Review Open Access
Copyright ©The Author(s) 2022. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Jul 14, 2022; 28(26): 3027-3046
Published online Jul 14, 2022. doi: 10.3748/wjg.v28.i26.3027
Tumor microenvironment involvement in colorectal cancer progression via Wnt/β-catenin pathway: Providing understanding of the complex mechanisms of chemoresistance
María Belén Novoa Díaz, María Julia Martín, Claudia Gentili, Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS)-INBIOSUR (CONICET-UNS), Bahía Blanca 8000, Argentina
María Julia Martín, Departamento de Química, Universidad Nacional del Sur (UNS)-INQUISUR (CONICET-UNS), Bahía Blanca 8000, Argentina
ORCID number: María Belén Novoa Díaz (0000-0002-6684-4076); María Julia Martín (0000-0001-5046-7686); Claudia Gentili (0000-0002-9957-7824).
Author contributions: Novoa Díaz MB, Martín MJ, and Gentili C contributed to the conceptualization, methodology, investigation, formal analysis, and visualization, wrote the original draft, and edited the manuscript; Gentili C obtained the resources and oversaw the project administration and funding acquisition.
Supported by Agencia Nacional de Promoción Científica y Tecnológica, No. PICT-2013-1441; Consejo Nacional de Investigaciones Científicas y Técnicas, No. PIP11220150100350; Instituto Nacional del Cáncer Asistencia Financiera II, RESOL 493/14, No. 2002-4395-14-1; Instituto Nacional del Cáncer Asistencia Financiera III-2016-2017, RESOL-2016-1006-E-APN-MS, No. 2002-3862-16-1; and Universidad Nacional del Sur (PGI), Argentina, No. 24/B230 and No. 24/B303.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: María Julia Martín, PhD, Research Assistant, Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS)-INBIOSUR (CONICET-UNS), San Juan 670, Bahía Blanca 8000, Argentina. julia.martin@uns.edu.ar
Received: December 21, 2021
Peer-review started: December 21, 2021
First decision: March 10, 2022
Revised: April 29, 2022
Accepted: June 20, 2022
Article in press: June 20, 2022
Published online: July 14, 2022

Abstract

Colorectal cancer (CRC) continues to be one of the main causes of death from cancer because patients progress unfavorably due to resistance to current therapies. Dysregulation of the Wnt/β-catenin pathway plays a fundamental role in the genesis and progression of several types of cancer, including CRC. In many subtypes of CRC, hyperactivation of the β-catenin pathway is associated with mutations of the adenomatous polyposis coli gene. However, it can also be associated with other causes. In recent years, studies of the tumor microenvironment (TME) have demonstrated its importance in the development and progression of CRC. In this tumor nest, several cell types, structures, and biomolecules interact with neoplastic cells to pave the way for the spread of the disease. Cross-communications between tumor cells and the TME are then established primarily through paracrine factors, which trigger the activation of numerous signaling pathways. Crucial advances in the field of oncology have been made in the last decade. This Minireview aims to actualize what is known about the central role of the Wnt/β-catenin pathway in CRC chemoresistance and aggressiveness, focusing on cross-communication between CRC cells and the TME. Through this analysis, our main objective was to increase the understanding of this complex disease considering a more global context. Since many treatments for advanced CRC fail due to mechanisms involving chemoresistance, the data here exposed and analyzed are of great interest for the development of novel and effective therapies.

Key Words: Colorectal cancer, β-catenin pathway, Tumor stroma, Tumor microenvironment factors, Cancer progression, Drug resistance

Core Tip: Currently, there is a high probability of failure in treatments for the advanced stages of colorectal cancer (CRC). For this reason, it is necessary to obtain a better understanding of CRC biology in a more global context for the future design of novel therapeutic approaches. The effects of the Wnt/β-catenin signaling pathway and the tumor microenvironment (TME) on the progression and chemoresistance of this disease were separately described in several review articles. Therefore, herein we comprehensively analyze the complex mechanisms of CRC chemoresistance triggered by TME factors that impact Wnt/β-catenin signaling.



INTRODUCTION

Colorectal cancer (CRC) is the third most frequent malignant disease worldwide in both men and women[1,2]. Data provided by the World Cancer Research Fund International indicate that CRC incidence is growing, and the prevalence of this pathology is expected to increase by 60% in the next 15 years[3]. Among all cancer-induced deaths, the ones associated with CRC reach values of 8%-9%[4,5]. These statistics rely mainly on the fact that 20% of patients show metastasis (stage IV) at the time of diagnosis and the overall survival in these patients is low due to failure in the treatments[5,6]. To improve the response of these patients to therapy, it is necessary to expand the knowledge about the mechanisms that play a critical role in the development, progression and chemoresistance of CRC.

In 2015, Guinney et al[7] proposed a classification of colorectal tumors based on four consensus molecular subtypes (CMSs). Groups were defined according to certain parameters such as clinical, genetic and molecular characteristics (Figure 1): The CMS1 group, also designated microsatellite instability immune, is characterized by high microsatellite instability and immune response. The canonical subtype (CMS2) represents 37% of cases and presents hyperactivation of the Wnt and Myc signaling pathways. The CMS3 (metabolic subtype) is characterized by a marked dysregulation of metabolic pathways. Finally, the CMS4 (mesenchymal subtype) group exhibits hyperactivation of the transforming growth factor-β (TGF-β) pathway and a marked switch from the epithelial to mesenchymal transition (EMT)[7,8]. Remarkably, 90% of CRC cases present with aberrant activation of the canonical Wnt/β-catenin signaling pathway[9-11]. This pathway is strictly regulated in physiological conditions and modulates fetal development and homeostasis in adult tissues[11]. Briefly, in the absence of Wnt ligand, β-catenin is continuously phosphorylated in the cytoplasm by a destruction complex comprised of adenomatous polyposis coli (APC), axis inhibition protein (Axin), glycogen synthase kinase 3 (GSK3), and casein kinase 1. Once phosphorylated, β-catenin is ubiquitinated and degraded by the proteasome. When Wnt ligand binds to its receptor, the destruction of β-catenin stops, so this protein accumulates in the cytosol and then translocates to the nucleus. Finally, β-catenin binds to transcription factors from the T-cell factor/lymphoid enhancer factor (TCF/LEF) family and promotes the transcription of several genes[12]. There are several published research as well as review articles on the involvement of the Wnt/β-catenin pathway in CRC development, progression, and chemoresistance[10,13,14]. Furthermore, this pathway became an unquestionable target for novel therapies for CRC. Despite this, to date, there are no approved treatments or clinical trials for CRC based on targeting the Wnt/β-catenin pathway[15].

Figure 1
Figure 1 Consensus molecular subtypes classification for colorectal tumors. BRAF: B-Raf proto-oncogene; CIMP: CpG island methylator phenotype; CNAs: Copy number alterations; EMT: Epithelial to mesenchymal transition; KRAS: Kirsten rat sarcoma 2 viral oncogene homolog; MSI: Microsatellite instability; TGF-β: Transforming growth factor-β; TP53: Tumor protein 53.

As extensively described in the open literature, hyperactivation of the Wnt/β-catenin pathway in CRC is directly associated with intrinsic causes such as mutations in the APC gene. This results in the nuclear accumulation of β-catenin and the induction of Wnt target genes that promote tumor progression[16]. In this contribution, we intend to analyze how Wnt/β-catenin aberrant activation can be induced or exacerbated by external causes.

In the last decade, the tumor microenvironment (TME) or tumor stroma has become relevant in the progression of CRC[17]. The TME is a niche composed of the extracellular matrix (ECM), a great variety of modified stromal cells and aberrant vasculature[9]. Intercellular communications between the tumor cells and the TME are mainly established through paracrine signaling[9,18]. These communications and factors are extrinsic and may directly affect CRC progression through the Wnt/β-catenin pathway. In this way, a wide range of novel therapeutic targets appears, expanding the possibilities of achieving effective treatments for CRC.

This work analyzed the available literature regarding the biomolecules associated with the TME that can modulate the Wnt/β-catenin signaling pathway in CRC cells, which allows a better understanding of the biological causes of the high morbidity and mortality of this pathology in a global context. Although antecedents of this particular analysis are registered in 2008[19], the enormous amount of information collected in recent years results in the emergence of new biomolecules, structures, and even processes related to the TME and the CRC cells that were previously unknown or unrelated. The comprehension of this information enables countless new possibilities for the development of new therapies for CRC.

DYNAMICS ESTABLISHED BETWEEN THE TME AND CRC CELLS

During CRC genesis, the TME and its associated signaling play a key role in tumor fate, since they facilitate the proliferation, invasion, metastasis, and chemoresistance processes. Specifically, the TME is composed of ECM proteins, mesenchymal stem cells, fibroblasts, cancer-associated fibroblasts (CAFs), endothelial cells (ECs), and tumor-infiltrating immune cells, with the last group comprising tumor-associated macrophages (TAMs), tumor-associated neutrophils, natural killer (NK) cells, regulatory T cells, myeloid-derived suppressor cells, and cytotoxic T lymphocytes (also known as CD8+ T cells), among others[5,20].

The TME cells and neoplastic cells continuously secrete and internalize factors that modulate the CRC development and contribute to the maintenance of the tumor ecosystem. These extracellular signals are schematized in Figure 2 and involve soluble proteins, insoluble proteins from the ECM, or compounds of variable nature loaded in vesicles[21]. Table 1 compiles the biomolecules well described in the literature that can be secreted by the TME cells and that allow communication with CRC cells. Some of these factors can travel freely through the tumor stroma space, whereas others are transported through membrane-bound vesicles known as extracellular vesicles (EVs)[22,23]. The EVs structure includes a lipid bilayer composed mainly of ceramide, cholesterol, sphingomyelin, phosphoglycerides, glycosphingolipids, phosphatidyl serine, phosphatidylethanolamine, mannose, N-linked glycans, polylactosamine, and sialic acid[22]. In recent years, EVs have acquired relevance as biological mediators in the communication between the TME and tumor cells. Concerning EVs related to CRC, their loads are considered prognostic factors or indicative of response to therapies. Recent studies have shown that EVs can carry large amounts of biomolecules from the TME cells to cancer cells and vice versa. Usual cargoes include cytokines/chemokines, angiogenic factors, ECM remodeling factors, and nucleic acids such as microRNAs (miRs), long non-coding RNAs (lncRNAs), and circular RNAs (cRNAs)[24-27]. Fluids from CRC patients such as plasma, saliva, and urine contain large amounts of secreted EVs. These molecules can stimulate or inhibit the expression of oncogenes and oncoproteins, thus affecting the phenotype of the neoplastic cells or altering the secretory profile of the TME cells[22,25,28]. Unlike cytokines and growth factors that act in the nearby cells, the EVs can influence both the primary colorectal tumor and metastatic niche remotely[22,29]. For instance, analysis of the plasma from CRC patients revealed that EVs are directly linked to the establishment of liver proinflammatory phenotype and liver metastasis, with this effect mediated by the modulation of macrophages by EVs[30]. The correlation between CD8+ T cell activation and high EVs concentrations in plasma has also been demonstrated, thus showing the role of EVs in the modulation of the immune response in CRC[31]. In conclusion, EVs have become a new source of therapeutic targets for CRC[22].

