Open access peer-reviewed chapter

Potential Toxicity of Nanoparticles for the Oral Delivery of Therapeutics

Written By

Iman M. Alfagih

Submitted: 19 May 2023 Reviewed: 24 May 2023 Published: 20 June 2023

DOI: 10.5772/intechopen.111946

From the Edited Volume

Toxicity of Nanoparticles - Recent Advances and New Perspectives

Edited by Mohammed Muzibur Rahman, Jamal Uddin, Abdullah Mohamed Asiri and Md Rezaur Rahman

Chapter metrics overview

50 Chapter Downloads

View Full Metrics

Abstract

Nanoparticles (NPs) offer a promising solution for orally delivering therapeutic substances due to their capability to surpass traditional drug delivery system (DDS) limitations like low solubility, bioavailability, and stability. However, the possible toxic effects of using NPs for oral therapeutic delivery raise significant concerns, as they might interact with biological systems unexpectedly. This chapter aims to comprehensively understand the potential toxicity of NPs employed in oral therapeutic delivery. Factors such as size, surface area, surface charge, and surface chemistry of NPs can impact their toxicity levels. Both in vitro and in vivo models have been utilised to evaluate NPs toxicity, with in vivo models being more suitable for anticipating human toxicity. The possible toxic consequences of different NPs varieties, including polymer, lipid, and metal NPs, have been documented. Ultimately, grasping the potential toxicity of NPs in oral therapeutic delivery is essential for creating safe and effective DDS.

Keywords

  • nanoparticles
  • oral drug delivery
  • cytotoxicity
  • inflammation
  • intestinal microbiota

1. Introduction

The use of nanoparticles (NPs) for oral drug delivery is a ground-breaking and rapidly growing area of research [1]. NPs are solid colloidal drug delivery systems (DDS) with sizes ranging from 1 nm to 1000 nm. They comprise polymers, lipids, carbon, silica, or metal encapsulating the drug moiety [2, 3, 4, 5]. Encapsulating drugs within NPs can increase stability, solubility, and bioavailability, ultimately boosting their therapeutic effectiveness [6]. Recent advances in biomedical research have led to the successful enhancement of therapeutic agents for treating various diseases. However, a significant challenge remains in efficiently delivering these agents to the target site [7]. NPs can be engineered with specific characteristics to optimise the DDS [4]. For example, biocompatible polymer NPs are coated to enhance their presence in the bloodstream or be equipped with unique ligands or antibodies that focus on specific target cells or tissues [2]. By directing drug delivery to specific locations within the body, NPs can reduce negative side effects and increase the effectiveness of treatments [8].

Oral drug administration is the most prevalent method and the favoured option for patients, as it is non-invasive, user-friendly, and well-accepted [9]. However, traditional forms like tablets and capsules may have a rapid and inadequately regulated drug release, potentially leading to drug degradation and alteration due to the gastrointestinal tract (GIT) environment (like, changes in pH, digestive enzymes, and microbiota) [10]. NPs can address these issues by safeguarding drugs from degradation and promoting their absorption through the intestinal barrier. Moreover, oral drugs can be directed to specific areas within the GIT to provide localised treatment for stomach and colorectal cancers, infections, inflammation, bowel disorders, gastro-duodenal ulcers, and gastroesophageal reflux issues [11]. GIT fluid consists of enzymes, acids, and other compounds that quickly decompose drugs, resulting in inadequate absorption and decreased effectiveness. Furthermore, a mucus layer in the GI tract can hinder drug absorption by acting as a barrier, restricting their access to the intestinal epithelium [12, 13]. Thus, using NPs as an oral DDS holds great promise due to their unique properties (which include good therapeutic properties, self-assembly, enhanced biocompatibility, serving as vehicles for antimicrobial agents, controlled drug release, reducing off-target effects, etc.) [2, 4, 14].

Although NPs-based DDS offer promising benefits, their safety must be thoroughly evaluated. The small size of NPs can result in interactions with biological systems that are not yet fully understood, possibly causing toxic consequences. Furthermore, NPs can accumulate in specific tissues or organs, leading to long-lasting damage. A recent study by Cabellos et al. [15] showed that mice exposed to silica NPs suffered inflammation and epithelial damage in the intestines and showed the expression of autophagy proteins [4, 16]. Various mechanisms can contribute to the potential toxicity of NPs. For example, they can cause oxidative stress by generating reactive oxygen species (ROS) that harm cell components [17]. NPs can also trigger inflammatory reactions by activating immune cells, causing tissue injury and malfunction [18]. Moreover, certain NPs might exhibit genotoxic effects by damaging DNA, potentially resulting in mutations and cancer [19]. However, it is crucial to meticulously assess the possible toxicity of NPs to ensure their safety and effectiveness [20, 21]. By refining the development and analysis of NPs, one can reduce toxicity and increase therapeutic advantages by modifying functional groups, coating with cell membranes, or using nanorobots [22]. For example, DDS can be developed using stable intestinal cell-derived exosomes that target colonic cells. It is even known that these exosomes can be found in faces. Lipid NPs derived from plants can also be used as an alternative strategy, thus providing a safe NPs delivery [23, 24]. Thereby, conducting comprehensive pre-clinical and clinical research is vital to evaluate the safety of NPs in the context of orally administered treatments. Such investigations should examine the biodistribution, pharmacokinetics, and toxicity of NPs in both animal models and human subjects [21, 25]. Specifically, close observation of NPs accumulation in different organs and tissues is necessary, along with monitoring any negative impacts on cellular and molecular functions [26]. Therefore, the present chapter delves into the diverse toxicological issues of orally administered NPs (from polymer, lipid, protein, carbon, silica, and metal-based NPs) and summarises the in vitro and in vivo assessments of the toxicity of oral NPs.

Advertisement

2. Structure of GIT and the interaction of nanoparticles

The GIT is a complex system responsible for nutrient absorption, immune function, and waste elimination, divided into several regions, including the mouth, oesophagus, stomach, small intestine, and large intestine. Each region of GIT presents unique anatomical characteristics and physiological conditions that can influence the interaction of NPs. The GIT represents a selective mucosal barrier, with an estimated surface area of 200 m2 in adult humans, that can potentially interact with ingested NPs [27]. The anatomical and physiological characteristics of each part of the GIT can affect NPs absorption and elimination [28]. For example, the stomach presents a tough barrier to drug absorption, with a strong acid environment (pH range of 1.0–2.5) that can degrade food, acid-labile drugs, and pathogenic microorganisms. Furthermore, the stomach has extrinsic epithelial cells and a mucin-bicarbonate barrier, which, combined with tight junctions beneath the intrinsic barrier, limits drug absorption. Furthermore, stomach pepsins can inactivate protein drugs [13]. The small intestine has a huge surface area due to the villi and microvilli in the intestinal lumen [29]. The small intestine is considered a major site for oral drug delivery due to its enormous surface and various transport routes [13]. The intestinal mucosa can recognise and transfer ectogenic antigens to the immune system. However, some challenges to drug delivery in the small intestine still arise from its unique physiology [30]. DDS that can increase their retention time in villi and microvilli, improve lipid solubility, and interact with a specific receptor or carrier can increase their overall bioavailability [13, 31]. Therefore, the unique anatomical and physiological features of each part of the GIT play a crucial role in the interaction and potential toxicity of NPs in the GIT.

The interaction of NPs with GIT depends on their physicochemical properties, including size, shape, surface charge, and surface chemistry (Table 1) [58]. Following oral administration, NPs encounter the mucous layer covering the GIT, which protects against harmful agents, such as pathogens and toxins [59]. However, NPs can interact with the mucous layer and penetrate the underlying tissue, resulting in potential toxicity [60].

Nanoparticles compositionToxic effectsIn vitro modelsIn vivo modelsReference
Polymers such as PLGA, PEG, chitosan, and othersCytotoxicity, genotoxicity, oxidative stress, inflammationCell lines such as HEK293, A549, and othersMice, rats, and Zebrafish[32, 33, 34, 35]
Phospholipids, cholesterol, and other lipidsCytotoxicity, genotoxicity, oxidative stress, inflammationCell lines such as HEK293, A549, and othersMice and rats[36, 37, 38, 39]
Silicon dioxide or silicon-based compoundsCytotoxicity, genotoxicity, oxidative stress, inflammationCell lines such as HepG2, BEAS-2B, and othersMice and rats; Caenorhabditis elegans[40, 41, 42, 43, 44, 45]
GoldCytotoxicity, genotoxicity, oxidative stress, inflammationCell lines such as MCF-7, HEK293, and othersMice and rats[46, 47, 48, 49, 50, 51]
SilverCytotoxicity, genotoxicity, oxidative stress, inflammationCell lines such as A549, RAW264.7, and othersMice and rats[52, 53, 54, 55]
Iron oxideCytotoxicity, genotoxicity, oxidative stress, inflammationCell lines such as RAW264.7, MCF-7, and othersMice and rats[56, 57]

Table 1.

