Next Article in Journal
Impact of Post-Operative Infection after CABG on Long-Term Survival
Next Article in Special Issue
Intraspinal and Intracranial Neurotuberculosis, Clinical and Imaging Characteristics and Outcomes in Hospitalized Patients: A Cohort Study (2000–2022)
Previous Article in Journal
The Comparison between the Composition of 100% Autologous Serum and 100% Platelet-Rich Plasma Eye Drops and Their Impact on the Treatment Effectiveness of Dry Eye Disease in Primary Sjogren Syndrome
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Interleukins (Cytokines) as Biomarkers in Colorectal Cancer: Progression, Detection, and Monitoring

1
Department of Biosciences, COMSATS University Islamabad (CUI), Islamabad 44000, Pakistan
2
Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, M. Strzody 9, 44-100 Gliwice, Poland
3
Centre for Organic and Nanohybrid Electronics, Silesian University of Technology, Konarskiego 22B, 44-100 Gliwice, Poland
4
Joint Doctoral School, Silesian University of Technology, Akademicka 2A, 44-100 Gliwice, Poland
5
Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Al Sharqia, Egypt
6
Department of Molecular Genetics, Faculty of Biology, Technical University of Kaiserslautern, Paul-Ehrlich Str. 24, 67663 Kaiserslautern, Germany
7
Faculty of Medicine and Pharmacy, University of Oradea, P-ta 1 Decembrie 10, 410087 Oradea, Romania
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2023, 12(9), 3127; https://doi.org/10.3390/jcm12093127
Submission received: 18 March 2023 / Revised: 18 April 2023 / Accepted: 20 April 2023 / Published: 25 April 2023
(This article belongs to the Special Issue Clinical Aspects of Infectious Diseases)

Abstract

:
Cancer is the primary cause of death in economically developed countries and the second leading cause in developing countries. Colorectal cancer (CRC) is the third most common cause of cancer-related deaths worldwide. Risk factors for CRC include obesity, a diet low in fruits and vegetables, physical inactivity, and smoking. CRC has a poor prognosis, and there is a critical need for new diagnostic and prognostic biomarkers to reduce related deaths. Recently, studies have focused more on molecular testing to guide targeted treatments for CRC patients. The most crucial feature of activated immune cells is the production and release of growth factors and cytokines that modulate the inflammatory conditions in tumor tissues. The cytokine network is valuable for the prognosis and pathogenesis of colorectal cancer as they can aid in the cost-effective and non-invasive detection of cancer. A large number of interleukins (IL) released by the immune system at various stages of CRC can act as “biomarkers”. They play diverse functions in colorectal cancer, and include IL-4, IL-6, IL-8, IL-11, IL-17A, IL-22, IL-23, IL-33, TNF, TGF-β, and vascular endothelial growth factor (VEGF), which are pro-tumorigenic genes. However, there are an inadequate number of studies in this area considering its correlation with cytokine profiles that are clinically useful in diagnosing cancer. A better understanding of cytokine levels to establish diagnostic pathways entails an understanding of cytokine interactions and the regulation of their various biochemical signaling pathways in healthy individuals. This review provides a comprehensive summary of some interleukins as immunological biomarkers of CRC.

1. Introduction

Cancer is a condition where cells proliferate uncontrollably [1,2]. It is the leading cause of death in economically developed countries and the second leading cause of death in developing countries [3]. Colorectal cancer (CRC) is the third most common cause of cancer death globally, with an estimated 2.2 million new cases and 1.1 million deaths expected over the next decade [4]. CRC accounts for 9.2% of cancer-related deaths, making it the second leading cause of cancer death [5,6,7]. Both genetic and environmental factors play a role in CRC causation [8]. Chronic colitis due to inflammatory bowel disease (IBD) is also associated with an increased risk of CRC [9]. CRC has a poor prognosis, and there is a critical need for new diagnostic and prognostic biomarkers to reduce CRC-related deaths [10,11]. Cytokine networks are largely involved in the prognosis and pathogenesis of CRC [12]. Tumors express cytokines with both antitumor and pro-tumor properties [13]. Cytokines’ ability to stimulate oncogenic signaling has shifted research focus towards their role in promoting cell proliferation and survival during tumorigenesis [14]. Immune cells activated in response to the tumor produce and release growth factors and cytokines to regulate the inflammatory environment in tumor tissues [15]. These inflammatory cytokines direct DNA damage in the epithelium [16]. They imply the pathogenesis and prognosis of CRC [12]. Interleukins have distinct roles in CRC progression through tumorigenesis [14] and are also involved in tumor growth, cancer cell invasion, and metastasis, while also inhibiting cancer through complex pathways [15]. In recent years, interleukins have gained significant attention due to their distinct roles in providing a new and promising strategy for CRC treatment [17].
Interestingly, interleukins are not the only biomarkers for CRC. Since the gut microbiome has been shown to play a crucial role in the development of CRC, several studies have investigated the use of gut microbes as biomarkers for CRC [18,19,20]. The presence of Fusobacterium nucleatum and Bacteroides fragilis in tumors has been reported in 43% and 24% of patients, respectively. The detection of these bacterial species has been found to correlate with the overall bacterial load, but further analysis of microbial signatures via diversity profiling suggests that their detection may be indicative of a specific microbial profile [21]. Consequently, the presence of F. nucleatum and clbA+ bacteria in stool samples can be used as a predictive marker for CRC with a relatively high specificity and moderate sensitivity [22]. It is important to note that these non-invasive screening approaches are still under development and more research is needed to validate their results in larger patient populations and confirm their efficacy compared to current screening methods. Several studies have also investigated the use of miRNA as biomarkers for CRC. For example, a study found that a panel of miRNAs (e.g., miR-21, miR-31, miR-146a, and miR-192) had a sensitivity and a specificity for detecting CRC [23]. Even though these studies demonstrate the potential of miRNA as a non-invasive biomarker for CRC, more research is still needed to validate these findings in larger patient populations [24,25,26]. The type of cancer and stage of the disease can impact the miRNA signature. Furthermore, the methods used to detect miRNA, such as RT-qPCR and microarray analysis, can also contribute to inconsistencies in results [24,27].
The point-of-care (POC) detection of cancer biomarkers provides an effective means for early-stage diagnosis. The cytokines, especially IL-6 and IL-8, have been used as potential biomarkers allowing the diagnosis of various benign oral lesions from malignant ones. Various electrochemical and optical biosensors have been developed to detect interleukins [28]. Studies on clinical units and mouse models helped to reveal many cytokines that have a high correlation with specific diseases, overlaying an efficient way to develop Biology Collector (BIOCO) in clinics to promote the research in cytokine development platforms [29]. Cytokines can play a key role in disease management and diagnosis as an immune mediator, similarly to pathophysiology-based sepsis, where pro- and anti-inflammatory cytokines have a double-edged function [30]. Recent studies have focused on using molecular testing to guide targeted treatments for CRC patients, but introducing novel molecular tests into routine clinical practices remains a huge challenge [31]. Interleukins, which can be detected in blood circulation even in the case of localized tumors, have the potential to serve as biomarkers for detecting cancers, predicting disease outcomes, and managing therapeutic choices [32]. The detection of specific cytokines in blood can cost-effectively and non-invasively assist in the detection of cancer, as well as in determining the appropriate therapy and monitoring the progression of the disease [31,32,33,34]. This review aims to assess the role of interleukins as biomarkers for the early possible detection of colorectal cancer by searching advanced published literature and studies regarding immunological biomarkers’ expressions and roles in colorectal cancer using keywords; colorectal cancer, gastric cancer, cytokine network, diagnostics, and monitoring.

2. Molecular Pathways and Cytokine Role in CRC

The tumor microenvironment (TME) plays an important role in the initiation and growth of tumors into metastatic states [35]. Tumor cells secrete cytokines that stimulate self-proliferation, drug resistance, and activation and differentiation of other cells in the tumor microenvironment [36,37]. Neutrophils are triggered by complement immunity and release IL-1β, which can activate myeloid cells to produce IL-6 and increase the IL-17A response. IL-33 can be secreted by vascular endothelial cells and tumor cells [17]. The host’s immune response causes chronic inflammation from time to time, which leads to tumor growth primarily due to interactions between tumors, immune, and other cells moderated by cytokine [38,39]. Cytokines alert immune cells in the presence of tissue damage and infections to stimulate the cells to release more cytokine signals leading to chronic inflammation [38]. Chronic inflammation promotes diverse cytokine secretion during various stages and types of cancer [32]. It causes DNA damage and DNA mutation by reactive oxygen and nitrogen species, along with the alteration of the epigenome of the cells [40,41].
Immunological biomarkers can be predictive, prognostic, or both [42]. Biomarkers are genes, gene products, cells, enzymes, molecules, or hormones that can be detected in tissues and body fluids (blood, urine, etc.) [43]. Cancer biomarkers are usually produced by the tumor or other body cells in response to the tumor [44]. They can be used for early cancer detection and patient state prediction [45]. Certain biomarkers can reveal genetic changes in cancerous cells, such as gene rearrangements or amplifications, mutations, and cell division, enabling cancer detection and response prediction to various treatments [46,47]. Prognostic immune cells include tumor-infiltrating lymphocytes, eosinophils, neutrophilic granulocytes, macrophages, and dendritic cells [48,49,50,51]. The epithelium of a tumor activates stromal cells to release cytokines, such as transforming growth factor, and cell signaling factors to develop a microenvironment for tumor progression and metastasis [52,53,54].
The development of colorectal cancer is also caused by specific mutations in oncogenes, tumor-suppressor genes, and genes associated with DNA repair mechanisms. However, approximately 70% of colorectal cancer cases follow a specific mutation and are transformed into a specific morphological sequence that starts with polyps and abnormal crypts, which then develop into early adenomas. The adenoma then progresses to advanced adenomas, eventually leading to colorectal cancer [55]. Therefore, genome instability is a fundamental feature of CRC. The pathogenic mechanisms leading to this situation can be divided into three different pathways (Figure 1): chromosomal instability (CIN), microsatellite instability (MSI), and CpG island methylator phenotype (CIMP) [56].
Mesenchymal cells of the gut, including endothelial cells, smooth muscle cells, mucosal immune cells, and subepithelial myofibroblasts, are combined with the tumor epithelial cells to regulate TME for the progression of CRC [57,58]. Table 1 represents main cancer cell types with the signaling pathways and target cells.
Many polymorphisms and somatically altered genes in CRC affect the KRAS- (kirsten rat sarcoma viral oncogene homolog), MYC- (cellular myelocytomatosis oncogene), Wnt-, mitogen-activated protein kinase (MAPK)-, or TGF-β/bone morphogenetic protein (BMP)-signaling pathways. For example, the nuclear factor κB (NF-κB), JNK, AP-1, and p38 MAPK pathways are activated by IL-1 receptor cytokine in CRC along with the activation of the Wnt signaling pathway by phosphorylation of GSK3β [12].

3. Advancements in Cytokine Detection and Monitoring Clinically

The study of cytokine secretion can lead to improved understanding of disease mechanisms and the development of new treatments. Elevated cytokine levels are a hallmark of many diseases such as diabetes, sepsis, cardiovascular diseases, neurodegenerative diseases, and cancers, and can be used as potential biomarkers [64,65,66,67,68]. A study found that a combination of IFN-γ, IP-10, ferritin, and 25-hydroxyvitamin D can be used to diagnose pediatric tuberculosis and to differentiate between TB and latent TB infection [69]. Inflammatory cytokines such as IL-6, IL-10, IL-1, TNF-α, and TGF-β are linked to the progression of heart failure and chronic kidney dysfunction, which are major global health problems with significant economic impacts [70]. Furthermore, inflammatory bowel diseases such as ulcerative colitis and Crohn’s disease result from an imbalance in pro- and anti-inflammatory cytokine interactions [29]. However, excessive cytokine release into the bloodstream from immune cells can cause cytokine release syndrome during immunotherapy and infections [71,72]. This makes detecting cytokines challenging due to their low concentration, instability, and complex networks [73]. Cytokine imbalances can also cause illnesses and prompt the need for precise and early treatment in severe conditions, so there is a demand for accurate, fast, and sensitive cytokine screening methods [74,75].
Common methods for cytokine quantification in clinical practice include ELISA and ELISpot [76]. Due to their crucial role in disease and human health, researchers are striving to develop tools for sensitive, multiplex cytokine detection [77]. For instance, recent studies have explored deployable devices with immunosensors on fiber optics [78] and stainless steel [79] for cytokine monitoring in mouse brain and spinal cords at pg/mL levels. An impedance aptasensor was developed for a highly sensitive detection of IL-6, with a detection limit of 1.6 pg/mL and a linear response of 5 pg/mL to 100 ng/mL [80]. A microfluidic technology based on single plex was designed to eliminate cross-reactivity and detect chemokines and cytokines in human and mouse samples [81]. Another chemiluminescent nanozyme immunoassay was developed for simultaneous detection of two chicken cytokines (IL-4 and IFN-γ) in serum samples with a range of 0.01–60 ng/mL for IFN-γ and IL-4, respectively, and detection limits of 2.9 pg/mL for IFN-γ and 3.2 pg/mL for IL-4 [82]. Clinical detection of serum biomarkers such as CA 19–9 and CA724 has a low specificity and sensitivity in gastric cancer diagnosis [83]. Studies show that the sensitivity, accuracy, and specificity of interleukins are as good as that of other markers such as carcinoembryonic antigen (CEA) and CA 19–9 [84]. This suggests that IL-6 may be a reliable biomarker for gastric cancer detection and diagnosis.
IL detection is commonly used as a biomarker for infections in clinical applications [85,86]. The most widely used tumor marker is a combination of ILs and CEA [87]. Tumor markers, found in body fluids, are widely used for early diagnosis, prognostic evaluation, and treatment monitoring of tumors [88,89], but CEA lacks specificity [90]. Combining inflammatory factors and tumor markers has shown a high potential in diagnosing pancreatic and CRC [91,92]. IL detection has also been analyzed for its individual role in the diagnosis of lung cancer and predicting lymph node metastasis [93]. They have shown promising results as a cancer biomarker in various types of cancer including gastric cancer, pancreatic cancer, and CRC. The use of ILs combined with other tumor markers such as CEA has the potential to enhance the accuracy of cancer diagnosis. In the case of thyroid cancer, the combination of IL-13 and IL-8 has been found to be effective in identifying the disease [94]. However, further research is needed to fully understand the genetic basis of interleukin levels as a cancer risk factor and to account for other factors that can impact the levels of interleukins in the body [95].
Alternation in the expression of interleukins such as IL-17, IL-22, and IL-1ß has been associated with various types of cancer [96,97]. Interleukins regulate the tumor microenvironment and are involved in tumor development and progression [98,99]. Not only locally, but their actions also occur distantly through circulation [100]. Interleukins promote cancer development by counteracting the immune response, mobilizing stromal, and immunosuppressive cells that support the tumor, inducing angiogenesis, and altering the response to therapeutic agents [101,102]. Their production is also triggered due to changes induced by cancer, alterations in metabolism, cell death, oxygen deficiency, and usage of anticancer drugs [102]. Figure 2 shows the immune response of cancer with cytokine release. The control of widespread cancer includes effective tools for the betterment of cancer burden by knowledge, early detection, suitable therapy along with a regular follow-up, and forecast measures by using cancer biomarkers [103]. The analysis of cytokines along with cancer-specific biomarkers has been put forward to improve cancer detection [32].

4. Cytokines’ Role in CRC

Some cytokines (other than interleukins) are also associated with the immune regulation of tumor cells and are highly expressed as compared to normal cells. They include forkhead box P3 (FOX P3), tumor necrosis factor-α (TNF-α), and interferon-gamma (IFN-γ).

4.1. Forkhead Box P3 (FOXP3)

The FoxP3 gene is expressed in regulatory T cells and is associated with cell development, transcription regulation, and DNA repair [104,105,106]. In addition to its basic role in immune responses, FOXP3 also plays a significant role in cancer development [107]. A high level of FOXP3 expression was observed in tumor cells compared to tumor-surrounding tissues, as detected through an immunohistochemistry assay [108]. FOXP3 levels are also higher in colorectal cancer tissues than in normal colorectal tissues [109], and its expression is associated with a poor prognosis compared to patients with low FOXP3 expression [110]. However, a high level of FOXP3 in tumor cells is associated with longer and disease-free survival [108]. Intra-tumoral CD4+ and FOXP3+ cell infiltration can be the most meaningful predictive factor in CRC patients [111]. Studies show that FOXP3 expression by cancer cells results in the secretion of cytokines such as IL-10 and TGFβ into the tumor microenvironment, which suppresses immunity [112].

4.2. Tumor Necrosis Factor-α (TNF-α)

Tumor necrosis factor-α (TNF-α) is a cytokine produced by macrophages and involved in various immune regulations [113]. It promotes the process of epithelial-to-mesenchymal transition (EMT) in colorectal cancer, thus promoting the metastasis of colorectal cancer [114,115,116]. Single-nucleotide polymorphisms in TNF-α genes are reportedly associated with the prognosis, therapy response, and survival of cancer patients [117]. High TNF-α gene expression is associated with Stage III and IV neoplasms compared to earlier tumor stages, and TNF-α expression is increased in the serum of CRC patients [118,119]. Statistical analysis shows that TNF-α mRNA expression levels are significantly higher in CRC compared to normal CRC tissue, and CRC patients with low TNF-α serum levels have a significantly higher survival rate compared to patients with high levels of TNF-α [118,120]. The examination of TNF-α levels in plasma can be used as a diagnostic factor for CRC instead of using other invasive tests [121].

4.3. Interferon-Gamma (IFN-γ)

The genetic variations in interferon-gamma (IFN-γ) and its receptor (IFN-γR) subunits are strongly associated with the risk of colorectal cancer and patient survival after diagnosis [122]. IFN-γ is a major activator of macrophages and an inducer of Class II major histocompatibility complex (MHC) molecules [123], with immune regulatory, antiviral, and antitumor properties [124,125]. Studies have shown that the deficiency of IFN-γ or its receptor promotes the development of colorectal cancer, whereas its specific expression activates innate immunity and inhibits tumorigenesis [122,124]. The specific expression of IFN-γ activates innate immunity and inhibits tumorigenesis [124] However, IFN-γ signaling can also compromise antitumor immunity by inducing immune checkpoint inhibitory molecules on T and tumor cells [125]. Furthermore, IFN-γ acts as a cytotoxic cytokine and initiates apoptosis in tumor cells [126]. The IFN-γ/Janus Kinase 2 (JAK)/signal transducer and activators of transcription (STAT) signaling pathway has been shown to induce programmed death-ligand 1 (PD-L1) expression in myeloid leukemia cells, pancreatic, and gastric cancer [127,128]. Increased PD-L1 expression levels have been found to be associated with a poor prognosis in patients with CRC [129]. Previous studies have also reported that PD-L1 expression on tumor-infiltrating immune cells is correlated with the survival of patients with CRC [130].

5. Interleukins in Colorectal Cancer

There are various interleukin families involved in CRC progression that have been studied as biomarkers (Table 2). They play different roles in colorectal cancer. For example, IFN-γ, interleukin-12 (IL-12), IL-15, IL-17F, and IL-18 inhibit CRC development [131,132,133,134]. On the other hand, IL-4, IL-6, IL-8, IL-11, IL-17A, IL-22, IL-23, IL-33, TNF, TGF-β, and vascular endothelial growth factor (VEGF) are pro-tumorigenic genes [135,136]. The contribution of IL-1, IL-9, IL-10, IL-21, and granulocyte-macrophage colony-stimulating factor and sargramostim (GM-CSF) to intestinal cancer remains unclear [135]. Figure 3 shows certain cytokines and their role in CRC.

5.1. Interleukin-1β

IL-1β is activated by immune response receptors to induce inflammatory responses [177]. As a pro-inflammatory cytokine, IL-1β is secreted by macrophages to promote cell proliferation [178,179]. Together with TNF-α, IL-1β is considered an “alarm cytokine” that triggers inflammatory responses by inducing other pro-inflammatory genes [180]. Polymorphisms in the IL-1β gene increase the risk of colon cancer development [181], while single-nucleotide polymorphisms (SNPs) associated with a high expression of IL-1Ra lead to better survival in patients with advanced CRC [182]. Mutations in the Nucleotide Binding Oligomerization Domain (NOD2) are also linked to severe forms of Cluster of Differentiation (CD) that secrete IL-1β, indicating its potential role in CRC progression [183]. Studies suggest that IL-1β promotes colon tumor growth by activating cancer stem cell (CSC) self-renewal and epithelial-to-mesenchymal transition (EMT) through the transcription factor Zinc Finger E-box binding homeobox 1 (Zeb1) [184]. Therefore, IL-1β and Zeb1 could be potential therapeutic targets for colon cancer treatment. In one study, IL-1β was found to be significantly increased in CRC tissues compared to normal tissues, leading to the hypothesis that IL-1β plays a tumorigenic role in CRC and is associated with a higher rate of survival [185].

5.2. Interleukin-17

The identification of only six members of the IL-17 family has been achieved, i.e., IL-17A, IL-17B, IL-17C, IL-17D, IL-17E, and IL-17F [186]. However, the extensive study of only IL-17A and IL-17F has been performed related to CRC development [187]. IL-17 is being considered a promoter in the progression of colorectal cancer [188]. Studies show that the serum levels of IL-17A were elevated in CRC patients in comparison to healthy individuals [189] or in the circulating tumor cells which also predicted poor survival [190]. Furthermore, its gene expression is reported to be higher in tumor tissues compared to normal mucosa [171]. IL-17A expression heightens the adenoma-to-carcinoma sequence (mutational activation of cancer genes) in the intestinal epithelium of CRC patients [191]. Poor prognosis has been shown when there is a high expression of these genes associated with Th17 in CRC tissues [171]. In vitro, IL-17 and TNF-α synergistically promote carcinogenesis by stimulating glycolysis and growth factor production by CRC cells [192]. The study showed that CD4+ T-cell-derived IL-17 promotes tumorigenesis in the intestine of mice [193]. IL-17-producing cells may facilitate the development of CRC by assisting angiogenesis through the stimulation of VEGF production by cancer cells [194]. These cells have seen an increase in the intestinal mucosa of CRC patients due to microbial misbalance, which indicates that they can be a sensitive prognostic indicator for CRC [195]. In an experiment on serum and tissues of 99 samples and 37 controls, high IL-17 expression was seen, predicting IL-17 as a valuable tumor marker in CRC patients [196]. The variant of IL-17A can be utilized as a screening marker to assess CRC risk while its expression can be used as a biomarker for early CRC detection [197]. Moreover, an elevated level of Th17 cells was found in almost 80% of sporadic colon cancer tissues of humans, which indicates that IL-17 expression can be among potential biomarkers as prognostic entities for future developments in sporadic CRC [198].

