Next Article in Journal
Breast Cancer Prediction Using Rotation Forest Algorithm Along with Finding the Influential Causes
Previous Article in Journal
Comparative Analysis of Knee Biomechanics in Total Knee Arthroplasty Patients Across Daily Activities
Previous Article in Special Issue
Validation of Madecassoside Synergy Significantly Enhanced Cryptotanshinone’s Therapeutic Efficacy Against Acne Vulgaris
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Fibroblast Growth Factor-Derived Peptides: Sources, Functions, and Applications

1
School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325000, China
2
Department of Rehabilitation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
3
National Engineering Research Center for Cell Growth Factor Drugs and Protein Biologics, Wenzhou 325000, China
4
National Key Laboratory of Macromolecular Drug Development and Manufacturing, Wenzhou 325000, China
*
Authors to whom correspondence should be addressed.
Bioengineering 2025, 12(10), 1019; https://doi.org/10.3390/bioengineering12101019
Submission received: 19 August 2025 / Revised: 20 September 2025 / Accepted: 23 September 2025 / Published: 25 September 2025

Abstract

Fibroblast growth factors (FGFs) play a crucial role in various biological processes, including tissue development, metabolic regulation, and injury repair. Previous studies have shown that certain peptides can exhibit similar biological functions to FGFs, whether they are fragments extracted from natural FGF molecules or derived peptides designed based on the structural characteristics of FGFs and their receptor molecules. These FGF-derived peptides have shown significant application potential in fields including tissue repair and regeneration, cancer therapy, metabolic regulation, neural recovery, and biological delivery. This article reviews the sources, bioactive functions, molecular mechanisms, and application prospects of FGF-derived peptides, aiming to provide new research ideas for subsequent structural optimization, drug delivery system development, and clinical translation of these peptides.

Graphical Abstract

1. Introduction

As a highly conserved protein family during biological evolution, fibroblast growth factors (FGFs) play a crucial role in embryonic development, tissue regeneration and repair, as well as the maintenance of metabolic homeostasis [1,2,3,4]. Since FGF1 was first isolated from the bovine pituitary in 1973 [5], the number of FGF family members has now expanded to 23, and 22 of them are encoded by the human genome (FGF15 in mice) [6]. Based on their sequence homology, FGFs can be classified into seven subfamilies, including the FGF1 subfamilies, the FGF4 subfamilies, the FGF7 subfamilies, the FGF8 subfamilies, the FGF9 subfamilies, the FGF11 subfamilies, and the FGF19 subfamilies [7]. According to their different modes of action, these seven subfamilies can be further divided into paracrine type (FGF1/4/7/8/9 subfamilies), endocrine type (FGF19 subfamily), and intracellular type (FGF11 subfamily) [8] (Figure 1). The molecular weights of FGF family members range from 17 to 34 kDa, and their sequence homology ranges from 13% to 71% [9]. The core regions of paracrine and intracellular FGFs are both composed of 12 parallel β strands, which fold to form a typical β-trifolium structure. Among them, the 10th and 11th β strands play a crucial role in promoting the binding of FGFs to heparan sulfate (HS). In contrast, because endocrine FGFs lack the 11th β chain, they are unable to form a complete β-trifolium structure; thus, their affinity for HS is significantly reduced [10,11,12,13] (Figure 1). Compared with the highly conservative core region, the N-terminal and C-terminal sequences of FGFs are highly diverse. This characteristic endows the FGF family proteins with rich functional properties [14].
Recent studies have shown that, obtained by extracting the natural sequences of FGFs or rationally designed based on the interaction interface between FGFs and their receptors (fibroblast growth factor receptors, FGFRs), some short peptides consisting of 4 to 28 amino acid residues can mimic or antagonize the biological functions of full-length FGFs. These short peptides are collectively referred to as FGF-derived peptides. Compared to full-length FGFs, FGF-derived peptides possess several advantages, including structural simplicity, low cost, ease of storage, low immunogenicity, high tissue permeability, and facile conjugation with biomaterials [15,16,17]. Currently, both naturally occurring and artificially designed FGF-derived peptides have made significant progress in the fields of tissue repair and regeneration, targeted disease therapy, and the development of novel biological delivery technologies (Table 1). They have gradually overcome, to some extent, the limitations of traditional FGF protein drugs in clinical applications [18]. Therefore, FGF-derived peptides hold great promise as an important direction for the development of next-generation therapeutic molecules.
This article provides a systematic review of the current research progress of FGF-derived peptides. The content covers their molecular sources, functional mechanisms, and applications in the treatment of various diseases. Although there are numerous research reports and discussions on related directions, providing highly valuable content, our review also offers a unique perspective, proposing a potential dual-mode mechanism of FGF’s action in the body. Specifically, we discuss that FGF may not only exert its effect by binding to FGFR in its full-length form to initiate downstream signal cascades but also may be hydrolyzed into biologically active short peptides. These peptides may have stronger tissue permeability and play a role in maintaining physiological homeostasis through finely regulatory effects. To our knowledge, this dual-action hypothesis has not been explicitly emphasized in previous reviews, which is precisely the uniqueness of this study. Additionally, the article also discusses the key challenges and future development directions currently faced by FGF-derived peptides. The aim is to provide new ideas for their further research and clinical translation.

2. Molecular Basis of the Functional Activity of FGF-Derived Peptides

Paracrine and endocrine FGFs exert a variety of biological functions by activating four major tyrosine kinase receptors (FGFR1, FGFR2, FGFR3, and FGFR4), as well as a truncated receptor lacking the intracellular tyrosine kinase domain (FGFRL1), thereby regulating downstream signal transduction [69]. FGFRs belong to the single-pass transmembrane protein family and are composed of an extracellular domain (ECD), a transmembrane domain (TMD), and an intracellular tyrosine kinase domain (TKD) [70]. The extracellular domain consists of three immunoglobulin-like subunits (DI, DII, and DIII), among which DII and DIII play key roles in ligand binding and receptor dimerization [71]. In FGFR1–3, alternative splicing of exons 7–9, which encode the DIII region, occurs during transcription, resulting in the b isoforms (FGFR1b–3b) encoded by exons 7 and 8, and the c isoforms (FGFR1c–3c) encoded by exons 7 and 9 [72] (Figure 2A). This selective splicing alters the amino acid composition at the ligand-binding pocket, thereby affecting ligand specificity. Furthermore, since FGFR isoforms are differentially distributed across various tissues and cell types, alternative splicing serves as an important mechanism by which FGFs achieve tissue-specific regulation [73].
Activation of the FGFR signaling pathway depends on receptor dimerization [74]. Paracrine FGFs induce receptor dimerization by stably interacting with FGFRs through binding to heparan sulfate [75] (Figure 2B). In contrast, the binding of endocrine FGFs to their receptors requires the synergistic action of heparan sulfate and Klotho family proteins (such as α-Klotho or β-Klotho) to enhance their affinity for FGFRs [76,77] (Figure 2C). FGF-FGFR signaling is transduced through the activation of four classical downstream pathways: (1) the MAPK/ERK pathway, which regulates cell proliferation and differentiation [78]; (2) the PI3K/AKT pathway, which promotes cell survival [79]; (3) the PLCγ/DAG pathway, which regulates cell migration [80]; and (4) the JAK/STAT pathway, which is involved in immune responses, tumor invasion, metastasis, and other processes [81], thereby mediating the biological functions of FGFs [82,83,84] (Figure 3, modified from [69,84]).
The concept of FGF-derived peptides originates from researchers’ efforts to explore the regulation of the FGF-FGFR signaling pathway, aiming to achieve therapeutic potential through targeted modulation of FGF biological functions. Specifically, FGF-derived peptides can act as targeted agonists by directly binding to FGFRs, thereby inducing receptor dimerization and activating downstream signaling pathways. At the same time, they may also function as competitive ligands, inhibiting the binding of natural FGFs to FGFRs and thus blocking the activation of the FGF-FGFR signaling pathway. Based on this concept, the development of FGF-derived peptides is primarily achieved through two strategies: The first strategy utilizes molecular docking software to analyze the key amino acid sequences at the FGF-FGFR interaction interface, allowing for the screening of derived peptides from natural FGFs (Table 1). The second strategy is based on the molecular structures of FGFs and FGFRs, employing high-throughput screening methods such as phage display technology to obtain artificial peptide sequences with high affinity [55,65,87]. However, due to the limitations of current phage libraries [88], such derived peptides typically consist of 6 to 12 amino acid residues. It is noteworthy that shorter peptide sequences may contain multiple FGFR binding sites or even interact with other receptors, thereby conferring novel biological functions. Therefore, the functional characteristics of FGF-derived peptides need to be systematically validated using various in vitro and in vivo experimental models to clarify their specificity and potential off-target effects.

3. Activation Functions of FGF-Derived Peptides

FGFs constitute a family of proteins that play critical roles in tissue development, metabolic regulation, and injury repair. Based on the biological functions of FGFs, researchers have designed and developed a series of FGF-derived peptides with similar activities to explore their potential applications in regenerative medicine and drug development.

3.1. FGF-Derived Peptides from Natural Sequences

Osteogenic Differentiation: The peptides derived from FGF2 have shown significant application prospects in bone and cartilage regeneration. Among them, the FGF2-derived peptide FP2 can activate the AKT and ERK signaling pathways by upregulating the phosphorylation levels of FGFR1 and the adaptor protein FRS2α, thereby significantly enhancing the proliferation and colony-forming ability of human umbilical cord Wharton’s jelly mesenchymal stem cells (hWJ-MSCs). At the same time, injecting hWJ-MSCs pre-treated with FP2 into the joint can effectively alleviate the arthritis symptoms of mice in the osteoarthritis model, demonstrating its good therapeutic potential [29]. Moreover, the FGF2-derived peptides F36 and F77 can also promote the differentiation of human bone marrow mesenchymal stem cells (hBM-MSCs) into osteoblasts [24]. It is worth noting that the short peptides F105 and F119 derived from FGF2 not only have strong heparin-binding ability but also can promote the differentiation of hBM-MSCs into osteoblasts in a dose-dependent manner and accelerate matrix mineralization [39].
Muscle Regeneration: Skeletal muscle satellite cells (SCs) are crucial stem cells during the muscle regeneration process and play a key role in the repair and regeneration of muscle fibers [89]. The FGF2-derived peptide-33 and its analogue peptide-33-13 can significantly promote the proliferation of mouse SCs. Pax7 is an important marker gene for maintaining the stemness of SCs, and its expression level directly influences the proliferation and survival of SCs. Studies have shown that peptide-33-13 can also significantly increase the proportion of cells with high expression of Pax7 in proliferating SCs [23]. These results indicate that FGF-derived peptides have significant potential in maintaining the stemness of SCs and promoting muscle regeneration.
Wound Repair: FGF-derived peptides also demonstrate broad application prospects in the field of wound repair. The FGF2-derived peptide FP2 can induce hWJ-MSCs to secrete large amounts of exosomes (FP2-exo). These exosomes can not only significantly promote the migration of human dermal fibroblasts but also effectively inhibit the inflammatory response induced by lipopolysaccharide (LPS) [90]. In addition, the FGF7-derived peptide KGFp can promote the differentiation of mouse BM-MSCs into keratinocytes by activating the ERK1/2, STAT3 and AKT signaling pathways. This peptide has high stability in the skin microenvironment. And, when used in combination with stem cells, it can significantly increase the closure rate of wounds in diabetic mouse models (79.3%) while also enhancing the hardness of the skin tissue [46]. These characteristics make KGFp a potential candidate drug for treating chronic wounds, such as diabetic foot ulcers.
Treatment of Multi-Organ Diseases: The FGF2-derived peptide FGF-P has shown remarkable efficacy in the prevention and treatment of acute radiation syndrome (ARS). FGF-P can significantly reduce sepsis and bleeding symptoms in animals with radiation-induced bone marrow syndrome and can also effectively alleviate their gastrointestinal inflammation and skin syndromes [32,33,34]. To further enhance the activity of this derivative peptide, researchers linked FGF-P with the heparin-binding sequence (RKRLDRIAR [91]) to develop a new active peptide F2A4-K-NS. This peptide exhibits similar angiogenic-promoting ability to FGF2 in both in vivo and in vitro vascularization models [35]. It not only promotes cell migration, proliferation, and gelatinase secretion but also significantly alleviates ulcerative colitis induced by dextran sulfate sodium (DSS) [36]. Furthermore, other researchers also report on peptide amphiphiles that incorporate FGF-P and peptide domains that drive its self-assembly into supramolecular nanoribbons. These FGF2-PA nanoribbons can significantly promote the proliferation and migration of human umbilical vein endothelial cells (HUVECs) by activating the FGFR1 signaling pathway. And it has the same extent as the native FGF-2 protein at certain concentrations [37].
Treatment of Neurodegenerative Diseases: In order to explore the neuroprotective potential of FGF2-derived peptides, researchers used an in vitro oxygen–glucose deprivation (OGD) model and an in vivo rat retinal ischemia–reperfusion (I/R) injury model to evaluate the neuroprotective effect of FGF2-derived peptide FK18 [30]. The results showed that FK18 significantly increased the viability of and attenuated the apoptosis of SH-SY5Y cells. It also markedly alleviated I/R-induced retinal neuronal apoptosis, damage to retinal ganglion cells (RGCs), and morphological and functional damage to the retina. In terms of the molecular mechanism, FK18 could activate the AKT signaling pathway under both physiological and pathological conditions, inhibit the mitochondrial translocation of the proapoptotic protein Bad, increase the expression of Bcl-2/Bax, and reduce the release of cytochrome c in mitochondria. Additionally, there are studies that have reported a series of short peptides, canofins. They are designed based on the interaction structure of FGF2 and FGFR1. They can promote neural synaptic growth by activating FGFRs and exert neuroprotective effects [22]. More importantly, canofins can also inhibit the receptor phosphorylation induced by FGF2, indicating that they have a unique mechanism of action as FGFR agonists.
The F3 region of neural cell adhesion molecule (NCAM) can bind to FGFRs and promote neurite outgrowth [92]. Studies have found that the sixth and seventh β-strands of FGFs are highly homologous to the sequence of this region. Based on this, researchers designed a series of short peptides called hexafins consisting of 16 amino acid residues [20]. Among these, hexafin1/2/3/8/9/10/17 can bind to FGFRs and activate their phosphorylation. Mechanistic studies revealed that basic and hydrophobic amino acids in hexafins play a key role in the binding with FGFRs. Unlike natural FGFs, the binding of hexafins to FGFRs can be inhibited by heparin. Functional experiments further confirmed that hexafin1/2/3/8/9/10/17 can induce neurite growth of cerebellar granule neurons, and the tetramer hexafin 10 has 1250 times higher activity than the monomer. In addition, hexafin1/3/9/10/17 can significantly increase the survival rate of cerebellar granule neurons. These findings suggest that Hexafins are expected to become candidate peptide drugs for regulating neural system functions through the FGFR pathway.
FGF-derived peptides can also be conjugated with other bioactive peptides to exert synergistic pharmacological effects. Researchers integrated the FGF2-derived peptide FGF-P and the laminin signaling peptide IKVAV at the termini of peptide amphiphiles (PAs) with different alkyl tails, constructing a biologically active scaffold. In a severe spinal cord injury mouse model, this scaffold significantly promoted angiogenesis, axonal regeneration, myelin formation, survival of motor neurons, and recovery of neurological function, demonstrating excellent therapeutic potential [38].
Metabolic Regulation: FGF23 is a crucial endocrine factor that plays a significant role in regulating phosphate homeostasis in the body [93]. Studies have shown that a short peptide derived from the C-terminal sequence of FGF23 can significantly reduce the phosphate levels in the serum of FGF23 knockout mice, and their efficacy is comparable to that of direct administration of FGF23. This finding indicates that this short peptide has the potential to be a new candidate drug for treating hyperphosphatemia, such as that occurring in chronic kidney disease. It is worth noting that, since this short peptide lacks the binding domain for FGFRs, it may function through non-classical signaling pathways or directly regulate related metabolic processes within cells [50]. Additionally, researchers have successfully constructed a 26-amino acid residue derived peptide PFNP by fusing the nuclear localization signal peptide of FGF1 with a cell membrane-permeable peptide (sequence: AAVALLPAVLLALLAP [94]). In vitro experiments have shown that this short peptide can stimulate DNA synthesis in cells but does not promote their proliferation [95]. Further mechanistic studies have revealed that PFNP does not bind to FGFRs and does not promote their phosphorylation, suggesting that the function of PFNP in stimulating DNA synthesis may be independent of the Ras signaling pathway [19]. Interestingly, PFNP also has the characteristic of significantly inhibiting animal appetite [96]. These research results all indicate that short peptides derived from endocrine FGFs have broad application prospects in the treatment of metabolic-related diseases such as chronic kidney disease and obesity.

