Minireviews Open Access
Copyright ©The Author(s) 2021. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Clin Cases. Dec 6, 2021; 9(34): 10438-10450
Published online Dec 6, 2021. doi: 10.12998/wjcc.v9.i34.10438
Role of immune escape in different digestive tumours
Xin-Zhu Du, Bin Wen, Lin Liu, Ying-Ting Wei, Kui Zhao, Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou Province, China
ORCID number: Xin-Zhu Du (0000-0003-0459-5024); Bin Wen (0000-0001-8413-0037); Lin Liu (0000-0001-7246-593X); Ying-Ting Wei (0000-0001-9434-7600); Kui Zhao (0000-0002-1639-0918).
Author contributions: Du XZ performed the literature search and wrote the manuscript; Wen B categorized the information; Liu L proofread the manuscript; Wei YT checked the information; and Zhao K revised the manuscript.
Conflict-of-interest statement: The authors have no conflicts of interest to disclose.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Kui Zhao, MD, Doctor, Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, No. 201 Dalian Road, Zunyi 563000, Guizhou Province, China. kuizhao95858@163.com
Received: July 25, 2021
Peer-review started: July 25, 2021
First decision: August 8, 2021
Revised: August 15, 2021
Accepted: October 18, 2021
Article in press: October 18, 2021
Published online: December 6, 2021

Abstract

A counterbalance between immune cells and tumour cells is key to fighting tumours, and immune escape is an important mechanism for the survival of tumour cells in the body. Tumor cells and their cytokines impair the activity of T cells, NK cells, macrophages and other immune cells through various ways, and change the expression of their own surface antigens so as to avoid the clearance of the immune system. Changes in major histocompatibility complex molecules, high expression of programmed death-ligand 1, and the presence of immunosuppressive cells in the tumor microenvironment (TME) are main means by which tumors impair the function of immune cells. During the development of tumours of the digestive system, different mechanisms acting on tumour cells, the TME, and immune cells lead to immune escape and promote tumour progression. In this paper, the mechanisms of immune escape in tumour cells of the digestive system are reviewed to provide a theoretical basis for the immunotherapy of gastrointestinal tumours.

Key Words: Gastrointestinal tumors, Immune escape, Immune cells, Tumor microenvironment, Molecular, Mechanism

Core Tip: To summarize and analyze the mechanisms of immune escape of tumor cells in the digestive system and provide help for immunotherapy. In this paper, the mechanisms can be analyzed from many aspects, including not only tumor cells themselves, but also immune cells and some other external factors. Through the summary of these mechanisms, we find some deficiencies in the research in this area, which may provide some ideas for the follow-up research.



INTRODUCTION

Digestive tumours are diseases with a high incidence worldwide, and the pathogenesis, clinical manifestations, and treatment of these tumours have been studied extensively. However, despite increased awareness and early screening for digestive tumours, only a few patients with distant metastases, such as colorectal cancer (CRC)[1], have long-term survival; CRC has the third-highest incidence of common tumours in the world and is the fourth leading cause of tumour-related death worldwide[2]. Therefore, it is particularly important to treat digestive tumours based on the root causes or pathogenesis. In recent years, many pieces of evidence have been obtained that support the view that the immune system plays an important role in tumorigenesis. Evading the surveillance of the immune system is also considered one of the markers of tumours. Some scholars believe that cancer immune editing includes three consecutive stages: Elimination, balance, and escape[3]. During the transformation of adenomas into malignant tumours, adenomatous dysplasia may represent an equilibrium phase, and malignant tumours may occur in the escape phase. Tumour cells escape attack from the immune system mainly by changing biological characteristics and microenvironments. Additionally, external factors can participate in immune escape. The mechanisms by which digestive tumours evade immune attack are summarized below (see Table 1).

Table 1 The mechanisms of immune escape in different digestive tumours.
Molecules/cells
Ref.
Tumor/cancer
Cells/cytokines
Up/down
Pathway/target
Outcomes
HLA-GBespalova et al[8], 2020; Liu et al[16], 2020CRCNK cells, T lymphocytes, and antigen-presenting cellsUpILT-2, ILt-4, and KIR2DL4By directly binding to the inhibitory receptors ILT-2, ILt-4, and KIR2DL4, leading to apoptosis of NK and T cells and weakening host immune defences
HLA-IZhao et al[11], 2011; Li et al[12], 2010; Özgül Özdemir et al[19], 2016Oesophageal malignant tumour, CRCCD8+ T cells, T lymphocytesDownTAADownregulates the expression of HLA-I and reduces the expression of tumour-associated antigen (TAA) on the surface of tumour cells, evading recognition and attack by immune cells
HLA-EHuang et al[17], 2017; Abd Hamid et al[18], 2019Early CRCCTLs and NK cellsUpCD94/NKG2AHLA-E is overexpressed on the surface of early CRC cells and can bind to the HLA-E receptor CD94/NKG2A, which is expressed on the surface of CTLs and NK cells, thus inhibiting their activity
PD-L1Calderaro et al[27], 2016Oesophageal carcinomaEGFRUpPI3K/AKT, EGFR-RAS-RAF-ERKBinding of the transmembrane protein - programmed death-ligand 1 (PD-L1) expressed in tumour cells or cells in the TME to PD-1 expressed on T cells can induce the production of immunosuppressive signals and decrease the proliferation of T cells, resulting in the depletion of T cells
Liu et al[30], 2020CRCCCL5Upp65/STAT3-CSN5-PD-L1Stabilizes PD-L1 in and out of cells through the p65/STAT3-CSN5-PD-L1 pathway mediated by NF-κB1 p65 (p65), which inhibits T-cell-mediated killing of HT29 tumour cells
Ghedini et al[31], 2018CRCFGFR2UpJAK/STAT3The tyrosine kinase domain initiates a series of intracellular signal cascade reactions, activates the JAK/STAT3 signalling pathway, and induces PD-L1 expression in CRC cells, thus participating in the occurrence and development of CRC
Li et al[34], 2019CRCCXCL5UpPIK3/AktThe binding of CXCL5 to CXCR2 on the surface of CRC cells promotes the movement of the CXCL5-CXCR2 axis, thus activating the PI3K/AKT signalling pathway and upregulating the expression of PD-L1 in CRC
Li et al[38], 2019Gallbladder malignant tumourT cellsUpPIK3/AktUpregulation of PD-L1 in gallbladder malignant tumour cells, activated the PIK3/Akt pathway, inhibited the cytotoxicity mediated by normal T cells, and promoted tumour growth and development
Galectin-9Wang et al[41], 2016; Halama et al[43], 2011Oesophageal carcinoma, CRCNK cellsDownRho/ROCK-1, F-actin polarizationThe low expression of Galectin-9 may lead to decreased activation or insufficient transport of NK cells to the tumour site
DKK2Xiao et al[44], 2018CRCNK cells, CD8+ T cellsUpSTATThe binding of DKK2 to LRP5 on the surface of NK cells leads to the disordering of STAT5 nuclear localization in NK cells and hinders the activation of NK cells
MDSCsGeiger et al[53], 2016CRCT cellsUpL-arginine The high expression of MDSCs consumes a large quantity of L-arginine, and the resulting depletion of L-arginine affects T-cell proliferation
Li et al[54], 2018Oesophageal carcinomaT cellsUpAkt1/rela/IL8 Oesophageal malignant tumour cells can guide MDSCs to migrate to the tumour site and promote tumour progression by activating the Akt1/rela/IL8 signalling pathway
Treg cellsChen et al[60], 2017CRCTCRUpCXCL13-CXCR5 axisHDCC mainly promotes the infiltration of Treg cells by binding to CXCR5 on the surface of Treg cells by secreting CXCL13, which initiates the CXCL13-CXCR5 axis, promotes the proliferation of Treg cells and the aggregation of Treg cells at the tumour site
TIM-3Shan et al[72], 2016Oesophageal carcinomaCD4+ Th1, CD8+ T cells, dysfunctional CD8+ T cells and FoxP3+ Treg cellsUpAKT/GSK-3 β/SnailA high expression of TIM-3 in tumour cells often indicates a poor prognosis of tumours
CD47Fujiwara-Tani et al[76], 2019Gastric tumour, CRCMacrophagesUpSirp αCD47 can prevent macrophage-mediated phagocytosis and antigen presentation by interacting with the receptor Sirp α expressed on macrophages, thus allowing tumour cells to escape the immune surveillance of macrophages
NF-κBOuyang et al[83], 2021; He et al[84], 2021Gallbladder malignant tumours, Pancreatic malignant tumours, CRCT cell granzyme B geneUpToll-like receptor 4, NF- κB/p65NF-κB realizes the immune escape of tumour cells by affecting the transcription of effector T cells at the cellular transcriptional level. NF-κB inhibits GZMB transcription in T cells, induces CTL dysfunction, and promotes tumour immune escape
IDO1+ Paneth cellsMezrich et al[90], 2010Oesophageal carcinoma, CRCCD8+ T cellsUpCanine uric acid, tryptophanIDO facilitates immune escape by locally increasing the level of canine uric acid derived from tumour epithelial cells and consuming tryptophan. The increased level of canine uric acid promotes the differentiation of Treg cells through the aromatic hydrocarbon receptor AhR29, and the depletion of tryptophan can lead to cell cycle arrest of T cells, both of which can inhibit the antitumour immune response
ALTERATIONS IN MAJOR HISTOCOMPATIBILITY COMPLEX MOLECULES INVOLVED IN IMMUNE ESCAPE

