Next Article in Journal
Role of Coactivator Associated Arginine Methyltransferase 1 (CARM1) in the Regulation of the Biological Function of 1,25-Dihydroxyvitamin D3
Next Article in Special Issue
Tissue Inhibitor of Metalloproteinases-1 Overexpression Mediates Chemoresistance in Triple-Negative Breast Cancer Cells
Previous Article in Journal
Characterization of Skeletal Muscle Biopsy and Derived Myoblasts in a Patient Carrying Arg14del Mutation in Phospholamban Gene
Previous Article in Special Issue
Inhibition of Bone Marrow-Mesenchymal Stem Cell-Induced Carbonic Anhydrase IX Potentiates Chemotherapy Efficacy in Triple-Negative Breast Cancer Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Clinical Significance and Role of CXCL1 Chemokine in Gastrointestinal Cancers

by
Jan Korbecki
1,2,
Mateusz Bosiacki
1,3,
Katarzyna Barczak
4,
Ryta Łagocka
4,
Dariusz Chlubek
1 and
Irena Baranowska-Bosiacka
1,*
1
Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
2
Department of Anatomy and Histology, Collegium Medicum, University of Zielona Góra, Zyty 28 St., 65-046 Zielona Góra, Poland
3
Department of Functional Diagnostics and Physical Medicine, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, Żołnierska 54 Str., 71-210 Szczecin, Poland
4
Department of Conservative Dentistry and Endodontics, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
*
Author to whom correspondence should be addressed.
Cells 2023, 12(10), 1406; https://doi.org/10.3390/cells12101406
Submission received: 24 April 2023 / Revised: 9 May 2023 / Accepted: 15 May 2023 / Published: 17 May 2023

Abstract

:
One area of cancer research is the interaction between cancer cells and immune cells, in which chemokines play a vital role. Despite this, a comprehensive summary of the involvement of C-X-C motif ligand 1 (CXCL1) chemokine (also known as growth-regulated gene-α (GRO-α), melanoma growth-stimulatory activity (MGSA)) in cancer processes is lacking. To address this gap, this review provides a detailed analysis of CXCL1’s role in gastrointestinal cancers, including head and neck cancer, esophageal cancer, gastric cancer, liver cancer (hepatocellular carcinoma (HCC)), cholangiocarcinoma, pancreatic cancer (pancreatic ductal adenocarcinoma), and colorectal cancer (colon cancer and rectal cancer). This paper presents the impact of CXCL1 on various molecular cancer processes, such as cancer cell proliferation, migration, and invasion, lymph node metastasis, angiogenesis, recruitment to the tumor microenvironment, and its effect on immune system cells, such as tumor-associated neutrophils (TAN), regulatory T (Treg) cells, myeloid-derived suppressor cells (MDSCs), and macrophages. Furthermore, this review discusses the association of CXCL1 with clinical aspects of gastrointestinal cancers, including its correlation with tumor size, cancer grade, tumor–node–metastasis (TNM) stage, and patient prognosis. This paper concludes by exploring CXCL1’s potential as a therapeutic target in anticancer therapy.

1. Introduction

Gastrointestinal tumors are a diverse group of cancers that affect organs responsible for digestion. These tumors are categorized based on their specific organ location, including head and neck cancer, esophageal cancer, gastric cancer, liver cancer, cholangiocarcinoma, pancreatic cancer, colon cancer, and rectal cancer. In 2021, there were an estimated 5.95 million new cases of these tumors, representing 30.9% of all cancer diagnoses [1]. Additionally, there were 4.06 million deaths caused by gastrointestinal tumors, accounting for 40.9% of all cancer-related deaths [1]. These high mortality rates highlight the need for more effective treatment options, which has led to increased research into potential therapeutic targets.
One promising area of research focuses on intercellular signaling within tumor tissue, specifically the interaction between cancer cells and the immune system [2,3,4,5,6,7]. Cytokines, extracellular signaling molecules that regulate various immune cells, play a critical role in this interaction [8]. Among cytokines, chemokines have chemotactic properties and are divided into four subfamilies based on a conservative motif at the N-terminus [9]. The CXC chemokine subfamily includes 16 representatives in humans, which are divided based on their ability to activate the CXCR receptors [9]. C-X-C motif receptor 2 (CXCR2) ligands, including C-X-C motif ligand 8 (CXCL8, interleukin-8 (IL-8)), are the most frequently studied chemokines, followed by CXCL1.
CXCL1 is a chemokine consisting of 73 amino acids and has a molecular weight of 8 kDa [10]. Its expression is regulated at both the transcription and CXCL1 mRNA stability levels [11,12]. This chemokine activates the CXCR2 receptor at concentrations of several nM [13], making CXCR2 its most significant receptor. At approximately 100-fold higher concentrations, CXCL1 can also activate the CXCR1 receptor [13]. However, the role of CXCR1 in the physiological and pathological functions of CXCL1 appears to be less significant.
Another receptor for CXCL1 is ACKR1 [14], though the importance of this receptor remains unclear. ACKR1 seems to regulate the availability of various chemokines, including CXCL1 [15], and may participate in the transport and distribution of CXCL1 within the intercellular space [16].
Activation of the CXCR2 receptor by CXCL1 triggers signal transduction. Heterotrimeric G proteins, particularly the inhibitory guanine nucleotide regulatory protein (Gαi), are directly activated by CXCR2 [17]. Intracellularly, many proteins bind directly to CXCR2 [18], playing a crucial role in signal transduction, with some signaling pathways operating independently of G proteins.
Activation of CXCR2 by CXCL1 induces cell migration. Among blood cells, neutrophils exhibit the highest expression of CXCR2, making CXCL1 an essential chemoattractant for neutrophils [9,19]. Furthermore, CXCL1 displays mitogenic properties, demonstrated on melanoma cells as one of the chemokine’s first identified properties. As a result, CXCL1 was initially referred to as melanoma growth-stimulatory activity (MGSA) [20].
Considerably less attention has been paid to other CXCR2 ligands in cancer research. As a result, knowledge of the significance of other CXCR2 ligands in cancer processes is incomplete. Moreover, there is a lack of available review articles that describe the significance of certain CXCR2 ligands in cancer processes, including CXCL1. With over 1300 papers available in the PubMed database on this chemokine in the context of cancer (https://pubmed.ncbi.nlm.nih.gov, accessed on 23 March 2023), it is difficult to obtain a clear picture of the current knowledge in this area. To address this gap, we have reviewed the significance of CXCL1 in tumors, with a focus on gastrointestinal tumors due to the sheer volume of available information.

2. Head and Neck Cancer

Head and neck cancer is a group of cancers located most often in the oral cavity, oropharynx, hypopharynx, nasopharynx, nasal cavity, and larynx [21,22]. Annually, approximately 890,000 new cases of head and neck cancer are diagnosed, with approximately 450,000 deaths [22]. The main form of this cancer is squamous cell carcinoma. Risk factors include cigarette smoking, heavy drinking, and oral infection by carcinogenic human papillomavirus (HPV) serotypes [23].
Head and neck squamous cell carcinoma [24,25,26,27], including oral squamous cell carcinoma (OSCC) [28,29,30,31] and larynx squamous cell carcinoma (LSCC) [32,33], show elevated CXCL1 expressions relative to healthy tissue. According to bioinformatics analysis, CXCL1 is considered one of the hub genes with one of the highest number of protein-protein associations in oral cancer [34], particularly in OSCC [35] and oral tongue squamous cell carcinoma (OTSCC) [36]. In patients with head and neck cancer, blood CXCL1 levels may be lower than in healthy individuals [37].
CXCL1 is secreted by cancer cells in head and neck cancer [38,39] including OSCC cells [28,39]. This occurs as a result of interleukin-1β (IL-1β) [40] and vascular endothelial growth factor (VEGF) and vascular endothelial growth factor receptor 1 (VEGFR1) activation on cancer cells [41]. VEGFR1 causes an increase in B-cell leukemia/lymphoma-2 (Bcl-2) expression in cancer cells; this protein activates nuclear factor κB (NF-κB), the transcription factor responsible for the increase in CXCL1 expression, as well as CXCL8 [42]. Other secretory factors that can increase CXCL1 expression in head and neck cancer cells are interleukin-1α (IL-1α) and epidermal growth factor (EGF), as suggested by experiments in mouse models [43]. CXCL1 expression is also increased by elevated Snail2 expression in cancer cells [44]. In addition to cancer cells, cancer-associated fibroblast (CAF) also produces CXCL1 in OSCC [31]. CXCL1 expression in these cells is increased by IL-1α [45] and IL-1β [31]. OSCC cancer cells induce the senescence of fibroblasts [46]; in this state, fibroblasts secrete CXCL1, which, in an autocrine manner, enhances the senescence of fibroblasts and participates in tumorigenic processes in OSCC.
CXCL1 may contribute to the formation of head and neck cancer. The expression of this chemokine is elevated in fibroblasts in oral submucous fibrosis [25], the precancerous lesions from which head and neck cancer arises. CXCL1 promotes tumorigenic processes in oral submucous fibrosis; it causes the proliferation and migration of keratinocytes, as well as increasing the stemness of these cells [25]. These processes lead to the transition of oral submucous fibrosis into head and neck cancer.
CXCL1 is also involved in tumorigenesis in head and neck cancer. CXCL1 increases the proliferation of OSCC cancer cells [40,45,47], which is associated with the activation of CXCR2 that transactivates epidermal growth factor receptor (EGFR) [40].
CXCL1 also causes OSCC cancer cells to migrate [31,46,47]. Specifically, OTSCC cancer cells cause an increase in CXCL1 expression in lymphatic endothelial cells (LEC) [48], which leads to head and neck cancer cells migrating into LEC, and consequently cancer cells migrating into lymphatic vessels and lymph node metastasis. This may explain the correlation of high CXCL1 expression in the tumor with the presence of lymph node metastasis in head and neck cancer [28,32].
CXCL1 increases the expression and secretion of matrix metalloproteinase (MMP)7 and MMP9 by OSCC cells [47], which promotes tumorigenesis.
CXCL1 is involved in angiogenesis in OSCC tumors [28,41]. Following an increase in the expression of CXCL1 in cancer cells [41] and endothelial cells [49] as a result of VEGF, CXCL1 acts on endothelial cells, causing angiogenesis, and on cancer cells, causing their migration toward endothelial cells. This means that CXCL1 is involved in the reciprocal interaction of OSCC cancer cells with endothelial cells. CXCL1 may also be responsible for the recruitment of TAN into the head and neck cancer tumor niche [50].
The high expression of CXCL1 in OSCC [28] and LSCC [32,33] tumors is correlated with lymph node metastasis. Additionally, in LSCC, CXCL1 is associated with the tumor–node–metastasis (TNM) stage and is inversely correlated with tumor histopathological grade [33]. At the same time, CXCL1 expression is not associated with T classification, or LSCC tumor size [32]. In LSCC, CXCL1 is not associated with tumor differentiation [32]. In contrast, in OSCC tumors, CXCL1 is not associated with mode of invasion [28] or tumor differentiation [28].
High levels of CXCL1 in the blood of patients with head and neck cancer are associated with failure of radiation therapy [37]. Higher CXCL1 expression in the tumor is also associated with a worse prognosis of head and neck squamous cell carcinoma (Table 1) [26,27,31,51], including LSCC [33].

3. Esophageal Cancer

More than 604,000 new cases of esophageal cancer are diagnosed annually, which accounts for 3.1% of all diagnosed cancer cases [1]. Each year, there are approximately 544,000 deaths resulting from this cancer, which constitutes 5.5% of all cancer-related deaths [1]. Esophageal cancer can be divided into two subtypes: esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma [53]. ESCC accounts for 70% of all esophageal cancers. Its risk factors include cigarette smoking and high alcohol consumption, while for esophageal adenocarcinoma it is obesity and gastro-oesophageal reflux disease [53]. Frequent damage to the esophagus from gastric contents may lead to Barrett’s esophagus and then to esophageal adenocarcinoma [53].
CXCL1 expression is elevated in ESCC tumors relative to healthy tissue [54,55], along with an elevated level of CXCL1 in the blood compared to healthy individuals [56]. Bioinformatic analyses have identified CXCL1 as one of the hub genes in ESCC [55,57]. A high level of CXCL1 in the blood is associated with a low risk of ESCC [58], which may reflect either antitumor mechanisms involving CXCL1 in the early stages of ESCC development or a protective effect. An elevated expression of CXCL1 has been observed in Barrett’s esophagus and esophageal adenocarcinoma compared to healthy tissue [59].
In ESCC tumors, CXCL1 is secreted by ESCC cells as a result of high NF-κB activation, mediated by several factors, for example, laminin subunit gamma 1 (LAMC1) [60] and early growth response-1 (EGR-1) [61]. By activating the receptor CXCR2, CXCL1 increases the expression of EGR-1, meaning that there is a positive feedback loop between EGR-1 and CXCL1 [61]. CXCL1 expression in ESCC cells is also increased by CAF [56]. It seems that CXCL1 in ESCC tumors comes mainly from CAF [56]. The expression of this chemokine in CAF is also increased by cancer cells.
Tumor-associated macrophages (TAM) are another source of CXCL1 in ESCC tumors [62]. CXCL1 expression is increased upon exposure of macrophages to secretory factors from ESCC cells, which results in the differentiation of these macrophages into TAM [62].
The increase in CXCL1 expression in Barrett’s esophagus and esophageal adenocarcinoma is also due to the frequent amplification of the CXCL1 gene as well as hypomethylation of its promoter [59]. Barrett’s esophagus is accompanied by a decreased expression of glutathione peroxidase 7 (GPX7) [63]. Due to the fact that GPX7 inhibits tumor necrosis factor-receptor 1 (TNFR1) activation when GPX7 expression is downregulated, there is increased tumor necrosis factor-α (TNF-α) activity, resulting in inflammatory responses in the esophagus when exposed to gastric contents. This activates NF-κB and results in the increased expression of proteins dependent on this transcription factor, such as CXCL1. Chronic inflammation leads to esophageal adenocarcinoma.
CXCL1 is involved in tumorigenic processes in ESCC. CXCL1 increases the proliferation of ESCC cells [54]. Through the CXCR2, CXCL1 increases the expression of EGR-1 in ESCC cells [61], a transcription factor that increases their proliferation. This effect of CXCL1 on ESCC cell proliferation is autocrine [54], as ESCC cells secrete CXCL1 that then acts on the same cells.
CXCL1 is also responsible for radio-resistance in ESCC cells [56]. This property of CXCL1 is dependent on extracellular signal-regulated kinase (ERK) mitogen-activated protein kinase (MAPK) activation and decreased superoxide dismutase 1 (SOD-1) expression, which leads to an increase in the level of reactive oxygen species (ROS) in ESCC cells which increases the expression of DNA repair enzymes when cells are exposed to radiation therapy [56].
CXCL1 acts on CAF. CXCL1 causes a change in the phenotype of CAF into inflammatory CAF through activation of the CXCR2-signal transducer and activator of transcription 3 (STAT3) pathway [60]. These altered CAF secrete factors such as IL-1β, interleukin-6 (IL-6), leukemia inhibitory factor (LIF), and granulocyte colony-stimulating factor (G-CSF), which contribute to tumorigenesis. CXCL1 also causes the recruitment of granulocytic-myeloid-derived suppressor cells (G-MDSCs) to the tumor niche in ESCC tumors [64].
CXCL1 expression in ESCC tumors is correlated with tumor size. A larger tumor shows higher CXCL1 expression [56]. Immunohistochemical studies have shown that ESCC tumors larger than 6.5 cm show higher CXCL1 expression than in the sample of ESCC tumors smaller than 6.5 cm. CXCL1 expression is also higher in ESCC stages II–IV than in stage I, showing that the expression of CXCL1 increases with tumor growth and development [57]. At the same time, CXCL1 expression in ESCC is not associated with the depth of invasion or lymph node metastasis [56]. Higher CXCL1 expression in CAF in ESCC tumors may be associated with a poorer prognosis (Table 2) [56], and higher CXCL1 expression in tumors may be associated with a better prognosis for ESCC patients [57]. However, there are also studies that show no association between CXCL1 expression in the tumor and prognosis for esophageal cancer patients [65].