3.2. Artificially Designed FGF-Derived Peptides

Wound Repair: The synthetic short peptide H1, identified through phage display technology, can specifically bind to FGFR2c and exhibit significant biological activity [56]. Studies have demonstrated that H1 promotes angiogenesis and migration of endothelial cells and accelerates the healing of full-thickness skin wounds in rats. Mechanistic investigations revealed that H1 activates the PI3K-AKT and MAPK-ERK1/2 signaling pathways, markedly upregulating the secretion of vascular endothelial growth factor (VEGF), thereby accelerating angiogenesis in the chick embryo chorioallantoic membrane model. Meanwhile, another short peptide, C19jun, which mimics the function of FGF2, is also capable of binding to FGFRs and inducing their dimerization. Further studies revealed that the mechanism of action of this peptide closely resembles that of natural FGF2, and its activation of the FGFR pathway similarly requires the assistance of heparin [52]. The artificially designed peptide FAP1, based on the crystal structure of the FGF2-FGFR1 complex, can promote the proliferation and migration of NIH3T3 cells. In vivo experiments demonstrated that FAP1 binds to FGFR1 to enhance collagen synthesis and promote the migration and proliferation of keratinocytes and fibroblasts, thereby significantly improving wound healing in diabetic mice [51].
Treatment of Neurodegenerative Diseases: The artificially designed short peptide CH02 can bind to and activate the FGFR2 signaling pathway while also exhibiting affinity for other FGFR subtypes [55]. In vitro experiments confirmed that CH02 can maintain the survival of sensory neurons and promote axonal growth. Animal studies showed that CH02 significantly promotes nerve regeneration and the recovery of sensory and motor functions in rats with dorsal root injuries. Mechanistic studies demonstrated that CH02 exerts its neuroprotective and regenerative effects by activating the AKT, ERK, and mTOR signaling pathways downstream of FGFRs. These findings provide important evidence for the potential application of CH02 in the treatment of neurodegenerative diseases.
Metabolic Regulation: FGF21 has attracted considerable attention due to its significant fat-reducing and glucose-lowering effects [97]. FGFR1c, together with β-Klotho, forms the core complex mediating the physiological functions of FGF21 [98]. Based on the structure of this complex, the agonist peptide F91-8A07 was designed, which can undergo self-dimerization through PEGylation. In vitro experiments demonstrated that this dimer exhibits superior lipid-lowering activity in primary human adipocytes compared to native FGF21. Moreover, F91-8A07 significantly upregulates the expression of Egr-1, a target gene of FGFR1c, in mice. These results suggest that this short peptide has potential application value in the treatment of obesity and diabetes [54].

4. Antagonistic Effects of FGF-Derived Peptides

In certain human diseases, abnormally elevated levels of FGFs within tissues may lead to excessive proliferation of target cells and disruption of metabolic homeostasis [99]. To address this, researchers have designed FGF-derived peptides based on the molecular structures and interaction mechanisms of FGFs and their receptors. By inhibiting the binding of ligands to receptors, these peptides offer novel strategies for disease treatment.

4.1. FGF-Derived Peptides from Natural Sequences

Cancer Therapy: The FGF2-derived peptide FREG specifically binds to the FGF2 molecule and reduces its bioactivity by competitively inhibiting the interaction between FGF2 and FGFRs [25]. Recent studies indicate that FREG can significantly suppress the proliferation, invasion of rat aortic smooth muscle cells and pulmonary metastasis of melanoma cells by the platelet-derived growth factor receptor-α (PDGFR-α) signaling pathway. Acute toxicity experiments confirmed the high safety profile of FREG, as even high doses did not induce pathological damage to liver, kidney, or lung tissues [26].
Angiogenesis inhibition is a crucial strategy in cancer therapy. A short peptide, 8b-13, designed based on the gN helix of the FGF8b molecule, can interfere with the binding of FGF8b to FGFRs, thereby inhibiting its bioactivity [47,48,49]. This peptide shows significant therapeutic potential in treating various cancers characterized by aberrant expression of FGF8b, such as prostate and breast cancer.
A cyclic short peptide, P2, designed based on the FGF1 sequence, can both promote the proliferation of BALB/c 3T3 cells and inhibit the interaction between FGF1 and FGFRs through competitive binding. The structural analysis of Nuclear magnetic resonance (NMR) results indicates that the specific amino acid in P2 and its characteristic cyclic conformation are crucial for maintaining its function [21]. A similar situation is that the linear short peptide BGF2, designed based on FGF2 and its cyclic derivative BGF1, which is formed through disulfide bonds, also exhibits different biological activities. The research shows that BGF1 can significantly inhibit the proliferation of HUVECs, 4T1 breast cancer cells, U87 glioblastoma cells, and SKOV3 ovarian cancer cells induced by FGF2, while the linear structure of BGF2 has no such effect [40]. This indicates that the molecular structure of FGF-derived peptides also has a certain degree of influence on the exertion of their biological functions.
The FGF2-derived peptide P5 and its cyclic derivative DcP5 can all inhibit the activation of the FGFR2 signaling pathway mediated by FGF2 to varying degrees [27]. P5 can inhibit the proliferation of DU145 prostate cancer cells in a FGFR2-dependent manner and significantly delay tumor growth in animal models. In contrast, since the cyclic DcP5 has a stronger binding affinity to FGFR2 and is more stable in the body, it can further enhance its anti-cancer activity. Additionally, some researchers have combined P5 with hyaluronic acid (HA) to prepare nanoparticles HA-P5. It can effectively repair acne lesions by inhibiting the FGFR signaling pathway, and its therapeutic effect is superior to the commercially available FGFR inhibitor AZD4547 [28].
Based on the FGF2 sequence and introducing cysteine modification at its C-terminus, the derived peptide bFGFp also has the ability to inhibit the binding of FGF2 to FGFR1. When researchers coupled bFGFp with bovine serum albumin (BSA) and maleimide-polyethylene glycol-phosphatidylethanolamine (Maleimide-PEG-PE), they obtained bFGFp-BSA and bFGFp-liposomes. These can not only efficiently bind to FGF2 but also be specifically taken up by NIH3T3 cells with high expression of FGFRs and do not induce cell proliferation [41]. After modifying it with methoxy polyethylene glycol-distearylphosphatidylethanolamine (mPEG-DSPE), this effect was further enhanced [100].
Active immunization targeting FGFs is an emerging direction in tumor immunotherapy. Researchers coupled 13 derivative peptides based on FGF2 with recombinant hepatitis B core antigen (HBcAg) virus-like particles (VLPs) and immunized mice. The experimental results showed that this method could induce mice to produce high-titer FGF2-specific antibodies and significantly increase the level of interferon-γ (IFN-γ) in the serum. At the same time, the number of immune effector cells (such as CD8+ IFN-γ+ cells, cytotoxic T lymphocytes, and CD4+ IFN-γ+ Th1 cells) in the mice’s bodies increased significantly after immunization. While the number of immunosuppressive cells (such as CD4+ CD25+ FOXP3+ Treg cells and Gr-1+ CD11b+ myeloid-derived suppressor cells) decreased significantly [31]. This immunization strategy provides an important reference for the development of new tumor immunotherapies.
Hair Follicle Repair: The growth process of hair follicles includes the anagen phase, catagen phase and telogen phase. This cyclical change plays an important role in the normal growth of hair [101]. Studies have shown that FGF5 is significantly highly expressed in the later stage of the active growth phase of hair follicles, and it promotes the transformation of hair follicles from the active growth phase to the regressive phase [102]. To inhibit the activity of FGF5, researchers based on the structure of FGF5 developed a derivative peptide P3 consisting of 10 amino acid residues. This peptide can effectively inhibit the proliferation of NIH3T3 and BALB/c 3T3 cells induced by FGF5 and can alleviate the inhibitory effect of FGF5 on the length of hair follicles and the growth of hair cells. These results indicate that P3 has potential application value for anti-hair loss and promoting hair growth [45].

