Next Article in Journal
Viewing Aggregation-Induced Emission of Metal Nanoclusters from Design Strategies to Applications
Next Article in Special Issue
Assessment of Carbon Nanotubes on Barrier Function, Ciliary Beating Frequency and Cytokine Release in In Vitro Models of the Respiratory Tract
Previous Article in Journal
Crosswise Stream of Cu-H2O Nanofluid with Micro Rotation Effects: Heat Transfer Analysis
Previous Article in Special Issue
The Effects of Titanium Dioxide Nanoparticles on Osteoblasts Mineralization: A Comparison between 2D and 3D Cell Culture Models
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The State of the Art and Challenges of In Vitro Methods for Human Hazard Assessment of Nanomaterials in the Context of Safe-by-Design

by
Nienke Ruijter
1,
Lya G. Soeteman-Hernández
1,
Marie Carrière
2,
Matthew Boyles
3,
Polly McLean
3,
Julia Catalán
4,5,
Alberto Katsumiti
6,
Joan Cabellos
7,
Camilla Delpivo
7,
Araceli Sánchez Jiménez
8,
Ana Candalija
7,
Isabel Rodríguez-Llopis
6,
Socorro Vázquez-Campos
7,
Flemming R. Cassee
1,9 and
Hedwig Braakhuis
1,*
1
National Institute for Public Health & the Environment (RIVM), 3721 MA Bilthoven, The Netherlands
2
Univ. Grenoble-Alpes, CEA, CNRS, SyMMES-CIBEST, 17 rue des Martyrs, 38000 Grenoble, France
3
Institute of Occupational Medicine (IOM), Edinburgh EH14 4AP, UK
4
Finnish Institute of Occupational Health, 00250 Helsinki, Finland
5
Department of Anatomy, Embryology and Genetics, University of Zaragoza, 50013 Zaragoza, Spain
6
GAIKER Technology Centre, Basque Research and Technology Alliance (BRTA), 48170 Zamudio, Spain
7
LEITAT Technological Center, 08225 Barcelona, Spain
8
Instituto Nacional de Seguridad y Salud en el Trabajo (INSST), 48903 Barakaldo, Spain
9
Institute for Risk Assessment Sciences (IRAS), Utrecht University, 3584 CS Utrecht, The Netherlands
*
Author to whom correspondence should be addressed.
Nanomaterials 2023, 13(3), 472; https://doi.org/10.3390/nano13030472
Submission received: 23 December 2022 / Revised: 16 January 2023 / Accepted: 18 January 2023 / Published: 24 January 2023
(This article belongs to the Special Issue Risk Assessment of Nanomaterials Toxicity)

Abstract

:
The Safe-by-Design (SbD) concept aims to facilitate the development of safer materials/products, safer production, and safer use and end-of-life by performing timely SbD interventions to reduce hazard, exposure, or both. Early hazard screening is a crucial first step in this process. In this review, for the first time, commonly used in vitro assays are evaluated for their suitability for SbD hazard testing of nanomaterials (NMs). The goal of SbD hazard testing is identifying hazard warnings in the early stages of innovation. For this purpose, assays should be simple, cost-effective, predictive, robust, and compatible. For several toxicological endpoints, there are indications that commonly used in vitro assays are able to predict hazard warnings. In addition to the evaluation of assays, this review provides insights into the effects of the choice of cell type, exposure and dispersion protocol, and the (in)accurate determination of dose delivered to cells on predictivity. Furthermore, compatibility of assays with challenging advanced materials and NMs released from nano-enabled products (NEPs) during the lifecycle is assessed, as these aspects are crucial for SbD hazard testing. To conclude, hazard screening of NMs is complex and joint efforts between innovators, scientists, and regulators are needed to further improve SbD hazard testing.

1. Introduction

The rapid expansion of the field of nanotechnology and its ever-growing number of applications has created a challenge for toxicologists and risk assessors. The continuous uncertainties surrounding nanomaterial (NM) safety, as well as the pace at which new NMs are developed, call for a more prevention-oriented strategy. The Safe-by-Design (SbD) concept is increasingly applied within the field of nanotechnology, as can be seen by the high number of EU funded nano-projects addressing SbD over the past years [1], and by its adoption in the EU Chemical Strategy for Sustainability as a strategy to meet the EU Green Deal ambitions [2,3].
SbD aims to reduce the human and environmental risk of a substance throughout its entire life cycle by minimizing or eliminating the hazard and/or by reducing exposure [4]. The concept of SbD consists of three pillars: safer materials and products, safer production, and safer use and end-of-life. For NMs, these were first described in the NanoReg2 project [5], and later in an internationally accepted working description of the OECD Safe Innovation Approach Report [6]. In practice, SbD is a two-step process: the first step is an early hazard and/or risk screening during the design phase of the innovation process of a new substance, NM, or product [7,8]. The second step is to take actions (SbD interventions) to reduce or minimize hazard, exposure, or both.
For NMs and nano-enabled products (NEPs), SbD interventions can be achieved in different ways. One option is to modify a NM in order to improve its safety profile. For example, Xia et al. (2011) showed that doping ZnO nanoparticles with iron reduces the shedding of harmful ions and reduces the toxicity of the particles upon pulmonary exposure [9]. Another example of a SbD intervention is applying a surface treatment to minimise NM biological reactivity, as has been successfully achieved for nano-SiO2 by adding silanol groups to the silica surface [10]. Reducing exposure is also a fundamental part of SbD and can be achieved by implementing procedural changes such as working in closed systems or using wet synthesis methods [5]. Reduced release and therefore minimized exposure can also be achieved by altering the design of the NEP, for example by improving the immobilization between the NM and the matrix, as was conducted for silver NMs onto cotton fabrics [11].
The above-mentioned examples can only be achieved after first assessing hazard and risk early in the innovation process, and then using this knowledge to integrate safety into the design of the NM, NEP, or production process. For many NMs, and especially for novel ones, hazards are largely unknown [12], and cannot be predicted only based on physicochemical (PC) characterisation. Therefore, carrying out suitable hazard testing at the early stages of product development is of utmost importance for SbD applicability. SbD hazard testing aims to identify hazard warnings in the early stages of the innovation process using simple in vitro methods. Once a product is designed and produced, the manufacturer should comply to the regulations and perform hazard and risk assessment accordingly.
Many strategies and frameworks for hazard assessment of NMs in the context of SbD have been proposed in recent years [13,14,15,16,17,18], some proposing specific in vitro assays, and some based only on a selection of toxicological endpoints to consider. However, no comprehensive investigation of the suitability of currently available in vitro assays for such strategies has been conducted thus far.
From previous studies on the mechanisms of action of NMs, it is known that transformation (e.g., dissolution), reactivity, inflammation, cytotoxicity, and genotoxicity are among the most important parameters and endpoints to evaluate when assessing the hazard of a NM, and therefore these are suggested to be measured in many available strategies and frameworks [13,19,20,21]. Selecting in vitro tests suitable for SbD hazard testing is not trivial. Only a few OECD test guidelines and ISO standards are available specifically for testing NMs. Due to the interfering behaviour of NMs with their surrounding environment and with the assay readout, routinely used toxicity assays (i.e., those used to test soluble chemicals) may prove unsuitable or may require optimizations and inclusion of extra controls [22]. In contrast with hazard assessment for soluble chemicals, NM testing requires additional steps, such as dispersion protocols and determining the dose delivered to the cells in submerged cell culture experiments [23]. Specifically for the purpose of SbD hazard testing, since it is performed early in the development of a NM/NEP, assays will not only have to be compatible with the NM to be tested, but should preferably also be fast, cost-effective, and able to correctly indicate hazard warnings.
This work provides a practical and critical evaluation of the suitability of most frequently used in vitro toxicity assays and the challenges for their use in NM SbD hazard testing. For this purpose, criteria for the suitability of methods for application in a SbD hazard testing strategy are established, leading to an evaluation of the methods currently in use for the parameters and endpoints identified as important for the mechanisms of action of NMs. This work is conducted under the umbrella of the Horizon2020 project SAbyNA which aims to develop a user-friendly platform for industry with optimal workflows to support the development of SbD NMs and NEPs. For this purpose, existing resources, such as in vitro assays are identified, distilled, and streamlined. This state of the art and evaluation of in vitro assays for SbD applicability can be used as an outlook for innovators, regulators, industry, and scientists of how early hazard testing of NMs and NEPs can be put into practice to eventually contribute to the design of SbD NEPs.

2. Criteria

A set of performance criteria is proposed to evaluate the suitability of in vitro methods for SbD hazard testing. The criteria were adapted from the widely used Good In Vitro Method Practices (GIVIMP) [24] and tailored to suit SbD hazard testing for NMs specifically. Figure 1 shows several key considerations for assay selection for SbD hazard testing.
Predictive: The first criterium is that an in vitro assay should be sufficiently predictive of the in vivo situation. This comparison is preferably made with human data, or alternatively using animal data. SbD hazard testing is carried out in an early stage of product development and is considered a first screening. The aim of SbD hazard testing is to detect early hazard warnings and not to derive a point of departure for risk assessment. Therefore, assays are sufficiently predictive for SbD hazard testing when they are able to indicate hazard warnings. Assays that are able to accurately rank NMs/NEPs based on their hazard potency are of extra value for SbD hazard testing, as this will allow comparison of candidate NMs and comparison with benchmark NMs.
Predictivity can be assessed by looking into the prediction accuracy of the assay. An assay’s accuracy to predict in vivo effects is a combination of its sensitivity and specificity. Sensitivity is the ability of the assay to detect true positives and specificity is the ability of the assay to detect true negatives.
Simple and cost effective: Simplicity and cost effectiveness are key for SbD hazard testing since these assays are to be performed in an early stage of NM/NEP product development. Ideally, an assay should be easy to perform, time-efficient and cost-effective.
Robust: An assay should give consistent and repeatable results between experimental repetitions and between different labs.
Compatible: An assay should preferably be compatible with a wide range of NMs, or at least its compatibility domain should be identified. Assays with optimized protocols specifically for NMs are preferred.
Readiness: Methods that are considered ‘ready to use’ and already standardized or (pre-)validated for NMs are prioritized.

3. Challenges of Testing NMs In Vitro for SbD Applicability

NMs are particulate matter, making NM in vitro testing by default more challenging than testing soluble chemicals. Several additional aspects need considering when testing NMs in vitro, including determining the behaviour of the NM in exposure medium, selecting a dispersion protocol to create stable suspensions which preferably mimic human exposure as much as possible, and assessing the potential interference of the NM with the assay components or optical readouts. Furthermore, elaborate characterization of the NM is required [25], but this will not be discussed further in this review. The fact that SbD hazard testing needs to be as simple as possible creates an important predicament that needs addressing. An overview of key aspects that should be taken into account when performing SbD hazard testing of NMs is shown in Figure 2. The most important aspects are discussed below.

3.1. Choice of Dispersion Protocol

Classically, in vitro toxicity evaluation is performed in cultured cells maintained in submerged conditions. To ensure reproducible and controlled exposure from one replicate to another, stable suspensions of well-dispersed NMs are prepared, sometimes requiring energy input to disrupt particle agglomerates. For SbD hazard testing, a prerequisite for the suitability of a dispersion procedure is that the SbD properties (e.g., coatings or surface treatments) of the dispersed NM/NEP are preserved, and that the resulting dispersed NM/NEP is relevant for human exposure in terms of size and other physicochemical properties.
The most commonly used dispersion procedure for NMs is via sonication, using either an ultrasonic bath, a probe, or a cup-type sonicator [26,27]. For SbD hazard testing, sonication would not be the preferred option in some specific cases. For instance, sonication can break multi-walled carbon nanotubes (MWCNT), causing a reduction of their length [27], and therefore leading to different toxicity profiles than the MWCNT that humans would be exposed to. Sonication has been used to produce MWCNTs with different lengths from a same initial batch of MWCNTs [28,29], and Hadrup et al. (2021) concluded that the length of the MWCNT is a major determinant of its toxicity [29]. Therefore, when assessing hazard properties of CNTs in vitro, sonication should be limited as much as possible, and in case sonication is used, NM physicochemical properties should be verified to ensure they still maintain exposure-relevant characteristics.
Another example where sonication would have to be carefully considered is when testing specific synthetic amorphous silicas, which are in some cases intentionally produced as agglomerates. Sonication can disrupt most of the agglomerates, reducing the overall hydrodynamic diameter, which constitutes a substantial modification of the initial material [30]. In an inhalation exposure scenario, a person would be exposed to these agglomerates, and therefore sonication would not be the preferred option. However, there are indications that these agglomerates disintegrate in the intestine [31]. In the case of ingestion, the gut epithelium is exposed to nano-sized silica, and sonication would result in an exposure-relevant material.
Dissolution is a major determinant of the toxicity of some NMs (e.g., release of toxic metal ions such as silver or copper ions) and decreasing the NM dissolution potential can be considered a SbD intervention. Sonication has been shown to enhance the dissolution of some metallic NMs, such as Cu, Mn, and Co [32], and the dissolution can be further increased when proteins are present in the solution during sonication [33]. Thus, dispersion procedures that involve sonication of NMs, especially in a medium that contains proteins, such as the procedure optimized within the Nanogenotox project [34], should be carefully considered in view of exposure scenarios when testing NMs that can potentially dissolve.
Some NMs are designed as core-shell structures (e.g., quantum dots (QDs)) where the shell reduces dissolution and leaching of potentially toxic elements from the core. The design of more robust shells, used as a SbD intervention, reduces the QDs dissolution rate and thereby their toxicity [35,36,37]. However, the core-shell boundary is a region of fragility and sonication could promote shell fragmentation and the release of core contents. Thus, sonication could result in a reduced effect of the SbD intervention, potentially resulting in an overestimation of toxicity in SbD hazard testing. Therefore, for core-shell NMs, sonication should not be recommended, unless humans are exposed to fragmented QDs.
Coating NMs with surface ligands or grafting them on an inert matrix such as cellulose has also been tested as a SbD intervention to produce safe(r) photocatalytic paints containing TiO2 NMs. Coating TiO2 NMs with polyethylene glycol (PEG), poly(acrylic acid) (PAA), or 3,4-dihydroxy-L-phenylalanine (DOPA) increased the stability of the doped paint and its resistance to weathering and abrasion, while their grafting on cellulose fibres enhanced their photocatalytic properties, thereby allowing for the reduction of the amount of NMs necessary to reach efficient photocatalysis [38]. Again, sonicating these surface-coated TiO2 NMs or TiO2-containing composites might lead to the reduction of the effect of SbD interventions. In addition to that, extensive sonication has been shown to alter the zeta potential of TiO2 and CeO2 NMs [39,40] and to cause re-agglomeration of Cu or Mn NMs [33,41]. It should in each case be investigated what the exposure-relevant form of the NM or NEP is.
Moreover, samples could be contaminated by the release of Al and Ti from the sonication probe upon extensive sonication, potentially leading to toxicity [30,42]. Finally, extensive sonication of NMs in a growth medium containing proteins or in water with bovine serum albumin (BSA) (as in the procedure optimized within the Nanogenotox project [34]) could promote the degradation of proteins, leading to the formation of large aggregates of degraded proteins [43].
To conclude, for SbD hazard testing sonication should be carefully considered, and exposure relevancy should always be kept in mind. If exposure-relevant and stable dispersions in the exposure medium are obtained using simple methods such as vortexing, dispersion via sonication might not be needed. In the case of NMs that quickly agglomerate and form large clumps, more controlled sonication methods might be appropriate. For example, a protocol using minimum material-specific energy to reach a stable dispersion as described by DeLoid et al. (2017) could be used [23]. NMs should subsequently be characterized to ensure that no PC changes were produced that deviate from the exposure-relevant material. The PC properties of the NM tested should reflect the exposure conditions, whether it be the pristine NM with SbD interventions or the agglomerated NM. However, it has to be noted that unstable suspensions could lead to difficulties with reproducibility and/or interferences with the assay readout.
Finally, it might be recommended to also perform in vitro assays after extensive sonication, as this might be required for regulatory risk assessment. By doing this, the first steps towards compiling a dossier for regulatory compliance are made, and this might already indicate if any issues can be foreseen for market entry. Additionally, extensive sonication may provide a worst-case scenario in in vitro assays, which could fit in a precautionary approach. OECD guidance on sample preparation [44] is currently being revised to include considerations for the choice of a specific dispersion protocol rather than applying extensive sonication by default.
For SbD hazard testing sonication should be carefully considered, and exposure relevancy should always be kept in mind. If exposure-relevant and stable dispersions in the exposure medium are obtained using simple methods such as vortexing, dispersion via sonication might not be needed. In the case of NMs that quickly agglomerate and form large clumps, more controlled sonication methods might be appropriate. NMs should subsequently be characterized to ensure that no PC changes were produced that deviate from the exposure-relevant material.

3.2. Influence of Medium Components

Supplementation of cell-culture medium with serum (i.e., foetal calf serum (FCS) and foetal bovine serum (FBS)) is common practice in cell culture procedures as it is required for cell growth and maintenance. When exposing the NMs in a test medium, constituents of the medium including proteins, amino acids, lipids, and sugars adsorb on the surface of NMs, leading to the formation of the so-called biomolecular corona [45]. This corona is highly dynamic and may change upon changing the composition of the test medium [46,47]. This dense layer of biological molecules can modify NM toxicity in several ways. Firstly, it could do so by masking the surface reactive sites of the NM [48]. Secondly, serum may stabilize the NM dispersion, leading to a lower dose delivered to the cells in in vitro assays, as has been shown for TiO2 NMs for example [49]. Lastly, a biomolecular corona may reduce NM surface energy, and thereby its cellular uptake via adhesion-induced endocytosis, as has been shown for SiO2 NMs [50,51].
These effects are clear when comparing the toxicity of NMs tested with and without serum. For instance, the cytotoxic potency of polystyrene NMs was found to decrease 2-fold when the exposure medium contained serum [52]. Similarly, the cytotoxicity of SiO2 NMs decreased up to 92%, and pro-inflammatory response decreased up to 87% when cells were exposed in medium with serum [53]. In addition, the species of origin of the serum could lead to different responses [54]. Addition of bovine serum albumin (BSA), a protein often used to help stabilize dispersions, has also been reported to reduce cytotoxicity [55,56].
In short, the addition of serum and BSA to the exposure medium may lead to lower toxicity in in vitro assays. Which approach is most suitable for SbD hazard testing should be explored further. Since SbD hazard testing is mostly focused on detecting hazard warnings, it could be argued that testing without serum is more appropriate, as it ensures a higher sensitivity. Additionally, when testing serum-free, a worst-case scenario could be mimicked without the protective effect of serum on NM-cell interaction. On the other hand, testing with serum is the more realistic approach as humans are rarely exposed to NMs without a biomolecular corona. Eventually, the route of (potential) human exposure should be taken into account when selecting an exposure protocol, as systemically injected NMs will immediately be covered by serum proteins, whereas inhaled NMs will come in contact with epithelial lining fluids, which contains a different set of biomolecules. For SbD hazard testing, exposure relevancy is important, and a biocorona could be applied which corresponds to the route of exposure, such as lung-lining fluid for pulmonary exposure. In the context of exposure relevance, and in the context of the 3Rs (replacement, reduction, and refining), the use of human serum or serum-free alternatives may be favoured over FBS.

3.3. Determining Dose Delivered to Cells

A particular challenge when testing NMs in vitro in adherent, submerged cell cultures is determining the delivered dose, i.e., the amount of material that reaches the cells. Settling of NMs depends on their density, size, and the properties of the cell-culture medium, as well as on their agglomeration state [57]. The latter is again influenced by the dispersion method used [58].
Determining the delivered dose in an in vitro experiment is an absolute requirement, even when performing a simple hazard screening for the purpose of SbD, and when performing high throughput screening (HTS) experiments. This is because the administered dose can differ substantially from the delivered dose that reaches the cells. For example, for particles that settle rapidly, the difference between administering 100 µL per well or 200 µL per well of the same concentration will mean a doubling of the amount of material per well, and thus a potential doubling of the delivered dose. Moreover, since sonication influences the agglomeration state, and the agglomeration state influences the settling rate and thus the dose delivered to cells, determining the delivered dose may help the comparison of data among independent experiments using different dispersion protocols. A visual representation of two example NMs with different settling rates is shown in Figure 3. For SbD hazard testing specifically, determination of delivered dose aids in a comparison to benchmark materials with known toxicity, as settling may differ greatly between NMs. The importance of determining the delivered dose was shown in a study by Pal et al. (2015), where a correction for the delivered dose led to a considerable change in the hazard ranking of a panel of NMs, after which the in vitro outcomes matched better with the in vivo results [59].
The delivered dose can be modelled using the DG [60] or ISD3 model [61], which are currently available in the DosiGUI software generated in the PATROLS project [62]. The DosiGUI is user friendly, however, these models also introduce uncertainty as they do not take into account some critical factors such as particle convection [63], or dispersion-stabilizing surface functionalization. Moreover, cell stickiness needs to be chosen from an arbitrary scale, which is often an unknown parameter that has a big effect on the modelled delivered dose [64].
These models require the effective density of the NM as input, which is the density of the NM in a dispersion. In the case of agglomerates, this includes the density of the medium trapped inside the agglomerate. The effective density can be measured using analytical ultracentrifugation (AUC) or using the volumetric centrifugation method (VCM). In order to adhere to the criteria of SbD hazard testing, the VCM is preferred over AUC, as it is easier, less costly, and does not require specialized equipment [65]. It is important to measure model input parameters precisely, as small differences in input as a result of instrument variation may lead to large differences in the modelled deposited dose [64]. Stable dispersions are a requirement for modelling the dose rate and final dose delivered to the cells, as the calculations are based on a one-size distribution. The accuracy of the model outcome—and thus the estimated deposited dose (rate)—is less accurate if the size distribution of the dispersion changes over time due to agglomeration or aggregation.
The need to determine the delivered dose adds an extra step to SbD hazard testing, leading to a reduction of the achievable simplicity. Determining the delivered dose is however a requirement, even for SbD hazard testing, as more precise dosimetry will allow for more informative hazard testing. This, however, only applies to submerged testing, and not to experiments in which, for instance, an air-liquid interface (ALI) exposure protocol is followed. For ALI exposures, a quartz crystal microbalance (QCM) may provide sensitive and accurate deposited-dose measurements [66].

3.4. SbD Hazard Testing of NEPs and NMs Released during the Life Cycle

One of the most important aspects of SbD is assessing the safety of a product along its entire life cycle (LC) [5]. Usually, only pristine NMs are included in toxicity assays. This could be insufficient, as humans are also exposed to NEPs, aged NM and/or NMs released during the product LC including production, use, and end-of-life. The physicochemical characteristics of the NMs released to the environment along the different stages of the LC can be very different in terms of shape, chemical composition, agglomeration state, and surface modification [67,68,69,70,71]. Moreover, NEPs and NMs released during the LC might pose a different hazard than pristine NMs [72,73,74,75]. Thus, gathering information on the characteristics and hazard of NEPs and NMs released during the LC is important for designing relevant SbD interventions.
Processes leading to the release of NMs from a NEP during the entire LC can be simulated under laboratory conditions, after which NMs can be collected (e.g., by using filters) and redispersed in liquid for toxicity testing [76] or collected in liquid suspensions directly [77]. Realistic, released NMs, relevant for consumer exposure during the use of NEPs, can be obtained by using standardized methods (e.g., abrasion and weathering) that are normally used to test the durability and performance of NEPs. In the case of abrasion processes, there are different instruments that can be used to simulate mild or hard abrasion. Experimental parameters (e.g., cycles of abrasion, abrasion materials, normal load at the top of the abrader, etc.) can be tuned to reflect closely the NEP use conditions.
Aging experiments simulate conditions to which a product could be exposed during its use phase and are usually performed in a weathering chamber under accelerated conditions of UV exposure and rain. The weathering conditions (e.g., duration of cycles of light and rain, duration of the experiment, etc.) can be selected to follow international standards or be customized. In order to obtain higher quantities of released material (worst-case scenario), NEPs can be fragmented and sieved [78].
Released aerosols can be size-separated by using e.g., a cascade impactor to ensure inhalable or respirable fractions of NMs; After which they are collected on filters [79]. Efforts should be made to ensure high extraction efficiency and minimal compositional alterations when extracting material from filters [80]. Another option which is less easy but mimics better a real-life exposure is the direct exposure of cells to the released material, as performed by Zarcone and colleagues [81] for diesel exhaust. This approach is however somewhat more labour-intensive for SbD hazard testing but might be useful for gaining a more fundamental insight into the toxicity of released materials (exposure-relevant material) without losing a fraction to filter extraction.
After obtaining and extracting the material from filters, the same actions should be taken as for pristine NM testing, such as an accurate dose determination, controlling for interference, endotoxin contamination testing, choosing an appropriate dispersion protocol, etc. For NEPs and NMs released during the LC, endotoxin contamination might pose an extra challenge as these materials are generally not produced in sterile environments. Finally, compatibility in submerged settings might pose additional challenges as a NEP matrix is often plastic-based and might float on culture medium.

Feasibility and Relevance

Obtaining sufficient amounts of NM that are released at a given stage of the LC can be challenging due to low emission rates, contamination with other substances (e.g., sanding material), as well as laboursome and time-consuming procedures. The question is whether performing SbD hazard testing on pristine NMs is sufficiently relevant when assessing the safety of a NM along its entire life cycle. There are some examples in literature in which both pristine and released NMs have been tested to assess and compare their hazards. In most cases, materials released during the LC induced less or equal toxicity as compared to the pristine NM, as has been shown in in vivo studies [75,82,83] as well as in vitro [84]. This means that testing pristine materials, albeit often far from representing the reality, can still represent a worst-case scenario. In this case, risk screening of NMs released from a NEP can be mainly based on emission rates combined with the hazard information of the pristine NM. However, it should be noted that there is very little known about the toxicity of released NMs as compared to pristine NMs, and the exception may prove the rule.
Similarly, testing pristine NMs may represent a worst-case scenario for aged NMs. For example, freshly ground quartz particles have been shown to induce higher levels of pulmonary inflammation and cytotoxicity as compared to aged quartz [85].
For now, it should be considered on a case-by-case basis whether testing forms other than the pristine NM is required. More research is needed to determine whether the use of the pristine NM in SbD hazard testing is sufficient due to its ‘worst-case’ nature, or whether testing aged NMs, and NMs released during LC is crucial for designing SbD interventions.
It should be considered on a case-by-case basis whether testing forms other than the pristine NM is required. More research is needed to determine whether the use of the pristine NM in SbD hazard testing is sufficient due to its ‘worst-case’ nature, or whether testing aged NMs, and NMs released during LC is crucial for designing SbD interventions.

