Metal–organic frameworks (MOFs), a new class of porous organic–organic hybrid materials controlled by self-assembly of metal atoms and organic pillars, are attracting considerable interest because of their specific properties. More recently, the advantages of different types of nanoscale metal–organic frameworks for the use of MOF nanoparticles in stomatology have been reported in the literature. This article covers the treatment of oral cancer, surface modification of implants, antibacterial dressings, and treatment of periodontitis and periodontal regeneration. It presents recent applications, future challenges, and prospects for MOFs in stomatology in four areas. It provides an overview of recent advances in the design and application of MOFs in stomatology from their intrinsic properties to different syntheses and their use as smart drug delivery systems or a combination of these.

The development of biotechnology and related novel biomaterials is a major driver of research and clinical innovation in contemporary stomatology.1–3 The use of nanomaterials in stomatology is growing in scope because of ongoing advancements in nanoscience.4,5 A wide range of nanomaterials have been created thus far for use as therapeutic and diagnostic agents, and because of their high cargo capacity, unique surface characterization, and customizable size, stomatology has advanced greatly.6,7 For instance, composite resins that can fix dental perforations have been created using a combination of silica and zirconia nanoparticles.8 Bacterial cell membranes, among other things, can be destroyed by adding antimicrobial quaternary ammonium salts to the resin polymers now used to manufacture teeth.9 It is important to remember that medications based on nanomaterials offer several advantages over other types of pharmaceuticals. such as regulated release, improved blood circulation, and increased accumulation, all of which improve therapeutic efficacy and reduce unfavorable effects. Currently, the majority of nanomaterials can be classified as organic (polymer nanoparticles, liposomes, micelles, and dendrimers) or inorganic (metal-based, metal–oxide-based, and carbon-based nanomaterials).10,11 However, these nanostructures have certain intrinsic drawbacks, such as the inability of inorganic materials to degrade and the low rate at which organic materials load drugs. Hybrid nanomaterials have been developed to overcome these difficulties in biological applications of pure inorganic or organic material’s biological uses.12,13

Metal–organic frameworks (MOFs) are a class of crystalline materials with structures that are formed by the coordination of metal ions with organic groups. The fact that they combine the benefits of organic and inorganic materials is significant because they increase their suitability for biomedicine.14,15 First, because of the relatively weak metal–ligand coordination bonds, MOFs are easier to biodegrade; second, because of their large and high porosity, goods have a higher loading efficiency; and third, MOFs can be easily modified and can be used to encapsulate or modify a variety of goods. Therefore, MOFs have attracted considerable interest in medical applications.16,17 In addition, a growing body of research has demonstrated the great promise of MOFs materials for a range of medical applications, including drug delivery, bioimaging, cancer diagnostics, and antibacterial coating. These results may also aid in the development of new approaches for oral disease diagnosis and treatment.18,19

Stomatology extends beyond dental concerns to encompass the comprehensive study and treatment of the entire mouth structure including the lips, cheeks, and jaws. This field addresses more intricate diseases and conditions that may necessitate medical intervention surpassing conventional dental care. With a focus on four areas of oral medicine (as illustrated in Fig. 1), this study intends to investigate the introduction of MOFs materials and their possible uses in Stomatology. These include the treatment of oral cancer, implant surface modification, antibacterial dressings, and the treatment and regeneration of periodontitis. In this paper, we present a comprehensive review of the current research progress on the properties and applications of Metal–Organic Frameworks (MOFs). We evaluate their efficacy in oral disease treatment across various research domains and analyze the potential prospects and challenges associated with their application in stomatology. Our findings aim to establish a solid foundation for the future wider and safer implementation of MOFs in the field of stomatology.

FIG. 1.

The process of drug loading and drug release from MOFs, and its application in stomatology (the figure was drawn by Figdraw).

FIG. 1.

The process of drug loading and drug release from MOFs, and its application in stomatology (the figure was drawn by Figdraw).

Close modal

Metal–organic frameworks (MOFs) are highly ordered crystalline porous coordination polymers.20–22 They consist of inorganic metal (such as transition and lanthanide metals) ions/clusters as nodes, which self-assemble with organic units (such as carboxylates, phosphonates, imidazolates, and phenolates) through coordination bonds to form porous materials with periodic network structure.23,24 Their unique structural properties, such as high specific surface area, large pore size, and high porosity, make them useful in a wide range of research, including gas storage and separation,25,26 chemical separation,27,28 catalysis,29,30 sensing,31,32 semiconductors,33,34 and bioimaging.35,36 In addition, in recent years, MOFs have shown a wide range of applications in the field of biomedicine.37,38 MOFs possess intrinsic biological activity by delivering specific metal ions and ligands, or catalyzing the generation of messenger molecules owing to their clear structure, ultra-high surface area and porosity, adjustable pore size, and easy chemical functionalization. Compared to polymeric structures, their pore size and porosity are significantly smaller and can be precisely controlled, making MOFs suitable for drug delivery and for sustaining or triggering the release of nanoparticles.39–41 

Metal–organic frameworks (MOFs) have proven to be highly effective drug delivery platforms, primarily owing to their remarkable drug-loading capabilities.42,43 Metal–organic frameworks (MOFs) have distinctive characteristics, including a well-organized structure, considerable surface area, and significant pore volume. These features facilitate the adsorption of multifunctional molecules, either on the outer surface or in the free channels of the MOFs, allowing the entrapment of these molecules within the scaffold. In addition, functional molecules can be integrated into metal–organic frameworks (MOFs) by covalent bonding using either one-pot synthesis or post-synthetic modification techniques.44,45

The use of endogenous stimuli, such as pH, to regulate the release of substances has been extensively investigated and applied in many drug delivery systems.46–48 The pH fluctuation at the site of inflammation, in comparison to standard physiological settings, modulates the binding process and, therefore, facilitates medication release.49,50 The coordinating bond by which a therapeutic agent is linked or loaded into metal–organic frameworks (MOFs) typically exhibits pH sensitivity, offering a more favorable delivery platform.51,52 In addition to pH, various stimuli including heat, light, electric and magnetic fields, and pressure have the potential to trigger changes in the physical and chemical characteristics of metal–organic frameworks (MOFs).53,54 Certain metal–organic frameworks (MOFs) have demonstrated photocatalytic capabilities, including the ability to switch photocurrents in response to light stimulation. These alterations encompass structural modifications resulting from bond breakage, as well as changes in the coordination number and mode of the metal ions and ligands. Consequently, these modifications facilitated drug release. The controlled release of therapeutic agents by light has gained interest because of its ability to achieve precise spatial and temporal control.55,56 In addition, pressure-responsive delivery has been explored as a means of achieving the same goal. The delivery mechanism encompasses structural modifications, including variations in resilience, crystallization, and morphology, in response to varying pressure situations.57,58

In summary, porous materials composed of metal ions and organic units are known as metal–organic frameworks (MOFs). However, not all MOFs are inherently active. In addition, MOFs have a large surface area, are well structured, and have porosity, which helps with multifunctional molecule adsorption and release. Covalent bonding or modification procedures can be utilized to insert functional molecules into MOFs, and endogenous stimuli, including pH, heat, light, electric and magnetic fields, and pressure can be employed to control the release of chemicals. Bond breaking and modifications to the coordination number and coordination pattern of metal ions and ligands can modify the characteristics of metal–organic frameworks (MOFs) and affect drug release. As a result, MOFs have been employed as a successful drug delivery technology that permits accurate control over the timing and location of drug release.

Titanium and its alloys have been widely recognized as optimal materials for dental implants because of their exceptional mechanical strength, commendable corrosion resistance, and biocompatibility.59 However, it is important to note that the absence of bioactivity on the surface of natural titanium implants may potentially increase the risk of infection and often lead to insufficient osseointegration. The significance of modifying implant surfaces lies in their ability to augment antibacterial activity, impede biofilm formation, mitigate peri-implant infections, boost osseointegration, and elevate the success rates of implant surgeries.60,61

We are aware of the potent bactericidal properties of metal ions, particularly those primarily composed of silver, copper, and zinc ions. Strong ion oxidation abilities allow these metal ions to quickly combine with the sulfhydryl group of the bacterial protease in the bacterial body, inactivating the protease and ultimately causing bacterial death. These metal ions can also destroy the bacterial cell membrane and metabolic process of the cell, thereby inhibiting bacterial reproduction. In addition, when the bacteria are eliminated, the metal ions may be left free to engage in further killing of additional bacteria until all the germs have been eliminated.62–65 

Because the metal nodes in MOFs have the ability to release metal ions as they expand, MOFs have intrinsic biological activity.66,67 The number of metal ions released has a direct correlation with the bactericidal efficiency of the released metal ions, which have significant antibacterial effects on oral bacterial strains, such as Clostridium nucleum, Porphyromonas gingivalis, Streptococcus mutans, and Streptococcus pyogenes. Applying ZIF-8 coatings, for instance, to titanium implants can release zinc ions and have a number of associated effects, including inhibiting the growth of S. mutans.68 It can also raise MG63 cell ALP activity, encourage mineralization of the extracellular matrix, and upregulate the expression of osteoblast genes. Furthermore, MOFs can provide long-lasting and persistent antibacterial effects by gradually releasing ions when they degrade due to their role as metal ion reservoir.69,70