5.3. Interleukin-22

Interleukin-22 has recently arisen as a novel part of CRC advancement as Th22 aggregation in patients showed relatedness with CRC advancement [199]. IL-22 in CRC tissue and serum or CRC tissue can be a prediction for the poor endurance of patients [14] elevating resistance to chemotherapy [200]. The polymorphisms in the IL-22 promoter are also linked with CRC risk [59]. Results of many studies suggest that IL-22 is involved in colon tumor maintenance as the analysis of IL-22 in human colon cancer showed that IL-22 mRNA expression in tumor tissue was more than two-fold higher than in normal tissue [201]. Their enhanced expression is related to the inflammation of colon mucosa in patients with gut infection or bowel diseases [202]. Excessive IL-22 in the cancer microenvironment leads to tumor growth with the activation of the STAT3 pathway [203], and the epigenetic activation of genes with a STAT3-dependent pathway maintains the CRC stem cells [59]. Levels of IL-22 in tumor tissues and blood are associated with chemoresistance and indicate a poor prognosis for patients having chemotherapy, so IL-22 may be a useful prognostic biomarker for CRC patients [200]. RORγt (necessary for IL-22 expression) and IL-17A expression (co-expressed with IL-22 sometimes) are associated with a bad prognosis of human CRC [171]. Modulation of IL-22 expression can also be due to various dietary components such as high fatty diet, and cruciferous vegetables; along with the microbiome, which has a substantial influence on IL-22 forming cells in CRC [14].

5.4. Interleukin-6

Interleukin-6 (IL-6) is rapidly produced in response to tissue sprains and infections, contributing to host defense through immune reactions [201]. Studies on mice with colitis-associated cancer have found that treatment with anti-IL-6 receptor antibodies reduces the incidence of cancer, suggesting that IL-6 may be a therapeutic target for colorectal cancer (CRC) [202]. IL-6 plays a central role in the development of colonic cancer [58], with its expression significantly elevated in CRC tissues compared to non-cancerous cells and associated with an increased risk of relapse [202,203]. Several meta-analyses have indicated that serum IL-6 may be a potential biomarker for the diagnosis of CRC, and circulating IL-6 in plasma is also increased in patients with CRC [154,204,205]. Targeting the IL-6/STAT3 pathway has been proposed as a possible strategy for CRC therapy, as its expression can be an important factor in establishing prognostics for clinical decisions [206,207,208]. Recently, many therapeutic strategies have been developed that target the IL-6/STAT3 pathway for the treatment of CRC [209]. The IL-6/JAK/STAT3 signaling pathway drives the metastasis, proliferation, survival, and invasiveness of tumor cells in the tumor microenvironment by suppressing the antitumor immune response. Thus, targeting this pathway can directly inhibit cancer cell growth and stimulate the antitumor immunity [210]. Cancer-associated fibroblasts (CAFs) induce IL-6 to activate the Jak1-STAT3 pathway in gastric cancer cells by paracrine signaling. This allows tumor cells to progressively resist apoptosis, increasing their survival and resistance to chemotherapy. The humanized monoclonal anti-IL-6R antibody Tocilizumab (an FDA-approved drug) inhibits the activation of the Jak1-STAT3 signaling pathway, increasing the effectiveness of chemotherapeutic drugs [211]. Figure 4 illustrates its activity.

5.5. Interleukin-23

IL-23 is a heterodimeric type 1 cytokine composed of IL-12/p40 and p19 subunit, which is chiefly secreted by macrophages, monocytes, and activated dendritic cells and is vital in mucosal immunity [212,213]. Elevated levels of IL-23 have been found in colon adenocarcinoma, and it promotes tumor growth by blocking cytotoxic T cells and initiating pro-inflammatory responses [214]. IL-23 mRNA has been reported to be increased in various human tumors, correlating with a poor prognosis [214,215]. IL-23 influences tumor cells via T-cell responses by positively affecting the STAT3 activity in tumor growth, elevating TH17 activity and regulatory T cells (Tregs) [216]. Reports showed that IL-23 is highly expressed in tumor tissues of humans from other organs, and its expression is correlated with a poor prognosis [171,214]. IL-23R protein has been detected in the cancerous colorectal cell line SW-480, and its expression is progressively elevated from normal to colorectal cancer tissue [217,218,219,220,221]. The equilibrium between the cytokines IL-23 and IL-12 is a significant shift from inflammation to tumorigenesis [216,222].

5.6. Interleukin-33

IL-33 is a member of the IL-1 superfamily of cytokines expressed in various organ systems, including the gastrointestinal tract [222]. It was identified as a receptor ST2 ligand in 2005, and its mRNA processing forms multiple isoforms of proteins, including a secreted soluble form (sST2), a transmembrane receptor (ST2L), and a variant form [223]. Myofibroblasts, smooth muscle cells, fibroblasts, epithelial cells, adipocytes, and endothelial cells (non-hematopoietic cells) are the main sources of IL-33 production [224,225]. IL-33 is highly expressed in the serum of cancer patients and is also found in cancer cells and cancer-associated fibroblasts (CAFs) [226]. Many studies have reported its role in metastasis and tumorous cell invasion, and inhibiting it in colon cancer cells resulted in reduced tumor growth, migration in vitro, and fewer tumor cells in vivo [147,227]. Overexpression of IL-33 in cancer can increase the antitumor immune response by activating CD8+ T and natural killer cells [228]. Similar studies have depicted an increase in IL-33 in colorectal cancer compared to normal tissues, and its antitumorigenic effect in CRC [147]. There is a positive correlation reported between human CRC development and IL-33/IL-1RL1 expression levels [147,229], which reduces tumor growth in skin cancer and CRC models [230,231].

5.7. Interleukin-15

IL-15 is a cytokine that can activate CD8+ T cells and natural killer cells, leading to cytolytic activity [232]. This cytokine has shown the potential to enhance antitumor responses in cancer models [233,234]. The presence of IL-15 expression in the tumor microenvironment (TME) is crucial for optimal antitumor responses [235,236]. Its loss in expression is associated with low T cell proliferation, low T cell density, and decreased survival [235]. The mRNA expression of IL-15 has been detected in colorectal cancer cells of humans, such as Colo320, WiDr, TCO, and DLD1, through reverse transcriptase-polymerase chain reaction (RT-PCR) [237]. IL-15 may have the potential to be used in cancer therapy, as it exhibits strong immune stimulatory functions in addition to its role as a growth factor that regulates homeostasis and lymphocyte function [238]. IL-15 has antitumor effects by activating the cytotoxicity of natural killer cells and producing other cytokines such as TNF-α and IFN-γ [239]. Deletion of IL-15 from CRC tissues results in fewer T cells compared to tumors where IL-15 is not deleted, as IL-15 induces T cell proliferation. Therefore, IL-15 deletion can be utilized as a prognostic biomarker in CRC [240].

5.8. Interleukin-18

The expression of IL-18 is reported to be low in colon cancer tissues and may be associated with tumor size, while also suppressing the proliferation of colon cancer [133]. Known as the “IFN-γ-inducing factor,” IL-18 induces IFN-γ expression in mice when treated with lipopolysaccharide [241]. The protein encoded by the IL-18 gene, located at 11q23.1, is responsible for pathogenic response and activation of host defense mechanisms [242]. IL-18 is mainly secreted by dendritic cells and macrophages, stimulating the production of interferon-γ (IFN-γ) by thymus-dependent lymphocytes (T cells) and natural killer cells (NK) [243]. IL-18 expression is elevated in the blood serum of CRC patients and is associated with tumor size, histological grade, and cancer cell metastasis, making it a potential indicator to predict CRC patients’ prognosis and survival time [143]. Gene expression of IL-8 is elevated (p < 0.05) in CRC patients compared to healthy individuals, detected using ELISA and real-time PCR [143].

5.9. Interleukin-13

IL-13 and IL-4 receptors may become attractive targets for the treatment of colorectal cancer [244,245]. High levels of IL-13Rα2 were detected by immunoblotting in metastatic colon cancer cell lines, and 66% of tumor samples in patients showed clear overexpression of IL-13Rα2 [246]. High levels of IL-13 and IL-13R expression are seen in 50% of Stage I–III CRC patients and are associated with longer survival time [244]. Its appearance is also related to high tumor stage and poor human CRC outcomes [246]. A study showed that IL-13 serum levels were significantly lower in advanced-stage patients, which are associated with a poorer prognosis [247]. Yet, another study with fecal samples showed higher IL-13 levels in 20 CRC patients compared to 20 healthy individuals [248]. IL-13 enhances the expression of EMT-promoting factor ZEB1 with a positive correlation between IL-13Rα1 and ZEB1 at mRNA levels in human CRC samples. Hence, the IL-13/IL-13Rα1/STAT6/ZEB1 pathway plays a critical role in promoting EMT and CRC aggressiveness [249]. Reports showed that IL-13R is involved in the local metastases process of colorectal cancer, while expression of IL-13 has an impact on survival. These interleukins and their receptors may become attractive targets for the treatment of colorectal cancer. [244]. Both expression level of IL-13Rα2 and IL-13Rα2-mediated signaling has been reported to cause cell survival, tumor proliferation, tumor progression, invasion, and metastasis [250].

5.10. Interleukin-4

Interleukin-4 (IL-4) is an anti-inflammatory and immunomodulatory cytokine that promotes cancer-directed immune surveillance [244]. IL-4R is expressed in human gastric cancer cell lines, such as CRL1739, and its expression contributes to local metastasis in colorectal cancer, making it an attractive target for CRC therapy [244]. IL-4 and IL-13 are cytokines that are structurally and functionally related, sharing common receptor subunits. They regulate immune responses and play a role in various human cancer pathogenesis, chemosensitivity, and prognosis [245,246,247]. The T allele of IL-4 rs2070874 is associated with a higher risk of gastrointestinal cancer [248]. IL-14 activates tumor-associated macrophages and suppressor cells containing tumor-promoting functions [251]. IL-4 is involved in the promotion of epithelial-to-mesenchymal transition (EMT) in CRC [146], while it inhibits the growth of GC cells, and its growth inhibitory effects are positively related to IL-4R expression in cell lines [252]. Moreover, IL-4 promotes EMT in CRC cell lines “HCT 116” and “RKO” through the STAT6 pathway [146].

5.11. Interleukin-8

Interleukin-8 (IL-8) is a chemokine that belongs to the CXC cytokine family and is markedly upregulated in colorectal cancer (CRC), contributing to enhanced invasion, tumor growth, and metastasis [253,254]. IL-8 has diverse biological actions, including promoting inflammation, infectious diseases, invasion, migration, angiogenesis, and proliferation [255]. Serum IL-8 levels are a promising biomarker for detecting CRC and identifying high-risk patients [256]. IL-8 induces CRC cell migration and proliferation through the ADAM-dependent pathway and disintegrin, where heparin-binding epidermal growth factor (EGF) acts as a major ligand [257]. IL-8 has a multifunctional role in CRC progression, including enhancing the survival of cancer cells, promoting tumor cell proliferation, and regulating adhesion and invasion [258,259,260]. Excessive expression of IL-8 in the cancer microenvironment promotes colon cancer growth and metastasis, but the absence of its receptor CXCR2 prevents cell growth [261]. The autocrine properties of IL-8/CXCR2-mediated activation facilitate the intrinsic mechanism of tumor cells to avoid stress-induced apoptosis [262]. Increased plasma levels of IL-8 are associated with a single-nucleotide polymorphism (SNP) in IL-8 at 251 bp upstream [263], with the IL-8T–251A polymorphism being individually associated with tumor reappearance risk [264]. IL-8 also regulates chemosensitivity and angiogenesis (in vitro and in vivo) in models of colon cancer [265]. Stage IV CRC shows almost 10-fold higher serum levels of IL-8 than normal individuals [265]. Enhanced serum expression of IL-8 is also found in liver and lung tissue damage along with metastatic CRC [265], suggesting that the systemic increase in IL-8 can be more important for prognosis than the local and cancer cell-derived IL-8 levels. IL-8 is highly expressed in CRC tissues but is differentially produced by tumor components depending on the genetic background of CRC. As IL-8 is a strong prognostic factor in CRC, it may be used for prognostic assessment and tailoring of therapeutic strategies in individual CRC patients [266].

5.12. Interleukin-11

Interleukin-11 (IL-11) belongs to the IL-6 family and has a wide range of functions, including hematopoiesis, bone development, tissue repair, and tumor development [267,268]. In some cancer cells, the IL-11 receptor (IL-11R) has been identified, which contains IL-11Rα1. When bound to IL-11 and gp130, this receptor transmits signals to the nucleus through Janus kinase (JAK) activation [269,270]. JAKs subsequently phosphorylate STAT3, which then enters the nucleus and activates the transcription of numerous target genes involved in the suppression of cell proliferation and apoptosis [158,271]. IL-11 production is regulated by various cytokines, such as TGFβ, IL-1β, IL-17A, and IL-22 [272,273,274,275]. A human study has suggested that a polymorphism of the IL-11 gene is linked to an increased susceptibility to ulcerative colitis (UC) in patients [276]. While mild UC patient’s exhibit increased IL-11 expression, severe UC patients show a decrease in expression [277]. A study using a human cancer database found that genes enriched in IL-11+ fibroblasts were elevated in human colorectal cancer, and the high expressions of several of these genes correlated with a reduced disease-free survival rate in colorectal cancer patients [278]. Previous studies have demonstrated that IL-11+ cells are derived from stromal or epithelial cells [53,158]. There is substantial evidence that IL-11 regulates tumor progression, cellular growth, and differentiation. While IL-11 has been suggested to become a therapeutically important molecule in the supportive care of cancer patients receiving chemotherapy [278], the study indicates that IL-11 may upregulate colorectal carcinoma cell growth and/or invasion, necessitating cautious attention to the therapeutic use of IL-11 [279]. Table 3 contains detailed description of interleukin families involved in CRC and their potential therapeutic strategies.

6. Discussion

Despite significant advances in treatment, mortality from colorectal cancer remains high, and 40–50% of patients eventually die due to the disease. The most significant impact on its incidence and mortality will come from extensive population screening [334]. Colorectal cancer is a complex and diverse group of disorders at the molecular level, involving signaling pathways with different patterns of genetic mutations [335]. Epigenetic modifications cause the progression of the disease along with the responses to specific therapies. As it is caused by the activation of multiple signaling pathways and cannot be targeted with a single treatment, combinations of conventional therapies with advanced inhibitors are immediately needed to target dysregulated pathways. It has been extensively recognized that immune system dysfunction, including abnormally expressed cytokines, is strongly associated with the progression and pathogenesis of colorectal cancer [336]. Diagnostic tests have many limitations. For example, fecal blood test screening suffers from low sensitivity for polyps, and colonoscopy is invasive [337,338].
The most important feature of activated immune cells is the production and release of growth factors and cytokines that modulate the inflammatory conditions [339,340,341,342,343,344,345,346]. Cancer-associated inflammation is a determining factor in disease progression and survival in CRC, contributing to invasion, tumor angiogenesis, and metastatic spread [347,348]. CRC biomarkers can be divided into two groups based on clinical criteria: “diagnostic biomarkers” for the detection or confirmation of the presence of the disease, and “clinical biomarkers” for the prediction of patients’ response to a specific treatment or their prognosis [349]. Although many novel therapeutic improvements have been anticipated to treat colon cancer, the survival rate is still unsatisfactory due to metastasis and tumor reappearance [44]. Molecular biomarkers are being explored for implementation in clinical practice in this period of precision cancer medicine [350]. These prognostic biomarkers are desired to assist patients and predict survival. A deeper understanding of CRC is required, and the efficiency of targeted therapies and the development of more efficient biomarkers provide an encouraging prospect for the future management of CRC [349]. We believe that with the discovery of more novel targeted therapeutics, the disease burden of CRC can be decreased in the future [351]. However, there have been only a few studies on point-of-care cytokine detection due to challenges such as low concentration, complex secretion process, thermal instability, and others [29,74]. Yet, studies have investigated the use of ILs as biomarkers for monitoring the response to treatment in CRC patients [12,352,353]. These studies demonstrate the potential of ILs as biomarkers for the diagnosis of CRC and these findings are based on studies that have been conducted on a small number of patients and more research is needed to confirm the accuracy of these results in larger patient populations.
Finding more effective prognostic markers and therapeutic targets for patients with advanced colorectal cancer [354] is important because the majority of patients with advanced colon cancer cannot undergo surgery. Subsequently, due to the widespread adoption of CRC screening in the population, many patients would be diagnosed at the preclinical stage through screening [355]. Given the rise in treatment costs, screening for colorectal cancer is a cost-saving tool in many countries [356]. Various genomic projects have acknowledged new potential molecular markers and targets for colorectal cancer to guide more specific treatments for patients [357]. Oncogenes involved in CRC are mainly well characterized; nevertheless, the effects of additional environmental factors in this disease are undefined. Molecular biomarkers have been investigated for the last 20 years with promising results. However, many drawbacks affect the consistency of the conclusions [358,359,360,361]. More detailed research is needed on the relationship between diet, microbiota, and CRC.
There is high heterogeneity and complexity in CRC; therefore, standard treatments including radiation/chemotherapy are only effective in only a few patient populations. Tumors can also have various core genetic causes which makes the protein expressions different in each patient along with their responses to generic treatments [362]. This intrinsic changeability of cancer lends to the growing field of precision and personalized medicine (PPM). Many steps are being taken in order to attain PPM data to distinguish molecular differences between tumors. These include “immunotherapy” to utilize the patient’s own immunity against cancer, containing cytokines, vaccines, checkpoint inhibitors, monoclonal antibodies (mAbs), and hematopoietic stem cell transplants (HSCTs) [363]. There is a growing category of PPM products known as “companion diagnostics (CDx)”, molecular assays that assess proteins, genes, or specific mutation levels to diagnose and suggest a specific and effective therapy for an individual’s condition [364].

7. Future Perspective

Extensive population screening is expected to have the most significant impact on colorectal cancer incidence and mortality. By developing and implementing new, more specific and sensitive biomarkers, clinicians can improve diagnostic strategies and detect CRC cases early in the disease, thereby improving the prognosis of thousands of patients. Several new potential molecular targets and markers for colorectal cancer have been identified through various genomic projects, providing guidance for more specific treatments. Although most of the major oncogenes involved in CRC are well characterized, the effects of additional environmental factors in this disease are still undefined. While molecular biomarker studies over the past two decades have shown promising results, some drawbacks limit the reliability of the conclusions. Therefore, further research is needed to investigate the relationship between diet, microbiota, and CRC in greater detail.

8. Conclusions

Previously, scientists have reported significant information on the various genes and proteins that contribute to cancer. Mutated genes and the identification of related environmental factors are key discoveries. Using molecular methods, important gene expressions can be determined and used as novel biomarkers to reduce cancer complications and treatment. Further studies are required to explore the pathways and mechanisms involved in the expressions of immunological biomarkers and their involvement in the development and progression of colorectal cancer. Detecting cytokines at the required detection limit for reliable results is challenging, but many efforts have been made to develop cytokine assays with more sensitivity. Research into cytokine quantification is still developing to find effective solutions for the accurate and real-time detection of multiple cytokines in vivo. The effects of promising targets on different immune cell populations are still poorly understood. Therefore, improving antitumor responses and suppressing immune cells that support tumor growth are the prospects for cytokine-based cancer treatment. However, identifying all environmental factors, pivotal genes, immune responses, and cytokine release at the cancer stage provides a comprehensive map for further efforts to reduce cancer in the future.

Author Contributions

Conceptualization, S.M.; original draft preparation, S.M., I.U.H., A.A.K. and G.Y. writing—review and editing, G.Y., K.K., I.U.H., S.C. and A.A.K. All authors have read and agreed to the published version of the manuscript.

Funding

The APC of this manuscript is supported by the University of Oradea, Romania.

