Next Article in Journal
Prediction of Antifungal Activity of Antimicrobial Peptides by Transfer Learning from Protein Pretrained Models
Next Article in Special Issue
The TGF-β/UCHL5/Smad2 Axis Contributes to the Pathogenesis of Placenta Accreta
Previous Article in Journal
A Cell-Based Optimised Approach for Rapid and Efficient Gene Editing of Human Pluripotent Stem Cells
Previous Article in Special Issue
TGF-β-3 Induces Different Effects from TGF-β-1 and -2 on Cellular Metabolism and the Spatial Properties of the Human Trabecular Meshwork Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

TGF-β Signaling in Progression of Oral Cancer

1
Department of Biomedical Engineering, Shenzhen University Medicine School, Shenzhen University, Shenzhen 518060, China
2
Key Laboratory of Optoelectronic Devices and Systems, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
3
Central Laboratory, Peking University School of Stomatology, Beijing 100081, China
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(12), 10263; https://doi.org/10.3390/ijms241210263
Submission received: 27 April 2023 / Revised: 26 May 2023 / Accepted: 13 June 2023 / Published: 17 June 2023
(This article belongs to the Special Issue TGF-β Signaling in Human Disease)

Abstract

:
Oral cancer is a common malignancy worldwide, accounting for 1.9% to 3.5% of all malignant tumors. Transforming growth factor β (TGF-β), as one of the most important cytokines, is found to play complex and crucial roles in oral cancers. It may act in a pro-tumorigenic and tumor-suppressive manner; activities of the former include cell cycle progression inhibition, tumor microenvironment preparation, apoptosis promotion, stimulation of cancer cell invasion and metastasis, and suppression of immune surveillance. However, the triggering mechanisms of these distinct actions remain unclear. This review summarizes the molecular mechanisms of TGF-β signal transduction, focusing on oral squamous cell and salivary adenoid systemic carcinomas as well as keratocystic odontogenic tumors. Both the supporting and contrary evidence of the roles of TGF-β is discussed. Importantly, the TGF-β pathway has been the target of new drugs developed in the past decade, some having demonstrated promising therapeutic effects in clinical trials. Therefore, the achievements of TGF-β pathway-based therapeutics and their challenges are also assessed. The summarization and discussion of the updated knowledge of TGF-β signaling pathways will provide insight into the design of new strategies for oral cancer treatment, leading to an improvement in oral cancer outcomes.

1. Introduction

Oral cancer, with over 300,000 diagnoses worldwide annually and 145,000 deaths, is the sixth most common cancer. This major subtype of head and neck cancer affects 1.6 million people worldwide [1,2], but the five-year survival rate has only improved from 54% to 66% thanks to advanced treatment in the past two decades [3]. Oral cancer is most common in patients aged 45 to 64, comprising approximately 50% of all cases, with incidence increasing with age [4,5,6]. Over half of patients are diagnosed at advanced stages [7]. Nowadays, the primary treatments for oral cancer remain surgery resection, radio/chemotherapy, or combination treatment. Various factors should be taken into consideration when choosing treatment strategies, such as the tumor location, tumor size, general health conditions of the patients, and the accessibility of different methods to the doctors [8]. Given the high recurrence and metastatic rates in patients with advanced cancer, new therapeutic strategies must be developed to improve treatments. In some cases of oral cancers and tongue cancers, traditional treatments are difficult to implement, because the lesion is full of blood vessels, there is high risk of metastasis, and the patients may suffer from large surgical trauma and severe psychological stress. Consequently, exploring the molecular mechanisms at play is crucially important.
Transforming growth factor β (TGF-β), a cytokine, regulates many cellular processes, including cell growth, differentiation, development, and migration. Substantial evidence indicates that the TGF-β signaling pathway has two distinct roles: pro-tumorigenic and tumor-suppressive [9,10]. On the one hand, depending on cellular context, TGF-β may exert a tumor-promoting effect. In cells of late-stage cancer, its activation can promote tumorigenesis that affects cell motility, immune system evasion, metastasis, and neoangiogenesis. On the other hand, the TGF-β pathway exhibits tumor-suppressing functions in healthy cells as well as in early stage tumor cells; it inhibits epithelial cell-cycle progression and promotes apoptosis [9,10,11,12,13,14,15]. The dual functionalities and complex outcomes of TGF-β signaling render the therapies employing anti–TGF-β agents challenging; medication must be provided cautiously, and patient selection based on suitable indications is difficult.
Deregulation of TGF-β signaling contributes to oral cancer’s pathophysiology [16,17]. TGF-β upregulation has been detected in specimens collected from oral cancer patients with metastases from the mouth to bones [18]. TGF-β stimulates oral cancer cell migration [19,20], which implies TGF-β’s important role in cancer progression. Here, we review and summarize recent studies of TGF-β signaling regulation in oral cancers. The possible TGF-β signaling pathway therapeutic targets were addressed, with the discussion of current achievements and challenges. This review will provide insight into future applications of TGF-β-based medications.

2. Search Strategy

A comprehensive search of the literature was performed using the following search engines: PubMed, Web of Science, Embase, and Google Scholar. We focused on articles on oral squamous cell carcinomas, salivary adenoid cystic carcinomas, and keratocystic odontogenic tumors. The keywords used for searching were: ‘(TGF-β) AND (oral squamous cell carcinomas)’, ‘(TGF-β) AND (salivary adenoid cystic carcinomas)’, ‘(TGF-β) and (keratocystic odontogenic tumors)’, ‘(TGF-β) and (oral cancer therapy)’ and ‘(TGF-β) and (oral cancer)’. After the removal of the duplicates, the reviews, comments, non-English articles, and those without full text, there were 127 research papers left. The quality of these studies was evaluated, and studies with insufficient experimental evidence or not closely related to the scope of this review were excluded. Finally, a total of 59 studies were selected for this review (Figure 1).

3. TGF-β Signaling Pathways

The TGF-β superfamily of cytokines in humans consists of over 30 factors, including the TGF-βs (TGF-β1, -2, and -3); anti-Müllerian hormone (aka Müllerian inhibitory factor); activins; inhibins; nodal agents; myostatin; bone morphogenetic proteins (BMPs); and sundry differentiation and growth factors, which are found in all multicellular organisms [9]. TGF-β ligands are the primary TGF-β signaling mediators. Inactive TGF-βs are released as homodimeric polypeptides that interact with latency-associated peptide (LAP) and latent TGF-β-binding protein, forming a complex known as the large latent complex (LLC) that helps promote extracellular sequestration. LAP proteolysis and release of LLC from the extracellular matrix (ECM) requires TGF-β activation to free active TGF-β, which then binds to certain receptors [21]. Matrix metalloproteinase (MMP)-2 and MMP-9 work to cleave latent TGF-β and ECM protein. Thrombospondin 1 activates latent TGF-β [22]. Alternatively, via a conformational change, the αvβ6 integrin is capable of regulating TGF-β activity; it does so by binding LAP’s RGD motif, which in turn induces the latent complex to release mature TGF-β [23,24].
Members of the TGF-β superfamily bind to heteromeric receptor complexes containing type I (TβRI) and type II (TβRII) receptors with serine/threonine-kinase activities (Figure 2). Auxiliary coreceptors (also termed type III receptors, TβRIII) aid TGF-β superfamily members’ approaches to signaling receptors [25]. Activated TGF-β ligands interact with TβRII to form a heterotetrameric, an active receptor complex that recruits and phosphorylates TβRI (aka activin receptor-like kinase anaplastic lymphoma kinase [ALK] 5) at specific residues of serine and threonine. This complex, acting as a functional receptor, thus regulates downstream SMAD-dependent canonical pathway activation as well as SMAD-independent or noncanonical pathway activation [10,26].
The SMADs (mothers against decapentaplegic homolog) comprise proteins that are grouped together because they transduce extracellular signals directly to the nucleus. Mammalian cells contain eight different SMADs of three categories: (1) receptor-regulated SMADs (R-SMADs), which include SMAD1, SMAD2, SMAD3, SMAD5, and SMAD8/9; (2) the common mediator SMAD (Co-SMAD) SMAD4; and (3) inhibitory SMADs, including SMAD6 and SMAD7 (Figure 2). The major function of the inhibitory SMAD is to downregulate receptor-mediated R-SMAD phosphorylation. This results in the prevention of complex formation with Co-SMAD. In the SMAD-dependent pathway, phosphorylated TβRI recruits and phosphorylates R-SMADs (SMAD2 and SMAD3). SMAD2 and SMAD3, in association with SMAD4, then modulate transcription. After activation, the SMAD complex is translocated to the nucleus, where it binds specific DNA domains known as SMAD-binding elements. Upon binding, R-SMAD–Co-SMAD complexes interact with other DNA-binding transcription factors to transactivate or repress target genes. In addition to such canonical signaling, the activated TGF-β receptor complex transmits signals via other SMAD-independent pathways, including the mitogen-activated protein kinase (MAPK) pathway, the c-Jun N-terminal kinase (JNK)/p38 pathway, and the phosphatidylinositol-3 kinase (PI3K)/protein kinase B (AKT) pathway; it transmits signals by tumor necrosis factor (TNF) receptor-associated factor 4 (TRAF4), TRAF6, and Rho GTPases [26,27]. Several signaling pathways are implicated in TGF-β signaling, including Notch, Wnt, PI3K/AKT, interferon, TNF, and renin-angiotensin system (aka RAS) pathways [9,28].

