Next Article in Journal
Telemedical Consultations in Palliative Care: Benefits through Knowledge Exchange and Intercollegiate Collaboration—Findings from the German oVID Project
Next Article in Special Issue
Significance of Cancer-Associated Fibroblasts in the Interactions of Cancer Cells with the Tumor Microenvironment of Heterogeneous Tumor Tissue
Previous Article in Journal
Thyroid Hormone Withdrawal versus Recombinant Human TSH as Preparation for I-131 Therapy in Patients with Metastatic Thyroid Cancer: A Systematic Review and Meta-Analysis
Previous Article in Special Issue
Cancer-Associated Fibroblasts: Master Tumor Microenvironment Modifiers
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Reciprocal Regulation of Cancer-Associated Fibroblasts and Tumor Microenvironment in Gastrointestinal Cancer: Implications for Cancer Dormancy

by
Shih-Hsuan Cheng
1,2,
Hsin-Ying Clair Chiou
3,4,
Jiunn-Wei Wang
2,5,6,7 and
Ming-Hong Lin
1,7,8,9,10,*
1
Department of Microbiology and Immunology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
2
Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
3
Teaching and Research Center, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung 812, Taiwan
4
Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
5
Department of Internal Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
6
Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
7
Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
8
Department of Post Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung City 807, Taiwan
9
Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
10
Master of Science Program in Tropical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(9), 2513; https://doi.org/10.3390/cancers15092513
Submission received: 3 March 2023 / Revised: 24 April 2023 / Accepted: 26 April 2023 / Published: 27 April 2023
(This article belongs to the Special Issue Crosstalk between Cancer-Associated Fibroblasts and Cancer Cells)

Abstract

:

Simple Summary

Gastrointestinal (GI) cancers are the leading cancer-related deaths worldwide. Despite improved survival rates with current treatments, recurrence remains a clinical challenge for patients with GI cancers. The unmet gap still needs to be further investigated. The tumor microenvironment (TME) is associated with the dormancy and awakening of tumor cells. Among them, the reciprocal regulation of cytokines/chemokines between cancer-associated fibroblasts (CAFs) and TME is a key driver of tumor progression, metastasis, and resistance to chemotherapy. In this review, we discuss the involvement of cytokines/chemokines produced by CAFs in the development of resistance to gastrointestinal cancer therapy, including tumor dormancy and tumor regeneration. Deciphering the crosstalk of CAFs and TME thus reduces the therapeutic recurrence of GI cancers.

Abstract

Gastrointestinal (GI) cancers remain a major cause of cancer-related deaths worldwide. Despite the progress made in current treatments, patients with GI cancers still have high recurrence rates after initial treatment. Cancer dormancy, which involves the entry and escape of cancer cells from dormancy, is linked to treatment resistance, metastasis, and disease relapse. Recently, the role of the tumor microenvironment (TME) in disease progression and treatment has received increasing attention. The crosstalk between cancer-associated fibroblasts (CAF)-secreted cytokines/chemokines and other TME components, for example, extracellular matrix remodeling and immunomodulatory functions, play crucial roles in tumorigenesis. While there is limited direct evidence of a relationship between CAFs and cancer cell dormancy, this review explores the potential of CAF-secreted cytokines/chemokines to either promote cancer cell dormancy or awaken dormant cancer cells under different conditions, and the therapeutic strategies that may be applicable. By understanding the interactions between cytokines/chemokines released by CAFs and the TME, and their impact on the entry/escape of cancer dormancy, researchers may develop new strategies to reduce the risk of therapeutic relapse in patients with GI cancers.

1. Introduction

Gastrointestinal (GI) cancer is a malignant disease that affects the gastrointestinal system, including the esophagus, liver, stomach, gallbladder and bile ducts, pancreas, gastrointestinal interstitial tumor (GIST), and colon. Despite advances in the treatment of GI cancers, they still account for 35% of all cancer-related deaths, posing a major threat to public health [1]. One of the most pressing challenges in treating GI cancers is the resistance of cancer cells to therapy.
The tumor microenvironment (TME) is a complex environment consisting of immune cells, cancer-associated fibroblasts (CAFs), the extracellular matrix (ECM), cytokines, chemokines, and growth factors, which play important roles in tumorigenesis, metastasis, and cancer cell resistance to therapy [2]. From among them, CAFs are the essential components of the TME in GI cancers as they can trigger the resistance of cancer cells to treatment [3].
Cancer recurrence can occur shortly after treatment or several years after the initial diagnosis or treatment, indicating that the cancer cells have been reactivated and are proliferating (also known as awakening) after a long period of latency (also known as dormancy) [4]. Cancer cell dormancy is an adaptive strategy that allows cancer cells to survive in disadvantageous conditions that they must deal with [5]. Dormant cancer cells (DCCs) may reactivate due to the crosstalk between cytokines/chemokines secreted by CAFs and other components of the TME, including the remodeling of the ECM and immunomodulatory functions.
Patients with gastrointestinal (GI) cancers have been experiencing shorter recurrence periods compared to certain other cancer types, such as breast cancer [6,7,8,9], mainly because of the insidious nature of their symptoms and the absence of effective early detection screening. Consequently, many cases are diagnosed at advanced stages, which results in fewer treatment options and a higher probability of developing drug resistance, with the alterations in the TME being a critical factor that contributes to the development of resistance in GI cancers [10,11]. In this review, we discuss the role of cytokines/chemokines secreted by CAFs in the TME of GI cancers and the generation of therapeutic resistance through the mechanism of cancer dormancy. Notably, the reciprocal regulation between CAF-derived cytokines/chemokines and the TME is not unique to GI tumors, and the underlying mechanisms may differ from those in other solid tumors due to the high heterogeneity of CAFs. Deshka et al. showed that CAF characteristics vary across solid tumor types and species. Nevertheless, CAFs can be classified into three superclusters: steady-state-like (SSL), mechanoresponsive (MR), and immunomodulatory (IM), using paired, same-cell chromatin accessibility and transcriptome analysis [12]. Additionally, Galbo et al. found that different CAF isoforms may activate their own unique molecular pathways to regulate the tumor microenvironment in various cancer types [13].
As direct evidence of the relationship between CAFs and cancer cell dormancy is limited, we have summarized the potential of cytokines/chemokines secreted by CAFs to induce or awaken DCCs under different conditions and the agents that may be applicable to resist it. Deciphering the crosstalk between CAFs and the TME in relation to the mechanisms of cancer cell dormancy may provide insightful strategies to further prevent and treat GI cancer recurrence. Our review aims to provide a better understanding of the role of CAFs in GI cancers, and to highlight potential therapeutic targets that could reduce the risk of recurrence in patients with GI cancers.

2. Reciprocal Regulation of CAF-Secreted Cytokines/Chemokines and the Gastrointestinal Tumor Microenvironment

The GI TME is a dynamic environment, dominated by CAFs through direct contact and indirect paracrine effects on the tumor–stroma crosstalk, ECM remodeling, and immune cells [14,15]. The multi-regulatory functions of cytokines/chemokines secreted by CAFs in the TME are depicted in Figure 1.

2.1. Crosstalk between Tumor and CAFs Contributes to Tumor Progression

Tumor cells set the conditions in the microenvironment and then take advantage of it, while CAFs maintain the microenvironment and further promote tumorigenesis [16]. The transforming growth factor beta (TGF-β) is a cytokine required for the induction of fibrotic responses and the activation of the tumor stroma, and it subsequently induces the formation of CAF phenotypes in the TME [17,18]. After fibroblast activation, TGF-β is secreted by CAFs and facilitates the further development of tumor cells [19]. Similarly, other factors released by tumor cells, such as the platelet-derived growth factor (PDGF) and fibroblast growth factor 2 (FGF-2), or inflammatory signals such as interleukin (IL)-1, IL-6 and tumor necrosis factor alpha (TNF-α), recruit and proliferate additional CAFs. Therefore, the activated CAFs shape a pro-tumorigenic TME through a variety of functions, which facilitates tumor progression [20].
Moreover, CXC ligand 12 (CXCL12, also called stromal cell-derived factor-1, SDF-1), which is produced by stromal cells and binds to C-X-C chemokine receptor type 4 (CXCR4) on the surface of tumor cells, has been reported to be essential for cancer pathogenesis. In gastric cancer (GC), the high expression of CXCL12 by CAFs may influence the malignancy of tumor cells [21]. More specifically, in a study of pancreatic cancer, tumor cells harboring the mutant KRASG12D communicated with CAFs via sonic hedgehog signaling, which stimulated the production of insulin-like growth factor 1 (IGF-1) in CAFs; in turn, reciprocal signaling affected tumor cell proliferation [22].
Recent studies have reported that CAFs can sense tumor cell genomic stress by expressing interferon (IFN)-β1, which leads to the increased production of associated factors such as CXC ligand 1 (CXCL1) and CXC ligand 10 (CXCL10), IL-6, IL-8, and IL-11 [23]. Among them, IL-6 and IL-8 are considered as indicators and therapeutic targets in GI-related cancers [24,25,26,27]. For example, CAF-secreted IL-6 enhances tumor cells’ epithelial–mesenchymal transition (EMT) via the activation of the Janus kinase 2/signal transducers and activators of the transcription 3 (JAK2/STAT3) pathway in GC [28]. Activation of STAT3 by CAF-derived IL-6/IL-11 promotes the development of colorectal tumors and is associated with a poor prognosis [29,30]. Furthermore, a meta-analysis including 12 studies showed that high IL-8 levels were significantly associated with shorter overall survival and progression-free survival in colorectal cancer (CRC) patients [26]. In summary, there is a bidirectional crosstalk between CAFs and tumor cells that shapes a dynamic TME through the release of cytokines/chemokines, thus aggravating the progression of GI cancers.

2.2. CAF-Mediated ECM Remodeling in Tumor Progression

The dysregulation of ECM homeostasis is associated with cancer progression and drug resistance [31]. CAF-derived cytokines/chemokines have an impact on ECM structure and function, and all of them are involved in tumor-promoting and metastasis-accelerating ECM remodeling. CAF-derived TGF-β upregulates matrix metalloproteinase 9 (MMP-9), which remodels ECM components and creates a favorable environment for tumor progression and metastasis [32]. Another enzyme regulated by TGF-β is lysyl oxidase (LOX), which cross-links collagen and elastin fibers. This leads to ECM stiffness and further feedback to TGF-β induction and contributes to a tumor-promoting inflammatory environment [33,34]. CAFs also secrete a disintegrin and metalloproteinase 9 (ADAM9), which has been implicated in increasing the proliferation and dissemination of CRC cells [35]. Recent literature suggests that ADAMs are considered biochemical markers of GI tumorigenesis due to their protein hydrolysis and adhesion activities [36,37]. Overall, CAF-derived cytokines/chemokines play a significant role in ECM remodeling, further contributing to tumor progression and metastasis.

2.3. Crosstalk between CAF-Derived Factors and Immune Cells in the TME

CAF-derived cytokines/chemokines play a critical role in modulating the immune response and are associated with immunosuppressive TME [38].
CAFs secrete various cytokines/chemokines that promote the recruitment and polarization of immune inhibitory cells, such as tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), and regulatory T cells. For example, monocyte chemotactic protein-1 (MCP-1), IL-6, CXCL12, and granulocyte-macrophage colony-stimulating factor (GM-CSF) promote the recruitment and polarization of these immune inhibitory cells [39,40,41]. In addition, CAFs secrete CXCL2 [42] and CXCL5 [43] to promote the PD-L1 expression of cancer cells via phosphatidylinositol-3-kinase (PI3K)/AKT signaling, which further facilitates immune deviation toward tolerance.
Moreover, CAFs also secrete TGF-β, indoleamine 2,3-dioxygenase (IDO), and prostaglandin E2 (PGE-2), which interfere with the activation of natural killer cells and thus suppress innate anti-tumor immunity [44,45]. CAFs secrete IL-6, CXCL12, and vascular endothelial growth factor (VEGF), which combat the activity of cytotoxic T lymphocytes and the maturation of dendritic cells (DCs) [46].
Understanding the mechanisms of CAF-induced immunosuppression and their interactions with immune cells within the TME can aid in identifying potential molecular targets for CAF-targeted therapy. Further exploration of these interactions may offer new therapeutic strategies to enhance the anti-tumor immune response in the TME.