Major histocompatibility complex (MHC) is a protein complex loaded with short peptides on the cell surface that can be recognized by the T-cell receptor (TCR)[3,4]. Research shows that MHC-I molecules play an important role in the acquired immune response of vertebrates and the occurrence and development of digestive system tumours[5]. For example, HLA-G, a nonclassical MHC-class I molecule, has been demonstrated to be expressed in digestive system tumours[6]. HLA-G expression was first identified at the maternal-foetal interface of cytotrophoblast cells and was subsequently discovered to be involved in organ transplantation, malignant transformation, and autoimmune diseases, while allowing tumours or viruses to evade immune responses[7]. HLA-G exerts an inhibitory effect on NK cells, T lymphocytes, and antigen-presenting cells mainly by directly binding to the inhibitory receptors ILT-2, ILt-4, and KIR2DL4[8]. In addition, soluble HLA-G (sHLA-G) binds to CD8 helper receptors, leading to apoptosis of NK and T cells and weakening host immune defences[9].

Unlike HLA-G, which participates in immune escape by affecting immune cell function (mainly NK and T cells), HLA-I, a classical MHC molecule, downregulates its expression in tumour cells and reduces the expression of tumour-associated antigen (TAA) on the surface of tumour cells, thus evading recognition and attack by immune cells[10]. This process is one of the mechanisms by which oesophageal malignant tumour cells escape immune surveillance of CD8+ T cells[11]. For example, aflatoxin G1 precisely reduces the expression of immunoproteasome LMP-2 in oesophageal malignant tumour cells, and the resulting downregulation of HLA-I expression on the surface of tumour cells hinders the recognition of T lymphocytes and enables tumour cells to escape immune surveillance[12]. Downregulation or complete suppression of the HLA-I gene leads to inefficient antigen presentation and a decrease in the recognition rate of cytotoxic T lymphocytes (CTLs)[13], which suggests that the deletion of HLA-I molecules may be one of the advantages of the host evasion of immune defence. In studies on gastric malignancies associated with the Epstein-Barr virus (EBV) infection, it was found that microRNA encoded by EBV decreased the antigen presentation function of MHC-I molecules, thus enabling cells infected with EBV to escape the killing effect of immune cells[14]. MHC-II levels have also been found to be significantly higher in almost all EBV-related gastric malignancies than in normal tissues, unlike MHC-I levels[15]. This result suggests that the upregulation of the MHC-II molecule may also be involved in immune escape. The expression of HLA-G in CRC cells is associated with high tumour grades and poor prognosis[16]. In addition to HLA-G, the upregulation of another nonclassical MHC molecule, HLA-E, in CRC has also been confirmed to participate in immune escape. In contrast to HLA-G, HLA-E is primarily involved in the immunosuppressive response to early CRC[17]. HLA-E is overexpressed on the surface of early CRC cells and can bind to the HLA-E receptor CD94/NKG2A, which is expressed on the surface of CTLs and NK cells, thus inhibiting their activity[18]. Additionally, HLA-E expression can inhibit cetuximab-mediated antibody cytotoxicity and promote the immune escape of CRC[19]. Both nonclassical and classical MHC molecules, such as HLA-I, are expressed on CRC cells. Prognostic studies of patients with CRC suggest a poor overall survival rate in patients with deletion or downregulation of HLA-I[20]. This result supports the hypothesis that HLA-I is involved in immune escape. In a study on gastric cancer, patients with loss of expression of MHC-I molecules on the surface of tumour cells had shorter overall survival than those with normally expressed MHC-I molecules[21]. In pancreatic malignancies, tumour cells actively degrade MHC-I through the autophagy-lysosomal system, resulting in an MHC-I deficiency and providing favourable conditions for immune escape[22]. The alteration of MHC molecules has been found to significantly affect the immunogenicity of many gastrointestinal tumour cells. However, there is little evidence to support that changes in MHC molecules are involved in the immune escape of liver tumours, and further investigation is required to determine whether MHC molecules are involved in the tumour progression.

CYTOKINES INVOLVED IN IMMUNE ESCAPE

Tumor microenvironment (TME) consists of an extracellular matrix and mesenchymal cells, which produce cytokines that play an intermediary role in promoting tumour progression[23,24]. Binding of the transmembrane protein - programmed death-ligand 1 (PD-L1) expressed in tumour cells or cells in the TME to programmed death 1 (PD-1) expressed on T cells can induce the production of immunosuppressive signals and decrease the proliferation of T cells, resulting in the depletion of T cells. This binding process is one of the most important mechanisms of immune escape of tumour cells in the digestive system[25-27].

In the TME, the cytokines involved in the immune escape of tumour cells do not act directly on tumour cells but use signalling pathways to achieve PD-L1 upregulation involved in immune escape via different mechanisms. In oesophageal malignancies, the activation of an EGFR-dependent PI3K/AKT pathway upregulates PD-L1 on the surface of tumour cells. In addition to the PI3K/AKT pathway, the upregulation of PD-L1 in oesophageal carcinoma is also affected by the EGFR-RAS-RAF-ERK and EGR-PLC-γ signalling pathways[28]. In gastric malignant tumour cells associated with EBV infection, the expression of PD-L1 is a common feature, and the overexpression of PD-L1 is associated with poor prognosis, but the mechanism is not clear. In addition, a significantly elevated PD-1+ lymphocyte count has been found in tumour stroma[29]. Some studies have shown a significant increase in the average expression level of PD-1 on T cells in peripheral blood and cancer tissues of patients with gastric cancer, suggesting that the PD-L1/PD-1 pathway is involved in immune escape in gastric cancer. Studies have also shown that a variety of cytokines participate in the immune escape process of CRC cells. For example, CCL5, a cytokine C-motif chemokine ligand 5 (CCL5) from tumour-associated macrophages (TAMs), stabilizes PD-L1 in and out of cells through the p65/STAT3-CSN5-PD-L1 pathway mediated by NF-κB1 p65 (p65), which inhibits T-cell-mediated killing of HT29 tumour cells, which is key for CRC cells to escape immune surveillance[30]. Not only do TAMs participate in the upregulation of PD-L1, but TAM subtype M2 macrophages also participate in the upregulation of PD-1 expression, which promotes the binding of PD-L1/PD-1. In addition, in the TME, VEGF-c is a growth factor involved in tumour-associated lymphangiogenesis that can also promote M2-mediated immune escape by changing the density of TAMs, as well as increasing the percentage of M2/M1 macrophages in TAMs and the M2 survival rate. The tumour-derived fibroblast growth factor receptor (FGFR)-2 binds to ligand fibroblast growth factors (FGFs) in the tumour microenvironment and undergoes dimerization (receptor pairing). The tyrosine kinase domain initiates a series of intracellular signal cascade reactions, activates the JAK/STAT3 signalling pathway, and induces PD-L1 expression in CRC cells, thus participating in the occurrence and development of CRC[31,32]. PD-L1 and FGFR2 have been found to be overexpressed in CRC and positively correlated with each other[33]. It has also been observed that the overexpression of FGFR2 in CRC not only increased the apoptosis rate of T cells but was also correlated with lymph node metastasis, clinical stage cancer, and a poor survival rate. In addition to the JAK/STAT3 signalling pathway, the PIK3/Akt pathway, which has a high activation, is also one of the means of PD-L1 upregulation, with chemokine-5 (CXCL5) from cancer-associated fibroblasts (CAFs) as the medium[34,35]. CXCL5 is an effective cytokine that affects the TME in many ways, of which the PI3K/AKT signalling pathway is the most common. The binding of CXCL5 to CXCR2 on the surface of CRC cells promotes the movement of the CXCL5-CXCR2 axis, thus activating the PI3K/AKT signalling pathway and upregulating the expression of PD-L1 in CRC[36]. In liver tumour cells, the expression of PD-L1 decreases antitumour immune ability and promotes the immune escape of tumour cells[37]; PD-L1 expression has been shown to be related to the invasiveness of tumour cells[27]. It has also been observed that PD-L1 expression was upregulated in gallbladder malignant tumour cells, activated the PIK3/Akt pathway, inhibited the cytotoxicity mediated by normal T cells, and promoted tumour growth and development[38]. PD-L1 expression is higher in invasive pancreatic malignant tumours, and the non-Smad-β signalling pathway mediated by the transforming growth factor in the TME leads to more invasive phenotypes and immunosuppression mediated by PD-L1[39]. All these results confirm that PD-L1 is involved in the immune escape of digestive system tumours.