4. Gastric Cancer

It is estimated that nearly 1.06 million new cases of gastric cancer were diagnosed in 2020, which accounted for 5.6% of all cancer diagnoses [1]. Further, in 2020, nearly 770,000 gastric cancer deaths were diagnosed, which accounted for 7.7% of cancer deaths [1]. The main cause of gastric cancer is Helicobacter pylori infection [66,67]. It is estimated that nearly 4.4 billion of the world’s population were infected with this bacterium in 2015 [68]. The largest percentage of the population infected with this bacterium is in Africa and the smallest in Oceania. H. pylori causes chronic inflammation, which leads to gastritis, which after some time develops into intestinal metaplasia, atypical hyperplasia, and, finally, gastric cancer [69]. An essential virulence factor in causing gastric damage by H. pylori is cytotoxin-associated gene A (CagA) [70], a protein that is guided to gastric epithelial cells via the type IV secretion system (TIVSS) [71]. CagA causes chronic inflammation, which is the basis for the development of gastric cancer. However, it is not the only virulence factor in H. pylori. Other virulence factors relevant to the pathogenesis of gastric cancer caused by H. pylori also include outer membrane inflammatory protein A (OipA), duodenal ulcer promoting gene A protein (DupA), and vacuolating cytotoxin A (VacA) [72].
H. pylori infection and the effects of this bacterium cause chronic inflammation in the gastric wall, which leads to increased levels of pro-inflammatory cytokines such as TNF-α, C-C motif ligand 5 (CCL5, regulated on activation, normally T cell expressed and secreted (RANTES)), C-C motif ligand 3 (CCL3, macrophage inflammatory protein 1α (MIP-1α)), C-X-C motif ligand 9 (CXCL9, monokine induced by interferon-γ (MIG)), C-C motif ligand 20 (CCL20, liver and activation-regulated chemokine (LARC)) and CXCR2 ligands such as CXCL8, C-X-C motif ligand 2 (CXCL2, growth-regulated gene-β (GRO-β)) and the described CXCL1 [69,73,74]. H. pylori increases the expression of CXCL1, as well as other CXCR2 ligands, in gastric epithelial cells [71,73,74,75]. The increased expression of CXCR2 ligands is higher in gastric epithelial cells than in gastric cancer cells [76], which shows that CXCL1 may be significant in the early stages of tumor formation. At the same time, the expression level of CXCL1 is correlated with the concentration of H. pylori [73] and decreases following antimicrobial therapy [77].
The mechanism for the increase in CXCL1 expression involves multiple pathways. H. pylori causes the translocation of CagA and fragments of peptidoglycan via TIVSS into gastric epithelial cells [71], which provokes an increase in CXCL1 expression [76]. The bacterium can also be recognized by toll-like receptor 2 (TLR2) independently of the Cag pathogenicity island, which increases the expression of CXCL1 in gastric epithelial cells [75]. EGFR, ERK MAPK, c-Jun N-terminal kinase (JNK) MAPK, and janus tyrosine kinase (JAK)/STAT are also important in this mechanism [75,78]. At the same time, TLR2 activation reduces CXCL1 expression in conventional dendritic cells (cDCs) [79]. Another mechanism for increasing CXCL1 expression is TNF-α inducing protein (Tip-α), secreted by H. pylori. This factor causes increases in the expression of chemokines such as C-C motif ligand 2 (CCL2, monocyte chemoattractant protein 1 (MCP-1)), C-C motif ligand 7 (CCL7, monocyte chemoattractant protein 3 (MCP-3)), CCL20, C-X-C motif ligand 10 (CXCL10, γ interferon inducible protein 10 (IP-10)), and CXCR2 ligands [80], which are associated with the activation of NF-κB by Tip-α. H. pylori may also indirectly increase the expression of CXCL1 and CXCL8. H. pylori activates NF-κB in a CagA-dependent manner which results in the increased expression of interleukin-32 (IL-32) [81] which increases CXCL1 expression. An increase in CXCL1 expression, similar to other CXCR2 ligands, causes neutrophils to infiltrate the gastric foveolar epithelium [82,83]. These cells also secrete CXCL1 and CXCL8 [84]. This means that neutrophils cause the infiltration of more neutrophils in H. pylori gastritis. Neutrophils are involved in inflammatory responses in gastritis; they secrete ROS and reactive nitrogen species (RNS), which damage tissue and cause gene instability [83].
Chronic inflammation leads to gastritis, which after some time develops into gastric intestinal metaplasia, then atypical hyperplasia, and, finally, gastric cancer [69]. The progression of H. pylori infection to gastric cancer is associated with the CXCL1/CXCL8-CXCR2 and p53 axis. H. pylori infection increases the expression of CXCR2, the receptor for CXCL1 [74], which is associated with the activation of nuclear factor-κB subunit 1 (NF-κB1).
H. pylori infection also increases the expression of CXCL1 as well as other ligands for CXCR2 [69,73,74,76,82]. The increased activation of the CXCL1/CXCL8-CXCR2 axis causes cellular senescence of gastric cells, which leads to atrophic mucosa. Importantly, cellular senescence is a process that inhibits proliferation and thus inhibits gastric cancer development; this process creates a positive feedback loop between CXCR2 and p53 [74] where p53 increases CXCR2 expression, while CXCR2 increases p53 activation. With this mechanism, CXCR2 increases proliferation and p53 blocks proliferation. In the next stage of gastric cancer development, H. pylori interfere with p53 function and increase p53 expression in gastric mucosa [85,86,87,88]. A disorder in the function of p53 is associated with H. pylori causing DNA damage [89], which leads to mutations in the TP53 gene [86,90]. This is partly due to the formation of the CagA complex with partitioning-defective 1b (PAR1b)/microtubule affinity-regulating kinase 2 (MARK2), which leads to a decrease in breast cancer type 1 susceptibility protein (BRCA1) function [91], which promotes DNA breakage and reduces DNA repair capabilities. H. pylori also disrupts the proper function of p53 by reducing the expression and function of upstream stimulatory factor 1 (USF1) and USF2, proteins that form a complex with p53 and are important in p53 function [92,93]. H. pylori decreases p53 protein expression by increasing degradation of the protein [87,94,95]—a result of the activation of human double minute 2 (HDM2)/mouse double minute 2 (MDM2) by CagA. The appearance of mutations in the TP53 gene leads to escape from senescence [74] and p53 no longer inhibiting proliferation. Activation of CXCR2 by CXCL1 and CXCL8 stimulates the proliferation of transformed cells, which leads to gastric cancer.
Gastric cancer tumors show a higher expression of CXCL1 and CXCL8 than healthy gastric tissue [96,97,98,99]; the expression of both of these chemokines is highest in diffuse-type gastric carcinoma [100]. CXCL1 levels are positively correlated with patient age [101]. As one of the risk factors for developing gastric cancer is age [66], there may be a link between CXCL1, age, and gastric cancer. In addition, as serum levels of CXCL1 are elevated in gastric cancer patients [102,103], plasma CXCL1 levels may be a marker of gastric cancer.
The elevated expression of CXCL1 in gastric cancer tumors is associated with an increase in the expression of this chemokine in an autocrine manner in gastric cancer cells [104,105]. Another important cause of increased CXCL1 levels is H. pylori infection [82]. There is also downregulation of microRNA (miR)-204 in gastric cancer tumors [106]; a microRNA that directly downregulates CXCL1 expression. Another important, if not the most important, source of CXCL1 in gastric cancer tumors is TAM [107]. CXCL1 in gastric cancer tumors also comes from fibroblasts [108] which increase CXCL1 expression under the influence of extracellular vesicles from gastric cancer cells.
The positive correlation of CXCL1 expression with worse overall survival of gastric cancer patients is related to the fact that CXCL1 participates in tumorigenic processes. CXCL1 increases gastric cancer cell proliferation, as shown by experiments on HGC27 cells [99], AGS, Hs746T [104]. Further, CXCL1 causes cancer cell migration [98,99,105,107,109]. However, the CXCL1/CXCL8-CXCR2 axis may also cooperate with other chemokines in gastric cancer cell migration, particularly C-X-C motif ligand 12 (CXCL12, stromal-derived factor-1 (SDF-1))-CXCR4. Both receptors, CXCR2 and CXCR4, activate NF-κB and STAT3 [107,110]. NF-κB activation increases CXCR4 expression, while STAT3 activation increases CXCR2 expression. Therefore, activation of either of these two receptors results in an increased expression of CXCR2 and CXCR4, which translates into an increase in gastric cancer cell migration and epithelial-to-mesenchymal transition (EMT) induction of these cells [110], an effect also significant in metastasis. The CXCR2-STAT3 pathway may be involved in angiogenesis in gastric cancer [98]; activated STAT3 increases the expression of VEGF [98]—the most important growth factor that causes angiogenesis.
Gastric cancer cells secrete factors that increase CXCL1 expression in LEC [109,111] as a result of NF-κB activation in LEC [111]. CXCL1 activates the focal adhesion kinase (FAK)-ERK MAPK-RhoA pathway in LEC, which leads to the reorganization of F-actin and to the migration and tube formation of LEC. That indicates that CXCL1 in gastric cancer causes lymphangiogenesis.
CXCL1 induces an increase in the expression of MMP2 and MMP9 in gastric cancer [97,109] and causes the migration of LEC, especially to lymphatic vessels [109], which results in lymphatic metastasis. For this reason, CXCL1 levels in gastric cancer tumors are positively correlated with lymphatic metastasis [97,101,109,112]. Serum CXCL1 levels are also positively correlated with lymph node metastasis [102].
CXCL1 acts on tumor-associated cells. CXCL1 causes the recruitment of G-MDSCs [113], bone marrow-derived mesenchymal cells [114], and neutrophils [82] to the tumor niche (Figure 1). Bone marrow-derived mesenchymal cells in the tumor niche are transformed into myofibroblasts. On the other hand, CXCL1 levels are not correlated with the number of TAN in a gastric cancer tumor [100]. For this reason, it is possible that another CXCR2 ligand is responsible for recruiting neutrophils into the tumor niche.
Gastric cancer cells can increase CXCL1 expression in tumor-associated cells. Gastric cancer cells secrete TNF-α, which increases CXCL1 expression in TAM [107]. Further, gastric cancer cells secrete extracellular vesicles that alter the expression of various genes in different cells in the tumor niche, for example extracellular vesicles that contain miR-155, miR-193b and miR-210, inducing the expression of chemokines CXCL1 and CXCL8 in fibroblasts [108]. At the same time, this mechanism does not take place in myofibroblasts.
CXCL1 is closely associated with tumorigenesis in gastric cancer, as CXCL1 expression is positively correlated with advanced TNM stage, i.e., T invasion stage, lymph node metastasis, as well as tumor size [97,98,101,107,109,112,115]. Likewise, serum CXCL1 levels are positively correlated with tumor stage and lymph node metastasis [97,102].
Higher CXCL1 expression in gastric cancer tumors is associated with worse overall survival (Table 3) [97,98,99,101,105,109,112,115], although there is also one study where high CXCL1 expression was associated with a better prognosis for gastric cancer patients [96]. In plasma, higher CXCL1 levels are also associated with poorer cumulative survival [97]. After anticancer treatment, high blood levels of CXCL1 are an indicator of future recurrence [116].

5. Liver Cancer

The most common liver cancer is HCC [117]. An estimated 782,000 new cases of HCC are diagnosed annually, meaning the incidence of this cancer is at 10 cases per 100,000 population per year. There are also 746,000 deaths caused by this cancer each year. Risk factors for HCC include [117]:
  • Chronic hepatitis B virus (HBV) infection;
  • Chronic hepatitis C virus (HCV) infection;
  • Consuming large amounts of alcohol, which leads to alcoholic liver disease (ALD), then to liver cirrhosis, and eventually HCC;
  • Obesity, which leads to non-alcoholic fatty liver disease (NAFLD), then liver cirrhosis, and, eventually, HCC.
Due to the significance of chronic inflammation in the liver, CXCL1 plays an important role in the onset and development of HCC.
CXCL1 is important in the emergence of liver cancer, as demonstrated by studies on genetic polymorphisms in patients with liver cirrhosis and HCC. Specifically, more patients with liver cirrhosis and HCC have the CXCL1 rs4074 A allele [118,119], implying that this allele is associated with a predisposition to these diseases. Individuals with this variant have higher levels of CXCL1 in their blood than those with the rs4074 G variant [119]. Further, the production of CXCL1 in response to HCV proteins is higher in individuals with this variant of the CXCL1 gene [118].
During exposure of the liver to agents leading to liver cirrhosis and HCC, NF-κB is activated and CXCL1 expression increases. In liver cells, there is p50:p50 NF-κB [120], which does not cause an increase in CXCL1 expression, but blocks changes in the expression of this chemokine. This is due to the simultaneous recruitment of p50:p50 NF-κB and histone deacetylase 1 (HDAC1) to the CXCL1 promoter [120]. This mechanism inhibits the increased expression of CXCL1 and also CXCL2, S100 calcium binding protein A8 (S100A8) and S100 calcium binding protein A9 (S100A9), which suppresses the formation of liver steatosis and HCC. Nevertheless, with chronic liver inflammation, this mechanism only delays the development of the disease.
Liver steatosis and HCC arise from chronic inflammation of the liver. This condition can be caused by either chronic HBV or HCV infection. HBV causes an increase in CXCL1 production in the liver. The hepatitis B X (HBx) antigen binds to the TGFB1 gene, which increases the expression of TGF-β1 in hepatocytes [121], a cytokine that activates hepatic stellate cells and more specifically increases the expression of CD147 in these cells [122]. This results in an increase in CXCL1 expression in hepatic stellate cells [123].
Further, chronic HCV infection is associated with increased CXCL1 expression in the liver. During HCV infection, hepatocytes produce CXCL1 [118]. Hepatic stellate cells increase CXCL1 production in HCV-infected hepatocytes [118].
Another cause of liver cirrhosis and HCC is frequent alcohol consumption. Alcohol causes an increase in CXCL1 expression in hepatocytes [124]. Further, CXCL1 expression in the liver of ALD patients is higher than in healthy individuals [125,126]. CXCL1 acts on hepatic stellate cells [123], which causes an increase in α smooth muscle actin (αSMA) and α1(I) collagen expression in these cells, leading to liver fibrosis [123,127].
CXCL1 can also cause liver fibrosis through another pathway. CXCL1 causes infiltration of the liver by neutrophils, which activate hepatic stellate cells via ROS. Then hepatic stellate cells start producing collagen, which leads to hepatic fibrosis [128], which is significant during alcoholic hepatitis as well as other liver diseases. For this reason, CXCL1 expression in the liver is positively correlated with liver fibrosis, for example, in patients with HCV infection [129]. On the other hand, another study showed that CXCL1 levels in the blood are negatively correlated with ISHAK fibrosis score [130], while CXCL8 levels in blood were positively correlated with this parameter.
Liver fibrosis is associated with liver cirrhosis [117]. Liver cirrhosis is a risk factor for HCC. This cancer, similar to liver cirrhosis, is characterized by inflammatory responses in which CXCL1 plays a significant role.
CXCL1 expression is higher in HCC tumors than in healthy liver tissue [131], and is higher in HCC than in liver cirrhosis [132]. In patients with liver cancer, blood CXCL1 levels are elevated compared to healthy individuals [132] and those with liver cirrhosis [132]. Blood CXCL1 level is also elevated in hepatitis B-related HCC, and for this reason may be used as a biomarker of this cancer [133].
A significant source of CXCL1 in HCC is myofibroblast-like cells [134], which also produce other secretory factors such as CXCL8 and IL-6 [134]. CXCL1 expression occurs in cancer cells [135,136], and CXCL1 and CXCL8 expression occurs in HCC cancer stem cells [137]. The expression of both chemokines is upregulated by neurotensin (NTS) through ERK MAPK activation, with it being CXCL8 that acts in an autocrine manner on cancer stem cells [137], increasing their proliferation. CXCL1 does not act on HCC cancer stem cells, only on other cells in the tumor niche.
In HCC tumors, many factors increase CXCL1 expression in cancer cells. In particular, CXCL1 itself is responsible for increases in NF-κB activation and thus its own expression in an autocrine manner [138]. Other factors that increase CXCL1 expression include chronic hypoxia [135], EGFR activation [139], interleukin-17 (IL-17) [140], saturated fatty acid (SFA) including palmitate [141], tumorigenesis-related downregulation of both solute carrier family 7 member 2 (SLC7A2) [142], and receptor-interacting protein kinase-3 (RIPK3) [136]. CXCL1 expression in HCC also depends on microRNA. It has been shown that miR-200a [143], a microRNA that directly downregulates CXCL1 expression, is downregulated in this tumor [143], and thus increases CXCL1 expression in HCC tumors.
Studies in mice have indicated that the elevated expression of CXCR2 ligands in the liver facilitates HCC formation in this organ [144]. In mice, the expression of CXCR2 ligands in the liver is highest a few days after birth. This indicates that CXCR2 ligands may create a favorable microenvironment for the development of pediatric liver cancer, particularly hepatoblastoma [144]. However, these results need to be confirmed on a human model.
CXCL1 increases the proliferation of HCC cells [131,138,145]. Studies on mice indicate that CXCL1 can affect HCC cancer stem cells, as CXCR2 ligands cause quiescence of these cells. These chemokines induce mechanistic target of rapamycin complex 1 (mTORC1) activation, which inhibits the differentiation of HCC cancer stem cells [146].
Further, mouse CXCR2 ligands increase the number of HCC cancer stem cells—cells that are resistant to chemotherapeutics, such as doxorubicin [146]. Therefore, CXCR2 ligands increase the resistance of HCC to treatment; nevertheless, studies need to be repeated in human models to see whether a human CXCR2 ligand is responsible for this process.
CXCL1 is also important in HCC tumor growth [138]. CXCL1 induces migration and EMT of HCC cells [131,143]. This property of CXCL1 is dependent on NF-κB activation [131]. CXCL1 also induces angiogenesis in this cancer [140].
CXCL1 also acts on tumor-associated cells. This chemokine induces the recruitment of neutrophils [147] and G-MDSCs [148] into the HCC tumor niche. This action of CXCL1 should be understood as part of a broader meshwork of tumor-associated mechanisms. CXCL1 induces the recruitment of neutrophils into the HCC tumor niche [147]; these cells secrete CCL2 and C-C motif ligand 17 (CCL17, thymus and activation-regulated chemokine (TARC)), which induce the recruitment of monocytes and Treg, respectively [149]. Monocytes differentiate into TAM, which secrete pro-angiogenic VEGF [150], while Treg participate in cancer immunoevasion [6].
The great importance of CXCL1 in tumorigenesis is confirmed by the correlation of CXCL1 expression with various clinical parameters of the cancer in question. The level of CXCL1 expression increases with HCC tumor growth. There is a positive correlation of CXCL1 expression in the tumor with TNM classification stages, macrovascular invasion, microvascular invasion, and distant metastasis [131,143]. Higher CXCL1 expression in the tumor is associated with a worse prognosis for liver cancer patients [151], including HCC (Table 4) [131,143,147,152].

6. Cholangiocarcinoma

Cholangiocarcinoma is a tumor located in the biliary tree [153]. The incidence of this type of cancer varies by world region. For the US, it is at 0.72 to 1.67 per 100,000 population. In contrast, the highest incidence is in Northeast Thailand, with 80 cases per 100,000 population [153]. Cholangiocarcinoma can be divided by tumor location into intrahepatic cholangiocarcinoma and extrahepatic cholangiocarcinoma. Factors that increase the likelihood of cholangiocarcinoma include sclerosing cholangitis, liver cirrhosis, Opisthorchis viverrini infection, HBV or HCV infection, and heavy alcohol consumption [153,154]. Studies of patients with cholangiocarcinoma have shown that this is one of the few cancers in which CXCL1 may have anticancer properties.
CXCL1 inhibits the proliferation of OCUG-1 and HuCCT1 cholangiocarcinoma cells [155] and does not affect the proliferation of KMBC cells [156]. CXCL1 also inhibits the migration of cholangiocarcinoma cancer cells [155]. The source of CXCL1 in cholangiocarcinoma tumors may be mesenchymal stem cells (MSC), as cancer cells secrete extracellular vesicles that act on MSC, causing their fibroblastic differentiation and increasing the expression of secretory factors such as CXCL1, CCL2, and IL-6 [156].
CXCL1 may also have pro-cancer properties in cholangiocarcinoma. Liver CXCR2 ligands regulate cancer immune evasion in cholangiocarcinoma, which originates in the gut. Disruption in gut barrier function leads to the passage of commensal gut bacteria into the liver, which activates toll-like receptor 4 (TLR4) on cells in that organ. This leads to an increase in the expression of CXCR2 ligands in the liver, as shown by experiments in mice [157]. Chronic inflammation, in particular chronic upregulation of CXCR2 ligand expression in the liver, leads to the accumulation of MDSCs in this organ [157]. As cells have immunosuppressive properties, and therefore this leads to cancer immune evasion in cholangiocarcinoma and promotes the development of this cancer. Nevertheless, this has only been shown in mouse experiments and therefore we need to study this cancer mechanism on human models to show which human CXCR2 ligand is responsible for this process.
CXCL1 expression does not increase with tumor stage [155]. The level of CXCL1 expression in the tumor is negatively correlated with distant metastasis, and higher CXCL1 expression in the tumor is a better [155] or worse prognosis for patients [65]. Higher CXCR2 expression is also associated with a better prognosis for patients with cholangiocarcinoma (Table 5) [155]. On the other hand, one study indicates that increased CXCR2 expression in the tumor is associated with a worse prognosis for patients with intrahepatic cholangiocellular carcinoma [158].