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. Safarzadeh, E.; Sandoghchian, S.; Baradaran, B. Herbal medicine as inducers of apoptosis in cancer treatment. Adv. Pharm. Bull. 2014, 4, 421–427. [Google Scholar] [CrossRef] [PubMed]
  2. Shewach, D.S.; Kuchta, R.D. Introduction to cancer chemotherapeutics. Chem. Rev. 2009, 109, 2859–2861. [Google Scholar] [CrossRef] [PubMed]
  3. Jemal, A.; Bray, F.; Center, M.M.; Ferlay, J.; Ward, E.; Forman, D. Global cancer statistics. CA Cancer J. Clin. 2011, 61, 69–90. [Google Scholar] [CrossRef] [PubMed]
  4. Arnold, M.; Sierra, M.S.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global patterns and trends in colorectal cancer incidence and mortality. Gut 2017, 66, 683–691. [Google Scholar] [CrossRef]
  5. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef]
  6. Keum, N.; Giovannucci, E. Global burden of colorectal cancer: Emerging trends.; risk factors and prevention strategies. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 713–732. [Google Scholar] [CrossRef]
  7. 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]
  8. Williams, R.; White, P.; Nieto, J.; Vieira, D.; Francois, F.; Hamilton, F. Colorectal Cancer in African Americans: An Update. Clin. Transl. Gastroenterol. 2016, 7, e185. [Google Scholar] [CrossRef]
  9. Jess, T.; Rungoe, C.; Peyrin-Biroulet, L. Risk of colorectal cancer in patients with ulcerative colitis: A meta-analysis of population-based cohort studies. Clin. Gastroenterol. Hepatol. 2012, 10, 639–645. [Google Scholar] [CrossRef]
  10. Marventano, S.; Forjaz, M.J.; Grosso, G.; Mistretta, A.; Giorgianni, G.; Platania, A.; Gangi, S.; Basile, F.; Biondi, A. Health related quality of life in colorectal cancer patients: State of the art. BMC Surg. 2013, 13, S15. [Google Scholar] [CrossRef]
  11. Vacante, M.; Borzì, A.M.; Basile, F.; Biondi, A. Biomarkers in colorectal cancer: Current clinical utility and future perspectives. World J. Clin. Cases 2018, 6, 869–881. [Google Scholar] [CrossRef] [PubMed]
  12. Mager, L.F.; Wasmer, M.H.; Rau, T.T.; Krebs, P. Cytokine-Induced Modulation of Colorectal Cancer. Front. Oncol. 2016, 6, 96. [Google Scholar] [CrossRef] [PubMed]
  13. Lee, S.; Margolin, K. Cytokines in cancer immunotherapy. Cancers 2011, 3, 3856–3893. [Google Scholar] [CrossRef] [PubMed]
  14. Koltsova, E.K.; Grivennikov, S.I. IL-22 Gets to the Stem of Colorectal Cancer. Immunity 2014, 40, 639–641. [Google Scholar] [CrossRef]
  15. Wang, J.; Li, D.; Cang, H.; Guo, B. Crosstalk between cancer and immune cells: Role of tumor-associated macrophages in the tumor microenvironment. Cancer Med. 2019, 8, 4709–4721. [Google Scholar] [CrossRef] [PubMed]
  16. Kawanishi, S.; Ohnishi, S.; Ma, N.; Hiraku, Y.; Murata, M. Crosstalk between DNA Damage and Inflammation in the Multiple Steps of Carcinogenesis. Int. J. Mol. Sci. 2017, 18, 1808. [Google Scholar] [CrossRef] [PubMed]
  17. Li, J.; Huang, L.; Zhao, H.; Yan, Y.; Lu, J. The Role of Interleukins in Colorectal Cancer. Int. J. Biol. Sci. 2020, 16, 2323–2339. [Google Scholar] [CrossRef]
  18. Veziant, J.; Villéger, R.; Barnich, N.; Bonnet, M. Gut Microbiota as Potential Biomarker and/or Therapeutic Target to Improve the Management of Cancer: Focus on Colibactin-Producing Escherichia coli in Colorectal Cancer. Cancers 2021, 13, 2215. [Google Scholar] [CrossRef]
  19. Kim, J.; Lee, H.K. Potential Role of the Gut Microbiome In Colorectal Cancer Progression. Front. Immunol. 2022, 12, 807648. [Google Scholar] [CrossRef]
  20. Olovo, C.V.; Huang, X.; Zheng, X.; Xu, M. Faecal microbial biomarkers in early diagnosis of colorectal cancer. J. Cell. Mol. Med. 2021, 25, 10783–10797. [Google Scholar] [CrossRef]
  21. Rye, M.S.; Garrett, K.L.; Holt, R.A.; Platell, C.F.; McCoy, M.J. Fusobacterium nucleatum and Bacteroides fragilis detection in colorectal tumours: Optimal target site and correlation with total bacterial load. PLoS ONE 2022, 17, e0262416. [Google Scholar] [CrossRef] [PubMed]
  22. Eklöf, V.; Löfgren-Burström, A.; Zingmark, C.; Edin, S.; Larsson, P.; Karling, P.; Alexeyev, O.; Rutegård, J.; Wikberg, M.L.; Palmqvist, R. Cancer-associated fecal microbial markers in colorectal cancer detection. Int. J. Cancer 2017, 141, 2528–2536. [Google Scholar] [CrossRef] [PubMed]
  23. Nassar, F.J.; Msheik, Z.S.; Itani, M.M.; Helou, R.E.; Hadla, R.; Kreidieh, F.; Bejjany, R.; Mukherji, D.; Shamseddine, A.; Nasr, R.R.; et al. Circulating miRNA as Biomarkers for Colorectal Cancer Diagnosis and Liver Metastasis. Diagnostics 2021, 11, 341. [Google Scholar] [CrossRef]
  24. Condrat, C.E.; Thompson, D.C.; Barbu, M.G.; Bugnar, O.L.; Boboc, A.; Cretoiu, D.; Suciu, N.; Cretoiu, S.M.; Voinea, S.C. miRNAs as Biomarkers in Disease: Latest Findings Regarding Their Role in Diagnosis and Prognosis. Cells 2020, 9, 276. [Google Scholar] [CrossRef] [PubMed]
  25. Staiteieh, S.A.; Akil, L.; Al Khansa, R.; Nasr, R.; Sagheer, A.Z.; Houshaymi, B.; Merhi, R.A. Study of microRNA expression profiling as biomarkers for colorectal cancer patients in Lebanon. Mol. Clin. Oncol. 2022, 16, 39. [Google Scholar] [CrossRef] [PubMed]
  26. Sur, D.; Advani, S.; Braithwaite, D. MicroRNA panels as diagnostic biomarkers for colorectal cancer: A systematic review and meta-analysis. Front. Med. 2022, 9, 915226. [Google Scholar] [CrossRef]
  27. Precazzini, F.; Detassis, S.; Imperatori, A.S.; Denti, M.A.; Campomenosi, P. Measurements Methods for the Development of MicroRNA-Based Tests for Cancer Diagnosis. Int. J. Mol. Sci. 2021, 22, 1176. [Google Scholar] [CrossRef]
  28. Kaur, J.; Preethi, M.; Srivastava, R.; Borse, V. Role of IL-6 and IL-8 biomarkers for optical and electrochemical based point-of-care detection of oral cancer. Biosens. Bioelectron. X 2022, 11, 100212. [Google Scholar] [CrossRef]
  29. Liu, G.; Jiang, C.; Lin, X.; Yang, Y. Point-of-care detection of cytokines in cytokine storm management and beyond: Significance and challenges. View 2021, 2, 20210003. [Google Scholar] [CrossRef]
  30. Chaudhry, H.; Zhou, J.; Zhong, Y.; Ali, M.M.; McGuire, F.; Nagarkatti, P.S.; Nagarkatti, M. Role of cytokines as a double-edged sword in sepsis. In Vivo 2013, 27, 669–684. [Google Scholar]
  31. Febbo, P.G.; Ladanyi, M.; Aldape, K.D.; De Marzo, A.M.; Hammond, M.E.; Hayes, D.F.; Lafrate, A.J.; Kelley, R.K.; Marcucci, G.; Ogino, S.; et al. NCCN Task Force report: Evaluating the clinical utility of tumor markers in oncology. J. Natl. Compr. Cancer Netw. 2011, 9, S1–S32. [Google Scholar] [CrossRef]
  32. Kartikasari, A.E.R.; Huertas, C.S.; Mitchell, A.; Plebanski, M. Tumor-Induced Inflammatory Cytokines and the Emerging Diagnostic Devices for Cancer Detection and Prognosis. Front. Oncol. 2021, 11, 692142. [Google Scholar] [CrossRef] [PubMed]
  33. Capone, F.; Guerriero, E.; Sorice, A.; Colonna, G.; Ciliberto, G.; Costantini, S. Serum Cytokinome Profile Evaluation: A Tool to Define New Diagnostic and Prognostic Markers of Cancer Using Multiplexed Bead-Based Immunoassays. Mediat. Inflamm. 2016, 2016, 3064643. [Google Scholar] [CrossRef] [PubMed]
  34. Schett, G.; Elewaut, D.; McInnes, I.B.; Dayer, J.M.; Neurath, M.F. How cytokine networks fuel inflammation: Toward a cytokine-based disease taxonomy. Nat. Med. 2013, 19, 822–824. [Google Scholar] [CrossRef] [PubMed]
  35. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed]
  36. Jia, D.; Li, L.; Andrew, S.; Allan, D.; Li, X.; Lee, J.; Ji, Z.; Yao, Z.; Gadde, S.; Figeys, D.; et al. An autocrine inflammatory forward-feedback loop after chemotherapy withdrawal facilitates the repopulation of drug-resistant breast cancer cells. Cell Death Dis. 2017, 8, e2932. [Google Scholar] [CrossRef]
  37. Nengroo, M.A.; Verma, A.; Datta, D. Cytokine chemokine network in tumor microenvironment: Impact on CSC properties and therapeutic applications. Cytokine 2022, 156, 155916. [Google Scholar] [CrossRef]
  38. Furman, D.; Campisi, J.; Verdin, E.; Carrera-Bastos, P.; Targ, S.; Franceschi, C.; Ferrucci, L.; Gilroy, D.W.; Fasano, A.; Miller, G.W.; et al. Chronic inflammation in the etiology of disease across the life span. Nat. Med. 2019, 25, 1822–1832. [Google Scholar] [CrossRef]
  39. Greten, F.R.; Grivennikov, S.I. Inflammation and Cancer: Triggers, Mechanisms, and Consequences. Immunity 2019, 51, 27–41. [Google Scholar] [CrossRef]
  40. Sokolova, O.; Naumann, M. Crosstalk between DNA Damage and Inflammation in the Multiple Steps of Gastric Carcinogenesis. Curr. Top. Microbiol. Immunol. 2019, 421, 107–137. [Google Scholar] [CrossRef]
  41. Yang, Z.H.; Dang, Y.Q.; Ji. G. Role of epigenetics in transformation of inflammation into colorectal cancer. World J. Gastroenterol. 2019, 25, 2863–2877. [Google Scholar] [CrossRef] [PubMed]
  42. Martin, B.; Märkl, B. Immunologic Biomarkers and Biomarkers for Immunotherapies in Gastrointestinal Cancer. Visc. Med. 2019, 35, 3–10. [Google Scholar] [CrossRef] [PubMed]
  43. Rhea, J.M.; Molinaro, R.J. Cancer biomarkers: Surviving the journey from bench to bedside. MLO Med. Lab. Obs. 2011, 43, 10–12. [Google Scholar] [PubMed]
  44. Nagata, H.; Ishihara, S.; Hata, K.; Murono, K.; Kaneko, M.; Yasuda, K.; Otani, K.; Watanabe, T. Survival and Prognostic Factors for Metachronous Peritoneal Metastasis in Patients with Colon Cancer. Ann. Surg. Oncol. 2017, 24, 1269–1280. [Google Scholar] [CrossRef] [PubMed]
  45. Goossens, N.; Nakagawa, S.; Sun, X.; Hoshida, Y. Cancer biomarker discovery and validation. Transl. Cancer Res. 2015, 4, 256. [Google Scholar] [PubMed]
  46. Sarhadi, V.K.; Armengol, G. Molecular Biomarkers in Cancer. Biomolecules 2022, 12, 1021. [Google Scholar] [CrossRef]
  47. Herceg, Z.; Hainaut, P. Genetic and epigenetic alterations as biomarkers for cancer detection, diagnosis and prognosis. Mol. Oncol. 2007, 1, 26–41. [Google Scholar] [CrossRef]
  48. Harbaum, L.; Pollheimer, M.J.; Kornprat, P.; Lindtner, R.A.; Bokemeyer, C.; Langner, C. Peritumoral eosinophils predict recurrence in colorectal cancer. Mod. Pathol. 2015, 28, 403–413. [Google Scholar] [CrossRef]
  49. HSU, C.-P.; Chung, Y.-C. Influence of Interleukin-6 on the Invasiveness of Human Colorectal Carcinoma. Anticancer. Res. 2006, 26, 4607–4614. Available online: https://ar.iiarjournals.org/content/anticanres/26/6B/4607.full.pdf (accessed on 17 March 2023).
  50. Waniczek, D.; Lorenc, Z.; Śnietura, M.; Wesecki, M.; Kopec, A.; Muc-Wierzgoń, M. Tumor-Associated Macrophages and Regulatory T Cells Infiltration and the Clinical Outcome in Colorectal Cancer. Arch. Immunol. Ther. Exp. 2017, 65, 445–454. [Google Scholar] [CrossRef]
  51. Wikberg, M.L.; Ling, A.; Li, X.; Öberg, Å.; Edin, S.; Palmqvist, R. Neutrophil infiltration is a favorable prognostic factor in early stages of colon cancer. Hum. Pathol. 2017, 68, 193–202. [Google Scholar] [CrossRef] [PubMed]
  52. Terzić, J.; Grivennikov, S.; Karin, E.; Karin. M. Inflammation and colon cancer. Gastroenterology 2010, 138, 2101–2114.e5. [Google Scholar] [CrossRef]
  53. Calon, A.; Espinet, E.; Palomo-Ponce, S.; Tauriello, D.V.; Iglesias, M.; Céspedes, M.V. Dependency of colorectal cancer on a TGF-β-driven program in stromal cells for metastasis initiation. Cancer Cell 2012, 22, 571–584. [Google Scholar] [CrossRef] [PubMed]
  54. Guo, S.; Deng, C.X. Effect of Stromal Cells in Tumor Microenvironment on Metastasis Initiation. Int. J. Biol. Sci. 2018, 14, 2083–2093. [Google Scholar] [CrossRef] [PubMed]
  55. Nguyen, L.H.; Goel, A.; Chung, D.C. Pathways of Colorectal Carcinogenesis. Gastroenterology 2020, 158, 291–302. [Google Scholar] [CrossRef]
  56. Pino, M.S.; Chung, D.C. The chromosomal instability pathway in colon cancer. Gastroenterology 2010, 138, 2059–2072. [Google Scholar] [CrossRef]
  57. Powell, D.W.; Adegboyega, P.A.; Di Mari, J.F.; Mifflin, R.C. Epithelial cells and their neighbors I. Role of intestinal myofibroblasts in development repair and cancer. Am. J. Physiol. Gastrointest. Liver Physiol. 2005, 289, G2–G7. [Google Scholar] [CrossRef]
  58. Koliaraki, V.; Pallangyo, C.K.; Greten, F.R.; Kollias, G. Mesenchymal Cells in Colon Cancer. Gastroenterology 2017, 152, 964–979. [Google Scholar] [CrossRef]
  59. Kryczek, I.; Lin, Y.; Nagarsheth, N.; Peng, D.; Zhao, L.; Zhao, E.; Vatan, L.; Szeliga, W.; Dou, Y.; Owens, S.; et al. IL-22(+) CD4(+) T cells promote colorectal cancer stemness via STAT3 transcription factor activation and induction of the methyltransferase DOT1L. Immunity 2014, 40, 772–784. [Google Scholar] [CrossRef]
  60. Todaro, M.; Gaggianesi, M.; Catalano, V.; Benfante, A.; Iovino, F.; Biffoni, M.; Apuzzo, T.; Sperduti, I.; Volpe, S.; Cocorullo, G.; et al. CD44v6 is a marker of constitutive and reprogrammed cancer stem cells driving colon cancer metastasis. Cell Stem Cell 2014, 14, 342–356. [Google Scholar] [CrossRef]
  61. Lotti, F.; Jarrar, A.M.; Pai, R.K.; Hitomi, M.; Lathia, J.; Mace, A.; Rich, J.N. Chemotherapy activates cancer-associated fibroblasts to maintain colorectal cancer-initiating cells by IL-17A. J. Exp. Med. 2013, 210, 2851–2872. [Google Scholar] [CrossRef] [PubMed]
  62. Chen, K.; Liu, Q.; Tsang, L.L.; Ye, Q.; Chan, H.C.; Sun, Y.; Jiang, X. Human MSCs promotes colorectal cancer epithelial–mesenchymal transition and progression via CCL5/β-catenin/Slug pathway. Cell Death Dis. 2017, 8, e2819. [Google Scholar] [CrossRef] [PubMed]
  63. Vermeulen, L.; De Sousa, E.M.F.; van der Heijden, M.; Cameron, K.; de Jong, J.H.; Borovski, T.; Tuynman, J.B.; Todaro, M.; Merz, C.; Rodermond, H.; et al. Wnt activity defines colon cancer stem cells and is regulated by the microenvironment. Nat. Cell Biol. 2010, 12, 468–476. [Google Scholar] [CrossRef] [PubMed]
  64. Schulte, W.; Jürgen, B.; Richard, B. Cytokines in sepsis: Potent immunoregulators and potential therapeutic targets—An updated view. Mediat. Inflamm. 2013, 2013, 165974. [Google Scholar] [CrossRef] [PubMed]
  65. Sprague, A.H.; Khalil, R.A. Inflammatory cytokines in vascular dysfunction and vascular disease. Biochem. Pharmacol. 2009, 78, 539–552. [Google Scholar] [CrossRef] [PubMed]
  66. Rea, I.M.; Gibson, D.S.; McGilligan, V.; McNerlan, S.E. Alexander HD, Ross OA. Age and Age-Related Diseases: Role of Inflammation Triggers and Cytokines. Front. Immunol. 2018, 9, 586. [Google Scholar] [CrossRef]
  67. Nedoszytko, B.; Sokołowska-Wojdyło, M.; Ruckemann-Dziurdzińska, K.; Roszkiewicz, J.; Nowicki, R.J. Chemokines and cytokines network in the pathogenesis of the inflammatory skin diseases: Atopic dermatitis, psoriasis and skin mastocytosis. Postępy Dermatol. I Alergol. 2014, 31, 84–91. [Google Scholar] [CrossRef]
  68. Kabel, A.M. Relationship between cancer and cytokines. J. Cancer Res. Treat. 2014, 2, 41–43. [Google Scholar]
  69. Comella-Del-Barrio, P.; Abellana, R.; Villar-Hernández, R.; Jean Coute, M.D.; Sallés Mingels, B.; Canales Aliaga, L.; Narcisse, M.; Gautier, J.; Ascaso, C.; Latorre, I.; et al. A model based on the combination of IFN-γ, IP-10, ferritin and 25-hydroxyvitamin D for discriminating latent from active tuberculosis in children. Front. Microbiol. 2019, 10, 1855. [Google Scholar] [CrossRef]
  70. Pugliese, N.R.; Fabiani, I.; Conte, L.; Nesti, L.; Masi, S.; Natali, A.; Colombo, P.C.; Pedrinelli, R.; Dini, F.L. Persistent congestion, renal dysfunction and inflammatory cytokines in acute heart failure: A prognosis study. J. Cardiovasc. Med. 2020, 21, 494–502. [Google Scholar] [CrossRef]
  71. Shimabukuro-Vornhagen, A.; Gödel, P.; Subklewe, M.; Stemmler, H.J.; Schlößer, H.A.; Schlaak, M.; Kochanek, M.; Böll, B.; von Bergwelt-Baildon, M.S. Cytokine release syndrome. J. Immunother. Cancer 2018, 6, 56. [Google Scholar] [CrossRef] [PubMed]
  72. Moore, J.B.; Carl, H.J. Cytokine release syndrome in severe COVID-19. Science 2020, 368, 473–474. [Google Scholar] [CrossRef] [PubMed]
  73. Simpson, S.; Kaislasuo, J.; Guller, S.; Pal, L. Thermal stability of cytokines: A review. Cytokine 2020, 125, 154829. [Google Scholar] [CrossRef] [PubMed]
  74. Liu, G.; Qi, M.; Hutchinson, M.R.; Yang, G.; Goldys, E.M. Recent advances in cytokine detection by immunosensing. Biosens. Bioelectron. 2016, 79, 810–821. [Google Scholar] [CrossRef] [PubMed]
  75. Monastero, R.N.; Srinivas, P. Cytokines as biomarkers and their respective clinical cutoff levels. Int. J. Inflam. 2017, 2017, 4309485. [Google Scholar] [CrossRef] [PubMed]
  76. Kouwenhoven, M.; Ozenci, V.; Teleshova, N.; Hussein, Y.; Huang, Y.M.; Eusebio, A.; Link, H. Enzyme-linked immunospot assays provide a sensitive tool for detection of cytokine secretion by monocytes. Clin. Diagn. Lab. Immunol. 2001, 8, 1248–1257. [Google Scholar] [CrossRef] [PubMed]
  77. Stenken, J.A.; Poschenrieder, A.J. Bioanalytical chemistry of cytokines–a review. Anal. Chim. Acta 2015, 853, 95–115. [Google Scholar] [CrossRef]
  78. Zhang, K.; Baratta, M.V.; Liu, G.; Frank, M.G.; Leslie, N.R.; Watkins, L.R.; Maier, S.F.; Hutchinson, M.R.; Goldys, E.M. A novel platform for in vivo detection of cytokine release within discrete brain regions. Brain Behav. Immun. 2018, 71, 18–22. [Google Scholar] [CrossRef]
