Open Access

Challenges and perspectives for immunotherapy in oesophageal cancer: A look to the future (Review)

  • Authors:
    • Andrea Lampis
    • Margherita Ratti
    • Michele Ghidini
    • Milko B. Mirchev
    • Ali Fuat Okuducu
    • Nicola Valeri
    • Jens Claus Hahne
  • View Affiliations

  • Published online on: April 8, 2021     https://doi.org/10.3892/ijmm.2021.4930
  • Article Number: 97
  • Copyright : © Lampis et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY 4.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Oesophageal cancer is one of the most aggressive malignancies with limited treatment options, thus resulting in a high morbidity and mortality. With 5‑year survival rates of only 5‑10%, oesophageal cancer holds a dismal prognosis for patients. In order to improve overall survival, the early diagnosis and tools for patient stratification for personalized treatment are urgent needs. A minority of oesophageal cancers belong to the spectrum of Lynch syndrome‑associated cancers and are characterized by microsatellite instability (MSI). Microsatellite instability is a consequence of defective mismatch repair protein functions and it has been well characterized in other gastrointestinal tumours, such as colorectal and gastric cancer. In the latter, high levels of MSI are associated with a better prognosis and with an increased benefit to immune‑based therapies. Therefore, similar therapeutic approaches could offer an opportunity of treatment for oesophageal cancer patients with MSI. Apart from immune checkpoint inhibitors, other immunotherapies such as adoptive T‑cell transfer, peptide vaccine and oncolytic viruses are under investigation in oesophageal cancer patients. In the present review, the rationale and current knowledge about immunotherapies in oesophageal cancer are summarised.

1. Introduction

The incidence of oesophageal cancer has rapidly increased over the past years and it is currently the fifth most common type of cancer worldwide with a very high mortality rate (1,2). Oesophageal cancer is subdivided into two groups according to its histological appearance: Oesophageal squamous cell carcinoma (predominant in western countries) and oesophageal adenocarcinoma (most common form in Asia) (3,4). Thus far, no molecular markers for prognosis or treatment efficacy have been discovered for squamous cell carcinoma. For oesophageal adenocarcinoma, the human epidermal growth factor receptor-2 (HER-2) status has been proven to be an efficient biomarker. HER-2 is scored by immunohistochemistry for protein expression or fluorescent in situ hybridization for HER2 gene amplification (5). If positive, a targeted therapy option with trastuzumab for HER-2 is the treatment of choice (6,7). Nevertheless, oesophageal cancer is mostly treated by radiation in combination with chemotherapy or surgery. However, the 5-year overall survival rate remains very poor and is only at 5-10% (8,9). Nonetheless, surgery is not applicable in approximately half of patients as distant metastases are already present at the time of diagnosis (10). The most commonly used chemotherapeutic agents for oesophageal cancer treatment are 5′-fluorouracil and platinum agents in combination with radiotherapy (11,12).

The major risk factors for oesophageal cancer are represented by smoking, the consumption of hot tea, red meat consumption, poor oral health, low intake of fresh fruits and vegetables, alcohol abuse, obesity, nass use (a chewing tobacco product), opium consumption and low socioeconomic status (13-24). The majority of these risk factors induce gene mutations which can be recognized by the immune system (25). In addition, a minority of oesophageal cancers belong to the spectrum of Lynch syndrome-associated cancers and are characterized by microsatellite instability (MSI) (26). Therefore, the use of immunotherapy approaches in oesophageal cancer appears to be an attractive novel therapeutic strategy.

The identified main reasons for the high mortality rate of patients with oesophageal cancer are mainly the late stage of diagnosis (13) and the key role of tumour microenvironment in this type of cancer (27), where the surrounding stromal cells seem to exert an important influence in supporting tumour cell survival (27). Apart from cancer-associated fibroblasts, that are able to support tumour growth and metastasis by altering the extracellular matrix by secreting growth factors and cytokines, several immune cells [e.g., myeloid-derived suppressor cells, tumour-associated macrophages and regulatory T-cells (TREGS)] are involved in support the development of oesophageal cancer (27). Therapies targeting the tumour microenvironment and/or the immune system may thus be able to increase the survival of patients with oesophageal cancer. Over the past years, immunotherapy in particular has revolutionized the management and outcome of several types of cancer, such as melanoma, lung, gastric and kidney cancer (28). Therefore, it may be advantageous to explore the benefits from immunotherapy for oesophageal cancer. The identification and selection of robust biomarkers predicting clinical benefit are also mandatory before commencing immunotherapy treatment, as even though generally well-tolerated compared to standard therapies, immunotherapy is associated occasionally with severe toxic side-effects, such as cutaneous, gastrointestinal, endocrine and hepatic toxicity. Thus, only patients with oesophageal cancer who have the highest likelihood of benefit from immunotherapy should be offered this therapeutic regimen. For example, it is well-established that immune checkpoint inhibitors are particularly effective against mismatch repair-deficient tumours (29). In general, tumours with MSI have a higher mutation rate, which increases the probability for the immune system to recognize tumour cells (29-31). Recently, several reviews have summarized the current knowledge on immunotherapy and cancer (32-34). The present review focuses on the current state of the use of immunotherapy in oesophageal cancer.

2. Biological background of tumour immunotherapy

The immune system is a highly complex and specialised biological network including specific cells, protein and organs and is usually composed of two types: Adaptive (specific) and innate (non-specific) (35). In recognising and preventing the spread of cancer cells, the innate immunity components, such as natural killer (NK) cells, dendritic cells and macrophages are of pivotal importance; nevertheless, T-cells from the adaptive immune are recruited in order to track and kill tumour cells (35,36). Recently, a new model that provides a mechanistic explanation of this interaction termed 'cancer-immunity cycle' has been suggested (35). According to this model, dead cancer cells release antigens that in turn are recognised by antigen-presenting cells (particularly by dendritic cells). This results in the priming and activation of dendritic cells and T-cells in lymph nodes, followed by the recruitment of helper T-cells [cluster of differentiation (CD)4+-T-cells] and cytotoxic T-lymphocytes (CD8+-T-cells) at the tumour site. Following the infiltration of the tumour microenvironment, immune cells recognize and attack tumour cells that results in the release of further tumour antigens. The whole cancer-immunity cycle is fine-tuned by different stimulating and inhibitory factors, such as chemokines, cytokines, metabolic compounds, surface proteins and immune checkpoint receptors to prevent autoimmunity (37).

Cancer cells use different strategies to escape the immune system, and to capture and reprogram immune cells, leading to immune evasion. Among these strategies is the mechanisms of shedding of MHC class I chain-related protein A and B (MICA and MICB) from tumour cells into the tumour microenvironment as protection against NK cell-mediated killing (38-40). In addition, tumour cells express immune checkpoint proteins, such as programmed cell death 1 ligand 1 (PD-L1) and receptors, such as cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) on the surface, but also secrete exosomes which contains these immune checkpoint regulators. After binding to proteins expressed on the immune cells (T-cells, B-cells and myeloid cells) the checkpoint regulators exert an inhibitory signal and lead to the suppression of the immune response (41-43).

Furthermore, cancer cells, as well as tumour-associated macrophages are able to secrete chemokines, such as chemokine (C-C motif) ligand (CCL)-17 and CCl-22 which attract a subpopulation of T-cells, the so-called TREGs. TREGs are known to regulate and suppress the activity of other immune cells and to help preventing autoimmune reactions under healthy conditions (44,45). In tumour tissue, TREGs protect cancer cells and foster tumour growth (46,47). Moreover, CD8+-T-cells are inhibited directly by myeloid-derived suppressor cells (MDSCs) which are stimulated by tumour-derived growth factors (48,49). In addition, stromal cells in the tumour microenvironment inhibit the function of the immune system further supporting tumour progression and metastasis (50).

3. Immunotherapy in oesophageal cancer

The immune system is a complex network of interacting cells and biochemical signals that orchestrate the recognition and attack of external antigens, whilst preventing autoimmune reactions. Under physiological conditions, this is guaranteed by a fine-tuned interplay between immune cells and a balance between stimulatory and inhibitory signals (51,52). Cancer cells often find a way to de-regulate the balanced immune system by manipulating signalling pathways to evade from immune surveillance. To overcome the mechanisms of tumour immune evasion and use the immune system as weapon against cancer, either agonists of stimulatory receptors or antagonists of inhibitory signals can be used (41). Nevertheless, according to currently available study results, only a subset of oesophageal cancer patients may benefit from immunotherapy (53). Therefore, there is an urgent need to identify biomarkers for the prediction of the benefit from immunotherapy, so that patients can be selected for treatment and those who have no benefit from immunotherapy are spared from side-effects (e.g., cutaneous, gastrointestinal, endocrine and hepatic toxicity) and therapy failure. In light of this scenario, currently, several clinical trials are underway to evaluate the efficacy of different immunotherapies combined with other treatment options in oesophageal cancer patients (Table I) with the aim to increase the therapeutic option for oesophageal cancer patients. The majority of these studies are ongoing Phase 2 studies and the results have not been published yet.