In some digestive system tumours (such as an oesophageal malignant tumour, CRC), some cytokines can also promote immune escape by affecting the function of immune cells. For example, Galectin-9 is a widely expressed protein in the TME that plays a dual role in the immune escape of tumour cells. Galectin-9 can not only affect the activity of NK cells through the signalling pathway mediated by TIM-3 (an immunosuppressive molecule) but can also bind to effector CD8+ T cells expressing TIM-3 molecules in the TME, leading to apoptosis and promoting the occurrence of antitumour immunosuppression[40]. However, Galectin-9, as a protective factor against tumours, can also increase the recruitment of NK cells by affecting the expression of Rho/ROCK-1 and F-actin polarization[41]. Most tumour cells express Galectin-9 but at a lower positive rate and expression level than in normal tissues. In a prognostic study of patients with an oesophageal malignant tumour, it was found that low expression of Galectin-9 was closely related to poor prognosis[42]. These results suggest that the low expression of Galectin-9 may lead to decreased activation or insufficient transport of NK cells to the tumour site[43], which may be a new mechanism of tumour cell immune escape. In CRC, transcriptional activator (STAT) is the key regulator of NK cell functional activation, and dickkopf-associated protein 2 (DKK2), which is exploited via the high expression of a secretory protein in CRC tissue to affect the function of NK cells. The binding of DKK2 to LRP5 on the surface of NK cells leads to the disordering of STAT5 nuclear localization in NK cells and hinders the activation of NK cells[44]. DKK2 can also block the activation of CD8+ T cells, not by direct action on T cells but by indirect regulation of CD8+ T cells after direct interaction with NK cells[45]. In addition, DKK2 can inhibit the antitumour immune response by inhibiting the activation of CD8+ T cells mediated by IL-15[46].

Some studies have shown that, in addition to programmed death ligands, apoptosis antigen 1 (Fas) is also involved in the immune escape of digestive system tumours. Fas promotes apoptosis of tumour cells, whereas Fas ligand (FasL) has a protective effect on Fas-mediated apoptosis[17]. It has been found that FasL expressed by pancreatic tumor cells can avoid immune surveillance by inducing apoptosis of infiltrating lymphocytes around tumour tissue[47], but relatively few studies on FasL have been conducted to date.

IMMUNOSUPPRESSIVE CELLS INVOLVED IN IMMUNE ESCAPE

There are two important immunosuppressive cells in the TME, myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Treg cells). These two types of cells function both interdependently and independently during the process of suppression of antitumour immunity by digestive system tumour cells[48-50]. As the most common immunosuppressive cells, MDSCs exert inhibitory effects, such as on the production of anti-TAA antibodies and T cells and the function of NK cells, mainly through the transforming growth factor-β[51]. However, in digestive system tumours, MDSCs participate in immune escape mainly by inhibiting T-cell proliferation[52]. An important factor related to the inhibitory effect of MDSCs on T cells is L-arginine, an amino acid essential for T-cell proliferation and normal function. The high expression of MDSCs consumes a large quantity of L-arginine, and the resulting depletion of L-arginine affects T-cell proliferation[53]. Many scholars have confirmed that MDSCs are involved in the immune escape of digestive system tumours. Studies have shown that oesophageal malignant tumour cells can guide MDSCs to migrate to the tumour site and promote tumour progression by activating the Akt1/rela/IL8 signalling pathway[54]. The immunosuppressive effect of MDSCs on T cells in gastric tumour tissue has been related to the transforming growth factor β[55], and the upregulated expression of MDSCs in liver tumour cells has been related to poor prognosis[56].

In addition to affecting T-cell function, another important role of MDSCs is to stimulate the development of another key immunosuppressive cell, Treg cells, which can directly inhibit the TCR-mediated immune response, leading to antitumour suppression[57-59]. The CRC process involves another cell related to Treg cells, HDCC, a myeloid cell expressing histidine decarboxylase[60]. HDCC mainly promotes the infiltration of Treg cells by binding to CXCR5 on the surface of Treg cells by secreting CXCL13, which initiates the CXCL13-CXCR5 axis, promotes the proliferation of Treg cells and the aggregation of Treg cells at the tumour site. HDCC can also affect the function of Treg T cells directly or indirectly by regulating the function of CD8C T cells and thus plays an important role in inhibiting antitumour immunity. CD70+CAFs may also play a role in immune escape by promoting the aggregation of Treg cells and increasing the migration ability of Treg cells[61].

MDSCs and Treg cells complement each other. MDSCs can induce the production of Treg cells; conversely, Treg cells can stimulate the production of MDSCs through positive feedback of the transforming growth factor-β[51]. MDSCs can not only damage the activation of T cells by producing O2 and iNOs but also cooperate with the VEGF to induce angiogenesis around tumour cells and directly stimulate tumour growth and metastasis[62-65]. In the TME, many favourable factors act as "fertile soil" for the growth of MDSCs to promote the immune escape of digestive system tumours. Factors such as PGE2, IL-6, IL-10, LTB4, and histamine are involved in the induction of MDSCs; local hypoxia and low pH in the TME can stimulate the expression of MDSCs, and S100A9 is a proinflammatory molecule that can induce an immunosuppressive microenvironment by regulating the chemotaxis and activation of MDSCs in digestive system tumours[66].

IMMUNOSUPPRESSIVE MOLECULES INVOLVED IN IMMUNE ESCAPE

Some immunosuppressive molecules in tumour cells, immune cells, and other immune-related cells, such as TIM-3, CD47, and NF-kB, play a key role in the process of immune escape of tumours.

TIM-3 is a coinhibitory receptor that can be expressed not only in immune cells (including immunosuppressive cells) and tumour cells but also in other cells. TIM-3 acts as a negative regulator in immune cells (such as CD4+ Th1 and CD8+ T cells) and thus plays an important role in T-cell depletion in a variety of environments[67], whereas TIM-3 expression is promoted in immunosuppressive cells (such as dysfunctional CD8+ T cells and FoxP3+ Treg cells, two key cells in tumour development)[68,69]. TIM-3 is also expressed in cell types other than the two abovementioned cell types[70], including myeloid cells and digestive system tumour cells themselves. A high expression of TIM-3 in tumour cells often indicates a poor prognosis of tumours[71], and studies have shown that TIM-3 can induce metastasis of oesophageal malignant tumours through the AKT/GSK-3 β/Snail signalling pathway[72]. In this regard, the TIM-3 molecule is a very suitable target for antitumour immunotherapy, and an in-depth study of TIM-3 may help identify new directions in immunotherapy.