  79. Arman, A.; Deng, F.; Goldys, E.M.; Liu, G.; Hutchinson, M.R. In vivo intrathecal IL-1β quantification in rats: Monitoring the molecular signals of neuropathic pain. Brain Behav. Immun. 2020, 88, 442–450. [Google Scholar] [CrossRef]
  80. Tertis, M.; Leva, P.I.; Bogdan, D.; Suciu, M.; Graur, F.; Cristea, C. Impedimetric aptasensor for the label-free and selective detection of Interleukin-6 for colorectal cancer screening. Biosens. Bioelectron. 2019, 137, 123–132. [Google Scholar] [CrossRef]
  81. Aldo, P.; Marusov, G.; Svancara, D.; David, J.; Mor, G. Simple Plex™: A novel multi-analyte, automated microfluidic immunoassay platform for the detection of human and mouse cytokines and chemokines. Am. J. Reprod. Immunol. 2016, 75, 6. [Google Scholar] [CrossRef] [PubMed]
  82. Zhong, Y.; Tang, X.; Li, J.; Lan, Q.; Min, L.; Ren, C.; Hu, X.; Torrente, R.M.; Gao, W.; Yang, Z. A nanozyme tag enabled chemiluminescence imaging immunoassay for multiplexed cytokine monitoring. Chem. Commun. 2018, 54, 13813–13816. [Google Scholar] [CrossRef] [PubMed]
  83. Emoto, S.; Ishigami, H.; Yamashita, H.; Yamaguchi, H.; Kaisaki, S.; Kitayama, J. Clinical significance of CA125 and CA72-4 in gastric cancer with peritoneal dissemination. Gastric Cancer 2012, 15, 154–161. [Google Scholar] [CrossRef] [PubMed]
  84. Liang, Y.; Wang, W.; Fang, C.; Raj, S.S.; Hu, W.M.; Li, Q.W.; Zhou, Z.W. Clinical significance and diagnostic value of serum CEA, CA19-9 and CA72-4 in patients with gastric cancer. Oncotarget 2016, 7, 49565–49573. [Google Scholar] [CrossRef] [PubMed]
  85. Ulusoy, H.; Kangalgil, M.; Küçük, A.O.; Özdemir, A.; Karahan, S.C.; Yaman, S.Ö.; Yavuz, H.B.; Ok, Ü. Effects of different lipid emulsions on serum adipokines, inflammatory markers and mortality in critically ill patients with sepsis: A prospective observational cohort study. Clin. Nutr. 2021, 40, 4569–4578. [Google Scholar] [CrossRef]
  86. He, R.; Zhu, Q.; Wang, Y.; Chen, G.; Chen, S.; Wang, Y. Influence of respiratory function training under the mode of mutual-assisted patients on postoperative pulmonary infection and immune function on lung cancer. Am. J. Transl. Res. 2021, 13, 9260–9268. [Google Scholar]
  87. Diamandis, E.P.; Hoffman, B.R.; Sturgeon, C.M. National academy of clinical biochemistry laboratory medicine practice guidelines for the use of tumor markers. Clin. Chem. 2008, 54, 1935–1939. [Google Scholar] [CrossRef]
  88. Hing, J.X.; Mok, C.W.; Tan, P.T.; Sudhakar, S.S.; Seah, C.M.; Lee, W.P.; Tan, S.M. Clinical utility of tumour marker velocity of cancer antigen 15-3 (CA 15-3) and carcinoembryonic antigen (CEA) in breast cancer surveillance. Breast 2020, 52, 95–101. [Google Scholar] [CrossRef]
  89. Diaz, C.B.; Marono, S.H.; Olivas, M.L.G.; Sosa, M.A.; Soto, R.G.; Arrieta, O. Association of neurologic manifestations and CEA levels with the diagnosis of brain metastases in lung cancer patients. Clin. Transl. Oncol. 2019, 21, 1538–1542. [Google Scholar] [CrossRef]
  90. Song, X.; Liang, B.; Wang, C.; Shi, S. Clinical value of color Doppler ultrasound combined with serum CA153, CEA and TSGF detection in the diagnosis of breast cancer. Exp. Ther. Med. 2020, 20, 1822–1828. [Google Scholar] [CrossRef]
  91. Sakamoto, T.; Saito, H.; Amisaki, M.; Tokuyasu, N.; Honjo, S.; Fujiwara, Y. Combined preoperative platelet-to-lymphocyte ratio and serum carbohydrate antigen 19-9 level as a prognostic factor in patients with resected pancreatic cancer. Hepatobiliary Pancreat. Dis. Int. 2019, 18, 278–284. [Google Scholar] [CrossRef] [PubMed]
  92. Peng, H.X.; Yang, L.; He, B.S.; Pan, Y.Q.; Ying, H.Q.; Sun, H.L.; Lin, K.; Hu, X.X.; Xu, T.; Wang, S.K. Combination of preoperative NLR, PLR and CEA could increase the diagnostic efficacy for I-III stage CRC. J. Clin. Lab. Anal. 2017, 31, e22075. [Google Scholar] [CrossRef] [PubMed]
  93. Yan, X.; Han, L.; Zhao, R.; Fatima, S.; Zhao, L.; Gao, F. Prognosis value of IL-6, IL-8, and IL-1β in serum of patients with lung cancer: A fresh look at interleukins as a biomarker. Heliyon 2022, 8, e09953. [Google Scholar] [CrossRef] [PubMed]
  94. Xi, C.; Zhang, G.Q.; Sun, Z.K.; Song, H.J.; Shen, C.T.; Chen, X.Y.; Sun, J.W.; Qiu, Z.L.; Luo, Q.Y. Interleukins in Thyroid Cancer: From Basic Researches to Applications in Clinical Practice. Front. Immunol. 2020, 11, 1124. [Google Scholar] [CrossRef]
  95. Provatopoulou, X.; Georgiadou, D.; Sergentanis, T.N.; Kalogera, E.; Spyridakis, J.; Gounaris, A.; Zografos, G.N. Interleukins as markers of inflammation in malignant and benign thyroid disease. Inflamm. Res. 2014, 63, 667–674. [Google Scholar] [CrossRef]
  96. Rébé, C.; Ghiringhelli, F. Interleukin-1β and Cancer. Cancers 2020, 12, 1791. [Google Scholar] [CrossRef]
  97. Valeri, M.; Raffatellu, M. Cytokines IL-17 and IL-22 in the host response to infection. Pathog. Dis. 2016, 74, ftw111. [Google Scholar] [CrossRef]
  98. Anestakis, D.; Petanidis, S.; Kalyvas, S.; Nday, C.M.; Tsave, O.; Kioseoglou, E.; Salifoglou, A. Mechanisms and Applications of Ιnterleukins in Cancer Immunotherapy. Int. J. Mol. Sci. 2015, 16, 1691–1710. [Google Scholar] [CrossRef]
  99. Zarogoulidis, P.; Lampaki, S.; Yarmus, L.; Kioumis, I.; Pitsiou, G.; Katsikogiannis, N.; Hohenforst-Schmidt, W.; Li, Q.; Huang, H.; Sakkas, A. Interleukin-7 and interleukin-15 for cancer. J. Cancer 2014, 5, 765–773. [Google Scholar] [CrossRef]
  100. Todoric, J.; Antonucci, L.; Karin, M. Targeting Inflammation in Cancer Prevention and Therapy. Cancer Prev. Res. 2016, 9, 895. [Google Scholar] [CrossRef]
  101. Coussens, L.M.; Werb, Z. Inflammation and cancer. Nature 2002, 420, 860–867. [Google Scholar] [CrossRef] [PubMed]
  102. Mantovani, A.; Allavena, P.; Sica, A.; Balkwill, F. Cancer-related inflammation. Nature 2008, 454, 436–444. [Google Scholar] [CrossRef] [PubMed]
  103. Ilyin, S.E.; Belkowski, S.M.; Plata-Salamán, C.R. Biomarker discovery and validation: Technologies and integrative approaches. Trends Biotechnol. 2004, 22, 411–416. [Google Scholar] [CrossRef] [PubMed]
  104. Brunkow, M.E.; Jeffery, E.W.; Hjerrild, K.A.; Paeper, B.; Clark, L.B.; Yasayko, S.A.; Wilkinson, J.E.; Galas, D.; Ziegler, S.F.; Ramsdell, F. Disruption of a new forkhead/winged-helix protein.; scurfin.; results in the fatal lymphoproliferative disorder of the scurfy mouse. Nat. Genet. 2001, 27, 68–73. [Google Scholar] [CrossRef]
  105. Katoh, M.; Igarashi, M.; Fukuda, H.; Nakagama, H.; Katoh, M. Cancer genetics and genomics of human FOX family genes. Cancer Lett. 2013, 328, 198–206. [Google Scholar] [CrossRef]
  106. Tian, T.; Wang, M.; Zheng, Y.; Yang, T.; Zhu, W.; Li, H.; Lin, S.; Liu, K.; Xu, P.; Deng, Y.; et al. Association of two FOXP3 polymorphisms with breast cancer susceptibility in Chinese Han women. Cancer Manag. Res. 2018, 10, 867–872. [Google Scholar] [CrossRef]
  107. Luo, Q.; Zhang, S.; Wei, H.; Pang, X.; Zhang, H. Roles of Foxp3 in the occurrence and development of cervical cancer. Int. J. Clin. Exp. Pathol. 2015, 8, 8717–8730. [Google Scholar]
  108. Sun, X.; Feng, Z.; Wang, Y.; Qu, Y.; Gai, Y. Expression of Foxp3 and its prognostic significance in colorectal cancer. Int. J. Immunopathol. Pharmacol. 2017, 30, 201–206. [Google Scholar] [CrossRef]
  109. Liu, Y.; Lan, Q.; Lu, L.; Chen, M.; Xia, Z.; Ma, J.; Wang, J.; Fan, H.; Shen, Y.; Ryffel, B. Phenotypic and functional characteristic of a newly identified CD8+ Foxp3− CD103+ regulatory T cells. J. Mol. Cell Biol. 2014, 6, 81–92. [Google Scholar] [CrossRef]
  110. Kim, M.; Grimmig, T.; Grimm, M.; Lazariotou, M.; Meier, E.; Rosenwald, A.; Tsaur, I.; Blaheta, R.; Heemann, U.; Germer, C.T. Expression of Foxp3 in colorectal cancer but not in Treg cells correlates with disease progression in patients with colorectal cancer. PLoS ONE 2013, 8, e53630. [Google Scholar] [CrossRef]
  111. Kuwahara, T.; Hazama, S.; Suzuki, N.; Yoshida, S.; Tomochika, S.; Nakagami, Y.; Matsui, H.; Shindo, Y.; Kanekiyo, S.; Tokumitsu, Y. Intratumoural-infiltrating CD4 + and FOXP3 + T cells as strong positive predictive markers for the prognosis of resectable colorectal cancer. Br. J. Cancer 2019, 121, 659–665. [Google Scholar] [CrossRef] [PubMed]
  112. Grimmig, T.; Kim, M.; Germer, C.-T.; Gasser, M.; Maria Waaga-Gasser, A. The role of FOXP3 in disease progression in colorectal cancer patients. Oncoimmunology 2013, 2, e24521. [Google Scholar] [CrossRef] [PubMed]
  113. Bessis, N.; Chiocchia, G.; Kollias, G.; Minty, A.; Fournier, C.; Fradelizi, D.; Boissier, M.C. Modulation of proinflammatory cytokine production in tumour necrosis factor-alpha (TNF-alpha)-transgenic mice by treatment with cells engineered to secrete IL-4.; IL-10 or IL-13. Clin. Exp. Immunol. 1998, 111, 391–396. [Google Scholar] [CrossRef] [PubMed]
  114. Bates, R.C.; Mercurio, A.M. Tumor necrosis factor-alpha stimulates the epithelial-to-mesenchymal transition of human colonic organoids. Mol. Biol. Cell. 2003, 14, 1790–1800. [Google Scholar] [CrossRef] [PubMed]
  115. Ou, B.; Zhao, J.; Guan, S.; Feng, H.; Wangpu, X.; Zhu, C.; Zong, Y.; Ma, J.; Sun, J.; Shen, X.; et al. CCR4 promotes metastasis via ERK/NF-κB/MMP13 pathway and acts downstream of TNF-α in colorectal cancer. Oncotarget 2016, 7, 47637. [Google Scholar] [CrossRef] [PubMed]
  116. Jovanovic, D.V.; Di Battista, J.A.; Martel-Pelletier, J.; Jolicoeur, F.C.; He, Y.; Zhang, M.; Mineau, F.; Pelletier, J.P. IL-17 stimulates the production and expression of proinflammatory cytokines.; IL-beta and TNF-alpha. by human macrophages. J. Immunol. 1998, 160, 3513–3521. [Google Scholar] [CrossRef]
  117. Hunter, K. Host genetics influence tumour metastasis. Nat. Rev. Cancer 2006, 6, 141–146. [Google Scholar] [CrossRef]
  118. Al Obeed, O.A.; Alkhayal, K.A.; Al Sheikh, A.; Zubaidi, A.M.; Vaali-Mohammed, M.A.; Boushey, R.; Mckerrow, J.H.; Abdulla, M.H. Increased expression of tumor necrosis factor-α is associated with advanced colorectal cancer stages. World J. Gastroenterol. 2014, 20, 18390–18396. [Google Scholar] [CrossRef]
  119. Grimm, M.; Kim, M.; Rosenwald, A.; von Raden, B.H.; Tsaur, I.; Meier, E.; Heemann, U.; Germer, C.T.; Gasser, M.; Waaga-Gasser, A.M. Tumour-mediated TRAIL-Receptor expression indicates effective apoptotic depletion of infiltrating CD8+ immune cells in clinical colorectal cancer. Eur. J. Cancer 2010, 46, 2314–2323. [Google Scholar] [CrossRef]
  120. Stanilov, N.; Miteva, L.; Dobreva, Z.; Stanilova, S. Colorectal cancer severity and survival in correlation with tumour necrosis factor-alpha. Biotechnol. Biotechnol. Equip. 2014, 28, 911–917. [Google Scholar] [CrossRef]
  121. Warsinggih; Limanu, F.; Labeda, I.; Lusikooy, R.E.; Mappincara; Faruk, M. The relationship of tumor necrosis factor alpha levels in plasma toward the stage and differentiation degree in colorectal cancer. Med. Clínica Práctica 2021, 4, 100224. [Google Scholar] [CrossRef]
  122. Wang, L.; Wang, Y.; Song, Z.; Chu, J.; Qu, X. Deficiency of interferon-gamma or its receptor promotes colorectal cancer development. J. Interferon Cytokine Res. 2015, 35, 273–280. [Google Scholar] [CrossRef] [PubMed]
  123. Kosmidis, C.; Sapalidis, K.; Koletsa, T.; Kosmidou, M.; Efthimiadis, C.; Anthimidis, G.; Varsamis, N.; Michalopoulos, N.; Koulouris, C.; Atmatzidis, S.; et al. Interferon-γ and Colorectal Cancer: An up-to date. J. Cancer 2018, 9, 232–238. [Google Scholar] [CrossRef]
  124. Liu, S.; Yu, X.; Wang, Q.; Liu, Z.; Xiao, Q.; Hou, P.; Hu, Y.; Hou, W.; Yang, Z.; Guo, D.; et al. Specific expression of interferon-γ induced by synergistic activation mediator-derived systems activates innate immunity and inhibits tumorigenesis. J. Microbiol. Biotechnol. 2017, 27, 1855–1866. [Google Scholar] [CrossRef] [PubMed]
  125. Schroder, K.; Hertzog, P.J.; Ravasi, T.; Hume, D.A. Interferon-gamma: An overview of signals.; mechanisms and functions. J. Leukoc. Biol. 2004, 75, 163–189. [Google Scholar] [CrossRef]
  126. Jorgovanovic, D.; Song, M.; Wang, L.; Zhang, Y. Roles of IFN-γ in tumor progression and regression: A review. Biomark. Res. 2020, 8, 49. [Google Scholar] [CrossRef]
  127. Bellucci, R.; Martin, A.; Bommarito, D.; Wang, K.; Hansen, S.H.; Freeman, G.J.; Ritz, J. Interferon-γ-induced activation of JAK1 and JAK2 suppresses tumor cell susceptibility to NK cells through upregulation of PD-L1 expression. Oncoimmunology 2015, 4, e1008824. [Google Scholar] [CrossRef]
  128. Mimura, K.; The, J.L.; Okayama, H.; Shiraishi, K.; Kua, L.F.; Koh, V.; Smoot, D.T.; Ashktorab, H.; Oike, T.; Suzuki, Y.; et al. PD-L1 expression is mainly regulated by interferon gamma associated with JAK-STAT pathway in gastric cancer. Cancer Sci. 2018, 109, 43–53. [Google Scholar] [CrossRef]
  129. Zhao, T.; Li, Y.; Zhang, J.; Zhang, B. PD-L1 expression increased by IFN-γ via JAK2-STAT1 signaling and predicts a poor survival in colorectal cancer. Oncol. Lett. 2020, 20, 1127–1134. [Google Scholar] [CrossRef]
  130. Calik, I.; Calik, M.; Turken, G.; Ozercan, I.H.; Dagli, A.F.; Artas, G.; Sarikaya, B. Intratumoral Cytotoxic T-Lymphocyte Density and PD-L1 Expression Are Prognostic Biomarkers for Patients with Colorectal Cancer. Medicina 2019, 55, 723. [Google Scholar] [CrossRef]
  131. Carson, W.E.; Dierksheide, J.E.; Jabbour, S.; Anghelina, M.; Bouchard, P.; Ku, G.; Yu, H.; Baumann, H.; Shah, M.H.; Cooper, M.A.; et al. Coadministration of interleukin-18 and interleukin-12 induces a fatal inflammatory response in mice: Critical role of natural killer cell interferon-γ production and STAT-mediated signal transduction. Blood 2000, 96, 1465–1473. [Google Scholar] [CrossRef]
  132. Chaisavaneeyakorn, S.; Othoro, C.; Shi, Y.P.; Otieno, J.; Chaiyaroj, S.C.; Lal, A.A.; Udhayakumar, V. Relationship between plasma Interleukin-12 (IL-12) and IL-18 levels and severe malarial anemia in an area of holoendemicity in western Kenya. Clin. Diagn. Lab. Immunol. 2003, 10, 362–366. [Google Scholar] [CrossRef] [PubMed]
  133. Feng, X.; Zhang, Z.; Sun, P.; Song, G.; Wang, L.; Sun, Z.; Yuan, N.; Wang, Q.; Lun, L. Interleukin-18 Is a Prognostic Marker and Plays a Tumor Suppressive Role in Colon Cancer. Disease Markers 2020, 2020, 6439614. [Google Scholar] [CrossRef] [PubMed]
  134. Braumüller, H.; Mauerer, B.; Andris, J.; Berlin, C.; Wieder, T.; Kesselring, R. The Cytokine Network in Colorectal Cancer: Implications for New Treatment Strategies. Cells 2023, 12, 138. [Google Scholar] [CrossRef] [PubMed]
  135. Briukhovetska, D.; Dörr, J.; Endres, S.; Libby, P.; Dinarello, C.A.; Kobold, S. Interleukins in cancer: From biology to therapy. Nat. Rev. Cancer 2021, 21, 481–499. [Google Scholar] [CrossRef]
  136. Chen, X.W.; Zhou, S.F. Inflammation, cytokines, the IL-17/IL-6/STAT3/NF-κB axis, and tumorigenesis. Drug. Des. DevelTher. 2015, 9, 2941–2946. [Google Scholar] [CrossRef]
  137. Cheng, K.J.; Mejia, M.E.H.; Khong, T.L.; Mohd, Z.S.; Thavagnanam, S.; Ibrahim, Z.A. IL-1α and colorectal cancer pathogenesis: Enthralling candidate for anti-cancer therapy. Crit. Rev. Oncol. Hematol. 2021, 163, 103398. [Google Scholar] [CrossRef]
  138. Li, Y.; Wang, L.; Pappan, L. IL-1β promotes stemness and invasiveness of colon cancer cells through Zeb1 activation. Mol. Cancer 2012, 11, 87. [Google Scholar] [CrossRef]
  139. Lubberink, M.; Golla, S.S.; Jonasson, M.; Rubin, K.; Glimelius, B.; Sörensen, J.; Nygren, P. (15)O-Water PET Study of the Effect of Imatinib.; a Selective Platelet-Derived Growth Factor Receptor Inhibitor.; Versus Anakinra.; an IL-1R Antagonist.; on Water-Perfusable Tissue Fraction in Colorectal Cancer Metastases. J. Nucl. Med. 2015, 56, 1144–1149. [Google Scholar] [CrossRef]
  140. Pastille, E.; Wasmer, M.H.; Adamczyk, A.; Vu, V.P.; Mager, L.F.; Phuong, N.N.T.; Palmieri, V.; Simillion, C.; Hansen, W.; Kasper, S.; et al. The IL-33/ST2 pathway shapes the regulatory T cell phenotype to promote intestinal cancer. Mucosal Immunol. 2019, 12, 990–1003. [Google Scholar] [CrossRef]
  141. Bergman, M.; Levin, G.S.; Bessler, H.; Djaldetti, M.; Salman, H. Resveratrol affects the cross talk between immune and colon cancer cells. Biomed. Pharmacother. 2013, 67, 43–47. [Google Scholar] [CrossRef] [PubMed]
  142. Idris, A.; Ghazali, N.B.; Koh, D. Interleukin 1β—A Potential Salivary Biomarker for Cancer Progression? Biomark. Cancer 2015, 7, BIC.S25375. [Google Scholar] [CrossRef] [PubMed]
  143. Li, B.; Wang, F.; Ma, C.; Hao, T.; Geng, L.; Jiang, H. Predictive value of IL-18 and IL-10 in the prognosis of patients with colorectal cancer. Oncol. Lett. 2019, 18, 713–719. [Google Scholar] [CrossRef] [PubMed]
  144. Nikiteas, N.; Yannopoulos, A.; Chatzitheofylaktou, A.; Tsigris, C. Heterozygosity for Interleukin-18 -607 A/C Polymorphism is Associated with Risk for Colorectal Cancer. Anticancer Res. 2007, 27, 3849–3853. Available online: https://ar.iiarjournals.org/content/anticanres/27/6B/3849.full.pdf (accessed on 17 March 2023). [PubMed]
  145. Misa, B.I.; Diakowska, D.; Korpacka, K.M. Local and Systemic IL-7 Concentration in Gastrointestinal-Tract Cancers. Medicina 2019, 55, 262. [Google Scholar] [CrossRef] [PubMed]
