Back to Journals » International Journal of Nanomedicine » Volume 20
Polymeric Nanomedicines in Diabetic Wound Healing: Applications and Future Perspectives
Authors Chen Z , Chan K, Li X , Gong L, Ma Y, Huang C, Lu Y , Wang L, Piao C
Received 23 December 2024
Accepted for publication 29 April 2025
Published 22 May 2025 Volume 2025:20 Pages 6423—6446
DOI https://doi.org/10.2147/IJN.S514000
Checked for plagiarism Yes
Review by Single anonymous peer review
Peer reviewer comments 2
Editor who approved publication: Dr Xing Zhang
Zeyao Chen,1 Kakei Chan,1 Xin Li,1,2 Li Gong,3 Yingjie Ma,4 Chiwen Huang,5 Yan Lu,6 Li Wang,1 Chunli Piao1
1Department of Endocrinology, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, People’s Republic of China; 2Department of Gynecology, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, People’s Republic of China; 3Department of Diabetes, Shenzhen Bao’an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, People’s Republic of China; 4First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China; 5Faculty of Science, The University of Queensland, Brisbane, Queensland, Australia; 6Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese, Guangzhou, People’s Republic of China
Correspondence: Chunli Piao, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, 518000, People’s Republic of China, Tel +86-18819075590, Email [email protected]
Abstract: The management of diabetic wound continues to pose significant clinical obstacles, primarily attributed to bacterial infections, excessive inflammation, oxidative stress, and impaired angiogenesis. These pathological factors not only severely affect patient well-being but also create considerable burden on medical services. Current managements often show limited efficacy, necessitating the exploration of alternative therapeutic strategies. Polymeric nanomedicines (PNs), owing to their nanoscale properties, enhanced cellular uptake, stability, bioavailability, and biocompatibility, have been broadly utilized for diabetic wound treatment. PNs demonstrate remarkable capabilities in microbial inhibition, inflammation regulation, oxidative stress mitigation, and vascular network formation, particularly when combined with various agents, including organic substances (eg, exosomes), inorganic substances (eg, metals), and biomaterials (eg, chitosan, hyaluronic acid, and hydrogels). This article systematically examines recent progress in PN-based interventions for diabetic wound recovery, highlighting the pivotal role of PNs in mitigating bacterial infection, modulating inflammatory responses, and promoting cellular regeneration. Additionally, we provide a novel perspective on the multifunctionality of PNs and their potential for overcoming the limitations of conventional therapies. Overall, PNs represent an innovative and promising approach to diabetic wound management, outperforming conventional therapies in stability, targeted delivery, and multifunctionality. In the future, investigations should concentrate on refining PNs formulations and administration strategies so as to enhance biocompatibility, and conducting well-designed clinical trials to validate their therapeutic efficacy.
Keywords: polymeric nanomedicines, nanotechnology, diabetic wounds healing, drug delivery
Introduction
Diabetes mellitus is a chronic metabolic disease marked by hyperglycemia, usually due to loss of insulin secretion or insulin resistance with relative insufficient insulin secretion.1 In a hyperglycemic state, the immune system is suppressed, increasing the risk of infection and causing a continuous inflammatory response, which impairs wound healing in patients with diabetes. Diabetic foot ulcers (DFUs) is a typical representative of the wound in patients with diabetes and is one of the prevalent complications of diabetes, with a risk ranging from 15% to 25% in adult patients with diabetes.2 According to the latest meta-analysis, the global incidence of DFUs is approximately 6.3%,3 with the incidence in patients with type 2 diabetes reaching 34%.4 In some high - risk regions, the incidence can be even higher. For example, in certain African regions, the incidence of DFUs among diabetic patients may exceed 10%. Additionally, the prevalence tends to increase with age, with elderly diabetic patients having a notably higher risk.5 In a 5-year longitudinal multiracial cohort study conducted in Singapore, the mean hospitalized time of patients with DFUs was 13.3 days, and that with minor and major amputation were 20.5 days and 59.6 days respectively. Among the inpatients with DFUs, it was estimated that the yearly medical expenditure of each patient was US $ 3368 in average. The average costs for patients with minor and major amputation were US $ 10,468, and US $ 30,131 per year respectively.2 As the incidence of diabetes increases, the risk of DFUs rises subsequently. This not only affects patients’ quality of life, but also poses a major challenge to the healthcare system.6
Current clinical strategies for diabetic wound include glycemic control, local wound care, antibiotic therapy, negative pressure wound therapy (NPWT), growth factor therapy, biomaterials, skin substitutes, physical therapy, surgical interventions, and education of daily lifestyles.7–9 However, these strategies still have their limitations. Among them, long-term use of antibiotics may contribute to the development of drug resistance, as well as disruption of the normal balance of skin flora; If the technique is poor or the pressure selection is inappropriate, NPWT also has adverse effects such as toxic shock, aggravation of wound infection, hemorrhage, necrosis, and allergy;10 Although growth factors can accelerate the growth of granulation tissue, they cannot address the underlying causes of diabetic wounds, such as neuropathy and vascular disease;11 Mesenchymal stem cells (MSCs) are also associated with high cost, uncertain treatment efficacy and potential tumor risks.12 In recent years, nanotechnology serves as one of the innovative approaches in diabetic wound treatment.13 Polymeric nanomedicines (PNs) usually refer to nanoscale (1–100 nm) drug carriers which are engineered by polymer materials. Due to their nanostructures, good stability, and biocompatibility, PNs show great potential in wound healing. Beyond controlling drug release, improving drug stability, and enhancing bioavailability, PNs offer multifunctional benefits, including anti-inflammatory, antimicrobial, and pro-angiogenic effects. Their functionalized design further enables targeted therapy, improving microenvironmental barriers in diabetic wound healing and significantly enhancing therapeutic outcomes.14–18
The Underlying Mechanism of Diabetic Wound Healing
Normally, the process of wound healing is categorized into four stages: hemostasis, inflammation, proliferation, and remodeling, and often referred to as the “healing cascade”. During hemostasis, platelets are activated and aggregate to form fibrin clots, while cytokines are released to promote blood clotting and recruit inflammatory cell.19 As the inflammatory response progresses, neutrophils are gradually replaced by macrophages, which promote angiogenesis and tissue repair by secreting growth factors.20 The proliferative phase involves fibroblasts and epithelial cells in tissue remodeling, while the remodeling phase involves collagen transformation and extracellular matrix reconstitution to enhance tissue strength and function. The formation of growth factors and extracellular matrix has a significant influence on the entire wound recovery process, as they promote the efficient migration and proliferation of various cell types, thereby accelerating wound repair.
In contrast, wound healing in diabetic patients is distinct from that in individuals without diabetes, primarily due to the pathophysiological conditions associated with diabetes. Diabetes mellitus is a long-term metabolic disorder marked by persistent high blood sugar levels, typically resulting from impaired insulin secretion, insulin resistance, or a combination of both. The glucose-rich environment in patients with diabetes promotes the long-term innate immunity which affect the aerobic glycolysis of macrophages, the pentose phosphate pathway, and the tricarboxylic acid cycle. This, in turn, macrophages’ ability to engulf and eliminate bacteria is affected. Hyperglycemia may also increase the accumulation of advanced glycation end products (AGEs), which change the redox state of the wound and the immune response, affecting the clearance of pathogens.21 These factors increase the risk of bacterial infection and make it more difficult to remove pathogens once infection occurs,22,23 resulting in rapid local wound progression, necrotized infection, and a heightened risk of amputation in individuals with DFUs.8
In the hemostatic stage of diabetic wound healing, hyperglycemia impairs vascular endothelial cell function, reducing the production of nitric oxide (NO), which in turn decreases blood vessel dilation and reduces blood supply to the wound. It also leads to non-enzymatic glycosylation of platelet membrane proteins and lipids, alters the fluidity and receptor function of platelet membranes, decreases chemokine synthesis, and affects the aggregation and adhesion of platelets.24
During the early stage of inflammation, hyperglycemia reduces the expression of damage-associated molecular patterns (DAMPs), hydrogen peroxide (H2O2) and chemokines such as C-X-C motif chemokine ligand 4 (CXCL4), CXCL8, CXCL10, CXCL12, and CXCL3 at the wound site. This diminishes the activity of chemokines, leading to reduced recruitment of inflammatory cells and unbalanced expression of inflammatory mediators, including tumor necrosis factor-alpha (TNF-α), Interleukin-1 beta (IL-1β), and Interleukin-6 (IL-6) around diabetic wounds. With the increase of reactive oxygen species (ROS) production, a chronic inflammatory state is fostered.25,26
In the later stages of inflammation, hyperglycemia affects macrophage polarization, preventing the conversion of M1 macrophages to the healing-promoting M2 type,20 thus prolonging the inflammatory phase. During the proliferative phase, hyperglycemia reduces fibroblast proliferation and migration by affecting growth factor signaling. Additionally, hyperglycemia affects angiogenesis, reducing angiogenesis and hindering tissue regeneration. It also inhibits the activity of fibroblasts, reduces collagen synthesis, increases non-enzymatic glycosylation of collagen and other extracellular matrix proteins, further delaying wound healing.
In the remodeling stage, collagen synthesis in fibroblasts is inhibited, and the abnormal activity of matrix metalloproteinases, along with non-enzymatic glycosylation, leads to abnormal collagen structure and reduced scar tissue stability. The persistent inflammatory response results in poor scar tissue formation. Additionally, reduced levels of provascular factors such as vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), and transforming growth factor-beta (TGF-β) in diabetes result in decreased angiogenesis at the wound site. This leads to lowered expression of PDGF and its receptors, as well as a reduced angiopoietin 1/angiopoietin 2/ tie 2 (Ang1/Ang2/Tie2) ratio, which interferes with the maturation and stability of the vascular system, further delaying the healing process27 (Figure 1).
![]() |
Figure 1 Phases of physiological and diabetic wound healing. |
The primary distinction comparing a diabetic wound to a normal wound lies in the fact that diabetic wound healing is heavily influenced by internal factors such as hyperglycemia, inflammatory mediators, macrophages, provascular factors, all of which contribute to delayed healing. Unlike normal wound healing, which progresses through well-regulated and coordinated stages, the healing process in diabetic wounds is often prolonged and complicated, requiring targeted therapeutic strategies to overcome these unique challenges.
Types of Polymeric Nanomedicines
With the rapid advancement of nanotechnology, polymeric nanomedicines have found widespread applications across various fields, including biomedicine, drug delivery, and material science. In the realm of wound treatment, PNs exhibit significant prospects and substantial therapeutic benefits.
