locally advanced or metastatic esophageal carcinoma. Int J Colorectal Dis 2003; 18: 330–334. [DOI] [PubMed] [Google Scholar]
- 133. Burkart C, Bokemeyer C, Klump B, et al. A phase II trial of weekly irinotecan in cisplatin-refractory esophageal cancer. Anticancer Res 2007; 27: 2845–2848. [PubMed] [Google Scholar]
- 134. Müller M, Posch F, Kiem D, et al. Benefit of second-line therapy for advanced esophageal squamous cell carcinoma: a tri-center propensity score analysis. Ther Adv Med Oncol 2021; 13: 17588359211039930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity 2013; 39: 1–10. [DOI] [PubMed] [Google Scholar]
- 136. Ooki A, Shinozaki E, Yamaguchi K. Immunotherapy in colorectal cancer: Current and Future Strategies. J Anus Rectum Colon 2021; 5: 11–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Teng MW, Ngiow SF, Ribas A, et al. Classifying cancers based on T-cell infiltration and PD-L1. Cancer Res 2015; 75: 2139–2145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Xu RH, Wang F, Cui C, et al. 1373MO JUPITER-06: A randomized, double-blind, phase III study of toripalimab versus placebo in combination with first-line chemotherapy for treatment naive advanced or metastatic esophageal squamous cell carcinoma (ESCC). Ann Oncol 2021; 32: S1041. [Google Scholar]
- 139. Qu HX, Zhao LP, Zhan SH, et al. Clinicopathological and prognostic significance of programmed cell death ligand 1 (PD-L1) expression in patients with esophageal squamous cell carcinoma: a meta-analysis. J Thorac Dis 2016; 8: 3197–3204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Doi T, Piha-Paul SA, Jalal SI, et al. Safety and antitumor activity of the anti-programmed death-1 antibody pembrolizumab in patients with advanced esophageal carcinoma. J Clin Oncol 2018; 36: 61–67. [DOI] [PubMed] [Google Scholar]
- 141. Shah MA, Kojima T, Hochhauser D, et al. Efficacy and safety of pembrolizumab for heavily pretreated patients with advanced, metastatic adenocarcinoma or squamous cell carcinoma of the esophagus: the Phase 2 KEYNOTE-180 study. JAMA Oncol 2019; 5: 546–550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Kudo T, Hamamoto Y, Kato K, et al. Nivolumab treatment for oesophageal squamous-cell carcinoma: an open-label, multicentre, phase 2 trial. Lancet Oncol 2017; 18: 631–639. [DOI] [PubMed] [Google Scholar]
- 143. Doki Y, Ajani JA, Kato K, et al. Nivolumab combination therapy in advanced esophageal squamous-cell carcinoma. New Engl J Med 2022; 386: 449–462. [DOI] [PubMed] [Google Scholar]
- 144. Zhang B, Qi L, Wang X, et al. Phase II clinical trial using camrelizumab combined with apatinib and chemotherapy as the first-line treatment of advanced esophageal squamous cell carcinoma. Cancer Commun 2020; 40: 711–720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Luo H, Lu J, Bai Y, et al. Effect of camrelizumab vs Placebo added to chemotherapy on survival and progression-free survival in patients with advanced or metastatic esophageal squamous cell carcinoma: the ESCORT-1st Randomized Clinical Trial. JAMA 2021; 326: 916–925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Xu J, Bai Y, Xu N, et al. Tislelizumab plus chemotherapy as first-line treatment for advanced esophageal squamous cell carcinoma and gastric/gastroesophageal junction adenocarcinoma. Clin Cancer Res 2020; 26: 4542–4550. [DOI] [PubMed] [Google Scholar]
- 147. Yoon H, Kato K, Raymond E, et al. LBA-1 RATIONALE-306: Randomized, global, placebo-controlled, double-blind phase 3 study of tislelizumab plus chemotherapy versus chemotherapy as first-line treatment for advanced or metastatic esophageal squamous cell carcinoma (ESCC). Ann Oncol 2022; 33: S375. [DOI] [PubMed] [Google Scholar]
- 148. Xu J, Li Y, Fan Q, et al. Clinical and biomarker analyses of sintilimab versus chemotherapy as second-line therapy for advanced or metastatic esophageal squamous cell carcinoma: a randomized, open-label phase 2 study (ORIENT-2). Nat Commun 2022; 13: 857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Shen L, Lu ZH, Wang JY, et al. LBA52 Sintilimab plus chemotherapy versus chemotherapy as first-line therapy in patients with advanced or metastatic esophageal squamous cell cancer: first results of the phase III ORIENT-15 study. Ann Oncol 2021; 32: S1330. [Google Scholar]
- 150. Xu J, Liu R, Zhang Y, et al. Efficacy and safety of KN046 plus paclitaxel/cisplatin as first-line treatment for unresectable locally advanced, recurrent or metastatic esophageal squamous cell carcinoma (ESCC). J Clin Oncol 2021; 39: 4062–4062. [Google Scholar]
- 151. Galluzzi L, Buqué A, Kepp O, et al. Immunological effects of conventional chemotherapy and targeted anticancer agents. Cancer Cell 2015; 28: 690–714. [DOI] [PubMed] [Google Scholar]
- 152. Tsuchikawa T, Miyamoto M, Yamamura Y, et al. The immunological impact of neoadjuvant chemotherapy on the tumor microenvironment of esophageal squamous cell carcinoma. Ann Surg Oncol 2012; 19: 1713–1719. [DOI] [PubMed] [Google Scholar]