7. Pancreatic Cancer

In 2020 alone, more than 495,000 new cases of pancreatic cancer were diagnosed, accounting for 2.6% of all cancers [1]. This cancer has a high mortality rate, with only 4% of patients surviving 5 years after diagnosis [159]. In 2020, there were 466,000 such deaths, which accounted for 4.7% of deaths caused by all cancers [1]. In pancreatic cancer, mutations occur during tumorigenesis, in particular in the Kirsten rat sarcoma viral oncogene homologue (KRAS) oncogene, increasing the activity of this protein [159,160,161,162]. Mutations also occurred in the CDKN2A, TP53, SMAD4, and BRCA2 genes [159,162]. This leads to the formation of a tumor.
In pancreatic cancer tumors [163], particularly in pancreatic ductal adenocarcinoma, there may be an upregulation of CXCL1 expression relative to healthy tissue [164,165,166]. However, other studies have shown that in humans, CXCL1 expression in pancreatic ductal adenocarcinoma tumors is not different relative to healthy tissue [167].
CXCL1 expression is found in pancreatic cancer cells [168,169]. Mutation in the KRAS gene, common in pancreatic cancer [160,161,162], leads to an increase in CXCL1 expression as well as other CXCR2 ligands [170,171]. Other factors cause an increase in CXCL1 expression in pancreatic tumor cells. In particular, an increase in CXCL1 expression may be induced by the elevated expression of p63 [169], a protein belonging to the p53 transcription factor family, with similar properties to p53 [172]. p63 binds to the promoter of the CXCL1 gene which increases the expression of CXCL1. In a cancer cell, CXCL1 expression may also be due to the interaction of this cell with other non-cancerous cells in the tumor niche. CXCL1 expression in cancer cells can be increased by IL-17 [173] and also apolipoprotein E (ApoE) secreted by inflammatory CAF into the tumor microenvironment [174]. ApoE activates the low-density lipoprotein receptor (LDLR) on the tumor cell, which leads to an increase in NF-κB activation and an increase in CXCL1 expression in that cell [174].
CXCL1 expression in cancer cells may also depend on pancreatic stellate cells found in a healthy pancreas that are recruited to the cancer niche during pancreatic cancer tumor development. These cells are an essential component of the cancer stroma in pancreatic cancer and play an important role in its tumorigenesis [175]. Pancreatic stellate cells secrete exosomes that increase CXCL1 expression in pancreatic cancer cells [176]. Increased CXCL1 expression in pancreatic cancer tumors may also be caused by a disruption in the function of metaplastic tuft cells, which are solitary chemosensory cells [177].
Another source of CXCL1 in pancreatic tumors is inflammatory CAF [178,179]; in pancreatic cancer, these are stromal fibroblasts [167]. CAF in pancreatic tumors originate mainly from resident pancreatic stellate cells [180]. CAF can be divided into two types based on their phenotype: inflammatory CAF and myofibroblastic CAF. If CAF come into direct contact with cancer cells, they are transformed into myofibroblastic CAF [179]. Polarization of CAF to inflammatory CAF occurs in cells that do not come into direct contact with pancreatic cancer cells [179]. Under the influence of secretory factors from cancer cells, the expression of sequestosome-1 (sqstm1) in CAF is reduced [180], which leads to increased levels of ROS in CAF cells and results in their senescence and the acquisition of a senescence-associated secretory phenotype (SASP) by these cells [180,181,182]. The factor responsible for this process may be IL-1α secreted by cancer cells [169]. IL-1α expression in the cancer cell is dependent on p63. IL-1α enhances inflammatory CAF and downregulates the myofibroblastic CAF phenotype. Subsequently, inflammatory CAF secrete factors such as CXCL1 that help maintain their phenotype [183].
CXCL1 expression in the tumor may also be increased by recruited monocytes [184]. In particular, macrophages produce significant amounts of CXCL1 at the metastatic site of pancreatic ductal adenocarcinoma [185]. Monocytes produce CXCL1 and CXCL8 under the influence of IL-35, which is associated with activation by IL-35 of the GP130:IL12RB2 heterodimer, which activates the pSTAT1:pSTAT4 heterodimer. The pSTAT1:pSTAT4 complex attaches to the promoters of the aforementioned chemokines, thus increasing their expression.
CXCL1 is significant in the early stages of pancreatic cancer transformation. Very often mutations in the KRAS gene occur at the beginning of tumorigenesis [171], followed by oncogene-induced senescence [186] which involves the activation of NF-κB which increases the expression of CXCR2 ligands such as CXCL1 [171,186]. This means that such a cell is in a senescent state and exhibits the senescence-associated secretory phenotype (SASP) [181,182]. Increased secretion of CXCR2 ligands results in the activation of this receptor. At an early stage of tumorigenesis, this induces inhibition of the proliferation of such an altered pre-cancerous cell and prevents tumor development [186]. Further, the activation of NF-κB leads to the expression of factors that have pro-inflammatory effects, including an increase in the number of antitumor macrophages with M1 polarity. In the early stages of tumorigenesis, p53 and p16/retinoblastoma (Rb) pathways are responsible for inhibiting proliferation and further development of pancreatic cancer. When mutations occur in the TP53 gene, a senescence bypass occurs [186], an event common during tumorigenesis in pancreatic cancer as approximately 76% of pancreatic tumors show a mutation in TP53 [162]. Then, activation of CXCR2 by CXCL1 causes the proliferation of the transformed cell and thus further development of pancreatic cancer [171].
In advanced pancreatic ductal adenocarcinoma tumors, CXCL1 also has pro-tumorigenic properties. Other CXCR2 ligands may also either be responsible or co-responsible for the properties of CXCL1. In particular, this refers to C-X-C motif ligand 5 (CXCL5, epithelial neutrophil-activating protein 78 (ENA78)) and CXCL8, which expression in pancreatic ductal adenocarcinoma tumors are the highest among CXCR2 ligands [166]. The low importance of CXCL1 in the development of pancreatic cancer tumors has been shown in animal experiments [167] and it has been suggested that this chemokine is more important in pancreatic cancer metastasis [167].
This effect of CXCL1 on pancreatic cancer cells can be negated if they have a high expression of atypical chemokine receptor 1 (ACKR1, Duffy antigen receptor for chemokines (DARC)) [187], an atypical receptor for various chemokines including CXCL1 [188] which inhibits the activity of CXCR2 and thus the action of CXCL1. Through activation of the receptor CXCR2, CXCL1 increases pancreatic ductal adenocarcinoma proliferation [171]. This effect may still be weak as in some studies CXCR2 did not affect the proliferation of pancreatic ductal adenocarcinoma cells [189] and was only weakly involved in the tumor growth of this cancer [167].
However, CXCL1 may be important in other cancer processes. CXCL1, together with other CXCR2 ligands, causes angiogenesis [170,173,190], and it may also be important in the metastasis of pancreatic cancer. In addition, this chemokine is important in cancer cell survival in lung metastasis [173]. One study shows that CXCR2 gene knockout in mice leads to increased liver metastasis arising from pancreatic ductal adenocarcinoma [190]. As another available study shows opposite results [167], CXCL1 may have a role in metastasis only to certain organs and in certain models.
CXCL1 induces the recruitment of neutrophils into the tumor niche of pancreatic ductal adenocarcinoma [164,169,185] and G-MDSCs [177] but not monocytes and macrophages [167]. CXCL1 enhances the polarization of M2 macrophages [191,192,193,194] which inhibit the antitumor immune response more strongly than TAM not exposed to CXCL1. At the same time, neutrophils recruitment to pancreatic ductal adenocarcinoma is in balance with macrophage recruitment. When TAM recruitment is reduced by decreasing CCL2 expression in the tumor niche, there is an increase in CXCR2 ligand expression, which leads to increased neutrophils recruitment [164].
Further, CXCL1 can act on CAF where it enhances inflammatory CAF, decreases myofibroblastic-CAF phenotype [183], and lowers the expression of αSMA and production of ECM proteins. Inflammatory CAF also secrete increased amounts of various secretory factors, such as CXCR2 ligands [183]. This is associated with NF-κB activation in these cells. CXCL1 also causes an increase in connective tissue growth factor (CTGF) expression and secretion from CAF [195]. This growth factor is involved in tumorigenesis [196], causing proliferation and invasiveness of cancer cells, and participates in angiogenesis.
CXCL1 is also involved in chemotherapy resistance in pancreatic cancer patients. Some anticancer drugs, such as gemcitabine, increase the expression of CXCL1 and CXCL2 in pancreatic ductal adenocarcinoma tumor cells [185,192]. This has important implications for the recurrence of this cancer following chemotherapy with gemcitabine, where an increase is shown in the expression of CXCR2 ligands, including CXCL1, in pancreatic ductal adenocarcinoma [192] and in the metastatic site of this tumor [185]. This is associated with the increased expression of RIPK1 and RIPK3, the components of necroptosis [192]. CXCL1 increases M2 polarization of macrophages [191,192,193,194], which have immunosuppressive properties and inhibit the antitumor immune system response augmented by chemotherapy. Increased CXCL1 expression also recruits neutrophils to the metastasis site; these cells secrete growth arrest specific 6 (Gas6) [185], a secretory factor activating the Axl receptor tyrosine kinase that causes tumor cell proliferation and thus the regrowth of the metastatic site following chemotherapy.
CXCL1 expression in tumors increases with the tumor stage [165]. CXCL1 expression level in tumor cells is positively correlated with N stage, T stage, and distant metastasis, but not with perineural invasion [163]. CXCL1 expression in tumor stroma is positively correlated with perineural invasion, N stage, and T stage [163]. The high importance of CXCL1 in tumor processes is reflected in the impact of this chemokine on patient prognosis. Higher CXCL1 expression in a tumor means a worse prognosis for patients (Table 6) [163]. However, there are also studies that show no association between CXCL1 expression in the tumor and prognosis for pancreatic cancer patients [65,165].

8. Colorectal Cancer

Colorectal cancer is one of the three most commonly diagnosed cancers. An estimated 1.88 million new cases of this cancer were diagnosed in 2020 alone, which accounted for 9.8% of all cancers diagnosed [1]. Further, there were nearly 916,000 deaths caused by this cancer in 2020, which accounted for 9.2% of deaths caused by all cancers. Risk factors for this type of cancer include genetic factors and diet [197]. Genetic predisposition accounts for 10% to 20% of colorectal cancer cases [197]. Further, infection with certain intestinal bacteria may be associated with an increased likelihood of developing colorectal cancer [198]. As this cancer is associated with inflammation, one of the components of its pathogenesis is CXCL1.
There is elevated CXCL1 expression in colorectal cancer tumors compared to healthy tissue [199,200,201,202,203,204,205,206,207,208,209,210]. In addition, CXCL1 may also be a marker of early-stage colorectal cancer [211]. However, serum CXCL1 levels are reduced in patients with colorectal cancer [212]. In contrast, serum CXCL8 levels are increased in the same patients [212]. Some studies show that CXCL1 expression is higher in colon cancer tumors of elderly patients older than 65 years compared to patients younger than 65 years [205,213], and at least one study shows an inverse relationship [214].
In colorectal cancer, CXCL1 is produced by a variety of cells [215,216,217]. In the tumor niche, myofibroblasts [218], tumor-associated dendritic cells (TADC) [219], and TAM [220] may also be responsible for CXCL1 production, with various factors responsible for CXCL1 expression in these cells. In approximately 35% of colorectal cancer tumor cases, SMAD family member 4 (SMAD4) expression is reduced [212]. As SMAD4 reduces CXCL1 expression by decreasing inhibitor of NF-κB kinase β (IKKβ) activity and affecting glycogen synthase kinase-3β (GSK-3β), a decrease in SMAD4 expression in a tumor cell increases CXCL1 expression. An elevated CXCL1 expression in a colorectal cancer tumor may also be due to hypoxia-inducible factor-2 (HIF-2) which has been shown to be responsible for the expression of CXCR2 ligands in colorectal tumors in a murine model [221].
Other factors increasing CXCL1 expression include IL-22, secreted by CD4+ and CD8+ polyfunctional T cells [222], and cell migration inducing hyaluronidase 1 (CEMIP, KIAA1199), a protein that acts on hyaluronic acid and activates transforming growth factor-β receptor 1 and 2 (TGFBR1/2) which stimulates CXCL1 expression [223]. CXCL1 expression in colorectal cancer cells is also increased by prostaglandin E2 (PGE2) [215,216] in a process that depends on the EGFR-MAPK pathway. Further, mutation in Ras proteins can cause an increase in the activity of these proteins and lead to an increase in CXCL1 expression in colorectal cancer cells [217,218]. Elevated CXCL1 expression in colorectal cancer cells may also be the result of high basal NF-κB activation [224] associated with the overexpression of UEV1A, an enzyme that catalyzes poly-ubiquitination, which results in the activation of NF-κB. This transcription factor directly attaches to the CXCL1 promoter and then increases the expression of this chemokine.
Increased CXCL1 expression in colorectal cancer cells may also be associated with adiponectin [225,226]. CXCL12-CXCR4 axis [227], and microRNAs, e.g., miR-302e in colorectal cancer [208], miR-145-5p in colorectal cancer [228], and miR-1-3p in rectal adenocarcinoma [229], whose expression is decreased in colorectal cancer tumors. These microRNAs directly reduce CXCL1 expression. That is, the downregulation of the expression of these microRNAs is followed by an increase in CXCL1 expression.
CXCL1 in colorectal cancer tumors participates in tumorigenic processes. Due to links in tumor mechanisms with other proteins, CXCL1 is considered a hub gene in colorectal cancer [229,230]. CXCL1 increases cancer cell proliferation [104,201,208,231,232] and increases glycolytic enzyme expressions, such as glucose transporter 1 (GLUT1), hexokinase 2 (HK2), and lactate dehydrogenase A (LDHA) [233]. In addition, in colorectal cancer cells, and in fibroblasts it decreases the expression of fibulin-1, a matrix protein that characterizes normal intestinal tissue [201]—the reduced expression allows colorectal cancer to develop.
CXCL1 also causes the migration and invasion of colorectal cancer cells [208,219,231,232], which is associated with an increase in MMP7 expression, and CXCL1 causing EMT of cancer cells [219]. For this reason, CXCL1 is associated with so-called “tumor budding” [234]—the appearance of either single cancer cells or small clusters of cancer cells at the invasive front of tumors [235].
CXCL1 may also facilitate the formation of metastasis by inducing an increase in parathyroid hormone-like hormone (PTHLH) expression in colorectal cancer cells [219]—a factor that causes bone remodeling during the formation of bone metastasis. CXCL1 is also carried with blood from colorectal cancer cells to the liver where it causes the infiltration of MDSCs to this organ by [220]. These cells form a pre-metastatic niche in the liver which facilitates the metastasis to this organ. High levels of CXCL1 in the blood are correlated with levels of circulating tumor cells [236], which indicates that CXCL1 is associated with a high likelihood of colorectal cancer metastasis.
CXCL1 also affects cells in the tumor niche in colorectal cancer. CXCL1 causes the recruitment of neutrophils [50,222,223] and G-MDSCs [216,237] into the tumor niche. The latter are cells with immunosuppressive properties that inhibit the antitumor immune response; in particular, they reduce the effect of cytotoxic CD8+ T cells. In addition, CXCL1 in colorectal cancer tumors causes angiogenesis [215] and stromal fibroblast senescence [226] which alters the secretory phenotype of these cells and thus enhances cancer tumor growth.
The level of colorectal tumor development cannot be definitely linked to the level of CXCL1 expression in the tumor, as some studies show a positive correlation and some a negative correlation between CXCL1 levels and colorectal tumor grade. Some studies show that CXCL1 expression in colorectal tumors is positively correlated with lymph node metastasis, tumor size, and tumor stage [233,238,239]. A higher level of CXCL1 in the blood of a colorectal cancer patient is associated with a higher TNM stage and may be an indicator of lung metastasis [236,240]. However, there are also studies where the elevated CXCL1 expression in a colorectal tumor is inversely correlated with lymph node metastasis [214,218] or tumor stage [202,207]. Some studies on colon cancer have shown that the higher the tumor stage is, the lower the level of CXCL1 in the tumor [205,209,213]. Finally, CXCL1 expression in liver metastasis is lower than in the primary colorectal tumor [202]. The cited results demonstrate that different research groups report negative and positive correlations of CXCL1 expression in the tumor with tumor stage, i.e., contradictory results. The results of studies on the relationship between CXCL1 levels in the tumor and prognosis for the colorectal cancer patient are varied. Some show that a higher CXCL1 expression in the tumor is associated with a worse prognosis for the patient [233,238]. Further, the prognosis is worse for rectal adenocarcinoma patients with a high CXCL1 expression in the tumor [229]. Other studies have shown that CXCL1 is associated with a worse prognosis only in patients with stage III colorectal cancer [204] or only in stage IV [218]. Still, other studies have found no association of CXCL1 expression in colorectal cancer tumors [206,239] and in blood [212] with patient prognosis (Table 7). Finally, there are also articles that indicate that high CXCL1 expression in the tumor is associated with a better prognosis for the colon cancer patient [209,213,241].

9. CXCL1 as a Therapeutic Target in Anticancer Therapy of Gastrointestinal Tumors

As described above, CXCL1 plays a significant role in the molecular processes of gastrointestinal tumors. In theory, it is possible to develop anticancer therapy targeting CXCL1. However, the most important receptor for CXCL1 is CXCR2 [9], which is also activated by other CXC chemokines, including CXCL2, CXCL3, CXCL5, CXCL6, CXCL7, and CXCL8. Therefore, a better therapeutic approach would be to target CXCR2 instead of CXCL1, which would block the effects of not only CXCL1 but also the other CXC chemokines listed above.
The most well-known and commonly tested CXCR2 antagonist as an antitumor agent is SB225002 (N-(2-hydroxy-4-nitrophenyl)-N’-(2-bromophenyl)urea) [242]. This compound has demonstrated antitumor activity, reducing the viability of SCC158 and HN30 oral squamous cell carcinoma cell lines [243]. It has also been shown to have no toxicity to the immortalized keratinocyte lineage HaCaT. In addition, SB225002 reduced the proliferation and migration of RBE and SSP25 intrahepatic cholangiocellular carcinoma cell lines in vitro [158]. Moreover, SB225002 inhibited the growth of cholangiocellular carcinoma tumors in mice [158]. However, it should be noted that SB225002 is not only a CXCR2 inhibitor but also binds to β-tubulin [244,245], leading to destabilization of microtubules and antimitotic activity of SB225002.
Additionally being tested are other CXCR2 antagonists that also inhibit CXCR1 activity. CXCL1 only activates CXCR2 at low concentrations, while CXCL8 activates both CXCR1 and CXCR2 at low concentrations [246]. Therefore, the use of dual CXCR1/CXCR2 antagonists allows for the inhibition not only of CXCL1 and other CXCR2 ligands but also of CXCL8. SCH-527123 is one such compound being tested [247,248]. SCH-527123 inhibits the proliferation and migration of colorectal cancer cell lines HCT116 and Caco2 in vitro [249]. SCH-527123 also sensitizes these cells to anticancer drugs, as demonstrated by experiments involving oxaliplatin [249]. In vivo studies in mice have confirmed that SCH-527123 exhibits anticancer activity and sensitizes colorectal cancer cells to oxaliplatin [249].
CXCR2 antagonists are currently under investigation in clinical trials as potential therapeutic agents (Table 8). On the ClinicalTrials.gov website (https://clinicaltrials.gov/ct2/home, accessed on 5 May 2023), a search using the receptor name “CXCR2” revealed 29 distinct clinical trials involving CXCR2 antagonists. Additional search results appear when entering the names of specific drugs.
Typically, a particular CXCR2 antagonist is examined for its efficacy in treating a specific group of diseases. For instance, SX-682 is being studied as a potential anticancer agent, while Ladarixin is being explored as a treatment for type 1 diabetes. Danirixin (GSK1325756), on the other hand, is being tested for lung diseases such as COPD and influenza. AZD5069 is under investigation as both an anticancer drug and a treatment for lung diseases such as COPD and asthma. Meanwhile, SB656933 is being examined as a potential therapeutic option for COPD and cystic fibrosis.

10. Future Perspectives

Among CXCR2 ligands, research predominantly focuses not on CXCL1 but on another CXCR2 ligand, CXCL8. As of 5 May 2023, the PubMed database (https://pubmed.ncbi.nlm.nih.gov) contained over 6-fold more scientific publications discussing CXCL8 in the context of cancer compared to CXCL1. This testifies to significant knowledge gaps remain regarding the role of CXCL1 in the oncogenic mechanisms of gastrointestinal neoplasms. Future research should also explore the interrelationships among individual CXCR2 ligands in tumorigenic processes. In humans, there are seven distinct CXCR2 ligands, which exhibit similar functions; however, differences among them warrant further investigation.

11. Conclusions

The role of CXCL1 in gastrointestinal malignancies has been extensively investigated and numerous studies have assessed the clinical implications of this chemokine in these tumors. Additionally, research has demonstrated the involvement of CXCL1 in a multitude of molecular mechanisms associated with these neoplasms. Presently, clinical trials exploring anticancer therapies incorporating CXCR2 antagonists are in preliminary phases. In the forthcoming years, these therapeutic agents are expected to become included in widely available cancer treatment.