Metal ions produced by MOFs can control angiogenesis by influencing vascular endothelial growth factor, hypoxia-inducing factor, angiogenesis-related genes, endothelial cells, and macrophage immune modulation.71–74 In addition to their strong angiogenic effects, copper, magnesium, strontium, cobalt, and other metal ions stimulate osteogenesis via angiogenesis.75,76 For instance, the ZIF-8/Cu precursor was carefully pyrolyzed to create a CuO@ZnO composite, which was subsequently grafted onto a titanium alloy that had been treated with PDA. It is possible to deliberately release zinc (Zn2+) and copper (Cu2+) ions to cause bacteria to produce too many intracellular reactive oxygen species (ROS), which will prevent bacteria from forming biofilms. By increasing vascular endothelial growth factor (VEGF) expression, liberated Cu2+ ions can induce angiogenesis in human umbilical vein endothelial cells (HUVECs). Furthermore, the CuO@ZnO-coated titanium markedly enhanced human bone marrow mesenchymal stem cells (hBMSC) adhesion, proliferation, and osteogenesis.77 

In addition, MOFs can achieve antibacterial and pro-new bone-formation effects by releasing metal ions, such as magnesium, zinc, silver, and copper. They can also increase the expression of anti-inflammatory genes and inhibit the expression of pro-inflammatory genes, thereby exerting anti-inflammatory effects.78–80 

For instance, Shen et al. fabricated Mg/Zn-MOF74 hybridized organic skeleton coatings on alkali-heat-treated titanium (AT) surfaces. The composite coatings of Mg/Zn-MOF74 exhibited excellent stability, and the modified samples demonstrated sensitivity to bacterial acidic microenvironments, displaying strong antimicrobial activity against both Escherichia coli and Staphylococcus aureus. Moreover, the coatings displayed promising early anti-inflammatory properties compared to natural Ti substrates modified which MOF74 significantly increased the expression of anti-inflammatory genes, such as IL-10 and IL-1ra, while decreasing the expression of pro-inflammatory genes, such as IL-1β and TNFα.81 

In vivo experiments were conducted by implanting different substrates [Ti, alkali treated titanium (AT), and AT-Mg/Zn3] in the presence of Sta. aureus for three days after implantation to investigate their antibacterial and anti-inflammatory capacities. Impressions and agar plates revealed minimal bacteria on the surface of the AT-Mg/Zn3 implants, as well as in the surrounding medullary cavity. Corresponding quantitative analysis further confirmed that AT-Mg/Zn3 implants exhibited the highest bacterial inhibition rate (>90%) among the three groups (p < 99.9%). In addition, hematoxylin-eosin (HE) staining and CD68 staining images showed a higher clustering of mononuclear cells (indicated by green arrows or red fluorescent markers) around Ti and AT implants compared to the AT-Mg/Zn3 group in both the epiphysis and medullary cavity regions.

These results further validated that, during the early stages of implantation, AT-Mg/Zn3 implants possessed remarkable antibacterial and anti-inflammatory properties and significantly enhanced new bone formation at infected and non-infected sites.81 

Interferon-gamma (IFN-γ), interleukin 6 (IL-6), and tumor necrosis factor alpha (TNF-α) are pro-inflammatory substances released by inflammatory cells, such as neutrophils, white blood cells, and macrophages, when they congregate in the affected area during the inflammatory response. These variables impair the metabolism of the mitochondrial electron transport chain (ETC) due to the release of reactive oxygen species (ROS).82,83

MOFs can also efficiently control inflammation by assembling compounds with antioxidant effects to offset the excess ROS produced by the inflammatory response and altering the organic ligands that coordinate with metal ions or clusters. For instance, large numbers of manganese porphyrin molecules are regularly assembled into two-dimensional manganese oxide films (2D MOFs), which exhibit unique anti-inflammatory and biomineralizing capabilities, along with SOD and catalase-like activities.84 

MOFs can upregulate genes related to osteogenesis, leading to an increase in the formation of mineralized nodes and the promotion of osteogenesis by accelerating extracellular matrix mineralization and protein translation. The expressions of alkaline phosphatase (ALP), collagen I (COLI), osteopontin (OPN), and osteocalcin (OCN) genes were all up-regulated after doping the Mg-MOF74 coating with suitable Zn2+ (especially AT-Mg/Zn3).81 Calcium–strontium metal–organic frameworks (Ca–Sr MOFs) can increase alkaline phosphatase (ALP) production. Furthermore, the concomitant release of calcium and strontium from these MOFs can generate signals that stimulate bone formation, thereby facilitating bone mineralization.85 

Because of their exceptional qualities, such as large porosity and high specific surface area, metal–organic frameworks (MOFs) can be employed as efficient drug transporters. These characteristics improve the biological qualities of medications and other bioactive substances by facilitating their effective transit and collection. For instance, adding zeolite imidazolidine skeleton8 containing naringenin to the titanium substrate can increase the antibacterial capabilities of the coating and dramatically boost osteoblast proliferation and osteogenic differentiation.86 

In conclusion, because their metal nodes discharge metal ions, metal–organic frameworks (MOFs) are intrinsically bioactive substances with potent bactericidal properties. These metal ions can bind to the bacterial protease, inactivate it, and kill bacteria. They can also damage bacterial cell membranes and metabolism, preventing bacteria from reproducing. The long-lasting antibacterial activity of MOFs is attributed to their metal–ion reservoirs. Furthermore, MOFs can enhance angiogenesis and bone formation by releasing metal ions. They can also control the immune cells by minimizing oxidative stress and inflammatory reactions. Its large specific surface area and high porosity make it an effective drug-delivery vehicle. In addition to being antibacterial and anti-inflammatory, MOFs can promote angiogenesis and bone formation and have good biocompatibility when used as a covering. As a result, MOFs have several potential uses in implant surface modification.

Oral squamous cell carcinoma (OSCC) is a prevalent and fatal malignancy affecting the maxillofacial region. Its aggressive growth pattern and propensity for metastasis make it challenging to achieve complete eradication by surgical resection alone.87,88 Chemotherapy is widely recognized as a highly efficacious therapeutic approach for eradicating cancer cells and mitigating the risk of tumor recurrence and metastasis. However, a significant drawback associated with traditional chemotherapy is the necessity to administer high drug dosages due to inadequate biodistribution, thereby leading to the frequent occurrence of side effects that are dose-dependent.89,90 This necessitates the investigation of innovative and effective drug delivery systems (DDS). An effective approach to address the issue of drug buildup in healthy tissues is to enhance the targeted delivery of chemotherapeutic agents specifically toward cancer cells.91,92 The implementation of a focused drug delivery system has the potential to mitigate the adverse effects associated with chemotherapeutic agents and enhance the efficacy of treatment for oral squamous cell carcinoma (OSCC). The intricate nature of the tumor microenvironment, characterized by diminished oxygen levels, acidic pH, and heightened levels of hydrogen peroxide (H2O2) and glutathione (GSH), frequently poses challenges to the effectiveness of cancer therapy.93,94 To enhance the precision of anticancer therapy, it is possible to focus on targeting the microenvironmental stimuli. An illustration of this may be seen in the difference in pH levels between mildly acidic malignant settings and typical tissue environments, which provides a conceptual foundation for the development of drug delivery systems that respond to pH changes. In the context of local oral cancer treatment, a combination of doxorubicin (Dox) and celecoxib (Cel) was introduced into a system known as Dox/Cel/MOFs@Gel. The Dox/Cel/MOFs@Gel composite materials exhibited a notable ability to encapsulate Dox and Cel, excellent pH-responsive behavior, effectively induced apoptosis in cancer cells, and regulated tumor angiogenesis. The use of this localized therapeutic approach resulted in a substantial decrease in systemic toxicity and did not induce any noteworthy harm to adjacent organs.95 

The hydrogen sulfide (H2S) pathway is activated within the tumor microenvironment. A high level of reduced hydrogen sulfide (H2S) leads to the depletion of the generated hydroxyl radicals and impairs the effectiveness of chemodynamic therapy (CDT). To prevent the depletion of the hydroxyl radicals, the investigators incorporated Cu2+ ions into the MOF structure to produce CuS precipitates, which are insoluble when exposed to H2S, thereby improving the efficacy of CDT.96 

The elevated concentration of H2O2 in tumor cells is another characteristic of the tumor microenvironment, according to which researchers have developed nanocomplexes based on metal–organic frameworks (MOFs) of titanium (Ti) that are capable of releasing CO based on the level of hydrogen peroxide present in the tumor. This nanocomplex enables both the controlled release of CO triggered by H2O2 within the tumor microenvironment and real-time monitoring of CO release through fluorescence imaging. This targeted release mechanism enables the nanocomplex to efficiently eradicate tumor cells while safeguarding normal cells.97 Furthermore, it has been shown that metal–organic frameworks (MOFs) incorporating transition metal ions possess the capability to interact with hydrogen peroxide, thereby resulting in the production of detrimental hydroxyl radicals.98 

Metal–organic frameworks (MOFs) can act as carriers for drug molecules and can be modified on their surfaces to precisely target cancer cells, thus facilitating accurate medication delivery. Folic acid incorporation is one of the several surface modifications that have emerged as the most frequently employed strategy for targeted therapy in recent research and clinical practice. The overexpression of folate receptors has been observed on the cell membranes of cancer cells. Folate-modified MOFs have a distinctive inclination toward, which can enable the precise distribution of medicine, leading to its release within malignant tissue.99,100

Furthermore, the modification of MOF at the cellular level can also be utilized to attain a targeted therapeutic outcome, as demonstrated by the utilization of CXCR2-containing dental pulp stem cells (DPSCs) membranes to modify/coat MOFs, leading to the production of MOF@DPSCM. This study revealed that CAL27 cells have the capacity to secrete CXCL8. This secretion leads to the upregulation of CXCR2 receptors on DPSC cellular membranes, ultimately resulting in the attraction of DPSCs to OSCC sites where CAL27 cells are present. In contrast to other metal–organic frameworks (MOFs), the MOF@DPSCM demonstrated a distinct capability to selectively adhere to and effectively penetrate CAL27 cells.101 

In addition to the CDT treatment, metal–organic frameworks (MOFs) have the potential to be used in photodynamic therapy. MOFs consisting of porphyrin-based ligands have been developed as potential approaches for near-infrared (NIR) photodynamic therapy (PDT) treatment.102 These ligands act as photosensitizers, whereas the incorporation of high-Z metal ions into the MOF scaffolds of porphyrins serves as a radiosensitizer. This allows for the combination therapy of radiation therapy (RT) and PDT.103 

In summary, metal–organic frameworks (MOFs) include notable characteristics, such as a substantial specific surface area, considerable pore size, and significant porosity. These attributes enable MOFs to accommodate therapeutic substances efficiently, including antibiotics and natural compounds. The self-assembly of inorganic clusters within MOFs occurs through coordination bonds with organic units, facilitating the manipulation of their physical characteristics and chemical properties. In addition to incorporating desired functional groups into the organic ligand through pre-designed ligands or post-synthetic modification techniques, controlled release of the enclosed drug can be accomplished by subjecting the system to various external stimuli, including but not limited to pH, light, pressure, ions, magnetic fields, and temperature. Furthermore, the explicit organization of the content facilitates the examination of the dynamics between the host and client. Due to its distinctive characteristics, MOFs are widely regarded as highly promising contenders in the field of medication delivery and cancer therapy.

Periodontal disease is frequently correlated with the depletion of alveolar bone and the presence of microbial infections within periodontal tissues.104,105

The abundance and complexity of oral micro-organisms are significant.106,107 The combination of periodontal surgery with oral broad-spectrum antibiotics is commonly used to prevent or minimize postoperative infections. However, it is worth noting that the systemic drug concentration for target site treatment is quite low, leading to significant side effects and the potential for bacterial imbalance resulting from excessive medication administration. Hence, the implementation of a localized drug delivery system is crucial for effective management of periodontal diseases.108–110 An optimal local drug delivery system should exhibit consistent and sustained drug release, exhibit prolonged therapeutic efficacy, and mitigate hazardous adverse reactions and dosing frequency.111,112

Due to their substantial drug encapsulation capacity and minimal premature release, MOFs have been identified as promising candidates for local drug delivery systems in periodontics. ZIF-8 was chosen as the carrier for ceftazidime encapsulation. During the assay, the composite material demonstrated sustained release of ceftazidime for one week, producing a bactericidal effect against E. coli.113 

In another study, composite microspheres composed of CD-MOF nanocrystals embedded in a biocompatible polymer (PAA) matrix were loaded with two drugs, ibuprofen (IBU) and lansoprazole (LPZ). The microspheres were spherical in shape and continued to release drugs over a long period. In addition, they exhibited reduced cytotoxicity.114 

Photodynamic therapy (PDT) is currently utilized in clinical practice for the treatment of a range of diseases, with particular emphasis on its application in antibacterial treatments. Consequently, PDT is anticipated to serve as a promising alternative to the existing clinical therapies for periodontitis.115 

Certain metal–organic frameworks (MOFs) have shown photocatalytic properties such as the capacity to change the optical flow in response to light stimulation, as previously described.116,117 These modifications include shifts in the coordination numbers and coordination patterns of the metal ions and ligands, as well as structural modifications caused by bond breakage.118,119 Two-dimensional porphyrin metal–organic frameworks (MOF) have the greatest potential for PDT. For example, two-dimensional MOF nanosheets composed of atoms and layers of ferric oxide can be combined with a polyethylene glycol matrix to create an ointment. Reactive oxygen species and released ions combine with light exposure to provide a broad spectrum of antibacterial action. This antibacterial activity has the capacity to reduce inflammation and encourage angiogenesis and is effective against a variety of oral infections, including P. gingivalis and Pseudomonas nucleomonas.120 

Furthermore, the ability of photocontrolled medicinal medications to achieve precise spatiotemporal control has drawn public interest. After loading the medication, MOFs can be used to create photosensitive composite materials by adding photosensitizers. This allows for the controlled release of drug molecules and the achievement of antibacterial properties. For example, AuNR@SiO2@UIO-66 composite microspheres were created using UIO-66 as the framework. Drugs can be released from the composite using two distinct mechanisms: activation in response to near-infrared (NIR) light and slow passive release at low concentrations, followed by quick active release at high doses. Furthermore, gram-positive and gram-negative bacteria are significantly inhibited by a light-responsive material.121 

In summary, because oral bacteria are numerous and intricate, broad-spectrum antibiotics are frequently administered to prevent infection during periodontal surgery. However, local delivery systems are essential because low systemic doses may cause adverse effects and bacterial imbalances. Because of their distinct benefits, metal–organic frameworks (MOFs) are regarded as excellent candidate materials for the treatment of periodontal disorders. In addition, the use of photodynamic therapy (PDT) in the management of periodontitis is promising. Certain MOFs, such as two-dimensional porphyrin MOFs, that have antimicrobial capabilities and stimulate angiogenesis possess photocatalytic properties. MOFs can be employed as photosensitive composite materials by adding photosensitizers to enable the precise release of medicinal molecules and their antibacterial characteristics. Research on photo-controlled pharmaceuticals has also attracted attention.

Periodontitis frequently coincides with alveolar bone depletion, prompting significant interest in the field of periodontal regeneration.122,123 In the realm of biological strategies, the prevailing methodology for tissue engineering involves the integration of scaffolds, cells, and biologically active substances, such as drugs and growth factors to facilitate tissue regeneration. Numerous drugs and growth factors have been subjected to experimentation in the realm of periodontal regeneration. These conventional biological substances can be altered or amalgamated with inorganic materials and polymers to create composite materials. The objective is to establish a suitable microenvironment and scaffold system that can effectively induce periodontal regeneration.124,125

Compared to conventional materials, metal–organic frameworks (MOFs) possess inherent bioactivity due to their ability to release specifically targeted metal ions. By integrating nanoscale metal–organic frameworks (NMOFs) into engineered scaffolds, it is possible to alter the functional groups, porosity, roughness, and hydrophilicity of the scaffolds, thereby enhancing the adhesion and interaction of mesenchymal stem cells (MSCs).126,127

For instance, the incorporation of copper-loaded zeolitic imidazolate framework-8 (ZIF-8) onto poly l-lactic acid (PLLA) frameworks resulted in improved osteogenic differentiation in contrast to pure PLLA frameworks while also providing protection against bacterial infection.128 

Magnesium-based metal–organic framework (MOF)-doped cement has demonstrated favorable antimicrobial properties, potentially facilitating bone growth and reducing the risk of infection in wounds.129 

When used in combination with implant materials, MOFs can be loaded with drug molecules to provide additional antibacterial or anti-infective properties. The incorporation of levofloxacin-loaded ZIF-8 particles into implant materials can improve the osseointegration and antimicrobial capacity of the material,130 and the incorporation of dexamethasone-loaded ZIF-8 into cellulose-hydroxyapatite nanocomposites can prolong and control the release of the dexamethasone material and improve resistance to infection.131 

MOFs can potentially increase the osteogenic capability of scaffold materials by carrying proteins and drug molecules. By incorporating BMP-2-loaded ZIF-8 nanoparticles into porous scaffolds of polypropylene glycol and mesoporous bioactive glass (PLGA/MBG), enhanced osteogenic activity, promotion of new bone formation, and repair of bone defects can be achieved through slow and sustained release of bone morphogenetic protein 2 (BMP-2).132 

Mesenchymal stem cells (MSCs) are able to adhere, develop, and specialize in an extracellular environment that is conducive to their growth when placed in nanoscale metal–organic frameworks (MOFs), according to prior research.133 The combination of MOFs and bone scaffold materials can promote the growth of new bones and provide anti-infection properties. However, several researchers have questioned whether MOFs can be utilized as bone scaffold materials. To assess the potential of UIO-66 nanoparticles to stimulate bone growth in vivo, one study used a rabbit bone defect model. cd34-expressing cells were successfully transformed into osteoblasts by the nanomaterial, which also showed exceptional biocompatibility. This process leads to the stimulation of the BMP-2/SMAD pathway in vivo, osteoblast differentiation, and the eventual creation of bone tissue.134 The regeneration of alveolar bone is currently a popular area of study in periodontal therapy, although the notion that MOFs can serve as bone scaffolds on their own is not well supported by available data. Nevertheless, this study offers a new concept for using MOFs for periodontal regeneration.

Furthermore, there is research indicating that metal–organic frameworks (MOFs) have potential research value in the field of endodontics, in addition to their application in periodontal treatment. Incorporation of olivetol (OLV) into γ-cyclodextrin metal–organic frameworks (γ-CD-MOFs) allows for effective management of dentin hypersensitivity. The therapeutic agent penetrates the pulp from the dentin layer without damaging the structural integrity of the tooth and demonstrates analgesic properties.135 

In conclusion, metal–organic frameworks (MOFs) are a new biomaterial with potential research value in periodontal regeneration and endodontics. MOFs can modify the functional groups, porosity, roughness, and hydrophilicity of scaffolds, thereby enhancing stem cell adhesion and interactions. In addition, MOFs can be loaded with drug molecules and proteins, providing additional antimicrobial, anti-infective, and osteogenic capabilities. This research has shown that MSCs can develop and specialize in an extracellular environment that promotes their growth when placed in nanoscale MOFs. However, further research is needed to support the use of MOFs as bone-scaffold materials. In addition, MOFs have potential in the field of endodontics for the treatment of dentin hypersensitivity by adding olive alcohol. Currently, the use of MOFs for the treatment of oral diseases is restricted to in vitro and in vivo studies. Further studies are needed to confirm the efficacy of these drugs.

The oral and maxillofacial regions are vulnerable areas of the human body, which are susceptible to injury in various contexts, such as daily activities, occupational hazards, traffic accidents, and warfare.136,137 Given the unique anatomical structure of this region, any harm inflicted on it can facilitate the entry of microorganisms into the oral cavity, leading to potential infections. An ideal wound dressing is capable of maintaining a moist environment, avoiding wound dryness while preventing retting and absorption of wound drainage, providing protection against bacterial infection, stimulating angiogenesis and connective tissue growth, promoting gas exchange, increasing epidermal migration, maintaining adequate blood flow temperature, maintaining an electrical gradient, non-adhesion, rapid separation and debridement, non-allergenic, non-toxic, and sterile. These factors are beneficial for promoting wound healing.138,139 Currently, compared to other nanomaterials, MOFs have great potential for antimicrobial dressing applications because of their increased drug load, targeted delivery of surface conditioning, and controlled release of wound healing agents, which are less toxic.140–143 

A study was conducted on the development of a dressing using Ag–metal–organic framework-loaded chitosan nanoparticles and a poly(vinyl alcohol)/sodium alginate/chitosan (PACS) bilayered hydrogel. Ag@MOF served as a reservoir of Ag ions, which were released continuously, resulting in the long-lasting antibacterial properties of the dressing. This facilitated quick wound healing.144 In another investigation, a polymeric CMC matrix was combined with tetracycline-loaded UIO-66, forming a dressing that sustained slow drug release in both artificial sweat and simulated wound exudate. This dressing showed effective antimicrobial activity against E. coli and Sta. aureus while also displaying good biocompatibility.145 

The findings from these studies indicate that MOFs have the potential to significantly enhance wound antimicrobial dressings through both their inherent properties and those acquired through further research.

Metal–organic frameworks (MOFs) have garnered significant attention in the biomedical field due to their favorable structural properties, including a well-defined structure, large specific surface area, remarkable porosity, and the ability to readily alter pore size and function. These unique properties make MOFs promising candidates for drug delivery and oral cancer therapy as well as potential treatments for periodontal and endodontic diseases, thanks to their antimicrobial, anti-infective, and osteogenic capabilities either by themselves or when loaded with cargoes. Despite notable progress in this area, there remain several challenges that require further investigation, such as complex functionalization techniques that can impact functional molecule activity during the synthesis of MOFs using organic ligands. In addition, researchers must consider drug loading/release kinetics along with in vivo toxicity/degradation mechanisms/pharmacokinetics of MOF nanoparticles. Further studies are necessary to rationalize MOFs while improving biostability/biocompatibility/therapeutic effects so that they may be applied more quickly toward diagnosing/treating oral diseases.

This study was supported by Hangzhou Normal University Affiliated Hospital. This work was carried out under the Research Program Peiyuan Plan of Hangzhou Normal University Affiliated Hospital.

The authors have no conflicts to disclose.

Minghe Zheng: Conceptualization (equal); Formal analysis (equal); Investigation (equal); Methodology (equal); Project administration (equal); Writing – original draft (equal). Ru Li: Investigation (equal); Project administration (equal); Resources (equal). Jiaye Wang: Validation (equal); Visualization (equal). Yanlin Huang: Project administration(equal), Resources (equal). Mingfang Han: Methodology (equal); Project administration (equal). Zehui Li: Formal analysis (equal); Writing – review & editing (lead).

The data that support the findings of this study are available within the article.

