Article Search
닫기

Journal of Cancer Prevention

Review

J Cancer Prev 2020; 25(4): 213-222

Published online December 30, 2020

https://doi.org/10.15430/JCP.2020.25.4.213

© Korean Society of Cancer Prevention

Recent Advances in the Development of TGF-β Signaling Inhibitors for Anticancer Therapy

Ho-Jae Lee

Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, Gachon University College of Medicine, Incheon, Korea

Correspondence to :
Ho-Jae Lee, E-mail: hojlee@gachon.ac.kr, https://orcid.org/0000-0003-2170-355X

Received: December 1, 2020; Revised: December 13, 2020; Accepted: December 14, 2020

TGF-β is a multifunctional cytokine that plays an important role in both physiologic and pathologic processes, including cancer. Importantly, TGF-β has a dual role in tumorigenesis, acting as a tumor suppressor or a tumor promoter, depending on the stage of tumor development. The aberrantly upregulated production of TGF-β has been strongly implicated in tumor progression, angiogenesis, and metastasis, as well as immune evasion. Therefore, hyperactivated TGF-β signaling is considered a potential therapeutic target for cancer therapy. Numerous inhibitors of overactivated TGF-β signaling have been developed, and some of them are currently in clinical trials. This review focuses on the TGF-β signaling that contributes to tumor progression and immune evasion in the tumor microenvironment and presents recent achievements on TGF-β signaling inhibition as a single or combined therapeutic approach in cancer therapy.

Keywords: TGF-β, Tumor microenvironment, Tumor escape, Antineoplastic agents, Immune checkpoint inhibitors

TGF-β was initially identified by its ability to transform and induce anchorage-independent growth of non-transformed fibroblasts [1]. TGF-β is now known to be a multifunctional cytokine that regulates various physiological processes including, cell growth, differentiation, apoptosis, embryonic development, immune responses, and the synthesis of extracellular matrix proteins [2]. Although the effects of the TGF-β signaling depend on the cellular and tissue context, dysregulation of the TGF-β signaling pathway has been involved in the development of human diseases, including cancer [3]. In recent decades, there has been a growing emphasis on the role of the TGF-β signaling pathway in promoting tumor development and progression and inducing an immunosuppressive tumor microenvironment [4,5]. Multiple lines of preclinical evidence suggest that the inhibition of the TGF-β signaling pathway exhibits antitumor activity in various animal models. Notably, high levels of TGF-β expression are correlated with poor prognosis and metastasis and associated with resistance to chemotherapy and shorter survival [6,7].

Based on this rationale, several types of TGF-β signaling inhibitors have been developed, and some of them are currently in clinical trials for cancer treatment. Immunotherapy has recently led to a paradigm shift in cancer therapy, amongst which immune checkpoint inhibitors are the most representative agents approved for treating various advanced solid tumors [8]. Of note, many recent studies have demonstrated that the TGF-β signaling pathway plays a pivotal role in cancer immune escape and resistance to the immune checkpoint inhibitor therapy [9,10]. Therefore, concurrent inhibition of the TGF-β and the immune checkpoint pathway may be a useful strategy to enhance the therapeutic efficacy of immunotherapy.

TGF-β is synthesized as a latent complex consisting of a mature TGF-β homodimer non-covalently linked to the latency associated protein (LAP) forming the small latent complex (SLC) and the latent TGF-β binding molecule (LTBP), which interacts with extracellular matrix (ECM) [11]. Latent TGF-β can be activated by proteolytic cleavage of LAP by furin, plasmin, and matrix metalloproteinases (MMPs), or by binding to thrombospondin-1 (TSP-1) or αvβ6 integrin [12,13]. Once released, active TGF-β binds to a serine/threonine kinase type II receptor (TGF-β RII), which recruits the type I receptor (TGF-β RI) and activates it by phosphorylation. Activated TGF-β RI phosphorylates the receptor-regulated Smads (R-Smads), Smad2, and Smad3. Subsequently, phosphorylated R-Smads form a complex with Smad4 and then translocate into the nucleus. In the nucleus, the heteromeric Smad complex regulates transcription through its ability to interact with DNA-binding and recruit transcription cofactors [14]. Besides the Smad-dependent canonical pathway, TGF-β receptors can also activate Smad-independent pathways such as mitogen-activated protein kinases (MAPKs), phosphatidylinositol 3-kinase-AKT (PI-3K/AKT), NF-κB, the small GTPases Rac/Cdc42 and RhoA in a context-dependent manner [15].

TGF-β plays an important but paradoxical role in carcinogenesis and tumor development (Fig. 1). In normal or early stages of tumors, TGF-β acts as a tumor suppressor by inhibiting proliferation and inducing apoptosis of cancer cells. However, during progression, cancer cells become resistant to TGF-β-induced antitumor effects through the acquisition of mutations in the signaling components of the pathway, and then the TGF-β pathway shifts from tumor-suppressive to tumor-promoting functions [16]. In late-stage tumors, TGF-β signaling promotes epithelial-mesenchymal transition (EMT) and invasion, evasion of immune surveillance, and metastatic spread [4]. Also, the tumor-promoting function of TGF-β depends on its effect on the cancer cells and the tumor microenvironment [17].

Figure 1. The dual role of TGF-β signaling in tumor progression. TGF-β induces apoptosis and inhibits the proliferation of normal or nontransformed cells but loses its growth inhibitory properties as cells progress to later stages of tumorigenesis. In later stages of tumor progression, TGF-β is actively secreted by cancer cells or stromal cells and acts as a tumor promoter by promoting EMT, cancer cell migration and invasion, and the formation of an immunosuppressive tumor microenvironment. EMT, epithelial-mesenchymal transition.

Role of TGF-β in tumor microenvironment

Solid tumors are not merely composed of cancer cells alone, but they are complex and heterogeneous structures composed of malignant cells that recruit various normal cells, such as fibroblasts, endothelial cells, and immune cells, to promote tumor growth. ECM serves as a structural framework for cell migration and as a reservoir for latent TGF-β, and modulates its activation and turnover [18]. The tumor stroma is a critical component of the tumor microenvironment and is recognized as an essential contributor to tumor growth, metastasis, and immune evasion [19].

TGF-β is normally present at high concentrations in the tumor microenvironment. The increased TGF-β is attributable to the production by both cancer cells and stromal cells, especially cancer-associated fibroblasts (CAFs), immune cells, such as regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) as well as tumor-associated macrophages (TAMs) [20]. The interplay between these cells within the tumor microenvironment promotes tumor progression by stimulating survival signaling of cancer cells, promoting EMT, angiogenesis, metastasis, and immune evasion [21,22].