3. CAF-Secreted Cytokines/Chemokines Inducing Cancer Cell Dormancy

Cancer cell dormancy refers to the state in which cancer cells survive through slow proliferation or slow cycling [47]. DCCs themselves activate mechanisms such as autophagy or immune evasion, which is the result of their interaction with the TME. In other words, dormancy can be considered an adaptive response to microenvironmental stress [48], where CAF-secreted cytokines/chemokines may influence DCCs directly or indirectly by interaction with the TME (Figure 2). The activation of the extracellular signal-regulated kinase (ERK) pathway has been demonstrated to facilitate G0-G1-S-phase transition and promote cell division. In contrast, high levels of p38 activity have been implicated as negative regulators of growth by inhibiting ERK, inducing G0/G1 arrest, or triggering apoptosis. As a result, the ratio between p38 signaling and the ERK determines whether tumor cells proliferate or enter a dormant state [49,50].
CAFs are one of the sources of TGF-β2, which has been implicated in the maintenance of cancer cell dormancy [51]. TGF-β2 induces SMAD1/2/5 to upregulate p27 via TGFβ-RIII, which in turn activates p38 to promote dormancy [52]. In addition, Wu et al. reported that TGF-β2 signaling decreased the proliferation of both CAFs and tumor cells by activating the G1/S checkpoint in CRC [53]. In addition, an elevated production of TGF-β2 by CAFs was observed when heat shock factor (HSF)-1 was activated [54]. HSF-1 is a transcription factor that responds to a state of cellular hypoxia and is associated with dormancy through the induction of cell cycle arrest and the maintenance of cell survival through the activation of autophagy [55,56]. Moreover, CAFs secrete growth differentiation factor 10 (GDF-10) [57], which promotes cell cycle arrest by binding to the TGF-βRIII receptor and then activating the p38MAPK signaling pathway [58]. Furthermore, Shirai et al. found that bone morphogenetic protein 4 (BMP-4) induced G1 arrest in diffuse gastric cancer cells via SMAD-induced p21 [59]. Similarly, Yokoyama et al. reported that the autocrine signaling of BMP-4 may be a therapeutic target in CRC [60]. In summary, in the GI cancer TME, those CAF-secreted factors mentioned above promote or maintain cancer cell dormancy by upregulating the p38 pathway and/or inducing cell cycle arrest.
Recently, Owen et al. revealed a key role of intrinsic interferon (IFN) signaling in maintaining the dormant state of tumor cells [61]. IFN-β secreted by CAFs drives the sustained activation of the type I interferon receptor/interferon regulatory factor 7 pathway, triggering the chemotherapy-induced dormancy of breast cancer cells [62]. In line with this, Guo et al. suggested that IRF7 is associated with immune cell infiltration in gastric adenocarcinoma, which serves as a prognostic biomarker [63]. Therefore, IFN-β may play a role in inducing cancer cell dormancy in the GI microenvironment. Additionally, CAFs can produce IFN-γ, which activates STAT1 and thus induces cells to enter G0/G1 growth arrest by reducing the cyclin D1/CDK4 complex [64]. On the other hand, IFN-γ can mediate cancer cell dormancy via the indoleamine-2,3-dioxygenase/kynurenine/aryl hydrocarbon receptor (AhR)/p27 pathway in a STAT1-independent manner [65]. Taken together, it is suggested that CAF-secreted IFN-γ is critical for the induction of cancer cell dormancy.
The Wnt pathway, an inducer of proliferation, is involved in the reactivation of DCCs. Dickkopf-related protein 1 (DKK-1), which is secreted by CAFs, is an inhibitor of Wnt signaling, and its expression has been reported to be a prognostic factor in the prediction of tumor recurrence and survival in patients with advanced GC [66]. Malladi et al. found that isolated latency-competent cancer cells from early-stage human lung and breast cancers enter a dormant state by expressing a Sox-dependent stemness state and the Wnt inhibitor DKK1 [67].
Another feature of DCCs is the activation of autophagy in response to different forms of stress and the promotion of cancer cell survival during growth arrest, eventually making autophagy integral to the survival of DCCs. Therefore, it is essential to maintain autophagic flux in cancer cells that are in a dormant state [68,69]. In both in vitro and in vivo research models, CAFs produce IGF-1/IGF-2 to induce autophagic dormancy, promoting cancer cell recovery from radiation-induced damage [70] and protecting cells from chemotherapeutic stress. In addition, Rajbhandari et al. reported that autocrine IGF-1 signaling mediated the dormancy of pancreatic tumor cells in the absence of oncogenic drivers [71]. Lu et al. reported that IL-6 induces autophagy through the expression of NS5ATP9, which upregulates IL-6 levels and further induces autophagy [72].
Finally, the balance of pro- and anti-angiogenic factors produced by CAFs, such as VEGF for the former and thrombospondin-1 (TSP-1) for the latter, is also involved in the regulation of cancer cell dormancy [73]. The loss of TSP-1 has been shown to promote escape from cancer dormancy [74,75], in which higher levels of TSP-1 in the lesion tissues of patients with metastatic CRC have been reported to result in the remission of mortality [76].

4. CAF-Secreted Cytokines/Chemokines Awakening Cancer Cell Dormancy

Dormancy escape can be stimulated by evolving interactions with TME components, with CAF-driven cytokines/chemokines being one of the main driving forces [47]. Awakening is the process of forcing DCCs back into the cell cycle, shifting the imbalance from a non-proliferating state of p38 MAPK induction to a proliferating state of ERK upregulation [77].
The CAF-secreted urokinase-type plasminogen activator (uPA) binds to its receptor (uPAR), activating the integrin and epidermal growth factor receptor (EGFR) pathways and resulting in the subsequent activation of ERK and reduction of p38 activity, which promotes tumor cell proliferation [78]. Likewise, the inhibition of uPAR-activated FAK signaling leads to low levels of ERK and induces cancer cell dormancy. Taken together, the uPA/uPAR axis is critical in the awakening of DCCs [79].
In addition to the ERK pathway, the previously mentioned Wnt signaling is an indicator of cell proliferation, and it also reactivates DCCs [80]. Tenascin C is known to be initially produced by initiated metastatic tumor cells and later secreted by CAFs to support the growth of metastatic cells by promoting the Wnt signaling pathway; this appears to reflect the awakening of DCCs [81]. In this regard, the knockdown of Tenascin C inhibits ERK signaling and thereby inhibits the metastasis of gastric cancer. Furthermore, studies have reported that CAFs secrete periostin and hepatocyte growth factor (HGF), which promote the recruitment of Wnt ligands and thus increase Wnt presentation in cancer cells, including colon and gastric cancer [82,83,84], suggesting that the reawakening and metastasis of dormant cancer cells are likewise promoted [85]. Many studies on gastrointestinal cancers have shown that the upregulation of periostin and HGF is associated with increasing cancer cell malignancy, the elevating of metastatic capacity, and worsening patient prognosis [86,87,88]; thus, it can be said that both periostin and HGF allow dormant cancer cells to reawaken and metastasize. In addition, galectin-1 (Gal-1) secreted by CAFs binds to integrin β1, leading to the activation of the Wnt/β-catenin signaling pathway associated with the metastatic process in GC cells [89,90].
Chronic inflammation is known to promote the development and progression of cancer [91] and is now also considered to be a major driver in the awakening of dormant cancer cells [92]. During chronic inflammation, the TME is enriched with inflammatory factors secreted by tumor cells, CAFs, and immune cells, such as IL-6, IL-8, IL-1β, IL-10, and IL-17, creating a microenvironment conducive to dormancy escape as well as reactivating cancer cell expansion [92]. For example, Khazali et al. demonstrated that IL-8 can mediate cancer cell dormancy escape using an in vitro 3D liver microphysiological system [93]. Furthermore, IL-17 secreted by CAFs upregulates JAK2/STAT3 signaling, which promotes gastric cancer cell malignancy [94]. Similarly, IL-17 has been shown to promote the transformation of dormant gastric cancer stem cells [95]. Cyclooxygenase 2 (COX-2) is a pro-inflammatory mediator and a key mediator of inflammatory pathways that awaken DCCs [96,97]. COX-2 is also abundant in the TME of GI cancers [98], where CAFs are a major source of COX-2 [99].
Regarding IL-6, as described in the article above, it is secreted by CAFs, which promote cancer cell dormancy by inducing autophagy. Nevertheless, most studies have concluded that IL-6 is a key player in the inflammatory process, serving as a pro-inflammatory and pro-metastatic factor and thus promoting the escape of DCCs [100]. For example, a study by Shaashua et al. showed that spontaneous micrometastatic cells returned to a dormant state after IL-6 was removed during a minimally invasive resectioning of the primary tumor, highlighting the role of IL-6 in dormancy escape [101]. Furthermore, Esposito et al. showed that IL-6 disrupts the dormant phenotype mediated by the tumor suppressor aplasia Ras homolog member (ARH-I) by upregulating an onco-miRNA miR-1305 [102]. In sum, IL-6 plays a role in controlling cancer cell dormancy or awakening, depending on the interaction between the stage of the tumor cell progression and the TME.
CAFs secrete chemokines, which act as mediators of inflammation and influence the entry/escape of dormancy. As mentioned earlier, the interaction between immune and tumor cells in the TME has important implications for tumor development and treatment, and the chemokines secreted by CAFs act as activators for this [34]. Romero-Moreno et al. used in vitro co-culture to simulate the final stage of metastasis of dormant breast cancer cells to the bone. They found that the increase in CXCL5 at the metastatic site created a favorable and supportive microenvironment that promoted the reactivation and recolonization of DCCs [103]. Similarly, CXCL5 and its receptor, CXCR2, have been shown to be associated with metastasis in many gastrointestinal cancer models [104,105] and may be potential therapeutic targets for TME [106,107]. Adamski et al. showed that CXCL12, CXCL16, and CX3CL1 result in the delayed entry and exit of GBM cells from temozolomide-induced cellular dormancy. In other words, CXCL12, CXCL16, and CX3CL1 facilitate the escape of cells from the dormant state [108]. Furthermore, there is evidence that CCXR4 is absent or low in dormant gastric cancer cells [95] or metastatic breast cancer cells [109]. On the contrary, the increased expression of CXCL12, a ligand for CXCR4, facilitates the escape of cancer cells from dormancy. Similarly, Peng et al. showed that the activation of TGF-β via integrin αvβ6 further stimulates CXCL12 secretion by CAFs, leading to CRC invasion via the CXCL12/CXCR4-dependent axis [110].
Notably, some CAFs release factors that play opposite roles in the TME, either by mediating dormancy or by reversing it; in addition to IL-6, these also include TGF-β. In contrast to the ability of TGF-β2 to promote dormancy, TGF-β1 has been shown in several studies to be associated with tumor cell proliferation [111] and the awakening of dormant tumor cells [112]. For example, in sprouting neovascularization, TGF-β1 blocked the quiescent effects of TSP-1 and promoted micrometastasis [112]. Interestingly, the TGF-β1 ligand antagonist Coco was shown to reactivate dormant breast cancer cells in the lung, but not in the bone or brain, suggesting the organ-specific regulation of cancer cell dormancy [113]. Similarly, CAF-secreted IGF-2 has a dual role in dormancy entry/escape. In contrast to the ability of IGF-2 to promote dormancy through the induction of autophagy, CAFs produce high levels of IGF-2, which is thought to promote distant metastasis by stimulating the formation of a pre-metastatic ecotone [114]. Furthermore, in an esophageal cancer model, tumor-derived IGF-2 induces CAFs to produce VEGF, creating a general environment for metastasis by stimulating the formation of a pre-metastatic ecotone [115]. It was thus concluded that IGF-2 plays a role in dormancy escape.
Another dominant ability of CAFs to drive dormancy escape is their ability to synthesize and remodel ECM, further affecting the balance between ECM-mediated cell dormancy and proliferation [116]. For example, CAFs assemble the ECM protein fibronectin (FN), which promotes dormancy escape by inhibiting the p38 signaling pathway and activating ERK [117,118]. In detail, CAFs secrete MMP-2, MMP-9, and type I collagen, which are important processes that promote cellular dormancy escape by inducing cytoskeletal reorganization and β1/β4 integrin signaling [52,119,120]. In addition, CAFs secrete LOX and lysyl oxidase-like (LOXL2), which are involved in collagen deposition. This determines the matrix stiffness and influences tumor cell adhesion to the ECM, thus allowing the transition from a dormant to a proliferative state [121,122,123]. Moreover, CAF-secreted Gremlin 1 (GREM1), a BMP antagonist, was found to be upregulated in GI cancers [124]. GREM1 has also been detected in the demyelinating invasion front of CRC, suggesting a potential role in cancer metastasis [124,125]. In addition, GREM1 enables dormant breast cancer cells to escape the quiescence imposed by BMP signaling and thus initiate metastasis [113].
Furthermore, other cell types in TME are involved in ECM remodeling. For example, Albrengues et al. discovered that, in an inflammatory state, neutrophil elastase and MMP-9, two neutrophil extracellular trap (NET) proteases, can sequentially cleave laminin (LN), which can result in the activation of integrin α3β1 signaling and the proliferation of dormant cancer cells. The proteolytic remodeling of LN can be prevented by blocking NET remodeling with antibodies, which can prevent the awakening of dormant cells [120]. Moreover, MMPs can disrupt blood vessels in metastatic tissues, leading to the production of TGF-β1 and ECM molecules, such as periostin and FN, by endothelial cells and contributing to the deposition of ECM components [112]. These findings emphasize the importance of the interaction between the TME and ECM in the development of cancer dormancy escape and metastasis.
In addition to the previous discussion, the composition of the ECM plays an important role in chemoresistance and immunosuppression. For example, the aberrant expression of FN can activate cellular signaling pathways, such as PI3K/Akt, ERK, and FAK signaling, through interaction with tumor cell α5β1 or β1 integrins, which leads to tumor cell resistance to chemotherapeutic agents [126]. Additionally, cancer cell adhesion to the ECM can result in a drug resistance phenotype, termed cell adhesion-mediated drug resistance [127]. For instance, the activation of the LN and its receptor mGr1-Ag/37LRP pathway can reduce drug accumulation and inhibit apoptosis, resulting in GC cell resistance [128]. Moreover, collagen has been suggested as a predictor and a strategic target for the improvement of the efficacy of immune checkpoint inhibitors in anti-PD-1 responses by promoting the infiltration and accumulation of effector CD8+ T cells in tumors [129,130].

5. Therapeutic Strategies Targeting CAF-Secreted Cytokines/Chemokines That Maintain or Awaken Dormancy

In early studies, cancer cell dormancy had already been identified as a key driver of drug resistance and cancer relapse, yet DCC remains difficult to monitor in patients, representing a gap in the fight against drug resistance. Dormancy entry/escape is a dynamic interaction between tumor cells and the TME, and the factors secreted by CAFs play an essential role in this process. Therefore, one strategy to combat cancer relapse may be to select factors secreted by CAFs as therapeutic targets (Figure 3) [131]. Here, we focus on relevant drugs that target factors secreted by CAFs and may therefore help combat dormancy-related mechanisms, particularly in GI cancers.

