Tooling, Instrumentation, Equipment Challenges in Nanotoxicology

The nanotechnology sub-field of nanotoxicology examines the potential toxic effects of nanomaterials, especially for monitoring, addressing, mitigating safety concerns.

Introduction

The rapid expansion of nanotechnology has introduced a diverse array of engineered nanomaterials (ENMs) into various sectors, including medicine, electronics, consumer products, and environmental remediation.1 This proliferation promises significant technological and economic benefits but concurrently raises concerns regarding potential risks to human health and the environment.1 Nanotoxicology, the field dedicated to assessing these risks, faces unique challenges stemming from the distinct physicochemical properties of materials at the nanoscale (typically 1-100 nm).7 Unlike their bulk chemical counterparts, ENMs exhibit properties such as high surface area-to-volume ratios, quantum effects, and unique surface reactivities, which govern their interactions with biological systems in complex ways.7 Consequently, traditional toxicological assessment methods often prove inadequate or yield unreliable results when applied to ENMs.13

Ensuring the safe and sustainable development of nanotechnology necessitates robust and reliable methods for characterizing ENMs, quantifying exposure, and evaluating potential hazards.16 However, significant gaps exist in the available tooling, instrumentation, and methodologies required for comprehensive safety assessment.5 These limitations hinder the ability to generate consistent, comparable data, establish clear structure-activity relationships, perform accurate risk assessments, and develop harmonized regulatory guidelines.1 The dire need for improved assessment capabilities is underscored by the limited exploitation of potentially beneficial nanotechnologies due to safety uncertainties 16 and the historical precedents where initially promising materials later revealed significant hazards.2

This report aims to provide a comprehensive analysis of the most significant technical barriers currently impeding nanotoxicology safety assessment. Drawing upon recent scientific literature and expert opinions, it identifies and elaborates on up to 100 specific tooling, instrumentation, and methodological quandaries. These challenges are broadly categorized into issues related to ENM characterization, dosimetry and exposure quantification, assessment of biological effects, and methodological standardization and predictive modeling. By detailing these persistent problems and their underlying causes, this report seeks to illuminate the critical areas requiring innovation and investment to advance the field and support the responsible integration of nanotechnology into society.

Section 1: Barriers in Nanomaterial Characterization for Toxicological Assessment

Comprehensive physicochemical characterization forms the bedrock of any meaningful nanotoxicological investigation.11 Understanding properties like size, shape, surface chemistry, and stability is crucial because these parameters dictate how an ENM interacts with biological components, influencing its uptake, distribution, persistence, and ultimate toxic potential.12 However, a major hurdle lies in performing this characterization under conditions relevant to biological or environmental exposure.1 ENMs are not static entities; they undergo dynamic transformations (e.g., aggregation, dissolution, protein corona formation) upon contact with complex media, meaning their properties in situ or in vivo can differ dramatically from those measured in pristine conditions.4 Many current analytical tools struggle to cope with the complexities of biological matrices and the dynamic nature of ENMs, leading to significant uncertainties in linking specific material properties to observed biological effects.

1.1 Challenges in Size and Size Distribution Measurement

Accurately determining the size and size distribution of ENMs within relevant biological or environmental media remains a fundamental challenge. Techniques commonly employed, such as Dynamic Light Scattering (DLS) and Transmission Electron Microscopy (TEM), face significant limitations when applied outside of simple, clean systems.21 DLS, which measures hydrodynamic size based on light scattering fluctuations, struggles with polydisperse samples (common for ENMs), interference from matrix components (proteins, salts), and low ENM concentrations.16 TEM provides high-resolution images but requires extensive sample preparation (drying, staining, vacuum) that can introduce artifacts like aggregation or alter particle structure, and it offers only static snapshots of potentially dynamic systems, often requiring laborious analysis for statistical relevance.21

Furthermore, distinguishing between the primary size of individual nanoparticles, the size of aggregates (strongly bound clusters), and agglomerates (loosely bound clusters) in situ is difficult yet critical, as these different forms can exhibit distinct biological behaviors and toxicities.16 Current methodologies often lack the capability for real-time, in situ monitoring of size changes during exposure experiments. This inability to track size dynamics accurately within the exposure environment represents a major bottleneck, given that particle size significantly influences cellular uptake mechanisms, translocation across barriers, biodistribution patterns, and clearance rates.11 Without tools to measure the relevant size metric (primary vs. aggregate/agglomerate) dynamically in the relevant environment, understanding these critical size-dependent biological processes is severely hampered.

1.2 Challenges in Shape and Morphology Analysis

Nanomaterial shape is another critical determinant of biological interaction, influencing processes like cellular uptake, phagocytosis rates, and inflammatory responses.20 For instance, high-aspect-ratio nanomaterials (HARN), such as carbon nanotubes or nanofibers 2, often elicit different biological responses compared to spherical particles of similar composition and volume. However, quantitatively characterizing complex morphologies, especially within biological or environmental matrices, remains challenging.

Imaging techniques like TEM and SEM can visualize shape but face difficulties in providing statistically robust, quantitative shape descriptors (e.g., aspect ratio, sphericity, surface curvature) for large particle populations in situ without time-consuming manual analysis or sophisticated image analysis algorithms that are not yet standardized.24 Furthermore, there is a lack of consensus on which specific shape metrics are most relevant for predicting biological interactions and a corresponding lack of standardized methods to measure these parameters reliably and efficiently.20 This hinders the development of clear shape-activity relationships.

1.3 Challenges in Surface Chemistry and Charge Characterization

The surface of an ENM is its primary interface with the biological world, making surface chemistry (composition, functional groups, coatings, defects) and surface charge critical factors in determining interactions with cells and biomolecules.7 However, characterizing these surface properties in situ is exceptionally difficult because surfaces are highly susceptible to modification upon contact with biological fluids (e.g., adsorption of proteins forming a corona, binding of ions). Techniques like X-ray Photoelectron Spectroscopy (XPS) or Time-of-Flight Secondary Ion Mass Spectrometry (ToF-SIMS), which provide detailed surface chemical information, typically require high vacuum conditions and are not readily applicable to hydrated, complex biological samples.

Measuring surface charge, often represented by the zeta potential, is also problematic in physiologically relevant media.16 Biological fluids typically have high ionic strength, which screens surface charges and compresses the electrical double layer, making zeta potential measurements difficult to perform and interpret reliably. Measurements are often conducted in simplified, low-ionic-strength buffers, which may not accurately reflect the effective surface charge experienced by the ENM in vivo or in vitro in complex culture media. This lack of tools for relevant surface characterization limits the understanding of how surface properties drive biological responses.

1.4 Challenges in Assessing Dynamic Transformations (Aggregation, Dissolution, Corona Formation)

Perhaps the most pervasive challenge in ENM characterization for nanotoxicology stems from their dynamic nature in biological and environmental systems. ENMs rarely remain in their pristine, as-synthesized state upon introduction into complex media. They can aggregate or agglomerate, changing their effective size and surface area 16; they can dissolve, releasing potentially toxic ions 7; and they rapidly adsorb proteins and other biomolecules to form a “corona” that dictates their biological identity.4 Assessing these transformations dynamically and in situ is crucial but technically demanding.

Robust tools for monitoring aggregation/agglomeration kinetics in real-time under relevant exposure conditions are lacking, yet the aggregation state significantly impacts dosimetry, transport, and biological activity. Similarly, accurately measuring dissolution rates and distinguishing between toxicity caused by the particles themselves versus the released ions is difficult, especially for complex multicomponent ENMs or within complex biological matrices where ions may already be present or interact with matrix components.23 Techniques like single-particle ICP-MS (sp-ICP-MS) show promise for differentiating dissolved ions from particles but require further development, validation, and wider accessibility.26 Characterizing the dynamically evolving biomolecule corona in situ is also challenging; current methods often involve isolating the ENM-corona complex, which may perturb the corona structure and composition.21 This fundamental mismatch between the dynamic reality of nano-bio interactions and the predominantly static capabilities of many characterization tools hinders progress in understanding mechanisms and predicting effects.

1.5 Challenges in Distinguishing Engineered vs. Background Nanoparticles

For realistic exposure assessment, particularly in environmental or occupational settings, it is essential to distinguish target ENMs from naturally occurring or incidentally produced nanoparticles (e.g., from combustion or weathering) that may be present at much higher concentrations.1 Many ENMs lack unique analytical signatures, making their specific detection and quantification in complex mixtures extremely challenging. This requires the development and application of highly sensitive and specific analytical techniques, such as advanced mass spectrometry methods (e.g., ICP-MS coupled with separation techniques, isotope labeling) or specific functionalization/labeling strategies. However, such sophisticated methods are often complex, expensive, and not readily available for routine monitoring, limiting the ability to accurately assess real-world ENM exposures.17

Section 2: Barriers in Dosimetry and Exposure Quantification

Accurate dosimetry—the measurement or estimation of the dose of a substance received by a biological system—is fundamental to toxicology. It allows for the establishment of quantitative dose-response relationships, informs risk assessment, and is essential for extrapolating findings between experimental systems (e.g., in vitro to in vivo).5 For ENMs, however, dosimetry presents unique and significant challenges that go beyond those encountered with soluble chemicals.7 Issues arise from the particulate nature of the dose, complex delivery dynamics (e.g., sedimentation, diffusion, aggregation), difficulties in defining the most relevant dose metric, and challenges in measuring the dose that actually reaches the target site (the biologically effective dose).

2.1 Defining Relevant Dose Metrics

A primary challenge is the lack of consensus on the most appropriate dose metric for expressing ENM exposure and predicting biological effects.11 While mass concentration (e.g., µg/mL or µg/cm²) is the most commonly reported metric due to ease of measurement and convention, it may not be the most biologically relevant parameter for nanoparticles. Other metrics, such as particle number concentration, total surface area, surface reactivity, or even the number of specific surface sites, might be better predictors of toxicity for certain ENMs and endpoints, as toxicity is often driven by surface interactions.11

However, measuring these alternative metrics accurately and routinely, especially in situ or in vivo, poses significant technical difficulties. Furthermore, the most relevant dose metric might not be constant; it could vary depending on the ENM type, the biological system, the exposure route, the endpoint measured, and even the transformation state of the ENM within the biological compartment.12 This ambiguity complicates the comparison of results across studies and hinders the development of predictive models and regulatory guidelines. Resolving this requires not only developing tools to measure various metrics but also systematic research to determine which metric(s) best correlate with biological outcomes for different scenarios.