Polymers are generally classified into natural and synthetic types based on their composition. Natural polymers, such as proteins (eg, collagen, gelatin, silk fibroin, keratin, and natural rubber) and polysaccharides (eg, chitin, chitosan, starch, alginate, cellulose, and hyaluronic acid), have been widely utilized in nanomedicine. On the other hand, synthetic polymers, characterized by well-defined and controllable chemical structures, are also increasingly utilized in nanomedicines. Examples of synthetic polymers include poly(lactic acid-co-glycolic acid)-b-poly(ethylene glycol)-b-poly(lactic acid-co-glycolic acid) (PLGA-PEG-PLGA), polyvinyl alcohol (PVA), polycaprolactone (PCL), and polyethylene glycol (PEG), etc.28
Polymeric nanomedicines can be roughly divided into 6 categories according to their structural differences, including polymer conjugates, dendrimers, polymeric nanocapsules, nanogels, polymeric micelles(PMs), and nanoparticles(NPs)28 (Figure 2). These nanomedicines provide multiple benefits, such as enhanced bioavailability, extended circulation time, and improved solubility of poorly water-soluble drugs, leading to significant breakthroughs in areas such as targeted delivery (both active and passive) and controlled drug release.29
Polymer Conjugates
Polymer–protein conjugates and polymer–drug conjugates, characterized by the conjugation of therapeutic molecules or functional moieties to polymers, offer various advantages including extended circulation times, targeted delivery, controlled release, and decreased immunogenicity.30
Polymer–protein conjugates mainly conjugate proteins to polymers for delivering proteins, antibodies, and peptides to enhance their stability, and alert the pharmacokinetics and targeting ability. PEG, a polymer with high water solubility, flexibility, lack of charge, and biocompatibility, is commonly used for polymer-protein conjugation. PEGylation hinders the interaction between the protein and plasma proteins, enzymes, and the phagocytic system, thereby preventing rapid clearance.31,32
Polymer–drug conjugates typically consist of multiple drugs conjugated to a single polymer due to the significantly smaller molecular weight of drugs compared to proteins. As a result, polymers can alter pharmacokinetic properties, improve solubility, and enable controlled release kinetics of the conjugated drugs, thereby promoting both diagnostic and therapeutic performances.33 Functionalizing polymer conjugates with specific bioactive ligands both enhances therapeutic efficacy and reduces side effects on healthy tissues.34 For instance, amino groups of AS1411 aptamers have been conjugated to carboxymethyl chitosan via an esterification reaction, creating a targeted drug delivery system for tumor cells.35
Dendrimers
Dendrimers are hyperbranched, unimolecular, 3D polymeric macromolecules which consist of a central core surrounded by convergent reactive chain-ends, with a readily modifiable surface.36 Thus, dendrimers commonly serve as versatile carriers for small molecule drugs, which can either be physically encapsulated within the dendrimer cavities or chemically conjugated to the surface functional groups, depending on the specific structures and properties of the drugs.37
Additionally, the encapsulation of drug molecules within the internal cavities of dendrimers can significantly enhance the stability of the drugs, protecting against the degradation during blood circulation until they reach the target site.38 Furthermore, dendrimers facilitate site-specific drug delivery through targeting ligands conjugated to their surfaces, thereby minimizing nonspecific toxicity to adjacent tissues.39 Among the various types, the most extensively studied dendrimers are non-biodegradable, cationic, amine-terminated polyamidoamine (PAMAM) dendrimers.40
Polymeric Nanocapsules
Polymeric nanocapsules feature a liquid or solid core enclosed by a polymeric shell.40 With their core-shell microstructure, the drug-loading efficiency can be increased effectively.41 The polymeric shell protects against degradation or burst release caused by factors such as pH, temperature, and enzymatic activity. Furthermore, it can facilitate specific interactions with targeted biomolecules, thereby achieving precise drug delivery.42–44
Common natural polymers used as nanocapsules include polysaccharides, chitosan and protein.45–47 To date, synthetic polymers that have been widely utilized include aliphatic polyesters, Eudragit® polymers and PEG.48 Eudragit® RS100, can effectively neutralize the negative charge of DNA, thus facilitating its transport across cell membranes without causing molecular degradation.49 Furthermore, the incorporation of PEG on the surface of nanocapsules enhance their stability in biological media while simultaneously reducing immunogenicity.50 Hence, the encapsulation of photosensitive drugs, such as desonide and ketoprofen, within the oil core of Eudragit®RL 100 nanocapsules, can effectively prevent photodegradation of the drug under UV radiation.51
By modulating the interactions between cells and the drug, these nanocapsules improve bioavailability compared to free, unloaded drugs.52 Additionally, polymeric nanocapsules offer a reliable delivery mechanism that maintains therapeutic drug concentrations over extended periods, enhancing patient convenience.53
Nanogels
Nanogels are composed of hydrophilic polymeric networks at submicron scales, which allows encapsulation of hydrophilic and lipophilic medicines, DNA sequences, small interfering RNA (siRNA), peptides, and proteins.54–56
Active targeting in nanogels is achieved by conjugating ligands, such as antibodies or aptamers, that specifically bind to biological receptors on target cell surfaces.57 It has also been indicated that cells exposed to nanogels composed of biocompatible natural polymers, including alginate, dextran, pullulan, and hyaluronic acid, exhibit a high survival time in the cellular environment and low toxicity.58–60 Multi-stimuli responsive nanogels are capable of releasing their drug payloads at specific locations by undergoing changes in their configuration, size, and physicochemical properties in response to various stimuli such as pH, temperature, redox conditions, and light.61 For instance, dual temperature/pH-sensitive nanogels have been developed using temperature-responsive poly(N-isopropylacrylamide) P(NIPAAm) and N,N-dimethylaminoethyl methacrylate (DMAEMA), which contains amino groups that exhibit pH-responsive behavior to facilitate the release of anticancer drugs.62 Lian et al synthesized poly(ethylene glycol)-graft-dextran (CDP) nanogels through cross-linking with 3,30-dithiodipropionic acid (DTPA), enabling dual reduction-triggered and pH-responsive drug delivery for cancer therapy.63
Polymeric Micelles(PMs)
Polymeric micelles (PMs) are formed by the spontaneous self-assembly of amphiphilic polymers into nanostructures ranging from 20 to 200 nm in size.64 These micelles are comprised of a hydrophobic inner core for entrapping poorly-water soluble-drugs, and a outer hydrophilic shell for isolating the drug from the surrounding environment.65 The hydrophilic outer surface can be further functionalized with a variety of targeting ligands, such as folate (FOL), monoclonal antibodies (mAb), and monosaccharides (eg, mannose, glucose, and fructose), enabling pH/temperature responsive drug delivery.66
Recently, two or more distinct amphiphilic polymers are commonly combined in micelles.67 Mixed micellar formulations are utilized to achieve better thermodynamic and kinetic stability, enhance drug loading capacity, and provide more accurate size control and incorporation of various modifications.68–70 The derivatives of poly(ethylene oxide)–poly(propylene oxide)-poly(ethylene oxide) (PEO–PPO–PEO) block copolymers are classic amphiphilic materials used for the preparation of polymeric mixed micelles.70 Other relevant amphiphilic macromolecules employed in the construction of PMs include PEG-based molecules such as poly(lactic acid) (PLA), PLGA, and PCL, which have been approved by the US Food and Drug Administration (FDA) for various biomedical applications in humans.71
Polymer Nanoparticles
Polymer nanoparticles are colloidal carriers with nanoscale dimensions. They enable the enhancement of hydrophobic agents delivery, promote an extended circulation and modify the biodistribution of encapsulated therapeutics.72 They also possess greater structural complexity and offer enhanced flexibility through the design of both core and surface components, in contrast to water-soluble dendrimers.
Solid polymer nanoparticles are typically fabricated through precipitation or emulsification, often with the addition of a surfactant. Due to their solid structure, these nanoparticle-based drug carriers provide distinct advantages, including agent encapsulation within the hydrophobic core, higher drug loading capacity, and controlled drug release through diffusion or regulated polymer degradation.73,74
Applications of PNs in the Treatment of Diabetic Wound Healing
The complex pathophysiology of diabetic wounds poses a significant challenge for clinical treatment. The main manifestations include long-term inflammatory reactions, elevated levels of reactive oxygen species, continuous bacterial colonization that often develops into difficult-to-treat biofilms, sustained oxidative stress, and reduced neovascularization under hyperglycemic conditions (Figure 3).8 Polymeric nanomedicines hold significant promise for treating diabetic wounds. As research progresses, PNs are emerging as a novel and effective approach to addressing diabetic wounds and enhancing tissue repair, offering new therapeutic possibilities for managing chronic wounds in diabetic patients (Table 1).
![]() |
Table 1 Summary of Various Polymeric Nanomedicines Used for the Treatment of Diabetic Wound Healing |
![]() |
Figure 3 Scheme illustrating the application of polymeric nanomedicines in the treatment of Diabetic Wound Healing. |
Wound Infection Control
Diabetic patients are especially susceptible to wound infections due to multidrug-resistant organisms(MDROs). The combined use of other wound infection drugs may further exacerbate bacterial resistance to antibiotics.105 In light of these challenges, antibacterial PNs offer a promising therapeutic approach. By loading metal nanoparticles or antibiotics, PNs inhibit resistance and enhance antibacterial effects. The small size and surface modifications enable them to penetrate and disrupt bacterial biofilms while improving antibiotic delivery and reducing chronic infection risks. Additionally, PNs could serve as carriers for antimicrobial agents, prolonging drug release and increasing antibacterial efficacy.
PNs loaded with metal nanoparticles can interfere with bacterial quorum sensing and suppress antibiotic resistance, thereby enhancing antibacterial efficacy.106 Lin and Qu’s team developed a CGH hydrogel to eradicate MDROs and promote diabetic wound healing. The hydrogel is synthesized in situ by crosslinking copper nanoclusters (CuNCs) with oxidized hyaluronic acid (HA-ALD) while simultaneously loading glucose oxidase (GOx).75 The dressing releases CuNCs to catalyze the Fenton reaction, generating hydroxyl radicals(⋅OH) with broad-spectrum antibacterial activity, thereby inhibiting bacterial proliferation. GOx reduces glucose and optimized Fenton reaction to enhance antibacterial effect. Animal studies indicated that this hydrogel, in combination with electrical stimulation, could inhibit bacterial infection, promote angiogenesis, and accelerate wound healing. It can also be applied to irregular wound shapes and establish a sustained sterile environment. Nissren et al innovatively integrate SrO-CoO bimetallic oxide nanoparticles with Guggul gum grafted polyacrylamide hydrogels (SG), developing a hydrogel with enhanced antibacterial properties for wound infection control.76 This hydrogel synergistically utilizes strontium to promote tissue repair and cobalt to induce angiogenesis. It effectively inhibits S. aureus, E. coli, and P. aeruginosa, reducing bacterial colonization in wounds. Additionally, it also exhibits sustained drug release (naproxen: 10%-21%), potentially lowering dressing frequency and infection risk. Furthermore, the hydrogel demonstrates no cytotoxicity to healthy cells, making it a promising biomaterial for chronic diabetic wound care.
Due to the hyperglycemic state, bacterial biofilm (BBF) infections exist in approximately 90% of patients with diabetic wounds, prolonging the inflammatory stage and ultimately leading to wound deterioration.107 PNs provide an advanced approach for combating biofilm infections. Due to their unique size and physiochemical characteristics, they can penetrate and disrupt biofilms, helping to eradicate BBF, eliminate infections, and break this vicious cycle of inflammation, thereby accelerating wound healing. Li’s team used gallic acid-modified chitosan (GC), in which palladium ions coordinate with divalent ions through amino and catechol groups on its side chains, to construct a microenvironment-adaptive nanodecoy (GC@Pd) via an in-situ reduction process mediated by ascorbic acid.77 In the weakly acidic environment of biofilm-associated infections, GC@Pd induces bacterial aggregation and generates ROS and heat through its oxidase-like activity and photothermal effects, thereby synergistically eliminating the BBF. In vivo experiments and transcriptomic analysis confirmed that GC@Pd promotes the shift of diabetic wounds from the inflammatory to the proliferative phase by eradicating biofilm infections and reducing inflammatory responses, providing a promising therapeutic approach for treating biofilm infections in chronic diabetic wounds.
PNs can also assist in improving the utilization of other antibacterial substances. Yoo et al developed polyethylenimine/diazeniumdiolate (PEI/NONOate)-doped PLGA nanoparticles (PLGA-PEI/NO NPs) that can bind firmly to the biofilm matrix to facilitate the delivery of NO to wounds infected with methicillin-resistant Staphylococcus aureus (MRSA) biofilm.78 As an NO donor, these nanoparticles provide a sustained release of NO, extending the release over 4 days, effectively inhibiting bacterial biofilm formation and enhancing wound healing outcomes. PNs can also enhance drug delivery efficacy by loading antibiotics. Yao and Lin et al prepared PLA/SCS/PDA-GS nanofiber membranes, in which the antibiotic gentamicin sulfate (GS) was decorated.79 In vitro studies showed that the GS-loaded nanofiber membrane had efficient antibacterial ability against Staphylococcus aureus.
In addition, PNs can be combined with mechanical methods so as to enhance penetration against biofilm-associated infections. Dai and Ju et al designed an integrated therapeutic and preventive-based nanozyme microneedle (Fe2C/GOx@MNs) for the healing of diabetic wounds that were infected by MRSA biofilm.80 These soluble tips, with sufficient mechanical strength, improve the penetration capability of Fe2C nanoparticles (Fe2C NPs) and GOx for effective biofilm elimination. Meanwhile, the use of a chitosan backing layer provides excellent antibacterial properties, preventing bacterial re-invasion during the wound healing process to a great extent. Most importantly, Fe2C/GOx@MNs demonstrated biofilm clearance and reinfection prevention capabilities in a diabetic mouse model of MRSA biofilm infection, indicating its promising clinical potential in promoting the healing of infected wounds in diabetic patients. Such advancements in nanomedicine provide a novel strategy to combat the risks associated with chronic diabetic wounds, enhancing infection management and tissue regeneration through effective antimicrobial action.