3.5. Challenging NMs and Advanced Materials

Hydrophobic NMs, NMs with low material density, multi-component NMs, and other advanced materials yet to be invented may show poor compatibility with commonly used in vitro assays. Applying a single standardized exposure method to all types of NMs will inherently give biased outcomes. For SbD hazard testing it is therefore important to consider the compatibility of NMs with challenging physicochemical properties and to be prepared for future novel advanced materials.

3.5.1. Hydrophobic Particles

Since cells are always cultured in aqueous culture medium, hydrophobic particles can be of extra difficulty to test. Carbon nanotubes (CNTs) and graphene-based particles, for example, are notorious for being difficult to disperse in culture medium. Ethanol pre-wetting, using different dispersion media [86], and adjusting sonication time and frequency have been shown to improve dispersibility of NMs. However, for some NMs a stable dispersion can never be achieved in cell-culture medium. For example, some CNTs are specifically designed to agglomerate in order to reduce their dustiness and thereby improve their safety. For cases such as these, dry exposures at the air-liquid interface (ALI) should be considered when focussing on potentially respirable NMs. This requires a cell type that can be cultured on membranes in medium on the basal side, while being exposed to the air on the apical side. For the generation of a dry (dust) aerosol for ALI exposure, several methods are available [87]. However, it should be kept in mind that if a dust cannot be generated in a laboratory setting, inhalation of the NM is very unlikely. Thus, in these cases the relevance of an inhalation study should be reconsidered.

3.5.2. Buoyant NMs

NMs with a density lower than cell-culture medium (e.g., certain types of plastic particles or agglomerates with a low effective density) will float and do not settle over time, resulting in no contact with adhering cells in a classical, submerged in vitro setup. This will likely lead to an underestimation of the potency of NMs such as nano-plastics and liposomes [88]. A solution to solve the problem of buoyant particles is to perform an inverted ‘overhead’ cell culture, where the cells are not cultured on the bottom of a culture dish, but upside down on top of the exposure medium. With this approach, it was possible to produce a dose response for several floating particles, whereas the traditional approach did not show any results [88,89]. For buoyant NMs, where inhalation is the relevant exposure route, ALI exposures can also be an option.

3.5.3. Multicomponent NMs and Other Advanced Materials

In the past years, the more complex multicomponent nanomaterials (MCNM) have gained popularity. These next generation NMs consist of two or more materials or substances, giving rise to properties (e.g., reactivity) that are not equal to the sum of the properties of each component [90]. There are still many knowledge gaps when it comes to the toxicity of these and other novel advanced materials, which is why the concept of SbD is a suitable prevention-oriented approach. Whether these materials are compatible with the available toxicity assays is unknown, and this might pose challenges for future SbD hazard testing. Theoretically, MCNMs could exhibit multiple types of assay interference, attributed to the individual components of the MCNM. It is important to be aware of these challenges and to always assess interference.

4. Evaluation of In Vitro Methods for SbD Hazard Testing

4.1. Cytotoxicity

Measuring cell viability or cytotoxicity is a fundamental part of most hazard assessment strategies and integrated approaches to testing and assessment (IATA’s) for several reasons. Firstly, cytotoxic potency (for example LC50) gives an indication of the relative hazard of a NM. Secondly, cytotoxicity assays allow for the selection of appropriate sub-lethal doses for further mechanistic testing (e.g., genotoxicity and inflammation). Lastly, for several mechanistic assays such as genotoxicity assays, cytotoxicity measurements are a requirement for the correct interpretation of the results. Cell viability can be determined by the measurement of various cellular parameters, such as mitochondrial activity, lysosomal integrity, and membrane integrity. Different endpoints should be included to assess cytotoxicity [91,92,93], as results from different assays do not always correspond [94].

4.1.1. Most Frequently Used Assays, Strengths and Limitations

The most-used approaches for measuring cytotoxicity or cell viability in vitro include measuring mitochondrial activity (examples are MTT, MTS, XTT, and WST-1 assays), release of cytoplasm components (examples are LDH and AK), lysosomal integrity (Neutral red uptake), apoptosis markers (caspase 3/7), and stains that can specifically enter apoptotic and/or necrotic cells (Trypan blue, Propidium iodide, and Annexin V). Propidium iodide and Annexin V can be combined to determine plasma membrane restructuring which can be representative of either necrosis or apoptosis specifically. Most cytotoxicity assays are relatively simple, can be carried out in a 96-well microplate format, could be used for HTS, and have commercial kits available. An exception are the assays that require microscopic evaluation of a certain staining, as this is more labour-intensive.
For many cytotoxicity and viability assays, NMs can interfere with assay reagents and/or the optical readout [95,96,97,98]. Therefore, potential interference of the NMs with assays should always be assessed [92,99]. The elimination of NMs via high-speed centrifugation may reduce optical interference [92]. Alternatively, since mitochondrial activity is measured intracellularly, NMs can be washed away from cells prior to incubation with the reagent to avoid interaction of the NMs with the reagent [100]. For products measured in the supernatant (e.g., LDH and AK), washing is not feasible, but centrifugation can help remove larger NMs and thereby reduce optical interference. However, some NMs are known to inactivate or adsorb LDH directly [101]. If interference still occurs after taking precautions, it is advised to perform another type of cytotoxicity test, as the subtraction of the average background signal of the NMs will reduce the accuracy of the outcome [97,102,103]. For specific NMs, some assays might prove not to be compatible.
Much effort has been put into the optimization and standardization of in vitro cytotoxicity assays specifically for NMs in the past years. An ISO standard for the MTS assay was published in 2018 [104]. In 2021, an ISO standard was published for impedance measurements for NMs specifically [105]. This assay involves growing cells on an electrode during exposure to the NM. The detachment of cells, indicating cytotoxicity, is measured as a decrease in electrochemical impedance, as demonstrated in the assessment of poly-lactic acid NM-induced toxicity in A549 epithelial cells [106]. This assay is less prone to interferences as no optical readout and no assay reagents are required. However, it does require specialized equipment not available in many labs. Internationally standardized and harmonized standard operating procedures (SOPs) for other cytotoxicity assays have not been published to date.
In the NanoReg project, an interlaboratory study for the MTS assay was carried out, and acceptable robustness levels were found depending on the cell type. The human alveolar cell-line A549 showed a good agreement in cytotoxicity between labs, whereas the differentiated human monocyte cell-line THP-1 (dTHP-1) showed varying results and a poor robustness [107]. In a large interlaboratory study by Piret et al. (2017), a good robustness was found for the MTS assay and ATP content measurements. These comparisons were carried out using both A549 as well as dTHP-1 cell lines, and two different NMs. The authors stressed the importance of avoiding interference of the NM with the assay in order to obtain more reliable results, and a lower inter-laboratory variability. They also found that the caspase-3/7 assay showed a high inter-laboratory variability [100].
A large interlaboratory study of eight labs studied how to improve the robustness of the LDH and MTS assay. After a first round of experiments, adaptations to the protocols were made and robustness increased significantly within and between laboratories. Changes made to the protocols included the optimization of the differentiation of THP-1 cells and centrifugation after incubation with MTS reagent to remove NMs [108]. These findings on the MTS assay were confirmed in another interlaboratory study using the A549 cell line. Additional sources of variability were identified in this study. A549 cells from two different suppliers showed a large difference in cytotoxicity in response to polystyrene NMs. Also, the inclusion of serum effectuated large differences in cytotoxicity as compared with serum-free experiments. Moreover, differences in pipetting techniques (e.g., harsh aspiration vs. gentle pipetting and completely removing medium vs. partially removing medium before MTS incubation) and dispersion protocols were identified as causing differences in results between laboratories [52]. The importance of more elaborate and detailed SOPs was again stressed in a recent inter-laboratory study, where the inclusion of several acceptance criteria was found to improve the robustness of the MTS assay, such as maximum acceptable variations between replicates, minimum cell survival, and maximum interference levels [102].

4.1.2. Predictivity and Relevance

Whether in vitro cytotoxicity assays are predictive of in vivo acute toxicity has been studied for years for soluble chemicals. For NMs, however, there are only a few studies that correlate in vitro cytotoxicity with in vivo toxicity. Therefore, in this section we have included not only studies that correlate in vitro cytotoxicity with in vivo markers of cell death (apoptosis, necrosis etc.), but also with any type of in vivo toxicity.
In general, the predictivity of cytotoxicity assays depends on the mechanism of action of the NM, as well as on the cell type used for the in vitro study [109,110,111]. NMs which exert their effect through the shedding of toxic ions are usually also cytotoxic in vitro [100]. In a comprehensive comparison study, in vitro cytotoxicity was compared to in vivo lung inflammation for several different particles, using comparable doses. LDH release and trypan blue exclusion assays were able to predict the inflammation-inducing effects of ion-shedding NMs, but not of poorly soluble NMs [112]. However, in another study, in vitro LDH release in response to poorly soluble TiO2 NMs correlated well to the in vivo number of polymorphonuclear cells (PMN) in BALF. This correlation was only present when the dose was expressed as surface area, and not when using mass as dose metric [113].
The toxic effects of CNTs in vivo upon inhalation are not easily predicted using in vitro cytotoxicity assays, unless the toxicity is caused by metal impurities [109]. Also for CNTs, the in vitro effects differ between cell types [114]. Other carbon-based materials, such as diesel exhaust also do not show an accurate correlation of cytotoxic response with in vivo effects. LDH release from A549 cells and LDH measured in BALF from rats upon instillation with diesel exhaust did not correspond, and even showed an opposite ranking in toxicity [115]. However, the suspensions used were not purely the particle fraction and contained other substances such as lube oil (which floats on culture medium), possibly causing the contrasting rankings.
The choice of cell type is crucial for performing a predictive in vitro cytotoxicity assay. For example, the WST-1 and NRU assays were able to establish an accurate ranking in toxicity of Ag, Au, SiO2, and MWCNTs, but IC50 values differed between the cell types used [114]. In a study by Sayes and colleagues, in vitro LDH release did not correlate with rat pulmonary LDH release and inflammation (% PMNs) for rat primary pulmonary macrophages and rat pulmonary epithelial L2 cells grown in mono-cultures. However, when grown in co-culture, in vivo LDH release and inflammation were accurately predicted via the in vitro LDH release for crystalline silica and ZnO (but not for amorphous silica) [94]. A similar study also showed a good correlation for this co-culture model for ZnO NM, but only at the highest (particle overload) dose [116].
When choosing a cell line, immune cells are found to give a higher prediction accuracy than fibroblasts [114]. When macrophages are thought to be involved in the toxicity of NM, it is especially important to select an immune cell type for testing cytotoxicity. THP-1 cells which were differentiated to macrophages (dTHP-1) showed a higher sensitivity for cytotoxic effects as compared to A549 cells (alveolar cell line) for a panel of 24 NMs [117]. Cho and colleagues found that differentiated peripheral blood mononuclear cells and isolated lung macrophages performed better compared to cell lines such as dTHP-1, A549, and 16-HBE [112]. The fact that primary cells are more sensitive than cell lines is generally accepted. Despite this, primary cells are more difficult to work with and more expensive, and will therefore most likely be disfavoured for SbD hazard testing.

4.1.3. Overview of Needs and Knowledge Gaps

Table 1 shows a summary of how the different cytotoxicity assays perform in terms of the criteria for SbD hazard testing. As cytotoxicity measurements are a requirement for several mechanistic assays, they are crucial for SbD hazard testing. Simple cytotoxicity assays—although optimizations for NMs are needed—serve as a good starting point for detecting hazard warnings in SbD hazard testing. It is recommended to include at least two different cytotoxicity assays as different assays measure different mechanisms [92,118]. A combination of a mitochondrial activity assay and a membrane-integrity assay is recommended.
Prediction accuracy of cytotoxicity assays should be investigated further. It was shown that predictivity depends on the cell type used and the mode of action (MOA) of the NM. Assay applicability domains should be mapped in more detail to understand which toxic effects can be predicted with in vitro cytotoxicity assays and which cannot. We also found that protocol optimization improves assay robustness. Moreover, interferences are quite common for cytotoxicity assays, and they can be avoided by taking the right precautions and including the right controls, which is crucial even when performing SbD hazard testing. Together, this indicates the need for optimised and standardized protocols for NMs specifically. This will in turn also aid the determination of the prediction accuracy of assays.
As cytotoxicity measurements are a requirement for several mechanistic assays, they are crucial for SbD hazard testing. Simple cytotoxicity assays—although optimizations for NMs are needed—serve as a good starting point for detecting hazard warnings in SbD hazard testing. It is recommended to include at least two different cytotoxicity assays as different assays measure different mechanisms A combination of a mitochondrial activity assay and a membrane integrity assay is recommended.

4.2. Dissolution

Although in vitro testing of dissolution is a measure of a PC property, and not directly a measure of toxicity, the results obtained can be used to infer potential toxicity, or even potential pathogenicity. This is through consideration of a material’s biodurability or its transformation to ions or molecules. In this context, biodurability may be accompanied with biopersistence, which historically has been linked to the fibre pathogenicity paradigm such as that relating to asbestos, CNTs, and other respirable fibres [119], but also in relation to poorly soluble particles such as TiO2. Long-term inhalation exposure to poorly soluble particles can induce impaired clearance and chronic inflammation that might even progress to cancer, as has been observed for TiO2 in rats [120]. The human relevance of these results is a topic currently receiving a resurgence in interest within the scientific community [121]. Conversely, rapid dissolution of a substance can indicate exposure to potentially harmful soluble components, such as metal ions, which can be released in body compartments that are otherwise inaccessible.
Information on dissolution in relevant conditions is greatly beneficial for the hazard assessment of NMs and in defining SbD interventions. Information on dissolution is already a requirement of REACH and EFSA [21] and dissolution rates are a valuable criterion within all of the current risk assessment tools available for NM hazard assessment and can also be used for grouping/read-across [14,16,18].

4.2.1. Most Frequently Used Assays, Strengths and Limitations

There are various methods used in in vitro testing of dissolution, acellular and cellular, which have not changed significantly for some time. There are a number of guidance documents including ones from ISO (ISO 19057:2017) and OECD (OECD GD No. 318, specifically for environmental studies) which provide the start of standardization of these techniques. The output of various EU projects (e.g., GRACIOUS, Gov4Nano and BIORIMA) will also greatly impact the development of this methodology.
For acellular testing, it is possible to test within static systems [122] or flow-through (dynamic) systems [123,124]. The application of these methods is extremely diverse, as the formulation of different simulant fluids may facilitate the simulation of any biological compartment, including extracellular and intracellular compartments, and any exposure route of interest including oral, dermal, or inhalation [125], with recommendations made within an ISO technical report (ISO 19057:2017). There are a number of differences, both subtle and substantial, in the simulant fluids used that determine the accuracy of in vivo prediction. For example, components such as citric acid have a significant effect on the dissolution of certain metals, and inclusion of proteins/serums will also have an effect on dissolution [126]. These considerations have been recently reviewed [127]. Two recently completed projects (nanoGRAVUR and GRACIOUS) identify the abiotic flow-through system ISO/TR 19057:2017 as the most relevant system [124,128], with a technology readiness level (TRL) identified as high/medium for metals using Inductively Coupled Plasma Mass Spectroscopy (ICP-MS) analysis and medium/low for materials such as CNTs that require techniques such as Transmission Electron Microscopy (TEM) or X-Ray Photoelectron Spectroscopy (XPS).
Although there are currently no accepted methods for in vitro cellular dissolution testing, various studies have been conducted and these may be more reflective of the in vivo response following inhalation of particles, although there have been concerns raised with the cellular methods.

Acellular Methods

Acellular dissolution can certainly be considered simple, considering the practical requirements. However, each methodology has different demands and associated limitations. For example, the solutes released during dissolution within a static system, especially those of a basic nature, may cause enhanced nucleation, precipitation, changes in localised pH, and/or saturation effects preventing further dissolution [129,130,131,132]; this behaviour is unlikely to reflect the in vivo behaviour, demonstrating clear limitations of static systems. Although dynamic systems, by design, circumvent these issues, they are not without limitations, and there are a number of factors which may affect their reproducibility [132]. NMs may pass through filters used in flow-through systems, leading to misinterpretation of results and potential false-positive results [133], or filters may become blocked and ruptured by components of the more complex fluids such as proteins or lipids [134]. Practically, dynamic systems are cumbersome due to the high volume of liquids required in long-running tests [135]. Although a comparison of acellular methods is not often made, when done so the findings have been confounding. For example, the dissolution rate of gold nanoparticles has been found similar in static and dynamic systems [136], while the solubility of BaSO4 has been shown to differ in static and dynamic systems [137].
It is often reported that distinction between different material forms is possible, with a level of sensitivity allowing for a distinction between dissolution rates leading to grouping, as has been suggested for fibrous NMs within the GRACIOUS project [138]. This approach has been aligned with previously established methodology for man-made vitreous fibres (MMVF) and asbestos, whereby NMs biodurability can be defined by the respective dissolution in alveolar fluid and lysosomal fluid. Good comparability has already been found for in vitro dissolution of MMVF materials and in vivo biopersistence [139], allowing confidence in this approach. Heavy influencers of sensitivity include the analytical method used to detect released ions, as well as high background measurements caused by the complexity of fluids used, although this can be alleviated through the removal (or reduction) of specific metal components within the fluid, in line with the solutes expected to be released from the test material [140]. The current use of dissolution within RA tools may not be so greatly impacted by sensitivity, as the thresholds used are very broad and/or rather elementary, using a ranking based on dissolution time (ANSES, Swiss Precautionary Matrix) or soluble concentration (GUIDEnano). Advances have been made recently to include threshold decisions based on dissolution rate [21,123]. Nevertheless, unless very significant changes are made to the particle to result in very different dissolution behaviour, it is unlikely that the sensitivity of the thresholds used will be dynamic enough to provide meaningful SbD decisions on dissolution behaviour.
In terms of compatibility, the potential of acellular tests is broad, and other than particular hydrophobic materials, it is difficult to list examples that could not be tested. In fact, these acellular methods have been used for some time to resolve the time-kinetic release of metals within complex materials, such as man-made fibres, or from occupational dusts such as welding fumes [141]. The biological predictivity of acellular tests, although not always established within the literature, has been demonstrated to a relatively high level, with various promising outcomes. For example, the solubility of BaSO4 in the dynamic system was considered to reproduce what is known for the solubility of BaSO4 in vivo, while the results of the static system underestimated this [137]. With the use of a lysosomal simulant fluid, the dynamic dissolution system was also shown to replicate cellular dissolution of BaSO4 (in conjunction with SrCO3 and ZnO) in rat macrophage models [123], and similarly acellular dissolution of MoO3 in the same lysosomal simulant fluid was found comparable to dissolution within mouse macrophage models [142]. These studies, and others, have demonstrated that by using various simulated biological fluids of intracellular compartments and/or lung lining fluid, a number of correlations with either cellular assays or in vivo exposures can be attained [123,143,144,145,146]; however, it should be acknowledged that there is an equal number of studies that have shown no correlation, raising the concern for appropriate fluid selection [127].

Cellular Methods

The basic principle of the cellular method is simple and can be performed cheaply as typical assessments investigate dissolution within cells up to 24 h. The difficulty with this methodology is the success of analysing the ions released. There are various options for separating cells and supernatant, such as centrifugal ultrafiltration and cloud point extraction [147], however it is not always as straightforward as the acellular assessments, as released ions may form complexes with biomolecules and therefore separation may be hampered [148]. Additional concerns may arise from the complexing of ions to biomolecules. Therefore, studies have often opted to determine both the ion concentration and the NM concentration to aim to avoid false positives or false negatives [149].
Koltermann-Jülly et al. (2018) found that macrophage-assisted dissolution in vitro was only applicable for an exposure period of 1–2 days, which they believe is too short and may be responsible for the low amounts of ions detected for the NMs tested [123]. The authors state that using cellular systems gives no additional benefit to the abiotic flow-through system with regards to predicting the in vivo response. This conclusion, however, is based only on the three materials tested. Moreover, when a cellular method uses uptake as an inference of dissolution, overestimating particle concentrations may occur, when measurements are not only of internalised particles but also of those adhered to the cell membrane. However, this is likely to be resolved by following well-described methodology which includes steps to limit this interference such as ensuring thorough washing and etching of the cells prior to analysis to remove adhered particles [150]. Further promising methodologies for this include the isolation of NMs and ions from cells using Triton X-114-based cloud point extraction as has previously been conducted for intracellular Ag NMs and Ag+ isolation [147,149].

4.2.2. Overview of Needs and Knowledge Gaps

Table 2 shows a summary of how the different dissolution assays perform in terms of the criteria for SbD hazard testing. There is a wealth of studies available for interpretation of in vitro dissolution methods, and although there are promising findings, there are still too many uncertainties to be sure of which model is most appropriate or reliable for specific materials. For SbD hazard testing, the use of a static system would be preferred, due to its simplicity. Although correlations with in vivo outcomes have been shown for static and flow-through systems, this is not always the case, and therefore requires further attention. Going forward, assessing predictivity will be important, especially when assessing novel materials; however, auspiciously, as shown above, for some substances a strong relationship between acellular, cellular, and in vivo findings has already been observed. It has been previously suggested that specific in vitro methods (e.g., specific fluid choices) should be selected based on feasible degradation pathways [142] which could be dependent upon specific degradation routes e.g., complexation, protonation, or to establish robust and fully accurate biological simulations [127]. Moreover, if these methods are to be applied in grouping and read-across approaches, the development of reliable and robust methods for determining particle dissolution rates has been considered paramount [123].
There is a wealth of studies available for interpretation of in vitro dissolution methods, and although there are promising findings, there are still too many uncertainties to be sure of which model is most appropriate or reliable for specific materials. For SbD hazard testing, the use of a static system would be preferred, due to its simplicity.

4.3. Oxidative Potential and Oxidative Stress

The oxidative potential (OP) of a NM is a chemical property that defines the ability of a NM to form potentially toxic species such as hydroxyl (OH) and superoxide (O2) radicals and hydrogen peroxide (H2O2) (collectively called reactive oxygen species (ROS)), or reactive nitrogen species (RNS) through redox reactions. This parameter is part of many NM hazard assessment strategies and grouping approaches due to its potential as a predictor of toxicity [13,14,15,18]. The pros and cons of OP assays in NM research have extensively been reviewed previously [156,157].
Oxidative stress (OS) is a cellular state in which the amount of ROS, caused by NM OP or the release of reactive ions, overwhelms the cells’ antioxidant capacity, potentially leading to the oxidation of biomolecules, inflammation, and oxidative DNA damage [158,159]. OS is seen as an important key event in the mode of action of many NMs and is therefore important to quantify as an early warning indictor [156,160,161].

4.3.1. Most Frequently Used Assays, Strengths and Limitations

Acellular Methods

In an acellular assay, OP is usually measured as a rate of depletion of a reductor. OP assays do not measure reductor depletion by OP only, since the release of reactive ions by dissolution will also lead to a depletion. Multiple acellular assays have been proposed to evaluate NM OP. The acellular dichloro-dihydro-fluorescein (DCFH) assay, electron paramagnetic resonance (EPR), electron spin resonance (ESR) spectroscopy, and the Ferric Reducing Ability of Serum (FRAS) assay are frequently used and have been evaluated extensively in literature [162,163,164,165,166,167]. An additional assay which is especially relevant for measuring NM OP is the haemolysis assay. For this assay, red blood cells are isolated from whole blood, and the ability of NMs to disturb their membranes is measured through absorbance. This assay requires whole blood, but no cell culturing, making it an easy and cost-effective method. No interferences have been reported in this assay, but it has not been studied extensively for NMs. There is no information available on robustness, and there is no publicly available standardized protocol.
Out of the other acellular assays, standardized protocols are only available for EPR/ESR (i.e., ISO 18827:2017). These assays require relatively expensive equipment, not always available in standard laboratories. FRAS and DCFH are considered simple and low-cost assays, requiring equipment that is present in any standard biological lab [165,166,168]. The FRAS method was originally developed to measure the ferric reduction in blood plasma (FRAP) [169]. It has been adapted to be used with serum, optimized for smaller volumes [165] and for multi-dose measurements, while showing good sensitivity and reproducibility for several metal-bearing NMs [163]. Recently, the FRAS protocol was successfully adapted to measure the reactivity of graphene-based materials by adding a filtration step for NMs of very low density [168]. Interlaboratory studies for the FRAS assay have not yet been performed.
The robustness of the acellular DCFH assay using different NMs was evaluated in a recent inter-laboratory study. A good robustness was found for the positive control NMs when normalizing fluorescence values between labs. However, for the other NMs, interlaboratory reproducibility differed per particle type [170]. Several papers reported NM interference with, for example, the fluorescent readout of the DCFH assay [162,166,171]. Zhao and Riediker (2014) identified several other factors that could reduce the reliability of the acellular DCFH assay, such as the use of different dispersing agents, as well as using too-high concentrations of NM [172]. Interference by way of NM flocculation or optical interference has been noted regarding the FRAS assay when testing various NM pigments [173], while no NM interference has been reported for ESR/EPR.
The EPR, DCFH, and FRAS assays have recently been evaluated for NM-grouping purposes. Results showed that the sensitivity of the methods greatly depends on the type of particle studied. For example, CuO, BaSO4, and Mn2O3 were consistent in their reactivity level across the three methods, but ZnO and CeO2 only showed a response in the FRAS assay, and not in EPR and DCFH measurements [173]. This might suggest that reactive species produced by certain NMs are captured better by some assays than by others, or that the FRAS assay is more sensitive in general. The latter has been confirmed in several studies that showed that the FRAS and ESR/EPR perform better than the DCFH assay in terms of sensitivity [163,171,174,175].
The choice of assay should depend on the goal of testing. For example, if one would like to know which types of radicals are formed in order to know what to change in the NM design as a SbD intervention, ESR/EPR measurements with different spin traps will provide the most informative results [176]. The FRAS assay can provide a more general image of ROS generating potential, as a result of the cocktail of antioxidants that is present in serum. The DCFH assay is especially sensitive for one-electron oxidizing species (such as hydroxyl radicals) [177]. It should be taken into account that these assays measure the OP of the NM in the specific environment required by the assay. OP is greatly influenced by the exposure environment, and therefore the OP measured in the assay may not fully reflect the OP in a real-life exposure scenario. A relevant protein corona could be applied to ensure exposure relevancy, as is described in Section 3.2.