2.2 Quantifying Delivered Dose In Vitro

In in vitro cell-based assays, quantifying the dose of ENMs that actually reaches and interacts with the cells is notoriously difficult.21 Unlike soluble chemicals that distribute relatively uniformly in culture media, ENMs are subject to complex transport phenomena, including sedimentation (gravitational settling), diffusion, and aggregation/agglomeration, all influenced by particle properties (size, density, surface charge) and media composition (viscosity, ionic strength, protein content). Consequently, the nominal concentration added to the media often bears little resemblance to the concentration at the cell surface over time.

While computational models (e.g., based on distorted grid or particle-dynamics approaches) have been developed to estimate the delivered dose, they require accurate input parameters (e.g., effective particle density, agglomeration state in media) that are often difficult to measure or unavailable.21 There is a lack of standardized, validated, and easy-to-use tools for real-time monitoring of ENM concentration and state (e.g., size distribution, aggregation) directly at the cell-culture interface during exposure. Furthermore, quantifying the internalized dose (e.g., number or mass of particles per cell) remains challenging, especially for large cell populations needed for statistical power.24 Techniques like flow cytometry can measure cell-associated fluorescence (if particles are labeled) but require careful calibration against lower-throughput, high-resolution methods like electron microscopy to translate fluorescence intensity into particle number or mass per cell.24 This persistent uncertainty in in vitro dosimetry is a major factor contributing to the unreliability of in vitro data and the difficulty in extrapolating findings to in vivo situations.

2.3 Quantifying Dose and Biodistribution In Vivo

Determining the fate of ENMs within a whole organism (in vivo) presents another set of significant challenges. Non-invasive imaging techniques (e.g., MRI, PET, SPECT, optical imaging) can potentially track ENM distribution over time, but often lack the required sensitivity (requiring high doses) or spatial resolution, may necessitate specific labeling of the ENMs (which could alter their behavior), and are generally expensive and complex.5

Traditional methods involve sacrificing animals at different time points and measuring ENM content in various tissues and organs, typically using techniques like Inductively Coupled Plasma Mass Spectrometry (ICP-MS) or Atomic Absorption Spectroscopy (AAS) for elemental analysis. While sensitive for detecting elements, these methods usually cannot distinguish between intact nanoparticles, aggregated/transformed particles, or dissolved ions/degradation products, limiting the understanding of the form of the material present in the tissue.12 Tracking the translocation of ENMs across critical biological barriers—such as the alveolar epithelium in the lung, the intestinal wall, the blood-brain barrier, or the placenta—is particularly challenging but crucial for understanding systemic exposure and potential toxicity in sensitive organs.7

Furthermore, the development and validation of physiologically based pharmacokinetic (PBPK) models, which aim to computationally simulate ENM absorption, distribution, metabolism, and excretion (ADME), are hampered by a lack of necessary experimental data for parameterization.5 Specifically, data on ENM transport rates across membranes, dissolution kinetics in vivo, interactions with specific cell types, and clearance mechanisms are often missing for the vast array of existing ENMs. Without reliable internal dosimetry data and predictive models, relating external exposure levels to target organ doses and subsequent health risks remains highly uncertain.

2.4 Assessing Real-World Exposure Scenarios

Bridging the gap between laboratory studies and real-world human or environmental exposures requires tools and methods capable of measuring ENMs in complex, uncontrolled environments. A significant barrier is the lack of portable, affordable, real-time monitoring instruments that can specifically detect and quantify target ENMs in occupational air or environmental samples, while distinguishing them from the often much higher background of natural and incidental nanoparticles.15 Current occupational exposure assessment often relies on collecting integrated samples (e.g., on filters) for subsequent, time-consuming laboratory analysis, which provides limited information on exposure dynamics or peak concentrations.

Another major challenge lies in characterizing the release of ENMs from consumer products throughout their lifecycle (use, disposal, degradation) and understanding their subsequent fate, transport, and transformation in environmental compartments like soil, water, and air.17 Methods are needed to simulate and measure these processes under realistic conditions. Additionally, most experimental toxicology studies utilize relatively high, acute exposures, whereas real-world exposures are often chronic and at low doses. Developing and validating experimental models that can effectively mimic these realistic, long-term, low-dose scenarios to assess potential cumulative effects remains a difficult but important challenge.12

Section 3: Barriers in Assessing Biological Interactions and Toxicological Endpoints

Understanding the potential hazards of ENMs requires assessing their interactions with biological systems at multiple levels—from molecular interactions (e.g., with DNA, proteins) and cellular responses (e.g., oxidative stress, inflammation, apoptosis) to effects at the tissue, organ, and whole-organism level.2 However, applying standard toxicological assays, originally developed for soluble chemicals, to particulate ENMs is fraught with difficulties. ENMs can interfere with assay chemistries and detection systems, leading to unreliable results, and may induce toxicity through unique mechanisms not captured by conventional endpoints.13 Developing sensitive, specific, and relevant methods to probe ENM-induced biological effects remains a critical area of need.

3.1 Nanomaterial Interference with Assays

A pervasive problem in in vitro nanotoxicology is the propensity of ENMs to interfere with the assays themselves, leading to false-positive or false-negative results.13 This interference can manifest in several ways:

  • Optical Interference: Many ENMs (e.g., carbon-based materials, metallic nanoparticles) can absorb or scatter light within the wavelengths used by common detection methods like spectrophotometry (e.g., MTT, MTS, WST viability assays), fluorometry (e.g., ROS probes like DCFH-DA, resazurin assays), or luminometry (e.g., ATP-based assays). This can directly confound absorbance, fluorescence, or luminescence readings, leading to inaccurate assessment of cytotoxicity or other endpoints.13
  • Chemical Interference: ENMs can adsorb assay reagents (dyes, substrates, antibodies) onto their large surface area, reducing their availability to react or be detected. Conversely, some ENMs possess intrinsic chemical properties, such as catalytic or redox activity, that can directly react with assay components or mimic the biological endpoint being measured. For example, redox-active ENMs can directly oxidize or reduce probes used to measure cellular oxidative stress (ROS), leading to results that do not reflect the actual intracellular redox state.7 ENMs can also denature proteins, potentially interfering with enzyme-based assays (like LDH release) or ELISAs.25
  • Physical Interference: ENMs can physically interact with cellular structures or assay components in ways that cause artifacts. For instance, in the comet assay for genotoxicity, dense ENMs might impede DNA migration during electrophoresis, or ENMs might induce DNA damage during sample processing steps rather than through a biological mechanism within the cell.29 Similarly, ENMs could interfere with chromosome visualization or scoring in the micronucleus assay.

Addressing these interferences requires meticulous experimental design, including the use of appropriate particle-only controls (in acellular systems) and careful validation of each assay for the specific ENM and conditions being tested.13 This adds significant complexity, time, and cost to in vitro testing and raises fundamental questions about the validity of data generated using unadapted standard protocols. The systemic nature of these interferences challenges the reliability of many existing in vitro datasets and necessitates either laborious assay adaptation or the development of entirely new, interference-resistant measurement platforms.

3.2 Sensitivity and Specificity of Current Assays

Beyond interference, many standard toxicological assays may lack the sensitivity required to detect subtle biological perturbations caused by ENMs at low, realistic exposure concentrations.12 Cytotoxicity assays, for example, often require relatively high doses to show significant effects, potentially missing sub-lethal stress responses or adaptive changes occurring at lower concentrations. There is a need for more sensitive methods capable of detecting early molecular initiating events or key events in adverse outcome pathways (AOPs) triggered by ENM exposure.28

Furthermore, many current assays measure rather general endpoints (e.g., overall cell death, total ROS production) and may lack the specificity to elucidate the precise mechanisms of ENM toxicity. Understanding whether toxicity arises from the particle itself, released ions, physical disruption, specific receptor interactions, or other pathways requires more targeted molecular probes and assays. The development and validation of sensitive and specific biomarkers of ENM exposure or effect, applicable across different ENM types and biological systems, remain important goals that are currently unmet.

3.3 Limitations in High-Throughput Screening (HTS) / High-Content Analysis (HCA)

Given the vast number and diversity of ENMs being developed, high-throughput screening (HTS) and high-content analysis (HCA) approaches are theoretically attractive for rapidly assessing potential hazards and prioritizing materials for further testing.16 HTS/HCA platforms employ automation, robotics, and miniaturized assays (typically in microplate formats) to test many substances at multiple concentrations simultaneously.29 However, adapting these platforms for reliable ENM testing faces substantial hurdles.26

Maintaining stable, homogenous ENM dispersions during automated liquid handling is challenging, as particles can aggregate or settle in storage containers or assay plates.29 The assay interference issues discussed previously are often exacerbated in HTS formats due to miniaturization and reliance on automated optical readouts.26 Critically, integrating the necessary ENM characterization (e.g., size, aggregation state, dissolution) within the HTS workflow and under assay conditions is technically difficult but essential for interpreting results correctly.26 Validating the biological relevance and predictive capacity of HTS/HCA endpoints for in vivo toxicity also remains a major challenge, requiring comparison with lower-throughput assays and animal studies, which is often lacking.26 Finally, the initial investment and operational complexity of establishing and running robust nano-specific HTS/HCA platforms can be considerable.30 These combined difficulties currently limit the practical utility and reliability of HTS/HCA for comprehensive ENM safety assessment, despite the clear need for such approaches.

3.4 Assessing Complex Endpoints (Genotoxicity, Immunotoxicity, Chronic Effects)

Evaluating more complex toxicological endpoints poses additional challenges for ENMs. Standard genotoxicity assays (e.g., Ames bacterial mutation test, in vitro micronucleus test, comet assay) often require adaptation and careful interpretation when applied to ENMs due to potential interference, unique mechanisms of DNA damage (e.g., secondary genotoxicity via inflammation or oxidative stress), and difficulties in ensuring particle delivery to the target genetic material.4 Distinguishing direct genotoxic effects from indirect effects remains difficult.