The upregulation of CD47 expression in some digestive system tumours has been confirmed[73-75] and is closely related to the occurrence of gastric tumours associated with EBV infection[74]. CD47 can prevent macrophage-mediated phagocytosis and antigen presentation by interacting with the receptor Sirp α expressed on macrophages, thus allowing tumour cells to escape the immune surveillance of macrophages[76]. Furthermore, CD47 promotes the proliferation and metastasis of CRC cells by increasing aerobic glycolysis and activating the MAPK signalling pathway[77]. When blocking the PD-L1/PD-1 axis with an anti-PD-1 antibody, CD47Sirp α signal transduction can be weakened[78], suggesting that there may be a common pathway between the PD-L1/PD-1 axis and CD47-mediated immune escape, which is extremely significant for the further study of anti-immunotherapy.

Another common immunosuppressive molecule, NF-κB, is widely found in tumour cells of the digestive system and participates in immune escape mainly by affecting the function of T cells. In a study on the effect of microtubule-associated serine/threonine kinase (MAST1) on gastric malignant tumours, it was found that tumour cells after MAST1 gene knockout exerted an antitumour effect by downregulating the expression of NF-κB p65, suggesting that NF-κB may be involved in the immune escape of tumour cells[79,80]. NF-κB1 p50 (hereinafter referred to as p50) realizes the immune escape of tumour cells by affecting the transcription of effector T cells at the cellular transcriptional level[81,82]. P50 is a transcriptional inhibitor of the T cell granzyme B gene (GZMB). By binding to an unknown κB element in the GZMBb promoter, p50 inhibits GZMB transcription in T cells, induces CTL dysfunction, and promotes tumour immune escape. The activation of p50 and the expression of GZMB have been found to be negatively correlated with the degree of T-cell infiltration in CRC; that is, in CRC with a high level of p50 activation, the expression of GZMB was downregulated and T-cell infiltration decreased, whereas in CRC with a low level of p50 activation, the expression of GZMB was upregulated and T-cell infiltration increased. During the development of colitis into CRC, the colitis-related immune response may first activate p50 and damage the function of CTL effectors, leading to the immune escape of transformed epithelial cells and tumour development. In addition, in gallbladder malignant tumours, the downregulated expression of miR-146b-5p increases the expression of Toll-like receptor 4 (TLR4) and indirectly activates the NF-κB signalling pathway, which regulates tumour development[83]. Tumour proliferation, epithelial transformation, and stem cell-like characteristics were found to be inhibited when the phosphorylation pathway of NF-κB/p65 was blocked in pancreatic malignant tumour cells[84], which suggests that NF-κB is involved in the development of pancreatic malignant tumours. Therefore, the activation of NF-kB is considered to be a key link in the carcinogenesis of the human digestive system[85].

OTHER PROCESSES INVOLVED IN IMMUNE ESCAPE

The interaction between tumour cells and immune cells plays an important role in the occurrence of inflammation-related tumours. The reaction between tumour cells and immune cells has two facets: CTLs activated by antitumour action inhibit tumour growth, while chronic inflammation creates a microenvironment that promotes tumour cell growth and invasion. Studies have shown that an elevated level of the inflammatory cytokine IL-6 can upregulate the expression of the cell adhesion molecule ICAM1 through the STAT3/5, ERK, and Rho-ROCK signalling pathways and promote the formation of chronic inflammation and lymphocyte death in tumour tissue, which enables the tumour to evade immune attack[86].

The inflammatory process induces tumour or immune cells to release cytokines[87] and is part of tumour immunosuppression, which plays an important role in tumour immune escape of the digestive system. In addition to the mechanism of the inflammatory reaction, another type of cell, the STAT1-dependent indole-2-dioxygenase-1 (IDO1)+ Paneth cell, plays an essential role in immune escape. In oesophageal malignant tumours, the expression of IDO has been found to impair the function of CD8+ T cells and promote the immune escape of oesophageal malignant tumours[88,89]. IDO facilitates immune escape by locally increasing the level of canine uric acid derived from tumour epithelial cells and consuming tryptophan. The increased level of canine uric acid promotes the differentiation of Treg cells through the aromatic hydrocarbon receptor AhR29[90], and the depletion of tryptophan can lead to cell cycle arrest of T cells[91], both of which can inhibit the antitumour immune response. In addition, IDO participates in the immune escape of colitis-associated CRC, where IDO1+ Paneth cells exist in both cancer tissue and normal intestinal recess[92]. IDO1+ Paneth cells can be used as local immunosuppressants to prevent the abnormal activation of immune cells by bacteria and promote tumour progression.

In recent years, in-depth study of the digestive system has resulted in the identification of factors related to immune escape. For example, in a study on the relationship between intestinal flora and CRC, it was found that Clostridium could expand myeloid immune cells, inhibit the proliferation of T cells in CRC and induce T-cell apoptosis[93]. In blood circulation, platelets inhibit the immune response of T cells to CRC through the GARP-TGF-b axis; thus, drugs such as clopidogrel can improve the immunosuppressive response through antiplatelet aggregation[94]. In the TME, high levels of insulin and epidermal growth factor may be risk factors for tumour escape. When the TME is anoxic, lactic acid produced by tumour cells can weaken the differentiation and effector function of T cells and monocytes[95]. In addition, some RNAs can promote immune escape by affecting the function of immune cells. For example, microRNA-21 induces immunosuppression of CRC by increasing the levels of IL-10 and PGE2 (PGE2 inhibits DCs, macrophages, neutrophils, CTLs, TH1 cells, and NK cells and stimulates the production of MDSCs, Treg cells, and TH2)[96], and circRNA participates in the immune escape of liver tumour cells by regulating the function of NK cells[97].

CONCLUSION

The immune system is the body’s protective barrier,and immune escape is really the umbrella for tumors. The immune escape of tumour cells in the digestive system is mainly realized by changing the expression of MHC molecules through various mechanisms; affecting the function of T cells, NK cells, and other immune cells; stimulating the activation and accumulation of immunosuppressive cells; and finally promote the immune escape of tumour cells, which is the key breakthrough of tumour immunotherapy. Among carcinogenic mechanisms, the immune escape mechanism of tumour cells provides a reliable basis for the further study of immunotherapy, which is expected to become a milestone in the history of tumour treatment that goes beyond surgical treatment, radiotherapy, and chemotherapy. Existing studies have confirmed that immune escape is involved in the development of most digestive system tumors, but it has been rarely reported in liver cancer, whether the immune escape is involved in the formation of it needs further study in the future. Although the development of digestive system tumours is closely related to the immune escape mechanism, it is difficult to block tumour progression at the root using a single locking mechanism; thus, many aspects and multiple dimensions of the immune escape mechanism need to be studied in the future. In-depth exploration of various mechanisms will help lay a theoretical foundation for further progress in immunotherapy.

ACKNOWLEDGEMENTS

The authors thank Professor Tuo BG (Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College) for professional assistance.