While EMT is a physiological process required for normal embryonic development and wound healing in which epithelial cells differentiate into mesenchymal cells, EMT also plays a crucial role in tumor progression and metastasis. TGF-β, which is secreted by cancer cells in an autocrine loop, or a paracrine fashion by stromal and immune cells in the tumor microenvironment, is a potent inducer of EMT [23,24]. TGF-β induces EMT by activating the Smad-dependent and/or Smad-independent transcriptional pathway, which induces the expression of EMT-activating transcription factors, such as Snail (SNAI1), Slug (SNAI2), Zeb1 (TCF8), Zeb2 (SIP1), and Twist [25,26]. The Smad-independent signaling pathways, such as those mediated by the Rho-Rock1 and the AKT, play important roles in promoting cancer cell migration and the manifestation of invasive cellular phenotypes [27]. Cancer cells undergoing EMT acquire invasive properties and enter the surrounding stroma, thereby creating a favorable microenvironment for successful metastasis. EMT is also associated with the acquisition of cancer stem cell-like characteristics and thereby conferring a more aggressive phenotype and resistance to radiotherapy or chemotherapy [28,29].

Angiogenesis in the tumor microenvironment is an essential process for tumor growth and metastasis. New blood vessels can supply the nutrients and oxygen to the cancer cells and allow them to have access to the blood system and metastasize to distant sites [30]. TGF-β signaling plays an essential role in normal angiogenesis and vascular integrity, and also neovascularization in the tumor microenvironment [31]. TGF-β directly induces the expression of VEGF, the most potent proangiogenic mediator [32]. TGF-β also recruits endothelial cells, CAFs, and TAMs, which in turn produce proangiogenic factors [33]. TGF-β-mediated MAPK activation promotes cancer cell invasion and tumor angiogenesis through the upregulated expression of MMP-9, which plays an essential role in ECM degradation during tumor angiogenesis and metastasis [34].

CAFs are among the most abundant stromal cell types within the tumor microenvironment and promote tumor progression by supporting cancer cell growth and ECM remodeling, tumor-promoting inflammation, and metastasis [35]. TGF-β promotes the differentiation of fibroblasts to myofibroblasts and stimulates the conversion of normal fibroblasts to CAFs. Activated CAFs enhance angiogenesis by secreting proangiogenic factors, such as VEGF, hypoxia-inducible factor, platelet-derived growth factor, and stromal cell-derived factor-1 [36]. The high density of activated CAFs in the t is closely associated with poor survival, therapy resistance, and recurrence in multiple solid cancers [37,38].

Role of TGF-β in tumor immune evasion

Tumors can escape the host immune system. The malignant tumor cells effectively modulate cellular processes by releasing soluble factors, such as immunosuppressive cytokines (e.g., TGF-β) to protect antitumor immune effector cells. Besides promoting tumor invasion and angiogenesis, TGF-β also plays a critical role in regulating the immune response in the tumor microenvironment [25,39]. TGF-β functions primarily as an immunosuppressive cytokine (Fig. 2). The first in vivo evidence that TGF-β signaling regulates immune response was demonstrated by knockout studies [40,41]. The lack of TGF-β1 results in severe inflammatory responses by activating the immune cell population and the infiltration of lymphocytes and macrophages in various organs.

Figure 2. TGF-β secretion in the tumor microenvironment and its effects on immune cells. TGF-β is secreted by cancer cells and stromal cells and exerts immunosuppressive roles for the immune cells involved in the antitumor immune responses. Treg, regulatory T cells; Th17, T helper 17 cells; CTL, cytotoxic T lymphocytes; NK, natural killer cells; MΦ, macrophages; DC, dendritic cells.

Previous studies have demonstrated that TGF-β, secreted by cancer cells and neighboring immune cells, such as Tregs and myeloid suppressor cells, suppress the antitumor activity of effector cells, including CD8+ T cells, natural killer (NK) cells, and macrophages [39]. CD8+ T cells are referred to as cytotoxic T lymphocytes (CTLs), given their ability to kill target cells. TGF-β suppresses the growth and differentiation of CTLs through Smad-dependent downregulation of c-Myc and IL-2, and upregulation of p21 and p27 [42]. NK cells can kill malignant cells without activation process and enhance antitumor effects by secreting cytokines such as IFN-γ and tumor necrosis factor-α (TNF-α). TGF-β signaling inhibits NK cell functions in multiple ways. TGF-β suppresses the expression of IFN-γ in NK cells, thereby blocking Th2 responses [43]. TAMs are also an important source of TGF-β in the tumor microenvironment and are closely associated with poor prognosis and resistance to antitumor agents. TGF-β inhibits the inflammatory response of macrophages mediated by TNF-α/NF-κB pathway [44]. TGF-β signaling also plays a critical role in inducing Tregs, which suppress the function of effector T cells and participate in cancer immune evasion [45]. TGF-β induces a forkhead box protein 3 (Foxp3), a transcription factor essential for developing Tregs, through Smad3 activation [46].

Recently published data from Tauriello et al. [47] showed that the inhibition of TGF-β signaling with a TGF-β RI kinase inhibitor triggers the infiltration of CTLs, and renders tumors susceptible to immune checkpoint therapies. Likewise, the combination of TGF-β and immune checkpoint pathway inhibitors enhances CTL response and drives potent antitumor immunity and tumor regression [48]. This evidence may imply that the high abundance of TGF-β in the tumor microenvironment represents the most general and primary mechanism for cancer cells to escape the host immune system.

The main strategies for the therapeutic targeting the TGF-β signaling pathway can be exploited by suppressing the production of TGF-β, the activity of TGF-β, the interaction between TGF-β ligand and its receptors, or the kinase activity of TGF-β receptor (Fig. 3). Several types of TGF-β signaling inhibitors are currently under clinical trials in various cancer types, either singly or in combination with other anticancer agents, including immune checkpoint inhibitors (Table 1).