Table I

Clinical trials investigating immunotherapeutic options in combination with other therapeutic options in oesophageal cancer patients.

Table I

Clinical trials investigating immunotherapeutic options in combination with other therapeutic options in oesophageal cancer patients.

Immunotherapeutic optionStage of developmentCombined withClinicalTrials.gov. identifierStart timeCurrent status
Immune checkpoint inhibitor treatment
NivolumabPhase 1/2 ChemoradiotherapyNCT032786262017Active, but not recruiting
Phase 1/2 ChemoradiotherapyNCT035447362018Recruiting
Phase 2Chemoradiotherapy + IpilimumabNCT036049912018Recruiting
Phase 2Chemoradiotherapy + IpilimumabNCT034372002018Recruiting
Phase 3Ipilimumab + ChemotherapyNCT31431532017Recruiting
Phase 2IpilimumabNCT034162442018Recruiting
Phase 1/2Ipilimumab + INCAGN01949NCT032411732017Completed in 2020, but no results published
Phase 1Chemoradiotherapy + RelatlimabNCT030446132017Recruiting
Phase 2Chemoradiotherapy + CetuximabNCT042294592020Recruiting
Phase 1ChemotherapyNCT039144432019Recruiting
Phase 3After chemotherapyNCT025692422015Active, but not recruiting
Phase 3After chemoradiotherapy and surgeryNCT027434942016Active but not recruiting
Phase 1+ RucaparibNCT039950172019Recruiting
Phase 1+ MogamulizumabNCT029466712016Completed in 2020, but no results published
Phase 1+ MogamulizumabNCT024761232015Completed in 2020, but no results published
Phase 1/2RadiotherapyNCT035447362018Recruiting
PembrolizumabPhase 2 ChemoradiotherapyNCT028440752016Active, but not recruiting
Phase 2 ChemoradiotherapyNCT030644902017Recruiting
Phase 2 ChemoradiotherapyNCT033222672017Recruiting
Phase 1 ChemoradiotherapyNCT037923472019Active, but not recruiting
Phase 2Chemoradiotherapy + chemotherapyNCT029982682016Active, but not recruiting
Phase 2After chemoradiotherapy and surgeryNCT028440752016Active but not recruiting
Phase 2After chemoradiotherapy and surgeryNCT033222672017Recruiting
Phase 2Chemotherapy + TrastuzumabNCT029545362016Recruiting
Phase 2+ EpacadostatNCT035924072018Withdrawn due to safety concerns
Phase 3ChemotherapyNCT031897192017Active, but not recruiting
Phase 3ChemotherapyNCT038811112019Withdrawn due to protocol amendment
Phase 2ChemotherapyNCT044372122020Recruiting
Phase 2RadiotherapyNCT028305942016Active, but not recruiting
Phase 1Brachytherapy: 16Gy/2FNCT026428092015Active, but not recruiting
Phase 1DKN-01NCT020131542013Active, but not recruiting
Phase 1/2INCAGN01876 + EpacadostatNCT032773522017Completed in 2020, but no results published
Phase 2TadalafilNCT039933532019Recruiting
Phase 2+ CRS-207NCT031225482019Terminated because of low enrolment and lack of clinical activity in other CRS-207 studies
CamrelizumabPhase 2RadiotherapyNCT032006912017Recruiting
Phase 2RadiotherapyNCT031873142017Recruiting
Phase 2ChemotherapyNCT039179662019Not yet recruiting
Phase 3ChemotherapyNCT036910902018Recruiting
Phase 2 ChemoradiotherapyNCT043909452020Recruiting
Phase 3 ChemoradiotherapyNCT044269552020Recruiting
Phase 3 ChemoradiotherapyNCT044044912020Recruiting
Phase 2After chemoradiotherapyNCT038176582019Not yet recruiting
Phase 1After chemoradiotherapyNCT039850462019Recruiting
Phase 2After chemoradiotherapyNCT042869582020Recruiting
Phase 2ApatinibNCT037368632019Not yet recruiting
Phase 2Apatinib + chemotherapyNCT036037562018Recruiting
Phase 2NimotuzumabNCT037661782018Recruiting
SintilimabPhase 1/2ChemotherapyNCT039469692019Recruiting
Phase 3ChemotherapyNCT037481342018Recruiting
Phase 1 ChemoradiotherapyNCT039400012019Recruiting
SpartalizumabPhase 1LGK974NCT013511032011Recruiting
Phase 1/2LAG525NCT024602242015Active, but not recruiting
Phase 2LAG525NCT033657912017Active, but not recruiting
Phase 2MCS110NCT037854962018Active, but not recruiting
Phase 1TNO155NCT040005292019Recruiting
TislelizumabPhase 2ChemotherapyNCT034695572018Active, but not recruiting
Phase 3ChemotherapyNCT037834422018Recruiting
Phase 3 ChemoradiotherapyNCT039575902019Recruiting
ToripalimabPhase 2ChemotherapyNCT039856702019Recruiting
Phase 3ChemotherapyNCT038299692019Recruiting
Phase 2 ChemoradiotherapyNCT040060412019Recruiting
Phase 2 ChemoradiotherapyNCT040051702019Recruiting
Phase 2 ChemoradiotherapyNCT040841582019Recruiting
Phase 2 ChemoradiotherapyNCT041778752019Recruiting
Phase 2After Chemoradiotherapy + surgeryNCT044372122020Recruiting
HLX-10Phase 3ChemotherapyNCT039588902019Recruiting
AvelumabPhase 1/2 ChemoradiotherapyNCT034902922018Recruiting
Phase 2 ChemoradiotherapyNCT038009532019Not yet recruiting
Phase 2Chemotherapy before surgeryNCT033990712018Recruiting
AtezolizumabPhase 2 ChemoradiotherapyNCT030878642017Completed in 2020, but no results published
Phase 1 ChemoradiotherapyNCT037843262018Recruiting
Phase 2ChemotherapyNCT034488352018Recruiting
Phase 1/2CabozantinibNCT031709602017Recruiting
Phase 1/2KY1044NCT038295012019Recruiting
Phase 1/2DKN-01NCT041667212019Recruiting
DurvalumabPhase 2 ChemoradiotherapyNCT029620632016Recruiting
Phase 2 ChemoradiotherapyNCT037778132018Recruiting
Phase 2Chemoradiotherapy + chemotherapyNCT027352392016Active but not recruiting
Phase 2After chemoradiotherapyNCT040545182019Not yet recruiting
Phase 2After chemoradiotherapy + surgeryNCT026390652015Active but not recruiting
Phase 2After chemoradiotherapy + surgeryNCT025204532015Active but not recruiting
Phase 2Chemoradiotherapy + TremelimumabNCT033774002017Active but not recruiting
Phase 1Chemotherapy + TremelimumabNCT026582142013Active but not recruiting
Phase 2TremelimumabNCT032922502017Recruiting
Phase 2TremelimumabNCT039821732019Not yet recruiting
Phase 2TremelimumabNCT041599742019Recruiting
Phase 1/2Tremelimumab + SBRTNCT032124692017Recruiting
SHR-1316Phase 2ChemotherapyNCT037325082018Recruiting
Phase 2NimotuzumabNCT037661782019Not yet recruiting

Adoptive T-cell therapy
Phase 1HER2Bi-armed T-cells + IL-2NCT026623482016Unknown
Phase 1/2CAR-T combined with PD-1 knockout T-cellsNCT037063262018Recruiting
Phase 1CAR-T combined with CAdVEC (oncolytic adenovirus)NCT037402562018Recruiting
Phase 1TCR-T + Cyclophosphamide + FludarabineNCT028692172016Recruiting
Phase 1TCR-T + + Fludarabine CyclophosphamideNCT023665462015Active but not recruiting
Phase 1TCR-T + + Fludarabine CyclophosphamideNCT020966142017Unknown
Phase 1TCR-T + RadiotherapyNCT031329222017Recruiting
Phase 1TCR-T + TrastuzumabNCT036805602018Suspended by the sponsor

Peptide vaccine
Phase 1+ ChemotherapyNCT006323332011Unknown
Phase 2+ Toll-like receptor 9 agonistNCT006692922010Unknown
Phase 2+ Granulocyte-macrophage colony stimulating factorNCT000122462013Terminated without any published results

Oncolytic virus
Oncolytic measles virusPhase 1+ 5-Fluorocytosine + anti-PD-1 checkpoint inhibitorNCT041953732020Withdrawn
Oncolytic adenovirusPhase 1CAdVEC combined with CAR-TNCT037402562018Recruiting
Phase 1Telomelysin + radiotherapyNCT032130542017Recruiting
Phase 2Telomelysin + PembrolizumabNCT039210212019Recruiting

[i] ClinicalTrials.gov was accessed in November, 2020.