  146. Chen, J.; Gong, C.; Mao, H.; Li, Z.; Fang, Z.; Chen, Q.; Lin, M.; Jiang, X.; Hu, Y.; Wang, W.; et al. E2F1/SP3/STAT6 axis is required for IL-4-induced epithelial-mesenchymal transition of colorectal cancer cells. Int. J. Oncol. 2018, 53, 567–578. [Google Scholar] [CrossRef] [PubMed]
  147. Liu, X.; Zhu, L.; Lu, X.; Bian, H.; Wu, X.; Yang, W.; Qin, Q. IL-33/ST2 pathway contributes to metastasis of human colorectal cancer. Biochem. Biophys. Res. Commun. 2014, 453, 486–492. [Google Scholar] [CrossRef]
  148. Rocca, Y.S.; Roberti, M.P.; Juliá, E.P.; Pampena, M.B.; Bruno, L.; Rivero, S.; Huertas, E.; Sánchez Loria, F.; Pairola, A.; Caignard, A.; et al. Phenotypic and Functional Dysregulated Blood NK Cells in Colorectal Cancer Patients Can Be Activated by Cetuximab Plus IL-2 or IL-15. Front. Immunol. 2016, 7, 413. [Google Scholar] [CrossRef]
  149. Wang, C.; Lu, Y.; Chen, L.; Gao, T.; Yang, Q.; Zhu, C.; Chen, Y. Th9 cells are subjected to PD-1/PD-L1-mediated inhibition and are capable of promoting CD8 T cell expansion through IL-9R in colorectal cancer. Int. Immunopharmacol. 2020, 78, 106019. [Google Scholar] [CrossRef]
  150. Fukushima, Y.; Iinuma, H.; Tsukamoto, M.; Matsuda, K.; Hashiguchi, Y. Clinical significance of microRNA-21 as a biomarker in each Dukes’ stage of colorectal cancer. Oncol. Rep. 2015, 33, 573–582. [Google Scholar] [CrossRef]
  151. Steele, N.; Anthony, A.; Saunders, M.; Esmarck, B.; Ehrnrooth, E.; Kristjansen, P.E.; Nihlén, A.; Hansen, L.T.; Cassidy, J. A phase 1 trial of recombinant human IL-21 in combination with cetuximab in patients with metastatic colorectal cancer. Br. J. Cancer 2012, 106, 793–798. [Google Scholar] [CrossRef] [PubMed]
  152. Stolfi, C.; Rizzo, A.; Franzè, E.; Rotondi, A.; Fantini, M.C.; Sarra, M.; Caruso, R.; Monteleone, I.; Sileri, P.; Franceschilli, L. Involvement of interleukin-21 in the regulation of colitis-associated colon cancer. J. Exp. Med. 2011, 208, 2279–2290. [Google Scholar] [CrossRef] [PubMed]
  153. Belluco, C.; Nitti, D.; Frantz, M.; Toppan, P.; Basso, D.; Plebani, M.; Lise, M.; Jessup, J.M. Interleukin-6 Blood Level Is Associated with Circulating Carcinoembryonic Antigen and Prognosis in Patients with Colorectal Cancer. Ann. Surg. Oncol. 2000, 7, 133–138. [Google Scholar] [CrossRef] [PubMed]
  154. Knüpfer, H.; Preiss, R. Serum interleukin-6 levels in colorectal cancer patients—A summary of published results. Int. J. Color. Dis. 2010, 25, 135–140. [Google Scholar] [CrossRef]
  155. St John, M.A.; Li, Y.; Zhou, X.; Denny, P.; Ho, C.M.; Montemagno, C.; Shi, W.; Qi, F.; Wu, B.; Sinha, U.; et al. Interleukin 6 and Interleukin 8 as Potential Biomarkers for Oral Cavity and Oropharyngeal Squamous Cell Carcinoma. Arch. Otolaryngol. Head Neck Surg. 2004, 130, 929–935. [Google Scholar] [CrossRef] [PubMed]
  156. Vainer, N.; Dehlendorff, C.; Johansen, J.S. Systematic literature review of IL-6 as a biomarker or treatment target in patients with gastric, bile duct, pancreatic and colorectal cancer. Oncotarget 2018, 9, 29820–29841. [Google Scholar] [CrossRef]
  157. Huynh, J.; Chand, A.; Ernst, M. IL-11 as a therapeutic target to treat colorectal cancer. Cancer Res. 2018, 78, 5733. [Google Scholar] [CrossRef]
  158. Putoczki, T.L.; Thiem, S.; Loving, A.; Busuttil, R.A.; Wilson, N.J.; Ziegler, P.K.; Nguyen, P.M.; Preaudet, A.; Farid, R.; Edwards, K.M.; et al. Interleukin-11 is the dominant IL-6 family cytokine during gastrointestinal tumorigenesis and can be targeted therapeutically. Cancer Cell 2013, 24, 257–271. [Google Scholar] [CrossRef]
  159. Yoshizaki, A.; Nakayama, T.; Yamazumi, K.; Yakata, Y.; Taba, M.; Sekine, I. Expression of interleukin (IL)-11 and IL-11 receptor in human colorectal adenocarcinoma: IL-11 up-regulation of the invasive and proliferative activity of human colorectal carcinoma cells. Int. J. Oncol. 2006, 29, 869–876. [Google Scholar] [CrossRef]
  160. Sun, Q.; Sun, F.; Wang, B.; Liu, S.; Niu, W.; Liu, E.; Peng, C.; Wang, J.; Gao, H.; Liang, B.; et al. Interleukin-8 promotes cell migration through integrin αvβ6 upregulation in colorectal cancer. Cancer Lett. 2014, 354, 245–253. [Google Scholar] [CrossRef]
  161. Shimizu, M.; Tanaka, N. IL-8-induced O-GlcNAc modification via GLUT3 and GFAT regulates cancer stem cell-like properties in colon and lung cancer cells. Oncogene 2019, 38, 1520–1533. [Google Scholar] [CrossRef] [PubMed]
  162. Xia, W.; Chen, W.; Zhang, Z.; Wu, D.; Wu, P.; Chen, Z.; Li, C.; Huang, J. Prognostic value, clinicopathologic features and diagnostic accuracy of interleukin-8 in colorectal cancer: A meta-analysis. PLoS ONE 2015, 10, e0123484. [Google Scholar] [CrossRef] [PubMed]
  163. Miteva, L.D.; Stanilov, N.S.; Deliysky, T.S.; Stanilova, S.A. Significance of−1082A/G polymorphism of IL10 gene for progression of colorectal cancer and IL-10 expression. Tumor Biol. 2014, 35, 12655–12664. [Google Scholar] [CrossRef] [PubMed]
  164. O’Hara, R.J.; Greenman, J.; MacDonald, A.W.; Gaskell, K.M.; Topping, K.P.; Duthie, G.S.; Kerin, M.J.; Lee, P.W.; Monson, J.R.J. Advanced colorectal cancer is associated with impaired interleukin 12 and enhanced interleukin 10 production. Clin. Cancer Res. 1998, 4, 1943–1948. Available online: https://clincancerres.aacrjournals.org/content/clincanres/4/8/1943.full.pdf (accessed on 17 March 2023). [PubMed]
  165. Shouval, D.S.; Biswas, A.; Goettel, J.A.; McCann, K.; Conaway, E.; Redhu, N.S.; Mascanfroni, I.D.; Al Adham, Z.; Lavoie, S.; Ibourk, M.; et al. Interleukin-10 receptor signaling in innate immune cells regulates mucosal immune tolerance and anti-inflammatory macrophage function. Immunity 2014, 40, 706–719. [Google Scholar] [CrossRef] [PubMed]
  166. Doulabi, H.; Rastin, M.; Shabahangh, H.; Maddah, G.; Abdollahi, A.; Nosratabadi, R.; Esmaeili, S.A.; Mahmoudi, M. Analysis of Th22.; Th17 and CD4(+)cells co-producing IL-17/IL-22 at different stages of human colon cancer. Biomed. Pharmacother. 2018, 103, 1101–1106. [Google Scholar] [CrossRef] [PubMed]
  167. Yu, L.Z.; Wang, H.Y.; Yang, S.P.; Yuan, Z.P.; Xu, F.Y.; Sun, C.; Shi, R.H. Expression of interleukin-22/STAT3 signaling pathway in ulcerative colitis and related carcinogenesis. World J. Gastroenterol. WJG 2013, 19, 2638. [Google Scholar] [CrossRef]
  168. Wang, C.; Gong, G.; Sheh, A.; Muthupalani, S.; Bryant, E.M.; Puglisi, D.A.; Holcombe, H.; Conaway, E.A.; Parry, N.A.P.; Bakthavatchalu, V.; et al. Interleukin-22 drives nitric oxide-dependent DNA damage and dysplasia in a murine model of colitis-associated cancer. Mucosal Immunol. 2017, 10, 1504–1517. [Google Scholar] [CrossRef]
  169. Moseley, T.A.; Haudenschild, D.R.; Rose, L.; Reddi, A.H. Interleukin-17 family and IL-17 receptors. Cytokine Growth Factor Rev. 2013, 14, 155–174. [Google Scholar] [CrossRef]
  170. Razi, S.; Baradaran, N.B.; Keshavarz-Fathi, M.; Rezaei, N. IL-17 and colorectal cancer: From carcinogenesis to treatment. Cytokine 2019, 116, 7–12. [Google Scholar] [CrossRef]
  171. Alinejad, V.; Dolati, S.; Motallebnezhad, M.; Yousefi, M. The role of IL17B-IL17RB signaling pathway in breast cancer. Biomed. Pharmacother. 2017, 88, 795–803. [Google Scholar] [CrossRef] [PubMed]
  172. Shamoun, L.; Skarstedt, M.; Andersson, R.E.; Wågsäter, D.; Dimberg, J. Association study on IL-4, IL-4Rα and IL-13 genetic polymorphisms in Swedish patients with colorectal cancer. Clin. Chim. Acta 2018, 487, 101–106. [Google Scholar] [CrossRef] [PubMed]
  173. Neurath, M.F. IL-23 in inflammatory bowel diseases and colon cancer. Cytokine Growth Factor Rev. 2019, 45, 1–8. [Google Scholar] [CrossRef] [PubMed]
  174. Zhang, L.; Li, J.; Li, L.; Zhang, J.; Wang, X.; Yang, C.; Li, Y.; Lan, F.; Lin, P. IL-23 selectively promotes the metastasis of colorectal carcinoma cells with impaired Socs3 expression via the STAT5 pathway. Carcinogenesis 2014, 35, 1330–1340. [Google Scholar] [CrossRef]
  175. Suzuki, H.; Ogawa, H.; Miura, K.; Haneda, S.; Watanabe, K.; Ohnuma, S.; Sasaki, H.; Sase, T.; Kimura, S.; Kajiwara, T.; et al. IL-23 directly enhances the proliferative and invasive activities of colorectal carcinoma. Oncol. Lett. 2012, 4, 199–204. [Google Scholar] [CrossRef]
  176. Hu, W.H.; Chang, C.D.; Liu, T.T.; Chen, H.H.; Hsiao, C.C.; Kang, H.Y.; Chuang, J.H. Association of sarcopenia and expression of interleukin-23 in colorectal cancer survival. Clin. Nutr. 2021, 40, 5322–5326. [Google Scholar] [CrossRef]
  177. Voronov, E.; Apte, R.N. IL-1 in Colon Inflammation.; Colon Carcinogenesis and Invasiveness of Colon Cancer. Cancer Microenviron. 2015, 8, 187–200. [Google Scholar] [CrossRef]
  178. Cohen, I.; Rider, P.; Carmi, Y.; Braiman, A.; Dotan, S.; White, M.R.; Voronov, E.; Martin, M.U.; Dinarello, C.A.; Apte, R.N. Differential release of chromatin-bound IL-1α discriminates between necrotic and apoptotic cell death by the ability to induce sterile inflammation. Proc. Natl. Acad. Sci. USA 2010, 107, 2574–2579. [Google Scholar] [CrossRef]
  179. Rider, P.; Carmi, Y.; Guttman, O.; Braiman, A.; Cohen, I.; Voronov, E.; White, M.R.; Dinarello, C.A.; Apte, R.N. IL-1alpha and IL-1beta recruit different myeloid cells and promote different stages of sterile inflammation. J. Immunol. 2011, 187, 4835–4843. [Google Scholar] [CrossRef]
  180. Voronov, E.; Yaron, C.; Ron, N.A. The role IL-1 in tumor-mediated angiogenesis. Front. Physiol. 2014, 5, 114. [Google Scholar] [CrossRef]
  181. Gunter, M.J.; Canzian, F.; Landi, S.; Chanock, S.J.; Sinha, R.; Rothman, N. Inflammation-related gene polymorphisms and colorectal adenoma. Cancer Epidemiol. Biomark. Prev. 2006, 15, 1126–1131. [Google Scholar] [CrossRef] [PubMed]
  182. Graziano, F.; Ruzzo, A.; Canestrari, E.; Loupakis, F.; Santini, D.; Rulli, E.; Humar, B.; Galluccio, N.; Bisonni, R.; Floriani, I.; et al. Variations in the interleukin-1 receptor antagonist gene impact on survival of patients with advanced colorectal cancer. Pharm. J. 2009, 9, 78–84. [Google Scholar] [CrossRef] [PubMed]
  183. Maeda, S.; Hsu, L.C.; Liu, H.; Bankston, L.A.; Iimura, M.; Kagnoff, M.F.; Eckmann, L.; Karin, M. Nod2 mutation in Crohn’s disease potentiates NF-kappaB activity and IL-1beta processing. Science 2005, 307, 734–738. [Google Scholar] [CrossRef] [PubMed]
  184. Li, Y.-P.; Philip, S. Proinflammatory cytokines tumor necrosis factor-alpha and IL-6, but not IL-1, down-regulate the osteocalcin gene promoter. J. Immunol. 1992, 148, 788–794. [Google Scholar] [CrossRef] [PubMed]
  185. Chen, Y.; Yang, Z.; Deng, B.; Wu, D.; Quan, Y.; Min, Z. Interleukin 1β/1RA axis in colorectal cancer regulates tumor invasion, proliferation and apoptosis via autophagy. Oncol. Rep. 2020, 43, 908–918. [Google Scholar] [CrossRef]
  186. Kolls, J.K.; Anders, L. Interleukin-17 family members and inflammation. Immunity 2004, 21, 467–476. [Google Scholar] [CrossRef]
  187. Feng, H.; Ying, R.; Chai, T.; Chen, H.; Ju, H. The association between IL-17 gene variants and risk of colorectal cancer in a Chinese population: A case-control study. Biosci. Rep. 2019, 39, BSR20190013. [Google Scholar] [CrossRef]
  188. Wu, D.; Wu, P.; Huang, Q.; Liu, Y.; Ye, J.; Huang, J. Interleukin-17: A promoter in colorectal cancer progression. Clin. Dev. Immunol. 2013, 2013, 436307. [Google Scholar] [CrossRef]
  189. Nemati, K.; Golmoghaddam, H.; Hosseini, S.V.; Ghaderi, A.; Doroudchi, M. Interleukin-17FT7488 allele is associated with a decreased risk of colorectal cancer and tumor progression. Gene 2015, 561, 88–94. [Google Scholar] [CrossRef]
  190. Tseng, J.Y.; Yang, C.Y.; Liang, S.C.; Liu, R.S.; Yang, S.H.; Lin, J.K.; Chen, Y.M.; Wu, Y.C.; Jiang, J.K.; Lin, C.H. Interleukin-17A modulates circulating tumor cells in tumor draining vein of colorectal cancers and affects metastases. Clin. Cancer Res. 2014, 20, 2885–2897. [Google Scholar] [CrossRef]
  191. Cui, G.; Yuan, A.; Goll, R.; Florholmen, J. IL-17A in the tumor microenvironment of the human colorectal adenoma-carcinoma sequence. Scand. J. Gastroenterol. 2012, 47, 1304–1312. [Google Scholar] [CrossRef] [PubMed]
  192. Straus, D.S. TNFα and IL-17 cooperatively stimulate glucose metabolism and growth factor production in human colorectal cancer cells. Mol. Cancer 2013, 12, 78. [Google Scholar] [CrossRef] [PubMed]
  193. Chae, W.J.; Gibson, T.F.; Zelterman, D.; Hao, L.; Henegariu, O.; Bothwell, A.L. Ablation of IL-17A abrogates progression of spontaneous intestinal tumorigenesis. Proc. Natl. Acad. Sci. USA 2010, 107, 5540–5544. [Google Scholar] [CrossRef] [PubMed]
  194. Liu, J.; Duan, Y.; Cheng, X.; Chen, X.; Xie, W.; Long, H.; Lin, Z.; Zhu, B. IL-17 is associated with poor prognosis and promotes angiogenesis via stimulating VEGF production of cancer cells in colorectal carcinoma. Biochem. Res. Commun. 2011, 407, 348–354. [Google Scholar] [CrossRef]
  195. Sobhani, I.; Tap, J.; Roudot-Thoraval, F.; Roperch, J.P.; Letulle, S.; Langella, P.; Corthier, G.; Van Nhieu, J.T.; Furet, J.P. Microbial dysbiosis in colorectal cancer (CRC) patients. PLoS ONE 2011, 6, e16393. [Google Scholar] [CrossRef]
  196. Radosavljevic, G.; Ljujic, B.; Jovanovic, I.; Srzentic, Z.; Pavlovic, S.; Zdravkovic, N.; Milovanovic, M.; Bankovic, D.; Knezevic, M.; Acimovic, L.; et al. Interleukin-17 may be a valuable serum tumor marker in patients with colorectal carcinoma. Neoplasma 2010, 57, 135–144. [Google Scholar] [CrossRef]
  197. Al Obeed, O.A.; Vaali-Mohamed, M.A.; Alkhayal, K.A.; Bin Traiki, T.A.; Zubaidi, A.M.; Arafah, M.; Harris, R.A.; Khan, Z.; Abdulla, M.H. IL-17 and colorectal cancer risk in the Middle East: Gene polymorphisms and expression. Cancer Manag. Res. 2018, 10, 2653. [Google Scholar] [CrossRef]
  198. Le Gouvello, S.; Bastuji-Garin, S.; Aloulou, N.; Mansour, H.; Chaumette, M.T.; Berrehar, F.; Seikour, A.; Charachon, A.; Karoui, M.; Leroy, K.; et al. High prevalence of Foxp3 and IL17 in MMR-proficient colorectal carcinomas. Gut 2008, 57, 772–779. [Google Scholar] [CrossRef]
  199. Huang, Y.H.; Cao, Y.F.; Jiang, Z.Y.; Zhang, S.; Gao, F. Th22 cell accumulation is associated with colorectal cancer development. World J. Gastroenterol. 2015, 21, 4216–4224. [Google Scholar] [CrossRef]
  200. Wu, T.; Wang, Z.; Liu, Y.; Mei, Z.; Wang, G.; Liang, Z.; Cui, A.; Hu, X.; Cui, L.; Yang, Y.; et al. Interleukin 22 protects colorectal cancer cells from chemotherapy by activating the STAT3 pathway and inducing autocrine expression of interleukin 8. Clin. Immunol. 2014, 154, 116–126. [Google Scholar] [CrossRef]
  201. Harrison, C. IL-22: Linking inflammation and cancer. Nat. Rev. Drug. Discov. 2013, 12, 505. [Google Scholar] [CrossRef] [PubMed]
  202. Thompson, C.L.; Plummer, S.J.; Tucker, T.C.; Casey, G.; Li, L. Interleukin-22 genetic polymorphisms and risk of colon cancer. Cancer Causes Control 2010, 21, 1165–1170. [Google Scholar] [CrossRef] [PubMed]
  203. Jiang, R.; Wang, H.; Deng, L.; Hou, J.; Shi, R.; Yao, M.; Gao, Y.; Yao, A.; Wang, X.; Yu, L.; et al. IL-22 is related to development of human colon cancer by activation of STAT3. BMC Cancer 2013, 13, 59. [Google Scholar] [CrossRef] [PubMed]
  204. Bobe, G.; Albert, P.S.; Sansbury, L.B.; Lanza, E.; Schatzkin, A.; Colburn, N.H.; Cross, A.J. Interleukin-6 as a Potential Indicator for Prevention of High-Risk Adenoma Recurrence by Dietary Flavonols in the Polyp Prevention Trial. Cancer Prev. Res. 2010, 3, 764–775. [Google Scholar] [CrossRef] [PubMed]
  205. Kim, S.; Keku, T.O.; Martin, C.; Galanko, J.; Woosley, J.T.; Schroeder, J.C.; Satia, J.A.; Halabi, S.; Sandler, R.S. Circulating Levels of Inflammatory Cytokines and Risk of Colorectal Adenomas. Cancer Res. 2008, 68, 323–328. [Google Scholar] [CrossRef] [PubMed]
  206. Guo, Y.; Xu, F.; Lu, T.; Duan, Z.; Zhang, Z. Interleukin-6 signaling pathway in targeted therapy for cancer. Cancer Treat. Rev. 2012, 38, 904–910. [Google Scholar] [CrossRef]
  207. Waetzig, G.H.; Rose-John, S. Hitting a complex target: An update on interleukin-6 trans-signalling. Expert Opin. Ther. Targets 2012, 16, 225–236. [Google Scholar] [CrossRef]
  208. Wang, Z.; Wu, P.; Wu, D.; Zhang, Z.; Hu, G.; Zhao, S.; Lai, Y.; Huang, J. Prognostic and clinicopathological significance of serum interleukin-6 expression in colorectal cancer: A systematic review and meta-analysis. OncoTargets Ther. 2015, 8, 3793–3801. [Google Scholar] [CrossRef]
  209. Waldner, M.J.; Foersch, S.; Neurath, M.F. Interleukin-6--a key regulator of colorectal cancer development. Int. J. Biol. Sci. 2012, 8, 1248–1253. [Google Scholar] [CrossRef]
  210. Johnson, D.E.; O’Keefe, R.A.; Grandis, J.R. Targeting the IL-6/JAK/STAT3 signalling axis in cancer. Nat. Rev. Clin. Oncol. 2018, 15, 234–248. [Google Scholar] [CrossRef]
  211. Ham, I.H.; Oh, H.J.; Jin, H.; Bae, C.A.; Jeon, S.M.; Choi, K.S.; Son, S.Y.; Han, S.U.; Brekken, R.A.; Lee, D. Targeting interleukin-6 as a strategy to overcome stroma-induced resistance to chemotherapy in gastric cancer. Mol. Cancer 2019, 18, 68. [Google Scholar] [CrossRef] [PubMed]
  212. Hue, S.; Ahern, P.; Buonocore, S.; Kullberg, M.C.; Cua, D.J.; McKenzie, B.S.; Powrie, F.; Maloy, K.J. Interleukin-23 drives innate and T cell-mediated intestinal inflammation. J. Exp. Med. 2006, 203, 2473–2483. [Google Scholar] [CrossRef] [PubMed]