- 153. Akagi K, Oki E, Taniguchi H, et al. Nationwide large-scale investigation of microsatellite instability status in more than 18,000 patients with various advanced solid cancers. J Clin Oncol 2020; 38: 803–803. [Google Scholar]
- 154. Leone AG, Petrelli F, Ghidini A, et al. Efficacy and activity of PD-1 blockade in patients with advanced esophageal squamous cell carcinoma: a systematic review and meta-analysis with focus on the value of PD-L1 combined positive score. ESMO Open 2022; 7: 100380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Dinh HQ, Pan F, Wang G, et al. Integrated single-cell transcriptome analysis reveals heterogeneity of esophageal squamous cell carcinoma microenvironment. Nat Commun 2021; 12: 7335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Zheng Y, Chen Z, Han Y, et al. Immune suppressive landscape in the human esophageal squamous cell carcinoma microenvironment. Nat Commun 2020; 11: 6268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157. Zappasodi R, Merghoub T, Wolchok JD. Emerging concepts for immune checkpoint blockade-based combination therapies. Cancer Cell 2018; 34: 690–598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Kojima T, Yamazaki K, Kato K, et al. Phase I dose-escalation trial of Sym004, an anti-EGFR antibody mixture, in Japanese patients with advanced solid tumors. Cancer Sci 2018; 109: 3253–3262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Liu R, Liu L, Zhao C, et al. Larotinib in patients with advanced and previously treated esophageal squamous cell carcinoma with epidermal growth factor receptor overexpression or amplification: an open-label, multicenter phase 1b study. BMC Gastroenterol 2021; 21: 398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160. Huang J, Fan Q, Lu P, et al. Icotinib in patients with pretreated advanced esophageal squamous cell carcinoma with EGFR overexpression or EGFR gene amplification: a single-arm, multicenter Phase 2 Study. J Thorac Oncol 2016; 11: 910–917. [DOI] [PubMed] [Google Scholar]
- 161. Hong MH, Heo SG, Lee Y, et al. Phase 2 study of afatinib among patients with recurrent and/or metastatic esophageal squamous cell carcinoma. Cancer 2020; 126: 4521–4531. [DOI] [PubMed] [Google Scholar]
- 162. Ilson DH, Kelsen D, Shah M, et al. A phase 2 trial of erlotinib in patients with previously treated squamous cell and adenocarcinoma of the esophagus. Cancer 2011; 117: 1409–1414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. Janmaat ML, Gallegos-Ruiz MI, Rodriguez JA, et al. Predictive factors for outcome in a phase II study of gefitinib in second-line treatment of advanced esophageal cancer patients. J Clin Oncol 2006; 24: 1612–1619. [DOI] [PubMed] [Google Scholar]
- 164. Dutton SJ, Ferry DR, Blazeby JM, et al. Gefitinib for oesophageal cancer progressing after chemotherapy (COG): a phase 3, multicentre, double-blind, placebo-controlled randomised trial. Lancet Oncol 2014; 15: 894–904. [DOI] [PubMed] [Google Scholar]
- 165. Lu M, Wang X, Shen L, et al. Nimotuzumab plus paclitaxel and cisplatin as the first line treatment for advanced esophageal squamous cell cancer: a single centre prospective phase II trial. Cancer Sci 2016; 107: 486–490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Lorenzen S, Schuster T, Porschen R, et al. Cetuximab plus cisplatin-5-fluorouracil versus cisplatin-5-fluorouracil alone in first-line metastatic squamous cell carcinoma of the esophagus: a randomized phase II study of the Arbeitsgemeinschaft Internistische Onkologie. Ann Oncol 2009; 20: 1667–1673. [DOI] [PubMed] [Google Scholar]
- 167. Moehler M, Maderer A, Thuss-Patience PC, et al. Cisplatin and 5-fluorouracil with or without epidermal growth factor receptor inhibition panitumumab for patients with non-resectable, advanced or metastatic oesophageal squamous cell cancer: a prospective, open-label, randomised phase III AIO/EORTC trial (POWER). Ann Oncol 2020; 31: 228–235. [DOI] [PubMed] [Google Scholar]
- 168. Kim HS, Kim SM, Kim H, et al. Phase II clinical and exploratory biomarker study of dacomitinib in recurrent and/or metastatic esophageal squamous cell carcinoma. Oncotarget 2015; 6: 44971–44984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169. Cohen SJ, Feng Y, Catalano PJ, et al. E2208: Randomized phase II study of paclitaxel with or without the anti-IGF-IR antibody cixutumumab (IMC-A12) as second-line treatment for patients with metastatic esophageal or GE junction cancer. J Clin Oncol 2014; 32: 4020–4020. [Google Scholar]