Author Contributions

Conceptualization, J.K. and I.B.-B.; investigation, M.B., K.B. and R.Ł.; writing—original draft preparation, J.K. and I.B.-B.; writing—review and editing, I.B.-B. and J.K.; supervision and funding acquisition, D.C. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the statutory budget of the Department of Biochemistry and Medical Chemistry Pomeranian Medical University in Szczecin, Poland.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Sabry, M.; Lowdell, M.W. Tumor-primed NK cells: Waiting for the green light. Front. Immunol. 2013, 4, 408. [Google Scholar] [CrossRef] [PubMed]
  3. Coffelt, S.B.; Wellenstein, M.D.; de Visser, K.E. Neutrophils in cancer: Neutral no more. Nat. Rev. Cancer 2016, 16, 431–446. [Google Scholar] [CrossRef]
  4. Kumar, V.; Patel, S.; Tcyganov, E.; Gabrilovich, D.I. The Nature of Myeloid-Derived Suppressor Cells in the Tumor Microenvironment. Trends Immunol. 2016, 37, 208–220. [Google Scholar] [CrossRef] [PubMed]
  5. Hansen, M.; Andersen, M.H. The role of dendritic cells in cancer. Semin. Immunopathol. 2017, 39, 307–316. [Google Scholar] [CrossRef]
  6. Najafi, M.; Farhood, B.; Mortezaee, K. Contribution of regulatory T cells to cancer: A review. J. Cell Physiol. 2019, 234, 7983–7993. [Google Scholar] [CrossRef] [PubMed]
  7. Khalaf, K.; Hana, D.; Chou, J.T.; Singh, C.; Mackiewicz, A.; Kaczmarek, M. Aspects of the Tumor Microenvironment Involved in Immune Resistance and Drug Resistance. Front. Immunol. 2021, 12, 656364. [Google Scholar] [CrossRef]
  8. Dinarello, C.A. Historical insights into cytokines. Eur. J. Immunol. 2007, 37 (Suppl. S1), S34–S45. [Google Scholar] [CrossRef]
  9. Hughes, C.E.; Nibbs, R.J.B. A guide to chemokines and their receptors. FEBS J. 2018, 285, 2944–2971. [Google Scholar] [CrossRef]
  10. Balentien, E.; Han, J.H.; Thomas, H.G.; Wen, D.Z.; Samantha, A.K.; Zachariae, C.O.; Griffin, P.R.; Brachmann, R.; Wong, W.L.; Matsushima, K.; et al. Recombinant expression, biochemical characterization, and biological activities of the human MGSA/gro protein. Biochemistry 1990, 29, 10225–10233. [Google Scholar]
  11. Shattuck, R.L.; Wood, L.D.; Jaffe, G.J.; Richmond, A. MGSA/GRO transcription is differentially regulated in normal retinal pigment epithelial and melanoma cells. Mol. Cell Biol. 1994, 14, 791–802. [Google Scholar] [PubMed]
  12. Herjan, T.; Hong, L.; Bubenik, J.; Bulek, K.; Qian, W.; Liu, C.; Li, X.; Chen, X.; Yang, H.; Ouyang, S.; et al. IL-17-receptor-associated adaptor Act1 directly stabilizes mRNAs to mediate IL-17 inflammatory signaling. Nat. Immunol. 2018, 19, 354–365. [Google Scholar] [CrossRef]
  13. Loetscher, P.; Seitz, M.; Clark-Lewis, I.; Baggiolini, M.; Moser, B. Both interleukin-8 receptors independently mediate chemotaxis. Jurkat cells transfected with IL-8R1 or IL-8R2 migrate in response to IL-8, GRO alpha and NAP-2. FEBS Lett. 1994, 341, 187–192. [Google Scholar] [CrossRef] [PubMed]
  14. Szabo, M.C.; Soo, K.S.; Zlotnik, A.; Schall, T.J. Chemokine class differences in binding to the Duffy antigen-erythrocyte chemokine receptor. J. Biol. Chem. 1995, 270, 25348–25351. [Google Scholar] [CrossRef]
  15. Fukuma, N.; Akimitsu, N.; Hamamoto, H.; Kusuhara, H.; Sugiyama, Y.; Sekimizu, K. A role of the Duffy antigen for the maintenance of plasma chemokine concentrations. Biochem. Biophys. Res. Commun. 2003, 303, 137–139. [Google Scholar] [CrossRef]
  16. Lee, J.S.; Frevert, C.W.; Wurfel, M.M.; Peiper, S.C.; Wong, V.A.; Ballman, K.K.; Ruzinski, J.T.; Rhim, J.S.; Martin, T.R.; Goodman, R.B. Duffy antigen facilitates movement of chemokine across the endothelium in vitro and promotes neutrophil transmigration in vitro and in vivo. J. Immunol. 2003, 170, 5244–5251. [Google Scholar] [CrossRef] [PubMed]
  17. Kuwano, Y.; Adler, M.; Zhang, H.; Groisman, A.; Ley, K. Gαi2 and Gαi3 differentially regulate arrest from flow and chemotaxis in mouse neutrophils. J. Immunol. 2016, 196, 3828–3833. [Google Scholar] [CrossRef]
  18. Raman, D.; Neel, N.F.; Sai, J.; Mernaugh, R.L.; Ham, A.J.; Richmond, A.J. Characterization of chemokine receptor CXCR2 interacting proteins using a proteomics approach to define the CXCR2 “chemosynapse”. Methods Enzymol. 2009, 460, 315–330. [Google Scholar]
  19. Moser, B.; Clark-Lewis, I.; Zwahlen, R.; Baggiolini, M. Neutrophil-activating properties of the melanoma growth-stimulatory activity. J. Exp. Med. 1990, 171, 1797–1802. [Google Scholar] [CrossRef]
  20. Richmond, A.; Lawson, D.H.; Nixon, D.W.; Chawla, R.K. Characterization of autostimulatory and transforming growth factors from human melanoma cells. Cancer Res. 1985, 45, 6390–6394. [Google Scholar]
  21. Argiris, A.; Karamouzis, M.V.; Raben, D.; Ferris, R.L. Head and neck cancer. Lancet 2008, 371, 1695–1709. [Google Scholar] [CrossRef]
  22. Chow, L.Q.M. Head and Neck Cancer. N. Engl. J. Med. 2020, 382, 60–72. [Google Scholar] [CrossRef] [PubMed]
  23. Rettig, E.M.; D’Souza, G. Epidemiology of head and neck cancer. Surg. Oncol. Clin. N. Am. 2015, 24, 379–396. [Google Scholar] [CrossRef] [PubMed]
  24. Kolář, M.; Szabo, P.; Dvořánková, B.; Lacina, L.; Gabius, H.J.; Strnad, H.; Sáchová, J.; Vlček, C.; Plzák, J.; Chovanec, M.; et al. Upregulation of IL-6, IL-8 and CXCL-1 production in dermal fibroblasts by normal/malignant epithelial cells in vitro: Immunohistochemical and transcriptomic analyses. Biol. Cell 2012, 104, 738–751. [Google Scholar] [CrossRef]
  25. Ye, M.Y.; Chen, M.Y.; Chang, Y.H.; Huang, J.S.; Huang, T.T.; Wong, T.Y.; Hong, T.M.; Chen, Y.L. Growth-regulated oncogene-α from oral submucous fibrosis fibroblasts promotes malignant transformation of oral precancerous cells. J. Oral Pathol. Med. 2018, 47, 880–886. [Google Scholar] [CrossRef]
  26. Jing, F.; Wang, J.; Zhou, L.; Ning, Y.; Xu, S.; Zhu, Y. Bioinformatics analysis of the role of CXC ligands in the microenvironment of head and neck tumor. Aging 2021, 13, 17789–17817. [Google Scholar] [CrossRef] [PubMed]
  27. Li, Y.; Wu, T.; Gong, S.; Zhou, H.; Yu, L.; Liang, M.; Shi, R.; Wu, Z.; Zhang, J.; Li, S. Analysis of the Prognosis and Therapeutic Value of the CXC Chemokine Family in Head and Neck Squamous Cell Carcinoma. Front. Oncol. 2021, 10, 570736. [Google Scholar] [CrossRef]
  28. Shintani, S.; Ishikawa, T.; Nonaka, T.; Li, C.; Nakashiro, K.; Wong, D.T.; Hamakawa, H. Growth-regulated oncogene-1 expression is associated with angiogenesis and lymph node metastasis in human oral cancer. Oncology 2004, 66, 316–322. [Google Scholar] [CrossRef]
  29. Ye, H.; Yu, T.; Temam, S.; Ziober, B.L.; Wang, J.; Schwartz, J.L.; Mao, L.; Wong, D.T.; Zhou, X. Transcriptomic dissection of tongue squamous cell carcinoma. BMC Genom. 2008, 9, 69. [Google Scholar] [CrossRef]
  30. Chakrabarti, S.; Multani, S.; Dabholkar, J.; Saranath, D. Whole genome expression profiling in chewing-tobacco-associated oral cancers: A pilot study. Med. Oncol. 2015, 32, 60. [Google Scholar] [CrossRef]
  31. Wei, L.Y.; Lee, J.J.; Yeh, C.Y.; Yang, C.J.; Kok, S.H.; Ko, J.Y.; Tsai, F.C.; Chia, J.S. Reciprocal activation of cancer-associated fibroblasts and oral squamous carcinoma cells through CXCL1. Oral Oncol. 2019, 88, 115–123. [Google Scholar] [CrossRef] [PubMed]
  32. Liu, S.; Wang, X.; Xu, X. Expression of GRO-1 and its relationship with VEGF in squamous cell carcinoma of larynx. Lin Chuang Er Bi Yan Hou Ke Za Zhi 2006, 20, 541–544. [Google Scholar] [PubMed]
  33. Han, L.; Liu, W.; Chen, Y.; Wu, H.; Zhang, Y.; Jiang, B. GROα expression and its prognostic implications in laryngeal squamous cell carcinoma. Neoplasma 2015, 62, 152–158. [Google Scholar] [CrossRef] [PubMed]
  34. Reyimu, A.; Chen, Y.; Song, X.; Zhou, W.; Dai, J.; Jiang, F. Identification of latent biomarkers in connection with progression and prognosis in oral cancer by comprehensive bioinformatics analysis. World J. Surg. Oncol. 2021, 19, 240. [Google Scholar] [CrossRef]
  35. Yang, B.; Dong, K.; Guo, P.; Guo, P.; Jie, G.; Zhang, G.; Li, T. Identification of Key Biomarkers and Potential Molecular Mechanisms in Oral Squamous Cell Carcinoma by Bioinformatics Analysis. J. Comput. Biol. 2020, 27, 40–54. [Google Scholar] [CrossRef]
  36. Sun, W.; Qiu, Z.; Huang, W.; Cao, M. Gene expression profiles and protein-protein interaction networks during tongue carcinogenesis in the tumor microenvironment. Mol. Med. Rep. 2018, 17, 165–171. [Google Scholar] [CrossRef]
  37. Brøndum, L.; Eriksen, J.G.; Singers Sørensen, B.; Mortensen, L.S.; Toustrup, K.; Overgaard, J.; Alsner, J. Plasma proteins as prognostic biomarkers in radiotherapy treated head and neck cancer patients. Clin. Transl. Radiat. Oncol. 2017, 2, 46–52. [Google Scholar] [CrossRef]
  38. Michiels, K.; Schutyser, E.; Conings, R.; Lenaerts, J.P.; Put, W.; Nuyts, S.; Delaere, P.; Jacobs, R.; Struyf, S.; Proost, P.; et al. Carcinoma cell-derived chemokines and their presence in oral fluid. Eur. J. Oral Sci. 2009, 117, 362–368. [Google Scholar] [CrossRef]
  39. Wolff, H.A.; Rolke, D.; Rave-Fränk, M.; Schirmer, M.; Eicheler, W.; Doerfler, A.; Hille, A.; Hess, C.F.; Matthias, C.; Rödel, R.M.; et al. Analysis of chemokine and chemokine receptor expression in squamous cell carcinoma of the head and neck (SCCHN) cell lines. Radiat. Environ. Biophys. 2011, 50, 145–154. [Google Scholar] [CrossRef]
  40. Lee, C.H.; Syu, S.H.; Liu, K.J.; Chu, P.Y.; Yang, W.C.; Lin, P.; Shieh, W.Y. Interleukin-1 beta transactivates epidermal growth factor receptor via the CXCL1-CXCR2 axis in oral cancer. Oncotarget 2015, 6, 38866–38880. [Google Scholar] [CrossRef]
  41. Kaneko, T.; Zhang, Z.; Mantellini, M.G.; Karl, E.; Zeitlin, B.; Verhaegen, M.; Soengas, M.S.; Lingen, M.; Strieter, R.M.; Nunez, G.; et al. Bcl-2 orchestrates a cross-talk between endothelial and tumor cells that promotes tumor growth. Cancer Res. 2007, 67, 9685–9693. [Google Scholar] [CrossRef]
  42. Karl, E.; Warner, K.; Zeitlin, B.; Kaneko, T.; Wurtzel, L.; Jin, T.; Chang, J.; Wang, S.; Wang, C.Y.; Strieter, R.M.; et al. Bcl-2 acts in a proangiogenic signaling pathway through nuclear factor-kappaB and CXC chemokines. Cancer Res. 2005, 65, 5063–5069. [Google Scholar] [CrossRef]
  43. Loukinova, E.; Chen, Z.; Van Waes, C.; Dong, G. Expression of proangiogenic chemokine Gro 1 in low and high metastatic variants of Pam murine squamous cell carcinoma is differentially regulated by IL-1alpha, EGF and TGF-beta1 through NF-kappaB dependent and independent mechanisms. Int. J. Cancer 2001, 94, 637–644. [Google Scholar] [CrossRef]
  44. Lyons, J.G.; Patel, V.; Roue, N.C.; Fok, S.Y.; Soon, L.L.; Halliday, G.M.; Gutkind, J.S. Snail up-regulates proinflammatory mediators and inhibits differentiation in oral keratinocytes. Cancer Res. 2008, 68, 4525–4530. [Google Scholar] [CrossRef]
  45. Bae, J.Y.; Kim, E.K.; Yang, D.H.; Zhang, X.; Park, Y.J.; Lee, D.Y.; Che, C.M.; Kim, J. Reciprocal interaction between carcinoma-associated fibroblasts and squamous carcinoma cells through interleukin-1α induces cancer progression. Neoplasia 2014, 16, 928–938. [Google Scholar] [CrossRef]
  46. Kim, E.K.; Moon, S.; Kim, D.K.; Zhang, X.; Kim, J. CXCL1 induces senescence of cancer-associated fibroblasts via autocrine loops in oral squamous cell carcinoma. PLoS ONE 2018, 13, e0188847. [Google Scholar] [CrossRef]
  47. Khurram, S.A.; Bingle, L.; McCabe, B.M.; Farthing, P.M.; Whawell, S.A. The chemokine receptors CXCR1 and CXCR2 regulate oral cancer cell behaviour. J. Oral Pathol. Med. 2014, 43, 667–674. [Google Scholar] [CrossRef]
  48. Zhang, Z.; Pan, J.; Li, L.; Han, B.; Xiao, W. Altered phenotype of lymphatic endothelial cells induced by highly metastatic OTSCC cells contributed to the lymphatic metastasis of OTSCC cells. Cancer Sci. 2010, 101, 686–692. [Google Scholar] [CrossRef]
  49. Warner, K.A.; Miyazawa, M.; Cordeiro, M.M.; Love, W.J.; Pinsky, M.S.; Neiva, K.G.; Spalding, A.C.; Nör, J.E. Endothelial cells enhance tumor cell invasion through a crosstalk mediated by CXC chemokine signaling. Neoplasia 2008, 10, 131–139. [Google Scholar] [CrossRef]
  50. Brú, A.; Souto, J.C.; Alcolea, S.; Antón, R.; Remacha, A.; Camacho, M.; Soler, M.; Brú, I.; Porres, A.; Vila, L. Tumour cell lines HT-29 and FaDu produce proinflammatory cytokines and activate neutrophils in vitro: Possible applications for neutrophil-based antitumour treatment. Mediat. Inflamm. 2009, 2009, 817498. [Google Scholar] [CrossRef]
  51. Dufies, M.; Grytsai, O.; Ronco, C.; Camara, O.; Ambrosetti, D.; Hagege, A.; Parola, J.; Mateo, L.; Ayrault, M.; Giuliano, S.; et al. New CXCR1/CXCR2 inhibitors represent an effective treatment for kidney or head and neck cancers sensitive or refractory to reference treatments. Theranostics 2019, 9, 5332–5346. [Google Scholar] [CrossRef]
  52. Cancer Genome Atlas Research Network; Weinstein, J.N.; Collisson, E.A.; Mills, G.B.; Shaw, K.R.; Ozenberger, B.A.; Ellrott, K.; Shmulevich, I.; Sander, C.; Stuart, J.M. The Cancer Genome Atlas Pan-Cancer analysis project. Nat. Genet. 2013, 45, 1113–1120. [Google Scholar] [CrossRef] [PubMed]
  53. Smyth, E.C.; Lagergren, J.; Fitzgerald, R.C.; Lordick, F.; Shah, M.A.; Lagergren, P.; Cunningham, D. Oesophageal cancer. Nat. Rev. Dis. Prim. 2017, 3, 17048. [Google Scholar] [CrossRef] [PubMed]
  54. Wang, B.; Hendricks, D.T.; Wamunyokoli, F.; Parker, M.I. A growth-related oncogene/CXC chemokine receptor 2 autocrine loop contributes to cellular proliferation in esophageal cancer. Cancer Res. 2006, 66, 3071–3077. [Google Scholar] [CrossRef] [PubMed]
  55. Feng, Z.; Qu, J.; Liu, X.; Liang, J.; Li, Y.; Jiang, J.; Zhang, H.; Tian, H. Integrated bioinformatics analysis of differentially expressed genes and immune cell infiltration characteristics in Esophageal Squamous cell carcinoma. Sci. Rep. 2021, 11, 16696. [Google Scholar] [CrossRef]
  56. Zhang, H.; Yue, J.; Jiang, Z.; Zhou, R.; Xie, R.; Xu, Y.; Wu, S. CAF-secreted CXCL1 conferred radioresistance by regulating DNA damage response in a ROS-dependent manner in esophageal squamous cell carcinoma. Cell Death Dis. 2017, 8, e2790. [Google Scholar] [CrossRef]
  57. Hasib, F.M.Y. Esophageal squamous cell carcinoma: Integrated bioinformatics analysis for differential gene expression with identification of hub genes and lncRNA. Biochem. Biophys. Rep. 2022, 30, 101262. [Google Scholar] [CrossRef]
  58. Aversa, J.; Song, M.; Shimazu, T.; Inoue, M.; Charvat, H.; Yamaji, T.; Sawada, N.; Pfeiffer, R.M.; Karimi, P.; Dawsey, S.M.; et al. Prediagnostic circulating inflammation biomarkers and esophageal squamous cell carcinoma: A case-cohort study in Japan. Int. J. Cancer 2020, 147, 686–691. [Google Scholar] [CrossRef]
  59. Alvarez, H.; Opalinska, J.; Zhou, L.; Sohal, D.; Fazzari, M.J.; Yu, Y.; Montagna, C.; Montgomery, E.A.; Canto, M.; Dunbar, K.B.; et al. Widespread hypomethylation occurs early and synergizes with gene amplification during esophageal carcinogenesis. PLoS Genet. 2011, 7, e1001356. [Google Scholar] [CrossRef]
  60. Fang, L.; Che, Y.; Zhang, C.; Huang, J.; Lei, Y.; Lu, Z.; Sun, N.; He, J. LAMC1 upregulation via TGFβ induces inflammatory cancer-associated fibroblasts in esophageal squamous cell carcinoma via NF-κB-CXCL1-STAT3. Mol. Oncol. 2021, 15, 3125–3146. [Google Scholar] [CrossRef]
  61. Wang, B.; Khachigian, L.M.; Esau, L.; Birrer, M.J.; Zhao, X.; Parker, M.I.; Hendricks, D.T. A key role for early growth response-1 and nuclear factor-kappaB in mediating and maintaining GRO/CXCR2 proliferative signaling in esophageal cancer. Mol. Cancer Res. 2009, 7, 755–764. [Google Scholar] [CrossRef]