Assessing immunotoxicity is also complex, as ENMs can interact with various components of the immune system, potentially causing immunosuppression, immunostimulation (including adjuvant effects), or hypersensitivity reactions. The formation of the protein corona plays a critical, yet poorly understood, role in mediating these interactions.4 Developing standardized in vitro and in vivo models that adequately capture the complexity of immune responses to ENMs is an ongoing challenge.

Finally, assessing the potential for long-term, chronic health effects resulting from biopersistent ENMs or repeated low-dose exposures is severely limited by the lack of validated methods and models.12 Most nanotoxicology studies focus on acute or sub-acute exposures and may miss delayed or cumulative effects. Similarly, assessing effects on potentially sensitive subpopulations (e.g., individuals with pre-existing conditions) or during critical windows of development (e.g., prenatal exposure 27) requires specialized models and approaches that are not yet widely established or validated for ENMs.

Section 4: Barriers in Methodological Standardization and Predictive Models

Progress in nanotoxicology safety assessment is significantly hampered by a lack of standardization across methods, protocols, and reporting practices. This deficiency leads to poor data comparability and reproducibility, hinders regulatory acceptance of generated data, and impedes the development of reliable predictive models needed to manage the ever-increasing number of novel ENMs.1 Concurrently, while advanced in vitro and in silico models offer promise for reducing reliance on animal testing and improving predictive capabilities, their development, validation, and application for ENMs face specific technical hurdles.

4.1 Lack of Standardized Protocols and Methods

Despite years of research, a major barrier remains the absence of widely accepted, formally validated standard operating procedures (SOPs) for many critical aspects of nanotoxicology testing.4 This includes protocols for ENM dispersion, characterization in relevant media, dosimetry determination, conducting specific in vitro and in vivo toxicity assays (accounting for interferences), and data reporting. This lack of standardization contributes significantly to the variability and frequent inconsistencies observed in the nanotoxicology literature.14

Achieving inter-laboratory reproducibility is extremely difficult when labs use different protocols, variations in critical reagents (e.g., cell culture media, dispersion agents), different cell lines or animal strains, or subtly different equipment settings.32 While international organizations like the Organisation for Economic Co-operation and Development (OECD) and the International Organization for Standardization (ISO) are working on developing standardized test guidelines and guidance documents for nanomaterials 33, progress is often slow, struggling to keep pace with the rapid innovation in nanomaterial synthesis and application. Furthermore, validating these standardized methods across the diverse range of ENMs poses a significant challenge.

4.2 Scarcity of Reference Materials

Method validation, instrument calibration, quality assurance, and inter-laboratory comparisons rely heavily on the availability of appropriate reference materials (RMs) and certified reference materials (CRMs). However, there is a significant scarcity of well-characterized, stable, and widely accessible RMs/CRMs for many common types of ENMs.17 Developing such materials is challenging due to the difficulty in producing ENMs with highly consistent and stable physicochemical properties (size, shape, surface chemistry, purity) in sufficient quantities.

Moreover, existing RMs often represent pristine, as-synthesized materials, whereas reference materials mimicking ENMs in more relevant states—such as aged ENMs, ENMs dispersed in complex matrices, or ENMs bearing a specific biomolecule corona—are largely unavailable but critically needed for validating methods under realistic conditions. Without adequate RMs/CRMs, ensuring the accuracy and comparability of measurements across different studies and laboratories remains a major obstacle.17

4.3 Challenges in Developing and Validating Advanced In Vitro Models

There is a strong push, driven by ethical considerations (3Rs principle: Replacement, Reduction, Refinement of animal testing) and efficiency needs, to develop more physiologically relevant in vitro models that can better predict in vivo human responses.4 These include 3D cell cultures, co-cultures of different cell types, organoid models, microfluidic organ-on-a-chip systems, and models incorporating biological barriers (e.g., lung, gut, blood-brain barrier).5 While promising, creating and validating these models specifically for nanotoxicology presents unique challenges.

Replicating the complex microenvironment of in vivo tissues—including multicellular architecture, extracellular matrix components, physiological flow conditions, and tissue-specific functions—is inherently difficult.16 Incorporating realistic immune system components, which play a crucial role in responses to ENMs, adds another layer of complexity. For ENM testing, specific challenges arise in achieving controlled and quantifiable delivery of particles to target cells within these complex 3D or microfluidic systems, and in performing in situ characterization of ENMs and cellular responses within these miniaturized, often optically challenging, environments.37 Furthermore, standardized methods for rigorously validating the physiological relevance and predictive capacity of these advanced models for various ENM types and toxicity endpoints are still lacking.4 Overcoming these technical hurdles is essential to realize the potential of advanced in vitro models for improving nanotoxicology assessment and reducing animal use.

4.4 Limitations in Computational Modeling (QSAR, PBPK)

Computational modeling approaches, such as quantitative structure-activity relationship (QSAR) modeling and physiologically based pharmacokinetic (PBPK) modeling, hold great potential for predicting ENM behavior and toxicity, prioritizing testing, and aiding risk assessment.5 QSAR models attempt to correlate physicochemical properties (descriptors) of ENMs with their biological activity, while PBPK models simulate the ADME processes within an organism. However, the application of these models to ENMs is currently limited by several factors.

Developing robust QSAR models requires large datasets of high-quality, standardized experimental data covering a diverse range of ENMs and endpoints, which are often unavailable.5 Identifying and quantifying the most relevant descriptors for ENMs is also more complex than for traditional chemicals, needing to encompass parameters like size distribution, shape, surface area, surface chemistry, dissolution rate, and potentially corona characteristics.20 For PBPK models, accurately parameterizing the models to capture ENM-specific processes—such as transport across biological barriers, cellular uptake rates, intracellular trafficking, dissolution and transformation kinetics in vivo, and clearance pathways—is a major bottleneck due to insufficient experimental data.5 Computationally modeling the formation of the biomolecule corona and its dynamic influence on ENM fate and interaction is another significant challenge. Effective use of computational toxicology for ENMs requires better integration between model development and targeted experimental data generation to fill critical knowledge gaps and validate model predictions.5

Section 5: Prioritized Tooling Barriers in Nanotoxicology (Detailed Explanations)

This section details specific tooling, instrumentation, and methodological barriers identified as significant impediments to nanotoxicology safety assessment, drawing from the preceding analysis and supporting literature. The barriers are roughly prioritized based on their perceived impact, frequency of mention in recent expert reviews, and fundamental nature.

Characterization Barriers (Primarily Section 1 related)

  1. Lack of Real-Time, In Situ Size/Aggregation Monitoring Tools: Measuring ENM size distribution and aggregation/agglomeration state dynamically within complex biological media (cell culture media, blood, lung fluid) during exposure is crucial but lacks robust tools. Standard methods like DLS are confounded by matrices, and EM provides only static snapshots after disruptive sample preparation.16 This prevents accurate understanding of the particle form interacting with cells/tissues over time, hindering dosimetry and mechanism studies. Persistence: Technical difficulty of non-invasive, real-time measurement in optically complex, dynamic liquid environments.
  2. Difficulty Quantifying Dissolution Kinetics In Situ: Determining the rate and extent of ENM dissolution and ion release within relevant biological fluids is critical for distinguishing particulate vs. ionic toxicity but remains challenging. Techniques like sp-ICP-MS show promise but require further validation, standardization, and accessibility, and may struggle with complex matrices or very low dissolution rates.23 Lack of reliable dissolution data hinders hazard assessment, especially for metal/metal oxide ENMs. Persistence: Analytical challenges in detecting low ion concentrations against background, separating ions from particles in situ, and matrix effects.
  3. Inadequate Tools for In Situ Surface Chemistry Analysis: Characterizing the surface functional groups, coatings, defects, and adsorbed biomolecules (corona) of ENMs directly within biological media is vital as the surface dictates interactions, yet current surface-sensitive techniques (XPS, ToF-SIMS) typically require vacuum and are unsuitable for liquids.16 This limits understanding of how surface properties change dynamically and influence biological fate. Persistence: Fundamental incompatibility of high-vacuum surface analysis techniques with hydrated biological samples.
  4. Unreliable Zeta Potential Measurement in Biological Media: Assessing surface charge via zeta potential in high ionic strength, protein-rich biological fluids is highly problematic due to charge screening and molecular adsorption, making results difficult to interpret and often irrelevant to the in vivo situation.16 Measurements are often made in unrealistic low-salt buffers. This hinders understanding the role of electrostatic interactions in ENM behavior. Persistence: Physical chemistry limitations of electrophoresis-based measurements in conductive, complex media.
  5. Challenges in Characterizing the Biomolecule Corona Dynamically: The protein/biomolecule corona formed on ENMs in biological fluids dictates their biological identity, but characterizing its composition and structure dynamically in situ without altering it during isolation/analysis is extremely difficult.4 Current methods provide snapshots and may introduce artifacts. This limits understanding of corona-mediated uptake, transport, and toxicity. Persistence: The labile nature of the corona and limitations of analytical techniques to probe it non-invasively in complex media.
  6. Difficulty Distinguishing Aggregates vs. Agglomerates In Situ: Differentiating between strongly bound aggregates and loosely bound agglomerates in suspension is important as they may have different transport properties and biological effects, but current tools often measure only an overall hydrodynamic size.16 Techniques providing structural information (e.g., advanced microscopy, scattering methods) are often not suitable for routine in situ analysis. Persistence: Lack of techniques combining size measurement with binding energy/structural information in relevant media.
  7. Lack of Standardized Methods for HARN Morphology Quantification: Quantifying the critical morphological parameters (length, diameter, aspect ratio distribution) of high-aspect-ratio nanomaterials (HARN) like nanotubes or nanofibers reliably and efficiently, especially in complex matrices, is challenging.2 Manual EM analysis is laborious; automated image analysis lacks standardization. This hinders structure-toxicity relationship studies for potentially hazardous fibrous ENMs. Persistence: Complexity of HARN structures and limitations of automated image analysis algorithms.
  8. Difficulty Detecting ENMs at Low Concentrations in Complex Matrices: Identifying and quantifying specific ENMs at environmentally or biologically relevant low concentrations against a high background of natural/incidental particles or matrix components is a major analytical challenge.1 This requires highly sensitive and specific techniques often unavailable for routine use. Persistence: Lack of unique analytical signatures for many ENMs and inherent limitations in sensitivity/specificity of analytical instruments in complex samples.
  9. Inability to Characterize ENM Transformations During Lifecycle: Assessing how ENMs change (e.g., degradation, leaching, surface modification) as they are released from products and interact with environmental compartments (aging) requires specialized methods that are largely underdeveloped.17 This limits realistic environmental risk assessment. Persistence: Complexity of simulating diverse environmental conditions and analyzing trace levels of transformed ENMs in complex environmental matrices.
  10. Lack of Tools for In Situ Surface Reactivity Measurement: Directly measuring the surface reactivity (e.g., redox potential, catalytic activity) of ENMs within biological media is difficult but important for understanding mechanisms like oxidative stress.7 Reactivity can change upon interaction with biomolecules. Persistence: Challenges in developing probes or sensors for surface reactivity compatible with complex biological environments.
  11. Limitations of TEM for Statistically Relevant Analysis: While providing high resolution, TEM analysis typically images only a very small, potentially unrepresentative sample fraction and requires extensive, often manual, analysis to obtain statistically robust data on size, shape, or aggregation state distributions.21 Persistence: Inherent trade-off between resolution and sample throughput in electron microscopy.
  12. Limitations of DLS in Polydisperse/Complex Samples: DLS struggles to accurately resolve size distributions in highly polydisperse samples (common for ENMs in biological media due to aggregation) and is very sensitive to interference from large particles or matrix components.16 Persistence: Fundamental limitations of the technique based on light scattering intensity being proportional to the sixth power of radius.
  13. Challenges in Characterizing ENM Purity/Impurities: Determining the presence and nature of impurities (e.g., residual catalysts, reagents, endotoxins) associated with ENMs is critical as impurities can confound toxicity assessments, but comprehensive impurity analysis requires multiple, often complex, analytical techniques. Persistence: Difficulty in detecting and identifying diverse potential impurities at trace levels within the nanoparticle matrix.
  14. Difficulty Assessing Nanoparticle Coating Stability/Integrity: Verifying the stability and integrity of surface coatings on ENMs within biological media over time is crucial, as coating loss can dramatically alter properties and toxicity, but methods for non-destructively monitoring coating integrity in situ are limited.7 Persistence: Challenges in probing the interface between the core particle and the coating layer in complex environments.
  15. Lack of Standardized Dispersion Protocols: Achieving consistent and relevant ENM dispersions for toxicological testing is critical but hampered by the lack of validated, standardized dispersion protocols applicable across different ENM types and test media.15 Dispersion state significantly impacts effective dose and results. Persistence: ENM properties vary widely, requiring tailored dispersion approaches; achieving stability in complex biological media is inherently difficult.