In the following section, the main immunotherapy approaches that have been studied thus far will be discussed (Fig. 1).

4. Immune checkpoint inhibitors

Immune checkpoints are of pivotal importance to prevent autoimmunity reactions by the inhibition of antigen recognition via T-cell receptors (TCRs) (41,54,55). Cancer cells use immune checkpoint proteins to inactivate the adaptive immune system by blocking tumour specific T-cells and escape from immune surveillance. Thus far, the immune checkpoint receptors programmed cell death protein 1 (PD-1; also known as CD279) and CTLA-4 (also known as CD152) have been found to be associated with the inhibition and downregulation of T-cell activity (41,54,55).

PD-1 receptor is highly expressed on T-cells, B-cells and NK cells. The ligand for PD-1 receptor is PD-L1 often also termed B7-homolog 1 (B7-H1) or CD274. This molecule is expressed in peripheral tissues following exposure to inflammatory cytokines and limits T-cell activity (56). Furthermore, interleukin (IL)-18, an inflammatory cytokine that accumulates in the tumour microenvironment, results in the upregulation of PD-L1 in activated mature NK cells and triggers immunosuppression (57). In melanoma, lung, breast, pancreatic, gastric, colon, ovarian and oesophageal cancers, PD-L1 is often found overexpressed on cancer cells (58). This enables tumour cells to interact with PD-1 receptors on T-cells and this interaction prevents T-cell activation, proliferation and ultimately leading to T-cell apoptosis (41).

The expression of CTLA-4 receptor is restricted to activated T-cells (e.g., TREGs), whereas the homolog CD28 is also expressed on non-activated T-cells. Ligands for both receptors are the immunoglobulin proteins B7-1 (CD80) and B7-2 (CD86), which are expressed early during the immune response on antigen-presenting cells, such as macrophages and dendritic cells or on B-cells and monocytes, respectively. CTLA-4 receptor has a higher affinity for ligands and competing with CD28 on ligand binding; the interaction between B7-1 or B7-2 with CD28 results in T-cell activation, whereas the interaction with CTLA-4 inhibits T-cell activation at an early stage (59,60).

It has been widely proven that PD-L1 expression is one of the key mechanisms through which several cancers evade the immune response; thus, it is not surprising that inhibitors of PD-L1 and PD-1 have been identified thus far as one of most efficient and broadly used immunotherapies for cancer (61-71). Recently, a monoclonal antibody targeting PD-1, pembrolizumab, has been approved for the treatment of oesophageal and oesophagogastric junction adenocarcinoma by the US Food and Drug Administration (FDA) (8). The prerequisite for the treatment of oesophageal cancer with pembrolizumab is either a proven PD-L1 expression on the cancer cells and a high MSI, or a proven defective mismatch repair system. Therefore, most probably, the subgroup of Lynch syndrome-associated oesophageal cancers patients may benefit from this new treatment option. According to a previous study, it is possible to predict the efficacy of pembrolizumab in patients with oesophageal cancer by using a six-gene interferon-γ gene expression signature (72). This offers the possibility to stratify oesophageal cancer patients and limit the targeted treatment to the group that will most probably benefit from the anti-PD-L1 therapy.

Earlier in 2020, the FDA approved nivolumab, a fully human monoclonal antibody against PD-1 (73) for patients with unresectable advanced, recurrent or metastatic oesophageal squamous cell carcinoma as a second line following 5′-fluorouracil- and platinum-based chemotherapy. The overall survival benefit is 2.5 months according to a phase 3 clinical study (74).

Currently, combination therapies with anti-PD1 and anti-CTLA-4 antibodies are forthcoming (75). According to the first preliminary results from clinical studies (NCT02743494, CheckMate 648 and CheckMate 649) the combination of nivolumab with the anti CDLA-4 antibody, ipilimumab, led to an improved clinical response in oesophageal cancer compared to treatment with nivolumab alone (76,77). The combination of nivolumab and ipilimumab appears to be safe; nevertheless, it must be considered that CTLA-4 blockade results in more severe and more common side-effects than it is the case for targeting PD-1/PD-L1 alone. Therefore, the development of novel strategies for reducing serious adverse side-effects is an urgent need and the first steps need to be carefully controlled (78).

As a potential biomarker for prediction of the response to immune checkpoint inhibitor therapy, the total amount of PD-1+ CD4+ T-cells in the tumour microenvironment is discussed. According to the presence or absence of CD4+ T-cells and PD-1 expression in the tumour microenvironment, a stratification of patients is possible. The absence of CD4+ T-cells and PD-1 expression results in immunological ignorance; in a situation where only one component (either CD4+ T-cells or PD-1) is expressed, immunological tolerance exists and only in the case of a PD-1+ tumour microenvironment containing CD4+ T-cells an adoptive immune resistance is present that is most likely to respond to immune checkpoint inhibitor therapy (79).

5. Adoptive T-cell therapy

Adoptive T-cell therapy is a personalized approach of immunotherapy. T-cells are collected from the tumour or peripheral blood of a patient and the isolated T-cells are stimulated in vitro with IL-2. After this ex-vivo expansion, the cancer patient receives his own autologous immune cells as an infusion (80). In addition, T-cells can be also genetically modified after collection from the patient either by introducing chimeric antigen receptor (CAR T-cells) or transducing antigen-specific TCR cells (TCR T-cells). In all cases, the expanded or modified T-cells exert an improved tumour-specific immunity (81-83). In several trials, a regression of tumours has been demonstrated following persistent adoptive T-cell therapy (84,85). In a first clinical trial based on adoptive T-cell therapy for patients with recurrent or advanced oesophageal cancer, the patients received (on a fortnight basis) activated T-cells administered into primary tumours or metastatic lymph nodes; this therapy was found to be safe and in one third of the patients, a significant tumour regression was observed (86). In another study, based on TCR T-cells, oesophageal cancer patients with minimal tumours survived >27 months; nevertheless, after 2 months of treatment, several patients exhibited tumour progression even if the autologous T-cells persist for a long period of time; therefore, TCR T-cell therapy appears to have a benefit only for oesophageal cancer patients with minimal lesions (87).

6. Peptide vaccine

Peptide vaccines are therapeutic cancer vaccines which aim to increase immunogenic cancer-specific antigens, leading to the activation of cancer antigen-specific T-cells in vivo (59,76,88). For the successful use of peptide vaccines, the characterization of tumour-specific T-cells and the use of immunogenic tumour-associated antigens are a prerequisite (89). As tumour-associated antigens, either recombinant short peptides, whole-cell tumour lysates or full-length proteins can be used (90,91). The length of the used peptide has at least in part an influence on the efficiency of the immune response (92). It has been well-established that short peptides composed of 8-11 amino acids induce major histocompatibility complex (MHC) class-I-restricted antigen-specific CD8+ T-cell reaction via direct binding to human leukocyte antigen (HLA)-I molecules (93). By contrast, longer peptides (25-50 amino acids) are usually presented by MHC class-I and class-II molecules on antigen-presenting cells to CD8+ or CD4+ T-cell, respectively (94). This results in a broader and longer lasting immune response by generating cytotoxic T-lymphocytes as well as long-living memory CD8+ T-cells (95).

In a modified approach, dendritic cells isolated from the peripheral blood of a cancer patient are presented to tumour-associated antigens ex vivo and after loading with the antigens the dendritic cells, are re-injected into patients (91,96). This strategy was evaluated in a pre-clinical study as possible novel treatment option for oesophageal tumours (97). Dendritic cells from oesophageal cancer patients have been pulsed with Wilms' tumour 1 peptide ex vivo and used as a vaccine. The patients were treated in parallel with the chemotherapeutic agent, picibanil. In this exploratory study, 15 patients were included; the median progression-free survival and overall survival were 4.1 and 7.0 months, respectively. This treatment was well-tolerated and no severe adverse events related to the vaccinations were observed (97). Based on this promising result, a phase II clinical trial is in preparation.

Even with the first-generation of peptide vaccines which have been based on highly expressed non-mutant tumour-associated antigens of tumour cells [such as melanoma antigen gene (MAGE) and New York oesophageal squamous cell carcinoma-1 (NY-ESO-1) proteins] an immune response was induced and led to clinical positive effect (98-100). The advantage of these peptides is that they are only expressed in male germ-line cells and placenta under physiological conditions; however, a number of tumours, among these oesophageal cancer, express these proteins as well. Therefore, they represent very promising targets for cancer immunotherapy (101-103).