Footnotes

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

Specialty type: Immunology

Country/Territory of origin: China

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): C

Grade D (Fair): D

Grade E (Poor): 0

P-Reviewer: Wang Z, Zharikov YO S-Editor: Yan JP L-Editor: A P-Editor: Yan JP

References
1.  Cao H, Xu E, Liu H, Wan L, Lai M. Epithelial-mesenchymal transition in colorectal cancer metastasis: A system review. Pathol Res Pract. 2015;211:557-569.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 215]  [Cited by in F6Publishing: 269]  [Article Influence: 29.9]  [Reference Citation Analysis (0)]
2.  Dekker E, Tanis PJ, Vleugels JLA, Kasi PM, Wallace MB. Colorectal cancer. Lancet. 2019;394:1467-1480.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1570]  [Cited by in F6Publishing: 2068]  [Article Influence: 413.6]  [Reference Citation Analysis (2)]
3.  Rodríguez JA. HLA-mediated tumor escape mechanisms that may impair immunotherapy clinical outcomes via T-cell activation. Oncol Lett. 2017;14:4415-4427.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 48]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
4.  Zajonc DM. Unconventional Peptide Presentation by Classical MHC Class I and Implications for T and NK Cell Activation. Int J Mol Sci. 2020;21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 3]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
5.  Kuznetsov A, Voronina A, Govorun V, Arapidi G. Critical Review of Existing MHC I Immunopeptidome Isolation Methods. Molecules. 2020;25.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 12]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
6.  Swets M, Seneby L, Boot A, van Wezel T, Gelderblom H, van de Velde CJ, van den Elsen PJ, Kuppen PJ. Promoter methylation and mRNA expression of HLA-G in relation to HLA-G protein expression in colorectal cancer. Hum Immunol. 2016;77:764-772.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 9]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
7.  Xu X, Zhou Y, Wei H. Roles of HLA-G in the Maternal-Fetal Immune Microenvironment. Front Immunol. 2020;11:592010.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 73]  [Article Influence: 18.3]  [Reference Citation Analysis (0)]
8.  Bespalova O, Bakleicheva M, Ivashchenko T, Tral T, Tolibova G, Kogan I. Expression of HLA-G and KIR2DL4 receptor in chorionic villous in missed abortion. Gynecol Endocrinol. 2020;36:43-47.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
9.  Kirana C, Ruszkiewicz A, Stubbs RS, Hardingham JE, Hewett PJ, Maddern GJ, Hauben E. Soluble HLA-G is a differential prognostic marker in sequential colorectal cancer disease stages. Int J Cancer. 2017;140:2577-2586.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 23]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
10.  Swets M, König MH, Zaalberg A, Dekker-Ensink NG, Gelderblom H, van de Velde CJ, van den Elsen PJ, Kuppen PJ. HLA-G and classical HLA class I expression in primary colorectal cancer and associated liver metastases. Hum Immunol. 2016;77:773-779.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 29]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
11.  Zhao X, Sun Q, Tian H, Cong B, Jiang X, Peng C. Loss of heterozygosity at 6p21 and HLA class I expression in esophageal squamous cell carcinomas in China. Asian Pac J Cancer Prev. 2011;12:2741-2745.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
12.  Li Z, Cui J, Zhang X, Kang W. Aflatoxin G1 reduces the molecular expression of HLA-I, TAP-1 and LMP-2 of adult esophageal epithelial cells in vitro. Toxicol Lett. 2010;195:169-173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
13.  Garrido F. HLA Class-I Expression and Cancer Immunotherapy. Adv Exp Med Biol. 2019;1151:79-90.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 30]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
14.  Ghasemi F, Gameiro SF, Tessier TM, Maciver AH, Mymryk JS. High Levels of Class I Major Histocompatibility Complex mRNA Are Present in Epstein-Barr Virus-Associated Gastric Adenocarcinomas. Cells. 2020;9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 17]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
15.  Ghasemi F, Tessier TM, Gameiro SF, Maciver AH, Cecchini MJ, Mymryk JS. High MHC-II expression in Epstein-Barr virus-associated gastric cancers suggests that tumor cells serve an important role in antigen presentation. Sci Rep. 2020;10:14786.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 21]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
16.  Liu L, Wang L, Zhao L, He C, Wang G. The Role of HLA-G in Tumor Escape: Manipulating the Phenotype and Function of Immune Cells. Front Oncol. 2020;10:597468.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 21]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
17.  Huang R, Zhang D, Li F, Xiao Z, Wu M, Shi D, Xiang P, Bao Z. Loss of Fas expression and high expression of HLA-E promoting the immune escape of early colorectal cancer cells. Oncol Lett. 2017;13:3379-3386.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 18]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
18.  Abd Hamid M, Wang RZ, Yao X, Fan P, Li X, Chang XM, Feng Y, Jones S, Maldonado-Perez D, Waugh C, Verrill C, Simmons A, Cerundolo V, McMichael A, Conlon C, Wang X, Peng Y, Dong T. Enriched HLA-E and CD94/NKG2A Interaction Limits Antitumor CD8+ Tumor-Infiltrating T Lymphocyte Responses. Cancer Immunol Res. 2019;7:1293-1306.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 41]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
19.  Özgül Özdemir RB, Özdemir AT, Oltulu F, Kurt K, Yiğittürk G, Kırmaz C. A comparison of cancer stem cell markers and nonclassical major histocompatibility complex antigens in colorectal tumor and noncancerous tissues. Ann Diagn Pathol. 2016;25:60-63.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
20.  Anderson P, Aptsiauri N, Ruiz-Cabello F, Garrido F. HLA class I loss in colorectal cancer: implications for immune escape and immunotherapy. Cell Mol Immunol. 2021;18:556-565.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 46]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
21.  Goeppert B, Frauenschuh L, Zucknick M, Roessler S, Mehrabi A, Hafezi M, Stenzinger A, Warth A, Pathil A, Renner M, Schirmacher P, Weichert W. Major histocompatibility complex class I expression impacts on patient survival and type and density of immune cells in biliary tract cancer. Br J Cancer. 2015;113:1343-1349.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 43]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
22.  Yamamoto K, Venida A, Yano J, Biancur DE, Kakiuchi M, Gupta S, Sohn ASW, Mukhopadhyay S, Lin EY, Parker SJ, Banh RS, Paulo JA, Wen KW, Debnath J, Kim GE, Mancias JD, Fearon DT, Perera RM, Kimmelman AC. Autophagy promotes immune evasion of pancreatic cancer by degrading MHC-I. Nature. 2020;581:100-105.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 335]  [Cited by in F6Publishing: 585]  [Article Influence: 146.3]  [Reference Citation Analysis (0)]
23.  Arneth B. Tumor Microenvironment. Medicina (Kaunas). 2019;56.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 262]  [Cited by in F6Publishing: 578]  [Article Influence: 115.6]  [Reference Citation Analysis (0)]
24.  Zhang Y, Lou Y, Wang J, Yu C, Shen W. Research Status and Molecular Mechanism of the Traditional Chinese Medicine and Antitumor Therapy Combined Strategy Based on Tumor Microenvironment. Front Immunol. 2020;11:609705.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 61]  [Article Influence: 20.3]  [Reference Citation Analysis (0)]
25.  Wang C, Weng M, Xia S, Zhang M, Chen C, Tang J, Huang D, Yu H, Sun W, Zhang H, Lai M. Distinct roles of programmed death ligand 1 alternative splicing isoforms in colorectal cancer. Cancer Sci. 2021;112:178-193.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 19]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
26.  Juneja VR, McGuire KA, Manguso RT, LaFleur MW, Collins N, Haining WN, Freeman GJ, Sharpe AH. PD-L1 on tumor cells is sufficient for immune evasion in immunogenic tumors and inhibits CD8 T cell cytotoxicity. J Exp Med. 2017;214:895-904.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 429]  [Cited by in F6Publishing: 557]  [Article Influence: 79.6]  [Reference Citation Analysis (0)]