4.2. Artificially Designed FGF-Derived Peptides

Cancer Therapy: The application of phage display technology in cancer-targeted therapy is becoming increasingly widespread. Through this technology, peptide antagonists that can specifically bind to target proteins can be screened out [103]. For example, the artificially synthesized peptide AP8, designed using this approach and capable of specifically binding to FGF1, can block the activation of the ERK1/2 and AKT signaling pathways in breast cancer cells and vascular endothelial cells induced by FGF1. And, by inhibiting the expression of proliferating cell nuclear antigen (PCNA) and Cyclin D1, it can cause the cell cycle to be arrested at the G0/G1 phase [61]. Clinical studies have shown that the level of FGF2 in the serum of breast cancer patients is significantly elevated [104]. The derivative peptide P7, obtained by screening using the phage surface display technology and specifically binding to FGF2, can also block the activation of the ERK and P38 signaling pathways, effectively inhibiting the proliferation of breast cancer cells induced by FGF2 [62]. In addition, P7 can also inhibit the promotion effect of FGF2 on the transformation of epithelial ovarian cancer cells from the G0/G1 phase to the S phase and suppress the expression of Cyclin D1 and the activation of the MAPK and AKT signaling pathways, thereby playing a role in the treatment of ovarian cancer [63]. At the same time, P7 can also act as a chemosensitizer, improving the resistance of colorectal cancer patients to irinotecan hydrochloride (CPT-11) [64]. Moreover, the truncated form of P7, P7Δ, which is an artificially synthesized peptide with high homology to the DIII domain of FGFR1c and FGFR2c, also shows high affinity for FGF2 and can inhibit the proliferation of cells induced by FGF2 and the formation of new blood vessels [65].
Abnormally high expression of FGF3 has been observed in a large number of breast cancer patients [105]. The specific derivative peptide FP16, designed against FGF3, can inhibit the expression of Cyclin D1 and PCNA, causing the cell cycle to be arrested at the G0/G1 phase, thereby inhibiting the proliferation of cancer cells caused by FGF3 overexpression [66]. FGF8b is considered a potential therapeutic target for prostate cancer [106]. The artificially synthesized peptide P12, which is highly homologous to the DIII domain of FGFR3c, can also inhibit the expression of PCNA and Cyclin D1, thereby blocking the activation of ERK1/2 and AKT pathways in prostate cancer cells and vascular endothelial cells [67]. The overexpression of FGF9 is closely related to the occurrence of gastric cancer and bladder cancer [107,108]. The derivative short peptide P4, which is highly homologous to the extracellular domain of FGFR3c, can inhibit the proliferation, migration and invasion of tumor cells induced by FGF9, and can increase the sensitivity of gastric cancer cells to chemotherapy drugs [68]. In addition, the derivative peptide F8 designed for the FGFR1-FGFR1 complex can reduce the proliferation efficiency of BA/F3 cells (highly expressing FGFR1c) induced by FGF1 by more than 40%, showing its potential therapeutic value in FGFR1-overexpressing tumors [53].
Osteogenic Differentiation: Mutations in the FGFR3 gene can lead to various human skeletal dysplasia syndromes, including achondroplasia, hypochondroplasia, and thanatophoric dysplasia [109]. The highly specific derivative peptide P3 developed based on the extracellular domain of FGFR3 can effectively inhibit the abnormal activation of the FGFR3 pathway and its downstream tyrosine kinase activity. Moreover, P3 can also alleviate the bone growth disorders caused by the lethal chondrodysplasia type II disease [57].
Metabolic Regulation: The derivative peptide 23-b6 designed for the FGFR-Klotho complex can act as an antagonist of FGF23. It can alleviate the metabolic disorders caused by abnormal phosphate uptake of FGF23 by inhibiting the ERK pathway and upregulating the expression of type II sodium phosphate cotransporter proteins (NaPi-2a and NaPi-2c) in the opossum kidney cells [59].
Hair Follicle Repair: FGF18 is highly expressed during the hair follicle resting phase and plays an important role in regulating the hair follicle growth cycle [110]. The short peptide GPIGS, obtained through functional screening of bacterial conditioned medium, exhibits the effect of promoting hair regeneration in mice [111]. Subsequent studies revealed that the dipeptide derived from this short peptide has similar receptor specificity to FGF18 and can activate FGFR1c, FGFR3c and FGFR4 while competitively inhibiting the regulatory effect of FGF18. This dipeptide also has great potential in regulating skin physiological functions, promoting hair growth and accelerating wound healing [60].

5. Drug Delivery

To enhance the transfection efficiency of the target gene, researchers constructed a novel gene transfection vector by coupling the nuclear localization signal peptide of FGF3 with the polyamidoamine (PAMAM) dendrimers [42]. The experimental results showed that it was significantly superior to similar polyethyleneimine (PEI) materials in terms of cell transfection efficiency and had extremely low cytotoxicity. Additionally, the derivative peptide K16SP, which fused 16 lysine residues (K16) with the FGF4 signal peptide (SP), could efficiently and non-covalently deliver complete macromolecular proteins (such as immunoglobulin G, β-galactosidase, and green fluorescent protein) into mammalian cells [43]. There are also reports that, when the molecular complex composed of FGF4 signal peptide and polylysine is combined with fluorescently labeled oligonucleotides (ONs), this complex can significantly enhance the cellular uptake of ONs in a dose-dependent and non-endocytic manner [44]. Similarly, when the short peptide targeting FGFR is coupled with the polyethyleneimine–polyethylene glycol (PEI-PEG) complexes, this conjugate also exhibits excellent DNA delivery capabilities [58]. These research findings indicate that the short peptides derived from FGFs have significant potential value in the development of efficient and low-toxicity drug delivery systems.

6. Conclusions and Future Perspectives

FGFs are a class of key signaling molecules that regulate body development, metabolic homeostasis and tissue repair in the human body. However, natural FGFs are limited in clinical application due to their high production cost, poor stability and complex delivery requirements. In contrast, FGF-derived peptides, as functional alternatives to natural FGFs, have shown broad application potential in the fields of tissue repair, regenerative medicine, disease treatment and new technology development at present.
Although these FGF-derived active short peptides currently lack significant sequence homology or structural similarity, they still possess the ability to effectively activate FGFRs and their downstream signaling pathways, ultimately achieving biological functions similar to those of natural FGFs. Interestingly, upon reviewing the literature, we discovered that the functional short peptides derived from specific FGFs (such as FGF2), although containing only 5 to 25 amino acid residues, were able to collectively span almost the entire amino acid sequence of the corresponding full-length protein [20,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41] (Table 1). This observation suggests that FGFs may exert their biological activities not only in their full-length form but also through combinations of shorter functional peptides. Indeed, pharmacokinetic studies have shown that FGFs generally possess a short in vivo half-life and are readily hydrolyzed by various endogenous proteases into peptide fragments of different lengths [112,113,114]. These findings support the hypothesis that FGF-mediated biological functions can be achieved through multiple molecular forms, encompassing both the intact proteins and their constituent peptide assemblies. In addition, these hydrolyzed peptides can reach lesion sites that are difficult for full-length proteins to access, performing regulatory functions (Figure 4). Therefore, systematically mining active short peptides from FGFs and clarifying their mechanisms of action is a research direction worthy of in-depth exploration.
It is worth noting that the biological activity of FGF-derived peptides is usually achieved through the FGF-FGFR pathway, but some of these peptides’ activities do not rely on the activation of FGFRs [19,23]. This multiple regulatory mechanism enables FGF-derived peptides to retain their disease-treatment capabilities while avoiding the toxic side effects caused by the activation of FGFRs [115]. In addition, due to their small molecular weight and low immunogenicity, FGF-derived peptides also show promising application prospects in the treatment of neurological diseases.
The biological activity assessment of FGF-derived peptides is a key part of their preclinical research. Currently, the detection methods mainly rely on various cell lines and animal models (Table 1). However, due to the diversity of the reported model types and drug dosages at this stage, a unified standard has not yet been established. Researchers need to select appropriate models based on specific research goals and mechanisms of action. Compared with full-length FGFs, FGF-derived peptides can partially or fully simulate the biological functions of FGFs by regulating the FGF-FGFR signaling pathway and may also activate other signaling pathways [19,23], showing new biological activities. Therefore, the selection of activity detection methods should comprehensively consider the target of the derived peptide, expected function and research background. It should be noted that the effective working concentration of FGF-derived peptides in in vitro and in vivo experiments is usually 2 to 3 orders of magnitude higher than that of full-length FGFs. This may be related to their smaller molecular weight, simplified structure and changes in the binding sites with FGFRs [46]. Until now, the structural basis underlying the biological activity of these peptides remains unclear. Therefore, it is necessary to investigate whether these peptides may have off-target effects or cause side effects different from those of the natural FGF protein. Meanwhile, optimizing activity in conjunction with standardized risk assessment will support both the development of FGF-derived peptide medicines and their clinical translation.
Although FGF-derived peptides exhibit good stability and ease of storage in vitro, their in vivo application still faces challenges such as rapid degradation and short half-life. Studies have shown that chemical modifications can significantly improve their stability. For example, cyclization of short peptides can enhance molecular rigidity and increase resistance to proteolytic degradation [116]; N-terminal methylation can improve resistance to biodegradation [117]; and C-terminal amidation (introduction of an -NH2 group) can further enhance proteolytic stability [118]. In addition, polyethylene glycol (PEG) modification not only inhibits proteolytic hydrolysis but also improves the biodistribution and solubility of peptides, reduces renal clearance, and prolongs the in vivo half-life [119]. These modification strategies provide technical support for the clinical development of FGF-derived peptides.
The efficient large-scale production of FGF-derived peptides is a prerequisite for their clinical translation. Currently, solid-phase peptide synthesis (SPPS) is the mainstream method for producing short peptides [120]. This technique uses individual amino acids as raw materials and prepares pure peptides through coupling, cleavage, and purification steps, ultimately obtaining stable products through freeze-drying [121]. Therefore, it is suitable for the large-scale production of short FGF-derived peptides. For long FGF-derived peptides, gene engineering methods are usually adopted, with recombinant expression in E. coli or yeast expression systems for preparation [122,123]. In recent years, semi-synthetic strategies combining chemical and biological synthesis have gradually emerged [124,125,126]. This approach can obtain polypeptide precursors with specific tags through microbial expression systems and then prepare target peptides through in vitro enzymatic or chemical modification. This scheme can significantly improve the large-scale production efficiency of derived peptides, thereby accelerating their application process in clinical translation.
Recent studies have demonstrated that the therapeutic efficacy of FGF-derived peptides can be further enhanced through their combination with biomaterials [31,37,38,41]. Therefore, in future development, by referring to the established strategies of other bioactive peptides, efforts should be made to integrate FGF-derived peptides with biological materials. A broad range of biomaterials can serve as potential candidates, including lipid-based nanocarriers, hydrogels, chitosan, PEG, dendrimers, and trehalose-based glycopolymers. These can offer multiple advantages: (1) provide protection for peptides against enzymatic degradation and accidental thiol/disulfide bond exchange reactions; (2) extend the peptide’s half-life and reduce adverse reactions; (3) enhance the physical stability of the peptides, including solubility and anti-aggregation properties, and resist environmental pressures; (4) increase topical bioavailability and therapeutic efficacy [127,128,129,130,131]. However, several issues still need to be further addressed clearly, such as the release kinetics of peptides in materials, biocompatibility and stability in various tissues [132,133,134,135].
The development of artificial intelligence (AI) technologies has opened up new directions for the design of new proteins and short peptides [136,137] and has made it possible to design FGF-derived peptides tailored for various clinical applications. Using AI software such as RFDiffusion (v1.1.0), it is now feasible to move beyond traditional strategies based on natural FGF sequence truncation or phage display and to rationally design high-affinity short peptides targeting specific molecules [138]. Subsequent high-throughput functional screening enables the rapid identification of bioactive peptides with desired functions. For example, TGF-β receptor type II (TGF-βRII)-binding peptides designed using tools such as AlphaFold (v2.3.2) and Rosetta (v1.0.1) have demonstrated sub-nanomolar affinity for the receptor and have been applied in cancer therapy [139]. In the future, research into peptide-based therapeutics, including those based on FGFs and other growth factors, is likely to enter an exciting new stage of innovation and application.
In summary, FGF-derived peptides, as novel and efficient therapeutic molecules designed based on native FGFs, demonstrate broad prospects in regenerative medicine, metabolic regulation, and neuroprotection. They overcome the limitations of traditional protein-based drugs and offer new possibilities for precise and effective disease intervention. In the future, with continued advances in chemical modification techniques, scalable production processes, and AI-assisted design, FGF-derived peptides are expected to accelerate the transition from basic research to clinical application, providing innovative solutions for human health.

Author Contributions

Conceptualization and funding acquisition, X.-K.L. and Z.-J.S.; writing—original draft preparation, C.-K.C. and Z.-Y.S.; writing—review and editing, C.-K.C. and C.-B.C.; investigation, C.-K.C.; supervision oversight, Z.-J.S. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Zhejiang Provincial Top Discipline Program (School of Pharmaceutical Sciences, Wenzhou Medical University), Wenzhou Medical University Talent Research Start-up Project (89225016) and the Horizontal Project of the National Engineering Research Center for Cell Growth Factor Drugs and Protein Biologics, Wenzhou Medical University (Shaanxi Anke Yin Biotechnology Co., Ltd.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study.

Acknowledgments

During the preparation of this work, the authors used ChatGPT 5.0 in order to improve the readability and language of the manuscript. After using this tool, the authors reviewed and edited the content as needed and take full responsibility for the content of the published article.

Conflicts of Interest

The authors declare that they have no conflicts of interest. Author ZhiJian Su has received research grants from Shaanxi Anke Yin Biotechnology Co., Ltd.