  213. Oppmann, B.; Lesley, R.; Blom, B.; Timans, J.C.; Xu, Y.; Hunte, B.; Vega, F.; Yu, N.; Wang, J.; Singh, K.; et al. Novel p19 protein engages IL-12p40 to form a cytokine, IL-23, with biological activities similar as well as distinct from IL-12. Immunity 2000, 13, 715–725. [Google Scholar] [CrossRef] [PubMed]
  214. Langowski, J.L.; Zhang, X.; Wu, L.; Mattson, J.D.; Chen, T.; Smith, K.; Basham, B.; McClanahan, T.; Kastelein, R.A.; Oft, M. IL-23 promotes tumour incidence and growth. Nature 2006, 442, 461–465. [Google Scholar] [CrossRef] [PubMed]
  215. Liu, Z.; Zhang, J.Y.; Yang, Y.J.; Chang, K.; Wang, Q.F.; Kong, Y.Y.; Dai, B. High IL-23+ cells infiltration correlates with worse clinical outcomes and abiraterone effectiveness in patients with prostate cancer. Asian J. Androl. 2022, 24, 147–153. [Google Scholar] [CrossRef] [PubMed]
  216. Kortylewski, M.; Xin, H.; Kujawski, M.; Lee, H.; Liu, Y.; Harris, T.; Drake, C.; Pardoll, D.; Yu, H. Regulation of the IL-23 and IL-12 Balance by Stat3 Signaling in the Tumor Microenvironment. Cancer Cell 2009, 15, 114–123. [Google Scholar] [CrossRef] [PubMed]
  217. Grivennikov, S.I.; Wang, K.; Mucida, D.; Stewart, C.A.; Schnabl, B.; Jauch, D.; Taniguchi, K.; Yu, G.Y.; Osterreicher, C.H.; Hung, K.E.; et al. Adenoma-linked barrier defects and microbial products drive IL-23/IL-17-mediated tumour growth. Nature 2012, 491, 254–258. [Google Scholar] [CrossRef] [PubMed]
  218. Liang, S.C.; Tan, X.Y.; Luxenberg, D.P.; Karim, R.; Dunussi-Joannopoulos, K.; Collins, M.; Fouser, L.A. Interleukin (IL)-22 and IL-17 are coexpressed by Th17 cells and cooperatively enhance expression of antimicrobial peptides. J. Exp. Med. 2006, 203, 2271–2279. [Google Scholar] [CrossRef] [PubMed]
  219. Buonocore, S.; Ahern, P.P.; Uhlig, H.H.; Ivanov, I.I.; Littman, D.R.; Maloy, K.J.; Powrie, F. Innate lymphoid cells drive interleukin-23-dependent innate intestinal pathology. Nature 2010, 464, 1371–1375. [Google Scholar] [CrossRef] [PubMed]
  220. McGovern, D.; Powrie, F. The IL23 axis plays a key role in the pathogenesis of IBD. Gut 2007, 56, 1333–1336. [Google Scholar] [CrossRef] [PubMed]
  221. Lan, F.; Zhang, L.; Wu, J.; Zhang, J.; Zhang, S.; Li, K.; Qi, Y.; Lin, P. IL-23/IL-23R: Potential mediator of intestinal tumor progression from adenomatous polyps to colorectal carcinoma. Int. J. Color. Dis. 2011, 26, 1511–1518. [Google Scholar] [CrossRef] [PubMed]
  222. Lopetuso, L.R.; Scaldaferri, F.; Pizarro, T.T. Emerging role of the interleukin (IL)-33/ST2 axis in gut mucosal wound healing and fibrosis. Fibrogenesis Tissue Repair 2012, 5, 18. [Google Scholar] [CrossRef] [PubMed]
  223. Garlanda, C.; Dinarello, C.A.; Mantovani, A. The interleukin-1 family: Back to the future. Immunity 2013, 39, 1003–1018. [Google Scholar] [CrossRef] [PubMed]
  224. Schmitz, J.; Owyang, A.; Oldham, E.; Song, Y.; Murphy, E.; McClanahan, T.K.; Zurawski, G.; Moshrefi, M.; Qin, J.; Li, X.; et al. IL-33.; an interleukin-1-like cytokine that signals via the IL-1 receptor-related protein ST2 and induces T helper type 2-associated cytokines. Immunity 2005, 23, 479–490. [Google Scholar] [CrossRef]
  225. Wood, I.S.; Wang, B.; Trayhurn, P. IL-33.; a recently identified interleukin-1 gene family member.; is expressed in human adipocytes. Biochem. Biophys. Res. Commun. 2009, 384, 105–109. [Google Scholar] [CrossRef]
  226. Yangngam, S.; Thongchot, S.; Pongpaibul, A.; Vaeteewoottacharn, K.; Pinlaor, S.; Thuwajit, P.; Okada, S.; Hermoso, M.A.; Thuwajit, C. High level of interleukin-33 in cancer cells and cancer-associated fibroblasts correlates with good prognosis and suppressed migration in cholangiocarcinoma. J. Cancer 2020, 11, 6571–6581. [Google Scholar] [CrossRef]
  227. Fang, K.-M.; Yang, C.-S.; Lin, T.-C.; Chan, T.-C.; Tzeng, S.-F. Induced interleukin-33 expression enhances the tumorigenic activity of rat glioma cells. Neuro-Oncology 2014, 16, 552–566. [Google Scholar] [CrossRef]
  228. Gao, X.; Wang, X.; Yang, Q.; Zhao, X.; Wen, W.; Li, G.; Lu, J.; Qin, W.; Qi, Y.; Xie, F.; et al. Tumoral expression of IL-33 inhibits tumor growth and modifies the tumor microenvironment through CD8+ T and NK cells. J. Immunol. 2015, 194, 438–445. [Google Scholar] [CrossRef]
  229. Cui, G.; Qi, H.; Gundersen, M.D.; Yang, H.; Christiansen, I.; Sørbye, S.W.; Goll, R.; Florholmen, J. Dynamics of the IL-33/ST2 network in the progression of human colorectal adenoma to sporadic colorectal cancer. Cancer Immunol. Immunother. 2015, 64, 181–190. [Google Scholar] [CrossRef]
  230. O’Donnell, C.; Mahmoud, A.; Keane, J.; Murphy, C.; White, D.; Carey, S.; O’Riordain, M.; Bennett, M.W.; Brint, E.; Houston, A. An antitumorigenic role for the IL-33 receptor, ST2L, in colon cancer. Br. J. Cancer. 2016, 114, 37–43. [Google Scholar] [CrossRef]
  231. Lucarini, V.; Ziccheddu, G.; Macchia, I.; La Sorsa, V.; Peschiaroli, F.; Buccione, C.; Sistigu, A.; Sanchez, M.; Andreone, S.; D’Urso, M.T.; et al. IL-33 restricts tumor growth and inhibits pulmonary metastasis in melanoma-bearing mice through eosinophils. Oncoimmunology 2017, 6, e1317420. [Google Scholar] [CrossRef] [PubMed]
  232. Santana Carrero, R.M.; Beceren-Braun, F.; Rivas, S.C.; Hegde, S.M.; Gangadharan, A.; Plote, D.; Pham, G.; Anthony, S.M.; Schluns, K.S. IL-15 is a component of the inflammatory milieu in the tumor microenvironment promoting antitumor responses. Proc. Natl. Acad. Sci. USA 2019, 116, 599–608. [Google Scholar] [CrossRef] [PubMed]
  233. Klebanoff, C.A.; Finkelstein, S.E.; Surman, D.R.; Lichtman, M.K.; Gattinoni, L.; Theoret, M.R.; Grewal, N.; Spiess, P.J.; Antony, P.A.; Palmer, D.C. IL-15 enhances the in vivo antitumor activity of tumor-reactive CD8+ T cells. Proc. Natl. Acad. Sci. USA 2004, 101, 1969–1974. [Google Scholar] [CrossRef] [PubMed]
  234. Yu, P.; Bamford, R.N.; Waldmann, T.A. IL-15-dependent CD8+ CD122+ T cells ameliorate experimental autoimmune encephalomyelitis by modulating IL-17 production by CD4+ T cells. Eur. J. Immunol. 2014, 44, 3330–3341. [Google Scholar] [CrossRef] [PubMed]
  235. Mlecnik, B.; Bindea, G.; Angell, H.K.; Sasso, M.S.; Obenauf, A.C.; Fredriksen, T.; Lafontaine, L.; Bilocq, A.M.; Kirilovsky, A.; Tosolini, M.; et al. Functional network pipeline reveals genetic determinants associated with in situ lymphocyte proliferation and survival of cancer patients. Sci. Transl. Med. 2014, 6, 228ra237. [Google Scholar] [CrossRef] [PubMed]
  236. Curran, M.A.; Geiger, T.L.; Montalvo, W.; Kim, M.; Reiner, S.L.; Al-Shamkhani, A.; Sun, J.C.; Allison, J.P. Systemic 4-1BB activation induces a novel T cell phenotype driven by high expression of Eomesodermin. J. Exp. Med. 2013, 210, 743–755. [Google Scholar] [CrossRef]
  237. Kuniyasu, H.; Oue, N.; Nakae, D.; Tsutsumi, M.; Denda, A.; Tsujiuchi, T.; Yokozaki, H.; Yasui, W. Interleukin-15 expression is associated with malignant potential in colon cancer cells. Pathobiology 2001, 69, 86–95. [Google Scholar] [CrossRef]
  238. Chen, J.; Li, Z.Y.; Huang, S.Y.; Petersen, E.; Song, H.Q.; Zhou, D.H.; Zhu, X.Q. Protective efficacy of Toxoplasma gondiicalcium-dependent protein kinase 1 (TgCDPK1) adjuvated with recombinant IL-15 and IL-21 against experimental toxoplasmosis in mice. BMC Infect. Dis. 2014, 14, 487. [Google Scholar] [CrossRef]
  239. Berger, C.; Berger, M.; Hackman, R.C.; Gough, M.; Elliott, C.; Jensen, M.C.; Riddell, S.R. Safety and immunologic effects of IL-15 administration in nonhuman primates. Blood 2009, 114, 2417–2426. [Google Scholar] [CrossRef]
  240. Pilipow, K.; Roberto, A.; Roederer, M.; Waldmann, T.A.; Mavilio, D.; Lugli, E. IL15 and T-cell Stemness in T-cell-Based Cancer Immunotherapy. Cancer Res. 2015, 75, 5187–5193. [Google Scholar] [CrossRef]
  241. Lusty, E.; Poznanski, S.M.; Kwofie, K.; Mandur, T.S.; Lee, D.A.; Richards, C.D.; Ashkar, A.A. IL-18/IL-15/IL-12 synergy induces elevated and prolonged IFN-γ production by ex vivo expanded NK cells which is not due to enhanced STAT4 activation. Mol. Immunol. 2017, 88, 138–147. [Google Scholar] [CrossRef] [PubMed]
  242. Yasuda, K.; Nakanishi, K.; and Tsutsui, H. Interleukin-18 in Health and Disease. Int. J. Mol. Sci. 2019, 20, 649. [Google Scholar] [CrossRef] [PubMed]
  243. Gosmann, C.; Frazer, I.H.; Mattarollo, S.R.; Blumenthal, A. IL-18, but not IL-12, induces production of IFN-γ in the immunosuppressive environment of HPV16 E7 transgenic hyperplastic skin. J. Investig. Dermatol. 2014, 134, 2562–2569. [Google Scholar] [CrossRef]
  244. Formentini, A.; Braun, P.; Fricke, H.; Link, K.-H.; Henne-Bruns, D.; Kornmann, M. Expression of interleukin-4 and interleukin-13 and their receptors in colorectal cancer. Int. J. Color. Dis. 2012, 27, 1369–1376. [Google Scholar] [CrossRef]
  245. Shi, J.; Song, X.; Traub, B.; Luxenhofer, M.; Kornmann, M. Involvement of IL-4, IL-13 and their receptors in pancreatic cancer. Int. J. Mol. Sci. 2021, 22, 2998. [Google Scholar] [CrossRef]
  246. Barderas, R.; Bartolomé, R.A.; Fernandez-Acenero, M.J.; Torres, S.; Casal, J.I. High Expression of IL-13 Receptor α2 in Colorectal Cancer Is Associated with Invasion.; Liver Metastasis.; and Poor PrognosisRole of IL-13Rα2 in Colorectal Cancer Metastasis. Cancer Res. 2012, 72, 2780–2790. [Google Scholar] [CrossRef] [PubMed]
  247. Jin, R.; Hu, S.; Liu, X.; Guan, R.; Lu, L.; Lin, R. Intranasal instillation of miR-410 targeting IL-4/IL-13 attenuates airway inflammation in OVA-induced asthmatic mice. Mol. Med. Rep. 2019, 19, 895–900. [Google Scholar] [CrossRef]
  248. Wang, H.P.; Wang, Y.Y.; Pan, J.; Cen, R.; Cai, Y.K. Evaluation of specific fecal protein biochips for the diagnosis of colorectal cancer. World J. Gastroenterol. 2014, 20, 1332–1339. [Google Scholar] [CrossRef]
  249. Cho, Y.A.; Kim, J. Association of IL4, IL13, and IL4R polymorphisms with gastrointestinal cancer risk: A meta-analysis. J. Epidemiol. 2017, 27, 215–220. [Google Scholar] [CrossRef]
  250. Song, X.; Traub, B.; Shi, J.; Kornmann, M. Possible Roles of Interleukin-4 and -13 and Their Receptors in Gastric and Colon Cancer. Int. J. Mol. Sci. 2021, 22, 727. [Google Scholar] [CrossRef]
  251. Terada, H.; Urano, T.; Konno, H. Association of interleukin-8 and plasminogen activator system in the progression of colorectal cancer. Eur. Surg. Res. 2005, 37, 166–172. [Google Scholar] [CrossRef] [PubMed]
  252. Baggiolini, M.; Clark-Lewis, I. Interleukin-8, a chemotactic and inflammatory cytokine. FEBS Lett. 1992, 307, 97–101. [Google Scholar] [CrossRef] [PubMed]
  253. Ning, Y.; Lenz, H.-J. Targeting IL-8 in colorectal cancer. Expert Opin. Ther. Targets. 2012, 16, 491–497. [Google Scholar] [CrossRef] [PubMed]
  254. Jin, W.J.; Xu, J.M.; Xu, W.L.; Gu, D.H.; Li, P.W. Diagnostic value of interleukin-8 in colorectal cancer: A case-control study and meta-analysis. World J. Gastroenterol. 2014, 20, 16334. [Google Scholar] [CrossRef]
  255. Itoh, Y.; Joh, T.; Tanida, S.; Sasaki, M.; Kataoka, H.; Itoh, K.; Oshima, T.; Ogasawara, N.; Togawa, S.; Wada, T.; et al. IL-8 promotes cell proliferation and migration through metalloproteinase-cleavage proHB-EGF in human colon carcinoma cells. Cytokine 2005, 29, 275–282. [Google Scholar] [CrossRef]
  256. Rubie, C.; Frick, V.O.; Pfeil, S.; Wagner, M.; Kollmar, O.; Kopp, B.; Graber, S.; Rau, B.M.; Schilling, M.K. Correlation of IL-8 with induction.; progression and metastatic potential of colorectal cancer. World J. Gastroenterol. 2007, 13, 4996. [Google Scholar] [CrossRef]
  257. Galffy, G.; Mohammed, K.A.; Dowling, P.A.; Nasreen, N.; Ward, M.J.; Antony, V.B. Interleukin 8: An autocrine growth factor for malignant mesothelioma. Cancer Res. 1999, 59, 367–371. [Google Scholar]
  258. Brew, R.; Erikson, J.S.; West, D.C.; Kinsella, A.R.; Slavin, J.; Christmas, S.E. Interleukin-8 as an autocrine growth factor for human colon carcinoma cells in vitro. Cytokine 2000, 12, 78–85. [Google Scholar] [CrossRef]
  259. Haqqani, A.S.; Sandhu, J.K.; Birnboim, H.C. Expression of interleukin-8 promotes neutrophil infiltration and genetic instability in mutatect tumors. Neoplasia 2000, 2, 561–568. [Google Scholar] [CrossRef]
  260. Bazzichetto, C.; Milella, M.; Zampiva, I.; Simionato, F.; Amoreo, C.A.; Buglioni, S.; Pacelli, C.; Le Pera, L.; Colombo, T.; Bria, E.; et al. Interleukin-8 in Colorectal Cancer: A Systematic Review and Meta-Analysis of Its Potential Role as a Prognostic Biomarker. Biomedicines 2022, 10, 2631. [Google Scholar] [CrossRef]
  261. Lee, Y.S.; Choi, I.; Ning, Y.; Kim, N.Y.; Khatchadourian, V.; Yang, D.; Chung, H.K.; Choi, D.; LaBonte, M.J.; Ladner, R.D.; et al. Interleukin-8 and its receptor CXCR2 in the tumour microenvironment promote colon cancer growth.; progression and metastasis. Br. J. Cancer 2012, 106, 1833–1841. [Google Scholar] [CrossRef] [PubMed]
  262. Maxwell, P.J.; Gallagher, R.; Seaton, A.; Wilson, C.; Scullin, P.; Pettigrew, J.; Stratford, I.J.; Williams, K.J.; Johnston, P.G.; Waugh, D.J. HIF-1 and NF-κB-mediated upregulation of CXCR1 and CXCR2 expression promotes cell survival in hypoxic prostate cancer cells. Oncogene 2007, 26, 7333–7345. [Google Scholar] [CrossRef]
  263. Moreno-Guerrero, S.S.; Ramírez-Pacheco, A.; Rocha-Ramírez, L.M.; Hernández-Pliego, G.; Eguía-Aguilar, P.; Escobar-Sánchez, M.A.; Reyes-López, A.; Juárez-Villegas, L.E.; Sienra-Monge, J.J.L. Association of Genetic Polymorphisms and Serum Levels of IL-6 and IL-8 with the Prognosis in Children with Neuroblastoma. Cancers 2021, 13, 529. [Google Scholar] [CrossRef] [PubMed]
  264. Gordon, M.A.; Gil, J.; Lu, B.; Zhang, W.; Yang, D.; Yun, J.; Schneider, S.; Groshen, S.; Iqbal, S.; Press, O.A.; et al. Genomic profiling associated with recurrence in patients with rectal cancer treated with chemoradiation. Pharmacogenomics 2006, 7, 67–88. [Google Scholar] [CrossRef] [PubMed]
  265. Ning, Y.; Manegold, P.C.; Hong, Y.K.; Zhang, W.; Pohl, A.; Lurje, G.; Winder, T.; Yang, D.; LaBonte, M.J.; Wilson, P.M.; et al. Interleukin-8 is vivo in colon cancer cell line models. Int. J. Cancer 2011, 128, 2038–2049. [Google Scholar] [CrossRef] [PubMed]
  266. Ackermann, A.; Lafferton, B.; Plotz, G.; Zeuzem, S.; Brieger, A. Expression and secretion of the pro-inflammatory cytokine IL-8 is increased in colorectal cancer cells following the knockdown of non-erythroid spectrin αII. Int. J. Oncol. 2020, 56, 1551–1564. [Google Scholar] [CrossRef]
  267. Howlett, M.; Giraud, A.S.; Lescesen, H.; Jackson, C.B.; Kalantzis, A.; Van Driel, I.R.; Robb, L.; Van der Hoek, M.; Ernst, M.; Minamoto, T.; et al. The interleukin-6 family cytokine interleukin-11 regulates homeostatic epithelial cell turnover and promotes gastric tumor development. Gastroenterology 2009, 136, 967–977. [Google Scholar] [CrossRef]
  268. Jones, S.A.; Jenkins, B.J. Recent insights into targeting the IL-6 cytokine family in inflammatory diseases and cancer. Nat. Rev. Immunol. 2018, 18, 773–789. [Google Scholar] [CrossRef]
  269. Yamazumi, K.; Nakayama, T.; Kusaba, T.; Wen, C.Y.; Yoshizaki, A.; Yakata, Y.; Nagayasu, T.; Sekine, I. Expression of interleukin-11 and interleukin-11 receptor alpha in human colorectal adenocarcinoma; immunohistochemical analyses and correlation with clinicopathological factors. World J. Gastroenterol. 2006, 12, 317–321. [Google Scholar] [CrossRef]
  270. West, N.R. Coordination of immune-stroma crosstalk by IL-6 family cytokines. Front. Immunol. 2019, 10, 1093. [Google Scholar] [CrossRef]
  271. Kiessling, S.; Muller-Newen, G.; Leeb, S.N.; Hausmann, M.; Rath, H.C.; Strater, J.; Spottl, T.; Schlottmann, K.; Grossmann, J.; Montero-Julian, F.A.; et al. Functional expression of the interleukin-11 receptor α-chain and evidence of antiapoptotic effects in human colonic epithelial cells. J. Biol. Chem. 2004, 279, 10304–10315. [Google Scholar] [CrossRef] [PubMed]
  272. Nishina, T.; Deguchi, Y.; Ohshima, D.; Wakami, T.; Masato, O.; Shigeyuki, S.; Saroshi, U.; Soh, Y.; Mika, K.; Eri, N.; et al. Interleukin-11-expressing fibroblasts have a unique gene signature correlated with poor prognosis of colorectal cancer. Nat. Commun. 2021, 12, 2281. [Google Scholar] [CrossRef] [PubMed]
  273. Andoh, A.; Zhang, Z.; Inatomi, O.; Fujino, S.; Deguchi, Y.; Araki, Y.; Tsujikawa, T.; Kitoh, K.; Kim-Mitsuyama, S.; Takayanagi, A.; et al. Interleukin-22.; a member of the IL-10 subfamily.; induces inflammatory responses in colonic subepithelial myofibroblasts. Gastroenterology 2005, 129, 969–984. [Google Scholar] [CrossRef]
  274. Bamba, S.; Andoh, A.; Yasui, H.; Makino, J.; Kim, S.; Fujiyama, Y. Regulation of IL-11 expression in intestinal myofibroblasts: Role of c-Jun AP-1-and MAPK-dependent pathways. Am. J. Physiol. Gastrointest. Liver Physiol. 2003, 285, G529–G538. [Google Scholar] [CrossRef] [PubMed]
  275. Putoczki, T.; Ernst, M. More than a sidekick: The IL-6 family cytokine IL-11 links inflammation to cancer. J. Leukoc. Biol. 2010, 88, 1109–1117. [Google Scholar] [CrossRef]
  276. Klein, W.; Tromm, A.; Griga, T.; Fricke, H.; Folwaczny, C.; Hocke, M.; Eitner, K.; Marx, M.; Duerig, N.; Epplen, J.T. A polymorphism in the IL11 gene is associated with ulcerative colitis. Genes Immun. 2002, 3, 494–496. [Google Scholar] [CrossRef]
  277. Sabzevary-Ghahfarokhi, M.; Shohan, M.; Shirzad, H.; Rahimian, G.; Bagheri, N.; Soltani, A.; Deris, F.; Ghatreh-Samani, M.; Razmara, E. The expression analysis of Fra-1 gene and IL-11 protein in Iranian patients with ulcerative colitis. BMC Immunol. 2018, 19, 17. [Google Scholar] [CrossRef]
  278. Nishina, T.; Deguchi, Y.; Ohshima, D.; Takeda, W.; Ohtsuka, M.; Shichino, S.; Ueha, S.; Yamazaki, S.; Kawauchi, M.; Nakamura, E.; et al. Interleukin-11 is a Marker for Both Cancer-and Inflammation-Associated Fibroblasts that Contribute to Colorectal Cancer Progression. bioRxiv 2020. [Google Scholar] [CrossRef]
  279. Ault, P.; Kantarjian, H.; Welch, M.A.; Giles, F.; Rios, M.B.; Cortes, J. Interleukin 11 may improve thrombocytopenia associated with imatinib mesylate therapy in chronic myelogenous leukemia. Leuk. Res. 2004, 28, 613–618. [Google Scholar] [CrossRef]
  280. Espinosa-Cotton, M.; Rodman Iii, S.N.; Ross, K.A.; Jensen, I.J.; Sangodeyi-Miller, K.; McLaren, A.J.; Dahl, R.A.; Gibson-Corley, K.N.; Koch, A.T.; Fu, Y.X.; et al. Interleukin-1 alpha increases anti-tumor efficacy of cetuximab in head and neck squamous cell carcinoma. J. Immunother. Cancer 2019, 7, 79. [Google Scholar] [CrossRef]
  281. Malik, A.; Sharma, D.; Zhu, Q.; Karki, R.; Guy, C.S.; Vogel, P.; Kanneganti, T.D. IL-33 regulates the IgA-microbiota axis to restrain IL-1α-dependent colitis and tumorigenesis. J. Clin. Investig. 2016, 126, 4469–4481. [Google Scholar] [CrossRef] [PubMed]
  282. Maywald, R.L.; Doerner, S.K.; Pastorelli, L.; De, S.C.; Benton, S.M.; Dawson, E.P.; Lanza, D.G.; Berger, N.A.; Markowitz, S.D.; Lenz, H.J.; et al. IL-33 activates tumor stroma to promote intestinal polyposis. Proc. Natl. Acad. Sci. USA 2015, 112, E2487–E2496. [Google Scholar] [CrossRef] [PubMed]
  283. Schiering, C.; Krausgruber, T.; Chomka, A.; Fröhlich, A.; Adelmann, K.; Wohlfert, E.A.; Pott, J.; Griseri, T.; Bollrath, J.; Hegazy, A.N.; et al. The alarmin IL-33 promotes regulatory T-cell function in the intestine. Nature 2014, 513, 564–568. [Google Scholar] [CrossRef] [PubMed]
  284. Taniguchi, S.; Elhance, A.; Van Duzer, A.; Kumar, S.; Leitenberger, J.J.; Oshimori, N. Tumor-initiating cells establish an IL-33-TGF-β niche signaling loop to promote cancer progression. Science 2020, 369, eaay1813. [Google Scholar] [CrossRef]
  285. Herzyk, D.J.; Bugelski, P.J.; Hart, T.K.; Wier, P.J. Preclinical safety of recombinant human interleukin-18. Toxicol. Pathol. 2003, 31, 554–561. [Google Scholar] [CrossRef]
  286. Zhu, B.; Luo, J.; Jiang, Y.; Yu, L.; Liu, M.; Fu, J. Prognostic significance of nomograms integrating IL-37 expression, neutrophil level, and MMR status in patients with colorectal cancer. Cancer Med. 2018, 7, 3682–3694. [Google Scholar] [CrossRef]
  287. Weinstein, A.M.; Giraldo, N.A.; Petitprez, F.; Julie, C.; Lacroix, L.; Peschaud, F.; Emile, J.F.; Marisa, L.; Fridman, W.H.; Storkus, W.J.; et al. Association of IL-36γ with tertiary lymphoid structures and inflammatory immune infiltrates in human colorectal cancer. Cancer Immunol Immunother. 2019, 68, 109–120. [Google Scholar] [CrossRef]
  288. Kinoshita, F.; Tagawa, T.; Akamine, T.; Takada, K.; Yamada, Y.; Oku, Y.; Kosai, K.; Ono, Y.; Tanaka, K.; Wakasu, S.; et al. Interleukin-38 promotes tumor growth through regulation of CD8+ tumor-infiltrating lymphocytes in lung cancer tumor microenvironment. Cancer Immunol. Immunother. 2021, 70, 123–135. [Google Scholar] [CrossRef]
  289. van de Veerdonk, F.L.; de Graaf, D.M.; Joosten, L.A.; Dinarello, C.A. Biology of IL-38 and its role in disease. Immunol. Rev. 2018, 281, 191–196. [Google Scholar] [CrossRef]
  290. Damoiseaux, J. The IL-2–IL-2 receptor pathway in health and disease: The role of the soluble IL-2 receptor. Clin. Immunol. 2020, 218, 108515. [Google Scholar] [CrossRef]
  291. Suzuki, A.; Leland, P.; Joshi, B.H.; Puri, R.K. Targeting of IL-4 and IL-13 receptors for cancer therapy. Cytokine 2015, 75, 79–88. [Google Scholar] [CrossRef] [PubMed]
  292. Krzystek-Korpacka, M.; Zawadzki, M.; Neubauer, K.; Bednarz-Misa, I.; Górska, S.; Wiśniewski, J.; Witkiewicz, W.; Gamian, A. Elevated systemic interleukin-7 in patients with colorectal cancer and individuals at high risk of cancer: Association with lymph node involvement and tumor location in the right colon. Cancer Immunol. Immunother. 2017, 66, 171–179. [Google Scholar] [CrossRef] [PubMed]
  293. Lin, J.; Zhu, Z.; Xiao, H.; Wakefield, M.R.; Ding, V.A.; Bai, Q.; Fang, Y. The role of IL-7 in Immunity and Cancer. Anticancer Res. 2017, 37, 963–967. [Google Scholar] [PubMed]
  294. Angkasekwinai, P.; Chen, D. IL-9-producing T cells: Potential players in allergy and cancer. Nat. Rev. Immunol. 2021, 21, 37–48. [Google Scholar] [CrossRef] [PubMed]
  295. Liu, D.; Song, L.; Wei, J.; Courtney, A.N.; Gao, X.; Marinova, E.; Guo, L.; Heczey, A.; Asgharzadeh, S.; Kim, E.; et al. IL-15 protects NKT cells from inhibition by tumor-associated macrophages and enhances antimetastatic activity. J. Clin. Investig. 2012, 122, 2221–2233. [Google Scholar] [CrossRef] [PubMed]
  296. Waldmann, T.A. Cytokines in cancer immunotherapy. Cold Spring Harb. Perspect. Biol. 2018, 10, a028472. [Google Scholar] [CrossRef] [PubMed]
  297. Dolgin, E. First CD123-targeted drug approved after wowing in rare cancer. Nat. Biotechnol. 2019, 37, 202–204. [Google Scholar] [CrossRef]
  298. Testa, U.; Elvira, P.; Germana, C. CD123 as a therapeutic target in the treatment of hematological malignancies. Cancers 2019, 11, 1358. [Google Scholar] [CrossRef]
  299. Grisaru-Tal, S.; Itan, M.; Klion, A.D.; Munitz, A. A new dawn for eosinophils in the tumour microenvironment. Nat. Rev. Cancer 2020, 20, 594–607. [Google Scholar] [CrossRef]
  300. Hirano, T. IL-6 in inflammation, autoimmunity and cancer. Int. Immunol. 2021, 33, 127–148. [Google Scholar] [CrossRef]
  301. Cui, G.; Yuan, A.; Sun, Z.; Zheng, W.; Pang, Z. IL-1β/IL-6 network in the tumor microenvironment of human colorectal cancer. Pathol. Res. Pract. 2018, 214, 986–992. [Google Scholar] [CrossRef] [PubMed]
  302. Lin, Y.; He, Z.; Ye, J.; Liu, Z.; She, X.; Gao, X.; Liang, R. Progress in Understanding the IL-6/STAT3 Pathway in Colorectal Cancer. OncoTargets Ther. 2020, 13, 13023–13032. [Google Scholar] [CrossRef] [PubMed]
  303. Xu, J.; Ye, Y.; Zhang, H.; Szmitkowski, M.; Mäkinen, M.J.; Li, P.; Xia, D.; Yang, J.; Wu, Y.; Wu, H. Diagnostic and Prognostic Value of Serum Interleukin-6 in Colorectal Cancer. Medicine 2016, 95, e2502. [Google Scholar] [CrossRef]
  304. Grivennikov, S.I. IL-11: A prominent pro-tumorigenic member of the IL-6 family. Cancer Cell 2013, 24, 145–147. [Google Scholar] [CrossRef]
  305. Naing, A.; Infante, J.R.; Papadopoulos, K.P.; Chan, I.H.; Shen, C.; Ratti, N.P.; Rojo, B.; Autio, K.A.; Wong, D.J.; Patel, M.R.; et al. PEGylated IL-10 (Pegilodecakin) induces systemic immune activation, CD8+ T cell invigoration and polyclonal T cell expansion in cancer patients. Cancer Cell 2018, 34, 775–791. [Google Scholar] [CrossRef]
  306. Ouyang, W.; O’Garra, A. IL-10 family cytokines IL-10 and IL-22: From basic science to clinical translation. Immunity 2019, 50, 871–891. [Google Scholar] [CrossRef]
  307. Dumoutier, L.; Lejeune, D.; Colau, D.; Renauld, J.C. Cloning and characterization of IL-22 binding protein.; a natural antagonist of IL-10-related T cell-derived inducible factor/IL-22. J. Immunol. 2001, 166, 7090–7095. [Google Scholar] [CrossRef] [PubMed]
  308. Markota, A.; Stefan, E.; Sebastian, K. Targeting interleukin-22 for cancer therapy. Hum. Vaccines Immunother. 2018, 14, 2012–2015. [Google Scholar] [CrossRef]
  309. Hu, H.J.; Liang, X.; Li, H.L.; Du, C.M.; Hao, J.L.; Wang, H.Y.; Gu, J.F.; Ni, A.M.; Sun, L.Y.; Xiao, J.; et al. The armed oncolytic adenovirus ZD55-IL-24 eradicates melanoma by turning the tumor cells from the self-state into the nonself-state besides direct killing. Cell Death Dis. 2020, 11, 1022. [Google Scholar] [CrossRef]
  310. Menezes, M.E.; Bhatia, S.; Bhoopathi, P.; Das, S.K.; Emdad, L.; Dasgupta, S.; Dent, P.; Wang, X.Y.; Sarkar, D.; Fisher, P.B. MDA-7/IL-24: Multifunctional cancer killing cytokine. Anticancer Genes 2014, 818, 127–153. [Google Scholar]
  311. Larochette, V.; Miot, C.; Poli, C.; Beaumont, E.; Roingeard, P.; Fickenscher, H.; Jeannin, P.; Delneste, Y. IL-26, a cytokine with roles in extracellular DNA-induced inflammation and microbial defense. Front. Immunol. 2019, 10, 204. [Google Scholar] [CrossRef] [PubMed]
  312. Trotter, T.N.; Shuptrine, C.W.; Tsao, L.C.; Marek, R.D.; Acharya, C.; Wei, J.P.; Yang, X.Y.; Lei, G.; Wang, T.; Lyerly, H.K.; et al. IL26, a noncanonical mediator of DNA inflammatory stimulation, promotes TNBC engraftment and progression in association with neutrophils. Cancer Res. 2020, 80, 3088–3100. [Google Scholar] [CrossRef] [PubMed]
  313. Yan, J.; Smyth, M.J.; Teng, M.W.L. Interleukin (IL)-12 and IL-23 and their conflicting roles in cancer. Cold Spring Harb. Perspect. Biol. 2018, 10, a028530. [Google Scholar] [CrossRef]
  314. Hewitt, S.L.; Bai, A.; Bailey, D.; Ichikawa, K.; Zielinski, J.; Karp, R.; Apte, A.; Arnold, K.; Zacharek, S.J.; Iliou, M.S.; et al. Durable anticancer immunity from intratumoral administration of IL-23, IL-36γ, and OX40L mRNAs. Sci. Transl. Med. 2019, 11, eaat9143. [Google Scholar] [CrossRef] [PubMed]
  315. Elessawi, D.F.; Alkady, M.M.; Ibrahim, I.M. Diagnostic and prognostic value of serum IL-23 in colorectal cancer. Arab J. Gastroenterol. 2019, 20, 65–68. [Google Scholar] [CrossRef] [PubMed]
  316. Chihara, N. Dysregulated T cells in multiple sclerosis. Clin. Exp. Neuroimmunol. 2018, 9, 20–29. [Google Scholar] [CrossRef]
  317. Figueiredo, M.L.; Figueiredo Neto, M.; Salameh, J.W.; Decker, R.E.; Letteri, R.; Chan-Seng, D.; Emrick, T. Ligand-mediated targeting of cytokine Interleukin-27 enhances its bioactivity in vivo. Mol. Ther. Methods Clin. Dev. 2020, 17, 739–751. [Google Scholar] [CrossRef] [PubMed]
  318. Kourko, O.; Seaver, K.; Odoardi, N.; Basta, S.; Gee, K. IL-27, IL-30, and IL-35: A cytokine triumvirate in cancer. Front. Oncol. 2019, 9, 969. [Google Scholar] [CrossRef]
  319. Park, Y.J.; Ryu, H.; Choi, G.; Kim, B.S.; Hwang, E.S.; Kim, H.S.; Chung, Y. IL-27 confers a protumorigenic activity of regulatory T cells via CD39. Proc. Natl. Acad. Sci. USA 2019, 116, 3106–3111. [Google Scholar] [CrossRef]
  320. Li, X.; Shao, Y.; Sha, X.; Fang, P.; Kuo, Y.M.; Andrews, A.J.; Li, Y.; Yang, W.Y.; Maddaloni, M.; Pascual, D.W.; et al. IL-35 (interleukin-35) suppresses endothelial cell activation by inhibiting mitochondrial reactive oxygen species-mediated site-specific acetylation of H3K14 (histone 3 lysine 14). Arterioscler. Thromb. Vasc. Biol. 2018, 38, 599–609. [Google Scholar] [CrossRef]
  321. Sawant, D.V.; Yano, H.; Chikina, M.; Zhang, Q.; Liao, M.; Liu, C.; Callahan, D.J.; Sun, Z.; Sun, T.; Tabib, T.; et al. Adaptive plasticity of IL-10+ and IL-35+ Treg cells cooperatively promotes tumor T cell exhaustion. Nat. Immunol. 2019, 20, 724–735. [Google Scholar] [CrossRef] [PubMed]
  322. Vitiello, G.A.; George, M. Targeting the interleukin-17 immune axis for cancer immunotherapy. J. Exp. Med. 2020, 217, e20190456. [Google Scholar] [CrossRef] [PubMed]
  323. Bie, Q.; Jin, C.; Zhang, B.; Dong, H. IL-17B: A new area of study in the IL-17 family. Mol. Immunol. 2017, 90, 50–56. [Google Scholar] [CrossRef] [PubMed]
  324. Jungnickel, C.; Schmidt, L.H.; Bittigkoffer, L.; Wolf, L.; Wolf, A.; Ritzmann, F.; Kamyschnikow, A.; Herr, C.; Menger, M.D.; Spieker, T.; et al. IL-17C mediates the recruitment of tumor-associated neutrophils and lung tumor growth. Oncogene 2017, 36, 4182–4190. [Google Scholar] [CrossRef]
  325. Liu, R.; Lauridsen, H.M.; Amezquita, R.A.; Pierce, R.W.; Jane-Wit, D.; Fang, C.; Pellowe, A.S.; Kirkiles-Smith, N.C.; Gonzalez, A.L.; Pober, J.S. IL-17 promotes neutrophil-mediated immunity by activating microvascular pericytes and not endothelium. J. Immunol. 2016, 197, 2400–2408. [Google Scholar] [CrossRef] [PubMed]
  326. Tong, Z.; Yang, X.O.; Yan, H.; Liu, W.; Niu, X.; Shi, Y.; Fang, W.; Xiong, B.; Wan, Y.; Dong, C. A protective role by interleukin-17F in colon tumorigenesis. PLoS ONE 2012, 7, e34959. [Google Scholar] [CrossRef] [PubMed]
  327. Lazear, H.M.; Schoggins, J.W.; Diamond, M.S. Shared and distinct functions of type I and type III interferons. Immunity 2019, 50, 907–923. [Google Scholar] [CrossRef]
  328. Bakouny, Z.; Toni, K.C. IL-8 and cancer prognosis on immunotherapy. Nat. Med. 2020, 26, 650–651. [Google Scholar] [CrossRef]
  329. Ford, R.; Tamayo, A.; Martin, B.; Niu, K.; Claypool, K.; Cabanillas, F.; Ambrus, J., Jr. Identification of B-cell growth factors (interleukin-14; high molecular weight-B-cell growth factors) in effusion fluids from patients with aggressive B-cell lymphomas. Blood 1995, 86, 283–293. [Google Scholar] [CrossRef]
  330. Richmond, J.; Tuzova, M.; Cruikshank, W.; Center, D. Regulation of cellular processes by interleukin-16 in homeostasis and cancer. J. Cell. Physiol. 2014, 229, 139–147. [Google Scholar] [CrossRef]
  331. Sloot, Y.J.E.; Smit, J.W.; Joosten, L.A.B.; Netea-Maier, R.T. Insights into the role of IL-32 in cancer. Semin. Immunol. 2018, 38, 24–32. [Google Scholar] [CrossRef] [PubMed]
  332. Baghdadi, M.; Endo, H.; Takano, A.; Ishikawa, K.; Kameda, Y.; Wada, H.; Miyagi, Y.; Yokose, T.; Ito, H.; Nakayama, H.; et al. High co-expression of IL-34 and M-CSF correlates with tumor progression and poor survival in lung cancers. Sci. Rep. 2018, 8, 418. [Google Scholar] [CrossRef] [PubMed]
  333. Franzè, E.; Laudisi, F.; Di Grazia, A.; Marônek, M.; Bellato, V.; Sica, G.; Monteleone, G. Macrophages produce and functionally respond to interleukin-34 in colon cancer. Cell Death Discov. 2020, 6, 117. [Google Scholar] [CrossRef] [PubMed]
  334. Mármol, I.; Sánchez-de-Diego, C.; Pradilla Dieste, A.; Cerrada, E.; Rodriguez Yoldi, M.J. Colorectal Carcinoma: A General Overview and Future Perspectives in Colorectal Cancer. Int. J. Mol. Sci. 2017, 18, 197. [Google Scholar] [CrossRef]
  335. Bardhan, K.; Liu, K. Epigenetics and colorectal cancer pathogenesis. Cancers 2013, 5, 676–713. [Google Scholar] [CrossRef]
  336. Waldner, M.; Schimanski, C.C.; Neurath, M.F. Colon cancer and the immune system: The role of tumor invading T cells. World J. Gastroenterol. 2006, 12, 7233–7238. [Google Scholar] [CrossRef]
  337. Klintrup, K.; Mäkinen, J.M.; Kauppila, S.; Väre, P.O.; Melkko, J.; Tuominen, H.; Tuppurainen, K.; Mäkelä, J.; Karttunen, T.J.; Mäkinen, M.J. Inflammation and prognosis in colorectal cancer. Eur. J. Cancer 2005, 41, 2645–2654. [Google Scholar] [CrossRef]
  338. Levin, R.; Brown, M.J.; Kashtock, M.E.; Jacobs, D.E.; Whelan, E.A.; Rodman, J.; Schock, M.R.; Padilla, A.; Sinks, T. Lead exposures in US children, 2008: Implications for prevention. Environ. Health Perspect. 2008, 116, 1285–1293. [Google Scholar] [CrossRef]
  339. Zhao, H.; Wu, L.; Yan, G.; Chen, Y.; Zhou, M.; Wu, Y.; Li, Y. Inflammation and tumor progression: Signaling pathways and targeted intervention. Signal Transduct. Target. Ther. 2021, 6, 263. [Google Scholar] [CrossRef]
  340. Saber, S.; Alomar, S.Y.; Yahya, G. Blocking prostanoid receptors switches on multiple immune responses and cascades of inflammatory signaling against larval stages in snail fever. Environ. Sci. Pollut. Res. 2022, 29, 43546–43555. [Google Scholar] [CrossRef]
  341. Youssef, M.E.; El-Azab, M.F.; Abdel-Dayem, M.A.; Yahya, G.; Alanazi, I.S.; Saber, S. Electrocardiographic and histopathological characterizations of diabetic cardiomyopathy in rats. Env. Sci. Pollut. Res. Int. 2022, 29, 25723–25732. [Google Scholar] [CrossRef]
  342. Saber, S.; Yahya, G.; Gobba, N.A.; Sharaf, H.; Alshaman, R.; Alattar, A.; Amin, N.A.; El-Shedody, R.; Aboutouk, F.H.; Abd El-Galeel, Y.; et al. The Supportive Role of NSC328382, a P2X7R Antagonist, in Enhancing the Inhibitory Effect of CRID3 on NLRP3 Inflammasome Activation in Rats with Dextran Sodium Sulfate-Induced Colitis. J. Inflamm. Res. 2021, 14, 3443–3463. [Google Scholar] [CrossRef] [PubMed]
  343. Saber, S.; Abd El-Fattah, E.E.; Yahya, G.; Gobba, N.A.; Maghmomeh, A.O.; Khodir, A.E.; Mourad, A.A.E.; Saad, A.S.; Mohammed, H.G.; Nouh, N.A.; et al. A Novel Combination Therapy Using Rosuvastatin and Lactobacillus Combats Dextran Sodium Sulfate-Induced Colitis in High-Fat Diet-Fed Rats by Targeting the TXNIP/NLRP3 Interaction and Influencing Gut Microbiome Composition. Pharmaceuticals 2021, 14, 341. [Google Scholar] [CrossRef] [PubMed]
  344. Youssef, M.E.; Abdel-Reheim, M.A.; Morsy, M.A.; El-Daly, M.; Atwa, G.M.K.; Yahya, G.; Cavalu, S.; Saber, S.; Ahmed Gaafar, A.G. Ameliorative Effect of Dabigatran on CFA-Induced Rheumatoid Arthritis via Modulating Kallikrein-Kinin System in Rats. Int. J. Mol. Sci. 2022, 23, 10297. [Google Scholar] [CrossRef]