- 170. Kato K, Doi T, Kojima T, et al. Phase II study of BKM120 in patients with advanced esophageal squamous cell carcinoma (EPOC1303). J Clin Oncol 2017; 35: 4069–4069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Huang J, Xiao J, Fang W, et al. Anlotinib for previously treated advanced or metastatic esophageal squamous cell carcinoma: a double-blind randomized phase 2 trial. Cancer Med 2021; 10: 1681–1689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172. Chu L, Chen Y, Liu Q, et al. A Phase II study of apatinib in patients with chemotherapy-refractory esophageal squamous cell carcinoma (ESO-Shanghai 11). Oncologist 2021; 26: e925–e935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173. Yanwei L, Feng H, Ren P, et al. Safety and efficacy of apatinib monotherapy for unresectable, metastatic esophageal cancer: a Single-Arm, Open-Label, Phase II Study. Oncologist 2020; 25: e1464–e1472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Karasic TB, O’Hara MH, Teitelbaum UR, et al. Phase II trial of palbociclib in patients with advanced esophageal or gastric cancer. Oncologist 2020; 25: e1864–e1868. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175. Li JC, Zhao YH, Wang XY, et al. Clinical significance of the expression of EGFR signaling pathway-related proteins in esophageal squamous cell carcinoma. Tumour Biol 2014; 35: 651–657. [DOI] [PubMed] [Google Scholar]
- 176. Hanawa M, Suzuki S, Dobashi Y, et al. EGFR protein overexpression and gene amplification in squamous cell carcinomas of the esophagus. Int J Cancer 2006; 118: 1173–1180. [DOI] [PubMed] [Google Scholar]
- 177. Kato H, Arao T, Matsumoto K, et al. Gene amplification of EGFR, HER2, FGFR2 and MET in esophageal squamous cell carcinoma. Int J Oncol 2013; 42: 1151–1158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178. Yu WW, Guo YM, Zhu M, et al. Clinicopathological and prognostic significance of EGFR over-expression in esophageal squamous cell carcinoma: a meta-analysis. Hepatogastroenterology 2011; 58: 426–431. [PubMed] [Google Scholar]
- 179. Guo K, Wang WP, Jiang T, et al. Assessment of epidermal growth factor receptor mutation/copy number and K-ras mutation in esophageal cancer. J Thorac Dis 2016; 8: 1753–1763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180. Petty RD, Dahle-Smith A, Stevenson DAJ, et al. Gefitinib and EGFR gene copy number aberrations in esophageal cancer. J Clin Oncol 2017; 35: 2279–2287. [DOI] [PubMed] [Google Scholar]
- 181. Cui D, Zhu Y, Yan D, et al. Dual inhibition of cMET and EGFR by microRNA-338-5p suppresses metastasis of esophageal squamous cell carcinoma. Carcinogenesis 2021; 42: 995–1007. [DOI] [PubMed] [Google Scholar]
- 182. Han L, Cui D, Li B, et al. MicroRNA-338-5p reverses chemoresistance and inhibits invasion of esophageal squamous cell carcinoma cells by targeting Id-1. Cancer Sci 2019; 110: 3677–3688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183. Neijssen J, Cardoso RM, Chevalier KM, et al. Discovery of amivantamab (JNJ-61186372), a bispecific antibody targeting EGFR and MET. J Biol Chem 2021; 296: 100641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184. Park K, Haura EB, Leighl NB, et al. Amivantamab in EGFR exon 20 insertion–mutated Non–Small-Cell lung cancer progressing on platinum Chemotherapy: Initial results from the CHRYSALIS Phase I Study. J Clin Oncol 2021; 39: 3391–3402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185. Ward MD, Leahy DJ. Kinase activator-receiver preference in ErbB heterodimers is determined by intracellular regions and is not coupled to extracellular asymmetry. J Biol Chem 2015; 290: 1570–1579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186. Zhou J, Wu Z, Wong G, et al. CDK4/6 or MAPK blockade enhances efficacy of EGFR inhibition in oesophageal squamous cell carcinoma. Nat Commun 2017; 8: 13897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187. Dieci MV, Arnedos M, Andre F, et al. Fibroblast growth factor receptor inhibitors as a cancer treatment: from a biologic rationale to medical perspectives. Cancer Discov 2013; 3: 264–279. [DOI] [PubMed] [Google Scholar]
- 188. Helsten T, Elkin S, Arthur E, et al. The FGFR landscape in cancer: analysis of 4,853 tumors by next-generation sequencing. Clin Cancer Res 2016; 22: 259–267. [DOI] [PubMed] [Google Scholar]
- 189. Ooki A, Yamaguchi K. The beginning of the era of precision medicine for gastric cancer with fibroblast growth factor receptor 2 aberration. Gastric Cancer 2021; 24: 1169–1183. [DOI] [PubMed] [Google Scholar]
- 190. Wang Y, Wu Y, Li J, et al. Prognostic and clinicopathological significance of FGFR1 gene amplification in resected esophageal squamous cell carcinoma: a meta-analysis. Ann Transl Med 2019; 7: 669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191. Goyal L, Shi L, Liu LY, et al. TAS-120 overcomes resistance to ATP-competitive FGFR inhibitors in patients with FGFR2 fusion–positive intrahepatic cholangiocarcinoma. Cancer Discov 2019; 9: 1064–1079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192. He Y, Sun MM, Zhang GG, et al. Targeting PI3K/Akt signal transduction for cancer therapy. Signal Transduct Target Ther 2021; 6: 425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193. Fruman DA, Rommel C. PI3K and cancer: lessons, challenges and opportunities. Nat Rev Drug Discov 2014; 13: 140–156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194. Vasan N, Razavi P, Johnson JL, et al. Double PIK3CA mutations in cis increase oncogenicity and sensitivity to PI3Kα inhibitors. Science 2019; 366: 714–723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195. Li B, Tsao SW, Chan KW, et al. Id1-induced IGF-II and its autocrine/endocrine promotion of esophageal cancer progression and chemoresistance–implications for IGF-II and IGF-IR-targeted therapy. Clin Cancer Res 2014; 20: 2651–2662. [DOI] [PubMed] [Google Scholar]