Abbreviations

The following abbreviations are used in this manuscript:
AIartificial intelligence
ARSacute radiation syndrome
BSAbovine serum albumin
CAMchorioallantoic membrane
DSPEdistearoyl phosphatidylethanolamine
DSSdextran sulfate sodium
ECDextracellular domain
FGFfibroblast growth factor
FGFRfibroblast growth factor receptor
HBcAghepatitis B core antigen
hBM-MSCshuman bone marrow mesenchymal stem cells
HSheparan sulfate
HUVECshuman umbilical vein endothelial cells
hWJ-MSCshuman Wharton’s jelly mesenchymal stem cells
I/Rischemia–reperfusion
IFN-γinterferon-γ
LPSlipopolysaccharide
NCAMneural cell adhesion molecule
NLSnuclear localization signal
OGDoxygen-glucose deprivation
ONsoligonucleotides
PAMAMpolyamidoamine
PAspeptide amphiphiles
PCNAproliferating cell nuclear antigen
PDGFR-αplatelet-derived growth factor receptor-α
PEphosphatidylethanolamine
PEGpolyethylene glycol
PEIpolyethylenimine
SCsskeletal muscle satellite cells
SPsignal peptide
SPPSsolid-phase peptide synthesis
TGF-βRIItransforming growth factor-β receptor type II
TKDtyrosine kinase domain
TMDtransmembrane domain
VEGFvascular endothelial growth factor
VLPsvirus-like particles

