Fact-checked by Grok 2 weeks ago

Transforming growth factor

Transforming growth factors (TGFs) are cytokines that include transforming growth factor alpha (TGF-α), a member of the (EGF) family that binds to the EGF receptor to promote , and the (TGF-β) superfamily, which comprises a large group of multifunctional cytokines that regulate essential cellular processes, including , , , , and extracellular matrix production, and is conserved across metazoans with 33 members encoded in the and mouse genomes. Originally discovered in 1978 as a growth factor (SGF) secreted by virus-transformed murine fibroblasts, TGF-β was purified and characterized in the early 1980s from sources such as bovine and human platelets, revealing its dual capacity to promote anchorage-independent growth in cells while inhibiting in transformed ones. The three mammalian isoforms—TGF-β1, TGF-β2, and TGF-β3—share high (over 70% identity) and are synthesized as inactive precursors that require proteolytic to form bioactive disulfide-linked homodimers of approximately 25 . TGF-β signaling is initiated upon binding to heterotetrameric complexes of type I and type II serine/ receptors on the surface, leading to of receptor-activated Smad proteins (R-Smads: Smad2/3 for TGF-β or Smad1/5/8 for related members like BMPs), which complex with Smad4 to translocate to the and modulate target in a context-dependent manner. This pathway, often integrated with non-canonical routes such as MAPK or PI3K/Akt signaling, enables versatile responses that vary by type, developmental stage, and microenvironment, underscoring TGF-β's role as a master regulator of tissue . In health, TGF-β is indispensable for embryonic development—guiding processes like gastrulation, organogenesis, and hematopoiesis—while also promoting wound healing through epithelial-mesenchymal transition (EMT) and immune tolerance by inducing regulatory T cells (Tregs) and suppressing pro-inflammatory responses. Dysregulated signaling contributes to numerous pathologies: it acts as a tumor suppressor in early carcinogenesis by inhibiting cell growth but paradoxically drives metastasis and fibrosis in advanced stages via EMT and extracellular matrix deposition, as seen in conditions like Marfan syndrome, systemic sclerosis, and various cancers. Therapeutic strategies targeting TGF-β, including neutralizing antibodies (e.g., fresolimumab) and small-molecule kinase inhibitors (e.g., galunisertib), are under investigation for fibrotic, inflammatory, and oncologic diseases, highlighting its clinical relevance.

Introduction

Definition and classes

Transforming growth factors (TGFs) are polypeptide growth factors that induce phenotypic changes in cells, originally identified for their capacity to transform normal fibroblasts into a tumorigenic state by promoting anchorage-independent growth. The term "transforming growth factor" was coined in the based on assays demonstrating this transformation of non-transformed cells in both and soft conditions. TGFs comprise two principal classes—TGF-α and TGF-β—that are unrelated structurally or genetically, despite sharing the "transforming" nomenclature, and they mediate their effects through distinct receptor mechanisms. TGF-α consists of a single member that functions as a and binds to the (), a , to elicit its biological responses. In contrast, TGF-β constitutes a superfamily of over 30 structurally related members that assemble into disulfide-linked dimers and signal primarily through heterotetrameric complexes of serine/ receptors, such as type I and type II TGF-β receptors. This dimeric configuration is essential for the bioactivity of TGF-β ligands, distinguishing it fundamentally from the monomeric TGF-α.

Historical background

The discovery of transforming growth factor (TGF) activity dates back to 1978, when Joseph E. De Larco and George J. Todaro identified polypeptides secreted by murine sarcoma virus-transformed fibroblasts that induced a transformed in normal indicator cells, including loss of contact inhibition and anchorage-independent growth in soft agar. These factors, initially termed sarcoma growth factors, represented the first demonstration of autocrine growth stimulation by tumor cells and laid the foundation for distinguishing TGF-α, which mimicked (EGF) effects. Independently in the same year, Robert W. Holley and colleagues reported a growth-inhibitory activity from density-arrested BSC-1 cells that regulated proliferation in a density-dependent manner, later recognized as an early description of TGF-β-like inhibition. Subsequent isolations clarified the distinct classes of TGFs. In 1980, De Larco and Todaro extended their work by isolating TGF polypeptides from both virally and chemically transformed cells using acid-ethanol extraction, separating activities into EGF-competing (TGF-α) and non-competing (TGF-β) fractions that synergistically promoted anchorage-independent growth. By 1981, Anita B. Roberts and colleagues purified a novel TGF-β from non-neoplastic murine tissues, such as kidneys, highlighting its presence in normal cells and its potent anchorage-independent growth promotion when combined with TGF-α or EGF, shifting focus from purely oncogenic roles. TGF-α was further isolated from chemically transformed cells in related studies, confirming its production by transformed lines. Key advancements in the 1980s involved purification and structural characterization. In 1983, Hans Marquardt and colleagues purified TGF-α from the conditioned medium of transformed cells and determined its partial , revealing to EGF and establishing it as a distinct for the EGF receptor. For TGF-β, Rik Derynck and colleagues reported the full cDNA in 1985 from human sources, uncovering its precursor structure and dimeric form, which enabled recombinant production and broader functional studies. These milestones built on the 1986 in or awarded to Stanley Cohen and for discovering EGF and , contextualizing TGFs within the expanding family of polypeptide growth regulators. By the , research had evolved the understanding of TGFs from factors primarily linked to oncogenesis—evident in their overproduction by transformed cells—to multifunctional cytokines involved in diverse processes like immune regulation, , and , as demonstrated in seminal reviews and studies integrating their signaling pathways. This shift was propelled by high-impact work from labs like those of Roberts, Sporn, and Derynck, emphasizing TGF-β's context-dependent effects on and across normal and pathological states.