Inflammatory Response Regulation
Chronic inflammation is a key pathological manifestation of diabetic wounds, with the duration and quality of wound healing being closely associated with the severity of the inflammatory response.108 In diabetic patients, hyperglycemia triggers the activation of inflammasomes, leading to sustained expression of M1 macrophages, which in turn perpetuates a prolonged pro-inflammatory state. This chronic inflammatory milieu significantly delays wound healing, impairs tissue regeneration, and increases the risk of infection.109,110
Macrophages, as the key cells in the regulation of wound inflammation, play a critical role in regulating inflammatory responses, removing infections, and promoting tissue repair.111 Therefore, PNs regulate inflammation mainly by influencing macrophage polarization and modulating key inflammatory pathways. Some PNs are designed to shift macrophages from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype, which promotes tissue repair.82 Additionally, PNs can inhibit the NF-κB and MAPK signaling pathways, reducing excessive pro-inflammatory cytokine release (TNF-α, IL-6, IL-1β) and enhancing the expression of anti-inflammatory factors (IL-10, TGF-β), ultimately creating a more favorable wound-healing environment. Liao and Ouyang’s research team developed an injectable chitosan@puerarin (C@P) nanofiber hydrogel for this purpose.81 Puerarin is widely recognized for its anti-inflammatory effects, and when combined with the natural polymer chitosan, it addresses issues such as low hydrophilicity, poor bioavailability, and low permeability in hydrogels with only Puerarin. This combination enhances the effectiveness of diabetic wound recovery. The findings suggest that it can effectively mitigate inflammation and promote wound recovery by inhibiting M1 macrophage polarization, which is mediated by miR-29a/b1. Liu, Mi, and Shahbazi et al discovered that MLN4924, a low-concentration neddylation compound, can suppress the polarization of M1 macrophages. The researchers loaded MLN4924 within PLGA nanoparticles coated with biomimetic macrophage membranes, creating M-NPs/MLN4924. This formulation combines the beneficial properties of polymer nanomaterials with the unique characteristics of macrophage membranes.82 By incorporating M-NPs/MLN4924 into hydrogels and applying them in a diabetic mouse model, it was found to inhibit macrophage polarization into the inflammatory M1 phenotype via receptors on the macrophage membrane, such as Tumor Necrosis Factor Receptor 1(TNFR1), Interleukin-6 Receptor(IL-6R), and Toll-like Receptor 4(TLR4). The promotion of polarization towards the anti-inflammatory M2 phenotype reduced the secretion of inflammatory factors and significantly improved wound repair in diabetic mice, thereby accelerating diabetic wound healing.
Chronic inflammation of diabetic wounds is closely related to inflammatory cytokines and signaling pathways. PNs can inhibit classical inflammatory signaling pathways, including NF-κB, MAPK, JAK-STAT et al,112,113 reducing the production of excessive proinflammatory mediators such as IL-1β, IL-6, TNF-α etc, which in turn lowers excessive inflammatory responses and promotes the release of anti-inflammatory factors (IL-10, TGF-β) which facilitate wound healing. Nabarawi et al developed environmentally friendly zinc oxide nanoparticles (ZnO-NPs) by utilizing Althaea officinalis flowers, which were then integrated into a 2% chitosan (CS) gel to form the A.O-ZnO-NPs CS gel for wound repair.83 In diabetic rat models, the treatment led to a significant reduction in TNF-α, IL-6, and IL-1β expressions, along with an increase in IL-10 levels. The gel, compared to the control group, led to a 1.9-fold increase in the serum levels of anti-inflammatory cytokine IL-10 levels, highlighting its efficacy and superiority in alleviating inflammatory signs. This demonstrates the potential of eco-friendly green synthesis of PNs and its therapeutic approach for facilitating diabetic wound recovery. Zhao et al utilized CS as a carrier to load curcumin, which exhibits a wide range of biological activities, including anti-inflammatory and antioxidant properties, and created polymer nanoparticles Cur-CS-NPs.84 The findings showed that Cur-CS-NPs exhibited sustained drug release and effective cell uptake in the diabetic model, significantly reducing the release of inflammatory factors from macrophages and attenuating local inflammation at the site of the diabetic wound. Liu’s team oxidized hyaluronic acid (HA) and combined it with gelatin (GEL) and cordycepin (COR), followed by modification with dopamine(DA), ultimately forming nanofiber membranes (COR/OHDA/GEL) through electrostatic spinning technique.85 The constructed COR/OHDA/GEL nanofiber membranes significantly reduced the expression of inflammatory factors such as TNF-α, IL-1β, and IL-6 in macrophages by inhibiting the TLR4/NF-κB signal pathway, thereby modulating the inflammatory response to promote diabetic wound healing.
Antioxidative Stress
In diabetic wound healing, the high-glucose environment increases mitochondrial oxygen consumption and impairs its function.114 Sustained elevated blood glucose levels can also lead to excessive protein glycation, resulting in the formation of advanced AGEs, all of which contribute to a high level of ROS.77 The accumulation of ROS can trigger excessive oxidative stress, causing damage in cells responsible for wound healing and disrupt the entire wound healing process. PNs have demonstrated significant antioxidant properties, acting as efficient ROS scavengers to reduce oxidative stress. They can also possess inherent antioxidant capabilities, protecting cells from damage. Additionally, PNs enhance mitochondrial function, supporting energy production, and promote an increased local oxygen supply, creating a more favorable environment for wound healing. These mechanisms collectively improve wound healing outcomes by reducing oxidative damage and facilitating tissue regeneration.
ROS Scavenging
During the healing process of diabetic wounds, oxidative stress activated by the abnormal accumulation of ROS is one of the main factors leading to difficult wound healing. Utilizing antioxidative enzymes is one of the effective strategies for scavenging ROS.115 PNs can mimic enzymatic activities to scavenge ROS, thereby reducing oxidative stress-related damage. Wang and Liu et al developed a multifunctional hydrogel named MOF/CGA@GP-CS(MCGC) by constructing metal–organic framework (MOF)-nanozymes anchored with the natural antibacterial agent chlorogenic acid(CGA) in genipin-crosslinked chitosan hydrogels.86 These nanozymes exhibit catalase-like activity, converting excess H₂O₂ in wounds into dissolved oxygen. This not only effectively removes ROS but also generates oxygen at the wound site. This approach addresses the limitations of natural enzymes, such as fragility and high cost, thereby enhancing wound healing outcomes. In vitro experiments confirmed that MCGC reduced ROS levels in high-glucose induced bacterial infection and increased antioxidative enzyme activity, significantly lowering oxidative stress markers and regulating oxidative stress responses.
Prussian Blue Nanoparticles (PBNPs) display remarkable ROS scavenging capabilities, mimicking the activities of catalase (CAT), peroxidase (POD), and superoxide dismutase (SOD), thereby protecting mitochondria from the damage associated with oxidative stress. Chen et al developed a thermosensitive poly (d,l-lactide)-poly(ethylene glycol)-poly(d,l-lactide) (PDLLA-PEG-PDLLA) hydrogel (PLEL)-based wound dressing which is loaded with PBNPs.87 After injecting PBNPs@PLEL into the site of injury, PBNPs could release slowly, maintaining a sustained antioxidant activity. Both in vitro and in vivo investigations revealed that PBNPs@PLEL effectively eliminate ROS, reduce cellular damage, protect mitochondrial function, and preserve the endogenous NRF2/HO-1 antioxidant signaling pathway, thereby promoting diabetic wound healing.
Antioxidant
In response to oxidative stress during diabetes, PNs can serve as stable, sustained-release antioxidants to regulate redox balance. Chen and Ren et al synthesized novel tea polyphenol nanospheres (TPN) and encapsulated them in a PVA/alginate hydrogel (TPN@H) to solve the issue of green tea polyphenols(TP) being easily oxidized, providing a gel material that can prevent oxidation.87 In diabetic rat models, TPN@H promoted collagen deposition and maturation, as well as the formation of granulation tissue, to a greater extent compared to the control group. TPN@H also facilitated wound healing and regulated immune responses. Furthermore, TPN@H helped regulate the PI3K/AKT signaling pathway to promote diabetic wound healing.
Mitochondrial Function Improvement
Excessive ROS are not only a result of mitochondrial dysfunction but also cause further damage to the mitochondria.116 PNs can reduce the production of ROS by improving mitochondrial function, thereby inhibiting the sources of ROS generation. Guo and Tao’s team developed a double-network hydrogel (FH-M@S), constructed with pluronic F127 diacrylate (F127DA) and hyaluronic acid methacrylate (HAMA), enhanced by mesoporous polydopamine nanoparticles (MPDA NPs) loaded with SS31, a mitochondrial-targeting peptide that attaches to the inner mitochondrial membrane to maintain mitochondrial function and reduce mitochondrial ROS production.89 This gel exerts a synergistic effect on full-thickness wound healing under diabetic conditions through near-infrared photothermal antibacterial action and mitochondrial maintenance. In in vivo experiments, FH-M@S consistently demonstrated optimal wound closure effects.
Resveratrol (RES) exhibits antioxidative properties, particularly in mitochondrial protection. Wang et al an injectable, light-curable silk-based nanocomposite hydrogel (SS/MPDA@RES) by integrating RES-loaded mesoporous polydopamine nanoparticles (MPDA) into silk microfibers.90 It was found that through direct injection and in situ visible light treatment, SS/MPDA@RES markedly promoted wound healing in diabetic rats. This gel demonstrated effective scavenging of excessive ROS, thereby safeguarding mitochondrial function, restoring ATP production, and rebalancing redox homeostasis.
Increased Topical Oxygen Supply
Increasing the topical oxygen supply to the wound can enhance cellular aerobic metabolism, leading to more ATP for cellular energy and increasing the activity of antioxidative enzymes to scavenge ROS and reduce oxidative stress.117 Currently, there is a growing focus on developing PNs to enhance topical oxygen supply for improving wound oxygenation and facilitating the healing of diabetic wounds. Muhammad et al developed oxygen-generating polymeric nanofibers based on PCL, loaded with inorganic sodium percarbonate (SPC) salt that can chemically generate oxygen in situ.91 The results indicated that PCL-SPC wound dressing can continuously produce oxygen at the wound site for up to 10 days. Experiments using the chorioallantoic membrane assay demonstrated that this oxygen-releasing dressing stimulates angiogenesis and shows great potential for wound healing in diabetic rat models. Han et al utilized charged quaternized chitosan(QCS) and hyaluronic acid to layer black phosphorus(BP) nanosheets and hemoglobin(Hb) onto electrospun poly-l-lactide(PLLA) nanofibers, creating a sequence of multifunctional wound dressings (coded as PQBH-n).92 Both in vitro and in vivo studies confirmed their excellent abilities in facilitating wound healing. These PN-based dressings, combined with near-infrared (NIR)-assisted oxygen delivery, enable on-demand oxygen release, effectively improving the hypoxic microenvironment of wounds.
PNs offer significant advantages in mitigating oxidative stress, with high stability and prolonged drug circulation time. This enhances their ability to regulate oxidative stress responses, thereby promoting a more favorable microenvironment for tissue regeneration and accelerating the healing process.
Acceleration of Angiogenesis
In diabetic wounds, PNs can promote the generation of cells and growth factors, as well as facilitate drug delivery, thereby accelerating angiogenesis. They promote fibroblast proliferation, migration, and extracellular matrix deposition, all of which are essential for vascular regeneration. Additionally, PNs regulate the sustained release of key pro-angiogenic growth factors to enhance endothelial cell proliferation and stabilize newly formed blood vessels. Some polymer-based delivery systems ensure the continuous release of these factors at the wound site, improving endothelial cell proliferation and new capillary formation.
Jain and Dandekar et al prepared starch-based nanofibrous scaffolds by electrospinning starch and polyvinyl alcohol (PVOH) in a 30:70% w/w ratio.93 Cellular assays with L929 mouse fibroblast cells indicated that the scaffolds promoted faster growth of dermal cells, thus accelerating vascular regeneration. PNs can also promote the synthesis of fibroblasts and their deposition in the extracellular matrix, which is crucial to angiogenesis. Zou, Li, and Zheng et al incorporated turmeric-derived nanoparticles (TDNPs) into a permeable aerogel(AG) made from cellulose nanofibers and sodium alginate, developing a wound dressing named TDNPs@AG.93 This dressing enhances fibroblast proliferation and migration by activating the Nrf2/HO-1 signaling pathway, promoting beneficial interactions between macrophages and fibroblasts that increase the formation of extracellular matrix and skin tissue remodeling, thereby accelerating vascular regeneration.
In addition, PNs are capable of facilitating the release of growth factors, including vascular endothelial VEGF, fibroblast growth factor (FGF) and platelet-derived growth factor (PDGF), all of which accelerate angiogenesis and facilitate wound healing.118 Préat et al encapsulated VEGF in PLGA nanoparticles (PLGA-VEGF NP) for treatment of diabetic wounds. Moreover, lactate, a degradation product of PLGA, can enhance neovascularization.118 Under the sustained combinations of VEGF and lactate, wounds in diabetic rats under treatment of PLGA-VEGF NP demonstrated higher collagen content and re-epithelialization, contributing to angiogenesis and wound healing. Chen et al developed a chitin whisker (CW)/carboxymethyl chitosan nanoparticles (CMCS NPs)/thermosensitive hydroxybutyl chitosan (HBC) composite hydrogel (CW/NPs/HBC-HG), which encapsulates recombinant human epidermal growth factor (rhEGF).118 Prolonged cell proliferation was observed for up to 5 days within this dressing. When applied to the wound, rhEGF is gradually released into the gel network, extending the duration of EGF penetration into the wound. In a diabetic rat model of chronic wound healing, this dressing accelerated re-epithelialization, collagen deposition and angiogenesis.