27.  Calderaro J, Rousseau B, Amaddeo G, Mercey M, Charpy C, Costentin C, Luciani A, Zafrani ES, Laurent A, Azoulay D, Lafdil F, Pawlotsky JM. Programmed death ligand 1 expression in hepatocellular carcinoma: Relationship With clinical and pathological features. Hepatology. 2016;64:2038-2046.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 260]  [Cited by in F6Publishing: 302]  [Article Influence: 37.8]  [Reference Citation Analysis (0)]
28.  Zhang W, Pang Q, Yan C, Wang Q, Yang J, Yu S, Liu X, Yuan Z, Wang P, Xiao Z. Induction of PD-L1 expression by epidermal growth factor receptor-mediated signaling in esophageal squamous cell carcinoma. Onco Targets Ther. 2017;10:763-771.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 39]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
29.  Saito R, Abe H, Kunita A, Yamashita H, Seto Y, Fukayama M. Overexpression and gene amplification of PD-L1 in cancer cells and PD-L1+ immune cells in Epstein-Barr virus-associated gastric cancer: the prognostic implications. Mod Pathol. 2017;30:427-439.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 115]  [Article Influence: 16.4]  [Reference Citation Analysis (0)]
30.  Liu C, Yao Z, Wang J, Zhang W, Yang Y, Zhang Y, Qu X, Zhu Y, Zou J, Peng S, Zhao Y, Zhao S, He B, Mi Q, Liu X, Zhang X, Du Q. Macrophage-derived CCL5 facilitates immune escape of colorectal cancer cells via the p65/STAT3-CSN5-PD-L1 pathway. Cell Death Differ. 2020;27:1765-1781.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 102]  [Article Influence: 20.4]  [Reference Citation Analysis (0)]
31.  Ghedini GC, Ronca R, Presta M, Giacomini A. Future applications of FGF/FGFR inhibitors in cancer. Expert Rev Anticancer Ther. 2018;18:861-872.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 45]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
32.  Babina IS, Turner NC. Advances and challenges in targeting FGFR signalling in cancer. Nat Rev Cancer. 2017;17:318-332.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 419]  [Cited by in F6Publishing: 469]  [Article Influence: 67.0]  [Reference Citation Analysis (0)]
33.  Li P, Huang T, Zou Q, Liu D, Wang Y, Tan X, Wei Y, Qiu H. FGFR2 Promotes Expression of PD-L1 in Colorectal Cancer via the JAK/STAT3 Signaling Pathway. J Immunol. 2019;202:3065-3075.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 99]  [Article Influence: 19.8]  [Reference Citation Analysis (0)]
34.  Li Z, Zhou J, Zhang J, Li S, Wang H, Du J. Cancer-associated fibroblasts promote PD-L1 expression in mice cancer cells via secreting CXCL5. Int J Cancer. 2019;145:1946-1957.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 91]  [Article Influence: 18.2]  [Reference Citation Analysis (0)]
35.  Yildirim K, Colak E, Aktimur R, Gun S, Taskin MH, Nigdelioglu A, Aktimur SH, Karagöz F, Ozlem N. Clinical Value of CXCL5 for Determining of Colorectal Cancer. Asian Pac J Cancer Prev. 2018;19:2481-2484.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 10]  [Reference Citation Analysis (0)]
36.  Sun X, Ng TTH, Sham KWY, Zhang L, Chan MTV, Wu WKK, Cheng CHK. Bufalin, a Traditional Chinese Medicine Compound, Prevents Tumor Formation in Two Murine Models of Colorectal Cancer. Cancer Prev Res (Phila). 2019;12:653-666.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 25]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
37.  He G, Zhang H, Zhou J, Wang B, Chen Y, Kong Y, Xie X, Wang X, Fei R, Wei L, Chen H, Zeng H. Peritumoural neutrophils negatively regulate adaptive immunity via the PD-L1/PD-1 signalling pathway in hepatocellular carcinoma. J Exp Clin Cancer Res. 2015;34:141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 121]  [Cited by in F6Publishing: 156]  [Article Influence: 17.3]  [Reference Citation Analysis (0)]
38.  Li M, Liu F, Zhang F, Zhou W, Jiang X, Yang Y, Qu K, Wang Y, Ma Q, Wang T, Bai L, Wang Z, Song X, Zhu Y, Yuan R, Gao Y, Liu Y, Jin Y, Li H, Xiang S, Ye Y, Zhang Y, Jiang L, Hu Y, Hao Y, Lu W, Chen S, Gu J, Zhou J, Gong W, Wang X, Liu X, Liu C, Liu H. Genomic ERBB2/ERBB3 mutations promote PD-L1-mediated immune escape in gallbladder cancer: a whole-exome sequencing analysis. Gut. 2019;68:1024-1033.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 91]  [Article Influence: 18.2]  [Reference Citation Analysis (0)]
39.  Hussain SM, Kansal RG, Alvarez MA, Hollingsworth TJ, Elahi A, Miranda-Carboni G, Hendrick LE, Pingili AK, Albritton LM, Dickson PV, Deneve JL, Yakoub D, Hayes DN, Kurosu M, Shibata D, Makowski L, Glazer ES. Role of TGF-β in pancreatic ductal adenocarcinoma progression and PD-L1 expression. Cell Oncol (Dordr). 2021;44:673-687.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 7]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
40.  Sasidharan Nair V, Toor SM, Taha RZ, Shaath H, Elkord E. DNA methylation and repressive histones in the promoters of PD-1, CTLA-4, TIM-3, LAG-3, TIGIT, PD-L1, and galectin-9 genes in human colorectal cancer. Clin Epigenetics. 2018;10:104.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 60]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
41.  Wang Y, Sun J, Ma C, Gao W, Song B, Xue H, Chen W, Chen X, Zhang Y, Shao Q, Wang Q, Zhao L, Liu J, Wang X, Wang H, Yang M, Qu X. Reduced Expression of Galectin-9 Contributes to a Poor Outcome in Colon Cancer by Inhibiting NK Cell Chemotaxis Partially through the Rho/ROCK1 Signaling Pathway. PLoS One. 2016;11:e0152599.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 28]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
42.  Hou N, Ma J, Li W, Zhao L, Gao Q, Mai L. T-cell immunoglobulin and mucin domain-containing protein-3 and galectin-9 protein expression: Potential prognostic significance in esophageal squamous cell carcinoma for Chinese patients. Oncol Lett. 2017;14:8007-8013.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 6]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
43.  Halama N, Braun M, Kahlert C, Spille A, Quack C, Rahbari N, Koch M, Weitz J, Kloor M, Zoernig I, Schirmacher P, Brand K, Grabe N, Falk CS. Natural killer cells are scarce in colorectal carcinoma tissue despite high levels of chemokines and cytokines. Clin Cancer Res. 2011;17:678-689.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 196]  [Cited by in F6Publishing: 216]  [Article Influence: 16.6]  [Reference Citation Analysis (0)]
44.  Xiao Q, Wu J, Wang WJ, Chen S, Zheng Y, Yu X, Meeth K, Sahraei M, Bothwell ALM, Chen L, Bosenberg M, Chen J, Sexl V, Sun L, Li L, Tang W, Wu D. DKK2 imparts tumor immunity evasion through β-catenin-independent suppression of cytotoxic immune-cell activation. Nat Med. 2018;24:262-270.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 93]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
45.  Crouse J, Xu HC, Lang PA, Oxenius A. NK cells regulating T cell responses: mechanisms and outcome. Trends Immunol. 2015;36:49-58.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 137]  [Cited by in F6Publishing: 154]  [Article Influence: 17.1]  [Reference Citation Analysis (0)]
46.  Wu GZ, Pan CX, Jiang D, Zhang Q, Li Y, Zheng SY. Clinicopathological significance of Fas and Fas ligand expressions in esophageal cancer. Am J Cancer Res. 2015;5:2865-2871.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Lu C, Paschall AV, Shi H, Savage N, Waller JL, Sabbatini ME, Oberlies NH, Pearce C, Liu K. The MLL1-H3K4me3 Axis-Mediated PD-L1 Expression and Pancreatic Cancer Immune Evasion. J Natl Cancer Inst. 2017;109.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 157]  [Article Influence: 22.4]  [Reference Citation Analysis (0)]
48.  Karakasheva TA, Waldron TJ, Eruslanov E, Kim SB, Lee JS, O'Brien S, Hicks PD, Basu D, Singhal S, Malavasi F, Rustgi AK. CD38-Expressing Myeloid-Derived Suppressor Cells Promote Tumor Growth in a Murine Model of Esophageal Cancer. Cancer Res. 2015;75:4074-4085.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 93]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
49.  Shitara K, Nishikawa H. Regulatory T cells: a potential target in cancer immunotherapy. Ann N Y Acad Sci. 2018;1417:104-115.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 133]  [Cited by in F6Publishing: 141]  [Article Influence: 23.5]  [Reference Citation Analysis (0)]
50.  Chang AL, Miska J, Wainwright DA, Dey M, Rivetta CV, Yu D, Kanojia D, Pituch KC, Qiao J, Pytel P, Han Y, Wu M, Zhang L, Horbinski CM, Ahmed AU, Lesniak MS. CCL2 Produced by the Glioma Microenvironment Is Essential for the Recruitment of Regulatory T Cells and Myeloid-Derived Suppressor Cells. Cancer Res. 2016;76:5671-5682.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 336]  [Cited by in F6Publishing: 402]  [Article Influence: 50.3]  [Reference Citation Analysis (0)]