Figure 2
Figure 2 Extracellular signal molecules secreted by the tumor microenvironment and neoplastic cells. The traffic of soluble and insoluble factors allows crosstalk between tumor cells and their environment modulating colorectal cancer development and progression.
Table 1 Factors secreted by the tumor microenvironment and their role in colorectal cancer progression.
Released/secreted components
Source, TME cell
Known effects in CRC
Ref.
Growth factors
TGF-βCAFs; TIICs1; MSCs1Proliferation on tumor and stromal cells in late stages of tumorigenesis. EMT program and CSC-like traits. Metastasis, vasculogenesis and angiogenesis[5,21,136]
BMPsCAFsAnti-tumor activity. Or pro-tumor activity, induce CSCs phenotype, EMT program and chemoresistance. Differentiation of colon CSCs[5,137,138]
HGFCAFs; TIICs; MSCs1Invasion, metastasis and stemness[21,54,139,140]
VEGFECs; CAFs; TIICsAngiogenesis, invasiveness, metastasis[104,140,141]
FGFCAFs; MSCsCAFs profiles. Tumor growth and metastasis[142,143]
PDGFCAFsTumor growth and metastasis[144]
TNF-αTIICsProliferation. Growth arrest and cancer cell death, angiogenesis and metastasis[141,145]
Cytokines
IL-1TIICsAngiogenesis and metastasis[141,146]
IL-2TIICsAnti-tumor activity[1,147]
IL-6TIICs; MSCs; CAFsProliferation, angiogenesis and metastasis[1,141]
IL-8TIICs; MSCsTumor growth, angiogenesis and chemoresistance[1,148]
IL-17CAFs; TIICsAnti-tumor or pro-tumor activity. Invasion and self-renewal of CSCs[1,104,149]
IL-18TIICsAnti-tumor activity[150]
IL-22TIICsProliferation, invasion and stemness[1,104]
IL-33ECs; TIICsAnti-tumor activity Suppresses tumorigenesis. Or pro-tumor activity. Angiogenesis and metastasis. Tumor growth through immunosuppressive microenvironment favoring[1,104,151]
CCL2TIICs; MCSTumor progression[152]
CCL5TIICsTumor progression. Acts on tumor cells and TAMs[153]
CCL7CAFsProliferation, invasion, and migration[154]
CXCL12CAFs; MSCsProliferation and invasion[142,155]
PTHrPUndefined TME cellsProliferation, invasion, angiogenesis, migration and chemoresistance[34,35,66,156,157]
OsteopontinTIICsMetastasis, stemness and chemoresistance[85]
Prostaglandins
PGs (like PGE2)CAFs; TIICs; MSCsResistance to apoptosis, increased proliferation, angiogenesis and metastasis[21,32]
Signaling pathways ligands
NOTCH ligands (Jagged-1; Jagged-2; DLL4)ECsCSCs phenotype, EMT program and metastasis[104,158]
WNT ligands (Wnt2, Wnt5)CAFsInvasion, metastasis and angiogenesis[44,45,54]
Enzymes
Serine proteinases (like MMPs)TAMs; TANsInvasion and angiogenesis[140,145]
Immunosuppressive enzymes (like iNOS)TIICsTumor progression. Inhibitory effect on the immune system, apoptosis of immune cells[39]
Receptors
TLRsCAFs, ECsInflammatory-mediated tumorigenesis[41,43]
RNA molecules
miR-92a-3pCAFsCSCs phenotype, EMT program and chemoresistance[24]
lncRNA H19CAFsStemness and chemoresistance[25]
miR-155MSCs1Migration[21,159]
miR-375MSCs1Chemoresistance[21,160]
cRNACAFsTumor progression or anti-tumor activity[26,27]

On the other hand, the soluble molecules that are directly secreted to the biological fluids exert their actions locally and participate in the communication between the tumor and the microenvironment. These factors are prostaglandins, cytokines, and other paracrine factors, which have distinctive modes of action. Prostaglandins and cytokines act especially as immunomodulators, by promoting or inhibiting the progression of CRC[1,32,33]. Growth factors such as TGF-β, vascular endothelial growth factor (VEGF), and EGF promote the transformation and development of neoplastic cells by inducing proliferation, invasion, and migration responses and also by affecting the TME[21]. In previous works in vitro using two CRC cell lines with different grades of aggressiveness, we demonstrated that parathyroid hormone-related peptide (PTHrP) acts as a paracrine factor by inducing the survival, proliferation, migration, cell cycle progression, angiogenesis, and EMT program. Moreover, our in vivo studies in CRC tumor xenografts revealed that the intratumor administration of PTHrP modulates the expression of several tumorigenic markers, which are involved in the same cell responses observed in vitro[5,34-37].

Other proteins with different functions were studied in relation to CRC and the TME. In recent years, it has been reported that both neoplastic cells and TME cells can synthesize and secrete enzymes that participate in tumorigenesis. Muñoz-Galván et al[38] demonstrated that phospholipase D2 (PLD2) enzyme is overexpressed and secreted by CRC cells. PLD2 in the extracellular space modulates the phenotype and secretory profile of CAFs, thus contributing to promote stemness in tumor cells[38]. Other enzymes such as nitric oxide synthase are capable of inducing immunomodulatory effects[39]. Moreover, enzymes present in the TME, such as metalloproteases, enable the invasion and migration of cancer cells[39]. These enzymes act mainly by altering the structure and function of the ECM, thus allowing the activation of other factors that amplify the pro-tumor signals[40]. Receptor proteins such as Toll-like receptors (TLRs) play a fundamental role in maintaining epithelial barrier homeostasis in the gut and mediating inflammatory and immune responses[41]. TLRs are also expressed in fibroblasts and ECs in the TME and in CRC cells. The dysregulation in TLR pathways is associated with inflammation-driven carcinogenesis[42]. In response to this evidence, currently, there are clinical trials that include TLRs agonists for the treatment of CRC (www.clinicaltrials.gov)[41,43].

The abovementioned factors generally exert their effects by modulating several signaling pathways in the tumor[1,44-46]. In accordance with the focus of this contribution, in the next sections, we further discuss which of these biomolecules participate in aberrant activation of the β-catenin signaling pathway in CRC, and how these findings have provided new opportunities for the development of more efficient therapies.

TME FACTORS AND CONDITIONS THAT MODULATE THE WNT/β-CATENIN PATHWAY IN CRC

It is clear that the signaling triggered by factors that mediate the communication between TME and CRC cells is of great importance. Since Wnt/β-catenin is involved in most of the processes related to CRC genesis and progression[8,47], in this section we focus on the factors from the TME able to modulate this pathway. Figure 3 schematizes the complex interrelationships between the TME and CRC cells that lead to the activation of β-catenin and the consequent effects on disease progression.

Figure 3
Figure 3 Influence of tumor microenvironment-derived factors in the activation of β-catenin pathways. Several cytokines, growth factors, and small nuclei acid molecules secreted by stromal cells induce aberrantly activation of β-catenin and its nuclear translocation in colorectal cancer (CRC) cells promoting events associated with an aggressive phenotype of the tumor cells. Il-37 represses the expression of β-catenin and its transcriptional activity. Osteopontin is statistically associated with the expression of β-catenin in CRC and is known to induce its activation in other types of cancer. More studies are necessary to confirm positive feedback between the secretion of this tumor microenvironment (TME) factor and β-catenin signaling pathways in CRC. In addition to these factors, the microbiota and the hypoxia in the TME, also participate in the modulation of β-catenin activation. This figure is original for this work. BMP: Bone morphogenic protein; CCAL: Colorectal cancer-associated lncRNA; cRNA: Circular RNA; CXCL: C-X-C motif chemokine ligand; HGF: Hepatocyte growth factor; IL: Interleukin; lncRNA: Long non-coding RNA; OPN: Osteopontin; PTHrP: Parathyroid Hormone-related Peptide; TNF-α: Tumor necrosis factor-α; Wnt: Wingless protein.

Other β-catenin regulation, independently of the destruction complex actions described in the introduction section, involves phosphorylation by other kinases in different amino acids, such as Ser-552 and Ser-675, resulting in the stabilization and nuclear accumulation of this protein[48]. Mutations in the gene that encodes the β-catenin protein (CTNNB1 gene) will produce structural alterations and its consequent hyperactivation and nuclear accumulation, events directly associated with CRC genesis and progression[7,49]. Voorneveld and collaborators[50] demonstrated that nuclear β-catenin is found predominantly in cells at the invasive front of CRC tumors. Other investigations have proven that the signals that exacerbate aberrant β-catenin function independently of others than those produced by the tumor can come from the TME[44,51].

Activation of the Wnt pathway by TME factors was initially studied a decade ago. Vermeulen and colleagues showed that CAF-derived hepatocyte growth factor (HGF) activates Wnt/β-catenin signaling and subsequently the clonogenicity in cancer stem cells (CSCs) isolated from CRC patients that were transiently transfected with TCF/LEF luciferase reporter vector. Moreover, HGF restored the CSC phenotype in more differentiated tumor cells both in vitro and in vivo[52]. Essex and collaborators recently replicated these studies, emphasizing the relevance of this pathway and the interaction of tumor cells with their stroma, and in the promotion and development of CRC[53]. As stated in Table 1, various Wnt ligands are secreted mostly by CAFs[44,45,54,55]. A recent study confirmed that Wnt2 acts in an autocrine manner, generating morphogenetic changes in fibroblasts and contributing to the invasive and metastatic capacity of CRC-derived cells[44]. In vitro experiments revealed that the treatment of DLD1 and HCT116 cells (both derived from CRC) with conditioned medium obtained from CAFs transfected with small interfering RNA targeting Wnt2, significantly decreased cell invasion and migration[45]. It is of clinical relevance that these ligands are overexpressed in the tumor stroma of CRC patients[45,53]. Moreover, the aberrant expression of Wnt ligand is correlated with a worse prognosis[44]. EVs play a key role in activating Wnt signaling in CRC. On the one hand, EVs can transport mutant β-catenin and activate the Wnt signaling pathway in the recipient cells, thus promoting CRC progression[56]. Regarding the effects of EVs on TME cells, Hu and colleagues demonstrated in vitro that fibroblast-derived exosomes (one type of EVs) promote the tumor growth of CSCs upon treatment with 5-fluorouracil (5-FU) or oxaliplatin (OXA)[57]. Further investigations of the same group in subcutaneous xenografts achieved through HT-29 CRC-derived cells and fibroblast co-implantation demonstrated that stromal fibroblasts can secrete exosomes loaded with Wnt ligands. Once these exosomes interact with differentiated CRC cells, Wnt ligands induce the phenotypic reversion of CRC cells to CSCs, which includes features such as the expression of CSCs markers and elevated Wnt activity[57].

Hypoxia is a prevalent condition in the solid tumor TME, which involves oxygen pressures of less than 5-10 mmHg. The hypoxic TME can deeply influence CRC, and these effects may be mediated by β-catenin. For instance, it was found in vitro that Wnt/β-catenin signaling is responsible for the hypoxia-induced self-renewal of colorectal stem cells[58]. Furthermore, Huang and collaborators observed that hypoxic CRC cells can secrete exosomes enriched with Wnt4 Ligands. These exosomes can activate β-catenin signaling in normoxic CRC cells and stimulate prometastatic behaviors such as cell migration and invasion[59]. In another work, the authors demonstrated that Wnt4-loaded exosomes secreted by the tumor cells promoted angiogenesis through the proliferation and migration of ECs. These effects were induced in conditions of hypoxia and mediated by the activation of Wnt/β-catenin signaling[60].

Autophagy is a physiological process through which normal cells degrade intracellular components to maintain cellular homeostasis. It is postulated as an alternative to cell death when the apoptotic machinery fails and is associated with both CRC and the TME, with controversial findings[61-63]. TME hypoxia induces autophagy and also activates several tumor escape mechanisms[62]. Some of the molecular mechanisms involved in the modulation of autophagy are phosphoinositide 3-kinases (PI3K)/protein kinase B (also known as Akt)/mammalian target of rapamycin, TGF-β, Notch, and Wnt/β-catenin signaling[63]. According to evidence, autophagy induced in the early stages of tumor development prevents the infiltration of immune cells as well as tumor cell death. Autophagy can also promote tumor progression in the advanced stages of the disease[63]. On the other hand, it has been reported that the relationship between autophagy and increased lymphocytic infiltration in the TME is mediated by the autophagy-related protein Vps34 in CRC. The modulation of this autophagy-related protein would improve the efficacy of immunotherapies[64]. However, it is still necessary to conduct additional studies on the interrelationship between the Wnt/β-catenin signaling pathway and autophagy, and which TME factors can mediate these events in CRC.