Some studies on the toxic effects of various nanoparticles on different models.

2.1 Influence of NPs size on GIT toxicity

Smaller NPs possess a higher surface area-to-volume ratio, promoting better engagement with the mucous layer and increased absorption into the tissue [61]. Conversely, larger NPs may interact less with the mucous layer and could be eliminated from the GIT before reaching the tissue. Zhao et al. [62] noted that a 100 nm nano-vaccine showed superior pharmacokinetic effectiveness compared to a 500 nm nano-vaccine when an alum adjuvant was administered. The toxicities of NPs are inversely proportional to their size, and they are generally more toxic than large particles of the same chemical substance [63].

2.2 Influence of NPs shape and aspect ratio on GIT toxicity

It was reported that the shape of NPs can impact their association with the GIT as well. NPs with round shapes have been discovered to exhibit a higher level of interaction with the mucous layer compared to alternative forms, such as cylindrical or cubic shapes [61]. This occurs because round NPs possess a larger surface area-to-volume ratio, which can increase their connection with the mucous layer [4]. In addition to size, the configuration and proportion of NPs are essential factors in determining their cytotoxic effects in vivo [64]. Vedhanayagam et al. [65] investigated the impact of various zinc oxide NPs, such as spheres, needle, rod, hexagonal, star, flower, doughnut, circular discs, and cube, on the healing process of wounds. The researchers found that the spherical structure of zinc dioxide (ZnO) within a cross-linked collagen framework leads to improved re-epithelization and more rapid collagen accumulation. A higher aspect ratio of NPs is believed to be linked to increased cytotoxicity due to decreased clearance and enhanced bioavailability of these particles [66]. NPs with higher aspect ratios often exhibit cytotoxicity patterns that resemble those of asbestos. These particles can cause macrophage cell death during phagocytosis and, similar to asbestos fibres, can contribute to cancer formation [67].

2.3 Influence of NPs charge on GIT toxicity

NPs with positive charges have been shown to have a stronger connection with the negatively charged mucous layer compared to those with a negative charge or no charge at all. Researchers have explored using charged polymers to temporarily open tight junctions and enhance drug delivery across the intestinal epithelial barrier. For example, chitosan (a cationic polymer) has been shown to promote the paracellular transport of NPs [68]. NPs’ bioavailability and absorption have been studied only a few times after oral administration with regard to surface charge, hydrophobicity, and shape. NPs with a positive charge demonstrated increased absorption and movement by enterocytes compared to those with a negative or neutral charge, as well as a notable increase in oral bioavailability in vivo [69]. Furthermore, rod-shaped gold NPs (AuNPs) and DNA struggle to permeate or enter cells because of their charge. To enhance uptake, both AuNPs and DNA have undergone surface modification by adding lipid layers, while DNA has also been electrostatically attached to cationic liposomes, facilitating transport into cells [21].

2.4 Influence of NPs surface chemistry on GIT toxicity

Numerous NPs undergo modifications to alter their surface chemistry for specific objectives [70]. For instance, applying a polyethylene glycol (PEG) coating on NPs surfaces to establish water-attracting surface chemistry lessened the intense interaction with mucus components and increased particle movement across the mucus and mucosa [69]. Furthermore, Hasse and colleagues reported that peptide-coated silver NPs (20 nm) were more cytotoxic than citrate-coated silver NPs of the same size. They evaluated cytotoxicity in a human leukaemia cell line using the WST-1 assay [71]. It was perceived that the toxic effects of AgHECp (Silver hexagonal close placed), Ag pristine, and AgPVP (silver polyvinylpyrrolidone) were examined in A431 cell lines (human epidermoid carcinoma) and HaCaT (human keratinocytes) cell lines [71]. It has been revealed that smaller particles with negative surface charge and increased ionic content, such as AgPVP, were highly toxic. Similarly, toxic effects were observed for the larger particles, which were pure AgNP with a negative surface charge and ion content comparable to AgPVP [71]. Additionally, NPs featuring a hydrophobic surface exhibit a more robust interaction with the mucous layer than their hydrophilic counterparts. A cationic polysaccharide with mucoadhesive properties, chitosan has been extensively investigated. Chitosan’s mucoadhesive potency is limited due to its low water solubility and low mucoadhesive strength, so it must be chemically modified to optimise its mucoadhesive properties [72].

Advertisement

3. Toxicological concern with oral nanoparticles

3.1 Polymer-based nanoparticles

Polymer-based NPs (PNPs) can interact with GIT cells and tissues, resulting in inflammation, oxidative stress, and damage to the intestinal barrier. Various poly(lactide-co-glycolic acid) NPs (PLGA) were examined for toxicity in THP-1 macrophages similar to human cells. When used as an NPs stabiliser, chitosan polymer conferred significant cytotoxicity to PLGA NPs, despite being slightly cytotoxic [73]. Research has shown that NPs exhibit varying levels of impact on intestinal cells, intestinal flora, and the intestinal barrier, confirming that NPs cause damage to the digestive system [45, 74]. Polystyrene NPs administration has been found to trigger the TOS-MAPK/NF-kB signalling pathway in macrophage RAW 264.7, leading to inflammation with pro-inflammatory and cytotoxic potential activity [35]. Furthermore, exposure to polystyrene NPs have been shown to disrupt the balance of cell populations within the intestinal cells of zebrafish [75]. Existing data on the toxic effects of ingested zinc oxide (ZnO) NPs on intestinal models need to be more consistent. Several studies have found negative biological effects, such as increased intestinal inflammation, reduced cell viability, and mitochondrial membrane depolarization, resulting from the treatment with ZnO NPs. However, modified ZnO NPs do not cause significant cell damage [76, 77].

3.2 Lipid-based nanoparticles

Lipid-based nanoparticles (LNPs) have gained attention as potential DDS due to their ability to encapsulate hydrophobic medications and shield them from degradation [78]. Cationic lipids, for instance, show significant potential as carriers for delivering delicate substances such as nucleic acids, but certain cationic lipids can lead to cytotoxicity [36]. The impact of hydrophobic chains on lipid toxicity has yet to be thoroughly investigated, impeding the development of less harmful lipids [79]. Solid lipid nanoparticles (SLNs) are among the most prevalent types of LNPs employed in drug delivery. SLNs have been discovered to cause toxicity in the GIT, resulting in inflammation, oxidative stress, and damage to the intestinal barrier [80]. Numerous studies have indicated that the interaction between LNPs and GIT can trigger an immune response, modify the gut microbiota, and induce toxicity [79, 81]. A recent study discovered that oral administration of altered RNA-LNPs led to the expression of pro-inflammatory cytokines (such as interleukin (IL) -6 and macrophage inflammation protein 2 (MIP-2)) and chemokines in the intestines of unexposed mice [39]. Ball et al. [82] documented that LNPs can compromise the stability of the intestinal epithelial barrier and result in intestinal inflammation.

3.3 Silica-based nanoparticles

Silicon-based nanoparticles (SiNPs) can interact with GIT cells and tissues, causing inflammation, oxidative stress, and damage to the intestinal barrier [83]. Various factors influence the toxicity of SiNPs in GIT, including their dimensions, surface area, surface charge, and chemical makeup [84]. Cellular absorption of silica dioxide NPs (SiO2NPs) depends on the size of the particles, particularly in the range of 30-50 nm. SiO2 NPs within the 30-50 nm size range can be carriers in multiple applications [40, 41]. An in vivo study revealed that ingesting SiO2NPs led to GIT inflammation and increased permeability, causing intestinal contents to leak into the bloodstream and disrupting the microbiota-gut-brain axis [85]. Guo et al. [83] discovered that exposure to SiO2NPs affected nutrient transportation, ROS production, barrier function, gene expression, and microvilli structure. To assess the potential toxicity of SiNPs, examining their interactions with gut cells, uptake, and impact on GIT function and microbiota is important. The nanoscale SiO2 found in the E551 food additive could uniquely impact the absorption and distribution of SiO2 within the human body [86]. Numerous in vitro studies have shown that SiO2NPs can produce cytotoxic effects in cultured human cell lines, such as glioblastoma cells, depending on their size, shape, and dose [87]. Furthermore, SiO2NPs, inhaled or ingested, can infiltrate cells and engage with cellular membranes or organelles, leading to mammalian cell death through oxidative stress, endoplasmic reticulum stress, and apoptosis [42, 43]. A different instance of SiNPs that exhibit toxicity in the GIT involves mesoporous silica NPs (M-SiNPs) [44]. M-SiNPs have been identified to induce toxicity in the GIT, with inflammation, oxidative stress, and harm to the intestinal barrier. SiNPs administration has been found to cause intestinal inflammation by interfering with the hydrolysis and metabolism of nutrient peptides in Caenorhabditis elegans (an invertebrate nematode) [45]. Furthermore, Ogawa et al. [88] revealed that administering 10-nm SiNPs provoked intestinal inflammation by activating an apoptosis-associated speck-like protein with a CARD (caspase activation and recruitment domain) inflammasome.