References

  1. Liu, Y.; Liu, Y.; Deng, J.; Li, W.; Nie, X. Fibroblast Growth Factor in Diabetic Foot Ulcer: Progress and Therapeutic Prospects. Front. Endocrinol. 2021, 12, 744868. [Google Scholar] [CrossRef] [PubMed]
  2. Itoh, N.; Ornitz, D.M. Fibroblast Growth Factors: From Molecular Evolution to Roles in Development, Metabolism and Disease. J. Biochem. 2011, 149, 121–130. [Google Scholar] [CrossRef]
  3. Jin, L.; Yang, R.; Geng, L.; Xu, A. Fibroblast Growth Factor–Based Pharmacotherapies for the Treatment of Obesity-Related Metabolic Complications. Annu. Rev. Pharmacol. Toxicol. 2023, 63, 359–382. [Google Scholar] [CrossRef]
  4. Phan, P.; Saikia, B.B.; Sonnaila, S.; Agrawal, S.; Alraawi, Z.; Kumar, T.K.S.; Iyer, S. The Saga of Endocrine FGFs. Cells 2021, 10, 2418. [Google Scholar] [CrossRef] [PubMed]
  5. Armelin, H.A. Pituitary Extracts and Steroid Hormones in the Control of 3T3 Cell Growth. Proc. Natl. Acad. Sci. USA 1973, 70, 2702–2706. [Google Scholar] [CrossRef]
  6. Li, X.; Lu, W.; Kharitonenkov, A.; Luo, Y. Targeting the FGF19–FGFR4 Pathway for Cholestatic, Metabolic, and Cancerous Diseases. J. Intern. Med. 2024, 295, 292–312. [Google Scholar] [CrossRef]
  7. Biadun, M.; Sidor, S.; Kalka, M.; Karelus, R.; Sochacka, M.; Krowarsch, D.; Opalinski, L.; Zakrzewska, M. Production and Purification of Recombinant Long Protein Isoforms of FGF11 Subfamily. J. Biotechnol. 2025, 403, 9–16. [Google Scholar] [CrossRef]
  8. Ornitz, D.M.; Itoh, N. New Developments in the Biology of Fibroblast Growth Factors. WIREs Mech. Dis. 2022, 14, e1549. [Google Scholar] [CrossRef]
  9. Ornitz, D.M.; Itoh, N. Fibroblast Growth Factors. Genome Biol. 2001, 2, reviews3005.1. [Google Scholar] [CrossRef]
  10. Liu, Y.; Ma, J.; Beenken, A.; Srinivasan, L.; Eliseenkova, A.V.; Mohammadi, M. Regulation of Receptor Binding Specificity of FGF9 by an Autoinhibitory Homodimerization. Structure 2017, 25, 1325–1336.e3. [Google Scholar] [CrossRef] [PubMed]
  11. Olsen, S.K.; Garbi, M.; Zampieri, N.; Eliseenkova, A.V.; Ornitz, D.M.; Goldfarb, M.; Mohammadi, M. Fibroblast Growth Factor (FGF) Homologous Factors Share Structural but Not Functional Homology with FGFs. J. Biol. Chem. 2003, 278, 34226–34236. [Google Scholar] [CrossRef]
  12. Goetz, R.; Beenken, A.; Ibrahimi, O.A.; Kalinina, J.; Olsen, S.K.; Eliseenkova, A.V.; Xu, C.; Neubert, T.A.; Zhang, F.; Linhardt, R.J.; et al. Molecular Insights into the Klotho-Dependent, Endocrine Mode of Action of Fibroblast Growth Factor 19 Subfamily Members. Mol. Cell. Biol. 2007, 27, 3417–3428. [Google Scholar] [CrossRef]
  13. Asada, M.; Shinomiya, M.; Suzuki, M.; Honda, E.; Sugimoto, R.; Ikekita, M.; Imamura, T. Glycosaminoglycan Affinity of the Complete Fibroblast Growth Factor Family. Biochim. Biophys. Acta (BBA) Gen. Subj. 2009, 1790, 40–48. [Google Scholar] [CrossRef] [PubMed]
  14. Goetz, R.; Mohammadi, M. Exploring Mechanisms of FGF Signalling through the Lens of Structural Biology. Nat. Rev. Mol. Cell Biol. 2013, 14, 166–180. [Google Scholar] [CrossRef] [PubMed]
  15. Xiao, W.; Jiang, W.; Chen, Z.; Huang, Y.; Mao, J.; Zheng, W.; Hu, Y.; Shi, J. Advance in Peptide-Based Drug Development: Delivery Platforms, Therapeutics and Vaccines. Signal Transduct. Target. Ther. 2025, 10, 74. [Google Scholar] [CrossRef]
  16. Lubell, W.D. Peptide-Based Drug Development. Biomedicines 2022, 10, 2037. [Google Scholar] [CrossRef]
  17. Apostolopoulos, V.; Bojarska, J.; Chai, T.-T.; Elnagdy, S.; Kaczmarek, K.; Matsoukas, J.; New, R.; Parang, K.; Lopez, O.P.; Parhiz, H.; et al. A Global Review on Short Peptides: Frontiers and Perspectives. Molecules 2021, 26, 430. [Google Scholar] [CrossRef]
  18. Ternier, G.; Shahzad, K.; Edirisinghe, O.; Okoto, P.; Alraawi, Z.; Sonnaila, S.; Phan, P.; Adams, P.D.; Thallapuranam, S.K. Fibroblast Growth Factors: Roles and Emerging Therapeutic Applications. Curr. Drug Targets 2025, 26, 551–570. [Google Scholar] [CrossRef] [PubMed]
  19. Komi, A.; Ishisaki, A.; Suzuki, M.; Imamura, T. A Permeable FGF-1 Nuclear Localization Sequence Peptide Induces DNA Synthesis Independently of Ras Activation. Exp. Cell Res. 2003, 283, 91–100. [Google Scholar] [CrossRef]
  20. Li, S.; Christensen, C.; Køhler, L.B.; Kiselyov, V.V.; Berezin, V.; Bock, E. Agonists of Fibroblast Growth Factor Receptor Induce Neurite Outgrowth and Survival of Cerebellar Granule Neurons. Dev. Neurobiol. 2009, 69, 837–854. [Google Scholar] [CrossRef]
  21. Kiyota, S.; Franzoni, L.; Nicastro, G.; Benedetti, A.; Oyama, S.; Viviani, W.; Gambarini, A.G.; Spisni, A.; Miranda, M.T.M. Introduction of a Chemical Constraint in a Short Peptide Derived from Human Acidic Fibroblast Growth Factor Elicits Mitogenic Structural Determinants. J. Med. Chem. 2003, 46, 2325–2333. [Google Scholar] [CrossRef]
  22. Manfè, V.; Kochoyan, A.; Bock, E.; Berezin, V. Peptides Derived from Specific Interaction Sites of the Fibroblast Growth Factor 2—FGF Receptor Complexes Induce Receptor Activation and Signaling. J. Neurochem. 2010, 114, 74–86. [Google Scholar] [CrossRef] [PubMed]
  23. Fujii, I.; Kinoshita, R.; Akiyama, H.; Nakamura, A.; Iwamori, K.; Fukada, S.; Honda, H.; Shimizu, K. Discovery of Fibroblast Growth Factor 2-Derived Peptides for Enhancing Mice Skeletal Muscle Satellite Cell Proliferation. Biotechnol. J. 2024, 19, 2400278. [Google Scholar] [CrossRef]
  24. Lee, J.; Choo, J.; Choi, Y.; Shim, I.; Lee, S.; Seol, Y.; Chung, C.; Park, Y. Effect of Immobilized Cell-binding Peptides on Chitosan Membranes for Osteoblastic Differentiation of Mesenchymal Stem Cells. Biotech. Appl. Biochem. 2009, 52, 69–77. [Google Scholar] [CrossRef] [PubMed]
  25. Facchiano, A.; Russo, K.; Facchiano, A.M.; De Marchis, F.; Facchiano, F.; Ribatti, D.; Aguzzi, M.S.; Capogrossi, M.C. Identification of a Novel Domain of Fibroblast Growth Factor 2 Controlling Its Angiogenic Properties. J. Biol. Chem. 2003, 278, 8751–8760. [Google Scholar] [CrossRef] [PubMed]
  26. Aguzzi, M.S.; Faraone, D.; D’Arcangelo, D.; De Marchis, F.; Toietta, G.; Ribatti, D.; Parazzoli, A.; Colombo, P.; Capogrossi, M.C.; Facchiano, A. The FGF-2-Derived Peptide FREG Inhibits Melanoma Growth in Vitro and in Vivo. Mol. Ther. 2011, 19, 266–273. [Google Scholar] [CrossRef]
  27. Zhang, Y.; Ouyang, M.; Wang, H.; Zhang, B.; Guang, W.; Liu, R.; Li, X.; Shih, T.; Li, Z.; Cao, J.; et al. A Cyclic Peptide Retards the Proliferation of DU145 Prostate Cancer Cells in Vitro and in Vivo through Inhibition of FGFR2. MedComm 2020, 1, 362–375. [Google Scholar] [CrossRef]
  28. Su, Z.; Zhang, Y.; Cao, J.; Sun, Y.; Cai, Y.; Zhang, B.; He, L.; Zhang, Z.; Xie, J.; Meng, Q.; et al. Hyaluronic Acid-FGF2-Derived Peptide Bioconjugates for Suppression of FGFR2 and AR Simultaneously as an Acne Antagonist. J. Nanobiotechnol. 2023, 21, 55. [Google Scholar] [CrossRef]
  29. Lee, S.B.; Abdal Dayem, A.; Kmiecik, S.; Lim, K.M.; Seo, D.S.; Kim, H.-T.; Kumar Biswas, P.; Do, M.; Kim, D.-H.; Cho, S.-G. Efficient Improvement of the Proliferation, Differentiation, and Anti-Arthritic Capacity of Mesenchymal Stem Cells by Simply Culturing on the Immobilized FGF2 Derived Peptide, 44-ERGVVSIKGV-53. J. Adv. Res. 2024, 62, 119–141. [Google Scholar] [CrossRef]
  30. Xiong, S.; Xu, Y.; Ma, M.; Wang, H.; Wei, F.; Gu, Q.; Xu, X. Neuroprotective Effects of a Novel Peptide, FK18, under Oxygen-Glucose Deprivation in SH-SY5Y Cells and Retinal Ischemia in Rats via the Akt Pathway. Neurochem. Int. 2017, 108, 78–90. [Google Scholar] [CrossRef]
  31. Shu, C.; Sun, P.; Xie, H.; Huang, W.; Qi, J.; Ma, Y. Virus-Like Particles Presenting the FGF-2 Protein or Identified Antigenic Peptides Promoted Antitumor Immune Responses in Mice. Int. J. Nanomed. 2020, 15, 1983–1996. [Google Scholar] [CrossRef]
  32. Zhang, L.; Sun, W.; Wang, J.; Zhang, M.; Yang, S.; Tian, Y.; Vidyasagar, S.; Peña, L.A.; Zhang, K.; Cao, Y.; et al. Mitigation Effect of an FGF-2 Peptide on Acute Gastrointestinal Syndrome after High-Dose Ionizing Radiation. Int. J. Radiat. Oncol. Biol. Phys. 2010, 77, 261–268. [Google Scholar] [CrossRef]
  33. Casey-Sawicki, K.; Zhang, M.; Kim, S.; Zhang, A.; Zhang, S.B.; Zhang, Z.; Singh, R.; Yang, S.; Swarts, S.; Vidyasagar, S.; et al. A Basic Fibroblast Growth Factor Analog for Protection and Mitigation against Acute Radiation Syndromes. Health Phys. 2014, 106, 704–712. [Google Scholar] [CrossRef]
  34. Zhang, K.; Tian, Y.; Yin, L.; Zhang, M.; Beck, L.A.; Zhang, B.; Okunieff, P.; Zhang, L.; Vidyasagar, S. Fibroblast Growth Factor-Peptide Improves Barrier Function and Proliferation in Human Keratinocytes After Radiation. Int. J. Radiat. Oncol. Biol. Phys. 2011, 81, 248–254. [Google Scholar] [CrossRef][Green Version]
  35. Lin, X.; Takahashi, K.; Campion, S.; Liu, Y.; Gustavsen, G.; Peña, L.; Zamora, P. Synthetic Peptide F2A4-K-NS Mimics Fibroblast Growth Factor-2 in Vitro and Is Angiogenic in Vivo. Int. J. Mol. Med. 2006, 17, 833–839. [Google Scholar] [CrossRef] [PubMed][Green Version]
  36. Lin, X.; Zamora, P.O.; Takahashi, K.; Lui, Y. Alleviation of Experimental Ulcerative Colitis with the Synthetic Peptide, F2A4-K-NS (Fibratide). Dig. Dis. Sci. 2007, 52, 2054–2062. [Google Scholar] [CrossRef] [PubMed]
  37. Rubert Pérez, C.M.; Álvarez, Z.; Chen, F.; Aytun, T.; Stupp, S.I. Mimicking the Bioactivity of Fibroblast Growth Factor-2 Using Supramolecular Nanoribbons. ACS Biomater. Sci. Eng. 2017, 3, 2166–2175. [Google Scholar] [CrossRef]
  38. Álvarez, Z.; Kolberg-Edelbrock, A.N.; Sasselli, I.R.; Ortega, J.A.; Qiu, R.; Syrgiannis, Z.; Mirau, P.A.; Chen, F.; Chin, S.M.; Weigand, S.; et al. Bioactive Scaffolds with Enhanced Supramolecular Motion Promote Recovery from Spinal Cord Injury. Science 2021, 374, 848–856. [Google Scholar] [CrossRef] [PubMed]
  39. Lee, J.; Choo, J.; Choi, Y.; Lee, K.; Min, D.; Pi, S.; Seol, Y.; Lee, S.; Jo, I.; Chung, C.; et al. Characterization of the Surface Immobilized Synthetic Heparin Binding Domain Derived from Human Fibroblast Growth Factor-2 and Its Effect on Osteoblast Differentiation. J. Biomed. Mater. Res. 2007, 83, 970–979. [Google Scholar] [CrossRef]
  40. Allahmoradi, H.; Asghari, S.M.; Ahmadi, A.; Assareh, E.; Nazari, M. Anti-Tumor and Anti-Metastatic Activity of the FGF2 118–126 Fragment Dependent on the Loop Structure. Biochem. J. 2022, 479, 1285–1302. [Google Scholar] [CrossRef]
  41. Terada, T.; Mizobata, M.; Kawakami, S.; Yabe, Y.; Yamashita, F.; Hashida, M. Basic Fibroblast Growth Factor-Binding Peptide as a Novel Targeting Ligand of Drug Carrier to Tumor Cells. J. Drug Target. 2006, 14, 536–545. [Google Scholar] [CrossRef]
  42. Lee, J.; Jung, J.; Kim, Y.-J.; Lee, E.; Choi, J.S. Gene Delivery of PAMAM Dendrimer Conjugated with the Nuclear Localization Signal Peptide Originated from Fibroblast Growth Factor 3. Int. J. Pharm. 2014, 459, 10–18. [Google Scholar] [CrossRef]
  43. Mahlum, E.; Mandal, D.; Halder, C.; Maran, A.; Yaszemski, M.J.; Jenkins, R.B.; Bolander, M.E.; Sarkar, G. Engineering a Noncarrier to a Highly Efficient Carrier Peptide for Noncovalently Delivering Biologically Active Proteins into Human Cells. Anal. Biochem. 2007, 365, 215–221. [Google Scholar] [CrossRef]
  44. Dokka, S.; Toledo-Velasquez, D.; Shi, X.; Wang, L.; Rojanasakul, Y. Cellular Delivery of Oligonucleotides by Synthetic Import Peptide Carrier. Pharm. Res. 1997, 14, 1759–1764. [Google Scholar] [CrossRef]
  45. Ito, C.; Saitoh, Y.; Fujita, Y.; Yamazaki, Y.; Imamura, T.; Oka, S.; Suzuki, S. Decapeptide with Fibroblast Growth Factor (FGF)-5 Partial Sequence Inhibits Hair Growth Suppressing Activity of FGF-5. J. Cell. Physiol. 2003, 197, 272–283. [Google Scholar] [CrossRef] [PubMed]
  46. Choudhury, S.; Madhu Krishna, M.; Sen, D.; Ghosh, S.; Basak, P.; Das, A. 3D Porous Polymer Scaffold-Conjugated KGF-Mimetic Peptide Promotes Functional Skin Regeneration in Chronic Diabetic Wounds. ACS Appl. Mater. Interfaces 2024, 16, 37418–37434. [Google Scholar] [CrossRef] [PubMed]
  47. Li, T.; Luo, W.; He, D.; Wang, R.; Huang, Y.; Zeng, X.; Wang, W.; Chen, X.; Gao, S.; Yu, Y.; et al. A Short Peptide Derived from the gN Helix Domain of FGF8b Suppresses the Growth of Human Prostate Cancer Cells. Cancer Lett. 2013, 339, 226–236. [Google Scholar] [CrossRef]