Because about 90% of patients with CRC present with mutations in genes associated with the Wnt pathway[65], more efforts have been made to identify other molecules with the ability to regulate β-catenin. In line with this evidence, our research group also reported interesting results regarding the participation of PTHrP in the activation of signaling pathways associated with β-catenin. Our published data showed that the exposition of cells derived from colorectal adenocarcinoma, to exogenous PTHrP increases β-catenin expression and upregulates cyclin D1 and c-Myc oncogenes with the concomitant cell proliferation induction[66]. Regarding the role of this cytokine in the TME, we demonstrated in vitro that tumor conditioned media from PTHrP-exposed CRC cells promotes pro-angiogenic characteristics in the stromal EC line HMEC-1 through VEGF modulation[35]. In addition, we recently found that PTHrP modulates the protein expression and secretion from the HMEC-1 cells of secreted protein acidic and cysteine-rich, a TME factor involved in the EMT program. This event promotes molecular and morphological changes associated with EMT in cells derived from CRC[36]. In this context, we believe that cross-communication may be initially established through overexpression and secretion of PTHrP by the tumor, which in turn affects the behavior of neoplastic cells and TME cells inducing the subsequent release from them of this cytokine and other factors. More studies are needed to elucidate which stromal cells can secrete PTHrP and participate in the modulation of the β-catenin pathway. Several authors have also described the relationship between PTHrP and activation of the β-catenin signaling pathway in other types of cancer. In prostate cancer-derived cells, PTHrP downregulates the Wnt inhibitor, DKK1, promoting aberrant activation of this pathway[67]. Other research has shown that PTHrP modulates activation of the canonical Wnt/β-catenin signaling in the mammary mesenchyme[68]. Moreover, Johnson and collaborators reported that Wnt activation promotes PTHrP expression in breast and lung tumor cells[69].

Tumor small and long non-coding RNAs are other factors transported by EVs, which are also involved in the modulation of β-catenin signaling. Colorectal cancer-associated lncRNA (CCAL) and H19 (lncRNA19) expressed by CAFs are capable of stimulating aberrant activation of β-catenin in tumor cells[9,25]. Besides, the overexpression of miR-103 and miR-107 has been detected in the plasma and tumor tissue of patients with CRC[70]. In HCT116 and HT29 cell lines, miR-103/107 overexpression enhanced β-catenin activity and its nuclear translocation. These molecules act by inhibiting Axin2, a negative regulator of the Wnt/β-catenin pathway, and in this way, β-catenin is stabilized and its activation is prolonged[70]. Another study showed that miR-92a-3p secreted by CAFs, induces β-catenin nuclear translocation and the expression of target genes related to stemness, the EMT program, and chemoresistance in the human CRC cell lines SW480, SW620, and LOVO. Moreover, the high expression of exosomal miR-92a-3p in serum predicts metastasis and chemotherapy resistance in CRC patients[24]. The overexpression of miR-100, miR-125b, miR-27a, miR-135, and MIR100HG in CRC tumors modulates the canonical Wnt pathway, promoting events such as proliferation and invasion[71,72].

In the last decade, research on small non-coding RNA molecules has focused on cRNAs. Increasing studies have shown that these molecules intervene in the development and progression of various types of tumors including CRC[73]. Results obtained by Zhang et al[73] show that circAGFG1 activates the Wnt/β-catenin pathway in SW480 and HCT116 cells promoting the transcription of the CTNNB1 gene. Two years earlier, Fang and his group revealed that circ_100290 promotes CRC progression through activation of the Wnt/β-catenin pathway[74]. Other cRNAs involved in the activation of this signaling pathway in CRC are Hsa_circ_0005075[75], circMTO1[76] and Circ-PRKDC[72]. On the other hand, cRNAs such as circPRKDC and cirITCH act by inhibiting the activity of the β-catenin pathway[77,78]. The action of most of these molecules is mediated by miRNAs[26,79].

HGF and its receptor RTK Met are associated with β-catenin-related pathways in CRC cells, promoting a CSC phenotype, metastasis, and drug resistance[46,51,80,81]. Other factors including tumor necrosis factor-α (TNF-α) also stimulate the main regulatory mechanism of β-catenin activation in CRC cells. Wei and colleagues demonstrated that under TNF-α action, the CSCs derived from the HT29 cell line increase the survival rate and their invasive capacity via the Wnt pathway. These actions are mediated by the modulation of β-catenin related proteins expression, such as c-Myc, cyclin D1, E-cadherin, and vimentin, which are cell cycle regulators associated with the stimulation of cell proliferation and/or the progress of the EMT program[82]. Molecules released from tumor cells including PLD2, modify the secretome of CAFs by promoting the release of the following factors: Macrophage migration inhibitory factor (MIF), HGF, and CCL2. The results shown in this article confirm that the treatment of CRC-derived cells (HCT116 and LS180) with these three factors induces the activation of the Wnt/β-catenin signaling pathway. The authors of this work observed a reduction of β-catenin phosphorylation together with an increase in its total expression after treatment with CCL2, HGF, and MIF, as well as a decrease and an increase in the expression of Axin1 and c-Myc, respectively, consistent with Wnt activation. This research also demonstrated that gene transcription induced by the Wnt signaling pathway correlates with high expression of PLD2 and CAFs-derived factors in CRC patients[38].

Osteopontin (OPN) is another factor that plays an important role in the regulatory mechanism of β-catenin. It is a glycoprotein synthesized by a great variety of cells and is overexpressed in CRC. This protein is capable of activating β-catenin via Akt and subsequently GSK3β, or by recruiting and activating mitogen-activated protein kinase (MAPK)[83,84]. Youssef and Osman analyzed tumor samples and found an association between the expression of nuclear OPN/β-catenin and markers related to poor prognosis in CRC[85]. In the last decade, an association has also been established between OPN expression and the activation and nuclear translocation of β-catenin in other types of cancer[83,84]. Recent research has shown that this protein is a target gene of the Wnt pathway[86] and in these cases, positive feedback from the system can be generated. Based on the evidence analyzed in the previous lines, positive feedback is highly probable between OPN secretion and the modulation of the Wnt/β-catenin pathway in CRC.

Other cytokines and growth factors released by the TME cells promote activation of the canonical Wnt pathway in cells derived from human CRC[87]. Bone morphogenetic proteins (BMPs) are cytokines closely linked to angiogenesis, EMT program, and induction of CSCs[88]. Voorneveld and colleagues transfected HCT116, HT29, LS174T, RKO, and SW480 cells with the BMPR2 plasmid, increasing BMP signaling. Hyperactivation of this pathway only downregulated Wnt signaling in HCT116 and LS174T cells, both were SMAD4-positive and had wild type p53. In the remaining cell lines, the Wnt pathway was activated by BMP signaling. This work demonstrated that modulation of Wnt/β-catenin signaling by the BMP pathway is dependent on SMAD4 and p53 status[5,50]. Regarding the role of interleukins (ILs), neoplastic cells promote the release of IL-1β, mainly from TME macrophages. In HCT116 cells, this cytokine induces β-catenin activation and consequently the expression of target genes, through nuclear factor kappa B (NF-KB)/Akt signaling pathway[87,89]. IL-6 secreted by TME cells induces activation of the Wnt/β-catenin pathway through the signal transducer and activator of transcription 3/extracellular signal-related kinase (ERK) pathway favoring the promotion of the aggressive phenotype of SW48 cells[90]. Chemokines such as C-X-C Motif Chemokine Ligand 12 (CXCL12) are overexpressed in CRC. Song et al[91] showed that the increased expression of this cytokine and its receptor (CXCR4) in tumor cells induces activation of the canonical Wnt pathway, possibly mediating the processes of proliferation, survival, and invasion[91]. However, not all cytokines act by promoting aberrant activation of the Wnt pathway. IL-37 mainly expressed by TAMs[92] has been observed to inhibit the proliferation, migration, and CSC phenotype by directly repressing the expression of β-catenin and consequently its translocation to the nucleus[49].

Bacteria, fungi, and viruses constitute the microbiota residing within the gastrointestinal tract. However, the gut microbiota and their metabolites can be altered in the TME and directly influence CRC progression[93,94]. Various bacteria have been implicated in these processes by modulating the β-catenin pathway. Fusobacterium nucleatum is a CRC inductor and suppressor of NK cell activity[93,95] Recently, it was reported that F. nucleatum can stimulate annexin A1 protein, specifically expressed in CRC cells. Then annexin A1 can induce β-catenin nuclear accumulation and finally the exacerbation of CRC cell proliferation[96]. On the opposite side, as extensively analyzed by Li et al[93] in their review article, other bacteria could be beneficial for CRC treatment or its prevention. For example, Lactobacillus and Clostridium butyricum are probiotics that have inhibitory effects on CRC progression via modulation of the β-catenin pathway[93]. This background shows that the complex interrelations between tumor cells and their microenvironment (including the microbiota) involve the participation of one of the most outstanding signaling pathways in the promotion and development of CRC, the β-catenin pathway. Therefore, it is important to consider these phenomena in the development of new treatments that focus on inhibiting or silencing the activators/exacerbators of this pathway.

ROLE OF β-CATENIN AND THE TME IN THE CHEMORESISTANCE OF CRC

Among the most used chemotherapeutic drugs, approved as first- and second-line adjuvants in CRC, are 5-FU, irinotecan (CPT-11), OXA, capecitabine, and leucovorin[97,98]. Depending on the CRC subtype, these drugs are also used in combination with monoclonal antibodies, such as bevacizumab or cetuximab[5]. On certain occasions, signs of a poor prognosis appear. Therefore, neoadjuvant or adjuvant chemotherapy must be implemented[99]. Despite this, patients with advanced grades of metastatic disease continue to lag far behind in successful treatment. One of the main causes of failure in treatment is the chemoresistance observed in about half of patients with CRC[99,100]. In this regard, the cell responses that mediate drug resistance are associated with hypoxic conditions, autophagy, induction of the CSC phenotype, and the EMT program[101,102]. At the molecular level, the development of drug-resistant tumors involves alterations of Wnt/β-catenin among other important signaling pathways such as Hedgehog and Notch. It also includes disturbances in the expression of anti-apoptotic proteins, the overexpression of drug transporters of the ATP-binding cassette (ABC) family, increased aldehyde dehydrogenase 1 enzyme activity and also the release of factors from the TME[99,103]. Regarding the Wnt pathway, its aberrant activation involves induction of the CSC phenotype, the EMT program, the expression of proteins from the ABC family and factors from the TME, all of these events being related to drug resistance[99]. Furthermore, various biological mediators released by stromal cells are capable of inducing chemoresistance through the Wnt/β-catenin pathway in tumor cells[9,25,104]. In this regard, in vitro studies made by Hu and colleagues revealed that miR-92a-3p secreted from CAFs activates the Wnt/β-catenin signaling, decreasing the sensitivity to the combination of 5-FU/OXA in SW480, SW620, and Lovo CRC-derived cells[24]. Moreover, in vivo studies have demonstrated that exosomes secreted by fibroblasts can load Wnt ligands, thus promoting CSC features and resistance to 5-FU or OXA. On the contrary, the inhibition of Wnt release reverted the observed effects[57]. CAFs are capable of inducing drug resistance by transferring small nucleic acid molecules such as H19 or CCAL lncRNA and activating β-catenin in neighboring cells[25]. H19 lncRNA can also trigger autophagy via SIRT1 and consequently induce resistance to 5-FU in CRC cells[105]. Other miRNAs such as miR-27[106], miR-103, and miR-107 decrease the sensitivity of HCT116 cells to 5-FU and OXA, through hyperactivation of the β-catenin pathway[9]. Zhou and colleagues reported that the inhibition of the Wnt/β-catenin signaling pathway by the overexpression of miR-506 decreases the chemoresistance response of HCT116 cells to OXA. The mechanism that explains this effect is that the gene and protein expression of the transporter of the ABC family and multidrug resistance protein 1/P-glycoprotein are blocked[107]. Several cRNAs are also involved in the acquisition of drug resistance by CRC cells through modulation of the β-catenin axis. For instance, circ-PRKDC participates in the development of chemoresistance to 5-FU in SW620 and SW480 cell lines by regulation of miR-375 and the Wnt/β-catenin pathway[77]. These molecules constitute a novel target for the development of alternative therapies for CRC, given their role in the communication between tumor cells and the TME and their participation in the regulation of Wnt/β-catenin[26,108].

Macrophages also play a crucial role in the development of resistance to drugs. IL-1β released by TAMs activates the canonical Wnt pathway through NF-κB/Akt and subsequently induces the expression of genes associated with chemoresistance. This has been observed in several cell lines derived from CRC such as HCT116, Hke-3, SW480, and RKO[109,110].

In our laboratory, we previously reported that PTHrP, acting as a cytokine, induces chemoresistance to CPT-11 through ERK MAPK and Akt pathways in the CRC cell lines Caco-2 and HCT116[34]. Moreover, as described in previous sections, we also registered nuclear β-catenin nuclear-increased expression after PTHrP treatment in CRC cells[37]. In view of these data, we hypothesize that this protein also participates in the resistance to CPT-11. This is our current research focus.