3.4 Metallic-based nanoparticles

Numerous scientific investigations have been conducted on the therapeutic applications of gold nanoparticles (AuNPs), silver nanoparticles (AgNPs) and superparamagnetic iron oxide nanoparticles (SPIONPs) [55, 89]. They possess several characteristics that make them attractive as DDS. Specifically, they can be easily synthesised in various sizes and shapes, their surfaces can be functionalized with different elements such as polymers, peptides, targeting ligands, imaging probes, and more, and they are generally considered safe for certain in vivo biological applications [55].

AgNPs exhibit distinct properties related to toxicity, surface plasmon resonance, and electrical resistance [90]. In a study by Vandebriel et al. [91], repeated exposure to AgNPs resulted in cytotoxic effects on various rat cells. Intending to create new anticancer treatments, Azizi et al. [92] developed albumin-coated AgNPs, which were found to be specifically taken up by cancerous cells and trigger apoptosis. Furthermore, through numerous pathways, AgNPs are highly effective against gram-positive and gram-negative bacteria. They help address the problem of drug resistance often seen with traditional antibiotics due to their unique mode of action [54]. Clear evidence of systemic toxicity from AgNPs ingested orally or through intravenous routes has been demonstrated, and the toxic effects are related to the amount of Ag+ released during exposure [93]. Moreover, the significant surface area of AgNPs that release Ag+ ions is a critical aspect that contributes to the cytotoxic behaviour. As widely recognised, smaller AgNPs exhibit a faster pace of silver ion (Ag+) dissolution in the nearby microenvironment, owing to their greater surface area-to-volume ratio. This results in increased bioavailability, improved distribution, and increased toxicity compared to larger AgNPs [40, 94]. Research examining the anti-inflammatory properties of AgNPs has shown a considerable decrease in wound inflammation, adjustment of fibrogenic cytokines, a reduction of pro-inflammatory cytokines, and cell death in inflammatory cells [52, 53]. NPs with diameters below 100 nm have been documented to be mainly taken up by endocytosis in epithelial cells. Within these cells, AgNPs can induce oxidative stress, DNA damage, and inflammation [95]. Jeong et al. [96] observed an increase in goblet cells in the intestines and a significant release of mucus granules in mice treated with oral AgNPs (60 nm) at a 30 mg/kg body weight per day for 28 days. Furthermore, AgNPs administered orally (5-20 nm) for 21 days in mice (20 mg/kg body weight) disrupted the microvilli of epithelial cells and affected the intestinal glands [93].

Various in vivo studies have been conducted to assess the possible toxic effects of AuNPs, but the findings still need to be definitive [51]. Factors such as size, shape, surface properties, stabilising coatings, and administration aspects (dosage, duration, and method of administration) can lead to the varying toxic effects of AuNP in vivo [46]. Research has shown that smaller AuNPs (5–15 nm) exhibit a more extensive organ distribution in rodents compared to larger AuNPs (50–100 nm), suggesting a higher risk of toxicity in vivo for smaller AuNPs [47]. Research conducted by Goodman and colleagues [97] indicated that cationic AuNPs interact with the negatively charged cellular membrane, causing damage to the intestinal membrane. Furthermore, citrate-capped AuNP (13 nm in diameter) were harmful to human lung carcinoma cells while not affecting human liver carcinoma cells at the same dose [98]. As additional information is collected, it has been recommended that proper consideration be given to surface chemistry and dosages of Au and AgNPs to utilise them in biomedical applications efficiently [49, 50].

SPIONPs have become increasingly popular in numerous biomedical applications, such as magnetic resonance imaging, targeted drug or gene delivery, and hyperthermia [57]. However, SPIONPs can potentially cause cytotoxicity, negatively impacting vital cellular components such as mitochondria, the nucleus, and DNA [99]. Research examining the influence of various surface coatings on cell behaviour and structure revealed that dextran-magnetite (Fe3O4) NPs lead to cell death and decreased proliferation, similar to the effects of uncoated iron oxide particles [56]. Contact with SPIONPs has been linked to considerable harmful consequences, including inflammation, developing apoptotic structures, compromised mitochondrial functioning (MTT), membrane leakage, ROS production, extensive chromosomal aberration, and condensation [57].

Advertisement

4. In vitro tests to evaluate the toxicity of nanoparticles

The in vitro evaluation of the toxicity of NPs serves as a standard approach for identifying acute hazards associated with potentially harmful NPs during this screening process. However, these methods can only partially replace in vivo evaluations. At the same time, in vitro analyses report the immediate harmful effects of NPs in specific cellular settings, in vivo animal models monitor biodistribution and bioaccumulation pathways, which are not accessible through in vitro observations. A comprehensive understanding of NPs toxicity and potential risk requires using both methods [100]. In vitro, NPs toxicity assessment is a critical technique that offers benefits such as reduced cost, quicker results, and minimal ethical issues [101].

4.1 Size and surface charge evaluation

Various analytical approaches exist to examine the toxicological properties of NPs, with two key techniques frequently employed to provide crucial quantitative data: dynamic light scattering (DLS) and zeta potential (ZP) analysis. The ZP and, subsequently, the surface charge of a particle play an important role in inferring potential toxicity, promoting combined DLS-ZP systems as vital tools for preliminary biocompatibility assessments. Other in vitro tests utilised to determine the size and surface charge of NPs encompass scanning electron microscopy (SEM), transmission electron microscopy (TEM), atomic force spectroscopy (AFM), Fourier transform infrared spectroscopy (FTIR), X-ray diffraction (XRD), surface-enhanced Raman spectroscopy (SERS) and solid-state nuclear magnetic resonance spectroscopy (SSNMR) for compositional analysis, as well as fluorometry for photonic characteristics [102].

4.2 Proliferation assay

Proliferation analysis is conducted by evaluating cell metabolic function to determine cell metabolism using 3-(4-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), the most commonly utilised tetrazolium bromide for evaluating NPs toxicity [103]. This test uses tetrazolium salt to gauge cellular metabolism. The intricate chemical preparation of the MTT assay has led to its substitution for the Alamar blue assay, which is simpler to prepare and measures the cellular redox potential [104]. However, its success has been hindered due to an unclear operating mechanism. Other evaluations include the [3H] thymidine incorporation method to assess cell proliferation, which is typically avoided due to its toxicity [105]. The cologenic assay is another method that counts proliferating cells by visually inspecting them after exposure to NPs [106].

4.3 Apoptosis assay

One primary indicator of nanoparticles (NPs)-induced cell toxicity is apoptosis. This process, triggered by NPs, can lead to cellular renewal and accelerated ageing in mammalian hosts [100]. Commonly used markers for apoptosis include phosphatidylserine (PS), which moves to the cell membrane’s outer layer, and caspase activation. Research has shown that silver NP can induce apoptosis in cells such as mouse embryonic stem cells [107] and stimulate the activation of Caspase 3 and 9 when applied to Drosophila melanogaster larvae [108]. Once host signalling pathways are initiated, active caspases provide various assays, such as cleavable substrates with fluorogenic and chromogenic labels, immunoblotting, immunofluorescence, and affinity assays with connected reporter units [109].

Numerous in vitro tests evaluate NPs toxicity, such as the Annexin V assay, which binds to PS and is a barrier against coagulation cascades. There are also various other techniques for examining apoptosis in cells or tissues exposed to NPs, including the comet assay, the TUNEL (terminal deoxynucleotidyl transferase (TdT) Nick-End labelling) staining technique, and analysing morphological alterations [110, 111, 112]. A decrease in cell size and DNA fragmentation are signs of apoptosis in NPs-treated cells. DNA gel electrophoresis is the most straightforward test for identifying cellular abnormalities. DNA fragmentation with uneven DNA sizes in agarose gel signifies necrosis-mediated cell death, while ladder-like electrophoretic DNA patterns indicate apoptosis-induced cell death in NPs-treated cells [113, 114]. Such changes were observed in human HepG2 hepatoma cells exposed to silica NPs [115]. Furthermore, the single-cell gel electrophoresis assay (SCGE), also known as the comet assay, is used to detect the mutagenic potential of NPs-treated cells by identifying DNA breaks in Drosophila tertiolecta exposed to SiO and TiO NPs [70, 116].