  48. Lin, X.; Song, L.; He, D.; Zeng, X.; Wu, J.; Luo, W.; Yang, Q.; Wang, J.; Wang, T.; Cai, J.; et al. An FGF8b-Mimicking Peptide with Potent Antiangiogenic Activity. Mol. Med. Rep. 2017, 16, 894–900. [Google Scholar] [CrossRef]
  49. Liu, H.; Lin, X.; Huang, T.; Song, L.; Zhu, C.; Ma, H.; Long, T.; Zeng, H.; Li, R.; Wang, H.; et al. A Short Peptide Reverses the Aggressive Phenotype of Prostate Cancer Cells. Eur. J. Pharmacol. 2018, 838, 129–137. [Google Scholar] [CrossRef]
  50. Berndt, T.J.; Craig, T.A.; McCormick, D.J.; Lanske, B.; Sitara, D.; Razzaque, M.S.; Pragnell, M.; Bowe, A.E.; O’Brien, S.P.; Schiavi, S.C.; et al. Biological Activity of FGF-23 Fragments. Pflug. Arch. Eur. J. Physiol. 2007, 454, 615–623. [Google Scholar] [CrossRef]
  51. Farooq, M.; Hwang, M.; Khan, A.W.; Batool, M.; Ahmad, B.; Kim, W.; Kim, M.S.; Choi, S. Identification of a Novel Fibroblast Growth Factor Receptor-Agonistic Peptide and Its Effect on Diabetic Wound Healing. Life Sci. 2025, 364, 123432. [Google Scholar] [CrossRef]
  52. Ballinger, M.D.; Shyamala, V.; Forrest, L.D.; Deuter-Reinhard, M.; Doyle, L.V.; Wang, J.; Panganiban-Lustan, L.; Stratton, J.R.; Apell, G.; Winter, J.A.; et al. Semirational Design of a Potent, Artificial Agonist of Fibroblast Growth Factor Receptors. Nat. Biotechnol. 1999, 17, 1199–1204. [Google Scholar] [CrossRef] [PubMed]
  53. Lipok, M.; Szlachcic, A.; Kindela, K.; Czyrek, A.; Otlewski, J. Identification of a Peptide Antagonist of the FGF 1– FGFR 1 Signaling Axis by Phage Display Selection. FEBS Open Bio 2019, 9, 914–924. [Google Scholar] [CrossRef] [PubMed]
  54. Sakamoto, K.; Kawata, Y.; Masuda, Y.; Umemoto, T.; Ito, T.; Asami, T.; Takekawa, S.; Ohtaki, T.; Inooka, H. Discovery of an Artificial Peptide Agonist to the Fibroblast Growth Factor Receptor 1c/βKlotho Complex from Random Peptide T7 Phage Display. Biochem. Biophys. Res. Commun. 2016, 480, 55–60. [Google Scholar] [CrossRef] [PubMed]
  55. Zhao, Y.; Wang, Q.; Xie, C.; Cai, Y.; Chen, X.; Hou, Y.; He, L.; Li, J.; Yao, M.; Chen, S.; et al. Peptide Ligands Targeting FGF Receptors Promote Recovery from Dorsal Root Crush Injury via AKT/mTOR Signaling. Theranostics 2021, 11, 10125–10147. [Google Scholar] [CrossRef]
  56. Zhao, Y.; Wang, Q.; Jin, Y.; Li, Y.; Nie, C.; Huang, P.; Li, Z.; Zhang, B.; Su, Z.; Hong, A.; et al. Discovery and Characterization of a High-Affinity Small Peptide Ligand, H1, Targeting FGFR2IIIc for Skin Wound Healing. Cell Physiol. Biochem. 2018, 49, 1074–1089. [Google Scholar] [CrossRef]
  57. Jin, M.; Yu, Y.; Qi, H.; Xie, Y.; Su, N.; Wang, X.; Tan, Q.; Luo, F.; Zhu, Y.; Wang, Q.; et al. A Novel FGFR3-Binding Peptide Inhibits FGFR3 Signaling and Reverses the Lethal Phenotype of Mice Mimicking Human Thanatophoric Dysplasia. Hum. Mol. Genet. 2012, 21, 5443–5455. [Google Scholar] [CrossRef]
  58. Rao, G.A.; Tsai, R.; Roura, D.; Hughes, J.A. Evaluation of the Transfection Property of a Peptide Ligand for the Fibroblast Growth Factor Receptor as Part of PEGylated Polyethylenimine Polyplex. J. Drug Target. 2008, 16, 79–89. [Google Scholar] [CrossRef]
  59. Huang, T.; Lin, X.; Li, Q.; Luo, W.; Song, L.; Tan, X.; Wang, W.; Li, X.; Wu, X. Selection of a Novel FGF23-Binding Peptide Antagonizing the Inhibitory Effect of FGF23 on Phosphate Uptake. Appl. Microbiol. Biotechnol. 2015, 99, 3169–3177. [Google Scholar] [CrossRef]
  60. Yamada, R.; Fukumoto, R.; Noyama, C.; Fujisawa, A.; Oka, S.; Imamura, T. An Epidermis-permeable Dipeptide Is a Potential Cosmetic Ingredient with Partial Agonist/Antagonist Activity toward Fibroblast Growth Factor Receptors. J. Cosmet. Dermatol. 2020, 19, 477–484. [Google Scholar] [CrossRef]
  61. Dai, X.; Cai, C.; Xiao, F.; Xiong, Y.; Huang, Y.; Zhang, Q.; Xiang, Q.; Lou, G.; Lian, M.; Su, Z.; et al. Identification of a Novel aFGF-Binding Peptide with Anti-Tumor Effect on Breast Cancer from Phage Display Library. Biochem. Biophys. Res. Commun. 2014, 445, 795–801. [Google Scholar] [CrossRef]
  62. Wu, X. A Novel bFGF Antagonist Peptide Inhibits Breast Cancer Cell Growth. Mol. Med. Report. 2012, 6, 210–214. [Google Scholar] [CrossRef][Green Version]
  63. Huang, Y.; Wang, T.; Lin, X.; Wang, H.; Li, R.; Zeng, X.; Zhu, C.; Chen, L.; Guo, Q.; Liu, H.; et al. Antitumor Effect of a Short Peptide on P53-Null SKOV3 Ovarian Cancer Cells. Anti Cancer Drugs 2019, 30, 973–982. [Google Scholar] [CrossRef] [PubMed]
  64. Luo, W.; Yu, Y.; Wang, R.; He, D.; Wang, C.; Zeng, X.; Chen, X.; Tan, X.; Huang, T.; Wu, X. P7 Peptides Targeting bFGF Sensitize Colorectal Cancer Cells to CPT-11. Int. J. Mol. Med. 2014, 33, 194–200. [Google Scholar] [CrossRef] [PubMed][Green Version]
  65. Wu, X.; Yan, Q.; Huang, Y.; Huang, H.; Su, Z.; Xiao, J.; Zeng, Y.; Wang, Y.; Nie, C.; Yang, Y.; et al. Isolation of a Novel Basic FGF-Binding Peptide with Potent Antiangiogenetic Activity. J. Cell Mol. Med. 2010, 14, 351–356. [Google Scholar] [CrossRef]
  66. Wang, W.; Chen, T.; Li, H.; Chen, Y.; Wu, Z.; Feng, T.; Zhang, X.; Zhong, Q.; Zhong, Q.; Li, G.; et al. Screening a Novel FGF3 Antagonist Peptide with Anti-Tumor Effects on Breast Cancer from a Phage Display Library. Mol. Med. Rep. 2015, 12, 7051–7058. [Google Scholar] [CrossRef]
  67. Wang, W.; Chen, X.; Li, T.; Li, Y.; Wang, R.; He, D.; Luo, W.; Li, X.; Wu, X. Screening a Phage Display Library for a Novel FGF8b-Binding Peptide with Anti-Tumor Effect on Prostate Cancer. Exp. Cell Res. 2013, 319, 1156–1164. [Google Scholar] [CrossRef]
  68. Wang, J.; Tan, X.; Guo, Q.; Lin, X.; Huang, Y.; Chen, L.; Zeng, X.; Li, R.; Wang, H.; Wu, X. FGF9 Inhibition by a Novel Binding Peptide Has Efficacy in Gastric and Bladder Cancer per Se and Reverses Resistance to Cisplatin. Pharmacol. Res. 2020, 152, 104575. [Google Scholar] [CrossRef] [PubMed]
  69. Xie, Y.; Su, N.; Yang, J.; Tan, Q.; Huang, S.; Jin, M.; Ni, Z.; Zhang, B.; Zhang, D.; Luo, F.; et al. FGF/FGFR Signaling in Health and Disease. Signal Transduct. Target. Ther. 2020, 5, 181. [Google Scholar] [CrossRef] [PubMed]
  70. Turner, N.; Grose, R. Fibroblast Growth Factor Signalling: From Development to Cancer. Nat. Rev. Cancer 2010, 10, 116–129. [Google Scholar] [CrossRef]
  71. Plotnikov, A.N.; Schlessinger, J.; Hubbard, S.R.; Mohammadi, M. Structural Basis for FGF Receptor Dimerization and Activation. Cell 1999, 98, 641–650. [Google Scholar] [CrossRef] [PubMed]
  72. Zhang, X.; Ibrahimi, O.A.; Olsen, S.K.; Umemori, H.; Mohammadi, M.; Ornitz, D.M. Receptor Specificity of the Fibroblast Growth Factor Family. J. Biol. Chem. 2006, 281, 15694–15700. [Google Scholar] [CrossRef]
  73. Park, J.S.; Choi, J.; Cao, L.; Mohanty, J.; Suzuki, Y.; Park, A.; Baker, D.; Schlessinger, J.; Lee, S. Isoform-Specific Inhibition of FGFR Signaling Achieved by a de-Novo-Designed Mini-Protein. Cell Rep. 2022, 41, 111545. [Google Scholar] [CrossRef]
  74. Chen, G.; Chen, L.; Li, X.; Mohammadi, M. FGF-Based Drug Discovery: Advances and Challenges. Nat. Rev. Drug Discov. 2025, 24, 335–357. [Google Scholar] [CrossRef]
  75. Schlessinger, J.; Plotnikov, A.N.; Ibrahimi, O.A.; Eliseenkova, A.V.; Yeh, B.K.; Yayon, A.; Linhardt, R.J.; Mohammadi, M. Crystal Structure of a Ternary FGF-FGFR-Heparin Complex Reveals a Dual Role for Heparin in FGFR Binding and Dimerization. Mol. Cell 2000, 6, 743–750. [Google Scholar] [CrossRef]
  76. Kuro-o, M. The Klotho Proteins in Health and Disease. Nat. Rev. Nephrol. 2019, 15, 27–44. [Google Scholar] [CrossRef]
  77. Chen, L.; Fu, L.; Sun, J.; Huang, Z.; Fang, M.; Zinkle, A.; Liu, X.; Lu, J.; Pan, Z.; Wang, Y.; et al. Structural Basis for FGF Hormone Signalling. Nature 2023, 618, 862–870. [Google Scholar] [CrossRef]
  78. Wen, X.; Jiao, L.; Tan, H. MAPK/ERK Pathway as a Central Regulator in Vertebrate Organ Regeneration. Int. J. Mol. Sci. 2022, 23, 1464. [Google Scholar] [CrossRef] [PubMed]
  79. Glaviano, A.; Foo, A.S.C.; Lam, H.Y.; Yap, K.C.H.; Jacot, W.; Jones, R.H.; Eng, H.; Nair, M.G.; Makvandi, P.; Geoerger, B.; et al. PI3K/AKT/mTOR Signaling Transduction Pathway and Targeted Therapies in Cancer. Mol. Cancer 2023, 22, 138. [Google Scholar] [CrossRef]
  80. Mandal, S.; Bandyopadhyay, S.; Tyagi, K.; Roy, A. Recent Advances in Understanding the Molecular Role of Phosphoinositide-Specific Phospholipase C Gamma 1 as an Emerging Onco-Driver and Novel Therapeutic Target in Human Carcinogenesis. Biochim. Biophys. Acta (BBA) Rev. Cancer 2021, 1876, 188619. [Google Scholar] [CrossRef] [PubMed]
  81. Philips, R.L.; Wang, Y.; Cheon, H.; Kanno, Y.; Gadina, M.; Sartorelli, V.; Horvath, C.M.; Darnell, J.E.; Stark, G.R.; O’Shea, J.J. The JAK-STAT Pathway at 30: Much Learned, Much More to Do. Cell 2022, 185, 3857–3876. [Google Scholar] [CrossRef] [PubMed]
  82. Ornitz, D.M.; Itoh, N. The Fibroblast Growth Factor Signaling Pathway. WIREs Dev. Biol. 2015, 4, 215–266. [Google Scholar] [CrossRef] [PubMed]
  83. Santhana Kumar, K.; Brunner, C.; Schuster, M.; Kopp, L.L.; Gries, A.; Yan, S.; Jurt, S.; Moehle, K.; Bruns, D.; Grotzer, M.; et al. Discovery of a Small Molecule Ligand of FRS2 That Inhibits Invasion and Tumor Growth. Cell Oncol. 2023, 46, 331–356. [Google Scholar] [CrossRef]
  84. Peng, M.; Deng, J.; Li, X. Clinical Advances and Challenges in Targeting FGF/FGFR Signaling in Lung Cancer. Mol. Cancer 2024, 23, 256. [Google Scholar] [CrossRef]
  85. Katoh, M.; Loriot, Y.; Brandi, G.; Tavolari, S.; Wainberg, Z.A.; Katoh, M. FGFR-Targeted Therapeutics: Clinical Activity, Mechanisms of Resistance and New Directions. Nat. Rev. Clin. Oncol. 2024, 21, 312–329. [Google Scholar] [CrossRef]
  86. Edirisinghe, O.; Ternier, G.; Alraawi, Z.; Suresh Kumar, T.K. Decoding FGF/FGFR Signaling: Insights into Biological Functions and Disease Relevance. Biomolecules 2024, 14, 1622. [Google Scholar] [CrossRef]
  87. Jafari, B.; Hamzeh-Mivehroud, M.; Morris, M.B.; Dastmalchi, S. Exploitation of Phage Display for the Development of Anti-Cancer Agents Targeting Fibroblast Growth Factor Signaling Pathways: New Strategies to Tackle an Old Challenge. Cytokine Growth Factor. Rev. 2019, 46, 54–65. [Google Scholar] [CrossRef]
  88. Istomina, P.V.; Gorchakov, A.A.; Paoin, C.; Yamabhai, M. Phage Display for Discovery of Anticancer Antibodies. New Biotechnol. 2024, 83, 205–218. [Google Scholar] [CrossRef]
  89. Sousa-Victor, P.; García-Prat, L.; Muñoz-Cánoves, P. Control of Satellite Cell Function in Muscle Regeneration and Its Disruption in Ageing. Nat. Rev. Mol. Cell Biol. 2022, 23, 204–226. [Google Scholar] [CrossRef]
  90. Lee, Y.; Lim, K.-M.; Bong, H.; Lee, S.-B.; Jeon, T.-I.; Lee, S.-Y.; Park, H.-S.; Kim, J.-Y.; Song, K.; Kang, G.-H.; et al. The Immobilization of an FGF2-Derived Peptide on Culture Plates Improves the Production and Therapeutic Potential of Extracellular Vesicles from Wharton’s Jelly Mesenchymal Stem Cells. Int. J. Mol. Sci. 2024, 25, 10709. [Google Scholar] [CrossRef]
  91. Verrecchio, A.; Germann, M.W.; Schick, B.P.; Kung, B.; Twardowski, T.; San Antonio, J.D. Design of Peptides with High Affinities for Heparin and Endothelial Cell Proteoglycans. J. Biol. Chem. 2000, 275, 7701–7707. [Google Scholar] [CrossRef]
  92. Kiselyov, V.V.; Skladchikova, G.; Hinsby, A.M.; Jensen, P.H.; Kulahin, N.; Soroka, V.; Pedersen, N.; Tsetlin, V.; Poulsen, F.M.; Berezin, V.; et al. Structural Basis for a Direct Interaction between FGFR1 and NCAM and Evidence for a Regulatory Role of ATP. Structure 2003, 11, 691–701. [Google Scholar] [CrossRef]
  93. Cherian, K.E.; Paul, T.V. Inherited Fibroblast Growth Factor 23 Excess. Best Pract. Res. Clin. Endocrinol. Metab. 2024, 38, 101844. [Google Scholar] [CrossRef]
  94. Lin, Y.-Z.; Yao, S.; Veach, R.A.; Torgerson, T.R.; Hawiger, J. Inhibition of Nuclear Translocation of Transcription Factor NF-κB by a Synthetic Peptide Containing a Cell Membrane-Permeable Motif and Nuclear Localization Sequence. J. Biol. Chem. 1995, 270, 14255–14258. [Google Scholar] [CrossRef]
  95. Komi, A.; Suzuki, M.; Imamura, T. Permeable FGF-1 Nuclear Localization Signal Peptide Stimulates DNA Synthesis in Various Cell Types but Is Cell-Density Sensitive and Unable to Support Cell Proliferation. Exp. Cell Res. 1998, 243, 408–414. [Google Scholar] [CrossRef]
  96. Li, A.-J.; Tsuboyama, H.; Komi, A.; Ikekita, M.; Imamura, T. Strong Suppression of Feeding by a Peptide Containing Both the Nuclear Localization Sequence of Fibroblast Growth Factor-1 and a Cell Membrane-Permeable Sequence. Neurosci. Lett. 1998, 255, 41–44. [Google Scholar] [CrossRef] [PubMed]