- 196. Sulzmaier FJ, Jean C, Schlaepfer DD. FAK in cancer: mechanistic findings and clinical applications. Nat Rev Cancer 2014; 14: 598–610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197. Zhang L, Zhao D, Wang Y, et al. Focal adhesion kinase (FAK) inhibitor-defactinib suppresses the malignant progression of human esophageal squamous cell carcinoma (ESCC) cells via effective blockade of PI3K/AKT axis and downstream molecular network. Mol Carcinog 2021; 60: 113–124. [DOI] [PubMed] [Google Scholar]
- 198. Linger RM, Keating AK, Earp HS, et al. TAM receptor tyrosine kinases: biologic functions, signaling, and potential therapeutic targeting in human cancer. Adv Cancer Res 2008; 100: 35–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199. Hsieh MS, Yang PW, Wong LF, et al. The AXL receptor tyrosine kinase is associated with adverse prognosis and distant metastasis in esophageal squamous cell carcinoma. Oncotarget 2016; 7: 36956–36970. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200. Elkabets M, Pazarentzos E, Juric D, et al. AXL mediates resistance to PI3Kα inhibition by activating the EGFR/PKC/mTOR axis in head and neck and esophageal squamous cell carcinomas. Cancer Cell 2015; 27: 533–546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201. Goel HL, Mercurio AM. VEGF targets the tumour cell. Nat Rev Cancer 2013; 13: 871–882. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202. Jain RK. Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science 2005; 307: 58–62. [DOI] [PubMed] [Google Scholar]
- 203. Chen M, Cai E, Huang J, et al. Prognostic value of vascular endothelial growth factor expression in patients with esophageal cancer: A systematic review and meta-analysis. Cancer Epidemiol Biomarkers Prev 2012; 21: 1126–1134. [DOI] [PubMed] [Google Scholar]
- 204. Wang YM, Xu X, Tang J, et al. Apatinib induces endoplasmic reticulum stress-mediated apoptosis and autophagy and potentiates cell sensitivity to paclitaxel via the IRE-1α-AKT-mTOR pathway in esophageal squamous cell carcinoma. Cell Biosci 2021; 11: 124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205. Syed YY. Anlotinib: First Global Approval. Drugs 2018; 78: 1057–1062. [DOI] [PubMed] [Google Scholar]
- 206. Turner NC, Ro J, André F, et al. Palbociclib in hormone-receptor-positive advanced breast cancer. New Engl J Med 2015; 373: 209–219. [DOI] [PubMed] [Google Scholar]
- 207. Wang J, Li Q, Yuan J, et al. CDK4/6 inhibitor-SHR6390 exerts potent antitumor activity in esophageal squamous cell carcinoma by inhibiting phosphorylated Rb and inducing G1 cell cycle arrest. J Transl Med 2017; 15: 127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208. Qie S, Yoshida A, Parnham S, et al. Targeting glutamine-addiction and overcoming CDK4/6 inhibitor resistance in human esophageal squamous cell carcinoma. Nat Commun 2019; 10: 1296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209. Hayes JD, Dinkova-Kostova AT. The Nrf2 regulatory network provides an interface between redox and intermediary metabolism. Trends Biochem Sci 2014; 39: 199–218. [DOI] [PubMed] [Google Scholar]
- 210. Zhu M, Fahl WE. Functional characterization of transcription regulators that interact with the electrophile response element. Biochem Biophys Res Commun 2001; 289: 212–219. [DOI] [PubMed] [Google Scholar]
- 211. Fu J, Xiong Z, Huang C, et al. Hyperactivity of the transcription factor Nrf2 causes metabolic reprogramming in mouse esophagus. J Biol Chem 2019; 294: 327–340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212. Maher JM, Dieter MZ, Aleksunes LM, et al. Oxidative and electrophilic stress induces multidrug resistance-associated protein transporters via the nuclear factor-E2-related factor-2 transcriptional pathway. Hepatology 2007; 46: 1597–1610. [DOI] [PubMed] [Google Scholar]
- 213. Shen H, Yang Y, Xia S, et al. Blockage of Nrf2 suppresses the migration and invasion of esophageal squamous cell carcinoma cells in hypoxic microenvironment. Dis Esophagus 2014; 27: 685–692. [DOI] [PubMed] [Google Scholar]
- 214. Kawasaki Y, Okumura H, Uchikado Y, et al. Nrf2 is useful for predicting the effect of chemoradiation therapy on esophageal squamous cell carcinoma. Ann Surg Oncol 2014; 21: 2347–2352. [DOI] [PubMed] [Google Scholar]
- 215. Rojo de la Vega M, Dodson M, Chapman E, et al. NRF2-targeted therapeutics: new targets and modes of NRF2 regulation. Curr Opin Toxicol 2016; 1: 62–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216. Ren D, Villeneuve NF, Jiang T, et al. Brusatol enhances the efficacy of chemotherapy by inhibiting the nrf2-mediated defense mechanism. Proc Natl Acad Sci USA 2011; 108: 1433–1438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217. Tsuchida K, Tsujita T, Hayashi M, et al. Halofuginone enhances the chemo-sensitivity of cancer cells by suppressing NRF2 accumulation. Free Radic Biol Med 2017; 103: 236–247. [DOI] [PubMed] [Google Scholar]