Dosimetry Barriers (Primarily Section 2 related)

  1. Lack of Consensus on Relevant Dose Metric(s): No universal agreement exists on the most biologically relevant metric (mass, number, surface area, etc.) for quantifying ENM dose, hindering comparisons and modeling.11 The best metric likely varies with ENM type and endpoint. Persistence: Complex relationship between multiple physicochemical properties and biological activity; difficulty measuring alternative metrics routinely.
  2. Inability to Reliably Quantify Delivered Dose In Vitro: Determining the actual amount of ENM reaching cells in culture over time is highly uncertain due to complex transport dynamics (sedimentation, diffusion, aggregation) not captured by nominal concentration.21 This undermines the accuracy and relevance of in vitro dose-response data. Persistence: Difficulty in measuring or accurately modeling particle transport in dynamic culture systems.
  3. Lack of Tools for Real-Time Dose Monitoring at Cell Interface: Instruments or methods to directly monitor ENM concentration and state (e.g., size) at the cell surface in vitro in real-time are lacking. This prevents accurate assessment of the dose cells actually experience during exposure. Persistence: Technical challenges of non-invasive measurement at the microscale in complex liquid environments.
  4. Difficulty Quantifying Internalized Dose per Cell: Accurately measuring the number or mass of ENMs taken up by individual cells, especially for large populations needed for HTS or statistical power, remains challenging.24 Methods like flow cytometry require complex calibration; EM is low-throughput. Persistence: Limitations of current techniques to quantify intracellular particulates rapidly and accurately across many cells.
  5. Limited Sensitivity/Resolution of In Vivo Imaging Techniques: Non-invasively tracking and quantifying ENM biodistribution in vivo is hampered by the limited sensitivity (requiring high doses) and spatial resolution of current imaging modalities (MRI, PET, SPECT, optical).5 Persistence: Fundamental physical limitations of imaging techniques in detecting small quantities deep within tissues.
  6. Inability to Differentiate Particle States In Vivo: Quantifying ENMs in tissues post-mortem (e.g., via ICP-MS) typically measures total elemental content, failing to distinguish intact particles from ions or transformed species.12 This limits understanding of the biologically active form. Persistence: Lack of techniques combining elemental quantification with structural/chemical state information in complex tissue matrices.
  7. Challenges Tracking Translocation Across Biological Barriers: Methods to accurately quantify ENM movement across critical biological barriers (lung, gut, BBB, placenta) in vivo or in advanced in vitro models are limited, hindering assessment of systemic exposure and target organ toxicity.7 Persistence: Difficulty accessing barrier interfaces and measuring low levels of translocation non-invasively or in complex models.
  8. Lack of Validated PBPK Models for ENMs: Development of reliable PBPK models to predict ENM internal dosimetry is hindered by insufficient experimental data for parameterization (e.g., transport rates, dissolution kinetics in vivo) across the diverse range of ENMs.5 Persistence: Complexity of ENM ADME processes and lack of standardized data generation for model inputs.
  9. Absence of Portable, Real-Time ENM Exposure Monitors: Tools for specific, real-time monitoring of occupational or environmental exposure to airborne ENMs, distinguishing them from background particles, are largely unavailable.15 This limits accurate exposure assessment and risk management. Persistence: Difficulty developing sensors with sufficient specificity, sensitivity, and portability for diverse ENMs in complex aerosol environments.
  10. Difficulty Mimicking Chronic Low-Dose Exposures: Designing and conducting experiments that realistically simulate long-term, low-dose human or environmental exposures is challenging logistically and technically, yet crucial for assessing chronic health risks.12 Persistence: Cost and duration of chronic studies; difficulty maintaining stable low concentrations and relevant exposure conditions over time.
  11. Poor Quantification of Dermal Exposure/Uptake: Methods for accurately quantifying ENM deposition on skin and subsequent penetration through the stratum corneum under realistic exposure conditions (e.g., from cosmetics, occupational contact) are underdeveloped. Persistence: Complexity of skin barrier properties and challenges in non-invasively measuring penetration.
  12. Challenges in Assessing Inhalation Dosimetry: Accurately modeling or measuring the deposition patterns and delivered dose of inhaled ENMs in different regions of the respiratory tract is complex, influenced by particle properties (size, shape, density) and breathing patterns.11 Persistence: Complexity of aerosol dynamics and lung geometry; limitations of current deposition models and measurement techniques.
  13. Difficulty Measuring Dose in Complex Environmental Media: Quantifying ENM concentrations and bioavailability in realistic environmental matrices like soil, sediment, or surface waters is challenging due to strong matrix interactions and background interference.1 Persistence: Heterogeneity of environmental samples and difficulty extracting/detecting ENMs without altering them.
  14. Lack of Tools to Measure Biologically Effective Dose: Determining the actual dose reaching the specific molecular or cellular target and eliciting a response (the biologically effective dose) is extremely difficult but arguably the most relevant metric. Current methods typically measure applied or tissue-level dose.7 Persistence: Requires subcellular resolution tracking and quantification, often beyond current capabilities.
  15. Uncertainty in Dose Extrapolation Across Species: Extrapolating dose-response relationships from animal models to humans is complicated for ENMs due to potential species differences in physiology, ADME processes, and sensitivity, requiring better dosimetry data and validated scaling approaches.5 Persistence: Biological differences between species and lack of comparative ENM pharmacokinetic data.

Effects Assessment Barriers (Primarily Section 3 related)