TGF-α

Molecular structure

Transforming growth factor alpha (TGF-α) is synthesized as a transmembrane precursor protein, known as pro-TGF-α, consisting of approximately 160 amino acid residues in humans. This precursor includes a signal peptide, an extracellular domain, a transmembrane region, and a short cytoplasmic tail; proteolytic cleavage at specific sites releases the mature, soluble form of TGF-α, which comprises 50 amino acids. As a member of the epidermal growth factor (EGF) family, TGF-α shares structural homology with EGF, featuring a conserved EGF-like motif that enables high-affinity binding to the EGF receptor (EGFR). The core of the mature TGF-α structure is defined by an EGF-like domain characterized by six conserved cysteine residues that form three intramolecular bonds. These bridges stabilize a compact fold essential for receptor interaction and biological activity, with the cysteines positioned to create a characteristic "knuckle" motif typical of EGF family ligands. The , which encodes TGF-α, is located on the short arm of human at position 2p13. This single produces primarily one canonical isoform through limited , resulting in minor variants that do not significantly alter the mature protein sequence. Insights from crystallographic studies reveal that mature TGF-α adopts a beta-sheet-rich conformation, featuring a three-stranded antiparallel beta-sheet formed by an N-terminal strand and a central beta-ribbon.00940-6) This structure facilitates tight binding to the extracellular domain of , with a (Kd) of approximately 1 nM, underscoring its potent mitogenic properties.

Biological functions

Transforming growth factor alpha (TGF-α) primarily functions as a ligand for the (), binding to it with high affinity to initiate signaling cascades that promote epithelial , , and , particularly in and mucosal tissues. In of the , TGF-α stimulates and , contributing to epidermal thickening and repair processes. Similarly, in mucosal epithelia such as the and gastric lining, TGF-α enhances cell motility more potently than (EGF), facilitating wound closure through EGFR-mediated recycling and sustained signaling. This receptor activation also influences by modulating maturation in stratified epithelia. TGF-α plays a key role in development and through autocrine and paracrine mechanisms. During pubertal mammogenesis and , TGF-α expression in epithelial and stromal cells drives ductal elongation and alveolar via local signaling loops that amplify activity in adjacent tissues. In , TGF-α is secreted by , macrophages, and platelets at injury sites, promoting epithelial re-epithelialization through autocrine stimulation of and paracrine of in surrounding cells. TGF-α contributes to angiogenesis indirectly by inducing vascular endothelial growth factor (VEGF) expression in endothelial and epithelial cells. Through EGFR signaling, TGF-α activates transcription factors like AP-2 in keratinocytes, leading to upregulated VEGF production that supports vessel formation in epithelial contexts. In vivo assays demonstrate TGF-α's superior potency over EGF in eliciting angiogenic responses, highlighting its role in vascular support for epithelial tissues. Experimental evidence from transgenic models underscores TGF-α's proliferative effects in . Overexpression of TGF-α targeted to the basal results in , , and spontaneous formation, with neonates exhibiting scaly and alopecia due to unchecked . These mice develop epidermal thickening without requiring additional oncogenic mutations, confirming TGF-α's direct role in driving hyperproliferative disorders.

TGF-β

Isoforms and structure

In mammals, three isoforms of transforming growth factor β (TGF-β) exist: TGF-β1, TGF-β2, and TGF-β3, each encoded by distinct located on different —TGFB1 on 19q13.2, TGFB2 on 1q41, and TGFB3 on 14q24.3. These isoforms share high conservation in their regions but exhibit differences in their regulatory elements and expression patterns. Each TGF-β isoform is synthesized as a precursor proprotein of approximately 390–414 , consisting of a , a latency-associated (LAP) domain, and a C-terminal mature domain. The mature TGF-β is a -linked homodimer (with rare heterodimers possible) of about 25 kDa, formed by proteolytic cleavage of the proprotein, where the LAP (roughly 250–300 ) remains noncovalently associated with the mature domain, while intramolecular bonds stabilize the dimeric structure within both LAP and the mature . This latent configuration prevents premature activity until further processing. TGF-β belongs to a larger superfamily of over 30 secreted signaling proteins in humans, including bone morphogenetic proteins (BMPs), activins, growth differentiation factors (GDFs), and nodals, all characterized by a conserved in their mature domains. This motif features nine conserved residues per : eight forming intramolecular bonds that create a compact β-sheet structure with a central , and the ninth enabling an interchain bond to stabilize the homodimeric assembly. Biosynthesis begins with translation of the proprotein in the , where two monomers dimerize via bonds—two in the LAP region and one in the mature domain—forming a pro-TGF-β homodimer. This dimeric proprotein is then secreted extracellularly as a large latent complex (LLC), comprising the mature TGF-β noncovalently bound to LAP and covalently linked via bonds to latent TGF-β-binding protein (LTBP), which aids in matrix association. Unlike the monomeric TGF-α, the dimeric latent form of TGF-β ensures regulated .

Activation and signaling

TGF-β is synthesized as a latent precursor complex consisting of the mature TGF-β dimer non-covalently associated with its propeptide, known as the latency-associated (LAP), which maintains inactivity by preventing receptor . Activation occurs through proteolytic cleavage of LAP, primarily mediated by such as αvβ6 and αvβ8, which exert mechanical force or facilitate enzymatic release, as well as by matrix metalloproteinases (MMPs) like MMP-2 and MMP-9, and , which directly degrade LAP to liberate active TGF-β. Thrombospondin-1 (TSP-1) further facilitates this process by to LAP and promoting conformational changes that expose the active dimer for receptor . Upon activation, mature TGF-β binds to cell surface receptors, forming a heterotetrameric complex composed of two type II receptors (TβRII) and two type I receptors (TβRI, also known as ALK5), both of which are serine/ receptors. binding to TβRII induces its constitutive activity to phosphorylate and recruit TβRI, leading to transphosphorylation of TβRI's glycine-serine (GS) domain and activation of its function within the complex. In the canonical signaling pathway, activated TβRI phosphorylates receptor-regulated Smads (R-Smads), specifically Smad2 and Smad3, at their C-terminal SSXS motifs, enabling them to form heteromeric complexes with the common mediator Smad (Co-Smad4). These complexes translocate to the , where they bind to Smad-binding elements (SBEs) in target promoters, characterized by the palindromic CAGAC motif, to regulate transcription of genes involved in cellular processes. TGF-β signaling also engages non-canonical pathways independent of Smads, involving crosstalk with (MAPK) cascades such as ERK, JNK, and p38, which modulate and stress responses; the PI3K/Akt pathway, which influences survival and metabolism; and Rho GTPases, which regulate cytoskeletal dynamics and cell motility. A simplified representation of the canonical pathway kinetics can be expressed as: \text{TGF-β (Ligand)} + \text{TβRI/TβRII (Receptor)} \to \text{p-Smad2/3} \to \text{[Gene expression](/page/Gene_expression)} This arrow diagram illustrates the sequential activation steps without detailed rate constants.