1
V.
Grassia
and
L.
Nucci
, “
New materials in oral surgery
,”
Materials
13
(
5
),
1034
(
2020
).
2
Q.
Ding
,
J.
Cui
,
H.
Shen
et al, “
Advances of nanomaterial applications in oral and maxillofacial tissue regeneration and disease treatment
,”
WIREs Nanomed. Nanobiotechnol.
13
,
e1669
(
2020
).
3
Y.
Zhao
and
S.
Jalili
, “
Dextran, as a biological macromolecule for the development of bioactive wound dressing materials: A review of recent progress and future perspectives
,”
Int. J. Biol. Macromol.
207
,
666
682
(
2022
).
4
T.
Barot
,
D.
Rawtani
, and
P.
Kulkarni
,
Nanotechnology-based materials as emerging trends for dental applications
.
Rev. Adv, Mater. Sci.
2021
;
60
(
1
):
173
189
.
5
Z.
Chen
,
Z.
Chu
,
Y.
Jiang
et al, “
Recent advances on nanomaterials for antibacterial treatment of oral diseases
,”
Materials Today Bio
20
,
100635
(
2023
).
6
M. J.
Virlan
,
D.
Miricescu
,
R.
Radulescu
et al, “
Organic nanomaterials and their applications in the treatment of oral diseases
,”
Molecules
21
(
2
),
207
(
2016
).
7
J. J.
Chokkattu
,
S.
Neeharika
, and
M.
Rameshkrishnan
, “
Applications of nanomaterials in dentistry: A review
,”
J. Int. Soc. Prev. Community Dent.
13
(
1
),
32
41
(
Jan-Feb 2023
).
8
A.
Alshamrani
,
A.
Alhotan
,
E.
Kelly
, and
A.
Ellakwa
, “
Mechanical and biocompatibility properties of 3D-printed dental resin reinforced with glass silica and zirconia nanoparticles: In vitro study
,”
Polymers
15
(
11
),
2523
(
2023
).
9
H.
Zhang
,
S.
Zhao
,
A.
Li
et al, “
Structure-dependent antimicrobial mechanism of quaternary ammonium resins and a novel synthesis of highly efficient antimicrobial resin
,”
Sci. Total Environ.
768
,
144450
(
2021
).
10
P. K. P.
Sreenivasalu
,
C. P.
Dora
,
R.
Swami
et al, “
Nanomaterials in dentistry: Current applications and future scope
,”
Nanomaterials
12
(
10
),
1676
(
2022
).
11
S.
Chu
,
J.
Wang
, and
F.
Gao
, “
The application of chitosan nanostructures in stomatology
,”
Molecules
26
(
20
),
6315
(
2021
).
12
R.
Onnainty
,
B.
Onida
,
P.
Páez
,
M.
Longhi
,
A.
Barresi
, and
G.
Granero
, “
Targeted chitosan-based bionanocomposites for controlled oral mucosal delivery of chlorhexidine
,”
Int. J. Pharm.
509
,
408
418
(
2016
).
13
T.
Jiang
,
W.
Su
,
Y.
Li
et al, “
Research progress on nanomaterials for tissue engineering in oral diseases
,”
J. Funct. Biomater.
14
(
8
),
404
(
2023
).
14
A.
Ahmed
,
D.
McHugh
, and
C.
Papatriantafyllopoulou
, “
Synthesis and biomedical applications of highly porous metal-organic frameworks
,”
Molecules
27
(
19
),
6585
(
2022
).
15
W.
Chai
,
X.
Chen
,
J.
Liu
et al, “
Recent progress in functional metal-organic frameworks for bio-medical application
,”
Regener. Biomater.
11
,
rbad115
(
2024
).
16
Abánades Lázaro
and
R. S.
Forgan
, “
Application of zirconium MOFs in drug delivery and biomedicine
,”
Coord. Chem. Rev.
380
,
230
259
(
2019
).
17
D.
Zhao
,
W.
Zhang
,
Z. H.
Wu
, and
H.
Xu
, “
Nanoscale metal–organic frameworks and their nanomedicine applications
,”
Front. Chem.
9
,
834171
(
2022
).
18
H. S.
Wang
,
Y. H.
Wang
, and
Y.
Ding
, “
Development of biological metal-organic frameworks designed for biomedical applications: From bio-sensing/bio-imaging to disease treatment
,”
Nanoscale Adv.
2
(
9
),
3788
3797
(
2020
).
19
A. A.
Vodyashkin
,
A. V.
Sergorodceva
,
P.
Kezimana
, and
Y. M.
Stanishevskiy
, “
Metal-organic framework (MOF)—A universal material for biomedicine
,”
Int. J. Mol. Sci.
24
(
9
),
7819
(
2023
).
20
S. T.
Meek
,
J. A.
Greathouse
, and
M. D.
Allendorf
, “
Metal-organic frameworks: A rapidly growing class of versatile nanoporous materials
,”
Adv. Mater.
23
(
2
),
249
267
(
2011
).
21
H. C.
Zhou
,
J. R.
Long
, and
O. M.
Yaghi
, “
Introduction to metal-organic frameworks
,”
Chem. Rev.
112
(
2
),
673
674
(
2012
).
22
H.
Furukawa
,
K. E.
Cordova
,
M.
O'Keeffe
, and
O. M.
Yaghi
, “
The chemistry and applications of metal-organic frameworks
,”
Science
341
(
6149
),
1230444
(
2013
).
23
T. R.
Cook
,
Y. R.
Zheng
, and
P. J.
Stang
, “
Metal-organic frameworks and self-assembled supramolecular coordination complexes: Comparing and contrasting the design, synthesis, and functionality of metal-organic materials
,”
Chem. Rev.
113
(
1
),
734
777
(
2013
).
24
Y.
Feng
,
Q.
Chen
,
M.
Jiang
, and
J.
Yao
, “
Tailoring the properties of UiO-66 through defect engineering: A review
,”
Ind. Eng. Chem. Res.
58
(
38
),
17646
17659
(
2019
).
25
T.
Jia
,
Y.
Gu
, and
F.
Li
, “
Progress and potential of metal-organic frameworks (MOFs) for gas storage and separation: A review
,”
J. Environ. Chem. Eng.
10
(
5
),
108300
(
2022
).
26
J.
Zheng
,
X.
Cui
,
Q.
Yang
,
Q.
Ren
,
Y.
Yang
, and
H.
Xing
, “
Shaping of ultrahigh-loading MOF pellet with a strongly anti-tearing binder for gas separation and storage
,”
Chem. Eng. J.
354
,
1075
1082
(
2018
).
27
P. M.
Bhatt
,
V.
Guillerm
,
S. J.
Datta
,
A.
Shkurenko
, and
M.
Eddaoudi
, “
Topology meets reticular chemistry for chemical separations: MOFs as a case study
,”
Chem
6
(
7
),
1613
1633
(
2020
).
28
B. J.
Furlong
and
M. J.
Katz
, “
Bistable dithienylethene-based metal-organic framework illustrating optically induced changes in chemical separations
,”
J. Am. Chem. Soc.
139
(
38
),
13280
13283
(
2017
).
29
S.
Jin
, “
How to effectively utilize MOFs for electrocatalysis
,”
ACS Energy Lett.
4
(
6
),
1443
1445
(
2019
).
30
P.
Gao
,
X.-Y.
Sun
,
B.
Liu
,
H.-T.
Lian
,
X.-Q.
Liu
, and
J.-S.
Shen
, “
Cu MOF-based catalytic sensing for formaldehyde
,”
J. Mater. Chem. C
6
(
30
),
8105
8114
(
2018
).
31
M. X.
Wu
,
C.
Wei
,
X. H.
Wang
,
Q. Q.
Xia
,
H.
Wang
, and
X.
Liu
, “
Construction and sensing amplification of raspberry-shaped MOF@MOF
,”
Inorg. Chem.
61
(
11
),
4705
4713
(
2022
).
32
Q.
Cao
,
Y.
Xiao
,
N.
Liu
et al, “
Synthesis of yolk/shell heterostructures MOF@MOF as biomimetic sensing platform for catechol detection
,”
Sens. Actuators, B
329
,
129133
(
2021
).
33
M.
Alvaro
,
E.
Carbonell
,
B.
Ferrer
,
F. X.
Llabrés i Xamena
, and
H.
Garcia
, “
Semiconductor behavior of a metal-organic framework (MOF)
,”
Chem.—Eur. J.
13
(
18
),
5106
5112
(
2007
).
34
C.
Zhao
,
X.
Pan
,
Z.
Wang
, and
C.-C.
Wang
, “
1 + 1 > 2: A critical review of MOF/bismuth-based semiconductor composites for boosted photocatalysis
,”
Chem. Eng. J.
417
,
128022
(
2021
).
35
K.
Naskar
,
A. K.
Bhanja
,
S.
Paul
,
K.
Pal
, and
C.
Sinha
, “
Trace quantity detection of H2PO4 by fluorescent metal–organic framework (F-MOF) and bioimaging study
,”
Cryst. Growth Des.
20
(
10
),
6453
6460
(
2020
).
36
C.
Hao
,
X.
Wu
,
M.
Sun
et al, “
Chiral core-shell upconversion nanoparticle@MOF nanoassemblies for quantification and bioimaging of reactive oxygen species in vivo
,”
J. Am. Chem. Soc.
141
(
49
),
19373
19378
(
2019
).
37
M.
Pourmadadi
,
M. M.
Eshaghi
,
S.
Ostovar
et al, “
UiO-66 metal-organic framework nanoparticles as gifted MOFs to the biomedical application: A comprehensive review
,”
J. Drug Delivery Sci. Technol.
76
,
103758
(
2022
).
38
K.
Chattopadhyay
,
M.
Mandal
, and
D. K.
Maiti
, “
A review on zirconium-based metal–organic frameworks: Synthetic approaches and biomedical applications
,”
Mater. Adv.
5
(
1
),
51
67
(
2024
).
39
W.
Zhao
,
J.
Deng
,
Y.
Ren
,
L.
Xie
,
W.
Li
et al, “
Antibacterial application and toxicity of metal–organic frameworks
,”
Nanotoxicology
15
(
3
),
311
330
(
2020
).
40
S.
Xian
,
Y.
Lin
,
H.
Wang
, and
J.
Li
, “
Calcium-based metal–organic frameworks and their potential applications
,”
Small
17
,
2005165
(
2020
).
41
J.
Liu
,
D.
Wu
,
N.
Zhu
,
Y.
Wu
, and
G.
Li
, “
Antibacterial mechanisms and applications of metal-organic frameworks and their derived nanomaterials
,”
Trends Food Sci. Technol.
109
,
413
(
2021
).
42
C. Y.
Sun
,
C.
Qin
,
X. L.
Wang
, and
Z. M.
Su
, “
Metal-organic frameworks as potential drug delivery systems
,”
Expert Opin. Drug Delivery
10
(
1
),
89
101
(
2013
).
43
X.
Cai
,
X.
Bao
, and
Y.
Wu
, “
Metal-organic frameworks as intelligent drug nanocarriers for cancer therapy
,”
Pharmaceutics
14
(
12
),
2641
(
2022
).
44
Y.
Sun
,
L.
Zheng
,
Y.
Yang
et al, “
Metal-organic framework nanocarriers for drug delivery in biomedical applications
,”
Nanomicro Lett.
12
(
1
),
103
(
2020
).
45
S.
Mallakpour
,
E.
Nikkhoo
, and
C. M.
Hussain
, “
Application of MOF materials as drug delivery systems for cancer therapy and dermal treatment
,”
Coord. Chem. Rev.
451
,
214262
(
2022
).
46
C.
Yang
,
X.
Wu
,
J.
Liu
, and
B.
Ding
, “
Stimuli-responsive nucleic acid nanostructures for efficient drug delivery
,”
Nanoscale
14
(
48
),
17862
17870
(
2022
).
47
G.
Liu
,
J. F.
Lovell
,
L.
Zhang
, and
Y.
Zhang
, “
Stimulus-responsive nanomedicines for disease diagnosis and treatment
,”
Int. J. Mol. Sci.
21
(
17
),
6380
(
2020
).
48
S.
Khan
,
Y.
Vahdani
,
A.
Hussain
et al, “
Polymeric micelles functionalized with cell penetrating peptides as potential pH-sensitive platforms in drug delivery for cancer therapy: A review
,”
Arab. J. Chem.
14
(
8
),
103264
(
2021
).
49
M.
Parsaei
,
K.
Akhbari
,
E.
Tylianakis
, and
G. E.
Froudakis
, “
Effects of fluorinated functionalization of linker on quercetin encapsulation, release and hela cell cytotoxicity of Cu-based MOFs as smart pH-stimuli nanocarriers
,”
Chem.—Eur. J.
30
,
e202301630
(
2023
).
50
J.
Schnabel
,
R.
Ettlinger
, and
H.
Bunzen
, “
Zn-MOF-74 as pH-responsive drug-delivery system of arsenic trioxide
,”
ChemNanoMat
6
(
8
),
1229
1236
(
2020
).
51
J.
Wang
,
L.
Liu
,
L.
Yin
, and
L.
Chen
, “
Acid-triggered synergistic chemo-photodynamic therapy systems based on metal-coordinated supramolecular interaction
,”
J. Biomed. Mater. Res., Part A
106
(
11
),
2955
2962
(
2018
).
52
Q.
Ding
,
Y.
Liu
,
C.
Shi
,
J.
Xiao
,
W.
Dai
et al, “
Applications of ROS-induced Zr-MOFs platform in multimodal synergistic therapy
,”
Mini-Rev. Med. Chem.
21
,
1718
(
2021
).
53
M.
Rodr??guez
,
J. L.
Vila-Jato
, and
D.
Torres
, “
Design of a new multiparticulate system for potential site-specific and controlled drug delivery to the colonic region
,”
J. Controlled Release
55
(
1
),
67
(
1998
).
54
D.
Ni
,
J.
Lin
,
N.
Zhang
et al, “
Combinational application of metal-organic frameworks-based nanozyme and nucleic acid delivery in cancer therapy
,”
WIREs Nanomed. Nanobiotechnol.
14
(
3
),
e1773
(
2022
).
55
C.
Yuan
,
Y.
Li
,
Z.
Xu
et al, “
Imaging-guided synergistic photo-chemotherapy using doxorubicin-loaded gadolinium porphyrin-based metal–organic framework nanosheets
,”
ACS Appl. Nano Mater.
5
(
10
),
15318
15327
(
2022
).
56
J.
Huang
,
Z.
Xu
,
Y.
Jiang
, et al “
Metal organic framework-coated gold nanorod as an on-demand drug delivery platform for chemo-photothermal cancer therapy
”,
J Nanobiotechnology.
19
,
219
(
2021
).
57
K.
Jiang
,
L.
Zhang
,
Q.
Hu
et al, “
Pressure controlled drug release in a Zr-cluster-based MOF
,”
J. Mater. Chem. B
4
(
39
),
6398
6401
(
2016
).
58
Y.
Liu
,
P.
Lei
,
X.
Liao
, and
C.
Wang
, “
Nanoscale metal–organic frameworks as smart nanocarriers for cancer therapy
,”
J. Nanostruct. Chem.
14
,
1
19
(
2022
).
59
M. E.
Hoque
,
N. N.
Showva
,
M.
Ahmed
et al, “
Titanium and titanium alloys in dentistry: Current trends, recent developments, and future prospects
,”
Heliyon
8
(
11
),
e11300
(
2022
).
60
W.
Zhou
,
X.
Peng
,
Y.
Ma
et al, “
Two-staged time-dependent materials for the prevention of implant-related infections
,”
Acta Biomater.
101
,
128
140
(
2020
).
61
R. C. S.
Silva
,
A.
Agrelli
,
A. N.
Andrade
et al, “
Titanium dental implants: An overview of applied nanobiotechnology to improve biocompatibility and prevent infections
,”
Materials
15
(
9
),
3150
(
2022
).
62
M.
Godoy-Gallardo
,
U.
Eckhard
,
L. M.
Delgado
et al, “
Antibacterial approaches in tissue engineering using metal ions and nanoparticles: From mechanisms to applications
,”
Bioact. Mater.
6
(
12
),
4470
4490
(
2021
).
63
X.
Ma
,
S.
Zhou
,
X.
Xu
, and
Q.
Du
, “
Copper-containing nanoparticles: Mechanism of antimicrobial effect and application in dentistry-a narrative review
,”
Front. Surg.
9
,
905892
(
2022
).
64
P.
Anna
,
G.
Angeliki
,
P.
George
,
P.
Anastasia
, and
G. N.
Karanikolos
, “
Ion-based metal/graphene antibacterial agents comprising of mono-ionic and bi-ionic silver and copper species
,”
Langmuir
34
,
11156
(
2018
).
65
J.
Yan
,
D.
Xia
,
W.
Zhou
et al, “
pH-responsive silk fibroin-based CuO/Ag micro/nano coating endows polyetheretherketone with synergistic antibacterial ability, osteogenesis, and angiogenesis
,”
Acta Biomater.
115
,
220
234
(
2020
).
66
N.
Kaur
,
P.
Tiwari
,
K. S.
Kapoor
,
A. K.
Saini
,
V.
Sharma
, and
S. M.
Mobin
, “
Metal–organic framework based antibiotic release and antimicrobial response: An overview
,”
CrystEngComm
22
,
7513
(
2020
).
67
Y.
Zhu
,
K.
Mao
,
J.
Rong
et al, “
In situ deposition of Ag/Zn-MOFs on the surface of non-woven fabrics for effective antibacterial activity
,”
J. Ind. Text.
52
,
152808372211096
(
2022
).
68
P.
Cao
,
X.
Wu
,
W.
Zhang
,
L.
Zhao
,
W.
Sun
, and
Z.
Tang
, “
Killing oral bacteria using metal–organic frameworks
,”
Ind. Eng. Chem. Res.
59
(
4
),
1559
1567
(
2020
).
69
M.
Shen
,
F.
Forghani
,
X.
Kong
et al, “
Antibacterial applications of metal–organic frameworks and their composites
,”
Compr. Rev. Food Sci. Food Saf.
19
(
4
),
1397
(
2020
).
70
J.
Chen
,
X.
Zhang
,
C.
Huang
et al, “
Osteogenic activity and antibacterial effect of porous titanium modified with metalorganic framework films
,”
J. Biomed. Mater. Res., Part A
105
,
834
(
2017
).
71
Y.
Zhou
,
S.
Han
et al, “
Accelerated host angiogenesis and immune responses by ion release from mesoporous bioactive glass
,”
J. Mater. Chem. B
6
(
20
),
3274
3284
(
2018
).
72
H.
Zhang
,
Y.
Zhao
,
Y.
Zhang
,
R.
Hang
,
X.
Yao
, and
R.
Hang
, “
Exosomes derived from macrophages upon cobalt ion stimulation promote angiogenesis
,”
Colloids Surf., B
203
,
111742
(
2021
).
73
H.
Qin
,
J.
Weng
,
B.
Zhou
et al, “
Magnesium ions promote in vitro rat bone marrow stromal cell angiogenesis through notch signaling
,”
Biol. Trace Elem. Res.
201
,
2823
2842
(
2022
).
74
X.
Wang
,
B.
Gao
,
S.
Xia
et al, “
Surface-functionalized zinc MOFs delivering zinc ion and hydrogen sulfide as tailored anti-hindlimb ischemic nanomedicine
,”
Appl. Mater. Today
32
,
101843
(
2023
).
75
C.
Xu
,
Y.
Kang
,
S.
Guan
,
X.
Dong
,
D.
Jiang
, and
M.
Qi
, “
Iron-based metal–organic framework as a dual cooperative release system for enhanced vascularization and bone regeneration
,”
Chin. Chem. Lett.
34
(
5
),
107825
(
2023
).
76
X.
Chen
,
S.
He
,
Y.
Dong
et al, “
Cobalt-doped layered hydroxide coating on titanium implants promotes vascularization and osteogenesis for accelerated fracture healing
,”
Mater. Today Bio
24
,
100912
(
2024
).
77
Y.
Si
,
H.
Liu
,
H.
Yu
,
X.
Jiang
, and
D.
Sun
, “
MOF-derived CuO@ZnO modified titanium implant for synergistic antibacterial ability, osteogenesis and angiogenesis
,”
Colloids Surf., B
219
,
112840
(
2022
).
78
D.
Xiao
,
F.
Yang
,
Q.
Zhao
et al, “
Fabrication of a Cu/Zn co-incorporated calcium phosphate scaffold-derived GDF-5 sustained release system with enhanced angiogenesis and osteogenesis properties
,”
RSC Adv.
8
(
52
),
29526
29534
(
2018
).
79
M.
Wang
,
X.
Zhou
,
Y.
Li
et al, “
Triple-synergistic MOF-nanozyme for efficient antibacterial treatment
,”
Bioact. Mater.
17
,
289
299
(
2022
).
80
B.
Yan
,
J.
Tan
,
H.
Zhang
et al, “
Constructing fluorine-doped Zr-MOF films on titanium for antibacteria, anti-inflammation, and osteogenesis
,”
Biomater. Adv.
134
,
112699
(
2022
).
81
X.
Shen
,
Y.
Zhang
,
P.
Ma
et al, “
Fabrication of magnesium/zinc-metal organic framework on titanium implants to inhibit bacterial infection and promote bone regeneration
,”
Biomaterials
212
,
1
16
(
2019
).
82
A.
Gomez
,
A.
Serrano
,
E.
Salero
et al, “
Tumor necrosis factor-alpha and interferon-gamma induce inflammasome-mediated corneal endothelial cell death
,”
Exp. Eye Res.
207
,
108574
(
2021
).
83
U. N.
Das
, “
Inflammatory bowel disease as a disorder of an imbalance between pro- and anti-inflammatory molecules and deficiency of resolution bioactive lipids
,”
Lipids Health Dis.
15
,
11
(
2016
).
84
B.
Yang
,
H.
Yao
,
J.
Yang
,
C.
Chen
, and
J.
Shi
, “
Construction of a two-dimensional artificial antioxidase for nanocatalytic rheumatoid arthritis treatment
,”
Nat. Commun.
13
(
1
),
1988
(
2022
).
85
N.
Joseph
,
H. D.
Lawson
,
K. J.
Overholt
et al, “
Synthesis and characterization of CaSr-metal organic frameworks for biodegradable orthopedic applications
,”
Sci. Rep.
9
(
1
),
13024
(
2019
).
86
L.
Wang
,
F.
Dai
,
Y.
Yang
, and
Z.
Zhang
, “
Zeolitic imidazolate framework-8 with encapsulated naringin synergistically improves antibacterial and osteogenic properties of Ti implants for osseointegration
,”
ACS Biomater. Sci. Eng.
8
,
3797
(
2022
).
87
Z.
Ling
,
B.
Cheng
, and
X.
Tao
, “
Epithelial-to-mesenchymal transition in oral squamous cell carcinoma: Challenges and opportunities
,”
Int. J. Cancer
148
(
7
),
1548
1561
(
2021
).
88
R.
Baillie
,
S. T.
Tan
, and
T.
Itinteang
, “
Cancer stem cells in oral cavity squamous cell carcinoma: A review
,”
Front. Oncol.
7
,
112
(
2017
).
89
X. G.
Wang
,
Z. Y.
Dong
,
H.
Cheng
et al, “
A multifunctional metal-organic framework based tumor targeting drug delivery system for cancer therapy
,”
Nanoscale
7
(
38
),
16061
16070
(
2015
).
90
A.
Klimpel
,
T.
Lutzenburg
, and
I.
Neundorf
, “
Recent advances of anti-cancer therapies including the use of cell-penetrating peptides
,”
Curr. Opin. Pharmacol.
47
,
8
13
(
2019
).
91
A. K.
Deshantri
,
A.
Varela Moreira
,
V.
Ecker
et al, “
Nanomedicines for the treatment of hematological malignancies
,”
J. Controlled Release
287
,
194
215
(
2018
).
92
A.
Usmani
,
A.
Mishra
, and
M.
Ahmad
, “
Nanomedicines: A theranostic approach for hepatocellular carcinoma
,”
Artif. Cells, Nanomed., Biotechnol.
46
(
4
),
680
690
(
2018
).
93
T.
Xu
, “
Research progress based on regulation of tumor microenvironment redox and drug-loaded metal-organic frameworks
,”
Oxid. Med. Cell. Longevity
2022
,
7302883
.
94
Y.
Lee
,
S.
Lee
, and
S.
Jon
, “
Biotinylated bilirubin nanoparticles as a tumor microenvironment-responsive drug delivery system for targeted cancer therapy
,”
Adv. Sci.
5
(
6
),
1800017
(
2018
).
95
G.
Tan
,
Y.
Zhong
,
L.
Yang
,
Y.
Jiang
,
J.
Liu
, and
F.
Ren
, “
A multifunctional MOF-based nanohybrid as injectable implant platform for drug synergistic oral cancer therapy
,”
Chem. Eng. J.
390
,
124446
(
2020
).
96
C.
Wang
,
F.
Xue
,
M.
Wang
,
L.
An
,
D.
Wu
, and
Q.
Tian
, “
2D Cu-bipyridine MOF nanosheet as an agent for colon cancer therapy: A three-in-one approach for enhancing chemodynamic therapy
,”
ACS Appl. Mater. Interfaces
14
(
34
),
38604
38616
(
2022
).
97
Z.
Jin
,
P.
Zhao
,
J.
Zhang
et al, “
Intelligent metal carbonyl metal–organic framework nanocomplex for fluorescent traceable H2O2-triggered CO delivery
,”
Chem.—Eur. J.
24
(
45
),
11667
11674
(
2018
).
98
X.
Di
,
Z.
Pei
,
Y.
Pei
, and
T. D.
James
, “
Tumor microenvironment-oriented MOFs for chemodynamic therapy
,”
Coord. Chem. Rev.
484
,
215098
(
2023
).
99
P.
Pandit
,
S.
Bhagat
,
P.
Rananaware
et al, “
Iron oxide nanoparticle encapsulated; folic acid tethered dual metal organic framework-based nanocomposite for MRI and selective targeting of folate receptor expressing breast cancer cells
,”
Microporous Mesoporous Mater.
340
,
112008
(
2022
).
100
A.
Ahmed
,
A.
Karami
,
R.
Sabouni
,
G. A.
Husseini
, and
V.
Paul
, “
pH and ultrasound dual-responsive drug delivery system based on PEG–folate-functionalized Iron-based metal–organic framework for targeted doxorubicin delivery
,”
Colloids Surf,. A
626
,
127062
(
2021
).
101
D.
Zhou
,
Y.
Chen
,
W.
Bu
et al, “
Modification of metal-organic framework nanoparticles using dental pulp mesenchymal stem cell membranes to target oral squamous cell carcinoma
,”
J. Colloid Interface Sci.
601
,
650
660
(
2021
).
102
J.
Deng
,
S.
Xu
,
W.
Hu
,
X.
Xun
,
L.
Zheng
, and
M.
Su
, “
Tumor targeted, stealthy and degradable bismuth nanoparticles for enhanced X-ray radiation therapy of breast cancer
,”
Biomaterials
154
,
24
33
(
2018
).
103
J.
Liu
,
Y.
Yang
,
W.
Zhu
et al, “
Nanoscale metal–organic frameworks for combined photodynamic & radiation therapy in cancer treatment
,”
Biomaterials
97
,
1
9
(
2016
).
104
S.
Tsuchida
,
M.
Satoh
,
M.
Takiwaki
, and
F.
Nomura
, “
Ubiquitination in periodontal disease: A review
,”
Int. J. Mol. Sci.
18
(
7
),
1476
(
2017
).
105
H. R.
Rajeshwari
,
D.
Dhamecha
,
S.
Jagwani
et al, “
Local drug delivery systems in the management of periodontitis: A scientific review
,”
J. Controlled Release
307
,
393
409
(
2019
).
106
X.
Li
,
Y.
Liu
,
X.
Yang
,
C.
Li
, and
Z.
Song
, “
The oral microbiota: Community composition, influencing factors, pathogenesis, and interventions
,”
Front. Microbiol.
13
,
895537
(
2022
).
107
E.
Caselli
,
C.
Fabbri
,
M.
D'Accolti
et al, “
Defining the oral microbiome by whole-genome sequencing and resistome analysis: The complexity of the healthy picture
,”
BMC Microbiol.
20
(
1
),
120
(
2020
).
108
M.
Amato
,
S.
Santonocito
,
A.
Polizzi
et al, “
Local delivery and controlled release drugs systems: A new approach for the clinical treatment of periodontitis therapy
,”
Pharmaceutics
15
(
4
),
1312
(
2023
).
109
D. S.
Yang
,
K. T.
You
,
S. H.
Pi
,
M. Y.
Lee
,
Y. O.
You
,
H. K.
You
, and
H. S.
Shin
, “
Effects of several biodegradable controlled-release local delivery drugs on the treatment of periodontitis
,”
J. Korean Acad. Periodontol.
29
(
4
),
725
735
(
2014
).
110
Y.
Wei
,
Y.
Deng
,
S.
Ma
,
M.
Ran
,
Y.
Jia
et al, “
Local drug delivery systems as therapeutic strategies against periodontitis: A systematic review
,”
J. Controlled Release
333
(
4
),
269
(
2021
).
111
D.
Steinberg
and
M.
Friedman
, “
Sustained-release delivery of antimicrobial drugs for the treatment of periodontal diseases: Fantasy or already reality?
,”
Periodontology 2000
84
(
1
),
176
187
(
2020
).
112
B.
Wang
,
J.
Wang
,
J.
Shao
et al, “
A tunable and injectable local drug delivery system for personalized periodontal application
,”
J. Controlled Release
324
,
134
(
2020
).
113
D. F.
Sava Gallis
,
K. S.
Butler
,
J. O.
Agola
,
C. J.
Pearce
, and
A. A.
McBride
, “
Antibacterial countermeasures via metal–organic framework-supported sustained therapeutic release
,”
ACS Appl. Mater. Interfaces
11
(
8
),
7782
7791
(
2019/02/ 2019
).
114
H.
Li
,
N.
Lv
,
X.
Li
et al, “
Composite CD-MOF nanocrystals-containing microspheres for sustained drug delivery
,”
Nanoscale
9
(
22
),
7454
7463
(
2017
).
115
M. A.
Lopez
,
P. C.
Passarelli
,
M.
Marra
et al, “
Antimicrobial efficacy of photodynamic therapy (PDT) in periodontitis and peri-implantitis: A systematic review
,”
J. Biol. Regul. Homeost. Agents
34
(
5 Suppl 3
),
59
65
(
2020
), part of Special Issue: Technology in Medicine.
116
X.
Gao
,
G.
Ji
,
R.
Cui
, and
Z.
Liu
, “
Controlled synthesis of MOFs@MOFs core–shell structure for photodynamic therapy and magnetic resonance imaging
,”
Mater. Lett.
237
,
197
199
(
2019
).
117
Y.
Zhao
,
X.
Jiang
,
X.
Liu
,
X.
Liu
,
Z.
Liu
, and
X.
Liu
, “
Application of photo-responsive metal-organic framework in cancer therapy and bioimaging
,”
Front. Bioeng. Biotechnol.
10
,
1031986
(
2022
).
118
Y.
Ye
,
Y.
Zhao
,
Y.
Sun
, and
J.
Cao
, “
Recent progress of metal-organic framework-based photodynamic therapy for cancer treatment
,”
Int. J. Nanomed.
17
,
2367
2395
(
2022
).
119
X.
Zheng
,
J.
Zhong
,
M. Y.
Dong
,
Y.
Wen
, and
A. Z.
Chen
, “
Synthesis of porphyrin-based 2D ytterbium metal organic frameworks for efficient photodynamic therapy
,”
RSC Adv.
12
(
53
),
34318
34324
(
2022
).
120
J.
Li
,
S.
Song
,
J.
Meng
et al, “
2D MOF periodontitis photodynamic ion therapy
,”
J. Am. Chem. Soc.
143
(
37
),
15427
15439
(
2021
).
121
X.
Han
,
G.
Boix
,
M.
Balcerzak
et al, “
Antibacterial films based on MOF composites that release iodine passively or upon triggering by near-infrared light
,”
Adv. Funct. Mater.
32
,
2112902
(
19
) (
2022
).
122
F. F.
Farook
,
H.
Alodwene
,
R.
Alharbi
et al, “
Reliability assessment between clinical attachment loss and alveolar bone level in dental radiographs
,”
Clin. Exp. Dent. Res.
6
(
6
),
596
601
(
2020
).
123
K.
Sun
,
H.
Shen
,
Y.
Liu
,
H.
Deng
,
H.
Chen
, and
Z.
Song
, “
Assessment of alveolar bone and periodontal status in peritoneal dialysis patients
,”
Front. Physiol.
12
,
759056
(
2021
).
124
Y.
Liang
,
X.
Luan
, and
X.
Liu
, “
Recent advances in periodontal regeneration: A biomaterial perspective
,”
Bioact. Mater.
5
(
2
),
297
308
(
2020
).
125
Y.
Zhu
,
L.
Zhao
, and
T.
Ngai
, “
Multiphasic membranes/scaffolds for periodontal guided tissue regeneration
,”
Macromol. Mater. Eng.
308
,
2300081
(
10
) (
2023
).
126
M. A.
Fardjahromi
,
H.
Nazari
,
S. M. A.
Tafti
,
A.
Razmjou
,
S.
Mukhopadhyay
, and
M. E.
Warkiani
, “
Metal-organic framework-based nanomaterials for bone tissue engineering and wound healing
,”
Mater. Today Chem.
23
,
23
(
2022
).
127
M.
Shyngys
,
J.
Ren
,
X.
Liang
,
J.
Miao
,
A.
Blocki
, and
S.
Beyer
, “
Metal-organic framework (MOF)-based biomaterials for tissue engineering and regenerative medicine
,”
Front. Bioeng. Biotechnol.
9
,
603608
(
2021
).
128
F.
Zou
,
J.
Jiang
,
F.
Lv
,
X.
Xia
, and
X.
Ma
, “
Preparation of antibacterial and osteoconductive 3D-printed PLGA/Cu(I)@ZIF-8 nanocomposite scaffolds for infected bone repair
,”
J. Nanobiotechnol.
18
(
1
),
39
(
2020
).
129
B.
Wang
,
H.
Chen
,
S.
Peng
et al, “
Multifunctional magnesium-organic framework doped biodegradable bone cement for antibacterial growth, inflammatory regulation and osteogenic differentiation
,”
J. Mater. Chem. B
11
(
13
),
2872
2885
(
2023
).
130
B.
Tao
,
W.
Zhao
,
C.
Lin
et al, “
Surface modification of titanium implants by ZIF-8@Levo/LBL coating for inhibition of bacterial-associated infection and enhancement of in vivo osseointegration
,”
Chem. Eng. J.
390
,
124621
(
2020
).
131
Y.
Wang
,
D.
Zhang
,
M.
Jia
et al, “
ZIF-8 nanoparticles coated with macrophage-derived microvesicles for sustained, targeted delivery of dexamethasone to arthritic joints
,”
J. Drug Targeting
30
(
9
),
1006
1016
(
2022/10/06 2022
).
132
R.
Ma
,
Y.
Su
,
R.
Cao
,
K.
Wang
, and
P.
Yang
, “
Enhanced osteogenic activity and bone repair ability of PLGA/MBG scaffolds doped with ZIF-8 nanoparticles loaded with BMP-2
,”
Int. J. Nanomed.
18
,
5055
5072
(
2023
).
133
M. D.
Telgerd
,
M.
Sadeghinia
,
G.
Birhanu
et al, “
Enhanced osteogenic differentiation of mesenchymal stem cells on metal–organic framework based on copper, zinc, and imidazole coated poly-l-lactic acid nanofiber scaffolds
,”
J. Biomed. Mater. Res., Part A
107
,
1841
(
2019
).
134
A. A.
Sadek
,
M.
Abd-Elkareem
,
H. N.
Abdelhamid
,
S.
Moustafa
, and
K.
Hussein
, “
Enhancement of critical-sized bone defect regeneration using UiO-66 nanomaterial in rabbit femurs
,”
BMC Vet. Res.
18
(
1
),
260
(
2022
).
135
J.
Rodriguez-Martinez
,
M. J.
Sanchez-Martin
, and
M.
Valiente
, “
Efficient controlled release of cannabinoids loaded in γ-CD-MOFs and DPPC liposomes as novel delivery systems in oral health
,”
Microchim. Acta
190
(
4
),
125
(
2023
).
136
L. M.
Santos
,
I. M.
Bernardino
,
A. V.
Ferreira Porto
,
K. G.
Nórbrega Barbosa
,
L.
Marques da Nóbrega
, and
S.
d’Avila
, “
Aggression using a knife or other sharp instruments and oral-maxillofacial traumas: Incidence, risk factors and epidemiological trends
,”
J. Oral Maxillofac. Surg.
76
,
1953
(
2018
).
137
T. U.
Khan
,
S.
Rahat
,
Z. A.
Khan
,
L.
Shahid
,
S. S.
Banouri
, and
N.
Muhammad
, “
Etiology and pattern of maxillofacial trauma
,”
PLoS One
17
(
9
),
e0275515
(
2022
).
138
M.
Siavash
and
A.
Noursina
, “
The ideal wound dressing
,”
Burns
49
(
7
),
1780
1781
(
2023
).
139
A.
Moeini
,
P.
Pedram
,
P.
Makvandi
,
M.
Malinconico
, and
G.
Gomez d’Ayala
, “
Wound healing and antimicrobial effect of active secondary metabolites in chitosan-based wound dressings: A review
,”
Carbohydr. Polym.
233
,
115839
(
2020
).
140
R.
Rezaee
,
M.
Montazer
,
A.
Mianehro
, and
M.
Mahmoudirad
, “
Single-step synthesis and characterization of Zr-MOF onto wool fabric: Preparation of antibacterial wound dressing with high absorption capacity
,”
Fibers Polym.
23
(
2
),
404
412
(
2021
).
141
F.
Cheng
,
X.
Yi
,
J.
Dai
et al, “
Photothermal MXene@Zn-MOF-decorated bacterial cellulose-based hydrogel wound dressing for infectious wound healing
,”
Cell Rep. Phys. Sci.
4
(
10
),
101619
(
2023
).
142
C.
Liu
,
C.
He
,
M.
Li
et al, “
2D MOF based-heterostructure with hierarchical architecture as antibacterial wound dressing
,”
Int. J. Pharm.
651
,
123745
(
2024
).
143
A.
Bigham
,
N.
Islami
,
A.
Khosravi
,
A.
Zarepour
,
S.
Iravani
, and
A.
Zarrabi
, “
MOFs and MOF-based composites as next-generation materials for wound healing and dressings
,”
Small
e2311903
(published online
2024
).
144
M.
Zhang
,
G.
Wang
,
D.
Wang
et al, “
Ag@MOF-loaded chitosan nanoparticle and polyvinyl alcohol/sodium alginate/chitosan bilayer dressing for wound healing applications
,”
Int. J. Biol. Macromol.
175
,
481
494
(
2021
).
145
S.
Javanbakht
,
M.
Nabi
,
M.
Shadi
,
M. M.
Amini
, and
A.
Shaabani
, “
Carboxymethyl cellulose/tetracycline@UiO-66 nanocomposite hydrogel films as a potential antibacterial wound dressing
,”
Int. J. Biol. Macromol.
188
,
811
819
(
2021
).