  1. Pervasive Assay Interference (Optical): ENMs frequently absorb or scatter light, interfering with standard optical detection methods (absorbance, fluorescence, luminescence) used in many cytotoxicity, proliferation, and reporter gene assays.13 This requires extensive controls or alternative detection methods. Persistence: Inherent optical properties of many ENM types (e.g., carbon materials, metals).
  2. Pervasive Assay Interference (Chemical/Adsorption): ENMs can adsorb assay reagents or possess intrinsic reactivity (e.g., redox activity) that confounds assays measuring endpoints like oxidative stress (ROS), enzyme activity (LDH), or cytokine levels.7 Persistence: High surface area and reactivity of ENMs leading to non-specific binding and chemical interactions.
  3. Pervasive Assay Interference (Physical): ENMs can physically hinder processes in certain assays, such as DNA migration in the comet assay or interaction with detection antibodies in ELISAs.29 Persistence: Particulate nature and potential for steric hindrance or non-specific binding.
  4. Lack of Validated Interference Controls: While the need for controls is recognized, standardized, universally applicable control experiments to definitively rule out ENM interference across all assay types are lacking.13 Persistence: Interference mechanisms can be complex and assay-specific.
  5. Insufficient Sensitivity for Low-Dose Effects: Many standard toxicity assays lack the sensitivity to detect subtle biological effects or adaptive responses occurring at low, environmentally or occupationally relevant ENM concentrations.12 Persistence: Assays often designed to detect overt toxicity, not subtle pathway perturbations.
  6. Lack of Specific Mechanistic Assays: Current tools often measure general toxicity endpoints, making it difficult to identify specific molecular initiating events or key events in adverse outcome pathways (AOPs) for ENMs.28 Persistence: Complexity of ENM interactions; need for development of targeted pathway-specific assays validated for ENMs.
  7. Difficulties Adapting HTS for ENM Dispersions: Automated liquid handling systems in HTS platforms struggle to maintain stable, homogenous ENM dispersions, leading to inaccurate dosing.29 Persistence: Tendency of ENMs to aggregate/settle in miniaturized formats and aqueous media over time.
  8. Integrating Characterization into HTS Workflows: Performing necessary ENM characterization (size, aggregation, etc.) within the high-speed, automated HTS workflow is technically challenging but crucial for data interpretation.26 Persistence: Mismatch between the speed of HTS and the time/complexity required for most ENM characterization techniques.
  9. Amplified Assay Interference in HTS: Interference issues (optical, chemical) are often magnified in HTS formats due to smaller volumes, reliance on automated readouts, and potential interactions with plate materials.26 Persistence: Miniaturization reduces signal-to-noise ratios, making assays more susceptible to interference.
  10. Lack of HTS/HCA Validation for Predictive Power: Establishing the correlation and predictive validity of results from high-throughput in vitro screens for in vivo ENM toxicity remains a major hurdle, limiting their regulatory acceptance.26 Persistence: Biological complexity gap between simple in vitro HTS models and whole organisms; lack of sufficient comparative data.
  11. Challenges in Adapting Standard Genotoxicity Assays: Assays like Ames, micronucleus, and comet require significant adaptation and careful interpretation for ENMs due to interference potential and unique (potentially indirect) mechanisms of genotoxicity.4 Persistence: Difficulty ensuring particle delivery to DNA and distinguishing direct vs. indirect effects.
  12. Underdeveloped Immunotoxicity Assessment Methods: Standardized, validated in vitro or in vivo models specifically suited for assessing the diverse potential immunotoxic effects of ENMs (inflammation, allergy, suppression) are lacking.4 Persistence: Complexity of the immune system and ENM interactions with its components (including corona effects).
  13. Lack of Methods for Chronic Toxicity Assessment: Validated experimental models and protocols for evaluating long-term health effects from chronic or repeated low-dose ENM exposure are scarce.12 Most studies focus on acute effects. Persistence: Cost, time, and technical challenges of conducting long-term studies, especially with potentially biopersistent materials.
  14. Limited Tools for Assessing Neurotoxicity: Evaluating potential neurotoxic effects of ENMs, particularly those crossing the blood-brain barrier, requires specialized models (e.g., advanced in vitro BBB models, relevant neuronal cell types) and sensitive endpoints that are still under development.37 Persistence: Complexity of the CNS and BBB; difficulty modeling neuronal networks in vitro.
  15. Difficulties Assessing Reproductive/Developmental Toxicity: Investigating ENM effects on reproduction and development, including placental transfer and impacts on offspring, requires complex in vivo studies or advanced in vitro models that are challenging to implement and validate.4 Persistence: Ethical constraints and complexity of developmental processes.
  16. Lack of High-Content Endpoints for Subtle Effects: While HCA offers multiparametric analysis, developing and validating high-content imaging endpoints that reliably capture subtle, sub-lethal cellular changes induced by ENMs remains challenging.26 Persistence: Requires sophisticated image analysis and understanding of subtle morphological/functional markers of toxicity.
  17. Difficulty Assessing Fiber Toxicity Mechanisms: Elucidating the specific mechanisms by which high-aspect-ratio ENMs (fibers) cause toxicity (e.g., frustrated phagocytosis, chronic inflammation) requires specialized assays and imaging techniques capable of probing particle-cell interactions at high resolution over time. Persistence: Challenges in visualizing dynamic interactions involving long fibers and specific cell types (e.g., macrophages).
  18. Inadequate Assessment of Cardiovascular Effects: Methods to assess potential cardiovascular impacts of ENMs (e.g., effects on endothelial function, thrombosis, atherosclerosis), particularly following inhalation exposure, require refinement and validation. Persistence: Complexity of cardiovascular system responses and challenges in modeling systemic effects in vitro.
  19. Challenges in Assessing Ecotoxicity in Relevant Models: Evaluating ENM toxicity in environmentally relevant species and complex ecosystems (e.g., soil mesocosms, aquatic communities) is hampered by difficulties in controlling exposure, characterizing ENMs in environmental media, and measuring relevant ecological endpoints.17 Persistence: Complexity of ecosystem interactions and matrix effects in environmental samples.
  20. Lack of Tools for Real-Time Monitoring of Cellular Responses: Methods for continuously monitoring key cellular responses (e.g., ROS production, calcium flux, membrane potential) in real-time during ENM exposure in vitro are limited but needed to understand dynamic effects.31 Persistence: Need for non-invasive, biocompatible sensors compatible with cell culture and microscopy.

Standardization and Modeling Barriers (Primarily Section 4 related)

  1. Absence of Validated SOPs for Core Assays: Lack of widely accepted, validated Standard Operating Procedures (SOPs) for fundamental nanotoxicology tasks (dispersion, characterization, key toxicity assays) hinders data reproducibility and comparability.4 Persistence: Slow pace of formal standardization processes relative to rapid ENM development; difficulty validating across diverse ENMs.
  2. Poor Inter-Laboratory Reproducibility: Studies often show significant variability in results between different laboratories testing the same ENM, largely due to lack of standardized protocols and methods.14 Persistence: Subtle differences in materials, protocols, equipment, and analysis can lead to divergent outcomes.
  3. Scarcity of Certified Reference Materials (CRMs): Lack of well-characterized, stable, and widely available ENM reference materials impedes method validation, quality control, and instrument calibration.17 Persistence: Difficulty producing ENMs with sufficient homogeneity, stability, and relevance to industrial materials.
  4. Lack of RMs for Transformed/Matrix-Bound ENMs: Reference materials representing ENMs in realistic states (e.g., aged, with corona, in environmental matrices) are virtually non-existent but needed for validating methods under relevant conditions.17 Persistence: Extreme difficulty in producing stable, well-characterized materials mimicking complex, dynamic states.
  5. Difficulty Validating Advanced In Vitro Models: Establishing the physiological relevance and predictive capacity of complex in vitro models (3D, OoC) for ENM toxicity compared to in vivo outcomes lacks standardized validation strategies.4 Persistence: Complexity of models and in vivo systems; lack of benchmark data and validation frameworks.
  6. Challenges Integrating Monitoring in Advanced Models: Incorporating real-time ENM characterization and biological response monitoring tools within complex microfluidic or 3D culture systems is technically demanding.37 Persistence: Miniaturization and complexity of models limit access for conventional analytical tools.
  7. Lack of Data for ENM QSAR Model Training: Insufficient high-quality, standardized data linking comprehensive ENM physicochemical properties to toxicological endpoints hinders the development and validation of predictive QSAR models.5 Persistence: Cost and effort required to generate large, reliable datasets covering diverse ENMs and endpoints with full characterization.
  8. Complexity of Descriptors for ENM QSAR: Identifying and quantifying the complex set of descriptors (size, shape, surface, corona, etc.) needed to capture ENM properties relevant to toxicity is much harder than for simple molecules.20 Persistence: Multifaceted nature of ENM properties influencing biological interactions.
  9. Parameterization Bottlenecks for ENM PBPK Models: Lack of essential experimental data (e.g., transport rates, dissolution kinetics, clearance pathways in vivo) prevents accurate parameterization of PBPK models for most ENMs.5 Persistence: Difficulty obtaining necessary in vivo kinetic data for diverse ENMs.
  10. Difficulty Modeling Corona Effects: Computationally simulating the formation of the biomolecule corona and predicting its impact on ENM fate and toxicity is highly complex and lacks validated modeling approaches. Persistence: Dynamic nature and complex composition of the corona; limited understanding of corona-cell interactions.
  11. Lack of Standardized Reporting Guidelines: Inconsistent and incomplete reporting of experimental details (especially ENM characterization and dosimetry) in publications makes it difficult to interpret, compare, and reproduce findings.16 Persistence: Lack of enforcement of comprehensive reporting standards in journals and research communities.
  12. Slow Pace of Formal Standardization (OECD, ISO): Development and adoption of internationally recognized standard test guidelines for ENMs through bodies like OECD and ISO lags behind the rapid pace of nanotechnology innovation.33 Persistence: Consensus-based standardization processes are inherently slow and complex, especially for novel materials.
  13. Difficulty Harmonizing Definitions: Achieving global consensus on the definition of a “nanomaterial” for regulatory purposes remains challenging, impacting consistent classification and assessment.1 Persistence: Different stakeholder needs and scientific complexities in setting size or property boundaries.
  14. Lack of Tools for Data Integration and Management: Efficient tools and platforms for integrating, managing, and analyzing the large, complex datasets generated in nanotoxicology (characterization, omics, imaging, toxicology) are underdeveloped. Persistence: Heterogeneity of data types and formats; need for specialized nanoinformatics tools.32
  15. Challenges in Validating In Silico Models: Rigorous validation of computational models (QSAR, PBPK, docking) against independent experimental data is often lacking, limiting confidence in their predictions.5 Persistence: Scarcity of suitable validation datasets; complexity of model validation procedures.
  16. Need for Standardized Negative/Benchmark Controls: Lack of well-defined, widely accepted negative control nanoparticles (demonstrably non-toxic under specific conditions) and benchmark materials with known toxicity profiles hinders assay validation and relative hazard ranking. Persistence: Difficulty ensuring complete lack of bioactivity or producing materials with highly consistent, known toxicity.
  17. Difficulty Incorporating Realistic Exposure Routes in In Vitro Models: Many advanced in vitro models struggle to accurately mimic realistic exposure routes like inhalation (requiring air-liquid interface culture) or ingestion (requiring complex gut models). Persistence: Technical challenges in recreating complex physiological interfaces and exposure dynamics in vitro.
  18. Lack of Standardized Methods for Assessing Nanoparticle Stability: Validated protocols for assessing the colloidal stability and physicochemical integrity of ENMs over time in relevant storage conditions and test media are needed but often lacking. Persistence: Stability depends heavily on specific ENM properties and media composition.
  19. Difficulty Linking In Vitro Assays to AOPs: Systematically linking results from specific in vitro assays to key events within established or putative Adverse Outcome Pathways (AOPs) for ENMs requires better mechanistic understanding and validated assays for key events.28 Persistence: AOP development for ENMs is still in early stages; linking assays requires mechanistic validation.
  20. Lack of Read-Across Frameworks for ENMs: Developing reliable read-across approaches (predicting toxicity of one ENM based on data from similar ones) is hampered by the complexity of ENM properties and lack of standardized data/descriptors.17 Persistence: Difficulty defining similarity criteria for complex ENMs; lack of sufficient data for robust analogue identification.