The second-generation of peptide vaccines is an effort for a more personalized medicine with the aim of targeting mutated antigens that are patient-specific. In this approach, mutations which have been accumulated during tumour development are the basis for the vaccine generation (104). In the context of oesophageal cancer, a large number of genetic mutations are present which result in specific neo-antigens (105). The main challenge is to identify mutated epitopes derived from tumour neo-antigens for developing a patient-specific vaccine (106,107). The vaccine peptides are patient-specific and they differ completely among patients. Therefore, batch production will not be possible and it will never become a conventional drug (104). The advantage is that neo-antigen vaccines result in a potent T-cell response and induce a new population of specific T-cells in cancer patients that are able to kill cancer cells without damaging healthy tissues (104,108,109). Furthermore, pre-clinical trials are forthcoming with an aim to induce the T-cell response by ribonucleic acid (RNA)-based vaccine coding for multiple neo-epitopes (110). Another novel strategy combines the use of long-peptide vaccines with checkpoint inhibitor administration (111). The aim in both cases, is to increase the repertoire of CD8+ and CD4+ T-cell directed against the tumour. These personalized approaches have the potential to offer novel therapeutic options with high specificity and low toxicity for cancer patients who are resistant to current therapies.

Peptide vaccines have been used in several clinical trials in patients with oesophageal squamous carcinoma. Different peptides have been administered simultaneously to patients, which resulted in a significant induced CD8+ T-cell response. Clinical benefit, as well as an increased overall survival was observed in the majority of patients (112,113). Peptide vaccinations can be combined with other therapeutic options in patients with oesophageal tumours. One example is the use of a peptide vaccine to suppress the recurrence of oesophageal cancer following curative resection. In a previous study, the 5-year relapse-free survival of oesophageal cancer patients was 44.6% in patients that received the vaccination compared to the ones that did not receive the vaccination (31.6% relapse-free survival) (114). Of special interest is the peptide vaccine, S-588410, which is composed of 5 HLA-A*2402-restricted epitope peptides derived from the onco-antigens, DEPDC1, MPHOSPH1, URLC10, CDCA1 and KOC1. All these antigens are up-regulated in the context of oesophageal cancer (115,116). In previous studies, it was proven that each of these 5 peptides has the capacity to induce a peptide-specific activation of CD8+ T-cells in different tumours, among these oesophageal cancer (112,113,117,118). In an exploratory study based on 15 patients with oesophageal tumours, an increased immune response in tumour tissue was observed following vaccination with S-588410. Following a median of 5 injections of S-588410, peptide-specific CD8+ T-cells for all peptides included in this vaccination were induced in all patients. The number of functional T-lymphocytes (CD8+ and CD4+ T-cells) was found to be increased in blood, as well as in tumour biopsies. In parallel, a higher PD-L1 expression in the tumour microenvironment was observed (115). Most probably, the increased PD-L1 expression was related to interferon (IFN)-γ produced by infiltrated CD8+ T-cells into the tumour area. The accumulation of effective T-cells and IFN-γ production in the tumour microenvironment most probably favour the change from an immune 'desert' into an immune-inflamed tumour microenvironment (93). It is tempting to speculate about the therapeutic potential of combining peptide vaccines, such as S-588410 with immune-checkpoint inhibitors in patients with oesophageal cancer (54,79).

7. Oncolytic viruses

Oncolytic virus therapy is still in its infancy, but it has already proven its potential. In general, oncolytic viruses infect and replicate selectively in tumour cells and induce tumour cell lysis (119,120). Talimogene laherparepvec is the first FDA-approved oncolytic viral therapy for the treatment of patients with advanced melanoma (121). Recently, the efficacy of a telomerase-specific oncolytic virus (telomelysin OBP-301) in combination with radiotherapy was investigated in a Phase I/II study for the treatment of elderly patients with oesophageal squamous cell carcinoma. According to the first results, this viral therapy was well-tolerated and demonstrated efficient tumour regression (122,123). Based on this success, several other clinical trials with various oncolytic viruses for the treatment of patients with oesophageal cancer are ongoing (Table I).

8. Conclusion and perspectives

In oesophageal cancer, as in most other tumour diseases, the therapeutic options are limited and therapeutic success is only achieved for a short period of time before resistance appears. Therefore, novel therapeutic options, such as the addition of immunotherapy to the treatment of tumours are an urgent need. Albeit some success of immunotherapy in oesophageal cancer treatment and the approval of pembrolizumab and nivolumab by the FDA, it is noteworthy to mention that immunotherapy is often associated with severe toxic side-effects; the most frequent ones are cutaneous, gastrointestinal, endocrine and hepatic toxicity. Therefore, a careful monitoring and follow-up of patients under immunotherapy is required and if necessary, the patient must receive effective measures to manage the side-effects. An advantage for patients with oesophageal cancer could be a combination of immunotherapy with surgery, chemotherapy and radiotherapy. Recently, the advantage from radiotherapy in parallel with immune checkpoint inhibitor treatment was already demonstrated (124).

A prerequisite for improving the success and efficiency of immunotherapy is the knowledge about robust biomarkers predicting clinical benefit before treatment and enabling stratification of oesophageal cancer patients in such a manner that the best possible immunotherapy can be applied to each patient. One possibility could be the multiplexed immunohistochemical staining of adaptive immune (CD3, CD4, CD8 and CD45RO) and immune checkpoint biomarkers [inducible T-cell costimulatory molecule (ICOS), indoleamine-2,3-di- oxygenase-1 (IDO-1), PD-L1 and PD-1] in combination with digital pathology quantitation (125). Furthermore, it is well-established that immunotherapies are resulting in an increased tumour burden and/or emergence of new tumour lesions in the short-term. Therefore, the currently used evaluation system for therapeutically success is most probably not applicable for immunotherapies; thus, it may be prudent to consider a different system for this novel type of therapy.

Availability of data and materials

Not applicable.

Authors' contributions

JCH, AL and NV were involved in the conceptualization of the study. JCH, MG, MR and AL were involved in the writing and preparation of the original draft. JCH, NV, MBM and AFO were involved in the writing, reviewing and editing of the manuscript. All authors have read and agreed to the published version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

NV received speaker honorarium from the companies, Bayer, Eli-Lilly, Pfizer and Merck. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results. All other authors (AL, MR, MG, MBM, AFO and JCH) declare that they have no competing interests.

Acknowledgments

Not applicable.

Funding

No funding was received.

Abbreviations:

B7-H1

B7-homolg1;

CAR

chimeric antigen receptor;

CCL

chemokine (C-C motif) ligand;

CD

cluster of differentiation;

CTLA-4

cytotoxic T lymphocyte-associated antigen-4 (CD152)

FDA

US Food and Drug Administration;

HER-2

human epidermal growth factor receptor-2;

HLA

human leukocyte antigen;

ICOS

inducible T-cell costimulatory molecule;

IDO-1

indoleamine-2,3-dioxygenase-1;

IFN

interferon; IL-18, interleukin-18;

MAGE

melanoma antigen gene;

MDSC

myeloid-derived suppressor cell;

MHC

major histocompatibility complex;

MICA

MHC class I chain-related protein A;

MICB

MHC class I chain-related protein B;

MSI

microsatellite instability;

NK

cells natural killer cells;

NY-ESO-1

New York oesophageal squamous cell carcinoma-1 protein;

PD-1

programmed cell death protein 1 (CD279);

PD-L1

programmed cell death 1 ligand 1 (CD274)

RNA

ribonucleic acid;

TCR

T-cell receptor;

TREGs

regulatory T-cells

References

1 

de Vos-Geelen J, Hoebers FJP, Geurts SME, Hoeben A, de Greef BTA, Voncken FEM, Bogers JHA, Braam PM, Muijs CKT, de Jong MA, et al: A national study to assess outcomes of definitive chemoradiation regimens in proximal esophageal cancer. Acta Oncol. 59:895–903. 2020. View Article : Google Scholar : PubMed/NCBI

2 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. Feb 4–2021.Epub ahead of print. View Article : Google Scholar : PubMed/NCBI