4.4 In vitro assessment of oxidative stress

As a result of the high surface area-to-volume ratio imposed by NPs, enhanced reactivity promotes intracellular damage due to oxidative stress. Detection of oxidative events through ROS measurement, protein carbonyl content, genotoxicity, and inflammatory markers reveal the potential of NPs to produce harmful toxicants in the host [100]. Beyond the natural levels, ROS levels that are produced in the cytoplasm can be detected using ROS-sensitive dyes such as nonionic, non-polar, membrane permeable fluorophore 2′,7′-dichlorofluorescein diacetate (DCFH-DA) [117]. After cellular entry of DCFH-DA, the fluorophore is hydrolysed enzymatically by cytosolic esterase into non-fluorescent polar analogue dichlorofluorescein (DCFH). It is oxidised by cellular ROS into highly fluorescent dichlorofluorescein (DCF) that can be monitored [118]. The use of other agents, such as 2,2,6,6-tetramethylpiperidine (TEMP), detect ROS by reacting with the stable O2 radical, which can be detected using X-band electron paramagnetic resonance (EPR) [119]. However, the use of DCFH-DA is advantageous over that of TEMP because of its high cost. Other available assays to detect ROS induced due to NPs can be detected using lipid hydroperoxide, Amplex Red assay, measurement of antioxidant depletion by 5,5′-dithiobis-(2-nitrobenzoic acid) (DTNB) and superoxide dismutase (SOD) activity by Nitro blue tetrazolium assay [120].

4.5 Necrosis assay

Necrosis disrupts the integrity of the membrane and is commonly used to determine the viability of cells. Necrosis is measured using two dyes Neutral Red and trypan Blue. Neutral red (2-amino-3 methyl-7-dimethyl-amino-phenazonium-chloride) is a weakly cationic supravital dye that, at slightly acid pH, yields a deep red colour [121]. The mechanism of action of neutral red includes diffusion through the plasma membrane and concentrates on the binding of lysosomes through electrostatic, hydrophobic bonds [101]. Any disruption in the cell membrane brought about by NPs results in reduced uptake of neutral red, differentiating live and necrotic cells. Studies have used neutral red to detect necrotic cells induced when exposed to silver NPs [122]. The other dye used for detecting necrotic cells induced through NPs is trypan blue which enters dead cells while being excluded by live cells and thus is used to detect the stability of the cell membrane. Trypan blue has been used to detect the cytotoxicity of cells exerted by Zn NPs [123].

4.6 Viability assays

Lactate dehydrogenase (LDH) is an enzyme produced by living cells that regulates pyruvate and lactate levels through the oxidation of nicotinamide adenine dinucleotide (NAD). When cells are lysed due to the toxic effects of NPs and other agents, LDH is released into the surroundings and maintains the enzyme activity. The usual conversion of pyruvate to lactate in the presence of LDH follows the NADH + pyruvate NAD + + lactate. Spectroscopic methods can monitor the NADH complementary reaction with tetrazolium salts. This time-dependent change in spectroscopic absorbance through enzyme-linked immunosorbent assay (ELISA) tests infers the extent of cellular trauma [100]. Common tetrazolium salts include iodonitrotetrazolium (INT), 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), and 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxy-phenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) which rival the throughput of the 51Cr assay and exhibit a threefold increase in sensitivity over ultraviolet NADH assays [124]. In addition to these, luminogenic glycylphenylalanyl-aminofluorocoumarin (GF-AFC) and firefly luciferase ATP assays provide non-toxic and highly sensitive options for assessing cell viability [125].

Advertisement

5. In vivo tests to evaluate the toxicity of nanoparticles

In vivo toxicity assessment methods provide more reliable data than in vitro models, and often yield conflicting results [126]. Some examples of in vivo toxicity assessment methods for NPs include biodistribution, clearance, haematology, serum chemistry, and histopathology [101]. For example, researchers have studied the toxicity of aluminium oxide NPs. These NPs have been analysed for their cell toxicity, immunotoxicity, and genotoxicity using in vivo models [126]. Tetrazolium-based assays such as MTT, MTS, and WST-1 have been used to evaluate the cytotoxicity of these NPs [126]. Manganese oxide nanomaterials have also been examined for their potentially harmful effects [127]. One study evaluated the toxicity of a newly synthesised nanomaterial called GNA35 and its precursor Mn3O4 using in vitro models representing the respiratory, GIT, and skin systems of the human body [127]. The study provided information on the potential health risks associated with these nanomaterials, highlighting the need for further evaluation using in vivo models [127].

In vivo toxicity assessment of various NPs is crucial for assessing their safety in biomedical and industrial applications. Here are some examples of in vivo toxicity assessment of various NPs:

Polymer NPs, often used for drug delivery systems, have been evaluated for toxicity using in vivo models. A study utilised zebrafish embryos as a model to investigate the toxicity of polystyrene NPs. The study found that NPs demonstrated dose-dependent toxicity, leading to developmental and behavioural abnormalities in zebrafish embryos [46].

Lipid-based NPs are commonly used in drug delivery systems because of their biocompatibility and low toxicity. In a study by Lama et al., researchers evaluated the in vivo toxicity of lipid NPs in rats by measuring their biodistribution, clearance, and histopathology [128]. They found lipid NPs were well tolerated, with no significant adverse effects observed [101].

Protein NPs, such as albumin-based NPs, have been developed for targeted drug delivery. In vivo studies in mice have shown that these NPs can accumulate in tumour tissues and effectively deliver drugs without causing significant systemic toxicity. However, further evaluation of protein NPs using in vivo models is necessary to ensure the safety of these drug delivery systems [129].

Silica NPs have many applications, from drug delivery to cosmetics. In a study by Yu et al., researchers assessed the in vivo toxicity of silica NPs in mice by assessing their biodistribution, clearance, and inflammatory response. They found that silica NPs were distributed primarily in the liver, spleen, and kidneys and induced a transient inflammatory response, suggesting potential risks associated with their use [130].

AuNPs have been studied for their potential applications in various fields, including drug delivery and diagnostics. In vivo toxicity evaluation of gold NPs has been carried out using different animal models. For example, a study investigated the biodistribution and toxicity of AuNPs in mice using various doses and sizes. The results showed that, depending on size and dose, AuNPs could accumulate in organs such as the liver, spleen, lungs, and kidneys, leading to potential toxicity problems [46, 129].

AgNPs have also been investigated for potential applications in medicine and industry. An example of in vivo toxicity assessment of AgNPs is a study that evaluated silver NPs stabilised with gum Arabic protein (AgNPs-GP) in Daphnia, a small freshwater crustacean model. The results indicated that AgNPs-GP exhibited dose-dependent toxicity, causing adverse effects on Daphnia’s survival and reproduction [131].

Iron oxide NPs (SPIONPs) are widely used in biomedical applications, such as drug delivery and magnetic resonance imaging. In a study by Mahmoudi et al., the researchers evaluated the in vivo toxic effects of SPIONPs in mice by observing their biodistribution, retention, and clearance [132]. They found that SPIONPs did not exhibit significant toxicity, but NPs tended to accumulate in the liver and spleen, which can cause long-term adverse effects [57].

Advertisement

6. Conclusion

Throughout the years, the remarkable properties of NPs have led to their recognition as marvels of contemporary science because of their extensive applications. The employment of NPs in the biomedical field has contributed significantly to overcoming numerous diseases and improving the quality of human life. Unlike other delivery methods, orally administering NPs exposes them to various biological and chemical conditions, such as enzyme and microbiota-related digestion and fluctuations in ionic strength and pH levels. Nanotoxicity generally stems from the instability of the NPs and distinct physicochemical properties. Numerous factors, such as the type of NPs, particle and pore size, the extent of modification, frequency and amount of dosage, and administration timing, influence them. Other crucial factors include cell type, cell condition, organ distribution, animal condition, and delivery duration, presenting individual differences in cellular and in vivo contexts. NPs toxicity mechanisms include oxidative stress, inflammation, and apoptosis; however, studies of the implicated signal pathways are still relatively scarce. Due to the numerous variables involved, there currently needs to be a standardised approach to toxicity research. Investigations of NPs toxicity should consider their actual applications and refine safety evaluation indicators for both in vitro and in vivo testing. As observed, a limited amount of data is available for specific NPs types used in DDS. Consequently, before introducing NPs into the therapeutic market, it is essential to evaluate the risk-benefit ratio.