  62. Urakawa, N.; Utsunomiya, S.; Nishio, M.; Shigeoka, M.; Takase, N.; Arai, N.; Kakeji, Y.; Koma, Y.; Yokozaki, H. GDF15 derived from both tumor-associated macrophages and esophageal squamous cell carcinomas contributes to tumor progression via Akt and Erk pathways. Lab. Investig. 2015, 95, 491–503. [Google Scholar] [CrossRef]
  63. Peng, D.F.; Hu, T.L.; Soutto, M.; Belkhiri, A.; El-Rifai, W. Loss of glutathione peroxidase 7 promotes TNF-α-induced NF-κB activation in Barrett’s carcinogenesis. Carcinogenesis 2014, 35, 1620–1628. [Google Scholar] [CrossRef]
  64. Qin, G.; Lian, J.; Huang, L.; Zhao, Q.; Liu, S.; Zhang, Z.; Chen, X.; Yue, D.; Li, L.; Li, F.; et al. Metformin blocks myeloid-derived suppressor cell accumulation through AMPK-DACH1-CXCL1 axis. Oncoimmunology 2018, 7, e1442167. [Google Scholar] [CrossRef]
  65. Tang, Z.; Li, C.; Kang, B.; Gao, G.; Li, C.; Zhang, Z. GEPIA: A web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 2017, 45, W98–W102. [Google Scholar] [CrossRef]
  66. Smyth, E.C.; Nilsson, M.; Grabsch, H.I.; van Grieken, N.C.; Lordick, F. Gastric cancer. Lancet 2020, 396, 635–648. [Google Scholar] [CrossRef]
  67. Yan, L.; Chen, Y.; Chen, F.; Tao, T.; Hu, Z.; Wang, J.; You, J.; Wong, B.C.Y.; Chen, J.; Ye, W. Effect of Helicobacter pylori Eradication on Gastric Cancer Prevention: Updated Report from a Randomized Controlled Trial with 26.5 Years of Follow-up. Gastroenterology 2022, 163, 154–162.e3. [Google Scholar] [CrossRef]
  68. Hooi, J.K.Y.; Lai, W.Y.; Ng, W.K.; Suen, M.M.Y.; Underwood, F.E.; Tanyingoh, D.; Malfertheiner, P.; Graham, D.Y.; Wong, V.W.S.; Wu, J.C.Y.; et al. Global Prevalence of Helicobacter pylori Infection: Systematic Review and Meta-Analysis. Gastroenterology 2017, 153, 420–429. [Google Scholar] [CrossRef]
  69. Yin, H.; Chu, A.; Liu, S.; Yuan, Y.; Gong, Y. Identification of DEGs and transcription factors involved in H. pylori-associated inflammation and their relevance with gastric cancer. PeerJ 2020, 8, e9223. [Google Scholar] [CrossRef] [PubMed]
  70. Ni, H.K.; Liao, L.M.; Huang, R.L.; Zhou, W. The relationship between gastric cancer and Helicobacter pylori cytotoxin-related gene A genotypes. Cell Mol. Biol. 2020, 66, 1–4. [Google Scholar] [CrossRef] [PubMed]
  71. Cover, T.L.; Lacy, D.B.; Ohi, M.D. The Helicobacter pylori Cag Type IV Secretion System. Trends Microbiol. 2020, 28, 682–695. [Google Scholar] [CrossRef]
  72. de Brito, B.B.; da Silva, F.A.F.; Soares, A.S.; Pereira, V.A.; Santos, M.L.C.; Sampaio, M.M.; Neves, P.H.M.; de Melo, F.F. Pathogenesis and clinical management of Helicobacter pylori gastric infection. World J. Gastroenterol. 2019, 25, 5578–5589. [Google Scholar] [CrossRef] [PubMed]
  73. Yamaoka, Y.; Kita, M.; Kodama, T.; Sawai, N.; Tanahashi, T.; Kashima, K.; Imanishi, J. Chemokines in the gastric mucosa in Helicobacter pylori infection. Gut 1998, 42, 609–617. [Google Scholar] [CrossRef] [PubMed]
  74. Cai, Q.; Shi, P.; Yuan, Y.; Peng, J.; Ou, X.; Zhou, W.; Li, J.; Su, T.; Lin, L.; Cai, S.; et al. Inflammation-Associated Senescence Promotes Helicobacter pylori-Induced Atrophic Gastritis. Cell Mol. Gastroenterol. Hepatol. 2021, 11, 857–880. [Google Scholar] [CrossRef]
  75. Tran, C.T.; Garcia, M.; Garnier, M.; Burucoa, C.; Bodet, C. Inflammatory signaling pathways induced by Helicobacter pylori in primary human gastric epithelial cells. Innate Immun. 2017, 23, 165–174. [Google Scholar] [CrossRef]
  76. Mustapha, P.; Paris, I.; Garcia, M.; Tran, C.T.; Cremniter, J.; Garnier, M.; Faure, J.P.; Barthes, T.; Boneca, I.G.; Morel, F.; et al. Chemokines and antimicrobial peptides have a cag-dependent early response to Helicobacter pylori infection in primary human gastric epithelial cells. Infect. Immun. 2014, 82, 2881–2889. [Google Scholar] [CrossRef]
  77. Suzuki, H.; Mori, M.; Sakaguchi, A.A.; Suzuki, M.; Miura, S.; Ishii, H. Enhanced levels of C-X-C chemokine, human GROalpha, in Helicobacter pylori-associated gastric disease. J. Gastroenterol. Hepatol. 1998, 13, 516–520. [Google Scholar] [CrossRef]
  78. Sierra, J.C.; Asim, M.; Verriere, T.G.; Piazuelo, M.B.; Suarez, G.; Romero-Gallo, J.; Delgado, A.G.; Wroblewski, L.E.; Barry, D.P.; Peek, R.M., Jr.; et al. Epidermal growth factor receptor inhibition downregulates Helicobacter pylori-induced epithelial inflammatory responses, DNA damage and gastric carcinogenesis. Gut 2018, 67, 1247–1260. [Google Scholar] [CrossRef]
  79. Neuper, T.; Frauenlob, T.; Sarajlic, M.; Posselt, G.; Wessler, S.; Horejs-Hoeck, J. TLR2, TLR4 and TLR10 Shape the Cytokine and Chemokine Release of H. pylori-Infected Human DCs. Int. J. Mol. Sci. 2020, 21, 3897. [Google Scholar] [CrossRef]
  80. Kuzuhara, T.; Suganuma, M.; Kurusu, M.; Fujiki, H. Helicobacter pylori-secreting protein Tipalpha is a potent inducer of chemokine gene expressions in stomach cancer cells. J. Cancer Res. Clin. Oncol. 2007, 133, 287–296. [Google Scholar] [CrossRef]
  81. Sakitani, K.; Hirata, Y.; Hayakawa, Y.; Serizawa, T.; Nakata, W.; Takahashi, R.; Kinoshita, H.; Sakamoto, K.; Nakagawa, H.; Akanuma, M.; et al. Role of interleukin-32 in Helicobacter pylori-induced gastric inflammation. Infect. Immun. 2012, 80, 3795–3803. [Google Scholar] [CrossRef] [PubMed]
  82. Sieveking, D.; Mitchell, H.M.; Day, A.S. Gastric epithelial cell CXC chemokine secretion following Helicobacter pylori infection in vitro. J. Gastroenterol. Hepatol. 2004, 19, 982–987. [Google Scholar] [CrossRef] [PubMed]
  83. Lee, I. Critical pathogenic steps to high risk Helicobacter pylori gastritis and gastric carcinogenesis. World J. Gastroenterol. 2014, 20, 6412–6419. [Google Scholar] [CrossRef] [PubMed]
  84. Eck, M.; Schmausser, B.; Scheller, K.; Toksoy, A.; Kraus, M.; Menzel, T.; Müller-Hermelink, H.K.; Gillitzer, R. CXC chemokines Gro(alpha)/IL-8 and IP-10/MIG in Helicobacter pylori gastritis. Clin. Exp. Immunol. 2000, 122, 192–199. [Google Scholar] [CrossRef] [PubMed]
  85. Satoh, K.; Kihira, K.; Kawata, H.; Tokumaru, K.; Kumakura, Y.; Ishino, Y.; Kawakami, S.; Inoue, K.; Kojima, T.; Satoh, Y.; et al. p53 expression in the gastric mucosa before and after eradication of Helicobacter pylori. Helicobacter 2001, 6, 31–36. [Google Scholar] [CrossRef]
  86. Li, J.H.; Shi, X.Z.; Lv, S.; Liu, M.; Xu, G.W. Effect of Helicobacter pylori infection on p53 expression of gastric mucosa and adenocarcinoma with microsatellite instability. World J. Gastroenterol. 2005, 11, 4363–4366. [Google Scholar] [CrossRef]
  87. Shu, X.; Yang, Z.; Li, Z.H.; Chen, L.; Zhou, X.D.; Xie, Y.; Lu, N.H. Helicobacter pylori Infection Activates the Akt-Mdm2-p53 Signaling Pathway in Gastric Epithelial Cells. Dig. Dis. Sci. 2015, 60, 876–886. [Google Scholar] [CrossRef]
  88. Triantafyllou, K.; Papadopoulos, V.; Emanouil, T.; Gkolfakis, P.; Damaskou, V.; Tziatzios, G.; Panayiotides, I.G.; Vafiadis, I.; Ladas, S.D. Eradication of Helicobacter pylori Infection Restores ki67, p53, and Cyclin D1 Immunoreactivity in the Human Gastric Epithelium. Clin. Med. Insights Gastroenterol. 2016, 9, 73–78. [Google Scholar] [CrossRef]
  89. Toller, I.M.; Neelsen, K.J.; Steger, M.; Hartung, M.L.; Hottiger, M.O.; Stucki, M.; Kalali, B.; Gerhard, M.; Sartori, A.A.; Lopes, M.; et al. Carcinogenic bacterial pathogen Helicobacter pylori triggers DNA double-strand breaks and a DNA damage response in its host cells. Proc. Natl. Acad. Sci. USA 2011, 108, 14944–14949. [Google Scholar] [CrossRef]
  90. André, A.R.; Ferreira, M.V.; Mota, R.M.; Ferrasi, A.C.; Pardini, M.I.; Rabenhorst, S.H. Gastric adenocarcinoma and Helicobacter pylori: Correlation with p53 mutation and p27 immunoexpression. Cancer Epidemiol. 2010, 34, 618–625. [Google Scholar] [CrossRef]
  91. Imai, S.; Ooki, T.; Murata-Kamiya, N.; Komura, D.; Tahmina, K.; Wu, W.; Takahashi-Kanemitsu, A.; Knight, C.T.; Kunita, A.; Suzuki, N.; et al. Helicobacter pylori CagA elicits BRCAness to induce genome instability that may underlie bacterial gastric carcinogenesis. Cell Host Microbe 2021, 29, 941–958.e10. [Google Scholar] [CrossRef]
  92. Bussière, F.I.; Michel, V.; Mémet, S.; Avé, P.; Vivas, J.R.; Huerre, M.; Touati, E.H. pylori-induced promoter hypermethylation downregulates USF1 and USF2 transcription factor gene expression. Cell Microbiol. 2010, 12, 1124–1133. [Google Scholar] [CrossRef]
  93. Costa, L.; Corre, S.; Michel, V.; Le Luel, K.; Fernandes, J.; Ziveri, J.; Jouvion, G.; Danckaert, A.; Mouchet, N.; Da Silva Barreira, D.; et al. USF1 defect drives p53 degradation during Helicobacter pylori infection and accelerates gastric carcinogenesis. Gut 2020, 69, 1582–1591. [Google Scholar] [CrossRef]
  94. Wei, J.; Nagy, T.A.; Vilgelm, A.; Zaika, E.; Ogden, S.R.; Romero-Gallo, J.; Piazuelo, M.B.; Correa, P.; Washington, M.K.; El-Rifai, W.; et al. Regulation of p53 tumor suppressor by Helicobacter pylori in gastric epithelial cells. Gastroenterology 2010, 139, 1333–1343. [Google Scholar] [CrossRef]
  95. Abu-Lubad, M.A.; Helaly, G.F.; Haddadin, W.J.; Jarajreh, D.A.K.; Aqel, A.A.; Al-Zeer, M.A. Loss of p53 Expression in Gastric Epithelial Cells of Helicobacter pylori-Infected Jordanian Patients. Int. J. Microbiol. 2022, 2022, 7779770. [Google Scholar] [CrossRef]
  96. Junnila, S.; Kokkola, A.; Mizuguchi, T.; Hirata, K.; Karjalainen-Lindsberg, M.L.; Puolakkainen, P.; Monni, O. Gene expression analysis identifies over-expression of CXCL1, SPARC, SPP1, and SULF1 in gastric cancer. Genes Chromosom. Cancer 2010, 49, 28–39. [Google Scholar] [CrossRef]
  97. Cheng, W.L.; Wang, C.S.; Huang, Y.H.; Tsai, M.M.; Liang, Y.; Lin, K.H. Overexpression of CXCL1 and its receptor CXCR2 promote tumor invasion in gastric cancer. Ann. Oncol. 2011, 22, 2267–2276. [Google Scholar] [CrossRef]
  98. Wei, Z.W.; Xia, G.K.; Wu, Y.; Chen, W.; Xiang, Z.; Schwarz, R.E.; Brekken, R.A.; Awasthi, N.; He, Y.L.; Zhang, C.H. CXCL1 promotes tumor growth through VEGF pathway activation and is associated with inferior survival in gastric cancer. Cancer Lett. 2015, 359, 335–343. [Google Scholar] [CrossRef]
  99. Chen, X.; Jin, R.; Chen, R.; Huang, Z. Complementary action of CXCL1 and CXCL8 in pathogenesis of gastric carcinoma. Int. J. Clin. Exp. Pathol. 2018, 11, 1036–1045. [Google Scholar]
  100. Eck, M.; Schmausser, B.; Scheller, K.; Brändlein, S.; Müller-Hermelink, H.K. Pleiotropic effects of CXC chemokines in gastric carcinoma: Differences in CXCL8 and CXCL1 expression between diffuse and intestinal types of gastric carcinoma. Clin. Exp. Immunol. 2003, 134, 508–515. [Google Scholar] [CrossRef]
  101. Yamamoto, Y.; Kuroda, K.; Sera, T.; Sugimoto, A.; Kushiyama, S.; Nishimura, S.; Togano, S.; Okuno, T.; Yoshii, M.; Tamura, T.; et al. The Clinicopathological Significance of the CXCR2 Ligands, CXCL1, CXCL2, CXCL3, CXCL5, CXCL6, CXCL7, and CXCL8 in Gastric Cancer. Anticancer Res. 2019, 39, 6645–6652. [Google Scholar] [CrossRef] [PubMed]
  102. Jung, J.J.; Noh, S.; Jeung, H.C.; Jung, M.; Kim, T.S.; Noh, S.H.; Roh, J.K.; Chung, H.C.; Rha, S.Y. Chemokine growth-regulated oncogene 1 as a putative biomarker for gastric cancer progression. Cancer Sci. 2010, 101, 2200–2206. [Google Scholar] [CrossRef] [PubMed]
  103. Wang, Q.; Li, D.; Zhang, W.; Tang, B.; Li, Q.Q.; Li, L. Evaluation of proteomics-identified CCL18 and CXCL1 as circulating tumor markers for differential diagnosis between ovarian carcinomas and benign pelvic masses. Int. J. Biol. Mrk. 2011, 26, 262–273. [Google Scholar] [CrossRef] [PubMed]
  104. Fujisawa, N.; Sakao, Y.; Hayashi, S.; Hadden, W.A., 3rd; Harmon, C.L.; Miller, E.J. alpha-Chemokine growth factors for adenocarcinomas; a synthetic peptide inhibitor for alpha-chemokines inhibits the growth of adenocarcinoma cell lines. J. Cancer Res. Clin. Oncol. 2000, 126, 19–26. [Google Scholar] [CrossRef]
  105. Wang, L.; Zhang, C.; Xu, J.; Wu, H.; Peng, J.; Cai, S.; He, Y. CXCL1 gene silencing inhibits HGC803 cell migration and invasion and acts as an independent prognostic factor for poor survival in gastric cancer. Mol. Med. Rep. 2016, 14, 4673–4679. [Google Scholar] [CrossRef]
  106. Shrestha, S.; Yang, C.D.; Hong, H.C.; Chou, C.H.; Tai, C.S.; Chiew, M.Y.; Chen, W.L.; Weng, S.L.; Chen, C.C.; Chang, Y.A.; et al. Integrated MicroRNA-mRNA Analysis Reveals miR-204 Inhibits Cell Proliferation in Gastric Cancer by Targeting CKS1B, CXCL1 and GPRC5A. Int. J. Mol. Sci. 2017, 19, 87. [Google Scholar] [CrossRef]
  107. Zhou, Z.; Xia, G.; Xiang, Z.; Liu, M.; Wei, Z.; Yan, J.; Chen, W.; Zhu, J.; Awasthi, N.; Sun, X.; et al. A C-X-C Chemokine Receptor Type 2-Dominated Cross-talk between Tumor Cells and Macrophages Drives Gastric Cancer Metastasis. Clin. Cancer Res. 2019, 25, 3317–3328. [Google Scholar] [CrossRef]
  108. Naito, Y.; Yamamoto, Y.; Sakamoto, N.; Shimomura, I.; Kogure, A.; Kumazaki, M.; Yokoi, A.; Yashiro, M.; Kiyono, T.; Yanagihara, K.; et al. Cancer extracellular vesicles contribute to stromal heterogeneity by inducing chemokines in cancer-associated fibroblasts. Oncogene 2019, 38, 5566–5579. [Google Scholar] [CrossRef]
  109. Wang, Z.; Wang, Z.; Li, G.; Wu, H.; Sun, K.; Chen, J.; Feng, Y.; Chen, C.; Cai, S.; Xu, J.; et al. CXCL1 from tumor-associated lymphatic endothelial cells drives gastric cancer cell into lymphatic system via activating integrin β1/FAK/AKT signaling. Cancer Lett. 2017, 385, 28–38. [Google Scholar] [CrossRef]
  110. Xiang, Z.; Zhou, Z.J.; Xia, G.K.; Zhang, X.H.; Wei, Z.W.; Zhu, J.T.; Yu, J.; Chen, W.; He, Y.; Schwarz, R.E.; et al. A positive crosstalk between CXCR4 and CXCR2 promotes gastric cancer metastasis. Oncogene 2017, 36, 5122–5133. [Google Scholar] [CrossRef]
  111. Xu, J.; Zhang, C.; He, Y.; Wu, H.; Wang, Z.; Song, W.; Li, W.; He, W.; Cai, S.; Zhan, W. Lymphatic endothelial cell-secreted CXCL1 stimulates lymphangiogenesis and metastasis of gastric cancer. Int. J. Cancer 2012, 130, 787–797. [Google Scholar] [CrossRef]
  112. Kasashima, H.; Yashiro, M.; Nakamae, H.; Masuda, G.; Kinoshita, H.; Morisaki, T.; Fukuoka, T.; Hasegawa, T.; Nakane, T.; Hino, M.; et al. Clinicopathologic significance of the CXCL1-CXCR2 axis in the tumor microenvironment of gastric carcinoma. PLoS ONE 2017, 12, e0178635. [Google Scholar] [CrossRef]
  113. Zhou, X.; Fang, D.; Liu, H.; Ou, X.; Zhang, C.; Zhao, Z.; Zhao, S.; Peng, J.; Cai, S.; He, Y.; et al. PMN-MDSCs accumulation induced by CXCL1 promotes CD8+ T cells exhaustion in gastric cancer. Cancer Lett. 2022, 532, 215598. [Google Scholar] [CrossRef]
  114. Kasashima, H.; Yashiro, M.; Nakamae, H.; Kitayama, K.; Masuda, G.; Kinoshita, H.; Fukuoka, T.; Hasegawa, T.; Nakane, T.; Hino, M.; et al. CXCL1-Chemokine (C-X-C Motif) Receptor 2 Signaling Stimulates the Recruitment of Bone Marrow-Derived Mesenchymal Cells into Diffuse-Type Gastric Cancer Stroma. Am. J. Pathol. 2016, 186, 3028–3039. [Google Scholar] [CrossRef]
  115. Xiang, Z.; Jiang, D.P.; Xia, G.G.; Wei, Z.W.; Chen, W.; He, Y.; Zhang, C.H. CXCL1 expression is correlated with Snail expression and affects the prognosis of patients with gastric cancer. Oncol. Lett. 2015, 10, 2458–2464. [Google Scholar] [CrossRef]
  116. Chen, X.; Chen, R.; Jin, R.; Huang, Z. The role of CXCL chemokine family in the development and progression of gastric cancer. Int. J. Clin. Exp. Pathol. 2020, 13, 484–492. [Google Scholar]
  117. Forner, A.; Reig, M.; Bruix, J. Hepatocellular carcinoma. Lancet 2018, 391, 1301–1314. [Google Scholar] [CrossRef]
  118. Nischalke, H.D.; Berger, C.; Luda, C.; Müller, T.; Berg, T.; Coenen, M.; Krämer, B.; Körner, C.; Trebicka, J.; Grünhage, F.; et al. The CXCL1 rs4074 A allele is associated with enhanced CXCL1 responses to TLR2 ligands and predisposes to cirrhosis in HCV genotype 1-infected Caucasian patients. J. Hepatol. 2012, 56, 758–764. [Google Scholar] [CrossRef]
  119. Nischalke, H.D.; Berger, C.; Lutz, P.; Langhans, B.; Wolter, F.; Eisenhardt, M.; Krämer, B.; Kokordelis, P.; Glässner, A.; Müller, T.; et al. Influence of the CXCL1 rs4074 A allele on alcohol induced cirrhosis and HCC in patients of European descent. PLoS ONE 2013, 8, e80848. [Google Scholar] [CrossRef]
  120. Wilson, C.L.; Jurk, D.; Fullard, N.; Banks, P.; Page, A.; Luli, S.; Elsharkawy, A.M.; Gieling, R.G.; Chakraborty, J.B.; Fox, C.; et al. NFκB1 is a suppressor of neutrophil-driven hepatocellular carcinoma. Nat. Commun. 2015, 6, 6818. [Google Scholar] [CrossRef]
  121. Yoo, Y.D.; Ueda, H.; Park, K.; Flanders, K.C.; Lee, Y.I.; Jay, G.; Kim, S.J. Regulation of transforming growth factor-beta 1 expression by the hepatitis B virus (HBV) X transactivator. Role in HBV pathogenesis. J. Clin. Investig. 1996, 97, 388–395. [Google Scholar] [CrossRef] [PubMed]
  122. Li, H.Y.; Ju, D.; Zhang, D.W.; Li, H.; Kong, L.M.; Guo, Y.; Li, C.; Wang, X.L.; Chen, Z.N.; Bian, H. Activation of TGF-β1-CD147 positive feedback loop in hepatic stellate cells promotes liver fibrosis. Sci. Rep. 2015, 5, 16552. [Google Scholar] [CrossRef] [PubMed]
  123. Shi, W.P.; Ju, D.; Li, H.; Yuan, L.; Cui, J.; Luo, D.; Chen, Z.N.; Bian, H. CD147 Promotes CXCL1 Expression and Modulates Liver Fibrogenesis. Int. J. Mol. Sci. 2018, 19, 1145. [Google Scholar] [CrossRef] [PubMed]
  124. Jiang, Y.; Xi, Y.; Li, Y.; Zuo, Z.; Zeng, C.; Fan, J.; Zhang, D.; Tao, H.; Guo, Y. Ethanol promoting the upregulation of C-X-C Motif Chemokine Ligand 1 (CXCL1) and C-X-C Motif Chemokine Ligand 6 (CXCL6) in models of early alcoholic liver disease. Bioengineered 2022, 13, 4688–4701. [Google Scholar] [CrossRef]
  125. Maltby, J.; Wright, S.; Bird, G.; Sheron, N. Chemokine levels in human liver homogenates: Associations between GRO alpha and histopathological evidence of alcoholic hepatitis. Hepatology 1996, 24, 1156–1160. [Google Scholar] [CrossRef]
  126. Roh, Y.S.; Zhang, B.; Loomba, R.; Seki, E. TLR2 and TLR9 contribute to alcohol-mediated liver injury through induction of CXCL1 and neutrophil infiltration. Am. J. Physiol. Gastrointest. Liver Physiol. 2015, 309, G30–G41. [Google Scholar] [CrossRef]
  127. Stefanovic, L.; Brenner, D.A.; Stefanovic, B. Direct hepatotoxic effect of KC chemokine in the liver without infiltration of neutrophils. Exp. Biol. Med. 2005, 230, 573–586. [Google Scholar] [CrossRef]
  128. Casini, A.; Ceni, E.; Salzano, R.; Biondi, P.; Parola, M.; Galli, A.; Foschi, M.; Caligiuri, A.; Pinzani, M.; Surrenti, C. Neutrophil-derived superoxide anion induces lipid peroxidation and stimulates collagen synthesis in human hepatic stellate cells: Role of nitric oxide. Hepatology 1997, 25, 361–367. [Google Scholar] [CrossRef]
  129. Asselah, T.; Bièche, I.; Laurendeau, I.; Paradis, V.; Vidaud, D.; Degott, C.; Martinot, M.; Bedossa, P.; Valla, D.; Vidaud, M.; et al. Liver gene expression signature of mild fibrosis in patients with chronic hepatitis C. Gastroenterology 2005, 129, 2064–2075. [Google Scholar] [CrossRef]
  130. Johansson, S.; Talloen, W.; Tuefferd, M.; Darling, J.M.; Scholliers, A.; Fanning, G.; Fried, M.W.; Aerssens, J. Plasma levels of growth-related oncogene (CXCL1-3) associated with fibrosis and platelet counts in HCV-infected patients. Aliment. Pharmacol. Ther. 2015, 42, 1111–1121. [Google Scholar] [CrossRef]
  131. Cao, Z.; Fu, B.; Deng, B.; Zeng, Y.; Wan, X.; Qu, L. Overexpression of Chemokine (C-X-C) ligand 1 (CXCL1) associated with tumor progression and poor prognosis in hepatocellular carcinoma. Cancer Cell Int. 2014, 14, 86. [Google Scholar] [CrossRef]
  132. Han, K.Q.; Han, H.; He, X.Q.; Wang, L.; Guo, X.D.; Zhang, X.M.; Chen, J.; Zhu, Q.G.; Nian, H.; Zhai, X.F.; et al. Chemokine CXCL1 may serve as a potential molecular target for hepatocellular carcinoma. Cancer Med. 2016, 5, 2861–2871. [Google Scholar] [CrossRef]
  133. Wu, F.X.; Wang, Q.; Zhang, Z.M.; Huang, S.; Yuan, W.P.; Liu, J.Y.; Ban, K.C.; Zhao, Y.N. Identifying serological biomarkers of hepatocellular carcinoma using surface-enhanced laser desorption/ionization-time-of-flight mass spectroscopy. Cancer Lett. 2009, 279, 163–170. [Google Scholar] [CrossRef]
  134. Slany, A.; Haudek-Prinz, V.; Zwickl, H.; Stättner, S.; Grasl-Kraupp, B.; Gerner, C. Myofibroblasts are important contributors to human hepatocellular carcinoma: Evidence for tumor promotion by proteome profiling. Electrophoresis 2013, 34, 3315–3325. [Google Scholar] [CrossRef]
  135. Ye, L.Y.; Chen, W.; Bai, X.L.; Xu, X.Y.; Zhang, Q.; Xia, X.F.; Sun, X.; Li, G.G.; Hu, Q.D.; Fu, Q.H.; et al. Hypoxia-Induced Epithelial-to-Mesenchymal Transition in Hepatocellular Carcinoma Induces an Immunosuppressive Tumor Microenvironment to Promote Metastasis. Cancer Res. 2016, 76, 818–830. [Google Scholar] [CrossRef]
  136. Li, Y.M.; Liu, Z.Y.; Wang, J.C.; Yu, J.M.; Li, Z.C.; Yang, H.J.; Tang, J.; Chen, Z.N. Receptor-Interacting Protein Kinase 3 Deficiency Recruits Myeloid-Derived Suppressor Cells to Hepatocellular Carcinoma through the Chemokine (C-X-C Motif) Ligand 1-Chemokine (C-X-C Motif) Receptor 2 Axis. Hepatology 2019, 70, 1564–1581. [Google Scholar] [CrossRef]
  137. Tang, K.H.; Ma, S.; Lee, T.K.; Chan, Y.P.; Kwan, P.S.; Tong, C.M.; Ng, I.O.; Man, K.; To, K.F.; Lai, P.B.; et al. CD133(+) liver tumor-initiating cells promote tumor angiogenesis, growth, and self-renewal through neurotensin/interleukin-8/CXCL1 signaling. Hepatology 2012, 55, 807–820. [Google Scholar] [CrossRef]
  138. Han, K.Q.; He, X.Q.; Ma, M.Y.; Guo, X.D.; Zhang, X.M.; Chen, J.; Han, H.; Zhang, W.W.; Zhu, Q.G.; Zhao, W.Z. Targeted silencing of CXCL1 by siRNA inhibits tumor growth and apoptosis in hepatocellular carcinoma. Int. J. Oncol. 2015, 47, 2131–2140. [Google Scholar] [CrossRef]
  139. Ueda, S.; Basaki, Y.; Yoshie, M.; Ogawa, K.; Sakisaka, S.; Kuwano, M.; Ono, M. PTEN/Akt signaling through epidermal growth factor receptor is prerequisite for angiogenesis by hepatocellular carcinoma cells that is susceptible to inhibition by gefitinib. Cancer Res. 2006, 66, 5346–5353. [Google Scholar] [CrossRef]
  140. Liu, L.; Sun, H.; Wu, S.; Tan, H.; Sun, Y.; Liu, X.; Si, S.; Xu, L.; Huang, J.; Zhou, W.; et al. IL-17A promotes CXCR2-dependent angiogenesis in a mouse model of liver cancer. Mol. Med. Rep. 2019, 20, 1065–1074. [Google Scholar] [CrossRef]
  141. Dahlquist, K.J.V.; Voth, L.C.; Fee, A.J.; Stoeckman, A.K. An Autocrine Role for CXCL1 in Progression of Hepatocellular Carcinoma. Anticancer Res. 2020, 40, 6075–6081. [Google Scholar] [CrossRef] [PubMed]
  142. Xia, S.; Wu, J.; Zhou, W.; Zhang, M.; Zhao, K.; Liu, J.; Tian, D.; Liao, J. SLC7A2 deficiency promotes hepatocellular carcinoma progression by enhancing recruitment of myeloid-derived suppressors cells. Cell Death Dis. 2021, 12, 570. [Google Scholar] [CrossRef] [PubMed]
  143. Cui, X.; Li, Z.; Gao, J.; Gao, P.J.; Ni, Y.B.; Zhu, J.Y. Elevated CXCL1 increases hepatocellular carcinoma aggressiveness and is inhibited by miRNA-200a. Oncotarget 2016, 7, 65052–65066. [Google Scholar] [CrossRef] [PubMed]
  144. Fan, L.; Pan, Q.; Yang, W.; Koo, S.C.; Tian, C.; Li, L.; Lu, M.; Brown, A.; Ju, B.; Easton, J.; et al. A developmentally prometastatic niche to hepatoblastoma in neonatal liver mediated by the Cxcl1/Cxcr2 axis. Hepatology 2022, 76, 1275–1290. [Google Scholar] [CrossRef]
  145. Vansaun, M.N.; Mendonsa, A.M.; Lee Gorden, D. Hepatocellular proliferation correlates with inflammatory cell and cytokine changes in a murine model of nonalchoholic fatty liver disease. PLoS ONE 2013, 8, e73054. [Google Scholar] [CrossRef]
  146. Wolf, B.; Krieg, K.; Falk, C.; Breuhahn, K.; Keppeler, H.; Biedermann, T.; Schmid, E.; Warmann, S.; Fuchs, J.; Vetter, S.; et al. Inducing Differentiation of Premalignant Hepatic Cells as a Novel Therapeutic Strategy in Hepatocarcinoma. Cancer Res. 2016, 76, 5550–5561. [Google Scholar] [CrossRef]
  147. Li, L.; Xu, L.; Yan, J.; Zhen, Z.J.; Ji, Y.; Liu, C.Q.; Lau, W.Y.; Zheng, L.; Xu, J. CXCR2-CXCL1 axis is correlated with neutrophil infiltration and predicts a poor prognosis in hepatocellular carcinoma. J. Exp. Clin. Cancer Res. 2015, 34, 129. [Google Scholar] [CrossRef]
  148. Zhao, W.; Xu, Y.; Xu, J.; Wu, D.; Zhao, B.; Yin, Z.; Wang, X. Subsets of myeloid-derived suppressor cells in hepatocellular carcinoma express chemokines and chemokine receptors differentially. Int. Immunopharmacol. 2015, 26, 314–321. [Google Scholar] [CrossRef]
  149. Zhou, S.L.; Zhou, Z.J.; Hu, Z.Q.; Huang, X.W.; Wang, Z.; Chen, E.B.; Fan, J.; Cao, Y.; Dai, Z.; Zhou, J. Tumor-Associated Neutrophils Recruit Macrophages and T-Regulatory Cells to Promote Progression of Hepatocellular Carcinoma and Resistance to Sorafenib. Gastroenterology 2016, 150, 1646–1658.e17. [Google Scholar] [CrossRef]
  150. Chanmee, T.; Ontong, P.; Konno, K.; Itano, N. Tumor-associated macrophages as major players in the tumor microenvironment. Cancers 2014, 6, 1670–1690. [Google Scholar] [CrossRef]
  151. Shen, Q.; Hu, G.; Wu, J.; Lv, L. A new clinical prognostic nomogram for liver cancer based on immune score. PLoS ONE 2020, 15, e0236622. [Google Scholar] [CrossRef]
  152. Wang, J.; Zhang, C.; Chen, X.; Li, Y.; Li, A.; Liu, D.; Li, F.; Luo, T. Functions of CXC chemokines as biomarkers and potential therapeutic targets in the hepatocellular carcinoma microenvironment. Transl. Cancer Res. 2021, 10, 2169–2187. [Google Scholar] [CrossRef]
  153. Krasinskas, A.M. Cholangiocarcinoma. Surg. Pathol. Clin. 2018, 11, 403–429. [Google Scholar] [CrossRef]
  154. Shaib, Y.; El-Serag, H.B. The epidemiology of cholangiocarcinoma. Semin Liver Dis. 2004, 24, 115–125. [Google Scholar] [CrossRef]
  155. Yamamoto, Y.; Sugimoto, A.; Maruo, K.; Tsujio, G.; Sera, T.; Kushiyama, S.; Nishimura, S.; Kuroda, K.; Togano, S.; Eguchi, S.; et al. CXCR2 signaling might have a tumor-suppressive role in patients with cholangiocarcinoma. PLoS ONE 2022, 17, e0266027. [Google Scholar] [CrossRef]
  156. Haga, H.; Yan, I.K.; Takahashi, K.; Wood, J.; Zubair, A.; Patel, T. Tumour cell-derived extracellular vesicles interact with mesenchymal stem cells to modulate the microenvironment and enhance cholangiocarcinoma growth. J. Extracell. Vesicles 2015, 4, 24900. [Google Scholar] [CrossRef]
  157. Zhang, Q.; Ma, C.; Duan, Y.; Heinrich, B.; Rosato, U.; Diggs, L.P.; Ma, L.; Roy, S.; Fu, Q.; Brown, Z.J.; et al. Gut Microbiome Directs Hepatocytes to Recruit MDSCs and Promote Cholangiocarcinoma. Cancer Discov. 2021, 11, 1248–1267. [Google Scholar] [CrossRef]
  158. Sueoka, H.; Hirano, T.; Uda, Y.; Iimuro, Y.; Yamanaka, J.; Fujimoto, J. Blockage of CXCR2 suppresses tumor growth of intrahepatic cholangiocellular carcinoma. Surgery 2014, 155, 640–649. [Google Scholar] [CrossRef]
  159. Vincent, A.; Herman, J.; Schulick, R.; Hruban, R.H.; Goggins, M. Pancreatic cancer. Lancet 2011, 378, 607–620. [Google Scholar] [CrossRef]
  160. Nagawkar, S.S.; Abu-Funni, S.; Simon, E.; Bick, T.; Prinz, E.; Sabo, E.; Ben-Izhak, O.; Hershkovitz, D. Intratumor Heterogeneity of KRAS Mutation Status in Pancreatic Ductal Adenocarcinoma Is Associated with Smaller Lesions. Pancreas 2016, 45, 876–881. [Google Scholar] [CrossRef]
  161. Wang, Z.; Zhang, C.; Nagee, K.; Mohammadi, A.; Monteiro, C. Characterization of KRAS Mutation in Acinar and Langerhans Islet Cells of Patients with Pancreatic Ductal Adenocarcinoma. Pancreas 2016, 45, 337–341. [Google Scholar] [CrossRef] [PubMed]
  162. Choi, M.H.; Mejlænder-Andersen, E.; Manueldas, S.; El Jellas, K.; Steine, S.J.; Tjensvoll, K.; Sætran, H.A.; Knappskog, S.; Hoem, D.; Nordgård, O.; et al. Mutation analysis by deep sequencing of pancreatic juice from patients with pancreatic ductal adenocarcinoma. BMC Cancer 2019, 19, 11. [Google Scholar] [CrossRef]
  163. Lian, S.; Zhai, X.; Wang, X.; Zhu, H.; Zhang, S.; Wang, W.; Wang, Z.; Huang, J. Elevated expression of growth-regulated oncogene-alpha in tumor and stromal cells predicts unfavorable prognosis in pancreatic cancer. Medicine 2016, 95, e4328. [Google Scholar] [CrossRef] [PubMed]
  164. Nywening, T.M.; Belt, B.A.; Cullinan, D.R.; Panni, R.Z.; Han, B.J.; Sanford, D.E.; Jacobs, R.C.; Ye, J.; Patel, A.A.; Gillanders, W.E.; et al. Targeting both tumour-associated CXCR2+ neutrophils and CCR2+ macrophages disrupts myeloid recruitment and improves chemotherapeutic responses in pancreatic ductal adenocarcinoma. Gut 2018, 67, 1112–1123. [Google Scholar] [CrossRef]
  165. Huang, J.; Chen, Z.; Ding, C.; Lin, S.; Wan, D.; Ren, K. Prognostic Biomarkers and Immunotherapeutic Targets among CXC Chemokines in Pancreatic Adenocarcinoma. Front. Oncol. 2021, 11, 711402. [Google Scholar] [CrossRef] [PubMed]
  166. Saxena, S.; Molczyk, C.; Purohit, A.; Ehrhorn, E.; Goel, P.; Prajapati, D.R.; Atri, P.; Kaur, S.; Grandgenett, P.M.; Hollingsworth, M.A.; et al. Differential expression profile of CXC-receptor-2 ligands as potential biomarkers in pancreatic ductal adenocarcinoma. Am. J. Cancer Res. 2022, 12, 68–90. [Google Scholar]
  167. Steele, C.W.; Karim, S.A.; Leach, J.D.G.; Bailey, P.; Upstill-Goddard, R.; Rishi, L.; Foth, M.; Bryson, S.; McDaid, K.; Wilson, Z.; et al. CXCR2 Inhibition Profoundly Suppresses Metastases and Augments Immunotherapy in Pancreatic Ductal Adenocarcinoma. Cancer Cell 2016, 29, 832–845. [Google Scholar] [CrossRef]
  168. Takamori, H.; Oades, Z.G.; Hoch, O.C.; Burger, M.; Schraufstatter, I.U. Autocrine growth effect of IL-8 and GROalpha on a human pancreatic cancer cell line, Capan-1. Pancreas 2000, 21, 52–56. [Google Scholar] [CrossRef]
  169. Somerville, T.D.; Biffi, G.; Daßler-Plenker, J.; Hur, S.K.; He, X.Y.; Vance, K.E.; Miyabayashi, K.; Xu, Y.; Maia-Silva, D.; Klingbeil, O.; et al. Squamous trans-differentiation of pancreatic cancer cells promotes stromal inflammation. eLife 2020, 9, e53381. [Google Scholar] [CrossRef]
  170. Matsuo, Y.; Campbell, P.M.; Brekken, R.A.; Sung, B.; Ouellette, M.M.; Fleming, J.B.; Aggarwal, B.B.; Der, C.J.; Guha, S. K-Ras promotes angiogenesis mediated by immortalized human pancreatic epithelial cells through mitogen-activated protein kinase signaling pathways. Mol. Cancer Res. 2009, 7, 799–808. [Google Scholar] [CrossRef]
  171. Purohit, A.; Varney, M.; Rachagani, S.; Ouellette, M.M.; Batra, S.K.; Singh, R.K. CXCR2 signaling regulates KRAS(G¹²D)-induced autocrine growth of pancreatic cancer. Oncotarget 2016, 7, 7280–7296. [Google Scholar] [CrossRef]
  172. Woodstock, D.L.; Sammons, M.A.; Fischer, M. p63 and p53: Collaborative Partners or Dueling Rivals? Front. Cell Dev. Biol. 2021, 9, 701986. [Google Scholar] [CrossRef]
  173. Wu, H.H.; Hwang-Verslues, W.W.; Lee, W.H.; Huang, C.K.; Wei, P.C.; Chen, C.L.; Shew, J.Y.; Lee, E.Y.; Jeng, Y.M.; Tien, Y.W.; et al. Targeting IL-17B-IL-17RB signaling with an anti-IL-17RB antibody blocks pancreatic cancer metastasis by silencing multiple chemokines. J. Exp. Med. 2015, 212, 333–349. [Google Scholar] [CrossRef]
  174. Kemp, S.B.; Carpenter, E.S.; Steele, N.G.; Donahue, K.L.; Nwosu, Z.C.; Pacheco, A.; Velez-Delgado, A.; Menjivar, R.E.; Lima, F.; The, S.; et al. Apolipoprotein E Promotes Immune Suppression in Pancreatic Cancer through NF-κB-Mediated Production of CXCL1. Cancer Res. 2021, 81, 4305–4318. [Google Scholar] [CrossRef]
  175. Hrabák, P.; Kalousová, M.; Krechler, T.; Zima, T. Pancreatic stellate cells—Rising stars in pancreatic pathologies. Physiol. Res. 2021, 70, S597–S616. [Google Scholar] [CrossRef]
  176. Takikawa, T.; Masamune, A.; Yoshida, N.; Hamada, S.; Kogure, T.; Shimosegawa, T. Exosomes Derived from Pancreatic Stellate Cells: MicroRNA Signature and Effects on Pancreatic Cancer Cells. Pancreas 2017, 46, 19–27. [Google Scholar] [CrossRef]