3 

Lin Y, Totsuka Y, He Y, Kikuchi S, Qiao Y, Ueda J, Wei W, Inoue M and Tanaka H: Epidemiology of esophageal cancer in Japan and China. J Epidemiol. 23:233–242. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Napier KJ, Scheerer M and Misra S: Esophageal cancer: A Review of epidemiology, pathogenesis, staging workup and treatment modalities. World J Gastrointest Oncol. 6:112–120. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Bartley AN, Washington MK, Ventura CB, Ismaila N, Colasacco C, Benson AB III, Carrato A, Gulley ML, Jain D, Kakar S, et al: HER2 testing and clinical decision making in gastroesophageal adenocarcinoma: Guideline from the college of American pathologists American society for clinical pathology, and American society of clinical oncology. Arch Pathol Lab Med. 140:1345–1363. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Bang YJ, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki A, Lordick F, Ohtsu A, Omuro Y, Satoh T, et al: Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): A phase 3, open-label, randomised controlled trial. Lancet. 376:687–697. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Lordick F, Mariette C, Haustermans K, Obermannova R and Arnold D; ESMO Guidelines Committee: Oesophageal cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 27(Suppl 5): v50–v57. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Brar G and Shah MA: The role of pembrolizumab in the treatment of PD-L1 expressing gastric and gastroesophageal junction adenocarcinoma. Therap Adv Gastroenterol. 12:17562848198697672019. View Article : Google Scholar : PubMed/NCBI

9 

Le Bras GF, Farooq MH, Falk GW and Andl CD: Esophageal cancer: The latest on chemoprevention and state of the art therapies. Pharmacol Res. 113(Pt A): 236–244. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Short MW, Burgers KG and Fry VT: Esophageal cancer. Am Fam Physician. 95:22–28. 2017.PubMed/NCBI

11 

Kitagawa Y, Uno T, Oyama T, Kato K, Kato H, Kawakubo H, Kawamura O, Kusano M, Kuwano H, Takeuchi H, et al: Esophageal cancer practice guidelines 2017 edited by the Japan esophageal society: Part 2. Esophagus. 16:25–43. 2019. View Article : Google Scholar :

12 

Wang T, Yu J, Liu M, Chen Y, Zhu C, Lu L, Wang M, Min L, Liu X, Zhang X, et al: The benefit of taxane-based therapies over fluoropyrimidine plus platinum (FP) in the treatment of esophageal cancer: A meta-analysis of clinical studies. Drug Des Devel Ther. 13:539–553. 2019. View Article : Google Scholar : PubMed/NCBI

13 

Zhang Y: Epidemiology of esophageal cancer. World J Gastroenterol. 19:5598–5606. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Blot WJ: Invited commentary: More evidence of increased risks of cancer among alcohol drinkers. Am J Epidemiol. 150:1138–1140; discussion 1141. 1999. View Article : Google Scholar : PubMed/NCBI

15 

Guanrei Y and Songliang Q: Endoscopic surveys in high-risk and low-risk populations for esophageal cancer in China with special reference to precursors of esophageal cancer. Endoscopy. 19:91–95. 1987. View Article : Google Scholar : PubMed/NCBI

16 

Lagergren J and Lagergren P: Recent developments in esophageal adenocarcinoma. CA Cancer J Clin. 63:232–248. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Duggan C, Onstad L, Hardikar S, Blount PL, Reid BJ and Vaughan TL: Association between markers of obesity and progression from Barrett's esophagus to esophageal adenocarcinoma. Clin Gastroenterol Hepatol. 11:934–943. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Carr JS, Zafar SF, Saba N, Khuri FR and El-Rayes BF: Risk factors for rising incidence of esophageal and gastric cardia adenocarcinoma. J Gastrointest Cancer. 44:143–151. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Lofdahl HE, Lu Y, Lagergren P and Lagergren J: Risk factors for esophageal adenocarcinoma after antireflux surgery. Ann Surg. 257:579–582. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Mao WM, Zheng WH and Ling ZQ: Epidemiologic risk factors for esophageal cancer development. Asian Pac J Cancer Prev. 12:2461–2466. 2011.

21 

D'Onofrio V, Bovero E and Iaquinto G: Characterization of acid and alkaline reflux in patients with Barrett's esophagus. G.O.S.P.E. Operative Group for the study of Esophageal Precancer. Dis Esophagus. 10:16–22; discussion 22-3. 1997.PubMed/NCBI

22 

Fassan M, Realdon S, Cascione L, Hahne JC, Munari G, Guzzardo V, Arcidiacono D, Lampis A, Brignola S, Dal Santo L, et al: Circulating microRNA expression profiling revealed miR-92a-3p as a novel biomarker of Barrett's carcinogenesis. Pathol Res Pract. 216:1529072020. View Article : Google Scholar : PubMed/NCBI

23 

Lin J, Zeng R, Cao W, Luo R, Chen J and Lin Y: Hot beverage and food intake and esophageal cancer in southern China. Asian Pac J Cancer Prev. 12:2189–2192. 2011.

24 

Nieman KM, Romero IL, Van Houten B and Lengyel E: Adipose tissue and adipocytes support tumorigenesis and metastasis. Biochim Biophys Acta. 1831:1533–1541. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Lawrence MS, Stojanov P, Polak P, Kryukov GV, Cibulskis K, Sivachenko A, Carter SL, Stewart C, Mermel CH, Roberts SA, et al: Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature. 499:214–218. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Ratti M, Lampis A, Hahne JC, Passalacqua R and Valeri N: Microsatellite instability in gastric cancer: Molecular bases, clinical perspectives, and new treatment approaches. Cell Mol Life Sci. 75:4151–4162. 2018. View Article : Google Scholar : PubMed/NCBI

27 

Lin EW, Karakasheva TA, Hicks PD, Bass AJ and Rustgi AK: The tumor microenvironment in esophageal cancer. Oncogene. 35:5337–5349. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Ammannagari N and Atasoy A: Current status of immunotherapy and immune biomarkers in gastro-esophageal cancers. J Gastrointest Oncol. 9:196–207. 2018. View Article : Google Scholar : PubMed/NCBI

29 

Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, Skora AD, Luber BS, Azad NS, Laheru D, et al: PD-1 Blockade in tumors with Mismatch-Repair deficiency. N Engl J Med. 372:2509–2520. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Kim WK, Park M, Park M, Kim YJ, Shin N, Kim HK, You KT and Kim H: Identification and selective degradation of neopeptide-containing truncated mutant proteins in the tumors with high microsatellite instability. Clin Cancer Res. 19:3369–3382. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Puzzoni M, Silvestris N, Leone F, Giampieri R, Faloppi L, Demurtas L, Dell'Aquila E, Marino D, Brunetti O, Garattini SK, et al: The immune revolution in gastrointestinal tumours: Leading the way or just following? Target Oncol. 11:593–603. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Li P, Xu W, Liu F, Zhu H, Zhang L, Ding Z, Liang H and Song J: The emerging roles of IDO2 in cancer and its potential as a therapeutic target. Biomed Pharmacother. 137:1112952021. View Article : Google Scholar : PubMed/NCBI

33 

Zhou H, Jiang M, Yuan H, Ni W and Tai G: Dual roles of myeloid-derived suppressor cells induced by Toll-like receptor signaling in cancer. Oncol Lett. 21:1492021. View Article : Google Scholar : PubMed/NCBI

34 

Abou Khouzam R, Brodaczewska K, Filipiak A, Zeinelabdin NA, Buart S, Szczylik C, Kieda C and Chouaib S: Tumor hypoxia regulates immune Escape/Invasion: Influence on angiogenesis and potential impact of hypoxic biomarkers on cancer therapies. Front Immunol. 11:6131142021. View Article : Google Scholar : PubMed/NCBI

35 

Gajewski TF, Schreiber H and Fu YX: Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol. 14:1014–1022. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Ando M, Ito M, Srirat T, Kondo T and Yoshimura A: Memory T cell, exhaustion, and tumor immunity. Immunol Med. 43:1–9. 2020. View Article : Google Scholar

37 

Chen DS and Mellman I: Oncology meets immunology: The cancer-immunity cycle. Immunity. 39:1–10. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Hahne JC, Meyer SR, Gambaryan S, Walter U, Dietl J, Engel JB and Honig A: Immune escape of AKT overexpressing ovarian cancer cells. Int J Oncol. 42:1630–1635. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Boutet P, Aguera-Gonzalez S, Atkinson S, Pennington CJ, Edwards DR, Murphy G, Reyburn HT and Valés-Gómez M: Cutting edge: The metalloproteinase ADAM17/TNF-alpha-converting enzyme regulates proteolytic shedding of the MHC class I-related chain B protein. J Immunol. 182:49–53. 2009. View Article : Google Scholar

40 

Waldhauer I, Goehlsdorf D, Gieseke F, Weinschenk T, Wittenbrink M, Ludwig A, Stevanovic S, Rammensee HG and Steinle A: Tumor-associated MICA is shed by ADAM proteases. Cancer Res. 68:6368–6376. 2008. View Article : Google Scholar : PubMed/NCBI

41 

Pardoll DM: The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 12:252–264. 2012. View Article : Google Scholar : PubMed/NCBI

42 

Ricklefs FL, Alayo Q, Krenzlin H, Mahmoud AB, Speranza MC, Nakashima H, Hayes JL, Lee K, Balaj L, Passaro C, et al: Immune evasion mediated by PD-L1 on glioblastoma-derived extracellular vesicles. Sci Adv. 4:eaar27662018. View Article : Google Scholar : PubMed/NCBI

43 

Theodoraki MN, Yerneni SS, Hoffmann TK, Gooding WE and Whiteside TL: Clinical significance of PD-L1(+) exosomes in plasma of head and neck cancer patients. Clin Cancer Res. 24:896–905. 2018. View Article : Google Scholar

44 

Corthay A: How do regulatory T cells work? Scand J Immunol. 70:326–336. 2009. View Article : Google Scholar : PubMed/NCBI

45 

Romano M, Fanelli G, Albany CJ, Giganti G and Lombardi G: Past, present, and future of regulatory T Cell therapy in transplantation and autoimmunity. Front Immunol. 10:432019. View Article : Google Scholar : PubMed/NCBI

46 

Togashi Y, Shitara K and Nishikawa H: Regulatory T cells in cancer immunosuppression-implications for anticancer therapy. Nat Rev Clin Oncol. 16:356–371. 2019. View Article : Google Scholar : PubMed/NCBI

47 

Engel JB, Honig A, Kapp M, Hahne JC, Meyer SR, Dietl J and Segerer SE: Mechanisms of tumor immune escape in triple-negative breast cancers (TNBC) with and without mutated BRCA 1. Arch Gynecol Obstet. 289:141–147. 2014. View Article : Google Scholar

48 

Gabrilovich DI and Nagaraj S: Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol. 9:162–174. 2009. View Article : Google Scholar : PubMed/NCBI

49 

Gabrilovich DI: Myeloid-Derived suppressor cells. Cancer Immunol Res. 5:3–8. 2017. View Article : Google Scholar : PubMed/NCBI

50 

Karakasheva TA, Dominguez GA, Hashimoto A, Lin EW, Chiu C, Sasser K, Lee JW, Beatty GL, Gabrilovich DI and Rustgi AK: CD38+ M-MDSC expansion characterizes a subset of advanced colorectal cancer patients. JCI Insight. 3:e970222018. View Article : Google Scholar :

51 

Shi T, Ma Y, Yu L, Jiang J, Shen S, Hou Y and Wang T: Cancer immunotherapy: A focus on the regulation of immune check- points. Int J Mol Sci. 19:13892018. View Article : Google Scholar

52 

Kim ES, Kim JE, Patel MA, Mangraviti A, Ruzevick J and Lim M: Immune checkpoint modulators: An emerging anti-glioma armamentarium. J Immunol Res. 2016:46836072016. View Article : Google Scholar

53 

Vivaldi C, Catanese S, Massa V, Pecora I, Salani F, Santi S, Lencioni M, Vasile E, Falcone A and Fornaro L: Immune check-point inhibitors in esophageal cancers: Are we finally finding the right path in the mist? Int J Mol Sci. 21:16582020. View Article : Google Scholar

54 

Sharma P and Allison JP: The future of immune checkpoint therapy. Science. 348:56–61. 2015. View Article : Google Scholar : PubMed/NCBI

55 

Topalian SL, Drake CG and Pardoll DM: Immune checkpoint blockade: A common denominator approach to cancer therapy. Cancer Cell. 27:450–461. 2015. View Article : Google Scholar : PubMed/NCBI

56 

Disis ML: Mechanism of action of immunotherapy. Semin Oncol. 41(Suppl 5): S3–S13. 2014. View Article : Google Scholar : PubMed/NCBI

57 

Senju H, Kumagai A, Nakamura Y, Yamaguchi H, Nakatomi K, Fukami S, Shiraishi K, Harada Y, Nakamura M, Okamura H, et al: Effect of IL-18 on the expansion and phenotype of human natural killer cells: Application to cancer immunotherapy. Int J Biol Sci. 14:331–340. 2018. View Article : Google Scholar : PubMed/NCBI

58 

Patel SP and Kurzrock R: PD-L1 expression as a predictive biomarker in cancer immunotherapy. Mol Cancer Ther. 14:847–856. 2015. View Article : Google Scholar : PubMed/NCBI

59 

Mimura K, Yamada L, Ujiie D, Hayase S, Tada T, Hanayama H, Thar Min AK, Shibata M, Momma T, Saze Z, et al: Immunotherapy for esophageal squamous cell carcinoma: A review. Fukushima J Med Sci. 64:46–53. 2018. View Article : Google Scholar : PubMed/NCBI

60 

Redman JM, Gibney GT and Atkins MB: Advances in immunotherapy for melanoma. BMC Med. 14:202016. View Article : Google Scholar : PubMed/NCBI

61 

Figueroa-Protti L, Soto-Molinari R, Calderon-Osorno M, Mora J and Alpizar-Alpizar W: Gastric cancer in the Era of immune checkpoint blockade. J Oncol. 2019:10797102019. View Article : Google Scholar : PubMed/NCBI

62 

Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, Drake CG, Camacho LH, Kauh J, Odunsi K, et al: Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 366:2455–2465. 2012. View Article : Google Scholar : PubMed/NCBI

63 

Marchetti A, Di Lorito A and Buttitta F: Why anti-PD1/PDL1 therapy is so effective? Another piece in the puzzle. J Thorac Dis. 9:4863–4866. 2017. View Article : Google Scholar

64 

Akinleye A and Rasool Z: Immune checkpoint inhibitors of PD-L1 as cancer therapeutics. J Hematol Oncol. 12:922019. View Article : Google Scholar : PubMed/NCBI

65 

Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, Wolchok JD, Hersey P, Joseph RW, Weber JS, et al: Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med. 369:134–144. 2013. View Article : Google Scholar : PubMed/NCBI

66 

Robert C, Ribas A, Wolchok JD, Hodi FS, Hamid O, Kefford R, Weber JS, Joshua AM, Hwu WJ, Gangadhar TC, et al: Anti-progra mmed-death-receptor-1 treatment with pembrolizumab in ipilimumab-refractory advanced melanoma: A randomised dose-comparison cohort of a phase 1 trial. Lancet. 384:1109–1117. 2014. View Article : Google Scholar : PubMed/NCBI

67 

Maleki Vareki S, Garrigos C and Duran I: Biomarkers of response to PD-1/PD-L1 inhibition. Crit Rev Oncol Hematol. 116:116–124. 2017. View Article : Google Scholar : PubMed/NCBI

68 

Kato R, Yamasaki M, Urakawa S, Nishida K, Makino T, Morimoto-Okazawa A, Kawashima A, Iwahori K, Suzuki S, Ueda R, et al: Increased Tim-3(+) T cells in PBMCs during nivolumab therapy correlate with responses and prognosis of advanced esophageal squamous cell carcinoma patients. Cancer Immunol Immunother. 5467:1673–1683. 2018. View Article : Google Scholar

69 

Brahmer JR, Rodriguez-Abreu D, Robinson AG, Hui R, Csőszi T, Fülöp A, Gottfried M, Peled N, Tafreshi A, Cuffe S, et al: Health-related quality-of-life results for pembrolizumab versus chemotherapy in advanced, PD-L1-positive NSCLC (KEYNOTE-024): A multicentre, international, randomised, open-label phase 3 trial. Lancet Oncol. 18:1600–1609. 2017. View Article : Google Scholar : PubMed/NCBI

70 

Seiwert TY, Burtness B, Mehra R, Weiss J, Berger R, Eder JP, Heath K, McClanahan T, Lunceford J, Gause C, et al: Safety and clinical activity of pembrolizumab for treatment of recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-012): An open-label, multicentre, phase 1b trial. Lancet Oncol. 17:956–965. 2016. View Article : Google Scholar : PubMed/NCBI

71 

Fuchs CS, Doi T, Jang RW, Muro K, Satoh T, Machado M, Sun W, Jalal SI, Shah MA, Metges JP, et al: Safety and efficacy of pembrolizumab monotherapy in patients with previously treated advanced gastric and gastroesophageal junction cancer: Phase 2 Clinical KEYNOTE-059 Trial. JAMA Oncol. 4:e1800132018. View Article : Google Scholar : PubMed/NCBI

72 

Doi T, Piha-Paul SA, Jalal SI, Saraf S, Lunceford J, Koshiji M and Bennouna J: Safety and antitumor activity of the Anti-Programmed Death-1 antibody pembrolizumab in patients with advanced esophageal carcinoma. J Clin Oncol. 36:61–67. 2018. View Article : Google Scholar

73 

Hamanishi J, Mandai M, Ikeda T, Minami M, Kawaguchi A, Murayama T, Kanai M, Mori Y, Matsumoto S, Chikuma S, et al: Safety and antitumor activity of Anti-PD-1 Antibody, Nivolumab, in patients with platinum-resistant ovarian cancer. J Clin Oncol. 33:4015–4022. 2015. View Article : Google Scholar : PubMed/NCBI

74 

Kato K, Cho BC, Takahashi M, Okada M, Lin CY, Chin K, Kadowaki S, Ahn MJ, Hamamoto Y, Doki Y, 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. 20:1506–1517. 2019. View Article : Google Scholar : PubMed/NCBI

75 

Das R, Verma R, Sznol M, Boddupalli CS, Gettinger SN, Kluger H, Callahan M, Wolchok JD, Halaban R, Dhodapkar MV and Dhodapkar KM: Combination therapy with anti-CTLA-4 and anti-PD-1 leads to distinct immunologic changes in vivo. J Immunol. 194:950–959. 2015. View Article : Google Scholar

76 

Tanaka T, Nakamura J and Noshiro H: Promising immunotherapies for esophageal cancer. Expert Opin Biol Ther. 17:723–733. 2017. View Article : Google Scholar : PubMed/NCBI

77 

Janjigian YY, Bendell J, Calvo E, Kim JW, Ascierto PA, Sharma P, Ott PA, Peltola K, Jaeger D, Evans J, et al: CheckMate-032 study: Efficacy and safety of nivolumab and nivolumab plus ipilimumab in patients with metastatic esophagogastric cancer. J Clin Oncol. 36:2836–2844. 2018. View Article : Google Scholar : PubMed/NCBI

78 

Zhao Y, Yang W, Huang Y, Cui R, Li X and Li B: Evolving roles for Targeting CTLA-4 in cancer immunotherapy. Cell Physiol Biochem. 47:721–734. 2018. View Article : Google Scholar : PubMed/NCBI

79 

Darvin P, Toor SM, Sasidharan Nair V and Elkord E: Immune checkpoint inhibitors: Recent progress and potential biomarkers. Exp Mol Med. 50:1–11. 2018. View Article : Google Scholar : PubMed/NCBI

80 

Rosenberg SA and Restifo NP: Adoptive cell transfer as personalized immunotherapy for human cancer. Science. 348:62–68. 2015. View Article : Google Scholar : PubMed/NCBI

81 

Alsina M, Moehler M and Lorenzen S: Immunotherapy of esophageal cancer: Current status, many trials and innovative strategies. Oncol Res Treat. 41:266–271. 2018. View Article : Google Scholar : PubMed/NCBI

82 

Miliotou AN and Papadopoulou LC: CAR T-cell Therapy: A New Era in cancer immunotherapy. Curr Pharm Biotechnol. 19:5–18. 2018. View Article : Google Scholar : PubMed/NCBI

83 

Walseng E, Koksal H, Sektioglu IM, Fåne A, Skorstad G, Kvalheim G, Gaudernack G, Inderberg EM and Wälchli S: A TCR-based Chimeric antigen receptor. Sci Rep. 7:107132017. View Article : Google Scholar : PubMed/NCBI

84 

Robbins PF, Dudley ME, Wunderlich J, El-Gamil M, Li YF, Zhou J, Huang J, Powell DJ Jr and Rosenberg SA: Cutting edge: Persistence of transferred lymphocyte clonotypes correlates with cancer regression in patients receiving cell transfer therapy. J Immunol. 173:7125–7130. 2004. View Article : Google Scholar : PubMed/NCBI

85 

Dudley ME, Yang JC, Sherry R, Hughes MS, Royal R, Kammula U, Robbins PF, Huang J, Citrin DE, Leitman SF, et al: Adoptive cell therapy for patients with metastatic melanoma: Evaluation of intensive myeloablative chemoradiation preparative regimens. J Clin Oncol. 26:5233–5239. 2008. View Article : Google Scholar : PubMed/NCBI

86 

Toh U, Yamana H, Sueyoshi S, Tanaka T, Niiya F, Katagiri K, Fujita H, Shirozou K and Itoh K: Locoregional cellular immunotherapy for patients with advanced esophageal cancer. Clin Cancer Res. 6:4663–4673. 2000.

87 

Kageyama S, Ikeda H, Miyahara Y, Imai N, Ishihara M, Saito K, Sugino S, Ueda S, Ishikawa T, Kokura S, et al: Adoptive Transfer of MAGE-A4 T-cell Receptor Gene-Transduced lymphocytes in patients with recurrent esophageal cancer. Clin Cancer Res. 21:2268–2277. 2015. View Article : Google Scholar : PubMed/NCBI

88 

Li W, Joshi MD, Singhania S, Ramsey KH and Murthy AK: Peptide vaccine: Progress and challenges. Vaccines (Basel). 2:515–536. 2014. View Article : Google Scholar

89 

Masopust D and Schenkel JM: The integration of T cell migration, differentiation and function. Nat Rev Immunol. 13:309–320. 2013. View Article : Google Scholar : PubMed/NCBI

90 

Aranda F, Vacchelli E, Eggermont A, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G and Galluzzi L: Trial Watch: Peptide vaccines in cancer therapy. Oncoimmunology. 2:e266212013. View Article : Google Scholar

91 

Tacken PJ, de Vries IJ, Torensma R and Figdor CG: Dendritic-cell immunotherapy: From ex vivo loading to in vivo targeting. Nat Rev Immunol. 7:790–802. 2007. View Article : Google Scholar : PubMed/NCBI

92 

Hos BJ, Tondini E, van Kasteren SI and Ossendorp F: Approaches to improve chemically defined synthetic peptide vaccines. Front Immunol. 9:8842018. View Article : Google Scholar : PubMed/NCBI

93 

Chen DS and Mellman I: Elements of cancer immunity and the cancer-immune set point. Nature. 541:321–330. 2017. View Article : Google Scholar : PubMed/NCBI

94 

Blander JM: Regulation of the cell biology of antigen cross-presentation. Annu Rev Immunol. 36:717–753. 2018. View Article : Google Scholar : PubMed/NCBI

95 

Quakkelaar ED and Melief CJ: Experience with synthetic vaccines for cancer and persistent virus infections in nonhuman primates and patients. Adv Immunol. 114:77–106. 2012. View Article : Google Scholar : PubMed/NCBI

96 

Kono K: Current status of cancer immunotherapy. J Stem Cells Regen Med. 10:8–13. 2014. View Article : Google Scholar : PubMed/NCBI

97 

Fujiwara S, Wada H, Miyata H, Kawada J, Kawabata R, Nishikawa H, Gnjatic S, Sedrak C, Sato E, Nakamura Y, et al: Clinical trial of the intratumoral administration of labeled DC combined with systemic chemotherapy for esophageal cancer. J Immunother. 35:513–521. 2012. View Article : Google Scholar : PubMed/NCBI

98 

Robbins PF, Kassim SH, Tran TL, Crystal JS, Morgan RA, Feldman SA, Yang JC, Dudley ME, Wunderlich JR, Sherry RM, et al: A pilot trial using lymphocytes genetically engineered with an NY-ESO-1-reactive T-cell receptor: Long-term follow-up and correlates with response. Clin Cancer Res. 21:1019–1027. 2015. View Article : Google Scholar

99 

Daudi S, Eng KH, Mhawech-Fauceglia P, Morrison C, Miliotto A, Beck A, Matsuzaki J, Tsuji T, Groman A, Gnjatic S, et al: Expression and immune responses to MAGE antigens predict survival in epithelial ovarian cancer. PLoS One. 9:e1040992014. View Article : Google Scholar : PubMed/NCBI

100 

Kawabata R, Wada H, Isobe M, Saika T, Sato S, Uenaka A, Miyata H, Yasuda T, Doki Y, Noguchi Y, et al: Antibody response against NY-ESO-1 in CHP-NY-ESO-1 vaccinated patients. Int J Cancer. 120:2178–2184. 2007. View Article : Google Scholar : PubMed/NCBI

101 

Veit JA, Heine D, Thierauf J, Lennerz J, Shetty S, Schuler PJ, Whiteside T, Beutner D, Meyer M, Grünewald I, et al: Expression and clinical significance of MAGE and NY-ESO-1 cancer-testis antigens in adenoid cystic carcinoma of the head and neck. Head Neck. 38:1008–1016. 2016. View Article : Google Scholar : PubMed/NCBI

102 

Thomas R, Al-Khadairi G, Roelands J, Hendrickx W, Dermime S, Bedognetti D and Decock J: NY-ESO-1 based immunotherapy of cancer: Current perspectives. Front Immunol. 9:9472018. View Article : Google Scholar : PubMed/NCBI

103 

Bujas T, Marusic Z, Peric Balja M, Mijic A, Kruslin B and Tomas D: MAGE-A3/4 and NY-ESO-1 antigens expression in metastatic esophageal squamous cell carcinoma. Eur J Histochem. 55:e72011. View Article : Google Scholar : PubMed/NCBI

104 

Zhang H, Zhou X, Liu D, Zhu Y, Ma Q and Zhang Y: Progress and challenges of poersonalized neoantigens in the clinical treatment of tumors. Med Drug Disc. 6:1000302020. View Article : Google Scholar

105 

Huang TX and Fu L: The immune landscape of esophageal cancer. Cancer Commun (Lond). 39:792019. View Article : Google Scholar

106 

Tran E, Robbins PF and Rosenberg SA: 'Final common pathway' of human cancer immunotherapy: Targeting random somatic mutations. Nat Immunol. 18:255–262. 2017. View Article : Google Scholar : PubMed/NCBI

107 

Butterfield LH: Cancer vaccines. BMJ. 350:h9882015. View Article : Google Scholar : PubMed/NCBI

108 

Han XJ, Ma XL, Yang L, Wei YQ, Peng Y and Wei XW: Progress in neoantigen targeted cancer immunotherapies. Front Cell Dev Biol. 8:7282020. View Article : Google Scholar : PubMed/NCBI

109 

Jiang T, Shi T, Zhang H, Hu J, Song Y, Wei J, Ren S and Zhou C: Tumor neoantigens: From basic research to clinical applications. J Hematol Oncol. 12:932019. View Article : Google Scholar : PubMed/NCBI

110 

Sahin U, Derhovanessian E, Miller M, Kloke BP, Simon P, Löwer M, Bukur V, Tadmor AD, Luxemburger U, Schrörs B, et al: Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature. 547:222–226. 2017. View Article : Google Scholar : PubMed/NCBI

111 

Ott PA, Hu Z, Keskin DB, Shukla SA, Sun J, Bozym DJ, Zhang W, Luoma A, Giobbie-Hurder A, Peter L, et al: An immunogenic personal neoantigen vaccine for patients with melanoma. Nature. 547:217–221. 2017. View Article : Google Scholar : PubMed/NCBI

112 

Iinuma H, Fukushima R, Inaba T, Tamura J, Inoue T, Ogawa E, Horikawa M, Ikeda Y, Matsutani N, Takeda K, et al: Phase I clinical study of multiple epitope peptide vaccine combined with chemoradiation therapy in esophageal cancer patients. J Transl Med. 12:842014. View Article : Google Scholar : PubMed/NCBI

113 

Kono K, Iinuma H, Akutsu Y, Tanaka H, Hayashi N, Uchikado Y, Noguchi T, Fujii H, Okinaka K, Fukushima R, et al: Multicenter, phase II clinical trial of cancer vaccination for advanced esophageal cancer with three peptides derived from novel cancer-testis antigens. J Transl Med. 10:1412012. View Article : Google Scholar : PubMed/NCBI

114 

Yasuda T, Nishiki K, Yoshida K, Shiraishi O, Iwama M, Kato H, Yasuda A, Shinkai M, Chiba Y, Okuno K and Nakamura Y: Cancer peptide vaccine to suppress postoperative recurrence in esophageal SCC patients with induction of antigen-specific CD8+T cell. J Clin Oncol. 35(Suppl 15): e146352017. View Article : Google Scholar

115 

Murahashi M, Hijikata Y, Yamada K, Tanaka Y, Kishimoto J, Inoue H, Marumoto T, Takahashi A, Okazaki T, Takeda K, et al: Phase I clinical trial of a five-peptide cancer vaccine combined with cyclophosphamide in advanced solid tumors. Clin Immunol. 166-167:48–58. 2016. View Article : Google Scholar : PubMed/NCBI

116 

Harao M, Hirata S, Irie A, Senju S, Nakatsura T, Komori H, Ikuta Y, Yokomine K, Imai K, Inoue M, et al: HLA-A2-restricted CTL epitopes of a novel lung cancer-associated cancer testis antigen, cell division cycle associated 1, can induce tumor-reactive CTL. Int J Cancer. 123:2616–2625. 2008. View Article : Google Scholar : PubMed/NCBI

117 

Obara W, Eto M, Mimata H, Kohri K, Mitsuhata N, Miura I, Shuin T, Miki T, Koie T, Fujimoto H, et al: A phase I/II study of cancer peptide vaccine S-288310 in patients with advanced urothelial carcinoma of the bladder. Ann Oncol. 28:798–803. 2017. View Article : Google Scholar

118 

Yoshitake Y, Fukuma D, Yuno A, Hirayama M, Nakayama H, Tanaka T, Nagata M, Takamune Y, Kawahara K, Nakagawa Y, et al: Phase II clinical trial of multiple peptide vaccination for advanced head and neck cancer patients revealed induction of immune responses and improved OS. Clin Cancer Res. 21:312–321. 2015. View Article : Google Scholar

119 

Ungerechts G, Engeland CE, Buchholz CJ, Eberle J, Fechner H, Geletneky K, Holm PS, Kreppel F, Kühnel F, Lang KS, et al: Virotherapy research in Germany: From engineering to translation. Hum Gene Ther. 28:800–819. 2017. View Article : Google Scholar : PubMed/NCBI

120 

Moehler M, Goepfert K, Heinrich B, Breitbach CJ, Delic M, Galle PR and Rommelaere J: Oncolytic virotherapy as emerging immunotherapeutic modality: Potential of parvovirus h-1. Front Oncol. 4:922014. View Article : Google Scholar : PubMed/NCBI

121 

Conry RM, Westbrook B, McKee S and Norwood TG: Talimogene laherparepvec: First in class oncolytic virotherapy. Hum Vaccin Immunother. 14:839–846. 2018. View Article : Google Scholar : PubMed/NCBI

122 

Tanabe S, Tazawa H, Kagawa S, Noma K, Takehara K, Koujima T, Kashima H, Kato T, Kuroda S, Kikuchi S, et al: Phase I/II trial of endoscopic intratumoral administration of OBP-301, a novel telomerase-specific oncolytic virus, with radiation in elderly esophageal cancer patients. Cancer Res. 75:Abstract CT1232015.

123 

Nemunaitis J, Tong AW, Nemunaitis M, Senzer N, Phadke AP, Bedell C, Adams N, Zhang YA, Maples PB, Chen S, et al: A phase I study of telomerase-specific replication competent oncolytic adenovirus (telomelysin) for various solid tumors. Mol Ther. 18:429–434. 2010. View Article : Google Scholar :

124 

Sharabi AB, Lim M, DeWeese TL and Drake CG: Radiation and checkpoint blockade immunotherapy: Radiosensitisation and potential mechanisms of synergy. Lancet Oncol. 16:e498–e509. 2015. View Article : Google Scholar : PubMed/NCBI

125 

Humphries MP, Craig SG, Kacprzyk R, Fisher NC, Bingham V, McQuaid S, Murray GI, McManus D, Turkington RC, James J and Salto-Tellez M: The adaptive immune and immune checkpoint landscape of neoadjuvant treated esophageal adenocarcinoma using digital pathology quantitation. BMC Cancer. 20:5002020. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2021
Volume 47 Issue 6

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lampis A, Ratti M, Ghidini M, Mirchev MB, Okuducu AF, Valeri N and Hahne JC: Challenges and perspectives for immunotherapy in oesophageal cancer: A look to the future (Review). Int J Mol Med 47: 97, 2021
APA
Lampis, A., Ratti, M., Ghidini, M., Mirchev, M.B., Okuducu, A.F., Valeri, N., & Hahne, J.C. (2021). Challenges and perspectives for immunotherapy in oesophageal cancer: A look to the future (Review). International Journal of Molecular Medicine, 47, 97. https://doi.org/10.3892/ijmm.2021.4930
MLA
Lampis, A., Ratti, M., Ghidini, M., Mirchev, M. B., Okuducu, A. F., Valeri, N., Hahne, J. C."Challenges and perspectives for immunotherapy in oesophageal cancer: A look to the future (Review)". International Journal of Molecular Medicine 47.6 (2021): 97.
Chicago
Lampis, A., Ratti, M., Ghidini, M., Mirchev, M. B., Okuducu, A. F., Valeri, N., Hahne, J. C."Challenges and perspectives for immunotherapy in oesophageal cancer: A look to the future (Review)". International Journal of Molecular Medicine 47, no. 6 (2021): 97. https://doi.org/10.3892/ijmm.2021.4930