Cross-Cutting and Emerging Barriers

  1. Assessing Combined Exposures/Mixture Toxicity: Evaluating the effects of ENMs in combination with other chemicals or nanoparticles (mixture toxicity) is crucial for realistic scenarios but lacks established methodologies.32 Persistence: Exponential increase in complexity when considering mixtures; potential for synergistic/antagonistic interactions.
  2. Evaluating Effects of Weathered/Aged ENMs: Understanding how environmental weathering or aging processes alter ENM properties and toxicity requires methods to simulate aging and test transformed materials, which are underdeveloped.17 Persistence: Complexity of environmental transformation processes; difficulty creating relevant aged materials in the lab.
  3. Characterizing and Testing Nanoplastics: Assessing the risks of nanoplastics requires specific analytical tools for detection and characterization in environmental/biological samples and relevant toxicity testing models, which are emerging but not yet mature.4 Persistence: Extreme diversity of plastic types, shapes, sizes, and associated chemicals; low concentrations in environmental samples.
  4. Assessing Advanced/Complex Nanomaterials: Evaluating the safety of next-generation ENMs (e.g., multi-component, hybrid, surface-patterned, self-assembling) poses new challenges for characterization and toxicity testing beyond simpler materials.17 Persistence: Increasing complexity of material design outpaces development of assessment methods.
  5. Lack of Tools for Single-Cell Analysis: Methods to analyze ENM uptake, localization, and cellular responses at the single-cell level are needed to understand population heterogeneity but are often complex and low-throughput.24 Persistence: Requires high-resolution imaging or specialized flow cytometry/spectroscopy techniques.
  6. Difficulty Assessing Endotoxin Contamination: Reliably detecting and quantifying endotoxin contamination on ENMs is critical as it can confound inflammatory responses, but standard LAL assays can suffer from ENM interference. Persistence: Interference of ENMs with the Limulus Amebocyte Lysate (LAL) assay components or detection.
  7. Lack of Rapid Screening Tools for Manufacturing: Need for fast, reliable, potentially online tools to monitor critical ENM properties (e.g., size, purity) during manufacturing for quality control and ensuring consistency relevant to safety assessment. Persistence: Integrating complex characterization tools into rapid manufacturing processes is challenging.
  8. Challenges in Assessing Biodegradability/Persistence: Determining the rate and extent of ENM degradation in biological or environmental systems is crucial for assessing long-term risks but lacks standardized methods applicable across diverse ENM types.16 Persistence: Slow degradation rates; complexity of degradation pathways; difficulty tracking degradation products.
  9. Linking Physicochemical Properties to Corona Composition: Understanding precisely how ENM physicochemical properties influence the composition of the adsorbed biomolecule corona requires systematic studies and advanced analytical tools, but predictive capability is limited.4 Persistence: Complexity of protein-surface interactions and combinatorial possibilities.
  10. Inadequate Tools for Studying Subcellular Localization: Determining the precise location of ENMs within cells (e.g., specific organelles, nucleus) and correlating localization with effects requires high-resolution imaging (e.g., EM, super-resolution microscopy) combined with specific labeling, which is technically demanding.24 Persistence: Resolution limits of light microscopy; artifacts in EM preparation.
  11. Difficulty Assessing Effects on Microbiome: Evaluating the impact of ingested or environmentally released ENMs on microbial communities (e.g., gut microbiome, soil microbes) requires integrating ENM characterization with metagenomic/metabolomic analyses, presenting methodological challenges. Persistence: Complexity of microbial ecosystems and their interactions with ENMs.
  12. Lack of Validated In Silico Tools for Corona Prediction: Computational tools to accurately predict the composition and structure of the biomolecule corona based on ENM properties and biological fluid composition are still in early development and lack validation. Persistence: Complexity of protein folding, adsorption dynamics, and competitive binding at surfaces.
  13. Challenges in High-Throughput Genotoxicity Screening: Adapting genotoxicity assays for reliable high-throughput screening of ENMs faces significant hurdles related to interference, particle delivery, and assay sensitivity/specificity in miniaturized formats.29 Persistence: Combines HTS challenges with specific difficulties of genotoxicity assays for ENMs.
  14. Need for Better In Vitro Blood-Brain Barrier Models: Current in vitro BBB models often lack the full complexity (multiple cell types, flow, matrix) and validated permeability characteristics needed for reliable assessment of ENM translocation.37 Persistence: Difficulty recreating the intricate structure and function of the neurovascular unit in vitro.
  15. Need for Better In Vitro Lung Models (Air-Liquid Interface): While ALI models exist, standardizing them for ENM aerosol exposure, ensuring relevant deposition dosimetry, and incorporating immune components remains challenging for routine inhalation toxicity assessment. Persistence: Technical complexity of maintaining ALI cultures and delivering aerosols reproducibly.
  16. Lack of Tools to Assess Nanoparticle-Drug Interactions: For nanomedicines, understanding potential interactions between the nanoparticle carrier and co-administered drugs, or effects on drug metabolism enzymes, requires specific assays that are not well-established. Persistence: Requires evaluating complex interactions involving particles, drugs, and metabolic systems.
  17. Difficulty Assessing Long-Term Fate of Non-Degradable ENMs: Predicting the long-term accumulation, potential sequestration sites, and chronic effects of biopersistent, non-degradable ENMs within the body is extremely challenging due to lack of long-term data and tracking methods.12 Persistence: Requires very long studies or highly predictive models currently unavailable.
  18. Lack of Standardized Bioavailability Assays: Methods to assess the fraction of ENMs that becomes available for uptake and interaction after administration (bioavailability), particularly via ingestion or environmental exposure, are not standardized. Persistence: Depends heavily on ENM transformation and interactions within complex exposure media (e.g., gut fluid, soil pore water).
  19. Challenges in Assessing Nanomaterial Effects on Protein Conformation: Tools to directly assess if ENMs induce functionally relevant changes in protein structure (e.g., denaturation, fibrillation, cryptic epitope exposure) upon adsorption are needed but limited.25 Persistence: Requires sensitive biophysical techniques applicable to surface-adsorbed proteins in complex media.
  20. Need for Multiplexed Toxicity Assays: Developing assays capable of simultaneously measuring multiple toxicity endpoints or pathway activations in response to ENM exposure would increase efficiency and mechanistic insight, but requires careful validation to avoid compounded interference issues.26 Persistence: Technical complexity of developing robust multiplexed assays, especially for HTS.
  21. Difficulty Validating Alternative Species Models (Zebrafish, C. elegans): While models like zebrafish embryos or C. elegans offer potential for higher throughput in vivo screening, validating their relevance and predictive power for mammalian/human toxicity across diverse ENMs requires more systematic comparative studies.4 Persistence: Biological differences between invertebrates/fish larvae and mammals; need for cross-species validation data.
  22. Lack of Tools for Assessing ENM Effects on Epigenetics: Investigating potential epigenetic modifications induced by ENMs (e.g., changes in DNA methylation, histone modification) requires specialized molecular assays adapted and validated for use with ENM exposures. Persistence: Field of nano-epigenetics is nascent; requires sensitive molecular biology techniques applied to ENM contexts.
  23. Difficulty Assessing Effects on Cell-Cell Communication: Evaluating how ENMs might disrupt signaling pathways or communication between different cell types (e.g., in co-cultures or tissue models) requires sophisticated models and analytical tools. Persistence: Requires monitoring complex intercellular interactions in the presence of potentially interfering ENMs.
  24. Lack of Standardized Protocols for Nanomedicine Quality Control: Ensuring consistent quality and safety-relevant physicochemical properties (size, drug load, surface properties) batch-to-batch for clinical-grade nanomedicines requires robust, validated QC methods often lacking standardization.8 Persistence: Complexity of nanomedicine formulations; need for stringent regulatory standards.
  25. Challenges in Life Cycle Assessment (LCA) Data: Performing comprehensive LCAs for ENMs to assess overall environmental impact is hindered by lack of data on manufacturing inputs/outputs, release rates during use/disposal, and environmental fate/toxicity. Persistence: Requires data collection across entire product lifecycle, often involving proprietary information.
  26. Inadequate Consideration of Biological Variability: Many studies use single cell lines or animal strains, failing to account for inter-individual variability in response to ENMs due to genetic background, age, or health status.7 Persistence: Requires testing across diverse populations/strains, increasing experimental complexity and cost.
  27. Lack of Tools for Assessing Mitochondrial Toxicity: Specific methods to evaluate ENM impacts on mitochondrial function (respiration, membrane potential, dynamics) beyond general ROS or ATP assays are needed, as mitochondria are frequent targets.31 Persistence: Requires specialized assays often involving live-cell imaging or isolated mitochondria, potentially subject to interference.
  28. Difficulty Assessing Lysosomal Interactions and Fate: Understanding ENM trafficking to lysosomes, lysosomal stability/damage, and subsequent cellular consequences requires specific probes and imaging techniques adapted for nanoparticle tracking. Persistence: Requires tracking particles within dynamic organelles at high resolution.
  29. Lack of Frameworks for Integrating Multi-Omics Data: Effectively integrating large datasets from different omics platforms (genomics, transcriptomics, proteomics, metabolomics) to understand ENM modes of action requires advanced bioinformatics tools and analytical frameworks specifically adapted for nanotoxicology data.8 Persistence: Complexity of multi-omics data integration and interpretation in the context of ENM exposures.
  30. Need for Improved Data Sharing and Accessibility: Lack of open, accessible databases and standardized formats for sharing raw and processed nanotoxicology data hinders meta-analyses, model development, and overall progress in the field.17 Persistence: Requires community agreement on data standards, infrastructure investment, and incentives for data sharing.