51.  Lee CR, Kwak Y, Yang T, Han JH, Park SH, Ye MB, Lee W, Sim KY, Kang JA, Kim YC, Mazmanian SK, Park SG. Myeloid-Derived Suppressor Cells Are Controlled by Regulatory T Cells via TGF-β during Murine Colitis. Cell Rep. 2016;17:3219-3232.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 93]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
52.  Veglia F, Perego M, Gabrilovich D. Myeloid-derived suppressor cells coming of age. Nat Immunol. 2018;19:108-119.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 896]  [Cited by in F6Publishing: 1142]  [Article Influence: 190.3]  [Reference Citation Analysis (0)]
53.  Geiger R, Rieckmann JC, Wolf T, Basso C, Feng Y, Fuhrer T, Kogadeeva M, Picotti P, Meissner F, Mann M, Zamboni N, Sallusto F, Lanzavecchia A. L-Arginine Modulates T Cell Metabolism and Enhances Survival and Anti-tumor Activity. Cell. 2016;167:829-842.e13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 716]  [Cited by in F6Publishing: 935]  [Article Influence: 116.9]  [Reference Citation Analysis (0)]
54.  Li P, Chen X, Qin G, Yue D, Zhang Z, Ping Y, Wang D, Zhao X, Song M, Zhao Q, Li J, Liu S, Zhang C, Lian J, Cao L, Li F, Huang L, Wang L, Yang L, Huang J, Li H, Zhang B, Zhang Y. Maelstrom Directs Myeloid-Derived Suppressor Cells to Promote Esophageal Squamous Cell Carcinoma Progression via Activation of the Akt1/RelA/IL8 Signaling Pathway. Cancer Immunol Res. 2018;6:1246-1259.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 22]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
55.  Moaaz M, Lotfy H, Elsherbini B, Motawea MA, Fadali G. TGF-β Enhances the Anti-inflammatory Effect of Tumor- Infiltrating CD33+11b+HLA-DR Myeloid-Derived Suppressor Cells in Gastric Cancer: A Possible Relation to MicroRNA-494. Asian Pac J Cancer Prev. 2020;21:3393-3403.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 4]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
56.  Ma C, Zhang Q, Greten TF. MDSCs in liver cancer: A critical tumor-promoting player and a potential therapeutic target. Cell Immunol. 2021;361:104295.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 23]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
57.  Hanoteau A, Newton JM, Krupar R, Huang C, Liu HC, Gaspero A, Gartrell RD, Saenger YM, Hart TD, Santegoets SJ, Laoui D, Spanos C, Parikh F, Jayaraman P, Zhang B, Van der Burg SH, Van Ginderachter JA, Melief CJM, Sikora AG. Tumor microenvironment modulation enhances immunologic benefit of chemoradiotherapy. J Immunother Cancer. 2019;7:10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 57]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
58.  Fujimoto H, Saito Y, Ohuchida K, Kawakami E, Fujiki S, Watanabe T, Ono R, Kaneko A, Takagi S, Najima Y, Hijikata A, Cui L, Ueki T, Oda Y, Hori S, Ohara O, Nakamura M, Saito T, Ishikawa F. Deregulated Mucosal Immune Surveillance through Gut-Associated Regulatory T Cells and PD-1+ T Cells in Human Colorectal Cancer. J Immunol. 2018;200:3291-3303.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 22]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
59.  Langhans B, Nischalke HD, Krämer B, Dold L, Lutz P, Mohr R, Vogt A, Toma M, Eis-Hübinger AM, Nattermann J, Strassburg CP, Gonzalez-Carmona MA, Spengler U. Role of regulatory T cells and checkpoint inhibition in hepatocellular carcinoma. Cancer Immunol Immunother. 2019;68:2055-2066.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 89]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
60.  Chen X, Takemoto Y, Deng H, Middelhoff M, Friedman RA, Chu TH, Churchill MJ, Ma Y, Nagar KK, Tailor YH, Mukherjee S, Wang TC. Histidine decarboxylase (HDC)-expressing granulocytic myeloid cells induce and recruit Foxp3+ regulatory T cells in murine colon cancer. Oncoimmunology. 2017;6:e1290034.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 31]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
61.  Jacobs J, Deschoolmeester V, Zwaenepoel K, Flieswasser T, Deben C, Van den Bossche J, Hermans C, Rolfo C, Peeters M, De Wever O, Lardon F, Siozopoulou V, Smits E, Pauwels P. Unveiling a CD70-positive subset of cancer-associated fibroblasts marked by pro-migratory activity and thriving regulatory T cell accumulation. Oncoimmunology. 2018;7:e1440167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 30]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
62.  Hirsch L, Flippot R, Escudier B, Albiges L. Immunomodulatory Roles of VEGF Pathway Inhibitors in Renal Cell Carcinoma. Drugs. 2020;80:1169-1181.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 40]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
63.  OuYang LY, Wu XJ, Ye SB, Zhang RX, Li ZL, Liao W, Pan ZZ, Zheng LM, Zhang XS, Wang Z, Li Q, Ma G, Li J. Tumor-induced myeloid-derived suppressor cells promote tumor progression through oxidative metabolism in human colorectal cancer. J Transl Med. 2015;13:47.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 113]  [Cited by in F6Publishing: 124]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
64.  Kumagai Y, Tachikawa T, Higashi M, Sobajima J, Takahashi A, Amano K, Ishibashi KI, Mochiki E, Yakabi K, Tamaru JI, Ishida H. Chondromodulin-1 and vascular endothelial growth factor-A expression in esophageal squamous cell carcinoma: accelerator and brake theory for angiogenesis at the early stage of cancer progression. Esophagus. 2020;17:159-167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
65.  Baba Y, Nomoto D, Okadome K, Ishimoto T, Iwatsuki M, Miyamoto Y, Yoshida N, Baba H. Tumor immune microenvironment and immune checkpoint inhibitors in esophageal squamous cell carcinoma. Cancer Sci. 2020;111:3132-3141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in F6Publishing: 132]  [Article Influence: 33.0]  [Reference Citation Analysis (0)]
66.  Huang M, Wu R, Chen L, Peng Q, Li S, Zhang Y, Zhou L, Duan L. S100A9 Regulates MDSCs-Mediated Immune Suppression via the RAGE and TLR4 Signaling Pathways in Colorectal Carcinoma. Front Immunol. 2019;10:2243.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 60]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
67.  Tang R, Rangachari M, Kuchroo VK. Tim-3: A co-receptor with diverse roles in T cell exhaustion and tolerance. Semin Immunol. 2019;42:101302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 85]  [Article Influence: 21.3]  [Reference Citation Analysis (0)]
68.  Liu JF, Wu L, Yang LL, Deng WW, Mao L, Wu H, Zhang WF, Sun ZJ. Blockade of TIM3 relieves immunosuppression through reducing regulatory T cells in head and neck cancer. J Exp Clin Cancer Res. 2018;37:44.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 78]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
69.  Ono M. Control of regulatory T-cell differentiation and function by T-cell receptor signalling and Foxp3 transcription factor complexes. Immunology. 2020;160:24-37.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 84]  [Article Influence: 21.0]  [Reference Citation Analysis (0)]
70.  Das M, Zhu C, Kuchroo VK. Tim-3 and its role in regulating anti-tumor immunity. Immunol Rev. 2017;276:97-111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 385]  [Cited by in F6Publishing: 539]  [Article Influence: 77.0]  [Reference Citation Analysis (0)]
71.  Hong MH, Shin SJ, Shin SK, Kim DJ, Zo JI, Shim YM, Lee SE, Cho BC, Park SY, Choi YL, Kim HR. High CD3 and ICOS and low TIM-3 expression predict favourable survival in resected oesophageal squamous cell carcinoma. Sci Rep. 2019;9:20197.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 10]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
72.  Shan B, Man H, Liu J, Wang L, Zhu T, Ma M, Xv Z, Chen X, Yang X, Li P. TIM-3 promotes the metastasis of esophageal squamous cell carcinoma by targeting epithelial-mesenchymal transition via the Akt/GSK-3β/Snail signaling pathway. Oncol Rep. 2016;36:1551-1561.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 40]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
73.  Steinert G, Schölch S, Niemietz T, Iwata N, García SA, Behrens B, Voigt A, Kloor M, Benner A, Bork U, Rahbari NN, Büchler MW, Stoecklein NH, Weitz J, Koch M. Immune escape and survival mechanisms in circulating tumor cells of colorectal cancer. Cancer Res. 2014;74:1694-1704.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 157]  [Cited by in F6Publishing: 166]  [Article Influence: 16.6]  [Reference Citation Analysis (0)]