Physiological roles

Embryonic development

Transforming growth factor β (TGF-β) signaling plays a critical role in by regulating induction through activin-like members of the superfamily, which promote the formation of mesodermal tissues from presumptive in embryos.81582-2) In Xenopus laevis, activin, a TGF-β-related , induces in animal cap assays by activating downstream pathways that specify mesodermal fates, establishing the foundational organization during early embryogenesis.80149-X) This process is conserved across , where graded concentrations of these signals dictate ventral formation, highlighting TGF-β's morphogenetic influence in initiating body axis patterning. Isoform-specific functions of TGF-β further underscore its indispensable contributions to . In contrast, TGF-β2 is essential for cardiac septation, as its absence leads to incomplete atrioventricular canal septation and outflow tract defects, resulting in conotruncal malformations. TGF-β2 also governs eye formation by regulating and development through epithelial-mesenchymal interactions in the optic vesicle. Meanwhile, TGF-β3 drives secondary fusion by inducing medial edge remodeling, ensuring proper craniofacial closure without which cleft ensues. Members of the broader TGF-β superfamily extend these roles in embryonic patterning. Bone morphogenetic proteins (BMPs), such as BMP4, establish dorsoventral polarity by forming a ventral-to-dorsal signaling gradient that specifies tissue fates, with high BMP levels promoting ventral mesoderm and while low levels allow dorsal neural induction. Nodal, another superfamily member, orchestrates left-right asymmetry by asymmetrically expressing in the left , directing organ situs through a feedback loop with inhibitors like Lefty. Knockout studies have revealed the non-redundant necessities of TGF-β2 and TGF-β3, as double-null mice exhibit early embryonic lethality around E15.5 due to severe midline fusion defects resulting in failure of ventral body wall closure, extrathoracic heart position, liver extrusion, and skeletal abnormalities such as lack of distal ribs and sternal primordia. Single knockouts confirm isoform specificity: TGF-β2-null embryos display pulmonary and cardiac anomalies leading to perinatal lethality, while TGF-β3-null mice survive gestation but suffer complete cleft palate penetrance. These findings, derived from targeted disruptions in mice, emphasize TGF-β's coordinated orchestration of embryonic viability and morphogenesis.

Tissue homeostasis and repair

Transforming growth factor-β (TGF-β) plays a crucial role in maintaining in adult organisms by regulating immune responses and preventing excessive cellular . In the , TGF-β suppresses T-cell through Smad-dependent mechanisms that inhibit the expression of cytolytic factors such as perforin and , as well as interferon-γ (IFN-γ). This suppression helps sustain and prevent . Additionally, TGF-β promotes the differentiation and function of regulatory T cells (Tregs) by inducing expression via Smad3 signaling, often in conjunction with interleukin-2 (IL-2), thereby enhancing immunosuppressive activity and contributing to peripheral immune . TGF-β also supports tissue architecture by modulating () production in fibroblasts, which is essential for structural integrity. It upregulates the synthesis of key components, including type I and type III collagens and , through transcriptional activation of their genes, leading to increased matrix deposition and remodeling. This process maintains the mechanical properties of tissues like and connective structures under steady-state conditions, ensuring long-term without pathological accumulation. In , TGF-β orchestrates the resolution of , formation of , and re-epithelialization to restore integrity after . During the inflammatory , it recruits and modulates immune cells to excessive responses, transitioning to fibroblast activation for granulation tissue development, where ECM production supports vascularization and provisional matrix formation. In the re-epithelialization stage, TGF-β isoforms, particularly TGF-β1 and TGF-β3, stimulate keratinocyte and while inhibiting to ensure orderly closure of the bed. Furthermore, TGF-β enforces homeostatic balance in epithelial tissues by inhibiting cell proliferation, thereby preventing hyperplasia in organs such as the skin and intestine. In keratinocytes, it induces cell cycle arrest at the G1 phase via Smad-mediated upregulation of cyclin-dependent kinase inhibitors like p15 and p21, maintaining epidermal thickness and barrier function. Similarly, in intestinal epithelia, TGF-β signaling limits enterocyte turnover and promotes barrier maintenance, counteracting proliferative signals to sustain mucosal homeostasis. These inhibitory effects highlight TGF-β's role as a guardian of adult tissue steadiness.