Cellular Methods

Cell-based assays can directly measure the intracellular ROS, irrespective of their origin (i.e., as a result of the surface chemistry of the NM, as a cell-generated signalling molecule, or as a defence mechanism of the cell within the phagolysosome), for example through use of the cellular DCFH-DA assay. Other options include the assessment of the effect of these radicals on biomolecules such as lipids (e.g., lipid peroxidation) and proteins (e.g., protein carbonylation), cellular antioxidant status (e.g., glutathione (GSH:GSSG ratio)), and antioxidant gene regulation (HO-1 expression and Nrf-2 reporter cell lines), of which the latter two are extensively described in Boyles et al. (2016) [178].
It has been suggested that OS measured in a cell-system has advantages over measuring the OP in acellular systems. By measuring in a cellular environment, the cells’ ability to defend itself against the induced OS is taken into account, the ROS’ generated genotoxicity can be assessed, and other mechanisms other than the OP that lead to OS are captured as well [156]. Other mechanisms leading to OS, such as through mitochondrial perturbation, have been shown for chemicals extracted from diesel exhaust particles [179,180], yet there is no convincing evidence that NMs are capable of inducing OS through mechanisms other than OP or ion release.

4.3.2. Predictivity and Relevance

For SbD hazard testing, it would be desirable to be able to predict human health effects or at least effects that are observed in studies in experimental animals with simple, fast, and cheap acellular OP assays. The ability of acellular assays to predict cellular oxidative stress and in vivo oxidative stress markers is quite good, as shown in a comprehensive review by Moller et al. (2010), but not for all particles and all test systems [160]. It has been shown that NMs can induce different types of ROS [181] and therefore it depends on the type of NM and their MOA whether assays can predict cellular and in vivo effects. For example, data derived with the haemolysis assay correlate very well with in vivo pulmonary inflammation for a panel of 13 metal oxide NMs (92% prediction accuracy), whereas EPR (69% prediction accuracy) and DCFH (77% prediction accuracy) results showed lower correlations [164]. The haemolysis assay was able to predict in vivo pro-inflammatory responses of both NMs that act through soluble ions as well as NMs that act through surface reactivity with a prediction accuracy of 62.5% for a panel of eight NMs [112]. The FRAS assay has even been shown to be able to correctly distinguish OPs between several types of CNTs [175]. In a study comparing pulmonary inflammation (PMN influxes) upon inhalation of a range of NMs with acellular ESR and DCFH results, the correlation was reasonable; however, here it was concluded that ESR measurements in macrophages give a higher prediction accuracy than the acellular assays [182]. For SiO2 NMs, EPR results correlated very well with in vitro cellular cytotoxicity [183]. ESR also correlated well with in vitro protein carbonylation for a large panel of NMs [184]. However, ESR as well as the FRAS assay were able to accurately predict only 50% of the in vivo outcomes for a panel of 35 NMs [162].
False positives in acellular OP assays (when compared to in vivo outcomes) can be explained by the fact that cells and organisms can resolve ROS to a certain extent. Therefore, effort should go towards establishing thresholds for these assays. False negatives in acellular OP assays can be explained by the fact that other mechanisms other than OP can lead to pulmonary inflammation as well, which cannot be detected by these assays. The large variation between prediction accuracies between studies could be explained by the differences between the NM panels tested. Each assay has a specific applicability domain and prediction accuracy will therefore depend on the NM types and the resulting types of ROS.
In general, cellular assays show a higher prediction accuracy than acellular assays, and a combination of both might perform even better [157,162,185]. However, for SbD hazard testing, acellular assays may already give a good indication of toxicity and could serve as a valuable initial screening in the very early stages of NM development.

4.3.3. Overview of Needs and Knowledge Gaps

Table 3 shows a summary of how the different OP assays perform in terms of the criteria for SbD hazard testing. There are clear indications that only measuring acellular reactivity would be sufficient for SbD hazard testing, when cellular testing is already performed for cytotoxicity, genotoxicity, and pro-inflammatory effects. OP assays can be used to categorize the materials and to explore in more depth if the OP can or should be reduced in a SbD intervention. Mapping the prediction accuracy for each assay, as well as an applicability domain will help understand which assays can be used to predict which specific effects.
There are clear indications that only measuring acellular reactivity would be sufficient for SbD hazard testing, when cellular testing is already performed for cytotoxicity, genotoxicity, and pro-inflammatory effects.

4.4. Inflammation

Many IATAs and testing strategies include the measurement of inflammatory potential using NMs since this is generally accepted as one of the key mechanisms of NM toxicity [15,19,20]. Pulmonary inflammation in response to NM exposure has been shown to lead to several adverse health effects, such as fibrosis as well as lung cancer in animal studies [120,186,187]. For oral exposure, inflammation is a key parameter in NM toxicity as well [188]. However, this section will focus on assays targeting the pulmonary route of exposure only.

4.4.1. Most Frequently Used Assays, Strengths and Limitations

It is impossible to capture the complexity of an in vivo inflammatory response in an in vitro model, where recruitment of inflammatory cells other than those already present cannot occur. It is however possible to detect the cytokines responsible for this recruitment in an in vitro experiment. The most widely used approach to assess inflammatory responses in in vitro assays is measuring cytokine production or secretion, using, for instance, an enzyme-linked immunosorbent assay (ELISA), RT-qPCR, or multiplex-based immunoassays [189] after exposing cultured cells to NMs. Measuring the levels of pro-inflammatory cytokines and other inflammatory mediators may give insight into the mechanisms of the immunomodulatory effects of NMs in vitro, such as inflammasome activation or dendritic cell maturation. Cytokines of specific interest for NM pulmonary toxicity are, amongst others, IL-8 as markers for neutrophil recruitment [190], IL-1β as a marker for NLRP3 inflammasome activation [191], and TNF-α as a marker for macrophage activation [192].
Cytokine release can be measured in e.g., epithelial cells, macrophages, and dendritic cells, cultured in mono- and co-cultures. Cells can be exposed in a submerged setup or at the air-liquid interface (ALI), where cells are cultured in contact with the air and exposed to aerosols on the apical side whilst kept in medium on the basal side, better resembling the physiological environment of cells in the respiratory tract (Figure 4).
A critical factor when assessing pro-inflammatory responses in cell models is that some NMs can interfere with common in vitro assays. SWCNT and MWCNT can non-specifically adsorb TNF-α and IL-8 to their surface, and TiO2 NMs have been described to be able to adsorb IL-8, thus causing a false-negative result in ELISA assays [99,118]. This effect has also been observed for Ag NM in combination with TNF-α and IL-8 [100]. NMs are also known to interfere with the components of the ELISA. This problem can be overcome by centrifugation to remove the NMs from the supernatant before performing the ELISA. It is also essential to test the NMs for endotoxin contamination, as endotoxins can induce inflammation at very low concentrations, leading to false positive results [193,194], especially since NMs are generally not produced in a sterile environment.
Despite the relevance of pro-inflammatory effects of NMs in human health, there is currently no validated test method available to investigate inflammatory responses in vitro. Submerged assays have been used far longer compared to the relatively new ALI models, and thus more advances in standardization and optimization have been accomplished. Only a few studies have been performed to show the robustness of one of these protocols. At the ALI, Calu-3 cells with and without macrophages (either differentiated THP-1 cells (dTHP-1) or primary cells) showed high reproducibility in seven participating labs based on measurements of membrane integrity and mitochondrial activity. Cytokine release however showed higher variability, although similar trends between the seven labs were observed [195]. The reproducibility between labs after exposure of A549 cells at the ALI to lipopolysaccharide (LPS) as a positive control was found to be quite low. However, after protocol optimizations, special training of personnel for cell handling, and homogenization of disposables and reagents, the reproducibility increased [196]. The reproducibility of results between labs when using dTHP-1 cells is a frequent topic of debate. Not only do they show varying responses to NMs, but also to a positive control such as LPS, as shown in a large inter-laboratory study [100]. In another large inter-laboratory study by Xia et al. (2013), it was shown that good results can be obtained when using very detailed protocols and using the same batch of serum and cells. They also showed that cell-culture conditions and the duration of differentiation greatly affect the variability of dTHP-1 cells between labs [108].
A frequently used alternative for dTHP-1 macrophages are monocyte derived macrophages (MDMs), derived from donor blood. Even though they are considered more predictive of the in vivo situation, they are also known for their donor-to-donor variation. The same holds true for the use of commercially available primary epithelial cells, which are considered more relevant, but also show considerable variation [197].
In ALI exposure systems, there are many other factors that may contribute to an increased variability, such as the accuracy of the microbalance in the exposure system, the quality of the nebulizer used, the method of sample preparation, etc. A comprehensive overview of factors that can influence reliability and robustness can be found in Petersen et al. (2021) [198].
In terms of predictivity, commercially available primary cells generally give a good indication of in vivo effects for known inflammation-inducing particles. Studies have shown that the pro-inflammatory effects of quartz [197], Ag NMs [199], SiO2 NMs [200], and Pd and Cu NMs [201] are accurately predicted using primary cell models. Co-cultures of cell lines are also able to predict in vivo responses in many cases, as has been shown for quartz [94,202], CuO [112], and ZnO [94]. In this latter study, it was shown that co-cultures perform better than the two cell types separately, showing the importance of interplay between epithelial cells and immune cells. The addition of macrophages seems to be crucial in order to capture a much wider domain of immunological responses as compared to epithelial cells only, as has been shown in multiple studies [108,203,204]. Epithelial cell lines in mono-cultures were not able to predict the toxic effects of quartz [205], but did accurately detect Ag NMs’ pro-inflammatory effects [112].
In short, primary cells are the most sensitive, followed by co-culture systems with macrophages, and then mono-cultures. There are however some studies that prove otherwise. Cho et al. (2013) showed that cell lines performed similar to primary alveolar macrophages and differentiated PBMCs in terms of accuracy [112]. The A549 epithelial cell line in tri-culture with inflammatory cells did not pick up the pro-inflammatory effect of Ag NMs [206]. Mono-cultures of the epithelial cell line 16-HBE better predicted in vivo effects of Ag NMs than when in co-culture with dTHP-1 cells [207]. Furthermore, CeO2, Co3O4, and NiO NMs induced an increase in granulocytes in BALF, whereas no pro-inflammatory effects were seen in submerged mono- and co-cultures [112]. Finally, BEAS-2B and dTHP-1 cells were able to predict a ranking in pro-inflammatory effects of several types of CNTs which corresponded to in vivo markers of lung fibrosis in two separate studies [208,209]. This could mean that cell lines could be suitable for SbD hazard testing. Likely, different modes of action of toxicity require different levels of complexity in a cell model. In order to be sure about the predictive capacity of the different cell types, more types of NMs should be tested.
The exposure method chosen will also impact the predictivity of the method. Exposing ALI-cultured cells is generally considered a more sensitive approach, since it is more physiologically relevant, as has been shown in multiple studies [210,211,212]. However, for SbD hazard testing it is desirable to work with a model that is as simple and cost-effective as possible. This disfavours the use of primary cells and favours simple submerged exposure systems as opposed to the more complex ALI cultures. There are strong indications that simple submerged models could be predictive enough for SbD hazard testing. For example, in a study by Loret et al. (2016) they concluded that, indeed, co-cultures were more sensitive than monocultures, and that ALI exposures were more sensitive than submerged cultures. However, the general ranking of the NMs in terms of their toxicity was similar across the various exposure methods [213]. A study by Di Ianni et al. (2021) showed a strong correlation between in vitro submerged co-cultures and in vivo results when testing CNTs [190]. In a study by Herzog et al. (2014), the pro-inflammatory effects of Ag NMs were not detected in ALI culture conditions, but were detected under submerged conditions, suggesting a better performance of the submerged model [214]. Submerged and ALI exposures performed equally well for cytotoxicity in response to TiO2 [215]. In a study by Panas et al. (2014), submerged conditions were more sensitive in detecting the pro-inflammatory effects of SiO2 NMs as compared to ALI conditions [216]. Altogether, the potential for submerged experiments to predict in vivo responses has been shown in multiple studies, and is worth exploring further, especially for SbD hazard testing.
The differences in sensitivity between the ALI and submerged exposures can be explained by many (potentially confounding) factors. Firstly, certain cell types such as A549 are suggested to produce surfactant at the ALI, but not under submerged conditions, making them more vulnerable to toxic effects in a submerged experiment [217]. Secondly, the effective dose in submerged experiments is not always (correctly) calculated, and this may lead to a skewed comparison to ALI and in vivo results. Thirdly, the medium used in the submerged experiments may have an impact on NM behaviour in terms of the protein corona and dissolution rate, which does not occur, or occurs differently, in ALI experiments. Lastly, studies finding a good correlation between an in vitro model and in vivo results are more likely to be published, leading to publication bias. In a comprehensive overview of different cell types and exposure methods by McLean et al. (in preparation), it was shown that for quartz hazard prediction, the strength of in vitro prediction of in vivo responses was highly inconsistent, and largely dependent upon the data and study quality, which highlighted a need for robust SOPs which take into account numerous requirements for in vitro/in vivo extrapolation (McLean et al., in preparation).
There are several advantages of using ALI over submerged exposures. Particle alterations due to interaction with medium (such as the formation of a protein corona, dissolution, agglomeration) are no longer an issue, and calculating the deposited dose is much easier as compared to submerged experiments [212,218]. ALI exposures are compatible with a wider range of NMs, including hydrophobic particles, as exposures can be performed using a powder. Without having to take into consideration their behaviour in medium, abrasion products of NEPs can also directly be applied, making this type of model especially interesting for assessing life-cycle considerations, as is crucial for SbD hazard testing. Using ALI exposures is however more time-consuming and less high-throughput. Additionally, robustness of deposited dose after nebulization in an ALI setup may be low, depending on the NM used [219].

4.4.2. Overview of Needs and Knowledge Gaps

Table 4 shows a summary of how the different inflammation assays perform in terms of the criteria for SbD hazard testing. For SbD hazard testing, it is crucial to include tests that are predictive yet simple and cost-effective. Therefore, based on the current literature, the use of a submerged co-culture model including at least a type of macrophage might be the most suitable. However, more research is needed to confirm that simple methods are predictive enough for early hazard screening by testing data-rich NMs. Moreover, novel and advanced NMs should be tested in the available cell models in order to determine the compatibility of the cell models and readouts with different types of NMs. For a better predictivity, avoiding issues with dosimetry and medium interactions, and for hydrophobic particles, a simple ALI experiment can be set-up for SbD hazard testing.
A short-lived inflammatory response is beneficial to help clear NMs from the lung, and macrophage recruitment may not necessarily be a hazard warning. There is still a poor understanding of which amount of inflammation could be considered an adverse outcome, especially when measured in vitro. Establishing meaningful thresholds for these assays is important. Moreover, since pulmonary inflammation is mostly a chronic adverse effect, more work should be focusing on predicting chronic effects with in vitro assays, with which a promising start has been made in the PATROLS project [220].
For SbD hazard testing, it is crucial to include tests that are predictive yet simple and cost-effective. Therefore, based on current literature, the use of a submerged co-culture model including at least a type of macrophage might be the most suitable. However, more research is needed to confirm that simple methods are predictive enough for early hazard screening.

4.5. Genotoxicity

One of the main safety concerns related to NMs is their possible genotoxicity [221,222]. Genotoxicity describes the capacity of a chemical or physical agent to produce genetic damage that, if left unrepaired, may lead to cancer [223]. Therefore, every mutagen is potentially carcinogenic [222].
Due to the important consequences to human health, mutagenicity is a hazard endpoint required in all product regulations (chemicals, biocides, pharmaceuticals, medical devices, food additives, cosmetics, etc.) [224]. The assessment of genotoxicity is based on validated in vitro assays, which can be followed up by validated in vivo assays, depending on the in vitro outcome and the regulation involved [225]. Therefore, genotoxicity assessment at an early stage of innovation is highly advised. In fact, genotoxicity is a key endpoint in most of the testing strategies developed for NMs [13,19,221,226,227].

4.5.1. Most Frequently Used Assays, Strengths and Limitations

The mutagenicity of chemicals is usually evaluated on the basis of a battery of standard genotoxicity assays, able to detect gene mutations, chromosomal damage, and aneuploidy, as all these different mechanisms need to be considered in the assessment [221,224,227]. A core in vitro battery comprising the Ames test (detecting bacterial gene mutations) plus the in vitro micronucleus test (detecting chromosomal damage and aneuploidy) was already proposed 10 years ago for soluble chemicals [228]. Nearly 100% (958 out of 962) of rodent carcinogens or in vivo genotoxins were correctly detected with these two tests, which makes this battery a particularly sensitive combination [224]. However, the specificity of both assays together was unacceptably low (12.0%), giving rise to a high rate of false-positive results [229]. Hence, most of the EU regulations require a follow-up in vivo study when in vitro positive results are obtained [224]. In the case of NMs, the Ames test does not appear to be a suitable method as some NMs may not be able to penetrate through the bacterial wall, whereas others may kill the bacteria due to their bactericidal effects [230,231,232,233,234]. Based on this evidence, results obtained with this method should be followed up with other gene mutation assays using mammalian cells [21,235], or better yet, the Ames test should be avoided for NMs.
A roadmap for the genotoxicity testing of NMs was suggested some years ago [236], followed by guidance and common considerations [237]. There are two OECD TGs for assessing in vitro mammalian gene mutations: the In vitro Mammalian Cell Gene Mutation Tests using the Hprt and xprt genes (OECD TG 476), and the In vitro Mammalian Cell Gene Mutation Tests Using the Thymidine Kinase Gene (OECD TG 490). The latter, also called the mouse lymphoma assay (MLA), can detect a broader spectrum of genetic damage than the former, including chromosome rearrangements, deletions, and mitotic recombination [236]. Both assays are time-consuming, requiring long culture times (e.g., 10–14 days before counting colony formation), which has probably precluded an extensive use of these assays. For soluble chemicals, the sensitivity and specificity of the MLA assay was reported to be 73.1 and 39.0%, respectively, resulting in a prediction accuracy of 62.9 % [224]. In the case of NMs, given the low number of studies performed with these assays, and the wide variety of NMs included in these few studies, it has not been possible to draw any conclusions concerning the relative sensitivity of the various reporter genes to the potential mutagenicity of NMs [236]. Nevertheless, there are ongoing efforts to adapt the HPRT assay for use with NMs, e.g., within the EU H2020 RiskGone project, where round robin activities are ongoing.
Among the assays detecting chromosome damage, the In vitro Mammalian Cell Micronucleus (MN) Test (OECD TG 487) has been the most extensively used in nanotoxicology [221,230]. The assay detects chromosome mutations induced by either clastogenic or aneugenic agents. In addition, it also detects most mutagenic events as most mechanisms leading to gene mutation also induce chromosome mutations [228]. For soluble chemicals, the sensitivity and specificity of the MN assay was reported to be 78.7 and 30.8%, respectively [229], resulting in a concordance of 67.8%. In the case of NMs, there are few papers that evaluated a similar material, and there is a substantial variation in the methodology applied, which precludes raising conclusions on the reproducibility and predictability of this assay [236].
The most classically used version of the MN assay is the cytochalasin-blocked MN assay, which includes the use of cytochalasin B, a cytokinesis blocking agent that enables the identification of dividing cells [238]. However, as cytochalasin B may impair NM intracellular internalization, leading to false-negative results in the MN assay [239], it is recommended to successively treat the cells with NMs, and then with this agent [240,241]. Since cells should undergo mitosis for binucleated cells to form, the use of serum in exposure medium is recommended. The need of both proliferating cells and cells accumulating NMs in the MN assay has been illustrated recently while optimizing the MN assay on 3D cell models. The 3D EpiDerm™ skin model accumulates less NMs than 2D skin cells, resulting in less MNs upon exposure to genotoxic ZnO NMs [242]. Moreover, HepG2 spheroids still hold the capacity to proliferate while HepaRG spheroids do not, and genotoxic ZnO NMs do not show a positive outcome in the MN assay in 3D HepaRG while they do on the 3D HepG2 model [242,243]. An adaptation of this TG for NMs, within the OECD project 4.95 (‘Guidance Document on the Adaptation of In vitro Mammalian Cell Based Genotoxicity TGs for Testing of Manufactured Nanomaterials’), is currently ongoing based on previous recommendations [236]. One of the first round robin studies on the in vitro MN assay was performed within the NanoGenotox project [244], involving 12 laboratories, and comparing the genotoxicity of three reference materials. Relatively reproducible results were obtained in some cases, but they were material- and cell line-specific. A similar conclusion was reached by Louro et al. (2016) on four benchmark MWCNTs in two lung epithelial cell lines [245]. One reason could be the low fold increase over control values, as was also pointed out in the genotoxicity assessment performed by Elespuru et al. (2018) [236]. Currently, round robin tests are planned within the OECD project 4.95 and the EU H2020 RiskGone project.
Lately, the MN assay has been applied in co-culture systems involving inflammatory cells (e.g., THP-1 cells) and target cells (e.g., lung epithelial cells) allowing the evaluation of the mechanisms of action (primary vs. secondary) underlying NMs’ genotoxicity [246,247]. Genotoxins operating by a secondary mechanism of action, mediated by inflammation, are assumed to have a threshold response [248].
Classically, the MN assay has involved a labour-intensive manual scoring under the microscope. However, the speed of the analyses can nowadays be increased by using automated microscope scoring platforms [249], or flow cytometry [250,251,252]. The latter has recently been adapted to NMs [253].
The other validated method for assessing chromosome damage is the In vitro Mammalian Chromosome Aberration (CA) Test (OECD TG 473). This assay has been used much less because it is more time-consuming and requires a significant level of expertise to score the aberrations [236]. Furthermore, the CA assay does not detect aneugens, while the MN assay does (OECD TG 473, 2016). Hence, the CA assay would not be recommended for SbD hazard testing.
Furthermore, HTS approaches that could be applied to the testing of NMs are currently in development. These methods are non-OECD-guideline methods but proved efficient to detect potential NM genotoxicity. The comet assay is by far the most employed among these assays, and it could complement the recommended in vitro mutagenicity assays [236]. During the past decade, some effort has been dedicated to increase its throughput, with the highest achieved in the 96-minigel version using gel bond films [254]. It has been optimized and successfully applied to assess the genotoxicity of NMs within the FP7 NanoREG project [255].
Lastly, one commonly used genotoxicity assay for NMs is the immunolabelling of DNA repair protein foci, such as gamma-H2AX, which form during DNA double-strand break repair. The background of DNA double-strand breaks in cells is generally very low (although some exceptions exist, such as in some cancer cell lines), which makes these assays very sensitive. High-throughput versions of the assays exist. Foci can be counted using automated microscopy platforms or flow cytometry, with the advantage of their rapidity and possibility of analysing other cell parameters such as cell viability or apoptosis simultaneously [256]. Such high-throughput methods have rarely been applied on advanced 3D cell models for assessing NM genotoxicity because they necessitate additional steps. For example, a high-throughput comet assay can be performed on 3D cells after enzymatic and mechanical dissociation of the spheroid [257], reducing simplicity. Still, such advanced models could increase the predictivity of the assay. For example, Ag NMs cause significant DNA damage in a 2D liver-cell system, while the outcome of the comet assay is insignificant in 3D HepG2 spheroids, which is similar to most of the in vivo studies published up to now reporting Ag NM genotoxicity via comet assay [258].

4.5.2. Overview of Needs and Knowledge Gaps

Table 5 shows a summary of how the different genotoxicity assays perform in terms of the criteria for SbD hazard testing. One of the main problems for determining the sensitivity and predictability of the genotoxicity assays when assessing NMs is the absence of nano-sized particulate controls. NM-specific controls have rarely been demonstrated [236,259], making comparisons among labs difficult. Furthermore, it is not possible to establish historical positive control ranges that would confirm the sensitivity of the tests [259]. Based on the currently available information, we follow the recommendations by Elespuru et al. (2018) [236] to use the MN assay in combination with a gene-mutation assay (HPRT or MLA). In the meantime, further optimizations of genotoxicity assays for testing NMs are ongoing.
For advanced models, such as 3D models, the 3D skin comet and micronucleus assays are sufficiently validated for conventional chemicals and individual OECD TGs could start being developed [260]. However, the 3D airway and liver models are still lacking assays that could measure micronuclei and gene mutations, respectively [260]. Working with NMs raises additional technical hurdles that need to be overcome. Nevertheless, advanced models will offer advantages over current assays, especially by mimicking better the human body response and being able to evaluate modes of actions, e.g., secondary genotoxicity [261].
Based on the currently available information, we follow the recommendations by Elespuru et al. (2018) to use the MN assay in combination with a gene mutation assay (HPRT or MLA). In the meantime, further optimizations of genotoxicity assays for testing NMs are ongoing.

5. Discussion and Outlook

The development, manufacturing, and use of NMs with novel properties and potentially undesired health risks are growing at a rapid rate. One way to reduce potential adverse effects caused by NMs is to incorporate SbD in NM development processes. Within a SbD approach, the potential hazards of a NM throughout the life cycle are assessed at an early stage of product innovation. Although advancements have been made in terms of nano-specific risk assessment strategies [13,19,138,227], several challenges remain when applying these strategies to SbD:
  • Current hazard and risk-assessment strategies are not easily applied in an early hazard assessment for SbD applicability, as the proposed and required assays are too time-consuming and costly to be performed early in the development process of a NM.
  • The suitability for SbD hazard testing of currently available in vitro assays in terms of predictivity, cost-effectiveness, sensitivity, specificity, robustness, and compatibility are largely unclear.
In this review, in vitro toxicity assays have been critically assessed in terms of their suitability for SbD hazard testing. The main purpose of SbD hazard testing is the identification of early hazard warnings and obtaining a general idea of the potential hazards of a novel NM, NEP, or components released thereof during the LC. It therefore serves as a first screening during the early stages of the development of a new NM or NEP. For SbD hazard testing, a balance needs to be sought between simplicity and comprehensive testing that addresses all concerns (Figure 5). The more elaborate the assessment, the more uncertainty is minimized, and the more the testing becomes too complex for the purpose of SbD.

5.1. Assay Predictivity

5.1.1. Early Hazard Warnings

Correlating in vitro effects to in vivo potency has not yet been possible [109] and is not a requirement for SbD hazard testing. The identification of hazard warnings and detecting the most potent NMs is more relevant, and in this review it was shown that many assays are capable of doing this. The prediction accuracy of the evaluated assays in many cases depends on the type of particle, sample preparation, as well as the type of cell system used. Effects of NMs which assert their effect through ion shedding, such as Ag and ZnO NMs, were most accurately predicted across all toxicity endpoints. Current in vitro assays were less capable of predicting the effects of NMs acting through surface reactivity, and fibre-like NMs. Another important finding across several toxicity outcomes was the better predictivity of primary cells as compared to cell lines, as well as better predictivity of macrophage-like cells as compared to other cell types. However, there are clear indications that simple submerged assays might be suitable for prediction of adverse effects in vivo, as was also shown in a recent review by Di Ianni et al. (2022) [262].