5.1. Prevention of Dormant Cancer Cell Reawakening by Targeting Their Crosstalk with Their Supportive Microenvironment

Maintaining tumor cells in a dormant state prevents metastatic cancer from recurring. In other words, it is necessary to develop new therapeutic strategies that prevent cancer recurrence by maintaining a dormant state, which could be based on indefinitely blocking the crosstalk between CAF-driven signaling and the supportive microenvironment involved in dormancy escape [132].
First, uPAR may be an attractive target for maintaining the dormant state of cancer cells and thus inhibiting the metastatic process of cancer [133,134]. For example, the antibody ATN-292 reduces the migration of human pancreatic cancer cells by inhibiting the binding of uPA to uPAR [135]. Qin et al. developed a novel anti-uPAR monoclonal antibody that exerts antitumor effects in GC by blocking the uPA/uPAR interaction [136]. In addition, the small molecule uPA inhibitor, WX-671, was tested in a phase II trial in patients with locally advanced non-metastatic pancreatic cancer; however, in the study, the combination of WX-671 and gemcitabine, a nucleoside analog used as a chemotherapeutic agent, was shown to be well tolerated, but the survival outcomes of the combination group were similar to those in the gemcitabine-only group [137].
The abundance of TGF-β1 in the TME drives dormancy escape, while CAFs are the main source of TGF-β in the TME. Agents that inhibit TGF-β1, including those that interfere with the ligand–receptor interactions or the activity of the receptor kinase, can be used as a strategy to keep tumor cells in a dormant state. Some of these agents have been or are being clinically evaluated for the treatment of gastrointestinal cancers [138]. To cite an instance, SRK-181 was developed to specifically bind the pro-segment of TGF-β1, which effectively prevents the dissociation of the mature TGF-β1 from its pro-segment. This results in the maintenance of TGF-β1 in a latent state without impacting the activation of TGF-β2 or TGF-β3 [139]. Based on current evidence, it is postulated that SRK-181 exerts its inhibitory effects systemically, suggesting its potential to modulate TGF-β1 signaling pathways, including canonical or non-canonical pathways [140]. Preclinical data suggest that SRK-181 may overcome primary resistance to checkpoint inhibitors [139,141,142]. Moreover, LY2157299 is a small molecule inhibitor of TGF-βRI kinase, also known as Galunisertib, which demonstrated efficacy in a Phase Ib/II trial in combination with gemcitabine in patients with unresectable pancreatic cancer, with minimal increase in toxicity [143,144,145]. Another TGF-βRI inhibitor, Ki26894, also known as Kirin, has been shown to inhibit a GC tumor through the reduction of invasive and bone metastases [146].
CAFs secrete pro-inflammatory cytokines, most of which promote dormancy. Many anti-inflammatory therapies have been extensively studied and applied in cancer treatment. Among these, non-steroidal anti-inflammatory molecules such as sulindac [147,148,149] and celecoxib [150,151] exert their influence by inhibiting COX-2 activity, and they have shown positive responses in preclinical and clinical studies on gastrointestinal cancers. Sulindac is currently undergoing a phase III clinical trial (NCT01349881) investigating its use alone or in combination with eflornithine to reduce the three-year event rate of adenomas and second primary colorectal cancer in patients previously treated for stage 0 to III colon or rectal cancer. With regard to celecoxib, a meta-analysis study has demonstrated its potential in improving response rates to standard chemotherapy among patients diagnosed with advanced colorectal cancer. Nevertheless, it is worth noting that this improvement has not been shown to increase one-year survival rates [152]. Currently, another phase III clinical trial is underway to further evaluate the efficacy of oxaliplatin, leucovorin calcium, and fluorouracil in combination with or without celecoxib, in patients with stage III colon cancer who have undergone surgery (NCT01150045) [153]. Given their presumed anti-inflammatory properties, these drugs may also hold potential in inhibiting dormancy escape [154,155,156].
CAF-mediated ECM remodeling is an important factor in dormancy escape [157,158]. ECM-associated molecules, such as collagen and FN, regulate the roles of integrins in the transition from dormancy to proliferation through binding [159]. Therefore, an indirect approach that could be considered is to inhibit integrin activation. Aberrant integrin expression is common in GI cancers and is thought to be associated with cancer metastasis [160]. For example, α5β1 is upregulated in GC [161] and CRC [162]. Clinically, a phase Ib trial of the anti-α5β1 integrin antibody Volociximab with carboplatin and paclitaxel in advanced non-small cell lung cancer has shown a better prognosis [163]. THR-687, a pan-αv/α5β1 inhibitor, has been tested in clinical trials for diabetic macular edema [164] and retinal vascular disease. OS2966, a monoclonal antibody directed against the β1 integrin, has entered clinical trials for the treatment of high-grade glioma [165]. Consequently, these integrin-targeting drugs have been linked to the mechanism by which CAFs regulate cancer cell dormancy, and further research is needed to determine whether they are effective in preventing the awakening of DCCs.
In addition, the inhibition of LOX or LOXL2 may be an option for preventing the awakening of DCCs [166]. Benjamin et al. showed that Lox activity promotes chemoresistance in pancreatic cancer cells. They found that an elevated level of Lox activity results in an increase in the organization of collagen fibers, which reduces the benefit of gemcitabine [167]. Zhao et al. showed that the β-aminopropionitrile (BAPN) inhibition of LOX in GC cells reduced the expression and activity of MMP-2 and MMP-9 [168]. Similarly, Li et al. showed that the inhibition of LOX by BAPN hampered the outgrowth of GC liver metastasis [169]. Simtuzumab is a humanized monoclonal antibody to IgG4 with an anti-LOXL2 function, and it has been tested in phase II trials in combination with gemcitabine and FOLFIRI (folinic acid, fluorouracil, and irinotecan) in patients with pancreatic [170] and colorectal [171] cancers, respectively. In these clinical trials, Simtuzumab was generally well tolerated. However, it is possible that Simtuzumab only targeted extracellular LOXL2 and therefore demonstrated limited clinical benefit [172]. Recently, epigallocatechin gallate (EGCG) has been shown to inhibit LOXL2 and TGF-β1, and clinical trials have shown that EGCG reduces TGF-β1 signaling and collagen accumulation [173], potentially inhibiting dormancy escape.
Another way to prevent dormant cells from awakening is to strengthen the dormant environment. As mentioned above, TSP-1 drives tumor cells into a dormant state [174]. Therefore, the administration of the TSP-1 mimetic peptide prosaposin generates a microenvironment that inhibits cancer metastasis [175]. Furthermore, the combination of 5-AZA with trans-retinoic acid (ATRA) restores the NR2F1-induced dormant program and induces TGF-β2, thereby inhibiting cancer metastasis by restoring the TGF-β-SMAD4 signaling [176]. As for therapeutic approaches that directly target components of the ECM, one example is the inhibition of FN biosynthesis. Tomasini-Johansson et al. developed a small peptide inhibitor, pUR4B, which effectively limits the polymerization of FN in the matrix [177]. pUR4B was also demonstrated by Hielscher and colleagues in co-cultures of fibroblasts and breast cancer cells to reduce not only the deposition of FN in the ECM and organization, but also to inhibit the deposition of other stromal proteins [178]. However, to date, neither strategy has been validated in studies of GI cancers.

5.2. Awakening Dormant Cancer Cells to Re-Sensitize Them to Therapy

Based on a growing understanding of the mechanisms of cancer dormancy, the prevention of cancer cells from entering dormancy or forcing DCCs to awaken, thereby increasing cellular sensitivity to therapeutic agents, is a novel strategy that may be developed to combat the recurrence of drug-resistant disease. The inhibition of dormancy-promoting factors influenced by CAFs may be one way to achieve this. For example, TGF-β2 binds to the TGF-βRIII signaling pathway and induces tumor cells to enter a dormant state. AP 12009 (also known as Trabedersen), an antisense oligonucleotide (ASO) targeting TGF-β2 mRNA, has been shown to be safe in phase I/II studies in patients with pancreatic and colorectal cancer [179].
CAF-secreted DKK-1 is a key factor in the promotion of cancer cell dormancy. DKN-01 is a humanized monoclonal antibody that binds to and blocks the activity of the DKK-1, and it has been evaluated in several clinical trials for GI cancers. A phase II study of DKN-01 in combination with Tislelizumab with or without chemotherapy as first line therapy for patients with metastatic gastric cancer or gastroesophageal junction adenocarcinoma is ongoing [180,181]. Another a phase Ib clinical trial showed that DKN-01 in combination with pembrolizumab was well tolerated in advanced esophageal cancer patients [182]. In addition, a phase II study of DKN-01 in combination with standard-of-care bevacizumab (Avastin) and chemotherapy as second-line treatment for patients with advanced CRC (NCT05480306) is ongoing. However, in these clinical trials, the effectiveness of the treatment may depend on the amount of DKK1 that is expressed by the patient’s own tumor.
Other factors secreted by CAFs, such as GDF-10 and BMP4, have been reported to be involved in the promotion of cancer cell dormancy. Treatment targeting these factors has not been investigated in GI cancers [113,183,184]. In the future, related antibodies, RNA interference, or inhibitors may have potential therapeutic effects and become treatment options for GI cancers. In conclusion, by inhibiting the CAF-derived factors that promote dormancy in the TME, tumor cells can be prevented from entering or awakened from dormancy. It is recommended that this be carried out early in the treatment process to prevent the tumor cells from acquiring a strong malignant capacity. Table 1 summarizes the factors secreted by CAFs that have been targeted by agents used in GI cancers. The table highlights potential strategies that could be applied to target cancer dormancy at different stages.

6. Conclusions

The communication between cytokines/chemokines secreted by CAFs and other components of the TME, as well as ECM remodeling and immune modulation, are involved in the development of cancer (Figure 1), which includes resistance to treatment, dormancy, and metastasis. Metastatic recurrence is a significant problem in patients with GI cancers and is a leading cause of death. Cancer dormancy was initially identified in breast cancer, where cancer cells remain inactive for a prolonged period before recurrence [185]. However, less research has been performed on dormancy associated with GI-related cancers.
This review focused on how CAF secretion affects the GI TME and its impact on cancer dormancy (Figure 2). The decision to enter or exit cancer dormancy depends on the dynamics of the TME, with CAF secretion playing a critical role. Therefore, CAFs are potential targets for therapeutic strategies in the treatment of cancer and the prevention of recurrence and metastasis. However, some CAFs have dual roles in the cancer cell dormancy/awakening mechanism [186,187]. Early in treatment, CAFs may help cancer cells enter dormancy and survive treatment, while at later stages, CAFs create a microenvironment conducive to metastasis, thus allowing cancer cells to escape from dormancy and metastasize with greater malignancy. Further studies are needed to evaluate the specific responses of each secreted factor in detail.
This review summarized several potential approaches to targeting CAF secretion, such as maintaining cancer cells in a dormant state indefinitely or awakening DCCs to re-sensitize them to treatment (Figure 3). However, these approaches have their complexities and require the consideration of the treatment window and patient stratification. Ideally, the former should be used in the post-treatment phase, and it may require a long period of continuous application. The latter should be used from the early stages of treatment and in combination with other treatments such as chemotherapy or immunotherapy [188].
Taken together, it is reasonable to believe that CAF-driven secretion in the TME can be used as a strategy for combating GI cancer recurrence, which scientifically and theoretically provides an insightful basis for the development of anti-cancer treatment.

Author Contributions

Writing—review, conceptualization, and figure composition, S.-H.C.; writing—review and supervision, J.-W.W.; writing—review and editing, H.-Y.C.C.; writing—review, conceptualization, and supervision, M.-H.L. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Grants from the National Science and Technology Council, Taiwan (111-2314-B-037-048-MY3 and 109-2314-B-037-034-MY3), and Kaohsiung Medical University Hospital (KMUH110-0T05).