In the past few years, research on exosomes in the treatment of diabetic wounds has gathered significant attention. Guo and Hu’s team engineered PEG/Ag/CNT-M+E hydrogel loaded with exosomes and metformin (MET). Within the gel, hydroxyl-modified multiwalled carbon nanotubes act as the conductive material, establishing hydrogen bonds with thiol, resulting in a stable 3D structure for exosomes and MET.119 This 3D network, characterized by a highly interconnected porous structure, facilitates drug release and delivery, maintaining the integrity of microvessels and barrier function while promoting cell proliferation and angiogenesis. Compared to the relatively short half-life of exosomes, the fusion with PNs exhibit a higher stability with slower degradation and prolonged drug release.
Additionally, PNs can be utilized for gene therapy to achieve targeted treatment. Harmon et al presented a method of gene delivery to diabetic wounds utilizing polymeric chitosan to deliver a plasmid encoding human CA5-HIF-1α, a degradation resistant form of HIF-1α.98 These HIF/CPs nanoparticles enhanced the stability of the human CA5-HIF-1α-encoding plasmid in high-glucose environments, prevented oxygen-dependent degradation by prolyl hydroxylases, and induced an increased number of CD31+ vessel structures in healed tissue, thereby promoting angiogenesis at the wound site and improving wound healing.
In summary, PNs enhance angiogenesis in diabetic wound healing by promoting fibroblast proliferation, migration, and extracellular matrix deposition, all of which are critical for vascular regeneration. PNs also stimulate the sustained release of growth factors such as VEGF, FGF, and PDGF, which further accelerate angiogenesis and tissue repair. Additionally, PNs enable the stable delivery of therapeutic plasmids in high-glucose environments, enhancing gene therapy efficacy and promoting angiogenesis at the wound site.
Glycemic Control
In diabetic wounds, hyperglycemia-induced cytotoxicity directly impedes the wound healing process. A high tropical glucose environment can lead to rapid bacterial growth, induce persistent inflammation, and hinder angiogenesis, with the severity positively correlated with the duration of hyperglycemia exposure.120 Therefore, regulation of the hyperglycemic microenvironment is crucial. Due to the complexity of the diabetic wound microenvironment, conventional glycemic therapeutic options are often limited in effectiveness.121 PNs present a novel and effective approach for blood glucose control in diabetic wounds. They not only help regulate the hyperglycemic environment but also modulate local glucose concentrations within the wound, thereby promoting wound healing.
Topical application of insulin can reduce topical glucose levels and improve diabetic wound recovery. However, its utilization is often hindered by the lack of an appropriate carrier capable of consistently and efficiently delivering insulin to the wound site.122 PNs can serve as a carrier while addressing the issues of short half-life and insufficient biological activity of insulin in the skin. Abdelkader et al constructed a poly(vinyl alcohol) (PVA)-borate hydrogel that contains human insulin encapsulated in PLGA nanoparticles, with 33.86 μg insulin loaded per milligram.123 By comparing the wound healing rates with applied free insulin and that with nano-encapsulated insulin in diabetic rats to their controls, it was found that after 10 days of experiment, the percentage of wound injury indices with insulin-PLGA NP and free insulin were 29.15 and 12.16% respectively. Li et al developed a functionalized silk fibroin (SF) dressing with sustained bioactive insulin release for injury healing in patients with diabetes.99 By encapsulating insulin within the inner layer of SF microparticles, they created a system ensuring a sustained insulin release for a duration of one month without altering the insulin’s original molecular conformation and native bioactivity. This approach not only helps to maintain the bioactivity of the insulin but also improves its stability.
PNs can also enhance wound healing and reduce the likelihood of amputation by loading GOx to degrade excessive glucose at the wound site.100 Li, Yang, and Jiang et al developed a holistic therapeutic nanozyme system(AHAMA/CS-GOx@Zn-POM) that integrated an aldehyde and methacrylic anhydride-modified hyaluronic acid hydrogel (AHAMA) and chitosan nanoparticles (CS NPs), combining GOx with zinc-based polymetallic oxonate nanozyme(Zn-POM) to modulate the hyperglycemic conditions and reprogram the immune microenvironment.101 As GOx oxidizes glucose, hydrogen peroxide and gluconic acid are produced, helping to degrade excessive glucose at the wound site, maintaining a local blood glucose homeostasis, and mitigating the detrimental effects of the hyperglycemic microenvironment on healing.
The utilization of PNs in glycemic control can improve the bioavailability and safety of hypoglycemic agents, aiding in the control of baseline blood glucose levels and preventing further wound infections.124 Zahedi et al synthesized crosslinked carboxymethyl chitosan nanoparticles (CMCS NPs) that contain metformin.102 These CMCS NPs enable sustained release of MET through the degradation of the biopolymer, effectively exerting glucose-lowering effects. In diabetic rats, CMCS NPs inhibited weight loss, reduced blood glucose levels, and promoted the regeneration of pancreatic islets. PNs can also optimize the administration of insulin and help maintain the blood glucose homeostasis in diabetic patients. Victoria C et al investigated an oral insulin nanoformulation using insulin-conjugated silver sulfide quantum dots coated with chitosan/glucose polymer (CS/GS). The formulation showed a dose-dependent effect and marked sensitivity to hydrolytic enzymes, particularly β-glucosidase, which triggers the degradation of CS/GS to release insulin.103 Notably, insulin is released only when blood glucose levels are low, reducing the occurrence of hypoglycemic episodes while enhancing the bioavailability of insulin.125 This creates favorable conditions for wound healing.
PNs can also be used externally to regulate hyperglycemia. Lee’s team developed a hydrogel nanofilm caging system (GC/HA@GEL) via a tyrosinase-mediated enzymatic reaction, encapsulating glycol chitosan(GC) and HA.104 It was found that GC/HA@GEL effectively encapsulates pancreatic β-cells, playing a vital role in regulating glucose homeostasis.
Smart Wound Care Approach
With the advancement of PN technology, researchers are striving to develop smart therapeutic measures that offer continuous blood glucose monitoring and enhanced drug delivery efficacy (Table 2). This will become a significant component of diabetes wound management in the future.
![]() |
Table 2 Description of Various Smart Wound Care Approach with Polymeric Nanomedicines |
Smart Responsive PNs
Smart responsive PNs can respond to specific external stimuli or internal environmental changes, including temperature, light, and magnetic fields, as well as internal changes such as pH and glucose enzyme levels. These responses help activate cellular activity, promote drug release and monitor wound conditions, thereby maximizing their therapeutic efficacy.
Polymers with phenylboronic acid (PBA) are common glucose-responsive materials. PBA can form reversible boronated ester bonds with the cis-diol structure in glucose molecules, which gives PBA high selectivity and sensitivity to glucose,134 resulting in glucose-responsive PNs. Wu et al combined PBA-modified hyaluronic acid with polyethylene glycol diacrylates (PEG-DA) to develop a novel hybrid hydrogel (PEG-DA/HA-PBA).134 This hydrogel, loaded with the highly antioxidative myricetin (MY), enabled glucose-triggered MY release and effectively scavenged ROS(>80.0%), thereby remodeling the oxidative environment of wounds.
The topical temperature of diabetic wounds may vary due to factors such as inflammatory responses. Polymers based on N-isopropylacrylamide (NIPAM) can be prepared as temperature-responsive PNs to sense changes in wound temperature and release drugs accordingly.135 Bao’s team dispersed polydopamine nanoparticles into methacrylated gelatin and NIPAM monomers, loading them with the drug linagliptin, to create a temperature-sensitive hydrogel(LIN@PG10@PDA-NIR). The drug release rate was controlled through near-infrared laser irradiation.127 This hydrogel exhibits excellent thermosensitive and photothermal properties, promoting cell migration and the expression of angiogenic factors for diabetic wound healing. Zhong et al encapsulated Fe3O4@SiO2 particles with MXene, which has excellent near-infrared absorption capabilities, to form MNPs@MXene. These particles were combined with silver nanoparticles and loaded into a poly(N-isopropylacrylamide) (PNIPAM) and alginate dual-network hydrogel system. PNIPAM is a temperature-sensitive polymer that dehydrates and contracts below its lower critical solution temperature.128 Under the influence of near-infrared light and an alternating magnetic field, temperature within the system rapidly increases, allowing for controlled release of AgNPs. This approach demonstrated promising therapeutic effects in subcutaneous infected wounds in a diabetic rat model.
The pH of normal skin generally ranges from 4 to 6, while diabetic wounds tend to be more alkaline, with pH values between 7 and 9.77 The pH value can serve as a real-time indicator to monitor the progression of wound healing in diabetes, making the development of pH-responsive PNs crucial for optimal therapeutic effects. Tian and Sun et al created a pH-responsive dressing (Cell-An/PCL-Ch) consisting of a pH-sensitive, hydrophilic nanofibrous layer and an antibacterial, hydrophobic PCL bottom layer.129 This dressing can continuously monitor pH changes in the exudate of diabetic wounds. Real-time pH monitoring can be achieved through a program integrated into smartphones, simplifying the medical care of diabetic wounds.
ROS-responsive biomaterials can be triggered by ROS in the injured tissue to release drugs that modulate ROS levels, alleviate oxidative stress, and promote tissue regeneration.136 Liang et al synthesized a hyaluronic acid-based ROS-responsive composite multifunctional wound dressing (HA@Cur@Ag), which incorporated curcumin liposomes and silver nanoparticles (AgNPs). Experimental validation showed that the HA@Cur@Ag hydrogel effectively modulated the oxidative stress response in diabetic wounds and promoted tissue repair. Transcriptomic sequencing revealed that this dressing significantly inhibited the activation of the TNF/NF-κB signaling pathway, reducing oxidative stress and inflammation in diabetic wounds.137 The ROS-responsive dressing also controlled the release of curcumin liposomes and silver nanoparticles, demonstrating antioxidative, antibacterial, and anti-inflammatory properties.
To further enhance therapeutic efficacy, multiple responsive polymeric nanomedicines have been designed. By integrating multiple response mechanisms, PNs can adapt to the complex environment of wound, enabling precise drug release and synergistic therapy. Xue and Shang et al constructed self-adaptive hydrogels (AuNCs@PBA-Sa) based on marine-derived gold clusterzyme (AuNCs). Marine mussel-derived L-3,4-dihydroxyphenylalanine (DOPA) with intrinsic antioxidative properties was used as the functional ligand to prepare AuNCs with SOD-like activity. This ligand forms boronate ester bonds with phenylboronic acid-modified marine-derived sodium alginate(PBA-Sa), imparting ROS/glucose responsiveness to AuNCs@PBA-Sa.97 This allows the dressing to respond to degradation and control the release of AuNCs. Additionally, it significantly enhances the ability of AuNCs to remove free radicals and exhibits excellent SOD-like enzyme activity, efficiently regulating the pathological conditions associated with chronic wounds, such as oxidative stress, immune dysregulation, and excessive inflammation. Luo et al developed a polytrimethylene carbonate (PTMC)/polyvinylpyrrolidone (PVP) nanofibrous dressing (C-PPZS) which loaded with simvastatin-loaded ZIF-8 nanoparticles (ZIF-8@SIM NPs) for wound healing. This dressing responds to multiple stimuli, including the release of Zn2+ and SIM in acidic environments, mechanical contraction induced by liquid, and enhanced contractility with temperature elevation.138 The results indicated that C-PPZS facilitated wound healing, making it an effective strategy for chronic wound management.
In addition to the above-mentioned response mechanisms, there are other mechanisms such as hypoxia response, H2O2 response, and ultrasound response,130,139–141 that require further research. Developing more efficient and safer multiple responsive PNs offers new hope for wound treatment in diabetic patients.
Integrated Monitoring and Management
Diabetic wounds are characterized by dynamic changes, and their healing process is affected by multiple factors, including blood glucose levels, infection, inflammatory response, and vascular lesions. Moreover, most wound care relies on visual judgment, which is often not consistent with real-time monitoring, management, and dynamic treatment.142 The integrated diagnosis and treatment provided by PNs offer a viable solution to the challenge. Real-time monitoring of wound conditions enables timely and targeted interventions, thereby facilitating the healing of diabetic wounds.
Dai et al developed an intelligent lipoic acid-modified chitosan hydrogel (LAMC/CD-C@M@P) for pH monitoring and accelerated healing of diabetic wounds. The carbon quantum dots (CDs) in the hydrogel exhibit stable photoluminescence properties that are pH-dependent, allowing the dressing to monitor pH levels. The ceria oxide-molybdenum disulfide nanoparticles with a polydopamine layer (C@M@P) provide photothermal antibacterial effects.132 When an alkaline environment is detected, likely indicating bacterial infection, the dressing utilizes its photothermal and near-infrared assisted antibacterial properties to address the issue. Additionally, it effectively scavenges ROS, which alleviates inflammatory responses and reduces oxidative stress, thereby promoting the repair of diabetic wounds. Jiang, Yi, and Haick et al combined advanced polymer nanomaterials with electro-controlled devices to develop a wireless wound management system. This system consists of a customized smartphone app, wearable bioelectronics, and a theragnostic patch (Thera-patch). The patch includes pH and glucose sensors, allowing for continuous monitoring of the status of diabetic wounds, and provides personalized and precise treatment through iontophoresis and electrical stimulation based on the wound conditions.133 It enables a closed-loop drug delivery while monitoring multiple wound-related biomarkers, improving real-time monitoring and targeted treatment of diabetic wounds, ultimately facilitate wound healing.
With the in-depth research of polymer nanomedicine, real-time monitoring for patients with diabetic wounds can be established, enabling more targeted therapeutic strategies and bringing new hope for the full recovery of diabetic wounds.
Future Perspectives
Although many studies have shown the potential of PNs for diabetic wounds, there are still limitations to address in future research.
Safety
The regulation of nanomedicines is an important bottleneck in their clinical translation. Due to the unique nature of nanomaterials, it is difficult to fully apply traditional drug regulatory frameworks. For example, several guidelines have been issued by organisations such as the FDA and EMA, emphasising quality control, non-clinical safety and pharmacokinetic studies of nanomedicines.5 Some PNs materials are still in animal experimentation stage, and their effects on humans have not been fully examined. To date, the United States Food and Drug Administration has approved only a liposome-like nanoformulation of insulin, a hepatic-directed vesicle insulin, for the management of diabetic wounds. However, this formulation has not been commercialized due to multiple adverse reactions observed in clinical trials.143 Additionally, most PNs tend to accumulate mainly in the liver, spleen, or kidneys, posing potential toxicity risks to the human body.144–146 Therefore, it is essential to enhance the loading capacity and encapsulation efficiency of PNs and investigate their specific metabolic pathways via surface modification and targeting ligands to reduce the non-specific accumulation of PNs and minimize collateral metabolites.147,148
Some nanomaterials may exhibit cytotoxicity at high concentrations.149 To address this, Zhang et al designed a “microcage” based on neutrophil extracellular traps (NETs) to improve diabetic wound healing by integrating methacryloylgel (GelMA) hydrogel microspheres with cationic polyethylenimine (PEI)-functionalized mesoporous polydopamine (mPDA), named mPDA-PEI@GelMA. This design enables the removal of NETs from nanoparticles and diabetic wound surfaces in a non-contact manner, effectively reducing chronic diabetic wound-related pro-inflammatory responses, enhancing wound healing processes, minimizing biotoxicity, and ensuring high biosecurity.150
In future studies, it is essential to conduct large-scale clinical trials to assess the safety and efficacy of PNs for the treatment of diabetic wounds. Improving the biocompatibility of PNs while reducing their potential cytotoxicity through surface modification, functionalization, or novel biodegradable materials is crucial for their clinical application.151
Stability
The stability of polymeric nanomedicines directly affects their efficacy and safety. Nanoparticles may lose their function due to physicochemical changes (eg, aggregation, degradation) during storage and transport. For example, liposomal nanodrugs are prone to drug leakage or lipid oxidation during long-term storage, which affects their stability.152 Moreover, the interactions between various bioactive chemicals loaded on PNs are not well understood, particularly in terms of drug mass loading ratios, non-specific binding, aggregation phenomena, and the potential loss of biological activity in clinical applications. For instance, Krishna Yadav’s research group developed a temperature-responsive self-shrinking nanofiber/hydrogel wound dressing. However, the temperature-sensitive response of this material is prone to being influenced by fluctuations in external temperature.153 Therefore, future PN development should focus on studying the interaction mechanisms between PNs and bioactive substances, including intermolecular forces, hydrogen bonding, and hydrophobic interactions, through molecular dynamics simulation and quantum chemical calculations. This will clarify the interaction mechanisms and improve PN stability in diabetic wound treatments.154,155
Erigi et al employed PRISM theory in conjunction with molecular dynamics simulations to investigate the structure and phase diagram of nanorods at a 1% volume fraction within a polymer melt. Through a quantitative comparison of the resulting phase diagrams, both methods revealed that the formation of contact aggregates under conditions of low polymer-nanorod attraction (γ), and bridging aggregates when the attraction was higher.156 The stability of polymers and nanorod formation composites is different in γ, which suggests new directions for PN preparations in diabetic wound treatment.
Cost
The research and development process of PNs, such as designing, synthesizing, testing, and optimizing recipes, involve highly specialized equipment and technicians, which increases overall costs. Moreover, the raw materials required for PN formulations are often more expensive than traditional materials, especially if they contain rare or high-purity materials. The commercialization of PNs is also subject to strict regulations and standards, requiring additional testing and documentation, further driving up costs. Additionally, personalized PN formulations for patients significantly increases production costs due to the need for special parameters.156 Mahmoudi et al proposed that due to the differences in physiological and immune responses between men and women, gender-specific factors should be considered when designing PNs for the treatment of diabetic wounds to improve treatment efficacy.157
Future research could focus on simplifying the synthesis process of PNs to reduce production time and cost. Optimizing synthesis conditions and parameters will promote the application of PNs in diabetic wound healing.90,101,158
Validity
Passive skin administration poses challenges due to the self-protection function of human skin. Future advancements in PNs should focus on integrating smart, multifunctional nanomedicine strategies to enhance their therapeutic potential. One promising direction is the development of multi-stimuli-responsive PNs, which can react to wound-specific conditions such as pH, temperature, glucose levels, and oxidative stress to achieve on-demand drug release. These adaptive systems can further enhance therapeutic efficacy while minimizing off-target effects. Another key research avenue is the combination of PNs with regenerative medicine, including gene therapy, stem cell therapy, and 3D bioprinting.159–161 PNs can serve as carriers for genetic materials, growth factors, or exosomes, promoting cellular proliferation, tissue remodeling, and neovascularization in chronic wounds. Additionally, integrating PNs with real-time monitoring systems could enable the development of wearable wound dressings, allowing continuous tracking of wound conditions and dynamic drug delivery.
Future studies may explore the interaction between electronic signals and biological signals to improve the healing progress of wounds.162 Additionally, to better understand the key biomarkers of diabetic wounds, advanced imaging technologies and bioanalytical methods can be used to explore subcellular models.163 This will enable tracking the distribution, metabolism, and excretion pathways of PNs in vivo and target subcellular-scale indicators.
Conclusions
Diabetic wound healing is a multifaceted process influenced by hyperglycemia, infections, inflammation, oxidative stress, and vascular complications. PNs have emerged as a promising approach due to their unique physicochemical properties and excellent biocompatibility, enabling enhanced anti-inflammatory, antioxidant, pro-angiogenic, and antimicrobial effects. Particularly, their antibacterial action plays a crucial role in combating multidrug-resistant infections, facilitating biofilm removal, ultimately accelerating wound healing and lowering amputation risk. By leveraging their nanoscale characteristics and tailored drug delivery capabilities, PNs offer enhanced bioavailability, targeted action, and prolonged therapeutic effects, improving therapeutic efficacy and combating antibiotic resistance. PNs hold significant potential for advancing patient care and clinical outcomes.
Future studies should focus on developing novel PNs with multiple biomaterials while enabling smart wound care in response to specific stimuli. The field of multiple PNs or combining with alternative materials for integrated therapeutic effects, while maintaining biocompatibility, stability, and functionality, remains a key priority. Rigorous preclinical and clinical trials are essential to validate their efficacy and safety, paving the way for their adoption in diabetic wound management. Addressing these gaps will enhance wound healing and drive advancements in nanomedicine for chronic wound care.
Funding
This work was supported by National Natural Science Foundation of China (No.82374380), and Sanming Project of Medicine in Shenzhen(No.SZZYSM202202010).
Disclosure
The authors report no conflicts of interest in this work.
References
1. Taylor R. Understanding the cause of type 2 diabetes. Lancet Diabetes Endocrinol. 2024;12(9):664–673. doi:10.1016/S2213-8587(24)00157-8
2. Lo ZJ, Surendra NK, Saxena A, Car J. Clinical and economic burden of diabetic foot ulcers: a 5‐year longitudinal multi‐ethnic cohort study from the tropics. Int Wound J. 2021;18(3):375–386. doi:10.1111/iwj.13540
3. McDermott K, Fang M, Boulton AJM, Selvin E, Hicks CW. Etiology, epidemiology, and disparities in the burden of diabetic foot ulcers. Diabetes Care. 2023;46(1):209–221. doi:10.2337/dci22-0043
4. Miranda C, Da Ros R, Marfella R. Update on prevention of diabetic foot ulcer. Arch Med Sci Atheroscler Dis. 2021;6(1):e123–e131. doi:10.5114/amsad.2021.107817
5. Kilic M, Olgun N, Dündar M, et al. Prevalence, risk level and risk factors of diabetic foot ulcer among adult individuals with diabetes in the Southeastern Anatolia Region of Turkiye. J Tissue Viability. 2025;34(1):100839. doi:10.1016/j.jtv.2024.12.003
6. Jodheea-Jutton A, Hindocha S, Bhaw-Luximon A. Health economics of diabetic foot ulcer and recent trends to accelerate treatment. Foot. 2022;52:101909. doi:10.1016/j.foot.2022.101909
7. Olszewska A, Duan J, Javorovic J, et al. Manufacture and initial characterisation of RAPIDTM biodynamic haematogel, an autologous platelet and leukocyte-rich plasma gel for diabetic foot ulcers. Gels. 2024;10(9):572. doi:10.3390/gels10090572
8. Armstrong DG, Tan TW, Boulton AJM, Bus SA. Diabetic foot ulcers: a review. JAMA. 2023;330(1):62. doi:10.1001/jama.2023.10578
9. Armstrong DG, Boulton AJM, Bus SA. Diabetic foot ulcers and their recurrence. N Engl J Med. 2017;376(24):2367–2375. doi:10.1056/NEJMra1615439
10. Normandin S, Safran T, Winocour S, et al. Negative pressure wound therapy: mechanism of action and clinical applications. Semin Plast Surg. 2021;35(03):164–170. doi:10.1055/s-0041-1731792
11. Zheng SY, Wan XX, Kambey PA, et al. Therapeutic role of growth factors in treating diabetic wound. World J Diabetes. 2023;14(4):364–395. doi:10.4239/wjd.v14.i4.364
12. Mudgal SK, Kumar S, Sharma SK, et al. A network meta-analysis of randomized controlled trials on the comparative efficacy of stem cells therapy for diabetic foot ulcer healing. Int J Lower Extremity Wounds. 2024:15347346241286216. 10.1177/15347346241286216
13. Shi S, Hu L, Hu D, Ou X, Huang Y. Emerging nanotherapeutic approaches for diabetic wound healing. IJN. 2024;19:8815–8830. doi:10.2147/IJN.S476006
14. Huang F, Lu X, Yang Y, et al. Microenvironment‐based diabetic foot ulcer nanomedicine. Adv Sci. 2023;10(2):2203308. doi:10.1002/advs.202203308
15. Pinto S, Viegas J, Cristelo C, et al. Bioengineered nanomedicines targeting the intestinal fc receptor achieve the improved glucoregulatory effect of semaglutide in a type 2 diabetic mice model. ACS Nano. 2024:
16. Yan Y, Cai H, Yang M. The application of nanotechnology for the diagnosis and treatment of endocrine disorders: a review of current trends, toxicology and future perspective. IJN. 2024;19:9921–9942. doi:10.2147/IJN.S477835
17. Li J, Liu Y, Geng K, Lu X, Shen X, Guo Q. ROS-responsive nanoparticles with antioxidative effect for the treatment of diabetic retinopathy. J biomater Sci Poly ed. 2024;1–22. doi:10.1080/09205063.2024.2406628
18. Xie S, Huang K, Peng J, et al. Self‐propelling nanomotors integrated with biofilm microenvironment‐activated NO release to accelerate healing of bacteria‐infected diabetic wounds. Adv Healthcare Materials. 2022;11(19):2201323. doi:10.1002/adhm.202201323
19. Sorg H, Sorg CGG. Skin wound healing: of players, patterns, and processes. Eur Surg Res. 2023;64(2):141–157. doi:10.1159/000528271
20. Sharifiaghdam M, Shaabani E, Faridi-Majidi R, De Smedt SC, Braeckmans K, Fraire JC. Macrophages as a therapeutic target to promote diabetic wound healing. Mol Ther. 2022;30(9):2891–2908. doi:10.1016/j.ymthe.2022.07.016
21. Edgar L, Akbar N, Braithwaite AT, et al. Hyperglycemia induces trained immunity in macrophages and their precursors and promotes atherosclerosis. Circulation. 2021;144(12):961–982. doi:10.1161/CIRCULATIONAHA.120.046464
22. Eming SA, Wynn TA, Martin P. Inflammation and metabolism in tissue repair and regeneration. Science. 2017;356(6342):1026–1030. doi:10.1126/science.aam7928
23. Holl J, Kowalewski C, Zimek Z, et al. Chronic diabetic wounds and their treatment with skin substitutes. Cells. 2021;10(3):655. doi:10.3390/cells10030655
24. Nass N, Vogel K, Hofmann B, Presek P, Silber RE, Simm A. Glycation of PDGF results in decreased biological activity. Int J Biochem Cell Biol. 2010;42(5):749–754. doi:10.1016/j.biocel.2010.01.012
25. Li H, Meng Y, He S, et al. Macrophages, chronic inflammation, and insulin resistance. Cells. 2022;11(19):3001. doi:10.3390/cells11193001
26. Feng Y, Chen L, Luo Q, Wu M, Chen Y, Shi X. Involvement of microRNA-146a in diabetic peripheral neuropathy through the regulation of inflammation. DDDT. 2018;12:171–177. doi:10.2147/DDDT.S157109
27. Okonkwo U, DiPietro L. Diabetes and wound angiogenesis. IJMS. 2017;18(7):1419. doi:10.3390/ijms18071419
28. Rowe RC, Sheskey PJ, Owen SC. Handbook of Pharmaceutical Excipients.
29. Kim JK, Kim HJ, Chung JY, Lee JH, Young SB, Kim YH. Natural and synthetic biomaterials for controlled drug delivery. Arch Pharmacal Res. 2014;37(1):60–68. doi:10.1007/s12272-013-0280-6
30. Zhu X, Anquillare ELB, Farokhzad OC, Shi J. Chapter 22 - polymer- and protein-based nanotechnologies for cancer theranostics. In: Chen X, Wong S, editors. Cancer Theranostics. Oxford: Academic Press; 2014:419–436. doi:10.1016/B978-0-12-407722-5.00022-0
31. Guo H, Mi P. Polymer–drug and polymer–protein conjugated nanocarriers: design, drug delivery, imaging, therapy, and clinical applications. WIREs Nanomed Nanobiotechnol. 2024;16(4):e1988. doi:10.1002/wnan.1988
32. Knop K, Hoogenboom R, Fischer D, Schubert US. Poly(ethylene glycol) in drug delivery: pros and cons as well as potential alternatives. Angew Chem Int Ed. 2010;49(36):6288–6308. doi:10.1002/anie.200902672
33. Wen P, Ke W, Dirisala A, Toh K, Tanaka M, Li J. Stealth and pseudo-stealth nanocarriers. Adv Drug Deliv Rev. 2023;198:114895. doi:10.1016/j.addr.2023.114895
34. Yameen B, Choi WI, Vilos C, Swami A, Shi J, Farokhzad OC. Insight into nanoparticle cellular uptake and intracellular targeting. J Control Release. 2014;190:485–499. doi:10.1016/j.jconrel.2014.06.038
35. Rață DM, Cadinoiu AN, Atanase LI, et al. “in vitro” behaviour of aptamer-functionalized polymeric nanocapsules loaded with 5-fluorouracil for targeted therapy. Mater Sci Eng C. 2019;103:109828. doi:10.1016/j.msec.2019.109828
36. Ekladious I, Colson YL, Grinstaff MW. Polymer–drug conjugate therapeutics: advances, insights and prospects. Nat Rev Drug Discov. 2019;18(4):273–294. doi:10.1038/s41573-018-0005-0
37. Tomalia DA. Dendrimer research. Science. 1991;252(5010):1231. doi:10.1126/science.252.5010.1231-b
38. Chauhan AS. Dendrimer nanotechnology for enhanced formulation and controlled delivery of resveratrol. Ann N Y Acad Sci. 2015;1348(1):134–140. doi:10.1111/nyas.12816
39. Svenson S, Chauhan AS. Dendrimers for enhanced drug solubilization. Nanomed. 2008;3(5):679–702. doi:10.2217/17435889.3.5.679
40. McNerny DQ, Leroueil PR, Baker JR. Understanding specific and nonspecific toxicities: a requirement for the development of dendrimer‐based pharmaceuticals. WIREs Nanomed Nanobiotechnol. 2010;2(3):249–259. doi:10.1002/wnan.79
41. Frank LA, Gazzi RP, de Andrade Mello P, Buffon A, Pohlmann AR, Guterres SS. Imiquimod-loaded nanocapsules improve cytotoxicity in cervical cancer cell line. Eur J Pharm Biopharm. 2019;136:9–17. doi:10.1016/j.ejpb.2019.01.001
42. Raffin Pohlmann A, Weiss V, Mertins O, Pesce da Silveira N, Stanisçuaski Guterres S. Spray-dried indomethacin-loaded polyester nanocapsules and nanospheres: development, stability evaluation and nanostructure models. Eur J Pharm Sci. 2002;16(4):305–312. doi:10.1016/S0928-0987(02)00127-6
43. Trindade IC, Pound-Lana G, Pereira DGS, et al. Mechanisms of interaction of biodegradable polyester nanocapsules with non-phagocytic cells. Eur J Pharm Sci. 2018;124:89–104. doi:10.1016/j.ejps.2018.08.024
44. Teixeira M, Alonso MJ, Pinto MMM, Barbosa CM. Development and characterization of PLGA nanospheres and nanocapsules containing xanthone and 3-methoxyxanthone. Eur J Pharm Biopharm. 2005;59(3):491–500. doi:10.1016/j.ejpb.2004.09.002
45. Belbekhouche S, Mansour O, Carbonnier B. Promising sub-100 nm tailor made hollow chitosan/poly(acrylic acid) nanocapsules for antibiotic therapy. J Colloid Interface Sci. 2018;522:183–190. doi:10.1016/j.jcis.2018.03.061
46. Guterres SS, Alves MP, Pohlmann AR. Polymeric nanoparticles, nanospheres and nanocapsules, for cutaneous applications. Drug Target Insights. 2007;2:147–157.
47. Mukhopadhyay P, Chakraborty S, Bhattacharya S, Mishra R, Kundu PP. pH-sensitive chitosan/alginate core-shell nanoparticles for efficient and safe oral insulin delivery. Int J Biol Macromol. 2015;72:640–648. doi:10.1016/j.ijbiomac.2014.08.040
48. Gaber M, Hany M, Mokhtar S, Helmy MW, Elkodairy KA, Elzoghby AO. Boronic-targeted albumin-shell oily-core nanocapsules for synergistic aromatase inhibitor/herbal breast cancer therapy. Mater Sci Eng C. 2019;105:110099. doi:10.1016/j.msec.2019.110099
49. Fustier C, Chang T. PEG-PLA nanocapsules containing a nanobiotechnological complex of polyhemoglobin-tyrosinase for the depletion of tyrosine in melanoma: preparation and in vitro characterisation. J Nanomed Biother Discov. 2012;2:1–9. doi:10.4172/2155-983X.1000103
50. de Gomes MG, Pando Pereira M, Guerra Teixeira FE, et al. Assessment of unloaded polymeric nanocapsules with different coatings in female rats: influence on toxicological and behavioral parameters. Biomed Pharmacother. 2020;121:109575. doi:10.1016/j.biopha.2019.109575
51. Rosa P, Friedrich ML, Dos Santos J, et al. Desonide nanoencapsulation with açai oil as oil core: physicochemical characterization, photostability study and in vitro phototoxicity evaluation. J Photochem Photobiol, B. 2019;199:111606. doi:10.1016/j.jphotobiol.2019.111606
52. Huang P, Du S, Lin Y, et al. Identification of three potential circRNA biomarkers of polycystic ovary syndrome by bioinformatics analysis and validation. Int J Gen Med. 2021;14:5959–5968. doi:10.2147/IJGM.S324126
53. Gomes MLS, Da Silva Nascimento N, Borsato DM, et al. Long-lasting anti-platelet activity of cilostazol from poly(ε-caprolactone)-poly(ethylene glycol) blend nanocapsules. Mater Sci Eng C. 2019;94:694–702. doi:10.1016/j.msec.2018.10.029
54. Bzowska M, Karabasz A, Szczepanowicz K. Encapsulation of camptothecin into pegylated polyelectrolyte nanocarriers. Colloids Surf A. 2018;557:36–42. doi:10.1016/j.colsurfa.2018.05.070
55. Sahu P, Kashaw SK, Kushwah V, Sau S, Jain S, Iyer AK. pH responsive biodegradable nanogels for sustained release of bleomycin. Bioorg Med Chem. 2017;25(17):4595–4613. doi:10.1016/j.bmc.2017.06.038
56. Zhang W, Tung CH. Sequence-independent DNA nanogel as a potential drug carrier. Macromol Rapid Commun. 2017;38(20). doi:10.1002/marc.201700366
57. Ding F, Mou Q, Ma Y, et al. A crosslinked nucleic acid nanogel for effective siRNA delivery and antitumor therapy. Angew Chem Int Ed. 2018;57(12):3064–3068. doi:10.1002/anie.201711242
58. Jin B, Zhou X, Li X, Lin W, Chen G, Qiu R. Self-assembled modified soy protein/dextran nanogel induced by ultrasonication as a delivery vehicle for riboflavin. Molecules. 2016;21(3):282. doi:10.3390/molecules21030282
59. Su S, Wang H, Liu X, Wu Y, Nie G. iRGD-coupled responsive fluorescent nanogel for targeted drug delivery. Biomaterials. 2013;34(13):3523–3533. doi:10.1016/j.biomaterials.2013.01.083
60. El-Feky GS, El-Banna ST, El-Bahy GS, Abdelrazek EM, Kamal M. Alginate coated chitosan nanogel for the controlled topical delivery of silver sulfadiazine. Carbohydr Polym. 2017;177:194–202. doi:10.1016/j.carbpol.2017.08.104
61. Nakahashi-Ouchida R, Yuki Y, Kiyono H. Cationic pullulan nanogel as a safe and effective nasal vaccine delivery system for respiratory infectious diseases. Hum Vaccines Immunother. 2018;14(9):2189–2193. doi:10.1080/21645515.2018.1461298
62. Chiang WH, Thang HV, Huang WC, Huang YF, Chern CS, Chiu HC. Dual stimuli-responsive polymeric hollow nanogels designed as carriers for intracellular triggered drug release. Langmuir. 2012;28(42):15056–15064. doi:10.1021/la302903v
63. Salehi R, Rasouli S, Hamishehkar H. Smart thermo/pH responsive magnetic nanogels for the simultaneous delivery of doxorubicin and methotrexate. Int J Pharm. 2015;487(1):274–284. doi:10.1016/j.ijpharm.2015.04.051
64. Cai M, Zhu K, Qiu Y, Liu X, Chen Y, Luo X. pH and redox-responsive mixed micelles for enhanced intracellular drug release. Colloids Surf B. 2014;116:424–431. doi:10.1016/j.colsurfb.2014.01.012
65. Torchilin VP. Micellar nanocarriers: pharmaceutical perspectives. Pharm Res. 2007;24(1):1–16. doi:10.1007/s11095-006-9132-0
66. Kwon GS. Polymeric micelles for delivery of poorly water-soluble compounds. Crit Rev Ther Drug Carrier Syst. 2003;20(5):357–403. doi:10.1615/CritRevTherDrugCarrierSyst.v20.i5.20
67. Torchilin VP. Structure and design of polymeric surfactant-based drug delivery systems. J Control Release. 2001;73(2):137–172. doi:10.1016/S0168-3659(01)00299-1
68. Lo CL, Lin SJ, Tsai HC, et al. Mixed micelle systems formed from critical micelle concentration and temperature-sensitive diblock copolymers for doxorubicin delivery. Biomaterials. 2009;30(23):3961–3970. doi:10.1016/j.biomaterials.2009.04.002
69. Kim SH, Tan JPK, Nederberg F, et al. Mixed micelle formation through stereocomplexation between enantiomeric poly(lactide) block copolymers. Macromolecules. 2009;42(1):25–29. doi:10.1021/ma801739x
70. Yang L, Wu X, Liu F, Duan Y, Li S. Novel biodegradable polylactide/poly(ethylene glycol) micelles prepared by direct dissolution method for controlled delivery of anticancer drugs. Pharm Res. 2009;26(10):2332–2342. doi:10.1007/s11095-009-9949-4
71. Kabanov AV, Batrakova EV, Alakhov VY. Pluronic® block copolymers for overcoming drug resistance in cancer. Adv Drug Deliv Rev. 2002;54(5):759–779. doi:10.1016/S0169-409X(02)00047-9
72. Torchilin VP. PEG-based micelles as carriers of contrast agents for different imaging modalities. Adv Drug Deliv Rev. 2002;54(2):235–252. doi:10.1016/S0169-409X(02)00019-4
73. Griset AP, Walpole J, Liu R, Gaffey A, Colson YL, Grinstaff MW. Expansile nanoparticles: synthesis, characterization, and in vivo efficacy of an acid-responsive polymeric drug delivery system. J Am Chem Soc. 2009;131(7):2469–2471. doi:10.1021/ja807416t
74. Petros RA, DeSimone JM. Strategies in the design of nanoparticles for therapeutic applications. Nat Rev Drug Discovery. 2010;9(8):615–627. doi:10.1038/nrd2591
75. Li X, Meng Z, Guan L, et al. Glucose-responsive hydrogel optimizing Fenton reaction to eradicate multidrug-resistant bacteria for infected diabetic wound healing. Chem Eng J. 2024;487:150545. doi:10.1016/j.cej.2024.150545
76. Sajid A, Amjad M, Manzoor Q, et al. Synthesis of bimetallic oxides (SrO-CoO) nanoparticles decorated polyacrylamide hydrogels for controlled drug release and wound healing applications. Int J Biol Macromol. 2024;274:133194. doi:10.1016/j.ijbiomac.2024.133194
77. Chen L, Peng M, He W, et al. Microenvironment‐adaptive nanodecoy synergizes bacterial eradication, inflammation alleviation, and immunomodulation in promoting biofilm‐associated diabetic chronic wound healing cascade. Aggregate. 2024;5(6):e640. doi:10.1002/agt2.640
78. Hasan N, Cao J, Lee J. PEI/NONOates-doped PLGA nanoparticles for eradicating methicillin-resistant staphylococcus aureus biofilm in diabetic wounds via binding to the biofilm matrix. Materials Science and Engineering: C. 2019;103:109741. doi:10.1016/j.msec.2019.109741
79. Yu H, Li Y, Pan Y, et al. Multifunctional porous poly (L-lactic acid) nanofiber membranes with enhanced anti-inflammation, angiogenesis and antibacterial properties for diabetic wound healing. J Nanobiotechnol. 2023;21(1):110. doi:10.1186/s12951-023-01847-w
80. Sun C, Zhou X, Liu C, et al. An integrated therapeutic and preventive nanozyme‐based microneedle for biofilm‐infected diabetic wound healing. Adv Healthcare Materials. 2023;12(30):2301474. doi:10.1002/adhm.202301474
81. Zeng X. Chitosan@puerarin hydrogel for accelerated wound healing in diabetic subjects by miR-29ab1 mediated inflammatory axis suppression. Bioact Mater. 2023.
82. Zeng R, Lv B, Lin Z, et al. Neddylation suppression by a macrophage membrane-coated nanoparticle promotes dual immunomodulatory repair of diabetic wounds. Bioact Mater. 2024;34:366–380. doi:10.1016/j.bioactmat.2023.12.025
83. Elhabal SF, Abdelaal N, Elrefai MFM, et al. Green synthesis of zinc oxide nanoparticles from althaea officinalis flower extract coated with chitosan for potential healing effects on diabetic wounds by inhibiting TNF-α and IL-6/IL-1β signaling pathways. Int J Nanomed. 2024.
84. Li F, Shi Y, Liang J, Zhao L. Curcumin-loaded chitosan nanoparticles promote diabetic wound healing via attenuating inflammation in a diabetic rat model. J Biomater Appl. 2019;34(4):476–486. doi:10.1177/0885328219860929
85. Wang N. Dopamine-grafted oxidized hyaluronic acid/gelatin/cordycepin nanofiber membranes modulate the TLR4/NF-kB signaling pathway to promote diabetic wound healing. Int J Biol Macromol. 2024.
86. Wei Y, Chen H, Zhou Z, et al. Kill two birds with one stone: dual‐metal MOF‐nanozyme‐decorated hydrogels with ROS‐scavenging, oxygen‐generating, and antibacterial abilities for accelerating infected diabetic wound healing. Small. 2024;20(48):2403679. doi:10.1002/smll.202403679
87. Xu Z, Liu Y, Ma R, et al. Thermosensitive hydrogel incorporating Prussian blue nanoparticles promotes diabetic wound healing via ROS scavenging and mitochondrial function restoration. ACS Appl Mater Interfaces. 2022;14(12):14059–14071. doi:10.1021/acsami.1c24569
88. Chen G, He L, Zhang P. Encapsulation of green tea polyphenol nanospheres in PVA/alginate hydrogel for promoting wound healing of diabetic rats by regulating PI3K/AKT pathway. Materials Science and Engineering: C. 2020;110:110686. doi:10.1016/j.msec.2020.110686
89. Deng QS, Gao Y, Rui BY, et al. Double-network hydrogel enhanced by SS31-loaded mesoporous polydopamine nanoparticles: symphonic collaboration of near-infrared photothermal antibacterial effect and mitochondrial maintenance for full-thickness wound healing in diabetes mellitus. Bioact Mater. 2023;27:409–428. doi:10.1016/j.bioactmat.2023.04.004
90. Chen S, Lei W, Liu Q, et al. Silk-based nanocomposite hydrogel balances immune homeostasis via targeting mitochondria for diabetic wound healing. Chem Eng J. 2024;498:155884. doi:10.1016/j.cej.2024.155884
91. Zehra M, Zubairi W, Hasan A, et al. Oxygen generating polymeric nano fibers that stimulate angiogenesis and show efficient wound healing in a diabetic wound model. IJN. 2020;15:3511–3522. doi:10.2147/IJN.S248911
92. Zhao Y, Tian C, Liu Y, et al. All-in-one bioactive properties of photothermal nanofibers for accelerating diabetic wound healing. Biomaterials. 2023;295:122029. doi:10.1016/j.biomaterials.2023.122029
93. Waghmare VS, Wadke PR, Dyawanapelly S, Deshpande A, Jain R, Dandekar P. Starch based nanofibrous scaffolds for wound healing applications. Bioact Mater. 2018;3(3):255–266. doi:10.1016/j.bioactmat.2017.11.006
94. Wu B, Pan W, Luo S, et al. Turmeric‐derived nanoparticles functionalized aerogel regulates multicellular networks to promote diabetic wound healing. Adv Sci. 2024;11(18):2307630. doi:10.1002/advs.202307630
95. Chereddy KK, Lopes A, Koussoroplis S, et al. Combined effects of PLGA and vascular endothelial growth factor promote the healing of non-diabetic and diabetic wounds. Nanomed Nanotechnol Biol Med. 2015;11(8):1975–1984. doi:10.1016/j.nano.2015.07.006
96. Xia G, Liu Y, Tian M, et al. Nanoparticles/thermosensitive hydrogel reinforced with chitin whiskers as a wound dressing for treating chronic wounds. J Mater Chem B. 2017;5(17):3172–3185. doi:10.1039/C7TB00479F
97. Wang T, Wen M, Li N, Zhang L, Xue Y, Shang L. Marine‐derived nanozyme‐crosslinked self‐adaptive hydrogels for programmed regulating the regeneration process. Adv Funct Mater. 2024;34(39):2405257. doi:10.1002/adfm.202405257
98. Born LJ, Bengali S, Hsu ATW, et al. Chitosan particles complexed with CA5-HIF-1α plasmids increase angiogenesis and improve wound healing. IJMS. 2023;24(18):14095. doi:10.3390/ijms241814095
99. Li X, Liu Y, Zhang J, You R, Qu J, Li M. Functionalized silk fibroin dressing with topical bioactive insulin release for accelerated chronic wound healing. Mater Sci Eng C. 2017;72:394–404. doi:10.1016/j.msec.2016.11.085
100. Wu Y, Lyu Y, Li L, et al. Unimolecular cascaded multienzyme conjugates modulate the microenvironment of diabetic wound to promote healing. Biomacromolecules. 2024;25(1):43–54. doi:10.1021/acs.biomac.3c00698
101. Pu C, Wang Y, Xiang H, et al. Zinc-based polyoxometalate nanozyme functionalized hydrogels for optimizing the hyperglycemic-immune microenvironment to promote diabetic wound regeneration. J Nanobiotechnol. 2024;22(1):611. doi:10.1186/s12951-024-02840-7
102. Lari AS, Zahedi P, Ghourchian H, Khatibi A. Microfluidic-based synthesized carboxymethyl chitosan nanoparticles containing metformin for diabetes therapy: in vitro and in vivo assessments. Carbohydr Polym. 2021;261:117889. doi:10.1016/j.carbpol.2021.117889
103. Hunt NJ, Lockwood GP, Heffernan SJ, et al. Oral nanotherapeutic formulation of insulin with reduced episodes of hypoglycaemia. Nat Nanotechnol. 2024;19(4):534–544. doi:10.1038/s41565-023-01565-2
104. Kim M, Kim H, sun LY, et al. Novel enzymatic cross-linking–based hydrogel nanofilm caging system on pancreatic cell spheroid for long-term blood glucose regulation. Sci Adv. 2021;7(26). doi:10.1126/sciadv.abf7832
105. Giurazza R, Mazza MC, Andini R, Sansone P, Pace MC, Durante-Mangoni E. Emerging treatment options for multi-drug-resistant bacterial infections. Life. 2021;11(6):519. doi:10.3390/life11060519
106. Ye Y, Zheng Q, Wang Z, et al. Metal-phenolic nanoparticles enhance low temperature photothermal therapy for bacterial biofilm in superficial infections. J Nanobiotechnol. 2024;22(1):713. doi:10.1186/s12951-024-02985-5
107. Dörr S, Holland-Letz AK, Weisser G, Chatzitomaris A, Lobmann R. Bacterial diversity, antibiotic resistance, and the risk of lower limb amputation in younger and older individuals with diabetic foot infection. Int J Lower Extremity Wounds. 2023;22(1):63–71. doi:10.1177/1534734621992290
108. Louiselle AE, Niemiec SM, Zgheib C, Liechty KW. Macrophage polarization and diabetic wound healing. Transl Res. 2021;236:109–116. doi:10.1016/j.trsl.2021.05.006
109. Aitcheson SM, Frentiu FD, Hurn SE, Edwards K, Murray RZ. Skin wound healing: normal macrophage function and macrophage dysfunction in diabetic wounds. Molecules. 2021;26(16):4917. doi:10.3390/molecules26164917
110. Audu CO, Melvin WJ, Joshi AD, et al. Macrophage-specific inhibition of the histone demethylase JMJD3 decreases STING and pathologic inflammation in diabetic wound repair. Cell Mol Immunol. 2022;19(11):1251–1262. doi:10.1038/s41423-022-00919-5
111. Liu G, Yan D, Yang L, et al. The effect of miR-471-3p on macrophage polarization in the development of diabetic cardiomyopathy. Life Sci. 2021;268:118989. doi:10.1016/j.lfs.2020.118989
112. Liu X, Guo C, Yang W, et al. Composite microneedles loaded with astragalus membranaceus polysaccharide nanoparticles promote wound healing by curbing the ROS/NF-κB pathway to regulate macrophage polarization. Carbohydr Polym. 2024;345:122574. doi:10.1016/j.carbpol.2024.122574
113. Chen J, Chen L, She Z, Zeng F, Wu S. A multifunctional nanoaggregate-based system for detection of rheumatoid arthritis via optoacoustic/NIR-II fluorescent imaging and therapy via inhibiting JAK-STAT/NF-κB/NLRP3 pathways. Aggregate. 2024;5(1):e419. doi:10.1002/agt2.419
114. Wang S, Zhang Y, Zhong Y, et al. Accelerating diabetic wound healing by ROS-scavenging lipid nanoparticle–mRNA formulation. Proc Natl Acad Sci USA. 2024;121(22):e2322935121. doi:10.1073/pnas.2322935121
115. Halliwell B. Understanding mechanisms of antioxidant action in health and disease. Nat Rev Mol Cell Biol. 2024;25(1):13–33. doi:10.1038/s41580-023-00645-4
116. Sies H, Mailloux RJ, Jakob U. Fundamentals of redox regulation in biology. Nat Rev Mol Cell Biol. 2024;25(9):701–719. doi:10.1038/s41580-024-00730-2
117. Yang Z, Ren K, Chen Y, Quanji X, Cai C, Yin J. Oxygen-generating hydrogels as oxygenation therapy for accelerated chronic wound healing. Adv Healthcare Mater. 2024;13(3):2302391. doi:10.1002/adhm.202302391
118. Zubair M, Ahmad J. Role of growth factors and cytokines in diabetic foot ulcer healing: a detailed review. Rev Endocr Metab Disord. 2019;20(2):207–217. doi:10.1007/s11154-019-09492-1
119. Zhang Y, Li M, Wang Y, et al. Exosome/metformin-loaded self-healing conductive hydrogel rescues microvascular dysfunction and promotes chronic diabetic wound healing by inhibiting mitochondrial fission. Bioact Mater. 2023;26:323–336. doi:10.1016/j.bioactmat.2023.01.020
120. Kaur M, Misra S, Swarnkar P, et al. Understanding the role of hyperglycemia and the molecular mechanism associated with diabetic neuropathy and possible therapeutic strategies. Biochem Pharmacol. 2023;215:115723. doi:10.1016/j.bcp.2023.115723
121. Wang X, Sng MK, Foo S, et al. Early controlled release of peroxisome proliferator-activated receptor β/δ agonist GW501516 improves diabetic wound healing through redox modulation of wound microenvironment. J Control Release. 2015;197:138–147. doi:10.1016/j.jconrel.2014.11.001
122. Zhu Y, Cankova Z, Iwanaszko M, Lichtor S, Mrksich M, Ameer GA. Potent laminin-inspired antioxidant regenerative dressing accelerates wound healing in diabetes. Proc Natl Acad Sci. 2018;115(26):6816–6821. doi:10.1073/pnas.1804262115
123. Abdelkader DH, Tambuwala MM, Mitchell CA, et al. Enhanced cutaneous wound healing in rats following topical delivery of insulin-loaded nanoparticles embedded in poly(vinyl alcohol)-borate hydrogels. Drug Deliv Transl Res. 2018;8(5):1053–1065. doi:10.1007/s13346-018-0554-0
124. Zhao X, Zhang H, Li J, et al. Orally administered saccharide-sequestering nanocomplex to manage carbohydrate metabolism disorders. Sci Adv. 2021;7(14):eabf7311. doi:10.1126/sciadv.abf7311
125. Ji K, Wei X, Kahkoska AR, et al. An orally administered glucose-responsive polymeric complex for high-efficiency and safe delivery of insulin in mice and pigs. Nat Nanotechnol. 2024;19(1):1–12. doi:10.1038/s41565-024-01764-5
126. Xu Z, Liu G, Huang J, Wu J. Novel glucose-responsive antioxidant hybrid hydrogel for enhanced diabetic wound repair. ACS Appl Mater Interfaces. 2022;14(6):7680–7689. doi:10.1021/acsami.1c23461
127. Zhao W, Qiang L, Zhang C, et al. Near-infrared stimuli-responsive hydrogel promotes cell migration for accelerated diabetic wound healing. ACS Appl Mater Interfaces. 2024;16(38):50175–50187. doi:10.1021/acsami.4c05852
128. Yang X, Zhang C, Deng D, Gu Y, Wang H, Zhong Q. Multiple stimuli‐responsive MXene‐based hydrogel as intelligent drug delivery carriers for deep chronic wound healing. Small. 2022;18(5):2104368. doi:10.1002/smll.202104368
129. Xu Z, Fan J, Tian W, et al. Cellulose‐based pH‐responsive janus dressing with unidirectional moisture drainage for exudate management and diabetic wounds healing. Adv Funct Mater. 2024;34(3):2307449. doi:10.1002/adfm.202307449
130. Chen H, Guo Q, Chu Y, et al. Smart hypoxia-responsive transformable and charge-reversible nanoparticles for the deep penetration and tumor microenvironment modulation of pancreatic cancer. Biomaterials. 2022;287:121599. doi:10.1016/j.biomaterials.2022.121599
131. Liu L, Yang T, Wang S, et al. Preparation and design of intelligent multi-response PTMC/PVP composite nanofibrous dressing to promote wound closure and its applications in diabetic wounds. Chem Eng J. 2024;495:153872. doi:10.1016/j.cej.2024.153872
132. Wang Y, Liu K, Wei W, Dai H. A multifunctional hydrogel with photothermal antibacterial and AntiOxidant activity for smart monitoring and promotion of diabetic wound healing. Adv Funct Mater. 2024;34(38):2402531. doi:10.1002/adfm.202402531
133. Gong X, Yang J, Zheng Y, et al. Polymer hydrogel‐based multifunctional theranostics for managing diabetic wounds. Adv Funct Mater. 2024;34(26):2315564. doi:10.1002/adfm.202315564
134. Wang Q, Fu M, Guan Y, James TD, Zhang Y. Mechanistic insights into the novel glucose-sensitive behavior of
135. Xu X, Liu Y, Fu W, et al. Poly(N-isopropylacrylamide)-based thermoresponsive composite hydrogels for biomedical applications. Polymers. 2020;12(3):580. doi:10.3390/polym12030580
136. Zhou J, Fang C, Rong C, Luo T, Liu J, Zhang K. Reactive oxygen species-sensitive materials: a promising strategy for regulating inflammation and favoring tissue regeneration. Smart Mater Med. 2023;4:427–446. doi:10.1016/j.smaim.2023.01.004
137. Shi C, Zhang Y, Wu G, et al. Hyaluronic acid‐based reactive oxygen species‐responsive multifunctional injectable hydrogel platform accelerating diabetic wound healing. Adv Healthcare Materials. 2023:2302626. 10.1002/adhm.202302626
138. Liang Y, Li M, Yang Y, Qiao L, Xu H, Guo B. pH/glucose dual responsive metformin release hydrogel dressings with adhesion and self-healing via dual-dynamic bonding for athletic diabetic foot wound healing. ACS Nano. 2022;16(2):3194–3207. doi:10.1021/acsnano.1c11040
139. Yang N, Xiao W, Song X, Wang W, Dong X. Recent advances in tumor microenvironment hydrogen peroxide-responsive materials for cancer photodynamic therapy. Nano-Micro Lett. 2020;12(1):15. doi:10.1007/s40820-019-0347-0
140. Arrizabalaga JH, Smallcomb M, Abu-Laban M, et al. Ultrasound-responsive hydrogels for on-demand protein release. ACS Appl Bio Mater. 2022;5(7):3212–3218. doi:10.1021/acsabm.2c00192
141. Cao J, Wu J, Mu J, et al. ROS filter coating scaffold protects 3D mesenchymal stem cell spheroids for dual-phase treatment of spinal cord injury. Chem Eng J. 2023;462:142192. doi:10.1016/j.cej.2023.142192
142. Yang Y, Huang K, Wang M, et al. Ubiquitination flow repressors: enhancing wound healing of infectious diabetic ulcers through stabilization of polyubiquitinated hypoxia‐inducible factor‐1α by theranostic nitric oxide nanogenerators. Adv Mater. 2021;33(45):2103593. doi:10.1002/adma.202103593
143. Andreadi A, Lodeserto P, Todaro F, et al. Nanomedicine in the treatment of diabetes. IJMS. 2024;25(13):7028. doi:10.3390/ijms25137028
144. Zhang YN, Poon W, Tavares AJ, McGilvray ID, Chan WCW. Nanoparticle–liver interactions: cellular uptake and hepatobiliary elimination. J Control Release. 2016;240:332–348. doi:10.1016/j.jconrel.2016.01.020
145. Ullah S, Burki S, Munir AB, Yousaf G, Shafique M. Nanocarrier-based localized and effective treatment of renal disorders: currently employed targeting strategies. Nanomedicine. 2024;19(4):345–361. doi:10.2217/nnm-2023-0251
146. Su S, Kang P M. Recent advances in nanocarrier-assisted therapeutics delivery systems. Pharmaceutics. 2020;12(9):837. doi:10.3390/pharmaceutics12090837
147. Anwar M, Muhammad F, Akhtar B. Biodegradable nanoparticles as drug delivery devices. J Drug Delivery Sci Technol. 2021;64:102638. doi:10.1016/j.jddst.2021.102638
148. Sahu KK, Pradhan M, Singh D, Singh MR, Yadav K. Non-viral nucleic acid delivery approach: a boon for state-of-the-art gene delivery. J Drug Delivery Sci Technol. 2023;80:104152. doi:10.1016/j.jddst.2023.104152
149. Wang B, He X, Zhang Z, Zhao Y, Feng W. Metabolism of nanomaterials in vivo: blood circulation and organ clearance. Acc Chem Res. 2013;46(3):761–769. doi:10.1021/ar2003336
150. Xiao Y, Ding T, Fang H, et al. Innovative bio‐based hydrogel microspheres micro‐cage for neutrophil extracellular traps scavenging in diabetic wound healing. Adv Sci. 2024;11(21):2401195. doi:10.1002/advs.202401195
151. Lee ES, Shin JM, Son S, et al. Recent advances in polymeric nanomedicines for cancer immunotherapy. Adv Healthcare Materials. 2019;8(4):1801320. doi:10.1002/adhm.201801320
152. Kafetzis KN, Papalamprou N, McNulty E, et al. The effect of cryoprotectants and storage conditions on the transfection efficiency, stability, and safety of lipid-based nanoparticles for mRNA and DNA delivery. Adv Healthcare Mater. 2023;12(18):e2203022. doi:10.1002/adhm.202203022
153. Huang Y, Song M, Li X, et al. Temperature-responsive self-contraction nanofiber/hydrogel composite dressing facilitates the healing of diabetic-infected wounds. Mater Today Bio. 2024;28:101214. doi:10.1016/j.mtbio.2024.101214
154. Abarca-Cabrera L, Fraga-García P, Berensmeier S. Bio-nano interactions: binding proteins, polysaccharides, lipids and nucleic acids onto magnetic nanoparticles. Biomater Res. 2021;25(1):12. doi:10.1186/s40824-021-00212-y
155. Wagner M, Krieger A, Minameyer M, et al. Multiresponsive polymer nanoparticles based on disulfide bonds. Macromolecules. 2021;54(6):2899–2911. doi:10.1021/acs.macromol.1c00299
156. Erigi U, Dhumal U, Tripathy M. Phase behavior of polymer–nanorod composites: a comparative study using PRISM theory and molecular dynamics simulations. The Journal of Chemical Physics. 2021;154(12):124903. doi:10.1063/5.0038186
157. Mahmoudi N, Nisbet DR, Pastar I, Gould L, Matoori S, Mahmoudi M. Sex-specific nanomedicine- and biomaterials-based therapies of chronic wounds. Nat Rev Bioeng. 2024;2(6):447–449. doi:10.1038/s44222-024-00191-4
158. Jing Y, Zhao Z, Cao X, Sun Q, Yuan Y, Li T. Ultraflexible, cost-effective and scalable polymer-based phase change composites via chemical cross-linking for wearable thermal management. Nat Commun. 2023;14(1):8060. doi:10.1038/s41467-023-43772-4
159. Wei Hu S, Ding T, Tang H, Guo H, Cui W, Shu Y. Nanobiomaterial vectors for improving gene editing and gene therapy. Mater Today. 2023;66:114–136. doi:10.1016/j.mattod.2023.04.011
160. Zhao H, Xu J, Yuan H, et al. 3D printing of artificial skin patches with bioactive and optically active polymer materials for anti-infection and augmenting wound repair. Mater Horiz. 2022;9(1):342–349. doi:10.1039/D1MH00508A
161. Jin Y, Han G, Gao Y, et al. Serum-tolerant polymeric complex for stem-cell transfection and neural differentiation. Nat Commun. 2025;16(1):2022. doi:10.1038/s41467-025-57278-8
162. Harun-Or-Rashid M, Aktar M, Hossain M, et al. Recent advances in micro- and nano-drug delivery systems based on natural and synthetic biomaterials. Polymers. 2023;15(23):4563. doi:10.3390/polym15234563
163. Huang Y, Geng H, Wu Z, et al. An Ag2S@ZIF-van nanosystem for NIR-II imaging of bacterial-induced inflammation and treatment of wound bacterial infection. Biomater Sci. 2022;10(14):3972–3980. doi:10.1039/d2bm00550f
© 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The
full terms of this license are available at https://www.dovepress.com/terms.php
and incorporate the Creative Commons Attribution
- Non Commercial (unported, 4.0) License.
By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted
without any further permission from Dove Medical Press Limited, provided the work is properly
attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.
Recommended articles

Recent Advances in the Pharmaceutical and Biomedical Applications of Cyclodextrin-Capped Gold Nanoparticles
Abdellatif AAH, Ahmed F, Mohammed AM, Alsharidah M, Al-Subaiyel A, Samman WA, Alhaddad AA, Al-Mijalli SH, Amin MA, Barakat H, Osman SK
International Journal of Nanomedicine 2023, 18:3247-3281
Published Date: 14 June 2023
Nanotechnology-Enhanced Pharmacotherapy for Intervertebral Disc Degeneration Treatment
Shi S, Ou X, Liu C, Li R, Zheng Q, Hu L
International Journal of Nanomedicine 2024, 19:14043-14058
Published Date: 27 December 2024
Progress in the Application of Novel Nanomaterials in Targeted Therapy for Liver Cancer
Wei X, Cao W, Wang S, Zhang Y, Gao Z, Wang S, Yao L, Zhang Z, Li X, Deng W, Xie Y, Li M
International Journal of Nanomedicine 2025, 20:2623-2643
Published Date: 3 March 2025