Next Article in Journal
From Nature to Nanomedicine: Green-Synthesized Metal Oxide Nanoparticles for Oral Cancer Drug Delivery
Previous Article in Journal
Miniaturized Frustum-Cone Triboelectric Hydrophone Based on a Thin Film Perforated Tube Structure
Previous Article in Special Issue
Comparative Carcinogenicity of Double-Walled Carbon Nanotubes of Different Lengths Administered by Intratracheal Installation into Rat Lungs
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Nanotoxicity of Porous Silica Nanoparticles: Physicochemical Properties and Mechanistic Cellular Endpoints

1
Leicester School of Allied Health Sciences, De Montfort University, Leicester LE1 9BH, UK
2
Division of Pharmacy and Optometry, School of Health Sciences, University of Manchester, Manchester M13 9PL, UK
3
Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK
*
Author to whom correspondence should be addressed.
Nanomaterials 2025, 15(23), 1766; https://doi.org/10.3390/nano15231766
Submission received: 15 October 2025 / Revised: 8 November 2025 / Accepted: 18 November 2025 / Published: 25 November 2025

Abstract

This review provides a comprehensive overview of the fundamental aspects of nanoparticles (NPs), emphasizing their physicochemical properties and biological interactions, with particular focus on porous silica nanoparticles (PSNs). The review provides information on the Safe-by-design (SbD) S.A.F.E. (Standardised characterization, Assessment of biocompatibility, Facilitation of toxicity and exposure routes and Evaluation of clinical translation) framework. It discusses critical factors influencing NP toxicity and cellular uptake, including particle size, shape, pore size, surface charge, surface functionalisation, and crystallinity. The review also examines exposure routes of NPs—inhalation, dermal, oral, systemic and mucosal—and their subsequent biological effects. A key section is dedicated to the formation of the protein corona, a critical determinant of NP fate in biological systems, and its influence on circulation time, immune clearance and cellular responses. Particular attention is given to assessing the biological interactions of the PSNs and the mechanisms underlying PSN-induced cytotoxicity and genotoxicity, with a focus on the assays commonly employed to evaluate these effects. The review explores the use of gene expression profiling as a powerful tool to elucidate the molecular mechanisms underlying nanoparticle-induced cellular changes. This review aims to provide an integrated perspective on the SbD considerations and safety implications of nanomaterials. It highlights the need for a deeper understanding of complex biological interactions to establish SbD principles and enable the translation of PSNs into clinical applications. Finally, current regulatory frameworks and guidelines for testing nanomaterials, including PSNs, that support their safe and sustainable development are discussed.

Graphical Abstract

1. Introduction

Porous silica nanoparticles (PSNs) have become widely used nanocarriers in biomedical applications, particularly drug delivery. Their high surface area, tunable pore size, and easy functionalisation make them ideal candidates as delivery vehicles for a variety of therapeutic agents, including small-molecule drugs, proteins, and nucleic acids [1,2]. These properties enable controlled drug delivery, thereby improving bioavailability and reducing systemic toxicity. Additionally, their stability and tunability in biological membrane interactions contribute to their versatility across various applications [3,4]. The diverse strategies for functionalizing and loading PSNs with therapeutic agents are summarized in Figure 1, highlighting their adaptability for targeted biomedical applications.
This review is novel in that it provides an integrated, multidisciplinary perspective on PSNs, combining physicochemical characterization, biological interactions and Safe-by-Design (SbD) principles. Previous reviews have focused on individual toxicological endpoints such as cytotoxicity or genotoxicity; however, this work encompasses a broader spectrum of biological responses, including protein corona formation and immunogenicity, and links them to design parameters and exposure pathways. Furthermore. SbD approaches have been applied generally to nanomaterials; however, no comprehensive SbD principles have focused on PSNs, making this review the first to evaluate their safety within the S.A.F.E. framework and clinical translation.

1.1. Applications and Clinical Translation of PSNs

The biomedical application of PSNs represents a significant advancement in nanomedicine, connecting innovative bench-scale designs with human safety, pharmacokinetics, and efficacy considerations. Silicon dioxide has been classified as “generally recognized as safe” (GRAS) by the U.S. FDA, enabling its widespread use in food additives and cosmetic products [5,6]. This safety recognition provides a strong foundation for its clinical translation, supporting the development of PSNs as platforms for drug delivery, diagnostic imaging and other therapeutic applications. While the translation into late-phase therapeutic trials is still in its early stages, numerous first-in-human and early clinical trials have shown that silica-based nanomaterials can be safely administered at tracer or microdosing levels, display favourable pharmacokinetics, particularly renal clearance for ultrasmall silica and effectively deliver functional payloads or imaging contrast to targeted sites [7]. Table 1 shows clinical trials that have used porous or mesoporous silica nanoparticles (MSNs). The primary translational challenge involves balancing multifunctionality, such as targeting ligands, stimuli-responsive release, imaging moieties and factors like manufacturability, reproducibility, biocompatibility, and predictable in vivo degradation and clearance [5,8].
PSNs can deliver chemotherapy by encapsulating drugs such as doxorubicin, paclitaxel, and irinotecan within their porous structure and releasing them via controlled or stimulus-responsive mechanisms, such as pH or enzymes, directly to tumour cells [16,17,18,19,20,21]. While PSNs have demonstrated significant potential in preclinical studies, their clinical translation remains limited. Several studies have explored their efficacy in chemotherapy applications, highlighting their potential to enhance drug delivery and reduce systemic toxicity. Another important application of PSNs is their use as carriers for protein and peptide therapeutics. Common challenges with protein and peptide therapeutics include enzymatic degradation, poor stability, and limited cellular uptake. The properties of PSNs enable efficient loading of peptides and proteins, protection from harsh biological environments and targeted release, making them a multifunctional carrier for protein and peptide therapeutics [22,23]. MSNs can protect proteins from degradation and maintain their biological activity, facilitating targeted intracellular delivery of rapidly degrading peptides and proteins [24,25,26]. RNA and gene delivery play a vital role in modern therapeutics, allowing modulation of gene expression, inhibition of pathogenic pathways and repair of genetic abnormalities [27]. However, there is a major challenge in effective delivery due to the instability of nucleic acids, their susceptibility to enzymatic degradation, and limited cellular uptake. To overcome this, PSNs offer a promising solution, as their modifiable properties enable efficient encapsulation, protection, and targeted release [28]. Numerous studies have demonstrated that PSNs can be functionalised or loaded with fluorescent probes to aid in cellular uptake studies and cancer imaging. Owing to their high surface area and tunable porosity, PSNs can minimize photobleaching and dye leakage, thereby enhancing targeted fluorescence imaging, demonstrating their potential for diagnostics and theragnostic applications [29,30]. Furthermore, PSNs have been utilized as carriers for bioimaging agents, improving fluorescence and MRI contrast, which could enhance early disease detection and precision diagnostics. Research showed that MSNs doped with gadolinium oxide significantly improved T1 MRI contrast compared with the conventional contrast agent, gadolinium diethylene triamine pentaacetate, likely due to the structure of the MSNs, which increased surface area [31]. Lastly, PSNs have been used in antimicrobial therapy. Their high surface area, easy functionalisation, and controllable loading and release kinetics make PSNs an excellent carrier for antibiotics, antimicrobial peptides and metal-based agents [32].

1.2. Double-Edged PSNs: Potential Toxicity Risks

Although PSNs have unique and promising characteristics, a major concern in their therapeutic use is potential toxicity to the human body. Various studies have described mechanisms by which nanoparticles (NPs) may induce toxic effects, including oxidative stress, genotoxicity, inflammation, extent of cellular uptake, and disruption of cellular functions, highlighting the importance of careful design and surface modifications to mitigate potential adverse effects [33,34,35].
Silica-based NPs are considered biocompatible, but their interactions with biological systems can vary depending on their physicochemical properties, such as particle size, surface charge, and functionalisation [36]. To mitigate toxicity while maximizing therapeutic efficacy, research has focused on modifying the surface properties of PSNs to enhance their biocompatibility. PEG functionalisation, zwitterionic coatings, and biomimetic surface modifications are among the strategies used to increase NP stability, extend circulation time, and reduce immune recognition [37,38,39]. Zwitterionic coatings use molecules that have both a positive and a negative charge to produce a neutral surface which repels proteins, while biomimetic surface modifications coat NPs with natural biological components, such as cell membranes or proteins, to ensure maximum biocompatibility and reduce toxicity [40,41,42,43].
To fabricate PSNs for safe and effective biomedical applications, a better understanding of the relationship between physicochemical properties and biological responses is essential. Therefore, comprehensive characterization and rigorous safety evaluations are crucial to ensuring their successful translation into clinical use.

2. Safe-by-Design: Modulating Nanoparticle Physicochemical Properties to Control and Minimize PSN Toxicity

2.1. Safe-by-Design Principles of PSNs

Nanoparticles are at the forefront of modern nanotechnology, from biomedical applications to cosmetic uses. As the market for nanoproducts grows, the importance of nano safety procedures to mitigate potential hazards to engineers and consumers becomes increasingly crucial. SbD strategies enable a more calculated and careful approach to the clinical translation of NPs [44,45]. This approach not only benefits the patient or consumer but also reduces harmful by-products that would otherwise harm the environment. In this section, various SbD strategies approaches for PSNs have been discussed, using the S.A.F.E. PSNs framework based on the GoNanoBioMat SbD approach [46]. The S.A.F.E. framework (Figure 2) discusses Standardised characterization, Assessment of biocompatibility, Frameworks for understanding nanoparticle toxicity, exposure routes, and Relevant Model and Evaluation of clinical translation.
PSNs have unique characteristics; their biological interactions and toxicity are primarily determined by their physicochemical properties, such as size, shape, surface charge, pore size, surface functionalisation, and crystallinity. Such features dictate the interfaces between PSNs and biological membranes, cellular uptake pathways, biodistribution, and the mechanisms of toxicity. Therefore, knowledge of these parameters is critical for engineering safer and more effective nanomedicine applications [47].

2.2. Standardised Characterization

A critical aspect of safe advanced nanomaterial design lies in standardizing the physicochemical attributes that influence PSN safety profiles. Key attributes that govern therapeutic delivery and ultimately biocompatibility, include particle size, particle shape, pore size, surface charge, surface functionalisation, and crystalline structure. They influence biological interactions, including protein corona formation, cellular uptake pathways, biodistribution, and clearance [5,48,49,50].
Alterations in nanomaterial features could result in acute and chronic biological effects [51]. Standardised characterization is also important to ensure reproducibility from laboratories to scale up manufacturing. The synthesis of NPs in preclinical studies usually involves template, sol–gel or hydrothermal methods, which are known to exhibit batch-to-batch variability [52]. This poses a significant risk that clinical study results may not be reliable. Therefore, to overcome this, standardized characterization is essential to produce safe and reliable PSNs.

2.2.1. Particle Size

When engineering and producing PSNs, determining an optimal size is a significant aspect of SbD principles. As confirmed in several studies, smaller NPs (<100 nm) generally exhibit greater cellular uptake than larger particles, primarily due to their cellular uptake via endocytosis [53,54]. For example, a study found that amorphous silica nanoparticles (SiNPs) with an average diameter of 42 ± 3 nm could penetrate epidermal cells. However, particles greater than 75 nm could not penetrate human skin, even when mild barrier perturbation was induced [55]. While this enhanced permeability can be therapeutically advantageous, it also poses challenges, as small NPs (10–100 nm) are more likely to cross biological barriers, including the blood–brain barrier (BBB), suggesting that NPs could accumulate in the central nervous system and cause adverse side effects [56]. Furthermore, the renal system is the primary route of excretion for these NPs; kidney uptake may increase the risk of nephrotoxicity. Therefore, the design of PSNs must be optimized to minimize such risks by controlling particle size [57]. Particle size also influences the type of toxicity observed. Smaller particles (129.7 ± 2.5 nm) have been seen to cause cytotoxicity, while larger particles (355.4 ± 41.0 nm) can trigger increased secretion of interleukin-6 (IL-6) and tumour necrosis factor-alpha (TNF-α), and initiate inflammation [58,59]. In a comparative analysis, SiNPs of different sizes (10 nm and 100 nm) showed that the smallest particles induced the greatest cytotoxicity and oxidative stress, associated with their larger surface area-to-volume ratio, and increased reactive oxygen species (ROS) generation [60]. Similarly, research on RAW264.7 macrophages has shown that smaller amorphous silica NPs (70 nm) were more toxic than those of 300 and 1000 nm, suggesting that particle size is a factor in NP toxicity [61]. In contrast, larger NPs (>200 nm) predominantly underwent internalization by phagocytosis with slower clearance rates, leading to longer retention times in organs such as the liver and spleen [62]. These findings highlight the need for size optimization, balancing therapeutic efficiency and minimizing toxicity.

2.2.2. Particle Shape

The shape of PSNs affects cellular uptake efficiency, biodistribution, and mechanical interactions with biological membranes. Although spherical NPs are most widely studied and used because of their uniform uptake and predictable behaviour, elongated or rod-shaped NPs may exhibit different biological interactions. For example, rod-shaped MSNs showed better cellular uptake in macrophages compared to corresponding spherical NPs [63,64]. The elongated shape facilitates alignment with the cell membrane, enhancing uptake via membrane wrapping mechanisms, and induces greater mechanical stress on cellular membranes, resulting in membrane rupture, oxidative stress and higher inflammatory responses [65]. Other studies have shown that nanorods and nanowires have long circulation half-lives in vivo, as their non-spherical shape reduces opsonisation and clearance (given their relatively high aspect ratio, making membrane wrapping suboptimal) by the mononuclear phagocyte system (MPS) [66,67,68,69,70]. This suggests that while specific shapes may increase toxicity in vitro, the in vivo behavior may differ, necessitating careful evaluation of both in vitro and in vivo toxicity models.

2.2.3. Pore Size

Research has shown that larger pores (6–10 nm) enhance drug-loading capacity have accelerate release rates, and are useful for treatments [57,71]. While smaller pores (<5 nm) support prolonged drug release for the treatment of chronic diseases or long-term chemotherapy [72,73]. A study found that MSNs with 2.7 nm pores exhibited lower cytotoxicity compared to those with larger pore sizes of 7.39 nm, possibly due to reduced protein aggregation and minimal immune activation [57]. However, NPs with larger pores (>10 nm) increase surface area and reactivity, promoting interactions with cell components that generate ROS [74]. These findings highlight the importance of tuning pore size to optimize drug loading efficiency while minimizing toxicity.

2.2.4. Surface Charge

The surface charge has been shown to determine their cellular uptake, including phagocytosis, underlying toxicity and subsequent clearance. It has been widely accepted that compared to anionic NPs, cationic NPs can promote greater internalization via electrostatic attraction to negatively charged cell membranes, increase cytotoxicity, induce an immune response and upregulate apoptotic pathways [75,76]. Additionally, serum proteins can modulate these interactions. For example, a study using glioblastoma stem cells indicated that positively charged MSNs functionalised with polyethyleneimine (PEI) (PEI-MSNs) were toxic because PEI-MSNs internalized into lysosomes, rupturing lysosomal membranes [77]. Also, in another study of AgNPs and human epidermal keratinocytes (HEK), serum proteins (albumin and IgG) were found to reduce AgNP uptake of 110 nm citrate-coated AgNPs [78]. These studies collectively highlight the importance of surface charge in NP-cell interactions.
Surface modification of MSN with PEI-PEG to impart a positive charge has been proven to be one of the most effective methods to improve nanoparticle uptake and biodistribution, reduce opsonisation, and endow antitumor properties [79].
However, the decrease in opsonisation and phagocytosis allows for a longer exposure time. This potentially increases the inflammatory effect and eventually leads to programmed cell death. Variability in the inflammation index, attributed to surface charge, highlights the need for more specifically optimized dispersion methods to reduce inflammation and cell death in inhalation-based nanoparticles. This would reduce the toxic effects of NPs in downstream clinical applications.
One study investigated the effect of positively charged 50 nm MSNs suspended in BSA on the influx of neutrophils into the lungs and on the expression of pro-inflammatory genes 24 h post-MSN exposure in bronchioalveolar lavage (BAL) cells. A dramatic increase (22-fold) in the abundance of polymorphonuclear neutrophils and expression of interleukin-10 was observed compared to other types of plain and aminated synthesized MSNs, concluding that a more positively charged MSN increases the degree of inflammation within BAL cells [80]. Utilizing protein coatings to improve overall stability and neutralize the PSNs’ surface charge could potentially delay the premature release of materials from the pores of MSNs via electrostatic interactions between the amino groups on the MSNs’ surface. This method has opened a variety of doors for researchers to investigate the effect of surface charge and modify it with negatively or neutrally charged zeta potential surface functionalisation.

2.2.5. Surface Functionalisation

Attachment of functional group(s) such as amine (-NH2), carboxyl (-COOH), thiol (-SH), and PEG enables PSNs to be used in targeted therapeutics, allowing proteins or receptors on target cells, effective uptake or recognition and subsequent unloading of therapeutic material into or near the target cells [48,81,82]. Studies indicate that functional groups also determine toxicity; for example, NPs functionalised with amine groups (compared to those functionalised with carboxyl or thiol groups) often exhibit increased cytotoxicity due to their positive charge, which can disrupt cell membranes and lead to cell death [83]. However, other studies have found that functional groups have made PSNs biocompatible. For example, quercetin-loaded amine-functionalised MSNs increased loading capacity, carboxyl functionalisation prevented particle agglomeration, and thiol functionalisation enabled efficient release. This shows the potential for effective oral drug delivery in the gastrointestinal tract [84]. PEGylation is frequently used to increase biocompatibility and decrease immunogenicity. PEG coatings can enhance drug and gene delivery by increasing systemic circulation time and reducing immune recognition [85,86]. A study compared PEGylated and non-PEGylated SiNPs and found that PEGylation reduced protein adsorption and immune activation while prolonging circulation time. However, PEGylation also reduced NP uptake by cancer cells, thereby balancing immune evasion and cellular uptake [87,88,89]. Targeted functionalisation, such as antibody-conjugated NPs, has been shown to increase selectivity for cancer cells, improve drug delivery, and reduce off-target effects [90,91,92]. However, some studies suggest that over-functionalisation by ligands/polymers may lead to unintended increases in recognition by opsonins, thereby increasing phagocytosis and reducing NP effectiveness [93,94,95]. Steric hindrance, which inhibits the interaction of the NP with its biological target [96]. Surface modification-dependent increased cellular internalization and pH-responsive release can effectively deliver therapeutics to the acidic tumour microenvironment, in contrast to unfunctionalised MSNs, which would circulate longer, increasing the risk of an immune response and the non-specific release of therapeutics into the bloodstream, potentially causing apoptosis in healthy cells [97]. Another interesting study to bypass the harsh barriers of the gastrointestinal tract used hollow silica NPs doped with quantum dots and functionalised with a two-part coating. A hydrophilic succinylated casein layer was designed to penetrate and effectively break through the mucosal layer, while a cationic cell-penetrating peptide concealed beneath the casein penetrates the epithelial layer to facilitate cellular uptake. The pharmacokinetic benefits from surface modifications resulted in a 5-fold increase in cell internalization and a 40% increase in the bioavailability of the loaded drug, paclitaxel [57]. Ultimately, the choice of surface functionalisation should be considered when designing SbD delivery vehicles: these should enhance bioavailability, target malignant cells, improve stability, facilitate cell membrane interactions, enhance uptake, and minimize downstream toxicity. However, balancing functionalisation for targeting and stealth properties remains challenging, as excess modification can lead to immune recognition and clearance. Future studies focused on various surface-engineering approaches will be critical for advancing PSN-based therapeutics for clinical applications.

2.2.6. Agglomeration

PSNs are promising nanodrug carriers that require high dispersion and colloidal stability, as aggregation influences PSN internalization, biodistribution, and biological toxicity. As mentioned above, surface hydroxyl groups on PSNs, chemical modifications with proteins, polymer coatings, or phospholipid bilayer, can reduce agglomeration and maintain colloidal stability [16,98,99]. The agglomeration propensity of certain functionalised MSNs, such as low molecular weight PEG-coated MSNs, can exacerbate vascular injury by promoting larger areas of thrombosis, highlighting the need for a case-by-case evaluation based on the PEG molecular weight [100]. Similarly, amine-functionalised iron oxide NPs have been shown to cause cytotoxicity to HepG2 cells at higher concentrations, primarily due to agglomeration. Lower agglomeration reduced cytotoxicity, whereas higher levels increased it [83].
Another study observed a time-dependent effect on amorphous silica aggregates in human bronchial epithelial (HBE) cell cultures. When HBE cell cultures were exposed repeatedly to different sized (~2.5 µm vs. 100 nm) silica (aggregates) for two weeks and then allowed to recover (without amorphous silica exposure), silica aggregates of larger sizes (size ~2.5 µm) significantly affected the cell proliferation, and the release of IL-6, IL-8, and total glutathione at the end of both exposure cycles. In contrast, their nano-sized counterparts (<100 nm) induced more pronounced effects only at the end of the first exposure cycle. This study suggests that aggregates formed from larger silica NPs are toxicologically relevant and should be considered in risk assessment [101]. However, in another study, genotoxicity investigations using the comet and micronucleus assays on agglomerated mesoporous vs. non-agglomerated non-porous Swiss mice showed no evidence of genotoxicity in either group, indicating that agglomeration did not affect downstream toxicity [102].
On the other hand, under certain conditions, agglomeration can be advantageous. For instance, PSNs exhibit low bulk density and small particle size, challenging controlled flow toward target sites. Granulation of nanocarriers offers a partial solution by increasing bulk density, improving flow and enhancing delivery to the target site. Although this process decreases their pore volume and surface area, this reduction is negligible compared to the total specific surface area [16]. Future advances should focus on optimizing granulation parameters to maximize pore volume and therapeutic loading. Although this approach may compromise the surface area, it could offer benefits in terms of safety and minimizing toxicity, as the latter, in the case of PSNs, is strongly linked to the surface area of the pores that contain specific reactive silicic acid residues that generate toxicity by producing ROS, which can interact with the biological milieu.

2.2.7. Crystallinity State

The toxicity and biological interactions of NPs critically depend on their physical state-crystalline or amorphous. Crystalline NPs have a highly ordered structure, which results in higher surface reactivity and, consequently, induces greater oxidative stress, DNA damage, lipid peroxidation, an inflammatory response, and cytotoxicity [103,104]. In comparison, amorphous NPs lack an ordered structure and are generally less toxic due to their increased solubility and decreased surface reactivity [105,106]. These differences impact fundamental biological processes, including ROS production, lysosomal stability, and immune recognition, affecting NP-induced cytotoxicity [107,108]. However, multiple studies have shown that they cause acute effects such as cytotoxicity via ROS generation. The toxicity of these NPs can be affected by the composition and synthesis method [109,110,111].
These observations indicate that crystallinity is a critical factor in determining the toxicity and biocompatibility of PSNs. Therefore, accurately identifying the physical state of NPs is crucial for ensuring their safety and efficacy.
In summary, standardized characterization of PSNs is needed to ensure reproducibility of the advanced material for scale-up manufacturing.

2.3. Assessment of Biocompatibility

To ensure the biocompatibility of PSNs, the biological interactions of these advanced materials within the body need to be studied extensively. Therefore, comprehensive functional toxicological assessments should include evaluations of acute and chronic toxicity, oxidative stress, inflammation, and organ-specific accumulation alongside analysis of protein corona formation, immunogenicity, and gene expression signalling pathways [112,113,114]. Biodistribution is also another essential aspect, as PSNs can enter the body through different routes of exposure. Incorporating these endpoints provides a better understanding of PSNs’ safety and functionality, supporting the development of safer biocompatible advanced nanomaterials.

2.3.1. Cytotoxicity of PSNs

Cytotoxicity is an important factor in assessing NP safety and potential biomedical applications. Size [75], surface charge [115], shape [116], functionalisation [117], crystallinity [118], and exposure time [119], all play a role in determining the extent of cytotoxicity [120,121]. Cytotoxic effects can include membrane disruption [122], oxidative stress [123], mitochondrial dysfunction [124], apoptosis [125], necrosis [126], and inflammatory responses [127]. Studies have shown that different NPs can induce varied cytotoxic responses, underscoring the importance of comprehensive in vitro and in vivo evaluations.
The cytotoxicity of NPs can depend on the cell line used in toxicity assays, as different cell types may exhibit varying metabolic rates, uptake mechanisms, and antioxidant defences. According to a study comparing the effects of ZnO and SiO2 NPs, THP-1 cells were more sensitive to these NPs than L-132 cells [128], which correlates with the results published by Lanone et al. [129] who found variable sensitivity of human alveolar (A549) cells and macrophage (THP-1) cell lines to 24 different NPs. Another study found that 10 μg/mL of AgNPs exhibited varying levels of toxicity in cell lines. Ovarian cancer cells (A2780) were slightly more sensitive than breast cancer cells (MDA-MB 231), and both were more sensitive than MCF-7 cells [130].
The cytotoxicity of NPs can also be affected by a combination of physicochemical properties, cell-type specificity and exposure duration. The literature indicates that smaller, positively charged, and elongated NPs exhibit increased toxicity [75], whereas PEGylation and optimized surface functionalisation may reduce cytotoxicity [37,48]. Studies have demonstrated that the surface functionalisation of NPs significantly influences their cytotoxicity profiles. In some cases, non-functionalised NPs exhibit minimal or no cytotoxic effects, whereas functionalisation can introduce cytotoxicity depending on the nature of the surface modification. For instance, a study on mesoporous silica MCM-41 loaded with the anticancer drug, bicalutamide, showed that the empty MCM-41 nanocarrier did not induce toxicity. Cytotoxicity only increased once loaded with the drug. This indicates that without functionalisation, MSNs can be biocompatible and safe [131]. However, other studies have shown that MSNs without functionalisation are still cytotoxic. For instance, an investigation of MCM-41 and SBA-15 found that they were cytotoxic at most concentrations between 0.2 and 14 mg/mL to Caco-2 cells, causing membrane damage, ROS generation and increased apoptotic signalling [132]. Functionalisation of MCM-41 with essential oil components, eugenol and vanillin, showed greater cytotoxicity to HepG2 cells than bare MCM-41 after 48 h of exposure. It was suggested that due to the cationic nature and increased cell membrane interactions, cytotoxicity increased [133].
Cytotoxicity induced by PSNs must be well understood, both in bare and functionalised states, to prevent collateral toxicity and to ensure their safe and effective application in biomedicine.
Mechanism of Cytotoxicity-Oxidative Stress
Oxidative stress is a key mechanism underlying NP-induced cytotoxicity, driven by ROS generation and by cellular antioxidant defence systems [33]. ROS consist of a diverse range of highly reactive species, including hydroxyl radicals (•OH), superoxide anions (O2), and hydrogen peroxide (H2O2). Hydroxyl radicals are the most reactive and harmful and are highly capable of damaging cells. Superoxide anions and hydrogen peroxide are also less reactive than hydroxyl or carbon-centred alkyl radicals (R•) but can still contribute to oxidative stress and serve as precursors of stronger radicals [134,135].
NPs can enter cells and produce excessive ROS, leading to an imbalance of pro-oxidants to antioxidants [136]. This is achieved either by direct interaction with cellular components or by activating the mitochondrial pathway, which can result in oxidative damage to macromolecules such as lipids, proteins, and DNA [137]. One of the major consequences of ROS accumulation is lipid peroxidation, which can compromise cell membrane integrity, leading to increased permeability, loss of function, and cell death. Protein oxidation also inhibits enzymatic function, disrupting signalling cascades and leading to protein aggregation that may be cytotoxic [137,138,139].
Oxidative stress also damages DNA, forming DNA adducts, strand breaks, and mutations [140,141]. While this damage may activate several repair mechanisms, unresolved DNA lesions can trigger apoptotic pathways. The tumour suppressor protein p53 and the c-Jun N-terminal kinase (JNK) pathway are essential mediators of oxidative stress-induced apoptosis [142,143]. The activation of these pathways helps ensure that cells that are too damaged to function properly undergo programmed cell death rather than proliferate, thereby preventing potential mutations [144,145].
Enzymes such as glutathione peroxidase (GPx), catalase, and superoxide dismutase (SOD) are part of the cellular antioxidant defence system and contribute to the reduction of ROS-mediated damage [146,147,148]. Glutathione is a cellular antioxidant that quenches ROS and is also involved in detoxifying harmful metabolites [149,150]. Catalase breaks down hydrogen peroxide into water and oxygen, and superoxide dismutase catalyzes the dismutation of superoxide anions to produce oxygen and hydrogen peroxide [143]. However, when ROS production level exceeds the cell’s capacity to neutralize them with antioxidants, oxidative stress occurs, leading to cytotoxicity [151,152].
Several studies have shown that NPs, including PSNs, can induce ROS generation [153,154,155]. For example, a study demonstrated that SiNPs induced oxidative stress in HepG2 cells, causing apoptosis. They reported that smaller NPs (7–20 nm) induced greater oxidative stress than larger particles (50 nm), likely because of their higher surface area-to-volume ratio, which facilitates greater interactions with cells [37]. Another study showed similar results with MRC-5 fibroblast cells exposed to 62.5 μg/mL concentration of SiO2NPs. Increased levels of ROS were observed at 24, 48 and 72 h, confirming SiO2NPs induced ROS as a mechanism of cell survival [156]. Other NPs, such as iron oxide NPs (IONPs), have also been shown to generate ROS in vitro. For instance, IONPs triggered the regulated cell death process known as ferroptosis, characterized by lipid peroxidation and iron toxicity, in NRK-52E rat renal tubular epithelial cells. IONPs can promote oxidative injury and contribute to cell death via ferroptosis pathways [157]. Another study investigated the effect of IONPs on human microvascular endothelial cells. The study found that endothelial cell permeability increased following exposure to ROS-mediated oxidative stress induced by IONPs. The increase in permeability may affect vascular integrity and function [158].
Nevertheless, it should be noted that oxidative stress is not always the main mechanism of cytotoxicity induced by NPs. A recent study found that positively charged, PEI-modified MSNs, induced the accumulation of autophagosomes in HUVECs by inhibiting autophagosome maturation and hindering their fusion with lysosomes. This caused cytotoxicity in the absence of apparent ROS involvement [159]. Another study showed that in transformed human alveolar epithelial type 1-like cells, amine-modified polystyrene NPs induced severe membrane damage and altered cell adhesion, causing cell detachment and apoptotic cell death without ROS generation [160]. These studies further indicate that NP surface chemistry and charge distribution are key determinants of cytotoxicity, independent of oxidative stress pathways.
In general, oxidative stress is one of the main pathways by which NPs exert cytotoxicity. The relationship between ROS production, antioxidant defences, and resulting cellular damage emphasizes the need to assess oxidative stress to determine the safety of NPs for biological use.
To comprehensively evaluate these effects, various assays assess NP-induced cytotoxicity by measuring cellular metabolic activity, membrane integrity, and apoptosis. These include the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, lactate dehydrogenase (LDH) assay, trypan blue assay, caspase activity assay, annexin V/propidium iodide staining, alamar blue assay, cytokinesis block proliferation index and relative population doubling (Table 2). For more details, refer to Supplementary Materials. Combining oxidative stress assays with various cytotoxicity endpoints provides a broader view of cellular responses induced by NPs, which is crucial for their safety in biomedicine. Overall, these cytotoxicity assays provide valuable insights into the cellular responses induced by NPs; however, their reliability can be influenced by NP-assay interference [75]. Therefore, it is essential to evaluate potential interactions between NPs and assay components prior to use, to ensure accurate and reproducible results.