Pathological roles

Cancer

Transforming growth factor β (TGF-β) exhibits a paradoxical dual role in cancer, functioning as a tumor suppressor in early stages while promoting progression in advanced malignancies. In normal epithelial cells and premalignant lesions, TGF-β inhibits cell proliferation by inducing growth arrest, thereby preventing tumor initiation. This suppressive effect is mediated through canonical Smad signaling, where TGF-β ligands bind to type I and II receptors, leading to phosphorylation of Smad2 and Smad3. These phosphorylated Smads form complexes with Smad4, translocate to the nucleus, and transcriptionally upregulate cyclin-dependent kinase inhibitors such as p15^INK4B and p21^WAF1/CIP1. p15^INK4B sequesters CDK4/6, while p21^WAF1/CIP1 inhibits CDK2-cyclin E/A complexes, enforcing G1-phase arrest and cytostasis. Studies in transgenic models demonstrate that loss of TGF-β responsiveness, such as through Smad4 mutations, accelerates epithelial tumorigenesis, underscoring its role in early suppression. In contrast, during advanced carcinogenesis, TGF-β signaling shifts to favor tumor progression, enabling epithelial-to-mesenchymal transition (EMT), invasion, metastasis, angiogenesis, and immune evasion. EMT is induced via Smad-dependent and non-Smad pathways, including upregulation of transcription factors Twist and Snail, which repress E-cadherin expression and enhance cell motility and invasiveness. This facilitates metastatic dissemination, as evidenced in breast cancer, where TGF-β promotes chemoattraction to bone through factors such as CXCR4 and PTHrP. Angiogenesis is supported by TGF-β-induced vascular endothelial growth factor (VEGF) secretion from tumor and stromal cells, increasing microvascular density and nutrient supply to hypoxic tumors. Additionally, TGF-β suppresses antitumor immunity by inducing apoptosis in T and B lymphocytes and impairing cytotoxic T-cell function, creating an immunosuppressive microenvironment that shields advanced tumors. Transforming growth factor α (TGF-α), a distinct in the TGF superfamily, contributes to oncogenesis primarily through autocrine and paracrine activation of the (EGFR). Overexpression of TGF-α is observed in , where it is elevated in bone metastatic lesions and primary tumors compared to benign tissues, stimulating EGFR on bone marrow mesenchymal stem cells to produce pro-metastatic cytokines like IL-6, VEGF, and TGF-β1. In colon cancer, TGF-α overexpression in tumor cells leads to constitutive EGFR , enhancing , lymphangiogenesis via VEGFC from recruited macrophages, and liver metastasis in orthotopic models. This EGFR hyperactivation drives proliferative and angiogenic signaling, promoting remodeling. Clinically, elevated serum TGF-β1 levels serve as a prognostic for poor outcomes in specific carcinomas. In pancreatic ductal , high TGF-β1 expression correlates with reduced survival and increased . Similarly, in , plasma TGF-β1 levels are markedly higher in patients versus those with alone and associate with shorter survival, enhanced , and tumor vascularity. These correlations highlight TGF-β1 as a potential indicator of aggressive disease progression.

Fibrosis and immune disorders

Transforming growth factor-β (TGF-β), particularly isoform TGF-β1, plays a central role in the of through persistent activation of the Smad signaling pathway, which drives the of fibroblasts into and excessive deposition of (ECM) components such as and . In this process, TGF-β binds to its receptors, leading to phosphorylation of Smad2 and Smad3, which form a complex with Smad4 and translocate to the to upregulate profibrotic genes, while also inhibiting matrix-degrading enzymes through inhibitors of metalloproteinases (TIMPs). This mechanism is evident in chronic fibrotic diseases, where sustained Smad signaling perpetuates myofibroblast activation and ECM accumulation, contrasting with its transient role in normal repair. In , such as (IPF), TGF-β induces (EMT) in alveolar epithelial cells and activates resident fibroblasts, resulting in progressive lung scarring and impaired gas exchange. Similarly, in (CKD), TGF-β promotes renal by enhancing pericyte-to-myofibroblast transition and macrophage infiltration, leading to and tubular atrophy. In liver cirrhosis, TGF-β stimulates hepatic stellate cells to secrete , amplifying and in response to chronic injury from toxins or viruses. Among TGF-β isoforms, TGF-β1 predominates in systemic sclerosis, where elevated levels and in fibroblasts contribute to widespread and through upregulated production and vascular dysfunction. In contrast, TGF-β2 is the primary isoform implicated in ocular fibrosis, driving proliferative vitreoretinopathy and subretinal scarring in conditions like age-related by promoting hyalocyte activation and synthesis in the vitreous and . TGF-β also dysregulates immune responses in various disorders; it promotes the differentiation of T helper 17 (Th17) cells in the presence of interleukin-6 (IL-6), enhancing their production of IL-17 and contributing to autoimmunity, as seen in multiple sclerosis where pathogenic Th17 cells infiltrate the central nervous system and exacerbate inflammation. Conversely, TGF-β inhibits allergic responses by restraining T follicular helper 2 (Tfh2) cell differentiation and reducing IgE production, thereby preventing excessive Th2-driven immunity in conditions like atopic dermatitis. However, in HIV infection, excess TGF-β1 from regulatory T cells suppresses antiviral immunity, promotes lymphoid tissue fibrosis, and accelerates disease progression to AIDS by inhibiting CD4+ T cell function and cytokine responses. Genetic variations in the TGFB1 gene further link TGF-β to fibrotic disorders; polymorphisms such as T869C (resulting in the Leu10Pro variant) are associated with increased risk of diabetic nephropathy by elevating TGF-β1 expression and promoting renal ECM accumulation. Similarly, certain TGFB1 polymorphisms, including those in the promoter region like -509C/T, have been implicated in susceptibility to keloid scarring, where they correlate with dysregulated TGF-β1 levels and excessive dermal fibrosis following injury.