4. Role of TGF-β in Oral Squamous Cell Carcinomas

Oral squamous cell carcinomas (OSCCs) occurring in the mucus membranes of the mouth and/or oral cavity account for >90% of all oral malignancies [29]. OSCCs can develop from oral precancerous lesions such as leucoplakia and erythroplakia [30]. In most patients, when OSCC is diagnosed, the disease is advanced (stages III and IV) [31]. In OSCC, the role of TGF-β signaling is complex because TGF-β evidences both anti- and protumorigenic activities. Early in carcinogenesis, TGF-β acts as a potent tumor suppressor. Yet, during advanced disease stages, TGF-β may promote cancer cell growth and/or metastasis growth, thus advancing tumor progression [32].
In early stage OSCC, TGF-β exhibits potent inhibitory growth effects; it promotes apoptosis, reconstitutes the tumor microenvironment, and suppresses cell cycle progression via G1 arrest [33]. The mechanisms of such TGF-β-mediated arrest differ depending on cell type and cell differentiation stage. However, in most squamous carcinoma cells, TGF-β suppresses cell-cycle proliferation via G1 arrest, including the inhibition of MYC and cyclin-dependent kinase (CDK) 4 (CDK4), and repression of cyclin activities and induction of the CDK inhibitors p21 (aka CIP1) and p15 (aka INK4B) [34]. TGF-β represses MYC transcription, which encodes a transcriptional activator of genes required for cell growth and proliferation. The SMAD2/SMAD3/SMAD4 complex acts together with Sp1 of the p15 promoter to induce p15 gene expression in response to TGF-β1 [34,35]. The p15 protein suppresses the kinase activities of the cyclin D–CDK4/6 complex and inhibits phosphorylation of intracellular target retinoblastoma protein, which is necessary for G1/S progression. TGF-β1-induced p21 plays a critical role in the inhibition of cyclin-CDK kinase activities, increasing p27 binding to cyclin E-CDK2 and inhibiting the expression of cyclin D1, CDK4, and CDK6 to mediate cell cycle arrest in G1 [36,37]. Wang et al. found that TGF-β1 rapidly increased p15 and p21 expression in both high- and low-metastatic OSCC cell lines. OSCC growth arrest might be thus induced by TGF-β1 via multiple pathways [38].
Studies on TGF-β-mediated tumor suppression revealed that loss of TGF-β signaling components is associated with carcinoma progression [10,39,40]. In OSCC patients, TβRII expression is significantly lower in metastases than in the primary tumors, where TβRII expression is downregulated compared to the normal oral epithelium [16]. Fukai et al. found that decreased TβRII expression correlates with high cell migration, high invasiveness, and poor prognosis of OSCC [36]. In OSCC patients, it is common that TβRII and TβRIII lack expression in oral epithelium and stroma; however, the TβRI expression levels in OSCC do not change significantly compared to normal tissue [16,41]. Mutation of genes encoding SMAD transcription factors (SMAD2, SMAD4) revokes TGF-β signaling’s tumor-suppressive effects and triggers OSCC progression [42].
During advanced carcinogenesis stages, TGF-β promotes OSCC invasion and metastasis, acting by many pathways, including suppression of immune surveillance, induction of the epithelial-mesenchymal transition (EMT), and modulation of the tumor microenvironment to promote carcinoma-cell proliferation. The EMT results in cell–cell adhesion loss, thus increasing cell mobility and promoting metastasis. During EMT, cells lose E-cadherin as well as other components of epithelial cell junctions. Metastasis is accelerated by the EMT, which is associated with increased cell migration and invasion, and increased cell-substrate adhesion. The overexpression of TGF-β ligands is common in OSCC as it promotes EMT and cell invasion in vitro [43,44]. Of all the inducers of EMT-activating transcription factors, including Slug, Snail, Twist, and Zeb1, TGF-β is among the best known [45]. Sun et al. found that TGF-β1 regulates MMP-9 expression to enhance the EMT of OSCC cells by inducing the expression of Snail/Ets-1 [43]. TGF-β1 induces MMP-9 expression via the SMAD/myosin light chain kinase pathway and upregulates Slug by acting on extracellular-regulated protein kinases (ERK) 1/2 to promote OSCC invasion and metastasis [46,47].
Immune surveillance is crucial in carcinogenesis. TGF-β and the cytokine interleukin (IL)-10 are typical anti-inflammatory immunosuppressive cytokines that promote immune system evasion by neoplastic cells [48,49]. In OSCC, TGF-β and IL-10 upregulation, combined with the downregulation of natural killer (NK) cell-activating cytokines such as IL-2 and NK receptors, modulate the immune escape of NK cells [50,51]. TGF-β downregulates IL-2 expression and thus suppresses T-cell proliferation, activation, and differentiation [52]. In a study of head and neck squamous cell carcinoma using an orthotopic murine model, Dasgupta et al. found that TGF-β1 reduces the expression of the NK-activating receptor NKG2D on the NK cell surface and inhibits NK cell functions [53]. TH17 cells produce IL-17 and participate in various inflammatory reactions. TH17 cell numbers and upregulated IL-17 levels correlate with the extent of OSCC malignancy. TGF-β plays a proinflammatory role via its effects on cells of the innate immune system as well as on TH17 cells [54,55].
In addition, TGF-β induces macrophages to differentiate into the M2 phenotype in the OSCC tumor microenvironment. A predominance of M2 macrophages triggers local immunosuppression and more lymph node involvement and reduces survival [56]. In tumor microenvironments, cancer-associated fibroblasts (CAFs) are central to immunosuppression. In an ex vivo model of OSCC, TGF-β, IL-10, and arginase-1 caused CAFs to induce CD14 myeloid cells to develop an M2-like phenotype, ultimately suppressing T-cell proliferation and further promoting tumorigenesis [57].

5. Role of TGF-β in Salivary Adenoid Cystic Carcinoma

One of the most common malignant tumors of the salivary glands is salivary adenoid cystic carcinoma (SACC), which is associated with perineural and perivascular invasion, distant metastasis, and poor prognosis. SACC exhibits certain specific characteristics, including multiple local recurrences, indolent growth, and high metastasis incidence [58,59,60]. Treatment failure, as in most solid tumors, is generally the result of distant metastasis at an early disease stage [61]. TGF-β both suppresses and promotes SACC activities. However, further systematic research on TGF-β signaling in SACC is needed.
TβRIII expression is markedly repressed in SACC patients. Xu et al. demonstrated that transient TβRIII overexpression induces SACC m apoptosis and G2/M arrest that inhibit cell growth and migration; TβRIII, scaffolding protein arrestin 2 (β-arrestin 2), and IκBα form a complex that is an important negative regulator of NF-κB signaling and promoting the TβRIII-mediated inhibition of SACC migration and progression [62]. A member of the TGF-β superfamily, Runt-related transcription factor 3 (RUNX3) acts as a tumor suppressor gene and induces apoptosis. After CpG island hypermethylation of the promoter region in various carcinomas such as SACC, RUNX3 expression is decreased or even abolished [63]. Poor prognosis, a tumor located in a salivary gland, and SACC recurrence all correlate highly with RUNX3 downregulation associated with DNA hypermethylation and protein mislocalization. RUNX3 mislocalization may be a novel mechanism of RUNX3 gene inactivation that impairs TGF-β signaling [63]. TGF-β-stimulated clone 22 (TSC-22) induces differentiation, thus negatively regulating salivary-gland cancer cell growth. In SACC patients, TSC-22 was hyper expressed in the inner ductal cells of tubular structures. Nuclear translocation of TSC-22 is essential for the induction of salivary-gland cancer cell apoptosis [64]. Dong et al. found that the upregulation of TGF-β1 and phospho-SMAD2, combined with the downregulation of membrane β-catenin, is associated with pulmonary metastasis in SACC patients; they also found that TGF-β1 promotes the invasion and migration of SACC cell lines via the SMAD pathway, inducing the EMT and changing the morphology of SACC cells [65]. The redox protein thioredoxin 1 is critical in terms of cellular redox homeostasis regulation and the promotion of antiapoptotic functions. One study found that TGF-β-induced EMT is mediated by thioredoxin 1, and the EMT further promotes SACC metastasis by stabilizing Slug and Snail, two transcription factors associated with PI3K/AKT/glycogen synthase kinase 3β signaling [66]. TRAF6 mediates SACC progression by activating SMAD2/SMAD3/p38/JNK/ERK signaling cascades [67].

6. Role of TGF-β in Keratocystic Odontogenic Tumors

Keratocystic odontogenic tumors (KCOTs) are benign odontogenic cystic epithelial neoplasms that comprise approximately 11% of all oral cysts [68]. KCOTs are single-cell or multicell, rapidly growing, and intraosseous tumors associated with nevoid basal cell carcinoma syndrome (NBCCS). Human KCOTs arise in two ways: sporadically, or as components of this syndrome. In contrast to other maxillary cysts, such as epidermoid and radicular cysts, KCOTs have high recurrence rates. They may transform malignantly and exhibit a characteristic lining of parakeratinized, stratified squamous epithelium [69,70,71,72].
Some human KCOTs obtain PTCH1 mutations that may also occur in tumors associated with NBCCS as well as in the sporadic forms of retinoblastoma, basal cell carcinoma, and medulloblastoma [73,74]. PTCH1 mutations may derepress Smoothened (Smo) and constitutive Sonic Hedgehog (Shh) signal activities [74]. When Shh binds Patched that is encoded by PTCH1, then the inhibition of Smo by Patched ceases. Upon the activation of the Hedgehog pathway, the downstream transcription factor Gli will enter the nucleus to initiate the transcription of downstream effector genes, including PTCH1 per se as well as Wnt and TGF-β/BMP family genes [75]. The aberrant inactivation of PTCH1 triggers constitutive activation of the Hedgehog pathway, causing a series of developmental abnormalities and tumorigenesis [74,76]. It is unknown whether other signal transduction events occur during KCOT initiation and progression. It is believed that tooth development is closely related to dental epithelial status. However, the pathology that bridges tooth root development to KCOT tumorigenesis remains under investigation.
Previous studies have suggested that TGF-β might play roles in various lesions, including odontogenic cysts, by regulating epithelial cell differentiation [77]. TGF-β may be involved in local invasiveness [78], biological behaviors [79], and the proliferation and/or differentiation of various odontogenic cysts and tumors. It has been suggested that TGF-β expression may differ among these lesions, thus affecting epithelial-mesenchymal interaction as well as extracellular matrix formation and growth [77,80,81]. The expression of TGF-β1 and its receptors correlated negatively with the differentiation of OSCC [82] and other cancers [83,84], but differentiation was further enhanced in terminally differentiated mucosal keratinocytes [85]. Santos et al. found that the level of immunoreactive TGF-β1 in connective tissue is higher in KCOTs than in other cysts [86].
TGF-β acts in all stages of tooth development by the regulation (paracrine and autocrine) of epithelial-mesenchymal interactions, regulation of odontoblast proliferation, maintenance of homeostasis of odontogenic epithelium, and facilitation of terminal differentiation of ameloblasts and odontoblasts [79,87,88,89]. Gao et al. suggested that SMAD4 serves as a tumor suppressor gene of the TGF-β/BMP pathway, plays important roles in epithelial–mesenchymal interactions that control root development, and suppresses multiple KCOTs in the jaw [90]. A reduction in SMAD4 expression changes the fate of odontoblasts, dramatically reactivates Hedgehog signaling within the hyperplastic epithelium, and alters the fates of the Hertwig epithelial root sheath and the epithelial rests of Malassez, thus triggering the formation of multiple jaw odontogenic cysts [81]. Yamada et al. reported that TGF-β and IL-1α in KCOT fluid act together to induce receptor activator of NF-κB ligand (aka RANKL) expression in stromal fibroblasts and to promote osteoclast formation that might mediate jawbone resorption [91]. Zhong et al. found that the presence of both Slug and MMP-9 in KCOT samples indicates that the EMT profoundly affects local invasion by KCOTs [92].

7. TGF-β as a Therapeutic Biomarker

The TGF-β signaling pathway plays both tumor-promoting and tumor-suppressing roles in cancer (Figure 3). TGF-β potently inhibits epithelial cell growth via the induction of various proteins that inhibit the cell cycle. Thus, during early oral carcinogenesis, TGF-β strongly suppresses tumors, maintaining tissue homeostasis and inhibiting tumor progression [93,94,95]. Patient stratification based on gene expression has revealed that the highest TGF-β pathway activity is associated with the worst prognosis. TGF-β is notably overexpressed in aggressive and invasive cancers; it triggers malignant effects in established cancers by induction of the EMT and metastasis [96]. Thus, TGF-β promotes tumor invasion, growth, and metastasis. The dual TGF-β cancer phenotypes are in part dependent on the disease progression stage (Figure 3).
TGF-β may be a potential cancer-drug target when TGF-β is overexpressed and thus promotes tumor growth. However, the usefulness of such drugs is compromised by the dual roles that TGF-β signaling plays in cancer. TGF-β promotes fibrosis, angiogenesis, and metastasis as well as suppressing host immune responses, thus enhancing the tumor microenvironment at all carcinogenetic stages. The TGF-β pathway is therefore a candidate for a microenvironment-targeted cancer treatment.
Aberrant TGF-β signaling contributes to the pathogenesis of many diseases. Several TGF-β pathway inhibitors have been preclinically investigated, with some currently under clinical evaluation. For such applications, the downregulation of TGF-β expression and the inhibition of TGF-β-receptor kinase activation is used to suppress TGF-β signaling [97]. Various methods have been used to antagonize TGF-β actions, such as the usage of antisense oligonucleotides that target specific TGF-β signaling components. These treatments are either provided intravenously or engineered into immune cells to prevent TGF-β synthesis [98,99]. Monoclonal antibodies of the anti–TGF-β receptor prevent ligand–receptor interactions [100], and small-molecule inhibitors of TGF-β receptor kinase prevent signal transduction. Engineered mutant TGF-β ligands [98], recombinant soluble β-glycan [101,102], decorin [103], and a soluble TβRII:Fc fusion protein [104,105,106] all inhibit TGF-β activity or signaling.
TGF-β inhibitors suppress tumor development and reduce fibrosis in tumor microenvironments. A dense fibrotic stroma is a characteristic of several tumors, including pancreatic ductal adenocarcinomas. TGF-β regulates the functions of stromal cells in fibrotic tissues and tumor microenvironments by driving tissue angiogenesis, inducing immune cell recruitment, and promoting fibroblast differentiation into myofibroblasts [107]. TGF-β mediates myofibroblast differentiation and activates resident/infiltrating fibroblasts. CAFs and stellate cells are activated by mitogenic and fibrogenic stimulants, including TGF-β, platelet-derived growth factor, and Shh released by cancer cells. CAFs and stellate cells, when activated, emit factors that promote tumor growth, invasion, and metastasis [108]. In the Panc-1 orthotopic pancreatic ductal adenocarcinoma model, TGF-βRI inhibition by SD-208 significantly reduced the biological activities of fibroblasts and suppressed tumor growth [109]. Medicherla et al. found that the blocking of TGF-β signaling with galunisertib or LY2109761 reduces tumor stromal content and inhibits the synthesis of connective tissue growth factor by downregulating fibroblastic-cell proliferation in hepatocellular carcinomas [110]. Pirfenidone (5-methyl-1-phenylpyridin-2-one, brand name Esbriet), an orally available pyridine derivative, was approved in 2014 by the US Food and Drug Administration to treat idiopathic pulmonary fibrosis (Table 1). Although the precise biological target of pirfenidone remains unclear, pirfenidone inhibits TGF-β production, thereby reducing collagen synthesis and fibroblast proliferation and delaying idiopathic pulmonary fibrosis development [111]. Fluorofenidone, which is a structural analog of pirfenidone, also inhibits TGF-β expression. Fluorofenidone suppresses the TGF-β-induced or -associated EMT and attenuates myofibroblast differentiation [112,113]. Malignant transformation to OSCC develops in approximately 7% of patients with oral submucous fibrosis. In such patients, small-molecule TGF-β inhibitors that may treat oral cancers are an alternative to surgery.
The inhibition of TGF-β signaling is an emerging cancer therapy strategy. Most small-molecule inhibitors of such signaling specifically target the type I receptor of TGF-β to inhibit the phosphorylation of SMAD2 and SMAD3 with the maintenance of at least some noncanonical responses. TβRI kinase inhibitors affect the immune response, invasion/metastasis, and extracellular matrix production. SB-431542 inhibited the kinase activities of TβRI, ALK4, and ALK7; inhibited the phosphorylation of SMAD2/SMAD3; and reduced cell proliferation and migration [114,115,116]. TEW-7197 (vactosertib) is a competitive inhibitor of ATP that inhibits kinase activity by binding the hydrophobic ATP pocket of the TβRI intracellular domain. In a mouse model, TEW-7197 effectively inhibited melanoma development and lymph-node metastasis. In addition to blocking the phosphorylation of SMAD2/SMAD3, TEW-7197 enhanced the antimelanoma cytotoxic T-lymphocyte immune response by increasing the proliferation of CD8+ T lymphocytes in mice with melanoma by ubiquitin-mediated SMAD4 degradation [117]. SMAD2 phosphorylation induced by TGF-β1 was downregulated by LY2157299 in human HLE and HLF hepatocellular carcinoma cells. Additionally, LY2157299 significantly inhibited cancer-cell metastasis and proliferation [118].
Despite the accumulated evidence of TGF-β-targeting therapeutics, it should also be noticed that inhibitors of TGF-β receptor kinases sometimes have poor pharmacokinetics and pharmacodynamics. They target TGF-β receptors non-specifically, and inhibit related type I receptors for several other TGF-β-related proteins equally effectively [115], and may also inhibit other kinases, such as p38 MAPK [119]. The lack of specificity and poor pharmacokinetics of current TGF-β receptor kinase inhibitors lead to potential challenges with dosing strategies. Due to the aforementioned issues of the TGF-β-pathway-targeted therapeutics, it is critical to identify reliable biomarkers that predict response to TGF-β inhibitors. Predictive capacities may differ depending on the tumor microenvironment and overall blood biomarker spectrum of individual patients [120]. Zinc finger E-box-binding homeobox 1 (ZEB1) is a critical activator of the EMT associated with cancer. The direct targeting of ZEB1 by miR-101 induced EMT, and the downregulation of miR-101 promoted OSCC growth and metastasis [121]. MiR-455-5p suppresses UBE2B and thus contributes to oral tumorigenesis by binding specific promoter regions of SMAD3 [122]. αvβ6 regulates TGF-β activity by binding the RGD sequence of LAP, triggering cancer progression [123,124]. The blockade of αvβ6 did not affect tumor cell proliferation in vitro but inhibited tumor growth in vivo [125]. The microenvironment thus plays a role in such regulation; further work in this realm is necessary.

8. Discussion and Conclusions

In the tumor microenvironment, TGF-β is becoming an important and clinically actionable means of immune evasion [126]. Meanwhile, TGF-β signaling inhibition is also an emerging strategy for cancer therapy. There are three potential strategies: (1) inhibiting TGF-β signaling components or modulating factors; (2) blocking the interaction between TGF-β and other signaling pathways in cancer; and (3) normalizing tumor microenvironment homeostasis by downregulating stromal stimulation resulting from excess TGF-β produced by tumors and tumor-related tissues.
TGF-β-pathway inhibitors have been investigated in the preclinical setting, some of which are now in the clinical development phase [127]. However, unlike preclinical results, TGF-β blocking progress in clinical trials has always been difficult, and many trials have failed to replicate success in animal models. Due to the dual role of the TGF-β signaling pathway, the window of effective TGF-β targeting is therefore evidently small, which poses a clear challenge in selecting patients at the right time. The ideal evaluation method will enable the identification of individuals who will benefit from TGF-targeted therapy, as well as excluding patients in whom TGF-targeting will create limited or even detrimental effects. Alternatively, another potential reason for the relative lack of success in clinical trials is that immune-suppressive TGF-β signaling may be more nuanced than previously realized, and additional factors must be considered when designing anti-TGF-β therapies. Since the effects of TGF-β signaling are both localized and rapid [15], it is important to choose the dose and frequency of administration to support the sustained inhibition of the TGF-β receptors and prevent the reactivation of TGF-β signals in the tumor microenvironment. In addition, the identification of reliable and predictable biomarkers of response to TGF-β inhibitors is also a critical issue.
TGF-β signaling plays a key role in cancer progression and high TGF-β signaling activity has been linked to resistance to multiple anticancer therapies, including chemotherapy and molecularly targeted therapies [127]. Enhanced intratumoral TGF-β signaling is also a barrier in patients in response to immunotherapy. Given the role of TGF-β signaling in non-pathophysiological processes, it is not surprising that systemic inhibition of TGF-β signaling would accompany the onset of adverse events. However, current early clinical trials of TGF-β inhibitors show that adverse events are generally controllable, such that therapies respond significantly and persistently when combined with immune checkpoint inhibitors. In the future, the inhibition of TGF-β with dual-specific drugs or using drug interference with the composition of TGF-β signaling with restricted tissue expression are expected to lead to new therapies. Furthermore, combining TGF-β inhibitors with cytotoxic drugs, radiotherapy, adoptive T cell transfer therapy (such as CAR-T), immune checkpoint inhibitors (such as PD-1/PD-L1 antibodies), cancer vaccines, viral vectors, nanoparticles, or oncolytic viruses may provide additional opportunities.

9. Future Implications

Our understanding of oral cancer has been greatly improved by the developments in molecular biotechnology and human genetics. TGF-β signaling plays a complex role in oral cancer progression and development, as it can serve as a tumor promoter or suppressor, depending on the progression stage of oral cancer. However, much remains to be investigated in the future, particularly the cross-talk between the TGF-β pathway and other signaling pathways. Further elucidation of regulatory tumor-host interactions may identify more useful therapeutic targets and aid in the accurate timing of TGF-β-targeted therapy and clinical development of new TGF-β inhibitors.
TGF-β and other components of the TGF-β signaling pathway are also therapeutic antitumor candidates. Future work on the TGF-β signaling mechanisms that operate in oral cancer may reveal new therapeutic targets, such as small-molecule TGF-β inhibitors, that broaden the range of drug development and even prevent or cure oral cancer.

Author Contributions

Conceptualization, literature search, formal analysis and writing-original draft preparation, Y.G.; writing-review and editing, Y.G., Y.C. and F.C.; visualization, Y.G. and T.X.; supervision, Y.C. and F.C.; funding acquisition, Y.C. and F.C. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Guangdong Young Innovative Talents Project (2022KQNCX067); Innovation and Entrepreneurship Training Program for College Students (202210590017X); Shenzhen Overseas Talent Program (827-000511); The National Natural Science Foundation of China (No. 81991501) and National Key R&D Program of China (SQ2022YFE010688).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We are grateful to Huisheng Zhang for his kind support and constant encouragement.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ramu, A.K.; Ali, D.; Alarifi, S.; Abuthakir, M.H.S.; Abdul, B.A.A. Reserpine inhibits DNA repair, cell proliferation, invasion and induces apoptosis in oral carcinogenesis via modulation of TGF-β signaling. Life Sci. 2021, 264, 118730. [Google Scholar] [CrossRef] [PubMed]
  2. Rivera, C. Essentials of oral cancer. Int. J. Clin. Exp. Pathol. 2015, 8, 11884–11894. [Google Scholar] [PubMed]
  3. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2016. CA Cancer J. Clin. 2016, 66, 7–30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. van Dijk, B.A.; Brands, M.T.; Geurts, S.M.; Merkx, M.A.; Roodenburg, J.L. Trends in oral cavity cancer incidence, mortality, survival and treatment in the Netherlands. Int. J. Cancer 2016, 139, 574–583. [Google Scholar] [CrossRef] [Green Version]
  5. Derbi, H.A.; Kruger, E.; Tennant, M. Incidence of oral cancer in Western Australia (1982–2009): Trends and regional variations. Asia-Pac. J. Clin. Oncol. 2016, 12, e305–e310. [Google Scholar] [CrossRef]
  6. Zhang, J.; Gao, F.; Yang, A.K.; Chen, W.K.; Chen, S.W.; Li, H.; Zhang, X.; Yang, Z.Y.; Chen, X.L.; Song, M. Epidemiologic characteristics of oral cancer: Single-center analysis of 4097 patients from the Sun Yat-sen University Cancer Center. Chin. J. Cancer 2016, 35, 24. [Google Scholar] [CrossRef] [Green Version]
  7. Huang, C.; Song, H.; Lai, L. The role and mechanism of microRNA-18a-5p in oral squamous cell carcinoma. Mol. Med. Rep. 2019, 20, 1637–1644. [Google Scholar] [CrossRef] [Green Version]
  8. Zhu, X.; Cao, Y.; Liu, W.; Ju, X.; Zhao, X.; Jiang, L.; Ye, Y.; Jin, G.; Zhang, H. Stereotactic body radiotherapy plus pembrolizumab and trametinib versus stereotactic body radiotherapy plus gemcitabine for locally recurrent pancreatic cancer after surgical resection: An open-label, randomised, controlled, phase 2 trial. Lancet Oncol. 2022, 23, e105–e115. [Google Scholar] [CrossRef]
  9. Massagué, J. TGFβ in cancer. Cell 2008, 134, 215–230. [Google Scholar] [CrossRef] [Green Version]
  10. Ikushima, H.; Miyazono, K. TGF-beta signalling: A complex web in cancer progression. Nat. Rev. Cancer 2010, 10, 415–424. [Google Scholar] [CrossRef]
  11. Pickup, M.; Novitskiy, S.; Moses, H.L. The roles of TGF-β in the tumour microenvironment. Nat. Rev. Cancer 2013, 13, 788–799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Derynck, R.; Akhurst, R.J.; Balmain, A. TGF-beta signaling in tumor suppression and cancer progression. Nat. Genet. 2001, 29, 117–129. [Google Scholar] [CrossRef]
  13. Calon, A.; Espinet, E.; Palomo-Ponce, S.; Tauriello, D.V.; Iglesias, M.; Céspedes, M.V.; Sevillano, M.; Nadal, C.; Jung, P.; Zhang, X.H.; et al. Dependency of colorectal cancer on a TGF-β-driven program in stromal cells for metastasis initiation. Cancer Cell 2012, 22, 571–584. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Principe, D.R.; Doll, J.A.; Bauer, J.; Jung, B.; Munshi, H.G.; Bartholin, L.; Pasche, B.; Lee, C.; Grippo, P.J. TGF-β: Duality of function between tumor prevention and carcinogenesis. J. Natl. Cancer Inst. 2014, 106, djt369. [Google Scholar] [CrossRef]
  15. Giannos, P.; Kechagias, K.S.; Gal, A. Identification of prognostic gene biomarkers in non-small cell lung cancer progression by integrated bioinformatics analysis. Biology 2021, 10, 1200. [Google Scholar] [CrossRef]
  16. Paterson, I.C.; Matthews, J.B.; Huntley, S.; Robinson, C.M.; Fahey, M.; Parkinson, E.K.; Prime, S.S. Decreased expression of TGF-β cell surface receptors during progression of human oral squamous cell carcinoma. J. Pathol. 2001, 193, 458–467. [Google Scholar] [CrossRef]
  17. Prime, S.S.; Davies, M.; Pring, M.; Paterson, I.C. The role of TGF-beta in epithelial malignancy and its relevance to the pathogenesis of oral cancer (part II). Crit. Rev. Oral Biol. Med. 2004, 15, 337–347. [Google Scholar] [CrossRef]
  18. Takahashi, K.; Akatsu, Y.; Podyma-Inoue, K.A.; Matsumoto, T.; Watabe, T. Targeting all transforming growth factor-β isoforms with a Fc chimeric receptor impairs tumor growth and angiogenesis of oral squamous cell cancer. J. Biol. Chem. 2020, 295, 12559–12572. [Google Scholar] [CrossRef]
  19. Nakamura, R.; Kayamori, K.; Oue, E.; Sakamoto, K.; Harada, K.; Yamaguchi, A. Transforming growth factor-β synthesized by stromal cells and cancer cells participates in bone resorption induced by oral squamous cell carcinoma. Biochem. Biophys. Res. Commun. 2015, 458, 777–782. [Google Scholar] [CrossRef] [PubMed]
  20. Takahashi, K.; Podyma-Inoue, K.A.; Takao, C.; Yoshimatsu, Y.; Muramatsu, T.; Inazawa, J.; Watabe, T. Regulatory role of transforming growth factor-β signals in the migration and tumor formation of HOC313-LM cells, an oral squamous cell carcinoma. J. Stomatol. Soc. Jpn. 2018, 85, 52–61. [Google Scholar]
  21. Derynck, R.; Miyazono, K. The TGF-β Family; Cold Spring Harbor Lab Press: Cold Spring Harbor, NY, USA, 2008. [Google Scholar]
  22. Schultz-Cherry, S.; Ribeiro, S.; Gentry, L.; Murphy-Ullrich, J.E. Thrombospondin binds and activates the small and large forms of latent transforming growth factor-beta in a chemically defined system. J. Biol. Chem. 1994, 269, 26775–26782. [Google Scholar] [CrossRef] [PubMed]
  23. Annes, J.P.; Rifkin, D.B.; Munger, J.S. The integrin alphavbeta6 binds and activates latent TGFbeta3. FEBS Lett. 2002, 511, 65–68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Shi, M.; Zhu, J.; Wang, R.; Chen, X.; Mi, L.; Walz, T.; Springer, T.A. Latent TGF-β structure and activation. Nature 2011, 474, 343–349. [Google Scholar] [CrossRef] [Green Version]
  25. Li, M.O.; Wan, Y.Y.; Sanjabi, S.; Robertson, A.; Flavell, R.A. Transforming growth factor-beta regulation of immune responses. Annu. Rev. Immunol. 2006, 24, 99–146. [Google Scholar] [CrossRef]
  26. Colak, S.; Ten Dijke, P. Targeting TGF-β signaling in cancer. Trends Cancer 2017, 3, 56–71. [Google Scholar] [CrossRef]
  27. 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] [Green Version]
  28. Akhurst, R.J.; Hata, A. Targeting the TGFbeta signalling pathway in disease. Nat. Rev. Drug Discov. 2012, 11, 790–811. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Warnakulasuriya, S. Global epidemiology of oral and oropharyngeal cancer. Oral Oncol. 2009, 45, 309–316. [Google Scholar] [CrossRef]