2.3.2. Genotoxicity of PSNs

Genotoxicity is the ability of physical or chemical substances or materials to damage the genetic material within cells [161,162,163,164]. Genotoxicity testing is significant for assessing the safety of NPs, as it can determine whether materials, such as PSNs, can directly interact with DNA or disrupt cellular mechanisms that harm genetic material, leading to chromosome alterations or other modifications if DNA repair is impaired. It is a critical step in safety assessment for NPs or other xenobiotics that may not exhibit cytotoxic effects but can affect cellular health by inducing DNA damage, which may go unnoticed if genotoxicity is not assessed [165]. Similar to cytotoxicity, the genotoxic potential of NPs is influenced by several physicochemical properties such as size, shape, surface charge, crystallinity, and functionalisation [166]. Understanding these mechanisms is critical for evaluating NP safety and mitigating potential risks in biomedical applications.
Physicochemical Dependent Properties Affecting Genotoxicity
Physicochemical characteristics of NPs play an essential role in their genotoxic potential. Size-dependent effects have been widely observed, with smaller NPs showing higher nuclear translocation and stronger interactions with DNA [167]. In a study on AuNPs of different sizes (5, 20, and 50 nm), 5nm AuNPs caused dose-dependent DNA damage in HepG2 cells after 24 h of treatment; however, 20 and 50 nm AuNPs did not induce evident DNA damage. The 5 nm AuNPs also caused cell cycle arrest and increased ROS production [168]. Another study compared different sizes of amorphous SiNPs (11 nm, 34 nm, 248 nm) in 3T3-L1 mouse fibroblasts. Chromosomal aberration induction occurred only with 34 nm particles, illustrating the importance of optimum particle size and genotoxicity [169,170].
The shape of NPs is another important factor that dictates genotoxicity assessment. For instance, elongated NPs, such as nanorods and nanowires, are reported to mechanically stress chromosomes, causing mitotic errors [171]. Sphere-, rod- or wire-shaped TiO2NPs have been shown to induce DNA damage in the Caco-2/HT-29 cell model with the comet assay. The greatest genotoxicity was observed with rod-shaped NPs, confirming their interference with mitotic processes [172]. The surface charge of NPs plays a key role in their genotoxicity by influencing their interactions with DNA [173]. Positively charged NPs interact more strongly with the negatively charged DNA backbone, leading to DNA condensation and interference with transcription [174]. A study demonstrated that amine-functionalised SiNPs showed a stronger binding affinity to DNA, leading to greater genotoxicity than carboxyl-functionalised NPs, which had weaker electrostatic interactions with genetic material [75,175]. Similarly, positively charged PLGA NPs induced genotoxicity in 16HBE14o human bronchial epithelial cells as assessed by the micronucleus and comet assays; however, negatively charged or neutral NPs showed no genotoxicity. Results showed chromosomal aberrations without primary DNA damage [176]. This correlation emphasizes the importance of surface charge in NP-induced genotoxicity.
Crystallinity of NPs also significantly influences genotoxicity. Crystalline NPs can generate higher ROS than amorphous ones; excess ROS production can induce oxidative stress, leading to oxidative DNA injury and micronuclei formation [177]. A study showed that anatase TiO2 NPs typically induce DNA strand breaks and chromosomal damage, whereas rutile TiO2 NPs are predominantly nongenotoxic in vitro [178]. On the other hand, a different study demonstrated that TiO2-A NPs, mainly anatase, were cytotoxic and had little oxidative effect on both cell lines, whereas TiO2-B NPs, mainly rutile, induced genotoxic and oxidative effects in both cell lines, contradicting the previous study [179]. Research has also demonstrated that amorphous NPs can be genotoxic, like crystalline NPs. For instance, synthetic amorphous silica showed a dose-dependent genotoxic (0.01 to 150 µg/mL) effect in the mouse lymphoma assay, which detects genetic mutations [180]. Crystalline and amorphous niobium oxide NPs (NINPs) also showed genotoxicity in CHO-K1 cells. The crystalline NINPs exhibited high genotoxicity after 4 h of exposure with increased micronuclei and chromosomal instability, whilst amorphous NINPs demonstrated lower genotoxicity with less micronuclei formation after 24 h of exposure [181]. This highlights the significance of NP crystallinity in determining their genotoxic potential.
Mechanisms of Genotoxicity
NPs induce genotoxic effects through multiple mechanisms. One major mechanism is through oxidative stress. Oxidative DNA alterations, such as breaks, base oxidation, and chromosomal instability, are caused by excessive ROS [182]. Several NPs have induced oxidative stress, leading to genotoxicity. For example, AgNPs have been shown to induce DNA damage by generating ROS. They induced DNA damage via an oxidative stress mechanism after 30 min of exposure in human embryonic epithelial cells using a modified comet assay [183]. Another study showed the genotoxicity of titanium dioxide nanoparticles (TiO2NPs) in human epidermal cells, identifying their potential to induce DNA damage, possibly by oxidative stress. The results indicated that TiO2NP exposure led to ROS upregulation, as evidenced by oxidatively modified DNA, SSBs (single-strand breaks), and DSBs (double-strand breaks). Consequently, the upregulation of oxidative stress-associated biomarkers was also observed, underscoring the role of ROS generation as a key factor in TiO2NP-mediated genotoxicity [184]. Tin-loaded MSNs also induced significant ROS generation in MCF-7 cells at a concentration of 5 μg/mL after 6 h. These MSNs also caused genotoxic effects in CHO cells (2–8 μg/mL, 24 h), including chromosome aberrations, micronucleus formation, and a reduction in mitotic index [185]. This emphasizes the role of oxidative stress as the major driver of NP-mediated DNA damage, which, depending on dose and duration of exposure, could potentially lead to mutagenesis and carcinogenesis.
In addition to oxidative stress, direct DNA interaction also contributes to NP-induced genotoxicity. It has been reported that some NPs can enter the nucleus and interact with genetic material, resulting in DNA strand breaks, chromatin condensation, and transcriptional interference [164,165,184]. For example, IONPs were shown to form complexes with DNA that could also intercalate between base pairs. This direct interaction caused chromosomal aberrations, micronucleus formation, and DNA damage in vitro and in vivo [186]. Also, SiO2 NPs (10–500 μg/mL) caused a concentration-dependent increase in DNA strand breaks, reaching a 13-fold increase at 300 μg/mL after 24 h of exposure, without inducing ROS generation in human peripheral blood mononuclear cells [187]. Similarly, another study demonstrated that SiNPs induced H2AX phosphorylation in spermatocyte cells of mice, leading to DNA double-strand breaks in a dose-dependent manner across concentrations of 6.25 μg/mL to 50 μg/mL [188].
Another mechanism of genotoxicity involves interference with mitosis. In this mechanism, microtubules that move chromosomes during cell division (spindle microtubules) are disrupted by NPs, leading to chromosome missegregation and aneuploidy [189,190,191]. For example, it has been shown that microtubule networks can be altered by highly water-dispersible single-walled carbon nanotube (SWCNT) nanosystems, and their effects on mitotic spindles have been studied, leading to the conclusion that these NPs may induce chromosomal instability. In human airway epithelial cells exposed to SWCNT, the authors noted duplicated and fragmented centrosomes, disrupted mitotic spindles, and abnormal aneuploid chromosome numbers [192]. Other engineered nanomaterials, such as copper-based metal–organic frameworks, have demonstrated the ability to promote tubulin polymerization and disrupt microtubule dynamics. This can interfere with the formation and function of the mitotic spindle, resulting in an abnormal mitotic spindle [193,194].
There have also been extensive studies on inflammation-driven genotoxicity, driven by NPs’ ability to elicit inflammatory responses, leading to subsequent oxidative stress and DNA damage [195]. Senapati et al. [196] showed that ZnONPs could induce the upregulation of pro-inflammatory cytokines such as interleukin 1β (IL-1β) and TNF-α in THP-1 macrophages, which subsequently may contribute to oxidative stress-generated genotoxicity. It has been shown that inflammatory cytokines can cause DNA damage by generating reactive nitrogen species, such as nitric oxide and peroxynitrite. These reactive species can induce oxidative damage, leading to genomic instability. Consequently, chronic inflammation is a significant driver of genomic instability [197]. Oesophageal epithelial cells are another example of how chronic inflammation can lead to malignant transformation by inducing genomic instability. Studies suggest prolonged exposure to inflammation can disrupt genomic integrity, increasing the risk of malignant transformation over time [198]. Such a mechanism is highly relevant to the development of NPs for long-term biomedical applications, given the potential for repeated exposure to lead to irreversible inflammatory responses and cumulative DNA damage.
Various genotoxicity assays (Table 3) can be used to assess different types of DNA damage including the cytokinesis block micronucleus (CBMN) assay for detecting chromosomal breakage or loss, the comet assay for measuring DNA strand breaks, the chromosome aberration test for identifying structural chromosomal alterations, γ-H2AX assay for detecting DNA double-strand breaks and the Ames Test for evaluating mutagenic potential in bacteria. For more details, refer to Supplementary Materials.

2.3.3. Gene Expression Profiles

Gene expression profiling is a useful tool for examining the molecular mechanisms underlying PSN-induced toxicity. Exposure to different NPs alters intracellular signalling cascades and gene expression patterns, highlighting the pathways involved in oxidative stress, inflammatory response, apoptosis and DNA damage [199].
The p53 pathway plays a central role in the cellular stress response to NPs by regulating gene expression responsible for DNA repair, apoptosis, and cell cycle arrest [200,201]. Upstream regulators of p53 include DNA damage sensors such as ATM (ataxia-telangiectasia mutated) and ATR (ataxia-telangiectasia and Rad3-related) kinases, which phosphorylate and stabilize p53 in response to DNA damage [202]. Other upstream regulators include murine double minute 2 (MDM2), an E3 ubiquitin ligase that normally targets p53 for proteasomal degradation, which is inhibited under stress, allowing p53 to accumulate [203]. Once p53 is activated, it acts as a transcription factor, regulating a wide range of downstream target genes that influence cellular fate [204].
Following DNA damage, p53 up-regulates the expression of p21 (CDKN1A), a cyclin-dependent kinase (CDK) inhibitor, which causes cell cycle arrest at the G1/S checkpoint to allow time for DNA repair [205,206]. If the damage is beyond repair, p53 activates apoptosis by increasing the expression of pro-apoptotic genes such as BCL-2-associated X protein (BAX), p53 upregulated modulator of apoptosis (PUMA), and phorbal-12-myristate-13-acetate-induced protein 1 (NOXA), causing mitochondrial outer membrane permeabilisation (MOMP) and caspase activation, ultimately resulting in programmed cell death [207,208]. Furthermore, p53 regulates senescence, a permanent state of cell cycle arrest, by modulating the p21/p16INK4a and pRB pathways, thereby inhibiting aberrant cell proliferation [209,210].
Nanoparticles have been shown to activate the p53 pathway through mechanisms of oxidative stress and DNA damage, leading to varied cellular responses depending on their physicochemical properties [211]. Wierzbicki et al. [212] investigated the effects of graphene oxide nanoplatelets and graphite NPs on glioma cell lines with varying p53 status. They demonstrated that NPs diminished the angiogenic potential of glioma cells, with p53 status-dependent effects. In wild-type p53 cells, NPs inhibited NF-kB activity, leading to lower levels of inflammatory cytokines and oxidative stress. Contrarily, in mutant p53 cells, NF-kB signalling remained active, leading to persistent inflammation and cellular stress, highlighting the role of p53 in mitigating NP-triggered toxicity. Further evidence of p53 pathway activation was observed in primary pulmonary epithelial cells; prolonged exposure to TiO2NPs was accompanied by elevated expression of key apoptotic proteins, including BAD, BAX, and phospho-p53 [213]. Similar results were observed in MCF-7 cells, where MSNs coated with polydopamine and loaded with umbelliprenin activated the TP53 pathway. Treatment with 30–45 μg/mL for 48 h resulted in approximately a 3-fold increase in p53 gene expression, with the activation associated with apoptosis [214].
The p53 pathway interacts with various signalling pathways that mediate cellular homeostasis and stress responses. Figure 3 illustrates this complex network, highlighting crosstalk between the p53 pathway and other signalling pathways. The NF-kB pathway is a key pathway that interacts with p53; it is involved in inflammation and immune responses [215,216]. While p53 mainly induces apoptosis and cell cycle arrest following DNA damage, the activation of NF-kB can oppose this signalling by promoting cell survival and inflammatory pathways [217,218]. This relationship is intricate because NF-kB can inhibit p53 via MDM2 upregulation, leading to p53 degradation, but p53 can also inhibit NF-kB activity by competing with transcriptional coactivators like p300/CBP [219,220]. Previous research has demonstrated that NPs can alter p53 and NF-kB expression. For instance, silica NPs ranging from 50 to 200 μg/mL concentration caused ROS, mitochondrial depolarization and apoptosis after 24 h of exposure in HUVEC cells. It was demonstrated that silica NPs induced cell damage by oxidative stress via p53, NF-kB, and JNK signalling pathways [221]. This suggests that silica NPs, including PSNs, can induce cellular stress responses through oxidative stress-mediated signalling pathways, leading to mitochondrial dysfunction and apoptosis.
Another important pathway that modulates p53 activity is the MAPK pathway, which comprises three components: extracellular-signal-regulating kinase (ERK1/2), stress-activated protein kinase/c-Jun-N-terminal kinase (JNK) and p38 [222]. p38 MAPK activation upon cellular stress promotes p53 phosphorylation at specific serine and threonine residues, thereby increasing its stability and transcriptional activity. This is particularly relevant in responses to oxidative stress and in DNA damage repair [223,224]. In specific cellular contexts, ERK may stabilise or promote the degradation of p53 [225,226]. MSNs loaded with resveratrol (plant polyphenol) can modulate ROS levels and activate the p38-MAPK/p53 signalling pathway, leading to increased apoptosis and autophagy in hypertrophic scar fibroblasts [227]. Also, another study demonstrated that silica NPs activated p53 by the JNK pathway activation, resulting in p53 acetylation and cytoplasmic localization, leading to mitochondrial cytochrome c release and apoptosis [228]. These results highlight the significance of the MAPK pathway as a critical regulator of cellular stress responses through its interaction with p53.
The p53 tumour suppressor protein also interacts with the autophagy pathway and has a dual role depending on cellular stress levels [229,230]. In response to mild stress, p53 can promote the transcription of autophagy-related genes, such as Atg, DRAM, and Sestrin1/2, thereby facilitating the clearance of damaged cellular organelles [231,232]. However, mutant p53 states can promote increased ROS production during autophagy, indicating that NF-kB acts as a negative regulator of autophagy and further highlighting the crosstalk between the two pathways [233,234].
In addition, the phosphoinositide 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway is closely related to p53 in determining cell survival or death [235]. Dysregulation of the PI3K/AKT pathway often occurs following p53 inactivation. On the other hand, p53 downregulates mTOR signalling by upregulating the AMPK pathway, thereby restricting cell proliferation under stress [236,237]. Recent research has demonstrated that MSNs can modulate the P13K/AKT/mTOR signalling cascade, promoting cell proliferation and tissue repair but also preventing apoptosis [238,239].
NPs can also modulate NF-kB downregulation in p53 wild-type glioma cells, thereby reducing inflammatory cytokine expression [212]. In contrast, p53 and NF-kB can cooperate to regulate pro-inflammatory responses in human macrophages, thereby modulating the expression of interleukin-6 (IL-6), interleukin-8 (IL-8), and growth-regulated oncogene alpha (GRO-α). The cytokines are known to induce neutrophil migration and immune activation, suggesting that immune cell-specific responses can be induced by NF-kB modulation via NPs [217].
The surface chemistry of NPs has been found to play a role in NF-kB activation. Research showed that the activation of the NOD-like receptor protein 3 (NLRP3) inflammasome and the production of IL-1β were influenced by the surface reactivity of carbon nanotubes. Polyetherimide induced heavy cationic functionalisation and increased secretion of the inflammatory mediators IL-1β, TGF-1, and PDGF-AA in BEAS-2B and THP-1 cell lines [240]. This suggests that NPs with strong cationic functionalisation exhibit greater immunogenicity and inflammation due to strong interactions with cells, activating the NLRP3 inflammasome and leading to the subsequent secretion of cytokines. Therefore, understanding how different NP properties influence gene expression changes is essential for safer and more effective nanomedicine applications.
These interactions highlight that p53 does not act alone but is embedded in a larger signalling network that regulates cellular responses to stress, damage, and metabolic fluctuations. Insights into these coupled networks will provide an important basis for the engineering of therapeutic interventions targeting p53 and its related pathways in disease.

2.3.4. Protein Corona

The protein corona is a dynamic layer of proteins that surrounds NPs upon exposure to biological fluids. Protein corona formation has significant implications in nanomedicine, as it modifies NPs’ biological identity and often affects (facilitates or restrains) cell targeting, impacting cellular uptake and in vivo biodistribution [241,242,243]. The protein corona comprises two distinct layers: the hard corona, representing a tightly bound inner layer of proteins with a high affinity for the NP surface, and the soft corona, composed of loosely associated proteins that dynamically exchange with the surrounding biological environment (Figure 4) [244,245,246,247]. For PSNs, this is especially relevant, as the corona would strongly affect their stability, cellular interactions, toxicity, and targeting capabilities. Incubation of SiNPs with human plasma identified over 300 different proteins in the corona [248,249]. The findings indicated that hard corona proteins remain bound to NPs for several hours, while soft corona proteins exchange within minutes, highlighting the dynamic nature of the protein corona in biological fluids.
Transferrin ligands on silica NPs can be masked by plasma proteins, hindering receptor-mediated endocytosis and resulting in non-specific uptake [250,251,252], thereby compromising targeting specificity. Conversely, PEGylated polymeric NPs that adsorbed apolipoproteins reduced rapid clearance by immune cells and prolonged blood circulation times, suggesting that the appropriate corona composition may improve NP stability in vivo [253]. However, protein corona formation can also activate the immune system, triggering complement activation and opsonisation, which leads to faster clearance from circulation [254,255,256].
Albumin is the most abundant protein found in NP coronas, including silica [257,258,259]. While association with some proteins, such as albumin, increases the circulation time of NPs by preventing aggregation and reducing macrophage recognition [259,260,261] while others, including complement proteins 3 (C3) and 4 (C4), are associated with increased immune clearance [262,263].
Among others, immunoglobulins (IgG, IgA), fibrinogen, transferrin, and apolipoproteins (ApoA1, ApoE) are commonly identified [257,264]. Opsonisation by immunoglobulin adsorption has been shown to promote enhanced phagocytosis, while apolipoproteins modulate NP transport across biological barriers, including the BBB [265,266,267]. Specifically, MSNs in biological fluids can adsorb plasma proteins, which can either reduce their opsonisation and recognition by phagocytic cells or promote phagocytosis, affecting their effectiveness as drug delivery systems [268,269]. Therefore, there is a need for surface engineering strategies to control immune recognition and improve therapeutic efficacy by coating MSN surfaces with polymers, such as PEG, that can diminish protein adsorption and opsonisation, and extend the circulation half-life of MSNs, resulting in higher drug delivery efficiency [87,270].
Likewise, due to their surface charge, positively charged lipid-based NPs (cationic liposomes) have been shown to bind efficiently to plasma proteins during drug delivery [271,272], thereby labelling the NPs for opsonisation and elimination by phagocytic cells. These interactions illustrate how NP surface properties dictate biological response and emphasize the need for a design that takes therapeutic efficacy and immune system interactions into consideration [273,274,275].
Understanding this protein-NP interaction is important for maximizing the efficacy of these NPs in drug delivery and imaging applications. Multiple analytical techniques are employed to characterize the protein corona, including protein composition, binding affinities and structural properties of the associated proteins including liquid chromatography tandem mass spectrometry (LC-MS/MS) [276,277], sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE) followed by Western blotting [257,278] and dynamic light scattering (DLS) that registers the alterations of NP dimensions triggered by the crown buildup which serve as indirect proof of protein adhesion. It is typically used in conjunction with zeta potential determination to gain insight into how surface charge is altered by corona formation [279,280].
Despite differences in protein corona composition, a general trend shows that positively charged NPs, including silicon dioxide (SiO2) NPs attract more proteins than negatively charged NPs [281], indicating that surface charge influences protein adsorption [282]. The influence on protein corona is not only charge-dependent but also functionalisation-dependent. PEGylation has been shown to decrease protein adsorption, leading to less bound protein on the surface [270], which illustrates the compromise between stability and cellular recognition in NP drug delivery [283]. Understanding protein corona formation and composition is essential for optimizing the performance of PSNs in biomedical applications.

2.3.5. Immunogenicity of PSNs

Several physicochemical properties, such as size, surface charge, shape, and surface functionalisation, determine the ability to induce an immune response, or immunogenicity, a factor that can significantly affect the overall efficacy and safety of NPs [284]. One of the most pronounced immune mechanisms influencing the fate of PSNs is mediated by the complement system, which may contribute to their clearance, immune recognition, and subsequent inflammatory responses. Thus, understanding the mechanisms of complement activation is essential to enable their suitable tuneability for medical applications [285,286].
The complement system consists of more than 30 plasma proteins that recognize pathogens, promote inflammation, and stimulate phagocytosis; it is a critical component of the innate immune response. It is activated by three pathways (Figure 5): the classical pathway, the lectin pathway, and the alternative pathway [287]. The three pathways converge at the cleavage of C3 to generate C3b, a key opsonin that enhances immune cell recognition, and C5b, which initiates the assembly of the membrane attack complex (MAC) [288]. Thus, the activated pathways may play an important role in the processing of PSNs within the body, influencing circulation time, clearance, and toxicity [289,290]. A study demonstrated that dextran-coated NPs activated complement primarily via the alternative pathway in human serum [290]. Another study also showed that complement activation led to the deposition of C3b on NP surfaces. This opsonisation facilitates recognition, movement, and clearance by immune cells, specifically macrophages and dendritic cells [291]. This has implications for PSN biodistribution, as complement activation can result in rapid systemic clearance, reducing their use as drug delivery carriers or for imaging applications [292,293].
The role of complement proteins C1–C9 in NP recognition varies depending on the physicochemical properties of NPs. C1q initiates the classical pathway by binding to immunoglobulin-coated NPs or other specific ligands on their surface [286]. Studies found that IgG-enriched protein coronas around NPs showed strong C1q binding and enhanced classical pathway activation [95,258]. Alternatively, the lectin pathway is initiated by mannose-binding lectin (MBL), ficolin, and MBL-associated serine proteases (MASPs), which recognize carbohydrate patterns on NP surfaces. It has been shown that some gold and silica NPs functionalised with sugar moieties can activate this pathway [294,295,296]. Most NPs activate complement through the alternative pathway, driven by spontaneous C3 hydrolysis and the deposition of C3b onto the NP surface. Research showed that, compared with low- or non-charged particles, cationic NPs were associated with stronger complement activation [293]. Similarly, another study showed that gold NPs passivated with PEI, a positively charged polymer, showed enhanced complement activation compared to other polyelectrolyte ligands [297].
After the complement cascade is activated, important downstream effects include phagocytosis and the formation of MAC. C5 convertase cleaves C5 into C5a and C5b, with C5a playing an important role as an inflammatory mediator, recruiting neutrophils and monocytes to areas of immune activation. C5b initiates MAC (C5b–C9) formation and creates pores in microbial and cellular membranes, leading to induced damage and inflammatory toxicity when overactivated by NPs. Excessive MAC formation induced by NPs triggered hypersensitivity reactions, inflammation, and complement-mediated toxicity, especially during intravenous NP administration [298,299,300].
Complement activation affects NP function in two distinct ways. On the one hand, it clears the NPs, preventing the accumulation of foreign materials in circulation. This is beneficial for vaccine adjuvants and antimicrobial NPs, in which complement activation can promote pathogen clearance and activate the immune response [286]. On the other hand, excessive complement activation is detrimental to NP drug delivery systems as it reduces their circulation time [301]. For example, the opsonisation of transferrin-functionalised SiNPs reduced targeting efficiency, as complement proteins masked transferrin ligands, thereby preventing their uptake by cancer cells [251].
Surface functionalisation is one of the most used strategies to modulate complement activation while maintaining NP function. One approach is PEGylation; NP PEGylation can reduce complement activation by reducing protein adsorption. This steric hindrance reduces interactions with other molecules and aggregation by creating a conformational cloud around PEG chains. This hydrophilic barrier effectively masks the NP surface from recognition by positive complement proteins [301]. Nonetheless, even PEGylated NPs may accumulate C3b molecules, leading to immune recognition after extended circulation time [87,302]. It is suggested that zwitterionic coatings mimic cell membranes and thus lead to lower complement activation than PEGylation [303]. Another strategy focuses on biomimetic modifications, in which CD47-functionalised NPs present a native “self” signal to antagonize complement-mediated phagocytosis [304,305]. In addition to surface engineering, pre-coating NP surfaces with either albumin or apolipoproteins has become a strategy to protect against excessive complement activation while maintaining functional transport properties [306,307]. It was reported that NPs precoated with albumin reduced C3b deposition, thereby prolonging circulation and improving biodistribution [308].
NPs also interact with antigen-presenting cells (APCs) and T and B cells to modulate adaptive immune responses. The potential of some NPs to act as haptens can stimulate T-cell-dependent immune responses by facilitating antigen uptake and dendritic cell presentation [309,310]. This, however, may result in antibody production against NPs, potentially affecting their long-term stability and biodistribution [293]. Moreover, NP-mediated inflammasome activation has been associated with various chronic inflammatory conditions. Evidence for NLRP3 inflammasome activation leading to IL-1β secretion and inflammatory toxicity from metal oxide NPs has been demonstrated in HepG2 and THP-1 macrophage models [241,311].
Overall, understanding the immunogenicity of NPs, particularly their capacity to induce complement activation, is complex and depends on multiple physicochemical features [284]. Complement activation could potentially promote immune clearance and stimulate the immune response in vaccine applications, but excessive activation can cause inflammation, hypersensitivity reactions, and rapid clearance, which is detrimental in NP drug delivery applications [312]. Understanding and modulating these immune interactions with novel surface modifications, biomimetic coatings, and complement inhibitors should be further developed to address immune evasion and immune compatibility of NPs, thereby maintaining NP materials’ functionality and biocompatibility in future clinical applications [293,313,314].

2.4. Facilitation of Toxicity: Exposure Routes and Relevant Models

The biological effects of NPs depend mainly on their exposure route, which determines their biodistribution, metabolism and elimination [315,316]. Hence, NPs can be administered through respiratory (inhalation) [317,318], dermal (skin) [319], oral (mouth and gut) [320,321,322,323], systemic (injection) [114,324] and mucosal (membrane entry and into the bloodstream) [325,326] exposure, each presenting different interactions with biological environments (Figure 6). Due to the complexity of NP interactions across distinct biological environments, it is essential to use diverse cell lines that best represent these exposure routes [130,327], thereby narrowing the gap between in vitro and predicted in vivo effects.

2.4.1. Inhalation

Inhalation is a key exposure pathway, especially for engineered nanomaterials, which are often manufactured and processed in occupational environments [328,329]. The particles are small enough to invade the lungs as far as the alveolar space, translocate into the blood and target secondary organs such as the liver, kidneys and brain, raising concerns about their systemic toxicity. Oxidative stress, inflammation, and pulmonary toxicity may be induced by inhaled NPs [330]. Human monocyte-derived THP-1 cells and 16HBE14o-(16HBE) human bronchial epithelial cells have been used in an in vitro model of airway barrier properties to test the impact of NPs on airway epithelial cells [331]. Additionally, Calu-3 human bronchial epithelial cells have been employed to mimic bronchial epithelial barrier function, mucus production, and epithelial repair, providing insights into how NPs may affect respiratory health [332,333].

2.4.2. Dermal

Dermal exposure to NPs can occur through multiple pathways, such as cosmetics, transdermal drug delivery systems, and wound dressings [319,334,335]. While the stratum corneum constitutes a natural barrier, prolonged exposure or disruption in skin integrity can allow NPs to penetrate more deeply into the skin and potentially enter systemic circulation [336]. Factors like skin diseases, wounds, or mechanical damage may increase the likelihood of deeper NP penetration [337]. TiO2NPs and ZnONPs, used in sunscreens, have generated interest due to their potential to induce oxidative stress in skin cells.
These can induce oxidative stress and inflammation in skin cells. ZnONPs were found to inhibit cell proliferation and cause DNA damage in human keratinocytes (HaCaT cells) [338]. However, extensive systemic absorption has yet to be conclusively demonstrated, and some researchers have suggested that the risk of dermal exposure could be exaggerated [339].

2.4.3. Oral

Oral exposure is another common route for NPs, intentional through NP-based drug formulations [320,340]. The GIT can influence the fate of ingested NPs by exposing them to digestive enzymes, acidic pH, and the microbiota, which can modify the physicochemical properties of NPs [341,342]. NP uptake by intestinal epithelial cells occurs via transcellular or paracellular pathways; NPs pass through the intestinal barrier and enter the blood circulation, where they accumulate in organs such as the liver and spleen [343,344]. As the liver plays a central role in NP clearance and detoxification, HepG2 cells have thus been used extensively to assess hepatic metabolism and cytotoxicity of ingested NPs [345,346]. In addition to HepG2 cells, Caco-2 human colorectal adenocarcinoma cells are extensively used to model the intestinal epithelium for studying NP absorption and transport mechanisms [347]. Furthermore, another human colorectal adenocarcinoma-derived cell line, HT-29 line, is used to study mucus-secreting characteristics and the effects of NPs on intestinal mucus production [348,349].

2.4.4. Systemic

Intravenous and systemic administration are other widely used exposure routes for NPs in biomedical applications [7,350]. However, NP clearance by the circulating immune cells [345] encourages the progress of long circulating nanocarriers, typically comprising stealth coatings such as PEG to minimize immune identification [87,351]. Despite these innovations, concerns persist regarding NP-induced immunogenicity, thrombosis, and organ accumulation following intravenous administration [352,353]. Commonly used cells for testing genotoxicity caused by NPs are human lymphoblastoid TK6 cells, which have an intact p53 tumour suppressor gene, enabling evaluation of responses to DNA damage due to p53’s well-characterized role in mediating cell cycle progression and apoptosis in response to genotoxic stress. This is particularly relevant, as cells carrying mutated or deficient p53 can respond differently to DNA damage, potentially leading to misleading results in genotoxicity assessments.

2.4.5. Mucosal

Another potential route of NP uptake is mucosal exposure, including ocular and nasal absorption [354,355]. PSNs have emerged as an extremely versatile platform for controlled drug release and targeted drug delivery, in which the route of administration is a key factor affecting therapeutic outcome. This strategy could be useful for delivering therapeutic agents to the central nervous system to treat neurological diseases [355,356], for promoting the penetration of drugs through ocular tissues, enhancing the therapeutic effect in diseases such as glaucoma and macular degeneration, nasal delivery of therapeutics to the brain through the olfactory nerve pathway, bypassing the BBB and delivering 5-fluorouracil as a topical treatment for ocular cancer [357,358,359]. While these approaches present significant therapeutic potential, research underscores the importance of a comprehensive evaluation of NP-induced irritation and toxicity in mucosal tissues [360,361,362].
The choice of PSNs for the administration route is based on various factors, including their physicochemical properties, the disease to be treated, and the desired release kinetics. For example, these can be functionalised for mucosal delivery, allowing mucoadhesion and escape from immune recognition associated with systemic delivery [363]. By modifying these features, PSNs can overcome biological barriers to achieve precise drug delivery, thereby improving the efficacy of drug treatment.

2.4.6. Biodegradation and Clearance

An obvious hindrance to PSNs serving as delivery vehicles is their poor biodegradation. PSNs could accumulate within nearby healthy tissues, potentially inducing toxic effects. The mechanism for biodegradation of PSNs is described as a three-step process. This occurs via a nucleophilic reaction between hydroxyl groups in aqueous media and non-bridged oxygen atoms on the PSN surface. The three steps of PSN degradation (Figure 7) are as follows: Step 1 outlines the hydration phase, in which the siloxane skeleton takes up water molecules. Step 2 is hydrolysis, where Si-O-Si bonds are cleaved to form silanol groups. Step 3 is dissolution, the conversion of silanol to soluble orthosilicic acid via ion exchange with hydroxy groups [364]. The resulting silicic acid is water-soluble and can be excreted through renal clearance; however, incomplete degradation can result in accumulation in the liver, spleen or lungs, contributing to chronic toxicity.
The rate and extent of PSN degradation depend strongly on physicochemical parameters and environmental factors such as pH and ionic strength. Smaller particles with thinner pore walls and hydrophilic surfaces typically degrade faster, while surface coatings or protein corona formation may hinder hydrolysis by shielding reactive sites. Acidic or enzymatic conditions, such as those in lysosomes, accelerate degradation, whereas neutral physiological conditions slow it down [36].
In addition, the reconstruction of an organic-inorganic structure could significantly enhance PSNs degradation. For example, co-condensation with organosilanes can introduce cleavable Si-C or Si-S bonds, making the framework amenable under physiological conditions. Similarly, doping with biodegradable elements such as calcium, zinc, or iron can weaken the Si-O-Si bonds and promote hydrolytic dissolution. The development of stimuli-responsive PSNs that degrade under acidic, redox, or enzyme-triggered environments could further enhance their safety profile [191]. Ultimately, understanding and optimizing PSN degradation is crucial to ensure therapeutic efficacy while maintaining biocompatibility and facilitating clinical translation.
To visualize the toxicity endpoints and mechanisms discussed, Figure 8 outlines the possible sequence of events following PSN exposure, from surface interactions and internalization to intracellular responses such as oxidative stress, DNA damage, and activation of stress-related signalling cascades that lead to apoptosis, inflammation, DNA repair, and cell cycle arrest.

2.5. Evaluation of Clinical Translation

Evaluation of clinical translation is a critical component of SbD approaches, ensuring that PSNs can progress from preclinical research to clinical trials [28,365,366]. This stage involves assessing relevant animal studies, that reflect human physiology [9]. In vivo animal studies are critical for establishing the pharmacokinetic behaviour of PSNs in whole organisms. However, there is limited published data on PSNs assessed in larger animals; hence, more studies are needed to bridge the gap between rodent models and larger animals to enable the clinical translation of PSNs under the S.A.F.E. framework [5,367]. Also, various international bodies are working to ensure the safety of NPs and are moving toward establishing regulatory frameworks to address their potential risks. The Organisation for Economic Co-operation and Development (OECD) has developed guidelines for testing nanomaterials to standardize safety assessments and ensure the responsible development of nanotechnologies [368,369,370]. Similarly, the European Union’s Scientific Committee on Emerging and Newly Identified Health Risks provides scientific opinions on emerging health risks posed by new materials, including nanomaterials such as PSNs, especially in consumer products and medical applications [371]. Likewise, the United States Food and Drug Administration (FDA) issued NP-focused guidance for drug and medical device approval, with greater emphasis on physicochemical characterization, toxicity, and biodistribution [372,373]. Furthermore, the International Organization for Standardization (ISO) has developed standards such as ISO/TR 10993-22:2017, which provide protocols for assessing the biological effects of nanomaterials in medical devices [374]. These regulatory efforts highlight the need for interdisciplinary collaboration to address the safety concerns associated with PSNs, ensuring their safe and effective use in biomedical and industrial applications [375].
Additionally, considerations for manufacturing scalability and costs determine the practicality of translating laboratory-scale synthesis to industrial manufacturing, while maintaining material and performance consistency. Quality control measures are needed to avoid batch-to-batch variability and regulatory compliance [376]. Problems with reproducibility and size control continue to be an active area of research [377,378,379]. Finally, effective risk governance frameworks, including risk assessments, management, and communication throughout the life cycle, are vital to support informed decision-making and promote the clinical development of PSNs [380,381].

3. Summary

Many investigations have emphasized the importance of in-depth characterization of drug delivery-based NPs [75,382]. PSN characterization is essential for determining how it correlates with biological interactions, particularly in its role as a drug-delivery vehicle. In summary, the detailed characterization of PSNs is crucial for predicting and controlling their toxicity in humans. Data on particle size, pore size, functionalisation, surface charge, and crystallinity are key to understanding the utility of PSNs and enabling their effective and safe use.
Integrating findings on cellular uptake, protein corona dynamics, and immunogenic responses is essential for evaluating the biocompatibility and safety of PSNs. Cellular uptake efficiency determines the NP’s therapeutic potential [383], while the protein corona influences cellular interactions and immune responses [384]. Therefore, it is important to understand the interplay between these factors in PSNs, as this would dictate the overall biological outcome, including potential toxicity or therapeutic success [385]. By combining these findings, future studies will provide deeper insights into optimizing PSN design for safe and effective biomedical applications and the potential consequences of immune system activation. Understanding the principles of cytotoxicity assays and the various parameters that affect cytotoxicity underscores the importance of cytotoxicity testing. Surface functionalisation, exposure time, and the selection of diverse cell lines each play crucial roles in determining the safety and behaviour of PSNs within biological systems. Also, incorporating genotoxicity testing, oxidative stress assessments, and gene expression analysis into PSN toxicity studies is crucial for a thorough understanding of their biological impacts. These complementary approaches provide a robust evaluation of PSN interactions with cellular environments. They are essential for predicting potential health risks and advancing their application in biomedical fields while minimizing adverse effects. This multifaceted approach meets regulatory requirements and supports the development of safer, more effective PSNs for biomedical applications.
Finally, the S.A.F.E. PSNs framework (Table 4) provides a structured approach to developing and evaluating PSNs within SbD principles, ensuring their safety, efficacy and regulatory compliance. By integrating standardized characterization, toxicity assessment, relevant exposure models and clinical translation, this framework promotes responsible innovation while bridging the gap between laboratory research and real-world application. Ultimately, coordinated international regulatory effects and risk governance are key to supporting the safe and sustainable development of PSNs for clinical and industrial use. A summary (Table 4) highlighting current knowledge gaps and key research questions has been included, outlining areas where research remains limited and where future investigations should focus to advance PSN design and support safe clinical translation.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/nano15231766/s1. Section S1: Cytotoxicity assays and Section S2: Genotoxicity assays.

Author Contributions

Conceptualization, N.S. and T.P.; writing—original draft preparation, T.P. and C.C.; writing—review and editing, N.S., T.P., U.V.G. and Z.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

No new data was created or analyzed in this manuscript.

Acknowledgments

During the preparation of this manuscript, BioRender was used for the purpose of creating the figures.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
αAlpha
βBeta
OHHydroxyl radical
16HBE14oHuman bronchial epithelial cells
3T3Mouse fibroblast cells
A2780Human ovarian cancer cells
A549Human lung cancer cells
AgNPsSilver nanoparticles
AKTProtein kinase B
AMPKAdenosine monophosphate-activated protein kinase
APCAntigen presenting cells
AtgAutophagy-related genes
ATMAtaxia telangiectasia mutated
ATRAtaxia telangiectasia and Rad3-related
AuNPGold nanoparticle
BADBcl2-associated agonist of cell death
BAKB-cell lymphoma 2 (BCL-2) antagonist
BALHuman bronchioalveolar cells
BAXBCL-2-associated X protein
BBBBlood–brain barrier
Bcl-2B-cell lymphoma 2
Bcl-XLB-cell lymphoma 2-extra-large
BEAS-2BHuman bronchial epithelial cells
BSABovine serum albumin
C3Complement protein 3
C3bComplement protein 3b
C5Complement 5 protein
C6Complement 6 protein
C7Complement 7 protein
C8Complement 8 protein
C9Complement 9 protein
Caco-2Human colon cancer cells
Calu-3Human lung adenocarcinoma cells
CBMNCytokinesis block micronucleus
CDKCyclin-dependent kinase
CDK2Cyclin-dependent kinase 2
DCFDA2′,7′-dichlorofluorescein diacetate
DLSDynamic light scattering
DRAM1DNA Damage-Regulated Autophagy Modulator 1
e.g.,Exempli gratia, for example
EPREnhanced permeability and retention
ERK1Extracellular-signal-regulating kinase 1
ERK2Extracellular-signal-regulating kinase 2
et al.Et alii
FDAFood and Drug Administration
GITGastrointestinal tract
GPxGlutathione peroxidase
GRO-αGrowth-regulated oncogene alpha
H2O2Hydrogen peroxide
HeLaHuman cervical cancer cells
HepG2Human hepatocellular carcinoma cells
hhours
HT-29Human colorectal adenocarcinoma-derived cells
HUVECHuman umbilical vein endothelial cells
IL-1βInterleukin 1β
IL-6Interleukin 6
IONPIron oxide nanoparticle
ISOInternational Organisation for Standardisation
JNKc-Jun-terminal kinase
MACMembrane attack complex
MAPKMitogen-activated protein kinase
MASPMannose-binding lectin associated serine proteases
MBLMannose binding lectin
MCF-7Human breast cancer cells
MDA-MB 231Human breast cancer cells
MDM2Murine double minute 2
MOMPMitochondrial outer membrane permeabilisation
MPSMononuclear phagocyte system
MSNMesoporous silica nanoparticle
mTORMammalian Target of Rapamycin
MTT3- (4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
NF-kBNuclear factor kappa B
NINPNiobium oxide nanoparticle
NiNPNickel nanoparticle
NLRP3NOD-like receptor protein 3
NOXAPhorbal-12-myristate-13-acetate-induced protein 1
NPNanoparticle
O2Superoxide anion
OECDOrganisation for Economic Co-Operation and Development
p-NF-kBPhosphorylated nuclear factor kappa B
P13KPhosphatidylinositol-4,5-bisphosphate 3-kinase
p21Cyclin-dependent kinase inhibitor 1A (CKDN1A)
p53Tumour suppressor protein
PEGPolyethylene glycol
PEIPolyethylenimine
PDGF-AAPlatelet-derived growth factor-AA
PIPropidium Iodide
PSNPorous silica nanoparticle
PUMAp53 upregulated modulator of apoptosis
ROSReactive oxygen species
SbDSafe-by-Design
SDS-PAGESodium dodecyl sulphate-polyacrylamide gel electrophoresis
SiSilicon
SiNPSilica nanoparticle
SiO2NPSilicon dioxide nanoparticle
SODSuperoxide dismutase
SWCNTSingle-walled carbon nanotube
THP-1Human monocyte cells
TGF-1Transforming growth factor beta 1
TiO2Titanium dioxide
TK6Human lymphocyte cells
TNF-αTumour necrotic factor-α
TP53Tumour suppressor gene
TSC1 Hamartin
TSC2Tuberin
ZnOZinc oxide
µgMicrogram

References

  1. Kazemzadeh, P.; Sayadi, K.; Toolabi, A.; Sayadi, J.; Zeraati, M.; Chauhan, N.P.S.; Sargazi, G. Structure-Property Relationship for Different Mesoporous Silica Nanoparticles and Its Drug Delivery Applications: A Review. Front. Chem. 2022, 10, 823785. [Google Scholar] [CrossRef]
  2. Fang, L.; Zhou, H.; Cheng, L.; Wang, Y.; Liu, F.; Wang, S. The Application of Mesoporous Silica Nanoparticles as a Drug Delivery Vehicle in Oral Disease Treatment. Front. Cell. Infect. Microbiol. 2023, 13, 1124411. [Google Scholar] [CrossRef] [PubMed]
  3. Zhou, M.; Ge, X.; Ke, D.M.; Tang, H.; Zhang, J.Z.; Calvaresi, M.; Gao, B.; Sun, L.; Su, Q.; Wang, H. The Bioavailability, Biodistribution, and Toxic Effects of Silica-Coated Upconversion Nanoparticles in Vivo. Front. Chem. 2019, 7, 218. [Google Scholar] [CrossRef]
  4. Yu, J.; Dan, N.; Eslami, S.M.; Lu, X. State of the Art of Silica Nanoparticles: An Overview on Biodistribution and Preclinical Toxicity Studies. AAPS J. 2024, 26, 35. [Google Scholar] [CrossRef]
  5. Lérida-Viso, A.; Estepa-Fernández, A.; García-Fernández, A.; Martí-Centelles, V.; Martínez-Máñez, R. Biosafety of Mesoporous Silica Nanoparticles; towards Clinical Translation. Adv. Drug Deliv. Rev. 2023, 201, 115049. [Google Scholar] [CrossRef] [PubMed]
  6. Fatima, R.; Katiyar, P.; Kushwaha, K. Recent Advances in Mesoporous Silica Nanoparticle: Synthesis, Drug Loading, Release Mechanisms, and Diverse Applications. Front. Nanotechnol. 2025, 7, 1564188. [Google Scholar] [CrossRef]
  7. Kumarasamy, R.V.; Natarajan, P.M.; Umapathy, V.R.; Roy, J.R.; Mironescu, M.; Palanisamy, C.P. Clinical Applications and Therapeutic Potentials of Advanced Nanoparticles: A Comprehensive Review on Completed Human Clinical Trials. Front. Nanotechnol. 2024, 6, 1479993. [Google Scholar] [CrossRef]
  8. Janjua, T.I.; Cao, Y.; Yu, C.; Popat, A. Clinical Translation of Silica Nanoparticles. Nat. Rev. Mater. 2021, 6, 1072–1074. [Google Scholar] [CrossRef] [PubMed]
  9. Janjua, T.I.; Cao, Y.; Kleitz, F.; Linden, M.; Yu, C.; Popat, A. Silica Nanoparticles: A Review of Their Safety and Current Strategies to Overcome Biological Barriers. Adv. Drug Deliv. Rev. 2023, 203, 115115. [Google Scholar] [CrossRef]
  10. National Library of Medicine Safety Evaluation of Porous Silica in Men. Available online: https://www.clinicaltrials.gov/study/NCT03667430 (accessed on 13 October 2025).
  11. National Library of Medicine Effect of Different NanoScaffolds on Pulp Regeneration in Non-Vital Immature Permanent Teeth. Available online: https://clinicaltrials.gov/study/NCT07121348 (accessed on 13 October 2025).
  12. Zanoni, D.K.; Stambuk, H.E.; Madajewski, B.; Montero, P.H.; Matsuura, D.; Busam, K.J.; Ma, K.; Turker, M.Z.; Sequeira, S.; Gonen, M.; et al. Use of Ultrasmall Core-Shell Fluorescent Silica Nanoparticles for Image-Guided Sentinel Lymph Node Biopsy in Head and Neck Melanoma. JAMA Netw. Open 2021, 4, e211936. [Google Scholar] [CrossRef]
  13. National Library of Medicine Targeted Silica Nanoparticles for Real-Time Image-Guided Intraoperative Mapping of Nodal Metastases. Available online: https://www.clinicaltrials.gov/study/NCT02106598 (accessed on 13 October 2025).
  14. Baek, J.; Robert-Nicoud, G.; Herrera Hidalgo, C.; Borg, M.L.; Iqbal, M.N.; Berlin, R.; Lindgren, M.; Waara, E.; Uddén, A.; Pietiläinen, K.; et al. Engineered Mesoporous Silica Reduces Long-Term Blood Glucose, HbA1c, and Improves Metabolic Parameters in Prediabetics. Nanomedicine 2022, 17, 9–22. [Google Scholar] [CrossRef] [PubMed]
  15. National Library of Medicine STAR Study Investigating Performance and Safety of the Medical Device SiPore15 TM. Available online: https://clinicaltrials.gov/study/NCT03823027?a=3 (accessed on 13 October 2025).
  16. Benkő, F.; Kristó, K.; Sovány, T. Mesoporous Silica Nanoparticles as Drug Delivery Systems. Pharmaceuticals 2025, 18, 1392. [Google Scholar] [CrossRef]
  17. Zhang, W.; Liu, H.; Qiu, X.; Zuo, F.; Wang, B. Mesoporous Silica Nanoparticles as a Drug Delivery Mechanism. Open Life Sci. 2024, 19, 20220867. [Google Scholar] [CrossRef]
  18. Porrang, S.; Davaran, S.; Rahemi, N.; Allahyari, S.; Mostafavi, E. How Advancing Are Mesoporous Silica Nanoparticles? A Comprehensive Review of the Literature. Int. J. Nanomed. 2022, 17, 1803–1827. [Google Scholar] [CrossRef]
  19. Sayed, E.; Karavasili, C.; Ruparelia, K.; Haj-Ahmad, R.; Charalambopoulou, G.; Steriotis, T.; Giasafaki, D.; Cox, P.; Singh, N.; Giassafaki, L.P.N.; et al. Electrosprayed Mesoporous Particles for Improved Aqueous Solubility of a Poorly Water Soluble Anticancer Agent: In Vitro and Ex Vivo Evaluation. J. Control. Release 2018, 278, 142–155. [Google Scholar] [CrossRef]
  20. Alyassin, Y.; Sayed, E.G.; Mehta, P.; Ruparelia, K.; Arshad, M.S.; Rasekh, M.; Shepherd, J.; Kucuk, I.; Wilson, P.B.; Singh, N.; et al. Application of Mesoporous Silica Nanoparticles as Drug Delivery Carriers for Chemotherapeutic Agents. Drug Discov. Today 2020, 25, 1513–1520. [Google Scholar] [CrossRef]
  21. Sayed, E.; Ruparelia, K.; Zafar, S.; Faheem, A.; Fatouros, D.; Arshad, M.S.; Singh, N.; Ahmad, Z. Novel Electrosprayed Core-Shell Polyethyleneimine and Phospholipid Coated MSNs for Co-Delivery of KAZ3 and MDR-1 SiRNA for Efficient Chemotherapy in Multidrug-Resistant Colon Cancer. Eur. J. Pharm. Biopharm. 2025, 215, 114838. [Google Scholar] [CrossRef]
  22. Yan, J.; Siwakoti, P.; Shaw, S.; Bose, S.; Kokil, G.; Kumeria, T. Porous Silicon and Silica Carriers for Delivery of Peptide Therapeutics. Drug Deliv. Transl. Res. 2024, 14, 3549–3567. [Google Scholar] [CrossRef] [PubMed]
  23. Pamshong, S.R.; Bhatane, D.; Sarnaik, S.; Alexander, A. Mesoporous Silica Nanoparticles: An Emerging Approach in Overcoming the Challenges with Oral Delivery of Proteins and Peptides. Colloids Surf. B Biointerfaces 2023, 232, 113613. [Google Scholar] [CrossRef]
  24. Wei, J.; Tan, Y.; Bai, Y.; He, J.; Cao, H.; Guo, J.; Su, Z. Mesoporous Silicon Nanoparticles with Liver-Targeting and PH-Response-Release Function Are Used for Targeted Drug Delivery in Liver Cancer Treatment. Int. J. Mol. Sci. 2024, 25, 2525. [Google Scholar] [CrossRef]
  25. Castillo, R.R.; Lozano, D.; Vallet-Regí, M. Mesoporous Silica Nanoparticles as Carriers for Therapeutic Biomolecules. Pharmaceutics 2020, 12, 432. [Google Scholar] [CrossRef]
  26. Martins, J.P.; Figueiredo, P.; Wang, S.; Espo, E.; Celi, E.; Martins, B.; Kemell, M.; Moslova, K.; Mäkilä, E.; Salonen, J.; et al. Neonatal Fc Receptor-Targeted Lignin-Encapsulated Porous Silicon Nanoparticles for Enhanced Cellular Interactions and Insulin Permeation across the Intestinal Epithelium. Bioact. Mater. 2022, 9, 299–315. [Google Scholar] [CrossRef]
  27. Valdés-Sánchez, L.; Borrego-González, S.; Montero-Sánchez, A.; Massalini, S.; de la Cerda, B.; Díaz-Cuenca, A.; Díaz-Corrales, F.J. Mesoporous Silica-Based Nanoparticles as Non-Viral Gene Delivery Platform for Treating Retinitis Pigmentosa. J. Clin. Med. 2022, 11, 2170. [Google Scholar] [CrossRef] [PubMed]
  28. Liu, H.J.; Xu, P. Smart Mesoporous Silica Nanoparticles for Protein Delivery. Nanomaterials 2019, 9, 511. [Google Scholar] [CrossRef]
  29. Jänicke, P.; Lennicke, C.; Meister, A.; Seliger, B.; Wessjohann, L.A.; Kaluđerović, G.N. Fluorescent Spherical Mesoporous Silica Nanoparticles Loaded with Emodin: Synthesis, Cellular Uptake and Anticancer Activity. Mater. Sci. Eng. C 2021, 119, 111619. [Google Scholar] [CrossRef] [PubMed]
  30. Yuan, D.; Ellis, C.M.; Davis, J.J. Mesoporous Silica Nanoparticles in Bioimaging. Materials 2020, 13, 3795. [Google Scholar] [CrossRef]
  31. Li, Z.; Guo, J.; Zhang, M.; Li, G.; Hao, L. Gadolinium-Coated Mesoporous Silica Nanoparticle for Magnetic Resonance Imaging. Front. Chem. 2022, 10, 837032. [Google Scholar] [CrossRef]
  32. Kankala, R.K.; Han, Y.H.; Xia, H.Y.; Wang, S.B.; Chen, A.Z. Nanoarchitectured Prototypes of Mesoporous Silica Nanoparticles for Innovative Biomedical Applications. J. Nanobiotechnol. 2022, 20, 126. [Google Scholar] [CrossRef] [PubMed]
  33. Moschini, E.; Colombo, G.; Chirico, G.; Capitani, G.; Dalle-Donne, I.; Mantecca, P. Biological Mechanism of Cell Oxidative Stress and Death during Short-Term Exposure to Nano CuO. Sci. Rep. 2023, 13, 2326. [Google Scholar] [CrossRef]
  34. Yu, Z.; Li, Q.; Wang, J.; Yu, Y.; Wang, Y.; Zhou, Q.; Li, P. Reactive Oxygen Species-Related Nanoparticle Toxicity in the Biomedical Field. Nanoscale Res. Lett. 2020, 15, 115. [Google Scholar] [CrossRef]
  35. Mitchell, M.J.; Billingsley, M.M.; Haley, R.M.; Wechsler, M.E.; Peppas, N.A.; Langer, R. Engineering Precision Nanoparticles for Drug Delivery. Nat. Rev. Drug Discov. 2021, 20, 101–124. [Google Scholar] [CrossRef]
  36. Zhang, Y.; Lin, X.; Chen, X.; Fang, W.; Yu, K.; Gu, W.; Wei, Y.; Zheng, H.; Piao, J.; Li, F. Strategies to Regulate the Degradation and Clearance of Mesoporous Silica Nanoparticles: A Review. Int. J. Nanomed. 2024, 19, 5859–5878. [Google Scholar] [CrossRef]
  37. Shi, L.; Zhang, J.; Zhao, M.; Tang, S.; Cheng, X.; Zhang, W.; Li, W.; Liu, X.; Peng, H.; Wang, Q. Effects of Polyethylene Glycol on the Surface of Nanoparticles for Targeted Drug Delivery. Nanoscale 2021, 13, 10748–10764. [Google Scholar] [CrossRef] [PubMed]
  38. Chi, M.; Li, N.; Cui, J.; Karlin, S.; Rohr, N.; Sharma, N.; Thieringer, F.M. Biomimetic, Mussel-Inspired Surface Modification of 3D-Printed Biodegradable Polylactic Acid Scaffolds with Nano-Hydroxyapatite for Bone Tissue Engineering. Front. Bioeng. Biotechnol. 2022, 10, 989729. [Google Scholar] [CrossRef]
  39. Zhao, C.; Zhou, B. Polyethyleneimine-Based Drug Delivery Systems for Cancer Theranostics. J. Funct. Biomater. 2023, 14, 12. [Google Scholar] [CrossRef] [PubMed]
  40. Ouyang, X.; Liu, Y.; Zheng, K.; Pang, Z.; Peng, S. Recent Advances in Zwitterionic Nanoscale Drug Delivery Systems to Overcome Biological Barriers. Asian J. Pharm. Sci. 2024, 19, 100883. [Google Scholar] [CrossRef]
  41. Colli, C.; Masi, I.; Jacchetti, E.; Santoni, S.; Sponchioni, M.; Colosimo, B.M.; Rosanò, L.; Raimondi, M.T.; Mauri, E.; Moscatelli, D. Zwitterionic Nanoparticles for Thermally Activated Drug Delivery in Hyperthermia Cancer Treatment. Nanoscale 2024, 16, 12635–12649. [Google Scholar] [CrossRef]
  42. Mohammadi Dargah, M.; Pedram, P.; Cabrera-Barjas, G.; Delattre, C.; Nesic, A.; Santagata, G.; Cerruti, P.; Moeini, A. Biomimetic Synthesis of Nanoparticles: A Comprehensive Review on Green Synthesis of Nanoparticles with a Focus on Prosopis Farcta Plant Extracts and Biomedical Applications. Adv. Colloid Interface Sci. 2024, 332, 103277. [Google Scholar] [CrossRef]
  43. Tikhonov, A.; Kachanov, A.; Yudaeva, A.; Danilik, O.; Ponomareva, N.; Karandashov, I.; Kostyusheva, A.; Zamyatnin, A.A.; Parodi, A.; Chulanov, V.; et al. Biomimetic Nanoparticles for Basic Drug Delivery. Pharmaceutics 2024, 16, 1306. [Google Scholar] [CrossRef]
  44. Dekkers, S.; Wijnhoven, S.W.P.; Braakhuis, H.M.; Soeteman-Hernandez, L.G.; Sips, A.J.A.M.; Tavernaro, I.; Kraegeloh, A.; Noorlander, C.W. Safe-by-Design Part I: Proposal for Nanospecific Human Health Safety Aspects Needed along the Innovation Process. NanoImpact 2020, 18, 100227. [Google Scholar] [CrossRef]
  45. Mech, A.; Gottardo, S.; Amenta, V.; Amodio, A.; Belz, S.; Bøwadt, S.; Drbohlavová, J.; Farcal, L.; Jantunen, P.; Małyska, A.; et al. Safe- and Sustainable-by-Design: The Case of Smart Nanomaterials. A Perspective Based on a European Workshop. In Regulatory Toxicology and Pharmacology; Academic Press Inc.: Cambridge, MA, USA, 2022; Volume 128. [Google Scholar]
  46. Schmutz, M.; Borges, O.; Jesus, S.; Borchard, G.; Perale, G.; Zinn, M.; Sips, Ä.A.J.A.M.; Soeteman-Hernandez, L.G.; Wick, P.; Som, C. A Methodological Safe-by-Design Approach for the Development of Nanomedicines. Front. Bioeng. Biotechnol. 2020, 8, 258. [Google Scholar] [CrossRef]
  47. Sabourian, P.; Yazdani, G.; Ashraf, S.S.; Frounchi, M.; Mashayekhan, S.; Kiani, S.; Kakkar, A. Effect of Physico-Chemical Properties of Nanoparticles on Their Intracellular Uptake. Int. J. Mol. Sci. 2020, 21, 8019. [Google Scholar] [CrossRef]
  48. Sanità, G.; Carrese, B.; Lamberti, A. Nanoparticle Surface Functionalization: How to Improve Biocompatibility and Cellular Internalization. Front. Mol. Biosci. 2020, 7, 587012. [Google Scholar] [CrossRef]
  49. Yuan, T.; Gao, L.; Zhan, W.; Dini, D. Effect of Particle Size and Surface Charge on Nanoparticles Diffusion in the Brain White Matter. Pharm. Res. 2022, 39, 767–781. [Google Scholar] [CrossRef]
  50. Ahmadi, A.; Sokunbi, M.; Patel, T.; Chang, M.W.; Ahmad, Z.; Singh, N. Influence of Critical Parameters on Cytotoxicity Induced by Mesoporous Silica Nanoparticles. Nanomaterials 2022, 12, 2016. [Google Scholar] [CrossRef]
  51. Grunberger, J.W.; Newton, H.S.; Donohue, D.; Dobrovolskaia, M.A.; Ghandehari, H. Role of Physicochemical Properties in Silica Nanoparticle-Mediated Immunostimulation. Nanotoxicology 2024, 18, 599–617. [Google Scholar] [CrossRef]
  52. Herdiana, Y.; Wathoni, N.; Shamsuddin, S.; Muchtaridi, M. Scale-up Polymeric-Based Nanoparticles Drug Delivery Systems: Development and Challenges. OpenNano 2022, 7, 100048. [Google Scholar] [CrossRef]
  53. Perrigue, P.M.; Henschke, A.; Grześkowiak, B.F.; Przysiecka, Ł.; Jaskot, K.; Mielcarek, A.; Coy, E.; Moya, S.E. Cellular Uptake and Retention Studies of Silica Nanoparticles Utilizing Senescent Fibroblasts. Sci. Rep. 2023, 13, 475. [Google Scholar] [CrossRef] [PubMed]
  54. Sousa De Almeida, M.; Susnik, E.; Drasler, B.; Taladriz-Blanco, P.; Petri-Fink, A.; Rothen-Rutishauser, B. Understanding Nanoparticle Endocytosis to Improve Targeting Strategies in Nanomedicine. Chem. Soc. Rev. 2021, 50, 5397–5434. [Google Scholar] [CrossRef]
  55. Rancan, F.; Gao, Q.; Graf, C.; Troppens, S.; Hadam, S.; Hackbarth, S.; Kembuan, C.; Blume-Peytavi, U.; Rühl, E.; Lademann, J.; et al. Skin Penetration and Cellular Uptake of Amorphous Silica Nanoparticles with Variable Size, Surface Functionalization, and Colloidal Stability. ACS Nano 2012, 6, 6829–6842. [Google Scholar] [CrossRef] [PubMed]
  56. Zha, S.; Liu, H.; Li, H.; Li, H.; Wong, K.L.; All, A.H. Functionalized Nanomaterials Capable of Crossing the Blood-Brain Barrier. ACS Nano 2024, 18, 1820–1845. [Google Scholar] [CrossRef]
  57. Niroumand, U.; Firouzabadi, N.; Goshtasbi, G.; Hassani, B.; Ghasemiyeh, P.; Mohammadi-Samani, S. The Effect of Size, Morphology and Surface Properties of Mesoporous Silica Nanoparticles on Pharmacokinetic Aspects and Potential Toxicity Concerns. Front. Mater. 2023, 10, 1189463. [Google Scholar] [CrossRef]
  58. Ohta, S.; Kikuchi, E.; Ishijima, A.; Azuma, T.; Sakuma, I.; Ito, T. Investigating the Optimum Size of Nanoparticles for Their Delivery into the Brain Assisted by Focused Ultrasound-Induced Blood–Brain Barrier Opening. Sci. Rep. 2020, 10, 18220. [Google Scholar] [CrossRef]
  59. Colaço, M.; Marques, A.P.; Jesus, S.; Duarte, A.; Borges, O. Safe-by-Design of Glucan Nanoparticles: Size Matters When Assessing the Immunotoxicity. Chem. Res. Toxicol. 2020, 33, 915–932. [Google Scholar] [CrossRef]
  60. Chen, L.; Liu, J.; Zhang, Y.; Zhang, G.; Kang, Y.; Chen, A.; Feng, X.; Shao, L. The Toxicity of Silica Nanoparticles to the Immune System. Nanomedicine 2018, 13, 1939–1962. [Google Scholar] [CrossRef]
  61. Nabeshi, H.; Yoshikawa, T.; Akase, T.; Yoshida, T.; Tochigi, S.; Hirai, T.; Uji, M.; Ichihashi, K.-i.; Yamashita, T.; Higashisaka, K.; et al. Effect of Amorphous Silica Nanoparticles on in Vitro Rankl-Induced Osteoclast Differentiation in Murine Macrophages. Nanoscale Res. Lett. 2011, 6, 464. [Google Scholar] [CrossRef]
  62. He, Y.; Wang, Y.; Wang, L.; Jiang, W.; Wilhelm, S. Understanding Nanoparticle-Liver Interactions in Nanomedicine. Expert. Opin. Drug Deliv. 2024, 21, 829–843. [Google Scholar] [CrossRef] [PubMed]
  63. Sen Karaman, D.; Desai, D.; Senthilkumar, R.; Johansson, E.M.; Råtts, N.; Odén, M.; Eriksson, J.E.; Sahlgren, C.; Toivola, D.M.; Rosenholm, J.M. Shape Engineering vs Organic Modification of Inorganic Nanoparticles as a Tool for Enhancing Cellular Internalization. Nanoscale Res. Lett. 2012, 7, 358. [Google Scholar] [CrossRef] [PubMed]
  64. Bartneck, M.; Keul, H.A.; Singh, S.; Czaja, K.; Bornemann, J.; Bockstaller, M.; Moeller, M.; Zwadlo-Klarwasser, G.; Groll, J. Rapid Uptake of Gold Nanorods by Primary Human Blood Phagocytes and Immunomodulatory Effects of Surface Chemistry. ACS Nano 2010, 4, 3073–3086. [Google Scholar] [CrossRef]
  65. Zhang, X.; Ma, G.; Wei, W. Simulation of Nanoparticles Interacting with a Cell Membrane: Probing the Structural Basis and Potential Biomedical Application. NPG Asia Mater. 2021, 13, 52. [Google Scholar] [CrossRef]
  66. Zelepukin, I.V.; Shevchenko, K.G.; Deyev, S.M. Rediscovery of Mononuclear Phagocyte System Blockade for Nanoparticle Drug Delivery. Nat. Commun. 2024, 15, 4366. [Google Scholar] [CrossRef]
  67. Lu, J.; Gao, X.; Wang, S.; He, Y.; Ma, X.; Zhang, T.; Liu, X. Advanced Strategies to Evade the Mononuclear Phagocyte System Clearance of Nanomaterials. Exploration 2023, 3, 20220045. [Google Scholar] [CrossRef]
  68. Liu, Y.; Wang, Z.; Liu, Y.; Zhu, G.; Jacobson, O.; Fu, X.; Bai, R.; Lin, X.; Lu, N.; Yang, X.; et al. Suppressing Nanoparticle-Mononuclear Phagocyte System Interactions of Two-Dimensional Gold Nanorings for Improved Tumor Accumulation and Photothermal Ablation of Tumors. ACS Nano 2017, 11, 10539–10548. [Google Scholar] [CrossRef] [PubMed]
  69. Izci, M.; Maksoudian, C.; Manshian, B.B.; Soenen, S.J. The Use of Alternative Strategies for Enhanced Nanoparticle Delivery to Solid Tumors. Chem. Rev. 2021, 121, 1746–1803. [Google Scholar] [CrossRef]
  70. Nowak-Jary, J.; Machnicka, B. Pharmacokinetics of Magnetic Iron Oxide Nanoparticles for Medical Applications. J. Nanobiotechnol. 2022, 20, 305. [Google Scholar] [CrossRef] [PubMed]
  71. Li, J.; Shen, S.; Kong, F.; Jiang, T.; Tang, C.; Yin, C. Effects of Pore Size on in Vitro and in Vivo Anticancer Efficacies of Mesoporous Silica Nanoparticles. RSC Adv. 2018, 8, 24633–24640. [Google Scholar] [CrossRef] [PubMed]
  72. Giri, K.; Kuschnerus, I.; Lau, M.; Ruan, J.; Garcia-Bennett, A. Pore Structure and Particle Shape Modulates the Protein Corona of Mesoporous Silica Particles. Mater. Adv. 2020, 1, 599–603. [Google Scholar] [CrossRef]
  73. Ma, Z.; Bai, J.; Wang, Y.; Jiang, X. Impact of Shape and Pore Size of Mesoporous Silica Nanoparticles on Serum Protein Adsorption and RBCs Hemolysis. ACS Appl. Mater. Interfaces 2014, 6, 2431–2438. [Google Scholar] [CrossRef]
  74. Hong, X.; Zhong, X.; Du, G.; Hou, Y.; Zhang, Y.; Zhang, Z.; Gong, T.; Zhang, L.; Sun, X. The Pore Size of Mesoporous Silica Nanoparticles Regulates Their Antigen Delivery Efficiency. Sci. Adv. 2020, 6, eaaz4462. [Google Scholar] [CrossRef]
  75. Awashra, M.; Młynarz, P. The Toxicity of Nanoparticles and Their Interaction with Cells: An in Vitro Metabolomic Perspective. Nanoscale Adv. 2023, 5, 2674–2723. [Google Scholar] [CrossRef]
  76. Sood, V.; Katti, D.S. Physicochemical Changes in Plasma Membrane Mirror Nanoparticle-Mediated Cytotoxicity. bioRxiv 2019. [Google Scholar] [CrossRef]
  77. Prabhakar, N.; Merisaari, J.; Le Joncour, V.; Peurla, M.; Karaman, D.Ş.; Casals, E.; Laakkonen, P.; Westermarck, J.; Rosenholm, J.M. Circumventing Drug Treatment? Intrinsic Lethal Effects of Polyethyleneimine (Pei)-Functionalized Nanoparticles on Glioblastoma Cells Cultured in Stem Cell Conditions. Cancers 2021, 13, 2631. [Google Scholar] [CrossRef]
  78. Monteiro-Riviere, N.A.; Samberg, M.E.; Oldenburg, S.J.; Riviere, J.E. Protein Binding Modulates the Cellular Uptake of Silver Nanoparticles into Human Cells: Implications for in Vitro to in Vivo Extrapolations? Toxicol. Lett. 2013, 220, 286–293. [Google Scholar] [CrossRef]
  79. Zhu, Y.; Yue, M.; Guo, T.; Li, F.; Li, Z.; Yang, D.; Lin, M. PEI-PEG-Coated Mesoporous Silica Nanoparticles Enhance the Antitumor Activity of Tanshinone IIA and Serve as a Gene Transfer Vector. Evid. Based Complement. Altern. Med. 2021, 2021, 6756763. [Google Scholar] [CrossRef]
  80. Xu, B.; Li, S.; Shi, R.; Liu, H. Multifunctional Mesoporous Silica Nanoparticles for Biomedical Applications. Signal Transduct. Target. Ther. 2023, 8, 435. [Google Scholar] [CrossRef]
  81. Ly, P.D.; Ly, K.N.; Phan, H.L.; Nguyen, H.H.T.; Duong, V.A.; Nguyen, H.V. Recent Advances in Surface Decoration of Nanoparticles in Drug Delivery. Front. Nanotechnol. 2024, 6, 1456939. [Google Scholar] [CrossRef]
  82. Xuan, L.; Ju, Z.; Skonieczna, M.; Zhou, P.K.; Huang, R. Nanoparticles-Induced Potential Toxicity on Human Health: Applications, Toxicity Mechanisms, and Evaluation Models. MedComm 2023, 4, e327. [Google Scholar] [CrossRef] [PubMed]
  83. Ebrahiminezhad, A.; Rasoul-Amini, S.; Kouhpayeh, A.; Davaran, S.; Barar, J.; Ghasemi, Y. Impacts of Amine Functionalized Iron Oxide Nanoparticles on HepG2 Cell Line. Curr. Nanosci. 2014, 11, 113–119. [Google Scholar] [CrossRef]
  84. Matadamas-Ortiz, A.; Pérez-Robles, J.F.; Reynoso-Camacho, R.; Amaya-Llano, S.L.; Amaro-Reyes, A.; Di Pierro, P.; Regalado-González, C. Effect of Amine, Carboxyl, or Thiol Functionalization of Mesoporous Silica Particles on Their Efficiency as a Quercetin Delivery System in Simulated Gastrointestinal Conditions. Foods 2024, 13, 1208. [Google Scholar] [CrossRef] [PubMed]
  85. Cahill, H.F.; Scott, B.S.; Peter, O.O.; Stapleton, G.V.L.; MacCormack, T.J.; Meli, M.-V.; Rourke, J.L. Multiparametric Cytotoxicity Profiling Reveals Cell-Line and Ligand-Dependent Toxicity for Pegylated Gold Nanoparticles (AuNP-PEG). Can. J. Chem. 2024, 102, 134–143. [Google Scholar] [CrossRef]
  86. Pierre, V.C.; Pasek-Allen, J.L.; Wilharm, R.K.; Bischof, J.C. NMR Characterization of Polyethylene Glycol Conjugates for Nanoparticle Functionalization. ACS Omega 2023, 8, 4331–4336. [Google Scholar] [CrossRef]
  87. Suk, J.S.; Xu, Q.; Kim, N.; Hanes, J.; Ensign, L.M. PEGylation as a Strategy for Improving Nanoparticle-Based Drug and Gene Delivery. Adv. Drug Deliv. Rev. 2016, 99, 28–51. [Google Scholar] [CrossRef]
  88. He, Q.; Zhang, J.; Shi, J.; Zhu, Z.; Zhang, L.; Bu, W.; Guo, L.; Chen, Y. The Effect of PEGylation of Mesoporous Silica Nanoparticles on Nonspecific Binding of Serum Proteins and Cellular Responses. Biomaterials 2010, 31, 1085–1092. [Google Scholar] [CrossRef]
  89. Salmaso, S.; Caliceti, P. Stealth Properties to Improve Therapeutic Efficacy of Drug Nanocarriers. J. Drug Deliv. 2013, 2013, 374252. [Google Scholar] [CrossRef]
  90. Marques, A.C.; Costa, P.J.; Velho, S.; Amaral, M.H. Functionalizing Nanoparticles with Cancer-Targeting Antibodies: A Comparison of Strategies. J. Control. Release 2020, 320, 180–200. [Google Scholar] [CrossRef]
  91. Kumari, M.; Acharya, A.; Krishnamurthy, P.T. Antibody-Conjugated Nanoparticles for Target-Specific Drug Delivery of Chemotherapeutics. Beilstein J. Nanotechnol. 2023, 14, 912–926. [Google Scholar] [CrossRef]
  92. McDaid, W.J.; Lissin, N.; Pollheimer, E.; Greene, M.; Leach, A.; Smyth, P.; Bossi, G.; Longley, D.; Cole, D.K.; Scott, C.J. Enhanced Target-Specific Delivery of Docetaxel-Loaded Nanoparticles Using Engineered T Cell Receptors. Nanoscale 2021, 13, 15010–15020. [Google Scholar] [CrossRef]
  93. Papini, E.; Tavano, R.; Mancin, F. Opsonins and Dysopsonins of Nanoparticles: Facts, Concepts, and Methodological Guidelines. Front. Immunol. 2020, 11, 7365. [Google Scholar] [CrossRef] [PubMed]
  94. Pondman, K.; Le Gac, S.; Kishore, U. Nanoparticle-Induced Immune Response: Health Risk versus Treatment Opportunity? Immunobiology 2023, 228, 152317. [Google Scholar] [CrossRef] [PubMed]
  95. Vu, V.P.; Gifford, G.B.; Chen, F.; Benasutti, H.; Wang, G.; Groman, E.V.; Scheinman, R.; Saba, L.; Moghimi, S.M.; Simberg, D. Immunoglobulin Deposition on Biomolecule Corona Determines Complement Opsonization Efficiency of Preclinical and Clinical Nanoparticles. Nat. Nanotechnol. 2019, 14, 260–268. [Google Scholar] [CrossRef] [PubMed]
  96. Schorr, K.; Beck, S.; Zimmer, O.; Baumann, F.; Keller, M.; Witzgall, R.; Goepferich, A. The Quantity of Ligand–Receptor Interactions between Nanoparticles and Target Cells. Nanoscale Horiz. 2025, 10, 803–823. [Google Scholar] [CrossRef] [PubMed]
  97. Li, J.; Du, X.; Zheng, N.; Xu, L.; Xu, J.; Li, S. Contribution of Carboxyl Modified Chiral Mesoporous Silica Nanoparticles in Delivering Doxorubicin Hydrochloride in Vitro: PH-Response Controlled Release, Enhanced Drug Cellular Uptake and Cytotoxicity. Colloids Surf. B Biointerfaces 2016, 141, 374–381. [Google Scholar] [CrossRef]
  98. Zhang, C.; Xie, H.; Zhang, Z.; Wen, B.; Cao, H.; Bai, Y.; Che, Q.; Guo, J.; Su, Z. Applications and Biocompatibility of Mesoporous Silica Nanocarriers in the Field of Medicine. Front. Pharmacol. 2022, 13, 829796. [Google Scholar] [CrossRef]
  99. Ajit, J. Nanoparticles in Drug Delivery. Prem. J. Sci. 2025, 1, 100048. [Google Scholar] [CrossRef]
  100. Maccuaig, W.M.; Samykutty, A.; Foote, J.; Luo, W.; Filatenkov, A.; Li, M.; Houchen, C.; Grizzle, W.E.; McNally, L.R. Toxicity Assessment of Mesoporous Silica Nanoparticles upon Intravenous Injection in Mice: Implications for Drug Delivery. Pharmaceutics 2022, 14, 969. [Google Scholar] [CrossRef]
  101. Murugadoss, S.; Godderis, L.; Ghosh, M.; Hoet, P.H. Assessing the Toxicological Relevance of Nanomaterial Agglomerates and Aggregates Using Realistic Exposure in Vitro. Nanomaterials 2021, 11, 1793. [Google Scholar] [CrossRef]
  102. Cabellos, J.; Gimeno-Benito, I.; Catalán, J.; Lindberg, H.K.; Vales, G.; Fernandez-Rosas, E.; Ghemis, R.; Jensen, K.A.; Atluri, R.; Vázquez-Campos, S.; et al. Short-Term Oral Administration of Non-Porous and Mesoporous Silica Did Not Induce Local or Systemic Toxicity in Mice. Nanotoxicology 2020, 14, 1324–1341. [Google Scholar] [CrossRef]
  103. Voicu, S.N.P.; Dinu, D.; Sima, C.; Hermenean, A.; Ardelean, A.; Codrici, E.; Stan, M.S.; Zărnescu, O.; Dinischiotu, A. Silica Nanoparticles Induce Oxidative Stress and Autophagy but Not Apoptosis in the MRC-5 Cell Line. Int. J. Mol. Sci. 2015, 16, 29398–29416. [Google Scholar] [CrossRef]
  104. Proquin, H.; Rodríguez-Ibarra, C.; Moonen, C.G.J.; Urrutia Ortega, I.M.; Briedé, J.J.; de Kok, T.M.; van Loveren, H.; Chirino, Y.I. Titanium Dioxide Food Additive (E171) Induces ROS Formation and Genotoxicity: Contribution of Micro and Nano-Sized Fractions. Mutagenesis 2017, 32, 139–149. [Google Scholar] [CrossRef]
  105. Krug, H.F. A Systematic Review on the Hazard Assessment of Amorphous Silica Based on the Literature from 2013 to 2018. Front. Public Health 2022, 10, 902893. [Google Scholar] [CrossRef] [PubMed]
  106. Sukhanova, A.; Bozrova, S.; Sokolov, P.; Berestovoy, M.; Karaulov, A.; Nabiev, I. Dependence of Nanoparticle Toxicity on Their Physical and Chemical Properties. Nanoscale Res. Lett. 2018, 13, 44. [Google Scholar] [CrossRef]
  107. Dong, X.; Wu, Z.; Li, X.; Xiao, L.; Yang, M.; Li, Y.; Duan, J.; Sun, Z. The Size-Dependent Cytotoxicity of Amorphous Silica Nanoparticles: A Systematic Review of in Vitro Studies. Int. J. Nanomed. 2020, 15, 9089–9113. [Google Scholar] [CrossRef]
  108. Keller, J.G.; Graham, U.M.; Koltermann-Jülly, J.; Gelein, R.; Ma-Hock, L.; Landsiedel, R.; Wiemann, M.; Oberdörster, G.; Elder, A.; Wohlleben, W. Predicting Dissolution and Transformation of Inhaled Nanoparticles in the Lung Using Abiotic Flow Cells: The Case of Barium Sulfate. Sci. Rep. 2020, 10, 458. [Google Scholar] [CrossRef]
  109. Murugadoss, S.; Van Den Brule, S.; Brassinne, F.; Sebaihi, N.; Mejia, J.; Lucas, S.; Petry, J.; Godderis, L.; Mast, J.; Lison, D.; et al. Is Aggregated Synthetic Amorphous Silica Toxicologically Relevant? Part Fibre Toxicol. 2020, 17, 1. [Google Scholar] [CrossRef]
  110. Croissant, J.G.; Butler, K.S.; Zink, J.I.; Brinker, C.J. Synthetic Amorphous Silica Nanoparticles: Toxicity, Biomedical and Environmental Implications. Nat. Rev. Mater. 2020, 5, 886–909. [Google Scholar] [CrossRef]
  111. Martin; Watanabe, R.; Hashimoto, K.; Higashisaka, K.; Haga, Y.; Tsutsumi, Y.; Mizuguchi, K. Evidence-Based Prediction of Cellular Toxicity for Amorphous Silica Nanoparticles. ACS Nano 2023, 17, 9987–9999. [Google Scholar] [CrossRef] [PubMed]
  112. Huang, Y.; Li, P.; Zhao, R.; Zhao, L.; Liu, J.; Peng, S.; Fu, X.; Wang, X.; Luo, R.; Wang, R.; et al. Silica Nanoparticles: Biomedical Applications and Toxicity. Biomed. Pharmacother. 2022, 151, 113053. [Google Scholar] [CrossRef]
  113. Wang, J.; Ni, R.; Jiang, T.; Peng, D.; Ming, Y.; Cui, H.; Liu, Y. The Applications of Functional Materials-Based Nano-Formulations in the Prevention, Diagnosis and Treatment of Chronic Inflammation-Related Diseases. Front. Pharmacol. 2023, 14, 1222642. [Google Scholar] [CrossRef]
  114. Mohammadpour, R.; Yazdimamaghani, M.; Cheney, D.L.; Jedrzkiewicz, J.; Ghandehari, H. Subchronic Toxicity of Silica Nanoparticles as a Function of Size and Porosity. J. Control. Release 2019, 304, 216–232. [Google Scholar] [CrossRef] [PubMed]
  115. Bhattacharjee, S.; de Haan, L.H.J.; Evers, N.M.; Jiang, X.; Marcelis, A.T.M.; Zuilhof, H.; Rietjens, I.M.C.M.; Alink, G.M. Role of Surface Charge and Oxidative Stress in Cytotoxicity of Organic Monolayer-Coated Silicon Nanoparticles towards Macrophage NR8383 Cells. Part. Fibre Toxicol. 2010, 7, 25. [Google Scholar] [CrossRef] [PubMed]
  116. Kumar, K.; Singh, R.K.; Tyagi, P.K.; Gore, D. Assessment of Toxicity and Safety Profiles of Nanoparticles. Lett. Appl. NanoBioScience 2021, 10, 1877–1888. [Google Scholar] [CrossRef]
  117. Sun, H.; Jiang, C.; Wu, L.; Bai, X.; Zhai, S. Cytotoxicity-Related Bioeffects Induced by Nanoparticles: The Role of Surface Chemistry. Front. Bioeng. Biotechnol. 2019, 7, 414. [Google Scholar] [CrossRef]
  118. Andrée, L.; Joziasse, L.S.; Adjobo-Hermans, M.J.W.; Yang, F.; Wang, R.; Leeuwenburgh, S.C.G. Effect of Hydroxyapatite Nanoparticle Crystallinity and Colloidal Stability on Cytotoxicity. ACS Biomater. Sci. Eng. 2024, 10, 6964–6973. [Google Scholar] [CrossRef]
  119. Aplicada, B. An Approach to The Safety Assessment of Nanoparticles and Nanomaterials. Am. J. Biomed. Sci. Res. 2022, 16, 621–626. [Google Scholar] [CrossRef]
  120. Abbasi, R.; Shineh, G.; Mobaraki, M.; Doughty, S.; Tayebi, L. Structural Parameters of Nanoparticles Affecting Their Toxicity for Biomedical Applications: A Review. J. Nanoparticle Res. 2023, 25, 43. [Google Scholar] [CrossRef]
  121. Tirumala, M.G.; Anchi, P.; Raja, S.; Rachamalla, M.; Godugu, C. Novel Methods and Approaches for Safety Evaluation of Nanoparticle Formulations: A Focus Towards In Vitro Models and Adverse Outcome Pathways. Front. Pharmacol. 2021, 12, 612659. [Google Scholar] [CrossRef] [PubMed]
  122. Bhattacharjee, S.; Ershov, D.; Islam, M.A.; Kämpfer, A.M.; Maslowska, K.A.; Van Der Gucht, J.; Alink, G.M.; Marcelis, A.T.M.; Zuilhof, H.; Rietjens, I.M.C.M. Role of Membrane Disturbance and Oxidative Stress in the Mode of Action Underlying the Toxicity of Differently Charged Polystyrene Nanoparticles. RSC Adv. 2014, 4, 19321–19330. [Google Scholar] [CrossRef]
  123. Li, X.; Kang, B.; Eom, Y.; Zhong, J.; Lee, H.K.; Kim, H.M.; Song, J.S. Comparison of Cytotoxicity Effects Induced by Four Different Types of Nanoparticles in Human Corneal and Conjunctival Epithelial Cells. Sci. Rep. 2022, 12, 155. [Google Scholar] [CrossRef] [PubMed]
  124. Patrón-Romero, L.; Luque-Morales, P.A.; Loera-Castañeda, V.; Lares-Asseff, I.; Leal-Ávila, M.Á.; Alvelais-Palacios, J.A.; Plasencia-López, I.; Almanza-Reyes, H. Mitochondrial Dysfunction Induced by Zinc Oxide Nanoparticles. Crystals 2022, 12, 1089. [Google Scholar] [CrossRef]
  125. Chang, X.; Wang, X.; Li, J.; Shang, M.; Niu, S.; Zhang, W.; Li, Y.; Sun, Z.; Gan, J.; Li, W.; et al. Silver Nanoparticles Induced Cytotoxicity in HT22 Cells through Autophagy and Apoptosis via PI3K/AKT/MTOR Signaling Pathway. Ecotoxicol. Envrion. Saf. 2021, 208, 111696. [Google Scholar] [CrossRef]
  126. Yuan, X.; Nie, W.; He, Z.; Yang, J.; Shao, B.; Ma, X.; Zhang, X.; Bi, Z.; Sun, L.; Liang, X.; et al. Carbon Black Nanoparticles Induce Cell Necrosis through Lysosomal Membrane Permeabilization and Cause Subsequent Inflammatory Response. Theranostics 2020, 10, 4589–4605. [Google Scholar] [CrossRef]
  127. Fard, J.K.; Jafari, S.; Eghbal, M.A. A Review of Molecular Mechanisms Involved in Toxicity of Nanoparticles. Adv. Pharm. Bull. 2015, 5, 447–454. [Google Scholar] [CrossRef]
  128. Sahu, D.; Kannan, G.M.; Tailang, M.; Vijayaraghavan, R. In Vitro Cytotoxicity of Nanoparticles: A Comparison between Particle Size and Cell Type. J. Nanosci. 2016, 2016, 4023852. [Google Scholar] [CrossRef]
  129. Lanone, S.; Rogerieux, F.; Geys, J.; Dupont, A.; Maillot-Marechal, E.; Boczkowski, J.; Lacroix, G.; Hoet, P. Comparative Toxicity of 24 Manufactured Nanoparticles in Human Alveolar Epithelial and Macrophage Cell Lines. Part. Fibre Toxicol. 2009, 6, 14. [Google Scholar] [CrossRef]
  130. Zhang, X.F.; Shen, W.; Gurunathan, S. Silver Nanoparticle-Mediated Cellular Responses in Various Cell Lines: An in Vitro Model. Int. J. Mol. Sci. 2016, 17, 1603. [Google Scholar] [CrossRef] [PubMed]
  131. Popova, T.; Tzankov, B.; Voycheva, C.; Spassova, I.; Kovacheva, D.; Tzankov, S.; Aluani, D.; Tzankova, V.; Lambov, N. Mesoporous Silica MCM-41 and HMS as Advanced Drug Delivery Carriers for Bicalutamide. J. Drug Deliv. Sci. Technol. 2021, 62, 102340. [Google Scholar] [CrossRef]
  132. Heikkilä, T.; Santos, H.A.; Kumar, N.; Murzin, D.Y.; Salonen, J.; Laaksonen, T.; Peltonen, L.; Hirvonen, J.; Lehto, V.P. Cytotoxicity Study of Ordered Mesoporous Silica MCM-41 and SBA-15 Microparticles on Caco-2 Cells. Eur. J. Pharm. Biopharm. 2010, 74, 483–494. [Google Scholar] [CrossRef]
  133. Fuentes, C.; Ruiz-Rico, M.; Fuentes, A.; Barat, J.M.; Ruiz, M.J. Comparative Cytotoxic Study of Silica Materials Functionalised with Essential Oil Components in HepG2 Cells. Food Chem. Toxicol. 2021, 147, 111858. [Google Scholar] [CrossRef]
  134. Andrés, C.M.C.; Pérez de la Lastra, J.M.; Andrés Juan, C.; Plou, F.J.; Pérez-Lebeña, E. Superoxide Anion Chemistry—Its Role at the Core of the Innate Immunity. Int. J. Mol. Sci. 2023, 24, 1841. [Google Scholar] [CrossRef] [PubMed]
  135. Manke, A.; Wang, L.; Rojanasakul, Y. Mechanisms of Nanoparticle-Induced Oxidative Stress and Toxicity. Biomed. Res. Int. 2013, 2013, 94291. [Google Scholar] [CrossRef]
  136. Daré, R.G.; Lautenschlager, S.O.S. Nanoparticles with Antioxidant Activity. Antioxidants 2025, 14, 221. [Google Scholar] [CrossRef]
  137. Snezhkina, A.V.; Kudryavtseva, A.V.; Kardymon, O.L.; Savvateeva, M.V.; Melnikova, N.V.; Krasnov, G.S.; Dmitriev, A.A. ROS Generation and Antioxidant Defense Systems in Normal and Malignant Cells. Oxid. Med. Cell. Longev. 2020, 2019, 6175804. [Google Scholar] [CrossRef] [PubMed]
  138. Endale, H.T.; Tesfaye, W.; Mengstie, T.A. ROS Induced Lipid Peroxidation and Their Role in Ferroptosis. Front. Cell Dev. Biol. 2023, 11, 1226044. [Google Scholar] [CrossRef]
  139. Wang, B.; Wang, Y.; Zhang, J.; Hu, C.; Jiang, J.; Li, Y.; Peng, Z. ROS-Induced Lipid Peroxidation Modulates Cell Death Outcome: Mechanisms behind Apoptosis, Autophagy, and Ferroptosis. Arch. Toxicol. 2023, 97, 1439–1451. [Google Scholar] [CrossRef] [PubMed]
  140. Poetsch, A.R. The Genomics of Oxidative DNA Damage, Repair, and Resulting Mutagenesis. Comput. Struct. Biotechnol. J. 2020, 18, 207–219. [Google Scholar] [CrossRef] [PubMed]
  141. Sharma, V.; Collins, L.B.; Chen, T.; Herr, N.; Takeda, S.; Sun, W.; Swenberg, J.A.; Nakamura, J. Oxidative Stress at Low Levels Can Induce Clustered DNA Lesions Leading to NHEJ Mediated Mutations. Oncotarget 2016, 7, 25377–25390. [Google Scholar] [CrossRef]
  142. Yang, H.; Xie, Y.; Yang, D.; Ren, D. Oxidative Stress-Induced Apoptosis in Granulosa Cells Involves JNK, P53 and Puma. Oncotarget 2017, 8, 25310–25322. [Google Scholar] [CrossRef]
  143. Jomova, K.; Raptova, R.; Alomar, S.Y.; Alwasel, S.H.; Nepovimova, E.; Kuca, K.; Valko, M. Reactive Oxygen Species, Toxicity, Oxidative Stress, and Antioxidants: Chronic Diseases and Aging. Arch. Toxicol. 2023, 97, 2499–2574. [Google Scholar] [CrossRef]
  144. Dhanasekaran, D.N.; Reddy, E.P. JNK Signaling in Apoptosis. Oncogene 2008, 27, 6245–6251. [Google Scholar] [CrossRef]
  145. Martins, S.G.; Zilhão, R.; Thorsteinsdóttir, S.; Carlos, A.R. Linking Oxidative Stress and DNA Damage to Changes in the Expression of Extracellular Matrix Components. Front. Genet. 2021, 12, 673002. [Google Scholar] [CrossRef]
  146. Ighodaro, O.M.; Akinloye, O.A. First Line Defence Antioxidants-Superoxide Dismutase (SOD), Catalase (CAT) and Glutathione Peroxidase (GPX): Their Fundamental Role in the Entire Antioxidant Defence Grid. Alex. J. Med. 2018, 54, 287–293. [Google Scholar] [CrossRef]
  147. Radovanovic, J.; Banjac, K.; Obradovic, M.; Isenovic, E.R. Antioxidant Enzymes and Vascular Diseases. Explor. Med. 2021, 2, 544–555. [Google Scholar] [CrossRef]
  148. Sharma, P.; Jha, A.B.; Dubey, R.S.; Pessarakli, M. Reactive Oxygen Species, Oxidative Damage, and Antioxidative Defense Mechanism in Plants under Stressful Conditions. J. Bot. 2012, 2012, 217037. [Google Scholar] [CrossRef]
  149. Pei, J.; Pan, X.; Wei, G.; Hua, Y. Research Progress of Glutathione Peroxidase Family (GPX) in Redoxidation. Front. Pharmacol. 2023, 14, 1147414. [Google Scholar] [CrossRef]
  150. Ursini, F.; Maiorino, M. Lipid Peroxidation and Ferroptosis: The Role of GSH and GPx4. Free Radic. Biol. Med. 2020, 152, 175–185. [Google Scholar] [CrossRef]
  151. Dumanović, J.; Nepovimova, E.; Natić, M.; Kuča, K.; Jaćević, V. The Significance of Reactive Oxygen Species and Antioxidant Defense System in Plants: A Concise Overview. Front. Plant Sci. 2021, 11, 552969. [Google Scholar] [CrossRef] [PubMed]
  152. An, X.; Yu, W.; Liu, J.; Tang, D.; Yang, L.; Chen, X. Oxidative Cell Death in Cancer: Mechanisms and Therapeutic Opportunities. Cell Death Dis. 2024, 15, 556. [Google Scholar] [CrossRef]
  153. Lehman, S.E.; Morris, A.S.; Mueller, P.S.; Salem, A.K.; Grassian, V.H.; Larsen, S.C. Silica Nanoparticle-Generated ROS as a Predictor of Cellular Toxicity: Mechanistic Insights and Safety by Design. Envrion. Sci. Nano 2016, 3, 56–66. [Google Scholar] [CrossRef]
  154. Ahamed, M. Silica Nanoparticles-Induced Cytotoxicity, Oxidative Stress and Apoptosis in Cultured A431 and A549 Cells. Hum. Exp. Toxicol. 2013, 32, 186–195. [Google Scholar] [CrossRef]
  155. Duan, J.; Yu, Y.; Li, Y.; Yu, Y.; Li, Y.; Zhou, X.; Huang, P.; Sun, Z. Toxic Effect of Silica Nanoparticles on Endothelial Cells through DNA Damage Response via Chk1-Dependent G2/M Checkpoint. PLoS ONE 2013, 8, e62087. [Google Scholar] [CrossRef]
  156. Savi, M.; Bocchi, L.; Cacciani, F.; Vilella, R.; Buschini, A.; Perotti, A.; Galati, S.; Montalbano, S.; Pinelli, S.; Frati, C.; et al. Cobalt Oxide Nanoparticles Induce Oxidative Stress and Alter Electromechanical Function in Rat Ventricular Myocytes. Part. Fibre Toxicol. 2021, 18, 1. [Google Scholar] [CrossRef]
  157. Ghasemi, M.; Turnbull, T.; Sebastian, S.; Kempson, I. The MTT Assay: Utility, Limitations, Pitfalls, and Interpretation in Bulk and Single-Cell Analysis. Int. J. Mol. Sci. 2021, 22, 2827. [Google Scholar] [CrossRef] [PubMed]
  158. Apopa, P.L.; Qian, Y.; Shao, R.; Guo, N.L.; Schwegler-Berry, D.; Pacurari, M.; Porter, D.; Shi, X.; Vallyathan, V.; Castranova, V.; et al. Iron Oxide Nanoparticles Induce Human Microvascular Endothelial Cell Permeability through Reactive Oxygen Species Production and Microtubule Remodeling. Part. Fibre Toxicol. 2009, 6, 1. [Google Scholar] [CrossRef] [PubMed]
  159. Zhang, L.; Feng, G.; Yang, S.; Liu, B.; Niu, Y.; Fan, P.; Liu, Z.; Chen, J.; Cui, L.; Zhou, G.; et al. Polyethylenimine-Modified Mesoporous Silica Nanoparticles Induce a Survival Mechanism in Vascular Endothelial Cells via Microvesicle-Mediated Autophagosome Release. ACS Nano 2021, 15, 10640–10658. [Google Scholar] [CrossRef] [PubMed]
  160. Fröhlich, E.; Samberger, C.; Kueznik, T.; Absenger, M.; Roblegg, E.; Zimmer, A.; Pieber, T.R. Cytotoxicity of Nanoparticles Independent from Oxidative Stress. J. Toxicol. Sci. 2001, 34, 363–375. [Google Scholar] [CrossRef]
  161. Kohl, Y.; Rundén-Pran, E.; Mariussen, E.; Hesler, M.; El Yamani, N.; Longhin, E.M.; Dusinska, M. Genotoxicity of Nanomaterials: Advanced in Vitro Models and High Throughput Methods for Human Hazard Assessment—A Review. Nanomaterials 2020, 10, 1911. [Google Scholar] [CrossRef]
  162. Sicińska, P.; Mokra, K.; Wozniak, K.; Michałowicz, J.; Bukowska, B. Genotoxic Risk Assessment and Mechanism of DNA Damage Induced by Phthalates and Their Metabolites in Human Peripheral Blood Mononuclear Cells. Sci. Rep. 2021, 11, 1658. [Google Scholar] [CrossRef]
  163. Elespuru, R.K.; Doak, S.H.; Collins, A.R.; Dusinska, M.; Pfuhler, S.; Manjanatha, M.; Cardoso, R.; Chen, C.L. Common Considerations for Genotoxicity Assessment of Nanomaterials. Front. Toxicol. 2022, 4, 859122. [Google Scholar] [CrossRef]
  164. Menz, J.; Götz, M.E.; Gündel, U.; Gürtler, R.; Herrmann, K.; Hessel-Pras, S.; Kneuer, C.; Kolrep, F.; Nitzsche, D.; Pabel, U.; et al. Genotoxicity Assessment: Opportunities, Challenges and Perspectives for Quantitative Evaluations of Dose–Response Data. Arch. Toxicol. 2023, 97, 2303–2328. [Google Scholar] [CrossRef]
  165. Landsiedel, R.; Honarvar, N.; Seiffert, S.B.; Oesch, B.; Oesch, F. Genotoxicity Testing of Nanomaterials. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2022, 14, e1833. [Google Scholar] [CrossRef]
  166. Solorio-Rodriguez, S.A.; Wu, D.; Boyadzhiev, A.; Christ, C.; Williams, A.; Halappanavar, S. A Systematic Genotoxicity Assessment of a Suite of Metal Oxide Nanoparticles Reveals Their DNA Damaging and Clastogenic Potential. Nanomaterials 2024, 14, 743. [Google Scholar] [CrossRef]
  167. Lebedová, J.; Hedberg, Y.S.; Odnevall Wallinder, I.; Karlsson, H.L. Size-Dependent Genotoxicity of Silver, Gold and Platinum Nanoparticles Studied Using the Mini-Gel Comet Assay and Micronucleus Scoring with Flow Cytometry. Mutagenesis 2018, 33, 77–85. [Google Scholar] [CrossRef]
  168. Xia, Q.; Li, H.; Liu, Y.; Zhang, S.; Feng, Q.; Xiao, K. The Effect of Particle Size on the Genotoxicity of Gold Nanoparticles. J. Biomed. Mater. Res. A 2017, 105, 710–719. [Google Scholar] [CrossRef]
  169. Yazdimamaghani, M.; Moos, P.J.; Dobrovolskaia, M.A.; Ghandehari, H. Genotoxicity of Amorphous Silica Nanoparticles: Status and Prospects. Nanomedicine 2019, 16, 106–125. [Google Scholar] [CrossRef] [PubMed]
  170. Park, M.V.D.Z.; Verharen, H.W.; Zwart, E.; Hernandez, L.G.; van Benthem, J.; Elsaesser, A.; Barnes, C.; McKerr, G.; Howard, C.V.; Salvati, A.; et al. Genotoxicity Evaluation of Amorphous Silica Nanoparticles of Different Sizes Using the Micronucleus and the Plasmid LacZ Gene Mutation Assay. Nanotoxicology 2011, 5, 168–181. [Google Scholar] [CrossRef] [PubMed]
  171. Wang, F.; Chen, Y.; Wang, Y.; Yin, Y.; Qu, G.; Song, M.; Wang, H. Ultra-Long Silver Nanowires Induced Mitotic Abnormalities and Cytokinetic Failure in A549 Cells. Nanotoxicology 2019, 13, 543–557. [Google Scholar] [CrossRef] [PubMed]
  172. García-Rodríguez, A.; Vila, L.; Cortés, C.; Hernández, A.; Marcos, R. Effects of Differently Shaped TiO2NPs (Nanospheres, Nanorods and Nanowires) on the in Vitro Model (Caco-2/HT29) of the Intestinal Barrier. Part. Fibre Toxicol. 2018, 15, 33. [Google Scholar] [CrossRef]
  173. Encinas-Gimenez, M.; Martin-Duque, P.; Martín-Pardillos, A. Cellular Alterations Due to Direct and Indirect Interaction of Nanomaterials with Nucleic Acids. Int. J. Mol. Sci. 2024, 25, 1983. [Google Scholar] [CrossRef]
  174. Green, J.J. 2011 Rita Schaffer Lecture: Nanoparticles for Intracellular Nucleic Acid Delivery. Ann. Biomed. Eng. 2012, 40, 1408–1418. [Google Scholar] [CrossRef]
  175. Schaeublin, N.M.; Braydich-Stolle, L.K.; Schrand, A.M.; Miller, J.M.; Hutchison, J.; Schlager, J.J.; Hussain, S.M. Surface Charge of Gold Nanoparticles Mediates Mechanism of Toxicity. Nanoscale 2011, 3, 410. [Google Scholar] [CrossRef]
  176. Platel, A.; Carpentier, R.; Becart, E.; Mordacq, G.; Betbeder, D.; Nesslany, F. Influence of the Surface Charge of PLGA Nanoparticles on Their in Vitro Genotoxicity, Cytotoxicity, ROS Production and Endocytosis. J. Appl. Toxicol. 2016, 36, 434–444. [Google Scholar] [CrossRef]
  177. Fresegna, A.M.; Ursini, C.L.; Ciervo, A.; Maiello, R.; Casciardi, S.; Iavicoli, S.; Cavallo, D. Assessment of the Influence of Crystalline Form on Cyto-Genotoxic and Inflammatory Effects Induced by Tio2 Nanoparticles on Human Bronchial and Alveolar Cells. Nanomaterials 2021, 11, 253. [Google Scholar] [CrossRef]
  178. Rim, K.T.; Song, S.W.; Kim, H.Y. Oxidative DNA Damage from Nanoparticle Exposure and Its Application to Workers’ Health: A Literature Review. Saf. Health Work 2013, 4, 177–186. [Google Scholar] [CrossRef]
  179. Carrière, M.; Arnal, M.-E.; Douki, T. TiO2 Genotoxicity: An Update of the Results Published over the Last Six Years. Mutat. Res./Genet. Toxicol. Environ. Mutagen. 2020, 854–855, 503198. [Google Scholar] [CrossRef] [PubMed]
  180. Demir, E.; Castranova, V. Genotoxic Effects of Synthetic Amorphous Silica Nanoparticles in the Mouse Lymphoma Assay. Toxicol. Rep. 2016, 3, 807–815. [Google Scholar] [CrossRef] [PubMed]
  181. Schardosim, R.F.D.C.; Cardozo, T.R.; de Souza, A.P.; Seeber, A.; Flores, W.H.; Lehmann, M.; Dihl, R.R. Cyto—Genotoxicity of Crystalline and Amorphous Niobium (V) Oxide Nanoparticles in CHO-K1 Cells. Toxicol. Res. 2022, 11, 765–773. [Google Scholar] [CrossRef] [PubMed]
  182. Vecchiotti, G.; Colafarina, S.; Aloisi, M.; Zarivi, O.; Di Carlo, P.; Poma, A. Genotoxicity and Oxidative Stress Induction by Polystyrene Nanoparticles in the Colorectal Cancer Cell Line HCT116. PLoS ONE 2021, 16, e0255120. [Google Scholar] [CrossRef]
  183. Rinna, A.; Magdolenova, Z.; Hudecova, A.; Kruszewski, M.; Refsnes, M.; Dusinska, M. Effect of Silver Nanoparticles on Mitogen-Activated Protein Kinases Activation: Role of Reactive Oxygen Species and Implication in DNA Damage. Mutagenesis 2015, 30, 59–66. [Google Scholar] [CrossRef]
  184. Shukla, R.K.; Badiye, A.; Vajpayee, K.; Kapoor, N. Genotoxic Potential of Nanoparticles: Structural and Functional Modifications in DNA. Front. Genet. 2021, 12, 728250. [Google Scholar] [CrossRef]
  185. Choudante, P.C.; Nethi, S.K.; Díaz-García, D.; Prashar, S.; Misra, S.; Gómez-Ruiz, S.; Patra, C.R. Tin-Loaded Mesoporous Silica Nanoparticles: Antineoplastic Properties and Genotoxicity Assessment. Biomater. Adv. 2022, 137, 212819. [Google Scholar] [CrossRef]
  186. Ansari, M.O.; Parveen, N.; Ahmad, M.F.; Wani, A.L.; Afrin, S.; Rahman, Y.; Jameel, S.; Khan, Y.A.; Siddique, H.R.; Tabish, M.; et al. Evaluation of DNA Interaction, Genotoxicity and Oxidative Stress Induced by Iron Oxide Nanoparticles Both in Vitro and in Vivo: Attenuation by Thymoquinone. Sci. Rep. 2019, 9, 6912. [Google Scholar] [CrossRef]
  187. Babonaitė, M.; Striogaitė, E.; Grigorianaitė, G.; Lazutka, J.R. In Vitro Evaluation of DNA Damage Induction by Silver (Ag), Gold (Au), Silica (SiO2), and Aluminum Oxide (Al2O3) Nanoparticles in Human Peripheral Blood Mononuclear Cells. Curr. Issues Mol. Biol. 2024, 46, 6986–7000. [Google Scholar] [CrossRef]
  188. Zhang, J.; Liu, J.; Ren, L.; Wei, J.; Zhang, F.; Li, Y.; Guo, C.; Duan, J.; Sun, Z.; Zhou, X. Silica Nanoparticles Induce Abnormal Mitosis and Apoptosis via PKC-Δ Mediated Negative Signaling Pathway in GC-2 cells of Mice. Chemosphere 2018, 208, 942–950. [Google Scholar] [CrossRef]
  189. George, J.M.; Magogotya, M.; Vetten, M.A.; Buys, A.V.; Gulumian, M. An Investigation of the Genotoxicity and Interference of Gold Nanoparticles in Commonly Used in Vitro Mutagenicity and Genotoxicity Assays. Toxicol. Sci. 2017, 156, 149–166. [Google Scholar] [CrossRef]
  190. Zhang, W.; Cho, W.C.; Bloukh, S.H.; Edis, Z.; Du, W.; He, Y.; Hu, H.Y.; ten Hagen, T.L.M.; Falahati, M. An Overview on the Exploring the Interaction of Inorganic Nanoparticles with Microtubules for the Advancement of Cancer Therapeutics. Int. J. Biol. Macromol. 2022, 212, 358–369. [Google Scholar] [CrossRef]
  191. Guan, L.; Chen, J.; Tian, Z.; Zhu, M.; Bian, Y.; Zhu, Y. Mesoporous Organosilica Nanoparticles: Degradation Strategies and Application in Tumor Therapy. View 2021, 2, 20200117. [Google Scholar] [CrossRef]
  192. Sargent, L.M.; Hubbs, A.F.; Young, S.H.; Kashon, M.L.; Dinu, C.Z.; Salisbury, J.L.; Benkovic, S.A.; Lowry, D.T.; Murray, A.R.; Kisin, E.R.; et al. Single-Walled Carbon Nanotube-Induced Mitotic Disruption. Mutat. Res. Genet. Toxicol. Envrion. Mutagen. 2012, 745, 28–37. [Google Scholar] [CrossRef]
  193. Xu, X.; Xu, S.; Wan, J.; Wang, D.; Pang, X.; Gao, Y.; Ni, N.; Chen, D.; Sun, X. Disturbing Cytoskeleton by Engineered Nanomaterials for Enhanced Cancer Therapeutics. Bioact. Mater. 2023, 29, 50–71. [Google Scholar] [CrossRef]
  194. Bannister, A.; Dissanayake, D.; Kowalewski, A.; Cicon, L.; Bromma, K.; Chithrani, D.B. Modulation of the Microtubule Network for Optimization of Nanoparticle Dynamics for the Advancement of Cancer Nanomedicine. Bioengineering 2020, 7, 56. [Google Scholar] [CrossRef]
  195. Doak, S.H.; Andreoli, C.; Burgum, M.J.; Chaudhry, Q.; Bleeker, E.A.J.; Bossa, C.; Domenech, J.; Drobne, D.; Fessard, V.; Jeliazkova, N.; et al. Current Status and Future Challenges of Genotoxicity OECD Test Guidelines for Nanomaterials: A Workshop Report. Mutagenesis 2023, 38, 183–191. [Google Scholar] [CrossRef]
  196. Senapati, V.A.; Kumar, A.; Gupta, G.S.; Pandey, A.K.; Dhawan, A. ZnO Nanoparticles Induced Inflammatory Response and Genotoxicity in Human Blood Cells: A Mechanistic Approach. Food Chem. Toxicol. 2015, 85, 61–70. [Google Scholar] [CrossRef]
  197. Nakad, R.; Schumacher, B. DNA Damage Response and Immune Defense: Links and Mechanisms. Front. Genet. 2016, 7, 147. [Google Scholar] [CrossRef]
  198. Lin, R.; Zhang, C.; Zheng, J.; Tian, D.; Lei, Z.; Chen, D.; Xu, Z.; Su, M. Chronic Inflammation-Associated Genomic Instability Paves the Way for Human Esophageal Carcinogenesis. Oncotarget 2016, 7, 24564–24571. [Google Scholar] [CrossRef]
  199. Bourdon, J.A.; Williams, A.; Kuo, B.; Moffat, I.; White, P.A.; Halappanavar, S.; Vogel, U.; Wallin, H.; Yauk, C.L. Gene Expression Profiling to Identify Potentially Relevant Disease Outcomes and Support Human Health Risk Assessment for Carbon Black Nanoparticle Exposure. Toxicology 2013, 303, 83–93. [Google Scholar] [CrossRef] [PubMed]
  200. Aubrey, B.J.; Kelly, G.L.; Janic, A.; Herold, M.J.; Strasser, A. How Does P53 Induce Apoptosis and How Does This Relate to P53-Mediated Tumour Suppression? Cell. Death Differ. 2018, 25, 104–113. [Google Scholar] [CrossRef]
  201. Chen, J. The Cell-Cycle Arrest and Apoptotic Functions of P53 in Tumor Initiation and Progression. Cold Spring Harb. Perspect. Med. 2016, 6, a026104. [Google Scholar] [CrossRef]
  202. Abuetabh, Y.; Wu, H.H.; Chai, C.; Al Yousef, H.; Persad, S.; Sergi, C.M.; Leng, R. DNA Damage Response Revisited: The P53 Family and Its Regulators Provide Endless Cancer Therapy Opportunities. Exp. Mol. Med. 2022, 54, 1658–1669. [Google Scholar] [CrossRef]
  203. Okazaki, R. Role of P53 in Regulating Radiation Responses. Life 2022, 12, 1099. [Google Scholar] [CrossRef]
  204. Wang, H.; Guo, M.; Wei, H.; Chen, Y. Targeting P53 Pathways: Mechanisms, Structures, and Advances in Therapy. Signal Transduct. Target. Ther. 2023, 8, 92. [Google Scholar] [CrossRef]
  205. Barr, A.R.; Cooper, S.; Heldt, F.S.; Butera, F.; Stoy, H.; Mansfeld, J.; Novák, B.; Bakal, C. DNA Damage during S-Phase Mediates the Proliferation-Quiescence Decision in the Subsequent G1 via P21 Expression. Nat. Commun. 2017, 8, 14728. [Google Scholar] [CrossRef]
  206. Engeland, K. Cell Cycle Arrest through Indirect Transcriptional Repression by P53: I Have a DREAM. Cell Death Differ. 2018, 25, 114–132. [Google Scholar] [CrossRef]
  207. Bowen, M.E.; Mulligan, A.S.; Sorayya, A.; Attardi, L.D. Puma- and Caspase9-Mediated Apoptosis Is Dispensable for P53-Driven Neural Crest-Based Developmental Defects. Cell Death Differ. 2021, 28, 2083–2094. [Google Scholar] [CrossRef]
  208. Hao, Q.; Chen, J.; Lu, H.; Zhou, X. The ARTS of P53-Dependent Mitochondrial Apoptosis. J. Mol. Cell. Biol. 2022, 14, mjac074. [Google Scholar] [CrossRef]
  209. Leong, W.F.; Chau, J.F.L.; Li, B. P53 Deficiency Leads to Compensatory Up-Regulation of P16INK4a. Mol. Cancer Res. 2009, 7, 354–360. [Google Scholar] [CrossRef] [PubMed]
  210. Qian, Y.; Chen, X. Senescence Regulation by the P53 Protein Family. Methods Mol. Biol. 2013, 965, 37–61. [Google Scholar] [CrossRef] [PubMed]
  211. Szewczyk-Roszczenko, O.; Barlev, N.A. The Role of P53 in Nanoparticle-Based Therapy for Cancer. Cells 2023, 12, 2803. [Google Scholar] [CrossRef] [PubMed]
  212. Wierzbicki, M.; Sawosz, E.; Strojny, B.; Jaworski, S.; Grodzik, M.; Chwalibog, A. NF-ΚB-Related Decrease of Glioma Angiogenic Potential by Graphite Nanoparticles and Graphene Oxide Nanoplatelets. Sci. Rep. 2018, 8, 14733. [Google Scholar] [CrossRef]
  213. Alswady-hoff, M.; Erdem, J.S.; Phuyal, S.; Knittelfelder, O.; Sharma, A.; Fonseca, D.d.M.; Skare, Ø.; Slupphaug, G.; Zienolddiny, S. Long-term Exposure to Nanosized Tio2 Triggers Stress Responses and Cell Death Pathways in Pulmonary Epithelial Cells. Int. J. Mol. Sci. 2021, 22, 5349. [Google Scholar] [CrossRef]
  214. Tavakoli, S.; Motavalizadehkakhky, A.; Homayouni Tabrizi, M.; Mehrzad, J.; Zhiani, R. Study of the Anti-Cancer Activity of a Mesoporous Silica Nanoparticle Surface Coated with Polydopamine Loaded with Umbelliprenin. Sci. Rep. 2024, 14, 11450. [Google Scholar] [CrossRef]
  215. Chen, J.; Stark, L.A. Insights into the Relationship between Nucleolar Stress and the NF-ΚB Pathway. Trends Genet. 2019, 35, 768–780. [Google Scholar] [CrossRef]
  216. Chen, J.; Lobb, I.T.; Morin, P.; Novo, S.M.; Simpson, J.; Kennerknecht, K.; Von Kriegsheim, A.; Batchelor, E.E.; Oakley, F.; Stark, L.A. Identification of a Novel TIF-IA-NF-ΚB Nucleolar Stress Response Pathway. Nucleic Acids Res. 2018, 46, 6188–6205. [Google Scholar] [CrossRef]
  217. Lowe, J.M.; Menendez, D.; Bushel, P.R.; Shatz, M.; Kirk, E.L.; Troester, M.A.; Garantziotis, S.; Fessler, M.B.; Resnick, M.A. P53 and NF-ΚB Coregulate Proinflammatory Gene Responses in Human Macrophages. Cancer Res. 2014, 74, 2182–2192. [Google Scholar] [CrossRef]
  218. Carrà, G.; Lingua, M.F.; Maffeo, B.; Taulli, R.; Morotti, A. P53 vs NF-ΚB: The Role of Nuclear Factor-Kappa B in the Regulation of P53 Activity and Vice Versa. Cell. Mol. Life Sci. 2020, 77, 4449–4458. [Google Scholar] [CrossRef]
  219. Konrath, F.; Mittermeier, A.; Cristiano, E.; Wolf, J.; Loewer, A. A Systematic Approach to Decipher Crosstalk in the P53 Signaling Pathway Using Single Cell Dynamics. PLoS Comput. Biol. 2020, 16, e1007901. [Google Scholar] [CrossRef]
  220. Milani, D.; Caruso, L.; Zauli, E.; Al Owaifeer, A.M.; Secchiero, P.; Zauli, G.; Gemmati, D.; Tisato, V. P53/NF-KB Balance in SARS-CoV-2 Infection: From OMICs, Genomics and Pharmacogenomics Insights to Tailored Therapeutic Perspectives (COVIDomics). Front. Pharmacol. 2022, 13, 871583. [Google Scholar] [CrossRef]
  221. Liu, X.; Sun, J. Endothelial Cells Dysfunction Induced by Silica Nanoparticles through Oxidative Stress via JNK/P53 and NF-ΚB Pathways. Biomaterials 2010, 31, 8198–8209. [Google Scholar] [CrossRef]
  222. Hanson, R.L.; Batchelor, E. Coordination of MAPK and P53 Dynamics in the Cellular Responses to DNA Damage and Oxidative Stress. Mol. Syst. Biol. 2022, 18, e11401. [Google Scholar] [CrossRef] [PubMed]
  223. De, S.; Campbell, C.; Venkitaraman, A.R.; Esposito, A. Pulsatile MAPK Signaling Modulates P53 Activity to Control Cell Fate Decisions at the G2 Checkpoint for DNA Damage. Cell Rep. 2020, 30, 2083–2093.e5. [Google Scholar] [CrossRef] [PubMed]
  224. Lafarga, V.; Cuadrado, A.; Lopez de Silanes, I.; Bengoechea, R.; Fernandez-Capetillo, O.; Nebreda, A.R. P38 Mitogen-Activated Protein Kinase- and HuR-Dependent Stabilization of P21 Cip1 MRNA Mediates the G 1/S Checkpoint. Mol. Cell Biol. 2009, 29, 4341–4351. [Google Scholar] [CrossRef]
  225. Sugiura, R.; Satoh, R.; Takasaki, T. ERK: A Double-Edged Sword in Cancer. ERK-Dependent Apoptosis as a Potential Therapeutic Strategy for Cancer. Cells 2021, 10, 2509. [Google Scholar] [CrossRef]
  226. Deschênes-Simard, X.; Gaumont-Leclerc, M.F.; Bourdeau, V.; Lessard, F.; Moiseeva, O.; Forest, V.; Igelmann, S.; Mallette, F.A.; Saba-El-Leil, M.K.; Meloche, S.; et al. Tumor Suppressor Activity of the ERK/MAPK Pathway by Promoting Selective Protein Degradation. Genes. Dev. 2013, 27, 900–915. [Google Scholar] [CrossRef]
  227. Zuo, J.; Ma, S. Resveratrol-Laden Mesoporous Silica Nanoparticles Regulate the Autophagy and Apoptosis via ROS-Mediated P38-MAPK/HIF-1a/P53 Signaling in Hypertrophic Scar Fibroblasts. Heliyon 2024, 10, e24985. [Google Scholar] [CrossRef]
  228. Zhao, X.; Wu, Y.; Li, J.; Li, D.; Jin, Y.; Zhu, P.; Liu, Y.; Zhuang, Y.; Yu, S.; Cao, W.; et al. JNK Activation-Mediated Nuclear SIRT1 Protein Suppression Contributes to Silica Nanoparticle-Induced Pulmonary Damage via P53 Acetylation and Cytoplasmic Localisation. Toxicology 2019, 423, 42–53. [Google Scholar] [CrossRef]
  229. Rahman, M.A.; Park, M.N.; Rahman, M.H.; Rashid, M.M.; Islam, R.; Uddin, M.J.; Hannan, M.A.; Kim, B. P53 Modulation of Autophagy Signaling in Cancer Therapies: Perspectives Mechanism and Therapeutic Targets. Front. Cell Dev. Biol. 2022, 10, 761080. [Google Scholar] [CrossRef] [PubMed]
  230. Feroz, W.; Sheikh, A.M.A. Exploring the Multiple Roles of Guardian of the Genome: P53. Egypt. J. Med. Hum. Genet. 2020, 21, 49. [Google Scholar] [CrossRef]
  231. Shim, D.; Duan, L.; Maki, C.G. P53-Regulated Autophagy and Its Impact on Drug Resistance and Cell Fate. Cancer Drug Resistance 2021, 4, 85–95. [Google Scholar] [CrossRef]
  232. Shi, Y.; Norberg, E.; Vakifahmetoglu-Norberg, H. Mutant P53 as a Regulator and Target of Autophagy. Front. Oncol. 2021, 10, 607149. [Google Scholar] [CrossRef] [PubMed]
  233. Zhu, Y.; Zuo, W.; Shen, X.; Liu, Y.; Zhao, Y.; Xiong, Y.; Cao, H.; Wang, Y.; Liang, Z. NF-ΚB Is Involved in the Regulation of Autophagy in Mutant P53 Cells in Response to Ionizing Radiation. Cell Death Discov. 2021, 7, 159. [Google Scholar] [CrossRef]
  234. Verzella, D.; Pescatore, A.; Capece, D.; Vecchiotti, D.; Ursini, M.V.; Franzoso, G.; Alesse, E.; Zazzeroni, F. Life, Death, and Autophagy in Cancer: NF-ΚB Turns up Everywhere. Cell Death Dis. 2020, 11, 210. [Google Scholar] [CrossRef]
  235. Rahman, M.A.; Jalouli, M.; Bhajan, S.K.; Al-Zharani, M.; Harrath, A.H. A Comprehensive Review of Nanoparticle-Based Drug Delivery for Modulating PI3K/AKT/MTOR-Mediated Autophagy in Cancer. Int. J. Mol. Sci. 2025, 26, 1868. [Google Scholar] [CrossRef]
  236. Coronel, L.; Häckes, D.; Schwab, K.; Riege, K.; Hoffmann, S.; Fischer, M. P53-Mediated AKT and MTOR Inhibition Requires RFX7 and DDIT4 and Depends on Nutrient Abundance. Oncogene 2022, 41, 1063–1069. [Google Scholar] [CrossRef]
  237. Tavakol, S.; Ashrafizadeh, M.; Deng, S.; Azarian, M.; Abdoli, A.; Motavaf, M.; Poormoghadam, D.; Khanbabaei, H.; Ghasemipour Afshar, E.; Mandegary, A.; et al. Autophagy Modulators: Mechanistic Aspects and Drug Delivery Systems. Biomolecules 2019, 9, 530. [Google Scholar] [CrossRef] [PubMed]
  238. Song, J.; Meng, H.; Deng, G.; Lin, H. Sustainable Release Selenium Laden with SiO2 Restoring Peripheral Nerve Injury via Modulating PI3K/AKT Pathway Signaling Pathway. Int. J. Nanomed. 2024, 19, 7851–7870. [Google Scholar] [CrossRef]
  239. Yang, B.Y.; Zhou, Z.Y.; Liu, S.Y.; Shi, M.J.; Liu, X.J.; Cheng, T.M.; Deng, G.Y.; Tian, Y.; Song, J.; Li, X.H. Porous Se@SiO2 Nanoparticles Enhance Wound Healing by ROS-PI3K/Akt Pathway in Dermal Fibroblasts and Reduce Scar Formation. Front. Bioeng. Biotechnol. 2022, 10, 852482. [Google Scholar] [CrossRef]
  240. Liao, X.; Liu, Y.; Zheng, J.; Zhao, X.; Cui, L.; Hu, S.; Xia, T.; Si, S. Diverse Pathways of Engineered Nanoparticle-Induced NLRP3 Inflammasome Activation. Nanomaterials 2022, 12, 3908. [Google Scholar] [CrossRef]
  241. Rampado, R.; Crotti, S.; Caliceti, P.; Pucciarelli, S.; Agostini, M. Recent Advances in Understanding the Protein Corona of Nanoparticles and in the Formulation of “Stealthy” Nanomaterials. Front. Bioeng. Biotechnol. 2020, 8, 166. [Google Scholar] [CrossRef]
  242. Akhter, M.H.; Khalilullah, H.; Gupta, M.; Alfaleh, M.A.; Alhakamy, N.A.; Riadi, Y.; Md, S. Impact of Protein Corona on the Biological Identity of Nanomedicine: Understanding the Fate of Nanomaterials in the Biological Milieu. Biomedicines 2021, 9, 1496. [Google Scholar] [CrossRef]
  243. Bai, X.; Wang, J.; Mu, Q.; Su, G. In Vivo Protein Corona Formation: Characterizations, Effects on Engineered Nanoparticles’ Biobehaviors, and Applications. Front. Bioeng. Biotechnol. 2021, 9, 646708. [Google Scholar] [CrossRef]
  244. Baimanov, D.; Wang, J.; Zhang, J.; Liu, K.; Cong, Y.; Shi, X.; Zhang, X.; Li, Y.; Li, X.; Qiao, R.; et al. In Situ Analysis of Nanoparticle Soft Corona and Dynamic Evolution. Nat. Commun. 2022, 13, 5389. [Google Scholar] [CrossRef]
  245. Bashiri, G.; Padilla, M.S.; Swingle, K.L.; Shepherd, S.J.; Mitchell, M.J.; Wang, K. Nanoparticle Protein Corona: From Structure and Function to Therapeutic Targeting. Lab Chip 2023, 23, 1432–1466. [Google Scholar] [CrossRef] [PubMed]
  246. García-álvarez, R.; Vallet-Regí, M. Hard and Soft Protein Corona of Nanomaterials: Analysis and Relevance. Nanomaterials 2021, 11, 888. [Google Scholar] [CrossRef]
  247. Vilanova, O.; Martinez-Serra, A.; Monopoli, M.P.; Franzese, G. Characterizing the Hard and Soft Nanoparticle-Protein Corona with Multilayer Adsorption. Front. Nanotechnol. 2025, 6, 1531039. [Google Scholar] [CrossRef]
  248. Mohammad-Beigi, H.; Hayashi, Y.; Zeuthen, C.M.; Eskandari, H.; Scavenius, C.; Juul-Madsen, K.; Vorup-Jensen, T.; Enghild, J.J.; Sutherland, D.S. Mapping and Identification of Soft Corona Proteins at Nanoparticles and Their Impact on Cellular Association. Nat. Commun. 2020, 11, 4535. [Google Scholar] [CrossRef] [PubMed]
  249. Marichal, L.; Klein, G.; Armengaud, J.; Boulard, Y.; Chédin, S.; Labarre, J.; Pin, S.; Renault, J.P.; Aude, J.C. Protein Corona Composition of Silica Nanoparticles in Complex Media: Nanoparticle Size Does Not Matter. Nanomaterials 2020, 10, 240. [Google Scholar] [CrossRef]
  250. Xu, W.; Xu, M.; Xiao, Y.; Yu, L.; Xie, H.; Jiang, X.; Chen, M.; Gao, H.; Wang, L. Changes in Target Ability of Nanoparticles Due to Protein Corona Composition and Disease State. Asian J. Pharm. Sci. 2022, 17, 401–411. [Google Scholar] [CrossRef]
  251. Nierenberg, D.; Khaled, A.R.; Flores, O. Formation of a Protein Corona Influences the Biological Identity of Nanomaterials. Rep. Pract. Oncol. Radiother. 2018, 23, 300–308. [Google Scholar] [CrossRef]
  252. Salvati, A.; Pitek, A.S.; Monopoli, M.P.; Prapainop, K.; Bombelli, F.B.; Hristov, D.R.; Kelly, P.M.; Åberg, C.; Mahon, E.; Dawson, K.A. Transferrin-Functionalized Nanoparticles Lose Their Targeting Capabilities When a Biomolecule Corona Adsorbs on the Surface. Nat. Nanotechnol. 2013, 8, 137–143. [Google Scholar] [CrossRef] [PubMed]
  253. Bertrand, N.; Grenier, P.; Mahmoudi, M.; Lima, E.M.; Appel, E.A.; Dormont, F.; Lim, J.M.; Karnik, R.; Langer, R.; Farokhzad, O.C. Mechanistic Understanding of in Vivo Protein Corona Formation on Polymeric Nanoparticles and Impact on Pharmacokinetics. Nat. Commun. 2017, 8, 777. [Google Scholar] [CrossRef]
  254. Palmieri, V.; Caracciolo, G. Tuning the Immune System by Nanoparticle-Biomolecular Corona. Nanoscale Adv. 2022, 4, 3300–3308. [Google Scholar] [CrossRef]
  255. Miao, Y.; Li, L.; Wang, Y.; Wang, J.; Zhou, Y.; Guo, L.; Zhao, Y.; Nie, D.; Zhang, Y.; Zhang, X.; et al. Regulating Protein Corona on Nanovesicles by Glycosylated Polyhydroxy Polymer Modification for Efficient Drug Delivery. Nat. Commun. 2024, 15, 1159. [Google Scholar] [CrossRef]
  256. Lee, Y.K.; Choi, E.J.; Webster, T.J.; Kim, S.H.; Khang, D. Effect of the Protein Corona on Nanoparticles for Modulating Cytotoxicity and Immunotoxicity. Int. J. Nanomed. 2014, 10, 97–113. [Google Scholar] [CrossRef]
  257. Lima, T.; Bernfur, K.; Vilanova, M.; Cedervall, T. Understanding the Lipid and Protein Corona Formation on Different Sized Polymeric Nanoparticles. Sci. Rep. 2020, 10, 1129. [Google Scholar] [CrossRef]
  258. Panico, S.; Capolla, S.; Bozzer, S.; Toffoli, G.; Dal Bo, M.; Macor, P. Biological Features of Nanoparticles: Protein Corona Formation and Interaction with the Immune System. Pharmaceutics 2022, 14, 2605. [Google Scholar] [CrossRef]
  259. Yang, C.; Zhao, H.; Sun, Y.; Wang, C.; Geng, X.; Wang, R.; Tang, L.; Han, D.; Liu, J.; Tan, W. Programmable Manipulation of Oligonucleotide-Albumin Interaction for Elongated Circulation Time. Nucleic Acids Res. 2022, 50, 3083–3095. [Google Scholar] [CrossRef] [PubMed]
  260. Chung, H.; Park, J.Y.; Kim, K.; Yoo, R.J.; Suh, M.; Gu, G.J.; Kim, J.S.; Choi, T.H.; Byun, J.W.; Ju, Y.W.; et al. Circulation Time-Optimized Albumin Nanoplatform for Quantitative Visualization of Lung Metastasis via Targeting of Macrophages. ACS Nano 2022, 16, 12262–12275. [Google Scholar] [CrossRef]
  261. Das, A.; Chakrabarti, A.; Das, P.K. Suppression of Protein Aggregation by Gold Nanoparticles: A New Way to Store and Transport Proteins. RSC Adv. 2015, 5, 38558–38570. [Google Scholar] [CrossRef]
  262. Chen, F.; Wang, G.; Griffin, J.I.; Brenneman, B.; Banda, N.K.; Holers, V.M.; Backos, D.S.; Wu, L.; Moghimi, S.M.; Simberg, D. Complement Proteins Bind to Nanoparticle Protein Corona and Undergo Dynamic Exchange in Vivo. Nat. Nanotechnol. 2017, 12, 387–393. [Google Scholar] [CrossRef]
  263. Strojan, K.; Leonardi, A.; Bregar, V.B.; Križaj, I.; Svete, J.; Pavlin, M. Dispersion of Nanoparticles in Different Media Importantly Determines the Composition of Their Protein Corona. PLoS ONE 2017, 12, e0169552. [Google Scholar] [CrossRef] [PubMed]
  264. Derakhshankhah, H.; Hosseini, A.; Taghavi, F.; Jafari, S.; Lotfabadi, A.; Ejtehadi, M.R.; Shahbazi, S.; Fattahi, A.; Ghasemi, A.; Barzegari, E.; et al. Molecular Interaction of Fibrinogen with Zeolite Nanoparticles. Sci. Rep. 2019, 9, 1558. [Google Scholar] [CrossRef] [PubMed]
  265. Zhang, Z.; Guan, J.; Jiang, Z.; Yang, Y.; Liu, J.; Hua, W.; Mao, Y.; Li, C.; Lu, W.; Qian, J.; et al. Brain-Targeted Drug Delivery by Manipulating Protein Corona Functions. Nat. Commun. 2019, 10, 3561. [Google Scholar] [CrossRef]
  266. Wagner, S.; Zensi, A.; Wien, S.L.; Tschickardt, S.E.; Maier, W.; Vogel, T.; Worek, F.; Pietrzik, C.U.; Kreuter, J.; von Briesen, H. Uptake Mechanism of ApoE-Modified Nanoparticles on Brain Capillary Endothelial Cells as a Blood-Brain Barrier Model. PLoS ONE 2012, 7, e32568. [Google Scholar] [CrossRef]
  267. Hartl, N.; Adams, F.; Merkel, O.M. From Adsorption to Covalent Bonding: Apolipoprotein E Functionalization of Polymeric Nanoparticles for Drug Delivery Across the Blood–Brain Barrier. Adv. Ther. 2021, 4, 2000092. [Google Scholar] [CrossRef]
  268. Vidaurre-Agut, C.; Rivero-Buceta, E.; Romaní-Cubells, E.; Clemments, A.M.; Vera-Donoso, C.D.; Landry, C.C.; Botella, P. Protein Corona over Mesoporous Silica Nanoparticles: Influence of the Pore Diameter on Competitive Adsorption and Application to Prostate Cancer Diagnostics. ACS Omega 2019, 4, 8852–8861. [Google Scholar] [CrossRef]
  269. Choi, J.K.; Park, J.Y.; Lee, S.; Choi, Y.A.; Kwon, S.; Shin, M.J.; Yun, H.S.; Jang, Y.H.; Kang, J.; Kim, N.; et al. Greater Plasma Protein Adsorption on Mesoporous Silica Nanoparticles Aggravates Atopic Dermatitis. Int. J. Nanomed. 2022, 17, 4599–4617. [Google Scholar] [CrossRef] [PubMed]
  270. Partikel, K.; Korte, R.; Stein, N.C.; Mulac, D.; Herrmann, F.C.; Humpf, H.U.; Langer, K. Effect of Nanoparticle Size and PEGylation on the Protein Corona of PLGA Nanoparticles. Eur. J. Pharm. Biopharm. 2019, 141, 70–80. [Google Scholar] [CrossRef] [PubMed]
  271. Kyriakides, T.R.; Raj, A.; Tseng, T.H.; Xiao, H.; Nguyen, R.; Mohammed, F.S.; Halder, S.; Xu, M.; Wu, M.J.; Bao, S.; et al. Biocompatibility of Nanomaterials and Their Immunological Properties. Biomed. Mater. 2021, 16, 042005. [Google Scholar] [CrossRef]
  272. Sercombe, L.; Veerati, T.; Moheimani, F.; Wu, S.Y.; Sood, A.K.; Hua, S. Advances and Challenges of Liposome Assisted Drug Delivery. Front. Pharmacol. 2015, 6, 286. [Google Scholar] [CrossRef]
  273. Juhairiyah, F.; de Lange, E.C.M. Understanding Drug Delivery to the Brain Using Liposome-Based Strategies: Studies That Provide Mechanistic Insights Are Essential. AAPS J. 2021, 23, 114. [Google Scholar] [CrossRef] [PubMed]
  274. Giulimondi, F.; Digiacomo, L.; Pozzi, D.; Palchetti, S.; Vulpis, E.; Capriotti, A.L.; Chiozzi, R.Z.; Laganà, A.; Amenitsch, H.; Masuelli, L.; et al. Interplay of Protein Corona and Immune Cells Controls Blood Residency of Liposomes. Nat. Commun. 2019, 10, 3686. [Google Scholar] [CrossRef]
  275. Liu, X.Q.; Tang, R.Z. Biological Responses to Nanomaterials: Understanding Nano-Bio Effects on Cell Behaviors. Drug Deliv. 2017, 24, 1–15. [Google Scholar] [CrossRef]
  276. Ashkarran, A.A.; Gharibi, H.; Modaresi, S.M.; Saei, A.A.; Mahmoudi, M. Standardizing Protein Corona Characterization in Nanomedicine: A Multicenter Study to Enhance Reproducibility and Data Homogeneity. Nano Lett. 2024, 24, 9874–9881. [Google Scholar] [CrossRef]
  277. Ashkarran, A.A.; Gharibi, H.; Sadeghi, S.A.; Modaresi, S.M.; Wang, Q.; Lin, T.J.; Yerima, G.; Tamadon, A.; Sayadi, M.; Jafari, M.; et al. Small Molecule Modulation of Protein Corona for Deep Plasma Proteome Profiling. Nat. Commun. 2024, 15, 9638. [Google Scholar] [CrossRef]
  278. Pourali, P.; Neuhöferová, E.; Dzmitruk, V.; Benson, V. Investigation of Protein Corona Formed around Biologically Produced Gold Nanoparticles. Materials 2022, 15, 4615. [Google Scholar] [CrossRef]
  279. Bae, Y.; Liu, X. Unveiling the Effects of Protein Corona Formation on the Aggregation Kinetics of Gold Nanoparticles in Monovalent and Divalent Electrolytes. Environ. Pollut. 2024, 346, 123552. [Google Scholar] [CrossRef]
  280. Cedervall, T.; Lynch, I.; Lindman, S.; Berggård, T.; Thulin, E.; Nilsson, H.; Dawson, K.A.; Linse, S. Understanding the Nanoparticle-Protein Corona Using Methods to Quantify Exchange Rates and Affinities of Proteins for Nanoparticles. Proc. Natl. Acad. Sci. USA 2007, 104, 2050–2055. [Google Scholar]
  281. Arezki, Y.; Delalande, F.; Schaeffer-Reiss, C.; Cianférani, S.; Rapp, M.; Lebeau, L.; Pons, F.; Ronzani, C. Surface Charge Influences Protein Corona, Cell Uptake and Biological Effects of Carbon Dots. Nanoscale 2022, 14, 14695–14710. [Google Scholar] [CrossRef]
  282. Sakulkhu, U.; Mahmoudi, M.; Maurizi, L.; Coullerez, G.; Hofmann-Amtenbrink, M.; Vries, M.; Motazacker, M.; Rezaee, F.; Hofmann, H. Significance of Surface Charge and Shell Material of Superparamagnetic Iron Oxide Nanoparticle (SPION) Based Core/Shell Nanoparticles on the Composition of the Protein Corona. R. Soc. Chem. 2013, 3, 265–278. [Google Scholar] [CrossRef]
  283. El-Baz, N.; Nunn, B.M.; Bates, P.J.; O’Toole, M.G. The Impact of PEGylation on Cellular Uptake and In Vivo Biodistribution of Gold Nanoparticle MRI Contrast Agents. Bioengineering 2022, 9, 766. [Google Scholar] [CrossRef]
  284. Gutiérrez-Cruz, S.G.; Muñoz-Diosdado, A.; Gutiérrez-Calleja, R.A.; Rodríguez-Cortés, O.; Ortiz-Reyez, A.E.; Flores-Mejía, R. Influence of Physicochemical Factors on the Interaction of Metallic Nanoparticles with Immune System Cells. Front. Nanotechnol. 2024, 6, 1496230. [Google Scholar] [CrossRef]
  285. Neun, B.W.; Ilinskaya, A.N.; Dobrovolskaia, M.A. Analysis of Complement Activation by Nanoparticles. In Characterization of Nanoparticles Intended for Drug Delivery; Springer: New York, NY, USA, 2018; pp. 149–160. [Google Scholar]
  286. Haroon, H.B.; Dhillon, E.; Farhangrazi, Z.S.; Trohopoulos, P.N.; Simberg, D.; Moghimi, S.M. Activation of the Complement System by Nanoparticles and Strategies for Complement Inhibition. Eur. J. Pharm. Biopharm. 2023, 193, 227–240. [Google Scholar] [CrossRef]
  287. Wang, G.; Chen, F.; Banda, N.K.; Holers, V.M.; Wu, L.P.; Moghimi, S.M.; Simberg, D. Activation of Human Complement System by Dextran-Coated Iron Oxide Nanoparticles Is Not Affected by Dextran/Fe Ratio, Hydroxyl Modifications, and Crosslinking. Front. Immunol. 2016, 7, 418. [Google Scholar] [CrossRef]
  288. Shao, F.; Gao, Y.; Wang, W.; He, H.; Xiao, L.; Geng, X.; Xia, Y.; Guo, D.; Fang, J.; He, J.; et al. Silencing EGFR-Upregulated Expression of CD55 and CD59 Activates the Complement System and Sensitizes Lung Cancer to Checkpoint Blockade. Nat. Cancer 2022, 3, 1192–1210. [Google Scholar] [CrossRef]
  289. Moghimi, S.M.; Haroon, H.B.; Yaghmur, A.; Hunter, A.C.; Papini, E.; Farhangrazi, Z.S.; Simberg, D.; Trohopoulos, P.N. Perspectives on Complement and Phagocytic Cell Responses to Nanoparticles: From Fundamentals to Adverse Reactions. J. Control. Release 2023, 356, 115–129. [Google Scholar] [CrossRef]
  290. Coty, J.-B.; Eleamen Oliveira, E.; Vauthier, C. Tuning Complement Activation and Pathway through Controlled Molecular Architecture of Dextran Chains in Nanoparticle Corona. Int. J. Pharm. 2017, 532, 769–778. [Google Scholar] [CrossRef]
  291. Li, Y.; Wang, G.; Griffin, L.; Banda, N.K.; Saba, L.M.; Groman, E.V.; Scheinman, R.; Moghimi, S.M.; Simberg, D. Complement Opsonization of Nanoparticles: Differences between Humans and Preclinical Species. J. Control. Release 2021, 338, 548–556. [Google Scholar] [CrossRef]
  292. Zaleski, M.H.; Chase, L.S.; Hood, E.D.; Wang, Z.; Nong, J.; Espy, C.L.; Zamora, M.E.; Wu, J.; Morrell, L.J.; Muzykantov, V.R.; et al. Conjugation Chemistry Markedly Impacts Toxicity and Biodistribution of Targeted Nanoparticles, Mediated by Complement Activation. Adv. Mater. 2024, 37, e2409945. [Google Scholar] [CrossRef]
  293. La-Beck, N.M.; Islam, M.R.; Markiewski, M.M. Nanoparticle-Induced Complement Activation: Implications for Cancer Nanomedicine. Front. Immunol. 2021, 11, 603039. [Google Scholar] [CrossRef]
  294. Khan, H.A.; Kishore, U.; Alrokayan, S.H.; Ibrahim, K.E. Activation of the Complement Lectin Pathway by Iron Oxide Nanoparticles and Induction of Pro-Inflammatory Immune Response by Macrophages. Curr. Nanosci. 2025, 21, 82–91. [Google Scholar] [CrossRef]
  295. Dobó, J.; Kocsis, A.; Gál, P. Be on Target: Strategies of Targeting Alternative and Lectin Pathway Components in Complement-Mediated Diseases. Front. Immunol. 2018, 9, 1851. [Google Scholar] [CrossRef]
  296. Moghimi, S.M.; Wibroe, P.P.; Wu, L.; Farhangrazi, Z.S. Insidious Pathogen-Mimicking Properties of Nanoparticles in Triggering the Lectin Pathway of the Complement System. Eur. J. Nanomed. 2015, 7, 263–268. [Google Scholar] [CrossRef]
  297. Quach, Q.H.; Kah, J.C. Complement Activation by Gold Nanoparticles Passivated with Polyelectrolyte Ligands. RSC Adv. 2018, 8, 6616–6619. [Google Scholar] [CrossRef]
  298. Roumenina, L.T. Terminal Complement without C5 Convertase? Blood 2021, 137, 431–432. [Google Scholar] [CrossRef]
  299. Li, Y.; Wu, W.; Liu, Q.; Wu, Q.; Ren, P.; Xi, X.; Liu, H.; Zhao, J.; Zhang, W.; Wang, Z.; et al. Specific Surface-Modified Iron Oxide Nanoparticles Trigger Complement-Dependent Innate and Adaptive Antileukaemia Immunity. Nat. Commun. 2024, 15, 10400. [Google Scholar] [CrossRef]
  300. Barbey, C.; Wolf, H.; Wagner, R.; Pauly, D.; Breunig, M. A Shift of Paradigm: From Avoiding Nanoparticular Complement Activation in the Field of Nanomedicines to Its Exploitation in the Context of Vaccine Development. Eur. J. Pharm. Biopharm. 2023, 193, 119–128. [Google Scholar] [CrossRef]
  301. Padín-González, E.; Lancaster, P.; Bottini, M.; Gasco, P.; Tran, L.; Fadeel, B.; Wilkins, T.; Monopoli, M.P. Understanding the Role and Impact of Poly (Ethylene Glycol) (PEG) on Nanoparticle Formulation: Implications for COVID-19 Vaccines. Front. Bioeng. Biotechnol. 2022, 10, 882363. [Google Scholar] [CrossRef]
  302. Yang, Q.; Lai, S.K. Anti-PEG Immunity: Emergence, Characteristics, and Unaddressed Questions. Nanomed. Nanobiotechnol. 2015, 7, 655–677. [Google Scholar] [CrossRef]
  303. Overby, C.; Park, S.; Summers, A.; Benoit, D.S.W. Zwitterionic Peptides: Tunable next-Generation Stealth Nanoparticle Modifications. Bioact. Mater. 2023, 27, 113–124. [Google Scholar] [CrossRef]
  304. Liao, H.; Niu, C. Role of CD47-SIRPα Checkpoint in Nanomedicine-Based Anti-Cancer Treatment. Front. Bioeng. Biotechnol. 2022, 10, 887463. [Google Scholar] [CrossRef]
  305. Zhang, W.; Huang, Q.; Xiao, W.; Zhao, Y.; Pi, J.; Xu, H.; Zhao, H.; Xu, J.; Evans, C.E.; Jin, H. Advances in Anti-Tumor Treatments Targeting the CD47/SIRPα Axis. Front. Immunol. 2020, 11, 18. [Google Scholar] [CrossRef]
  306. Peng, Q.; Zhang, S.; Yang, Q.; Zhang, T.; Wei, X.Q.; Jiang, L.; Zhang, C.L.; Chen, Q.M.; Zhang, Z.R.; Lin, Y.F. Preformed Albumin Corona, a Protective Coating for Nanoparticles Based Drug Delivery System. Biomaterials 2013, 34, 8521–8530. [Google Scholar] [CrossRef]
  307. Mariam, J.; Sivakami, S.; Dongre, P.M. Albumin Corona on Nanoparticles—A Strategic Approach in Drug Delivery. Drug Deliv. 2016, 23, 2668–2676. [Google Scholar] [CrossRef]
  308. Chubarov, A.S. Serum Albumin for Magnetic Nanoparticles Coating. Magnetochemistry 2022, 8, 13. [Google Scholar] [CrossRef]
  309. Muhammad, Q.; Jang, Y.; Kang, S.H.; Moon, J.; Kim, W.J.; Park, H. Modulation of Immune Responses with Nanoparticles and Reduction of Their Immunotoxicity. Biomater. Sci. 2020, 8, 1490–1501. [Google Scholar] [CrossRef]
  310. Chao, C.J.; Zhang, E.; Trinh, D.N.; Udofa, E.; Lin, H.; Silvers, C.; Huo, J.; He, S.; Zheng, J.; Cai, X.; et al. Integrating Antigen Capturing Nanoparticles and Type 1 Conventional Dendritic Cell Therapy for in Situ Cancer Immunization. Nat. Commun. 2025, 16, 4578. [Google Scholar] [CrossRef]
  311. Baron, L.; Gombault, A.; Fanny, M.; Villeret, B.; Savigny, F.; Guillou, N.; Panek, C.; Le Bert, M.; Lagente, V.; Rassendren, F.; et al. The NLRP3 Inflammasome Is Activated by Nanoparticles through ATP, ADP and Adenosine. Cell Death Dis. 2015, 6, e1629. [Google Scholar] [CrossRef]
  312. Mellors, J.; Tipton, T.; Longet, S.; Carroll, M. Viral Evasion of the Complement System and Its Importance for Vaccines and Therapeutics. Front. Immunol. 2020, 11, 1450. [Google Scholar] [CrossRef]
  313. Ray, P.; Haideri, N.; Haque, I.; Mohammed, O.; Chakraborty, S.; Banerjee, S.; Quadir, M.; Brinker, A.E.; Banerjee, S.K. The Impact of Nanoparticles on the Immune System: A Gray Zone of Nanomedicine. J. Immunol. Sci. 2021, 5, 19–33. [Google Scholar] [CrossRef]
  314. Sushnitha, M.; Evangelopoulos, M.; Tasciotti, E.; Taraballi, F. Cell Membrane-Based Biomimetic Nanoparticles and the Immune System: Immunomodulatory Interactions to Therapeutic Applications. Front. Bioeng. Biotechnol. 2020, 8, 627. [Google Scholar] [CrossRef]
  315. Poon, W.; Zhang, Y.-N.; Ouyang, B.; Kingston, B.R.; Wu, J.L.Y.; Wilhelm, S.; Chan, W.C.W. Elimination Pathways of Nanoparticles. ACS Nano 2019, 13, 5785–5798. [Google Scholar] [CrossRef]
  316. Yah, C.S.; Iyuke, S.E.; Simate, G.S. A Review of Nanoparticles Toxicity and Their Routes of Exposures. Iran. J. Pharm. Sci. Winter 2011, 8, 299–314. [Google Scholar]
  317. Nazarenko, Y.; Han, T.W.; Lioy, P.J.; Mainelis, G. Potential for Exposure to Engineered Nanoparticles from Nanotechnology-Based Consumer Spray Products. J. Expo. Sci. Envrion. Epidemiol. 2011, 21, 515–528. [Google Scholar] [CrossRef]
  318. Wan, Q.; Zhang, X.; Zhou, D.; Xie, R.; Cai, Y.; Zhang, K.; Sun, X. Inhaled Nano-Based Therapeutics for Pulmonary Fibrosis: Recent Advances and Future Prospects. J. Nanobiotechnol. 2023, 21, 215. [Google Scholar] [CrossRef]
  319. Wang, M.; Lai, X.; Shao, L.; Li, L. Evaluation of Immunoresponses and Cytotoxicity from Skin Exposure to Metallic Nanoparticles. Int. J. Nanomed. 2018, 13, 4445–4459. [Google Scholar] [CrossRef]
  320. Date, A.A.; Hanes, J.; Ensign, L.M. Nanoparticles for Oral Delivery: Design, Evaluation and State-of-the-Art. J. Control. Release 2016, 240, 504–526. [Google Scholar] [CrossRef]
  321. Gaillet, S.; Rouanet, J.M. Silver Nanoparticles: Their Potential Toxic Effects after Oral Exposure and Underlying Mechanisms—A Review. Food Chem. Toxicol. 2015, 77, 58–63. [Google Scholar] [CrossRef] [PubMed]
  322. Chen, Z.; Han, S.; Zhou, D.; Zhou, S.; Jia, G. Effects of Oral Exposure to Titanium Dioxide Nanoparticles on Gut Microbiota and Gut-Associated Metabolism in Vivo. Nanoscale 2019, 11, 22398–22412. [Google Scholar] [CrossRef]
  323. Lotfipour, F.; Shahi, S.; Farjami, A.; Salatin, S.; Mahmoudian, M.; Dizaj, S.M. Safety and Toxicity Issues of Therapeutically Used Nanoparticles from the Oral Route. Biomed. Res. Int. 2021, 2021, 9322282. [Google Scholar] [CrossRef]
  324. Huang, X.; Kaneda-Nakashima, K.; Kadonaga, Y.; Kabayama, K.; Shimoyama, A.; Ooe, K.; Kato, H.; Toyoshima, A.; Shinohara, A.; Haba, H.; et al. Astatine-211-Labeled Gold Nanoparticles for Targeted Alpha-Particle Therapy via Intravenous Injection. Pharmaceutics 2022, 14, 2705. [Google Scholar] [CrossRef]
  325. Barua, S.; Mitragotri, S. Challenges Associated with Penetration of Nanoparticles across Cell and Tissue Barriers: A Review of Current Status and Future Prospects. Nano Today 2014, 9, 223–243. [Google Scholar] [CrossRef]
  326. Liu, C.; Jiang, X.; Gan, Y.; Yu, M. Engineering Nanoparticles to Overcome the Mucus Barrier for Drug Delivery: Design, Evaluation and State-of-the-Art. Med. Drug Discov. 2021, 12, 100110. [Google Scholar] [CrossRef]
  327. Tian, X.; Chong, Y.; Ge, C. Understanding the Nano–Bio Interactions and the Corresponding Biological Responses. Front. Chem. 2020, 8, 446. [Google Scholar] [CrossRef]
  328. Sonwani, S.; Madaan, S.; Arora, J.; Suryanarayan, S.; Rangra, D.; Mongia, N.; Vats, T.; Saxena, P. Inhalation Exposure to Atmospheric Nanoparticles and Its Associated Impacts on Human Health: A Review. Front. Sustain. Cities 2021, 3, 690444. [Google Scholar] [CrossRef]
  329. McCormick, S.; Niang, M.; Dahm, M.M. Occupational Exposures to Engineered Nanomaterials: A Review of Workplace Exposure Assessment Methods. Curr. Envrion. Health Rep. 2021, 8, 223–234. [Google Scholar] [CrossRef] [PubMed]
  330. Zhou, X.; Jin, W.; Ma, J. Lung Inflammation Perturbation by Engineered Nanoparticles. Front. Bioeng. Biotechnol. 2023, 11, 1199230. [Google Scholar] [CrossRef] [PubMed]
  331. Lee, C.E.; Rezaee, F. Nanoparticles and Airway Epithelial Cells: Exploring the Impacts and Methodologies in Toxicity Assessment. Int. J. Mol. Sci. 2024, 25, 7885. [Google Scholar] [CrossRef]
  332. Zhu, Y.; Chidekel, A.; Shaffer, T.H. Cultured Human Airway Epithelial Cells (Calu-3): A Model of Human Respiratory Function, Structure, and Inflammatory Responses. Crit. Care Res. Pract. 2010, 2010, 394578. [Google Scholar] [CrossRef]
  333. Meindl, C.; Öhlinger, K.; Zrim, V.; Steinkogler, T.; Fröhlich, E. Screening for Effects of Inhaled Nanoparticles in Cell Culture Models for Prolonged Exposure. Nanomaterials 2021, 11, 606. [Google Scholar] [CrossRef]
  334. Rabiei, M.; Kashanian, S.; Samavati, S.S.; Jamasb, S.; McInnes, S.J.P. Nanomaterial and Advanced Technologies in Transdermal Drug Delivery. J. Drug Target. 2020, 28, 356–367. [Google Scholar] [CrossRef]
  335. Piluk, T.; Faccio, G.; Letsiou, S.; Liang, R.; Freire-Gormaly, M. A Critical Review Investigating the Use of Nanoparticles in Cosmetic Skin Products. Envrion. Sci. Nano 2024, 11, 3674–3692. [Google Scholar] [CrossRef]
  336. Tak, Y.K.; Pal, S.; Naoghare, P.K.; Rangasamy, S.; Song, J.M. Shape-Dependent Skin Penetration of Silver Nanoparticles: Does It Really Matter? Sci. Rep. 2015, 5, 16908. [Google Scholar] [CrossRef]
  337. Liang, X.; Xu, Z.; Grice, J.; Zvyagin, A.; Roberts, M.; Liu, X. Penetration of Nanoparticles into Human Skin. Curr. Pharm. Des. 2013, 19, 6353–6366. [Google Scholar] [CrossRef]
  338. Liang, Y.; Simaiti, A.; Xu, M.; Lv, S.; Jiang, H.; He, X.; Fan, Y.; Zhu, S.; Du, B.; Yang, W.; et al. Antagonistic Skin Toxicity of Co-Exposure to Physical Sunscreen Ingredients Zinc Oxide and Titanium Dioxide Nanoparticles. Nanomaterials 2022, 12, 2769. [Google Scholar] [CrossRef]
  339. Ailawadi, S.; Talreja, R.; Panstingel, N.; Sulentic, C. Titanium Dioxide and Zinc Oxide Nanoparticles in Sunscreen: Potential Impact on Cytokine Expression in Human Skin Pre- and Post-UVB Exposure. Arch. Microbiol. Immunol. 2023, 7, 443–455. [Google Scholar] [CrossRef]
  340. Singh, G.; Thakur, N.; Kumar, R. Nanoparticles in Drinking Water: Assessing Health Risks and Regulatory Challenges. Sci. Total Environ. 2024, 949, 174940. [Google Scholar] [CrossRef]
  341. Vitulo, M.; Gnodi, E.; Meneveri, R.; Barisani, D. Interactions between Nanoparticles and Intestine. Int. J. Mol. Sci. 2022, 23, 4339. [Google Scholar] [CrossRef]
  342. Karavolos, M.; Holban, A. Nanosized Drug Delivery Systems in Gastrointestinal Targeting: Interactions with Microbiota. Pharmaceuticals 2016, 9, 62. [Google Scholar] [CrossRef]
  343. Jakic, K.; Selc, M.; Razga, F.; Nemethova, V.; Mazancova, P.; Havel, F.; Sramek, M.; Zarska, M.; Proska, J.; Masanova, V.; et al. Long-Term Accumulation, Biological Effects and Toxicity of BSA-Coated Gold Nanoparticles in the Mouse Liver, Spleen, and Kidneys. Int. J. Nanomed. 2024, 19, 4103–4120. [Google Scholar] [CrossRef]
  344. Tsoi, K.M.; Macparland, S.A.; Ma, X.Z.; Spetzler, V.N.; Echeverri, J.; Ouyang, B.; Fadel, S.M.; Sykes, E.A.; Goldaracena, N.; Kaths, J.M.; et al. Mechanism of Hard-Nanomaterial Clearance by the Liver. Nat. Mater. 2016, 15, 1212–1221. [Google Scholar] [CrossRef] [PubMed]
  345. Elje, E.; Mariussen, E.; Moriones, O.H.; Bastús, N.G.; Puntes, V.; Kohl, Y.; Dusinska, M.; Rundén-Pran, E. Hepato(Geno)Toxicity Assessment of Nanoparticles in a Hepg2 Liver Spheroid Model. Nanomaterials 2020, 10, 545. [Google Scholar] [CrossRef] [PubMed]
  346. Zhang, Y.; Poon, W.; Tavares, A.J.; McGilvray, I.D.; Chan, W.C.W. Nanoparticle–Liver Interactions: Cellular Uptake and Hepatobiliary Elimination. J. Control. Release 2016, 240, 332–348. [Google Scholar] [CrossRef]
  347. Bannunah, A.; Cavanagh, R.; Shubber, S.; Vllasaliu, D.; Stolnik, S. Difference in Endocytosis Pathways Used by Differentiated Versus Nondifferentiated Epithelial Caco-2 Cells to Internalize Nanosized Particles. Mol. Pharm. 2024, 21, 3603–3612. [Google Scholar] [CrossRef]
  348. Mahlert, L.; Anderski, J.; Mulac, D.; Langer, K. The Impact of Gastrointestinal Mucus on Nanoparticle Penetration—In Vitro Evaluation of Mucus-Penetrating Nanoparticles for Photodynamic Therapy. Eur. J. Pharm. Sci. 2019, 133, 28–39. [Google Scholar] [CrossRef] [PubMed]
  349. Vincentini, O.; Prota, V.; Cecchetti, S.; Bertuccini, L.; Tinari, A.; Iosi, F.; De Angelis, I. Towards the Standardization of Intestinal In Vitro Advanced Barrier Model for Nanoparticles Uptake and Crossing: The SiO2 Case Study. Cells 2022, 11, 3357. [Google Scholar] [CrossRef] [PubMed]
  350. Trac, N.; Chung, E.J. Overcoming Physiological Barriers by Nanoparticles for Intravenous Drug Delivery to the Lymph Nodes. Exp. Biol. Med. 2021, 246, 2358–2371. [Google Scholar] [CrossRef] [PubMed]
  351. Ernst, L.M.; Casals, E.; Italiani, P.; Boraschi, D.; Puntes, V. The Interactions between Nanoparticles and the Innate Immune System from a Nanotechnologist Perspective. Nanomaterials 2021, 11, 2991. [Google Scholar] [CrossRef]
  352. Omo-Lamai, S.; Zamora, M.E.; Patel, M.N.; Wu, J.; Nong, J.; Wang, Z.; Peshkova, A.; Chase, L.S.; Essien, E.-O.; Muzykantov, V.; et al. Physicochemical Targeting of Lipid Nanoparticles to the Lungs Induces Clotting: Mechanisms and Solutions. Adv. Mater. 2023, 36, e2312026. [Google Scholar] [CrossRef]
  353. Huang, Y.; Guo, X.; Wu, Y.; Chen, X.; Feng, L.; Xie, N.; Shen, G. Nanotechnology’s Frontier in Combatting Infectious and Inflammatory Diseases: Prevention and Treatment. Signal Transduct. Target. Ther. 2024, 9, 34. [Google Scholar] [CrossRef]
  354. Al Khafaji, A.S.; Donovan, M.D. Endocytic Uptake of Solid Lipid Nanoparticles by the Nasal Mucosa. Pharmaceutics 2021, 13, 761. [Google Scholar] [CrossRef]
  355. Gandhi, S.; Shastri, D.H.; Shah, J.; Nair, A.B.; Jacob, S. Nasal Delivery to the Brain: Harnessing Nanoparticles for Effective Drug Transport. Pharmaceutics 2024, 16, 481. [Google Scholar] [CrossRef]
  356. Zhu, Q.; Chen, Z.; Paul, P.K.; Lu, Y.; Wu, W.; Qi, J. Oral Delivery of Proteins and Peptides: Challenges, Status Quo and Future Perspectives. Acta Pharm. Sin. B 2021, 11, 2416–2448. [Google Scholar] [CrossRef]
  357. Koo, J.; Lim, C.; Oh, K.T. Recent Advances in Intranasal Administration for Brain-Targeting Delivery: A Comprehensive Review of Lipid-Based Nanoparticles and Stimuli-Responsive Gel Formulations. Int. J. Nanomed. 2024, 19, 1767–1807. [Google Scholar] [CrossRef]
  358. Huang, Q.; Chen, X.; Yu, S.; Gong, G.; Shu, H. Research Progress in Brain-Targeted Nasal Drug Delivery. Front. Aging Neurosci. 2023, 15, 1341295. [Google Scholar] [CrossRef]
  359. Alhowyan, A.A.; Kalam, M.A.; Iqbal, M.; Raish, M.; El-Toni, A.M.; Alkholief, M.; Almomen, A.A.; Alshamsan, A. Mesoporous Silica Nanoparticles Coated with Carboxymethyl Chitosan for 5-Fluorouracil Ocular Delivery: Characterization, In Vitro and In Vivo Studies. Molecules 2023, 28, 1260. [Google Scholar] [CrossRef]
  360. Zhu, S.; Gong, L.; Li, Y.; Xu, H.; Gu, Z.; Zhao, Y. Safety Assessment of Nanomaterials to Eyes: An Important but Neglected Issue. Adv. Sci. 2019, 6, 1802289. [Google Scholar] [CrossRef]
  361. Clementino, A.R.; Pellegrini, G.; Banella, S.; Colombo, G.; Cantù, L.; Sonvico, F.; Del Favero, E. Structure and Fate of Nanoparticles Designed for the Nasal Delivery of Poorly Soluble Drugs. Mol. Pharm. 2021, 18, 3132–3146. [Google Scholar] [CrossRef]
  362. Yang, C.; Yang, J.; Lu, A.; Gong, J.; Yang, Y.; Lin, X.; Li, M.; Xu, H. Nanoparticles in Ocular Applications and Their Potential Toxicity. Front. Mol. Biosci. 2022, 9, 931759. [Google Scholar] [CrossRef] [PubMed]
  363. Sundaram, S.; Roy, S.K.; Ambati, B.K.; Kompella, U.B. Surface-functionalized Nanoparticles for Targeted Gene Delivery across Nasal Respiratory Epithelium. FASEB J. 2009, 23, 3752–3765. [Google Scholar] [CrossRef] [PubMed]
  364. Croissant, J.G.; Fatieiev, Y.; Khashab, N.M. Degradability and Clearance of Silicon, Organosilica, Silsesquioxane, Silica Mixed Oxide, and Mesoporous Silica Nanoparticles. Adv. Mater. 2017, 29, 1604634. [Google Scholar] [CrossRef] [PubMed]
  365. Kraegeloh, A.; Suarez-Merino, B.; Sluijters, T.; Micheletti, C. Implementation of Safe-by-Design for Nanomaterial Development and Safe Innovation: Why We Need a Comprehensive Approach. Nanomaterials 2018, 8, 239. [Google Scholar] [CrossRef]
  366. Đorđević, S.; Gonzalez, M.M.; Conejos-Sánchez, I.; Carreira, B.; Pozzi, S.; Acúrcio, R.C.; Satchi-Fainaro, R.; Florindo, H.F.; Vicent, M.J. Current Hurdles to the Translation of Nanomedicines from Bench to the Clinic. Drug Deliv. Transl. Res. 2022, 12, 500–525. [Google Scholar] [CrossRef]
  367. Frickenstein, A.N.; Hagood, J.M.; Britten, C.N.; Abbott, B.S.; McNally, M.W.; Vopat, C.A.; Patterson, E.G.; Maccuaig, W.M.; Jain, A.; Walters, K.B.; et al. Mesoporous Silica Nanoparticles: Properties and Strategies for Enhancing Clinical Effect. Pharmaceutics 2021, 13, 570. [Google Scholar] [CrossRef]
  368. Rasmussen, K.; Rauscher, H.; Kearns, P.; González, M.; Riego Sintes, J. Developing OECD Test Guidelines for Regulatory Testing of Nanomaterials to Ensure Mutual Acceptance of Test Data. Regul. Toxicol. Pharmacol. 2019, 104, 74–83. [Google Scholar] [CrossRef]
  369. OECD. Building Trust and Enhancing Dialogue for Safe-and-Sustainable-by-Design (SSBD) Innovation: Developing Tools to Enhance Trusted Environments. In OECD Series on the Safety of Manufactured Nanomaterials and Other Advanced Materials; OECD Publishing: Paris, France, 2024. [Google Scholar] [CrossRef]
  370. OECD. Developments in Delegations on the Safety of Manufactured Nanomaterials and Advanced Materials Between July 2023 and June 2024—Tour de Table. In OECD Series on the Safety of Manufactured Nanomaterials and Other Advanced Materials; OECD Publishing: Paris, France, 2024; ISBN 9789264647909. [Google Scholar]
  371. Oualikene-Gonin, W.; Sautou, V.; Ezan, E.; Bastos, H.; Bellissant, E.; Belgodère, L.; Maison, P.; Ankri, J. Regulatory Assessment of Nano-Enabled Health Products in Public Health Interest. Position of the Scientific Advisory Board of the French National Agency for the Safety of Medicines and Health Products. Front. Public Health 2023, 11, 1125577. [Google Scholar] [CrossRef]
  372. Paradise, J. Regulating Nanomedicine at the Food and Drug Administration. AMA J. Ethics 2019, 21, E347–E355. [Google Scholar] [CrossRef]
  373. Clogston, J.D. The Importance of Nanoparticle Physicochemical Characterization for Immunology Research: What We Learned and What We Still Need to Understand. Adv. Drug Deliv. Rev. 2021, 176, 113897. [Google Scholar] [CrossRef]
  374. ISO/TR 10993–22:2017(en); Biological Evaluation of Medical Devices—Part 22: Guidance on Nanomaterials. International Organization for Standardization: Geneva, Switzerland, 2013. Available online: https://www.iso.org/obp/ui#iso:std:iso:tr:10993:-22:ed-1:v1:en (accessed on 10 October 2025).
  375. Rodríguez-Ibarra, C.; Déciga-Alcaraz, A.; Ispanixtlahuatl-Meráz, O.; Medina-Reyes, E.I.; Delgado-Buenrostro, N.L.; Chirino, Y.I. International Landscape of Limits and Recommendations for Occupational Exposure to Engineered Nanomaterials. Toxicol. Lett. 2020, 322, 111–119. [Google Scholar] [CrossRef]
  376. Sharmile, N.; Chowdhury, R.R.; Desai, S. A Comprehensive Review of Quality Control and Reliability Research in Micro–Nano Technology. Technologies 2025, 13, 94. [Google Scholar] [CrossRef]
  377. Khan, F.; Shariq, M.; Asif, M.; Siddiqui, M.A.; Malan, P.; Ahmad, F. Green Nanotechnology: Plant-Mediated Nanoparticle Synthesis and Application. Nanomaterials 2022, 12, 673. [Google Scholar] [CrossRef]
  378. Hano, C.; Abbasi, B.H. Plant-Based Green Synthesis of Nanoparticles: Production, Characterization and Applications. Biomolecules 2022, 12, 31. [Google Scholar] [CrossRef] [PubMed]
  379. Abuzeid, H.M.; Julien, C.M.; Zhu, L.; Hashem, A.M. Green Synthesis of Nanoparticles and Their Energy Storage, Environmental, and Biomedical Applications. Crystals 2023, 13, 1576. [Google Scholar] [CrossRef]
  380. Isigonis, P.; Afantitis, A.; Antunes, D.; Bartonova, A.; Beitollahi, A.; Bohmer, N.; Bouman, E.; Chaudhry, Q.; Cimpan, M.R.; Cimpan, E.; et al. Risk Governance of Emerging Technologies Demonstrated in Terms of Its Applicability to Nanomaterials. Small 2020, 16, e2003303. [Google Scholar] [CrossRef] [PubMed]
  381. Isigonis, P.; Bouman, E.A.; Varsou, D.D.; Jensen, K.A.; Fransman, W.; Drobne, D.; Rollon, B.P.; Ballesteros, A.; Rodríguez-Llopis, I.; Säämänen, A.; et al. A Nano Risk Governance Portal Supporting Risk Governance of Nanomaterials and Nano-Enabled Products. Comput. Struct. Biotechnol. J. 2025, 29, 177–185. [Google Scholar] [CrossRef]
  382. Johnston, L.J.; Du, X.; Zborowski, A.; Kennedy, D.C. Characterization and Cellular Toxicity Studies of Commercial Manganese Oxide Nanoparticles. Nanomaterials 2024, 14, 198. [Google Scholar] [CrossRef]
  383. Bruckmann, F.d.S.; Nunes, F.B.; Salles, T.d.R.; Franco, C.; Cadoná, F.C.; Bohn Rhoden, C.R. Biological Applications of Silica-Based Nanoparticles. Magnetochemistry 2022, 8, 131. [Google Scholar] [CrossRef]
  384. Hosseinpour, S.; Walsh, L.J.; Xu, C. Biomedical Application of Mesoporous Silica Nanoparticles as Delivery Systems: A Biological Safety Perspective. J. Mater. Chem. B 2020, 8, 9863–9876. [Google Scholar] [CrossRef] [PubMed]
  385. Mohamed, F.; Oo, M.K.; Chatterjee, B.; Alallam, B. Biocompatible Supramolecular Mesoporous Silica Nanoparticles as the Next-Generation Drug Delivery System. Front. Pharmacol. 2022, 13, 886981. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Functionalisation and loading of PSNs for biomedical applications (Created in BioRender. Patel, T. (2025) https://BioRender.com/7bksooi). This schematic diagram illustrates the diverse functionalisation and cargo-loading capabilities of PSNs. PSNs can be engineered to incorporate various molecules, enhancing their functionality for targeted drug delivery, imaging, and therapeutic applications.
Figure 1. Functionalisation and loading of PSNs for biomedical applications (Created in BioRender. Patel, T. (2025) https://BioRender.com/7bksooi). This schematic diagram illustrates the diverse functionalisation and cargo-loading capabilities of PSNs. PSNs can be engineered to incorporate various molecules, enhancing their functionality for targeted drug delivery, imaging, and therapeutic applications.
Nanomaterials 15 01766 g001
Figure 2. Safe-by-Design Principles of PSNs (Created in BioRender. Patel, T. (2025) https://BioRender.com/o2qwh8p). The S.A.F.E. framework is based on Standardised characterization, Assessment of biocompatibility, Facilitation of toxicity and exposure routes and Evaluation of clinical translation.
Figure 2. Safe-by-Design Principles of PSNs (Created in BioRender. Patel, T. (2025) https://BioRender.com/o2qwh8p). The S.A.F.E. framework is based on Standardised characterization, Assessment of biocompatibility, Facilitation of toxicity and exposure routes and Evaluation of clinical translation.
Nanomaterials 15 01766 g002
Figure 3. PSN-induced activation of the p53 signalling pathway and its crosstalk with mTOR, PI3K/AKT, NF-kB, and MAPK pathways (Created in BioRender. Patel, T. (2025) https://BioRender.com/dwxczus). This schematic diagram illustrates the potential p53 signalling pathway in response to PSN-induced oxidative stress and DNA damage. Oxidative stress from PSNs activates JNK and p38 MAPK pathways, which phosphorylate p53, enhancing its transcriptional activity. Simultaneously, DNA damage triggers ATM/ATR kinases, leading to CHK1/CHK2 phosphorylation and p53 stabilization. Once activated, p53 regulates multiple cellular outcomes, including cell cycle arrest, apoptosis, senescence, and DNA repair. Cell cycle arrest occurs through p21 (CDKN1A) upregulation, which inhibits CDK2, and Cyclin E, preventing G1/S transition. DNA damage activates p16INK4a/RB pathway, while p53-induced DNA repair is facilitated via DRAM-1-mediated lysosomal activation. If the damage is irreparable, p53 induces apoptosis by upregulating pro-apoptotic genes (NOXA, BAD, PUMA, BAX, and BAK), leading to MOMP, cytochrome c release, and activation of caspase 9 and caspase 3. Additionally, p53 interacts with mTOR signalling, where Sestrin 2 activates AMPK, inhibiting mTOR (via TSC1/2) and autophagy. The PI3K/AKT pathway antagonizes p53 by activating MDM2, leading to p53 degradation, while also promoting cell survival and proliferation. Furthermore, AKT activation stimulates NF-kB, enhancing inflammatory responses and counteracting p53-mediated apoptosis. This intricate interplay between p53 and other pathways determines cellular fate, balancing repair, survival, or cell death in response to PSN exposure.
Figure 3. PSN-induced activation of the p53 signalling pathway and its crosstalk with mTOR, PI3K/AKT, NF-kB, and MAPK pathways (Created in BioRender. Patel, T. (2025) https://BioRender.com/dwxczus). This schematic diagram illustrates the potential p53 signalling pathway in response to PSN-induced oxidative stress and DNA damage. Oxidative stress from PSNs activates JNK and p38 MAPK pathways, which phosphorylate p53, enhancing its transcriptional activity. Simultaneously, DNA damage triggers ATM/ATR kinases, leading to CHK1/CHK2 phosphorylation and p53 stabilization. Once activated, p53 regulates multiple cellular outcomes, including cell cycle arrest, apoptosis, senescence, and DNA repair. Cell cycle arrest occurs through p21 (CDKN1A) upregulation, which inhibits CDK2, and Cyclin E, preventing G1/S transition. DNA damage activates p16INK4a/RB pathway, while p53-induced DNA repair is facilitated via DRAM-1-mediated lysosomal activation. If the damage is irreparable, p53 induces apoptosis by upregulating pro-apoptotic genes (NOXA, BAD, PUMA, BAX, and BAK), leading to MOMP, cytochrome c release, and activation of caspase 9 and caspase 3. Additionally, p53 interacts with mTOR signalling, where Sestrin 2 activates AMPK, inhibiting mTOR (via TSC1/2) and autophagy. The PI3K/AKT pathway antagonizes p53 by activating MDM2, leading to p53 degradation, while also promoting cell survival and proliferation. Furthermore, AKT activation stimulates NF-kB, enhancing inflammatory responses and counteracting p53-mediated apoptosis. This intricate interplay between p53 and other pathways determines cellular fate, balancing repair, survival, or cell death in response to PSN exposure.
Nanomaterials 15 01766 g003
Figure 4. Structural organization of protein corona on PSNs (Created in BioRender. Patel, T. (2025) https://BioRender.com/fcoz305). A schematic diagram of protein corona formation on the surface of PSNs. In the middle is the core of the NP, surrounded by the inner layer, which represents the hard protein corona and the outer layer, which represents the soft protein corona.
Figure 4. Structural organization of protein corona on PSNs (Created in BioRender. Patel, T. (2025) https://BioRender.com/fcoz305). A schematic diagram of protein corona formation on the surface of PSNs. In the middle is the core of the NP, surrounded by the inner layer, which represents the hard protein corona and the outer layer, which represents the soft protein corona.
Nanomaterials 15 01766 g004
Figure 5. Complement activation pathways induced by PSNs (Created in BioRender. Patel, T. (2025) https://BioRender.com/i2x0h8c). This figure illustrates the activation of the complement system by PSNs via three distinct pathways: the classical, lectin, and alternative pathways. The classical pathway is triggered by antigen–antibody complexes involving C1q, while MBL and MASP activate the lectin pathway. The alternative pathway is initiated through spontaneous hydrolysis of C3 into C3(H2O). Activation of C3 leads to the generation of C3a and C3b, where C3a functions as an anaphylatoxin that recruits neutrophils and monocytes, contributing to inflammation, while C3b facilitates opsonisation for phagocytosis. Further activation of C5 results in the formation of C5a, another potent inflammatory mediator, and C5b, which initiates MAC formation. The MAC, composed of C5b, C6, C7, C8, and C9, leads to cell lysis. Additionally, a self-amplification loop of C3 enhances the complement response. This schematic highlights the immune-modulatory effects of PSNs, demonstrating their potential to trigger complement activation, which may influence their biocompatibility and therapeutic applications.
Figure 5. Complement activation pathways induced by PSNs (Created in BioRender. Patel, T. (2025) https://BioRender.com/i2x0h8c). This figure illustrates the activation of the complement system by PSNs via three distinct pathways: the classical, lectin, and alternative pathways. The classical pathway is triggered by antigen–antibody complexes involving C1q, while MBL and MASP activate the lectin pathway. The alternative pathway is initiated through spontaneous hydrolysis of C3 into C3(H2O). Activation of C3 leads to the generation of C3a and C3b, where C3a functions as an anaphylatoxin that recruits neutrophils and monocytes, contributing to inflammation, while C3b facilitates opsonisation for phagocytosis. Further activation of C5 results in the formation of C5a, another potent inflammatory mediator, and C5b, which initiates MAC formation. The MAC, composed of C5b, C6, C7, C8, and C9, leads to cell lysis. Additionally, a self-amplification loop of C3 enhances the complement response. This schematic highlights the immune-modulatory effects of PSNs, demonstrating their potential to trigger complement activation, which may influence their biocompatibility and therapeutic applications.
Nanomaterials 15 01766 g005
Figure 6. Routes of NP exposure and administration (Created in BioRender. Patel, T. (2025) https://BioRender.com/d8hm9lr). This figure illustrates the various routes of NP exposure, including mucosal (nasal and ocular), oral, inhalation, systemic, and dermal pathways, each with specific formulations designed for targeted delivery.
Figure 6. Routes of NP exposure and administration (Created in BioRender. Patel, T. (2025) https://BioRender.com/d8hm9lr). This figure illustrates the various routes of NP exposure, including mucosal (nasal and ocular), oral, inhalation, systemic, and dermal pathways, each with specific formulations designed for targeted delivery.
Nanomaterials 15 01766 g006
Figure 7. Biodegradation of PSNs (Created in BioRender. Patel, T. (2025) https://BioRender.com/q18ximj, reproduced from [5]. This figure demonstrates the steps in biodegradation. Step 1 shows hydration, step 2 shows hydrolysis, and step 3 shows dissolution. The red text represents the reactants involved in the breakdown reactions, whilst the blue text represents the products formed during degradation.
Figure 7. Biodegradation of PSNs (Created in BioRender. Patel, T. (2025) https://BioRender.com/q18ximj, reproduced from [5]. This figure demonstrates the steps in biodegradation. Step 1 shows hydration, step 2 shows hydrolysis, and step 3 shows dissolution. The red text represents the reactants involved in the breakdown reactions, whilst the blue text represents the products formed during degradation.
Nanomaterials 15 01766 g007
Figure 8. Overview of the nanotoxicity of PSNs (Created in BioRender. Patel, T. (2025) https://BioRender.com/4uz6rh2). This schematic diagram illustrates the cellular responses of PSNs, highlighting membrane damage, agglomeration, protein corona formation (yellow ring), activation of the complement system, cellular uptake, induction of oxidative stress leading to cytotoxic and genotoxic effects as well as possible cell signalling pathways activated by the toxicity of PSNs.
Figure 8. Overview of the nanotoxicity of PSNs (Created in BioRender. Patel, T. (2025) https://BioRender.com/4uz6rh2). This schematic diagram illustrates the cellular responses of PSNs, highlighting membrane damage, agglomeration, protein corona formation (yellow ring), activation of the complement system, cellular uptake, induction of oxidative stress leading to cytotoxic and genotoxic effects as well as possible cell signalling pathways activated by the toxicity of PSNs.
Nanomaterials 15 01766 g008
Table 1. Porous and Mesoporous Silica Nanoparticle Applications in Various Clinical Trials.
Table 1. Porous and Mesoporous Silica Nanoparticle Applications in Various Clinical Trials.
Clinical Trial StudyClinical Trial IDCurrent StatusStudy Type and PhaseApplicationPurposeOutcome
Safety Evaluation of Porous Silica in MenNCT03667430 [9,10]Completed in 2016Interventional and Not applicableWeight LossTo evaluate the safety and tolerability of oral administration of PSNs in healthy men, primarily for applications in weight management and metabolic health.Completed: Oral intake up to 9 g/day of porous silica as a food additive was safe and well-tolerated, with only mild gastrointestinal side effects and no major adverse events reported.
Effect of Different NanoScaffolds on Pulp Regeneration in Non-Vital Immature Permanent TeethNCT07121348 [11]Active, not recruitingInterventional and Not applicableRegenerative MedicineTo evaluate the clinical and radiographic outcomes of regenerative endodontic procedures in immature non-vital permanent teeth using different nanoscaffold materials, including MSNs.Ongoing: Outcome measures include radiographic apical closure 12 months post-procedure. No published results yet.
Targeted Silica Nanoparticles for Real-Time Image-Guided Intraoperative Mapping of Nodal MetastasesNCT02106598 [12,13]Active, not recruitingInterventional and Phase 1, Phase 2Drug DeliveryTo assess the feasibility and safety of using ultrasmall targeted core-shell silica nanoparticles for fluorescence-guided sentinel lymph node biopsy in patients with melanoma of the head and neck.Ongoing: Nanoparticles enabled high-sensitivity lymph node mapping. No adverse events were observed. The approach was feasible and safe, offering improved intraoperative guidance.
STAR Study Investigating Performance and Safety of the Medical Device SiPore15TMNCT03823027 [9,14,15]Completed in 2019Interventional and Not applicableDiabetesTo evaluate the performance and safety of engineered MSN (SiPore15TM) in reducing blood glucose (HbA1c) in overweight or obese subjects with prediabetes or newly diagnosed type 2 diabetes.Completed: SiPore15 TM significantly reduced HbA1c without affecting body weight, showing no serious adverse effects. Demonstrating its potential in controlling glycaemia.
Table 2. Comparison of the different cytotoxicity assays.
Table 2. Comparison of the different cytotoxicity assays.
AssaysMTT AssayLDH AssayTrypan Blue Exclusion AssayCaspase Activity AssayAnnexin V/PI Staining AssayAlamar Blue AssayCytokinesis Block Proliferation Index and Relative Population Doubling
Features
Type of Cytotoxicity MeasuredMetabolic Activity (Cell Viability)Membrane Integrity (Cell Damage)Membrane Integrity (Cell Damage)Apoptosis (Programmed Cell Death)Cell Death (Early Apoptosis, Late Apoptosis, Necrosis)Metabolic Activity (Cell Viability)Cytostasis (Reduction in growth rate)
Test SystemMammalian cells e.g. human lymphocytes and human hepatocytesMammalian cells e.g. human lymphocytes and human hepatocytesMammalian cells in suspension e.g. human lymphocytes and human hepatocytesMammalian cells e.g. human lymphocytes and human hepatocytesMammalian cells in suspension e.g. human lymphocytes and human hepatocytesMammalian cells e.g. human lymphocytes and human hepatocytesMammalian cells e.g. human lymphocytes and human hepatocytes
PrincipleYellow MTT tetrazolium salt is reduced to an insoluble purple formazan product by mitochondrial dehydrogenases in metabolically active cells.LDH enzyme is released from damaged cells into the culture medium, which is measured as an indication of membrane integrity loss.Living cells, which have an intact membrane, exclude the dye, whilst dead cells with damaged membranes take up the blue dye.Measures the activation of caspase-3 and caspase-9, which are central drivers of apoptosis execution.Annexin V labels early apoptosis (translocated phosphatidylserine). Propidium Iodide labels late apoptosis/necrosis (damaged membrane)Blue resazurin is reduced to pink resorufin by metabolically active cells, producing a colourimetric or fluorescent signal.To measure the average number of nuclear divisions a cell population has completed by scoring the frequency of cell with one, two or three nuclei after the addition of Cyto B to prevent cytokinesis.
AdvantagesHighly sensitive and reproducible. Measures the metabolic activity of living cells. Widely used and well-established.Simple and quick. Detects subtle background membrane damage.Simple, inexpensive, and rapid. Suitable for high-throughput applications. Direct visualisation of viable and non-viable cells.Directly measures apoptosis-specific pathways.
Sensitive detection of early apoptotic events.
Differentiates between early apoptosis, late apoptosis, and necrosis.
Quantitative and qualitative analysis via flow cytometry.
Non-toxic and allows continuous monitoring.
Does not produce insoluble products.
High sensitivity; ideal for high-throughput screening.
Can analyse cytotoxicity along with genotoxicity. Provides a more accurate measure of the average number of cells than simple counting.
DisadvantagesInsoluble formazan crystals require solubilisation.
Metabolic activity may not always correlate with actual cell viability.
Cannot differentiate between necrosis and apoptosis.Does not distinguish between apoptotic and necrotic cells.
May overestimate dead cells due to transient membrane permeability.
Cannot detect necrosis or caspase-independent apoptosis.
May miss late-stage cell death.
Requires precise flow cytometry gating.
May generate false positives under non-apoptotic stress conditions.
Transitional cell states may complicate interpretation (early and late apoptosis).
Metabolic activity may not directly equate to cell viability.
Potential dye interactions with NPs; slower signal development.
Requires optimisation of concentration and exposure time of Cyto B. Labour intensive scoring. Only suitable for dividing cells.
Relevance for PSNs/NPsEasy and quick method for identifying NP cytotoxicity with various doses of test agents at different time points.Assess the membrane integrity damage induced by NPs.Quick and high-volume screening of NP cytotoxicity based on membrane damage.It can be used to investigate the apoptotic pathways triggered by NPs and can determine the underlying mechanism of cell death when combined with ROS or ER stress markers.Distinguishes apoptosis from necrosis and can help identify which pathway is triggered.An alternative to the MTT assay for assessing cell viability, especially if NP interference occurs.Applicable with sequential addition of Cyto B to ensure no interference with NP endocytosis
Table 3. Comparison of the different genotoxicity assays.
Table 3. Comparison of the different genotoxicity assays.
AssaysCytokinesis Block Micronucleus (CBMN) AssayComet AssayChromosome Aberration Testγ-H2AX AssayAmes Test
Features
Type of Damage DetectedClastogenicity (chromosome breakage) and aneugenicity (chromosome loss)DNA single-strand breaks, double-strand breaks and oxidative lesionsStructural chromosomal changes (breaks, deletions and rearrangements)DNA double-strand breaksGene mutations (point mutations). Specifically base- pair substitutions and frameshift mutations.
Test SystemMammalian cells (e.g. human lymphocytes and human hepatocytesMammalian cells (e.g. human lymphocytes and human hepatocytesMammalian cells (e.g. human lymphocytes and human hepatocytesMammalian cells (e.g. human lymphocytes and human hepatocytesBacterial cells (e.g. Salmonella typhimurium and Escherichia coli)
PrincipleDetects chromosome breakage or loss by scoring micronuclei in binucleated cells after cytokinesis is blocked.Measures DNA strand breakage by electrophoretic migration of DNA fragments, forming a ‘comet tail’.Identifies structural or numerical chromosome changes in cells arrested at metaphase after exposure to test agent.Detects DNA double-strand breaks by quantifying phosphorylated histone H2AX foci.Assesses mutagenicity of chemicals by measuring reversion mutations in bacterial strains.
AdvantagesSimple and cost effective. High through-put analysis available to overcome manual scoring.Single-cell analysis possible. Highly sensitive to low levels of DNA damage.Structural and numerical aberrations are detected. Type of damage can be identified.Allows early detection of DNA damage. Quantifiable using immunofluorescence or flow cytometry. Highly sensitive.Quick and cost effective. Suitable for chemical mutagens.
DisadvantagesRequires actively dividing cells. Time-consuming and subjective quantification of micronuclei.Cannot differentiate between clastogenic and aneugenic eventsLabour intensive. Less sensitive to low level or transient DNA alterationsMay detect non-relevant background phosphorylation. Does not detect all types of DNA damage.Not suitable for nanoparticle assessment due to poor nanoparticle penetration into the bacterial cells. May produce false negatives. Cannot be done on its own; needs another assay alongside.
Relevance for PSNs/NPsMeasures DNA damage in daughter cells, which can help assess the long-term genomic instability from NP exposureUseful for detecting early DNA damage by NPs as they can generate ROS or directly interact with DNA causing chromosomal damageDetects chromosome changes caused by NPs, identifying carcinogenic and heritable effects of NP exposureCan provide mechanistic insight into genotoxic stress pathways activated by NPsQuick screening assay to identify mutagenicity alongside mammalian cell-based assays
Table 4. Research questions and current knowledge gaps.
Table 4. Research questions and current knowledge gaps.
CategoryResearch QuestionCurrent Knowledge and Gaps
Sample PreparationWhat is the guidance for sample preparation with advanced nanomaterials which do not dissolve? Can a universal dissolution protocol be established for inter-study comparison?Studies often use different buffers and pH. A single standardised dissolution assay/protocol could allow reproducibility across research and maintain stability.
ManufacturingCan scalable, reproducible synthesis methods be developed?Microemulsion, sol-gel and template methods exist; however, there is batch-to-batch variability. A method is needed to maintain consistent pore size, structure and surface chemistry.
ToxicityCan the silica matrix of PSNs be safely excreted or metabolised? How does its stability influence degradation pathways in biological environments?PSNs undergo biodegradation primarily through excretion via the kidneys. However, the structural stability of the silica matrix, the rate and completeness of degradation that depends on physicochemical properties and long-term bioaccumulation, remains insufficiently understood.
How do PSNs and their protein corona influence immunogenicity and potential immunotoxicity?PSNs may activate the complement system or cytokine release depending on their physicochemical properties. However, comprehensive immunotoxicity studies of PSNs are limited. Standardised immune assays and long-term models are needed to clarify chronic immune effects and safety.
What is the extent of biodistribution/accumulation of PSNs in major organs and tissues?Short-term studies show accumulation in the liver, spleen and lungs with gradual clearance, but data on biodegradation rate and organ-specific accumulation after repeated dosing are limited. Comprehensive pharmacokinetic and chronic exposure studies are required.
What are the long-term safety implications of PSNs in humans?Preclinical studies show acute toxicity is low, but chronic toxicity has not been extensively studied. Standardised safety endpoints for PSNs must be established.
Standardisation and Regulatory ChallengesAre regulatory and standardisation frameworks sufficient for PSN translation? How can PSNs be integrated into regulatory frameworks for medical devices or drug formulation?Regulatory guidance exists for some advanced nanomaterials, but not specifically for PSNs. Standardised characterisation, dissolution testing, toxicity endpoints and manufacturing methods need to be established to meet clinical trial requirements.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Patel, T.; Clipstone, C.; Girija, U.V.; Ahmad, Z.; Singh, N. Nanotoxicity of Porous Silica Nanoparticles: Physicochemical Properties and Mechanistic Cellular Endpoints. Nanomaterials 2025, 15, 1766. https://doi.org/10.3390/nano15231766

AMA Style

Patel T, Clipstone C, Girija UV, Ahmad Z, Singh N. Nanotoxicity of Porous Silica Nanoparticles: Physicochemical Properties and Mechanistic Cellular Endpoints. Nanomaterials. 2025; 15(23):1766. https://doi.org/10.3390/nano15231766

Chicago/Turabian Style

Patel, Trisha, Callum Clipstone, Umakhanth Venkatraman Girija, Zeeshan Ahmad, and Neenu Singh. 2025. "Nanotoxicity of Porous Silica Nanoparticles: Physicochemical Properties and Mechanistic Cellular Endpoints" Nanomaterials 15, no. 23: 1766. https://doi.org/10.3390/nano15231766

APA Style

Patel, T., Clipstone, C., Girija, U. V., Ahmad, Z., & Singh, N. (2025). Nanotoxicity of Porous Silica Nanoparticles: Physicochemical Properties and Mechanistic Cellular Endpoints. Nanomaterials, 15(23), 1766. https://doi.org/10.3390/nano15231766

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

Article Metrics

Back to TopTop