Conclusion

The comprehensive assessment of engineered nanomaterial safety remains a formidable challenge, significantly impeded by a wide array of tooling, instrumentation, and methodological barriers. This report has detailed approximately 100 such obstacles spanning the critical areas of ENM characterization, dosimetry and exposure quantification, biological effects assessment, and methodological standardization and predictive modeling. The analysis highlights several deeply ingrained, cross-cutting issues: the dynamic and transformative nature of ENMs in biological and environmental systems, which defies static measurement approaches 12; the pervasive interference of ENMs with conventional analytical techniques and toxicological assays 13; the persistent difficulties in establishing relevant dose metrics and measuring the biologically effective dose 11; the significant gap between simplified in vitro models and complex in vivo realities 5; and a critical lack of standardization in protocols, reference materials, and data reporting practices that undermines data quality and comparability.15

Overcoming these multifaceted barriers is paramount for advancing nanotoxicology from a largely descriptive field towards a more predictive and quantitative science. Progress necessitates a concerted, interdisciplinary effort involving materials scientists, chemists, biologists, toxicologists, physicists, engineers, and computational modelers. Targeted investment is required to develop novel analytical technologies capable of in situ, real-time characterization and monitoring in complex matrices. Robust validation of existing and emerging assays, including advanced in vitro models and HTS platforms, is crucial to ensure data reliability and relevance. Furthermore, significant international cooperation and commitment are needed to accelerate the development and adoption of standardized methodologies, reference materials, and harmonized reporting guidelines, thereby building a foundation for robust data integration, predictive modeling, and globally accepted risk assessment frameworks.6

Addressing the technical quandaries outlined in this report is not merely an academic exercise; it is essential for fostering public confidence, enabling responsible innovation in nanotechnology, ensuring regulatory preparedness, and ultimately protecting human health and environmental integrity in the face of rapidly evolving nanoscale materials and applications.1 While challenges remain significant, continued focus on overcoming these instrumentation and methodological limitations will pave the way for a safer and more sustainable nanotechnology future.

Table 1: Summary of Prioritized Nanotoxicology Tooling Barriers

Barrier ID Concise Barrier Title Primary Challenge Area Key Affected Aspect Approx. Significance
1 Lack of Real-Time, In Situ Size/Aggregation Monitoring Characterization In Situ Analysis, Dosimetry High
2 Difficulty Quantifying Dissolution Kinetics In Situ Characterization In Situ Analysis, Mechanism High
3 Inadequate Tools for In Situ Surface Chemistry Analysis Characterization In Situ Analysis, Mechanism High
4 Unreliable Zeta Potential Measurement in Biological Media Characterization In Situ Analysis, Bio-interactions High
5 Challenges Characterizing Biomolecule Corona Dynamically Characterization In Situ Analysis, Biological Identity High
6 Difficulty Distinguishing Aggregates vs. Agglomerates In Situ Characterization In Situ Analysis, Dosimetry Medium
7 Lack of Standardized HARN Morphology Quantification Characterization Structure-Activity, Fiber Safety Medium
8 Difficulty Detecting Low ENM Conc. in Complex Matrices Characterization Exposure Assessment, In Situ Analysis High
9 Inability to Characterize ENM Transformations During Lifecycle Characterization Environmental Risk Assessment Medium
10 Lack of Tools for In Situ Surface Reactivity Measurement Characterization Mechanism (Ox. Stress), In Situ Medium
11 Limitations of TEM for Statistically Relevant Analysis Characterization Quantitative Analysis, Throughput Medium
12 Limitations of DLS in Polydisperse/Complex Samples Characterization Size Analysis Accuracy Medium
13 Challenges in Characterizing ENM Purity/Impurities Characterization Hazard Identification, Confounding Medium
14 Difficulty Assessing Coating Stability/Integrity Characterization Material Stability, Bio-interactions Medium
15 Lack of Standardized Dispersion Protocols Characterization Reproducibility, Effective Dose High
16 Lack of Consensus on Relevant Dose Metric(s) Dosimetry Dose-Response, Risk Assessment High
17 Inability to Reliably Quantify Delivered Dose In Vitro Dosimetry In Vitro Relevance, IVIVE High
18 Lack of Tools for Real-Time Dose Monitoring at Cell Interface Dosimetry In Vitro Accuracy, Dynamic Effects High
19 Difficulty Quantifying Internalized Dose per Cell Dosimetry Cellular Dose, HTS/HCA High
20 Limited Sensitivity/Resolution of In Vivo Imaging Dosimetry Biodistribution, Non-invasive Medium
21 Inability to Differentiate Particle States In Vivo Dosimetry Biodistribution, Mechanism High
22 Challenges Tracking Translocation Across Barriers Dosimetry Systemic Exposure, Target Organs High
23 Lack of Validated PBPK Models for ENMs Dosimetry Predictive Modeling, IVIVE High
24 Absence of Portable, Real-Time ENM Exposure Monitors Dosimetry Occupational/Environmental Exposure High
25 Difficulty Mimicking Chronic Low-Dose Exposures Dosimetry Realistic Risk Assessment High
26 Poor Quantification of Dermal Exposure/Uptake Dosimetry Dermal Risk Assessment Medium
27 Challenges in Assessing Inhalation Dosimetry Dosimetry Inhalation Risk Assessment Medium
28 Difficulty Measuring Dose in Complex Environmental Media Dosimetry Ecotoxicology, Environmental Risk Medium
29 Lack of Tools to Measure Biologically Effective Dose Dosimetry Mechanism, Target Site Dose High
30 Uncertainty in Dose Extrapolation Across Species Dosimetry Human Risk Assessment, IVIVE Medium
31 Pervasive Assay Interference (Optical) Effects Assessment In Vitro Data Reliability High
32 Pervasive Assay Interference (Chemical/Adsorption) Effects Assessment In Vitro Data Reliability High
33 Pervasive Assay Interference (Physical) Effects Assessment In Vitro Data Reliability High
34 Lack of Validated Interference Controls Effects Assessment Assay Validation, Reproducibility High
35 Insufficient Sensitivity for Low-Dose Effects Effects Assessment Realistic Risk Assessment High
36 Lack of Specific Mechanistic Assays Effects Assessment Mechanism Elucidation, AOPs High
37 Difficulties Adapting HTS for ENM Dispersions Effects Assessment HTS Feasibility, Dosing Accuracy High
38 Integrating Characterization into HTS Workflows Effects Assessment HTS Interpretation, Reliability High
39 Amplified Assay Interference in HTS Effects Assessment HTS Reliability High
40 Lack of HTS/HCA Validation for Predictive Power Effects Assessment HTS Utility, Regulatory Acceptance High
41 Challenges Adapting Standard Genotoxicity Assays Effects Assessment Genotoxicity Assessment Reliability High
42 Underdeveloped Immunotoxicity Assessment Methods Effects Assessment Immunotoxicity Assessment High
43 Lack of Methods for Chronic Toxicity Assessment Effects Assessment Long-term Risk Assessment High
44 Limited Tools for Assessing Neurotoxicity Effects Assessment Neurotoxicity Assessment Medium
45 Difficulties Assessing Repro/Developmental Toxicity Effects Assessment Repro/Devo Risk Assessment Medium
46 Lack of High-Content Endpoints for Subtle Effects Effects Assessment HCA Utility, Early Effects Detection Medium
47 Difficulty Assessing Fiber Toxicity Mechanisms Effects Assessment Fiber Safety Assessment Medium
48 Inadequate Assessment of Cardiovascular Effects Effects Assessment Cardiovascular Risk Assessment Medium
49 Challenges Assessing Ecotoxicity in Relevant Models Effects Assessment Ecotoxicology, Environmental Risk Medium
50 Lack of Tools for Real-Time Monitoring of Cellular Responses Effects Assessment Dynamic Effects, Mechanism Medium
51 Absence of Validated SOPs for Core Assays Standardization/Models Reproducibility, Comparability High
52 Poor Inter-Laboratory Reproducibility Standardization/Models Data Reliability, Consensus Building High
53 Scarcity of Certified Reference Materials (CRMs) Standardization/Models Method Validation, QC High
54 Lack of RMs for Transformed/Matrix-Bound ENMs Standardization/Models Relevant Method Validation High
55 Difficulty Validating Advanced In Vitro Models Standardization/Models In Vitro Relevance, Predictive Power High
56 Challenges Integrating Monitoring in Advanced Models Standardization/Models Model Utility, Mechanistic Insight Medium
57 Lack of Data for ENM QSAR Model Training Standardization/Models Predictive Modeling (QSAR) High
58 Complexity of Descriptors for ENM QSAR Standardization/Models Predictive Modeling (QSAR) High
59 Parameterization Bottlenecks for ENM PBPK Models Standardization/Models Predictive Modeling (PBPK), IVIVE High
60 Difficulty Modeling Corona Effects Standardization/Models Predictive Modeling, Bio-interactions High
61 Lack of Standardized Reporting Guidelines Standardization/Models Data Interpretation, Reproducibility High
62 Slow Pace of Formal Standardization (OECD, ISO) Standardization/Models Regulatory Harmonization High
63 Difficulty Harmonizing Definitions Standardization/Models Regulatory Consistency Medium
64 Lack of Tools for Data Integration and Management Standardization/Models Nanoinformatics, Data Analysis Medium
65 Challenges in Validating In Silico Models Standardization/Models Model Reliability, Acceptance High
66 Need for Standardized Negative/Benchmark Controls Standardization/Models Assay Validation, Relative Ranking Medium
67 Difficulty Incorporating Realistic Exposure Routes In Vitro Standardization/Models In Vitro Model Relevance Medium
68 Lack of Standardized Methods for Assessing Stability Standardization/Models Material Consistency, Data Quality Medium
69 Difficulty Linking In Vitro Assays to AOPs Standardization/Models Mechanistic Understanding, Prediction High
70 Lack of Read-Across Frameworks for ENMs Standardization/Models Efficient Assessment, Grouping High
71 Assessing Combined Exposures/Mixture Toxicity Cross-Cutting Realistic Risk Assessment High
72 Evaluating Effects of Weathered/Aged ENMs Cross-Cutting Environmental Risk Assessment Medium
73 Characterizing and Testing Nanoplastics Cross-Cutting Emerging Contaminant Risk High
74 Assessing Advanced/Complex Nanomaterials Cross-Cutting Future Preparedness, Innovation Safety High
75 Lack of Tools for Single-Cell Analysis Cross-Cutting Heterogeneity, Mechanistic Insight Medium
76 Difficulty Assessing Endotoxin Contamination Cross-Cutting Confounding Factors, Assay Validity Medium
77 Lack of Rapid Screening Tools for Manufacturing Cross-Cutting Quality Control, Consistency Medium
78 Challenges Assessing Biodegradability/Persistence Cross-Cutting Long-term Risk, Environmental Fate High
79 Linking Physicochemical Properties to Corona Composition Cross-Cutting Predictive Understanding, Bio-ID High
80 Inadequate Tools for Studying Subcellular Localization Cross-Cutting Mechanism, Target Sites Medium
81 Difficulty Assessing Effects on Microbiome Cross-Cutting Gut/Environmental Health Medium
82 Lack of Validated In Silico Tools for Corona Prediction Cross-Cutting Predictive Modeling, Bio-ID High
83 Challenges in High-Throughput Genotoxicity Screening Cross-Cutting HTS Utility, Genotoxicity Assessment High
84 Need for Better In Vitro Blood-Brain Barrier Models Cross-Cutting Neurotoxicity Assessment, IVIVE High
85 Need for Better In Vitro Lung Models (ALI) Cross-Cutting Inhalation Toxicity, IVIVE High
86 Lack of Tools for Nanoparticle-Drug Interactions Cross-Cutting Nanomedicine Safety/Efficacy Medium
87 Difficulty Assessing Long-Term Fate of Non-Degradable ENMs Cross-Cutting Chronic Risk, Biopersistence High
88 Lack of Standardized Bioavailability Assays Cross-Cutting Exposure Assessment, Dose Medium
89 Challenges Assessing ENM Effects on Protein Conformation Cross-Cutting Mechanism, Autoimmunity Potential Medium
90 Need for Multiplexed Toxicity Assays Cross-Cutting Efficiency, Mechanistic Insight Medium
91 Difficulty Validating Alternative Species Models Cross-Cutting 3Rs, Predictive Power Medium
92 Lack of Tools for Assessing ENM Effects on Epigenetics Cross-Cutting Long-term Effects, Mechanism Low
93 Difficulty Assessing Effects on Cell-Cell Communication Cross-Cutting Tissue Function, Complex Responses Low
94 Lack of Standardized Protocols for Nanomedicine QC Cross-Cutting Nanomedicine Safety, Consistency High
95 Challenges in Life Cycle Assessment (LCA) Data Cross-Cutting Sustainability, Environmental Impact Medium
96 Inadequate Consideration of Biological Variability Cross-Cutting Population Risk, Susceptibility Medium
97 Lack of Tools for Assessing Mitochondrial Toxicity Cross-Cutting Mechanism, Cellular Energetics Medium
98 Difficulty Assessing Lysosomal Interactions and Fate Cross-Cutting Intracellular Trafficking, Mechanism Medium
99 Lack of Frameworks for Integrating Multi-Omics Data Cross-Cutting Systems Toxicology, Mechanistic Insight High
100 Need for Improved Data Sharing and Accessibility Cross-Cutting Collaboration, Modeling, Transparency High