  97. She, Q.-Y.; Bao, J.-F.; Wang, H.-Z.; Liang, H.; Huang, W.; Wu, J.; Zhong, Y.; Ling, H.; Li, A.; Qin, S.-L. Fibroblast Growth Factor 21: A “Rheostat” for Metabolic Regulation? Metabolism 2022, 130, 155166. [Google Scholar] [CrossRef]
  98. Adams, A.C.; Cheng, C.C.; Coskun, T.; Kharitonenkov, A. FGF21 Requires Βklotho to Act In Vivo. PLoS ONE 2012, 7, e49977. [Google Scholar] [CrossRef]
  99. Ruan, R.; Li, L.; Li, X.; Huang, C.; Zhang, Z.; Zhong, H.; Zeng, S.; Shi, Q.; Xia, Y.; Zeng, Q.; et al. Unleashing the Potential of Combining FGFR Inhibitor and Immune Checkpoint Blockade for FGF/FGFR Signaling in Tumor Microenvironment. Mol. Cancer 2023, 22, 60. [Google Scholar] [CrossRef]
  100. Terada, T.; Mizobata, M.; Kawakami, S.; Yamashita, F.; Hashida, M. Optimization of Tumor-Selective Targeting by Basic Fibroblast Growth Factor-Binding Peptide Grafted PEGylated Liposomes. J. Control. Release 2007, 119, 262–270. [Google Scholar] [CrossRef]
  101. Kimura-Ueki, M.; Oda, Y.; Oki, J.; Komi-Kuramochi, A.; Honda, E.; Asada, M.; Suzuki, M.; Imamura, T. Hair Cycle Resting Phase Is Regulated by Cyclic Epithelial FGF18 Signaling. J. Investig. Dermatol. 2012, 132, 1338–1345. [Google Scholar] [CrossRef]
  102. Higgins, C.A.; Petukhova, L.; Harel, S.; Ho, Y.Y.; Drill, E.; Shapiro, L.; Wajid, M.; Christiano, A.M. FGF5 Is a Crucial Regulator of Hair Length in Humans. Proc. Natl. Acad. Sci. USA 2014, 111, 10648–10653. [Google Scholar] [CrossRef]
  103. Saw, P.E.; Song, E.-W. Phage Display Screening of Therapeutic Peptide for Cancer Targeting and Therapy. Protein Cell 2019, 10, 787–807. [Google Scholar] [CrossRef]
  104. Suh, J.; Kim, D.; Lee, Y.; Jang, J.; Surh, Y. Fibroblast Growth Factor-2, Derived from Cancer-associated Fibroblasts, Stimulates Growth and Progression of Human Breast Cancer Cells via FGFR1 Signaling. Mol. Carcinog. 2020, 59, 1028–1040. [Google Scholar] [CrossRef]
  105. Dienstmann, R.; Rodon, J.; Prat, A.; Perez-Garcia, J.; Adamo, B.; Felip, E.; Cortes, J.; Iafrate, A.J.; Nuciforo, P.; Tabernero, J. Genomic Aberrations in the FGFR Pathway: Opportunities for Targeted Therapies in Solid Tumors. Ann. Oncol. 2014, 25, 552–563. [Google Scholar] [CrossRef] [PubMed]
  106. Mattila, M.; Harkonen, P. Role of Fibroblast Growth Factor 8 in Growth and Progression of Hormonal Cancer. Cytokine Growth Factor Rev. 2007, 18, 257–266. [Google Scholar] [CrossRef] [PubMed]
  107. Chen, H.; Ren, C.; Han, C.; Wang, D.; Chen, Y.; Fu, D. Expression and Prognostic Value of miR-486-5p in Patients with Gastric Adenocarcinoma. PLoS ONE 2015, 10, e0119384. [Google Scholar] [CrossRef]
  108. Knowles, M.A. Novel Therapeutic Targets In Bladder Cancer: Mutation and Expression of Fgf Receptors. Future Oncol. 2008, 4, 71–83. [Google Scholar] [CrossRef]
  109. Wrobel, W.; Pach, E.; Ben-Skowronek, I. Advantages and Disadvantages of Different Treatment Methods in Achondroplasia: A Review. Int. J. Mol. Sci. 2021, 22, 5573. [Google Scholar] [CrossRef]
  110. Zhao, J.; Lin, H.; Wang, L.; Guo, K.; Jing, R.; Li, X.; Chen, Y.; Hu, Z.; Gao, S.; Xu, N. Suppression of FGF5 and FGF18 Expression by Cholesterol-Modified siRNAs Promotes Hair Growth in Mice. Front. Pharmacol. 2021, 12, 666860. [Google Scholar] [CrossRef]
  111. Tsuruda, A.; Kawano, Y.; Maekawa, T.; Oka, S. A Short Peptide GPIGS Promotes Proliferation of Hair Bulb Keratinocytes and Accelerates Hair Regrowth in Mice. Biol. Pharm. Bull. 2005, 28, 485–489. [Google Scholar] [CrossRef][Green Version]
  112. Cai, J.; Dou, G.; Zheng, L.; Yang, T.; Jia, X.; Tang, L.; Huang, Y.; Wu, W.; Li, X.; Wang, X. Pharmacokinetics of Topically Applied Recombinant Human Keratinocyte Growth Factor-2 in Alkali-Burned and Intact Rabbit Eye. Exp. Eye Res. 2015, 136, 93–99. [Google Scholar] [CrossRef]
  113. Sun, J.; Wu, J.; Jin, H.; Ying, T.; Jin, W.; Fan, M.; Zhou, J.; Chen, H.; Jin, L.; Zhou, J. Structure-Guided Design, Generation, and Biofunction of PEGylated Fibroblast Growth Factor 2 Variants for Wound Healing. Nanoscale 2020, 12, 18200–18213. [Google Scholar] [CrossRef]
  114. Xia, X.; Babcock, J.P.; Blaber, S.I.; Harper, K.M.; Blaber, M. Pharmacokinetic Properties of 2nd-Generation Fibroblast Growth Factor-1 Mutants for Therapeutic Application. PLoS ONE 2012, 7, e48210. [Google Scholar] [CrossRef]
  115. Subbiah, V.; Verstovsek, S. Clinical Development and Management of Adverse Events Associated with FGFR Inhibitors. Cell Rep. Med. 2023, 4, 101204. [Google Scholar] [CrossRef]
  116. Shinbara, K.; Liu, W.; Van Neer, R.H.P.; Katoh, T.; Suga, H. Methodologies for Backbone Macrocyclic Peptide Synthesis Compatible With Screening Technologies. Front. Chem. 2020, 8, 447. [Google Scholar] [CrossRef]
  117. Li, X.; Wang, N.; Liu, Y.; Li, W.; Bai, X.; Liu, P.; He, C.-Y. Backbone N-Methylation of Peptides: Advances in Synthesis and Applications in Pharmaceutical Drug Development. Bioorg. Chem. 2023, 141, 106892. [Google Scholar] [CrossRef]
  118. Fischer, N.H.; Nielsen, D.S.; Palmer, D.; Meldal, M.; Diness, F. C-Terminal Lactamization of Peptides. Chem. Commun. 2021, 57, 895–898. [Google Scholar] [CrossRef]
  119. Andrianov, A.K. Noncovalent PEGylation of Protein and Peptide Therapeutics. WIREs Nanomed. Nanobiotechnol. 2023, 15, e1897. [Google Scholar] [CrossRef]
  120. Martin, V.; Egelund, P.H.G.; Johansson, H.; Thordal Le Quement, S.; Wojcik, F.; Sejer Pedersen, D. Greening the Synthesis of Peptide Therapeutics: An Industrial Perspective. RSC Adv. 2020, 10, 42457–42492. [Google Scholar] [CrossRef]
  121. Noki, S.; De La Torre, B.G.; Albericio, F. Safety-Catch Linkers for Solid-Phase Peptide Synthesis. Molecules 2024, 29, 1429. [Google Scholar] [CrossRef]
  122. Zhang, Z.-X.; Nong, F.-T.; Wang, Y.-Z.; Yan, C.-X.; Gu, Y.; Song, P.; Sun, X.-M. Strategies for Efficient Production of Recombinant Proteins in Escherichia Coli: Alleviating the Host Burden and Enhancing Protein Activity. Microb. Cell Fact. 2022, 21, 191. [Google Scholar] [CrossRef]
  123. Eskandari, A.; Nezhad, N.G.; Leow, T.C.; Rahman, M.B.A.; Oslan, S.N. Current Achievements, Strategies, Obstacles, and Overcoming the Challenges of the Protein Engineering in Pichia Pastoris Expression System. World J. Microbiol. Biotechnol. 2024, 40, 39. [Google Scholar] [CrossRef]
  124. Stevens, T.A.; Tomaleri, G.P.; Hazu, M.; Wei, S.; Nguyen, V.N.; DeKalb, C.; Voorhees, R.M.; Pleiner, T. A Nanobody-Based Strategy for Rapid and Scalable Purification of Human Protein Complexes. Nat. Protoc. 2024, 19, 127–158. [Google Scholar] [CrossRef]
  125. Silverman, A.D.; Karim, A.S.; Jewett, M.C. Cell-Free Gene Expression: An Expanded Repertoire of Applications. Nat. Rev. Genet. 2020, 21, 151–170. [Google Scholar] [CrossRef]
  126. Hartrampf, N.; Saebi, A.; Poskus, M.; Gates, Z.P.; Callahan, A.J.; Cowfer, A.E.; Hanna, S.; Antilla, S.; Schissel, C.K.; Quartararo, A.J.; et al. Synthesis of Proteins by Automated Flow Chemistry. Science 2020, 368, 980–987. [Google Scholar] [CrossRef]
  127. Haddadzadegan, S.; Dorkoosh, F.; Bernkop-Schnürch, A. Oral Delivery of Therapeutic Peptides and Proteins: Technology Landscape of Lipid-Based Nanocarriers. Adv. Drug Deliv. Rev. 2022, 182, 114097. [Google Scholar] [CrossRef]
  128. Shan, B.; Wu, F. Hydrogel-Based Growth Factor Delivery Platforms: Strategies and Recent Advances. Adv. Mater. 2024, 36, 2210707. [Google Scholar] [CrossRef]
  129. Mohammadi, Z.; Eini, M.; Rastegari, A.; Tehrani, M.R. Chitosan as a Machine for Biomolecule Delivery: A Review. Carbohydr. Polym. 2021, 256, 117414. [Google Scholar] [CrossRef]
  130. Li, C.; Li, T.; Tian, X.; An, W.; Wang, Z.; Han, B.; Tao, H.; Wang, J.; Wang, X. Research Progress on the PEGylation of Therapeutic Proteins and Peptides (TPPs). Front. Pharmacol. 2024, 15, 1353626. [Google Scholar] [CrossRef]
  131. Holz, E.; Darwish, M.; Tesar, D.B.; Shatz-Binder, W. A Review of Protein- and Peptide-Based Chemical Conjugates: Past, Present, and Future. Pharmaceutics 2023, 15, 600. [Google Scholar] [CrossRef]
  132. Zhao, J.; Qiu, P.; Wang, Y.; Wang, Y.; Zhou, J.; Zhang, B.; Zhang, L.; Gou, D. Chitosan-Based Hydrogel Wound Dressing: From Mechanism to Applications, a Review. Int. J. Biol. Macromol. 2023, 244, 125250. [Google Scholar] [CrossRef]
  133. Cao, H.; Duan, L.; Zhang, Y.; Cao, J.; Zhang, K. Current Hydrogel Advances in Physicochemical and Biological Response-Driven Biomedical Application Diversity. Signal Transduct. Target. Ther. 2021, 6, 426. [Google Scholar] [CrossRef]
  134. Abdel-Mageed, H.M.; Abd El Aziz, A.E.; Mohamed, S.A.; AbuelEzz, N.Z. The Tiny Big World of Solid Lipid Nanoparticles and Nanostructured Lipid Carriers: An Updated Review. J. Microencapsul. 2022, 39, 72–94. [Google Scholar] [CrossRef]
  135. Li, X.; Naeem, A.; Xiao, S.; Hu, L.; Zhang, J.; Zheng, Q. Safety Challenges and Application Strategies for the Use of Dendrimers in Medicine. Pharmaceutics 2022, 14, 1292. [Google Scholar] [CrossRef]
  136. Lauko, A.; Pellock, S.J.; Sumida, K.H.; Anishchenko, I.; Juergens, D.; Ahern, W.; Jeung, J.; Shida, A.; Hunt, A.; Kalvet, I.; et al. Computational Design of Serine Hydrolases. Science 2025, 388, eadu2454. [Google Scholar] [CrossRef]
  137. Liu, H.; Song, Z.; Zhang, Y.; Wu, B.; Chen, D.; Zhou, Z.; Zhang, H.; Li, S.; Feng, X.; Huang, J.; et al. De Novo Design of Self-Assembling Peptides with Antimicrobial Activity Guided by Deep Learning. Nat. Mater. 2025, 24, 1295–1306. [Google Scholar] [CrossRef]
  138. Watson, J.L.; Juergens, D.; Bennett, N.R.; Trippe, B.L.; Yim, J.; Eisenach, H.E.; Ahern, W.; Borst, A.J.; Ragotte, R.J.; Milles, L.F.; et al. De Novo Design of Protein Structure and Function with RFdiffusion. Nature 2023, 620, 1089–1100. [Google Scholar] [CrossRef]
  139. Yang, W.; Hicks, D.R.; Ghosh, A.; Schwartze, T.A.; Conventry, B.; Goreshnik, I.; Allen, A.; Halabiya, S.F.; Kim, C.J.; Hinck, C.S.; et al. Design of High-Affinity Binders to Immune Modulating Receptors for Cancer Immunotherapy. Nat. Commun. 2025, 16, 2001. [Google Scholar] [CrossRef]
Figure 1. A brief overview of FGFs. There are 22 identified FGFs, which are classified into seven subfamilies. Structure diagrams of FGF9 (PDB ID: 5W59), FGF12 (PDB ID: 1Q1U) and FGF19 (PDB ID: 2P23).
Figure 1. A brief overview of FGFs. There are 22 identified FGFs, which are classified into seven subfamilies. Structure diagrams of FGF9 (PDB ID: 5W59), FGF12 (PDB ID: 1Q1U) and FGF19 (PDB ID: 2P23).
Bioengineering 12 01019 g001
Figure 2. Schematic illustration of FGFRs and FGF-induced FGFR dimerization. (A) Structure of FGFRs and FGFRL1, as well as alternative splicing during gene expression. (B) Paracrine FGFs (represented by FGF2) promote FGFR dimerization. (C) Endocrine FGFs (represented by FGF23) promote FGFR dimerization. Structure diagrams of FGF2-FGFR1c-HS (PDB ID: 1FQ9) and FGF23-FGFR1c-HS-aKlotho (PDB ID: 7YSH). FGFs are colored in orange; FGFR in light blue; Klotho in light purple; and HS in purple. In the FGF23-FGFR1c-HS-aKlotho complex, the primary FGFR (FGFRp) is colored in green, and the secondary FGFR (FGFRs) in light blue.
Figure 2. Schematic illustration of FGFRs and FGF-induced FGFR dimerization. (A) Structure of FGFRs and FGFRL1, as well as alternative splicing during gene expression. (B) Paracrine FGFs (represented by FGF2) promote FGFR dimerization. (C) Endocrine FGFs (represented by FGF23) promote FGFR dimerization. Structure diagrams of FGF2-FGFR1c-HS (PDB ID: 1FQ9) and FGF23-FGFR1c-HS-aKlotho (PDB ID: 7YSH). FGFs are colored in orange; FGFR in light blue; Klotho in light purple; and HS in purple. In the FGF23-FGFR1c-HS-aKlotho complex, the primary FGFR (FGFRp) is colored in green, and the secondary FGFR (FGFRs) in light blue.
Bioengineering 12 01019 g002
Figure 3. Schematic illustration of the molecular basis and signaling pathways underlying the functional activity of FGF-derived peptides [85,86]. The dashed box shows the binding of FGF-derived peptides to FGFRs.
Figure 3. Schematic illustration of the molecular basis and signaling pathways underlying the functional activity of FGF-derived peptides [85,86]. The dashed box shows the binding of FGF-derived peptides to FGFRs.
Bioengineering 12 01019 g003