5.1.2. Hazard Ranking

For an assay to be able to establish an accurate ranking in toxicity is valuable for SbD hazard testing, as this allows the use of the assay for comparison between candidate NMs, and for comparison to benchmark materials with known toxicity. There are indications that in vitro cytotoxicity assays are able to predict an adequate ranking in toxicity which corresponds to in vivo pulmonary inflammation [114]. Also, the detection of cytokines at the ALI could potentially detect a ranking that corresponds to in vivo pro-inflammatory mediators [211]. However, both of these studies could not draw any definitive conclusions on comparable rankings. Two studies showed that simple submerged cell lines are able to produce accurate rankings in pro-inflammatory effects that corresponded to in vivo markers of fibrosis [208,209]. It is important to note that the accuracy of toxicity rankings is hugely dependent on the calculation of the dose delivered to the cells, and this should therefore always be carried out [59].

5.1.3. Applicability Domains

A low prediction accuracy of an assay could be improved by exploring the exact applicability of the assay. Ensuring that the specific MOA that caused the in vivo toxicity can be detected using the assay will reduce the rate of false negative outcomes. The applicability domain of each assay should be well-understood (which assay can predict what kind of in vivo (human) toxicity) to be able to use assays that are fit-for-purpose. When looking at the transition to animal free testing in general, this is one of the issues that needs addressing for soluble chemicals as well [263]. In that respect, in vitro toxicity testing of NMs should be mechanism-based by looking for specific effects or MOAs [264]. If the applicability domain of assays and cell models is established with more certainty, it is possible to combine assays into a strategy to holistically assess NM toxicity in vitro. In such a strategy, assay and cell type selection would be facilitated by a combination of applicability domains and the most relevant exposure route.

5.1.4. Prediction Accuracy

Prediction accuracy in terms of sensitivity and specificity should be determined for more assays, to increase knowledge about assay reliability. Determination of accuracy is a crucial step during the validation process of in vitro assays in general [265]. For example, for regulatory genotoxicity testing, it is known that in vitro assays have a high sensitivity but low specificity. This means that there is a high chance of false positives, and positives should always be confirmed in an in vivo study. A better understanding of the prediction accuracies of in vitro assays used for SbD hazard testing would help enormously with the interpretation of results.

5.1.5. Challenges in Assessing Predictivity

Although in vitro assays have been used for some time to test NM toxicity, not all the criteria could be evaluated properly due to the lack of or limited availability of high-quality data. Especially for predictivity, there is a data gap that needs to be filled in order to correctly interpret results. Several factors complicate the assessment of prediction accuracy of in vitro assays. Since there is a lack of human data on NM toxicity, predictivity of assays is at present evaluated compared to in vivo data derived from studies in experimental animals. This means that an in vitro model comprised of human cells is being compared to animal data, to predict a human response. The relevance of this approach is questionable, due to the differences between humans and experimental animals [266,267]. The lack of deposited dose calculations, interference controls, proper characterization, and varying sample preparation protocols (e.g., the use of serum, different media, different dispersion techniques) across the literature add another layer of complexity to assessing the predictivity of assays. Since these factors can have such an impact on assay outcome, assay standardization will aid in determining assay predictivity for adverse human health effects. Additionally, the lack of clear positive and negative controls for NMs hampers the assessment of prediction accuracy.
Finally, it must be noted that in the papers reviewed here, an optimistic perspective is given about the predictivity of in vitro assays for toxicity and adverse health outcomes in vivo. We should however be cautious, as negative results or results with a low correlation with known in vivo or human health responses may not reach publication: a phenomenon known as publication bias.

5.2. Outlook for Innovators, Regulators, and Industry Based on Current Knowledge

An overview of the most important knowns and unknowns with regards to NM SbD hazard testing is summarized in Table 6. Figure 6 shows what we think is the road forward towards successfully putting SbD hazard testing into practice. The successful implementation of SbD hazard testing requires efforts from innovators, regulators, as well as from industry.

5.2.1. A Change in Mindset towards Purpose-Driven Innovations

The current European policy landscape (the European Green Deal, the European Chemical Strategy for Sustainability and the Zero Pollution Action Plan [2,3,268]) demands a new mindset for innovating. SbD provides an approach aiming at developing safer NMs and NEPs by integrating safety into the innovation process and material development in a LC thinking approach, from design to end-of-life. Any innovation that does not have a green or sustainable purpose will not survive.

5.2.2. Starting In Silico: Databases and SARs

SbD hazard assessment should first and foremost be based on material knowledge and material–activity relationships. Before even starting in vitro experiments, an elaborate evaluation of available physicochemical data should be performed [220]. Here, certain hazard warnings could already be noticed. For example, the structure–activity relationship (SAR) of high aspect ratio NMs (HARNs) and mesothelioma risk is widely accepted [138]. It would be unnecessary to perform hazard testing on HARNs, as it would already be clear beforehand that this material raises a hazard warning. Another potential hazard warning would be respirable crystalline silica particles, due to their structure–activity relationship with silicosis and lung cancer [269,270]. Knowledge on structure activity relationships is especially important for the identification of potential hazards and application of SbD interventions.
For novel advanced materials however, limited information on these tox-driving properties is available, and the SbD decisions are mostly based on SbD hazard-testing outcomes.

5.2.3. Importance of Experimental Design

The physical aspects of NMs add another dimension to the complexity of toxicity testing. It should always be considered that the way the experiment is carried out (dispersion protocol, medium type, addition of serum) affects the outcomes and that the behaviour of the particle in the culture dish (settling, agglomerating, floating, dissolution, formation of protein corona) should always be analysed [23,49,53,58,59]. Checking and accounting for assay interference is crucial, also for SbD hazard testing and high throughput screening, where it is often overlooked [97]. This makes SbD hazard testing for NMs more challenging than that of soluble chemicals.

5.2.4. Combinations of Assays

An integrated approach to testing and assessment (IATA) that can combine information from multiple sources (available data, in silico tools, in vitro assays) is the way forward towards an effective early hazard identification of NMs and NEPs and for the development of SbD interventions. This review discusses simple assays since it focusses on the initial stages of innovation. However, at more advanced stages, SbD hazard testing may also include approaches that are not as simple and cost-effective [264]. With regards to the transition to animal-free alternatives, the focus on simplicity as is required for SbD hazard testing should not create a barrier for the development of more realistic and innovative cell models with potentially better predictivity, such as induced pluripotent stem cells (iPSCs) and organoids.
For inflammatory potential, chronic inflammation (leading to tissue damage and remodelling as well as loss of functions) is the adverse outcome of concern, which is presently not captured by one or more in vitro tests. An acute pro-inflammatory effect in an in vitro assay as measured by cytokine secretion, in combination with slow dissolution, indicating high bio-persistency [14], might together indicate that the NM induces chronic inflammation. Combining assay outcomes in SbD hazard testing should be further explored.

5.2.5. Thresholds for Toxicity

In order to raise hazard warnings and to interpret results from combinations of assays, thresholds are needed. This is especially challenging for inflammatory potential assays. Macrophages are the major defence mechanism against foreign materials, and their activation is crucial for the clearance of NMs [192]. It is unclear when a beneficial immune response turns into persistent pulmonary inflammation in vivo, and how to predict this in vitro.
Previously established frameworks have made a step towards generating thresholds for toxicity. The Nanoreg2 framework and the Swiss Precautionary Matrix score NMs as low, medium, or high hazard according to their fold change increase as compared to a negative control [271]. The Nanoreg2 framework adds a scoring system that allows for combining outcomes of different assays, and subsequent comparison of different NMs. With both approaches, a significantly positive response in an assay might still lead to a classification as low hazard.
Since SbD hazard testing is performed as an early screening, and its main goal is determining early hazard warnings, a zero-tolerance principle might be more suitable in this case (as is common practice in the pharmaceutical industry). For primary genotoxicity, a zero-tolerance principle is already in place in regulatory risk assessment, as genotoxic carcinogens are regarded as having no threshold and thus an acceptable exposure level cannot be derived [224]. For SbD hazard testing, it could be argued that a worst-case approach would be suitable for the other endpoints as well, meaning that any indication of inflammation, reactivity, or cytotoxicity at relevant doses would raise a hazard warning. Here it is important to consider the possibility of false negatives produced in the assays.
For SbD hazard testing, the inclusion of benchmark NMs with known in vivo toxicity is recommended to compare the new NM to existing information. Thresholds could be set according to the response of the benchmark NM in a specific assay. Alternatively, an appropriate ranking in potency of NMs could be useful for making SbD decisions when comparing several candidate NMs.

5.2.6. Assay Standardization

In Figure 6, assay standardization is represented connecting many important aspects. As mentioned throughout this review, assay standardization is a key need for the further development of SbD hazard testing, as well as for putting SbD into practice. Firstly, we showed that in vitro-in vivo comparisons are hampered by the lack of standardized protocols. Moreover, fundamental research into structure–activity relationships will benefit from standardized protocols as well. Assay standardization will result in more high-quality fundamental data on the MOAs of toxicity of NMs, which will in turn aid the refining of SbD hazard testing. Ultimately, standardization will increase the chances of industrial use and acceptance of these assays into existing legal frameworks, which will make incorporating SbD approaches more appealing for manufacturers [272].
On the contrary, the complexity of NM toxicity testing hampers the standardization of assays. It is for example impossible to create one exposure method suitable for all NMs, especially considering NMs of the future which will possess yet unknown properties. A case-by-case or targeted approach will be needed for specific NMs with incompatible PC properties. In some cases, standardization may not be feasible, but guidance will be of great help.
Assay standardization should be followed by assay validation in order to improve our understanding of the robustness, predictivity, and compatibility of the assays. The ongoing work in the OECD’s Working Party on Manufactured Nanomaterials [273], the Malta Initiative [274], and work in ongoing European projects such as NanoHarmony [275], Nanomet [276] and Gov4Nano [277] are currently supporting the standardization efforts.

5.2.7. Compatibility (NEPs and Novel Materials)

Safety along the LC as well as keeping pace with the rapid emergence of advanced materials are important hallmarks of SbD. Consumers are most likely exposed to NEPs and not pristine NMs. Therefore, assay optimization is needed to be able to test NEPs in an accurate way. Assay compatibility with NEPs and novel advanced materials needs to be studied further. More data on how NMs can change over the LC and the possible risks they may pose during this process is very much needed. This will help determine whether testing only pristine NMs may be sufficient for SbD hazard testing.

5.2.8. Gathering Experimental Data following FAIR Principles

Since SbD hazard testing will involve the generation of large datasets, it is important to ensure that the data gathered from the different in vitro assays are adequately collected using templates that support FAIR principles, and that the data is findable, accessible, interoperable and reusable. Guidance for finding these templates can be found in the GoFair initiative and guidance on experimental workflows design and implementation can be found within the NanoCommons initiative.

5.2.9. The Chemical Strategy for Sustainability

Although this review covers cytotoxicity, dissolution, oxidative potential, inflammatory potential, and genotoxicity, the Chemical Strategy for Sustainability has put forth extra endpoints to ensure the ambition towards a toxic-free environment and protection against the most harmful chemicals is fulfilled [3]. One of these endpoints is endocrine disruption. Under REACH, endocrine disruptors are identified as substances of very high concern alongside chemicals known to cause cancer, mutations, and toxicity to reproduction. Work is ongoing by ECHA to develop classification and labelling criteria for endocrine disruption [278]. From a NM-perspective, there is increasing evidence showing endocrine disruption and reproductive impairments caused by NMs such as nano plastics [279,280], and this warrants further attention.
Although this review is only focused on SbD, sustainability impacts should also be considered early in the innovation process. Safe-and-sustainable-by-design is a central element of the European Chemical Strategy for Sustainability and it demands the optimization of safety and sustainability interventions in the design of NMs, NEPs, and all processes in a life-cycle approach.

6. Conclusions

This review provides the first building blocks towards an early hazard testing strategy for SbD applicability and is the first detailed state of the art analysis of in vitro assays against performance criteria (simplicity and cost effectiveness, predictivity, robustness, compatibility, and readiness) for SbD hazard testing. The most important conclusions are:
  • Based on current knowledge, primary cell models and more physiologically relevant exposure methods provide better predictions of in vivo results. However, the aim of SbD hazard testing is to detect early hazard warnings using simple methods. There are strong indications that simpler assays, such as acellular OP assays, static dissolution assays, and simple submerged cell-based assays for cytotoxicity, genotoxicity, and inflammation give sufficiently accurate information for identifying early hazard warnings or even hazard rankings, when carried out correctly.
  • The suitability of these simple assays for SbD hazard testing has to be further confirmed in future studies. More model comparisons between simple, complex, and in vivo models are needed to investigate whether simple in vitro models are indeed sufficiently predictive and suitable for SbD hazard testing, preferably using standardized methods. Additionally, the applicability domain of in vitro assays to detect NM toxicity should be mapped more precisely to correctly interpret results.
  • Assay standardization proved to be critical for the progression of SbD hazard testing as it will improve in vitro-in vivo comparisons, improve fundamental knowledge on NM toxicity, support industrial use, and is a first step towards regulatory acceptance.
  • Simplicity is not always feasible when testing NMs, even though it has been put forward as one of the criteria for SbD hazard testing. Dispersion protocols, dose delivered to cells, compatibility issues, interferences, testing NEPs and NMs released along the LC, etc., all complicate SbD hazard testing of NMs and reduce achievable simplicity. Innovators, industry, regulators, and policymakers should realize that the hazard assessment of NMs and advanced materials is complex and that in vitro tests need to be further developed, tested, and evaluated to assess their suitability in identifying potential hazards.

Author Contributions

All authors contributed to the conceptualization of the manuscript. The preparation of the different sections of the original draft was done by the following authors: Introduction by N.R. and L.G.S.-H.; Criteria by N.R.; Choice of dispersion protocol by M.C. and J.C. (Joan Cabellos); the use of serum and stabilizers by N.R.; determining dose delivered to cells by N.R.; SbD hazard testing of NEPs and NMs released during the life cycle by M.C., C.D., A.S.J. and N.R.; challenging NMs and advanced materials by N.R.; cytotoxicity by H.B. and N.R.; Dissolution by M.B. and P.M.; oxidative potential and oxidative stress by A.K., J.C (Joan Cabellos) and N.R.; inflammation by A.K., A.C. and N.R.; genotoxicity by M.C. and J.C. (Julia Catalán); discussion and outlook by N.R., H.B. and L.G.S.-H.; conclusions by N.R., L.G.S.-H., F.R.C. and H.B. Afterwards, in several iterations all authors read, commented, and proposed revisions to the entire manuscript, which were compiled by N.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the SAbyNA project, European Union’s Horizon 2020 research and innovation program under grant agreement No 862419, and by the Dutch Ministry of Infrastructure and Water Management.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