  177. Hoffman, M.T.; Kemp, S.B.; Salas-Escabillas, D.J.; Zhang, Y.; Steele, N.G.; The, S.; Long, D.; Benitz, S.; Yan, W.; Margolskee, R.F.; et al. The Gustatory Sensory G-Protein GNAT3 Suppresses Pancreatic Cancer Progression in Mice. Cell Mol. Gastroenterol. Hepatol. 2021, 11, 349–369. [Google Scholar] [CrossRef]
  178. Erez, N.; Truitt, M.; Olson, P.; Arron, S.T.; Hanahan, D. Cancer-Associated Fibroblasts Are Activated in Incipient Neoplasia to Orchestrate Tumor-Promoting Inflammation in an NF-kappaB-Dependent Manner. Cancer Cell 2010, 17, 135–147. [Google Scholar] [CrossRef]
  179. Öhlund, D.; Handly-Santana, A.; Biffi, G.; Elyada, E.; Almeida, A.S.; Ponz-Sarvise, M.; Corbo, V.; Oni, T.E.; Hearn, S.A.; Lee, E.J.; et al. Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. J. Exp. Med. 2017, 214, 579–596. [Google Scholar] [CrossRef]
  180. Shao, C.; Tu, C.; Cheng, X.; Xu, Z.; Wang, X.; Shen, J.; Chai, K.; Chen, W. Inflammatory and Senescent Phenotype of Pancreatic Stellate Cells Induced by Sqstm1 Downregulation Facilitates Pancreatic Cancer Progression. Int. J. Biol. Sci. 2019, 15, 1020–1029. [Google Scholar] [CrossRef]
  181. Acosta, J.C.; O’Loghlen, A.; Banito, A.; Guijarro, M.V.; Augert, A.; Raguz, S.; Fumagalli, M.; Da Costa, M.; Brown, C.; Popov, N.; et al. Chemokine signaling via the CXCR2 receptor reinforces senescence. Cell 2008, 133, 1006–1018. [Google Scholar] [CrossRef] [PubMed]
  182. Guo, H.; Liu, Z.; Xu, B.; Hu, H.; Wei, Z.; Liu, Q.; Zhang, X.; Ding, X.; Wang, Y.; Zhao, M.; et al. Chemokine receptor CXCR2 is transactivated by p53 and induces p38-mediated cellular senescence in response to DNA damage. Aging Cell 2013, 12, 1110–1121. [Google Scholar] [CrossRef] [PubMed]
  183. Awaji, M.; Saxena, S.; Wu, L.; Prajapati, D.R.; Purohit, A.; Varney, M.L.; Kumar, S.; Rachagani, S.; Ly, Q.P.; Jain, M.; et al. CXCR2 signaling promotes secretory cancer-associated fibroblasts in pancreatic ductal adenocarcinoma. FASEB J. 2020, 34, 9405–9418. [Google Scholar] [CrossRef] [PubMed]
  184. Huang, C.; Li, Z.; Li, N.; Li, Y.; Chang, A.; Zhao, T.; Wang, X.; Wang, H.; Gao, S.; Yang, S.; et al. Interleukin 35 Expression Correlates with Microvessel Density in Pancreatic Ductal Adenocarcinoma, Recruits Monocytes, and Promotes Growth and Angiogenesis of Xenograft Tumors in Mice. Gastroenterology 2018, 154, 675–688. [Google Scholar] [CrossRef] [PubMed]
  185. Bellomo, G.; Rainer, C.; Quaranta, V.; Astuti, Y.; Raymant, M.; Boyd, E.; Stafferton, R.; Campbell, F.; Ghaneh, P.; Halloran, C.M.; et al. Chemotherapy-induced infiltration of neutrophils promotes pancreatic cancer metastasis via Gas6/AXL signalling axis. Gut 2022, 71, 2284–2299. [Google Scholar] [CrossRef]
  186. Lesina, M.; Wörmann, S.M.; Morton, J.; Diakopoulos, K.N.; Korneeva, O.; Wimmer, M.; Einwächter, H.; Sperveslage, J.; Demir, I.E.; Kehl, T.; et al. RelA regulates CXCL1/CXCR2-dependent oncogene-induced senescence in murine Kras-driven pancreatic carcinogenesis. J. Clin. Investig. 2016, 126, 2919–2932. [Google Scholar] [CrossRef]
  187. Maeda, S.; Kuboki, S.; Nojima, H.; Shimizu, H.; Yoshitomi, H.; Furukawa, K.; Miyazaki, M.; Ohtsuka, M. Duffy antigen receptor for chemokines (DARC) expressing in cancer cells inhibits tumor progression by suppressing CXCR2 signaling in human pancreatic ductal adenocarcinoma. Cytokine 2017, 95, 12–21. [Google Scholar] [CrossRef]
  188. Neote, K.; Mak, J.Y.; Kolakowski, L.F., Jr.; Schall, T.J. Functional and biochemical analysis of the cloned Duffy antigen: Identity with the red blood cell chemokine receptor. Blood 1994, 84, 44–52. [Google Scholar] [CrossRef]
  189. Sano, M.; Ijichi, H.; Takahashi, R.; Miyabayashi, K.; Fujiwara, H.; Yamada, T.; Kato, H.; Nakatsuka, T.; Tanaka, Y.; Tateishi, K.; et al. Blocking CXCLs-CXCR2 axis in tumor-stromal interactions contributes to survival in a mouse model of pancreatic ductal adenocarcinoma through reduced cell invasion/migration and a shift of immune-inflammatory microenvironment. Oncogenesis 2019, 8, 8. [Google Scholar] [CrossRef]
  190. Purohit, A.; Saxena, S.; Varney, M.; Prajapati, D.R.; Kozel, J.A.; Lazenby, A.; Singh, R.K. Host Cxcr2-Dependent Regulation of Pancreatic Cancer Growth, Angiogenesis, and Metastasis. Am. J. Pathol. 2021, 191, 759–771. [Google Scholar] [CrossRef]
  191. Miyake, M.; Hori, S.; Morizawa, Y.; Tatsumi, Y.; Nakai, Y.; Anai, S.; Torimoto, K.; Aoki, K.; Tanaka, N.; Shimada, K.; et al. CXCL1-Mediated Interaction of Cancer Cells with Tumor-Associated Macrophages and Cancer-Associated Fibroblasts Promotes Tumor Progression in Human Bladder Cancer. Neoplasia 2016, 18, 636–646. [Google Scholar] [CrossRef]
  192. Seifert, L.; Werba, G.; Tiwari, S.; Giao Ly, N.N.; Alothman, S.; Alqunaibit, D.; Avanzi, A.; Barilla, R.; Daley, D.; Greco, S.H.; et al. The necrosome promotes pancreatic oncogenesis via CXCL1 and Mincle-induced immune suppression. Nature 2016, 532, 245–249. [Google Scholar] [CrossRef]
  193. Di Mitri, D.; Mirenda, M.; Vasilevska, J.; Calcinotto, A.; Delaleu, N.; Revandkar, A.; Gil, V.; Boysen, G.; Losa, M.; Mosole, S.; et al. Re-education of Tumor-Associated Macrophages by CXCR2 Blockade Drives Senescence and Tumor Inhibition in Advanced Prostate Cancer. Cell Rep. 2019, 28, 2156–2168.e5. [Google Scholar] [CrossRef]
  194. Hu, J.; Zhao, Q.; Kong, L.Y.; Wang, J.; Yan, J.; Xia, X.; Jia, Z.; Heimberger, A.B.; Li, S. Regulation of tumor immune suppression and cancer cell survival by CXCL1/2 elevation in glioblastoma multiforme. Sci. Adv. 2021, 7, eabc2511. [Google Scholar] [CrossRef]
  195. Ijichi, H.; Chytil, A.; Gorska, A.E.; Aakre, M.E.; Bierie, B.; Tada, M.; Mohri, D.; Miyabayashi, K.; Asaoka, Y.; Maeda, S.; et al. Inhibiting Cxcr2 disrupts tumor-stromal interactions and improves survival in a mouse model of pancreatic ductal adenocarcinoma. J. Clin. Investig. 2011, 121, 4106–4117. [Google Scholar] [CrossRef]
  196. Aikawa, T.; Gunn, J.; Spong, S.M.; Klaus, S.J.; Korc, M. Connective tissue growth factor-specific antibody attenuates tumor growth, metastasis, and angiogenesis in an orthotopic mouse model of pancreatic cancer. Mol. Cancer Ther. 2006, 5, 1108–1116. [Google Scholar] [CrossRef]
  197. Dekker, E.; Tanis, P.J.; Vleugels, J.L.A.; Kasi, P.M.; Wallace, M.B. Colorectal cancer. Lancet 2019, 394, 1467–1480. [Google Scholar] [CrossRef]
  198. Kwong, T.N.Y.; Wang, X.; Nakatsu, G.; Chow, T.C.; Tipoe, T.; Dai, R.Z.W.; Tsoi, K.K.K.; Wong, M.C.S.; Tse, G.; Chan, M.T.V.; et al. Association between Bacteremia from Specific Microbes and Subsequent Diagnosis of Colorectal Cancer. Gastroenterology 2018, 155, 383–390.e8. [Google Scholar] [CrossRef]
  199. Cuenca, R.E.; Azizkhan, R.G.; Haskill, S. Characterization of GRO alpha, beta and gamma expression in human colonic tumours: Potential significance of cytokine involvement. Surg. Oncol. 1992, 1, 323–329. [Google Scholar] [CrossRef]
  200. Baier, P.K.; Eggstein, S.; Wolff-Vorbeck, G.; Baumgartner, U.; Hopt, U.T. Chemokines in human colorectal carcinoma. Anticancer Res. 2005, 25, 3581–3584. [Google Scholar]
  201. Wen, Y.; Giardina, S.F.; Hamming, D.; Greenman, J.; Zachariah, E.; Bacolod, M.D.; Liu, H.; Shia, J.; Amenta, P.S.; Barany, F.; et al. GROalpha is highly expressed in adenocarcinoma of the colon and down-regulates fibulin-1. Clin. Cancer Res. 2006, 12, 5951–5959. [Google Scholar] [CrossRef] [PubMed]
  202. Rubie, C.; Frick, V.O.; Wagner, M.; Schuld, J.; Gräber, S.; Brittner, B.; Bohle, R.M.; Schilling, M.K. ELR+ CXC chemokine expression in benign and malignant colorectal conditions. BMC Cancer 2008, 8, 178. [Google Scholar] [CrossRef] [PubMed]
  203. Erreni, M.; Bianchi, P.; Laghi, L.; Mirolo, M.; Fabbri, M.; Locati, M.; Mantovani, A.; Allavena, P. Expression of chemokines and chemokine receptors in human colon cancer. Methods Enzymol. 2009, 460, 105–121. [Google Scholar] [PubMed]
  204. Oladipo, O.; Conlon, S.; O’Grady, A.; Purcell, C.; Wilson, C.; Maxwell, P.J.; Johnston, P.G.; Stevenson, M.; Kay, E.W.; Wilson, R.H.; et al. The expression and prognostic impact of CXC-chemokines in stage II and III colorectal cancer epithelial and stromal tissue. Br. J. Cancer 2011, 104, 480–487. [Google Scholar] [CrossRef] [PubMed]
  205. Sun, Y.L.; Zhang, Y.; Guo, Y.C.; Yang, Z.H.; Xu, Y.C. A Prognostic Model Based on the Immune-related Genes in Colon Adenocarcinoma. Int. J. Med. Sci. 2020, 17, 1879–1896. [Google Scholar] [CrossRef]
  206. Yang, J.; Gao, S.; Qiu, M.; Kan, S. Integrated Analysis of Gene Expression and Metabolite Data Reveals Candidate Molecular Markers in Colorectal Carcinoma. Cancer Biother. Radiopharm. 2020, 37, 907–916. [Google Scholar] [CrossRef]
  207. Yu, L.; Yang, X.; Xu, C.; Sun, J.; Fang, Z.; Pan, H.; Han, W. Comprehensive analysis of the expression and prognostic value of CXC chemokines in colorectal cancer. Int. Immunopharmacol. 2020, 89, 107077. [Google Scholar] [CrossRef]
  208. Chen, B.; Song, L.; Nie, X.; Lin, F.; Yu, Z.; Kong, W.; Qi, X.; Wang, W. CXCL1 Regulated by miR-302e Is Involved in Cell Viability and Motility of Colorectal Cancer via Inhibiting JAK-STAT Signaling Pathway. Front. Oncol. 2021, 10, 577229. [Google Scholar] [CrossRef]
  209. Gong, Y.Z.; Ma, H.; Ruan, G.T.; Zhu, L.C.; Liao, X.W.; Wang, S.; Yan, L.; Huang, W.; Huang, K.T.; Xie, H.; et al. Diagnosis and prognostic value of C-X-C motif chemokine ligand 1 in colon adenocarcinoma based on the Cancer Genome Atlas and Guangxi cohort. J. Cancer 2021, 12, 5506–5518. [Google Scholar] [CrossRef]
  210. Yang, M.Q.; Bai, L.L.; Wang, Z.; Lei, L.; Zheng, Y.W.; Li, Z.H.; Huang, W.J.; Liu, C.C.; Xu, H.T. DEK is highly expressed in breast cancer and is associated with malignant phenotype and progression. Oncol. Lett. 2021, 21, 440. [Google Scholar] [CrossRef]
  211. Sipos, F.; Germann, T.M.; Wichmann, B.; Galamb, O.; Spisák, S.; Krenács, T.; Tulassay, Z.; Molnár, B.; Műzes, G. MMP3 and CXCL1 are potent stromal protein markers of dysplasia-carcinoma transition in sporadic colorectal cancer. Eur. J. Cancer Prev. 2014, 23, 336–343. [Google Scholar] [CrossRef]
  212. Ogawa, R.; Yamamoto, T.; Hirai, H.; Hanada, K.; Kiyasu, Y.; Nishikawa, G.; Mizuno, R.; Inamoto, S.; Itatani, Y.; Sakai, Y.; et al. Loss of SMAD4 Promotes Colorectal Cancer Progression by Recruiting Tumor-Associated Neutrophils via the CXCL1/8-CXCR2 Axis. Clin. Cancer Res. 2019, 25, 2887–2899. [Google Scholar] [CrossRef]
  213. Liu, K.; Lai, M.; Wang, S.; Zheng, K.; Xie, S.; Wang, X. Construction of a CXC Chemokine-Based Prediction Model for the Prognosis of Colon Cancer. Biomed Res. Int. 2020, 2020, 6107865. [Google Scholar] [CrossRef]
  214. Chiu, S.T.; Hsieh, F.J.; Chen, S.W.; Chen, C.L.; Shu, H.F.; Li, H. Clinicopathologic correlation of up-regulated genes identified using cDNA microarray and real-time reverse transcription-PCR in human colorectal cancer. Cancer Epidemiol. Biomark. Prev. 2005, 14, 437–443. [Google Scholar] [CrossRef]
  215. Wang, D.; Wang, H.; Brown, J.; Daikoku, T.; Ning, W.; Shi, Q.; Richmond, A.; Strieter, R.; Dey, S.K.; DuBois, R.N. CXCL1 induced by prostaglandin E2 promotes angiogenesis in colorectal cancer. J. Exp. Med. 2006, 203, 941–951. [Google Scholar] [CrossRef]
  216. Katoh, H.; Wang, D.; Daikoku, T.; Sun, H.; Dey, S.K.; Dubois, R.N. CXCR2-expressing myeloid-derived suppressor cells are essential to promote colitis-associated tumorigenesis. Cancer Cell 2013, 24, 631–644. [Google Scholar] [CrossRef]
  217. Khan, S.; Cameron, S.; Blaschke, M.; Moriconi, F.; Naz, N.; Amanzada, A.; Ramadori, G.; Malik, I.A. Differential gene expression of chemokines in KRAS and BRAF mutated colorectal cell lines: Role of cytokines. World J. Gastroenterol. 2014, 20, 2979–2994. [Google Scholar] [CrossRef]
  218. le Rolle, A.F.; Chiu, T.K.; Fara, M.; Shia, J.; Zeng, Z.; Weiser, M.R.; Paty, P.B.; Chiu, V.K. The prognostic significance of CXCL1 hypersecretion by human colorectal cancer epithelia and myofibroblasts. J. Transl. Med. 2015, 13, 199. [Google Scholar] [CrossRef]
  219. Hsu, Y.L.; Chen, Y.J.; Chang, W.A.; Jian, S.F.; Fan, H.L.; Wang, J.Y.; Kuo, P.L. Interaction between Tumor-Associated Dendritic Cells and Colon Cancer Cells Contributes to Tumor Progression via CXCL1. Int. J. Mol. Sci. 2018, 19, 2427. [Google Scholar] [CrossRef]
  220. Wang, D.; Sun, H.; Wei, J.; Cen, B.; DuBois, R.N. CXCL1 Is Critical for Premetastatic Niche Formation and Metastasis in Colorectal Cancer. Cancer Res. 2017, 77, 3655–3665. [Google Scholar] [CrossRef]
  221. Triner, D.; Xue, X.; Schwartz, A.J.; Jung, I.; Colacino, J.A.; Shah, Y.M. Epithelial Hypoxia-Inducible Factor 2α Facilitates the Progression of Colon Tumors through Recruiting Neutrophils. Mol. Cell Biol. 2017, 37, e00481-16. [Google Scholar] [CrossRef] [PubMed]
  222. Tosti, N.; Cremonesi, E.; Governa, V.; Basso, C.; Kancherla, V.; Coto-Llerena, M.; Amicarella, F.; Weixler, B.; Däster, S.; Sconocchia, G.; et al. Infiltration by IL22-Producing T Cells Promotes Neutrophil Recruitment and Predicts Favorable Clinical Outcome in Human Colorectal Cancer. Cancer Immunol. Res. 2020, 8, 1452–1462. [Google Scholar] [CrossRef] [PubMed]
  223. Wang, H.; Zhang, B.; Li, R.; Chen, J.; Xu, G.; Zhu, Y.; Li, J.; Liang, Q.; Hua, Q.; Wang, L.; et al. KIAA1199 drives immune suppression to promote colorectal cancer liver metastasis by modulating neutrophil infiltration. Hepatology 2022, 76, 967–981. [Google Scholar] [CrossRef] [PubMed]
  224. Wu, Z.; Neufeld, H.; Torlakovic, E.; Xiao, W. Uev1A-Ubc13 promotes colorectal cancer metastasis through regulating CXCL1 expression via NF-κB activation. Oncotarget 2018, 9, 15952–15967. [Google Scholar] [CrossRef] [PubMed]
  225. Farquharson, A.J.; Steele, R.J.; Carey, F.A.; Drew, J.E. Novel multiplex method to assess insulin, leptin and adiponectin regulation of inflammatory cytokines associated with colon cancer. Mol. Biol. Rep. 2012, 39, 5727–5736. [Google Scholar] [CrossRef]
  226. Cai, L.; Xu, S.; Piao, C.; Qiu, S.; Li, H.; Du, J. Adiponectin induces CXCL1 secretion from cancer cells and promotes tumor angiogenesis by inducing stromal fibroblast senescence. Mol. Carcinog. 2016, 55, 1796–1806. [Google Scholar] [CrossRef]
  227. Urosevic, J.; Blasco, M.T.; Llorente, A.; Bellmunt, A.; Berenguer-Llergo, A.; Guiu, M.; Cañellas, A.; Fernandez, E.; Burkov, I.; Clapés, M.; et al. ERK1/2 Signaling Induces Upregulation of ANGPT2 and CXCR4 to Mediate Liver Metastasis in Colon Cancer. Cancer Res. 2020, 80, 4668–4680. [Google Scholar] [CrossRef]
  228. Zhuang, W.; Niu, T.; Li, Z. MicroRNA miR-145-5p regulates cell proliferation and cell migration in colon cancer by inhibiting chemokine (C-X-C motif) ligand 1 and integrin α2. Bioengineered 2021, 12, 9909–9917. [Google Scholar] [CrossRef]
  229. Lv, Q.Y.; Zou, H.Z.; Xu, Y.Y.; Shao, Z.Y.; Wu, R.Q.; Li, K.J.; Deng, X.; Gu, D.N.; Jiang, H.X.; Su, M.; et al. Expression levels of chemokine (C-X-C motif) ligands CXCL1 and CXCL3 as prognostic biomarkers in rectal adenocarcinoma: Evidence from Gene Expression Omnibus (GEO) analyses. Bioengineered 2021, 12, 3711–3725. [Google Scholar] [CrossRef]
  230. Zhao, Z.W.; Fan, X.X.; Yang, L.L.; Song, J.J.; Fang, S.J.; Tu, J.F.; Chen, M.J.; Zheng, L.Y.; Wu, F.Z.; Zhang, D.K.; et al. The identification of a common different gene expression signature in patients with colorectal cancer. Math. Biosci. Eng. 2019, 16, 2942–2958. [Google Scholar] [CrossRef]
  231. Li, A.; Varney, M.L.; Singh, R.K. Constitutive expression of growth regulated oncogene (gro) in human colon carcinoma cells with different metastatic potential and its role in regulating their metastatic phenotype. Clin. Exp. Metastasis 2004, 21, 571–579. [Google Scholar] [CrossRef]