References

  1. 1. Yetisgin AA, Cetinel S, Zuvin M, Kosar A, Kutlu O. Therapeutic nanoparticles and their targeted delivery applications. Molecules. 2020;25(9):2193. DOI: 10.3390/molecules25092193
  2. 2. Patra JK et al. Nano based drug delivery systems: Recent developments and future prospects. Journal of Nanobiotechnology. 2018;16(1):71. DOI: 10.1186/s12951-018-0392-8
  3. 3. Nasrollahzadeh M, Sajadi SM, Sajjadi M, Issaabadi Z. An Introduction to Nanotechnology, Interface Science and Technology. Amsterdam, The Netherlands: Elsevier; 2019. pp. 1-27. DOI: 10.1016/b978-0-12-813586-0.00001-8
  4. 4. Mitchell MJ, Billingsley MM, Haley RM, Wechsler ME, Peppas NA, Langer R. Engineering precision nanoparticles for drug delivery. Nature Reviews. Drug Discovery. 2021;20(2):101-124. DOI: 10.1038/s41573-020-0090-8
  5. 5. Carissimi G, Montalbán MG, Fuster MG, Víllora G. Silk Fibroin Nanoparticles: Synthesis and Applications as Drug Nanocarriers, 21st Century Nanostructured Materials - Physics, Chemistry, Classification, and Emerging Applications in Industry, Biomedicine, and Agriculture. London: IntechOpen; 2022. DOI: 10.5772/intechopen.100386
  6. 6. Palmerston Mendes L, Pan J, Torchilin VP. Dendrimers as Nanocarriers for nucleic acid and drug delivery in cancer therapy. Molecules. 2017;22(9):1401. DOI: 10.3390/molecules22091401
  7. 7. Kadam RS, Bourne DWA, Kompella UB. Nano-advantage in enhanced drug delivery with biodegradable nanoparticles: Contribution of reduced clearance. Drug Metabolism and Disposition. 2012;40(7):1380-1388. DOI: 10.1124/dmd.112.044925
  8. 8. Obeid MA, Al Qaraghuli MM, Alsaadi M, Alzahrani AR, Niwasabutra K, Ferro VA. Delivering natural products and biotherapeutics to improve drug efficacy. Therapeutic Delivery. 2017;8(11):947-956. DOI: 10.4155/tde-2017-0060
  9. 9. Zhan S, Paik A, Onyeabor F, Ding B, Prabhu S, Wang J. Oral bioavailability evaluation of Celastrol-encapsulated silk fibroin nanoparticles using an optimized LC-MS/MS method. Molecules. 2020;25(15):3422. DOI: 10.3390/molecules25153422
  10. 10. Alqahtani MS, Kazi M, Alsenaidy MA, Ahmad MZ. Advances in Oral drug delivery. Frontiers in Pharmacology. 2021;12:618411. DOI: 10.3389/fphar.2021.618411
  11. 11. Mudie DM, Amidon GL, Amidon GE. Physiological parameters for oral delivery and in vitro testing. Molecular Pharmaceutics. 2010;7(5):1388-1405. DOI: 10.1021/mp100149j
  12. 12. Majumder J, Taratula O, Minko T. Nanocarrier-based systems for targeted and site specific therapeutic delivery. Advanced Drug Delivery Reviews. 2019;144:57-77. DOI: 10.1016/j.addr.2019.07.010
  13. 13. Lou J et al. Advances in Oral drug delivery systems: Challenges and opportunities. Pharmaceutics. 2023;15(2):484. DOI: 10.3390/pharmaceutics15020484
  14. 14. Joseph TM et al. Nanoparticles: Taking a unique position in medicine. Nanomaterials (Basel). 2023;13(3):574. DOI: 10.3390/nano13030574
  15. 15. Cabellos J et al. Short-term oral administration of non-porous and mesoporous silica did not induce local or systemic toxicity in mice. Nanotoxicology. 2020;14(10):1324-1341. DOI: 10.1080/17435390.2020.1818325
  16. 16. Sharma S, Parveen R, Chatterji BP. Toxicology of nanoparticles in drug delivery. Current Pathobiology Reports. 2021;9(4):133-144. DOI: 10.1007/s40139-021-00227-z
  17. 17. Zhang Q et al. A superoxide dismutase/catalase mimetic nanomedicine for targeted therapy of inflammatory bowel disease. Biomaterials. 2016;105:206-221. DOI: 10.1016/j.biomaterials.2016.08.010
  18. 18. Vong LB, Mo J, Abrahamsson B, Nagasaki Y. Specific accumulation of orally administered redox nanotherapeutics in the inflamed colon reducing inflammation with dose–response efficacy. Journal of Controlled Release. 2015;210:19-25. DOI: 10.1016/j.jconrel.2015.05.275
  19. 19. Geiser M et al. Ultrafine particles cross cellular membranes by nonphagocytic mechanisms in lungs and in cultured cells. Environmental Health Perspectives. 2005;113(11):1555-1560. DOI: 10.1289/ehp.8006
  20. 20. Fu PP, Xia Q, Hwang H-M, Ray PC, Yu H. Mechanisms of nanotoxicity: Generation of reactive oxygen species. Journal of Food and Drug Analysis. 2014;22(1):64-75. DOI: 10.1016/j.jfda.2014.01.005
  21. 21. Yusuf A, Almotairy ARZ, Henidi H, Alshehri OY, Aldughaim MS. Nanoparticles as drug delivery systems: A review of the implication of nanoparticles’ physicochemical properties on responses in biological systems. Polymers (Basel). 2023;15(7):1596. DOI: 10.3390/polym15071596
  22. 22. Lu W, Yao J, Zhu X, Qi Y. Nanomedicines: Redefining traditional medicine. Biomedicine & Pharmacotherapy. 2021;134:111103. DOI: 10.1016/j.biopha.2020.111103
  23. 23. Hosseini A et al. Exosome-inspired targeting of cancer cells with enhanced affinity. Journal of Materials Chemistry B. 2016;4(4):768-778. DOI: 10.1039/c5tb01741f
  24. 24. Teng Y et al. Plant-derived Exosomal MicroRNAs shape the gut microbiota. Cell Host & Microbe. 2018;24(5):637-652.e8. DOI: 10.1016/j.chom.2018.10.001
  25. 25. Xia W et al. Targeted delivery of drugs and genes using polymer Nanocarriers for cancer therapy. International Journal of Molecular Sciences. 2021;22(17):9118. DOI: 10.3390/ijms22179118
  26. 26. Quinson J et al. Investigating particle size effects in catalysis by applying a size-controlled and surfactant-free synthesis of colloidal nanoparticles in alkaline ethylene glycol: Case study of the oxygen reduction reaction on Pt. ACS Catalysis. 2018;8(7):6627-6635. DOI: 10.1021/acscatal.8b00694
  27. 27. Bergin IL, Witzmann FA. Nanoparticle toxicity by the gastrointestinal route: Evidence and knowledge gaps. International Journal of Biomedical Nanoscience and Nanotechnology. 2013;3(1-2):163-210. DOI: 10.1504/IJBNN.2013.054515
  28. 28. Greenwood-Van Meerveld B, Johnson AC, Grundy D. Gastrointestinal Physiology and Function, Gastrointestinal Pharmacology. New York, USA: Springer International Publishing; 2017. pp. 1-16. DOI: 10.1007/164_2016_118
  29. 29. Lim YF et al. An exploration of the microrheological environment around the distal ileal villi and proximal colonic mucosa of the possum (Trichosurus vulpecula). Journal of the Royal Society, Interface. 2013;10(81):20121008. DOI: 10.1098/rsif.2012.1008
  30. 30. Azman M, Sabri AH, Anjani QK, Mustaffa MF, Hamid KA. Intestinal absorption study: Challenges and absorption enhancement strategies in improving Oral drug delivery. Pharmaceuticals (Basel). 2022;15(8):975. DOI: 10.3390/ph15080975
  31. 31. Mowat AM, Agace WW. Regional specialization within the intestinal immune system. Nature Reviews. Immunology. 2014;14(10):667-685. DOI: 10.1038/nri3738
  32. 32. Brannon-Peppas L, Blanchette JO. Nanoparticle and targeted systems for cancer therapy. Advanced Drug Delivery Reviews. 2012;64:206-212. DOI: 10.1016/j.addr.2012.09.033
  33. 33. Padhi S, Behera A. Cellular Internalization and Toxicity of Polymeric Nanoparticles, Environmental Chemistry for a Sustainable World. New York, USA: Springer International Publishing; 2022. pp. 473-488. DOI: 10.1007/978-3-031-14848-4_17
  34. 34. Yu J, Chen L, Gu W, Liu S, Wu B. Heterogeneity effects of nanoplastics and lead on zebrafish intestinal cells identified by single-cell sequencing. Chemosphere. 2022;289:133133. DOI: 10.1016/j.chemosphere.2021.133133