The authors would like to thank Rob Vandebriel and Agnes Oomen for their valuable suggestions and critical review of the manuscript.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Falk, A.; Cassee, F.R.; Valsami-Jones, E. Safe-by-Design and EU Funded NanoSafety Projects; European Commission: Brussels, Belgium, 2021. [Google Scholar] [CrossRef]
  2. European Commission. Communication from the Commission to the European Parliament, the Council, the European Economic and Social Committee and the Committee of the Regions: The European Green Deal; European Commission: Brussels, Belgium, 2019. [Google Scholar]
  3. European Commission. Chemicals Strategy for Sustainability—Towards a Toxic-Free Environment; European Commission: Brussels, Belgium, 2020. [Google Scholar]
  4. Gottardo, S.; Alessandrelli, M.; Amenta, V.; Atluri, R.; Barberio, G.; Bekker, C.; Bergonzo, P.; Bleeker, E.; Booth, A.M.; Borges, T.; et al. NANoREG Framework for the Safety Assessment of Nanomaterials; European Commission Joint Research Centre: Brussels, Belgium, 2017. [Google Scholar]
  5. Sánchez Jiménez, A.; Puelles, R.; Pérez-Fernández, M.; Gómez-Fernández, P.; Barruetabeña, L.; Jacobsen, N.R.; Suarez-Merino, B.; Micheletti, C.; Manier, N.; Trouiller, B.; et al. Safe(r) by design implementation in the nanotechnology industry. NanoImpact 2020, 20, 100267. [Google Scholar] [CrossRef]
  6. OECD. Moving Towards a Safe(r) Innovation Approach (SIA) for More Sustainable Nanomaterials and Nano-Enabled Products; Series on the Safety of Manufactured Nanomat; OECD: Paris, France, 2020. [Google Scholar]
  7. Schmutz, M.; Borges, O.; Jesus, S.; Borchard, G.; Perale, G.; Zinn, M.; Sips, Ä.A.J.A.M.; Soeteman-Hernandez, L.G.; Wick, P.; Som, C. A Methodological Safe-by-Design Approach for the Development of Nanomedicines. Front. Bioeng. Biotechnol. 2020, 8, 258. [Google Scholar] [CrossRef]
  8. Soeteman-Hernandez, L.G.; Apostolova, M.D.; Bekker, C.; Dekkers, S.; Grafström, R.C.; Groenewold, M.; Handzhiyski, Y.; Herbeck-Engel, P.; Hoehener, K.; Karagkiozaki, V.; et al. Safe innovation approach: Towards an agile system for dealing with innovations. Mater. Today Commun. 2019, 20, 100548. [Google Scholar] [CrossRef]
  9. Xia, T.; Zhao, Y.; Sager, T.; George, S.; Pokhrel, S.; Li, N.; Schoenfeld, D.; Meng, H.; Lin, S.; Wang, X.; et al. Decreased Dissolution of ZnO by Iron Doping Yields Nanoparticles with Reduced Toxicity in the Rodent Lung and Zebrafish Embryos. ACS Nano 2011, 5, 1223–1235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Rubio, L.; Pyrgiotakis, G.; Beltran-Huarac, J.; Zhang, Y.; Gaurav, J.; DeLoid, G.; Spyrogianni, A.; Sarosiek, K.A.; Bello, D.; Demokritou, P. Safer-by-design flame-sprayed silicon dioxide nanoparticles: The role of silanol content on ROS generation, surface activity and cytotoxicity. Part. Fibre Toxicol. 2019, 16, 40. [Google Scholar] [CrossRef]
  11. Zhou, J.; Cai, D.; Xu, Q.; Zhang, Y.; Fu, F.; Diao, H.; Liu, X. Excellent binding effect of l-methionine for immobilizing silver nanoparticles onto cotton fabrics to improve the antibacterial durability against washing. RSC Adv. 2018, 8, 24458–24463. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Hjorth, R.; van Hove, L.; Wickson, F. What can nanosafety learn from drug development? The feasibility of “safety by design”. Nanotoxicology 2017, 11, 305–312. [Google Scholar] [CrossRef] [Green Version]
  13. Dekkers, S.; Wijnhoven, S.W.P.; Braakhuis, H.M.; Soeteman-Hernandez, L.G.; Sips, A.J.A.M.; Tavernaro, I.; Kraegeloh, A.; Noorlander, C.W. Safe-by-Design part I: Proposal for nanospecific human health safety aspects needed along the innovation process. NanoImpact 2020, 18, 100227. [Google Scholar] [CrossRef]
  14. Di Cristo, L.; Oomen, A.G.; Dekkers, S.; Moore, C.; Rocchia, W.; Murphy, F.; Johnston, H.J.; Janer, G.; Haase, A.; Stone, V.; et al. Grouping Hypotheses and an Integrated Approach to Testing and Assessment of Nanomaterials Following Oral Ingestion. Nanomaterials 2021, 11, 2623. [Google Scholar] [CrossRef]
  15. Arts, J.H.E.; Hadi, M.; Irfan, M.-A.; Keene, A.M.; Kreiling, R.; Lyon, D.; Maier, M.; Michel, K.; Petry, T.; Sauer, U.G.; et al. A decision-making framework for the grouping and testing of nanomaterials (DF4nanoGrouping). Regul. Toxicol. Pharmacol. 2015, 71, S1–S27. [Google Scholar] [CrossRef] [Green Version]
  16. Stone, V.; Gottardo, S.; Bleeker, E.A.J.; Braakhuis, H.; Dekkers, S.; Fernandes, T.; Haase, A.; Hunt, N.; Hristozov, D.; Jantunen, P.; et al. A framework for grouping and read-across of nanomaterials- supporting innovation and risk assessment. Nano Today 2020, 35, 100941. [Google Scholar] [CrossRef]
  17. Rauscher, H.; Rasmussen, K.; Riego Sintes, J.; Sala, S. Safe and Sustainable by Design Chemicals and Materials; Publications Office of the EU: Luxembourg, 2022. [Google Scholar]
  18. Braakhuis, H.M.; Murphy, F.; Ma-Hock, L.; Dekkers, S.; Keller, J.; Oomen, A.G.; Stone, V. An Integrated Approach to Testing and Assessment to Support Grouping and Read-Across of Nanomaterials After Inhalation Exposure. Appl. In Vitro Toxicol. 2021, 7, 112–128. [Google Scholar] [CrossRef] [PubMed]
  19. Dekkers, S.; Oomen, A.G.; Bleeker, E.A.; Vandebriel, R.J.; Micheletti, C.; Cabellos, J.; Janer, G.; Fuentes, N.; Vázquez-Campos, S.; Borges, T.; et al. Towards a nanospecific approach for risk assessment. Regul. Toxicol. Pharmacol. 2016, 80, 46–59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Halappanavar, S.; Nymark, P.; Krug, H.F.; Clift, M.J.D.; Rothen-Rutishauser, B.; Vogel, U. Non-Animal Strategies for Toxicity Assessment of Nanoscale Materials: Role of Adverse Outcome Pathways in the Selection of Endpoints. Small 2021, 17, 2007628. [Google Scholar] [CrossRef]
  21. EFSA Scientific Committee; More, S.; Bampidis, V.; Benford, D.; Bragard, C.; Halldorsson, T.; Hernández-Jerez, A.; Hougaard Bennekou, S.; Koutsoumanis, K.; Lambré, C. Guidance on risk assessment of nanomaterials to be applied in the food and feed chain: Human and animal health. EFSA J. 2021, 19, e06768. [Google Scholar]
  22. Gulumian, M.; Cassee, F.R. Safe by design (SbD) and nanotechnology: A much-discussed topic with a prudence? Part. Fibre Toxicol. 2021, 18, 32. [Google Scholar] [CrossRef]
  23. DeLoid, G.M.; Cohen, J.M.; Pyrgiotakis, G.; Demokritou, P. Preparation, characterization, and in vitro dosimetry of dispersed, engineered nanomaterials. Nat. Protoc. 2017, 12, 355–371. [Google Scholar] [CrossRef]
  24. Pedersen, E.; Fant, K. Guidance Document on Good In Vitro Method Practices (GIVIMP): Series on Testing and Assessment No. 286; OECD: Paris, France, 2018. [Google Scholar]
  25. 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]
  26. Taurozzi, J.S.; Hackley, V.A.; Wiesner, M.R. Ultrasonic dispersion of nanoparticles for environmental, health and safety assessment—Issues and recommendations. Nanotoxicology 2011, 5, 711–729. [Google Scholar] [CrossRef]
  27. Kaur, I.; Ellis, L.-J.; Römer, I.; Tantra, R.; Carriere, M.; Allard, S.; Mayne-L’Hermite, M.; Minelli, C.; Unger, W.; Potthoff, A.; et al. Dispersion of Nanomaterials in Aqueous Media: Towards Protocol Optimization. J. Vis. Exp. 2017, 130, e56074. [Google Scholar] [CrossRef] [Green Version]
  28. Simon-Deckers, A.; Gouget, B.; Mayne-L’Hermite, M.; Herlin-Boime, N.; Reynaud, C.; Carrière, M. In vitro investigation of oxide nanoparticle and carbon nanotube toxicity and intracellular accumulation in A549 human pneumocytes. Toxicology 2008, 253, 137–146. [Google Scholar] [CrossRef]
  29. Hadrup, N.; Knudsen, K.B.; Carriere, M.; Mayne-L’Hermite, M.; Bobyk, L.; Allard, S.; Miserque, F.; Pibaleau, B.; Pinault, M.; Wallin, H.; et al. Safe-by-design strategies for lowering the genotoxicity and pulmonary inflammation of multiwalled carbon nanotubes: Reduction of length and the introduction of COOH groups. Environ. Toxicol. Pharmacol. 2021, 87, 103702. [Google Scholar] [CrossRef]
  30. Retamal Marín, R.R.; Babick, F.; Lindner, G.-G.; Wiemann, M.; Stintz, M. Effects of Sample Preparation on Particle Size Distributions of Different Types of Silica in Suspensions. Nanomaterials 2018, 8, 454. [Google Scholar] [CrossRef] [Green Version]
  31. Peters, R.; Kramer, E.; Oomen, A.G.; Herrera Rivera, Z.E.; Oegema, G.; Tromp, P.C.; Fokkink, R.; Rietveld, A.; Marvin, H.J.P.; Weigel, S.; et al. Presence of Nano-Sized Silica during In Vitro Digestion of Foods Containing Silica as a Food Additive. ACS Nano 2012, 6, 2441–2451. [Google Scholar] [CrossRef]
  32. Cappellini, F.; Hedberg, Y.; McCarrick, S.; Hedberg, J.; Derr, R.; Hendriks, G.; Wallinder, I.O.; Karlsson, H.L. Mechanistic insight into reactivity and (geno)toxicity of well-characterized nanoparticles of cobalt metal and oxides. Nanotoxicology 2018, 12, 602–620. [Google Scholar] [CrossRef]
  33. Pradhan, S.; Hedberg, J.; Blomberg, E.; Wold, S.; Wallinder, I.O. Effect of sonication on particle dispersion, administered dose and metal release of non-functionalized, non-inert metal nanoparticles. J. Nanopart. Res. 2016, 18, 285. [Google Scholar] [CrossRef] [Green Version]
  34. Alstrup Jensen, K.; Kembouche, Y.; Christiansen, E.; Jacobsen, N.; Wallin, H.; Guiot, C.; Spalla, O.; Witschger, O. Final Protocol for Producing Suitable Manufactured Nanomaterial Exposure Media; NANOGENOTOX deliverable report 3; EAHC: Luxembourg, 2011. [Google Scholar]
  35. Wegner, K.D.; Dussert, F.; Truffier-Boutry, D.; Benayad, A.; Beal, D.; Mattera, L.; Ling, W.L.; Carrière, M.; Reiss, P. Influence of the Core/Shell Structure of Indium Phosphide Based Quantum Dots on Their Photostability and Cytotoxicity. Front. Chem. 2019, 7, 466. [Google Scholar] [CrossRef] [Green Version]
  36. Dussert, F.; Wegner, K.D.; Moriscot, C.; Gallet, B.; Jouneau, P.-H.; Reiss, P.; Carriere, M. Evaluation of the Dermal Toxicity of InZnP Quantum Dots Before and After Accelerated Weathering: Toward a Safer-By-Design Strategy. Front. Toxicol. 2021, 3, 636976. [Google Scholar] [CrossRef]
  37. Tarantini, A.; Wegner, K.D.; Dussert, F.; Sarret, G.; Beal, D.; Mattera, L.; Lincheneau, C.; Proux, O.; Truffier-Boutry, D.; Moriscot, C.; et al. Physicochemical alterations and toxicity of InP alloyed quantum dots aged in environmental conditions: A safer by design evaluation. NanoImpact 2019, 14, 100168. [Google Scholar] [CrossRef]
  38. Rosset, A.; Bartolomei, V.; Laisney, J.; Shandilya, N.; Voisin, H.; Morin, J.; Michaud-Soret, I.; Capron, I.; Wortham, H.; Brochard, G.; et al. Towards the development of safer by design TiO2-based photocatalytic paint: Impacts and performances. Environ. Sci. Nano 2021, 8, 758–772. [Google Scholar] [CrossRef]
  39. Roebben, G.; Ramirez-Garcia, S.; Hackley, V.A.; Roesslein, M.; Klaessig, F.; Kestens, V.; Lynch, I.; Garner, C.M.; Rawle, A.; Elder, A.; et al. Interlaboratory comparison of size and surface charge measurements on nanoparticles prior to biological impact assessment. J. Nanopart. Res. 2011, 13, 2675–2687. [Google Scholar] [CrossRef]
  40. Bihari, P.; Vippola, M.; Schultes, S.; Praetner, M.; Khandoga, A.G.; Reichel, C.A.; Coester, C.; Tuomi, T.; Rehberg, M.; Krombach, F. Optimized dispersion of nanoparticles for biological in vitro and in vivo studies. Part. Fibre Toxicol. 2008, 5, 14. [Google Scholar] [CrossRef] [Green Version]
  41. Sharma, G.; Kodali, V.; Gaffrey, M.; Wang, W.; Minard, K.R.; Karin, N.J.; Teeguarden, J.G.; Thrall, B.D. Iron oxide nanoparticle agglomeration influences dose rates and modulates oxidative stress-mediated dose–response profiles in vitro. Nanotoxicology 2014, 8, 663–675. [Google Scholar] [CrossRef] [Green Version]
  42. Betts, J.N.; Johnson, M.G.; Rygiewicz, P.T.; King, G.A.; Andersen, C.P. Potential for metal contamination by direct sonication of nanoparticle suspensions. Environ. Toxicol. Chem. 2013, 32, 889–893. [Google Scholar] [CrossRef]
  43. Gülseren, I.; Güzey, D.; Bruce, B.D.; Weiss, J. Structural and functional changes in ultrasonicated bovine serum albumin solutions. Ultrason. Sonochem. 2007, 14, 173–183. [Google Scholar] [CrossRef] [PubMed]
  44. OECD. Guidance on sample preparation and dosimetry for the safety testing of manufactured nanomaterials. In Proceedings of Tour de Table at the 7th Meeting of the Working Party on Manufactured Nanomaterials No. 36; Series on the Safety of Manufactured Nanomaterials; OECD: Paris, France, 2012. [Google Scholar]
  45. Shannahan, J.H. Nanoparticle–Biocorona. In Encyclopedia of Nanotechnology; Bhushan, B., Ed.; Springer: Dordrecht, The Netherlands, 2014; pp. 1–4. [Google Scholar] [CrossRef]
  46. Strojan, K.; Leonardi, A.; Bregar, V.B.; Križaj, I.; Svete, J.; Pavlin, M. Dispersion of Nanoparticles in Different Media Importantly Determines the Composition of Their Protein Corona. PLoS ONE 2017, 12, e0169552. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Boyles, M.S.P.; Kristl, T.; Andosch, A.; Zimmermann, M.; Tran, N.; Casals, E.; Himly, M.; Puntes, V.; Huber, C.G.; Lütz-Meindl, U.; et al. Chitosan functionalisation of gold nanoparticles encourages particle uptake and induces cytotoxicity and pro-inflammatory conditions in phagocytic cells, as well as enhancing particle interactions with serum components. J. Nanobiotechnol. 2015, 13, 84. [Google Scholar] [CrossRef] [Green Version]
  48. McConnell, K.I.; Shamsudeen, S.; Meraz, I.M.; Mahadevan, T.S.; Ziemys, A.; Rees, P.; Summers, H.D.; Serda, R.E. Reduced Cationic Nanoparticle Cytotoxicity Based on Serum Masking of Surface Potential. J. Biomed. Nanotechnol. 2016, 12, 154–164. [Google Scholar] [CrossRef] [Green Version]
  49. Vranic, S.; Gosens, I.; Jacobsen, N.R.; Jensen, K.A.; Bokkers, B.; Kermanizadeh, A.; Stone, V.; Baeza-Squiban, A.; Cassee, F.R.; Tran, L.; et al. Impact of serum as a dispersion agent for in vitro and in vivo toxicological assessments of TiO2 nanoparticles. Arch. Toxicol. 2017, 91, 353–363. [Google Scholar] [CrossRef]
  50. 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] [Green Version]
  51. Lesniak, A.; Fenaroli, F.; Monopoli, M.P.; Åberg, C.; Dawson, K.A.; Salvati, A. Effects of the Presence or Absence of a Protein Corona on Silica Nanoparticle Uptake and Impact on Cells. ACS Nano 2012, 6, 5845–5857. [Google Scholar] [CrossRef]
  52. Elliott, J.T.; Rösslein, M.; Song, N.W.; Toman, B.; Kinsner-Ovaskainen, A.; Maniratanachote, R.; Salit, M.L.; Petersen, E.J.; Sequeira, F.; Romsos, E.L.; et al. Toward achieving harmonization in a nano-cytotoxicity assay measurement through an interlaboratory comparison study_suppl. ALTEX-Altern. Anim. Exp. 2017, 34, 201–218. [Google Scholar] [CrossRef] [Green Version]
  53. Wiemann, M.; Vennemann, A.; Venzago, C.; Lindner, G.-G.; Schuster, T.B.; Krueger, N. Serum Lowers Bioactivity and Uptake of Synthetic Amorphous Silica by Alveolar Macrophages in a Particle Specific Manner. Nanomaterials 2021, 11, 628. [Google Scholar] [CrossRef]
  54. Pisani, C.; Rascol, E.; Dorandeu, C.; Gaillard, J.-C.; Charnay, C.; Guari, Y.; Chopineau, J.; Armengaud, J.; Devoisselle, J.-M.; Prat, O. The species origin of the serum in the culture medium influences the in vitro toxicity of silica nanoparticles to HepG2 cells. PLoS ONE 2017, 12, e0182906. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Žūkienė, R.; Snitka, V. Zinc oxide nanoparticle and bovine serum albumin interaction and nanoparticles influence on cytotoxicity in vitro. Colloids Surf. B Biointerfaces 2015, 135, 316–323. [Google Scholar] [CrossRef] [PubMed]
  56. Kittler, S.; Greulich, C.; Gebauer, J.S.; Diendorf, J.; Treuel, L.; Ruiz, L.; Gonzalez-Calbet, J.M.; Vallet-Regi, M.; Zellner, R.; Köller, M.; et al. The influence of proteins on the dispersability and cell-biological activity of silver nanoparticles. J. Mater. Chem. 2010, 20, 512. [Google Scholar] [CrossRef]
  57. Pyrgiotakis, G.; Blattmann, C.O.; Pratsinis, S.; Demokritou, P. Nanoparticle–Nanoparticle Interactions in Biological Media by Atomic Force Microscopy. Langmuir 2013, 29, 11385–11395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Cohen, J.; DeLoid, G.; Pyrgiotakis, G.; Demokritou, P. Interactions of engineered nanomaterials in physiological media and implications for in vitro dosimetry. Nanotoxicology 2013, 7, 417–431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Pal, A.K.; Bello, D.; Cohen, J.; Demokritou, P. Implications of in vitro dosimetry on toxicological ranking of low aspect ratio engineered nanomaterials. Nanotoxicology 2015, 9, 871–885. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. DeLoid, G.M.; Cohen, J.M.; Pyrgiotakis, G.; Pirela, S.V.; Pal, A.; Liu, J.; Srebric, J.; Demokritou, P. Advanced computational modeling for in vitro nanomaterial dosimetry. Part. Fibre Toxicol. 2015, 12, 32. [Google Scholar] [CrossRef] [Green Version]
  61. Thomas, D.G.; Smith, J.N.; Thrall, B.D.; Baer, D.R.; Jolley, H.; Munusamy, P.; Kodali, V.; Demokritou, P.; Cohen, J.; Teeguarden, J.G. ISD3: A particokinetic model for predicting the combined effects of particle sedimentation, diffusion and dissolution on cellular dosimetry for in vitro systems. Part. Fibre Toxicol. 2018, 15, 6. [Google Scholar] [CrossRef] [Green Version]
  62. Botte, E.; Vagaggini, P.; Zanoni, I.; Gardini, D.; Costa, A.L.; Ahluwalia, A.D. An integrated pipeline and multi-model graphical user interface for accurate nano-dosimetry. bioRxiv 2021. [Google Scholar] [CrossRef]
  63. Lison, D.; Thomassen, L.C.J.; Rabolli, V.; Gonzalez, L.; Napierska, D.; Seo, J.W.; Kirsch-Volders, M.; Hoet, P.; Kirschhock, C.E.A.; Martens, J.A. Nominal and Effective Dosimetry of Silica Nanoparticles in Cytotoxicity Assays. Toxicol. Sci. 2008, 104, 155–162. [Google Scholar] [CrossRef]
  64. Keller, J.G.; Quevedo, D.F.; Faccani, L.; Costa, A.L.; Landsiedel, R.; Werle, K.; Wohlleben, W. Dosimetry in vitro—exploring the sensitivity of deposited dose predictions vs. affinity, polydispersity, freeze-thawing, and analytical methods. Nanotoxicology 2021, 15, 21–34. [Google Scholar] [CrossRef]
  65. DeLoid, G.; Cohen, J.M.; Darrah, T.; Derk, R.; Rojanasakul, L.; Pyrgiotakis, G.; Wohlleben, W.; Demokritou, P. Estimating the effective density of engineered nanomaterials for in vitro dosimetry. Nat. Commun. 2014, 5, 3514. [Google Scholar] [CrossRef] [Green Version]
  66. Ding, Y.; Weindl, P.; Lenz, A.-G.; Mayer, P.; Krebs, T.; Schmid, O. Quartz crystal microbalances (QCM) are suitable for real-time dosimetry in nanotoxicological studies using VITROCELL®Cloud cell exposure systems. Part. Fibre Toxicol. 2020, 17, 44. [Google Scholar] [CrossRef]
  67. Yokel, R.A.; MacPhail, R.C. Engineered nanomaterials: Exposures, hazards, and risk prevention. J. Occup. Med. Toxicol. 2011, 6, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Basinas, I.; Jiménez, A.S.; Galea, K.S.; Van Tongeren, M.; Hurley, F. A Systematic Review of the Routes and Forms of Exposure to Engineered Nanomaterials. Ann. Work. Expo. Health 2018, 62, 639–662. [Google Scholar] [CrossRef] [Green Version]
  69. Schneider, T.; Brouwer, D.H.; Koponen, I.K.; Jensen, K.A.; Fransman, W.; Van Duuren-Stuurman, B.; van Tongeren, M.; Tielemans, E. Conceptual model for assessment of inhalation exposure to manufactured nanoparticles. J. Expo. Sci. Environ. Epidemiol. 2011, 21, 450–463. [Google Scholar] [CrossRef]
  70. Duncan, T.V.; Pillai, K. Release of Engineered Nanomaterials from Polymer Nanocomposites: Diffusion, Dissolution, and Desorption. ACS Appl. Mater. Interfaces 2015, 7, 2–19. [Google Scholar] [CrossRef] [PubMed]
  71. Al-Kattan, A.; Wichser, A.; Vonbank, R.; Brunner, S.; Ulrich, A.; Zuin, S.; Arroyo, Y.; Golanski, L.; Nowack, B. Characterization of materials released into water from paint containing nano-SiO2. Chemosphere 2015, 119, 1314–1321. [Google Scholar] [CrossRef]
  72. Murray, A.R.; Kisin, E.R.; Tkach, A.V.; Yanamala, N.; Mercer, R.; Young, S.-H.; Fadeel, B.; Kagan, V.E.; Shvedova, A.A. Factoring-in agglomeration of carbon nanotubes and nanofibers for better prediction of their toxicity versus asbestos. Part. Fibre Toxicol. 2012, 9, 10. [Google Scholar] [CrossRef] [Green Version]
  73. Noël, A.; Charbonneau, M.; Cloutier, Y.; Tardif, R.; Truchon, G. Rat pulmonary responses to inhaled nano-TiO2: Effect of primary particle size and agglomeration state. Part. Fibre Toxicol. 2013, 10, 48. [Google Scholar] [CrossRef] [Green Version]
  74. Murugadoss, S.; Brassinne, F.; Sebaihi, N.; Petry, J.; Cokic, S.M.; Van Landuyt, K.L.; Godderis, L.; Mast, J.; Lison, D.; Hoet, P.H.; et al. Agglomeration of titanium dioxide nanoparticles increases toxicological responses in vitro and in vivo. Part. Fibre Toxicol. 2020, 17, 10. [Google Scholar] [CrossRef]
  75. Saber, A.T.; Jacobsen, N.R.; Mortensen, A.; Szarek, J.; Jackson, P.; Madsen, A.M.; Jensen, K.A.; Koponen, I.K.; Brunborg, G.; Gützkow, K.B.; et al. Nanotitanium dioxide toxicity in mouse lung is reduced in sanding dust from paint. Part. Fibre Toxicol. 2012, 9, 4. [Google Scholar] [CrossRef] [Green Version]
  76. Pal, A.K.; Watson, C.Y.; Pirela, S.V.; Singh, D.; Chalbot, M.-C.G.; Kavouras, I.; Demokritou, P. Linking Exposures of Particles Released from Nano-Enabled Products to Toxicology: An Integrated Methodology for Particle Sampling, Extraction, Dispersion, and Dosing. Toxicol. Sci. 2015, 146, 321–333. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Kim, S.; Jaques, P.A.; Chang, M.; Froines, J.R.; Sioutas, C. Versatile aerosol concentration enrichment system (VACES) for simultaneous in vivo and in vitro evaluation of toxic effects of ultrafine, fine and coarse ambient particles Part I: Development and laboratory characterization. J. Aerosol Sci. 2001, 32, 1281–1297. [Google Scholar] [CrossRef]
  78. Nowack, B.; Boldrin, A.; Caballero, A.; Hansen, S.F.; Gottschalk, F.; Heggelund, L.; Hennig, M.; Mackevica, A.; Maes, H.; Navratilova, J.; et al. Meeting the Needs for Released Nanomaterials Required for Further Testing—The SUN Approach. Environ. Sci. Technol. 2016, 50, 2747–2753. [Google Scholar] [CrossRef] [Green Version]
  79. Demokritou, P.; Lee, S.J.; Ferguson, S.T.; Koutrakis, P. A compact multistage (cascade) impactor for the characterization of atmospheric aerosols. J. Aerosol Sci. 2004, 35, 281–299. [Google Scholar] [CrossRef]
  80. Bein, K.J.; Wexler, A.S. A high-efficiency, low-bias method for extracting particulate matter from filter and impactor substrates. Atmos. Environ. 2014, 90, 87–95. [Google Scholar] [CrossRef] [Green Version]
  81. Zarcone, M.C.; Duistermaat, E.; van Schadewijk, A.; Jedynska, A.; Hiemstra, P.S.; Kooter, I.M. Cellular response of mucociliary differentiated primary bronchial epithelial cells to diesel exhaust. Am. J. Physiol.-Lung Cell. Mol. Physiol. 2016, 311, L111–L123. [Google Scholar] [CrossRef] [Green Version]
  82. Wohlleben, W.; Brill, S.; Meier, M.W.; Mertler, M.; Cox, G.; Hirth, S.; Von Vacano, B.; Strauss, V.; Treumann, S.; Wiench, K.; et al. On the Lifecycle of Nanocomposites: Comparing Released Fragments and their In-Vivo Hazards from Three Release Mechanisms and Four Nanocomposites. Small 2011, 7, 2384–2395. [Google Scholar] [CrossRef] [PubMed]
  83. Smulders, S.; Luyts, K.; Brabants, G.; Van Landuyt, K.; Kirschhock, C.E.A.; Smolders, E.; Golanski, L.; Vanoirbeek, J.; Hoet, P.H. Toxicity of Nanoparticles Embedded in Paints Compared with Pristine Nanoparticles in Mice. Toxicol. Sci. 2014, 141, 132–140. [Google Scholar] [CrossRef]
  84. Smulders, S.; Luyts, K.; Brabants, G.; Golanski, L.; Martens, J.; Vanoirbeek, J.; Hoet, P.H. Toxicity of nanoparticles embedded in paints compared to pristine nanoparticles, in vitro study. Toxicol. Lett. 2015, 232, 333–339. [Google Scholar] [CrossRef]
  85. Shoemaker, D.A.; Pretty, J.R.; Ramsey, D.M.; McLaurin, J.L.; Khan, A.; Teass, A.W.; Castranova, V.; Pailes, W.H.; Dalal, N.S.; Miles, P.R. Particle activity and in vivo pulmonary response to freshly milled and aged alpha-quartz. Scand. J. Work. Environ. Health 1995, 21 (Suppl. 2), 15–18. [Google Scholar] [PubMed]
  86. Buford, M.C.; Hamilton, R.F.; Holian, A. A comparison of dispersing media for various engineered carbon nanoparticles. Part. Fibre Toxicol. 2007, 4, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Polk, W.W.; Sharma, M.; Sayes, C.M.; Hotchkiss, J.A.; Clippinger, A.J. Aerosol generation and characterization of multi-walled carbon nanotubes exposed to cells cultured at the air-liquid interface. Part. Fibre Toxicol. 2016, 13, 20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Watson, C.Y.; DeLoid, G.M.; Pal, A.; Demokritou, P. Buoyant Nanoparticles: Implications for Nano-Biointeractions in Cellular Studies. Small 2016, 12, 3172–3180. [Google Scholar] [CrossRef] [Green Version]
  89. Stock, V.; Böhmert, L.; Dönmez, M.H.; Lampen, A.; Sieg, H. An inverse cell culture model for floating plastic particles. Anal. Biochem. 2020, 591, 113545. [Google Scholar] [CrossRef]
  90. Saleh, N.B.; Aich, N.; Plazas-Tuttle, J.; Lead, J.R.; Lowry, G.V. Research strategy to determine when novel nanohybrids pose unique environmental risks. Environ. Sci. Nano 2015, 2, 11–18. [Google Scholar] [CrossRef]
  91. Kroll, A.; Pillukat, M.H.; Hahn, D.; Schnekenburger, J. Current in vitro methods in nanoparticle risk assessment: Limitations and challenges. Eur. J. Pharm. Biopharm. 2009, 72, 370–377. [Google Scholar] [CrossRef] [PubMed]
  92. Stone, V.; Johnston, H.; Schins, R.P.F. Development of in vitro systems for nanotoxicology: Methodological considerations. Crit. Rev. Toxicol. 2009, 39, 613–626. [Google Scholar] [CrossRef] [PubMed]
  93. Hillegass, J.M.; Shukla, A.; Lathrop, S.A.; MacPherson, M.B.; Fukagawa, N.K.; Mossman, B.T. Assessing nanotoxicity in cells in vitro. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2010, 2, 219–231. [Google Scholar] [CrossRef] [Green Version]
  94. Sayes, C.; Reed, K.L.; Warheit, D.B. Assessing Toxicity of Fine and Nanoparticles: Comparing In Vitro Measurements to In Vivo Pulmonary Toxicity Profiles. Toxicol. Sci. 2007, 97, 163–180. [Google Scholar] [CrossRef] [Green Version]
  95. Guadagnini, R.; Halamoda Kenzaoui, B.; Walker, L.; Pojana, G.; Magdolenova, Z.; Bilanicova, D.; Saunders, M.; Juillerat-Jeanneret, L.; Marcomini, A.; Huk, A.; et al. Toxicity screenings of nanomaterials: Challenges due to interference with assay processes and components of classic in vitro tests. Nanotoxicology 2015, 9 (Suppl. 1), 13–24. [Google Scholar] [CrossRef]
  96. Ribeiro, A.R.; Leite, P.E.; Falagan-Lotsch, P.; Benetti, F.; Micheletti, C.; Budtz, H.C.; Jacobsen, N.R.; Lisboa-Filho, P.N.; Rocha, L.A.; Kühnel, D.; et al. Challenges on the toxicological predictions of engineered nanoparticles. NanoImpact 2017, 8, 59–72. [Google Scholar] [CrossRef]
  97. Andraos, C.; Yu, I.J.; Gulumian, M. Interference: A Much-Neglected Aspect in High-Throughput Screening of Nanoparticles. Int. J. Toxicol. 2020, 39, 397–421. [Google Scholar] [CrossRef]
  98. Savage, D.T.; Hilt, J.Z.; Dziubla, T.D. In vitro methods for assessing nanoparticle toxicity. In Nanotoxicity; Methods in Molecular Biology; Zhang, Q., Ed.; Humana Press: New York, NY, USA, 2019; Volume 1894, pp. 1–29. [Google Scholar] [CrossRef]
  99. Kroll, A.; Pillukat, M.H.; Hahn, D.; Schnekenburger, J. Interference of engineered nanoparticles with in vitro toxicity assays. Arch. Toxicol. 2012, 86, 1123–1136. [Google Scholar] [CrossRef]
  100. Piret, J.-P.; Bondarenko, O.M.; Boyles, M.S.P.; Himly, M.; Ribeiro, A.R.; Benetti, F.; Smal, C.; Lima, B.; Potthoff, A.; Simion, M.; et al. Pan-European inter-laboratory studies on a panel of in vitro cytotoxicity and pro-inflammation assays for nanoparticles. Arch. Toxicol. 2017, 91, 2315–2330. [Google Scholar] [CrossRef]
  101. Han, X.; Gelein, R.; Corson, N.; Wade-Mercer, P.; Jiang, J.; Biswas, P.; Finkelstein, J.N.; Elder, A.; Oberdörster, G. Validation of an LDH assay for assessing nanoparticle toxicity. Toxicology 2011, 287, 99–104. [Google Scholar] [CrossRef] [Green Version]
  102. Nelissen, I.; Haase, A.; Anguissola, S.; Rocks, L.; Jacobs, A.; Willems, H.; Riebeling, C.; Luch, A.; Piret, J.-P.; Toussaint, O.; et al. Improving Quality in Nanoparticle-Induced Cytotoxicity Testing by a Tiered Inter-Laboratory Comparison Study. Nanomaterials 2020, 10, 1430. [Google Scholar] [CrossRef]
  103. Labouta, H.I.; Sarsons, C.; Kennard, J.; Gomez-Garcia, M.J.; Villar, K.; Lee, H.; Cramb, D.T.; Rinker, K.D. Understanding and improving assays for cytotoxicity of nanoparticles: What really matters? RSC Adv. 2018, 8, 23027–23039. [Google Scholar] [CrossRef] [Green Version]
  104. ISO 19007:2018; Nanotechnologies—In Vitro MTS Assay for Measuring the Cytotoxic Effect of Nanoparticles. International Organization for Standardization: Geneva, Switzerland, 2018.
  105. ISO/TS 21633:2021; Label-Free Impedance Technology to Assess the Toxicity of Nanomaterials In Vitro. International Organization for Standardization: Geneva, Switzerland, 2021.
  106. Da Luz, C.M.; Boyles, M.S.P.; Falagan-Lotsch, P.; Pereira, M.R.; Tutumi, H.R.; Santos, E.D.O.; Martins, N.B.; Himly, M.; Sommer, A.; Foissner, I.; et al. Poly-lactic acid nanoparticles (PLA-NP) promote physiological modifications in lung epithelial cells and are internalized by clathrin-coated pits and lipid rafts. J. Nanobiotechnol. 2017, 15, 11. [Google Scholar] [CrossRef] [Green Version]
  107. NANoREG. Deliverable D 5.06. Identification and Optimization of the Most Suitable In Vitro Methodology. 2016. Available online: https://www.rivm.nl/sites/default/files/2019-01/NANoREG_D5_06_DR_Identification_and_optimization_of_the_most_suitable_in_vitro_methodology_Yo3qmgPoS9aEh1-9l2OVcA.pdf (accessed on 14 November 2022).
  108. Xia, T.; Hamilton, R.F.; Bonner, J.C.; Crandall, E.D.; Elder, A.; Fazlollahi, F.; Girtsman, T.A.; Kim, K.; Mitra, S.; Ntim, S.A.; et al. Interlaboratory Evaluation of In Vitro Cytotoxicity and Inflammatory Responses to Engineered Nanomaterials: The NIEHS Nano GO Consortium. Environ. Health Perspect. 2013, 121, 683–690. [Google Scholar] [CrossRef] [Green Version]
  109. Landsiedel, R.; Sauer, U.G.; Ma-Hock, L.; Schnekenburger, J.; Wiemann, M. Pulmonary toxicity of nanomaterials: A critical comparison of published in vitro assays and in vivo inhalation or instillation studies. Nanomedicine 2014, 9, 2557–2585. [Google Scholar] [CrossRef]
  110. Sohaebuddin, S.K.; Thevenot, P.T.; Baker, D.; Eaton, J.W.; Tang, L. Nanomaterial cytotoxicity is composition, size, and cell type dependent. Part. Fibre Toxicol. 2010, 7, 22. [Google Scholar] [CrossRef] [Green Version]
  111. Kroll, A.; Dierker, C.; Rommel, C.; Hahn, D.; Wohlleben, W.; Schulze-Isfort, C.; Göbbert, C.; Voetz, M.; Hardinghaus, F.; Schnekenburger, J. Cytotoxicity screening of 23 engineered nanomaterials using a test matrix of ten cell lines and three different assays. Part. Fibre Toxicol. 2011, 8, 9. [Google Scholar] [CrossRef] [Green Version]
  112. Cho, W.-S.; Duffin, R.; Bradley, M.; Megson, I.L.; MacNee, W.; Lee, J.K.; Jeong, J.; Donaldson, K. Predictive value of in vitro assays depends on the mechanism of toxicity of metal oxide nanoparticles. Part. Fibre Toxicol. 2013, 10, 55. [Google Scholar] [CrossRef] [Green Version]
  113. Han, X.; Corson, N.; Wade-Mercer, P.; Gelein, R.; Jiang, J.; Sahu, M.; Biswas, P.; Finkelstein, J.N.; Elder, A.; Oberdörster, G. Assessing the relevance of in vitro studies in nanotoxicology by examining correlations between in vitro and in vivo data. Toxicology 2012, 297, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Mannerström, M.; Zou, J.; Toimela, T.; Pyykkö, I.; Heinonen, T. The applicability of conventional cytotoxicity assays to predict safety/toxicity of mesoporous silica nanoparticles, silver and gold nanoparticles and multi-walled carbon nanotubes. Toxicol. In Vitro 2016, 37, 113–120. [Google Scholar] [CrossRef]
  115. Seagrave, J.; McDonald, J.D.; Mauderly, J.L. In vitro versus in vivo exposure to combustion emissions. Exp. Toxicol. Pathol. 2005, 57, 233–238. [Google Scholar] [CrossRef] [PubMed]
  116. Warheit, D.B.; Sayes, C.M.; Reed, K.L. Nanoscale and Fine Zinc Oxide Particles: Can In Vitro Assays Accurately Forecast Lung Hazards following Inhalation Exposures? Environ. Sci. Technol. 2009, 43, 7939–7945. [Google Scholar] [CrossRef]
  117. Lanone, S.; Rogerieux, F.; Geys, J.; Dupont, A.; Maillot-Marechal, E.; Boczkowski, J.; Lacroix, G.; Hoet, P. Comparative toxicity of 24 manufactured nanoparticles in human alveolar epithelial and macrophage cell lines. Part. Fibre Toxicol. 2009, 6, 14. [Google Scholar] [CrossRef] [PubMed]
  118. Monteiro-Riviere, N.A.; Inman, A.O.; Zhang, L.W. Limitations and relative utility of screening assays to assess engineered nanoparticle toxicity in a human cell line. Toxicol. Appl. Pharmacol. 2009, 234, 222–235. [Google Scholar] [CrossRef] [PubMed]
  119. Donaldson, K.; Tran, C.L. An introduction to the short-term toxicology of respirable industrial fibres. Mutat. Res./Fundam. Mol. Mech. Mutagen. 2004, 553, 5–9. [Google Scholar] [CrossRef] [PubMed]
  120. Heinrich, U.; Fuhst, R.; Rittinghausen, S.; Creutzenberg, O.; Bellmann, B.; Koch, W.; Levsen, K. Chronic inhalation exposure of Wistar rats and two different strains of mice to diesel exhaust, carbon black and titanium dioxide. Inhal. Toxicol. 1995, 7, 23. [Google Scholar] [CrossRef]
  121. Driscoll, K.E.; Borm, P.J.A. Expert workshop on the hazards and risks of poorly soluble low toxicity particles. Inhal. Toxicol. 2020, 32, 53–62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Shin, H.U.; Stefaniak, A.B.; Stojilovic, N.; Chase, G.G. Comparative dissolution of electrospun Al2O3 nanofibres in artificial human lung fluids. Environ. Sci. Nano 2015, 2, 251–261. [Google Scholar] [CrossRef] [Green Version]