Table 1 . Currently ongoing clinical trials of TGF-β signaling pathway inhibitors for cancer treatment

Drug nameTarget typeTreatmentNCT number
Trabedersen (AP 12009)TGF-β2AS-ODNAdvanced pancreatic, colorectal cancer, melanoma (phase I)NCT00844064(completed)
Fresolimumab (GC1008)TGF-β1 mAbNSCLC (phase I/II)NCT02581787
AVID200TGF-β1/3 trapMalignant solid tumor (phase I)NCT03834662
ABBV-151GARP/TGF-β1 mAbAdvanced or metastatic solid tumor (phase I)NCT03821935
SRK-181TGF-β1mAbAdvanced solid tumor (phase I); alone or in combination with anti-PD-L1NCT04291079
Bintrafusp alfa (M7824)TGF-β trapAdvanced NSCLC (phase III)NCT03631706
Biliary tract cancer (phase II/III)NCT04066491
Thymoma and thymic carcinoma (phase II)NCT04417660
Urothelial carcinoma (phase II)NCT04501094
HPV associated cancers (phase II)NCT03427411
Cervical cancer (phase II)NCT04246489
Triple-negative breast cancer (phase II)NCT04489940
Esophageal squamous cell carcinoma (phase II)NCT04595149
Advanced biliary tract cancer (phase II)NCT03833661
Head and neck squamous cell carcinoma (phase II)NCT04428047
Cervical cancer (phase II)NCT04246489
Advanced colorectal cancers (phase II)NCT04491955
Metastatic colorectal cancer (phase I/II)NCT03436563
Advanced Kaposi sarcoma (phase I/II)NCT04303117
Advanced pancreatic cancer (phase I/II)NCT04327986
Galunisertib (LY2157299)TGF-β RI kinase inhibitorMetastatic prostate cancer (phase II)NCT02452008
Advanced hepatocellular carcinoma (phase II)NCT02178358
Recurrent glioblastoma (phase II)NCT01582269
Advanced hepatocellular carcinoma (phase I)NCT02906397
Triple-negative breast cancer (phase I)NCT02672475
Carcinosarcoma of uterus or ovary (phase I); in combination with Paclitaxel/CarboplatinNCT03206177
Rectal adenocarcinoma (phase I)NCT02688712
Recurrent malignant glioma (phase I)NCT01682187
Vactosertib (TEW-7197)TGF-β RI kinase inhibitorMyeloproliferative neoplasms (phase II)NCT04103645
Metastatic colorectal or gastric cancer (phase I/II); in combination with PembrolizumabNCT03724851
Advanced NSCLC (phase I/II); in combination with DurvalumabNCT03732274
Metastatic gastric cancer (phase I/II); in combination with PaclitaxelNCT03698825
Myelodysplastic syndrome (phase I/II)NCT03074006
Refractory multiple myeloma (phase I); in combination with PomalidomideNCT03143985
Advanced desmoid tumor (phase I); in combination with ImatinibNCT03802084
Urothelial carcinoma (phase I); in combination with DurvalumabNCT04064190(not yet recruiting)
PD-L1 positive NSCLC (phase II); in combination with PembrolizumabNCT04515979(not yet recruiting)
Pancreatic ductal adenocarcinoma (phase I);in combination with nal-IRI+5FU/LVNCT04258072(not yet recruiting)

AS-ODN, antisense oligonucleotides; NCT, National Clinical Trial; NSCLC, non-small cell lung cancer; mAb, monoclonal antibody; GARP, glycoprotein A repetitions predominant; anti-PD-L1, anti-programmed death-ligand 1; HPV, human papillomavirus; TGF-β-RI, TGF-β receptor type I; nal-IRI+5FU/LV, nanoliposomal irinotecan plus 5-fluorouracil/leucovorin. Data from ClinicalTrial.gov (https://www.clinicaltrials.gov/).



Figure 3. Mechanisms and examples of TGF-β signaling inhibitors being explored in clinical trials. mAb, monoclonal antibody; TGF-β RI, TGF-β receptor type I; TGF-β RII, TGF-β receptor type II.

Clinical application of TGF-β signaling inhibitors for cancer therapy

Antisense oligonucleotides can effectively and specifically block the expression and secretion of TGF-β. Trabedersen (AP 12009, OT-101), an antisense oligonucleotide designed to human TGF-β2, has been evaluated in phase I/II clinical trials in patients with various types of advanced solid tumors, including pancreatic and colorectal cancers, glioma, and malignant melanoma [49-51]. Since it is well established that the TGF-β2 isoform plays a pivotal role in the progression of pancreatic cancer and glioma, Trabedersen effectively inhibited proliferation and migration of cancer cells and reversed TGF-β-mediated immune suppression of cancer cells [50]. Although initial clinical trials of Trabedersen demonstrated encouraging results, subsequent trials have been disappointing due to significant side effects and insufficient targeted delivery of antisense oligonucleotide. The TGF-β1-specific antisense oligonucleotide, AP 11014 has been developed for the treatment of non-small cell lung cancer (NSCLC), colorectal cancer, and prostate cancer [52].

Monoclonal antibodies (mAbs) targeting ligands are particularly effective in blocking ligand-receptor binding and subsequent ligand-induced signaling. The human mAbs, Metelimumab (CAT-192), Lerdelimumab (CAT-152), and Fresolimumab (GC1008) have been developed and evaluated in clinical trials. Metelimumab and Lerdelimumab are recombinant humanized immunoglobulin (Ig) G4 mAbs that neutralize TGF-β1 and TGF-β2 isoforms, respectively. Although Metelimumab and Lerdelimumab have not entered clinical trials for the treatment of cancers, both mAbs were awarded orphan drug status to treat scleroderma and the prevention of postoperative scarring after glaucoma surgery [53,54]. Fresolimumab (GC-1008) is a human pan TGF-β neutralizing mAb that was evaluated in clinical studies in patients with advanced solid tumors, including melanoma, renal, and squamous cell carcinoma [55,56]. Fresolimumab showed an acceptable safety profile, and there is evidence for its antitumor activity. Recent positron emission tomography (PET) image analysis in patients with glioblastoma and breast cancer using an 89Zr-labeled Fresolimumab has demonstrated that the inhibition of TGF-β provides survival benefit in the context of radiation treatment [57]. Decoy receptors, such as decorin and bone morphogenetic protein and activin membrane-bound inhibitor (BAMBI) inhibit TGF-β signaling by competing with the TGF-β receptor for ligand binding [14]. AVID200, a protein composed of TGF-β receptor ectodomain fused to the human IgG fragment crystallizable domain, is currently in phase I trials for patients with advanced and metastatic solid tumors (NCT03834662). AVID200 was designed to have the specificity that binds and neutralizes TGF-β1 and TGF-β3 isoforms, which can reverse immunosuppressive effects of TGF-β in the tumor microenvironment as well as resistance to anticancer therapies [58].

Specific inhibition of the activity of TGF-β RI, activin-like kinase 5 (ALK5) by small molecules is an attractive therapeutic strategy for developing anticancer drugs. Although several small molecule inhibitors such as SB-431542, SB-505124 (GlaxoSmithKline; Brentford, UK), and LY36493/LY580276 (Eli Lilly and Company; Indianapolis, IN, USA) have been developed and evaluated in preclinical and clinical trials, many of them exhibit low selectivity and undesired side effects, especially cardiac toxicity [59]. However, ALK5 inhibitors, such as Galunisertib and Vactosertib, are currently undergoing clinical trials in cancer treatment through clinical pharmacokinetic and pharmacodynamic studies to establish a safe and effective therapeutic window.

Galunisertib (LY2157299) is an oral ALK5 inhibitor with an IC50 of 50 nM that specifically inhibits the activation of the downstream molecule, Smad, thereby blocking the TGF-β signaling pathway [60]. The antitumor activity of Galunisertib has been shown in various animal models for breast and colorectal cancers and hepatocellular carcinoma either alone or in combination with other standard anticancer agents [21,61]. Galunisertib is one of the most extensively studied small molecule ALK5 inhibitors in clinical development and shows a safety profile from various clinical trials [62,63]. Two phase Ib/IIa clinical trials were conducted with Galunisertib combined with gemcitabine or with temozolomide-based radiochemotherapy in patients with advanced pancreatic cancer or with malignant glioma, respectively [64,65]. When given together, both treatments had an acceptable safety and tolerability profile, although the clinical benefits are not statistically significant. A recent phase II study has demonstrated that Galunisertib combined with a multikinase inhibitor Sorafenib prolonged the overall survival of over 14 months in patients with advanced hepatocellular carcinoma [66]. Galunisertib is currently being evaluated in a phase I clinical trial in patients with carcinosarcoma of the uterus or ovary combined with Paclitaxel or Carboplatin (NCT03206177). A phase II clinical trial is ongoing with Galunisertib combined with Enzalutamide, a nonsteroidal antiandrogen, in patients with metastatic castration-resistant prostate cancer (NCT02452008).

Vactosertib (TEW-7197) is a novel orally bioavailable inhibitor of ALK5 with an IC50 of 11 nM that was designed to have more potent and specificity in comparison to other small molecule inhibitors [67]. The antitumor activity of Vactosertib has been demonstrated in various types of cancers, including breast and lung cancer, melanoma, and hepatocellular carcinoma [68-70]. Vactosertib prolonged the survival of breast tumor-bearing mice by inhibiting invasion and metastasis and improved the ability to elicit CTL activity [69]. A recent study showed that combination treatment of Vactosertib with nal-IRI + 5-FU/LV, a chemotherapy regimen for advanced pancreatic cancer, significantly improved the overall survival of mice by suppressing the invasion of pancreatic cancer cells [71]. Using RNA-sequencing analysis, this study has demonstrated that Vactosertib significantly induces the CCDC80 gene, which regulates E-cadherin expression, and the ectopic expression of CCDC80 suppresses migration and expression of EMT markers in pancreatic cancer cells. Based on the promising results of antitumor effects in various preclinical models, Vactosertib has been tested first in a human phase I study in patients with advanced solid tumors (NCT02160106) [72,73]. Vactosertib is currently evaluated in phase I/II clinical trials for various cancer types combined with conventional chemotherapeutic agents. Two phase I clinical trials are currently evaluating the safety and efficacy of Vactosertib in combination with Paclitaxel in patients with metastatic gastric cancer (NCT03698825) and in combination with Pomalidomide, a thalidomide analog in patients with relapsed and refractory multiple myeloma (NCT03143985). Vactosertib combined with Imatinib is currently investigated in clinical trials for patients with advanced desmoid tumor (NCT03802084).

Combination therapy of immune checkpoint and TGF-β signaling pathway inhibition

Cancer immunotherapy is currently more focused on targeting immune inhibitory checkpoints, such as programmed death-1 (PD-1) or its main ligand PD-L1, or cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) [74]. Several mAbs that block the immune checkpoint pathway are currently approved in specific clinical indications for immunotherapy. Although a subset of patients achieves durable clinical responses after immunotherapy, the majority of patients show little response or resistance. As described above, the aberrant TGF-β signaling contributes to tumor immune escape and immune checkpoint inhibitor resistance. Therefore, concurrent inhibition of TGF-β and immune checkpoint pathway might be a promising strategy for increasing therapeutic efficacy.

One approach targeting TGF-β signaling is to block the release of active TGF-β from the latent complex or the TGF-β ligand activation. Glycoprotein A repetitions predominant (GARP) is a transmembrane protein that serves as the cell surface docking receptor for latent TGF-β [75]. GARP is abundantly expressed on the surface of activated Tregs and platelets and plays an important role in the bioavailability and activation of TGF-β [76]. ABBV-151 is a mAb that binds to the GARP-TGF-β complex and blocks the release of TGF-β. Preclinical data have shown that targeting both GARP-TGF-β and PD-1 improves antitumor effects compared with anti-PD-1 alone. ABBV-151 was evaluated to determine the safety, efficacy, pharmacokinetics, and pharmacodynamics in patients with locally advanced or metastatic solid tumors as a single agent or combined with an anti-PD-1 mAb (NCT03821935). SRK-181 is a humanized mAb that selectively binds to latent TGF-β1 and inhibits its activation [77]. The combined treatment of SRK-181 with an anti-PD-1 antibody demonstrated significant antitumor response and survival benefit in bladder tumor-bearing mice by increasing intratumoral CD8+ T cells and decreasing immunosuppressive myeloid cells. SRK-181 is currently undergoing clinical trials in patients with locally advanced or metastatic solid tumors as an independent agent and combination with anti-PD-L1 antibody (NCT04291079).

Bintrafusp alfa (M7824) is a novel first-in-class bifunctional fusion protein composed of the extracellular domain of TGF-β RII that functions as a TGF-β sequestering or trap fused to a human IgG1 mAb against PD-L1 [78]. Bintrafusp alfa not only inhibits PD-L1 signaling but also blocks the TGF-β-mediated immune evasion in the tumor microenvironment. Preclinical data have demonstrated that Bintrafusp alfa induces immune cell infiltration and antibody-dependent cell-mediated cytotoxicity for a wide range of human carcinoma cells, including those of lung, urothelial bladder, and breast origin [79-81]. According to this study, TGF-β reduces NK cell activation and NK cell-mediated cytotoxicity of tumor cells. The immunosuppressive activities of TGF-β were reduced by Bintrafusp alfa but not by anti-PD-L1 [78]. Bintrafusp alfa prolonged survival of mice bearing EMT-6 breast tumors and suppressed spontaneous lung metastasis more efficiently than did treatment with either anti-PD-L1 or TGF-β trap alone [79]. Bintrafusp alfa treatment reduced expression of α-smooth muscle actin (α-SMA), a maker of CAFs, which serve as a significant tumor microenvironment component and contribute to drug resistance. Bintrafusp alfa also promoted the antitumor immune activity of tumor-infiltrating lymphocytes, CD8+ T cells, and NK cells by enhancing their cytotoxic activity. Bintrafusp alfa has undergone phase I clinical trials in patients with various types of advanced solid tumors, including NSCLC, biliary tract cancer, and hepatocellular carcinoma [82-84]. Results from these clinical trials showed that the treatment with Bintrafusp alfa had a tolerable safety profile and preliminary encouraging evidence for antitumor activity in patients with advanced NSCLC and hepatocellular carcinoma. Bintrafusp alfa is currently investigated in multiple phase II clinical trials in patients with advanced solid tumors including colorectal, head and neck squamous cell, cervical, triple-negative breast cancer, and NSCLC as a single therapy and also in combination with other standard chemotherapeutic agents. Interestingly, a phase III clinical study has been undertaken to evaluate the efficacy and safety of Bintrafusp alfa compared with Pembrolizumab, an anti-PD-1 mAb, as the first-line treatment for patients with advanced NSCLC that has high PD-L1 expression (NCT03631706).