Therapeutic implications

Inhibitors and agonists

Small molecule inhibitors targeting the TGF-β pathway primarily act by blocking the kinase activity of transforming growth factor-β receptor type I (TβRI, also known as ALK5), thereby preventing downstream Smad signaling and cellular responses such as , , and extracellular matrix production. (LY2157299 monohydrate), an orally bioavailable ATP-competitive inhibitor, selectively binds to TβRI with high potency ( ≈ 60 nM), inhibiting its autophosphorylation and the subsequent of Smad2/3, which disrupts TGF-β-induced epithelial-to-mesenchymal transition and tumor invasion in preclinical models. Similarly, SB-431542 is a potent, selective that inhibits TβRI activity ( = 94 nM) as well as related receptors ALK4 and ALK7, blocking Smad2/3 and nuclear translocation to suppress TGF-β-mediated activation and deposition in models. Monoclonal antibodies provide an alternative approach by directly neutralizing TGF-β ligands or sequestering them via receptor traps, offering isoform-specific or pan-TGF-β inhibition with reduced off-target effects on related pathways. Fresolimumab (GC1008), a human IgG4 monoclonal antibody, binds with high affinity to all three active isoforms of TGF-β (TGF-β1, -β2, and -β3), preventing their interaction with TβRII and subsequent receptor complex formation, which inhibits Smad signaling and preclinical fibrotic responses in renal and pulmonary models. Bintrafusp alfa (M7824), a bifunctional fusion protein, combines a human IgG1 anti-PD-L1 antibody with the extracellular domain of TβRII as a "trap" for soluble TGF-β, enabling localized sequestration at PD-L1-expressing tumor sites; this dual mechanism blocks TGF-β-induced immunosuppression and epithelial-to-mesenchymal transition while enhancing immune cell infiltration in syngeneic mouse tumor models. To mitigate fibrosis risks in regenerative contexts, intracellular inhibitors such as TAT-Smad7—a cell-penetrating fusion protein of the inhibitory Smad7 with the HIV Tat peptide—overexpress Smad7 to competitively block TGF-β-induced Smad2/3 activation, reducing collagen production and inflammatory infiltration in radiation-induced fibrosis and oral mucositis models. Agonists of TGF-β signaling are explored primarily for regenerative applications like , where controlled activation promotes tissue repair, though their use is constrained by the risk of excessive . Recombinant TGF-β1, administered topically or locally, enhances formation and re-epithelialization in preclinical dermal wound models by stimulating proliferation and synthesis via Smad-dependent pathways, but chronic exposure elevates risk through sustained activation and matrix accumulation. Pathway-specific agonists within the TGF-β superfamily, such as bone morphogenetic proteins (), target distinct receptors (e.g., BMPR1A/BMPR2) to promote osteogenesis while avoiding canonical TGF-β effects like . Recombinant human (rh), a potent BMP agonist, binds BMP type I and II receptors to activate Smad1/5/8 signaling, inducing mesenchymal stem cell differentiation into osteoblasts and enhancing bone formation in preclinical and models, providing a rationale for its use in orthopedic repair independent of TGF-β inhibition strategies. However, post-approval surveillance has identified risks including ectopic bone formation and potential malignancy.

Clinical applications and recent developments

Pirfenidone, an indirect inhibitor of TGF-β signaling, was approved by the U.S. (FDA) in 2014 for the treatment of (IPF), a condition driven by excessive TGF-β-mediated in the lungs. Clinical trials demonstrated that pirfenidone reduces the decline in forced and prolongs in IPF patients by modulating TGF-β-induced deposition. Similarly, recombinant human bone morphogenetic protein-2 (rhBMP-2), a TGF-β superfamily member, received FDA approval in 2002 as part of the INFUSE Bone Graft system for anterior lumbar interbody fusion in spinal surgeries, promoting formation and fusion success rates comparable to autograft in , though with noted risks of adverse events. In , TGF-β inhibitors have advanced to clinical trials for cancers where the pathway promotes tumor progression and immune evasion. Galunisertib, a small-molecule TGF-β receptor I , was evaluated in a phase II randomized trial for recurrent , combined with , showing median of 1.8 months but no overall survival benefit over lomustine alone. Bintrafusp alfa, a bifunctional targeting and TGF-β, underwent phase III testing in treatment-naive, PD-L1-high advanced non-small cell lung cancer (NSCLC), but failed to demonstrate superior efficacy compared to in 2023 interim analysis, with ongoing evaluations of safety profiles including manageable immune-related adverse events and continued trials in other indications such as and cancers as of 2025. Post-2020 developments highlight innovative applications of TGF-β modulation. In CAR-T , combined /TGF-β blockade has been shown to prevent T cell exhaustion and enhance expansion in solid tumors, as demonstrated in preclinical models and early clinical studies from 2023, potentially improving efficacy against immunosuppressive microenvironments. For , isoform-specific targeting of TGF-β2, which drives fibrosis and elevated , has progressed with preclinical evidence that inhibits TGF-β2-induced extracellular matrix production in human cells, supporting its exploration in antifibrotic therapies as reviewed in 2023. Additionally, artificial intelligence-driven discovery has identified novel inhibitors like the TNIK modulator INS018_055, which disrupts TGF-β/Smad-mediated in preclinical fibrotic models, with phase I trials initiated in 2022 for conditions such as and . As of May 2025, Agomab Therapeutics reported interim phase IIa data for AGMB-129, an oral TGF-β inhibitor, showing promising antifibrotic activity in systemic sclerosis. Despite these advances, clinical translation of TGF-β inhibitors faces challenges, including dose-limiting toxicities such as bleeding due to impaired and vascular integrity, observed in trials of agents like bintrafusp alfa and fresolimumab. Ongoing emphasizes isoform-selective and combination strategies to mitigate these risks while preserving therapeutic benefits.