  123. Koltermann-Jülly, J.; Keller, J.G.; Vennemann, A.; Werle, K.; Müller, P.; Ma-Hock, L.; Landsiedel, R.; Wiemann, M.; Wohlleben, W. Abiotic dissolution rates of 24 (nano)forms of 6 substances compared to macrophage-assisted dissolution and in vivo pulmonary clearance: Grouping by biodissolution and transformation. NanoImpact 2018, 12, 29–41. [Google Scholar] [CrossRef]
  124. Keller, J.G.; Persson, M.; Müller, P.; Ma-Hock, L.; Werle, K.; Arts, J.; Landsiedel, R.; Wohlleben, W. Variation in dissolution behavior among different nanoforms and its implication for grouping approaches in inhalation toxicity. NanoImpact 2021, 23, 100341. [Google Scholar] [CrossRef]
  125. Plumlee, G.S.; Morman, S.A.; Ziegler, T.L. The Toxicological Geochemistry of Earth Materials: An Overview of Processes and the Interdisciplinary Methods Used to Understand Them. Rev. Miner. Geochem. 2006, 64, 5–57. [Google Scholar] [CrossRef] [Green Version]
  126. Krystek, P.; Kettler, K.; van der Wagt, B.; de Jong, W.H. Exploring influences on the cellular uptake of medium-sized silver nanoparticles into THP-1 cells. Microchem. J. 2015, 120, 45–50. [Google Scholar] [CrossRef]
  127. Innes, E.; Yiu, H.H.P.; McLean, P.; Brown, W.; Boyles, M. Simulated biological fluids—A systematic review of their biological relevance and use in relation to inhalation toxicology of particles and fibres. Crit. Rev. Toxicol. 2021, 51, 217–248. [Google Scholar] [CrossRef] [PubMed]
  128. Wohlleben, W.; Hellack, B.; Nickel, C.; Herrchen, M.; Hund-Rinke, K.; Kettler, K.; Riebeling, C.; Haase, A.; Funk, B.; Kühnel, D.; et al. The nanoGRAVUR framework to group (nano)materials for their occupational, consumer, environmental risks based on a harmonized set of material properties, applied to 34 case studies. Nanoscale 2019, 11, 17637–17654. [Google Scholar] [CrossRef] [PubMed]
  129. Alexander, I.C.; Brown, R.C.; Jubb, G.A.; Pickering, P.; Hoskins, J.A. Durability of ceramic and novel man-made mineral fibers. Environ. Health Perspect. 1994, 102, 67–71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Christensen, V.R.; Jensen, S.L.; Guldberg, M.; Kamstrup, O. Effect of chemical composition of man-made vitreous fibers on the rate of dissolution in vitro at different pHs. Environ. Health Perspect. 1994, 102, 83–86. [Google Scholar] [CrossRef]
  131. Spitler, G.; Spitz, H.; Glasser, S.; Hoffman, M.K.; Bowen, J. In Vitro Dissolution of Uranium-contaminated Soil in Simulated Lung Fluid Containing a Pulmonary Surfactant. Health Phys. 2015, 108, 336–343. [Google Scholar] [CrossRef] [Green Version]
  132. Searl, A.; Cullen, R.T. An enzymatic tissue digestion method for fibre biopersistence studies. Ann. Occup. Hyg. 1997, 41, 721–727. [Google Scholar] [CrossRef]
  133. Utembe, W.; Potgieter, K.; Stefaniak, A.B.; Gulumian, M. Dissolution and biodurability: Important parameters needed for risk assessment of nanomaterials. Part. Fibre Toxicol. 2015, 12, 11. [Google Scholar] [CrossRef] [Green Version]
  134. Ansoborlo, E.; Hengé-Napoli, M.H.; Chazel, V.; Gibert, R.; Guilmette, R.A. Review and Critical Analysis of Available In Vitro Dissolution Tests. Health Phys. 1999, 77, 638–645. [Google Scholar] [CrossRef]
  135. Farrugia, C. Flow-through dissolution testing: A comparison with stirred beaker methods. Chronic Ill 2002, 6, 17–19. [Google Scholar]
  136. Braydich-Stolle, L.K.; Breitner, E.K.; Comfort, K.K.; Schlager, J.J.; Hussain, S.M. Dynamic Characteristics of Silver Nanoparticles in Physiological Fluids: Toxicological Implications. Langmuir 2014, 30, 15309–15316. [Google Scholar] [CrossRef]
  137. Keller, J.; Peijnenburg, W.; Werle, K.; Landsiedel, R.; Wohlleben, W. Understanding Dissolution Rates via Continuous Flow Systems with Physiologically Relevant Metal Ion Saturation in Lysosome. Nanomaterials 2020, 10, 311. [Google Scholar] [CrossRef] [Green Version]
  138. Murphy, F.; Dekkers, S.; Braakhuis, H.; Ma-Hock, L.; Johnston, H.; Janer, G.; di Cristo, L.; Sabella, S.; Jacobsen, N.R.; Oomen, A.G.; et al. An integrated approach to testing and assessment of high aspect ratio nanomaterials and its application for grouping based on a common mesothelioma hazard. NanoImpact 2021, 22, 100314. [Google Scholar] [CrossRef]
  139. Hesterberg, T.W.; Hart, G.A. Lung Biopersistence and In Vitro Dissolution Rate Predict the Pathogenic Potential of Synthetic Vitreous Fibers. Inhal. Toxicol. 2000, 12 (Suppl. 3), 91–97. [Google Scholar] [CrossRef]
  140. Boyles, M.S.P.; Brown, D.; Knox, J.; Horobin, M.; Miller, M.R.; Johnston, H.J.; Stone, V. Assessing the bioactivity of crystalline silica in heated high-temperature insulation wools. Inhal. Toxicol. 2018, 30, 255–272. [Google Scholar] [CrossRef] [Green Version]
  141. Ellingsen, D.G.; Chashchin, M.; Berlinger, B.; Fedorov, V.; Chashchin, V.; Thomassen, Y. Biological monitoring of welders’ exposure to chromium, molybdenum, tungsten and vanadium. J. Trace Elem. Med. Biol. 2017, 41, 99–106. [Google Scholar] [CrossRef]
  142. Gray, E.P.; Browning, C.L.; Wang, M.; Gion, K.D.; Chao, E.Y.; Koski, K.J.; Kane, A.B.; Hurt, R.H. Biodissolution and cellular response to MoO3 nanoribbons and a new framework for early hazard screening for 2D materials. Environ. Sci. Nano 2018, 5, 2545–2559. [Google Scholar] [CrossRef]
  143. Chazel, V.; Houpert, P.; Paquet, F.; Ansoborlo, E.; Hengé-Napoli, M.H. Experimental Determination of the Solubility of Industrial UF4 Particles. Radiat. Prot. Dosim. 2000, 92, 289–294. [Google Scholar] [CrossRef]
  144. Chazel, V.; Gerasimo, P.; Debouis, V.; Laroche, P.; Paquet, F. Characterisation and dissolution of depleted uranium aerosols produced during impacts of kinetic energy penetrators against a tank. Radiat. Prot. Dosim. 2003, 105, 163–166. [Google Scholar] [CrossRef]
  145. Stefaniak, A.B.; Guilmette, R.A.; Day, G.A.; Hoover, M.D.; Breysse, P.N.; Scripsick, R.C. Characterization of phagolysosomal simulant fluid for study of beryllium aerosol particle dissolution. Toxicol. In Vitro 2005, 19, 123–134. [Google Scholar] [CrossRef]
  146. Heim, K.E.; Danzeisen, R.; Verougstraete, V.; Gaidou, F.; Brouwers, T.; Oller, A.R. Bioaccessibility of nickel and cobalt in synthetic gastric and lung fluids and its potential use in alloy classification. Regul. Toxicol. Pharmacol. 2020, 110, 104549. [Google Scholar] [CrossRef]
  147. Yu, S.-J.; Chao, J.-B.; Sun, J.; Yin, Y.-G.; Liu, J.-F.; Jiang, G.-B. Quantification of the Uptake of Silver Nanoparticles and Ions to HepG2 Cells. Environ. Sci. Technol. 2013, 47, 3268–3274. [Google Scholar] [CrossRef]
  148. Su, C.-K.; Sun, Y.-C. Considerations of inductively coupled plasma mass spectrometry techniques for characterizing the dissolution of metal-based nanomaterials in biological tissues. J. Anal. At. Spectrom. 2015, 30, 1689–1705. [Google Scholar] [CrossRef]
  149. Jiang, X.; Miclăuş, T.; Wang, L.; Foldbjerg, R.; Sutherland, D.; Autrup, H.; Chen, C.; Beer, C. Fast intracellular dissolution and persistent cellular uptake of silver nanoparticles in CHO-K1 cells: Implication for cytotoxicity. Nanotoxicology 2015, 9, 181–189. [Google Scholar] [CrossRef]
  150. Malysheva, A.; Ivask, A.; Doolette, C.L.; Voelcker, N.H.; Lombi, E. Cellular binding, uptake and biotransformation of silver nanoparticles in human T lymphocytes. Nat. Nanotechnol. 2021, 16, 926–932. [Google Scholar] [CrossRef]
  151. Lehuede, P.; de Meringo, A.; Bernstein, D.M. Comparison of the chemical evolution of MMVF following inhalation exposure in rats and acellular in vitro dissolution. Inhal. Toxicol. 1997, 9, 495–523. [Google Scholar] [CrossRef]
  152. Luoto, K.; Holopainen, M.; Karppinen, K.; Perander, M.; Savolainen, K. Dissolution of man-made vitreous fibers in rat alveolar macrophage culture and Gamble’s saline solution: Influence of different media and chemical composition of the fibers. Environ. Health Perspect. 1994, 102 (Suppl. 5), 103–107. [Google Scholar] [CrossRef] [Green Version]
  153. Zhong, L.; Liu, X.; Hu, X.; Chen, Y.; Wang, H.; Lian, H.-Z. In vitro inhalation bioaccessibility procedures for lead in PM2.5 size fraction of soil assessed and optimized by in vivo-in vitro correlation. J. Hazard. Mater. 2020, 381, 121202. [Google Scholar] [CrossRef]
  154. Guldberg, M.; Jensen, S.L.; Knudsen, T.; Steenberg, T.; Kamstrup, O. High-Alumina Low-Silica HT Stone Wool Fibers: A Chemical Compositional Range with High Biosolubility. Regul. Toxicol. Pharmacol. 2002, 35, 217–226. [Google Scholar] [CrossRef]
  155. Takenaka, S.; Karg, E.; Roth, C.; Schulz, H.; Ziesenis, A.; Heinzmann, U.; Schramel, P.; Heyder, J. Pulmonary and systemic distribution of inhaled ultrafine silver particles in rats. Environ. Health Perspect. 2001, 109 (Suppl. 4), 547–551. [Google Scholar] [CrossRef] [Green Version]
  156. Ayres, J.G.; Borm, P.; Cassee, F.R.; Castranova, V.; Donaldson, K.; Ghio, A.; Harrison, R.M.; Hider, R.; Kelly, F.; Kooter, I.M.; et al. Evaluating the Toxicity of Airborne Particulate Matter and Nanoparticles by Measuring Oxidative Stress Potential—A Workshop Report and Consensus Statement. Inhal. Toxicol. 2008, 20, 75–99. [Google Scholar] [CrossRef]
  157. Hellack, B.; Nickel, C.; Albrecht, C.; Kuhlbusch, T.; Boland, S.; Baeza-Squiban, A.; Wohlleben, W.; Schins, R.P.F. Analytical methods to assess the oxidative potential of nanoparticles: A review. Environ. Sci. Nano 2017, 4, 1920–1934. [Google Scholar] [CrossRef]
  158. Møller, P.; Danielsen, P.H.; Karottki, D.G.; Jantzen, K.; Roursgaard, M.; Klingberg, H.; Jensen, D.M.; Vest Christophersen, D.; Hemmingsen, J.G.; Cao, Y.; et al. Oxidative stress and inflammation generated DNA damage by exposure to air pollution particles. Mutat. Res./Rev. Mutat. Res. 2014, 762, 133–166. [Google Scholar] [CrossRef]
  159. Nel, A.; Xia, T.; Mädler, L.; Li, N. Toxic Potential of Materials at the Nanolevel. Science 2006, 311, 622–627. [Google Scholar] [CrossRef] [Green Version]
  160. Møller, P.; Jacobsen, N.R.; Folkmann, J.K.; Danielsen, P.H.; Mikkelsen, L.; Hemmingsen, J.G.; Vesterdal, L.K.; Forchhammer, L.; Wallin, H.; Loft, S. Role of oxidative damage in toxicity of particulates. Free Radic. Res. 2010, 44, 1–46. [Google Scholar] [CrossRef]
  161. Song, B.; Zhou, T.; Yang, W.; Liu, J.; Shao, L. Contribution of oxidative stress to TiO 2 nanoparticle-induced toxicity. Environ. Toxicol. Pharmacol. 2016, 48, 130–140. [Google Scholar] [CrossRef]
  162. Bahl, A.; Hellack, B.; Wiemann, M.; Giusti, A.; Werle, K.; Haase, A.; Wohlleben, W. Nanomaterial categorization by surface reactivity: A case study comparing 35 materials with four different test methods. NanoImpact 2020, 19, 100234. [Google Scholar] [CrossRef]
  163. Gandon, A.; Werle, K.; Neubauer, N.; Wohlleben, W. Surface reactivity measurements as required for grouping and read-across: An advanced FRAS protocol. J. Phys. Conf. Ser. 2017, 838, 012033. [Google Scholar] [CrossRef] [Green Version]
  164. Lu, S.; Duffin, R.; Poland, C.; Daly, P.; Murphy, F.; Drost, E.; MacNee, W.; Stone, V.; Donaldson, K. Efficacy of Simple Short-Term In Vitro Assays for Predicting the Potential of Metal Oxide Nanoparticles to Cause Pulmonary Inflammation. Environ. Health Perspect. 2009, 117, 241–247. [Google Scholar] [CrossRef] [Green Version]
  165. Rogers, E.J.; Hsieh, S.F.; Organti, N.; Schmidt, D.; Bello, D. A high throughput in vitro analytical approach to screen for oxidative stress potential exerted by nanomaterials using a biologically relevant matrix: Human blood serum. Toxicol. In Vitro 2008, 22, 1639–1647. [Google Scholar] [CrossRef] [PubMed]
  166. Sauvain, J.-J.; Rossi, M.J.; Riediker, M. Comparison of Three Acellular Tests for Assessing the Oxidation Potential of Nanomaterials. Aerosol Sci. Technol. 2013, 47, 218–227. [Google Scholar] [CrossRef] [Green Version]
  167. Hsieh, S.-F.; Bello, D.; Schmidt, D.F.; Pal, A.K.; Stella, A.; Isaacs, J.A.; Rogers, E.J. Mapping the Biological Oxidative Damage of Engineered Nanomaterials. Small 2013, 9, 1853–1865. [Google Scholar] [CrossRef] [PubMed]
  168. Achawi, S.; Feneon, B.; Pourchez, J.; Forest, V. Assessing biological oxidative damage induced by graphene-based materials: An asset for grouping approaches using the FRAS assay. Regul. Toxicol. Pharmacol. 2021, 127, 105067. [Google Scholar] [CrossRef] [PubMed]
  169. Benzie, I.F.F.; Strain, J.J. The ferric reducing ability of plasma (FRAP) as a measure of “antioxidant power”: The FRAP assay. Anal. Biochem. 1996, 239, 70–76. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Boyles, M.; Murphy, F.; Mueller, W.; Wohlleben, W.; Jacobsen, N.R.; Braakhuis, H.; Giusti, A.; Stone, V. Development of a standard operating procedure for the DCFH2-DA acellular assessment of reactive oxygen species produced by nanomaterials. Toxicol. Mech. Methods 2022, 32, 439–452. [Google Scholar] [CrossRef]
  171. Pal, A.K.; Bello, D.; Budhlall, B.; Rogers, E.; Milton, D.K. Screening for Oxidative Stress Elicited by Engineered Nanomaterials: Evaluation of Acellular DCFH Assay. Dose-Response 2012, 10, 308–330. [Google Scholar] [CrossRef]
  172. Zhao, J.; Riediker, M. Detecting the oxidative reactivity of nanoparticles: A new protocol for reducing artifacts. J. Nanopart. Res. 2014, 16, 2493. [Google Scholar] [CrossRef] [Green Version]
  173. Seleci, D.A.; Tsiliki, G.; Werle, K.; Elam, D.A.; Okpowe, O.; Seidel, K.; Bi, X.; Westerhoff, P.; Innes, E.; Boyles, M.; et al. Determining nanoform similarity via assessment of surface reactivity by abiotic and in vitro assays. NanoImpact 2022, 26, 100390. [Google Scholar] [CrossRef]
  174. Janer, G.; Landsiedel, R.; Wohlleben, W. Rationale and decision rules behind the ECETOC NanoApp to support registration of sets of similar nanoforms within REACH. Nanotoxicology 2021, 15, 145–166. [Google Scholar] [CrossRef]
  175. Pal, A.K.; Hsieh, S.-F.; Khatri, M.; Isaacs, J.A.; Demokritou, P.; Gaines, P.; Schmidt, D.F.; Rogers, E.J.; Bello, D. Screening for oxidative damage by engineered nanomaterials: A comparative evaluation of FRAS and DCFH. J. Nanopart. Res. 2014, 16, 2167. [Google Scholar] [CrossRef]
  176. Hawkins, C.L.; Davies, M.J. Detection and characterisation of radicals in biological materials using EPR methodology. Biochim. Biophys. Acta (BBA)—Gen. Subj. 2014, 1840, 708–721. [Google Scholar] [CrossRef]
  177. Kalyanaraman, B.; Darley-Usmar, V.; Davies, K.J.A.; Dennery, P.A.; Forman, H.J.; Grisham, M.B.; Mann, G.E.; Moore, K.; Roberts, L.J., II; Ischiropoulos, H. Measuring reactive oxygen and nitrogen species with fluorescent probes: Challenges and limitations. Free Radic. Biol. Med. 2012, 52, 1–6. [Google Scholar] [CrossRef] [Green Version]
  178. Boyles, M.S.P.; Ranninger, C.; Reischl, R.; Rurik, M.; Tessadri, R.; Kohlbacher, O.; Duschl, A.; Huber, C.G. Copper oxide nanoparticle toxicity profiling using untargeted metabolomics. Part. Fibre Toxicol. 2016, 13, 49. [Google Scholar] [CrossRef] [Green Version]
  179. Hiura, T.S.; Li, N.; Kaplan, R.; Horwitz, M.; Seagrave, J.-C.; Nel, A.E. The Role of a Mitochondrial Pathway in the Induction of Apoptosis by Chemicals Extracted from Diesel Exhaust Particles. J. Immunol. 2000, 165, 2703–2711. [Google Scholar] [CrossRef] [Green Version]
  180. Xia, T.; Korge, P.; Weiss, J.N.; Li, N.; Venkatesen, M.I.; Sioutas, C.; Nel, A. Quinones and Aromatic Chemical Compounds in Particulate Matter Induce Mitochondrial Dysfunction: Implications for Ultrafine Particle Toxicity. Environ. Health Perspect. 2004, 112, 1347–1358. [Google Scholar] [CrossRef]
  181. Čapek, J.; Roušar, T. Detection of Oxidative Stress Induced by Nanomaterials in Cells—The Roles of Reactive Oxygen Species and Glutathione. Molecules 2021, 26, 4710. [Google Scholar] [CrossRef]
  182. Rushton, E.K.; Jiang, J.; Leonard, S.S.; Eberly, S.; Castranova, V.; Biswas, P.; Elder, A.; Han, X.; Gelein, R.; Finkelstein, J.; et al. Concept of Assessing Nanoparticle Hazards Considering Nanoparticle Dosemetric and Chemical/Biological Response Metrics. J. Toxicol. Environ. Health A 2010, 73, 445–461. [Google Scholar] [CrossRef]
  183. Lehman, S.E.; Morris, A.S.; Mueller, P.S.; Salem, A.K.; Grassian, V.H.; Larsen, S.C. Silica nanoparticle-generated ROS as a predictor of cellular toxicity: Mechanistic insights and safety by design. Environ. Sci. Nano 2016, 3, 56–66. [Google Scholar] [CrossRef] [Green Version]
  184. Driessen, M.D.; Mues, S.; Vennemann, A.; Hellack, B.; Bannuscher, A.; Vimalakanthan, V.; Riebeling, C.; Ossig, R.; Wiemann, M.; Schnekenburger, J.; et al. Proteomic analysis of protein carbonylation: A useful tool to unravel nanoparticle toxicity mechanisms. Part. Fibre Toxicol. 2015, 12, 36. [Google Scholar] [CrossRef] [Green Version]
  185. Riebeling, C.; Wiemann, M.; Schnekenburger, J.; Kuhlbusch, T.A.J.; Wohlleben, W.; Luch, A.; Haase, A. A redox proteomics approach to investigate the mode of action of nanomaterials. Toxicol. Appl. Pharmacol. 2016, 299, 24–29. [Google Scholar] [CrossRef] [PubMed]
  186. Reuzel, P.G.J.; Bruijntjes, J.P.; Feron, V.J.; Woutersen, R.A. Subchronic inhalation toxicity of amorphous silicas and quartz dust in rats. Food Chem. Toxicol. 1991, 29, 341–354. [Google Scholar] [CrossRef]
  187. Muhle, H.; Kittel, B.; Ernst, H.; Mohr, U.; Mermelstein, R. Neoplastic lung lesions in rat after chronic exposure to crystalline silica. Scand. J. Work Environ. Health 1995, 21, 27–29. [Google Scholar]
  188. Brand, W.; Peters, R.J.B.; Braakhuis, H.M.; Maślankiewicz, L.; Oomen, A.G. Possible effects of titanium dioxide particles on human liver, intestinal tissue, spleen and kidney after oral exposure. Nanotoxicology 2020, 14, 985–1007. [Google Scholar] [CrossRef]
  189. Elsabahy, M.; Wooley, K.L. Cytokines as biomarkers of nanoparticle immunotoxicity. Chem. Soc. Rev. 2013, 42, 5552–5576. [Google Scholar] [CrossRef] [Green Version]
  190. Di Ianni, E.; Erdem, J.S.; Møller, P.; Sahlgren, N.M.; Poulsen, S.S.; Knudsen, K.B.; Zienolddiny, S.; Saber, A.T.; Wallin, H.; Vogel, U.; et al. In vitro-in vivo correlations of pulmonary inflammogenicity and genotoxicity of MWCNT. Part. Fibre Toxicol. 2021, 18, 25. [Google Scholar] [CrossRef]
  191. Sun, B.; Wang, X.; Ji, Z.; Li, R.; Xia, T. NLRP3 Inflammasome Activation Induced by Engineered Nanomaterials. Small 2013, 9, 1595–1607. [Google Scholar] [CrossRef]
  192. Harkema, J.R.; Wagner, J.G. Pathology of the Respiratory System. In Toxicologic Pathology for Non-Pathologists; Humana: New York, NY, USA, 2019; pp. 311–354. [Google Scholar] [CrossRef]
  193. Li, Y.; Fujita, M.; Boraschi, D. Endotoxin Contamination in Nanomaterials Leads to the Misinterpretation of Immunosafety Results. Front. Immunol. 2017, 8, 472. [Google Scholar] [CrossRef]
  194. Dobrovolskaia, M.A.; Germolec, D.R.; Weaver, J.L. Evaluation of nanoparticle immunotoxicity. Nat. Nanotechnol. 2009, 4, 411–414. [Google Scholar] [CrossRef]
  195. Braakhuis, H.M.; Gremmer, E.R.; Bannuscher, A.; Drasler, B.; Keshavan, S.; Barbara Rothen-Rutishauser, B.B.; Verlohner, A.; Landsiedel, R.; Meldrum, K.; Doak, S.H.; et al. Transferability and reproducibility of exposed air-liquid interface co-culture lung models. NanoImpact, 2022; in preparation. [Google Scholar]
  196. Barosova, H.; Meldrum, K.; Karakocak, B.B.; Balog, S.; Doak, S.H.; Petri-Fink, A.; Clift, M.J.D.; Rothen-Rutishauser, B. Inter-laboratory variability of A549 epithelial cells grown under submerged and air-liquid interface conditions. Toxicol. In Vitro 2021, 75, 105178. [Google Scholar] [CrossRef]
  197. Bessa, M.J.; Brandão, F.; Fokkens, P.; Cassee, F.R.; Salmatonidis, A.; Viana, M.; Vulpoi, A.; Simon, S.; Monfort, E.; Teixeira, J.P.; et al. Toxicity assessment of industrial engineered and airborne process-generated nanoparticles in a 3D human airway epithelial in vitro model. Nanotoxicology 2021, 15, 542–557. [Google Scholar] [CrossRef]
  198. Petersen, E.J.; Sharma, M.; Clippinger, A.J.; Gordon, J.; Katz, A.; Laux, P.; Leibrock, L.B.; Luch, A.; Matheson, J.; Stucki, A.O.; et al. Use of Cause-and-Effect Analysis to Optimize the Reliability of In Vitro Inhalation Toxicity Measurements Using an Air–Liquid Interface. Chem. Res. Toxicol. 2021, 34, 1370–1385. [Google Scholar] [CrossRef]
  199. Guo, C.; Buckley, A.; Marczylo, T.; Seiffert, J.; Römer, I.; Warren, J.; Hodgson, A.; Chung, K.F.; Gant, T.; Smith, R.; et al. The small airway epithelium as a target for the adverse pulmonary effects of silver nanoparticle inhalation. Nanotoxicology 2018, 12, 539–553. [Google Scholar] [CrossRef] [Green Version]
  200. Sandberg, W.J.; Låg, M.; Holme, J.A.; Friede, B.; Gualtieri, M.; Kruszewski, M.; Schwarze, P.E.; Skuland, T.; Refsnes, M. Comparison of non-crystalline silica nanoparticles in IL-1β release from macrophages. Part. Fibre Toxicol. 2012, 9, 32. [Google Scholar] [CrossRef] [Green Version]
  201. Svensson, C.R.; Ameer, S.S.; Ludvigsson, L.; Ali, N.; Alhamdow, A.; Messing, M.E.; Pagels, J.; Gudmundsson, A.; Bohgard, M.; Sanfins, E.; et al. Validation of an air–liquid interface toxicological set-up using Cu, Pd, and Ag well-characterized nanostructured aggregates and spheres. J. Nanopart. Res. 2016, 18, 86. [Google Scholar] [CrossRef] [Green Version]
  202. Endes, C.; Schmid, O.; Kinnear, C.; Mueller, S.; Camarero-Espinosa, S.; Vanhecke, D.; Foster, E.J.; Petri-Fink, A.; Rothen-Rutishauser, B.; Weder, C.; et al. An in vitro testing strategy towards mimicking the inhalation of high aspect ratio nanoparticles. Part. Fibre Toxicol. 2014, 11, 40. [Google Scholar] [CrossRef] [Green Version]
  203. Hufnagel, M.; Neuberger, R.; Wall, J.; Link, M.; Friesen, A.; Hartwig, A. Impact of Differentiated Macrophage-Like Cells on the Transcriptional Toxicity Profile of CuO Nanoparticles in Co-Cultured Lung Epithelial Cells. Int. J. Mol. Sci. 2021, 22, 5044. [Google Scholar] [CrossRef]
  204. Clift, M.J.D.; Endes, C.; Vanhecke, D.; Wick, P.; Gehr, P.; Schins, R.P.F.; Petri-Fink, A.; Rothen-Rutishauser, B. A Comparative Study of Different In Vitro Lung Cell Culture Systems to Assess the Most Beneficial Tool for Screening the Potential Adverse Effects of Carbon Nanotubes. Toxicol. Sci. 2014, 137, 55–64. [Google Scholar] [CrossRef]
  205. Braakhuis, H.M.; He, R.; Vandebriel, R.J.; Gremmer, E.R.; Zwart, E.; Vermeulen, J.P.; Fokkens, P.; Boere, J.; Gosens, I.; Cassee, F.R. An Air-liquid Interface Bronchial Epithelial Model for Realistic, Repeated Inhalation Exposure to Airborne Particles for Toxicity Testing. J. Vis. Exp. 2020, 159, e61210. [Google Scholar] [CrossRef]
  206. Herzog, F.; Clift, M.J.D.; Piccapietra, F.; Behra, R.; Schmid, O.; Petri-Fink, A.; Rothen-Rutishauser, B. Exposure of silver-nanoparticles and silver-ions to lung cells in vitro at the air-liquid interface. Part. Fibre Toxicol. 2013, 10, 11. [Google Scholar] [CrossRef] [Green Version]
  207. Braakhuis, H.M.; Giannakou, C.; Peijnenburg, W.J.G.M.; Vermeulen, J.; van Loveren, H.; Park, M.V.D.Z. Simplein vitromodels can predict pulmonary toxicity of silver nanoparticles. Nanotoxicology 2016, 10, 770–779. [Google Scholar] [CrossRef]
  208. Li, R.; Wang, X.; Ji, Z.; Sun, B.; Zhang, H.; Chang, C.H.; Lin, S.; Meng, H.; Liao, Y.-P.; Wang, M.; et al. Surface Charge and Cellular Processing of Covalently Functionalized Multiwall Carbon Nanotubes Determine Pulmonary Toxicity. ACS Nano 2013, 7, 2352–2368. [Google Scholar] [CrossRef] [Green Version]
  209. Wang, X.; Xia, T.; Ntim, S.A.; Ji, Z.; Lin, S.; Meng, H.; Chung, C.-H.; George, S.; Zhang, H.; Wang, M.; et al. Dispersal State of Multiwalled Carbon Nanotubes Elicits Profibrogenic Cellular Responses That Correlate with Fibrogenesis Biomarkers and Fibrosis in the Murine Lung. ACS Nano 2011, 5, 9772–9787. [Google Scholar] [CrossRef] [Green Version]
  210. Diabaté, S.; Armand, L.; Murugadoss, S.; Dilger, M.; Fritsch-Decker, S.; Schlager, C.; Béal, D.; Arnal, M.-E.; Biola-Clier, M.; Ambrose, S.; et al. Air–Liquid Interface Exposure of Lung Epithelial Cells to Low Doses of Nanoparticles to Assess Pulmonary Adverse Effects. Nanomaterials 2020, 11, 65. [Google Scholar] [CrossRef]
  211. Loret, T.; Rogerieux, F.; Trouiller, B.; Braun, A.; Egles, C.; Lacroix, G. Predicting the in vivo pulmonary toxicity induced by acute exposure to poorly soluble nanomaterials by using advanced in vitro methods. Part. Fibre Toxicol. 2018, 15, 25. [Google Scholar] [CrossRef]
  212. Lenz, A.-G.; Karg, E.; Brendel, E.; Hinze-Heyn, H.; Maier, K.L.; Eickelberg, O.; Stoeger, T.; Schmid, O. Inflammatory and Oxidative Stress Responses of an Alveolar Epithelial Cell Line to Airborne Zinc Oxide Nanoparticles at the Air-Liquid Interface: A Comparison with Conventional, Submerged Cell-Culture Conditions. BioMed Res. Int. 2013, 2013, 652632. [Google Scholar] [CrossRef] [Green Version]
  213. Loret, T.; Peyret, E.; Dubreuil, M.; Aguerre-Chariol, O.; Bressot, C.; le Bihan, O.; Amodeo, T.; Trouiller, B.; Braun, A.; Egles, C.; et al. Air-liquid interface exposure to aerosols of poorly soluble nanomaterials induces different biological activation levels compared to exposure to suspensions. Part. Fibre Toxicol. 2016, 13, 58. [Google Scholar] [CrossRef] [Green Version]
  214. Herzog, F.; Loza, K.; Balog, S.; Clift, M.J.D.; Epple, M.; Gehr, P.; Petri-Fink, A.; Rothen-Rutishauser, B. Mimicking exposures to acute and lifetime concentrations of inhaled silver nanoparticles by two different in vitro approaches. Beilstein J. Nanotechnol. 2014, 5, 1357–1370. [Google Scholar] [CrossRef] [Green Version]
  215. Medina-Reyes, E.I.; Delgado-Buenrostro, N.L.; Leseman, D.L.; Déciga-Alcaraz, A.; He, R.; Gremmer, E.R.; Fokkens, P.H.B.; Flores-Flores, J.O.; Cassee, F.R.; Chirino, Y.I. Differences in cytotoxicity of lung epithelial cells exposed to titanium dioxide nanofibers and nanoparticles: Comparison of air-liquid interface and submerged cell cultures. Toxicol. In Vitro 2020, 65, 104798. [Google Scholar] [CrossRef]
  216. Panas, A.; Comouth, A.; Saathoff, H.; Leisner, T.; Al-Rawi, M.; Simon, M.; Seemann, G.; Dössel, O.; Mülhopt, S.; Paur, H.-R.; et al. Silica nanoparticles are less toxic to human lung cells when deposited at the air–liquid interface compared to conventional submerged exposure. Beilstein J. Nanotechnol. 2014, 5, 1590–1602. [Google Scholar] [CrossRef] [Green Version]
  217. Öhlinger, K.; Kolesnik, T.; Meindl, C.; Gallé, B.; Absenger-Novak, M.; Kolb-Lenz, D.; Fröhlich, E. Air-liquid interface culture changes surface properties of A549 cells. Toxicol. In Vitro 2019, 60, 369–382. [Google Scholar] [CrossRef]
  218. Upadhyay, S.; Palmberg, L. Air-Liquid Interface: Relevant In Vitro Models for Investigating Air Pollutant-Induced Pulmonary Toxicity. Toxicol. Sci. 2018, 164, 21–30. [Google Scholar] [CrossRef] [Green Version]
  219. Bannuscher, A.; Schmid, O.; Drasler, B.; Rohrbasser, A.; Braakhuis, H.M.; Meldrum, K.; Zwart, E.P.; Gremmer, E.R.; Birk, B.; Rissel, M.; et al. An inter-laboratory effort to harmonize the cell-delivered in vitro dose of aerosolized materials. NanoImpact 2022, 28, 100439. [Google Scholar] [CrossRef]
  220. Doak, S.H.; Clift, M.J.; Costa, A.; Delmaar, C.; Gosens, I.; Halappanavar, S.; Kelly, S.; Pejinenburg, W.J.; Rothen-Rutishauser, B.; Schins, R.P. The Road to Achieving the European Commission’s Chemicals Strategy for Nanomaterial Sustainability—A PATROLS Perspective on New Approach Methodologies. Small 2022, 18, 2200231. [Google Scholar] [CrossRef]
  221. Catalán, J.; Stockmann-Juvala, H.; Norppa, H. A theoretical approach for a weighted assessment of the mutagenic potential of nanomaterials. Nanotoxicology 2017, 11, 964–977. [Google Scholar] [CrossRef]
  222. Kohl, Y.; Rundén-Pran, E.; Mariussen, E.; Hesler, M.; El Yamani, N.; Longhin, E.M.; Dusinska, M. Genotoxicity of Nanomaterials: Advanced In Vitro Models and High Throughput Methods for Human Hazard Assessment—A Review. Nanomaterials 2020, 10, 1911. [Google Scholar] [CrossRef]
  223. Hartwig, A.; Arand, M.; Epe, B.; Guth, S.; Jahnke, G.; Lampen, A.; Martus, H.-J.; Monien, B.; Rietjens, I.M.C.M.; Schmitz-Spanke, S.; et al. Mode of action-based risk assessment of genotoxic carcinogens. Arch. Toxicol. 2020, 94, 1787–1877. [Google Scholar] [CrossRef]
  224. Steiblen, G.; van Benthem, J.; Johnson, G. Strategies in genotoxicology: Acceptance of innovative scientific methods in a regulatory context and from an industrial perspective. Mutat. Res./Genet. Toxicol. Environ. Mutagen. 2020, 853, 503171. [Google Scholar] [CrossRef]
  225. Catalán, J.; Norppa, H. Safety Aspects of Bio-Based Nanomaterials. Bioengineering 2017, 4, 94. [Google Scholar] [CrossRef] [Green Version]
  226. Stone, V.; Pozzi-Mucelli, S.; Tran, L.; Aschberger, K.; Sabella, S.; Vogel, U.; Poland, C.; Balharry, D.; Fernandes, T.; Gottardo, S.; et al. ITS-NANO—Prioritising nanosafety research to develop a stakeholder driven intelligent testing strategy. Part. Fibre Toxicol. 2014, 11, 9. [Google Scholar] [CrossRef] [Green Version]
  227. Dusinska, M.; Boland, S.; Saunders, M.; Juillerat-Jeanneret, L.; Tran, L.; Pojana, G.; Marcomini, A.; Volkovova, K.; Tulinska, J.; Knudsen, L.E.; et al. Towards an alternative testing strategy for nanomaterials used in nanomedicine: Lessons from NanoTEST. Nanotoxicology 2015, 9, 118–132. [Google Scholar] [CrossRef] [Green Version]
  228. Kirkland, D.; Reeve, L.; Gatehouse, D.; Vanparys, P. A core in vitro genotoxicity battery comprising the Ames test plus the in vitro micronucleus test is sufficient to detect rodent carcinogens and in vivo genotoxins. Mutat. Res./Genet. Toxicol. Environ. Mutagen. 2011, 721, 27–73. [Google Scholar] [CrossRef]
  229. Kirkland, D.; Aardema, M.; Henderson, L.; Müller, L. Evaluation of the ability of a battery of three in vitro genotoxicity tests to discriminate rodent carcinogens and non-carcinogens I. Sensitivity, specificity and relative predictivity. Mutat. Res./Genet. Toxicol. Environ. Mutagen. 2005, 584, 1–256. [Google Scholar] [CrossRef]
  230. Doak, S.H.; Manshian, B.; Jenkins, G.J.S.; Singh, N. In vitro genotoxicity testing strategy for nanomaterials and the adaptation of current OECD guidelines. Mutat. Res. Toxicol. Environ. Mutagen. 2012, 745, 104–111. [Google Scholar] [CrossRef]
  231. Clift, M.J.D.; Raemy, D.O.; Endes, C.; Ali, Z.; Lehmann, A.D.; Brandenberger, C.; Petri-Fink, A.; Wick, P.; Parak, W.J.; Gehr, P.; et al. Can the Ames test provide an insight into nano-object mutagenicity? Investigating the interaction between nano-objects and bacteria. Nanotoxicology 2013, 7, 1373–1385. [Google Scholar] [CrossRef]
  232. George, J.M.; Magogotya, M.; Vetten, M.A.; Buys, A.V.; Gulumian, M. From the cover: An investigation of the genotoxicity and interference of gold nanoparticles in commonly used in vitro mutagenicity and genotoxicity assays. Toxicol. Sci. 2017, 156, 149–166. [Google Scholar]
  233. Landsiedel, R.; Kapp, M.D.; Schulz, M.; Wiench, K.; Oesch, F. Genotoxicity investigations on nanomaterials: Methods, preparation and characterization of test material, potential artifacts and limitations—Many questions, some answers. Mutat. Res./Rev. Mutat. Res. 2009, 681, 241–258. [Google Scholar] [CrossRef]
  234. Magdolenova, Z.; Collins, A.; Kumar, A.; Dhawan, A.; Stone, V.; Dusinska, M. Mechanisms of genotoxicity. A review of in vitro and in vivo studies with engineered nanoparticles. Nanotoxicology 2014, 8, 233–278. [Google Scholar] [CrossRef]
  235. Pfuhler, S.; Elespuru, R.; Aardema, M.J.; Doak, S.H.; Maria Donner, E.; Honma, M. Genotoxicity of nanomaterials: Refining strategies and tests for hazard identification. Environ. Mol. Mutagen. 2013, 54, 229. [Google Scholar] [CrossRef]
  236. Elespuru, R.; Pfuhler, S.; Aardema, M.J.; Chen, T.; Doak, S.H.; Doherty, A.; Farabaugh, C.S.; Kenny, J.; Manjanatha, M.; Mahadevan, B.; et al. Genotoxicity Assessment of Nanomaterials: Recommendations on Best Practices, Assays, and Methods. Toxicol. Sci. 2018, 164, 391–416. [Google Scholar] [CrossRef]
  237. Elespuru, R.K.; Doak, S.H.; Collins, A.R.; Dusinska, M.; Pfuhler, S.; Manjanatha, M.; Cardoso, R.; Chen, C.L. Common Considerations for Genotoxicity Assessment of Nanomaterials. Front. Toxicol. 2022, 4, 859122. [Google Scholar] [CrossRef] [PubMed]
  238. Fenech, M.; Morley, A.A. Cytokinesis-block micronucleus method in human lymphocytes: Effect of in vivo ageing and low dose X-irradiation. Mutat. Res./Fundam. Mol. Mech. Mutagen. 1986, 161, 193–198. [Google Scholar] [CrossRef] [PubMed]
  239. Doak, S.; Griffiths, S.; Manshian, B.; Singh, N.; Williams, P.; Brown, A.; Jenkins, G. Confounding experimental considerations in nanogenotoxicology. Mutagenesis 2009, 24, 285–293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  240. ECHA. Guidance on Information Requirements and Chemical Safety Assessment; Appendix R7-1 for Nanomaterials Applicable to Chapter R7a Endpoint Specific Guidance ECHA-21-H-04-EN; ECHA: Helsinki, Finland, 2021. [Google Scholar]
  241. Dusinska, M.; Mariussen, E.; Rundén-Pran, E.; Hudecova, A.M.; Elje, E.; Kazimirova, A.; El Yamani, N.; Dommershausen, N.; Tharmann, J.; Fieblinger, D.; et al. In Vitro Approaches for Assessing the Genotoxicity of Nanomaterials. In Nanotoxicity; Methods in Molecular Biology; Springer: Cham, Switzerland, 2019; Volume 1894, pp. 83–122. [Google Scholar] [CrossRef]