Growth factors are also involved in the therapy resistance mediated by β-catenin. Activation of the c-MET pathway through HGF signaling is related to drug resistance in many types of tumors[111]. Several works have already shown that HGF is associated with both Met and β-catenin pathways[52,54]. Studies published by Woo and collaborators showed that this factor is related to resistance to CPT-11 in CRC cells[112]. Since the HGF/Met axis plays an important role in CRC, molecules such as inhibitors, neutralizing antibodies, and antagonists of this pathway have been designed. To date, pre-clinical trials have failed and demonstrated poor efficacy[111,113]. One of the possible causes of the resistance of CRC cells to HGF pathway inhibitors is the activation of the Wnt/β-catenin signaling pathway[113]. All of these data highlight the need for consideration of the mitogenic pathways and paracrine factors released by both the tumor and the TME as targets to revert the chemoresistance.

Li et al[114] made a relevant contribution in the field of basic sciences related to CRC therapy and TME cells. They observed that the pharmacological treatment of CAFs inhibits their recruitment and increases the sensitivity of tumor cells to OXA[114]. Chemotherapy combined with treatments that inhibit CAFs recruitment and/or their activity may represent a novel method for the improvement of tumor response to chemotherapy[114]. This area of research is practically vacant and needs to be further explored since other unknown factors derived from TME may be able to stimulate the nuclear translocation of β-catenin and modulate the events related to the chemoresistance of CRC cells.

THE TME AND β-CATENIN PATHWAY: FUTURE PERSPECTIVES FOR CRC MANAGEMENT

In the last decade, therapeutic inhibition of the β-catenin pathway has been considered an invaluable tool for developing new therapy regimens for patients with CRC[115-117]. Despite this, as previously stated, there are still no approved drugs for the treatment of CRC or clinical trials based on the targeting of this signaling pathway[15,117]. For these reasons, comprehension of the regulatory mechanisms of the Wnt/β-catenin signaling pathway will not only expand the knowledge about the pathogenesis and evolution of CRC but also improve the treatment with the implementation of new targeted therapies. Many small molecules with β-catenin inhibitory effects have been developed such as tankyrase inhibitors (TNKSi), R-spondin inhibitors, and porcupine enzyme inhibitors. tankyrases (TNKSs) favor Wnt signaling by inducing Axin degradation. Therefore, TNKSi including XAV939 (XAV), JW55, NVP-TNKS656, and GOO7-LK are effective approaches for inhibiting this pathway in the preclinical stage of validation[118,119]. Moon and collaborators recently discovered that β-catenin expression influences the response of CRC-derived cells to MEK inhibitors, and that TNKSi allows the resistance to this treatment to be exceeded in CRC cell lines with mutant KRAS and PI3K. These results demonstrate the possible efficacy of combined treatments with inhibitors of MEK and β-catenin signaling pathways. However, this kind of therapeutic strategy is not yet in the clinical trial phase[118], and there is even controversy on the implementation of this type of scheme[116]. Drugs that disable enzymes related to Wnt secretion are another possibility to inhibit the Wnt/β-catenin. These compounds are denominated IWP-2, LGK974, or ETC-159 and inhibit porcupine enzyme actions. Some R-spondin inhibitors, such as OMP-131R10 (rosmantuzumab), are also currently under investigation for the treatment of CRC. When this ligand binds to the receptor leucine rich repeat containing G protein-coupled receptor 4 (LGR4) or LGR5, it enhances Wnt signaling, so blocking this point would consequently decrease the transcriptional activity of β-catenin[119].

One of the therapeutic targets that is being studied for CRC treatment is the use of antibodies or small molecules for the inhibition of Wnt ligands, like OMP-54F28, OMP-18R5, and OTSA101[116]; the disruption of the transcriptional activity of β-catenin using small molecules that prevent the binding of this protein with the nuclear transcription factors TCF and LEF, such as LF3 and 2,4-diamino-quinazoline; or pharmacological drugs that stimulate β-catenin proteasomal degradation as MSAB[119,120].

In addition to the hyper-activating mutations affecting the β-catenin pathway in CRC, as previously mentioned, cross-communication through paracrine signals between tumor cells and TME cells amplify these effects. The Wnt/β-catenin pathway is key for immune cells differentiation and functioning[120], the aberrant activation of this signaling alters the activation and downregulation of the immune response, especially related to dendritic cells (DCs) and T cells[119]. In the TME, the β-catenin pathway-dependent production of cytokines, such as IL-10, by DCs has been shown to induce immune tolerance through CD4+ and CD8+ T cells[121]. Moreover, it was reported that β-catenin signaling is activated in tumors with an inflammatory microenvironment, immune evasion and poor infiltration of CD8+ T cells[119,122,123]. In CRC, this immune response is favored by the activation of β-catenin, Myc, and RAS[7,124,125]. Currently known mechanisms that mediate immune exclusion and evasion through the Wnt/β-catenin pathway are: Modulating the production and release of cytokines from DCs and consequently diminishing CD8+ T cells infiltration; stimulating the production and release of soluble factors like Snail and IL-1β from tumor cells and TAMs, respectively; and increasing regulatory T cells survival, which are also effective inhibitors of CD8+ T cells[120,126]. To date, there are some inhibitors related to the immune response that are approved for clinical use, one of them is programmed death-1[127]. However, although immunomodulation can be effective in the treatment of CRC, resistance often occurs. Given that several of these drugs target CD8+ T cells, one of the explanations for the failure of these therapies may be the hyperactivation of the β-catenin pathway in the TME[128]. From this point of view, the current research focuses on the importance of the development of anticancer therapies that target the Wnt/β-catenin pathway as a checkpoint to improve the efficacy of immunotherapies, mainly by restoring T-cell infiltration[119,120,122].

In previous sections, we described the role of several TME-derived factors that activates the β-catenin signaling pathway. Currently, clinical research is aimed at blocking or inhibiting the effects of these molecules on CRC. In our laboratory, we have found that the selective inhibition of the HGF receptor, the RTK Met, suppresses β-catenin phosphorylation at domains that favor its nuclear translocation induced by PTHrP. Concerning this, Rimassa and his research group[129] explored in a clinical phase II study the effect of an oral selective Met inhibitor (tivantinib) and found that, in combination with another drug, at least 10% of patients with CRC respond better to the therapy. This drug is a possible emerging treatment that is already in phase III study in other types of tumors[130]. Another drug that inhibits the activity of the Met receptor by inducing the degradation of β-catenin is celecoxib[116]. However, recent evidence indicates that the combined treatment of celecoxib with the usual chemotherapy did not improve overall survival or progression-free time in patients with CRC[131].

On the other hand, a large number of clinical trials have been developed using cytokine-based immunotherapy for CRC. Different drugs targeting IL, prostaglandins, CXCL, TNF-α and TGF-β superfamily factors are in clinical trials. Although it has not yet been proven that they generate an effect as monotherapy or in combined treatments with commonly used drugs, future expectations are very optimistic[132,133].

Employing non-coding RNA such as miRNAs or cRNAs is a promising solution to overcome CRC drug resistance. It has been seen that miR505, miR199a/b and miR320 decreased sensitivity to cisplatin and OXA by modulating the activity of the Wnt/β-catenin pathway. Others, such as miR30-5p, favor chemosensitivity in CRC cells[134]. In this work, we have also described the role of TME-derived cRNAs in activating the β-catenin signaling pathway and its influence on CRC development and progression. Nevertheless, there is still a lot to explore about its potential role in future therapies for CRC. In this regard, recent research indicates that knockdown circ-PRKDC decreases the resistance of CRC-derived cells to 5-FU by modulating the Wnt/β-catenin pathway[77]. These results could postulate a new strategy to deal with multidrug resistance in CRC. Additionally, Viralippurath and collaborators propose that gene therapy could be aimed at silencing these oncogenic cRNAs or increasing the effects of those molecules that act as tumor suppressors[26]. Despite these findings, to date, there are no available therapies that use these small molecules to evade the strategies of tumor cells against chemotherapeutic drugs. More studies are needed regarding the role of non-coding RNAs in CRC chemoresistance as well as for the development of delivery strategies for these molecules in new targeted therapies. Figure 4 schematizes current studies related to the disruption of the interrelationships between TME and CRC cells that trigger β-catenin activation and are involved in treatment failure.

Figure 4
Figure 4 Pharmacological targeting of the tumor microenvironment factors involved in the activation of β-catenin signaling in colorectal cancer. Several therapeutic strategies to block or inhibit the induction of the β-catenin pathway by tumor microenvironment factors in colorectal cancer (CRC) are currently under clinical study. This figure is original for this work. APC: Adenomatous polyposis coli; BMP: Bone morphogenic protein; CK1α: Casein kinase 1 alpha; CXCL: C-X-C motif chemokine ligand; GSK3β: Glycogen Synthase Kinase 3 Beta; HGF: Hepatocyte growth factor; IL: Interleukin; PTHrP: Parathyroid Hormone-related Peptide; PG: prostaglandin; TCF/LEF: T-cell factor/lymphoid enhancer factor; TGF-β: Transforming growth factor-beta; TNF-α: Tumor necrosis factor-α; Wnt: Wingless protein.

As previously mentioned, autophagy is a mechanism associated with chemoresistance in CRC[105]. Recent investigations aim to find drugs that help reverse the autophagy process and overcome chemoresistance in CRC[135]. According to Pérez-Plasencia et al[62], the involvement of Wnt/β-catenin signaling in the regulation of autophagy was demonstrated in several types of cancer, such as leukemia, hepatocarcinoma, squamous cell carcinoma, lung cancer and prostate cancer[62]. However, it is just a matter of time before the relationship between these two pathways in CRC will be considered crucial for future pharmacological treatments. In their work, the authors postulate several therapeutic approaches based on drugs that are capable of simultaneously disrupting components of β-catenin and autophagy pathways[61].

From what we have analyzed in this work, it is important to highlight that concerning CRC, the achievement of successful future therapies will involve the use of combined pharmacological compounds that inhibit not only the canonical β-catenin signaling pathway but also the TME- derived signaling.

CONCLUSION

The cells of the TME are mainly responsible for triggering the mechanisms that determine a worse prognosis of cancer disease. These cells and the biological mediators released in the neoplastic context regulate the expression of ligands and modulate the activity of pro-tumor signaling pathways. TME cells control the fate of the tumor cells through permanent bidirectional communication. It is known that a considerable proportion of CRC patients evolve unfavorably due to chemoresistance. To improve CRC therapies, it is necessary to expand the knowledge about the mechanisms that play a critical role in the development, progression and chemoresistance of CRC.

Given that the factors associated with the TME activate the β-catenin signaling pathway, it will be possible in the future to improve the response of CRC patients to treatment. This goal could be achieved through new therapeutic interventions based on interrupting the crosstalk between the stroma and the tumor that control the β-catenin pathway activation. From the exhaustive search of the bibliography made by the authors of this work, it is concluded that there are multiple vacant areas in the research related to chemoresistance induced by cells from the tumor environment. Even to date, new types of biomarkers are continuously identified and related to a worse prognosis and aggressiveness of CRC. For this reason, it is imperative to strengthen in vitro and preclinical studies to support clinical trials in order to achieve novel therapeutic approaches considering the background of each patient and personalized medicine.

ACKNOWLEDGEMENTS

The authors would like to thank Antonela Rossi Bertone for her helpful advice. Being a teacher of English graduated from Instituto Superior Juan XXIII (from Bahia Blanca city, Argentina) and currently working at San Cayetano school (in the same city), she revised the paper and made useful comments as regards the spelling, grammar and punctuation of this manuscript. MBND and MJM thanks CONICET for the fellowships.