Figure 4. Schematic representation of the binding interaction between FGF-derived peptides and FGFR. Full-length FGF is colored in orange; various FGF-derived peptides are colored in pink, light red, yellow, blue, purple, light purple, green, dark green and light green, respectively; FGFR is colored in grey.
Figure 4. Schematic representation of the binding interaction between FGF-derived peptides and FGFR. Full-length FGF is colored in orange; various FGF-derived peptides are colored in pink, light red, yellow, blue, purple, light purple, green, dark green and light green, respectively; FGFR is colored in grey.
Bioengineering 12 01019 g004
Table 1. Fibroblast growth factor-derived peptides.
Table 1. Fibroblast growth factor-derived peptides.
Peptide Name [Refs.]Peptide
Sequence
OriginCorresponding ResiduesWorking
Concentration
Mode of ActionModelApplication
FGF1 NLS [19]NYKKPKLHuman FGF121~2720~100 μg/mLfusion with a cell-penetrating peptideIn Vitro Model: NIH3T3 Cell LineMetabolic Regulation
Hexafin 1 [20]TGQYLAMDTDGLLYGSHuman FGF176~910~100 μmol/LTetramerIn Vitro Model: Neurite Outgrowth Assay Using Cerebellar Granule NeuronsNeural Functional Recovery
peptide 1 [21]SKKHAEKNWFhuman FGF1114~12310~200 μmol/LCyclic peptideIn Vitro Model: 3T3 Cell Proliferation AssayRational Design of Peptides
Canofin 1 [22]HFKDPKRLYCKhuman FGF225~350~100 μmol/LTetramerIn Vitro Model: Neurite Outgrowth Assay of Cerebellar Granule NeuronsNeural Functional Recovery
Peptide 33 [23]CKNGGFFhuman FGF234~4010~400 μmol/LLinear monomerIn Vitro Model: Skeletal Muscle Satellite Cell Proliferation AssayMuscle Regeneration
Peptide 33-13 [23]CKNGGFFLRIHPDhuman FGF234~4610~400 μmol/LLinear monomerIn Vitro Model: Skeletal Muscle Satellite Cell Proliferation AssayMuscle Regeneration
F36 [24]PDGRVDhuman FGF245~50100 μmol/L (conjugation dose)immobilized on the surface of a chitosan membraneIn Vitro Model: Human Mesenchymal Stem Cell Adhesion and Osteogenic Differentiation AssayBone Regeneration
FREG [25,26]DPHIKLQLQAEhuman FGF257~670~100 ng/mL(cells)
3~6 mg·kg−1·d−1 (animals)
Linear monomerIn Vitro Model: Human Melanoma Cell Proliferation and Invasion Assay
In Vivo Model: Melanoma Mouse Model
Tumor Suppression
P5 and DcP5 [27,28]LQLQAEERhuman FGF262~695~15 μmol/L (cells)
10 mg/kg (animals)
20~200 μmol/L (conjugation dose)
Linear monomer
Cyclic peptide
conjugated with the polysaccharide hyaluronic acid
In Vitro Model: DU145 Prostate Cancer Cell Proliferation Model
In Vivo Models: Tumor Model and Acne Model
Tumor Suppression
Alleviation of Acne
FP2 [29]ERGVVSIKGVhuman FGF268~770.05 μg/mL (conjugation dose)fused with mussel adhesive protein
immobilized on the surface of the culture plate
In Vitro Model: Proliferation and Differentiation of Human Wharton’s Jelly-Derived Mesenchymal Stem Cells
In Vivo Model: Osteoarthritis Model
Bone Regeneration
Hexafin 2 [20]ANRYLAMKEDGRLLAShuman FGF279~940~100 μmol/LTetramerIn Vitro Model: Neurite Outgrowth Assay of Cerebellar Granule NeuronsNeural Functional Recovery
F77 [24]KEDGRLLhuman FGF286~92100 μmol/L (conjugation dose)immobilized on the surface of a chitosan membraneIn Vitro Model: Adhesion and Osteogenic Differentiation of Human Mesenchymal Stem CellsBone Regeneration
FK18 [30]FFFERLESNNYNTYRSRKhuman FGF2102~1190~100 μg/mLLinear monomerIn Vitro Model: Oxygen-Glucose Deprivation (OGD) Model in SH-SY5Y Cells
In Vivo Model: Retinal Ischemia Model
Neural Functional Recovery
Peptide 12 [31]FFFERLESNNYNTYRSRKYSSWYVAhuman FGF2102~12650 μg/animalconjugated with VLPsIn Vivo Model: Breast Tumor ModelTumor Suppression
FGF-P [32,33,34,35,36,37,38]YRSRKYSSWYVALKRhuman FGF2115~129200 ng/mL (cells)
0~20 mg/kg (animals)
3~6 mmol/L (conjugation dose)
40~400 ng/sample (fusion protein)
Linear monomer
conjugated with PA
fused with a heparin-binding sequence
In Vitro Model: Proliferation and Migration Assays Using Hs-27 Fibroblasts and Keratinocytes
In Vivo Model: Total Body Irradiation (TBI) Model, Bone Marrow Syndrome Model, Skin Burn Injury Model, Spinal Cord Injury (SCI) Model
Multi-Organ Repair
Spinal Cord Injury Repair
Tissue Regeneration
F105 [39]YKRSRYThuman FGF2120~114100 μmol/L (conjugation dose)immobilized on the surface of the culture plateIn Vitro Model: Adhesion and Osteogenic Differentiation of Human Mesenchymal Stem CellsBone Regeneration
BGF1 [40]CLKRTGQYKLChuman FGF2127~1350~1.8 mmol/L (cells)
2~10 mg/kg (animals)
Cyclic peptideIn Vitro Model: Proliferation models of human umbilical vein endothelial cells (HUVECs), 4T1 breast cancer cells, U87 glioblastoma cells, and SKOV3 ovarian cancer cells
In Vivo Model: 4T1 Breast Cancer Model
Tumor Suppression
bFGFp [41]KRTGQYKLChuman FGF2128~135100 mg/mL (conjugation dose)conjugated with bovine serum albumin or liposomesIn Vitro Model: 3T3 Cell Proliferation AssayTumor Suppression
F119 [39]KRTGQYKLGSKTGPGQKhuman FGF2128~144100 μmol/L (conjugation dose)immobilized on the surface of the culture plateIn Vitro Model: Adhesion and Osteogenic Differentiation of Human Mesenchymal Stem CellsBone Regeneration
Canofin 3 [22]KTGPGQKAILhuman FGF2138~1470~100 μmol/LTetramerIn Vitro Model: Neurite Outgrowth Assay of Cerebellar Granule NeuronsNeural Functional Recovery
Canofin 2 [22]FLPMSAKShuman FGF2147~1550~100 μmol/LTetramerIn Vitro Model: Neurite Outgrowth Assay of Cerebellar Granule NeuronsNeural Functional Recovery
FGF3 NLS [42]RRRKhuman FGF344~470~100 μg/mLconjugated with PAMAMIn Vitro Model: Transfection of HEK293 and HeLa CellsCell Transfection
Hexafin 3 [20]SGRYLAMNKRGRLYKShuman FGF393~1080~10 μmol/LTetramerIn Vitro Model: Neurite Outgrowth Assay of Cerebellar Granule NeuronsNeural Functional Recovery
SP and IP [43,44]AAVALLPAVLLALLAPhuman FGF47~220~80 μmol/L (conjugation dose)
0~500 μmol/L (conjugation dose)
conjugated with lysine
conjugated with PL
In Vitro Model: Protein Delivery into 143B, TE85, MG63, and FOB Cells; Oligonucleotide Delivery into A549 CellsProtein Delivery
Oligonucleotide Delivery
P3 [45]VGIGFHLQIYhuman FGF595~1041~1000 mmol/L (cells)
5 μg/subject (animals)
Linear monomerIn Vitro Model: 3T3 Cell Proliferation Assay
In Vivo Model: Depilated Mouse Model
Hair Follicle Repair
KGFp [46]KELILENHYNTYAhuman FGF7140~1521~100 ng/mL (cells)Linear monomer
conjugated to a 3D porous scaffold
In Vitro Model: Migration and Differentiation of Human Bone Marrow Mesenchymal Stem Cells
In Vivo Model: Chronic Wound Model in Type 2 Diabetic Mice
Tissue Repair
8b-13 [47,48,49]PNFTQHVREQSLVhuman FGF830~421~125 nmol/LLinear monomerIn Vitro Model: Proliferation Assay of PC-3 and DU-145 Prostate Cancer CellsTumor Suppression
Hexafin 8 [20]TGLYICMNKKGKLIAKhuman FGF8104~1190~10 μmol/LTetramerIn Vitro Model: Neurite Outgrowth Assay of Cerebellar Granule NeuronsNeural Functional Recovery
Hexafin 9 [20]SGLYLGMNEKGELYGShuman FGF9112~1270~100 μmol/LTetramerIn Vitro Model: Neurite Outgrowth Assay of Cerebellar Granule NeuronsNeural Functional Recovery
Hexafin 10 [20]SNYYLAMNKKGKLYGShuman FGF10128~1430~10 μmol/LTetramerIn Vitro Model: Neurite Outgrowth Assay of Cerebellar Granule NeuronsNeural Functional Recovery
Hexafin 17 [20]SEKYICMNKRGKLIGKhuman FGF1793~1080~10 μmol/LTetramerIn Vitro Model: Neurite Outgrowth Assay of Cerebellar Granule NeuronsNeural Functional Recovery
FGF23 Peptide [50]AEDDSERDPLNVLKPRARMTPAPAShuman FGF23181~2050.2 nmol/LLinear monomerIn Vivo Model: Hyperphosphatemia in Fgf23−/− MiceMetabolic Regulation
FAP1 [51]RERNEVNHYRTYComputational Design of Peptide Derivatives Targeting Human FGFR1c1~100 ng/mL (cells)
1~10 mg/kg (animals)
Linear monomerIn Vitro Model: Proliferation and Migration Assays Using NIH 3T3 Cells
In Vivo Model: Diabetic Mouse Wound Healing Model
Tissue Repair
C19jun [52]AESGDDYCVLVFTDSAWTKICDWSHFRNPhage Display Technology for Screening FGFR1c-Binding Peptide Derivatives0~10 nmol/Lfused with human c-Jun residues for expression
Dimer
In Vitro Model: Swiss 3T3 Cell Proliferation and Neurite Outgrowth in Neuronal CellsTissue Repair
F8 [53]ACSLNHTVNCPhage Display Technology for Screening FGFR1c-Binding Peptide Derivatives0~10 μmol/LCyclic peptideIn Vitro Model: BA/F3 Cell Proliferation AssayTumor Suppression
F91-8A07 [54]LPGRTCREYPDLWWVRCYPhage Display Technology for Screening FGFR1c/β-Klotho-Binding Peptide Derivatives0~1000 μmol/L (cells)
0~1000 nmol/kg (animals)
DimerIn Vitro Model: Primary Human Adipocyte Model
In Vivo Model: Mouse Model
Metabolic Regulation
CH02 [55]GPANVETPhage Display Technology for Screening FGFR2c-Binding Peptide Derivatives0~40 μmol/LLinear monomerIn Vitro Model: Neurite Outgrowth Assay of Dorsal Root Ganglion (DRG) Neurons
In Vivo Model: Rat Dorsal Root Compression Injury Model
Neural Functional Recovery
H1 [56]SNFLHLGPhage Display Technology for Screening FGFR2c-Binding Peptide Derivatives0~20 μmol/L (cells)
0~1000 μmol/L (animals)
Linear monomerIn Vitro Model: 3T3 Cell Proliferation and Migration Assay
In Vivo Model: Full-Thickness Excisional Wound Model
Tissue Repair
P3 [57]VSPPLTLGQLLSPhage Display Technology for Screening FGFR3-Binding Peptide Derivatives0~50 μmol/L (cells)
100 μg·kg−1·d−1 (animals)
Linear monomerIn Vitro Model: ATDC5 Cell Proliferation and Chondrogenic Differentiation Model
In Vivo Model: TDII Mouse Lethal Phenotype Model
Bone Regeneration
peptide [58]MQLPLATPhage Display Technology for Screening FGFR-Binding Peptide Derivatives10–20 μg/mL (conjugation dose)Conjugation with PEI-PEGIn Vitro Model: B16F10 Cell Transfection ModelCell Transfection
23-b6 [59]SSPPKSPPhage Display Technology for Screening FGFR-Klotho-Binding Peptide Derivatives0~0.1 μmol/LLinear monomerIn Vitro Model: Phosphate Uptake Assay in Renal Proximal Tubule CellsMetabolic Regulation
Pro-Ile [60]PIFunctional Screening of Human FGFR-Binding Peptide Derivatives Using Bacterial Conditioned Medium0~1 mmol/LLinear monomerIn Vitro Model: Keratinocyte Proliferation AssayHair Follicle Repair
AP8 [61]AGNWTPIPhage Display Technology for Screening FGF1-Binding Peptide Derivatives0~16 μmol/LLinear monomerIn Vitro Model: Proliferation Assay of Breast Cancer Cells and Human Umbilical Vein Endothelial CellsTumor Suppression
P7 [62,63,64]PLLQATLGGGSPhage Display Technology for Screening FGF2-Binding Peptide Derivatives0~16 μmol/L (cells)Linear monomerIn Vitro Model: Proliferation and Migration Assay of MDA-MB-231 Breast Cancer CellsTumor Suppression
P7Δ [65]PLLQATLPhage Display Technology for Screening FGF2-Binding Peptide Derivatives0~16 μmol/L (cells)
1 μmol/L (animals)
Linear monomerIn Vitro Model: Proliferation Assay of BALB/c 3T3 Cells
In Vivo Model: Chick Embryo Chorioallantoic Membrane (CAM) Assay
Tumor Suppression
FP16 [66]VLWLKNRPhage Display Technology for Screening FGF3-Binding Peptide Derivatives0~16 μmol/LLinear monomerIn Vitro Model: Proliferation Assay of MDA-MB-231 and T47D Breast Cancer CellsTumor Suppression
P12 [67]HSQAAVPPhage Display Technology for Screening FGF8b-Binding Peptide Derivatives0~16 μmol/LLinear monomerIn Vitro Model: Proliferation Assay of PC-3 and HUVECsTumor Suppression
P4 [68]NVFTVSPPhage Display Technology for Screening FGF9-Binding Peptide Derivatives0~16 μmol/LLinear monomerIn Vitro Model: Proliferation Assay of SGC-7901 Gastric Cancer Cells and RT-112 Bladder Cancer CellsTumor Suppression
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Cao, C.-K.; Shi, Z.-Y.; Chen, C.-B.; Li, X.-K.; Su, Z.-J. Fibroblast Growth Factor-Derived Peptides: Sources, Functions, and Applications. Bioengineering 2025, 12, 1019. https://doi.org/10.3390/bioengineering12101019

AMA Style

Cao C-K, Shi Z-Y, Chen C-B, Li X-K, Su Z-J. Fibroblast Growth Factor-Derived Peptides: Sources, Functions, and Applications. Bioengineering. 2025; 12(10):1019. https://doi.org/10.3390/bioengineering12101019

Chicago/Turabian Style

Cao, Cheng-Kun, Zhong-Yuan Shi, Chuan-Bang Chen, Xiao-Kun Li, and Zhi-Jian Su. 2025. "Fibroblast Growth Factor-Derived Peptides: Sources, Functions, and Applications" Bioengineering 12, no. 10: 1019. https://doi.org/10.3390/bioengineering12101019

APA Style

Cao, C.-K., Shi, Z.-Y., Chen, C.-B., Li, X.-K., & Su, Z.-J. (2025). Fibroblast Growth Factor-Derived Peptides: Sources, Functions, and Applications. Bioengineering, 12(10), 1019. https://doi.org/10.3390/bioengineering12101019

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop