Next Article in Journal
Genome-Wide Identification and Comprehensive Analysis of Ubiquitin-Specific Protease Gene Family in Soybean (Glycine max)
Previous Article in Journal
The Role of Cytokines in Orthodontic Tooth Movement
Previous Article in Special Issue
Characterization and Hemocompatibility of α, β, and γ Cyclodextrin-Modified Magnetic Nano-Adsorbents
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Analyzing Molecular Determinants of Nanodrugs’ Cytotoxic Effects

1
FZU—Institute of Physics of the Czech Academy of Sciences, 182 21 Prague, Czech Republic
2
Faculty of Mathematics and Physics, Charles University, 121 16 Prague, Czech Republic
3
Institute for Clinical & Experimental Medicine (IKEM), 140 21 Prague, Czech Republic
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(14), 6687; https://doi.org/10.3390/ijms26146687
Submission received: 29 May 2025 / Revised: 4 July 2025 / Accepted: 9 July 2025 / Published: 11 July 2025
(This article belongs to the Special Issue Molecular Research on Nanotoxicology)

Abstract

Nanodrugs hold great promise for targeted therapies, but their potential for cytotoxicity remains a major area of concern, threatening both patient safety and clinical translation. In this systematic review, we conducted a systematic investigation of nanotoxicity studies—identified through an AI-assisted screening procedure using Scopus, PubMed, and Elicit AI—to establish the molecular determinants of nanodrug-induced cytotoxicity. Our findings reveal three dominant and linked mechanisms that consistently act in a range of nanomaterials: oxidative stress, inflammatory signaling, and lysosomal disruption. Key nanomaterial properties like chemical structure, size, shape, surface charge, tendency to aggregate, and biocorona formation control these pathways, modulating cellular uptake, reactive oxygen species generation, cytokine release, and subcellular injury. Notably, the most frequent mechanism was oxidative stress, which often initiated downstream inflammatory and apoptotic signaling. By linking these toxicity pathways with particular nanoparticle characteristics, our review presents necessary guidelines for safer, more biocompatible nanodrug formulation design. This extensive framework acknowledges the imperative necessity for mechanistic toxicity assessment in nanopharmaceutical design and underscores the strength of AI tools in driving systematic toxicology studies.

Graphical Abstract

1. Introduction

Nanomedicine holds enormous promise to transform today’s healthcare through its potential to enable targeted therapy, specific diagnostics, and smart drug delivery systems [1,2]. According to the National Institutes of Health, nanomedicine is a branch of nanotechnology designed for highly targeted medical intervention at the molecular level either to cure disease or repair damaged tissue [3]. Using materials and mechanisms on the nanoscale, nanomedicine offers new solutions to complex medical challenges. One of the basic advantages of nanomedicine is the ability to simplify the delivery of therapeutic agents in order to improve their therapeutic activity and minimize undesired side effects [1,2,4,5]. Many active pharmaceutical ingredients (APIs) have poor solubility, decreasing their therapeutic effectiveness and increasing the risk of undesired side reactions [6]. Nanotechnology overcomes this problem through improving drug solubility by encapsulation in lipid or polymer-based carriers, which are commonly referred to as nanocarriers [1,2,4,5]. These systems enhance the bioavailability of drugs, reduce the required dose, lower treatment costs, and reduce toxicity [1,2,4,5,6]. Additionally, nanocarriers also facilitate targeted delivery. By functionalizing the surfaces of nanoparticles (NPs) with targeting ligands, one can target nanodrugs to particular cell types, such as cancer or immune cells. This targeted delivery not only enhances therapeutic specificity but also reduces off-target effects, thereby enhancing patient outcomes [4,7,8].
Given these advantages, engineered nanoparticles have attracted growing attention in biomedical studies and clinical translation. As of 2022, the U.S. Food and Drug Administration (FDA) had approved 23 nanoparticle-based drugs, with many more under investigation in clinical trials [8,9,10]. Most of the approved nanomedicines are founded on developed platforms such as liposomes, micelles, polymeric nanoparticles, and inorganic particles [6,7,8]. Notwithstanding the impressive clinical value demonstrated by these systems, they are not without limitations.
Despite heavy investment and progress—driven in part by initiatives like the National Nanotechnology Initiative—the number of clinically approved nanomedicines is far lower than initially anticipated [6]. One major obstacle is the translational gap between preclinical (especially animal-based) studies and successful human applications [11,12,13]. This gap occurs primarily due to the absence of complete understanding of interspecies variances in physiological and pathological responses, which directly affect how nanomedicines behave in the human body [14]. Some studies state that current preclinical models overestimate treatment effects as they fail to replicate human pathophysiology properly [15]. Besides species differences, patient-to-patient variation (i.e., genetic variation, disease status, and immune status) also prevents clinical translation [6]. Other technical challenges include nanoparticle size and shape heterogeneity, instability under physiological conditions, and long-term safety and cytotoxicity issues [5,10,16,17,18,19]. Variations in nanoparticle properties can undermine reproducibility and therapeutic effectiveness, while chemical and physical instability can impact shelf life and bioactivity. In addition, with the development of nanomedicine comes growing concern over potential toxicological implications, demanding ever greater rigorous investigation of nanoparticle safety [10,16,17,18,19]. Finally, regulatory and clinical framework weaknesses are due to improper biosafety evaluations, poor patient stratification, and the absence of nanomedicine-specific guidelines [20]. Together, the findings explain the complex mechanisms responsible for the absence of correlation between successful animal trials and successful human applications in the progress of nanomedicine [21]. These limitations have had practical consequences. Safety issues and clinical failures have led to the market withdrawal of several nanodrugs [10,19,22,23,24,25,26]. In fact, FDA-approved nanomedicines dropped from over 50 in 2016 to just 23 in 2022 [9,10,27,28]. This decline underscores the pressing need for additional innovation in nanoparticle design, manufacturing scale-up, and safety testing.
One of the most important barriers to broader clinical application is the potential cytotoxicity of nanodrugs [9,10,27,28]. While adverse drug reactions, along with toxicity, are not unique to nanomedicines (they are also prevalent for conventional therapies [29,30,31]) the unique properties of nanoscale materials present some new safety concerns. An understanding of drug toxicity on the cellular and molecular levels is required for the development of safer formulations, the identification of predictive biomarkers, and the reduction in late-stage clinical failures [32]. Nanomaterials can cross cell membranes, biological barriers, and tissue interfaces more easily compared to materials at the larger size scale [6,33]. Extensive biokinetic research, however, suggests that particle size alone may have little influence on overall biodistribution [34]. Further basic understanding of complex interactions between nanomaterials and human biological systems is thus a priority for maximizing clinical efficacy and guaranteeing safety. Although a growing number of reviews cover the general toxicity and safety of nanomaterials [6,35,36,37], there have been relatively few that have systematically discussed cellular and molecular mechanisms of cytotoxicity for clinically approved nanodrugs [2,10]. This review strives to fill this gap by presenting a critical overview of molecular determinants of nanodrug-induced cytotoxicity. By doing so, we aim to gain valuable insights into the determinants of their safety profiles and inform the design of next-generation nanotherapeutics.

2. Materials and Methods

In order to systematically assess the current literature on nanodrug-induced cytotoxicity, we employed a multi-step search strategy combining traditional database searching with AI-driven tools, as well as manual reference list screening. The principal literature search was conducted using the Scopus and PubMed databases. To achieve greater coverage, we manually screened eligible articles’ reference lists, thus allowing us to include supplementary relevant studies that could have otherwise been missed in light of varying keyword indexing. To supplement the depth and efficiency of our review, we incorporated Elicit AI (https://elicit.com, accessed on 5 May 2025), an artificial intelligence-based research assistant built on top of the Semantic Scholar corpus (https://www.semanticscholar.org/, accessed on 5 May 2025), which includes over 126 million scholarly publications. Elicit AI was specifically programmed to identify research on nanodrug-induced cytotoxicity. This yielded 498 papers initially, which were subsequently screened based on predetermined inclusion criteria. A complete list of these studies is given in the Supplementary Materials (Table S1).
The entire selection, screening, and data extraction workflow is summarized in Supplementary Materials (Figure S1), which presents a flowchart constructed in accordance with systematic reviews guidelines. The figure distinctly describes each phase of the review process—ranging from initial identification of records to final inclusion—marking methodological rigor and reproducibility. We applied the following inclusion criteria to screen articles retrieved by Elicit AI:
  • Investigated FDA-approved or clinically used nanodrugs in therapeutic uses.
  • Addressed molecular mechanisms of cytotoxicity induced by nanodrugs (e.g., protein corona formation, oxidative stress, inflammation, lysosomal damage).
  • Provided experimental evidence (in vitro or in vivo) and not solely theoretical or computational.
  • Provided a comprehensive toxicity or safety profile, such as dose–response curves or IC50 values.
  • Analyzed nanodrug metabolism, degradation, or biodistribution in biological systems.
  • Focused on therapeutic applications, excluding environmental or non-medical studies.
  • Were original research articles or systematic reviews/meta-analyses.
  • Addressed molecular-level toxicity mechanisms, beyond drug delivery efficacy.
Each publication was evaluated holistically against these criteria, with inclusion decisions based on overall relevance and scientific merit. To prioritize studies for in-depth analysis, Elicit AI was used to assign a relevance score to each publication. These scores, presented in Supplementary Materials (Table S2), reflect how well each study aligned with the selection criteria. Applying a cutoff score of ≥3.5, we identified 98 high-priority articles for detailed evaluation.
For all these 98 studies, Elicit AI facilitated the quantitative and qualitative data extraction on nanomaterial cytotoxicity. Data extracted were:
  • Toxicity rates or IC50 values.
  • Dose-dependent toxicity responses.
  • Molecular mechanisms (e.g., oxidative stress, inflammatory signaling, lysosomal disruption).
  • Affected cellular structures (e.g., mitochondria, lysosomes, membrane integrity).
  • Biomarkers of oxidative stress and inflammation.
  • Observed immunomodulatory effects.
In reports describing several pathways of toxicity, Elicit AI was instructed to order mechanisms by reported rank or frequency of citation. If no explicit molecular mechanisms were reported, the report was labeled as “No specific molecular mechanisms reported.”
To ensure consistent and targeted data extraction, Elicit AI was guided using the following screening questions:
Therapeutic Nanodrug Focus: Does the study investigate FDA-approved or clinically used nanodrugs and their cytotoxic effects at the molecular level?
Molecular Interaction Analysis: Are molecular-scale interactions (e.g., protein corona formation) between nanomaterials and cells examined?
Experimental Validation: Is the research based on empirical (in vivo or in vitro) data rather than computational or theoretical models?
Safety Assessment: Is a comprehensive toxicity or safety profile provided?
Biological Fate: Does the study examine nanodrug degradation, metabolism, or biodistribution in biological systems?
Study Type Relevance: Is the context therapeutic (medical) rather than environmental or non-clinical?
Evidence Synthesis: Is the publication an original research article or a systematic review/meta-analysis?
Mechanism Analysis: Does the study focus on molecular mechanisms of cytotoxicity rather than drug delivery efficiency alone?
This structured, AI-assisted methodology ensured a robust, unbiased, and reproducible selection and analysis of literature addressing nanodrug-induced cytotoxicity.
During manuscript preparation, ChatGPT 4o (OpenAI) was used for grammar refinement, academic style enhancement, English translation, and consistency checks. All content generated or edited with AI assistance was thoroughly reviewed and revised by the authors. The authors accept full responsibility for the scientific content, interpretation, and integrity of the work. It is important to note that AI tools, including Elicit AI Pro and ChatGPT 4o, do not meet the criteria for authorship and are therefore not listed as co-authors. The authors maintain full accountability for the originality, accuracy, and scholarly validity of this manuscript.
It is important to acknowledge several limitations of this study. Primarily, the analysis is based on a qualitative synthesis of cytotoxicity data, which restricts the ability to identify consistent toxicity trends or establish clear dose–response relationships across studies. Our objective, however, was to provide a broad overview of the cytotoxicity associated with nanodrugs and to highlight common molecular mechanisms underlying these effects. Furthermore, the study incorporates findings from both in vitro and in vivo models, which vary in their physiological relevance to human biology. While this approach offers a broader perspective, it also introduces variability that may limit the generalizability of the findings. In addition, the study intentionally excludes data related to environmental or non-medical nanotoxicity—such as inhalation risks from industrial nanomaterials. While this may omit certain mechanistic insights relevant to human exposure in broader contexts, the exclusion was deliberate in order to maintain a focused analysis on the pharmaceutical applications of nanomaterials.

3. Types of Nanodrugs Approved for Clinical Applications

Prior to providing an overview of molecular mechanisms involved in the possible nanodrug-induced cytotoxicity of nanodrugs used in the clinic, let us first define a “nanodrug” and provide an overview of different classes of existing nanodrugs utilized for clinical use. We start with definitions of the key terms “nanoparticle,” “nanomedicine,” and “nanodrug.” A review of the literature indicates that some of these terms are differently defined in the scientific community and the regulatory arena [10,38,39]. In the majority of the scientific literature, nanoparticles are mainly characterized based on their size and related physicochemical characteristics [10,38,39]. While some (mostly referring to regulatory documents) consider nanoparticles to be 1–100 nanometers in size, others consider the size limit up to 1000 nanometers [10,38,39]. Notably, the European Medicines Agency (EMA) considers the size range of nanoparticles as 0.2–100 nm [40]. Conversely, the term nanomedicine is broadly defined in terms of function, rather than dimension [39,41,42]. It is utilized to refer to the application of nanotechnology for diagnosing disease, treating disease, and preventing disease. The definition of nanomedicine also depends on context. In certain contexts, it includes the therapeutic application of nanomaterials—including diagnostic, therapeutic, and monitoring applications [39]. In more general contexts, it is referred to as a multi-disciplinary science that utilizes nanotechnology to enable new imaging, diagnosis, treatment, tissue repair, and regeneration of biological systems [41]. Likewise, the term nanodrug is not standardized. Most commonly, it is used to describe a drug product that utilizes nanoscale materials (i.e., nanoparticles or nanocarriers) to improve drug delivery, efficacy, and safety [10,38,39].

3.1. Defining the Terminology of Nanodrugs

It is interesting to point out here that the FDA has not adopted official regulatory definitions of the terms “nanotechnology,” “nanomaterial,” or “nanoscale” [43]. The FDA did suggest, however, a working definition for products involving the application of nanotechnology. The product uses nanotechnology, according to the agency, if it matches two criteria:
  • “a material or end product is engineered to have at least one external dimension, or an internal or surface structure, in the nanoscale range (approximately 1 nm to 100 nm);
  • a material or end product is engineered to exhibit properties or phenomena, including physical or chemical properties or biological effects, that are attributable to its dimension (s), even if these dimensions fall outside the nanoscale range, up to one micrometer (1000 nm)” [43].
This definition highlights both structural properties and nanoscale-dependent functional properties, with flexibility to accommodate emerging nanotechnologies in pharmaceutical and biomedical applications. The following are the implications of this definition. Firstly, the drug product has to be deliberately designed to possess certain dimensions or to have special properties [43]. Secondly, it must possess at least one external dimension, or an internal or surface structure, in the nanoscale range (roughly 1 nm to 100 nm) [43]. Thirdly, the product should possess attributes or phenomena (physical, chemical, or biological effects, for example) that can be attributed to its nanoscale size [43]. Finally, even end products or materials with sizes beyond the conventional nanoscale range (up to 1000 nm) can be referred to as nanotechnology-based if the nanoscale-like properties are directly attributable to their engineered size [43].
From this analysis, we suggest a working definition of a nanodrug by combining the FDA [43] definition of nanotechnology products and the National Cancer Institute definition of a drug [44]. We thus define a nanodrug as: A substance that involves the application of nanotechnology—as defined by the FDA—and is aimed at use in the prevention, diagnosis, treatment, or mitigation of the symptoms of a disease or abnormal physiological condition.
We find it necessary to have a concise definition of a nanodrug to start with. Such a definition offers a shared paradigm within which the systematic and unbiased determination of the pharmaceutical products that have been approved so far, and which encompass nanotechnology can be conducted. It also allows for the categorization of approved nanodrugs by the type of nanomaterials incorporated in their formulations.
Bearing in mind the terminology of nanodrug as defined above, we systematically searched the literature to find clinically approved nanodrugs [4,5,6,9,10,26,28,45,46,47]. We found that 25 approved drugs meet the criteria laid down by our definition. Additionally, approximately 10 other nanodrugs have been approved on a national scale in single countries—for example, CosmoFer and Monofer, which have been approved in certain member states of the EU [4,5,6,9,10,26,28,45,46,47]. However, for the purpose of analysis, we considered only nanodrugs approved by either FDA or EMA. The latter are presented in Table 1.
Our analysis (Table 1) clearly demonstrates that lipid-based, polymer-based, and inorganic NPs are the most commonly used nanocarriers in the formulation of clinically approved nanodrugs (Figure 1). Their structural and functional flexibility makes them capable of delivering a wide range of therapeutic molecules, including small-molecule drugs, nucleic acids, and antibodies [4,5,6,9,10,26,28,45,46,47].
These nanocarriers (Figure 1) enhance conventional therapies by enabling more targeted and efficient delivery, thereby enhancing therapeutic efficacy and minimizing potential side effects [4,5,6,9,10,26,28,45,46,47]. Notably, they break biological barriers, improve the solubility of poorly water-soluble drugs, and prolong blood circulation time [4,5,6,9,10,26,28,45,46,47]. Thanks to these advantages, these nanocarriers are applied in many therapeutic areas with exceptional significance in cancer treatment and the management of chronic diseases [4,5,6,9,10,26,28,45,46,47]. Next, we briefly describe each category of nanocarriers.

3.2. Lipid-Based NPs

Lipid-based NPs are a prominent class of drug delivery systems composed primarily of lipids that self-assemble into nanoscale structures. They play an important role in drug delivery applications [4,9,28], as well as in gene therapy involving nucleic acids such as mRNA, siRNA, and DNA [4,5,6,9,10,26,28,45,46,47]. These NPs are typically spherical and consist of at least one phospholipid bilayer surrounding an aqueous core. Lipid-based NPs are widely utilized due to their many advantages in biological systems, including excellent biocompatibility and biodegradability, high drug-loading capacity, and a broad range of tunable physicochemical properties [65]. Moreover, they can reduce systemic toxicity while enhancing the efficacy and specificity of therapeutic agents [4,5,6,9,10,26,28,45,46,47]. As a result, lipid-based nanoparticles represent the most commonly used class among clinically approved nanomedicines (Table 1) [4,5,6,9,10,26,28,45,46,47].
However, these systems are not without limitations. They can trigger immune responses [66,67], and have been associated with increased rates of programmed cell death and potential degradation of nucleic acids such as DNA [68], which may be undesirable depending on the therapeutic context.
Liposomes are a well-established and extensively studied subclass of lipid-based NPs. They were first described in the 1960s [9,27,50] and remain among the most versatile drug delivery platforms [69]. Structurally, liposomes are unilamellar or multilamellar vesicles that mimic biological cell membranes. The phospholipid bilayer allows for the encapsulation of hydrophobic drugs, while the aqueous core is suitable for hydrophilic compounds [70]. Furthermore, the aqueous interior can be used to carry other therapeutic or diagnostic agents such as nucleic acids, proteins, or imaging compounds [71,72]. Several factors influence liposome stability, including particle size, shape, surface charge, lipid composition, and surface characteristics. Surface modifications, such as PEGylation, reduce opsonization and help prevent rapid clearance by the reticuloendothelial system, thereby extending circulation time and enhancing drug delivery efficiency [4,71,72]. Thanks to their versatility and established safety profiles, liposomes are widely used in clinical applications, including as carriers for chemotherapeutics, antibiotics, and gene therapy agents [4,71,72,73].
Lipid nanoparticles (LNPs) are a newer generation of lipid-based delivery systems, particularly suited for nucleic acid therapeutics [74]. Unlike liposomes, LNPs are typically non-lamellar and possess a micellar internal structure. Their formulation usually consists of four primary components: (1) a cationic or ionizable lipid for complexation with negatively charged nucleic acids, (2) phospholipids to maintain structural integrity, (3) cholesterol to improve particle stability and facilitate membrane fusion, and (4) PEGylated lipids to enhance circulation time, reduce aggregation, and control particle size [75]. LNPs are highly effective for nucleic acid delivery and can be engineered for targeted transfection [76,77]. Nevertheless, they face some limitations, including a generally lower loading capacity compared to liposomes and challenges in achieving optimal biodistribution. In particular, LNPs may accumulate in the liver and spleen, which could limit their therapeutic applications [10,74].

3.3. Polymer-Based NPs

Polymeric NPs are nanoscale drug delivery systems synthesized from natural or synthetic polymers [4,5,6,9,10,26,28,45,46,47]. Widely applied in medicine and pharmaceuticals, they are used for targeted drug delivery, cancer therapy, gene therapy, and diagnostics [4,5,6,9,10,26,28,45,46,47]. Their appeal lies in their biocompatibility, biodegradability, and formulation flexibility, which allows for diverse structural configurations and physicochemical properties [4,5,6,9,10,26,28,45,46,47]. This flexibility enables targeted delivery, controlled release, and surface modification, making polymeric NPs ideal for co-delivery applications [78] and precise control over drug loading and release kinetics [79]. Therapeutic agents can be incorporated via multiple strategies: encapsulation, matrix entrapment, chemical conjugation, or surface adsorption [4,5,6,9,10,26,28,45,46,47]. However, limitations include potential toxicity, nanoparticle aggregation, and the formation of toxic degradation by-products [4,5,6,9,10,26,28,45,46,47].
Several methods are available to tailor polymeric NP characteristics for specific applications. Emulsification involves dissolving a polymer and drug in a volatile organic solvent (e.g., dichloromethane), then emulsifying it into an aqueous phase [80,81]. Solvent evaporation leads to the formation of solid NPs, often used to encapsulate hydrophobic drugs. Nanoprecipitation entails dissolving the polymer and drug in a water-miscible solvent (e.g., acetone) and adding it to water under stirring, leading to spontaneous nanoparticle formation [82]. This method is efficient for small molecules and hydrophobic drugs. Ionic gelation is primarily used with natural polymers like chitosan [83,84]. The polymer is dissolved in water and mixed with a crosslinking agent such as tripolyphosphate, forming nanoparticles suitable for encapsulating biologically active compounds [83,84]. Microfluidics offers precise control over formulation by manipulating fluid streams in microchannels [85,86]. This technique enables high reproducibility and customization of nanoparticles.
Polymeric NPs are available in several structural forms, each suited to specific therapeutic goals, e.g., nanospheres, nanocapsules, dendrimers, micelles, polymersomes, nanohydrogels [4,5,6,9,10,26,28,45,46,47]. Nanospheres represent solid matrix systems in which drugs are uniformly dispersed, offering sustained or controlled release [4,5,6,9,10,26,28,45,46,47]. Nanocapsules feature a central core containing the active agent, surrounded by a polymeric shell [4,5,6,9,10,26,28,45,46,47]. These are especially effective for protecting sensitive drugs and enabling targeted release. Dendrimers are highly branched macromolecules with a central core and layered branches [87]. They offer multiple surface sites for attaching drugs, ligands, or imaging agents, and are widely used for delivering nucleic acids and small molecules. Dendrimers allow precise control over size, shape, molecular weight, and surface chemistry [88]. Micelles are self-assembled structures from amphiphilic block copolymers, consisting of a hydrophobic core and hydrophilic shell [89]. Ideal for encapsulating poorly water-soluble drugs, they improve bioavailability and efficacy [4,5,6,9,10,26,28,45,46,47]. Polymersomes are vesicle-like carriers composed of block copolymers, structurally similar to liposomes but with greater stability and loading capacity [90]. They are especially suited for dual drug delivery, as hydrophobic drugs can be integrated into the membrane and hydrophilic agents encapsulated in the core [91,92]. They also demonstrate efficient cytosolic delivery [93]. However, clinical translation remains limited due to production complexity and potential toxic by-products [94]. Nanohydrogels are three-dimensional hydrophilic polymer networks that swell in water [95]. They offer excellent biocompatibility and are well-suited for the controlled release of biomolecules, with promising applications in scaffold development and wound healing [96].

3.4. Inorganic NPs

Inorganic NPs are composed of materials such as metals (e.g., gold), metal oxides (e.g., iron), semiconductors (e.g., quantum dots), or silica [97]. Their main advantage lies in their tunable sizes, structures, and geometries, as well as their distinct optical, magnetic, electrical, and physical properties. These unique characteristics make inorganic NPs ideal for multimodal, stimuli-responsive, and targeted imaging applications [98]. Depending on their core composition, inorganic NPs serve different purposes. Metallic and silica NPs are commonly explored for drug delivery, thanks to their structural stability and high potential for surface functionalization [97]. Metal oxide NPs, particularly iron oxides, are used as magnetic resonance imaging (MRI) contrast agents due to their superparamagnetic properties, though many have been withdrawn from the market for safety or efficacy reasons [26,97].
To date, iron oxide nanoparticles remain the only FDA-approved inorganic NPs for clinical use (Table 1). Despite their potential, inorganic NPs face significant barriers to clinical translation, including low solubility, oxidative stress induction, and genotoxicity or cytotoxicity linked to heavy metal content [10,26,99]. Interestingly, in addition to iron oxide NPs, the EMA has recently approved hafnium oxide NPs for the treatment of squamous cell carcinoma (see Table 1) [64,100].
Generally, metal nanoparticles (e.g., gold, silver, platinum, and copper) interact strongly with light, making them valuable in diagnostics and imaging [4,5,6,9,10,26,28,45,46,47]. Their surfaces can be easily functionalized with biomolecules, enabling targeted delivery [4,5,6,9,10,26,28,45,46,47]. Gold nanoparticles are the most extensively studied, and can be synthesized in various morphologies, including nanospheres, nanorods, nanostars, nanoshells, and nanocages [101]. Their size and shape play a critical role in determining both their optical behavior and biological interactions [102]. Gold nanoparticles show good stability in biological environments, are generally non-toxicity and chemically inert [101]. Strong affinity for functional groups like thiols (-SH) and amines (-NH2), enabling conjugation with polyethylene glycol (PEG), proteins, or drugs [101]. However, despite their promise, no gold NP-based nanodrugs have received full regulatory approval, primarily due to unresolved issues regarding long-term biodistribution, clearance, and potential gene-related toxicity [103]. Nonetheless, several formulations are currently in clinical trials [104,105,106].
In contrast, iron oxide NPs-typically composed of magnetite (Fe3O4) or maghemite (Fe2O3)—are more broadly represented among FDA-approved nanomedicines [4,5,6,9,10,26,28,45,46,47]. Their approval largely stems from their use in treating iron deficiency in conditions such as anemia and chronic kidney disease (CKD) (Table 1), as their iron content is metabolizable by the human body. Iron oxide NPs are superparamagnetic, making them well-suited for MRI [107]. Additionally, they are being explored for drug delivery, thermal therapies (due to their ability to produce localized heating under an external magnetic field) and antimicrobial applications (although research is still ongoing, and evidence remains preliminary) [108,109].

4. General Cytotoxic Characteristics of Nanomaterials

The interactions between nanomaterials and biological systems are governed by a complex interplay of intrinsic physicochemical properties that are fundamental to the design and function of the nanomaterials.
Key among these is the chemical composition of the nanoparticle core, the presence of trace impurities, and the specific strategies used for surface functionalization (Figure 2). Surface characteristics (including surface charge, hydrophobicity, and the attachment of targeting ligands) further modulate the biological identity and reactivity of nanomaterials. Apart from the surface properties, size and shape are also critical determinants of the biological activity of nanoparticles and influence cellular uptake, biodistribution, and clearing mechanisms. Nanomaterials agglomerate in biological environments as well, greatly altering their effective size and surface area, and consequently their interactions with cells and tissues (Figure 2). Another key factor is biomolecular corona formation, an evolving layer of protein and other biomolecules that adsorbs onto the nanoparticle surface when exposed to biological fluid (Figure 2). The corona remodels the surface properties of the nanoparticle and is essential for mediating its detection and processing by the immune system and other cell components. Taken together, these characteristics determine the nano-bio interface and hence the pharmacokinetics, biodistribution, cellular interactions, and potential toxicological impact of nanomaterials on biological systems.
To comprehensively evaluate the toxic potential of clinically approved nanodrugs, it is crucial first to develop a clear idea of the general principles of nanomaterial-induced cytotoxicity. This means critically examining the physicochemical properties and biological processes that govern the cellular responses towards nanomaterials. In this regard, this section will present an overview of the most significant factors (such as particle size, shape, surface charge, composition, and functionalization) engaged in their cytotoxicity. In outlining these basic components, we wish to create the correct context for the interpretation of toxicity profiles evidenced in clinical application.

4.1. Cytotoxic Effects Determined by Nanomaterial Chemical Composition

The chemical composition of nanomaterials is a key determinant of their toxicity, as it governs critical properties such as solubility, redox activity, ionization behavior, and interactions with biological macromolecules [6]. These characteristics collectively shape their toxicological profile and help elucidate the mechanisms by which nanomaterials may induce adverse biological effects [6]. For instance, studies on zinc oxide nanoparticles (nano-ZnO) have demonstrated embryotoxic effects, primarily due to their dissolution into toxic Zn2+ ions [110]. Indeed, a recent comprehensive study evaluated the toxicity of 29 rare earth oxide and transition metal oxide nanoparticles in liver cells [111]. The results showed that pro-oxidative transition metal oxide NPs induced apoptosis in hepatocytes, while rare earth oxide NPs triggered pyroptosis in Kupffer cells and other macrophages [111]. These distinct cell death pathways are likely driven by differences in solubility, redox behavior, and catalytic activity among the nanoparticles [111,112]. However, a common feature underlying the toxic response was the excessive generation of reactive oxygen species (ROS) [111]. Depending on their capacity to induce ROS, nanoparticles triggered distinct cytotoxic pathways [111].
While certain nanomaterials (such as copper oxide, silver NPs, and quantum dots) pose toxicity risks due to the release of harmful metal ions [113], others, like gold NPs, demonstrate significantly greater biocompatibility [101]. For instance, gold NPs have been shown to be non-toxic to RAW264.7 macrophages and can even reduce ROS levels, suggesting potential anti-inflammatory properties [114]. In contrast, soluble metallic nanoparticles tend to increase ROS generation and cytotoxicity due to their ion-releasing behavior [111]. Non-metallic nanomaterials, such as mesoporous silica nanoparticles, generally exhibit lower toxicity [6]. This is largely attributed to the presence of surface silanol groups that modulate interactions with biological molecules [115]. Similarly, liposomes—due to their excellent biocompatibility—have been successfully employed in clinical settings as drug carriers, enhancing therapeutic efficacy while minimizing side effects [4,5,6,9,10,26,28,45,46,47].
Furthermore, impurities also play a critical role in determining nanomaterial toxicity. For instance, residual metal catalysts in carbon nanotubes (CNTs) can enhance ROS production, contributing to adverse biological effects [6,116]. Similarly, gold nanoparticles synthesized with cetyltrimethylammonium bromide (CTAB) can be toxic if residual CTAB is not adequately removed [117]. Organic contaminants, particularly endotoxins like lipopolysaccharides (LPS), are another source of concern [6]. LPS are resistant to standard sterilization and can activate Toll-like receptor 4 (TLR4) on immune cells, triggering the release of pro-inflammatory cytokines [6]. Therefore, accurate nanotoxicity assessment must include rigorous screening and removal of LPS to avoid misinterpretation of inflammatory potential [118,119].

4.2. Cytotoxic Effects Determined by Nanomaterial Size

Particle size is another determinant in the interaction between NPs and biological systems [120,121,122]. It influences not only cellular uptake but also distribution, diffusion, and biological reactivity [123]. Smaller NPs have a higher surface area-to-volume ratio, enhancing their catalytic activity and interaction with biological environments [124,125]. For instance, gold NPs of 10-16 nm can penetrate the cytoplasm, while particles <6 nm may even reach the cell nucleus, increasing potential toxicity [126].
Although NPs around 50 nm are considered optimal for intracellular uptake, both smaller (15–30 nm) and larger (70–240 nm) particles tend to show reduced internalization [127]. A consensus has now been reached regarding the primary mechanisms by which NPs enter cells [128]. There are five major pathways: phagocytosis, macropinocytosis, clathrin-mediated endocytosis (CME), fast endophilin-mediated endocytosis (FEME), and clathrin-independent carrier/glycosylphosphatidylinositol-anchored protein-enriched early endocytic compartment (CLIC/GEEC) endocytosis [128]. CME, FEME, CLIC/GEEC pathways typically internalize NPs in the 60–100 nm range, while macropinocytosis and phagocytosis accommodates larger particles >250 nm via large vesicles (0.5–1.5 μm) [128,129,130]. In addition to size, surface characteristics such as hydrophobicity, ligand presence, and the cell type also influence uptake [128,129,130]. For example, Herceptin-conjugated gold NPs (40–50 nm) demonstrated enhanced cellular internalization compared to both smaller (2–10 nm) and larger (80–100 nm) particles, underscoring the role of ligand density, binding affinity, and membrane wrapping in cellular uptake [131].
The ideal NP size for biomedical applications generally ranges from 10 to 100 nm, with ~50 nm being frequently used for drug delivery due to efficient uptake and favorable pharmacokinetics [132,133]. Size also plays a pivotal role in systemic and organ-specific responses [132,133]. For instance, particles between 10 and 50 nm can be efficiently cleared by the kidneys, whereas larger particles are more likely to accumulate in organs like the liver, spleen, or lungs [26,132,133,134].
Toxicological studies confirm that smaller NPs, such as 20 nm TiO2, induce stronger inflammatory responses in the lungs compared to larger particles (250 nm), largely due to their greater surface area and reactivity [135,136]. Furthermore, particles <100 nm are more prone to tissue accumulation, leading to potential oxidative stress, inflammation, and organ toxicity [6,137]. Recent findings also suggest that prolonged exposure to small NPs may cause histopathological changes, particularly in the liver [6,10,26]. Notably, silica NPs exhibited higher hepatotoxicity compared to silver or zinc oxide NP composites [138,139].

4.3. Cytotoxic Effects Determined by Nanomaterial Shape

NPs exhibit a variety of shapes—such as nanospheres, nanotubes, nanosheets, nanowires, and nanocubes—which significantly influence their toxicokinetics and cytotoxicity [6,140]. Shape affects cellular uptake, cytotoxicity, biodegradation, and organ-specific toxic effects [6]. Computational simulations suggest that nanospheres are internalized most efficiently via endocytosis, followed by nanocubes, nanotubes, and nanodiscs, likely due to the lower membrane deformation energy required by highly symmetrical structures [121,141]. Experimental studies on gold NPs support this, showing that nanospheres result in the lowest cell viability, with prismatic gold NPs slightly less toxic [6,123]. Cubic and rod-shaped gold NPs demonstrated similar bioavailability across a size range of 10–100 nm [123]. Despite reduced internalization, some gold NPs induced notable cytotoxicity due to mechanical damage from sharp edges [6,142].
In mesoporous silica NPs, adjusting the aspect ratio of rod-shaped particles has been shown to decrease hepatic accumulation and enhance renal clearance, regardless of administration route [6]. While oral administration of mesoporous silica NPs typically results in minimal organ toxicity, shape-dependent nephrotoxicity remains a concern and may cause significant kidney injury [143,144].
It is worth noting here that particle shape also influences the exposure of crystallographic planes, biocorona formation, and catalytic activity—all factors contributing to the overall toxicity profile [6,145].

4.4. Cytotoxic Effects Determined by Nanomaterial Surface Properties

Nanomaterials interact with biological systems primarily through their surface, making surface characteristics an important determinant of their behavior and toxicity [145]. Critical surface properties include charge, hydrophobicity, and the presence of specific functional groups or atoms [6,145]. In biomedical applications, NPs are frequently modified post-synthesis to enhance colloidal stability, biocompatibility, safety, and pharmacokinetics [6,145]. These modifications significantly influence the nanomaterials’ interfacial behavior and toxicological profile [145,146]. For instance, rendering NP surfaces hydrophilic and electro-neutral can improve their ability to traverse mucus barriers by reducing interactions with negatively charged, hydrophobic mucin fibers [6,145,147]. In contrast, hydrophobic and positively charged NPs more effectively penetrate epithelial barriers [6,145,147]. Therefore, surface properties must be tailored to match the target biological environment for optimal absorption and delivery [6,145,147,148].
Surface features also influence biodistribution and excretion by affecting protein interactions, biocorona formation, and colloidal stability of NPs [6,149]. For example, PEG-coated gold NPs show enhanced stability in the bloodstream and more uniform tissue distribution [4,6,101,114,149]. Conversely, polyethyleneimine (PEI)-coated gold NPs tend to aggregate and are rapidly cleared by the mononuclear phagocyte system (MPS), leading to accumulation in the liver and spleen and reduced systemic availability [4,6,101,114,149].
Surface charge also affects cellular uptake and intracellular trafficking of NPs [6,123]. Positively charged NPs are internalized more readily due to strong electrostatic interactions with the negatively charged cell membrane [150,151]. Surface ligands further enhance targeting by directing NPs to specific cell types or subcellular compartments [152,153]. Additionally, surface coatings can modulate oxidative stress executed by NPs. For example, silver NPs coated with PEG or bovine serum albumin (BSA) generate fewer ROS and exhibit reduced cytotoxicity then noncoated NPs [154,155].

4.5. Cytotoxicity Driven by Aggregation/Agglomeration

Nanomaterial aggregation and agglomeration is another factor influencing cytotoxicity, with emphasis on effects on biodistribution and cellular uptake [6]. Generally, agglomerates are defined as loosely bound particles and aggregates—as chemically or tightly bound particles [6]. In fact, the toxicological behavior of aggregates and agglomerates is not yet fully understood and warrants further investigation [156,157,158].
Agglomerated nanomaterials may enter cells via different internalization pathways compared to individual nanoparticles, leading to altered interactions with cellular receptors and membrane proteins [159]. Both aggregates and agglomerates have demonstrated notable cytotoxic potential in biological systems [158,159,160]. The biological effects of aggregated or agglomerated nanomaterials are closely tied to their physicochemical characteristics, which shape their interactions with cells, proteins, and tissues, ultimately influencing their toxicological outcomes [110,158,159,160,161].

4.6. Impact of Biocorona Formation on Cytotoxicity

The protein corona, or biocorona, forms rapidly when nanoparticles come into contact with biological fluids, resulting in the adsorption of various biomolecules onto their surface [162,163,164,165]. This dynamic layer redefines the NP’s biological identity and significantly alters its interactions with cellular systems [162,163,164,165]. The biocorona critically influences the biokinetics and biological fate of nanoparticles, including their biodistribution and clearance [162,163,164,165,166,167]. For example, glucose-coated iron oxide NPs preferentially accumulate in the liver, while PEG-coated counterparts demonstrate broader tissue distribution [6,162,163,164,165,166]. These differences stem from variations in biocorona composition, affecting degradation pathways and macrophage-mediated uptake [6,162,163,164,165,166].
At the cellular level, the biocorona reduces nonspecific interactions with membranes, preserving membrane integrity and modifying internalization routes [6,162,163,164,165,166]. Unlike bare nanoparticles, coronated NPs are less likely to disrupt the lipid bilayer directly, which mitigates membrane damage and can improve biocompatibility [168,169]. Moreover, the biocorona plays a pivotal role in modulating cytotoxicity by altering NP bioavailability, immune recognition, and intracellular interactions [170,171,172]. It can either mask or expose reactive surface sites, influencing the extent of oxidative stress, inflammation, or augment cell death execution triggered by NPs [6,162,163,164,165,166]. In summary, the formation of the protein corona is one of the main determinants of NPs behavior in biological systems, governing not only cellular uptake mechanisms but also the safety and efficacy of nanomaterials in biomedical applications.

4.7. Effects of Degradation and Metabolization Products on Cytotoxicity

Other players that largely influence cytotoxicity of nanomaterials are the degradation of nanomaterials and their metabolic byproducts, along with the long-term implications of their biotransformation in biological systems [6]. Once internalized by cells, nanomaterials are subjected to harsh biochemical environments, notably enzymes in hepatic cells and within the acidic, oxidative, and ion-rich environment of lysosomes—particularly in cells of the MPS [10,26,134]. These compartments are rich in enzymes, acids, oxidative agents, and metal ions, all of which facilitate the breakdown or transformation of nanomaterials [10,26,134,172,173,174].
The extent, rate, and pathway of nanomaterial degradation significantly influence their toxicological profile [6,10,26,134,172,173,174]. These processes alter the materials’ physicochemical characteristics (including size, surface chemistry, and solubility) which in turn affect their biodistribution, cellular uptake, and interaction with biomolecules [6,10,26,116,134,172,173,174]. Importantly, degradation products themselves may exhibit distinct or enhanced toxic effects, depending on their chemical composition, reactivity, and accumulation in target tissues [6,10,26,116,134,172,173,174]. For instance, mesoporous silica NPs with a lower aspect ratio degrade more rapidly in simulated body fluids, leading to increased systemic absorption, preferential hepatic accumulation, and severe renal toxicity following oral exposure [143].
One of the most extensively studied cases of degradation-driven toxicity involves silver NPs [6]. Their primary toxic mechanism is linked to ion release through lysosomal dissolution [6,139,154,175]. In the acidic environment of lysosomes, silver NPs readily dissolve into Ag+ ions [6,139,154,175]. This dissolution disrupts lysosomal membrane integrity, resulting in leakage of both silver ions and lysosomal enzymes into the cytoplasm [6,139,154,175,176]. Once in the cytosol, Ag+ ions contribute to mitochondrial dysfunction, oxidative stress, and apoptosis induction [6,139,154,175,176,177]. Furthermore, these ions can react with organic acids to form Ag-O-complexes, which may subsequently convert into Ag-S-species through interactions with thiol-containing proteins, such as those containing cysteine residues [6,139,154,175,176,177]. These chemical transformations exacerbate mitochondria-mediated apoptosis and influence the intracellular retention or excretion of silver [6,139,154,175,176,177,178].
Understanding nanomaterial degradation is essential for predicting long-term biological effects, especially for materials designed for chronic exposure or therapeutic use [6]. Accumulation of toxic byproducts, organ-specific degradation kinetics, and incomplete clearance can all contribute to persistent cytotoxicity or delayed onset of adverse outcomes [6,10,26]. Therefore, both the parent nanomaterial and its degradation/metabolic products must be considered when assessing safety and biocompatibility [6].

5. Defining Molecular Determinants of Nanodrugs’ Cytotoxic Effects

Having defined the term nanodrug, outlined the major categories of clinically approved nanodrugs, and examined the general cytotoxic properties of nanomaterials, we proceeded to conduct a systematic literature search, screening, and data extraction focused on nanodrugs (using the definition provided in Section 3.1) and their associated cytotoxicity. To assist in this process, we employed Elicit AI Pro, an AI-powered research tool, to identify and extract relevant molecular determinants underlying nanodrug-induced cytotoxic effects. A comprehensive description of how Elicit AI Pro was utilized for data extraction and analysis is provided in Section 2.

5.1. Key Molecular Pathways Determining Cytotoxicity of Nanodrugs

Using Elicit AI Pro, we analyzed a broad range of studies that consistently highlight several key molecular mechanisms driving nanoparticle-induced cytotoxicity. These mechanisms are often interconnected and may trigger cascading cellular effects, amplifying toxic outcomes. A summary of the principal molecular pathways identified as contributors to nanoparticle-induced cytotoxicity is presented in Table 2.
It is worth noting that our analysis revealed that nanodrug platforms exhibit formulation-dependent cytotoxicity profiles, with active targeting strategies generally outperforming passive approaches in terms of tumor selectivity and therapeutic efficacy [6,10,35,110,138,142,179,184,188]. Active targeting uses receptor-mediated endocytosis, enabling enhanced uptake of nanocarriers by tumor cells and facilitating drug delivery at lower concentrations [6,10,35,110,138,142,179,184,188]. Clinical studies of approved nanodrugs have revealed notable differences in cytotoxicity and molecular interaction profiles across formulations [6,10,35,110,138,142,179,184,188]. Nanocarriers functionalized with active targeting ligands—such as aptamers, folate, transferrin, and prostate-specific membrane antigen—have demonstrated increased cellular internalization and induced apoptosis in tumor cells at significantly lower drug doses compared to their non-targeted counterparts [189]. For example, apoferritin nanocages achieved IC50 values ranging from 14 to 225 nM, compared to 14 to 68 nM for the free drug, suggesting a concentration-dependent but improved delivery efficiency [190]. Carbon nitride dot-based platforms induced apoptosis at 50 nM, markedly outperforming free doxorubicin, which required concentrations up to 1000 nM [190]. Liposomal formulations and polymer-drug conjugates demonstrated IC50 values between 20 and 2000 nM, often exhibiting reversible or attenuated cytotoxicity, improving safety margins [191]. Squalenoylated lipid nanoparticles allowed for a fivefold increase in the maximum tolerated dose, significantly reducing cardiotoxicity commonly associated with anthracycline-based therapies [192].
The mechanistic basis for these enhanced outcomes centers on specific molecular interactions between nanoparticle surfaces and tumor cell receptors [6,10,35,110,138,142,179,184,188]. Key receptors implicated in targeted delivery include heat shock protein 70 (HSP70), transferrin receptor 1 (TfR1), folate receptors, and integrins [6,10,35,110,138,142,179,184,188]. Targeting these molecules facilitates preferential tumor accumulation, minimizes systemic exposure, and reduces off-target toxicity [6,10,35,110,138,142,179,184,188]. In contrast, passively targeted nanodrugs rely on the enhanced permeability and retention (EPR) effect to accumulate in tumor tissue [193,194,195]. While this mechanism can be effective, it lacks the precision of receptor-mediated targeting and often results in broader biodistribution and higher systemic toxicity [193,194,195]. It is worth noting here that studies challenged the efficacy of EPR effect, identifying a high degree of heterogeneity and limited experimental data from patients on the effectiveness of this mechanism [193,194,195]. In fact, tumor microenvironment, e.g., vascular density, extracellular matrix composition, and interstitial pressure, are main factors in determining the extent of nanoparticle extravasation [193,194,195,196,197]. Imaging assessments and tailored nanocarrier design emerge as critical considerations to address the inherent heterogeneity of the EPR effect as reported in both preclinical and clinical settings [193,194,195]. Finally, therapeutically optimal cytotoxicity relies to a great extent on the targeting strategy and is largely characterized by the mechanism of action of the API. Typically, the data indicate that nanodrug preparations featuring well-established active targeting strategies are generally more superior regarding their selectivity toward tumors, cytotoxicity profiles, and therapeutic indexes than those relying entirely on passive targeting mechanisms.
While nanodrugs offer enhanced efficacy and targeted delivery, their cytotoxic potential can also contribute to adverse drug reactions (ADRs). Assessing and understanding nanodrug-induced toxicity is therefore essential for ensuring patient safety and guiding the development of safer nanopharmaceuticals. Studying cytotoxicity at the cellular and molecular levels enables researchers to pinpoint the specific pathways and mechanisms by which nanodrugs exert toxic effects. Such mechanistic insights are critical for predicting, mitigating, and ultimately preventing ADRs during both preclinical development and clinical application. To address this, we conducted a systematic analysis of current literature, using defined criteria for nanodrugs (as outlined in Section 3.1), with the aim of identifying shared molecular features and common signaling pathways linked to ADR-related cytotoxicity. This effort focuses on elucidating the cellular mechanisms most frequently implicated in the toxic responses to nanodrug formulations.
Our findings, summarized in Table 2, reveal that three major molecular mechanisms consistently underlie nanoparticle-induced cytotoxicity: oxidative stress pathways (characterized by excessive generation of ROS, leading to mitochondrial dysfunction, lipid peroxidation, and DNA damage), inflammatory signaling cascades (involving activation of pro-inflammatory cytokines and key pathways such as NF-κB and mitogen-activated protein kinases-MAPK, which contribute to chronic inflammation and tissue damage) and lysosomal dysfunction (resulting from nanoparticle accumulation or degradation within lysosomes, causing membrane destabilization, leakage of enzymes, and disruption of cellular homeostasis) (see details in Table 2).
Oxidative stress consistently emerges as a predominant mechanism of nanotoxicity across numerous studies (Figure 3) [6,10,35,120,122,136,138,176,179,184]. Alongside inflammation and DNA damage, oxidative stress has been identified as a key contributor to the cytotoxic effects of nanodrugs (Figure 3) [6,10,35,120,122,136,138,176,179,184]. A central driver of oxidative stress is the excessive generation and/or accumulation of ROS (highly reactive molecules that can damage essential cellular components) [6,10,35,120,122,136,138,176,179,184]. The specific mechanisms by which nanoparticles induce ROS production vary depending on their physicochemical properties, including size, shape, surface charge, and material composition [6,10,35,120,122,136,138,176,179,184]. Some nanoparticles directly produce ROS through surface redox reactions or photoactivation, while others indirectly increase ROS levels by disrupting cellular antioxidant defenses (Figure 3) [179,198].
For example, platinum NPs have been shown to mimic the activity of NADPH oxidase, an enzyme complex involved in ROS generation, potentially amplifying oxidative stress in exposed cells [198]. However, multiple studies coherently report that rising levels of ROS compromise redox balance [6,10,35,120,122,136,138,176,179,184]. Elevated ROS levels coincide with glutathione depletion, loss of mitochondrial membrane potential, lipid peroxidation, and damage to DNA and proteins (Figure 3) [6,10,35,120,122,136,138,176,179,184]. Thus, current evidence indicates that nanodrug-induced oxidative stress mediates cytotoxicity primarily via ROS generation and impaired antioxidant defenses (Figure 3) [6,10,35,120,122,136,138,176,179,184].
Inflammation represents another major mechanism of nanoparticle-induced toxicity and is frequently observed alongside oxidative stress and DNA damage (Figure 4) [6,10,35,120,122,136,138,176,179,184]. Numerous studies have demonstrated that NPs can activate a wide array of inflammatory signaling pathways, most notably NF-κB, MAPK, and phosphatidylinositol 3-kinase/Akt (PI3K/Akt) pathways [179,199]. These activations result in the upregulation and release of pro-inflammatory cytokines such as TNF-α, IL-6, and IL-8, contributing to both local and systemic inflammatory responses (Figure 4) [179]. Exposure to nanoparticles has been shown to modulate gene and protein expression in inflammatory cascades, including cyclooxygenase-2 (COX-2) and downstream MAPK/PI3K/Akt pathways [199]. This dysregulation contributes to an environment of chronic inflammation, which can drive further cytotoxicity and impair cellular homeostasis [6,10,35,120,122,136,138,176,179,184]. A particularly well-characterized mechanism involves the activation of the NLR family pyrin domain containing 3 (NLRP3) inflammasome, especially in immune and epithelial cells [200,201,202,203,204]. For instance, amino-modified polystyrene NPs have been shown to destabilize lysosomes and induce ROS production, both of which contribute to NLRP3 inflammasome activation [200,201,202,203,204]. This leads to the release of interleukin-1β (IL-1β) and initiates pyroptotic or necrotic cell death pathways (Figure 4) [200,201,202,203,204]. Similarly, zinc oxide NPs have been reported to stimulate NF-κB and MAPK signaling in monocytes, leading to the production of TNF-α and IL-1β, enhanced oxidative stress, and DNA damage [205]. Additional studies highlight shifts in cytokine profiles—including increased levels of nitric oxide (NO), IL-6, and inducible nitric oxide synthase (iNOS)—and changes in macrophage polarization, which further link inflammation to nanoparticle-mediated cytotoxicity [206].
Importantly, a growing body of research suggests that these inflammatory effects can be modulated by therapeutic interventions [200,201,202,203,204]. The use of ROS scavengers, nonsteroidal anti-inflammatory drugs (NSAIDs), small-molecule pathway inhibitors, and optimized nanoparticle formulations has shown promise in attenuating NP-induced inflammation and mitigating associated cytotoxicity [200,201,202,203,204,207]. Therefore, the relationship between oxidative stress and inflammation is particularly noteworthy, as ROS generation frequently acts as a trigger for inflammatory cascades. This creates a positive feedback loop in which inflammation enhances ROS production, and vice versa, amplifying cellular stress, injury, and death [200,201,202,203,204].
Next, lysosomal dysfunction has emerged as a critical intracellular mechanism underlying nanodrug-induced cytotoxicity (Figure 5). Numerous studies have identified lysosomal damage, membrane permeabilization, and impaired enzymatic activity as common cellular responses to nanoparticle exposure (Figure 5) [6,10,35,120,122,136,138,176,179,184]. Lysosomes, essential for cellular homeostasis, act as key sites for the degradation and recycling of biomolecules [208]. However, due to their acidic environment and membrane-bound architecture, they are also particularly vulnerable to disruption by internalized nanomaterials (Figure 5) [6,10,35,120,122,136,138,176,179,184]. The mechanisms by which nanoparticles induce lysosomal dysfunction are diverse and highly dependent on particle composition, surface charge, and degradation behavior [6,10,35,120,122,136,138,176,179,184]. Certain nanoparticles—particularly those with cationic surface charges or acid-sensitive moieties—tend to accumulate within lysosomes [10,180]. This accumulation can lead to lysosomal membrane permeabilization (LMP), leakage of hydrolytic enzymes such as cathepsins into the cytoplasm, and subsequent activation of cell death pathways, including apoptosis and necrosis (Figure 5) [10,180,209]. Other nanoparticles may disrupt lysosomal pH regulation, impair autophagic flux, or interfere with exocytosis, thereby disturbing the cell’s waste management and stress response systems (Figure 5) [181].
Studies have shown that cationic liposomes and pH-responsive nanogels can induce LMP, triggering cathepsin release, mitochondrial membrane depolarization, and excessive ROS generation [6,10,35,120,122,136,138,176,179,184,210]. For example, zinc oxide NPs have been reported to impair autophagy and reduce mitochondrial membrane potential—a process mitigated by antioxidant treatment such as N-acetylcysteine, implicating oxidative stress as a downstream consequence of lysosomal damage [211]. Similarly, iron oxide nanoparticles have been shown to promote ROS formation via Fenton-type reactions, leading to mitochondrial dysfunction, DNA damage, and enhanced cytotoxic or anticancer effects [26,212]. Notably, these events often follow or coincide with lysosomal disruption, reinforcing the idea that lysosomal dysfunction serves as both a trigger and amplifier of oxidative stress [10,26]. Lysosomal impairment triggered by NPs is typically characterized by membrane destabilization or rupture, inhibition of autophagic flux, disruption of lysosomal enzyme activity, altered exocytosis or intracellular trafficking (Figure 5) [6,10,35,120,122,136,138,176,179,184,210]. These events initiate a cascade of intracellular disturbances that ultimately result in cell death, as confirmed by markers of apoptosis, necrosis, and inflammatory responses [6,10,35,120,122,136,138,176,179,184,210]. Quantitative analyses, including IC50 values and biomarker profiling, further support the cytotoxic impact of lysosomal damage in various cell types [6,10,35,120,122,136,138,176,179,184,210].
Collectively, the evidence suggests that lysosomal dysfunction is a pivotal contributor to nanodrug cytotoxicity, closely interconnected with oxidative stress and mitochondrial impairment. This crosstalk between organelle systems exacerbates cellular damage and underscores the importance of lysosomal stability in the safe design of nanodrug platforms.
It is important to stress the need for summarizing representative research on cytotoxicity with close attention to material-specific trends, NP types, and their correlations with cytotoxic effects. While Table 2 outlines major molecular pathways in nanoparticle-induced cytotoxicity, it is a generalized picture lacking stratification by NP type. For this reason, we present Table 3 and Table 4, which summarizes representative research with particular attention to stratification by nanoparticle type.

5.2. Predictive Frameworks Determining Cytotoxicity of Nanodrugs

While oxidative stress, inflammation, and lysosomal destabilization are distinct molecular signatures of nanoparticle toxicity, previous reviews have primarily approached these mechanisms in general contexts or relative to preclinical material rather than clinically relevant nanodrug formulations [10,35,116,120,136,145,176,179,184]. The aim of this work is to move beyond repetition through the identification of formulation-dependent molecular mechanisms and uncovering under-valued mechanistic properties related to active versus passive targeting, carrier type, and intracellular trafficking behavior. In order to achieve this, we conducted a systematic re-review of the literature that solely focused on nanodrugs (defined as in Section 3.1), supplemented by Elicit platform-facilitated pathway extraction with AI support. This allowed us to compare which different nanodrug classes selectively modulate cytotoxic pathways, quantify biomarkers, and evaluate underexplored mechanisms like autophagy inhibition and inflammasome activation. Our aim was to develop an even finer-grained, comparative model of predicting and averting ADRs at the molecular level.
Having identified the key molecular drivers of cytotoxicity induced by nanodrugs (e.g., oxidative stress, damage and barrier dysfunction of the membrane, lysosomal impairment, activation of pro-inflammatory cascades, and mitochondrial injury) we applied the Elicit platform for data extraction from foundational literature (see Table S2) and based on summarized data generated a qualitative comparative matrix. We constructed a qualitative comparative matrix (Table 4) that associates specific nanodrug classes (e.g., liposomes, polymer-drug conjugates, inorganic nanoparticles) with their most prevalent cytotoxic mechanisms (e.g., ROS generation, lysosomal membrane disruption, cytokine release). This enables comparison across formulation-type toxicological profiles and uncovers the relative importance of each molecular stimulus toward total cytotoxicity. We grouped each cytotoxic mechanism into three categories based on its reported association strength: strong (consistently reported or well-known as a first-order mechanism), moderate (consistently reported), and weak (occasionally reported).
This was conducted according to how frequently and prominently each mechanism appeared in the literature. The association strength was qualitatively assessed in the studies referenced. As many publications did not report specific IC50 values or quantitative ROS measurements, the resulting matrix is presented in a qualitative format.
It needs to be noted that due to the structural and compositional diversity of nanomaterials (i.e., metal, polymer, and lipid-based platforms) their interactions with biological systems, and the resulting toxic effects, are highly context-dependent [6,10,35,110,138,142,179,184,188]. They may vary with the route of administration, biological environment, and nanomaterial architecture [6,10,35,110,138,142,179,184,188,236]. As current evidence indicates, there could be no universally applicable nanodrug platform, given that cytotoxic mechanisms are largely determined by the kind of nanomaterial being used [10,21]. However, it is still possible to generalize existing cytotoxicity data by stratifying it into classes of nanomaterials, whereby common mechanistic trends are established for comparable platforms and even finding cross-platforms similarities.
Summarizing evidence in Section 4 and Section 5, it is obvious that cytotoxicity of nanomaterials is strongly influenced by numerous physicochemical parameters like particle size, charge on the surface, composition of the material, coating on the surface, and shape [6,10,35,110,138,142,179,184,188]. Below, we briefly outline each of these parameters’ contribution. There is a rapidly emerging consensus in the literature that the smaller NPs will be more cytotoxic compared to larger NPs [26,132,133,134]. For example, 1.4 nm gold NPs have been found to be toxic, while nanoparticles with diameters of 15 nm or larger are typically found to have negligible or no toxicity [237,238,239]. Such a size-dependent effect is largely due to increased surface area, cellular uptake, and increased interactions with intracellular components [6,10,35,110,138,142,179,184,188]. Positively charged NPs are found to show increased cytotoxicity due to increased electrostatic interactions with the negatively charged cell membranes [182]. These interactions can cause enhanced membrane disruption and facilitate cellular uptake, with the potential to produce down-stream toxicities [182]. Surface modifications such as PEGylation can prevent nonspecific interactions and cytotoxicity [240]. However, they also have the potential to introduce new problematics such as immunogenic reactions or alterations in biodistribution [240]. The presence and integrity of a protein corona, a biomolecular layer that develops on the NP surface when it comes into contact with biological fluids, also make significant impacts on cellular interactions, recognition, and immunity by the immune system [240]. Toxicity can be material-specific. For instance, cadmium-based quantum dots (e.g., CdTe) are more toxic than quantum dots that are protected by multiple layers (e.g., CdSe/ZnS) [187]. Metal oxide nanoparticles have been shown to produce ROS, which play a role in oxidative stress and cellular damage [241,242]. Carbon nanomaterials, such as carbon nanotubes, particularly needle-like structures, have been said to exhibit unusual and in most instances increased toxicity due to their shape and stiffness [110,116,185]. Hydrophobic surfaces also contribute to cell membrane disruption and hence become more cytotoxic [182]. The disruption may enable unregulated entry of nanoparticles into cells and organelles [182]. Additionally, size plays a crucial role in immune recognition and cellular uptake [6,10,35,110,138,142,179,184,188]. Furthermore, spherical NPs are more readily internalized by macrophages [186]. High-aspect-ratio needle-like carbon nanotubes, however, have been associated with toxicity, which could be due to frustrated phagocytosis and persistent cellular stress [243,244].
Based on the reviewed literature and our systematic analysis, we have summarized the key cytotoxicity mechanisms associated with different classes of nanodrugs (Figure 6). Each class of nanomaterial has characteristic biological interactions and cellular effects. Metal NPs are primarily recognized as potent generators of ROS, inducing oxidative stress, lipid peroxidation, DNA damage, and leading to apoptosis or necrosis (Figure 6). Quantum dots, by release of the heavy metal core components, show high mitochondrial toxicity by disrupting membrane potential, inhibiting ATP synthesis, and initiating intrinsic apoptotic pathways (Figure 6). Carbon nanotubes and lipid NPs will most likely cause robust inflammatory reactions, partly due to their geometry, surface characteristics, and immune recognition, often in combination with cytokine release and macrophage activation. Polymeric NPs tend to accumulate in lysosomes and cause lysosomal destabilization, which may lead to leakage of the enzyme into the cytoplasm and initiation of cell death pathways (Figure 6).
Based on our systematic analysis (supported by the data presented in Table 4 and the cytotoxicity mechanisms illustrated in Figure 6) we propose a semi-quantitative model for evaluating the cytotoxic risk of nanomaterials (Figure 7). This model, titled the Toxicity Index Scoring System, integrates representative physicochemical properties, biological response indicators, and surface modification parameters to give a composite risk score of 0 to 21 points. Depending on the overall score, nanomaterials receive low, moderate, or high cytotoxicity risk categories.
The compositional and structural characteristics of nanoparticles (particularly particle size, shape, surface charge, and material composition) are important determinants of their biological behavior. These parameters dictate the interactions of nanoparticles with cells, tissues, and organelles, and thus establish their toxicological profiles. In addition to these intrinsic properties, we incorporate biomarker-based parameters reflecting cellular responses commonly encountered in both in vitro and in vivo toxicological studies. These include the levels of oxidative stress, inflammatory cytokine release, and damage to organelles or membranes—each a functional indicator of nanoparticle-induced cytotoxicity. Finally, the modifying factors such as surface PEGylation, biodegradable coatings, or protein corona formation are also considered, as these may decrease or increase cytotoxic potential. Taken together, this structured Toxicity Index Scoring System constitutes a semi-quantitative, evidence-based framework for the direct comparison of nanoparticle cytotoxicity across different material types. It is designed to support preclinical safety screening, safer-by-design nanomaterials, and inform risk assessment strategies in the fields of nanomedicine, toxicology, and environmental health sciences.

6. Outlook and Perspectives

Our analysis reveals that several classes of nanodrugs—including liposomes, albumin-based nanospheres, and iron oxide nanoparticles—have already received FDA approval and are in clinical use, while others are progressing through various stages of clinical translation [4,5,6,9,10,26,28,45,46,47]. The choice of nanodrug platform and targeting strategy should be carefully tailored to tumor type, receptor expression patterns, and the broader clinical context to optimize therapeutic efficacy and safety [4,5,6,9,10,26,28,45,46,47].
We observed notable differences among nanodrugs in terms of cytotoxicity and molecular interactions. These variations arise from the complex interplay of multiple factors, including nanoparticle size, shape, surface charge, functionalization, and composition [6,10,26,116,134,172,173,174]. For instance, cadmium-containing quantum dots consistently demonstrate higher toxicity compared to other nanomaterials [187]. Additionally, many studies report a dose-dependent increase in toxicity, with higher concentrations of nanoparticles inducing more severe biological effects [199]. Despite the diversity of nanomaterials and their applications, our analysis identified three predominant molecular mechanisms that underlie nanoparticle-induced cytotoxicity: oxidative stress pathways, inflammatory signaling cascades, lysosomal dysfunction. These shared mechanisms are summarized in Table 2. Understanding these pathways provides a foundation for the rational design of risk mitigation strategies. We outlined tentatively proposed risk mitigation strategies in Table 5 that are tailored to nanoparticle type and cytotoxic mechanisms.
A recurring concern across different nanodrug categories is the generation of cytotoxic ROS, which is closely linked to oxidative stress and often acts as a central mediator of cytotoxicity [6,10,35,120,122,136,138,176,179,184]. ROS generation is frequently observed across a wide range of nanomaterials and often intersects with inflammatory and lysosomal pathways, forming feedback loops that amplify cellular damage [6,10,35,120,122,136,138,176,179,184]. This interplay is visualized in Figure 3, Figure 4, Figure 5 and Figure 6 and underscores oxidative stress as a core contributor to nanotoxicity.
While some safety concerns—such as ROS generation—are common across many nanodrug types, others are material-specific [6,10,35,120,122,136,138,176,179,184]. For example, carbon nanotubes have been associated with asbestos-like effects, while zinc oxide nanoparticles present toxicity related to their dissolution and ion release [246]. Among the most commonly reported strategies to mitigate these risks are surface modifications, which improve biocompatibility and reduce immune recognition (Table 5). Controlling physical parameters such as size and shape also emerged as critical to minimizing toxicity. These insights highlight the complexity of nanodrug safety and emphasize the need for comprehensive, material-specific approaches to toxicological assessment and risk management in nanomedicine development.
Overall, based on the structured assessment presented in this study, we have built a preliminary, concise, and structured checklist that aims to decrease nanodrug-induced toxicity (see Supplementary Materials: Cytotoxicity Checklist). Our approach was inspired by the MIRIBEL guidelines (Minimum Information Reporting in Bio-Nano Experimental Literature), on which we propose laying the foundations to report experimental studies on bio–nano interactions [249]. Building on these thoroughly established guidelines, we provisionally propose a supplementary checklist aimed at reducing cytotoxicity specifically (see Supplementary Materials: Cytotoxicity Checklist). Even though we are aware of the shortcomings of our checklist—most prominently its insufficiency of specificity regarding the exact physicochemical characteristics of NPs—we believe that even a preliminary structure of this kind constitutes an innovative input, especially given that, as far as we can tell, such a checklist has not yet been offered in the literature.
This checklist serves as a systematic methodology for minimizing the toxicity of the nanodrug without compromising therapeutic efficacy, thus allowing the construction of more secure nanopharmaceuticals for clinical use. The underlying principles of the checklist are: prioritizing active targeting over passive targeting to optimize selectivity; physicochemical optimization (e.g., particle size > 15 nm, neutral or negative charge, spherical shape); application of surface modification technologies such as PEGylation or biodegradable coatings; avoidance of key mechanisms of toxicity such as oxidative stress, inflammation, and lysosomal dysfunction; application of evidence-based risk assessment such as a Toxicity Index Scoring System (Figure 7); safety validation through rigorous mechanistic studies; incorporation of material-specific risks into the design phase. We hope this proposed checklist serves as a starting point for further refinement and validation within the scientific community.
Looking ahead, novel nanomaterials are being developed to enhance therapeutic performance while reducing adverse effects. Notably, advances in molecular self-assembly and nucleic acid nanotechnology have introduced new possibilities for precision nanomedicine [250,251,252,253,254,255,256]. Programmable biopolymeric materials (such as DNA nanocarriers) are particularly promising due to their inherent biocompatibility, structural precision, and molecular programmability [250,251,252,253,254,255,257]. DNA nanostructures (DNs) provide a versatile platform for nanoscale drug delivery, enabling controlled cargo release, improved targeting, and reduced systemic toxicity [36,254,258,259]. Although still in early stages of clinical development, DNA nanocarriers have demonstrated remarkable potential in enhancing drug delivery mechanisms and interacting with biological systems [36,254,258]. For example, functionalized DNA origami have been instrumental in dissecting nanoscale processes such as immune receptor activation [260,261,262,263], enzymatic activity [264,265], and spatial tolerances in antibody–antigen interactions [266]. Furthermore, DNA nanotechnology offers synthesis of highly versatile platforms for the precise modulation of cellular functions by targeting and regulating specific organelles through rational design [259,267,268,269,270]. This innovative approach represents a paradigm shift in nanomedicine, introducing a new class of nanocarriers with the potential to transform therapeutic strategies and significantly broaden the scope of biomedical interventions [271].
Overall, our analysis emphasizes that nanodrug-induced cytotoxicity results from a multifaceted interaction of cellular pathways, particularly those involving mitochondrial function, lysosomal integrity, ROS production, inflammation and membrane dynamics. By identifying common molecular mechanisms and emerging mitigation strategies, this work lays the groundwork for designing safer, more effective nanomedicines tailored to both clinical needs and biomedical applications.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/ijms26146687/s1.

Author Contributions

Conceptualization, O.L.; methodology, O.L. and M.L.; software, A.C., P.E. and O.L.; validation, H.A., M.L. and O.L.; formal analysis, A.C., P.E. and H.A.; investigation, A.C., P.E. and H.A.; resources, O.L.; data curation, O.L.; writing—original draft preparation, O.L.; writing—review and editing, A.C., P.E., H.A. and M.L.; visualization, A.C., P.E., H.A. and O.L.; supervision, O.L.; project administration, O.L.; funding acquisition, O.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Operational Program Johannes Amos Comenius financed by European Structural and Investment Funds and the Czech Ministry of Education, Youth and Sports (Project no. SENDISO-CZ.02.01.01/00/22_008/0004596).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study.

Acknowledgments

The authors acknowledge the MH CZ—DRO Institute for Clinical and Experimental Medicine—IKEM, IN 00023001. Figure 1, Figure 2, Figure 3, Figure 4, Figure 5, Figure 6 and Figure 7 and the graphical abstract were created using BioRender.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
APIsactive pharmaceutical ingredients
FDAU.S. Food and Drug Administration
NPsNanoparticles
EMAEuropean Medicines Agency
VZVVaricella-Zoster Virus
SARS-CoV-2Severe acute respiratory syndrome coronavirus 2
AML-MRCAcute myeloid leukemia with myelodysplasia-related changes
t-AMLTherapy-related acute myeloid leukemia
MACMycobacterium avium complex
NTMNontuberculous mycobacteria
CKDChronic kidney disease
PEGPolyethylene glycol
PLGAPoly(lactic-co-glycolic) acid
CNTsCarbon nanotubes
CTABCetyltrimethylammonium bromide
LPSLipopolysaccharides
TLR4Toll-like receptor 4
CMEClathrin-mediated endocytosis
FEMEFast endophilin-mediated endocytosis
CLIC/GEECClathrin-independent carrier/glycosylphosphatidylinositol-anchored protein-enriched early endocytic compartment endocytosis
PEIPolyethyleneimine
MPSMononuclear phagocyte system
BSABovine serum albumin
ROSReactive Oxygen Species
EREndoplasmic Reticulum
CDKsCyclin-dependent kinases
NF-κBNuclear factor kappa-light-chain-enhancer of activated B cells
IL-6Interleukin 6
IL-8Interleukin 8
IL-1βInterleukin-1β
TNF-αTumor necrosis factor α.
HSP70Heat shock protein 70
TfR1Transferrin receptor 1
EPREnhanced permeability and retention
MAPKMitogen-activated protein kinases
LMPLysosomal membrane permeabilization
UPRUnfolded protein response
COX-2Cyclooxygenase-2
NLRP3NLR family pyrin domain containing 3
NSAIDsNonsteroidal anti-inflammatory drugs
DNsDNA nanostructures

References

  1. Bhatia, S.N.; Chen, X.; Dobrovolskaia, M.A.; Lammers, T. Cancer nanomedicine. Nat. Rev. Cancer 2022, 22, 550–556. [Google Scholar] [CrossRef] [PubMed]
  2. Younis, M.A.; Tawfeek, H.M.; Abdellatif, A.A.H.; Abdel-Aleem, J.A.; Harashima, H. Clinical translation of nanomedicines: Challenges, opportunities, and keys. Adv. Drug Deliv. Rev. 2022, 181, 114083. [Google Scholar] [CrossRef]
  3. Available online: https://commonfund.nih.gov/nanomedicine (accessed on 27 January 2025).
  4. 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] [PubMed]
  5. Fan, D.; Cao, Y.; Cao, M.; Wang, Y.; Cao, Y.; Gong, T. Nanomedicine in cancer therapy. Signal Transduct. Target. Ther. 2023, 8, 293. [Google Scholar] [CrossRef]
  6. Havelikar, U.; Ghorpade, K.B.; Kumar, A.; Patel, A.; Singh, M.; Banjare, N.; Gupta, P.N. Comprehensive insights into mechanism of nanotoxicity, assessment methods and regulatory challenges of nanomedicines. Discov. Nano 2024, 19, 165. [Google Scholar] [CrossRef]
  7. Wang, J.; Li, Y.Y.; Nie, G.J. Multifunctional biomolecule nanostructures for cancer therapy. Nat. Rev. Mater. 2021, 6, 766–783. [Google Scholar] [CrossRef] [PubMed]
  8. Szebeni, J.; Storm, G.; Ljubimova, J.Y.; Castells, M.; Phillips, E.J.; Turjeman, K.; Barenholz, Y.; Crommelin, D.J.A.; Dobrovolskaia, M.A. Applying lessons learned from nanomedicines to understand rare hypersensitivity reactions to mRNA-based SARS-CoV-2 vaccines. Nat. Nanotechnol. 2022, 17, 337–346. [Google Scholar] [CrossRef]
  9. Anselmo, A.C.; Mitragotri, S. Nanoparticles in the clinic: An update post COVID-19 vaccines. Bioeng. Transl. Med. 2021, 6, e10246. [Google Scholar] [CrossRef]
  10. Uzhytchak, M.; Smolková, B.; Lunova, M.; Frtús, A.; Jirsa, M.; Dejneka, A.; Lunov, O. Lysosomal nanotoxicity: Impact of nanomedicines on lysosomal function. Adv. Drug Deliv. Rev. 2023, 197, 114828. [Google Scholar] [CrossRef]
  11. Herrmann, I.; Li, Z.A.; Bahal, R.; Conde, J. Translating nanomedicines from the lab to the clinic. Cell Rep. Phys. Sci. 2025, 6, 102357. [Google Scholar] [CrossRef]
  12. Dordevic, 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] [PubMed]
  13. Mitragotri, S.; Lammers, T.; Bae, Y.H.; Schwendeman, S.; De Smedt, S.; Leroux, J.C.; Peer, D.; Kwon, I.C.; Harashima, H.; Kikuchi, A.; et al. Drug delivery research for the future: Expanding the nano horizons and beyond. J. Control. Release 2017, 246, 183–184. [Google Scholar] [CrossRef]
  14. Hua, S.; de Matos, M.B.C.; Metselaar, J.M.; Storm, G. Current trends and challenges in the clinical translation of nanoparticulate nanomedicines: Pathways for translational development and commercialization. Front. Pharmacol. 2018, 9, 790. [Google Scholar] [CrossRef] [PubMed]
  15. Skrypnyk, N.I.; Siskind, L.J.; Faubel, S.; de Caestecker, M.P. Bridging translation for acute kidney injury with better preclinical modeling of human disease. Am. J. Physiol.-Ren. Physiol. 2016, 310, F972–F984. [Google Scholar] [CrossRef]
  16. Joudeh, N.; Linke, D. Nanoparticle classification, physicochemical properties, characterization, and applications: A comprehensive review for biologists. J. Nanobiotechnol. 2022, 20, 262. [Google Scholar] [CrossRef] [PubMed]
  17. Wu, L.B.; Zhang, J.; Watanabe, W. Physical and chemical stability of drug nanoparticles. Adv. Drug Deliv. Rev. 2011, 63, 456–469. [Google Scholar] [CrossRef]
  18. 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]
  19. Wilhelm, S.; Tavares, A.J.; Dai, Q.; Ohta, S.; Audet, J.; Dvorak, H.F.; Chan, W.C.W. Analysis of nanoparticle delivery to tumours. Nat. Rev. Mater. 2016, 1, 16014. [Google Scholar] [CrossRef]
  20. Rosenblum, D.; Joshi, N.; Tao, W.; Karp, J.M.; Peer, D. Progress and challenges towards targeted delivery of cancer therapeutics. Nat. Commun. 2018, 9, 1410. [Google Scholar] [CrossRef]
  21. Sun, D.X.; Zhou, S.; Gao, W. What went wrong with anticancer nanomedicine design and how to make it right. ACS Nano 2020, 14, 12281–12290. [Google Scholar] [CrossRef]
  22. Wáng, Y.X.J.; Idée, J.M. A comprehensive literatures update of clinical researches of superparamagnetic resonance iron oxide nanoparticles for magnetic resonance imaging. Quant. Imaging Med. Surg. 2017, 7, 88–122. [Google Scholar] [CrossRef]
  23. Kendall, M.; Lynch, I. Long-term monitoring for nanomedicine implants and drugs. Nat. Nanotechnol. 2016, 11, 206–210. [Google Scholar] [CrossRef] [PubMed]
  24. Rubin, R. Black box warning for anemia drug. JAMA 2015, 313, 1704. [Google Scholar] [CrossRef]
  25. Sun, T.; Kang, Y.Y.; Liu, J.; Zhang, Y.L.; Ou, L.L.; Liu, X.N.; Lai, R.F.; Shao, L.Q. Nanomaterials and hepatic disease: Toxicokinetics, disease types, intrinsic mechanisms, liver susceptibility, and influencing factors. J. Nanobiotechnol. 2021, 19, 108. [Google Scholar] [CrossRef]
  26. Frtús, A.; Smolková, B.; Uzhytchak, M.; Lunova, M.; Jirsa, M.; Kubinová, S.; Dejneka, A.; Lunov, O. Analyzing the mechanisms of iron oxide nanoparticles interactions with cells: A road from failure to success in clinical applications. J. Control. Release 2020, 328, 59–77. [Google Scholar] [CrossRef] [PubMed]
  27. Bobo, D.; Robinson, K.J.; Islam, J.; Thurecht, K.J.; Corrie, S.R. Nanoparticle-based medicines: A review of FDA-approved materials and clinical trials to date. Pharm. Res. 2016, 33, 2373–2387. [Google Scholar] [CrossRef] [PubMed]
  28. Anselmo, A.C.; Mitragotri, S. Nanoparticles in the clinic: An update. Bioeng. Transl. Med. 2019, 4, e10143. [Google Scholar] [CrossRef]
  29. Aronson, J.K.; Ferner, R.E. Joining the DoTS: New approach to classifying adverse drug reactions. BMJ 2003, 327, 1222–1225. [Google Scholar] [CrossRef]
  30. Coleman, J.J.; Pontefract, S.K. Adverse drug reactions. Clin. Med. 2016, 16, 481–485. [Google Scholar] [CrossRef]
  31. Liebler, D.C.; Guengerich, F.P. Elucidating mechanisms of drug-induced toxicity. Nat. Rev. Drug Discov. 2005, 4, 410–420. [Google Scholar] [CrossRef]
  32. Pognan, F.; Beilmann, M.; Boonen, H.C.M.; Czich, A.; Dear, G.; Hewitt, P.; Mow, T.; Oinonen, T.; Roth, A.; Steger-Hartmann, T.; et al. The evolving role of investigative toxicology in the pharmaceutical industry. Nat. Rev. Drug Discov. 2023, 22, 317–335. [Google Scholar] [CrossRef] [PubMed]
  33. Helal-Neto, E.; de Barros, A.O.D.; Saldanha-Gama, R.; Brandao-Costa, R.; Alencar, L.M.R.; dos Santos, C.C.; Martínez-Máñez, R.; Ricci-Junior, E.; Alexis, F.; Morandi, V.; et al. Molecular and cellular risk assessment of healthy human cells and cancer human cells exposed to nanoparticles. Int. J. Mol. Sci. 2020, 21, 230. [Google Scholar] [CrossRef]
  34. Laux, P.; Tentschert, J.; Riebeling, C.; Braeuning, A.; Creutzenberg, O.; Epp, A.; Fessard, V.; Haas, K.H.; Haase, A.; Hund-Rinke, K.; et al. Nanomaterials: Certain aspects of application, risk assessment and risk communication. Arch. Toxicol. 2018, 92, 121–141. [Google Scholar] [CrossRef]
  35. Thu, H.E.; Haider, M.; Khan, S.; Sohail, M.; Hussain, Z. Nanotoxicity induced by nanomaterials: A review of factors affecting nanotoxicity and possible adaptations. OpenNano 2023, 14, 100190. [Google Scholar] [CrossRef]
  36. Jiang, W.; Wang, Y.; Wargo, J.A.; Lang, F.F.; Kim, B.Y.S. Considerations for designing preclinical cancer immune nanomedicine studies. Nat. Nanotechnol. 2021, 16, 6–15. [Google Scholar] [CrossRef] [PubMed]
  37. Singh, A.V.; Laux, P.; Luch, A.; Sudrik, C.; Wiehr, S.; Wild, A.-M.; Santomauro, G.; Bill, J.; Sitti, M. Review of emerging concepts in nanotoxicology: Opportunities and challenges for safer nanomaterial design. Toxicol. Mech. Methods 2019, 29, 378–387. [Google Scholar] [CrossRef]
  38. Owen, A.; Rannard, S.; Bawa, R.; Feng, S.-S. Interdisciplinary nanomedicine publications through interdisciplinary peer-review. J. Interdiscip. Nanomed. 2016, 1, 4–8. [Google Scholar] [CrossRef]
  39. Tinkle, S.; McNeil, S.E.; Mühlebach, S.; Bawa, R.; Borchard, G.; Barenholz, Y.; Tamarkin, L.; Desai, N. Nanomedicines: Addressing the scientific and regulatory gap. Ann. New York Acad. Sci. 2014, 1313, 35–56. [Google Scholar] [CrossRef]
  40. Available online: https://www.ema.europa.eu/en/news/european-medicines-agency-publishes-reflection-paper-general-issues-consideration-regarding-coated (accessed on 27 January 2025).
  41. Liz-Marzán, L.M.; Nel, A.E.; Brinker, C.J.; Chan, W.C.W.; Chen, C.Y.; Chen, X.D.; Ho, D.A.; Hu, T.; Kataoka, K.; Kotov, N.A.; et al. What do we mean when we say nanomedicine? ACS Nano 2022, 16, 13257–13259. [Google Scholar] [CrossRef]
  42. Mulvaney, P. Nanoscience vs nanotechnology-Defining the field. ACS Nano 2015, 9, 2215–2217. [Google Scholar] [CrossRef]
  43. Available online: https://www.fda.gov/regulatory-information/search-fda-guidance-documents/considering-whether-fda-regulated-product-involves-application-nanotechnology (accessed on 27 January 2025).
  44. Available online: https://www.cancer.gov/publications/dictionaries/cancer-terms/def/drug (accessed on 27 January 2025).
  45. Onoue, S.; Yamada, S.; Chan, H.K. Nanodrugs: Pharmacokinetics and safety. Int. J. Nanomed. 2014, 9, 1025–1037. [Google Scholar] [CrossRef] [PubMed]
  46. Wang, B.L.; Hu, S.Q.; Teng, Y.; Chen, J.L.; Wang, H.Y.; Xu, Y.Z.; Wang, K.Y.; Xu, J.G.; Cheng, Y.Z.; Gao, X. Current advance of nanotechnology in diagnosis and treatment for malignant tumors. Signal Transduct. Target. Ther. 2024, 9, 200. [Google Scholar] [CrossRef]
  47. Shan, X.T.; Gong, X.; Li, J.; Wen, J.Y.; Li, Y.P.; Zhang, Z.W. Current approaches of nanomedicines in the market and various stage of clinical translation. Acta Pharm. Sin. B 2022, 12, 3028–3048. [Google Scholar] [CrossRef] [PubMed]
  48. Khurana, N.; Sünner, T.; Hubbard, O.; Imburgia, C.; Stoddard, G.J.; Yellepeddi, V.; Ghandehari, H.; Watt, K.M. Micellar encapsulation of propofol reduces its adsorption on extracorporeal membrane oxygenator (ECMO) circuit. AAPS J. 2023, 25, 52. [Google Scholar] [CrossRef]
  49. Kumari, S.; Kumar, V.; Tiwari, R.K.; Ravidas, V.; Pandey, K.; Kumar, A. Amphotericin B: A drug of choice for Visceral Leishmaniasis. Acta Trop. 2022, 235, 106661. [Google Scholar] [CrossRef]
  50. Alberto, A.G.; Shira, G.-P.; Shadan, M.; Ninh, M.L.-B. Thirty years from FDA approval of pegylated liposomal doxorubicin (Doxil/Caelyx): An updated analysis and future perspective. BMJ Oncol. 2025, 4, e000573. [Google Scholar]
  51. Sirks, M.J.; Subhi, Y.; Rosenberg, N.; Hollak, C.E.M.; Boon, C.J.F.; Diederen, R.M.H.; Yzer, S.; Norel, J.; de Jong-Hesse, Y.; Schlingemann, R.O.; et al. Perspectives and update on the global shortage of verteporfin (visudyne®). Ophthalmol. Ther. 2024, 13, 1821–1831. [Google Scholar] [CrossRef]
  52. Strom, J.B.; Mulvagh, S.L.; Porter, T.R.; Wei, K.; Stout, J.L.; Main, M.L. Contemporary safety of ultrasound enhancing agents in a nationwide analysis. J. Am. Heart Assoc. Logo 2025, 14, e039480. [Google Scholar] [CrossRef]
  53. Singh, A.P.; Biswas, A.; Shukla, A.; Maiti, P. Targeted therapy in chronic diseases using nanomaterial-based drug delivery vehicles. Signal Transduct. Target. Ther. 2019, 4, 33. [Google Scholar] [CrossRef]
  54. Layon, S.A.; Williams, A.D.; Burns, H.R.; Parham, M.J.; Monson, L.A.; Mohammad, S.; Buchanan, E.P. Managing donor site pain after alveolar bone grafting: A comparative analysis of liposomal bupivacaine (exparel), bupivacaine-soaked gelfoam, and ON-Q ropivacaine. J. Craniofacial Surg. 2025, 36, 1291–1296. [Google Scholar] [CrossRef]
  55. Brown, M.B.; Blair, H.A. Liposomal irinotecan: A review as first-line therapy in metastatic pancreatic adenocarcinoma. Drugs 2025, 85, 255–262. [Google Scholar] [CrossRef] [PubMed]
  56. Liu, Z.C.; Fu, Z.; Liu, Y.H.; Jiao, Y. Biomaterial-based drug delivery: Evaluating the safety profiles of liposomal Vyxeos. Drug Deliv. 2025, 32, 2494781. [Google Scholar] [CrossRef]
  57. Curtis, J.R.; Conrad, D.M.; Krueger, W.S.; Gara, A.P.; Winthrop, K.L. Real-world data on the use of the Shingrix vaccine among patients with inflammatory arthritis and risk of cardiovascular events following herpes zoster. Arthritis Res. Ther. 2025, 27, 108. [Google Scholar] [CrossRef] [PubMed]
  58. Vaikkathillam, P.; Sajeevan, A.; Mohan, S.; Solomon, A.P.; Rajan, P.P.; Manjusree, S.; Kumar, P.; Thomas, S. Genomic analysis of colistin and carbapenem resistant Klebsiella pneumoniae GC29. Microb. Pathog. 2025, 199, 107220. [Google Scholar] [CrossRef] [PubMed]
  59. Hawley, J.J.; Allen, S.L.; Thompson, D.M.; Schwarz, A.J.; Tranquart, F.J.M. Commercially available ultrasound contrast agents: Factors contributing to favorable outcomes with ultrasound-mediated drug delivery and ultrasound localization microscopy imaging. Investig. Radiol. 2025. [Google Scholar] [CrossRef]
  60. Stukan, I.; Zuk, A.; Pukacka, K.; Mierzejewska, J.; Pawlowski, J.; Kowalski, B.; Dabkowska, M. Wolf in sheep’s clothing: Taming cancer’s resistance with human serum albumin? Int. J. Nanomed. 2025, 20, 3493–3525. [Google Scholar] [CrossRef]
  61. Paik, J.; Duggan, S.T.; Keam, S.J. Triamcinolone acetonide extended-release: A review in osteoarthritis pain of the knee. Drugs 2019, 79, 455–462. [Google Scholar] [CrossRef]
  62. Borges, G.S.M.; Lima, F.A.; Carneiro, G.; Goulart, G.A.C.; Ferreira, L.A.M. All-trans retinoic acid in anticancer therapy: How nanotechnology can enhance its efficacy and resolve its drawbacks. Expert Opin. Drug Deliv. 2021, 18, 1335–1354. [Google Scholar] [CrossRef]
  63. Macdougall, I.C.; Comin-Colet, J.; Breymann, C.; Spahn, D.R.; Koutroubakis, I.E. Iron sucrose: A wealth of experience in treating iron deficiency. Adv. Ther. 2020, 37, 1960–2002. [Google Scholar] [CrossRef]
  64. Germain, M.; Caputo, F.; Metcalfe, S.; Tosi, G.; Spring, K.; Åslund, A.K.O.; Pottier, A.; Schiffelers, R.; Ceccaldi, A.; Schmid, R. Delivering the power of nanomedicine to patients today. J. Control. Release 2020, 326, 164–171. [Google Scholar] [CrossRef]
  65. 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] [PubMed]
  66. Inglut, C.T.; Sorrin, A.J.; Kuruppu, T.; Vig, S.; Cicalo, J.; Ahmad, H.; Huang, H.C. Immunological and toxicological considerations for the design of liposomes. Nanomaterials 2020, 10, 190. [Google Scholar] [CrossRef] [PubMed]
  67. Janos, S.; Moghimi, S.M. Liposome triggering of innate immune responses: A perspective on benefits and adverse reactions. J. Liposome Res. 2009, 19, 85–90. [Google Scholar]
  68. Moraes-Lacerda, T.; de Jesus, M.B. Mechanisms of solid lipid nanoparticles-triggered signaling pathways in eukaryotic cells. Colloids Surf. B Biointerfaces 2022, 220, 112863. [Google Scholar] [CrossRef]
  69. Allen, T.M.; Cullis, P.R. Liposomal drug delivery systems: From concept to clinical applications. Adv. Drug Deliv. Rev. 2013, 65, 36–48. [Google Scholar] [CrossRef]
  70. Hua, S.; Wu, S.Y. The use of lipid-based nanocarriers for targeted pain therapies. Front. Pharmacol. 2013, 4, 143. [Google Scholar] [CrossRef] [PubMed]
  71. Monteiro, N.; Martins, A.; Reis, R.L.; Neves, N.M. Liposomes in tissue engineering and regenerative medicine. J. R. Soc. Interface 2014, 11, 20140459. [Google Scholar] [CrossRef]
  72. Shafiei, M.; Ansari, M.N.M.; Abd Razak, S.I.; Khan, M.U.A. A comprehensive review on the applications of exosomes and liposomes in regenerative medicine and tissue engineering. Polymers 2021, 13, 2529. [Google Scholar] [CrossRef]
  73. Gomes-da-Silva, L.C.; Fonseca, N.A.; Moura, V.; de Lima, M.C.P.; Simoes, S.; Moreira, J.N. Lipid-based nanoparticles for siRNA delivery in cancer therapy: Paradigms and challenges. Accounts Chem. Res. 2012, 45, 1163–1171. [Google Scholar] [CrossRef] [PubMed]
  74. Tenchov, R.; Bird, R.; Curtze, A.E.; Zhou, Q.Q. Lipid nanoparticles-From liposomes to mRNA vaccine delivery, a landscape of research diversity and advancement. ACS Nano 2021, 15, 16982–17015. [Google Scholar] [CrossRef]
  75. Cheng, X.W.; Lee, R.J. The role of helper lipids in lipid nanoparticles (LNPs) designed for oligonucleotide delivery. Adv. Drug Deliv. Rev. 2016, 99, 129–137. [Google Scholar] [CrossRef]
  76. Berraondo, P.; Martini, P.G.V.; Avila, M.A.; Fontanellas, A. Messenger RNA therapy for rare genetic metabolic diseases. Gut 2019, 68, 1323–1330. [Google Scholar] [CrossRef]
  77. Patel, S.; Ryals, R.C.; Weller, K.K.; Pennesi, M.E.; Sahay, G. Lipid nanoparticles for delivery of messenger RNA to the back of the eye. J. Control. Release 2019, 303, 91–100. [Google Scholar] [CrossRef] [PubMed]
  78. Afsharzadeh, M.; Hashemi, M.; Mokhtarzadeh, A.; Abnous, K.; Ramezani, M. Recent advances in co-delivery systems based on polymeric nanoparticle for cancer treatment. Artif. Cells Nanomed. Biotechnol. 2018, 46, 1095–1110. [Google Scholar] [CrossRef]
  79. Patra, J.K.; Das, G.; Fraceto, L.F.; Campos, E.V.R.; Rodriguez-Torres, M.D.P.; Acosta-Torres, L.S.; Diaz-Torres, L.A.; Grillo, R.; Swamy, M.K.; Sharma, S.; et al. Nano based drug delivery systems: Recent developments and future prospects. J. Nanobiotechnol. 2018, 16, 71. [Google Scholar] [CrossRef]
  80. Nguyen, J.; Steele, T.W.J.; Merkel, O.; Reul, R.; Kissel, T. Fast degrading polyesters as siRNA nano-carriers for pulmonary gene therapy. J. Control. Release 2008, 132, 243–251. [Google Scholar] [CrossRef] [PubMed]
  81. Brown, S.B.; Wang, L.; Jungels, R.R.; Sharma, B. Effects of cartilage-targeting moieties on nanoparticle biodistribution in healthy and osteoarthritic joints. Acta Biomater. 2020, 101, 469–483. [Google Scholar] [CrossRef] [PubMed]
  82. Zhang, C.-x.; Cheng, Y.; Liu, D.-z.; Liu, M.; Cui, H.; Zhang, B.-l.; Mei, Q.-b.; Zhou, S.-y. Mitochondria-targeted cyclosporin A delivery system to treat myocardial ischemia reperfusion injury of rats. J. Nanobiotechnol. 2019, 17, 18. [Google Scholar] [CrossRef]
  83. Dong, Y.; Ng, W.K.; Shen, S.; Kim, S.; Tan, R.B.H. Scalable ionic gelation synthesis of chitosan nanoparticles for drug delivery in static mixers. Carbohydr. Polym. 2013, 94, 940–945. [Google Scholar] [CrossRef]
  84. He, C.B.; Yue, H.M.; Xu, L.; Liu, Y.F.; Song, Y.D.; Tang, C.; Yin, C.H. siRNA release kinetics from polymeric nanoparticles correlate with RNAi efficiency and inflammation therapy via oral delivery. Acta Biomater. 2020, 103, 213–222. [Google Scholar] [CrossRef]
  85. Zhang, L.B.; Beatty, A.; Lu, L.; Abdalrahman, A.; Makris, T.M.; Wang, G.R.; Wang, Q. Microfluidic-assisted polymer-protein assembly to fabricate homogeneous functionalnanoparticles. Mater. Sci. Eng. C-Mater. Biol. Appl. 2020, 111, 110768. [Google Scholar] [CrossRef] [PubMed]
  86. Kucuk, I.; Edirisinghe, M. Microfluidic preparation of polymer nanospheres. J. Nanopart. Res. 2014, 16, 2626. [Google Scholar] [CrossRef]
  87. Palmerston Mendes, L.; Pan, J.; Torchilin, V.P. Dendrimers as nanocarriers for nucleic acid and drug delivery in cancer therapy. Molecules 2017, 22, 1401. [Google Scholar] [CrossRef] [PubMed]
  88. Abbasi, E.; Aval, S.F.; Akbarzadeh, A.; Milani, M.; Nasrabadi, H.T.; Joo, S.W.; Hanifehpour, Y.; Nejati-Koshki, K.; Pashaei-Asl, R. Dendrimers: Synthesis, applications, and properties. Nanoscale Res. Lett. 2014, 9, 247. [Google Scholar] [CrossRef] [PubMed]
  89. Ghosh, B.; Biswas, S. Polymeric micelles in cancer therapy: State of the art. J. Control. Release 2021, 332, 127–147. [Google Scholar] [CrossRef]
  90. Rideau, E.; Dimova, R.; Schwille, P.; Wurm, F.R.; Landfester, K. Liposomes and polymersomes: A comparative review towards cell mimicking. Chem. Soc. Rev. 2018, 47, 8572–8610. [Google Scholar] [CrossRef]
  91. Thambi, T.; Deepagan, V.G.; Ko, H.; Lee, D.S.; Park, J.H. Bioreducible polymersomes for intracellular dual-drug delivery. J. Mater. Chem. 2012, 22, 22028–22036. [Google Scholar] [CrossRef]
  92. Kim, H.O.; Kim, E.; An, Y.; Choi, J.; Jang, E.; Choi, E.B.; Kukreja, A.; Kim, M.H.; Kang, B.; Kim, D.J.; et al. A biodegradable polymersome containing Bcl-xL siRNA and doxorubicin as a dual delivery vehicle for a synergistic anticancer effect. Macromol. Biosci. 2013, 13, 745–754. [Google Scholar] [CrossRef]
  93. Huang, Z.H.; Teng, W.; Liu, L.S.; Wang, L.C.; Wang, Q.M.; Dong, Y.G. Efficient cytosolic delivery mediated by polymersomes facilely prepared from a degradable, amphiphilic, and amphoteric copolymer. Nanotechnology 2013, 24, 265104. [Google Scholar] [CrossRef]
  94. Gouveia, M.G.; Wesseler, J.P.; Ramaekers, J.; Weder, C.; Scholten, P.B.V.; Bruns, N. Polymersome-based protein drug delivery—Quo vadis? Chem. Soc. Rev. 2023, 52, 728–778. [Google Scholar] [CrossRef]
  95. Hamidi, M.; Azadi, A.; Rafiei, P. Hydrogel nanoparticles in drug delivery. Adv. Drug Deliv. Rev. 2008, 60, 1638–1649. [Google Scholar] [CrossRef] [PubMed]
  96. Nunes, D.; Andrade, S.; Ramalho, M.J.; Loureiro, J.A.; Pereira, M.C. Polymeric nanoparticles-loaded hydrogels for biomedical applications: A Systematic review on in vivo findings. Polymers 2022, 14, 1010. [Google Scholar] [CrossRef]
  97. Sonkar, C.; Ranjan, R.; Mukhopadhyay, S. Inorganic nanoparticle-based nanogels and their biomedical applications. Dalton Trans. 2025, 54, 6346–6360. [Google Scholar] [CrossRef]
  98. Kim, D.; Kim, J.; Park, Y.I.; Lee, N.; Hyeon, T. Recent development of inorganic nanoparticles for biomedical imaging. ACS Cent. Sci. 2018, 4, 324–336. [Google Scholar] [CrossRef] [PubMed]
  99. Wu, L.; Wen, W.; Wang, X.; Huang, D.; Cao, J.; Qi, X.; Shen, S. Ultrasmall iron oxide nanoparticles cause significant toxicity by specifically inducing acute oxidative stress to multiple organs. Part. Fibre Toxicol. 2022, 19, 24. [Google Scholar] [CrossRef] [PubMed]
  100. Bonvalot, S.; Rutkowski, P.L.; Thariat, J.; Carrère, S.; Ducassou, A.; Sunyach, M.-P.; Agoston, P.; Hong, A.; Mervoyer, A.; Rastrelli, M.; et al. NBTXR3, a first-in-class radioenhancer hafnium oxide nanoparticle, plus radiotherapy versus radiotherapy alone in patients with locally advanced soft-tissue sarcoma (Act.In.Sarc): A multicentre, phase 2–3, randomised, controlled trial. Lancet Oncol. 2019, 20, 1148–1159. [Google Scholar] [CrossRef]
  101. Yang, W.; Liang, H.; Ma, S.; Wang, D.; Huang, J. Gold nanoparticle based photothermal therapy: Development and application for effective cancer treatment. Sustain. Mater. Technol. 2019, 22, e00109. [Google Scholar] [CrossRef]
  102. Wang, J.X.; Potocny, A.M.; Rosenthal, J.; Day, E.S. Gold nanoshell-linear tetrapyrrole conjugates for near infrared-activated dual photodynamic and photothermal therapies. ACS Omega 2020, 5, 926–940. [Google Scholar] [CrossRef]
  103. Falagan-Lotsch, P.; Grzincic, E.M.; Murphy, C.J. One low-dose exposure of gold nanoparticles induces long-term changes in human cells. Proc. Natl. Acad. Sci. USA 2016, 113, 13318–13323. [Google Scholar] [CrossRef]
  104. Stern, J.M.; Kibanov Solomonov, V.V.; Sazykina, E.; Schwartz, J.A.; Gad, S.C.; Goodrich, G.P. Initial evaluation of the safety of nanoshell-directed photothermal therapy in the treatment of prostate disease. Int. J. Toxicol. 2016, 35, 38–46. [Google Scholar] [CrossRef]
  105. Rastinehad, A.R.; Anastos, H.; Wajswol, E.; Winoker, J.S.; Sfakianos, J.P.; Doppalapudi, S.K.; Carrick, M.R.; Knauer, C.J.; Taouli, B.; Lewis, S.C.; et al. Gold nanoshell-localized photothermal ablation of prostate tumors in a clinical pilot device study. Proc. Natl. Acad. Sci. USA 2019, 116, 18590–18596. [Google Scholar] [CrossRef] [PubMed]
  106. Kumthekar, P.; Ko, C.H.; Paunesku, T.; Dixit, K.; Sonabend, A.M.; Bloch, O.; Tate, M.; Schwartz, M.; Zuckerman, L.; Lezon, R.; et al. A first-in-human phase 0 clinical study of RNA interference–based spherical nucleic acids in patients with recurrent glioblastoma. Sci. Transl. Med. 2021, 13, eabb3945. [Google Scholar] [CrossRef] [PubMed]
  107. Jang, J.; Nah, H.; Lee, J.-H.; Moon, S.H.; Kim, M.G.; Cheon, J. Critical enhancements of MRI contrast and hyperthermic effects by dopant-controlled magnetic nanoparticles. Angew. Chem.-Int. Edit. 2009, 48, 1234–1238. [Google Scholar] [CrossRef]
  108. Arias, L.S.; Pessan, J.P.; Vieira, A.P.M.; de Lima, T.M.T.; Delbem, A.C.B.; Monteiro, D.R. Iron oxide nanoparticles for biomedical applications: A perspective on synthesis, drugs, antimicrobial activity, and toxicity. Antibiotics 2018, 7, 46. [Google Scholar] [CrossRef]
  109. Gudkov, S.V.; Burmistrov, D.E.; Serov, D.A.; Rebezov, M.B.; Semenova, A.A.; Lisitsyn, A.B. Do iron oxide nanoparticles have significant antibacterial properties? Antibiotics 2021, 10, 884. [Google Scholar] [CrossRef]
  110. Kang, M.; Lim, C.-H.; Han, J.-H. Comparison of toxicity and deposition of nano-sized carbon black aerosol prepared with or without dispersing sonication. Toxicol. Res. 2013, 29, 121–127. [Google Scholar] [CrossRef]
  111. Mirshafiee, V.; Sun, B.B.; Chang, C.H.; Liao, Y.P.; Jiang, W.; Jiang, J.H.; Liu, X.S.; Wang, X.; Xia, T.; Nel, A.E. Toxicological profiling of metal oxide nanoparticles in liver context reveals pyroptosis in Kupffer cells and macrophages versus apoptosis in hepatocytes. ACS Nano 2018, 12, 3836–3852. [Google Scholar] [CrossRef] [PubMed]
  112. Uzhytchak, M.; Smolková, B.; Lunova, M.; Jirsa, M.; Frtús, A.; Kubinová, S.; Dejneka, A.; Lunov, O. Iron oxide nanoparticle-induced autophagic flux is regulated by interplay between p53-mTOR axis and Bcl-2 signaling in hepatic cells. Cells 2020, 9, 1015. [Google Scholar] [CrossRef]
  113. 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]
  114. Arvizo, R.; Resham, B.; Mukherjee, P. Gold nanoparticles: Opportunities and challenges in nanomedicine. Expert Opin. Drug Deliv. 2010, 7, 753–763. [Google Scholar] [CrossRef]
  115. Cui, M.; Wiraja, C.; Qi, L.W.; Ting, S.C.W.; Jana, D.; Zheng, M.; Hu, X.; Xu, C. One-step synthesis of amine-coated ultra-small mesoporous silica nanoparticles. Nano Res. 2020, 13, 1592–1596. [Google Scholar] [CrossRef]
  116. Liu, Y.; Zhao, Y.L.; Sun, B.Y.; Chen, C.Y. Understanding the toxicity of carbon nanotubes. Accounts Chem. Res. 2013, 46, 702–713. [Google Scholar] [CrossRef] [PubMed]
  117. Carnovale, C.; Bryant, G.; Shukla, R.; Bansal, V. Identifying trends in gold nanoparticle toxicity and uptake: Size, shape, capping ligand, and biological corona. ACS Omega 2019, 4, 242–256. [Google Scholar] [CrossRef]
  118. Wang, D.; Peng, Y.; Li, Y.; Kpegah, J.K.S.K.; Chen, S. Multifunctional inorganic biomaterials: New weapons targeting osteosarcoma. Front. Mol. Biosci. 2023, 9, 1105540. [Google Scholar] [CrossRef]
  119. Della Camera, G.; Liu, T.; Yang, W.; Li, Y.; Puntes, V.F.; Gioria, S.; Italiani, P.; Boraschi, D. Induction of innate memory in human monocytes exposed to mixtures of bacterial agents and nanoparticles. Int. J. Mol. Sci. 2022, 23, 14655. [Google Scholar] [CrossRef] [PubMed]
  120. Dhawan, A.; Sharma, V. Toxicity assessment of nanomaterials: Methods and challenges. Anal. Bioanal. Chem. 2010, 398, 589–605. [Google Scholar] [CrossRef]
  121. Öztürk, K.; Kaplan, M.; Çalis, S. Effects of nanoparticle size, shape, and zeta potential on drug delivery. Int. J. Pharm. 2024, 666, 124799. [Google Scholar] [CrossRef] [PubMed]
  122. Awashra, M.; Mlynarz, P. The toxicity of nanoparticles and their interaction with cells: An in vitro metabolomic perspective. Nanoscale Adv. 2023, 5, 2674–2723. [Google Scholar] [CrossRef]
  123. He, C.B.; Hu, Y.P.; Yin, L.C.; Tang, C.; Yin, C.H. Effects of particle size and surface charge on cellular uptake and biodistribution of polymeric nanoparticles. Biomaterials 2010, 31, 3657–3666. [Google Scholar] [CrossRef]
  124. Bai, X.; Liu, F.; Liu, Y.; Li, C.; Wang, S.Q.; Zhou, H.Y.; Wang, W.Y.; Zhu, H.; Winkler, D.A.; Yan, B. Toward a systematic exploration of nano-bio interactions. Toxicol. Appl. Pharmacol. 2017, 323, 66–73. [Google Scholar] [CrossRef]
  125. Cheng, L.-C.; Jiang, X.; Wang, J.; Chen, C.; Liu, R.-S. Nano–bio effects: Interaction of nanomaterials with cells. Nanoscale 2013, 5, 3547–3569. [Google Scholar] [CrossRef] [PubMed]
  126. Huo, S.D.; Jin, S.B.; Ma, X.W.; Xue, X.D.; Yang, K.N.; Kumar, A.; Wang, P.C.; Zhang, J.C.; Hu, Z.B.; Liang, X.J. Ultrasmall gold nanoparticles as carriers for nucleus-based gene therapy due to size-dependent nuclear entry. ACS Nano 2014, 8, 5852–5862. [Google Scholar] [CrossRef] [PubMed]
  127. Foroozandeh, P.; Aziz, A.A. Insight into cellular uptake and intracellular trafficking of nanoparticles. Nanoscale Res. Lett. 2018, 13, 339. [Google Scholar] [CrossRef]
  128. Rennick, J.J.; Johnston, A.P.R.; Parton, R.G. Key principles and methods for studying the endocytosis of biological and nanoparticle therapeutics. Nat. Nanotechnol. 2021, 16, 266–276. [Google Scholar] [CrossRef]
  129. Donahue, N.D.; Acar, H.; Wilhelm, S. Concepts of nanoparticle cellular uptake, intracellular trafficking, and kinetics in nanomedicine. Adv. Drug Deliv. Rev. 2019, 143, 68–96. [Google Scholar] [CrossRef]
  130. Zhu, M.T.; Nie, G.J.; Meng, H.; Xia, T.; Nel, A.; Zhao, Y.L. Physicochemical properties determine nanomaterial cellular uptake, transport, and fate. Acc. Chem. Res. 2013, 46, 622–631. [Google Scholar] [CrossRef]
  131. Jiang, W.; Kim, B.Y.S.; Rutka, J.T.; Chan, W.C.W. Nanoparticle-mediated cellular response is size-dependent. Nat. Nanotechnol. 2008, 3, 145–150. [Google Scholar] [CrossRef] [PubMed]
  132. Chaparro, D.; Goudeli, E. Design of engineered nanoparticles for biomedical applications by computational modeling. Nanoscale 2025, 17, 9705–9737. [Google Scholar] [CrossRef]
  133. Dolai, J.; Mandal, K.; Jana, N.R. Nanoparticle size effects in biomedical applications. ACS Appl. Nano Mater. 2021, 4, 6471–6496. [Google Scholar] [CrossRef]
  134. Zhang, Y.N.; 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]
  135. Renwick, L.C.; Donaldson, K.; Clouter, A. Impairment of alveolar macrophage phagocytosis by ultrafine particles. Toxicol. Appl. Pharmacol. 2001, 172, 119–127. [Google Scholar] [CrossRef]
  136. Buzea, C.; Pacheco, I.I.; Robbie, K. Nanomaterials and nanoparticles: Sources and toxicity. Biointerphases 2007, 2, MR17–MR71. [Google Scholar] [CrossRef] [PubMed]
  137. Bakhshi, S.; Shoari, A.; Alibolandi, P.; Ganji, M.; Ghazy, E.; Rahdar, A.; Fathi-karkan, S.; Pandey, S. Emerging innovations in vincristine-encapsulated nanoparticles: Pioneering a new era in oncological therapeutics. J. Drug Deliv. Sci. Technol. 2024, 91, 105270. [Google Scholar] [CrossRef]
  138. Kim, I.-Y.; Joachim, E.; Choi, H.; Kim, K. Toxicity of silica nanoparticles depends on size, dose, and cell type. Nanomed. Nanotechnol. Biol. Med. 2015, 11, 1407–1416. [Google Scholar] [CrossRef]
  139. Chen, L.Q.; Fang, L.; Ling, J.; Ding, C.Z.; Kang, B.; Huang, C.Z. Nanotoxicity of silver nanoparticles to red blood cells: Size dependent adsorption, uptake, and hemolytic activity. Chem. Res. Toxicol. 2015, 28, 501–509. [Google Scholar] [CrossRef] [PubMed]
  140. Huang, X.L.; Li, L.L.; Liu, T.L.; Hao, N.J.; Liu, H.Y.; Chen, D.; Tang, F.Q. The shape effect of mesoporous silica nanoparticles on biodistribution, clearance, and biocompatibility. ACS Nano 2011, 5, 5390–5399. [Google Scholar] [CrossRef]
  141. Gratton, S.E.A.; Ropp, P.A.; Pohlhaus, P.D.; Luft, J.C.; Madden, V.J.; Napier, M.E.; DeSimone, J.M. The effect of particle design on cellular internalization pathways. Proc. Natl. Acad. Sci. USA 2008, 105, 11613–11618. [Google Scholar] [CrossRef]
  142. Kus-Liskiewicz, M.; Fickers, P.; Ben Tahar, I. Biocompatibility and cytotoxicity of gold nanoparticles: Recent advances in methodologies and regulations. Int. J. Mol. Sci. 2021, 22, 10952. [Google Scholar] [CrossRef]
  143. Li, L.L.; Liu, T.L.; Fu, C.H.; Tan, L.F.; Meng, X.W.; Liu, H.Y. Biodistribution, excretion, and toxicity of mesoporous silica nanoparticles after oral administration depend on their shape. Nanomed. Nanotechnol. Biol. Med. 2015, 11, 1915–1924. [Google Scholar] [CrossRef]
  144. Li, Y.; Kröger, M.; Liu, W.K. Shape effect in cellular uptake of PEGylated nanoparticles: Comparison between sphere, rod, cube and disk. Nanoscale 2015, 7, 16631–16646. [Google Scholar] [CrossRef]
  145. Sharifi, S.; Behzadi, S.; Laurent, S.; Laird Forrest, M.; Stroeve, P.; Mahmoudi, M. Toxicity of nanomaterials. Chem. Soc. Rev. 2012, 41, 2323–2343. [Google Scholar] [CrossRef] [PubMed]
  146. Yan, L.; Zhao, F.; Wang, J.; Zu, Y.; Gu, Z.; Zhao, Y. A safe-by-design strategy towards safer nanomaterials in nanomedicines. Adv. Mater. 2019, 31, 1805391. [Google Scholar] [CrossRef] [PubMed]
  147. Wu, L.; Shan, W.; Zhang, Z.R.; Huang, Y. Engineering nanomaterials to overcome the mucosal barrier by modulating surface properties. Adv. Drug Deliv. Rev. 2018, 124, 150–163. [Google Scholar] [CrossRef]
  148. Li, M.; Zhang, H.; Hou, Y.; Wang, X.; Xue, C.; Li, W.; Cai, K.; Zhao, Y.; Luo, Z. State-of-the-art iron-based nanozymes for biocatalytic tumor therapy. Nanoscale Horiz. 2020, 5, 202–217. [Google Scholar] [CrossRef]
  149. Kiaie, S.H.; Zolbanin, N.M.; Ahmadi, A.; Bagherifar, R.; Valizadeh, H.; Kashanchi, F.; Jafari, R. Recent advances in mRNA-LNP therapeutics: Immunological and pharmacological aspects. J. Nanobiotechnol. 2022, 20, 276. [Google Scholar] [CrossRef]
  150. Cai, R.; Ren, J.Y.; Ji, Y.L.; Wang, Y.L.; Liu, Y.; Chen, Z.G.; Sabet, Z.F.; Wu, X.C.; Lynch, I.; Chen, C.Y. Corona of thorns: The surface chemistry-mediated protein corona perturbs the recognition and immune response of macrophages. ACS Appl. Mater. Interfaces 2020, 12, 1997–2008. [Google Scholar] [CrossRef]
  151. Augustine, R.; Hasan, A.; Primavera, R.; Wilson, R.J.; Thakor, A.S.; Kevadiya, B.D. Cellular uptake and retention of nanoparticles: Insights on particle properties and interaction with cellular components. Mater. Today Commun. 2020, 25, 101692. [Google Scholar] [CrossRef]
  152. Jerzykiewicz, J.; Czogalla, A. Polyethyleneimine-based lipopolyplexes as carriers in anticancer gene therapies. Materials 2022, 15, 179. [Google Scholar] [CrossRef] [PubMed]
  153. Manshian, B.B.; Pokhrel, S.; Mädler, L.; Soenen, S.J. The impact of nanoparticle-driven lysosomal alkalinization on cellular functionality. J. Nanobiotechnol. 2018, 16, 85. [Google Scholar] [CrossRef]
  154. Vukovic, B.; Milic, M.; Dobrosevic, B.; Milic, M.; Ilic, K.; Pavicic, I.; Seric, V.; Vrcek, I.V. Surface stabilization affects toxicity of silver nanoparticles in human peripheral blood mononuclear cells. Nanomaterials 2020, 10, 1390. [Google Scholar] [CrossRef]
  155. Mihalache, R.; Verbeek, J.; Graczyk, H.; Murashov, V.; van Broekhuizen, P. Occupational exposure limits for manufactured nanomaterials, a systematic review. Nanotoxicology 2017, 11, 7–19. [Google Scholar] [CrossRef]
  156. Kuhlbusch, T.A.J.; Asbach, C.; Fissan, H.; Göhler, D.; Stintz, M. Nanoparticle exposure at nanotechnology workplaces: A review. Part. Fibre Toxicol. 2011, 8, 22. [Google Scholar] [CrossRef] [PubMed]
  157. Missaoui, W.N.; Arnold, R.D.; Cummings, B.S. Toxicological status of nanoparticles: What we know and what we don’t know. Chem.-Biol. Interact. 2018, 295, 1–12. [Google Scholar] [CrossRef]
  158. 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] [PubMed]
  159. Bruinink, A.; Wang, J.; Wick, P. Effect of particle agglomeration in nanotoxicology. Arch. Toxicol. 2015, 89, 659–675. [Google Scholar] [CrossRef] [PubMed]
  160. Sridhar, K.; Inbaraj, B.S.; Chen, B.H. Recent advances on nanoparticle based strategies for improving carotenoid stability and biological activity. Antioxidants 2021, 10, 713. [Google Scholar] [CrossRef]
  161. Biola-Clier, M.; Beal, D.; Caillat, S.; Libert, S.; Armand, L.; Herlin-Boime, N.; Sauvaigo, S.; Douki, T.; Carriere, M. Comparison of the DNA damage response in BEAS-2B and A549 cells exposed to titanium dioxide nanoparticles. Mutagenesis 2017, 32, 161–172. [Google Scholar] [CrossRef]
  162. Ke, P.C.; Lin, S.; Parak, W.J.; Davis, T.P.; Caruso, F. A decade of the protein corona. ACS Nano 2017, 11, 11773–11776. [Google Scholar] [CrossRef]
  163. Barz, M.; Parak, W.J.; Zentel, R. Concepts and approaches to reduce or avoid protein corona formation on nanoparticles: Challenges and opportunities. Adv. Sci. 2024, 11, e2402935. [Google Scholar] [CrossRef]
  164. Wheeler, K.E.; Chetwynd, A.J.; Fahy, K.M.; Hong, B.S.; Tochihuitl, J.A.; Foster, L.A.; Lynch, I. Environmental dimensions of the protein corona. Nat. Nanotechnol. 2021, 16, 617–629. [Google Scholar] [CrossRef]
  165. Baimanov, D.; Cai, R.; Chen, C.Y. Understanding the chemical nature of nanoparticle-protein interactions. Bioconjug. Chem. 2019, 30, 1923–1937. [Google Scholar] [CrossRef]
  166. Stepien, G.; Moros, M.; Pérez-Hernández, M.; Monge, M.; Gutiérrez, L.; Fratila, R.M.; Heras, M.D.; Guillén, S.M.; Lanzarote, J.J.P.; Solans, C.; et al. Effect of surface chemistry and associated protein corona on the long-term biodegradation of iron oxide nanoparticles in vivo. ACS Appl. Mater. Interfaces 2018, 10, 4548–4560. [Google Scholar] [CrossRef] [PubMed]
  167. Smolková, B.; MacCulloch, T.; Rockwood, T.F.; Liu, M.H.; Henry, S.J.W.; Frtús, A.; Uzhytchak, M.; Lunova, M.; Hof, M.; Jurkiewicz, P.; et al. Protein corona inhibits endosomal escape of functionalized DNA nanostructures in living cells. ACS Appl. Mater. Interfaces 2021, 13, 46375–46390. [Google Scholar] [CrossRef] [PubMed]
  168. Lesniak, A.; Salvati, A.; Santos-Martinez, M.J.; Radomski, M.W.; Dawson, K.A.; Åberg, C. Nanoparticle adhesion to the cell membrane and its effect on nanoparticle uptake efficiency. J. Am. Chem. Soc. 2013, 135, 1438–1444. [Google Scholar] [CrossRef]
  169. Wang, L.; Hartel, N.; Ren, K.X.; Graham, N.A.; Malmstadt, N. Effect of protein corona on nanoparticle-plasma membrane and nanoparticle-biomimetic membrane interactions. Environ. Sci. Nano 2020, 7, 963–974. [Google Scholar] [CrossRef]
  170. Persaud, I.; Shannahan, J.H.; Raghavendra, A.J.; Alsaleh, N.B.; Podila, R.; Brown, J.M. Biocorona formation contributes to silver nanoparticle induced endoplasmic reticulum stress. Ecotoxicol. Environ. Saf. 2019, 170, 77–86. [Google Scholar] [CrossRef]
  171. Huang, Z.; Wu, L.; Wang, W.; Zhou, Y.; Zhang, X.; Huang, Y.; Pan, X.; Wu, C. Unraveling the pulmonary drug delivery carriers in inhalable nanostructures. J. Nanopart. Res. 2022, 24, 10. [Google Scholar] [CrossRef]
  172. Tan, Y.; Zhu, X.Y.; Wu, D.; Song, E.Q.; Song, Y. Compromised autophagic effect of polystyrene nanoplastics mediated by protein corona was recovered after lysosomal degradation of corona. Environ. Sci. Technol. 2020, 54, 11485–11493. [Google Scholar] [CrossRef]
  173. Adhipandito, C.F.; Cheung, S.H.; Lin, Y.H.; Wu, S.H. Atypical renal clearance of nanoparticles larger than the kidney filtration threshold. Int. J. Mol. Sci. 2021, 22, 11182. [Google Scholar] [CrossRef]
  174. Sohal, I.S.; Cho, Y.K.; O’Fallon, K.S.; Gaines, P.; Demokritou, P.; Bello, D. Dissolution behavior and biodurability of ingested engineered nanomaterials in the gastrointestinal environment. ACS Nano 2018, 12, 8115–8128. [Google Scholar] [CrossRef]
  175. Lee, S.H.; Jun, B.H. Silver nanoparticles: Synthesis and application for nanomedicine. Int. J. Mol. Sci. 2019, 20, 865. [Google Scholar] [CrossRef] [PubMed]
  176. Egbuna, C.; Parmar, V.K.; Jeevanandam, J.; Ezzat, S.M.; Patrick-Iwuanyanwu, K.C.; Adetunji, C.O.; Khan, J.; Onyeike, E.N.; Uche, C.Z.; Akram, M.; et al. Toxicity of nanoparticles in biomedical application: Nanotoxicology. J. Toxicol. 2021, 2021, 954443. [Google Scholar] [CrossRef] [PubMed]
  177. Wang, L.M.; Zhang, T.L.; Li, P.Y.; Huang, W.X.; Tang, J.L.; Wang, P.Y.; Liu, J.; Yuan, Q.X.; Bai, R.; Li, B.; et al. Use of synchrotron radiation-analytical techniques to reveal chemical origin of silver-nanoparticle cytotoxicity. ACS Nano 2015, 9, 6532–6547. [Google Scholar] [CrossRef] [PubMed]
  178. Bondarenko, O.; Juganson, K.; Ivask, A.; Kasemets, K.; Mortimer, M.; Kahru, A. Toxicity of Ag, CuO and ZnO nanoparticles to selected environmentally relevant test organisms and mammalian cells in vitro: A critical review. Arch. Toxicol. 2013, 87, 1181–1200. [Google Scholar] [CrossRef]
  179. Khanna, P.; Ong, C.; Bay, B.H.; Baeg, G.H. Nanotoxicity: An interplay of oxidative stress, inflammation and cell death. Nanomaterials 2015, 5, 1163–1180. [Google Scholar] [CrossRef]
  180. Won, Y.Y.; Sharma, R.; Konieczny, S.F. Missing pieces in understanding the intracellular trafficking of polycation/DNA complexes. J. Control. Release 2009, 139, 88–93. [Google Scholar] [CrossRef]
  181. Liu, N.; Tang, M. Toxic effects and involved molecular pathways of nanoparticles on cells and subcellular organelles. J. Appl.Toxicol. 2020, 40, 16–36. [Google Scholar] [CrossRef]
  182. Lee, E.; Lee, M.; Kwon, S.; Kim, J.; Kwon, Y. Systematic and mechanistic analysis of AuNP-induced nanotoxicity for risk assessment of nanomedicine. Nano Converg. 2022, 9, 27. [Google Scholar] [CrossRef]
  183. Chen, M.; Wu, T.S. Nanoparticles and neurodegeneration: Insights on multiple pathways of programmed cell death regulated by nanoparticles. Sci. Total Environ. 2024, 912, 168739. [Google Scholar] [CrossRef]
  184. Nowak-Jary, J.; Machnicka, B. Comprehensive analysis of the potential toxicity of magnetic iron oxide nanoparticles for medical applications: Cellular mechanisms and systemic effects. Int. J. Mol. Sci. 2024, 25, 12013. [Google Scholar] [CrossRef]
  185. Xu, B.; Wu, S.; Wang, Y.; Ji, Y.; Liang, S.; Wang, C.; Tian, X. Paradoxical roles of carbon nanotubes in cancer therapy and carcinogenesis. J. Nanotheranostics 2024, 5, 84–98. [Google Scholar] [CrossRef]
  186. Aula, S.; Lakkireddy, S.; Jamil, K.; Kapley, A.; Swamy, A.V.N.; Lakkireddy, H.R. Biophysical, biopharmaceutical and toxicological significance of biomedical nanoparticles. RSC Adv. 2015, 5, 47830–47859. [Google Scholar] [CrossRef]
  187. Winnik, F.M.; Maysinger, D. Quantum dot cytotoxicity and ways to reduce it. Acc. Chem. Res. 2013, 46, 672–680. [Google Scholar] [CrossRef]
  188. Liang, M.M.; Fan, K.L.; Zhou, M.; Duan, D.M.; Zheng, J.Y.; Yang, D.L.; Feng, J.; Yan, X.Y. H-ferritin-nanocaged doxorubicin nanoparticles specifically target and kill tumors with a single-dose injection. Proc. Natl. Acad. Sci. USA 2014, 111, 14900–14905. [Google Scholar] [CrossRef]
  189. Chakraborty, S.; Chakraborty, A.; Mukherjee, B.; Besra, S.E.; Dewanjee, S.; Mukherjee, A.; Sen, R.; Ojha, P.K.; Kumar, V.; Shaw, T.K.; et al. Assessment of superiority of HSP70-targeting aptamer-functionalized drug-nanocarrier over non-targeted commercially available counterpart in HCC therapy: In vitro and in vivo investigations and molecular modeling. Life Sci. 2023, 317, 121467. [Google Scholar] [CrossRef] [PubMed]
  190. Kurzatkowska, K.; Pazos, M.A.; Herschkowitz, J.I.; Hepel, M. Cancer-targeted controlled delivery of chemotherapeutic anthracycline derivatives using apoferritin nanocage carriers. Int. J. Mol. Sci. 2021, 22, 1362. [Google Scholar] [CrossRef] [PubMed]
  191. Scomparin, A.; Salmaso, S.; Eldar-Boock, A.; Ben-Shushan, D.; Ferber, S.; Tiram, G.; Shmeeda, H.; Landa-Rouben, N.; Leor, J.; Caliceti, P.; et al. A comparative study of folate receptor-targeted doxorubicin delivery systems: Dosing regimens and therapeutic index. J. Control. Release 2015, 208, 106–120. [Google Scholar] [CrossRef]
  192. Maksimenko, A.; Dosio, F.; Mougin, J.; Ferrero, A.; Wack, S.; Reddy, L.H.; Weyn, A.A.; Lepeltier, E.; Bourgaux, C.; Stella, B.; et al. A unique squalenoylated and nonpegylated doxorubicin nanomedicine with systemic long-circulating properties and anticancer activity. Proc. Natl. Acad. Sci. USA 2014, 111, E217–E226. [Google Scholar] [CrossRef]
  193. Man, F.; Lammers, T.; de Rosales, R.T.M. Imaging nanomedicine-based drug delivery: A review of clinical studies. Mol. Imaging. Biol. 2018, 20, 683–695. [Google Scholar] [CrossRef]
  194. Prabhakar, U.; Maeda, H.; Jain, R.K.; Sevick-Muraca, E.M.; Zamboni, W.; Farokhzad, O.C.; Barry, S.T.; Gabizon, A.; Grodzinski, P.; Blakey, D.C. Challenges and key considerations of the enhanced permeability and retention effect for nanomedicine drug delivery in oncology. Cancer Res. 2013, 73, 2412–2417. [Google Scholar] [CrossRef]
  195. Perry, J.L.; Reuter, K.G.; Luft, J.C.; Pecot, C.V.; Zamboni, W.; DeSimone, J.M. Mediating passive tumor accumulation through particle size, tumor type, and location. Nano Lett. 2017, 17, 2879–2886. [Google Scholar] [CrossRef] [PubMed]
  196. Ouyang, B.; Poon, W.; Zhang, Y.N.; Lin, Z.P.; Kingston, B.R.; Tavares, A.J.; Zhang, Y.W.; Chen, J.; Valic, M.S.; Syed, A.M.; et al. The dose threshold for nanoparticle tumour delivery. Nat. Mater. 2020, 19, 1362–1371. [Google Scholar] [CrossRef] [PubMed]
  197. Nguyen, L.N.M.; Lin, Z.P.; Sindhwani, S.; MacMillan, P.; Mladjenovic, S.M.; Stordy, B.; Ngo, W.; Chan, W.C.W. The exit of nanoparticles from solid tumours. Nat. Mater. 2023, 22, 1261–1272. [Google Scholar] [CrossRef]
  198. Petty, H.R. Could nanoparticles that mimic the NADPH oxidase be used to kill tumor cells? Nanomedicine 2016, 11, 1631–1634. [Google Scholar] [CrossRef] [PubMed]
  199. Klaper, R.; Arndt, D.; Bozich, J.; Dominguez, G. Molecular interactions of nanomaterials and organisms: Defining biomarkers for toxicity and high-throughput screening using traditional and next-generation sequencing approaches. Analyst 2014, 139, 882–895. [Google Scholar] [CrossRef]
  200. Gupta, G.; Kaur, J.; Bhattacharya, K.; Chambers, B.J.; Gazzi, A.; Furesi, G.; Rauner, M.; Fuoco, C.; Orecchioni, M.; Delogu, L.G.; et al. Exploiting mass spectrometry to unlock the mechanism of nanoparticle-induced inflammasome activation. ACS Nano 2023, 17, 17451–17467. [Google Scholar] [CrossRef]
  201. Lunov, O.; Syrovets, T.; Loos, C.; Nienhaus, G.U.; Mailänder, V.; Landfester, K.; Rouis, M.; Simmet, T. Amino-functionalized polystyrene nanoparticles activate the NLRP3 inflammasome in human macrophages. ACS Nano 2011, 5, 9648–9657. [Google Scholar] [CrossRef]
  202. 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]
  203. Aschner, M.; Skalny, A.V.; Martins, A.C.; Tizabi, Y.; Zaitseva, I.P.; Santamaria, A.; Lu, R.Z.; Gluhcheva, Y.Y.; Tinkov, A.A. The role of NLRP3 inflammasome activation in proinflammatory and cytotoxic effects of metal nanoparticles. Arch. Toxicol. 2025, 99, 1287–1314. [Google Scholar] [CrossRef]
  204. Yazdi, A.S.; Guarda, G.; Riteau, N.; Drexler, S.K.; Tardivel, A.; Couillin, I.; Tschopp, J. Nanoparticles activate the NLR pyrin domain containing 3 (Nlrp3) inflammasome and cause pulmonary inflammation through release of IL-1α and IL-1β. Proc. Natl. Acad. Sci. USA 2010, 107, 19449–19454. [Google Scholar] [CrossRef]
  205. 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] [PubMed]
  206. Rangasami, V.K.; Samanta, S.; Parihar, V.S.; Asawa, K.; Zhu, K.Y.; Varghese, O.P.; Teramura, Y.; Nilsson, B.; Hilborn, J.; Harris, R.A.; et al. Harnessing hyaluronic acid-based nanoparticles for combination therapy: A novel approach for suppressing systemic inflammation and to promote antitumor macrophage polarization. Carbohydr. Polym. 2021, 254, 117291. [Google Scholar] [CrossRef] [PubMed]
  207. Sarfraz, M.; Roa, W.; Bou-Chacra, N.; Löbenberg, R. Inflammation caused by nanosized delivery systems: Is there a benefit? Mol. Pharm. 2016, 13, 3270–3278. [Google Scholar] [CrossRef]
  208. Xu, H.; Ren, D. Lysosomal physiology. Annu. Rev. Physiol. 2015, 77, 57–80. [Google Scholar] [CrossRef]
  209. Levada, K.; Pshenichnikov, S.; Omelyanchik, A.; Rodionova, V.; Nikitin, A.; Savchenko, A.; Schetinin, I.; Zhukov, D.; Abakumov, M.; Majouga, A.; et al. Progressive lysosomal membrane permeabilization induced by iron oxide nanoparticles drives hepatic cell autophagy and apoptosis. Nano Converg. 2020, 7, 17. [Google Scholar] [CrossRef]
  210. Yang, K.X.; Lu, Y.; Xie, F.Y.; Zou, H.; Fan, X.Y.; Li, B.H.; Li, W.; Zhang, W.; Mei, L.; Feng, S.; et al. Cationic liposomes induce cell necrosis through lysosomal dysfunction and late-stage autophagic flux inhibition. Nanomedicine 2016, 11, 3117–3137. [Google Scholar] [CrossRef]
  211. He, G.; Ma, Y.; Zhu, Y.; Yong, L.; Liu, X.; Wang, P.; Liang, C.; Yang, C.; Zhao, Z.; Hai, B.; et al. Cross talk between autophagy and apoptosis contributes to ZnO nanoparticle-induced human osteosarcoma cell death. Adv. Healthc. Mater. 2018, 7, 1800332. [Google Scholar] [CrossRef] [PubMed]
  212. Li, H.; Zhang, H.; He, X.; Zhao, P.; Wu, T.; Xiahou, J.; Wu, Y.; Liu, Y.; Chen, Y.; Jiang, X.; et al. Blocking spatiotemporal crosstalk between subcellular organelles for enhancing anticancer therapy with nanointercepters. Adv. Mater. 2023, 35, 2211597. [Google Scholar] [CrossRef]
  213. Li, J.J.; Zou, L.; Hartono, D.; Ong, C.N.; Bay, B.H.; Yung, L.Y.L. Gold nanoparticles induce oxidative damage in lung fibroblasts in vitro. Adv. Mater. 2008, 20, 138–142. [Google Scholar] [CrossRef]
  214. Avalos, A.; Haza, A.I.; Mateo, D.; Morales, P. Cytotoxicity and ROS production of manufactured silver nanoparticles of different sizes in hepatoma and leukemia cells. J. Appl. Toxicol. 2014, 34, 413–423. [Google Scholar] [CrossRef]
  215. Ahamed, M.; Posgai, R.; Gorey, T.J.; Nielsen, M.; Hussain, S.M.; Rowe, J.J. Silver nanoparticles induced heat shock protein 70, oxidative stress and apoptosis in Drosophila melanogaster. Toxicol. Appl. Pharm. 2010, 242, 263–269. [Google Scholar] [CrossRef]
  216. Guo, D.W.; Zhu, L.Y.; Huang, Z.H.; Zhou, H.X.; Ge, Y.; Ma, W.J.; Wu, J.; Zhang, X.Y.; Zhou, X.F.; Zhang, Y.; et al. Anti-leukemia activity of PVP-coated silver nanoparticles via generation of reactive oxygen species and release of silver ions. Biomaterials 2013, 34, 7884–7894. [Google Scholar] [CrossRef] [PubMed]
  217. Huo, L.L.; Chen, R.; Zhao, L.; Shi, X.F.; Bai, R.; Long, D.X.; Chen, F.; Zhao, Y.L.; Chang, Y.Z.; Chen, C.Y. Silver nanoparticles activate endoplasmic reticulum stress signaling pathway in cell and mouse models: The role in toxicity evaluation. Biomaterials 2015, 61, 307–315. [Google Scholar] [CrossRef]
  218. Choi, A.O.; Brown, S.E.; Szyf, M.; Maysinger, D. Quantum dot-induced epigenetic and genotoxic changes in human breast cancer cells. J. Mol. Med.-JMM 2008, 86, 291–302. [Google Scholar] [CrossRef]
  219. Lam, C.W.; James, J.T.; McCluskey, R.; Hunter, R.L. Pulmonary toxicity of single-wall carbon nanotubes in mice 7 and 90 days after intratracheal instillation. Toxicol. Sci. 2004, 77, 126–134. [Google Scholar] [CrossRef] [PubMed]
  220. Morimoto, Y.; Horie, M.; Kobayashi, N.; Shinohara, N.; Shimada, M. Inhalation toxicity assessment of carbon-based nanoparticles. Acc. Chem. Res. 2013, 46, 770–781. [Google Scholar] [CrossRef] [PubMed]
  221. Cui, D.X.; Tian, F.R.; Ozkan, C.S.; Wang, M.; Gao, H.J. Effect of single wall carbon nanotubes on human HEK293 cells. Toxicol. Lett. 2005, 155, 73–85. [Google Scholar] [CrossRef]
  222. Alazzam, A.; Mfoumou, E.; Stiharu, I.; Kassab, A.; Darnel, A.; Yasmeen, A.; Sivakumar, N.; Bhat, R.; Al Moustafa, A.E. Identification of deregulated genes by single wall carbon-nanotubes in human normal bronchial epithelial cells. Nanomed. Nanotechnol. Biol. Med. 2010, 6, 563–569. [Google Scholar] [CrossRef]
  223. Wang, J.Y.; Sun, P.P.; Bao, Y.M.; Liu, J.W.; An, L.J. Cytotoxicity of single-walled carbon nanotubes on PC12 cells. Toxicol. Vitro 2011, 25, 242–250. [Google Scholar] [CrossRef]
  224. Pacurari, M.; Yin, X.J.; Zhao, J.S.; Ding, M.; Leonard, S.S.; Schwegier-Berry, D.; Ducatman, B.S.; Sbarra, D.; Hoover, M.D.; Castranova, V.; et al. Raw single-wall carbon nanotubes induce oxidative stress and activate MAPKs, AP-1, NF-κB, and Akt in normal and malignant human mesothelial cells. Environ. Health Perspect. 2008, 116, 1211–1217. [Google Scholar] [CrossRef]
  225. Sarkar, A.; Sil, P.C. Iron oxide nanoparticles mediated cytotoxicity via PI3K/AKT pathway: Role of quercetin. Food Chem. Toxicol. 2014, 71, 106–115. [Google Scholar] [CrossRef]
  226. Shukla, S.; Jadaun, A.; Arora, V.; Sinha, R.K.; Biyani, N.; Jain, V.K. In vitro toxicity assessment of chitosan oligosaccharide coated iron oxide nanoparticles. Toxicol. Rep. 2015, 2, 27–39. [Google Scholar] [CrossRef] [PubMed]
  227. Park, E.J.; Park, K. Oxidative stress and pro-inflammatory responses induced by silica nanoparticles in vivo and in vitro. Toxicol. Rep. 2009, 184, 18–25. [Google Scholar] [CrossRef] [PubMed]
  228. Liu, X.; Sun, J.A. 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]
  229. Duan, J.C.; Yu, Y.B.; Yu, Y.; Wang, J.; Geng, W.J.; Jiang, L.Z.; Li, Q.L.; Zhou, X.Q.; Sun, Z.W. Silica nanoparticles induce autophagy and endothelial dysfunction via the PI3K/Akt/mTOR signaling pathway. Int. J. Nanomed. 2014, 9, 5131–5141. [Google Scholar] [CrossRef] [PubMed]
  230. Shrivastava, R.; Raza, S.; Yadav, A.; Kushwaha, P.; Flora, S.J.S. Effects of sub-acute exposure to TiO2, ZnO and Al2O3 nanoparticles on oxidative stress and histological changes in mouse liver and brain. Drug Chem. Toxicol. 2014, 37, 336–347. [Google Scholar] [CrossRef]
  231. Park, E.J.; Yi, J.; Chung, Y.H.; Ryu, D.Y.; Choi, J.; Park, K. Oxidative stress and apoptosis induced by titanium dioxide nanoparticles in cultured BEAS-2B cells. Toxicol. Lett. 2008, 180, 222–229. [Google Scholar] [CrossRef]
  232. González-Esquivel, A.E.; Charles-Niño, C.L.; Pacheco-Moisés, F.P.; Ortiz, G.G.; Jaramillo-Juárez, F.; Rincón-Sánchez, A.R. Beneficial effects of quercetin on oxidative stress in liver and kidney induced by titanium dioxide (TiO) nanoparticles in rats. Toxicol. Mech. Methods 2015, 25, 166–175. [Google Scholar] [CrossRef]
  233. Roy, R.; Parashar, V.; Chauhan, L.K.S.; Shanker, R.; Das, M.; Tripathi, A.; Dwivedi, P.D. Mechanism of uptake of ZnO nanoparticles and inflammatory responses in macrophages require PI3K mediated MAPKs signaling. Toxicol. Vitr. 2014, 28, 457–467. [Google Scholar] [CrossRef]
  234. Fukui, H.; Horie, M.; Endoh, S.; Kato, H.; Fujita, K.; Nishio, K.; Komaba, L.K.; Maru, J.; Miyauhi, A.; Nakamura, A.; et al. Association of zinc ion release and oxidative stress induced by intratracheal instillation of ZnO nanoparticles to rat lung. Chem.-Biol. Interact. 2012, 198, 29–37. [Google Scholar] [CrossRef]
  235. Wu, W.D.; Samet, J.M.; Peden, D.B.; Bromberg, P.A. Phosphorylation of p65 is required for zinc oxide nanoparticle-induced interleukin 8 expression in human bronchial epithelial cells. Environ. Health Perspect. 2010, 118, 982–987. [Google Scholar] [CrossRef] [PubMed]
  236. Elblová, P.; Lunova, M.; Dejneka, A.; Jirsa, M.; Lunov, O. Impact of mechanical cues on key cell functions and cell-nanoparticle interactions. Discov. Nano 2024, 19, 106. [Google Scholar] [CrossRef]
  237. Karlsson, H.L.; Gliga, A.R.; Calléja, F.M.G.R.; Gonçalves, C.S.A.G.; Wallinder, I.O.; Vrieling, H.; Fadeel, B.; Hendriks, G. Mechanism-based genotoxicity screening of metal oxide nanoparticles using the ToxTracker panel of reporter cell lines. Part. Fibre Toxicol. 2014, 11, 41. [Google Scholar] [CrossRef] [PubMed]
  238. Gliga, A.R.; Skoglund, S.; Wallinder, I.O.; Fadeel, B.; Karlsson, H.L. Size-dependent cytotoxicity of silver nanoparticles in human lung cells: The role of cellular uptake, agglomeration and Ag release. Part. Fibre Toxicol. 2014, 11, 11. [Google Scholar] [CrossRef] [PubMed]
  239. Wolfram, J.; Zhu, M.T.; Yang, Y.; Shen, J.L.; Gentile, E.; Paolino, D.; Fresta, M.; Nie, G.J.; Chen, C.Y.; Shen, H.F.; et al. Safety of nanoparticles in medicine. Curr. Drug Targets 2015, 16, 1671–1681. [Google Scholar] [CrossRef]
  240. Chen, Y.C.; Su, Y.C.; Roffler, S.R. Polyethylene glycol immunogenicity in nanomedicine. Nat. Rev. Bioeng. 2025, 1–19. [Google Scholar] [CrossRef]
  241. Fröhlich, E. Cellular targets and mechanisms in the cytotoxic action of non-biodegradable engineered nanoparticles. Curr. Drug Metab. 2013, 14, 976–988. [Google Scholar] [CrossRef]
  242. Kozenkova, E.; Levada, K.; Efremova, M.V.; Omelyanchik, A.; Nalench, Y.A.; Garanina, A.S.; Pshenichnikov, S.; Zhukov, D.G.; Lunov, O.; Lunova, M.; et al. Multifunctional Fe3O4-Au nanoparticles for the MRI diagnosis and potential treatment of liver cancer. Nanomaterials 2020, 10, 1646. [Google Scholar] [CrossRef]
  243. Awasthi, S.; Srivastava, A.; Kumar, D.; Pandey, S.K.; Mubarak, N.M.; Dehghani, M.H.; Ansari, K. An insight into the toxicological impacts of carbon nanotubes (CNTs) on human health: A review. Environ. Adv. 2024, 18, 100601. [Google Scholar] [CrossRef]
  244. Bhattacharya, K.; Andón, F.T.; El-Sayed, R.; Fadeel, B. Mechanisms of carbon nanotube-induced toxicity: Focus on pulmonary inflammation. Adv. Drug Deliv. Rev. 2013, 65, 2087–2097. [Google Scholar] [CrossRef]
  245. Seabra, A.B.; Durán, N. Nanotoxicology of metal oxide nanoparticles. Metals 2015, 5, 934–975. [Google Scholar] [CrossRef]
  246. Andón, F.T.; Fadeel, B. Programmed cell death: Molecular mechanisms and implications for safety assessment of nanomaterials. Acc. Chem. Res. 2013, 46, 733–742. [Google Scholar] [CrossRef] [PubMed]
  247. Witika, B.A.; Makoni, P.A.; Matafwali, S.K.; Chabalenge, B.; Mwila, C.; Kalungia, A.C.; Nkanga, C.I.; Bapolisi, A.M.; Walker, R.B. Biocompatibility of biomaterials for nanoencapsulation: Current approaches. Nanomaterials 2020, 10, 1649. [Google Scholar] [CrossRef] [PubMed]
  248. Setyawati, M.I.; Tay, C.Y.; Leong, D.T. The gap between endothelial cells: Key to the quick escape of nanomaterials? Nanomedicine 2014, 9, 1591–1594. [Google Scholar] [CrossRef]
  249. Faria, M.; Björnmalm, M.; Thurecht, K.J.; Kent, S.J.; Parton, R.G.; Kavallaris, M.; Johnston, A.P.R.; Gooding, J.J.; Corrie, S.R.; Boyd, B.J.; et al. Minimum information reporting in bio-nano experimental literature. Nat. Nanotechnol. 2018, 13, 777–785. [Google Scholar] [CrossRef]
  250. Islas, P.; Platnich, C.M.; Gidi, Y.; Karimi, R.; Ginot, L.; Saliba, D.; Luo, X.; Cosa, G.; Sleiman, H.F. Automated synthesis of DNA nanostructures. Adv. Mater. 2024, 36, 2403477. [Google Scholar] [CrossRef]
  251. Luo, X.; Saliba, D.; Yang, T.X.; Gentile, S.; Mori, K.; Islas, P.; Das, T.; Bagheri, N.; Porchetta, A.; Guarne, A.; et al. Minimalist design of wireframe DNA nanotubes: Tunable geometry, size, chirality, and dynamics. Angew. Chem.-Int. Edit. 2023, 62, e202309869. [Google Scholar] [CrossRef]
  252. Wang, J.; Xie, M.; Ouyang, L.; Li, J.; Wang, L.; Fan, C.; Chao, J. Artificial molecular communication network based on DNA nanostructures recognition. Nat. Commun. 2025, 16, 244. [Google Scholar] [CrossRef]
  253. Liu, X.; Shi, B.; Gao, Y.; Zhu, S.T.; Yan, Q.L.; Liu, X.G.; Shi, J.Y.; Li, Q.; Wang, L.H.; Li, J.; et al. Ultrabright near-infrared fluorescent DNA frameworks for near-single-cell cancer imaging. Nat. Photonics 2024, 19, 79–88. [Google Scholar] [CrossRef]
  254. Knappe, G.A.; Wamhoff, E.C.; Bathe, M. Functionalizing DNA origami to investigate and interact with biological systems. Nat. Rev. Mater. 2023, 8, 123–138. [Google Scholar] [CrossRef]
  255. Stephanopoulos, N. Hybrid nanostructures from the self-assembly of proteins and DNA. Chem 2020, 6, 364–405. [Google Scholar] [CrossRef]
  256. Elblová, P.; Andelová, H.; Lunova, M.; Anthi, J.; Henry, S.J.W.; Tu, X.Y.; Dejneka, A.; Jirsa, M.; Stephanopoulos, N.; Lunov, O. Geometrically constrained cytoskeletal reorganisation modulates DNA nanostructures uptake. J. Mater. Chem. B 2025, 13, 2335–2351. [Google Scholar] [CrossRef]
  257. Frtus, A.; Smolkova, B.; Uzhytchak, M.; Lunova, M.; Jirsa, M.; Henry, S.J.W.; Dejneka, A.; Stephanopoulos, N.; Lunov, O. The interactions between DNA nanostructures and cells: A critical overview from a cell biology perspective. Acta Biomater. 2022, 146, 10–22. [Google Scholar] [CrossRef] [PubMed]
  258. Lacroix, A.; Sleiman, H.F. DNA nanostructures: Current challenges and opportunities for cellular delivery. ACS Nano 2021, 15, 3631–3645. [Google Scholar] [CrossRef] [PubMed]
  259. Elblová, P.; Lunova, M.; Henry, S.J.W.; Tu, X.Y.; Calé, A.; Dejneka, A.; Havelková, J.; Petrenko, Y.; Jirsa, M.; Stephanopoulos, N.; et al. Peptide-coated DNA nanostructures as a platform for control of lysosomal function in cells. Chem. Eng. J. 2024, 498, 155633. [Google Scholar] [CrossRef] [PubMed]
  260. Veneziano, R.; Moyer, T.J.; Stone, M.B.; Wamhoff, E.C.; Read, B.J.; Mukherjee, S.; Shepherd, T.R.; Das, J.; Schief, W.R.; Irvine, D.J.; et al. Role of nanoscale antigen organization on B-cell activation probed using DNA origami. Nat. Nanotechnol. 2020, 15, 716–723. [Google Scholar] [CrossRef]
  261. Hellmeier, J.; Platzer, R.; Eklund, A.S.; Schlichthaerle, T.; Karner, A.; Motsch, V.; Schneider, M.C.; Kurz, E.; Bamieh, V.; Brameshuber, M.; et al. DNA origami demonstrate the unique stimulatory power of single pMHCs as T cell antigens. Proc. Natl. Acad. Sci. USA 2021, 118, e2016857118. [Google Scholar] [CrossRef]