Footnotes

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

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country/Territory of origin: Argentina

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B, B, B, B

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Koustas E, Greece; Pan JS, China; Rahimi-Movaghar E, Iran; Scurtu RR, Romania; Ying HQ, China S-Editor: Fan JR L-Editor: Filipodia P-Editor: Fan JR

References
1.  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.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin. 2020;70:7-30.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Erratum: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2020;70:313.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Siegel RL, Miller KD, Fedewa SA, Ahnen DJ, Meester RGS, Barzi A, Jemal A. Colorectal cancer statistics, 2017. CA Cancer J Clin. 2017;67:177-193.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Villalba M, Evans SR, Vidal-Vanaclocha F, Calvo A. Role of TGF-β in metastatic colon cancer: it is finally time for targeted therapy. Cell Tissue Res. 2017;370:29-39.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Martín MJ, Gentili C, Lassalle V. In vitro Biological Tests as the First Tools To Validate Magnetic Nanotheranostics for Colorectal Cancer Models. ChemMedChem. 2020;15:1003-1017.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Guinney J, Dienstmann R, Wang X, de Reyniès A, Schlicker A, Soneson C, Marisa L, Roepman P, Nyamundanda G, Angelino P, Bot BM, Morris JS, Simon IM, Gerster S, Fessler E, De Sousa E Melo F, Missiaglia E, Ramay H, Barras D, Homicsko K, Maru D, Manyam GC, Broom B, Boige V, Perez-Villamil B, Laderas T, Salazar R, Gray JW, Hanahan D, Tabernero J, Bernards R, Friend SH, Laurent-Puig P, Medema JP, Sadanandam A, Wessels L, Delorenzi M, Kopetz S, Vermeulen L, Tejpar S. The consensus molecular subtypes of colorectal cancer. Nat Med. 2015;21:1350-1356.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Stastna M, Janeckova L, Hrckulak D, Kriz V, Korinek V. Human Colorectal Cancer from the Perspective of Mouse Models. Genes (Basel). 2019;10.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Yuan S, Tao F, Zhang X, Zhang Y, Sun X, Wu D. Role of Wnt/β-Catenin Signaling in the Chemoresistance Modulation of Colorectal Cancer. Biomed Res Int. 2020;2020:9390878.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Flores-Hernández E, Velázquez DM, Castañeda-Patlán MC, Fuentes-García G, Fonseca-Camarillo G, Yamamoto-Furusho JK, Romero-Avila MT, García-Sáinz JA, Robles-Flores M. Canonical and non-canonical Wnt signaling are simultaneously activated by Wnts in colon cancer cells. Cell Signal. 2020;72:109636.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Bian J, Dannappel M, Wan C, Firestein R. Transcriptional Regulation of Wnt/β-Catenin Pathway in Colorectal Cancer. Cells. 2020;9.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Anthony CC, Robbins DJ, Ahmed Y, Lee E. Nuclear Regulation of Wnt/β-Catenin Signaling: It's a Complex Situation. Genes (Basel). 2020;11.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Chatterjee A, Paul S, Bisht B, Bhattacharya S, Sivasubramaniam S, Paul MK. Advances in targeting the WNT/β-catenin signaling pathway in cancer. Drug Discov Today. 2022;27:82-101.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Voloshanenko O, Schwartz U, Kranz D, Rauscher B, Linnebacher M, Augustin I, Boutros M. β-catenin-independent regulation of Wnt target genes by RoR2 and ATF2/ATF4 in colon cancer cells. Sci Rep. 2018;8:3178.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Disoma C, Zhou Y, Li S, Peng J, Xia Z. Wnt/β-catenin signaling in colorectal cancer: Is therapeutic targeting even possible? Biochimie. 2022;195:39-53.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Krishnamurthy N, Kurzrock R. Targeting the Wnt/beta-catenin pathway in cancer: Update on effectors and inhibitors. Cancer Treat Rev. 2018;62:50-60.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Li J, Chen D, Shen M. Tumor Microenvironment Shapes Colorectal Cancer Progression, Metastasis, and Treatment Responses. Front Med (Lausanne). 2022;9:869010.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  van Pelt GW, Sandberg TP, Morreau H, Gelderblom H, van Krieken JHJM, Tollenaar RAEM, Mesker WE. The tumour-stroma ratio in colon cancer: the biological role and its prognostic impact. Histopathology. 2018;73:197-206.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Huang D, Du X. Crosstalk between tumor cells and microenvironment via Wnt pathway in colorectal cancer dissemination. World J Gastroenterol. 2008;14:1823-1827.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Sandberg TP, Stuart MPME, Oosting J, Tollenaar RAEM, Sier CFM, Mesker WE. Increased expression of cancer-associated fibroblast markers at the invasive front and its association with tumor-stroma ratio in colorectal cancer. BMC Cancer. 2019;19:284.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Laplagne C, Domagala M, Le Naour A, Quemerais C, Hamel D, Fournié JJ, Couderc B, Bousquet C, Ferrand A, Poupot M. Latest Advances in Targeting the Tumor Microenvironment for Tumor Suppression. Int J Mol Sci. 2019;20.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Siveen KS, Raza A, Ahmed EI, Khan AQ, Prabhu KS, Kuttikrishnan S, Mateo JM, Zayed H, Rasul K, Azizi F, Dermime S, Steinhoff M, Uddin S. The Role of Extracellular Vesicles as Modulators of the Tumor Microenvironment, Metastasis and Drug Resistance in Colorectal Cancer. Cancers (Basel). 2019;11.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Gonda A, Kabagwira J, Senthil GN, Wall NR. Internalization of Exosomes through Receptor-Mediated Endocytosis. Mol Cancer Res. 2019;17:337-347.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Hu JL, Wang W, Lan XL, Zeng ZC, Liang YS, Yan YR, Song FY, Wang FF, Zhu XH, Liao WJ, Liao WT, Ding YQ, Liang L. CAFs secreted exosomes promote metastasis and chemotherapy resistance by enhancing cell stemness and epithelial-mesenchymal transition in colorectal cancer. Mol Cancer. 2019;18:91.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Ren J, Ding L, Zhang D, Shi G, Xu Q, Shen S, Wang Y, Wang T, Hou Y. Carcinoma-associated fibroblasts promote the stemness and chemoresistance of colorectal cancer by transferring exosomal lncRNA H19. Theranostics. 2018;8:3932-3948.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Viralippurath Ashraf J, Sasidharan Nair V, Saleh R, Elkord E. Role of circular RNAs in colorectal tumor microenvironment. Biomed Pharmacother. 2021;137:111351.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Gu C, Lu H, Qian Z. Matrine reduces the secretion of exosomal circSLC7A6 from cancer-associated fibroblast to inhibit tumorigenesis of colorectal cancer by regulating CXCR5. Biochem Biophys Res Commun. 2020;527:638-645.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Herrera M, Llorens C, Rodríguez M, Herrera A, Ramos R, Gil B, Candia A, Larriba MJ, Garre P, Earl J, Rodríguez-Garrote M, Caldés T, Bonilla F, Carrato A, García-Barberán V, Peña C. Differential distribution and enrichment of non-coding RNAs in exosomes from normal and Cancer-associated fibroblasts in colorectal cancer. Mol Cancer. 2018;17:114.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Wortzel I, Dror S, Kenific CM, Lyden D. Exosome-Mediated Metastasis: Communication from a Distance. Dev Cell. 2019;49:347-360.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Shao Y, Chen T, Zheng X, Yang S, Xu K, Chen X, Xu F, Wang L, Shen Y, Wang T, Zhang M, Hu W, Ye C, Yu X, Shao J, Zheng S. Colorectal cancer-derived small extracellular vesicles establish an inflammatory premetastatic niche in liver metastasis. Carcinogenesis. 2018;39:1368-1379.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Abu N, Othman N, Ab Razak NS, Bakarurraini NAAR, Nasir SN, Soh JEC, Mazlan L, Azman ZAM, Jamal R. Extracellular Vesicles Derived From Colorectal Cancer Affects CD8 T Cells: An Analysis Based on Body Mass Index. Front Cell Dev Biol. 2020;8:564648.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Karpisheh V, Nikkhoo A, Hojjat-Farsangi M, Namdar A, Azizi G, Ghalamfarsa G, Sabz G, Yousefi M, Yousefi B, Jadidi-Niaragh F. Prostaglandin E2 as a potent therapeutic target for treatment of colon cancer. Prostaglandins Other Lipid Mediat. 2019;144:106338.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Landskron G, De la Fuente López M, Dubois-Camacho K, Díaz-Jiménez D, Orellana-Serradell O, Romero D, Sepúlveda SA, Salazar C, Parada-Venegas D, Quera R, Simian D, González MJ, López-Köstner F, Kronberg U, Abedrapo M, Gallegos I, Contreras HR, Peña C, Díaz-Araya G, Roa JC, Hermoso MA. Interleukin 33/ST2 Axis Components Are Associated to Desmoplasia, a Metastasis-Related Factor in Colorectal Cancer. Front Immunol. 2019;10:1394.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Martín MJ, Gigola G, Zwenger A, Carriquiriborde M, Gentil F, Gentili C. Potential therapeutic targets for growth arrest of colorectal cancer cells exposed to PTHrP. Mol Cell Endocrinol. 2018;478:32-44.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Calvo N, Carriere P, Martín MJ, Gigola G, Gentili C. PTHrP treatment of colon cancer cells promotes tumor associated-angiogenesis by the effect of VEGF. Mol Cell Endocrinol. 2019;483:50-63.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Carriere P, Calvo N, Novoa Díaz MB, Lopez-Moncada F, Herrera A, Torres MJ, Alonso E, Gandini NA, Gigola G, Contreras HR, Gentili C. Role of SPARC in the epithelial-mesenchymal transition induced by PTHrP in human colon cancer cells. Mol Cell Endocrinol. 2021;530:111253.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Novoa Díaz MB, Carriere PM, Martín MJ, Calvo N, Gentili C. Involvement of parathyroid hormone-related peptide in the aggressive phenotype of colorectal cancer cells. World J Gastroenterol. 2021;27:7025-7040.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Muñoz-Galván S, Lucena-Cacace A, Perez M, Otero-Albiol D, Gomez-Cambronero J, Carnero A. Tumor cell-secreted PLD increases tumor stemness by senescence-mediated communication with microenvironment. Oncogene. 2019;38:1309-1323.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Molinier-Frenkel V, Castellano F. Immunosuppressive enzymes in the tumor microenvironment. FEBS Lett. 2017;591:3135-3157.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Cox TR. The matrix in cancer. Nat Rev Cancer. 2021;21:217-238.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Sameer AS, Nissar S. Toll-Like Receptors (TLRs): Structure, Functions, Signaling, and Role of Their Polymorphisms in Colorectal Cancer Susceptibility. Biomed Res Int. 2021;2021:1157023.