Ravi et al. [85] reported a similar bifunctional antibody-ligand trap comprising an antibody targeting CTLA-4 or PD-L1 fused to an ectodomain of TGF-β RII. This fusion antibody inhibits tumor growth more efficiently than either anti-CTLA-4 or anti-PD-1 alone and the combination of anti-CTLA-4 and anti-PD-1 in human melanoma and triple-negative breast cancer models, respectively. The anti-CTLA-4-TGF-β RII fusion protein significantly reduced and counteracted tumor-infiltrating Tregs and activating tumor-reactive IFN-γ-expressing CD8+ T cells and memory cells.

Besides mAbs and TGF-β traps, the small molecule inhibitors of TGF-β receptor kinase are also currently tested in clinical trials combined with immune checkpoint inhibitors. The phase I/II study of Galunisertib combined with Nivolumab, an anti-PD-1 antibody, was investigated in patients with refractory NSCLC and hepatocellular carcinoma (NCT02423343), and with Durvalumab, an anti-PD-L1 antibody with metastatic pancreatic cancer (NCT02734160). A phase Ib/IIa trial of the combination of Vactosertib with Durvalumab, an anti-PD-L1 antibody, is being conducted in patients with advanced NSCLC (NCT03732274). A phase II clinical trial of Vactosertib combined with Pembrolizumab to treat PD-L1 positive NSCLC (NCT04515979) and a phase I trial of Vactosertib combined with Durvalumab to treat advanced urothelial carcinoma (NCT04064190) are scheduled to begin shortly to evaluate their therapeutic benefits.

TGF-β is a pleiotropic cytokine that can display either tumor-suppressive or tumor-promoting effects in cancer, depending on the stages of tumor and cellular context. Cancer cells also frequently exhibit aberrant regulation of TGF-β signaling either by upregulated expression of TGF-β or by activation of downstream pathways mediated by crosstalk with other signaling pathways. Increasing evidence demonstrates that the TGF-β pathway primarily contributes to generating a favorable microenvironment for tumor growth, angiogenesis, metastasis, and evasion from immune surveillance throughout various tumor progression steps. Therefore, targeting the tumor microenvironment, including immune checkpoint molecules, has led to a paradigm shift in cancer therapy. Although cancer immunotherapies such as immune checkpoint inhibition have shown remarkable promise in the management of various types of malignancies, their efficacy is still limited by various immune-resistant mechanisms. Recent clinical studies of Bintrafusp alfa and small molecule inhibitor of ALK5 combined with an immune checkpoint inhibitor have demonstrated enhanced antitumor efficacy in advanced solid tumors. These results suggest that TGF-β signaling inhibition overcome therapeutic resistance and ultimately enhances the antitumor immune responses. In conclusion, use of TGF-β signaling pathway inhibitors as a single treatment or combined with chemotherapeutic or immunotherapeutic agents is a promising cancer treatment strategy. The challenge going forward is to identify reliable biomarkers for predicting the therapeutic response and determining the patient selection and optimal timing of treatment.

This work was supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education, Science and Technology (NRF-2016R1D1A1B 03935001).