Works cited

  1. Challenges in Nanomaterials Characterization Frontiers Research Topic, accessed April 17, 2025, https://www.frontiersin.org/research-topics/6198/challenges-in-nanomaterials-characterization/magazine
  2. Nanotoxicity: a challenge for future medicine - PMC, accessed April 17, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7379444/
  3. Challenges in Nanomaterials Characterization - Frontiers, accessed April 17, 2025, https://www.frontiersin.org/research-topics/6198/challenges-in-nanomaterials-characterization
  4. Editorial: Methods and protocols in nanotoxicology: volume II - Frontiers, accessed April 17, 2025, https://www.frontiersin.org/journals/toxicology/articles/10.3389/ftox.2024.1532023/pdf
  5. Toxicokinetics, dose-response and risk assessment of nanomaterials: methodology, challenges, and future perspectives - PMC - PubMed Central, accessed April 17, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9699155/
  6. Ethical and legal challenges in nanomedical innovations: a scoping review - PMC, accessed April 17, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10213273/
  7. Review of emerging concepts in nanotoxicology: opportunities and challenges for safer nanomaterial design, accessed April 17, 2025, https://www.tandfonline.com/doi/full/10.1080/15376516.2019.1566425
  8. Comprehensive insights into mechanism of nanotoxicity, assessment methods and regulatory challenges of nanomedicines, accessed April 17, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11452581
  9. Nanotoxicology and Exposure in the Occupational Setting - Scientific Research Publishing, accessed April 17, 2025, https://www.scirp.org/journal/paperinformation?paperid=58445
  10. Challenges in assessing nanomaterial toxicology: a personal perspective - PubMed, accessed April 17, 2025, https://pubmed.ncbi.nlm.nih.gov/20799267/
  11. The New Toxicology of Sophisticated Materials: Nanotoxicology and Beyond - PMC, accessed April 17, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3145386/
  12. From Dose to Response: In Vivo Nanoparticle Processing and Potential Toxicity - PMC, accessed April 17, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6376403/
  13. The State of the Art and Challenges of In Vitro Methods for Human Hazard Assessment of Nanomaterials in the Context of Safe-by-Design - PubMed Central, accessed April 17, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9920318/
  14. Toxicity of Nanoparticles and an Overview of Current Experimental Models - PMC, accessed April 17, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4689276/
  15. Chapter 1: Nanotoxicology: Challenges for Biologists - RSC Books, accessed April 17, 2025, https://books.rsc.org/books/edited-volume/1834/chapter/2251956/Nanotoxicology-Challenges-for-Biologists
  16. Precision Nanotoxicology in Drug Development: Current Trends and Challenges in Safety and Toxicity Implications of Customized Multifunctional Nanocarriers for Drug-Delivery Applications, accessed April 17, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9697541/
  17. Challenges in Nanomaterial Characterization – From Definition to Analysis - ResearchGate, accessed April 17, 2025, https://www.researchgate.net/publication/360706994_Challenges_in_Nanomaterial_Characterization_-_From_Definition_to_Analysis
  18. Research Strategies for Safety Evaluation of Nanomaterials, Part VIII: International Efforts to Develop Risk-Based Safety Evaluations for Nanomaterials - Oxford Academic, accessed April 17, 2025, https://academic.oup.com/toxsci/article/92/1/23/1643002
  19. NNI Environmental, Health, & Safety-Related Documents, accessed April 17, 2025, https://www.nano.gov/ehsdocuments
  20. A Review on Conventional and Advanced Methods for Nanotoxicology Evaluation of Engineered Nanomaterials - MDPI, accessed April 17, 2025, https://www.mdpi.com/1420-3049/26/21/6536
  21. At the Crossroads of Nanotoxicology in vitro: Past Achievements and Current Challenges - Oxford Academic, accessed April 17, 2025, https://academic.oup.com/toxsci/article-pdf/147/1/5/10916079/kfv106.pdf
  22. In Vivo Toxicological Analysis of MnFe2O4@poly(tBGE-alt-PA) Composite as a Hybrid Nanomaterial for Possible Biomedical Use - ACS Publications, accessed April 17, 2025, https://pubs.acs.org/doi/10.1021/acsabm.2c00983
  23. Nanomaterial Characterization in Complex Media—Guidance and Application - MDPI, accessed April 17, 2025, https://www.mdpi.com/2079-4991/13/5/922
  24. Quantification of Nanoparticle Dose and Vesicular Inheritance in Proliferating Cells ACS Nano - ACS Publications, accessed April 17, 2025, https://pubs.acs.org/doi/10.1021/nn4019619
  25. In Vitro Methods for Assessing Nanoparticle Toxicity - PMC, accessed April 17, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6984383/
  26. High throughput toxicity screening and intracellular detection of nanomaterials - PMC, accessed April 17, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5215403/
  27. Safety and Toxicity Implications of Multifunctional Drug Delivery Nanocarriers on Reproductive Systems In Vitro and In Vivo - Frontiers, accessed April 17, 2025, https://www.frontiersin.org/journals/toxicology/articles/10.3389/ftox.2022.895667/full
  28. Novel Methods and Approaches for Safety Evaluation of Nanoparticle Formulations: A Focus Towards In Vitro Models and Adverse Outcome Pathways - Frontiers, accessed April 17, 2025, https://www.frontiersin.org/journals/pharmacology/articles/10.3389/fphar.2021.612659/full
  29. High throughput toxicity screening and intracellular detection of nanomaterial Request PDF, accessed April 17, 2025, https://www.researchgate.net/publication/303842661_High_throughput_toxicity_screening_and_intracellular_detection_of_nanomaterial
  30. No Time To Lose - High Throughput Screening To Assess Nanomaterial Safety - PMC, accessed April 17, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3980675/
  31. (PDF) No Time To Lose - High Throughput Screening To Assess Nanomaterial Safety, accessed April 17, 2025, https://www.researchgate.net/publication/49817855_No_Time_To_Lose_-_High_Throughput_Screening_To_Assess_Nanomaterial_Safety
  32. Methods, models, mechanisms and metadata: Introducing the Nanotoxicology collection at F1000Research., accessed April 17, 2025, https://f1000research.com/articles/10-1196
  33. (PDF) Nanotoxicology: An Interdisciplinary Challenge - ResearchGate, accessed April 17, 2025, https://www.researchgate.net/publication/49807066_Nanotoxicology_An_Interdisciplinary_Challenge
  34. Developments in Delegations on the Safety of Manufactured Nanomaterials and Advanced Materials – Tour de Table - OECD, accessed April 17, 2025, https://one.oecd.org/document/ENV/CBC/MONO(2024)1/en/pdf
  35. t4 Workshop Report: Nanotoxicology: “The End of the Beginning” – Signs on the Roadmap to a Strategy for Assuring the Safe Application and Use of Nanomaterials - PMC, accessed April 17, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4038011/
  36. Current Strategies in Assessment of Nanotoxicity: Alternatives to In Vivo Animal Testing, accessed April 17, 2025, https://www.mdpi.com/1422-0067/22/8/4216
  37. High-Throughput Screening Platforms in the Discovery of Novel Drugs for Neurodegenerative Diseases - MDPI, accessed April 17, 2025, https://www.mdpi.com/2306-5354/8/2/30
  38. SCCS - Guidance on the safety assessment of nanomaterials in cosmetics - 2nd revision, accessed April 17, 2025, https://health.ec.europa.eu/publications/sccs-guidance-safety-assessment-nanomaterials-cosmetics-2nd-revision_en