74.  Abe H, Saito R, Ichimura T, Iwasaki A, Yamazawa S, Shinozaki-Ushiku A, Morikawa T, Ushiku T, Yamashita H, Seto Y, Fukayama M. CD47 expression in Epstein-Barr virus-associated gastric carcinoma: coexistence with tumor immunity lowering the ratio of CD8+/Foxp3+ T cells. Virchows Arch. 2018;472:643-651.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 10]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
75.  Rodríguez MM, Fiore E, Bayo J, Atorrasagasti C, García M, Onorato A, Domínguez L, Malvicini M, Mazzolini G. 4Mu Decreases CD47 Expression on Hepatic Cancer Stem Cells and Primes a Potent Antitumor T Cell Response Induced by Interleukin-12. Mol Ther. 2018;26:2738-2750.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 37]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
76.  Fujiwara-Tani R, Sasaki T, Ohmori H, Luo Y, Goto K, Nishiguchi Y, Mori S, Nakashima C, Mori T, Miyagawa Y, Kawahara I, Fujii K, Kishi S, Tatsumoto N, Kuniyasu H. Concurrent Expression of CD47 and CD44 in Colorectal Cancer Promotes Malignancy. Pathobiology. 2019;86:182-189.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 22]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
77.  Hu T, Liu H, Liang Z, Wang F, Zhou C, Zheng X, Zhang Y, Song Y, Hu J, He X, Xiao J, King RJ, Wu X, Lan P. Tumor-intrinsic CD47 signal regulates glycolysis and promotes colorectal cancer cell growth and metastasis. Theranostics. 2020;10:4056-4072.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 59]  [Article Influence: 14.8]  [Reference Citation Analysis (0)]
78.  Yu GT, Bu LL, Huang CF, Zhang WF, Chen WJ, Gutkind JS, Kulkarni AB, Sun ZJ. PD-1 blockade attenuates immunosuppressive myeloid cells due to inhibition of CD47/SIRPα axis in HPV negative head and neck squamous cell carcinoma. Oncotarget. 2015;6:42067-42080.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 89]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
79.  An CX, Xie SP, Li HL, Hu YH, Niu R, Zhang LJ, Jiang Y, Li Q, Zhou YN. Knockdown of Microtubule Associated Serine/threonine Kinase Like Expression Inhibits Gastric Cancer Cell Growth and Induces Apoptosis by Activation of ERK1/2 and Inactivation of NF-κB Signaling. Curr Med Sci. 2021;41:108-117.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
80.  Zhao H, Xu J, Wang Y, Jiang R, Li X, Zhang L, Che Y. Knockdown of CEACAM19 suppresses human gastric cancer through inhibition of PI3K/Akt and NF-κB. Surg Oncol. 2018;27:495-502.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
81.  Suresh R, Barakat DJ, Barberi T, Zheng L, Jaffee E, Pienta KJ, Friedman AD. NF-κB p50-deficient immature myeloid cell (p50-IMC) adoptive transfer slows the growth of murine prostate and pancreatic ductal carcinoma. J Immunother Cancer. 2020;8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
82.  Lu C, Klement JD, Smith AD, Yang D, Waller JL, Browning DD, Munn DH, Liu K. p50 suppresses cytotoxic T lymphocyte effector function to regulate tumor immune escape and response to immunotherapy. J Immunother Cancer. 2020;8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
83.  Ouyang B, Pan N, Zhang H, Xing C, Ji W. miR146b5p inhibits tumorigenesis and metastasis of gallbladder cancer by targeting Tolllike receptor 4 via the nuclear factorκB pathway. Oncol Rep. 2021;45.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 8]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
84.  He Y, Peng X, Zheng L, Tang Y, Li J, Huang X. Asiaticoside inhibits epithelial-mesenchymal transition and stem cell-like properties of pancreatic cancer PANC-1 cells by blocking the activation of p65 and p38MAPK. J Gastrointest Oncol. 2021;12:196-206.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 10]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
85.  Mortezaee K, Najafi M, Farhood B, Ahmadi A, Shabeeb D, Musa AE. NF-κB targeting for overcoming tumor resistance and normal tissues toxicity. J Cell Physiol. 2019;234:17187-17204.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 59]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
86.  Wang S, Li L, Zhou Y, He Y, Wei Y, Tao A. Heterotypic cell-in-cell structures in colon cancer can be regulated by IL-6 and lead to tumor immune escape. Exp Cell Res. 2019;382:111447.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
87.  Huang HW, Chang CC, Wang CS, Lin KH. Association between Inflammation and Function of Cell Adhesion Molecules Influence on Gastrointestinal Cancer Development. Cells. 2021;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 14]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
88.  Loeser H, Kraemer M, Gebauer F, Bruns C, Schröder W, Zander T, Alakus H, Hoelscher A, Buettner R, Lohneis P, Quaas A. Indoleamine 2,3-Dioxygenase (IDO) Expression Is an Independent Prognostic Marker in Esophageal Adenocarcinoma. J Immunol Res. 2020;2020:2862647.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 7]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
89.  Zhou S, Zhao L, Liang Z, Liu S, Li Y, Yang H, Liu M, Xi M. Indoleamine 2,3-dioxygenase 1 and Programmed Cell Death-ligand 1 Co-expression Predicts Poor Pathologic Response and Recurrence in Esophageal Squamous Cell Carcinoma after Neoadjuvant Chemoradiotherapy. Cancers (Basel). 2019;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 21]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
90.  Mezrich JD, Fechner JH, Zhang X, Johnson BP, Burlingham WJ, Bradfield CA. An interaction between kynurenine and the aryl hydrocarbon receptor can generate regulatory T cells. J Immunol. 2010;185:3190-3198.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 942]  [Cited by in F6Publishing: 1087]  [Article Influence: 77.6]  [Reference Citation Analysis (0)]
91.  Platten M, Nollen EAA, Röhrig UF, Fallarino F, Opitz CA. Tryptophan metabolism as a common therapeutic target in cancer, neurodegeneration and beyond. Nat Rev Drug Discov. 2019;18:379-401.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 445]  [Cited by in F6Publishing: 691]  [Article Influence: 138.2]  [Reference Citation Analysis (0)]
92.  Pflügler S, Svinka J, Scharf I, Crncec I, Filipits M, Charoentong P, Tschurtschenthaler M, Kenner L, Awad M, Stift J, Schernthanner M, Bischl R, Herndler-Brandstetter D, Glitzner E, Moll HP, Casanova E, Timelthaler G, Sibilia M, Gnant M, Lax S, Thaler J, Müller M, Strobl B, Mohr T, Kaser A, Trajanoski Z, Heller G, Eferl R. IDO1+ Paneth cells promote immune escape of colorectal cancer. Commun Biol. 2020;3:252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 18]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
93.  Nosho K, Sukawa Y, Adachi Y, Ito M, Mitsuhashi K, Kurihara H, Kanno S, Yamamoto I, Ishigami K, Igarashi H, Maruyama R, Imai K, Yamamoto H, Shinomura Y. Association of Fusobacterium nucleatum with immunity and molecular alterations in colorectal cancer. World J Gastroenterol. 2016;22:557-566.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 232]  [Cited by in F6Publishing: 238]  [Article Influence: 29.8]  [Reference Citation Analysis (3)]
94.  Servais L, Wéra O, Dibato Epoh J, Delierneux C, Bouznad N, Rahmouni S, Mazzucchelli G, Baiwir D, Delvenne P, Lancellotti P, Oury C. Platelets contribute to the initiation of colitis-associated cancer by promoting immunosuppression. J Thromb Haemost. 2018;16:762-777.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 22]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
95.  Deng J, Li J, Sarde A, Lines JL, Lee YC, Qian DC, Pechenick DA, Manivanh R, Le Mercier I, Lowrey CH, Varn FS, Cheng C, Leib DA, Noelle RJ, Mabaera R. Hypoxia-Induced VISTA Promotes the Suppressive Function of Myeloid-Derived Suppressor Cells in the Tumor Microenvironment. Cancer Immunol Res. 2019;7:1079-1090.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 118]  [Article Influence: 23.6]  [Reference Citation Analysis (0)]
96.  Liu H, Wang J, Tao Y, Li X, Qin J, Bai Z, Chi B, Yan W, Chen X. Curcumol inhibits colorectal cancer proliferation by targeting miR-21 and modulated PTEN/PI3K/Akt pathways. Life Sci. 2019;221:354-361.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 65]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
97.  Tang Y, Jiang M, Jiang HM, Ye ZJ, Huang YS, Li XS, Qin BY, Zhou RS, Pan HF, Zheng DY. The Roles of circRNAs in Liver Cancer Immunity. Front Oncol. 2020;10:598464.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 11]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]