Acknowledgments

All the figures were created with BioRender (BioRender.com). The Agreement numbers of publication and license rights for the following images are: Figure 1, WN25AO820V; Figure 2, BB25ANSC0H; Figure 3, PR25ANI6J0.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Arnold, M.; Abnet, C.C.; Neale, R.E.; Vignat, J.; Giovannucci, E.L.; McGlynn, K.A.; Bray, F. Global Burden of 5 Major Types of Gastrointestinal Cancer. Gastroenterology 2020, 159, 335–349.e15. [Google Scholar] [CrossRef] [PubMed]
  2. Xing, F.; Saidou, J.; Watabe, K. Cancer associated fibroblasts (CAFs) in tumor microenvironment. Front. Biosci. 2010, 15, 166–179. [Google Scholar] [CrossRef] [PubMed]
  3. Ham, I.H.; Lee, D.; Hur, H. Cancer-Associated Fibroblast-Induced Resistance to Chemotherapy and Radiotherapy in Gastrointestinal Cancers. Cancers 2021, 13, 1172. [Google Scholar] [CrossRef] [PubMed]
  4. Klein, C.A. Cancer progression and the invisible phase of metastatic colonization. Nat. Rev. Cancer 2020, 20, 681–694. [Google Scholar] [CrossRef]
  5. Pranzini, E.; Raugei, G.; Taddei, M.L. Metabolic Features of Tumor Dormancy: Possible Therapeutic Strategies. Cancers 2022, 14, 547. [Google Scholar] [CrossRef]
  6. Van den Berg, I.; Coebergh van den Braak, R.R.J.; van Vugt, J.L.A.; Ijzermans, J.N.M.; Buettner, S. Actual survival after resection of primary colorectal cancer: Results from a prospective multicenter study. World J. Surg. Oncol. 2021, 19, 96. [Google Scholar] [CrossRef]
  7. Okusaka, T. Treatment for postoperative recurrence of pancreatic cancer: A narrative review. Chin. Clin. Oncol. 2022, 11, 19. [Google Scholar] [CrossRef]
  8. Pang, H.-Y.; Yan, M.-H.; Chen, L.-H.; Chen, X.-F.; Chen, Z.-X.; Zhang, S.-R.; Sun, H. Detection of asymptomatic recurrence following curative surgery improves survival in patients with gastric cancer: A systematic review and meta-analysis. Front. Oncol. 2022, 12, 1011683. [Google Scholar] [CrossRef]
  9. Pedersen, R.N.; Esen, B.; Mellemkjær, L.; Christiansen, P.; Ejlertsen, B.; Lash, T.L.; Nørgaard, M.; Cronin-Fenton, D. The Incidence of Breast Cancer Recurrence 10-32 Years After Primary Diagnosis. J. Natl. Cancer Inst. 2022, 114, 391–399. [Google Scholar] [CrossRef]
  10. Marin, J.J.; Al-Abdulla, R.; Lozano, E.; Briz, O.; Bujanda, L.; Banales, J.M.; Macias, R.I. Mechanisms of Resistance to Chemotherapy in Gastric Cancer. Anti-Cancer Agents Med. Chem. 2016, 16, 318–334. [Google Scholar] [CrossRef]
  11. Ruan, T.; Liu, W.; Tao, K.; Wu, C. A Review of Research Progress in Multidrug-Resistance Mechanisms in Gastric Cancer. OncoTargets Ther. 2020, 13, 1797–1807. [Google Scholar] [CrossRef]
  12. Foster, D.S.; Januszyk, M.; Delitto, D.; Yost, K.E.; Griffin, M.; Guo, J.; Guardino, N.; Delitto, A.E.; Chinta, M.; Burcham, A.R.; et al. Multiomic analysis reveals conservation of cancer-associated fibroblast phenotypes across species and tissue of origin. Cancer Cell 2022, 40, 1392–1406.e7. [Google Scholar] [CrossRef]
  13. Galbo, P.M., Jr.; Zang, X.; Zheng, D. Molecular Features of Cancer-associated Fibroblast Subtypes and their Implication on Cancer Pathogenesis, Prognosis, and Immunotherapy Resistance. Clin. Cancer Res. 2021, 27, 2636–2647. [Google Scholar] [CrossRef]
  14. Liu, T.; Zhou, L.; Li, D.; Andl, T.; Zhang, Y. Cancer-Associated Fibroblasts Build and Secure the Tumor Microenvironment. Front. Cell Dev. Biol. 2019, 7, 60. [Google Scholar] [CrossRef]
  15. Huang, L.; Xu, A.M.; Liu, S.; Liu, W.; Li, T.J. Cancer-associated fibroblasts in digestive tumors. World J. Gastroenterol. 2014, 20, 17804–17818. [Google Scholar] [CrossRef]
  16. Kuzet, S.E.; Gaggioli, C. Fibroblast activation in cancer: When seed fertilizes soil. Cell Tissue Res. 2016, 365, 607–619. [Google Scholar] [CrossRef]
  17. Caja, L.; Dituri, F.; Mancarella, S.; Caballero-Diaz, D.; Moustakas, A.; Giannelli, G.; Fabregat, I. TGF-β and the Tissue Microenvironment: Relevance in Fibrosis and Cancer. Int. J. Mol. Sci. 2018, 19, 1294. [Google Scholar] [CrossRef]
  18. Liu, F.L.; Mo, E.P.; Yang, L.; Du, J.; Wang, H.S.; Zhang, H.; Kurihara, H.; Xu, J.; Cai, S.H. Autophagy is involved in TGF-β1-induced protective mechanisms and formation of cancer-associated fibroblasts phenotype in tumor microenvironment. Oncotarget 2016, 7, 4122–4141. [Google Scholar] [CrossRef]
  19. Yoon, H.; Tang, C.-M.; Banerjee, S.; Delgado, A.L.; Yebra, M.; Davis, J.; Sicklick, J.K. TGF-β1-mediated transition of resident fibroblasts to cancer-associated fibroblasts promotes cancer metastasis in gastrointestinal stromal tumor. Oncogenesis 2021, 10, 13. [Google Scholar] [CrossRef]
  20. Kalluri, R. The biology and function of fibroblasts in cancer. Nat. Rev. Cancer 2016, 16, 582–598. [Google Scholar] [CrossRef]
  21. Qin, Y.; Wang, F.; Ni, H.; Liu, Y.; Yin, Y.; Zhou, X.; Gao, G.; Li, Q.; Qi, X.; Li, J. Cancer-associated fibroblasts in gastric cancer affect malignant progression via the CXCL12-CXCR4 axis. J. Cancer 2021, 12, 3011–3023. [Google Scholar] [CrossRef] [PubMed]
  22. Sperb, N.; Tsesmelis, M.; Wirth, T. Crosstalk between Tumor and Stromal Cells in Pancreatic Ductal Adenocarcinoma. Int. J. Mol. Sci. 2020, 21, 5486. [Google Scholar] [CrossRef] [PubMed]
  23. Arwert, E.N.; Milford, E.L.; Rullan, A.; Derzsi, S.; Hooper, S.; Kato, T.; Mansfield, D.; Melcher, A.; Harrington, K.J.; Sahai, E. STING and IRF3 in stromal fibroblasts enable sensing of genomic stress in cancer cells to undermine oncolytic viral therapy. Nat. Cell Biol. 2020, 22, 758–766. [Google Scholar] [CrossRef] [PubMed]
  24. Briukhovetska, D.; Dörr, J.; Endres, S.; Libby, P.; Dinarello, C.A.; Kobold, S. Interleukins in cancer: From biology to therapy. Nat. Rev. Cancer 2021, 21, 481–499. [Google Scholar] [CrossRef] [PubMed]
  25. Rašková, M.; Lacina, L.; Kejík, Z.; Venhauerová, A.; Skaličková, M.; Kolář, M.; Jakubek, M.; Rosel, D.; Smetana, K., Jr.; Brábek, J. The Role of IL-6 in Cancer Cell Invasiveness and Metastasis-Overview and Therapeutic Opportunities. Cells 2022, 11, 3698. [Google Scholar] [CrossRef]
  26. Bazzichetto, C.; Milella, M.; Zampiva, I.; Simionato, F.; Amoreo, C.A.; Buglioni, S.; Pacelli, C.; Le Pera, L.; Colombo, T.; Bria, E.; et al. Interleukin-8 in Colorectal Cancer: A Systematic Review and Meta-Analysis of Its Potential Role as a Prognostic Biomarker. Biomedicines 2022, 10, 2631. [Google Scholar] [CrossRef]
  27. Najdaghi, S.; Razi, S.; Rezaei, N. An overview of the role of interleukin-8 in colorectal cancer. Cytokine 2020, 135, 155205. [Google Scholar] [CrossRef]
  28. Wu, X.; Tao, P.; Zhou, Q.; Li, J.; Yu, Z.; Wang, X.; Li, J.; Li, C.; Yan, M.; Zhu, Z.; et al. IL-6 secreted by cancer-associated fibroblasts promotes epithelial-mesenchymal transition and metastasis of gastric cancer via JAK2/STAT3 signaling pathway. Oncotarget 2017, 8, 20741–20750. [Google Scholar] [CrossRef]
  29. Zhong, B.; Cheng, B.; Huang, X.; Xiao, Q.; Niu, Z.; Chen, Y.F.; Yu, Q.; Wang, W.; Wu, X.J. Colorectal cancer-associated fibroblasts promote metastasis by up-regulating LRG1 through stromal IL-6/STAT3 signaling. Cell Death Dis. 2021, 13, 16. [Google Scholar] [CrossRef]
  30. Heichler, C.; Scheibe, K.; Schmied, A.; Geppert, C.I.; Schmid, B.; Wirtz, S.; Thoma, O.M.; Kramer, V.; Waldner, M.J.; Büttner, C.; et al. STAT3 activation through IL-6/IL-11 in cancer-associated fibroblasts promotes colorectal tumour development and correlates with poor prognosis. Gut 2020, 69, 1269–1282. [Google Scholar] [CrossRef]
  31. Walker, C.; Mojares, E.; Del Río Hernández, A. Role of Extracellular Matrix in Development and Cancer Progression. Int. J. Mol. Sci. 2018, 19, 28. [Google Scholar] [CrossRef]
  32. Foster, D.S.; Jones, R.E.; Ransom, R.C.; Longaker, M.T.; Norton, J.A. The evolving relationship of wound healing and tumor stroma. JCI Insight 2018, 3, 99911. [Google Scholar] [CrossRef]
  33. Tenti, P.; Vannucci, L. Lysyl oxidases: Linking structures and immunity in the tumor microenvironment. Cancer Immunol. Immunother. CII 2020, 69, 223–235. [Google Scholar] [CrossRef]
  34. Marozzi, M.; Parnigoni, A.; Negri, A.; Viola, M.; Vigetti, D.; Passi, A.; Karousou, E.; Rizzi, F. Inflammation, Extracellular Matrix Remodeling, and Proteostasis in Tumor Microenvironment. Int. J. Mol. Sci. 2021, 22, 8102. [Google Scholar] [CrossRef]
  35. Ao, T.; Mochizuki, S.; Kajiwara, Y.; Yonemura, K.; Shiraishi, T.; Nagata, K.; Shinto, E.; Okamoto, K.; Nearchou, I.P.; Shimazaki, H.; et al. Cancer-associated fibroblasts at the unfavorable desmoplastic stroma promote colorectal cancer aggressiveness: Potential role of ADAM9. Int. J. Cancer 2022, 150, 1706–1721. [Google Scholar] [CrossRef]
  36. Oh, S.; Park, Y.; Lee, H.J.; Lee, J.; Lee, S.H.; Baek, Y.S.; Chun, S.K.; Lee, S.M.; Kim, M.; Chon, Y.E.; et al. A Disintegrin and Metalloproteinase 9 (ADAM9) in Advanced Hepatocellular Carcinoma and Their Role as a Biomarker During Hepatocellular Carcinoma Immunotherapy. Cancers 2020, 12, 745. [Google Scholar] [CrossRef]
  37. Łukaszewicz-Zając, M.; Pączek, S.; Mroczko, B. A Disintegrin and Metalloproteinase (ADAM) Family-Novel Biomarkers of Selected Gastrointestinal (GI) Malignancies? Cancers 2022, 14, 2307. [Google Scholar] [CrossRef]
  38. Mao, X.; Xu, J.; Wang, W.; Liang, C.; Hua, J.; Liu, J.; Zhang, B.; Meng, Q.; Yu, X.; Shi, S. Crosstalk between cancer-associated fibroblasts and immune cells in the tumor microenvironment: New findings and future perspectives. Mol. Cancer 2021, 20, 131. [Google Scholar] [CrossRef]
  39. Mace, T.A.; Ameen, Z.; Collins, A.; Wojcik, S.; Mair, M.; Young, G.S.; Fuchs, J.R.; Eubank, T.D.; Frankel, W.L.; Bekaii-Saab, T.; et al. Pancreatic cancer-associated stellate cells promote differentiation of myeloid-derived suppressor cells in a STAT3-dependent manner. Cancer Res. 2013, 73, 3007–3018. [Google Scholar] [CrossRef]
  40. Cho, H.; Seo, Y.; Loke, K.M.; Kim, S.-W.; Oh, S.-M.; Kim, J.-H.; Soh, J.; Kim, H.S.; Lee, H.; Kim, J.; et al. Cancer-Stimulated CAFs Enhance Monocyte Differentiation and Protumoral TAM Activation via IL6 and GM-CSF Secretion. Clin. Cancer Res. 2018, 24, 5407–5421. [Google Scholar] [CrossRef]
  41. Zhang, R.; Qi, F.; Zhao, F.; Li, G.; Shao, S.; Zhang, X.; Yuan, L.; Feng, Y. Cancer-associated fibroblasts enhance tumor-associated macrophages enrichment and suppress NK cells function in colorectal cancer. Cell Death Dis. 2019, 10, 273. [Google Scholar] [CrossRef] [PubMed]
  42. Inoue, C.; Miki, Y.; Saito, R.; Hata, S.; Abe, J.; Sato, I.; Okada, Y.; Sasano, H. PD-L1 Induction by Cancer-Associated Fibroblast-Derived Factors in Lung Adenocarcinoma Cells. Cancers 2019, 11, 1257. [Google Scholar] [CrossRef] [PubMed]
  43. Li, Z.; Zhou, J.; Zhang, J.; Li, S.; Wang, H.-S.; Du, J. Cancer-associated fibroblasts promote PD-L1 expression in mice cancer cells via secreting CXCL5. Int. J. Cancer 2019, 145, 1946–1957. [Google Scholar] [CrossRef] [PubMed]
  44. Ziani, L.; Chouaib, S.; Thiery, J. Alteration of the Antitumor Immune Response by Cancer-Associated Fibroblasts. Front. Immunol. 2018, 9, 414. [Google Scholar] [CrossRef]
  45. Huang, X.; Zhang, F.; Wang, X.; Liu, K. The Role of Indoleamine 2, 3-Dioxygenase 1 in Regulating Tumor Microenvironment. Cancers 2022, 14, 2756. [Google Scholar] [CrossRef]
  46. Diab, M.; El-Rayes, B.F. The heterogeneity of CAFs and immune cell populations in the tumor microenvironment of pancreatic adenocarcinoma. J. Cancer Metastasis Treat. 2022, 8, 42. [Google Scholar] [CrossRef]