  232. Bandapalli, O.R.; Ehrmann, F.; Ehemann, V.; Gaida, M.; Macher-Goeppinger, S.; Wente, M.; Schirmacher, P.; Brand, K. Down-regulation of CXCL1 inhibits tumor growth in colorectal liver metastasis. Cytokine 2012, 57, 46–53. [Google Scholar] [CrossRef]
  233. Zhuo, C.; Wu, X.; Li, J.; Hu, D.; Jian, J.; Chen, C.; Zheng, X.; Yang, C. Chemokine (C-X-C motif) ligand 1 is associated with tumor progression and poor prognosis in patients with colorectal cancer. Biosci. Rep. 2018, 38, BSR20180580. [Google Scholar] [CrossRef]
  234. Guil-Luna, S.; Mena, R.; Navarrete-Sirvent, C.; López-Sánchez, L.M.; Khouadri, K.; Toledano-Fonseca, M.; Mantrana, A.; Guler, I.; Villar, C.; Díaz, C.; et al. Association of Tumor Budding with Immune Evasion Pathways in Primary Colorectal Cancer and Patient-Derived Xenografts. Front. Med. 2020, 7, 264. [Google Scholar] [CrossRef]
  235. Mitrovic, B.; Schaeffer, D.F.; Riddell, R.H.; Kirsch, R. Tumor budding in colorectal carcinoma: Time to take notice. Mod. Pathol. 2012, 25, 1315–1325. [Google Scholar] [CrossRef]
  236. Divella, R.; Daniele, A.; Abbate, I.; Bellizzi, A.; Savino, E.; Simone, G.; Giannone, G.; Giuliani, F.; Fazio, V.; Gadaleta-Caldarola, G.; et al. The presence of clustered circulating tumor cells (CTCs) and circulating cytokines define an aggressive phenotype in metastatic colorectal cancer. Cancer Causes Control 2014, 25, 1531–1541. [Google Scholar] [CrossRef]
  237. Tannenbaum, C.S.; Rayman, P.A.; Pavicic, P.G.; Kim, J.S.; Wei, W.; Polefko, A.; Wallace, W.; Rini, B.I.; Morris-Stiff, G.; Allende, D.S.; et al. Mediators of Inflammation-Driven Expansion, Trafficking, and Function of Tumor-Infiltrating MDSCs. Cancer Immunol. Res. 2019, 7, 1687–1699. [Google Scholar] [CrossRef]
  238. Ogata, H.; Sekikawa, A.; Yamagishi, H.; Ichikawa, K.; Tomita, S.; Imura, J.; Ito, Y.; Fujita, M.; Tsubaki, M.; Kato, H.; et al. GROα promotes invasion of colorectal cancer cells. Oncol. Rep. 2010, 24, 1479–1486. [Google Scholar]
  239. Yang, X.; Wei, Y.; Sheng, F.; Xu, Y.; Liu, J.; Gao, L.; Yang, J.; Sun, X.; Huang, J.; Guo, Q. Comprehensive analysis of the prognosis and immune infiltration for CXC chemokines in colorectal cancer. Aging 2021, 13, 17548–17567. [Google Scholar] [CrossRef]
  240. Divella, R.; Daniele, A.; DE Luca, R.; Simone, M.; Naglieri, E.; Savino, E.; Abbate, I.; Gadaleta, C.D.; Ranieri, G. Circulating Levels of VEGF and CXCL1 Are Predictive of Metastatic Organotropismin in Patients with Colorectal Cancer. Anticancer Res. 2017, 37, 4867–4871. [Google Scholar]
  241. Gong, B.; Kao, Y.; Zhang, C.; Sun, F.; Gong, Z.; Chen, J. Identification of Hub Genes Related to Carcinogenesis and Prognosis in Colorectal Cancer Based on Integrated Bioinformatics. Mediat. Inflamm. 2020, 2020, 5934821. [Google Scholar] [CrossRef] [PubMed]
  242. White, J.R.; Lee, J.M.; Young, P.R.; Hertzberg, R.P.; Jurewicz, A.J.; Chaikin, M.A.; Widdowson, K.; Foley, J.J.; Martin, L.D.; Griswold, D.E.; et al. Identification of a potent, selective non-peptide CXCR2 antagonist that inhibits interleukin-8-induced neutrophil migration. J. Biol. Chem. 1998, 273, 10095–10098. [Google Scholar] [CrossRef] [PubMed]
  243. Romanini, J.; Mielcke, T.R.; Leal, P.C.; Figueiredo, C.P.; Calixto, J.B.; Morrone, F.B.; Batista, E.L., Jr.; Campos, M.M. The role of CXCR2 chemokine receptors in the oral squamous cell carcinoma. Investig. New Drugs 2012, 30, 1371–1378. [Google Scholar] [CrossRef] [PubMed]
  244. Goda, A.E.; Koyama, M.; Sowa, Y.; Elokely, K.M.; Yoshida, T.; Kim, B.Y.; Sakai, T. Molecular mechanisms of the antitumor activity of SB225002: A novel microtubule inhibitor. Biochem. Pharmacol. 2013, 85, 1741–1752. [Google Scholar] [CrossRef]
  245. Goda, A.E.; Sakai, T. Molecular insights into the microtubules depolymerizing activity of the IL-8 receptor B antagonist SB225002. Eur. Rev. Med. Pharmacol. Sci. 2022, 26, 3726–3734. [Google Scholar]
  246. Ahuja, S.K.; Murphy, P.M. The CXC chemokines growth-regulated oncogene (GRO) alpha, GRObeta, GROgamma, neutrophil-activating peptide-2, and epithelial cell-derived neutrophil-activating peptide-78 are potent agonists for the type B, but not the type A, human interleukin-8 receptor. J. Biol. Chem. 1996, 271, 20545–20550. [Google Scholar] [CrossRef]
  247. Dwyer, M.P.; Yu, Y.; Chao, J.; Aki, C.; Chao, J.; Biju, P.; Girijavallabhan, V.; Rindgen, D.; Bond, R.; Mayer-Ezel, R.; et al. Discovery of 2-hydroxy-N,N-dimethyl-3-{2-[[(R)-1-(5-methylfuran-2-yl)propyl]amino]-3,4-dioxocyclobut-1-enylamino}benzamide (SCH 527123): A potent, orally bioavailable CXCR2/CXCR1 receptor antagonist. J. Med. Chem. 2006, 49, 7603–7606. [Google Scholar] [CrossRef]
  248. Gonsiorek, W.; Fan, X.; Hesk, D.; Fossetta, J.; Qiu, H.; Jakway, J.; Billah, M.; Dwyer, M.; Chao, J.; Deno, G.; et al. Pharmacological characterization of Sch527123, a potent allosteric CXCR1/CXCR2 antagonist. J. Pharm. Exp. Ther. 2007, 322, 477–485. [Google Scholar] [CrossRef]
  249. Ning, Y.; Labonte, M.J.; Zhang, W.; Bohanes, P.O.; Gerger, A.; Yang, D.; Benhaim, L.; Paez, D.; Rosenberg, D.O.; Nagulapalli Venkata, K.C.; et al. The CXCR2 antagonist, SCH-527123, shows antitumor activity and sensitizes cells to oxaliplatin in preclinical colon cancer models. Mol. Cancer Ther. 2012, 11, 1353–1364. [Google Scholar] [CrossRef]
Figure 1. The role of CXCL1 in gastric cancer pathogenesis. Helicobacter pylori infection induces inflammatory reactions within the gastric wall, leading to gastritis and atrophic mucosa. These sequelae of H. pylori infection are partially mediated by CXCL1. The bacterium upregulates this chemokine’s expression through its virulence factors (Tip-α and CagA) and activation of TLR2. The persistent inflammatory state culminates in gastric cancer. Gastric cancer cells express CXCL1, which in turn amplifies its own production. Other sources of CXCL1 in gastric tumors include tumor-associated macrophages (TAMs), cancer-associated fibroblasts (CAFs), and lymphatic endothelial cells (LECs). CXCL1 participates in tumorigenic processes, promoting the migration of gastric cancer cells. Furthermore, CXCL1 facilitates the recruitment of granulocytic myeloid-derived suppressor cells (G-MDSCs) and neutrophils. CXCL1 also influences LECs, resulting in lymphangiogenesis and the migration of gastric cancer cells to lymphatic vessels, ultimately leading to lymph node metastasis.
Figure 1. The role of CXCL1 in gastric cancer pathogenesis. Helicobacter pylori infection induces inflammatory reactions within the gastric wall, leading to gastritis and atrophic mucosa. These sequelae of H. pylori infection are partially mediated by CXCL1. The bacterium upregulates this chemokine’s expression through its virulence factors (Tip-α and CagA) and activation of TLR2. The persistent inflammatory state culminates in gastric cancer. Gastric cancer cells express CXCL1, which in turn amplifies its own production. Other sources of CXCL1 in gastric tumors include tumor-associated macrophages (TAMs), cancer-associated fibroblasts (CAFs), and lymphatic endothelial cells (LECs). CXCL1 participates in tumorigenic processes, promoting the migration of gastric cancer cells. Furthermore, CXCL1 facilitates the recruitment of granulocytic myeloid-derived suppressor cells (G-MDSCs) and neutrophils. CXCL1 also influences LECs, resulting in lymphangiogenesis and the migration of gastric cancer cells to lymphatic vessels, ultimately leading to lymph node metastasis.
Cells 12 01406 g001
Table 1. The effect of CXCL1 expression level on the survival of patients with head and neck cancer.
Table 1. The effect of CXCL1 expression level on the survival of patients with head and neck cancer.
Type of TumorEffect on the Survival at High CXCL1 ExpressionNumber of Patients in the StudyNotesReferences
Head and neck cancer: squamous cell carcinomaWorse prognosis491OS, DFS
Data from the Cancer Genome Atlas (TCGA) dataset
[31,52]
Head and neck cancer: squamous cell carcinomaWorse prognosis494OS, DFS/PFS
Data from TCGA dataset
[51,52]
Head and neck cancer: squamous cell carcinomaWorse prognosis499OS,
statistically insignificant difference in RFS
Data from Kaplan–Meier plotter
[26,27]
Head and neck cancer: larynx squamous cell carcinomaWorse prognosis135OS[33]
DFS—disease-free survival; OS—overall survival; PFS—progression-free survival; RFS—relapse-free survival; red color—worse prognosis.
Table 2. The effect of CXCL1 expression level on the survival of patients with esophageal cancer.
Table 2. The effect of CXCL1 expression level on the survival of patients with esophageal cancer.
Type of TumorEffect on the Survival at High CXCL1 ExpressionNumber of Patients in the StudyNotesReferences
Esophageal cancer: esophageal squamous cell carcinomaWorse prognosis141OS,[56]
Esophageal cancer: esophageal squamous cell carcinomaBetter prognosis877OS
Data from the Kaplan–Meier Plotter database (http://kmplot.com access date: 16 April 2022)
[57]
Esophageal carcinomaNo significant impact on prognosis92RFS and OS
Data from the gene expression profiling interactive analysis (GEPIA) database
[65]
OS—overall survival; RFS—relapse-free survival; red color—worse prognosis; blue color—better prognosis.
Table 3. The effect of CXCL1 expression level on the survival of patients with gastric cancer.
Table 3. The effect of CXCL1 expression level on the survival of patients with gastric cancer.
Type of TumorEffect on the Survival at High CXCL1 ExpressionNumber of Patients in the StudyNotesReferences
Gastric cancerWorse prognosis98OS[98]
Gastric cancerWorse prognosis56Cumulative survival,
CXCL1 in the tumor, as well as plasma CXCL1 level
[97]
Gastric cancerBetter prognosis34OS[96]
Gastric cancerWorse prognosis572OS,
Data from the public database
[108]
Gastric cancerWorse prognosis155OS[107]
Gastric cancerWorse prognosis127OS[115]
Gastric cancerWorse prognosis590OS[101]
Gastric cancerWorse prognosis100OS[105]
Gastric cancerWorse prognosis263OS,
Only for stage I patients
[112]
Gastric cancerWorse prognosis105OS[109]
Gastric cancerWorse prognosis72OS[99]
OS—overall survival; red color—worse prognosis; blue color—better prognosis.
Table 4. The effect of CXCL1 expression level on the survival of patients with liver cancer.
Table 4. The effect of CXCL1 expression level on the survival of patients with liver cancer.
Type of TumorEffect on the Survival at High CXCL1 ExpressionNumber of Patients in the StudyNotesReferences
Liver cancerWorse prognosis346OS,
Data from TCGA dataset
[52,151]
Liver cancer: HCCWorse prognosis182OS
Data from the Kaplan–Meier plotter database
[152]
Liver cancer: HCCWorse prognosis48OS[131]
Liver cancer: HCCWorse prognosis119OS, DFS[143]
Liver cancer: HCCWorse prognosis259OS, RFS
Patients with a high CXCL1 expression in a tumor together with a high CXCR2 expression
[147]
DFS—disease-free survival; OS—overall survival; RFS—relapse-free survival; red color—worse prognosis.
Table 5. The effect of CXCL1 expression level on the survival of patients with cholangiocarcinoma.
Table 5. The effect of CXCL1 expression level on the survival of patients with cholangiocarcinoma.
Type of TumorEffect on the Survival at High CXCL1 ExpressionNumber of Patients in the StudyNotesReferences
CholangiocarcinomaBetter prognosis165OS[155]
CholangiocarcinomaWorse prognosis18RFS
No effect at OS,
Data from the GEPIA database
[65]
OS—overall survival; RFS—relapse-free survival; red color—worse prognosis; blue color—better prognosis.
Table 6. The effect of CXCL1 expression level on the survival of patients with pancreatic cancer.
Table 6. The effect of CXCL1 expression level on the survival of patients with pancreatic cancer.
Type of TumorEffect on the Survival at High CXCL1 ExpressionNumber of Patients in the StudyNotesReferences
Pancreatic cancerWorse prognosis160OS[163]
Pancreatic cancer: pancreatic adenocarcinomaNo significant impact on prognosis178OS, DFS[165]
Pancreatic adenocarcinomaNo significant impact on prognosis90OS, RFS
Data from the GEPIA database
[65]
DFS—disease-free survival; OS—overall survival; RFS—relapse-free survival; red color—worse prognosis.
Table 7. The effect of CXCL1 expression level on the survival of patients with colorectal cancer.
Table 7. The effect of CXCL1 expression level on the survival of patients with colorectal cancer.
Type of TumorEffect on the Survival at High CXCL1 ExpressionNumber of Patients in the StudyNotesReferences
Colorectal cancerWorse prognosis62OS[238]
Colorectal cancerWorse prognosis91RFS, analysis only in stage III patients. In other stages, CXCL1 expression is not related to prognosis[204]
Colorectal cancerNo significant impact on prognosis163RFS, OS
analysis only in stage II patients
[204]
Colorectal cancerNo significant impact on prognosis270OS, Data from the GEPIA database[65,206]
Colorectal cancerNo significant impact on prognosis362RFS, OS
Data from the GEPIA database
[65,239]
Colorectal cancerWorse prognosis276OS, DFS[233]
Colorectal cancerNo significant impact on prognosis125OS, RFS[212]
Colorectal cancerWorse prognosis45OS,
only stage IV patients
[218]
Colorectal cancerNo significant impact on prognosis70OS,
Only stage II and III patients
[218]
Colorectal cancer: colon cancerBetter prognosis438OS, data from TCGA[52,209,213]
Colorectal cancer: colon adenocarcinomaNo significant impact on prognosis171OS, RFS[209]
Colorectal cancer: rectal adenocarcinomaWorse prognosis304OS
Data from the TCGA
database
[52,229]
DFS—disease-free survival; OS—overall survival; RFS—relapse-free survival; red color—worse prognosis; blue color—better prognosis.
Table 8. CXCR2 inhibitors in selected clinical trials. Source: ClinicalTrials.gov NIH U.S. National Library of Medicine website.
Table 8. CXCR2 inhibitors in selected clinical trials. Source: ClinicalTrials.gov NIH U.S. National Library of Medicine website.
Disease for Which the Drug Is Being TestedDrug NameClinical Trial PhaseClinicalTrials.gov
Identifier
Metastatic Castration-Resistant Prostate CancerAZD5069I and IINCT03177187
Myelodysplastic SyndromesSX-682INCT04245397
Pancreatic Ductal AdenocarcinomaSX-682INCT04477343
Melanoma Stage III and Stage IVSX-682INCT03161431
Inflammatory ResponseRIST4721INCT04105959
Chronic Obstructive Pulmonary Disease (COPD)Danirixin (GSK1325756)INCT01453478
COPDDanirixin (GSK1325756)INCT03136380
COPDDanirixin (GSK1325756)IINCT03250689
COPDDanirixin (GSK1325756)IINCT02130193
COPDNavarixin (SCH 527123, MK-7123)IINCT01006616
COPDQBM076IINCT01972776
InfluenzaDanirixin (GSK1325756)IINCT02469298
InfluenzaDanirixin (GSK1325756)IINCT02927431
Respiratory Syncytial Virus (RSV) InfectionsDanirixin (GSK1325756)INCT02201303
COPDSB-656933INCT00504439
Cystic FibrosisSB-656933IINCT00903201
Type 1 DiabetesLadarixinIINCT05035368
Type 1 DiabetesLadarixinIIINCT04628481
Bullous PemphigoidDF2156AIINCT01571895
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Korbecki, J.; Bosiacki, M.; Barczak, K.; Łagocka, R.; Chlubek, D.; Baranowska-Bosiacka, I. The Clinical Significance and Role of CXCL1 Chemokine in Gastrointestinal Cancers. Cells 2023, 12, 1406. https://doi.org/10.3390/cells12101406

AMA Style

Korbecki J, Bosiacki M, Barczak K, Łagocka R, Chlubek D, Baranowska-Bosiacka I. The Clinical Significance and Role of CXCL1 Chemokine in Gastrointestinal Cancers. Cells. 2023; 12(10):1406. https://doi.org/10.3390/cells12101406

Chicago/Turabian Style

Korbecki, Jan, Mateusz Bosiacki, Katarzyna Barczak, Ryta Łagocka, Dariusz Chlubek, and Irena Baranowska-Bosiacka. 2023. "The Clinical Significance and Role of CXCL1 Chemokine in Gastrointestinal Cancers" Cells 12, no. 10: 1406. https://doi.org/10.3390/cells12101406

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop