TY - JOUR AU - Xin-Hua, Feng, AB - Abstract Signals from the transforming growth factor-β (TGF-β) superfamily mediate a broad spectrum of cellular processes and are deregulated in many diseases, including cancer. TGF-β signaling has dual roles in tumorigenesis. In the early phase of tumorigenesis, TGF-β has tumor suppressive functions, primarily through cell cycle arrest and apoptosis. However, in the late stage of cancer, TGF-β acts as a driver of tumor progression and metastasis by increasing tumor cell invasiveness and migration and promoting chemo-resistance. Here, we briefly review the mechanisms and functions of TGF-β signaling during tumor progression and discuss the therapeutic potentials of targeting the TGF-β pathway in cancer. TGF-β, cancer, EMT, immune, cancer treatment Introduction The transforming growth factor-β (TGF-β) family secreted factors are comprised of over 30 members in mammals, including activins, nodals, bone morphogenetic proteins (BMPs), and growth and differentiation factors (GDFs). They control numerous aspects of animal biology including embryonic development, organogenesis, cell fate decisions, immune modulation, stress responses, and stem cell function [1–3]. Because of its widespread functional diversity, malfunctions of TGF-β signaling are associated with human diseases including cancer, fibrosis, systemic sclerosis, and hereditary disorders [4–7]. TGF-β often inhibits cell proliferation and stimulates differentiation in normal cells, therefore acting as a tumor suppressor [1,2]. In contrast, in advanced cancer, it promotes tumor progression and metastasis, thus serving as an oncogenic factor [8,9]. Researchers have made tremendous effort in elucidating mechanisms underlying these contradictory phenomena. Recent studies have uncovered complex interactions at various levels by using combinatorial approaches from molecular and cellular to animal models and human samples. This review will summarize the mechanisms of TGF-β signaling from tumor suppression to tumor promotion and discuss the implication of this process for understanding cancer biology and therapies. TGF-β Superfamily Signaling Pathway On the cell surface, the TGF-β ligand first binds to the high affinity receptors and stabilizes the formation of a hetero-tetrameric complex of type I and type II serine/threonine kinase receptors [1,2]. For example, TGF-β first binds to the TGF-β type II receptor (i.e. TβRII) and induces the formation of the hetero-tetrameric complex of TβRII and the TGF-β type I receptor (i.e. TβRI). In the receptor complex, TβRII acts as an upstream kinase to phosphorylate TβRI in the serine-rich GS motif, leading to the activation of TβRI (Fig. 1). The human genome encodes seven type I receptors (i.e. ALK1–7) and five type II receptors (i.e. TβRII, ActRII, ActRIIB, BMPRII, and AMHRII) [1,2]. A TGF-β ligand often acts through specific combinations of the type I and type II receptors (Fig. 2). The ligand-mediated interactions between the ligand and the receptors as well as between the type I and type II receptors dictate the specificity of receptor signaling. Figure 1. View largeDownload slide The TGF-β signaling pathway TGF-β is presented to TβRII, which leads to the formation of a hetero-tetrameric complex between the serine/threonine kinases TβRI and TβRII. The constitutive active TβRII phosphorylates TβRI, which in turn recruits, phosphorylates and activates Smad2 and Smad3. Activated Smad2 and/or Smad3 bind(s) to Smad4 to form a heterotrimer. The Smad complex becomes accumulated in the nucleus and drives transcription in cooperation with large number of specific transcription factors or cofactors in a context-dependent manner. In addition to the canonical Smad-mediated signaling, TGF-β also activates several other signaling cascades such as TRAF6–TAK1–p38/JNK, RhoA–Rhock1, and Par6. Figure 1. View largeDownload slide The TGF-β signaling pathway TGF-β is presented to TβRII, which leads to the formation of a hetero-tetrameric complex between the serine/threonine kinases TβRI and TβRII. The constitutive active TβRII phosphorylates TβRI, which in turn recruits, phosphorylates and activates Smad2 and Smad3. Activated Smad2 and/or Smad3 bind(s) to Smad4 to form a heterotrimer. The Smad complex becomes accumulated in the nucleus and drives transcription in cooperation with large number of specific transcription factors or cofactors in a context-dependent manner. In addition to the canonical Smad-mediated signaling, TGF-β also activates several other signaling cascades such as TRAF6–TAK1–p38/JNK, RhoA–Rhock1, and Par6. Figure 2. View largeDownload slide Schematic illustration of the selective binding of members of TGF-β superfamily receptors TGF-β ligands bind to specific combination of TβRII-TβRI heterotetramers at the surface. Subsequent activation of R-Smads is shown at right. While ActRII and ActRIIB are encoded by different genes, BMPRII and BMPRIIB are two isoforms encoded by the same gene. Figure 2. View largeDownload slide Schematic illustration of the selective binding of members of TGF-β superfamily receptors TGF-β ligands bind to specific combination of TβRII-TβRI heterotetramers at the surface. Subsequent activation of R-Smads is shown at right. While ActRII and ActRIIB are encoded by different genes, BMPRII and BMPRIIB are two isoforms encoded by the same gene. Smad proteins are the major intracellular mediators for TGF-β superfamily signaling [10]. They are classified into three groups. The first group is the receptor-activated Smads (R-Smads). Once activated by the type II receptor, the type I receptor phosphorylates R-Smads. For instance, Smad1, 5, and 8 are primarily activated by the BMP-specific type I receptors, while Smad2 and Smad3 are activated by the TGF-β/activin-specific type I receptors (Fig. 2). This specificity of type I receptor-mediated R-Smad activation is determined by the paired interactions of the L45 loop on the type I receptor and the L3 loop of the R-Smads as well as the GS motif of the receptor and the basic patch on the R-Smads. The second group is the common mediator Smad (Co-Smad, Smad4). Activated R-Smads form a complex with Smad4 and become accumulated the nucleus [10,11]. As the activated Smad complexes have a weak binding affinity for DNA, additional transcription factors are required for the high affinity interaction and specificity on chromatin [2,10,12]. A variety of transcription factor families have been reported to act in concert with Smad proteins such as p300/CBP, Sp1, AP1, Ets, and Fox proteins in transcription [10]. The third Smad group includes inhibitory Smads (I-Smads, e.g. Smad6 and Smad7). The inhibitory Smads are often induced by the TGF-β/BMP family signaling and act in a negative feedback loop [1,13]. While Smad6 mainly inhibits BMP signaling, Smad7 is able to prevent both BMP and TGF-β signaling. TGF-β has also been shown to signal independently of Smads, as it can activate signaling from other signaling molecules or pathways such as TAK1 (TGF-β-associated kinase 1), Erk (extracellular signal regulated kinase), p38, MAPK (mitogen-activated protein kinase), and Akt [14,15]. TGF-β Cytostatic Signaling in Normal Cells and Early Stages of Cancer TGF-β plays a crucial role in cancer, acting as a tumor suppressor during the initial stages of tumorigenesis. In normal epithelial cells, TGF-β can inhibit cell proliferation, while it also promotes cell differentiation and apoptosis. TGF-β inhibits cell proliferation mainly through two transcriptional events, i.e. induction of cyclin-dependent kinase (CDK) inhibitors and suppression of c-Myc expression. In epithelial, endothelial and neuronal cells, TGF-β induces expression of the CDK inhibitors such as p15INK4b and p21WAF1 [16–21]. It has been well established that cooperative actions of the Smad complex with transcription factor Sp1 activate transcription of p15INK4b and p21WAF1 in response to TGF-β [16–21]. p15INK4b inhibits cell cycle progression at the late G1 phase by preventing the formation of cyclin D complexes with CDK4 or CDK6 [22]. p21CIP1 inhibits the formation of cyclin E complexes with CDK2 [22]. In haematopoietic cells, TGF-β mediates its growth inhibition through up-regulation of the CDK inhibitor p57 [23]. Simultaneously, TGF-β down-regulates expression of the c-Myc oncogene which promotes cell proliferation. c-Myc is a major transcriptional factor of cell growth and division. In epithelial cells, downregulation of c-Myc expression is mediated by the TGF-β-induced Smad complex in cooperation with the transcription factors p107, E2F4/E2F5 [24], and CCAAT/enhancer binding protein β (C/EBPβ) [25,26]. Interestingly, C/EBPβ is required for both the repression of c-Myc expression and activation of p15INK4b expression [27]. In proliferating cells, c-Myc can also directly bind to Smad2/3 and blocks the transcriptional activities of Smad2/3 in transactivating transcription of p15INK4b and p21WAF1, which also explains why c-Myc is repressed by TGF-β [28]. In addition, the transcription factor c-Myc-interacting Zn finger protein-1 (Miz-1) recruits c-Myc as a repressor to the transcriptional start regions of the p15INK4b and p21WAF1 promoters [29,30]. Smad-independent pathways of TGF-β have been also implicated in the anti-proliferative response [14]. For example, TGF-β dephosphorylates p70S6K by PP2A, which leads to cell cycle arrest [31]. In addition to the regulatory role of TGF-β in cell cycle progression, TGF-β can also trigger apoptosis in a variety of cell types under physiological circumstances. However, the molecular mechanisms remain less defined. It has been reported that TGF-β increases expression of death-associated protein kinase DAPK in hepatoma cells [32] and expression of the signaling factor GADD45β (growth arrest and DNA damage 45β) in hepatocytes [33]. It also promotes the activation of death receptor FAS and binding of the pro-apoptotic effector Bim to Bcl-2 and Bcl-XL in gastric carcinoma cell lines [34]. In hepatocytes and B lymphocytes, TGF-β triggers apoptosis through increasing expression of phosphatase MKP2, which enhances the pro-apoptotic effect of Bim [35]. It is generally thought that TGF-β-mediated expression of these apoptotic regulators is mediated or coordinated by the canonical Smad signaling. In addition, TGF-β-induced apoptosis also involves the TRAF6–TAK1–JNK/p38 pathway in some cell types [36,37]. The TβRII-TβRI complex leads to auto-ubiquitylation of TRAF6, and active TRAF6 subsequently activates TAK1 by polyubiquitylation. Activated TAK1 phosphorylates MKK3 or MKK6, which in turn activates p38 MAPK and then leads to apoptosis [38,39]. TGF-β can be also linked to the tumor suppressor p53 [40], which in turn is regulated by p38 MAPK and Smads. In hepatocytes, TGF-β induces cell death through reactive oxygen species (ROS) production [41]. TGF-β-induced ROS production can promote apoptosis through the modulation of various members of the Bcl-2 family [42]. TGF-β may also induce cellular differentiation in some tumors. TGF-β regulates differentiation by controlling the expression of Id1 protein, which has been reported to enhance Ras-driven mammary tumorigenesis in mice bypassing senescence [43]. In an xenograft model using a Ras-transformed human breast epithelial cell line, TGF-β can down-regulate Id1, thereby suppressing tumor formation by Ras signaling and imposing a less proliferative phenotype [44]. These findings suggest that Id1 repression mediates cell differentiation as a tumor suppressive response to TGF-β. Due to its potent tumor suppressive effects, TGF-β signaling is lost in cancers, which is a hallmark in cancer [45]. Mutations or deletions of genes encoding components of the pathway frequently occur in a variety of cancers such as colon, pancreatic, and gastric cancers. Taking Smad4 as an example, its mutations or deletions are found in about half of pancreatic cancer patients [8,9]. However, such somatic mutations are rare in other cancers such as breast, prostate, and skin cancers [46]. Because loss of TGF-β responses is common in all types of cancers, there are alternative mechanisms underlying TGF-β resistance in those cancer types without somatic mutations in the TGF-β pathway. Much effort has been made to understand the tumor suppressive functions of TGF-β in those cancers without genetic inactivation of Smads or TGF-β receptors. Accumulating evidence has demonstrated that the tumor suppressor functions of Smads are compromised by oncogene products such as c-Ski, Bcl-6, c-Myc, Evi-1, and Stat3 through direct Smad–oncoprotein interactions [47–53], suggesting that activation of oncogenes can suppress TGF-β growth inhibitory response. Since different sets of oncogenes were found in different cancer types, it is anticipated that more interactions between oncogenes and the TGF-β pathway will be identified. Such studies will provide more insights into the loss of TGF-β tumor suppressive signaling in a particular cancer. TGF-β Tumor-promoting Signaling in Advanced Cancer In many cancers, there are mutations or deletions of TGF-β receptors or Smads in the TGF-β pathway, which lead to inactivation or perturbation of signaling responses [1,8]. Tumors can exploit certain aspects of TGF-β signaling to actively promote tumor cell. Different types of tumors such as gliomas, breast cancer, and prostate cancer seem to obtain mutations preferentially not in the core components of TGF-β signaling [1,8]. Such human tumors retain the ability to exploit TGF-β signaling to induce signaling that promotes EMT, tumor invasion, metastatic dissemination, and evasion of the immune system [1,8]. Under these conditions, tumors with such a signature are highly aggressive. EMT is crucial for the embryonic development. It is also important during wound healing, fibrosis, and cancer progression. Cells undergoing EMT are characterized by loss of E-cadherin expression and epithelial cell junctions, and a rearrangement of the cytoskeleton into a mesenchymal pattern. TGF-β is a key driver of EMT in epithelial cancers [1,8]. TGF-β-induced EMT supports tumor invasion and dissemination by releasing tumor cells into the surrounding environment and promoting their motility. In many cancers, TGF-β-induces EMT transcriptionally regulates E-cadherin, Snail, N-cadherin, and vimentin [54,55]. It also induces the expression of Sox4 and promotes mesenchymal programs, tumor progression and invasiveness in breast cancer [56]. More importantly, TGF-β-induced Snail or Twist1 can in turn drive epigenetic changes that influence EMT [54,55]. TGF-β increases cell motility, dissemination, and metastasis by regulating gene expression of integrin in both lung and breast cancer [57–60]. In HCC, TGF-β-driven EMT promotes cell dissemination and intrahepatic metastasis by inducing Snail1, which confers ability of resistance of apoptosis [61]. Additionally, TGF-β promotes CXCR4 expression in HCC cells, driving cell migration and invasion [62]. In prostate cancer, TGF-β increases the metastasis by repressing the expression of E-cadherin and promoting the expression of N-cadherin, ZEB1, TWIST, fibronectin, and Snail1 [63–65]. During the final stage of metastasis, mesenchymal-to-epithelial transition (MET), which is the reverse process of EMT, is also required to colonize distant sites and form metastases [66]. TGF-β has also been reported to regulate Id1 expression which promotes metastatic colonization in breast cancer cells [67] and repress Twist expression in basal breast cancer cells which infiltrate the lung parenchyma [68]. Although it is well established that TGF-β mediates EMT before cell dissemination from the primary tumor, it is unknown when or where MET occurs and to what extent TGF-β contributes in this process. Collectively, EMT driven by TGF-β confers invasiveness, motility, and progenitor-like features in cancer cell. In addition, EMT also contributes to chemo-resistance in breast and pancreatic cancer [69]. TGF-β signaling drives cell motility and local invasion in non-epithelial cancers as in epithelial cancers. In glioblastoma, TGF-β has been reported to activate EMT drivers ZEB1 and Snail1 [70] as well as surface molecules such as cadherin-11 [71] and integrin [72], which together promote cell motility and local invasion. Moreover, activation of integrin also contributes to TGF-β activation and its transcriptional responses in a positive feedback loop [73]. In particular, TGF-β–Smad2–CITED1-mediated transcription promotes melanoma amoeboid invasion [74]. Taken together, many different studies implicate TGF-β signaling in metastatic dissemination. However, conflicting results have been reported in mouse models. Different tumor types and the use of different interfering strategies sometimes yield opposing results in the potential of TGF-β signaling to initiate the tumor metastasis. For example, TβRII depletion by targeted gene inactivation or dominant-negative interference increases metastasis in polyoma middle-T antigen (PyMT)-tumors [75], but inhibits metastasis of prostate cancer xenografted in mice [76]. In addition, expression of a dominant-negative TβRII promotes metastasis in ErbB2/Neu mouse mammary tumors [77], but expression of activated TGF-β1 has been reported to enhance metastasis in the same model [78]. Overall, the contribution of TGF-β signaling to metastasis still requires further and more comprehensive investigation. On the whole, TGF-β inhibits cell proliferation and stimulates differentiation in normal cells, thus acting as a tumor suppressor. On the contrary, it induces tumor progression and metastasis in advanced cancer, thus serving as a tumor promoter. The dual role of TGF-β on cancer can be explained by the pleiotropic nature of TGF-β. Smad4 may serve as a switch between tumor-suppressive and tumor-promoting activities of TGF-β in pancreatic cancer cells [79]. Mechanistically, TGF-β increases the expression of Sox4 in a Smad4-independent manner, which cooperates with the epithelial lineage determinant Kruppel like factor 5 (Klf5) to promote oncogenic growth. Restoration of Smad4 activity destroys the Sox4–KLF5 interaction and changes Sox4 to induce the transcription of pro-apoptotic genes through a lethal EMT [79]. The tumor-promoting effects of TGF-β on cancer can also be achieved through TGF-β-mediated regulation on tumor microenvironment, including the stroma, and immune responses. Effect of TGF-β on Immune Response in Cancers Cancer progression is dependent on escaping immune surveillance, while TGF-β plays an integral role in regulating immune responses and tumor microenvironment [1,3,80]. TGF-β has pleiotropic effects on both innate and adaptive immune cells, including dendritic cells, macrophages, natural killer (NK) cells, and CD4+ and CD8+ T cells. TGF-β has been reported in stimulating the differentiation of the immune-suppressive regulatory (Treg) cells [1,3]. Given these fundamental roles in immune regulation, TGF-β is therefore an attractive therapeutic target in many immune disorders and in cancer. TGF-β has a profound impact on the myeloid lineage. TGF-β induces monocyte recruitment and promotes differentiation and polarization from M1 into M2 tumor-associated macrophages (TAMs). The M2 TAMs can successively secrete TGF-β supporting tumor promotion [81]. Tumor-derived TGF-β also promotes tumor-associated neutrophils (TANs) [82]. These findings suggest that TGF-β regulates N1–N2 polarization of neutrophils, which in turn decreases activation of intra-tumoral CD8+ T cells. Furthermore, TGF-β signaling can inhibit DC migration and induce their apoptosis [83–85]. The NK cells are a type of cytotoxic lymphocyte critical to the innate immune system [86]. TGF-β has been reported to inhibit NK cell proliferation and function. And in this way, it contributes to a permissive microenvironment for tumor progression. In lung and colorectal cancer, TGF-β inhibits the cytotoxic function of NK cells by down-regulating the NK cell-specific receptor, NKG2D [87]. Moreover, TGF-β decreases NK cell-mediated tumor cell death by inducing repression of MHC class I expression in tumor cells [88]. Overall, increased TGF-β expression within the tumor microenvironment can lead to reduced NK cell cytotoxic activity, decrease the recognition of tumor cells by NK cells and thus block the NK cell-mediated clearance of tumor cells. TGF-β secreted by cancer cells can impact T cell activity by regulating their transcriptional profile. The effect of TGF-β on T cells has been clearly demonstrated by genetic ablation or attenuation of TGF-β signaling. Transgenic mice with a dominant-negative TβRII under a T cell-specific promoter exhibit spontaneous T cell differentiation and autoimmune disease [89]. TGF-β represses cytotoxic gene expression, such as granzyme A, granzyme B, perforin, IFN-γ, and FasL in cytotoxic T cells (CTLs), resulting in tumor cells to escape from immune surveillance [90]. Blockade of TGF-β signaling in T cells supports tumor-specific CD8+ cytotoxic T cells and the clonal expansion of CD8+ T cells in vivo [91]. Inhibition of proliferation and repression of the cytotoxic gene program in are two distinct effects, which ultimately favor tumor progression [91]. TGF-β can act on T helper cell differentiation. TGF-β induces FoxP3 expression and generates induced Treg cells, which supports their phenotype and suppressive functions. In lung tumor, TGF-β secreted by lung cancer cells induces Treg cells in tumor microenvironment [92]. In HCC, TGF-β promotes the differentiation of Treg cells, and the blockade of TGF-β decreases Treg cells in liver tissues and reduces HCC progression [93]. In addition to Treg cells, TGF-β and IL-6 cooperatively induce Th17 cells, which are involved in inflammation [94,95] and inhibit IL-2-dependent T cell proliferation [96]. Moreover, myeloid-derived suppressor cells (MDSCs) can secrete TGF-β [81]. Thus, TGF-β helps tumor cells escape from the immune surveillance and support tumor progression. In general, TGF-β is a potent physiological immune-suppressor in humans. The immunosuppressive response to TGF-β allows tumors to escape from the anti-tumor immune response. In mouse models that systemically lack TGF-β1 also die of systemic inflammation and severe autoimmunity [97,98]. Increased TGF-β in the tumor microenvironment represents a primary mechanism of immune evasion in colon cancer [99]. TGF-β shapes the tumor microenvironment to restrain anti-tumor immunity by restricting T cell infiltration [100]. Generally, TGF-β may be considered an appealing therapeutic target in the context of cancer immunomodulation. TGF-β Targeted Cancer Therapy Drugs that target many different components of the canonical TGF-β signaling pathway have been developed. For instance, strategies include interference with the ligand expression or activity, including ligand traps and antisense oligonucleotides, and inhibition of the intracellular signaling components such as receptor kinase inhibitors and anti-Smad peptide aptamers. Each type has distinct advantages and disadvantages that have to be considered in evaluating their potential for use in the clinic. Fully humanized pan-TGF-β monoclonal neutralizing antibodies have been developed by Genzyme, including Lerdelimumab [101,102], Metelimumab [103], and Fresolimumab (GC1008) [104]. Fresolimumab has shown partial and stable response in a phase I study of advanced melanoma [105]. Fresolimumab had been effectively delivered to recurrent high-grade gliomas but did not result in clinical benefit, probably due to breakdown of the blood brain barrier in this neoplastic tissue [106]. Results to date have provided evidence that Fresolimumab is well tolerated and is not associated with irreversible, life-threatening complications. It indicates that antibody targeting TGF-β may be a novel and productive therapeutic strategy for the treatment of certain cancers. Antisense oligonucleotides have been engineered into immune cells to prevent TGF-β synthesis. Trabedersen (AP12009) is one good example of antisense oligonucleotide that specifically inhibits TGF-β2 expression. In an initial Phase I/II study in patients with high-grade glioma, Trabedersen shows a significant survival benefit over standard chemotherapy [107]. Belagenpumatucel-L, which is an allogeneic tumor cell vaccine that inhibits expression of TGF-β2 and demonstrates enhancement of tumor antigen recognition, is well tolerated and shows survival advantages in a phase II trial in NSCLC at different stages [108]. Although the Phase III trial investigating belagenpumatucel-L in stage III/IV patients did not meet its primary end point, some of the patients experience a survival benefit [109]. Many preclinical studies evaluating TβRI inhibitor drugs have been undertaken [110]. Galunisertib (LY2157299) is a small molecule inhibitor selectively inhibiting the kinase activity of TβRI. It has been investigated either as monotherapy or in combination with standard anti-tumor regimens in different cancer patients, such as glioblastoma, pancreatic cancer, and hepatocellular carcinoma [111]. Galunisertib has completed Phase I [112] and is currently under investigation in several Phase II trials. In the initial Phase I trial this compound showed promising activity in patients with glioblastoma [112]. However, in Phase II trials it failed to show improvement compare with lomustine in second line glioblastoma [113]. On the other side, galunisertib did show superiority over gemcitabine in patients with pancreas cancer [114]. And it has been well tolerated as a first-in-class, oral cancer therapy [111]. All this indicate galunisertib remains a promising compound in clinical development. Another strategy to block TGF-β signaling for cancer is to interfere with the TGF-β signaling molecules such as Smads. This may be achieved by using peptide aptamers. The Trx-SARA aptamer have been designed that bind to Smad2 and Smad3, consequently disrupting their interactions with Smad4. Treatment with Trx-SARA can reduce the level of Smad2/3 in complex with Smad4 after TGF-β stimulation. Furthermore, Trx-SARA treatment has been reported to inhibit TGF-β-induced EMT in NMuMG murine mammary epithelial cells in vitro [115]. Although TGF-β signaling inhibition results in a significant reduction in metastasis in mouse models, clinically the effects have been less obvious as it hoped for. Additionally, it is more difficult to demonstrate inhibition of primary tumors. All these facts suggest that combinatorial therapy may increase the efficacy of TGF-β inhibitors in a clinical setting. Thus, in the next stage of drug development, major efforts may involve the most efficacious drug combinations of TGF-β blockade for each oncological disease application with radiation, chemotherapy, pathway targeted therapies and immunotherapy. Some studies have shown the benefits of combined treatment. For instance, the TβRI/II kinase inhibitor LY2109761 in combination with temozolomine and radiotherapy in a glioblastoma model shows delayed tumor growth compared to controls [116]. Therapeutic co-administration of TGF-β blocker and anti-PD-L1 antibodies in stromal cells facilitates T cell penetration into the center of tumors, and provokes vigorous anti-tumor immunity and tumor regression [100]. On the other hand, as there is considerable variation in both therapeutic and toxicological responses among different patients, development of predictive biomarkers of TGF-β blockade is vitally important. Conclusions and Perspectives The role of TGF-β signaling in tumor cells and stroma has been established based on in vitro and in vivo studies. Strong evidence indicates that in early cancer progression, TGF-β plays a tumor suppressive role, but in later stages it is a potent pro-metastatic mediator. Therefore, a comprehensive understanding of the TGF-β biology in cancer is required to design successful therapeutic approaches. Nowadays, several inhibitors of the TGF-β pathway are being developed and clinically tested for a number of cancers such as glioma, pancreatic cancer, NSCLC, HCC, and melanoma [117]. Although TGF-β targeting agents, such as galunisertib, have shown dramatic therapeutic effects in animal cancer models and in some cancer patients, it is still not clear how the therapeutic effect in cancer patients is achieved. Therefore, it is crucial to consider the complexities in TGF-β signaling when applying TGF-β-related small molecules or protein drugs in clinics. While most translational research has often centered on the pro-tumorigenic of TGF-β on tumor cells in the past, the role of TGF-β to regulate immune evasion has been overlooked. As more and more immune checkpoint inhibitors show promising results in clinical trials, the role of TGF-β as an immune regulator is becoming more valuably investigated. For instance, because TGF-β restricts T cell infiltration and thus attenuates tumor response to PD-L1 blockade, combination of TGF-β-blocking antibody and anti-PD-L1 antibody facilitates penetration of CD8+ T cells in tumors and causes tumor regression [100]. In conclusion, the future of TGF-β inhibitors in cancer therapy as tumor microenvironment targeting agents may hold great promises and open new challenges in cancer therapy. Funding This research was partly supported by the grants from the National Natural Science Foundation of China (No. 31771546), the Zhejiang Provincial Natural Science Foundation (No. LY17C07002) to S.G., the National Natural Science Foundation of China (Nos. 91540205, 31730057, and 31571447) to X.H.F., and the Fundamental Research Funds for the Central Universities. References 1 Derynck R , Miyazono K . The Biology of the TGF-β Family . New York, NY, USA : Cold Spring Harbor Laboratory Press , 2017 , 1164 . 2 Massagué J . TGFβ signalling in context . Nat Rev Mol Cell Biol 2012 , 13 : 616 – 630 . Google Scholar Crossref Search ADS PubMed 3 Chen W , Ten Dijke P . Immunoregulation by members of the TGFβ superfamily . Nat Rev Immunol 2016 , 16 : 723 – 740 . Google Scholar Crossref Search ADS PubMed 4 Wakefield LM , Hill CS . Beyond TGFβ: roles of other TGFβ superfamily members in cancer . Nat Rev Cancer 2013 , 13 : 328 – 341 . Google Scholar Crossref Search ADS PubMed 5 Bierie B , Moses HL . Tumour microenvironment: TGFbeta: the molecular Jekyll and Hyde of cancer . Nat Rev Cancer 2006 , 6 : 506 – 520 . Google Scholar Crossref Search ADS PubMed 6 Lafyatis R . Transforming growth factor β—at the centre of systemic sclerosis . Nat Rev Rheumatol 2014 , 10 : 706 – 719 . Google Scholar Crossref Search ADS PubMed 7 McDonald J , Wooderchak-Donahue W , VanSant Webb C , Whitehead K , Stevenson DA , Bayrak-Toydemir P . Hereditary hemorrhagic telangiectasia: genetics and molecular diagnostics in a new era . Front Genet 2015 , 6 : 1 . Google Scholar Crossref Search ADS PubMed 8 Massagué J . TGFbeta in cancer . Cell 2008 , 134 : 215 – 230 . Google Scholar Crossref Search ADS PubMed 9 Derynck R , Akhurst RJ , Balmain A . TGF-beta signaling in tumor suppression and cancer progression . Nat Genet 2001 , 29 : 117 – 129 . Google Scholar Crossref Search ADS PubMed 10 Feng XH , Derynck R . Specificity and versatility in TGF-beta signaling through Smads . Annu Rev Cell Dev Biol 2005 , 21 : 659 – 693 . Google Scholar Crossref Search ADS PubMed 11 Watanabe M , Masuyama N , Fukuda M , Nishida E . Regulation of intracellular dynamics of Smad4 by its leucine-rich nuclear export signal . EMBO Rep 2000 , 1 : 176 – 182 . Google Scholar Crossref Search ADS PubMed 12 Massagué J , Chen YG . Controlling TGF-beta signaling . Genes Dev 2000 , 14 : 627 – 44 . Google Scholar PubMed 13 Yan X , Liu Z , Chen Y . Regulation of TGF-beta signaling by Smad7 . Acta Biochim Biophys Sin 2009 , 41 : 263 – 272 . Google Scholar Crossref Search ADS PubMed 14 Derynck R , Zhang YE . Smad-dependent and Smad-independent pathways in TGF-beta family signalling . Nature 2003 , 425 : 577 – 584 . Google Scholar Crossref Search ADS PubMed 15 Zhang YE . Mechanistic insight into contextual TGF-β signaling . Curr Opin Cell Biol 2018 , 51 : 1 – 7 . Google Scholar Crossref Search ADS PubMed 16 Pardali K , Kurisaki A , Morén A , ten Dijke P , Kardassis D , Moustakas A . Role of Smad proteins and transcription factor Sp1 in p21(Waf1/Cip1) regulation by transforming growth factor-beta . J Biol Chem 2000 , 275 : 29244 – 29256 . Google Scholar Crossref Search ADS PubMed 17 Hannon GJ , Beach D . p15INK4B is a potential effector of TGF-beta-induced cell cycle arrest . Nature 1994 , 371 : 257 – 261 . Google Scholar Crossref Search ADS PubMed 18 Datto MB , Li Y , Panus JF , Howe DJ , Xiong Y , Wang XF . Transforming growth factor beta induces the cyclin-dependent kinase inhibitor p21 through a p53-independent mechanism . Proc Natl Acad Sci USA 1995 , 92 : 5545 – 5549 . Google Scholar Crossref Search ADS PubMed 19 Li JM , Datto MB , Shen X , Hu PP , Yu Y , Wang XF . Sp1, but not Sp3, functions to mediate promoter activation by TGF-beta through canonical Sp1 binding sites . Nucleic Acids Res 1998 , 26 : 2449 – 2456 . Google Scholar Crossref Search ADS PubMed 20 Hu PP , Shen X , Huang D , Liu Y , Counter C , Wang XF . The MEK pathway is required for stimulation of p21(WAF1/CIP1) by transforming growth factor-beta . J Biol Chem 1999 , 274 : 35381 – 35387 . Google Scholar Crossref Search ADS PubMed 21 Feng XH , 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 Search ADS PubMed 22 Siegel PM , Massagué J . Cytostatic and apoptotic actions of TGF-beta in homeostasis and cancer . Nat Rev Cancer 2003 , 3 : 807 – 820 . Google Scholar Crossref Search ADS PubMed 23 Scandura JM , Boccuni P , Massague J , Nimer SD . Transforming growth factor beta-induced cell cycle arrest of human hematopoietic cells requires p57KIP2 up-regulation . Proc Natl Acad Sci USA 2004 , 101 : 15231 – 15236 . Google Scholar Crossref Search ADS PubMed 24 Frederick JP , Liberati NT , Waddell DS , Shi Y , Wang XF . Transforming growth factor beta-mediated transcriptional repression of c-myc is dependent on direct binding of Smad3 to a novel repressive Smad binding element . Mol Cell Biol 2004 , 24 : 2546 – 2559 . Google Scholar Crossref Search ADS PubMed 25 Gomis RR , Alarcón C , Nadal C , Van Poznak C , Massagué J . C/EBPbeta at the core of the TGFbeta cytostatic response and its evasion in metastatic breast cancer cells . Cancer Cell 2006 , 10 : 203 – 214 . Google Scholar Crossref Search ADS PubMed 26 Chen YG , Lui HM , Lin SL , Lee JM , Ying SY . Regulation of cell proliferation, apoptosis, and carcinogenesis by activin . Exp Biol Med (Maywood) 2002 , 227 : 75 – 87 . Google Scholar Crossref Search ADS PubMed 27 Gomis RR , Alarcon C , He W , Wang Q , Seoane J , Lash A , Massague J , et al. . A FoxO-Smad synexpression group in human keratinocytes . Proc Natl Acad Sci USA 2006 , 103 : 12747 – 12752 . Google Scholar Crossref Search ADS PubMed 28 Feng XH , Liang YY , 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 Search ADS PubMed 29 Seoane J , Pouponnot C , Staller P , Schader M , Eilers M , Massagué J . TGFbeta influences Myc, Miz-1 and Smad to control the CDK inhibitor p15INK4b . Nat Cell Biol 2001 , 3 : 400 – 408 . Google Scholar Crossref Search ADS PubMed 30 Seoane J , Le HV , Massagué J . Myc suppression of the p21(Cip1) Cdk inhibitor influences the outcome of the p53 response to DNA damage . Nature 2002 , 419 : 729 – 734 . Google Scholar Crossref Search ADS PubMed 31 Petritsch C , Beug H , Balmain A , Oft M . TGF-beta inhibits p70 S6 kinase via protein phosphatase 2A to induce G(1) arrest . Genes Dev 2000 , 14 : 3093 – 3101 . Google Scholar Crossref Search ADS PubMed 32 Jang CW , Chen CH , Chen CC , Chen J , Su YH , Chen RH . TGF-beta induces apoptosis through Smad-mediated expression of DAP-kinase . Nat Cell Biol 2002 , 4 : 51 – 58 . Google Scholar Crossref Search ADS PubMed 33 Takekawa M , Tatebayashi K , Itoh F , Adachi M , Imai K , Saito H . Smad-dependent GADD45beta expression mediates delayed activation of p38 MAP kinase by TGF-beta . EMBO J 2002 , 21 : 6473 – 6482 . Google Scholar Crossref Search ADS PubMed 34 Ohgushi M , Kuroki S , Fukamachi H , O'reilly LA , Kuida K , Strasser A , Yonehara S , et al. . Transforming growth factor beta-dependent sequential activation of Smad, Bim, and caspase-9 mediates physiological apoptosis in gastric epithelial cells . Mol Cell Biol 2005 , 25 : 10017 – 10028 . Google Scholar Crossref Search ADS PubMed 35 Ramesh S , Qi XJ , Wildey GM , Robinson J , Molkentin J , Letterio J , Howe PH , et al. . TGF beta-mediated BIM expression and apoptosis are regulated through SMAD3-dependent expression of the MAPK phosphatase MKP2 . EMBO Rep 2008 , 9 : 990 – 997 . Google Scholar Crossref Search ADS PubMed 36 Sorrentino A , Thakur N , Grimsby S , Marcusson A , von Bulow V , Schuster N , Zhang S , et al. . The type I TGF-beta receptor engages TRAF6 to activate TAK1 in a receptor kinase-independent manner . Nat Cell Biol 2008 , 10 : 1199 – 1207 . Google Scholar Crossref Search ADS PubMed 37 Yamashita M , Fatyol K , Jin C , Wang X , Liu Z , Zhang YE . TRAF6 mediates Smad-independent activation of JNK and p38 by TGF-beta . Mol Cell 2008 , 31 : 918 – 924 . Google Scholar Crossref Search ADS PubMed 38 Yamaguchi K , Shirakabe K , Shibuya H , Irie K , Oishi I , Ueno N , Taniguchi T , et al. . Identification of a member of the MAPKKK family as a potential mediator of TGF-beta signal transduction . Science 1995 , 270 : 2008 – 2011 . Google Scholar Crossref Search ADS PubMed 39 Shibuya H , Yamaguchi K , Shirakabe K , Tonegawa A , Gotoh Y , Ueno N , Irie K , et al. . TAB1: an activator of the TAK1 MAPKKK in TGF-beta signal transduction . Science 1996 , 272 : 1179 – 1182 . Google Scholar Crossref Search ADS PubMed 40 Zhang S , Ekman M , Thakur N , Bu S , Davoodpour P , Grimsby S , Tagami S , et al. . TGFbeta1-induced activation of ATM and p53 mediates apoptosis in a Smad7-dependent manner . Cell Cycle 2006 , 5 : 2787 – 2795 . Google Scholar Crossref Search ADS PubMed 41 Sánchez A , Álvarez AM , Benito M , Fabregat I . Apoptosis induced by transforming growth factor-beta in fetal hepatocyte primary cultures: involvement of reactive oxygen intermediates . J Biol Chem 1996 , 271 : 7416 – 7422 . Google Scholar Crossref Search ADS PubMed 42 Ramjaun AR , Tomlinson S , Eddaoudi A , Downward J . Upregulation of two BH3-only proteins, Bmf and Bim, during TGF beta-induced apoptosis . Oncogene 2007 , 26 : 970 – 981 . Google Scholar Crossref Search ADS PubMed 43 Swarbrick A , Roy E , Allen T , Bishop JM . Id1 cooperates with oncogenic Ras to induce metastatic mammary carcinoma by subversion of the cellular senescence response . Proc Natl Acad Sci USA 2008 , 105 : 5402 – 5407 . Google Scholar Crossref Search ADS PubMed 44 Tang B , Yoo N , Vu M , Mamura M , Nam JS , Ooshima A , Du Z , et al. . Transforming growth factor-beta can suppress tumorigenesis through effects on the putative cancer stem or early progenitor cell and committed progeny in a breast cancer xenograft model . Cancer Res 2007 , 67 : 8643 – 8652 . Google Scholar Crossref Search ADS PubMed 45 Hanahan D , Weinberg RA . Hallmarks of cancer: the next generation . Cell 2011 , 144 : 646 – 674 . Google Scholar Crossref Search ADS PubMed 46 Ikushima H , Miyazono K . TGFbeta signalling: a complex web in cancer progression . Nat Rev Cancer 2010 , 10 : 415 – 424 . Google Scholar Crossref Search ADS PubMed 47 Hirai H , Izutsu K , Kurokawa M , Mitani K . Oncogenic mechanisms of Evi-1 protein . Cancer Chemother Pharmacol 2001 , 48 : S35 – S40 . Google Scholar Crossref Search ADS PubMed 48 Feng XH , Liang YY , Liang M , Zhai W , Lin X . Direct interaction of c-Myc with Smad2 and Smad3 to inhibit TGF-beta-mediated induction of the CDK inhibitor p15(Ink4B) . Mol Cell 2002 , 9 : 133 – 143 . Google Scholar Crossref Search ADS PubMed 49 Alliston T , Ko TC , Cao Y , Liang YY , Feng XH , Chang C , Derynck R , et al. . Repression of bone morphogenetic protein and activin-inducible transcription by Evi-1 . J Biol Chem 2005 , 280 : 24227 – 24237 . Google Scholar Crossref Search ADS PubMed 50 Wang D , Long J , Dai F , Liang M , Feng XH , Lin X . BCL6 represses Smad signaling in transforming growth factor-beta resistance . Cancer Res 2008 , 68 : 783 – 789 . Google Scholar Crossref Search ADS PubMed 51 Deheuninck J , Luo K . Ski and SnoN, potent negative regulators of TGF-beta signaling . Cell Res 2009 , 19 : 47 – 57 . Google Scholar Crossref Search ADS PubMed 52 Kiyono K , Suzuki HI , Morishita Y , Komuro A , Iwata C , Yashiro M , Hirakawa K , et al. . c-Ski overexpression promotes tumor growth and angiogenesis through inhibition of transforming growth factor-beta signaling in diffuse-type gastric carcinoma . Cancer Sci 2009 , 100 : 1809 – 1816 . Google Scholar Crossref Search ADS PubMed 53 Wang G , Yu Y , Sun C , Liu T , Liang T , Zhan L , Lin X , et al. . Erratum: STAT3 selectively interacts with Smad3 to antagonize TGF-β signalling . Oncogene 2016 , 35 : 4422 . Google Scholar Crossref Search ADS PubMed 54 Moustakas A , Heldin CH . Mechanisms of TGFβ-induced epithelial-mesenchymal transition . J Clin Med 2016 , 5 : 63 . Google Scholar Crossref Search ADS 55 Moustakas A , Heldin P . TGFβ and matrix-regulated epithelial to mesenchymal transition . Biochim Biophys Acta 2014 , 1840 : 2621 – 2634 . Google Scholar Crossref Search ADS PubMed 56 Vervoort SJ , Lourenço AR , van Boxtel R , Coffer PJ . SOX4 mediates TGF-β-induced expression of mesenchymal markers during mammary cell epithelial to mesenchymal transition . PLoS One 2013 , 8 : e53238 . Google Scholar Crossref Search ADS PubMed 57 Bae GY , Hong SK , Park JR , Kwon OS , Kim KT , Koo J , Oh E , et al. . Chronic TGFβ stimulation promotes the metastatic potential of lung cancer cells by Snail protein stabilization through integrin β3-Akt-GSK3β signaling . Oncotarget 2016 , 7 : 25366 – 76 . Google Scholar PubMed 58 Mise N , Savai R , Yu H , Schwarz J , Kaminski N , Eickelberg O . Zyxin is a transforming growth factor-β (TGF-β)/Smad3 target gene that regulates lung cancer cell motility via integrin α5β1 . J Biol Chem 2012 , 287 : 31393 – 31405 . Google Scholar Crossref Search ADS PubMed 59 Salvo E , Garasa S , Dotor J , Morales X , Peláez R , Altevogt P , Rouzaut A , et al. . Combined targeting of TGF-β1 and integrin β3 impairs lymph node metastasis in a mouse model of non-small-cell lung cancer . Mol Cancer 2014 , 13 : 112 . Google Scholar Crossref Search ADS PubMed 60 Parvani JG , Gujrati MD , Mack MA , Schiemann WP , Lu ZR . Silencing β3 integrin by targeted ECO/siRNA nanoparticles inhibits EMT and metastasis of triple-negative breast cancer . Cancer Res 2015 , 75 : 2316 – 2325 . Google Scholar Crossref Search ADS PubMed 61 Franco DL , Mainez J , Vega S , Sancho P , Murillo MM , de Frutos CA , del Castillo G , et al. . Snail1 suppresses TGF-beta-induced apoptosis and is sufficient to trigger EMT in hepatocytes . J Cell Sci 2010 , 123 : 3467 – 3477 . Google Scholar Crossref Search ADS PubMed 62 Bertran E , Crosas-Molist E , Sancho P , Caja L , Lopez-Luque J , Navarro E , Egea G , et al. . Overactivation of the TGF-β pathway confers a mesenchymal-like phenotype and CXCR4-dependent migratory properties to liver tumor cells . Hepatology 2013 , 58 : 2032 – 2044 . Google Scholar Crossref Search ADS PubMed 63 Thakur N , Gudey SK , Marcusson A , Fu JY , Bergh A , Heldin CH , Landström M , et al. . TGFβ-induced invasion of prostate cancer cells is promoted by c-Jun-dependent transcriptional activation of Snail1 . Cell Cycle 2014 , 13 : 2400 – 2414 . Google Scholar Crossref Search ADS PubMed 64 Pu H , Horbinski C , Hensley PJ , Matuszak EA , Atkinson T , Kyprianou N . PARP-1 regulates epithelial-mesenchymal transition (EMT) in prostate tumorigenesis . Carcinogenesis 2014 , 35 : 2592 – 2601 . Google Scholar Crossref Search ADS PubMed 65 Shiota M , Zardan A , Takeuchi A , Kumano M , Beraldi E , Naito S , Zoubeidi A , et al. . Clusterin mediates TGF-β-induced epithelial-mesenchymal transition and metastasis via Twist1 in prostate cancer cells . Cancer Res 2012 , 72 : 5261 – 5272 . Google Scholar Crossref Search ADS PubMed 66 Polyak K , Weinberg RA . Transitions between epithelial and mesenchymal states: acquisition of malignant and stem cell traits . Nat Rev Cancer 2009 , 9 : 265 – 273 . Google Scholar Crossref Search ADS PubMed 67 Gupta GP , Perk J , Acharyya S , de Candia P , Mittal V , Todorova-Manova K , Gerald WL , et al. . ID genes mediate tumor reinitiation during breast cancer lung metastasis . Proc Natl Acad Sci USA 2007 , 104 : 19506 – 19511 . Google Scholar Crossref Search ADS PubMed 68 Stankic M , Pavlovic S , Chin Y , Brogi E , Padua D , Norton L , Massagué J , et al. . TGF-β-Id1 signaling opposes Twist1 and promotes metastatic colonization via a mesenchymal-to-epithelial transition . Cell Rep 2013 , 5 : 1228 – 1242 . Google Scholar Crossref Search ADS PubMed 69 Zheng X , Carstens JL , Kim J , Scheible M , Kaye J , Sugimoto H , Wu CC , et al. . Epithelial-to-mesenchymal transition is dispensable for metastasis but induces chemoresistance in pancreatic cancer . Nature 2015 , 527 : 525 – 530 . Google Scholar Crossref Search ADS PubMed 70 Nana AW , Yang PM , Lin HY . Overview of transforming growth factor β superfamily involvement in glioblastoma initiation and progression . Asian Pac J Cancer Prev 2015 , 16 : 6813 – 6823 . Google Scholar Crossref Search ADS PubMed 71 Schulte JD , Srikanth M , Das S , Zhang J , Lathia JD , Yin L , Rich JN , et al. . Cadherin-11 regulates motility in normal cortical neural precursors and glioblastoma . PLoS One 2013 , 8 : e70962 . Google Scholar Crossref Search ADS PubMed 72 Cosset EC , Godet J , Entz-Werlé N , Guérin E , Guenot D , Froelich S , Bonnet D , et al. . Involvement of the TGFβ pathway in the regulation of α5 β1 integrins by caveolin-1 in human glioblastoma . Int J Cancer 2012 , 131 : 601 – 611 . Google Scholar Crossref Search ADS PubMed 73 Roth P , Silginer M , Goodman SL , Hasenbach K , Thies S , Maurer G , Schraml P , et al. . Integrin control of the transforming growth factor-β pathway in glioblastoma . Brain 2013 , 136 : 564 – 576 . Google Scholar Crossref Search ADS PubMed 74 Cantelli G , Orgaz JL , Rodriguez-Hernandez I , Karagiannis P , Maiques O , Matias-Guiu X , Nestle FO , et al. . TGF-β-induced transcription sustains amoeboid melanoma migration and dissemination . Curr Biol 2015 , 25 : 2899 – 2914 . Google Scholar Crossref Search ADS PubMed 75 Forrester E , Chytil A , Bierie B , Aakre M , Gorska AE , Sharif-Afshar AR , Muller WJ , et al. . Effect of conditional knockout of the type II TGF-beta receptor gene in mammary epithelia on mammary gland development and polyomavirus middle T antigen induced tumor formation and metastasis . Cancer Res 2005 , 65 : 2296 – 2302 . Google Scholar Crossref Search ADS PubMed 76 Zhang F , Lee J , Lu S , Pettaway CA , Dong Z . Blockade of transforming growth factor-beta signaling suppresses progression of androgen-independent human prostate cancer in nude mice . Clin Cancer Res 2005 , 11 : 4512 – 4520 . Google Scholar Crossref Search ADS PubMed 77 Novitskiy SV , Forrester E , Pickup MW , Gorska AE , Chytil A , Aakre M , Polosukhina D , et al. . Attenuated transforming growth factor beta signaling promotes metastasis in a model of HER2 mammary carcinogenesis . Breast Cancer Res 2014 , 16 : 425 . Google Scholar Crossref Search ADS PubMed 78 Muraoka RS , Koh Y , Roebuck LR , Sanders ME , Brantley-Sieders D , Gorska AE , Moses HL , et al. . Increased malignancy of Neu-induced mammary tumors overexpressing active transforming growth factor beta1 . Mol Cell Biol 2003 , 23 : 8691 – 8703 . Google Scholar Crossref Search ADS PubMed 79 David CJ , Huang YH , Chen M , Su J , Zou Y , Bardeesy N , Iacobuzio-Donahue CA , et al. . TGF-β tumor suppression through a lethal EMT . Cell 2016 , 164 : 1015 – 1030 . Google Scholar Crossref Search ADS PubMed 80 Pickup M , Novitskiy S , Moses HL . The roles of TGFβ in the tumour microenvironment . Nat Rev Cancer 2013 , 13 : 788 – 799 . Google Scholar Crossref Search ADS PubMed 81 Condamine T , Ramachandran I , Youn JI , Gabrilovich DI . Regulation of tumor metastasis by myeloid-derived suppressor cells . Annu Rev Med 2015 , 66 : 97 – 110 . Google Scholar Crossref Search ADS PubMed 82 Fridlender ZG , Sun J , Kim S , Kapoor V , Cheng G , Ling L , Worthen GS , et al. . Polarization of tumor-associated neutrophil phenotype by TGF-beta: "N1" versus "N2" TAN . Cancer Cell 2009 , 16 : 183 – 194 . Google Scholar Crossref Search ADS PubMed 83 Ito M , Minamiya Y , Kawai H , Saito S , Saito H , Nakagawa T , Imai K , et al. . Tumor-derived TGFbeta-1 induces dendritic cell apoptosis in the sentinel lymph node . J Immunol 2006 , 176 : 5637 – 5643 . Google Scholar Crossref Search ADS PubMed 84 Imai K , Minamiya Y , Koyota S , Ito M , Saito H , Sato Y , Motoyama S , et al. . Inhibition of dendritic cell migration by transforming growth factor-β1 increases tumor-draining lymph node metastasis . J Exp Clin Cancer Res 2012 , 31 : 3 . Google Scholar Crossref Search ADS PubMed 85 Kobie JJ , Wu RS , Kurt RA , Lou S , Adelman MK , Whitesell LJ , Ramanathapuram LV , et al. . Transforming growth factor beta inhibits the antigen-presenting functions and antitumor activity of dendritic cell vaccines . Cancer Res 2003 , 63 : 1860 – 1864 . Google Scholar PubMed 86 Kiessling R , Klein E , Pross H , Wigzell H . "Natural" killer cells in the mouse. II. Cytotoxic cells with specificity for mouse Moloney leukemia cells. Characteristics of the killer cell . Eur J Immunol 1975 , 5 : 117 – 121 . Google Scholar Crossref Search ADS PubMed 87 Lee JC , Lee KM , Kim DW , Heo DS . Elevated TGF-beta1 secretion and down-modulation of NKG2D underlies impaired NK cytotoxicity in cancer patients . J Immunol 2004 , 172 : 7335 – 7340 . Google Scholar Crossref Search ADS PubMed 88 Ma D , Niederkorn JY . Transforming growth factor-beta down-regulates major histocompatibility complex class I antigen expression and increases the susceptibility of uveal melanoma cells to natural killer cell-mediated cytolysis . Immunology 1995 , 86 : 263 – 269 . Google Scholar PubMed 89 Gorelik L , Flavell RA . Transforming growth factor-beta in T-cell biology . Nat Rev Immunol 2002 , 2 : 46 – 53 . Google Scholar Crossref Search ADS PubMed 90 Thomas DA , 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 Search ADS PubMed 91 Gorelik L , Flavell RA . Immune-mediated eradication of tumors through the blockade of transforming growth factor-beta signaling in T cells . Nat Med 2001 , 7 : 1118 – 1122 . Google Scholar Crossref Search ADS PubMed 92 Hao NB , Lü MH , Fan YH , Cao YL , Zhang ZR , Yang SM . Macrophages in tumor microenvironments and the progression of tumors . Clin Dev Immunol 2012 , 2012 : 948098 . Google Scholar Crossref Search ADS PubMed 93 Shen Y , Wei Y , Wang Z , Jing Y , He H , Yuan J , Li R , et al. . TGF-β regulates hepatocellular carcinoma progression by inducing Treg cell polarization . Cell Physiol Biochem 2015 , 35 : 1623 – 1632 . Google Scholar Crossref Search ADS PubMed 94 Mangan PR , Harrington LE , O'quinn DB , Helms WS , Bullard DC , Elson CO , Hatton RD , et al. . Transforming growth factor-beta induces development of the T(H)17 lineage . Nature 2006 , 441 : 231 – 234 . Google Scholar Crossref Search ADS PubMed 95 Veldhoen M , Hocking RJ , Atkins CJ , Locksley RM , Stockinger B . TGFbeta in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17-producing T cells . Immunity 2006 , 24 : 179 – 189 . Google Scholar Crossref Search ADS PubMed 96 Kehrl JH , Wakefield LM , Roberts AB , Jakowlew S , Alvarez-Mon M , Derynck R , Sporn MB , et al. . Production of transforming growth factor beta by human T lymphocytes and its potential role in the regulation of T cell growth . J Exp Med 1986 , 163 : 1037 – 1050 . Google Scholar Crossref Search ADS PubMed 97 Diebold RJ , Eis MJ , Yin M , Ormsby I , Boivin GP , Darrow BJ , Saffitz JE , et al. . Early-onset multifocal inflammation in the transforming growth factor beta 1-null mouse is lymphocyte mediated . Proc Natl Acad Sci USA 1995 , 92 : 12215 – 12219 . Google Scholar Crossref Search ADS PubMed 98 Shull MM , Ormsby I , Kier AB , Pawlowski S , Diebold RJ , Yin M , Allen R , et al. . Targeted disruption of the mouse transforming growth factor-beta 1 gene results in multifocal inflammatory disease . Nature 1992 , 359 : 693 – 699 . Google Scholar Crossref Search ADS PubMed 99 Tauriello DVF , Palomo-Ponce S , Stork D , Berenguer-Llergo A , Badia-Ramentol J , Iglesias M , Sevillano M , et al. . TGFβ drives immune evasion in genetically reconstituted colon cancer metastasis . Nature 2018 , 554 : 538 – 543 . Google Scholar Crossref Search ADS PubMed 100 Mariathasan S , Turley SJ , Nickles D , Castiglioni A , Yuen K , Wang Y , Kadel III EE , et al. . TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells . Nature 2018 , 554 : 544 – 548 . Google Scholar Crossref Search ADS PubMed 101 Mead AL , Wong TTL , Cordeiro MF , Anderson IK , Khaw PT . Evaluation of anti-TGF-beta 2 antibody as a new postoperative anti-scarring agent in glaucoma surgery . Invest Ophthalmol Vis Sci 2003 , 44 : 3394 – 3401 . Google Scholar Crossref Search ADS PubMed 102 Thompson JE , Vaughan TJ , Williams AJ , Wilton J , Johnson KS , Bacon L , Green JA , et al. . A fully human antibody neutralising biologically active human TGF beta 2 for use in therapy . J Immunol Methods 1999 , 227 : 17 – 29 . Google Scholar Crossref Search ADS PubMed 103 Cordeiro MF , Gay JA , Khaw PT . Human anti-transforming growth factor-beta2 antibody: a new glaucoma anti-scarring agent . Invest Ophthalmol Vis Sci 1999 , 40 : 2225 – 2234 . Google Scholar PubMed 104 Morris JC , Shapiro GI , Tan AR , Lawrence DP , Olencki TE , Dezube BJ , Hsu FJ , et al. . Phase I/II study of GC1008: a human anti-transforming growth factor-beta (TGFβ) monoclonal antibody (MAb) in patients with advanced malignant melanoma (MM) or renal cell carcinoma (RCC) . J Clin Oncol 2008 , 26 : 9028 . Google Scholar Crossref Search ADS 105 Morris JC , Tan AR , Olencki TE , Shapiro GI , Dezube BJ , Reiss M , Hsu FJ , et al. . Phase I study of GC1008 (fresolimumab): a human anti-transforming growth factor-beta (TGFβ) monoclonal antibody in patients with advanced malignant melanoma or renal cell carcinoma . PLoS One 2014 , 9 : e90353 . Google Scholar Crossref Search ADS PubMed 106 den Hollander MW , Bensch F , Glaudemans AWJM , Oude Munnink TH , Enting RH , den Dunnen WFA , Heesters MAAM , et al. . TGF-β antibody uptake in recurrent high-grade glioma imaged with 89Zr-Fresolimumab PET . J Nucl Med 2015 , 56 : 1310 – 1314 . Google Scholar Crossref Search ADS PubMed 107 Hau P , Jachimczak P , Schlingensiepen R , Schulmeyer F , Jauch T , Steinbrecher A , Brawanski A , et al. . Inhibition of TGF-beta 2 with AP 12009 in recurrent malignant gliomas: from preclinical to phase I/II studies . Oligonucleotides 2007 , 17 : 201 – 212 . Google Scholar Crossref Search ADS PubMed 108 Nemunaitis J , Dillman RO , Schwarzenberger PO , Senzer N , Cunningham C , Cutler J , Tong A , et al. . Phase II study of belagenpumatucel-L, a transforming growth factor beta-2 antisense gene-modified allogeneic tumor cell vaccine in non-small-cell lung cancer . J Clin Oncol 2006 , 24 : 4721 – 4730 . Google Scholar Crossref Search ADS PubMed 109 Rijavec E , Biello F , Genova C , Barletta G , Maggioni C , Dal Bello MG , Coco S , et al. . Belagenpumatucel-L for the treatment of non-small cell lung cancer . Expert Opin Biol Ther 2015 , 15 : 1371 – 1379 . Google Scholar Crossref Search ADS PubMed 110 Akhurst RJ , Hata A . Targeting the TGFβ signalling pathway in disease . Nat Rev Drug Discov 2012 , 11 : 790 – 811 . Google Scholar Crossref Search ADS PubMed 111 Herbertz S , Sawyer JS , Stauber AJ , Gueorguieva I , Driscoll KE , Estrem ST , Cleverly AL , et al. . Clinical development of galunisertib (LY2157299 monohydrate), a small molecule inhibitor of transforming growth factor-beta signaling pathway . Drug Design Dev Ther 2015 , 9 : 4479 – 4499 . 112 Rodon J , Carducci MA , Sepulveda-Sánchez JM , Azaro A , Calvo E , Seoane J , Braña I , et al. . First-in-human dose study of the novel transforming growth factor-β receptor I kinase inhibitor LY2157299 monohydrate in patients with advanced cancer and glioma . Clin Cancer Res 2015 , 21 : 553 – 560 . Google Scholar Crossref Search ADS PubMed 113 Brandes AA , Carpentier AF , Kesari S , Sepulveda-Sanchez JM , Wheeler HR , Chinot O , Cher L , et al. . A phase II randomized study of galunisertib monotherapy or galunisertib plus lomustine compared with lomustine monotherapy in patients with recurrent glioblastoma . Neuro Oncol 2016 , 18 : 1146 – 1156 . Google Scholar Crossref Search ADS PubMed 114 Melisi D , Garcia-Carbonero R , Macarulla T , Pezet D , Deplanque G , Fuchs M , et al. . A randomized phase II, double-blind study to evaluate the efficacy and safety of galunisertib plus gemcitabine (GG) or gemcitabine plus placebo (GP) in patients with unresectable pancreatic cancer (PC) . Cancer Res 2016 , 76(14 Supplement): CT068 – CT068 . 115 Zhao BM , Hoffmann FM . Inhibition of transforming growth factor-beta1-induced signaling and epithelial-to-mesenchymal transition by the Smad-binding peptide aptamer Trx-SARA . Mol Biol Cell 2006 , 17 : 3819 – 3831 . Google Scholar Crossref Search ADS PubMed 116 Zhang M , Kleber S , Rohrich M , Timke C , Han N , Tuettenberg J , Martin-Villalba A , et al. . Blockade of TGF-β signaling by the TGFβR-I kinase inhibitor LY2109761 enhances radiation response and prolongs survival in glioblastoma . Cancer Res 2011 , 71 : 7155 – 7167 . Google Scholar Crossref Search ADS PubMed 117 Giannelli G , Mikulits W , Dooley S , Fabregat I , Moustakas A , Ten Dijke P , Portincasa P , et al. . The rationale for targeting TGF-β in chronic liver diseases . Eur J Clin Invest 2016 , 46 : 349 – 361 . Google Scholar Crossref Search ADS PubMed © The Author(s) 2018. Published by Oxford University Press on behalf of the Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic.oup.com/journals/pages/open_access/funder_policies/chorus/standard_publication_model) TI - TGF-β signaling in cancer JF - Acta Biochimica et Biophysica Sinica DO - 10.1093/abbs/gmy092 DA - 2018-10-01 UR - https://www.deepdyve.com/lp/oxford-university-press/tgf-signaling-in-cancer-aFS0h0QD0c SP - 941 VL - 50 IS - 10 DP - DeepDyve ER -