  345. Zohny, M.H.; Cavalu, S.; Youssef, M.E.; Kaddah, M.M.Y.; Mourad, A.A.E.; Gaafar, A.G.A.; El-Ahwany, E.; Amin, N.A.; Arakeep, H.M.; Shata, A.; et al. Coomassie brilliant blue G-250 dye attenuates bleomycin-induced lung fibrosis by regulating the NF-κB and NLRP3 crosstalk: A novel approach for filling an unmet medical need. Biomed. Pharmacother. 2022, 148, 112723. [Google Scholar] [CrossRef] [PubMed]
  346. Abd El-Fattah, E.E.; Saber, S.; Mourad, A.A.E.; El-Ahwany, E.; Amin, N.A.; Cavalu, S.; Yahya, G.; Saad, A.S.; Alsharidah, M.; Shata, A.; et al. The dynamic interplay between AMPK/NFκB signaling and NLRP3 is a new therapeutic target in inflammation: Emerging role of dapagliflozin in overcoming lipopolysaccharide-mediated lung injury. Biomed. Pharmacother. 2022, 147, 112628. [Google Scholar] [CrossRef] [PubMed]
  347. Dranoff, G. Cytokines in cancer pathogenesis and cancer therapy. Nat. Rev. Cancer 2004, 4, 11–22. [Google Scholar] [CrossRef]
  348. Balkwill, F.; Mantovani, A. Inflammation and cancer: Back to Virchow? Lancet 2001, 357, 539–545. [Google Scholar] [CrossRef] [PubMed]
  349. Ogunwobi, O.O.; Mahmood, F.; Akingboye, A. Biomarkers in Colorectal Cancer: Current Research and Future Prospects. Int. J. Mol. Sci. 2020, 21, 5311. [Google Scholar] [CrossRef]
  350. Louie, A.D.; Huntington, K.; Carlsen, L.; Zhou, L.; El-Deiry, W.S. Integrating Molecular Biomarker Inputs Into Development and Use of Clinical Cancer Therapeutics. Front. Pharmacol. 2021, 12, 747194. [Google Scholar] [CrossRef]
  351. Huang, Z.; Yang, M. Molecular Network of Colorectal Cancer and Current Therapeutic Options. Front. Oncol. 2022, 12, 852927. [Google Scholar] [CrossRef]
  352. Czajka-Francuz, P.; Francuz, T.; Cisoń-Jurek, S.; Czajka, A.; Fajkis, M.; Szymczak, B.; Kozaczka, M.; Malinowski, K.P.; Zasada, W.; Wojnar, J.; et al. Serum cytokine profile as a potential prognostic tool in colorectal cancer patients-one center study. Rep. Pract. Oncol. Radiother. 2020, 25, 867–875. [Google Scholar] [CrossRef] [PubMed]
  353. West, N.R.; McCuaig, S.; Franchini, F.; Powrie, F. Emerging cytokine networks in colorectal cancer. Nat. Rev. Immunol. 2015, 15, 615–629. [Google Scholar] [CrossRef] [PubMed]
  354. Wei, X.; Zhang, Y.; Yang, Z.; Sha, Y.; Pan, Y.; Chen, Y.; Cai, L. Analysis of the role of the interleukins in colon cancer. Biol. Res. 2020, 53, 20. [Google Scholar] [CrossRef] [PubMed]
  355. Lindebjerg, J.; Merete, O.; Claus, B. Colorectal cancers detected through screening are associated with lower stages and improved survival. Dan. Med. J. 2014, 61, A4758. [Google Scholar] [PubMed]
  356. Vogelaar, L.I.; van Ballegooijen, M.; Zauber, A.G.; Habbema, J.D.; Kuipers, E.J. Effect of rising chemotherapy costs on the cost savings of colorectal cancer screening. J. Natl. Cancer Inst. 2009, 101, 1412–1422. [Google Scholar] [CrossRef]
  357. Wu, Z.; Li, Y.; Zhang, Y.; Hu, H.; Wu, T.; Liu, S.; Chen, W.; Xie, S.; Lu, Z. Colorectal Cancer Screening Methods and Molecular Markers for Early Detection. Technol. Cancer Res. Treat. 2020, 19, 1533033820980426. [Google Scholar] [CrossRef]
  358. Wu, N.; Yang, X.; Zhang, R.; Li, J.; Xiao, X.; Hu, Y.; Chen, Y.; Yang, F.; Lu, N.; Wang, Z.; et al. Dysbiosis signature of fecal microbiota in colorectal cancer patients. Microb. Ecol. 2013, 66, 462–470. [Google Scholar] [CrossRef]
  359. Rahman, M.M.; Islam, M.R.; Shohag, S.; Ahasan, M.T.; Sarkar, N.; Khan, H.; Hasan, A.M.; Cavalu, S.; Rauf, A. Microbiome in cancer: Role in carcinogenesis and impact in therapeutic strategies. Biomed. Pharmacother. 2022, 149, 112898. [Google Scholar] [CrossRef]
  360. Cavalu, S.; Popa, A.; Bratu, I.; Borodi, G.; Maghiar, A. New Evidences of Key Factors Involved in Silent Stones Etiopathogenesis and Trace Elements: Microscopic, Spectroscopic, and Biochemical Approach. Biol. Trace Elem. Res. 2015, 168, 311–320. [Google Scholar] [CrossRef]
  361. Kappen, J.; Skorupa, M.; Krukiewicz, K. Conducting Polymers as Versatile Tools for the Electrochemical Detection of Cancer Biomarkers. Biosensors 2022, 13, 31. [Google Scholar] [CrossRef] [PubMed]
  362. Burney, I.A.; Lakhtakia, R. Precision medicine: Where have we reached and where are we headed? Sultan Qaboos Univ. Med. J. 2017, 17, e255–e258. [Google Scholar] [CrossRef] [PubMed]
  363. Maciejko, L.; Smalley, M.; Goldman, A. Cancer immunotherapy and personalized medicine: Emerging technologies and biomarker-based approaches. J. Mol. Biomark. Diagn. 2017, 8, 350. [Google Scholar] [CrossRef] [PubMed]
  364. Verma, M. Personalized medicine and cancer. J. Pers. Med. 2012, 2, 1–14. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Molecular alterations and Genetic changes cause various pathways: chromosomal instability (CIN), microsatellite instability (MSI), and CpG island methylator phenotype (CIMP,) which lead to metastasis, angiogenesis, drug resistance, immunosuppression, and inflammation in tumors.
Figure 1. Molecular alterations and Genetic changes cause various pathways: chromosomal instability (CIN), microsatellite instability (MSI), and CpG island methylator phenotype (CIMP,) which lead to metastasis, angiogenesis, drug resistance, immunosuppression, and inflammation in tumors.
Jcm 12 03127 g001
Figure 2. Immune cells influencing the tumor, leading to acute and chronic inflammation. Many cytokines (shown in form of colored dots in the figure) play their roles in this regard to either suppress or progress the tumor.
Figure 2. Immune cells influencing the tumor, leading to acute and chronic inflammation. Many cytokines (shown in form of colored dots in the figure) play their roles in this regard to either suppress or progress the tumor.
Jcm 12 03127 g002
Figure 3. Cytokine networks in the pathogenesis of colorectal cancer. Cytokines expressed by tumor and/or stromal cells cluster to form networks with antitumor, pro-tumor, or bivalent properties. IFN-γ, interleukin-12 (IL-12), IL-15, IL-17F, and IL-18 inhibit CRC development. IL-4, IL-6, IL-8, IL-11, IL-17A, IL-22, IL-23, IL-33, TNF, TGF-β, and VEGF are pro-tumorigenic. The contribution of IL-1, IL-9 IL-10, IL-21, and GM-CSF to intestinal cancer remains unclear.
Figure 3. Cytokine networks in the pathogenesis of colorectal cancer. Cytokines expressed by tumor and/or stromal cells cluster to form networks with antitumor, pro-tumor, or bivalent properties. IFN-γ, interleukin-12 (IL-12), IL-15, IL-17F, and IL-18 inhibit CRC development. IL-4, IL-6, IL-8, IL-11, IL-17A, IL-22, IL-23, IL-33, TNF, TGF-β, and VEGF are pro-tumorigenic. The contribution of IL-1, IL-9 IL-10, IL-21, and GM-CSF to intestinal cancer remains unclear.
Jcm 12 03127 g003
Figure 4. Schematic display of Tocilizumab that inhibits the activation of the Jak1-STAT3 signaling pathway in gastric/colorectal cancer cells. The Jak1-STAT3 pathway is activated by IL-6 (shown by colored dots in the figure).
Figure 4. Schematic display of Tocilizumab that inhibits the activation of the Jak1-STAT3 signaling pathway in gastric/colorectal cancer cells. The Jak1-STAT3 pathway is activated by IL-6 (shown by colored dots in the figure).
Jcm 12 03127 g004
Table 1. Cancer cell types in TME and the specific soluble factors released by them. Soluble factors involved in the promotion of cancer stem ness pathways.
Table 1. Cancer cell types in TME and the specific soluble factors released by them. Soluble factors involved in the promotion of cancer stem ness pathways.
TME CellsSoluble FactorsTarget CellsSignaling PathwayBiological EffectsReference
CD4+IL-22CRCSTAT3/DOT1L
(Signal transducer and activator of transcription)/(Disruptor of telomeric silencing 1-like)
Stemness gene regulation[59]
CAF
(Cancer-associated fibroblasts)
HGF/SDF1
(Hepatocyte growth factor)/Stromal cell-derived factor-1
Cancer stem cells (CSC)Wnt/β-cateninClonogenic activity and expression of
CD44v6
[60]
Endothelial cellsJAG1
(Jagged)
CRCNotchCD133 expression, tumorigenicity and chemoresistance[61]
MSC
(Mesenchymal stem cells)
PGE2
(Prostaglandin E2)
CRCWnt/β-cateninEMT (Epithelial-to-mesenchymal transition) and invasion[62]
MyofibroblastsHGFCSCWnt/β-cateninClonogenicity[63]
Table 2. Major interleukins involved in CRC progression and studied as biomarkers.
Table 2. Major interleukins involved in CRC progression and studied as biomarkers.
CytokineFunctional Effect in CRCExpression PatternsReference
IL-1αPromotes metastasis and the chemosensitivityJcm 12 03127 i001[136,137]
IL-1βPromotes the proliferation of colon cancer cells, tumorigenesis, and alters the tumor microenvironmentJcm 12 03127 i002[137,138,139]
IL-18Antitumorigenic properties and release of other signalsJcm 12 03127 i003[140,141]
IL-2, IL-7, IL-9, IL-15Antitumor activity, promote EMT, proliferation, invasion, and metastasisIL-4, IL-7 upregulated, IL-9 downregulated, IL-2 in between[142,143,144,145,146,147]
IL-21Activation of immune response biomarkersJcm 12 03127 i004
(Potential for biomarker)
[148,149,150]
IL-6Promotes mitosis, proliferation, metastasis, migration, and angiogenesisJcm 12 03127 i004[151,152,153,154]
IL-11Facilitates the proliferation of CRCJcm 12 03127 i005[155,156,157]
IL-8Promotes cell proliferation, angiogenesis, cancer metastasis, chemoresistance, antianoikis, maintains CCSC propertiesJcm 12 03127 i006[158,159,160]
IL-10Pathogenesis and progressionJcm 12 03127 i007[141,161,162,163]
IL-22Dominant role in CRC tumorigenesis, antiapoptosis, and cell proliferationJcm 12 03127 i008[164,165,166]
IL-17aPromotes cell cycle progression and angiogenesisJcm 12 03127 i009[167]
IL-17bPromotes tumorJcm 12 03127 i010[168,169,170,171]
IL-4Overexpressed in early CRC, tumor developmentJcm 12 03127 i011[172]
IL-23Overexpressed in CRC tissue and predictive for CRC metastasisJcm 12 03127 i012[173,174,175,176]
Upwards arrow just showed upregulation of the genes while downwards show suppression or downregulation.
Table 3. Interleukin families in colorectal cancer.
Table 3. Interleukin families in colorectal cancer.
Interleukin FamilyReceptorsCytokinePotential Effect in CRCTherapeutic StrategyReference
IL-1R1
IL-1R2
IL-1αMetastasis promotion with chemosensitivity. Promotes antitumor immunity and carcinogenesis (inflammatory)Therapeutic neutralization in order to tackle severe illness in clinical trials[102,139,140,280]
IL-2R1
IL-1R2
ILR3
sIL-IR2
sIL-IR3
IL-1βProliferation of cancer cells of colon and promotion of tumorigenesis. Altering the tumor microenvironmentTherapeutic neutralization to manage cytokine release syndrome (CRS) in clotting time and the prevention of cancer[102,140,141]
IL-1 super familyIL-1R4
(ST2)
IL-33Protumor, maintenance of intestinal microbiota, tumor microenvironment change, TH2 polarization, Treg cell function, promotion of
Angiogenesis and enhancement of colon cancer cell stemness, maintain intestinal microbiota.
Preclinical neutralization[15,281,282,283,284]
IL-18 subfamilyIL-1R5–IL-1R7 IL-18BP
IL-1R5 (IL-18Rα IL-18Rβ)
IL-18Antitumor, activates lymphocytes to produce IFN-γ, improve the integrity of intestinal barrier and induces apoptosis to
act on NK cells
Preclinical engineered rIL-18 or by combined with activated clotting time, hindered by IL-18BP[140,285]
IL-1R8–IL-1R5IL-37Antitumor attributes, inhibit the colon cancer cell development by stopping β-catenin.Not explored[135,286]
IL-1R6IL-36αAntitumor [287]
IL-36 subfamilyIL-1R6IL-36γAntitumor, inflammatory immune infiltrates promotion, promote inflammation (TH1-type) inhibited by IL-36RaPreclinical rIL-36γ as an alternative to IL-1[287]
IL-38IL-1R6–IL-1R9ImmunosuppressiveNot explored[288,289]
IL-2Rα, IL-2Rβ/IL-2Rγ, IL-2Rα/IL-2Rβ/IL-2Rγ
sIL-2RαIL-2/IL-15Rβ–γc IL-2Rα–IL-2/IL-15Rβ–γc
IL-2Antitumor, NK and T cell growth factor, inhibit T cell responses by maintaining Treg cells and AICD inductionrIL-2 approved for monotherapy. Engineered to avoid side effects and to be used in ACT[147,148,290]
Type (IL-4Rα/γc) and Type (IL-4Rα/IL-13Rα1)IL-4Promote epithelial-to-mesenchymal transition (EMT), metastasis and invasion. Promotes inflammation of TH2-type and polarization of TH9. Promotes cancer cell growth upon overexpression of IL-4R.IL-4R- targeting to bear cancer cells and block signaling. Antitumor TH9 cells production for ACT[146,291]
IL-2 familyIL-7R (IL-7Rα/γc)IL-7Metastasis promotion
Antitumoural: NK growth factor and T cell production
rIL-7 in combination with interleukins or ACT[145,292,293]
IL-9R (IL-9Rα/γc)IL-9Antitumor action
Pleiotropic
Preclinical TH9 cells in ACT[15,109,294]
IL-15R (IL-15Rα/IL-15Rβ/γc)IL-15Proliferation and angiogenesis inhibition, Antitumor activity by activating lymphocytes to produce IFN γ,
Promote apoptosis
rIL-15 or analogues in combination with interleukins or ACT[295,296]
IL-21R–γc
heterodimers of IL-21R and γc
IL-21Enhances cytotoxicity of CTLs, Antitumor activityCombination therapies with rIL-21 in clinical trials[296]
IL-3 familyIL-3Rα–βcIL-3Promotes malignancy as a
haematopoietic factor
Target CD123-bearing cells by fused to toxins[297,298,299]
IL-6 familygp130 IL-6R
IL-6Rα–gp130 (classic) sIL-6Rα–gp130 (trans)
IL-6Mitosis promotion, metastasis, proliferation, making the microenvironment for metastasis, activates tumor outgrowth and carcinogenesis, mediates cytokine release syndrome (CRS), Cachexia promotionNeutralization to manage CRS in ACT, cachexia[300,301,302,303]
gp130 IL-11Ra
IL-11Rα–gp130 (classic) sIL-11Rα–gp130 (trans)
IL-11Proliferation of CRC
Promotes inflammation by inducing carcinogenesis and cancer progression
Preclinical neutralization and gp130 common receptor blockade[158,304]
IL-31Rα–OSMRβIL-31TH2-type cytokine, evidently tumorigenicUnexplored[287]
IL-10 familyIL-10RA and IL-10RBIL-10Promotes cytotoxicity, inhibits antitumor responsesrIL-10 to increase cytotoxicity in trials[163,165,305,306]
IL-10RB and IL-22R /IL-22BPIL-22Promote tumorigenesis, antiapoptosis and cell proliferation
Peritumoral: promotion of carcinoma progression
Preclinical neutralization[114,306,307,308]
IL-20Rα–IL-20RβIL-22Rα1–IL-20RβIL-24Induces autophagy of cancer and apoptosisPreclinical rIL-24 combined with oncolytic virus[309,310]
IL-20Rα–IL-10RβIL-26Pro-tumoral through TH17 cells and neutrophilsPreclinical neutralization[311,312]
IL-12Rβ1–IL-12Rβ2IL-12Antitumoral: the main driver of TH1-type immunity, amplification and initiation of productionEngineered or combined with other interleukins in trials[286,313]
IL-12 familyIL-23R–IL-12Rβ1IL-23Mainly pro-tumoral: direct and indirect effect via TH17 cells and TH22 cellsNeutralization in trials, enhances CAR T cell cytotoxicity[314,315]
IL-27RαIL-27 and IL-30Pleiotropic: induces cytotoxicity and NK cell yet enhances T cell and Treg cell activityNeutralization and engineered rIL-27 in trials[316,317,318,319]
IL-12Rβ2–gp130gp130–gp130IL-27Rα–IL-12Rβ2IL-35Treg cell-mediated suppression of T cell responses and promotion of metastatic colonizationPreclinical neutralization with checkpoint inhibitors and other therapies[320,321]
IL-17 familyIL-17RA–IL-17RCIL-17A/FCell cycle progression and angiogenesis, facilitate the development indirectly and change the tissue environment and microbiota of CRCNeutralization in clinical trials[169,322]
IL-17RIL-17RBIL-17bCarcinogenesis, immunosuppression, EMT [170,323]
IL-17RA–IL-17REIL-17cMostly pro-tumoral, but antitumoral propertiesNot explored[324]
UnknownIL-17 D [325]
IL-17RIL-17e [170]
IL-17RA–IL-17RBIL-25
IL-17RIL-17fTumor suppression effect possibly by inhibiting tumor angiogenesis [326]
IFN-γ familyIL-28A and IL-28B
IL-28Rα–IL-10Rβ
IL-28Rα
IL-10Rβ
IL-29
Antitumoral: induces apoptosis of malignant cellsPreclinical gene therapy using IL-28 and IL-29[327]
CXCR1, CXCR2ACKR1/DARCIL-8Attracts neutrophils and mediates the suppressive environmentTherapeutic neutralization in clinical trials[328]
IL-13Rα1–IL-4RαIL-13Rα2 TH2-type cytokineTargeting or blocking IL-13R[291]
IL-14α and IL-14βIL-14RGrowth factor in B cell and in lymphomaNot explored[329]
CD4IL-16Pro-tumoral: proliferation of lymphoma and chemoattractantScarce preclinical evidence[330]
Other interleukinsUnknownIL-32Pleiotropic in action but depends on cancer type and isoform.Preclinical antitumor effects in combination[331]
CSF1RIL-34Pro-tumoral: immune suppression, cancer progression, and resistance of therapyPreclinical neutralization to lessen the pro-tumor effects[332,333]
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

Maryam, S.; Krukiewicz, K.; Haq, I.U.; Khan, A.A.; Yahya, G.; Cavalu, S. Interleukins (Cytokines) as Biomarkers in Colorectal Cancer: Progression, Detection, and Monitoring. J. Clin. Med. 2023, 12, 3127. https://doi.org/10.3390/jcm12093127

AMA Style

Maryam S, Krukiewicz K, Haq IU, Khan AA, Yahya G, Cavalu S. Interleukins (Cytokines) as Biomarkers in Colorectal Cancer: Progression, Detection, and Monitoring. Journal of Clinical Medicine. 2023; 12(9):3127. https://doi.org/10.3390/jcm12093127

Chicago/Turabian Style

Maryam, Sajida, Katarzyna Krukiewicz, Ihtisham Ul Haq, Awal Ayaz Khan, Galal Yahya, and Simona Cavalu. 2023. "Interleukins (Cytokines) as Biomarkers in Colorectal Cancer: Progression, Detection, and Monitoring" Journal of Clinical Medicine 12, no. 9: 3127. https://doi.org/10.3390/jcm12093127

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