  242. Wills, J.W.; Hondow, N.; Thomas, A.D.; Chapman, K.E.; Fish, D.; Maffeis, T.G.; Penny, M.W.; Brown, R.A.; Jenkins, G.J.S.; Brown, A.P.; et al. Genetic toxicity assessment of engineered nanoparticles using a 3D in vitro skin model (EpiDerm™). Part. Fibre Toxicol. 2016, 13, 50. [Google Scholar] [CrossRef] [Green Version]
  243. Conway, G.E.; Shah, U.-K.; Llewellyn, S.; Cervena, T.; Evans, S.J.; Al Ali, A.S.; Jenkins, G.J.; Clift, M.J.D.; Doak, S.H. Adaptation of the in vitro micronucleus assay for genotoxicity testing using 3D liver models supporting longer-term exposure durations. Mutagenesis 2020, 35, 319–330. [Google Scholar] [CrossRef]
  244. ANSES. Nanogenotox Final Report—Facilitating the Safety Evaluation of Manufactured Nanomaterials by Characterising Their Potential Genotoxic Hazard; ANSES: Buenos Aires, Argentina, 2013. [Google Scholar]
  245. Louro, H.; Pinhão, M.; Santos, J.; Tavares, A.; Vital, N.; Silva, M.J. Evaluation of the cytotoxic and genotoxic effects of benchmark multi-walled carbon nanotubes in relation to their physicochemical properties. Toxicol. Lett. 2016, 262, 123–134. [Google Scholar] [CrossRef]
  246. Evans, S.J.; Clift, M.J.D.; Singh, N.; Wills, J.W.; Hondow, N.; Wilkinson, T.S.; Burgum, M.J.; Brown, A.P.; Jenkins, G.J.; Doak, S.H. In vitro detection of in vitro secondary mechanisms of genotoxicity induced by engineered nanomaterials. Part. Fibre Toxicol. 2019, 16, 8. [Google Scholar] [CrossRef] [Green Version]
  247. Burgum, M.J.; Clift, M.J.D.; Evans, S.J.; Hondow, N.; Tarat, A.; Jenkins, G.J.; Doak, S.H. Few-layer graphene induces both primary and secondary genotoxicity in epithelial barrier models in vitro. J. Nanobiotechnol. 2021, 19, 24. [Google Scholar] [CrossRef]
  248. SCOEL. Methodology for Derivation of Occupational Exposure Limits of Chemical Agents; European Commission, Ed.; SCOEL: Brussels, Belgium, 2017. [Google Scholar]
  249. Decordier, I.; Papine, A.; Loock, K.V.; Plas, G.; Soussaline, F.; Kirsch-Volders, M. Automated image analysis of micronuclei by IMSTAR for biomonitoring. Mutagenesis 2011, 26, 163–168. [Google Scholar] [CrossRef] [Green Version]
  250. Nüsse, M.; Kramer, J. Flow cytometric analysis of micronuclei found in cells after irradiation. Cytometry 1984, 5, 20–25. [Google Scholar] [CrossRef]
  251. Nüsse, M.; Marx, K. Flow cytometric analysis of micronuclei in cell cultures and human lymphocytes: Advantages and disadvantages. Mutat. Res./Genet. Toxicol. Environ. Mutagen. 1997, 392, 109–115. [Google Scholar] [CrossRef] [PubMed]
  252. Nüsse, M.; Recknagel, S.; Beisker, W. Micronuclei induced by 2-chlorobenzylidene malonitrile contain single chromosomes as demonstrated by the combined use of flow cytometry and immunofluorescent staining with anti-kinetochore antibodies. Mutagenesis 1992, 7, 57–67. [Google Scholar] [CrossRef] [PubMed]
  253. Franz, P.; Bürkle, A.; Wick, P.; Hirsch, C. Exploring Flow Cytometry-Based Micronucleus Scoring for Reliable Nanomaterial Genotoxicity Assessment. Chem. Res. Toxicol. 2020, 33, 2538–2549. [Google Scholar] [CrossRef]
  254. Azqueta, A.; Gutzkow, K.B.; Priestley, C.C.; Meier, S.; Walker, J.S.; Brunborg, G.; Collins, A.R. A comparative performance test of standard, medium- and high-throughput comet assays. Toxicol. In Vitro 2013, 27, 768–773. [Google Scholar] [CrossRef] [PubMed]
  255. García-Rodríguez, A.; Rubio, L.; Vila, L.; Xamena, N.; Velázquez, A.; Marcos, R.; Hernández, A. The Comet Assay as a Tool to Detect the Genotoxic Potential of Nanomaterials. Nanomaterials 2019, 9, 1385. [Google Scholar] [CrossRef] [Green Version]
  256. Nelson, B.C.; Wright, C.W.; Ibuki, Y.; Moreno-Villanueva, M.; Karlsson, H.L.; Hendriks, G.; Sims, C.M.; Singh, N.; Doak, S.H. Emerging Metrology for High-Throughput Nanomaterial Genotoxicology Mutagenesis Advance Access. Mutagenesis 2017, 32, 215. [Google Scholar] [CrossRef] [Green Version]
  257. Elje, E.; Mariussen, E.; Moriones, O.H.; Bastús, N.G.; Puntes, V.; Kohl, Y.; Dusinska, M.; Rundén-Pran, E. Hepato(Geno)Toxicity Assessment of Nanoparticles in a HepG2 Liver Spheroid Model. Nanomaterials 2020, 10, 545. [Google Scholar] [CrossRef] [Green Version]
  258. Rodriguez-Garraus, A.; Azqueta, A.; Vettorazzi, A.; de Cerain, A.L. Genotoxicity of Silver Nanoparticles. Nanomaterials 2020, 10, 251. [Google Scholar] [CrossRef] [Green Version]
  259. Warheit, D.B.; Donner, E.M. Rationale of genotoxicity testing of nanomaterials: Regulatory requirements and appropriateness of available OECD test guidelines. Nanotoxicology 2010, 4, 409–413. [Google Scholar] [CrossRef]
  260. Pfuhler, S.; van Benthem, J.; Curren, R.; Doak, S.H.; Dusinska, M.; Hayashi, M.; Heflich, R.H.; Kidd, D.; Kirkland, D.; Luan, Y.; et al. Use of in vitro 3D tissue models in genotoxicity testing: Strategic fit, validation status and way forward. Report of the working group from the 7th International Workshop on Genotoxicity Testing (IWGT). Mutat. Res./Genet. Toxicol. Environ. Mutagen. 2020, 850–851, 503135. [Google Scholar] [CrossRef]
  261. Evans, S.J.; Clift, M.J.; Singh, N.; de Oliveira Mallia, J.; Burgum, M.; Wills, J.W.; Wilkinson, T.S.; Jenkins, G.J.; Doak, S.H. Critical review of the current and future challenges associated with advanced in vitro systems towards the study of nanoparticle (secondary) genotoxicity. Mutagenesis 2017, 32, 233–241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  262. Di Ianni, E.; Jacobsen, N.R.; Vogel, U.; Møller, P. Predicting nanomaterials pulmonary toxicity in animals by cell culture models: Achievements and perspectives. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2022, 14, e1794. [Google Scholar] [CrossRef]
  263. Clewell, R.A.; McMullen, P.D.; Adeleye, Y.; Carmichael, P.L.; Andersen, M.E. Pathway based toxicology and fit-for-purpose assays. In Validation of Alternative Methods for Toxicity Testing; Springer: Cham, Switzerland, 2016; pp. 205–230. [Google Scholar]
  264. Nymark, P.; Bakker, M.; Dekkers, S.; Franken, R.; Fransman, W.; García-Bilbao, A.; Greco, D.; Gulumian, M.; Hadrup, N.; Halappanavar, S.; et al. Toward Rigorous Materials Production: New Approach Methodologies Have Extensive Potential to Improve Current Safety Assessment Practices. Small 2020, 16, e1904749. [Google Scholar] [CrossRef] [PubMed]
  265. OECD. Guidance Document on the Validation and International Acceptance of New or Updated Test Methods for Hazard Assessment; OECD Environment, Health and Safety Publications Series on Testing and Assessment; OECD: Paris, France, 2015. [Google Scholar]
  266. Clift, M.J.D.; Jenkins, G.J.S.; Doak, S.H. An Alternative Perspective towards Reducing the Risk of Engineered Nanomaterials to Human Health. Small 2020, 16, e2002002. [Google Scholar] [CrossRef] [PubMed]
  267. Van der Zalm, A.J.; Barroso, J.; Browne, P.; Casey, W.; Gordon, J.; Henry, T.R.; Kleinstreuer, N.C.; Lowit, A.B.; Perron, M.; Clippinger, A.J. A framework for establishing scientific confidence in new approach methodologies. Arch. Toxicol. 2022, 96, 2865–2879. [Google Scholar] [CrossRef]
  268. European Commission. Pathway to a Healthy Planet for All. EU Action Plan: ’Towards Zero Pollution for Air, Water and Soil’—Communication from the Commission to the European Parliament, the Council, the European Economic and Social Committee and the Committee of the Regions; European Commission: Brussels, Belgium, 2021. [Google Scholar]
  269. Napierska, D.; Thomassen, L.C.J.; Lison, D.; Martens, J.A.; Hoet, P.H. The nanosilica hazard: Another variable entity. Part. Fibre Toxicol. 2010, 7, 39. [Google Scholar] [CrossRef] [Green Version]
  270. IARC. Arsenic, metals, fibres, and dusts. IARC Monogr. Eval. Carcinog. Risks Hum. 2012, 100, 11–465. [Google Scholar]
  271. Höck, J.; Behra, R.; Bergamin, L.; Bourqui-Pittet, M.; Bosshard, C.; Epprecht, T.; Furrer, V.; Frey, S.; Gautschi, M.; Hofmann, H.; et al. Guidelines on the Precautionary Matrix for Synthetic Nanomaterials; Federal Office of Public Health: Bern, Switzerland, 2018. [Google Scholar]
  272. Pavlicek, A.; Part, F.; Gressler, S.; Rose, G.; Gazsó, A.; Ehmoser, E.-K.; Huber-Humer, M. Testing the Applicability of the Safe-by-Design Concept: A Theoretical Case Study Using Polymer Nanoclay Composites for Coffee Capsules. Sustainability 2021, 13, 13951. [Google Scholar] [CrossRef]
  273. OECD. Testing Programme of Manufactured Nanomaterials. Available online: https://www.oecd.org/chemicalsafety/nanosafety/testing-programme-manufactured-nanomaterials.htm (accessed on 14 November 2022).
  274. NanoSafetyCluster. The Malta Initiative. Available online: https://www.nanosafetycluster.eu/international-cooperation/the-malta-initiative/ (accessed on 14 November 2022).
  275. NanoHarmony. Available online: https://nanoharmony.eu/ (accessed on 14 November 2022).
  276. OECD. Nanomet. Available online: https://www.oecd.org/chemicalsafety/nanomet/ (accessed on 14 November 2022).
  277. Gov4Nano. Available online: https://www.gov4nano.eu/ (accessed on 14 November 2022).
  278. EFSA; Andersson, N.; Arena, M.; Auteri, D.; Barmaz, S.; Grignard, E.; Kienzler, A.; Lepper, P.; Lostia, A.M.; Munn, S. Guidance for the identification of endocrine disruptors in the context of Regulations (EU) No 528/2012 and (EC) No 1107/2009. EFSA J. 2018, 16, e05311. [Google Scholar]
  279. Shen, F.; Li, D.; Guo, J.; Chen, J. Mechanistic toxicity assessment of differently sized and charged polystyrene nanoparticles based on human placental cells. Water Res. 2022, 223, 118960. [Google Scholar] [CrossRef]
  280. Priyam, A.; Singh, P.P.; Gehlout, S. Role of Endocrine-Disrupting Engineered Nanomaterials in the Pathogenesis of Type 2 Diabetes Mellitus. Front. Endocrinol. 2018, 9, 704. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Considerations for assay selection for SbD hazard testing.
Figure 1. Considerations for assay selection for SbD hazard testing.
Nanomaterials 13 00472 g001
Figure 2. Overview of aspects that might have to be considered when performing SbD hazard testing, showing that simple testing can be challenging to achieve.
Figure 2. Overview of aspects that might have to be considered when performing SbD hazard testing, showing that simple testing can be challenging to achieve.
Nanomaterials 13 00472 g002
Figure 3. Visual representation of two NMs with different properties, resulting in different doses delivered to the cells, when administered doses are equal.
Figure 3. Visual representation of two NMs with different properties, resulting in different doses delivered to the cells, when administered doses are equal.
Nanomaterials 13 00472 g003
Figure 4. Submerged (left) and ALI exposures (right) to NMs. Submerged exposures are considered easier, whereas ALI exposures are considered more physiologically relevant for inhalation (and dermal and intestinal) exposures.
Figure 4. Submerged (left) and ALI exposures (right) to NMs. Submerged exposures are considered easier, whereas ALI exposures are considered more physiologically relevant for inhalation (and dermal and intestinal) exposures.
Nanomaterials 13 00472 g004
Figure 5. The balance of SbD hazard testing. SbD aims to address safety at an early stage in the product development process. On the one hand, SbD tries to be comprehensive to address all concerns, while on the other hand the approach should be simple.
Figure 5. The balance of SbD hazard testing. SbD aims to address safety at an early stage in the product development process. On the one hand, SbD tries to be comprehensive to address all concerns, while on the other hand the approach should be simple.
Nanomaterials 13 00472 g005
Figure 6. Factors that became evident throughout this review that are crucial for putting SbD hazard testing into practice. Protocol standardization is key for SbD hazard testing, as well as for better understanding structure–activity relationships and prediction accuracies of assays.
Figure 6. Factors that became evident throughout this review that are crucial for putting SbD hazard testing into practice. Protocol standardization is key for SbD hazard testing, as well as for better understanding structure–activity relationships and prediction accuracies of assays.
Nanomaterials 13 00472 g006
Table 1. Evaluation of suitability of cytotoxicity assays for SbD hazard testing.
Table 1. Evaluation of suitability of cytotoxicity assays for SbD hazard testing.
Performance CriteriaMitochondrial Activity (MTT, MTS, XTT, WST-1, Alamar Blue)Cell Membrane Integrity (LDH)Cell Membrane Integrity Staining (Trypan Blue, Propidium Iodide, Annexin V)Lysosomal Integrity (Neutral Red Uptake)Caspase 3/7 Assay
Simplicity and costNanomaterials 13 00472 i001Easy and cost-effective, commercial kits available.Nanomaterials 13 00472 i001Easy and cost-effective, commercial kits available.Nanomaterials 13 00472 i001Microscopic evaluation is time-consuming. Using flow cytometry increases time efficiency.Nanomaterials 13 00472 i001Easy and cost-effective, commercial kits available.Nanomaterials 13 00472 i001Easy and cost-effective, commercial kits available.
Predictivity (Sensitivity and Specificity)Nanomaterials 13 00472 i001Nanomaterials 13 00472 i002Depends on the mechanism of toxicity of the particle, and the cell type used [110]. Macrophages seem more sensitive [114,117]. Assay better equipped to detect cytotoxicity of ion-shedding NMs [100]. Possibly suitable for making accurate rankings in toxicity [114]. Nanomaterials 13 00472 i001Nanomaterials 13 00472 i002Depends on the mechanism of toxicity of the particle, and the cell type used. LDH results have been shown to correlate with in vivo results for ion shedding NMs [112] as well as poorly soluble NMs [113]. Nanomaterials 13 00472 i003Not assessed for NMs specifically.Nanomaterials 13 00472 i003Not assessed for NMs specificallyNanomaterials 13 00472 i003Not assessed for NMs specifically.
RobustnessNanomaterials 13 00472 i001For MTS assay, decent robustness but depending on cell type used [107], and only when interferences are correctly avoided [100,108]. More elaborate SOPs and harmonization between labs enhance assay robustness [52,102].Nanomaterials 13 00472 i001Similar robustness as MTS assay [108].Nanomaterials 13 00472 i003Not assessed for NMs specifically.Nanomaterials 13 00472 i003Not assessed for NMs specifically.Nanomaterials 13 00472 i002One study showed high inter-laboratory variability [100].
CompatibilityNanomaterials 13 00472 i001Many NMs interfere with the substrate, the product, or the optical readout. Can be overcome by washing cells before incubation with reagent, and centrifugation to get rid of NMs [100,108].Nanomaterials 13 00472 i001Nanomaterials 13 00472 i002Many NMs interfere with the enzyme, the reagent, or the optical readout. Can be overcome via centrifugation. Washing not possible as LDH is measured in supernatant.Nanomaterials 13 00472 i001Nanomaterials 13 00472 i002NMs may interfere with the dye.Nanomaterials 13 00472 i001Nanomaterials 13 00472 i002NMs may interfere with the dye.Nanomaterials 13 00472 i001Nanomaterials 13 00472 i002NMs may interfere with the dye or the readout.
ReadinessNanomaterials 13 00472 i001ISO protocol for MTS assay.Nanomaterials 13 00472 i002No NM-specific standardized protocol available.Nanomaterials 13 00472 i002No NM-specific standardized protocol available.Nanomaterials 13 00472 i002No NM-specific standardized protocol available.Nanomaterials 13 00472 i002No NM-specific standardized protocol available.
Table 2. Evaluation of suitability of dissolution assays for SbD hazard testing.
Table 2. Evaluation of suitability of dissolution assays for SbD hazard testing.
Acellular AssaysCellular Assays
Performance CriteriaStatic Dissolution (e.g., OECD Series on Testing and Assessment No. 29)Flow-through/Dynamic DissolutionCellular In Vitro Dissolution
Simplicity and costNanomaterials 13 00472 i001Nanomaterials 13 00472 i001This system is the simplest and could be conducted by commercial laboratories without extensive investment in equipment.Nanomaterials 13 00472 i002Requires much greater effort with regards to setup, and also requires a large volume of fluid. Nanomaterials 13 00472 i001The basic principle of this method is simple and can be performed cheaply. Would be considered high throughput but as cellular will typically incur higher costs than acellular.
Predictivity (Sensitivity and Specificity)Nanomaterials 13 00472 i001Nanomaterials 13 00472 i002Static dissolution studies have been found to correlate with in vivo results in some instances [143,146,151], but in others poor correlation is observed [137,144,146,152,153]. Losses in sensitivity may arise due to any sample handling (e.g., acidifying the sample, filtration) or saturation of ions. For highly soluble materials, dissolution may continue during centrifugation steps, resulting in greater values of dissolution.Nanomaterials 13 00472 i001Nanomaterials 13 00472 i002Good correlation observed between flow-through system using a specific simulant fluid (modified Gamble’s) and intratracheal instillation in vivo [154], and for some particles dynamic dissolution in phagolysosomal simulate fluid (PSF) was a good predictor for short term inhalation study in rats [123] and intratracheal instillation in rats [137]. Losses in sensitivity may arise due to any sample handling (e.g., acidifying the sample). Additional concerns about losses in the system due to filtration.Nanomaterials 13 00472 i001Nanomaterials 13 00472 i002Results do appear to correlate well with in vivo in some instances (e.g., fast dissolution of Ag NMs in vivo [155] and in vitro [149]). Study by Koltermann-Jülly et al. (2018) found very low levels of dissolution in macrophages compared with the abiotic flow-through system and clearance in vivo [123]. Sensitivity relies on the capability of analysing released material. Additional concerns may arise from complexing of ions to biomolecules.
CompatibilityNanomaterials 13 00472 i001Nanomaterials 13 00472 i002The basic setup is compatible with many materials. Issues may arise with hydrophobic materials and with any material whereby sensitivity cannot be achieved for further analysis due to interference with components in the biofluid mixture (e.g., Ag NMs). Nanomaterials 13 00472 i001Nanomaterials 13 00472 i002The basic setup is compatible with many materials. Issues may arise with any material whereby sensitivity cannot be achieved for further analysis due to interference with components in the biofluid mixture or membranes used (e.g., Ag NMs).Nanomaterials 13 00472 i001Nanomaterials 13 00472 i002Most common analytical technique used is ICP-MS, therefore this methodology is the most compatible with metals. Carbon-based NMs such as CNT have used analytical techniques such as UV-Vis, Raman spectroscopy, and EM, however the sensitivity of these techniques is likely to be far less.
RobustnessNanomaterials 13 00472 i002Large variability between different biofluids.Nanomaterials 13 00472 i001Nanomaterials 13 00472 i002Can result in false positives and false negatives due to issues with the filtering system (i.e., due to NMs passing through pores or causing blockages in filters).Nanomaterials 13 00472 i003No evidence of inter-laboratory comparisons. Issues may arise due to inclusion of particles on the surface of the cell rather than internalised particles only.
ReadinessNanomaterials 13 00472 i001Nanomaterials 13 00472 i002OECD protocol but specifically for environmental studies. Various fluid compositions available.Nanomaterials 13 00472 i001Nanomaterials 13 00472 i002ISO protocol outlining basic methodology. TRL identified as high/medium for metals and medium/low for organic materials (e.g., CNT) [124].Nanomaterials 13 00472 i002Validated assays available but no standardized method.
Table 3. Evaluation of suitability of oxidative potential assays for SbD hazard testing.
Table 3. Evaluation of suitability of oxidative potential assays for SbD hazard testing.
Performance CriteriaFRASESR/EPRDCFH AcellularHaemolysis Assay
Simplicity and costNanomaterials 13 00472 i001Nanomaterials 13 00472 i001Very simple but needs large amounts of NM. Nanomaterials 13 00472 i001Nanomaterials 13 00472 i001Very simple, yet might be difficult to find lab with specialized ESR/EPR equipment. Nanomaterials 13 00472 i001Nanomaterials 13 00472 i001Very simple and only requires a fluorescence reader.Nanomaterials 13 00472 i001Nanomaterials 13 00472 i001Very simple and only requires absorbance reader and whole blood.
Predictivity (Sensitivity and Specificity)Nanomaterials 13 00472 i001The assay is able to detect NMs’ reactivity at low concentrations and in a dose-dependent manner with higher sensitivity compared to DCFH assay [163]. Could distinguish between CNT types [175]. Prediction accuracy reported: 50% [162].Nanomaterials 13 00472 i001Depending on spin trap used. Aids to identify specific ROS types, which could be useful for SbD interventions [176]. Prediction accuracies reported: 69% [164] and 50% [162]. Correlated well with in vitro cytotoxicity and protein carbonylation [183,184]. Nanomaterials 13 00472 i001Nanomaterials 13 00472 i002Lacks sensitivity as compared to FRAS and ESR/EPR [163,171,175]. However, protocol adaptations [172] show ameliorated sensitivity. Prediction accuracies reported: 77% [164].Nanomaterials 13 00472 i001Is thought to be able to detect OP of both surface reactive as well as ion-shedding NMs [112]. Showed very high prediction accuracy (92%) in one study [164].
CompatibilityNanomaterials 13 00472 i001Nanomaterials 13 00472 i001Good compatibility with a wide range of NMs. Optical interferences are largely avoided using a centrifugation step but have been reported [173]. Adapted method suggested for graphene-based materials [168]. Nanomaterials 13 00472 i001Nanomaterials 13 00472 i001Good compatibility with a wide range of NMs [162]. No interferences reported.Nanomaterials 13 00472 i002High background signals resulted from dye auto-oxidation [171]. NM interferences reported [162,166,184]. Adapted DCFH protocol reduces interferences [172]. Nanomaterials 13 00472 i001No interferences reported, yet might be expected due to absorbance readout.
RobustnessNanomaterials 13 00472 i003No interlaboratory study performed. Found to be reproducible and reliable within the same lab [163,165]Nanomaterials 13 00472 i003Not assessed for NMs specifically. Nanomaterials 13 00472 i001Nanomaterials 13 00472 i002Previously lacked robustness [175]. Interlaboratory round robin tests in GRACIOUS project showed satisfactory reproducibility for positive control NMs using optimized SOP [170]. Nanomaterials 13 00472 i003Not assessed for NMs specifically.
ReadinessNanomaterials 13 00472 i001Nanomaterials 13 00472 i002No NM-specific standardized protocol available. Gandon et al. (2017) protocol available [163].Nanomaterials 13 00472 i001ISO protocol available (ISO 18827:2017) Nanomaterials 13 00472 i002Nanomaterials 13 00472 i001No NM-specific standardized protocol available. Boyles et al. (2022) protocol available [170].Nanomaterials 13 00472 i002No NM-specific standardized protocol available.
Table 4. Evaluation of suitability of Inflammation assays for SbD hazard testing.
Table 4. Evaluation of suitability of Inflammation assays for SbD hazard testing.
Submerged Cell ModelsALI Cell Models
Performance CriteriaSubmerged Cytokine Release Mono-Culture Submerged Cytokine Release Co-Culture ALI Cytokine Release Mono-CulturesALI Cytokine Release Co-Cultures
Simplicity and costNanomaterials 13 00472 i001Simple and cost effective.Nanomaterials 13 00472 i001Simple and cost effective, however creating a co-culture requires more effort and experience than a mono-culture.Nanomaterials 13 00472 i002Requires specialized exposure equipment and a certain level of expertise.Nanomaterials 13 00472 i002Requires specialized exposure equipment and a certain level of expertise; creating a co-culture requires more effort and experience than a mono-culture.
Predictivity (Sensitivity and Specificity)Nanomaterials 13 00472 i001Good correlation with in vivo found [190]. Found to be more sensitive than ALI in several studies [214,216]. Accurate ranking found [208,209].Nanomaterials 13 00472 i001Combination of immune cell and epithelial cell more predictive than epithelial cell alone [94]. Accurate ranking found [213].Nanomaterials 13 00472 i001Generally good for primary cells. Lower predictivity of epithelial cell lines. ALI exposures found more sensitive than submerged in several studies [210,211,212]. Nanomaterials 13 00472 i001Co-cultures perform better than two cell types separately [94]. BMDL of this model comes closer to the in vivo BMDL compared to submerged [211].
RobustnessNanomaterials 13 00472 i003Large inter-laboratory variability for THP-1 cells [100,108], no inter-laboratory data on other cell types.Nanomaterials 13 00472 i003Not assessed for NMs specifically. Nanomaterials 13 00472 i002Low reproducibility but similar trends between labs [195]. Low reproducibility improved after protocol optimizations [196].Nanomaterials 13 00472 i002Low reproducibility but similar trends between labs [195].
CompatibilityNanomaterials 13 00472 i001NMs may interfere with ELISA [99,118].Nanomaterials 13 00472 i001NMs may interfere with ELISA [99,118].Nanomaterials 13 00472 i001Nanomaterials 13 00472 i001Compatible with a wide range of materials, including hydrophobic and low-density NMs. However, NMs may interfere with ELISA [99,118]. Nanomaterials 13 00472 i001Nanomaterials 13 00472 i001Compatible with a wide range of materials, as exposures do not necessarily require a dispersion. However, NMs may interfere with ELISA. Might be more suitable for NMs released form NEPs.
ReadinessNanomaterials 13 00472 i002No NM-specific standardized protocol available. Nanomaterials 13 00472 i002No NM-specific standardized protocol available.Nanomaterials 13 00472 i002No NM-specific standardized protocol available.Nanomaterials 13 00472 i002No NM-specific standardized protocol available.
Table 5. Evaluation of suitability of genotoxicity assays for SbD hazard testing.
Table 5. Evaluation of suitability of genotoxicity assays for SbD hazard testing.
Simple Cell ModelsMore Complex Cell Models
Performance CriteriaGene Mutations in Cell Lines (OECD TGs 476 and 490)Chromosome Damage in Cell Lines (OECD TGs 487 MN Assay)Gene Mutations in Advanced ModelsChromosome Damage in Advanced Models (OECD TG 487)
Simplicity and costNanomaterials 13 00472 i001Nanomaterials 13 00472 i002Time consuming, requiring long culture times (e.g., 10–14 days before counting colony formation). Relatively cheap.Nanomaterials 13 00472 i001Simple and relatively cheap. Analyses can be sped up using automatic image analysis systems and flow-cytometry.Nanomaterials 13 00472 i003Not used up to now, would necessitate 3D model dissociation before cell plating, i.e., simplicity reduced as compared to simple models.Nanomaterials 13 00472 i001Nanomaterials 13 00472 i002Relatively simple and cheap for advanced models. Would be more time consuming and expensive than 2D models [260].
Predictivity (Sensitivity and Specificity)Nanomaterials 13 00472 i003Conventional chemicals: adequate (62.9%) [224,229]. NMs: no conclusions can be reached [236,259].Nanomaterials 13 00472 i003Conventional chemicals: adequate (67.8%) [224,229] NMs: no conclusions can be reached [236,259].Nanomaterials 13 00472 i003Not used up to now, no conclusion can be reached. Nanomaterials 13 00472 i003Co-culture systems may allow the evaluation of the involved genotoxicity mechanisms of action [246,247]. They may be more predictive of an in vivo-like response [260]. 3D models do not seem to be appropriate for applying this assay due to the lack of cell proliferation [242,243]. When the 3D model involves proliferating cells, it is more sensitive than 2D models, due to higher metabolic activity [260].
RobustnessNanomaterials 13 00472 i003No inter-laboratory comparisons available for NMs. Ongoing comparisons within the EU H2020 RiskGone project.Nanomaterials 13 00472 i001Nanomaterials 13 00472 i002Relatively reproducible results in some cases, but material- and cell line-specific [244]. Future inter-laboratory comparisons under the OECD project 4.95.Nanomaterials 13 00472 i003Not used up to now, no conclusion can be reached.Nanomaterials 13 00472 i003Not enough studies available yet to allow reaching conclusions.
CompatibilityNanomaterials 13 00472 i003Too low number of studies to reach conclusions [236].Nanomaterials 13 00472 i001Suitable for different NMs (no interferences reported). No information about adequacy for complex materials.Nanomaterials 13 00472 i003No conclusion can be reached. Still, for NMs could prove unsuitable since only the cells at the periphery of the spheroid/organoid would be exposed to NMs.Nanomaterials 13 00472 i001Suitable for different NMs (no interferences reported). No information about adequacy for complex materials.
ReadinessNanomaterials 13 00472 i002No NM-specific standardized protocol available.Nanomaterials 13 00472 i002No NM-specific standardized protocol available.Nanomaterials 13 00472 i002No NM-specific standardized protocol available.Nanomaterials 13 00472 i002No NM-specific standardized protocol available.
Table 6. Overview of the most important findings in this review, including knowns and needs for SbD hazard testing.
Table 6. Overview of the most important findings in this review, including knowns and needs for SbD hazard testing.
What We Know for SbD Hazard TestingWhat We Need for SbD Hazard Testing
NM treatmentDispersion protocols-Sonication can destroy intrinsic NM properties that might be part of its safer design.
-Sonication can induce underestimation of toxicity by reducing the length of CNTs.
-Sonication can enhance dissolution and release of (toxic) ions.
-Sonication leads to a lower state of agglomeration.
-Consensus around dispersion protocols.
-Dispersion guidance which covers all relevant exposure conditions and takes into account SbD interventions.
Experimental design-Testing NMs with serum results in lower in vitro toxicity.
-Calculation of the dose delivered to the cells can have impact on toxicity ranking and is therefore also required for SbD hazard testing.
-Consensus and guidance for experimental design in the context of SbD.
Compatibility and LC-Humans are not only exposed to pristine NMs, but also to NEPs, aged NMs, and NMs released during the LC.
-Testing NMs released from NEPs may pose challenges in terms of feasibility and compatibility
-Compatibility of novel NMs with currently available in vitro assays unknown.
-Guidance on how to approach testing NMs with unknown compatibility.
-More research towards determining whether testing pristine NMs is sufficient for SbD hazard testing.
Assay protocolsCytotoxicity-More elaborate SOPs enhance robustness.
-Many NMs interfere with cytotoxicity assays, which should not be overlooked.
-In vivo effect of ion shedding NMs is sufficiently accurately predicted.
-Measuring cytotoxicity is useful for identifying hazard warnings.
-Further standardization and validation of cytotoxicity assays
-Thresholds for cytotoxicity in the context of SbD
-More focus on assays that do not pose interference issues.
-To confirm predictivity of cytotoxicity assays
Dissolution-Dissolution rate may infer bio-persistency, which is important information for SbD hazard testing.
-Predictivity largely depends on readout method as well as biological fluid choice.
-Static acellular dissolution seems to be the most appropriate method for SbD hazard testing, especially as they are rather simple, however some studies indicate otherwise.
-To confirm that measuring static acellular dissolution is indeed sufficiently predictive for SbD hazard testing.
-Meaningful thresholds for dissolution rates that allow for detection of differences that will lead to meaningful SbD decisions and interventions.
Oxidative Potential-Acellular assays might be predictive enough for SbD hazard testing.
-In vivo effects of ion-shedding NMs is sufficiently accurately predicted.
-There are indications that the haemolysis assay can accurately predict effects of surface-reactive NMs.
-FRAS and ESR assays are more sensitive than DCFH. -FRAS assay can provide accurate ranking.
-To confirm that measuring acellular OP is predictive enough for SbD testing. -Meaningful thresholds for OP in the context of SbD.
Inflammation-The use of a type of immune cell is crucial (using only epithelial cells is not sufficient).
-Primary cell models have better predictivity, but (immune) cell lines may suffice for SbD hazard testing.
-Co-cultures seem to perform better than mono-cultures.
-More elaborate SOPs enhance robustness.
-More work needed to develop in vitro models that can predict chronic inflammation.
-Thresholds for in vitro inflammation in the context of SbD.
-To confirm that submerged mono-cultures of macrophage cell lines are predictive enough.
Genotoxicity-Prediction accuracies very well established for soluble chemicals, but not for NMs.
-It is important that the cell model of choice is capable of NM uptake.
-The absence of NM positive controls makes determination of prediction accuracy challenging.
-To determine prediction accuracies of assays for NM specifically.
-Round robin initiatives to test robustness of assays.
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