  262. Fang, T.; Alvelid, J.; Spratt, J.; Ambrosetti, E.; Testa, I.; Teixeira, A. Spatial regulation of T-Cell signaling by programmed death-ligand 1 on wireframe DNA origami flat sheets. ACS Nano 2021, 15, 3441–3452. [Google Scholar] [CrossRef]
  263. Dong, R.; Aksel, T.; Chan, W.; Germain, R.N.; Vale, R.D.; Douglas, S.M. DNA origami patterning of synthetic T cell receptors reveals spatial control of the sensitivity and kinetics of signal activation. Proc. Natl. Acad. Sci. USA 2021, 118, e2109057118. [Google Scholar] [CrossRef]
  264. Rosier, B.J.H.M.; Markvoort, A.J.; Audenis, B.G.; Roodhuizen, J.A.L.; den Hamer, A.; Brunsveld, L.; de Greef, T.F.A. Proximity-induced caspase-9 activation on a DNA origami-based synthetic apoptosome. Nat. Catal. 2020, 3, 295–306. [Google Scholar] [CrossRef]
  265. Klein, W.P.; Thomsen, R.P.; Turner, K.B.; Walper, S.A.; Vranish, J.; Kjems, J.; Ancona, M.G.; Medintz, I.L. Enhanced catalysis from multienzyme cascades assembled on a DNA origami triangle. ACS Nano 2019, 13, 13677–13689. [Google Scholar] [CrossRef] [PubMed]
  266. Shaw, A.; Hoffecker, I.T.; Smyrlaki, I.; Rosa, J.; Grevys, A.; Bratlie, D.; Sandlie, I.; Michaelsen, T.E.; Andersen, J.T.; Högberg, B. Binding to nanopatterned antigens is dominated by the spatial tolerance of antibodies. Nat. Nanotechnol. 2019, 14, 184–190. [Google Scholar] [CrossRef]
  267. Li, F.; Liu, Y.J.; Dong, Y.H.; Chu, Y.W.; Song, N.C.; Yang, D.Y. Dynamic assembly of DNA nanostructures in living cells for mitochondrial interference. J. Am. Chem. Soc. 2022, 144, 4667–4677. [Google Scholar] [CrossRef] [PubMed]
  268. Dong, Y.H.; Li, F.; Lv, Z.Y.; Li, S.; Yuan, M.H.; Song, N.C.; Liu, J.Q.; Yang, D.Y. Lysosome interference enabled by proton-driven dynamic assembly of DNA nanoframeworks inside cells. Angew. Chem.-Int. Edit. 2022, 61, e202207770. [Google Scholar] [CrossRef] [PubMed]
  269. Yang, S.; Cheng, Y.; Liu, M.X.; Tang, J.P.; Li, S.Q.; Huang, Y.; Kou, X.H.; Yao, C.; Yang, D.Y. Sequential assembly of DNA nanoparticles inside cells enables lysosome interference and cell behavior regulation. Nano Today 2024, 56, 102224. [Google Scholar] [CrossRef]
  270. Yuan, M.H.; Dong, Y.H.; Lv, Z.Y.; Liu, J.Q.; Liu, M.X.; Xu, M.D.; Guo, X.C.; Yao, C.; Yang, D.Y. Controlled sequential assembly of DNA nanoparticles inside cells enabling mitochondrial interference. Adv. Funct. Mater. 2024, 34, 2312880. [Google Scholar] [CrossRef]
  271. Elblová, P.; Anthi, J.; Liu, M.H.; Lunova, M.; Jirsa, M.; Stephanopoulos, N.; Lunov, O. DNA nanostructures for rational regulation of cellular organelles. JACS Au 2025, 5, 1591–1616. [Google Scholar] [CrossRef]
Figure 1. Schematic illustration of major nanoparticle types used in clinically approved nanodrugs. Each nanoparticle type includes several subtypes, a selection of which is highlighted here. These nanoparticle systems exhibit unique advantages and limitations related to cargo loading capacity, delivery efficiency, and patient-specific response profiles. Created with BioRender version 2.0.
Figure 1. Schematic illustration of major nanoparticle types used in clinically approved nanodrugs. Each nanoparticle type includes several subtypes, a selection of which is highlighted here. These nanoparticle systems exhibit unique advantages and limitations related to cargo loading capacity, delivery efficiency, and patient-specific response profiles. Created with BioRender version 2.0.
Ijms 26 06687 g001
Figure 2. Summary of factors affecting nanoparticle cytotoxicity and biocompatibility. Created with BioRender.
Figure 2. Summary of factors affecting nanoparticle cytotoxicity and biocompatibility. Created with BioRender.
Ijms 26 06687 g002
Figure 3. Schematic illustration of ROS role in cytotoxicity triggered by nanoparticles. Several factors contribute to the generation of ROS induced by nanoparticles, including their active surface area, size, photoactivation properties, associated toxins, metal ion dissolution, and interactions with biomolecules. ROS: reactive oxygen species; LMP: lysosomal membrane permeabilization; UPR: unfolded protein response. Created with BioRender.
Figure 3. Schematic illustration of ROS role in cytotoxicity triggered by nanoparticles. Several factors contribute to the generation of ROS induced by nanoparticles, including their active surface area, size, photoactivation properties, associated toxins, metal ion dissolution, and interactions with biomolecules. ROS: reactive oxygen species; LMP: lysosomal membrane permeabilization; UPR: unfolded protein response. Created with BioRender.
Ijms 26 06687 g003
Figure 4. Role of inflammatory responses in cytotoxicity triggered by nanoparticles. ROS: reactive oxygen species; LMP: lysosomal membrane permeabilization; IL-6: Interleukin 6; IL-8: Interleukin 8; TNF-α: Tumor necrosis factor α; IL-1β: interleukin-1β. Created with BioRender.
Figure 4. Role of inflammatory responses in cytotoxicity triggered by nanoparticles. ROS: reactive oxygen species; LMP: lysosomal membrane permeabilization; IL-6: Interleukin 6; IL-8: Interleukin 8; TNF-α: Tumor necrosis factor α; IL-1β: interleukin-1β. Created with BioRender.
Ijms 26 06687 g004
Figure 5. Role of lysosomal dysfunction in cytotoxicity triggered by nanoparticles. Nanoparticles may disrupt autophagy by overloading or damaging the lysosomal compartment, or by altering the cytoskeleton, thereby impairing autophagosome–lysosome fusion. Additionally, nanoparticles can compromise lysosomal stability by inducing oxidative stress, alkalization, osmotic swelling, or by exerting detergent-like effects on the lysosomal membrane, ultimately leading to LMP. ROS: reactive oxygen species; LMP: lysosomal membrane permeabilization; UPR: unfolded protein response. Created with BioRender.
Figure 5. Role of lysosomal dysfunction in cytotoxicity triggered by nanoparticles. Nanoparticles may disrupt autophagy by overloading or damaging the lysosomal compartment, or by altering the cytoskeleton, thereby impairing autophagosome–lysosome fusion. Additionally, nanoparticles can compromise lysosomal stability by inducing oxidative stress, alkalization, osmotic swelling, or by exerting detergent-like effects on the lysosomal membrane, ultimately leading to LMP. ROS: reactive oxygen species; LMP: lysosomal membrane permeabilization; UPR: unfolded protein response. Created with BioRender.
Ijms 26 06687 g005
Figure 6. Generalized key cytotoxicity mechanisms associated with different classes of nanodrugs. ROS: reactive oxygen species; LMP: lysosomal membrane permeabilization; UPR: unfolded protein response. Created with BioRender.
Figure 6. Generalized key cytotoxicity mechanisms associated with different classes of nanodrugs. ROS: reactive oxygen species; LMP: lysosomal membrane permeabilization; UPR: unfolded protein response. Created with BioRender.
Ijms 26 06687 g006
Figure 7. Proposed Toxicity Index Scoring System. QDs: quantum dots; NPs: nanoparticles; CNTs: carbon nanotubes; pt/pts: point/points. Created with BioRender.
Figure 7. Proposed Toxicity Index Scoring System. QDs: quantum dots; NPs: nanoparticles; CNTs: carbon nanotubes; pt/pts: point/points. Created with BioRender.
Ijms 26 06687 g007
Table 1. Clinically approved nanodrugs and their general characteristics.
Table 1. Clinically approved nanodrugs and their general characteristics.
Trade NameAPINanocarrierYear of ApprovalApplicationsCompanyRef.
Lipid-Based NPs
DiprivanPropofolNanoemulsionFDA 1989Sedation or anesthesiaFresenius Kabi[9,48]
DoxilDoxorubicinPEGylated
liposome
FDA 1995Metastatic ovarian cancer
HIV-associated Kaposi’s sarcoma
Multiple myeloma
Janssen Pharmaceuticals[9]
AbelcetAmphotericin BLipid complexFDA 1995Invasive severe fungal infectionLeadiant Biosciences Inc.[49]
CaelyxDoxorubicinPEGylated liposomeEMA 1996Metastatic breast cancer
Ovarian cancer
AIDS-associated Kaposi’s sarcoma
Multiple myeloma
Eagle Pharmaceuticals[50]
AmBisomeAmphotericin BUnilamellar liposomeFDA 1997Fungal and/or protozoal infection
Anti-leishmanial
Gilead Sciences, Inc.[9]
MyocetDoxorubicinLiposomeEMA 2000Metastatic breast cancerCHEPLAPHARM Arzneimittel GmbH[9,10]
VisudyneVerteporfinUnilamellar liposomeFDA 2000
EMA 2000
Decreased vision
Macular degeneration
Pathologic myopia
Bausch + Lomb[51]
DefinityPerflutrenPhospholipid-stabilized microbubbleFDA 2001Ultrasound contrast agentLantheus Medical Imaging[52]
MepactMifamurtideLiposomeEMA 2009Non-metastatic osteosarcoma and myosarcomaTakeda Pharmaceuticals[53]
ExparelBupivacaineLiposomeFDA 2011
EMA 2020
Pain managementPacira Biosciences[54]
OnivydeIrinotecanLiposomeFDA 2015
EMA 2016
Metastatic breast cancerIpsen[55]
VyxeosDaunorubicin and cytarabine (1:5 ratio)LiposomeFDA 2017
EMA 2018
Acute myeloid leukemia
AML-MRC
t-AML
Jazz Pharmaceuticals[56]
OnpattroPatisiran sodiumLipid nanoparticleFDA 2018
EMA 2018
Hereditary transthyretin-mediated amyloidosisAlnylam Pharmaceuticals, Inc.[9,10]
ShingrixRecombinant VZV glycoprotein ELiposomeEMA 2018Prevention of shingles and post-herpetic neuralgiaGlaxoSmithKline[57]
ArikayceAmikacinLiposomeFDA 2018
EMA 2020
NTM lung disease caused by MACInsmed Incorporated[58]
Comirnaty (BNT162b2)mRNA encoding SARS-CoV-2 spikePEGylated lipid nanoparticleFDA 2021
EMA 2022
Prevention of coronavirus 2 infection (SARS-CoV-2 vaccine)Pfizer, Inc.[9]
Spikevax (mRNA-1273)mRNA encoding SARS-CoV-2 spikePEGylated lipid nanoparticleFDA 2022
EMA 2022
Prevention of coronavirus 2 infection (SARS-CoV-2 vaccine)Moderna, Inc.[9]
Polymer-based NPs
OptisonPerflutrenAlbumin-stabilized microbubbleFDA 1997
EMA 1998
Ultrasound contrast agentGE Healthcare[59]
AbraxanePaclitaxelAlbumin-bound nanoparticleFDA 2005, 2012, 2013
EMA 2008
Metastatic breast cancer
Lung cancer
Metastatic pancreatic adenocarcinoma
Eli Lilly Company[60]
ZilrettaTriamcinolone acetonidePLGA microsphereFDA 2017Knee osteoarthritisPacira Biosciences[61]
ApealeaPaclitaxelMicelleFDA 2018Ovarian cancer
Peritoneal cancer
Fallopian tube cancer
Oasmia Pharmaceutical[62]
Inorganic NPs
VenoferIron sucroseColloidal iron sucroseFDA 2000Iron deficiency in CKDAmerican Regent[63]
FerahemeFerumoxytolDextran-based nanoparticleFDA 2009Iron deficiency in CKDCovis Group S.a.r.l[26]
InjectaferFerric carbocymaltoseColloidal formulationFDA 2013Iron deficiency in anemiaVifor Pharma[26]
HensifyHafnium oxideInorganic nanoparticleEMA 2019Squamous cell carcinomaNanobiotix[64]
API: Active Pharmaceutical Ingredient; FDA: U.S. Food and Drug Administration; EMA: European Medicines Agency; VZV: Varicella-Zoster Virus; SARS-CoV-2: Severe acute respiratory syndrome coronavirus 2; AML-MRC: Acute myeloid leukemia with myelodysplasia-related changes; t-AML: Therapy-related acute myeloid leukemia; MAC: Mycobacterium avium complex; NTM: Nontuberculous mycobacteria; CKD: Chronic kidney disease; PEG: Polyethylene glycol; PLGA: poly(lactic-co-glycolic) acid.
Table 2. Key molecular pathways mediating nanoparticle-induced cytotoxicity.
Table 2. Key molecular pathways mediating nanoparticle-induced cytotoxicity.
Molecular PathwayAffected Cell ComponentsKey MediatorsClinical ImplicationsRef.
Oxidative stressMitochondria, DNA, proteins, lipidsROS, antioxidant enzymesPotential for widespread cellular damage, mutagenesis[179]
Inflammatory responseCell membrane, cytokine signaling pathwaysNF-κB, TNF-α, IL-6, IL-8Chronic inflammation, tissue damage[179]
Lysosomal dysfunctionLysosomes, autophagy machineryCathepsins, autophagy-related proteinsDisruption of cellular waste management, potential trigger for cell death[10,180,181]
Membrane disruptionPlasma membrane, organelle membranesMembrane lipids, membrane proteinsAltered cellular permeability, potential for cell lysis[182]
Mitochondrial dysfunctionMitochondriaElectron transport chain components, ATP synthaseEnergy metabolism disruption, potential trigger for apoptosis[181]
DNA damageNucleus, mitochondrial DNADNA repair enzymes, p53Mutagenesis, potential carcinogenesis[179]
ApoptosisWhole cellCaspases, Bcl-2 family proteinsProgrammed cell death, potential for tissue damage[181,182]
AutophagyAutophagosomes, lysosomesLC3, p62, Beclin-1Altered cellular homeostasis, potential protective or destructive effects[10,181]
FerroptosisCell membrane, mitochondriaIron, lipid peroxidesIron-dependent cell death, potential for tissue-specific effects[183]
ER StressERUnfolded protein response proteinsProtein folding disruption, potential trigger for apoptosis[181]
Cytoskeleton disruptionActin filaments, microtubulesActin, tubulinAltered cell morphology and motility[184]
Cell cycle regulationNucleus, cytoplasmCyclins, CDKsAltered cell proliferation, potential for carcinogenesis[184,185]
Immune modulationImmune cells, cytokine signaling pathwaysToll-like receptors, complement proteinsAltered immune responses, potential for immunotoxicity[186]
Epigenetic changesNucleus, chromatinHistone modifying enzymes, DNALong-term alterations in gene expression[187]
ROS: Reactive Oxygen Species; ER: Endoplasmic Reticulum; CDKs: cyclin-dependent kinases; NF-κB: nuclear factor kappa-light-chain-enhancer of activated B cells; IL-6: interleukin 6; IL-8: interleukin 8; TNF-α: tumor necrosis factor α.
Table 3. Molecular basis of nanoparticle cytotoxicity by particle type.
Table 3. Molecular basis of nanoparticle cytotoxicity by particle type.
Nanoparticle TypeMolecular PathwaysCell ModelRef.
Gold nanoparticlesOxidative stress
Membrane disruption
DNA damage
ER stress
Cell cycle regulation
Embryonic lung fibroblasts *
Human neuroblastoma cells *
Human monocytes *
Human neutrophils *
Human endothelial cells *
[181,182,183,213]
Silver nanoparticlesOxidative stress
Lysosomal dysfunction
Mitochondrial dysfunction
DNA damage
Apoptosis
Autophagy
ER stress
Human leukemia cells *
Human HCC *
D. melanogaster **
AML cell lines *
Embryonic lung fibroblasts *
Human hepatoblastoma cells *
AML patient samples **
Human bronchial epithelial cells *
Mouse (lung, liver, kidney) **
Rat **
Human neuroblastoma cells *
[181,213,214,215,216,217]
Quantum dotsOxidative stress
Mitochondrial dysfunction
Apoptosis
ER stress
Epigenetic changes
Human breast carcinoma *
Endometrial cancer cells *
Rat neuronal cells *
Mouse **
Rat neuronal cells *
[181,182,187,218]
Carbon nanotubesOxidative stress
Inflammatory responses
Lysosomal dysfunction
Mitochondrial dysfunction
DNA damage
Apoptosis
Rat embryonic lung fibroblasts *
Normal and malignant mesothelial cells *
Mouse and rat lungs **
Mouse hepatoblastoma cells *
Lung epithelial cells *
Human embryonic kidney cells *
Bronchial epithelial cells *
[181,219,220,221,222,223,224]
Iron oxide nanoparticlesOxidative stress
Mitochondrial dysfunction
DNA damage
Apoptosis
Cytoskeleton dysregulation
Cell cycle regulation
Human breast carcinoma *
Human cervical carcinoma *
Human lung adenocarcinoma *
Human embryonic kidney cells *
Human neuroblastoma cells *
Mouse hepatocytes *
Breast cells (cancerous and non-cancerous) *
Healthy lung cells *
Liver cancer cells *
Mouse **
Umbilical vein endothelial cells *
[99,183,184,225,226]
Silica nanoparticlesOxidative stress
Inflammatory responses
Mitochondrial dysfunction
Apoptosis
Autophagy
Mouse macrophage-like cells *
HUVEC *
Mice (peritoneal macrophages) **, Mouse macrophage-like cells *
Human endothelial cells *
Human hepatic cell line *
Human HCC *
[181,227,228,229]
Titanium dioxide nanoparticlesOxidative stress
Inflammatory responses
Mitochondrial dysfunction
Apoptosis
Autophagy
ER stress
Mouse erythrocytes, brain, liver **
Bronchial epithelial cells *
Rat liver and kidney **
Rat **
Human monocytes *
Human neuroblastoma cells *
Mouse **
[181,183,230,231,232]
Zinc oxide nanoparticlesOxidative stress
Inflammatory responses
Apoptosis
Autophagy
Ferroptosis
Mouse erythrocytes, brain, liver **
Rat lung **,
Human lung adenocarcinoma *
Human neuroblastoma cells *
Mouse macrophages *
Bronchial epithelial cells *
Zebrafish embryos **
Embryonic lung fibroblasts *
Mouse **
[181,182,183,230,233,234,235]
Aluminum oxide nanoparticlesOxidative stress
Autophagy
Ferroptosis
Mouse erythrocytes, brain, liver **
Mouse **
Rat **
[181,183,230]
* in vitro model; ** in vivo model; HCC: Hepatocellular carcinoma; AML: Acute myeloid leukemia; HUVEC: Human umbilical vein endothelial cells.
Table 4. Qualitative comparative matrix showing the relationship between nanodrug types and their predominant cytotoxic mechanisms.
Table 4. Qualitative comparative matrix showing the relationship between nanodrug types and their predominant cytotoxic mechanisms.
Nanodrug
Category
Cytotoxic TriggerRef.
Oxidative StressLysosomal
Dysfunction
Membrane
Disruption
Inflammatory
Response
Mitochondrial
Dysfunction
Liposomes+++++++++[186]
Metal NPs (Au, Ag)++++++++++++[182]
Quantum dots+++++++++++[187]
Carbon nanotubes++++++++++[116]
Polymeric NPs++++++++[180]
Iron oxide NPs++++++++++[184]
Silica NPs+++++++++[138]
+++ Strong association (frequently reported/primary mechanism); ++ Moderate association (commonly reported); + Weak association (occasionally reported).
Table 5. Risk mitigation strategies to counteract nanoparticle-induced cytotoxicity.
Table 5. Risk mitigation strategies to counteract nanoparticle-induced cytotoxicity.
Nanodrug CategorySafety ConcernsMolecular BasisMitigation ApproachesRef.
Metal nanoparticles (e.g., gold, silver)Size-dependent toxicity, ROS generationOxidative stress, membrane disruptionSize optimization, surface coating (e.g., PEGylation)[182,245]
Quantum dotsHeavy metal toxicity, long-term accumulationCadmium-induced cellular damage, ROS generationUse of cadmium-free QDs, surface passivation[187]
Carbon nanotubesAsbestos-like effects, inflammatory responseMembrane damage, ROS generation, NF-κB activationFunctionalization, length control, use of biodegradable CNTs[179,246]
Polymeric nanoparticlesPotential immunogenicity, complement activationProtein corona formation, inflammatory responseStealth coatings, immunomodulatory strategies[180,186]
LiposomesComplement activation, potential cardiotoxicityLipid peroxidation, membrane fusionPEGylation, use of non-toxic lipids, size optimization[186]
Iron oxide nanoparticlesROS generation, potential for iron overloadFenton reaction, disruption of iron homeostasisSurface coating, controlled biodegradation[184,186,241]
Silica nanoparticlesHemolysis, liver toxicityMembrane interactions, ROS generationSurface modification, size control[247]
DendrimersCationic toxicity, hemolysisMembrane disruption, mitochondrial dysfunctionSurface modification, use of biodegradable cores[4,6,142]
Titanium dioxide nanoparticlesPulmonary inflammation, potential carcinogenicityROS generation, DNA damageSurface coating, shape control[241,248]
Zinc oxide nanoparticlesDissolution-related toxicity, ROS generationZn2+ release, oxidative stressSurface passivation, controlled dissolution[6,181]
ROS: Reactive Oxygen Species; NF-κB: Nuclear factor kappa-light-chain-enhancer of activated B cells; PEG: Polyethylene glycol; QDs: Quantum dots; CNTs: Carbon nanotubes.
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

Calé, A.; Elblová, P.; Andělová, H.; Lunova, M.; Lunov, O. Analyzing Molecular Determinants of Nanodrugs’ Cytotoxic Effects. Int. J. Mol. Sci. 2025, 26, 6687. https://doi.org/10.3390/ijms26146687

AMA Style

Calé A, Elblová P, Andělová H, Lunova M, Lunov O. Analyzing Molecular Determinants of Nanodrugs’ Cytotoxic Effects. International Journal of Molecular Sciences. 2025; 26(14):6687. https://doi.org/10.3390/ijms26146687

Chicago/Turabian Style

Calé, Alicia, Petra Elblová, Hana Andělová, Mariia Lunova, and Oleg Lunov. 2025. "Analyzing Molecular Determinants of Nanodrugs’ Cytotoxic Effects" International Journal of Molecular Sciences 26, no. 14: 6687. https://doi.org/10.3390/ijms26146687

APA Style

Calé, A., Elblová, P., Andělová, H., Lunova, M., & Lunov, O. (2025). Analyzing Molecular Determinants of Nanodrugs’ Cytotoxic Effects. International Journal of Molecular Sciences, 26(14), 6687. https://doi.org/10.3390/ijms26146687

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