  30. Chen, Y.J.; Lin, S.C.; Kao, T.; Chang, C.S.; Hong, P.S.; Shieh, T.M.; Chang, K.W. Genome-wide profiling of oral squamous cell carcinoma. J. Pathol. 2004, 204, 326–332. [Google Scholar] [CrossRef] [PubMed]
  31. Oliveira, M.L.C.; Wagner, V.P.; Sant’Ana Filho, M.; Carrard, V.C.; Hugo, F.N.; Martins, M.D. A 10-year analysis of the oral squamous cell carcinoma profile in patients from public health centers in Uruguay. Braz. Oral Res. 2015, 29, 1–8. [Google Scholar] [CrossRef]
  32. Jimi, E.; Furuta, H.; Matsuo, K.; Tominaga, K.; Takahashi, T.; Nakanishi, O. The cellular and molecular mechanisms of bone invasion by oral squamous cell carcinoma. Oral Dis. 2011, 17, 462–468. [Google Scholar] [CrossRef] [PubMed]
  33. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646. [Google Scholar] [CrossRef] [Green Version]
  34. Feng, X.H.; Liang, Y.Y.; Liang, M.; Zhai, W.; Lin, X. Direct interaction of c-Myc with Smad2 and Smad3 to inhibit TGF-β–mediated induction of the CDK inhibitor p15(Ink4B). Mol. Cell 2016, 63, 1089. [Google Scholar] [CrossRef] [PubMed]
  35. Feng, X.H.; Lin, X.; Derynck, R. Smad2, Smad3 and Smad4 cooperate with Sp1 to induce p15(Ink4B) transcription in response to TGF-beta. EMBO J. 2000, 19, 5178–5193. [Google Scholar] [CrossRef] [Green Version]
  36. Malliri, A.; Yeudall, W.A.; Nikolic, M.; Crouch, D.H.; Parkinson, E.K.; Ozanne, B. Sensitivity to transforming growth factor beta 1–induced growth arrest is common in human squamous cell carcinoma cell lines: C-MYC down-regulation and p21waf1 induction are important early events. Cell Growth Differ. 1996, 7, 1291–1304. [Google Scholar]
  37. Peng, B.; Fleming, J.B.; Breslin, T.; Grau, A.M.; Fojioka, S.; Abbruzzese, J.L.; Evans, D.B.; Ayers, D.; Wathen, K.; Wu, T.; et al. Suppression of tumorigenesis and induction of p15(ink4b) by Smad4/DPC4 in human pancreatic cancer cells. Clin. Cancer Res. 2002, 8, 3628–3638. [Google Scholar] [PubMed]
  38. Wang, X.; Sun, W.; Bai, J.; Ma, L.; Yu, Y.; Geng, J.; Qi, J.; Shi, Z.; Fu, S. Growth inhibition induced by transforming growth factor-β1 in human oral squamous cell carcinoma. Mol. Biol. Rep. 2009, 36, 861. [Google Scholar] [CrossRef] [PubMed]
  39. Böttinger, E.P.; Jakubczak, J.L.; Roberts, I.S.; Mumy, M.; Hemmati, P.; Bagnall, K.; Merlino, G.; Wakefield, L.M. Expression of a dominant-negative mutant TGF-β type II receptor in transgenic mice reveals essential roles for TGF-β in regulation of growth and differentiation in the exocrine pancreas. EMBO J. 1997, 16, 2621–2633. [Google Scholar] [CrossRef] [Green Version]
  40. Amendt, C.; Schirmacher, P.; Weber, H.; Blessing, M. Expression of a dominant negative type II TGF-β receptor in mouse skin results in an increase in carcinoma incidence and an acceleration of carcinoma development. Oncogene 1998, 17, 25–34. [Google Scholar] [CrossRef] [Green Version]
  41. Fukai, Y.; Fukuchi, M.; Masuda, N.; Osawa, H.; Kuwano, H. Reduced expression of transforming growth factor-beta receptors is an unfavorable prognostic factor in human esophageal squamous cell carcinoma. Int. J. Cancer 2003, 104, 161–166. [Google Scholar] [CrossRef]
  42. Qiu, W.; Schönleben, F.; Li, X.; Su, G.H. Disruption of transforming growth factor beta–Smad signaling pathway in head and neck squamous cell carcinoma as evidenced by mutations of SMAD2 and SMAD4. Cancer Lett. 2007, 245, 163–170. [Google Scholar] [CrossRef] [Green Version]
  43. Sun, L.; Diamond, M.E.; Ottaviano, A.J.; Joseph, M.J.; Ananthanarayan, V.; Munshi, H.G. Transforming growth factor-1 promotes matrix metalloproteinase-9–mediated oral cancer invasion through snail expression. Mol. Cancer Res. 2008, 6, 10–20. [Google Scholar] [CrossRef] [Green Version]
  44. Qiao, B.; Johnson, N.W.; Gao, J. Epithelial–mesenchymal transition in oral squamous cell carcinoma triggered by transforming growth factor-β1 is Snail family–dependent and correlates with matrix metalloproteinase-2 and -9 expressions. Int. J. Oncol. 2010, 37, 663–668. [Google Scholar]
  45. Padua, D.; Massagué, J. Roles of TGFβ in metastasis. Cell Res. 2009, 19, 89–102. [Google Scholar] [CrossRef] [PubMed]
  46. Sinpitaksakul, S.N.; Pimkhaokham, A.; Sanchavanakit, N.; Pavasant, P. TGF-beta1 induced MMP-9 expression in HNSCC cell lines via Smad/MLCK pathway. Biochem. Biophys. Res. Commun. 2008, 371, 713–718. [Google Scholar] [CrossRef] [PubMed]
  47. Joseph, M.J.; Dangi-Garimella, S.; Shields, M.A.; Diamond, M.E.; Sun, L.; Koblinski, J.E.; Munshi, H.G. Slug is a downstream mediator of transforming growth factor-β1–induced matrix metalloproteinase-9 expression and invasion of oral cancer cells. J. Cell. Biochem. 2009, 108, 726–736. [Google Scholar] [CrossRef]
  48. Arantes, D.A.C.; Costa, N.L.; Mendonça, E.F.; Silva, T.A.; Batista, A.C. Overexpression of immunosuppressive cytokines is associated with poorer clinical stage of oral squamous cell carcinoma. Arch. Oral Biol. 2016, 61, 28–35. [Google Scholar] [CrossRef] [PubMed]
  49. Thomas, D.A.; Massagué, J. TGF-beta directly targets cytotoxic T cell functions during tumor evasion of immune surveillance. Cancer Cell 2005, 8, 369–380. [Google Scholar] [CrossRef] [Green Version]
  50. Gaur, P.; Singh, A.K.; Shukla, N.K.; Das, S.N. Inter-relation of Th1, Th2, Th17 and Treg cytokines in oral cancer patients and their clinical significance. Hum. Immunol. 2014, 75, 330–337. [Google Scholar] [CrossRef]
  51. Dutta, A.; Banerjee, A.; Saikia, N.; Phookan, J.; Baruah, M.N.; Baruah, S. Negative regulation of natural killer cell in tumor tissue and peripheral blood of oral squamous cell carcinoma. Cytokine 2015, 76, 123–130. [Google Scholar] [CrossRef]
  52. Sun, Y.; Liu, N.; Guan, X.; Wu, H.; Sun, Z.; Zeng, H. Immunosuppression induced by chronic inflammation and the progression to oral squamous cell carcinoma. Mediators Inflamm. 2016, 2016, 5715719. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Dasgupta, S.; Bhattacharya-Chatterjee, M.; O’Malley, B.W., Jr.; Chatterjee, S.K. Inhibition of NK cell activity through TGF-beta 1 by down-regulation of NKG2D in a murine model of head and neck cancer. J. Immunol. 2005, 175, 5541–5550. [Google Scholar] [CrossRef] [Green Version]
  54. Lee, J.J.; Chang, Y.L.; Lai, W.L.; Ko, J.Y.; Kuo, M.Y.; Chiang, C.P.; Azuma, M.; Chen, C.W.; Chia, J.S. Increased prevalence of interleukin-17–producing CD4+ tumor infiltrating lymphocytes in human oral squamous cell carcinoma. Head Neck 2011, 33, 1301–1308. [Google Scholar] [CrossRef]
  55. Martinez, G.J.; Zhang, Z.; Reynolds, J.M.; Tanaka, S.; Chen, D. Smad2 positively regulates the generation of Th17 cells. J. Biol. Chem. 2010, 285, 29039–29043. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Costa, N.L.; Valadares, M.C.; Souza, P.P.C.; Mendonca, E.F.; Oliveira, J.C.; Silva, T.A.; Batista, A.C. Tumor-associated macrophages and the profile of inflammatory cytokines in oral squamous cell carcinoma. Oral Oncol. 2013, 49, 216–223. [Google Scholar] [CrossRef]
  57. Takahashi, H.; Sakakura, K.; Kudo, T.; Toyoda, M.; Kaira, K.; Oyama, T.; Chikamatsu, K. Cancer-associated fibroblasts promote an immunosuppressive microenvironment through the induction and accumulation of protumoral macrophages. Oncotarget 2017, 8, 8633. [Google Scholar] [CrossRef] [Green Version]
  58. Spiro, R.H. Salivary neoplasms: Overview of a 35-year experience with 2807 patients. Head Neck Surg. 1986, 8, 177–184. [Google Scholar] [CrossRef]
  59. Ciccolallo, L.; Licitra, L.; Cantú, G.; Gatta, G.; EUROCARE Working Group. Survival from salivary glands adenoid cystic carcinoma in European populations. Oral Oncol. 2009, 45, 669–674. [Google Scholar] [CrossRef] [PubMed]
  60. Tian, Z.; Li, L.; Wang, L.; Hu, Y.; Li, J. Salivary gland neoplasms in oral and maxillofacial regions: A 23-year retrospective study of 6982 cases in an eastern Chinese population. Int. J. Oral Maxillofac. Surg. 2010, 39, 235–242. [Google Scholar] [CrossRef]
  61. Liu, J.; Shao, C.; Tan, M.L.; Mu, D.; Ferris, R.L.; Ha, P.K. Molecular biology of adenoid cystic carcinoma. Head Neck 2012, 34, 1665–1677. [Google Scholar] [CrossRef]
  62. Xu, D.; Li, D.; Lu, Z.; Dong, X.; Wang, X. Type III TGF-β receptor inhibits cell proliferation and migration in salivary glands adenoid cystic carcinoma by suppressing NF-κB signaling. Oncol. Rep. 2016, 35, 267–274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Sasahira, T.; Kurihara, M.; Yamamoto, K.; Bhawal, U.K.; Kirita, T.; Kuniyasu, H. Downregulation of runt-related transcription factor 3 associated with poor prognosis of adenoid cystic and mucoepidermoid carcinomas of the salivary gland. Cancer Sci. 2011, 102, 492–497. [Google Scholar] [CrossRef]
  64. Doi, Y.; Kawamata, H.; Ono, Y.; Fujimori, T.; Imai, Y. Expression and cellular localization of TSC-22 in normal salivary glands and salivary gland tumors: Implications for tumor cell differentiation. Oncol. Rep. 2008, 19, 609–616. [Google Scholar] [CrossRef] [PubMed]
  65. Dong, L.; Ge, X.Y.; Wang, Y.X.; Yang, L.Q.; Li, S.L.; Yu, G.Y.; Gao, Y.; Fu, J. Transforming growth factor-β and epithelial–mesenchymal transition are associated with pulmonary metastasis in adenoid cystic carcinoma. Oral Oncol. 2013, 49, 1051–1058. [Google Scholar] [CrossRef]
  66. Jiang, Y.; Feng, X.; Zheng, L.; Li, S.L.; Ge, X.Y.; Zhang, J.G. Thioredoxin 1 mediates TGF-β–induced epithelial–mesenchymal transition in salivary adenoid cystic carcinoma. Oncotarget 2015, 6, 25506. [Google Scholar] [CrossRef] [Green Version]
  67. Liang, Y.; Jiao, J.; Liang, L.; Zhang, J.; Lu, Y.; Xie, H.; Liang, Q.; Wan, D.; Duan, L.; Wu, Y.; et al. Tumor necrosis factor receptor–associated factor 6 mediated the promotion of salivary adenoid cystic carcinoma progression through SMAD-p38-JNK signaling pathway induced by TGF-β. J. Oral Pathol. Med. 2018, 47, 583–589. [Google Scholar] [CrossRef] [PubMed]
  68. Thompson, L. World Health Organization classification of tumours: Pathology and genetics of head and neck tumours (editorial). Ear Nose Throat J. 2006, 85, 74. [Google Scholar] [CrossRef] [Green Version]
  69. Madras, J.; Lapointe, H. Keratocystic odontogenic tumour: Reclassification of the odontogenic keratocyst from cyst to tumour. J. Can. Dent. Assoc. 2008, 74, 165. [Google Scholar] [PubMed]
  70. Shear, M. The aggressive nature of the odontogenic keratocyst: Is it a benign cystic neoplasm? Part 1. Clinical and early experimental evidence of aggressive behaviour. Oral Oncol. 2002, 38, 219–226. [Google Scholar] [CrossRef]
  71. Shear, M. The aggressive nature of the odontogenic keratocyst: Is it a benign cystic neoplasm? Part 2. Proliferation and genetic studies. Oral Oncol. 2002, 38, 323–331. [Google Scholar] [CrossRef]
  72. Shear, M. The aggressive nature of the odontogenic keratocyst: Is it a benign cystic neoplasm? Part 3. Immunocytochemistry of cytokeratin and other epithelial cell markers. Oral Oncol. 2002, 38, 407–415. [Google Scholar] [CrossRef] [PubMed]
  73. Hahn, H.; Wicking, C.; Zaphiropoulous, P.G.; Gailani, M.R.; Bale, A.E. Mutations of the human homolog of Drosophila patched in the nevoid basal cell carcinoma syndrome. Cell 1996, 85, 841–851. [Google Scholar] [CrossRef] [Green Version]
  74. Barreto, D.; Gomez, R.; Bale, A.; Boson, W.; De Marco, L. Ptch gene mutations in odontogenic keratocysts. J. Dent. Res. 2000, 79, 1418–1422. [Google Scholar] [CrossRef] [PubMed]
  75. Grachtchouk, M.; Liu, J.; Wang, A.; Wei, L.; Bichakjian, C.K.; Garlick, J.; Paulino, A.F.; Giordano, T.; Dlugosz, A.A. Odontogenic keratocysts arise from quiescent epithelial rests and are associated with deregulated hedgehog signaling in mice and humans. Am. J. Pathol. 2006, 169, 806–814. [Google Scholar] [CrossRef] [Green Version]
  76. Rahnama, F.; Toftgård, R.; Zaphiropoulos, P.G. Distinct roles of ptch2 splice variants in hedgehog signalling. Biochem. J. 2004, 378, 325–334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Piattelli, A.; Rubini, C.; Fioroni, M.; Favero, L.; Strocchi, R. Expression of transforming growth factor-beta 1 (TGF-beta 1) in odontogenic cysts. Int. Endod. J. 2004, 37, 7–11. [Google Scholar] [CrossRef]
  78. Kurioka, K.; Wato, M.; Iseki, T.; Tanaka, A.; Morita, S. Differential expression of the epithelial mesenchymal transition factors Snail, Slug, Twist, TGF-β, and E-cadherin in ameloblastoma. Med. Mol. Morphol. 2017, 50, 68–75. [Google Scholar] [CrossRef]
  79. Kumamoto, H.; Yoshida, M.; Ooya, K. Immunohistochemical detection of hepatocyte growth factor, transforming growth factor-beta and their receptors in epithelial odontogenic tumors. J. Oral Pathol. Med. 2002, 31, 539–548. [Google Scholar] [CrossRef]
  80. Iezzi, G.; Piattelli, A.; Rubini, C.; Artese, L.; Goteri, G.; Perrotti, V.; Carinci, F. Expression of transforming growth factor beta1 in ameloblastomas. J. Craniofac. Surg. 2008, 19, 1618–1621. [Google Scholar] [CrossRef]
  81. Karathanasi, V.; Tosios, K.I.; Nikitakis, N.G.; Piperi, E.; Koutlas, I.; Trimis, G.; Sklavounou, A. TGF-β1, Smad-2/-3, Smad-1/-5/-8, and Smad-4 signaling factors are expressed in ameloblastomas, adenomatoid odontogenic tumors, and calcifying cystic odontogenic tumors: An immunohistochemical study. J. Oral Pathol. Med. 2013, 42, 415–423. [Google Scholar] [CrossRef]
  82. Meng, W.; Xia, Q.; Wu, L.; Chen, S.; He, X.; Zhang, L.; Gao, Q.; Zhou, H. Downregulation of TGF-beta receptor types II and III in oral squamous cell carcinoma and oral carcinoma–associated fibroblasts. BMC Cancer 2011, 11, 88. [Google Scholar] [CrossRef] [Green Version]
  83. Mhawech-Fauceglia, P.; Kesterson, J.; Wang, D.; Akers, S.; DuPont, N.C.; Clark, K.; Lele, S.; Liu, S. Expression and clinical significance of the transforming growth factor-beta signalling pathway in endometrial cancer. Histopathology 2011, 59, 63–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Iancu, I.V.; Botezatu, A.; Goia-Ruşanu, C.D.; Stanescu, A.; Huica, I.; Nistor, E.; Anton, G.; Plesa, A. TGF-beta signalling pathway factors in HPV-induced cervical lesions. Roum. Arch. Microbiol. Immunol. 2010, 69, 113–118. [Google Scholar]
  85. Min, B.M.; Woo, K.M.; Lee, G.; Park, N.H. Terminal differentiation of normal human oral keratinocytes is associated with enhanced cellular TGF-β and phospholipase c-γ1 levels and apoptotic cell death. Exp. Cell Res. 1999, 249, 377–385. [Google Scholar] [CrossRef]
  86. Santos, P.P.A.; Nonaka, C.F.W.; Freitas, R.A.; Pereira Pinto, L.; Souza, L.B. Immunohistochemical analysis of myofibroblasts, TGF-β1, and IFN-γ in epithelial odontogenic lesions. J. Oral Pathol. Med. 2017, 46, 365–370. [Google Scholar] [CrossRef]
  87. Yoshida, M.; Kumamoto, H.; Ooya, K.; Mayanagi, H. Immunohistochemical analysis of benign mixed and mesenchymal odontogenic tumors. Oral Med. Pathol. 2003, 8, 125–132. [Google Scholar] [CrossRef] [Green Version]
  88. Takata, T.; Miyauchi, M.; Ogawa, I.; Kudo, Y.; Takekoshi, T.; Zhao, M.; Sato, S.; Nikai, H.; Komiyama, K. Immunoexpression of transforming growth factor beta in desmoplastic ameloblastoma. Virchows Arch. 2000, 436, 319–323. [Google Scholar] [CrossRef]
  89. Heikinheimo, K.; Happonen, R.; Miettinen, P.; Ritvos, O. Transforming growth factor beta 2 in epithelial differentiation of developing teeth and odontogenic tumors. J. Clin. Investig. 1993, 91, 1019–1027. [Google Scholar] [CrossRef] [PubMed]
  90. Gao, Y.; Yang, G.; Weng, T.; Du, J.; Wang, X.; Zhou, J.; Wang, S.; Yang, X. Disruption of Smad4 in odontoblasts causes multiple keratocystic odontogenic tumors and tooth malformation in mice. Mol. Cell. Biol. 2009, 29, 5941–5951. [Google Scholar] [CrossRef] [Green Version]
  91. Yamada, C.; Aikawa, T.; Okuno, E.; Miyagawa, K.; Amano, K.; Takahata, S.; Kimata, M.; Okura, M.; Iida, S.; Kogo, M. TGF-β in jaw tumor fluids induces RANKL expression in stromal fibroblasts. Oral Oncol. 2016, 49, 499–508. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Zhong, W.Q.; Chen, G.; Zhang, W.; Ren, J.G.; Wu, Z.X.; Zhao, Y.; Liu, B.; Zhao, Y.F. Epithelial–mesenchymal transition in keratocystic odontogenic tumor: Possible role in locally aggressive behavior. BioMed Res. Int. 2015, 2015, 168089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Hu, M.G.; Hu, G.F.; Yong, K.; Tsuji, T.; Wong, D. Role of p12(cdk2-ap1) in transforming growth factor-beta1–mediated growth suppression. Cancer Res. 2004, 64, 490–499. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Game, S.; Stone, A.; Scully, C.; Prime, S. Tumour progression in experimental oral carcinogenesis is associated with changes in EGF and TGF-β receptor expression and altered responses to these growth factors. Carcinogenesis 1990, 11, 965–973. [Google Scholar] [CrossRef]
  95. Paterson, I.C.; Davies, M.; Stone, A.; Huntley, S.; Smith, E.; Pring, M.; Eveson, J.W.; Robinson, C.M.; Parkinson, E.K.; Prime, S.S. TGF-β1 acts as a tumor suppressor of human malignant keratinocytes independently of Smad 4 expression and ligand-induced G1 arrest. Oncogene 2002, 21, 1616–1624. [Google Scholar] [CrossRef] [Green Version]
  96. Dasgupta, S.; Bhattacharya-Chatterjee, M.; O’Malley, B.W., Jr.; Chatterjee, S.K. Tumor metastasis in an orthotopic murine model of head and neck cancer: Possible role of TGF-beta 1 secreted by the tumor cells. J. Cell. Biochem. 2006, 97, 1036–1051. [Google Scholar] [CrossRef]
  97. Neuzillet, C.; Tijeras-Raballand, A.; Cohen, R.; Cros, J.; Faivre, S.; Raymond, E.; de Gramont, A. Targeting the TGFβ pathway for cancer therapy. Pharmacol. Ther. 2015, 147, 22–31. [Google Scholar] [CrossRef] [Green Version]
  98. Hinck, A.P. Structure-guided engineering of TGF-βs for the development of novel inhibitors and probing mechanism. Bioorg. Med. Chem. 2018, 26, 5239–5246. [Google Scholar] [CrossRef] [PubMed]
  99. Seystahl, K.; Papachristodoulou, A.; Burghardt, I.; Schneider, H.; Hasenbach, K.; Janicot, M.; Roth, P.; Weller, M. Biological role and therapeutic targeting of TGF-β3 in glioblastoma. Mol. Cancer Ther. 2017, 16, 1177–1186. [Google Scholar] [CrossRef] [Green Version]
  100. Giri, S.N.; Hyde, D.M.; Hollinger, M.A. Effect of antibody to transforming growth factor beta on bleomycin induced accumulation of lung collagen in mice. Thorax 1993, 48, 959–966. [Google Scholar] [CrossRef] [Green Version]
  101. Gronau, T.; Krüger, K.; Prein, C.; Aszodi, A.; Gronau, I.; Iozzo, R.V.; Mooren, F.C.; Clausen-Schaumann, H.; Bertrand, J.; Pap, T.; et al. Forced exercise–induced osteoarthritis is attenuated in mice lacking the small leucine-rich proteoglycan decorin. Ann. Rheum. Dis. 2017, 76, 442–449. [Google Scholar] [CrossRef]
  102. Bandyopadhyay, A.; López-Casillas, F.; Malik, S.N.; Montiel, J.L.; Mendoza, V.; Yang, J.; Sun, L.Z. Antitumor activity of a recombinant soluble betaglycan in human breast cancer xenograft. Cancer Res. 2002, 62, 4690–4695. [Google Scholar] [PubMed]
  103. Yamaguchi, Y.; Mann, D.M.; Ruoslahti, E. Negative regulation of transforming growth factor-β by the proteoglycan decorin. Nature 1990, 346, 281–284. [Google Scholar] [CrossRef]
  104. Villarreal, M.M.; Kim, S.K.; Barron, L.; Kodali, R.; Baardsnes, J.; Hinck, C.S.; Krzysiak, T.C.; Henen, M.A.; Pakhomova, O.; Mendoza, V.; et al. Binding properties of the transforming growth factor-β coreceptor betaglycan: Proposed mechanism for potentiation of receptor complex assembly and signaling. Biochemistry 2016, 55, 6880–6896. [Google Scholar] [CrossRef]
  105. Isaka, Y.; Akagi, Y.; Ando, Y.; Tsujie, M.; Sudo, T.; Ohno, N.; Border, W.A.; Noble, N.A.; Kaneda, Y.; Hori, M.; et al. Gene therapy by transforming growth factor-beta receptor–IgG Fc chimera suppressed extracellular matrix accumulation in experimental glomerulonephritis. Kidney Int. 1999, 55, 465–475. [Google Scholar] [CrossRef] [Green Version]
  106. Zheng, H.; Wang, J.; Koteliansky, V.E.; Gotwals, P.J.; Hauer-Jensen, M. Recombinant soluble transforming growth factor β type II receptor ameliorates radiation enteropathy in mice. Gastroenterology 2000, 119, 1286–1296. [Google Scholar] [CrossRef]
  107. Ueno, H.; Sakamoto, T.; Nakamura, T.; Qi, Z.; Astuchi, N.; Takeshita, A.; Shimizu, K.; Ohashi, H. A soluble transforming growth factor beta receptor expressed in muscle prevents liver fibrogenesis and dysfunction in rats. Hum. Gene Ther. 2000, 11, 33–42. [Google Scholar] [CrossRef] [PubMed]
  108. Tirado-Rodriguez, B.; Ortega, E.; Segura-Medina, P.; Huerta-Yepez, S. TGF-β: An important mediator of allergic disease and a molecule with dual activity in cancer development. J. Immunol. Res. 2014, 2014, 318481. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Duner, S.; Lopatko Lindman, J.; Ansari, D.; Gundewar, C.; Andersson, R. Pancreatic cancer: The role of pancreatic stellate cells in tumor progression. Pancreatology 2010, 10, 673–681. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Medicherla, S.; Li, L.; Ma, J.Y.; Kapoun, A.M.; Higgins, L.S. Antitumor activity of TGF-beta inhibitor is dependent on the microenvironment. Anticancer Res. 2007, 27, 4149–4157. [Google Scholar] [PubMed]
  111. 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]
  112. Taniguchi, H.; Ebina, M.; Kondoh, Y.; Ogura, T.; Azuma, A.; Suga, M.; Taguchi, Y.; Takahashi, H.; Nakata, K.; Sato, A.; et al. Pirfenidone in idiopathic pulmonary fibrosis. Eur. Respir. J. 2010, 35, 821–829. [Google Scholar] [CrossRef]
  113. Peng, Y.; Li, L.; Zhang, X.; Xie, M.; Yang, C.; Tu, S.; Shen, H.; Hu, G.; Tao, L.; Yang, H. Fluorofenidone affects hepatic stellate cell activation in hepatic fibrosis by targeting the TGF-β1/Smad and MAPK signaling pathways. Exp. Ther. Med. 2019, 18, 41–48. [Google Scholar] [CrossRef] [Green Version]
  114. Yuan, Q.; Wang, L.; Zhang, F.; Wang, R.; Fu, X.; Peng, Z.; Ning, W.; Hu, G.; Wang, Z.; Tao, L. Fluorofenidone suppresses epithelial–mesenchymal transition and the expression of connective tissue growth factor via inhibiting TGF-β/Smads signaling in human proximal tubular epithelial cells. Pharmazie 2011, 66, 961–967. [Google Scholar]
  115. Inman, G.J.; Nicolás, F.J.; Callahan, J.F.; Harling, J.D.; Gaster, L.M.; Reith, A.D.; Laping, N.J.; Hill, C.S. SB-431542 is a potent and specific inhibitor of transforming growth factor-beta superfamily type I activin receptor-like kinase (ALK) receptors ALK4, ALK5, and ALK7. Mol. Pharmacol. 2002, 62, 65–74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Laping, N.; Grygielko, E.; Mathur, A.; Butter, S.; Bomberger, J.; Tweed, C.; Martin, W.; Fornwald, J.; Lehr, R.; Harling, J.; et al. Inhibition of transforming growth factor (TGF)-β1–induced extracellular matrix with a novel inhibitor of the TGF-β type I receptor kinase activity: SB-431542. Mol. Pharmacol. 2002, 62, 58–64. [Google Scholar] [CrossRef] [Green Version]
  117. Tanaka, H.; Shinto, O.; Yashiro, M.; Yamazoe, S.; Iwauchi, T.; Muguruma, K.; Kubo, N.; Ohira, M.; Hirakawa, K. Transforming growth factor β signaling inhibitor, SB-431542, induces maturation of dendritic cells and enhances anti-tumor activity. Oncol. Rep. 2010, 24, 1637–1643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Yoon, J.H.; Jung, S.M.; Park, S.H.; Kato, M.; Yamashita, T.; Lee, I.K.; Sudo, K.; Nakae, S.; Han, J.S.; Kim, O.H.; et al. Activin receptor-like kinase5 inhibition suppresses mouse melanoma by ubiquitin degradation of Smad4, thereby derepressing eomesodermin in cytotoxic T lymphocytes. EMBO Mol. Med. 2013, 5, 1720–1739. [Google Scholar] [CrossRef] [PubMed]
  119. Yingling, J.M.; McMillen, W.T.; Yan, L.; Huang, H.; Sawyer, J.S.; Graff, J.; Clawson, D.K.; Britt, K.S.; Anderson, B.D.; Beight, D.W.; et al. Preclinical assessment of galunisertib (LY2157299 monohydrate), a first-in-class transforming growth factor-β receptor type I inhibitor. Oncotarget 2018, 9, 6659–6677. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Dituri, F.; Mazzocca, A.; Fernando, J.; Peidrò, F.J.; Papappicco, P.; Fabregat, I.; De Santis, F.; Paradiso, A.; Sabbà, C.; Giannelli, G. Differential inhibition of the TGF-β signaling pathway in HCC cells using the small molecule inhibitor LY2157299 and the D10 monoclonal antibody against TGF-β receptor type II. PLoS ONE 2013, 8, e67109, Erratum in PLoS ONE 2013, 8, e67109. [Google Scholar] [CrossRef]
  121. Necchi, A.; Giannatempo, P.; Mariani, L.; Farè, E.; Raggi, D.; Pennati, M.; Zaffaroni, N.; Crippa, F.; Marchianò, A.; Nicolai, N.; et al. PF-03446962, a fully-human monoclonal antibody against transforming growth-factor β (TGFβ) receptor ALK1, in pre-treated patients with urothelial cancer: An open label, single-group, phase 2 trial. Investig. New Drugs 2014, 32, 555–560. [Google Scholar] [CrossRef] [PubMed]
  122. Wu, B.; Lei, D.; Wang, L.; Yang, X.; Jia, S.; Yang, Z.; Shan, C.; Yang, X.; Zhang, C.; Lu, B. MiRNA-101 inhibits oral squamous-cell carcinoma growth and metastasis by targeting zinc finger E-box binding homeobox 1. Am. J. Cancer Res. 2016, 6, 1396–1407. [Google Scholar] [PubMed]
  123. Cheng, C.M.; Shiah, S.G.; Huang, C.C.; Hsiao, J.R.; Chang, J.Y. Up-regulation of miR-455-5p by the TGF-β–SMAD signalling axis promotes the proliferation of oral squamous cancer cells by targeting UBE2B. J. Pathol. 2016, 240, 38–49. [Google Scholar] [CrossRef] [PubMed]
  124. Munger, J.S.; Huang, X.; Kawakatsu, H.; Griffiths, M.J.; Dalton, S.L.; Wu, J.; Pittet, J.F.; Kaminski, N.; Garat, C.; Matthay, M.A.; et al. The integrin alpha v beta 6 binds and activates latent TGF beta 1: A mechanism for regulating pulmonary inflammation and fibrosis. Cell 1999, 96, 319–328. [Google Scholar] [CrossRef] [Green Version]
  125. Koopman Van Aarsen, L.A.; Leone, D.R.; Ho, S.; Dolinski, B.M.; McCoon, P.E.; LePage, D.J.; Kelly, R.; Heaney, G.; Rayhorn, P.; Reid, C.; et al. Antibody-mediated blockade of integrin αvβ6 inhibits tumor progression in vivo by a transforming growth factor-β–regulated mechanism. Cancer Res. 2008, 68, 561–570. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Bierie, B.; Moses, H.L. Tumour microenvironment: TGF-beta: The molecular Jekyll and Hyde of cancer. Nat. Rev. Cancer 2006, 6, 506–520. [Google Scholar] [CrossRef] [PubMed]
  127. Derynck, R.; Turley, S.J.; Akhurst, R.J. TGF-β biology in cancer progression and immunotherapy. Nat. Rev. Clin. Oncol. 2021, 18, 9–34. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Flowchart of the search strategy and the literature selection process.
Figure 1. Flowchart of the search strategy and the literature selection process.
Ijms 24 10263 g001
Figure 2. Overview of TGF-β signaling pathway. TGF-β signaling is transduced through 2 pathways: canonical (SMAD dependent) and noncanonical (SMAD independent). Signal transduction initiates by binding TGF-β to TβRII, which recruits and transphosphorylates TβRI. Either canonical or noncanonical signaling pathways can subsequently be activated by the TβRI–TβRII complex. After phosphorylation, TβRI becomes activated, then activates the canonical pathway through phosphorylation of R-SMADs (SMAD2 and SMAD3). R-SMADs form heteromeric complexes with Co-SMAD (SMAD4) and translocate to the nucleus, where they bind to site-specific recognition sequences (SBEs) and regulate the expression of TGF-β-signaling-pathway target genes. R-SMADs modulate the biogenesis of microRNA (miRNA) by promoting the processing of primary miRNA into precursor miRNA in the nucleus. mG, 5′ capping of messenger RNA (mRNA); AAAAA, 3′ polyadenylation of mRNA. In noncanonical pathways, TGF-β signaling activates SMAD-independent pathways such as the renin–angiotensin system (RAS)/ERK, PI3K/AKT, JNK and p38, NF-κB, and Rho. TGF-β signaling can be influenced by other pathways (e.g., RAS, Wnt, Hedgehog, interferon, TNF, Notch).
Figure 2. Overview of TGF-β signaling pathway. TGF-β signaling is transduced through 2 pathways: canonical (SMAD dependent) and noncanonical (SMAD independent). Signal transduction initiates by binding TGF-β to TβRII, which recruits and transphosphorylates TβRI. Either canonical or noncanonical signaling pathways can subsequently be activated by the TβRI–TβRII complex. After phosphorylation, TβRI becomes activated, then activates the canonical pathway through phosphorylation of R-SMADs (SMAD2 and SMAD3). R-SMADs form heteromeric complexes with Co-SMAD (SMAD4) and translocate to the nucleus, where they bind to site-specific recognition sequences (SBEs) and regulate the expression of TGF-β-signaling-pathway target genes. R-SMADs modulate the biogenesis of microRNA (miRNA) by promoting the processing of primary miRNA into precursor miRNA in the nucleus. mG, 5′ capping of messenger RNA (mRNA); AAAAA, 3′ polyadenylation of mRNA. In noncanonical pathways, TGF-β signaling activates SMAD-independent pathways such as the renin–angiotensin system (RAS)/ERK, PI3K/AKT, JNK and p38, NF-κB, and Rho. TGF-β signaling can be influenced by other pathways (e.g., RAS, Wnt, Hedgehog, interferon, TNF, Notch).
Ijms 24 10263 g002
Figure 3. Relationship between genes related to TGF-β signaling pathway and pathogenesis of oral cancers. The diagram outlines the relationship between genes related to the TGF-β signaling pathway and the pathogenesis of oral cancers, including OSCC, salivary adenoid systemic carcinoma, and KCOTs. Genes indicated in red might promote disease progression; genes indicated in blue might suppress disease progression.
Figure 3. Relationship between genes related to TGF-β signaling pathway and pathogenesis of oral cancers. The diagram outlines the relationship between genes related to the TGF-β signaling pathway and the pathogenesis of oral cancers, including OSCC, salivary adenoid systemic carcinoma, and KCOTs. Genes indicated in red might promote disease progression; genes indicated in blue might suppress disease progression.
Ijms 24 10263 g003
Table 1. Overview of clinical trials for TGF-β targeting agents.
Table 1. Overview of clinical trials for TGF-β targeting agents.
DrugTypeTargetsDisease ApplicationsClinical Trial IdentifierPhase
Trabedersen (AP12009)Antisense oligoTGF-β2 ligandMelanoma, pancreatic cancer, colorectal carcinomaNCT00844064Phase I
GlioblastomaNCT00431561Phase I/IIB
GlioblastomaNCT00761280Phase III
Belagen-pumatucel-L (Lucanix)Antisense
gene-modified
allogeneic
tumor cell
vaccine
TGF-β2Non-small cell lung cancerNCT00676507Phase III/IV
Lung neoplasm, bronchogenic carcinomaNCT01058785Phase II
Fresolimumab (GC-1008)Humanized antibodyTGF-β1, TGF-β2, TGF-β3Renal cell carcinomaNCT00923169Phase I
Renal cell carcinoma, melanomaNCT00356460Phase I
Primary brain tumorsNCT01472731Phase II
Metastatic breast cancerNCT01401062Phase II
Non-small cell lung carcinomaNCT02581787Phase I/II
Pleural malignant mesotheliomaNCT01112293Phase II
Galunisertib (LY2157299)Small moleculeTβRI kinaseAdvanced solid tumors, pancreatic cancerNCT01373164Phase I/II
Hepatocellular carcinomaNCT02240433Phase I
Advanced solid tumorsNCT01722825Phase I
Malignant gliomaNCT01220271Phase I
Hepatocellular carcinomaNCT01246986Phase II
Advanced hepatocellular carcinomaNCT02178358Phase II
Pancreatic neoplasmsNCT02154646Phase I
Metastatic pancreatic cancerNCT02734160Phase I
Metastatic androgen receptor negative (AR-) triple negative breast cancerNCT02672475Phase I
Recurrent malignant gliomaNCT01682187Phase I
Advanced solid tumors, non-small cell lung carcinoma, hepatocellular carcinomaNCT02423343Phase I/II
Advanced solid tumorsNCT02304419Phase I
PirfenidoneSmall molecule, not TGF-β specific IPF, glomerulosclerosis and diabetic kidney disease, pathological skin scarringMultiple trialsPhase III
TEW-7197Small moleculeTβRI kinaseAdvanced solid tumorsNCT02160106Phase I
Myelodysplastic syndromesNCT03074006Phase I
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

Guo, Y.; Xu, T.; Chai, Y.; Chen, F. TGF-β Signaling in Progression of Oral Cancer. Int. J. Mol. Sci. 2023, 24, 10263. https://doi.org/10.3390/ijms241210263

AMA Style

Guo Y, Xu T, Chai Y, Chen F. TGF-β Signaling in Progression of Oral Cancer. International Journal of Molecular Sciences. 2023; 24(12):10263. https://doi.org/10.3390/ijms241210263

Chicago/Turabian Style

Guo, Yuanyuan, Tiansong Xu, Yujuan Chai, and Feng Chen. 2023. "TGF-β Signaling in Progression of Oral Cancer" International Journal of Molecular Sciences 24, no. 12: 10263. https://doi.org/10.3390/ijms241210263

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