References

  1. [1]
    TGF-β and the TGF-β Family: Context-Dependent Roles in Cell and ...
    This review introduces the reader to the TGF-β family with its complexity of names and biological activities.
  2. [2]
    TGF-β signaling in health, disease and therapeutics - Nature
    Mar 22, 2024 · The signal transduction of TGF-β can stimulate diverse cellular responses and is particularly critical to embryonic development, wound healing, ...
  3. [3]
    The Discovery and Early Days of TGF-β: A Historical Perspective
    This review summarizes the discovery of TGF-β, the early developments in its molecular and biological characterization with its many biological activities.Discovery Of Tgf-β · Three Tgf-βs · The Tgf-β ``superfamily''
  4. [4]
    Transforming Growth Factor Protein | TGFB Peptide - Prospec Bio
    ... TGF) are not structurally or genetically related. They act through different receptor mechanisms and they do not always induce cellular transformation.
  5. [5]
    Recombinant Human TGF-α - Cell Guidance Systems
    Molecular Weight, 5.6 kDa ; Structure, Monomer ; Source, E. coli ; Accession Number, P01135 ; Purity, ≥95% determined by reducing and non-reducing SDS-PAGE.
  6. [6]
    Biochemistry, Epidermal Growth Factor Receptor - StatPearls - NCBI
    Dec 3, 2023 · The epidermal growth factor receptor (EGFR) family is a subclass of receptor tyrosine kinase (RTK) proteins and consists of 4 members.
  7. [7]
    TGF-β – an excellent servant but a bad master
    Sep 3, 2012 · Transforming Growth Factor-β (TGF-β) is a key player in cell proliferation, differentiation and apoptosis. The importance of this regulation is ...Missing: etymology | Show results with:etymology<|control11|><|separator|>
  8. [8]
    Regulation of TGF-β receptor activity | Cell & Bioscience | Full Text
    Mar 15, 2012 · TGF-β signaling is initiated by the binding of TGF-β to its serine and threonine kinase receptors, the type II (TβRII) and type I (TβRI) ...
  9. [9]
    An engineered transforming growth factor β (TGF-β) monomer ... - NIH
    The TGF-β monomers are described as curled left hands, with the heel formed by a 3½ turn α-helix (α3) and the four fingers formed by the β-strands that extend ...
  10. [10]
  11. [11]
  12. [12]
    Transforming growth factor alpha: expression, regulation ... - PubMed
    TGF alpha is a 6 kDa polypeptide mitogen that interacts with the epidermal growth factor receptor and activates its intrinsic tyrosine kinase.Missing: monomer | Show results with:monomer
  13. [13]
    Human transforming growth factor-alpha: precursor structure and ...
    Transforming growth factor-alpha (TGF-alpha) is secreted by many human tumors and can induce the reversible transformation of nontransformed cell lines.Missing: disulfide bonds crystal EGFR affinity
  14. [14]
    From wavy hair to naked proteins: The role of transforming growth ...
    TGFA protein is synthesized as a 160 amino acid precursor. A 23 amino acid long signal peptide (SP) is followed by a 16 amino acid pro-region (PR). The ...Missing: length | Show results with:length
  15. [15]
    Cloning and Biological Activity of Epigen, a Novel Member of the ...
    Examination of the disulfide bonding arrangement in EGF and TGFα indicated that the six cysteine residues form three pairs of disulfide bonds, with substitution ...
  16. [16]
    Gene ResultTGFA transforming growth factor alpha [ (human)] - NCBI
    Aug 19, 2025 · TGFA is up-regulated in tumor cells of SCC but not BCC of the skin. TGFA is also overexpressed in asymptomatic epidermis adjacent to both SCC ...Missing: chromosome | Show results with:chromosome
  17. [17]
    TGFA - Protransforming growth factor alpha - Homo sapiens (Human)
    Jul 21, 1986 · TGF alpha is a mitogenic polypeptide that is able to bind to the EGF receptor/EGFR and to act synergistically with TGF beta to promote anchorage-independent ...Missing: monomer | Show results with:monomer
  18. [18]
    Low-affinity ligands of the epidermal growth factor receptor are long ...
    Nov 26, 2024 · The high-affinity ligands, with apparent Kd values ranging from 0.1 to 1 nM, are EGF, transforming growth factor α (TGF-α), betacellulin (BTC), ...
  19. [19]
  20. [20]
  21. [21]
  22. [22]
  23. [23]
  24. [24]
  25. [25]
    Targeted overexpression of transforming growth factor alpha in the ...
    Dec 1, 1993 · TGF-alpha transgene were often smaller than normal littermates and had precocious eyelid opening and wrinkled, scaly skin with diffuse alopecia.
  26. [26]
    7040 - Gene ResultTGFB1 transforming growth factor beta 1 [ (human)]
    Sep 27, 2025 · TGFB1 encodes a secreted ligand that regulates cell proliferation, differentiation, and growth. It is often upregulated in tumor cells.
  27. [27]
    7042 - Gene ResultTGFB2 transforming growth factor beta 2 [ (human)]
    Sep 27, 2025 · TGFB2 is located in genes previously associated with chronic obstructive pulmonary disease susceptibility. Data indicate that glioma cell ...
  28. [28]
    TGF-β: from latent to active - ScienceDirect.com
    The isoforms of TGF-β are encoded as large precursor proteins that are 390–412 amino acids [4], [5], [6], [12] in size and each isoform is the product of a ...
  29. [29]
    Regulation of the Bioavailability of TGF-β and TGF-β-Related Proteins
    Within the endoplasmic reticulum, the dimeric pro-TGF-β is linked to a single LTBP by a pair of disulfide bonds between LTBP and LAP (Miyazono et al. 1991; ...Missing: homodimer | Show results with:homodimer
  30. [30]
    Structural Biology and Evolution of the TGF-β Family - PMC
    We review the evolution and structure of members of the transforming growth factor β (TGF-β) family, antagonistic or agonistic modulators, and receptors ...
  31. [31]
    TGF-β Signaling - MDPI
    Dimerization of pro-TGF-β takes place in the ER lumen via three disulfide bonds (black dots), two in the prodomain and one in the mature domain. The dimeric pro ...
  32. [32]
    Transforming growth factor-β receptors: versatile mechanisms of ...
    Feb 13, 2024 · Transforming growth factor-β (TGF-β) signaling is initiated by activation of transmembrane TGF-β receptors (TGFBR), which deploys Smad2/3 ...
  33. [33]
    Thrombospondin-1 regulation of latent TGF-β activation
    Latent TGF-β can be activated through proteolysis (including plasmin and matrix metalloproteinases); binding to integrins βvβ1, βvβ3, βvβ5, βvβ8, or βvβ6 in ...Missing: cleavage | Show results with:cleavage
  34. [34]
    Signaling Receptors for TGF-β Family Members - PubMed Central
    Transforming growth factor β (TGF-β) family members signal via heterotetrameric complexes of type I and type II dual specificity kinase receptors.
  35. [35]
    The roles and regulatory mechanisms of TGF-β and BMP signaling ...
    Jan 24, 2024 · In the TGF-β and BMP signaling pathways, the dimeric ligands bind to heterotetrameric receptors comprising two type I and two type II receptors.
  36. [36]
    Transcriptional control by the TGF-β/Smad signaling system - PMC
    The SBE CAGAC sequence is calculated to be present on average once every 1024 bp in the genome, or about once in the regulatory region of any average size gene.
  37. [37]
    Non-Smad Signaling Pathways of the TGF-β Family - PMC
    Non-Smad pathways include activation of Erk MAPK, JNK, p38 MAPK, IKK, PI3K-Akt, and Rho-like GTPases, in response to TGF-β.
  38. [38]
    A Rate Equation Approach to Elucidate the Kinetics and Robustness ...
    In this article, we apply the rate equation methodology to the Transforming Growth Factor β (TGF-β) pathway in endothelial cells. The members of the TGF-β ...
  39. [39]
    Roles of TGF-β family signaling in stem cell renewal and differentiation
    Dec 30, 2008 · During early stages of embryogenesis, three germ layers, ectoderm, mesoderm, and endoderm, are generated during gastrulation. At the ...
  40. [40]
    TGF-β Family Signaling in Embryonic and Somatic Stem-Cell ...
    Transforming growth factor β1 selectively regulates early murine hematopoietic progenitors and inhibits the growth of IL-3-dependent myeloid leukemia cell lines ...
  41. [41]
    Double-Outlet Right Ventricle and Overriding Tricuspid Valve Reflect ...
    In summary, TGF-β2–knockout mice exhibited a range of cardiovascular anomalies that resulted from failure of normal completion of looping and septation of ...
  42. [42]
    TGFβ2 knockout mice have multiple developmental defects that are ...
    In this paper, we show that TGFβ2 plays essential roles in the development of a wide range of tissues, including craniofacial, axial and appendicular skeleton, ...
  43. [43]
    Transforming growth factor–β3 is required for secondary palate fusion
    Dec 1, 1995 · Mice lacking TGF–β3 exhibit an incompletely penetrant failure of the palatal shelves to fuse leading to cleft palate.
  44. [44]
    BMP Signaling: Lighting up the Way for Embryonic Dorsoventral ...
    It is well-known that the morphogen gradient created by BMP signaling activity is crucial for DV axis patterning across a diverse set of vertebrates.
  45. [45]
    TGF-beta superfamily signaling and left-right asymmetry - PubMed
    During early embryogenesis, the TGF-beta-like protein Nodal (or a Nodal-related ortholog) is expressed by the left lateral plate mesoderm and provides essential ...
  46. [46]
    Tgfbeta2 -/- Tgfbeta3 -/- double knockout mice display ... - PubMed
    Tgfbeta2 (+/-) Tgfbeta3 (-/-) littermates show normal rib and sternum development, normal anterior body wall fusion, and are still alive on E18.5.Missing: β2 β3 1990s
  47. [47]
    TGFβ2 knockout mice have multiple developmental defects that are ...
    Jul 1, 1997 · No significant embryonic lethality was observed. In contrast, the ... B. L.. (. 1990. ). In situ hybridization analysis of TGF beta 3 RNA ...
  48. [48]
    The Role of TGFβ Signaling in Wound Epithelialization - PMC - NIH
    In unwounded skin, TGFβ signaling contributes to tissue homeostasis through regulation of the keratinocyte cell cycle and inhibition of proliferation. During ...
  49. [49]
    Critical Role of Transforming Growth Factor Beta in Different Phases ...
    Epidermal restoration begins with keratinocyte migration and proliferation stimulated by TGF-α, followed by neoepithelium differentiation and basement membrane ...
  50. [50]
    Transforming Growth Factor-β Mediates Intestinal Healing and ...
    In vitro studies suggest that transforming growth factor (TGF)-β has potent effects on gastrointestinal mucosal integrity, wound repair, and neoplasia.Missing: skin | Show results with:skin
  51. [51]
    The Dual Role of TGFβ in Human Cancer: From Tumor Suppression ...
    TGFβ acts as a tumor suppressor in normal cells and early carcinomas, while it promotes tumor metastasis in more advance stages of cancer.
  52. [52]
    TGF-α Overexpression in Breast Cancer Bone Metastasis and ...
    In the process, breast cancer cells and nontumor tissue macrophages produce TGF-α to stimulate BMSCs that express the receptor EGFR, and activated BMSCs ...Missing: colon | Show results with:colon
  53. [53]
    Modification of the Primary Tumor Microenvironment by ...
    These data show that activation of TGFα-EGFR signaling in colon cancer cells creates a microenvironment that is conducive for metastasis.Colon Cancer Cell Lines And... · Elisa For Tgfα Expression · Results
  54. [54]
    Perspectives of TGF-β inhibition in pancreatic and hepatocellular ...
    In the first part of this review, we provide a comprehensive overview of the roles played by the TGF-β pathway and its deregulation in PDAC and HCC development ...
  55. [55]
    Transforming growth factor–β in tissue fibrosis - PubMed Central - NIH
    This review discusses the role of TGF-β in fibrosis, highlighting mechanisms of TGF-β activation and signaling, the cellular targets of TGF-β actions,
  56. [56]
    Targeting fibrosis: mechanisms and clinical trials - Nature
    Jun 30, 2022 · TGF-β promotes fibrosis through diverse mechanisms, including activation of resident fibroblasts, promotion of cell apoptosis, and induction of ...
  57. [57]
    TGF‐β signaling and the fibrotic response - The FASEB Journal - Wiley
    May 1, 2004 · As TGF-β induces fibroblasts to synthesize and contract ECM, this cytokine has long been believed to be a central mediator of the fibrotic ...
  58. [58]
    Transforming growth factor β--at the centre of systemic sclerosis
    Transforming growth factor β (TGF-β) has long been implicated in fibrotic diseases, including the multisystem fibrotic disease systemic sclerosis (SSc).
  59. [59]
    TGF-β Superfamily Signaling in the Eye: Implications for Ocular ...
    Inadequate TGF-β signaling has been associated with defective angiogenesis, vascular barrier function, unfavorable inflammatory responses, and tissue fibrosis.
  60. [60]
    Intricacies of TGF-β signaling in Treg and Th17 cell biology - Nature
    May 23, 2023 · Cutting edge: regulatory T cells induce CD4 + CD25-Foxp3- T cells or are self-induced to become Th17 cells in the absence of exogenous TGF-beta.Missing: paper | Show results with:paper
  61. [61]
    TGFβ prevents IgE-mediated allergic disease by restraining T ...
    Jan 19, 2024 · We demonstrate that LDS mutations lead to reduced TGFβ signaling and elevated total and allergen-specific IgE, despite the presence of wild-type ...
  62. [62]
    The Role of Transforming Growth Factor Beta-1 in the Progression of ...
    Sustained elevations in circulating TGF-β1 are believed to contribute not only to immunosuppression and progression to AIDS, but also to residual ...
  63. [63]
    Transforming growth factor-β1 and diabetic nephropathy - PMC - NIH
    The T869C polymorphism in the human TGF-β1 gene, leading to the L10P variant of the coding protein, is associated with an increased risk of diabetic nephropathy ...Missing: keloid | Show results with:keloid
  64. [64]
    Association of TGFβ1 and SMAD4 variants in the etiology of keloid ...
    Aug 7, 2025 · ... TGF-beta receptor SNPs. Therefore, these TGF-beta receptor polymorphisms are unlikely to be associated with keloid scarring. It is possible ...Missing: TGFB1 | Show results with:TGFB1
  65. [65]
    Clinical development of galunisertib (LY2157299 monohydrate), a ...
    Galunisertib (LY2157299 monohydrate) is an oral small molecule inhibitor of the TGF-β receptor I kinase that specifically downregulates the phosphorylation of ...
  66. [66]
    Targeting TGFβ signal transduction for cancer therapy - Nature
    Jan 8, 2021 · Galunisertib (LY2157299) is an orally available small-molecule inhibitor that selectively binds to TβRI (and weakly to TβRII) and inhibits its ...
  67. [67]
    A Specific Inhibitor of TGF-β Receptor Kinase, SB-431542, as ... - NIH
    SB-431542 efficiently blocks the tumor-promoting effects of TGF-β including cell motility, migration, invasion, and vascular endothelial growth factor (VEGF) ...
  68. [68]
    Phase I Study of GC1008 (Fresolimumab): A Human Anti ...
    Mar 11, 2014 · GC1008 is a human anti-TGFβ monoclonal antibody that neutralizes all isoforms of TGFβ. Here, the safety and activity of GC1008 was evaluated in patients with ...
  69. [69]
    Dual targeting of TGF-β and PD-L1 via a bifunctional anti-PD ... - NIH
    Feb 19, 2020 · Bintrafusp alfa consists of an IgG 1 targeting programmed death ligand 1 (PD-L1) moiety fused via peptide linkers to the extracellular domain of two TGF-β ...
  70. [70]
    Transforming growth factor beta (TGF‐β) isoforms in wound healing ...
    Dec 24, 2015 · TGF-β1 may mediate fibrosis in adults' wounds, while TGF-β3 may promote scarless healing in the fetus and reduced scarring in adults.
  71. [71]
    Smad7 Promotes Healing of Radiotherapy-Induced Oral Mucositis ...
    In contrast, Tat-Smad7 still reduced F4/80+ macrophages in FaDu tumors, possibly by blocking RT-induced TGFβ signaling through Smad2/Smad3 and by direct ...
  72. [72]
    Bone Morphogenetic Protein-2 in Development ... - PubMed Central
    Bone morphogenetic proteins (BMPs) are multi-functional growth factors belonging to the Transforming Growth Factor-Beta (TGF-β) superfamily.
  73. [73]
    [PDF] 022535Orig1s000 - accessdata.fda.gov
    Oct 6, 2014 · Pirfenidone is proposed for the treatment of idiopathic pulmonary fibrosis (IPF), a relentlessly progressive and fatal disease that is at ...
  74. [74]
    Pirfenidone anti-fibrotic effects are partially mediated by the ... - NIH
    Apr 14, 2020 · Pirfenidone is a pleiotropic molecule approved to treat idiopathic pulmonary fibrosis (IPF). Pirfenidone has demonstrated to downregulate ...
  75. [75]
    [PDF] SUMMARY OF SAFETY AND EFFECTIVENESS DATA
    Jan 10, 2002 · The InFUSE™ Bone Graft/LT-CAGE™ Lumbar Tapered Fusion Device is for spinal fusion in patients with degenerative disc disease at one level from ...
  76. [76]
    A Phase II randomized study of galunisertib monotherapy or ...
    Feb 21, 2016 · The secondary efficacy outcome measures included PFS, response rates, and change in tumor size. The median PFS was 1.8 months (95% CI: 1.7–1.8) ...
  77. [77]
    Bintrafusp Alfa Versus Pembrolizumab in Patients With Treatment ...
    First-line treatment with bintrafusp alfa did not exhibit superior efficacy compared with pembrolizumab in patients with PD-L1–high, advanced NSCLC.
  78. [78]
    Combined PD-L1/TGFβ blockade allows expansion and ... - Nature
    Aug 5, 2023 · Knocking out endogenous TGFβ receptor II (TGFBR2) in CAR T cells prevented the exhaustion of CARs. CD8 T cell-intrinsic TGFβ signaling has ...
  79. [79]
    Inhibition of TGF-β2–Induced Trabecular Meshwork Fibrosis ... - NIH
    Nov 17, 2023 · This study investigated the inhibitory effect of PFD on TM fibrosis and evaluated its efficacy in lowering intraocular pressure (IOP).Missing: review | Show results with:review
  80. [80]
    A small-molecule TNIK inhibitor targets fibrosis in preclinical and ...
    Mar 8, 2024 · We identify TRAF2- and NCK-interacting kinase (TNIK) as an anti-fibrotic target using a predictive artificial intelligence (AI) approach.
  81. [81]
    PF-06952229, a selective TGF-β-R1 inhibitor: preclinical ...
    Bleeding (including intratumoral bleeding) has been observed in clinical studies of other TGF-β inhibitors including fresolimumab and bintrafusp alfa.23, 24, 25 ...Original Research · Preclinical Findings · Clinical Findings
  82. [82]
    Model-informed approach for risk management of bleeding toxicities ...
    Sep 6, 2022 · While TGF-β inhibition has been reported to be associated with an increased risk of bleeding, the relative contribution of the three TGF-β ...Missing: review | Show results with:review