- 218. Bollong MJ, Yun H, Sherwood L, et al. A small molecule inhibits deregulated NRF2 transcriptional activity in cancer. ACS Chem Biol 2015; 10: 2193–2198. [DOI] [PubMed] [Google Scholar]
- 219. Singh A, Venkannagari S, Oh KH, et al. Small molecule inhibitor of NRF2 selectively intervenes therapeutic resistance in KEAP1-deficient NSCLC tumors. ACS Chem Biol 2016; 11: 3214–3225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220. Arlt A, Sebens S, Krebs S, et al. Inhibition of the Nrf2 transcription factor by the alkaloid trigonelline renders pancreatic cancer cells more susceptible to apoptosis through decreased proteasomal gene expression and proteasome activity. Oncogene 2013; 32: 4825–4835. [DOI] [PubMed] [Google Scholar]
- 221. Paiboonrungruang C, Simpson E, Xiong Z, et al. Development of targeted therapy of NRF2(high) esophageal squamous cell carcinoma. Cell Signal 2021; 86: 110105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222. L Suraweera T, Rupasinghe HPV, Dellaire G, et al. Regulation of Nrf2/are pathway by dietary flavonoids: A friend or Foe for cancer management? Antioxidants 2020; 9: 973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223. Bar-Peled L, Kemper EK, Suciu RM, et al. Chemical Proteomics identifies druggable vulnerabilities in a genetically defined cancer. Cell 2017; 171: 696–709.e23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224. Matés JM, Campos-Sandoval JA, de Los Santos-Jiménez J, et al. Glutaminases regulate glutathione and oxidative stress in cancer. Arch Toxicol 2020; 94: 2603–2623. [DOI] [PubMed] [Google Scholar]
- 225. Galan-Cobo A, Sitthideatphaiboon P, Qu X, et al. LKB1 and KEAP1/NRF2 pathways cooperatively promote metabolic reprogramming with enhanced glutamine dependence in KRAS-Mutant lung adenocarcinoma. Cancer Res 2019; 79: 3251–3267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226. Shen YA, Hong J, Asaka R, et al. Inhibition of the MYC-Regulated glutaminase metabolic axis is an effective synthetic lethal approach for treating chemoresistant ovarian cancers. Cancer Res 2020; 80: 4514–4526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227. Romero R, Sayin VI, Davidson SM, et al. Keap1 loss promotes kras-driven lung cancer and results in dependence on glutaminolysis. Nat Med 2017; 23: 1362–1368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228. Binkley MS, Jeon YJ, Nesselbush M, et al. KEAP1/NFE2L2 mutations predict lung cancer radiation resistance that can Be targeted by glutaminase inhibition. Cancer Discov 2020; 10: 1826–1841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229. Motzer RJ, Lee CH, Emamekhoo H, et al. ENTRATA: randomized, double-blind, phase II study of telaglenastat (tela; CB-839) + everolimus (E) vs placebo (pbo) + E in patients (pts) with advanced/metastatic renal cell carcinoma (mRCC). Ann Oncol 2019; 30: v889–v890. [Google Scholar]
- 230. Varghese S, Pramanik S, Williams LJ, et al. The glutaminase inhibitor CB-839 (Telaglenastat) enhances the antimelanoma activity of T-Cell-Mediated immunotherapies. Mol Cancer Ther 2021; 20: 500–511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231. Johnson R, Halder G. The two faces of Hippo: targeting the Hippo pathway for regenerative medicine and cancer treatment. Nat Rev Drug Discov 2014; 13: 63–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232. Ooki A, Del Carmen Rodriguez Pena M, Marchionni L, et al. YAP1 and COX2 coordinately regulate urothelial cancer stem-like cells. Cancer Res 2018; 78: 168–181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233. Nguyen CDK, Yi C. YAP/TAZ Signaling and Resistance to Cancer Therapy. Trends Cancer 2019; 5: 283–296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234. Kapoor A, Yao W, Ying H. Yap1 activation enables bypass of oncogenic Kras addiction in pancreatic cancer. Cell 2014; 158: 185–197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235. Ciamporcero E, Shen H, Ramakrishnan S, et al. YAP activation protects urothelial cell carcinoma from treatment-induced DNA damage. Oncogene 2016; 35: 1541–1553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236. Shibata M, Ooki A, Inokawa Y, et al. Concurrent targeting of potential cancer stem cells regulating pathways sensitizes lung adenocarcinoma to standard chemotherapy. Mol Cancer Ther 2020; 19: 2175–2185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237. Shao DD, Xue W, Krall EB, et al. KRAS and YAP1 converge to regulate EMT and tumor survival. Cell 2014; 158: 171–184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238. Kurppa KJ, Liu Y, To C, et al. Treatment-induced tumor dormancy through YAP-Mediated transcriptional reprogramming of the apoptotic pathway. Cancer Cell 2020; 37: 104–122.e12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239. Kitajima S, Asahina H, Chen T, et al. Overcoming resistance to dual innate immune and MEK inhibition downstream of KRAS. Cancer Cell 2018; 34: 439–452.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240. Wang L, Zhang Z, Yu X, et al. Unbalanced YAP-SOX9 circuit drives stemness and malignant progression in esophageal squamous cell carcinoma. Oncogene 2019; 38: 2042–2055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241. Muramatsu T, Imoto I, Matsui T, et al. YAP is a candidate oncogene for esophageal squamous cell carcinoma. Carcinogenesis 2011; 32: 389–398. [DOI] [PubMed] [Google Scholar]