  47. Phan, T.G.; Croucher, P.I. The dormant cancer cell life cycle. Nat. Rev. Cancer 2020, 20, 398–411. [Google Scholar] [CrossRef]
  48. Vera-Ramirez, L.; Hunter, K.W. Tumor cell dormancy as an adaptive cell stress response mechanism. F1000Research 2017, 6, 2134. [Google Scholar] [CrossRef]
  49. Aguirre-Ghiso, J.A.; Estrada, Y.; Liu, D.; Ossowski, L. ERK(MAPK) activity as a determinant of tumor growth and dormancy; regulation by p38(SAPK). Cancer Res. 2003, 63, 1684–1695. [Google Scholar] [CrossRef]
  50. Sosa, M.S.; Avivar-Valderas, A.; Bragado, P.; Wen, H.C.; Aguirre-Ghiso, J.A. ERK1/2 and p38α/β signaling in tumor cell quiescence: Opportunities to control dormant residual disease. Clin. Cancer Res. 2011, 17, 5850–5857. [Google Scholar] [CrossRef]
  51. Bragado, P.; Estrada, Y.; Parikh, F.; Krause, S.; Capobianco, C.; Farina, H.G.; Schewe, D.M.; Aguirre-Ghiso, J.A. TGF-β2 dictates disseminated tumour cell fate in target organs through TGF-β-RIII and p38α/β signalling. Nat. Cell Biol. 2013, 15, 1351–1361. [Google Scholar] [CrossRef]
  52. Barkan, D.; El Touny, L.H.; Michalowski, A.M.; Smith, J.A.; Chu, I.; Davis, A.S.; Webster, J.D.; Hoover, S.; Simpson, R.M.; Gauldie, J.; et al. Metastatic growth from dormant cells induced by a col-I-enriched fibrotic environment. Cancer Res. 2010, 70, 5706–5716. [Google Scholar] [CrossRef]
  53. Wu, J.; Fu, R.; Liu, Z.; Li, G.; Huang, X.; Xue, Y.; Xu, Y.; Sun, Y.; Zhao, J.; Mi, J. Cell proliferation downregulated by TGF-β2-triggered G1/S checkpoint in clinical CAFs. Cell Cycle 2017, 16, 172–178. [Google Scholar] [CrossRef]
  54. Scherz-Shouval, R.; Santagata, S.; Mendillo, M.L.; Sholl, L.M.; Ben-Aharon, I.; Beck, A.H.; Dias-Santagata, D.; Koeva, M.; Stemmer, S.M.; Whitesell, L.; et al. The reprogramming of tumor stroma by HSF1 is a potent enabler of malignancy. Cell 2014, 158, 564–578. [Google Scholar] [CrossRef]
  55. Butturini, E.; Carcereri de Prati, A.; Boriero, D.; Mariotto, S. Tumor Dormancy and Interplay with Hypoxic Tumor Microenvironment. Int. J. Mol. Sci. 2019, 20, 4305. [Google Scholar] [CrossRef]
  56. Qin, T.; Chen, K.; Li, J.; Qian, W.; Xiao, Y.; Wu, E.; Ma, J.; Chen, Z.; Wang, Z.; Ma, Q.; et al. Heat shock factor 1 inhibition sensitizes pancreatic cancer to gemcitabine via the suppression of cancer stem cell-like properties. Biomed. Pharmacother. 2022, 148, 112713. [Google Scholar] [CrossRef]
  57. Zhang, D.; Song, Y.; Li, D.; Liu, X.; Pan, Y.; Ding, L.; Shi, G.; Wang, Y.; Ni, Y.; Hou, Y. Cancer-associated fibroblasts promote tumor progression by lncRNA-mediated RUNX2/GDF10 signaling in oral squamous cell carcinoma. Mol. Oncol. 2022, 16, 780–794. [Google Scholar] [CrossRef]
  58. Yu-Lee, L.Y.; Yu, G.; Lee, Y.C.; Lin, S.C.; Pan, J.; Pan, T.; Yu, K.J.; Liu, B.; Creighton, C.J.; Rodriguez-Canales, J.; et al. Osteoblast-Secreted Factors Mediate Dormancy of Metastatic Prostate Cancer in the Bone via Activation of the TGFβRIII-p38MAPK-pS249/T252RB Pathway. Cancer Res. 2018, 78, 2911–2924. [Google Scholar] [CrossRef]
  59. Shirai, Y.T.; Ehata, S.; Yashiro, M.; Yanagihara, K.; Hirakawa, K.; Miyazono, K. Bone morphogenetic protein-2 and -4 play tumor suppressive roles in human diffuse-type gastric carcinoma. Am. J. Pathol. 2011, 179, 2920–2930. [Google Scholar] [CrossRef]
  60. Yokoyama, Y.; Watanabe, T.; Tamura, Y.; Hashizume, Y.; Miyazono, K.; Ehata, S. Autocrine BMP-4 Signaling Is a Therapeutic Target in Colorectal Cancer. Cancer Res. 2017, 77, 4026–4038. [Google Scholar] [CrossRef]
  61. Owen, K.L.; Gearing, L.J.; Zanker, D.J.; Brockwell, N.K.; Khoo, W.H.; Roden, D.L.; Cmero, M.; Mangiola, S.; Hong, M.K.; Spurling, A.J.; et al. Prostate cancer cell-intrinsic interferon signaling regulates dormancy and metastatic outgrowth in bone. EMBO Rep. 2020, 21, e50162. [Google Scholar] [CrossRef] [PubMed]
  62. Lan, Q.; Peyvandi, S.; Duffey, N.; Huang, Y.-T.; Barras, D.; Held, W.; Richard, F.; Delorenzi, M.; Sotiriou, C.; Desmedt, C.; et al. Type I interferon/IRF7 axis instigates chemotherapy-induced immunological dormancy in breast cancer. Oncogene 2019, 38, 2814–2829. [Google Scholar] [CrossRef] [PubMed]
  63. Guo, L.; Fang, T.; Jiang, Y.; Liu, D. IRF7 is a Prognostic Biomarker and Associated with Immune Infiltration in Stomach Adenocarcinoma. Int. J. Gen. Med. 2021, 14, 9887–9902. [Google Scholar] [CrossRef] [PubMed]
  64. Aqbi, H.F.; Wallace, M.; Sappal, S.; Payne, K.K.; Manjili, M.H. IFN-γ orchestrates tumor elimination, tumor dormancy, tumor escape, and progression. J. Leukoc. Biol. 2018, 103, 1219–1223. [Google Scholar] [CrossRef]
  65. Liu, Y.; Liang, X.; Yin, X.; Lv, J.; Tang, K.; Ma, J.; Ji, T.; Zhang, H.; Dong, W.; Jin, X.; et al. Blockade of IDO-kynurenine-AhR metabolic circuitry abrogates IFN-γ-induced immunologic dormancy of tumor-repopulating cells. Nat. Commun. 2017, 8, 15207. [Google Scholar] [CrossRef]
  66. Hong, S.A.; Yoo, S.H.; Lee, H.H.; Sun, D.S.; Won, H.S.; Kim, O.; Ko, Y.H. Prognostic value of Dickkopf-1 and ß-catenin expression in advanced gastric cancer. BMC Cancer 2018, 18, 506. [Google Scholar] [CrossRef]
  67. Malladi, S.; Macalinao, D.G.; Jin, X.; He, L.; Basnet, H.; Zou, Y.; de Stanchina, E.; Massagué, J. Metastatic Latency and Immune Evasion through Autocrine Inhibition of WNT. Cell 2016, 165, 45–60. [Google Scholar] [CrossRef]
  68. Smith, A.G.; Macleod, K.F. Autophagy, cancer stem cells and drug resistance. J. Pathol. 2019, 247, 708–718. [Google Scholar] [CrossRef]
  69. Akkoc, Y.; Peker, N.; Akcay, A.; Gozuacik, D. Autophagy and Cancer Dormancy. Front. Oncol. 2021, 11, 627023. [Google Scholar] [CrossRef]
  70. Wang, Y.; Gan, G.; Wang, B.; Wu, J.; Cao, Y.; Zhu, D.; Xu, Y.; Wang, X.; Han, H.; Li, X.; et al. Cancer-associated Fibroblasts Promote Irradiated Cancer Cell Recovery Through Autophagy. EBioMedicine 2017, 17, 45–56. [Google Scholar] [CrossRef]
  71. Rajbhandari, N.; Lin, W.C.; Wehde, B.L.; Triplett, A.A.; Wagner, K.U. Autocrine IGF1 Signaling Mediates Pancreatic Tumor Cell Dormancy in the Absence of Oncogenic Drivers. Cell Rep. 2017, 18, 2243–2255. [Google Scholar] [CrossRef]
  72. Lu, H.; Han, M.; Yuan, X.; Tursun, K.; Zhang, Y.; Li, Y.; Li, Z.; Feng, S.; Zhou, L.; Pan, Z.; et al. Role of IL-6-mediated expression of NS5ATP9 in autophagy of liver cancer cells. J. Cell. Physiol. 2018, 233, 9312–9319. [Google Scholar] [CrossRef]
  73. Natale, G.; Bocci, G. Does metronomic chemotherapy induce tumor angiogenic dormancy? A review of available preclinical and clinical data. Cancer Lett. 2018, 432, 28–37. [Google Scholar] [CrossRef]
  74. Pascual, G.; Avgustinova, A.; Mejetta, S.; Martín, M.; Castellanos, A.; Attolini, C.S.; Berenguer, A.; Prats, N.; Toll, A.; Hueto, J.A.; et al. Targeting metastasis-initiating cells through the fatty acid receptor CD36. Nature 2017, 541, 41–45. [Google Scholar] [CrossRef]
  75. Watt, M.J.; Clark, A.K.; Selth, L.A.; Haynes, V.R.; Lister, N.; Rebello, R.; Porter, L.H.; Niranjan, B.; Whitby, S.T.; Lo, J.; et al. Suppressing fatty acid uptake has therapeutic effects in preclinical models of prostate cancer. Sci. Transl. Med. 2019, 11, eaau5758. [Google Scholar] [CrossRef]
  76. Marisi, G.; Scarpi, E.; Passardi, A.; Nanni, O.; Pagan, F.; Valgiusti, M.; Casadei Gardini, A.; Neri, L.M.; Frassineti, G.L.; Amadori, D.; et al. IL-8 and thrombospondin-1 as prognostic markers in patients with metastatic colorectal cancer receiving bevacizumab. Cancer Manag. Res. 2018, 10, 5659–5666. [Google Scholar] [CrossRef]
  77. Park, S.Y.; Nam, J.S. The force awakens: Metastatic dormant cancer cells. Exp. Mol. Med. 2020, 52, 569–581. [Google Scholar] [CrossRef]
  78. Liu, D.; Aguirre Ghiso, J.; Estrada, Y.; Ossowski, L. EGFR is a transducer of the urokinase receptor initiated signal that is required for in vivo growth of a human carcinoma. Cancer Cell 2002, 1, 445–457. [Google Scholar] [CrossRef]
  79. Aguirre Ghiso, J.A. Inhibition of FAK signaling activated by urokinase receptor induces dormancy in human carcinoma cells in vivo. Oncogene 2002, 21, 2513–2524. [Google Scholar] [CrossRef]
  80. Katoh, M. Canonical and non-canonical WNT signaling in cancer stem cells and their niches: Cellular heterogeneity, omics reprogramming, targeted therapy and tumor plasticity (Review). Int. J. Oncol. 2017, 51, 1357–1369. [Google Scholar] [CrossRef]
  81. Oskarsson, T.; Acharyya, S.; Zhang, X.H.; Vanharanta, S.; Tavazoie, S.F.; Morris, P.G.; Downey, R.J.; Manova-Todorova, K.; Brogi, E.; Massagué, J. Breast cancer cells produce tenascin C as a metastatic niche component to colonize the lungs. Nat. Med. 2011, 17, 867–874. [Google Scholar] [CrossRef] [PubMed]
  82. Bao, S.; Ouyang, G.; Bai, X.; Huang, Z.; Ma, C.; Liu, M.; Shao, R.; Anderson, R.M.; Rich, J.N.; Wang, X.F. Periostin potently promotes metastatic growth of colon cancer by augmenting cell survival via the Akt/PKB pathway. Cancer Cell 2004, 5, 329–339. [Google Scholar] [CrossRef] [PubMed]
  83. Vermeulen, L.; De Sousa, E.M.F.; van der Heijden, M.; Cameron, K.; de Jong, J.H.; Borovski, T.; Tuynman, J.B.; Todaro, M.; Merz, C.; Rodermond, H.; et al. Wnt activity defines colon cancer stem cells and is regulated by the microenvironment. Nat. Cell Biol. 2010, 12, 468–476. [Google Scholar] [CrossRef] [PubMed]
  84. Cui, D.; Huang, Z.; Liu, Y.; Ouyang, G. The multifaceted role of periostin in priming the tumor microenvironments for tumor progression. Cell. Mol. Life Sci. CMLS 2017, 74, 4287–4291. [Google Scholar] [CrossRef] [PubMed]
  85. Yu, B.; Wu, K.; Wang, X.; Zhang, J.; Wang, L.; Jiang, Y.; Zhu, X.; Chen, W.; Yan, M. Periostin secreted by cancer-associated fibroblasts promotes cancer stemness in head and neck cancer by activating protein tyrosine kinase 7. Cell Death Dis. 2018, 9, 1082. [Google Scholar] [CrossRef]
  86. Moniuszko, T.; Wincewicz, A.; Koda, M.; Domysławska, I.; Sulkowski, S. Role of periostin in esophageal, gastric and colon cancer (Review). Oncol. Lett. 2016, 12, 783–787. [Google Scholar] [CrossRef]
  87. Koh, S.A.; Lee, K.H. Function of hepatocyte growth factor in gastric cancer proliferation and invasion. Yeungnam Univ. J. Med. 2020, 37, 73–78. [Google Scholar] [CrossRef]
  88. Zhang, H.; Feng, Q.; Chen, W.D.; Wang, Y.D. HGF/c-MET: A Promising Therapeutic Target in the Digestive System Cancers. Int. J. Mol. Sci. 2018, 19, 3295. [Google Scholar] [CrossRef]
  89. Chong, Y.; Tang, D.; Xiong, Q.; Jiang, X.; Xu, C.; Huang, Y.; Wang, J.; Zhou, H.; Shi, Y.; Wu, X.; et al. Galectin-1 from cancer-associated fibroblasts induces epithelial–mesenchymal transition through β1 integrin-mediated upregulation of Gli1 in gastric cancer. J. Exp. Clin. Cancer Res. 2016, 35, 175. [Google Scholar] [CrossRef]
  90. Zhang, X.; Zhang, C.; Ren, Z.; Zhang, F.; Xu, J.; Zhang, X.; Zheng, H. Curcumin Affects Gastric Cancer Cell Migration, Invasion and Cytoskeletal Remodeling Through Gli1-β-Catenin. Cancer Manag. Res. 2020, 12, 3795–3806. [Google Scholar] [CrossRef]
  91. Greten, F.R.; Grivennikov, S.I. Inflammation and Cancer: Triggers, Mechanisms, and Consequences. Immunity 2019, 51, 27–41. [Google Scholar] [CrossRef]
  92. Chernosky, N.M.; Tamagno, I. The Role of the Innate Immune System in Cancer Dormancy and Relapse. Cancers 2021, 13, 5621. [Google Scholar] [CrossRef]
  93. Khazali, A.S.; Clark, A.M.; Wells, A. Inflammatory cytokine IL-8/CXCL8 promotes tumour escape from hepatocyte-induced dormancy. Br. J. Cancer 2018, 118, 566–576. [Google Scholar] [CrossRef]
  94. Zhang, J.; Li, S.; Zhao, Y.; Ma, P.; Cao, Y.; Liu, C.; Zhang, X.; Wang, W.; Chen, L.; Li, Y. Cancer-associated fibroblasts promote the migration and invasion of gastric cancer cells via activating IL-17a/JAK2/STAT3 signaling. Ann. Transl. Med. 2020, 8, 877. [Google Scholar] [CrossRef]
  95. Jiang, Y.X.; Yang, S.W.; Li, P.A.; Luo, X.; Li, Z.Y.; Hao, Y.X.; Yu, P.W. The promotion of the transformation of quiescent gastric cancer stem cells by IL-17 and the underlying mechanisms. Oncogene 2017, 36, 1256–1264. [Google Scholar] [CrossRef]
  96. Baram, T.; Rubinstein-Achiasaf, L.; Ben-Yaakov, H.; Ben-Baruch, A. Inflammation-Driven Breast Tumor Cell Plasticity: Stemness/EMT, Therapy Resistance and Dormancy. Front. Oncol. 2020, 10, 614468. [Google Scholar] [CrossRef]
  97. Zheng, Y.; Comaills, V.; Burr, R.; Boulay, G.; Miyamoto, D.T.; Wittner, B.S.; Emmons, E.; Sil, S.; Koulopoulos, M.W.; Broderick, K.T.; et al. COX-2 mediates tumor-stromal prolactin signaling to initiate tumorigenesis. Proc. Natl. Acad. Sci. USA 2019, 116, 5223–5232. [Google Scholar] [CrossRef]
  98. Nagaraju, G.P.; El-Rayes, B.F. Cyclooxygenase-2 in gastrointestinal malignancies. Cancer 2019, 125, 1221–1227. [Google Scholar] [CrossRef]
  99. Zhu, Y.; Shi, C.; Zeng, L.; Liu, G.; Jiang, W.; Zhang, X.; Chen, S.; Guo, J.; Jian, X.; Ouyang, J.; et al. High COX-2 expression in cancer-associated fibiroblasts contributes to poor survival and promotes migration and invasiveness in nasopharyngeal carcinoma. Mol. Carcinog. 2020, 59, 265–280. [Google Scholar] [CrossRef]
  100. Tanaka, T.; Narazaki, M.; Kishimoto, T. IL-6 in inflammation, immunity, and disease. Cold Spring Harb. Perspect. Biol. 2014, 6, a016295. [Google Scholar] [CrossRef]
  101. Shaashua, L.; Eckerling, A.; Israeli, B.; Yanovich, G.; Rosenne, E.; Fichman-Horn, S.; Ben Zvi, I.; Sorski, L.; Haldar, R.; Satchi-Fainaro, R.; et al. Spontaneous regression of micro-metastases following primary tumor excision: A critical role for primary tumor secretome. BMC Biol. 2020, 18, 163. [Google Scholar] [CrossRef]
  102. Esposito, A.; Ferraresi, A.; Salwa, A.; Vidoni, C.; Dhanasekaran, D.N.; Isidoro, C. Resveratrol Contrasts IL-6 Pro-Growth Effects and Promotes Autophagy-Mediated Cancer Cell Dormancy in 3D Ovarian Cancer: Role of miR-1305 and of Its Target ARH-I. Cancers 2022, 14, 2142. [Google Scholar] [CrossRef] [PubMed]
  103. Romero-Moreno, R.; Curtis, K.J.; Coughlin, T.R.; Miranda-Vergara, M.C.; Dutta, S.; Natarajan, A.; Facchine, B.A.; Jackson, K.M.; Nystrom, L.; Li, J.; et al. The CXCL5/CXCR2 axis is sufficient to promote breast cancer colonization during bone metastasis. Nat. Commun. 2019, 10, 4404. [Google Scholar] [CrossRef] [PubMed]
  104. Zhao, J.; Ou, B.; Han, D.; Wang, P.; Zong, Y.; Zhu, C.; Liu, D.; Zheng, M.; Sun, J.; Feng, H.; et al. Tumor-derived CXCL5 promotes human colorectal cancer metastasis through activation of the ERK/Elk-1/Snail and AKT/GSK3β/β-catenin pathways. Mol. Cancer 2017, 16, 70. [Google Scholar] [CrossRef]
  105. Mao, Z.; Zhang, J.; Shi, Y.; Li, W.; Shi, H.; Ji, R.; Mao, F.; Qian, H.; Xu, W.; Zhang, X. CXCL5 promotes gastric cancer metastasis by inducing epithelial-mesenchymal transition and activating neutrophils. Oncogenesis 2020, 9, 63. [Google Scholar] [CrossRef]
  106. Zhang, W.; Wang, H.; Sun, M.; Deng, X.; Wu, X.; Ma, Y.; Li, M.; Shuoa, S.M.; You, Q.; Miao, L. CXCL5/CXCR2 axis in tumor microenvironment as potential diagnostic biomarker and therapeutic target. Cancer Commun. 2020, 40, 69–80. [Google Scholar] [CrossRef]
  107. Deng, J.; Jiang, R.; Meng, E.; Wu, H. CXCL5: A coachman to drive cancer progression. Front. Oncol. 2022, 12, 944494. [Google Scholar] [CrossRef]
  108. Adamski, V.; Hattermann, K.; Kubelt, C.; Cohrs, G.; Lucius, R.; Synowitz, M.; Sebens, S.; Held-Feindt, J. Entry and exit of chemotherapeutically-promoted cellular dormancy in glioblastoma cells is differentially affected by the chemokines CXCL12, CXCL16, and CX3CL1. Oncogene 2020, 39, 4421–4435. [Google Scholar] [CrossRef]
  109. Nobutani, K.; Shimono, Y.; Mizutani, K.; Ueda, Y.; Suzuki, T.; Kitayama, M.; Minami, A.; Momose, K.; Miyawaki, K.; Akashi, K.; et al. Downregulation of CXCR4 in Metastasized Breast Cancer Cells and Implication in Their Dormancy. PLoS ONE 2015, 10, e0130032. [Google Scholar] [CrossRef]
  110. Peng, C.; Zou, X.; Xia, W.; Gao, H.; Li, Z.; Liu, N.; Xu, Z.; Gao, C.; He, Z.; Niu, W.; et al. Integrin αvβ6 plays a bi-directional regulation role between colon cancer cells and cancer-associated fibroblasts. Biosci. Rep. 2018, 38, BSR20180243. [Google Scholar] [CrossRef]
  111. Yako, Y.Y.; Kruger, D.; Smith, M.; Brand, M. Cytokines as Biomarkers of Pancreatic Ductal Adenocarcinoma: A Systematic Review. PLoS ONE 2016, 11, e0154016. [Google Scholar] [CrossRef]
  112. Ghajar, C.M.; Peinado, H.; Mori, H.; Matei, I.R.; Evason, K.J.; Brazier, H.; Almeida, D.; Koller, A.; Hajjar, K.A.; Stainier, D.Y.; et al. The perivascular niche regulates breast tumour dormancy. Nat. Cell Biol. 2013, 15, 807–817. [Google Scholar] [CrossRef]
  113. Gao, H.; Chakraborty, G.; Lee-Lim, A.P.; Mo, Q.; Decker, M.; Vonica, A.; Shen, R.; Brogi, E.; Brivanlou, A.H.; Giancotti, F.G. The BMP inhibitor Coco reactivates breast cancer cells at lung metastatic sites. Cell 2012, 150, 764–779. [Google Scholar] [CrossRef]
  114. Belfiore, A.; Rapicavoli, R.V.; Le Moli, R.; Lappano, R.; Morrione, A.; De Francesco, E.M.; Vella, V. IGF2: A Role in Metastasis and Tumor Evasion from Immune Surveillance? Biomedicines 2023, 11, 229. [Google Scholar] [CrossRef]
  115. Xu, W.W.; Li, B.; Guan, X.Y.; Chung, S.K.; Wang, Y.; Yip, Y.L.; Law, S.Y.K.; Chan, K.T.; Lee, N.P.Y.; Chan, K.W.; et al. Cancer cell-secreted IGF2 instigates fibroblasts and bone marrow-derived vascular progenitor cells to promote cancer progression. Nat. Commun. 2017, 8, 14399. [Google Scholar] [CrossRef]
  116. Winkler, J.; Abisoye-Ogunniyan, A.; Metcalf, K.J.; Werb, Z. Concepts of extracellular matrix remodelling in tumour progression and metastasis. Nat. Commun. 2020, 11, 5120. [Google Scholar] [CrossRef]
  117. Aguirre-Ghiso, J.A.; Liu, D.; Mignatti, A.; Kovalski, K.; Ossowski, L. Urokinase receptor and fibronectin regulate the ERK(MAPK) to p38(MAPK) activity ratios that determine carcinoma cell proliferation or dormancy in vivo. Mol. Biol. Cell 2001, 12, 863–879. [Google Scholar] [CrossRef]
  118. Erdogan, B.; Ao, M.; White, L.M.; Means, A.L.; Brewer, B.M.; Yang, L.; Washington, M.K.; Shi, C.; Franco, O.E.; Weaver, A.M.; et al. Cancer-associated fibroblasts promote directional cancer cell migration by aligning fibronectin. J. Cell Biol. 2017, 216, 3799–3816. [Google Scholar] [CrossRef]
  119. Chen, R.; Huang, L.; Hu, K. Natural products remodel cancer-associated fibroblasts in desmoplastic tumors. Acta Pharm. Sin. B 2020, 10, 2140–2155. [Google Scholar] [CrossRef]
  120. Albrengues, J.; Shields, M.A.; Ng, D.; Park, C.G.; Ambrico, A.; Poindexter, M.E.; Upadhyay, P.; Uyeminami, D.L.; Pommier, A.; Küttner, V.; et al. Neutrophil extracellular traps produced during inflammation awaken dormant cancer cells in mice. Science 2018, 361, eaao4227. [Google Scholar] [CrossRef]
  121. Pickup, M.W.; Laklai, H.; Acerbi, I.; Owens, P.; Gorska, A.E.; Chytil, A.; Aakre, M.; Weaver, V.M.; Moses, H.L. Stromally derived lysyl oxidase promotes metastasis of transforming growth factor-β-deficient mouse mammary carcinomas. Cancer Res. 2013, 73, 5336–5346. [Google Scholar] [CrossRef] [PubMed]
  122. Liburkin-Dan, T.; Toledano, S.; Neufeld, G. Lysyl Oxidase Family Enzymes and Their Role in Tumor Progression. Int. J. Mol. Sci. 2022, 23, 6249. [Google Scholar] [CrossRef] [PubMed]
  123. Gao, Y.; Xiao, Q.; Ma, H.; Li, L.; Liu, J.; Feng, Y.; Fang, Z.; Wu, J.; Han, X.; Zhang, J.; et al. LKB1 inhibits lung cancer progression through lysyl oxidase and extracellular matrix remodeling. Proc. Natl. Acad. Sci. USA 2010, 107, 18892–18897. [Google Scholar] [CrossRef] [PubMed]
  124. Sneddon, J.B.; Zhen, H.H.; Montgomery, K.; van de Rijn, M.; Tward, A.D.; West, R.; Gladstone, H.; Chang, H.Y.; Morganroth, G.S.; Oro, A.E.; et al. Bone morphogenetic protein antagonist gremlin 1 is widely expressed by cancer-associated stromal cells and can promote tumor cell proliferation. Proc. Natl. Acad. Sci. USA 2006, 103, 14842–14847. [Google Scholar] [CrossRef] [PubMed]
  125. Karagiannis, G.S.; Berk, A.; Dimitromanolakis, A.; Diamandis, E.P. Enrichment map profiling of the cancer invasion front suggests regulation of colorectal cancer progression by the bone morphogenetic protein antagonist, gremlin-1. Mol. Oncol. 2013, 7, 826–839. [Google Scholar] [CrossRef]
  126. Wang, J.P.; Hielscher, A. Fibronectin: How Its Aberrant Expression in Tumors May Improve Therapeutic Targeting. J. Cancer 2017, 8, 674–682. [Google Scholar] [CrossRef]
  127. Damiano, J.S.; Cress, A.E.; Hazlehurst, L.A.; Shtil, A.A.; Dalton, W.S. Cell adhesion mediated drug resistance (CAM-DR): Role of integrins and resistance to apoptosis in human myeloma cell lines. Blood 1999, 93, 1658–1667. [Google Scholar] [CrossRef]
  128. Sun, L.; Liu, L.; Liu, X.; Wang, Y.; Li, M.; Yao, L.; Yang, J.; Ji, G.; Guo, C.; Pan, Y.; et al. Gastric cancer cell adhesion to laminin enhances acquired chemotherapeutic drug resistance mediated by MGr1-Ag/37LRP. Oncol. Rep. 2014, 32, 105–114. [Google Scholar] [CrossRef]
  129. Peng, D.H.; Rodriguez, B.L.; Diao, L.; Chen, L.; Wang, J.; Byers, L.A.; Wei, Y.; Chapman, H.A.; Yamauchi, M.; Behrens, C.; et al. Collagen promotes anti-PD-1/PD-L1 resistance in cancer through LAIR1-dependent CD8(+) T cell exhaustion. Nat. Commun. 2020, 11, 4520. [Google Scholar] [CrossRef]
  130. Baldari, S.; Di Modugno, F.; Nisticò, P.; Toietta, G. Strategies for Efficient Targeting of Tumor Collagen for Cancer Therapy. Cancers 2022, 14, 4706. [Google Scholar] [CrossRef]
  131. Czekay, R.P.; Cheon, D.J.; Samarakoon, R.; Kutz, S.M.; Higgins, P.J. Cancer-Associated Fibroblasts: Mechanisms of Tumor Progression and Novel Therapeutic Targets. Cancers 2022, 14, 1231. [Google Scholar] [CrossRef]
  132. Ghajar, C.M. Metastasis prevention by targeting the dormant niche. Nat. Rev. Cancer 2015, 15, 238–247. [Google Scholar] [CrossRef]
  133. Ma, Y.Y.; Tao, H.Q. Role of urokinase plasminogen activator receptor in gastric cancer: A potential therapeutic target. Cancer Biother. Radiopharm. 2012, 27, 285–290. [Google Scholar] [CrossRef]
  134. Zhai, B.-T.; Tian, H.; Sun, J.; Zou, J.-B.; Zhang, X.-F.; Cheng, J.-X.; Shi, Y.-J.; Fan, Y.; Guo, D.-Y. Urokinase-type plasminogen activator receptor (uPAR) as a therapeutic target in cancer. J. Transl. Med. 2022, 20, 135. [Google Scholar] [CrossRef]
  135. Bauer, T.W.; Liu, W.; Fan, F.; Camp, E.R.; Yang, A.; Somcio, R.J.; Bucana, C.D.; Callahan, J.; Parry, G.C.; Evans, D.B.; et al. Targeting of urokinase plasminogen activator receptor in human pancreatic carcinoma cells inhibits c-Met- and insulin-like growth factor-I receptor-mediated migration and invasion and orthotopic tumor growth in mice. Cancer Res. 2005, 65, 7775–7781. [Google Scholar] [CrossRef]
  136. Qin, L.; Wang, L.; Zhang, J.; Zhou, H.; Yang, Z.; Wang, Y.; Cai, W.; Wen, F.; Jiang, X.; Zhang, T.; et al. Therapeutic strategies targeting uPAR potentiate anti-PD-1 efficacy in diffuse-type gastric cancer. Sci. Adv. 2022, 8, eabn3774. [Google Scholar] [CrossRef]
  137. Heinemann, V.; Ebert, M.P.; Laubender, R.P.; Bevan, P.; Mala, C.; Boeck, S. Phase II randomised proof-of-concept study of the urokinase inhibitor upamostat (WX-671) in combination with gemcitabine compared with gemcitabine alone in patients with non-resectable, locally advanced pancreatic cancer. Br. J. Cancer 2013, 108, 766–770. [Google Scholar] [CrossRef]
  138. Sabbadini, F.; Bertolini, M.; De Matteis, S.; Mangiameli, D.; Contarelli, S.; Pietrobono, S.; Melisi, D. The Multifaceted Role of TGF-β in Gastrointestinal Tumors. Cancers 2021, 13, 3960. [Google Scholar] [CrossRef]
  139. Martin, C.J.; Datta, A.; Littlefield, C.; Kalra, A.; Chapron, C.; Wawersik, S.; Dagbay, K.B.; Brueckner, C.T.; Nikiforov, A.; Danehy, F.T., Jr.; et al. Selective inhibition of TGFβ1 activation overcomes primary resistance to checkpoint blockade therapy by altering tumor immune landscape. Sci. Transl. Med. 2020, 12, aay8456. [Google Scholar] [CrossRef]
  140. Budi, E.H.; Schaub, J.R.; Decaris, M.; Turner, S.; Derynck, R. TGF-β as a driver of fibrosis: Physiological roles and therapeutic opportunities. J. Pathol. 2021, 254, 358–373. [Google Scholar] [CrossRef]
  141. Welsh, B.T.; Faucette, R.; Bilic, S.; Martin, C.J.; Schürpf, T.; Chen, D.; Nicholls, S.; Lansita, J.; Kalra, A. Nonclinical Development of SRK-181: An Anti-Latent TGFβ1 Monoclonal Antibody for the Treatment of Locally Advanced or Metastatic Solid Tumors. Int. J. Toxicol. 2021, 40, 226–241. [Google Scholar] [CrossRef] [PubMed]
  142. Yap, T.A.; Barve, M.A.; Gainor, J.F.; Weekes, C.D.; Bockorny, B.; Ju, Y.; Faucette, R.; Bilic, S.; Lee-Hoeflich, S.T.; Song, G.; et al. First-in-human phase 1 trial (DRAGON) of SRK-181, a potential first-in-class selective latent TGFβ1 inhibitor, alone or in combination with anti-PD-(L)1 treatment in patients with advanced solid tumors. J. Clin. Oncol. 2021, 39, TPS3146. [Google Scholar] [CrossRef]
  143. Melisi, D.; Garcia-Carbonero, R.; Macarulla, T.; Pezet, D.; Deplanque, G.; Fuchs, M.; Trojan, J.; Oettle, H.; Kozloff, M.; Cleverly, A.; et al. Galunisertib plus gemcitabine vs. gemcitabine for first-line treatment of patients with unresectable pancreatic cancer. Br. J. Cancer 2018, 119, 1208–1214. [Google Scholar] [CrossRef] [PubMed]
  144. Gueorguieva, I.; Tabernero, J.; Melisi, D.; Macarulla, T.; Merz, V.; Waterhouse, T.H.; Miles, C.; Lahn, M.M.; Cleverly, A.; Benhadji, K.A. Population pharmacokinetics and exposure-overall survival analysis of the transforming growth factor-β inhibitor galunisertib in patients with pancreatic cancer. Cancer Chemother. Pharmacol. 2019, 84, 1003–1015. [Google Scholar] [CrossRef]
  145. Melisi, D.; Oh, D.Y.; Hollebecque, A.; Calvo, E.; Varghese, A.; Borazanci, E.; Macarulla, T.; Merz, V.; Zecchetto, C.; Zhao, Y.; et al. Safety and activity of the TGFβ receptor I kinase inhibitor galunisertib plus the anti-PD-L1 antibody durvalumab in metastatic pancreatic cancer. J. Immunother. Cancer 2021, 9, e002068. [Google Scholar] [CrossRef]
  146. Shinto, O.; Yashiro, M.; Kawajiri, H.; Shimizu, K.; Shimizu, T.; Miwa, A.; Hirakawa, K. Inhibitory effect of a TGFbeta receptor type-I inhibitor, Ki26894, on invasiveness of scirrhous gastric cancer cells. Br. J. Cancer 2010, 102, 844–851. [Google Scholar] [CrossRef]
  147. Meyskens, F.L., Jr.; McLaren, C.E.; Pelot, D.; Fujikawa-Brooks, S.; Carpenter, P.M.; Hawk, E.; Kelloff, G.; Lawson, M.J.; Kidao, J.; McCracken, J.; et al. Difluoromethylornithine plus sulindac for the prevention of sporadic colorectal adenomas: A randomized placebo-controlled, double-blind trial. Cancer Prev. Res. 2008, 1, 32–38. [Google Scholar] [CrossRef]
  148. Chandra, S.; Nymeyer, A.C.; Rice, P.F.; Gerner, E.W.; Barton, J.K. Intermittent Dosing with Sulindac Provides Effective Colorectal Cancer Chemoprevention in the Azoxymethane-Treated Mouse Model. Cancer Prev. Res. 2017, 10, 459–466. [Google Scholar] [CrossRef]
  149. Davis, J.S.; Kanikarla-Marie, P.; Gagea, M.; Yu, P.L.; Fang, D.; Sebastian, M.; Yang, P.; Hawk, E.; Dashwood, R.; Lichtenberger, L.M.; et al. Sulindac plus a phospholipid is effective for polyp reduction and safer than sulindac alone in a mouse model of colorectal cancer development. BMC Cancer 2020, 20, 871. [Google Scholar] [CrossRef]
  150. Guo, Q.; Li, Q.; Wang, J.; Liu, M.; Wang, Y.; Chen, Z.; Ye, Y.; Guan, Q.; Zhou, Y. A comprehensive evaluation of clinical efficacy and safety of celecoxib in combination with chemotherapy in metastatic or postoperative recurrent gastric cancer patients: A preliminary, three-center, clinical trial study. Medicine 2019, 98, e16234. [Google Scholar] [CrossRef]
  151. Wen, B.; Wei, Y.T.; Mu, L.L.; Wen, G.R.; Zhao, K. The molecular mechanisms of celecoxib in tumor development. Medicine 2020, 99, e22544. [Google Scholar] [CrossRef]
  152. Ye, S.Y.; Li, J.Y.; Li, T.H.; Song, Y.X.; Sun, J.X.; Chen, X.W.; Zhao, J.H.; Li, Y.; Wu, Z.H.; Gao, P.; et al. The Efficacy and Safety of Celecoxib in Addition to Standard Cancer Therapy: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Curr. Oncol. 2022, 29, 6137–6153. [Google Scholar] [CrossRef]
  153. Brown, J.C.; Ma, C.; Shi, Q.; Fuchs, C.S.; Meyer, J.; Niedzwiecki, D.; Zemla, T.; Couture, F.; Kuebler, P.; Kumar, P.; et al. Physical Activity in Stage III Colon Cancer: CALGB/SWOG 80702 (Alliance). J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2023, 41, 243–254. [Google Scholar] [CrossRef]
  154. Egashira, I.; Takahashi-Yanaga, F.; Nishida, R.; Arioka, M.; Igawa, K.; Tomooka, K.; Nakatsu, Y.; Tsuzuki, T.; Nakabeppu, Y.; Kitazono, T.; et al. Celecoxib and 2,5-dimethylcelecoxib inhibit intestinal cancer growth by suppressing the Wnt/β-catenin signaling pathway. Cancer Sci. 2017, 108, 108–115. [Google Scholar] [CrossRef]
  155. Trask, D.; Bock, J.; Goswami, P.; Menon, S. Celecoxib and sulindac sulfide enhance the cytotoxic effect of standard chemotherapeutic agents and possess anti-tumor activity mediated by p21cipl/waf1 expression. J. Clin. Oncol. 2004, 22, 3186. [Google Scholar] [CrossRef]
  156. Cha, B.K.; Kim, Y.S.; Hwang, K.E.; Cho, K.H.; Oh, S.H.; Kim, B.R.; Jun, H.Y.; Yoon, K.H.; Jeong, E.T.; Kim, H.R. Celecoxib and sulindac inhibit TGF-β1-induced epithelial-mesenchymal transition and suppress lung cancer migration and invasion via downregulation of sirtuin 1. Oncotarget 2016, 7, 57213–57227. [Google Scholar] [CrossRef]
  157. Paolillo, M.; Schinelli, S. Extracellular Matrix Alterations in Metastatic Processes. Int. J. Mol. Sci. 2019, 20, 4947. [Google Scholar] [CrossRef]
  158. Parker, A.L.; Cox, T.R. The Role of the ECM in Lung Cancer Dormancy and Outgrowth. Front. Oncol. 2020, 10, 1766. [Google Scholar] [CrossRef]
  159. Poltavets, V.; Kochetkova, M.; Pitson, S.M.; Samuel, M.S. The Role of the Extracellular Matrix and Its Molecular and Cellular Regulators in Cancer Cell Plasticity. Front. Oncol. 2018, 8, 431. [Google Scholar] [CrossRef]
  160. Hou, S.; Wang, J.; Li, W.; Hao, X.; Hang, Q. Roles of Integrins in Gastrointestinal Cancer Metastasis. Front. Mol. Biosci. 2021, 8, 708779. [Google Scholar] [CrossRef]
  161. Ren, J.; Xu, S.; Guo, D.; Zhang, J.; Liu, S. Increased expression of α5β1-integrin is a prognostic marker for patients with gastric cancer. Clin. Transl. Oncol. 2014, 16, 668–674. [Google Scholar] [CrossRef] [PubMed]
  162. Thongchot, S.; Singsuksawat, E.; Sumransub, N.; Pongpaibul, A.; Trakarnsanga, A.; Thuwajit, P.; Thuwajit, C. Periostin regulates autophagy through integrin α5β1 or α6β4 and an AKT-dependent pathway in colorectal cancer cell migration. J. Cell. Mol. Med. 2020, 24, 12421–12432. [Google Scholar] [CrossRef] [PubMed]
  163. Besse, B.; Tsao, L.C.; Chao, D.T.; Fang, Y.; Soria, J.C.; Almokadem, S.; Belani, C.P. Phase Ib safety and pharmacokinetic study of volociximab, an anti-α5β1 integrin antibody, in combination with carboplatin and paclitaxel in advanced non-small-cell lung cancer. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2013, 24, 90–96. [Google Scholar] [CrossRef] [PubMed]
  164. Khanani, A.M.; Patel, S.S.; Gonzalez, V.H.; Moon, S.J.; Jaffe, G.J.; Wells, J.A.; Kozma, P.; Dugel, P.U.; Maturi, R.K. Phase 1 Study of THR-687, a Novel, Highly Potent Integrin Antagonist for the Treatment of Diabetic Macular Edema. Ophthalmol. Sci. 2021, 1, 100040. [Google Scholar] [CrossRef]
  165. Nwagwu, C.D.; Immidisetti, A.V.; Bukanowska, G.; Vogelbaum, M.A.; Carbonell, A.M. Convection-Enhanced Delivery of a First-in-Class Anti-β1 Integrin Antibody for the Treatment of High-Grade Glioma Utilizing Real-Time Imaging. Pharmaceutics 2020, 13, 40. [Google Scholar] [CrossRef]
  166. Weidenfeld, K.; Schif-Zuck, S.; Abu-Tayeh, H.; Kang, K.; Kessler, O.; Weissmann, M.; Neufeld, G.; Barkan, D. Dormant tumor cells expressing LOXL2 acquire a stem-like phenotype mediating their transition to proliferative growth. Oncotarget 2016, 7, 71362–71377. [Google Scholar] [CrossRef]
  167. Le Calvé, B.; Griveau, A.; Vindrieux, D.; Maréchal, R.; Wiel, C.; Svrcek, M.; Gout, J.; Azzi, L.; Payen, L.; Cros, J.; et al. Lysyl oxidase family activity promotes resistance of pancreatic ductal adenocarcinoma to chemotherapy by limiting the intratumoral anticancer drug distribution. Oncotarget 2016, 7, 32100–32112. [Google Scholar] [CrossRef]
  168. Zhao, L.; Niu, H.; Liu, Y.; Wang, L.; Zhang, N.; Zhang, G.; Liu, R.; Han, M. LOX inhibition downregulates MMP-2 and MMP-9 in gastric cancer tissues and cells. J. Cancer 2019, 10, 6481–6490. [Google Scholar] [CrossRef]
  169. Li, Q.; Zhu, C.C.; Ni, B.; Zhang, Z.Z.; Jiang, S.H.; Hu, L.P.; Wang, X.; Zhang, X.X.; Huang, P.Q.; Yang, Q.; et al. Lysyl oxidase promotes liver metastasis of gastric cancer via facilitating the reciprocal interactions between tumor cells and cancer associated fibroblasts. EBioMedicine 2019, 49, 157–171. [Google Scholar] [CrossRef]
  170. Benson, A.B., 3rd; Wainberg, Z.A.; Hecht, J.R.; Vyushkov, D.; Dong, H.; Bendell, J.; Kudrik, F. A Phase II Randomized, Double-Blind, Placebo-Controlled Study of Simtuzumab or Placebo in Combination with Gemcitabine for the First-Line Treatment of Pancreatic Adenocarcinoma. Oncologist 2017, 22, 241-e15. [Google Scholar] [CrossRef]
  171. Hecht, J.R.; Benson, A.B., 3rd; Vyushkov, D.; Yang, Y.; Bendell, J.; Verma, U. A Phase II, Randomized, Double-Blind, Placebo-Controlled Study of Simtuzumab in Combination with FOLFIRI for the Second-Line Treatment of Metastatic KRAS Mutant Colorectal Adenocarcinoma. Oncologist 2017, 22, 243-e23. [Google Scholar] [CrossRef]
  172. Chopra, V.; Sangarappillai, R.M.; Romero-Canelón, I.; Jones, A.M. Lysyl Oxidase Like-2 (LOXL2): An Emerging Oncology Target. Adv. Ther. 2020, 3, 1900119. [Google Scholar] [CrossRef]
  173. Wei, Y.; Dong, W.; Jackson, J.; Ho, T.C.; Le Saux, C.J.; Brumwell, A.; Li, X.; Klesney-Tait, J.; Cohen, M.L.; Wolters, P.J.; et al. Blocking LOXL2 and TGFβ1 signalling induces collagen I turnover in precision-cut lung slices derived from patients with idiopathic pulmonary fibrosis. Thorax 2021, 76, 729–732. [Google Scholar] [CrossRef]
  174. Carlson, P.; Dasgupta, A.; Grzelak, C.A.; Kim, J.; Barrett, A.; Coleman, I.M.; Shor, R.E.; Goddard, E.T.; Dai, J.; Schweitzer, E.M.; et al. Targeting the perivascular niche sensitizes disseminated tumour cells to chemotherapy. Nat. Cell Biol. 2019, 21, 238–250. [Google Scholar] [CrossRef]
  175. Catena, R.; Bhattacharya, N.; El Rayes, T.; Wang, S.; Choi, H.; Gao, D.; Ryu, S.; Joshi, N.; Bielenberg, D.; Lee, S.B.; et al. Bone marrow-derived Gr1+ cells can generate a metastasis-resistant microenvironment via induced secretion of thrombospondin-1. Cancer Discov. 2013, 3, 578–589. [Google Scholar] [CrossRef]
  176. Singh, D.K.; Farias, E.; Carcamo, S.; Hasson, D.; Sun, D.; Cheung, J.; Nobre, A.R.; Kale, N.; Sosa, M.S.; Bernstein, E.; et al. Epigenetic reprogramming of DCCs into dormancy suppresses metastasis via restored TGFβ–SMAD4 signaling. bioRxiv 2021. [Google Scholar] [CrossRef]
  177. Tomasini-Johansson, B.R.; Kaufman, N.R.; Ensenberger, M.G.; Ozeri, V.; Hanski, E.; Mosher, D.F. A 49-residue peptide from adhesin F1 of Streptococcus pyogenes inhibits fibronectin matrix assembly. J. Biol. Chem. 2001, 276, 23430–23439. [Google Scholar] [CrossRef]
  178. Hielscher, A.; Ellis, K.; Qiu, C.; Porterfield, J.; Gerecht, S. Fibronectin Deposition Participates in Extracellular Matrix Assembly and Vascular Morphogenesis. PLoS ONE 2016, 11, e0147600. [Google Scholar] [CrossRef]
  179. Oettle, H.; Hilbig, A.; Seufferlein, T.; Tsianakas, A.; Luger, T.; Schmid, R.M.; von Wichert, G.; Endlicher, E.; Garbe, C.; Kaehler, K.K.; et al. Phase I/II study with trabedersen (AP 12009) monotherapy for the treatment of patients with advanced pancreatic cancer, malignant melanoma, and colorectal carcinoma. J. Clin. Oncol. 2011, 29, 2513. [Google Scholar] [CrossRef]
  180. Klempner, S.J.; Chao, J.; Uronis, H.E.; Sirard, C.A.; Kagey, M.; Baum, J.; Song, J.; Wang, J.; Kim, I.-H.; Lee, K.W.; et al. DKN-01 and tislelizumab ± chemotherapy as a first-line (1L) and second-line (2L) investigational therapy in advanced gastroesophageal adenocarcinoma (GEA): DisTinGuish Trial. J. Clin. Oncol. 2022, 40, 292. [Google Scholar] [CrossRef]
  181. Lee, K.-W.; Moehler, M.; Cunningham, D.; Wainberg, Z.; Uronis, H.; Oh, D.-Y.; Kim, I.-H.; Shim, B.; Sym, S.; Altura, R.; et al. Trial in progress: A phase 2, multicenter, open-label study of DKN-01 in combination with tislelizumab and chemotherapy as 1L therapy in patients with unresectable, locally advanced or metastatic gastric or gastroesophageal junction adenocarcinoma (G/GEJ; DisTinGuish). J. Clin. Oncol. 2023, 41, TPS484. [Google Scholar] [CrossRef]
  182. Klempner, S.J.; Bendell, J.C.; Villaflor, V.M.; Tenner, L.L.; Stein, S.M.; Rottman, J.B.; Naik, G.S.; Sirard, C.A.; Kagey, M.H.; Chaney, M.F.; et al. Safety, Efficacy, and Biomarker Results from a Phase Ib Study of the Anti-DKK1 Antibody DKN-01 in Combination with Pembrolizumab in Advanced Esophagogastric Cancers. Mol. Cancer Ther. 2021, 20, 2240–2249. [Google Scholar] [CrossRef] [PubMed]
  183. Senft, D.; Ronai, Z.A. Immunogenic, cellular, and angiogenic drivers of tumor dormancy—A melanoma view. Pigment Cell Melanoma Res. 2016, 29, 27–42. [Google Scholar] [CrossRef] [PubMed]
  184. Zhou, T.; Yu, L.; Huang, J.; Zhao, X.; Li, Y.; Hu, Y.; Lei, Y. GDF10 inhibits proliferation and epithelial-mesenchymal transition in triple-negative breast cancer via upregulation of Smad7. Aging 2019, 11, 3298–3314. [Google Scholar] [CrossRef]
  185. Banys, M.; Hartkopf, A.D.; Krawczyk, N.; Kaiser, T.; Meier-Stiegen, F.; Fehm, T.; Neubauer, H. Dormancy in breast cancer. Breast Cancer 2012, 4, 183–191. [Google Scholar] [CrossRef]
  186. Alkasalias, T.; Moyano-Galceran, L.; Arsenian-Henriksson, M.; Lehti, K. Fibroblasts in the Tumor Microenvironment: Shield or Spear? Int. J. Mol. Sci. 2018, 19, 1532. [Google Scholar] [CrossRef]
  187. Dai, L.; Li, M.; Zhang, W.-L.; Tang, Y.-J.; Tang, Y.-L.; Liang, X.-H. Fibroblasts in cancer dormancy: Foe or friend? Cancer Cell Int. 2021, 21, 184. [Google Scholar] [CrossRef]
  188. Sauer, S.; Reed, D.R.; Ihnat, M.; Hurst, R.E.; Warshawsky, D.; Barkan, D. Innovative Approaches in the Battle Against Cancer Recurrence: Novel Strategies to Combat Dormant Disseminated Tumor Cells. Front. Oncol. 2021, 11, 659963. [Google Scholar] [CrossRef]
Figure 1. Reciprocal regulation of CAF-secreted cytokines/chemokines and the tumor microenvironment (TME). The crosstalk between CAFs and TME contributes to tumor progression through three main mechanisms: (1) The behavior of CAFs can be influenced by cytokines produced by tumor cells. In turn, these cytokines stimulate CAFs to secrete pro-tumorigenic factors, which contribute to the tumorigenic process. (2) By producing and cross-linking ECM components, CAFs regulate the structure and function of the ECM to support tumor progression and metastasis. (3) CAF-derived factors play a regulatory role in immune cells, including tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), and dendritic cells (DCs). Together, these factors establish a tumor immune microenvironment (TIME) that is conducive to tumor growth.
Figure 1. Reciprocal regulation of CAF-secreted cytokines/chemokines and the tumor microenvironment (TME). The crosstalk between CAFs and TME contributes to tumor progression through three main mechanisms: (1) The behavior of CAFs can be influenced by cytokines produced by tumor cells. In turn, these cytokines stimulate CAFs to secrete pro-tumorigenic factors, which contribute to the tumorigenic process. (2) By producing and cross-linking ECM components, CAFs regulate the structure and function of the ECM to support tumor progression and metastasis. (3) CAF-derived factors play a regulatory role in immune cells, including tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), and dendritic cells (DCs). Together, these factors establish a tumor immune microenvironment (TIME) that is conducive to tumor growth.
Cancers 15 02513 g001
Figure 2. The ratio of p38 signaling to ERK determines whether cancer cells enter a dormant state or undergo proliferation. Various CAF-secreted factors can induce cancer cells to enter a dormant state, which is characterized by cell cycle arrest through upregulation of p21, as well as increased autophagy and inhibition of Wnt signaling. On the other hand, CAFs can secrete factors to promote the awakening of dormant cancer cells by restoring the cell cycle and promoting the Wnt signaling pathway. In addition, CAFs can promote the escape of dormant cancer cells through the synthesis and remodeling of the ECM. These processes promote cytoskeletal reorganization, β1 and α5β1 integrin signaling, stromal stiffness, and the adhesion of tumor cells to the ECM, ultimately leading to dormant cell escape. Some CAF-secreted factors, such as IL-6 and IGF-2, play a dual role.
Figure 2. The ratio of p38 signaling to ERK determines whether cancer cells enter a dormant state or undergo proliferation. Various CAF-secreted factors can induce cancer cells to enter a dormant state, which is characterized by cell cycle arrest through upregulation of p21, as well as increased autophagy and inhibition of Wnt signaling. On the other hand, CAFs can secrete factors to promote the awakening of dormant cancer cells by restoring the cell cycle and promoting the Wnt signaling pathway. In addition, CAFs can promote the escape of dormant cancer cells through the synthesis and remodeling of the ECM. These processes promote cytoskeletal reorganization, β1 and α5β1 integrin signaling, stromal stiffness, and the adhesion of tumor cells to the ECM, ultimately leading to dormant cell escape. Some CAF-secreted factors, such as IL-6 and IGF-2, play a dual role.
Cancers 15 02513 g002
Figure 3. Strategies to fight cancer recurrence. Targeting dormant cancer cells is a promising approach for preventing cancer recurrence. (1) Awakening dormant cancer cells and making them sensitive to treatment. This can be achieved by either blocking the secretion of dormant drivers by CAFs or by restoring cell proliferation signals. (2) Maintaining the permanent dormancy of cancer cells to prevent regeneration. This can be achieved by inhibiting the CAF-mediated dormancy escape pathway. By targeting these dormant cells, we can prevent cancer recurrence and improve patient outcomes.
Figure 3. Strategies to fight cancer recurrence. Targeting dormant cancer cells is a promising approach for preventing cancer recurrence. (1) Awakening dormant cancer cells and making them sensitive to treatment. This can be achieved by either blocking the secretion of dormant drivers by CAFs or by restoring cell proliferation signals. (2) Maintaining the permanent dormancy of cancer cells to prevent regeneration. This can be achieved by inhibiting the CAF-mediated dormancy escape pathway. By targeting these dormant cells, we can prevent cancer recurrence and improve patient outcomes.
Cancers 15 02513 g003
Table 1. Agent targeting CAF-secreted cytokines/chemokines that maintain or awaken cancer cell dormancy.
Table 1. Agent targeting CAF-secreted cytokines/chemokines that maintain or awaken cancer cell dormancy.
FunctionTargetAgentMechanismClinical TrialRef.
Maintain
Dormancy
uPARATN-292Blocking uPA/uPAR interaction to inhibit ERK signaling; preventing the reawakening of DCCsPreclinical development[135]
uPAWX-671Phase II
(NCT00499265)
[137]
TGF-β1SRK-181A selective antibody against TGF-β1Phase I
(NCT04291079)
[139,141,142]
TGF-βRILY2157299Inhibitor of TGF-βRI kinase, blocking the TGF-β1-induced dormancy escape signaling pathwayPhase Ib/II
(NCT02734160)
[143,144,145]
Ki26894Preclinical development[146]
COX-2SulindacNon-steroidal anti-inflammatory molecules that may inhibit inflammation-mediated escape from dormancyPhase III
(NCT01349881)
[147,148,149]
CelecoxibPhase III
(NCT01150045)
[152,153]
α5β1VolociximabInhibition of ECM remodeling involved in dormant escapePhase Ib
(NCT00654758)
[163]
THR-687Phase II
(NCT05063734)
[164]
β1 integrinOS2966Inhibition of ECM remodeling involved in dormant escapePhase I
(NCT04608812)
[165]
LOXBAPNInhibit the organization of collagen fibers; target the microenvironment that supports dormant awakeningPreclinical development[168,169]
LOXL2SimtuzumabInhibit the organization of collagen fibers; target the microenvironment that supports dormant awakeningPhase II
(NCT01472198)
Phase II
(NCT01479465)
[170,171]
TGF-β1/LOXL2EGCGInhibition both of TGF-β1 signaling and collagen accumulationPreclinical development[173]
TSP-1ProsaposinEnhancing the dormant driver as well as the microenvironmentsPreclinical development[175]
TGF-β25-AZA with ATRARestores the NR2F1-induced dormant program and induces TGF-β2, thereby inhibiting metastasis by restoring TGF-β-SMAD4 signalingPreclinical development[176]
Awaken
Dormancy
TGF-β2AP 12009An antisense oligonucleotide (ASO) targeting TGF-β2 mRNA; blocking the dormancy driverphase I/II
(NCT00844064)
[179]
DKK-1DKN-01A humanized antibody blocks DKK-1 activity and restores Wnt signaling and cell proliferationphase I
(NCT02013154)
phase II
(NCT04363801)
phase II
(NCT05480306)
[180,181,182]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Cheng, S.-H.; Chiou, H.-Y.C.; Wang, J.-W.; Lin, M.-H. Reciprocal Regulation of Cancer-Associated Fibroblasts and Tumor Microenvironment in Gastrointestinal Cancer: Implications for Cancer Dormancy. Cancers 2023, 15, 2513. https://doi.org/10.3390/cancers15092513

AMA Style

Cheng S-H, Chiou H-YC, Wang J-W, Lin M-H. Reciprocal Regulation of Cancer-Associated Fibroblasts and Tumor Microenvironment in Gastrointestinal Cancer: Implications for Cancer Dormancy. Cancers. 2023; 15(9):2513. https://doi.org/10.3390/cancers15092513

Chicago/Turabian Style

Cheng, Shih-Hsuan, Hsin-Ying Clair Chiou, Jiunn-Wei Wang, and Ming-Hong Lin. 2023. "Reciprocal Regulation of Cancer-Associated Fibroblasts and Tumor Microenvironment in Gastrointestinal Cancer: Implications for Cancer Dormancy" Cancers 15, no. 9: 2513. https://doi.org/10.3390/cancers15092513

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

Article Metrics

Back to TopTop