  35. 35. Chen J et al. Surface functionalization-dependent inflammatory potential of polystyrene nanoplastics through the activation of MAPK/NF-κB signaling pathways in macrophage raw 264.7. Ecotoxicology and Environmental Safety. 2023;251:114520. DOI: 10.1016/j.ecoenv.2023.114520
  36. 36. Dass CR. Lipoplex-mediated delivery of nucleic acids: Factors affecting in vivo transfection. Journal of Molecular Medicine. 2004;82(9):579-591. DOI: 10.1007/s00109-004-0558-8
  37. 37. El Moukhtari SH, Rodríguez-Nogales C, Blanco-Prieto MJ. Oral lipid nanomedicines: Current status and future perspectives in cancer treatment. Advanced Drug Delivery Reviews. 2021;173:238-251. DOI: 10.1016/j.addr.2021.03.004
  38. 38. Syama K, Jakubek ZJ, Chen S, Zaifman J, Tam YYC, Zou S. Development of lipid nanoparticles and liposomes reference materials (II): Cytotoxic profiles. Scientific Reports. 2022;12(1):18071. DOI: 10.1038/s41598-022-23013-2
  39. 39. Parhiz H et al. Added to pre-existing inflammation, mRNA-lipid nanoparticles induce inflammation exacerbation (IE). Journal of Controlled Release. 2022;344:50-61. DOI: 10.1016/j.jconrel.2021.12.027
  40. 40. Shang L, Nienhaus K, Nienhaus GU. Engineered nanoparticles interacting with cells: Size matters. Journal of Nanobiotechnology. 2014;12:5. DOI: 10.1186/1477-3155-12-5
  41. 41. McClements DJ, DeLoid G, Pyrgiotakis G, Shatkin JA, Xiao H, Demokritou P. The role of the food matrix and gastrointestinal tract in the assessment of biological properties of ingested engineered nanomaterials (iENMs): State of the science and knowledge gaps. NanoImpact. 2019;3-4:47-57. DOI: 10.1016/j.impact.2016.10.002
  42. 42. Hu H et al. Silicon dioxide nanoparticles induce insulin resistance through endoplasmic reticulum stress and generation of reactive oxygen species. Particle and Fibre Toxicology. 2019;16(1):41. DOI: 10.1186/s12989-019-0327-z
  43. 43. Liu J et al. Silica nanoparticles induce spermatogenesis disorders via L3MBTL2-DNA damage-p53 apoptosis and RNF8-ubH2A/ubH2B pathway in mice. Environmental Pollution. 2020;265:114974. DOI: 10.1016/j.envpol.2020.114974
  44. 44. Huang Y et al. Silica nanoparticles: Biomedical applications and toxicity. Biomedicine and Pharmacotherapy. 2022;151:113053. DOI: 10.1016/j.biopha.2022.113053
  45. 45. Piechulek A, Berwanger LC, von Mikecz A. Silica nanoparticles disrupt OPT-2/PEP-2-dependent trafficking of nutrient peptides in the intestinal epithelium. Nanotoxicology. 2019;13(8):1133-1148. DOI: 10.1080/17435390.2019.1643048
  46. 46. Adewale OB, Davids H, Cairncross L, Roux S. Toxicological behavior of gold nanoparticles on various models: Influence of physicochemical properties and other factors. International Journal of Toxicology. 2019;38(5):357-384. DOI: 10.1177/1091581819863130
  47. 47. De Jong WH, Hagens WI, Krystek P, Burger MC, Sips AJAM, Geertsma RE. Particle size-dependent organ distribution of gold nanoparticles after intravenous administration. Biomaterials. 2008;29(12):1912-1919. DOI: 10.1016/j.biomaterials.2007.12.037
  48. 48. Kim JH, Kim JH, Kim K-W, Kim MH, Yu YS. Intravenously administered gold nanoparticles pass through the blood–retinal barrier depending on the particle size, and induce no retinal toxicity. Nanotechnology. 2009;20(50):505101. DOI: 10.1088/0957-4484/20/50/505101
  49. 49. Alkilany AM, Murphy CJ. Toxicity and cellular uptake of gold nanoparticles: What we have learned so far? Journal of Nanoparticle Research. 2010;12(7):2313-2333. DOI: 10.1007/s11051-010-9911-8
  50. 50. Ferdous Z, Nemmar A. Health impact of silver nanoparticles: A review of the biodistribution and toxicity following various routes of exposure. International Journal of Molecular Sciences. 2020;21(7):23750. DOI: 10.3390/ijms21072375
  51. 51. Sun Q et al. Cytotoxicity and cellular responses of gold Nanorods to smooth muscle cells dependent on surface chemistry coupled action. Small. 2018;14(52):1803715. DOI: 10.1002/smll.201803715
  52. 52. David L et al. Green synthesis, characterization and anti-inflammatory activity of silver nanoparticles using European black elderberry fruits extract. Colloids and Surfaces. B, Biointerfaces. 2014;122:767-777. DOI: 10.1016/j.colsurfb.2014.08.018
  53. 53. Hebeish A, El-Rafie MH, EL-Sheikh MA, Seleem AA, El-Naggar ME. Antimicrobial wound dressing and anti-inflammatory efficacy of silver nanoparticles. International Journal of Biological Macromolecules. 2014;65:509-515. DOI: 10.1016/j.ijbiomac.2014.01.071
  54. 54. Le Ouay B, Stellacci F. Antibacterial activity of silver nanoparticles: A surface science insight. Nano Today. 2015;10(3):339-354. DOI: 10.1016/j.nantod.2015.04.002
  55. 55. Pasparakis G. Recent developments in the use of gold and silver nanoparticles in biomedicine. Wiley Interdisciplinary Reviews. Nanomedicine and Nanobiotechnology. 2022;14(5):e1817-e1817. DOI: 10.1002/wnan.1817
  56. 56. Berry CC, Wells S, Charles S, Curtis ASG. Dextran and albumin derivatised iron oxide nanoparticles: Influence on fibroblasts in vitro. Biomaterials. 2003;24(25):4551-4557. DOI: 10.1016/s0142-9612(03)00237-0
  57. 57. Singh N, Jenkins GJS, Asadi R, Doak SH. Potential toxicity of superparamagnetic iron oxide nanoparticles (SPION). Nano Reviews. 2010;1:1-15. DOI: 10.3402/nano.v1i0.5358
  58. 58. Sabourian P et al. Effect of physico-chemical properties of nanoparticles on their intracellular uptake. International Journal of Molecular Sciences. 2020;21(21):8019. DOI: 10.3390/ijms21218019
  59. 59. Date AA, Hanes J, Ensign LM. Nanoparticles for oral delivery: Design, evaluation and state-of-the-art. Journal of Controlled Release. 2016;240:504-526. DOI: 10.1016/j.jconrel.2016.06.016
  60. 60. Lai SK, Wang Y-Y, Hanes J. Mucus-penetrating nanoparticles for drug and gene delivery to mucosal tissues. Advanced Drug Delivery Reviews. 2009;61(2):158-171. DOI: 10.1016/j.addr.2008.11.002
  61. 61. Wang Y, Pi C, Feng X, Hou Y, Zhao L, Wei Y. The influence of nanoparticle properties on Oral bioavailability of drugs. International Journal of Nanomedicine. 2020;15:6295-6310. DOI: 10.2147/ijn.s257269
  62. 62. Zhao Z et al. A nanoparticle-based nicotine vaccine and the influence of particle size on its immunogenicity and efficacy. Nanomedicine. 2017;13(2):443-454. DOI: 10.1016/j.nano.2016.07.015
  63. 63. Mostafalou S, Mohammadi H, Ramazani A, Abdollahi M. Different biokinetics of nanomedicines linking to their toxicity; an overview. Daru. 2013;21(1):14. DOI: 10.1186/2008-2231-21-14
  64. 64. Abbasi R, Shineh G, Mobaraki M, Doughty S, Tayebi L. Structural parameters of nanoparticles affecting their toxicity for biomedical applications: A review. Journal of Nanoparticle Research. 2023;25(3):43. DOI: 10.1007/s11051-023-05690-w
  65. 65. Vedhanayagam M, Unni Nair B, Sreeram KJ. Collagen-ZnO scaffolds for wound healing applications: Role of dendrimer functionalization and nanoparticle morphology. ACS Applied Bio Materials. 2018;1(6):1942-1958. DOI: 10.1021/acsabm.8b00491
  66. 66. Shukla S et al. The impact of aspect ratio on the biodistribution and tumor homing of rigid soft-matter Nanorods. Advanced Healthcare Materials. 2015;4(6):874-882. DOI: 10.1002/adhm.201400641
  67. 67. Fubini B, Fenoglio I, Tomatis M, Turci F. Effect of chemical composition and state of the surface on the toxic response to high aspect ratio nanomaterials. Nanomedicine. 2011;6(5):899-920. DOI: 10.2217/nnm.11.80
  68. 68. Liu M et al. Efficient mucus permeation and tight junction opening by dissociable ‘mucus-inert’ agent coated trimethyl chitosan nanoparticles for oral insulin delivery. Journal of Controlled Release. 2016;222:67-77. DOI: 10.1016/j.jconrel.2015.12.008
  69. 69. Du X-J et al. The effect of surface charge on oral absorption of polymeric nanoparticles. Biomaterials Science. 2018;6(3):642-650. DOI: 10.1039/c7bm01096f
  70. 70. González-García LE et al. Nanoparticles surface chemistry influence on protein Corona composition and inflammatory responses. Nanomaterials (Basel). 2022;12(4):682. DOI: 10.3390/nano12040682
  71. 71. Marassi V et al. Silver nanoparticles as a medical device in healthcare settings: A five-step approach for candidate screening of coating agents. Royal Society Open Science. 2018;5(1):171113. DOI: 10.1098/rsos.171113
  72. 72. Prasher P et al. Targeting mucus barrier in respiratory diseases by chemically modified advanced delivery systems. Chemico-Biological Interactions. 2022;365:110048. DOI: 10.1016/j.cbi.2022.110048
  73. 73. Grabowski N et al. Surface coating mediates the toxicity of polymeric nanoparticles towards human-like macrophages. International Journal of Pharmaceutics. 2015;482(1-2):75-83. DOI: 10.1016/j.ijpharm.2014.11.042
  74. 74. Henson TE, Navratilova J, Tennant AH, Bradham KD, Rogers KR, Hughes MF. In vitro intestinal toxicity of copper oxide nanoparticles in rat and human cell models. Nanotoxicology. 2019;13(6):795-811. DOI: 10.1080/17435390.2019.1578428
  75. 75. Yu Z et al. Corrigendum: Yu et al. PPy@Fe(3)O(4) nanoparticles inhibit the proliferation and metastasis of CRC via suppressing the NF-κB signaling pathway and promoting ferroptosis. Frontiers in Bioengineering and Biotechnology. 2022;10:1094064. DOI: 10.3389/fbioe.2022.1094064
  76. 76. De Berardis B et al. Exposure to ZnO nanoparticles induces oxidative stress and cytotoxicity in human colon carcinoma cells. Toxicology and Applied Pharmacology. 2010;246(3):116-127. DOI: 10.1016/j.taap.2010.04.012
  77. 77. Mittag A, Singer A, Hoera C, Westermann M, Kämpfe A, Glei M. Impact of in vitro digested zinc oxide nanoparticles on intestinal model systems. Particle and Fibre Toxicology. 2022;19(1):39. DOI: 10.1186/s12989-022-00479-6
  78. 78. Seo Y et al. Recent Progress of lipid nanoparticles-based lipophilic drug delivery: Focus on surface modifications. Pharmaceutics. 2023;15(3):772. DOI: 10.3390/pharmaceutics15030772
  79. 79. Tenchov R, Bird R, Curtze AE, Zhou Q. Lipid nanoparticles–from liposomes to mRNA vaccine delivery, a landscape of research diversity and advancement. ACS Nano. 2021;15(11):16982-17015. DOI: 10.1021/acsnano.1c04996
  80. 80. Satapathy MK et al. Solid lipid nanoparticles (SLNs): An advanced drug delivery system targeting brain through BBB. Pharmaceutics. 2021;13(8):1183. DOI: 10.3390/pharmaceutics13081183
  81. 81. Lv H, Zhang S, Wang B, Cui S, Yan J. Toxicity of cationic lipids and cationic polymers in gene delivery. Journal of Controlled Release. 2006;114(1):100-109. DOI: 10.1016/j.jconrel.2006.04.014
  82. 82. Ball RL, Bajaj P, Whitehead KA. Oral delivery of siRNA lipid nanoparticles: Fate in the GI tract. Scientific Reports. 2018;8(1):1-12. DOI: 10.1038/s41598-018-20632-6
  83. 83. Guo Z, Martucci NJ, Liu Y, Yoo E, Tako E, Mahler GJ. Silicon dioxide nanoparticle exposure affects small intestine function in an in vitro model. Nanotoxicology. 2018;12(5):485-508. DOI: 10.1080/17435390.2018.1463407
  84. 84. Sakai-Kato K, Hidaka M, Un K, Kawanishi T, Okuda H. Physicochemical properties and in vitro intestinal permeability properties and intestinal cell toxicity of silica particles, performed in simulated gastrointestinal fluids. Biochimica et Biophysica Acta (BBA) - General Subjects. 2014;1840(3):1171-1180. DOI: 10.1016/j.bbagen.2013.12.014
  85. 85. Diao J et al. Silicon dioxide nanoparticles induced neurobehavioral impairments by disrupting microbiota-gut-brain axis. Journal of Nanobiotechnology. 2021;19(1):174. DOI: 10.1186/s12951-021-00916-2
  86. 86. Fruijtier-Pölloth C. The safety of nanostructured synthetic amorphous silica (SAS) as a food additive (E 551). Archives of Toxicology. 2016;90(12):2885-2916. DOI: 10.1007/s00204-016-1850-4
  87. 87. Kusaczuk M, Krętowski R, Naumowicz M, Stypułkowska A, Cechowska-Pasko M. Silica nanoparticle-induced oxidative stress and mitochondrial damage is followed by activation of intrinsic apoptosis pathway in glioblastoma cells. International Journal of Nanomedicine. 2018;13:2279-2294. DOI: 10.2147/IJN.S158393
  88. 88. Ogawa T et al. Oral intake of silica nanoparticles exacerbates intestinal inflammation. Biochemical and Biophysical Research Communications. 2021;534:540-546. DOI: 10.1016/j.bbrc.2020.11.047
  89. 89. Abedin MR et al. Polymer coated gold-ferric oxide superparamagnetic nanoparticles for theranostic applications. Journal of Nanobiotechnology. 2018;16(1):80. DOI: 10.1186/s12951-018-0405-7
  90. 90. Syafiuddin A, Salmiati, Salim MR, Kueh ABH, Hadibarata T, Nur H. A review of silver nanoparticles: Research trends, global consumption, synthesis, properties, and future challenges. Journal of the Chinese Chemical Society. 2017;64(7):732-756. DOI: 10.1002/jccs.201700067
  91. 91. Vandebriel RJ et al. Immunotoxicity of silver nanoparticles in an intravenous 28-day repeated-dose toxicity study in rats. Particle and Fibre Toxicology. 2014;11:21. DOI: 10.1186/1743-8977-11-21
  92. 92. Azizi M, Ghourchian H, Yazdian F, Bagherifam S, Bekhradnia S, Nyström B. Anti-cancerous effect of albumin coated silver nanoparticles on MDA-MB 231 human breast cancer cell line. Scientific Reports. 2017;7(1):5178. DOI: 10.1038/s41598-017-05461-3
  93. 93. van der Zande M et al. Distribution, elimination, and toxicity of silver nanoparticles and silver ions in rats after 28-day Oral exposure. ACS Nano. 2012;6(8):7427-7442. DOI: 10.1021/nn302649p
  94. 94. Recordati C et al. Tissue distribution and acute toxicity of silver after single intravenous administration in mice: Nano-specific and size-dependent effects. Particle and Fibre Toxicology. 2016;13:12. DOI: 10.1186/s12989-016-0124-x
  95. 95. De Matteis V. Exposure to inorganic nanoparticles: Routes of entry, immune response, biodistribution and In vitro/In vivo toxicity evaluation. Toxics. 2017;5(4):29. DOI: 10.3390/toxics5040029
  96. 96. Jeong GN, Jo UB, Ryu HY, Kim YS, Song KS, Yu IJ. Histochemical study of intestinal mucins after administration of silver nanoparticles in Sprague–Dawley rats. Archives of Toxicology. 2009;84(1):63-69. DOI: 10.1007/s00204-009-0469-0
  97. 97. Goodman CM, McCusker CD, Yilmaz T, Rotello VM. Toxicity of gold nanoparticles functionalized with cationic and anionic side chains. Bioconjugate Chemistry. 2004;15(4):897-900. DOI: 10.1021/bc049951i
  98. 98. Patra HK, Banerjee S, Chaudhuri U, Lahiri P, Kr A. Dasgupta, cell selective response to gold nanoparticles. Nanomedicine. 2007;3(2):111-119. DOI: 10.1016/j.nano.2007.03.005
  99. 99. Oberdörster G, Stone V, Donaldson K. Toxicology of nanoparticles: A historical perspective. Nanotoxicology. 2007;1(1):2-25. DOI: 10.1080/17435390701314761
  100. 100. Savage DT, Hilt JZ, Dziubla TD. In vitro methods for assessing nanoparticle toxicity. Methods in Molecular Biology. 2019;1894:1-29. DOI: 10.1007/978-1-4939-8916-4_1
