Next Article in Journal
G Protein-Coupled Receptor Kinase 2 Selectively Enhances β-Arrestin Recruitment to the D2 Dopamine Receptor through Mechanisms That Are Independent of Receptor Phosphorylation
Previous Article in Journal
The Genetic Background of Abnormalities in Metabolic Pathways of Phosphoinositides and Their Linkage with the Myotubular Myopathies, Neurodegenerative Disorders, and Carcinogenesis
Previous Article in Special Issue
Analysis of Acute and Chronic Methamphetamine Treatment in Mice on Gdnf System Expression Reveals a Potential Mechanism of Schizophrenia Susceptibility
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of the Transforming Growth Factor-β Signaling Pathway in Gastrointestinal Cancers

Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka 5458585, Japan
*
Author to whom correspondence should be addressed.
Biomolecules 2023, 13(10), 1551; https://doi.org/10.3390/biom13101551
Submission received: 21 September 2023 / Revised: 17 October 2023 / Accepted: 18 October 2023 / Published: 19 October 2023

Abstract

:
Transforming growth factor-β (TGF-β) has attracted attention as a tumor suppressor because of its potent growth-suppressive effect on epithelial cells. Dysregulation of the TGF-β signaling pathway is considered to be one of the key factors in carcinogenesis, and genetic alterations affecting TGF-β signaling are extraordinarily common in cancers of the gastrointestinal system, such as hereditary nonpolyposis colon cancer and pancreatic cancer. Accumulating evidence suggests that TGF-β is produced from various types of cells in the tumor microenvironment and mediates extracellular matrix deposition, tumor angiogenesis, the formation of CAFs, and suppression of the anti-tumor immune reaction. It is also being considered as a factor that promotes the malignant transformation of cancer, particularly the invasion and metastasis of cancer cells, including epithelial-mesenchymal transition. Therefore, elucidating the role of TGF-β signaling in carcinogenesis, cancer invasion, and metastasis will provide novel basic insight for diagnosis and prognosis and the development of new molecularly targeted therapies for gastrointestinal cancers. In this review, we outline an overview of the complex mechanisms and functions of TGF-β signaling. Furthermore, we discuss the therapeutic potentials of targeting the TGF-β signaling pathway for gastrointestinal cancer treatment and discuss the remaining challenges and future perspectives on targeting this pathway.

1. Introduction

Transforming growth factor-β (TGF-β) signaling is essential for the homeostasis of epithelial cells, stromal fibroblasts, and immune cells. A variety of cells produce and respond to TGF-β, which results in a complicated network that is closely related to tumor development during each process. Thus far, it has been accepted that TGF-β can have different, even paradoxical, roles according to cancer cell types and/or the tumor microenvironment (TME) [1]. In normal and precancerous cells, TGF-β mostly functions as a tumor suppressor by impairing cell growth, facilitating apoptosis, and sustaining genome stability [1]. In early cancer stages, TGF-β signaling represses the growth of tumor cells by inducing apoptosis. In contrast, in late-stage cancers, TGF-β is upregulated and converted into a central driver for tumor development and metastasis. The diverse activity of TGF-β signaling provides cancer-specific properties. Approximately 40% of genetic alterations in mediators of TGF-β signaling occur in gastrointestinal (GI) cancers [2,3,4]. Cancer cells that maintain the capacity of TGF-β signaling capitalize on platforms that induce plasticity, such as epithelial-to-mesenchymal transition (EMT) and stem-like self-renewal, which lead to composition and remodeling of the TME and immune evasion. These multifunctions regulate the progression of cancer cells, which is acknowledged as the “TGF-β paradox”, and have attracted great attention as a potential therapeutic target for GI cancers.
In this review, we summarize the current understanding of the various activations of TGF-β and its signaling pathways. We then highlight the recent progress and challenges in targeting TGF-β signaling and discuss the perspectives of TGF-β-targeting therapeutics that would be important in providing benefits from GI cancer therapies.

2. TGF-β Cell Biology

TGF-β generally exists as homodimers or heterodimers in the extracellular region. In the late 1970s, murine sarcoma virus-transformed cells were found to produce numerous growth factors, including TGF-β. TGF-β was discovered as a cytokine that induces normal fibrocyte transformation and anchorage-independent proliferation of fibroblast cell lines [5]. In recent decades, TGF-β has been generally acknowledged to be a multifunctional cytokine that regulates numerous important events in cancer cell activities, such as cell cycle progression, differentiation, adhesion, apoptosis, and immune tolerance [6,7]. Malignant cells can impair TGF-β-induced proliferative suppression by acquiring stimulating mutations, such as those of RAS, and mutations in tumor suppressor genes, such as p53 and retinoblastoma protein. TGF-β enhances forkhead box P3 gene expression [8] and reduces the gene expression of MHC class I-chain-related molecule A, interferon-gamma (IFNγ), natural cytotoxicity receptor 3 granzyme A/B, and perforin to repress immune activity [9,10,11]. In contrast, it increases the expression of interleukin (IL)-11 and metalloproteinase 9 to promote metastasis to certain organs [12,13]. TGF-β also facilitates angiogenesis by enhancing vascular endothelial growth factor (VEGF), connective tissue growth factor, and metalloproteinase 2 levels [14,15].
To elucidate TGF-β activity and targeting, it should be known that TGF-β is synthesized in the endoplasmic reticulum as part of a latent complex in the extracellular matrix (ECM) with no activity (Figure 1). It comprises a signal peptide in the large N-terminal protein named latency-associated peptide (LAP) and a short C-terminal mature peptide [16,17]. When the N-terminal signal peptide is removed, the pro-peptide accumulates into a disulfide-linked homodimer, and latent TGF-β is transformed into homodimeric ligands [18]. The large latent complex (LLC) consists of TGF-β, LAP, and latent TGF-β binding protein (LTBP). LTBP contributes to extracellular release, localization, and activation of latent TGF-β. Latent TGF-β is collected in the ECM and transforms into activated TGF-β under enzymatic and/or non-enzymatic activity. Activated TGF-β can only connect to the TGF receptor complex and then activate canonical and noncanonical TGF-β signal transduction. Latent TGF-β is activated by binding to integrins β1, β6, and β8 via the RGD (Arg Gly-Asp) sequence present in LAP. Meanwhile, LTBP binds to the ECM and is activated via mechanical tension. Integrins, such as αvβ1, αvβ3, αvβ5, αvβ6, and αvβ8, are the main activators of TGF-β ligand. Integrin family members have important roles in the detection and activation of TGF-β [19,20,21]. TGF-β function mediated by integrins (integrins αvβ6 and αvβ8) is crucial for tumorigenesis and immunity. Both αvβ6 and αvβ8 regulate TGF-β signaling via the RGD sequence present in LAP, depending on actin cytoskeleton-generated tensile strength [22]. In numerous cell types, TGF-β is secreted as a complex bound to LTBP and released outside the cells. Meanwhile, in mesenchymal stromal cells, regulatory T cells (Tregs), and vascular endothelial cells, TGF-β remains on the cell membrane surface-formed complex bound to glycoprotein-A repetitions predominant (GARP) via disulfide bonds. When TGF-β is stimulated by integrin αVβ8, it can activate adjacent cells, such as Tregs. In addition, LRRC33, a GARP-related molecule, is expressed in cells of the central nervous system [23]. LRRC33 is relevant to latent TGF-β and mediates TGF-β activity. A human neutralizing antibody targeting GARP has been developed and is expected to specifically regulate the immunosuppressive effects of Tregs.
Nowadays, it is considered that the functions of TGF-β are mainly divided into four categories: (1) inhibition of cell proliferation; (2) promotion of EMT; (3) interaction with TME; and (4) immunosuppression. TGF-β was discovered to suppress the growth of many cells, including epithelial cells, blood cells, lymphocytes, and vascular endothelial cells, in the mid-1980s, and its tumor-suppressing effects have attracted attention in relation to cancer for a long time.
Loss of the TGF-β tumor suppressive response plays an important role in developing human cancers. As a central player in TGF-β signal transduction, small mothers against decapentaplegic (SMAD) 4, which is also known as DPC4, which was detected as a tumor suppressor gene for pancreatic cancer (PC), is frequently mutated or deleted in GI cancer. On the contrary, in the mid-1990s, it was demonstrated that TGF-β promotes EMT, indicating that TGF-β enhances late-stage cancer progression [24].
TGF-β consists of a large group of organizationally linked cell regulatory proteins, which is called the TGF-β superfamily. The discovery of TGF-β family members and their signaling components has provided an understanding of the complicated biological functions of TGF-β. The TGF-β family comprises 33 members in mammals, such as TGF-β isoforms and activin, nodal and bone morphogenetic proteins (BMPs), and TGF-β family signaling contained in a variety of physiological processes. It is also associated with cancer development, including proliferation, differentiation, and migration [25]. TGF-β has three structurally similar mammalian genome-encoded isoforms, namely, TGF-β1, TGF-β2, and TGF-β3 [25,26]. All three isoforms have been said to elicit equivalent biological activities but also have different activated mechanisms [26]. According to the database from The Cancer Genome Atlas (TCGA), the TGF-β1 homodimer is the most widely examined isoform and is expressed in various kinds of human cancer. Moreover, the expression of TGF-β1 is the most closely associated with the TGF-β pathway compared with TGF-β2, and 3 [26]. Although other TGF-β superfamily members have similarly important roles in the regulation of biological procedures, they are not described much in this review.

3. TGF-β Signaling Pathways

3.1. The Receptor of the TGF-β Family

TGF-β signals are transmitted by the complex TGF-β receptor composed of TGF-β receptor type 1 (TGFBR1, also called ALK-5) and receptor type 2 (TGFBR2). Both TGFBR1 and TGFBR2 have two paired serine-threonine kinase domains that phosphorylate serine and threonine in the cell. TGF-β binding to a ligand induces the construction of tetrameric receptor complexes containing TGFBR1 dimers and TGFBR2 dimers, which phosphorylate TGFBR1 on a specific Gly/Ser-rich “GS sequence (glycine-serine) region and result in transmitting signals to the intracellular region [25] (Figure 1). Other members of the TGF-β family either bind to their own receptors or compete with other TGF-β family receptors to transduce and regulate signals. Activin mainly binds and activates type 1 receptors called ALK-4 and ALK-7, while BMP mainly activates ALK-1, -2, -3, and -6.

3.2. Smad Signaling Pathways

Although there are many TGF-β ligands, fewer receptors and downstream effectors, such as SMAD proteins, regulate intracellular signaling. A hetero-tetrameric complex of TGFBR2 and TGFBR1 results in the TGFBR2 kinase domain phosphorylating TGFBR1 and subsequently activating SMAD2/3 [25]. Consequently, activated SMAD2/3 facilitates complexes connecting to SMAD4, generating a trimeric complex that is then translocated into the nucleus, where they can exhibit and inhibit target gene expressions (Figure 1) [27]. Downstream factors of the SMAD signal pathway, particularly Smad2/Smad3, are considered to be important regulators of the TGF-β pathway in tissue fibrosis and tumorigenesis. In contrast, TGF-β produced SMAD6/SMAD7 to serve as a negative mediator of TGF-β/SMAD signals, which is relevant to TGFBR1, leading to inhibition of the activation of SMAD2/3. Furthermore, SMAD7 provokes TGF-β signals, thereby disrupting the structure of the SMAD-DNA conjugate in the nucleus [28] and blocking the translocation of the SMAD2/SMAD4 conjugate [29].
TGF-β can also introduce SMAD-independent noncanonical signaling pathways as well as the canonical SMAD pathway (Figure 1) [30]. TGFBR1 stimulates RHO small GTPases, which mediate the function of RHO-associated protein kinase and LIM kinase, thereby remodeling the actin cytoskeleton [31]. Similarly, TGFBR2 activates the cell polarity regulator PAR6, which mediates tight junctions and the migration/motility of cancer cells [31]. TGF-β also directly phosphorylates TGF-β-activated kinase 1 (TAK1), which results in prompt c-Jun NH2-terminal kinase (JNK), p38 mitogen-activated protein kinase (MAPK), and nuclear factor kappa-B (NF-κB) pathways. The PI3K/AKT/mTOR pathway is also induced as the downstream signaling pathway for noncanonical signals [31]. Furthermore, TGF-β activates Src homology domain 2-containing protein (SHC) and then stimulates growth factor receptor-binding protein 2 (GRB2) and son of sevenless (SOS), which leads to initiating the RAS, RAF, extracellular signal-regulated kinase (ERK), and MEK pathways [31]. The RAS-responsive element-binding protein 1 (RREB1) serves as a connection between the RAS and TGF-β pathways, which participate in the synchronized induction of a fibrogenic EMT pathway [32]. There is crosstalk between the canonical and noncanonical TGF-β pathways, and it is controlled by tyrosine kinase receptors [33]. Moreover, these pathways are also stimulated by TGF-β through inducing platelet-derived growth factor in an autocrine or paracrine manner [34].

4. Role of TGF-β Signaling in the Carcinogenesis and Progression of GI Cancer

TCGA dataset analysis shows that genetic alterations in TGF-β signaling are represented in about 30% of GI tumor specimens; in particular, mutations of TGFBR2 and SMAD4 are common in PC and colorectal cancers (CRCs) (Table 1) [2]. In PC, novel evidence suggests that several mutations in the TGF-β signaling genes, such as TGFBR1, TGFBR2, SMAD2, and SMAD4 genes, occur [35]. Notably, SMAD deletion or mutation is identified in 60% of PC patients, and an increased SMAD mutation drives early carcinogenesis and dissemination [36]. In CRC, SMAD4 is one of the most frequently mutated genes based on a series of high-throughput analyses. A recent study utilizing next-generation sequencing (NGS) including 123 non-MSI-high mCRC patients demonstrated an about 20% mutation occurrence of SMAD4 [37]. TGFBR2 mutations are also often shown in CRC [38]. Remarkably, TGFBR2 mutations are shown in 80–90% of CRC with microsatellite instability [39]. Carcinoembryonic antigen-associated cell adhesion molecule 5 (CEACAM5), a highly glycosylated protein of the CEACAM family, reduces the expression of TGF-β pathway members (TGFBR2, SMAD4, and SPTBN1), which alters the colonic microbiome to promote CRC [40]. By contrast, SMAD2 mutations have been reported in hepatocellular carcinoma (HCC) [41]. For SMAD4, about 70% of esophageal adenocarcinoma (EAC) associated with Barrett’s esophagus (BE) revealed loss of heterozygosity in a region involving SMAD2 and SMAD4 on chromosome 18q [42,43]. This loss of heterozygosity may be an early event in neoplastic transformation because it occurs in approximately 30–70% of patients with precancerous BE [43]. Another mechanism for reducing SMAD4 expression is the methylation of the SMAD4 promoter in both BE and EAC [44]. Genomic alterations of SMAD4 are more frequent in EAC (24%) than in esophageal squamous cell carcinoma (ECC) (8%). The noncanonical TGF-β and JNK signaling axis was hyperactivated in EC patients, and the genes mediated by these pathways were also upregulated in EC, suggesting new treatment tools for this refractory cancer [45]. The frequency of genetic alterations in TGF-β pathway-related genes in GI cancers was summarized in Table 1.
Since the TGF-β signaling pathway has diverse tumorigenic effects, understanding its mechanisms in carcinogenesis will lead to better diagnostic and predictive tools as well as therapeutic approaches.

5. Role of TGF-β Signaling in Epithelial-Mesenchymal Transition

EMT is a phenomenon in which keratinocytes differentiate into mesenchymal cells and contribute to the initiation processes, migration, and invasion of cancer cells [58]. During EMT, the down-regulation of epithelial markers such as E-cadherin, occludins, and claudins is shown, whereas mesenchymal markers such as N-cadherin, vimentin, and fibronectin are up-regulated. These alterations result in reduced cell adhesion, loss of polarity, tight junctions, and a highly invasive mesenchymal phenotype. TGF-β has a potent role in initiating tumorigenesis and metastatic ability of cancer cells by inducing the EMT via the activation of transcription factors such as SLUG, SNAIL, ZEB1/2, DMNT, and TWIST (Figure 2) [59]. TGF-β can also induce EMT in non-canonical signaling, for instance, by fascinating cytoskeletal remodeling, which results in the phosphorylation of ERK [60]. MET (mesenchymal-epithelial transition) is the reverse process of EMT. EMT not only transforms epithelial cells into full-form mesenchymal cells (full EMT) but often also partially acquires the characteristics of interstitial cells through intermediate hybrid states (partial EMT state, P-EMT). Furthermore, cells that have undergone EMT may acquire epithelial-like cell phenotypes due to MET. TGF-β vigorously regulates the transition between P-EMT and full-EMT positions in cancer cells [61]. Furthermore, long-term TGF-β stimulation, as often seen during clinical cancer progression, can stabilize EMT, in contrast to the reversible EMT induced by short-term TGF-β stimulation. Long-term TGF-β stimulation activates not only the SMAD pathway but also mTOR signaling, which stabilizes cancer stem cell (CSC)-like properties and anticancer drug resistance [62].
Accordingly, TGF-β is closely related to the infiltration and metastasis of GI cancer through EMT. In this section, we review the relationship between TGF-β and EMT in GI cancer. For example, in PC, it is conceivable that TGF-β induces EMT from an early stage due to KRAS abnormalities and shifts from a tumor suppressor to a tumor promoter [63]. Alterations in signaling or gene expression that facilitate EMT promote the CSC population and, consequently, the tumor-initiating capacity. The inhibitor of DNA binding 1 (ID1) protein acts as a cell differentiation suppressor and growth stimulator. Transcriptional dysregulation of ID1 regulates the escape of pancreatic ductal adenocarcinoma (PDA) from TGF-β tumor suppression and decouples TGF-β-induced EMT from apoptosis, which contributes progenitor-like characteristics to PDA cells [64]. In EC, cell division cycle associated 7, one of the copy number amplification genes, was shown to facilitate the EMT process and metastasis of EC through TGF-β signaling activation via inhibiting SMAD7 mRNA transcription and enhancing SMAD4 mRNA transcription [65]. Epithelium-specific ETS transcription factor 1 (ESE1) belongs to the ETS-domain transcription factor superfamily. In PC, an ESE1/anterior gradient protein 2 axis negatively interacts with TGF-β signaling to mediate the EMT phenotype by functioning with EMT drivers such as ZEB1/2 [66]. In addition, TGF-β1 upregulates acyl-CoA synthetases 3, leading to promoting ATP and suppressing NADPH, thereby maintaining redox homeostasis and facilitating the EMT and metastasis of CRC cells [67].

6. Role of TGF-β Signaling in the Tumor Microenvironment

During cancer development, the surrounding components in TME co-exist with cancer cells via paracrine and autocrine mechanisms. These active signaling networks in the TME confer cancer advancement, metastasis, and chemoresistance [68]. Hence, the TME is predicted to be useful as a tool for cancer therapy because of the enlarged and precise acknowledgement of the heterogeneity and complexity of cancer cells. The TME is organized into a variety of cell types, such as cancer-associated fibroblasts (CAFs), immune-related cells, endothelial cells, adipocytes, stromal cells, and the ECM, as well as a wide range of cytokines [69,70]. TGF-β is a potent cytokine that enables it to mediate the activity of most cells existing in the TME, which results in producing ECM components, stimulating CAFs, and facilitating angiogenesis (Figure 3).
Inflammation is a biological defensive reaction to a variety of damaging stimuli, including injured cells, pathogens, and toxic factors [71]. Chronic inflammation facilitates numerous pathological situations, such as tissue fibrosis [72]. The fibrotic procedure is characterized by altered mesenchymal transition and the growth of fibroblasts. One of the features of fibrosis is the transformation of fibroblasts into myofibroblasts, which express α-smooth muscle actin (α-SMA) and dysregulate the ECM. TGF-β has been shown to be crucial for the initiation of the fibrotic response and for the activity of the stroma surrounding cancer cells. TGF-β directly stimulates target cells to synthesize and transport ECM components such as fibronectin, laminins, and collagens [73]. TGF-β stimulation induces HCC cells to release an excess of soluble factors, which lead to target stromal cells. Connective tissue growth factor (CTGF), which is produced by TGF-β-stimulated HCC cells, promotes fibrosis, which results in enhancing tumor size in the in vivo model of HCC [74]. In scirrhous GC, TGF-β secreted from gastric fibroblasts affects the invasion ability of EMT and, in turn, prompts peritoneal metastasis [75].
As key components of the TME, CAFs are initiated by the activation or transformation of precursor cells in tumor tissues [76]. Activated CAFs provide a favorable environment to promote ECM remodeling, cancer cell growth, invasion, angiogenesis, metabolic reprogramming, stemness, and tumor metastasis. TGF-β signaling is a pivotal trigger of CAF function and construction and contributes to maintaining the morphological features and functional phenotypes of CAFs. In PCs, CAFs are identified as distant from primary tumors with low α-SMA expression and high levels of IL6, and myofibroblastic CAFs are located adjacent to tumor sites with high α-SMA expression and demonstrate a robust TGF-β reaction [77]. In stroma-abundant PC, branched chain α-ketoacid (BCKA) was positively related to the catabolism initiated by the metabolic reprogramming of CAFs and PC cells [78]. The TGF-β/SMAD5 axis induced the incorporation of ECM, which provided the amino acid precursors in CAFs, leading to BCKA secretion [78]. In HCC, CAFs stimulated the construction of vascular mimicry, which was significantly diminished when TGF-β signaling was eliminated [79]. In GC, several studies have demonstrated the relationship between TGF-β and stemness. In a recent study, fibroblasts activated by Helicobacter pylori promoted the differentiation of gastric epithelial cells, which is associated with CSCs. This result is partially dependent on TGF-β signaling and promotes tumorigenesis [80]. CAFs have been found to facilitate the aggressive phenotypes of scirrhous GC cells, which involve adhesion to mesothelial cells [81], migration and invasion [82], peritoneal dissemination [83], and the CSC phenotype [84]. Using a comprehensive study on the basis of large-scale clinical samples, including 18 datasets of 2320 patients with CRC from the TCGA and GEO databases, a recent study established the CAF-derived long non-coding RNA (CAFDL) signature in the TCGA-CRC training set, evaluated its predictive significance, and demonstrated that CAFDL was significantly positively associated with TGF-β1 [85]. TGF-β was shown to suppress lymphangiogenesis via inhibiting the expression of collagen and calcium-binding EGF domain-1 [86], a molecule implicated in the proteolytic activity and maturation of VEGF-C [87], showing a new mechanism by which a deficit of TGF-β signaling facilitates metastasis of CRC [86]. Accurate and effective therapy targeting the TGF-β pathway in CAFs is an assuring tool for GI cancers, and increasing attempts are ongoing to attain this purpose.

7. Role of TGF-β Signaling in the Immune System

TGF-β is also known as a crucial immunosuppressive cytokine [88]. Immunosuppression by TGF-β involves various immune cells, such as T cells, B cells, tumor-associated macrophages (TAMs), tumor-associated neutrophils, dendric cells (DCs), NK cells, and myeloid-derived suppressor cells (MDSCs) (Figure 3). TGF-β prompts macrophages to shift from an inflammatory (M1) to a tumor-trophic (M2) phenotype to be transformed into TAM, thus producing an immunosuppressive microenvironment [89,90]. In a similar way, TGF-β produces an N2 neutrophil phenotype, which facilitates cancer development [91]. Furthermore, up-regulation of TGF-β promotes immunosuppressive activity by translating naïve T cells to Treg cells [92]. TGF-β can inhibit the activation and cytotoxicity of NK cells and suppress their IFN-γ secretion [7]. IFN-γ is an activator of macrophages and promotes NK cells and neutrophils. As well, TGF-β represses the development of DCs and helper T cells [92]. Moreover, TGF-β can mediate the activation, proliferation, and apoptosis of B cells and regulate antibody switching in B cells [93]. Meanwhile, TGF-β can induce the expression of forkhead box P3, which leads to the production and differentiation of Treg from CD4+ T cells through IL-2 signaling. It also prompts differentiation into IL-17-producing helper T cells (Th17 cells) in the presence of IL-6 and IL-21 [92,94]. TGF-β produced by Tregs suppresses the differentiation and growth of effector T cells, has a role in inhibiting the function and development of cytotoxic CD8+ T cells by repressing the tumor antigen processing and presentation of DCs, and inhibits CD8+ T cell proliferation through blocking the expression of IFN-γ and IL2 [95,96,97]. Thus, TGF-β is known to regulate T cell differentiation depending on the existence of inflammatory cytokines. The blockade of the TGF-β pathway is an efficient tool in the treatment of cancer, which attenuates Treg-mediated anti-tumor activity, enhances T cell cytotoxicity, and prompts T cell movement into the center of the tumor, leading to inducing robust anti-tumor immunity and tumor suppression (Figure 3) [98]. In CRC, macrophage polarization to M2 phenotypes of TAMs via the TGF-β pathway was induced by the release of Collagen Triple Helix Repeat Containing 1 (CTHRC1), which led to liver metastasis in vivo models [99]. A previous study described that CD4+ CD25+ Tregs-derived IL-10 has been shown to inhibit microbially-induced colonic inflammation and CRC development [100]. In PC, TAMs promote migration and invasion of cancer cells by inducing EMT via the TGF-β-SMAD2/3/4-Snail signaling axis [101]. In EC patients, the effector immune responses correlated with the CD3+CD8+ T lymphocytes are disordered. CD8+ T lymphocyte repressed function could be associated with aberrant miR-21 production, which could be potentially involved in a pivotal target for immunotherapeutic strategies [102]. Collectively, these findings further imply the complications of the TGF-β pathway in the immune evasion of GI cancer and provide a pivotal role for immune cell modulation in the tumor-facilitating efficiencies of the TGF-β signaling pathway in GI cancer.

8. Role of Non-Coding RNAs Involved in TGF-β Signaling

The majority of RNA-transcribed but not-encoded proteins can be categorized as non-coding RNAs (ncRNAs) [103]. Depending on the shape, length, and situation, ncRNAs have been subdivided into diverse classes, such as microRNA (miRNA), long ncRNA (lncRNA), circular RNA (circRNA), and PIWI-interacting RNA (piRNA). They can modulate transcription factors to bind to promoters and thus regulate the progression of a variety of malignant tumors [103]. Growing evidence has shown that TGF-β signaling in tumor progression is related to the alteration of ncRNA regulation. In CRC, LINC00941 has tumorigenic functions and is highly expressed. Moreover, it facilitates the metastasis of CRC by activating the TGF-β/SMAD2/3 signaling pathway and preventing SMAD4 protein degradation [104]. LncRNA SBF2-AS1 regulates TGFBR1-mediated signaling by sponging miR-140-5p, which facilitates HCC progression [105]. LncRNA MBNL2-AS1, which forms a competing endogenous RNA network with miR-424-5p and SMAD7, downregulated the TGF-β/EMT pathway, thereby inhibiting the invasion ability of gastric cancer (GC) cells [106]. Circ GFRA1 (hsa_circ_005239), which originated from the GFRA1 (GDNF family receptor alpha1) on chromosome 10, is a type of cancer-related circRNA. The up-regulation of circGFRA1 significantly prompts tumor growth and intra-hepatic metastases in HCC. Meanwhile, similar binding sequences were identified between circGFRA1 and miR-498, and these expressions are reversely associated in HCC cells [107].
Abundant studies suggest that ncRNAs are mediated in response to TGF-β-induced EMT in GI cancer. For example, lncRNAs can mediate EMT advancement by phosphorylating TGF-β signaling in PC. For instance, lnc00462 facilitates PC invasion and metastasis via the miR-665/TGFBR1-TGFBR2/SMAD2/3 pathway [108]. As well, miR-492 induces EMT by stimulating TGF-β/SMAD3 signaling and the expression of NR2C1, a member of the steroid/thyroid hormone receptor superfamily, suggesting the significance of miR-492-based treatments for PC [109]. A recent paper described that the activation of CXCL12/CXCR7 promoted miR-146a-5p and miR-155-5p, derived from the exosome in CRC cells, to enhance the activation of CAFs via the JAK2-STAT2/NF-κB signaling pathway and further promote EMT and CRC metastasis to the lung [110]. Macrophage-derived exosomal miR-501-3p was shown to facilitate PC metastasis through the suppression of TGFBR3, which results in the activation of the TGF-β signaling pathway [111]. The interaction of ncRNAs and TGF-β signaling may provide a latent new target and promising diagnostic and prognostic markers for GI cancer. Table 2 summarizes non-coding RNAs involved in TGF-β signaling in GI cancers.

9. Discussion and Future Perspective

According to the World Health Organization, approximately 5.5 million new cases of GI cancer worldwide have been reported in 2020 [175]. Over 14 million new cancer cases are diagnosed annually, with 8.2 million deaths. Given the high mortality rates, the capacity to overcome GI cancer is urgently needed [176]. Many therapy strategies are available for patients with GI cancer, such as surgery, chemotherapy, and chemoradiotherapy [177]. However, 5-year overall survival (OS) remains unsatisfactory for patients with advanced stages, and effective methods to evaluate the prognosis of patients with GI cancer are still lacking [178]. Understanding the molecular mechanisms controlling the heterogeneity of GI cancer could lead to new therapeutic targets. We have evidence that relates TGF-β signaling to cancer biology, from tumorigenesis to TME and immune evasion in GI cancer. The importance of TGF-β signaling in GI cancer is represented by the high incidence of genetic alterations in the TGF-β pathways. Due to its pleiotropic activity as a tumor suppressor and active promoter of tumor development and metastasis, targeting TGF-β could provide new and promising prospects in GI cancer treatment.
Therapeutic strategies have normally been distinguished into three types: ligand, ligand-receptor binding, and intracellular transduction to disrupt TGF-β signaling. These strategies include the use of neutralizing antibodies, TGF-β ligand traps, small-molecule TGF-β inhibitors, antisense oligonucleotides, and vaccine-based therapy. Thus far, non-coding RNAs, kinase inhibitors, and natural compounds are familiar approaches that have been used for targeting TGF-β in clinical trials (Table 3). The remarkable capacities of anti-tumor agents with TGF-β-neutralizing antibodies and selective small-molecule TGF-β receptor kinase inhibitors have been proven in recent decades. In contrast, although TGF-β-targeted therapies have been efficacious in in vitro and in vivo examinations, the application of these pharmacological agents for approved subsequent clinical trials was unsatisfactory because of their systemic efficacy on normal tissues of TGF-β, which causes undesirable side effects such as fatigue, gingival bleeding, headache, epistaxis, and TGF-β-related skin lesions, including reversible cutaneous keratoacanthomas/squamous-cell carcinomas [179,180]. In particular, some TGF-β receptor inhibitors and neutralizing antibodies have caused cardiac toxicity in animal models. Although inhibition of TGF-β signaling has meaningful clinical capacity for treating advanced-stage cancers and metastasis, TGF-β blocking alone may promote tumor proliferation [181]. For instance, blocking TGF-β represses ECM deposition and decompresses blood vessels, leading to the recovery of blood perfusion. Enhanced blood perfusion provides a source of nutrients and oxygen to cancer cells and enhances tumor proliferation [181], which could enhance the ability of cancer cells to metastasize. Thus, the use of these inhibitors as monotherapies is still challenging, but combining them with other medications may be considerably more attractive. In PC, a recent phase II clinical trial presented that galunisertib combined with gemcitabine prolonged the outcome of patients at a dose of 300 mg/day [182]. Galunisertib also presented a satisfactory safety profile and improved OS when used in combination with sorafenib, implying its usefulness as a second-line therapy for HCC in a clinical phase II study [183]. In the meantime, in advanced rectal cancer, galunisertib improved the complete response rates to neoadjuvant chemoradiotherapy [184]. Vaccine-based therapy is an emerging new tool for GI cancer therapy. Vigil (Gemogenovatucel-T, FANG™, IND14205) is an autologous compound that consists of a plasmid encoding granulocyte-macrophage colony-stimulating factor and a bifunctional short hairpin RNA targeting furin convertase, which leads to suppressed TGF-β1 and TGF-β2 function. A non-randomized phase I trial evaluating the safety profile of Vigil in HCC patients who are surgically unresectable is now underway (NCT01061840).
Given that the inhibition of the TGF-β pathway induces tumor responses to immune checkpoint inhibitors (ICIs), including programmed death-1 (PD-1) or PD-1 ligand (PD-L1) antibodies, antitumorigenic immunotherapies have been developed to enhance the tumor-inhibitory effect of anti-PD1 treatment by suppressing TGF-β signaling [185]. Furthermore, stemness, EMT, and tumor resistance, which are closely related to TGF-β, are shown to be impaired by dual inhibition of TGF-β/PD-L1 [186]. Therefore, a variety of clinical studies are validating the effect of combining ICIs and TGF-β-targeting agents (Table 2) [92]. For example, the feasibility and usefulness of vactosertib or galunisertib combined with durvalumab, a monoclonal PD-L1 antibody, are under examination in clinical studies of CRC, PC, and GC [185]. Subsequently, M7824, a bispecific fusion peptide against PD-L1/TGF-β, localizes to TME, activates the immune function of Tregs, produces NK and CD8 + T cell activity, and increases the density of anti-tumor type 1 macrophages [187,188]. M7824-treated patients with recurrent gastro-esophageal or GC disease revealed 16% of the objective response rate (ORR) and 26% of the disease control rate. Responses are likely to depend on a high level of TGF-β in the tumor, regardless of the PD-L1 expression profile (NCT02699515) [189]. Monotherapy with SHR-1701, a fusion protein that includes an anti-PD-L1 mAb to which TGFBR2 ORR and 12-month OS of 20% and 54.5%, respectively, among cancers (NCT3710265) [190].
Thus far, no TGF-β-targeting agents have been approved by the FDA for GI cancer therapy. Although the dual blockage of ICIs and TGF-β signaling is well established, several challenges remain in terms of antagonizing TGF-β signaling in tumorigenesis. Limited knowledge of the interaction between proteins comprised in TGF-β secretion, activation, and signal crosstalk is insufficient for utilizing current TGF-β blockades in adjuvant combination therapies [191]. Indeed, the lack of dosing schedules and their dual activity in carcinogenesis make the use of TGF-β antagonists for the treatment of GI cancer challenging [192]. To promote efficient therapies that focus on tumor-promoting functions, screening genetic variants and the TME of each patient would provide great benefits in predicting the effectiveness of TGF-β antagonists. As such, the detection of dependable predictive biomarkers would provide a better strategy for patient choice. Alterations in the TME, such as matrix rigidity and tissue stiffness, can influence the effectiveness of TGF-β [193]. A better understanding of tissue mechanics in the TME may make it possible to provide the cellular response to TGF-β [194]. At present, the most attractive candidates for TGF-β signaling are traditional biomarkers such as SMAD2, SMAD4, and TGFBR2, as shown by several previous studies [195,196,197]. Meanwhile, the identification of blood-based biomarkers is important to select patients who will respond to TGF-β-targeting therapy and to decide the timing of the response of the patients to TGF-β inhibitors to increase therapeutic usefulness. In EC, TGF-β1 levels markedly increased after 40 Gy irradiation in patients who had radiation-induced pneumonitis. In contrast, patients with non-radiation-induced pneumonitis showed steady TGF-β1 levels during and after radiation therapy [198]. To date, clinical studies have assessed the value of the level of TGF-β as a diagnostic and prognostic biomarker in GI cancer (NCT01246986, NCT05500027, NCT04729725, and NCT04122937). Recent studies demonstrate that the secretion of TGF-β in small extracellular vesicles (sEV-TGF-β) contributes to tumor growth, development, and metastasis. Moreover, sEV-TGF-β is also closely related to immune evasion [199]. Interestingly, in non-small-cell lung cancer, the expression of extracellular vesicle (EV) TGF-β was associated with the prediction of efficacy of ICIs and shorter survival, which resulted in a higher accuracy than the existing circulating TGF-β and tissue PD-L1 in patients who are treated with ICIs, suggesting EV TGF-β as a latent predictive biomarker in immunotherapies involving dual TGF-β and PD-L1 blockade [200].
As previously described, reducing toxicity would be a key problem in applying the TGF-β blocking agents for clinical usage [201,202]. In melanoma, specifically blocking the surface-bound TGF-β on Tregs repressed anti-tumor cytotoxicity regulated by CD8+ T cells ex vivo [98]. Thus, a targeted disorder of tumor-enabling signaling within the TME or combining TGF-β antagonists with immuno-mediating therapies could be therapeutic strategies that conquer adverse events related to TGF-β blockade. Additionally, new small-molecule inhibitors of TGFBR1 with controllable toxicity are being developed. Based on the results that TGF-β blockade enhances response to radiotherapy [184], the use of TGF-β-targeted agents combined with chemoradiation could theoretically lead to the approval of TGF-β antagonists for clinical utility. For patients with PC, TGF-β-specific T cells showed significant clinical advantage and prolonged survival after the combined use of radiotherapy and ICIs [203]. Most recently, a comprehensive meta-analysis demonstrated an increase in progression-free survival, OS, and ORR when chemotherapy or radiotherapy was added to TGF-β antagonists in solid tumors [204]. Advancement in understanding the collective activities of TGF-β in specific tumor subtypes and various stages of GI cancer, identification of dependable and precise biomarkers, and evaluation of proper methods to reduce adverse events may provide beneficial insights into the clinical determinations in selecting patients who are sensitive to TGF-β-targeted therapies.

10. Conclusions

It is acknowledged that dysregulation of the TGF-β pathway is closely correlated with the progression of various GI cancers. The pleotropic activity of TGF-β is exploited by its capacity to regulate diverse biological processes, including EMT, angiogenesis, immune evasion, and malignant progression at a late stage. Nevertheless, a comprehensive understanding of the molecular mechanisms underlying its different roles in the advancement of GI cancers, as well as identifying reliable biomarkers and TGF-β antagonists exhibiting greater specificity and minimum toxicity, is essential in the development of more useful treatment approaches, possibly in combination with other antitumor agents, to enhance therapeutic efficacies and prolong the survival of patients with GI cancers.

Author Contributions

Conceptualization, T.M. and M.Y.; writing—original draft preparation, T.M. and M.Y.; writing—review and editing, T.M. and M.Y.; supervision, M.Y.; funding acquisition, M.Y. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the KAKENHI (Grant-in-Aid for Scientific Research), No. 18H02883.

Data Availability Statement

The data presented in this study are available in this article.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

Abbreviation Definition
TGF-βtransforming growth factor-β
TMEtumor microenvironment
GIgastrointestinal
EMTepithelial-to-mesenchymal transition
IFN-γinterferon-γ
ILinterleukin
VEGFvascular endothelial growth factor
ECMextracellular matrix
LAPlatency-associated peptide
LLClarge latent complex
LTBPlatent TGF-β-binding proteins
RGDArg-Gly-Asp
Tregsregulatory T cells
GARPglycoprotein-A repetition predominant Tregs
SMADsmall mothers against decapentaplegic
PCpancreatic cancer
TGFBR2TGF-β receptor type 2
TGFBR1TGF-β receptor type 1
TAK1TGF-β-activated kinase 1
JNKc-Jun NH2-terminal kinase
MAPKmitogen-activated protein kinase
NF-κBnuclear factor kappa-B
SHCSrc homology domain 2-containing protein
GRB2growth factor receptor-binding protein 2
SOSson of sevenless
ERKextracellular signal-regulated kinase
RREB1RAS-responsive element-binding rotein 1
PDGFplatelet-derived growth factor
CRCcolorectal cancer
NGSnext-generation sequencin
CEACAM5carcinoembryonic antigen-associated cell adhesion molecule 5
EACesophageal adenocarcinoma
BEBarrett’s esophagus
ECCesophageal squamous cell carcinoma
NcRNAnon-coding RNA
LncRNAlong non-coding RNA
CircRNAcircular RNA
HCChepatocellular carcinoma
METmesenchymal-epithelial transition
CSCcancer stem cell
ID1DNA binding 1
PDApancreatic ductal adenocarcinoma
ESE1ETS transcription factor 1
CAFscancer-associated fibroblasts
α-SMAα-smooth muscle actin
BCKAbranched chain α-ketoacid
GCgastric cancer
CAFDLCAF-derived long non-coding RN
OSoverall survival
ORRobjective response rate

References

  1. Tzavlaki, K.; Moustakas, A. TGF-β Signaling. Biomolecules 2020, 10, 487. [Google Scholar] [CrossRef] [PubMed]
  2. Korkut, A.; Zaidi, S.; Kanchi, R.S.; Rao, S.; Gough, N.R.; Schultz, A.; Li, X.; Lorenzi, P.L.; Berger, A.C.; Robertson, G.; et al. A Pan-Cancer Analysis Reveals High-Frequency Genetic Alterations in Mediators of Signaling by the TGF-β Superfamily. Cell Syst. 2018, 7, 422–437.e7. [Google Scholar] [CrossRef] [PubMed]
  3. Chen, J.; Zaidi, S.; Rao, S.; Chen, J.S.; Phan, L.; Farci, P.; Su, X.; Shetty, K.; White, J.; Zamboni, F.; et al. Analysis of Genomes and Transcriptomes of Hepatocellular Carcinomas Identifies Mutations and Gene Expression Changes in the Transforming Growth Factor-β Pathway. Gastroenterology 2018, 154, 195–210. [Google Scholar] [CrossRef] [PubMed]
  4. Waddell, N.; Pajic, M.; Patch, A.M.; Chang, D.K.; Kassahn, K.S.; Bailey, P.; Johns, A.L.; Miller, D.; Nones, K.; Quek, K.; et al. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature 2015, 518, 495–501. [Google Scholar] [CrossRef] [PubMed]
  5. Roberts, A.B.; Anzano, M.A.; Lamb, L.C.; Smith, J.M.; Sporn, M.B. New class of transforming growth factors potentiated by epidermal growth factor: Isolation from non-neoplastic tissues. Proc. Natl. Acad. Sci. USA 1981, 78, 5339–5343. [Google Scholar] [CrossRef] [PubMed]
  6. Xie, F.; Ling, L.; van Dam, H.; Zhou, F.; Zhang, L. TGF-β signaling in cancer metastasis. Acta Biochim. Biophys. Sin. 2018, 50, 121–132. [Google Scholar] [CrossRef]
  7. Batlle, E.; Massague, J. Transforming Growth Factor-β Signaling in Immunity and Cancer. Immunity 2019, 50, 924–940. [Google Scholar] [CrossRef]
  8. Zhou, L.; Lopes, J.E.; Chong, M.M.; Ivanov, I.I.; Min, R.; Victora, G.D.; Shen, Y.; Du, J.; Rubtsov, Y.P.; Rudensky, A.Y.; et al. TGF-β-induced Foxp3 inhibits T(H)17 cell differentiation by antagonizing RORgammat function. Nature 2008, 453, 236–240. [Google Scholar] [CrossRef]
  9. Castriconi, R.; Cantoni, C.; Della Chiesa, M.; Vitale, M.; Marcenaro, E.; Conte, R.; Biassoni, R.; Bottino, C.; Moretta, L.; Moretta, A. Transforming growth factor beta 1 inhibits expression of NKp30 and NKG2D receptors: Consequences for the NK-mediated killing of dendritic cells. Proc. Natl. Acad. Sci. USA 2003, 100, 4120–4125. [Google Scholar] [CrossRef]
  10. Thomas, D.A.; Massague, J. TGF-β directly targets cytotoxic T cell functions during tumor evasion of immune surveillance. Cancer Cell 2005, 8, 369–380. [Google Scholar] [CrossRef]
  11. Trotta, R.; Dal Col, J.; Yu, J.; Ciarlariello, D.; Thomas, B.; Zhang, X.; Allard, J., 2nd; Wei, M.; Mao, H.; Byrd, J.C.; et al. TGF-β utilizes SMAD3 to inhibit CD16-mediated IFN-gamma production and antibody-dependent cellular cytotoxicity in human NK cells. J. Immunol. 2008, 181, 3784–3792. [Google Scholar] [CrossRef]
  12. Pollari, S.; Leivonen, S.K.; Perala, M.; Fey, V.; Kakonen, S.M.; Kallioniemi, O. Identification of microRNAs inhibiting TGF-β-induced IL-11 production in bone metastatic breast cancer cells. PLoS ONE 2012, 7, e37361. [Google Scholar] [CrossRef] [PubMed]
  13. Wiercinska, E.; Naber, H.P.; Pardali, E.; van der Pluijm, G.; van Dam, H.; ten Dijke, P. The TGF-β/Smad pathway induces breast cancer cell invasion through the up-regulation of matrix metalloproteinase 2 and 9 in a spheroid invasion model system. Breast Cancer Res. Treat. 2011, 128, 657–666. [Google Scholar] [CrossRef] [PubMed]
  14. Goumans, M.J.; Liu, Z.; ten Dijke, P. TGF-β signaling in vascular biology and dysfunction. Cell Res. 2009, 19, 116–127. [Google Scholar] [CrossRef] [PubMed]
  15. Goumans, M.J.; Ten Dijke, P. TGF-β Signaling in Control of Cardiovascular Function. Cold Spring Harb. Perspect. Biol. 2018, 10, a022210. [Google Scholar] [CrossRef] [PubMed]
  16. Gentry, L.E.; Lioubin, M.N.; Purchio, A.F.; Marquardt, H. Molecular events in the processing of recombinant type 1 pre-pro-transforming growth factor beta to the mature polypeptide. Mol. Cell. Biol. 1988, 8, 4162–4168. [Google Scholar] [CrossRef] [PubMed]
  17. Derynck, R.; Jarrett, J.A.; Chen, E.Y.; Eaton, D.H.; Bell, J.R.; Assoian, R.K.; Roberts, A.B.; Sporn, M.B.; Goeddel, D.V. Human transforming growth factor-β complementary DNA sequence and expression in normal and transformed cells. Nature 1985, 316, 701–705. [Google Scholar] [CrossRef] [PubMed]
  18. Gray, A.M.; Mason, A.J. Requirement for activin A and transforming growth factor--β 1 pro-regions in homodimer assembly. Science 1990, 247, 1328–1330. [Google Scholar] [CrossRef] [PubMed]
  19. Olof Olsson, P.; Gustafsson, R.; Salnikov, A.V.; Gothe, M.; Zeller, K.S.; Friman, T.; Baldetorp, B.; Koopman, L.A.; Weinreb, P.H.; Violette, S.M.; et al. Inhibition of integrin α(V)β(6) changes fibril thickness of stromal collagen in experimental carcinomas. Cell Commun. Signal. 2018, 16, 36. [Google Scholar] [CrossRef]
  20. Nieberler, M.; Reuning, U.; Reichart, F.; Notni, J.; Wester, H.J.; Schwaiger, M.; Weinmuller, M.; Rader, A.; Steiger, K.; Kessler, H. Exploring the Role of RGD-Recognizing Integrins in Cancer. Cancers 2017, 9, 116. [Google Scholar] [CrossRef]
  21. Rapisarda, V.; Borghesan, M.; Miguela, V.; Encheva, V.; Snijders, A.P.; Lujambio, A.; O’Loghlen, A. Integrin Beta 3 Regulates Cellular Senescence by Activating the TGF-β Pathway. Cell Rep. 2017, 18, 2480–2493. [Google Scholar] [CrossRef]
  22. Dong, X.; Zhao, B.; Iacob, R.E.; Zhu, J.; Koksal, A.C.; Lu, C.; Engen, J.R.; Springer, T.A. Force interacts with macromolecular structure in activation of TGF-β. Nature 2017, 542, 55–59. [Google Scholar] [CrossRef] [PubMed]
  23. Ma, W.; Qin, Y.; Chapuy, B.; Lu, C. LRRC33 is a novel binding and potential regulating protein of TGF-β1 function in human acute myeloid leukemia cells. PLoS ONE 2019, 14, e0213482. [Google Scholar] [CrossRef] [PubMed]
  24. Hao, Y.; Baker, D.; Ten Dijke, P. TGF-β-Mediated Epithelial-Mesenchymal Transition and Cancer Metastasis. Int. J. Mol. Sci. 2019, 20, 2767. [Google Scholar] [CrossRef] [PubMed]
  25. Morikawa, M.; Derynck, R.; Miyazono, K. TGF-β and the TGF-β Family: Context-Dependent Roles in Cell and Tissue Physiology. Cold Spring Harb. Perspect. Biol. 2016, 8, a021873. [Google Scholar] [CrossRef] [PubMed]
  26. Flanders, K.C.; Yang, Y.A.; Herrmann, M.; Chen, J.; Mendoza, N.; Mirza, A.M.; Wakefield, L.M. Quantitation of TGF-β proteins in mouse tissues shows reciprocal changes in TGF-β1 and TGF-β3 in normal vs neoplastic mammary epithelium. Oncotarget 2016, 7, 38164–38179. [Google Scholar] [CrossRef] [PubMed]
  27. Derynck, R.; Zhang, Y.E. Smad-dependent and Smad-independent pathways in TGF-β family signalling. Nature 2003, 425, 577–584. [Google Scholar] [CrossRef] [PubMed]
  28. Shi, X.; Chen, F.; Yu, J.; Xu, Y.; Zhang, S.; Chen, Y.G.; Fang, X. Study of interaction between Smad7 and DNA by single-molecule force spectroscopy. Biochem. Biophys. Res. Commun. 2008, 377, 1284–1287. [Google Scholar] [CrossRef]
  29. De Ceuninck van Capelle, C.; Spit, M.; Ten Dijke, P. Current perspectives on inhibitory SMAD7 in health and disease. Crit. Rev. Biochem. Mol. Biol. 2020, 55, 691–715. [Google Scholar] [CrossRef]
  30. Zhang, Y.E. Non-Smad Signaling Pathways of the TGF-β Family. Cold Spring Harb. Perspect. Biol. 2017, 9, 128–139. [Google Scholar] [CrossRef]
  31. Tie, Y.; Tang, F.; Peng, D.; Zhang, Y.; Shi, H. TGF-β signal transduction: Biology, function and therapy for diseases. Mol. Biomed. 2022, 3, 45. [Google Scholar] [CrossRef] [PubMed]
  32. Su, J.; Morgani, S.M.; David, C.J.; Wang, Q.; Er, E.E.; Huang, Y.H.; Basnet, H.; Zou, Y.; Shu, W.; Soni, R.K.; et al. TGF-β orchestrates fibrogenic and developmental EMTs via the RAS effector RREB1. Nature 2020, 577, 566–571. [Google Scholar] [CrossRef] [PubMed]
  33. Lemmon, M.A.; Schlessinger, J. Cell signaling by receptor tyrosine kinases. Cell 2010, 141, 1117–1134. [Google Scholar] [CrossRef] [PubMed]
  34. Battegay, E.J.; Raines, E.W.; Seifert, R.A.; Bowen-Pope, D.F.; Ross, R. TGF-β induces bimodal proliferation of connective tissue cells via complex control of an autocrine PDGF loop. Cell 1990, 63, 515–524. [Google Scholar] [CrossRef] [PubMed]
  35. Riggins, G.J.; Kinzler, K.W.; Vogelstein, B.; Thiagalingam, S. Frequency of Smad gene mutations in human cancers. Cancer Res. 1997, 57, 2578–2580. [Google Scholar] [PubMed]
  36. Mueller, S.; Engleitner, T.; Maresch, R.; Zukowska, M.; Lange, S.; Kaltenbacher, T.; Konukiewitz, B.; Ollinger, R.; Zwiebel, M.; Strong, A.; et al. Evolutionary routes and KRAS dosage define pancreatic cancer phenotypes. Nature 2018, 554, 62–68. [Google Scholar] [CrossRef] [PubMed]
  37. Huang, Y.H.; Lin, P.C.; Su, W.C.; Chan, R.H.; Chen, P.C.; Lin, B.W.; Shen, M.R.; Chen, S.H.; Yeh, Y.M. Association between Altered Oncogenic Signaling Pathways and Overall Survival of Patients with Metastatic Colorectal Cancer. Diagnostics 2021, 11, 2308. [Google Scholar] [CrossRef] [PubMed]
  38. Markowitz, S.; Wang, J.; Myeroff, L.; Parsons, R.; Sun, L.; Lutterbaugh, J.; Fan, R.S.; Zborowska, E.; Kinzler, K.W.; Vogelstein, B.; et al. Inactivation of the type II TGF-β receptor in colon cancer cells with microsatellite instability. Science 1995, 268, 1336–1338. [Google Scholar] [CrossRef]
  39. Bellam, N.; Pasche, B. Tgf-β signaling alterations and colon cancer. Cancer Treat. Res. 2010, 155, 85–103. [Google Scholar] [CrossRef]
  40. Gu, S.; Zaidi, S.; Hassan, M.I.; Mohammad, T.; Malta, T.M.; Noushmehr, H.; Nguyen, B.; Crandall, K.A.; Srivastav, J.; Obias, V.; et al. Mutated CEACAMs Disrupt Transforming Growth Factor Beta Signaling and Alter the Intestinal Microbiome to Promote Colorectal Carcinogenesis. Gastroenterology 2020, 158, 238–252. [Google Scholar] [CrossRef]
  41. Ding, N.; Luo, H.; Zhang, T.; Peng, T.; Yao, Y.; He, Y. Correlation between SMADs and Colorectal Cancer Expression, Prognosis, and Immune Infiltrates. Int. J. Anal. Chem. 2023, 2023, 8414040. [Google Scholar] [CrossRef] [PubMed]
  42. Barrett, M.T.; Schutte, M.; Kern, S.E.; Reid, B.J. Allelic loss and mutational analysis of the DPC4 gene in esophageal adenocarcinoma. Cancer Res. 1996, 56, 4351–4353. [Google Scholar] [PubMed]
  43. Wu, T.T.; Watanabe, T.; Heitmiller, R.; Zahurak, M.; Forastiere, A.A.; Hamilton, S.R. Genetic alterations in Barrett esophagus and adenocarcinomas of the esophagus and esophagogastric junction region. Am. J. Pathol. 1998, 153, 287–294. [Google Scholar] [CrossRef] [PubMed]
  44. Onwuegbusi, B.A.; Aitchison, A.; Chin, S.F.; Kranjac, T.; Mills, I.; Huang, Y.; Lao-Sirieix, P.; Caldas, C.; Fitzgerald, R.C. Impaired transforming growth factor beta signalling in Barrett’s carcinogenesis due to frequent SMAD4 inactivation. Gut 2006, 55, 764–774. [Google Scholar] [CrossRef] [PubMed]
  45. Blum, A.E.; Venkitachalam, S.; Ravillah, D.; Chelluboyina, A.K.; Kieber-Emmons, A.M.; Ravi, L.; Kresak, A.; Chandar, A.K.; Markowitz, S.D.; Canto, M.I.; et al. Systems Biology Analyses Show Hyperactivation of Transforming Growth Factor-β and JNK Signaling Pathways in Esophageal Cancer. Gastroenterology 2019, 156, 1761–1774. [Google Scholar] [CrossRef] [PubMed]
  46. Zhao, M.; Mishra, L.; Deng, C.X. The role of TGF-β/SMAD4 signaling in cancer. Int. J. Biol. Sci. 2018, 14, 111–123. [Google Scholar] [CrossRef] [PubMed]
  47. Song, W.; Dai, W.J.; Zhang, M.H.; Wang, H.; Yang, X.Z. Comprehensive Analysis of the Expression of TGF-β Signaling Regulators and Prognosis in Human Esophageal Cancer. Comput. Math. Methods Med. 2021, 2021, 1812227. [Google Scholar] [CrossRef] [PubMed]
  48. Levy, L.; Hill, C.S. Alterations in components of the TGF-β superfamily signaling pathways in human cancer. Cytokine Growth Factor Rev. 2006, 17, 41–58. [Google Scholar] [CrossRef]
  49. Samanta, D.; Datta, P.K. Alterations in the Smad pathway in human cancers. Front. Biosci. (Landmark Ed) 2012, 17, 1281–1293. [Google Scholar] [CrossRef]
  50. Verma, R.; Agarwal, A.K.; Sakhuja, P.; Sharma, P.C. Microsatellite instability in mismatch repair and tumor suppressor genes and their expression profiling provide important targets for the development of biomarkers in gastric cancer. Gene 2019, 710, 48–58. [Google Scholar] [CrossRef]
  51. Shitara, Y.; Yokozaki, H.; Yasui, W.; Takenoshita, S.; Kuwano, H.; Nagamachi, Y.; Tahara, E. No mutations of the Smad2 gene in human sporadic gastric carcinomas. Jpn. J. Clin. Oncol. 1999, 29, 3–7. [Google Scholar] [CrossRef] [PubMed]
  52. Fleming, N.I.; Jorissen, R.N.; Mouradov, D.; Christie, M.; Sakthianandeswaren, A.; Palmieri, M.; Day, F.; Li, S.; Tsui, C.; Lipton, L.; et al. SMAD2, SMAD3 and SMAD4 mutations in colorectal cancer. Cancer Res. 2013, 73, 725–735. [Google Scholar] [CrossRef] [PubMed]
  53. Xu, Y.; Pasche, B. TGF-β signaling alterations and susceptibility to colorectal cancer. Hum. Mol. Genet. 2007, 16, R14–R20. [Google Scholar] [CrossRef] [PubMed]
  54. Troncone, E.; Monteleone, G. Smad7 and Colorectal Carcinogenesis: A Double-Edged Sword. Cancers 2019, 11, 612. [Google Scholar] [CrossRef] [PubMed]
  55. Truty, M.J.; Urrutia, R. Basics of TGF-β and pancreatic cancer. Pancreatology 2007, 7, 423–435. [Google Scholar] [CrossRef] [PubMed]
  56. Nagai, M.; Kawarada, Y.; Watanabe, M.; Iwase, T.; Muneyuki, T.; Yamao, K.; Fukutome, K.; Yatani, R. Analysis of microsatellite instability, TGF-β type II receptor gene mutations and hMSH2 and hMLH1 allele losses in pancreaticobiliary maljunction-associated biliary tract tumors. Anticancer Res. 1999, 19, 1765–1768. [Google Scholar] [PubMed]
  57. Zou, S.; Li, J.; Zhou, H.; Frech, C.; Jiang, X.; Chu, J.S.; Zhao, X.; Li, Y.; Li, Q.; Wang, H.; et al. Mutational landscape of intrahepatic cholangiocarcinoma. Nat. Commun. 2014, 5, 5696. [Google Scholar] [CrossRef] [PubMed]
  58. Heldin, C.H.; Vanlandewijck, M.; Moustakas, A. Regulation of EMT by TGFbeta in cancer. FEBS Lett. 2012, 586, 1959–1970. [Google Scholar] [CrossRef]
  59. Katsuno, Y.; Derynck, R. Epithelial plasticity, epithelial-mesenchymal transition, and the TGF-β family. Dev. Cell 2021, 56, 726–746. [Google Scholar] [CrossRef]
  60. Zavadil, J.; Bitzer, M.; Liang, D.; Yang, Y.C.; Massimi, A.; Kneitz, S.; Piek, E.; Bottinger, E.P. Genetic programs of epithelial cell plasticity directed by transforming growth factor-β. Proc. Natl. Acad. Sci. USA 2001, 98, 6686–6691. [Google Scholar] [CrossRef]
  61. Puram, S.V.; Parikh, A.S.; Tirosh, I. Single cell RNA-seq highlights a role for a partial EMT in head and neck cancer. Mol. Cell. Oncol. 2018, 5, e1448244. [Google Scholar] [CrossRef]
  62. Katsuno, Y.; Meyer, D.S.; Zhang, Z.; Shokat, K.M.; Akhurst, R.J.; Miyazono, K.; Derynck, R. Chronic TGF-β exposure drives stabilized EMT, tumor stemness, and cancer drug resistance with vulnerability to bitopic mTOR inhibition. Sci. Signal. 2019, 12, eaau8544. [Google Scholar] [CrossRef] [PubMed]
  63. Krantz, S.B.; Shields, M.A.; Dangi-Garimella, S.; Munshi, H.G.; Bentrem, D.J. Contribution of epithelial-to-mesenchymal transition and cancer stem cells to pancreatic cancer progression. J. Surg. Res. 2012, 173, 105–112. [Google Scholar] [CrossRef] [PubMed]
  64. Huang, Y.H.; Hu, J.; Chen, F.; Lecomte, N.; Basnet, H.; David, C.J.; Witkin, M.D.; Allen, P.J.; Leach, S.D.; Hollmann, T.J.; et al. ID1 Mediates Escape from TGFbeta Tumor Suppression in Pancreatic Cancer. Cancer Discov. 2020, 10, 142–157. [Google Scholar] [CrossRef] [PubMed]
  65. Li, H.; Wang, S.; Li, X.; Weng, Y.; Guo, D.; Kong, P.; Cheng, C.; Wang, Y.; Zhang, L.; Cheng, X.; et al. CDCA7 promotes TGF-β-induced epithelial-mesenchymal transition via transcriptionally regulating Smad4/Smad7 in ESCC. Cancer Sci. 2023, 114, 91–104. [Google Scholar] [CrossRef] [PubMed]
  66. Xu, H.J.; Bai, J.; Tian, Y.; Feng, X.; Chen, A.P.; Wang, J.; Wu, J.; Jin, X.R.; Zhang, F.; Quan, M.Y.; et al. ESE1/AGR2 axis antagonizes TGF-β-induced epithelial-mesenchymal transition in low-grade pancreatic cancer. Cancer Med. 2023, 12, 5979–5993. [Google Scholar] [CrossRef] [PubMed]
  67. Quan, J.; Cheng, C.; Tan, Y.; Jiang, N.; Liao, C.; Liao, W.; Cao, Y.; Luo, X. Acyl-CoA synthetase long-chain 3-mediated fatty acid oxidation is required for TGFbeta1-induced epithelial-mesenchymal transition and metastasis of colorectal carcinoma. Int. J. Biol. Sci. 2022, 18, 2484–2496. [Google Scholar] [CrossRef] [PubMed]
  68. Chen, F.; Zhuang, X.; Lin, L.; Yu, P.; Wang, Y.; Shi, Y.; Hu, G.; Sun, Y. New horizons in tumor microenvironment biology: Challenges and opportunities. BMC Med. 2015, 13, 45. [Google Scholar] [CrossRef]
  69. Hinshaw, D.C.; Shevde, L.A. The Tumor Microenvironment Innately Modulates Cancer Progression. Cancer Res. 2019, 79, 4557–4566. [Google Scholar] [CrossRef]
  70. 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]
  71. Furman, D.; Campisi, J.; Verdin, E.; Carrera-Bastos, P.; Targ, S.; Franceschi, C.; Ferrucci, L.; Gilroy, D.W.; Fasano, A.; Miller, G.W.; et al. Chronic inflammation in the etiology of disease across the life span. Nat. Med. 2019, 25, 1822–1832. [Google Scholar] [CrossRef] [PubMed]
  72. Wynn, T.A.; Ramalingam, T.R. Mechanisms of fibrosis: Therapeutic translation for fibrotic disease. Nat. Med. 2012, 18, 1028–1040. [Google Scholar] [CrossRef] [PubMed]
  73. Samarakoon, R.; Overstreet, J.M.; Higgins, P.J. TGF-β signaling in tissue fibrosis: Redox controls, target genes and therapeutic opportunities. Cell. Signal. 2013, 25, 264–268. [Google Scholar] [CrossRef] [PubMed]
  74. Mazzocca, A.; Fransvea, E.; Dituri, F.; Lupo, L.; Antonaci, S.; Giannelli, G. Down-regulation of connective tissue growth factor by inhibition of transforming growth factor beta blocks the tumor-stroma cross-talk and tumor progression in hepatocellular carcinoma. Hepatology 2010, 51, 523–534. [Google Scholar] [CrossRef] [PubMed]
  75. Yashiro, M.; Hirakawa, K. Cancer-stromal interactions in scirrhous gastric carcinoma. Cancer Microenviron. 2010, 3, 127–135. [Google Scholar] [CrossRef] [PubMed]
  76. Liao, Z.; Tan, Z.W.; Zhu, P.; Tan, N.S. Cancer-associated fibroblasts in tumor microenvironment—Accomplices in tumor malignancy. Cell. Immunol. 2019, 343, 103729. [Google Scholar] [CrossRef] [PubMed]
  77. Pereira, B.A.; Vennin, C.; Papanicolaou, M.; Chambers, C.R.; Herrmann, D.; Morton, J.P.; Cox, T.R.; Timpson, P. CAF Subpopulations: A New Reservoir of Stromal Targets in Pancreatic Cancer. Trends Cancer 2019, 5, 724–741. [Google Scholar] [CrossRef]
  78. Zhu, Z.; Achreja, A.; Meurs, N.; Animasahun, O.; Owen, S.; Mittal, A.; Parikh, P.; Lo, T.W.; Franco-Barraza, J.; Shi, J.; et al. Tumour-reprogrammed stromal BCAT1 fuels branched-chain ketoacid dependency in stromal-rich PDAC tumours. Nat. Metab. 2020, 2, 775–792. [Google Scholar] [CrossRef]
  79. Yang, J.; Lu, Y.; Lin, Y.Y.; Zheng, Z.Y.; Fang, J.H.; He, S.; Zhuang, S.M. Vascular mimicry formation is promoted by paracrine TGF-β and SDF1 of cancer-associated fibroblasts and inhibited by miR-101 in hepatocellular carcinoma. Cancer Lett. 2016, 383, 18–27. [Google Scholar] [CrossRef]
  80. Krzysiek-Maczka, G.; Targosz, A.; Szczyrk, U.; Wrobel, T.; Strzalka, M.; Brzozowski, T.; Czyz, J.; Ptak-Belowska, A. Long-Term Helicobacter pylori Infection Switches Gastric Epithelium Reprogramming Towards Cancer Stem Cell-Related Differentiation Program in Hp-Activated Gastric Fibroblast-TGFbeta Dependent Manner. Microorganisms 2020, 8, 1519. [Google Scholar] [CrossRef]
  81. Koyama, T.; Yashiro, M.; Inoue, T.; Nishimura, S.; Hirakawa, Y.S.C.K. TGF-β1 secreted by gastric fibroblasts up-regulates CD44H expression and stimulates the peritoneal metastatic ability of scirrhous gastric cancer cells. Int. J. Oncol. 2000, 16, 355–362. [Google Scholar] [CrossRef] [PubMed]
  82. Fuyuhiro, Y.; Yashiro, M.; Noda, S.; Matsuoka, J.; Hasegawa, T.; Kato, Y.; Sawada, T.; Hirakawa, K. Cancer-associated orthotopic myofibroblasts stimulates the motility of gastric carcinoma cells. Cancer Sci. 2012, 103, 797–805. [Google Scholar] [CrossRef] [PubMed]
  83. Yashiro, M.; Chung, Y.S.; Inoue, T.; Nishimura, S.; Matsuoka, T.; Fujihara, T.; Sowa, M. Hepatocyte growth factor (HGF) produced by peritoneal fibroblasts may affect mesothelial cell morphology and promote peritoneal dissemination. Int. J. Cancer 1996, 67, 289–293. [Google Scholar] [CrossRef]
  84. Hasegawa, T.; Yashiro, M.; Nishii, T.; Matsuoka, J.; Fuyuhiro, Y.; Morisaki, T.; Fukuoka, T.; Shimizu, K.; Shimizu, T.; Miwa, A.; et al. Cancer-associated fibroblasts might sustain the stemness of scirrhous gastric cancer cells via transforming growth factor-β signaling. Int. J. Cancer 2014, 134, 1785–1795. [Google Scholar] [CrossRef] [PubMed]
  85. Pan, H.; Pan, J.; Wu, J. Development and validation of a cancer-associated fibroblast-derived lncRNA signature for predicting clinical outcomes in colorectal cancer. Front. Immunol. 2022, 13, 934221. [Google Scholar] [CrossRef] [PubMed]
  86. Song, J.; Chen, W.; Cui, X.; Huang, Z.; Wen, D.; Yang, Y.; Yu, W.; Cui, L.; Liu, C.Y. CCBE1 promotes tumor lymphangiogenesis and is negatively regulated by TGFbeta signaling in colorectal cancer. Theranostics 2020, 10, 2327–2341. [Google Scholar] [CrossRef] [PubMed]
  87. Jeltsch, M.; Jha, S.K.; Tvorogov, D.; Anisimov, A.; Leppanen, V.M.; Holopainen, T.; Kivela, R.; Ortega, S.; Karpanen, T.; Alitalo, K. CCBE1 enhances lymphangiogenesis via A disintegrin and metalloprotease with thrombospondin motifs-3-mediated vascular endothelial growth factor-C activation. Circulation 2014, 129, 1962–1971. [Google Scholar] [CrossRef] [PubMed]
  88. Miyazono, K.; Katsuno, Y.; Koinuma, D.; Ehata, S.; Morikawa, M. Intracellular and extracellular TGF-β signaling in cancer: Some recent topics. Front. Med. 2018, 12, 387–411. [Google Scholar] [CrossRef]
  89. Gong, D.; Shi, W.; Yi, S.J.; Chen, H.; Groffen, J.; Heisterkamp, N. TGFbeta signaling plays a critical role in promoting alternative macrophage activation. BMC Immunol. 2012, 13, 31. [Google Scholar] [CrossRef]
  90. Kelly, A.; Gunaltay, S.; McEntee, C.P.; Shuttleworth, E.E.; Smedley, C.; Houston, S.A.; Fenton, T.M.; Levison, S.; Mann, E.R.; Travis, M.A. Human monocytes and macrophages regulate immune tolerance via integrin alphavbeta8-mediated TGFbeta activation. J. Exp. Med. 2018, 215, 2725–2736. [Google Scholar] [CrossRef]
  91. Fridlender, Z.G.; Sun, J.; Kim, S.; Kapoor, V.; Cheng, G.; Ling, L.; Worthen, G.S.; Albelda, S.M. Polarization of tumor-associated neutrophil phenotype by TGF-β: “N1” versus “N2” TAN. Cancer Cell 2009, 16, 183–194. [Google Scholar] [CrossRef] [PubMed]
  92. MaruYama, T.; Chen, W.; Shibata, H. TGF-β and Cancer Immunotherapy. Biol. Pharm. Bull. 2022, 45, 155–161. [Google Scholar] [CrossRef] [PubMed]
  93. Tamayo, E.; Alvarez, P.; Merino, R. TGFbeta Superfamily Members as Regulators of B Cell Development and Function-Implications for Autoimmunity. Int. J. Mol. Sci. 2018, 19, 3928. [Google Scholar] [CrossRef]
  94. Jiao, S.; Subudhi, S.K.; Aparicio, A.; Ge, Z.; Guan, B.; Miura, Y.; Sharma, P. Differences in Tumor Microenvironment Dictate T Helper Lineage Polarization and Response to Immune Checkpoint Therapy. Cell 2019, 179, 1177–1190.e13. [Google Scholar] [CrossRef]
  95. Takeuchi, M.; Kosiewicz, M.M.; Alard, P.; Streilein, J.W. On the mechanisms by which transforming growth factor-β 2 alters antigen-presenting abilities of macrophages on T cell activation. Eur. J. Immunol. 1997, 27, 1648–1656. [Google Scholar] [CrossRef] [PubMed]
  96. Das, L.; Levine, A.D. TGF-β inhibits IL-2 production and promotes cell cycle arrest in TCR-activated effector/memory T cells in the presence of sustained TCR signal transduction. J. Immunol. 2008, 180, 1490–1498. [Google Scholar] [CrossRef]
  97. McKarns, S.C.; Schwartz, R.H. Distinct effects of TGF-β 1 on CD4+ and CD8+ T cell survival, division, and IL-2 production: A role for T cell intrinsic Smad3. J. Immunol. 2005, 174, 2071–2083. [Google Scholar] [CrossRef] [PubMed]
  98. Budhu, S.; Schaer, D.A.; Li, Y.; Toledo-Crow, R.; Panageas, K.; Yang, X.; Zhong, H.; Houghton, A.N.; Silverstein, S.C.; Merghoub, T.; et al. Blockade of surface-bound TGF-β on regulatory T cells abrogates suppression of effector T cell function in the tumor microenvironment. Sci. Signal. 2017, 10, eaak9702. [Google Scholar] [CrossRef]
  99. Zhang, X.L.; Hu, L.P.; Yang, Q.; Qin, W.T.; Wang, X.; Xu, C.J.; Tian, G.A.; Yang, X.M.; Yao, L.L.; Zhu, L.; et al. CTHRC1 promotes liver metastasis by reshaping infiltrated macrophages through physical interactions with TGF-β receptors in colorectal cancer. Oncogene 2021, 40, 3959–3973. [Google Scholar] [CrossRef]
  100. Erdman, S.E.; Rao, V.P.; Poutahidis, T.; Ihrig, M.M.; Ge, Z.; Feng, Y.; Tomczak, M.; Rogers, A.B.; Horwitz, B.H.; Fox, J.G. CD4+CD25+ regulatory lymphocytes require interleukin 10 to interrupt colon carcinogenesis in mice. Cancer Res. 2003, 63, 6042–6050. [Google Scholar]
  101. Xiong, C.; Zhu, Y.; Xue, M.; Jiang, Y.; Zhong, Y.; Jiang, L.; Shi, M.; Chen, H. Tumor-associated macrophages promote pancreatic ductal adenocarcinoma progression by inducing epithelial-to-mesenchymal transition. Aging 2021, 13, 3386–3404. [Google Scholar] [CrossRef] [PubMed]
  102. Samiei, H.; Sadighi-Moghaddam, B.; Mohammadi, S.; Gharavi, A.; Abdolmaleki, S.; Khosravi, A.; Kokhaei, P.; Bazzazi, H.; Memarian, A. Dysregulation of helper T lymphocytes in esophageal squamous cell carcinoma (ESCC) patients is highly associated with aberrant production of miR-21. Immunol. Res. 2019, 67, 212–222. [Google Scholar] [CrossRef] [PubMed]
  103. Yan, H.; Bu, P. Non-coding RNA in cancer. Essays Biochem. 2021, 65, 625–639. [Google Scholar] [CrossRef] [PubMed]
  104. Wu, N.; Jiang, M.; Liu, H.; Chu, Y.; Wang, D.; Cao, J.; Wang, Z.; Xie, X.; Han, Y.; Xu, B. LINC00941 promotes CRC metastasis through preventing SMAD4 protein degradation and activating the TGF-β/SMAD2/3 signaling pathway. Cell Death Differ. 2021, 28, 219–232. [Google Scholar] [CrossRef] [PubMed]
  105. Li, Y.; Liu, G.; Li, X.; Dong, H.; Xiao, W.; Lu, S. Long non-coding RNA SBF2-AS1 promotes hepatocellular carcinoma progression through regulation of miR-140-5p-TGFBR1 pathway. Biochem. Biophys. Res. Commun. 2018, 503, 2826–2832. [Google Scholar] [CrossRef]
  106. Su, J.; Chen, D.; Ruan, Y.; Tian, Y.; Lv, K.; Zhou, X.; Ying, D.; Lu, Y. LncRNA MBNL1-AS1 represses gastric cancer progression via the TGF-β pathway by modulating miR-424-5p/Smad7 axis. Bioengineered 2022, 13, 6978–6995. [Google Scholar] [CrossRef] [PubMed]
  107. Lv, S.; Li, Y.; Ning, H.; Zhang, M.; Jia, Q.; Wang, X. CircRNA GFRA1 promotes hepatocellular carcinoma progression by modulating the miR-498/NAP1L3 axis. Sci. Rep. 2021, 11, 386. [Google Scholar] [CrossRef] [PubMed]
  108. Zhou, B.; Guo, W.; Sun, C.; Zhang, B.; Zheng, F. Linc00462 promotes pancreatic cancer invasiveness through the miR-665/TGFBR1-TGFBR2/SMAD2/3 pathway. Cell Death Dis. 2018, 9, 706. [Google Scholar] [CrossRef]
  109. Liu, S.; He, X.; Di, Y.; Li, Q.; Li, F.; Ma, Y.; Chen, L.; Gao, Y.; Xu, J.; Yang, S.; et al. NamiRNA-enhancer network of miR-492 activates the NR2C1-TGF-β/Smad3 pathway to promote epithelial-mesenchymal transition of pancreatic cancer. Carcinogenesis 2023, 44, 153–165. [Google Scholar] [CrossRef]
  110. Wang, D.; Wang, X.; Song, Y.; Si, M.; Sun, Y.; Liu, X.; Cui, S.; Qu, X.; Yu, X. Exosomal miR-146a-5p and miR-155-5p promote CXCL12/CXCR7-induced metastasis of colorectal cancer by crosstalk with cancer-associated fibroblasts. Cell Death Dis. 2022, 13, 380. [Google Scholar] [CrossRef]
  111. Yin, Z.; Ma, T.; Huang, B.; Lin, L.; Zhou, Y.; Yan, J.; Zou, Y.; Chen, S. Macrophage-derived exosomal microRNA-501-3p promotes progression of pancreatic ductal adenocarcinoma through the TGFBR3-mediated TGF-β signaling pathway. J. Exp. Clin. Cancer Res. 2019, 38, 310. [Google Scholar] [CrossRef]
  112. Tian, X.; Fei, Q.; Du, M.; Zhu, H.; Ye, J.; Qian, L.; Lu, Z.; Zhang, W.; Wang, Y.; Peng, F.; et al. miR-130a-3p regulated TGF-β1-induced epithelial-mesenchymal transition depends on SMAD4 in EC-1 cells. Cancer Med. 2019, 8, 1197–1208. [Google Scholar] [CrossRef] [PubMed]
  113. Cai, Y.; Ruan, W.; Ding, J.; Wei, N.; Wang, J.; Zhang, H.; Ma, N.; Weng, G.; Su, W.K.; Lin, Y.; et al. miR-93-5p regulates the occurrence and development of esophageal carcinoma epithelial cells by targeting TGFbetaR2. Int. J. Mol. Med. 2021, 47, 3. [Google Scholar] [CrossRef] [PubMed]
  114. Song, J.; Yang, P.; Li, X.; Zhu, X.; Liu, M.; Duan, X.; Liu, R. Esophageal Cancer-Derived Extracellular Vesicle miR-21-5p Contributes to EMT of ESCC Cells by Disorganizing Macrophage Polarization. Cancers 2021, 13, 4122. [Google Scholar] [CrossRef] [PubMed]
  115. Xu, R.; Zhou, X.M.; Li, Y.S.; Ren, L.; He, X.R. MicroRNA-181a promotes epithelial-mesenchymal transition in esophageal squamous cell carcinoma via the TGF-β/Smad pathway. Mol. Med. Rep. 2021, 23, 316. [Google Scholar] [CrossRef] [PubMed]
  116. Samiei, H.; Ajam, F.; Gharavi, A.; Abdolmaleki, S.; Kokhaei, P.; Mohammadi, S.; Memarian, A. Simultaneous disruption of circulating miR-21 and cytotoxic T lymphocytes (CTLs): Prospective diagnostic and prognostic markers for esophageal squamous cell carcinoma (ESCC). J. Clin. Lab. Anal. 2022, 36, e24125. [Google Scholar] [CrossRef] [PubMed]
  117. Luo, Y.; Wu, J.; Wu, Q.; Li, X.; Wu, J.; Zhang, J.; Rong, X.; Rao, J.; Liao, Y.; Bin, J.; et al. miR-577 Regulates TGF-β Induced Cancer Progression through a SDPR-Modulated Positive-Feedback Loop with ERK-NF-kappaB in Gastric Cancer. Mol. Ther. 2019, 27, 1166–1182. [Google Scholar] [CrossRef] [PubMed]
  118. Zhu, Q.L.; Li, Z.; Lv, C.M.; Wang, W. MiR-187 influences cisplatin-resistance of gastric cancer cells through regulating the TGF-β/Smad signaling pathway. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 9907–9914. [Google Scholar] [CrossRef] [PubMed]
  119. Bai, M.; Wang, P.; Yang, J.; Zuo, M.; Ba, Y. Identification of miR-135b as a novel regulator of TGFbeta pathway in gastric cancer. J. Physiol. Biochem. 2020, 76, 549–560. [Google Scholar] [CrossRef]
  120. Hu, W.; Zheng, X.; Liu, J.; Zhang, M.; Liang, Y.; Song, M. MicroRNA MiR-130a-3p promotes gastric cancer by targeting Glucosaminyl N-acetyl transferase 4 (GCNT4) to regulate the TGF-β1/SMAD3 pathway. Bioengineered 2021, 12, 11634–11647. [Google Scholar] [CrossRef]
  121. Zhu, M.; Zhang, N.; Ma, J.; He, S. Integration of exosomal miR-106a and mesothelial cells facilitates gastric cancer peritoneal dissemination. Cell Signal. 2022, 91, 110230. [Google Scholar] [CrossRef] [PubMed]
  122. Shichiri, K.; Oshi, M.; Ziazadeh, D.; Endo, I.; Takabe, K. High miR-200c expression is associated with suppressed epithelial-mesenchymal transition, TGF-β signaling and better survival despite enhanced cell proliferation in gastric cancer patients. Am. J. Cancer Res. 2023, 13, 3027–3040. [Google Scholar] [PubMed]
  123. Gao, S.; Zhang, Z.; Wang, X.; Ma, Y.; Li, C.; Liu, H.; Jing, C.; Li, L.; Guo, X. hsa-miR-875-5p inhibits tumorigenesis and suppresses TGF-β signalling by targeting USF2 in gastric cancer. J. Transl. Med. 2022, 20, 115. [Google Scholar] [CrossRef] [PubMed]
  124. Fricke, F.; Mussack, V.; Buschmann, D.; Hausser, I.; Pfaffl, M.W.; Kopitz, J.; Gebert, J. TGFBR2-dependent alterations of microRNA profiles in extracellular vesicles and parental colorectal cancer cells. Int. J. Oncol. 2019, 55, 925–937. [Google Scholar] [CrossRef] [PubMed]
  125. Ye, J.; Lei, J.; Fang, Q.; Shen, Y.; Xia, W.; Hu, X.; Xu, Q.; Yuan, H.; Huang, J.; Ni, C. miR-4666-3p and miR-329 Synergistically Suppress the Stemness of Colorectal Cancer Cells via Targeting TGF-β/Smad Pathway. Front. Oncol. 2019, 9, 1251. [Google Scholar] [CrossRef] [PubMed]
  126. Bayat, Z.; Ghaemi, Z.; Behmanesh, M.; Soltani, B.M. Hsa-miR-186-5p regulates TGFbeta signaling pathway through expression suppression of SMAD6 and SMAD7 genes in colorectal cancer. Biol. Chem. 2021, 402, 469–480. [Google Scholar] [CrossRef] [PubMed]
  127. Bartlett, B.; Gao, Z.; Schukking, M.; Menor, M.; Khadka, V.S.; Fabbri, M.; Fei, P.; Deng, Y. The miRNA Profile of Inflammatory Colorectal Tumors Identify TGF-β as a Companion Target for Checkpoint Blockade Immunotherapy. Front. Cell Dev. Biol. 2021, 9, 754507. [Google Scholar] [CrossRef]
  128. Bhome, R.; Emaduddin, M.; James, V.; House, L.M.; Thirdborough, S.M.; Mellone, M.; Tulkens, J.; Primrose, J.N.; Thomas, G.J.; De Wever, O.; et al. Epithelial to mesenchymal transition influences fibroblast phenotype in colorectal cancer by altering miR-200 levels in extracellular vesicles. J. Extracell. Vesicles 2022, 11, e12226. [Google Scholar] [CrossRef]
  129. Abkenar, B.R.; Mohammadi, A.; Amoli, H.A.; Soleimani, A.A.; Korani, M.; Mahmoodi, H.; Najafi, M. Non-coding RNAs are correlated to TGF-β receptor type 2 in patients with colorectal cancer. J. Gene Med. 2023, 25, e3472. [Google Scholar] [CrossRef]
  130. Zhao, P.; Ma, Y.G.; Zhao, Y.; Liu, D.; Dai, Z.J.; Yan, C.Y.; Guan, H.T. MicroRNA-552 deficiency mediates 5-fluorouracil resistance by targeting SMAD2 signaling in DNA-mismatch-repair-deficient colorectal cancer. Cancer Chemother. Pharmacol. 2019, 84, 427–439. [Google Scholar] [CrossRef]
  131. Kabiri, F.; Medlej, A.; Saleh, A.J.; Aghdami, N.; Khani, M.; Soltani, B.M. Downregulated miR-495-3p in colorectal cancer targets TGFbetaR1, TGFbetaR2, SMAD4 and BUB1 genes and induces cell cycle arrest. Cancer Treat. Res. Commun. 2023, 35, 100702. [Google Scholar] [CrossRef]
  132. Takahashi, K.; Koyama, K.; Ota, Y.; Iwamoto, H.; Yamakita, K.; Fujii, S.; Kitano, Y. The Interaction Between Long Non-coding RNA HULC and MicroRNA-622 via Transfer by Extracellular Vesicles Regulates Cell Invasion and Migration in Human Pancreatic Cancer. Front. Oncol. 2020, 10, 1013. [Google Scholar] [CrossRef] [PubMed]
  133. Sun, L.; Guo, Z.; Sun, J.; Li, J.; Dong, Z.; Zhang, Y.; Chen, J.; Kan, Q.; Yu, Z. MiR-133a acts as an anti-oncogene in Hepatocellular carcinoma by inhibiting FOSL2 through TGF-β/Smad3 signaling pathway. Biomed. Pharmacother. 2018, 107, 168–176. [Google Scholar] [CrossRef]
  134. Zhang, Q.; Huang, F.; Yao, Y.; Wang, J.; Wei, J.; Wu, Q.; Xiang, S.; Xu, L. Interaction of transforming growth factor-β-Smads/microRNA-362-3p/CD82 mediated by M2 macrophages promotes the process of epithelial-mesenchymal transition in hepatocellular carcinoma cells. Cancer Sci. 2019, 110, 2507–2519. [Google Scholar] [CrossRef] [PubMed]
  135. Zhang, J.; Zhu, Y.; Hu, L.; Yan, F.; Chen, J. miR-494 induces EndMT and promotes the development of HCC (Hepatocellular Carcinoma) by targeting SIRT3/TGF-β/SMAD signaling pathway. Sci. Rep. 2019, 9, 7213. [Google Scholar] [CrossRef] [PubMed]
  136. Zhao, Y.; Xu, Z.; Zhou, J.; Yang, H. miR-141 inhibits proliferation, migration and invasion in human hepatocellular carcinoma cells by directly downregulating TGFbetaR1. Oncol. Rep. 2019, 42, 1656–1666. [Google Scholar] [CrossRef] [PubMed]
  137. Hu, X.; Han, D.; Wang, Y.; Gu, J.; Wang, X.; Jiang, Y.; Yang, Y.; Liu, J. Phospho-Smad3L promotes progression of hepatocellular carcinoma through decreasing miR-140-5p level and stimulating epithelial-mesenchymal transition. Dig. Liver Dis. 2021, 53, 1343–1351. [Google Scholar] [CrossRef] [PubMed]
  138. Liu, H.T.; Luo, C.P.; Jiang, M.J.; Deng, Z.J.; Teng, Y.X.; Su, J.Y.; Pan, L.X.; Ma, L.; Guo, P.P.; Zhong, J.H. miR-17-5p slows progression of hepatocellular carcinoma by downregulating TGFbetaR2. Clin. Transl. Oncol. 2023, 25, 2960–2971. [Google Scholar] [CrossRef]
  139. Zhang, X.; Zhang, L.; Chen, M.; Liu, D. miR-324-5p inhibits gallbladder carcinoma cell metastatic behaviours by downregulation of transforming growth factor beta 2 expression. Artif. Cells Nanomed. Biotechnol. 2020, 48, 315–324. [Google Scholar] [CrossRef]
  140. Fu, X.; Chen, X.; Si, Y.; Yao, Y.; Jiang, Z.; Chen, K. Long non-coding RNA NCK1-AS1 is overexpressed in esophageal squamous cell carcinoma and predicts survival. Bioengineered 2022, 13, 8302–8310. [Google Scholar] [CrossRef]
  141. Feng, B.; Wang, G.; Liang, X.; Wu, Z.; Wang, X.; Dong, Z.; Guo, Y.; Shen, S.; Liang, J.; Guo, W. LncRNA FAM83H-AS1 promotes oesophageal squamous cell carcinoma progression via miR-10a-5p/Girdin axis. J. Cell. Mol. Med. 2020, 24, 8962–8976. [Google Scholar] [CrossRef]
  142. Zhang, X.; Wu, J. LINC00665 promotes cell proliferation, invasion, and metastasis by activating the TGF-β pathway in gastric cancer. Pathol. Res. Pract. 2021, 224, 153492. [Google Scholar] [CrossRef]
  143. Huang, D.; Zhang, K.; Zheng, W.; Zhang, R.; Chen, J.; Du, N.; Xia, Y.; Long, Y.; Gu, Y.; Xu, J.; et al. Long noncoding RNA SGO1-AS1 inactivates TGFbeta signaling by facilitating TGFB1/2 mRNA decay and inhibits gastric carcinoma metastasis. J. Exp. Clin. Cancer Res. 2021, 40, 342. [Google Scholar] [CrossRef] [PubMed]
  144. Li, D.; Xu, M.; Wang, Z.; Huang, P.; Huang, C.; Chen, Z.; Tang, G.; Zhu, X.; Cai, M.; Qin, S. The EMT-induced lncRNA NR2F1-AS1 positively modulates NR2F1 expression and drives gastric cancer via miR-29a-3p/VAMP7 axis. Cell Death Dis. 2022, 13, 84. [Google Scholar] [CrossRef] [PubMed]
  145. Miao, R.; Yao, Z.; Hu, B.; Jin, T.; Zhu, D.; Shi, Y.; Gong, Y.; Shao, S.; Shao, C. A novel long non-coding RNA XLOC_004787, is associated with migration and promotes cancer cell proliferation by downregulating mir-203a-3p in gastric cancer. BMC Gastroenterol. 2023, 23, 280. [Google Scholar] [CrossRef] [PubMed]
  146. Javanmard, A.R.; Dokanehiifard, S.; Bohlooli, M.; Soltani, B.M. LOC646329 long non-coding RNA sponges miR-29b-1 and regulates TGFbeta signaling in colorectal cancer. J Cancer Res. Clin. Oncol. 2020, 146, 1205–1215. [Google Scholar] [CrossRef] [PubMed]
  147. Wang, X.; Lai, Q.; He, J.; Li, Q.; Ding, J.; Lan, Z.; Gu, C.; Yan, Q.; Fang, Y.; Zhao, X.; et al. LncRNA SNHG6 promotes proliferation, invasion and migration in colorectal cancer cells by activating TGF-β/Smad signaling pathway via targeting UPF1 and inducing EMT via regulation of ZEB1. Int. J. Med. Sci. 2019, 16, 51–59. [Google Scholar] [CrossRef] [PubMed]
  148. Luo, K.; Geng, J.; Zhang, Q.; Xu, Y.; Zhou, X.; Huang, Z.; Shi, K.Q.; Pan, C.; Wu, J. LncRNA CASC9 interacts with CPSF3 to regulate TGF-β signaling in colorectal cancer. J. Exp. Clin. Cancer Res. 2019, 38, 249. [Google Scholar] [CrossRef] [PubMed]
  149. Xu, J.; Shao, T.; Song, M.; Xie, Y.; Zhou, J.; Yin, J.; Ding, N.; Zou, H.; Li, Y.; Zhang, J. MIR22HG acts as a tumor suppressor via TGFbeta/SMAD signaling and facilitates immunotherapy in colorectal cancer. Mol. Cancer 2020, 19, 51. [Google Scholar] [CrossRef] [PubMed]
  150. Shen, X.; Hu, X.; Mao, J.; Wu, Y.; Liu, H.; Shen, J.; Yu, J.; Chen, W. The long noncoding RNA TUG1 is required for TGF-β/TWIST1/EMT-mediated metastasis in colorectal cancer cells. Cell Death Dis. 2020, 11, 65. [Google Scholar] [CrossRef]
  151. Li, Q.; Yue, W.; Li, M.; Jiang, Z.; Hou, Z.; Liu, W.; Ma, N.; Gan, W.; Li, Y.; Zhou, T.; et al. Downregulating Long Non-coding RNAs CTBP1-AS2 Inhibits Colorectal Cancer Development by Modulating the miR-93-5p/TGF-β/SMAD2/3 Pathway. Front. Oncol. 2021, 11, 626620. [Google Scholar] [CrossRef] [PubMed]
  152. Yang, L.; Dong, X.; Liu, Z.; Tan, J.; Huang, X.; Wen, T.; Qu, H.; Wang, Z. VPS9D1-AS1 overexpression amplifies intratumoral TGF-β signaling and promotes tumor cell escape from CD8+ T cell killing in colorectal cancer. Elife 2022, 11, e79811. [Google Scholar] [CrossRef]
  153. Zhang, X.; Feng, W.; Zhang, J.; Ge, L.; Zhang, Y.; Jiang, X.; Peng, W.; Wang, D.; Gong, A.; Xu, M. Long non-coding RNA PVT1 promotes epithelial-mesenchymal transition via the TGF-β/Smad pathway in pancreatic cancer cells. Oncol. Rep. 2018, 40, 1093–1102. [Google Scholar] [CrossRef] [PubMed]
  154. Papoutsoglou, P.; Rodrigues-Junior, D.M.; Moren, A.; Bergman, A.; Ponten, F.; Coulouarn, C.; Caja, L.; Heldin, C.H.; Moustakas, A. The noncoding MIR100HG RNA enhances the autocrine function of transforming growth factor beta signaling. Oncogene 2021, 40, 3748–3765. [Google Scholar] [CrossRef] [PubMed]
  155. Ko, C.C.; Hsieh, Y.Y.; Yang, P.M. Long Non-Coding RNA MIR31HG Promotes the Transforming Growth Factor beta-Induced Epithelial-Mesenchymal Transition in Pancreatic Ductal Adenocarcinoma Cells. Int. J. Mol. Sci. 2022, 23, 6559. [Google Scholar] [CrossRef] [PubMed]
  156. Hu, M.L.; Wang, X.Y.; Chen, W.M. TGF-β1 upregulates the expression of lncRNA UCA1 and its downstream HXK2 to promote the growth of hepatocellular carcinoma. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 4846–4854. [Google Scholar] [CrossRef] [PubMed]
  157. Li, Y.; Guo, D.; Ren, M.; Zhao, Y.; Wang, X.; Chen, Y.; Liu, Y.; Lu, G.; He, S. Long non-coding RNA SNAI3-AS1 promotes the proliferation and metastasis of hepatocellular carcinoma by regulating the UPF1/Smad7 signalling pathway. J. Cell. Mol. Med. 2019, 23, 6271–6282. [Google Scholar] [CrossRef] [PubMed]
  158. Yang, X.; Cai, J.B.; Peng, R.; Wei, C.Y.; Lu, J.C.; Gao, C.; Shen, Z.Z.; Zhang, P.F.; Huang, X.Y.; Ke, A.W.; et al. The long noncoding RNA NORAD enhances the TGF-β pathway to promote hepatocellular carcinoma progression by targeting miR-202-5p. J. Cell. Physiol. 2019, 234, 12051–12060. [Google Scholar] [CrossRef]
  159. Dong, H.; Zhang, Y.; Xu, Y.; Ma, R.; Liu, L.; Luo, C.; Jiang, W. Downregulation of long non-coding RNA MEG3 promotes proliferation, migration, and invasion of human hepatocellular carcinoma cells by upregulating TGF-β1. Acta Biochim. Biophys. Sin. 2019, 51, 645–652. [Google Scholar] [CrossRef]
  160. Zhang, L.; Niu, H.; Ma, J.; Yuan, B.Y.; Chen, Y.H.; Zhuang, Y.; Chen, G.W.; Zeng, Z.C.; Xiang, Z.L. The molecular mechanism of LncRNA34a-mediated regulation of bone metastasis in hepatocellular carcinoma. Mol. Cancer 2019, 18, 120. [Google Scholar] [CrossRef]
  161. Sakai, S.; Ohhata, T.; Kitagawa, K.; Uchida, C.; Aoshima, T.; Niida, H.; Suzuki, T.; Inoue, Y.; Miyazawa, K.; Kitagawa, M. Long Noncoding RNA ELIT-1 Acts as a Smad3 Cofactor to Facilitate TGFbeta/Smad Signaling and Promote Epithelial-Mesenchymal Transition. Cancer Res. 2019, 79, 2821–2838. [Google Scholar] [CrossRef] [PubMed]
  162. Liang, J.; Liao, J.; Liu, T.; Wang, Y.; Wen, J.; Cai, N.; Huang, Z.; Xu, W.; Li, G.; Ding, Z.; et al. Comprehensive analysis of TGF-β-induced mRNAs and ncRNAs in hepatocellular carcinoma. Aging 2020, 12, 19399–19420. [Google Scholar] [CrossRef] [PubMed]
  163. Huang, W.J.; Tian, X.P.; Bi, S.X.; Zhang, S.R.; He, T.S.; Song, L.Y.; Yun, J.P.; Zhou, Z.G.; Yu, R.M.; Li, M. The beta-catenin/TCF-4-LINC01278-miR-1258-Smad2/3 axis promotes hepatocellular carcinoma metastasis. Oncogene 2020, 39, 4538–4550. [Google Scholar] [CrossRef] [PubMed]
  164. Chen, E.; Zhou, B.; Bian, S.; Ni, W.; Chen, Z. The lncRNA B3GALT5-AS1 Functions as an HCC Suppressor by Regulating the miR-934/UFM1 Axis. J. Oncol. 2021, 2021, 1776432. [Google Scholar] [CrossRef]
  165. Chen, Z.; Xiang, L.; Li, L.; Ou, H.; Fang, Y.; Xu, Y.; Liu, Q.; Hu, Z.; Huang, Y.; Li, X.; et al. TGF-β1 induced deficiency of linc00261 promotes epithelial-mesenchymal-transition and stemness of hepatocellular carcinoma via modulating SMAD3. J. Transl. Med. 2022, 20, 75. [Google Scholar] [CrossRef] [PubMed]
  166. Zhang, Z.; Zhou, Y.; Jia, Y.; Wang, C.; Zhang, M.; Xu, Z. PRR34-AS1 promotes exosome secretion of VEGF and TGF-β via recruiting DDX3X to stabilize Rab27a mRNA in hepatocellular carcinoma. J. Transl. Med. 2022, 20, 491. [Google Scholar] [CrossRef] [PubMed]
  167. Zhang, L.; Song, X.; Chen, X.; Wang, Q.; Zheng, X.; Wu, C.; Jiang, J. Circular RNA CircCACTIN Promotes Gastric Cancer Progression by Sponging MiR-331-3p and Regulating TGFBR1 Expression. Int. J. Biol. Sci. 2019, 15, 1091–1103. [Google Scholar] [CrossRef] [PubMed]
  168. Qiu, S.; Li, B.; Xia, Y.; Xuan, Z.; Li, Z.; Xie, L.; Gu, C.; Lv, J.; Lu, C.; Jiang, T.; et al. CircTHBS1 drives gastric cancer progression by increasing INHBA mRNA expression and stability in a ceRNA- and RBP-dependent manner. Cell Death Dis. 2022, 13, 266. [Google Scholar] [CrossRef]
  169. Zhou, Y.; Zhang, Q.; Liao, B.; Qiu, X.; Hu, S.; Xu, Q. circ_0006089 promotes gastric cancer growth, metastasis, glycolysis, and angiogenesis by regulating miR-361-3p/TGFB1. Cancer Sci. 2022, 113, 2044–2055. [Google Scholar] [CrossRef]
  170. Li, F.; Tang, H.; Zhao, S.; Gao, X.; Yang, L.; Xu, J. Circ-E-Cad encodes a protein that promotes the proliferation and migration of gastric cancer via the TGF-β/Smad/C-E-Cad/PI3K/AKT pathway. Mol. Carcinog. 2023, 62, 360–368. [Google Scholar] [CrossRef]
  171. Shang, A.; Gu, C.; Wang, W.; Wang, X.; Sun, J.; Zeng, B.; Chen, C.; Chang, W.; Ping, Y.; Ji, P.; et al. Exosomal circPACRGL promotes progression of colorectal cancer via the miR-142-3p/miR-506-3p- TGF-β1 axis. Mol. Cancer 2020, 19, 117. [Google Scholar] [CrossRef] [PubMed]
  172. Zheng, L.; Liang, H.; Zhang, Q.; Shen, Z.; Sun, Y.; Zhao, X.; Gong, J.; Hou, Z.; Jiang, K.; Wang, Q.; et al. circPTEN1, a circular RNA generated from PTEN, suppresses cancer progression through inhibition of TGF-β/Smad signaling. Mol. Cancer 2022, 21, 41. [Google Scholar] [CrossRef]
  173. Shi, X.; Yang, J.; Liu, M.; Zhang, Y.; Zhou, Z.; Luo, W.; Fung, K.M.; Xu, C.; Bronze, M.S.; Houchen, C.W.; et al. Circular RNA ANAPC7 Inhibits Tumor Growth and Muscle Wasting via PHLPP2-AKT-TGF-β Signaling Axis in Pancreatic Cancer. Gastroenterology 2022, 162, 2004–2017.e2. [Google Scholar] [CrossRef] [PubMed]
  174. Zhang, B.; Liu, Z.; Cao, K.; Shan, W.; Liu, J.; Wen, Q.; Wang, R. Circ-SPECC1 modulates TGFbeta2 and autophagy under oxidative stress by sponging miR-33a to promote hepatocellular carcinoma tumorigenesis. Cancer Med. 2020, 9, 5999–6008. [Google Scholar] [CrossRef] [PubMed]
  175. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  176. Peery, A.F.; Crockett, S.D.; Murphy, C.C.; Lund, J.L.; Dellon, E.S.; Williams, J.L.; Jensen, E.T.; Shaheen, N.J.; Barritt, A.S.; Lieber, S.R.; et al. Burden and Cost of Gastrointestinal, Liver, and Pancreatic Diseases in the United States: Update 2018. Gastroenterology 2019, 156, 254–272.e11. [Google Scholar] [CrossRef] [PubMed]
  177. Japanese Gastric Cancer Association. Japanese gastric cancer treatment guidelines 2014 (ver. 4). Gastric Cancer 2017, 20, 1–19. [Google Scholar] [CrossRef]
  178. McFarland, E.G.; Levin, B.; Lieberman, D.A.; Pickhardt, P.J.; Johnson, C.D.; Glick, S.N.; Brooks, D.; Smith, R.A. Revised colorectal screening guidelines: Joint effort of the American Cancer Society, U.S. Multisociety Task Force on Colorectal Cancer, and American College of Radiology. Radiology 2008, 248, 717–720. [Google Scholar] [CrossRef]
  179. Teixeira, A.F.; Ten Dijke, P.; Zhu, H.J. On-Target Anti-TGF-β Therapies Are Not Succeeding in Clinical Cancer Treatments: What Are Remaining Challenges? Front. Cell Dev. Biol. 2020, 8, 605. [Google Scholar] [CrossRef]
  180. Morris, J.C.; Tan, A.R.; Olencki, T.E.; Shapiro, G.I.; Dezube, B.J.; Reiss, M.; Hsu, F.J.; Berzofsky, J.A.; Lawrence, D.P. Phase I study of GC1008 (fresolimumab): A human anti-transforming growth factor-β (TGFbeta) monoclonal antibody in patients with advanced malignant melanoma or renal cell carcinoma. PLoS ONE 2014, 9, e90353. [Google Scholar] [CrossRef]
  181. Papageorgis, P.; Stylianopoulos, T. Role of TGFbeta in regulation of the tumor microenvironment and drug delivery (review). Int. J. Oncol. 2015, 46, 933–943. [Google Scholar] [CrossRef]
  182. 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]
  183. Kelley, R.K.; Gane, E.; Assenat, E.; Siebler, J.; Galle, P.R.; Merle, P.; Hourmand, I.O.; Cleverly, A.; Zhao, Y.; Gueorguieva, I.; et al. A Phase 2 Study of Galunisertib (TGF-β1 Receptor Type I Inhibitor) and Sorafenib in Patients with Advanced Hepatocellular Carcinoma. Clin. Transl. Gastroenterol. 2019, 10, e00056. [Google Scholar] [CrossRef] [PubMed]
  184. Yamazaki, T.; Gunderson, A.J.; Gilchrist, M.; Whiteford, M.; Kiely, M.X.; Hayman, A.; O’Brien, D.; Ahmad, R.; Manchio, J.V.; Fox, N.; et al. Galunisertib plus neoadjuvant chemoradiotherapy in patients with locally advanced rectal cancer: A single-arm, phase 2 trial. Lancet Oncol. 2022, 23, 1189–1200. [Google Scholar] [CrossRef]
  185. Bai, X.; Yi, M.; Jiao, Y.; Chu, Q.; Wu, K. Blocking TGF-β Signaling to Enhance the Efficacy of Immune Checkpoint Inhibitor. OncoTargets Ther. 2019, 12, 9527–9538. [Google Scholar] [CrossRef]
  186. Singh, A.; Settleman, J. EMT, cancer stem cells and drug resistance: An emerging axis of evil in the war on cancer. Oncogene 2010, 29, 4741–4751. [Google Scholar] [CrossRef] [PubMed]
  187. Knudson, K.M.; Hicks, K.C.; Luo, X.; Chen, J.Q.; Schlom, J.; Gameiro, S.R. M7824, a novel bifunctional anti-PD-L1/TGFbeta Trap fusion protein, promotes anti-tumor efficacy as monotherapy and in combination with vaccine. Oncoimmunology 2018, 7, e1426519. [Google Scholar] [CrossRef] [PubMed]
  188. Paz-Ares, L.; Kim, T.M.; Vicente, D.; Felip, E.; Lee, D.H.; Lee, K.H.; Lin, C.C.; Flor, M.J.; Di Nicola, M.; Alvarez, R.M.; et al. Bintrafusp Alfa, a Bifunctional Fusion Protein Targeting TGF-β and PD-L1, in Second-Line Treatment of Patients With NSCLC: Results From an Expansion Cohort of a Phase 1 Trial. J. Thorac. Oncol. 2020, 15, 1210–1222. [Google Scholar] [CrossRef]
  189. Kang, Y.K.; Bang, Y.J.; Kondo, S.; Chung, H.C.; Muro, K.; Dussault, I.; Helwig, C.; Osada, M.; Doi, T. Safety and Tolerability of Bintrafusp Alfa, a Bifunctional Fusion Protein Targeting TGFbeta and PD-L1, in Asian Patients with Pretreated Recurrent or Refractory Gastric Cancer. Clin. Cancer Res. 2020, 26, 3202–3210. [Google Scholar] [CrossRef]
  190. Liu, D.; Zhou, J.; Wang, Y.; Li, M.; Jiang, H.; Liu, Y.; Yin, X.; Ge, M.; Xiang, X.; Ying, J.; et al. Bifunctional anti-PD-L1/TGF-βRII agent SHR-1701 in advanced solid tumors: A dose-escalation, dose-expansion, and clinical-expansion phase 1 trial. BMC Med. 2022, 20, 408. [Google Scholar] [CrossRef]
  191. Kim, B.G.; Malek, E.; Choi, S.H.; Ignatz-Hoover, J.J.; Driscoll, J.J. Novel therapies emerging in oncology to target the TGF-β pathway. J. Hematol. Oncol. 2021, 14, 55. [Google Scholar] [CrossRef]
  192. Sheen, Y.Y.; Kim, M.J.; Park, S.A.; Park, S.Y.; Nam, J.S. Targeting the Transforming Growth Factor-β Signaling in Cancer Therapy. Biomol. Ther. 2013, 21, 323–331. [Google Scholar] [CrossRef] [PubMed]
  193. Leight, J.L.; Wozniak, M.A.; Chen, S.; Lynch, M.L.; Chen, C.S. Matrix rigidity regulates a switch between TGF-β1-induced apoptosis and epithelial-mesenchymal transition. Mol. Biol. Cell 2012, 23, 781–791. [Google Scholar] [CrossRef] [PubMed]
  194. Sun, N.; Taguchi, A.; Hanash, S. Switching Roles of TGF-β in Cancer Development: Implications for Therapeutic Target and Biomarker Studies. J. Clin. Med. 2016, 5, 109. [Google Scholar] [CrossRef] [PubMed]
  195. Busch, S.; Acar, A.; Magnusson, Y.; Gregersson, P.; Ryden, L.; Landberg, G. TGF-β receptor type-2 expression in cancer-associated fibroblasts regulates breast cancer cell growth and survival and is a prognostic marker in pre-menopausal breast cancer. Oncogene 2015, 34, 27–38. [Google Scholar] [CrossRef] [PubMed]
  196. Suzuki, R.; Fukui, T.; Kishimoto, M.; Miyamoto, S.; Takahashi, Y.; Takeo, M.; Mitsuyama, T.; Sakaguchi, Y.; Uchida, K.; Nishio, A.; et al. Smad2/3 linker phosphorylation is a possible marker of cancer stem cells and correlates with carcinogenesis in a mouse model of colitis-associated colorectal cancer. J. Crohns Colitis 2015, 9, 565–574. [Google Scholar] [CrossRef] [PubMed]
  197. Voorneveld, P.W.; Jacobs, R.J.; Kodach, L.L.; Hardwick, J.C. A Meta-Analysis of SMAD4 Immunohistochemistry as a Prognostic Marker in Colorectal Cancer. Transl. Oncol. 2015, 8, 18–24. [Google Scholar] [CrossRef] [PubMed]
  198. Li, J.; Mu, S.; Mu, L.; Zhang, X.; Pang, R.; Gao, S. Transforming growth factor-β-1 is a serum biomarker of radiation-induced pneumonitis in esophageal cancer patients treated with thoracic radiotherapy: Preliminary results of a prospective study. OncoTargets Ther. 2015, 8, 1129–1136. [Google Scholar] [CrossRef]
  199. Hosseini, R.; Hosseinzadeh, N.; Asef-Kabiri, L.; Akbari, A.; Ghezelbash, B.; Sarvnaz, H.; Akbari, M.E. Small extracellular vesicle TGF-β in cancer progression and immune evasion. Cancer Gene Ther. 2023, 30, 1309–1322. [Google Scholar] [CrossRef]
  200. de Miguel-Perez, D.; Russo, A.; Gunasekaran, M.; Buemi, F.; Hester, L.; Fan, X.; Carter-Cooper, B.A.; Lapidus, R.G.; Peleg, A.; Arroyo-Hernandez, M.; et al. Baseline extracellular vesicle TGF-β is a predictive biomarker for response to immune checkpoint inhibitors and survival in non-small cell lung cancer. Cancer 2023, 129, 521–530. [Google Scholar] [CrossRef]
  201. Mitra, M.S.; Lancaster, K.; Adedeji, A.O.; Palanisamy, G.S.; Dave, R.A.; Zhong, F.; Holdren, M.S.; Turley, S.J.; Liang, W.C.; Wu, Y.; et al. A Potent Pan-TGFbeta Neutralizing Monoclonal Antibody Elicits Cardiovascular Toxicity in Mice and Cynomolgus Monkeys. Toxicol. Sci. 2020, 175, 24–34. [Google Scholar] [CrossRef]
  202. Rak, G.D.; White, M.R.; Augustine-Rauch, K.; Newsome, C.; Graziano, M.J.; Schulze, G.E. Intermittent dosing of the transforming growth factor beta receptor 1 inhibitor, BMS-986260, mitigates class-based cardiovascular toxicity in dogs but not rats. J. Appl. Toxicol. 2020, 40, 931–946. [Google Scholar] [CrossRef]
  203. Mortensen, R.E.J.; Holmstrom, M.O.; Lisle, T.L.; Hasselby, J.P.; Willemoe, G.L.; Met, O.; Marie Svane, I.; Johansen, J.; Nielsen, D.L.; Chen, I.M.; et al. Pre-existing TGF-β-specific T-cell immunity in patients with pancreatic cancer predicts survival after checkpoint inhibitors combined with radiotherapy. J. Immunother. Cancer 2023, 11, e006432. [Google Scholar] [CrossRef]
  204. Rahavi, H.; Alizadeh-Navaei, R.; Tehrani, M. Efficacy of therapies targeting TGF-β in solid tumors: A systematic review and meta-analysis of clinical trials. Immunotherapy 2023, 15, 283–292. [Google Scholar] [CrossRef]
Figure 1. Schematic diagram of transforming growth factor-β (TGF-β) signaling pathways. A latent complex in the collagen comprised an N-terminal peptide with a latency-associated peptide (LAP) and a mature C-terminal fragment. The N-terminal signal peptide is removed, the pro-peptide accumulates into a disulfide-linked homodimer, and latent TGF-β is transformed into homodimeric ligands. This latent TGF-β molecule can create a complex with latent TGF-β-binding protein (LTBP) that mediates its deposition to the extracellular matrix (ECM), mediating the release of active TGF-β via interaction. TGF-β can also be activated by anchoring to glycoprotein-A repetitions predominant (GARP). Activated TGF-β phosphorylates TGF-β type 2 receptor (TGFBR2), which recruits TGF-β type 1 receptor (TGFBR1) and connects to a tetrameric complex composed of TGF receptors and then activates canonical and noncanonical TGF-β pathways. In the case of the canonical SMAD pathway, TGBFR1 recruits and phosphorylates specific SMADs such as SMAD2 and SMAD3, which can form heteromeric complexes with SMAD4. Then, SMAD2/3 and the SMAD4 complex translate into the nucleus to regulate target gene transcription. TGF-β also activates non-canonical pathways when connected to other downstream factors, such as SHC/GRB2/SODS-MAPK pathways, RHO-like GTPase signaling pathways, TRAF4/6, PAR6, and PI3K/AKT pathways.
Figure 1. Schematic diagram of transforming growth factor-β (TGF-β) signaling pathways. A latent complex in the collagen comprised an N-terminal peptide with a latency-associated peptide (LAP) and a mature C-terminal fragment. The N-terminal signal peptide is removed, the pro-peptide accumulates into a disulfide-linked homodimer, and latent TGF-β is transformed into homodimeric ligands. This latent TGF-β molecule can create a complex with latent TGF-β-binding protein (LTBP) that mediates its deposition to the extracellular matrix (ECM), mediating the release of active TGF-β via interaction. TGF-β can also be activated by anchoring to glycoprotein-A repetitions predominant (GARP). Activated TGF-β phosphorylates TGF-β type 2 receptor (TGFBR2), which recruits TGF-β type 1 receptor (TGFBR1) and connects to a tetrameric complex composed of TGF receptors and then activates canonical and noncanonical TGF-β pathways. In the case of the canonical SMAD pathway, TGBFR1 recruits and phosphorylates specific SMADs such as SMAD2 and SMAD3, which can form heteromeric complexes with SMAD4. Then, SMAD2/3 and the SMAD4 complex translate into the nucleus to regulate target gene transcription. TGF-β also activates non-canonical pathways when connected to other downstream factors, such as SHC/GRB2/SODS-MAPK pathways, RHO-like GTPase signaling pathways, TRAF4/6, PAR6, and PI3K/AKT pathways.
Biomolecules 13 01551 g001
Figure 2. Epithelial–mesenchymal transition (EMT) process. Epithelial cells show apical-basal polarity, and they are connected strongly through tight junctions and adherence junctions, and desmosomes are bound to the extracellular membranes. TGF-β activates the SMAD2/3/4 complex and then regulates EMT-inducing transcription factors such as SNAIL, TWIST, Slug, DMNT, and ZEB. These factors repress epithelial genes and include mesenchymal genes. Expression of these genes leads to cellular alterations, including disassembly of epithelial cell-cell junctions and deficiency of apical-basal cell polarity, where cells become disseminated with the ability for invasion. Activation of non-canonical signaling responses such as MAP kinases and the PI3K/Akt pathway in response to TGF-β has been connected to TGF-β-induced EMT via their mediation of cytoskeleton organization, cell growth, apoptosis, motility, or invasion. Through EMT, the down-regulation of epithelial markers such as E-cadherin, occludins, and claudins is shown, whereas mesenchymal markers such as N-cadherin, vimentin, and fibronectin are up-regulated.
Figure 2. Epithelial–mesenchymal transition (EMT) process. Epithelial cells show apical-basal polarity, and they are connected strongly through tight junctions and adherence junctions, and desmosomes are bound to the extracellular membranes. TGF-β activates the SMAD2/3/4 complex and then regulates EMT-inducing transcription factors such as SNAIL, TWIST, Slug, DMNT, and ZEB. These factors repress epithelial genes and include mesenchymal genes. Expression of these genes leads to cellular alterations, including disassembly of epithelial cell-cell junctions and deficiency of apical-basal cell polarity, where cells become disseminated with the ability for invasion. Activation of non-canonical signaling responses such as MAP kinases and the PI3K/Akt pathway in response to TGF-β has been connected to TGF-β-induced EMT via their mediation of cytoskeleton organization, cell growth, apoptosis, motility, or invasion. Through EMT, the down-regulation of epithelial markers such as E-cadherin, occludins, and claudins is shown, whereas mesenchymal markers such as N-cadherin, vimentin, and fibronectin are up-regulated.
Biomolecules 13 01551 g002
Figure 3. Tumor microenvironment-related cells and extracellular elements as regulators of cancer progression: The schematic illustrates how the interaction between the tumor cells and the surrounding stroma cells involves myeloid-derived suppressor cells (MDSCs), macrophages, T cells, B cells, natural killer (NK) cells, dendritic cells (DCs), and cancer-associated fibroblasts (CAFs). TGF-β modulates cancer immunity. TGF-β suppresses T-helper 1 (Th1), M1, and N1 but facilitates Th2, M2, and N2 differentiation and advancement in the tumor microenvironment. As well, TGF-β inhibits NK and DC functions.
Figure 3. Tumor microenvironment-related cells and extracellular elements as regulators of cancer progression: The schematic illustrates how the interaction between the tumor cells and the surrounding stroma cells involves myeloid-derived suppressor cells (MDSCs), macrophages, T cells, B cells, natural killer (NK) cells, dendritic cells (DCs), and cancer-associated fibroblasts (CAFs). TGF-β modulates cancer immunity. TGF-β suppresses T-helper 1 (Th1), M1, and N1 but facilitates Th2, M2, and N2 differentiation and advancement in the tumor microenvironment. As well, TGF-β inhibits NK and DC functions.
Biomolecules 13 01551 g003
Table 1. Frequency of genetic alterations in TGF-β pathway-related genes in GI cancers. GI cancer = gastrointestinal cancer; CRC = colorectal cancer; PC = pancreatic cancer; EC = esophageal cancer; GC = gastric cancer; HCC = hepatocellular carcinoma; TGF-β = transforming growth factor-β; Ref = reference; N.A. = not applicable.
Table 1. Frequency of genetic alterations in TGF-β pathway-related genes in GI cancers. GI cancer = gastrointestinal cancer; CRC = colorectal cancer; PC = pancreatic cancer; EC = esophageal cancer; GC = gastric cancer; HCC = hepatocellular carcinoma; TGF-β = transforming growth factor-β; Ref = reference; N.A. = not applicable.
Cancer TypeTGF-β 1/2/3TGFBR1TGFBR2SMAD1SMAD2SMAD4Ref
ECMutation 4%
Down-regulated 20%
Mutation 1%
Down-regulated 5%
Mutation 1%
Down-regulated 7%
Mutation 1%
Down-regulated 8%
Mutation 1%
Down-regulated 20%
Mutation 6–22%
Complete deletion 9.8%
[46,47,48]
GCMutation 4–13%
Up-regulated 22.8%
Mutation 1%
Loss 7%
Down-regulated 45%N.A.Mutation 0%Mutation 6.0%
Up-regulated 0.2%
Complete deletion 3.7%
LOH 61%
[48,49,50,51]
CRCMutation 1%
Loss 8%
Up-regulated 58.8/26.4/23.5%
Mutation 1%
Down-regulated 8%
Mutation 11–30%Mutation 8.6%Mutation 2.3–10.3%Mutation 12–38%
Complete deletion 0.9%
LOH 61%
[39,48,49,52,53,54]
PCMutation 1%
Loss 20%
Up-regulated 47%
Mutation 1–5%
Down-regulated 20%
Mutation 4–7%
Somatic alteration 4.1%
N.A.N.A.Mutation 20.4%
Deletion 13%
LOH 90%
[49,55]
HCC TGF-β 1/2/3Mutation 1%Mutation 3.0%Mutation 3.0%Mutation 0.8–6%
Complete deletion 0.2%
[48,49]
Biliary tract
cancer
Mutation 1%
Down-regulated 4%
N.A.Mutation 50%N.A.N.A.Mutation 3.9–16%[48,56,57]
Table 2. ncRNAs associated with TGF-β signaling in GI cancer for the past 5 years. ncRNAs = non-coding RNAs; TGF-β = transforming growth factor-β; GI cancer = gastrointestinal cancer; Ref = reference; CRC = colorectal cancer; PC = pancreatic cancer; EC = esophageal cancer; GC = gastric cancer; HCC = hepatocellular carcinoma; lncRNA = long non-coding RNA; circRNA = circular RNA; piRNA = PIWI interacting RNA; EMT = epithelial-mesenchymal transition; MSI = microsatelite instability; ICB = immune checkpoint blockade; EndMT = endothelial-to-mesenchymal transition; TAM = tumor-associated macrophage; EV = extracellular vesicles; Down = downregulation; Up = upregulation; N.A. = not applicable.
Table 2. ncRNAs associated with TGF-β signaling in GI cancer for the past 5 years. ncRNAs = non-coding RNAs; TGF-β = transforming growth factor-β; GI cancer = gastrointestinal cancer; Ref = reference; CRC = colorectal cancer; PC = pancreatic cancer; EC = esophageal cancer; GC = gastric cancer; HCC = hepatocellular carcinoma; lncRNA = long non-coding RNA; circRNA = circular RNA; piRNA = PIWI interacting RNA; EMT = epithelial-mesenchymal transition; MSI = microsatelite instability; ICB = immune checkpoint blockade; EndMT = endothelial-to-mesenchymal transition; TAM = tumor-associated macrophage; EV = extracellular vesicles; Down = downregulation; Up = upregulation; N.A. = not applicable.
Cancer TypeAlterationsInteractionCommentsRef
miRNA
miR-130a-3pECDownSuppressionTGF-β/miR-130a-3p/SMAD4 pathway mediated by EMT.[112]
miR-93-5pECUpActivationPrompts the migration and inhibits the apoptosis by targeting TGFβR2.[113]
miR-21-5pECUpActivationActivates M2 macrophages and, in turn, EMT through TGF-β/Smad2 signaling.[114]
miR-181aECUpActivationPromotes EMT through the TGF-β/SMAD pathway. [115]
miR-21ECUpActivationTGF-βin CTLs may serve as an excellent biomarker.[116]
miR-577GCUpActivationTGF-β-miR-577-SDPR axis enhances metastasis via the NF-κB pathway. [117]
miR-187GCUpSuppressionSuppresses CDDP resistance. [118]
miR-135bGCUpActivationPrompts growth and migration by negatively modulating the expression of TGFBR2.[119]
miR-130a-3pGCUpActivationTGF-β1/SMAD3 signaling via miR-130a-3p increased proliferation.[120]
miR-106aGCUpActivationCancer-derived exosomes transport miR-106a to peritoneal mesothelial cells.[121]
miR-200cGCUpSuppressionNegatively associated with EMT, angiogenesis, and hypoxia.[122]
miR-875-5pGCUpSuppressionDownregulates USF2, leading to repression of TGF-β signaling.[123]
miR-31-3pCRCUpActivationPositively related to TGFBR2 deficiency in MSI CRC.[124]
miR-4666-3p and miR-329CRCDownSuppressionRegulate CRC stemness.[125]
miR-186-5pCRCUpSuppressionSuppresses cell cycle via inhibition of SMAD6/7. [126]
miRNA-146b, miRNA-155, and miRNA-22CRCN.A.N.A.Three miRNAs identify SMAD2, SMAD4, and TGFBR2 as companions for ICB.[127]
miR-200CRCUpActivationEV-encapsulated miR-200 regulates tumor-stroma crosstalk in CRC.[128]
miR-20a-5pCRCDownSuppressionNegatively correlated with LAMTOR5-AS and TGFBR2.[129]
miR-552CRCDownSuppressionSuppresses 5-FU resistance.[130]
miR-495-3pCRCUpSuppressionNegatively mediates TGFβR1, TGFβR2, and SMAD4 genes.[131]
miR-501-3pPCUpActivationM2 macrophage-derived exosomal miR-501-3p suppresses the TGFBR3 gene pathway. [111]
miR-21PCUpActivationM2 macrophage-derived exosomal miR-501-3p suppresses the TGFBR3 gene pathway. [111]
miR-622,PCUpSuppressionTargets HULC and suppresses invasion by blocking EMT signaling through EV transfer.[132]
miR-492PCUpActivationPromotes malignant behavior through the NR2C1-TGF-β/Smad3 pathway. [109]
miR-133aHCCUpSuppressionRepressed malignant behavior of HCC via TGF-β/Smad3 signaling.[133]
miR-362-3pHCCUpActivationRegulates EMT by modulating CD82 and TAM.[134]
miR-494HCCUpActivationRegulates EndMT via SIRT3/TGF-β/SMAD signaling.[135]
miR-141HCCUpSuppressionBlocks growth and invasion by directly downregulating TGFβR1.[136]
miR-140-5pHCCDownSuppressionServes as an EMT inhibitor.[137]
miR-17-5pHCCDownSuppressionInhibits TGFβR2 expression and EMT.[138]
miR-324-5pGallbladder cancerDownSuppressionTargets TGFB2 expression to inhibit GBC cell metastatic behaviors.[139]
lnc RNAs
NCK1-AS1ECUpActivationPrompts migration and invasion.[140]
FAM83H-AS1ECUpActivationInduced by TGF-β and increased migration and invasion.[141]
LINC00665GCUpActivationIncreases growth, invasion, and metastasis.[142]
SGO1-AS1GCUpSuppressionSuppresses the TGF-β pathway and impaired gastric carcinoma metastasis.[143]
Nr2F1-AS1GCUpActivationPromotes progression by regulating the miR-29a-3p/VAMP7 axis induced by EMT[144]
MBNL1-AS1GCDownSuppressionRegulates the miR-424-5p/Smad7 axis and prompts TGF-β/EMT pathways.[106]
XLOC_004787 GCUpActivationEnhances growth and migration via TGF-signaling and blockade of mir-203a-3p.[145]
LOC646329CRCUpActivationSuppresses CRC development via sponging miR-29b and modulates TGFB signaling.[146]
SNHG6CRCUpActivationStimulates TGF-β/Smad signaling via targeting UPF1 and inducing EMT.[147]
CASC9CRCUpActivationPredicts the prognostis for patients with CRC.[148]
MIR-22HGCRCDownSuppressionRelated to CD8A and induced T cell infiltration.[149]
TUG1CRCUpActivationTUG1/TWIST1/EMT signaling is associated with CRC metastasis prompted by TGF-β. [150]
CTBP1-AS2CRCUpActivationStimulates the TGF-β/SMAD2/3 pathway by blocking miR-93-5p.[151]
LINC00941CRCUpActivationStimulates the TGF-β/SMAD2/3 axis.[104]
VPS9D1-AS1CRCUpActivationVPS9D1-AS1/TGF-β signaling enhances tumor proliferation and immune evasion. [152]
PVT1PCUpActivationServes as an oncogene via EMT via the TGF-β/Smad pathway.[153]
Linc00462PCUpActivationEnhances TGFBR1/2 expression and activates the SMAD2/3 pathway.[108]
MIR100HGPCUpActivationRegulates TGFβ signaling through TGFβ1 production.[154]
MIR31HGPCUpActivationMIR31HG inhibits TGFβ-induced EMT and cancer cell migration.[155]
SBF2-AS1HCCUpActivationRegulates TGFBR1 via sponging miR-140-5p.[105]
UCA1HCCUpActivationTGF-β1 prompts the growth of HCC via the upregulation of UCA1 and HXK2.[156]
SNAI3-AS1HCCUpActivationRegulates UPF1 and activates the TGF-β/Smad pathway, leading to HCC progression.[157]
NORADHCCUpActivationRegulates the TGF-β pathway to stimulate HCC development by miR-202-5p.[158]
MEG3HCCUpSuppressionIncrease growth, migration, and invasion.[159]
LncRNA34aHCCUpActivationLncRNA34a is positively associated with bone metastasis.[160]
ELIT-1HCCUpActivationPositively regulates TGFβ/Smad3 signaling and EMT.[161]
SLC7A11-AS1HCCUpActivationSLC7A11-AS1 and hsa_circ_0006123 are implicated in the EMT induced by TGF-β.[162]
LINC01278HCCUpActivationβ-catenin/TCF-4-LINC01278-miR-1258-Smad2/3 feedback loop is associated with HCC metastasis.[163]
B3GALT5-AS1 HCCUpActivationSuppresses CD4 T cell invasion through TGF-β signaling.[164]
LINC00261HCCUpSuppressionRepresses EMT and stem-like change by blocking TGF-β1/SMAD3 signaling.[165]
PRR34-AS1HCCUpActivationPrompts the exosome secretion of VEGF and TGF-β.[166]
circ RNAs
circCACTINGCUpActivationThe circCACTIN/miR-331-3p/TGFBR1 axis promotes migration, invasion, and EMT.[167]
circTHBS1GCUpActivationPromotes malignant behavior and EMT.[168]
circ_0006089GCUpActivationPrompts glycolysis and angiogenesis via the miR-361-3p/TGFB1 pathway.[169]
Circ-E-CadGCUpActivationRegulates growth, migration, and EMT via PI3K/AKT signaling.[170]
circPACRGLCRCUpActivationExosomal circPACRGL enhanced malignant behavior via the miR-142-3p/miR-506-3p-TGF-β1 axis.[171]
circPTEN1CRCDownSuppressionDisordered SMAD4 interaction with SMAD2/associated with EMT upon TGF-β activation.[172]
circANAPC7PCDownSuppressionRepresses tumor proliferation via the CREB-miR-373-PHLPP2 axis and TGF-β down-regulation.[173]
circSPECC1HCCUpActivationMediates TGFβ2 and autophagy via miR-33a under oxidative stress.[174]
circGFRA1HCCUpActivationRegulates the miR-498/NAP1L3 axis, which led to HCC development.[107]
Table 3. Clinical trials of pharmacological strategies targeting TGF-β for GI cancer. GI cancer = gastrointestinal cancer; TGF-β = transforming growth factor-β; TGFBR1 = TGF-β2 type receptor; TGFBR2 = TGF-β2 type receptor; PD-L1 = programmed death-1 ligand; CRC = colorectal cancer; PC = pancreatic cancer; EC = esophageal cancer; GC = gastric cancer; GEJC = gastroesophageal junction cancer; HCC = hepatocellular carcinoma; CCC = cholangiocellular carcinoma; COX = cyclooxygenase.
Table 3. Clinical trials of pharmacological strategies targeting TGF-β for GI cancer. GI cancer = gastrointestinal cancer; TGF-β = transforming growth factor-β; TGFBR1 = TGF-β2 type receptor; TGFBR2 = TGF-β2 type receptor; PD-L1 = programmed death-1 ligand; CRC = colorectal cancer; PC = pancreatic cancer; EC = esophageal cancer; GC = gastric cancer; GEJC = gastroesophageal junction cancer; HCC = hepatocellular carcinoma; CCC = cholangiocellular carcinoma; COX = cyclooxygenase.
StrategyAgentsTargetCancer TypeIdentifierStatusResultsComments
Antibody
SAR439459TGF-β 1/2/3CRC, HCCNCT03192345IbTermninatedMonotherapy
+Cemiplimab
NIS793TGF-β 1/2/3PCNCT04390763IIActiveMonotherapy
+spartalizumab
NIS793TGF-β 1/2/3PCNCT04935359IIIActive, not-recruitingGemcitabine and nab-paclitaxel ± NIS793
NIS793TGF-β 1/2/3PCNCT05417386IRecruiting+FOLFIRINOX
NIS793TGF-β 1/2/3CRCNCT04952753IIActive, not-recruiting+bevacizumab with mFOLFOX6 or FOLFIRI
ABBV151GARP:TGF-β1PC
HCC
NCT03821935IRecruitingMonotherapy
+ABBV151
GT90001ALK-1GC, EC, HCCNCT04984668I/IIRecruiting+KN046
ALK-1HCCNCT05178043IIRecruiting+NivolumabGT90001
Ligand trap
M7824TGF-β/PD-L1CCC/gallbladder cancerNCT03833661IIActiveMonotherepy
M7824TGF-β/PD-L1Biliary tract cancerNCT04066491IIRecruiting+gemcitabine and cisplatin
M7824TGF-β/PD-L1PCNCT03451773IICompleted+gemcitabine
M7824TGF-β/PD-L1PCNCT04327986ITerminated+M9241 and radiotherapy
M7824TGF-β/PD-L1CRCNCT03436563IIActive, not recruitingMonotherapy
M7824TGF-β/PD-L1Small bowel cancer, CRCNCT04491955IIActive+N-803, M9241, and CV301
M7824TGF-β/PD-L1Small bowel cancer, CRCNCT04708470I/IIRecruitingEntinostat and NHS-IL12 ± M7824
M7824TGF-β/PD-L1ECNCT04595149IIRecruiting+paclitaxel, carboplatin, and radiotherapy
M7824TGF-β/PD-L1Intrahepatic cholangiocarcinomaNCT04708067IRecruiting+Hypofractionated radiation
Small-
molecule inhibitors
GalunisertibTGFBR1HCCNCT02906397IActive +radiotherapy
GalunisertibTGFBR1Rectal cancerNCT02688712IIActive, not recruiting +fluorouracil/capecitabine and radiotherapy
GalunisertibTGFBR1PCNCT02734160IITerminated+durvalumab
GalunisertibTGFBR1HCCNCT01246986IIActive+sorafenib/ramucirumab
GalunisertibTGFBR1PCNCT01373164IICompleted+gemcitabine
VactosertibTGFBR1HCCNCT02160106ICompletedMonotherapy
VactosertibTGFBR1CRCNCT03844750IRecruiting+pembrolizumab
VactosertibTGFBR1GCNCT03698825IIRecruiting+paclitaxel
VactosertibTGFBR1GCNCT04656002IINot yet recruiting+paclitaxel and ramucirumab
VactosertibTGFBR1GCNCT04893252IIRecruiting+durvalumab
VactosertibTGFBR1PCNCT04258072IIActive+irinotecan, fluorouracil and leucovorin
LY3200882TGFBR1CRCNCT04031872IIActive+capecitabine
PF06952229TGFBR1 (ALK1)CRCNCT02116894ICompleted+regorafenib
PF06952229TGFBR1 (ALK1)PC, CRC, HCCNCT03685591IRecruitingMonotherapy
GFH018TGFBR1PC, EC, CRC, HCC, Biliary tract cancerNCT04914286I/IIRecruiting+Toripalimab
TEW-7197ALK4/5PCNCT03666832I/IIRecruiting+FOLFOX
SHR-1701TGFBR2/PD-L1CRCNCT04856787II/IIIRecruiting+BP102 and capecitabine/oxaliplatin
SHR-1701TGFBR2/PD-L1Rectal cancerNCT05300269IIRecruiting+capecitabine/oxaliplatin
SHR-1701TGFBR2/PD-L1GCNCT05149807IEnrolling by invitationMonotherapy
SHR-1701TGFBR2PD-L1HCCNCT04679038I/IIRecruiting+Famitinib
SHR-1701TGFBR2/PD-L1GC/GEJCNCT04950322IIIRecruiting+capecitabine/oxaliplatin
Antisense oligonucleotide
STP705TGF-β/COX-2HCC, CCCNCT04676633IActive, not recruitingMonotherapy
TrabedersenTGF-β mRNA/TGFBR2CRCNCT00844064ICompletedMonotherapy
Vaccine
vigilTGFβI/TGFβIILiver cancerNCT01061840ICompletedMonotherapy
STP705TGF-β/COX-2HCC, CCCNCT04676633IActive, not recruitingMonotherapy
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

Matsuoka, T.; Yashiro, M. The Role of the Transforming Growth Factor-β Signaling Pathway in Gastrointestinal Cancers. Biomolecules 2023, 13, 1551. https://doi.org/10.3390/biom13101551

AMA Style

Matsuoka T, Yashiro M. The Role of the Transforming Growth Factor-β Signaling Pathway in Gastrointestinal Cancers. Biomolecules. 2023; 13(10):1551. https://doi.org/10.3390/biom13101551

Chicago/Turabian Style

Matsuoka, Tasuku, and Masakazu Yashiro. 2023. "The Role of the Transforming Growth Factor-β Signaling Pathway in Gastrointestinal Cancers" Biomolecules 13, no. 10: 1551. https://doi.org/10.3390/biom13101551

APA Style

Matsuoka, T., & Yashiro, M. (2023). The Role of the Transforming Growth Factor-β Signaling Pathway in Gastrointestinal Cancers. Biomolecules, 13(10), 1551. https://doi.org/10.3390/biom13101551

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