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Angrini M, Varthaman A, Cremer I. Toll-Like Receptors (TLRs) in the Tumor Microenvironment (TME): A Dragon-Like Weapon in a Non-fantasy Game of Thrones. Adv Exp Med Biol. 2020;1263:145-173.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Kamal Y, Schmit SL, Frost HR, Amos CI. The tumor microenvironment of colorectal cancer metastases: opportunities in cancer immunotherapy. Immunotherapy. 2020;12:1083-1100.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Unterleuthner D, Neuhold P, Schwarz K, Janker L, Neuditschko B, Nivarthi H, Crncec I, Kramer N, Unger C, Hengstschläger M, Eferl R, Moriggl R, Sommergruber W, Gerner C, Dolznig H. Cancer-associated fibroblast-derived WNT2 increases tumor angiogenesis in colon cancer. Angiogenesis. 2020;23:159-177.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Aizawa T, Karasawa H, Funayama R, Shirota M, Suzuki T, Maeda S, Suzuki H, Yamamura A, Naitoh T, Nakayama K, Unno M. Cancer-associated fibroblasts secrete Wnt2 to promote cancer progression in colorectal cancer. Cancer Med. 2019;8:6370-6382.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Matsumoto K, Umitsu M, De Silva DM, Roy A, Bottaro DP. Hepatocyte growth factor/MET in cancer progression and biomarker discovery. Cancer Sci. 2017;108:296-307.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Goïta AA, Guenot D. Colorectal Cancer: The Contribution of CXCL12 and Its Receptors CXCR4 and CXCR7. Cancers (Basel). 2022;14.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Martín MJ, Calvo N, de Boland AR, Gentili C. Molecular mechanisms associated with PTHrP-induced proliferation of colon cancer cells. J Cell Biochem. 2014;115:2133-2145.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Amable G, Martínez-León E, Picco ME, Di Siervi N, Davio C, Rozengurt E, Rey O. Metformin inhibits β-catenin phosphorylation on Ser-552 through an AMPK/PI3K/Akt pathway in colorectal cancer cells. Int J Biochem Cell Biol. 2019;112:88-94.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Yan X, Zhao J, Zhang R. Interleukin-37 mediates the antitumor activity in colon cancer through β-catenin suppression. Oncotarget. 2017;8:49064-49075.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Voorneveld PW, Kodach LL, Jacobs RJ, van Noesel CJ, Peppelenbosch MP, Korkmaz KS, Molendijk I, Dekker E, Morreau H, van Pelt GW, Tollenaar RA, Mesker W, Hawinkels LJ, Paauwe M, Verspaget HW, Geraets DT, Hommes DW, Offerhaus GJ, van den Brink GR, Ten Dijke P, Hardwick JC. The BMP pathway either enhances or inhibits the Wnt pathway depending on the SMAD4 and p53 status in CRC. Br J Cancer. 2015;112:122-130.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Essex A, Pineda J, Acharya G, Xin H, Evans J; Reproducibility Project: Cancer Biology. Replication Study: Wnt activity defines colon cancer stem cells and is regulated by the microenvironment. Elife. 2019;8.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Vermeulen L, De Sousa E Melo F, van der Heijden M, Cameron K, de Jong JH, Borovski T, Tuynman JB, Todaro M, Merz C, Rodermond H, Sprick MR, Kemper K, Richel DJ, Stassi G, Medema JP. Wnt activity defines colon cancer stem cells and is regulated by the microenvironment. Nat Cell Biol. 2010;12:468-476.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Zhang L, Jiang X, Li Y, Fan Q, Li H, Jin L, Li L, Jin Y, Zhang T, Mao Y, Hua D. Clinical Correlation of Wnt2 and COL8A1 With Colon Adenocarcinoma Prognosis. Front Oncol. 2020;10:1504.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Koliaraki V, Pallangyo CK, Greten FR, Kollias G. Mesenchymal Cells in Colon Cancer. Gastroenterology. 2017;152:964-979.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Kalra H, Gangoda L, Fonseka P, Chitti SV, Liem M, Keerthikumar S, Samuel M, Boukouris S, Al Saffar H, Collins C, Adda CG, Ang CS, Mathivanan S. Extracellular vesicles containing oncogenic mutant β-catenin activate Wnt signalling pathway in the recipient cells. J Extracell Vesicles. 2019;8:1690217.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Hu Y, Yan C, Mu L, Huang K, Li X, Tao D, Wu Y, Qin J. Fibroblast-Derived Exosomes Contribute to Chemoresistance through Priming Cancer Stem Cells in Colorectal Cancer. PLoS One. 2015;10:e0125625.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Hu YB, Yan C, Mu L, Mi YL, Zhao H, Hu H, Li XL, Tao DD, Wu YQ, Gong JP, Qin JC. Exosomal Wnt-induced dedifferentiation of colorectal cancer cells contributes to chemotherapy resistance. Oncogene. 2019;38:1951-1965.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Dong HJ, Jang GB, Lee HY, Park SR, Kim JY, Nam JS, Hong IS. The Wnt/β-catenin signaling/Id2 cascade mediates the effects of hypoxia on the hierarchy of colorectal-cancer stem cells. Sci Rep. 2016;6:22966.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Huang Z, Yang M, Li Y, Yang F, Feng Y. Exosomes Derived from Hypoxic Colorectal Cancer Cells Transfer Wnt4 to Normoxic Cells to Elicit a Prometastatic Phenotype. Int J Biol Sci. 2018;14:2094-2102.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Huang Z, Feng Y. Exosomes Derived From Hypoxic Colorectal Cancer Cells Promote Angiogenesis Through Wnt4-Induced β-Catenin Signaling in Endothelial Cells. Oncol Res. 2017;25:651-661.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Pérez-Plasencia C, López-Urrutia E, García-Castillo V, Trujano-Camacho S, López-Camarillo C, Campos-Parra AD. Interplay Between Autophagy and Wnt/β-Catenin Signaling in Cancer: Therapeutic Potential Through Drug Repositioning. Front Oncol. 2020;10:1037.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Janji B, Berchem G, Chouaib S. Targeting Autophagy in the Tumor Microenvironment: New Challenges and Opportunities for Regulating Tumor Immunity. Front Immunol. 2018;9:887.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Pandurangan AK, Divya T, Kumar K, Dineshbabu V, Velavan B, Sudhandiran G. Colorectal carcinogenesis: Insights into the cell death and signal transduction pathways: A review. World J Gastrointest Oncol. 2018;10:244-259.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Janji B, Hasmim M, Parpal S, Berchem G, Noman MZ. Firing up the cold tumors by targeting Vps34. Oncoimmunology. 2020;9:1809936.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Rahmani F, Avan A, Hashemy SI, Hassanian SM. Role of Wnt/β-catenin signaling regulatory microRNAs in the pathogenesis of colorectal cancer. J Cell Physiol. 2018;233:811-817.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Zhang H, Yu C, Dai J, Keller JM, Hua A, Sottnik JL, Shelley G, Hall CL, Park SI, Yao Z, Zhang J, McCauley LK, Keller ET. Parathyroid hormone-related protein inhibits DKK1 expression through c-Jun-mediated inhibition of β-catenin activation of the DKK1 promoter in prostate cancer. Oncogene. 2014;33:2464-2477.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Hiremath M, Dann P, Fischer J, Butterworth D, Boras-Granic K, Hens J, Van Houten J, Shi W, Wysolmerski J. Parathyroid hormone-related protein activates Wnt signaling to specify the embryonic mammary mesenchyme. Development. 2012;139:4239-4249.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Johnson RW, Merkel AR, Page JM, Ruppender NS, Guelcher SA, Sterling JA. Wnt signaling induces gene expression of factors associated with bone destruction in lung and breast cancer. Clin Exp Metastasis. 2014;31:945-959.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Chen HY, Lang YD, Lin HN, Liu YR, Liao CC, Nana AW, Yen Y, Chen RH. miR-103/107 prolong Wnt/β-catenin signaling and colorectal cancer stemness by targeting Axin2. Sci Rep. 2019;9:9687.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Ba S, Xuan Y, Long ZW, Chen HY, Zheng SS. MicroRNA-27a Promotes the Proliferation and Invasiveness of Colon Cancer Cells by Targeting SFRP1 through the Wnt/β-Catenin Signaling Pathway. Cell Physiol Biochem. 2017;42:1920-1933.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Chu J, Fang X, Sun Z, Gai L, Dai W, Li H, Yan X, Du J, Zhang L, Zhao L, Xu D, Yan S. Non-Coding RNAs Regulate the Resistance to Anti-EGFR Therapy in Colorectal Cancer. Front Oncol. 2021;11:801319.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Zhang L, Dong X, Yan B, Yu W, Shan L. CircAGFG1 drives metastasis and stemness in colorectal cancer by modulating YY1/CTNNB1. Cell Death Dis. 2020;11:542.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Fang G, Ye BL, Hu BR, Ruan XJ, Shi YX. CircRNA_100290 promotes colorectal cancer progression through miR-516b-induced downregulation of FZD4 expression and Wnt/β-catenin signaling. Biochem Biophys Res Commun. 2018;504:184-189.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Jin YD, Ren YR, Gao YX, Zhang L, Ding Z. Hsa_circ_0005075 predicts a poor prognosis and acts as an oncogene in colorectal cancer via activating Wnt/β-catenin pathway. Eur Rev Med Pharmacol Sci. 2019;23:3311-3319.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Ge Z, Li LF, Wang CY, Wang Y, Ma WL. CircMTO1 inhibits cell proliferation and invasion by regulating Wnt/β-catenin signaling pathway in colorectal cancer. Eur Rev Med Pharmacol Sci. 2018;22:8203-8209.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Chen H, Pei L, Xie P, Guo G. Circ-PRKDC Contributes to 5-Fluorouracil Resistance of Colorectal Cancer Cells by Regulating miR-375/FOXM1 Axis and Wnt/β-Catenin Pathway. Onco Targets Ther. 2020;13:5939-5953.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Su K, Yi Q, Dai X, Liu O. Circular RNA ITCH: An Emerging Multifunctional Regulator. Biomolecules. 2022;12.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Geng Y, Zheng X, Hu W, Wang Q, Xu Y, He W, Wu C, Zhu D, Jiang J. Hsa_circ_0009361 acts as the sponge of miR-582 to suppress colorectal cancer progression by regulating APC2 expression. Clin Sci (Lond). 2019;133:1197-1213.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Wang J, Du Y, Liu X, Cho WC, Yang Y. MicroRNAs as Regulator of Signaling Networks in Metastatic Colon Cancer. Biomed Res Int. 2015;2015:823620.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Tomida C, Yamagishi N, Nagano H, Uchida T, Ohno A, Hirasaka K, Nikawa T, Teshima-Kondo S. VEGF pathway-targeting drugs induce evasive adaptation by activation of neuropilin-1/cMet in colon cancer cells. Int J Oncol. 2018;52:1350-1362.  [PubMed]  [DOI]  [Cited in This Article: ]
82.  Wei X, Li X, Kong F, Ma L, Sui Y, Chen D, Xu F. [TNF-α activates Wnt signaling pathway to promote the invasion of human colon cancer stem cells]. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi. 2018;34:982-988.  [PubMed]  [DOI]  [Cited in This Article: ]
83.  Zheng Y, Zhou C, Yu XX, Wu C, Jia HL, Gao XM, Yang JM, Wang CQ, Luo Q, Zhu Y, Zhang Y, Wei JW, Sheng YY, Dong QZ, Qin LX. Osteopontin promotes metastasis of intrahepatic cholangiocarcinoma through recruiting MAPK1 and mediating Ser675 phosphorylation of β-Catenin. Cell Death Dis. 2018;9:179.  [PubMed]  [DOI]  [Cited in This Article: ]
84.  Yi J, Liu Y, Zhang L, Fang C. Secreted phosphoprotein-1 accelerates the progression of human colorectal cancer through activating β-catenin signaling. Oncol Lett. 2021;21:372.  [PubMed]  [DOI]  [Cited in This Article: ]
85.  Youssef NS, Osman WM. Relationship between osteopontin and β-catenin immunohistochemical expression and prognostic parameters of colorectal carcinoma. Int J Clin Exp Pathol. 2015;8:1503-1514.  [PubMed]  [DOI]  [Cited in This Article: ]
86.  Ishigamori R, Komiya M, Takasu S, Mutoh M, Imai T, Takahashi M. Osteopontin Deficiency Suppresses Intestinal Tumor Development in Apc-Deficient Min Mice. Int J Mol Sci. 2017;18.  [PubMed]  [DOI]  [Cited in This Article: ]
87.  Kaler P, Augenlicht L, Klampfer L. Macrophage-derived IL-1beta stimulates Wnt signaling and growth of colon cancer cells: a crosstalk interrupted by vitamin D3. Oncogene. 2009;28:3892-3902.  [PubMed]  [DOI]  [Cited in This Article: ]
88.  Bach DH, Park HJ, Lee SK. The Dual Role of Bone Morphogenetic Proteins in Cancer. Mol Ther Oncolytics. 2018;8:1-13.  [PubMed]  [DOI]  [Cited in This Article: ]
89.  Kaler P, Galea V, Augenlicht L, Klampfer L. Tumor associated macrophages protect colon cancer cells from TRAIL-induced apoptosis through IL-1beta-dependent stabilization of Snail in tumor cells. PLoS One. 2010;5:e11700.  [PubMed]  [DOI]  [Cited in This Article: ]
90.  Gao S, Hu J, Wu X, Liang Z. PMA treated THP-1-derived-IL-6 promotes EMT of SW48 through STAT3/ERK-dependent activation of Wnt/β-catenin signaling pathway. Biomed Pharmacother. 2018;108:618-624.  [PubMed]  [DOI]  [Cited in This Article: ]
91.  Song ZY, Gao ZH, Chu JH, Han XZ, Qu XJ. Downregulation of the CXCR4/CXCL12 axis blocks the activation of the Wnt/β-catenin pathway in human colon cancer cells. Biomed Pharmacother. 2015;71:46-52.  [PubMed]  [DOI]  [Cited in This Article: ]
92.  La Fleur L, Botling J, He F, Pelicano C, Zhou C, He C, Palano G, Mezheyeuski A, Micke P, Ravetch JV, Karlsson MCI, Sarhan D. Targeting MARCO and IL37R on Immunosuppressive Macrophages in Lung Cancer Blocks Regulatory T Cells and Supports Cytotoxic Lymphocyte Function. Cancer Res. 2021;81:956-967.  [PubMed]  [DOI]  [Cited in This Article: ]
93.  Li J, Zhang AH, Wu FF, Wang XJ. Alterations in the Gut Microbiota and Their Metabolites in Colorectal Cancer: Recent Progress and Future Prospects. Front Oncol. 2022;12:841552.  [PubMed]  [DOI]  [Cited in This Article: ]
94.  Yachida S, Mizutani S, Shiroma H, Shiba S, Nakajima T, Sakamoto T, Watanabe H, Masuda K, Nishimoto Y, Kubo M, Hosoda F, Rokutan H, Matsumoto M, Takamaru H, Yamada M, Matsuda T, Iwasaki M, Yamaji T, Yachida T, Soga T, Kurokawa K, Toyoda A, Ogura Y, Hayashi T, Hatakeyama M, Nakagama H, Saito Y, Fukuda S, Shibata T, Yamada T. Metagenomic and metabolomic analyses reveal distinct stage-specific phenotypes of the gut microbiota in colorectal cancer. Nat Med. 2019;25:968-976.  [PubMed]  [DOI]  [Cited in This Article: ]
95.  Kim YJ, Kim BK, Park SJ, Kim JH. Impact of Fusobacterium nucleatum in the gastrointestinal tract on natural killer cells. World J Gastroenterol. 2021;27:4879-4889.  [PubMed]  [DOI]  [Cited in This Article: ]
96.  Rubinstein MR, Baik JE, Lagana SM, Han RP, Raab WJ, Sahoo D, Dalerba P, Wang TC, Han YW. Fusobacterium nucleatum promotes colorectal cancer by inducing Wnt/β-catenin modulator Annexin A1. EMBO Rep. 2019;20.  [PubMed]  [DOI]  [Cited in This Article: ]
97.  Parsons S, Maldonado EB, Prasad V. Comparison of Drugs Used for Adjuvant and Metastatic Therapy of Colon, Breast, and Non-Small Cell Lung Cancers. JAMA Netw Open. 2020;3:e202488.  [PubMed]  [DOI]  [Cited in This Article: ]
98.  Chang CL, Yuan KS, Wu ATH, Wu SY. Adjuvant Therapy for High-Risk Stage II or III Colon Adenocarcinoma: A Propensity Score-Matched, Nationwide, Population-Based Cohort Study. Cancers (Basel). 2019;11.  [PubMed]  [DOI]  [Cited in This Article: ]
99.  Hu T, Li Z, Gao CY, Cho CH. Mechanisms of drug resistance in colon cancer and its therapeutic strategies. World J Gastroenterol. 2016;22:6876-6889.  [PubMed]  [DOI]  [Cited in This Article: ]
100.  Meyers BM, Cosby R, Quereshy F, Jonker D. Adjuvant Chemotherapy for Stage II and III Colon Cancer Following Complete Resection: A Cancer Care Ontario Systematic Review. Clin Oncol (R Coll Radiol). 2017;29:459-465.  [PubMed]  [DOI]  [Cited in This Article: ]
101.  Shibue T, Weinberg RA. EMT, CSCs, and drug resistance: the mechanistic link and clinical implications. Nat Rev Clin Oncol. 2017;14:611-629.  [PubMed]  [DOI]  [Cited in This Article: ]
102.  Erin N, Grahovac J, Brozovic A, Efferth T. Tumor microenvironment and epithelial mesenchymal transition as targets to overcome tumor multidrug resistance. Drug Resist Updat. 2020;53:100715.  [PubMed]  [DOI]  [Cited in This Article: ]
103.  Gupta R, Bhatt LK, Johnston TP, Prabhavalkar KS. Colon cancer stem cells: Potential target for the treatment of colorectal cancer. Cancer Biol Ther. 2019;20:1068-1082.  [PubMed]  [DOI]  [Cited in This Article: ]
104.  Tauriello DV, Calon A, Lonardo E, Batlle E. Determinants of metastatic competency in colorectal cancer. Mol Oncol. 2017;11:97-119.  [PubMed]  [DOI]  [Cited in This Article: ]
105.  Wang M, Han D, Yuan Z, Hu H, Zhao Z, Yang R, Jin Y, Zou C, Chen Y, Wang G, Gao X, Wang X. Long non-coding RNA H19 confers 5-Fu resistance in colorectal cancer by promoting SIRT1-mediated autophagy. Cell Death Dis. 2018;9:1149.  [PubMed]  [DOI]  [Cited in This Article: ]
106.  Barisciano G, Colangelo T, Rosato V, Muccillo L, Taddei ML, Ippolito L, Chiarugi P, Galgani M, Bruzzaniti S, Matarese G, Fassan M, Agostini M, Bergamo F, Pucciarelli S, Carbone A, Mazzoccoli G, Colantuoni V, Bianchi F, Sabatino L. miR-27a is a master regulator of metabolic reprogramming and chemoresistance in colorectal cancer. Br J Cancer. 2020;122:1354-1366.  [PubMed]  [DOI]  [Cited in This Article: ]
107.  Zhou H, Lin C, Zhang Y, Zhang X, Zhang C, Zhang P, Xie X, Ren Z. miR-506 enhances the sensitivity of human colorectal cancer cells to oxaliplatin by suppressing MDR1/P-gp expression. Cell Prolif. 2017;50.  [PubMed]  [DOI]  [Cited in This Article: ]
108.  Zhang Q, Wang W, Zhou Q, Chen C, Yuan W, Liu J, Li X, Sun Z. Roles of circRNAs in the tumour microenvironment. Mol Cancer. 2020;19:14.  [PubMed]  [DOI]  [Cited in This Article: ]
109.  Kaler P, Godasi BN, Augenlicht L, Klampfer L. The NF-κB/AKT-dependent Induction of Wnt Signaling in Colon Cancer Cells by Macrophages and IL-1β. Cancer Microenviron. 2009;2:69-80.  [PubMed]  [DOI]  [Cited in This Article: ]
110.  Yoshida GJ. Regulation of heterogeneous cancer-associated fibroblasts: the molecular pathology of activated signaling pathways. J Exp Clin Cancer Res. 2020;39:112.  [PubMed]  [DOI]  [Cited in This Article: ]
111.  Kim KH, Kim H. Progress of antibody-based inhibitors of the HGF-cMET axis in cancer therapy. Exp Mol Med. 2017;49:e307.  [PubMed]  [DOI]  [Cited in This Article: ]
112.  Hwang K, Yoon JH, Lee JH, Lee S. Recent Advances in Monoclonal Antibody Therapy for Colorectal Cancers. Biomedicines. 2021;9.  [PubMed]  [DOI]  [Cited in This Article: ]
113.  Huang X, Li E, Shen H, Wang X, Tang T, Zhang X, Xu J, Tang Z, Guo C, Bai X, Liang T. Targeting the HGF/MET Axis in Cancer Therapy: Challenges in Resistance and Opportunities for Improvement. Front Cell Dev Biol. 2020;8:152.  [PubMed]  [DOI]  [Cited in This Article: ]
114.  Li M, Li M, Yin T, Shi H, Wen Y, Zhang B, Chen M, Xu G, Ren K, Wei Y. Targeting of cancerassociated fibroblasts enhances the efficacy of cancer chemotherapy by regulating the tumor microenvironment. Mol Med Rep. 2016;13:2476-2484.  [PubMed]  [DOI]  [Cited in This Article: ]
115.  Novellasdemunt L, Antas P, Li VS. Targeting Wnt signaling in colorectal cancer. A Review in the Theme: Cell Signaling: Proteins, Pathways and Mechanisms. Am J Physiol Cell Physiol. 2015;309:C511-C521.  [PubMed]  [DOI]  [Cited in This Article: ]
116.  Shang S, Hua F, Hu ZW. The regulation of β-catenin activity and function in cancer: therapeutic opportunities. Oncotarget. 2017;8:33972-33989.  [PubMed]  [DOI]  [Cited in This Article: ]
117.  de Assis JV, Coutinho LA, Oyeyemi IT, Oyeyemi OT, Grenfell RFEQ. Diagnostic and therapeutic biomarkers in colorectal cancer: a review. Am J Cancer Res. 2022;12:661-680.  [PubMed]  [DOI]  [Cited in This Article: ]
118.  Moon JH, Hong SW, Kim JE, Shin JS, Kim JS, Jung SA, Ha SH, Lee S, Kim J, Lee DH, Park YS, Kim DM, Park SS, Hong JK, Kim DY, Kim EH, Jung J, Kim MJ, Kim SM, Deming DA, Kim K, Kim TW, Jin DH. Targeting β-catenin overcomes MEK inhibition resistance in colon cancer with KRAS and PIK3CA mutations. Br J Cancer. 2019;120:941-951.  [PubMed]  [DOI]  [Cited in This Article: ]
119.  Haseeb M, Pirzada RH, Ain QU, Choi S. Wnt Signaling in the Regulation of Immune Cell and Cancer Therapeutics. Cells. 2019;8.  [PubMed]  [DOI]  [Cited in This Article: ]
120.  Li X, Xiang Y, Li F, Yin C, Li B, Ke X. WNT/β-Catenin Signaling Pathway Regulating T Cell-Inflammation in the Tumor Microenvironment. Front Immunol. 2019;10:2293.  [PubMed]  [DOI]  [Cited in This Article: ]
121.  Hong Y, Manoharan I, Suryawanshi A, Majumdar T, Angus-Hill ML, Koni PA, Manicassamy B, Mellor AL, Munn DH, Manicassamy S. β-catenin promotes regulatory T-cell responses in tumors by inducing vitamin A metabolism in dendritic cells. Cancer Res. 2015;75:656-665.  [PubMed]  [DOI]  [Cited in This Article: ]
122.  Luke JJ, Bao R, Sweis RF, Spranger S, Gajewski TF. WNT/β-catenin Pathway Activation Correlates with Immune Exclusion across Human Cancers. Clin Cancer Res. 2019;25:3074-3083.  [PubMed]  [DOI]  [Cited in This Article: ]
123.  Spranger S, Bao R, Gajewski TF. Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature. 2015;523:231-235.  [PubMed]  [DOI]  [Cited in This Article: ]
124.  Lal N, White BS, Goussous G, Pickles O, Mason MJ, Beggs AD, Taniere P, Willcox BE, Guinney J, Middleton GW. KRAS Mutation and Consensus Molecular Subtypes 2 and 3 Are Independently Associated with Reduced Immune Infiltration and Reactivity in Colorectal Cancer. Clin Cancer Res. 2018;24:224-233.  [PubMed]  [DOI]  [Cited in This Article: ]
125.  Grasso CS, Giannakis M, Wells DK, Hamada T, Mu XJ, Quist M, Nowak JA, Nishihara R, Qian ZR, Inamura K, Morikawa T, Nosho K, Abril-Rodriguez G, Connolly C, Escuin-Ordinas H, Geybels MS, Grady WM, Hsu L, Hu-Lieskovan S, Huyghe JR, Kim YJ, Krystofinski P, Leiserson MDM, Montoya DJ, Nadel BB, Pellegrini M, Pritchard CC, Puig-Saus C, Quist EH, Raphael BJ, Salipante SJ, Shin DS, Shinbrot E, Shirts B, Shukla S, Stanford JL, Sun W, Tsoi J, Upfill-Brown A, Wheeler DA, Wu CJ, Yu M, Zaidi SH, Zaretsky JM, Gabriel SB, Lander ES, Garraway LA, Hudson TJ, Fuchs CS, Ribas A, Ogino S, Peters U. Genetic Mechanisms of Immune Evasion in Colorectal Cancer. Cancer Discov. 2018;8:730-749.  [PubMed]  [DOI]  [Cited in This Article: ]
126.  Pai SG, Carneiro BA, Mota JM, Costa R, Leite CA, Barroso-Sousa R, Kaplan JB, Chae YK, Giles FJ. Wnt/beta-catenin pathway: modulating anticancer immune response. J Hematol Oncol. 2017;10:101.  [PubMed]  [DOI]  [Cited in This Article: ]
127.  Payandeh Z, Khalili S, Somi MH, Mard-Soltani M, Baghbanzadeh A, Hajiasgharzadeh K, Samadi N, Baradaran B. PD-1/PD-L1-dependent immune response in colorectal cancer. J Cell Physiol. 2020;235:5461-5475.  [PubMed]  [DOI]  [Cited in This Article: ]
128.  Spranger S, Dai D, Horton B, Gajewski TF. Tumor-Residing Batf3 Dendritic Cells Are Required for Effector T Cell Trafficking and Adoptive T Cell Therapy. Cancer Cell. 2017;31:711-723.e4.  [PubMed]  [DOI]  [Cited in This Article: ]
129.  Rimassa L, Bozzarelli S, Pietrantonio F, Cordio S, Lonardi S, Toppo L, Zaniboni A, Bordonaro R, Di Bartolomeo M, Tomasello G, Dadduzio V, Tronconi MC, Piombo C, Giordano L, Gloghini A, Di Tommaso L, Santoro A. Phase II Study of Tivantinib and Cetuximab in Patients With KRAS Wild-type Metastatic Colorectal Cancer With Acquired Resistance to EGFR Inhibitors and Emergence of MET Overexpression: Lesson Learned for Future Trials With EGFR/MET Dual Inhibition. Clin Colorectal Cancer. 2019;18:125-132.e2.  [PubMed]  [DOI]  [Cited in This Article: ]
130.  Wu ZX, Yang Y, Teng QX, Wang JQ, Lei ZN, Lusvarghi S, Ambudkar SV, Yang DH, Chen ZS. Tivantinib, A c-Met Inhibitor in Clinical Trials, Is Susceptible to ABCG2-Mediated Drug Resistance. Cancers (Basel). 2020;12.  [PubMed]  [DOI]  [Cited in This Article: ]
131.  Meyerhardt JA, Shi Q, Fuchs CS, Meyer J, Niedzwiecki D, Zemla T, Kumthekar P, Guthrie KA, Couture F, Kuebler P, Bendell JC, Kumar P, Lewis D, Tan B, Bertagnolli M, Grothey A, Hochster HS, Goldberg RM, Venook A, Blanke C, O'Reilly EM, Shields AF. Effect of Celecoxib vs Placebo Added to Standard Adjuvant Therapy on Disease-Free Survival Among Patients With Stage III Colon Cancer: The CALGB/SWOG 80702 (Alliance) Randomized Clinical Trial. JAMA. 2021;325:1277-1286.  [PubMed]  [DOI]  [Cited in This Article: ]
132.  Turano M, Cammarota F, Duraturo F, Izzo P, De Rosa M. A Potential Role of IL-6/IL-6R in the Development and Management of Colon Cancer. Membranes (Basel). 2021;11.  [PubMed]  [DOI]  [Cited in This Article: ]
133.  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.  [PubMed]  [DOI]  [Cited in This Article: ]
134.  Vaghari-Tabari M, Majidinia M, Moein S, Qujeq D, Asemi Z, Alemi F, Mohamadzadeh R, Targhazeh N, Safa A, Yousefi B. MicroRNAs and colorectal cancer chemoresistance: New solution for old problem. Life Sci. 2020;259:118255.  [PubMed]  [DOI]  [Cited in This Article: ]
135.  Azwar S, Seow HF, Abdullah M, Faisal Jabar M, Mohtarrudin N. Recent Updates on Mechanisms of Resistance to 5-Fluorouracil and Reversal Strategies in Colon Cancer Treatment. Biology (Basel). 2021;10.  [PubMed]  [DOI]  [Cited in This Article: ]
136.  van Meeteren LA, Thorikay M, Bergqvist S, Pardali E, Stampino CG, Hu-Lowe D, Goumans MJ, ten Dijke P. Anti-human activin receptor-like kinase 1 (ALK1) antibody attenuates bone morphogenetic protein 9 (BMP9)-induced ALK1 signaling and interferes with endothelial cell sprouting. J Biol Chem. 2012;287:18551-18561.  [PubMed]  [DOI]  [Cited in This Article: ]
137.  Panda PK, Naik PP, Praharaj PP, Meher BR, Gupta PK, Verma RS, Maiti TK, Shanmugam MK, Chinnathambi A, Alharbi SA, Sethi G, Agarwal R, Bhutia SK. Abrus agglutinin stimulates BMP-2-dependent differentiation through autophagic degradation of β-catenin in colon cancer stem cells. Mol Carcinog. 2018;57:664-677.  [PubMed]  [DOI]  [Cited in This Article: ]
138.  Kim BR, Oh SC, Lee DH, Kim JL, Lee SY, Kang MH, Lee SI, Kang S, Joung SY, Min BW. BMP-2 induces motility and invasiveness by promoting colon cancer stemness through STAT3 activation. Tumour Biol. 2015;36:9475-9486.  [PubMed]  [DOI]  [Cited in This Article: ]
139.  Todaro M, Gaggianesi M, Catalano V, Benfante A, Iovino F, Biffoni M, Apuzzo T, Sperduti I, Volpe S, Cocorullo G, Gulotta G, Dieli F, De Maria R, Stassi G. CD44v6 is a marker of constitutive and reprogrammed cancer stem cells driving colon cancer metastasis. Cell Stem Cell. 2014;14:342-356.  [PubMed]  [DOI]  [Cited in This Article: ]
140.  Mizuno R, Kawada K, Itatani Y, Ogawa R, Kiyasu Y, Sakai Y. The Role of Tumor-Associated Neutrophils in Colorectal Cancer. Int J Mol Sci. 2019;20.  [PubMed]  [DOI]  [Cited in This Article: ]
141.  Yahaya MAF, Lila MAM, Ismail S, Zainol M, Afizan NARNM. Tumour-Associated Macrophages (TAMs) in Colon Cancer and How to Reeducate Them. J Immunol Res. 2019;2019:2368249.  [PubMed]  [DOI]  [Cited in This Article: ]
142.  Wang S, Miao Z, Yang Q, Wang Y, Zhang J. The Dynamic Roles of Mesenchymal Stem Cells in Colon Cancer. Can J Gastroenterol Hepatol. 2018;2018:7628763.  [PubMed]  [DOI]  [Cited in This Article: ]
143.  Yu Z, Lou L, Zhao Y. Fibroblast growth factor 18 promotes the growth, migration and invasion of MDAMB231 cells. Oncol Rep. 2018;40:704-714.  [PubMed]  [DOI]  [Cited in This Article: ]
144.  Gonzalez-Zubeldia I, Dotor J, Redrado M, Bleau AM, Manrique I, de Aberasturi AL, Villalba M, Calvo A. Co-migration of colon cancer cells and CAFs induced by TGFβ₁ enhances liver metastasis. Cell Tissue Res. 2015;359:829-839.  [PubMed]  [DOI]  [Cited in This Article: ]
145.  Vinnakota K, Zhang Y, Selvanesan BC, Topi G, Salim T, Sand-Dejmek J, Jönsson G, Sjölander A. M2-like macrophages induce colon cancer cell invasion via matrix metalloproteinases. J Cell Physiol. 2017;232:3468-3480.  [PubMed]  [DOI]  [Cited in This Article: ]
146.  Wu D, Ding Y, Wang T, Cui P, Huang L, Min Z, Xu M. Significance of Tumor-Infiltrating Immune Cells in the Prognosis of Colon Cancer. Onco Targets Ther. 2020;13:4581-4589.  [PubMed]  [DOI]  [Cited in This Article: ]
147.  Silva DA, Yu S, Ulge UY, Spangler JB, Jude KM, Labão-Almeida C, Ali LR, Quijano-Rubio A, Ruterbusch M, Leung I, Biary T, Crowley SJ, Marcos E, Walkey CD, Weitzner BD, Pardo-Avila F, Castellanos J, Carter L, Stewart L, Riddell SR, Pepper M, Bernardes GJL, Dougan M, Garcia KC, Baker D. De novo design of potent and selective mimics of IL-2 and IL-15. Nature. 2019;565:186-191.  [PubMed]  [DOI]  [Cited in This Article: ]
148.  Fisher RC, Bellamkonda K, Alex Molina L, Xiang S, Liska D, Sarvestani SK, Chakrabarti S, Berg A, Jorgensen ML, Hatala D, Chen S, Aiello A, Appelman HD, Scott EW, Huang EH. Disrupting Inflammation-Associated CXCL8-CXCR1 Signaling Inhibits Tumorigenicity Initiated by Sporadic- and Colitis-Colon Cancer Stem Cells. Neoplasia. 2019;21:269-281.  [PubMed]  [DOI]  [Cited in This Article: ]
149.  Wang X, Yang L, Huang F, Zhang Q, Liu S, Ma L, You Z. Inflammatory cytokines IL-17 and TNF-α up-regulate PD-L1 expression in human prostate and colon cancer cells. Immunol Lett. 2017;184:7-14.  [PubMed]  [DOI]  [Cited in This Article: ]
150.  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. Dis Markers. 2020;2020:6439614.  [PubMed]  [DOI]  [Cited in This Article: ]
151.  Pastille E, Wasmer MH, Adamczyk A, Vu VP, Mager LF, Phuong NNT, Palmieri V, Simillion C, Hansen W, Kasper S, Schuler M, Muggli B, McCoy KD, Buer J, Zlobec I, Westendorf AM, Krebs P. The IL-33/ST2 pathway shapes the regulatory T cell phenotype to promote intestinal cancer. Mucosal Immunol. 2019;12:990-1003.  [PubMed]  [DOI]  [Cited in This Article: ]
152.  De la Fuente López M, Landskron G, Parada D, Dubois-Camacho K, Simian D, Martinez M, Romero D, Roa JC, Chahuán I, Gutiérrez R, Lopez-K F, Alvarez K, Kronberg U, López S, Sanguinetti A, Moreno N, Abedrapo M, González MJ, Quera R, Hermoso-R MA. The relationship between chemokines CCL2, CCL3, and CCL4 with the tumor microenvironment and tumor-associated macrophage markers in colorectal cancer. Tumour Biol. 2018;40:1010428318810059.  [PubMed]  [DOI]  [Cited in This Article: ]
153.  Halama N, Zoernig I, Berthel A, Kahlert C, Klupp F, Suarez-Carmona M, Suetterlin T, Brand K, Krauss J, Lasitschka F, Lerchl T, Luckner-Minden C, Ulrich A, Koch M, Weitz J, Schneider M, Buechler MW, Zitvogel L, Herrmann T, Benner A, Kunz C, Luecke S, Springfeld C, Grabe N, Falk CS, Jaeger D. Tumoral Immune Cell Exploitation in Colorectal Cancer Metastases Can Be Targeted Effectively by Anti-CCR5 Therapy in Cancer Patients. Cancer Cell. 2016;29:587-601.  [PubMed]  [DOI]  [Cited in This Article: ]
154.  Noh GT, Kwon J, Kim J, Park M, Choi DW, Cho KA, Woo SY, Oh BY, Lee KY, Lee RA. Verification of the role of exosomal microRNA in colorectal tumorigenesis using human colorectal cancer cell lines. PLoS One. 2020;15:e0242057.  [PubMed]  [DOI]  [Cited in This Article: ]
155.  Ma J, Sun X, Wang Y, Chen B, Qian L. Fibroblast-derived CXCL12 regulates PTEN expression and is associated with the proliferation and invasion of colon cancer cells via PI3k/Akt signaling. Cell Commun Signal. 2019;17:119.  [PubMed]  [DOI]  [Cited in This Article: ]
156.  Calvo N, Martín MJ, de Boland AR, Gentili C. Involvement of ERK1/2, p38 MAPK, and PI3K/Akt signaling pathways in the regulation of cell cycle progression by PTHrP in colon adenocarcinoma cells. Biochem Cell Biol. 2014;92:305-315.  [PubMed]  [DOI]  [Cited in This Article: ]
157.  Calvo N, Carriere P, Martin MJ, Gentili C. RSK activation via ERK modulates human colon cancer cells response to PTHrP. J Mol Endocrinol. 2017;59:13-27.  [PubMed]  [DOI]  [Cited in This Article: ]
158.  Meurette O, Mehlen P. Notch Signaling in the Tumor Microenvironment. Cancer Cell. 2018;34:536-548.  [PubMed]  [DOI]  [Cited in This Article: ]
159.  Al-Haidari A, Algaber A, Madhi R, Syk I, Thorlacius H. MiR-155-5p controls colon cancer cell migration via post-transcriptional regulation of Human Antigen R (HuR). Cancer Lett. 2018;421:145-151.  [PubMed]  [DOI]  [Cited in This Article: ]
160.  Mussnich P, Rosa R, Bianco R, Fusco A, D'Angelo D. MiR-199a-5p and miR-375 affect colon cancer cell sensitivity to cetuximab by targeting PHLPP1. Expert Opin Ther Targets. 2015;19:1017-1026.  [PubMed]  [DOI]  [Cited in This Article: ]