  101. 101. Kumar V, Sharma N, Maitra SS. In vitro and in vivo toxicity assessment of nanoparticles. International Nano Letters. 2017;7(4):243-256. DOI: 10.1007/s40089-017-0221-3
  102. 102. Lin P-C, Lin S, Wang PC, Sridhar R. Techniques for physicochemical characterization of nanomaterials. Biotechnology Advances. 2014;32(4):711-726. DOI: 10.1016/j.biotechadv.2013.11.006
  103. 103. Sayes CM, Reed KL, Warheit DB. Assessing toxicity of fine and nanoparticles: Comparing In vitro measurements to In vivo pulmonary toxicity profiles. Toxicological Sciences. 2007;97(1):163-180. DOI: 10.1093/toxsci/kfm018
  104. 104. Punshon G, Vara D, Sales K, Kidane A, Salacinski H, Seifalian A. Interactions between endothelial cells and a poly(carbonate-silsesquioxane-bridge-urea)urethane. Biomaterials. 2005;26(32):6271-6279. DOI: 10.1016/j.biomaterials.2005.03.034
  105. 105. Hussain SM. Cellular toxicity of hydrazine in primary rat hepatocytes. Toxicological Sciences. 2002;69(2):424-432. DOI: 10.1093/toxsci/69.2.424
  106. 106. Casey A, Herzog E, Davoren M, Lyng FM, Byrne HJ, Chambers G. Spectroscopic analysis confirms the interactions between single walled carbon nanotubes and various dyes commonly used to assess cytotoxicity. Carbon N Y. 2007;45(7):1425-1432. DOI: 10.1016/j.carbon.2007.03.033
  107. 107. Ahamed M et al. DNA damage response to different surface chemistry of silver nanoparticles in mammalian cells. Toxicology and Applied Pharmacology. 2008;233(3):404-410. DOI: 10.1016/j.taap.2008.09.015
  108. 108. Ahamed M, Posgai R, Gorey TJ, Nielsen M, Hussain SM, Rowe JJ. Silver nanoparticles induced heat shock protein 70, oxidative stress and apoptosis in drosophila melanogaster. Toxicology and Applied Pharmacology. 2010;242(3):263-269. DOI: 10.1016/j.taap.2009.10.016
  109. 109. Kaufmann SH et al. Apoptosis-associated caspase activation assays. Methods. 2008;44(3):262-272. DOI: 10.1016/j.ymeth.2007.11.005
  110. 110. Jin Y, Kannan S, Wu M, Zhao JX. Toxicity of luminescent silica nanoparticles to living cells. Chemical Research in Toxicology. 2007;20(8):1126-1133. DOI: 10.1021/tx7001959
  111. 111. Mo Y, Lim L-Y. Paclitaxel-loaded PLGA nanoparticles: Potentiation of anticancer activity by surface conjugation with wheat germ agglutinin. Journal of Controlled Release. 2005;108(2-3):244-262. DOI: 10.1016/j.jconrel.2005.08.013
  112. 112. Pan Y et al. Size-dependent cytotoxicity of gold nanoparticles. Small. 2007;3(11):1941-1949. DOI: 10.1002/smll.200700378
  113. 113. Suman S, Pandey A, Chandna S. An improved non-enzymatic ‘DNA ladder assay’ for more sensitive and early detection of apoptosis. Cytotechnology. 2012;64(1):9-14. DOI: 10.1007/s10616-011-9395-0
  114. 114. Abdel-Khalek AA. Comparative evaluation of genotoxic effects induced by CuO bulk and Nano-particles in Nile tilapia, Oreochromis niloticus. Water, Air, & Pollution. 2015;227(1). DOI: 10.1007/s11270-015-2737-3
  115. 115. Yuan H et al. Sulfur nanoparticles improved plant growth and reduced mercury toxicity via mitigating the oxidative stress in Brassica napus L. Journal of Cleaner Production. 2021;318:128589. DOI: 10.1016/j.jclepro.2021.128589
  116. 116. Handy RD et al. Practical considerations for conducting ecotoxicity test methods with manufactured nanomaterials: What have we learnt so far? Ecotoxicology. 2012;21(4):933-972. DOI: 10.1007/s10646-012-0862-y
  117. 117. Keston AS, Brandt R. The fluorometric analysis of ultramicro quantities of hydrogen peroxide. Analytical Biochemistry. 1965;11(1):1-5. DOI: 10.1016/0003-2697(65)90034-5
  118. 118. Bass DA, Parce JW, Dechatelet LR, Szejda P, Seeds MC, Thomas M. Flow cytometric studies of oxidative product formation by neutrophils: A graded response to membrane stimulation. The Journal of Immunology. 1983;130(4):1910-1917. DOI: 10.4049/jimmunol.130.4.1910
  119. 119. Li M, Yin J-J, Wamer WG, Lo YM. Mechanistic characterization of titanium dioxide nanoparticle-induced toxicity using electron spin resonance. Journal of Food and Drug Analysis. 2014;22(1):76-85. DOI: 10.1016/j.jfda.2014.01.006
  120. 120. Hussain SM, Javorina AK, Schrand AM, Duhart HM, Ali SF, Schlager JJ. The interaction of manganese nanoparticles with PC-12 cells induces dopamine depletion. Toxicological Sciences. 2006;92(2):456-463. DOI: 10.1093/toxsci/kfl020
  121. 121. Monteiro-Riviere NA, Nemanich RJ, Inman AO, Wang YY, Riviere JE. Multi-walled carbon nanotube interactions with human epidermal keratinocytes. Toxicology Letters. 2005;155(3):377-384. DOI: 10.1016/j.toxlet.2004.11.004
  122. 122. Miranda RR et al. Toxicological interactions of silver nanoparticles and non-essential metals in human hepatocarcinoma cell line. Toxicology In Vitro. 2017;40:134-143. DOI: 10.1016/j.tiv.2017.01.003
  123. 123. Kononenko V et al. Comparative in vitro genotoxicity study of ZnO nanoparticles, ZnO macroparticles and ZnCl2 to MDCK kidney cells: Size matters. Toxicology In Vitro. 2017;40:256-263. DOI: 10.1016/j.tiv.2017.01.015
  124. 124. Babson AL, Babson SR. Kinetic colorimetric measurement of serum lactate dehydrogenase activity. Clinical Chemistry. 1973;19(7):766-769. DOI: 10.1093/clinchem/19.7.766
  125. 125. Niles AL, Moravec RA, Worzella TJ, Evans NJ, Riss TL. High-Throughput Screening Assays for the Assessment of Cytotoxicity, High-Throughput Screening Methods in Toxicity Testing. New Jersey, USA: John Wiley & Sons, Inc.; 2013. pp. 107-127. DOI: 10.1002/9781118538203.ch5
  126. 126. Bahadar H, Abdollahi M, Maqbool F, Baeeri M, Niaz K. Mechanistic overview of immune modulatory effects of environmental toxicants. Inflammation & Allergy-Drug Targets. 2015;13(6):382-386. DOI: 10.2174/1871528114666150529103003
  127. 127. Fernández-Pampín N et al. Toxicology assessment of manganese oxide nanomaterials with enhanced electrochemical properties using human in vitro models representing different exposure routes. Scientific Reports. 2022;12(1):20991. DOI: 10.1038/s41598-022-25483-w
  128. 128. Lama S, Merlin-Zhang O, Yang C. In vitro and In vivo models for evaluating the oral toxicity of nanomedicines. Nanomaterials (Basel). 2020;10(11):2177. DOI: 10.3390/nano10112177
  129. 129. Carnovale C, Bryant G, Shukla R, Bansal V. Identifying trends in gold nanoparticle toxicity and uptake: Size. ACS Omega. 2019;4(1):242-256. DOI: 10.1021/acsomega.8b03227
  130. 130. Adam A, Mertz D. Iron oxide@mesoporous silica Core-Shell nanoparticles as multimodal platforms for magnetic resonance imaging, magnetic hyperthermia, near-infrared light Photothermia, and drug delivery. Nanomaterials (Basel). 2023;13(8):1342. DOI: 10.3390/nano13081342
  131. 131. Maziero JS et al. Species-specific in vitro and in vivo evaluation of toxicity of silver nanoparticles stabilized with gum Arabic protein. International Journal of Nanomedicine. 2020;15:7359-7376. DOI: 10.2147/IJN.S250467
  132. 132. Mahmoudi M, Simchi A, Imani M, Milani AS, Stroeve P. An in vitro study of bare and poly(ethylene glycol)-co-fumarate-coated superparamagnetic iron oxide nanoparticles: A new toxicity identification procedure. Nanotechnology. 2009;20(22):225104. DOI: 10.1088/0957-4484/20/22/225104

Written By

Iman M. Alfagih

Submitted: 19 May 2023 Reviewed: 24 May 2023 Published: 20 June 2023