- 242. Liu L, Lu Z, Hu X, et al. Clinical significance of YAP1 and TAZ in esophageal squamous cell carcinoma. Medicine 2021; 100: e26597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243. Bailey DD, Zhang Y, van Soldt BJ. Use of hPSC-derived 3D organoids and mouse genetics to define the roles of YAP in the development of the esophagus. Development 2019; 146(23): dev178855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244. Zhao J, Li X, Yang Y, et al. Effect of YAP1 silencing on esophageal cancer. Onco Targets Ther 2016; 9: 3137–3146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245. Song S, Ajani JA, Honjo S, et al. Hippo coactivator YAP1 upregulates SOX9 and endows esophageal cancer cells with stem-like properties. Cancer Res 2014; 74: 4170–4182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246. Song S, Honjo S, Jin J, et al. The hippo coactivator YAP1 mediates EGFR overexpression and confers chemoresistance in esophageal cancer. Clin Cancer Res 2015; 21: 2580–2590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247. Song S, Xie M, Scott AW, et al. A novel YAP1 inhibitor targets CSC-enriched radiation-resistant cells and exerts strong antitumor activity in esophageal adenocarcinoma. Mol Cancer Ther 2018; 17: 443–454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248. Kaneda A, Seike T, Danjo T, et al. The novel potent TEAD inhibitor, K-975, inhibits YAP1/TAZ-TEAD protein-protein interactions and exerts an anti-tumor effect on malignant pleural mesothelioma. Am J Cancer Res 2020; 10: 4399–4415. [PMC free article] [PubMed] [Google Scholar]
- 249. Amidon B, Frosch H, Syed S, et al. Abstract P216: IK-930 mediated TEAD inhibition decreases and delays tumor growth and enhances targeted apoptosis in lung and colon cancer xenografts when combined with MEK or EGFR inhibitors. Mol Cancer Ther 2021; 20: 216–216. [Google Scholar]
- 250. Zanconato F, Battilana G, Forcato M, et al. Transcriptional addiction in cancer cells is mediated by YAP/TAZ through BRD4. Nat Med 2018; 24: 1599–1610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251. Wang W, Li N, Li X, et al. Tankyrase inhibitors target YAP by stabilizing Angiomotin family proteins. Cell Rep 2015; 13: 524–532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252. Macleod AR. Abstract ND11: The discovery and characterization of ION-537: A next generation antisense oligonucleotide inhibitor of YAP1 in preclinical cancer models. Cancer Res 2021; 81: ND11–ND11. [Google Scholar]
- 253. Xie Q, Chen J, Feng H, et al. YAP/TEAD-mediated transcription controls cellular senescence. Cancer Res 2013; 73: 3615–3624. [DOI] [PubMed] [Google Scholar]
- 254. Li F, Xu Y, Liu B, et al. YAP1-Mediated CDK6 activation confers radiation resistance in esophageal cancer – rationale for the combination of YAP1 and CDK4/6 inhibitors in esophageal cancer. Clin Cancer Res 2019; 25: 2264–2277. [DOI] [PubMed] [Google Scholar]
- 255. King RJ, Qiu F, Yu F, et al. Metabolic and immunological subtypes of esophageal cancer reveal potential therapeutic opportunities. Front Cell Dev Biol 2021; 9: 667852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256. Jiang C, Zhang L, Xu X, et al. Engineering a smart agent for enhanced immunotherapy effect by simultaneously blocking PD-L1 and CTLA-4. Adv Sci 2021; 8: e2102500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257. Anderson AC, Joller N, Kuchroo VK. Lag-3, tim-3, and TIGIT: Co-inhibitory receptors with specialized functions in immune regulation. Immunity 2016; 44: 989–1004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258. Koyama S, Akbay EA, Li YY, et al. Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints. Nat Commun 2016; 7: 10501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259. Rodriguez-Abreu D, Johnson ML, Hussein MA, et al. Primary analysis of a randomized, double-blind, phase II study of the anti-TIGIT antibody tiragolumab (tira) plus atezolizumab (atezo) versus placebo plus atezo as first-line (1L) treatment in patients with PD-L1-selected NSCLC (CITYSCAPE). J Clin Oncol 2020; 38: 9503–9503. [Google Scholar]
- 260. Huard B, Prigent P, Tournier M, et al. CD4/major histocompatibility complex class II interaction analyzed with cd4- and lymphocyte activation gene-3 (LAG-3)-Ig fusion proteins. Eur J Immunol 1995; 25: 2718–2721. [DOI] [PubMed] [Google Scholar]
- 261. Woo SR, Turnis ME, Goldberg MV, et al. Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape. Cancer Res 2012; 72: 917–927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262. Kang Y-K, Yamaguchi K, Oh DY, et al. An open-label, phase I trial of BI 754091 alone and in combination with BI 754111 in Asian patients (pts) with advanced solid tumors. J Clin Oncol 2020; 38: 3054–3054. [Google Scholar]
- 263. Zhu C, Anderson AC, Schubart A, et al. The tim-3 ligand galectin-9 negatively regulates T helper type 1 immunity. Nat Immunol 2005; 6: 1245–1252. [DOI] [PubMed] [Google Scholar]
- 264. Zhou G, Sprengers D, Boor PPC, et al. Antibodies against immune checkpoint molecules restore functions of tumor-infiltrating T cells in hepatocellular carcinomas. Gastroenterology 2017; 153: 1107–1119.e10. [DOI] [PubMed] [Google Scholar]
- 265. Ngiow SF, von Scheidt B, Akiba H, et al. Anti-TIM3 antibody promotes T cell IFN-γ-mediated antitumor immunity and suppresses established tumors. Cancer Res 2011; 71: 3540–3551. [DOI] [PubMed] [Google Scholar]
- 266. Vanneman M, Dranoff G. Combining immunotherapy and targeted therapies in cancer treatment. Nat Rev Cancer 2012; 12: 237–251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267. Schmidt EV, Chisamore MJ, Chaney MF, et al. Assessment of clinical activity of PD-1 checkpoint inhibitor combination therapies reported in clinical trials. JAMA Netw Open 2020; 3: e1920833. [DOI] [PubMed] [Google Scholar]
- 268. Bai LY, Chiu CF, Kadowaki S, et al. 1209P A phase II study of regorafenib in combination with nivolumab in patients with recurrent or metastatic solid tumors: results of the ESCC cohort. Ann Oncol 2022; 33: S1101–S1102. [Google Scholar]
- 269. Mo DC, Luo PH, Huang SX, et al. Safety and efficacy of pembrolizumab plus lenvatinib versus pembrolizumab and lenvatinib monotherapies in cancers: a systematic review. Int Immunopharmacol 2021; 91: 107281. [DOI] [PubMed] [Google Scholar]
- 270. Kumagai S, Koyama S, Nishikawa H. Antitumour immunity regulated by aberrant ERBB family signalling. Nat Rev Cancer 2021; 21: 181–197. [DOI] [PubMed] [Google Scholar]
- 271. Adachi Y, Kamiyama H, Ichikawa K, et al. Inhibition of FGFR reactivates IFNγ signaling in tumor cells to enhance the combined antitumor activity of lenvatinib with Anti-PD-1 antibodies. Cancer Res 2022; 82: 292–306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272. Kono M, Komatsuda H, Yamaki H, et al. Immunomodulation via FGFR inhibition augments FGFR1 targeting T-cell based antitumor immunotherapy for head and neck squamous cell carcinoma. OncoImmunology 2022; 11: 2021619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 273. Muro K, Kato K, Chin K, et al. 1241P phase ib study of futibatinib plus pembrolizumab in patients with advanced or metastatic solid tumors: tolerability results and antitumor activity in esophageal carcinoma. Ann Oncol 2022; 33: S1116–S580. [Google Scholar]
- 274. Feng GS, Hui CC, Pawson T. SH2-containing phosphotyrosine phosphatase as a target of protein-tyrosine kinases. Science 1993; 259: 1607–1611. [DOI] [PubMed] [Google Scholar]
- 275. Yuan X, Bu H, Zhou J, et al. Recent advances of SHP2 inhibitors in cancer therapy: Current Development and Clinical Application. J Med Chem 2020; 63: 11368–11396. [DOI] [PubMed] [Google Scholar]
- 276. Hui E, Cheung J, Zhu J, et al. T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science 2017; 355: 1428–1433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277. Li J, Jie HB, Lei Y, et al. PD-1/SHP-2 inhibits Tc1/Th1 phenotypic responses and the activation of T cells in the tumor microenvironment. Cancer Res 2015; 75: 508–518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278. Zhao M, Guo W, Wu Y, et al. SHP2 inhibition triggers anti-tumor immunity and synergizes with PD-1 blockade. Acta Pharm Sin B 2019; 9: 304–315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 279. Boyman O, Sprent J. The role of interleukin-2 during homeostasis and activation of the immune system. Nat Rev Immunol 2012; 12: 180–190. [DOI] [PubMed] [Google Scholar]
- 280. Abbas AK, Trotta E, R Simeonov D, et al. Revisiting IL-2: Biology and therapeutic prospects. Sci Immunol 2018; 3: eaat1482. [DOI] [PubMed] [Google Scholar]
- 281. Ptacin JL, Caffaro CE, Ma L, et al. An engineered IL-2 reprogrammed for anti-tumor therapy using a semi-synthetic organism. Nat Commun 2021; 12: 4785. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 282. Kim K, Skora AD, Li Z, et al. Eradication of metastatic mouse cancers resistant to immune checkpoint blockade by suppression of myeloid-derived cells. Proc Natl Acad Sci USA 2014; 111: 11774–11779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283. Shen L, Pili R. Class I histone deacetylase inhibition is a novel mechanism to target regulatory T cells in immunotherapy. OncoImmunology 2012; 1: 948–950. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 284. Orillion A, Hashimoto A, Damayanti N, et al. Entinostat neutralizes myeloid-derived suppressor cells and enhances the antitumor effect of PD-1 inhibition in murine models of lung and renal cell carcinoma. Clin Cancer Res 2017; 23: 5187–5201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 285. Maio M, Coral S, Fratta E, et al. Epigenetic targets for immune intervention in human malignancies. Oncogene 2003; 22: 6484–6488. [DOI] [PubMed] [Google Scholar]
- 286. Woods DM, Sodré AL, Villagra A, et al. HDAC inhibition upregulates PD-1 ligands in melanoma and augments immunotherapy with PD-1 blockade. Cancer Immunol Res 2015; 3: 1375–1385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 287. Gao Y, Nihira NT, Bu X, et al. Acetylation-dependent regulation of PD-L1 nuclear translocation dictates the efficacy of anti-PD-1 immunotherapy. Nat Cell Biol 2020; 22: 1064–1075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 288. Sullivan RJ, Moschos SJ, Johnson ML, et al. Abstract CT072: Efficacy and safety of entinostat (ENT) and pembrolizumab (PEMBRO) in patients with melanoma previously treated with anti-PD1 therapy. Cancer Res 2019; 79: CT072–CT072. [Google Scholar]
- 289. Gray JE, Saltos A, Tanvetyanon T, et al. Phase I/Ib study of Pembrolizumab Plus vorinostat in Advanced/metastatic Non-Small cell lung cancer. Clin Cancer Res 2019; 25: 6623–6632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 290. Rodriguez CP, Wu QV, Voutsinas J, et al. A Phase II trial of Pembrolizumab and vorinostat in recurrent metastatic head and neck squamous cell carcinomas and salivary gland cancer. Clin Cancer Res 2020; 26: 837–845. [DOI] [PubMed] [Google Scholar]
- 291. Lordick F, Mariette C, Haustermans K, et al. Oesophageal cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2016; 27: v50–v57. [DOI] [PubMed] [Google Scholar]
- 292. Gardner-Thorpe J, Hardwick RH, Dwerryhouse SJ. Salvage oesophagectomy after local failure of definitive chemoradiotherapy. Br J Surg 2007; 94: 1059–1066. [DOI] [PubMed] [Google Scholar]
- 293. Bedenne L, Michel P, Bouché O, et al. Chemoradiation followed by surgery compared with chemoradiation alone in squamous cancer of the esophagus: FFCD 9102. J Clin Oncol 2007; 25: 1160–1168. [DOI] [PubMed] [Google Scholar]
- 294. Stahl M, Stuschke M, Lehmann N, et al. Chemoradiation with and without surgery in patients with locally advanced squamous cell carcinoma of the esophagus. J Clin Oncol 2005; 23: 2310–2317. [DOI] [PubMed] [Google Scholar]
- 295. Crosby T, Hurt CN, Falk S, et al. Chemoradiotherapy with or without cetuximab in patients with oesophageal cancer (SCOPE1): a multicentre, phase 2/3 randomised trial. Lancet Oncol 2013; 14: 627–637. [DOI] [PubMed] [Google Scholar]
- 296. Suntharalingam M, Winter K, Ilson D, et al. Effect of the addition of cetuximab to paclitaxel, cisplatin, and radiation therapy for patients with esophageal cancer: the NRG oncology RTOG 0436 Phase 3 randomized clinical trial. JAMA Oncol 2017; 3: 1520–1528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 297. Wu SX, Wang LH, Luo HL, et al. Randomised phase III trial of concurrent chemoradiotherapy with extended nodal irradiation and erlotinib in patients with inoperable oesophageal squamous cell cancer. Eur J Cancer 2018; 93: 99–107. [DOI] [PubMed] [Google Scholar]
- 298. Xie C, Jing Z, Luo H, et al. Chemoradiotherapy with extended nodal irradiation and/or erlotinib in locally advanced oesophageal squamous cell cancer: long-term update of a randomised phase 3 trial. Br J Cancer 2020; 123: 1616–1624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 299. Meng X, Zheng A, Wang J, et al. Nimotuzumab plus concurrent chemo-radiotherapy versus chemo-radiotherapy in unresectable locally advanced esophageal squamous cell carcinoma (ESCC): interim analysis from a prospective, randomized-controlled, double-blinded, multicenter, and phase III clinical trial (NXCEL1311 study). J Clin Oncol 2022; 40: 4016–4016. [Google Scholar]
- 300. Krysko DV, Garg AD, Kaczmarek A, et al. Immunogenic cell death and DAMPs in cancer therapy. Nat Rev Cancer 2012; 12: 860–875. [DOI] [PubMed] [Google Scholar]
- 301. Twyman-Saint Victor C, Rech AJ, Maity A, et al. Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature 2015; 520: 373–377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 302. Klug F, Prakash H, Huber P, et al. Low-dose irradiation programs macrophage differentiation to an iNOS+/M1 phenotype that orchestrates effective T cell immunotherapy. Cancer Cell 2013; 24: 589–602. [DOI] [PubMed] [Google Scholar]
- 303. Antonia SJ, Villegas A, Daniel D. Durvalumab after chemoradiotherapy in Stage III Non-Small-Cell lung cancer. N Engl J Med 2017; 377: 1919–1929. [DOI] [PubMed] [Google Scholar]
- 304. Bando H, Kumagai S, Kotani D, et al. 1211P A multicenter phase II study of atezolizumab monotherapy following definitive chemoradiotherapy for unresectable locally advanced esophageal squamous cell carcinoma (EPOC1802). Ann Oncol 2022; 33: S1102–S1103. [Google Scholar]
- 305. Wang J, Cheng Y, Wu Y, et al. 1262TiP efficacy and safety of consolidative camrelizumab following definitive concurrent chemoradiotherapy in patients with locally advanced esophageal squamous cell cancer. Ann Oncol 2022; 33: S1124–S580. [Google Scholar]