Ruijter, N.; Soeteman-Hernández, L.G.; Carrière, M.; Boyles, M.; McLean, P.; Catalán, J.; Katsumiti, A.; Cabellos, J.; Delpivo, C.; Sánchez Jiménez, A.; et al. The State of the Art and Challenges of In Vitro Methods for Human Hazard Assessment of Nanomaterials in the Context of Safe-by-Design. Nanomaterials 2023, 13, 472. https://doi.org/10.3390/nano13030472

AMA Style

Ruijter N, Soeteman-Hernández LG, Carrière M, Boyles M, McLean P, Catalán J, Katsumiti A, Cabellos J, Delpivo C, Sánchez Jiménez A, et al. The State of the Art and Challenges of In Vitro Methods for Human Hazard Assessment of Nanomaterials in the Context of Safe-by-Design. Nanomaterials. 2023; 13(3):472. https://doi.org/10.3390/nano13030472

Chicago/Turabian Style

Ruijter, Nienke, Lya G. Soeteman-Hernández, Marie Carrière, Matthew Boyles, Polly McLean, Julia Catalán, Alberto Katsumiti, Joan Cabellos, Camilla Delpivo, Araceli Sánchez Jiménez, and et al. 2023. "The State of the Art and Challenges of In Vitro Methods for Human Hazard Assessment of Nanomaterials in the Context of Safe-by-Design" Nanomaterials 13, no. 3: 472. https://doi.org/10.3390/nano13030472

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