  1. Moses HL, Branum EL, Proper JA, Robinson RA. Transforming growth factor production by chemically transformed cells. Cancer Res 1981;41:2842-8.
    Pubmed
  2. Roberts AB. The ever-increasing complexity of TGF-beta signaling. Cytokine Growth Factor Rev 2002;13:3-5.
    Pubmed CrossRef
  3. Massagué J, Blain SW, Lo RS. TGFbeta signaling in growth control, cancer, and heritable disorders. Cell 2000;103:295-309.
    Pubmed CrossRef
  4. David CJ, Massagué J. Contextual determinants of TGFβ action in development, immunity and cancer. Nat Rev Mol Cell Biol 2018;19:419-35.
    Pubmed KoreaMed CrossRef
  5. Zhao H, Wei J, Sun J. Roles of TGF-β signaling pathway in tumor microenvirionment and cancer therapy. Int Immunopharmacol 2020;89(Pt B):107101.
    Pubmed CrossRef
  6. Chen XL, Chen ZQ, Zhu SL, Liu TW, Wen Y, Su YS, et al. Prognostic value of transforming growth factor-beta in patients with colorectal cancer who undergo surgery: a meta-analysis. BMC Cancer 2017;17:240.
    Pubmed KoreaMed CrossRef
  7. Marchini S, Fruscio R, Clivio L, Beltrame L, Porcu L, Fuso Nerini I, et al. Resistance to platinum-based chemotherapy is associated with epithelial to mesenchymal transition in epithelial ovarian cancer. Eur J Cancer 2013;49:520-30.
    Pubmed CrossRef
  8. Brower V. Checkpoint blockade immunotherapy for cancer comes of age. J Natl Cancer Inst 2015;107:djv069.
    Pubmed CrossRef
  9. Bai X, Yi M, Jiao Y, Chu Q, Wu K. Blocking TGF-β signaling to enhance the efficacy of immune checkpoint inhibitor. Onco Targets Ther 2019;12:9527-38.
    Pubmed KoreaMed CrossRef
  10. Santarpia M, González-Cao M, Viteri S, Karachaliou N, Altavilla G, Rosell R. Programmed cell death protein-1/programmed cell death ligand-1 pathway inhibition and predictive biomarkers: understanding transforming growth factor-beta role. Transl Lung Cancer Res 2015;4:728-42.
    Pubmed KoreaMed CrossRef
  11. Robertson IB, Horiguchi M, Zilberberg L, Dabovic B, Hadjiolova K, Rifkin DB. Latent TGF-β-binding proteins. Matrix Biol 2015;47:44-53.
    Pubmed KoreaMed CrossRef
  12. Gualandris A, Annes JP, Arese M, Noguera I, Jurukovski V, Rifkin DB. The latent transforming growth factor-beta-binding protein-1 promotes in vitro differentiation of embryonic stem cells into endothelium. Mol Biol Cell 2000;11:4295-308.
    Pubmed KoreaMed CrossRef
  13. Munger JS, Huang X, Kawakatsu H, Griffiths MJ, Dalton SL, Wu J, 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;96:319-28.
    Pubmed CrossRef
  14. Shi Y, Massagué J. Mechanisms of TGF-beta signaling from cell membrane to the nucleus. Cell 2003;113:685-700.
    Pubmed CrossRef
  15. Mu Y, Gudey SK, Landström M. Non-Smad signaling pathways. Cell Tissue Res 2012;347:11-20.
    Pubmed CrossRef
  16. Massagué J. TGFbeta in cancer. Cell 2008;134:215-30.
    Pubmed KoreaMed CrossRef
  17. Seoane J, Gomis RR. TGF-β family signaling in tumor suppression and cancer progression. Cold Spring Harb Perspect Biol 2017;9:a022277.
    Pubmed KoreaMed CrossRef
  18. Vogel V. Unraveling the mechanobiology of extracellular matrix. Annu Rev Physiol 2018;80:353-87.
    Pubmed CrossRef
  19. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011;144:646-74.
    Pubmed CrossRef
  20. Ahmadi A, Najafi M, Farhood B, Mortezaee K. Transforming growth factor-β signaling: tumorigenesis and targeting for cancer therapy. J Cell Physiol 2019;234:12173-87.
    Pubmed CrossRef
  21. Colak S, Ten Dijke P. Targeting TGF-β signaling in cancer. Trends Cancer 2017;3:56-71.
    Pubmed CrossRef
  22. Caja F, Vannucci L. TGFβ: a player on multiple fronts in the tumor microenvironment. J Immunotoxicol 2015;12:300-7.
    Pubmed CrossRef
  23. Fuxe J, Karlsson MC. TGF-β-induced epithelial-mesenchymal transition: a link between cancer and inflammation. Semin Cancer Biol 2012;22:455-61.
    Pubmed CrossRef
  24. Li H, Fan X, Houghton J. Tumor microenvironment: the role of the tumor stroma in cancer. J Cell Biochem 2007;101:805-15.
    Pubmed CrossRef
  25. Pickup M, Novitskiy S, Moses HL. The roles of TGFβ in the tumour microenvironment. Nat Rev Cancer 2013;13:788-99.
    Pubmed KoreaMed CrossRef
  26. Yu H, Shen Y, Hong J, Xia Q, Zhou F, Liu X. The contribution of TGF-β in Epithelial-Mesenchymal Transition (EMT): down-regulation of E-cadherin via snail. Neoplasma 2015;62:1-15.
    Pubmed CrossRef
  27. Dumont N, Bakin AV, Arteaga CL. Autocrine transforming growth factor-beta signaling mediates Smad-independent motility in human cancer cells. J Biol Chem 2003;278:3275-85.
    Pubmed CrossRef
  28. Iwatsuki M, Mimori K, Yokobori T, Ishi H, Beppu T, Nakamori S, et al. Epithelial-mesenchymal transition in cancer development and its clinical significance. Cancer Sci 2010;101:293-9.
    Pubmed CrossRef
  29. Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell 2008;133:704-15.
    Pubmed KoreaMed CrossRef
  30. Akhurst RJ, Derynck R. TGF-beta signaling in cancer--a double-edged sword. Trends Cell Biol 2001;11:S44-51.
    Pubmed CrossRef
  31. ten Dijke P, Arthur HM. Extracellular control of TGFbeta signalling in vascular development and disease. Nat Rev Mol Cell Biol 2007;8:857-69.
    Pubmed CrossRef
  32. Pertovaara L, Kaipainen A, Mustonen T, Orpana A, Ferrara N, Saksela O, et al. Vascular endothelial growth factor is induced in response to transforming growth factor-beta in fibroblastic and epithelial cells. J Biol Chem 1994;269:6271-4.
    Pubmed
  33. Sunderkötter C, Goebeler M, Schulze-Osthoff K, Bhardwaj R, Sorg C. Macrophage-derived angiogenesis factors. Pharmacol Ther 1991;51:195-216.
    Pubmed CrossRef
  34. Safina A, Vandette E, Bakin AV. ALK5 promotes tumor angiogenesis by upregulating matrix metalloproteinase-9 in tumor cells. Oncogene 2007;26:2407-22.
    Pubmed CrossRef
  35. Liao Z, Tan ZW, Zhu P, Tan NS. Cancer-associated fibroblasts in tumor microenvironment - accomplices in tumor malignancy. Cell Immunol 2019;343:103729.
    Pubmed CrossRef
  36. Ziani L, Chouaib S, Thiery J. Alteration of the antitumor immune response by cancer-associated fibroblasts. Front Immunol 2018;9:414.
    Pubmed KoreaMed CrossRef
  37. Fiori ME, Di Franco S, Villanova L, Bianca P, Stassi G, De Maria R. Cancer-associated fibroblasts as abettors of tumor progression at the crossroads of EMT and therapy resistance. Mol Cancer 2019;18:70.
    Pubmed KoreaMed CrossRef
  38. Calon A, Lonardo E, Berenguer-Llergo A, Espinet E, Hernando-Momblona X, Iglesias M, et al. Stromal gene expression defines poor-prognosis subtypes in colorectal cancer. Nat Genet 2015;47:320-9.
    Pubmed CrossRef
  39. Li MO, Wan YY, Sanjabi S, Robertson AK, Flavell RA. Transforming growth factor-beta regulation of immune responses. Annu Rev Immunol 2006;24:99-146.
    Pubmed CrossRef
  40. Diebold RJ, Eis MJ, Yin M, Ormsby I, Boivin GP, Darrow BJ, et al. Early-onset multifocal inflammation in the transforming growth factor beta 1-null mouse is lymphocyte mediated. Proc Natl Acad Sci USA 1995;92:12215-9.
    Pubmed KoreaMed CrossRef
  41. Geiser AG, Letterio JJ, Kulkarni AB, Karlsson S, Roberts AB, Sporn MB. Transforming growth factor beta 1 (TGF-beta 1) controls expression of major histocompatibility genes in the postnatal mouse: aberrant histocompatibility antigen expression in the pathogenesis of the TGF-beta 1 null mouse phenotype. Proc Natl Acad Sci USA 1993;90:9944-8.
    Pubmed KoreaMed CrossRef
  42. Wolfraim LA, Walz TM, James Z, Fernandez T, Letterio JJ. p21Cip1 and p27Kip1 act in synergy to alter the sensitivity of naive T cells to TGF-beta-mediated G1 arrest through modulation of IL-2 responsiveness. J Immunol 2004;173:3093-102.
    Pubmed CrossRef
  43. Laouar Y, Sutterwala FS, Gorelik L, Flavell RA. Transforming growth factor-beta controls T helper type 1 cell development through regulation of natural killer cell interferon-gamma. Nat Immunol 2005;6:600-7.
    Pubmed CrossRef
  44. Hong S, Lim S, Li AG, Lee C, Lee YS, Lee EK, et al. Smad7 binds to the adaptors TAB2 and TAB3 to block recruitment of the kinase TAK1 to the adaptor TRAF2. Nat Immunol 2007;8:504-13.
    Pubmed CrossRef
  45. Fang P, Li X, Dai J, Cole L, Camacho JA, Zhang Y, et al. Immune cell subset differentiation and tissue inflammation. J Hematol Oncol 2018;11:97.
    Pubmed KoreaMed CrossRef
  46. Ostroukhova M, Qi Z, Oriss TB, Dixon-McCarthy B, Ray P, Ray A. Treg-mediated immunosuppression involves activation of the Notch-HES1 axis by membrane-bound TGF-beta. J Clin Invest 2006;116:996-1004.
    Pubmed KoreaMed CrossRef
  47. Tauriello DVF, Palomo-Ponce S, Stork D, Berenguer-Llergo A, Badia-Ramentol J, Iglesias M, et al. TGFβ drives immune evasion in genetically reconstituted colon cancer metastasis. Nature 2018;554:538-43.
    Pubmed CrossRef
  48. Mariathasan S, Turley SJ, Nickles D, Castiglioni A, Yuen K, Wang Y, et al. TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 2018;554:544-8.
    Pubmed KoreaMed CrossRef
  49. Schlingensiepen KH, Jaschinski F, Lang SA, Moser C, Geissler EK, Schlitt HJ, et al. Transforming growth factor-beta 2 gene silencing with trabedersen (AP 12009) in pancreatic cancer. Cancer Sci 2011;102:1193-200.
    Pubmed CrossRef
  50. Bogdahn U, Hau P, Stockhammer G, Venkataramana NK, Mahapatra AK, Suri A, et al. Targeted therapy for high-grade glioma with the TGF-β2 inhibitor trabedersen: results of a randomized and controlled phase IIb study. Neuro Oncol 2011;13:132-42.
    Pubmed KoreaMed CrossRef
  51. Gachpazan M, Kashani H, Hassanian SM, Khazaei M, Khorrami S, Ferns GA, et al. Therapeutic potential of targeting transforming growth factor-beta in colorectal cancer: rational and progress. Curr Pharm Des 2019;25:4085-9.
    Pubmed CrossRef
  52. Akhurst RJ, Hata A. Targeting the TGFβ signalling pathway in disease. Nat Rev Drug Discov 2012;11:790-811.
    Pubmed KoreaMed CrossRef
  53. Nagaraj NS, Datta PK. Targeting the transforming growth factor-beta signaling pathway in human cancer. Expert Opin Investig Drugs 2010;19:77-91.
    Pubmed KoreaMed CrossRef
  54. Cordeiro MF. Technology evaluation: lerdelimumab, Cambridge Antibody Technology. Curr Opin Mol Ther 2003;5:199-203.
    Pubmed
  55. Lacouture ME, Morris JC, Lawrence DP, Tan AR, Olencki TE, Shapiro GI, et al. Cutaneous keratoacanthomas/squamous cell carcinomas associated with neutralization of transforming growth factor β by the monoclonal antibody fresolimumab (GC1008). Cancer Immunol Immunother 2015;64:437-46.
    Pubmed KoreaMed CrossRef
  56. Morris JC, Tan AR, Olencki TE, Shapiro GI, Dezube BJ, Reiss M, 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.
    Pubmed KoreaMed CrossRef
  57. Gonzalez-Junca A, Reiners O, Borrero-Garcia LD, Beckford-Vera D, Lazar AA, Chou W, et al. Positron emission tomography imaging of functional transforming growth factor β (TGFβ) activity and benefit of TGFβ inhibition in irradiated intracranial tumors [published online ahead of print September 29, 2020]. Int J Radiat Oncol Biol Phys :doi: 10.1016/j.ijrobp.2020.09.043.
    Pubmed CrossRef
  58. Joyce CE, Saadatpour A, Ruiz-Gutierrez M, Bolukbasi OV, Jiang L, Thomas DD, et al. TGFβ signaling underlies hematopoietic dysfunction and bone marrow failure in Shwachman-Diamond Syndrome. J Clin Invest 2019;129:3821-6.
    Pubmed KoreaMed CrossRef
  59. Anderton MJ, Mellor HR, Bell A, Sadler C, Pass M, Powell S, et al. Induction of heart valve lesions by small-molecule ALK5 inhibitors. Toxicol Pathol 2011;39:916-24.
    Pubmed CrossRef
  60. Sawyer JS, Anderson BD, Beight DW, Campbell RM, Jones ML, Herron DK, et al. Synthesis and activity of new aryl- and heteroaryl-substituted pyrazole inhibitors of the transforming growth factor-beta type I receptor kinase domain. J Med Chem 2003;46:3953-6.
    Pubmed CrossRef
  61. Herbertz S, Sawyer JS, Stauber AJ, Gueorguieva I, Driscoll KE, Estrem ST, et al. Clinical development of galunisertib (LY2157299 monohydrate), a small molecule inhibitor of transforming growth factor-beta signaling pathway. Drug Des Devel Ther 2015;9:4479-99.
    Pubmed KoreaMed CrossRef
  62. de Gramont A, Faivre S, Raymond E. Novel TGF-β inhibitors ready for prime time in onco-immunology. Oncoimmunology 2016;6:e1257453.
    Pubmed KoreaMed CrossRef
  63. Gueorguieva I, Cleverly AL, Stauber A, Sada Pillay N, Rodon JA, Miles CP, et al. Defining a therapeutic window for the novel TGF-β inhibitor LY2157299 monohydrate based on a pharmacokinetic/pharmacodynamic model. Br J Clin Pharmacol 2014;77:796-807.
    Pubmed KoreaMed CrossRef
  64. Wick A, Desjardins A, Suarez C, Forsyth P, Gueorguieva I, Burkholder T, et al. Phase 1b/2a study of galunisertib, a small molecule inhibitor of transforming growth factor-beta receptor I, in combination with standard temozolomide-based radiochemotherapy in patients with newly diagnosed malignant glioma. Invest New Drugs 2020;38:1570-9.
    Pubmed KoreaMed CrossRef
  65. Ikeda M, Takahashi H, Kondo S, Lahn MMF, Ogasawara K, Benhadji KA, et al. Phase 1b study of galunisertib in combination with gemcitabine in Japanese patients with metastatic or locally advanced pancreatic cancer. Cancer Chemother Pharmacol 2017;79:1169-77.
    Pubmed CrossRef
  66. Kelley RK, Gane E, Assenat E, Siebler J, Galle PR, Merle P, et al. A phase 2 study of galunisertib (TGF-β1 receptor type I inhibitor) and sorafenib in patients with advanced hepatocellular carcinoma. Clin Transl Gastroenterol 2019;10:e00056.
    Pubmed KoreaMed CrossRef
  67. Jin CH, Krishnaiah M, eenu D Sr, Subrahmanyam VB, Rao KS, Lee HJ, et al. Discovery of N-((4-([1,2,4]triazolo[1,5-a]pyridin-6-yl)-5-(6-methylpyridin-2-yl)-1H-imidazol-2-yl)methyl)-2-fluoroaniline (EW-7197): a highly potent, selective, and orally bioavailable inhibitor of TGF-β type I receptor kinase as cancer immunotherapeutic/antifibrotic agent. J Med Chem 2014;57:4213-38.
    Pubmed CrossRef
  68. Park SA, Kim MJ, Park SY, Kim JS, Lim W, Nam JS, et al. TIMP-1 mediates TGF-β-dependent crosstalk between hepatic stellate and cancer cells via FAK signaling. Sci Rep 2015;5:16492.
    Pubmed KoreaMed CrossRef
  69. Son JY, Park SY, Kim SJ, Lee SJ, Park SA, Kim MJ, et al. EW-7197, a novel ALK-5 kinase inhibitor, potently inhibits breast to lung metastasis. Mol Cancer Ther 2014;13:1704-16.
    Pubmed CrossRef
  70. Yoon JH, Jung SM, Park SH, Kato M, Yamashita T, Lee IK, 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;5:1720-39.
    Pubmed KoreaMed CrossRef
  71. Hong E, Park S, Ooshima A, Hong CP, Park J, Heo JS, et al. Inhibition of TGF-β signalling in combination with nal-IRI plus 5-Fluorouracil/Leucovorin suppresses invasion and prolongs survival in pancreatic tumour mouse models. Sci Rep 2020;10:2935.
    Pubmed KoreaMed CrossRef
  72. Jung SY, Yug JS, Clarke JM, Bauer TM, Keedy VL, Hwang S, et al. Population pharmacokinetics of vactosertib, a new TGF-β receptor type Ι inhibitor, in patients with advanced solid tumors. Cancer Chemother Pharmacol 2020;85:173-83.
    Pubmed CrossRef
  73. Jung SY, Hwang S, Clarke JM, Bauer TM, Keedy VL, Lee H, et al. Pharmacokinetic characteristics of vactosertib, a new activin receptor-like kinase 5 inhibitor, in patients with advanced solid tumors in a first-in-human phase 1 study. Invest New Drugs 2020;38:812-20.
    Pubmed CrossRef
  74. Postow MA, Callahan MK, Wolchok JD. Immune checkpoint blockade in cancer therapy. J Clin Oncol 2015;33:1974-82.
    Pubmed KoreaMed CrossRef
  75. Metelli A, Wu BX, Fugle CW, Rachidi S, Sun S, Zhang Y, et al. Surface expression of TGFβ docking receptor GARP promotes oncogenesis and immune tolerance in breast cancer. Cancer Res 2016;76:7106-17.
    Pubmed KoreaMed CrossRef
  76. Stockis J, Colau D, Coulie PG, Lucas S. Membrane protein GARP is a receptor for latent TGF-beta on the surface of activated human Treg. Eur J Immunol 2009;39:3315-22.
    Pubmed CrossRef
  77. Martin CJ, Datta A, Littlefield C, Kalra A, Chapron C, Wawersik S, et al. Selective inhibition of TGFβ1 activation overcomes primary resistance to checkpoint blockade therapy by altering tumor immune landscape. Sci Transl Med 2020;12:eaay8456.
    Pubmed CrossRef
  78. Jochems C, Tritsch SR, Pellom ST, Su Z, Soon-Shiong P, Wong HC, et al. Analyses of functions of an anti-PD-L1/TGFβR2 bispecific fusion protein (M7824). Oncotarget 2017;8:75217-31.
    Pubmed KoreaMed CrossRef
  79. Lan Y, Zhang D, Xu C, Hance KW, Marelli B, Qi J, et al. Enhanced preclinical antitumor activity of M7824, a bifunctional fusion protein simultaneously targeting PD-L1 and TGF-β. Sci Transl Med 2018;10:eaan5488.
    Pubmed CrossRef
  80. David JM, Dominguez C, McCampbell KK, Gulley JL, Schlom J, Palena C. A novel bifunctional anti-PD-L1/TGF-β Trap fusion protein (M7824) efficiently reverts mesenchymalization of human lung cancer cells. Oncoimmunology 2017;6:e1349589.
    Pubmed KoreaMed CrossRef
  81. Grenga I, Donahue RN, Gargulak ML, Lepone LM, Roselli M, Bilusic M, et al. Anti-PD-L1/TGFβR2 (M7824) fusion protein induces immunogenic modulation of human urothelial carcinoma cell lines, rendering them more susceptible to immune-mediated recognition and lysis. Urol Oncol 2018;36:93.e1-11.
    Pubmed KoreaMed CrossRef
  82. Doi T, Fujiwara Y, Koyama T, Ikeda M, Helwig C, Watanabe M, et al. Phase I study of the bifunctional fusion protein bintrafusp alfa in Asian patients with advanced solid tumors, including a hepatocellular carcinoma safety-assessment cohort. Oncologist 2020;25:e1292-302.
    Pubmed KoreaMed CrossRef
  83. Paz-Ares L, Kim TM, Vicente D, Felip E, Lee DH, Lee KH, et al. Bintrafusp alfa, a bifunctional fusion protein targeting TGF-β and PD-L1, in second-line treatment of patients with NSCLC: results from an expansion cohort of a phase 1 trial. J Thorac Oncol 2020;15:1210-22.
    Pubmed CrossRef
  84. Strauss J, Heery CR, Schlom J, Madan RA, Cao L, Kang Z, et al. Phase I trial of M7824 (MSB0011359C), a bifunctional fusion protein targeting PD-L1 and TGFβ, in advanced solid tumors. Clin Cancer Res 2018;24:1287-95.
    Pubmed CrossRef
  85. Ravi R, Noonan KA, Pham V, Bedi R, Zhavoronkov A, Ozerov IV, et al. Bifunctional immune checkpoint-targeted antibody-ligand traps that simultaneously disable TGFβ enhance the efficacy of cancer immunotherapy. Nat Commun 2018;9:741.
    Pubmed KoreaMed CrossRef

Share this article on :

Related articles in JCP

Most KeyWord